c Ovid 19 Treatment Guidelines
c Ovid 19 Treatment Guidelines
c Ovid 19 Treatment Guidelines
The Coronavirus Disease 2019 (COVID-19) Treatment Guidelines is published in an electronic format
that can be updated in step with the rapid pace and growing volume of information regarding the
treatment of COVID-19.
The COVID-19 Treatment Guidelines Panel (the Panel) is committed to updating this document to
ensure that health care providers, patients, and policy experts have the most recent information regarding
the optimal management of COVID-19 (see the Panel Roster for a list of Panel members).
New Guidelines sections and recommendations and updates to existing Guidelines sections are
developed by working groups of Panel members. All recommendations included in the Guidelines are
endorsed by a majority of Panel members (see Guidelines Development for additional details on the
development process).
Major revisions to the Guidelines within the past month are as follows:
Molnupiravir
The Panel notes that there is a lack of definitive data regarding the benefit of molnupiravir in patients
who are vaccinated and at high risk of progressing to severe COVID-19. Due to the fetal toxicities
that have been reported in animal studies of molnupiravir, 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). It is not yet known how often SARS-CoV-2 viral rebound occurs
after molnupiravir treatment.
On October 14, 2022, the Centers for Disease Control and Prevention (CDC) reported a rapid increase in
certain SARS-CoV-2 Omicron subvariants circulating in the United States1 that are likely to be resistant
to some anti-SARS-CoV-2 monoclonal antibodies (mAbs). The subvariants BQ.1 and BQ.1.1 are likely
to be resistant to bebtelovimab, and the subvariants BA.4.6, BA.2.75.2, BF.7, BQ.1, and BQ.1.1 are
likely to be resistant to tixagevimab plus cilgavimab (Evusheld). The anticipated loss of susceptibility is
based on knowledge about amino acid mutations that confer antibody resistance and on available data
from in vitro neutralization studies.2
Although the proportions of these potentially resistant SARS-CoV-2 subvariants are increasing, their
prevalence is currently low or moderate. The COVID-19 Treatment Guidelines Panel (the Panel)
continues to recommend bebtelovimab for the treatment of COVID-19 only when ritonavir-boosted
nirmatrelvir (Paxlovid) or remdesivir cannot be used in nonhospitalized adults who are at high risk
of progressing to severe COVID-19 (see Therapeutic Management of Nonhospitalized Adults With
COVID-19). Ritonavir-boosted nirmatrelvir, remdesivir, and molnupiravir are expected to be active
against these subvariants.
The Panel continues to recommend the anti-SARS-CoV-2 mAbs tixagevimab plus cilgavimab as
pre-exposure prophylaxis (PrEP) for eligible individuals (see Prevention of SARS-CoV-2 Infection).
Individuals who receive tixagevimab plus cilgavimab as PrEP should continue to take precautions to
avoid infection. If they experience signs and symptoms consistent with COVID-19, they should be
tested for SARS-CoV-2 and, if infected, promptly seek medical attention for consideration of antiviral
treatment.
The Panel will closely monitor the prevalence of circulating subvariants with marked reduction in
susceptibility to anti-SARS-CoV-2 mAbs. The recommendations for the use of bebtelovimab for the
treatment of COVID-19 and for the use of tixagevimab plus cilgavimab for PrEP will be updated if the
prevalence of these subvariants increases.
References
1. Centers for Disease Control and Prevention. Variant proportions. 2022. Available at:
https://covid.cdc.gov/covid-data-tracker/#variant-proportions. Accessed October 18, 2022.
2. Cao Y, Jian F, Wang J, et al. Imprinted SARS-CoV-2 humoral immunity induces convergent Omicron RBD
evolution. bioRxiv. 2022;Preprint. Available at:
https://www.biorxiv.org/content/10.1101/2022.09.15.507787v3.
The COVID-19 Treatment Guidelines were developed to provide clinicians with guidance on how
to care for patients with COVID-19. Because clinical information about the optimal management of
COVID-19 is evolving quickly, these Guidelines are updated frequently to reflect newly published data
and other authoritative information.
Panel Composition
Members of the COVID-19 Treatment Guidelines Panel (the Panel) are appointed by the Panel co-chairs
based on their clinical experience and expertise in patient management, translational and clinical
science, and/or the development of treatment guidelines. Panel members include representatives from
federal agencies, health care organizations, academic organizations, and professional societies. Federal
agencies and professional societies represented on the Panel include:
• American Association of Critical-Care Nurses
• American Association for Respiratory Care
• American College of Chest Physicians
• American College of Clinical Pharmacy
• 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 their societies have
endorsed all elements of these Guidelines.
The names, affiliations, and financial disclosures of the Panel members and ex officio members, as well
as members of the Guidelines support team, are provided in the Panel Roster and Financial Disclosure
sections of the Guidelines.
To develop the recommendations in these Guidelines, the Panel uses data from the rapidly growing
body of research on COVID-19. The Panel also relies heavily on experience with other diseases,
supplemented with the members’ evolving clinical experience with COVID-19.
In general, the recommendations in these Guidelines fall into the following categories:
• The Panel recommends using [blank] for the treatment of COVID-19 (rating).
Recommendations in this category are based on evidence that the potential benefits of using this
intervention outweigh the potential risks.
• There is insufficient evidence for the Panel to recommend either for or against the use of
[blank] for the treatment of COVID-19 (no rating). This statement is used when there are
currently not enough data to support a recommendation, or the available data are conflicting.
• The Panel recommends against the use of [blank] for the treatment of COVID-19, except
COVID-19 Treatment Guidelines 10
Epidemiology
The COVID-19 pandemic has exploded since cases were first reported in China in December 2019. As
of April 15, 2022, more than 503 million cases of COVID-19—caused by SARS-CoV-2 infection—have
been reported globally, including more than 6.2 million deaths.1
Individuals of all ages are at risk for SARS-CoV-2 infection and severe disease. However, the
probability of serious COVID-19 disease is higher in people aged ≥60 years, those living in a nursing
home or long-term care facility, and those with chronic medical conditions. In an analysis of more than
1.3 million laboratory-confirmed cases of COVID-19 that were reported in the United States between
January and May 2020, 14% of patients required hospitalization, 2% were admitted to the intensive care
unit, and 5% died.2 The percentage of patients who died was 12 times higher among those with reported
medical conditions (19.5%) than among those without medical conditions (1.6%), and the percentage
of patients who were hospitalized was 6 times higher among those with reported medical conditions
(45.4%) than among those without medical conditions (7.6%). Mortality was highest in patients aged
>70 years, regardless of the presence of chronic medical conditions. Data on comorbid health conditions
among patients with COVID-19 indicate that 32% had cardiovascular disease, 30% had diabetes, and
18% had chronic lung disease. Other conditions that may lead to a high risk for severe COVID-19
include cancer, kidney disease, liver disease (especially in patients with cirrhosis), obesity, sickle cell
disease, and other immunocompromising conditions. Transplant recipients and pregnant people are also
at a higher risk of severe COVID-19.3-10
Data from the United States suggest that racial and ethnic minorities experience higher rates of
COVID-19, subsequent hospitalization, and death.11-15 However, surveillance data that include race and
ethnicity are not available for most reported cases of COVID-19 in the United States.4,16 Factors that
contribute to the increased burden of COVID-19 in these populations may include over-representation
in work environments that confer higher risks of exposure to COVID-19, economic inequality (which
limits people’s ability to protect themselves against COVID-19 exposure), neighborhood disadvantage,17
and a lack of access to health care.16 Structural inequalities in society contribute to health disparities for
racial and ethnic minority groups, including higher rates of comorbid conditions (e.g., cardiac disease,
diabetes, hypertension, obesity, pulmonary diseases), which further increase the risk of developing
severe COVID-19.15
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 past SARS-CoV-2 infection or vaccination.
This viral evolution may increase the risk of reinfection or decrease the efficacy of vaccines.18 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 certain monoclonal antibodies (mAbs) that are being
considered for prevention and treatment.19-21
Since December 2020, the World Health Organization (WHO) has assigned Greek letter designations to
several identified variants. A SARS-CoV-2 variant designated as a variant of concern (VOC) displays
certain characteristics, such as increased transmissibility or virulence. In addition, vaccines and
COVID-19 Treatment Guidelines 12
Clinical Presentation
The estimated incubation period for COVID-19 is up to 14 days from the time of exposure, with a
median incubation period of 4 to 5 days.6,28,29 The spectrum of illness can range from asymptomatic
infection to severe pneumonia with acute respiratory distress syndrome and death. Among 72,314 people
with COVID-19 in China, 81% of cases were reported to be mild (defined in this study as no pneumonia
or mild pneumonia), 14% were severe (defined as dyspnea, respiratory frequency ≥30 breaths/min,
oxygen saturation ≤93%, a ratio of arterial partial pressure of oxygen to fraction of inspired oxygen
[PaO2/FiO2] <300 mm Hg, and/or lung infiltrates >50% within 24 to 48 hours), and 5% were critical
(defined as respiratory failure, septic shock, and/or multiple organ dysfunction syndrome or failure).30 In
a report on more than 370,000 confirmed COVID-19 cases with reported symptoms in the United States,
70% of patients experienced fever, cough, or shortness of breath; 36% had muscle aches; and 34%
reported headaches.2 Other reported symptoms have included, but are not limited to, diarrhea, dizziness,
rhinorrhea, anosmia, dysgeusia, sore throat, abdominal pain, anorexia, and vomiting.
The abnormalities seen in chest X-rays of patients with COVID-19 vary, but bilateral multifocal
COVID-19 Treatment Guidelines 13
References
1. Johns Hopkins University and Medicine. COVID-19 dashboard. 2021. Available at:
https://coronavirus.jhu.edu/map.html. Accessed February 15, 2022.
2. Stokes EK, Zambrano LD, Anderson KN, et al. Coronavirus disease 2019 case surveillance—United States,
January 22–May 30, 2020. MMWR Morb Mortal Wkly Rep. 2020;69(24):759-765. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32555134.
3. Cai Q, Chen F, Wang T, et al. Obesity and COVID-19 severity in a designated hospital in Shenzhen, China.
Diabetes Care. 2020;43(7):1392-1398. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32409502.
4. Centers for Disease Control and Prevention. COVID Data Tracker. 2022. Available at:
https://covid.cdc.gov/covid-data-tracker/#datatracker-home. Accessed April 15, 2022.
5. Garg S, Kim L, Whitaker M, et al. Hospitalization rates and characteristics of patients hospitalized with
laboratory-confirmed coronavirus disease 2019—COVID-NET, 14 states, March 1–30, 2020. MMWR Morb
Mortal Wkly Rep. 2020;69(15):458-464. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32298251.
6. 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.
7. Wu C, Chen X, Cai Y, et al. Risk factors associated with acute respiratory distress syndrome and death in
patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. 2020;180(7):934-943.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32167524.
8. Palaiodimos L, Kokkinidis DG, Li W, et al. Severe obesity, increasing age and male sex are independently
associated with worse in-hospital outcomes, and higher in-hospital mortality, in a cohort of patients with
COVID-19 in the Bronx, New York. Metabolism. 2020;108:154262. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32422233.
9. 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://www.ncbi.nlm.nih.gov/pubmed/33151921.
10. Centers for Disease Control and Prevention. COVID-19: people with certain medical conditions. 2021.
Available at: https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/people-with-medical-
conditions.html. Accessed April 13, 2022.
Summary Recommendations
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends using either a nucleic acid amplification test
(NAAT) or an antigen test with a sample collected from the upper respiratory tract (e.g., nasopharyngeal, nasal mid-
turbinate, anterior nasal) to diagnose acute SARS-CoV-2 infection (AIII).
• A NAAT should not be repeated in an asymptomatic person (with the exception of health care workers) within 90 days
of a previous SARS-CoV-2 infection, even if the person has had a significant exposure to SARS-CoV-2 (AIII).
• SARS-CoV-2 reinfection has been reported in people after an initial diagnosis of the infection; therefore, clinicians
should consider using a NAAT for those who have recovered from a previous infection and who present with
symptoms that are compatible with SARS-CoV-2 infection if there is no alternative diagnosis (BIII).
• The Panel recommends against diagnosing acute SARS-CoV-2 infection solely on the basis of serologic (i.e.,
antibody) test results (AIII).
• There is insufficient evidence for the Panel to recommend either for or against the use of SARS-CoV-2 serologic
testing to assess for immunity or to guide clinical decisions about using COVID-19 vaccines or anti-SARS-CoV-2
monoclonal antibodies.
Nucleic Acid Amplification Testing for Individuals With a Previous Positive SARS-CoV-2 Test
Result
NAATs can detect SARS-CoV-2 RNA in specimens obtained weeks to months after the onset of
COVID-19 symptoms.15,16 However, the likelihood of recovering replication-competent virus >10 days
from the onset of symptoms in those with mild disease and >20 days in those with severe disease is
very low.17,18 Furthermore, both virologic studies and contact tracing of high-risk contacts show a low
risk for SARS-CoV-2 transmission after these intervals.19,20 Based on these results, the Centers for
Disease Control and Prevention (CDC) does not recommend repeating NAATs in asymptomatic persons
within 90 days of a previous SARS-CoV-2 infection, even if the person has had a significant exposure
to SARS-CoV-2.21 An exception to this is for health care workers who meet the specific criteria found
in CDC guidance.22 If there are concerns that an immunocompromised health care worker may still be
infectious >20 days from the onset of SARS-CoV-2 infection, consulting local employee health services
References
1. Centers for Disease Control and Prevention. Testing strategies for SARS-CoV-2. 2021. Available at: https://
www.cdc.gov/coronavirus/2019-ncov/lab/resources/sars-cov2-testing-strategies.html. Accessed June 16, 2022.
2. Food and Drug Administration. Coronavirus disease 2019 (COVID-19) emergency use authorizations for
medical devices. 2021. Available at: https://www.fda.gov/medical-devices/emergency-use-authorizations-
medical-devices/coronavirus-disease-2019-covid-19-emergency-use-authorizations-medical-devices. Accessed
June 16, 2022.
3. Centers for Disease Control and Prevention. Interim guidelines for collecting and handling of clinical
specimens for COVID-19 testing. 2021. Available at: https://www.cdc.gov/coronavirus/2019-ncov/lab/
guidelines-clinical-specimens.html. Accessed June 16, 2022.
4. Centers for Disease Control and Prevention. Nucleic acid amplification tests (NAATs). 2021. Available at:
https://www.cdc.gov/coronavirus/2019-ncov/lab/naats.html. Accessed June 16, 2022.
5. 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 June 16, 2022.
6. Kucirka LM, Lauer SA, Laeyendecker O, Boon D, Lessler J. Variation in false-negative rate of reverse
transcriptase polymerase chain reaction-based SARS-CoV-2 tests by time since exposure. Ann Intern Med.
2020;173(4):262-267. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32422057.
7. Centers for Disease Control and Prevention. Science brief: emerging SARS-CoV-2 variants. 2021. Available
at: https://www.cdc.gov/coronavirus/2019-ncov/science/science-briefs/scientific-brief-emerging-variants.html.
Accessed June 16, 2022.
8. Chan PK, To WK, Ng KC, et al. Laboratory diagnosis of SARS. Emerg Infect Dis. 2004;10(5):825-831.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/15200815.
9. Tang P, Louie M, Richardson SE, et al. Interpretation of diagnostic laboratory tests for severe acute respiratory
syndrome: the Toronto experience. CMAJ. 2004;170(1):47-54. Available at: https://www.ncbi.nlm.nih.gov/
Vaccines
Vaccination is the most effective way to prevent SARS-CoV-2 infection. The COVID-19 Treatment
Guidelines Panel (the Panel) recommends COVID-19 vaccination as soon as possible for everyone who
is eligible according to the CDC’s Advisory Committee on Immunization Practices (AI). Three vaccines
are authorized or approved for use in the United States to prevent COVID-19. For primary and booster
vaccinations, the mRNA vaccines (i.e., BNT162b2 [Pfizer-BioNTech] or mRNA-1273 [Moderna]) are
preferable to the Ad26.COV2.S (Johnson & Johnson/Janssen) vaccine due to its risk of serious adverse
events.4 A primary series of COVID-19 vaccinations is recommended for everyone aged ≥6 months in
the United States. Certain groups of people should receive additional doses at specified intervals after
the primary series of vaccinations. The type and dose of vaccine and the timing of these additional doses
depend on the recipient’s age and underlying medical conditions. The CDC regularly updates the clinical
considerations for use of the COVID-19 vaccines that are currently approved by the Food and Drug
Administration (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
moderate in severity (i.e., they did not prevent vaccinated people from engaging in daily activities)
and resolved after 1 or 2 days. There have been a few reports of severe allergic reactions following
COVID-19 vaccination, including rare reports of patients who experienced anaphylaxis after receiving
an mRNA vaccine.6,7
Reports have suggested that there is an increased risk of thrombosis with thrombocytopenia syndrome
(TTS) in adults who have received the Ad26.COV2.S vaccine7 and, rarely, the mRNA-1273 vaccine.8
TTS is a rare but serious condition that causes blood clots in large blood vessels and low platelet levels.
Women aged 30 to 49 years should be aware of the increased risk of TTS. The American Society
of Hematology and the American Heart Association/American Stroke Association Stroke Council
leadership have published considerations that are relevant to the diagnosis and treatment of TTS that
occurs in people who receive the Ad26.COV2.S vaccine.9,10 These considerations include information
on administering a nonheparin anticoagulant and intravenous immunoglobulin to these patients. Given
the rarity of this syndrome and the unique treatment required, consider consulting a hematologist when
treating these patients.
Myocarditis and pericarditis after COVID-19 vaccination are rare, and most of the reported cases were
very mild and self-limiting.11 These conditions have occurred most often in male adolescents, young
adults, and people who have received mRNA vaccines.
Guillain-Barré syndrome (GBS) in people who received the Ad26.COV2.S vaccine is rare.11 GBS is
a neurologic disorder that causes muscle weakness and sometimes paralysis. Most people with GBS
fully recover, but some have permanent nerve damage. Onset typically occurs about 2 weeks after
vaccination. GBS has mostly been reported in men aged ≥50 years.
Pre-Exposure Prophylaxis
Anti-SARS-CoV-2 Monoclonal Antibodies
Vaccination remains the most effective way to prevent SARS-CoV-2 infection and should be considered
the first line of prevention. However, some individuals cannot or may not mount an adequate protective
response to COVID-19 vaccines. Others may not have been fully vaccinated because they have a history
of severe adverse reactions to a COVID-19 vaccine or its components.
On the basis of results from PROVENT, a large randomized controlled trial conducted when the major
circulating SARS-CoV-2 variants were Alpha, Beta, Delta, and Epsilon,19 the FDA issued an Emergency
Use Authorization (EUA) for the anti-SARS-CoV-2 monoclonal antibodies (mAbs) tixagevimab plus
cilgavimab (Evusheld).20 The EUA allows these anti-SARS-CoV-2 mAbs to be used as pre-exposure
prophylaxis (PrEP) for certain individuals who are immunocompromised and therefore may have
inadequate responses to COVID-19 vaccines or are unable to be fully vaccinated due to a history of
severe adverse reactions to a COVID-19 vaccine. A modification in the fragment crystallizable (Fc)
region gives these anti-SARS-CoV-2 mAbs prolonged half-lives, resulting in potential protection from
SARS-CoV-2 infection for up to 6 months, depending on the variant.
The PROVENT trial used tixagevimab 150 mg plus cilgavimab 150 mg, which was the dose initially
authorized by the FDA. However, in vitro data showed that some subvariants of the Omicron variant
had decreased susceptibility to tixagevimab plus cilgavimab. Because of these findings, in February
2022, the FDA revised the EUA to authorize the use of an increased dose of tixagevimab 300 mg
plus cilgavimab 300 mg.20-23 For patients who previously received a dose of tixagevimab 150 mg plus
cilgavimab 150 mg, the FDA EUA suggested administering an second dose; the specific dose depends
on the amount of time that has passed since the initial dose (see below). On June 30, 2022, the FDA
further revised the EUA to authorize repeated doses of tixagevimab 300 mg plus cilgavimab 300 mg to
be administered every 6 months.
When prescribing tixagevimab plus cilgavimab for SARS-CoV-2 PrEP, clinicians should be aware of the
following:
• Tixagevimab plus cilgavimab is authorized for use as PrEP in a population that was not well
represented in the PROVENT trial (i.e., a very small proportion of the participants were
immunocompromised).
• There are no clinical trial efficacy data on the use of tixagevimab 300 mg plus cilgavimab 300 mg
for the prevention of symptomatic COVID-19, and there are no data for any repeated dose at any
defined interval, including for the authorized schedule of repeat dosing every 6 months proposed
in the latest EUA. This dose and the strategy of repeating the dose every 6 months are based on
COVID-19 Treatment Guidelines 26
Post-Exposure Prophylaxis
Anti-SARS-CoV-2 Monoclonal Antibodies
• The Panel recommends against the use of bamlanivimab plus etesevimab and casirivimab
plus imdevimab for post-exposure prophylaxis (PEP), as the Omicron variant and its subvariants,
which are not susceptible to these agents, are currently the dominant SARS-CoV-2 variants
circulating in the United States (AIII).
Vaccination remains a highly effective way to prevent SARS-CoV-2 infection. However, despite the
widespread availability of COVID-19 vaccines, some individuals are not fully vaccinated or cannot
mount an adequate response to the vaccine. Some of these individuals, if infected, are at high risk of
progressing to serious COVID-19. Bamlanivimab plus etesevimab and casirivimab plus imdevimab have
previously received FDA EUAs for PEP; however, the Omicron variant and its subvariants are currently
the dominant SARS-CoV-2 variants circulating in the United States. The Panel recommends against the
use of these anti-SARS-CoV-2 mAbs because the Omicron variant and its subvariants are not susceptible
to them (AIII).
References
1. Centers for Disease Control and Prevention. Scientific brief: SARS-CoV-2 transmission. 2021. Available at:
https://www.cdc.gov/coronavirus/2019-ncov/science/science-briefs/sars-cov-2-transmission.html. Accessed
June 21, 2022.
2. Centers for Disease Control and Prevention. How to protect yourself and others. 2022. Available at: https://
www.cdc.gov/coronavirus/2019-ncov/prevent-getting-sick/prevention.html. Accessed September 9, 2022.
3. Centers for Disease Control and Prevention. Infection control guidance for healthcare professionals about
coronavirus (COVID-19). 2020. Available at: https://www.cdc.gov/coronavirus/2019-ncov/hcp/infection-
control.html. Accessed June 21, 2022.
4. Centers for Disease Control and Prevention. Johnson & Johnson’s Janssen COVID-19 vaccine: overview and
safety. 2022. Available at: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/different-vaccines/janssen.
html. Accessed June 21, 2022.
5. Centers for Disease Control and Prevention. Use of COVID-19 vaccines in the United States: interim clinical
considerations. 2022. Available at: https://www.cdc.gov/vaccines/covid-19/clinical-considerations/covid-19-
vaccines-us.html. Accessed June 21, 2022.
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 June 21, 2022.
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, 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.
9. American Society of Hematology. Vaccine-induced immune thrombotic thrombocytopenia. 2022. Available at:
https://www.hematology.org/covid-19/vaccine-induced-immune-thrombotic-thrombocytopenia. Accessed June
21, 2022.
10. Furie KL, Cushman M, Elkind MSV, et al. Diagnosis and management of cerebral venous sinus thrombosis
with vaccine-induced immune thrombotic thrombocytopenia. Stroke. 2021;52(7):2478-2482. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33914590.
11. Centers for Disease Control and Prevention. Selected adverse events reported after COVID-19 vaccination.
2022. Available at: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/safety/adverse-events.html. Accessed
June 21, 2022.
12. Centers for Disease Control and Prevention. COVID-19 vaccines while pregnant or breastfeeding. 2022.
Available at: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/recommendations/pregnancy.html. Accessed
June 21, 2022.
13. Society for Maternal-Fetal Medicine. Publications and clinical guidance. 2022. Available at:
https://www.smfm.org/covidclinical. Accessed June 21, 2022.
14. Shimabukuro TT, Kim SY, Myers TR, et al. Preliminary findings of mRNA COVID-19 vaccine safety in
pregnant persons. N Engl J Med. 2021;384(24):2273-2282. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33882218.
15. Zauche LH, Wallace B, Smoots AN, et al. Receipt of mRNA COVID-19 vaccines and risk of spontaneous
abortion. N Engl J Med. 2021;385(16):1533-1535. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34496196.
COVID-19 Treatment Guidelines 30
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 who have no
symptoms that are 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 who 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 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, and/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.
Patients who are aged ≥65 years are at a higher risk of progressing to severe COVID-19. Other
underlying conditions associated with a higher risk of severe COVID-19 include asthma, 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.1 Health care providers should closely monitor patients with
these conditions until they achieve clinical recovery.
The initial evaluation for patients may include chest imaging (e.g., X-ray, ultrasound or computed
tomography scan) and an electrocardiogram. 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.2-4
The definitions for the severity of illness categories also apply to pregnant patients. However, the
threshold for certain interventions is different for pregnant patients 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 adequate oxygen delivery to the fetus.5
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 through telemedicine or telephone visits. No
imaging or specific laboratory evaluations are routinely indicated in otherwise healthy patients with
mild COVID-19. Older patients and those with underlying comorbidities are at higher risk of disease
progression; therefore, health care providers should monitor these patients closely until clinical
recovery is achieved. 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 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 anti-
SARS-CoV-2 therapies. If bacterial pneumonia is suspected, administer empiric antibiotic treatment,
re-evaluate the patient daily, and de-escalate or stop antibiotics if further testing indicates the patient
does not have a bacterial infection.
Severe Illness
Patients with COVID-19 are considered to have severe illness if they have SpO2 <94% on room air
COVID-19 Treatment Guidelines 35
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.
Successful clinical management of a patient with COVID-19, as with any patient in the intensive care
unit (ICU), includes treating both the medical condition that initially resulted in ICU admission as
well as other comorbidities and nosocomial complications. For more information, see Critical Care for
Adults.
Persistent Symptoms
The prevalence of persistent post-COVID clinical signs and symptoms remains unclear. In a systematic
review of 25 observational cohort studies, prevalence varied widely (from 5% to 80%) and likely
reflected differences in study populations, case definitions, and data resources.43 Another large,
systematic review found a similar prevalence of post-COVID symptoms 6 months after initial infection
between studies from high-income or low- and middle-income countries and between studies in which
>60% or <60% of the patients were hospitalized.44
A prospective study conducted at the University of Washington investigated mostly outpatients with
laboratory-confirmed SARS-CoV-2 infection (150 participants had mild illness, 11 had no symptoms,
and 11 had moderate or severe disease that required hospitalization).45 Participants completed a
follow-up questionnaire 3 months to 9 months after illness onset; 33% of outpatients and 31% of
hospitalized patients reported ≥1 persistent symptom. Persistent symptoms were reported by 27% of the
patients aged 18 to 39 years, 30% of those aged 40 to 64 years, and 43% of those aged ≥65 years.
In these and other studies, the most commonly reported nonneurologic, persistent symptoms included
fatigue or muscle weakness, joint pain, chest pain, palpitations, shortness of breath, and cough.44-51 From
January 2020 to April 2021, the CDC conducted an internet-based survey of 3,135 noninstitutionalized
adults who self-reported receiving either a positive or negative SARS-CoV-2 test result.52 The study
found that fatigue, shortness of breath, and cough were commonly reported symptoms lasting >4 weeks
after onset. The prevalence of these symptoms among participants with a positive test result versus the
prevalence among participants with a negative test result was 22.5% versus 12% for fatigue, 15.5%
versus 5.2% for shortness of breath, and 14.5% versus 4.9% for cough.
Some of the reported symptoms overlap with post-intensive care syndrome symptoms that have been
described for patients without COVID-19. Prolonged symptoms and disabilities after COVID-19 have
also been reported in patients with milder illness, including outpatients.53,54
Patients who had breakthrough infection after COVID-19 vaccination are less likely to report symptoms
that persist ≥28 days than patients with SARS-CoV-2 infection who are unvaccinated.55,56 The COVID
Symptom Study, conducted from December 2020 to July 2021, included participants who used a mobile
application to report symptoms after breakthrough infections or reinfection.46 The investigators found
that the odds of reporting symptoms for ≥28 days was reduced by about half among participants who
received 2 vaccine doses, when compared with participants who received 1 or 0 vaccine doses.
A study of electronic health record data from 59 health care organizations, primarily in the United
States, compared the records of people who did not receive any vaccine doses with records of people
COVID-19 Treatment Guidelines 38
Cardiopulmonary Injury
A U.S. Department of Veterans Affairs (VA) study of a national health care database compared 153,760
veterans who survived the first 30 days of COVID-19 to contemporary and historical control cohorts
that had no evidence of SARS-CoV-2 infection.58 When compared with the control cohorts, patients
with history of COVID-19 had a greater incidence of post-acute cardiovascular outcomes (e.g.,
cerebrovascular disorder, dysrhythmia, inflammatory heart disease, ischemic heart disease, heart failure,
thromboembolic disease) at 12 months.
A prospective study of pulmonary function examined longitudinal data from the adult Copenhagen
General Population Study and found that pulmonary function declined faster for the 107 patients with
mostly mild COVID-19 than for a matched sample from the general population.59
Neuropsychiatric Impairment
Neuropsychiatric impairments have been reported among patients who have recovered from acute
COVID-19. Reported persistent neurologic symptoms include headaches, vision changes, hearing loss,
impaired mobility, numbness in extremities, restless legs syndrome, tremors, memory loss, cognitive
impairment, sleep difficulties, concentration problems, mood changes, and loss of smell or taste.60-63
One study in the United Kingdom administered cognitive tests to 84,285 participants who had recovered
from suspected or confirmed SARS-CoV-2 infection. These participants had worse performances across
multiple domains than would be expected for people with the same ages and demographic profiles; this
effect was observed even among those who had not been hospitalized.64 However, the study authors did
not report when the tests were administered in relation to the diagnosis of COVID-19.
A retrospective cohort study examined the electronic health records of 273,618 patients from 59 health
care organizations, primarily in the United States.65 The study reported that cognitive dysfunction
(defined using International Classification of Diseases, Tenth Revision codes) 3 to 6 months after
diagnosis was worse for people with COVID-19 than for people with influenza. Other studies have
reported high rates of anxiety and depression among patients who evaluated their psychiatric distress
using self-report scales.51,66 Reports also show that patients aged ≤60 years experienced more psychiatric
symptoms than patients aged >60 years.50,51
Metabolic Perturbations
There have been reports of new-onset diabetes after COVID-19.67 A study of a VA national health care
database analyzed the records of 181,280 people who survived the first 30 days of COVID-19 and
compared them to a contemporary control cohort that had no evidence of SARS-CoV-2 infection. People
with a history of COVID-19 had a 40% greater risk of diabetes 12 months after infection than people
in the control cohort.68 A CDC study of people aged <18 years reported that those with a history of
COVID-19 had an increased risk of diabetes >30 days after SARS-CoV-2 infection when compared with
those with no history of infection.69
Research on persistent symptoms and other conditions after COVID-19 is ongoing, including the
National Institutes of Health’s RECOVER Initiative, which aims to better characterize the prevalence,
characteristics, and pathophysiology of post-acute sequelae of SARS-CoV-2 and inform potential
therapeutic strategies.
References
1. Centers for Disease Control and Prevention. Underlying medical conditions associated with higher risk
for severe COVID-19: information for healthcare professionals. 2022. Available at: https://www.cdc.gov/
coronavirus/2019-ncov/hcp/clinical-care/underlyingconditions.html. Accessed September 16, 2022.
2. 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.
3. 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.
4. Casas-Rojo JM, Anton-Santos JM, Millan-Nunez-Cortes 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.
5. 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_
COVID_pos_preg_patients_2-2-21_(final).pdf.
6. Abbassi-Ghanavati M, Greer LG, Cunningham FG. Pregnancy and laboratory studies: a reference table for
clinicians. Obstet Gynecol. 2009;114(6):1326-1331. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19935037.
7. Anderson BL, Mendez-Figueroa H, Dahlke JD, et al. Pregnancy-induced changes in immune protection of the
genital tract: defining normal. Am J Obstet Gynecol. 2013;208(4):e321-e329. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23313311.
8. Riphagen S, Gomez X, Gonzalez-Martinez C, Wilkinson N, Theocharis P. Hyperinflammatory shock in
children during COVID-19 pandemic. Lancet. 2020;395(10237):1607-1608. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32386565.
9. 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.
10. Food and Drug Administration. Pulse oximeter accuracy and limitations: FDA safety communication. 2022.
Available at: https://www.fda.gov/medical-devices/safety-communications/pulse-oximeter-accuracy-and-
limitations-fda-safety-communication. Accessed August 11, 2022.
11. 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;Published online
ahead of print. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35679133.
12. 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,
COVID-19 Treatment Guidelines 40
The prioritization guidance in this section should be used only when logistical or supply constraints limit
the availability of therapies. When there are no supply or logistical constraints, the COVID-19 Treatment
Guidelines Panel (the Panel) recommends that therapies for the prevention or treatment of SARS-CoV-2
be prescribed for any eligible individual as recommended in these Guidelines.
At times during the pandemic, increased cases of COVID-19 and the emergence of new variants of
concern have resulted in logistical or supply constraints that made offering recommended therapies to all
eligible patients impossible. In these situations, prioritization of therapy for those who will benefit the
most becomes necessary. This section provides guidance on which individuals might receive the greatest
benefit from anti-SARS-CoV-2 therapeutics for treatment or prevention when access is limited.
The Panel has recommended several therapeutic agents for the treatment and prevention of SARS-CoV-2
infection in individuals who are at high risk for progression to severe COVID-19. These anti-SARS-
CoV-2 therapeutics have the greatest proven clinical benefit for nonhospitalized patients who have risk
factors for progression to severe COVID-19. The risks for progression are substantially higher for those
who are not vaccinated or who are vaccinated but not expected to mount an adequate immune response
to the vaccine.
The Food and Drug Administration provides a broad list of medical conditions or other factors as criteria
for use of anti-SARS-CoV-2 agents as treatment or pre-exposure prophylaxis (PrEP).
When it becomes necessary to triage patients for receipt of anti-SARS-CoV-2 therapies or preventive
strategies, the Panel suggests prioritizing:
• Treatment of COVID-19 in unvaccinated or incompletely vaccinated individuals with clinical risk
factors for severe illness and vaccinated individuals who are not expected to mount an adequate
immune response (see Immunocompromising Conditions below)
• Use of tixagevimab plus cilgavimab (Evusheld) as PrEP for individuals who are severely
immunocompromised over those who are moderately immunocompromised (see
Immunocompromising Conditions below)
Immunocompromising Conditions
The CDC website COVID-19 Vaccines for People Who Are Moderately or Severely Immunocompromised
provides a list of moderate or severe immunocompromising conditions.1
If, because of logistical constraints or supply limitations, anti-SARS-CoV-2 therapies cannot be provided
to all individuals who are moderately to severely immunocompromised, the Panel suggests prioritizing
their use for patients who are least likely to mount an adequate response to COVID-19 vaccination
or SARS-CoV-2 infection and who are at risk for severe outcomes, including (but not limited to) the
following populations:
• Patients who are within 1 year of receiving B cell-depleting therapies (e.g., rituximab, ocrelizumab,
ofatumumab, alemtuzumab)
COVID-19 Treatment Guidelines 46
References
1. Centers for Disease Control and Prevention. COVID-19 vaccines for people who are moderately or
severely immunocompromised. 2022. Available at: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/
recommendations/immuno.html. Accessed May 9, 2022.
2. Centers for Disease Control and Prevention. COVID-19 risks and vaccine information for older adults. 2021.
Available at: https://www.cdc.gov/aging/covid19/covid19-older-adults.html. Accessed December 22, 2021.
3. Rosenthal N, Cao Z, Gundrum J, Sianis J, Safo S. Risk factors associated with in-hospital mortality in a U.S.
national sample of patients with COVID-19. JAMA Netw Open. 2020;3(12):e2029058. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33301018.
4. Kompaniyets L, Agathis NT, Nelson JM, et al. Underlying medical conditions associated with severe
COVID-19 illness among children. JAMA Netw Open. 2021;4(6):e2111182. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34097050.
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
appears to be driven by a dysregulated immune/inflammatory response to SARS-CoV-2 that leads to
tissue damage. 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, while immunosuppressive/anti-inflammatory
therapies are likely to be more beneficial in the later stages of COVID-19.
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 adult patients. 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 adult patients
according to their disease severity and oxygen requirements.
Patients who require minimal Remdesivire (BIIa) For nonpregnant patients with D-dimer
conventional oxygen levels above the ULN who do not have
Hospitalized Most patients Use dexamethasone plus remdesivire (BIIa). If remdesivir cannot be an increased bleeding risk:
and Requires obtained, use dexamethasone (BI). • Therapeutic dose of heparing (CIIa)
Conventional Patients who are receiving Add PO baricitinibf or IV tocilizumabf to 1 of the options above (BIIa). For other patients:
Oxygend dexamethasone and who have • Prophylactic dose of heparin,
rapidly increasing oxygen needs unless contraindicated (AI); (BIII) for
and systemic inflammation pregnant patients
Most patients Promptly start 1 of the following, if not already initiated: For patients without an indication for
• Dexamethasone plus PO baricitinibf (AI) therapeutic anticoagulation:
Hospitalized and • Dexamethasone plus IV tocilizumab (BIIa) f • Prophylactic dose of heparin,
Requires HFNC unless contraindicated (AI); (BIII) for
If baricitinib, tofacitinib, tocilizumab, or sarilumab cannot be obtained: pregnant patients
Oxygen or NIV
• Dexamethasoneh (AI)
For patients who are started on a
Add remdesivir to 1 of the options above in certain patients (CIIa).i therapeutic dose of heparin in a non-
Most patients Promptly start 1 of the following, if not already initiated: ICU setting and then transferred to the
• Dexamethasone plus PO baricitinib (BIIa)f ICU, the Panel recommends switching
Hospitalized and
• Dexamethasone plus IV tocilizumab (BIIa) f to a prophylactic dose of heparin,
Requires MV or
unless there is another indication for
ECMO If baricitinib, tofacitinib, tocilizumab, or sarilumab cannot be obtained: therapeutic anticoagulation (BIII).
• Dexamethasoneh (AI)
Introduction
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. The Panel recognizes that there are times during the
COVID-19 pandemic when the distinction between outpatient and inpatient care may be less clear.
Patients with COVID-19 may receive care outside traditional ambulatory care or hospital settings if
there is a shortage of hospital beds, staff, or resources. In addition, asymptomatic SARS-CoV-2 infection
or mild disease may be diagnosed during a patient’s hospital admission for a non-COVID-19 condition.
Health care providers should use their judgment when deciding whether the guidance offered in this
section applies to individual patients.
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: telemedicine, remote monitoring, in-person visits, and home visits by nurses or other health
care providers.
Data from the United States show that racial and ethnic minorities experience higher rates of COVID-19,
COVID-19 Treatment Guidelines 52
Considerations in Pregnancy
Managing pregnant outpatients with COVID-19 is similar to managing nonpregnant patients (see
Special Considerations in Pregnancy). Clinicians should offer supportive care and therapeutics as
indicated, take steps to reduce the risk of SARS-CoV-2 transmission, and provide guidance on when
to seek an in-person evaluation. The American College of Obstetricians and Gynecologists (ACOG)
has published recommendations on how to use telehealth for prenatal care and how to modify routine
prenatal care when necessary to decrease the risk of SARS-CoV-2 transmission to patients, caregivers,
and staff.27
In pregnant patients, SpO2 should be maintained at 95% or above on room air at sea level; therefore,
the threshold for monitoring pregnant patients in an inpatient setting may be lower than in nonpregnant
patients. At this time, there are no changes to fetal monitoring recommendations in the outpatient setting,
and fetal surveillance and management should be similar to the fetal surveillance and management used
for pregnant patients with medical illness.28,29 However, these monitoring strategies can be discussed on
a case-by-case basis with an obstetrician. Pregnant and lactating patients should be given the opportunity
to participate in clinical trials of outpatients with COVID-19 to help inform decision-making in this
population.
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.
2. Gold JAW, Wong KK, Szablewski CM, et al. Characteristics and clinical outcomes of adult patients
hospitalized with COVID-19—Georgia, March 2020. MMWR Morb Mortal Wkly Rep. 2020;69(18):545-550.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32379729.
3. Gross CP, Essien UR, Pasha S, et al. Racial and ethnic disparities in population-level COVID-19 mortality. J
Gen Intern Med. 2020;35(10):3097-3099. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32754782.
4. Nayak A, Islam SJ, Mehta A, et al. Impact of social vulnerability on COVID-19 incidence and outcomes in the
United States. medRxiv. 2020;Preprint. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32511437.
5. Price-Haywood EG, Burton J, Fort D, Seoane L. Hospitalization and mortality among Black patients and
White patients with COVID-19. N Engl J Med. 2020;382(26):2534-2543. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32459916.
6. Azam TU, Berlin H, Anderson E, et al. Differences in inflammation, treatment, and outcomes between Black
COVID-19 Treatment Guidelines 56
Symptom Management
Treatment of symptoms includes using over-the-counter antipyretics, analgesics, or antitussives for
fever, headache, myalgias, and cough. Patients should be advised to drink fluids regularly to avoid
dehydration. Rest is recommended as needed during the acute phase of COVID-19, and ambulation
and other forms of activity should be increased according to the patient’s tolerance. Patients should be
educated about the variability in time to symptom resolution and complete recovery. When possible,
patients with symptoms of COVID-19 should be triaged via telehealth visits to determine whether they
require COVID-19-specific therapy and in-person care (AIII).
At a minimum, health care providers should use telehealth to closely follow patients with dyspnea,
and in-person monitoring of these patients should be considered (AIII). Patients with persistent or
progressive dyspnea, especially those who have an oxygen saturation measured by pulse oximetry
≤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).
Remdesivir
Remdesivir is a nucleotide prodrug of an adenosine analog that binds to the viral RNA-dependent
RNA polymerase and inhibits viral replication by terminating RNA transcription prematurely. It
is approved by the FDA for the treatment of COVID-19 in adults and children aged ≥28 days and
weighing ≥3 kg who are hospitalized with COVID-19 and for those with mild to moderate COVID-19
who are not hospitalized and are at high risk of progression to severe disease. In the PINETREE trial,
nonhospitalized patients with mild to moderate COVID-19 who were unvaccinated and at high risk
of progressing to severe disease received 3 days of intravenous (IV) remdesivir or placebo. Use of
remdesivir resulted in an 87% relative reduction in the risk of hospitalization or death.6 Remdesivir has
demonstrated activity in vitro and in animal studies against the Omicron variant and its subvariants.23-25
See Remdesivir for more information.
Recommendations
• The Panel recommends using remdesivir 200 mg IV on Day 1, followed by remdesivir 100 mg
IV once daily on Days 2 and 3 in nonhospitalized adults with mild to moderate COVID-19 who
are at high risk of disease progression; treatment should be initiated as soon as possible and within
7 days of symptom onset (BIIa).
• Remdesivir should be administered in a setting where severe hypersensitivity reactions, such as
anaphylaxis, can be managed. Patients should be monitored during the infusion and observed for
at least 1 hour after infusion as clinically appropriate.
• For patients who started on IV remdesivir during hospitalization but were discharged from the
hospital in stable condition, with or without supplemental oxygen, the Panel recommends against
continuing remdesivir after discharge (AIIa).
• For considerations in pregnancy, see Remdesivir.
Bebtelovimab
Bebtelovimab is a recombinant neutralizing human mAb that binds to the spike protein of SARS-CoV-2.
In vitro data suggest that bebtelovimab has activity against a broad range of SARS-CoV-2 variants,
including the Omicron variant and its subvariants.26,27 The FDA issued an EUA for bebtelovimab for the
treatment of mild to moderate COVID-19 in nonhospitalized adults and pediatric patients aged ≥12 years
and weighing ≥40 kg who are at high risk of disease progression. However, to date, the clinical trial data
for bebtelovimab are limited to a single Phase 2, randomized, placebo-controlled trial in patients with
COVID-19 who were at low risk of progressing to severe disease. The trial showed no unexpected safety
events, and patients who received bebtelovimab had more rapid viral decay than those who received the
placebo.
Recommendations
• The Panel recommends using a single bebtelovimab 175 mg IV injection as an alternative
therapy in nonhospitalized adults with mild to moderate COVID-19 who are at high risk of
disease progression ONLY 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 7 days of symptom onset (CIII).
• Bebtelovimab should be administered in a setting where severe hypersensitivity reactions, such as
anaphylaxis, can be managed. Patients should be monitored for at least 1 hour after the injection.
• For considerations in pregnancy, see Anti-SARS-CoV-2 Monoclonal Antibodies.
Although data on the efficacy of bebtelovimab in reducing hospitalization and deaths for patients
with COVID-19 who were at high risk of disease progression are limited, this agent has a mechanism
of action that is similar to other anti-SARS-CoV-2 mAbs that have been shown to reduce rates of
hospitalization or death among high-risk patients in Phase 3 trials. Therefore, the in vitro data and Phase
2 clinical trial data for bebtelovimab, along with the clinical efficacy data for other anti-SARS-CoV-2
mAbs, support the use of bebtelovimab in high-risk patients with COVID-19 when preferred treatment
options are not available, feasible to use, or clinically appropriate.
Molnupiravir
Molnupiravir is the oral prodrug of beta-D-N4-hydroxycytidine (NHC), a ribonucleoside that has
exhibited antiviral activity against SARS-CoV-2 in vitro and in clinical trials.28-30 NHC uptake by viral
RNA-dependent RNA-polymerases results in viral mutations and lethal mutagenesis.29,31 The FDA
issued an EUA for molnupiravir for the treatment of mild to moderate COVID-19 in nonhospitalized
patients aged ≥18 years who are at high risk of disease progression and for whom alternative treatment
options are not available, feasible to use, or clinically appropriate. Molnupiravir has activity against
Omicron subvariants based on in vitro and animal studies.14,25,30,32
As a mutagenic ribonucleoside antiviral agent, there is a theoretical risk that molnupiravir will be
metabolized by the human host cell and incorporated into the host DNA, leading to mutations.
Immunomodulators
For Nonhospitalized Patients With Mild to Moderate COVID-19
The Panel recommends against the use of dexamethasone or other systemic glucocorticoids to
treat outpatients with mild to moderate COVID-19 who do not require hospitalization or supplemental
oxygen (AIIb). However, patients who are receiving dexamethasone or another corticosteroid for
other indications should continue therapy for their underlying conditions as directed by their health care
providers (AIII).
Medicare and FDA data show a significant increase in the number of prescriptions for systemic
corticosteroids among nonhospitalized patients with COVID-1935 despite a lack of safety and efficacy
data on the use of systemic corticosteroids in this setting. Systemic glucocorticoids may cause
COVID-19 Treatment Guidelines 65
References
1. 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.
2. 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.
3. 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.
4. Centers for Disease Control and Prevention. Underlying medical conditions associated with higher risk
for severe COVID-19: information for healthcare professionals. 2022. Available at: https://www.cdc.gov/
coronavirus/2019-ncov/hcp/clinical-care/underlyingconditions.html. Accessed September 16, 2022.
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://www.ncbi.nlm.nih.gov/pubmed/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. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for
bebtelovimab. 2022. Available at: https://www.fda.gov/media/156152/download.
8. Pillaiyar T, Manickam M, Namasivayam V, Hayashi Y, Jung SH. An overview of severe acute respiratory
a
For a list of risk factors, see the CDC webpage Underlying Medical Conditions Associated With Higher Risk for Severe
COVID-19.
b
Corticosteroids that are prescribed for an underlying condition should be continued.
c
Evidence suggests that the benefit of remdesivir is greatest when the drug is given early in the course of COVID-19 (e.g.,
within 10 days of symptom onset).
d
Conventional oxygen refers to oxygen supplementation that is not HFNC oxygen, NIV, MV, or ECMO.
e
If these patients progress to requiring HFNC oxygen, NIV, MV, or ECMO, the full course of remdesivir should still be
completed.
f
If PO baricitinib and IV tocilizumab are not available or not feasible to use, PO tofacitinib can be used instead of PO
baricitinib (BIIa), and IV sarilumab can be used instead of IV tocilizumab (BIIa).
g
Contraindications for the use of therapeutic anticoagulation in patients with COVID-19 include a platelet count <50 x
109/L, Hgb <8 g/dL, the need for dual antiplatelet therapy, bleeding within the past 30 days that required an ED visit or
hospitalization, a history of a bleeding disorder, or an inherited or active acquired bleeding disorder.
h
If a JAK inhibitor or an anti-IL-6 receptor mAb is not readily available, start dexamethasone while waiting for the
additional immunomodulator to be acquired. If neither of the other immunomodulators can be obtained, use
dexamethasone alone.
i
Clinicians may consider adding remdesivir to 1 of the recommended immunomodulator combinations in patients who
require HFNC oxygen or NIV, including immunocompromised patients. The Panel recommends against the use of
remdesivir without immunomodulators in these patients (AIIa).
Key: CDC = Centers for Disease Control and Prevention; ECMO = extracorporeal membrane oxygenation; ED =
emergency department; HFNC = high-flow nasal cannula; Hgb = hemoglobin; ICU = intensive care unit; IL = interleukin;
IV = intravenous; JAK = Janus kinase; mAb = monoclonal antibody; MV = mechanical ventilation; NIV = noninvasive
ventilation; the Panel = the COVID-19 Treatment Guidelines Panel; PO = oral; ULN = upper limit of normal
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 appears to be also 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 and endothelial
dysfunction.
Below is a summary of the rationale for the COVID-19 Treatment Guidelines Panel’s (the Panel)
recommendations on the therapeutic management of hospitalized patients with COVID-19. For dosing
information for each of the recommended drugs, please refer to Table 2d below. For detailed information
regarding the therapies and evidence from clinical trials that support 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
are at high risk of progressing to severe COVID-19 may benefit from antiviral therapy.
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 patients who do not
require supplemental oxygen and 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 this population 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.
Other studies have not shown a clinical benefit of remdesivir in this group of hospitalized patients with
COVID-19. In ACTT-1, remdesivir showed no significant benefit in hospitalized patients with mild to
moderate disease. However, only 13% of the study population did not require supplemental oxygen.
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. See Table 4a for more
information.
The aggregate data on using remdesivir to treat this population show a faster time to recovery in patients
who received remdesivir but no clear evidence of a survival benefit. Given the impact on reducing
progression, the Panel finds that the available data support a recommendation for using remdesivir in
hospitalized patients with COVID-19 who are at risk of progressing to severe disease. For information
on medical conditions that confer high risk, see the Centers for Disease Control and Prevention webpage
People With Certain Medical Conditions.
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.2 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.3
There are no data to support the use of other systemic corticosteroids in hospitalized patients with
COVID-19. However, patients who are receiving corticosteroid treatment for an underlying condition
should continue to receive corticosteroids. See Table 6a for more information.
COVID-19 Treatment Guidelines 72
Recommendation
• For patients with COVID-19 who only require minimal conventional oxygen, the Panel
recommends using remdesivir without dexamethasone (BIIa).
In these patients, the hyperinflammatory state for which corticosteroids might be most beneficial
may not yet be present or fully developed. 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.4 Evidence from ACTT-1 suggests
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, or 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.5 In the Solidarity trial, in which approximately
two-thirds of the patients received corticosteroids, remdesivir significantly reduced mortality among the
subset of patients who were receiving conventional or HFNC oxygen at enrollment.6 See Table 4a for
more information.
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, dexamethasone 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.2 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 6a for more information.
Recommendation
• The Panel recommends adding a second immunomodulatory drug (e.g., baricitinib or
tocilizumab) to dexamethasone in patients who have rapidly increasing oxygen needs and
systemic inflammation (BIIa).
COVID-19 Treatment Guidelines 73
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).
• Patients who do not meet the criteria for therapeutic heparin noted above, including pregnant
individuals, should receive a prophylactic dose of heparin, unless this drug is contraindicated
(AI); (BIII) for pregnant patients.
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.
The multiplatform ATTACC/ACTIV-4a/REMAP-CAP trial reported more organ support-free days
for patients in the therapeutic heparin arm than in the usual care arm, but there was no difference
between the arms in mortality or length of hospitalization.12 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 endpoint of ICU admission and initiation of NIV or mechanical ventilation),
but the therapeutic dose of heparin reduced 28-day mortality.13 In the HEP-COVID trial, venous
thromboembolism (VTE), arterial thromboembolism, and death by Day 30 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 by Day 30 between the arms.14
Recommendation
• If PO baricitinib and IV tocilizumab are not available or not feasible to use, PO tofacitinib can
be used instead of PO baricitinib (BIIa), and IV sarilumab can be used instead of IV tocilizumab
(BIIa).
When neither baricitinib nor tocilizumab is available or feasible to use, the JAK inhibitor tofacitinib
or the IL-6 inhibitor sarilumab may be used as alternative agents for baricitinib or tocilizumab,
respectively. Tofacitinib decreased the risk for respiratory failure or death in the STOP-COVID trial,17
and sarilumab reduced mortality and the duration of organ support to the same degree as tocilizumab in
the REMAP-CAP trial.8
Recommendation
• For hospitalized patients who require HFNC oxygen or NIV and have certain medical conditions,
the Panel recommends adding remdesivir to 1 of the recommended immunomodulator
combinations (CIIa).
Although clinical trial data have not established a clear benefit of using remdesivir in patients who
require HFNC oxygen or NIV, the Panel’s recommendation reflects the balance of 2 factors. First, given
that these patients are routinely treated with 2 immunomodulators to prevent or mitigate inflammatory-
mediated injury, these treatments may impair the patient’s antiviral response, and directly treating the
virus with remdesivir may help improve outcomes. In this context, some Panel members would add
remdesivir to treatments for immunocompromised patients who require HFNC oxygen or NIV. In
addition, clinicians may extend the course of remdesivir beyond 5 days in this population based on
clinical response. In the Solidarity trial, remdesivir had a modest but statistically significant effect on
reducing the risk of death or progression to mechanical ventilation; however, these effects could not be
evaluated separately for patients who required conventional oxygen supplementation and those who
required HFNC oxygen or NIV.18 See Table 4a for more information.
Recommendation
• The Panel recommends against the use of remdesivir without immunomodulators in hospitalized
patients who require HFNC oxygen or NIV (AIIa).
In the ACTT-1 trial, hospitalized patients with COVID-19 received remdesivir or placebo without
immunomodulators. In the subgroup of 193 patients who required high-flow oxygen or NIV at
enrollment, there was no difference in time to recovery between patients in the remdesivir arm and
patients in the placebo arm (recovery rate ratio 1.09; 95% CI, 0.76–1.57). A post hoc analysis did not
show a survival benefit for remdesivir at Day 29 (HR 1.02; 95% CI, 0.54–1.91).4
The Panel recommends against using remdesivir without immunomodulators in patients who require
HFNC oxygen or NIV because there is uncertainty regarding whether using remdesivir by itself confers
a clinical benefit in this subgroup. Patients who are taking remdesivir and then progress to requiring
HFNC oxygen or NIV should complete the course of remdesivir. If these patients are not already
receiving 1 of the recommended immunomodulator combinations as part of their treatment, they should
initiate immunomodulatory therapy.
Use of Anticoagulants
• For patients without an indication for therapeutic anticoagulation, the Panel recommends using a
prophylactic dose of heparin as VTE prophylaxis, unless a contraindication exists (AI); (BIII)
for pregnant patients.
Recommendation
• The Panel recommends using dexamethasone alone for the treatment of patients with COVID-19
COVID-19 Treatment Guidelines 77
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 are started on a therapeutic dose of heparin in a non-ICU setting for the
management of COVID-19 and then transferred 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 an intermediate dose (e.g., enoxaparin 1 mg/kg daily)
and a therapeutic dose of anticoagulation for VTE prophylaxis in critically ill patients with
COVID-19, except in a clinical trial (BI).
Patients who required mechanical ventilation or ECMO were included in the multiplatform
REMAP-CAP/ACTIV-4a/ATTACC and INSPIRATION trials that studied therapeutic doses and
intermediate doses of heparin, respectively.19,20 Because these studies reported no benefits to using
intermediate or 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.
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. RECOVERY Collaborative Group, Horby P, Lim WS, et al. 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.
COVID-19 Treatment Guidelines 79
Data from the Centers for Disease Control and Prevention demonstrate a lower incidence of
SARS-CoV-2 infection, severe disease, and death in children compared with adults.1-4 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 the percentage among hospitalized
adults with COVID-19.5-16
Risk factors for severe COVID-19 have been identified through observational studies and meta-analyses
primarily conducted before the availability of COVID-19 vaccines. Risk factors include having ≥1
severe comorbid conditions, such as medical complexity with respiratory technology dependence, a
neurologic condition resulting in impaired mucociliary clearance, obesity (particularly severe obesity),
severe underlying cardiac or pulmonary disease, or severely immunocompromised status. However,
pediatric data on risk factors for severe COVID-19 are generally more limited and provide lower
certainty than data for adults.
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, which is in
contrast to COVID-19, the acute, primarily respiratory illness due to infection with SARS-CoV-2.
There are no results available from clinical trials that evaluated treatments for COVID-19 in children,
and data from observational studies are limited. 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.
Because of these differences in epidemiology and disease severity, the effect sizes for children are
likely to be smaller than those observed in adults; therefore, to produce a beneficial outcome, 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 children are based largely on adult safety and efficacy data from clinical trials, the child’s risk of
disease progression, and expert opinion. In general, the older the child and the more severe the disease,
the more reasonable it is to follow treatment recommendations for adult patients with COVID-19.
The Panel’s recommendations for the management of children with COVID-19 or MIS-C are
summarized in the tables below. Table 3a provides recommendations for the therapeutic management
of nonhospitalized children with COVID-19. The Panel’s recommendations are stratified by age (per
the Food and Drug Administration Emergency Use Authorizations) and risk level. See Therapeutic
Management of Nonhospitalized Children With COVID-19 for more information. Table 3b includes a
framework to help clinicians evaluate the risk for severe COVID-19 based on patient conditions and
COVID-19 vaccination status.
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
Bebtelovimab is the only anti-SARS-CoV-2 mAb active against the current dominant circulating Omicron subvariants. In
nonhospitalized adults, bebtelovimab may be used as an alternative therapy when none of the preferred therapies (i.e.,
ritonavir-boosted nirmatrelvir, remdesivir) are available, feasible to use, or clinically appropriate.
e
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.
f
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; mAb = monoclonal antibody; the Panel = the COVID-19 Treatment Guidelines
Panel
a
Unvaccinated = individuals who are not eligible for COVID-19 vaccination or are <2 weeks from the final dose of
the primary series. Vaccinated with primary series = individuals who completed the primary series of 2 or 3 doses
(the current CDC term is “fully vaccinated”) and are >2 weeks after the final dose of the primary series but have not
received a booster, if they are eligible for a booster. Children aged <5 years are not currently eligible for booster doses.
Vaccinated and up to date = individuals who received the recommended booster dose(s) if eligible or have completed
the primary series but are not yet eligible for a booster. See the CDC for more information.
b
The degree of risk conferred by obesity in younger children is less clear than it is in older adolescents.
c
Includes tracheostomy or NIV.
d
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
a
F or example, for children who are severely immunocompromised regardless of COVID-19 vaccination status and those
who are unvaccinated and have additional risk factors for progression (see Therapeutic Management of Nonhospitalized
Children With COVID-19).
b
The clinical benefit of remdesivir is greatest if it is initiated within 10 days of symptom onset. Remdesivir should be
given for 5 days or until hospital discharge, whichever comes first.
c
Conventional oxygen refers to oxygen supplementation that is not high-flow oxygen, NIV, MV, or ECMO.
d
Patients who are receiving NIV or MV at baseline and require a substantial increase in baseline support should be treated
per the recommendations for patients requiring new NIV or MV.
e
Tofacitinib is an alternative if baricitinib is not available (BIII).
f
For children who started receiving remdesivir before admission to the ICU, the remdesivir should be continued to
complete the treatment course.
Key: ECMO = extracorporeal membrane oxygenation; ICU = intensive care unit; MV = mechanical ventilation; NIV =
noninvasive ventilation
References
1. Centers for Disease Control and Prevention. COVID-19 weekly cases and deaths per 100,000
population by age, race/ethnicity, and sex. 2022. Available at: https://covid.cdc.gov/covid-data-
tracker/#demographicsovertime. Accessed July 26, 2022.
2. Centers for Disease Control and Prevention. Demographic trends of COVID-19 cases and deaths in the US
reported to CDC. 2022. Available at: https://covid.cdc.gov/covid-data-tracker/#demographics. Accessed July
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.
• Observational studies describe associations between severe COVID-19 and the presence of ≥1 comorbid conditions,
including 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.
• Most children hospitalized for severe COVID-19 have not been fully vaccinated or are not eligible for COVID-19
vaccination.
• 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, but suspected or probable cases of vertical transmission
have been described.
• 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 comorbid conditions.
• 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.
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.
This section 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, COVID-19
refers to the acute, primarily respiratory illness due to infection with SARS-CoV-2. MIS-C refers to the
postinfectious inflammatory condition.
For information on the prevention, treatment, and management of severe complications of COVID-19 in
children, see:
• Prevention of SARS-CoV-2 Infection
• Therapeutic Management of Hospitalized Children With COVID-19
• Therapeutic Management of Hospitalized Pediatric Patients With Multisystem Inflammatory
Syndrome in Children (MIS-C) (With Discussion on Multisystem Inflammatory Syndrome in
Adults [MIS-A])
• Introduction to Critical Care Management of Children With COVID-19
Epidemiology
Data from the Centers for Disease Control and Prevention (CDC) demonstrate that SARS-CoV-2
infection and severe disease and death due to COVID-19 occur less often in children than in adults.1-4
Age
Among all children, infants and adolescents have the highest risk of COVID-19-related ICU admission
or death. From March 2020 to mid-August 2021, U.S. children aged <5 years had the highest cumulative
COVID-19-related hospitalization rates, followed closely by adolescents.27 Children aged 5 to 11 years
had the lowest hospitalization rates. From July to August 2021, when the Delta variant was the dominant
VOC, 25% of 713 children admitted to 6 U.S. hospitals were aged <1 year, 17% were aged 1 to 4 years,
20% were aged 5 to 11 years, and 38% were aged 12 to 17 years.28 From March 2020 to mid-June 2021,
26.5% of 3,116 U.S. children hospitalized for COVID-19 were admitted to an ICU.27
An individual patient data meta-analysis reported that patients aged <1 year and those aged 10 to 14
years had the highest risks of ICU admission and death among hospitalized children with COVID-19.29
Another meta-analysis reported that neonates, but not infants aged 1 to 3 months, had an increased risk
of severe COVID-19 compared with other pediatric age groups.30 When Omicron was the dominant
circulating VOC, hospitalization rates among children and adolescents were higher than when the Delta
VOC was dominant, and they were highest for children aged <5 years.26,31 However, the proportion of
hospitalized children requiring ICU admission was significantly lower when the Omicron VOC was
dominant.
Comorbidities
Several chronic conditions are prevalent in hospitalized children with COVID-19. When the Delta
variant was the dominant VOC in the United States, 68% of hospitalized children had ≥1 underlying
medical condition, such as obesity (32%), asthma or reactive airway disease (16%), or feeding tube
dependence (8%). Obesity was present in approximately a third of hospitalized children aged 5 to 11
years, 60% of whom had 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.28
Meta-analyses and observational studies identified risk factors for ICU admission, mechanical
ventilation, or death among hospitalized children with COVID-19.30,32,33 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 having
more severe chronic disease (e.g., active cancer treated within the previous 3 months or asthma with
hospitalization within the previous 12 months), when compared with less severe conditions, increased
the risk of critical COVID-19 or death. The CDC has additional information on the underlying
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, deaths associated with COVID-19 have 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.36,37
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-19.29 In the same
study, an individual patient data meta-analysis reported that the risk of COVID-19-related death was
greater for children with 1 chronic condition than for those with no comorbid conditions, and the risk
increased substantially as the number of comorbidities increased.
Distinguishing MIS-C from other febrile illnesses in the community setting remains challenging, but
the presence of persistent fever, multisystem manifestations, and laboratory abnormalities could help
early recognition.60 The clinical spectrum of hospitalized cases has included younger children with
COVID-19 Treatment Guidelines 92
Post-COVID Conditions
Persistent symptoms after COVID-19 have been described in adults and are an active area of research in
children, although data on the incidence of post-COVID sequelae in children are limited and somewhat
conflicting (see Clinical Spectrum of SARS-CoV-2 Infection).65-67 Cardiac imaging studies have
described myocardial injury in young athletes who had only mild disease;68 additional studies are needed
to identify long-term cardiac sequelae.
The reported clinical manifestations and duration of post-COVID conditions in children are highly
variable.69 Not all studies included controls without SARS-CoV-2 infection, which makes determining
the true incidence a challenge. The incidence of post-COVID symptoms appears to increase with age.
The most common symptoms reported include persistent fatigue, headache, shortness of breath, sleep
disturbances, and altered sense of smell.
Among children, health care utilization increases following COVID-19. A Norwegian study of 10,279
children with and 275,859 without SARS-CoV-2 infection reported that primary care visits for children
aged 6 to 15 years increased for up to 3 months after a positive SARS-CoV-2 test result when compared
with controls.66,70 For preschool-age children, visits increased for up to 6 months.
In a study of 6,804 adolescents in England, 30% of the 3,065 participants who tested positive for
SARS-CoV-2 infection reported ≥3 symptoms at a 3-month follow-up visit.71 Common symptoms
included tiredness (39%), headache (23%), and shortness of breath (23%). In the same study, only 16%
of the 3,739 participants who tested negative for SARS-CoV-2 infection reported ≥3 symptoms at the
3-month follow-up visit.
A study in Denmark examined persistent symptoms among 16,836 children with and 16,620 children
without SARS-CoV-2 infection. The number of children with SARS-CoV-2 infection who reported
symptoms that persisted for >4 weeks increased as age increased. Among the preschool-age children,
more children in the control arm than in the SARS-CoV-2 arm reported experiencing symptoms that
persisted for >4 weeks.72
References
1. Centers for Disease Control and Prevention. COVID-19 weekly cases and deaths per 100,000
population by age, race/ethnicity, and sex. 2022. Available at: https://covid.cdc.gov/covid-data-
tracker/#demographicsovertime. Accessed July 26, 2022.
2. Centers for Disease Control and Prevention. Provisional COVID-19 deaths: focus on ages 0–18 years. 2022.
Available at: https://data.cdc.gov/NCHS/Provisional-COVID-19-Deaths-Focus-on-Ages-0-18-Yea/nr4s-juj3.
Accessed July 26, 2022.
3. Centers for Disease Control and Prevention. Demographic trends of COVID-19 cases and deaths in the US
reported to CDC. 2022. Available at: https://covid.cdc.gov/covid-data-tracker/#demographics. Accessed July
26, 2022.
4. Centers for Disease Control and Prevention. COVID-NET laboratory-confirmed COVID-19 hospitalizations.
2022. Available at: https://covid.cdc.gov/covid-data-tracker/#covidnet-hospitalization-network. Accessed July
26, 2022.
5. Couture A, Lyons BC, Mehrotra ML, et al. Severe acute respiratory syndrome coronavirus 2 seroprevalence
and reported coronavirus disease 2019 Cases in US children, August 2020–May 2021. Open Forum Infect Dis.
2022;9(3):ofac044. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35198651.
6. Clarke KEN, Jones JM, Deng Y, et al. Seroprevalence of infection-induced SARS-CoV-2 antibodies—United
States, September 2021–February 2022. MMWR Morb Mortal Wkly Rep. 2022;71(17):606-608. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35482574.
7. 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.
8. CDC COVID-19 Response Team. Coronavirus disease 2019 in children—United States, February 12–April 2,
2020. MMWR Morb Mortal Wkly Rep. 2020;69(14):422-426. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32271728.
9. Cui X, Zhang T, Zheng J, et al. Children with coronavirus disease 2019: a review of demographic, clinical,
laboratory, and imaging features in pediatric patients. J Med Virol. 2020;92(9):1501-1510. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32418216.
10. Livingston E, Bucher K. Coronavirus disease 2019 (COVID-19) in Italy. JAMA. 2020;323(14):1335. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/32181795.
11. Tagarro A, Epalza C, Santos M, et al. Screening and severity of coronavirus disease 2019 (COVID-19) in
children in Madrid, Spain. JAMA Pediatr. 2020;Published online ahead of print. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32267485.
12. DeBiasi RL, Song X, Delaney M, et al. Severe coronavirus disease-2019 in children and young adults in the
Washington, DC, metropolitan region. J Pediatr. 2020;223:199-203. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32405091.
13. Chao JY, Derespina KR, Herold BC, et al. Clinical characteristics and outcomes of hospitalized and critically
ill children and adolescents with coronavirus disease 2019 at a tertiary care medical center in New York City. J
Pediatr. 2020;223:14-19. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32407719.
14. Swann OV, Holden KA, Turtle L, et al. Clinical characteristics of children and young people admitted to
hospital with COVID-19 in United Kingdom: prospective multicentre observational cohort study. BMJ.
2020;370:m3249. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32960186.
15. Gotzinger F, Santiago-Garcia B, Noguera-Julian A, et al. COVID-19 in children and adolescents in Europe: a
multinational, multicentre cohort study. Lancet Child Adolesc Health. 2020;4(9):653-661. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32593339.
COVID-19 Treatment Guidelines 94
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, which
is in contrast to multisystem inflammatory syndrome in children (MIS-C), a 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 nonhospitalized children are based largely on adult
safety and efficacy data from clinical trials (see Table 3a). No pediatric comparative studies have been
published; therefore, all 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,
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
Bebtelovimab is the only anti-SARS-CoV-2 mAb active against the current dominant circulating Omicron subvariants. In
nonhospitalized adults, bebtelovimab may be used as an alternative therapy when none of the preferred therapies (i.e.,
ritonavir-boosted nirmatrelvir, remdesivir) are available, feasible to use, or clinically appropriate.
e
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.
f
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; mAb = monoclonal antibody; the Panel = the COVID-19 Treatment Guidelines
Panel
a
Unvaccinated = individuals who are not eligible for COVID-19 vaccination or are <2 weeks from the final dose of
the primary series. Vaccinated with primary series = individuals who completed the primary series of 2 or 3 doses
(the current CDC term is “fully vaccinated”) and are >2 weeks after the final dose of the primary series but have not
received a booster, if they are eligible for a booster. Children aged <5 years are not currently eligible for booster doses.
Vaccinated and up to date = individuals who received the recommended booster dose(s) if eligible or have completed
the primary series but are not yet eligible for a booster. See the CDC for more information.
b
The degree of risk conferred by obesity in younger children is less clear than it is in older adolescents.
c
Includes tracheostomy or NIV.
d
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
Vaccination Status
Because COVID-19 vaccines are highly effective in preventing severe disease, individuals who are not
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 groups experience disproportionate rates
of COVID-19 hospitalization and are less likely to receive specific therapies.25-28 These factors may be
relevant when making clinical decisions about treatment.29,30 See Special Considerations in Children for
more information.
Remdesivir
Remdesivir is approved by the FDA for use in hospitalized and nonhospitalized pediatric patients aged
≥28 days and weighing ≥3.0 kg.33 Remdesivir is expected to be active against the Omicron variant of
COVID-19 Treatment Guidelines 103
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):e202004780. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33883243.
3. 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.
4. 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.
5. 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.
6. Havers FP, Whitaker M, Self JL, et al. Hospitalization of adolescents aged 12–17 years with laboratory-
confirmed COVID-19—COVID-NET, 14 states, March 1, 2020–April 24, 2021. MMWR Morb Mortal Wkly
Rep. 2021;70(23):851-857. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34111061.
7. Wolf J, Abzug MJ, Wattier RL, et al. Initial guidance on use of monoclonal antibody therapy for treatment
of coronavirus disease 2019 in children and adolescents. J Pediatric Infect Dis Soc. 2021;10(5):629-634.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/33388760.
8. Graff K, Smith C, Silveira L, et al. Risk factors for severe COVID-19 in children. Pediatr Infect Dis J.
2021;40(4):e137-e145. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33538539.
9. Fernandes DM, Oliveira CR, Guerguis S, et al. Severe acute respiratory syndrome coronavirus 2 clinical
syndromes and predictors of disease severity in hospitalized children and youth. J Pediatr. 2021;230:23-31.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/33197493.
10. Gotzinger F, Santiago-Garcia B, Noguera-Julian A, et al. COVID-19 in children and adolescents in Europe: a
multinational, multicentre cohort study. Lancet Child Adolesc Health. 2020;4(9):653-661. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32593339.
11. Kompaniyets L, Agathis NT, Nelson JM, et al. Underlying medical conditions associated with severe
COVID-19 illness among children. JAMA Netw Open. 2021;4(6):e2111182. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34097050.
12. 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.
13. Mukkada S, Bhakta N, Chantada GL, et al. Global characteristics and outcomes of SARS-CoV-2 infection
in children and adolescents with cancer (GRCCC): a cohort study. Lancet Oncol. 2021;22(10):1416-1426.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34454651.
14. CDC COVID-19 Response Team. Coronavirus disease 2019 in children—United States, February 12–April 2,
2020. MMWR Morb Mortal Wkly Rep. 2020;69(14):422-426. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32271728.
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
adult safety and efficacy data from clinical trials, 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, adult data are most applicable to older children with severe COVID-19 and predominantly
lower respiratory tract disease. Extrapolation of adult data to children with SARS-CoV-2 infection who
present with clinical syndromes common to other respiratory viruses (e.g., bronchiolitis, croup, asthma)
is challenging. No evidence indicates that these syndromes should be managed differently when caused
by SARS-CoV-2 infection. Clinical judgment is needed when applying these recommendations to
patients, particularly young children.
Dexamethasone
Dexamethasone was evaluated in the RECOVERY trial, which was an open-label, randomized trial
conducted in the United Kingdom.15 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 for 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 for 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 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).
There is evidence demonstrating 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.16 There are no COVID-19-specific
data to 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 and croup
triggered by SARS-CoV-2.
The use of dexamethasone for the treatment of severe COVID-19 in children who are profoundly
immunocompromised has not been evaluated, and there is a potential risk of harm. Therefore, the
COVID-19 Treatment Guidelines 111
Baricitinib
The Janus kinase inhibitor baricitinib was recently approved by the FDA for the treatment of COVID-19
in hospitalized adults. An FDA Emergency Use Authorization (EUA) 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.17
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; 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.
This effect was most pronounced in patients who received high-flow oxygen or NIV.18 In the ACTT-4
trial, 1,010 patients were randomized 1:1 to receive baricitinib plus remdesivir or dexamethasone
plus remdesivir. The study reported no difference between the arms for the outcome of mechanical
ventilation-free survival.19
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).20 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% proportional reduction in mortality
(rate ratio 0.80; 95% CI, 0.72–0.89; P < 0.0001). See Kinase Inhibitors: Janus Kinase Inhibitors
and Bruton’s Tyrosine 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.21-24
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 for 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 in 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.25 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 Kinase Inhibitors: Janus Kinase Inhibitors and Bruton’s
Tyrosine 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,26 and a Phase
3, double-blind, randomized, placebo-controlled trial investigated the efficacy of using tofacitinib in
children with JIA.27 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., with pediatric
infectious disease, pediatric rheumatology) when considering administering tofacitinib to hospitalized
children with COVID-19.
Tocilizumab
Tocilizumab is an interleukin (IL)-6 inhibitor that has received an FDA EUA 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.28 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. 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.29 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)
COVID-19 Treatment Guidelines 113
Sarilumab
Sarilumab, a monoclonal antibody that blocks IL-6 receptors, is not authorized by the FDA for the
treatment of COVID-19. Data evaluating 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).
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 Centers for Disease Control and Prevention’s (CDC) case definition for MIS-C includes
“an individual aged <21 years.”1 The recommendations in this section encompass this age group. There
are no randomized controlled trials that compare treatment approaches for MIS-C. However, data from
descriptive and observational comparative effectiveness studies are available to guide treatment for
MIS-C. For information on the clinical manifestations of MIS-C, see Special Considerations in Children.
Key: AE = adverse effect; BMP = blood mineral panel; CBC = complete blood count; FDA = Food and Drug Administration;
IBW = ideal body weight; IV = intravenous; IVIG = intravenous immunoglobulin; LFT = liver function test; MIS-C =
multisystem inflammatory syndrome in children; PO = orally; Scr = serum creatinine; SUBQ = subcutaneously
References
1. Centers for Disease Control and Prevention. Information for healthcare providers about multisystem
inflammatory syndrome in children (MIS-C). 2021. Available at:
https://www.cdc.gov/mis/mis-c/hcp/index.html. Accessed February 7, 2022.
2. Centers for Disease Control and Prevention. Multisystem inflammatory syndrome in adults (MIS-A) case
definition information for healthcare providers. 2021. Available at:
https://www.cdc.gov/mis/mis-a/hcp.html. Accessed February 7, 2022.
3. Morris SB, Schwartz NG, Patel P, et al. Case series of multisystem inflammatory syndrome in adults
associated with SARS-CoV-2 infection—United Kingdom and United States, March–August 2020. MMWR
Morb Mortal Wkly Rep. 2020;69(40):1450-1456. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33031361.
4. 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.
5. Belhadjer Z, Auriau J, Meot 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.
6. Toubiana J, Poirault C, Corsia A, et al. Kawasaki-like multisystem inflammatory syndrome in children during
the COVID-19 pandemic in Paris, France: prospective observational study. BMJ. 2020;369:m2094. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/32493739.
COVID-19 Treatment Guidelines 125
Summary Recommendations
Hemodynamics
• For adults with COVID-19 and shock, the COVID-19 Treatment Guidelines Panel (the Panel) recommends using
dynamic parameters, skin temperature, capillary refilling time, and/or lactate levels over static parameters to assess
fluid responsiveness (BIIa).
• For the acute resuscitation of adults with COVID-19 and shock, the Panel recommends using buffered/balanced
crystalloids over unbalanced crystalloids (BIIa).
• For the acute resuscitation of adults with COVID-19 and shock, the Panel recommends against the initial use of
albumin for resuscitation (BI).
• For adults with COVID-19 and shock, the Panel recommends norepinephrine as the first-line vasopressor (AI).
• For adults with COVID-19 and shock, the Panel recommends titrating vasoactive agents to target a mean arterial
pressure (MAP) of 60 to 65 mm Hg over higher MAP targets (BI).
• The Panel recommends against using hydroxyethyl starches for intravascular volume replacement in adult patients
with COVID-19 and sepsis or septic shock (AI).
• When norepinephrine is available, the Panel recommends against using dopamine for adult patients with COVID-19
and 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 target or adding vasopressin (up to 0.03 units/min) (BIIa) to
decrease norepinephrine dosage.
• 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 the resources to do so 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 (NIV) or intubation and mechanical ventilation should be initiated (BIIa).
• For adults with COVID-19 and acute hypoxemic respiratory failure 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
NIV (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).
COVID-19 can progress to critical illness, including hypoxemic respiratory failure, acute respiratory
distress syndrome (ARDS), septic shock, cardiac dysfunction, thromboembolic disease, hepatic and/
or renal dysfunction, central nervous system disease, and exacerbation of underlying comorbidities in
both adults and children. In addition, multisystem inflammatory syndrome in adults (MIS-A) can occur
several weeks or months after SARS-CoV-2 infection, which can lead to critical illness.
Many of the initial recommendations for the management of critically ill adults with COVID-19 in
these Guidelines were extrapolated from experience with other causes of sepsis and respiratory failure.1
However, there is now a rapidly growing body of evidence regarding the management of critically ill
patients with COVID-19.
Treating patients with COVID-19 in the intensive care unit (ICU) often requires managing underlying
illnesses or COVID-19-related morbidities. As with any patient who is admitted to the ICU, clinicians
also need to focus on preventing ICU-related complications.
Acknowledgments
The Surviving Sepsis Campaign (SSC), an initiative supported by SCCM and the European Society of
Intensive Care Medicine, issued 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
COVID-19 Treatment Guidelines Panel (the Panel) has based the recommendations in this section 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. 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.
3. Sinha P, Matthay MA, Calfee CS. Is a “cytokine storm” relevant to COVID-19? JAMA Intern Med.
2020;180(9):1152-1154. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32602883.
4. Morris SB, Schwartz NG, Patel P, et al. Case series of multisystem inflammatory syndrome in adults
associated with SARS-CoV-2 infection—United Kingdom and United States, March-August 2020. MMWR
Morb Mortal Wkly Rep. 2020;69(40):1450-1456. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33031361.
5. Sansone M, Studahl M, Berg S, Gisslen M, Sundell N. Severe multisystem inflammatory syndrome
(MIS-C/A) after confirmed SARS-CoV-2 infection: a report of four adult cases. Infect Dis (Lond). 2022:1-6.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/35034549.
6. Martins A, Policarpo S, Silva-Pinto A, et al. SARS-CoV-2-related multisystem inflammatory syndrome in
adults. Eur J Case Rep Intern Med. 2021;8(11):003025. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34912751.
7. Belay ED, Godfred Cato S, Rao AK, et al. Multisystem inflammatory syndrome in adults after SARS-CoV-2
infection and COVID-19 vaccination. Clin Infect Dis. 2021. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34849680.
8. Zou F, Qian Z, Wang Y, Zhao Y, Bai J. Cardiac injury and COVID-19: a systematic review and meta-analysis.
CJC Open. 2020;2(5):386-394. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32838255.
9. Nishiga M, Wang DW, Han Y, Lewis DB, Wu JC. COVID-19 and cardiovascular disease: from basic
mechanisms to clinical perspectives. Nat Rev Cardiol. 2020;17(9):543-558. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32690910.
10. Llitjos JF, Leclerc M, Chochois C, et al. High incidence of venous thromboembolic events in anticoagulated
severe COVID-19 patients. J Thromb Haemost. 2020;18(7):1743-1746. Available at:
COVID-19 Treatment Guidelines 134
Most of the hemodynamic recommendations below are similar to those previously published in the
Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016.
Ultimately, adult patients with COVID-19 who require fluid resuscitation or hemodynamic management
of shock should be treated and managed identically to adult patients with septic shock.1
Recommendation
• For adults with COVID-19 and shock, the COVID-19 Treatment Guidelines Panel (the Panel)
recommends using dynamic parameters, skin temperature, capillary refilling 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),2 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). Dynamic parameters used in these trials included stroke volume variation (SVV), pulse
pressure variation (PPV), and stroke volume change with passive leg raise or fluid challenge. Passive leg
raising, followed by PPV and SVV, appears to predict fluid responsiveness with the greatest accuracy.3
The static parameters included components of early goal-directed therapy (e.g., central venous pressure,
mean arterial pressure [MAP]).
Resuscitation of patients with shock who do not have COVID-19 based on serum lactate levels has
been summarized in a systematic review and meta-analysis of seven randomized clinical trials (n =
1,301). Compared with central venous oxygen saturation-guided therapy, early lactate clearance-directed
therapy 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).4
Recommendation
• For the acute resuscitation of adults with COVID-19 and shock, the Panel recommends using
buffered/balanced crystalloids over unbalanced crystalloids (BIIa).
Rationale
A pragmatic randomized trial compared the use of balanced and unbalanced crystalloids for intravenous
(IV) fluid administration in critically ill adults without COVID-19 (n = 15,802). The rate of the
composite outcome of death, new renal-replacement therapy, or persistent renal dysfunction was
lower in the balanced crystalloids group than in the unbalanced crystalloids group (OR 0.90; 95% CI,
0.82–0.99; P = 0.04).5 A secondary analysis compared outcomes in a subset of patients with sepsis (n
= 1,641). Compared to treatment with unbalanced crystalloids, treatment with balanced crystalloids
resulted in fewer deaths (aOR 0.74; 95% CI, 0.59–0.93; P = 0.01) and more vasopressor-free and renal
replacement-free days.6 A subsequent meta-analysis of 21 non-COVID-19 randomized controlled trials
(n = 20,213) that included the pragmatic trial cited above compared balanced crystalloids to 0.9% saline
Recommendation
• For the acute resuscitation of adults with COVID-19 and shock, the Panel recommends against
the initial use of albumin for resuscitation (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 groups.8 In contrast, a meta-analysis of 17 non-COVID-19 randomized
controlled trials (n = 1,977) that compared the use of albumin to crystalloids specifically in patients with
sepsis observed a reduction in mortality among the patients who received albumin (OR 0.82; 95% CI,
0.67–1.0; P = 0.047).9 Given the higher cost of albumin and the lack of a definitive clinical benefit, the
Panel recommends against the routine use of albumin for initial acute resuscitation of patients with
COVID-19 and shock (BI).
Recommendation
• For adults with COVID-19 and shock, the Panel recommends norepinephrine as the first-choice
vasopressor (AI).
Rationale
Norepinephrine increases MAP due to its vasoconstrictive effects, with little change in heart rate
and less increase in stroke volume compared to dopamine. Dopamine increases MAP and cardiac
output, primarily due to an increase 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.10 It may also influence the endocrine
response via the hypothalamic pituitary axis and have immunosuppressive effects.11 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
(RR 0.89; 95% CI, 0.81–0.98) and a lower risk of arrhythmias (RR 0.48; 95% CI, 0.40–0.58) than
dopamine use.12 Although the beta-1 activity of dopamine would be useful in patients with myocardial
dysfunction, the greater risk of arrhythmias limits its use.13,14
Recommendation
• For adults with COVID-19 and shock, the Panel recommends titrating vasoactive agents to target a
MAP of 60 to 65 mm Hg, over higher MAP targets (BI).
Rationale
A recent individual patient-data meta-analysis of two, non-COVID-19 randomized controlled trials (n
= 894) comparing higher versus lower blood pressure targets for vasopressor therapy in adult patients
with shock reported no significant difference between the patients in the higher and lower target groups
in 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).15
The risk of arrhythmias was increased in patients allocated to the higher target group (OR 2.50; 95%
References
1. Rhodes A, Evans LE, Alhazzani W, et al. Surviving Sepsis Campaign: international guidelines for
management of sepsis and septic shock: 2016. Crit Care Med. 2017;45(3):486-552. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28098591.
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.
3. Bentzer P, Griesdale DE, Boyd J, MacLean K, Sirounis D, Ayas NT. Will this hemodynamically unstable
patient respond to a bolus of intravenous fluids? JAMA. 2016;316(12):1298-1309. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27673307.
4. Pan J, Peng M, Liao C, Hu X, Wang A, Li X. Relative efficacy and safety of early lactate clearance-guided
therapy resuscitation in patients with sepsis: a meta-analysis. Medicine (Baltimore). 2019;98(8):e14453.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/30813144.
5. Semler MW, Self WH, Wanderer JP, et al. Balanced crystalloids versus saline in critically ill adults. N Engl J
The COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations in this section were
informed by the recommendations in the Surviving Sepsis Campaign guidelines for managing sepsis and
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 of <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 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 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
demonstrate 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 to 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;Published online
ahead of print. 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, 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.
6. 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
and noninvasive positive pressure ventilation. A systematic review and meta-analysis. Am J Emerg Med.
2018;36(2):226-233. Available at: https://www.ncbi.nlm.nih.gov/pubmed/28780231.
7. Grieco DL, Menga LS, Cesarano M, et al. Effect of helmet noninvasive ventilation vs high-flow nasal oxygen
on days free of respiratory support in patients with COVID-19 and moderate to severe hypoxemic respiratory
COVID-19 Treatment Guidelines 147
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 intensive
care unit patients, 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
There are no clinical trials that have evaluated the use of empiric broad-spectrum antibiotics in patients
with severe or critical COVID-19 or other coronavirus infections. Routine, empiric use of antibiotics in
patients with severe or critical COVID-19 is not recommended (BIII); this recommendation is intended
to mitigate the unintended consequences of side effects and resistance. The use of antibiotics 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 antibiotics 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 central line-associated bloodstream infection). It is unclear whether using the corticosteroids or other
immunomodulatory agents that are recommended in the Guidelines should alter such approaches.
Immune-Based Therapy
See the Immunomodulators section for recommendations on the use of immunomodulators.
Adjunctive Therapy
Recommendations regarding the use of adjunctive therapies in critical care settings, including
antithrombotic therapy and vitamin C, can be found in Antithrombotic Therapy in Patients With
COVID-19, Therapeutic Management of Hospitalized Adults With COVID-19, and Vitamin C.
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
and refractory hypoxemia.
Rationale
ECMO has been used as a short-term rescue therapy in patients with acute respiratory distress syndrome
(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 a similar mortality to patients in a 2018 randomized
study who did not have COVID-19 but who had ARDS and received ECMO.3
However, a recent analysis reported worse outcomes over time among patients who required ECMO for
COVID-19.11 The analysis used data from 4,812 patients in the international Extracorporeal Life Support
Organization (ELSO) Registry who had COVID-19 and who received ECMO in 2020. 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.
Without data from controlled trials that have evaluated the use of ECMO in patients with COVID-19
and hypoxemic respiratory failure (e.g., ARDS), the benefits of ECMO cannot be clearly defined for this
patient population.
Clinicians who are interested in using ECMO should try to enter their patients into clinical trials
or clinical registries so that more informative data can be obtained. More information on the use of
ECMO in patients with COVID-19 can be found on ELSO’s Extracorporeal Membrane Oxygenation in
COVID-19 website and ClinicalTrials.gov.
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:
COVID-19 may lead to critical illness in children, including hypoxemic respiratory failure, acute
respiratory distress syndrome (ARDS), 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, management should follow the principles of pediatric critical care usually applied to non-
COVID-19-related illness, such as the Pediatric Acute Lung Injury Consensus Conference (PALICC)
recommendations and the Surviving Sepsis Campaign International Guidelines for the Management
of Septic Shock and Sepsis-Associated Organ Dysfunction in Children. 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
COVID-19 Treatment Guidelines 154
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
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 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. The studies found that the use of the protocols did not
demonstrate a significant benefit on outcomes, such as the duration of ventilation. 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 suggesting 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
COVID-19 Treatment Guidelines 155
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 non-COVID-19-specific treatment guidelines, such as the
Surviving Sepsis Campaign International Guidelines for the Management of Septic Shock and Sepsis-
Associated Organ Dysfunction in Children,22 the PALICC 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.
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 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 COVID-19 Treatment Guidelines Panel
(the Panel) recommends a target mean arterial pressure (MAP) between the fifth and fiftieth, or
greater than the fiftieth, percentiles for age (AIII).
Rationale
There are no clinical trials that 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. Therefore, for children with COVID-19 or MIS-C, clinicians should
use the same approach used for children without COVID-19 and target a MAP between the fifth and
fiftieth, or greater than the fiftieth, percentiles for age. When MAP cannot be reliably measured, systolic
blood pressure is a reasonable alternative.2
Recommendation
• The Panel recommends that, when available, a combination of serial clinical assessments; cardiac
ultrasound or echocardiography; and/or 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 non-COVID-19-related sepsis 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 use of
cardiac ultrasound in children with COVID-19 and MIS-C are limited to reports from case series.9
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 (CIIb).
Rationale
No published clinical trials directly compare balanced/buffered crystalloids with 0.9% saline
administered to children with sepsis of any etiology, although an international randomized trial is
underway (ClinicalTrials.gov Identifier NCT04102371). Two observational studies using administrative
data compared the use of balanced/buffered crystalloids to 0.9% saline in propensity-matched cohorts
of children with non-COVID-19-related severe sepsis or septic shock. 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. The study demonstrated no differences between the arms for 30-day
mortality or frequency of acute kidney injury.13
The other study compared patients receiving only balanced fluids with those receiving only 0.9%
saline. 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.14 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 or 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.
Rationale
Use of vasoactive infusions should be considered for children with shock due to COVID-19 if signs of
COVID-19 Treatment Guidelines 159
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 use of inodilators in children with 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 non-COVID-19-related sepsis, 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).
• The Panel recommends against using hydroxyethyl starches for intravascular volume
replacement in children with COVID-19 or MIS-C and sepsis or septic shock (AIII).
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):e17-26. 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 e141-144. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26711848.
13. Weiss SL, Keele L, Balamuth F, et al. Crystalloid fluid choice and clinical outcomes in pediatric sepsis: a
matched retrospective cohort study. J Pediatr. 2017;182:304-310 e310. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28063688.
The COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations in this section were
informed by recommendations from the Surviving Sepsis Campaign’s guidelines for managing adult
sepsis, pediatric sepsis, and COVID-19, as well as by recommendations from the 2015 Pediatric Acute
Lung Injury Consensus Conference (PALICC).
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 SpO2 index ≥12.3), an SpO2 <92% can be considered to minimize
exposure to a high fraction of inspired oxygen (FiO2), but prolonged periods of 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 2015 PALICC 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 recent 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, there was significant
heterogeneity across the included studies for populations, definitions of hyperoxia, and the timing of
assessments for mortality outcomes. For children with severe PARDS (i.e., those with an oxygenation
index ≥16 or SpO2 index ≥12.3),1 an SpO2 <92% can be considered to minimize exposure to a high FiO2.
Although no evidence clearly identifies a safe minimum SpO2 in children, prolonged exposure to SpO2
<88% should be avoided. When 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 adult observational studies that
also 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 children with PARDS who are
critically ill 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 PALICC recommend the use of NIV for children with respiratory failure
who have no indication for intubation.12,13
Furthermore, the response to NIV, particularly for children with more severe hypoxemia or high work
of breathing, should be gauged early (within the first several hours). 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.12
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 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 (adjusted HR 0.79; 95% CI, 0.54–1.16), although the results were not statistically significant.15
The other trial demonstrated that children treated with bubble CPAP ventilation had a lower risk of
mortality than children who received low-flow oxygen (relative risk 0.25; 95% CI, 0.07–0.89; P =
0.02).16 The results also indicated that for the composite outcome of treatment failure, there was no
difference between the use of HFNC oxygen and bubble CPAP (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 (M-WCAS) 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 (PICU).
Rationale
There are no high-quality pediatric data evaluating the effect of awake prone positioning on clinical
outcomes in children with COVID-19 or non-COVID-19-related illness. Awake prone positioning
may be considered for older children and adolescents (see Oxygenation and Ventilation for Adults). In
addition, pediatric clinicians should consider a child’s developmental stage and ability to comply with
the protocols for awake prone positioning.
Rationale
To optimize the safety of patients and health care workers and maximize first-attempt success,
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 for children with normal chest
wall compliance and ≤32 cm H2O for those with impaired chest wall compliance (AIII).
• The Panel recommends using a higher positive end-expiratory pressure (PEEP) strategy (i.e.,
10–15 cm H2O or higher in patients with severe PARDS) over a lower PEEP strategy, titrated
based on observed responses in oxygenation, hemodynamics, and respiratory system compliance
(BIIb).
• The Panel recommends permissive hypercapnia (e.g., pH 7.15–7.30), if needed, 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 against the routine use of inhaled nitric oxide (AIII).
Rationale
There is no evidence that ventilator management of children with PARDS due to COVID-19
should differ from ventilator management of patients with PARDS due to other causes. The Panel’s
recommendations are derived from the 2015 PALICC recommendations.1,3 Since the publication of the
PALICC recommendations, no randomized trials have provided significant new evidence, although
some observational data support some of the PALICC recommendations.
A large observational study conducted in 71 international PICUs reported that for patients with mild
to moderate acute respiratory distress syndrome (ARDS), less adherence to the recommended VT of 5
mL/kg to 8 mL/kg (or 3 mL/kg 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 ventilator protocol that includes suggested low PEEP/high FiO2
levels.26 The protocol suggests that for patients receiving FiO2 ≥0.6, a PEEP level of ≥10 cm H2O would
be implemented, which aligns with recommendations from PALICC. Two observational studies have
reported better clinical outcomes associated with use of the suggested (or higher) PEEP levels compared
to lower PEEP levels.25,27 The multicenter studies, which included nearly 1,500 pediatric patients with
ARDS, demonstrated that PEEP levels lower than those recommended by the ARDS Network were
associated with higher mortality.
Rationale
There is no evidence that fluid management in children 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 PALICC recommendation.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.29-31
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. 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, an increasing cumulative fluid
balance on Day 3 was associated with fewer ventilator-free days, but no association with mortality was
detected.29
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 (for 100
mL/kg on Day 5, OR 1.34; 95% CI, 1.11–1.61) after adjusting for oxygenation index, the number
of nonpulmonary organ failures, immunocompromised status, and vasopressor scores. Also, greater
cumulative fluid balance on Days 4 to 7 was associated with a lower probability of successful extubation
by Day 28.31 Collectively, the findings from these pediatric observational studies demonstrate the
potential harm of fluid overload in children 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.32 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 intensive care unit when compared with the liberal strategy arm.
However, no analysis of data from prospective pediatric trials delineates a causal relationship between a
specific, protocolized fluid management strategy, or the timing of such a strategy, and clinical outcomes.
Therefore, an individualized fluid management approach that is titrated to maintain intravascular volume
while preventing excessive positive fluid balance, as suggested by the 2015 PALICC recommendation,
is appropriate.1
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 PALICC recommendation.1 Since the publication of the 2015 PALICC
recommendation, no new data support significant changes to the recommendation.
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 inhaled nitric oxide as a rescue therapy; if no rapid improvement in
oxygenation is observed, 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 to 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 children 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 largely based on PALICC recommendations.1 Since the publication of the
2015 PALICC recommendations, many new trials evaluating these practices have been conducted.
One randomized controlled trial and 2 propensity-matched, observational studies have evaluated the use
of inhaled nitric oxide in patients with PARDS since the publication of the PALICC recommendations.
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.28
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. These results have been
corroborated by observational studies, which also reported more ventilator-free days for patients who
received inhaled nitric oxide.33,34 Although the evidence is insufficient to recommend the use of inhaled
nitric oxide for all patients with ARDS, in cases of severe hypoxemia, it can be considered as a rescue
therapy to potentially avoid the use of ECMO.
References
1. Pediatric Acute Lung Injury Consensus Conference Group. Pediatric acute respiratory distress syndrome:
consensus recommendations from the Pediatric Acute Lung Injury Consensus Conference. Pediatr Crit Care
Med. 2015;16(5):428-439. Available at: https://www.ncbi.nlm.nih.gov/pubmed/25647235.
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. Rimensberger PC, Cheifetz IM, Pediatric Acute Lung Injury Consensus Conference Group. Ventilatory
support in children with pediatric acute respiratory distress syndrome: proceedings from the Pediatric Acute
Lung Injury Consensus Conference. Pediatr Crit Care Med. 2015;16(5 suppl 1):S51-S60. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26035364.
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
accuracy in hospitalized children. JAMA Netw Open. 2022;5(3):e224584. Available at:
COVID-19 Treatment Guidelines 169
Recommendation
• The COVID-19 Treatment Guidelines Panel (the 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 are influenced
by the etiology and duration of 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 No randomized
trials evaluate the efficacy or benefit of ECMO for hypoxemic respiratory failure in children without
COVID-19 beyond the neonatal period. In an observational study of 122 children with severe pediatric
acute respiratory distress syndrome (PARDS), 90-day mortality for children treated with ECMO and for
those supported without ECMO was similar (25% vs. 30%).5
The Pediatric Acute Lung Injury Consensus Conference recommends considering ECMO for patients
with severe PARDS from reversible causes or for children who are candidates for lung transplantation.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 of evidence, to use venovenous ECMO for children with PARDS and refractory hypoxemia.7
Venoarterial ECMO has been used successfully for the treatment of refractory shock in children,
although no trials evaluate this approach, and the potential benefits must be weighed against risks of
bleeding or thromboembolic events.8-10 The Surviving Sepsis Campaign guidelines for children issued a
weak recommendation, based on very low quality of evidence, for use of venoarterial ECMO in children
with shock that is refractory to all other treatments; however, a standardized definition of refractory
shock in children is not available.7
Studies evaluating data on the use of ECMO in children with COVID-19 and MIS-C are limited to case
reports and case series.11-13 A publicly available registry for pediatric patients with COVID-19 on ECMO
is maintained by the multinational Extracorporeal Life Support Organization (ELSO). In-hospital
mortality at 90 days was about 30%, which is similar to reports from non-COVID-19 ECMO cohorts.14,15
ELSO has published guidelines for use of ECMO in COVID-19.16 In general, ECMO candidacy for
children with COVID-19 or MIS-C should be assessed using criteria similar to those used for other
causes of severe respiratory failure or shock. Cannulation approaches and management principles should
follow published international guidelines and local protocols for non-COVID-19 patients.
Pediatric clinicians should consider entering patients into clinical trials or registries to inform future
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, Sebastiao 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. Dalton HJ, Macrae DJ, Pediatric Acute Lung Injury Consensus Conference Group. Extracorporeal support in
children with pediatric acute respiratory distress syndrome: proceedings from the Pediatric Acute Lung Injury
Consensus Conference. Pediatr Crit Care Med. 2015;16(5 Suppl 1):S111-S117. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26035361.
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
Summary Recommendations
Remdesivir is the only antiviral drug that is approved by the Food and Drug Administration (FDA) for the treatment
of COVID-19. Ritonavir-boosted nirmatrelvir (Paxlovid), molnupiravir, and certain anti-SARS-CoV-2 monoclonal
antibodies (mAbs) have received Emergency Use Authorizations from the FDA for the treatment of COVID-19.
This section focuses on the COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations for using
small-molecule antiviral drugs to treat COVID-19. These recommendations are based on the available data. For
recommendations and information regarding the use of anti-SARS-CoV-2 mAbs, see Anti-SARS-CoV-2 Monoclonal
Antibodies.
Recommendations for Treating Nonhospitalized Adults
• 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 the following anti-SARS-CoV-2 therapies as alternative treatments for COVID-19. These drugs
should ONLY be used when neither of the preferred treatments are available, feasible to use, or clinically appropriate.
These drugs are listed in alphabetical order:
• Bebtelovimab (CIII)
• Molnupiravir (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).
• See Therapeutic Management of Nonhospitalized Adults With COVID-19 for detailed recommendations.
Recommendations for Treating Nonhospitalized Children
• See Therapeutic Management of Nonhospitalized Children With COVID-19 for the Panel’s recommendations on the
use of antiviral therapy in nonhospitalized children according to age and the risk for progression to severe COVID-19.
Recommendations for Treating Hospitalized Patients
• See Therapeutic Management of Hospitalized Adults With COVID-19 and Therapeutic Management of Hospitalized
Children With COVID-19 for the Panel’s recommendations on using remdesivir with or without immunomodulators in
certain hospitalized patients.
Antiviral Drugs 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:
• Interferons for nonhospitalized patients (AIIa)
• Interferon alfa or lambda for hospitalized patients (AIIa)
• Ivermectin (AIIa)
• Nitazoxanide (BIIa)
• The Panel recommends against the use of the following drugs for the treatment of COVID-19:
• Chloroquine or hydroxychloroquine and/or azithromycin for hospitalized (AI) and nonhospitalized patients (AIIa)
• Lopinavir/ritonavir and other HIV protease inhibitors for hospitalized (AI) and nonhospitalized patients (AIII)
• Systemic interferon beta for hospitalized patients (AI)
Antiviral Therapy
Because SARS-CoV-2 replication leads to many of the clinical manifestations of COVID-19, antiviral
therapies are being investigated for the treatment of COVID-19. These drugs prevent viral replication
through various mechanisms, including blocking SARS-CoV-2 entry, inhibiting the activity of
SARS-CoV-2 3-chymotrypsin-like protease (3CLpro) and RNA-dependent RNA polymerase (RdRp),
and causing lethal viral mutagenesis.1-3 Because viral replication may be particularly active early in the
course of COVID-19, antiviral therapy may have the greatest impact before the illness progresses to the
hyperinflammatory state that can characterize the later stages of disease, including critical illness.4 For
this reason, it is necessary to understand the role of antiviral medications in treating mild, moderate,
severe, and critical illness in order to optimize treatment for people with COVID-19.
The following sections describe the underlying rationale for using different antiviral medications,
provide the COVID-19 Treatment Guidelines Panel’s recommendations for using these medications to
treat COVID-19, and summarize the existing clinical trial data. Additional antiviral therapies will be
added to this section of the Guidelines as new evidence emerges.
References
1. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for
molnupiravir. 2022. Available at: https://www.fda.gov/media/155054/download.
2. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for Paxlovid.
2022. Available at: https://www.fda.gov/media/155050/download.
3. Remdesivir (Veklury) [package insert]. Food and Drug Administration. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/214787Orig1s015lbl.pdf.
4. Siddiqi HK, Mehra MR. COVID-19 illness in native and immunosuppressed states: a clinical-therapeutic
staging proposal. J Heart Lung Transplant. 2020;39(5):405-407. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32362390.
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 activity against SARS-CoV-2.1 In a rhesus macaque model of SARS-CoV-2
infection, remdesivir treatment was initiated soon after inoculation; the remdesivir-treated animals had
lower virus levels in the lungs and less lung damage than the control animals.2 Remdesivir is expected to
be active against the Omicron variant and its subvariants.3-6
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 high-risk,
nonhospitalized patients with mild to moderate COVID-19, 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.7 See Table 4d for more information.
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 remdesivir in 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 remdesivir with or without
immunomodulators in certain hospitalized patients, see Therapeutic Management of Hospitalized Adults
With COVID-19.
There are no data on using combinations of antiviral therapies or combinations of antiviral therapies and
anti-SARS-CoV-2 monoclonal antibodies for the treatment of COVID-19. Clinical trials are needed to
determine the role of combination therapy in certain patients.
Drug-Drug Interactions
Currently, no clinical drug-drug interaction studies of remdesivir have been conducted. In vitro,
remdesivir is a minor substrate of cytochrome P450 (CYP) 3A4 and a substrate of the drug transporters
organic anion transporting polypeptide (OATP) 1B1 and P-glycoprotein. It is also an inhibitor of
CYP3A4, OATP1B1, OATP1B3, and multidrug and toxin extrusion protein (MATE) 1.7
Minimal to no reduction in remdesivir exposure is expected when remdesivir is coadministered with
dexamethasone, according to information provided by Gilead Sciences (written communication, July
2020).
See Table 4d for more information.
Considerations in Pregnancy
Remdesivir should be offered to pregnant individuals if it is indicated.
While pregnant patients 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
patients have been reassuring. Among 86 pregnant and postpartum patients who were hospitalized with
severe COVID-19 and who received remdesivir through a compassionate use program, the therapy was
COVID-19 Treatment Guidelines 179
Considerations in Children
Please see Special Considerations in Children, Therapeutic Management of Nonhospitalized Children
With COVID-19, and Therapeutic Management of Hospitalized Children With COVID-19.
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. Williamson BN, Feldmann F, Schwarz B, et al. Clinical benefit of remdesivir in rhesus macaques infected with
SARS-CoV-2. Nature. 2020;585(7824):273-276. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32516797.
3. Vangeel L, Chiu W, De Jonghe S, et al. Remdesivir, molnupiravir and nirmatrelvir remain active against
SARS-CoV-2 Omicron and other variants of concern. Antiviral Res. 2022;198:105252. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35085683.
4. Gilead Sciences. Gilead statement on Veklury (remdesivir) and the SARS-CoV-2 Omicron variant. 2021.
Available at: https://www.gilead.com/news-and-press/company-statements/gilead-statement-on-veklury-
remdesivir-and-the-sars-cov-2-omicron-variant. Accessed June 7, 2022.
5. Takashita E, Kinoshita N, Yamayoshi S, et al. Efficacy of antiviral agents against the SARS-CoV-2 Omicron
subvariant BA.2. N Engl J Med. 2022;386(15):1475-1477. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35263535.
6. Takashita E, Yamayoshi S, Simon V, et al. Efficacy of antibodies and antiviral drugs against Omicron
BA.2.12.1, BA.4, and BA.5 subvariants. N Engl J Med. 2022;387(5):468-470. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35857646.
7. Remdesivir (Veklury) [package insert]. Food and Drug Administration. 2020. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/214787Orig1s000lbl.pdf.
8. 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.
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. Studies of hospitalized
patients are listed first, followed by 1 study of nonhospitalized patients.
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. The Food and Drug Administration (FDA) issued an
Emergency Use Authorization (EUA) for ritonavir-boosted nirmatrelvir on December 22, 2021, for the
treatment of COVID-19.3
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
(AIIa) and pediatric patients aged ≥12 years and weighing ≥40 kg (BIII) with mild to moderate
COVID-19 who are at high risk of disease progression.4 Treatment should be initiated as soon as
possible and within 5 days of symptom onset.
• For recommendations and a discussion on using ritonavir-boosted nirmatrelvir in nonhospitalized
children with COVID-19, see Therapeutic Management of Nonhospitalized Children With
COVID-19.
• Ritonavir-boosted nirmatrelvir may be used in patients who are hospitalized for a diagnosis other
than COVID-19, provided they have mild to moderate COVID-19, are at high risk of progressing to
severe disease, and are within 5 days of symptom onset.
Ritonavir-boosted nirmatrelvir has significant drug-drug interactions, primarily due to the ritonavir
component of the combination. 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.
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, and 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 Ontario COVID-19 Science Advisory Table
• The FDA EUA fact sheet and checklist for ritonavir-boosted nirmatrelvir
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.
Clinical Data
The EPIC-HR study was a multinational randomized trial that compared 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. Eligible patients were randomized within
5 days of symptom onset, were unvaccinated against COVID-19, and had at least 1 risk factor for
progression to severe disease.5 Patients were excluded if they used medications that were either highly
dependent upon CYP3A4 for clearance or strong inducers of CYP3A4.
A total of 2,246 patients enrolled in the trial. The mean age was 46 years, 51% of the patients were men,
and 72% were White. Forty-seven percent of the patients tested negative for SARS-CoV-2 antibodies,
and 66% started study treatment within 3 days of symptom onset.
Patients who were randomized within 3 days of symptom onset (n = 1,379) were included in the modified
intention-to-treat (mITT) analysis. COVID-19-related hospitalizations or all-cause deaths occurred by
Day 28 in 5 of 697 patients (0.72%) in the ritonavir-boosted nirmatrelvir arm and in 44 of 682 patients
(6.5%) in the placebo arm. Among the 2,085 patients who were randomized within 5 days of symptom
onset (mITT1 analysis), COVID-19-related hospitalizations and all-cause deaths occurred in 8 of 1,039
patients (0.77%) in the ritonavir-boosted nirmatrelvir arm and in 66 of 1,046 patients (6.3%) in the
placebo arm (89% relative risk reduction; 5.6% estimated absolute reduction; 95% CI, 7.2% to 4.0%; P <
0.001). There were no deaths in the ritonavir-boosted nirmatrelvir arm and 13 deaths in the placebo arm.
A total of 2,224 patients who received at least 1 dose of either ritonavir-boosted nirmatrelvir or placebo
were included in the EPIC-HR safety analysis set. Among these patients, dysgeusia and diarrhea
occurred more frequently in ritonavir-boosted nirmatrelvir recipients than in placebo recipients (6% vs.
COVID-19 Treatment Guidelines 191
Additional Considerations
• Nirmatrelvir must be administered with ritonavir to achieve sufficient therapeutic plasma
concentrations.
• Patients should complete the 5-day treatment course of ritonavir-boosted nirmatrelvir because
there are concerns that a shorter treatment course may be less effective or lead to resistance.
• If a patient requires hospitalization after starting treatment, the full 5-day treatment course of
ritonavir-boosted nirmatrelvir should be completed unless there are drug-drug interactions that
preclude its use.
• There are no data on using combinations of antiviral therapies or combinations of antiviral agents
and anti-SARS-CoV-2 monoclonal antibodies for the treatment of nonhospitalized patients with
COVID-19. Clinical trials are needed to determine whether combination therapy has a role in the
treatment of COVID-19.
SARS-CoV-2 Resistance
• Viral mutations that lead to substantial resistance to nirmatrelvir have been selected for in vitro
studies; the fitness of these mutations is unclear. Surveillance for the emergence of significant
resistance to nirmatrelvir is critical.
• Severely immunocompromised patients 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 or a longer treatment
duration in treating patients who are severely immunocompromised is not yet known.
Considerations in Pregnancy
Pregnancy is a risk factor for severe COVID-19.4 However, like many clinical trials of treatments for
COVID-19, the EPIC-HR trial excluded pregnant and lactating individuals. Ritonavir has been used
extensively during pregnancy in people with HIV and has a favorable safety profile during pregnancy.
The mechanisms of action for both nirmatrelvir and ritonavir and the results of animal studies of
ritonavir-boosted nirmatrelvir suggest that this regimen can be used safely in pregnant individuals.
Ritonavir-boosted nirmatrelvir should be offered to pregnant and recently pregnant patients with
COVID-19 who qualify for this therapy based on the results of a risk-benefit assessment. The risk-
benefit assessment for using ritonavir-boosted nirmatrelvir in pregnant patients may include factors
such as medical comorbidities, body mass index, vaccination status, and the number and severity of the
risk factors for severe disease. Obstetricians should be aware of potential drug-drug interactions when
prescribing this agent.
Lactation is not a contraindication for the use of ritonavir-boosted nirmatrelvir. There are no data on the
use of nirmatrelvir in lactating people, but the data from animal studies are reassuring. In a prebirth-
to-lactation study, an 8% decrease in body weight was observed on Postnatal Day 17 in the offspring
of rats who received nirmatrelvir and had systemic exposures that were 8 times higher than the clinical
exposures at the authorized human dose. This reduction in body weight was not seen in the offspring of
rats that had exposures that were 5 times higher than the clinical exposures at the authorized human dose.3
Studies of infants who were exposed to ritonavir through breast milk suggest that the amount of ritonavir
that transfers through breast milk is negligible and not considered clinically significant.24 The decision to
feed breast milk while taking ritonavir-boosted nirmatrelvir should take into consideration the benefits
of breastfeeding, the need for the medication, any underlying risks of infant exposure to the drug, and
the potential adverse outcomes of COVID-19.
Considerations in Children
For information on using ritonavir-boosted nirmatrelvir in pediatric patients, see Special Considerations
in Children, Therapeutic Management of Nonhospitalized Children With COVID-19, and Therapeutic
Management of Hospitalized Children With COVID-19.
Drug-Drug Interactions
Ritonavir-boosted nirmatrelvir has significant drug-drug interactions, primarily due to the ritonavir
component of the combination. Boosting with ritonavir, which is a strong CYP3A inhibitor and a
P-glycoprotein inhibitor, is required to increase the exposure of nirmatrelvir to a concentration that is
effective against SARS-CoV-2. However, it may also increase concentrations of certain concomitant
medications, thereby increasing the potential for serious and sometimes life-threatening drug toxicities.
Additionally, ritonavir is an inhibitor, inducer, and substrate of various other drug-metabolizing enzymes
COVID-19 Treatment Guidelines 193
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 M(pro) 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. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for Paxlovid.
2022. Available at: https://www.fda.gov/media/155050/download.
4. Centers for Disease Control and Prevention. People with certain medical conditions. 2022. Available at: https://
www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/people-with-medical-conditions.html. Accessed
September 6, 2022.
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://www.ncbi.nlm.nih.gov/pubmed/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 September 7, 2022.
9. Dryden-Peterson S, Kim A, Kim AY, et al. Nirmatrelvir plus ritonavir for early COVID-19 and hospitalization in a
large US health system. medRxiv. 2022;Preprint. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35734084.
10. Arbel R, Wolff Sagy Y, Hoshen M, et al. Nirmatrelvir use and severe COVID-19 outcomes during the Omicron
surge. N Engl J Med. 2022;387(9):790-798. Available at: https://www.ncbi.nlm.nih.gov/pubmed/36001529.
COVID-19 Treatment Guidelines 194
Ritonavir, a strong cytochrome P450 (CYP) 3A4 inhibitor and a P-glycoprotein inhibitor, is coadministered
with nirmatrelvir to increase the blood concentration of nirmatrelvir, thereby making it effective against
SARS-CoV-2. Ritonavir may also increase blood concentrations of certain concomitant medications.
Because ritonavir-boosted nirmatrelvir (Paxlovid) is the only highly effective oral antiviral for the treatment
of COVID-19, drug interactions that can be safely managed should not preclude the use of this medication.
Clinicians should be aware that many commonly used medications can be safely coadministered with
ritonavir-boosted nirmatrelvir despite its drug-drug interaction potential. Box 1 includes commonly
prescribed medications that are not expected to have clinically relevant interactions with ritonavir-boosted
nirmatrelvir.
Box 1. Select Outpatient Medications Not Expected to Have Clinically Relevant Interactions With
Ritonavir-Boosted Nirmatrelvir (Paxlovid)
This is not a comprehensive list of all the medications that are not expected to have clinically relevant
interactions with ritonavir-boosted nirmatrelvir.a
Medications Without Clinically Relevant Interactions
These medications may be coadministered without dose adjustment and without increased monitoring.a This list is not
inclusive of all noninteracting medications within each drug category.
Acid reducers Cardiovascular, continued Neuropsychiatric Respiratory
• Famotidine • Losartan • Amitriptyline • Corticosteroids (inhaled)
• Omeprazole • Metoprolol • Bupropion • Formoterol
• Pantoprazole • Prasugrel • Citalopram • Montelukast
Allergy Diabetes • Duloxetine Miscellaneous
• Cetirizine • Empagliflozin • Escitalopram • Allopurinol
• Diphenhydramine • Insulin • Fluoxetine • Contraceptives (oral)b
• Fexofenadine • Metformin • G
abapentin • Cyclobenzaprine
• Loratadine • Pioglitazone • Lorazepam • Donepezil
• Nortriptyline • Enoxaparin
Anti-infectives Immunosuppressants • Olanzapine
• Azithromycin • Abrocitinib • Finasteride
• Paroxetine • Levothyroxine
• Cidofovir • Baricitinib • Sertraline
• Hydroxychloroquine • Methotrexate • Most monoclonal antibody
• Venlafaxine productsc
• Tecovirimat • Mycophenolate
Pain • Ondansetron
• Valacyclovir • Prednisone
• Acetaminophen
Cardiovascular Lipid-modifiers • Aspirin
• Aspirin • Ezetimibe • Codeine
• Atenolol • Pitavastatin • Ibuprofen
• Carvedilol • Pravastatin • Meloxicam
• Furosemide Migraine • Naproxen
• Hydrochlorothiazide • Frovatriptan
• Irbesartan • Naratriptan
• Isosorbide dinitrate • Rizatriptan
• Lisinopril • Sumatriptan
References
1. 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.
2. 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.
3. 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.
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 ONLY
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).
• 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 However, this trial occurred before the emergence of the Omicron variant and its subvariants.
A secondary analysis of the patients who required hospitalization during the trial found a reduced need
for respiratory interventions among those who were randomized to receive molnupiravir compared to
those who received placebo.8 Molnupiravir has shown activity against the Omicron subvariants in vitro
and in animal studies.2,9-11
COVID-19 Treatment Guidelines 202
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.
• There are no data on using combination antiviral therapies or combinations of antiviral agents
and anti-SARS-CoV-2 monoclonal antibodies for the treatment of 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.
• It is not yet known how often viral rebound occurs in patients who have completed treatment with
molnupiravir.
Considerations in Children
The MOVe-OUT trial 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 was a multinational, 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 enrolled within 5 days of symptom onset. The trial was conducted before the emergence
of the Omicron variant and its subvariants. Pregnant people, lactating people, and children were
excluded from the study. Participants were randomized to receive molnupiravir 800 mg PO every 12
hours for 5 days or placebo.7 The primary composite outcome was all-cause hospitalization (defined as a
hospital stay >24 hours) or death by Day 29.
The final analysis included 1,433 participants; the median age was 43 years (with 17% aged >60 years).
Forty-nine percent of the participants were men, 57% were White, 50% were Hispanic/Latinx, and 5%
were Black or African American.7 Among the participants, 74% had a body mass index ≥30 and 16%
had diabetes. The time from COVID-19 symptom onset to randomization was ≤3 days in 48% of the
participants.
By Day 29, the use of molnupiravir reduced the risk of hospitalization or death by 31%, with 48 of 709
participants (6.8%) in the molnupiravir arm experiencing hospitalization or death compared with 68 of
699 participants (9.7%) in the placebo arm (-3.0% adjusted difference; 95% CI, -5.9% to -0.1%).7 The
molnupiravir arm had 1 death, and the placebo arm had 9 deaths. There were no significant differences
between the arms in the proportion of participants who experienced adverse events or serious adverse
events. A secondary analysis of data from the patients who were hospitalized during the trial revealed
COVID-19 Treatment Guidelines 204
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. 2021: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: a
randomized controlled clinical trial. Front Pharmacol. 2022;13:939573. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35784723.
3. Zhou S, Hill CS, Sarkar S, et al. Beta-d-N4-hydroxycytidine inhibits SARS-CoV-2 through lethal mutagenesis
but is also mutagenic to mammalian cells. J Infect Dis. 2021;224(3):415-419. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33961695.
4. Kabinger F, Stiller C, Schmitzova J, et al. Mechanism of molnupiravir-induced SARS-CoV-2 mutagenesis.
Nat Struct Mol Biol. 2021;28(9):740-746. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34381216.
5. Centers for Disease Control and Prevention. People with certain medical conditions. 2022. Available at:
https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/people-with-medical-conditions.html.
Accessed September 6, 2022.
6. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for
molnupiravir. 2022. Available at: https://www.fda.gov/media/155054/download.
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. Johnson MG, Puenpatom A, Moncada PA, et al. Effect of molnupiravir on biomarkers, respiratory
interventions, and medical services in COVID-19: a randomized, placebo-controlled trial. Ann Intern Med.
2022;175(8):1126-1134. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35667065.
9. Vangeel L, Chiu W, De Jonghe S, et al. Remdesivir, molnupiravir and nirmatrelvir remain active against
SARS-CoV-2 Omicron and other variants of concern. Antiviral Res. 2022;198:105252. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35085683.
10. Takashita E, Yamayoshi S, Simon V, et al. Efficacy of antibodies and antiviral drugs against Omicron
BA.2.12.1, BA.4, and BA.5 subvariants. N Engl J Med. 2022;387(5):468-470. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35857646.
11. Uraki R, Kiso M, Iida S, et al. Characterization and antiviral susceptibility of SARS-CoV-2 Omicron BA.2.
Nature. 2022;607(7917):119-127. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35576972.
12. Flisiak R, Zarębska-Michaluk D, Rogalska M, et al. Real-world experience with molnupiravir during the
period of SARS-CoV-2 Omicron variant dominance. Pharmacol Rep. 2022;Published online ahead of print.
Available at: https://pubmed.ncbi.nlm.nih.gov/36001284.
13. Yip CF, Lui GCY, Man Lai MS, et al. Impact of the use of oral antiviral agents on the risk of hospitalisation in
community COVID-19 patients. Clin Infect Dis. 2022;Published online ahead of print. Available at:
https://pubmed.ncbi.nlm.nih.gov/36031408.
14. Wong CKH, Au ICH, Lau KTK, et al. Real-world effectiveness of early molnupiravir or nirmatrelvir-ritonavir
in hospitalised patients with COVID-19 without supplemental oxygen requirement on admission during Hong
Kong’s Omicron BA.2 wave: a retrospective cohort study. Lancet Infect Dis. 2022. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/36029795.
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 it has been evaluated in clinical trials for the treatment of COVID-19. Interferon alfa has
been approved to treat hepatitis B and hepatitis C virus infections, and interferon lambda is not currently
approved by the FDA for any use. Both interferon alfa and lambda have also been evaluated for the
treatment of COVID-19.
Recommendations
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends against the use of systemic
interferon beta for the treatment of hospitalized patients with COVID-19 (AI).
• The Panel recommends against the use of interferon alfa or lambda for the treatment of
hospitalized patients with COVID-19, except in a clinical trial (AIIa).
• The Panel recommends against the use of interferons for the treatment of nonhospitalized
patients with mild or moderate COVID-19, except in a clinical trial (AIIa).
Rationale
Many of the early studies that evaluated the use of systemic interferons for the treatment of COVID-19
were conducted in early 2020, before the widespread use of remdesivir and corticosteroids. In addition,
these early 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 not demonstrated efficacy for interferons in the treatment of COVID-19, and
some of the trials suggested potential harm in patients with severe disease, such as those who were
on 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 for interferon beta-1a when this drug was
administered to hospitalized patients, approximately 50% of whom were on corticosteroids.5
Other interferons, including systemic interferon alfa or lambda and inhaled interferons, have also been
evaluated in patients with COVID-19; however, these interferons (with the exception of subcutaneous
interferon alfa) are not available in the United States. The trials that have evaluated interferon alfa and
interferon lambda have generally been small or moderate in size and have not been adequately powered
to assess whether these agents provide a clinical benefit for patients with COVID-19 (see Table 4b).
Clinical Trials
See ClinicalTrials.gov for a list of clinical trials that are evaluating the use of interferons for the
treatment of COVID-19.
Adverse Effects
The most frequent adverse effects of systemic interferon include flu-like symptoms, nausea, fatigue,
weight loss, hematological toxicities, elevated transaminases, and psychiatric problems (e.g., depression,
Drug-Drug Interactions
Additive toxicities may occur when systemic interferons are used concomitantly with other
immunomodulators and chemotherapeutic agents.6,7
Considerations in Pregnancy
According to analyses of data from several large pregnancy registries, exposure to interferon beta-1b
prior to conception or during pregnancy does not lead to an increased risk of adverse birth outcomes (e.g.,
spontaneous abortion, congenital anomaly).8,9 Exposure to interferon beta-1b did not influence birth weight,
height, or head circumference.10
Considerations in Children
There are currently not enough 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;Published online ahead of print. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34672949.
5. WHO Solidarity Trial Consortium, Pan H, Peto R, et al. 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. Interferon alfa-2b (Intron A) [package insert]. Food and Drug Administration. 2018. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/103132Orig1s5199lbl.pdf.
7. Interferon beta-1a (Rebif) [package insert]. Food and Drug Administration. 2019. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/103780s5204lbl.pdf.
8. Sandberg-Wollheim M, Alteri E, Moraga MS, Kornmann G. Pregnancy outcomes in multiple sclerosis following
subcutaneous interferon beta-1a therapy. Mult Scler. 2011;17(4):423-430. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21220368.
9. Hellwig K, Duarte Caron F, Wicklein EM, Bhatti A, Adamo A. Pregnancy outcomes from the global pharma-
covigilance database on interferon beta-1b exposure. Ther Adv Neurol Disord. 2020;13:1756286420910310.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32201504.
10. Burkill S, Vattulainen P, Geissbuehler Y, et al. The association between exposure to interferon-beta during pregnancy
and birth measurements in offspring of women with multiple sclerosis. PLoS One. 2019;14(12):e0227120. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/31887199.
COVID-19 Treatment Guidelines 207
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 • OS6 patients were excluded after 270
≤94% on room air, or supplemental oxygen) • 58% men; 32% Latino, 60% White, 17% Black patients were enrolled 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 IFN
beta-1a plus RDV in hospitalized patients
• Impaired renal function Primary Outcome: compared to RDV alone.
• Anticipated hospital discharge or transfer within 72 • Median time to recovery for both arms was 5 days (rate • The use of IFN beta-1a was associated
hours ratio 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 was >28 days in
• RDV 200 mg IV on Day 1, then RDV 100 mg IV once
IFN beta-1a arm and 9 days in placebo arm (rate ratio
daily for 9 days plus IFN beta-1a 44 µg SQ every other
0.40; 95% CI, 0.22–0.75; P = 0.0031).
day for up to 4 doses (n = 487)
• RDV 200 mg IV on Day 1, then RDV 100 mg IV once Secondary Outcomes:
daily for 9 days plus placebo (n = 482) • No difference between arms in clinical improvement at
14 days (OR 1.01; 95% CI, 0.79–1.28).
Primary Endpoint:
• No difference between arms in mortality by Day 28 in:
• Time to recovery by Day 28
• All patients: 5% vs. 3% (HR 1.33; 95% CI, 0.69–2.55)
Key Secondary Endpoints:
• Patients with OS6 at baseline: 21% vs. 12% (HR 1.74;
• Clinical status at Day 14, as measured by an OS 95% CI, 0.51–5.93)
• Mortality by Day 28
WHO Solidarity Trial: Multinational, Open-Label, Adaptive RCT of IV or SQ Interferon Beta-1a or Other Repurposed Drugs in Hospitalized Adults With
COVID-192
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Diagnosis of COVID-19 • 35% aged <50 years; 19% aged ≥70 years; 63% men • Open-label study
• Not expected to be transferred elsewhere within 72 • 70% on supplemental oxygen; 7% on ventilation • IFN beta-1a given as IV or SQ
hours • Approximately 50% received corticosteroids during the formulations at different doses
Interventions: study Interpretation:
• IFN beta-1a 44 µg SQ on day of randomization, Day 3, Primary Outcomes: • IFN beta-1a does not improve mortality
and Day 6 (n = 1,656) • In-hospital mortality was 11.9% for combined IFN beta- for hospitalized patients.
• IFN beta-1a 10 µg IV daily for 6 days for patients on 1a arms and 10.5% in SOC arm (rate ratio 1.16; 95% CI,
high-flow oxygen, ventilation, or ECMO (n = 394) 0.96–1.39).
• IFN beta-1a (either SQ or IV) and LPV/RTV 400 mg/50 • For IFN beta-1a only (without LPV/RTV) recipients
mg twice daily for 14 days (n = 651) vs. SOC recipients, rate ratio was 1.12 (95% CI,
• Local SOC (n = 2,050) 0.83–1.51).
• Among those on ventilation at entry, age-stratified
Primary Endpoint:
rate ratio for in-hospital mortality was 1.40 (95% CI,
• In-hospital mortality 0.93–2.11).
Key Secondary Endpoint: Secondary Outcome:
• Initiation of ventilation • 10% initiated ventilation in the combined IFN beta-1a
arms and SOC arm.
DisCoVeRy Solidarity Trial Add-On: Open-Label, Adaptive RCT of SQ Interferon Beta-1a Plus Lopinavir/Ritonavir, Lopinavir/Ritonavir, or Hydroxychloroquine
in Hospitalized Adults With COVID-19 in France3
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Positive PCR result for SARS-CoV-2 • Median age 63 years; 72% men • Open-label study
• Patients had pulmonary rales or crackles with SpO2 • 29% were obese; 26% with chronic cardiac disease; • Most patients had moderate disease
≤94% or they required supplemental oxygen 22% with DM • No IFN beta-1a arm without LPV/RTV
Interventions: • 36% had severe disease • Study stopped early for futility
• IFN beta-1a 44 ug SQ on Days 1, 3, and 6 plus LPV/RTV • Median of 9 days from symptom onset to randomization
Interpretation:
400 mg/100 mg PO twice daily for 14 days plus SOC (n • 30% received steroids during the study
= 145) • Compared to SOC alone, the use of IFN-
Primary Outcome: beta-1a plus LPV/RTV did not improve
• LPV/RTV 400 mg/100 mg PO twice daily for 14 days clinical status, rate of viral clearance, or
plus SOC (n = 145) • No difference in clinical status at Day 15 for any
intervention compared to SOC: time to viral clearance in hospitalized
• HCQ 400 mg twice on Day 1, then HCQ 400 mg daily for patients with COVID-19.
9 days plus SOC (n = 145) • IFN beta-1a plus LPV/RTV: aOR 0.69 (95% CI, 0.45–
1.04; P = 0.08)
• SOC alone, which included corticosteroids,
anticoagulants, or immunomodulatory agents but not • LPV/RTV: aOR 0.83 (95% CI, 0.55–1.26; P = 0.39)
antivirals (n = 148) • HCQ: aOR 0.93 (95% CI, 0.62–1.41; P = 0.75)
Primary Endpoint: Secondary Outcomes:
• Clinical status at Day 15, as measured by an OS • No difference in clinical status at Day 29 between the
arms.
Key Secondary Endpoints:
• No difference in rate and time to SARS-CoV-2 viral
• Clinical status at Day 29 clearance between the arms.
• Rate of SARS-CoV-2 viral clearance • Time to 2 OS-category improvement and hospital
• Time to SARS-CoV-2 viral clearance discharge by Day 29 was longer in LPV/RTV plus IFN
• Time to improvement of 2 OS categories beta-1a and LPV/RTV arms than in SOC arm.
• Time to hospital discharge
Single-Blind RCT of Peginterferon Lambda-1a for Treatment of Outpatients With Uncomplicated COVID-19 in the United States4
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 RT-PCR result for SARS-CoV-2 within 72 hours • 7% were asymptomatic
• PEG-IFN lambda-1a provided no
of enrollment • Median of 5 days of symptoms before randomization virologic or clinical benefit compared
Key Exclusion Criteria: Primary Outcome: to placebo among outpatients with
uncomplicated COVID-19.
• Current or imminent hospitalization • Median time to cessation of viral shedding was 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%
• Single dose of PEG-IFN lambda-1a 180 µg SQ (n = 60) for each arm
• Placebo (n = 60) • Time to resolution of symptoms: 8 days vs. 9 days
Primary Endpoint: (HR 0.94; 95% CI, 0.64–1.39)
• Time to first negative SARS-CoV-2 RT-PCR result Other Outcomes:
Key Secondary Endpoints: • Patients who received PEG-IFN lambda-1a were more
• Hospitalizations by Day 28 likely to have transaminase elevations than patients who
received placebo (25% vs. 8%; P = 0.027).
• Time to complete symptom resolution
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, Pan H, Peto R, et al. 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 212
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.
Proposed Mechanism of Action and Rationale for Use in Patients With COVID-19
Reports from in vitro studies suggest that ivermectin acts by inhibiting host importin alpha/beta-1
nuclear transport proteins, which are part of a key intracellular transport process.3,4 Viruses hijack the
process and enhance infection by suppressing the host’s antiviral response. In addition, ivermectin
docking may interfere with SARS-CoV-2 spike protein attachment to the human cell membrane.5
Some studies of ivermectin have also reported potential anti-inflammatory properties, which have been
postulated to be beneficial in people with COVID-19.6-8
Ivermectin has been shown to inhibit replication of SARS-CoV-2 in cell cultures.9 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.10,11 Although ivermectin appears to accumulate in lung
tissue, predicted systemic plasma and lung tissue concentrations are much lower than 2 µM, the half-
maximal inhibitory concentration (IC50) observed in vitro for ivermectin against SARS-CoV-2.12-15
Subcutaneous administration of ivermectin 400 µg/kg had no effect on SARS-CoV-2 viral loads in
hamsters.16 However, there was a reduction in olfactory deficit (measured using a food-finding test) and
a reduction in the interleukin (IL)-6:IL-10 ratio in lung tissues.
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 4c. The
Panel reviewed additional studies, but these studies are not summarized in Table 4c 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, except in
clinical trials (AIIa).
Rationale
The results of several randomized trials and retrospective cohort studies of ivermectin use in patients
with COVID-19 have been published in peer-reviewed journals or have been made available as
manuscripts ahead of peer review. Most of these studies, especially studies completed earlier in
the pandemic, had incomplete information and significant methodological limitations, which made
excluding common causes of bias difficult. Many of these studies have not been peer reviewed, and
some have now been retracted.
The Panel’s recommendation is primarily informed by recently published randomized controlled
trials.17-20 The primary outcomes of these trials showed that the use of ivermectin for the treatment of
COVID-19 had no clinical benefit. In TOGETHER, an adaptive platform trial conducted in Brazil, there
COVID-19 Treatment Guidelines 214
Clinical Trials
Several clinical trials evaluating the use of ivermectin for the treatment of COVID-19 are currently
underway or in development. Please see ClinicalTrials.gov for the latest information.
The clinical trials described in this table are RCTs that had the greatest impact on the Panel’s recommendation. The Panel reviewed other
clinical studies 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 the table.
TOGETHER: Double-Blind, Adaptive, RCT of Ivermectin in Nonhospitalized Patients With COVID-19 in Brazil27
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Positive SARS-CoV-2 antigen test • Median age 49 years; 46% aged ≥50 years; 58% • Health care facility capacity may have
• Within 7 days of symptom onset women; 95% “mixed race” influenced the number and duration
• Most prevalent risk factor: 50% with obesity of emergency setting visits and
• ≥1 high-risk factor for disease progression (e.g., aged hospitalizations.
>50 years, comorbidities, immunosuppression) • Symptom onset: 44% within 3 days
• No details on safety outcomes (e.g., type
Interventions: Primary Outcome: of treatment-emergent AEs) other than
• IVM 400 μg/kg PO per day for 3 days (n = 679) • Composite of emergency setting observation >6 hours grading were reported.
• Placebo (n = 679; not all participants received IVM or hospitalized within 28 days of randomization (ITT): Interpretation:
placebo) 100 (14.7%) in IVM arm vs. 111 (16.4%) in placebo arm
(relative risk 0.90; 95% CrI, 0.70–1.16) • In outpatients with recent COVID-19
Primary Endpoint: infection, IVM did not reduce the
• 171 (81%) of all events were hospitalizations (ITT) need for emergency setting visits or
• Composite of emergency setting observation >6
hours or hospitalized for COVID-19 within 28 days of Secondary Outcomes: hospitalization when compared with
randomization • No difference between IVM and placebo arms in: placebo.
Key Secondary Endpoints: • Viral clearance at Day 7 (relative risk 1.00; 95% CrI,
0.68–1.46)
• Viral clearance at Day 7
• All-cause mortality: 21 (3.1%) vs. 24 (3.5%) (relative
• All-cause mortality
risk 0.88; CrI, 0.49–1.55)
• Occurrence of AEs
• Occurrence of AEs
IVERCOR-COVID19: Double-Blind, Placebo-Controlled, Randomized Trial of Ivermectin in Nonhospitalized Patients With COVID-19 in Argentina28
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 • Enrolled a fairly 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 time from symptom onset: 4 days
• Oxygen supplementation 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. acquired SARS-CoV-2 infection, there
Interventions: 8.3% in placebo arm (OR 0.65; 95% CI, 0.32–1.31; P = was no evidence that IVM provided any
0.23) clinical benefit.
• Weight-based dose of IVM PO at enrollment and 24
hours later for a maximum total dose of 48 mg (n = Secondary Outcomes:
250) • Need for MV: 2% in IVM arm vs. 1% in placebo arm (P
• Placebo (n = 251) = 0.7)
Primary Endpoint: • All-cause mortality: 2% in IVM arm vs. 1% in placebo
arm (P = 0.7)
• Hospitalization for any reason
• Occurrence of AEs: 18% in IVM arm vs. 21% in placebo
Key Secondary Endpoints: arm (P = 0.6)
• Need for MV
• All-cause mortality
• Occurrence of AEs
I-TECH: Open-Label RCT of Ivermectin in Patients With Mild to Moderate COVID-19 in Malaysia29
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 dose COVID-19 vaccine; 52% received Interpretation:
• Aged ≥50 years 2 doses
• In patients with mild to moderate
•≥ 1 comorbidity • 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 duration of symptoms: 5 days including no evidence that IVM reduced
• Required supplemental oxygen 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 daily for 5 days plus SOC (n = 241) (relative risk 1.25; 95% CI, 0.87–1.80; P = 0.25)
• SOC (n = 249) Secondary Outcomes:
Primary Endpoint: • No difference between IVM plus SOC arm and SOC alone
• Progression to severe COVID-19 (i.e., hypoxia requiring arm in:
supplemental oxygen to maintain SpO2 ≥95%) • In-hospital, all-cause mortality: 3 (1.2%) vs. 10 (4.0%)
Key Secondary Endpoints: (relative risk 0.31; 95% CI, 0.09–1.11; P = 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
• Occurrence of AEs
0.78; 95% CI, 0.27–2.20; P = 0.79)
• Occurrence of AEs: 33 (13.7%) in the IVM plus SOC arm
vs. 11 (4.4%) in the SOC alone arm; most with diarrhea
(14 vs. 4)
Double-Blind, Placebo-Controlled, Randomized Trial of Ivermectin in Patients With Mild COVID-19 in Colombia30
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, • Due to low event rates, the primary
• Symptoms ≤7 days 8% in placebo arm; 39% men in IVM arm, 45% in endpoint changed from the proportion of
placebo 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 symptom onset to randomization: 5 days
• 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 300
• IVM 300 μg/kg PO per day for 5 days (n = 200) 79% in placebo arm μg/kg per day for 5 days did not improve
• Placebo PO (n = 198) Secondary Outcomes: the time to resolution of symptoms.
Primary Endpoint: • Clinical deterioration: no difference between arms
• Time to resolution of symptoms within 21 days • Escalation of care: no difference between arms
Key Secondary Endpoints: • Occurrence of AEs:
• Clinical deterioration • Discontinued treatment due to AEs: 8% in IVM arm vs.
3% in placebo arm
• Escalation of care
• No SAEs were related to intervention
• Occurrence of AEs
Double-Blind, Placebo-Controlled, Randomized Trial of Ivermectin in Patients With Mild to Moderate COVID-19 in India32
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 or 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 for 2 days (n = 55) • Mean 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:
Primary Outcome: and outcomes after discharge were not
• Negative SARS-CoV-2 RT-PCR result on Day 6 evaluated.
• Negative RT-PCR result on Day 6: 24% in IVM arm vs.
Key Secondary Endpoints: 32% in placebo arm (rate ratio 0.8; P = 0.348) Interpretation:
• Symptom resolution by Day 6 • IVM provided no significant virologic or
Secondary Outcomes:
• Discharge by Day 10 clinical benefit for patients with mild to
• Symptom resolution by Day 6: 84% in IVM arm vs. 90% moderate COVID-19.
• Need for ICU admission or MV in placebo arm (rate ratio 0.9; P = 0.36)
• In-hospital mortality • Discharge by Day 10: 80% in IVM arm vs. 74% in
placebo arm (rate ratio 1.1; P = 0.43)
• Need for ICU admission or MV: no difference between
arms
• 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 India33
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 Interpretation:
asymptomatic patients); 33% to 40% with moderate
Key Exclusion Criteria: • For patients who received IVM and those
COVID-19
• CrCl <30 mL/min who received placebo, there was no
• Median duration of symptoms: 4–5 days, similar across difference in the proportion of negative
• Transaminases >5 times ULN arms RT-PCR results at Day 5 or clinical
• MI, heart failure, QTc interval prolongation • 10% received concurrent antivirals (RDV, favipiravir, or outcomes.
• Severe comorbidity HCQ); no difference across arms
Interventions: Primary Outcomes:
• Single dose of IVM 24 mg PO (n = 51) • Negative RT-PCR result at Day 5: 48% in IVM 24 mg arm
• Single dose of IVM 12 mg PO (n = 49) vs. 35% in IVM 12 mg arm vs. 31% in placebo arm (P =
0.30)
• Placebo (n = 52)
• Decline of VL at Day 5: no significant difference between
Primary Endpoints: arms
• Negative RT-PCR result at Day 5 Secondary Outcomes:
• Decline of VL at Day 5 • Time to symptom resolution: no difference between arms
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 • Number of hospital-free days at Day 28: no difference
between arms
• Number of hospital-free days at Day 28
• Frequency of AEs: no difference between arms; no SAEs
• Frequency of AEs
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 symptoms • 90% of subjects screened were not
• Not hospitalized or receiving supplemental oxygen enrolled for various reasons.
Primary Outcomes:
• Recruitment stopped early because
Key Exclusion Criteria: • Number of SAEs: 0
of decline in the number of COVID-19
• CNS disease • Mean log10 reduction in VL at Day 7: 2.9 in IVM 1,200 μg/ cases.
• Receiving dialysis kg arm vs. 2.5 in IVM 600 μg/kg arm vs. 2.0 in placebo
arm (IVM 1,200 μg/kg vs. placebo, P = 0.099; IVM 600 Interpretations:
• Severe medical condition with <6 months survival μg/kg vs. placebo, P = 0.122) • A high dose of IVM (1,200 μg/kg)
prognosis appears to be safe but not well-tolerated;
• Use of warfarin, antiviral agents, CQ, or HCQ AE Outcomes:
34% discontinued therapy due to AEs.
• 14 (15.1%) discontinued treatment: 11 (34.4%) in IVM
Interventions: • There was no significant difference
1,200 μg/kg arm vs. 2 (6.9%) in IVM 600 μg/kg arm vs.
• IVM 1,200 μg/kg PO once daily for 5 days (n = 32) in reduction of VL between IVM and
1 (3.1%) in placebo arm
placebo arms.
• IVM 600 μg/kg plus placebo PO once daily for 5 days (n • All discontinuations in IVM 1,200 μg/kg arm due to
= 29) tolerability
• Placebo PO (n = 32)
Primary Endpoints:
• Number of SAEs
• Change in VL at Day 7
Double-Blind RCT of Ivermectin, Chloroquine, or Hydroxychloroquine in Hospitalized Adults With Severe COVID-19 in Brazil35
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 by CXR or CT • ICU admission: 28% vs. 22% vs. 21%
Key Exclusion Criterion: • Need for MV: 24% vs. 21% vs. 21%
• Cardiac arrhythmia • Mortality: 23% vs. 21% vs. 22%
Interventions: • Mean number of days of supplemental oxygen: 8 days
for each arm
• IVM 14 mg once daily for 3 days (n = 53)
• Occurrence of AEs: no difference between arms
• CQ 450 mg twice daily on Day 0, then once daily for 4
days (n = 61) • Baseline characteristics significantly associated with
mortality:
• HCQ 400 mg twice daily on Day 0, then once daily for 4
days (n = 54) • Aged >60 years (HR 2.4)
Endpoints: • DM (HR 1.9)
• Need for supplemental oxygen, MV, or ICU admission • BMI >33 (HR 2.0)
• Occurrence of AEs • SpO2 <90% (HR 5.8)
• Mortality
Key: AE = adverse event; ALT = alanine aminotransferase; BMI = body mass index; CNS = central nervous system; CQ = chloroquine; CrCl = creatinine clearance; CT
= computed tomography; CXR = chest X-ray; DM = diabetes mellitus; 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 = orally; 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
Molnupiravir
Authorized under an FDA EUA for the treatment of mild to moderate COVID-19 in high-risk individuals aged ≥18 years.
Dose Recommended in FDA EUA: • Diarrhea • Before initiating • Clinical drug-drug • MOV can be taken with or
• MOV 800 mg (4 200-mg capsules) PO • Nausea MOV, assess interaction studies of without food.
every 12 hours for 5 days pregnancy status MOV have not been • Sexually active individuals of
• Dizziness as clinically conducted. reproductive potential should
• Per the FDA, the 5-day course indicated. • Drug-drug interactions use effective contraception
of MOV has a low risk for • Monitor for related to hepatic during and following treatment
genotoxicity.3 See Molnupiravir for potential AEs. metabolism are not with MOV. See Molnupiravir for
details. expected. details.
• If MOV is prescribed for
a pregnant individual, the
prescribing clinician should
document that the risks and
benefits were discussed and
that the patient chose this
therapy. Pregnant patients
should also be offered the
opportunity to participate in
the pregnancy surveillance
program.
• During MOV treatment and
for 4 days after the final dose,
lactating people should not
breastfeed their infants.
• MOV is not authorized for use
in children aged <18 years due
to potential effects on bone and
cartilage growth.
• A list of clinical trials is
available: Molnupiravir
Interferon Alfa
Not approved by the FDA and not recommended by the Panel for the treatment of COVID-19. Currently under investigation in clinical trials.
IFN Alfa-2b • AEs associated with inhaled • Respiratory • Low potential for drug- • The nebulized formulation
Dose for COVID-19 in Clinical Trials: therapy (e.g., throat irritation, symptoms after drug interactions of IFN alfa has been the
cough, bronchospasm) inhalation formulation most used in
• Nebulized IFN alfa-2b 5 million clinical trials for the treatment
international units twice daily • Minimal systemic effects expected
of COVID-19. IFN alfa is
• The optimal duration of treatment is usually included as part of a
unclear. combination regimen.
• A list of clinical trials is
available: Interferon Alfa
Availability:
• Nebulized IFN alfa-2b is not
approved by the FDA for use in
the United States.
Interferon Beta
Not approved by the FDA and not recommended by the Panel for the treatment of COVID-19. Currently under investigation in clinical trials.
IFN Beta-1a • Flu-like symptoms (e.g., fever, • CBC with • Low potential for drug- • A list of clinical trials is
Dose for COVID-19 in Clinical Trials: fatigue, myalgia) differential drug interactions available: Interferon Beta
• IFN beta-1a 44 µg SUBQ or IV every • Leukopenia, neutropenia, • Liver enzymes •U se with caution with Availability
other day for up to 3 or 4 doses thrombocytopenia, lymphopenia • Worsening CHF other hepatotoxic
Brand Names of IFN Beta-1a
• Liver function abnormalities (ALT agents.
IFN Beta-1b • Depression, Products:
> AST) suicidal ideation • Reduce dose if ALT >5
Dose for COVID-19 in Clinical Trials: • Avonex, Plegridy, Rebif
• Injection site reactions times ULN.
• IFN beta-1b 8 million international units Brand Names of IFN Beta-1b
• Headache Products:
SUBQ every other day for up to 7 days
total • Hypertonia • Betaseron, Extavia
• Pain
• Rash
• Worsening depression
• Induction of autoimmunity
Interferon Lambda
Not approved by the FDA and not recommended by the Panel for the treatment of COVID-19. Currently under investigation in clinical trials.
PEG-IFN Lambda-1a • Liver function abnormalities • CBC with • Low potential for drug-drug • A list of clinical trials is
Dose for COVID-19 in Clinical Trials: • Injection site reactions differential interactions available: Interferon Lambda
• Single dose of PEG-IFN lambda-1a 180 • Liver enzymes • Use with caution with Availability:
µg SUBQ • Monitor for other hepatotoxic agents.
• PEG-IFN lambda-1a is not
potential AEs. approved by the FDA for use
in the United States.
Ivermectin
Not approved by the FDA and not recommended by the Panel for the treatment of COVID-19. Currently under investigation in clinical trials.
Dose for COVID-19 in Clinical Trials: • Dizziness • Monitor for • Minor CYP3A4 substrate • Generally given on an empty
• IVM 200–600 μg/kg PO as a single • Pruritis potential AEs. • P-gp substrate stomach with water; however,
dose or a once-daily dose for up to 5 administering IVM with food
• GI effects (e.g., nausea, diarrhea) increases its bioavailability.4
days
• Neurological AEs have been • A list of clinical trials is
reported when IVM has been used available: Ivermectin
to treat parasitic diseases, but it is
not clear whether these AEs were
caused by IVM or the underlying
conditions.
Key: AE = adverse event; ALT = alanine transaminase; AST = aspartate aminotransferase; CBC = complete blood count; CHF = congestive heart failure; CYP =
cytochrome P450; eGFR = estimated glomerular filtration rate; EUA = Emergency Use Authorization; FDA = Food and Drug Administration; GI = gastrointestinal;
HSR = hypersensitivity reaction; HTN = hypertension; IFN = interferon; IV = intravenous; IVM = ivermectin; mAb = monoclonal antibody; MATE = multidrug and toxin
extrusion protein; MOV = molnupiravir; OATP = organic anion transporting polypeptide; OTC = over the counter; the Panel = the COVID-19 Treatment Guidelines
Panel; PEG-IFN = pegylated interferon; P-gp = P-glycoprotein; PO = orally; RDV = remdesivir; RTV = ritonavir; SBECD = sulfobutylether-beta-cyclodextrin; SUBQ =
subcutaneous; ULN = upper limit of normal
References
1. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for Paxlovid. 2022. Available at:
https://www.fda.gov/media/155050/download.
2. Remdesivir (Veklury) [package insert]. Food and Drug Administration. 2020. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/
label/2020/214787Orig1s000lbl.pdf.
3. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for molnupiravir. 2022. Available at:
COVID-19 Treatment Guidelines 234
The SARS-CoV-2 genome encodes 4 major structural proteins: spike (S), envelope (E), membrane
(M), and nucleocapsid (N), as well as nonstructural and accessory proteins. The spike protein is further
divided into 2 subunits, S1 and S2, that mediate host cell attachment and invasion. Through its receptor-
binding domain (RBD), S1 attaches to angiotensin-converting enzyme 2 (ACE2) on the host cell; this
initiates a conformational change in S2 that results in virus-host cell membrane fusion and viral entry.1
Anti-SARS-CoV-2 monoclonal antibodies (mAbs) that target the spike protein have been shown to have
clinical benefits in treating SARS-CoV-2 infection. The anticipated activity of the different anti-SARS-
CoV-2 mAb therapies varies dramatically depending on the circulating variant. The recommendations
and discussion below pertain only to anti-SARS-CoV-2 mAb products for the treatment of COVID-19.
Currently, no product is available for post-exposure prophylaxis (PEP). For recommendations and
discussion regarding the use of tixagevimab plus cilgavimab (Evusheld) as pre-exposure prophylaxis
(PrEP), see Prevention of SARS-CoV-2 Infection.
The Omicron variant of concern (VOC) has become the dominant SARS-CoV-2 variant in the United
States.2 This variant and its subvariants have markedly reduced in vitro susceptibility to several anti-
SARS-CoV-2 mAbs, especially bamlanivimab plus etesevimab and casirivimab plus imdevimab.
Sotrovimab is active against the Omicron BA.1 and BA.1.1 subvariants, but it has substantially
decreased in vitro neutralization activity against the Omicron BA.2, BA.4, and BA.5 subvariants.
Bebtelovimab retains in vitro neutralization activity against circulating Omicron subvariants.3-5
Recommendations
The COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations for the use of anti-SARS-
CoV-2 mAbs are based on current knowledge of the in vitro activities of the available products against
the circulating SARS-CoV-2 variants and subvariants.
Bebtelovimab
• For nonhospitalized adults aged ≥18 years with mild to moderate COVID-19 who are at high risk
of progressing to severe disease, the Panel recommends using bebtelovimab 175 mg intravenous
(IV) injection as an alternative therapy ONLY when both ritonavir-boosted nirmatrelvir (Paxlovid)
and remdesivir are not available, feasible to use, or clinically appropriate (CIII).
• Treatment should be initiated as soon as possible and within 7 days of symptom onset.
• See the Centers for Disease Control and Prevention (CDC) webpage People With Certain Medical
Conditions for information on medical conditions that are associated with an increased risk of
progression to severe COVID-19 and Therapeutic Management of Nonhospitalized Adults With
COVID-19 for further guidance on the use of bebtelovimab.
• For recommendations for nonhospitalized children, see Therapeutic Management of
Nonhospitalized Children With COVID-19.
• Bebtelovimab is 1 of the treatment options that can be considered for adults aged ≥18 years with
mild to moderate COVID-19 who are hospitalized for a reason other than COVID-19 if they
otherwise meet the Food and Drug Administration (FDA) Emergency Use Authorization (EUA)
criteria for outpatient treatment.
Additional Considerations
• For information on medical conditions and other factors that are associated with an increased risk of
progression to severe COVID-19, see the CDC webpage People With Certain Medical Conditions.
The decision to use anti-SARS-CoV-2 mAbs for a patient should be based on an individualized
assessment of the risks and benefits.6 Not all of the conditions and factors considered to be high risk
were well-represented in the clinical trials that provide support for the mAb EUAs.
• Some rare medical conditions that are not listed on the CDC webpage Underlying Medical
Conditions Associated With Higher Risk for Severe COVID-19 and other factors (e.g.,
race, ethnicity) may be associated with a high risk of progressing to severe COVID-19. It is
important to note that the likelihood of developing severe COVID-19 increases when a person
has multiple high-risk conditions or comorbidities.7-10
• Previously published clinical trials that evaluated the use of anti-SARS-CoV-2 mAbs for the
treatment of COVID-19 largely enrolled an unvaccinated participant population. The risk of
progression to severe COVID-19 in high-risk patients is substantially greater for those who are
not vaccinated against COVID-19 and those who are vaccinated but do not mount an adequate
immune response to the vaccine due to an underlying immunocompromising condition.
• If indicated, treatment with anti-SARS-CoV-2 mAbs should be started as soon as possible after
SARS-CoV-2 infection is confirmed by an antigen test or a nucleic acid amplification test and
within 7 days of symptom onset.
• Anti-SARS-CoV-2 mAbs should be administered in a setting where severe hypersensitivity
reactions, such as anaphylaxis, can be managed. Patients should be monitored for at least 1 hour
after the injection.
• See Prioritization of Anti-SARS-CoV-2 Therapies for the Treatment and Prevention of COVID-19
When There Are Logistical or Supply Constraints for the Panel’s recommendations in situations
where therapies for the treatment of mild to moderate COVID-19, including anti-SARS-CoV-2
mAbs, cannot be offered to all eligible patients.
• Data are limited on the combined use of antiviral agents and anti-SARS-CoV-2 mAbs for the
treatment of nonhospitalized patients with COVID-19. Clinical trials are needed to determine
whether this combination therapy has a role in the treatment of COVID-19.
• Patients who are severely immunocompromised may have prolonged SARS-CoV-2 replication,
leading to more rapid viral evolution. There is a concern that using a single anti-SARS-CoV-2
mAb in these patients may result in the emergence of resistant virus. Additional studies are needed
to assess this risk.11,12
BEB TIX Plus CIL BAM Plus ETE CAS Plus IMD SOT
WHO Label Anti- Anti- Anti- Anti- Anti-
Years of Notable In Vitro In Vitro In Vitro In Vitro In Vitro
and Pango cipated cipated cipated cipated cipated
Circulation Mutations Suscept- Suscept- Suscept- Suscept- Suscept-
Lineage Clinical Clinical Clinical Clinical Clinical
ibilitya ibilitya ibilitya ibilitya ibilitya
Activity Activity Activity Activity Activity
Variants Currently or Recently Circulating in the United States
2021– Omicron BA.2 T376A, K417N, No change Active No change Active Marked Unlikely to Marked Unlikely to Marked Unlikely to
present N440K, E484A, reduction be active reduction be active reduction be active
Q493R, N501Y
2022– Omicron BA.4 BA.2 plus No change Active Moderate Activec Marked Unlikely to Marked Unlikely to Marked Unlikely to
present del69/70, L452R, reductionb reduction be active reduction be active reduction be active
F486V, Q493
reversion
2022– Omicron BA.5 BA.2 plus No change Active Moderate Activec Marked Unlikely to Marked Unlikely to Marked Unlikely to
present del69/70, L452R, reductionb reduction be active reduction be active reduction be active
F486V, Q493
reversion
Variants That Are Not Currently Circulating in the United States
2020–2021 Alpha B.1.1.7 N501Y No change Active No change Active No change Active No change Active No change Active
2020–2021 Beta B.1.351 K417N, E484K, No change Active No change Active Marked Unlikely to No changed Active No change Active
N501Y reduction be active
2020–2021 Gamma P.1 K417T, E484K, No change Active No change Active Marked Unlikely to No changed Active No change Active
N501Y reduction be active
2020–2021 Delta L452R, T478K No change Active No change Active No change Active No change Active No change Active
B.1.617.2,
non-AY.1/AY.2
2021–2022 Omicron K417N, N440K, No change Active Moderate Activec Marked Unlikely to Marked Unlikely to No change Active
B.1.1.529/ G446S, E484A, reductionb reduction be active reduction be active
BA.1 Q493R, N501Y
2021–2022 Omicron BA.1 plus R346K No change Active Moderate Activec Marked Unlikely to Marked Unlikely to No change Active
BA.1.1 reductionb reduction be active reduction be active
a
Based on the fold reduction in susceptibility reported in the FDA EUAs.6,16,24,25
b
Despite the moderately reduced in vitro susceptibility of TIX plus CIL, in vitro PK/PD modeling data suggest that the TIX 300 mg plus CIL 300 mg dose will retain
activity against the Omicron VOC.16
c
The duration of activity for TIX plus CIL against these subvariants is not well defined. TIX plus CIL should be given in repeat doses every 6 months if ongoing
protection is needed.
COVID-19 Treatment Guidelines 241
Clinical Trials
In placebo-controlled, randomized trials in nonhospitalized patients with mild to moderate COVID-19
symptoms and certain risk factors for disease progression, the use of anti-SARS-CoV-2 mAb products
reduced the risk of hospitalization and death (see Table 5a).6,13,24,25 These studies were conducted before
the widespread circulation of the Omicron VOC.
Bebtelovimab
Based on in vitro data, bebtelovimab is expected to have activity against a broad range of SARS-CoV-2
variants, including the Omicron VOC and its BA.1, BA.2, BA.4, and BA.5 subvariants.13-15 The Panel’s
recommendation for bebtelovimab is primarily based on laboratory data showing its potent activity
against the Omicron VOC (including the BA.1 and BA.2 subvariants) and other VOCs, as well as on
limited clinical trial data from the Phase 2 BLAZE-4 study.13
In treatment arms 9 to 11 in the Phase 2 BLAZE-4 trial, patients with COVID-19 who were at low risk
of disease progression were randomized to receive a single infusion of bamlanivimab plus etesevimab
plus bebtelovimab (n = 127), bebtelovimab alone (n = 125), or placebo (n = 128).13 Among these
individuals, the mean decline in viral load at Day 5 was greater in the 2 bebtelovimab arms than in the
placebo arm. The median time to sustained symptom resolution was 6 days in the bebtelovimab alone
arm and 8 days in the placebo arm (P = 0.003).
Large randomized controlled trials are needed to fully evaluate the efficacy of bebtelovimab in a
high-risk population. Nevertheless, when other therapeutic options are not available, feasible to use, or
clinically appropriate, in vitro susceptibility data and the antiviral activity and clinical benefits observed
in Phase 2 trials support the use of bebtelovimab for nonhospitalized patients with mild to moderate
COVID-19 who are at high risk of progressing to severe COVID-19. In addition, bebtelovimab has
mechanisms of action that are similar to those of other authorized anti-SARS-CoV-2 mAbs that have
shown definitive clinical benefits in this population.
Monitoring
Bebtelovimab should be administered by IV injection and should only be administered in health care
settings by qualified health care providers who have immediate access to emergency medical services
and medications that treat severe infusion-related reactions. Patients should be monitored for at least 1
hour after the injection.
Adverse Effects
Hypersensitivity, including anaphylaxis and infusion-related reactions, has been reported in patients who
received anti-SARS-CoV-2 mAbs. Rash, diarrhea, nausea, vomiting, dizziness, and pruritis have also
been reported.6,13,25,28
Drug-Drug Interactions
Drug-drug interactions are unlikely between the authorized anti-SARS-CoV-2 mAbs and medications
that are renally excreted or that are cytochrome P450 substrates, inhibitors, or inducers (see Table 5c).
Considerations in Pregnancy
The use of anti-SARS-CoV-2 mAbs can be considered for pregnant people with COVID-19, especially
those who have additional risk factors for severe disease.
As immunoglobulin (Ig) G mAbs, the authorized anti-SARS-CoV-2 mAbs would be expected to cross
the placenta. There are no pregnancy-specific data on the use of these mAbs; however, other IgG
products have been safely used in pregnant people when indicated. Therefore, authorized anti-SARS-
CoV-2 mAbs should not be withheld during pregnancy. When possible, pregnant and lactating people
should be included in clinical trials that are evaluating the use of anti-SARS-CoV-2 mAbs for the
treatment or prevention of COVID-19.
Considerations in Children
Please see Therapeutic Management of Nonhospitalized Children With COVID-19 for therapeutic
recommendations for children with COVID-19.
Drug Availability
Bebtelovimab is available for the treatment of COVID-19 and tixagevimab plus cilgavimab is available
for SARS-CoV-2 PrEP in all regions of the United States. The broad distribution of bamlanivimab plus
etesevimab, casirivimab plus imdevimab, and sotrovimab has paused in the United States because the
Omicron VOC has reduced susceptibility to these anti-SARS-CoV-2 mAbs.20,21
References
1. Jiang S, Hillyer C, Du L. Neutralizing antibodies against SARS-CoV-2 and other human coronaviruses. Trends
Immunol. 2020;41(5):355-359. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32249063.
2. Centers for Disease Control and Prevention. COVID data tracker: variant proportions. 2022. Available at:
https://covid.cdc.gov/covid-data-tracker/#variant-proportions. Accessed August 16, 2022.
3. Cao Y, Yisimayi A, Jian F, et al. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection.
Nature. 2022;Published online ahead of print. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35714668.
Key: BAM = bamlanivimab; bpm = beats per minute; BEB = bebtelovimab; CAS = casirivimab; DM = diabetes mellitus; ETE = etesevimab; IMD = imdevimab; IV =
intravenous; mAb = monoclonal antibody; PEP = post-exposure prophylaxis; PHVL = persistently high viral load; RCT = randomized controlled trial; RT-PCR = reverse
transcription polymerase chain reaction; SOT = sotrovimab; SpO2 = oxygen saturation; VL = viral load; VOC = variant of concern
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. 2021;Published online ahead
of print. 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 may contain antibodies to SARS-CoV-2 that
could help suppress viral replication.1 In August 2020, the Food and Drug Administration (FDA) issued
an Emergency Use Authorization (EUA) for COVID-19 convalescent plasma (CCP) for the treatment of
hospitalized patients with COVID-19. The EUA was revised in February 2021 to limit the authorization
to the use of high-titer CCP for the treatment of hospitalized patients with COVID-19 who are early
in their disease course or who have impaired humoral immunity.2 In December 2021, the EUA was
revised again to authorize the use of CCP only in outpatients or inpatients with COVID-19 who have
immunosuppressive disease or who are receiving immunosuppressive treatment. The testing criteria
used to identify CCP products that contain high levels of anti-SARS-CoV-2 antibodies (i.e., high-titer
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
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends against the use of CCP
that was collected prior to the emergence of the Omicron (B.1.1.529) variant for the treatment of
COVID-19 (AIII).
• The Panel recommends against the use of CCP for the treatment of COVID-19 in hospitalized,
immunocompetent patients (AI).
• There is insufficient evidence for the Panel to recommend either for or against the use of high-titer
CCP that was collected after the emergence of Omicron for the treatment of immunocompromised
patients and nonhospitalized, immunocompetent patients with COVID-19.
Rationale
Regarding the Use of COVID-19 Convalescent Plasma Collected Prior to the Emergence of
the Omicron Variant
The Omicron variant is the dominant SARS-CoV-2 variant currently circulating in the United States.
Although in vitro data suggest that the CCP collected from vaccinated and unvaccinated individuals who
have recovered from Omicron infection exhibits neutralizing activity against the Omicron variant,3-6 it
is not possible to extrapolate the potential clinical efficacy of CCP in the current clinical context. This is
due in part to the following factors:
• The current supply of CCP products in the United States was not generated from donors who had
recovered from Omicron infection.7
• Many CCP clinical trials were completed before the Omicron surge, and their results may not
inform the current clinical context.
• The current approaches to testing CCP titers do not account for potential differences in the
neutralizing activity of CCP products against currently circulating variants.
Furthermore, it is difficult to interpret the available data on the in vitro antiviral activity of CCP, since
the published studies use a variety of assays to characterize the neutralizing activity of CCP, and the
COVID-19 Treatment Guidelines 251
Considerations in Pregnancy
The safety and efficacy of using CCP during pregnancy have not been evaluated in clinical trials, and
published data on its use in pregnant individuals with COVID-19 are limited to case reports.51 Pathogen-
Considerations in Children
The safety and efficacy of CCP have not been systematically evaluated in pediatric patients. Published
literature on its use in children is limited to case reports and case series. A few clinical trials that are
evaluating the use of CCP in children are ongoing. The use of 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 FDA for use in immunocompetent patients.
As an alternative to CCP, several antiviral therapies are available for the treatment of children with
COVID-19 who are at high risk of progressing to severe disease. The use of these products in children
may be considered on a case-by-case basis. See Special Considerations in Children for more information.
Adverse Effects
The available data suggest that serious adverse reactions following the administration of CCP are
infrequent and consistent with the risks associated with plasma infusions for other indications. These
risks include transfusion-transmitted infections (e.g., HIV, hepatitis B, hepatitis C), allergic reactions,
anaphylactic reactions, febrile nonhemolytic reactions, transfusion-related acute lung injury, transfusion-
associated circulatory overload, and hemolytic reactions. Hypothermia, metabolic complications, and
post-transfusion purpura have also been described.2,18,54
Additional risks of CCP transfusion include a theoretical risk of antibody-dependent enhancement of
SARS-CoV-2 infection and a theoretical risk of long-term immunosuppression. In the CONCOR-1 trial,
higher levels of full transmembrane spike IgG were associated with worse outcomes, suggesting that the
use of CCP with nonfunctional anti-SARS-CoV-2 antibodies may be harmful.19 A subgroup analysis in
the REMAP-CAP trial showed potential harm in patients who received CCP transfusions more than 7
days after being hospitalized.20
When considering the use of CCP in patients with a history of severe allergic or anaphylactic transfusion
reactions, consultation with a transfusion medicine specialist is advised.
Clinical Trials
Several randomized clinical trials that are evaluating the use of CCP for the treatment of COVID-19 are
underway. Please see ClinicalTrials.gov for the latest 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. Lusvarghi S, Pollett SD, Neerukonda SN, et al. SARS-CoV-2 Omicron neutralization by therapeutic
antibodies, convalescent sera, and post-mRNA vaccine booster. bioRxiv. 2021;Preprint. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34981057.
4. Khan K, Karim F, Cele S, et al. Omicron infection of vaccinated individuals enhances neutralizing immunity
against the Delta variant. medRxiv. 2022;Preprint. Available at:
COVID-19 Treatment Guidelines 255
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for CCP.
The studies summarized below are those that have had the greatest impact on the Panel’s recommendations.
References
1. Writing Committee for the REMAP-CAP Investigators, Estcourt LJ, Turgeon AF, et al. 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.
2. Begin 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.
3. 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.
4. Sullivan DJ, Gebo KA, Shoham S, et al. Early outpatient treatment for COVID-19 with convalescent plasma. N Engl J Med. 2022;Published online
COVID-19 Treatment Guidelines 264
Recommendation
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel to recommend
either for or against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)
immunoglobulins for the treatment of COVID-19.
Rationale
Currently, there are no clinical data on the use of SARS-CoV-2 immunoglobulins. Trials evaluating
SARS-CoV-2 immunoglobulins are in development but not yet active and enrolling participants.
Proposed Mechanism of Action and Rationale for Use in Patients with COVID-19
Concentrated antibody preparations derived from pooled plasma collected from individuals who
have recovered from COVID-19 can be manufactured as SARS-CoV-2 immunoglobulin, which
could potentially suppress the virus and modify the inflammatory response. The use of virus-specific
immunoglobulins for other viral infections (e.g., cytomegalovirus [CMV] immunoglobulin for the
prevention of post-transplant CMV infection and varicella zoster immunoglobulin for postexposure
prophylaxis of varicella in individuals at high-risk) has proven to be safe and effective; however, there
are currently no clinical data on the use of such products for COVID-19. Potential risks may include
transfusion reactions. Theoretical risks may include antibody-dependent enhancement of infection.
Clinical Data
There are no clinical data on the use of SARS-CoV-2 immunoglobulins for the treatment of COVID-19.
Similarly, there are no clinical data on use of specific immunoglobulin or hyperimmunoglobulin
products in patients with severe acute respiratory syndrome (SARS) or Middle East respiratory
syndrome (MERS).
Considerations in Pregnancy
Pathogen-specific immunoglobulins are used clinically during pregnancy to prevent varicella zoster
virus (VZV) and rabies and have also been used in clinical trials of therapies for congenital CMV
infection.
Considerations in Children
Hyperimmunoglobulin has been used to treat several viral infections in children, including VZV,
respiratory syncytial virus, and CMV; efficacy data on their use for other respiratory viruses is limited.
• The information in this table is based on data from investigational trials evaluating these products for the treatment or prevention of
COVID-19. The table includes dose recommendations from the FDA EUAs for patients who meet specified criteria.
• There are limited or no data on 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 or prevention 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 combination therapies for the treatment or prevention of
COVID-19 are unknown. Clinicians are encouraged to report AEs to the FDA Medwatch program.
• For drug interaction information, please refer to product labels and visit the Liverpool COVID-19 Drug Interactions website.
• For the Panel’s recommendations on using the drugs listed in this table, please refer to the Anti-SARS-CoV-2 Monoclonal Antibodies,
Therapeutic Management of Nonhospitalized Adults With COVID-19, and Prevention of SARS-CoV-2 Infection sections of the Guidelines.
References
1. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for bebtelovimab. 2022. Available at:
https://www.fda.gov/media/156152/download.
2. Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for Evusheld (tixagevimab co-packaged with
cilgavimab). 2022. Available at: https://www.fda.gov/media/154701/download.
3. Marano G, Vaglio S, Pupella S, et al. Convalescent plasma: new evidence for an old therapeutic tool? Blood Transfus. 2016;14(2):152-157. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/26674811.
4. 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 for the COVID-19 Treatment Guidelines Panel’s (the
Panel) recommendations on the use of the following immunomodulators for hospitalized patients according to their
disease severity:
• Corticosteroids: dexamethasone
• Interleukin-6 inhibitors: tocilizumab (or sarilumab)
• Janus kinase (JAK) inhibitors: baricitinib (or tofacitinib)
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
• Fluvoxamine
• Granulocyte-macrophage colony-stimulating factor inhibitors for hospitalized patients
• Inhaled corticosteroids
The Panel recommends against the use of the following immunomodulators for the treatment of COVID-19, except in a
clinical trial:
• Baricitinib plus tocilizumab (AIII)
• Canakinumab (BIIa)
• Colchicine for nonhospitalized patients (BIIa)
• Intravenous immunoglobulin (IVIG) (non-SARS-CoV-2-specific) for the treatment of patients with acute COVID-19
(AIII). This recommendation should not preclude the use of IVIG for multisystem inflammatory syndrome in children
(MIS-C) or when it is otherwise indicated.
• Bruton’s tyrosine kinase inhibitors (e.g., acalabrutinib, ibrutinib, zanubrutinib) (AIII)
• JAK inhibitors other than baricitinib and tofacitinib (e.g., ruxolitinib) (AIII)
• Siltuximab (BIII)
The Panel recommends against the use of the following immunomodulators for the treatment of COVID-19:
• Colchicine for hospitalized patients (AI)
Multiple 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 has not shown any benefits and
may cause harm.3,4 The COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations for the
use of corticosteroids in hospitalized patients with COVID-19 are based on results from these clinical
trials (see Tables 4a and 4b for more information). There are no data to support the use of systemic
corticosteroids in nonhospitalized patients with COVID-19.
Recommendations
For Nonhospitalized Adults With COVID-19
• See Therapeutic Management of Nonhospitalized Adults With COVID-19 for the Panel’s
recommendations on the use of dexamethasone or other systemic corticosteroids in certain
nonhospitalized patients.
• 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 these patients.
• 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).
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. Mortality at 28 days was lower among the patients who received
COVID-19 Treatment Guidelines 273
Dose of Dexamethasone
The COVID STEROID 2 trial is the largest study to date that has investigated the use of different doses
of corticosteroids in people with COVID-19.6 This multicenter trial randomized hospitalized patients to
receive up to 10 days of once-daily dexamethasone 6 mg (n = 485) or dexamethasone 12 mg (n = 497).
The median number of days alive without life support at 28 days after randomization was 20.5 days
in the dexamethasone 6 mg arm (IQR 4.0–28.0 days) and 22.0 days in the dexamethasone 12 mg arm
(IQR 6.0–28.0 days), yielding an adjusted mean difference of 1.3 days (95% CI, 0–2.6; P = 0.07). No
differences were found in 28- or 90-day mortality between the arms. Approximately 12% of the patients
in each arm received either an interleukin-6 inhibitor or a kinase inhibitor during the study. While these
conventional analyses did not reach statistical significance, a preplanned Bayesian analysis found a
higher probability of benefit and a lower probability of harm for the 12-mg dose than for the 6-mg dose.7
A smaller randomized controlled trial reported a shorter time to clinical improvement and a lower
frequency of adverse events in patients with COVID-19 who received a lower dose of dexamethasone
(8 mg IV once daily) compared to those who received higher doses (8 mg IV 2 or 3 times daily). A
lower proportion of patients in the low-dose group died within 60 days compared to the intermediate-
and high-dose groups (17% vs. 30% and 41%, respectively; P = 0.06). It is worth noting that this study
included <200 participants.8
A third small, open-label, randomized trial (with <100 participants) found no difference in the median
number of ventilator-free days at 28 days after randomization between patients who received higher
COVID-19 Treatment Guidelines 274
Recommendation
• There is insufficient evidence for the Panel to recommend either for or against the use of inhaled
corticosteroids for the treatment of COVID-19.
Rationale
Inhaled budesonide was studied in 2 open-label randomized controlled trials in outpatients with mild
symptoms of COVID-19.22,23 The small STOIC trial suggested that initiating inhaled budesonide in
adult outpatients with mild COVID-19 may reduce the need for urgent care or emergency department
assessment or hospitalization.22 PRINCIPLE, a larger, open-label trial in nonhospitalized patients with
COVID-19 who were at high risk of disease progression, found that using inhaled budesonide did not
affect the rate of hospitalization or death but did reduce the time to self-reported recovery.23 The findings
from these trials should be interpreted with caution given the open-label design of the studies and other
limitations.
Inhaled ciclesonide was studied in 2 double-blind, randomized, placebo-controlled trials in outpatients
with mild COVID-19. The primary endpoint in 1 study was time to alleviation of COVID-19-related
symptoms. In this study, the use of inhaled ciclesonide did not reduce the time to self-reported recovery,
but the therapy did reduce the number of subsequent COVID-related emergency department visits
or hospitalizations. The robustness of this conclusion is uncertain given the small number of events,
which is likely due to the relatively small number of participants with comorbidities.24 In the smaller
CONTAIN study, the combined use of inhaled and intranasal ciclesonide did not improve the resolution
of fever and/or respiratory symptoms by Day 7.25
The studies described above that evaluated the use of inhaled corticosteroid therapy in outpatients with
mild COVID-19 have identified inconsistent effects of this therapy on subsequent hospitalization, and
similar placebo-controlled trials have not demonstrated that this therapy improves the time to symptom
resolution. The placebo-controlled studies did not enroll enough patients who were at high risk of
disease progression; therefore, further studies in this population are needed. For additional information
on these trials, see Table 4b.
Considerations in 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 women with threatened preterm
delivery.34,35
A short course of dexamethasone for the treatment of COVID-19 during pregnancy offers the potential
benefit of decreased maternal mortality and a low risk of fetal adverse effects. Therefore, the Panel
recommends using dexamethasone in hospitalized pregnant patients with COVID-19 who are
mechanically ventilated (AIII) or who require supplemental oxygen but are not mechanically ventilated
(BIII).
Considerations in Children
The safety and effectiveness of using dexamethasone or other corticosteroids for COVID-19 treatment
have not been sufficiently evaluated in pediatric patients. Caution is warranted when using data from
clinical trials that enrolled adults to inform treatment recommendations for children, particularly younger
children and those who are less severely ill. The Panel recommends using dexamethasone for children
with COVID-19 who require high-flow oxygen, noninvasive ventilation, mechanical ventilation, or
extracorporeal membrane oxygenation (BIII). Corticosteroids are not routinely recommended for
pediatric patients who require only low levels of oxygen support (i.e., administered via a nasal cannula
only) but could be considered on a case-by-case basis. The use of dexamethasone for the treatment of
severe COVID-19 in children who are profoundly immunocompromised has not been evaluated and may
be harmful; therefore, such use should be considered only if the benefit is expected to outweigh the risks.
The dexamethasone dosing regimen for pediatric patients is dexamethasone 0.15 mg/kg per dose (with a
maximum dose of 6 mg) once daily for up to 10 days. There is insufficient evidence to recommend either
for or against the use of inhaled corticosteroids in pediatric patients with COVID-19.
Methylprednisolone or another corticosteroid should be used in combination with IV immunoglobulin
for the initial treatment of multisystem inflammatory syndrome in children (MIS-C) (AIIb). The dosing
regimen for initial therapy is methylprednisolone 1 to 2 mg/kg IV once daily or another glucocorticoid
at an equivalent dose. See Therapeutic Management of Hospitalized Pediatric Patients With Multisystem
Inflammatory Syndrome in Children (MIS-C) (With Discussion on Multisystem Inflammatory
COVID-19 Treatment Guidelines 277
Clinical Trials
Several clinical trials that are evaluating the use of corticosteroids for the treatment of COVID-19 are
underway or in development. Please see ClinicalTrials.gov for the latest information.
References
1. WHO Rapid Evidence Appraisal for COVID-19 Therapies Working Group, Sterne JAC, Murthy S, et al.
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:
https://www.ncbi.nlm.nih.gov/pubmed/34492533.
3. RECOVERY Collaborative Group, Horby P, Lim WS, et al. 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.
4. Crothers K, DeFaccio R, Tate J, et al. Dexamethasone in hospitalised coronavirus-19 patients not on intensive
respiratory support. Eur Respir J. 2021;Published online ahead of print. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34824060.
5. Tomazini BM, Maia IS, Cavalcanti AB, et al. Effect of dexamethasone on days alive and ventilator-free in
patients with moderate or severe acute respiratory distress syndrome and COVID-19: the CoDEX randomized
clinical trial. JAMA. 2020;324(13):1307-1316. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32876695.
6. COVID STEROID 2 Trial Group, Munch MW, Myatra SN, et al. Effect of 12 mg vs 6 mg of dexamethasone
on the number of days alive without life support in adults with COVID-19 and severe hypoxemia: the COVID
STEROID 2 randomized trial. JAMA. 2021;326(18):1807-1817. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34673895.
7. Granholm A, Munch MW, Myatra SN, et al. Dexamethasone 12 mg versus 6 mg for patients with COVID-19
and severe hypoxaemia: a pre-planned, secondary Bayesian analysis of the COVID STEROID 2 trial.
Intensive Care Med. 2022;48(1):45-55. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34757439.
8. Toroghi N, Abbasian L, Nourian A, et al. Comparing efficacy and safety of different doses of dexamethasone
in the treatment of COVID-19: a three-arm randomized clinical trial. Pharmacol Rep. 2022;74(1):229-240.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34837648.
9. Maskin LP, Bonelli I, Olarte GL, et al. High- versus low-dose dexamethasone for the treatment of COVID-
19-related acute respiratory distress syndrome: a multicenter, randomized open-label clinical trial. J Intensive
Care Med. 2022;37(4):491-499. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34898320.
10. 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.
11. REMAP-CAP Investigators, Gordon AC, Mouncey PR, et al. 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.
12. 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.
13. Dequin PF, Heming N, Meziani F, et al. Effect of hydrocortisone on 21-day mortality or respiratory support
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.
RECOVERY: Open-Label RCT of Dexamethasone in Hospitalized Patients With COVID-19 in the United Kingdom1
Key Inclusion Criterion: Participant Characteristics: Key Limitations:
• Hospitalized with suspected or • Mean age 66 years; 64% men; 73% White • Open-label study
laboratory-confirmed SARS-CoV-2 • 56% had ≥1 comorbidity; 24% with DM • Published data did not include results for key
infection secondary endpoints (e.g., cause-specific
• 89% had laboratory-confirmed SARS-CoV-2 infection
Key Exclusion Criteria: mortality, need for renal replacement), AEs, and
• Median duration of DEX therapy: 7 days key subgroups (e.g., patients with comorbidities).
• Physician determination that risks of • At randomization:
participation were too great based on • Patients who required supplemental oxygen (but
patient’s medical history • 16% received MV or ECMO not MV) had variable severity of illness. It is
• 60% required supplemental oxygen but not MV unclear whether all patients in this group benefited
• An indication for corticosteroid therapy
from DEX or whether benefit is restricted to those
outside of the study • 24% required no supplemental oxygen
requiring higher levels of supplemental oxygen.
Interventions: • Received RDV: <1% in both arms
• Patients aged >80 years were preferentially
• DEX 6 mg IV or PO once daily plus SOC • Received tocilizumab or sarilumab: 2% in DEX arm vs. 3% in assigned to receive supplemental oxygen therapy
for up to 10 days or until discharge (n = SOC arm (and not MV).
2,104) Primary Outcome: • High mortality in this study may limit the
• SOC alone (n = 4,321) • All-cause mortality at 28 days: generalizability of results to populations with a
lower baseline mortality.
Primary Endpoint: • All patients: 23% in DEX arm vs. 26% in SOC arm (age-
• All-cause mortality at 28 days adjusted rate ratio 0.83; 95% CI, 0.75–0.93; P < 0.001) Interpretation:
• Patients who required MV or ECMO at randomization: 29% • In hospitalized patients with severe COVID-19
in DEX arm vs. 41% in SOC arm (rate ratio 0.64; 95% CI, who required supplemental oxygen, DEX reduced
0.51–0.81) mortality at 28 days. The greatest benefit was seen
• Patients who required supplemental oxygen but not MV at in those receiving MV at randomization.
randomization: 23% in DEX arm vs. 26% in SOC arm (rate • There was no survival benefit for DEX in patients
ratio 0.82; 95% CI, 0.72–0.94) who did not require supplemental oxygen at
• Patients who did not require supplemental oxygen at randomization.
CoDEX: Open-Label RCT of Dexamethasone in Patients With Moderate or Severe ARDS and COVID-19 in Brazil2
Key Inclusion Criteria: r andomization: 18% in DEX arm vs. 14% in SOC arm (rate Key Limitations:
• Confirmed or suspected SARS-CoV-2 ratio 1.19, 95% CI, 0.92–1.55) • Open-label study
infection Participant Characteristics: • Underpowered; enrollment stopped after release
• Received MV within 48 hours of meeting • Mean age 61 years; 63% men of data from the RECOVERY trial.
criteria for moderate to severe ARDS • Patients discharged before 28 days were not
• Comorbidities:
(PaO2/FiO2 ≤200 mm Hg) followed for rehospitalization or mortality.
• Obesity: 31% in DEX arm vs. 24% in SOC arm
Key Exclusion Criteria: • High mortality in this study may limit the
• DM: 38% in DEX arm vs. 47% in SOC arm
• Received immunosuppressive drugs in generalizability of results to populations with a
past 21 days • Vasopressor use: 66% in DEX arm vs. 68% in SOC arm lower baseline mortality.
• Death expected within 24 hours • Mean PaO2/FiO2: 131 mm Hg in DEX arm vs. 133 mm Hg in • More than one-third of those randomized to
SOC arm receive SOC also received corticosteroids.
Interventions: • Median duration of DEX therapy: 10 days Interpretation:
• DEX 20 mg IV once daily for 5 days, then • No patients received RDV or tocilizumab
DEX 10 mg IV once daily for 5 days or • Compared with SOC alone, DEX increased the
until ICU discharge (n = 151) • 35% in SOC arm received corticosteroids for indications such number of days alive and free of MV over 28 days
as bronchospasm or septic shock in patients with COVID-19 and moderate to severe
• SOC alone (n = 148)
Primary Outcome: ARDS.
Primary Endpoint:
• Mean number of days alive and free from MV by Day 28: 7 in
• Number of days alive and free from MV DEX arm vs. 4 in SOC arm (P = 0.04)
by Day 28
Secondary Outcomes:
Key Secondary Endpoints:
• No differences between arms in all-cause mortality (56% vs.
• All-cause mortality by Day 28 62%), number of ICU-free days, duration of MV, or score on
• Number of ICU-free days by Day 28 6-point OS
• Duration of MV by Day 28 • Mean SOFA score at Day 7: 6.1 in DEX arm vs. 7.5 in SOC arm
• Score on 6-point OS at Day 15 (P = 0.004)
• SOFA score at Day 7 Other Outcome:
• Post hoc analysis of probability of death or MV by Day 15:
68% in DEX arm vs. 80% in SOC arm (OR 0.46)
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 test result• Mean age 71 years; 95% men; 27% Black, 55% White • Retrospective observational study
for SARS-CoV-2 infection • 77% did not receive IRS within 48 hours • Because nearly all patients on MV or HFNC oxygen
Key Exclusion Criteria: • 83% admitted within 1 day after positive SARS-CoV-2 test received DEX, analysis was restricted to patients
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 patients who were not on supplemental oxygen
at baseline vs. 6 days for patients on low-flow nasal cannula • Differences between the arms in other therapies
• Hospital LOS of <48 hours received
oxygen
Interventions: • Received RDV: 43% of those who received DEX vs. 13% of Interpretation:
• Corticosteroids (95% of patients those who did not • In hospitalized patients with COVID-19, the use of
received DEX) administered within 48 • Received anticoagulants: 46% of those who received DEX vs. DEX was not associated with a mortality benefit
hours of admission (n = 7,507) 10% of those who did not among those who received low-flow nasal cannula
• No corticosteroids administered (n = oxygen during the first 48 hours after admission,
Primary Outcome: but it was associated with increased mortality
7,433)
• Risk of all-cause mortality at 90 days was higher in those who among those who received no supplemental
Primary Endpoint: received DEX: oxygen during the first 48 hours after admission.
• All-cause mortality at 90 days • For combination of those not on supplemental oxygen and
those on low-flow nasal cannula oxygen: HR 1.59; 95% CI,
1.39–1.81
• For those not on supplemental oxygen: HR 1.76; 95% CI,
1.47–2.12
• For those on low-flow nasal cannula oxygen: HR 1.08; 95%
CI, 0.86–1.36
COVID STEROID 2: Blinded RCT of Dexamethasone 12 mg Versus 6 mg in Hospitalized Adults With COVID-19 and Severe Hypoxemia in Denmark, India,
Sweden, and Switzerland4,5
Key Inclusion Criteria: Participant Characteristics: Key Limitation:
• Confirmed SARS-CoV-2 infection • Median age 65 years; 31% women • The randomized intervention period was <10 days
• Requiring oxygen ≥10 L/min, NIV, CPAP, • DM: 27% in 12 mg arm vs. 34% in 6 mg arm in some patients because the trial allowed up to 4
or MV days of DEX before enrollment.
• Median time from symptom onset to hospitalization: 7 days in
Key Exclusion Criteria: both arms Interpretation:
• Treated with DEX >6 mg (or equivalent) • Received ICU care: 78% in 12 mg arm vs. 81% in 6 mg arm • Among patients with COVID-19 and severe
hypoxemia, the use of DEX 12 mg once daily did
• Treated with a corticosteroid within past • Oxygen requirements: not result in more days alive without life support
5 days • 54% on oxygen via nasal cannula or face mask (median at 28 days than DEX 6 mg once daily.
• Invasive fungal infection or active TB flow rate 23 L/min)
Interventions: • 25% on NIV
• DEX 12 mg IV once daily for up to 10 • 21% on MV
days (n = 497) • 63% received RDV; 12% received IL-6 inhibitors or JAK
• DEX 6 mg IV once daily for up to 10 days inhibitors
(n = 485) • Median duration of DEX treatment: 7 days in both arms
Primary Endpoint: Primary Outcome:
• Number of days alive without life support • Median number of days alive without life support: 22.0 in 12
(MV, circulatory support, or kidney mg arm vs. 20.5 in 6 mg arm (adjusted mean difference 1.3
replacement therapy) at 28 days days; 95% CI, 0.0–2.6; P = 0.07)
Key Secondary Endpoints: • 63.9% Bayesian probability of clinically important benefit
and 0.3% Bayesian probability of clinically important harm
• Number of days alive without life support
for DEX 12 mg
at 90 days
Secondary Outcomes:
• Number of days alive and out of hospital
at 90 days • At 90 days:
• Mortality at 90 days • Median number of days alive without life support: 84 in 12
mg arm vs. 80 in 6 mg arm (P = 0.15)
• Mortality at 28 days
• Median number of days alive and out of hospital: 62 in 12
• SAEs at 28 days
mg arm vs. 48 in 6 mg arm (P = 0.09)
• Mortality: 32% in 12 mg arm vs. 38% in 6 mg arm
(adjusted relative risk 0.87; 99% CI, 0.70–1.07; P = 0.09)
COVID STEROID 2: Blinded RCT of Dexamethasone 12 mg Versus 6 mg in Hospitalized Adults With COVID-19 and Severe Hypoxemia in Denmark, India,
Sweden, and Switzerland4,5, continued
• At 28 days:
• Mortality: 27% in 12 mg arm vs. 32% in 6 mg arm
(adjusted relative risk 0.86; 99% CI, 0.68–1.08; P = 0.10)
• SAEs, including septic shock and invasive fungal infections:
11% in 12 mg arm vs. 13% in 6 mg arm (adjusted relative
risk 0.83; 99% CI, 0.54–1.29; P = 0.27)
CAPE COVID: Double-Blind RCT of Hydrocortisone Among Critically Ill Patients With COVID-19 in France6
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Laboratory-confirmed SARS-CoV-2 • Mean age 62 years; 70% men; median BMI 28 • Underpowered; enrollment stopped after release
infection or radiographically suspected • 96% had laboratory-confirmed SARS-CoV-2 infection of data from the RECOVERY trial, resulting in
COVID-19 with ≥1 of the following: limited power to detect differences between arms.
• Median symptom duration: 9–10 days
• MV with PEEP ≥5 cm H2O • Limited information about comorbidities
• Required MV at baseline: 81%
• PaO2/FiO2 <300 mm Hg and FiO2 Interpretation:
≥50% on HFNC • Received vasopressors: 24% in hydrocortisone arm vs. 18% in
placebo arm • The use of hydrocortisone did not reduce the
• PaO2/FiO2 <300 mm Hg on reservoir proportion of patients with COVID-19 and acute
mask oxygen • Received RDV or tocilizumab: <5%
respiratory failure who experienced treatment
• Pulmonary severity index >130 • Median duration of treatment with study drug: 11 days in failure by Day 21.
hydrocortisone arm vs. 13 days in placebo arm (P = 0.25)
Key Exclusion Criteria:
Primary Outcome:
• Septic shock
• Treatment failure by Day 21: 42% in hydrocortisone arm vs.
• Do-not-intubate orders 51% in placebo arm (P = 0.29)
Interventions: Secondary Outcomes:
• Continuous IV infusion of hydrocortisone • No difference between arms in need for intubation or prone
200 mg per day for 7 days, then 100 mg positioning (too few patients received ECMO or inhaled nitric
per day for 4 days, then 50 mg per day oxide for comparisons)
for 3 days. If patient improved by Day 4,
then IV infusion of hydrocortisone 200 • Among patients who did not require MV at baseline, 50%
mg per day for 4 days, then 100 mg per in hydrocortisone arm vs. 75% in placebo arm required
day for 2 days, then 50 mg per day for 2 subsequent MV
days (n = 76) • No difference between arms in proportion of patients with
• Placebo (n = 73) nosocomial infection by Day 28
CAPE COVID: Double-Blind RCT of Hydrocortisone Among Critically Ill Patients With COVID-19 in France6, continued
Primary Endpoint: • No difference between arms in clinical status on Day 21, but
• Treatment failure (death or dependency 15% died in hydrocortisone arm vs. 27% in placebo arm (P =
on MV or high-flow oxygen) by Day 21 0.06)
• Discharged from ICU by Day 21: 57% in hydrocortisone arm
Key Secondary Endpoints:
vs. 44% in placebo arm; 23% in both arms still required MV
• Need for MV, prone positioning, ECMO,
or inhaled nitric oxide
• Nosocomial infection by Day 28
• Clinical status on Day 21, as measured
by a 5-item scale:
• Death
• In ICU and on MV
• Required high-flow oxygen therapy
• Required low-flow oxygen therapy
• Discharged from ICU
REMAP-CAP: Randomized, Open-Label, Adaptive Trial of Hydrocortisone in Patients With Severe COVID-197
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Presumed or laboratory-confirmed • Mean age 60 years; 71% men; 53% White • Open-label study
SARS-CoV-2 infection • Mean BMI 29.7–30.9 • Enrollment stopped after release of data from the
• ICU admission for respiratory or • 50% to 64% required MV RECOVERY trial.
cardiovascular support
Primary Outcome: Interpretation:
Key Exclusion Criteria: • The use of hydrocortisone did not increase the
• No difference between arms in median number of organ
• Presumed imminent death support-free days at Day 21 (0 in each arm) median number of support-free days in either the
• Systemic corticosteroid use fixed-dose or the shock-dependent hydrocortisone
• Median adjusted ORs for primary outcome for hydrocortisone arms, although early termination limited the
• >36 hours since ICU admission arms compared to no hydrocortisone arm: study’s power to detect differences between the
Interventions: • OR 1.43 (95% CrI, 0.91–2.27) with 93% Bayesian study arms.
probability of superiority for fixed-dose hydrocortisone arm
• Hydrocortisone 50 mg IV every 6 hours
for 7 days (n = 137) • OR 1.22 (95% CrI, 0.76–1.94) with 80% Bayesian
probability of superiority for shock-dependent
• Shock-dependent hydrocortisone 50 mg
hydrocortisone arm
IV every 6 hours for duration of shock
for up to 28 days (n = 146) Key Secondary Outcome:
• No hydrocortisone (n = 101) • No difference between arms in in-hospital mortality (30% in
COVID-19 Treatment Guidelines 286
REMAP-CAP: Randomized, Open-Label, Adaptive Trial of Hydrocortisone in Patients With Severe COVID-197, continued
Primary Endpoint: fi xed-dose hydrocortisone arm vs. 26% in shock-dependent
• Number of days free from respiratory hydrocortisone arm vs. 33% in no hydrocortisone arm)
and cardiovascular support by Day 21
Key Secondary Endpoint:
• In-hospital mortality
Single-Blind RCT of Methylprednisolone in Hospitalized Patients With COVID-19 Pneumonia in China8
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Laboratory-confirmed SARS-CoV-2 • Mean age 56 years; 48% men • Small sample size
infection • Median time from symptom onset to randomization: 8 days • Terminated early because of decreasing incidence
• Pneumonia confirmed by chest CT scan • At randomization, 71% were receiving oxygen via nasal of COVID-19 pneumonia at study sites
• Hospitalized on general ward for <72 cannula Interpretation:
hours
Primary Outcome: • The incidence of clinical deterioration did not differ
Key Exclusion Criteria: • Clinical deterioration at 14 days: 4.8% in both arms (OR 1.0; between the methylprednisolone and placebo
• Severe immunosuppression 95% CI, 0.134–7.442; P = 1.00) arms.
• Corticosteroid use for other diseases Secondary Outcomes:
Interventions: • No differences (all P > 0.05) between methylprednisolone arm
• Methylprednisolone 1 mg/kg per day IV and placebo arm for:
for 7 days (n = 43) • Clinical cure at 14 days: 51% vs. 58%
• Saline (n = 43) • Median number of days to clinical cure: 14 vs. 12
Primary Endpoint: • ICU admission: 4.8% in both arms
• Clinical deterioration at 14 days • In-hospital mortality: 0% vs. 2.3%
Key Secondary Endpoints: • Median number of days hospitalized: 17 vs. 13
• Clinical cure at 14 days
• Time to clinical cure
• ICU admission
• In-hospital mortality
• Number of days hospitalized
Single-Blind RCT of 3 Doses of Dexamethasone in Hospitalized Patients With Moderate to Severe COVID-19 in Iran9
Key Inclusion Criteria: Participant Characteristics: Key Limitation:
• PCR-confirmed SARS-CoV-2 infection or • Mean age: 59 years in low-dose arm vs. 59 years in • Small sample size
CT scan showing lung involvement intermediate-dose arm vs. 56 years in high-dose arm
Interpretation:
• Moderate or severe COVID-19 • 50% men
• The time to clinical response was significantly
• Requirement for supplemental oxygen • 23% with DM shorter in the low-dose DEX arm than in the
Key Exclusion Criteria: • 75% received RDV intermediate- or high-dose arms. Patients in the
low-dose arm had a higher probability of survival
• Uncontrolled DM Primary Outcome: than those in the high-dose arm.
• Active fungal or parasitic infection • Mean number of days to clinical response: 4.3 in low-dose
• On MV or receiving vasopressor therapy arm vs. 5.3 in intermediate-dose arm vs. 6.1 in high-dose arm
(P = 0.025)
Interventions:
Secondary Outcomes:
• Low dose: DEX 8 mg IV once daily for up
to 10 days (n = 47) • Mortality at 60 days: 17% in low-dose arm vs. 30% in
intermediate-dose arm vs. 41% in high-dose arm (P = 0.06)
• Intermediate dose: DEX 8 mg IV twice
daily for up to 10 days (n = 40) • AEs (leukocytosis, hyperglycemia, and secondary infections)
occurred more frequently in intermediate-dose and high-dose
• High dose: DEX 8 mg IV 3 times a day arms than in low-dose arm; however, this result was not
for up to 10 days (n = 46) statistically significant.
Primary Endpoint:
• Time to clinical response, as measured
by OS
Key Secondary Endpoints:
• Mortality at 60 days
• Occurrence of AEs
Open-Label Randomized Trial of High-Dose Versus Low-Dose Dexamethasone in Patients With COVID-19-Related ARDS in Argentina10
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Laboratory-confirmed SAR-CoV-2 • Mean age: 60 years in high-dose arm vs. 63 years in low-dose • Small, open-label study
infection arm • Trial was prematurely terminated due to low
• ARDS • 30% women enrollment rate.
• On MV for <72 hours Primary Outcomes: Interpretation:
Key Exclusion Criteria: • Median number of ventilator-free days by Day 28: 0 for both • The use of a higher dose of DEX did not
• Presumed imminent death arms (P = 0.23) increase the median number of ventilator-free
• No difference between arms in mean duration of MV by days in patients with ARDS due to COVID-19.
• Immunosuppression However, the higher dose shortened the time to
Day 28 (19 ± 18 days in high-dose arm vs. 25 ± 22 days in
• Treatment with glucocorticoids low-dose arm; P = 0.078). Cumulative hazard of successful discontinuation of MV.
Interventions: discontinuation from MV was greater in high-dose arm than
• High dose: DEX 16 mg IV once daily for low-dose arm (adjusted subdistribution HR 1.84; 95% CI,
5 days, followed by DEX 8 mg IV once 1.31–2.5; P < 0.001).
daily for 5 days (n = 49) Secondary Outcome:
• Low dose: DEX 6 mg once daily IV for 10 • All-cause mortality:
days (n = 49) • By Day 28: 41% in high-dose arm vs. 39% in low-dose arm
Primary Endpoints: (P > 0.999)
• Number of ventilator-free days by Day 28 • By Day 90: 47% in both arms (P > 0.999)
• Time to discontinuation of MV
Key Secondary Endpoint:
• All-cause mortality by Day 28 and Day 90
Key: AE = adverse event; ARDS = acute respiratory distress syndrome; BMI = body mass index; CPAP = continuous positive airway pressure; CT = computed
tomography; DEX = dexamethasone; DM = diabetes mellitus; ECMO = extracorporeal membrane oxygenation; 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/FiO2 = ratio of arterial partial pressure of oxygen to fraction of inspired
oxygen; PCR = polymerase chain reaction; PEEP = positive end-expiratory pressure; PO = orally; RCT = randomized controlled trial; RDV = remdesivir; SAE = serious
adverse event; SOC = standard of care; SOFA = sequential organ failure assessment; TB = tuberculosis
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.
STOIC: Open-Label, Phase 2 RCT of Inhaled Budesonide in Nonhospitalized Adults with Early COVID-192, continued
Interventions: Primary Outcomes: Interpretation:
• Usual care plus budesonide 800 mcg inhaled twice daily until • Median duration of budesonide use: 7 days. • In adult outpatients with mild COVID-19,
symptom resolution (n = 73) • Percentage of patients with COVID-19-related inhaled budesonide may reduce the need
• Usual care (n = 73) urgent care visit or hospitalization: 1% in for urgent care or ED assessment and/or
budesonide arm vs.14% in usual care arm (relative hospitalization.
Primary Endpoint:
risk reduction 91%).
• COVID-19-related urgent care visit, including ED visit or
hospitalization
Phase 3, Double-Blind RCT of Inhaled Ciclesonide in Nonhospitalized Patients With COVID-193
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Aged ≥12 years • Mean age 43.3 years; 55.3% women; 86.3% White • ED or hospitalization outcome based on
• Positive SARS-CoV-2 molecular or antigen diagnostic test • Mean BMI 29.4 small number of events
result in previous 72 hours • 22.3% with HTN, 7.5% with type 2 DM • Primary endpoint of time to alleviation of
• ≥1 symptom of fever, cough, or dyspnea all symptoms based on participant self-
• Higher rates of DM and asthma in ciclesonide arm report
Key Exclusion Criteria: Primary Outcome: Interpretation:
• Taken inhaled or intranasal corticosteroid within 14 days • Median time to alleviation of all COVID-19-related • Inhaled ciclesonide did not reduce time to
of enrollment or systemic corticosteroid within 90 days of symptoms: 19.0 days in ciclesonide arm vs. 19.0
enrollment reported recovery.
days in placebo arm (HR 1.08; 95% CI, 0.84–
• Unable to use an inhaler 1.38). • The robustness of the conclusion that
inhaled ciclesonide reduced COVID-19-
Interventions: Secondary Outcomes: related ED visits or hospitalization is
• Ciclesonide MDI 160 µg/actuation, 2 actuations twice a day • By Day 30, percentage of patients in whom the uncertain; there were only a small number
for 30 days (n = 197) following outcomes occurred: of events, which is most likely due to the
• Placebo MDI twice a day for 30 days (n = 203) • Alleviation of COVID-19-related symptoms: relatively low rate of comorbidities in the
70.6% in ciclesonide arm vs. 63.5% in placebo study population.
Primary Endpoint:
arm.
• Time to alleviation of all COVID-19-related symptoms by Day
• Subsequent ED visit or hospital admission for
30
COVID-19: 1.0% in ciclesonide arm vs. 5.4% in
Key Secondary Endpoints: placebo arm (OR 0.18; 95% CI, 0.04–0.85).
• Alleviation of COVID-19-related symptoms by Day 30 • Hospital admission or death: 1.5% in ciclesonide
• ED visit or hospital admission for COVID-19 by Day 30 arm vs. 3.4% in placebo arm (OR 0.45; 95% CI,
0.11–1.84).
• Hospital admission or death by Day 30
• No deaths by Day 30 in either arm.
COVID-19 Treatment Guidelines 292
CONTAIN: Double-Blind RCT of Inhaled and Intranasal Ciclesonide in Nonhospitalized Patients With COVID-194
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 comorbid condition healthy population
• ≥1 symptom of fever, cough, or shortness of breath Primary Outcome: Interpretation:
• Symptom duration ≤6 days • Percentage of patients with resolution of fever • The use of inhaled ciclesonide plus
and all respiratory symptoms at Day 7: 40% in intranasal ciclesonide did not improve
Key Exclusion Criteria: resolution of fever and respiratory
ciclesonide arm vs. 35% in placebo arm (adjusted
• Already taking an inhaled corticosteroid or taken PO or IM risk difference 5.5%; 95% CI, -7.8% to 18.8%). symptoms in nonhospitalized patients
corticosteroids within 7 days of enrollment with COVID-19.
• Unable to use an inhaler Secondary Outcomes:
• No respiratory symptoms • Percentage of patients with resolution of fever
and all respiratory symptoms at Day 14: 66% in
• Use of oxygen at home ciclesonide arm vs. 58% in placebo arm (adjusted
• COVID-19 vaccinated risk difference 7.5%; 95% CI, -5.9% to 20.8%).
Interventions: • Percentage of patients who were admitted to the
hospital by Day 14: 6% in ciclesonide arm vs. 3%
• Ciclesonide MDI 600 µg/actuation and intranasal ciclesonide
in placebo arm (adjusted risk difference 2.3%;
100 µg, both twice a day for 14 days (n = 105)
95% CI, -3.0% to 7.6%).
• Saline placebo MDI and intranasal saline, both twice a day 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
Key: BMI = body mass index; CVD = cardiovascular disease; 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
References
1. 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:
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 and
sarilumab in hospitalized patients who require supplemental oxygen, high-flow oxygen,
noninvasive ventilation (NIV), or mechanical ventilation.
• The Panel recommends against the use of anti-IL-6 mAb therapy (i.e., siltuximab) for the
treatment of COVID-19, except in a clinical trial (BIII).
Additional Considerations
• 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, particularly those who have recently received other
biologic immunomodulating drugs, 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
• Known hypersensitivity to tocilizumab or sarilumab
• Tocilizumab and sarilumab should only be given in combination with a course of dexamethasone
or an alternative corticosteroid at a dose that is equivalent to dexamethasone 6 mg. See
Corticosteroids for more information.
• Some clinicians would assess the patient’s clinical response to dexamethasone before deciding
whether tocilizumab or sarilumab is needed.
• In both the REMAP-CAP and the RECOVERY trials, 29% of patients received a second dose of
tocilizumab at the discretion of their treating physician. However, there is insufficient evidence for
the Panel to recommend either for or against a second dose of tocilizumab.5,6
• Cases of severe and disseminated strongyloidiasis have been reported in patients with COVID-19
during treatment with tocilizumab and corticosteroids.7,8 Many clinicians would initiate empiric
COVID-19 Treatment Guidelines 295
Rationale
The results of the RECOVERY and REMAP-CAP trials provide consistent evidence that tocilizumab,
when coadministered with corticosteroids, offers a modest survival benefit in certain patients with
COVID-19 who are severely ill, who are rapidly deteriorating and have increasing oxygen needs, and
who have a significant inflammatory response.5,6 However, the Panel found it challenging to define the
specific patient populations that would benefit from this intervention. If tocilizumab is not available,
sarilumab may be used as an alternative because it has demonstrated a similar clinical benefit in
improving survival and reducing the duration of organ support in the REMAP-CAP trial.10 However, the
Panel recommends sarilumab only when tocilizumab is not available or is not feasible to use (BIIa)
because the evidence of efficacy is more extensive for tocilizumab than for sarilumab. In addition,
sarilumab is currently only approved for use as a subcutaneous (SQ) injection in the United States.
The data on the efficacy of siltuximab in patients with COVID-19 are currently limited.11
Sarilumab
Sarilumab is a recombinant humanized anti-IL-6 receptor mAb that is approved by the FDA for use
in patients with rheumatoid arthritis. It is available as an SQ formulation and is not approved for the
treatment of cytokine release syndrome.
Clinical Data for COVID-19
The clinical data on the use of sarilumab as a treatment for COVID-19 are summarized in Table 6c.
An adaptive Phase 2 and 3 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
COVID-19 Treatment Guidelines 297
References
1. Yoshikawa T, Hill T, Li K, Peters CJ, Tseng CT. 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. REMAP-CAP Investigators, Gordon AC, Mouncey PR, et al. 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.
6. 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.
7. 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.
8. 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.
9. Stauffer WM, Alpern JD, Walker PF. COVID-19 and dexamethasone: a potential strategy to avoid steroid-
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
• 45% on conventional oxygen
supplemental oxygen; and subsequently selected for secondary
• 41% on HFNC oxygen or NIV randomization/tocilizumab trial
• CRP ≥75 mg/L
• 14% on MV
Key Exclusion Criterion: Interpretation:
• 82% on corticosteroids
• Non-SARS-CoV-2 infection • Among hospitalized COVID-19 patients
Primary Outcomes: with hypoxemia and elevated CRP,
Interventions: • 28-day all-cause mortality: 31% in tocilizumab arm vs. 35% in usual tocilizumab was associated with reduced
• Single weight-based dose of tocilizumab care arm (rate ratio 0.85; 95% CI, 0.76–0.94; P = 0.003) all-cause mortality and shorter time to
(maximum 800 mg) and possible second • 28-day all-cause mortality among those who required MV at baseline: discharge alive.
dose (n = 2,022) 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 alive from hospital within 28 days: 57% in
tocilizumab arm vs. 50% in usual care arm (rate ratio 1.22; 95% CI,
Key Secondary Endpoints: 1.12–1.33; P < 0.0001)
• Time to discharge from hospital, alive, • Median time to discharge: 19 days in tocilizumab arm vs. 28 days in
within 28 days usual care arm
• Among those not on MV at enrollment, • Proportion not on MV at baseline who died or required MV within 28
death or receipt of MV within 28 days days: 35% in tocilizumab arm vs. 42% in usual care arm (rate ratio
0.84; 95% CI, 0.77–0.92; P < 0.0001)
COVID-19 Treatment Guidelines 301
REMAP-CAP: Open-Label, Adaptive-Platform RCT of Tocilizumab and Sarilumab in Adults With COVID-19 in 21 Countries in Europe and North America2,3
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 • 86% PCR-confirmed SARS-CoV-2 infection after which patients were randomized
COVID-19 to active arms only; enrollment in the
• Median 14 hours from ICU admission to enrollment tocilizumab and sarilumab arms was
• Receipt of MV, NIV, or cardiovascular • At baseline: partially nonconcurrent with the SOC
support arm, and although comparisons to the
• 68% on HFNC oxygen or NIV
Key Exclusion Criteria: • 32% on MV SOC arm were adjusted for time period,
• >24 hours after ICU admission there is a possibility of bias.
• On corticosteroids: 67% in SOC arm, 82% in tocilizumab arm,
• Presumption of imminent death 89% in sarilumab arm Interpretation:
• Immunosuppression Primary Outcomes • Among patients with respiratory
failure who were within 24 hours of
• ALT >5 times ULN Tocilizumab vs. SOC:
ICU admission, the tocilizumab and
Interventions: • Median organ support-free days: 7 in tocilizumab arm vs. 0 in SOC arm sarilumab arms had higher rates of in-
• SOC plus 1 of the following (drug selection • Improved composite outcome, by ordinal scale: median aOR 1.46 hospital survival and shorter durations
based on provider preference, availability, or (95% CrI, 1.13–1.87) of organ support than the SOC arm.
adaptive probability): • Highest CRP tercile: aOR 1.87 (95% CrI, 1.35–2.59) • These results were reported in a preprint
• Single dose tocilizumab 8 mg/kg IV and • Outcomes consistent across subgroups according to oxygen and are consistent with those for a
possible second dose in 12–24 hours (n requirement at baseline smaller cohort previously published in a
= 952) peer-reviewed article.
• Single dose sarilumab 400 mg IV (n = Sarilumab vs. SOC: • The treatment effect appeared to be
485) • Median organ support-free days: 9 in sarilumab arm vs. 0 in SOC arm strongest in the highest CRP tercile.
• SOC alone (n = 406) • Improved composite outcome, by ordinal scale: median aOR 1.50 • Tocilizumab and sarilumab were
(95% CrI, 1.13–2.00) similarly effective, with a 99%
Primary Endpoint:
• Highest CRP tercile: aOR 1.85 (95% CrI, 1.24–2.69) probability of noninferiority of sarilumab.
• Composite of in-hospital mortality and
organ support-free days to Day 21 (ordinal • Outcomes consistent across subgroups according to oxygen
scale) requirements at study entry
COVACTA: Double-Blind RCT of Tocilizumab in Hospitalized Adults With COVID-19 in 9 Countries in Europe and North America4
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• PCR-confirmed SARS-CoV-2 infection • Mean age 61 years; 70% men; 58% White • Modest power to detect differences in
• Hypoxemia • 30% on HFNC oxygen or NIV Day 28 clinical status
• Bilateral chest infiltrates • 38% on MV • More patients in placebo arm
than tocilizumab arm received
Key Exclusion Criteria: • 25% with multiorgan failure corticosteroids.
• Presumption of imminent death • Received corticosteroids at entry or during follow-up: 36% in
tocilizumab arm, 55% in placebo arm Interpretation:
• Presence of active non-SARS-CoV-2 • There was no difference in Day 28
infection Primary Outcome: clinical status or survival between the
Interventions: • Day 28 clinical status: no significant difference between arms (P = tocilizumab and placebo recipients.
0.31) • The median time to discharge was
• Single dose of tocilizumab 8 mg/kg and
possible second dose, plus SOC (n = 294) Secondary Outcomes: significantly shorter in the tocilizumab
• Placebo plus SOC (n = 144) • Median time to discharge: 20 days in tocilizumab arm vs. 28 days in arm than in the placebo arm.
placebo arm (HR 1.35; 95% CI, 1.02–1.79) • Although the result was not statistically
Primary Endpoint:
• Median ICU LOS: 9.8 days in tocilizumab arm vs. 15.5 days in significant, the tocilizumab arm had a
• Day 28 clinical status (ordinal score) shorter ICU LOS than the placebo arm.
placebo arm (difference 5.8 days, 95% CI, –15.0 to 2.9)
Key Secondary Endpoints: • Day 28 mortality: 20% in tocilizumab arm vs. 19% in placebo arm (P
• Time to discharge = 0.94)
• ICU LOS
• Day 28 mortality
EMPACTA: Double-Blind RCT of Tocilizumab in Hospitalized Adults With COVID-19 in 6 Countries in North America, South America, and Africa5
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
• Presumption of imminent death
• Corticosteroids: 80% in tocilizumab arm, 88% in placebo arm progression to MV, ECMO, or death by
• Receiving NIV or MV Day 28 but did not reduce 28-day all-
• RDV: 53% in tocilizumab arm, 59% in placebo arm
Interventions: cause mortality.
Primary Outcome:
• Single dose of tocilizumab 8 mg/kg plus
SOC, possible second dose (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,
• Placebo plus SOC (n = 128)
0.33–0.97; 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 (ordinal score) 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 States6
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Laboratory-confirmed SARS-CoV-2 • Median age 60 years; 58% men; 45% Hispanic/Latinx, 43% White • Wide confidence intervals due to small
infection • 50% with BMI ≥30; 49% with HTN; 31% with DM sample size and low event rates
• ≥2 of the following conditions: • 80% receiving oxygen ≤6 L/min; 4% receiving HFNC oxygen; 16% • Few patients received RDV or
• Fever >38°C receiving no supplemental oxygen corticosteroids
• Pulmonary infiltrates • Concomitant medications: Interpretation:
• Need for supplemental oxygen • Corticosteroids: 11% in tocilizumab arm, 6% in placebo arm • There was no benefit of tocilizumab in
• RDV: 33% in tocilizumab arm, 29% in placebo arm preventing MV or death, reducing the
• ≥1 of the following laboratory criteria: risk of clinical worsening, or reducing
• CRP ≥50 mg/L Primary Outcome: the time to discontinuation of oxygen.
• D-dimer >1,000 ng/mL • Day 28 MV or death: 11% in tocilizumab arm vs. 12% in placebo arm This could be due to the low rate of
(HR 0.83; 95% CI, 0.38–1.81; P = 0.64) concomitant corticosteroid use among
• LDH ≥250 U/L
the study participants.
• Ferritin >500 ng/mL Secondary Outcomes:
Key Exclusion Criteria: • Proportion with clinical worsening of disease by Day 28: 19% in
tocilizumab arm vs. 17% in placebo arm (HR 1.11; 95% CI, 0.59–
• Receipt of supplemental oxygen at rate >10 2.10; P = 0.73)
L/min
• Median days to discontinuation of oxygen: 5.0 in tocilizumab arm vs.
• Recent use of biologic agents or small- 4.9 in placebo arm (P = 0.69)
molecule immunosuppressive therapy
• 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:
• Receipt of MV or death, according to a time
to event analysis; data censored at Day 28
Key Secondary Endpoints:
• Clinical worsening by Day 28 (ordinal score)
• Discontinuation of supplemental oxygen
among patients receiving it at baseline
Double-Blind, RCT of Sarilumab in Hospitalized Adults With Severe or Critical COVID-19 in 11 Countries in Europe, North America, South America, and Asia7
Key Inclusion Criteria: Participant Characteristics: Key Limitation:
• COVID-19 pneumonia • Median age 59 years; 63% men; 77% White, 36% Hispanic/Latinx • Moderate sample size
• Requirement for supplemental oxygen or • 39% on HFNC oxygen, MV, or NIV Interpretation:
intensive care • 42% with BMI ≥30; 43% with HTN; 26% with type 2 DM • Sarilumab showed no mortality
Key Exclusion Criteria: • 20% received systemic corticosteroids before receiving intervention; benefit or reduction in time to clinical
• Low probability of surviving or remaining at 63% received ≥1 dose of corticosteroids during the study improvement in hospitalized adults with
study site COVID-19.
Primary Outcomes:
• Dysfunction of ≥2 organ systems and need • Median time to clinical improvement: 10 days in each sarilumab arm,
for ECMO or renal replacement therapy 12 days in placebo arm
Interventions: • Sarilumab 200 mg arm vs. placebo arm: HR 1.03; 95% CI,
• Sarilumab 400 mg IV (n = 173) 0.75–1.40; P = 0.96
• Sarilumab 200 mg IV (n = 159) • 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 at 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 scale
vs. placebo)
Key Secondary Endpoint:
• Survival at Day 29
REMDACTA: Double-Blind RCT of Tocilizumab and Remdesivir in Hospitalized Patients With Severe COVID‑19 Pneumonia in Brazil, Russia, Spain, and the
United States8
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Aged ≥12 years • Mean age 59 years; 40% in tocilizumab arm and 34% in placebo arm • During the trial, primary outcome
• PCR-confirmed SARS-CoV-2 infection aged ≥65 years; 63% men; 67% White changed from clinical status on Day
• Respiratory support: 28 to time to discharge or ready for
• Hospitalized with pneumonia confirmed discharge through Day 28
by CXR or CT and requiring supplemental • NIV or HFNC oxygen: 78% in tocilizumab arm, 83% in placebo
oxygen >6 L/min arm • Imbalances in patient characteristics at
baseline between arms
Key Exclusion Criteria: • MV or ECMO: 15% in tocilizumab arm, 11% in placebo arm
• Possible underrepresentation of patients
• eGFR <30 mL/min • Corticosteroid use: with rapidly progressive disease
• ALT or AST >5 times ULN • At baseline: 83% in tocilizumab arm, 86% in placebo arm Interpretation:
• Presence of non-SARS-CoV-2 infection • During trial: 88% in each arm • Compared with placebo plus RDV,
• Treatment with antivirals, CP, CQ, HCQ, JAK Primary Outcome: tocilizumab plus RDV did not shorten
inhibitors the time to discharge or ready for
• Time to discharge or ready for discharge through Day 28: 14 days in
Interventions: each arm (HR 0.97; 95% CI, 0.78–1.19; P = 0.74) discharge in patients with severe
COVID-19 pneumonia.
• Up to 10 days RDV plus: Secondary Outcomes:
• There was no difference in mortality
• Tocilizumab 8 mg/kg IV, with second • No difference between arms: between the arms.
dose within 8–24 hours if indicated (n =
• Proportion who required MV or died by Day 28: 29% in each arm;
434)
time to death not evaluable (HR 0.98; 95% CI, 0.72–1.34; P =
• Placebo (n = 215) 0.90)
Primary Endpoint: • Mean ordinal score for Day 14 clinical status: 2.8 in tocilizumab
• Time to discharge or ready for discharge arm vs. 2.9 in placebo arm (P = 0.72)
through Day 28 • Death by Day 28: 18% in tocilizumab arm vs. 20% in placebo arm;
time to death not evaluable (HR 0.95; 95% CI, 0.65–1.39; P =
Key Secondary Endpoints:
0.79)
• Time to MV or death through Day 28
• Day 14 clinical status (ordinal score)
• Time to death through Day 28
Key: ALT = alanine transaminase; AST = aspartate transaminase; BMI = body mass index; CP = convalescent plasma; CQ = chloroquine; CRP = C-reactive protein; CT
= computed tomography; CXR = chest X-ray; DM = diabetes mellitus; ECMO = extracorporeal membrane oxygenation; eGFR = estimated glomerular filtration rate;
HCQ = hydroxychloroquine; HFNC = high-flow nasal cannula; HTN = hypertension; ICU = intensive care unit; IL = interleukin; IV = intravenous; JAK = Janus kinase;
LDH = lactate dehydrogenase; LOS = length of stay; MV = mechanical ventilation; NIV = noninvasive ventilation; the Panel = the COVID-19 Treatment Guidelines
Panel; PCR = polymerase chain reaction; RCT = randomized controlled trial; RDV = remdesivir; SOC = standard of care; SpO2 = oxygen saturation; ULN = upper limit
of normal
COVID-19 Treatment Guidelines 307
Recommendations
• Baricitinib or tofacitinib is recommended in combination with dexamethasone in hospitalized
patients with evidence of inflammation and increasing oxygen needs. See Therapeutic Management
of Hospitalized Adults With COVID-19 for the COVID-19 Treatment Guidelines Panel’s (the
Panel) detailed recommendations and ratings on the use of baricitinib and tofacitinib.
• The Panel recommends against the use of JAK inhibitors other than baricitinib or tofacitinib
for the treatment of COVID-19, except in a clinical trial (AIII).
Rationale
Several large randomized controlled trials have demonstrated that some patients who require
supplemental oxygen and most patients who require a high-flow device, NIV, or mechanical ventilation
benefit from the use of dexamethasone in combination with baricitinib or tofacitinib.
In the RECOVERY trial, baricitinib was associated with a survival benefit among hospitalized
patients, with a treatment effect that was most pronounced among patients receiving NIV or oxygen
supplementation through a high-flow device.6 The COV-BARRIER trial also demonstrated a survival
benefit from baricitinib that was most pronounced among patients receiving high-flow oxygen or NIV.7
In the addendum to the COV-BARRIER trial, the benefit extended to patients receiving mechanical
ventilation.8 Data from the ACTT-29 and ACCT-410 trials support the overall safety of baricitinib and the
potential for benefit, but neither trial studied the drug in combination with dexamethasone as standard care.
The STOP-COVID study examined the use of tofacitinib in people with COVID-19 pneumonia who
were not receiving mechanical ventilation at the time of enrollment.11 The study demonstrated a survival
benefit in patients who received tofacitinib, nearly all of whom also received corticosteroids. Tofacitinib
has less clinical data support than baricitinib, but tofacitinib can be used as an alternative if baricitinib is
not available.
Clinical trial data on the use of JAK inhibitors, including baricitinib and tofacitinib, in patients with
Baricitinib
In May 2022, the FDA approved the use of baricitinib for the treatment of COVID-19 in hospitalized
adults requiring supplemental oxygen, NIV, mechanical ventilation, or ECMO.5 It is also FDA approved
for the treatment of rheumatoid arthritis.
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.13 Baricitinib has postulated antiviral effects by blocking SARS-CoV-2
from entering and infecting lung cells.14 In macaques infected with SARS-CoV-2, baricitinib reduced
inflammation and lung pathology, but an antiviral effect was not confirmed.15
Clinical Data for COVID-19
For additional details on clinical trial data for baricitinib, see Table 6d. For information on the Panel’s
recommendations for the use of baricitinib in hospitalized patients with COVID-19, see Therapeutic
Management of Hospitalized Adults With COVID-19.
Clinical Trials
Please see ClinicalTrials.gov for the latest information on studies of baricitinib for the treatment of
COVID-19.
Tofacitinib
Tofacitinib is the prototypical JAK inhibitor, predominantly selective for JAK1 and JAK3, with modest
activity against JAK2, and, as such, 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 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.16 Tofacitinib is also FDA approved for the treatment of psoriatic arthritis, juvenile
COVID-19 Treatment Guidelines 310
Ruxolitinib
Ruxolitinib is an oral JAK inhibitor selective for JAK1 and JAK2 that is currently approved for
myelofibrosis, polycythemia vera, and acute graft-versus-host disease.18 Like baricitinib, it can modulate
downstream inflammatory responses via JAK1/JAK2 inhibition and has exhibited dose-dependent
inhibition of IL-6-induced STAT3 phosphorylation.13 Ruxolitinib also has postulated antiviral effects by
blocking SARS-CoV-2 from entering and infecting lung cells.14
Clinical Data for COVID-19
A small, single-blind, Phase 2 randomized controlled trial in patients with COVID-19 in China
compared ruxolitinib 5 mg orally twice daily (n = 20) with placebo (administered as vitamin C 100 mg;
n = 21), both given in combination with standard of care. Treatment with ruxolitinib was associated with
a nonsignificant reduction in the median time to clinical improvement (12 days for ruxolitinib recipients
vs. 15 days for placebo recipients; P = 0.15), defined as a 2-point improvement on a 7-category ordinal
scale or hospital discharge. There was no difference between the arms in the median time to discharge
(17 days for ruxolitinib arm vs. 16 days for placebo arm; P = 0.94). Limitations of this study include
the small sample size.19 A Phase 3 trial of ruxolitinib in patients with COVID-19-associated acute
respiratory distress syndrome is currently in progress (ClinicalTrials.gov Identifier NCT04377620).
Clinical Trials
Please see ClinicalTrials.gov for the latest information on studies of ruxolitinib for the treatment of
COVID-19.
Considerations in Pregnancy
There is a paucity of data on the use of JAK inhibitors in pregnancy. As small-molecule drugs, JAK
inhibitors are likely to pass through the placenta; therefore, fetal risk cannot be ruled out.20 Decisions
regarding the administration of JAK inhibitors must include shared decision-making between pregnant
individuals and their health care providers, and potential maternal benefit and fetal risks should be
considered. In the decision-making process, factors to be considered include maternal COVID-19
severity, comorbidities, and gestational age. Pregnancy registries provide some outcome data on
tofacitinib use during pregnancy for other conditions (e.g., ulcerative colitis, rheumatoid arthritis,
psoriasis). Among the 33 cases reported, pregnancy outcomes were similar to those among the general
population.21-23
Considerations in Children
Please see Therapeutic Management of Hospitalized Children With COVID-19 for the Panel’s
recommendations regarding the use of baricitinib or tofacitinib in children.
Recommendation
• The Panel recommends against the use of BTK inhibitors for the treatment of COVID-19, except
in a clinical trial (AIII).
Acalabrutinib
Acalabrutinib is a second-generation, oral BTK inhibitor that is FDA approved to treat B-cell
malignancies (i.e., chronic lymphocytic leukemia/small lymphocytic lymphoma, mantle cell lymphoma).
It has a better toxicity profile than first-generation BTK inhibitors (e.g., ibrutinib) because it has less
off-target activity for other kinases.24 Acalabrutinib is proposed for use in patients with COVID-19
because it can modulate signaling that promotes inflammation.
Clinical Data for COVID-19
Data regarding acalabrutinib are limited to the results from a prospective case series of 19 patients with
severe COVID-19.25 The small sample size and lack of a control group limit evaluation of the data to
discern any clinical benefit.
Clinical Trials
Please see ClinicalTrials.gov for the latest information on studies of acalabrutinib for the treatment of
COVID-19.
Ibrutinib
Ibrutinib is a first-generation BTK inhibitor that is FDA approved to treat various B-cell malignancies26
and to prevent chronic graft-versus-host disease in stem cell transplant recipients.27 Based on results
from a small case series, ibrutinib has been theorized to reduce inflammation and protect against ensuing
lung injury in patients with COVID-19.28
Clinical Data for COVID-19
Data regarding ibrutinib are limited to those from an uncontrolled, retrospective case series of 6 patients
with COVID-19 who received the drug for a condition other than COVID-19.28 The small sample size
and lack of a control group limit evaluation of the data to discern any clinical benefit.
Clinical Trials
Please see ClinicalTrials.gov for the latest information on studies of ibrutinib for the treatment of
COVID-19.
Zanubrutinib
Zanubrutinib is a second-generation, oral BTK inhibitor that is FDA approved to treat mantle cell
lymphoma.29 It has been shown to have fewer toxicities than first-generation BTK inhibitors (e.g.,
ibrutinib) because it has less off-target activity for other kinases.30 Zanubrutinib is proposed to benefit
patients with COVID-19 by modulating signaling that promotes inflammation.
Clinical Data for COVID-19
There are no clinical data on the use of zanubrutinib to treat COVID-19.
Clinical Trials
Please see ClinicalTrials.gov for the latest information on studies of zanubrutinib for the treatment of
COVID-19.
COVID-19 Treatment Guidelines 312
Considerations in Pregnancy
There is a paucity of data on human pregnancy and BTK inhibitor use. In animal studies, acalabrutinib
and ibrutinib in doses exceeding the therapeutic human dose were associated with interference
with embryofetal development.26,31 Based on these data, use of BTK inhibitors that occurs during
organogenesis may be associated with fetal malformations. The impact of use later in pregnancy is
unknown. Risks of use should be balanced against potential benefits.
Considerations in Children
The safety and efficacy of BTK inhibitors have not been evaluated in pediatric patients with COVID-19,
and data on the use of the drugs in children with other conditions are extremely limited. The Panel
recommends against the use of BTK inhibitors for the treatment of COVID-19 in pediatric patients,
except in a clinical trial (AIII).
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
coronavirus disease 2019 (COVID-19): the perspectives of clinical immunologists from China. Clin Immunol.
2020;214:108393. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32222466.
4. Stebbing J, Phelan A, Griffin I, et al. COVID-19: combining antiviral and anti-inflammatory treatments.
Lancet Infect Dis. 2020;20(4):400-402. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32113509.
5. Baricitinib (Olumiant) [package insert]. Food and Drug Administration. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/207924s006lbl.pdf.
6. 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/.
7. 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.
8. Ely EW, Ramanan AV, Kartman CE, et al. Efficacy and safety of baricitinib plus standard of care for
the treatment of critically ill hospitalised adults with COVID-19 on invasive mechanical ventilation or
extracorporeal membrane oxygenation: an exploratory, randomised, placebo-controlled trial. Lancet Respir
Med. 2022;10(4):327-336. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35123660.
9. Kalil AC, Patterson TF, Mehta AK, et al. Baricitinib plus remdesivir for hospitalized adults with COVID-19. N
Engl J Med. 2021;384(9):795-807. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33306283.
10. Wolfe CR, Tomashek KM, Patterson TF, et al. Baricitinib versus dexamethasone for adults hospitalised with
COVID-19 (ACTT-4): a randomised, double-blind, double placebo-controlled trial. Lancet Respir Med.
2022;Published online ahead of print. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35617986.
11. Guimaraes PO, Quirk D, Furtado RH, et al. Tofacitinib in patients hospitalized with COVID-19 pneumonia. N
Engl J Med. 2021;385(5):406-415. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34133856.
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for
kinase inhibitors. The studies summarized below are those that have had the greatest impact on the Panel’s recommendations.
The information in this table may include data from preprints or articles that have not been peer reviewed. This section will be updated as new
information becomes available. Please see ClinicalTrials.gov for more information on clinical trials evaluating kinase inhibitors.
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 • Mean age 56 years; 35% women • Small sample size
infection • Median 10 days symptom onset to randomization • RDV not available during trial
• COVID-19 pneumonia on CXR or CT • At baseline: Interpretation:
• Hospitalized for <72 hours • 75% supplemental oxygen • Tofacitinib, when compared with
Key Exclusion Criteria: • 13% HFNC oxygen placebo, led to a lower risk of mortality
• Receiving NIV, MV, or ECMO at baseline or respiratory failure among hospitalized
• Use of glucocorticoids: 79% at baseline, 89% during hospitalization adults with COVID-19 pneumonia, most
• History of or current thrombosis of whom received glucocorticoids.
Primary Outcome:
• Immunosuppression or active cancer
• Mortality or respiratory failure through Day 28: 18% in tofacitinib arm
treatment
vs. 29% in placebo arm (risk ratio 0.63; 95% CI, 0.41–0.97; P = 0.04)
Interventions:
Secondary Outcome:
• Tofacitinib 10 mg PO twice daily for up to
• Mortality through Day 28: 2.8% in tofacitinib arm vs. 5.5% in placebo
14 days or until discharge (n = 144)
arm (HR 0.49; 95% CI, 0.15–1.63)
• Placebo (n = 145)
Primary Endpoint:
• Mortality or respiratory failure through Day
28
Key Secondary Endpoint:
• Mortality through Day 28
Key: AE = adverse event; ALC = absolute lymphocyte count; ALT = alanine transaminase; ANC = absolute neutrophil count; AST = aspartate transaminase; 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; IVIG = intravenous immunoglobulin; 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 = orally; 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:
COVID-19 Treatment Guidelines 321
Colchicine is an anti-inflammatory drug that is used to treat a variety of conditions, including gout,
recurrent pericarditis, and familial Mediterranean fever.1 Recently, the drug has been shown to
potentially reduce the risk of cardiovascular events in those with coronary artery disease.2 Colchicine
has several potential mechanisms of action, including reducing the chemotaxis of neutrophils, inhibiting
inflammasome signaling, and decreasing the production of cytokines, such as interleukin-1 beta.3
When colchicine is administered early in the course of COVID-19, these mechanisms could potentially
mitigate or prevent inflammation-associated manifestations of the disease. These anti-inflammatory
properties coupled with the drug’s limited immunosuppressive potential, favorable safety profile, and
widespread availability have prompted investigation of colchicine for the treatment of COVID-19.
Recommendations
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends against the use of colchicine
for the treatment of nonhospitalized patients with COVID-19, except in a clinical trial (BIIa).
• The Panel recommends against the use of colchicine for the treatment of hospitalized patients
with COVID-19 (AI).
Considerations in Children
Colchicine is most commonly used in children to treat periodic fever syndromes and autoinflammatory
conditions. Although colchicine is generally considered safe and well-tolerated in children, there are no
data on the use of the drug to treat pediatric acute COVID-19 or multisystem inflammatory syndrome in
children (MIS-C).
References
1. van Echteld I, Wechalekar MD, Schlesinger N, Buchbinder R, Aletaha D. Colchicine for acute gout. Cochrane
Database Syst Rev. 2014(8):CD006190. Available at: https://www.ncbi.nlm.nih.gov/pubmed/25123076.
2. Xia M, Yang X, Qian C. Meta-analysis evaluating the utility of colchicine in secondary prevention of coronary
artery disease. Am J Cardiol. 2021;140:33-38. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33137319.
3. Reyes AZ, Hu KA, Teperman J, et al. Anti-inflammatory therapy for COVID-19 infection: the case for
colchicine. Ann Rheum Dis. 2020;80(5):550-557. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33293273.
4. Tardif JC, Bouabdallaoui N, L’Allier PL, et al. Colchicine for community-treated patients with COVID-19
(COLCORONA): a Phase 3, randomised, double-blinded, adaptive, placebo-controlled, multicentre trial.
Lancet Respir Med. 2021;9(8):924-932. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34051877.
5. Dorward J, Yu L, Hayward G, et al. Colchicine for COVID-19 in the community (PRINCIPLE): a randomised,
controlled, adaptive platform trial. Br J Gen Pract. 2022;Published online ahead of print. Available at:
https://pubmed.ncbi.nlm.nih.gov/35440469/.
6. RECOVERY Collaborative Group. Colchicine in patients admitted to hospital with COVID-19
(RECOVERY): a randomised, controlled, open-label, platform trial. Lancet Respir Med. 2021;9(12):1419-
1426. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34672950.
7. Diaz R, Orlandini A, Castellana N, et al. Effect of colchicine vs usual care alone on intubation and
28-day mortality in patients hospitalized with COVID-19: a randomized clinical trial. JAMA Netw Open.
2021;4(12):e2141328. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34964849.
8. Deftereos SG, Giannopoulos G, Vrachatis DA, et al. Effect of colchicine vs standard care on cardiac and
inflammatory biomarkers and clinical outcomes in patients hospitalized with coronavirus disease 2019: the
GRECCO-19 randomized clinical trial. JAMA Netw Open. 2020;3(6):e2013136. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32579195.
9. Brunetti L, Diawara O, Tsai A, et al. Colchicine to weather the cytokine storm in hospitalized patients with
COVID-19. J Clin Med. 2020;9(9):2961. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32937800.
10. Salehzadeh F, Pourfarzi F, Ataei S. The impact of colchicine on the COVID-19 patients: a clinical trial study.
Research Square. 2020;Preprint. Available at: https://www.researchsquare.com/article/rs-69374/v1.
11. Sandhu T, Tieng A, Chilimuri S, Franchin G. A case control study to evaluate the impact of colchicine
on patients admitted to the hospital with moderate to severe COVID-19 infection. Can J Infect Dis Med
Microbiol. 2020;2020:8865954. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33133323.
12. Lopes MI, Bonjorno LP, Giannini MC, et al. Beneficial effects of colchicine for moderate to severe
COVID-19: a randomised, double-blinded, placebo-controlled clinical trial. RMD Open. 2021;7(1):e001455.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/33542047.
COVID-19 Treatment Guidelines 327
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 FDA-approved for the treatment of any infection.
Recommendation
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel) to
recommend either for or against the use of fluvoxamine for the treatment of COVID-19.
Rationale
Three randomized trials have studied the use of fluvoxamine for the treatment of nonhospitalized
patients with COVID-19. In STOP COVID, a contactless, double-blind randomized placebo-controlled
trial conducted in the United States among nonhospitalized adults with mild COVID-19 diagnosed
within 7 days of symptom onset, fluvoxamine (100 mg up to 3 times daily for 15 days) reduced clinical
deterioration at Day 15.3 Clinical deterioration was defined as shortness of breath plus oxygen saturation
(SpO2) <92% or hospitalization plus SpO2 <92%. This was a small study (≤80 participants per arm)
with limited cases of clinical deterioration and a short follow-up period. In addition, 24% of participants
stopped responding to surveys prior to Day 15.
The subsequent STOP COVID 2, a Phase 3 randomized controlled trial (ClinicalTrials.gov Identifier
NCT04668950) that enrolled >700 participants in the United States and Canada, was stopped for
futility by a data safety monitoring board after lower than expected case rates and treatment effect were
observed.4
TOGETHER is an adaptive platform, double-blind randomized placebo-controlled trial conducted
in Brazil.5 Nonhospitalized adults with COVID-19 and a known risk factor for progression to severe
disease were randomized to fluvoxamine 100 mg twice daily (n = 741) or placebo (n = 756) for 10
days. Fluvoxamine use was associated with a lower risk of the primary composite outcome of retention
in the emergency department for >6 hours or admission to a tertiary hospital (79 of 741 participants
[11%] in the fluvoxamine arm vs. 119 of 756 participants [16%] in the placebo arm [relative risk
0.68; 95% CrI, 0.52–0.88]). Of note, 87% of the primary outcome events were hospitalizations. There
was no statistically significant difference between study arms for the secondary outcomes of need for
hospitalization or time to symptom resolution. There was no significant difference in mortality between
study arms in the intention-to-treat (ITT) population (17 of 741 participants [2%] in the fluvoxamine
arm vs. 25 of 756 participants [3%] in the placebo arm [OR 0.69; 95% CI, 0.36–1.27]). In a secondary,
per-protocol analysis of participants who received >80% of possible doses, death was the outcome for
1 of 548 participants (<1%) in the fluvoxamine arm versus 12 of 618 participants (2%) in the placebo
COVID-19 Treatment Guidelines 329
Considerations in Pregnancy
Fluvoxamine is not thought to increase the risk of congenital abnormalities; however, the data on its use
in pregnancy are limited.7,8 The association of SSRI use in the late third trimester with a small, increased
risk of primary persistent pulmonary hypertension in newborns has not been excluded, although the
absolute risk is likely low.9 The risk of fluvoxamine use in pregnancy for the treatment of COVID-19
should be balanced with the potential benefit.
Considerations in Children
Fluvoxamine is approved by the FDA for the treatment of obsessive-compulsive disorder in children
aged ≥8 years.10 Adverse effects due to SSRI use seen in children are similar to those seen in adults,
although children and adolescents appear to have higher rates of behavioral activation and vomiting than
adults.11 There are no data on the use of fluvoxamine for the prevention or treatment of COVID-19 in
COVID-19 Treatment Guidelines 330
Clinical Trials
See ClinicalTrials.gov for the latest information on studies of fluvoxamine and COVID-19.
References
1. Rosen DA, Seki SM, Fernandez-Castaneda 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). 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. 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.
4. Lenze E. Fluvoxamine for early treatment of COVID-19: the STOP COVID clinical trials. 2021. Available
at: https://rethinkingclinicaltrials.org/news/august-20-2021-fluvoxamine-for-early-treatment-of-covid-19-the-
stop-covid-clinical-trials-eric-lenze-md/. Accessed December 8, 2021.
5. Reis G, Dos Santos Moreira-Silva EA, Silva DCM, 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. 2021;Published online ahead of print. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34717820.
6. Hemeryck A, Belpaire FM. Selective serotonin reuptake inhibitors and cytochrome P-450 mediated drug-drug
interactions: an update. Curr Drug Metab. 2002;3(1):13-37. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11876575.
7. Einarson A, Choi J, Einarson TR, Koren G. Incidence of major malformations in infants following
antidepressant exposure in pregnancy: results of a large prospective cohort study. Can J Psychiatry.
2009;54(4):242-246. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19321030.
8. Furu K, Kieler H, Haglund B, et al. Selective serotonin reuptake inhibitors and venlafaxine in early pregnancy
and risk of birth defects: population based cohort study and sibling design. BMJ. 2015;350:h1798. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/25888213.
9. Huybrechts KF, Bateman BT, Palmsten K, et al. Antidepressant use late in pregnancy and risk of persistent
pulmonary hypertension of the newborn. JAMA. 2015;313(21):2142-2151. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26034955.
10. Fluvoxamine maleate tablets [package insert]. Food and Drug Administration. 2019. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021519s012lbl.pdf.
11. Safer DJ, Zito JM. Treatment-emergent adverse events from selective serotonin reuptake inhibitors by age
group: children versus adolescents. J Child Adolesc Psychopharmacol. 2006;16(1-2):159-169. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16553536.
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations
for fluvoxamine. The studies summarized below are the randomized clinical trials that have had the greatest impact on the Panel’s
recommendations.
TOGETHER: Double-Blind, Adaptive RCT of Fluvoxamine in Nonhospitalized Patients With COVID-19 in Brazil1
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 emergency setting observation
• Laboratory-confirmed SARS-CoV-2 infection identified as mixed race endpoint has not been used in other studies
• 13% with uncontrolled HTN; 13% with type 2 DM; of interventions for nonhospitalized patients
• ≤7 days of symptoms who are at high risk for hospitalization and
50% with BMI ≥30 kg/m2
Key Exclusion Criteria: death
• Mean of 3.8 days from symptom onset to
• Use of an SSRI randomization • As this was an adaptive platform trial
• Severe mental illness where multiple investigational treatments
Primary Outcome: or placebos were being evaluated
• Cirrhosis, recent seizures, severe ventricular cardia • Proportion of patients who met the primary simultaneously, not all patients in the placebo
arrythmia composite endpoint: 11% in fluvoxamine arm vs. arm received a placebo that was matched
Interventions: 16% in placebo arm (relative risk 0.68; 95% CrI, to fluvoxamine by route of administration,
0.52–0.88) dosing frequency, or duration of therapy
• Fluvoxamine 100 mg PO twice daily for 10 days (n = 741)
Secondary Outcomes: • PP analyses are not randomized
• Placebo (route, dosing frequency, and duration for some
comparisons, and they introduce bias when
patients may have differed from fluvoxamine) (n = 756) • 87% of clinical events were hospitalizations.
adherence is associated with factors that
Primary Endpoint: • No difference between arms in COVID-19-related influence the outcome
• Composite endpoint of emergency setting observation hospitalizations: 10% in fluvoxamine arm vs. 13%
• Adherence was self-reported and not verified
for >6 hours or hospitalization due to progression of in placebo arm (OR 0.77; 95% CI, 0.55–1.05)
• No difference between arms in time to symptom Interpretation:
COVID-19 within 28 days after randomization
resolution. • Fluvoxamine reduced the proportion of
Key Secondary Endpoints: patients who met the composite endpoint of
• Adherence: 74% in fluvoxamine arm vs. 82% in
• Occurrence of COVID-19-related hospitalizations COVID-19-related hospitalization or retention
placebo arm (OR 0.62; 95% CI, 0.48–0.81). 11%
• Time to symptom resolution in fluvoxamine arm vs. 8% in placebo arm stopped in an emergency setting for >6 hours.
drug due to issues of tolerability.
TOGETHER: Double-Blind, Adaptive RCT of Fluvoxamine in Nonhospitalized Patients With COVID-19 in Brazil1, continued
• Proportion of patients who were adherent to study drugs, • Mortality (ITT): 2% in fluvoxamine arm • The use of fluvoxamine did not impact the incidence of
defined as receiving >80% of possible doses vs. 3% in placebo arm (OR 0.68; 95% COVID-19-related hospitalizations.
• Mortality in both the primary ITT population and a PP CI, 0.36–1.27) • It is difficult to define the clinical relevance of the >6-hour
population that included patients who took >80% of the • Mortality (PP): <1% in fluvoxamine emergency setting observation endpoint and apply it to
study medication doses arm vs. 2% in placebo arm (OR 0.09; practice settings in different countries.
95% CI, 0.01–0.47) • Fluvoxamine did not have a consistent impact on
mortality.
• Fluvoxamine did not impact time to symptom resolution.
STOP COVID: Double-Blind RCT of Fluvoxamine in Nonhospitalized Patients With COVID-19 in the United States2
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; • Ascertaining clinical deterioration was challenging
17% with asthma because all assessments were done remotely
Key Exclusion Criteria:
• Median of 4 days from symptom onset • 24% of patients stopped responding to follow-up prior to
• Immunocompromised to randomization Day 15 but were included in the final analysis
• Unstable medical comorbidities Primary Outcome: Interpretation:
Interventions: • Clinical deterioration: 0% in • Fluvoxamine reduced the proportion of patients who
• Fluvoxamine 50 mg PO for 1 dose, then fluvoxamine 100 fluvoxamine arm vs. 8.3% in placebo experienced clinical deterioration.
mg twice daily, then fluvoxamine 100 mg 3 times daily arm (absolute difference 8.7%; 95%
CI, 1.8% to 16.4%) • Due to significant limitations, it is difficult to draw
through Day 15 (n = 80)
definitive conclusions about the efficacy of using
• Placebo (n = 72) Secondary Outcome: fluvoxamine to treat COVID-19.
Primary Endpoint: • No patients in fluvoxamine arm
• Clinical deterioration within 15 days of randomization. and 4 patients in placebo arm were
Clinical deterioration was defined as: hospitalized.
• 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
References
1. Reis G, Dos Santos Moreira-Silva EA, Silva DCM, 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. 2021;Published online ahead of print.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34717820.
2. 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.
Recommendation
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel) to
recommend either for or against the use of GM-CSF inhibitors for the treatment of hospitalized
patients with COVID-19.
Rationale
Clinical data are lacking to definitively establish the potential benefits and risks associated with the use
of GM-CSF inhibitors in patients with COVID-19. Data from a double-blind randomized controlled trial
of lenzilumab did show a significant improvement in the primary endpoint of ventilator-free survival
through Day 28 among those who received the GM-CSF inhibitor.11 However, preliminary data from a
large, double-blind randomized trial of otilimab (primary endpoint: alive and free of respiratory failure
at Day 28) and published results of a small, double-blind, randomized trial of mavrilimumab (primary
endpoint: proportion alive and off supplemental oxygen at Day 14) did not show a survival benefit for
the GM-CSF inhibitors compared to placebo.12-14 The study populations differed; the lenzilumab and
mavrilimumab studies primarily included patients on room air or low-flow oxygen and excluded patients
receiving mechanical ventilation, whereas the otilimab study included only patients receiving high-flow
oxygen, noninvasive ventilation, or mechanical ventilation. Lenzilumab and mavrilimumab continue to
be investigated, whereas clinical development of otilimab for the treatment of COVID-19 has ceased.
Clinical Trials
See ClinicalTrials.gov for a list of ongoing clinical trials that are evaluating the use of GM-CSF
COVID-19 Treatment Guidelines 335
Adverse Effects
The primary risks associated with GM-CSF inhibitors being reported and evaluated are related to
bacterial infection. Other adverse events that have been reported with these agents include acute
kidney injury and elevated liver transaminases.10 Autoimmune pulmonary alveolar proteinosis has been
associated with a high-titer of anti-GM-CSF auto-antibodies.16
Considerations in Pregnancy
Pregnant patients have been excluded from clinical trials evaluating GM-CSF inhibitors for the
treatment of COVID-19. There is insufficient evidence to recommend for or against their use in pregnant
individuals with COVID-19.
Considerations in Children
There are no data on the use of GM-CSF inhibitors in children.
References
1. Mehta P, Porter JC, Manson JJ, et al. Therapeutic blockade of granulocyte macrophage colony-stimulating
factor in COVID-19-associated hyperinflammation: challenges and opportunities. Lancet Respir Med.
2020;8(8):822-830. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32559419.
2. Thwaites RS, Sanchez Sevilla Uruchurtu A, Siggins MK, et al. Inflammatory profiles across the spectrum of
disease reveal a distinct role for GM-CSF in severe COVID-19. Sci Immunol. 2021;6(57). Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33692097.
3. Hamilton JA. GM-CSF in inflammation and autoimmunity. Trends Immunol. 2002;23(8):403-408. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/12133803.
4. Lotfi N, Thome R, Rezaei N, et al. Roles of GM-CSF in the pathogenesis of autoimmune diseases: an update.
Front Immunol. 2019;10:1265. Available at: https://www.ncbi.nlm.nih.gov/pubmed/31275302.
5. 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.
6. Zhou Y, Fu B, Zheng X, et al. Pathogenic T-cells and inflammatory monocytes incite inflammatory storms in
severe COVID-19 patients. Natl Sci Rev. 2020;7(6):998-1002. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34676125.
7. De Luca G, Cavalli G, Campochiaro C, et al. GM-CSF blockade with mavrilimumab in severe COVID-19
pneumonia and systemic hyperinflammation: a single-centre, prospective cohort study. Lancet Rheumatol.
2020;2(8):e465-e473. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32835256.
8. Lang FM, Lee KM, Teijaro JR, Becher B, Hamilton JA. GM-CSF-based treatments in COVID-19: reconciling
opposing therapeutic approaches. Nat Rev Immunol. 2020;20(8):507-514. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32576980.
9. Temesgen Z, Assi M, Shweta FNU, et al. GM-CSF neutralization with lenzilumab in severe COVID-19
pneumonia: a case-cohort study. Mayo Clin Proc. 2020;95(11):2382-2394. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33153629.
10. Burmester GR, Feist E, Sleeman MA, et al. Mavrilimumab, a human monoclonal antibody targeting GM-CSF
receptor-alpha, in subjects with rheumatoid arthritis: a randomised, double-blind, placebo-controlled, phase I,
first-in-human study. Ann Rheum Dis. 2011;70(9):1542-1549. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21613310.
11. Temesgen Z, Burger CD, Baker J, et al. Lenzilumab in hospitalised patients with COVID-19 pneumonia
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for
GM-CSF inhibitors. The studies summarized below are those that have had the greatest impact on the Panel’s recommendations.
The information in this table may include data from preprints or articles that have not been peer reviewed. This section will be updated as new
information becomes available. Please see ClinicalTrials.gov for more information on clinical trials that are evaluating GM-CSF inhibitors.
LIVE-AIR: Double-Blind RCT of Lenzilumab in Hospitalized Patients With Severe COVID-19 Pneumonia in the United States and Brazil1,2
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Hospitalized with SARS-CoV-2 pneumonia • Mean age 61 years; 65% men; 72% White • Not powered to detect a survival
• SpO2 ≤94% on room air or required low-flow • 55% BMI ≥30 benefit
supplemental oxygen, HFNC oxygen, or NIV • At baseline: 41% received HFNC oxygen or NIV • Access to supportive care differed
across study sites
Key Exclusion Criteria: • 94% received corticosteroids; 72% received RDV; 69%
• MV or ECMO received corticosteroids and RDV Interpretation:
• Bacterial pneumonia, fungal or viral infection • Median CRP 79 mg/L • Lenzilumab improved ventilator-free
survival in participants with hypoxemia
• 48-hour survival not expected Primary Outcome: who were not receiving MV, with the
• Use of IL-1 inhibitors, IL-6 inhibitors, kinase inhibitors, • Survival without MV through Day 28: 84% in lenzilumab greatest benefit among those with
or mAbs within prior 8 weeks arm vs. 78% in placebo arm (HR 1.54; 95% CI, 1.02– lower CRP levels.
2.32; P = 0.040)
Interventions:
• 3 doses of lenzilumab 600 mg IV 8 hours apart (n = 236) Key Secondary Outcomes:
• Placebo (n = 243) • Mortality: 10% in lenzilumab arm vs. 14% in placebo arm
(HR 0.72; 95% CI, 0.42–1.23; P = 0.24)
Primary Endpoint:
• Incidence of death or requiring MV or ECMO: 15% in
• Survival without MV through Day 28 lenzilumab arm vs. 21% in placebo arm (HR 0.67; 95%
Key Secondary Endpoints: CI, 0.41–1.10; P = 0.11)
• Mortality Exploratory Outcome:
• Incidence of death or requiring MV or ECMO • Survival without MV for baseline CRP <150 mg/L: 90% in
lenzilumab arm vs. 79% in placebo arm (HR 2.54; 95%
Exploratory Endpoint:
CI, 1.46–4.41; P = 0.0009)
• Survival without MV, stratified by baseline CRP
COVID-19 Treatment Guidelines 338
MASH-COVID: Double-Blind RCT of Mavrilimumab in Hospitalized Patients With Severe COVID-19 Pneumonia and Systemic Hyperinflammation in the United
States3
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Hospitalization with SARS-CoV-2 pneumonia • Median age 57 years; 65% men; 40% African American • Very small sample size
• SpO2 <92% on room air or required supplemental oxygen • At baseline: • Ended early due to slow enrollment
• CRP >5 mg/dL • 50% required HFNC oxygen or NIV Interpretation:
Key Exclusion Criteria: • 65% received corticosteroids • Among participants with systemic
• MV • 75% received RDV hyperinflammation and severe
COVID-19 pneumonia, there was no
• ANC <1,500/mm3 Primary Outcome: evidence that use of mavrilimumab
• Uncontrolled bacterial infection • Alive and off supplemental oxygen at Day 14: 57% in improved supplemental oxygen–free
mavrilimumab arm vs. 47% in placebo arm (OR 1.48; survival by Day 14.
Interventions:
95% CI, 0.43–5.16; P = 0.76)
• Mavrilimumab 6 mg/kg as single IV infusion (n = 21)
Key Secondary Outcomes:
• Placebo (n = 19)
• Mortality at Day 28: 1 (5%) in mavrilimumab arm vs. 3
Primary Endpoint: (16%) in placebo arm (HR 3.72; 95% CI, 0.39–35.79; P =
• Alive and off supplemental oxygen at Day 14 0.22)
Key Secondary Endpoints: • Alive without respiratory failure at Day 28: 95% in
mavrilimumab arm vs. 79% in placebo arm (OR 5.33;
• Mortality at Day 28
95% CI, 0.54–52.7; P = 0.43)
• Alive without respiratory failure at Day 28
OSCAR: Double-Blind RCT of Otilimab in Patients With Severe COVID-19 Pneumonia in 17 Countries4
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Hospitalized with SARS-CoV-2 pneumonia • Mean age 59 years; 72% men; 66% White • Changes in SOC during study may
• Required HFNC oxygen, NIV, or MV ≤48 hours before • At baseline: have affected outcomes.
dosing • 77% received HFNC oxygen or NIV; 22% received MV Interpretation:
• CRP or ferritin >ULN • 83% received corticosteroids; 34% received RDV • For participants with severe COVID-19
Key Exclusion Criteria: pneumonia, use of otilimab did not
Primary Outcome: significantly reduce the probability of
• Death likely <48 hours • Alive and free of respiratory failure at Day 28: 71% in respiratory failure or death.
• Multiple organ failure otilimab arm vs. 67% in placebo arm (model-adjusted
• SOFA score >10 if in ICU difference 5.3%; 95% CI, -0.8 to 11.4; P = 0.09)
• ECMO Key Secondary Outcome:
• Dialysis • All-cause mortality at Day 60: 23% in otilimab arm vs.
• High-dose noradrenaline (>0.15 ug/kg/min) or equivalent 24% in placebo arm (model-adjusted difference -2.4%;
95% CI, -8.0 to 3.3; P = 0.41)
• >1 vasopressor
Interventions:
• Otilimab 90 mg IV as single infusion (n = 395)
• Placebo (n = 398)
Primary Endpoint:
• Alive and free of respiratory failure at Day 28
Key Secondary Endpoint:
• All-cause mortality at Day 60
Key: ANC = absolute neutrophil count; BMI = body mass index; CRP = C-reactive protein; ECMO = extracorporeal membrane oxygenation; GM-CSF = granulocyte-
macrophage colony-stimulating factor; HFNC = high-flow nasal cannula; ICU = intensive care unit; IL = interleukin; IV = intravenous; mAb = monoclonal antibody; MV
= mechanical ventilation; NIV = noninvasive ventilation; RCT = randomized controlled trial; RDV = remdesivir; SOC = standard of care; SOFA = sequential organ failure
assessment; SpO2 = oxygen saturation; ULN = upper limit of normal
References
1. Temesgen Z, Burger CD, Baker J, et al. Lenzilumab in hospitalised patients with COVID-19 pneumonia (LIVE-AIR): a Phase 3, randomised, placebo-
controlled trial. Lancet Respir Med. 2021. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34863332.
2. Temesgen Z, Kelley CF, Cerasoli F, et al. C reactive protein utilisation, a biomarker for early COVID-19 treatment, improves lenzilumab efficacy:
results from the randomised phase 3 ‘LIVE-AIR’ trial. Thorax. 2022. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35793833.
COVID-19 Treatment Guidelines 340
Recommendation
• The COVID-19 Treatment Guidelines Panel recommends against the use of non-severe acute
respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific intravenous immunoglobulin
(IVIG) for the treatment of acute COVID-19, except in a clinical trial (AIII). This
recommendation should not preclude the use of IVIG when otherwise indicated for the treatment
of complications that arise during the course of COVID-19.
Considerations in Pregnancy
IVIG is commonly used in pregnancy for other indications such as immune thrombocytopenia with an
acceptable safety profile.2,3
Considerations in Children
IVIG has been widely used in children for the treatment of a number of conditions. including Kawasaki
disease, and is generally safe.4 IVIG has been used in pediatric patients with COVID-19 and multiorgan
inflammatory syndrome in children (MIS-C), especially those with a Kawasaki disease-like presentation,
but the efficacy of IVIG in the management of MIS-C is still under investigation.
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-soluble urokinase plasminogen activator receptor (suPAR) levels ≥6 ng/mL were randomized
to receive either anakinra or placebo. The study found that patients who received anakinra had a lower
risk of clinical progression of COVID-19 than those who received placebo.4 CORIMUNO-ANA-1, a
randomized controlled trial that compared the use of anakinra to usual care in 116 hospitalized patients
who were hypoxemic but did not require high-flow oxygen or ventilation, was stopped early for futility.5
REMAP-CAP, an open-label, adaptive platform, randomized controlled trial that evaluated several
immunomodulators in patients with COVID-19 who required organ support, found that anakinra was not
effective in reducing the combined endpoint of in-hospital mortality and days of organ support.6 Although
the SAVE-MORE study suggests that suPAR levels could be used in risk stratification to identify
populations that could benefit from IL-1 inhibition, the laboratory assay that is used to assess suPAR levels
is not currently available in many countries, including the United States. After reviewing the results of the
studies discussed above and taking into consideration the fact that suPAR assays are not widely available
to guide the use of anakinra, the Panel has concluded that there is insufficient evidence to recommend
either for or against the use of anakinra for the treatment of COVID-19 in hospitalized patients.
Finally, CAN-COVID, a randomized controlled trial that evaluated canakinumab in hospitalized patients
with COVID-19 who were hypoxemic but did not require ventilatory support, reported that the use
of canakinumab did not improve the likelihood of survival without invasive mechanical ventilation.7
Because of these results, the Panel recommends against the use of canakinumab for the treatment of
COVID-19, except in a clinical trial (BIIa).
COVID-19 Treatment Guidelines 344
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 two IL-6 inhibitors, anakinra, interferon beta-1a, and a control group. 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.
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 invasive 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.6
Results
• Of the 2,274 participants who were randomized to one 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 invasive 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 noninvasive ventilation, with a few exceptions.
• The median number of organ support-free days was similar for patients who received anakinra and
COVID-19 Treatment Guidelines 345
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 a day on Days 1–3, 100 mg IV twice on
Day 4, and 100 mg IV once on Day 5) or usual care alone. Patients were eligible for enrollment if they
had laboratory-confirmed SARS-CoV-2 infection with COVID-19 pneumonia and they required >3 L/
min of supplemental oxygen. Patients who required high-flow oxygen, ventilation, or ICU admission
were excluded. The two coprimary outcomes were the proportion of patients who had died or who
needed noninvasive or invasive mechanical ventilation by Day 4 (score of >5 on the WHO-CPS) and the
proportion who survived without the need for noninvasive or invasive mechanical ventilation (including
high-flow oxygen) by Day 14.5
Results
• There was no difference between the anakinra plus usual care arm and the usual care alone arm in
the two coprimary outcomes: by Day 4, 36% of patients in the anakinra arm had died or required
high-flow oxygen or ventilation compared with 38% in the usual care arm (90% CrI, -17.1 to 12.0,
posterior probability of benefit 61%). By Day 14, 47% of patients in the anakinra arm had died or
required noninvasive or invasive mechanical ventilation compared to 51% in the usual care arm
(median HR 0.97; 90% CrI, 0.62–1.52; posterior probability of benefit 55%).
• Fifty-two percent of patients received corticosteroids at study entry.
• Serious adverse events occurred in 46% of patients in the anakinra arm compared to 38% in the
usual care arm; 11 of 59 patients (18.6%) in the anakinra arm experienced bacterial or fungal
infections compared to 4 of 55 patients (7.3%) who received usual care.
Limitations
• The limitations of this study include the small sample size, narrow eligibility criteria, and the
fact that many patients did not receive current standard-of-care therapy (e.g., corticosteroids,
remdesivir).
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 C-reactive protein
(≥ 20 mg/L) or ferritin (≥600 micrograms/L) levels. 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
COVID-19 Treatment Guidelines 346
Clinical Trials
See ClinicalTrials.gov for a list of clinical trials that are evaluating anakinra and canakinumab for the
treatment of COVID-19.
Adverse Effects
Headache, nausea, vomiting, and liver enzyme elevations can occur with both anakinra and
canakinumab.
Anakinra was not associated with any significant safety concerns when used in clinical trials for the
treatment of sepsis.12-14 Increased rates of infection were reported with prolonged anakinra use in
combination with tumor necrosis factor-alpha blockade, but not with short-term use.15
Considerations in Pregnancy
The data on using IL-1 inhibitors to treat COVID-19 in pregnant patients are currently limited. The
American College of Rheumatology recommends against the use of anakinra during pregnancy.16
Unintentional first-trimester exposure to anakinra is unlikely to be harmful, given the minimal transfer
of monoclonal antibodies across the placenta early in pregnancy.17
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. Anakinra (Kineret) [package insert]. Food and Drug Administration. 2012. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/103950s5136lbl.pdf.
4. 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. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34480127.
5. CORIMUNO-19 Collaborative Group. Effect of anakinra versus usual care in adults in hospital with
COVID-19 and mild-to-moderate pneumonia (CORIMUNO-ANA-1): a randomised controlled trial. Lancet
Respir Med. 2021;9(3):295-304. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33493450.
6. The REMAP-CAP Investigators, Derde LPG. 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.
7. Caricchio R, Abbate A, Gordeev I, et al. Effect of canakinumab vs placebo on survival without invasive
mechanical ventilation in patients hospitalized with severe COVID-19: a randomized clinical trial. JAMA.
2021;326(3):230-239. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34283183.
8. Aouba A, Baldolli A, Geffray L, et al. Targeting the inflammatory cascade with anakinra in moderate to severe
COVID-19 pneumonia: case series. Ann Rheum Dis. 2020;79(10):1381-1382. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32376597.
9. Cavalli G, De Luca G, Campochiaro C, et al. Interleukin-1 blockade with high-dose anakinra in patients with
COVID-19, acute respiratory distress syndrome, and hyperinflammation: a retrospective cohort study. Lancet
Rheumatol. 2020;2(6):e325-e331. Available at: https://pubmed.ncbi.nlm.nih.gov/32501454/.
10. Huet T, Beaussier H, Voisin O, et al. Anakinra for severe forms of COVID-19: a cohort study. Lancet
Rheumatol. 2020;2(7):e393-e400. Available at: https://pubmed.ncbi.nlm.nih.gov/32835245/.
11. Kooistra EJ, Waalders NJB, Grondman I, et al. Anakinra treatment in critically ill COVID-19 patients: a
prospective cohort study. Crit Care. 2020;24(1):688. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33302991.
Corticosteroid (Systemic)
Recommended by the Panel for the treatment of COVID-19 in certain hospitalized patients.
Dexamethasone Dose for COVID-19: • Hyperglycemia • Blood glucose • Moderate CYP3A4 inducer • If DEX is not available, an
(Systemic) • DEX 6 mg IV or PO • Secondary infections • BP • CYP3A4 substrate alternative corticosteroid (e.g.,
once daily for up to 10 • Reactivation of latent prednisone, methylprednisolone,
• Signs and hydrocortisone) can be used.
days or until hospital infections (e.g., HBV, symptoms of
discharge, whichever HSV, strongyloidiasis, new infection • The approximate total daily
comes first1 TB) dose equivalencies for these
glucocorticoids to DEX 6 mg (PO or
• Cases of disseminated IV) are:
strongyloidiasis have
been reported in • Prednisone 40 mg
patients with COVID-19 • Methylprednisolone 32 mg
during treatment with • Hydrocortisone 160 mg
corticosteroids and
tocilizumab. • A list of clinical trials is available:
Dexamethasone
COVID-19 Treatment Guidelines 350
Colchicine
Not recommended by the Panel for the treatment of COVID-19. Currently under investigation in clinical trials.
Colchicine Dose for COVID-19 in • Diarrhea • CBC • P-gp and CYP3A4 substrate • Use of colchicine should
COLCORONA Trial: • Nausea • Renal function • The risk of myopathy may be avoided in patients with
• Colchicine 0.5 mg twice be increased with the severe renal insufficiency.
• Vomiting • Hepatic function Patients with moderate renal
daily for 3 days, then once concomitant use of certain
daily for 27 days11 • Cramping HMG-CoA reductase inhibitors insufficiency who receive the
• Abdominal pain (e.g., atorvastatin, lovastatin, drug should be monitored
Dose for COVID-19 in for AEs.
• B
loating simvastatin) due to potential
COLCOVID Trial:
competitive interactions • A list of clinical trials is
• Colchicine 1.5 mg PO • Loss of appetite
mediated by P-gp and available: Colchicine
followed by 0.5 mg PO • Neuromyotoxicity CYP3A4 pathways.
within 2 hours of initial (rare)13 Availability:
• Fatal colchicine toxicity has
dose, then twice daily for • Blood dyscrasias (rare) • In the COLCORONA and
been reported in individuals
14 days or until hospital COLCOVID trials, 0.5 mg
with renal or hepatic
discharge, whichever colchicine tablets were used
impairment who used
comes first 12
for dosing; in the United
colchicine in conjunction
States, colchicine is available
with P-gp inhibitors or strong
as 0.6 mg tablets.
CYP3A4 inhibitors.
Corticosteroids (Inhaled)
Not recommended by the Panel for the treatment of COVID-19. Currently under investigation in clinical trials.
Budesonide Dose for COVID-19 in • Secondary infections • Signs of AEs • CYP3A4 substrate • A list of clinical trials is
(Inhaled) Clinical Trials: • Oral thrush involving the oral •D o not use with strong available: Inhaled Budesonide
• Budesonide 800 mcg oral • Systemic AEs (less mucosa or throat, CYP3A4 inhibitors.
inhalation twice daily until including thrush
common)
symptom resolution or for • Signs of systemic
up to 14 days14,15 corticosteroid
effects (e.g., adrenal
suppression)
Interleukin-1 Inhibitors
Not recommended by the Panel for the treatment of COVID-19. Currently under investigation in clinical trials.
Anakinra FDA-Approved Dose for • Neutropenia (particularly • CBC with • Use with TNF- • Anakinra for IV administration
Rheumatoid Arthritis: when used concomitantly differential blocking agents is not is not an approved
• Anakinra 100 mg SUBQ once with other agents that can • Liver enzymes recommended due formulation in the United
daily cause neutropenia) to increased risk of States.17
• Renal function; infection.
• Anaphylaxis and reduce dose if CrCl • A list of clinical trials is
Doses for COVID-19 in Clinical
angioedema <30 mL/min. available: Anakinra
Trials:
• Headache
• Dose and duration vary by
study. • Nausea
• Has also been used as IV • Diarrhea
infusion. • Sinusitis
• Arthralgia
• Flu-like symptoms
• Abdominal pain
• Injection site reactions
• Liver enzyme elevations
Canakinumab FDA-Approved Dose for • HSR • HSR • Binding of canakinumab • Canakinumab for IV
Systemic JIA: • Neutropenia • CBC with to IL-1 may increase administration is not an
• Canakinumab 4 mg/kg differential formation of CYP approved formulation in the
• Nasopharyngitis enzymes and alter United States.18
(maximum 300 mg) SUBQ • Liver enzymes
every 4 weeks 18 • D
iarrhea metabolism of drugs • A list of clinical trials is
• Respiratory tract that are CYP substrates. available: Canakinumab
infections • Use with TNF-
• Bronchitis blocking agents is not
recommended due to
• Gastroenteritis
potential increased risk
• Pharyngitis of infection.
• Musculoskeletal pain
• Vertigo
• Abdominal pain
• Injection site reactions
• Liver enzyme elevations
COVID-19 Treatment Guidelines 358
Key: AE = adverse event; ALC = absolute lymphocyte count; ALT = alanine transaminase; ANC = absolute neutrophil count; AST = aspartate aminotransferase; BP =
blood pressure; CBC = complete blood count; CPK = creatine phosphokinase; CrCl = creatinine clearance; CRP = C-reactive protein; 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; GI = gastrointestinal; HBV = hepatitis B virus; Hgb = hemoglobin; HSR =
hypersensitivity reaction; HSV = herpes simplex virus; HTN = hypertension; IL = interleukin; IV = intravenous; JIA = juvenile idiopathic arthritis; MAOI = monoamine
oxidase inhibitor; MDI = metered dose inhaler; MI= myocardial infarction; MV = mechanical ventilation; NaCl = sodium chloride; NIV = noninvasive ventilation;
NMBA = neuromuscular blocking agents; OAT = organic anion transporter; the Panel = the COVID-19 Treatment Guidelines Panel; PE = pulmonary embolism; P-gp=
P-glycoprotein; PLT = platelet count; PO = orally; SUBQ = subcutaneous; TB = tuberculosis; TNF = tumor necrosis factor; TRALI = transfusion-related acute lung
injury
References
1. Randomised Evaluation of COVID-19 Therapy (RECOVERY). Low-cost dexamethasone reduces death by up to one third in hospitalised patients with
severe respiratory complications of COVID-19. 2020. Available at: https://www.recoverytrial.net/news/low-cost-dexamethasone-reduces-death-by-up-
to-one-third-in-hospitalised-patients-with-severe-respiratory-complications-of-covid-19. Accessed August 3, 2022.
2. Baricitinib (Olumiant) [package insert]. Food and Drug Administration. 2022. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/
label/2022/207924s006lbl.pdf.
COVID-19 Treatment Guidelines 359
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).
Screening and Evaluation for Venous Thromboembolism
• There is insufficient evidence for the Panel to recommend either for or against routine screening for venous thrombo-
embolism (VTE) in patients with COVID-19 without signs or symptoms, regardless 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).
Anticoagulant Treatment for Thrombosis
• The Panel recommends that when diagnostic imaging is not possible, patients with COVID-19 who are highly
suspected to have thromboembolic disease be managed with therapeutic anticoagulation (AIII).
• The Panel recommends that patients with COVID-19 who require extracorporeal membrane oxygenation or
continuous renal replacement therapy or who have thrombosis related to catheters or extracorporeal filters be treated
with antithrombotic therapy as per the standard institutional protocols for those without COVID-19 (AIII).
Antithrombotic Therapy for Nonhospitalized Patients Without Evidence of Venous Thromboembolism
• In nonhospitalized patients with COVID-19, the Panel recommends against the use of anticoagulants 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.
• The Panel recommends against routinely continuing VTE prophylaxis after hospital discharge for patients with
COVID-19 unless they have another indication or are participating in a clinical trial (AIII). For patients discharged
after COVID-19-related hospitalization who are at high risk of VTE and at low risk of bleeding, there is insufficient
evidence for the Panel to recommend either for or against continuing anticoagulation unless another indication for
VTE prophylaxis exists.
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, because these 2 types of heparin have shorter half-lives and the effect can be reversed quickly,
they can be administered intravenously or subcutaneously, and they have fewer drug-drug interactions (AIII).
• 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 are a platelet count <50 x
109/L, 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 begun a therapeutic dose of heparin, treatment should continue for 14 days or until
they are transferred to the ICU or discharged from the hospital, whichever comes first.
COVID-19 Treatment Guidelines 362
Thrombolytic Therapy
Clinical trials are evaluating the effects of thrombolysis on mortality and the progression of COVID-19.
There is insufficient evidence for the Panel to recommend either for or against the use of thrombolytic
agents for VTE prophylaxis in hospitalized patients with COVID-19 outside of a clinical trial.
Hospitalized Children
A recent meta-analysis of publications on COVID-19 in children did not discuss VTE.37 Indications for
VTE prophylaxis in hospitalized children with COVID-19 should be the same as those for hospitalized
children without COVID-19 (BIII). For the Panel’s recommendations on the use of antithrombotic
therapy in children, see Therapeutic Management of Hospitalized Children With COVID-19 and
Therapeutic Management of Hospitalized Pediatric Patients With Multisystem Inflammatory Syndrome
in Children (MIS-C) (With Discussion on Multisystem Inflammatory Syndrome in Adults [MIS-A]).
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.
4. Tang N, Bai H, Chen X, et al. Anticoagulant treatment is associated with decreased mortality in severe
coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost. 2020;18(5):1094-1099. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32220112.
5. Nopp S, Moik F, Jilma B, Pabinger I, Ay C. Risk of venous thromboembolism in patients with COVID-19: a
systematic review and meta-analysis. Res Pract Thromb Haemost. 2020;4(7):1178-1191. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33043231.
6. Cohen AT, Davidson BL, Gallus AS, et al. Efficacy and safety of fondaparinux for the prevention of
venous thromboembolism in older acute medical patients: randomised placebo controlled trial. BMJ.
2006;332(7537):325-329. Available at: https://www.ncbi.nlm.nih.gov/pubmed/16439370.
7. Leizorovicz A, Cohen AT, Turpie AG, et al. Randomized, placebo-controlled trial of dalteparin for the
prevention of venous thromboembolism in acutely ill medical patients. Circulation. 2004;110(7):874-879.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/15289368.
8. Samama MM, Cohen AT, Darmon JY, et al. A comparison of enoxaparin with placebo for the prevention of
venous thromboembolism in acutely ill medical patients. N Engl J Med. 1999;341(11):793-800. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10477777.
9. Fraisse F, Holzapfel L, Couland JM, et al. Nadroparin in the prevention of deep vein thrombosis in acute
decompensated COPD. Am J Respir Crit Care Med. 2000;161(4):1109-1114. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10764298.
10. PROTECT Investigators for the Canadian Critical Care Trials Group, Australian and New Zealand Intensive
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for
antithrombotic 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 ≥2 • Median age 60 years; 57% men; mean BMI 30 • Open-label study
times ULN or any elevated D-dimer level and • 48% with HTN; 34% with DM; 7% with CVD • Only enrolled 12% of screened patients
SpO2 ≤93% on room air
• 91% had hypoxia; 6% received HFNC oxygen Interpretation:
• Hospitalized <5 days
• D-dimer: • Compared to prophylactic heparin, therapeutic
Key Exclusion Criteria: • 49% <2 times ULN heparin reduced mortality (a secondary endpoint)
• Indication for therapeutic anticoagulation • 51% ≥2 times ULN but had no effects on the composite primary
• Dual antiplatelet therapy endpoint of ICU admission, the need for NIV or
• 69% on corticosteroids MV, or death up to 28 days.
• High bleeding risk
Primary Outcome: • There were no differences between the arms in the
Interventions: • ICU admission, NIV or MV, or death: 16% in proportions of patients who experienced major
• Therapeutic UFH or LMWH for 28 days or until therapeutic arm vs. 22% in prophylactic arm (OR 0.69; bleeding events or VTE.
discharge or death (n = 228) 95% CI, 0.43–1.10)
• Prophylactic UFH or LMWH for 28 days or until Secondary Outcomes:
discharge or death (n = 237)
• All-cause death: 2% in therapeutic arm vs. 8% in
Primary Endpoint: prophylactic arm (OR 0.22; 95% CI, 0.07–0.65)
• Composite of ICU admission, NIV or MV, or • Mean organ support-free days: 26 days in therapeutic
death up to 28 days arm vs. 24 days in prophylactic arm (OR 1.41; 95% CI,
Key Secondary Endpoints: 0.9–2.21)
• All-cause death • No difference between arms for VTE (1% in therapeutic
arm vs. 3% in prophylactic arm) or major bleeding (1%
• Mean organ support-free days in therapeutic arm vs. 2% in prophylactic arm)
• VTE • Mean hospital-free days alive: 20 days in therapeutic
• Major bleeding event arm vs. 18 days in prophylactic arm (OR 1.09; 95% CI,
• Mean hospital-free days alive 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 supplemental oxygen • Median age 67 years; 54% men; mean BMI 30 • Open-label study
• D-dimer >4 times ULN or sepsis-induced • 60% with HTN; 37% with DM; 75 with CVD • Only enrolled 2% of screened patients
coagulopathy score of ≥4 • 64% received oxygen via nasal cannula; 15% received Interpretation:
• Hospitalized <72 hours high-flow oxygen or NIV; 5% received MV
• Compared to usual care, therapeutic LMWH
Key Exclusion Criteria: • 80% on corticosteroids reduced the incidence of VTE, ATE, and death.
• Indication for therapeutic anticoagulation Primary Outcomes: • For patients not in the ICU, therapeutic LMWH
• Dual antiplatelet therapy • Composite of VTE, ATE, and death within 32 days: 29% significantly reduced thrombotic events and did
in therapeutic arm vs. 42% in usual care arm (relative not increase major bleeding.
• High bleeding risk
risk 0.68; 95% CI, 0.49–0.96)
• CrCl <15 mL/min
• Death: 19% in therapeutic arm vs. 25% in usual
Interventions: care arm (relative risk 0.78; 95% CI, 0.49–1.23)
• Therapeutic LMWH until hospital discharge or • Thrombotic events: 11% in therapeutic arm vs.
primary endpoint met (n = 129) 29% in usual care arm (relative risk 0.37; 95% CI,
• Usual care of prophylactic or intermediate-dose 0.21–0.66)
LMWH until hospital discharge or primary • Non-ICU stratum composite of VTE, ATE, or death
endpoint met (n = 124) within 32 days: 17% in therapeutic arm vs. 36% in
Primary Endpoint: usual care arm (relative risk 0.46; 95% CI, 0.27–0.81)
• Composite of VTE, ATE, or death of any cause Key Safety Outcomes:
within 32 days of randomization • Major bleeding: 5% in therapeutic arm vs. 2% in usual
Key Safety Endpoint: care arm (relative risk 2.88, 95% CI, 0.59–14.02)
• Major bleeding • Non-ICU stratum major bleeding: 2% in both arms
ACTION: Open-Label RCT of Therapeutic Oral Anticoagulation (Rivaroxaban) in Hospitalized Patients With COVID-19 in Brazil4
Key Inclusion Criteria: Participant Characteristics: Key Limitations:
• Hospitalized for COVID-19 with elevated D-dimer • Median age 57 years; 60% men; mean BMI 30 • Open-label study
level • 49% with HTN; 24% with DM; 5% with coronary • Only enrolled 18% of screened patients
• Symptoms for ≤14 days disease • Longer duration of anticoagulation in the
Key Exclusion Criteria: • Critically ill: 7% in therapeutic arm; 5% in usual care rivaroxaban arm (30 days) than the prophylactic
arm anticoagulation arm (mean duration of 8 days)
• Indication for therapeutic anticoagulation
• 75% required oxygen: 60% low-flow oxygen; 8% HFNC Interpretation:
• CrCl <30 mL/min
oxygen; 1% NIV; 6% MV
• P2Y12 inhibitor therapy or aspirin >100 mg • When compared with usual care, therapeutic
• 83% on corticosteroids rivaroxaban did not reduce mortality, hospital
• High bleeding risk duration, oxygen use duration, or thrombosis.
Primary Outcomes:
Interventions: • Patients who received therapeutic rivaroxaban had
• Composite of time to death, hospital duration, and
• Therapeutic anticoagulation for 30 days: oxygen use duration: no difference between arms (win more clinically relevant nonmajor bleeding than
rivaroxaban 15 mg or 20 mg daily; if clinically ratio 0.86; 95% CI, 0.59–1.22) those who received usual care.
unstable, enoxaparin 1 mg/kg twice daily or UFH • The longer duration of therapy in the rivaroxaban
(n = 311) Secondary Outcomes:
arm may have influenced the difference in
• Usual care prophylactic anticoagulation with • No difference between therapeutic and prophylactic bleeding events.
enoxaparin or UFH during hospitalization (n = arms:
304) • Mortality: 11% vs. 8%
Primary Endpoint: • Thrombosis: 7% vs. 10%
• Hierarchical composite of time to death, hospital • Any bleeding: 12% in therapeutic arm vs. 3% in usual
duration, and oxygen use duration through Day care arm
30 • Major bleeding: 3% in therapeutic arm vs. 1% in usual
Key Secondary Endpoints: care arm
• Thrombosis, with and without all-cause death • Clinically relevant, nonmajor bleeding: 5% in
therapeutic arm vs. 1% in usual care arm
• Mortality
• Bleeding events
INSPIRATION: Open-Label RCT of Intermediate-Dose Versus Prophylactic-Dose Anticoagulant in Patients in Intensive Care With COVID-19 in Iran6
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 coronary • Not all patients received ICU-level care.
artery disease
Key Exclusion Criteria: Interpretation:
• 32% required NIV; 20% required MV
• Life expectancy <24 hours • Intermediate-dose anticoagulation did not
• 23% on vasopressors; 93% on corticosteroids; 60% significantly reduce VTE and ATE, the need for
• Indication for therapeutic anticoagulation
on RDV ECMO, or mortality.
• Overt bleeding
Primary Outcome: • Although the difference was nonsignificant,
Interventions: patients who received intermediate-dose
• Composite of adjudicated acute VTE, ATE, ECMO, or
• Intermediate-dose anticoagulation: enoxaparin 1 all-cause mortality: 46% in therapeutic arm vs. 44% in anticoagulation had more bleeding events than
mg/kg daily (n = 276) prophylactic arm (OR 1.06; 95% CI, 0.76–1.48) patients who received usual care.
• Prophylactic-dose anticoagulation (n = 286) Secondary Outcomes:
Primary Endpoint: • No difference between therapeutic and prophylactic
• Composite of adjudicated acute VTE, ATE, ECMO, arms:
or all-cause mortality within 30 days • All-cause mortality: 43% vs. 41%
Key Secondary Endpoints: • VTE: 3% both arms
• All-cause mortality • Major bleeding and clinically relevant nonmajor
• VTE bleeding: 6.3% vs. 3.1% (OR 2.02; 95% CI, 0.89–
4.61)
• Bleeding event
Key Exclusion Criteria: • 9.5% received at least 1 dose of a COVID-19 vaccine Interpretation:
• Severe renal or hepatic dysfunction Primary Outcome: • Thromboprophylaxis with enoxaparin did not
reduce the risk of hospitalization or death among
• Severe anemia or recent major bleeding • Composite of any untoward hospitalization and all- nonhospitalized, symptomatic patients with
• Receiving dual antiplatelet treatment cause death by Day 30: 8 (3%) in enoxaparin arm vs. 8 COVID-19.
(3%) in SOC arm (adjusted relative risk 0.98; 95% CI,
Interventions: 0.37–2.56; P = 0.96)
• Enoxaparin 40 mg SUBQ once daily for 14 days Key Secondary Outcomes:
(n = 234)
• Composite of major arterial and venous cardiovascular
• SOC (n = 238) events at 30 days: 2 (1%) in enoxaparin arm vs. 4 (2%)
Primary Endpoint: in SOC arm (relative risk 0.51; 95% CI, 0.09–2.74)
• Composite of any untoward hospitalization and • No major or clinically relevant nonmajor bleeding
all-cause death by Day 30 events occurred
Key Secondary Endpoints:
• Composite of major arterial and venous
cardiovascular events by Day 30
• Occurrence of bleeding events
Key: ATE = arterial thromboembolism; BMI = body mass index; CrCl = creatinine clearance; CVD = cardiovascular disease; DM = diabetes mellitus; ECMO =
extracorporeal membrane oxygenation; eGFR = estimated glomerular filtration rate; HFNC = high-flow nasal cannula; HTN = hypertension; ICU = intensive care unit;
LMWH = low-molecular-weight heparin; LOS = length of stay; MV = mechanical ventilation; NIV = noninvasive ventilation; the Panel = the COVID-19 Treatment
Guidelines Panel; RCT = randomized controlled trial; RDV = remdesivir; RT-PCR = reverse transcriptase polymerase chain reaction; SOC = standard of care; SpO2 =
oxygen saturation; SUBQ = subcutaneously; UFH = unfractionated heparin; ULN = upper limit of normal; VTE = venous thromboembolism
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 open-label design, reporting of
SARS-CoV-2 infection • 97% had laboratory-confirmed SARS-CoV-2 infection thrombotic and major bleeding events may have
influenced treatment allocation.
Key Exclusion Criteria: • At baseline:
• Hypersensitivity to aspirin • 33% on NIV or MV Interpretation:
• Recent history of major bleeding events • 34% on intermediate- or therapeutic-dose LMWH • In hospitalized patients with COVID-19, the use
of aspirin was not associated with reductions in
• Currently receiving aspirin or another antiplatelet • 60% on standard-dose LMWH 28-day mortality or the risk of progressing to MV
treatment • 7% received no thromboprophylaxis or death.
Interventions: • 94% on corticosteroids; 26% on RDV; 13% on
• Aspirin 150 mg once daily until discharge (n = tocilizumab; 6% on baricitinib
7,351) Primary Outcome:
• SOC alone (n = 7,541) • All-cause mortality at 28 days: 17% in both arms (rate
Primary Endpoint: ratio 0.96; 95% CI, 0.89–1.04; P = 0.35)
• All-cause mortality at 28 days Secondary Outcomes:
Key Secondary Endpoints: • Progression to MV or death at 28 days: 21% in aspirin
• Progression to MV or death at 28 days arm vs. 22% in SOC arm (risk ratio 0.96; 95% CI,
0.90–1.03)
• Major bleeding or 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: 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% on LMWH: • Different P2Y12 inhibitors used
• Within 48 hours of ICU admission • 19% on low-dose LMWH • Trial stopped for futility. Because equivalence for
Key Exclusion Criteria: • 59% on intermediate-dose LMWH aspirin and P2Y12 inhibitor arms was reached,
these arms were pooled for analyses.
• Bleeding risk sufficient to contraindicate • 12% therapeutic-dose LMWH
antiplatelet therapy • 98% on steroids; 21% on RDV; 44% on tocilizumab; Interpretation:
• CrCl <30 mL/min 11% on sarilumab • In critically ill patients with COVID-19, the use of
• Receiving antiplatelet therapy or NSAID • In P2Y12 inhibitor arm, 88.5% received clopidogrel, aspirin or a P2Y12 inhibitor did not reduce the
1.3% received ticagrelor, 1.3% received prasugrel, and number of organ support-free days or in-hospital
Interventions: mortality.
8.8% received an unknown P2Y12 inhibitor
• 1 of the following plus anticoagulation for 14 • Patients in pooled antiplatelet arm had more major
days or until hospital discharge, whichever came Primary Outcome: bleeding events than those in the control arm, but
first: • Data from aspirin and P2Y12 inhibitor arms were they had improved survival over 90 days.
• Aspirin 75–100 mg once daily (n = 565) pooled and reported as “pooled antiplatelet arm” in
final analysis:
• P2Y12 inhibitor (n = 455)
• Median number of organ support-free days: 7 in
• No antiplatelet therapy (control arm) (n = 529)
pooled antiplatelet arm and control arm (aOR 1.02;
Primary Endpoint: 95% CrI, 0.86–1.23; posterior probability of futility
• Number of organ support-free days by Day 21 96%)
Key Secondary Endpoints: Secondary Outcomes:
• Survival to hospital discharge • Survival to hospital discharge: 71.5% in pooled
antiplatelet arm vs. 67.9% in control arm (median-
• Survival to Day 90
adjusted OR 1.27; 95% CrI, 0.99–1.62; adjusted
• Major bleeding event by Day 14 absolute difference 5%; 95% CrI, -0.2% to 9.5%; 97%
posterior probability of 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 event by Day 14: 21 (2.1%) in pooled
antiplatelet arm vs. 2 (0.4%) in control arm (aOR 2.97;
95% CrI, 1.23–8.28; posterior probability of harm
99.4%)
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, Bradbury CA, Lawler PR, et al. 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.
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.
Vitamin D
• There is insufficient evidence for the Panel to recommend either for or against the use of vitamin D for the 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 for the
prevention of COVID-19, except in a clinical trial (BIII).
In addition to the antiviral medications and the immune-based therapies that are discussed elsewhere in
the COVID-19 Treatment Guidelines, adjunctive therapies are frequently used in the prevention and/or
treatment of COVID-19 or its complications. Some of these agents are being studied in clinical trials.
Some clinicians advocate for the use of vitamin and mineral supplements to treat respiratory viral
infections. Ongoing studies are evaluating the use of vitamin and mineral supplements for both the
treatment and prevention of SARS-CoV-2 infection.
The following sections describe the underlying rationale for using adjunctive therapies and summarize
the existing clinical trial data. Other adjunctive therapies will be added as new evidence emerges.
Since the most recent revision of this section, the results from several cohort studies, clinical trials,
and meta-analyses on the use of vitamin C in patients with COVID-19 have been published in peer-
reviewed journals or have been made available as manuscripts before peer review. However, most
of these studies had significant limitations, such as a small sample size or a lack of randomization or
blinding. In addition, the study designs had different doses or formulations of vitamin C and different
outcome measures, and the study populations included patients with varying concomitant medications
and COVID-19 disease severity. The studies summarized in this section have had the greatest impact
on the COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations. Results from adequately
powered, well-designed, and well-conducted clinical trials are needed to provide further guidance on the
role of vitamin C in the prevention and treatment of COVID-19.
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,1,2
including COVID-19.
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
No controlled trials have definitively demonstrated a clinical benefit of vitamin C in critically ill patients
with COVID-19, and the available observational data are inconclusive. Studies of vitamin C regimens
in patients with acute respiratory distress syndrome (ARDS) or sepsis not related to COVID-19 have
reported variable efficacy and few safety concerns.
Other Considerations
High circulating concentrations of vitamin C may affect the accuracy of point-of-care glucometers.16,17
References
1. 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.
2. 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.
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. 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.
5. 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.
6. 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.
7. Lamontagne F, Masse MH, Menard J, et al. Intravenous vitamin C in adults with sepsis in the intensive care
unit. N Engl J Med. 2022;386(25):2387-2398. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35704292.
8. Fowler AA III, Truwit JD, Hite RD, et al. Effect of vitamin C infusion on organ failure and biomarkers of
inflammation and vascular injury in patients with sepsis and severe acute respiratory failure: the CITRIS-ALI
randomized clinical trial. JAMA. 2019;322(13):1261-1270. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/31573637.
COVID-19 Treatment Guidelines 391
Since the last revision of this section, 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 COVID-19 Treatment
Guidelines Panel’s (the Panel) 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 rates), clear evidence that vitamin D supplementation provides protection against infection or
improves outcomes in patients with COVID-19 is still lacking.1,2
Recommendation
• There is insufficient evidence to recommend either for or against the use of vitamin D for the
prevention or treatment of COVID-19.
Rationale
Vitamin D is critical for bone and mineral metabolism. Because the vitamin D receptor is expressed
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.3 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 persons of Hispanic ethnicity and Black race.4 These
groups are overrepresented among cases of COVID-19 in the United States.5 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-19.6 High levels of vitamin D may cause
hypercalcemia and nephrocalcinosis.7
Since the last revision of this section, the results from some cohort studies and clinical trials on 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 had 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 these factors
make it difficult to compare results across studies. In addition, several studies used zinc in combination
with hydroxychloroquine, and the COVID-19 Treatment Guidelines Panel (the Panel) recommends
against the use of hydroxychloroquine for the treatment of COVID-19 (see Antiviral Drugs That Are
Approved, Authorized, or Under Evaluation for the Treatment of COVID-19).
The studies summarized below are those that have had the greatest impact on the Panel’s
recommendations. Results from adequately powered, well-designed, and well-conducted clinical trials
are needed to provide further guidance on the role of zinc in the prevention and treatment of COVID-19.
Recommendations
• 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).
Rationale
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 The relationship between zinc and COVID-19, including how zinc deficiency affects the
severity of COVID-19 and whether zinc supplements can improve clinical outcomes, is currently under
investigation. Zinc levels are difficult to measure accurately, as zinc is distributed as a component of
various proteins and nucleic acids.3 See ClinicalTrials.gov for more information about ongoing studies.
None of the results that are currently available from clinical trials provide evidence of a clinical benefit
of zinc for the treatment or prevention of COVID-19.
The recommended dietary allowance for elemental zinc is 11 mg daily for men and 8 mg daily for
nonpregnant women.4 The doses used in registered clinical trials for patients with COVID-19 vary
between studies, with a maximum dose of zinc sulfate 220 mg (50 mg of elemental zinc) twice daily.
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.4 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).
COVID-19 Treatment Guidelines 396
References
1. te Velthuis AJ, van den Worm SH, 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 August 23, 2022.
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.
7. Francis Z, Book G, Litvin C, Kalivas B. The COVID-19 pandemic and zinc-induced copper deficiency:
an important link. Am J Med. 2022;135(8):e290-e291. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/35367442.
8. 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/
COVID-19 Treatment Guidelines 397
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 these medications during acute management of COVID-19 unless discontinuation is
otherwise warranted by their clinical condition (AIIa for ACE inhibitors and ARBs; AIII for other medications).
• 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).
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 H-2
receptor antagonists,4 were hypothesized to offer potential as COVID-19 therapeutic agents. Others,
such as nonsteroidal anti-inflammatory agents (NSAIDs), 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 NSAIDs 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 these medications during acute management of COVID-19
unless discontinuation is otherwise warranted by their clinical condition (AIII). 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/or additive adverse effects.13 The decision to
continue or change a patient’s medications should be individualized based on their specific clinical
condition.
COVID-19 Treatment Guidelines 399
Summary Recommendations
Prevention of COVID-19
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends COVID-19 vaccination for all people who are
moderately or severely immunocompromised (AIII).
• All close contacts of people who are immunocompromised are strongly encouraged to be up to date on vaccination
against COVID-19 (AIII). Vaccinating household members, close contacts, and health care providers who provide care
for patients who are immunocompromised is important to protect these patients from infection.
• The Panel recommends using tixagevimab plus cilgavimab (Evusheld) as SARS-CoV-2 pre-exposure prophylaxis
(PrEP) for adults and adolescents who do not have SARS-CoV-2 infection or recent exposure to an individual with
SARS-CoV-2 infection and who are moderately to severely immunocompromised and may have an inadequate
immune response to COVID-19 vaccination (BIIb).
• There is insufficient evidence for the Panel to recommend either for or against the use of SARS-CoV-2 serologic
testing to assess for immunity or to guide clinical decisions about using COVID-19 vaccines or anti-SARS-CoV-2
monoclonal antibodies (mAbs) as PrEP in certain people.
General Management of Patients With COVID-19 Who Are Immunocompromised
• The Panel recommends that decisions regarding stopping or reducing the doses of immunosuppressive drugs
in patients with COVID-19 be made in consultation with the appropriate specialists; clinicians should consider
factors such as the underlying disease, the specific immunosuppressants being used, the potential for drug-drug
interactions, and the severity of COVID-19 (BIII).
Therapeutic Management of Nonhospitalized Patients With COVID-19 Who Are Immunocompromised
• The Panel recommends prompt treatment with antivirals or anti-SARS-CoV-2 mAbs for nonhospitalized patients with
mild to moderate COVID-19 who are immunocompromised (AIII). Specific recommendations are outlined in the text.
Therapeutic Management of Hospitalized Patients With COVID-19 Who Are Immunocompromised
• For most patients with COVID-19 who are immunocompromised, the Panel recommends using antiviral drugs and
immunomodulatory therapies at the doses and durations that are recommended for the general population (AIII).
• In some cases, immunomodulatory drug regimens may need to be adjusted to reduce the risk of drug-drug
interactions, overlapping toxicities, and secondary infections.
• There is insufficient evidence to guide clinical recommendations on using combination therapies (e.g., an antiviral
drug plus an anti-SARS-CoV-2 mAb) or extending the duration of treatment beyond the duration authorized by the
Food and Drug Administration.
• There is insufficient evidence for the Panel to recommend either for or against the use of high-titer COVID-19
convalescent plasma (CCP) in patients with COVID-19 who are immunocompromised. Some clinicians would
consider the use of CCP in patients who, in their clinical judgment, have severe or progressive COVID-19 and
an inadequate response to therapy. In these cases, clinicians should attempt to administer high-titer CCP from a
vaccinated donor who recently recovered from COVID-19 likely caused by a similar SARS-CoV-2 variant as the
patient.
• For people who are immunocompromised and have mild to moderate COVID-19 but are hospitalized for reasons
other than COVID-19, the Panel recommends promptly initiating treatment with an antiviral drug or an anti-SARS-
CoV-2 mAb (AIII).
Rating of Recommendations: A = Strong; B = Moderate; C = Weak
Rating of Evidence: I = One or more randomized trials without major limitations; IIa = Other randomized trials or
subgroup analyses of randomized trials; IIb = Nonrandomized trials or observational cohort studies; III = Expert opinion
Prevention of COVID-19
Vaccination
The Panel recommends COVID-19 vaccination for all people who are moderately or severely
immunocompromised (AIII). Vaccination is the most effective way to prevent serious outcomes and
death from SARS-CoV-2 infection, although there are certain special considerations for the timing of
vaccination and vaccine responses in people who are immunocompromised. Authorized and approved
COVID-19 vaccines in the United States are not live-virus vaccines, and they can be safely administered
to patients who are immunocompromised.16
The pivotal clinical trials that evaluated the safety and efficacy of the COVID-19 vaccines excluded
people who were severely immunocompromised; therefore, the data for this population are less robust.17,18
Observational data suggest that serological responses to vaccines may be blunted in patients who are
immunocompromised.19,20 Nevertheless, vaccination is still recommended, as it may confer partial
protection, including protection from vaccine-induced, cell-mediated immunity.4 See the Centers for
Disease Control and Prevention (CDC) webpage COVID-19 Vaccines for People Who Are Moderately or
Severely Immunocompromised for the current COVID-19 vaccination schedule for this population.
Vaccination of Close Contacts
All close contacts of people who are immunocompromised are strongly encouraged to be up to date on
vaccination against COVID-19 (AIII). Vaccinating household members, close contacts, and health care
workers who provide care to patients who are immunocompromised is important to protect these patients
from infection. There is evidence that vaccinated individuals who are infected with SARS-CoV-2 have
lower viral loads than unvaccinated individuals, and COVID-19 vaccines are associated with a reduction
in the number of SARS-CoV-2 infections not only among vaccinated individuals but also among their
household contacts.21-26
Vaccine Timing and Anti-SARS-CoV-2 Monoclonal Antibodies
Nonhospitalized patients who are immunocompromised may have received anti-SARS-CoV-2 mAbs
for the treatment of COVID-19. Vaccines can be administered at any time after anti-SARS-CoV-2 mAb
treatment.27 When tixagevimab plus cilgavimab (Evusheld) is being used as PrEP, it should not be
COVID-19 Treatment Guidelines 403
Pre-Exposure Prophylaxis
The Panel recommends using tixagevimab plus cilgavimab as SARS-CoV-2 PrEP for adults
and adolescents who do not have SARS-CoV-2 infection or recent exposure to an individual with
SARS-CoV-2 infection and who are moderately to severely immunocompromised and may have an
inadequate immune response to COVID-19 vaccination (BIIb). Information on dosing is available in
Prevention of SARS-CoV-2 Infection.
The FDA Emergency Use Authorization (EUA) for tixagevimab plus cilgavimab identifies a broad
group of immunocompromised individuals who are eligible for PrEP.31 Data suggest that some of
these individuals are at particularly high risk of inadequate vaccine responses and progression to
severe COVID-19 if infected. These individuals include, but are not limited to, those with hematologic
malignancies who are receiving active treatment, those who are within 1 year of receiving B cell-
depleting agents (e.g., rituximab, ocrelizumab, obinutuzumab, epratuzumab), CAR T-cell therapy
recipients, solid organ transplant recipients who are receiving immunosuppressive therapy, those with
severe combined immunodeficiencies, and those with HIV and low CD4 counts.4,9,32-36
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 (BIII). See Therapeutic
Management of Nonhospitalized Adults With COVID-19 for more information.
Prevention
The Panel recommends administering COVID-19 vaccines to pregnant individuals according to
the guidelines from the CDC and the Advisory Committee on Immunization Practices (ACIP) (AI).
COVID-19 vaccination is strongly recommended for pregnant individuals due to their increased
risk for severe disease.92,93 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.86 Pregnant individuals
who otherwise meet the criteria for PrEP with tixagevimab plus cilgavimab should not have this
therapy withheld due to their pregnancy status (AIII).
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 for severe COVID-19 and should be prioritized
for treatment. Providers should refer to the Panel’s recommendations in Therapeutic Management
of Nonhospitalized Adults With COVID-19 and Therapeutic Management of Hospitalized Adults
With COVID-19. Pregnant people who are immunocompromised comprise a heterogeneous group
of patients, ranging from those who are mildly immunocompromised to those who are severely
immunocompromised. The Panel recommends forming a collaborative, multidisciplinary team to make
decisions regarding the evaluation and management of pregnant patients, including the involvement of
a transplant or specialty provider, an obstetrician or maternal-fetal medicine specialist, a pediatrics or
neonatology specialist, and pharmacy services.
Treatment
Most children with mild to moderate COVID-19 will not progress to more severe illness and can be
managed with supportive care alone (AIII). Few children, if any, including children aged <18 years who
are immunocompromised, have been enrolled in clinical trials of treatments for COVID-19. 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
OpenSAFELY platform. Lancet Rheumatol. 2022;4(7):e490-e506. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/35698725.
8. Bastard P, Rosen LB, Zhang Q, et al. Autoantibodies against type I IFNs in patients with life-threatening
COVID-19. Science. 2020;370(6515). Available at: https://www.ncbi.nlm.nih.gov/pubmed/32972996.
9. Strangfeld A, Schafer M, Gianfrancesco MA, et al. Factors associated with COVID-19-related death in people
with rheumatic diseases: results from the COVID-19 Global Rheumatology Alliance physician-reported
registry. Ann Rheum Dis. 2021;80(7):930-942. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33504483.
10. Sharifian-Dorche M, Sahraian MA, Fadda G, et al. COVID-19 and disease-modifying therapies in patients
COVID-19 Treatment Guidelines 410
Summary Recommendations
• COVID-19 vaccination remains the most effective way to prevent SARS-CoV-2 infection and should be considered
the first line of prevention. The COVID-19 Treatment Guidelines Panel (the Panel) recommends COVID-19 vaccination
as soon as possible for everyone who is eligible (AI), including patients with active cancer and patients who are
receiving treatment for cancer (AIII).
• Because vaccine response rates may be lower in people with cancer, specific guidance on administering vaccines
to these individuals is provided by the Centers for Disease Control and Prevention’s Advisory Committee on
Immunization Practices. The Panel recommends following the most current COVID-19 vaccination schedule for
people with cancer.
• Vaccinating household members, close contacts, and health care providers who provide care to patients with cancer
is important to protect these patients from infection. All close contacts are strongly encouraged to get vaccinated
against COVID-19 as soon as possible (AIII).
• Patients with cancer are at high risk of progressing to serious COVID-19, and they may be eligible to receive anti-
SARS-CoV-2 monoclonal antibodies as pre-exposure prophylaxis (PrEP).
• The Panel recommends performing diagnostic molecular or antigen testing for SARS-CoV-2 in patients with cancer
who develop signs and symptoms that suggest acute COVID-19 (AIII). The Panel also recommends performing
diagnostic molecular testing in asymptomatic patients prior to procedures that require anesthesia and before
initiating cytotoxic chemotherapy and long-acting biologic therapy (BIII).
• The recommendations for treating COVID-19 in patients with cancer are the same as those for the general population
(AIII). See Therapeutic Management of Nonhospitalized Adults With COVID-19 and Therapeutic Management of
Hospitalized Adults With COVID-19 for more information.
• Clinicians should pay careful attention to potential overlapping toxicities and drug-drug interactions between drugs
that are used to treat COVID-19 (e.g., ritonavir-boosted nirmatrelvir [Paxlovid], dexamethasone) and cancer-directed
therapies, prophylactic antimicrobials, and other medications (AIII).
• Clinicians who are treating COVID-19 in patients with cancer should consult a hematologist or oncologist before
adjusting cancer-directed medications (AIII).
• 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 treatment (BIII).
People who are 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 (risk ratio 1.66; 95%
CI, 1.33–2.07) was higher in patients with cancer, 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 who are 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 It is unclear whether cancer survivors
Pre-Exposure Prophylaxis
Vaccination remains the most effective way to prevent SARS-CoV-2 infection and should be considered
the first line of prevention. However, some individuals, including some patients with cancer, cannot or
may not mount an adequate protective response to COVID-19 vaccines. These patients are at high risk
of progressing to severe COVID-19 and may be eligible to receive the anti-SARS-CoV-2 monoclonal
antibodies tixagevimab plus cilgavimab (Evusheld) as pre-exposure prophylaxis (PrEP). See Prevention
of SARS-CoV-2 Infection for more information.
General Guidance on Medical Care for Patients With Cancer During the COVID-19
Pandemic
Patients with cancer frequently engage with the health care system to receive treatment and supportive
care for cancer and/or treatment-related complications. Telemedicine can minimize the need for
COVID-19 Treatment Guidelines 419
Febrile Neutropenia
Patients with cancer and febrile neutropenia should undergo diagnostic molecular or antigen testing for
SARS-CoV-2, evaluation for other infectious agents, and treatment of neutropenic fever as standard of
care. Low-risk febrile neutropenia patients should be treated at home with oral antibiotics or intravenous
infusions of antibiotics to limit nosocomial exposure to SARS-CoV-2. Patients with high-risk febrile
neutropenia should be hospitalized per standard of care. Empiric antibiotics should be continued per
standard of care in patients who test positive for SARS-CoV-2. Clinicians should also continuously
evaluate neutropenic patients for emergent infections.
Medication 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 that are used to treat COVID-19 and cancer-directed therapies,
prophylactic antimicrobials, corticosteroids, and other medications (AIII).
COVID-19 Treatment Guidelines 421
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 meta-analysis of patient data. JCO Glob Oncol. 2020;6:799-808. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32511066.
COVID-19 Treatment Guidelines 422
Introduction
Treating COVID-19 in solid organ transplant, hematopoietic stem cell transplant (HCT), and cellular
immunotherapy recipients can be challenging due to the presence of coexisting medical conditions,
the potential for transplant-related cytopenias, and the need for chronic immunosuppressive therapy
to prevent graft rejection and graft-versus-host disease. Transplant recipients may also have a higher
risk of exposure to SARS-CoV-2 given their frequent contact with the health care system. Since
immunosuppressive agents modulate several aspects of the host’s immune response, the severity of
COVID-19 could potentially be affected by the type and intensity of the immunosuppressive effect of the
agent, as well as by specific combinations of immunosuppressive agents. Some transplant recipients have
medical comorbidities that have been associated with more severe cases of COVID-19 and a greater risk
of mortality, which makes the impact of transplantation on disease severity difficult to assess.
The International Society for Heart and Lung Transplantation, the American Society of Transplantation
(AST), the American Society for Transplantation and Cellular Therapy (ASTCT), and the European
Society for Blood and Marrow Transplantation (EBMT) provide guidance for clinicians who are caring
for transplant recipients with COVID-19 and guidance on screening potential donors and transplant or
cellular immunotherapy candidates. In addition, the American Society of Hematology offers guidance
regarding COVID-19 vaccination for transplant and cellular immunotherapy recipients. This section
of the COVID-19 Treatment Guidelines complements these sources and focuses on considerations
for managing COVID-19 in solid organ transplant, HCT, and cellular immunotherapy recipients.
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 recipients are the
same as those for nontransplant patients (AIII). See Therapeutic Management of Hospitalized Adults
With COVID-19 and Therapeutic Management of Nonhospitalized Adults With COVID-19 for more
information. The risks and benefits of each medication that is used to treat COVID-19 may be different
for transplant patients and nontransplant patients.
Pre-Exposure Prophylaxis
Vaccination remains the most effective way to prevent SARS-CoV-2 infection and should be considered
the first line of prevention. However, some individuals, including some transplant candidates and
recipients, cannot or may not mount an adequate protective response to COVID-19 vaccines. These
patients are at high risk of progressing to severe COVID-19 and may be eligible to receive the anti-
SARS-CoV-2 monoclonal antibodies (mAbs) tixagevimab plus cilgavimab (Evusheld) as pre-exposure
Assessing Donors
Living solid organ donors should be counseled on strategies to prevent infection and be monitored for
exposures and symptoms in the 14 days prior to a scheduled transplant.19 Living donors should undergo
SARS-CoV-2 reverse transcription polymerase chain reaction (RT-PCR) testing with a sample collected
from the respiratory tract within 3 days of donation. Deceased donors should be tested for SARS-CoV-2
infection using an RT-PCR assay of a sample taken from the upper respiratory tract within 72 hours of
death; ideally, the test should be performed as close to organ recovery as possible. Deceased donors can
be considered for donation if the results are negative. Lower respiratory sampling for COVID-19 testing
is required for potential lung transplant donors by the United Network for Organ Sharing.20
The Panel recommends following the guidance from medical professional organizations and assessing
all potential HCT donors for exposure to COVID-19 and clinical symptoms that are compatible with
COVID-19 before donation (AIII). HCT donors should practice good hygiene and avoid crowded places
and large gatherings during the 28 days prior to donation.21 Recommendations for screening HCT donors
are outlined in the ASTCT and EBMT guidelines.
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
that are used to prevent allograft rejection, antimicrobials that are used to prevent opportunistic
infections, and other medications. Dose modifications may be necessary for drugs that are 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.26 Clinicians who are treating COVID-19 in transplant patients should
consult a transplant specialist before adjusting immunosuppressive medications (AIII).
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
a narrow therapeutic index. 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.38
A 5-day course of ritonavir-boosted nirmatrelvir (Paxlovid) 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 medications, primarily due to the ritonavir component of the combination. Boosting
with ritonavir, a strong CYP3A inhibitor, is required to increase the exposure of nirmatrelvir to a
concentration that is effective against SARS-CoV-2. Ritonavir may also increase concentrations of
certain concomitant medications, including calcineurin and mTOR inhibitors, during the treatment
course and for ≥3 days after ritonavir is discontinued. Significant increases in the concentrations of these
drugs may lead to serious and sometimes life-threatening drug toxicities.
For nonhospitalized transplant patients who are receiving calcineurin or mTOR inhibitors as part of their
antirejection regimen, AST recommends either an anti-SARS-CoV-2 mAb or remdesivir as the first-line
therapy.39 If these drugs are not available or feasible to use, ritonavir-boosted nirmatrelvir may be used
with caution. Ritonavir-boosted nirmatrelvir should only be used when close monitoring of the patient
is possible, and clinicians should consult with transplant specialists during the treatment course. General
guidance for coadministering ritonavir-boosted nirmatrelvir with concomitant medications includes
temporarily withholding certain immunosuppressive agents (e.g., tacrolimus, everolimus, sirolimus) or
reducing the dosage of certain immunosuppressive agents (e.g., cyclosporine), monitoring the patient
closely for toxicities, and performing therapeutic drug monitoring (if possible) during and after the
treatment course of ritonavir-boosted nirmatrelvir.39,40
Some small case series have reported real-life success in using these recommendations to manage
COVID-19 Treatment Guidelines 432
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. Food and Drug Administration. Vaccines and related biological products advisory committee meeting.
February 26, 2021. 2021. Available at: https://www.fda.gov/media/146217/download.
4. Centers for Disease Control and Prevention. Current COVID-19 ACIP vaccine recommendations. 2021.
Available at: https://www.cdc.gov/vaccines/hcp/acip-recs/vacc-specific/covid-19.html. Accessed May 23,
2022.
5. 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.
6. Hallett AM, Greenberg RS, Boyarsky BJ, et al. SARS-CoV-2 messenger RNA vaccine antibody response
and reactogenicity in heart and lung transplant recipients. J Heart Lung Transplant. 2021;40(12):1579-1588.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34456108.
7. Mazzola A, Todesco E, Drouin S, et al. Poor antibody response after two doses of severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2) vaccine in transplant recipients. Clin Infect Dis. 2022;74(6):1093-
1096. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34166499.
8. Kamar N, Abravanel F, Marion O, et al. Three doses of an mRNA COVID-19 vaccine in solid-organ transplant
recipients. N Engl J of Med. 2021;385(7):661-662. Available at: https://pubmed.ncbi.nlm.nih.gov/34161700.
9. American Society of Transplantation. COVID-19 vaccine FAQ sheet. 2021. Available at:
https://www.myast.org/sites/default/files/2021_08_13%20COVID%20VACCINE%20FAQ-Prof8132021_
FINAL.pdf.
Summary Recommendations
There is current guidance from the Centers for Disease Control and Prevention, the American College of Obstetricians and
Gynecologists, and the Society for Maternal-Fetal Medicine detailing the management of pregnant patients with COVID-19.
This section of the COVID-19 Treatment Guidelines complements that guidance. The following are key considerations
regarding the management of COVID-19 in pregnancy:
• Pregnant people should be counseled about the increased risk for severe disease from SARS-CoV-2 infection and
receive recommendations on ways to protect themselves and their families from infection.
• 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 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).
• 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, and the
individual drug sections.
• There are limited data on the use of COVID-19 therapeutic agents in pregnant and lactating people. When making
decisions about treatment, pregnant or lactating people and their clinical teams should use a shared decision-
making process that takes several factors into consideration, 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 COVID-19 therapeutic agents during pregnancy, refer to the pregnancy considerations
subsections in Antiviral Therapy and Immunomodulators.
• 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 the future of breast milk delivery to the infant.
Rating of Recommendations: A = Strong; B = Moderate; C = Weak
Rating of Evidence: I = One or more randomized trials without major limitations; IIa = Other randomized trials or
subgroup analyses of randomized trials; IIb = Nonrandomized trials or observational cohort studies; III = Expert opinion
COVID-19 Vaccines
Pregnant people should be counseled about the benefits of COVID-19 vaccination, which include a
decreased risk of severe disease and hospitalization for the pregnant person and a decreased risk of
hospitalization for the infant in the first 6 months of life.12 The Society for Maternal-Fetal Medicine
(SMFM), the ACOG, and the CDC recommend that all eligible persons, including pregnant and lactating
individuals and those who are planning to become pregnant, receive a COVID-19 vaccine or vaccine
series. This includes booster doses, if the person is eligible. The CDC has published up-to-date guidance
regarding COVID-19 vaccination, including guidance for administering vaccines to pregnant and
lactating individuals. COVID-19 vaccines can be administered regardless of trimester and in concert
with other vaccines that are recommended during pregnancy.13
Pregnant people were not included in the initial COVID-19 vaccine studies. However, there is a growing
body of observational data that supports the efficacy and safety of administering COVID-19 vaccines
to this population. At this time, the mRNA COVID-19 vaccines and the recently authorized Novavax
vaccine are preferred over the Johnson & Johnson/Janssen vaccine for all eligible individuals, including
pregnant and lactating people.13,14 For the most up-to-date clinical recommendations, see the CDC
COVID-19 Treatment Guidelines 439
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.
Post-Delivery
Therapeutic management in postpartum patients should follow guidelines for nonpregnant patients.
However, the use of anticoagulation 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.20 Precautions should be taken to avoid transmission to the infant, including practicing good
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 patient is receiving therapeutic agents for
COVID-19 should be a joint 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 the future of breast milk delivery to the infant.
Specific guidance on the post-delivery management of infants born to individuals with known or
suspected SARS-CoV-2 infection, including breastfeeding recommendations, is provided by the CDC
and the American Academy of Pediatrics and in Special Considerations in Children.
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://www.ncbi.nlm.nih.gov/pubmed/33151921.
Introduction
Influenza activity during the 2021 to 2022 influenza season in the United States occurred in 2 waves
and extended from November 2021 through mid-June 2022.1 The overall severity of the 2021 to 2022
season was lower than the severity of seasonal influenza epidemics that occurred before the emergence
COVID-19 Treatment Guidelines 445
Influenza Vaccination
For Patients With Acute COVID-19 or Those Recovering From COVID-19
The Advisory Committee on Immunization Practices (ACIP) recommends offering an influenza vaccine
by the end of October to all people aged ≥6 months in the United States.4 People with acute COVID-19
should receive an inactivated influenza vaccine (BIII).
There are currently no data on the safety, immunogenicity, or efficacy of influenza vaccines in patients
with mild COVID-19 or those who are recovering from COVID-19. The safety and efficacy of
vaccinating people who have mild illnesses from other etiologies have been documented.5 Clinicians
should consider deferring influenza vaccination for symptomatic patients with COVID-19 until these
patients are no longer moderately or severely ill and have completed their COVID-19 isolation period.
People with asymptomatic SARS-CoV-2 infection or mild COVID-19 symptoms should seek influenza
vaccination when they no longer require isolation. They can be vaccinated sooner if they are in a health
care setting for other reasons.
It is not known whether administering dexamethasone or other immunomodulatory therapies to patients
with severe COVID-19 will affect the immune response to an influenza vaccine. Nevertheless, as long as
influenza viruses are circulating, people with COVID-19 should receive an influenza vaccine once they
have substantially improved or recovered from COVID-19. See the influenza vaccine recommendations
from the CDC, the ACIP, and the American Academy of Pediatrics.
References
1. Merced-Morales A, Daly P, Abd Elal AI, et al. Influenza activity and composition of the 2022–23 influenza
vaccine—United States, 2021–22 season. MMWR Morb Mortal Wkly Rep. 2022;71(29):913-919. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35862284.
2. Australian Government: Department of Health and Aged Care. Australian influenza surveillance
report. 2022. Available at: https://www1.health.gov.au/internet/main/publishing.nsf/
Content/666B3904B28E059BCA257D1700135B16/$File/Australian%20Influenza%20Surveillance%20
Report%20No.%2011%20-%2015%20August%20to%2028%20August%202022.pdf.
3. World Health Organization. Influenza update N° 427. 2022. Available at: https://cdn.who.int/media/docs/
default-source/influenza/influenza-updates/2022/2022_09_05_surveillance_update_427.pdf.
4. Grohskopf LA, Alyanak E, Ferdinands JM, et al. Prevention and control of seasonal influenza with vaccines:
recommendations of the Advisory Committee on Immunization Practices, United States, 2021–22 influenza
season. MMWR Recomm Rep. 2021;70(5):1-28. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34448800.
5. Centers for Disease Control and Prevention. Contraindications and precautions: general best practice
guidelines for immunization: best practices guidance of the Advisory Committee on Immunization Practices
(ACIP). 2022. Available at: https://www.cdc.gov/vaccines/hcp/acip-recs/general-recs/contraindications.html.
Accessed September 26, 2022.
6. 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.
7. 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.
COVID-19 Treatment Guidelines 448
Introduction
Approximately 1.2 million people in the United States are living with HIV. Most of these individuals
are in care, and many are on antiretroviral therapy (ART) and have well-controlled disease.1 Similar
to COVID-19, HIV disproportionately affects racial and ethnic minorities and people of lower
socioeconomic status in the United States;2 these demographic groups also appear to have a higher risk
of poor outcomes with COVID-19. In the general population, the individuals who are at the highest
risk of severe COVID-19 include those aged >60 years; those who are pregnant; those who have
received solid organ transplants; and those with comorbidities, such as cancer, obesity, diabetes mellitus,
cardiovascular disease, pulmonary disease, a history of smoking, chronic kidney disease, or chronic liver
disease.3 Many people with HIV have 1 or more comorbidities that may put them at increased risk for a
more severe course of COVID-19.
Information on SARS-CoV-2/HIV coinfection is evolving rapidly. The sections below outline the current
state of knowledge regarding preventing and diagnosing SARS-CoV-2 infection in people with HIV,
the treatment and clinical outcomes in people with HIV who develop COVID-19, and the management
of HIV during the COVID-19 pandemic. In addition to these Guidelines, the Department of Health
and Human Services Panel on Antiretroviral Guidelines for Adults and Adolescents has developed the
Guidance for COVID-19 and People With HIV.
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://www.ncbi.nlm.nih.gov/pubmed/31805031.
2. Meyerowitz EA, Kim AY, Ard KL, et al. Disproportionate burden of coronavirus disease 2019 among racial
minorities and those in congregate settings among a large cohort of people with HIV. AIDS.
2020;34(12):1781-1787. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32604138.
3. Centers for Disease Control and Prevention. COVID-19 information for specific groups of people. 2021.
Available at: https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/index.html. Accessed January 24,
2022.
4. Gervasoni C, Meraviglia P, Riva A, et al. Clinical features and outcomes of patients with human
immunodeficiency virus with COVID-19. Clin Infect Dis. 2020;71(16):2276-2278. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32407467.
5. Harter G, Spinner CD, Roider J, et al. COVID-19 in people living with human immunodeficiency virus: a case
series of 33 patients. Infection. 2020;48(5):681-686. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32394344.
6. Karmen-Tuohy S, Carlucci PM, Zervou FN, et al. Outcomes among HIV-positive patients hospitalized with
COVID-19. J Acquir Immune Defic Syndr. 2020;85(1):6-10. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32568770.
7. Patel VV, Felsen UR, Fisher M, et al. Clinical outcomes and inflammatory markers by HIV serostatus and viral
suppression in a large cohort of patients hospitalized with COVID-19. J Acquir Immune Defic Syndr.
2021;86(2):224-230. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33433966.
8. Shalev N, Scherer M, LaSota ED, et al. Clinical characteristics and outcomes in people living with human
immunodeficiency virus hospitalized for coronavirus disease 2019. Clin Infect Dis. 2020;71(16):2294-2297.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32472138.
9. Sigel K, Swartz T, Golden E, et al. Coronavirus 2019 and people living with human immunodeficiency virus:
outcomes for hospitalized patients in New York City. Clin Infect Dis. 2020;71(11):2933-2938. Available at: https://
www.ncbi.nlm.nih.gov/pubmed/32594164.
10. Stoeckle K, Johnston CD, Jannat-Khah DP, et al. COVID-19 in hospitalized adults with HIV. Open Forum
Infect Dis. 2020;7(8):ofaa327. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32864388.
11. Vizcarra P, Perez-Elias MJ, Quereda C, et al. Description of COVID-19 in HIV-infected individuals: a single-
centre, prospective cohort. Lancet HIV. 2020;7(8):e554-e564. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32473657.
12. Western Cape Department of Health in collaboration with the National Institute for Communicable Diseases,
South Africa. Risk factors for coronavirus disease 2019 (COVID-19) death in a population cohort study from the
Western Cape Province, South Africa. Clin Infect Dis. 2021;73(7):e2005-e2015. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32860699.
13. Bhaskaran K, Rentsch CT, MacKenna B, et al. HIV infection and COVID-19 death: a population-based cohort
COVID-19 Treatment Guidelines 456
Downloaded from https://www.covid19treatmentguidelines.nih.gov/ on 10/26/2022
analysis of UK primary care data and linked national death registrations within the OpenSAFELY platform.
Lancet HIV. 2021;8(1):e24-e32. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33316211.
14. Geretti AM, Stockdale AJ, Kelly SH, et al. Outcomes of coronavirus disease 2019 (COVID-19) related
hospitalization among people with human immunodeficiency virus (HIV) in the ISARIC World Health
Organization (WHO) clinical characterization protocol (UK): a prospective observational study. Clin Infect
Dis. 2021;73(7):e2095-e2106. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33095853.
15. Dandachi D, Geiger G, Montgomery MW, et al. Characteristics, comorbidities, and outcomes in a multicenter
registry of patients with human immunodeficiency virus and coronavirus disease 2019. Clin Infect Dis.
2021;73(7):e1964-e1972. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32905581.
16. Hoffmann C, Casado JL, Harter G, et al. Immune deficiency is a risk factor for severe COVID-19 in
people living with HIV. HIV Med. 2021;22(5):372-378. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/33368966.
17. Tesoriero JM, Swain CE, Pierce JL, et al. COVID-19 outcomes among persons living with or without
diagnosed HIV infection in New York State. JAMA Netw Open. 2021;4(2):e2037069. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33533933.
18. Sun J, Patel RC, Zheng Q, et al. COVID-19 disease severity among people with HIV infection or solid organ
transplant in the United States: a nationally-representative, multicenter, observational cohort study. medRxiv.
2021;Preprint. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34341798.
19. Bertagnolio S, Thwin SS, Silva R, et al. Clinical characteristics and prognostic factors in people living
with HIV hospitalized with COVID-19: findings from the WHO Global Clinical Platform. Presented
at: International AIDS Society. 2021. Virtual. Available at: https://theprogramme.ias2021.org/Abstract/
Abstract/2498.
20. 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.
21. 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.
22. 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.
23. Levy I, Wieder-Finesod A, Litchevsky V, et al. Immunogenicity and safety of the BNT162b2 mRNA
COVID-19 vaccine in people living with HIV-1. Clin Microbiol Infect. 2021;27(12):1851-1855. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34438069.
24. Woldemeskel BA, Karaba AH, Garliss CC, et al. The BNT162b2 mRNA vaccine elicits robust humoral and
cellular immune responses in people living with HIV. Clin Infect Dis. 2021;Published online ahead of print.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34293114.
25. Frater J, Ewer KJ, Ogbe A, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 (AZD1222) vaccine
against SARS-CoV-2 in HIV infection: a single-arm substudy of a Phase 2/3 clinical trial. Lancet HIV.
2021;8(8):e474-e485. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34153264.
26. Tan SS, Chew KL, Saw S, Jureen R, Sethi S. Cross-reactivity of SARS-CoV-2 with HIV chemiluminescent
assay leading to false-positive results. J Clin Pathol. 2021;74(9):614. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32907911.
27. Blanco JL, Ambrosioni J, Garcia F, et al. COVID-19 in patients with HIV: clinical case series. Lancet HIV.
2020;7(5):e314-e316. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32304642.
28. Coleman H, Snell LB, Simons R, Douthwaite ST, Lee MJ. Coronavirus disease 2019 and Pneumocystis
jirovecii pneumonia: a diagnostic dilemma in HIV. AIDS. 2020;34(8):1258-1260. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32501852.
29. Centers for Disease Control and Prevention. HIV surveillance report: estimated HIV incidence and prevalence
COVID-19 Treatment Guidelines 457