Skip to nav Skip to content

IDSA Guidelines on the Treatment and Management of Patients with COVID-19

Published by IDSA on 4/11/2020. Last updated,

COVID-19 Guideline, Part 2: Infection Prevention

COVID-19 Guideline, Part 3: Molecular Testing

COVID-19 Guideline, Part 4: Serologic Testing

COVID-19 Guideline, Part 5: Antigen Testing

Adarsh Bhimraj*, Rebecca L. Morgan**, Amy Hirsch Shumaker, Valery Lavergne**, Lindsey Baden, Vincent Chi-Chung Cheng, Kathryn M. Edwards, Rajesh Gandhi, Jason Gallagher, William J. Muller, John C. O’Horo, Shmuel Shoham, M. Hassan Murad**, Reem A. Mustafa**, Shahnaz Sultan**, Yngve Falck-Ytter**

*Corresponding Author  **Methodologist

 

February 22, 2022 - SPECIAL UPDATE ALERT

The following section has been updated based on newly available literature and approvals. This update will be fully integrated into the website soon; it is provided here for immediate use.

  • Lopinavir/Ritonavir (PDF): Two new recommendations on the use of lopinavir/ritonavir (prophylaxis for persons exposed to SARS-CoV-2; treatment for ambulatory patients with mild-to-moderate COVID-19) have been developed. This update has been endorsed by the Society for Healthcare Epidemiology of America and the Pediatric Infectious Diseases Society.

Update History

February 16, 2022

A revised recommendation was released on the use of remdesivir in patients (ambulatory or hospitalized) with mild-to-moderate COVID at high risk for progression to severe disease. This update has been endorsed by the Society for Healthcare Epidemiology of America and the Pediatric Infectious Diseases Society. This revised recommendation has been provided for immediate use and will be integrated into the website at a later date.

February 8, 2022

A new recommendation was released on the use of convalescent plasma in ambulatory patients with mild-to-moderate COVID-19 in ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease with no other treatment options. This recommendation has been provided for immediate use and will be integrated into the website at a later date.

February 3, 2022

Version 6.0.2 as been released and includes an update to the evidence profile for nirmatrelvir/ritonavir in ambulatory patients (outcome of COVID-19 related hospitalizations).

January 18, 2022

Version 6.0.1 as been released and includes endorsement from the Society for Infectious Diseases Pharmacists.

January 12, 2022

Version 6.0.0 as been released and includes new recommendations on the use of remdesivir for ambulatory patientstixagevimab/cilgavimab for pre-exposure prophylaxisnirmatrelvir/ritonavir in ambulatory patients, and molnupiravir for ambulatory patients. This update has been endorsed by the Society for Healthcare Epidemiology of America and the Pediatric Infectious Diseases Society.

December 30, 2021

A new recommendation was released on the use of molnupiravir for ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease who have no other treatment options. This recommendation was originally provided for immediate use and was later integrated into the website on January 12, 2022 as part of Version 6.0.0.

December 24, 2021

The following recommendation sections were added based on newly available literature and/or approvals. They were provided here for immediate use and were later integrated into the website on January 12, 2022 as part of Version 6.0.0.

  • Remdesivir: New recommendation on the use of remdesivir for ambulatory patients
  • Neutralizing Antibodies for Prophylaxis: New recommendation on the use of tixagevimab/cilgavimab for pre-exposure prophylaxis in adults at increased risk for inadequate immune response to COVID-19 vaccine or for whom COVID-19 vaccine is not recommended
  • Oral Antivirals (NEW): New recommendation on the use of nirmatrelvir/ritonavir in ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease
    • UPDATE (1/4/2022): The language in the above section has been updated, with "nirmatrelvir/ritonavir" replacing "oral antivirals". Additionally, information on the use of brand name as well as use in patients hospitalized for reasons other than COVID-19 has been added.
November 18, 2021

Version 5.6.0 as been released and includes revised recommendations on the use of convalescent plasma in hospitalized and ambulatory patients with COVID-19; this update has been endorsed by the Society for Healthcare Epidemiology of America, the Pediatric Infectious Diseases Society, and the Society of Infectious Diseases Pharmacists.

November 9, 2021

Version 5.5.3 has been released and contains a correction to the analysis for the outcome of mortality in ambulatory patients treated with fluvoxamine.

November 5, 2021

Version 5.5.2 has been released and includes updated literature for the use of fluvoxamine in ambulatory patients.

November 1, 2021

Version 5.5.1 has been released and includes endorsement from the Society of Infectious Diseases Pharmacists.

October 27, 2021

Version 5.5.0 has been released and contains a new recommendation on the use of fluvoxamine in ambulatory patients. This update has been endorsed by the Society for Healthcare Epidemiology of America and the Pediatric Infectious Diseases Society.

October 18, 2021

Version 5.4.1 has been released and contains a minor correction to the neutralizing antibodies section. Subcutaneous has been removed to the dosing for bamlanivimab/etesevimab.

October 15, 2021

Version 5.4.0 has been released and includes revised remarks and a new evidence profile for the use of baricitinib for critically ill patients requiring invasive mechanical ventilation. This update has been endorsed by the Society of Infectious Diseases Pharmacists.

October 1, 2021

Version 5.3.1 has been released and contains a correction to the certainty of evidence for the new recommendation on the use of hydroxychloroquine as post-exposure prophylaxis. This recommendation has a moderate--not low--certainty of evidence.

September 30, 2021

Version 5.3.0 has been released and includes a new recommendation on the use of hydroxychloroquine as post-exposure prophylaxis. This update has been endorsed by the Society for Healthcare Epidemiology of America and the Society of Infectious Diseases Pharmacists.

September 24, 2021

Version 5.2.1 has been released and features existing recommendations on the use of neutralizing SARS-CoV-2 antibodies separated by prophylaxis vs. treatment.

September 21, 2021

Version 5.2.0 has been released and includes a new recommendation on the use of sarilumab. This update has been endorsed by the Society for Healthcare Epidemiology of America, the Pediatric Infectious Diseases Society, and the Society of Infectious Diseases Pharmacists.

September 14, 2021

Version 5.1.2 has been released and includes endorsement from the Society of Infectious Diseases Pharmacists.

September 3, 2021

Version 5.1.1 has been released and includes endorsement from the Pediatric Infectious Diseases Society.

August 27, 2021

Version 5.1.0 has been released and includes revised remarks for the use of baricitinib and new recommendations on the use of tofacitinib.

August 25, 2021

Version 5.0.0 has been released and includes new and updated recommendations for neutralizing antibodies and ivermectin. This update has been endorsed by the Society for Healthcare Epidemiology of America.

June 25, 2021

Version 4.4.1 has been released and includes endorsement from the Pediatric Infectious Diseases Society.

June 23, 2021

Version 4.4.0 has been released and includes updated recommendations and literature summary on neutralizing antibodies.

June 3, 2021

Version 4.3.0 has been released and contains a new recommendation on the use of remdesivir.

May 3, 2021

Version 4.2.1 has been released and contains endorsement from the Pediatric Infectious Diseases Society.

April 14, 2021
Version 4.2.0 has been released and contains updated recommendations on the use of COVID-19 convalescent plasma, neutralizing antibodies and remarks on remdesivir.
 
April 5, 2021
Version 4.1.2 has been released and contains a revision to the number of studies included in Table 9 in the section on remdesivir.
 
March 18, 2021

Version 4.1.1 has been released and contains a revision to the number of studies found for ivermectin.

March 5, 2021

Version 4.1.0 has been released and contains a new recommendation on the use of bamlanivimab with etesevimab among ambulatory patients.

February 22, 2021

Version 4.0.0 has been released and contains a revised recommendation on the use of tocilizumab. 

February 18, 2021

Version 3.10.0 has been released and includes additional information on study eligibility for ivermectin.

February 10, 2021

Version 3.9.0 has been released and contains an updated literature review for tocilizumab.

February 5, 2021

Version 3.8.0 has been released and includes two new recommendations on the use of ivermectin.

February 3, 2021

Version 3.7.0 has been released and includes two new recommendations on the use of neutralizing monoclonal antibodies.

January 8, 2021

Version 3.6.0 has been realeased and includes new recommendations on the use of baricitinib and an updated literature review on hydroxychloroquine.

December 2, 2020

 Version 3.5.1 has been released and includes endorsement from the Pediatric Infectious Diseases Society.

November 22, 2020

Version 3.5.0 has been released and includes revisions to the sections on lopinavir/ritonavir, tocilizumab, and remdesivir.

November 18, 2020

Version 3.4.0 has been released and contains a new recommendation on the use of bamlanivimab.

September 25, 2020

Version 3.3.0 has been released and contains revised and new recommendations for the use of dexamethasone and a revised recommendation against the routine use of tocilizumab.

September 21, 2020

Version 3.2.1 has been released and includes endorsement from the Pediatric Infectious Diseases Society.

September 15, 2020

Version 3.2.0 has been released and contains a new recommendation on the use of remdesivir in patients with more moderate disease.

September 4, 2020

Version 3.1.0 has been released and contains additional information on convalescent plasma as well new and updated narrative summaries of treatments undergoing evaluation.

August 28, 2020

Version 3.0.1 has been released and includes endorsement from the Pediatric Infectious Diseases Society.

August 20, 2020

Version 3.0.0 of the guideline has been released and contains revised recommendations on hydroxychloroquine and hydroxychloroquine plus azithromycin.

June 25, 2020

Version 2.1.0 of the guideline has been released and includes revised recommendations on corticosteroids.

June 22, 2020

Version 2.0.0 of the guideline has been released and contains:

  • Revised recommendations on hydroxychloroquine and hydroxychloroquine plus azithromycin
  • Revised recommendations for convalescent plasma for treatment of COVID-19
  • New recommendations on the use of remdesivir
  • New recommendations for famotidine (not addressed in versions 1.0.0-1.0.4)
April 21, 2020

Version 1.0.4 of the guideline has been released.

Overview of COVID-19 Treatment Guidelines (Summary Table)

Overview of COVID-19 Interventions v6.0.png

 

Strengths of recommendation

Recommend (strong recommendation): Guideline panel is confident that the desirable effects of an intervention outweigh the undesirable effects. Most or all individuals will be best served by the recommended course of action.

Suggest (weak or conditional recommendation): Guideline panel after discussion concludes that the desirable effects probably outweigh undesirable effects, but appreciable uncertainty exists. Not all individuals will be best served by the recommended course of action and the caregiver needs to consider more carefully than usual the individual patient’s circumstances, preferences, and values.

Certainty of evidence

⨁⨁⨁⨁       high

⨁⨁⨁◯       moderate

⨁⨁◯◯        low

⨁◯◯◯        very low

See Figure 1 in the Executive Summary.

Abstract

Background: There are many pharmacologic therapies that are being used or considered for treatment of coronavirus disease 2019 (COVID-19). There is a need for frequently updated practice guidelines on their use, based on critical evaluation of rapidly emerging literature. 

Objective: There are many pharmacologic therapies that are being used or considered for treatment of coronavirus disease 2019 (COVID-19). There is a need for frequently updated practice guidelines on their use, based on critical evaluation of rapidly emerging literature.

Methods: In March 2020, the Infectious Diseases Society of America (IDSA) formed a multidisciplinary guideline panel of infectious disease clinicians, pharmacists, and methodologists with varied areas of expertise. The process followed a rapid recommendation checklist. The panel prioritized questions and outcomes. Then a systematic review of the peer-reviewed and grey literature was conducted. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach was used to assess the certainty of evidence and make recommendations.

Results: On April 11, 2020, IDSA released online initial treatment recommendations and narrative summaries of other treatments under evaluation. Since that time, the guideline panel and methodologists have continued to monitor the literature and issue updates and addendums to these guidelines in response to evolving research.

Conclusions: Since the inception of its work, the panel has expressed the overarching goal that patients be recruited into ongoing trials, which would provide much needed evidence on the efficacy and safety of various therapies for COVID-19, given that we could not make a determination whether the benefits outweigh harms for most treatments.

Keywords: coronavirus, SARS-CoV-2, COVID, COVID-19, pneumonia

Executive Summary and Background

Executive Summary

Coronavirus disease 2019 (COVID-19) is a pandemic with a rapidly increasing incidence of infections and deaths. Many pharmacologic therapies are being used or considered for treatment. Given the rapidity of emerging literature, the Infectious Diseases Society of America (IDSA) identified the need to develop living, frequently updated evidence-based guidelines to support patients, clinicians and other health-care professionals in their decisions about treatment and management of patients with COVID-19.

Summarized below are the recommendations with comments related to the clinical practice guideline for the treatment and management of COVID-19. A detailed description of background, methods, evidence summary and rationale that support each recommendation, and research needs can be found online in the full text. In brief, per Grading of Recommendations Assessment, Development and Evaluation (GRADE) methodology, recommendations are labeled as “strong” or “conditional”. The word “recommend” indicates strong recommendations and “suggest” indicates conditional recommendations. In situations where promising interventions were judged to have insufficient evidence of benefit to support their use and with potential appreciable harms or costs, the expert panel recommended their use in the context of a clinical trial. These recommendations acknowledge the current “knowledge gap” and aim at avoiding premature favorable recommendations for potentially ineffective or harmful interventions.

 

Hydroxychloroquine and Hydroxychloroquine+Azithromycin for Treatment

  • Recommendation 1: Among patients with COVID-19, the IDSA guideline panel recommends against hydroxychloroquine. (Strong recommendation, Moderate certainty of evidence)
    • Remark: Chloroquine is considered to be class equivalent to hydroxychloroquine.
  • Recommendation 2: Among hospitalized patients with COVID-19, the IDSA guideline panel recommends against hydroxychloroquine plus azithromycin. (Strong recommendation, Low certainty of evidence)
    • Remark Chloroquine is considered to be class equivalent to hydroxychloroquine


Hydroxychloroquine for Prophylaxis

  • Recommendation 3: In persons exposed to COVID-19, the IDSA guideline panel recommends against hydroxychloroquine. (Strong recommendation, Low certainty of evidence)


Lopinavir/Ritonavir

  • Recommendation 4: Among hospitalized patients with COVID-19, the IDSA guideline panel recommends against the use of the combination lopinavir/ritonavir. (Strong recommendation, Moderate certainty of evidence)
  • SPECIAL UPDATE ALERT (2/22/2022): This section has been updated based on newly available literature and approvals. This update will be fully integrated into the website soon; it is provided here for immediate use. Two new recommendations on the use of lopinavir/ritonavir in persons exposed to or with COVID-19 have been developed. The full updated section can be viewed here (PDF).


Glucocorticoids

  • Recommendation 5: Among hospitalized critically ill* patients with COVID-19, the IDSA guideline panel recommends dexamethasone rather than no dexamethasone. (Strong recommendation, Moderate certainty of evidence)
    • Remark: If dexamethasone is unavailable, equivalent total daily doses of alternative glucocorticoids may be used. Dexamethasone 6 mg IV or PO for 10 days (or until discharge) or equivalent glucocorticoid dose may be substituted if dexamethasone unavailable. Equivalent total daily doses of alternative glucocorticoids to dexamethasone 6 mg daily are methylprednisolone 32 mg and prednisone 40 mg.
  • Recommendation 6: Among hospitalized patients with severe** but not critical COVID-19, the IDSA guideline panel suggests dexamethasone rather than no dexamethasone. (Conditional recommendation, Moderate certainty of evidence)
    • Remark: Dexamethasone 6 mg IV or PO for 10 days (or until discharge) or equivalent glucocorticoid dose may be substituted if dexamethasone unavailable. Equivalent total daily doses of alternative glucocorticoids to dexamethasone 6 mg daily are methylprednisolone 32 mg and prednisone 40 mg.
  • Recommendation 7: Among hospitalized patients with non-severe*** COVID-19 without hypoxemia requiring supplemental oxygen, the IDSA guideline panel suggests against the use of glucocorticoids. (Conditional recommendation, Low certainty of evidence)
  • *Critical illness is defined as patients on mechanical ventilation and ECMO. Critical illness includes end organ dysfunction as is seen in sepsis/septic shock. In COVID-19, the most commonly reported form of end organ dysfunction is ARDS.
  • **Severe illness is defined as patients with SpO2 ≤94% on room air, including patients on supplemental oxygen.
  • ***Non-severe illness is defined as patient with a SpO2 > 94% not requiring supplemental oxygen.


Interleukin-6 Inhibitors

  • Recommendation 8: Among hospitalized adults with progressive severe* or critical** COVID-19 who have elevated markers of systemic inflammation, the IDSA guideline panel suggests tocilizumab in addition to standard of care (i.e., steroids) rather than standard of care alone. (Conditional recommendation, Low certainty of evidence)
    • Remarks:
      • Patients, particularly those who respond to steroids alone, who put a high value on avoiding possible adverse events of tocilizumab and a low value on the uncertain mortality reduction, would reasonably decline tocilizumab.
      • In the largest trial on the treatment of tocilizumab, criterion for systemic inflammation was defined as CRP ≥75 mg/L.
  • Recommendation 9: When tocilizumab is not available, for patients who would otherwise qualify for tocilizumab, the IDSA guideline panel suggests sarilumab in addition to standard of care (i.e., steroids) rather than standard of care alone. (Conditional recommendation, Very low certainty of evidence)
    • Remark: Patients, particularly those who respond to steroids alone, who put a high value on avoiding possible adverse events of sarilumab and a low value on the uncertain mortality reduction, would reasonably decline sarilumab.
  • *Severe illness is defined as patients with SpO2 ≤94% on room air, including patients on supplemental oxygen.
  • **Critical illness is defined as patients on mechanical ventilation and ECMO. Critical illness includes end organ dysfunction as is seen in sepsis/septic shock. In COVID-19, the most commonly reported form of end organ dysfunction is ARDS.


Convalescent Plasma

  • Recommendation 10: Among patients hospitalized with COVID-19, the IDSA guideline panel recommends against COVID-19 convalescent plasma. (Strong recommendation, Moderate certainty of evidence)
  • Recommendation 11: Among ambulatory patients with mild to moderate COVID-19, the IDSA guideline panel suggests against COVID-19 convalescent plasma outside of the context of a clinical trial. (Conditional recommendation, Low certainty of evidence)
    • SPECIAL UPDATE ALERT (2/8/2022): This recommendation has been updated based on newly available literature and approvals. This update will be fully integrated into the website soon; it is provided here for immediate use. Recommendation 11 has been revised to address the use of convalescent plasma in ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease with no other treatment options. The full updated section can be viewed here (PDF).


Remdesivir

  • Recommendation 12 (NEW): Among ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease, the IDSA guideline panel suggests remdesivir initiated within seven days of symptom onset rather than no remdesivir. (Conditional recommendation, Low certainty of evidence)
    • Remarks:
      • Dosing for remdesivir is 200 mg on day one followed by 100 mg on days two and three.
      • Patients with mild to moderate COVID-19 who are at high risk of progression to severe disease admitted to the hospital for reasons other than COVID-19 may also receive remdesivir
      • Options for treatment and management of ambulatory patients include nirmatrelvir/ritonavir, three-day treatment with remdesivir, molnupiravir, and neutralizing monoclonal antibodies. Patient specific factors (e.g., symptom duration, renal function, drug interactions), product availability, and institutional capacity and infrastructure should drive decision-making regarding choice of agent. Data for combination treatment do not exist in this setting.
    • SPECIAL UPDATE ALERT: This recommendation has been updated based on newly available literature and approvals. This update will be fully integrated into this document and the IDSA website soon; it is provided here for immediate use. Recommendation 12 has been revised to address the use of remdesivir in patients (ambulatory or hospitalized) with mild-to-moderate COVID-19 at high risk for progression to severe disease. The full updated section can be viewed on the here (PDF).

  • Recommendation 13a: In hospitalized patients with severe* COVID-19, the IDSA panel suggests remdesivir over no antiviral treatment. (Conditional recommendation, Moderate certainty of evidence)
    • Severity definition: *Severe illness is defined as patients with SpO2≤94% on room air.
  • Recommendation 13b: In patients with COVID-19 on invasive ventilation and/or ECMO, the IDSA panel suggests against the routine initiation of remdesivir (Conditional recommendation, Very low certainty of evidence)
  • Recommendation 14: In patients on supplemental oxygen but not on mechanical ventilation or ECMO, the IDSA panel suggests treatment with five days of remdesivir rather than 10 days of remdesivir. (Conditional recommendation, Low certainty of evidence)
  • Recommendation 15: In patients with COVID-19 admitted to the hospital without the need for supplemental oxygen and oxygen saturation >94% on room air, the IDSA panel suggests against the routine use of remdesivir. (Conditional recommendation, Very low certainty of evidence)


Famotidine

  • Recommendation 16: Among hospitalized patients with severe COVID-19, the IDSA panel suggests against famotidine use for the sole purpose of treating COVID-19 outside of the context of a clinical trial. (Conditional recommendation, Very low certainty of evidence)


Neutralizing Antibodies for Pre-Exposure and Post-Exposure Prophylaxis

  • Recommendation 17 (NEW): In moderately or severely immunocompromised individuals* at increased risk for inadequate immune response to COVID-19 vaccine or for whom COVID-19 vaccine is not recommended due to a documented serious adverse reaction to the vaccine, the IDSA guideline panel suggests pre-exposure prophylaxis with tixagevimab/cilgavimab rather than no tixagevimab/cilgavimab. (Conditional recommendation, Low certainty of evidence)
    • Remarks:
      • Dosing for tixagevimab/cilgavimab is 150 mg of tixagevimab & 150 mg of cilgavimab administered as two separate consecutive intramuscular injections once.
      • Local SARS-CoV-2 variant susceptibility should be considered.
  • Recommendation 18: In persons exposed to COVID-19 who are at high risk of progression to severe COVID-19, the IDSA guideline panel suggests post-exposure casirivimab/imdevimab rather than no casirivimab/imdevimab. (Conditional recommendation, low certainty of evidence)Remarks:
      • Dosing for casirivimab/imdevimab is casirivimab 600 mg & imdevimab 600 mg IV or SC once.
      • In the trial considered for this recommendation, participants were enrolled within 96 hours after a household contact received a diagnosis of SARS-CoV-2 infection.


Neutralizing Antibodies for Treatment

  • Recommendation 19: Among ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease, the IDSA guideline panel suggests bamlanivimab/etesevimab, casirivimab/imdevimab, or sotrovimab rather than no neutralizing antibody treatment. (Conditional recommendation, Moderate certainty of evidence)
    • Remarks:
      • Dosing for casirivimab/imdevimab is casirivimab 600 mg and imdevimab 600 mg IV. Subcutaneous injection is a reasonable alternative in patients for whom it cannot be given intravenously.
      • Dosing for sotrovimab is sotrovimab 500 IV once.
      • Dosing for bamlanivimab/etesevimab is bamlanivimab 700 mg and etesevimab 1400 mg IV once.
      • Patients with mild to moderate COVID-19 who are at high risk of progression to severe disease admitted to the hospital for reasons other than COVID-19 may also receive bamlanivimab/etesevimab, casirivimab/imdevimab, or sotrovimab.
      • Local variant susceptibility should be considered in the choice of the most appropriate neutralizing antibody therapy. Local availability of different monoclonal antibody combinations may be affected by predominance of local variants.
      • There are limited data on efficacy of bamlanivimab/etesevimab, casirivimab/imdevimab, or sotrovimab in high-risk patients under 18 years of age.
  • Recommendation 20: Among hospitalized patients with severe COVID-19, the IDSA guideline panel recommends against bamlanivimab monotherapy. (Strong recommendation, Moderate certainty of evidence)


JAK Inhibitors (Baricitinib, Tofacitinib)

  • Recommendation 21: Among hospitalized adults with severe* COVID-19 having elevated inflammatory markers, the IDSA panel suggests baricitinib rather than no baricitinib. (Conditional recommendation, Moderate certainty of evidence)
    • Remarks:
      • Baricitinib 4 mg per day (or appropriate renal dosing) up to 14 days or until discharge from hospital.
      • Baricitinib appears to demonstrate the most benefit in those with severe COVID-19 on high-flow oxygen/non-invasive ventilation at baseline.
      • Limited additional data suggest a mortality reduction even among patients requiring mechanical ventilation.
      • Patients who receive baricitinib for treatment of COVID-19 should not receive tocilizumab or other IL-6 inhibitors.
  • Recommendation 22: Among hospitalized patients with severe* COVID-19 who cannot receive a corticosteroid (which is standard of care) because of a contraindication, the IDSA guideline panel suggests use of baricitinib with remdesivir rather than remdesivir alone. (Conditional recommendation, Low certainty of evidence)
    • Remark: Baricitinib 4 mg daily dose for 14 days or until hospital discharge. The benefits of baricitinib plus remdesivir for persons on mechanical ventilation are uncertain.
  • Recommendation 23: Among hospitalized adults with severe** COVID-19, but not on non-invasive or invasive mechanical ventilation, the IDSA panel suggests tofacitinib rather than no tofacitinib. (Conditional recommendation, Low certainty of evidence)
    • Remarks:
      • Tofacitinib appears to demonstrate the most benefit in those with severe COVID-19 on supplemental or high-flow oxygen.
      • Patients treated with tofacitinib should be on at least prophylactic dose anticoagulant.
      • Patients who receive tofacitinib should not receive tocilizumab or other IL-6 inhibitor for treatment of COVID-19.
      • The STOP-COVID Trial did not include immunocompromised patients.
  • *Severe illness is defined as patients with SpO2 ≤94% on room air, including patients on supplemental oxygen, oxygen through a high-flow device, or non-invasive ventilation.
  • **Severe illness is defined as patients with SpO22 ≤94% on room air, including patients on supplemental oxygen or oxygen through a high-flow device.


Ivermectin

  • Recommendation 24: In hospitalized patients with COVID-19, the IDSA panel suggests against ivermectin outside of the context of a clinical trial. (Conditional recommendation, very low certainty of evidence)
  • Recommendation 25: In ambulatory persons with COVID-19, the IDSA panel suggests against ivermectin outside of the context of a clinical trial. (Conditional recommendation, very low certainty of evidence)


Fluvoxamine

  • Recommendation 26: Among ambulatory patients with COVID-19, the IDSA guideline panel recommends fluvoxamine only in the context of a clinical trial. (Knowledge gap)


Nirmatrelvir/Ritonavir

  • Recommendation 27 (NEW): In ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease, the IDSA guideline panel suggests nirmatrelvir/ritonavir initiated within five days of symptom onset rather than no nirmatrelvir/ritonavir. (Conditional recommendation, Low certainty of evidence)
    • Patients' medications need to be screened for serious drug interactions (i.e., medication reconciliation). Patients on ritonavir- or cobicistat-containing HIV or HCV regimens should continue their treatment as indicated.
    • Dosing based on renal function:
      • Estimated glomerular filtration rate (eGFR) > 60 ml/min: 300 mg nirmatrelvir/100 ritonavir every 12 hours for five days
      • eGFR ≤60 and ≥30 mL/min: 150 mg nirmatrelvir/100 mg ritonavir every 12 hours for five days
      • eGFR <30 mL/min: not recommended
    • Patients with mild to moderate COVID-19 who are at high risk of progression to severe disease admitted to the hospital for reasons other than COVID-19 may also receive nirmatrelvir/ritonavir
    • Options for treatment and management of ambulatory patients include nirmatrelvir/ritonavir, three-day treatment with remdesivir, molnupiravir, and neutralizing monoclonal antibodies. Patient-specific factors (e.g., symptom duration, renal function, drug interactions) as well as product availability should drive decision-making regarding choice of agent. Data for combination treatment do not exist in this setting.
    • Remarks:


Molnupiravir

  • Recommendation 28 (NEW): In ambulatory patients (≥18 years) with mild to moderate COVID-19 at high risk for progression to severe disease who have no other treatment options*, the IDSA guideline panel suggests molnupiravir initiated within five days of symptom onset rather than no molnupiravir. (Conditional recommendation, Low certainty of evidence)
    • Remarks:
      • Patients who put a higher value on the putative mutagenesis, adverse events or reproductive concerns, and a lower value on the uncertain benefits, would reasonably decline molnupiravir.
      • Molnupiravir 800 mg for five days.
      • Patients with mild to moderate COVID-19 who are at high risk of progression to severe disease admitted to the hospital for reasons other than COVID-19 may also receive molnupiravir.
      • Molnupiravir is not authorized under the FDA EUA for use in patients <18 years, because it may affect bone and cartilage growth.
      • Molnupiravir is not recommended under the FDA EUA for use during pregnancy.
      • Molnupiravir is not authorized under the FDA EUA for pre-exposure or post-exposure prevention of COVID-19 or for initiation of treatment in patients hospitalized due to COVID-19, because benefit of treatment has not been observed in individuals when treatment is started after hospitalization due to COVID-19.
    • *Other options for treatment and management of ambulatory patients include nirmatrelvir/ritonavir, three-day treatment with remdesivir, and neutralizing monoclonal antibodies. Patient-specific factors (e.g., symptom duration, renal function, drug interactions) as well as product availability should drive decision-making regarding choice of agent. Data for combination treatment do not exist in this setting.

 

Since the inception of its work, the panel has expressed the overarching goal that patients be recruited into ongoing trials, which would provide much needed evidence on the efficacy and safety of various therapies for COVID-19. The panel has determined that when an explicit trade-off between highly uncertain benefits and known putative harms of these therapeutic agents were considered, a net positive benefit was not reached and could possibly be negative (risk of excess harm). The panel acknowledges that enrolling patients in randomized controlled trials (RCTs) might not be feasible for many frontline providers due to limited access and infrastructure. Should lack of access to clinical trials exist, we encourage setting up local or collaborative registries to systematically evaluate the efficacy and safety of drugs to contribute to the knowledge base. Each clinician can play a role in advancing our understanding of this disease through a local registry or other data collection efforts.

Background

The first cases of COVID-19 were reported from Wuhan, China in early December 2019 [1], now known to be caused by a novel beta-coronavirus, named as Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Within a span of months, COVID-19 has become pandemic due to its transmissibility, spreading across continents with the number of cases and deaths rising daily [2]. Although most infected individuals exhibit a mild illness (80%+), 14% have serious and 5% have critical illness. Approximately 10% will require hospital admission due to COVID-19 pneumonia, of which approximately 10% will require ICU care, including invasive ventilation due to acute respiratory distress syndrome (ARDS) [3]. While mortality appears to be more common in older individuals and those with comorbidities, such as chronic lung disease, cardiovascular disease, hypertension and diabetes, young people with no comorbidities also appear to be at risk for critical illness including multi-organ failure and death.

There has been an expanding number of studies rapidly published online and in academic journals; however, some of these may be of limited quality and are pre-published without sufficient peer-review. Critical appraisal of the existing studies is needed to determine if the existing evidence is sufficient to support currently proposed management strategies.

Given the rapid global spread of SARS-CoV-2 and the difficulty for the overburdened front-line providers and policymakers to stay up to date on emerging literature, IDSA has recognized the necessity of developing a rapid guideline for the treatment of COVID-19. The guideline panel is using a methodologically rigorous process for evaluating the best available evidence and providing treatment recommendations. Two additional guidelines on diagnostic testing and infection prevention also have been developed. These guidelines will be frequently updated as substantive literature becomes available and are accessible on an easy to navigate web and device interface at http://www.idsociety.org/covid19guidelines.

There continue to be several ongoing trials evaluating therapeutic agents for the treatment of COVID-19. As data becomes available from these trials and if there is a preponderance of evidence to suggest the use of a therapeutic agent even in the context of clinical trials is no longer warranted it will be removed from future updates of the guideline (and the removal will be noted in the updated guidelines). If there is emerging evidence on the efficacy or safety of a therapeutic agent not mentioned in the current version of the guideline it will be included in future updates of the guideline.

These recommendations are intended to inform patients, clinicians, and other health professionals by providing the latest available evidence.

Methods and Search Results

This guideline was developed using the GRADE approach for evidence assessment. In addition, given the need for an urgent response to a major public health crisis, the methodological approach was modified according to the Guidelines International Network/McMaster checklist for the development of rapid recommendations [4].

Panel Composition

The initial guideline panel assembled in March 2020 was composed of nine members including infectious diseases specialists as well as experts in public health as well as other front-line clinicians, specializing in pharmacology, pediatrics, medical microbiology, preventive care, critical care, hepatology, nephrology and gastroenterology. Organizational representatives were included from the Society for Healthcare Epidemiology of America (SHEA), and the Pediatric Infectious Diseases Society (PIDS). In May 2020, an additional panel member was included as a representative from the Society of Infectious Diseases Pharmacists (SIDP). The Evidence Foundation provided technical support and guideline methodologists for the development of this guideline.

Disclosure and Management of Potential Conflicts of Interest

The conflict of interest (COI) review group for this guideline includes two representatives from IDSA who are responsible for reviewing, evaluating and approving all disclosures. All members of the expert panel have complied with the COI process for reviewing and managing conflicts of interest, which requires disclosure of any financial, intellectual, or other interest that might be construed as constituting an actual, potential, or apparent conflict, regardless of relevancy to the guideline topic. The assessment of disclosed relationships for possible COI is based on the relative weight of the financial relationship (i.e., monetary amount) and the relevance of the relationship (i.e., the degree to which an association might reasonably be interpreted by an independent observer as related to the topic or recommendation of consideration). The COI review group has ensured that the majority of the panel and chair is without potential relevant (related to the topic) conflicts for the duration of their term on the panel. The chair and all members of the technical team have been determined to be unconflicted.

Question Generation

Clinical questions included in this guideline were developed into a PICO format (Population, Intervention, Comparison, Outcomes) [5] and prioritized according to available evidence that met the minimum acceptable criteria (i.e., the body of evidence reported on at least a case-series design, case reports were excluded). Panel members prioritized patient-important outcomes such as mortality, development of ARDS (need for non-invasive or invasive ventilation) and clinical improvement (such as disease-oriented outcomes inferred by radiological findings or virologic cure), and severe adverse events (SAE) leading to treatment discontinuation. Serious adverse events are death, life threatening reactions, those that require hospitalization, result in disability or permanent damage or require an intervention to prevent permanent impairment [6]. Additional drug specific harms were evaluated when clinically relevant, including possible drug-drug reactions, if applicable.

Search Strategy

The National Institute for Health and Care Excellence (NICE) highly-sensitive search was reviewed by the methodologist in consultation with the technical team information specialist and was determined to have high sensitivity [7]. An additional term, COVID, was added to the search strategy used in addition to the treatment terms identified in the PICO questions (Supplementary Table s1). Ovid Medline and Embase were searched from 2019 through September 18, 2020. Horizon scans have been performed regularly during the evidence assessment and recommendation process to locate additional grey literature and manuscript pre-prints. Reference lists and literature suggested by panelists were reviewed for inclusion. No restrictions were placed on language or study type.

Screening and Study Selection

Two reviewers independently screened titles and abstracts, as well as eligible full-text studies. When acceptable RCTs of effectiveness were found, no additional non-randomized studies or non-comparative evidence (i.e., single arm case series) were sought. Evidence from single arm studies reporting on non-comparative rates of outcomes of interest were included if a historical control event rate could be estimated from the literature. Reviewers extracted relevant information into a standardized data extraction form.

For several interventions, no direct evidence was available other than case reports or mechanistic considerations. The panel either decided to include plausible indirect evidence and make a recommendation (e.g., from studies of SARS-CoV) or to provide a short narrative discussion of the intervention.

Data Collection and Analysis

Data extracted from the available evidence included: mortality, clinical progression or improvement as reported in the studies, virologic clearance, and adverse events. Where applicable, data were pooled using random effects model (fixed effects model for two or fewer trials or pooling of rates) using RevMan [8].

Risk of Bias and Certainty of Evidence

Risk of bias was assessed using the Cochrane Risk of Bias Tool for RCTs and the Risk of Bias Instrument for Non-randomized Studies – of Interventions (ROBINS-I) [9, 10]. The certainty of evidence was assessed using the GRADE approach [11]. Within GRADE, the body of evidence across each outcome is assessed for domains that may reduce or increase one’s certainty in the evidence. Factors that may reduce one’s certainty include risk of bias (study limitations), inconsistency (unexplained heterogeneity across study findings), indirectness (applicability or generalizability to the research question), imprecision (the confidence in the estimate of an effect to support a particular decision) or publication bias (selective publication of studies). One’s certainty in the evidence may be strengthened if the following considerations are present: large or very large magnitude of effect, evidence of a dose-response gradient, or opposing residual confounding. GRADE summary of findings tables were developed in GRADEpro Guideline Development Tool [12].

As higher quality direct evidence for clinical outcomes becomes available, outcomes previously deemed critical by the panel became less important for decision-making. For example, at the time of the first guideline, clinical improvement outcomes (e.g., need for mechanical ventilation) were not reported, only the results of radiographic findings. However, with the recent publication of RCTs and non-randomized studies reporting on direct measures of clinical improvement, results of radiographic studies were deemed to be less critical for decision making.

Evidence to Recommendations

The panel considered core elements of the GRADE evidence in the decision process, including Certainty of evidence and balance between desirable and undesirable effects. Additional domains were acknowledged where applicable (feasibility, resource use, acceptability). For all recommendations, the expert panelists reached consensus. Voting rules were agreed on prior to the panel meetings for situations when consensus could not be reached.

As per GRADE methodology, recommendations are labeled as “strong” or “conditional”. The words “we recommend” indicate strong recommendations and “we suggest” indicate conditional recommendations. Figure 1 provides the suggested interpretation of strong and weak recommendations for patients, clinicians, and healthcare policymakers. For recommendations where the comparators are not formally stated, the comparison of interest is implicitly referred to as “not using the intervention”. These recommendations acknowledge the current “knowledge gap” and aim at avoiding premature favorable recommendations for their use and to avoid encouraging the rapid diffusion of potentially ineffective or harmful interventions. Detailed suggestions about the specific research questions that should be addressed are found in the table (see Supplementary Table s2).

Figure 01.PNG

 

Review Process

This guideline has been rapidly reviewed and approved by the IDSA Board of Directors Executive Committee external to the guideline development panel. SHEA, PIDS, and SIDP have reviewed and provided endorsement of its contents.

Updating Process

Regular, frequent screening of the literature will take place to determine the need for revisions based on the likelihood that any new data will have an impact on the recommendations. When necessary, the entire expert panel is reconvened to discuss potential changes.

Changes to these guidelines will fall into one of two categories: update or amendment. An update involves a search for new studies, and if any new studies are found, they will be critically appraisal and the pertinent section will be removed and replaced with the updated section. An amendment involves a change or correction to the document, without any search for new studies and their appraisal. It will also involve changes made to clarify or explain a section based on “living” feedback from the readers.

Guideline revisions may result in major, minor, or “patch” version changes, defined as follows:

  • Major version (e.g., 1.0.0): Synonymous with a newly published version in the journal. This is usually called a "breaking version", i.e. prior recommendations may not be valid anymore.
  • Minor version (e.g., 1.1.0): Includes new information, maybe even added PICOs, but not a breaking version, i.e., existing recommendations are still valid, although new recommendations may be available.
  • Patch version (e.g., 1.0.1): Small changes, i.e., typos, adding words, removing words, but there are no material changes to the document or changes in recommendations.

Results

Systematic review and horizon scan of the literature identified 2030 references of which 48 informed the evidence base for these recommendations (Supplementary Figure s1). Characteristics of the included studies can be found in the supplementary materials.

Recommendations 1-2: Hydroxychloroquine and Hydroxychloroquine + azithromycin

Section last reviewed and updated 12/23/2020

Last literature search conducted 12/14/2020

Recommendation 1. Among hospitalized patients with COVID-19, the IDSA guideline panel recommends against hydroxychloroquine*. (Strong recommendation, Moderate certainty of evidence)

  • Remark: Chloroquine is considered to be class equivalent to hydroxychloroquine.

Recommendation 2. Among hospitalized patients with COVID-19, the IDSA guideline panel recommends against hydroxychloroquine* plus azithromycin. (Strong recommendation, Low certainty of evidence)

  • Remark: Chloroquine is considered to be class equivalent to hydroxychloroquine.

 

Why are hydroxychloroquine and hydroxychloroquine plus azithromycin considered for treatment?

Hydroxychloroquine (HCQ) and chloroquine are 4-aminoquinoline drugs developed in the mid-20th century for the treatment of malaria [13]. Hydroxychloroquine differs from chloroquine only in the addition of a hydroxyl group and is associated with a lower incidence of adverse effects with chronic use [13]. Both drugs have been used in the treatment of autoimmune diseases because of their immunomodulatory effects on several cytokines, including interleukin-1 (IL-1) and IL-6 [13]. There is some evidence that these drugs also have antiviral properties against many different viruses, including the coronaviruses [14, 15]. They have demonstrated in vitro activity against SARS-CoV-2, which range considerably between studies, but are generally within the range of predicted achievable tissue concentrations [14, 16-18]. The in vitro activity, the extensive use for other conditions, and widespread availability of generic versions of the drug made it an attractive option for treatment of COVID-19. Interest in combinations of HCQ with azithromycin (AZ) began when investigators in a small, uncontrolled study of hydroxychloroquine use for COVID-19 noticed a higher frequency of patients achieving virologic response in the six subjects who received AZ to prevent bacterial infection [19]. Azithromycin, widely utilized as an antibacterial agent, has also been shown to have in vitro antiviral activity against a variety of ribonucleic acid viruses [20-22]. While the exact mechanism of antiviral activity is unknown, possibilities include inhibiting endocytosis and limiting viral replication [23] and the induction of interferon [22, 24]. Macrolides have also been shown to have anti-inflammatory activity [25, 26].

Summary of the evidence

Our search identified eight RCTs and seven comparative cohort studies of hospitalized patients with confirmed COVID-19 treated with HCQ with reported mortality, clinical progression or clinical improvement, and adverse events outcomes [27-41] (Supplementary Table s3a) (Table 1).

In addition, we identified two RCTs, four comparative cohort studies, one case-control study, and three single-arm studies reporting adjusted analyses of hospitalized patients with confirmed COVID-19 treated with HCQ plus AZ with reported mortality, failure of virologic clearance (assessed with polymerase chain reaction [PCR] test), clinical improvement, and adverse events (i.e., significant QT prolongation leading to treatment discontinuation) [19, 27, 28, 37, 39, 41-45] (Supplementary Table s3b) (Table 2).

Benefits

Hydroxychloroquine

Five RCTs showed a trend toward mortality among patients with COVID-19 treated with HCQ compared to those who were not (relative risk [RR]: 1.08; 95% confidence interval [CI]: 0.99, 1.19, Moderate certainty in the evidence) (Table 1) [28, 29, 33].

Hydroxychloroquine + azithromycin

One RCT could not exclude the risk of in-hospital mortality among patients treated with HCQ+AZ compared to those not receiving HCQ or HCQ+AZ (hazard ratio [HR]: 0.64; 95% CI: 0.18, 2.21; Low certainty of evidence [CoE]) [28]. Three non-randomized studies failed to identify an association between treatment with HCQ+AZ and mortality: Ip reported an adjusted HR of 0.98 (95% CI: 0.75, 1.28); Magagnoli reported an adjusted HR in a subset after propensity score adjustment of 0.89 (95% CI: 0.45, 1.77); Rosenberg 2020 reported an adjusted HR of 1.35 (95% CI: 0.79, 2.40) [37, 39, 41]. As stated in the HCQ section, one non-randomized study reported a reduction in mortality among patients receiving HCQ+AZ (HR: 0.29; 95% CI: 0.22, 0.40); however, it failed to adjust for the critical confounder of disease severity and imbalances in steroid use [27]. As described in the HCQ section, similar methodologic concerns exist among patients allocated to HCQ+AZ in the Arshad study, leading to several sources of bias in interpreting their favorable results.

Harms

Hydroxychloroquine

One RCT reported that persons treated with HCQ experienced a longer time until hospital discharge (median 16 days compared with 13 days) and lower probability of being discharged alive within the 28-day study period (rate ratio: 0.92; 95% CI: 0.85, 0.99) [29]. In addition, persons treated with HCQ who were not on mechanical ventilation at baseline were more likely to be placed on mechanical ventilation during follow up (rate ratio: 1.10; 95% CI: 0.92, 1.31; Low CoE) [29, 32]. Across the body of evidence from four RCTs, treatment with HCQ may increase the risk of experiencing adverse events (RR: 2.36; 95% CI: 1.49, 3.75; Low CoE) and severe adverse events (adjusted odds ratio: 1.26; 95% CI: 0.56, 2.84; Low CoE) [28, 30, 31, 35]. One RCT and two non-randomized studies suggest increased risk of QT prolongation among patients treated with HCQ compared to those not receiving HCQ (RR: 8.47; 95% CI: 1.14, 63.03; Low CoE and RR: 2.89; 95% CI: 1.62, 5.16; Very low CoE, respectively) [28, 38, 39]. In addition, Rosenberg 2020 reported 16% of patients in the HCQ arm experienced arrhythmias compared with 10% in the non-HCQ arm (RR: 1.56; 95% CI: 0.97, 2.50; Very low CoE).

Gastrointestinal side effects occurred in 7% of patients in a prospective cohort study in 224 COVID-19 uninfected patients with systemic lupus erythematosus (SLE) who received either chloroquine or hydroxychloroquine for routine care [46].

While the 4-aminoquinolines, chloroquine and HCQ, have not been demonstrated to cause hemolysis in people with glucose-6-phosphate dehydrogenase (G6PD) deficiency [47, 48], case reports of hemolysis have emerged when these agents have been used for the treatment of COVID-19 [49-51]. It is possible that infection with SARS-CoV-2 may trigger hemolysis in G6PD deficient individuals in the absence of a 4-aminoquinolone. Caution should be exercised in administering these agents to G6PD deficient individuals with COVID-19, particularly if used for extended durations.

Renal clearance accounts for 15-25% of total clearance of HCQ; however, dose adjustments are not recommended with kidney dysfunction. Chloroquine and HCQ are metabolized by cytochrome P450 isoenzymes 2C8, 2D6, and 3A4 [52]. Therefore, inhibitors and inducers of these enzymes may result in altered pharmacokinetics of these agents.

Hydroxychloroquine + azithromycin

One RCT suggests increased risk of QT prolongation among patients treated with HCQ+AZ compared to those not receiving HCQ (RR: 8.50; 95% CI: 1.16, 62.31; Low CoE) [28]. Two studies described significant QT prolongation in 10 of 95 patients treated with HCQ+AZ, illustrating the high risk for clinically relevant arrhythmias with this treatment [43, 45]. In addition, several case reports of QT prolongation related to HCQ have also been published [53-56]. A case-control study of persons with COVID-19 treated with HCQ+AZ compared to healthy, untreated controls reported higher values of minimum (415 vs. 376 ms), mean (453 vs. 407 ms) and maximum QTc-interval (533 vs. 452 ms) among COVID-19 cases (n=22) compared to controls (n=34) [42].

Additional case reports have cited the risk of a prolonged QT prolongation, torsades de pointes, and ventricular tachycardia in patients without COVID-19 receiving AZ alone. In a large cohort study, patients taking a five-day course of AZ had an increased risk of sudden cardiac death with a HR of 2.71 (1.58-4.64) vs. 0.85 (0.45-1.60), compared to patients receiving either no antibiotic or amoxicillin, respectively [57]. Given the cumulative effect on cardiac conduction seen with HCQ and AZ, if this combination was used, baseline and follow-up electrocardiogram (ECG) monitoring would be indicated, as well as careful surveillance for other concomitant medications known to prolong the QT interval.

Azithromycin has a low risk for cytochrome P450 interactions [58]; however, additional pharmacologic adverse events including gastrointestinal effects and QT prolongation need to be carefully considered, particularly in the outpatient setting where frequent ECG monitoring is not feasible.

Providers are encouraged to visit resources such as https://www.covid19-druginteractions.org/ to aid in the evaluation and management of drug interactions with current and emerging investigational agents for COVID-19.

Other considerations

The panel agreed that the overall certainty of evidence against treatment with HCQ was moderate due to concerns with imprecision around the risk for a trend towards harms from increased mortality. When considering the addition of AZ, the overall certainty of the evidence was low; however, the panel recognized even greater concern with the toxicity. In addition, based on the moderate certainty of increased QT prolongation, the panel determined that this demonstrated certain harm with uncertain benefit; therefore, the panel made a strong recommendation against HCQ+AZ.

Conclusions and research needs for this recommendation

The guideline panel recommends against the use of either HCQ alone or in combination with AZ in the hospital setting as higher certainty benefits (e.g., mortality reduction) are now highly unlikely even if additional high quality RCTs would become available.

This recommendation does not address the use of azithromycin for secondary bacterial pneumonia in patients with COVID-19 (Supplementary Table s2).

Table 01.png

 

Table 02.png

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendation 3: Hydroxychloriquine as post-exposure prophylaxis

Section last reviewed and updated 9/23/2021

Last literature search conducted 9/21/2021

Recommendation 3: In persons exposed to COVID-19, the IDSA guideline panel recommends against hydroxychloroquine. (Strong recommendation, Low certainty of evidence)

 

Why is hydroxychloroquine considered for post-exposure prophylaxis?

There is some evidence that HCQ has antiviral properties against many different viruses, including the coronaviruses [14, 15]. It has demonstrated in vitro activity against SARS-CoV-2, which ranges considerably between studies, but is generally within the range of predicted achievable tissue concentrations [14, 16-18]. The in vitro activity, the extensive use for other conditions, and widespread availability of generic versions of the drug made it an attractive option for treatment and prophylaxis of COVID-19; however, at this point, HCQ has not been identified as effective for treatment of COVID-19.

Summary of the evidence

Our search identified three RCTs that reported on HCQ post-exposure prophylaxis of contacts of those diagnosed with SARS-CoV-2 infection [59-61]. Patients in these studies were randomized to HCQ or placebo or no additional treatment. All three studies evaluated for the presence of SARS-CoV-2 at day 14, two of the studies required a positive test for SARS-CoV-2, while one allowed symptoms suggestive of COVID-19 to meet the outcome when a test was not completed. Additional outcomes included hospitalization, mortality, and serious adverse events.

Benefits

Outpatients

Hydroxychloroquine appears to have trivial or no effect on the development of symptomatic SARS-CoV-2 infection at day 14 compared to no HCQ (RR: 0.95; 95% CI: 0.77, 1.16; moderate CoE). In addition, HCQ showed trivial or no effect on the rate of hospitalization (RR: 1.00; 95% CI: 0.47, 2.12; 3 fewer to 7 more hospitalizations in 1,000; low CoE) or mortality (RR: 0.45; 95% CI: 0.16, 1.28; 5 fewer to 2 more deaths in 1,000; low CoE).

Harms

There was no difference in serious adverse events in the HCQ rather than no HCQ for post-exposure prophylaxis (RR: 0.91; 95% CI: 0.47, 1.76; moderate CoE). Additional side effects and harms of HCQ (e.g., QT prolongation, arrhythmias, gastrointestinal effects) have been summarized in recommendation 1 (HCQ for treatment of hospitalized persons with COVID-19).

Other considerations

The panel made an explicit decision that:

  1. The primary outcome driving the decision for any post-exposure prophylaxis is the ability to prevent infection
  2. When the evidence demonstrates a very low likelihood of effective post-exposure prophylaxis, other outcomes become secondary
  3. When healthy persons are considered for preventive medications (such as would occur in post-exposure settings), a higher threshold for benefits is required and (even putative) harms become more important

The panel agreed that the overall certainty of the evidence against prophylaxis treatment with HCQ was moderate (failure to prevent infection) due to concerns with imprecision. The panel balanced the lack of clear benefit with the increased risk of harms from the body of evidence reported in the treatment section, in addition to the side effects reported in the trials to make a strong recommendation.

Conclusions and research needs for this recommendation

The guideline panel recommended against the use of HCQ as post-exposure prophylactic treatment for persons exposed to COVID-19.

 

Table 03.png

 

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendation 4: Lopinavir/ritonavir 

SPECIAL UPDATE ALERT (2/22/2022): This section has been updated based on newly available literature and approvals. This update will be fully integrated into the website soon; it is provided here for immediate use.

Two new recommendations on the use of lopinavir/ritonavir (prophylaxis for persons exposed to SARS-CoV-2; treatment for ambulatory patients with mild-to-moderate COVID-19) have been developed. The full updated section featuring the new recommendations can be viewed here (PDF).


Section last reviewed and updated 11/22/2020

Last literature search conducted 11/18/2020

Recommendation 4: Among hospitalized patients with COVID-19, the IDSA guideline panel recommends against the use of the combination lopinavir/ritonavir. (Strong recommendation, Moderate certainty of evidence)

The last literature search was conducted on November 18, 2020.

Why is lopinavir plus ritonavir considered for treatment?

Lopinavir/ritonavir is a protease inhibitor that was U.S. Food and Drug Administration (FDA)-approved for the treatment of HIV in September 2000. Ritonavir is added to the combination as a pharmacokinetic enhancer due to its strong inhibition of cytochrome P450 3A4, a metabolic pathway for lopinavir metabolism. Lopinavir/ritonavir demonstrated in vitro inhibition of SARS-CoV-1 and MERS-CoV replication [62-64]. A trial of lopinavir/ritonavir and ribavirin vs historical controls in SARS-CoV-1 patients, showed a reduced rate of ARDS and mortality in those receiving lopinavir/ritonavir. This study had limitations including a control group from early in the outbreak when management strategies likely differed significantly [65]. During the MERS outbreak, case reports cited efficacy of lopinavir/ritonavir with interferon in the management of MERS patients [66, 67]. During the early phase of COVID-19, triple combination of interferon beta-1b, lopinavir-ritonavir, and ribavirin shortened the duration of viral shedding and hospital stay in patients with mild to moderate COVID-19 in an open-label, randomized, phase II trial [68].

Summary of the evidence

Three RCTs reported on treatment with combination lopinavir/ritonavir or placebo for hospitalized patients with COVID-19 [32, 69, 70] (Table 4). The trials reported on the following outcomes: mortality, failure of clinical improvement (measured using a 7-point scale or hospital discharge), need for mechanical ventilation, and adverse events leading to treatment discontinuation.

Benefits

Among hospitalized patients with COVID-19, treatment with lopinavir/ritonavir failed to show or exclude a beneficial effect on mortality or need for invasive mechanical ventilation (RR: 1.00; 95% CI: 0.89, 1.13; moderate CoE and RR: 1.12; 95% CI: 0.93, 1.34; low CoE). Similarly, lopinavir/ritonavir may reduce failure of clinical improvement at 14 days, but it is uncertain (RR: 0.78; 95% CI: 0.63, 0.97; very low CoE).

Harms

RECOVERY reported 1/1588 serious adverse event due to treatment with lopinavir-ritonavir [70]; however, nearly 14% of lopinavir/ritonavir recipients in Cao 2020 were unable to complete the full 14-day course of administration. This was due primarily to gastrointestinal adverse events, including anorexia, nausea, abdominal discomfort, or diarrhea, as well as two serious adverse events, both acute gastritis. Two recipients had self-limited skin eruptions. Such side effects, including the risks of hepatic injury, pancreatitis, more severe cutaneous eruptions, and QT prolongation, and the potential for multiple drug interactions due to CYP3A inhibition, are well documented with this drug combination. The side-effect profile observed in these trials raise concerns about the use of higher or more prolonged lopinavir–ritonavir dose regimens in efforts to improve outcomes. 

Other considerations

The panel determined the certainty of evidence to be moderate due to concerns with imprecision. The guideline panel made a strong recommendation against treatment with the combination of lopinavir/ritonavir for hospitalized patients with COVID-19.

Conclusions and research needs for this recommendation

The guideline panel recommends against treatment with lopinavir/ritonavir in hospitalized patients with COVID-19.

 

Table 04.png

 

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendations 5-7: Glucocorticoids

Section last reviewed and updated 9/25/2020

Last literature search conducted 9/4/2020

Recommendation 5: Among hospitalized critically ill patients* with COVID-19, the IDSA guideline panel recommends dexamethasone rather than no dexamethasone. (Strong recommendation, Moderate certainty of evidence)

  • Remark: If dexamethasone is unavailable, equivalent total daily doses of alternative glucocorticoids may be used. Dexamethasone 6 mg IV or PO for 10 days (or until discharge) or equivalent glucocorticoid dose may be substituted if dexamethasone unavailable. Equivalent total daily doses of alternative glucocorticoids to dexamethasone 6 mg daily are methylprednisolone 32 mg and prednisone 40 mg.

Recommendation 6: Among hospitalized patients with severe**, but non-critical, COVID-19 the IDSA guideline panel suggests dexamethasone rather than no dexamethasone. (Conditional recommendation, Moderate certainty of evidence)

  • Remark: Dexamethasone 6 mg IV or PO for 10 days (or until discharge) or equivalent glucocorticoid dose may be substituted if dexamethasone unavailable. Equivalent total daily doses of alternative glucocorticoids to dexamethasone 6 mg daily are methylprednisolone 32 mg and prednisone 40 mg.

Recommendation 7: Among hospitalized patients with non-severe*** COVID-19 without hypoxemia requiring supplemental oxygen, the IDSA guideline panel suggests against the use of glucocorticoids. (Conditional recommendation, Low certainty of evidence)

Severity definitions:

  • *Critical illness is defined as patients on mechanical ventilation and ECMO. Critical illness includes end organ dysfunction as is seen in sepsis/septic shock. In COVID-19, the most commonly reported form of end organ dysfunction is ARDS
  • **Severe illness is defined as patients with SpO2 ≤94% on room air, including patients on supplemental oxygen.
  • ***Non-severe illness is defined as patient with a SpO2 > 94% not requiring supplemental oxygen.

 

The last literature search was conducted on September 4, 2020 and we identified eight RCTs and seven comparative non-randomized studies.

 

Why are corticosteroids considered for treatment?

In the early days of the SARS-CoV-2 pandemic, based on experience in both SARS and MERS, recommendations [71] cautioned against the use of systemic corticosteroids due to risk of worsening clinical status, delayed viral clearance, and adverse events [72-74]. Given the hyper-inflammatory state in COVID-19, immunomodulatory approaches, including steroids, continue to be evaluated to address both ARDS and systemic inflammation. ARDS stemming from dysregulated systemic inflammation may translate into prolonged ventilatory requirements and in-hospital mortality. In non-viral ARDS settings, there is increasing support for the role of steroids in the management of ARD [75]. A recent multicenter RCT in patients with moderate to severe ARDS demonstrated a reduced number of ventilatory days and reduction in mortality with use of a 10-day regimen of dexamethasone [76].

Summary of the evidence

Critical illness

Our search identified one systematic review that analyzed eight RCTs reporting on treatment with glucocorticoids among 1,844 critically ill patients with COVID-19 [77]. Three RCTs reported on patients treated with low- and high-dose dexamethasone [76, 78, 79]; three RCTs reported on patients treated with low-dose hydrocortisone [80-82]; and two RCTs reported on patients treated with high-dose methylprednisolone [77, 83]. The definition of critically ill varied across trials; however, the majority of patients had ARDS.

Severe and non-severe illness

Our search identified one RCT, one “partially” randomized trial, one prospective cohort, and five retrospective cohort studies [78, 84-90]. The RCT provided the best available evidence on treatment with corticosteroids for persons with COVID-19 [78] (Table 5, Table 6, and Table 7). Corral-Gudino et al. reported on a study that randomized patients to receive methylprednisolone or standard of care; however, patients expressing a preference for methylprednisolone were assigned to the same treatment arm [84]. Corral-Gudino et al. did not report the disaggregated results from the randomized trial; therefore, succumbing to the same potential for bias as reported subsequently for the non-randomized studies. The non-randomized studies had significant limitations with controlling for multiple co-interventions and disease severity at baseline [85-90]. All non-randomized studies had concerns with risk of bias due to lack of adjustment for critical confounders or potential for residual confounding. Timing of receipt, dose and duration of corticosteroids varied across studies.

The RECOVERY trial is a randomized trial among hospitalized patients in the United Kingdom [78]. In that study, 2,104 participants were randomized to receive dexamethasone (6 mg daily for up to 10 days) and 4,321 were randomized to usual care. The RECOVERY trial reported on the outcomes of mortality and hospital discharge. Participants and study staff were not blinded to the treatment arms.

Benefits

Critical illness

Among hospitalized, critically ill patients, the odds of mortality at 28 days was 34% less among patients treated with glucocorticoids than among patients not treated with glucocorticoids (OR: 0.66; 95% CI: 0.54; 0.82; high CoE). In addition, at 28 days, patients receiving dexamethasone were more likely to be discharged from the hospital (RR: 1.11; 95% CI: 1.04, 1.19; moderate CoE).

Severe illness

Among hospitalized patients, 28-day mortality was 17% lower in the group that received dexamethasone than in the group that did not receive dexamethasone (RR 0.83; 0.74-0.92; moderate CoE). In addition, at 28 days, patients receiving dexamethasone were more likely to be discharged from the hospital (RR: 1.11; 95% CI: 1.04, 1.19; moderate CoE).

Non-severe illness

In a sub-group analyses of patients without hypoxia not receiving supplemental oxygen, there was no evidence for benefit and a trend toward harm with dexamethasone in participants who were not on supplemental oxygen (RR 1.22; 0.86, 1.75; low CoE).

Harms

A systematic review of six studies did not report a difference in the events of serious adverse events experienced by patients randomized to receive treatment with glucocorticoids or no treatment with glucocorticoids (64/354 among those receiving glucocorticoids vs. 80/342 among those not receiving glucocorticoids).

Patients receiving a short course of steroids may experience hyperglycemia, neurological side effects (e.g., agitation/confusion), adrenal suppression, and risk of bacterial and fungal infection [85, 91, 92].

Other considerations

Critical illness

The panel agreed that the overall certainty of the evidence for treatment with glucocorticoids for patients with critical COVID-19 was moderate due to concerns with indirectness and imprecision.

Severe illness

The panel agreed the overall certainty of evidence for treatment with glucocorticoids for patients with severe COVID-19 as moderate due to concerns with indirectness since the evidence was from dexamethasone.

Non-severe illness

The panel agreed that the overall certainty of evidence for patients without hypoxemia requiring supplemental oxygen as low due to concerns with risk of bias (post hoc analysis) and imprecision.

The panel agreed the overall certainty of evidence for treatment with glucocorticoids for patients with severe COVID-19 as moderate due to concerns with indirectness since the evidence was from dexamethasone. The panel agreed that the overall certainty of evidence for patients without hypoxemia requiring supplemental oxygen as low due to concerns with risk of bias (post hoc analysis) and imprecision.

Conclusions and research needs for these recommendations

The guideline panel recommends dexamethasone for patients with critical COVID-19. The guideline panel suggests dexamethasone for patients with severe COVID-19. If dexamethasone is not available, then alternative glucocorticoids may be used (see details above). The guideline panel suggests against glucocorticoids for patients with COVID-19 without hypoxemia requiring supplemental oxygen.

Additional research is needed to inform the generalizability of treatment with different glucocorticoids for patients with COVID-19 (Supplementary Table s2).

 

Table 05.png

 

Table 06.png

 

Table 07.png

 

Supplementary Information

Study characteristics:

Risk of bias:

Recommendations 8-9: IL-6 inhibitors (tocilizumab and sarilumab)

Section last reviewed and updated on 9/14/2021

Last literature search conducted 8/31/2021

Recommendation 8: Among hospitalized adults with progressive severe* or critical** COVID-19 who have elevated markers of systemic inflammation, the IDSA guideline panel suggests tocilizumab in addition to standard of care (i.e., steroids) rather than standard of care alone. (Conditional recommendation, Low certainty of evidence)

  • Remarks:
    • Patients, particularly those who respond to steroids alone, who put a high value on avoiding possible adverse events of tocilizumab and a low value on the uncertain mortality reduction, would reasonably decline tocilizumab.
    • In the largest trial on the treatment of tocilizumab, criterion for systemic inflammation was defined as CRP ≥75 mg/L.

Recommendation 9: When tocilizumab is not available, for patients who would otherwise qualify for tocilizumab, the IDSA guideline panel suggests sarilumab in addition to standard of care (i.e., steroids) rather than standard of care alone. (Conditional recommendation, Very low certainty of evidence)

  • Remark: Patients, particularly those who respond to steroids alone, who put a high value on avoiding possible adverse events of sarilumab and a low value on the uncertain mortality reduction, would reasonably decline sarilumab.

 

Severity definitions:

  • *Severe illness is defined as patients with SpO2 ≤94% on room air, including patients on supplemental oxygen.
  • **Critical illness is defined as patients on mechanical ventilation and ECMO. Critical illness includes end organ dysfunction as is seen in sepsis/septic shock. In COVID-19, the most commonly reported form of end organ dysfunction is ARDS.

 

Why are are interleukin-6 (IL-6) receptor antagonists considered for treatment?

Some patients with COVID-19 develop a hyperinflammatory syndrome that is characterized by elevations in proinflammatory cytokines and multiorgan dysfunction also known as the immunopathology of SARS-CoV-2 infection. The significance of these findings is unclear, however early descriptions found that those with elevated IL-6 levels and evidence of hyperinflammation had increased rates of more severe disease [93, 94]. Tocilizumab, a monoclonal anti-IL-6-receptor blocking antibody, has been proposed as a therapeutic agent to mitigate hyperinflammation associated with COVID-19. Tocilizumab is FDA-approved for various rheumatologic conditions as well as cytokine release syndrome associated with CAR-T cell therapy.

Sarilumab, another IL-6 receptor antagonist, is currently FDA-approved for rheumatoid arthritis (RA).

Summary of the evidence

Tocilizumab

Our search identified eight RCTs (including pre-prints) that reported on patients with severe COVID-19 randomized to treatment with tocilizumab (8 mg/kg) or placebo/usual care [95-102]. Gordon 2020, Horby 2021, Rosas 2020, and Veiga 2021 allowed for patients to be on mechanical ventilation at randomization, whereas the other trials included patients with a lower disease severity (e.g., allowed supplemental oxygen but excluded those on higher levels of oxygen support) or included patients with severe COVID with an inflammatory phenotype.

One trial, RECOVERY, contributed the majority of the weight in the analysis [97]. RECOVERY trial participants must have demonstrated clinical evidence of progressive COVID-19, which was defined as <92% oxygen saturation on room air or receiving oxygen and C-reactive protein (CRP) ≥75 mg/L. Use of steroids was balanced across both the participants receiving tocilizumab or not receiving tocilizumab. Following recommendations for treatment with glucocorticoids, 82% of participants in both arms received dexamethasone. While RECOVERY did not blind participants or healthcare personnel to the randomized treatment arm, this likely would not introduce bias in the objective measurement of the outcome of mortality; however, it was considered as a risk of bias for more subjectively measured outcomes, clinical deterioration, along with the total body of evidence contributing to those outcomes (Table 8). There are limited safety data in the preliminary report.

Both RECOVERY and REMAP CAP (the two tocilizumab trials that reported a benefit) initiated treatment early (randomization at median of two days of hospitalization in RECOVERY; <24 hours in the ICU for REMAP-CAP), suggesting tocilizumab may be more beneficial early in people with rapidly progressive disease.

Sarilumab

We identified three RCTs that reported on patients with severe or critical COVID-19 randomized to treatment with sarilumab or placebo/usual care [95, 103, 104]. In addition, a pre-print network meta-analysis of 18 RCTs was identified that reported network estimates for sarilumab plus corticosteroids compared with usual care alone [105].

Benefits

Tocilizumab

Among hospitalized patients, tocilizumab showed a trend toward reduced mortality at 28 days compared to no tocilizumab treatment (RR: 0.91; 95% CI: 0.79, 1.04; moderate CoE). Tocilizumab demonstrated a lower relative risk of clinical deterioration, defined as death, need for mechanical ventilation, ECMO, or ICU admission, compared to placebo/usual care, RR: 0.83 (95% CI: 0.77, 0.89; moderate CoE). Four studies were not blinded, while in the remaining three trials healthcare personnel and outcome assessors were blinded. The panel noted that tocilizumab causes a decline in CRP levels, which if obtained would reveal the treatment arm designations of the patients, therefore introducing bias for the more subjectively measured outcomes of clinical deterioration and serious adverse events.

Sarilumab

Among hospitalized patients, sarilumab showed a trend toward reduced mortality at 28 days compared to usual care (network estimate OR: 0.80; 95% CI: 0.61, 1.04; low certainty of evidence). Sarilumab may reduce clinical deterioration, defined as progression to intubation, ECMO or death compared to usual care (RR: 0.67; 95% CI: 0.42, 1.05; very low CoE).

Harms

Serious adverse events among patients receiving tocilizumab or sarilumab did not differ from those receiving usual care (RR: 0.89; 95% CI: 0.74, 1.07; low CoE and RR: 1.03; 95% CI: 0.89, 1.18; low CoE, respectively). An additional trial attributed treatment with tocilizumab to three serious adverse events; however, did not report events among patients not receiving tocilizumab [97]. Previously, tocilizumab has been associated with gastrointestinal perforations in non-COVID-19 settings, and case reports of bowel perforations have recently emerged with the use of tocilizumab for COVID-19 [106-109]. Increased infection risks have been noted in uncontrolled studies, and it is possible that this risk may be compounded by the combination of glucocorticoids and tocilizumab. [110, 111].

Other considerations

While the overall certainty of evidence for the trend toward a reduction in mortality was moderate, the panel believes that differences in mortality rates across the trials may be the result of the differences in baseline severity of study participants and timing of tocilizumab receipt in the disease course. In REMAP-CAP, tocilizumab was administered within 24 hours of participants’ initiating organ support in an intensive care unit, raising the possibility that this may be the optimal time to administer the drug. In RECOVERY, tocilizumab was administered to participants with oxygen saturation <92% on room air or receiving oxygen therapy, and CRP ≥75 mg/L. Given the reduction in clinical deterioration and trend toward mortality reduction, the guideline panel made a conditional recommendation for treatment of adults with tocilizumab.

The use of tocilizumab, as with other therapeutic agents that can suppress the immune system, presents additional considerations and potential concerns when used in immunocompromised hosts. The panel did not conduct an analysis of available data to assess differences in efficacy and/or adverse effects of tocilizumab among oncology or other immunocompromised patients at this time.

The panel recognized the current shortage of tocilizumab and possible net benefit of treatment with sarilumab. 

Conclusions and research needs for this recommendation

The guideline panel suggests tocilizumab for hospitalized adults with COVID-19. When tocilizumab is not available and baricitinib is either not appropriate or available, the guideline panel suggests sarilumab for persons who would otherwise qualify for tocilizumab; however, it is acknowledged that patients, particularly those responding to steroids alone or baricitinib, who put a high value on avoiding the possible adverse events of sarilumab and a low value on the uncertain mortality reduction would reasonably decline sarilumab.

Additional research is needed to understand the efficacy of tocilizumab when taken at different times during the course of disease. For example, there are no data to guide recommendations in patient <18 years of age at this time. In addition, future studies are needed to inform the generalizability of tocilizumab with different IL-6 receptor inhibitors for patients with COVID-19 (Supplementary Table s2). At the time of update, preliminary data from a trial of treatment with sarilumab has been shared as a pre-print [95]; however, number of patients who received sarilumab is limited (n=45) and the published manuscript was not available for analysis or inclusion to inform this recommendation. Other studies of sarilumab have not been made available.

 

Table 08.png

 

Table 09.png

 

 

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendations 10-11: Convalescent plasma

SPECIAL UPDATE ALERT (2/8/2022): This section has been updated based on newly available literature and approvals. This update will be fully integrated into the website soon; it is provided here for immediate use.

Recommendation 11 has been revised to address the use of convalescent plasma in ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease with no other treatment options. The full updated section can be viewed here (PDF).


Section last reviewed and updated 11/11/2021

Last literature search conducted 10/31/2021

Recommendation 10: Among patients hospitalized with COVID-19, the IDSA guideline panel recommends against COVID-19 convalescent plasma. (Strong recommendation, Moderate certainty of evidence)

Recommendation 11: Among ambulatory patients with mild to moderate COVID-19, the IDSA guideline panel suggests against COVID-19 convalescent plasma outside of the context of a clinical trial. (Conditional recommendation, Low certainty of evidence)

 

Why is convalescent plasma considered for treatment?

Convalescent plasma has been used as passive immunotherapy for prevention and treatment of infections for over 100 years [112, 113]. The predominant proposed protective mechanism is thought to be pathogen neutralization, although antibody-dependent cellular cytotoxicity and enhanced phagocytosis may also play a role. With the advent of effective antimicrobial therapy (i.e., “the antibiotic era”), convalescent plasma fell out of favor. In recent years, interest in this approach has been revived as a means of addressing viral epidemics such as Ebola, SARS-CoV-1, and MERS. Studies of convalescent plasma derived from people who had recovered from those specific infections showed encouraging results but were typically small, non-randomized, and largely descriptive [114-116]. In the current pandemic, convalescent plasma obtained from individuals who have recovered from COVID-19 has been used in over 100,000 patients with moderate to severe infection as part of an expanded access program [117, 118]. When measurement of neutralizing antibody titers is available, the FDA recommends neutralizing antibody titers of ≥1:160. Assays to measure neutralizing antibody titers were not widely available early in the pandemic, so it is unclear if the plasma used in the context of the expanded access program had adequate titers of neutralizing antibodies meeting the FDA targets. Multiple prospective clinical trials are in progress utilizing plasma with an IgG enzyme-linked immunosorbent assay (ELISA) titer cutoff of ≥1:320. Titers at that level are seen in about 80% of donors [119]. The probability of obtaining a neutralizing antibody titer of ≥1:160 is highest (80% or greater) when the ELISA IgG titer is ≥1:1,350 [120]. In an analysis of the convalescent plasma expanded access program, higher levels of antibodies were associated with significant improvements in mortality compared to those receiving convalescent plasma with lower concentrations of neutralizing antibodies [117]. However, there was no placebo group in the study, so this result could be from increased mortality with low antibody titer plasma rather than improved mortality with high antibody titer plasma. Subgroup data from one open-label RCT reporting on plasma with anti-receptor-binding domain ELISA values corresponding to a high antibody titer cutoff resulted in a non-significant relative risk reduction in mortality of 5% (RR: 0.95; 95% CI: 0.73, 1.25) [121]. An additional subgroup analysis suggested unselected convalescent plasma (i.e., not limited to high-titer antibodies) may increase the relative risk for mortality by 49% (RR: 1.42; 95% CI: 0.92, 1.69).

An analysis of the convalescent plasma expanded access program suggests the most benefit is seen when convalescent plasma is given in the first three days from diagnosis [117]. In August 2020, the FDA issued an emergency use authorization (EUA) for investigational convalescent plasma for the treatment of COVID-19 in hospitalized patients [122]. In early February 2021, the FDA issued a revision to the EUA to limit the authorization to the use of high-titer COVID-19 convalescent plasma for the treatment of hospitalized patients early in the disease course [123].

Summary of the evidence

Our search identified and was informed by evidence from 20 RCTs and a large (n=20,000), single-arm registry study [112-116, 119, 120, 124-130], as they provided the best available evidence for the outcomes of mortality, need for mechanical ventilation, serious adverse events, and adverse events. Eighteen of those RCTs reported on convalescent plasma transfusions for patients hospitalized with COVID-19 (Table 10) [112-115, 119, 120, 124-127] and two RCTs [129, 130] reported on receipt of convalescent plasma by ambulatory persons with mild COVID-19 disease (Table 11) [116].

Eighteen trials randomized 17,232 patients hospitalized with COVID-19 to receive a transfusion with COVID-19 convalescent plasma [112-115, 119, 120, 124-127]. Several trials were open-label and/or had concerns with risk of bias due to lack of adjustment for critical confounders or potential for residual confounding (Supplementary Table s14a). Timing of receipt of COVID-19 convalescent plasma during the clinical course of the patients' illnesses varied across studies (Supplementary Table s13). One trial reported on 160 persons who received high-titer convalescent plasma less than 72 hours after the onset of symptoms of COVID-19 (mean age: 77.2 years; standard deviation: ±8.6 years) [116]. In addition, Joyner 2020 reported on safety outcomes of over 20,000 patients enrolled in the same FDA Expanded Access Program for COVID-19 convalescent plasma study.

Benefits

Hospitalized patients

Convalescent plasma transfusion appears to have trivial or no effect on mortality based on the body of evidence from RCTs (RR: 0.98; 95% CI: 0.93, 1.03; moderate CoE). Recipients of COVID-19 convalescent plasma may have a greater need for mechanical ventilation (RR: 1.10; 95% CI: 0.94, 1.29; low CoE); however, the evidence is uncertain because of concerns with risk of bias imprecision.

Ambulatory persons

Receipt of COVID-19 convalescent plasma failed to demonstrate or exclude a beneficial effect on COVID-19 related hospitalizations or medically-attended visits (emergency room or urgent care; RR 0.94; 95% CI: 0.73 to 1.22). Prior evidence showed a possible reduction of progression to severe respiratory disease (RR: 0.52; 95% CI: 0.29, 0.94; low CoE); however, the evidence remains uncertain, as oxygenation and respiration rates are surrogate measures of need for ventilation, morbidity, and death, and because of the fragility of the estimate due to the small number of events reported. Convalescent plasma transfusion failed to show or exclude a beneficial effect on mortality based on the body of evidence from two RCTs (RR: 0.78; 95% CI: 0.2, 3.1; low CoE); however, the evidence is uncertain due to concerns with fragility of the estimate due to the small number of events reported. Additional deaths beyond 15 days were reported in one RCT and included five deaths in the plasma group versus one in the placebo arm.

Harms

In the largest safety study (n=20,000), within four hours of completion of convalescent plasma transfusion, authors reported 146 serious adverse events classified as transfusion reactions (<1% of all transfusions) [128]. Of these, 63 deaths were reported (0.3%) with 13 judged as possibly or probably related to the transfusion. The non-mortality serious adverse events include 37 reports of transfusion-associated circulatory overload, 20 cases of transfusion-related acute lung injury, and 26 cases of severe allergic transfusion reactions.

Within seven days of transfusion, 1711 deaths were reported (mortality rate: 8.56%; 95% CI: 8.18, 8.95). In addition, 1136 serious adverse events were reported: 643 cardiac events (569 judged as unrelated to the transfusion), 406 sustained hypotensive events requiring intravenous (IV) pressor support, and 87 thromboembolic or thrombotic events (55 judged as unrelated to the transfusion).

Eleven trials among patients hospitalized for COVID-19 suggest increased adverse events among patients receiving convalescent plasma (RR: 1.08; 95% CI: 0.94, 1.26; low CoE); however, the evidence was uncertain due to concerns with lack of blinding. In addition, included studies lacked a standard definition for what met the definition of an adverse event. In ambulatory patients, serious adverse events were higher in the convalescent plasma group due to serious transfusion reactions (RR 5.9; 95% CI: 0.30, 118.0; low CoE), although the evidence is uncertain due to few events.

Other considerations

Hospitalized patients

The panel agreed that the overall certainty of evidence is moderate due to some remaining imprecision as the 95% CI crossed the threshold of 1% for plausible mortality reduction. The guideline panel recognized that unselected use of convalescent plasma appeared to have trivial to no beneficial effect from the now existing large body of evidence.

Ambulatory persons

The panel agreed that the overall certainty of evidence is low due to concerns with risk of bias and imprecision, which recognized the limited events and concerns with fragility. The guideline panel recognized the inability to exclude a meaningful beneficial or detrimental effect when plasma is given early in the course of COVID-19 disease.

Conclusions and research needs for this recommendation

The guideline panel suggests against COVID-19 convalescent plasma for persons hospitalized with COVID-19. In addition, the guideline panel suggests against COVID-19 convalescent plasma for ambulatory persons outside of the context of a clinical trial. Based on limited studies and mechanistic reasoning, COVID-19 convalescent plasma may be more effective if given at high titers early in course of hospitalization, in patients with undetectable or low levels of SARS-CoV-2 neutralizing antibodies, or in those with a humoral immune deficiency [131-136]. Current RCTs have not reported outcomes in such pre-specified subpopulations. Future studies in hospitalized patients should focus on patients with humoral immunodeficiencies early in the course of COVID-19. Future studies in hospitalized patients should also consider screening for SARS-CoV-2 neutralizing antibodies in all patients at entry into RCTs and assessing outcomes based on antibody levels.

Similarly, in ambulatory patients, convalescent plasma may be more effective if the product used contains high titers of neutralizing antibodies and is used early in clinical presentation or in subpopulations of patients who do not have an adequate humoral immune response even at later stages of disease [131]. There is a paucity of trials in this specific population of patients. Future studies in ambulatory patients should target these populations.

Additional clinical trials may be needed to also determine whether there is a benefit of treatment with COVID-19 convalescent plasma and at what dose (neutralizing antibody titers), especially for patients early in the disease course of COVID-19 (Supplementary Table s2).

 

Table 10.png

 

Table 11.png

 

 

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendations 12-15: Remdesivir

SPECIAL UPDATE ALERT (2/16/2022): This section has been updated based on newly available literature and approvals. This update will be fully integrated into the website soon; it is provided here for immediate use.

Recommendation 12 has been revised to address the use of remdesivir in patients (ambulatory or hospitalized) with mild-to-moderate COVID-19 at high risk for progression to severe disease. The full updated section can be viewed here (PDF).


Section last reviewed and updated 12/23/2021

Last literature search conducted 11/30/2021

Recommendation 12 (NEW, 12/23/2021): Among ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease, the IDSA guideline panel suggests remdesivir initiated within seven days of symptom onset rather than no remdesivir. (Conditional recommendation, Low certainty of evidence)

  • Remarks:
    • Dosing for remdesivir is 200 mg on day one followed by 100 mg on days two and three.
    • Patients with mild to moderate COVID-19 who are at high risk of progression to severe disease admitted to the hospital for reasons other than COVID-19 may also receive remdesivir
    • Options for treatment and management of ambulatory patients include nirmatrelvir/ritonavir, three-day treatment with remdesivir, molnupiravir, and neutralizing monoclonal antibodies. Patient-specific factors (e.g., symptom duration, renal function, drug interactions), product availability, and institutional capacity and infrastructure should drive decision-making regarding choice of agent. Data for combination treatment do not exist in this setting.

Recommendation 13a: In hospitalized patients with severe* COVID-19, the IDSA panel suggests remdesivir over no antiviral treatment. (Conditional recommendation, Moderate certainty of evidence)

  • Severity definition: *Severe illness is defined as patients with SpO2 ≤94% on room air.

Recommendation 13b: In patients with COVID-19 on invasive ventilation and/or ECMO, the IDSA panel suggests against the routine initiation of remdesivir (Conditional recommendation, Very low certainty of evidence)

Recommendation 14: In patients on supplemental oxygen but not on mechanical ventilation or ECMO, the IDSA panel suggests treatment with five days of remdesivir rather than 10 days of remdesivir. (Conditional recommendation, Low certainty of evidence)

Recommendation 15: In patients with COVID-19 admitted to the hospital without the need for supplemental oxygen and oxygen saturation >94% on room air, the IDSA panel suggests against the routine use of remdesivir. (Conditional recommendation, Very low certainty of evidence)

 

Why is remdesivir considered for treatment?

Remdesivir (GS-5734) is an antiviral drug with potent in vitro activity against a range of RNA viruses including MERS-CoV, SARS-CoV 1 & 2 [137-139]. Remdesivir acts by causing premature termination of viral RNA transcription [139]. Its use improved disease outcomes and reduced viral loads in SARS-CoV-1 infected mice [138]. In rhesus macaques, therapeutic treatment with remdesivir showed reduction in SARS-CoV-2 loads, pathologic changes, and progression of clinical disease [140]. In this same animal model, remdesivir treatment initiated 12 hours post-inoculation reduced clinical signs, virus replication in the lungs, and decreased the presence and severity of lung lesions.

Summary of the evidence

Ambulatory patients with mild to moderate disease who are at high risk for progression to severe COVID-19

One RCT compared treatment with three days of intravenous (IV) remdesivir (200 mg on day one followed by 100 mg on days two and three) or no remdesivir in unvaccinated patients [141]. The study enrolled patients at high risk for progression (e.g., obesity, diabetes mellitus, hypertension, immune compromise etc.) or age 60 years or older who were symptomatic seven days or less without prior treatment (e.g., monoclonal antibodies), but were not expected to receive oxygen at time of enrollment (>94% on room air). The outcomes assessed were mortality, hospitalizations for any cause, and COVID-19-related medically as well as serious adverse events.

Hospitalized patients with oxygen saturation >94% without supplemental oxygen

(Three RCTs compared treatment with five days of remdesivir (200 mg day one, 100 mg daily days 2-5), 10 days of remdesivir (200 mg day one, 100 mg daily days 2-10), or no remdesivir for patients hospitalized with oxygen saturation >94% on room air [32, 142, 143] (Table 15). The outcomes assessed were mortality, clinical improvement, and serious adverse events. Adaptive Covid-19 Treatment Trial (ACTT-1) and SOLIDARITY provided subgroup analyses among patients with mild to moderate disease [32, 142]. Randomization and lack of blinding failed to control for or balance receipt of co-interventions (e.g., treatment with dexamethasone, tocilizumab, hydroxychloroquine, and lopinavir/ritonavir) equally across arms in Spinner et al (2020) [143]. In addition, the Spinner et al did not adjust for severity of disease. 

Hospitalized patients with SpO2 ≤94% on room air

Three RCTs comparing treatment with remdesivir (200 mg day one, 100 mg daily days 2-10) against no remdesivir treatment [32, 142, 144], and one RCT comparing five days of treatment (200 mg day one, 100 mg daily days 2-5) against 10 days (200 mg day one, 100 mg daily days 2-10) of treatment [145] served as the best available evidence among hospitalized persons with severe COVID-19 (Tables 13a, 13b, Table 14). The outcomes assessed were mortality, time to clinical improvement, need for mechanical ventilation, serious adverse events, and adverse events leading to treatment discontinuation.

All trials used different definitions of severe disease for participants. ACTT-1 participants were considered to have severe disease if they required mechanical ventilation, supplemental oxygen, if SpO2 was 94% or lower while breathing ambient air, or if they had tachypnea (respiratory rate >24 breaths per minute) [142]. Within the SOLIDARITY trial (available only as a pre-print at this time), participants with severe disease were receiving mechanical ventilation [32]. In Wang 2020, severe participants had a SpO2 <94% while breathing room air or a ratio of arterial oxygen partial pressure to fractional inspired O2 of <300 mm Hg and radiologically confirmed pneumonia.

Updated analyses include the final analysis from the ACTT-1 and the interim analysis of the SOLIDARITY trial [32, 142]. SOLIDARITY reported mortality among persons remaining in hospital up to the duration of the study; however, among patients discharged before the end of the study, mortality may not have been collected completely. The study by Wang et al (2020) was stopped early due to lack of recruitment into the trial due to decreased incidence in China.

Randomization performed in Goldman 2020 failed to establish prognostic balance between baseline clinical status among the 397 patients randomized into the treatment arms, with patients in the 10-day arm more severely ill at study entry. Even with the adjusted analysis, residual confounding is possible. In addition, participants, healthcare workers, and outcome assessors were not blinded to the treatment arms.

Hospitalized patients on invasive ventilation and/or ECMO

Subgroups from SOLIDARITY and ACTT-1 reported on the outcomes of mortality, time to recovery and serious adverse events among patients on invasive ventilation or ECMO [32, 142] (Table 13b). The duration of ventilation at time of treatment with remdesivir was not reported in ACTT-1. This may introduce uncertainty when assessing outcomes of mortality or time to recovery.

In ACTT-1 [142], randomization was stratified by study site and disease severity at enrollment. Disease severity groups were mild to moderate COVID-19 (SpO2 >94%) and severe COVID-19 (SpO2 ≤94%). The severe COVID-19 stratum included patients who were hypoxemic with various degrees of severity including those requiring low flow oxygen by nasal cannula, those needing high-flow oxygen, non-invasive ventilation, invasive mechanical ventilation and ECMO. In addition to analyses on established strata, authors performed post hoc analyses for subgroups within the strata (e.g., receiving oxygen, receiving high-flow oxygen or noninvasive mechanical ventilation, or receiving mechanical ventilation or ECMO), which may introduce concerns with risk of bias and imprecision when making inferences on efficacy of remdesivir among these subgroups including mechanically ventilated patients.

Benefits

Ambulatory patients with mild to moderate disease who are at high risk for progression to severe COVID-19

Treatment with remdesivir for three days in ambulatory patients reduced hospitalizations and COVID-19-related medically attended visits throughout day 28 (HR: 0.28; 95% CI: 0.1, 0.75, low CoE; and HR: 0.19; 95% CI: 0.07, 0.56, low CoE, respectively). No deaths were observed.

Hospitalized patients with oxygen saturation >94% without supplemental oxygen

Treatment with a five- or ten-day course of remdesivir failed to show or to exclude a reduction in mortality when compared with no remdesivir (RR: 0.69; 95% CI: 0.36, 1.34; very low CoE). A five-day course of remdesivir may increase clinical improvement over no remdesivir (RR: 1.16; 95% CI: 1.00, 1.34; very low CoE) but a 10-day course of remdesivir was not associated with improved clinical status as compared with no remdesivir. Patients with mild to moderate disease receiving treatment with remdesivir had similar median time to recovery (median 5 vs. 5 days; Rate ratio: 1.22; 95% CI: 0.82, 1.81; very low CoE).

Hospitalized patients with SpO2 ≤94% on room air

The pooled analysis failed to show a mortality benefit at 28 days (RR: 0.92; 95% CI: 0.77, 1.10; low CoE) [32, 142, 144]. Patients receiving treatment with remdesivir trend toward greater clinical improvement at 28 days than patients not receiving remdesivir (RR: 1.13; 95% CI: 0.91, 1.41; low CoE) [144]. In addition, based on a post hoc analysis of patients with severe COVID-19, receiving treatment with remdesivir had a shorter median time to recovery (median 11 vs. 18 days; rate ratio: 1.31; 95% CI: 1.12, 1.52; low CoE) and decreased need for mechanical ventilation (RR: 0.57; 95% CI: 0.42, 0.79; moderate CoE) [142].

In the study by Goldman et al that compared five and ten days of treatment, the shorter course of remdesivir showed a trend toward decreased mortality (RR: 0.75; 95% CI: 0.51, 1.12; low CoE) and increased clinical improvement at 14 days (RR: 1.19; 95% CI: 1.01, 1.40; low CoE); however, the evidence is uncertain because the persons in the 10-day group had more severe disease at baseline and there is the possibility of residual confounding despite the adjusted analysis [145].

Hospitalized patients on invasive ventilation and/or ECMO

Treatment with remdesivir failed to show a reduction in mortality (RR: 1.23; 95% CI: 0.99, 1.53; low CoE). Similarly, remdesivir failed to show or exclude a reduction in time to recovery among patients on invasive ventilation and/or ECMO (HR: 0.98; 95% CI: 0.70, 1.36; very low CoE).

Harms

Ambulatory patients with mild to moderate disease who are at high risk for progression to severe COVID-19

As with other remdesivir studies published so far, three days of remdesivir infusions did not appear to be associated with a greater risk of serious adverse events compared to no remdesivir (RR: 0.27; 95% CI: 0.1, 0.7; moderate CoE).

Hospitalized patients with oxygen saturation >94% without supplemental oxygen

Patients treated with five days of remdesivir do not appear to experience greater serious adverse events than those not receiving remdesivir (RR: 0.64; 95% CI: 0.31, 1.31; very low CoE).

Hospitalized patients with SpO2 ≤94% on room air

Patients treated with remdesivir do not appear to experience greater serious adverse events (grade 3/4) than those not receiving remdesivir (RR: 0.87; 95% CI: 0.59, 1.28; moderate CoE) [142, 144].

Patients receiving five days of remdesivir may experience fewer serious adverse events and adverse events leading to treatment discontinuation than patients receiving 10 days of remdesivir (RR: 0.61; 0.44, 0.85; low CoE and RR: 0.44; 95% CI: 0.21, 0.95; low CoE, respectively); however, this evidence is uncertain because of the increased severity of disease among patients in the 10-day arm [145].

Hospitalized patients on invasive ventilation and/or ECMO

Patients on invasive ventilation and/or ECMO treated with remdesivir do not appear to experience greater serious adverse events than those not receiving remdesivir (RR: 0.79; 95% CI: 0.54, 1.16; moderate CoE).

Other considerations

Ambulatory patients with mild to moderate disease who are at high risk for progression to severe COVID-19

The panel agreed that the overall certainty of evidence for the treatment of ambulatory patients was low due to concerns about imprecision, as less than half of the original projected sample size was enrolled leading to few events and fragility of the effect estimate. However, compared to prior trials, giving remdesivir early in the course of the viral infection appears to have a robust effect within the limitation of a limited sample size. The panel agreed that benefits are likely to outweigh any potential harms in patients with COVID-19 who are at high risk for severe disease. The evidence confirms that using remdesivir early in the disease process when viral loads are high confers maximum benefit. It is critical to make a rapid diagnosis and treat ambulatory patients with COVID-19 early in the disease course.

Hospitalized patients with oxygen saturation >94% without supplemental oxygen

The panel agreed that the overall certainty of the evidence for treatment of patients with an oxygen saturation >94% with remdesivir compared to no remdesivir was very low due to concerns with study limitations and imprecision. Because of the study limitations and the relatively small effect of remdesivir in patients with moderate COVID-19, the panel suggests remdesivir not be used routinely in these patients. There is a need for more rigorous trials to assess the benefits and harms of remdesivir in patients with moderate COVID-19.

Hospitalized patients with SpO2 ≤94% on room air

The panel agreed that the overall certainty of the evidence for treatment of persons with severe disease with remdesivir compared to no remdesivir treatment was moderate due to concerns with imprecision. Given the inconsistent definition used in the evidence to describe baseline severity, the panel recognized a knowledge gap when assessing whether greater benefit could be attained for patients with oxygen saturation >94% and no supplemental oxygen; however, they agreed that the reported data supported the prioritization of remdesivir among persons with severe but not critical COVID-19.

The panel agreed on the overall certainty of the evidence for treatment with a five-day course compared to a 10-day course of treatment as low due to concerns with risk of bias and imprecision. The panel recognized the benefit of a shorter course of treatment, if providing similar or greater efficacy, on the availability of remdesivir. However, in a subgroup analysis of mechanically ventilated patients, the duration of treatment was 10 days in ACCT-1 trial; therefore, the panel recognized that a longer course of treatment could be desirable in this population.

Hospitalized patients on invasive ventilation and/or ECMO

The panel agreed on the overall certainty of the evidence for treatment of patients on invasive ventilation and/or ECMO with remdesivir as very low due to concerns with risk of bias and imprecision. The panel recognized that the estimates of effect for mortality and time to recovery exclude almost any benefit.

Pediatric use

The evidence for the use of remdesivir in children is limited. For ambulatory children at risk for severe disease, the RCT included 8 children age 12 to 18 years limiting our confidence in the available direct evidence for ambulatory care.

There are no randomized controlled data assessing efficacy of remdesivir for treatment of hospitalized pediatric patients with COVID-19. A report of 77 children who received remdesivir through compassionate use early in the pandemic found good tolerability in this population with a low rate of serious adverse events [146].

An ongoing study of remdesivir in children [147] is using 5 mg/kg on day one (maximum dose 200 mg) followed by 2.5 mg/kg daily in patients over 14 days of age, gestational age more than 37 weeks, and weight greater than or equal to 2.5 kg. The FDA EUA applies to patients weighing over 3.5 kg and applies to the lyophilized powder formulation only.

Conclusions and research needs for this recommendation

The guideline panel suggests remdesivir for ambulatory patients with mild to moderate disease who are at high risk for severe COVID-19.

The guideline panel suggests against remdesivir for routine treatment of patients with oxygen saturation >94% and no supplemental oxygen; however, strongly urges continued study through recruitment into RCTs.

The guideline panel suggests remdesivir rather than no remdesivir for treatment of severe COVID-19 in hospitalized patients with SpO2 <94% on room air. However, the guideline panel suggests against the routine initiation of remdesivir among patients on invasive ventilation and/or ECMO. Additional clinical trials are needed to provide increased certainty about the potential for both benefit and harms of treatment with remdesivir, as well as to understand the benefit of treatment based on disease severity.

Prescribing information in the United States recommends against use of remdesivir in patients with estimated glomerular filtration rate less than 30 mL per minute. This recommendation arises from concern about accumulation of the excipient (betadex sulfobutyl ether sodium) in such patients with potential for hepatic and renal toxicity due to that substance. Additional research into safety of remdesivir in patients with reduced renal function is needed to ascertain whether this concern is substantiated.

Immunocompromised patients who are unable to control viral replication may still benefit from remdesivir despite SpO2 that exceeds 94% on room air or a requirement for mechanical ventilation. Management of immunocompromised patients with uncontrolled viral replication is a knowledge gap and additional research into such populations is needed.

In addition, research is needed to address gaps in the evidence of effectiveness of remdesivir based on viral load.

 

Table 12.png

 

Table 13a.png

 

 

Table 13b.png

 

Table 14.png

 

Table 15.png

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendation 16: Famotidine

Section last reviewed and updated 6/22/2020

Last literature search conducted 6/18/2020

Recommendation 16: Among hospitalized patients with severe COVID-19, the IDSA panel suggests against famotidine use for the sole purpose of treating COVID-19 outside of the context of a clinical trial. (Conditional recommendation, very low certainty of evidence)

The last literature search was conducted on June 18, 2020 and we identified one non-randomized study in OVID. There were no new non-indexed RCTs available.

 

Why is famotidine considered for treatment?

Anecdotal reports from China suggest that patients infected with coronavirus who were receiving famotidine, a H2 receptor antagonist to treat conditions such as acid reflux and peptic ulcer disease, had improved survival versus those receiving proton pump inhibitors (PPIs) [148]. This post hoc finding summarized below has led to interest in the drug, though no predominant theory describing a mechanism for its efficacy yet exists. One theory is that famotidine, like many other compounds, binds and therefore inhibits the coronavirus main protease, 3C-like main protease (3CLpro) [149].

Summary of the evidence

Our search identified one cohort study that compared 84 patients treated with famotidine against 1,536 patients not receiving treatment with famotidine [150] (Table 16). Fifteen percent of patients in the famotidine group (13/84) started famotidine at home before presenting to the hospital. In addition, a subset of 420 patients not treated with famotidine were matched on baseline characteristics to the treated patients.

Benefits

Famotidine may decrease the composite outcome of death or intubation (HR: 0.42; 95% CI: 0.21, 0.85; very low CoE); however, the evidence is very uncertain (Table 16).

Harms

Famotidine is well tolerated. Common adverse events include diarrhea or constipation but occur in less than 5% of people. Severe adverse events occur in less than 1% of persons taking famotidine.

Other considerations

The panel determined that the certainty of evidence to be very low due to concerns with risk of bias, imprecision, and possible publication bias. The panel agreed that critically ill patients (i.e., mechanically ventilated) may have been more likely to receive PPIs than famotidine, thus potentially allocating more prognostically favorable patients to the famotidine group; however, the study did not report a protective effect associated with the use of PPIs.

Conclusions and research needs for this recommendation

The guideline panel suggests against famotidine for the sole purpose of treating COVID-19, unless in the context of a clinical trial. Additional clinical trials are needed to inform research for treatment with famotidine for patients with COVID-19 (Supplementary Table s2).

 

Table 16.png

 

Supplementary Information

Study characteristics:

Risk of bias:

Recommendation 17-18: Neutralizing Antibodies for Pre-Exposure and Post-Exposure Prophylaxis

Section last reviewed and updated 12/23/2021

Last literature search conducted 11/30/2021

Resources:

Recommendation 17 (NEW): In moderately or severely immunocompromised individuals* at increased risk for inadequate immune response to COVID-19 vaccine or for whom COVID-19 vaccine is not recommended due to a documented serious adverse reaction to the vaccine, the IDSA guideline panel suggests pre-exposure prophylaxis with tixagevimab/cilgavimab rather than no tixagevimab/cilgavimab. (Conditional recommendation, Low certainty of evidence)

*See Figure 2 below

 

Figure 02.PNG

 

 

Figure 03.PNG

 

 

Recommendation 18: In persons exposed to COVID-19 who are at high risk of progression to severe COVID-19, the IDSA guideline panel suggests post-exposure casirivimab/imdevimab rather than no casirivimab/imdevimab. (Conditional recommendation, Low certainty of evidence)

Remarks:

  • Dosing for casirivimab/imdevimab is casirivimab 600 mg & imdevimab 600 mg IV or SC once.
  • In the trial considered for this recommendation, participants were enrolled within 96 hours after a household contact received a diagnosis of SARS-CoV-2 infection.
  • Local SARS-CoV-2 variant susceptibility should be considered.

 

Figure 04.png

 

Why are neutralizing antibodies considered for prophylaxis?

Neutralizing antibodies directed at the receptor-binding domain of SARS-CoV-2 spike protein have been evaluated as prophylactic agents for COVID-19. In animal models there is evidence that antibody therapy may more rapidly reduce viral load in the upper and lower airways of infected animals resulting in reduced viral-induced pathology, demonstrating in vivo prophylactic and treatment efficacy  [151, 152]. Additionally, antibody mediated enhancement of disease has not been detected in animal models [152] but this potential phenomenon should be closely monitored in the future studies. In a large, randomized study of unvaccinated nursing home patients and staff where there was at least one confirmed case of COVID-19 at the facility, a single dose of either bamlanivimab appeared to significantly reduce the incidence of “mild or worse” COVID-19 amongst the nursing home residents [153].

Potential advantages of neutralizing antibodies include the ability to standardize the amount of neutralizing activity and the possibility of conferring protection more rapidly than with vaccine-induced immune responses (which generally take several weeks).

Summary of the evidence

Tixagevimab/cilgavimab

Our search identified one RCT reporting on pre-exposure prophylaxis (PrEP) with a single dose of intramuscular tixagevimab/cilgavimab administration in adults ≥18 years of age who are at increased risk of inadequate response to COVID-19 vaccination or SARS-CoV-2 infection [154, 155]. Patients included were those that were either age ≥60 years, immunocompromised, had severe renal or liver impairment, COPD, or those who had an increased risk of exposure including those working in healthcare or living in congregate living settings.  All participants had a negative SARS-CoV-2 serology test result at screening, had no history of SARS-CoV-2 infection, and had not received vaccine or biologic indicated for prevention of SARS-CoV-2 or COVID-19. Study participants received a single combined 300 mg intramuscular dose of the combination of tixagevimab (150 mg) /cilgavimab (150 mg).

Casirivimab/imdevimab

Our search identified one RCT reporting on post-exposure prophylaxis (PEP) with neutralizing antibodies (combination of casirivimab/imdevimab) for patients exposed to COVID-19 who are at high risk of progression to severe disease [156] (Table 18).

One RCT reported on 1,505 persons testing negative for SARS-CoV-2 infection (by reverse-transcriptase-quantitative polymerase-chain-reaction assay [RT-qPCR]) within 96 hours following household contact with a diagnosis of SARS-CoV-2 infection [156]. Of those included in the trial, 30.5% participants were categorized as having a high risk of COVID-19 (e.g., ≥65 years of age, body mass index [BMI] ≥35, chronic kidney disease, etc.). Participants in the treatment group received a total dose of 1200 mg of casirivimab/imdevimab subcutaneously.

Benefits

Tixagevimab/cilgavimab

PrEP with tixagevimab/cilgavimab appears to have little or no effect on mortality through a median of 6 months (RR: 0.50; 95% CI: 0.13, 2.0; absolute risk reduction: 1 fewer per 1,000 [from 2 fewer to 2 more]; moderate CoE). Symptomatic COVID-19 infection within six months after administration was reduced in those who received tixagevimab/cilgavimab compared to placebo (RR: 0.18; 95% CI: 0.09, 0.35; moderate CoE).

Casirivimab/imdevimab

Persons receiving PEP with casirivimab/imdevimab demonstrated an 81% relative risk reduction in development of symptomatic SARS-CoV-2 infection (RR: 0.19; 95% CI: 0.10, 0.35; moderate CoE). Of the 70 persons who developed symptomatic infection, those who received casirivimab/imdevimab rather than placebo experienced a shorter duration of symptoms (mean difference [MD]: -2.0 weeks; 95% CI: -2.91, -1.09; low CoE).

Harms

Tixagevimab/cilgavimab

Serious adverse events were not meaningfully different in those that received PrEP with tixagevimab/cilgavimab compared to placebo (RR: 1.09; 95% CI: 0.67, 1.78; moderate CoE).

Casirivimab/imdevimab

Serious treatment-emergent adverse events may be less frequent among persons receiving casirivimab/imdevimab compared to those receiving placebo; however, this may not be meaningfully different from those receiving placebo (RR: 0.66; 95% CI: 0.30, 1.47; low CoE).

Other considerations

Tixagevimab/cilgavimab

The panel agreed that the overall certainty of evidence for PrEP with tixagevimab/cilgavimab was low due to concerns with the generalizability of the trial population to the FDA-authorized indications (e.g., immunocompromised persons) and low number of events (fragility of results). The panel noted concerns with feasibility at different centers given the large number of potentially eligible individuals and supply constraints.

Casirivimab/imdevimab

The panel agreed that the overall certainty of evidence for post-exposure prophylaxis with casirivimab/imdevimab was low due to low number of events (fragility of results). The panel notes some indirectness between the trial participants (30.5% with any high-risk factor for COVID) and the current approved indications for post-exposure prophylaxis within the EUA.

Conclusions and research needs for this recommendation

Tixagevimab/cilgavimab

The guideline panel suggests PrEP with tixagevimab/cilgavimab in moderately or severely immunocompromised individuals at increased risk for inadequate immune response to COVID-19 vaccine or for whom COVID-19 vaccine is not recommended due to documented severe reactions to the COVID-19 vaccine. Data on the efficacy of pre-exposure prophylaxis specifically in immunocompromised individuals who have received COVID-19 vaccines are needed.

Casirivimab/imdevimab        

The guideline panel suggests PEP using casirivimab/imdevimab in persons exposed to COVID-19, who are at high risk of progression.

 

Table 17.png

 

Table 18.png

 

Supplementary Information

Study characteristics:

Risk of bias:

Recommendations 19-20: Neutralizing antibodies for treatment

Section last reviewed and updated 9/19/2021

Last literature search conducted 7/31/2021

Resources:

Recommendation 19: Among ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease, the IDSA guideline panel suggests bamlanivimab/etesevimab, casirivimab/imdevimab, or sotrovimab rather than no neutralizing antibody treatment. (Conditional recommendation, Moderate certainty of evidence)

  • Remarks:
    • Dosing for casirivimab/imdevimab is casirivimab 600 mg and imdevimab 600 mg IV. Subcutaneous injection is a reasonable alternative in patients for whom it cannot be given intravenously.
    • Dosing for sotrovimab is sotrovimab 500 IV once.
    • Dosing for bamlanivimab/etesevimab is bamlanivimab 700 mg and etesevimab 1400 mg IV.
    • Patients with mild to moderate COVID-19 who are at high risk of progression to severe disease admitted to the hospital for reasons other than COVID-19 may also receive bamlanivimab/etesevimab, casirivimab/imdevimab, or sotrovimab.
    • Local variant susceptibility should be considered in the choice of the most appropriate neutralizing antibody therapy. Local availability of different monoclonal antibody combinations may be affected by predominance of local variants.
    • There are limited data on efficacy of bamlanivimab/etesevimab, casirivimab/imdevimab, or sotrovimab in high-risk patients under 18 years of age.

Recommendation 20: Among hospitalized patients with severe COVID-19, the IDSA guideline panel recommends against bamlanivimab monotherapy. (Strong recommendation, Moderate certainty of evidence)

 

Figure 05.png

 

Why are neutralizing antibodies considered for treatment?

Neutralizing antibodies directed at the receptor-binding domain of SARS-CoV-2 spike protein have been evaluated as therapeutic agents for COVID-19. In animal models there is evidence that antibody therapy may more rapidly reduce viral load in the upper and lower airways of infected animals resulting in reduced viral-induced pathology [151, 152]. Additionally, antibody mediated enhancement of disease has not been detected in animal models [152] but this potential phenomenon should be closely monitored in the future studies.

Potential advantages of neutralizing antibodies include the ability to standardize the amount of neutralizing activity and the possibility of conferring protection more rapidly than with vaccine-induced immune responses (which generally take several weeks).

Antibody treatments have been and continue to be evaluated in both hospitalized and ambulatory patients. For outpatients, logistical challenges exist since the infrastructure for administration of IV infusions does not exist in most ambulatory care settings. There may also be concerns about spread of contagion when administering IV infusions in clinics. However, these challenges are being addressed in a number of outpatient infusion centers and availability of subcutaneous, or intramuscular administration options.

Summary of the evidence

Our search identified six publications of five RCTs reporting on treatment with neutralizing antibodies (bamlanivimab, combination of casirivimab/imdevimab, combination of bamlanivimab/etesevimab, or sotrovimab) for patients with COVID-19 [157-162] (Tables 19-21). Due to clinical heterogeneity of the outcome measures across studies, meta-analyses combining the different neutralizing antibodies were not considered appropriate.

One RCT, stopped early for futility, reported on hospitalized patients with COVID-19 randomized to treatment with either a single infusion of bamlanivimab (7000 mg) or placebo (ACTIV-3/TICO) [158]. One phase II/III RCT reported on non-hospitalized patients (adults as well as children age 12 and up) considered at high risk for progression to severe disease who were within three days of their first positive test for SARS-CoV-2 who were randomized to a single infusion of bamlanivimab 2800 mg/etesevimab 2800 mg or placebo [159]. One phase II RCT reported on non-hospitalized patients with recently diagnosed mild or moderate COVID-19 randomized to treatment with either a single infusion of neutralizing antibody bamlanivimab in one of three doses (700 mg, 2800 mg, or 7000 mg) or placebo [157].

One phase III RCT assessed a single infusion of either 1200 mg or 2400 mg of casirivimab/imdevimab in non-hospitalized participants with mild to moderate COVID-19 [161]. In the original phase of this trial, participants without risk factors for severe disease were included; however, 1,040 participants were removed after randomization and not analyzed as they had no risk factors for severe disease. In the amended phase of this investigation all participants were considered at high risk for severe disease. Another phase III RCT also reported on non-hospitalized participants with mild to moderate COVID-19 who were at risk for severe disease [160]. Participants in this study received a single infusion of sotrovimab 500 mg. Unlike previous studies, this study did exclude participants with immunocompromising conditions.

Benefits

Bamlanivimab/etesevimab

In ambulatory persons at high risk for severe COVID-19, bamlanivimab/etesevimab demonstrated an absolute mortality reduction of 1.9% (95% CI includes a minimum of 0.7% reduction in mortality) as no deaths were seen by day 29 in the 518 persons treated with bamlanivimab/etesevimab compared to 10 deaths in the 517 persons who received placebo. However, due to the small number of events (10, of which nine were believed to the result of COVID-19), the certainty of evidence was low due to imprecision. Bamlanivimab/etesevimab demonstrated a lower relative risk of COVID-19 related hospitalizations (defined as ≥24 hours of acute care) through day 29 compared to no bamlanivimab/etesevimab (RR: 0.30; 95% CI: 0.16, 0.59; low CoE). Ambulatory persons who received bamlanivimab/etesevimab had a lower relative risk of persistently high viral load at day seven compared to no bamlanivimab/etesevimab (RR: 0.34; 95% CI: 0.25-0.46; low CoE).

Casirivimab/imdevimab

Concerns were raised by the panel whether bias could have been introduced by excluding 1040 persons post-randomization (2400-mg dose group) due to lack of risk factors for severe disease. Therefore, the panel used the amended phase (1200-mg dose) full data set to inform the effect estimates as no exclusions were reported. Sensitivity analyses were carried out to test the robustness of this approach by either adding the 2400-mg to the 1200-mg dose data set or by formally pooling both effect estimates using fixed effects model; these sensitivity analyses resulted in little to no relevant differences in the findings. In addition, the amended phase lower dose (1200 mg) results also served as confirmation that the latest EUA recommended dosing appears to be equally effective as the previously authorized higher dose.

Among ambulatory persons with at least one risk factor for severe disease, there was no difference in 29-day mortality in persons treated with casirivimab/imdevimab compared to no casirivimab/imdevimab 1200 mg (RR: 1.02; 95% CI: 0.06, 16.20; low CoE). However, there was a lower relative risk of hospitalization in persons treated with casirivimab/imdevimab 1200 mg (RR: 0.27; CI: 0.11, 0.65; moderate CoE).

Sotrovimab

Among ambulatory persons with at least one risk factor for severe disease, sotrovimab demonstrated a lower relative risk of mortality compared to no sotrovimab (RR: 0.33; 95% CI: 0.01-8.19, low CoE). The low certainty of evidence was due to imprecision as there were no mortality events in those who received sotrovimab and one death in the placebo arm. Among ambulatory persons, sotrovimab use was associated with a lower relative risk of hospitalization, compared to no sotrovimab (RR: 0.14; 95% CI: 0.04-0.48; moderate CoE). Persons receiving sotrovimab had a lower progression to severe or critical disease compared to no sotrovimab (RR: 0.11; 95% CI: 0.02, 0.45; moderate CoE).

Bamlanivimab monotherapy

[NOTE: On April 16, 2021, FDA revoked EUA for monoclonal antibody bamlanivimab.] [163]

Among ambulatory persons, bamlanivimab demonstrated a lower relative risk of hospitalization, including visits to the emergency room, compared to no bamlanivimab (RR: 0.26; 95% CI: 0.09, 0.75; very low CoE). The very low certainty of evidence was due to indirectness, as the treatment may not have been provided to enough persons at risk of developing severe disease to be representative of the general population, and imprecision, due to few events recorded. Bamlanivimab may increase viral clearance at three days (MD: -0.49; 95% CI: -0.87, -0.11; low CoE); however, there may not be a meaningful difference at 11 days as measured by change from baseline SARS-CoV-2 viral load (MD: -0.22; 0.95: -0.60, 0.15; low CoE).

Among patients hospitalized for COVID-19, treatment with bamlanivimab compared to placebo failed to show or exclude a beneficial effect on mortality (HR: 2.00; 95% CI: 0.67, 5.99; moderate CoE). Clinical improvement, as defined as a decrease in a pulmonary ordinal scale, may not be meaningfully different among patients hospitalized for COVID-19 who received treatment with bamlanivimab or placebo (OR: 0.85; 0.56, 1.29; moderate CoE).

Harms

Bamlanivimab/etesevimab

Persons receiving bamlanivimab/etesevimab experienced more serious adverse events. However, this may not be meaningfully different from those receiving placebo (RR: 1.40; 95% CI: 0.45, 4.37; moderate CoE).

Casirivimab/imdevimab

Serious adverse events were less frequent among persons receiving casirivimab/imdevimab compared to those receiving placebo (RR: 0.34; 95% CI: 0.24, 0.48; moderate CoE).

Sotrovimab

Persons who received sotrovimab were less likely to experience serious adverse events compared to those receiving placebo (RR: 0.27; 95% CI: 0.12-0.63; moderate CoE).

Bamlanivimab monotherapy

Serious adverse events among ambulatory persons receiving bamlanivimab monotherapy may not be meaningfully different from those receiving placebo (RR: 0.15; 95% CI: 0.01, 3.78; low CoE). Persons receiving bamlanivimab did experience more infusion-related adverse events, including pruritus, flushing, rash, and facial swelling (RR: 1.62; 95% CI: 0.34, 7.70; low CoE).

Similarly, serious adverse events at five and 28 days among patients hospitalized for COVID-19 receiving bamlanivimab may not be meaningfully different from those receiving placebo (RR: 1.85; 95% CI: 0.34, 9.97; moderate CoE and RR: 0.93, 95% CI: 0.27, 3.14; moderate CoE, respectively). Similarly, infusion-related adverse events may not be meaningfully different between patients hospitalized for COVID-19 receiving bamlanivimab or placebo (OR: 1.64, 95% CI: 0.79, 3.44; moderate CoE).

Other considerations

Neutralizing antibodies for ambulatory persons

The panel agreed that the overall certainty of evidence for the treatment with bamlanivimab/etesevimab, casirivimab/imdevimab, and sotrovimab in ambulatory persons with COVID-19 at high risk for progression to severe disease (at least one risk factor) was moderate due to mostly low number of events (fragility of results). The results were driven by the number of avoided hospitalizations, as the number of deaths that occurred were too sparse to show a clear trend. Neutralizing antibodies were well tolerated, and serious adverse events were comparable or lower than placebo. The panel noted increased feasibility with the option of providing treatment with casirivimab/imdevimab through subcutaneous injections [156, 164].

​Casirivimab/imdevimab has been evaluated for the treatment of COVID-19 at doses of 1200 mg, 2400 mg, and 8000 mg. Across all treatment doses, there was a flat dose-response relationship for viral load and clinical outcomes. As part of the FDA Emergency Use Authorization, the use of casirivimab/imdevimab as an IV infusion is strongly recommended, however the subcutaneous route is authorized as an alternate route when IV infusion is not feasible and would result in a delay in treatment. Clinical outcomes of patients receiving casirivimab/imdevimab via the subcutaneous route for the treatment of COVID-19 have not been reported in available trials. A pre-print manuscript [164] evaluated early casirivimab/imdevimab 1200 mg versus placebo in asymptomatic outpatients with COVID-19 and demonstrated less hospitalizations in those receiving casirivimab/imdevimab compared to those receiving placebo, 0/100 versus 3/104, respectively (RR: 0.15; 95%CI: 0.01-2.84). Peak pharmacokinetic levels in those receiving subcutaneous casirivimab 600 mg/imdevimab 600 mg appear approximately 75% lower than after IV infusion [165].

Bamlanivimab monotherapy

The panel agreed that the overall certainty of evidence for treatment with bamlanivimab for ambulatory persons with COVID-19 is very low due to concerns with indirectness and imprecision.

The panel agreed that the overall certainty of evidence for treatment with bamlanivimab for patients hospitalized for COVID-19 is moderate due to concerns with fragility in the estimate from the small number of events reported. The guideline panel made a strong recommendation against treatment with bamlanivimab for patients hospitalized for COVID-19. The panel was moderately certain that any relevant benefit (reduction in mortality or clinical improvement) could be excluded.

SARS-CoV-2 variants and neutralizing monoclonal antibodies

The emergence and circulation of new SARS-CoV-2 genetic variants has been reported from the United States and other counties. The B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma), B.1.427/B.1.429 (epsilon) and B.1.617.2 (delta) variants circulating in the United States are classified as variants of concern as they may have potential clinical and public health implications. The B.1.525 (eta), B.1.526 (iota), B.1.526.1, B.1.617, B.1.617.1 (kappa), B.1.617.3 and P.2 (zeta) variants are classified as variants of interest [166]. In vitro neutralizing assays using SARS-CoV-2 or vesicular stomatitis virus-based pseudovirus showed that some of the variants had reduced susceptibility to neutralizing antibodies, either individually or in combination. There is limited data from clinical studies.

Bamlanivimab alone and the combination of bamlanivimab and etesevimab together had activity against pseudovirus expressing del69-70 + N501Y found in the B.1.1.7 variant (alpha). Pseudovirus expressing spike protein from the B.1.351 lineage (beta) or substitutions K417N + E484K + N501Y found in this lineage had reduced susceptibility to bamlanivimab and etesevimab together of >45-fold, and pseudovirus expressing K417T + E484K + N501Y found in the P.1 lineage (gamma) had reduced susceptibility to bamlanivimab and etesevimab together of >511-fold. Pseudovirus expressing spike protein from the B.1.427/B.1.429 lineages (epsilon), or the L452R substitution found in this lineage, had reduced susceptibility to bamlanivimab and etesevimab together of 7.7-fold or 7.4-fold, respectively [167]. In vitro neutralization studies showed that bamlanivimab lost activity against the delta variant, but etesevimab retained activity [168].

Casirivimab and imdevimab individually and together had neutralization activity against pseudovirus expressing all spike protein substitutions found in the B.1.1.7 lineage (alpha) and against pseudovirus expressing only N501Y found in B.1.1.7 (alpha) and other circulating lineages. Casirivimab and imdevimab together had neutralization activity against pseudovirus expressing all spike protein substitutions, or individual substitutions K417N, E484K or N501Y, found in the B.1.1351 lineage (beta), and against K417T+E484K, found in the P.1 lineage (gamma), although casirivimab alone, but not imdevimab, had reduced activity against pseudovirus expressing K417N or E484K, as indicated above. The E484K substitution is also found in the B.1.526 (iota) lineage. Casirivimab and imdevimab, individually and together, retained neutralization activity against the L452R substitution found in the B.1.427/B.1.429 lineages (epsilon) [165]. In in vitro neutralization studies, casirivimab and imdevimab remained active against the delta variant [168].

Pseudotype virus-like particle neutralization assays indicate that sotrovimab retains activity against the B.1.1.7, B.1.315, P.1, B.1.427/B.1.429, B1.526 & B.1.617 variant spike proteins. There is limited nucleotide sequencing data available from COMET ICE to comment on the clinical impact of variants on therapeutic response [169].

We have limited data on how in vitro neutralization activity of monoclonal antibodies against pseudovirus expressing spike protein substitutions or even in vitro neutralization activity against the SARS-CoV-2 variants correlates with clinical efficacy. Genotypic and phenotypic testing for variants and their correlation with patient important outcomes is being studied in clinical trials evaluating neutralizing antibodies. We still need further studies and surveillance data to understand the implications of SARS-CoV-2 variants on clinical efficacy of COVID-19 therapies.

Conclusions and research needs for this recommendation

The guideline panel suggests using bamlanivimab/etesevimab, casirivimab/imdevimab, or sotrovimab in mild to moderate COVID-19 ambulatory persons at high risk for developing severe disease as the expected benefits likely outweigh any potential harms (Tables 19-21).

The guideline panel recommends against use of bamlanivimab for patients hospitalized for COVID-19 (Table 23).

The guideline panel recognized the need for continued research and accrual of evidence, particularly trials on patient important outcomes (hospitalizations progressing to need for ventilation, or death), existing and new neutralizing antibodies, and outcomes with variants of concern  (Supplementary Table s2).

 

Table 19.png

 

Table 20.png

 

Figure 06a.png

 

Figure 06b.png

 

Figure 06c.png

 

Table 21.png

 

Table 22.png

 

Table 23.png

 

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendations 21-23: Janus kinase inhibitors (baricitinib and tofacitinib)

Bariticinib

Section last reviewed and updated 10/11/2021

Last literature search conducted 9/30/2021

Recommendation 21: Among hospitalized adults with severe* COVID-19 having elevated inflammatory markers, the IDSA panel suggests baricitinib rather than no baricitinib. (Conditional recommendation, Moderate certainty of evidence)

Remarks:

  • Baricitinib 4 mg per day (or appropriate renal dosing) up to 14 days or until discharge from hospital.
  • Baricitinib appears to demonstrate the most benefit in those with severe COVID-19 on high-flow oxygen/non-invasive ventilation at baseline.
  • Limited additional data suggest a mortality reduction even among patients requiring mechanical ventilation.
  • Patients who receive baricitinib for treatment of COVID-19 should not receive tocilizumab or other IL-6 inhibitors.

Recommendation 22: Among hospitalized patients with severe* COVID-19 who cannot receive a corticosteroid (which is standard of care) because of a contraindication, the IDSA guideline panel suggests use of baricitinib with remdesivir rather than remdesivir alone. (Conditional recommendation, Low certainty of evidence)

  • Remark: Baricitinib 4 mg daily dose for 14 days or until hospital discharge. The benefits of baricitinib plus remdesivir for persons on mechanical ventilation are uncertain.

*Severe illness is defined as patients with SpO2 ≤94% on room air, including patients on supplemental oxygen, oxygen through a high-flow device, or non-invasive ventilation.

 

Why is baricitinib considered for treatment?

Baricitinib, a selective Janus kinase 1 and 2 (JAK1 and JAK2, respectively) inhibitor currently FDA-approved for the treatment of RA, is being investigated in multiple studies for treatment of COVID-19. The proposed benefits of baricitinib in the management of COVID-19 may be two-fold as it has both anti-inflammatory and potential antiviral activity [170]. Janus kinase (JAK) mediates cytokine signaling, which contributes to inflammation; JAK inhibitors, therefore, may decrease cytokine-mediated inflammation. Baricitinib inhibits host intracellular membrane proteins AP2-associated protein kinase 1 (AAK1) and also binds cyclin G-associated kinase (GAK), both thought to play a role in receptor mediated endocytosis of many viruses including Ebola, dengue, hepatitis C, and SARS-CoV-2 [171-173]. Baricitinib has been evaluated in people with COVID-19 in both randomized and non-randomized studies [174-178].

Based on experience in clinical trials for RA, baricitinib has been associated with an increased risk of adverse effects including infections (especially upper respiratory tract infections), thrombosis, lymphopenia, anemia, increases in lipids, elevations in liver enzymes, and elevations in creatinine phosphokinase [170]. In clinical trials for RA, baricitinib was associated with a numerically higher risk of upper respiratory tract infections and herpes simplex and herpes zoster infections compared with placebo  [179]. Opportunistic infections such as herpes simplex, herpes zoster, and tuberculosis [180, 181] have been reported in patients taking baricitinib. Many of these side effects appear to be dose related, with increased incidence in patients taking baricitinib 4 mg compared with 2 mg. Patients enrolled in Adaptive COVID-19 Treatment Trial (ACTT-2) and COV-BARRIER received baricitinib 4 mg daily for two weeks or until discharge, a shorter duration than those taking the drug for RA.

Patients with COVID-19 have been found to have abnormalities in coagulation parameters and might have an elevated risk of thrombosis [182]. Baricitinib receipt was associated with an increased incidence of thrombosis when compared with placebo receipt in clinical trials for its FDA approval for RA, especially at a higher dose of 4 mg daily [170]. During the 16-week treatment period in RA trials, venous thromboembolism (VTE) occurred in five patients treated with baricitinib 4 mg daily, compared with zero in the 2 mg daily and placebo groups.  Arterial thrombosis occurred in two patients treated with baricitinib 4 mg, two patients treated with baricitinib 2 mg, and one patient on placebo. In ACTT-2, the percentage of patients reported to have VTE was numerically higher in the combination group (21 patients [4.1%] vs. 16 patients [3.1%]) although it was similar overall (absolute difference 1%, 95% CI -1.3 to 3.3) [183]. Of note, all patients in the trial were recommended to receive VTE prophylaxis if they had no contraindication. We do not have long-term data, especially on safety, development of the aforementioned adverse effects, and opportunistic infections from these two trials.

Summary of the evidence

Baricitinib

Our literature search identified one RCT that compared the use of baricitinib (4 mg daily dose up to 14 days) to placebo in hospitalized adults with severe COVID (NIAID OS: 4 – hospitalized, not requiring supplemental oxygen;  5 – hospitalized, requiring supplemental oxygen; or 6 – hospitalized, receiving non-invasive ventilation or high-flow oxygen devices) [178, 184]. In the COV-BARRIER trial, randomization was stratified by disease severity, age, region, and use of corticosteroids. Participants in both arms had >1 elevated inflammatory marker (CRP, d-dimer, LDH [lactate dehydrogenase], ferritin) and also received standard of care, which included corticosteroids in 79% and/or antivirals (e.g., remdesivir in 18.9%).

An additional presentation of baricitinib treatment for critically ill (OS-7) patients with COVID-19 pneumonia requiring invasive mechanical ventilation was identified that reported on the outcomes of mortality, need for invasive mechanical ventilation, days of hospitalization, and serious adverse events [185].

Baricitinib without corticosteroids, with remdesivir

Our literature search identified one RCT that reported on the use of baricitinib (4 mg daily dose) plus remdesivir in hospitalized patients with moderate and severe COVID-19 ([183]. This trial was conducted as the second stage of the ACTT-2, where subjects were randomized to receive combination therapy with baricitinib and remdesivir or remdesivir alone [183] (Table 26). Randomization was stratified by disease severity classified by an OS of clinical status (4+5 vs 6+7 [7 –patients with an ordinal scale of 6 (high-flow oxygen and non-invasive ventilation) or 7 (mechanical ventilation or ECMO). Mild to moderate disease was defined as patients with an ordinal scale of 4 (hospitalized, but not requiring supplemental oxygen) or 5 (requiring supplemental oxygen). The trial was initiated before corticosteroids were commonly used for severe COVID-19. 

Benefits

Baricitinib

Treatment of hospitalized patients with severe COVID-19 with baricitinib rather than no baricitinib reduced 60-day mortality (HR: 0.62; 95% CI: 0.47 to 0.83; moderate CoE). The odds of COVID-19 disease progression trends toward a reduction in persons receiving treatment with baricitinib (OR: 0.85; 95% CI: 0.67, 1.08; moderate CoE).

Treatment of critically ill hospitalized patients with baricitinib rather than no baricitinib reduced 60-day mortality (HR 0.56; 95% CI: 0.33 to 0.97; low CoE). However, the sample size for this sub-study was only 101 participants.

Baricitinib without corticosteroids, with remdesivir

In ACTT-2, the combination of baricitinib and remdesivir showed a trend towards lower mortality (4.7% vs. 7.1%; rate ratio: 0.65; 95% CI 0.39, 1.09; moderate CoE). In patients stratified within the severe COVID-19 pneumonia group, defined as 6 or 7 on the ordinal scale, subjects who received baricitinib and remdesivir were more likely to experience clinical recovery (defined as a value of <4 on the ordinal scale) at day 28 (69.3% vs. 59.7%; rate ratio 1.29; 95% CI 1.00, 1.66; moderate CoE). The original stratification was altered as 40 subjects were misclassified at baseline; however, re-analysis of the original stratified data produced a similar result. Patients in the baricitinib arm were less likely to require initiation of mechanical ventilation or ECMO through day 29 (10% vs. 15.2%; RR: 0.66; 95% CI 0.46, 0.93; low CoE). In summary, it appeared that patients requiring supplemental oxygen or non-invasive ventilation at baseline benefitted most from baricitinib; the benefit was less clear in patients already on mechanical ventilation.

Harms

The risk of serious adverse events in hospitalized patients with severe or critical COVID-19 receiving baricitinib was not greater than those not receiving baricitinib (RR: 0.82; 95% CI: 0.65, 1.03; moderate CoE and RR 0.70; 95% CI: 0.50 to 0.97, moderate CoE, respectively).

In ACTT-2, patients receiving baricitinib and remdesivir had a lower risk of developing any serious adverse events through day 28 (16% vs. 21%; RR 0.76; 95% CI 0.59, 0.99; moderate CoE) whether or not thought to be related to the study drug. In this trial, the overall rate of new infections was lower in the baricitinib plus remdesivir group compared with remdesivir alone (30 patients [5.9%] versus 57 patients [11.2%]) [183]. However, patients who received concomitant glucocorticoids had a higher incidence of serious or non-serious infections as compared with those who did not: 25.1% and 5.5%, respectively. It was not specified what proportion of these patients in the study were in the baricitinib combination group versus the control group.

Other considerations

Baricitinib

The panel agreed on the overall certainty of evidence as moderate due to concerns with imprecision, as some outcomes have concerns with fragility. The guideline panel recognized the resource implications based on the dose and duration reported in the trial (4 mg daily up to 14 days). Additional data from a small study of hospitalized patients with critical COVID-19 suggest consistent benefits; however, shares concerns with imprecision based on a small sample. Based on that the panel broadened the population in the recommendation to hospitalized adults with severe COVID-19 having elevated inflammatory markers, including those on mechanical ventilation.

Baricitinib without corticosteroids

The panel agreed that the overall certainty of evidence was low due to concerns with risk of bias, driven by the use of data from post hoc analyses and imprecision, which recognized the limited events and concerns with fragility in the group who likely benefited most (those requiring supplemental oxygen or non-invasive ventilation). The guideline panel noted the importance of suggesting baricitinib plus remdesivir as an option for persons unable to receive corticosteroids.

Conclusions and research needs for this recommendation

The guideline panel suggests baricitinib in addition to standard of care for patients hospitalized with severe COVID-19. The guideline panel suggests baricitinib with remdesivir for persons for whom corticosteroids are indicated but who cannot receive them due to a contraindication. Baricitinib plus remdesivir should be reserved for patients who cannot take corticosteroids because dexamethasone has been proven to reduce mortality in patients hospitalized with COVID-19 who require supplemental oxygen or mechanical ventilation and, for this reason, dexamethasone is recommended by the panel for this group. It is uncertain whether baricitinib plus remdesivir will have the same benefit as dexamethasone. As of the time of this narrative, there are no head-to-head trials evaluating either the combination of baricitinib plus tocilizumab or evaluating baricitinib compared to tocilizumab. Patients who received JAK inhibitors should not receive tocilizumab or other immunomodulators as no adequate evidence is available for its combined use.

Table 24.png

 

Table 25.png

 

Table 26.png

Supplementary Information

Study characteristics:

Risk of bias:


Tofacitinib

Section last reviewed and updated 8/21/2021

Last literature search conducted 7/31/2021

Recommendation 23: Among hospitalized adults with severe* COVID-19, but not on non-invasive or invasive mechanical ventilation, the IDSA panel suggests tofacitinib rather than no tofacitinib. (Conditional recommendation, Low certainty of evidence)

  • Remarks:
    • Tofacitinib appears to demonstrate the most benefit in those with severe COVID-19 on supplemental or high-flow oxygen.
    • Patients treated with tofacitinib should be on at least prophylactic dose anticoagulant.
    • Patients who receive tofacitinib should not receive tocilizumab or other IL-6 inhibitor for treatment of COVID-19.
    • The STOP-COVID Trial did not include immunocompromised patients.

*Severe illness is defined as patients with SpO2 ≤94% on room air, including patients on supplemental oxygen or oxygen through a high-flow device.

 

Why is tofacitinib considered for treatment?

Tofacitinib is a JAK inhibitor that preferentially inhibits JAK-1 and JAK-3 though it is active on all other JAK isoforms. It is FDA-approved for moderate to severe RA, active psoriatic arthritis, and moderate to severe ulcerative colitis. Like baricitinib, it is expected that JAK inhibition leads to downstream suppression of cytokine production, thereby modulating the inflammatory cascade that results in systemic inflammation in patients with severe COVID-19. See baricitinib section (above) for additional rationale on considerations for treatment.

Summary of the evidence

Our literature search identified one RCT that compared the use of tofacitinib 10 mg every 12 hours for up to 14 days or placebo [186]. Patients included were those who had laboratory-confirmed SARS-CoV-2 infection and evidence of COVID-19 pneumonia on imaging and who were hospitalized for less than 72 hours. Patients in this study could not be receiving non-invasive ventilation, mechanical ventilation, or ECMO at baseline. Additionally, patients with a history of or current thrombosis, personal or first-degree family history of blood clotting disorders, immunosuppression, any active cancer, or those with certain cytopenias were excluded from this trial. Patients who received other potent immunosuppressants, or other biologic agents were excluded, while the use of glucocorticoids for the management of COVID-19 was permitted. A composite outcome of death at day 28 or respiratory failure (defined as progression to NIAID ordinal scale 6, 7, or 8) was the primary outcome.

Benefits

Treatment of hospitalized patients with COVID-19 pneumonia with tofacitinib resulted in a lower risk of the composite outcome of death or respiratory failure compared to no tofacitinib (RR: 0.63; 95% CI: 0.41, 0.97; low CoE). However, results failed to show or to exclude a beneficial or detrimental effect on mortality alone (RR: 0.49; 95% CI: 0.15, 1.63; low CoE) or progression to mechanical ventilation or ECMO by day 28 (RR: 0.25; 95% CI: 0.03, 2.20; low CoE).

Harms

Patients who received tofacitinib experienced more serious adverse events; however, this may not be meaningfully different from those that received placebo (RR: 1.18; 95%CI: 0.64, 2.15; low CoE). Use of tofacitinib for other indications has shown an increase in thrombotic events which prompted a black box warning by the FDA [187, 188]. As COVID-19 infection itself increases the risk for VTE events; it is important to note that the patients studied were either on prophylactic or full dose anticoagulation during treatment with tofacitinib.

Tofacitinib carries four black boxed warnings for its labeled indications including a warning for 1) serious infections including tuberculosis, invasive fungal infections, bacterial, viral and other opportunistic pathogens; 2) mortality; 3) thrombosis; and 4) lymphoma and other malignancies, including an increased rate of EBV-mediated post-transplant lymphoproliferative disorder [187-190].

Other considerations

The panel agreed that the overall certainty of evidence was low due to concerns of imprecision, which recognized the limited number of events and concerns about fragility of the results in the group who likely would benefit the most (those requiring supplemental oxygen or oxygen through a high-flow device).

Conclusions and research needs for this recommendation

The guideline panel suggests tofacitinib in addition to standard of care for patient hospitalized for severe COVID-19. Due to the increased risk of VTE with treatment with tofacitinib, patients should receive at least prophylactic doses of anticoagulants during their hospital stay. Patients who received JAK inhibitors should not receive tocilizumab or other immunomodulators as no adequate evidence is available for its combined use.

 

Table 27.png

 

Supplementary Information

Study characteristics:

Risk of bias:

Recommendations 24-25: Ivermectin

Section last reviewed and updated 8/10/2021

Last literature search conducted 7/31/2021

Recommendation 24: In hospitalized patients with COVID-19, the IDSA panel suggests against ivermectin outside of the context of a clinical trial. (Conditional recommendation, very low certainty of evidence)

Recommendation 25: In ambulatory persons with COVID-19, the IDSA panel suggests against ivermectin outside of the context of a clinical trial. (Conditional recommendation, very low certainty of evidence)

 

Why is ivermectin considered for treatment?

Ivermectin is an anti-parasitic agent that is FDA-approved for onchocerciasis and strongyloidiasis and is used off-label for the treatment of many parasitic infections. Although it has in vitro activity against some viruses, including SARS-CoV-2, it has no proven therapeutic utility. In vitro activity against SARS-CoV-2 [191] requires concentrations considerably higher than those achieved in human plasma and lung tissue to reach the in vitro IC50 [192]. Ivermectin has been shown to have anti-inflammatory effects in in vitro and in vivo studies hence hypothesized to have a mechanism beyond its anti-viral effects in the treatment of COVID-19  [193, 194].

Since ivermectin is generally well tolerated, it was empirically evaluated in uncontrolled studies for COVID-19, alone and in combination with other off-label medications.

Summary of the evidence

Our search identified 15 studies in patients with COVID-19 with ages ranging between 8 and 86 years that reported on the outcomes of mortality, symptom resolution, viral clearance, and adverse events, and informed the evidence review for inpatient and outpatient therapy [195-212]. Eligible studies compared treatment with ivermectin against a placebo or standard of care. Studies comparing ivermectin to a non-placebo, active comparison (i.e., a different agent considered a possible treatment for COVID-19 infection by clinicians) or that did not provide a comparison arm were not included in these analyses. Several studies did not meet eligibility for inclusion in this review. Four trials compared ivermectin to hydroxychloroquine (comparison to treatment with evidence of harm) [213-215]; two trials examined ivermectin as prophylactic treatment [216, 217]; and three trials did not provide study data in a peer-reviewed, published or pre-print manuscript [215, 218, 219].

The studies that informed the recommendations for hospitalized patients included 10 RCTs [195-197, 199, 200, 205-209] and two non-randomized studies [198, 201]. Eight RCTs [197, 199, 202-204, 210-212] informed the recommendation for ambulatory persons. Each of them compared an active treatment arm of ivermectin to an inactive comparison (e.g., standard of care with or without placebo).

The evidence informing the recommendations for treating hospitalized and ambulatory persons with ivermectin reported on the use of a range of doses (100 mcg/kg/day to 400 mcg/kg/day) and durations (one day up to seven days). Among studies reporting on hospitalized patients, substantial heterogeneity was observed, introduced by one study (Supplementary Figure s9c) [195]. Ahmed 2021 treated patients with ivermectin for a duration of five days, rather than one day as used by the remaining studies. This may explain the heterogeneity between studies; however, excluding Ahmed 2021, any meaningful reduction in viral clearance was still not demonstrated by the summary estimate (Supplementary Figure s9d). Heterogeneity was not observed for other outcomes reported for hospitalized or ambulatory persons.

Among the RCTs, the risk of bias was high in two trials because of unsuccessful randomization into treatment and control groups. Hashim et al (2020) [199] inadequately randomized participants by allocating them to respective treatment arms on odd and even days, as well as assigning all critically ill patients to the ivermectin arm, and Podder et al (2020) [200] allocated participants based on odd or even registration numbers. In addition, across many RCTs, there were concerns due to lack of blinding of study personnel, which may lead to over- or under-estimates of treatment effects, particularly for subjective outcomes (e.g., symptom resolution, adverse events).

Benefits

Inpatients

The evidence from RCTs failed to show a reduction or increase in mortality among persons with COVID-19 (RR: 0.66; 95% CI: 0.31, 1.42; low CoE). In addition, the evidence from non-randomized studies cannot exclude no meaningful reduction in mortality among persons treated with ivermectin with COVID-19 (RR: 0.60; 95% CI: 0.37, 0.97, very low CoE). Persons receiving treatment with ivermectin rather than no ivermectin failed to demonstrate a beneficial or detrimental effect on symptom resolution or viral clearance at day seven (RR: 1.07; 95% CI: 0.69, 1.65; very low CoE and RR: 1.25; 95% CI: 0.72, 2.15; very low CoE, respectively).

Outpatients

The evidence is very uncertain, but ivermectin may reduce the time to recovery among outpatients with COVID-19 (mean difference: 3.46 days fewer; 95% CI: 5.40 to 1.52 days fewer; very low CoE). However, treatment with ivermectin failed to demonstrate a beneficial or detrimental effect on mortality, avoidance of progression to severe disease, or viral clearance at day seven (RR: 0.48; 95% CI: 0.13, 1.76; very low CoE, RR: 0.64; 95% CI: 0.26, 1.54; very low CoE, and RR: 1.13; 95% CI: 0.79, 1.62; very low CoE, respectively).

Harms

In doses typically used for the treatment of parasitic infections, ivermectin is well tolerated. We are unable to exclude the potential for adverse events in hospitalized and serious adverse events in non-hospitalized persons with COVID-19 treated with ivermectin rather than no ivermectin, (RR: 0.80; 95% CI: 0.39, 1.64; low CoE and RR: 0.99; 95% CI: 0.14, 6.96; low CoE, respectively).

Other considerations

The panel determined the certainty of evidence of treatment of ivermectin for hospitalized and non-hospitalized patients to be very low due to concerns with risk of bias (i.e., study limitations) and imprecision. In addition, there were concerns about publication bias, as the available evidence consisted mostly of positive trials of smaller size. The guideline panel made a conditional recommendation against treatment of COVID-19 with ivermectin outside of the context of a clinical trial for both patients with COVID-19 hospitalized or in the outpatient setting.

Conclusions and research needs for this recommendation

The guideline panel suggests against ivermectin for the treatment of hospitalized patients with COVID-19, unless in the context of a clinical trial. The guideline panel suggests against ivermectin for the treatment of outpatients with COVID-19, unless in the context of a clinical trial. Well-designed, adequately powered, and well-executed clinical trials are needed to inform decisions on treating COVID-19 with ivermectin (Supplementary Table s2).

Table 28.png

 

Table 29.png

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendation 26: Fluvoxamine

Section last reviewed and updated 11/8/2021

Last literature search conducted 10/31/2021

Recommendation 26: Among ambulatory patients with COVID-19, the IDSA guideline panel recommends fluvoxamine only in the context of a clinical trial. (Knowledge gap)

 

Why is fluvoxamine considered for treatment?

Fluvoxamine is a selective serotonin reuptake inhibitor (SSRI) which is currently FDA-approved for the treatment of obsessive-compulsive disorder. SSRIs have been shown to have affinity for Sigma-1 receptors, which have been demonstrated to modulate cytokine levels in animal models of septic shock [220]. Additionally, pharmacologic agents that act at Sigma-1 receptors have demonstrated in vitro activity against SARS-CoV-2 [221]. Amongst the SSRIs, fluvoxamine has been shown to have the high affinity for these receptors making it a potential repurposed drug option for the management of COVID-19 [222]. SSRIs like fluvoxamine may decrease uptake of serotonin from platelets during thrombosis, resulting in decreased neutrophil recruitment and platelet aggregation, which may be helpful in the early stages of COVID-19 [223, 224].

Summary of the evidence

Our search identified two RCTs that reported on ambulatory patients with SARS-CoV-2 infection [225, 226]. Patients in these studies were randomized to fluvoxamine or placebo/usual care. Both trials included symptomatic outpatients who tested positive for SARS-CoV-2 infection within seven days. Reis included patients who were at high risk for severe infection and utilized a composite primary outcome of hospitalization or emergency room visit lasting greater than six hours [226]. Additional outcomes reported in the two trials included mortality, hospitalization, emergency room visit lasting >6 hours, progression to oxygen saturation <92%, viral clearance, and serious adverse events.

Benefits

Outpatients

Among symptomatic ambulatory patients with COVID-19, fluvoxamine failed to demonstrate or to exclude a beneficial effect on mortality at 28 days compared to no fluvoxamine (RR: 0.69; 95% CI: 0.38, 1.27; low CoE). Fluvoxamine showed a reduction of the composite outcome of hospitalizations, emergency room visits lasting >6 hours, or oxygen saturation <92% (RR: 0.64; 0.50, 0.84; low CoE). When evaluating the effect on hospitalizations only, there was a trend toward less hospitalizations in fluvoxamine treated patients compared to those not receiving fluvoxamine (RR: 0.75; 95% CI: 0.57, 0.99; low CoE). Treatment with fluvoxamine failed to show a benefit in viral clearance at day seven (RR: 0.74; 0.52, 1.05; very low CoE).

Harms

The risk of serious adverse events in patients receiving fluvoxamine was not greater than those not receiving fluvoxamine (RR: 0.81; 95% CI: 0.59, 1.12; low CoE).

Other considerations

The panel agreed on the overall low certainty of evidence given the sparseness in mortality data and because upper boundary of the 95% confidence interval failed to exclude the risk of possible harms. The panel also had concerns about the generalizability/indirectness in the results surrounding hospitalization and emergency room visit >6 hours as one study [226] was partially conducted in patients with extended stays in emergency settings (mobile hospitals) to inform the primary endpoint, and it is unclear if resource constraints (possible contingency setting) may have affected the total number of events (i.e., emergency room stays and rates of hospitalization).

Conclusions and research needs for this recommendation

The guideline panel recommends fluvoxamine only in the context of a clinical trial to better delineate the effects of fluvoxamine on disease progression, such as need for hospital admission, ICU care, and ultimately, mortality.

Table 30.png

 

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Recommendation 27: Nirmatrelvir/ritonavir

Section last reviewed and updated 12/29/2021 

Last literature search conducted 12/28/2021

Resources:

Recommendation 27 (NEW): In ambulatory patients with mild to moderate COVID-19 at high risk for progression to severe disease, the IDSA guideline panel suggests nirmatrelvir/ritonavir initiated within five days of symptom onset rather than no nirmatrelvir/ritonavir. (Conditional recommendation, Low certainty of evidence)

Remarks:

  • Patients’ medications need to be screened for serious drug interactions (i.e., medication reconciliation). Patients on ritonavir- or cobicistat-containing HIV or HCV regimens should continue their treatment as indicated.
  • Dosing based on renal function:
    • Estimated glomerular filtration rate (eGFR) > 60 ml/min: 300 mg nirmatrelvir/100 ritonavir every 12 hours for five days
    • eGFR ≤60 and ≥30 mL/min: 150 mg nirmatrelvir/100 mg ritonavir every 12 hours for five days
    • eGFR <30 mL/min: not recommended
  • Patients with mild to moderate COVID-19 who are at high risk of progression to severe disease admitted to the hospital for reasons other than COVID-19 may also receive nirmatrelvir/ritonavir
  • Options for treatment and management of ambulatory patients include nirmatrelvir/ritonavir, three-day treatment with remdesivir, molnupiravir, and neutralizing monoclonal antibodies. Patient-specific factors (e.g., symptom duration, renal function, drug interactions) as well as product availability should drive decision-making regarding choice of agent. Data for combination treatment do not exist in this setting.

 

Figure 07.png

 

Why is nirmatrelvir/ritonavir considered for treatment?

Nirmatrelvir is an inhibitor to the main protease (Mpro) of SARS-CoV-2; inhibition of this enzyme blocks viral replication. Nirmatrelvir is a substrate of the cytochrome P450 3A4 isoenzyme system and is co-packaged with an HIV-1 protease inhibitor, ritonavir, a potent inhibitor of cytochrome P450 3A4. Coadministration results in higher concentrations and a longer half-life of nirmatrelvir, allowing for every 12-hour dosing. The U.S. Food and Drug Administration (FDA) granted emergency use authorization (EUA) to nirmatrelvir/ritonavir on December 22, 2021, for the treatment of mild to moderate COVID-19 in adults and pediatric patients who are at high risk for progression to severe COVID-19, including hospitalization or death [227].

Summary of the evidence

Our search identified one RCT reporting on treatment of mild to moderate COVID-19 in patients at high risk for progression to severe disease [227]. Data have not yet been published, but data to prepare this recommendation was extracted from the FDA EUA document.

Benefits

Nirmatrelvir/ritonavir

All-cause mortality through day 28 may be lower in patients receiving nirmatrelvir/ritonavir compared to no nirmatrelvir/ritonavir (RR: 0.04; 95% CI: 0.00, 0.69; low CoE). Patients treated with nirmatrelvir/ritonavir rather than no nirmatrelvir/ritonavir may have fewer COVID-19-related hospitalizations (RR: 0.12; 95% CI: 0.06, 0.26; low CoE). The composite endpoint of COVID-19-related hospitalizations or mortality was lower in patients receiving nirmatrelvir/ritonavir compared to no nirmatrelvir/ritonavir (RR: 0.12; 95% CI: 0.06, 0.25; low CoE).

Harms

Nirmatrelvir/ritonavir

Serious treatment-emergent adverse events were not reported in the FDA EUA.

Given co-formulation with ritonavir as a pharmacokinetic booster, there is potential for significant drug interactions. Contraindications exist between agents that can have their levels increased or decreased by nirmatrelvir and/or ritonavir and agents that can speed up the metabolism of the components of nirmatrelvir and/or ritonavir resulting in a loss of virologic response and possible resistance. These drug interactions can result in treatment failure or serious adverse events, which may lead to severe, life-threatening, or fatal events from greater exposures (i.e., higher levels) of concomitant medications. See Figures 8 and 9.

 

Figure 08.png

 

Figure 09.png

 

Other considerations

Nirmatrelvir/ritonavir

The panel agreed that the overall certainty of the evidence for the treatment of ambulatory patients was low; there are concerns with the inability to exclude potential risks to bias because of limited availability of study details within the EUA, and there is imprecision due to a low number of events reported. The EUA did not report safety data (e.g., adverse events or severe adverse events) from the trial. The panel agreed that the benefits are likely to outweigh any potential harms in patients with COVID-19 who are at high risk of severe disease; however, recognized concerns with drug interactions must be considered.

The evidence confirms that using nirmatrelvir/ritonavir early in the disease process when viral loads are high confers maximum benefit. It is critical to make a rapid diagnosis and treat ambulatory patients with COVID-19 early in the disease course.

Conclusions and research needs for this recommendation

Nirmatrelvir/ritonavir

The guideline panel suggests the use of nirmatrelvir/ritonavir for ambulatory patients with mild-to-moderate COVID-19 at high risk for progression to severe disease who are within five days of symptom onset. More data are needed on the potential adverse effects of this medication.

 

Table 31.png

 

Supplementary Information

Study characteristics:

Risk of bias:

Recommendation 28: Molnupiravir

Section last reviewed and updated 12/28/2021

Last literature search conducted 12/28/2021

Recommendation 28 (NEW): In ambulatory patients (≥18 years) with mild to moderate COVID-19 at high risk for progression to severe disease who have no other treatment options*, the IDSA guideline panel suggests molnupiravir initiated within five days of symptom onset rather than no molnupiravir. (Conditional recommendation, Low certainty of evidence)

Remarks:

  • Patients who put a higher value on the putative mutagenesis, adverse events or reproductive concerns, and a lower value on the uncertain benefits, would reasonably decline molnupiravir.
  • Molnupiravir 800 mg for five days.
  • Patients with mild to moderate COVID-19 who are at high risk of progression to severe disease admitted to the hospital for reasons other than COVID-19 may also receive molnupiravir.
  • Molnupiravir is not authorized under the FDA EUA for use in patients <18 years, because it may affect bone and cartilage growth.
  • Molnupiravir is not recommended under the FDA EUA for use during pregnancy.
  • Molnupiravir is not authorized under the FDA EUA for pre-exposure or post-exposure prevention of COVID-19 or for initiation of treatment in patients hospitalized due to COVID-19, because benefit of treatment has not been observed in individuals when treatment is started after hospitalization due to COVID-19.

*Other options for treatment and management of ambulatory patients include nirmatrelvir/ritonavir, three-day treatment with remdesivir, and neutralizing monoclonal antibodies. Patient-specific factors (e.g., symptom duration, renal function, drug interactions) as well as product availability should drive decision-making regarding choice of agent. Data for combination treatment do not exist in this setting.

 

Figure 10.png

 

Why is molnupiravir considered for treatment?

Molnupiravir is an oral antiviral that targets the genetic machinery that is responsible for SARS COV-2 replication. Molnupiravir is an oral pro-drug that is converted to β-D-N4-hydroxycytidine (NHC) which acts as a substrate for RNA-dependent RNA polymerase. After it is incorporated into the viral RNA, serial mutations develop, resulting in a virus that is less fit for ongoing viral replication. One phase 1 RCT evaluated the safety and tolerability of molnupiravir in healthy adults without COVID-19 [229]. The study reported molnupiravir to be well tolerated, with no increased reports of serious adverse events among persons in the molnupiravir arm compared to those receiving placebo. The FDA granted EUA to molnupiravir on December 23, 2021, for the treatment of mild to moderate COVID-19 in adults (≥18 years) who are at high risk for progression to severe COVID-19, including hospitalization or death.

Summary of the evidence

Two RCTs reported on treatment of unvaccinated patients with COVID-19 with either 800 mg of molnupiravir or placebo for five days [230, 231]. In one phase III trial (MOVe-OUT trial) reporting on the outcomes of death, hospitalization and serious adverse events, patients with mild to moderate COVID-19 received either molnupiravir or placebo within five days after the onset of symptoms. In the phase IIa trial reporting on the outcomes of death and serious adverse events in patients with symptom duration <7 days received molnupiravir or placebo.

Benefits

COVID-19-related mortality may be lower in patients receiving molnupiravir rather than placebo (RR: 0.11; 95% CI: 0.01, 0.86; low CoE). Similarly, COVID-19-related hospitalizations and the composite of all-cause hospitalization or death may trend towards a reduction among patients receiving molnupiravir rather than no molnupiravir (RR: 0.68; 95% CI: 0.48, 1.00; low CoE and HR: 0.69; 95% CI: 0.48, 1.01; low CoE, respectively).

Harms

Patients treated with molnupiravir may not experience greater serious adverse events than those receiving placebo (RR: 0.43; 95% CI: 0.17, 1.11; low CoE).

Based on findings from animal reproduction studies, molnupiravir may cause fetal harm when administered to pregnant individuals [232]. Other concerns with molnupiravir include the possibility of  viral mutagenesis in persons with compromised immune systems who are unable to clear the virus. Females of childbearing potential should be counseled to use a reliable method of contraception during treatment and for four days after the last dose. Men of reproductive potential who are sexually active with females of childbearing potential should be counseled to use a reliable method of contraception during treatment and for at least three months after the last dose of molnupiravir. It is also not recommended in children <18 years of age for the concern of bone growth.

Molnupiravir does not require renal or hepatic dose adjustment.

Other considerations

The panel agreed that the overall certainty of evidence for treatment of ambulatory patients was low, given concerns with imprecision, driven by few reported events and a relatively small effect.

The use of molnupiravir presents additional considerations and potential concerns regarding viral mutagenesis in immunocompromised persons and safety in persons of reproductive age, for which more data are needed to quantify such effects. The panel recognized that alternative treatment options exist with the possibility of greater benefit with a smaller known safety profile. The FDA required the manufacturers to conduct additional animal studies on the impact of the drug on spermatogenesis and to establish a pregnancy registry if the drug was inadvertently administered during pregnancy.

The evidence confirms that using molnupiravir early in the disease process when viral loads are high confers maximum benefit. It is critical to make a rapid diagnosis and treat ambulatory patients with COVID-19 early in the disease course.

Conclusions

The guideline panel suggests the use of molnupiravir for ambulatory patients with mild-to-moderate COVID-19 at high risk for progression to severe disease who are within five days of symptom onset and have no other treatment options. More data are needed on the potential adverse effects of this medication. The evidence supporting this recommendation will be reassessed with the release of updated published information from the MOVe-OUT study and other trials.

 

Table 32.png

 

Supplementary Information

Study characteristics:

Forest plots:

Risk of bias:

Narrative summaries of treatments undergoing evaluation

In addition to the clinical questions addressed above, the panel identified several treatments currently undergoing evaluation for which additional data are needed to rate recommendations. Narrative summaries for these treatments are provided below.

HIV antivirals

Last reviewed 4/11/2020; no updates made since 4/11/2020

In vitro antiviral activity of darunavir against SARS-CoV-2 showed no activity at clinically relevant concentrations. Three randomized, open-label clinical trials are currently listed on evaluating darunavir/cobicistat as a potential therapeutic option for COVID-19. Janssen, the manufacturer of darunavir/cobicistat has reported that one of these trials [233] has concluded that darunavir/cobicistat plus conventional treatments was not effective in achieving viral clearance at day seven post randomization, compared to conventional treatments alone. Clinical outcomes of this trial including rate of critical illness and mortality 14 days after randomization, have not been reported to date.

Lopinavir-ritonavir combined with interferon beta or other antivirals

Last reviewed and updated 9/4/2020

Lopinavir-ritonavir is a combination of protease inhibitors for the treatment of HIV infection. Lopinavir-ritonavir has been shown to have in vitro antiviral activity against beta-coronaviruses such as SARS-CoV, and MERS-CoV [62, 64, 65, 234]. Since lopinavir-ritonavir is not specifically designed for treatment of coronavirus, lopinavir-ritonavir alone may not demonstrate a difference from placebo in reducing viral load when treatment was initiated at a median of 13 days after symptoms onset [65]. In an open-label treatment trial, lopinavir-ritonavir with ribavirin reduced the mortality and requirement of intensive care support of hospitalized SARS patients compared with historical control [65]. Many interferons, especially interferon beta have been shown to have modest in-vitro antiviral activity against SARS-CoV and MERS-CoV [62, 234]. Lopinavir-ritonavir or interferon beta-1b has been shown to reduce viral load of MERS-CoV and improve lung pathology in a nonhuman primate model of common marmoset [64].

An RCT on the triple combination of lopinavir-ritonavir, ribavirin, and interferon beta-1b, compared with single agent lopinavir-ritonavir for 14 days was conducted in the treatment of 127 adult patients admitted to hospital with COVID-19 [68]. Patients who had NEWS2 of least one, and with symptom duration of 14 days or less were recruited and randomly assigned to either triple combination or control group in a ratio of 2:1. Treatment with triple combination was well tolerated and had a significantly shorter median time to suppress the viral load in nasopharyngeal specimen, and a significantly shorter time to alleviate symptoms, and resulted in shorter hospital stay. Since the median number of days from symptom onset to the start of study treatment was five days, only one patient in the control group received ventilator support and no patient died during the study. It is not possible to generalize the effectiveness of triple therapy in critically ill patients.

Lopinavir-ritonavir was further investigated in two retrospective cohort studies using HCQ [235] and arbidol [236], an indole-derivative licensed for decades in Russia and China against influenza, for comparison. Lopinavir-ritonavir was associated with more rapid viral clearance (median, 21 days vs. 28 days) than HCQ in 65 mild to moderate COVID-19 patients in South Korea, but there was no difference in time to clinical improvement [235]. Lopinavir-ritonavir was found to be inferior to arbidol in terms of viral clearance on day 14 after admission. But the number of patients was small (n=50) and all patients received atomized inhalation of recombinant human interferon-α2b injection. The efficacy of arbidol monotherapy remains uncertain [236].

Subcutaneous injection of interferon β-1a was used for the treatment of 42 severe COVID-19 adult patients in an open-label randomized clinical trial in Iran. Although there was no significant improvement in time to clinical response in the interferon-treated group, the overall mortality at 28 days was reduced in the interferon-treated then the control group (19% vs. 43.6%, p= 0.015) [237].

COVID-19 convalescent plasma for prophylaxis

Last reviewed and updated 9/4/2020

Studies of convalescent plasma for treatment of hospitalized patients with COVID-19 were discussed in a previous section. Use of convalescent plasma as prophylaxis in individuals with high-risk exposure to SARS-CoV-2 is under study, with at least five clinical trials in clinicaltrials.gov as of August 6, 2020 that include arms in which individuals exposed to SARS-CoV-2 but without disease may receive convalescent plasma [238-242]. Issues associated with regulatory concerns, safety, workflow, and trial design were recently reviewed [243]. Distinct from the polyclonal antibodies present in convalescent plasma, monoclonal antibodies specific for respiratory viruses have also been used in certain populations for protection against disease in specific high-risk populations [244, 245], and animal models have suggested utility in prophylaxis against SARS coronavirus infection [246]. There are multiple trials listed in clinicaltrials.gov of different SARS-CoV-2 monoclonal antibodies for treatment or prophylaxis, with other potential monoclonal antibodies in earlier stages of development. No data on safety or efficacy are yet reported.

Ribavirin

Last reviewed 4/11/2020; no updates made since 4/11/2020

There are only in vitro data available on the activity of ribavirin on SARS-CoV-2 currently. The EC50 (half maximal effective concentrations) was significantly higher than for chloroquine and remdesivir, so it appears less potent in vitro compared to these agents [16]. There are limited clinical studies in SARS-CoV-1 and MERS-CoV infections. In a systematic review of ribavirin treatment in patients infected with SARS-CoV-1, 26 studies were classified as inconclusive, and four showed possible harm [247]. In a retrospective observational study in patients with MERS-CoV infection, the combination of ribavirin and interferon, compared to no antiviral treatment, was not associated with improvement in the 90-day mortality or more rapid MERS-CoV RNA clearance [248].

Oseltamivir

Last reviewed 4/11/2020; no updates made since 4/11/2020

Oseltamivir is a neuraminidase inhibitor used for prophylaxis and treatment of influenza. Given its specificity for an enzyme not found on coronaviruses, it is unclear what the mechanism of action would be against COVID-19. However, this has been used in combinations of antiviral therapy in Wuhan [249] and continues to be explored as a therapeutic option as part of combination regimens. Two trials evaluating combination regimens are underway in Wuhan [250, 251] as well as a trial in Thailand proposing different combinations [252]. None of the trials or case reports have examined oseltamivir as monotherapy.

Intravenous immunoglobulin

Last reviewed and updated 9/4/2020

Intravenous immunoglobulin (IVIg) has been used as an adjuvant to treat a variety of pathogens either as a pooled product or in a concentrated more pathogen focused (hyperimmune) form. As the community from which a given batch of IVIg is derived from includes increasing numbers of individuals who have recovered from SARS-CoV-2, the possibility of protective antibodies being present in the pooled product is increased. However, the potential utility of IVIg for the treatment of SARS-CoV-2 is unknown at this time. Its use has been reported in a few patients with COVID-19 [253], but studies are needed to determine if there may be a role for IVIg in the treatment of SARS-CoV-2.

One open-label trial randomized patients with COVID-19 (SpO2 ≤96% on ≥4 liters O2 by nasal cannula but not on mechanical ventilation) to either three days of IVIg (n=16) or no IVIg (n=17) [254]. During the study period (30 days or hospital discharge), two patients in the IVIg arm and seven in the standard of care arm required mechanical ventilation, one patient in the IVIg arm and three patients in the standard of care arm died. No adverse events were reported in the IVIg arm. Co-treatments with remdesivir, convalescent plasma, and corticosteroids were balanced across arms at baseline; however, methylprednisolone was provided with each IVIg dose in the treatment arm, and co-interventions provided during the treatment period were unbalanced. One retrospective cohort reported on 58 patients who received IVIg; however, the study did not identify a standard of care group and multiple co-treatments were provided [255]. Two case series reported on eight patients [253, 256] with severe COVID-19 who received IVIg for five consecutive days. All patients were discharged from the hospital.

Should NSAIDS be stopped in patients with COVID-19?

Last reviewed and updated 9/4/2020

The role of nonsteroidal anti-inflammatory drugs (NSAIDs) in the management of SARS-CoV-2 was debated widely in the first few months of the COVID-19 pandemic. The discussion was prompted by warnings from European health officials regarding the possibility of increased risk of infection or severity of disease in those taking NSAIDS. These concerns were based on early unconfirmed reports in four patients and supported by theoretical mechanistic concerns about the role NSAIDs play in SARS-CoV-2 pathogenesis. Human coronaviruses, including SARS-CoV-2, use ACE2 to bind to human targets and gain entry into target cells [257]. It has been theorized that NSAIDs, due to upregulation in ACE2 in human target cells, may lead to an increased risk of infection or a more severe course of COVID-19 in those taking NSAIDs. In addition, there are well known risks of non-steroidal anti-inflammatory agents including cardiovascular, gastrointestinal and renal adverse events [258, 259]. In the setting of bacterial pneumonia, NSAIDs may impair recruitment of polymorphonuclear cells, resulting in a delayed inflammatory response and resolution of infection, however a causal relationship has not been established [260, 261].

A case-control study from Italy published in May 2020 did not demonstrate an increased risk of SARS-CoV-2 infection in those taking NSAIDs chronically (adjusted OR: 1.06; 95% CI 0.98, 1.15) [262]. In April 2020, the WHO produced a scientific brief detailing a systematic review that included 73 studies in patients with acute respiratory infections. While no direct studies for patients with MERS, SARS, or SARS-CoV-2 were available for analysis, there was no evidence of adverse events [263]. In a large registry trial that included data from five hospitals in Massachusetts, there was a lower risk of hospitalization in those with SARS-CoV-2 prescribed naproxen or ibuprofen, however it is difficult to determine if these patients were actively taking these medications at the time of COVID-19 diagnosis [264]. Randomized controlled trials are currently underway to better understand the safety of NSAIDs in the management of patients with COVID-19 [265, 266].

Should ACE inhibitors and ARBs for hypertension be stopped in patients with COVID-19?

Last reviewed and updated 9/4/2020

Angiotensin converting enzyme 2 (ACE2) is the entry receptor for SARS-CoV-2 on human cells. Animal experiments have shown mixed findings on the effect of angiotensin-converting enzyme inhibitors (ACEI) and angiotensin receptor blockers (ARBs) on ACE2 levels and activity, leading to two contrasting hypotheses in COVID-19 [267-269]. The harmful hypothesis is that ACEIs and ARBs may increase the risk of infection and severity of COVID-19 via increased ACE2 expression. On the contrary, infection with other coronaviruses have been shown to decrease ACE2 levels in vitro [270], which may lead to increased angiotensin II activity resulting in pulmonary, cardiovascular and other end organ damage in patients with COVID-19 [267, 271]. This has led to speculation about a beneficial hypothesis that ACEI and ARBs may have a therapeutic role in COVID-19, by inhibiting the renin-angiotensin-aldosterone axis.

There have been several recent observational studies on the effects of ACEIs and ARBs in patients tested for and diagnosed with COVID1-19. A multi-center retrospective study [272] evaluated 1,128 patents admitted to nine hospitals in Hubei province, China with COVID-19 including 188 (17%), who were on an ACEI or ARB. The risk of 28-day all-cause mortality was lower in ACEI/ARB group vs. non-ACEI/ARB group (IRD: -0.24; 95% CI: -0.43, -0.05). After adjusting the all-cause mortality was still lower in the ACEI/ARB group compared to the non-ACEI/ARB group (HR: 0.42; 95% CI 0.15, 0.89). Another single center retrospective study [273] among 1178 hospitalized patients with COVID-19, had 362 patients with hypertension and 115 were on ACEI/ARBs. There was no difference between those with severe vs. non-severe illness in use of ACEIs (9.2% vs. 10.1%; P = .80), and ARBs (24.9% vs. 21.2%; P = 0.40). There was also no difference between non-survivors and survivors in use of ACEIs (9.1% vs. 9.8%; P = 0.85) and ARBs (19.5% vs. 23.9%; P = 0.42).

Another study [274] among 1200 COVID-19 patients hospitalized in two hospitals in London, UK observed that chronic ACEI/ARB use was not associated with an increase in severity of COVID-19. Within their cohort of 1200 patients, 399 (33.3%) were on an ACEI/ARB and while unadjusted odds of critical care admission or death within 21 days were not significantly different between patients on ACEI/ARB vs. not (OR 0.83; 95% CI 0.64, 1.07), adjustment for age, sex and co-morbidities presented an OR of 0.63 (95% CI 0.47, 0.84, p<0.01) for the composite outcomes in patients on ACEI/ARB. An observational study from Italy [275] evaluated multiple predictors of in-hospital mortality in 311 patients with hypertension and COVID-19. The patients in this study were significantly older, with a higher BMI, comorbidities, and severity of disease. In a multivariate Cox regression analysis chronic use of ACEI and ARBs (aHR, 0.97; 95% CI: 0.68, 1.39; P = .88) were not associated with an increase in in-hospital mortality. A population-based case-control study [262] from Lombardy, Italy compared 6272 COVID-19 patients with 30,759 controls matched on sex, age, and municipality of residence. In a logistic-regression multivariate analysis, use of ARBs or ACEI did not show an association with COVID-19 among cases (aOR, 0.95, 95% CI 0.86 to 1.05 for ARBs and 0.96, 95% CI, 0.87 to 1.07 for ACEI). It also did not show an association with severe or fatal disease (for ARBs, aOR 0.83; 95% CI 0.63, 1.10; for ACEI, aOR 0.91; 95% CI 0.69, 1.21). Reynolds et al [276] analyzed data available for patients tested for COVID-19, available in the electronic medical records for New York University Langone Health system. In the study, 12,594 patients were tested, 5,894 (46.8%) were positive and 1,002 of these patients (17.0%) had severe illness. They performed propensity score matching and a Bayesian analysis to assess the relationship between various classes of antihypertensives including ACEI and ARBs and the likelihood of a positive COVID-19 test and severe disease. The study did not show a positive association for ACEI and ARBs with having a positive test for SARS-CoV-2 or developing severe infection. A retrospective cohort study using data from Danish national administrative registries, had an unadjusted 30-day mortality of 18.1% in the group with ACEI/ARB use compared to the 7.3% in the nonuser group, but the association was not significant after adjustment for age, sex and medical history (aHR 0.83; 95% CI: 0.67, 1.03). In that study, ACEI/ARB use compared with other antihypertensive agents was not significantly associated with higher incidence of COVID-19 (a HR 1.05 95% CI 0.80–1.36) [277]. One retrospective cohort study done in severe COVID-19 patient's showed ACEI/ARB use, after adjusting for other variables, to be independently associated with elevated creatinine >10.1 mg/L (OR 3.22; 95% CI: 2.28, 4.54). Consistent ACEI/ARB use was independently associated with AKI stage >1 (ALT ratio 3.28; 95% CI: 2.17, 4.94) [278].

Data from these observational studies suggest that ACEI and ARBs do not increase the risk of acquiring COVID-19, developing severe disease or death. One study showed possible increase risk of renal dysfunction in severe COVID-19. There are limitations though inherent to retrospective observational studies, especially differences in unmeasured prognostic factors between the compared groups that might be responsible for the difference in outcomes and not treatment with ACEI or ARBs. Most professional scientific and medical societies have recommended that ACEI or ARBs be continued in people who have an indication for these medications [279-281].

Antibacterials and antifungals

Last reviewed and updated 9/4/2020

Patients with COVID-19 often present to hospitals with viral pneumonia with accompanying febrile illness and respiratory symptoms. Differential diagnoses may include bacterial pneumonia, for which antibiotics are prescribed. Concerns for bacterial superinfections also exist. Studies performed early in the COVID-19 pandemic reported high percentages of antibiotic use in China (58-95%) [1, 249, 282], Spain (74%) [283], and New York (65%) [284]. These studies are not granular and do not report if they describe co-infection at presentation or the development of superinfection, limiting the ability to ascertain the reasons for antibiotic use.

Data reporting co-infection in patients presenting with COVID-19 for care is sparse. Rawson and colleagues reviewed 18 studies of human coronavirus infections reporting co-infections, of which nine were COVID-19 [285]. These cumulatively reported a bacterial and fungal co-infection rate of 8% (62/806). The studies evaluated were heterogeneous. One brief report of 393 patients in New York reported a bacteremia rate of 5.6%, which varied significantly between patients receiving invasive mechanical ventilation (15/126 [11.9%]) and those who were not (4/222 [1.8%]) [286]. Another study looked at 88,201 blood cultures performed during March 2020 in New York, comparing order volume, positivity, and etiologies between patients with COVID-19 and others during the time period [287]. The study found a significantly lower rate of bacteremia in COVID-19 patients (3.8%) than either COVID-19 negative (8%) or untested (7.1%) (p<0.001). When commensal skin organisms were excluded, the positivity rate in COVID-19 patients was 1.6% [287]. A study in Texas reviewed the use of antibiotics and incidence of coinfections in 147 PCR-positive COVID-19 patients [288]. Eighty-seven (59%) patients received empiric antibiotics, though none of the 47 (32%) patients with respiratory cultures had positive results. 112 patients (76%) had blood cultures collected also, and while nine were positive, eight of those were considered contaminants [288].

The apparent discordance between bacterial and fungal co-infection in patients with COVID-19 at presentation and the use of antibacterial therapy has potential negative effects, namely in antimicrobial resistance. Publications report on patients with severe and critical COVID-19 patients treated with immunomodulatory therapies, including corticosteroids, IL-6 antagonists, IL-1 antagonists, and others [289]. In one preprint examining outcomes of in a cohort of 154 patients receiving invasive mechanically ventilation, mortality was reduced in patients treated with tocilizumab (IPTW-adjusted model, HR 0.55; 95% CI 0.33, 0.90); however, superinfections were more commonly reported (54% vs. 26%, p<0.001), primarily due to ventilator-associated pneumonia [111]. Initiating and continuing empiric antibiotics at the time of admission may lead to superinfections that are antibiotic resistant [290].

Favipiravir

Last reviewed and updated 9/4/2020

Favipiravir is a purine analog that inhibits the RNA dependent RNA polymerase of influenza and other RNA viruses [291]. The drug is approved in Japan for treatment of influenza. However, because of its teratogenicity risk, favipiravir should not be given during pregnancy and there are substantial concerns about its use in women in child-bearing potential.

In terms of its potential role in COVID-19, favipiravir has in vitro activity against SARS-CoV-2 [16]. However, it is uncertain whether adequate drug levels can be achieved in vivo to inhibit SARS-CoV-2. There have been small clinical trials with this drug in people with COVID-19. In a non-randomized, open-label study in China [292], oral favipiravir was associated with shorter time to viral clearance and greater improvement in chest imaging than lopinavir/ritonavir (in both groups, the oral antiviral was given with aerosolized alpha-interferon). However, because the study was small and not randomized, it was not possible to conclude that favipiravir is effective in treating COVID-19. A randomized, open-label trial compared favipiravir to umifenovir, an antiviral approved in Russia and China, in people with COVID-19 [293]. The clinical recovery rate at day seven was not significantly different between the two groups. There appeared to be an impact of favipiravir in the sub-group of people who did not have critical illness, but more data are needed. An exploratory clinical trial, also conducted in China, randomized 30 hospitalized adults with COVID-19 into a baloxavir marboxil, favipiravir or control group. There was no apparent effect of favipiravir (or baloxivir) on viral clearance [294]. There are ongoing clinical trials assessing favipiravir for treatment of COVID-19.

Immunomodulatory agents

Last reviewed and updated 9/4/2020

Some patients with COVID-19 develop a hyperinflammatory state that may incorporate elements of cytokine release syndrome seen in conditions such as secondary hemophagocytic lymphohistiocytosis. The etiology is unclear, but patients who develop significantly elevated CRP, ferritin, and D-dimer levels with the syndrome have an increased risk of mortality, associated with respiratory failure, multiorgan dysfunction, and hypercoagulability. Numerous immunomodulatory agents are under investigation to address this immunologic complication.

IL-1 inhibitors: Anakinra is an FDA approved IL-1-beta inhibitor that is currently FDA-approved for rheumatoid arthritis and Neonatal-Onset Multisystem Inflammatory Disease. High- and low-dose anakinra was investigated in a recent retrospective cohort study in Italian patients with COVID-19, moderate to severe ARDS, and hyperinflammation. Patients receiving anakinra were compared to a historical control group with COVID-19 who fulfilled eligibility criteria for anakinra. The low-dose anakinra group was stopped early due to lack of effect. In the high-dose anakinra group, 3/29 (10%) patients died versus 7/16 (44%) in the historical control group, however there was no difference in the rates of mechanical ventilation-free survival [295]. Anakinra is being investigated in numerous trials including this randomized placebo-controlled trial [296]. Canakinumab is another IL-1-beta antagonist with limited human data for COVID-19 that is being studied in a phase III clinical trial [297, 298].

Janus kinase inhibitors: Baricitinib, a JAK inhibitor currently FDA-approved for the treatment of rheumatoid arthritis, is being investigated in multiple studies for COVID-19. The proposed benefits of baricitinib in the management of COVID-19 are two-fold as it has both anti-inflammatory and likely antiviral activity. Janus kinase mediates cytokine signaling which contributes to inflammation, which may reduce risk of the associated hyperinflammatory syndrome and ARDS. Baricitinib inhibits AAK1 and also binds GAK, both thought to play a role in receptor mediated endocytosis of many viruses including SARS-CoV-2 [173]. In an open-label non-randomized study from Italy, baricitinib with lopinavir/ritonavir (n=12) were compared to lopinavir/ritonavir (n=12) alone at one institution over two consecutive time periods. After two weeks in the baricitinib group, no patients required ICU transfer and 7/12 (58%) were discharged. In the lopinavir/ritonavir group, 4/12 (33%) required ICU transfer and only 1/12 patients were discharged by day 14. No serious adverse events or infections occurred in the baricitinib group [174]. In the ACTT-2 trial, baricitinib is being compared to remdesivir and numerous other RCTs are currently underway to better understand the role of baricitinib in the management of COVID-19 [299-303].

GM-CSF inhibitors: Monoclonal antibodies that bind to GM-CSF are under investigation for the treatment of hyperinflammation associated with COVID-19. GM-CSF inhibitors are postulated to disrupt the downstream signaling of pro-inflammatory cytokines. One agent, mavrilimumab was studied in a single center non-randomized cohort study in non-ventilated patients in Italy. Trial participants had SARS-CoV-2 infection with a PaO2: FiO2 ratio <300 mm Hg, pulmonary infiltrates, and evidence of hyperinflammation (CRP >100 mg/L or ferritin >900 µg/L and any increase in LDH). Patients in the treatment group received a single dose of mavrilimumab 6 mg/kg (n=13). A similar cohort managed by the same medical team received no mavrilimumab due to lack of consent and lack of access to mavrilimumab (n=26). Mortality rates were 0/13 in the mavrilimumab group and 7/26 (27%) died in the control group. Median days to clinical improvement (defined as a reduction of two or more points on the seven-point ordinal scale) was 8 (IQR: 5-11) versus 19 (IQR: 11 >28), in the mavrilimumab versus control groups, respectively. Mavrilimumab was well tolerated in all patients [304]. Randomized controlled trials are underway to investigate the role of GM-CSF inhibitors in the management of COVID-19 [305-307].

Complement inhibitors: ln mouse models of both SARS-CoV and MERS-CoV, complement activation has been shown to play a role in the pathogenesis of ARDS. Eculizumab, is a complement inhibitor that is already approved by the FDA for other conditions including myasthenia gravis and paroxysmal nocturnal hemoglobinuria, is currently being studied for the treatment of COVID-19 [308]. Ravulizumab, another complement inhibitor, is also being investigated in randomized trials for COVID-19 [300].

SARS-CoV-2 in children and treatment of multisystem inflammatory syndrome in children (MIS-C)

Last reviewed and updated 9/4/2020

Treatment

Compared with adults, children generally have milder illness from SARS-CoV-2 infection [309, 310]. However, severe illness does occur in children, even those with no predisposing factors [310, 311]. Among children admitted to the hospital for COVID-19, one-third are admitted to intensive care [310]. Despite this, clinical trials of therapeutic interventions for COVID-19 have almost exclusively focused on adult patients. For example, in the first of two recent studies of the antiviral remdesivir [312, 313], patients younger than 18 years were excluded [314], and the number of children between 12 and 18 years included in the analysis for the second paper was not reported [145]. These studies led to FDA EUA of remdesivir for both adults and children [315], with no published data available on either safety or efficacy in children under 12 years. A phase II/III open-label study in this population has started (the “CARAVAN” trial [147]). Future studies of both therapeutics and vaccines will need to include children to assure their safety and efficacy in this population.

Multisystem inflammatory syndrome in children

Multisystem inflammatory syndrome in children (MIS-C) or Pediatric Multisystem Inflammatory Syndrome is a rare acute inflammatory syndrome with some similarities to Kawasaki disease that has recently been reported in children. Reports from Europe and the United States generally describe critically ill children with fever, rash, conjunctivitis, abdominal complaints, shock, and significant cardiac dysfunction [316-328]. Case definitions have been developed to better characterize these patients (Table 33) [329, 330].

Patients with Kawasaki disease also present with fever and symptoms including rash, conjunctivitis, peripheral extremity changes, lymphadenopathy, and oral mucosal changes such as red, cracked lips and “strawberry tongue.” However, while Kawasaki disease and MIS-C share some similarities, there are also key differences [331]. Both are hyperinflammatory syndromes, both have findings of medium vessel vasculitis and both can present with the signs/symptoms described for Kawasaki disease. MIS-C is more likely to affect older children (average age 8-11 years versus younger than five years in Kawasaki disease), cause more severe disease (more patients presenting with shock), present frequently with gastrointestinal symptoms, includes some neurologic involvement, and more commonly causes cardiac myocarditis and ventricular dysfunction leading to hypotension or arrhythmias. In contrast, Kawasaki disease more commonly causes coronary artery dilatation. A small study of cytokine profiles in children distinguished MIS-C from severe COVID-19 based on a higher level of the combination of TNF-α and IL-10 in MIS-C patients [332].

Empiric treatment of MIS-C has generally involved immunomodulatory agents such as high-dose IV immunoglobulin (2 g/kg), corticosteroids, aspirin and rarely more targeted anti-inflammatory medications such as anakinra [316-319, 325, 326, 328]. Most of the children with MIS-C have had a history of prior SARS-CoV-2 infection several weeks earlier confirmed by viral detection of antibody testing or have had documented prior exposure to COVID-19, suggesting that this condition is a post-infectious immunologic phenomenon.

Future research should focus on how and why the immune system responds to SARS-CoV-2 causing a spectrum of illness in children, identifying genetic or environmental risk factors for MIS-C, and discovering optimum treatment for children with MIS-C. Multidisciplinary, collaborative approaches to data registries and clinical trials that promote evidence-based care for these children are needed.

 

Table 33.png

Discussion

During epidemics like the current COVID-19 pandemic, when there are no clinically proven treatments, the tendency is to use drugs based on in vitro antiviral activity, or on anti-inflammatory effects or based on limited observational studies. It is commendable that observational studies are done during an epidemic, but often they do not have concurrent controls, have a significant risk of bias, and use surrogate outcomes like viral clearance rather than patient-important outcomes. Medications that were thought to be effective based on in vitro studies and observational studies for other diseases were later proven to be ineffective in clinical trials [333].

Due to the understandable urgency in producing, synthesizing and disseminating data during the current pandemic, there has been a noticeable increase in fast track publication of studies. In addition to well-established concerns that may decrease our certainty in the available evidence, there may be additional issues that will ultimately influence the trustworthiness of that evidence, including: 1) Circumvention of usual research steps (delay of IRB approval [334], inclusion of same patients in several studies); 2) Limited peer-review process (the usual due diligence from editors and reviewers is side-stepped, potentially leading to unnoticed errors in data and calculations, incomplete reporting of methods and results, as well as underestimation of study limitations); 3) Increased potential for publication bias (in the interest of showing promising data and in the race to achieve recognition, there may be added inclination to publish positive results and disregard negative ones). The extent and impact of these considerations remain currently uncertain but were acknowledged in the development of this guideline. 

Despite these limitations, the recommendations in this guideline are based on evidence from the best available clinical studies with patient-important endpoints. The panel determined that when an explicit trade-off between the highly uncertain benefits (e.g., the panel was unable to confirm that HCQ increases viral cure or reduces mortality) and the known putative harms (QT prolongation and drug-drug interactions) were considered, a net positive benefit was not reached and could possibly be negative (risk of excess harm). The safety of drugs used for the treatment of COVID-19, especially in patients with cardiovascular disease, immunosuppressive conditions, or those who are critically ill with multi-organ failure has also not been studied. Drugs like AZ and HCQ can cause QT prolongation and potentially life-threatening arrhythmias. Steroids and IL-6 inhibitors can be immunosuppressive and potentially increase risk of secondary infections. Steroids may produce long term side effect such as osteonecrosis [335]. In instances where the panel could not make a determination whether the benefits outweigh harms, it is be ethical and prudent to enroll patients with COVID-19 in clinical trials, rather than use clinically unproven therapies [336]. There are multiple ongoing trials, some with adaptive designs, which potentially can quickly answer pressing questions on efficacy and safety of drugs in the treatment of patients with COVID-19.

We acknowledge that enrolling patients in RCTs might not be feasible for many frontline providers due to limited access and infrastructure. Should lack of access to clinical trials exist, we encourage setting up local or collaborative registries to systematically evaluate the efficacy and safety of drugs to contribute to the knowledge base. Without such evaluations we often attribute success to drugs and failure to disease (COVID-19) [333]. During such a pandemic, barriers to conducting studies and enrolling patients in trials for already overburdened front line providers should be minimized while ensuring the rights and safety of patients [337].

For clinical trials and observational studies, it is critical to determine a priori standardized and practical definitions of patient populations, clinical syndromes, disease severity and outcomes. Observational and non-experimental studies can sometimes answer questions not addressed by trials, but there is still a need for standardized definitions. For clinical syndromes clearly distinguishing between asymptomatic carrier state, upper respiratory tract infection and lower respiratory tract infection is important. Illness severity should be reasonably defined using readily available clinical criteria of end organ failure, like the degree of respiratory failure using SpO2 (percentage of oxyhemoglobin saturation) or PaO2:FiO2 ratios (partial pressure of oxygen in arterial blood: fractional percentage of inspired oxygen) for lower respiratory tract infection, as opposed to location-based severity determinations such as ICU admission, which can lead to bias based on resource limitations (i.e., bed availability) or regional/institutional practice patterns [338]. For outcomes of prophylaxis trials, the primary endpoint should be prevention of infection and for therapeutic trials patient centered outcomes like reduction of mortality (both short term and long term) [339]. Trials should also study treatments in high-risk populations or special populations like immunosuppressed patients, people with HIV, patients with cardiovascular comorbidities and pregnant women. The panel expressed the overarching goal that patients be recruited into ongoing trials, which would provide much needed evidence on the efficacy and safety of various therapies for COVID-19.

This is a living guideline that will be frequently updated as new data emerges. Updates and changes to the guideline will be posted to the IDSA website.

Notes

Acknowledgement

The expert panel thanks the Infectious Diseases Society of America for supporting guideline development, and specifically Cindy Sears, Dana Wollins, Genet Demisashi, and Rebecca Goldwater for their continued support throughout the guideline process. The panel would also like to acknowledge Haya Waseem, Kapeena Sivakumaran, and Nicholas J. Newman for supporting the evidence base for this guideline.

Financial Support

This project was funded in part by a cooperative agreement with the Centers for Disease Control and Prevention (CDC) (grant number 6 NU50CK000477-04-01). The CDC is an agency within the Department of Health and Human Services (HHS). The contents of this guideline do not necessarily represent the policy of CDC or HHS and should not be considered an endorsement by the Federal Government. 

COI Summary

The following list is a reflection of what has been reported to IDSA. To provide thorough transparency, IDSA requires full disclosure of all relationships, regardless of relevancy to the guideline topic. Evaluation of such relationships as potential conflicts of interest is determined by a review process which includes assessment by the Board of Directors liaison to the Standards and Practice Guideline Committee and, if necessary, the Conflicts of Interest and Ethics Committee. The assessment of disclosed relationships for possible COI is based on the relative weight of the financial relationship (i.e., monetary amount) and the relevance of the relationship (i.e., the degree to which an association might reasonably be interpreted by an independent observer as related to the topic or recommendation of consideration). The reader of these guidelines should be mindful of this when the list of disclosures is reviewed. L.B. receives research funding from the National Institutes of Health/National Institute of Allergy and Infectious Diseases, Bill and Melinda Gates Foundation, and Wellcome Trust, and serves as chair of the Antimicrobial Drug Advisory Committee of the Food and Drug Administration. V.C. receives research funding from the Health and Medical Research Fund. K. E. serves as a scientific advisor for Merck, Bionet, IBM, Sanofi, X4 Pharmaceuticals, Inc., Seqirus, Inc., Moderna, Inc. and Pfizer, and receives research funding from the Centers for Disease Control and Prevention and the National Institutes of Health. R. G. has served on a scientific advisory board for Gilead Sciences, Inc., serves on a scientific advisory board for Merck, and receives research funding from the NIH. J.G. serves in an advisory role for Qpex and Shionogi; receives research funding from Merk; previously served in an advisory role for Acceletrate, Achaogen, Astellas, Melinta, Nabriva, Paratek, scPharmaceutic, Spero and Tetraphase; and previously served on the speakers bureau for Astellas, Melinta, Merck and Shiongi. M.H.M receives research funding from the Agency for Healthcare Research and Quality, the Endocrine Society, the Society for Vascular Surgery and The American Society of Hematology and is a Board member for the Evidence Foundation. W.J.M. serves in an advisory role for Seqirus, Inc. and receives research funding from Ansun Biopharma, Astellas Pharma, AstraZeneca, Abbott Laboratories, Enanta Pharmaceuticals, Gilead Sciences, Janssen Pharmaceuticals, Karius, Merck, Melinta Therapeutics, Nabriva Therapeutics, Roche and Tetraphase Pharmaceuticals. S.S. serves as an advisory board member for Amplyx Pharmaceuticals, Inc.; as an advisor/consultant to ReViral Ltd.; receives research funding from Ansun BioPharma, F2G, Shire (now Takeda), University of Nebraska, Cidara Therapeutics; and has served as an advisor for Janssen Pharmaceutica and Acidophil. A.H.S. receives research funding from the U.S. Department of Veterans Affairs. Y.F.Y. receives honoraria for evidence reviews and teaching from the Evidence Foundation, honoraria for evidence reviews for the American Gastroenterological Association, and serves as a Director for the Evidence Foundation and for the U.S. GRADE Network. All other authors: no disclosures reported. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

IDSA Disclaimer

It is important to realize that guidelines cannot always account for individual variation among patients. They are assessments of current scientific and clinical information provided as an educational service; are not continually updated and may not reflect the most recent evidence (new evidence may emerge between the time information is developed and when it is published or read); should not be considered inclusive of all proper treatments methods of care, or as a statement of the standard of care; do not mandate any particular course of medical care; and are not intended to supplant physician judgment with respect to particular patients or special clinical situations. Whether and the extent to which to follow guidelines is voluntary, with the ultimate determination regarding their application to be made by the physician in the light of each patient’s individual circumstances. While IDSA makes every effort to present accurate, complete, and reliable information, these guidelines are presented “as is” without any warranty, either express or implied. IDSA (and its officers, directors, members, employees, and agents) assume no responsibility for any loss, damage, or claim with respect to any liabilities, including direct, special, indirect, or consequential damages, incurred in connection with these guidelines or reliance on the information presented.

The guidelines represent the proprietary and copyrighted property of IDSA. Copyright 2020 Infectious Diseases Society of America. All rights reserved. No part of these guidelines may be reproduced, distributed, or transmitted in any form or by any means, including photocopying, recording, or other electronic or mechanical methods, without the prior written permission of IDSA. Permission is granted to physicians and health care providers solely to copy and use the guidelines in their professional practices and clinical decision-making. No license or permission is granted to any person or entity, and prior written authorization by IDSA is required, to sell, distribute, or modify the guidelines, or to make derivative works of or incorporate the guidelines into any product, including but not limited to clinical decision support software or any other software product. Except for the permission granted above, any person or entity desiring to use the guidelines in any way must contact IDSA for approval in accordance with the terms and conditions of third-party use, in particular any use of the guidelines in any software product.

References

        1. Guan WJ, Ni ZY, Hu Y, et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med 2020.
        2. World Health Organization. Coronavirus disease 2019 (COVID-19) Situation Report - 75. Geneva: World Health Organization, 2020 4 April.
        3. Wu Z, McGoogan JM. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72314 Cases From the Chinese Center for Disease Control and Prevention. JAMA 2020.
        4. Morgan RL, Florez I, Falavigna M, et al. Development of rapid guidelines: 3. GIN-McMaster Guideline Development Checklist extension for rapid recommendations. Health Res Policy Syst 2018; 16(1): 63.
        5. Guyatt GH, Oxman AD, Kunz R, et al. GRADE guidelines: 2. Framing the question and deciding on important outcomes. J Clin Epidemiol 2011; 64(4): 395-400.
        6. U.S. Food and Drug Administration. What is a Serious Adverse Event? Available at: https://www.fda.gov/safety/reporting-serious-problems-fda/what-serious-adverse-event. Accessed 19 June 2020.
        7. National Institute for Health and Care Excellence. Scoping. Interim process and methods for developing rapid guidelines on COVID-19 (PMG35). London: National Institute for Health and Care Excellence, 2020.
        8. Wallace BC, Dahabreh IJ, Trikalinos TA, Lau J, Trow P, Schmid CH. Closing the gap between methodologists and end-users: R as a computational back-end. J Stat Softw 2012; 49(5): 1-15.
        9. Higgins JPT, Thomas J, Chandler J, et al. Cochrane Handbook for Systematic Reviews of Interventions. 2 ed. Chichester (UK): John Wiley & Sons, 2019.
        10. Sterne JA, Hernan MA, Reeves BC, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ 2016; 355: i4919.
        11. Guyatt G, Oxman AD, Akl EA, et al. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J Clin Epidemiol 2011; 64(4): 383-94.
        12. GRADEpro GDT. GRADEpro Guideline Development Tool [Software]. McMaster University, 2015 (developed by Evidence Prime, Inc.). Available at: https://gradepro.org/.
        13. Ben-Zvi I, Kivity S, Langevitz P, Shoenfeld Y. Hydroxychloroquine: from malaria to autoimmunity. Clin Rev Allergy Immunol 2012; 42(2): 145-53.
        14. Keyaerts E, Vijgen L, Maes P, Neyts J, Van Ranst M. In vitro inhibition of severe acute respiratory syndrome coronavirus by chloroquine. Biochem Biophys Res Commun 2004; 323(1): 264-8.
        15. Dyall J, Coleman CM, Hart BJ, et al. Repurposing of clinically developed drugs for treatment of Middle East respiratory syndrome coronavirus infection. Antimicrob Agents Chemother 2014; 58(8): 4885-93.
        16. 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): 269-71.
        17. Yao X, Ye F, Zhang M, et al. In Vitro Antiviral Activity and Projection of Optimized Dosing Design of Hydroxychloroquine for the Treatment of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Clin Infect Dis 2020.
        18. Vincent MJ, Bergeron E, Benjannet S, et al. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J 2005; 2: 69.
        19. Gautret P, Lagier JC, Parola P, et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents 2020; 56(1): 105949.
        20. Li C, Zu S, Deng YQ, et al. Azithromycin Protects against Zika virus Infection by Upregulating virus-induced Type I and III Interferon Responses. Antimicrob Agents Chemother 2019.
        21. Kouznetsova J, Sun W, Martinez-Romero C, et al. Identification of 53 compounds that block Ebola virus-like particle entry via a repurposing screen of approved drugs. Emerg Microbes Infect 2014; 3(12): e84.
        22. Gielen V, Johnston SL, Edwards MR. Azithromycin induces anti-viral responses in bronchial epithelial cells. Eur Respir J 2010; 36(3): 646-54.
        23. Tyteca D, Van Der Smissen P, Mettlen M, et al. Azithromycin, a lysosomotropic antibiotic, has distinct effects on fluid-phase and receptor-mediated endocytosis, but does not impair phagocytosis in J774 macrophages. Exp Cell Res 2002; 281(1): 86-100.
        24. Menzel M, Akbarshahi H, Bjermer L, Uller L. Azithromycin induces anti-viral effects in cultured bronchial epithelial cells from COPD patients. Sci Rep 2016; 6: 28698.
        25. Takizawa H, Desaki M, Ohtoshi T, et al. Erythromycin suppresses interleukin 6 expression by human bronchial epithelial cells: a potential mechanism of its anti-inflammatory action. Biochem Biophys Res Commun 1995; 210(3): 781-6.
        26. Schultz MJ. Macrolide activities beyond their antimicrobial effects: macrolides in diffuse panbronchiolitis and cystic fibrosis. J Antimicrob Chemother 2004; 54(1): 21-8.
        27. Arshad S, Kilgore P, Chaudhry ZS, et al. Treatment with hydroxychloroquine, azithromycin, and combination in patients hospitalized with COVID-19. Int J Infect Dis 2020; 97: 396-403.
        28. Cavalcanti AB, Zampieri FG, Rosa RG, et al. Hydroxychloroquine with or without Azithromycin in Mild-to-Moderate Covid-19. N Engl J Med 2020; 383: 2041-52.
        29. Horby P, Mafham M, Linsell L, et al. Effect of Hydroxychloroquine in Hospitalized Patients with COVID-19: Preliminary results from a multi-centre, randomized, controlled trial. medRxiv 2020.
        30. Chen J, Liu D, Liu L, et al. A pilot study of hydroxychloroquine in treatment of patients with moderate COVID-19. Journal of Zhejiang University (Medical Sciences) 2020; 49(2): 215-9.
        31. Chen Z, Hu J, Zhang Z, et al. Efficacy of hydroxychloroquine in patients with COVID-19: results of a randomized clinical trial. medRxiv 2020: Available at: https://doi.org/10.1101/2020.03.22.20040758 [Preprint 10 April 2020].
        32. 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: 497-511.
        33. Self WH, Semler MW, Leither LM, et al. Effect of Hydroxychloroquine on Clinical Status at 14 Days in Hospitalized Patients With COVID-19: A Randomized Clinical Trial. JAMA 2020; 324(21): 2165-76.
        34. Ulrich RJ, Troxel AB, Carmody E, et al. Treating COVID-19 With Hydroxychloroquine (TEACH): A Multicenter, Double-Blind Randomized Controlled Trial in Hospitalized Patients. Open Forum Infect Dis 2020; 7(10): ofaa446.
        35. Tang W, Cao Z, Han M, et al. Hydroxychloroquine in patients with mainly mild to moderate coronavirus disease 2019: open label, randomised controlled trial. BMJ 2020; 369: m1849.
        36. Geleris J, Sun Y, Platt J, et al. Observational Study of Hydroxychloroquine in Hospitalized Patients with Covid-19. N Engl J Med 2020.
        37. Magagnoli J, Narendran S, Pereira F, et al. Outcomes of hydroxychloroquine usage in United States veterans hospitalized with Covid-19. Med 2020; 1(1): 114-27.e3.
        38. Mahevas M, Tran V-T, Roumier M, et al. No evidence of clinical efficacy of hydroxychloroquine in patients hospitalized for COVID-19 infection with oxygen requirement: results of a study using routinely collected data to emulate a target trial. medRxiv 2020: Available at: https://doi.org/10.1101/2020.04.10.20060699 [Preprint 14 April 2020].
        39. Rosenberg ES, Dufort EM, Udo T, et al. Association of treatment with hydroxychloroquine or azithromycin with in-hospital mortality in patients with COVID-19 in New York state. JAMA 2020; 323(24): 2493-502.
        40. Yu B, Li C, Chen P, et al. Low dose of hydroxychloroquine reduces fatality of critically ill patients with COVID-19. Sci China Life Sci 2020.
        41. Ip A, Berry DA, Hansen E, et al. Hydroxychloroquine and Tocilizumab Therapy in COVID-19 Patients-An Observational Study. PloS One 2020; 15(8): e0237693.
        42. Cipriani A, Zorzi A, Ceccato D, et al. Arrhythmic profile and 24-hour QT interval variability in COVID-19 patients treated with hydroxychloroquine and azithromycin. Int J Cardiol 2020.
        43. Molina JM, Delaugerre C, Goff J, et al. No Evidence of Rapid Antiviral Clearance or Clinical Benefit with the Combination of Hydroxychloroquine and Azithromycin in Patients with Severe COVID-19 Infection. Médecine et Maladies Infectieuses 2020; 50(4): 384.
        44. Gautret P, Lagier JC, Parola P, et al. Clinical and microbiological effect of a combination of hydroxychloroquine and azithromycin in 80 COVID-19 patients with at least a six-day follow up: A pilot observational study. Travel Med Infect Dis 2020; 34: 101663.
        45. Chorin E, Dai M, Shulman E, et al. The QT Interval in Patients with SARS-CoV-2 Infection Treated with Hydroxychloroquine/Azithromycin. medRxiv 2020: Available at: https://doi.org/10.1101/2020.04.02.20047050 [Preprint 3 April 2020].
        46. Wang C, Fortin PR, Li Y, Panaritis T, Gans M, Esdaile JM. Discontinuation of antimalarial drugs in systemic lupus erythematosus. J Rheumatol 1999; 26(4): 808-15.
        47. Youngster I, Arcavi L, Schechmaster R, et al. Medications and glucose-6-phosphate dehydrogenase deficiency: an evidence-based review. Drug Saf 2010; 33(9): 713-26.
        48. Mohammad S, Clowse MEB, Eudy AM, Criscione-Schreiber LG. Examination of Hydroxychloroquine Use and Hemolytic Anemia in G6PDH-Deficient Patients. Arthritis Care Res (Hoboken) 2018; 70(3): 481-5.
        49. Beauverd Y, Adam Y, Assouline B, Samii K. COVID-19 infection and treatment with hydroxychloroquine cause severe haemolysis crisis in a patient with glucose-6-phosphate dehydrogenase deficiency. Eur J Haematol 2020.
        50. Kuipers MT, van Zwieten R, Heijmans J, et al. G6PD deficiency-associated hemolysis and methemoglobinemia in a COVID-19 patient treated with chloroquine. Am J Hematol 2020.
        51. Maillart E, Leemans S, Van Noten H, et al. A case report of serious haemolysis in a glucose-6-phosphate dehydrogenase-deficient COVID-19 patient receiving hydroxychloroquine. Infect Dis (Lond) 2020: 1-3.
        52. Rainsford KD, Parke AL, Clifford-Rashotte M, Kean WF. Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology 2015; 23(5): 231-69.
        53. Morgan ND, Patel SV, Dvorkina O. Suspected hydroxychloroquine-associated QT-interval prolongation in a patient with systemic lupus erythematosus. J Clin Rheumatol 2013; 19(5): 286-8.
        54. Chen CY, Wang FL, Lin CC. Chronic hydroxychloroquine use associated with QT prolongation and refractory ventricular arrhythmia. Clin Toxicol (Phila) 2006; 44(2): 173-5.
        55. Yelve K, Phatak S, Patil MA, Pazare AR. Syncope in a patient being treated for hepatic and intestinal amoebiasis. BMJ Case Rep 2012; 2012: bcr2012006687.
        56. Stas P, Faes D, Noyens P. Conduction disorder and QT prolongation secondary to long-term treatment with chloroquine. Int J Cardiol 2008; 127(2): e80-2.
        57. Ray WA, Murray KT, Hall K, Arbogast PG, Stein CM. Azithromycin and the risk of cardiovascular death. N Engl J Med 2012; 366(20): 1881-90.
        58. von Rosensteil NA, Adam D. Macrolide antibacterials. Drug interactions of clinical significance. Drug Saf 1995; 13(2): 105-22.
        59. Barnabas RV, Brown ER, Bershteyn A, et al. Hydroxychloroquine as Postexposure Prophylaxis to Prevent Severe Acute Respiratory Syndrome Coronavirus 2 Infection : A Randomized Trial. Ann Intern Med 2021; 174(3): 344-52
        60. Boulware DR, Pullen MF, Bangdiwala AS, et al. A Randomized Trial of Hydroxychloroquine as Postexposure Prophylaxis for Covid-19. N Engl J Med 2020; 383(6): 517-25.
        61. Mitja O, Corbacho-Monne M, Ubals M, et al. A Cluster-Randomized Trial of Hydroxychloroquine for Prevention of Covid-19. N Engl J Med 2021; 384(5): 417-27.
        62. Chen F, Chan KH, Jiang Y, et al. In vitro susceptibility of 10 clinical isolates of SARS coronavirus to selected antiviral compounds. J Clin Virol 2004; 31(1): 69-75.
        63. Wu CY, Jan JT, Ma SH, et al. Small molecules targeting severe acute respiratory syndrome human coronavirus. Proc Natl Acad Sci U S A 2004; 101(27): 10012-7.
        64. Chan JF, Yao Y, Yeung ML, et al. Treatment With Lopinavir/Ritonavir or Interferon-beta1b Improves Outcome of MERS-CoV Infection in a Nonhuman Primate Model of Common Marmoset. J Infect Dis 2015; 212(12): 1904-13.
        65. Chu CM, Cheng VC, Hung IF, et al. Role of lopinavir/ritonavir in the treatment of SARS: initial virological and clinical findings. Thorax 2004; 59(3): 252-6.
        66. Spanakis N, Tsiodras S, Haagmans BL, et al. Virological and serological analysis of a recent Middle East respiratory syndrome coronavirus infection case on a triple combination antiviral regimen. Int J Antimicrob Agents 2014; 44(6): 528-32.
        67. Kim UJ, Won EJ, Kee SJ, Jung SI, Jang HC. Combination therapy with lopinavir/ritonavir, ribavirin and interferon-alpha for Middle East respiratory syndrome. Antivir Ther 2016; 21(5): 455-9.
        68. 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): 1695-704.
        69. Cao B, Wang Y, Wen D, et al. A Trial of Lopinavir-Ritonavir in Adults Hospitalized with Severe Covid-19. N Engl J Med 2020.
        70. RECOVERY Collaborative Group, Horby PW, Mafham M, et al. Lopinavir–ritonavir in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. The Lancet 2020; 396(10259): 1345-52.
        71. World Health Organization. Clinical management of severe acute respiratory infection (SARI) when COVID-19 disease is suspected. Available at: https://apps.who.int/iris/bitstream/handle/10665/331446/WHO-2019-nCoV-clinical-2020.4-eng.pdf. Accessed 24 June 2020.
        72. Arabi YM, Mandourah Y, Al-Hameed F, et al. Corticosteroid Therapy for Critically Ill Patients with Middle East Respiratory Syndrome. Am J Respir Crit Care Med 2018; 197(6): 757-67.
        73. Lee N, Allen Chan KC, Hui DS, et al. Effects of early corticosteroid treatment on plasma SARS-associated Coronavirus RNA concentrations in adult patients. J Clin Virol 2004; 31(4): 304-9.
        74. Xiao JZ, Ma L, Gao J, et al. [Glucocorticoid-induced diabetes in severe acute respiratory syndrome: the impact of high dosage and duration of methylprednisolone therapy]. Zhonghua Nei Ke Za Zhi 2004; 43(3): 179-82.
        75. Laurent A, Bonnet M, Capellier G, Aslanian P, Hebert P. Emotional Impact of End-of-Life Decisions on Professional Relationships in the ICU: An Obstacle to Collegiality? Crit Care Med 2017; 45(12): 2023-30.
        76. Villar J, Ferrando C, Martinez D, et al. Dexamethasone treatment for the acute respiratory distress syndrome: a multicentre, randomised controlled trial. Lancet Respir Med 2020; 8(3): 267-76.
        77. WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) 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.
        78. RECOVERY Collaborative Group, Horby P, Lim WS, et al. Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med 2021; 384: 693-704.
        79. 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.
        80. Dequin PF, Heming N, Meziani F, et al. Effect of Hydrocortisone on 21-Day Mortality or Respiratory Support Among Critically Ill Patients With COVID-19: A Randomized Clinical Trial. JAMA 2020.
        81. Writing Committee for the REMAP-CAP Investigators, Angus DC, Derde L, et al. Effect of Hydrocortisone on Mortality and Organ Support in Patients With Severe COVID-19: The REMAP-CAP COVID-19 Corticosteroid Domain Randomized Clinical Trial. JAMA 2020.
        82. Petersen MW, Meyhoff TS, Helleberg M, et al. Low-dose hydrocortisone in patients with COVID-19 and severe hypoxia (COVID STEROID) trial-Protocol and statistical analysis plan. Acta Anaesthesiol Scand 2020.
        83. Jeronimo CMP, Farias MEL, Val FFA, et al. Methylprednisolone as Adjunctive Therapy for Patients Hospitalized With COVID-19 (Metcovid): A Randomised, Double-Blind, Phase IIb, Placebo-Controlled Trial. Clin Infect Dis 2020.
        84. Corral L, Bahamonde A, Arnaiz delas Revillas F, et al. GLUCOCOVID: A controlled trial of methylprednisolone in adults hospitalized with COVID-19 pneumonia. medRxiv 2020: 2020.06.17.20133579.
        85. Salton F, Confalonieri P, Santus P, et al. Prolonged low-dose methylprednisolone in patients with severe COVID-19 pneumonia. medRxiv 2020: 2020.06.17.20134031.
        86. Wang Y, Jiang W, He Q, et al. Early, low-dose and short-term application of corticosteroid treatment in patients with severe COVID-19 pneumonia: single-center experience from Wuhan, China. medRxiv 2020: 2020.03.06.20032342.
        87. 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.
        88. Fernandez-Cruz A, Ruiz-Antoran B, Munoz-Gomez A, et al. Impact of glucocorticoid treatment in SARS-CoV-2 infection mortality: a retrospective controlled cohort study. medRxiv 2020: 2020.05.22.20110544.
        89. Lu X, Chen T, Wang Y, et al. Adjuvant corticosteroid therapy for critically ill patients with COVID-19. medRxiv 2020: 2020.04.07.20056390.
        90. Yuan M, Xu X, Xia D, et al. Effects of Corticosteroid Treatment for Non-Severe COVID-19 Pneumonia: A Propensity Score-Based Analysis. Shock 2020.
        91. Henzen C, Suter A, Lerch E, Urbinelli R, Schorno XH, Briner VA. Suppression and recovery of adrenal response after short-term, high-dose glucocorticoid treatment. Lancet 2000; 355(9203): 542-5.
        92. Siemieniuk RA, Meade MO, Alonso-Coello P, et al. Corticosteroid Therapy for Patients Hospitalized With Community-Acquired Pneumonia: A Systematic Review and Meta-analysis. Ann Intern Med 2015; 163(7): 519-28.
        93. Chen G, Wu D, Guo W, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 2020; 130(5): 2620-9.
        94. Kalikshtein DB, Levantovskaia OM, Vyshenepol'skii I, Ol'shanskii A. [Coagulation and anticoagulation systems of the blood in allergic diseases]. Sov Med 1988; (9): 104-6.
        95. REMAP-CAP Investigators, Gordon AC, Mouncey PR, et al. Interleukin-6 Receptor Antagonists in Critically Ill Patients with Covid-19 – Preliminary report. medRxiv 2021: Available at: https://doi.org/10.1101/2021.01.07.21249390 [Preprint 9 January 2021].
        96. Hermine O, Mariette X, Tharaux PL, et al. Effect of Tocilizumab vs Usual Care in Adults Hospitalized With COVID-19 and Moderate or Severe Pneumonia: A Randomized Clinical Trial. JAMA Intern Med 2020; 181(1): 32-40.
        97. Horby PW, Pessoa-Amorim G, Peto L, et al. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): preliminary results of a randomised, controlled, open-label, platform trial. Lancet 2021; 397(10285): 1637-45.
        98. Rosas I, Bräu N, Waters M, et al. Tocilizumab in hospitalized patients with COVID-19 pneumonia. MedRxiv 2020: Available at: https://doi.org/10.1101/2020.08.27.20183442 [Preprint 12 September 2020].
        99. Salama C, Han J, Yau L, et al. Tocilizumab in Patients Hospitalized with Covid-19 Pneumonia. N Engl J Med 2021; 384(1): 20-30.
        100. Salvarani C, Dolci G, Massari M, et al. Effect of Tocilizumab vs Standard Care on Clinical Worsening in Patients Hospitalized With COVID-19 Pneumonia: A Randomized Clinical Trial. JAMA Intern Med 2020; 181(1): 24-31.
        101. Stone JH, Frigault MJ, Serling-Boyd NJ, et al. Efficacy of Tocilizumab in Patients Hospitalized with Covid-19. N Engl J Med 2020; 383: 2333-44.
        102. Veiga VC, Prats J, Farias DLC, et al. Effect of tocilizumab on clinical outcomes at 15 days in patients with severe or critical coronavirus disease 2019: randomised controlled trial. BMJ 2021; 372: n84.
        103. Lescure FX, Honda H, Fowler RA, et al. Sarilumab in patients admitted to hospital with severe or critical COVID-19: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med 2021; 9(5): 522-32.
        104. Sivapalasingam S, Lederer D, Bhore R, et al. A Randomized Placebo-Controlled Trial of Sarilumab in Hospitalized Patients with Covid-19. medRxiv 2021: Available at: https://doi.org/10.1101/2021.05.13.21256973 [Preprint 14 May 2021].
        105. Godolphin PJ, Fisher DJ, Berry LR, et al. Association between tocilizumab, sarilumab and all-cause mortality at 28 days in hospitalized patients with COVID-19: A network meta-analysis. medRxiv 2021: Available at: https://doi.org/10.1101/2021.08.26.21262523 [Preprint 28 August 2021].
        106. Rojo M, Cano-Valderrama O, Picazo S, et al. Gastrointestinal Perforation After Treatment With Tocilizumab : An Unexpected Consequence of COVID-19 Pandemic. Am Surg 2020; 86(6): 565-6.
        107. Gonzalvez Guardiola P, Diez Ares JA, Peris Tomas N, Sebastian Tomas JC, Navarro Martinez S. Intestinal perforation in patient with COVID-19 infection treated with tocilizumab and corticosteroids. Report of a clinical case. Cir Esp 2020.
        108. Ranchal P, Yates E, Gupta R, Aronow WS. Tocilizumab-Associated Bowel Perforation in SARS-CoV-2 Infection. Am J Ther 2020.
        109. Bruce-Hickman D, Sajeed SM, Pang YH, Seow CS, Chen W, Gulati Kansal M. Bowel ulceration following tocilizumab administration in a COVID-19 patient. BMJ Open Gastroenterol 2020; 7(1).
        110. Guaraldi G, Meschiari M, Cozzi-Lepri A, et al. Tocilizumab in patients with severe COVID-19: a retrospective cohort study. Lancet Rheumatol 2020; 2(8): e474-e84.
        111. Somers EC, Eschenauer GA, Troost JP, et al. Tocilizumab for treatment of mechanically ventilated patients with COVID-19. Clin Infect Dis 2020.
        112. Li L, Zhang W, Hu Y, et al. Effect of Convalescent Plasma Therapy on Time to Clinical Improvement in Patients With Severe and Life-threatening COVID-19: A Randomized Clinical Trial. JAMA 2020; 324(5): 460-70.
        1. Gharbharan A, Jordans CC, Geurts van Kessel C, et al. Effects of potent neutralizing antibodies from convalescent plasma in patients hospitalized for severe SARS-CoV-2 infection. Nat Commun 2021; 12(3189).
        2. AlQahtani M, Abdulrahman A, AlMadani A, et al. Randomized controlled trial of convalescent plasma therapy against standard therapy in patients with severe COVID-19 disease. Sci Rep 2021; 11: 9927.
        3. Avendaño-Solà C, Ramos-Martinez A, Munez-Rubio E, et al. A multicenter randomized open-label clinical trial for convalescent plasma in patients hospitalized with COVID-19 pneumonia. J Clin Invest 2021; 131(20).
        4. Libster R, Marc GP, Wappner D, et al. Prevention of severe COVID-19 in the elderly by early high-titer plasma. medRxiv 2020: Available at: https://doi.org/10.1101/2020.11.20.20234013 [Preprint 21 November 2020].
        5. Joyner MJ, Senefeld JW, Klassen SA, et al. Effect of convalescent plasma on mortality among hospitalized patients with COVID-19: initial three-month experience. medRxiv 2020: Available at: https://doi.org/10.1101/2020.08.12.20169359 [Preprint 12 August 2020].
        6. Expanded Access to Convalescent Plasma for the Treatment of Patients Team, Joyner M. Convalescent Plasma COVID-19 (Coronavirus) Treatment. Available at: https://www.uscovidplasma.org/. Accessed 1 November 2021.
        7. Ray Y, Paul SR, Bandopadhyay P, et al. Clinical and immunological benefits of convalescent plasma therapy in severe COVID-19: insights from a single center open label randomised control trial. medRxiv 2020: Available at: https://doi.org/10.1101/2020.11.25.20237883 [Preprint 29 November 2020].
        8. Simonovich VA, Burgos Pratx LD, Scibona P, et al. A Randomized Trial of Convalescent Plasma in Covid-19 Severe Pneumonia. N Engl J Med 2021; 384(7): 619-29.
        9. Bégin P, Callum J, Jamula E, et al. Convalescent plasma for hospitalized patients with COVID-19: an open-label, randomized controlled trial. Nat Med 2021: Available at: https://doi.org/10.1038/s41591-021-01488-2 [Epub ahead of print 9 September 2021].
        10. U.S. Food and Drug Administration. Recommendations for Investigational COVID-19 Convalescent Plasma. Available at: https://www.fda.gov/vaccines-blood-biologics/investigational-new-drug-ind-or-device-exemption-ide-process-cber/recommendations-investigational-covid-19-convalescent-plasma. Accessed 28 August 2020.
        11. U.S. Food and Drug Administration. FDA In Brief: FDA Updates Emergency Use Authorization for COVID-19 Convalescent Plasma to Reflect New Data. Available at: https://www.fda.gov/news-events/fda-brief/fda-brief-fda-updates-emergency-use-authorization-covid-19-convalescent-plasma-reflect-new-data. Accessed 30 March 2021.
        12. Agarwal A, Mukherjee A, Kumar G, et al. Convalescent plasma in the management of moderate covid-19 in adults in India: open label phase II multicentre randomised controlled trial (PLACID Trial). BMJ 2020; 371: m4232.
        13. O’Donnell MR, Grinsztejn B, Cummings MJ, et al. A randomized double-blind controlled trial of convalescent plasma in adults with severe COVID-19. J Clin Invest 2021; 131(13): e150646.
        14. 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): 2049-59.
        15. Balcells ME, Rojas L, Le Corre N, et al. Early versus deferred anti-SARS-CoV-2 convalescent plasma in patients admitted for COVID-19: A randomized phase II clinical trial. PLoS Med 2021; 18(3): e1003415.
        16. Joyner MJ, Bruno KA, Klassen SA, et al. Safety Update: COVID-19 Convalescent Plasma in 20,000 Hospitalized Patients. Mayo Clin Proc 2020; 95(9): 1888-97.
        17. Korley FK, Durkalski-Mauldin V, Yeatts SD, et al. Early Convalescent Plasma for High-Risk Outpatients with Covid-19. N Engl J Med 2021; 385(21): 1951-60.
        18. Libster R, Perez Marc G, Wappner D, et al. Early High-Titer Plasma Therapy to Prevent Severe Covid-19 in Older Adults. N Engl J Med 2021; 384(7): 610-8.
        19. de Candia P, Prattichizzo F, Garavelli S, et al. Effect of time and titer in convalescent plasma therapy for COVID-19. iScience 2021; 24(8): 102898.
        20. U.S. Food and Drug Administration. FDA Issues Emergency Use Authorization for Convalescent Plasma as Potential Promising COVID–19 Treatment, Another Achievement in Administration’s Fight Against Pandemic. Available at: https://www.fda.gov/news-events/press-announcements/fda-issues-emergency-use-authorization-convalescent-plasma-potential-promising-covid-19-treatment. Accessed 4 November 2021.
        21. Joyner MJ, Carter RE, Senefeld JW, et al. Convalescent Plasma Antibody Levels and the Risk of Death from Covid-19. N Engl J Med 2021; 384(11): 1015-27.
        22. Salazar E, Christensen PA, Graviss EA, et al. Significantly Decreased Mortality in a Large Cohort of Coronavirus Disease 2019 (COVID-19) Patients Transfused Early with Convalescent Plasma Containing High-Titer Anti-Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike Protein IgG. Am J Pathol 2021; 191(1): 90-107.
        23. Arnold Egloff SA, Junglen A, Restivo JS, et al. Convalescent plasma associates with reduced mortality and improved clinical trajectory in patients hospitalized with COVID-19. J Clin Invest 2021; 131(20).
        24. Thompson MA, Henderson JP, Shah PK, et al. Association of Convalescent Plasma Therapy With Survival in Patients With Hematologic Cancers and COVID-19. JAMA Oncol 2021; 7(8): 1167-75.
        25. Lo MK, Jordan R, Arvey A, et al. GS-5734 and its parent nucleoside analog inhibit Filo-, Pneumo-, and Paramyxoviruses. Sci Rep 2017; 7: 43395.
        26. Sheahan TP, Sims AC, Graham RL, et al. Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses. Sci Transl Med 2017; 9(396).
        27. Warren TK, Jordan R, Lo MK, et al. Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys. Nature 2016; 531(7594): 381-5.
        28. Williamson BN, Feldmann F, Schwarz B, et al. Clinical benefit of remdesivir in rhesus macaques infected with SARS-CoV-2. Nature 2020.
        29. Gottlieb RL, Vaca CE, Paredes R, et al. Early Remdesivir to Prevent Progression to Severe Covid-19 in Outpatients. N Engl J Med 2021: Available at: https://doi.org/10.1056/nejmoa2116846 [Epub ahead of print 22 December 2021].
        30. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the Treatment of Covid-19 - Final Report. N Engl J Med 2020; 383(19): 1813-26.
        31. 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): 1048-57.
        32. Wang Y, Zhang D, Du G, et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet 2020; 395(10236): 1569-78.
        33. Goldman JD, Lye DCB, Hui DS, et al. Remdesivir for 5 or 10 Days in Patients with Severe Covid-19. N Engl J Med 2020; 383: 1827-37.
        34. Goldman DL, Aldrich ML, Hagmann SHF, et al. Compassionate Use of Remdesivir in Children With Severe COVID-19. Pediatrics 2021; 147(5).
        35. Gilead Sciences, Inc. Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Efficacy of Remdesivir (GS-5734™) in Participants From Birth to < 18 Years of Age With Coronavirus Disease 2019 (COVID-19) (CARAVAN). Available at: https://www.clinicaltrials.gov/ct2/show/NCT04431453. Accessed 18 November 2020.
        36. Borrell B. New York clinical trial quietly tests heartburn remedy against coronavirus. Available at: https://www.sciencemag.org/news/2020/04/new-york-clinical-trial-quietly-tests-heartburn-remedy-against-coronavirus.
        37. Wu C, Liu Y, Yang Y, et al. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm Sin B 2020; 10(5): 766-88.
        38. Freedberg DE, Conigliaro J, Wang TC, et al. Famotidine use is associated with improved clinical outcomes in hospitalized COVID-19 patients: A propensity score matched retrospective cohort study. Gastroenterology 2020; 159(3): 1129-31.
        39. Jones BE, Brown-Augsburger PL, Corbett KS, et al. LY-CoV555, a rapidly isolated potent neutralizing antibody, provides protection in a non-human primate model of SARS-CoV-2 infection. bioRxiv 2020: Available at https://doi.org/10.1101/2020.09.30.318972 [Preprint 9 October 2020].
        40. Baum A, Ajithdoss D, Copin R, et al. REGN-COV2 antibodies prevent and treat SARS-CoV-2 infection in rhesus macaques and hamsters. Science 2020; 370(6520): 1110-5.
        41. Cohen MS, Nirula A, Mulligan MJ, et al. Effect of Bamlanivimab vs Placebo on Incidence of COVID-19 Among Residents and Staff of Skilled Nursing and Assisted Living Facilities: A Randomized Clinical Trial. JAMA 2021; 326(1): 46-55.
        42. U.S. Food and Drug Administration. FACT SHEET FOR HEALTHCARE PROVIDERS: EMERGENCY USE AUTHORIZATION FOR EVUSHELD™ (tixagevimab co-packaged with cilgavimab). Available at: https://www.fda.gov/media/154701/download. Accessed 22 December 2021.
        43. Levin M, Ustianowski A, De Wit S, et al. Intramuscular AZD7442 (tixagevimab/cilgavimab) for prevention of COVID-19. 2021: [Under review].
        44. O'Brien MP, Forleo-Neto E, Musser BJ, et al. Subcutaneous REGEN-COV Antibody Combination to Prevent Covid-19. N Engl J Med 2021; 385: 1184-95.
        45. Chen P, Nirula A, Heller B, et al. SARS-CoV-2 Neutralizing Antibody LY-CoV555 in Outpatients with Covid-19. N Engl J Med 2021; 384(3): 229-37.
        46. ACTIV-3/TICO LY-CoV555 Study Group, Grund B, Barkauskas CE, et al. A Neutralizing Monoclonal Antibody for Hospitalized Patients with Covid-19. N Engl J Med 2021; 384: 905-14.
        47. Dougan M, Nirula A, Azizad M, et al. The Impact of Bamlanivimab + Etesevimab Neutralizing Antibody Combination Treatment on Hospitalization Rates and Deaths Among High-Risk Patients Presenting With Mild-to-Moderate COVID-19 Illness. 2021: [Under review].
        48. Gupta A, Gonzalez-Rojas Y, Juarez E, et al. Early Covid-19 Treatment With SARS-CoV-2 Neutralizing Antibody Sotrovimab. N Engl J Med 2021; 385: 1941-50.
        49. Weinreich DM, Sivapalasingam S, Norton T, et al. REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19. N Engl J Med 2021; 384(3): 238-51.
        50. Regeneron Pharmaceuticals, Inc. Phase 3 Trial Shows Regen-CoV™ (Casirivimab with Imdevimab) Antibody Cocktail Reduced Hospitalization or Death by 70% in Non-Hospitalized COVID-19 Patients. Available at: https://investor.regeneron.com/news-releases/news-release-details/phase-3-trial-shows-regen-covtm-casirivimab-imdevimab-antibody. Accessed 9 April 2021.
        51. U.S. Food and Drug Administration. Coronavirus (COVID-19) Update: FDA Revokes Emergency Use Authorization for Monoclonal Antibody Bamlanivimab. Available at: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-revokes-emergency-use-authorization-monoclonal-antibody-bamlanivimab. Accessed 13 June 2021.
        52. O'Brien MP, Forleo-Neto E, Sarkar N, et al. Subcutaneous REGEN-COV Antibody Combination in Early SARS-CoV-2 Infection. medRxiv 2021: Available at: https://doi.org/10.1101/2021.06.14.21258569 [Preprint 14 June 2021].
        53. U.S. Food and Drug Administration. Fact Sheet for Health Care Providers: Emergency Use Authorization (EUA) of Regen-CoV™ (casirivimab with imdevimab). Available at: https://www.fda.gov/media/145611/download. Accessed 13 August 2021.
        54. Centers for Disease Control and Prevention. SARS-CoV-2 Variant Classifications and Definitions. Available at: https://www.cdc.gov/coronavirus/2019-ncov/variants/variant-info.html. Accessed 15 June 2021.
        55. U.S. Food and Drug Administration. Fact Sheet for Health Care Providers: Emergency Use Authorization (EUA) of Bamlanivimab and Etesevimab. Available at: https://www.fda.gov/media/145802/download. Accessed 9 April 2021.
        56. Planas D, Veyer D, Baidaliuk A, et al. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature 2021; 596(7871): 276-80.
        57. GlaxoSmithKline LLC, U.S. Food and Drug Administration. Fact Sheet for Healthcare Providers: Emergency Use Authorization (EUA) of Sotrovimab. Available at: https://www.fda.gov/media/149534/download. Accessed 15 June 2021.
        58. Jorgensen SCJ, Tse CLY, Burry L, Dresser LD. Baricitinib: A Review of Pharmacology, Safety, and Emerging Clinical Experience in COVID-19. Pharmacotherapy 2020; 40(8): 843-56.
        59. Bekerman E, Neveu G, Shulla A, et al. Anticancer kinase inhibitors impair intracellular viral trafficking and exert broad-spectrum antiviral effects. J Clin Invest 2017; 127(4): 1338-52.
        60. Neveu G, Ziv-Av A, Barouch-Bentov R, Berkerman E, Mulholland J, Einav S. AP-2-associated protein kinase 1 and cyclin G-associated kinase regulate hepatitis C virus entry and are potential drug targets. J Virol 2015; 89(8): 4387-404.
        61. Richardson P, Griffin I, Tucker C, et al. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet 2020; 395(10223): e30-e1.
        62. Cantini F, Niccoli L, Matarrese D, Nicastri E, Stobbione P, Goletti D. Baricitinib therapy in COVID-19: A pilot study on safety and clinical impact. J Infect 2020; 81(2): 318-56.
        63. Titanji BK, Farley MM, Mehta A, et al. Use of Baricitinib in Patients with Moderate and Severe COVID-19. Clin Infect Dis 2020.
        64. Rodriguez-Garcia JL, Sanchez-Nievas G, Arevalo-Serrano J, Garcia-Gomez C, Jimenez-Vizuete JM, Martinez-Alfaro E. Baricitinib improves respiratory function in patients treated with corticosteroids for SARS-CoV-2 pneumonia: an observational cohort study. Rheumatology (Oxford) 2021; 60(1): 399-407.
        65. Cantini F, Niccoli L, Nannini C, et al. Beneficial impact of Baricitinib in COVID-19 moderate pneumonia; multicentre study. J Infect 2020; 81(4): 647-79.
        66. Marconi VC, Ramanan AV, de Bono S, et al. Baricitinib plus Standard of Care for Hospitalized Adults with COVID-19. medRxiv 2021: Available at: https://doi.org/10.1101/2021.04.30.21255934 [Preprint 3 May 2021].
        67. OLUMIANT (baricitinib) tablet, for oral use (package insert). Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/207924s000lbl.pdf. Accessed 23 December 2020.
        68. King B, Maari C, Lain E, et al. Extended Safety Analysis of Baricitinib 2 mg in Adult Patients with Atopic Dermatitis: An Integrated Analysis from Eight Randomized Clinical Trials. Am J Clin Dermatol 2021; 22(3): 395-405.
        69. Winthrop KL, Harigai M, Genovese MC, et al. Infections in baricitinib clinical trials for patients with active rheumatoid arthritis. Ann Rheum Dis 2020; 79(10): 1290-7.
        70. Connors JM, Levy JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood 2020; 135(23): 2033-40.
        71. Kalil AC, Patterson TF, Mehta AK, et al. Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19. N Engl J Med 2021; 384: 795-807.
        72. 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: S2213-600(21)00331-3 [Epub ahead of print 31 August 2021]
        73. Ely EW, Ramanan AV, Kartman CE, et al. Baricitinib plus Standard of Care for Hospitalised Adults with COVID-19 on Invasive Mechanical Ventilation or Extracorporeal Membrane Oxygenation: Results of a Randomised, Placebo-Controlled Trial. medRxiv 2021: Available at: https://doi.org/10.1101/2021.10.11.21263897 [Preprint 12 October 2021].
        74. Guimaraes PO, Quirk D, Furtado RH, et al. Tofacitinib in Patients Hospitalized with Covid-19 Pneumonia. N Engl J Med 2021; 385(5): 406-15.
        75. U.S. Food and Drug Administration. Safety trial finds risk of blood clots in the lungs and death with higher dose of tofacitinib (Xeljanz, Xeljanz XR) in rheumatoid arthritis patients; FDA to investigate. Available at: https://www.fda.gov/drugs/drug-safety-and-availability/safety-trial-finds-risk-blood-clots-lungs-and-death-higher-dose-tofacitinib-xeljanz-xeljanz-xr. Accessed 23 July 2021.
        76. U.S. Food and Drug Administration. FDA approves Boxed Warning about increased risk of blood clots and death with higher dose of arthritis and ulcerative colitis medicine tofacitinib (Xeljanz, Xeljanz XR). Available at: https://www.fda.gov/drugs/drug-safety-and-availability/fda-approves-boxed-warning-about-increased-risk-blood-clots-and-death-higher-dose-arthritis-and. Accessed 23 July 2021.
        77. U.S. Food and Drug Administration. Initial safety trial results find increased risk of serious heart-related problems and cancer with arthritis and ulcerative colitis medicine Xeljanz, Xeljanz XR (tofacitinib). Available at: https://www.fda.gov/drugs/drug-safety-and-availability/initial-safety-trial-results-find-increased-risk-serious-heart-related-problems-and-cancer-arthritis. Accessed 23 July 2021.
        78. U.S. Food and Drug Administration. Xeljanz, Xeljanz XR (tofacitinib): Drug Safety Communication - Initial Safety Trial Results Find Increased Risk of Serious Heart-related Problems and Cancer with Arthritis and Ulcerative Colitis Medicine. Available at: https://www.fda.gov/safety/medical-product-safety-information/xeljanz-xeljanz-xr-tofacitinib-drug-safety-communication-initial-safety-trial-results-find-increased. Accessed 23 July 2021.
        79. Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antiviral Res 2020; 178: 104787.
        80. Bray M, Rayner C, Noel F, Jans D, Wagstaff K. Ivermectin and COVID-19: A report in Antiviral Research, widespread interest, an FDA warning, two letters to the editor and the authors' responses. Antiviral Res 2020; 178: 104805.
        81. Zhang X, Song Y, Ci X, et al. Ivermectin inhibits LPS-induced production of inflammatory cytokines and improves LPS-induced survival in mice. Inflamm Res 2008; 57(11): 524-9.
        82. Yan S, Ci X, Chen N, et al. Anti-inflammatory effects of ivermectin in mouse model of allergic asthma. Inflamm Res 2011; 60(6): 589-96.
        83. Ahmed S, Karim MM, Ross AG, et al. A five-day course of ivermectin for the treatment of COVID-19 may reduce the duration of illness. Int J Infect Dis 2020; 103: 214-6.
        84. Chachar AZK, Khan KA, Asif M, Tanveer K, Khaqan A, Basri R. Effectiveness of Ivermectin in SARS-CoV-2/COVID-19 Patients. Int J Sci 2020; 9(09): 31-5.
        85. Chaccour C, Casellas A, Blanco-Di Matteo A, et al. The effect of early treatment with ivermectin on viral load, symptoms and humoral response in patients with non-severe COVID-19: A pilot, double-blind, placebo-controlled, randomized clinical trial. EClinicalMedicine 2021; 32: 100720.
        86. Gorial FI, Mashhadani S, Sayaly HM, et al. Effectiveness of ivermectin as add-on therapy in COVID-19 management (pilot trial). medRxiv 2020: Available at: https://doi.org/10.1101/2020.07.07.20145979 [Preprint 8 July 2020].
        87. Hashim HA, Maulood MF, Rasheed AM, Fatak DF, Kabah KK, Abdulamir AS. Controlled randomized clinical trial on using Ivermectin with Doxycycline for treating COVID-19 patients in Baghdad, Iraq. medRxiv 2020: Available at: https://doi.org/10.1101/2020.10.26.20219345 [Preprint 27 October 2020].
        88. Podder CS, Chowdhury N, Sina MI, Ul Haque WMM. Outcome of ivermectin treated mild to moderate COVID-19 cases: a single-centre, open-label, randomised controlled study. IMC J Med Sci 2020; 14(2): 11-8.
        89. Rajter JC, Sherman MS, Fatteh N, Vogel F, Sacks J, Rajter JJ. Use of Ivermectin Is Associated With Lower Mortality in Hospitalized Patients With Coronavirus Disease 2019: The Ivermectin in COVID Nineteen Study. Chest 2021; 159(1): 85-92.
        90. Bukhari SKHS, Asghar A, Perveen N, et al. Efficacy of Ivermectin in COVID-19 Patients with Mild to Moderate Disease. medRxiv 2021: Available at: https://doi.org/10.1101/2021.02.02.21250840 [Preprint 5 February 2021].
        91. Ravikirti, Roy R, Pattadar C, et al. Ivermectin as a potential treatment for mild to moderate COVID-19–A double blind randomized placebo-controlled trial. medRxiv 2021: Available at: https://doi.org/10.1101/2021.01.05.21249310 [Preprint 9 January 2021].
        92. Lopez-Medina E, Lopez P, Hurtado IC, et al. Effect of Ivermectin on Time to Resolution of Symptoms Among Adults With Mild COVID-19: A Randomized Clinical Trial. JAMA 2021; 325(14): 1426-35.
        93. Pott-Junior H, Bastos Paoliello MM, Miguel AQC, et al. Use of ivermectin in the treatment of Covid-19: A pilot trial. Toxicol Rep 2021; 8: 505-10.
        94. Mohan A, Tiwari P, Suri T, Mittal S, Patel AA, Jain A. Ivermectin in mild and moderate COVID-19 (RIVET-COV): a randomized, placebo-controlled trial. Research Square 2021: Available at: https://doi.org/10.21203/rs.3.rs-191648/v1 [Preprint 2 February 2021].
        95. Beltran-Gonzalez JL, Gonzalez-Gamez M, Mendoza-Enciso E-A, et al. Efficacy and safety of Ivermectin and Hydroxychloroquine in patients with severe COVID-19. A randomized controlled trial. medRxiv 2021: Available at: https://doi.org/10.1101/2021.02.18.21252037 [Preprint 23 February 2021].
        96. Krolewiecki A, Lifschitz A, Moragas M, et al. Antiviral effect of high-dose ivermectin in adults with COVID-19: A proof-of-concept randomized trial. EClinicalMedicine 2021; 37: 100959.
        97. Abd-Elsalam S, Noor RA, Badawi R, et al. Clinical study evaluating the efficacy of ivermectin in COVID-19 treatment: A randomized controlled study. J Med Virol 2021; 93(10): 5833-8.
        98. Mahmud R, Rahman MM, Alam I, et al. Ivermectin in combination with doxycycline for treating COVID-19 symptoms: a randomized trial. J Int Med Res 2021; 49(5): 3000605211013550.
        99. Vallejos J, Zoni R, Bangher M, et al. Ivermectin to prevent hospitalizations in patients with COVID-19 (IVERCOR-COVID19) a randomized, double-blind, placebo-controlled trial. BMC Infect Dis 2021; 21(1): 635.
        100. Biber A, Mandelboim M, Harmelin G, et al. Favorable outcome on viral load and culture viability using Ivermectin in early treatment of non-hospitalized patients with mild COVID-19, A double-blind, randomized placebo-controlled trial. medRxiv 2021: Available at: https://doi.org/10.1101/2021.05.31.21258081 [Preprint 31 May 2021].
        101. Babalola OE, Bode CO, Ajayi AA, et al. Ivermectin shows clinical benefits in mild to moderate Covid19 disease: A randomised controlled double blind dose response study in Lagos. medRxiv 2021: Available at: https://doi.org/10.1101/2021.01.05.21249131 [Preprint 6 January 2021].
        102. Niaee MS, Gheibi N, Namdar P, et al. Ivermectin as an adjunct treatment for hospitalized adult COVID-19 patients: A randomized multi-center clinical trial. 2020: Available at: https://doi.org/10.21203/rs.3.rs-109670/v1 [Preprint 24 November 2020].
        103. Rezaie S. COVID-19 Update: Ivermectin. Available at: https://rebelem.com/covid-19-update-ivermectin/. Accessed 10 February 2021.
        104. Shoumann WM, Hegazy AA, Nafae RM, et al. Use of Ivermectin as a Potential Chemoprophylaxis for COVID-19 in Egypt: A Randomized Clinical Trial. JCDR 2021; 15(2): OC27-OC32.
        105. Peral de Bruno MdlA, Chala RE. Prophylaxis Covid-19 in Healthcare Agents by Intensive Treatment With Ivermectin and Iota-carrageenan (Ivercar-Tuc). Available at: https://clinicaltrials.gov/ct2/show/NCT04701710.
        106. Raad H. In vivo use of ivermectin (IVR) for treatment for corona virus infected patients (COVID-19): a randomized controlled trial. Available at: http://www.chictr.org.cn/showprojen.aspx?proj=54707
        107. Mahmud R. Clinical Trial of Ivermectin Plus Doxycycline for the Treatment of Confirmed Covid-19 Infection. Available at: https://clinicaltrials.gov/ct2/show/NCT04523831.
        108. 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).
        109. Gordon DE, Jang GM, Bouhaddou M, et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature 2020; 583(7816): 459-68.
        110. Ishima T, Fujita Y, Hashimoto K. Interaction of new antidepressants with sigma-1 receptor chaperones and their potentiation of neurite outgrowth in PC12 cells. Eur J Pharmacol 2014; 727: 167-73.
        111. Geiser F, Conrad R, Imbierowicz K, et al. Coagulation activation and fibrinolysis impairment are reduced in patients with anxiety and depression when medicated with serotonergic antidepressants. Psychiatry Clin Neurosci 2011; 65(5): 518-25.
        112. Duerschmied D, Suidan GL, Demers M, et al. Platelet serotonin promotes the recruitment of neutrophils to sites of acute inflammation in mice. Blood 2013; 121(6): 1008-15.
        113. 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): 2292-300.
        114. Reis G, dos Santos Moreira Silva EA, Medeiros Silva DC, et al. Effect of early treatment with fluvoxamine on risk of emergency care and hospitalisation among patients with COVID-19: the TOGETHER randomised, platform clinical trial. Lancet 2021; S2214-109X(21): 00448-4.
        115. U.S. Food and Drug Administration. Fact Sheet for Healthcare Providers: Emergency Use Authorization for Paxlovid™. Available at: https://www.fda.gov/media/155050/download. Accessed 22 December 2021.
        116. U.S. Food and Drug Administration. Important Paxlovid™ EUA Dispensing Information for Patients with Moderate Renal Impairment. Available at: https://www.fda.gov/media/155072/download. Accessed 22 December 2021.
        117. Painter WP, Holman W, Bush JA, et al. Human Safety, Tolerability, and Pharmacokinetics of Molnupiravir, a Novel Broad-Spectrum Oral Antiviral Agent with Activity Against SARS-CoV-2. Antimicrob Agents Chemother 2021; 65(5): e02428-20.
        118. 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 2021: Available at: https://doi.org/10.1056/nejmoa2116044 [Epub ahead of print 16 December 2021].
        119. Fischer WA, 2nd, 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://doi.org/10.1126/scitranslmed.abl7430 [Epub ahead of print 23 December 2021].
        120. U.S. Food and Drug Administration. Fact Sheet for Patients And Caregivers: Emergency Use Authorization (EUA) Of Molnupiravir For Coronavirus Disease 2019 (COVID-19). Available at: https://www.fda.gov/media/155055/download. Accessed 28 December 2021.
        121. Lu H. Efficacy and Safety of Darunavir and Cobicistat for Treatment of Pneumonia Caused by 2019-nCoV (DACO-nCoV). Available at: https://clinicaltrials.gov/ct2/show/NCT04252274?id=NCT04252274.
        122. Chan JF, Chan KH, Kao RY, et al. Broad-spectrum antivirals for the emerging Middle East respiratory syndrome coronavirus. J Infect 2013; 67(6): 606-16.
        123. Kim JW, Kim EJ, Kwon HH, et al. Lopinavir-ritonavir versus hydroxychloroquine for viral clearance and clinical improvement in patients with mild to moderate coronavirus disease 2019. Korean J Intern Med 2020.
        124. Zhu Z, Lu Z, Xu T, et al. Arbidol monotherapy is superior to lopinavir/ritonavir in treating COVID-19. J Infect 2020; 81(1): e21-e3.
        125. Davoudi-Monfared E, Rahmani H, Khalili H, et al. Efficacy and safety of interferon beta-1a in treatment of severe COVID-19: A randomized clinical trial. medRxiv 2020.
        126. Holland HK, Bashey A, Morris L, E, Solomon S, Solh M. Use of Convalescent Plasma for COVID-19. Available at: https://clinicaltrials.gov/ct2/show/NCT04408040.
        127. Justman JE, Zech J. Convalescent Plasma for COVID-19 Close Contacts. Available at: https://clinicaltrials.gov/ct2/show/NCT04390503.
        128. Mohamed MA, El Ekiaby M, Ez-Eldin AM, Hussein AM, Selim AM. Clinical Study for Efficacy of Anti-Corona VS2 Immunoglobulins Prepared From COVID19 Convalescent Plasma Prepared by VIPS Mini-Pool IVIG Medical Devices in Prevention of SARS-CoV-2 Infection in High Risk Groups as Well as Treatment of Early Cases of COVID19 Patients. Available at: https://clinicaltrials.gov/ct2/show/NCT04383548.
        129. Johns Hopkins University, Jain SK, Gordon O. Human Convalescent Plasma for High Risk Children Exposed or Infected With SARS-CoV-2 (COVID-19). Available at: https://clinicaltrials.gov/ct2/show/NCT04377672.
        130. Shoham S. Efficacy and Safety Human Coronavirus Immune Plasma (HCIP) vs. Control (SARS-CoV-2 Non-immune Plasma) Among Adults Exposed to COVID-19 (CSSC-001). Available at: https://clinicaltrials.gov/ct2/show/NCT04323800.
        131. Bloch EM, Shoham S, Casadevall A, et al. Deployment of convalescent plasma for the prevention and treatment of COVID-19. J Clin Invest 2020; 130(6): 2757-65.
        132. The IMpact-RSV Study Group. Palivizumab, a Humanized Respiratory Syncytial Virus Monoclonal Antibody, Reduces Hospitalization From Respiratory Syncytial Virus Infection in High-risk Infants. Pediatrics 1998; 102(3): 531-7.
        133. Feltes TF, Cabalka AK, Meissner HC, et al. Palivizumab prophylaxis reduces hospitalization due to respiratory syncytial virus in young children with hemodynamically significant congenital heart disease. J Pediatr 2003; 143(4): 532-40.
        134. ter Meulen J, Bakker AB, van den Brink EN, et al. Human monoclonal antibody as prophylaxis for SARS coronavirus infection in ferrets. Lancet 2004; 363(9427): 2139-41.
        135. Stockman LJ, Bellamy R, Garner P. SARS: systematic review of treatment effects. PLoS Med 2006; 3(9): e343.
        136. Arabi YM, Shalhoub S, Mandourah Y, et al. Ribavirin and Interferon Therapy for Critically Ill Patients With Middle East Respiratory Syndrome: A Multicenter Observational Study. Clin Infect Dis 2019.
        137. Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 2020; 395(10223): 507-13.
        138. Ning Q, Han M. A Prospective/Retrospective, Randomized Controlled Clinical Study of Antiviral Therapy in the 2019-nCoV Pneumonia. Available at: https://clinicaltrials.gov/ct2/show/NCT04255017.
        139. Ning Q, Han M. A Randomized, Open, Controlled Clinical Study to Evaluate the Efficacy of ASC09F and Ritonavir for 2019-nCoV Pneumonia. Available at: https://clinicaltrials.gov/ct2/show/NCT04261270.
        140. Kongsaengdao S, Sawanpanyalert N. Various Combination of Protease Inhibitors, Oseltamivir, Favipiravir, and Hydroxychloroquine for Treatment of COVID19 : A Randomized Control Trial (THDMS-COVID19). Available at: https://clinicaltrials.gov/ct2/show/NCT04303299.
        141. Cao W, Liu X, Bai T, et al. High-Dose Intravenous Immunoglobulin as a Therapeutic Option for Deteriorating Patients With Coronavirus Disease 2019. Open Forum Infect Dis 2020; 7(3): ofaa102.
        142. Sakoulas G, Geriak M, Kullar R, et al. Intravenous Immunoglobulin (IVIG) Significantly Reduces Respiratory Morbidity in COVID-19 Pneumonia: A Prospective Randomized Trial. medRxiv 2020.
        143. Xie Y, Cao S, Dong H, et al. Effect of regular intravenous immunoglobulin therapy on prognosis of severe pneumonia in patients with COVID-19. J Infect 2020; 81(2): 318-56.
        144. Mohtadi N, Ghaysouri A, Shirazi S, et al. Recovery of severely ill COVID-19 patients by intravenous immunoglobulin (IVIG) treatment: A case series. Virology 2020; 548: 1-5.
        145. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020; 181(2): 271-80 e8.
        146. Coxib, traditional NTC, Bhala N, et al. Vascular and upper gastrointestinal effects of non-steroidal anti-inflammatory drugs: meta-analyses of individual participant data from randomised trials. Lancet 2013; 382(9894): 769-79.
        147. Zhang X, Donnan PT, Bell S, Guthrie B. Non-steroidal anti-inflammatory drug induced acute kidney injury in the community dwelling general population and people with chronic kidney disease: systematic review and meta-analysis. BMC Nephrol 2017; 18(1): 256.
        148. Voiriot G, Dury S, Parrot A, Mayaud C, Fartoukh M. Nonsteroidal antiinflammatory drugs may affect the presentation and course of community-acquired pneumonia. Chest 2011; 139(2): 387-94.
        149. Voiriot G, Philippot Q, Elabbadi A, Elbim C, Chalumeau M, Fartoukh M. Risks Related to the Use of Non-Steroidal Anti-Inflammatory Drugs in Community-Acquired Pneumonia in Adult and Pediatric Patients. J Clin Med 2019; 8(6).
        150. Mancia G, Rea F, Ludergnani M, Apolone G, Corrao G. Renin-Angiotensin-Aldosterone System Blockers and the Risk of Covid-19. N Engl J Med 2020; 382(25): 2431-40.
        151. World Health Organization. The use of non-steroidal anti-inflammatory drugs (NSAIDs) in patients with COVID-19. Available at: https://www.who.int/news-room/commentaries/detail/the-use-of-non-steroidal-anti-inflammatory-drugs-(nsaids)-in-patients-with-covid-19. Accessed 18 June 2020.
        152. Castro VM, Ross RA, McBride SM, Perlis RH. Identifying common pharmacotherapies associated with reduced COVID-19 morbidity using electronic health records. medRxiv 2020.
        153. Adnet F, Slama Schwok A. Efficacy of Addition of Naproxen in the Treatment of Critically Ill Patients Hospitalized for COVID-19 Infection (ENACOVID). Available at: https://clinicaltrials.gov/ct2/show/NCT04325633.
        154. Beale R, Mazibuko N, Farrell C, King's College London. LIBERATE Trial in COVID-19 (LIBERATE). Available at: https://clinicaltrials.gov/ct2/show/NCT04334629.
        155. Vaduganathan M, Vardeny O, Michel T, McMurray JJV, Pfeffer MA, Solomon SD. Renin-Angiotensin-Aldosterone System Inhibitors in Patients with Covid-19. N Engl J Med 2020; 382(17): 1653-9.
        156. Ferrario CM, Jessup J, Chappell MC, et al. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2. Circulation 2005; 111(20): 2605-10.
        157. Ishiyama Y, Gallagher PE, Averill DB, Tallant EA, Brosnihan KB, Ferrario CM. Upregulation of angiotensin-converting enzyme 2 after myocardial infarction by blockade of angiotensin II receptors. Hypertension 2004; 43(5): 970-6.
        158. Dijkman R, Jebbink MF, Deijs M, et al. Replication-dependent downregulation of cellular angiotensin-converting enzyme 2 protein expression by human coronavirus NL63. J Gen Virol 2012; 93(Pt 9): 1924-9.
        159. Sodhi CP, Wohlford-Lenane C, Yamaguchi Y, et al. Attenuation of pulmonary ACE2 activity impairs inactivation of des-Arg(9) bradykinin/BKB1R axis and facilitates LPS-induced neutrophil infiltration. Am J Physiol Lung Cell Mol Physiol 2018; 314(1): L17-L31.
        160. Zhang P, Zhu L, Cai J, et al. Association of Inpatient Use of Angiotensin-Converting Enzyme Inhibitors and Angiotensin II Receptor Blockers With Mortality Among Patients With Hypertension Hospitalized With COVID-19. Circ Res 2020; 126(12): 1671-81.
        161. Li J, Wang X, Chen J, Zhang H, Deng A. Association of Renin-Angiotensin System Inhibitors With Severity or Risk of Death in Patients With Hypertension Hospitalized for Coronavirus Disease 2019 (COVID-19) Infection in Wuhan, China. JAMA Cardiol 2020.
        162. Bean DM, Kraljevic Z, Searle T, et al. Angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers are not associated with severe COVID-19 infection in a multi-site UK acute hospital trust. Eur J Heart Fail 2020; 22(6): 967-74.
        163. Tedeschi S, Giannella M, Bartoletti M, et al. Clinical Impact of Renin-angiotensin System Inhibitors on In-hospital Mortality of Patients With Hypertension Hospitalized for Coronavirus Disease 2019. Clin Infect Dis 2020; 71(15): 899-901.
        164. Reynolds HR, Adhikari S, Pulgarin C, et al. Renin-Angiotensin-Aldosterone System Inhibitors and Risk of Covid-19. N Engl J Med 2020; 382(25): 2441-8.
        165. Fosbol EL, Butt JH, Ostergaard L, et al. Association of Angiotensin-Converting Enzyme Inhibitor or Angiotensin Receptor Blocker Use With COVID-19 Diagnosis and Mortality. JAMA 2020.
        166. Oussalah A, Gleye S, Clerc Urmes I, et al. Long-Term ACE Inhibitor/ARB Use Is Associated with Severe Renal Dysfunction and Acute Kidney Injury in Patients with severe COVID-19: Results from a Referral Center Cohort in the North East of France. Clin Infect Dis 2020.
        167. European Society of Cardiology. Position Statement of the ESC Council on Hypertension on ACE-Inhibitors and Angiotensin Receptor Blockers. Available at: https://www.escardio.org/Councils/Council-on-Hypertension-(CHT)/News/position-statement-of-the-esc-council-on-hypertension-on-ace-inhibitors-and-ang. Accessed 3 August 2020.
        168. Association AH. Patients taking ACE-i and ARBs who contract COVID-19 should continue treatment, unless otherwise advised by their physician. Available at: https://newsroom.heart.org/news/patients-taking-ace-i-and-arbs-who-contract-covid-19-should-continue-treatment-unless-otherwise-advised-by-their-physician. Accessed 3 August 2020.
        169. International Society of Hypertension. A statement from the International Society of Hypertension on COVID-19. Available at: https://ish-world.com/news/a/A-statement-from-the-International-Society-of-Hypertension-on-COVID-19. Accessed 3 August 2020.
        170. 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): 1054-62.
        171. Rojo JMC, Santos JMA, Núñez-Cortés JM, et al. Clinical characteristics of patients hospitalized with COVID-19 in Spain: results from the SEMI-COVID-19 Network. medRxiv 2020.
        172. Argenziano MG, Bruce SL, Slater CL, et al. Characterization and clinical course of 1000 Patients with COVID-19 in New York: retrospective case series. medRxiv 2020.
        173. Rawson TM, Moore LSP, Zhu N, et al. Bacterial and fungal co-infection in individuals with coronavirus: A rapid review to support COVID-19 antimicrobial prescribing. Clin Infect Dis 2020.
        174. Goyal P, Choi JJ, Pinheiro LC, et al. Clinical Characteristics of Covid-19 in New York City. N Engl J Med 2020; 382(24): 2372-4.
        175. Sepulveda J, Westblade LF, Whittier S, et al. Bacteremia and Blood Culture Utilization during COVID-19 Surge in New York City. Journal of clinical microbiology 2020; 58(8).
        176. Wei W, Ortwine JK, Mang NS, Joseph C, Hall BC, Prokesch BC. Limited Role for Antibiotics in COVID-19: Scarce Evidence of Bacterial Coinfection. Available at SSRN 3622388 2020.
        177. McCreary EK, Pogue JM. Coronavirus Disease 2019 Treatment: A Review of Early and Emerging Options. Open Forum Infect Dis 2020; 7(4): ofaa105.
        178. Clancy CJ, Nguyen MH. COVID-19, superinfections and antimicrobial development: What can we expect? Clin Infect Dis 2020.
        179. Furuta Y, Gowen BB, Takahashi K, Shiraki K, Smee DF, Barnard DL. Favipiravir (T-705), a novel viral RNA polymerase inhibitor. Antiviral Res 2013; 100(2): 446-54.
        180. Cai Q, Yang M, Liu D, et al. Experimental Treatment with Favipiravir for COVID-19: An Open-Label Control Study. Engineering (Beijing) 2020.
        181. Chen C, Huang J, Cheng Z, et al. Favipiravir versus arbidol for COVID-19: a randomized clinical trial. MedRxiv 2020.
        182. Lou Y, Liu L, Qiu Y. Clinical Outcomes and Plasma Concentrations of Baloxavir Marboxil and Favipiravir in COVID-19 Patients: an Exploratory Randomized, Controlled Trial. medRxiv 2020.
        183. 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-e31.
        184. Chatham WW, Kendrach A, University of Alabama at Birmingham. Early Identification and Treatment of Cytokine Storm Syndrome in Covid-19. Available at: https://www.clinicaltrials.gov/ct2/show/NCT04362111.
        185. Ucciferri C, Auricchio A, Di Nicola M, et al. Canakinumab in a subgroup of patients with COVID-19. Lancet Rheumatol 2020; 2(8): e457-ee8.
        186. Novartis Pharamaceuticals. Study of Efficacy and Safety of Canakinumab Treatment for CRS in Participants With COVID-19-induced Pneumonia (CAN-COVID). Available at: https://clinicaltrials.gov/ct2/show/NCT04362813.
        187. National Institute of Allergy and Infectious Diseases (NIAID). Adaptive COVID-19 Treatment Trial 2 (ACTT-2). Available at: https://clinicaltrials.gov/ct2/show/NCT04401579.
        188. Hall F. mulTi-Arm Therapeutic Study in Pre-ICu Patients Admitted With Covid-19 - Repurposed Drugs (TACTIC-R) (TACTIC-R). Available at: https://clinicaltrials.gov/ct2/show/NCT04390464.
        189. Eli Lilly and Company. A Study of Baricitinib (LY3009104) in Participants With COVID-19 (COV-BARRIER). Available at: https://clinicaltrials.gov/ct2/show/NCT04421027.
        190. Lenz H-J, University of Southern California. Baricitinib, Placebo and Antiviral Therapy for the Treatment of Patients With Moderate and Severe COVID-19. Available at: https://clinicaltrials.gov/ct2/show/NCT04373044.
        191. Menichetti F, Ospedaliero A. Baricitinib Compared to Standard Therapy in Patients With COVID-19 (BARICIVID-19). Available at: https://clinicaltrials.gov/ct2/show/NCT04393051.
        192. 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-e73.
        193. Durrant C, Ahmed O, Humanigen, Inc. Phase 3 Study to Evaluate Efficacy and Safety of Lenzilumab in Patients With COVID-19. Available at: https://clinicaltrials.gov/ct2/show/NCT04351152.
        194. Xu C, Kumar P, Siegel M, et al. Study of TJ003234 (Anti-GM-CSF Monoclonal Antibody) in Subjects With Severe Coronavirus Disease 2019 (COVID-19). Available at: https://www.clinicaltrials.gov/ct2/show/NCT04341116.
        195. Dagna L, De Luca G, Ospedale San Raffaele. Mavrilimumab in Severe COVID-19 Pneumonia and Hyper-inflammation (COMBAT-19) (COMBAT-19). Available at: https://clinicaltrials.gov/ct2/show/NCT04397497.
        196. Bergeron-Lafaurie A, Azoulay E, Peffault de Latour R, Assistance Publique - Hôpitaux de Paris. CORIMUNO19-ECU: Trial Evaluating Efficacy and Safety of Eculizumab (Soliris) in Patients With COVID-19 Infection, Nested in the CORIMUNO-19 Cohort (CORIMUNO19-ECU). Available at: https://clinicaltrials.gov/ct2/show/NCT04346797.
        197. Dong Y, Mo X, Hu Y, et al. Epidemiology of COVID-19 Among Children in China. Pediatrics 2020; 145(6).
        198. Kim L, Whitaker M, O'Halloran A, et al. Hospitalization Rates and Characteristics of Children Aged <18 Years Hospitalized with Laboratory-Confirmed COVID-19 - COVID-NET, 14 States, March 1-July 25, 2020. MMWR Morb Mortal Wkly Rep 2020; 69(32): 1081-8.
        199. Shekerdemian LS, Mahmood NR, Wolfe KK, et al. Characteristics and Outcomes of Children With Coronavirus Disease 2019 (COVID-19) Infection Admitted to US and Canadian Pediatric Intensive Care Units. JAMA Pediatr 2020.
        200. National Institute of Allergy and Infectious Diseases (NIAID). Adaptive COVID-19 Treatment Trial (ACTT). Available at: https://clinicaltrials.gov/ct2/show/NCT04280705.
        201. Gilead Sciences, Inc. Study to Evaluate the Safety and Antiviral Activity of Remdesivir (GS-5734™) in Participants With Severe Coronavirus Disease (COVID-19). Available at: https://clinicaltrials.gov/ct2/show/NCT04292899.
        202. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the Treatment of Covid-19 - Preliminary Report. N Engl J Med 2020.
        203. U.S. Food and Drug Administration. Letter of FDA Emergency Use Authorization to Ashley Rhodes dated May 1, 2020. In: Rhoades A, Gilead Sciences, Inc., 2020.
        204. Belhadjer Z, Méot M, Bajolle F, et al. Acute heart failure in multisystem inflammatory syndrome in children (MIS-C) in the context of global SARS-CoV-2 pandemic. Circulation 2020; 142(5): 429-36.
        205. Deza Leon MP, Redzepi A, McGrath E, et al. COVID-19-Associated Pediatric Multisystem Inflammatory Syndrome. J Pediatric Infect Dis Soc 2020; 9(3): 407-8.
        206. Riphagen S, Gomez X, Gonzalez-Martinez C, Wilkinson N, Theocharis P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet (London, England) 2020; 395(10237): 1607-8.
        207. 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. The Lancet 2020; 395(10239): 1771-8.
        208. Blondiaux E, Parisot P, Redheuil A, et al. Cardiac MRI of Children with Multisystem Inflammatory Syndrome (MIS-C) Associated with COVID-19: Case Series. Radiology 2020: 202288.
        209. Greene AG, Saleh M, Roseman E, Sinert R. Toxic shock-like syndrome and COVID-19: A case report of multisystem inflammatory syndrome in children (MIS-C). Am J Emerg Med 2020.
        210. Pouletty M, Borocco C, Ouldali N, et al. Paediatric multisystem inflammatory syndrome temporally associated with SARS-CoV-2 mimicking Kawasaki disease (Kawa-COVID-19): a multicentre cohort. Ann Rheum Dis 2020; 79(8): 999-1006.
        211. Chiotos K, Bassiri H, Behrens EM, et al. Multisystem Inflammatory Syndrome in Children During the Coronavirus 2019 Pandemic: A Case Series. J Pediatric Infect Dis Soc 2020; 9(3): 393-8.
        212. Cabrero-Hernandez M, Garcia-Salido A, Leoz-Gordillo I, et al. Severe SARS-CoV-2 Infection in Children With Suspected Acute Abdomen: A Case Series From a Tertiary Hospital in Spain. Pediatr Infect Dis J 2020; 39(8): e195-e8.
        213. Feldstein LR, Rose EB, Horwitz SM, et al. Multisystem Inflammatory Syndrome in U.S. Children and Adolescents. N Engl J Med 2020; 383(4): 334-46.
        214. Dufort EM, Koumans EH, Chow EJ, et al. Multisystem Inflammatory Syndrome in Children in New York State. New England Journal of Medicine 2020.
        215. Whittaker E, Bamford A, Kenny J, et al. Clinical Characteristics of 58 Children With a Pediatric Inflammatory Multisystem Syndrome Temporally Associated With SARS-CoV-2. JAMA 2020; 324(3): 259-69.
        216. 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): 1074-80.
        217. Centers for Disease Control and Prevention. Multisystem Inflammatory Syndrome in Children (MIS-C) Associated with Coronavirus Disease 2019 (COVID-19). Available at: https://emergency.cdc.gov/han/2020/han00432.asp. Accessed 24 May 2020.
        218. Royal College of Paediatrics and Child Health. Guidance: Paediatric multisystem inflammatory syndrome temporally associated with COVID-19, 2020.
        219. Rowley AH. Understanding SARS-CoV-2-related multisystem inflammatory syndrome in children. Nat Rev Immunol 2020; 20(8): 453-4.
        220. Diorio C, Henrickson SE, Vella LA, et al. Multisystem inflammatory syndrome in children and COVID-19 are distinct presentations of SARS-CoV-2. J Clin Invest 2020.
        221. Kalil AC. Treating COVID-19-Off-Label Drug Use, Compassionate Use, and Randomized Clinical Trials During Pandemics. JAMA 2020.
        222. Bauchner H, Golub RM, Zylke J. Editorial Concern-Possible Reporting of the Same Patients With COVID-19 in Different Reports. JAMA 2020.
        223. Zhao R, Wang H, Wang X, Feng F. Steroid therapy and the risk of osteonecrosis in SARS patients: a dose-response meta-analysis. Osteoporos Int 2017; 28(3): 1027-34.
        224. Calain P. The Ebola clinical trials: a precedent for research ethics in disasters. J Med Ethics 2018; 44(1): 3-8.
        225. Cook D, Burns K, Finfer S, et al. Clinical research ethics for critically ill patients: a pandemic proposal. Crit Care Med 2010; 38(4 Suppl): e138-42.
        226. Fowler RA, Webb SA, Rowan KM, et al. Early observational research and registries during the 2009-2010 influenza A pandemic. Crit Care Med 2010; 38(4 Suppl): e120-32.
        227. Iwashyna TJ, McPeake J. Choosing outcomes for clinical trials: a pragmatic perspective. Curr Opin Crit Care 2018; 24(5): 428-33.

Supplementary Information

Supplementary Materials (PDF)

This website uses cookies

We use cookies to ensure that we give you the best experience on our website. Cookies facilitate the functioning of this site including a member login and personalized experience. Cookies are also used to generate analytics to improve this site as well as enable social media functionality.