Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Crit Care Med. Jul 9, 2021; 10(4): 81-101
Published online Jul 9, 2021. doi: 10.5492/wjccm.v10.i4.81
What we learned in the past year in managing our COVID-19 patients in intensive care units?
Jain Nitesh, Department of Medicine, Mayo Clinic Health System, Mankato, MN 56001, United States
Rahul Kashyap, Department of Anesthesiology, Mayo Clinic, Rochester, MN 55905, United States
Salim R Surani, Department of Medicine, Texas A&M University, Corpus Christi, TX 78404, United States
ORCID number: Jain Nitesh (0000-0003-0139-3515); Rahul Kashyap (0000-0002-4383-3411); Salim R Surani (0000-0001-7105-4266).
Author contributions: Surani SR and Kashyap R have contributed to the idea, review of data and manuscript; Nitesh J has also been involved in data search and initial write-up.
Conflict-of-interest statement: There are nothing to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Salim R Surani, FCCP, MD, Professor, Department of Medicine, Texas A&M University, 701 Ayers, Corpus Christi, TX 78404, United States. srsurani@gmail.com
Received: February 16, 2021
Peer-review started: February 16, 2021
First decision: March 31, 2021
Revised: April 7, 2021
Accepted: May 17, 2021
Article in press: May 17, 2021
Published online: July 9, 2021

Abstract

Coronavirus disease 2019 is a pandemic, was first recognized at Wuhan province, China in December 2019. The disease spread quickly across the globe, spreading stealthily from human to human through both symptomatic and asymptomatic individuals. A multisystem disease which appears to primarily spread via bio aerosols, it has exhibited a wide clinical spectrum involving multiple organ systems with the respiratory system pathology being the prime cause of morbidity and mortality. Initially unleashing a huge destructive trail at Wuhan China, Lombardy Italy and New York City, it has now spread to all parts of the globe and has actively thrived and mutated into new forms. Health care systems and Governments responded initially with panic, with containment measures giving way to mitigation strategies. The global medical and scientific community has come together and responded to this huge challenge. Professional medical societies quickly laid out “expert” guidelines which were conservative in their approach. Many drugs were re formulated and tested quickly with the help of national and international collaborative groups, helping carve out effective treatment strategies and help build a good scientific foundation for evidence-based medicine. Out of the darkness of chaos, we now have an orderly approach to manage this disease both from a public health preventive and therapeutic standpoint. With preventive measures such as masking and social distancing to the development of highly effective and potent vaccines, the public health success of such measures has been tempered by behavioral responses and resource mobilization. From a therapy standpoint, we now have drugs that were promising but now proven ineffective, and those that are effective when given early during viral pathogenesis or later when immune dysregulation has established, and the goal is to help reign in the destructive cascade. It has been a fascinating journey for mankind and our work here recapitulates the evolution of various aspects of critical care and other inpatient practices which continue to evolve.

Key Words: COVID-19, Respiratory support, Renal replacement therapy, Extracorporeal membrane oxygenator, Medications, Therapeutics

Core Tip: Severe acute respiratory syndrome coronavirus 2 transmission and the inpatient therapeutic management of coronavirus disease 2019 has been subject of immense research in the past one year. Our knowledge and understanding of the virus and the treatment of the disease continue to evolve. We attempt to summarize the progress made in a concise but comprehensive manner along with our insights into future directions.



INTRODUCTION

Coronavirus disease 2019 (COVID-19) is a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) first reported and widely believed to have originated at Wuhan in the Hubei province, China in late December 2019[1]. It started as a Zoonotic disease and gained a foothold in human population by person-to-person transmission, having evolved into a destructive pandemic infecting more than 100 million people and has caused more than 2.2 Million deaths till date[1,2].

A member of Beta coronaviruses, which includes SARS-CoV-1 and Middle East respiratory syndrome coronavirus (MERS-CoV) which have caused localized epidemics in the Asian continent, the SARS-CoV-2 rapidly spread across the globe and has now survived and evolved with mutants due to its ability to stealthily spread by airborne transmission, ability to survive in varying environmental conditions, causing asymptomatic or mild infection in humans with transmission characterized by the ability to infect early on during the prodromal phase of illness, aided generously by “super spreaders”[1,3,4].

The management of the disease has evolved with early conservative guidelines from experts to evidence-based recommendations which continue to evolve every day touching all aspects of care from the use of respiratory assist devices, medication including repurposed drugs, novel and controversial therapies as well as delivery of our critical care services. Here we attempt to capture some of these changes and present the current state of evidence of some of these therapies and services used in the management of COVID-19[5].

INFECTIVITY AND TRANSMISSION CHARACTERISTICS

Since the beginning of the pandemic SARS-CoV-2 duration of shedding, infectivity, and mechanism of transmission of infection have been very keenly studied as they have practical implications. We now have better knowledge and understanding of these characteristics. The viral RNA has been detected by reverse transcriptase-polymerase chain reaction testing from the upper respiratory tract for a mean of 17 d with a maximal duration of 83 d. Likewise, from the lower respiratory tract, the viral RNA has been detected for a mean duration of 17.2 d with a maximal duration of only 35 d. However more importantly the live virus has not been cultured beyond the 9th day of symptom in any study to date. Hence the maximal infectivity is likely in the first week from symptom onset and tapers off subsequently[6].

Respiratory transmission is now considered the predominant mode of infection. Droplets are large particles typically more than 5 microns which are heavier and drop within 6 feet, whereas aerosols are smaller than 5 microns and post evaporation remain suspended like pollens in the air having the ability to travel longer distances[7]. Our current understanding is that the virus is shed as particles across a wide range of sizes[8,9]. A longer duration, closer proximity, forced exhalation of air from a patient with high viral load is now considered necessary for cross-infection to occur with SARS-CoV-2[8]. Logically a “full high-level barrier protection” with Personal Protective Equipment (PPE), N95 mask & Negative pressure room may therefore be necessary when managing a highly symptomatic patient who is excessively coughing, is on high flow oxygen, noninvasive ventilation (NIV), Mechanical ventilator, is undergoing Bronchoscopy or has a Tracheostomy. In all these situations, a large amount of air is being mobilized across the mucosa covered with the virus, enhancing the possibility of viral aerosolization & infection[8]. In fact if the combination of “full barrier precautions” and adherence to clinical practice guidelines are strict, then the likelihood of infection with SARS-CoV-2 in clinical care areas for staff is substantially reduced or insignificant[10].

The role of respiratory assist devices and maneuvers in the pandemic

COVID-19 is a disease that affects multiple organ systems but primarily and disproportionately affects the Respiratory system. Early in the pandemic stemming from the Chinese experience, COVID-19 patients were intubated early when needing more than 5-6 L/min oxygen to avoid aerosolization of SARS-CoV-2 infection to staff and due to the anticipation, that these patients would deteriorate rapidly with the attendant risk of substantial hypoxia during intubation. However it is now apparent that such aggressive measures are not warranted as it places substantial burden on the need for critical care resources[11]. Although not proven to be causative, the early surge of COVID-19 cases in New York city and Italy in early 2020 was notable for very high mortality noted in intubated patients[12,13].

Adult respiratory distress syndrome (ARDS) is the dominant respiratory clinical syndrome seen in COVID-19 patients[13,14] with histopathology primarily characterized by diffuse alveolar damage very similar to SARS-CoV-1 and MERS-CoV infections[15]. ARDS related lung injury and Respiratory mechanics in COVID-19 appear to be similar to non-COVID-19 ARDS; nevertheless substantial controversy exists regarding management in literature which is intriguing and is addressed in our discussion[11,13,14].

Oxygen supplementation and NIV

It is generally accepted that low flow oxygen with a simple face mask or Cannula is used for supplemental oxygen as the first line of support when SaO2 is less than 88%. The next line of oxygen supplementation is through high flow nasal cannula (HFNC). It provides oxygen at a very high flow rates (40-80 L/min). This oxygen also is heated and humidified to simulate physiological conditions in the airway promoting patient comfort and tolerance[16]. HFNC is essentially a flow generator helping with muco- ciliary clearance in the airway and improves the Ventilatory function of the lung by providing low levels of functional “Positive end expiratory Pressure (PEEP)” in the respiratory tract[17]. A type 1 surgical mask can substantially reduce particulate aerosol contamination from nasal devices when placed over them[18]. The dispersion of aerosolized particles is higher than a simple mask for HFNC but much less when compared to NIV in simulated experiments[19,20].

NIV such as continuous positive airway pressure (CPAP) and Bi-level alveolar positive airway pressure (BIPAP) are the next line which provides pressure targeted ventilation. CPAP has traditionally been used in acute cardiogenic pulmonary edema by increasing functional residual capacity and therefore oxygenation and compliance. BIPAP in addition to the latter has also been used in acute exacerbation of the chronic obstructive pulmonary disease for counterbalancing inner PEEP with external PEEP and decreasing work of breathing by acting as an inhalation assist device[17]. Both modes of NIV have been traditionally used in obstructive sleep apnea and obesity hypoventilation syndrome[21]. CPAP and BIPAP must be used with a full-face mask to decrease the risk of aerosolization. BIPAP can also be used with a helmet mask (mostly available in Europe). They have been shown to have an acceptable level of aerosolization which can be further attenuated with the help of a well-fitting helmet mask[22].

In general, HFNC is preferred over NIV. HFNC is much more comfortable for the patient as it allows for speech, eating/drinking as well as comfort[17]. But NIV may be preferred in patients who have acute chronic obstructive pulmonary disease (COPD) exacerbation with hypercarbia, acute pulmonary edema and those who have sleep disordered breathing.

Evidence from non-COVID-19 literature for HFNC and NIV

In the FLORAL trial involving hypercpaneic patients with acute hypoxemic respiratory failure, HFNC was shown to decrease intubation rate which was statistically significant in a sub-group of patients with Pao2/Fio2 < 200 when compared to non-rebreather mask (≥ 10 L/min) or NIV. Mortality also favored the HFNC group at 90 d when compared to the other two groups in this study[23].

In another study, HFNC was non-inferior to NIV for preventing reintubation and post-extubation respiratory failure in high-risk adults[24].

In another randomised controlled trial involving high-risk adults, the combined use of HFNC and NIV prevented more extubation failures than HFNC alone[25] suggesting that the two modalities can complement each other.

In the LUNG SAFE study, about 15% of ARDS patients were treated with NIV. Failure of NIV was increasingly common with increasing severity of ARDS but mortality was especially higher in patients who had Pao2/Fio2 lower than 150 mmHg[26] and hence should be avoided in this subgroup of Moderate to Severe ARDS Patients.

In a systematic review and meta-analysis involving 25 studies and 3804 patients, the use of both helmet and face mask NIV was associated with decreased mortality and endotracheal intubation compared to standard oxygen therapy[27]. However, in sensitivity analysis excluding studies which included COPD exacerbation and congestive heart failure exacerbation, the observed benefit on mortality was not noted. The beneficial effect on mortality was also less certain with patients who had severe ARDS.

Evidence from COVID-19 literature for HFNC and NIV

Good quality data is lacking but some moderate sized retrospective observational studies have been published.

In Lombardy Italy, about 350 of 3988 patients with COVID-19 Pneumonia were treated with NIV, of which 50 percent required intubation. The mortality of the latter group was similar to patients who were intubated on admission to the intensive care units (ICU)[28].

In one published Italian retrospective observational study of 670 patients, the rate of intubation and adjusted mortality did not vary in patients who were treated with High flow oxygen, CPAP and BIPAP[29].

In a study of 110 patients who received non-invasive ventilation via helmet for two days, followed by the high flow nasal oxygen therapy or high flow oxygen alone, there was no difference in the ventilator free days at 28 d between NIV and high flow, but patient in the helmet NIV group had decrease in intubation and mechanical ventilation free days, with the P value of 0.03[30].

In a systematic review and meta-analysis of non-randomized cohort studies involving about 1897 critically ill patients, there was no statistically detectable difference on all-cause mortality between patients undergoing intubation without vs with a prior trial of HFNC/NIV [eight studies, 1128 deaths; 48.9% vs 42.5%; risk ratio (RR) 1.11, 95% confidence interval (CI): 0.99-1.25, P = 0.08][31].

Monitoring of patients on HFNC and NIV

Patients need to be carefully monitored when on supplemental oxygen devices like high flow or NIV. Intubation should not be withheld when appropriate criteria are met. It is estimated that about 20%-25% of patients can avoid intubation and help preserve Critical resources during the pandemic[17]. Further evidence is needed.

Early vs late intubation

The concept of early vs late intubation in COVID-19 pneumonia is controversial which has elicited a fascinating Pros-Con debate[32,33].

Early on, some professional organizations like the Royal College of Anesthetists & Intensive Care Society recommended early intubation to prevent the risk of high environmental contamination with other oxygenation and ventilatory adjuncts like NIV/HFNC[32].Others like the Society of Critical Care Medicine recommended careful monitoring with NIV/HFNC and intubation when the latter failed[34].

A failed NIV followed by intubation can be associated with an increased risk of complications during intubation like hypotension, desaturation, and aspiration with associated increased risk of mortality[35]. While some studies in non-COVID-19 hypoxemic respiratory failure show increased mortality with delayed intubation[35,36] others in COVID-19 hypoxemic respiratory failure showed no such increased mortality[13].

Proponents of early mechanical ventilation emphasize the possibility of “Patient self-inflicted Lung injury (P-SILI) “in the non-intubated critically ill patient with acute hypoxemic respiratory failure which is a collective term for the high minute ventilation, a high respiratory drive of the ARDS patient worsening the preexisting lung injury with increased vascular permeability along with local and global lung over distension[37]. P-SILI in a spontaneously breathing patient is akin to ventilator-induced lung injury in a mechanically ventilated patient[33] and is caused by high pleural pressures and trans pulmonary pressure swings. Lung protective ventilatory strategies using mechanical ventilation along with deep sedation and/or neuromuscular paralysis can prevent P-SILI[37,38]. The endotracheal tube helps gain good control over an unstable airway and regulate oxygen, pressure, and volume[39].

Opponents of early and liberal Mechanical ventilation offer many valid reasons. The concept of P-SILI is relatively new and the evidence supporting it is not very robust[33]. Mechanical ventilation brings along with it a host of complications like delirium secondary to sedation, hemodynamic instability secondary to decreased sympathetic drive and positive pressure ventilation, increased risk of infection, immobilization with increased risk of thromboembolism, neuromuscular paralysis, post-intensive care syndrome with its attendant physical and neurocognitive dysfunction[32]. Intubation and mechanical ventilation are associated with one of the highest risks of aerosolization[40] and for the patient, there is risk of procedure related hypotension, hypoxemia, cardiac arrest, and other complications[41]. During a pandemic conserving critical resources and their judicious use is important and intubating every patient with hypoxemic respiratory failure is going to be unethical[42,43].

No randomized control studies have been published on this topic. The definition of early vs late intubation is variable across studies. A few small single-center retrospective studies have reported variable outcomes for delayed vs early endotracheal intubation[44-47] with one study reporting worser mortality outcomes for delayed intubation and other three being equivocal.

In a systematic review and meta-analysis of non-randomized cohort studies involving about 9000 critically ill patients compared early (less than 24 h after ICU admission) vs late (more than 24 h after ICU admission) intubation found no difference in all-cause mortality(3981 deaths; 45.4% vs 39.1%; RR 1.07, 95%CI: 0.99-1.15, P = 0.08), duration of mechanical ventilation (1892 patients; MD - 0.58 d, 95%CI: 3.06-1.89 d, P = 0.65), ICU length of stay and renal replacement therapy (RRT)[31].

Due to limited data, the question apart from some lively, elegant and animated discussions between experts is probably unsettled[33,48].

Nebulization

SARS-CoV-2 virus transmission occurs predominantly through close contact, poor ventilated environment in a susceptible host via droplets/aerosols and less likely through fomites[6,7,9].Transmission via bio aerosols from medical procedures like Nebulization and Tracheostomy has been a very valid concern as discussed earlier[49].

As per the Global initiative for asthma & The Australian National Asthma Council, the recommendation is to use nebulization therapy only if unavoidable[50,51]. On the contrary, the British National Institute of Health Care and Excellence recommends that patients with COVID-19 can continue using nebulization therapy[52]. Such contrary guidelines and recommendations have sowed doubts in the minds of patients and professional health care practitioners. It is indicative of the fact that the evidence base for these contrary recommendations is not very strong.

Although a continuation of inhalational treatment for chronic respiratory diseases has been universally recommended[51], the optimal mode is less certain. Inhalers have been recommended as they seem to generate fewer aerosols, the drug is contained in the container and less likely to be contaminated by infectious particles, and they also have a low emitted dose[49]. However, either via normal exhalation or cough (determined by drug formulation characteristics) induced by the inhaled medication, inhalers can produce exhaled bio aerosols and hence they do not seem to be superior to nebulizer therapy[49].

Theoretically, nebulizer therapy produces an aerosol of the medication in the nebulizer container and hence should not produce infected aerosols unless the container or medication gets contaminated[49]. An aerosol droplet coming in contact with an infected mucous membrane, like in the lung stops being airborne and hence is no longer an aerosol[53]. Hence good hygiene precautions undertaken while using the nebulizer and while loading the medication should prevent the spread of infection by aerosolization[49,53]. Besides, other precautions to prevent bio aerosolization have been proposed such as the use of viral filters in the circuit of nebulizers/ventilators, use of vibratory mesh nebulizers which separate medication from patient interface including circuits, and good provider/patient hygiene and using mouthpiece with handheld devices[53]. Universally full barrier precautions as discussed earlier should be practiced to limit infection.

Bronchoscopy

At the beginning of the pandemic, many Pulmonary/Bronchology societies made recommendations for COVID-19, but were limited by generalizations, lack of exhaustiveness, and clear guidance was not available due to the novelty of the disease; extrapolation from previous coronavirus pandemics was required[54]. Almost all societies recommended deferring bronchoscopy in non-urgent cases, observing full barrier precautions when performing bronchoscopies, restricting the number of personnel who could be participating in the procedure, limit aerosol producing procedures like nebulization, use of atomizers and jet ventilation[55]. Peri procedurally recommendations included using sedation (or even paralytics when feasible) to avoid coughing, avoiding high flow and high shearing maneuvers, all intended to limit aerosolization. Flexible bronchoscopy is encouraged and rigid bronchoscopy is discouraged with post-procedure recommendations lacking consensus[54]. To avoid cross-contamination or accidental transmission, single-use flexible bronchoscopes are encouraged[54].The patient can wear a mask and a slot can be made for introducing the bronchoscope[54,55].

Certain acceptable indications for bronchoscopy in COVID-19 times include but not exhaustively, symptomatic airway stenosis, symptomatic hemoptysis, migrated stent, therapeutic aspiration of obstructive symptomatic secretions or masses, diagnosis of secondary infections in intubated COVID-19 patients, diagnosis of cancer, and diagnosis of infection in immunocompromised patients[55].

In a single-center, where 241 bronchoscopies were performed on 107 COVID-19 patients, 54 patients (50.5%) had Broncho Alveolar Lavage (BAL) with 35 patients (65%) demonstrating a positive culture. About 1/3rd of intubated patients required bronchoscopy presumably due to thickened white gelatinous secretions (likely due to heated air with less humidification as was recommended by guidelines) or bloody secretions due to high use of anticoagulants. BAL cultures were more likely to be positive (65%) compared to tracheal cultures (45%). 6% of BAL cultures also grew a second organism. The study showed a high rate of secondary infection in COVID-19 patients above and beyond that was diagnosed with tracheal cultures, indicating that under treatment may be driving higher mortality[56].

In another single-center series of 93 intubated patients, 101 bronchoscopies were performed which did not show increased secondary infection when compared to non-covid ventilator associated pneumonia[57].

In general, bronchoscopy has not shown any definitive increase in transmission when proper precautions have been observed[56,57].

Tracheostomy

Tracheostomy has been widely used across the globe for COVID-19 management. Initially, expert guidelines were made available which were very conservative in their recommendations but now we have better evidence to guide our decisions[58]. Certain pertinent issues concerned with Tracheostomy are addressed here.

The Indications for tracheostomy have traditionally not been well defined, dependent on multiple factors and individual circumstances[59]. In the current COVID-19 times, tracheostomies have been performed early (less than 7 to 10 d after intubation) and for very liberal indications with critical care resource utilization as a goal commensurate with principles of “Disaster management”[60-62]. However, guidelines based on several critical considerations including virology of transmission and infectiousness of the patient recommended the timing to be past 10 d and when patients show clinical improvement[59]. This is because it is difficult to predict the clinical trajectory of ARDS patients with COVID-19. After the patient has navigated the first few days of Critical illness and shown clinical improvement, but anticipate prolonged mechanical ventilation, with reasonable pulmonary reserves, the FiO2 less than 40% and PEEP less than 8, then tracheostomy can be considered[59,60,63,64]. Given that there are advantages and disadvantages to both early and late tracheostomy, and with relatively proven non-inferiority, the timing of tracheostomy like in non-COVID-19 patients has to be individualized[61,63]. In practice, a systematic review and meta-analysis encompassing 462 COVID-19 patients revealed that 250 patients (71.5%) received tracheostomy 14 d after intubation, which is consistent with conventional practice[65].

Tracheostomy can be performed by the “open or surgical” method in the operating room or by “Percutaneous dilatation” at the patient bedside. Initially, the recommendation was to use the “Open or Surgical” method to minimize exposure to bio aerosol which is potentially more with the percutaneous method[59,64]. However, with diligent and appropriate use of “Full barrier” precautions including PPE with or without a negative pressure room, the increased risk to healthcare personnel has not materialized and the emphasis is now to optimally use available resources as both methods have been proven to be safe[59,62,64,65]. In a pooled analysis of 3060 tracheostomies, 55.7% were created by the open method and 43.4% were created by the percutaneous method[65].

Post-procedural management guidelines suggest to limit staff exposure to bio aerosols have been published and it has been demonstrated that this can be implemented successfully by training new staff members unfamiliar with tracheostomy care, thereby helping free critical ICU resources when necessary[59,62,64].

Post tracheostomy outcome data in COVID-19 patients are now available. In a pooled analysis, of 2890 mechanically ventilated patients 54.9% were reported to have been successfully weaned, of 2628 patients 34.9% were successfully decannulated, and of 2980 patients 513 patients (13.1%) had died[65].

Overall tracheostomy in COVID-19 patients has evolved from the early time of guidelines recommending “abundant caution” to now practice and outcomes which seem to be more consistent with “regular order”.

Convalescent plasma and monoclonal antibody

Convalescent plasma has been used to treat many infectious diseases in the past like Influenza, MERS-CoV, Ebola Virus, Influenza, etc., but efficacy and evidence are not firmly established[66,67]. The goal of such passive immunization is to neutralize the infectious organism with the help of naturally formed and passively transferred antibodies[66]. Novel neutralizing monoclonal antibodies (nabs) and nano antibodies have also come into play during the coronavirus pandemic[68].

SARS-CoV-2 virus enters the cell via the angiotensin-converting enzyme 2 (ACE2) receptors on the respiratory and gastrointestinal tract epithelium. The SARS–CoV-2 virus has an outer “S” glycoprotein, with S1 and S2 subunits. The S1 subunit has a receptor binding domain along with receptor binding motif, the latter attaches to the ACE2 receptor in the host, and there is a conformational change in the S protein leading to S2 fusing with the host cell wall membrane followed by internalization of the virus into the host cell. The SARS-CoV-2 antibody in the convalescent plasma/nabs can halt the virus from multiplying and establishing a foothold in the host by interfering with receptor attachment, inhibiting wall fusion after attachment, and preventing uncoating of the virus once inside the cytoplasm[68,69].

With COVID-19, convalescent plasma has been widely used from the early days of the pandemic on a compassionate basis with regulatory approval[70]. However; results from various studies have been inconsistent.

Analysis of large observational data and different Randomized control studies show that when plasma with low SARS-CoV-2 antibody titer or when used later in the disease trajectory or both results in lack of survival benefit, does not halt the progression of the disease or help with stabilization of symptoms[70-72]. COVID-19 patients with moderate to severe ARDS, especially intubated patients do not derive any benefit from convalescent plasma[70-73].

On the contrary, when the plasma has high antibody titer, and patients receive early on at symptom onset in the community or even during early hospitalization when patients have mild to moderate disease, it results in better survival, disease stabilization and halts the progression of the disease[70,73,74].

As per Food and Drug Administration (FDA), high titter convalescent plasma corresponds to a neutralizing antibody titer of ≥ 250 in the Broad Institute's neutralizing antibody assay, a signal-to-cutoff of ≥ 12 in the Ortho VITROS immunoglobulin G (IgG) assay, or a level of ≥ 1:2880 in the Mount Sinai COVID-19 ELISA IgG Antibody Test[75].

The role of passive immunization with convalescent plasma or Neutralizing antibodies is to inhibit viral replication early in the disease when the host does not have sufficient antibodies of its own. Once the infection is established, native antibodies are formed and inflammatory processes are at work, at which point the passively transfused antibodies are not helpful[76].

Similarly neutralizing Monoclonal antibodies like Bamlanivimab were found to help reduce viral load, and hospitalization in recently diagnosed mild to moderate COVID-19 disease as outpatient especially in patients with co-morbidities across age groups, especially in elderly, but not useful in hospitalized severely ill COVID-19 patients[77]. In the yet to be published Blaze-2 trial, Bamlanivimab used as a prophylaxis in nursing home and assisted care home residents were found to decrease symptoms and even have a survival advantage when compared to placebo[78]. And although peer review is pending, this appears to be a promising therapy when used in high-risk patients either as prophylaxis or early disease complementing the huge anticipated benefit of vaccine administration on a large scale.

The FDA has updated its Emergency use authorization on February 4, 2021 and now limits the use of high titer COVID-19 convalescent plasma only for the treatment of hospitalized patients with COVID-19 early in the disease course and to those hospitalized patients who have impaired humoral immunity and cannot produce an adequate antibody response[79].

The recovery trial has reported its findings in a preprint article on the use of high titer convalescent plasma in hospitalized patients which is yet to be peer reviewed[80]. 5795 patients were randomly allocated to receive convalescent plasma and 5763 to usual care alone. There was no significant difference in 28-d mortality between the two groups: 1398 (24%) of 5795 patients allocated convalescent plasma and 1408 (24%) of 5763 patients allocated usual care died within 28 d (RR 1.00; 95%CI: 0.93-1.07; P = 0.93). Similarly there was no change in the proportion of patients discharged from hospital, progression of patients not on mechanical ventilation towards intubation, successful cessation from mechanical ventilation or need for RRT. However, the mean number of days from symptom onset was 9, and therefore likely the plasma was not used early enough in the disease course.

Glucocorticoids

Glucocorticoids are one of the oldest, well known, inexpensive, immunomodulatory agents with wide ranging immunosuppressive, anti-inflammatory and anti-allergic effect. They also have a multitude of adverse effects as well[81]. It was therefore natural to test their effectiveness as a therapeutic agent for COVID-19, and although some of the earlier studies did not show any benefit, the “RECOVERY Trial” was the earliest well conducted randomized sontrol trial that showed survival benefit in severely ill patients needing supplemental oxygen and ventilation[82]. The latter study showed that there was mortality benefit with use of dexamethasone.

A total of 2104 patients were assigned to receive dexamethasone and 4321 to receive usual care[77].

Overall 17 percent relative reduction in mortality (22.9 vs 25.7 percent, RR 0.83, 95%CI: 0.75-0.93),

Patients on invasive mechanical ventilation or (ECMO) at baseline–36 percent relative reduction (29.3 vs 41.4 percent, RR 0.64, 95%CI: 0.51-0.81). Age-adjusted analysis suggested a 12.3 percent absolute mortality reduction.

Patients on noninvasive oxygen therapy (including NIV) at baseline–18 percent relative reduction (23.3 vs 26.2 percent, RR 0.82, 95%CI: 0.72-0.94). Age-adjusted analysis suggested a 4.1 percent absolute mortality reduction.

Currently as per a pooled meta-analysis, the use of glucocorticoids is estimated to cause 31 fewer deaths per 1000 [odds ratio (OR) 0.87, 95%CI: 0.77 to 0.98; risk difference 31 fewer per 1000, 95%CI: 55 fewer to 5 fewer], risk of mechanical ventilation is reduced by 28 per 1000 (OR 0.73, 0.58 to 0.92; risk difference 28 fewer per 1000, 45 fewer to 9 fewer), and duration of hospital stay is reduced by almost 1 d (mean difference -0.99 d, -1.36 to -0.64), all results estimated to be of moderate certainty[83].

With this the use of glucocorticoids became well established as standard of care for the treatment of severely ill COVID-19 patients needing supplemental oxygen and or ventilation. This has been followed by the question whether the standard 6 milligram Dexamethasone per day therapy which was used in the RECOVERY TRIAL is sufficient a dose or if there is an incremental benefit by dose increase? Also, another pertinent question is whether there is any benefit of targeting any other specific immune pathways.

While Randomized control data involving the inhibition of complement C5 inhibitor, raviluzumab has not been shown to be of benefit as per preliminary unpublished data[84], the role of Interleukin-6 inhibitor, tocilizumab has been quite intriguing.

Tocilizumab

Tocilizumab is an interleukin 6 receptor antagonist monoclonal antibody that has been used to treat patients with COVID-19 respiratory and organ failure targeting a key step in inflammatory mediated damage[68]. Early treatment data in observational and randomized control studies, not involving many critically ill patients and without Glucocorticoid use showed that Tocilizumab was safe but did not have any significant Clinical outcomes[85-87]. There were six small trials which did not show any significant benefit from Tocilizumab[88]. However, data from “STOP COVID”-a large observational study and “REMAP CAP”-A well designed open label international randomized control study consisting of 803 patients, suggest that “the early use of Tocilizumab on entry to ICU” may have important survival and other outcome benefits in the short term which was not seen in less sick patients studied in randomized control trials outside the ICU[85-87,89]. This was especially noted in patients who had ICU admission within 3 d of symptom onset[89] or had evidence of organ failure on admission to ICU[87]. Participants in the Randomized, Embedded, Multi-factorial, Adaptive Platform Trial for Community-Acquired Pneumonia (REMAP-CAP) study also had a relatively larger proportion of patients on glucocorticoids (more than 80%) compared to other studies[86,87]. In “REMAP-CAP” Tocilizumab (n = 353) and Sarilumab (n = 48) each reduced in-hospital mortality compared with standard of care (28 and 22 vs 36 percent; OR for hospital survival 1.64, 95%CI: 1.14-2.35 for Tocilizumab and 2.01, 95%CI: 1.18-4.1 for Sarilumab).

The Tocilizumab arm of RECOVERY TRIAL reported preliminary results which are undergoing peer review[88]. This was an open label randomized placebo-controlled trial in which 82% patients took glucocorticoids like dexamethasone. 2022 patients received tocilizumab and 2094 received standard of care. To be eligible for randomization, patients with COVID-19 were to have hypoxia (SpO2 < 92%) and C-reactive protein more than 75 mg/dL.

Of 596 (29%) patients in the Tocilizumab group and 694 (33%) patients in the usual care group died (RR 0.86; 95%CI: 0.77-0.96; P = 0.007) at 28 d, an absolute difference of 4%. This translates into Numbers Needed to Treat for saving one life of 25.

Tocilizumab also increased the probability of being discharged alive within 28 d from 47% to 54% (RR 1.23, 95%CI: 1.12-1.34, P < 0.0001).

Among patients not on invasive mechanical ventilation when entered into the trial, Tocilizumab significantly reduced the chance of progressing to invasive mechanical ventilation or death from 38% to 33% (RR 0.85, 95%CI: 0.78-0.93, P = 0.0005).

Allocation to Tocilizumab reduced the use of all forms of dialysis (5% vs 7%, RR 0.75, 95%CI: 0.59-0.96, P = 0.02).

Tocilizumab did not have any effect on the chance of successful cessation of invasive mechanical ventilation.

These benefits were seen in all patient subgroups, including those requiring oxygen via a simple face mask through to those requiring mechanical ventilators in an intensive care unit.

Tocilizumab is estimated to reduce the relative risk of death by 14% and reduced the time spent in hospital by 5 d when used for patients on oxygen and in addition to the corticosteroid dexamethasone[90].

Taken together data from all 8 trials, use of tocilizumab was associated with 13% proportional reduction in 28-d mortality (death RR 0.87, 95%CI: 0.79-0.96, P = 0.005). It is noteworthy that these mortality benefits were noted in the RECOVERY TRIAL only in patients receiving concomitant steroids.

In summary, it appears that in severely ill COVID-19 patients with hypoxia accompanied by hyper inflammatory state, the early concomitant use of glucocorticoids and Tocilizumab improves outcomes including survival, organ support and progression of disease, suggesting additive or synergistic effect with these two agents.

This beneficial data appears to be quite specific for Tocilizumab, as the numbers of patients with Sarilumab in REMAP-CAP study were few. Trials involving Sarilumab are in progress and results are expected in the future[88].

The United Kingdom government and Center for disease control have expeditiously approved the use of Tocilizumab based on data from REMAP-CAP and RECOVERY TRIALS[90,91]. Other government and Professional societies are expected to update their guidelines soon as well.

Remdesivir

Remdesivir is an inhibitor of “viral RNA dependent RNA polymerase” which inhibits SARS-COV-2 in vitro[92] but has not been shown to decrease viral load when compared to placebo[93]. It has been studied extensively in clinical trials and the findings are summarized below.

The outcome data has been measured using the multipoint ordinal scale with each number denoting a particular “clinical status” and the changes are measured and reported accordingly[92-94].

In the international, multicentric auditory consonant trigram test-1 study conducted by the National Institute of Allergy and Infectious Diseases and others, 541 patients were assigned to Remdesivir and 521 to placebo in a double-blind placebo-controlled trial; the study drug was given intravenously for 10 d. A significant number of patients had severe disease with SpO2 less than 94% by definition and requiring supplemental oxygen. It reported a primary outcome of improved median recovery time of 10 d compared to 15 d with placebo. There was a trend to improvement in mortality which was not statistically significant, 11.4% and 15.2% in two groups, respectively [hazard ratio (HR) 0.73; 95%CI: 0.52-1.03] by day 29. In sub-group analysis, there was mortality benefit noted in patients who were on simple low flow oxygen, (HR 0.30; 95%CI: 0.14-0.64). Remdesivir also showed shorter hospital length of stay, reduced disease progression, and lesser utilization of respiratory assist devices like oxygen, invasive mechanical ventilation, and ECMO[92].

In the World health organization led SOLIDARITY trial[95], which was conducted at multiple sites in 30 countries, 11330 adults underwent randomization. Death occurred in 301 of 2743 patients receiving Remdesivir and in 303 of 2708 receiving its control (RR 0.95; 95%CI: 0.81-1.11; P = 0.50) showing no survival benefit. In this study which had good adherence, Remdesivir was given intravenously for 10 d. Remdesivir did not reduce the incidence of new ventilation.

In another randomized control trial, for patients with moderate clinical disease (Pulmonary infiltrates with SpO2 more than 94% by definition); Remdesivir did not demonstrate any difference in clinical status when compared to placebo after a 10-d course. Interestingly, the same study showed improvement in clinical status after a 5-d course. The study was confounded by open-label design and imbalances with co-therapy and therefore the significance is unknown[96].

Other randomized control trials did not show any difference in clinical status outcome between a 5 and a 10-d course of Remdesivir[33,34] and the drug is generally safe with no significant adverse effects[92,94,96,97].

Barcitinib, an oral selective Janus kinase inhibitor 1 and 2 inhibitors impair cell entry of the SARS-CoV-2 virus and inhibits cellular signaling pathway. It has been tested in RCT in combination with Remdesivir and compared to placebo it has improved median time to recovery by 1 d (RR for recovery, 1.16; 95%CI: 1.01-1.32; P = 0.03). At 15 d, time to recovery favors the drug combination. In sicker patients who are on NIV or high flow oxygen the time to recovery was 10 d compared to 18 d. (RR for recovery, 1.51; 95%CI: 1.10-2.08). However, given the lack of efficacy for survival, in practice, it can be used with Remdesivir, when steroids are contraindicated[98].

In summary in patients with severe disease (SpO2 less than 94% with pulmonary infiltrates) and risk of the hyper inflammatory response, Remdesivir may help improve time to clinical recovery and reduce duration of hospitalization, but does not improve survival[92-94,99-101]. It is likely not very helpful or may have very modest benefits in patients who have mild to moderate disease (Pulmonary infiltrates with SpO2 more than 94%)[34,96,100]. As per a meta-analysis, it may help to reduce the need for ventilation but the effect may not be large. It may help to reduce serious adverse events and may aid with some recovery. For non-ventilated patients, a 5 d course compared to 10 d course results in reduced costs, more benefits and less harm[101].

With lack of improvement in survival, the soft benefit of improvement in clinical status, the need to be given by intravenous infusion often as an inpatient over 5 d, lack of cost effectiveness and an endless number of patients with this pandemic, remdesivir is not an optimal answer where the treatment needs to be inexpensive, scalable and equitable[99,101,102]. However since it does reduce time to clinical recovery and reduces duration of hospitalization among survivors, it can help free up inpatient resources in a pandemic and hence gets approval from FDA and Infectious disease society of America[101,103].

Hydroxychloroquine

It is an immunomodulatory drug that has been used extensively in rheumatological disorders. It was repurposed for use in COVID-19 patients and many governments around the world including the United States allowed emergency authorization for its use. Its mechanism of action appears to be by inhibiting glycosylation of ACE2 receptors and increasing the pH of endosomes, in effect preventing virus entry into the cells[104,105].

Many studies have been performed with or without concomitant use of azithromycin compared to placebo after initial case reports and non-randomized studies showed efficacy for the drug against SARS-CoV-2[104]. However, none of the randomized control trials, systematic reviews, and meta-analyses, with or without Azithromycin has shown any benefit for Hydroxychloroquine with regards to survival[92,104,105]. Likewise, there is no benefit with regards to the length of hospitalization, virological cure rate, clinical status score based on a multipoint ordinal scale, need for mechanical ventilation, and radiological improvement[92,104,105]. There was concern over QT prolongation due to both hydroxychloroquine and azithromycin having those properties as well as concern for the possibility of other side effects without much proven benefit as noted before[104,106]. Currently, both these drugs are not used for COVID-19.

ECMO and COVID-19

ECMO is a resource-intensive therapy that has been used when conventional critical care management has failed to help the patient[107]. It has been used in previous pandemics like pandemic influenza A with variable success[108].

It is recommended by experts that ECMO be offered only at experienced centers that have adequate manpower and material resources as well as expertise in managing them, as every aspect of its care from patient selection, maintenance and liberation is highly specialized and nuanced[107]. In fact when regions are under crises level of care amid a surge of cases, then it may be difficult to offer highly resource-intensive therapies like ECMO[107].

The indications, contraindications, and general principles of ECMO care in COVID-19 remain the same[107] with some finer changes to approach and management. It is preferred that aerosolization of the virus is limited and hence transportation is restricted. Cannulation is best performed at the bedside in the ICU. Tracheostomy which is often performed to help lighten sedation and facilitate decannulation needs to be restricted. All personnel need to observe full barrier precautions[107]. Nevertheless, there is evidence that tracheostomy can be safely managed with standard full barrier precautions as mentioned elsewhere in this article and likely guidelines may change. The patient may not be able to be prone due to cannula and likewise, mobilization may be restricted[107].

Patients with COVID-19 often require deep sedation due to various factors and hence post ECMO delirium may need more supportive ICU care or discharge to specialized rehabilitation centers[107,109]. Veno venous ECMO is the most commonly used ECMO for respiratory failure and outcomes are better with this modality compared to veno arterial ECMO which is used only when concomitant circulatory support is necessary[107,109]. Given the high incidence of thrombosis in COVID-19, therapeutic anticoagulation keeping activated partial thromboplastin time 1.5 to 2.5 times normal is recommended often bordering on the higher side[107] to prevent clot formation in the oxygenator and other parts of the circuit.

Initially reports suggested poor outcomes with ECMO[110] with mortality in the range of 80%-100% but subsequently, a report from the Extracorporeal Life Support Organization registry which included only experienced centers suggested that the 90-d mortality in more than 1000 carefully selected patients was about 40% and this compares reasonably well with non-COVID-19 patients, indicating that when patient selection is optimal and with the application of best principles of standardized care, the outcomes can be optimal in COVID-19[109].

RRT

RRT is a term that denotes a process of replacing the non-endocrine function of the kidney in acute or chronic kidney injury/disease encompassing filtration across the permeable membrane, exchange of solute and electrolytes along with the removal of fluid[111]. There are different modalities which include standard intermittent hemodialysis (IHD), continuous RRT (CRRT), prolonged intermittent RRT (PIRRT), and peritoneal dialysis[112]. CRRT or its variates are preferred in critically ill patients due to their superior ability for fluid removal, causing less hemodynamic instability and consistent metabolic control[112]. It also provides for predictable dosing of medication in renal failure. However, CRRT is not superior to IHD when it comes to survival or Renal recovery[112].

CRRT functions by way of three different mechanisms namely convection, diffusion, and adsorption by the filtering membrane[113]. Different modalities or techniques which employ one of these machines are used such as simple diffusion (continuous venovenous hemodialysis), convection (continuous venovenous hemofiltration), or a combination of both (continuous venovenous hemodiafiltration)[114]. No one technique is superior to the other overall and employing any of them is a matter of availability, patient characteristics, and clinician judgment or preference[114]. Timing of RRT, whether early or late after diagnosis of acute kidney injury (AKI) and establishing indication for RRT has been an important question for many well-conducted clinical trials, largely demonstrating equivocal outcomes[113].

There is a paucity of COVID-19 data for RRT. Recommendations from guidelines have essentially been an extension from the non-COVID-19 population with emphasis on limiting staff exposure and optimal utilization of resources during the pandemic[114]. Full standard barrier precautions for staff taking care of ICU patients are recommended[114]. CRRT is ideal for ICU patients which can be managed by ICU nurses but if limited PIRRT can be used which will optimize resource utilization[114]. IHD consumes more specialized resources and equipment along with a dedicated dialysis nurse in full attendance for the duration of the session and is, therefore, less preferred[112]. Access to CRRT is essential with the right internal jugular vein being preferred especially if proning followed by femoral access, left internal jugular vein, and subclavian veins[112].

COVID-19 has been recognized as a prothrombotic disease having consequences for filter life, and as such regional citrate anticoagulation can be used if already in use in the institution. The latter should not be started if such practices are not already in vogue[113,115]. Systemic anticoagulation with low molecular weight heparin or Ultra fractionated heparin or other agents may be necessary to prolong the life of the circuit but specific evidence-based anticoagulation protocols are lacking in the literature[116]. Extracorporeal blood purification with RRT has been proposed as a therapeutic strategy to remove cytokines and other biological immune mediators to improve clinical outcomes. However, evidence for such therapies is currently lacking and is recommended only in the context of clinical trials[116,117].

In a systematic review of COVID-19 patients with AKI, involving 51 studies and 21531 patients, the incidence of AKI was found to be 12.3%. Patients with transplants had a higher rate of AKI at 38.9% (290 patients) and 39% in ICU patients (565 patients). Patients who did not survive had higher rates of AKI at 42% (1745 patients)[118].

RRT use was reported in 39 studies involving 17,664 patients. With overall use of 5.4% with higher rates noted in 16.3% in ICU patients (776 patients), and 15.6% in transplant patients (117 patients)[118]. AKI was more common in studies from North America, followed by Europe, and was least noted in China[118]. There is increasing evidence that both AKI and the need for RRT are important factors influencing survival in COVID-19 patients[112].

CONCLUSION

It was Sir William Osler who inspired by Thomas Carlisle said, “It is not our goal to see what lies dimly in the distance but to do what lies at hand”.

The COVID-19 pandemic has continued to teach us many important medical, social, political, economic, and humane lessons at a huge cost. Early on with a limited understanding of the virus, its transmission, spread in the community and the medical management of the disease, our response as a global community was reactive, guided by abundant caution. Medical practices and literature consisted of non-peer-reviewed articles, case reports, and case series consisting of incomplete and non-standardized data resulting in approaches and clinical management which were not scientifically sound, exposing patients to potentially nonbeneficial or even harmful treatment strategies[119,120].

Organized efforts to develop sound epidemiological, demographic, and evidence-based data resulted in governmental organizations (e.g., United Kingdom based Recovery trial), international trial networks (e.g., REMAP-CAP), The Society of Critical Care Medicine Discovery Viral Infection and Respiratory Illness Universal Study COVID-19 Registry and others who were well-positioned to rapidly deploy pragmatic trials, design data collection networks to meet data analytic needs in response to the COVID-19 pandemic[119,120].

As evident from our review, the application of sound scientific evidence-based management principles distilled from decades of research in the past, with some accommodations in practices specific to the SARS-CoV-2, mitigation strategies, along with the careful implementation of disaster management principles in times of surge have resulted in better and superior outcomes. This is borne out by the fact that although outcomes have varied highly between centers[121], they have generally improved with time[122], especially when health care delivery systems are not stressed due to surge[123]. This is evident by one organization's meticulous and highly diligent efforts to manage the pandemic by way of standardized, protocolized management principles accommodating new information as well as providing room for research opportunities[124]. This along with rapid large-scale effective immunization provides us hope to get back our lives and business back to normal soon.

Footnotes

Manuscript source: Invited manuscript

Corresponding Author's Membership in Professional Societies: Pulmonary Associates; and American College of Chest Physician.

Specialty type: Critical care medicine

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): A, A, A

Grade B (Very good): 0

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Cimen SG, Rodrigues AT S-Editor: Fan JR L-Editor: A P-Editor: Li JH

References
1.  Shah A, Kashyap R, Tosh P, Sampathkumar P, O'Horo JC. Guide to Understanding the 2019 Novel Coronavirus. Mayo Clin Proc. 2020;95:646-652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 51]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
2.  Medicine JHU  Coronavirus resource center. 2021. [cited 16 January 2021]. Available from: https://origin-coronavirus.jhu.edu/.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Lauring AS. Within-Host Viral Diversity: A Window into Viral Evolution. Annu Rev Virol. 2020;7:63-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
4.  Morris DH, Yinda KC, Gamble A, Rossine FW, Huang Q, Bushmaker T, Fischer RJ, Matson MJ, van Doremalen N, Vikesland PJ, Marr LC, Munster VJ, Lloyd-Smith JO. The effect of temperature and humidity on the stability of SARS-CoV-2 and other enveloped viruses. bioRxiv. 2020;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
5.  Singh R, Shaik L, Mehra I, Kashyap R, Surani S. Novel and Controversial Therapies in COVID-19. Open Respir Med J. 2020;14:79-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
6.  Cevik M, Tate M, Lloyd O, Maraolo AE, Schafers J, Ho A. SARS-CoV-2, SARS-CoV, and MERS-CoV viral load dynamics, duration of viral shedding, and infectiousness: a systematic review and meta-analysis. Lancet Microbe. 2021;2:e13-e22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 937]  [Cited by in F6Publishing: 909]  [Article Influence: 303.0]  [Reference Citation Analysis (0)]
7.  Klompas M, Baker MA, Rhee C. Airborne Transmission of SARS-CoV-2: Theoretical Considerations and Available Evidence. JAMA. 2020;324:441-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 260]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
8.  Klompas M, Baker M, Rhee C. What Is an Aerosol-Generating Procedure? JAMA Surg. 2021;156:113-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 89]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]
9.  Morawska L, Johnson GR, Ristovski ZD, Hargreaves M, Mengersen K, Corbett S, Chao CYH, Li Y, Katoshevski D. Size distribution and sites of origin of droplets expelled from the human respiratory tract during expiratory activities. J Aerosol Sci. 2009;40:256-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 701]  [Cited by in F6Publishing: 540]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
10.  Chandel A, Patolia S, Brown AW, Collins AC, Sahjwani D, Khangoora V, Cameron PC, Desai M, Kasarabada A, Kilcullen JK, Nathan SD, King CS. High-flow nasal cannula in COVID-19: Outcomes of application and examination of the ROX index to predict success. Res Care. 2020;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 82]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
11.  Rola P, Farkas J, Spiegel R, Kyle-Sidell C, Weingart S, Duggan L, Garrone M, Thomas A. Rethinking the early intubation paradigm of COVID-19: time to change gears? Clin Exp Emerg Med. 2020;7:78-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
12.  Richardson S, Hirsch JS, Narasimhan M, Crawford JM, McGinn T, Davidson KW;  the Northwell COVID-19 Research Consortium; Barnaby DP, Becker LB, Chelico JD, Cohen SL, Cookingham J, Coppa K, Diefenbach MA, Dominello AJ, Duer-Hefele J, Falzon L, Gitlin J, Hajizadeh N, Harvin TG, Hirschwerk DA, Kim EJ, Kozel ZM, Marrast LM, Mogavero JN, Osorio GA, Qiu M, Zanos TP. Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized With COVID-19 in the New York City Area. JAMA. 2020;323:2052-2059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6024]  [Cited by in F6Publishing: 6187]  [Article Influence: 1546.8]  [Reference Citation Analysis (0)]
13.  Grasselli G, Tonetti T, Protti A, Langer T, Girardis M, Bellani G, Laffey J, Carrafiello G, Carsana L, Rizzuto C, Zanella A, Scaravilli V, Pizzilli G, Grieco DL, Di Meglio L, de Pascale G, Lanza E, Monteduro F, Zompatori M, Filippini C, Locatelli F, Cecconi M, Fumagalli R, Nava S, Vincent JL, Antonelli M, Slutsky AS, Pesenti A, Ranieri VM;  collaborators. Pathophysiology of COVID-19-associated acute respiratory distress syndrome: a multicentre prospective observational study. Lancet Respir Med. 2020;8:1201-1208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 437]  [Article Influence: 109.3]  [Reference Citation Analysis (0)]
14.  Fan E, Beitler JR, Brochard L, Calfee CS, Ferguson ND, Slutsky AS, Brodie D. COVID-19-associated acute respiratory distress syndrome: is a different approach to management warranted? Lancet Respir Med. 2020;8:816-821.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 304]  [Article Influence: 76.0]  [Reference Citation Analysis (0)]
15.  Deshmukh V, Motwani R, Kumar A, Kumari C, Raza K. Histopathological observations in COVID-19: a systematic review. J Clin Pathol. 2021;74:76-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 103]  [Article Influence: 25.8]  [Reference Citation Analysis (0)]
16.  Spoletini G, Alotaibi M, Blasi F, Hill NS. Heated Humidified High-Flow Nasal Oxygen in Adults: Mechanisms of Action and Clinical Implications. Chest. 2015;148:253-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 227]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
17.  Raoof S, Nava S, Carpati C, Hill NS. High-Flow, Noninvasive Ventilation and Awake (Nonintubation) Proning in Patients With Coronavirus Disease 2019 With Respiratory Failure. Chest. 2020;158:1992-2002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 84]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
18.  Leonard S, Atwood CW Jr, Walsh BK, DeBellis RJ, Dungan GC, Strasser W, Whittle JS. Preliminary Findings on Control of Dispersion of Aerosols and Droplets During High-Velocity Nasal Insufflation Therapy Using a Simple Surgical Mask: Implications for the High-Flow Nasal Cannula. Chest. 2020;158:1046-1049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 75]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
19.  Hui DS, Chow BK, Lo T, Tsang OTY, Ko FW, Ng SS, Gin T, Chan MTV. Exhaled air dispersion during high-flow nasal cannula therapy versus CPAP via different masks. Eur Respir J. 2019;53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 221]  [Article Influence: 44.2]  [Reference Citation Analysis (0)]
20.  Hui DS, Hall SD, Chan MT, Chow BK, Tsou JY, Joynt GM, Sullivan CE, Sung JJ. Noninvasive positive-pressure ventilation: An experimental model to assess air and particle dispersion. Chest. 2006;130:730-740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 97]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
21.  Masa JF, Pépin JL, Borel JC, Mokhlesi B, Murphy PB, Sánchez-Quiroga Maria Á. Obesity hypoventilation syndrome. Eur Respir Rev. 2019;28:180097.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 122]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
22.  Ferioli M, Cisternino C, Leo V, Pisani L, Palange P, Nava S. Protecting healthcare workers from SARS-CoV-2 infection: practical indications. Eur Respir Rev. 2020;29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 241]  [Article Influence: 60.3]  [Reference Citation Analysis (0)]
23.  Frat JP, Thille AW, Mercat A, Girault C, Ragot S, Perbet S, Prat G, Boulain T, Morawiec E, Cottereau A, Devaquet J, Nseir S, Razazi K, Mira JP, Argaud L, Chakarian JC, Ricard JD, Wittebole X, Chevalier S, Herbland A, Fartoukh M, Constantin JM, Tonnelier JM, Pierrot M, Mathonnet A, Béduneau G, Delétage-Métreau C, Richard JC, Brochard L, Robert R;  FLORALI Study Group;  REVA Network. High-flow oxygen through nasal cannula in acute hypoxemic respiratory failure. N Engl J Med. 2015;372:2185-2196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1232]  [Cited by in F6Publishing: 1213]  [Article Influence: 134.8]  [Reference Citation Analysis (0)]
24.  Hernández G, Vaquero C, Colinas L, Cuena R, González P, Canabal A, Sanchez S, Rodriguez ML, Villasclaras A, Fernández R. Effect of Postextubation High-Flow Nasal Cannula vs Noninvasive Ventilation on Reintubation and Postextubation Respiratory Failure in High-Risk Patients: A Randomized Clinical Trial. JAMA. 2016;316:1565-1574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 308]  [Cited by in F6Publishing: 296]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
25.  Thille AW, Muller G, Gacouin A, Coudroy R, Decavèle M, Sonneville R, Beloncle F, Girault C, Dangers L, Lautrette A, Cabasson S, Rouzé A, Vivier E, Le Meur A, Ricard JD, Razazi K, Barberet G, Lebert C, Ehrmann S, Sabatier C, Bourenne J, Pradel G, Bailly P, Terzi N, Dellamonica J, Lacave G, Danin PÉ, Nanadoumgar H, Gibelin A, Zanre L, Deye N, Demoule A, Maamar A, Nay MA, Robert R, Ragot S, Frat JP;  HIGH-WEAN Study Group and the REVA Research Network. Effect of Postextubation High-Flow Nasal Oxygen With Noninvasive Ventilation vs High-Flow Nasal Oxygen Alone on Reintubation Among Patients at High Risk of Extubation Failure: A Randomized Clinical Trial. JAMA. 2019;322:1465-1475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 157]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
26.  Bellani G, Laffey JG, Pham T, Madotto F, Fan E, Brochard L, Esteban A, Gattinoni L, Bumbasirevic V, Piquilloud L, van Haren F, Larsson A, McAuley DF, Bauer PR, Arabi YM, Ranieri M, Antonelli M, Rubenfeld GD, Thompson BT, Wrigge H, Slutsky AS, Pesenti A;  LUNG SAFE Investigators;  ESICM Trials Group. Noninvasive Ventilation of Patients with Acute Respiratory Distress Syndrome. Insights from the LUNG SAFE Study. Am J Respir Crit Care Med. 2017;195:67-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 351]  [Article Influence: 50.1]  [Reference Citation Analysis (0)]
27.  Ferreyro BL, Angriman F, Munshi L, Del Sorbo L, Ferguson ND, Rochwerg B, Ryu MJ, Saskin R, Wunsch H, da Costa BR, Scales DC. Association of Noninvasive Oxygenation Strategies With All-Cause Mortality in Adults With Acute Hypoxemic Respiratory Failure: A Systematic Review and Meta-analysis. JAMA. 2020;324:57-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 230]  [Article Influence: 57.5]  [Reference Citation Analysis (0)]
28.  Grasselli G, Greco M, Zanella A, Albano G, Antonelli M, Bellani G, Bonanomi E, Cabrini L, Carlesso E, Castelli G, Cattaneo S, Cereda D, Colombo S, Coluccello A, Crescini G, Forastieri Molinari A, Foti G, Fumagalli R, Iotti GA, Langer T, Latronico N, Lorini FL, Mojoli F, Natalini G, Pessina CM, Ranieri VM, Rech R, Scudeller L, Rosano A, Storti E, Thompson BT, Tirani M, Villani PG, Pesenti A, Cecconi M;  COVID-19 Lombardy ICU Network. Risk Factors Associated With Mortality Among Patients With COVID-19 in Intensive Care Units in Lombardy, Italy. JAMA Intern Med. 2020;180:1345-1355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 850]  [Cited by in F6Publishing: 967]  [Article Influence: 241.8]  [Reference Citation Analysis (0)]
29.  Franco C, Facciolongo N, Tonelli R, Dongilli R, Vianello A, Pisani L, Scala R, Malerba M, Carlucci A, Negri EA, Spoladore G, Arcaro G, Tillio PA, Lastoria C, Schifino G, Tabbì L, Guidelli L, Guaraldi G, Ranieri VM, Clini E, Nava S. Feasibility and clinical impact of out-of-ICU noninvasive respiratory support in patients with COVID-19-related pneumonia. Eur Respir J. 2020;56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 170]  [Article Influence: 42.5]  [Reference Citation Analysis (0)]
30.  Grieco DL, Menga LS, Cesarano M, Rosà T, Spadaro S, Bitondo MM, Montomoli J, Falò G, Tonetti T, Cutuli SL, Pintaudi G, Tanzarella ES, Piervincenzi E, Bongiovanni F, Dell’Anna AM, Delle Cese L, Berardi C, Carelli S, Bocci MG, Montini L, Bello G, Natalini D, De Pascale G, Velardo M, Volta CA, Ranieri VM, Conti G, Maggiore SM, Antonelli M;  Group C-IGS. Effect of Helmet Noninvasive Ventilation vs High-Flow Nasal Oxygen on Days Free of Respiratory Support in Patients With COVID-19 and Moderate to Severe Hypoxemic Respiratory Failure: The HENIVOT Randomized Clinical Trial. JAMA. 2021;325:1731-1743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 249]  [Article Influence: 83.0]  [Reference Citation Analysis (0)]
31.  Papoutsi E, Giannakoulis VG, Xourgia E, Routsi C, Kotanidou A, Siempos II. Effect of timing of intubation on clinical outcomes of critically ill patients with COVID-19: a systematic review and meta-analysis of non-randomized cohort studies. Crit Care. 2021;25:121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 142]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
32.  Cabrini L, Ghislanzoni L, Severgnini P, Landoni G, Baiardo Redaelli M, Franchi F, Romagnoli S. Early versus late tracheal intubation in COVID-19 patients: a pro-con debate also considering heart-lung interactions. Minerva Cardioangiol. 2020;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
33.  Tobin MJ, Laghi F, Jubran A. Caution about early intubation and mechanical ventilation in COVID-19. Ann Intensive Care. 2020;10:78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 160]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
34.  Alhazzani W, Møller MH, Arabi YM, Loeb M, Gong MN, Fan E, Oczkowski S, Levy MM, Derde L, Dzierba A, Du B, Aboodi M, Wunsch H, Cecconi M, Koh Y, Chertow DS, Maitland K, Alshamsi F, Belley-Cote E, Greco M, Laundy M, Morgan JS, Kesecioglu J, McGeer A, Mermel L, Mammen MJ, Alexander PE, Arrington A, Centofanti JE, Citerio G, Baw B, Memish ZA, Hammond N, Hayden FG, Evans L, Rhodes A. Surviving Sepsis Campaign: guidelines on the management of critically ill adults with Coronavirus Disease 2019 (COVID-19). Crit Care Med. 2020;48:e440-e469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 634]  [Cited by in F6Publishing: 603]  [Article Influence: 150.8]  [Reference Citation Analysis (0)]
35.  Mosier JM, Sakles JC, Whitmore SP, Hypes CD, Hallett DK, Hawbaker KE, Snyder LS, Bloom JW. Failed noninvasive positive-pressure ventilation is associated with an increased risk of intubation-related complications. Ann Intensive Care. 2015;5:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 54]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
36.  Kangelaris KN, Ware LB, Wang CY, Janz DR, Zhuo H, Matthay MA, Calfee CS. Timing of Intubation and Clinical Outcomes in Adults With Acute Respiratory Distress Syndrome. Crit Care Med. 2016;44:120-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 137]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
37.  Brochard L, Slutsky A, Pesenti A. Mechanical Ventilation to Minimize Progression of Lung Injury in Acute Respiratory Failure. Am J Respir Crit Care Med. 2017;195:438-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 551]  [Cited by in F6Publishing: 664]  [Article Influence: 94.9]  [Reference Citation Analysis (0)]
38.  Marini JJ, Gattinoni L. Management of COVID-19 Respiratory Distress. JAMA. 2020;323:2329-2330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 706]  [Cited by in F6Publishing: 672]  [Article Influence: 168.0]  [Reference Citation Analysis (0)]
39.  Tobin MJ, Laghi F, Jubran A. Ventilatory failure, ventilator support, and ventilator weaning. Compr Physiol. 2012;2:2871-2921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 53]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
40.  Weissman DN, de Perio MA, Radonovich LJ Jr. COVID-19 and Risks Posed to Personnel During Endotracheal Intubation. JAMA. 2020;323:2027-2028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 102]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
41.  Yao W, Wang T, Jiang B, Gao F, Wang L, Zheng H, Xiao W, Yao S, Mei W, Chen X, Luo A, Sun L, Cook T, Behringer E, Huitink JM, Wong DT, Lane-Fall M, McNarry AF, McGuire B, Higgs A, Shah A, Patel A, Zuo M, Ma W, Xue Z, Zhang LM, Li W, Wang Y, Hagberg C, O'Sullivan EP, Fleisher LA, Wei H;  collaborators. Emergency tracheal intubation in 202 patients with COVID-19 in Wuhan, China: lessons learnt and international expert recommendations. Br J Anaesth. 2020;125:e28-e37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 216]  [Article Influence: 54.0]  [Reference Citation Analysis (0)]
42.  Jain NK, Tirupati R, Palabindala V.   COVID-19: Managing resource crunch and ethical challenges. The Hospitalist: Society of Hospital Medicine, 2020. [cited 16 January 2021]. Available from: https://www.the-hospitalist.org/hospitalist/article/220724/coronavirus-updates/covid-19-managing-resource-crunch-and-ethical.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Tan KS, Halpern NA.   United States Resource Availability for COVID-19. In: Neil A. Halpern M, MCCM, editor: Society of Critical care Medicine, 5/12/2020. [cited 16 January 2021]. Available from: https://www.sccm.org/Blog/March-2020/United-States-Resource-Availability-for-COVID-19?_zs=jxpjd1&_zl=w9pb6.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Matta A, Chaudhary S, Bryan Lo K, DeJoy R 3rd, Gul F, Torres R, Chaisson N, Patarroyo-Aponte G. Timing of Intubation and Its Implications on Outcomes in Critically Ill Patients With Coronavirus Disease 2019 Infection. Crit Care Explor. 2020;2:e0262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
45.  Pandya A, Kaur NA, Sacher D, O'Corragain O, Salerno D, Desai P, Sehgal S, Gordon M, Gupta R, Marchetti N, Zhao H, Patlakh N, Criner GJ, University T;  COVID-19 Research Group. Ventilatory Mechanics in Early vs Late Intubation in a Cohort of Coronavirus Disease 2019 Patients With ARDS: A Single Center's Experience. Chest. 2021;159:653-656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
46.  Siempos II, Xourgia E, Ntaidou TK, Zervakis D, Magira EE, Kotanidou A, Routsi C, Zakynthinos SG. Effect of Early vs. Delayed or No Intubation on Clinical Outcomes of Patients With COVID-19: An Observational Study. Front Med (Lausanne). 2020;7:614152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
47.  Lee YH, Choi KJ, Choi SH, Lee SY, Kim KC, Kim EJ, Lee J. Clinical Significance of Timing of Intubation in Critically Ill Patients with COVID-19: A Multi-Center Retrospective Study. J Clin Med. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
48.  Gattinoni L, Marini JJ, Camporota L. Reply to Tobin et al.: Respiratory Drive Measurements Do Not Signify Conjectural Patient Self-inflicted Lung Injury. Am J Respir Crit Care Med. 2021;203:143-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
49.  Fink JB, Ehrmann S, Li J, Dailey P, McKiernan P, Darquenne C, Martin AR, Rothen-Rutishauser B, Kuehl PJ, Häussermann S, MacLoughlin R, Smaldone GC, Muellinger B, Corcoran TE, Dhand R. Reducing Aerosol-Related Risk of Transmission in the Era of COVID-19: An Interim Guidance Endorsed by the International Society of Aerosols in Medicine. J Aerosol Med Pulm Drug Deliv. 2020;33:300-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 67]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
50.  National Asthma Council  Managing asthma during the COVID-19 (SARS-CoV-2) pandemic. [cited 23 August 2020]. Available from: https://www.asthmahandbook.org.au/clinical-issues/covid-19.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  al HKRPe  Global initiative for Asthma 2020 full report, 2020. [cited 23 January 2021]. Available from: https://ginasthma.org/.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  NICE  COVID-19 rapid guideline: severe asthma. UK: National institute of Clinical Excellence, United Kingdom, 04/03/2020. [cited 23 January 2021]. Available from: https://www.nice.org.uk/guidance/ng166.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Cazzola M, Ora J, Bianco A, Rogliani P, Matera MG. Guidance on nebulization during the current COVID-19 pandemic. Respir Med. 2021;176:106236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 26]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
54.  Lentz RJ, Colt H. Summarizing societal guidelines regarding bronchoscopy during the COVID-19 pandemic. Respirology. 2020;25:574-577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 65]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
55.  Vergnon JM, Trosini-Desert V, Fournier C, Lachkar S, Dutau H, Guibert N, Escarguel B, Froudarakis M;  French-Speaking Group on Thoracic Endoscopy (Groupe d’endoscopie de langue française GELF) of the French Language Respiratory Society (Société de pneumologie de langue française; SPLF). Bronchoscopy use in the COVID-19 era. Respir Med Res. 2020;78:100760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
56.  Chang SH, Jiang J, Kon ZN, Williams DM, Geraci TC, Smith DE, Cerfolio RJ, Zervos M, Bizekis C. Safety and Efficacy of Bronchoscopy in Critically Ill Patients With Coronavirus Disease 2019. Chest. 2021;159:870-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
57.  Torrego A, Pajares V, Fernández-Arias C, Vera P, Mancebo J. Bronchoscopy in Patients with COVID-19 with Invasive Mechanical Ventilation: A Single-Center Experience. Am J Respir Crit Care Med. 2020;202:284-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
58.  Chiesa-Estomba CM, Lechien JR, Calvo-Henríquez C, Fakhry N, Karkos PD, Peer S, Sistiaga-Suarez JA, Gónzalez-García JA, Cammaroto G, Mayo-Yánez M, Parente-Arias P, Saussez S, Ayad T. Systematic review of international guidelines for tracheostomy in COVID-19 patients. Oral Oncol. 2020;108:104844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 44]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
59.  McGrath BA, Brenner MJ, Warrillow SJ, Pandian V, Arora A, Cameron TS, Añon JM, Hernández Martínez G, Truog RD, Block SD, Lui GCY, McDonald C, Rassekh CH, Atkins J, Qiang L, Vergez S, Dulguerov P, Zenk J, Antonelli M, Pelosi P, Walsh BK, Ward E, Shang Y, Gasparini S, Donati A, Singer M, Openshaw PJM, Tolley N, Markel H, Feller-Kopman DJ. Tracheostomy in the COVID-19 era: global and multidisciplinary guidance. Lancet Respir Med. 2020;8:717-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 265]  [Article Influence: 66.3]  [Reference Citation Analysis (1)]
60.  Mattioli F, Fermi M, Ghirelli M, Molteni G, Sgarbi N, Bertellini E, Girardis M, Presutti L, Marudi A. Tracheostomy in the COVID-19 pandemic. Eur Arch Otorhinolaryngol. 2020;277:2133-2135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 63]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
61.  Kwak PE, Connors JR, Benedict PA, Timen MR, Wang B, Zhang Y, Youlios S, Sureau K, Persky MJ, Rafeq S, Angel L, Amin MR. Early Outcomes From Early Tracheostomy for Patients With COVID-19. JAMA Otolaryngol Head Neck Surg. 2021;147:239-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 59]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
62.  Queen Elizabeth Hospital Birmingham COVID-19 airway team. Safety and 30-day outcomes of tracheostomy for COVID-19: a prospective observational cohort study. Br J Anaesth. 2020;125:872-879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 73]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
63.  Mattioli F, Marudi A, Ghirelli M, Molteni G, Sgarbi N, Valerini S, Girardis M, Presutti L, Fermi M. Reply to "Indications and timing for tracheostomy in patients with SARS CoV2-related" by Ferri et al. Eur Arch Otorhinolaryngol. 2020;277:2405-2406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
64.  McGrath BA, Brenner MJ, Warrillow SJ. Tracheostomy for COVID-19: business as usual? Br J Anaesth. 2020;125:867-871.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
65.  Benito DA, Bestourous DE, Tong JY, Pasick LJ, Sataloff RT. Tracheotomy in COVID-19 Patients: A Systematic Review and Meta-analysis of Weaning, Decannulation, and Survival. Otolaryngol Head Neck Surg. 2021;194599820984780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
66.  Marano G, Vaglio S, Pupella S, Facco G, Catalano L, Liumbruno GM, Grazzini G. Convalescent plasma: new evidence for an old therapeutic tool? Blood Transfus. 2016;14:152-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 157]  [Reference Citation Analysis (0)]
67.  Winkler AM, Koepsell SA. The use of convalescent plasma to treat emerging infectious diseases: focus on Ebola virus disease. Curr Opin Hematol. 2015;22:521-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
68.  Jiang S, Zhang X, Yang Y, Hotez PJ, Du L. Neutralizing antibodies for the treatment of COVID-19. Nat Biomed Eng. 2020;4:1134-1139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 79]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
69.  Nguyen AA, Habiballah SB, Platt CD, Geha RS, Chou JS, McDonald DR. Immunoglobulins in the treatment of COVID-19 infection: Proceed with caution! Clin Immunol. 2020;216:108459.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 78]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
70.  Joyner MJ, Carter RE, Senefeld JW, Klassen SA, Mills JR, Johnson PW, Theel ES, Wiggins CC, Bruno KA, Klompas AM, Lesser ER, Kunze KL, Sexton MA, Diaz Soto JC, Baker SE, Shepherd JRA, van Helmond N, Verdun NC, Marks P, van Buskirk CM, Winters JL, Stubbs JR, Rea RF, Hodge DO, Herasevich V, Whelan ER, Clayburn AJ, Larson KF, Ripoll JG, Andersen KJ, Buras MR, Vogt MNP, Dennis JJ, Regimbal RJ, Bauer PR, Blair JE, Paneth NS, Fairweather D, Wright RS, Casadevall A. Convalescent Plasma Antibody Levels and the Risk of Death from Covid-19. N Engl J Med. 2021;384:1015-1027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 401]  [Cited by in F6Publishing: 371]  [Article Influence: 123.7]  [Reference Citation Analysis (0)]
71.  Li L, Zhang W, Hu Y, Tong X, Zheng S, Yang J, Kong Y, Ren L, Wei Q, Mei H, Hu C, Tao C, Yang R, Wang J, Yu Y, Guo Y, Wu X, Xu Z, Zeng L, Xiong N, Chen L, Man N, Liu Y, Xu H, Deng E, Zhang X, Li C, Wang C, Su S, Zhang L, Wu Y, Liu Z. 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;324:460-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 854]  [Cited by in F6Publishing: 857]  [Article Influence: 214.3]  [Reference Citation Analysis (0)]
72.  Agarwal A, Mukherjee A, Kumar G, Chatterjee P, Bhatnagar T, Malhotra P;  PLACID Trial Collaborators. Convalescent plasma in the management of moderate covid-19 in adults in India: open label phase II multicentre randomised controlled trial (PLACID Trial). BMJ. 2020;371:m3939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 426]  [Cited by in F6Publishing: 443]  [Article Influence: 110.8]  [Reference Citation Analysis (0)]
73.  Simonovich VA, Burgos Pratx LD, Scibona P, Beruto MV, Vallone MG, Vázquez C, Savoy N, Giunta DH, Pérez LG, Sánchez MDL, Gamarnik AV, Ojeda DS, Santoro DM, Camino PJ, Antelo S, Rainero K, Vidiella GP, Miyazaki EA, Cornistein W, Trabadelo OA, Ross FM, Spotti M, Funtowicz G, Scordo WE, Losso MH, Ferniot I, Pardo PE, Rodriguez E, Rucci P, Pasquali J, Fuentes NA, Esperatti M, Speroni GA, Nannini EC, Matteaccio A, Michelangelo HG, Follmann D, Lane HC, Belloso WH;  PlasmAr Study Group. A Randomized Trial of Convalescent Plasma in Covid-19 Severe Pneumonia. N Engl J Med. 2021;384:619-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 642]  [Cited by in F6Publishing: 611]  [Article Influence: 203.7]  [Reference Citation Analysis (0)]
74.  Libster R, Pérez Marc G, Wappner D, Coviello S, Bianchi A, Braem V, Esteban I, Caballero MT, Wood C, Berrueta M, Rondan A, Lescano G, Cruz P, Ritou Y, Fernández Viña V, Álvarez Paggi D, Esperante S, Ferreti A, Ofman G, Ciganda Á, Rodriguez R, Lantos J, Valentini R, Itcovici N, Hintze A, Oyarvide ML, Etchegaray C, Neira A, Name I, Alfonso J, López Castelo R, Caruso G, Rapelius S, Alvez F, Etchenique F, Dimase F, Alvarez D, Aranda SS, Sánchez Yanotti C, De Luca J, Jares Baglivo S, Laudanno S, Nowogrodzki F, Larrea R, Silveyra M, Leberzstein G, Debonis A, Molinos J, González M, Perez E, Kreplak N, Pastor Argüello S, Gibbons L, Althabe F, Bergel E, Polack FP;  Fundación INFANT–COVID-19 Group. Early High-Titer Plasma Therapy to Prevent Severe Covid-19 in Older Adults. N Engl J Med. 2021;384:610-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 655]  [Cited by in F6Publishing: 632]  [Article Influence: 210.7]  [Reference Citation Analysis (0)]
75.  FDA  US Food and Drug Administration RDMH. FDA, 2021. [cited 23 January 2021]. Available from: https://www.fda.gov/.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Bloch EM, Shoham S, Casadevall A, Sachais BS, Shaz B, Winters JL, van Buskirk C, Grossman BJ, Joyner M, Henderson JP, Pekosz A, Lau B, Wesolowski A, Katz L, Shan H, Auwaerter PG, Thomas D, Sullivan DJ, Paneth N, Gehrie E, Spitalnik S, Hod EA, Pollack L, Nicholson WT, Pirofski LA, Bailey JA, Tobian AA. Deployment of convalescent plasma for the prevention and treatment of COVID-19. J Clin Invest. 2020;130:2757-2765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 574]  [Cited by in F6Publishing: 548]  [Article Influence: 137.0]  [Reference Citation Analysis (0)]
77.  EUA for Bamlanivimab-A Monoclonal Antibody for COVID-19. JAMA. 2021;325:880-881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 54]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
78.  Company ELa  Lilly's neutralizing antibody bamlanivimab (LY-CoV555) prevented COVID-19 at nursing homes in the BLAZE-2 trial, reducing risk by up to 80 percent for residents. 2021. [cited 23 January 2021]. Available from: https://www.prnewswire.com/news-releases/lillys-neutralizing-antibody-bamlanivimab-ly-cov555-prevented-covid-19-at-nursing-homes-in-the-blaze-2-trial-reducing-risk-by-up-to-80-percent-for-residents-301212159.html.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Hinton RDM, Scientist C, Administration FaD.   USA: FDA, 2021. [cited 23 January 2021]. Available from: https://www.fda.gov/home.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Horby PW, Estcourt L, Peto L, Emberson JR, Staplin N, Spata E, Pessoa-Amorim G, Campbell M, Roddick A, Brunskill NE, George T, Zehnder D, Tiberi S, Aung NN, Uriel A, Widdrington J, Koshy G, Brown T, Scott S, Baillie JK, Buch MH, Chappell LC, Day JN, Faust SN, Jaki T, Jeffery K, Juszczak E, Lim WS, Montgomery A, Mumford A, Rowan K, Thwaites G, Mafham M, Roberts D, Haynes R, Landray MJ.   Convalescent plasma in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. 2021 Preprint. Available from: medRxiv: 2021: 2021.2003.2009.21252736.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Strehl C, Ehlers L, Gaber T, Buttgereit F. Glucocorticoids-All-Rounders Tackling the Versatile Players of the Immune System. Front Immunol. 2019;10:1744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 142]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
82.  RECOVERY Collaborative Group. , Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, Staplin N, Brightling C, Ustianowski A, Elmahi E, Prudon B, Green C, Felton T, Chadwick D, Rege K, Fegan C, Chappell LC, Faust SN, Jaki T, Jeffery K, Montgomery A, Rowan K, Juszczak E, Baillie JK, Haynes R, Landray MJ. Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med. 2021;384:693-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6502]  [Cited by in F6Publishing: 6654]  [Article Influence: 2218.0]  [Reference Citation Analysis (1)]
83.  Siemieniuk RA, Bartoszko JJ, Ge L, Zeraatkar D, Izcovich A, Kum E, Pardo-Hernandez H, Rochwerg B, Lamontagne F, Han MA, Liu Q, Agarwal A, Agoritsas T, Chu DK, Couban R, Darzi A, Devji T, Fang B, Fang C, Flottorp SA, Foroutan F, Ghadimi M, Heels-Ansdell D, Honarmand K, Hou L, Hou X, Ibrahim Q, Khamis A, Lam B, Loeb M, Marcucci M, McLeod SL, Motaghi S, Murthy S, Mustafa RA, Neary JD, Qasim A, Rada G, Riaz IB, Sadeghirad B, Sekercioglu N, Sheng L, Sreekanta A, Switzer C, Tendal B, Thabane L, Tomlinson G, Turner T, Vandvik PO, Vernooij RW, Viteri-García A, Wang Y, Yao L, Ye Z, Guyatt GH, Brignardello-Petersen R, Martinez JPD, Cusano E. Drug treatments for covid-19: living systematic review and network meta-analysis. BMJ. 2020;370:m2980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 441]  [Cited by in F6Publishing: 484]  [Article Influence: 121.0]  [Reference Citation Analysis (0)]
84.  INC AP  Alexion Provides Update on Phase 3 Study of ULTOMIRIS® (ravulizumab-cwvz) in Hospitalized Patients with Severe COVID-19. 2021. [cited 23 January 2021]. Available from: https://pipelinereview.com/index.php/2021011477170/Antibodies/Alexion-Provides-Update-on-Phase-3-Study-of-ULTOMIRIS-ravulizumab-cwvz-in-Hospitalized-Patients-with-Severe-COVID-19.html.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Parr JB. Time to Reassess Tocilizumab's Role in COVID-19 Pneumonia. JAMA Intern Med. 2021;181:12-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 61]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
86.  Gordon AC, Mouncey PR, Al-Beidh F, Rowan KM, Nichol AD, Arabi YM, Annane D, Beane A, van Bentum-Puijk W, Berry LR, Bhimani Z, Bonten MJM, Bradbury CA, Brunkhorst FM, Buzgau A, Cheng AC, Detry MA, Duffy EJ, Estcourt LJ, Fitzgerald M, Goossens H, Haniffa R, Higgins AM, Hills TE, Horvat CM, Lamontagne F, Lawler PR, Leavis HL, Linstrum KM, Litton E, Lorenzi E, Marshall JC, Mayr FB, McAuley D, McGlothlin A, McGuinness SP, McVerry BJ, Montgomery SK, Morpeth SC, Murthy S, Orr K, Parke RL, Parker JC, Patanwala AE, Pettilä V, Rademaker E, Santos MS, Saunders CT, Seymour CW, Shankar-Hari M, Sligl WI, Turgeon AF, Turner AM, van de Veerdonk FL, Zarychanski R, Green C, Lewis RJ, Angus DC, McArthur CJ, Berry S, Webb SA, Derde LPG.   Interleukin-6 Receptor Antagonists in Critically Ill Patients with Covid-19 – Preliminary report. 2021 Preprint. Available from: medRxiv: 2021: 2021.2001.2007.21249390.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Investigators, Gordon AC, Mouncey PR, Al-Beidh F, Rowan KM, Nichol AD, Arabi YM, Annane D, Beane A, van Bentum-Puijk W, Berry LR, Bhimani Z, Bonten MJM, Bradbury CA, Brunkhorst FM, Buzgau A, Cheng AC, Detry MA, Duffy EJ, Estcourt LJ, Fitzgerald M, Goossens H, Haniffa R, Higgins AM, Hills TE, Horvat CM, Lamontagne F, Lawler PR, Leavis HL, Linstrum KM, Litton E, Lorenzi E, Marshall JC, Mayr FB, McAuley DF, McGlothlin A, McGuinness SP, McVerry BJ, Montgomery SK, Morpeth SC, Murthy S, Orr K, Parke RL, Parker JC, Patanwala AE, Pettilä V, Rademaker E, Santos MS, Saunders CT, Seymour CW, Shankar-Hari M, Sligl WI, Turgeon AF, Turner AM, van de Veerdonk FL, Zarychanski R, Green C, Lewis RJ, Angus DC, McArthur CJ, Berry S, Webb SA, Derde LPG. Interleukin-6 Receptor Antagonists in Critically Ill Patients with Covid-19. N Engl J Med 2021; 384: 1491-1502. .  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1175]  [Cited by in F6Publishing: 1150]  [Article Influence: 383.3]  [Reference Citation Analysis (0)]
88.  Horby PW, Campbell M, Staplin N, Spata E, Emberson JR, Pessoa-Amorim G, Peto L, Brightling CE, Sarkar R, Thomas K, Jeebun V, Ashish A, Tully R, Chadwick D, Sharafat M, Stewart R, Rudran B, Baillie JK, Buch MH, Chappell LC, Day JN, Furst SN, Jaki T, Jeffery K, Juszczak E, Lim WS, Montgomery A, Mumford A, Rowan K, Thwaites G, Mafham M, Haynes R, Landray MJ.   Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): preliminary results of a randomised, controlled, open-label, platform trial. 2021 Preprint. Available from: medRxiv 2021: 2021.2002.2011.21249258.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Gupta S, Wang W, Hayek SS, Chan L, Mathews KS, Melamed ML, Brenner SK, Leonberg-Yoo A, Schenck EJ, Radbel J, Reiser J, Bansal A, Srivastava A, Zhou Y, Finkel D, Green A, Mallappallil M, Faugno AJ, Zhang J, Velez JCQ, Shaefi S, Parikh CR, Charytan DM, Athavale AM, Friedman AN, Redfern RE, Short SAP, Correa S, Pokharel KK, Admon AJ, Donnelly JP, Gershengorn HB, Douin DJ, Semler MW, Hernán MA, Leaf DE;  STOP-COVID Investigators. Association Between Early Treatment With Tocilizumab and Mortality Among Critically Ill Patients With COVID-19. JAMA Intern Med. 2021;181:41-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 313]  [Article Influence: 104.3]  [Reference Citation Analysis (0)]
90.  Government U  Thousands more NHS patients to get life-saving COVID-19 treatment. 2021. [cited 23 January 2021]. Available from: https://www.nhs.uk/conditions/coronavirus-covid-19/.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  CDC  CDC Treatment guidelines for Covid-19. 2021. [cited 23 January 2021]. Available from: https://www.cdc.gov/coronavirus/2019-ncov/index.html.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, Hohmann E, Chu HY, Luetkemeyer A, Kline S, Lopez de Castilla D, Finberg RW, Dierberg K, Tapson V, Hsieh L, Patterson TF, Paredes R, Sweeney DA, Short WR, Touloumi G, Lye DC, Ohmagari N, Oh MD, Ruiz-Palacios GM, Benfield T, Fätkenheuer G, Kortepeter MG, Atmar RL, Creech CB, Lundgren J, Babiker AG, Pett S, Neaton JD, Burgess TH, Bonnett T, Green M, Makowski M, Osinusi A, Nayak S, Lane HC;  ACTT-1 Study Group Members. Remdesivir for the Treatment of Covid-19 - Final Report. N Engl J Med. 2020;383:1813-1826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5013]  [Cited by in F6Publishing: 4713]  [Article Influence: 1178.3]  [Reference Citation Analysis (0)]
93.  Wang Y, Zhang D, Du G, Du R, Zhao J, Jin Y, Fu S, Gao L, Cheng Z, Lu Q, Hu Y, Luo G, Wang K, Lu Y, Li H, Wang S, Ruan S, Yang C, Mei C, Wang Y, Ding D, Wu F, Tang X, Ye X, Ye Y, Liu B, Yang J, Yin W, Wang A, Fan G, Zhou F, Liu Z, Gu X, Xu J, Shang L, Zhang Y, Cao L, Guo T, Wan Y, Qin H, Jiang Y, Jaki T, Hayden FG, Horby PW, Cao B, Wang C. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;395:1569-1578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2337]  [Cited by in F6Publishing: 2361]  [Article Influence: 590.3]  [Reference Citation Analysis (0)]
94.  WHO Solidarity Trial Consortium. , Pan H, Peto R, Henao-Restrepo AM, Preziosi MP, Sathiyamoorthy V, Abdool Karim Q, Alejandria MM, Hernández García C, Kieny MP, Malekzadeh R, Murthy S, Reddy KS, Roses Periago M, Abi Hanna P, Ader F, Al-Bader AM, Alhasawi A, Allum E, Alotaibi A, Alvarez-Moreno CA, Appadoo S, Asiri A, Aukrust P, Barratt-Due A, Bellani S, Branca M, Cappel-Porter HBC, Cerrato N, Chow TS, Como N, Eustace J, García PJ, Godbole S, Gotuzzo E, Griskevicius L, Hamra R, Hassan M, Hassany M, Hutton D, Irmansyah I, Jancoriene L, Kirwan J, Kumar S, Lennon P, Lopardo G, Lydon P, Magrini N, Maguire T, Manevska S, Manuel O, McGinty S, Medina MT, Mesa Rubio ML, Miranda-Montoya MC, Nel J, Nunes EP, Perola M, Portolés A, Rasmin MR, Raza A, Rees H, Reges PPS, Rogers CA, Salami K, Salvadori MI, Sinani N, Sterne JAC, Stevanovikj M, Tacconelli E, Tikkinen KAO, Trelle S, Zaid H, Røttingen JA, Swaminathan S. Repurposed Antiviral Drugs for Covid-19 - Interim WHO Solidarity Trial Results. N Engl J Med. 2021;384:497-511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1601]  [Cited by in F6Publishing: 1614]  [Article Influence: 538.0]  [Reference Citation Analysis (0)]
95.  Veiga VC, Prats JAGG, Farias DLC, Rosa RG, Dourado LK, Zampieri FG, Machado FR, Lopes RD, Berwanger O, Azevedo LCP, Avezum Á, Lisboa TC, Rojas SSO, Coelho JC, Leite RT, Carvalho JC, Andrade LEC, Sandes AF, Pintão MCT, Castro CG, Santos SV, de Almeida TML, Costa AN, Gebara OCE, de Freitas FGR, Pacheco ES, Machado DJB, Martin J, Conceição FG, Siqueira SRR, Damiani LP, Ishihara LM, Schneider D, de Souza D, Cavalcanti AB, Scheinberg P. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 254]  [Article Influence: 84.7]  [Reference Citation Analysis (0)]
96.  Spinner CD, Gottlieb RL, Criner GJ, Arribas López JR, Cattelan AM, Soriano Viladomiu A, Ogbuagu O, Malhotra P, Mullane KM, Castagna A, Chai LYA, Roestenberg M, Tsang OTY, Bernasconi E, Le Turnier P, Chang SC, SenGupta D, Hyland RH, Osinusi AO, Cao H, Blair C, Wang H, Gaggar A, Brainard DM, McPhail MJ, Bhagani S, Ahn MY, Sanyal AJ, Huhn G, Marty FM;  GS-US-540-5774 Investigators. Effect of Remdesivir vs Standard Care on Clinical Status at 11 Days in Patients With Moderate COVID-19: A Randomized Clinical Trial. JAMA. 2020;324:1048-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 820]  [Cited by in F6Publishing: 846]  [Article Influence: 211.5]  [Reference Citation Analysis (0)]
97.  Goldman JD, Lye DCB, Hui DS, Marks KM, Bruno R, Montejano R, Spinner CD, Galli M, Ahn MY, Nahass RG, Chen YS, SenGupta D, Hyland RH, Osinusi AO, Cao H, Blair C, Wei X, Gaggar A, Brainard DM, Towner WJ, Muñoz J, Mullane KM, Marty FM, Tashima KT, Diaz G, Subramanian A;  GS-US-540-5773 Investigators. Remdesivir for 5 or 10 Days in Patients with Severe Covid-19. N Engl J Med. 2020;383:1827-1837.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 987]  [Cited by in F6Publishing: 915]  [Article Influence: 228.8]  [Reference Citation Analysis (0)]
98.  Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V, Marconi VC, Ruiz-Palacios GM, Hsieh L, Kline S, Tapson V, Iovine NM, Jain MK, Sweeney DA, El Sahly HM, Branche AR, Regalado Pineda J, Lye DC, Sandkovsky U, Luetkemeyer AF, Cohen SH, Finberg RW, Jackson PEH, Taiwo B, Paules CI, Arguinchona H, Erdmann N, Ahuja N, Frank M, Oh MD, Kim ES, Tan SY, Mularski RA, Nielsen H, Ponce PO, Taylor BS, Larson L, Rouphael NG, Saklawi Y, Cantos VD, Ko ER, Engemann JJ, Amin AN, Watanabe M, Billings J, Elie MC, Davey RT, Burgess TH, Ferreira J, Green M, Makowski M, Cardoso A, de Bono S, Bonnett T, Proschan M, Deye GA, Dempsey W, Nayak SU, Dodd LE, Beigel JH;  ACTT-2 Study Group Members. Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19. N Engl J Med. 2021;384:795-807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1159]  [Cited by in F6Publishing: 1178]  [Article Influence: 392.7]  [Reference Citation Analysis (1)]
99.  Bartoszko JJ, Siemieniuk RAC, Kum E, Qasim A, Zeraatkar D, Ge L, Han MA, Sadeghirad B, Agarwal A, Agoritsas T, Chu DK, Couban R, Darzi AJ, Devji T, Ghadimi M, Honarmand K, Izcovich A, Khamis A, Lamontagne F, Loeb M, Marcucci M, McLeod SL, Motaghi S, Murthy S, Mustafa RA, Neary JD, Pardo-Hernandez H, Rada G, Rochwerg B, Switzer C, Tendal B, Thabane L, Vandvik PO, Vernooij RWM, Viteri-García A, Wang Y, Yao L, Ye Z, Guyatt GH, Brignardello-Petersen R. Prophylaxis against covid-19: living systematic review and network meta-analysis. BMJ. 2021;373:n949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 65]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
100.  Young B, Tan TT, Leo YS. The place for remdesivir in COVID-19 treatment. Lancet Infect Dis. 2021;21:20-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 71]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
101.  Kaka AS, MacDonald R, Greer N, Vela K, Duan-Porter W, Obley A, Wilt TJ. Major Update: Remdesivir for Adults With COVID-19: A Living Systematic Review and Meta-analysis for the American College of Physicians Practice Points. Ann Intern Med. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 52]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
102.  Dyer O. Covid-19: Remdesivir has little or no impact on survival, WHO trial shows. BMJ. 2020;371:m4057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 79]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
103.  Harrington DP, Baden LR, Hogan JW. A Large, Simple Trial Leading to Complex Questions. N Engl J Med. 2021;384:576-577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
104.  Ghazy RM, Almaghraby A, Shaaban R, Kamal A, Beshir H, Moursi A, Ramadan A, Taha SHN. A systematic review and meta-analysis on chloroquine and hydroxychloroquine as monotherapy or combined with azithromycin in COVID-19 treatment. Sci Rep. 2020;10:22139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 61]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
105.  Self WH, Semler MW, Leither LM, Casey JD, Angus DC, Brower RG, Chang SY, Collins SP, Eppensteiner JC, Filbin MR, Files DC, Gibbs KW, Ginde AA, Gong MN, Harrell FE Jr, Hayden DL, Hough CL, Johnson NJ, Khan A, Lindsell CJ, Matthay MA, Moss M, Park PK, Rice TW, Robinson BRH, Schoenfeld DA, Shapiro NI, Steingrub JS, Ulysse CA, Weissman A, Yealy DM, Thompson BT, Brown SM;  National Heart; Lung, and Blood Institute PETAL Clinical Trials Network, Steingrub J, Smithline H, Tiru B, Tidswell M, Kozikowski L, Thornton-Thompson S, De Souza L, Hou P, Baron R, Massaro A, Aisiku I, Fredenburgh L, Seethala R, Johnsky L, Riker R, Seder D, May T, Baumann M, Eldridge A, Lord C, Shapiro N, Talmor D, O’Mara T, Kirk C, Harrison K, Kurt L, Schermerhorn M, Banner-Goodspeed V, Boyle K, Dubosh N, Filbin M, Hibbert K, Parry B, Lavin-Parsons K, Pulido N, Lilley B, Lodenstein C, Margolin J, Brait K, Jones A, Galbraith J, Peacock R, Nandi U, Wachs T, Matthay M, Liu K, Kangelaris K, Wang R, Calfee C, Yee K, Hendey G, Chang S, Lim G, Qadir N, Tam A, Beutler R, Levitt J, Wilson J, Rogers A, Vojnik R, Roque J, Albertson T, Chenoweth J, Adams J, Pearson S, Juarez M, Almasri E, Fayed M, Hughes A, Hillard S, Huebinger R, Wang H, Vidales E, Patel B, Ginde A, Baduashvili A, McKeehan J, Finck L, Higgins C, Howell M, Douglas I, Haukoos J, Hiller T, Lyle C, Cupelo A, Caruso E, Camacho C, Gravitz S, Finigan J, Griesmer C, Park P, Hyzy R, Nelson K, McDonough K, Olbrich N, Williams M, Kapoor R, Nash J, Willig M, Ford H, Gardner-Gray J, Ramesh M, Moses M, Ng Gong M, Aboodi M, Asghar A, Amosu O, Torres M, Kaur S, Chen JT, Hope A, Lopez B, Rosales K, Young You J, Mosier J, Hypes C, Natt B, Borg B, Salvagio Campbell E, Hite RD, Hudock K, Cresie A, Alhasan F, Gomez-Arroyo J, Duggal A, Mehkri O, Hastings A, Sahoo D, Abi Fadel F, Gole S, Shaner V, Wimer A, Meli Y, King A, Terndrup T, Exline M, Pannu S, Robart E, Karow S, Hough C, Robinson B, Johnson N, Henning D, Campo M, Gundel S, Seghal S, Katsandres S, Dean S, Krol O, Jouzestani M, Huynh P, Yealy D, Scholl D, Adams P, McVerry B, Huang D, Angus D, Schooler J, Moore S, Files C, Miller C, Gibbs K, LaRose M, Flores L, Koehler L, Morse C, Sanders J, Langford C, Nanney K, MdalaGausi M, Yeboah P, Morris P, Sturgill J, Seif S, Cassity E, Dhar S, de Wit M, Mason J, Goodwin A, Hall G, Grady A, Chamberlain A, Brown S, Bledsoe J, Leither L, Peltan I, Starr N, Fergus M, Aston V, Montgomery Q, Smith R, Merrill M, Brown K, Armbruster B, Harris E, Middleton E, Paine R, Johnson S, Barrios M, Eppensteiner J, Limkakeng A, McGowan L, Porter T, Bouffler A, Leahy JC, deBoisblanc B, Lammi M, Happel K, Lauto P, Self W, Casey J, Semler M, Collins S, Harrell F, Lindsell C, Rice T, Stubblefield W, Gray C, Johnson J, Roth M, Hays M, Torr D, Zakaria A, Schoenfeld D, Thompson T, Hayden D, Ringwood N, Oldmixon C, Ulysse C, Morse R, Muzikansky A, Fitzgerald L, Whitaker S, Lagakos A, Brower R, Reineck L, Aggarwal N, Bienstock K, Freemer M, Maclawiw M, Weinmann G, Morrison L, Gillespie M, Kryscio R, Brodie D, Zareba W, Rompalo A, Boeckh M, Parsons P, Christie J, Hall J, Horton N, Zoloth L, Dickert N, Diercks D. Effect of Hydroxychloroquine on Clinical Status at 14 Days in Hospitalized Patients With COVID-19: A Randomized Clinical Trial. JAMA. 2020;324:2165-2176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 291]  [Article Influence: 72.8]  [Reference Citation Analysis (0)]
106.  Borba MGS, Val FFA, Sampaio VS, Alexandre MAA, Melo GC, Brito M, Mourão MPG, Brito-Sousa JD, Baía-da-Silva D, Guerra MVF, Hajjar LA, Pinto RC, Balieiro AAS, Pacheco AGF, Santos JDO Jr, Naveca FG, Xavier MS, Siqueira AM, Schwarzbold A, Croda J, Nogueira ML, Romero GAS, Bassat Q, Fontes CJ, Albuquerque BC, Daniel-Ribeiro CT, Monteiro WM, Lacerda MVG;  CloroCovid-19 Team. Effect of High vs Low Doses of Chloroquine Diphosphate as Adjunctive Therapy for Patients Hospitalized With Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection: A Randomized Clinical Trial. JAMA Netw Open. 2020;3:e208857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 706]  [Cited by in F6Publishing: 675]  [Article Influence: 168.8]  [Reference Citation Analysis (0)]
107.  Shekar K, Badulak J, Peek G, Boeken U, Dalton HJ, Arora L, Zakhary B, Ramanathan K, Starr J, Akkanti B, Antonini MV, Ogino MT, Raman L, Barret N, Brodie D, Combes A, Lorusso R, MacLaren G, Müller T, Paden M, Pellegrino V;  ELSO Guideline Working Group. Extracorporeal Life Support Organization Coronavirus Disease 2019 Interim Guidelines: A Consensus Document from an International Group of Interdisciplinary Extracorporeal Membrane Oxygenation Providers. ASAIO J. 2020;66:707-721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 244]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
108.  Zangrillo A, Biondi-Zoccai G, Landoni G, Frati G, Patroniti N, Pesenti A, Pappalardo F. Extracorporeal membrane oxygenation (ECMO) in patients with H1N1 influenza infection: a systematic review and meta-analysis including 8 studies and 266 patients receiving ECMO. Crit Care. 2013;17:R30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 135]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
109.  Barbaro RP, MacLaren G, Boonstra PS, Iwashyna TJ, Slutsky AS, Fan E, Bartlett RH, Tonna JE, Hyslop R, Fanning JJ, Rycus PT, Hyer SJ, Anders MM, Agerstrand CL, Hryniewicz K, Diaz R, Lorusso R, Combes A, Brodie D;  Extracorporeal Life Support Organization. Extracorporeal membrane oxygenation support in COVID-19: an international cohort study of the Extracorporeal Life Support Organization registry. Lancet. 2020;396:1071-1078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 617]  [Cited by in F6Publishing: 577]  [Article Influence: 144.3]  [Reference Citation Analysis (0)]
110.  Haiduc AA, Alom S, Melamed N, Harky A. Role of extracorporeal membrane oxygenation in COVID-19: A systematic review. J Card Surg. 2020;35:2679-2687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
111.  Hechanova LA  Texas Tech University. Merck Manual Professional version. USA: Merck Manual, December 2020. [cited 16 January 2021]. Available from: https://www.merckmanuals.com/professional/.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Ostermann M, Lumlertgul N, Forni LG, Hoste E. What every Intensivist should know about COVID-19 associated acute kidney injury. J Crit Care. 2020;60:91-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
113.  Raza A, Estepa A, Chan V, Jafar MS. Acute Renal Failure in Critically Ill COVID-19 Patients With a Focus on the Role of Renal Replacement Therapy: A Review of What We Know So Far. Cureus. 2020;12:e8429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
114.  Adapa S, Aeddula NR, Konala VM, Chenna A, Naramala S, Madhira BR, Gayam V, Balla M, Muppidi V, Bose S. COVID-19 and Renal Failure: Challenges in the Delivery of Renal Replacement Therapy. J Clin Med Res. 2020;12:276-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 43]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
115.  Shankaranarayanan D, Muthukumar T, Barbar T, Bhasin A, Gerardine S, Lamba P, Leuprecht L, Neupane SP, Salinas T, Shimonov D, Varma E, Liu F. Anticoagulation Strategies and Filter Life in COVID-19 Patients Receiving Continuous Renal Replacement Therapy: A Single-Center Experience. Clin J Am Soc Nephrol. 2020;16:124-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
116.  Nadim MK, Forni LG, Mehta RL, Connor MJ Jr, Liu KD, Ostermann M, Rimmelé T, Zarbock A, Bell S, Bihorac A, Cantaluppi V, Hoste E, Husain-Syed F, Germain MJ, Goldstein SL, Gupta S, Joannidis M, Kashani K, Koyner JL, Legrand M, Lumlertgul N, Mohan S, Pannu N, Peng Z, Perez-Fernandez XL, Pickkers P, Prowle J, Reis T, Srisawat N, Tolwani A, Vijayan A, Villa G, Yang L, Ronco C, Kellum JA. COVID-19-associated acute kidney injury: consensus report of the 25th Acute Disease Quality Initiative (ADQI) Workgroup. Nat Rev Nephrol. 2020;16:747-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 433]  [Cited by in F6Publishing: 362]  [Article Influence: 90.5]  [Reference Citation Analysis (0)]
117.  Ronco C, Reis T. Kidney involvement in COVID-19 and rationale for extracorporeal therapies. Nat Rev Nephrol. 2020;16:308-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 313]  [Article Influence: 78.3]  [Reference Citation Analysis (0)]
118.  Yang X, Tian S, Guo H. Acute kidney injury and renal replacement therapy in COVID-19 patients: A systematic review and meta-analysis. Int Immunopharmacol. 2021;90:107159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 28]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
119.  Walkey AJ, Kumar VK, Harhay MO, Bolesta S, Bansal V, Gajic O, Kashyap R. The Viral Infection and Respiratory Illness Universal Study (VIRUS): An International Registry of Coronavirus 2019-Related Critical Illness. Crit Care Explor. 2020;2:e0113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
120.  Walkey AJ, Sheldrick RC, Kashyap R, Kumar VK, Boman K, Bolesta S, Zampieri FG, Bansal V, Harhay MO, Gajic O. Guiding Principles for the Conduct of Observational Critical Care Research for Coronavirus Disease 2019 Pandemics and Beyond: The Society of Critical Care Medicine Discovery Viral Infection and Respiratory Illness Universal Study Registry. Crit Care Med. 2020;48:e1038-e1044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
121.  Domecq JP, Lal A, Sheldrick CR, Kumar VK, Boman K, Bolesta S, Bansal V, Harhay MO, Garcia MA, Kaufman M, Danesh V, Cheruku S, Banner-Goodspeed VM, Anderson HL 3rd, Milligan PS, Denson JL, St Hill CA, Dodd KW, Martin GS, Gajic O, Walkey AJ, Kashyap R. Outcomes of Patients With Coronavirus Disease 2019 Receiving Organ Support Therapies: The International Viral Infection and Respiratory Illness Universal Study Registry. Crit Care Med. 2021;49:437-448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 78]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
122.  Anesi GL, Jablonski J, Harhay MO, Atkins JH, Bajaj J, Baston C, Brennan PJ, Candeloro CL, Catalano LM, Cereda MF, Chandler JM, Christie JD, Collins T, Courtright KR, Fuchs BD, Gordon E, Greenwood JC, Gudowski S, Hanish A, Hanson Iii CW, Heuer M, Kinniry P, Kornfield ZN, Kruse GB, Lane-Fall M, Martin ND, Mikkelsen ME, Negoianu D, Pascual JL, Patel MB, Pugliese SC, Qasim ZA, Reilly JP, Salmon J, Schweickert WD, Scott MJ, Shashaty MGS, Sicoutris CP, Wang JK, Wang W, Wani AA, Anderson BJ, Gutsche JT. Characteristics, Outcomes, and Trends of Patients With COVID-19-Related Critical Illness at a Learning Health System in the United States. Ann Intern Med. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 77]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
123.  Bravata DM, Perkins AJ, Myers LJ, Arling G, Zhang Y, Zillich AJ, Reese L, Dysangco A, Agarwal R, Myers J, Austin C, Sexson A, Leonard SJ, Dev S, Keyhani S. Association of Intensive Care Unit Patient Load and Demand With Mortality Rates in US Department of Veterans Affairs Hospitals During the COVID-19 Pandemic. JAMA Netw Open. 2021;4:e2034266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 167]  [Article Influence: 55.7]  [Reference Citation Analysis (0)]
124.  O’Horo JC, Cerhan JR, Cahn EJ, Bauer PR, Temesgen Z, Ebbert J, Abril A, Abu Saleh OM, Assi M, Berbari EF, Bierle DM, Bosch W, Burger CD, Cano Cevallos EJ, Clements CM, Carmona Porquera EM, Castillo Almeida NE, Challener DW, Chesdachai S, Comba IY, Corsini Campioli CG, Crane SJ, Dababneh AS, Enzler MJ, Fadel HJ, Ganesh R, De Moraes AG, Go JR, Gordon JE, Gurram PR, Guru PK, Halverson EL, Harrison MF, Heaton HA, Hurt R, Kasten MJ, Lee AS, Levy ER, Libertin CR, Mallea JM, Marshall WF, Matcha G, Meehan AM, Franco PM, Morice WG, O’Brien JJ, Oeckler R, Ommen S, Oravec CP, Orenstein R, Ough NJ, Palraj R, Patel BM, Pureza VS, Pickering B, Phelan DM, Razonable RR, Rizza S, Sampathkumar P, Sanghavi DK, Sen A, Siegel JL, Singbartl K, Shah AS, Shweta FNU, Speicher LL, Suh G, Tabaja H, Tande A, Ting HH, Tontz RC, Vaillant JJ, Vergidis P, Warsame MY, Yetmar ZA, Zomok CD, Williams AW, Badley AD. Outcomes of COVID-19 With the Mayo Clinic Model of Care and Research. Mayo Clin Proc. 2021;96:601-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]