A narrative review of COVID-19-related acute respiratory distress syndrome (CARDS): “typical” or “atypical” ARDS?
Review Article

A narrative review of COVID-19-related acute respiratory distress syndrome (CARDS): “typical” or “atypical” ARDS?

Dan Pu1#, Xiaoqian Zhai1#, Yuwen Zhou2, Yao Xie3, Liansha Tang2, Liyuan Yin1, Hangtian Liu4, Lu Li1

1Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China; 2Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; 3Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China; 4Data Science and Big Data Technology, Chengdu University of Information Technology, Chengdu, China

Contributions: (I) Conception and design: D Pu, X Zhai, L Li; (II) Administrative support: L Li; (III) Provision of study materials or patients: D Pu, X Zhai, Y Zhou, Y Xie, L Tang; (IV) Collection and assembly of data: Y Zhou, Y Xie, L Tang, L Yin, H Liu; (V) Data analysis and interpretation: D Pu, X Zhai, L Yin, H Liu; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work and should be considered as co-first authors.

Correspondence to: Lu Li. Lung Cancer Center, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China. Email: windflower1991@vip.163.com.

Background and Objective: The coronavirus disease of 2019 (COVID-19) is highly infectious and mainly involves the respiratory system, with some patients rapidly progress to acute respiratory distress syndrome (ARDS), which is the leading cause of death in COVID-19 patients. Hence, fully understanding the features of COVID-19-related ARDS (CARDS) and early management of this disease would improve the prognosis and reduce the mortality of severe COVID-19. With the development of recent studies which have focused on CARDS, whether CARDS is “typical” or “atypical” ARDS has become a hotly debated topic.

Methods: We searched for relevant literature from 1999 to 2021 published in PubMed by using the following keywords and their combinations: “COVID-19”, “CARDS”, “ARDS”, “pathophysiological mechanism”, “clinical manifestations”, “prognosis”, and “clinical trials”. Then, we analyzed, compared and highlighted the differences between classic ARDS and CARDS from all of the aspects above.

Key Content and Findings: Classical ARDS commonly occurs within 1 week after a predisposing cause, yet the median time from symptoms onset to CARDS is longer than that of classical ARDS, manifesting within a period of 9.0–12.0 days. Although the lung mechanics exhibited in CARDS grossly match those of classical ARDS, there are some atypical manifestations of CARDS: the severity of hypoxemia seemed not to be proportional to injury of lung mechanics and an increase of thrombogenic processes. Meanwhile, some patients’ symptoms do not correspond with the extent of the organic injury: a chest computed tomography (CT) will reveal the severe and diffuse lung injuries, yet the clinical presentations of patients can be mild.

Conclusions: Despite the differences between the CARDS and ARDS, in addition to the treatment of antivirals, clinicians should continue to follow the accepted evidence-based framework for managing all ARDS cases, including CARDS.

Keywords: Coronavirus disease of 2019 (COVID-19); acute respiratory distress syndrome (ARDS); COVID-19-related ARDS (CARDS); pathophysiological mechanism; prognosis


Submitted Jul 14, 2022. Accepted for publication Aug 08, 2022.

doi: 10.21037/atm-22-3717


Introduction

The coronavirus disease of 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is an ongoing global pandemic (1-3). Predominantly, COVID-19 patients experience mild clinical symptoms; however, 19% of those infected could experience severe or fatal symptoms, particularly COVID-19-related acute respiratory distress syndrome (CARDS) (4). It seems that CARDS is the main cause of death for COVID-19 patients. Hence, a research emphasis has been placed on the pathogenesis, clinical manifestations, and prognostic factors of CARDS. With the development of such studies, controversy has emerged surrounding whether CARDS is simply “typical” acute respiratory distress syndrome (ARDS), or is a particular subtype of ARDS with “atypical” pathophysiological and clinical features. It is increasingly inferred that CARDS might be different from other viral-driven related phenotypes. In a matched cohort study, Chiumello et al. demonstrated arterial oxygen partial pressure/fractional inspired oxygen (PaO2/FiO2) ratio was related to the proportion of non-aerated lung tissue in patients with “typical” ARDS, while there was no such correlation in CARDS patients. Furthermore, in CARDS, the severity of hypoxemia seemed not to be proportional to injury of lung mechanics, which was another point of distinction from classic ARDS (5). However, despite the above differences, there have been no study suggesting that the standard approaches used to manage classic ARDS should be modified for the management of CARDS. Rather, recent study has asserted that clinicians should continue to follow the accepted evidence-based framework for managing all ARDS cases, including CARDS (6). However, management approaches for CARDS are continually evolving with the accumulation of clinical experience (7). In this study, we capture the current understanding of CARDS, including pathophysiological mechanism, manifestations, diagnosis and differential diagnosis, treatment, as well as management and prognosis biomarkers. We further highlight and elucidate the differences between typical ARDS and atypical CARDS from the above aspects. We present the following article in accordance with the Narrative Review reporting checklist (available at https://atm.amegroups.com/article/view/10.21037/atm-22-3717/rc).


Methods

The present study was conducted through the digital libraries of West China Hospital, Sichuan University, Jiangsu, China. We collected the associated literature about the pathophysiological mechanism, clinical manifestations, diagnosis, treatment, as well as the management and prognosis biomarkers of CARDS. All the data were collected from National Center for Biotechnology Information (NCBI) database, PubMed. For data collection, we used some Medical Subject Headings (MeSH) terms and their combinations in title/abstract: “COVID-19”, “CARDS”, "ARDS", “pathophysiological mechanism”, “clinical manifestations”, “prognosis biomarkers”, and “clinical trials”. Table 1 describes the study sequence and details.

Table 1

The search strategy summary

Items Specification
Date of search 2022-01-04 to 2022-05-30
Databases and other sources searched NCBI PubMed
Search terms used (including MeSH and free text search terms) “COVID-19”, “CARDS”, "ARDS", “pathophysiological mechanism”, “clinical manifestations”, “prognosis biomarkers”, and “clinical trials”
Timeframe 1999 to 2021
Inclusion and exclusion criteria The study collected the relevant literature published in English from 1999 to 2021. The literatures of COVID-19 and ARDS was mainly covered
Selection process Dan Pu, Xiaoqian Zhai and Yuwen Zhou jointly collected and assembled the data. Then Yao Xie, Liansha Tang and Liyuan Yina conducted the classification and analysis of the information. Finally, all authors reached an agreement on the manuscript
Any additional considerations None

MeSH, Medical Subject Headings; NCBI, National Center for Biotechnology Information; COVID-19, coronavirus disease of 2019; CARDS, COVID-19-associated acute respiratory distress syndrome; ARDS, acute respiratory distress syndrome.


Mechanism of CARDS

Although the lung mechanics exhibited in CARDS grossly match those of classical ARDS (8), there are some atypical manifestations of CARDS (Figure 1): the separation between the maintenance of relatively good respiratory mechanics and the severity of the hypoxemia and an increase of thrombogenic processes (9-11). Based on different pathophysiology, patients with CARDS are divided into two phenotypes: L phenotype, with almost normal lung compliance, and H phenotype, with reduced lung compliance (12).

Figure 1 Comparison of CARDS and ARDS. Created with BioRender.com. CARDS, COVID-19-related acute respiratory distress syndrome; ARDS, acute respiratory distress syndrome; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; DAD, diffuse alveolar damage.

The hypoxemia caused by the L phenotype is likely attributed to a pulmonary vascular dysregulation leading to a mismatched ratio of ventilation to perfusion rather than an inherent problem of the pulmonary alveoli (10,13). This is probably related to the binding properties of SARS-CoV-2 and the angiotensin-converting enzyme 2 (ACE2) receptors of endothelial cells and arterial smooth muscle cells. Type L patients are responsive to the increase of FiO2 to reverse hypoxemia.

To the contrary, the H phenotype is attributed to hyper-inflammation that causes direct damage to the lungs and eventually leads to ARDS. The process of pathophysiology is as follows: the first 7–10 days (exudative stage) show interstitial/alveolar lymphocytic infiltration in the lung, scarcity of neutrophils (14), multinucleated syncytial cells, diffuse alveolar damage (DAD) with necrosis of endothelial and types I/II epithelial cells, and widespread microthrombosis with microangiopathy (15,16). These changes eventually lead to pulmonary edema, hyaline membrane formation, decreased pulmonary compliance, and difficulties in pulmonary ventilation and gas exchange. Type H patients should be treated as severe ARDS cases, including higher positive end-expiratory pressure (PEEP), if compatible with hemodynamics, prone positioning, and extracorporeal support.


Clinical manifestation

Based on clinical data available, the incidence of CARDS ranges from 14% to 67% (17-21). Classical ARDS commonly occurs within 1 week after a predisposing cause, yet researchers have reported that the median time from symptoms onset to CARDS is longer than that of classical ARDS, manifesting within a period of 9.0–12.0 days (1,17,22-24) (Figure 1). Similar to the classical ARDS, the clinical manifestations of CARDS usually include fever, dry cough, cyanosis of the mouth and lips, and respiratory distress which cannot be adjusted by normal oxygen therapy (25,26). A distinguishing feature of COVID-19 disease is that a lack of dyspnea is recognized in the most critical cases (26), the major reason for which is the direct neurotoxic impact of the virus and a general response caused within the infectious context (27). There are two mechanisms associated with it: one is the direct invasion of SARS-CoV-2 into ACE2-expressing brain cells in the limbic system (especially in the insula), and the second is the indirect toxic effect on the cortical network, which plays a major role in expressing the sensation of dyspnea, through cytokine storm (28). Apart from typical clinical manifestations, CARDS presents some atypical symptoms: it always manifests severe hypoxemia with well-preserved lung mechanics, although in classical ARDS, severe hypoxemia is always associated with poor lung compliance (29,30). Meanwhile, some patients’ symptoms do not correspond with the extent of the organic injury: a chest computed tomography (CT) will reveal the severe and diffuse lung injuries, yet the clinical presentations of patients can be mild (29). Similarly, the results of laboratory tests, including indexes of hemodynamics and tissue perfusion, may be relatively stable in some patients with CARDS (21).


Diagnosis

Clinical diagnosis of CARDS is the same as that of classical ARDS, which is based on the Berlin criteria on the prerequisite of molecular diagnosis of COVID-19 (31). Usually, ARDS is divided into mild [200 mmHg < PaO2/FiO2 ≤300 mmHg with PEEP or continuous positive airway pressure (CPAP) ≥5 cmH2O], moderate (100 mmHg < PaO2/FiO2 ≤200 mmHg with PEEP ≥5 cmH2O), and severe (PaO2/FiO2 ≤100 mmHg with PEEP ≥5 cmH2O) (24). However, some researchers have indicated that the Berlin definition is not fully suitable for the diagnosis and grading of CARDS because of its poor specificity to the diagnosis of DAD—the common histopathologic hallmark of ARDS which is more frequent in CARDS (32,33). The incidence of identification of DAD increases from less than 50% to 69% when Berlin-based diagnosis of classic ARDS is made after 3 days from the symptom onset. However, the median time of occurrence for CARDS is 9 days (34).


Hematologic tests

Abnormalities of hematologic indexes of CARDS present similarly to those of classical ARDS. Lymphopenia, neutrophilia, and thrombocytopenia are observed in blood routine and the deviation from the normal values always indicates the severity (18,35,36). Inflammatory responses lead to the overexpression of infection-related biomarkers including interleukin-6 (IL-6), interleukin-8 (IL-8), vascular endothelial growth factor (VEGF), monocyte chemoattractant protein-1 (MCP-1), granulocyte colony-stimulating factor (GCSF), tumor necrosis factor-α (TNF-α), complement 3 (C3), macrophage inflammatory protein 1-alpha (MIP1A), interferon (IFN)-inducible protein-10 (IP10), C-reactive protein (CRP), serum amyloid A, ferritin, and hepcidin. Among these, median IL-6 levels in hyper-inflammatory ARDS have been shown to be 10- to 200-fold higher than those in CARDS patients (37). Of note, higher concentration of these biomarkers is usually related to more severe disease (1,38-41).


Pathology manifestation

In both ARDS and CARDS, DAD is the major pathological manifestation, and was detectable in all lobes with a prominent distribution in middle and lower lobes (42). Differing from neutrophil infiltration in DAD of ARDS, patients who have died from CARDS have exhibited infiltration of lymphocytes and plasmacytes and a lack of neutrophils in both lungs (12,42) (Figure 1). Meanwhile, patients with DAD in CARDS developed more thrombotic complications compared to those with classical ARDS (43,44) (Figure 1). In addition to different features in DAD, CARDS presents other atypical pathological features. Enlarged pneumocytes and multinucleated syncytial cells have been observed, indicating viral-induced cytopathic-like changes in the absence of intranuclear or intracytoplasmic viral inclusions (42).


Although early studies have shown that CARDS has a unique function that distinguishes it from the historical ARDS, emerging evidence suggests that the respiratory mechanisms of ARDS patients are roughly similar, regardless of SARS-Cov2 infection. However, in terms of treatment, it should not be ignored that CARDS and ARDS involve some different pathophysiological aspects. In ARDS patients, due to the hyper-inflammation, the stability of the alveoli is always reduced, accompanied by pulmonary edema, pulmonary blood flow damage, and pulmonary blood vessel obstruction, resulting in hypoxemia. However, in CARDS, lung compliance remains almost normal, along with increased lung microvascular and macrovascular thrombosis when COVID-19 is severe. From this perspective, the breathing strategy is determined based on the homogeneity and heterogeneity in ARDS and CARDS.

Prevention of venous thromboembolism

A prospective cohort study (44) showed that the incidence of thrombotic complications in COVID-19 intensive care unit (ICU) patients is very high. Some 42.6% of CARDS patients were diagnosed with clinically relevant thrombotic complications during their stay in the ICU, especially, pulmonary embolism (16.7%) (45). Therefore, it is recommended that all COVID-19 patients admitted to the ICU should be strictly administered thromboprophylaxis where there are no contraindications, even in the absence of substantiating evidence (46). To date, several guidelines have recommended low molecular weight heparin for treatment severe COVID-19 patients with hypercoagulation.

Streptokinase, one of the thrombolytics, was recently found to promote lung function and oxygenation, and decrease ICU stay length and mortality in a randomized controlled trial (RCT) recruiting 60 ARDS patients without response to standard therapy (47). Besides, the results of previous phase I clinical trial reported that tissue plasminogen activator (tPA) used as salvage therapy for ARDS yielded significant improvements in PaO2/FiO2 (48). Preliminary studies have reported that tPA may improve recovery of ARDS patients and reduce COVID-19-related mortality (49,50). Anti-thrombosis therapy, as a potential therapy, is being evaluated in a few studies of CARDS patients on the basis of the pulmonary microthrombosis reports.


Respiratory support

Respiratory support plays an important role in the management of patients with CARDS and ARDS. Although the risk factors of CARDS associated with respiratory failure that require mechanical ventilation are not clearly described in the limited literature available, the risk factors associated with severe disease include older age (>60 years), male gender, and complications such as diabetes, chronic respiratory diseases, cardiovascular diseases, malignant tumors, and immunodeficiency (51). In addition, early in the pandemic of COVID-19, research demonstrated the differences between CARDS and classical ARDS and suggested different ventilatory management in CARDS. However, according to the gradually enriched understanding of CARDS, it is reasonable to employ a similar strategy at this stage, despite residual gray areas in the understanding of COVID-19 (52,53).

Respiratory support of mild ARDS

Patients with PaO2/FiO2 <300 mmHg should be administrated oxygen treatment immediately. A high-flow nasal cannula (HFNC) or a mask for oxygen supplementation could be implemented at the initial stage and the respiratory distress and/or hypoxemia should be promptly re-evaluated in time in the case of delayed intubation. In the absence of indications for tracheal intubation, patients with CARDS who cannot obtain a HFNC should undergo a close monitoring test of nasal intermittent positive pressure ventilation (NIPPV). For patients with persistent hypoxemia who have not undergone tracheal intubation despite increased supplemental oxygen demand, an awake prone position should be considered to improve oxygenation (CIIa), as reports have indicated that awake prone positioning could improve the overall median oxygen saturation of these patients (30). Nonetheless, considering the risk of viral transmission to other patients and health care workers, HFNC are not implemented in emergency rooms, unless negative pressure single rooms are available. A study has confirmed that HFNC could improve the outcome of patients with acute hypoxemic respiratory failure when the optimal oxygen saturation (SpO2) ranged from 92% to 96% (54).

Respiratory support of moderate-severe ARDS

For adult patients with moderate-to-severe ARDS, it is recommended to ventilate in the prone position for 12–16 hours a day (BIIa). This ventilation strategy can evenly distribute the gas-tissue ratio and lung stress and strain distribution, yielding a significant improvement in arterial blood gas (55). Some clinical trials have convincingly shown that ARDS patients with PaO2/FiO2 <150 mmHg can reduce mortality by ventilating prone for at least 16 hours a day (56,57). To date, no study has described the clinical course of prone position ventilation in patients with COVID-19. However, we infer that these patients should also benefit from prone ventilation.

Patients with CARDS that have indications as follows for invasive mechanical ventilation should receive endotracheal intubation: (I) respiratory distress aggravation: when hypoxemia cannot be improved (SPO2 ≤93%), the frequency of breathing ≥35 times/minute, or the tidal volume (Vt) is too large (>9 to 10 mL/kg weight) under HFNC or NIPPV treatment. (II) Tissue hypoxia or lactic acid elevation: the performances of tissue hypoxia aggravate, such as increasing lactic acid or decreasing central venous oxygen saturation (ScvO2). (III) Hemodynamic instability or consciousness disorder. However, invasive mechanical ventilation has two sides. On the one hand, it potentially saves lives. On the other hand, it also causes ventilator-induced lung injury (VILI), exacerbating lung damage in ARDS patients and eventually leading to multiple organ failure. Reducing VILI may be achieved through low Vt ventilation (4–8 mL/kg), plateau pressures <30 cmH2O, a conservative fluid strategy, higher PEEP. The recommendation comes from the experience of non-CARDS experience whereby higher PEEP in patients with moderate (PaO2/FiO2 100–200 mmHg) and severe ARDS (PaO2/FiO2 <100 mmHg) leads to lower rates of ICU mortality (58).

Furthermore, in ARDS patients, the analgesia and sedation effects should be adjusted according to the specific needs of the patient to avoid lung injury during the ventilation. Early neuromuscular blocking has been suggested to reduce 90-day mortality (59), although evidence remains controversial (60). Therefore, it is suggested that neuromuscular blocking agents (NMBA) should be used in patients with persistent ventilator dyssynchrony, and patients needing ongoing deep sedation prone ventilation, or persistently high plateau pressures (61).

An extracorporeal membrane oxygenator (ECMO) should be taken into consideration if a patient present with persistent hypoxemia, regardless of whether lung protective ventilation measures have been taken or not. The venovenous ECMO (vv-ECMO) has traditionally been used to provide adequate tissue oxygenation, to support patients with severe ARDS. However, the efficacy of ECMO in patients with ARDS remains controversial. A study showed that the use of ECMO in severe ARDS patients does not significantly reduce the 60-day mortality compared with conventional mechanical ventilation strategies including ECMO as a rescue therapy (62). There has been one study that demonstrated that the use of vv-ECMO in adults with severe ARDS can reduce 60-day mortality when compared with conventional mechanical ventilation, despite a moderate risk of major bleeding (63). Recently, guidelines have recommended that ECMO is used in the treatment of severe ARDS patients (64). However, in CARDS patients, the role of vv-ECMO is uncertain. One study demonstrated that the mortality rate was similar between ARDS and CARDS patients with vv-ECMO, which indicated that the use of vv-ECMO can achieve a similar effect in CARDS and ARDS patients (65). Patients with severe COVID-19 receiving ECMO support should be monitored closely for multiple drug-resistant bacteria (Klebsiella pneumoniae, Baumann bacillus, Pseudomonas aeruginosa, etc.) infections which will lead to poor prognosis. Due to the uncertain benefits and possible risks, the National Institutes of Health (NIH) guideline neither recommend nor advise against the use of ECMO in patients with COVID-19 and refractory hypoxemia.


Pharmacological therapy of CARDS

The pharmacologic therapy of CARDS does not appear “atypical”. Most therapies of CARDS take the therapies of “typical” ARDS caused by other factors for reference, due to the similar critical physiopathologic processes such as cytokine storm. Actually, the pivotal point of combating CARDS may be anti-COVID-19 therapy itself. Here, we have summarized the following pharmacologic therapies of CARDS.

Antivirals

Remdesivir (GS-5734), as a broad-spectrum antiviral drug, was regarded the most promising drug for treating severe COVID-19, but disappointing results have emerged. Study published in the Lancet reported no observable effect of remdesivir on the recovery or mortality of hospitalized patients with COVID-19 compared to placebo controls (66). Thus, Remdesivir was fails to be used to treat severe COVID-19.


Anti-inflammatory and immunoregulatory therapy

Corticosteroids

Owing to the anti-inflammatory and immune-modulating properties, corticosteroids have been widely applied for CARDS. However, the application of corticosteroids on CARDS patients remains controversial. The World Health Organization (WHO) (67) conducted a prospective meta-analysis, which pooled data from seven trials (RECOVERY, CoDEX, REMAP-CAP, CAPE COVID, and another 3 trials), to evaluate the response of corticosteroids to critically ill COVID-19 patients. Among these trials, over 50% of patients came from the RECOVERY trial. The RECOVERY trial confirmed for the first time that low-dose dexamethasone (6 mg/d, no more than 10 days) can reduce less than 28-day mortality among patients receiving invasive mechanical ventilation or oxygen alone (68). Based on that, medical officers in some countries have recommended to use glucocorticoids in patients with serious COVID-19 (69). In the CoDEX trial, intravenous dexamethasone was shown to shorten the ventilator-free days of patients with CARDS (70). The REMAP-CAP trial indicated that hydrocortisone contributed to improved organ support-free days within 21 days (71). Another retrospective study found that low-dose corticosteroid therapy may help to reduce the risk of in-hospital death in CARDS patients (72). The above studies indicated that the use of steroids was related to the reduced mortality in critically ill patients with COVID-19, and corticosteroids could be administrated in general treatment in CARDS patients (73). Nevertheless, another study found that patients with mild ARDS could not benefit form corticosteroids administration (74). Furthermore, specific virus types may lead to different responses for corticosteroids. Prior studies have found that corticosteroids were linked to delayed viral clearance in SARS and Middle East respiratory syndrome (MERS) (75,76), which increased concerns that corticosteroids might damage host response to COVID-19. In addition, patients with influenza pneumonia who received corticosteroids experienced increased mortality (77). In summary, further study is required to explore the harmful or beneficial effects of corticosteroids for this application (78). Furthermore, re-evaluation in combination with novel therapeutic strategy, such as new anti-viral therapies, immunomodulatory agents, and potential monoclonal antibodies (mAbs), may display considerable efficacy as well.

Anti-cytokine storm drugs

As the critical pathophysiologic progress in CARDS, the cytokine storm is triggered by the colossal release of pro-inflammatory cytokines, including IL-6, IL-1, TNF-α, and so on. This leads to a hyperactive and dysregulated immune reaction causing organ dysfunction. Therefore, the immunomodulatory drugs are of vital importance in anti-cytokine storm therapy.

Tocilizumab (TCZ), one of the most studied IL-6 inhibitors, has been identified to exert a great therapeutic effect in CARDS patients. A Chinese open-label, noncontrolled study showed that 21 severe COVID-19 patients who were treated with TCZ (400 mg IV infusion) harbored normal lymphocyte counts and improved oxygenation, manifesting the potential role of TCZ (79). Besides, Roche has approved TCZ to enter into a phase III RCT (COVACTA) in severe, hospitalized COVID-19 patients (80). Another IL-6 inhibitor, sarilumab, has been initiated clinical trials (NCT04315298, NCT04327388) in severe cases of SARS-CoV-2. Furthermore, novel mAb that inhibits IL-6 receptor (TZLS-501) is currently under investigation.

Anakinra (ANK) is an inhibitor of IL-1, which is considered a potential life-saving therapy for patients using non-invasive ventilation outside of the ICU (81). A small-scale respective study indicated that 55.6% of CARDS patients may benefit from high dose (100 mg every 6 h) subcutaneous ANK therapy (82). Especially in patients with elevated aminotransferases, ANK might become a potential alternative for non-response TCZ patients.

Granulocyte-macrophage colony-stimulating factor (GM-CSF) plays a cardinal role in inflammatory modulation. When binding to the receptor (GM-CSFR-α), various pro-inflammatory pathways are activated, inducing the release of pro-inflammatory cytokines (83). A single-center, prospective cohort study found that mavrilimumab (an anti-GM-CSFR-α mAb) can improve the clinical outcome of patients with severe COVID-19 (84).

The functions of various IFN subtypes are different. In ARDS, IFN-γ has proinflammatory functions whereas IFN-b1a has antiviral and immunomodulatory functions (85,86). As such, the application of inhaled IFN-b1a formulation (SNG001) in CARDS is being evaluated in a clinical trial (NCT04385095).

Vitamin C and D might act as strong immunosuppressive agents in inhibiting cytokine release syndrome in COVID-19 (87). A retrospective study identified that high-dose (6 g IV infusion q12h D1 and 6 g qd for the following 4 days) vitamin C may decrease the death rate without adverse events in COVID-19 patients (88). In addition, the lack of vitamin D has been found to be associated with ARDS, due to lower 25(OH)D concentration (89). Currently, various RCTs utilizing either oral 25(OH)D or vitamin D are ongoing (90).

Mesenchymal stem cell (MSC) therapy

Violent cytokines release is attributed to T cells hyper-activation in CARDS patients, which lead to cytotoxic effects on the respiratory system (91). MSC could reduce the activation of T cells, B cells, natural killer (NK) cells, and dendritic cells (DC) by secreting cytokines. In addition, the induction of neutrophils, macrophages, and anti-inflammatory regulatory Treg cells is also the performance of MSC’s immune modulation function. Currently, MSCs which are mainly derived from bone marrow (BM), umbilical cord (UC), adipose tissue (AD), and dental pulp (DP) have been involved in the clinical trials against COVID-19 in many countries (92,93). Preclinical research has identified that BM-MSCs have the regeneration and anti-inflammatory impact on damaged pulmonary alveolar and endothelial cells, through the release of extracellular vesicle (EV) and paracrine factors and transfer of mitochondria (94,95). Therefore, MSCs are shown to have the potential for treating severe COVID-19 cases. Previous studies have shown that MSC therapy improves the symptom and function of COVID-19 patients with ARDS and obtains a significant survival rate (95-97). Besides, exosomes originated from allogeneic BM-MSCs have been investigated to treat severely compromised COVID-19 patients in clinical trials (98-101). These results showed excellent ability and safety to downregulate cytokine storm, reconstitute immunity, and restore oxygenation.

Convalescent human plasma and intravenous immunoglobulin (IVIG)

Convalescent plasma enriched in antibodies is obtained from recovered COVID-19 patient, and then transfused to the infected patients. One study reported four of five severe COVID-19 cases who were transfused with convalescent plasma resolved the symptoms of CARDS at 12 days after transfusion (102). However, a Chinese study revealed that convalescent plasma therapy did not achieve a statistically significant clinical improvement within 28 days (103). Although in August 2020, the Food and Drug Authority (FDA) approved the use of convalescent plasma for the treatment of critically ill COVID-19 patients (104), this therapy still needs further research. In terms of IVIG, previous evidence has suggested that a high dose of IVIG therapy should be considered in patients with severe COVID-19 (105). Furthermore, the National Health Service (NHS) specialty guideline also recommended the use of IVIG for the treatment of COVID-19 patients with sepsis (106); IVIG is considered an important therapeutic approach.


Advanced potential therapy

Repositioning drugs

Currently, some advanced potential drugs have emerged to treat CARDS, which are mainly inferred from the mechanism of action with limited supporting clinical evidence. However, these therapies may provide a novel direction for developing repositioning drugs which could fight CARDS.

Opioids

Although opioids are most exploited in the analgesic aspect, they could also exert triple effects in combating COVID-19. The benefits of utilizing opioids contain addressing the unfavorable side-effects [cough (107) and dyspnea (108)], overcoming the virus infection cycle (inhibit lysosomal acidification) (109), and the host reactions to the virus-elicited pathogenesis (anti-excessive inflammation) (110). In addition, hydromorphone and oxycodone could alleviate the subjective perception of CARDS as well as reduce anxiety effects (111,112). Therefore, further investigation is warranted to evaluate the long-term therapeutic effects of opioids.

Radiotherapy (RT)

The evidence in literature has revealed that RT may be effective for mitigating CARDS (113). Firstly, low-dose radiation (LDR) is conducive to decelerate viral loading and replication (114) in the pulmonary epithelial cells by oxygen metabolism. In addition, LDR could enhance the recruitment of circulating immune cells and further promote the antiviral environment (115). As for the LDR, a single total dose of 0.3–0.5 Gy would be of great benefit for CARDS patients (116). However, the optimal dose of RT should be assessed carefully and repetitively in the future clinical trials.

Poly ADP-ribose polymerase (PARP) inhibitors (PARPis)

PARPs take part in various cellular processes, containing cell death, autophagy, antiviral response, and immune function. Some researchers (117,118) have verified that PARPi could decrease the critical interleukins in COVID-19-induced cytokine storm. Furthermore, PARPis has the ability of counteracting cell death induced by SARS-CoV-2 and inflammation and supporting cell survival (119). The benefits of PARPis have been supported by great therapeutic effects in animal models of VILI and ARDS (120,121). Thus, PARPis may potentiate the effectiveness of anti-cytokine storm therapy (122-124) and would be beneficial for SARS-CoV-2 patients.


Prognosis

Mortality and survival

Although most COVID-19 patients experience mild symptoms, 19% could present with intractable conditions and develop severe or fatal symptoms, particularly CARDS (4). In developed countries, the mortality rate of cards can reach 30–50% (125-129), which is consistent with the mortality from non-COVID ARDS in the ICU. Moreover, in some cases, the mortality rate of patients with CARDS can reach 70% (130). In addition, a study showed that the 90-day mortality rate of CARDS increased with the severity of CARDS (30%, 34%, and 50% for mild, moderate, and severe cards, respectively) (125). Another unfortunate reality is that patients with CARDS who are accompanied with acute kidney injury (AKI), acute heart injury, and septic shock will face significantly reduced mortality (131).

Sequelae

Patients who have survived CARDS may further experience many medical consequences post intensive care therapies. Most common long-term consequences of CARDS include impaired weaning from mechanical ventilation, ventilator-diaphragmatic interactions, ICU-acquired weakness (132-134), consequences from other organ injury (131), other neuromuscular disorders (e.g., dysphagia) (135,136) and/or thromboembolic complications (137). All the above “sequelae” of CARDS not only damage the survival and health of patients, but also place a huge burden on the social medical system. Hence, it is necessary to identify biological markers that predict individual prognosis.

Prognosis biomarkers

The most common combined biomarkers that have been used are age, lymphocyte percentage, monocyte percentage, CRP, procalcitonin, and serum albumin. These are regarded as independent predictors of a more severe illness course (138,139). In addition, based on the pathogenesis of CARDS: endothelial injury, epithelial injury, inflammatory cascade, and coagulation cascade (140), there are some the most commonly promising prognostic biomarkers: lactate dehydrogenase [LDH; weakens the immune response to viral infection (141)]; ferritin (142); D-dimer (143); proinflammatory cytokines (they often are measured as part of a panel and have insufficient specificity to serve as a stand-alone biomarker (144,145); nicotinamide phosphoribosyltransferase (eNAMPT) (146); soluble intercellular adhesion molecule-1 (sICAM-1) [an early pulmonary endothelial injury maker of CARDS (147)]; angiopoeitin-2 (Ang-2) in plasma, and receptor for advanced glycation end products (RAGE) in plasma (148).


Summary

The COVID-19 is an unprecedented pandemic and can lead to fatal clinical outcomes, particularly CARDS. Researchers have made great efforts to improve the prognosis of severe COVID-19 and reduce the mortality caused by CARDS. Among them, a large number of studies have attempted to determine whether CARDS is “typical” or “atypical” ARDS. As of the time of writing, we have delineated the pathophysiological mechanism, manifestations, diagnosis, treatment, management, and prognosis biomarkers of CARDS based on the past 1-year development of this disease, and further highlighted and elucidated the differences between typical ARDS and atypical CARDS from all of the above aspects. Despite the differences between the CARDS and ARDS, in addition to the treatment of antivirals, clinicians should continue to follow the accepted evidence-based framework for managing all ARDS cases, including CARDS. In fact, our study is still not comprehensive, such as failing to explain the mechanistic differences between CARDS and ARDS in more depth. However, we still help clarify the homogeneity and heterogeneity between CARDS with other cause ARDS, which may provide direction for the future basic research and help to optimize the clinical management framework of this disease.


Acknowledgments

Funding: This work was supported by the 1·3·5 Project for Disciplines of Excellence-Clinical Research Incubation Project, West China Hospital, Sichuan University (No. 2020HXFH046) and the Post-Doctor Research Project, West China Hospital, Sichuan University (No. 2020HXBH131).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://atm.amegroups.com/article/view/10.21037/atm-22-3717/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-22-3717/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395:497-506. [Crossref] [PubMed]
  2. Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 2020;395:565-74. [Crossref] [PubMed]
  3. Zhu N, Zhang D, Wang W, et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N Engl J Med 2020;382:727-33. [Crossref] [PubMed]
  4. Wu Z, McGoogan JM. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72 314 Cases From the Chinese Center for Disease Control and Prevention. JAMA 2020;323:1239-42. [Crossref] [PubMed]
  5. Chiumello D, Busana M, Coppola S, et al. Physiological and quantitative CT-scan characterization of COVID-19 and typical ARDS: a matched cohort study. Intensive Care Med 2020;46:2187-96. [Crossref] [PubMed]
  6. Goligher EC, Ranieri VM, Slutsky AS. Is severe COVID-19 pneumonia a typical or atypical form of ARDS? And does it matter? Intensive Care Med 2021;47:83-5. [Crossref] [PubMed]
  7. Matthay MA, Aldrich JM, Gotts JE. Treatment for severe acute respiratory distress syndrome from COVID-19. Lancet Respir Med 2020;8:433-4. [Crossref] [PubMed]
  8. Grasselli G, Tonetti T, Protti A, et al. Pathophysiology of COVID-19-associated acute respiratory distress syndrome: a multicentre prospective observational study. Lancet Respir Med 2020;8:1201-8. [Crossref] [PubMed]
  9. Karmouty-Quintana H, Thandavarayan RA, Keller SP, et al. Emerging Mechanisms of Pulmonary Vasoconstriction in SARS-CoV-2-Induced Acute Respiratory Distress Syndrome (ARDS) and Potential Therapeutic Targets. Int J Mol Sci 2020;21:8081. [Crossref] [PubMed]
  10. Gattinoni L, Chiumello D, Rossi S. COVID-19 pneumonia: ARDS or not? Crit Care 2020;24:154. [Crossref] [PubMed]
  11. Lin SH, Zhao YS, Zhou DX, et al. Coronavirus disease 2019 (COVID-19): cytokine storms, hyper-inflammatory phenotypes, and acute respiratory distress syndrome. Genes Dis 2020;7:520-7. [Crossref] [PubMed]
  12. Montenegro F, Unigarro L, Paredes G, et al. Acute respiratory distress syndrome (ARDS) caused by the novel coronavirus disease (COVID-19): a practical comprehensive literature review. Expert Rev Respir Med 2021;15:183-95. [Crossref] [PubMed]
  13. Potus F, Mai V, Lebret M, et al. Novel insights on the pulmonary vascular consequences of COVID-19. Am J Physiol Lung Cell Mol Physiol 2020;319:L277-88. [Crossref] [PubMed]
  14. Kassirian S, Taneja R, Mehta S. Diagnosis and Management of Acute Respiratory Distress Syndrome in a Time of COVID-19. Diagnostics (Basel) 2020;10:1053. [Crossref] [PubMed]
  15. Li X, Ma X. Acute respiratory failure in COVID-19: is it "typical" ARDS? Crit Care 2020;24:198. [Crossref] [PubMed]
  16. Hariri L, Hardin CC. Covid-19, Angiogenesis, and ARDS Endotypes. N Engl J Med 2020;383:182-3. [Crossref] [PubMed]
  17. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 2020;395:1054-62. [Crossref] [PubMed]
  18. Hong KS, Lee KH, Chung JH, et al. Clinical Features and Outcomes of 98 Patients Hospitalized with SARS-CoV-2 Infection in Daegu, South Korea: A Brief Descriptive Study. Yonsei Med J 2020;61:431-7. [Crossref] [PubMed]
  19. Sun P, Qie S, Liu Z, et al. Clinical characteristics of hospitalized patients with SARS-CoV-2 infection: A single arm meta-analysis. J Med Virol 2020;92:612-7. [Crossref] [PubMed]
  20. Chand S, Kapoor S, Orsi D, et al. COVID-19-Associated Critical Illness-Report of the First 300 Patients Admitted to Intensive Care Units at a New York City Medical Center. J Intensive Care Med 2020;35:963-70. [Crossref] [PubMed]
  21. Cummings MJ, Baldwin MR, Abrams D, et al. Epidemiology, clinical course, and outcomes of critically ill adults with COVID-19 in New York City: a prospective cohort study. Lancet 2020;395:1763-70. [Crossref] [PubMed]
  22. Wang D, Hu B, Hu C, et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA 2020;323:1061-9. [Crossref] [PubMed]
  23. Ferrando C, Suarez-Sipmann F, Mellado-Artigas R, et al. Clinical features, ventilatory management, and outcome of ARDS caused by COVID-19 are similar to other causes of ARDS. Intensive Care Med 2020;46:2200-11. [Crossref] [PubMed]
  24. ARDS Definition Task Force. Acute respiratory distress syndrome: the Berlin Definition. JAMA 2012;307:2526-33. [PubMed]
  25. Mao L, Jin H, Wang M, et al. Neurologic Manifestations of Hospitalized Patients With Coronavirus Disease 2019 in Wuhan, China. JAMA Neurol 2020;77:683-90. [Crossref] [PubMed]
  26. De Vito EL. Possible Role of Corollary Discharge in Lack of Dyspnea in Patients With COVID-19 Disease. Front Physiol 2021;12:719166. [Crossref] [PubMed]
  27. Bertran Recasens B, Martinez-Llorens JM, Rodriguez-Sevilla JJ, et al. Lack of dyspnea in patients with Covid-19: another neurological conundrum? Eur J Neurol 2020;27:e40. [Crossref] [PubMed]
  28. Nouri-Vaskeh M, Sharifi A, Khalili N, et al. Dyspneic and non-dyspneic (silent) hypoxemia in COVID-19: Possible neurological mechanism. Clin Neurol Neurosurg 2020;198:106217. [Crossref] [PubMed]
  29. Gattinoni L, Coppola S, Cressoni M, et al. COVID-19 Does Not Lead to a "Typical" Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med 2020;201:1299-300. [Crossref] [PubMed]
  30. Gattinoni L, Chiumello D, Caironi P, et al. COVID-19 pneumonia: different respiratory treatments for different phenotypes? Intensive Care Med 2020;46:1099-102. [Crossref] [PubMed]
  31. Taleghani N, Taghipour F. Diagnosis of COVID-19 for controlling the pandemic: A review of the state-of-the-art. Biosens Bioelectron 2021;174:112830. [Crossref] [PubMed]
  32. Thompson BT, Chambers RC, Liu KD. Acute Respiratory Distress Syndrome. N Engl J Med 2017;377:562-72. [Crossref] [PubMed]
  33. Barton LM, Duval EJ, Stroberg E, et al. COVID-19 Autopsies, Oklahoma, USA. Am J Clin Pathol 2020;153:725-33. [Crossref] [PubMed]
  34. Thille AW, Esteban A, Fernández-Segoviano P, et al. Comparison of the Berlin definition for acute respiratory distress syndrome with autopsy. Am J Respir Crit Care Med 2013;187:761-7. [Crossref] [PubMed]
  35. Ware LB, Calfee CS. Biomarkers of ARDS: what's new? Intensive Care Med 2016;42:797-9. [Crossref] [PubMed]
  36. Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 2020;395:507-13. [Crossref] [PubMed]
  37. Sinha P, Matthay MA, Calfee CS. Is a "Cytokine Storm" Relevant to COVID-19? JAMA Intern Med 2020;180:1152-4. [Crossref] [PubMed]
  38. Terpstra ML, Aman J, van Nieuw Amerongen GP, et al. Plasma biomarkers for acute respiratory distress syndrome: a systematic review and meta-analysis*. Crit Care Med 2014;42:691-700. [Crossref] [PubMed]
  39. Moore JB, June CH. Cytokine release syndrome in severe COVID-19. Science 2020;368:473-4. [Crossref] [PubMed]
  40. Goodman RB, Pugin J, Lee JS, et al. Cytokine-mediated inflammation in acute lung injury. Cytokine Growth Factor Rev 2003;14:523-35. [Crossref] [PubMed]
  41. Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol 2004;202:145-56. [Crossref] [PubMed]
  42. Xu Z, Shi L, Wang Y, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med 2020;8:420-2. [Crossref] [PubMed]
  43. Bradley BT, Maioli H, Johnston R, et al. Histopathology and ultrastructural findings of fatal COVID-19 infections in Washington State: a case series. Lancet 2020;396:320-32. [Crossref] [PubMed]
  44. Helms J, Tacquard C, Severac F, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med 2020;46:1089-98. [Crossref] [PubMed]
  45. Goodwin CM, Hoffman JA. Deep vein thrombosis and stress ulcer prophylaxis in the intensive care unit. J Pharm Pract 2011;24:78-88. [Crossref] [PubMed]
  46. McClave SA, Martindale RG, Vanek VW, et al. Guidelines for the Provision and Assessment of Nutrition Support Therapy in the Adult Critically Ill Patient: Society of Critical Care Medicine (SCCM) and American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). JPEN J Parenter Enteral Nutr 2009;33:277-316. [Crossref] [PubMed]
  47. Abdelaal Ahmed Mahmoud A, Mahmoud HE, Mahran MA, et al. Streptokinase Versus Unfractionated Heparin Nebulization in Patients With Severe Acute Respiratory Distress Syndrome (ARDS): A Randomized Controlled Trial With Observational Controls. J Cardiothorac Vasc Anesth 2020;34:436-43. [Crossref] [PubMed]
  48. Hardaway RM, Harke H, Tyroch AH, et al. Treatment of severe acute respiratory distress syndrome: a final report on a phase I study. Am Surg 2001;67:377-82. [PubMed]
  49. Choudhury R, Barrett CD, Moore HB, et al. Salvage use of tissue plasminogen activator (tPA) in the setting of acute respiratory distress syndrome (ARDS) due to COVID-19 in the USA: a Markov decision analysis. World J Emerg Surg 2020;15:29. [Crossref] [PubMed]
  50. Wang J, Hajizadeh N, Moore EE, et al. Tissue plasminogen activator (tPA) treatment for COVID-19 associated acute respiratory distress syndrome (ARDS): A case series. J Thromb Haemost 2020;18:1752-5. [Crossref] [PubMed]
  51. Combes A, Hajage D, Capellier G, et al. Extracorporeal Membrane Oxygenation for Severe Acute Respiratory Distress Syndrome. N Engl J Med 2018;378:1965-75. [Crossref] [PubMed]
  52. Griffiths MJ, Evans TW. Inhaled nitric oxide therapy in adults. N Engl J Med 2005;353:2683-95. [Crossref] [PubMed]
  53. Adhikari NK, Dellinger RP, Lundin S, et al. Inhaled nitric oxide does not reduce mortality in patients with acute respiratory distress syndrome regardless of severity: systematic review and meta-analysis. Crit Care Med 2014;42:404-12. [Crossref] [PubMed]
  54. Raimondi N, Vial MR, Calleja J, et al. Evidence-based guidelines for the use of tracheostomy in critically ill patients. J Crit Care 2017;38:304-18. [Crossref] [PubMed]
  55. Sud S, Friedrich JO, Adhikari NK, et al. Effect of prone positioning during mechanical ventilation on mortality among patients with acute respiratory distress syndrome: a systematic review and meta-analysis. CMAJ 2014;186:E381-90. [Crossref] [PubMed]
  56. Guérin C, Reignier J, Richard JC, et al. Prone positioning in severe acute respiratory distress syndrome. N Engl J Med 2013;368:2159-68. [Crossref] [PubMed]
  57. Pan A, Liu L, Wang C, et al. Association of Public Health Interventions With the Epidemiology of the COVID-19 Outbreak in Wuhan, China. JAMA 2020;323:1915-23. [Crossref] [PubMed]
  58. Saguil A, Fargo MV. Acute Respiratory Distress Syndrome: Diagnosis and Management. Am Fam Physician 2020;101:730-8. [PubMed]
  59. Caputo ND, Strayer RJ, Levitan R. Early Self-Proning in Awake, Non-intubated Patients in the Emergency Department: A Single ED's Experience During the COVID-19 Pandemic. Acad Emerg Med 2020;27:375-8. [Crossref] [PubMed]
  60. Briel M, Meade M, Mercat A, et al. Higher vs lower positive end-expiratory pressure in patients with acute lung injury and acute respiratory distress syndrome: systematic review and meta-analysis. JAMA 2010;303:865-73. [Crossref] [PubMed]
  61. Alhazzani W, Møller MH, Arabi YM, et al. Surviving Sepsis Campaign: guidelines on the management of critically ill adults with Coronavirus Disease 2019 (COVID-19). Intensive Care Med 2020;46:854-87. [Crossref] [PubMed]
  62. Guan WJ, Zhong NS. Clinical Characteristics of Covid-19 in China. N Engl J Med 2020;382:1861-2. Reply. [PubMed]
  63. An P, Ji M, Ren H, et al. Prevention of COVID-19 in patients with inflammatory bowel disease in Wuhan, China. Lancet Gastroenterol Hepatol 2020;5:525-7. [Crossref] [PubMed]
  64. Ruan Q, Yang K, Wang W, et al. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med 2020;46:846-8. [Crossref] [PubMed]
  65. Bednarczyk JM, Fridfinnson JA, Kumar A, et al. Incorporating Dynamic Assessment of Fluid Responsiveness Into Goal-Directed Therapy: A Systematic Review and Meta-Analysis. Crit Care Med 2017;45:1538-45. [Crossref] [PubMed]
  66. Wang Y, Zhang D, Du G, et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet 2020;395:1569-78. [Crossref] [PubMed]
  67. Meta-analysis A. JAMA 2020;324:1330-41. WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group; Sterne JAC, Murthy S, et al Association Between Administration of Systemic Corticosteroids and Mortality Among Critically Ill Patients With COVID-19:. [Crossref] [PubMed]
  68. RECOVERY Collaborative Group. Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med 2021;384:693-704. [Crossref] [PubMed]
  69. COVID-19 treatment guidelines. Bethesda: National Institutes of Health. 2020. Available online: https://www.covid19treatmentguidelines.nih.gov/
  70. Tomazini BM, Maia IS, Cavalcanti AB, et al. Effect of Dexamethasone on Days Alive and Ventilator-Free in Patients With Moderate or Severe Acute Respiratory Distress Syndrome and COVID-19: The CoDEX Randomized Clinical Trial. JAMA 2020;324:1307-16. [Crossref] [PubMed]
  71. Angus DC, Derde L, Al-Beidh F, et al. Effect of Hydrocortisone on Mortality and Organ Support in Patients With Severe COVID-19: The REMAP-CAP COVID-19 Corticosteroid Domain Randomized Clinical Trial. JAMA 2020;324:1317-29. [Crossref] [PubMed]
  72. Wu C, Hou D, Du C, et al. Corticosteroid therapy for coronavirus disease 2019-related acute respiratory distress syndrome: a cohort study with propensity score analysis. Crit Care 2020;24:643. [Crossref] [PubMed]
  73. Prescott HC, Rice TW. Corticosteroids in COVID-19 ARDS: Evidence and Hope During the Pandemic. JAMA 2020;324:1292-5. [Crossref] [PubMed]
  74. Lewis SR, Pritchard MW, Thomas CM, et al. Pharmacological agents for adults with acute respiratory distress syndrome. Cochrane Database Syst Rev 2019;7:CD004477. [Crossref] [PubMed]
  75. Lee N, Allen Chan KC, Hui DS, et al. Effects of early corticosteroid treatment on plasma SARS-associated Coronavirus RNA concentrations in adult patients. J Clin Virol 2004;31:304-9. [Crossref] [PubMed]
  76. Arabi YM, Mandourah Y, Al-Hameed F, et al. Corticosteroid Therapy for Critically Ill Patients with Middle East Respiratory Syndrome. Am J Respir Crit Care Med 2018;197:757-67. [Crossref] [PubMed]
  77. Ye Z, Wang Y, Colunga-Lozano LE, et al. Efficacy and safety of corticosteroids in COVID-19 based on evidence for COVID-19, other coronavirus infections, influenza, community-acquired pneumonia and acute respiratory distress syndrome: a systematic review and meta-analysis. CMAJ 2020;192:E756-67. [Crossref] [PubMed]
  78. Matthay MA, Wick KD. Corticosteroids, COVID-19 pneumonia, and acute respiratory distress syndrome. J Clin Invest 2020;130:6218-21. [Crossref] [PubMed]
  79. Xu X, Han M, Li T, et al. Effective treatment of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci U S A 2020;117:10970-5. [Crossref] [PubMed]
  80. Roche initiates phase III clinical trial of Actemra/RoActemra plus remdesivir in hospitalised patients with severe COVID-19 pneumonia. 2020. Available online: https://www.roche.com/media/releases/med-cor-2020-05-28.htm
  81. Cavalli G, De Luca G, Campochiaro C, et al. Interleukin-1 blockade with high-dose anakinra in patients with COVID-19, acute respiratory distress syndrome, and hyperinflammation: a retrospective cohort study. Lancet Rheumatol 2020;2:e325-31. [Crossref] [PubMed]
  82. Iglesias-Julián E, López-Veloso M, de-la-Torre-Ferrera N, et al. High dose subcutaneous Anakinra to treat acute respiratory distress syndrome secondary to cytokine storm syndrome among severely ill COVID-19 patients. J Autoimmun 2020;115:102537. [Crossref] [PubMed]
  83. Hamilton JA. GM-CSF-Dependent Inflammatory Pathways. Front Immunol 2019;10:2055. [Crossref] [PubMed]
  84. De Luca G, Cavalli G, Campochiaro C, et al. GM-CSF blockade with mavrilimumab in severe COVID-19 pneumonia and systemic hyperinflammation: a single-centre, prospective cohort study. Lancet Rheumatol 2020;2:e465-73. [Crossref] [PubMed]
  85. Liu B, Bao L, Wang L, et al. Anti-IFN-γ therapy alleviates acute lung injury induced by severe influenza A (H1N1) pdm09 infection in mice. J Microbiol Immunol Infect 2021;54:396-403. [Crossref] [PubMed]
  86. Ranieri VM, Pettilä V, Karvonen MK, et al. Effect of Intravenous Interferon β-1a on Death and Days Free From Mechanical Ventilation Among Patients With Moderate to Severe Acute Respiratory Distress Syndrome: A Randomized Clinical Trial. JAMA 2020;323:725-33. [Crossref] [PubMed]
  87. Boretti A, Banik BK. Intravenous vitamin C for reduction of cytokines storm in acute respiratory distress syndrome. PharmaNutrition 2020;12:100190. [Crossref] [PubMed]
  88. Gao D, Xu M, Wang G, et al. The efficiency and safety of high-dose vitamin C in patients with COVID-19: a retrospective cohort study. Aging (Albany NY) 2021;13:7020-34. [Crossref] [PubMed]
  89. Grant WB, Lahore H, McDonnell SL, et al. Evidence that Vitamin D Supplementation Could Reduce Risk of Influenza and COVID-19 Infections and Deaths. Nutrients 2020;12:988. [Crossref]
  90. Quesada-Gomez JM, Entrenas-Castillo M, Bouillon R. Vitamin D receptor stimulation to reduce acute respiratory distress syndrome (ARDS) in patients with coronavirus SARS-CoV-2 infections: Revised Ms SBMB 2020_166. J Steroid Biochem Mol Biol 2020;202:105719. [Crossref] [PubMed]
  91. Mallis P, Michalopoulos E, Chatzistamatiou T, et al. Mesenchymal stromal cells as potential immunomodulatory players in severe acute respiratory distress syndrome induced by SARS-CoV-2 infection. World J Stem Cells 2020;12:731-51. [Crossref] [PubMed]
  92. Tovar I, Guerrero R, López-Peñalver JJ, et al. Rationale for the Use of Radiation-Activated Mesenchymal Stromal/Stem Cells in Acute Respiratory Distress Syndrome. Cells 2020;9:2015. [Crossref] [PubMed]
  93. Can A, Coskun H. The rationale of using mesenchymal stem cells in patients with COVID-19-related acute respiratory distress syndrome: What to expect. Stem Cells Transl Med 2020;9:1287-302. [Crossref] [PubMed]
  94. Hayes M, Masterson C, Devaney J, et al. Therapeutic efficacy of human mesenchymal stromal cells in the repair of established ventilator-induced lung injury in the rat. Anesthesiology 2015;122:363-73. [Crossref] [PubMed]
  95. Simonson OE, Mougiakakos D, Heldring N, et al. In Vivo Effects of Mesenchymal Stromal Cells in Two Patients With Severe Acute Respiratory Distress Syndrome. Stem Cells Transl Med 2015;4:1199-213. [Crossref] [PubMed]
  96. Wilson JG, Liu KD, Zhuo H, et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med 2015;3:24-32. [Crossref] [PubMed]
  97. Hashemian SR, Aliannejad R, Zarrabi M, et al. Mesenchymal stem cells derived from perinatal tissues for treatment of critically ill COVID-19-induced ARDS patients: a case series. Stem Cell Res Ther 2021;12:91. [Crossref] [PubMed]
  98. Qu W, Wang Z, Hare JM, et al. Cell-based therapy to reduce mortality from COVID-19: Systematic review and meta-analysis of human studies on acute respiratory distress syndrome. Stem Cells Transl Med 2020;9:1007-22. [Crossref] [PubMed]
  99. Tsuchiya A, Takeuchi S, Iwasawa T, et al. Therapeutic potential of mesenchymal stem cells and their exosomes in severe novel coronavirus disease 2019 (COVID-19) cases. Inflamm Regen 2020;40:14. [Crossref] [PubMed]
  100. O'Driscoll L. Extracellular vesicles from mesenchymal stem cells as a Covid-19 treatment. Drug Discov Today 2020;25:1124-5. [Crossref] [PubMed]
  101. Sengupta V, Sengupta S, Lazo A, et al. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev 2020;29:747-54. [Crossref] [PubMed]
  102. Shen C, Wang Z, Zhao F, et al. Treatment of 5 Critically Ill Patients With COVID-19 With Convalescent Plasma. JAMA 2020;323:1582-9. [Crossref] [PubMed]
  103. Li L, Zhang W, Hu Y, et al. Effect of Convalescent Plasma Therapy on Time to Clinical Improvement in Patients With Severe and Life-threatening COVID-19: A Randomized Clinical Trial. JAMA 2020;324:460-70. [Crossref] [PubMed]
  104. Tanne JH. Covid-19: FDA approves use of convalescent plasma to treat critically ill patients. BMJ 2020;368:m1256. [Crossref] [PubMed]
  105. Cao W, Liu X, Bai T, et al. High-Dose Intravenous Immunoglobulin as a Therapeutic Option for Deteriorating Patients With Coronavirus Disease 2019. Open Forum Infect Dis 2020;7:ofaa102. [Crossref] [PubMed]
  106. Alhazzani W, Møller MH, Arabi YM, et al. Surviving Sepsis Campaign: Guidelines on the Management of Critically Ill Adults with Coronavirus Disease 2019 (COVID-19). Crit Care Med 2020;48:e440-69. [Crossref] [PubMed]
  107. Wong CM, Lim KH, Liam CK. Assessment and management of chronic cough. Med J Malaysia 2002;57:515-23; quiz 524-5. [PubMed]
  108. Johnson MJ, Hui D, Currow DC. Opioids, Exertion, and Dyspnea: A Review of the Evidence. Am J Hosp Palliat Care 2016;33:194-200. [Crossref] [PubMed]
  109. HoaglandDAClarkeDJBMøllerRModulating the transcriptional landscape of SARS-CoV-2 as an effective method for developing antiviral compounds.Biorxiv 2020. doi: .10.1101/2020.07.12.199687
  110. Allard P, Lamontagne C, Bernard P, et al. How effective are supplementary doses of opioids for dyspnea in terminally ill cancer patients? A randomized continuous sequential clinical trial. J Pain Symptom Manage 1999;17:256-65. [Crossref] [PubMed]
  111. Pang GS, Qu LM, Tan YY, et al. Intravenous Fentanyl for Dyspnea at the End of Life: Lessons for Future Research in Dyspnea. Am J Hosp Palliat Care 2016;33:222-7. [Crossref] [PubMed]
  112. Woodcock AA, Gross ER, Gellert A, et al. Effects of dihydrocodeine, alcohol, and caffeine on breathlessness and exercise tolerance in patients with chronic obstructive lung disease and normal blood gases. N Engl J Med 1981;305:1611-6. [Crossref] [PubMed]
  113. Dhawan G, Kapoor R, Dhawan R, et al. Low dose radiation therapy as a potential life saving treatment for COVID-19-induced acute respiratory distress syndrome (ARDS). Radiother Oncol 2020;147:212-6. [Crossref] [PubMed]
  114. Candas D, Fan M, Nantajit D, et al. CyclinB1/Cdk1 phosphorylates mitochondrial antioxidant MnSOD in cell adaptive response to radiation stress. J Mol Cell Biol 2013;5:166-75. [Crossref] [PubMed]
  115. Li JJ. Mitigating Coronavirus-Induced Acute Respiratory Distress Syndrome by Radiotherapy. iScience 2020;23:101215. [Crossref] [PubMed]
  116. Calabrese EJ, Dhawan G, Kapoor R, et al. Radiotherapy treatment of human inflammatory diseases and conditions: Optimal dose. Hum Exp Toxicol 2019;38:888-98. [Crossref] [PubMed]
  117. Fatokun AA, Dawson VL, Dawson TM. Parthanatos: mitochondrial-linked mechanisms and therapeutic opportunities. Br J Pharmacol 2014;171:2000-16. [Crossref] [PubMed]
  118. Ke Y, Wang C, Zhang J, et al. The Role of PARPs in Inflammation-and Metabolic-Related Diseases: Molecular Mechanisms and Beyond. Cells 2019;8:1047. [Crossref] [PubMed]
  119. Curtin N, Bányai K, Thaventhiran J, et al. Repositioning PARP inhibitors for SARS-CoV-2 infection(COVID-19); a new multi-pronged therapy for acute respiratory distress syndrome? Br J Pharmacol 2020;177:3635-45. [Crossref] [PubMed]
  120. Vaschetto R, Kuiper JW, Musters RJ, et al. Renal hypoperfusion and impaired endothelium-dependent vasodilation in an animal model of VILI: the role of the peroxynitrite-PARP pathway. Crit Care 2010;14:R45. [Crossref] [PubMed]
  121. Kim JH, Suk MH, Yoon DW, et al. Inflammatory and transcriptional roles of poly (ADP-ribose) polymerase in ventilator-induced lung injury. Crit Care 2008;12:R108. [Crossref] [PubMed]
  122. Luo P, Liu Y, Qiu L, et al. Tocilizumab treatment in COVID-19: A single center experience. J Med Virol 2020;92:814-8. [Crossref] [PubMed]
  123. Sethi GS, Dharwal V, Naura AS. Poly(ADP-Ribose)Polymerase-1 in Lung Inflammatory Disorders: A Review. Front Immunol 2017;8:1172. [Crossref] [PubMed]
  124. Sethi GS, Sharma S, Naura AS. PARP inhibition by olaparib alleviates chronic asthma-associated remodeling features via modulating inflammasome signaling in mice. IUBMB Life 2019;71:1003-13. [Crossref] [PubMed]
  125. COVID-ICU Group on behalf of the REVA Network and the COVID-ICU Investigators. Clinical characteristics and day-90 outcomes of 4244 critically ill adults with COVID-19: a prospective cohort study. Intensive Care Med 2021;47:60-73. [Crossref]
  126. Gupta S, Hayek SS, Wang W, et al. Factors Associated With Death in Critically Ill Patients With Coronavirus Disease 2019 in the US. JAMA Intern Med 2020;180:1436-47. [Crossref] [PubMed]
  127. Grimaldi D, Aissaoui N, Blonz G, et al. Characteristics and outcomes of acute respiratory distress syndrome related to COVID-19 in Belgian and French intensive care units according to antiviral strategies: the COVADIS multicentre observational study. Ann Intensive Care 2020;10:131. [Crossref] [PubMed]
  128. Ioannou GN, Locke E, Green P, et al. Risk Factors for Hospitalization, Mechanical Ventilation, or Death Among 10 131 US Veterans With SARS-CoV-2 Infection. JAMA Netw Open 2020;3:e2022310. [Crossref] [PubMed]
  129. Bellan M, Patti G, Hayden E, et al. Fatality rate and predictors of mortality in an Italian cohort of hospitalized COVID-19 patients. Sci Rep 2020;10:20731. [Crossref] [PubMed]
  130. Yang X, Yu Y, Xu J, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med 2020;8:475-81. [Crossref] [PubMed]
  131. Rodriguez-Morales AJ, Cardona-Ospina JA, Gutiérrez-Ocampo E, et al. Clinical, laboratory and imaging features of COVID-19: A systematic review and meta-analysis. Travel Med Infect Dis 2020;34:101623. [Crossref] [PubMed]
  132. Hermans G, Van den Berghe G. Clinical review: intensive care unit acquired weakness. Crit Care 2015;19:274. [Crossref] [PubMed]
  133. Schefold JC, Bierbrauer J, Weber-Carstens S. Intensive care unit-acquired weakness (ICUAW) and muscle wasting in critically ill patients with severe sepsis and septic shock. J Cachexia Sarcopenia Muscle 2010;1:147-57. [Crossref] [PubMed]
  134. Schefold JC, Wollersheim T, Grunow JJ, et al. Muscular weakness and muscle wasting in the critically ill. J Cachexia Sarcopenia Muscle 2020;11:1399-412. [Crossref] [PubMed]
  135. Dziewas R, Warnecke T, Zürcher P, et al. Dysphagia in COVID-19 -multilevel damage to the swallowing network? Eur J Neurol 2020;27:e46-7. [Crossref] [PubMed]
  136. Zuercher P, Moret CS, Dziewas R, et al. Dysphagia in the intensive care unit: epidemiology, mechanisms, and clinical management. Crit Care 2019;23:103. [Crossref] [PubMed]
  137. Malas MB, Naazie IN, Elsayed N, et al. Thromboembolism risk of COVID-19 is high and associated with a higher risk of mortality: A systematic review and meta-analysis. EClinicalMedicine 2020;29:100639. [Crossref] [PubMed]
  138. Bime C, Casanova N, Oita RC, et al. Development of a biomarker mortality risk model in acute respiratory distress syndrome. Crit Care 2019;23:410. [Crossref] [PubMed]
  139. Pregernig A, Müller M, Held U, et al. Prediction of mortality in adult patients with sepsis using six biomarkers: a systematic review and meta-analysis. Ann Intensive Care 2019;9:125. [Crossref] [PubMed]
  140. Bime C, Camp SM, Casanova N, et al. The acute respiratory distress syndrome biomarker pipeline: crippling gaps between discovery and clinical utility. Transl Res 2020;226:105-15. [Crossref] [PubMed]
  141. Ding J, Karp JE, Emadi A. Elevated lactate dehydrogenase (LDH) can be a marker of immune suppression in cancer: Interplay between hematologic and solid neoplastic clones and their microenvironments. Cancer Biomark 2017;19:353-63. [Crossref] [PubMed]
  142. Henry BM, de Oliveira MHS, Benoit S, et al. Hematologic, biochemical and immune biomarker abnormalities associated with severe illness and mortality in coronavirus disease 2019 (COVID-19): a meta-analysis. Clin Chem Lab Med 2020;58:1021-8. [Crossref] [PubMed]
  143. Wu C, Chen X, Cai Y, et al. Risk Factors Associated With Acute Respiratory Distress Syndrome and Death in Patients With Coronavirus Disease 2019 Pneumonia in Wuhan, China. JAMA Intern Med 2020;180:934-43. [Crossref] [PubMed]
  144. Figueira Gonçalves JM, Hernández Pérez JM, Acosta Sorensen M, et al. Biomarkers of acute respiratory distress syndrome in adults hospitalised for severe SARS-CoV-2 infection in Tenerife Island, Spain. BMC Res Notes 2020;13:555. [Crossref] [PubMed]
  145. McGonagle D, Sharif K, O'Regan A, et al. The Role of Cytokines including Interleukin-6 in COVID-19 induced Pneumonia and Macrophage Activation Syndrome-Like Disease. Autoimmun Rev 2020;19:102537. [Crossref] [PubMed]
  146. Quijada H, Bermudez T, Kempf CL, et al. Endothelial eNAMPT amplifies pre-clinical acute lung injury: efficacy of an eNAMPT-neutralising monoclonal antibody. Eur Respir J 2021;57:2002536. [Crossref] [PubMed]
  147. Spadaro S, Fogagnolo A, Campo G, et al. Markers of endothelial and epithelial pulmonary injury in mechanically ventilated COVID-19 ICU patients. Crit Care 2021;25:74. [Crossref] [PubMed]
  148. van der Zee P, Rietdijk W, Somhorst P, et al. A systematic review of biomarkers multivariately associated with acute respiratory distress syndrome development and mortality. Crit Care 2020;24:243. [Crossref] [PubMed]

(English Language Editor: J. Jones)

Cite this article as: Pu D, Zhai X, Zhou Y, Xie Y, Tang L, Yin L, Liu H, Li L. A narrative review of COVID-19-related acute respiratory distress syndrome (CARDS): “typical” or “atypical” ARDS? Ann Transl Med 2022;10(16):908. doi: 10.21037/atm-22-3717

Download Citation