Tuberc Respir Dis > Volume 85(4); 2022 > Article
Yoon and Uh: Post-Coronavirus Disease 2019 Pulmonary Fibrosis: Wait or Needs Intervention

Abstract

Coronavirus disease 2019 (COVID-19) has become a major health burden worldwide, with over 450 million confirmed cases and 6 million deaths. Although the acute phase of COVID-19 management has been established, there is still a long way to go to evaluate the long-term clinical course or manage complications due to the relatively short outbreak of the virus. Pulmonary fibrosis is one of the most common respiratory complications associated with COVID-19. Scarring throughout the lungs after viral or bacterial pulmonary infection have been commonly observed, but the prevalence of post- COVID-19 pulmonary fibrosis is rapidly increasing. However, there is limited information available about post-COVID-19 pulmonary fibrosis, and there is also a lack of consensus on what condition should be defined as post-COVID-19 pulmonary fibrosis. During a relatively short follow-up period of approximately 1 year, lesions considered related to pulmonary fibrosis often showed gradual improvement; therefore, it is questionable at what time point fibrosis should be evaluated. In this review, we investigated the epidemiology, risk factors, pathogenesis, and management of post-COVID-19 pulmonary fibrosis.

Introduction

Coronavirus disease 2019 (COVID-19), caused by the novel coronavirus strain severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has continued to spread throughout the world, disrupting our daily lives worldwide since it first broke out in Wuhan, China, in 2019. As of March 2022, more than 450 million cumulative confirmed cases and more than 6 million cumulative deaths have been reported globally due to COVID-19 to date. As the COVID-19 pandemic continues for more than 2 years, the long-term prognosis and clinical course of COVID-19 survivors is attracting attention in addition to the management of the acute phase. Certain people complain of various symptoms for several weeks after acute SARS-CoV-2 infection, which is collectively called post-acute COVID-19 syndrome or “long COVID [1].” Post-acute COVID-19 affects several organ systems; however, the respiratory system is most commonly involved. COVID-19 survivors complain of a spectrum of respiratory symptoms, ranging from mild dyspnea to difficulty weaning from ventilator, due to lung sequelae [2,3].
In COVID-19-related acute respiratory distress syndrome (ARDS) survivors, fatal respiratory complications including failure of withdrawing oxygen supply and home ventilation are frequently observed [4,5]. These complications are presumed to be related to fibrotic lung damage from SARS-CoV-2. Lung sequelae can occur after pulmonary infection; however, fibrotic changes in the lungs after COVID-19 are more frequently observed (range, 20%-70%) [6-14]. However, the clinical course of patients with fibrotic changes induced by COVID-19 may be different from that of patients diagnosed with progressive fibrosing interstitial lung disease (PF-ILD). The radiological features of pulmonary fibrosis generally include distinctive patterns such as architectural distortion, traction bronchiectasis/bronchiolectasis, parenchymal band, reticulation, and honeycombing on computed tomography (CT) [15]. In patients with PF-ILD, fibrotic lesions undergo progression or stabilization, whereas fibrotic lesions gradually improve after pulmonary infection. Therefore, it is challenging to detect pulmonary fibrosis after pulmonary infection. Therefore, previous studies reporting pulmonary fibrosis after COVID-19 favor the term “fibrosis-like pattern” over the term “fibrosis.” In this study, we reviewed post-COVID-19 pulmonary fibrosis with various aspects.

Definition and Epidemiology

The incidence of post-COVID-19 pulmonary fibrosis varies depending on several factors such as population characteristics, fibrosis definition, and observational duration (Table 1). Post-COVID-19 pulmonary fibrosis was mostly characterized by radiological features including reticulation, traction bronchiectasis, parenchymal band, architectural distortion, and honeycombing change, which are also generally considered fibrotic lesions in PF-ILD. A study by Zou et al. [16] revealed that fibrosis, defined as the presence of ground-glass opacity (GGO), evaluated by artificial intelligence-assisted high-resolution computed tomography (HR-CT), was present in 84% of COVID-19 patients at discharge CT. However, the proportion of patients with GGO was 82%, whereas the proportions of patients with reticulation and honeycombing were 30% and 36%, respectively [16]. Since the incidence of post-COVID-19 pulmonary fibrosis differs depending on which lesion is fibrotic, a universal consensus on its definition can help standardize the study.
According to the timing of follow-up, the incidence of post-COVID-19 pulmonary fibrosis was relatively high at midterm (6-9 months) follow-up, whereas it was slightly lower at long-term (more than 1 year) follow-up. Li et al. [13] reported that in 287 patients diagnosed with COVID-19 pneumonia, fibrotic lesion was detected in 88%, 74%, 80%, 68%, and 62% patients, whereas the resolution of fibrotic lesion was observed in 5%, 20%, 18%, 38%, and 49% at 0-30, 31-60, 61-90, 91-120, and >120 days after disease onset. In addition, the proportion of GGO was greatly decreased from 91% in 31-60 days to 65% in >120 days, whereas the proportion of parenchymal bands (95% in 31-60 days and 94% in >120 days) and traction bronchiectasis (14% in 31-60 days and 14% in >120 days) remained stable after the 30-day follow-up [13]. However, in a prospective Italian study (n=118), the proportion of fibrosis-like lesion including reticular pattern with or without honeycombing significantly increased from 55% at baseline to 72%, at the 6-month follow-up [7]. However, in a long-term follow-up Chinese study (n=64), the proportion of reticular abnormalities and traction bronchiectasis did not significantly change from 6 months to 1 year of follow-up [17]. In 209 patients with COVID pneumonia including 22 with ARDS, the most frequently observed predominant CT lesion was GGO during the 1-year follow-up, but the predominant rate of GGO gradually decreased from 83% in discharge CT to 22% after the 1-year follow-up [18]. Meanwhile, the predominance of reticular abnormalities that first appeared at 11% on discharge CT reduced to 3% at 3 months and was maintained during the 1-year follow-up, showing similar rates at the 7-month and 12-month follow-up of 2% and 2%, respectively [18]. Parenchymal bands, another radiological finding of fibrosis, was were confirmed at 0.5%, after the 3-month follow-up; however, it increased to 1% at the 7-month follow-up and remained unchanged until the 1-year follow-up [18]. These findings collectively suggested that as GGOs improve comprehensively, some of them may change to fibrotic lesions because of scarring of the lungs between the 3-month and 6-month follow-up after COVID-19 pneumonia. Once fibrotic lesions occur, they do not improve between the mid- to long-term follow-up, and additional follow-up is needed to identify its accurate incidence. In a 15-year prospective chest CT follow-up of patients with SARS-CoV-1 on April 2003 (n=27), abnormal CT lesions gradually decreased from 2003 to 2018, but remained stable during 2004 and 2018 [19]. This finding suggests that the specific part of pulmonary fibrosis after COVID-19 may be difficult to spontaneously improve without progression.
The characteristics of the included study population is noteworthy. There is a possibility that the incidence of fibrosis is higher in studies that include more patients with severe pneumonia, particularly in patients receiving mechanical ventilation [20]. Therefore, when estimating the incidence of post-COVID-19 pulmonary fibrosis, an approach based on the stratification of fibrosis risk factors, including disease severity, is needed.

Risk Factors

Several risk factors for post-COVID-19 pulmonary fibrosis associated with an individual and disease itself have been proposed in previous studies (Table 2) [6,7,11,12,14,16,20-24]. Aul et al. [20] demonstrated that in COVID-19 patients with available chest CT at 6-week follow-up (n=117), application of invasive ventilation (adjusted odds ratio [OR], 3.48; 95% confidence interval [CI], 1.16-10.49) and persistent breathlessness (adjusted OR, 5.25; 95% CI, 1.86-14.81) were independent risk factors for post-COVID-19 pulmonary fibrosis after adjusting for covariates with a p-value of <0.05 in univariate analysis (male sex, persistent myalgia, peak white blood cell [WBC] count, and high-risk chest X-ray during inpatient COVID-19 admission). Yasin et al. [14] showed that old age (OR, 3.37; 95% CI, 0.76-14.55), CT severity score for opacity estimation (OR, 2.38; 95% CI, 1.18-4.41), presence of consolidation on the initial CT (OR, 1.91; 95% CI, 0.63-4.35), elevated D-dimer level (OR, 1.98; 95% CI, 1.01-10.19), and admission to intensive care unit (ICU) (OR, 6.77; 95% CI, 1.77-25.88) were significantly associated with the occurrence of post-COVID-19 pulmonary fibrosis according to multivariable analysis at median 1.5-month follow-up after discharge in a retrospective Egyptian study (n=21). Zou et al. [16] reported that increased interleukin (IL)-6 level in the acute phase (hazard ratio [HR], 1.081; 95% CI, 1.021-1.144) and decreased serum albumin level (HR, 0.821; 95% CI, 0.734-0.918) were independently associated with post-COVID-19 pulmonary fibrosis in patients with COVID-19 pneumonia (n=248) according to discharge HR-CT. In a prospective 3-month follow-up study (n=173), severe COVID-19 pneumonia (oxygen saturation <90% on room air or signs of severe respiratory distress) according to the World Health Organization classification (OR, 2.40; 95% CI, 1.27-4.51) and the presence of consolidation on the initial CT (OR, 2.84; 95% CI, 1.20-6.73) were significantly associated with the development of post-COVID-19 pulmonary fibrosis according to multivariable analysis [12].
A 6-month follow-up prospective study (n=118) by Caruso et al. [7] revealed that male sex (OR, 0.03; 95% CI, 0.00-0.89), cough (OR, 0.08; 95% CI, 0.01-0.88), lymphocytosis (OR, 0.08; 95% CI, 0.01-0.86), and quantitative chest CT-based well-aerated lung volume (OR, 0.44; 95% CI, 0.01-1.19) were independent protective factors of fibrotic-like changes on multivariable analysis. Yu et al. [22] exhibited that in a retrospective Chines study (n=32; median days from discharge to last follow-up, 9 days), fibrosis was observed in 44% of patients, and patients with fibrosis showed more frequent abnormal radiological features including interstitial thickening, air bronchogram, irregular interface, coarse reticular pattern, and parenchymal bands on worst CT images than those without fibrosis. Although it is not known to be directly related to the development of fibrosis, cigarette smoking and alcohol consumption are potential risk factors for post-COVID-19 pulmonary fibrosis because of their association with the progression of severe pneumonia [25,26]. Overall, post-COVID-19 pulmonary fibrosis can frequently develop in patients with risk factors such as old age, multiple comorbidities, severe pneumonia, and invasive ventilation. Because the previous studies included heterogeneous patients with COVID-19 pneumonia and the definition of fibrosis as well as the observation period was also different, further long-term studies using a common fibrosis criterion are needed to identify risk factors for post-COVID-19 pulmonary fibrosis.

Pathogenesis

The pathogenesis of post-COVID-19 pulmonary fibrosis has not yet been clarified, and multiple pathways are thought to be involved. In idiopathic pulmonary fibrosis (IPF), abnormal wound healing processes, including injury, inflammation, and repair, are key pathways for fibrosis [27]. Acute lung injuries destruct the basement membrane (BM) of the alveolar-capillary barriers and induce inflammatory responses by releasing fibroblast-activating cytokines, chemokines, and growth factors from the recruited inflammatory cells and fibroblasts. Substantial repair processes including angiogenesis and fibroblast activation lead to the deposition of the extracellular matrix (ECM). In normal wound healing response, the repair integrity of the alveolar-capillary barrier BM terminates collagen disposition by activating the fibrolytic process [28]. However, when BM fails to restore its integrity due to persistent or severe injury, fibroblasts and inflammatory pathways are continuously activated, resulting in ECM deposition [29]. These processes collectively contribute to destroyed lung architecture and scarring with fibrosis. Due to an imbalanced wound healing response, excessive deposition of ECM is key to progressive fibrosis in IPF.

1. Inflammatory markers

Severity of pneumonia is regarded as a risk factor for post-COVID-19 pulmonary fibrosis [6,11,12-14]. A study by Huang et al. [11] found that the post-COVID-19 fibrosis group had elevated levels of serum inflammatory markers including WBC, neutrophils, D-dimer, C-reactive protein (CRP), and procalcitonin compared with the non-fibrosis group among patients with COVID pneumonia (n=81). Furthermore, the presence of ARDS, disease severity of pneumonia, or serum inflammatory markers were independent prognostic factors for fibrosis secondary to COVID-19 [6,7,12,14,16]. Therefore, hyperinflammatory status induced by SARS-CoV-2 might be a key pathogenesis of fibrosis. The “cytokine storm” is a phenomenon in which excessive production of proinflammatory cytokines is caused by the hyperimmune reaction of the host to SARS-CoV-2 [30]. The three most crucial mediators of the cytokine storm due to COVID-19 are IL-1, IL-6, and tumor necrosis factor-α (TNF-α). In a Chinese study by Huang et al. [31] (n=41), the initial plasma levels of IL-1β and TNF-α were significantly higher in patients with COVID-19 than in the healthy population, and the level of TNF-α particularly increased in ICU patients (n=13) compared with that in non-ICU patients (n=28). Another Chines study by Chen et al. [32] (n=21) showed that patients with severe COVID-19 had elevated levels of IL-6 and TNF-α as well as increased alanine aminotransferase, lactate dehydrogenase (LDH), CRP, and D-dimer levels and decreased albumin and lymphocyte levels compared with patients with moderate COVID-19. These laboratory findings are similar to the predictive factors for post-COVID-19 pulmonary findings [7,14,16]. TNF-α induces loss of expression on fibroblast Thy-1 surface, leading to myofibroblast differentiation [33]. In addition, TNF-α expression increased in the lungs in IPF [34] and anti-TNF-α antibody diminished pulmonary fibrosis in both bleomycin-induced and silica-induced pulmonary fibrosis in a mouse model [35,36]. In COVID-19, IL-6 level is higher in severe ICU cases and non-survived patients [37-39]. IL-6 stimulates the profibrotic pathway in fibroblasts in patients with IPF, while it enhances apoptosis pathway in normal fibroblasts [40]. Circulating IL-6 levels significantly increased in bleomycin-induced pulmonary fibrosis mice than that in control mice [41]. In vitro, the pathway mediated by soluble IL-6Rα activates proliferation of fibroblasts and production of ECM proteins [42]. Among the cytokines released during COVID-19, profibrotic cytokines were also included, leading to pulmonary fibrosis.
Krebs von den Lungen-6 antigen (KL-6) is one of the biomarkers for ARDS and interstitial lung disease, disrupting the alveolar-capillary barrier [43,44]. Peng et al. [45] reported that the initial serum KL-6 level was significantly higher in patients with fibrosis at discharge CT (n=19) than in patients without fibrosis (n=94). They also observed that serum KL-6 level was elevated in severe cases and not in moderate and mild cases compared with that in controls [45]. Collectively, these findings suggest that patients with severe disease enough to destroy the alveolar-capillary barrier are susceptible to the occurrence of post-COVID-19 pulmonary fibrosis. Therefore, the integrity of the alveolar-capillary barrier may be one of the factors in the pathogenesis of post-COVID-19 pulmonary fibrosis, similar to that in IPF.

2. Renin-angiotensin-aldosterone system

SARS-CoV-2 invades human cells by binding to angiotensin-converting enzyme (ACE) 2, which is a functional receptor mostly expressed on the surface of heart, kidney, and lung cells [46]. Once infected with the virus, the activity of ACE, which is a key regulator of the reninangiotensin-aldosterone system (RAAS), is downregulated by various mechanisms. ACE2 converts angiotensin II to its physiological antagonist angiotensin-(1-7), resulting in vasodilatory, anti-inflammatory, anti-fibrotic, and anti-apoptotic effects [46,47]. Consequently, the activation of vasoconstrictor, proinflammatory, profibrotic, and proliferative effects by the activation of angiotensin II may contribute to post-COVID-19 pulmonary fibrosis. In addition, Xu et al. [48] confirmed that mRNA transcripts of transforming growth factor beta-1 and connective tissue growth factor were significantly elevated in the alveolar epithelium following SARS-CoV-2 binding to ACE2. Because transforming growth factor β (TGF-β) is a potent profibrotic cytokine that induces myofibroblast differentiation, type II alveolar epithelial cell apoptosis, ECM remodeling, it is considered to play a key role in lung fibrogenesis in IPF [49,50]. Therefore, due to SARS-CoV-2 infection, RAAS imbalance may cause post-COVID-19 pulmonary fibrosis by activating the TGF-β pathway.

3. Oxidative stress

Oxidative stress can also contribute to the fibrogenic pathway in post-COVID-19 pulmonary fibrosis. ARDS, which requires high oxygen supply, is a predictive factor for post-COVID-19 pulmonary fibrosis [6]. A case-control study by Farghaly et al. [51] (n=64) demonstrated that patients with COVID-19 undergoing long-term oxygen therapy showed significantly higher CT score, which quantitatively evaluates the extent of abnormal lesion on chest CT than those in patients not undergoing long-term oxygen therapy on chest CT at 1 month (9.6 [oxygen] vs. 6.1 [no-oxygen], p=0.014), and 6 months (4.7 vs. 2.0, p=0.037), after discharge. Hyperoxia induces reactive oxygen species generation in mitochondria [52], with inhibition of oxidative phosphorylation and reduction of adenosine triphosphate level [53], similar to IPF pathogenesis in mitochondria [54]. Meanwhile, in a 4-month follow-up U.S. study (n=76), 10% decrease in an age-adjusted telomere length increased 35% development of fibrotic-like pattern (95% CI, 1.06-1.72) along with sequential organ failure assessment score, LDH level, and duration of invasive ventilation on multivariable analysis [24]. Telomere shortening or mutation is associated with the development of IPF [55]. Taking these points into account, it is possible that post-COVID-19 pulmonary fibrosis may also share a common pathogenetic mechanism with IPF. However, unlike IPF where the abnormal healing process persists due to repeated lung injury, post-COVID-19 pulmonary fibrosis does not progress even if the fibrosis is not significantly improved because additional lung damage does not occur once the active infection subsides. Further clinical studies may be useful to confirm the pathogenesis of post-COVID-19 pulmonary fibrosis.

Clinical Manifestation

Patients with post-COVID-19 pulmonary fibrosis exhibit a wide spectrum of clinical manifestations, ranging from asymptomatic to failure of withdrawing oxygen supply, depending on the extent of fibrosis, follow-up timing, or comorbidities. The most commonly observed symptoms after COVID-19 include fatigue, cough, dyspnea, and sleep disturbance [56,57]. Li et al. [13] reported that at 3-month follow-up, the proportion of symptomatic patients was the highest at 44%, followed by fatigue (21%), cough (15%), and exercise limitation (9%) in patients with fibrosis (n=114). Another 3-month follow-up study revealed that 33.3% patients with had grade ≥2 dyspnea among patients with COVID-19 (n=48) [8]. However, post-COVID-19-related symptoms tend to gradually improve over time [17,51,56,57]. Farghaly et al. [51] demonstrated that the overall mean dyspnea scores for survived patients with post-COVID-19 pulmonary fibrosis at discharge and at the 6-month follow-up were 2.8 and 1.1, respectively. A study by Han et al. [17] also observed that rate of patients with sputum expectoration significantly decreased from 15% in 6 months to 5% in 1 year among patients with severe COVID-19 (n=62). Although statistical significances were not found, the proportion of patients with dry cough (8% in 6 months and 5% in 1 year) and exertional dyspnea (21% in 6 months and 15% in 1 year) also numerically decreased [17].
The most common abnormality in pulmonary function test in post-COVID-19 was impairment of diffusing capacity for carbon monoxide (DLCO) [8,24,58,59]. At 3-month follow-up following discharge, out of the total patients (n=48), 91.3% had fibrosis on chest CT and DLCO was slightly impaired at 61.0% predicted, while other lung functions including forced vital capacity (FVC) and forced expiratory volume per 1 second (FEV1) were within the normal range [8]. At the 4-month follow-up (n=76), patients with fibrotic-like pattern (n=32) also showed decreased DLCO and more weight loss than patients with non-fibrotic-like changes (n=13) or normal CT images (n=31) [24]. Since DLCO was significantly correlated with the extent of traction bronchiectasis (r=−0.49) or reticulation (r=−0.64) on chest CT in patients after COVID-19 [24], DLCO impairment can be frequently observed particularly in patients with post-COVID-19 pulmonary fibrosis. Other lung function abnormalities in FVC, FEV1, and total lung capacity (TLC) can be observed [23,57,60]. In an Italian retrospective study, patients with fibrosis (n=23) had a lower FVC predicted value (50% predicted [fibrosis] vs. 90% predicted [non-fibrosis], p<0.001) compared with patients without fibrosis (n=67) at the 8-week follow-up [60].
Like the symptoms, abnormal lung function also gradually recovered over time [56,57]. The proportion of patients with DLCO of <80% predicted significantly decreased from 31% in 60 days to 21% in 100 days after COVID-19 diagnosis in a prospective, multicenter, observational study (n=145) [56]. Meanwhile, in a Chinese study, the number of patients with impaired lung function including FEV1, FVC, and TLC decreased overtime during 1-year follow-up, particularly that of patients receiving high-flow nasal cannula, invasive or non-invasive mechanical ventilation [57]. However, the proportion of patients with impaired DLCO (<80% predicted) did not change significantly, rather increased numerically from the 6-month to 12-month follow-up [57]. These findings suggested that pulmonary function deterioration due to pulmonary fibrosis may not easily recover during midterm follow-up.

Treatment and Prevention

1. Anti-fibrotics

To date, no effective method has been demonstrated for the treatment or prevention of post-COVID-19 pulmonary fibrosis. Anti-fibrotic agents for the treatment of IPF have been proposed. In IPF, pirfenidone and nintedaninb are recommended to reduce the decline rate of FVC based on clinical trials [61,62]. Nintedanib is a selective tyrosine kinase inhibitor for the receptors of vascular endothelial growth factor, platelet-derived growth factor, and fibroblast growth factor [63], which were significantly elevated in patients with COVID-19 [31]. In a small Japanese interventional study including patients with ventilated COVID-19 pneumonia, the rate of high-attenuation areas on chest CT were not different at the initiation of mechanical ventilation, but significantly lower in patients on nintedanib (n=30) compared with that in controls (n=30), after extubation [64]. In addition, some cases in which fibrosis improved after nintedanib administration have been reported [65,66]. Pirfenidone inhibits profibrotic pathways mostly targeting TGF-β and its downstream pathways, leading to blockage of fibroblast proliferation, myofibroblast transdifferentiation, and collagen deposition [67]. Pirfenidone also has broad anti-inflammatory and anti-oxidative effects by modulating anti-inflammatory cells and cytokines. Zhang et al. [68] revealed that in patients with severe COVID-19 pneumonia, no significant differences in chest CT images between the pirfenidone (n=73) and placebo group (n=73) at 4 weeks of treatment, but pirfenidone group showed lower levels of inflammatory markers including IL-2R, TNF-α, and D-dimer. Other clinical trials showing that anti-fibrotic agents inhibit various profibrotic pathways such as bisamide derivative of dicarboxylic acid, LYT-100, and collagen-polyvinylpyrrolidone and are ongoing [69]. Further long-term clinical trials in different populations can verify the effect of anti-fibrotic agents.

2. Immunomodulatory drugs

There are methods to modify disease-related risk factors to prevent the occurrence of post-COVID-19 pulmonary fibrosis. In particular, because the severity of pneumonia or the occurrence of ARDS is related to fibrosis after COVID-19 [6,7,12,14,16], the administration of corticosteroids or IL-6 receptor inhibitors may be considered to prevent fibrosis by reducing the severity of pneumonia [70,71]. In selective patients with persistent post-COVID-19 interstitial change (n=30), a 6-week prolonged use of corticosteroids improved the clinical course, including lung function and exercise capacity, with tolerability [72]. However, the appropriate dosage and duration of steroids in post-COVID-19 condition remain unclear. In addition, treatment with mesenchymal stem cells, spironolactone, long-term oxygen therapy, and ozone therapy are underway to improve fibrosis.

3. Rehabiltation

Pulmonary rehabilitation can improve lung function, exercise capacity, dyspnea score, and quality of life in patients with ILD [73]. According to a Chinese open randomized controlled study including patients with a post-COVID-19 condition (≥6 months after diagnosis), those who underwent pulmonary rehabilitation (n=36) had a significantly improved lung function, exercise capacity, quality of life, and anxiety/depression assessment score at 6 weeks compared with those before treatment or compared with controls (n=36) [74]. Pulmonary rehabilitation is beneficial in many aspects for patients with a history of ICU admission for respiratory failure, but there is insufficient evidence whether these results can be equally generalized to patients recovering from COVID-19 [75]. Lung transplantation can be an alternative option for end-stage patients with post-COVID-19 pulmonary fibrosis [76,77].
Several ongoing clinical trials have been registered at ClinicalTrials.gov for identifying the efficacy of agents to prevent or diminish secondary fibrogenesis after COVID-19 (Table 3) [78-90]. The efficacy of several trial interventions is being investigated, and the results are expected to be confirmed over a period of months to years.

Conclusion

Approximately 2 years have passed since the outbreak of COVID-19 in 2019, but it is insufficient to evaluate complications. A consensus on the definition of post-COVID-19 pulmonary fibrosis should precede further investigation of its pathogenesis, incidence, and treatment. Furthermore, it is necessary to establish management by observing the results of interventions currently in progress for fibrosis.

Notes

Authors’ Contributions

Conceptualization: Yoon HY, Uh ST. Methodology: Yoon HY. Investigation: Yoon HY. Writing - original draft preparation: Yoon HY. Writing - review and editing: Yoon HY, Uh ST. Approval of final manuscript: all authors.

Conflicts of Interest

No potential conflict of interest relevant to this article was reported.

Funding

This study was supported by the Soonchunhyang University Research Fund.

Table 1.
Definition and incidence of post-COVID-19 pulmonary fibrosis
Study Study design and subject Site Definition Follow-up Incidence (%)
Zou et al. (2021) [16] Retrospective, n=284 China Ground-glass opacity, linear opacity, interlobular septal thickening, reticulation, honeycombing, or bronchiectasis At discharge 84
Yasin et al. (2021) [14] Retrospective, n=210 Egypt NA Median 42 days after discharge 48
ICU admission: 52 (25%)
van Gassel et al. (2021) [8] Retrospective, n=48 Netherland Coarse fibrous bands with or without obvious parenchymal distortion 3 months after discharge 67
MV: 48 (100%) Bronchiectasis/bronchiolectasis
Gulati and Lakhani (2021) [9] Retrospective, n=12 USA Architectural distortion with traction bronchiectasis or honeycombing 3 months after symptoms onset 58-100
MV: 6 (50%)
COMEBAC Study Group (2021) [10] Prospective, n=117 France NA 3-4 months after discharge 19
MV: 51 (28.8%)
Huang et al. (2021) [11] Prospective, n=81 China Parenchymal bands, irregular interfaces Median 58 days after discharge 52
NIV: 21 (25.9%) Reticular opacities
MV: 14 (17.3%) Traction bronchiectasis ± honeycombing
ICU: 45 (55.6%)
Nabahati et al. (2021) [12] Prospective, n=173 Iran Traction bronchiectasis, honeycombing, parenchymal bands, interlobar septal thickening 3, 6 months after discharge 3 mo: 52
6 mo: 68
Li et al. (2021) [13] Retrospective, n=287 China Parenchymal bands, irregular interfaces, reticulation, traction bronchiectasis 3-5 months after symptom onset 2-3 mo: 80
3-4 mo: 68
>4 mo: 62
Han et al. (2021) [6] Prospective, n=114 China Traction bronchiectasis 6 months after diagnosis 35
NIV: 24 (21%) Parenchymal bands ± honeycomb
MV: 4 (3.5%)
Caruso et al. (2021) [7] Prospective, n=118 Italy Reticular pattern ± honeycombing 6 months after diagnosis 72
NIV: 53 (61%)
MV: 34 (39%)
Han et al. (2021) [17] Prospective, n=62 China Reticular abnormalities, traction bronchiectasis 6, 12 months after discharge 6 mo: 44-58
12 mo: 44-52

COVID-19: coronavirus disease 2019; ICU: intensive care unit; NA: not available; MV: mechanical ventilator; NIV: non-invasive pulmonary ventilator.

Table 2.
Potential risk factor for post-COVID-19 pulmonary fibrosis
Risk factor
Demographics Sex
Old age
Smoking
Alcohol abuse
Comorbidities (diabetes, obesity, hypertension, chronic lung diseases, chronic liver disease, cardiovascular diseases, and cerebrovascular disease)
Length of telomere
Disease-related factors Use of high-flow oxygen or invasive or non-invasive ventilation
Long duration of hospital or ICU stay
Unstable initial vital signs (tachycardia, high fever, tachypnea)
Presence of ARDS
Absence of cough as the initial presentation
Persistent dyspnea
Laboratory factors IL-6, albumin, WBC, lymphocyte, neutrophil, eosinophil, NLR, total bilirubin, CRP, LDH, D-dimer, BNP, procalcitonin
Radiological factors Extent of abnormal lesion in initial chest CT
Presence of consolidation, reticulation, parenchymal band, interstitial thickening, irregular interface, pleural effusion, and poor-aerated lung volume

COVID-19: coronavirus disease 2019; ICU: intensive care unit; ARDS: acute respiratory distress syndrome; IL: interleukin; WBC: white blood cell; NLR: neutrophil-lymphocyte ratio; CRP: C-reactive protein; LDH: lactic dehydrogenase; BNP: B-type natriuretic peptide; CT: computed tomography.

Table 3.
Ongoing clinical trial numbers for treatment of post-COVID-19 pulmonary fibrosis
Medication NCT No. Site Status
Nintedanib NCT04541680 [78] France Recruiting, Phase 3
NCT04856111 [79] India Recruiting, Phase 4
NCT04619680 [80] USA Recruiting, Phase 4
Pirfenidone NCT04607928 [81] Spain Recruiting, Phase 2
NCT04856111 [79] India Recruiting, Phase 4
NCT04607928 [82] China Recruiting, Phase 3
Treamid NCT04527354 [83] Russia Completed, Phase 2
Cholchicine NCT04818489 [84] Egypt Completed, Phase 2
EV-Pure+WJ-Pure NCT05387239 [85] USA Recruiting, Phase 1
Sirolimus NCT04948203 [86] USA Recruiting, Phase 2 and 3
Mineralocorticoid receptor antagonist NCT04912011 [87] Poland Recruiting, Phase 4
Autologous monocytes NCT04805086 [88] UK Recruiting, Phase 1 and 2
LYT-100 NCT04652518 [89] USA, Argentina, Brazil, Moldova, Republic of Philippines, Romania, Ukraine, UK Recruiting, Phase 2
Collagen-Polyvinylpyrrolidone NCT0451716213 [90] Mexico Recruiting, Phase 1 and 2

COVID-19: coronavirus disease 2019.

References

1. Nalbandian A, Sehgal K, Gupta A, Madhavan MV, McGroder C, Stevens JS, et al. Post-acute COVID-19 syndrome. Nat Med 2021;27:601-15.
crossref pmid pmc pdf
2. Carfi A, Bernabei R, Landi F; Gemelli Against COVID-19 Post-Acute Care Study Group. Persistent symptoms in patients after acute COVID-19. JAMA 2020;324:603-5.
crossref pmid pmc
3. Tenforde MW, Kim SS, Lindsell CJ, Billig Rose E, Shapiro NI, Files DC, et al. Symptom duration and risk factors for delayed return to usual health among outpatients with COVID-19 in a multistate health care systems network - United States, March-June 2020. MMWR Morb Mortal Wkly Rep 2020;69:993-8.
crossref pmid pmc
4. Chopra V, Flanders SA, O’Malley M, Malani AN, Prescott HC. Sixty-day outcomes among patients hospitalized with COVID-19. Ann Intern Med 2021;174:576-8.
crossref pmid pmc
5. Martin-Villares C, Perez Molina-Ramirez C, Bartolome-Benito M, Bernal-Sprekelsen M; COVID ORL ESP Collaborative Group. Outcome of 1890 tracheostomies for critical COVID-19 patients: a national cohort study in Spain. Eur Arch Otorhinolaryngol 2021;278:1605-12.
pmid
6. Han X, Fan Y, Alwalid O, Li N, Jia X, Yuan M, et al. Six-month follow-up chest CT findings after severe COVID-19 pneumonia. Radiology 2021;299:E177-86.
crossref pmid pmc
7. Caruso D, Guido G, Zerunian M, Polidori T, Lucertini E, Pucciarelli F, et al. Post-acute sequelae of COVID-19 pneumonia: six-month chest CT follow-up. Radiology 2021;301:E396-405.
crossref pmid pmc
8. van Gassel RJ, Bels JL, Raafs A, van Bussel BC, van de Poll MC, Simons SO, et al. High prevalence of pulmonary sequelae at 3 months after hospital discharge in mechanically ventilated survivors of COVID-19. Am J Respir Crit Care Med 2021;203:371-4.
crossref pmid pmc
9. Gulati A, Lakhani P. Interstitial lung abnormalities and pulmonary fibrosis in COVID-19 patients: a short-term follow-up case series. Clin Imaging 2021;77:180-6.
crossref pmid pmc
10. Writing Committee for the COMEBAC Study Group, Morin L, Savale L, Pham T, Colle R, Figueiredo S, et al. Four-month clinical status of a cohort of patients after hospitalization for COVID-19. JAMA 2021;325:1525-34.
crossref pmid pmc
11. Huang W, Wu Q, Chen Z, Xiong Z, Wang K, Tian J, et al. The potential indicators for pulmonary fibrosis in survivors of severe COVID-19. J Infect 2021;82:e5-7.
crossref pmid pmc
12. Nabahati M, Ebrahimpour S, Khaleghnejad Tabari R, Mehraeen R. Post-COVID-19 pulmonary fibrosis and its predictive factors: a prospective study. Egypt J Radiol Nuclear Med 2021;52:248.
crossref pdf
13. Li X, Shen C, Wang L, Majumder S, Zhang D, Deen MJ, et al. Pulmonary fibrosis and its related factors in discharged patients with new corona virus pneumonia: a cohort study. Respir Res 2021;22:203.
crossref pmid pmc pdf
14. Yasin R, Gomaa AA, Ghazy T, Hassanein SA, Ibrahem RAL, Khalifa MH. Predicting lung fibrosis in post-COVID-19 patients after discharge with follow-up chest CT findings. Egypt J Radiol Nucl Med 2021;52:118.
crossref pdf
15. Hansell DM, Bankier AA, MacMahon H, McLoud TC, Muller NL, Remy J. Fleischner Society: glossary of terms for thoracic imaging. Radiology 2008;246:697-722.
crossref pmid
16. Zou JN, Sun L, Wang BR, Zou Y, Xu S, Ding YJ, et al. The characteristics and evolution of pulmonary fibrosis in COVID-19 patients as assessed by AI-assisted chest HRCT. PLoS One 2021;16:e0248957.
crossref pmid pmc
17. Han X, Fan Y, Alwalid O, Zhang X, Jia X, Zheng Y, et al. Fibrotic interstitial lung abnormalities at 1-year follow-up CT after severe COVID-19. Radiology 2021;301:E438-40.
crossref pmid
18. Pan F, Yang L, Liang B, Ye T, Li L, Li L, et al. Chest CT patterns from diagnosis to 1 year of follow-up in patients with COVID-19. Radiology 2022;302:709-19.
crossref pmid pmc
19. Zhang P, Li J, Liu H, Han N, Ju J, Kou Y, et al. Long-term bone and lung consequences associated with hospital-acquired severe acute respiratory syndrome: a 15-year follow-up from a prospective cohort study. Bone Res 2020;8:8.
crossref pmid pmc pdf
20. Aul DR, Gates DJ, Draper DA, Dunleavy DA, Ruickbie DS, Meredith DH, et al. Complications after discharge with COVID-19 infection and risk factors associated with development of post-COVID pulmonary fibrosis. Respir Med 2021;188:106602.
crossref pmid pmc
21. Cocconcelli E, Bernardinello N, Giraudo C, Castelli G, Giorgino A, Leoni D, et al. Characteristics and prognostic factors of pulmonary fibrosis after COVID-19 pneumonia. Front Med (Lausanne) 2021;8:823600.
crossref pmid pmc
22. Yu M, Liu Y, Xu D, Zhang R, Lan L, Xu H. Prediction of the development of pulmonary fibrosis using serial thin-section CT and clinical features in patients discharged after treatment for COVID-19 pneumonia. Korean J Radiol 2020;21:746-55.
crossref pmid pmc pdf
23. Guler SA, Ebner L, Aubry-Beigelman C, Bridevaux PO, Brutsche M, Clarenbach C, et al. Pulmonary function and radiological features 4 months after COVID-19: first results from the national prospective observational Swiss COVID-19 lung study. Eur Respir J 2021;57:2003690.
crossref pmid pmc
24. McGroder CF, Zhang D, Choudhury MA, Salvatore MM, D’Souza BM, Hoffman EA, et al. Pulmonary fibrosis 4 months after COVID-19 is associated with severity of illness and blood leucocyte telomere length. Thorax 2021;76:1242-5.
crossref pmid pmc
25. Liu W, Tao ZW, Wang L, Yuan ML, Liu K, Zhou L, et al. Analysis of factors associated with disease outcomes in hospitalized patients with 2019 novel coronavirus disease. Chin Med J (Engl) 2020;133:1032-8.
crossref pmid pmc
26. Da BL, Im GY, Schiano TD. Coronavirus disease 2019 hangover: a rising tide of alcohol use disorder and alcohol-associated liver disease. Hepatology 2020;72:1102-8.
crossref pmid pdf
27. Wilson MS, Wynn TA. Pulmonary fibrosis: pathogenesis, etiology and regulation. Mucosal Immunol 2009;2:103-21.
crossref pmid pmc pdf
28. Myers JL, Katzenstein AL. Ultrastructural evidence of alveolar epithelial injury in idiopathic bronchiolitis obliterans-organizing pneumonia. Am J Pathol 1988;132:102-9.
pmid pmc
29. Strieter RM, Mehrad B. New mechanisms of pulmonary fibrosis. Chest 2009;136:1364-70.
crossref pmid pmc
30. Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R. The COVID-19 cytokine storm: what we know so far. Front Immunol 2020;11:1446.
crossref pmid pmc
31. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395:497-506.
crossref pmid pmc
32. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 2020;130:2620-9.
crossref pmid pmc
33. Kis K, Liu X, Hagood JS. Myofibroblast differentiation and survival in fibrotic disease. Expert Rev Mol Med 2011;13:e27.
crossref pmid pmc
34. Piguet PF, Ribaux C, Karpuz V, Grau GE, Kapanci Y. Expression and localization of tumor necrosis factor-alpha and its mRNA in idiopathic pulmonary fibrosis. Am J Pathol 1993;143:651-5.
pmid pmc
35. Piguet PF, Collart MA, Grau GE, Kapanci Y, Vassalli P. Tumor necrosis factor/cachectin plays a key role in bleomycin-induced pneumopathy and fibrosis. J Exp Med 1989;170:655-63.
crossref pmid pmc pdf
36. Piguet PF, Collart MA, Grau GE, Sappino AP, Vassalli P. Requirement of tumour necrosis factor for development of silica-induced pulmonary fibrosis. Nature 1990;344:245-7.
crossref pmid pdf
37. Ruan Q, Yang K, Wang W, Jiang L, Song J. 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 pmid pmc pdf
38. Gao Y, Li T, Han M, Li X, Wu D, Xu Y, et al. Diagnostic utility of clinical laboratory data determinations for patients with the severe COVID-19. J Med Virol 2020;92:791-6.
crossref pmid pmc pdf
39. Chen L, Liu HG, Liu W, Liu J, Liu K, Shang J, et al. Analysis of clinical features of 29 patients with 2019 novel coronavirus pneumonia. Zhonghua Jie He He Hu Xi Za Zhi 2020;43:203-8.
pmid
40. Moodley YP, Misso NL, Scaffidi AK, Fogel-Petrovic M, McAnulty RJ, Laurent GJ, et al. Inverse effects of interleukin-6 on apoptosis of fibroblasts from pulmonary fibrosis and normal lungs. Am J Respir Cell Mol Biol 2003;29:490-8.
crossref pmid
41. Shieh JM, Tseng HY, Jung F, Yang SH, Lin JC. Elevation of IL-6 and IL-33 levels in serum associated with lung fibrosis and skeletal muscle wasting in a bleomycin-induced lung injury mouse model. Mediators Inflamm 2019;2019:7947596.
crossref pmid pmc pdf
42. Le TT, Karmouty-Quintana H, Melicoff E, Le TT, Weng T, Chen NY, et al. Blockade of IL-6 Trans signaling attenuates pulmonary fibrosis. J Immunol 2014;193:3755-68.
crossref pmid pmc
43. Nathani N, Perkins GD, Tunnicliffe W, Murphy N, Manji M, Thickett DR. Kerbs von Lungren 6 antigen is a marker of alveolar inflammation but not of infection in patients with acute respiratory distress syndrome. Crit Care 2008;12:R12.
crossref pmid pmc
44. Zhang H, Chen L, Wu L, Huang J, Li H, Wang X, et al. Diagnostic and prognostic predictive values of circulating KL-6 for interstitial lung disease: a PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore) 2020;99:e19493.
pmid pmc
45. Peng DH, Luo Y, Huang LJ, Liao FL, Liu YY, Tang P, et al. Correlation of Krebs von den Lungen-6 and fibronectin with pulmonary fibrosis in coronavirus disease 2019. Clin Chim Acta 2021;517:48-53.
crossref pmid pmc
46. Beyerstedt S, Casaro EB, Rangel EB. COVID-19: angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. Eur J Clin Microbiol Infect Dis 2021;40:905-19.
crossref pmid pmc pdf
47. Peiro C, Moncada S. Substituting angiotensin-(1-7) to prevent lung damage in SARS-CoV-2 infection? Circulation 2020;141:1665-6.
crossref pmid pmc
48. Xu J, Xu X, Jiang L, Dua K, Hansbro PM, Liu G. SARS-CoV-2 induces transcriptional signatures in human lung epithelial cells that promote lung fibrosis. Respir Res 2020;21:182.
crossref pmid pmc pdf
49. Yue X, Shan B, Lasky JA. TGF-beta: titan of lung fibrogenesis. Curr Enzym Inhib 2010;6:10.2174/10067.
pmc
50. Fernandez IE, Eickelberg O. The impact of TGF-beta on lung fibrosis: from targeting to biomarkers. Proc Am Thorac Soc 2012;9:111-6.
crossref pmid
51. Farghaly S, Badedi M, Ibrahim R, Sadhan MH, Alamoudi A, Alnami A, et al. Clinical characteristics and outcomes of post-COVID-19 pulmonary fibrosis: a case-control study. Medicine (Baltimore) 2022;101:e28639.
pmid pmc
52. Turrens JF. Mitochondrial formation of reactive oxygen species. J Physiol 2003;552:335-44.
crossref pmid pmc
53. Das KC. Hyperoxia decreases glycolytic capacity, glycolytic reserve and oxidative phosphorylation in MLE-12 cells and inhibits complex I and II function, but not complex IV in isolated mouse lung mitochondria. PLoS One 2013;8:e73358.
crossref pmid pmc
54. Bueno M, Calyeca J, Rojas M, Mora AL. Mitochondria dysfunction and metabolic reprogramming as drivers of idiopathic pulmonary fibrosis. Redox Biol 2020;33:101509.
crossref pmid pmc
55. Bilgili H, Bialas AJ, Gorski P, Piotrowski WJ. Telomere abnormalities in the pathobiology of idiopathic pulmonary fibrosis. J Clin Med 2019;8:1232.
crossref pmid pmc
56. Sonnweber T, Sahanic S, Pizzini A, Luger A, Schwabl C, Sonnweber B, et al. Cardiopulmonary recovery after COVID-19: an observational prospective multicentre trial. Eur Respir J 2021;57:2003481.
crossref pmid pmc
57. Huang L, Yao Q, Gu X, Wang Q, Ren L, Wang Y, et al. 1-year outcomes in hospital survivors with COVID-19: a longitudinal cohort study. Lancet 2021;398:747-58.
crossref pmid pmc
58. Liang L, Yang B, Jiang N, Fu W, He X, Zhou Y, et al. Three-month follow-up study of survivors of coronavirus disease 2019 after discharge. J Korean Med Sci 2020;35:e418.
crossref pmid pmc pdf
59. Huang Y, Tan C, Wu J, Chen M, Wang Z, Luo L, et al. Impact of coronavirus disease 2019 on pulmonary function in early convalescence phase. Respir Res 2020;21:163.
crossref pmid pmc pdf
60. Marvisi M, Ferrozzi F, Balzarini L, Mancini C, Ramponi S, Uccelli M. First report on clinical and radiological features of COVID-19 pneumonitis in a Caucasian population: factors predicting fibrotic evolution. Int J Infect Dis 2020;99:485-8.
crossref pmid pmc
61. King TE Jr, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med 2014;370:2083-92.
crossref pmid
62. Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014;370:2071-82.
crossref pmid
63. Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 2015;45:1434-45.
crossref pmid pmc
64. Umemura Y, Mitsuyama Y, Minami K, Nishida T, Watanabe A, Okada N, et al. Efficacy and safety of nintedanib for pulmonary fibrosis in severe pneumonia induced by COVID-19: an interventional study. Int J Infect Dis 2021;108:454-60.
crossref pmid pmc
65. Ogata H, Nakagawa T, Sakoda S, Ishimatsu A, Taguchi K, Kadowaki M, et al. Nintedanib treatment for pulmonary fibrosis after coronavirus disease 2019. Respirol Case Rep 2021;9:e00744.
crossref pmid pmc pdf
66. Marwah V, Choudhary R, Malik V, Pemmaraju A, Peter D. Early experience of nintedanib in COVID-19 ARDS-related pulmonary fibrosis: a case series. Adv Respir Med 2021;89:589-96.
crossref pmid
67. Ruwanpura SM, Thomas BJ, Bardin PG. Pirfenidone: molecular mechanisms and potential clinical applications in lung disease. Am J Respir Cell Mol Biol 2020;62:413-22.
crossref pmid
68. Zhang F, Wei Y, He L, Zhang H, Hu Q, Yue H, et al. A trial of pirfenidone in hospitalized adult patients with severe coronavirus disease 2019. Chin Med J (Engl) 2021;135:368-70.
crossref pmid pmc
69. Bazdyrev E, Rusina P, Panova M, Novikov F, Grishagin I, Nebolsin V. Lung fibrosis after COVID-19: treatment prospects. Pharmaceuticals (Basel) 2021;14:807.
crossref pmid pmc
70. Recovery Collaborative Group; Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, et al. Dexamethasone in hospitalized patients with COVID-19. N Engl J Med 2021;384:693-704.
crossref pmid pmc
71. Gupta S, Padappayil RP, Bansal A, Daouk S, Brown B. Tocilizumab in patients hospitalized with COVID-19 pneumonia: systematic review and meta-analysis of randomized controlled trials. J Investig Med 2022;70:55-60.
crossref pmid pmc
72. Myall KJ, Mukherjee B, Castanheira AM, Lam JL, Benedetti G, Mak SM, et al. Persistent post-COVID-19 interstitial lung disease: an observational study of corticosteroid treatment. Ann Am Thorac Soc 2021;18:799-806.
crossref pmid pmc
73. Dowman L, Hill CJ, Holland AE. Pulmonary rehabilitation for interstitial lung disease. Cochrane Database Syst Rev 2014;6:CD006322.
crossref
74. Liu K, Zhang W, Yang Y, Zhang J, Li Y, Chen Y. Respiratory rehabilitation in elderly patients with COVID-19: a randomized controlled study. Complement Ther Clin Pract 2020;39:101166.
crossref pmid pmc
75. Goodwin VA, Allan L, Bethel A, Cowley A, Cross JL, Day J, et al. Rehabilitation to enable recovery from COVID-19: a rapid systematic review. Physiotherapy 2021;111:4-22.
crossref pmid pmc
76. Chen JY, Qiao K, Liu F, Wu B, Xu X, Jiao GQ, et al. Lung transplantation as therapeutic option in acute respiratory distress syndrome for coronavirus disease 2019-related pulmonary fibrosis. Chin Med J (Engl) 2020;133:1390-6.
crossref pmid pmc
77. Bharat A, Querrey M, Markov NS, Kim S, Kurihara C, Garza-Castillon R, et al. Lung transplantation for patients with severe COVID-19. Sci Transl Med 2020;12:eabe4282.
crossref pmid pmc
78. Nintedanib for the treatment of SARS-Cov-2 induced pulmonary fibrosis [Internet]. Bethesda: U.S. National Library of Medicine; 2020 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04541680.

79. Pirfenidone vs. nintedanib for fibrotic Llung disease after coronavirus disease-19 pneumonia (PINCER) [Internet]. Bethesda: U.S. National Library of Medicine; 2021 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04856111.

80. The study of the use of nintedanib in slowing lung disease in patients with fibrotic or non-fibrotic interstitial lung disease related to COVID-19 (ENDCOV-I) [Internet]. Bethesda: National Library of Medicine; 2020 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04619680.

81. Pirfenidone compared to placebo in post-COVID19 pulmonary fibrosis COVID-19 (FIBRO-COVID) [Internet]. Bethesda: National Library of Medicine; 2020 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04607928.

82. A study to evaluate the efficacy and safety of pirfenidone with novel coronavirus infection [Internet]. Bethesda: National Library of Medicine; 2020 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04282902.

83. Pilot study to assess efficacy and safety of treamid in the rehabilitation of patients after COVID-19 pneumonia [Internet]. Bethesda: U.S. National Library of Medicine; 2020 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04527354.

84. Colchicine and post-COVID-19 pulmonary fibrosis [Internet]. Bethesda: U.S. National Library of Medicine; 2021 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04818489.

85. Safety and effectiveness of EV-Pure + WJ-Pure treatment on pulmonary fibrosis secondary to COVID-19 [Internet]. Bethesda: U.S. National Library of Medicine; 2022 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT05387239.

86. Assessing the efficacy of sirolimus in patients with COVID-19 pneumonia for prevention of post-COVID fibrosis [Internet]. Bethesda: U.S. National Library of Medicine; 2021 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04948203.

87. Mineralocorticoid receptor antagonist and pulmonary fibrosis in COVID-19 [Internet]. Bethesda: U.S. National Library of Medicine; 2021 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04912011.

88. The MONACO cell therapy study: monocytes as an anti-fibrotic treatment after COVID-19 (MONACO) [Internet]. Bethesda: U.S. National Library of Medicine; 2021 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04805086.

89. LYT-100 in post-acute COVID-19 respiratory disease [Internet]. Bethesda: U.S. National Library of Medicine; 2020 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04652518.

90. Intramuscular effect of polymerized type I collagen on the cytokine storm in COVID-19 patients [Internet]. Bethesda: U.S. National Library of Medicine; 2020 [cited 2022 Apr 20]. Available from: https://ClinicalTrials.gov/show/NCT04517162.



ABOUT
ARTICLE & TOPICS
Article category

Browse all articles >

Topics

Browse all articles >

BROWSE ARTICLES
FOR CONTRIBUTORS
Editorial Office
101-605, 58, Banpo-daero, Seocho-gu (Seocho-dong, Seocho Art-Xi), Seoul 06652, Korea
Tel: +82-2-575-3825, +82-2-576-5347    Fax: +82-2-572-6683    E-mail: katrdsubmit@lungkorea.org                

Copyright © 2024 by The Korean Academy of Tuberculosis and Respiratory Diseases. All rights reserved.

Developed in M2PI

Close layer
prev next