Next Article in Journal
Cardiac Arrest Survival Postresuscitation In-Hospital (CASPRI) Score Predicts Neurological Favorable Survival in Emergency Department Cardiac Arrest
Previous Article in Journal
Updates on Molecular and Biochemical Development and Progression of Prostate Cancer
Previous Article in Special Issue
Infection of Human Cells by SARS-CoV-2 and Molecular Overview of Gastrointestinal, Neurological, and Hepatic Problems in COVID-19 Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunologic Dysregulation and Hypercoagulability as a Pathophysiologic Background in COVID-19 Infection and the Immunomodulating Role of Colchicine

by
Dimitrios A. Vrachatis
1,†,
Konstantinos A. Papathanasiou
1,†,
Sotiria G. Giotaki
1,
Konstantinos Raisakis
2,
Charalampos Kossyvakis
2,
Andreas Kaoukis
2,
Fotis Kolokathis
1,
Gerasimos Deftereos
2,
Konstantinos E. Iliodromitis
3,4,
Dimitrios Avramides
2,
Harilaos Bogossian
3,4,
Gerasimos Siasos
1,
George Giannopoulos
5,
Bernhard Reimers
6,
Alexandra Lansky
7,
Jean-Claude Tardif
8 and
Spyridon Deftereos
1,*
1
Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece
3
Evangelisches Krankenhaus Hagen-Haspe, Clinic for Cardiology and Electrophysiology, 58135 Hagen, Germany
4
Department of Cardiology, University of Witten/Herdecke, 58455 Witten, Germany
5
Medical School, Artistotle University of Thessaloniki, 54124 Thessaloniki, Greece
6
Humanitas Research Hospital IRCCS, Rozzano, 20089 Milan, Italy
7
Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
8
Montreal Heart Institute, Université de Montréal, Montreal, QC H1T 1C8, Canada
*
Author to whom correspondence should be addressed.
Equal contribution.
J. Clin. Med. 2021, 10(21), 5128; https://doi.org/10.3390/jcm10215128
Submission received: 20 September 2021 / Revised: 20 October 2021 / Accepted: 29 October 2021 / Published: 31 October 2021

Abstract

:
In 2020, SARS-COV-2 put health systems under unprecedented resource and manpower pressure leading to significant number of deaths. Expectedly, researchers sought to shed light on the pathophysiologic background of this novel disease (COVID-19) as well as to facilitate the design of effective therapeutic modalities. Indeed, early enough the pivotal role of inflammatory and thrombotic pathways in SARS-COV-2 infection has been illustrated. The purpose of this article is to briefly present the epidemiologic and clinical features of COVID-19, analyze the pathophysiologic importance of immunologic dysregulation and hypercoagulability in developing disease complications and finally to present an up-to-date systematic review of colchicine’s immunomodulating capacity in view of hindering coronavirus complications.

1. Introduction: Coronavirus Disease 19 (COVID-19) Pandemic

On December 2019 Wuhan, China came to public attention due to emerging cases of fever of unknown origin and on 7 January 2020, WHO revealed the isolation of a new coronavirus strain (SARS-COV-2). At the end of the same month, China declared 7000 cases of COVID-19 infection and the virus had already spread to eighteen countries worldwide, hence establishing a new pandemic [1]. By early September 2021, more than 200 million people have been infected and the death toll has already surpassed 4 million [2].

2. COVID-19 Clinical and Pathophysiological Aspects

2.1. COVID-19 Infection: Clinical Course

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-COV-2) belongs to the coronaviridae family of viruses. All four subfamilies (α,β,γ,δ) of coronaviridae viruses affect humans or animals with respiratory tract, gastrointestinal tract and central nervous system infections [1]. Humans are susceptible only to subfamilies alpha and beta, with SARS-CoV and Middle-East Respiratory Syndrome (MERS)-CoV being examples of beta coronaviruses. SARS-COV-2 belongs to the beta subfamily and is a single RNA virus with an external envelope [1]. Both viral and host genetic variations are critical in understanding transmissibility, pathogenicity and severity of COVID-19 infection [3,4]. Taking advantage of the molecular mimicry between glycoprotein S [5,6] and angiotensin converting enzyme-2 (ACE-2) membrane receptor, SARS-COV-2 invades host cells and affects many organs (heart, endothelium, liver, lungs, gastrointestinal tract, kidney, bladder) [7]. Contaminated droplets represent the mainstay of viral transmission between humans [8]. The viral inoculation varies with a mean time to symptom development of five days. Comorbidities, obesity and age are implicated in disease severity, complication development and bacterial superinfection [9,10]. Approximately 80% of infected patients develop mild flu-like or diarrhea manifestations, 15% present with pneumonia and hypoxemia, and the remaining 5% suffer multi-organ failure with high mortality (up to 50%) [10]. The imaging hallmark is widespread and peripherally located ground glass opacities in the lungs, detected in 75% of cases on computed tomography imaging [1]. The second immunologic phase of COVID-19 infection commonly occurs during the second week after symptom onset, and patients may suffer a wide variety of complications such as acute respiratory distress syndrome, acute kidney injury, myocardial injury, sepsis, rhabdomyolysis, liver failure and venous thromboembolic disease [11]. Lymphocytopenia, prothrombin time prolongation and elevated LDH are typical laboratory alterations. Marked cytokine release (IL-1B, IL-1RA, IL-6, IL-7, IL-8, IL-2R, TNF-a) is also detected in serum [12,13]. The diagnosis is confirmed by molecular testing of nasopharyngeal swab or sputum for SARS-COV-2 RNA. During the first year of the pandemic (2020), preventive measures such isolation of infected cases, social distancing and hand hygiene were adopted. Vaccines became publicly available late in 2020 and additional efforts are underway [14].
The main therapeutic modalities tested against SARS-COV-2 fall in the following categories (Table 1): (1) active or passive immunization via vaccination and convalescent plasma, or immunoglobulins, respectively; (2) antiviral drugs (fusion inhibitors, protease inhibitors, RNA polymerase inhibitors, endosomal acidification inhibitors); (3) symptom-based device therapies such as ECMO (extracorporeal membrane oxygenation), ALS (artificial liver system), cytokine filters; (4) immunomodulation: colchicine, azithromycin, corticosteroids, interferons a or b, thalidomide, JAK inhibitors, IL-1 or IL-6 inhibitors, BTK inhibitors and cell therapies [15,16,17,18,19].

2.2. COVID-19 Pathophysiology: Immunologic Dysregulation and Hypercoagulability

The infection has been suggested to have an initial flu-like phase followed by a second immunologic stage [13]. The underlying pathophysiologic component of the latter features a marked cytokine release, thus researchers named it cytokine storm or cytokine release syndrome (alternatively known as macrophage activation syndrome or secondary hemophagocytic lymphohistiocytosis) [20]. This immunologic dysregulation differentiates SARS-COV-2 from previous coronaviruses and influenza, and is also critically involved in complication development and increased mortality [20]. Viral infections, in general, trigger an immunologic reaction that culminates in inflammasome activation and IL1-b and IL-18 production [21,22]. SARS-COV-2 leads to hyperactivation of innate and adaptive immune reactions [19]. After invading lung alveolar cells, the virus takes advantage of the host enzymatic machinery to replicate and ultimately brings about cell lysis and neighbouring cell invasion. Antigen-presenting cells (dendritic cells and macrophages) activate CD4+ lymphocytes, which in turn mobilize CD8+ lymphocytes and then neutrophils and macrophages. CD8+ lymphocytes destroy infected alveolar cells, while neutrophil and macrophage activation leads to the systematic inflammatory reaction syndrome (SIRs) and cytokine release syndrome [23]. Complement component C5a, acting as chemoattractant of neutrophils, potentiates the development of a deleterious hypercytokinaemia [24]. T-helper 17 polarization, occurring after neutrophil hyperactivation, seems to hold a central role in triggering a vicious cycle of inflammation and fibrosis [25]. The prevailing mechanisms employed to explain the immunologic dysregulation apparent in COVID-19 infection are (a) immune system escape [26]; (b) reduced INF-γ release [27,28]; (c) NETosis: a complex apoptotic mechanism which leads to extracellular neutrophilic traps [29,30]; (d) pyroptosis: another apoptotic pattern induced by rapid viral replication and cytokine overexcretion [31]. The final immunologic outcome is a state of reduced lymphocyte activity counterbalanced by macrophage hyperactivation. [20]. Severe cases feature a hypercoagulable state that resembles the prothrombotic manifestations of disseminated intravascular coagulation [32].
At least four contributory mechanisms to hypercoagulability are under investigation (Figure 1): autoimmunity and anti-phospholipid antibodies [33,34]; cytokine storm-mediated thrombin overproduction [35]; increased tissue factor and plasminogen activator inhibitor-1 levels due to ARDS-related tissue damage [36]; and ACE2 receptor-mediated endothelial dysfunction and “endothelitis” [37]. The endothelium is central to coagulation physiology, and its dysfunction explains at least partly the clinical complications (venous thromboembolic disease) and the laboratory hallmarks (elevated d-dimer, prothrombin time prolongation) of severe COVID-19 infection cases [12,32]. Of note, inflammation and coagulation share molecular pathways involving cytokines such as IL-8, IL-6, IL-1b [20] and complement components C3 and C5 [24,38]. Immune complexes containing antibodies against SARS-COV2 spike protein are also implicated in COVID-19 hypercoagulability through enhanced of FcγRIIA signaling [39,40]. Besides an hypercoagulative state, the COVID-19-related hyperinflammatory status may culminate in lung pneumonitis and/or fibrosis and/or vasculitis-like coronary and skin lesions [41].

3. Colchicine and COVID-19 Infection

3.1. Brief History, Pharmacology and COVID-19 Hypothesis

According to German archeologist Ebers, the first clinical application of colchicine dates back to 16 century BC, when Egyptians used the herb Colchicum autumnale for the treatment of swelling [42].
In 1987, Rodriguez de la Serna et al. rekindled physicians interest in studying colchicine, since they reported its beneficial role in managing recurrent pericarditis [43]. Katsilambros examined the antiviral properties of colchicine in a variety of infections (herpes zoster virus, mumps, echovirus and coxsackievirus among others) since 1955 [44], yet a recent systematic review suggested that the clinical value of using colchicine for infectious disease management is still inconclusive [45].
Through the centuries colchicine was proven effective in alleviating many inflammatory diseases such as gout, pericarditis, familial Mediterranean fever, Adamantiades-Behcet’s disease, post-operative pericarditis and a variety of dermatologic conditions [14,46,47,48,49]. SARS-CoV-2-induced hypercytokinaemia might also be positively affected by colchicine ’s anti-inflammatory properties [50].
Colchicine is a lipophilic alkaloid and it is metabolized in the liver and primarily excreted via the gastrointestinal tract (80%) [51]. To minimize side effects and drug-drug interactions, physicians should meticulously examine co-administration of medications that are substrates of either CYP3A4 enzyme or P-glycoprotein transporter. Colchicine is neither an inhibitor nor an inducer of these two metabolic pathways, yet substances strongly affecting those two enzymes might increase colchicine plasma concentration to toxic levels [48]. Diarrhea, nausea and vomiting are common side effects after colchicine administration, but most cases are mild and manageable, since it is a dose-related phenomenon. Infrequently, transient myelosuppression or myopathy may occur in the setting of severe kidney or severe liver disease, particularly if the contra-indication related to the use of concomitant medications like clarithromycin is not respected (azithromycin can be used) [52]. Two recent meta-analyses are reassuring and suggest the relative safety of colchicine administration [53,54]. In addition, the two large COLCOT and LoDoCo2 trials, which included a total of more than 10,000 patients with coronary artery disease followed for approximately 2 years, have also shown the safety of low-dose colchicine [55,56].
Colchicine seems to possess pleiotropic mechanisms of action [42,51,52,57,58,59] such as: (1) inhibition of neutrophil chemotaxis, extravasation and degranulation via tubulin depolymerization, (2) reduced IL-1β and IL-18 secretion through NLRP3 (NOD-, LRR- and pyrin domain-containing protein) inflammasome inhibition in neutrophils and macrophages, (3) inhibition of platelet-neutrophil and platelet-monocyte aggregates (notably, platelet to platelet interactions are spared and normal coagulation mechanisms remain intact), (4) maturation of dendritic antigen-presenting cells, (5) anti-fibrotic effects (colchicine induces Bcl-2 gene expression and down-regulates ROCK pathway as well as transforming growth factor-β1), (6) endothelial protection via reduced Lp-PLA2 activity and enhanced nitric oxide levels [60], (7) T-cell inhibition via downregulation of L-selectin [61], (8) anti-vascular endothelial growth factor effect [62,63], (9) calcium homeostasis regulation in cardiomyocytes [64], and (10) suppression of NETs [65,66]. Regarding COVID-19 infection, inhibition of clathrin-mediated endocytocis (at least partly regulated via tubulin polymerization) may be involved in its potentially favorable effects [67,68]. Furthermore, the protease furin belonging to the PCSK (pro-protein convertase subtilisin/kexin) family activates the spike protein of SARS-COV-2 and is implicated in endothelial tropism of the virus [69]. Interestingly, colchicine has been suspected to inhibit the enzymatic activity of furin [69]. Aiming to repurpose existing drugs against SARS-COV-2, Peele et al. carried out an extensive molecular dynamics analysis and found that colchicine is a potent inhibitor of viral assembly by exerting high binding affinity with its membrane (M) protein [70]. The role of tissue factor overexpression is increasingly recognized in COVID-19-related coagulopathy, and the former might be affected by colchicine [71]. Lastly, colchicine reduced acute lung injury in a rat model of acute respiratory distress syndrome [72].

3.2. Clinical Results with Colchicine in COVID-19 Patients

The aforementioned multifaceted actions of colchicine, affecting critical stages of inflammation (innate, adaptive immunity, angiogenesis, fibrosis) as well as cardiomyocyte and endothelial functions, encouraged many investigators to put colchicine to the test against COVID-19 infection (see also Figure 2).
A systematic PubMed search was carried out (up to 2 September 2021) using the following query ‘’colchicine’’ AND ‘’COVID’’ OR ‘’COVID-19’’ OR ‘’SARS-COV 2’’ OR ‘’betacoronavirus’’. References cited in the articles initially identified by this query were reviewed in order to identify any supplemental studies (“snowball procedure”). Out of 172 results, 27 studies reported outcomes regarding colchicine utilization against SARS-COV-2 and are discussed in the following sections (Figure 3).

3.2.1. Studies Evaluating Colchicine as Prevention against COVID-19

As could be predicted by known pathophysiology and its properties, studies that evaluated colchicine against the risk of infection with SARS-COV-2 showed no protective effect. Bourguiba et al. conducted a retrospective survey of FMF (Familial Mediterranean Fever) patients on chronic colchicine treatment (1 mg daily) in France from March to May 2020. They identified 27 infected patients and found neither increased risk for severe infection due to FMF history nor protection after chronic colchicine administration [73]. The small number of patients and the observational study design with no controls are the major limitations of this study. Similarly, Güven et al. retrospectively examined data from 34 FMF patients diagnosed with SARS-COV-2 between March and December 2020 in Turkey and their findings were in accordance with the above-mentioned study [74]. In addition, Gendelman et al. retrospectively investigated 14,520 subjects from the Maccabi Health Services database in Israel (from 23 February 2020 to 31 March 2020) and found no difference in the rate of COVID-19 infection between colchicine users and non-users [75].
The protective role of colchicine against COVID-19-related hospital admissions was also investigated in a retrospective observational study in Spain from March 2020 to May 2020. After evaluating 9379 rheumatologic patients on colchicine (71.6% female, median age 61.1 years, 8% diabetes), Madrid-García et al. found no statistically significant change in hospitalization rates (2.96% in colchicine users vs. 1.34% in controls) [76].
A Turkish cohort of 404 pediatric patients suffering from chronic inflammatory disease was investigated in a retrospective manner between 11 March and 15 May 2020. 90% of the study population had a diagnosis of FMF and 93% were on colchicine. Among 376 colchicine users, 6 patients contracted COVID-19 infection and only one of them required hospitalization [77]. The retrospective design, short follow-up and lack of control group are the major limitations of this analysis. A cross-sectional study conducted from June 2020 to February 2021 evaluated the features of 822 patients with FMF and suggested that colchicine non-compliance was not associated with COVID-19 hospitalization. Of note, this Turkish cohort investigated a relatively young population without major comorbidities (mean age 35.6 years, female gender 62.9%, cardiovascular disease 0.2%, and diabetes mellitus 1.2%) [78].
Finally, a small (n = 36) retrospective case-control study from Jerusalem revealed that neither FMF history nor colchicine administration at baseline affected COVID-19-related length of hospitalization, mortality, oxygen support needs and mechanical ventilation [79].

3.2.2. Colchicine for the Treatment or Prevention of COVID-19 Complications

Early in the COVID-19 pandemic, colchicine has been proposed as a potential means to alleviate the cytokine storm and hence prevent COVID-19 complications [80,81]. Up to April 2020, only case reports and small case series had been published providing a signal of benefit [82,83,84,85,86]. In the open-label, randomized GRECCO-19 study, Deftereos et al. observed longer time to clinical deterioration of COVID-19 with colchicine compared to the control group [87]. In particular, only 1.8% of patients taking colchicine suffered clinical deterioration as compared to 14.0% of the patients on conventional treatment. Interestingly, the secondary endpoint of peak d-dimer concentration was reduced by colchicine [88]. This observation should be further investigated in view of the hypercoagulable state associated with COVID-19 infection and the prognostic value of d-dimer level in this setting [89].
The results of the double-blind, randomized controlled COLCORONA trial then became available. COLCORONA focused on 4488 community-treated patients who were randomly assigned to receive colchicine (0.5 mg twice daily for 3 days and once daily for the following 27 days) or placebo for 28 days [90]. The effect of colchicine compared to placebo on the primary endpoint (death or hospitalization) was not statistically significant for the entire population (odds ratio 0.79, 95% CI 0.61–1.03, p = 0.081). However, in a predefined analysis of patients with PCR-proven diagnosis (4159 patients), the benefit of colchicine on the primary endpoint was statistically significant (4.6% vs. 6.0%, respectively; odds ratio 0.75, 95% CI 0.57–0.99, p = 0.042). This effect was primarily driven by hospitalizations rather than deaths. Although not a pre-specified outcome, all-cause hospitalizations were lowered by colchicine administration and reached statistical significance (4.9% vs. 6.3%; p = 0.04). The number needed to treat with colchicine in order to prevent a death or hospital admission was 25 and 29 in those with cardiovascular disease and diabetes mellitus, respectively.
Recovery is an ongoing trial that investigated, among its various arms, colchicine versus standard of care therapy (94% of patients were on systemic corticosteroid treatment) and found no additional benefit on 28-day mortality (risk ratio 1.02; 95% CI 0.94–1.11; p = 0.63). Of note, a high mortality rate was observed in both treatment arms (21%), suggesting that this hospitalized population suffered from severe COVID-19 [91]. One study limitation is that colchicine and systemic steroids are rarely used clinically in combination for any inflammatory disease.
The ECLA PHRI COLCOVID results were announced at the European Society of Cardiology Virtual Congress in August 2021 [92]. This open-label study randomized 1277 hospitalized adults (mean age 62 years, 64% male) to standard care with or without colchicine (loading dose of 2 mg on day 1 and 0.5 mg twice daily for 2 weeks). At 28 days of follow-up, the effect of colchicine on the co-primary endpoint of death or mechanical ventilation did not reach statistical significance (HR: 0.83; 95% CI: 0.67–1.02; p = 0.08). Of note, the occurrence of the composite endpoint of new intubation or death due to respiratory failure was reduced in the colchicine group compared to the control group (HR 0.79%; 95% CI 0.63–0.99; p < 0.05) [92]. These results are concordant with those of other studies in hospitalized patients with SARS-COV-2 that have shown benefits of colchicine on ordinal scale clinical deterioration [92] and need for supplemental oxygen [92] (see Table 2).
Finally, Karatza et al. conducted a pharmacokinetic simulation analysis showing that 0.5 mg of colchicine twice daily achieves therapeutic plasma levels with minimal toxicity [101].

3.2.3. Meta-Analyses

A series of six studies of patients hospitalized for COVID-19 were meta-analyzed by Vrachatis et al. and a mortality risk reduction was found in those treated with colchicine (OR 0.35; 95% CI 0.24–0.52) [102]. Updated meta-analyses confirmed these findings. Salah et al. conducted a meta-analysis of 8 studies (5259 patients) and found that colchicine reduced mortality (3.2% vs. 8.3%; RR: 0.62; 95% CI: 0.48–0.81) [103]. Similar results were reported by Hariyanto et al. [104]. Chiu et al. included six studies (3 observational studies and 3 randomized trials) having included 5033 patients and reported mortality risk reduction with colchicine administration (OR 0.36; 95% CI: 0.17–0.76), but this survival benefit was no longer significant when analyzing only randomized trial data (OR 0.49; 95% CI: 0.20–1.24) [105]. More recent meta-analyses of COVID-19 studies have also reported benefits of colchicine administration on mortality [106,107,108,109]. Of note, Nawangsih et al. demonstrated that colchicine’s survival benefit is more pronounced in younger adults yet, remains statistically important irrespective of diabetes and coronary artery disease history [108]. The incorporation of RECOVERY trial findings in a meta-analysis conducted by Lien et al. did not alter the afore-mentioned survival benefit among colchicine treated patients [109] (see also Table 3).
Recently, the Italian Society of Anti-Infective Therapy and the Italian Society of Pulmonology guidelines highlighted that colchicine may be considered in outpatients with COVID-19, if other randomized trials replicate COLCORONA’s favorable findings [110].
Finally, a limitation of our study is that we opted for data published in MEDLINE instead of unpublished papers or studies published in other databases. Another limitation is the fact that the presented data predate the new era of vaccination against SARS-COV2.

4. Conclusions

SARS-COV-2 not infrequently triggers a massive release of cytokines, capable of causing severe lung injury and multi-organ dysfunction and establishing a detrimental prothrombotic state. Colchicine is an old, safe, inexpensive but powerful anti-inflammatory medication with favorable pleiotropic and cardioprotective effects. Currently available data have shown colchicine’s potential in SARS-COV2 infected patients. Further research will clarify its role in the ongoing COVID-19 pandemic.

Author Contributions

Conceptualization, D.A.V., K.A.P., S.G.G., G.G. and S.D.; methodology, D.A.V., K.A.P., S.G.G., K.R., C.K., A.K. and F.K.; software D.A.V., K.A.P., S.G.G., G.D.; validation not applicable; formal analysis not applicable; investigation not applicable; resources not applicable; data curation D.A.V., K.A.P., S.G.G., K.R., C.K., A.K. and G.D.; writing—original draft preparation, D.A.V, K.A.P., S.G.G., K.R, C.K., A.K, F.K.; writing—review and editing, K.E.I., D.A., H.B., G.S., G.G., B.R., A.L., J.-C.T., S.D.; visualization, K.A.P., S.G.G. and G.D.; supervision, G.S., G.G., and S.D.; project administration, D.A.V., K.A.P.,S.G.G.,G.D. and S.D.; funding acquisition, not applicable. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available on request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harapan, H.; Itoh, N.; Yufika, A.; Winardi, W.; Keam, S.; Te, H.; Megawati, D.; Hayati, Z.; Wagner, A.L.; Mudatsir, M. Coronavirus disease 2019 (COVID-19): A literature review. J. Infect. Public Health 2020, 13, 667–673. [Google Scholar] [CrossRef]
  2. COVID-19 Map—Johns Hopkins Coronavirus Resource Center n.d. Available online: https://coronavirus.jhu.edu/map.html (accessed on 2 September 2021).
  3. Peacock, T.P.; Penrice-Randal, R.; Hiscox, J.A.; Barclay, W.S. SARS-CoV-2 one year on: Evidence for ongoing viral adaptation. J. Gen. Virol. 2021, 102, 001584. [Google Scholar] [CrossRef]
  4. Fricke-Galindo, I.; Falfán-Valencia, R. Genetics Insight for COVID-19 Susceptibility and Severity: A Review. Front. Immunol. 2021, 12, 622176. [Google Scholar] [CrossRef] [PubMed]
  5. Beniac, D.R.; Andonov, A.; Grudeski, E.; Booth, T.F. Architecture of the SARS coronavirus prefusion spike. Nat. Struct. Mol. Biol. 2006, 13, 751–752. [Google Scholar] [CrossRef] [Green Version]
  6. Gilzad-Kohan, H.; Jamali, F. Anti-Inflammatory Properties of Drugs Used to Control COVID-19 and their Effects on the Renin-Angiotensin System and Angiotensin-Converting Enzyme-2. J. Pharm. Pharm. Sci. 2020, 23, 259–277. [Google Scholar] [CrossRef] [PubMed]
  7. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef] [Green Version]
  8. Zou, L.; Ruan, F.; Huang, M.; Liang, L.; Huang, H.; Hong, Z.; Yu, J.; Kang, M.; Song, Y.; Xia, J.; et al. SARS-CoV-2 viral load in upper respiratory specimens of infected patients. N. Engl. J. Med. 2020, 382, 1177–1179. [Google Scholar] [CrossRef] [PubMed]
  9. Korakas, E.; Ikonomidis, I.; Kousathana, F.; Balampanis, K.; Kountouri, A.; Raptis, A.; Palaiodimou, L.; Kokkinos, A.; Lambadiari, V. Obesity and COVID-19: Immune and metabolic derangement as a possible link to adverse clinical outcomes. Am. J. Physiol. Metab. 2020, 319, E105–E109. [Google Scholar] [CrossRef]
  10. Wu, Z.; McGoogan, J.M. Characteristics of and Important Lessons from the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72314 Cases from the Chinese Center for Disease Control and Prevention. JAMA 2020, 323, 1239–1242. [Google Scholar] [CrossRef] [PubMed]
  11. Berlin, D.A.; Gulick, R.M.; Martinez, F.J. Severe Covid-19. N. Engl. J. Med. 2020, 383, 2451–2460. [Google Scholar] [CrossRef]
  12. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  13. Cantini, F.; Goletti, D.; Petrone, L.; Najafi Fard, S.; Niccoli, L.; Foti, R. Immune Therapy, or Antiviral Therapy, or Both for COVID-19: A Systematic Review. Drugs 2020, 80, 1929–1946. [Google Scholar] [CrossRef] [PubMed]
  14. Kabir, M.A.; Ahmed, R.; Chowdhury, R.; Asher Iqbal, S.M.; Paulmurugan, R.; Demirci, U.; Asghar, W. Management of COVID-19: Current Status and Future Prospects. Microbes Infect. 2021, 23, 104832. [Google Scholar] [CrossRef] [PubMed]
  15. Elhusseiny, K.M.; Abd-Elhay, F.A.-E.; Kamel, M.G. Possible therapeutic agents for COVID-19: A comprehensive review. Expert Rev. Anti. Infect. Ther. 2020, 18, 1005–1020. [Google Scholar] [CrossRef]
  16. Fragkou, P.C.; Belhadi, D.; Peiffer-Smadja, N.; Moschopoulos, C.D.; Lescure, F.-X.; Janocha, H.; Karofylakis, E.; Yazdanpanah, Y.; Mentré, F.; Skevaki, C.; et al. Review of trials currently testing treatment and prevention of COVID-19. Clin. Microbiol. Infect. 2020, 26, 988–998. [Google Scholar] [CrossRef] [PubMed]
  17. Magro, G. COVID-19: Review on latest available drugs and therapies against SARS-CoV-2. Coagulation and inflammation cross-talking. Virus Res. 2020, 286, 198070. [Google Scholar] [CrossRef] [PubMed]
  18. Andreou, A.; Trantza, S.; Filippou, D.; Filippou, D.; Sipsas, N.; Tsiodras, S. COVID-19: The potential role of copper and N-acetylcysteine (NAC) in a combination of candidate antiviral treatments against SARS-CoV-2. In Vivo 2020, 34, 1567–1588. [Google Scholar] [CrossRef] [PubMed]
  19. Rizk, J.G.; Kalantar-Zadeh, K.; Mehra, M.R.; Lavie, C.J.; Rizk, Y.; Forthal, D.N. Pharmaco-Immunomodulatory Therapy in COVID-19. Drugs 2020, 80, 1267–1292. [Google Scholar] [CrossRef] [PubMed]
  20. Soy, M.; Keser, G.; Atagündüz, P.; Tabak, F.; Atagündüz, I.; Kayhan, S. Cytokine storm in COVID-19: Pathogenesis and overview of anti-inflammatory agents used in treatment. Clin. Rheumatol. 2020, 39, 2085–2094. [Google Scholar] [CrossRef] [PubMed]
  21. Schnappauf, O.; Chae, J.J.; Kastner, D.L.; Aksentijevich, I. The Pyrin Inflammasome in Health and Disease. Front. Immunol. 2019, 10, 1745. [Google Scholar] [CrossRef]
  22. Ribeiro, S.A.; Lopes, C.; Amaral, R.; Amaral, A. The therapeutic potential of colchicine in the complications of COVID19. Could the immunometabolic properties of an old and cheap drug help? Metab. Open 2020, 7, 100045. [Google Scholar] [CrossRef] [PubMed]
  23. Yuki, K.; Fujiogi, M.; Koutsogiannaki, S. COVID-19 pathophysiology: A review. Clin. Immunol. 2020, 215, 108427. [Google Scholar] [CrossRef] [PubMed]
  24. Das, A.; Rana, S. The role of human C5a as a non-genomic target in corticosteroid therapy for management of severe COVID19. Comput. Biol. Chem. 2021, 92, 107482. [Google Scholar] [CrossRef] [PubMed]
  25. Sarmiento-Monroy, J.C.; Parra-Medina, R.; Garavito, E.; Rojas-Villarraga, A. T Helper 17 Response to Severe Acute Respiratory Syndrome Coronavirus 2: A Type of Immune Response with Possible Therapeutic Implications. Viral Immunol. 2021, 34, 190–200. [Google Scholar] [CrossRef] [PubMed]
  26. Huang, C.-F.; Hsieh, S.-M.; Pan, S.-C.; Huang, Y.-S.; Chang, S.-C. Dose-Related Aberrant Inhibition of Intracellular Perforin Expression by S1 Subunit of Spike Glycoprotein That Contains Receptor-Binding Domain from SARS-CoV-2. Microorganisms 2021, 9, 1303. [Google Scholar] [CrossRef]
  27. Jenner, A.L.; Aogo, R.A.; Alfonso, S.; Crowe, V.; Deng, X.; Smith, A.P.; Morel, P.A.; Davis, C.L.; Smith, A.M.; Craig, M. COVID-19 virtual patient cohort suggests immune mechanisms driving disease outcomes. PLoS Pathog. 2021, 17, e1009753. [Google Scholar] [CrossRef] [PubMed]
  28. Severa, M.; Diotti, R.A.; Etna, M.P.; Rizzo, F.; Fiore, S.; Ricci, D.; Iannetta, M.; Sinigaglia, A.; Lodi, A.; Mancini, N.; et al. Differential plasmacytoid dendritic cell phenotype and type I Interferon response in asymptomatic and severe COVID-19 infection. PLoS Pathog. 2021, 17, e1009878. [Google Scholar] [CrossRef] [PubMed]
  29. Barnes, B.J.; Adrover, J.M.; Baxter-Stoltzfus, A.; Borczuk, A.; Cools-Lartigue, J.; Crawford, J.M.; Daßler-Plenker, J.; Guerci, P.; Huynh, C.; Knight, J.S.; et al. Targeting potential drivers of COVID-19: Neutrophil extracellular traps. J. Exp. Med. 2020, 217, e20200652. [Google Scholar] [CrossRef]
  30. Obermayer, A.; Jakob, L.-M.; Haslbauer, J.D.; Matter, M.S.; Tzankov, A.; Stoiber, W. Neutrophil Extracellular Traps in Fatal COVID-19-Associated Lung Injury. Dis. Markers 2021, 2021, 1–10. [Google Scholar] [CrossRef]
  31. Ferreira, A.C.; Soares, V.C.; de Azevedo-Quintanilha, I.G.; Dias, S.d.S.G.; Fintelman-Rodrigues, N.; Sacramento, C.Q.; Mattos, M.; de Freitas, C.S.; Temerozo, J.R.; Teixeira, L.; et al. SARS-CoV-2 engages inflammasome and pyroptosis in human primary monocytes. Cell Death Discov. 2021, 7, 43. [Google Scholar] [CrossRef]
  32. Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 844–847. [Google Scholar] [CrossRef] [Green Version]
  33. Foret, T.; Dufrost, V.; Salomon Du Mont, L.; Costa, P.; Lefevre, B.; Lacolley, P.; Regnault, V.; Zuily, S.; Wahl, D. Systematic Review of Antiphospholipid Antibodies in COVID-19 Patients: Culprits or Bystanders? Curr. Rheumatol. Rep. 2021, 23, 65. [Google Scholar] [CrossRef] [PubMed]
  34. Hollerbach, A.; Müller-Calleja, N.; Pedrosa, D.; Canisius, A.; Sprinzl, M.F.; Falter, T.; Rossmann, H.; Bodenstein, M.; Werner, C.; Sagoschen, I.; et al. Pathogenic lipid-binding antiphospholipid antibodies are associated with severity of COVID-19. J. Thromb. Haemost. 2021, 19, 2335–2347. [Google Scholar] [CrossRef] [PubMed]
  35. Spiezia, L.; Boscolo, A.; Poletto, F.; Cerruti, L.; Tiberio, I.; Campello, E.; Navalesi, P.; Simioni, P. COVID-19-Related Severe Hypercoagulability in Patients Admitted to Intensive Care Unit for Acute Respiratory Failure. Thromb. Haemost. 2020, 120, 998–1000. [Google Scholar] [CrossRef]
  36. Ozolina, A.; Sarkele, M.; Sabelnikovs, O.; Skesters, A.; Jaunalksne, I.; Serova, J.; Ievins, T.; Bjertnaes, L.J.; Vanags, I. Activation of Coagulation and Fibrinolysis in Acute Respiratory Distress Syndrome: A Prospective Pilot Study. Front. Med. 2016, 3, 64. [Google Scholar] [CrossRef]
  37. Fogarty, H.; Townsend, L.; Ni Cheallaigh, C.; Bergin, C.; Martin-Loeches, I.; Browne, P.; Bacon, C.L.; Gaule, R.; Gillett, A.; Byrne, M.; et al. COVID19 coagulopathy in Caucasian patients. Br. J. Haematol. 2020, 189, 1044–1049. [Google Scholar] [CrossRef]
  38. Risitano, A.M.; Mastellos, D.C.; Huber-Lang, M.; Yancopoulou, D.; Garlanda, C.; Ciceri, F.; Lambris, J.D. Complement as a target in COVID-19? Nat. Rev. Immunol. 2020, 20, 343–344. [Google Scholar] [CrossRef] [Green Version]
  39. Bye, A.P.; Hoepel, W.; Mitchell, J.L.; Jégouic, S.; Loureiro, S.; Sage, T.; de Taeye, S.; van Gils, M.; Kriek, N.; Cooper, N.; et al. Aberrant glycosylation of anti-SARS-CoV-2 IgG is a pro-thrombotic stimulus for platelets. bioRxiv 2021. [Google Scholar] [CrossRef]
  40. Nazy, I.; Jevtic, S.D.; Moore, J.C.; Huynh, A.; Smith, J.W.; Kelton, J.G.; Arnold, D.M. Platelet-activating immune complexes identified in critically ill COVID-19 patients suspected of heparin-induced thrombocytopenia. J. Thromb. Haemost. 2021, 19, 1342–1347. [Google Scholar] [CrossRef] [PubMed]
  41. González-Gay, M.A.; Mayo, J.; Castañeda, S.; Cifrián, J.M.; Hernández-Rodríguez, J. Tocilizumab: From the rheumatology practice to the fight against COVID-19, a virus infection with multiple faces. Expert Opin. Biol. Ther. 2020, 20, 717–723. [Google Scholar] [CrossRef] [PubMed]
  42. Slobodnick, A.; Shah, B.; Krasnokutsky, S.; Pillinger, M.H. Update on colchicine, 2017. Rheumatology 2018, 57, i4–i11. [Google Scholar] [CrossRef] [Green Version]
  43. Bayes-Genis, A.; Adler, Y.; De Luna, A.B.; Imazio, M. Colchicine in Pericarditis. Eur. Heart J. 2017, 38, 1706–1709. [Google Scholar] [CrossRef] [Green Version]
  44. Katsilabros, L. La colchicine et ses dérivés contre les viroses. Arch. Inst. Pasteur Hell. 1958, 4, 139–145. [Google Scholar]
  45. McEwan, T.; Robinson, P.C. A systematic review of the infectious complications of colchicine and the use of colchicine to treat infections. Semin. Arthritis Rheum. 2021, 51, 101–112. [Google Scholar] [CrossRef]
  46. Deftereos, S.G.; Siasos, G.; Giannopoulos, G.; Vrachatis, D.A.; Angelidis, C.; Giotaki, S.G.; Gargalianos, P.; Giamarellou, H.; Gogos, C.; Daikos, G.; et al. The Greek study in the effects of colchicine in COvid-19 complications prevention (GRECCO-19 study): Rationale and study design. Hell. J. Cardiol. 2020, 61, 42–45. [Google Scholar] [CrossRef] [PubMed]
  47. Deftereos, S.G.; Vrachatis, D.A.; Angelidis, C.; Vrettou, A.-R.; Sarri, E.K.; Giotaki, S.G.; Varytimiadi, E.; Kossyvakis, C.; Kotsia, E.; Deftereos, G.S.; et al. The Role of Colchicine in Treating Postoperative and Post-catheter Ablation Atrial Fibrillation. Clin. Ther. 2019, 41, 21–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Schenone, A.L.; Menon, V. Colchicine in Pericardial Disease: From the Underlying Biology and Clinical Benefits to the Drug-Drug Interactions in Cardiovascular Medicine. Curr. Cardiol. Rep. 2018, 20, 62. [Google Scholar] [CrossRef] [PubMed]
  49. Vrachatis, D.A.; Giannopoulos, G.; Deftereos, S.G. Colchicine: Conventional and Contemporary Indications. Curr. Pharm. Des. 2018, 24, 647. [Google Scholar] [CrossRef] [PubMed]
  50. Papadopoulos, C.; Patoulias, D.; Teperikidis, E.; Mouselimis, D.; Tsarouchas, A.; Toumpourleka, M.; Boulmpou, A.; Bakogiannis, C.; Doumas, M.; Vassilikos, V.P. Colchicine as a Potential Therapeutic Agent Against Cardiovascular Complications of COVID-19: An Exploratory Review. SN Compr. Clin. Med. 2020, 2, 1419–1429. [Google Scholar] [CrossRef] [PubMed]
  51. Angelidis, C.; Kotsialou, Z.; Kossyvakis, C.; Vrettou, A.-R.; Zacharoulis, A.; Kolokathis, F.; Kekeris, V.; Giannopoulos, G. Colchicine Pharmacokinetics and Mechanism of Action. Curr. Pharm. Des. 2018, 24, 659–663. [Google Scholar] [CrossRef] [PubMed]
  52. Leung, Y.Y.; Yao Hui, L.L.; Kraus, V.B. Colchicine-Update on mechanisms of action and therapeutic uses. Semin. Arthritis Rheum. 2015, 45, 341–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Stewart, S.; Yang, K.C.K.; Atkins, K.; Dalbeth, N.; Robinson, P.C. Adverse events during oral colchicine use: A systematic review and meta-analysis of randomised controlled trials. Arthritis Res. Ther. 2020, 22, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Andreis, A.; Imazio, M.; Avondo, S.; Casula, M.; Paneva, E.; Piroli, F.; De Ferrari, G.M. Adverse events of colchicine for cardiovascular diseases. J. Cardiovasc. Med. 2021, 22, 637–644. [Google Scholar] [CrossRef]
  55. Tardif, J.C.; Kouz, S.; Waters, D.D.; Bertrand, O.F.; Diaz, R.; Maggioni, A.P.; Pinto, F.J.; Ibrahim, R.; Gamra, H.; Kiwan, G.S.; et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N. Engl. J. Med. 2019, 381, 2497–2505. [Google Scholar] [CrossRef] [PubMed]
  56. Nidorf, S.M.; Fiolet, A.T.L.; Mosterd, A.; Eikelboom, J.W.; Schut, A.; Opstal, T.S.J.; The, S.H.K.; Xu, X.-F.; Ireland, M.A.; Lenderink, T.; et al. Colchicine in Patients with Chronic Coronary Disease. N. Engl. J. Med. 2020, 383, 1838–1847. [Google Scholar] [CrossRef] [PubMed]
  57. Shah, B.; Allen, N.; Harchandani, B.; Pillinger, M.; Katz, S.; Sedlis, S.P.; Echagarruga, C.; Samuels, S.K.; Morina, P.; Singh, P.; et al. Effect of Colchicine on Platelet-Platelet and Platelet-Leukocyte Interactions: A Pilot Study in Healthy Subjects. Inflammation 2016, 39, 182–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Deftereos, S.; Giannopoulos, G.; Papoutsidakis, N.; Panagopoulou, V.; Kossyvakis, C.; Raisakis, K.; Cleman, M.W.; Stefanadis, C. Colchicine and the Heart. J. Am. Coll. Cardiol. 2013, 62, 1817–1825. [Google Scholar] [CrossRef] [Green Version]
  59. Sandbo, N.; Ngam, C.; Torr, E.; Kregel, S.; Kach, J.; Dulin, N. Control of Myofibroblast Differentiation by Microtubule Dynamics through a Regulated Localization of mDia2. J. Biol. Chem. 2013, 288, 15466–15473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Das, U.N. Bioactive Lipids in COVID-19-Further Evidence. Arch. Med. Res. 2021, 52, 107–120. [Google Scholar] [CrossRef] [PubMed]
  61. Perico, N.; Ostermann, D.; Bontempeill, M.; Morigi, M.; Amuchastegui, C.S.; Zoja, C.; Akalin, E.; Sayegh, M.H.; Remuzzi, G. Colchicine interferes with L-selectin and leukocyte function-associated antigen-1 expression on human T lymphocytes and inhibits T cell activation. J. Am. Soc. Nephrol. 1996, 7, 594–601. [Google Scholar] [CrossRef] [PubMed]
  62. Atta, H.M.; El-Rehany, M.A.; Abdel Raheim, S.R.; Fouad, R.; Galal, A.M.F. Colchicine inhibits intimal hyperplasia and leukocyte VEGF expression in dogs. J. Surg. Res. 2008, 146, 184–189. [Google Scholar] [CrossRef]
  63. Marzo-Mas, A.; Falomir, E.; Murga, J.; Carda, M.; Marco, J.A. Effects on tubulin polymerization and down-regulation of c-Myc, hTERT and VEGF genes by colchicine haloacetyl and haloaroyl derivatives. Eur. J. Med. Chem. 2018, 150, 591–600. [Google Scholar] [CrossRef] [PubMed]
  64. Lu, Y.Y.; Chen, Y.C.; Kao, Y.H.; Lin, Y.K.; Yeh, Y.H.; Chen, S.A.; Chen, Y.J. Colchicine modulates calcium homeostasis and electrical property of HL-1 cells. J. Cell. Mol. Med. 2016, 20, 1182–1190. [Google Scholar] [CrossRef] [PubMed]
  65. Kow, C.S.; Hasan, S.S. Colchicine as an adjunct to heparin for prophylaxis of venous thromboembolism in patients with COVID-19. Rheumatol. Int. 2021, 41, 677–678. [Google Scholar] [CrossRef]
  66. Apostolidou, E.; Skendros, P.; Kambas, K.; Mitroulis, I.; Konstantinidis, T.; Chrysanthopoulou, A.; Nakos, K.; Tsironidou, V.; Koffa, M.; Boumpas, D.T.; et al. Neutrophil extracellular traps regulate IL-1β-mediated inflammation in familial Mediterranean fever. Ann. Rheum. Dis. 2016, 75, 269–277. [Google Scholar] [CrossRef]
  67. Kaksonen, M.; Roux, A. Mechanisms of clathrin-mediated endocytosis. Nat. Rev. Mol. Cell Biol. 2018, 19, 313–326. [Google Scholar] [CrossRef] [PubMed]
  68. Milewska, A.; Nowak, P.; Owczarek, K.; Szczepanski, A.; Zarebski, M.; Hoang, A.; Berniak, K.; Wojarski, J.; Zeglen, S.; Baster, Z.; et al. Entry of Human Coronavirus NL63 into the Cell. J. Virol. 2017, 92, e01933-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. AbdelMassih, A.F.; Ye, J.; Kamel, A.; Mishriky, F.; Ismail, H.-A.; Ragab, H.A.; El Qadi, L.; Malak, L.; Abdu, M.; El-Husseiny, M.; et al. A multicenter consensus: A role of furin in the endothelial tropism in obese patients with COVID-19 infection. Obes. Med. 2020, 19, 100281. [Google Scholar] [CrossRef] [PubMed]
  70. Peele, K.A.; Kumar, V.; Parate, S.; Srirama, K.; Lee, K.W.; Venkateswarulu, T.C. Insilico drug repurposing using FDA approved drugs against Membrane protein of SARS-CoV-2. J. Pharm. Sci. 2021, 110, 2346–2354. [Google Scholar] [CrossRef]
  71. Cañas, C.A.; Cañas, F.; Bautista-Vargas, M.; Bonilla-Abadía, F. Role of Tissue Factor in the Pathogenesis of COVID-19 and the Possible Ways to Inhibit It. Clin. Appl. Thromb. 2021, 27, 107602962110039. [Google Scholar] [CrossRef]
  72. Dupuis, J.; Sirois, M.G.; Rhéaume, E.; Nguyen, Q.T.; Clavet-Lanthier, M.-É.; Brand, G.; Mihalache-Avram, T.; Théberge-Julien, G.; Charpentier, D.; Rhainds, D.; et al. Colchicine reduces lung injury in experimental acute respiratory distress syndrome. PLoS ONE 2020, 15, e0242318. [Google Scholar] [CrossRef] [PubMed]
  73. Bourguiba, R.; Delplanque, M.; Vinit, C.; Ackermann, F.; Savey, L.; Grateau, G.; Hentgen, V.; Georgin-lavialle, S. Clinical course of COVID-19 in a cohort of 342 familial Mediterranean fever patients with a long-term treatment by colchicine in a French endemic area. Ann. Rheum. Dis. 2021, 80, 539–540. [Google Scholar] [CrossRef] [PubMed]
  74. Güven, S.C.; Erden, A.; Karakaş, Ö.; Armağan, B.; Usul, E.; Omma, A.; Küçükşahin, O. COVID-19 outcomes in patients with familial Mediterranean fever: A retrospective cohort study. Rheumatol. Int. 2021, 41, 715–719. [Google Scholar] [CrossRef] [PubMed]
  75. Gendelman, O.; Amital, H.; Bragazzi, N.L.; Watad, A.; Chodick, G. Continuous hydroxychloroquine or colchicine therapy does not prevent infection with SARS-CoV-2: Insights from a large healthcare database analysis. Autoimmun. Rev. 2020, 19, 102566. [Google Scholar] [CrossRef]
  76. Madrid-García, A.; Pérez, I.; Colomer, J.I.; León-Mateos, L.; Jover, J.A.; Fernández-Gutiérrez, B.; Abásolo-Alcazar, L.; Rodríguez-Rodríguez, L. Influence of colchicine prescription in COVID-19-related hospital admissions: A survival analysis. Ther. Adv. Musculoskelet. Dis. 2021, 13, 1759720X2110026. [Google Scholar] [CrossRef]
  77. Haslak, F.; Yildiz, M.; Adrovic, A.; Sahin, S.; Koker, O.; Aliyeva, A.; Barut, K.; Kasapcopur, O. Management of childhood-onset autoinflammatory diseases during the COVID-19 pandemic. Rheumatol. Int. 2020, 40, 1423–1431. [Google Scholar] [CrossRef]
  78. Günendi, Z.; Yurdakul, F.G.; Bodur, H.; Cengiz, A.K.; Uçar, Ü.; Çay, H.F.; Şen, N.; Keskin, Y.; Gürer, G.; Melikoğlu, M.A.; et al. The impact of COVID-19 on familial Mediterranean fever: A nationwide study. Rheumatol. Int. 2021, 41, 1447–1455. [Google Scholar] [CrossRef] [PubMed]
  79. Kharouf, F.; Ishay, Y.; Kenig, A.; Bitan, M.; Ben-Chetrit, E. Incidence and course of COVID-19 hospitalizations among patients with familial Mediterranean fever. Rheumatology 2021, 69, SI85–SI89. [Google Scholar] [CrossRef]
  80. Deftereos, S.; Giannopoulos, G.; Vrachatis, D.A.; Siasos, G.; Giotaki, S.G.; Cleman, M.; Dangas, G.; Stefanadis, C. Colchicine as a potent anti-inflammatory treatment in COVID-19: Can we teach an old dog new tricks? Eur. Hear. J. Cardiovasc. Pharmacother. 2020, 6, 255. [Google Scholar] [CrossRef]
  81. Giannopoulos, G.; Vrachatis, D.A.; Deftereos, S.G. Myocardial Injury in COVID-19—Can We Successfully Target Inflammation? JAMA Cardiol. 2020, 5, 1069. [Google Scholar] [CrossRef]
  82. Dabbagh, M.F.; Aurora, L.; D’Souza, P.; Weinmann, A.J.; Bhargava, P.; Basir, M.B. Cardiac Tamponade Secondary to COVID-19. JACC Case Rep. 2020, 2, 1326–1330. [Google Scholar] [CrossRef] [PubMed]
  83. Recalcati, S.; Piconi, S.; Franzetti, M.; Barbagallo, T.; Prestinari, F.; Fantini, F. Colchicin treatment of COVID-19 presenting with cutaneous rash and myopericarditis. Dermatol. Ther. 2020, 33, e13891. [Google Scholar] [CrossRef]
  84. Montealegre-Gómez, G.; Garavito, E.; Gómez-López, A.; Rojas-Villarraga, A.; Parra-Medina, R. Colchicine: A potential therapeutic tool against COVID-19. Experience of 5 patients. Reumatol. Clínica 2020, 17, 371–375. [Google Scholar] [CrossRef]
  85. Gandolfini, I.; Delsante, M.; Fiaccadori, E.; Zaza, G.; Manenti, L.; Degli Antoni, A.; Peruzzi, L.; Riella, L.V.; Cravedi, P.; Maggiore, U. COVID-19 in kidney transplant recipients. Am. J. Transplant. 2020, 20, 1941–1943. [Google Scholar] [CrossRef]
  86. Della-Torre, E.; Della-Torre, F.; Kusanovic, M.; Scotti, R.; Ramirez, G.A.; Dagna, L.; Tresoldi, M. Treating COVID-19 with colchicine in community healthcare setting. Clin. Immunol. 2020, 217, 108490. [Google Scholar] [CrossRef]
  87. Deftereos, S.G.; Giannopoulos, G.; Vrachatis, D.A.; Siasos, G.D.; Giotaki, S.G.; Gargalianos, P.; Metallidis, S.; Sianos, G.; Baltagiannis, S.; Panagopoulos, P.; et al. Effect of Colchicine vs. Standard Care on Cardiac and Inflammatory Biomarkers and Clinical Outcomes in Patients Hospitalized With Coronavirus Disease 2019. JAMA Netw. Open 2020, 3, e2013136. [Google Scholar] [CrossRef] [PubMed]
  88. Bikdeli, B.; Madhavan, M.V.; Jimenez, D.; Chuich, T.; Dreyfus, I.; Driggin, E.; Nigoghossian, C.D.; Ageno, W.; Madjid, M.; Guo, Y.; et al. COVID-19 and Thrombotic or Thromboembolic Disease: Implications for Prevention, Antithrombotic Therapy, and Follow-Up: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2020, 75, 2950–2973. [Google Scholar] [CrossRef]
  89. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; 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, 1054–1062. [Google Scholar] [CrossRef]
  90. Tardif, J.-C.; Bouabdallaoui, N.; L’Allier, P.L.; Gaudet, D.; Shah, B.; Pillinger, M.H.; Lopez-Sendon, J.; da Luz, P.; Verret, L.; Audet, S.; et al. Colchicine for community-treated patients with COVID-19 (COLCORONA): A phase 3, randomised, double-blinded, adaptive, placebo-controlled, multicentre trial. Lancet Respir. Med. 2021, 9, 924–932. [Google Scholar] [CrossRef]
  91. Horby, P.W.; Campbell, M.; Spata, E.; Emberson, J.R.; Staplin, N.; Pessoa-Amorim, G.; Peto, L.; Wiselka, M.; Wiffen, L.; Tiberi, S.; et al. Colchicine in patients admitted to hospital with COVID-19 (RECOVERY): A randomised, controlled, open-label, platform trial. medRxiv 2021. [Google Scholar] [CrossRef]
  92. Colchicine in Patients Hospitalized with COVID-19—ECLA PHRI COLCOVID n.d. Available online: https://www.acc.org/Latest-in-Cardiology/Clinical-Trials/2021/08/26/01/09/ECLA-PHRI-COLCOVID (accessed on 29 August 2021).
  93. Lopes, M.I.; Bonjorno, L.P.; Giannini, M.C.; Amaral, N.B.; Menezes, P.I.; Dib, S.M.; Gigante, S.L.; Benatti, M.N.; Rezek, U.C.; Emrich-Filho, L.L.; et al. Beneficial effects of colchicine for moderate to severe COVID-19: A randomised, double-blinded, placebo-controlled clinical trial. RMD Open 2021, 7, e001455. [Google Scholar] [CrossRef]
  94. Mareev, V.Y.; Orlova, Y.A.; Plisyk, A.G.; Pavlikova, E.P.; Akopyan, Z.A.; Matskeplishvili, S.T.; Malakhov, P.S.; Krasnova, T.N.; Seredenina, E.M.; Potapenko, A.V.; et al. Proactive anti-inflammatory therapy with colchicine in the treatment of advanced stages of new coronavirus infection. The first results of the COLORIT study. Kardiologiia 2021, 61, 15–27. [Google Scholar] [CrossRef]
  95. Brunetti, L.; Diawara, O.; Tsai, A.; Firestein, B.L.; Nahass, R.G.; Poiani, G.; Schlesinger, N. Colchicine to Weather the Cytokine Storm in Hospitalized Patients with COVID-19. J. Clin. Med. 2020, 9, 2961. [Google Scholar] [CrossRef]
  96. Scarsi, M.; Piantoni, S.; Colombo, E.; Airó, P.; Richini, D.; Miclini, M.; Bertasi, V.; Bianchi, M.; Bottone, D.; Civelli, P.; et al. Association between treatment with colchicine and improved survival in a single-centre cohort of adult hospitalised patients with COVID-19 pneumonia and acute respiratory distress syndrome. Ann. Rheum. Dis. 2020, 79, 1286–1289. [Google Scholar] [CrossRef]
  97. Sandhu, T.; Tieng, A.; Chilimuri, S.; Franchin, G. A Case Control Study to Evaluate the Impact of Colchicine on Patients Admitted to the Hospital with Moderate to Severe COVID-19 Infection. Can. J. Infect. Dis. Med. Microbiol. 2020, 2020, 1–9. [Google Scholar] [CrossRef] [PubMed]
  98. Kevorkian, J.-P.; Lopes, A.; Sène, D.; Riveline, J.-P.; Vandiedonck, C.; Féron, F.; Nassarmadji, K.; Mouly, S.; Mauvais-Jarvis, F.; Gautier, J.-F.; et al. Oral corticoid, aspirin, anticoagulant, colchicine, and furosemide to improve the outcome of hospitalized COVID-19 patients—The COCAA-COLA cohort study. J. Infect. 2021, 82, 276–316. [Google Scholar] [CrossRef]
  99. Manenti, L.; Maggiore, U.; Fiaccadori, E.; Meschi, T.; Antoni, A.D.; Nouvenne, A.; Ticinesi, A.; Cerundolo, N.; Prati, B.; Delsante, M.; et al. Reduced mortality in COVID-19 patients treated with colchicine: Results from a retrospective, observational study. PLoS ONE 2021, 16, e0248276. [Google Scholar] [CrossRef] [PubMed]
  100. García-Posada, M.; Aruachan-Vesga, S.; Mestra, D.; Humánez, K.; Serrano-Coll, H.; Cabrales, H.; Faccini, Á.; Mattar, S. Clinical outcomes of patients hospitalized for COVID-19 and evidence-based on the pharmacological management reduce mortality in a region of the Colombian Caribbean. J. Infect. Public Health 2021, 14, 696–701. [Google Scholar] [CrossRef] [PubMed]
  101. Karatza, E.; Ismailos, G.; Karalis, V. Colchicine for the treatment of COVID-19 patients: Efficacy, safety, and model informed dosage regimens. Xenobiotica 2021, 51, 643–656. [Google Scholar] [CrossRef]
  102. Vrachatis, D.A.; Giannopoulos, G.V.; Giotaki, S.G.; Raisakis, K.; Kossyvakis, C.; Iliodromitis, K.E.; Reimers, B.; Tousoulis, D.; Cleman, M.; Stefanadis, C.; et al. Impact of colchicine on mortality in patients with COVID-19: A meta-analysis. Hell. J. Cardiol. 2021. [Epub ahead of print]. [Google Scholar] [CrossRef] [PubMed]
  103. Salah, H.M.; Mehta, J.L. Meta-analysis of the Effect of Colchicine on Mortality and Mechanical Ventilation in COVID-19. Am. J. Cardiol. 2021, 145, 170–172. [Google Scholar] [CrossRef]
  104. Hariyanto, T.I.; Halim, D.A.; Jodhinata, C.; Yanto, T.A.; Kurniawan, A. Colchicine treatment can improve outcomes of coronavirus disease 2019 (COVID-19): A systematic review and meta-analysis. Clin. Exp. Pharmacol. Physiol. 2021, 48, 823–830. [Google Scholar] [CrossRef]
  105. Chiu, L.; Chow, R.; Chiu, N.; Lo, C.-H.; Aggarwal, R.; Lee, J.; Choi, Y.-G.; Lam, H.; Prsic, E.H.; Shin, H.J. Colchicine use in patients with COVID-19: A systematic review and meta-analysis. medRxiv 2021. [Google Scholar] [CrossRef]
  106. Golpour, M.; Mousavi, T.; Alimohammadi, M.; Mosayebian, A.; Shiran, M.; Alizadeh Navaei, R.; Rafiei, A. The effectiveness of Colchicine as an anti-inflammatory drug in the treatment of coronavirus disease 2019: Meta-analysis. Int. J. Immunopathol. Pharmacol. 2021, 35, 205873842110317. [Google Scholar] [CrossRef] [PubMed]
  107. Elshafei, M.N.; El-Bardissy, A.; Khalil, A.; Danjuma, M.; Mubasher, M.; Abubeker, I.Y.; Mohamed, M.F.H. Colchicine use might be associated with lower mortality in COVID-19 patients: A meta-analysis. Eur. J. Clin. Invest. 2021, 51, e13645. [Google Scholar] [CrossRef] [PubMed]
  108. Nawangsih, E.N.; Kusmala, Y.Y.; Rakhmat, I.I.; Handayani, D.R.; Juliastuti, H.; Wibowo, A.; Lim, M.A.; Pranata, R. Colchicine and mortality in patients with coronavirus disease 2019 (COVID-19) pneumonia: A systematic review, meta-analysis, and meta-regression. Int. Immunopharmacol. 2021, 96, 107723. [Google Scholar] [CrossRef] [PubMed]
  109. Lien, C.-H.; Lee, M.-D.; Weng, S.-L.; Lin, C.-H.; Liu, L.Y.-M.; Tai, Y.-L.; Lei, W.-T.; Liu, J.-M.; Huang, Y.-N.; Chi, H.; et al. Repurposing Colchicine in Treating Patients with COVID-19: A Systematic Review and Meta-Analysis. Life 2021, 11, 864. [Google Scholar] [CrossRef]
  110. Bassetti, M.; Giacobbe, D.R.; Bruzzi, P.; Barisione, E.; Centanni, S.; Castaldo, N.; Corcione, S.; De Rosa, F.G.; Di Marco, F.; Gori, A.; et al. Clinical Management of Adult Patients with COVID-19 Outside Intensive Care Units: Guidelines from the Italian Society of Anti-Infective Therapy (SITA) and the Italian Society of Pulmonology (SIP). Infect. Dis. Ther. 2021, 30, 1–49. [Google Scholar] [CrossRef]
Figure 1. Contributory mechanisms of hypercoagulability in COVID 19 infection are autoimmunity and anti-phospholipid antibodies, cytokine storm-mediated thrombin overproduction, increased tissue factor and plasminogen activator inhibitor-2 levels due to ARDS-related tissue damage and ACE2 receptor-mediated endothelial dysfunction. APA anti-phospholipid antibodies, IL1RA interleukin 1 receptor agonist, IL-2R interleukin 2 receptor, ARDS acute respiratory distress syndrome, TNF tissue necrosis factor, ACE angiotensin converting enzyme, TF tissue factor, PAI-2 plasminogen activator inhibitor-2.
Figure 1. Contributory mechanisms of hypercoagulability in COVID 19 infection are autoimmunity and anti-phospholipid antibodies, cytokine storm-mediated thrombin overproduction, increased tissue factor and plasminogen activator inhibitor-2 levels due to ARDS-related tissue damage and ACE2 receptor-mediated endothelial dysfunction. APA anti-phospholipid antibodies, IL1RA interleukin 1 receptor agonist, IL-2R interleukin 2 receptor, ARDS acute respiratory distress syndrome, TNF tissue necrosis factor, ACE angiotensin converting enzyme, TF tissue factor, PAI-2 plasminogen activator inhibitor-2.
Jcm 10 05128 g001
Figure 2. Colchicine’s mechanisms of actions generate the multifaceted hypothesis against COVID-19 infection.
Figure 2. Colchicine’s mechanisms of actions generate the multifaceted hypothesis against COVID-19 infection.
Jcm 10 05128 g002
Figure 3. PubMed Query Flowchart.
Figure 3. PubMed Query Flowchart.
Jcm 10 05128 g003
Table 1. Therapeutic modalities against COVID-19 infection.
Table 1. Therapeutic modalities against COVID-19 infection.
Therapeutic StrategyAgent/Approach Tested
Immunizationactive immunity: vaccines
passive immunity: plasma, immunoglobulins
Antiviral Agentsfusion inhibitors
RNA polymerase inhibitors
protease inhibitors
endosome acidification inhibitors
Supportive CareECMO
artificial liver system (ALS)
cytokine filters
thromboprophylaxis
ImmunomodulationInterferons
Steroids
Colchicine
Macrolides
JAK inhibitors
BTK inhibitors
IL-6 inhibitors
IL-1 inhibitors
Anti-GM-CSF Abs
Thalidomide
Cell therapies
ECMO, extracorporeal membrane oxygenation; JAK, Janus kinase; BTK, bruton tyrosine kinase; IL, interleukin; anti-GM-CSF Abs, antibodies against granulocyte-macrophage colony stimulating factor.
Table 2. Main characteristics and outcomes of completed studies investigating the impact of colchicine in SARS-COV-2 infected patients.
Table 2. Main characteristics and outcomes of completed studies investigating the impact of colchicine in SARS-COV-2 infected patients.
Author Country YearDesignnAge
(%)
BMI (kg/m2)DM (%)Colchicine DosageSoCFindingsOutcome
Randomized Controlled Trials
Deftereos SG [87]
Greece
April 2020
prospective, open-label, RCT1056441.927.510.5L:2 mg
M: 0.5 mg BD
up to 21 days
HCQ ± AZM ± LPV/r ± tocilizumabWHO-OSCD: 1.8% vs. 14%
OR 0.11; 95% CI: 0.01–0.96; p: 0.02
Positive
Tardif JC et al. [90]
Multinational
March 2020–January 2021
double-blinded RCT
non-hospitalized patients
4488 5453.93019.90.5 mg BD 3d
0.5 mg OD 27d
HCQ
OACs
Anti-platelets
Composite of death or hospitalization
4.7% vs. 5.8%
OR:0.79; 95% CI: 0.61–1.03; p: 0.081
Secondary endpoint, Mechanical ventilation
OR: 0.53; 95% CI: 0.25–1.09
secondary analysis PCR-confirmed COVID-19
Composite of death or hospitalization 4.6% vs. 6%
OR: 0.75; 95% CI: 0.57–0.99; p: 0.042
Any hospital admission (not pre-specified)
OR: 0.76; 95% CI: 0.58–0.99; p: 0.04
Neutral
Lopes M. et al. [93]
Brazil
April–August 2020
double blinded RCT72555431.6390.5 mg q 8 h 5 days
0.5 mg BD 5 days
AZM 500mg OD up to 7 days
HCQ 400 mg BD daily for 2 days, then 400 mg OD up to 8 days
Need for supplemental oxygen
4.0 days vs. 6.5 days; p < 0.001
Time of hospitalization
7.0 days vs. 9.0 days; p: 0.003
Positive
Mareev VY et al. [94]
(COLORIT study)
Russia
prospective comparative RCT43613030.411.61 mg/day for 3 days
0.5 mg/day for 14 days
NAPrimary endpoint
SHOCS-COVID score change day 12
8 to 2 vs. 7 to 7 (p: 0.002)
Secondary outcomes
Hospital stay: 13 vs. 17.5; p: 0.079
Any oxygen support: decreased from 50% to 9.5%; p: 0.011
Deaths: 0 vs. 2; p: 0.467
Positive
Horby PW et al. [91]
Pre-print
RECOVERY
NCT04381936 Multinational
November 2020–March 2021
open-label, RCT1134063.430.5NA25.5L: 1 mg + 0.5 mg 12 h later
M: 0.5 mg BD up to 10 days
corticosteroids
remdesivir
tocilizumab
convalescent plasma
baricitinib
aspirin
Primary outcome
28-day all-cause mortality
RR: 1.01; 95% CI: 0.93–1.0; p: 0.77
Secondary outcomes
time to discharge 10 vs. 10 d
invasive mechanical ventilation 11 vs. 11%
Neutral
Observational Studies
Brunetti L et al. [95]
USA
March–May 2020
propensity matched retrospective observational cohort6662.934.830.721.2L: 1.2 mg
M: 0.6 mg BD
AZM, HCQ ± remdesivir or tocilizumab28-days mortality
OR:0.20; 95% Cl:0.05–0.80; p: 0.023
WHO-OSCI days 14 and 28
57.6% vs. 51.5%; p: 0.621
Not requiring supplemental oxygen on days 14 and 28
54.5% vs. 54.5%, p: 1.0
Hospital discharge by day 28
OR: 5.0; 95% CI: 1.25–20.1; p: 0.023
Positive
Scarsi M et al. [96]
Italy
March–April 2020
retrospective, case-control
observational study
2627036.5NANAM: 1 mg/dayHCQ ±
dexamethasone ±
LPV/r
21 days survival rate:
84.2% vs. 63.6%, p: 0.001
adj HR: 0.151; 95% CI: 0.062–0.368; p < 0.0001
Positive
Sandhu T et al. [97]
USA
March–May 2020
prospective comparative cohort study112674327.5420.6 mg BD 3 days
0.6 mg OD for 9 days
HCQ, steroids, oseltmamivir
Excluded if on: lamivudine dolutegravir, tocilizumab convalescent plasma
Mortality: 49.1% vs. 72.9%; p: 0.002
Intubations: 52.8% vs. 73.6%; p: 0.006
Discharge rate: 50.9% vs. 27.1%; p: 0.002
Positive
Kevorkian JP et al. [98]
(COCAA-COLA study)
France
January–November 2020
observational cohort study6866 2227 44Prednisone
1 mg/kg/day
Furosemide
80 mg/day
Salicylate
75 mg/day
Colchicine
L: 1.5 mg
M: 0.5 mg q 8 h
Rivaroxaban or Apixaban
dexamethasone
(6 mg OD for up to 10 days)
LMWH
Primary composite endpoint
OR: 0.097; 95% CI: 0.001–0.48; p: 0.0009
  • requirement of HFOT: 13% vs. 4%; p: 0.38
  • non-invasive mechanical ventilation: 13% vs. 0%; p: 0.07
  • invasive mechanical ventilation: 15% vs. 4%; p: 0.21
  • 28-day mortality: 5% vs. 0%; p: 0.5
Positive
Manenti L et al. [99]
Italy
February–April 2020
retrospective cohort study
age & sex matched
14161.52927.517M: 1 mg/day
up to 21 days
ABX, antivirals, HCQ, i.v steroids, tocilizumab21 days mortality
adj HR: 0.24; 95% CI: 0.09–0.67; p: 0.006
WHO-OSCI:
adj relative improvement rate 1.80
95% CI: 1.00–3.22; p: 0.048
Positive
García-Posada M et al. [100]
Colombia
May–August 2020
descriptive
observational study
2096039NA25.3L:2 mg
M: 0.5 mg BD
up to 20 days
varying combinations of
ABX corticosteroids
LMWH
or tocilizumab
All-cause mortality:
(combination of ABX, LMWH, colchicine, corticosteroids)
OR: 0.26; 95% CI: 0.08−0.71; p < 0.05
positive
Abbreviations; n: number of study participants, female symbol, DM: diabetes mellitus, SoC: standard of care, L: loading dose, M: maintenance dose, ABX: antibiotics, BD: twice daily, OD: once daily, RCT: randomized clinical trial, WHO-OSCI: ordinal scale clinical improvement, WHO-OSCD: ordinal scale clinical deterioration, LPV/r: Lopinavir /ritonavir, AZM: azithromycin, HCQ: hydroxychloroquine, OACs: oral anticoagulants, LMWH: low molecular weight heparin, HFOT: high flow oxygen therapy, SHOCS-COVID score: assessment of the patient’s clinical condition, computed tomography (CT) score of the lung tissue damage, CRP changes, D-dimer changes.
Table 3. 8 meta-analyses reporting effect of colchicine administration in COVID-19 mortality. (Abbreviations: OR odds ratio, RR relative risk, CI confidence interval, n/R not reported, n population, RCTs randomized clinical trials).
Table 3. 8 meta-analyses reporting effect of colchicine administration in COVID-19 mortality. (Abbreviations: OR odds ratio, RR relative risk, CI confidence interval, n/R not reported, n population, RCTs randomized clinical trials).
Meta-AnalysisJournalnStudies IncludedMortality Effect
Vrachatis et al., 2021 [102]Hell J Cardiol8816 studies
3 cohorts
2 RCTs
1 case-control
OR 0.35
(95% CI: 0.24–0.52; p < 0.05)
Salah et al., 2021 [103]Am J Cardiol52598 studies
4 cohorts
3 RCTs
1 case-control
RR 0.62
(95% CI: 0.48–0.81; p = 0.0005)
Hariyanto et al., 2021 [104]Clin Exp Pharmacol Physiol57788 studies
4 cohorts
3 RCTs
1 case-control
OR 0.43
(95% CI: 0.32–0.58; p = n/R)
Chiu et al., 2021 [105]medRxiv (pre-print)50336 studies
3 cohorts
3 RCTs
OR 0.36
(95% CI: 0.17–0.76; p = n/R)
Golpour et al., 2021 [106]Int J Immunopathol Pharmacol567810 studies
5 cohorts
4 RCTs
1 case-control
RR 0.365
(95% CI: 0.555–0.748; p < 0.05)
Elshafei et al., 2021 [107]Eur J Clin Invest55229 studies
4 cohorts
4 RCTs
1 case-control
OR 0.35
(95% CI: 0.25–0.48; p = n/R)
Nawangsih et al., 2021 [108]Int Immunopharmacol55308 studies
5 cohorts
3 RCTs
OR 0.47
(95% CI: 0.31–0.72; p = 0.001)
Lien et al., 2021 [109]Life (Basel)17,20511 studies
7 cohorts
4 RCTs
OR 0.57
(95% CI: 0.38–0.87; p < 0.01)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vrachatis, D.A.; Papathanasiou, K.A.; Giotaki, S.G.; Raisakis, K.; Kossyvakis, C.; Kaoukis, A.; Kolokathis, F.; Deftereos, G.; Iliodromitis, K.E.; Avramides, D.; et al. Immunologic Dysregulation and Hypercoagulability as a Pathophysiologic Background in COVID-19 Infection and the Immunomodulating Role of Colchicine. J. Clin. Med. 2021, 10, 5128. https://doi.org/10.3390/jcm10215128

AMA Style

Vrachatis DA, Papathanasiou KA, Giotaki SG, Raisakis K, Kossyvakis C, Kaoukis A, Kolokathis F, Deftereos G, Iliodromitis KE, Avramides D, et al. Immunologic Dysregulation and Hypercoagulability as a Pathophysiologic Background in COVID-19 Infection and the Immunomodulating Role of Colchicine. Journal of Clinical Medicine. 2021; 10(21):5128. https://doi.org/10.3390/jcm10215128

Chicago/Turabian Style

Vrachatis, Dimitrios A., Konstantinos A. Papathanasiou, Sotiria G. Giotaki, Konstantinos Raisakis, Charalampos Kossyvakis, Andreas Kaoukis, Fotis Kolokathis, Gerasimos Deftereos, Konstantinos E. Iliodromitis, Dimitrios Avramides, and et al. 2021. "Immunologic Dysregulation and Hypercoagulability as a Pathophysiologic Background in COVID-19 Infection and the Immunomodulating Role of Colchicine" Journal of Clinical Medicine 10, no. 21: 5128. https://doi.org/10.3390/jcm10215128

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop