Article Data

  • Views 5770
  • Dowloads 1204

Reviews

Open Access Special Issue

Therapeutic agents for ARDS

  • Elena Campaña-Duel1,2
  • Marta Camprubí-Rimblas1,3,*,

1Critical Care Research Center, Parc Taulí Hospital Universitari, 08208 Sabadell, Spain

2Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain

3CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain

DOI: 10.22514/sv.2022.041 Vol.18,Issue 5,September 2022 pp.12-32

Submitted: 31 December 2021 Accepted: 23 March 2022

Published: 08 September 2022

(This article belongs to the Special Issue New Insights in Acute Respiratory Distress Syndrome)

*Corresponding Author(s): Marta Camprubí-Rimblas E-mail: mcamprubi@tauli.cat

Abstract

In spite of supportive care of patients with acute respiratory distress syndrome (ARDS), morbidity and mortality of these patients are considerable and effective therapies centred in ARDS pathophysiology are needed. Substantial progress in pharmacological therapies has been noticed, however, several studies have not been successfully translated to the clinics. Nonetheless, many preclinical and clinical studies are ongoing. In this review, pharmacological therapies underlying ARDS pathophysiology are summarized: therapies targeting the alveolocapillary membrane, mucolytics, bronchodilators, immunomodulators, anticoagulants and fibrinolytics, aspirin, and other treatments are discussed, including both, studies with beneficial and controversial results, and ongoing trials. In addition, a section concerning preclinical studies is included. An enlarged understanding of ARDS pathophysiology and its fundamental pathways and mechanism, together with the identification of ARDS subsets of patients and phenotypes will maximise patient response to a specific treatment.


Keywords

Acute respiratory distress syndrome (ARDS); Acute lung injury; COVID-19; Sepsis


Cite and Share

Elena Campaña-Duel,Marta Camprubí-Rimblas. Therapeutic agents for ARDS. Signa Vitae. 2022. 18(5);12-32.

References

[1] Matthay MA, Zemans RL, Zimmerman GA, Arabi YM, Beitler JR, Mercat A, et al. Acute respiratory distress syndrome. Nature Reviews Disease Primers. 2019; 5: 18.

[2] Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Internal Medicine. 2020; 180: 934–943.

[3] Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016; 315: 788.

[4] Villar J, Blanco J, Añón JM, Santos-Bouza A, Blanch L, Ambrós A, et al. The ALIEN study: incidence and outcome of acute respiratory distress syndrome in the era of lung protective ventilation. Intensive Care Medicine. 2011; 37: 1932–1941.

[5] Herridge MS, Tansey CM, Matté A, Tomlinson G, Diaz-Granados N, Cooper A, et al. Functional disability 5 years after acute respiratory distress syndrome. New England Journal of Medicine. 2011; 364: 1293–1304.

[6] Gonzales JN, Lucas R, Verin AD. The acute respiratory distress syndrome: mechanisms and perspective therapeutic approaches. Austin Journal of Vascular Medicine. 2015; 2: 1009.

[7] Ware LB, Matthay MA. The acute respiratory distress syndrome. New England Journal of Medicine. 2000; 342: 1334–1349.

[8] Millar FR, Summers C, Griffiths MJ, Toshner MR, Proudfoot AG. The pulmonary endothelium in acute respiratory distress syndrome: insights and therapeutic opportunities. Thorax. 2016; 71: 462–473.

[9] Hrenak J, Simko F. Renin-angiotensin system: an important player in the pathogenesis of acute respiratory distress syndrome. International Journal of Molecular Science. 2020; 21: 8038.

[10] Khan A, Benthin C, Zeno B, Albertson TE, Boyd J, Christie JD, et al. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Critical Care. 2017; 21: 234.

[11] Cross LJM, Matthay MA. Biomarkers in acute lung injury: insights into the pathogenesis of acute lung injury. Critical Care Clinics. 2011; 27: 355–377.

[12] Wong MH, Johnson MD. Differential response of primary alveolar type I and type II cells to LPS stimulation. PLoS One. 2013; 8: e55545.

[13] Lopez-Rodriguez E, Gay-Jordi G, Mucci A, Lachmann N, Serrano-Mollar A. Lung surfactant metabolism: early in life, early in disease and target in cell therapy. Cell and Tissue Research. 2017; 367: 721–735.

[14] Ware L. Pathophysiology of acute lung injury and the acute respiratory distress syndrome. Seminars in Respiratory and Critical Care Medicine. 2006; 27: 337–349.

[15] Lewis JF, Jobe AH. Surfactant and the adult respiratory distress syndrome. American Review of Respiratory Disease. 1993; 147: 218–233.

[16] Rodriguez-Fanjul J, Jordan I, Balaguer M, Batista-Muñoz A, Ramon M, Bobillo-Perez S. Early surfactant replacement guided by lung ultrasound in preterm newborns with RDS: the ULTRASURF randomised controlled trial. European Journal of Pediatrics. 2020; 179: 1913–1920.

[17] Anzueto A, Baughman RP, Guntupalli KK, Weg JG, Wiedemann HP, Raventós AA, et al. Aerosolized surfactant in adults with sepsis-induced acute respiratory distress syndrome. New England Journal of Medicine. 1996; 334: 1417–1422.

[18] Spragg RG, Lewis JF, Walmrath H, Johannigman J, Bellingan G, Laterre P, et al. Effect of recombinant surfactant protein c-based surfactant on the acute respiratory distress syndrome. New England Journal of Medicine. 2004; 351: 884–892.

[19] Meng H, Sun Y, Lu J, Fu S, Meng Z, Scott M, et al. Exogenous surfactant may improve oxygenation but not mortality in adult patients with acute lung injury/acute respiratory distress syndrome: a meta-analysis of 9 clinical trials. Journal of Cardiothoracic and Vascular Anesthesia. 2012; 26: 849–856.

[20] Taut FJH, Rippin G, Schenk P, Findlay G, Wurst W, Häfner D, et al. A search for subgroups of patients with ARDS who may benefit from surfactant replacement therapy. Chest. 2008; 134: 724–732.

[21] Piva S, DiBlasi RM, Slee AE, Jobe AH, Roccaro AM, Filippini M, et al. Surfactant therapy for COVID-19 related ARDS: a retrospective case–control pilot study. Respiratory Research. 2021; 22: 20.

[22] Ware L, Matthay M. Alveolar fluid clearance is impaired in the majority of patients with acute lung injury and the acute respiratory distress syndrome. American Journal of Respiratory and Critical Care Medicine. 2001; 163: 1376–1383.

[23] Shabbir W, Scherbaum-Hazemi P, Tzotzos S, Fischer B, Fischer H, Pietschmann H, et al. Mechanism of action of novel lung edema therapeutic AP301 by activation of the epithelial sodium channel. Molecular Pharmacology. 2013; 84: 899–910.

[24] Krenn K, Croize A, Klein KU. Oral inhalation of AP301 peptide activates pulmonary oedema clearance: initial results from a phase IIa clinical trial in mechanically ventilated ICU patients. European Respiratory Journal. 2014; 44: 1386.

[25] Krenn K, Lucas R, Croizé A, Boehme S, Klein KU, Hermann R, et al. Inhaled AP301 for treatment of pulmonary edema in mechanically ventilated patients with acute respiratory distress syndrome: a phase IIa randomized placebo-controlled trial. Critical Care. 2017; 21: 194.

[26] Schmid B, Kredel M, Ullrich R, Krenn K, Lucas R, Markstaller K, et al. Safety and preliminary efficacy of sequential multiple ascending doses of solnatide to treat pulmonary permeability edema in patients with moderate-to-severe ARDS—a randomized, placebo-controlled, double-blind trial. Trials. 2021; 22: 643.

[27] Ware LB, Matthay MA. Keratinocyte and hepatocyte growth factors in the lung: roles in lung development, inflammation, and repair. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2002; 282: L924–L940.

[28] Franco-Montoya M, Bourbon JR, Durrmeyer X, Lorotte S, Jarreau P, Delacourt C. Pulmonary effects of keratinocyte growth factor in newborn rats exposed to hyperoxia. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2009; 297: L965–L976.

[29] McAuley DF, Cross LM, Hamid U, Gardner E, Elborn JS, Cullen KM, et al. Keratinocyte growth factor for the treatment of the acute respiratory distress syndrome (KARE): a randomised, double-blind, placebo-controlled phase 2 trial. The Lancet Respiratory Medicine. 2017; 5: 484–491.

[30] Cynober L. Citrulline: just a biomarker or a conditionally essential amino acid and a pharmaconutrient in critically ill patients? Critical Care. 2013; 17: 122.

[31] Afshari A, Brok J, Møller AM, Wetterslev J. Inhaled nitric oxide for acute respiratory distress syndrome and acute lung injury in adults and children. Anesthesia & Analgesia. 2011; 112: 1411–1421.

[32] Wagenaar GTM, Hiemstra PS, Gosens R. Therapeutic potential of soluble guanylate cyclase modulators in neonatal chronic lung disease. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2015; 309: L1037–L1040.

[33] Enomoto M, Jain A, Pan J, Shifrin Y, Van Vliet T, McNamara PJ, et al. Newborn rat response to single vs. combined cGMP-dependent pulmonary vasodilators. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2014; 306: L207–L215.

[34] Haeberle H, Prohaska S, Martus P, Straub A, Zarbock A, Marx G, et al. Therapeutic iloprost for the treatment of acute respiratory distress syndrome (ARDS) (the ThIlo trial): a prospective, randomized, multicenter phase II study. Trials. 2020; 21: 242.

[35] Ware LB. VEGF levels in the alveolar compartment do not distinguish be-tween ARDS and hydrostatic pulmonary oedema. European Respiratory Journal. 2005; 26: 101–105.

[36] Watanabe M, Boyer JL, Crystal RG. Genetic delivery of bevacizumab to suppress vascular endothelial growth factor-induced high-permeability pulmonary edema. Human Gene Therapy. 2009; 20: 598–610.

[37] Patel G, Pielykh D, Patel SM, Patel MJ, Bhavsar K, Koritala T. COVID-19 coronavirus-induced atypical pneumonia: efficacy of the monoclonal antibody bevacizumab in moderate to severe cases. Cureus. 2021; 13: e18317.

[38] Häberle HA. Levosimendan—a 20-year experience. Anasthesiol Inten-sivmed Notfallmed Schmerzther. 2021; 56: 414–426. (In German)

[39] Morelli A, Teboul J, Maggiore SM, Vieillard-Baron A, Rocco M, Conti G, et al. Effects of levosimendan on right ventricular afterload in patients with acute respiratory distress syndrome: a pilot study. Critical Care Medicine. 2006; 34: 2287–2293.

[40] Shankar-Hari M, Santhakumaran S, Prevost AT, Ward JK, Marshall T, Bradley C, et al. Defining phenotypes and treatment effect heterogeneity to inform acute respiratory distress syndrome and sepsis trials: secondary analyses of three RCTs. NIHR Journals Library: Southampton. 2021.

[41] Ghorbi M, Rashidi M, Olapour A, Javaherforooshzadeh F, Akhondzadeh R. Effect of N-acetylcysteine on the treatment of acute respiratory distress syndrome in mechanically ventilated patients admitted to the intensive care unit. Medical Journal of the Islamic Republic of Iran. 2021; 35: 87.

[42] Taher A, Lashgari M, Sedighi L, Rahimi-bashar F, Poorolajal J, Mehrpooya M. A pilot study on intravenous N-acetylcysteine treatment in patients with mild-to-moderate COVID19-associated acute respiratory distress syndrome. Pharmacological Reports. 2021; 73: 1650–1659.

[43] Denning NL, Aziz M, Gurien SD, Wang P. DAMPs and NETs in sepsis. Frontiers in Immunology. 2019; 10: 2536.

[44] Paul K, Rietschel E, Ballmann M, Griese M, Worlitzsch D, Shute J, et al. Effect of treatment with dornase alpha on airway inflammation in patients with cystic fibrosis. American Journal of Respiratory and Critical Care Medicine. 2004; 169: 719–725.

[45] Perkins GD, McAuley DF, Thickett DR, Gao F. The beta-agonist lung injury trial (BALTI): a randomized placebo-controlled clinical trial: a randomized placebo-controlled clinical trial. American Journal of Respiratory and Critical Care Medicine. 2006; 173: 281–287.

[46] Gates S, Perkins GD, Lamb SE, Kelly C, Thickett DR, Young JD, et al. Beta-agonist lung injury TrIal-2 (BALTI-2): a multicentre, randomised, double-blind, placebo-controlled trial and economic evaluation of intravenous infusion of salbutamol versus placebo in patients with acute respiratory distress syndrome. Health Technology Assessment. 2013; 17: v–vi, 1–87.

[47] Smith FG, Perkins GD, Gates S, Young D, McAuley DF, Tunnicliffe W, et al. Effect of intravenous β-2 agonist treatment on clinical outcomes in acute respiratory distress syndrome (BALTI-2): a multicentre, randomised controlled trial. The Lancet. 2012; 379: 229–235.

[48] National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network, Matthay MA, Brower RG, Carson S, Douglas IS, Eisner M, et al. Randomized, placebo-controlled clinical trial of an aerosolized β2-agonist for treatment of acute lung injury. American Journal of Respiratory and Critical Care Medicine. 2011; 184: 561–568.

[49] Hraiech S, Yoshida T, Annane D, Duggal A, Fanelli V, Gacouin A, et al. Myorelaxants in ARDS patients. Intensive Care Medicine. 2020; 46: 2357–2372.

[50] Papazian L, Forel J, Gacouin A, Penot-Ragon C, Perrin G, Loundou A, et al. Neuromuscular blockers in early acute respiratory distress syndrome. New England Journal of Medicine. 2010; 363: 1107–1116.

[51] Moss M, Huang DT, Brower RG, Ferguson ND, Ginde AA, Gong MN, et al. Early neuromuscular blockade in the acute respiratory distress syndrome. New England Journal of Medicine. 2019; 380: 1997–2008.

[52] Alhazzani W, Belley-Cote E, Møller MH, Angus DC, Papazian L, Arabi YM, et al. Neuromuscular blockade in patients with ARDS: a rapid practice guideline. Intensive Care Medicine. 2020; 46: 1977–1986.

[53] Choi H, Shin B, Yoo H, Suh GY, Cho JH, Kim HK, et al. Early corticosteroid treatment for postoperative acute lung injury after lung cancer surgery. Therapeutic Advances in Respiratory Disease. 2019; 13: 175346661984025.

[54] Sun S, Liu D, Zhang H, Zhang X, Wan B. Effect of different doses and time-courses of corticosteroid treatment in patients with acute respiratory distress syndrome: a meta-analysis. Experimental and Therapeutic Medicine. 2019; 18: 4637–4644.

[55] Mammen MJ, Aryal K, Alhazzani W, Alexander PE. Corticosteroids for patients with acute respiratory distress syndrome: a systematic review and meta-analysis of randomized trials. Polish Archives of Internal Medicine. 2020; 130: 276–286.

[56] Ni Y, Chen G, Sun J, Liang B, Liang Z. The effect of corticosteroids on mortality of patients with influenza pneumonia: a systematic review and meta-analysis. Critical Care. 2019; 23: 99.

[57] Tsai M, Yang K, Chan M, Kao K, Wang H, Perng W, et al. Impact of corticosteroid treatment on clinical outcomes of influenza-associated ARDS: a nationwide multicenter study. Annals of Intensive Care. 2020; 10: 26.

[58] Torres A, Sibila O, Ferrer M, Polverino E, Menendez R, Mensa J, et al. Effect of corticosteroids on treatment failure among hospitalized patients with severe community-acquired pneumonia and high inflammatory response. JAMA. 2015; 313: 677.

[59] Siemieniuk RAC, Meade MO, Alonso-Coello P, Briel M, Evaniew N, Prasad M, et al. Corticosteroid therapy for patients hospitalized with community-acquired pneumonia. Annals of Internal Medicine. 2015; 163: 519–528.

[60] Villar J, Ferrando C, Martínez D, Ambrós A, Muñoz T, Soler JA, et al. Dexamethasone treatment for the acute respiratory distress syndrome: a multicentre, randomised controlled trial. Lancet Respiratory Medicine. 2020; 8: 267–276.

[61] Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, et al. Dexamethasone in hospitalized patients with Covid-19. New England Journal of Medicine. 2021; 384: 693–704.

[62] Tongyoo S, Permpikul C, Mongkolpun W, Vattanavanit V, Udompanturak S, Kocak M, et al. Hydrocortisone treatment in early sepsis-associated acute respiratory distress syndrome: results of a randomized controlled trial. Critical Care. 2016; 20: 329.

[63] Meduri UG, Headley SA, Golden E, Carson SJ, Umberger RA, Kelso T, et al. Effect of prolonged methylprednisolone therapy in unresolving acute respiratory distress syndrome: a randomized controlled trial. Survey of Anesthesiology. 1999; 43: 135–136.

[64] Meduri GU, Golden E, Freire AX, Taylor E, Zaman M, Carson SJ, et al. Methylprednisolone infusion in early severe ARDS: results of a randomized controlled trial. Chest. 2007; 131: 954–963.

[65] Festic E, Carr GE, Cartin-Ceba R, Hinds RF, Banner-Goodspeed V, Bansal V, et al. Randomized clinical trial of a combination of an inhaled corticosteroid and beta agonist in patients at risk of developing the acute respiratory distress syndrome. Critical Care Medicine. 2017; 45: 798–805.

[66] Mohamed H, Meguid MA. Effect of nebulized budesonide on respiratory mechanics and oxygenation in acute lung injury/acute respiratory distress syndrome: Randomized controlled study. Saudi Journal of Anaesthesia. 2017; 11: 9.

[67] Yeh TF, Chen CM, Wu SY, Husan Z, Li TC, Hsieh WS, et al. Intratracheal administration of budesonide/surfactant to prevent bronchopulmonary dysplasia. American Journal of Respiratory and Critical Care Medicine. 2016; 193: 86–95.

[68] Kothe TB, Sadiq FH, Burleyson N, Williams HL, Anderson C, Hillman NH. Surfactant and budesonide for respiratory distress syndrome: an observational study. Pediatric Research. 2020; 87: 940–945.

[69] Oh TK, Song I, Choi S. Prior statin therapy and mortality among critically ill patients: a systemic review and meta-analysis of cohort studies. Annals of Translational Medicine. 2020; 8: 396–396.

[70] Calfee CS, Delucchi KL, Sinha P, Matthay MA, Hackett J, Shankar-Hari M, et al. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. The Lancet Respiratory Medicine. 2018; 6: 691–698.

[71] Sinha P, Delucchi KL, Thompson BT, McAuley DF, Matthay MA, Calfee CS. Latent class analysis of ARDS subphenotypes: a secondary analysis of the statins for acutely injured lungs from sepsis (SAILS) study. Intensive Care Medicine. 2018; 44: 1859–1869.

[72] Ryter SW, Otterbein LE, Morse D, Choi AMK. Heme oxygenase/carbon monoxide signaling path-ways: regulation and functional significance. Molecular and Cellular Biochemistry. 2002; 234–235: 249–263.

[73] Jung S, Moon J, Xu J, Ifedigbo E, Ryter SW, Choi AMK, et al. Carbon monoxide negatively regulates NLRP3 inflammasome activation in macrophages. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2015; 308: L1058–L1067.

[74] Jahn N, Lamberts RR, Busch CJ, Voelker MT, Busch T, Koel-Simmelink MJA, et al. Inhaled carbon monoxide protects time-dependently from loss of hypoxic pulmonary vasoconstriction in endotoxemic mice. Respiratory Research. 2015; 16: 119.

[75] Fredenburgh LE, Kraft BD, Hess DR, Harris RS, Wolf MA, Suliman HB, et al. Effects of inhaled CO administration on acute lung injury in baboons with pneumococcal pneumonia. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2015; 309: L834–L846.

[76] Islam D, Huang Y, Fanelli V, Delsedime L, Wu S, Khang J, et al. Identification and modulation of microenvironment is crucial for effective mesenchymal stromal cell therapy in acute lung injury. American Journal of Respiratory and Critical Care Medicine. 2019; 199: 1214–1224.

[77] Bellingan G, Jacono F, Bannard-Smith J, Brealey D, Meyer N, Thickett D, et al. Primary analysis of a phase 1/2 study to assess multistem® cell therapy, a regenerative advanced therapy medicinal product (ATMP), in acute respiratory distress syndrome (MUST-ARDS). B14. Late Breaking Clinical Trials. 2019; 199: A7353.

[78] Matthay MA, Calfee CS, Zhuo H, Thompson BT, Wilson JG, Levitt JE, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. The Lancet Respiratory Medicine. 2019; 7: 154–162.

[79] Leng Z, Zhu R, Hou W, Feng Y, Yang Y, Han Q, et al. Transplantation of ACE2- mesenchymal stem cells improves the outcome of patients with COVID-19 Pneumonia. Aging and Disease. 2020; 11: 216.

[80] Chen J, Zhang X, Xie J, Xue M, Liu L, Yang Y, et al. Overexpression of TGFβ1 in murine mesenchymal stem cells improves lung inflammation by impacting the Th17/Treg balance in LPS-induced ARDS mice. Stem Cell Research & Therapy. 2020; 11: 311.

[81] Fowler AA 3rd, Truwit JD, Hite RD, Morris PE, DeWilde C, Priday A, et al. Effect of vitamin C infusion on organ failure and biomarkers of inflammation and vascular injury in patients with sepsis and severe acute respiratory failure: the CITRIS-ALI randomized clinical trial: The CITRIS-ALI randomized clinical trial. JAMA 2019; 322: 1261–1270.

[82] Chiscano-Camón L, Ruiz-Rodriguez JC, Ruiz-Sanmartin A, Roca O, Ferrer R. Vitamin C levels in patients with SARS-CoV-2-associated acute respiratory distress syndrome. Critical Care. 2020; 24: 522.

[83] Ji M, Chen T, Wang B, Chen M, Ding Q, Chen L, et al. Effects of ulinastatin combined with mechanical ventilation on oxygen metabolism, inflammation and stress response and antioxidant capacity of ARDS. Experimental and Therapeutic Medicine. 2018; 15: 4665–4670.

[84] Christie JD, Vaslef S, Chang PK, May AK, Gunn SR, Yang S, et al. A randomized dose-escalation study of the safety and anti-inflammatory activity of the p38 mitogen-activated protein kinase inhibitor dilmapimod in severe trauma subjects at risk for acute respiratory distress syndrome. Critical Care Medicine. 2015; 43: 1859–1869.

[85] Grimes JM, Grimes KV. P38 MAPK inhibition: a promising therapeutic approach for COVID-19. Journal of Molecular and Cellular Cardiology. 2020; 144: 63–65.

[86] Jimenez-Guardeño JM, Nieto-Torres JL, DeDiego ML, Regla-Nava JA, Fernandez-Delgado R, Castaño-Rodriguez C, et al. The PDZ-binding motif of severe acute respiratory syndrome coronavirus envelope protein is a determinant of viral pathogenesis. PLoS Pathogens. 2014; 10: e1004320.

[87] Bowman ER, Cameron CMA, Avery A, Gabriel J, Kettelhut A, Hecker M, et al. Levels of soluble CD14 and tumor necrosis factor receptors 1 and 2 may be predictive of death in severe coronavirus disease 2019. The Journal of Infectious Diseases. 2021; 223: 805–810.

[88] Proudfoot A, Bayliffe A, O’Kane CM, Wright T, Serone A, Bareille PJ, et al. Novel anti-tumour necrosis factor receptor-1 (TNFR1) domain antibody prevents pulmonary inflammation in experimental acute lung injury. Thorax. 2018; 73: 723–730.

[89] McElvaney OJ, Curley GF, Rose-John S, McElvaney NG. Interleukin-6: obstacles to targeting a complex cytokine in critical illness. The Lancet Respiratory Medicine. 2021; 9: 643–654.

[90] Morrondo CD, Zarza LP, Gil JG, Pinto Tasende JA, Diez PD, López JMB. Benefit of tocilizumab therapy for adult-onset still disease complicated with acute respiratory distress syndrome. JCR: Journal of Clinical Rheumatology. 2016; 22: 291–293.

[91] Ziegler CGK, Allon SJ, Nyquist SK, Mbano IM, Miao VN, Tzouanas CN, et al. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell. 2020; 181: 1016–1035.e19.

[92] Monk PD, Marsden RJ, Tear VJ, Brookes J, Batten TN, Mankowski M, et al. Safety and efficacy of inhaled nebulised interferon beta-1a (SNG001) for treatment of SARS-CoV-2 infection: a randomised, double-blind, placebo-controlled, phase 2 trial. The Lancet Respiratory Medicine. 2021; 9: 196–206.

[93] Ranieri VM, Pettilä V, Karvonen MK, Jalkanen J, Nightingale P, Brealey D, et al. Effect of intravenous interferon β-1a on death and days free from mechanical ventilation among patients with moderate to severe acute respiratory distress syndrome. JAMA. 2020; 323: 725.

[94] Rizzo AN, Sammani S, Esquinca AE, Jacobson JR, Garcia JGN, Letsiou E, et al. Imatinib attenuates inflammation and vascular leak in a clinically relevant two-hit model of acute lung injury. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2015; 309: L1294–L1304.

[95] Stephens RS, Johnston L, Servinsky L, Kim BS, Damarla M. The tyrosine kinase inhibitor imatinib prevents lung injury and death after intravenous LPS in mice. Physiological Reports. 2015; 3: e12589.

[96] Nejat R, Sadr AS. Are losartan and imatinib effective against SARS-CoV2 pathogenesis? a pathophysiologic-based in silico study. In Silico Pharmacology. 2021; 9: 1.

[97] Emadi A, Chua JV, Talwani R, Bentzen SM, Baddley J. Safety and efficacy of imatinib for hospitalized adults with COVID-19: a structured summary of a study protocol for a randomised controlled trial. Trials. 2020; 21: 897.

[98] Li Y, Li H, Liu S, Pan P, Su X, Tan H, et al. Pirfenidone ameliorates lipopolysaccharide-induced pulmonary inflammation and fibrosis by blocking NLRP3 inflammasome activation. Molecular Immunology. 2018; 99: 134–144.

[99] Bode C, Peukert K, Schewe J, Putensen C, Latz E, Steinhagen F. Tetracycline alleviates acute lung injury by inhibition of NLRP3 inflammasome. European Respiratory Journal. 2019; 54: PA2175.

[100] Cao F, Tian X, Li Z, Lv Y, Han J, Zhuang R, et al. Suppression of NLRP3 inflammasome by erythropoietin via the EPOR/JAK2/STAT3 pathway contributes to attenuation of acute lung injury in mice. Frontiers in Pharmacology. 2020; 11: 306.

[101] Hamilton JA. GM-CSF-dependent inflammatory pathways. Frontiers in Immunology. 2019; 10: 2055.

[102] Stanley E, Lieschke GJ, Grail D, Metcalf D, Hodgson G, Gall JA, et al. Granulocyte/macrophage colony-stimulating factor-deficient mice show no major perturbation of hematopoiesis but develop a characteristic pulmonary pathology. Proceedings of the National Academy of Sciences. 1994; 91: 5592–5596.

[103] Paine R, Wilcoxen SE, Morris SB, Sartori C, Baleeiro CEO, Matthay MA, et al. Transgenic overexpression of granulocyte macrophage-colony stimulating factor in the lung prevents hyperoxic lung injury. The American Journal of Pathology. 2003; 163: 2397–2406.

[104] Cakarova L, Marsh LM, Wilhelm J, Mayer K, Grimminger F, Seeger W, et al. Macrophage tumor necrosis factor-alpha induces epithelial expression of granulocyte-macrophage colony-stimulating factor: impact on alveolar epithelial repair: impact on alveolar epithelial repair. American Journal of Respiratory and Critical Care Medicine. 2009; 180: 521–532.

[105] Paine R, Standiford TJ, Dechert RE, Moss M, Martin GS, Rosenberg AL, et al. A randomized trial of recombinant human granulocyte-macrophage colony stimulating factor for patients with acute lung injury. Critical Care Medicine. 2012; 40: 90–97.

[106] Herold S, Hoegner K, Vadász I, Gessler T, Wilhelm J, Mayer K, et al. Inhaled granulocyte/macrophage colony-stimulating factor as treatment of pneumonia-associated acute respiratory distress syndrome. American Journal of Respiratory and Critical Care Medicine. 2014; 189: 609–611.

[107] Cremer PC, Abbate A, Hudock K, McWilliams C, Mehta J, Chang SY, et al. Mavrilimumab in patients with severe COVID-19 pneumonia and systemic hyperinflammation (MASH-COVID): an investigator initiated, multicentre, double-blind, randomised, placebo-controlled trial. The Lancet Rheumatology. 2021; 3: e410–e418.

[108] Mehta P, Porter JC, Manson JJ, Isaacs JD, Openshaw PJM, McInnes IB, et al. Therapeutic blockade of granulocyte macrophage colony-stimulating factor in COVID-19-associated hyperinflammation: chal-lenges and opportunities. The Lancet Respiratory Medicine. 2020; 8: 822–830.

[109] Zemans RL, Matthay MA. What drives neutrophils to the alveoli in ARDS? Thorax. 2017; 72: 1–3.

[110] Havemann K, Gramse M. Physiology and pathophysiology of neutral proteinases of human granulocytes. Advances in Experimental Medicine and Biology. 1984; XIII: 1–20.

[111] Bédard M, McClure CD, Schiller NL, Francoeur C, Cantin A, Denis M. Release of interleukin-8, interleukin-6, and colony-stimulating factors by upper airway epithelial cells: implications for cystic fibrosis. American Journal of Respiratory Cell and Molecular Biology. 1993; 9: 455–462.

[112] Zeiher BG, Matsuoka S, Kawabata K, Repine JE. Neutrophil elastase and acute lung injury: prospects for sivelestat and other neutrophil elastase inhibitors as therapeutics. Critical Care Medicine. 2002; 30: S281–S287.

[113] Hagio T, Nakao S, Matsuoka H, Matsumoto S, Kawabata K, Ohno H. Inhibition of neutrophil elastase activity attenuates complement-mediated lung injury in the hamster. European Journal of Pharmacology. 2001; 426: 131–138.

[114] Hagio T, Matsumoto S, Nakao S, Abiru T, Ohno H, Kawabata K. Elastase inhibition reduced death associated with acid aspiration-induced lung injury in hamsters. European Journal of Pharmacology. 2004; 488: 173–180.

[115] Xiao X-G, Zu H-G, Li Q-G, Huang P. Sivelestat sodium hydrate attenuates acute lung injury by decreasing systemic inflammation in a rat model of severe burns. European Review for Medical and Pharmacological Sciences. 2016; 20: 528–536.

[116] Maki C, Inoue Y, Ishihara T, Hirano Y, Kondo Y, Sueyoshi K, et al. Evaluation of appropriate indications for the use of sivelestat sodium in acute respiratory distress syndrome: a retrospective cohort study. Acute Medicine & Surgery. 2019; 7: e471.

[117] Pu S, Wang D, Liu D, Zhao Y, Qi D, He J, et al. Effect of sivelestat sodium in patients with acute lung injury or acute respiratory distress syndrome: a meta-analysis of randomized controlled trials. BMC Pulmonary Medicine. 2017; 17: 148.

[118] Wang T, Zhu Z, Liu Z, Yi L, Yang Z, Bian W, et al. Plasma neutrophil elastase and elafin as prognostic biomarker for acute respiratory distress syndrome. Shock. 2017; 48: 168–174.

[119] Small DM, Zani M, Quinn DJ, Dallet-Choisy S, Glasgow AM, O’Kane C, et al. A functional variant of elafin with improved anti-inflammatory activity for pulmonary inflammation. Molecular Therapy. 2015; 23: 24–31.

[120] Camprubí-Rimblas M, Tantinyà N, Bringué J, Guillamat-Prats R, Artigas A. Anticoagulant therapy in acute respiratory distress syndrome. Annals of Translational Medicine. 2018; 6: 36.

[121] Juschten J, Tuinman PR, Juffermans NP, Dixon B, Levi M, Schultz MJ. Nebulized anticoagulants in lung injury in critically ill patients—an updated systematic review of preclinical and clinical studies. Annals of Translational Medicine. 2017; 5: 444.

[122] Frantzeskaki F, Armaganidis A, Orfanos SE. Immunothrombosis in acute respiratory distress syndrome: cross talks between inflammation and coagulation. Respiration. 2017; 93: 212–225.

[123] Morris PE, Steingrub JS, Huang BY, Tang S, Liu PM, Rhode PR, et al. A phase I study evaluating the pharmacokinetics, safety and tolerability of an antibody-based tissue factor antagonist in subjects with acute lung injury or acute respiratory distress syndrome. BMC Pulmonary Medicine. 2012; 12: 5.

[124] Abraham E, Reinhart K, Opal S, Demeyer I, Doig C, Rodriguez AL, et al. Efficacy and safety of tifacogin (recombinant tissue factor pathway inhibitor) in severe sepsis. JAMA. 2003; 290: 238.

[125] Wunderink RG, Laterre P, Francois B, Perrotin D, Artigas A, Vidal LO, et al. Recombinant tissue factor pathway inhibitor in severe community-acquired pneumonia. American Journal of Respiratory and Critical Care Medicine. 2011; 183: 1561–1568.

[126] Rezaie AR, Giri H. Antithrombin: an anticoagulant, anti‐inflammatory and antibacterial serpin. Journal of Thrombosis and Haemostasis. 2020; 18: 528–533.

[127] Wiedermann CJ. Clinical review: molecular mechanisms underlying the role of antithrombin in sepsis. Critical Care. 2006; 10: 209.

[128] Warren BL, Eid A, Singer P, Pillay SS, Carl P, Novak I, et al. Caring for the critically ill patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial. JAMA. 2001; 286: 1869–1878.

[129] Eid A, Wiedermann CJ, Kinasewitz GT. Early administration of high-dose antithrombin in severe sepsis: single center results from the kybersept-trial. Anesthesia & Analgesia. 2008; 107: 1633–1638.

[130] Camprubí‐Rimblas M, Tantinyà N, Guillamat‐Prats R, Bringué J, Puig F, Gómez MN, et al. Effects of nebulized antithrombin and heparin on inflammatory and coagulation alterations in an acute lung injury model in rats. Journal of Thrombosis and Haemostasis. 2020; 18: 571–583.

[131] Cornet A, Smit E, Beishuizen A, Groeneveld ABJ. The role of heparin and allied compounds in the treatment of sepsis. Thrombosis and Haemostasis. 2007; 98: 579–586.

[132] Gray E, Hogwood J, Mulloy B. The anticoagulant and antithrombotic mechanisms of heparin. Heparin - a Century of Progress. 2012; 1: 43–61.

[133] Dixon B, Santamaria JD, Campbell DJ. A phase 1 trial of nebulised heparin in acute lung injury. Critical Care. 2008; 12: R64.

[134] Dixon B, Schultz MJ, Hofstra JJ, Campbell DJ, Santamaria JD. Nebulized heparin reduces levels of pulmonary coagulation activation in acute lung injury. Critical Care. 2010; 14: 445.

[135] Dixon B, Schultz MJ, Smith R, Fink JB, Santamaria JD, Campbell DJ. Nebulized heparin is associated with fewer days of mechanical ventilation in critically ill patients: a randomized controlled trial. Critical Care. 2010; 14: R180.

[136] Dixon B, Smith RJ, Campbell DJ, Moran JL, Doig GS, Rechnitzer T, et al. Nebulised heparin for patients with or at risk of acute respiratory distress syndrome: a multicentre, randomised, double-blind, placebo-controlled phase 3 trial. The Lancet Respiratory Medicine. 2021; 9: 360–372.

[137] Glas GJ, Muller J, Binnekade JM, Cleffken B, Colpaert K, Dixon B, et al. HEPBURN - investigating the efficacy and safety of nebulized heparin versus placebo in burn patients with inhalation trauma: study protocol for a multi-center randomized controlled trial. Trials. 2014; 15: 91.

[138] Vincent JL, Ramesh MK, Ernest D, LaRosa SP, Pachl J, Aikawa N, et al. A randomized, double-blind, placebo-controlled, Phase 2b study to evaluate the safety and efficacy of recombinant human soluble thrombomodulin, ART-123, in patients with sepsis and suspected disseminated intravascular coagulation. Critical Care Medicine. 2013; 41: 2069–2079.

[139] Miyoshi S, Ito R, Katayama H, Dote K, Aibiki M, Hamada H, et al. Combination therapy with sivelestat and recombinant human soluble thrombomodulin for ARDS and DIC patients. Drug Design, Development and Therapy. 2014; 8: 1211–1219.

[140] Vincent J, Francois B, Zabolotskikh I, Daga MK, Lascarrou J, Kirov MY, et al. Effect of a recombinant human soluble thrombomodulin on mortality in patients with sepsis-associated coagulopathy. JAMA. 2019; 321: 1993.

[141] François B, Fiancette M, Helms J, Mercier E, Lascarrou J, Kayanoki T, et al. Efficacy and safety of human soluble thrombomodulin (ART-123) for treatment of patients in France with sepsis-associated coagulopathy: post hoc analysis of SCARLET. Annals of Intensive Care. 2021; 11: 53.

[142] Heslet L, Andersen JS, Sengeløv H, Dahlbäck B, Dalsgaard-Nielsen J. Inhalation of activated protein C: a possible new adjunctive intervention in acute respiratory distress syndrome. Biologics. 2007; 1: 465–472.

[143] Ranieri VM, Thompson BT, Barie PS, Dhainaut J, Douglas IS, Finfer S, et al. Drotrecogin alfa (Activated) in adults with septic shock. New England Journal of Medicine. 2012; 366: 2055–2064.

[144] Abdelaal Ahmed Mahmoud A, Mahmoud HE, Mahran MA, Khaled M. Streptokinase versus unfractionated heparin nebulization in patients with severe acute respiratory distress syndrome (ARDS): a randomized controlled trial with observational controls. Journal of Cardiothoracic and Vascular Anesthesia. 2020; 34: 436–443.

[145] Kor DJ, Carter RE, Park PK, Festic E, Banner-Goodspeed VM, Hinds R, et al. Effect of aspirin on development of ards in at-risk patients presenting to the emergency department. JAMA. 2016; 315: 2406.

[146] Bai X, Hippensteel J, Leavitt A, Maloney JP, Beckham D, Garcia C, et al. Hypothesis: alpha-1-antitrypsin is a promising treatment option for COVID-19. Medical Hypotheses. 2021; 146: 110394.

[147] McEvoy NL, Clarke JL, Mc Elvaney OJ, Mc Elvaney OF, Boland F, Hyland D, et al. A randomised, double-blind, placebo-controlled, pilot trial of intravenous plasma purified alpha-1 antitrypsin for SARS-CoV-2-induced acute respiratory distress syndrome: a structured summary of a study protocol for a randomised, controlled trial. Trials. 2021; 22: 288.

[148] Scheraga RG, Southern BD, Grove LM, Olman MA. The role of transient receptor potential vanilloid 4 in pulmonary inflammatory diseases. Frontiers in Immunology. 2017; 8: 503.

[149] Alvarez DF, King JA, Weber D, Addison E, Liedtke W, Townsley MI. Transient receptor potential vanilloid 4-mediated disruption of the alveolar septal barrier. Circulation Research. 2006; 99: 988–995.

[150] Scheraga RG, Abraham S, Grove LM, Southern BD, Crish JF, Perelas A, et al. TRPV4 Protects the Lung from bacterial pneumonia via MAPK molecular pathway switching. The Journal of Immunology. 2020; 204: 1310–1321.

[151] Balakrishna S, Song W, Achanta S, Doran SF, Liu B, Kaelberer MM, et al. TRPV4 inhibition counteracts edema and inflammation and improves pulmonary function and oxygen saturation in chemically induced acute lung injury. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2014; 307: L158–L172.

[152] Abd-Elaziz K, Jesenak M, Vasakova M, Diamant Z. Revisiting matrix metalloproteinase 12: its role in pathophysiology of asthma and related pulmonary diseases. Current Opinion in Pulmonary Medicine. 2021; 27: 54–60.

[153] Jerath A, Ferguson ND, Cuthbertson B. Inhalational volatile-based sedation for COVID-19 pneumonia and ARDS. Intensive Care Medicine. 2020; 46: 1563–1566.

[154] Eckle T, Füllbier L, Wehrmann M, Khoury J, Mittelbronn M, Ibla J, et al. Identification of ectonucleotidases CD39 and CD73 in innate protection during acute lung injury. The Journal of Immunology. 2007; 178: 8127–8137.

[155] Chen C, Penuelas O, Quinn K, Cheng K, Li C, Zhang H, et al. Protective effects of adenosine A2A receptor agonist in ventilator-induced lung injury in rats. Critical Care Medicine. 2009; 37: 2235–2241.

[156] Folkesson HG, Kuzenko SR, Lipson DA, Matthay MA, Simmons MA. The adenosine 2a receptor agonist GW328267C improves lung function after acute lung injury in rats. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2012; 303: L259–L271.

[157] Eckle T, Hughes K, Ehrentraut H, Brodsky KS, Rosenberger P, Choi D, et al. Crosstalk between the equilibrative nucleoside transporter ENT2 and alveolar Adora2b adenosine receptors dampens acute lung injury. The FASEB Journal. 2013; 27: 3078–3089.

[158] Wang W, Chen N-Y, Ren D, Davies J, Philip K, Eltzschig HK, et al. Enhancing extracellular adenosine levels restores barrier function in acute lung injury through expression of focal adhesion proteins. Frontiers in Molecular Biosciences. 2021; 8: 636678.

[159] Coughlin SR. Thrombin signalling and protease-activated receptors. Nature. 2000; 407: 258–264.

[160] Li Z, Yin M, Zhang H, Ni W, Pierce RW, Zhou HJ, et al. BMX represses thrombin-PAR1–mediated endothelial permeability and vascular leakage during early sepsis. Circulation Research. 2020; 126: 471–485.

[161] Natarajan K, Gottipati KR, Berhane K, Samten B, Pendurthi U, Boggaram V. Proteases and oxidant stress control organic dust induction of inflammatory gene expression in lung epithelial cells. Respiratory Research. 2016; 17: 137.

[162] Shebuski RJ, Kilgore KS. Role of inflammatory mediators in thrombo-genesis. Journal of Pharmacology and Experimental Therapeutics. 2002; 300: 729–735.

[163] Okajima K. Regulation of inflammatory responses by activated protein C: the molecular mechanism(s) and therapeutic implications. Clinical Chemistry and Laboratory Medicine (CCLM). 2004; 42: 132–141.

[164] Khoufache K, Berri F, Nacken W, Vogel AB, Delenne M, Camerer E, et al. PAR1 contributes to influenza a virus pathogenicity in mice. Journal of Clinical Investigation. 2013; 123: 206–214.

[165] Jabaudon M, Berthelin P, Pranal T, Roszyk L, Godet T, Faure J, et al. Receptor for advanced glycation end-products and ARDS prediction: a multicentre observational study. Scientific Reports. 2018; 8: 2603.

[166] Uchida T, Shirasawa M, Ware LB, Kojima K, Hata Y, Makita K, et al. Receptor for advanced glycation end-products is a marker of type I cell injury in acute lung injury. American Journal of Respiratory and Critical Care Medicine. 2006; 173: 1008–1015.

[167] Blondonnet R, Audard J, Belville C, Clairefond G, Lutz J, Bouvier D, et al. RAGE inhibition reduces acute lung injury in mice. Scientific Reports. 2017; 7: 7208.

[168] Audard J, Godet T, Blondonnet R, Joffredo J, Paquette B, Belville C, et al. Inhibition of the receptor for advanced glycation end-products in acute respiratory distress syndrome: a randomised laboratory trial in piglets. Scientific Reports. 2019; 9: 9227.

[169] Li J, Wang K, Huang B, Li R, Wang X, Zhang H, et al. The receptor for advanced glycation end products mediates dysfunction of airway epithelial barrier in a lipopolysaccharides-induced murine acute lung injury model. International Immunopharmacology. 2021; 93: 107419.

[170] Remy KE, Cortés-Puch I, Solomon SB, Sun J, Pockros BM, Feng J, et al. Haptoglobin improves shock, lung injury, and survival in canine pneumonia. JCI Insight. 2018; 3: e123013.

[171] Yang F, Haile DJ, Berger FG, Herbert DC, Van Beveren E, Ghio AJ. Haptoglobin reduces lung injury associated with exposure to blood. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2003; 284: L402–L409.

[172] Zheng S, D’Souza VK, Bartis D, Dancer RCA, Parekh D, Naidu B, et al. Lipoxin A4 promotes lung epithelial repair whilst inhibiting fibroblast proliferation. ERJ Open Research. 2016; 2: 00079–02015.

[173] Mei HX, Ye Y, Xu HR, Xiang SY, Yang Q, Ma HY, et al. LXA4 inhibits lipopolysaccharide-induced inflammatory cell accumulation by resident macrophages in mice. Journal of Inflammation Research. 2021; 14: 1375–1385.

[174] Yang J, Li M, Chen X, Lian Q, Wang Q, Gao F, et al. Lipoxin a4 ameliorates lipopolysaccharide-induced lung injury through stimulating epithelial proliferation, reducing epithelial cell apoptosis and inhibits epithelial-mesenchymal transition. Respiratory Research. 2019; 20: 192.

[175] Zhang HW, Wang Q, Mei HX, Zheng SX, Ali AM, Wu QX, et al. RvD1 ameliorates LPS-induced acute lung injury via the suppression of neutrophil infiltration by reducing CXCL2 expression and release from resident alveolar macrophages. International Immunopharmacology. 2019; 76: 105877.

[176] Liao Z, Dong J, Wu W, Yang T, Wang T, Guo L, et al. Resolvin D1 attenuates inflammation in lipopolysaccharide-induced acute lung injury through a process involving the PPARγ/NF-κB pathway. Respiratory Research. 2012; 13: 110.

[177] Yang Q, Xu HR, Xiang SY, Zhang C, Ye Y, Shen CX, et al. Resolvin conjugates in tissue regeneration 1 promote alveolar fluid clearance by activating alveolar epithelial sodium channels and Na, K-ATPase in lipopolysaccharide-induced acute lung injury. The Journal of Pharmacology and Experimental Therapeutics. 2021; 379: 156–165.

[178] Heijnen NFL, Bergmans DCJJ, Schultz MJ, Bos LDJ. Precision medicine in Acute Respiratory Distress Syndrome. Signa Vitae. 2021. (in press)

[179] Zhai R, Bonda WLM, Matute-Bello G, Jabaudon M. From preclinical to clinical models of acute respiratory distress syndrome. Signa Vitae. 2022; 18; 3–14.


Abstracted / indexed in

Science Citation Index Expanded (SciSearch) Created as SCI in 1964, Science Citation Index Expanded now indexes over 9,200 of the world’s most impactful journals across 178 scientific disciplines. More than 53 million records and 1.18 billion cited references date back from 1900 to present.

Journal Citation Reports/Science Edition Journal Citation Reports/Science Edition aims to evaluate a journal’s value from multiple perspectives including the journal impact factor, descriptive data about a journal’s open access content as well as contributing authors, and provide readers a transparent and publisher-neutral data & statistics information about the journal.

Chemical Abstracts Service Source Index The CAS Source Index (CASSI) Search Tool is an online resource that can quickly identify or confirm journal titles and abbreviations for publications indexed by CAS since 1907, including serial and non-serial scientific and technical publications.

Index Copernicus The Index Copernicus International (ICI) Journals database’s is an international indexation database of scientific journals. It covered international scientific journals which divided into general information, contents of individual issues, detailed bibliography (references) sections for every publication, as well as full texts of publications in the form of attached files (optional). For now, there are more than 58,000 scientific journals registered at ICI.

Geneva Foundation for Medical Education and Research The Geneva Foundation for Medical Education and Research (GFMER) is a non-profit organization established in 2002 and it works in close collaboration with the World Health Organization (WHO). The overall objectives of the Foundation are to promote and develop health education and research programs.

Scopus: CiteScore 1.0 (2022) Scopus is Elsevier's abstract and citation database launched in 2004. Scopus covers nearly 36,377 titles (22,794 active titles and 13,583 Inactive titles) from approximately 11,678 publishers, of which 34,346 are peer-reviewed journals in top-level subject fields: life sciences, social sciences, physical sciences and health sciences.

Embase Embase (often styled EMBASE for Excerpta Medica dataBASE), produced by Elsevier, is a biomedical and pharmacological database of published literature designed to support information managers and pharmacovigilance in complying with the regulatory requirements of a licensed drug.

Submission Turnaround Time

Conferences

Top