135
Views
35
CrossRef citations to date
0
Altmetric
Review

Cytokine Storm and Mucus Hypersecretion in COVID-19: Review of Mechanisms

, , , , ORCID Icon, ORCID Icon, & show all
Pages 175-189 | Published online: 22 Jan 2021

References

  • Tian S, Hu W, Niu L, Liu H, Xu H, Xiao SY. Pulmonary pathology of early Phase 2019 novel coronavirus (COVID-19) pneumonia in two patients with lung cancer. J Thorac Oncol. 2020;15:700–704. doi:10.1016/j.jtho.2020.02.010
  • Bernheim A, Mei X, Huang M. Chest CT findings in coronavirus disease-19 (COVID-19): relationship to duration of infection. Radiology. 2020;295.
  • Khashkhosha HK, Elhadi M. A hypothesis on the role of the human immune system in covid-19 published online ahead of print, [2020 Jul 1]. Med Hypotheses. 2020;143:110066. doi:10.1016/j.mehy.2020.110066
  • Earhart AP, Holliday ZM, Hofmann HV, Schrum AG. Consideration of dornase alfa for the treatment of severe COVID-19 acute respiratory distress syndrome. New Microbes New Infect. 2020;35:100689. doi:10.1016/j.nmni.2020.100689
  • Zhu N, Zhang D, Wang W, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med. 2020;382(8):727–733. doi:10.1056/NEJMoa2001017
  • Costela-Ruiz VJ, Illescas-Montes R, Puerta-Puerta JM, Ruiz C, Melguizo-Rodríguez L. SARS-CoV-2 infection: the role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev. 2020;54:62–75. doi:10.1016/j.cytogfr.2020.06.001
  • Jiang Y, Xu J, Zhou C, et al. Characterization of cytokine/chemokine profiles of severe acute respiratory syndrome. Am J Respir Crit Care Med. 2005;171(8):850–857. doi:10.1164/rccm.200407-857OC
  • Upadhyay J, Tiwari N, Ansari MN. Role of inflammatory markers in corona virus disease (COVID-19) patients: a review. Exp Biol Med. 2020;245(15):1368–1375. doi:10.1177/1535370220939477
  • Ye Z, Zhang Y, Wang Y, et al. Chest CT manifestations of new coronavirus disease 2019 (COVID-19): a pictorial review. Eur Radiol. 2020;1–9.
  • Li X, Ma X. Acute respiratory failure in COVID-19: is it “typical” ARDS? Crit Care. 2020;24(1):198. doi:10.1186/s13054-020-02911-9
  • Holtzman MJ, Battaile JT, Patel AC. Immunogenetic programs for viral induction of mucous cell metaplasia. Am J Respir Cell Mol Biol. 2006;35(1):29–39. doi:10.1165/rcmb.2006-0092SF
  • Vetrugno L, Baciarello M, Bignami E, et al. The “pandemic” increase in lung ultrasound use in response to Covid-19: can we complement computed tomography findings? A narrative review. Ultrasound J. 2020;12:39. doi:10.1186/s13089-020-00185-4
  • Lai SK, Wang YY, Wirtz D, Hanes J. Micro- and macrorheology of mucus. Adv Drug Deliv Rev. 2009;61(2):86–100. doi:10.1016/j.addr.2008.09.012
  • Shukla SD, Mahmood MQ, Weston Set al. The main rhinovirus respiratory tract adhesion site (ICAM-1) is upregulated in smokers and patients with chronic airflow limitation (CAL). Respir Res2017;18(1):6 doi:10.1186/s12931-016-0483-8
  • Ohar JA, Donohue JF, Spangenthal S. The role of guaifenesin in the management of chronic mucus hypersecretion associated with stable chronic bronchitis: a comprehensive review. Chronic Obstr Pulm Dis. 2019;6(4):341–349.
  • Hodges RR, Dartt DA. Conjunctival goblet cells. Encyclopedia Eye. 2010;369–376.
  • Knoop KA, Newberry RD. Goblet cells: multifaceted players in immunity at mucosal surfaces. Mucosal Immunol. 2018;11:1551–1557. doi:10.1038/s41385-018-0039-y
  • Zanin M, Baviskar P, Webster R, Webby R. The interaction between respiratory pathogens and mucus. Cell Host Microbe. 2016;19(2):159–168. doi:10.1016/j.chom.2016.01.001
  • Adler KB, Tuvim MJ, Dickey BF. Regulated mucin secretion from airway epithelial cells. Front Endocrinol. 2013;4:129. doi:10.3389/fendo.2013.00129
  • Chen C, Thai P, Yoneda K, et al. A peptide that inhibits the function of Myristoylated Alanine-Rich C Kinase Substrate (MARCKS) reduces lung cancer metastasis. Oncogene. 2014;33:3696–3706. doi:10.1038/onc.2013.336
  • Wickström C, Davies JR, Eriksen GV, et al. MUC5B is a major gel-forming, oligomeric mucin from human salivary gland, respiratory tract and endocervix: identification of glycoforms and C-terminal cleavage. Biochem J. 1998;15(334):685–693. doi:10.1042/bj3340685
  • Hovenberg HW, Davies JR. Carlstedt.Different mucins are produced by the surface epithelium and the submucosa in human trachea: identification of MUC5AC as a major mucin from the goblet cells. I Biochem J. 1996;15(318):319–324. doi:10.1042/bj3180319
  • Dabbagh K, Takeyama K, Lee HM, et al. IL-4 induces mucin gene expression and goblet cell metaplasia in vitro and in vivo. J Immunol. 1999;162:6233–6237.
  • Xiang J, Rir-Sim-Ah J, Tesfaigzi Y. IL-9 and IL-13 induce mucous cell metaplasia that is reduced by IFN-gamma in a Bax-mediated pathway. Am J Respir Cell Mol Biol. 2008;38(3):310–317. doi:10.1165/rcmb.2007-0078OC
  • Cerveri I, Brusasco V. Revisited role for mucus hypersecretion in the pathogenesis of COPD. Eur Respir Rev. 2010;19(116):109–112. doi:10.1183/09059180.00002710
  • Wen FQ, Shen YC. Expectorant therapy revisited in chronic obstructive pulmonary disease. Zhonghua Jie He He Hu Xi Za Zhi. 2011;34(4):243–245.
  • Jartti T, Bønnelykke K, Elenius V, Feleszko W. Role of viruses in asthma. Semin Immunopathol. 2020;42(1):61–74. doi:10.1007/s00281-020-00781-5
  • Grunstein MM, Hakonarson H, Maskeri N, et al. Autocrine cytokine signaling mediates effects of rhinovirus on airway responsiveness. Am J Physiol. 2000;6:1146–L1153.
  • Baños-Lara Mdel R, Piao B, Guerrero-Plata A. Differential mucin expression by respiratory syncytial virus and human metapneumovirus infection in human epithelial cells. Mediators Inflamm. 2015;2015:347292.
  • Persson BD, Jaffe AB, Fearns R, Danahay H. Respiratory syncytial virus can infect basal cells and alter human airway epithelial differentiation. PLoS One. 2014;9(7):e102368. doi:10.1371/journal.pone.0102368
  • Stokes KL, Currier MG, Sakamoto K, et al. The respiratory syncytial virus fusion protein and neutrophils mediate the airway mucin response to pathogenic respiratory syncytial virus infection. J Virol. 2013;87(18):10070–10082. doi:10.1128/JVI.01347-13
  • Farooqi FI, Morgan RC, Dhawan N, Dinh J, Yatzkan G, Michel G. Airway hygiene in COVID-19 pneumonia: treatment responses of 3 critically Ill cruise ship employees. Am J Case Rep. 2020;21:e926596. doi:10.12659/AJCR.926596
  • Sungnak W, Huang N, Bécavin C, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020;26:681–687. doi:10.1038/s41591-020-0868-6
  • Bustamante-Marin XM, Ostrowski LE. Cilia and mucociliary clearance. Cold Spring Harb Perspect Biol. 2017;9(4):a028241. doi:10.1101/cshperspect.a028241
  • Arumugham V Immunological mechanisms explaining the role of IgE, mast cells, histamine, elevating ferritin, IL-6, D-dimer, VEGF levels in COVID-19 and dengue, potential treatments such as mast cell stabilizers, antihistamines, Vitamin C, hydroxychloroquine, ivermectin and azithromycin. 2020.
  • Prompetchara E, Ketloy C, Palaga T. Immune responses in COVID-19 and potential vaccines: lessons learned from SARS and MERS epidemic. Asian Pac J Allergy Immunol.
  • Cohn L, Homer RJ, Marinov A, Rankin J, Bottomly K. Induction of airway mucus production By T helper 2 (Th2) cells: a critical role for interleukin 4 in cell recruitment but not mucus production. J Exp Med. 1997;186(10):1737–1747. doi:10.1084/jem.186.10.1737
  • Thai P, Chen Y, Dolganov G, Wu R. Differential regulation of MUC5AC/Muc5ac and hCLCA-1/mGob-5 expression in airway epithelium. Am J Respir Cell Mol Biol. 2005;33(6):523–530. doi:10.1165/rcmb.2004-0220RC
  • Kreda SM, Davis CW, Rose MC. CFTR, mucins, and mucus obstruction in cystic fibrosis. Cold Spring Harb Perspect Med. 2012;2(9):a009589. doi:10.1101/cshperspect.a009589
  • Saint-Criq V, Gray MA. Role of CFTR in epithelial physiology. Cell Mol Life Sci. 2017;74(1):93–115.
  • Almughem FA, Aldossary AM, Tawfik EA, et al. Cystic fibrosis: overview of the current development trends and innovative therapeutic strategies. Pharmaceutics. 2020;12(7):616.
  • Jarosz-Griffiths HH, Scambler T, Wong CH, et al. Different CFTR modulator combinations downregulate inflammation differently in cystic fibrosis. Elife. 2020;9:e54556. doi:10.7554/eLife.54556
  • Griffin DO, Jensen A, Khan M, et al. Pulmonary embolism and increased levels of d-dimer in patients with coronavirus disease. Emerg Infect Dis. 2020;26(8):1941–1943. doi:10.3201/eid2608.201477
  • Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China [published correction appears in Lancet. 2020 Jan 30]. Lancet. 2020;395(10223):497–506. doi:10.1016/S0140-6736(20)30183-5
  • Liu K, Fang YY, Deng Y, et al. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei Province. Chin Med J (Engl). 2020;133(9):1025–1031. doi:10.1097/CM9.0000000000000744
  • Wang W, Liu X, Wu S, et al. The definition and risks of cytokine release syndrome in 11 COVID-19-affected critically ill patients with pneumonia: analysis of disease characteristics. J Infect Dis. 2020;jiaa387.
  • Allinson JP, Hardy R, Donaldson GC, et al. The presence of chronic mucus hypersecretion across adult life in relation to chronic obstructive pulmonary disease development. Am J Respir Crit Care Med. 2016;193(6):662–667. doi:10.1164/rccm.201511-2210OC
  • Busse PJ, Zhang TF. Kamal, et al. Chronic exposure to TNF-alpha increases airway mucus gene expression in vivo. J Allergy Clin Immunol. 2005; 116(6):1256–63.
  • Girija ASS, Shankar EM, Larsson M. Could SARS-CoV-2-induced hyperinflammation magnify the severity of Coronavirus Disease (CoViD-19) leading to acute respiratory distress syndrome? Front Immunol. 2020;11:1206. doi:10.3389/fimmu.2020.01206
  • Xia Y, Cai P, Yu F, et al. IL-4-induced caveolin-1-containing lipid rafts aggregation contributes to MUC5AC synthesis in bronchial epithelial cells. Respir Res. 2017;18:174. doi:10.1186/s12931-017-0657-z
  • Gharavi NM, Alva JA, Kevin P, et al. Role of the JAK/STAT pathway in the regulation of IL-8 transcription by oxidized phospholipids in vitro and in atherosclerosis in vivo. J Biol Chem. 2007;282(43):31460–31468. doi:10.1074/jbc.M704267200
  • Yu H, Li Q, Kolosov VP, et al. Interleukin-13 induces mucin 5AC production involving STAT6/SPDEF in human airway epithelial cells. Cell Commun Adhes. 2010;17(4–6):83–92. doi:10.3109/15419061.2010.551682
  • Grunig G, Warnock M, Wakil AE, et al. Requirement for IL-13 independently of IL-4 in experimental asthma. Science. 1998;282:2261–2264. doi:10.1126/science.282.5397.2261
  • Kelly-Welch AE, Hanson EM, Boothby MR, et al. Interleukin-4 and interleukin-13 signalling connection maps. Science. 2003;300:1527–1528. doi:10.1126/science.1085458
  • Seibold MA. Interleukin-13 stimulation reveals the cellular and functional plasticity of the airway epithelium. Ann Am Thorac Soc. 2018;15(2):S98–S102.
  • Nakanishi K. Unique action of Interleukin-18 on T cells and other immune cells. Front Immunol. 2018. 9:763. doi:10.3389/fimmu.2018.00763
  • Goswami R, Kaplan MH. A brief history of IL-9. J Immunol. 2011;186(6):3283–3288. doi:10.4049/jimmunol.1003049
  • Demoulin JB, Uyttenhove C, Van Roost E, et al. A single tyrosine of the interleukin-9 (IL-9) receptor is required for STAT activation, antiapoptotic activity, and growth regulation by IL-9. Mol Cell Biol. 1996;16(9):4710–4716.
  • Fung M, Chu Y, Fink J, et al. IL-2- and STAT5-regulated cytokine gene expression in cells expressing the Tax protein of HTLV-1. Oncogene. 2005;24:4624–4633. doi:10.1038/sj.onc.1208507
  • Wilson MS, Pesce JT, Ramalingam TR, et al. Suppression of murine allergic airway disease by IL-2: anti-IL-2monoclonal antibody-induced regulatory T cells. J Immunol. 2008;181(10):6942–6954. doi:10.4049/jimmunol.181.10.6942
  • Bao L, Zhang H, Chan LS. The involvement of the JAK-STAT signaling pathway in chronic inflammatory skin disease atopic dermatitis. JAKSTAT. 2013;2(3):e24137.
  • Li L, Zhao GD, Shi Z, Qi LL, Zhou LY, Fu ZX. The Ras/Raf/MEK/ERK signaling pathway and its role in the occurrence and development of HCC. Oncol Lett. 2016;12(5):3045–3050. doi:10.3892/ol.2016.5110
  • Paunovic V, Harnett MM. Mitogen-activated protein kinases as therapeutic targets for rheumatoid arthritis. Drugs. 2013;73(2):101–115.
  • Lillehoj EP, Kato K, Lu W, Kim KC. Cellular and molecular biology of airway mucins. Int Rev Cell Mol Biol. 2013;303:139–202.
  • Burgos-Blasco B, Güemes-Villahoz N, Santiago JL, et al. Hypercytokinemia in COVID-19: tear cytokine profile in hospitalized COVID-19 patients. Exp Eye Res. 2020;200:108253.
  • Lee JJ, McGarry MP, Farmer SC, et al. Interleukin-5 expression in the lung epithelium of transgenic mice leads to pulmonary changes pathognomonic of asthma. J Exp Med. 1997;185(12):2143–2156. doi:10.1084/jem.185.12.2143
  • Tesmer LA, Lundy SK, Sarkar S, Fox DA. Th17 cells in human disease. Immunol Rev. 2008;223:87–113.
  • Hynes GM, Hinks TSC. The role of interleukin-17 in asthma: a protective response? ERJ Open Res. 2020;6(2):00364–2019. doi:10.1183/23120541.00364-2019
  • Coperchini F, Chiovato L, Croce L, Magri F, Rotondi M. The cytokine storm in COVID-19: an overview of the involvement of the chemokine/chemokine-receptor system. Cytokine Growth Factor Rev. 2020;53:25–32. doi:10.1016/j.cytogfr.2020.05.003
  • Zhang D, Facchinetti V, Wang X, Huang Q, Qin J, Su B. Identification of MEKK2/3 serine phosphorylation site targeted by the Toll-like receptor and stress pathways. EMBO J. 2006;25(1):97–107.
  • Chen Y, Garvin LM, Nickola TJ, Watson AM, Colberg-Poley AM, Rose MC. IL-1β induction of MUC5AC gene expression is mediated by CREB and NF-κB and repressed by dexamethasone. Am J Physiol Lung Cell Mol Physiol. 2014;306(8):L797–807. doi:10.1152/ajplung.00347.2013
  • Price MM, Oskeritzian CA, Falanga YT, et al. A specific sphingosine kinase 1 inhibitor attenuates airway hyperresponsiveness and inflammation in a mast cell-dependent murine model of allergic asthma. J Allergy Clin Immunol. 2013;131(2):501–11.e1. doi:10.1016/j.jaci.2012.07.014
  • Kankaanranta H, Ilmarinen P, Zhang X, et al. Tumour necrosis factor-α regulates human eosinophil apoptosis via ligation of TNF-receptor 1 and balance between NF-κB and AP-1. PLoS One. 2014;9(2):e90298. doi:10.1371/journal.pone.0090298
  • Wang X, Lin Y. Tumor necrosis factor and cancer, buddies or foes? Acta Pharmacol Sin. 2008;29(11):1275–1288. doi:10.1111/j.1745-7254.2008.00889.x
  • Wang IJ, Wu CY, Hu FR. Effect of proinflammatory cytokines on the human MUC5AC promoter activity in vitro and in vivo. Clin Ophthalmol. 2007;1(1):71–77.
  • Thai P, Loukoianov A, Wachi S, Wu R. Regulation of airway mucin gene expression. Annu Rev Physiol. 2008;70:405–429. doi:10.1146/annurev.physiol.70.113006.100441
  • Shishikura Y, Koarai A, Aizawa H, et al. Extracellular ATP is involved in dsRNA-induced MUC5AC production via P2Y2R in human airway epithelium. Respir Res. 2016;17(1):121. doi:10.1186/s12931-016-0438-0
  • Xue L, Barrow A, Fleming VM, et al. Leukotriene E4 activates human Th2 cells for exaggerated proinflammatory cytokine production in response to prostaglandin D2. J Immunol. 2012;188(2):694–702.
  • Huber M. Activation/Inhibition of mast cells by supra-optimal antigen concentrations. Cell Commun Signal. 2013;11(1):7. doi:10.1186/1478-811X-11-7
  • Rådmark O, Samuelsson B. 5-Lipoxygenase: mechanisms of regulation. J Lipid Res. 2009;50 Suppl(Suppl):S40–5.
  • Hedi H, Norbert G. 5-lipoxygenase pathway, dendritic cells, and adaptive immunity. J Biomed Biotechnol. 2004;2004(2):99–105. doi:10.1155/S1110724304310041
  • Leckie MJ, Ten Brinke A, Khan J, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;35(6):2144–2148. doi:10.1016/S0140-6736(00)03496-6
  • Deeks ED, Brusselle G. Reslizumab in eosinophilic asthma: a review. Drugs. 2017;77:777–784.
  • Brightling CE, Bleecker ER, Panettieri RA Jr, et al. Benralizumab for chronic obstructive pulmonary disease and sputum eosinophilia: a randomised, double-blind, placebo-controlled, phase 2 study. Lancet Respir Med. 2014;2:891–901. doi:10.1016/S2213-2600(14)70187-0
  • Arora S, Ahmad S, Irshad R, et al. TLRs in pulmonary diseases. Life Sci. 2019;15(233):116671. doi:10.1016/j.lfs.2019.116671
  • Wang L, Quan Y, Yue Y, Heng X, Che F. Interleukin-37: a crucial cytokine with multiple roles in disease and potentially clinical therapy. Oncol Lett. 2018;15(4):4711–4719.
  • Zhang L, Zhang J, Gao P. The potential of interleukin-37 as an effective therapeutic agent in asthma. Respir Res. 2017;18(1):192. doi:10.1186/s12931-017-0675-x
  • Nold MF, Nold-Petry CA. Zepp, et al. IL-37 is a fundamental inhibitor of innate immunity. Nat Immunol. 2010;11:1014–1022. doi:10.1038/ni.1944
  • Nold-Petry CA, Lo CY, Rudloff I, et al. IL-37 requires the receptors IL-18Ralpha and IL-1R8 (SIGIRR) to carry out its multifaceted anti-inflammatory program upon innate signal transduction. Nat Immunol. 2015;16:354–365. doi:10.1038/ni.3103
  • Abdalla AE, Li Q, Xie L, Xie J. Biology of IL-27 and its role in the host immunity against Mycobacterium tuberculosis. Int J Biol Sci. 2015;11(2):168–175. doi:10.7150/ijbs.10464
  • Bosmann M, Ward PA. Modulation of inflammation by interleukin-27. J Leukoc Biol. 2013;94(6):1159–1165. doi:10.1189/jlb.0213107
  • de Almeida Nagata DE, Demoor T, Ptaschinski C, et al. IL-27R-mediated regulation of IL-17 controls the development of respiratory syncytial virus-associated pathogenesis. Am J Pathol. 2014;184(6):1807–1818. doi:10.1016/j.ajpath.2014.02.004
  • Ito T, Tanaka T, Nakamaru K, et al. Interleukin-35 promotes the differentiation of regulatory T cells and suppresses Th2 response in IgG4-related type 1 autoimmune pancreatitis. J Gastroenterol. 2020;55(8):789–799. doi:10.1007/s00535-020-01689-5
  • Huang A, Cheng L, He M, Nie J, Wang J, Jiang K. Interleukin-35 on B cell and T cell induction and regulation. J Inflamm (Lond). 2017;14(16). doi:10.1186/s12950-017-0164-5
  • Heinrich PC, Behrmann I, Müller-Newen G, Schaper F, Graeve L. Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway. Biochem J. 1998;334(Pt 2):297–314. doi:10.1042/bj3340297
  • Seelaender M, Neto JC, Pimentel GD, Goldszmid RS, Lira FS. Inflammation in the disease: mechanism and therapies 2014. Mediators Inflamm. 2015;2015:169852. doi:10.1155/2015/169852
  • Van de Veerdonk FL, Stoeckman AK, Wu G, et al. IL-38 binds to the IL-36 receptor and has biological effects on immune cells similar to IL-36 receptor antagonist. Proc Natl Acad Sci U S A. 2012;109(8):3001–3005. doi:10.1073/pnas.1121534109
  • Pavord ID, Chanez P, Criner GJ, et al. Mepolizumab for Eosinophilic Chronic Obstructive Pulmonary Disease. N Engl J Med. 2017;377:1613–1629.
  • Queen D, Ediriweera C, Liu L. Function and regulation of IL-36 signaling in inflammatory diseases and cancer development. Front Cell Dev Biol. 2019;7:317. doi:10.3389/fcell.2019.00317
  • Boutet MA, Nerviani A, Pitzalis C. IL-36, IL-37, and IL-38 cytokines in skin and joint inflammation: a comprehensive review of their therapeutic potential. Int J Mol Sci. 2019;20(6):1257. doi:10.3390/ijms20061257
  • Madonna S, Girolomoni G, Dinarello CA, Albanesi C. The significance of IL-36 hyperactivation and IL-36R targeting in psoriasis. Int J Mol Sci. 2019;20(13):3318. doi:10.3390/ijms20133318
  • Bassoy EY, Towne JE, Gabay C. Regulation and function of interleukin-36 cytokines. Immunol Rev. 2018;281(1):169–178. doi:10.1111/imr.12610
  • Atal S, Fatima Z. IL-6 inhibitors in the treatment of serious COVID-19: a promising therapy? Pharmaceut Med. 2020;34(4):223–231.
  • Goldbach-Mansky R. Blocking interleukin-1 in rheumatic diseases. Ann N Y Acad Sci. 2009;1182(1):111–123. doi:10.1111/j.1749-6632.2009.05159.x
  • Sciascia S, Aprà F, Baffa A, et al. Pilot prospective open, single-arm multicentre study on off-label use of tocilizumab in patients with severe COVID-19. Clin Exp Rheumatol. 2020;38(3):529–532.
  • Russell B, Moss C, George G, et al. Associations between immune-suppressive and stimulating drugs and novel COVID-19-a systematic review of current evidence. Ecancermedicalscience. 2020;14:1022. doi:10.3332/ecancer.2020.1022
  • Kim JH, Gupta SC, Park B, Yadav VR, Aggarwal BB. Turmeric (Curcuma longa) inhibits inflammatory nuclear factor (NF)-κB and NF-κB-regulated gene products and induces death receptors leading to suppressed proliferation, induced chemosensitization, and suppressed osteoclastogenesis. Mol Nutr Food Res. 2012;56(3):454–465. doi:10.1002/mnfr.201100270
  • Ghandadi M, Sahebkar A. Curcumin: an effective inhibitor of interleukin-6. Curr Pharm Des. 2017;23(6):921–931. doi:10.2174/1381612822666161006151605
  • Mao QQ, Xu XY, Cao SY, et al. Bioactive compounds and bioactivities of ginger (Zingiber officinale Roscoe). Foods. 2019;8(6):185. doi:10.3390/foods8060185
  • Ohishi T, Goto S, Monira P, Isemura M, Nakamura Y. Anti-inflammatory action of green tea. Antiinflamm Antiallergy Agents Med Chem. 2016;15(2):74–90. doi:10.2174/1871523015666160915154443
  • Cabrita I, Benedetto R, Schreiber R, Kunzelmann K. Niclosamide repurposed for the treatment of inflammatory airway disease. JCI Insight. 2019;4(15):e128414. doi:10.1172/jci.insight.128414
  • Tufts Medical Center. Niclosamide for mild to moderate COVID-19. NLM identifier: NCT04399356. Available from: https://clinicaltrials.gov/ct2/show/NCT04399356. Accessed September 1, 2020.
  • Miklossy G, Hilliard TS, Turkson J. Therapeutic modulators of STAT signalling for human diseases. Nat Rev Drug Discov. 2013;12(8):611–629. doi:10.1038/nrd4088
  • Yale University. Tofacitinib for Treatment of Moderate COVID-19 (I-TOMIC). NLM identifier: NCT04415151. Available from: https://clinicaltrials.gov/ct2/show/NCT04415151. Accessed July 29, 2020..
  • Gerriets V, Bansal P, Goyal A, et al. Tumor Necrosis Factor (TNF) Inhibitors. [Updated 2020 Jul 4]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2020 Jan. Available from: https://www.ncbi.nlm.nih.gov/books/NBK482425/. Accessed December 11, 2020.
  • Tufts Medical Center. A Phase 2 trial of infliximab in coronavirus disease 2019 (COVID-19). NLM identifier: NCT04425538. Available from: https://clinicaltrials.gov/ct2/show/NCT04425538. Accessed June 1, 2020.
  • Choy EH, De Benedetti F, Takeuchi T, Hashizume M, John MR, Kishimoto T. Translating IL-6 biology into effective treatments. Nat Rev Rheumatol. 2020;16(6):335–345. doi:10.1038/s41584-020-0419-z
  • Genentech, Inc. A study to evaluate the efficacy and safety of tocilizumab in hospitalized participants with COVID-19 pneumonia. NLM identifier: NCT04372186. Available from: https://clinicaltrials.gov/ct2/show/NCT04372186. Accessed May 14, 2020.
  • Wu KK, Dao H Jr. Off-label dermatologic uses of IL-17 inhibitors [published online ahead of print, 2020 Mar 9]. J Dermatolog Treat. 2020;1–7. doi:10.1080/09546634.2020.1737638
  • Lomonosov Moscow State University Medical Research and Educational Center. COLchicine versus ruxolitinib and secukinumab in open prospective randomized trial (COLORIT). NLM identifier: NCT04403243. Available from: https://clinicaltrials.gov/ct2/show/NCT04403243. Accessed May 8, 2020.
  • Fenini G, Contassot E, French LE. Potential of IL-1, IL-18 and inflammasome inhibition for the treatment of inflammatory skin diseases. Front Pharmacol. 2017;8:278. doi:10.3389/fphar.2017.00278
  • University Hospital, Basel, Switzerland. Canakinumab in patients with COVID-19 and Type 2 diabetes (CanCovDia). NLM identifier: NCT04510493. Available from: https://clinicaltrials.gov/ct2/show/NCT04510493. Accessed September 2020.