228
Views
10
CrossRef citations to date
0
Altmetric
Original Research

The Effect of Nintedanib on T-Cell Activation, Subsets and Functions

ORCID Icon, &
Pages 997-1011 | Published online: 08 Mar 2021

References

  • LuzinaIG, ToddNW, IaconoAT, AtamasSP. Roles of T lymphocytes in pulmonary fibrosis. J Leukoc Biol. 2008;83(2):237–244. doi:10.1189/jlb.070750417962367
  • PerrosF, DorfmullerP, MontaniD, et al. Pulmonary lymphoid neogenesis in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2012;185(3):311–321. doi:10.1164/rccm.201105-0927OC22108206
  • Marchal-Sommé J, UzunhanY, Marchand-AdamS, et al. Cutting edge: nonproliferating mature immune cells form a novel type of organized lymphoid structure in idiopathic pulmonary fibrosis. J Immunol. 2006;176(10):5735–5739. doi:10.4049/jimmunol.176.10.573516670278
  • WellsAU, LorimerS, MajumdarS, et al. Fibrosing alveolitis in systemic sclerosis: increase in memory T-cells in lung interstitium. Eur Respir J. 1995;8(2):266–271. doi:10.1183/09031936.95.080202667758562
  • PoppW, RitschkaL, ScherakO, et al. Bronchoalveolar lavage in rheumatoid arthritis and secondary Sjögren’s syndrome. Lung. 1990;168(4):221–231. doi:10.1007/BF02719696
  • FischerA, Du BoisR. du Bois R. Interstitial lung disease in connective tissue disorders. Lancet. 2012;380(9842):689–698. doi:10.1016/S0140-6736(12)61079-422901890
  • Fernández PérezER, BrownKK. Fibrotic hypersensitivity pneumonitis. Curr Respir Care Rep. 2014;3(4):170–178. doi:10.1007/s13665-014-0094-0
  • HeukelsP, MoorCC, von der ThusenJH, WijsenbeekMS, KoolM. Inflammation and immunity in IPF pathogenesis and treatment. Respir Med. 2019;25:79–91. doi:10.1016/j.rmed.2018.12.015
  • O’ReillyS, HugleT, van LaarJM. T cells in systemic sclerosis: a reappraisal. Rheumatology. 2012;51(9):1540–1549. doi:10.1093/rheumatology/kes09022577083
  • CopeAP, Schulze-KoopsH, AringerM. The central role of T cells in rheumatoid arthritis. Clin Exp Rheumatol. 2007;25(5 Suppl 46):S4–11.17977483
  • BarreraL, MendozaF, ZunigaJ, et al. Functional diversity of T-cell subpopulations in subacute and chronic hypersensitivity pneumonitis. Am J Respir Crit Care Med. 2008;177(1):44–55. doi:10.1164/rccm.200701-093OC17947613
  • TanjoreH, XuXC, PolosukhinVV, et al. Contribution of epithelial-derived fibroblasts to bleomycin-induced lung fibrosis. Am J Respir Crit Care Med. 2009;180(7):657–665. doi:10.1164/rccm.200903-0322OC19556518
  • SpagnoloP, RossiG, CavazzaA, et al. Hypersensitivity pneumonitis: a comprehensive review. J Investig Allergology Clin Immunol. 2015;25(4):237–250.
  • ZhangM, ZhangS. T cells in fibrosis and fibrotic diseases. Front Immunol. 2020;11:1142. doi:10.3389/fimmu.2020.0114232676074
  • HilbergF, RothGJ, KrssakM, et al. BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res. 2008;68(12):4774–4782. doi:10.1158/0008-5472.CAN-07-630718559524
  • HilbergF, Tontsch-GruntU, BaumA, et al. Triple angiokinase inhibitor nintedanib directly inhibits tumor cell growth and induces tumor shrinkage via blocking oncogenic receptor tyrosine kinases. J Pharmacol Exp Ther. 2018;364(3):494–503. doi:10.1124/jpet.117.24412929263244
  • WollinL, WexE, PautschA, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45(5):1434–1445. doi:10.1183/09031936.0017491425745043
  • RicheldiL. du Bois RM, Raghu G, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014;370(22):2071–2082. doi:10.1056/NEJMoa140258424836310
  • DistlerO, HighlandKB, GahlemannM, et al. Nintedanib for systemic sclerosis–associated interstitial lung disease. N Engl J Med. 2019;380(26):2518–2528. doi:10.1056/NEJMoa190307631112379
  • FlahertyKR, WellsAU, CottinV, et al. Nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med. 2019;381(18):1718–1727. doi:10.1056/NEJMoa190868131566307
  • HostettlerKE, ZhongJ, PapakonstantinouE, et al. Anti-fibrotic effects of nintedanib in lung fibroblasts derived from patients with idiopathic pulmonary fibrosis. Respir Res. 2014;15(1):157. doi:10.1186/s12931-014-0157-325496490
  • WollinL, MailletI, QuesniauxV, HolwegA, RyffelB. Antifibrotic and anti-inflammatory activity of the tyrosine kinase inhibitor nintedanib in experimental models of lung fibrosis. J Pharmacol Exp Ther. 2014;349(2):209–220. doi:10.1124/jpet.113.20822324556663
  • AckermannM, KimYO, WagnerWL, et al. Effects of nintedanib on the microvascular architecture in a lung fibrosis model. Angiogenesis. 2017;20(3):359–372. doi:10.1007/s10456-017-9543-z28283856
  • HuangJ, MaierC, ZhangY, et al. Nintedanib inhibits macrophage activation and ameliorates vascular and fibrotic manifestations in the Fra2 mouse model of systemic sclerosis. Ann Rheum Dis. 2017;76(11):1941–1948. doi:10.1136/annrheumdis-2016-21082328814429
  • LeeHY, HurJ, KimIK, et al. Effect of nintedanib on airway inflammation and remodeling in a murine chronic asthma model. Exp Lung Res. 2017;43(4–5):187–196. doi:10.1080/01902148.2017.133914128696800
  • WollinL-S, TomsicC, ErbKJ. The effect of nintedanib in a mouse model of severe allergic lung inflammation and remodeling. Am J Respir Crit Care Med. 2018;197:A1062.
  • NelAE. T-cell activation through the antigen receptor. Part 1: signaling components, signaling pathways, and signal integration at the T-cell antigen receptor synapse. J Allergy Clin Immunol. 2002;109(5):758–770. doi:10.1067/mai.2002.12425911994696
  • StrausDB, WeissA. Genetic evidence for the involvement of the lck tyrosine kinase in signal transduction through the T cell antigen receptor. Cell. 1992;70(4):585–593. doi:10.1016/0092-8674(92)90428-F1505025
  • LovattM, FilbyA, ParraviciniV, WerlenG, PalmerE, ZamoyskaR. Lck regulates the threshold of activation in primary T cells, while both Lck and Fyn contribute to the magnitude of the extracellular signal-related kinase response. Mol Cell Biol. 2006;26(22):8655. doi:10.1128/MCB.00168-0616966372
  • YasudaK, NagafukuM, ShimaT, et al. Cutting edge: Fyn is essential for tyrosine phosphorylation of Csk-binding protein/phosphoprotein associated with glycolipid-enriched microdomains in lipid rafts in resting T cells. J Immunol. 2002;169(6):2813–2817. doi:10.4049/jimmunol.169.6.281312218089
  • MolinaTJ, KishiharaK, SiderovskiDP, et al. Profound block in thymocyte development in mice lacking p56lck. Nature. 1992;357(6374):161–164. doi:10.1038/357161a01579166
  • SerafinV, CapuzzoG, MilaniG, et al. Glucocorticoid resistance is reverted by LCK inhibition in pediatric T-cell acute lymphoblastic leukemia. Blood. 2017;130(25):2750–2761. doi:10.1182/blood-2017-05-78460329101238
  • WindS, SchmidU, FreiwaldM, et al. Clinical pharmacokinetics and pharmacodynamics of nintedanib. Clin Pharmacokinet. 2019;58(9):1131–1147. doi:10.1007/s40262-019-00766-031016670
  • ZumwaldeNA, DomaeE, MescherMF, ShimizuY. ICAM-1-dependent homotypic aggregates regulate CD8 T cell effector function and differentiation during T cell activation. J Immunol. 2013;191(7):3681–3693. doi:10.4049/jimmunol.120195423997225
  • FigueiredoAS, SchumacherA. The T helper type 17/regulatory T cell paradigm in pregnancy. Immunology. 2016;148(1):13–21. doi:10.1111/imm.1259526855005
  • Re SL, LisonD, HuauxF. CD4+ T lymphocytes in lung fibrosis: diverse subsets, diverse functions. J Leukoc Biol. 2013;93(4):499–510. doi:10.1189/jlb.051226123159927
  • SeddonB, LegnameG, TomlinsonP, ZamoyskaR. Long-term survival but impaired homeostatic proliferation of naive T cells in the absence of p56lck. Science. 2000;290(5489):127–131. doi:10.1126/science.290.5489.12711021796
  • RossyJ, WilliamsonDJ, GausK. How does the kinase Lck phosphorylate the T cell receptor? Spatial organization as a regulatory mechanism. Front Immunol. 2012;3:167. doi:10.3389/fimmu.2012.0016722723799
  • LanzavecchiaA, IezziG, ViolaA. From TCR engagement to T cell activation: a kinetic view of T cell behavior. Cell. 1999;96(1):1–4. doi:10.1016/S0092-8674(00)80952-69989490
  • LeeK-H, DinnerAR, TuC, et al. The immunological synapse balances T cell receptor signaling and degradation. Science. 2003;302(5648):1218–1222. doi:10.1126/science.108650714512504
  • VarmaR, CampiG, YokosukaT, SaitoT, DustinML. T cell receptor-proximal signals are sustained in peripheral microclusters and terminated in the central supramolecular activation cluster. Immunity. 2006;25(1):117–127. doi:10.1016/j.immuni.2006.04.01016860761
  • EisenT, ShparykY, MacleodN, et al. Effect of small angiokinase inhibitor nintedanib (BIBF 1120) on QT interval in patients with previously untreated, advanced renal cell cancer in an open-label, Phase II study. Invest New Drugs. 2013;31(5):1283–1293. doi:10.1007/s10637-013-9962-723625328
  • MrossK, StefanicM, GmehlingD, et al. Phase I study of the angiogenesis inhibitor BIBF 1120 in patients with advanced solid tumors. Clin Cancer Res. 2010;16(1):311–319. doi:10.1158/1078-0432.CCR-09-069420028771
  • OkazakiT, NakaoA, NakanoH, et al. Impairment of bleomycin-induced lung fibrosis in CD28-deficient mice. J Immunol. 2001;167(4):1977–1981. doi:10.4049/jimmunol.167.4.197711489978
  • HuauxF, LiuT, McGarryB, UllenbruchM, XingZ, PhanSH. Eosinophils and T lymphocytes possess distinct roles in bleomycin-induced lung injury and fibrosis. J Immunol. 2003;171(10):5470–5481. doi:10.4049/jimmunol.171.10.547014607953
  • BarbarinV, ArrasM, MissonP, et al. Characterization of the effect of interleukin-10 on silica-induced lung fibrosis in mice. Am J Respir Cell Mol Biol. 2004;31(1):78–85. doi:10.1165/rcmb.2003-0299OC14975940
  • ChristensenPJ, GoodmanRE, PastorizaL, MooreB, ToewsGB. Induction of lung fibrosis in the mouse by intratracheal instillation of fluorescein isothiocyanate is not T-cell-dependent. Am J Pathol. 1999;155(5):1773–1779. doi:10.1016/S0002-9440(10)65493-410550334
  • GalatiD, De MartinoM, TrottaA, et al. Peripheral depletion of NK cells and imbalance of the Treg/Th17 axis in idiopathic pulmonary fibrosis patients. Cytokine. 2014;66(2):119–126. doi:10.1016/j.cyto.2013.12.00324418172
  • NakashimaT, JinninM, YamaneK, et al. Impaired IL-17 signaling pathway contributes to the increased collagen expression in scleroderma fibroblasts. J Immunol. 2012;188(8):3573–3583. doi:10.4049/jimmunol.110059122403442
  • ChizzoliniC, DufourAM, BrembillaNC. Is there a role for IL-17 in the pathogenesis of systemic sclerosis? Immunol Lett. 2018;195:61–67. doi:10.1016/j.imlet.2017.09.00728919455
  • GrussHJ, ScottC, RollinsBJ, BrachMA, HerrmannF. Human fibroblasts express functional IL-2 receptors formed by the IL-2R alpha- and beta-chain subunits: association of IL-2 binding with secretion of the monocyte chemoattractant protein-1. J Immunol. 1996;157(2):851–857.8752938
  • KangR, TangD, LotzeMT, Zeh IIIHJ. Autophagy is required for IL-2-mediated fibroblast growth. Exp Cell Res. 2013;319(4):556–565. doi:10.1016/j.yexcr.2012.11.01223195496
  • SaitoA, OkazakiH, SugawaraI, YamamotoK, TakizawaH. Potential action of IL-4 and IL-13 as fibrogenic factors on lung fibroblasts in vitro. Int Arch Allergy Immunol. 2003;132(2):168–176. doi:10.1159/00007371814600429
  • SuS, ZhaoQ, HeC, et al. miR-142-5p and miR-130a-3p are regulated by IL-4 and IL-13 and control profibrogenic macrophage program. Nat Commun. 2015;6(1):8523. doi:10.1038/ncomms952326436920
  • WynnTA. Integrating mechanisms of pulmonary fibrosis. J Exp Med. 2011;208(7):1339–1350. doi:10.1084/jem.2011055121727191
  • GuoJ, YaoH, LinX, et al. IL-13 induces YY1 through the AKT pathway in lung fibroblasts. PLoS One. 2015;10(3):e0119039. doi:10.1371/journal.pone.011903925775215
  • EsnaultS, BernauK, TorrEE, BochkovYA, JarjourNN, SandboN. RNA-sequencing analysis of lung primary fibroblast response to eosinophil-degranulation products predicts downstream effects on inflammation, tissue remodeling and lipid metabolism. Respir Res. 2017;18(1):188. doi:10.1186/s12931-017-0669-829126429
  • HashimotoT, NakamuraM, OshikaY, et al. Interleukin-10 relieves the inhibitory effects of interferon-γ on normal human lung fibroblasts. Int J Mol Med. 2001;7(2):149–154.11172617
  • KhannaD, SpinoC, JohnsonS, et al. Abatacept in early diffuse cutaneous systemic sclerosis - results of a Phase 2 investigator-initiated, multicenter, double-blind randomized placebo-controlled trial. Arthritis Rheumatol. 2020;72(1):125–136. doi:10.1002/art.4105531342624