Target inhibition of galectin-3 by inhaled TD139 in patients with idiopathic pulmonary fibrosis

Nikhil Hirani, Alison C MacKinnon, Lisa Nicol, Paul Ford, Hans Schambye, Anders Pedersen, Ulf J Nilsson, Hakon Leffler, Tariq Sethi, Susan Tantawi, Lise Gravelle, Robert J Slack, Ross Mills, Utsa Karmakar, Duncan Humphries, Fredrik Zetterberg, Lucy Keeling, Lyn Paul, Philip L Molyneaux, Feng Li, Wendy Funston, Ian A Forrest, A John Simpson, Michael A Gibbons, Toby M Maher, Nikhil Hirani, Alison C MacKinnon, Lisa Nicol, Paul Ford, Hans Schambye, Anders Pedersen, Ulf J Nilsson, Hakon Leffler, Tariq Sethi, Susan Tantawi, Lise Gravelle, Robert J Slack, Ross Mills, Utsa Karmakar, Duncan Humphries, Fredrik Zetterberg, Lucy Keeling, Lyn Paul, Philip L Molyneaux, Feng Li, Wendy Funston, Ian A Forrest, A John Simpson, Michael A Gibbons, Toby M Maher

Abstract

Galectin (Gal)-3 is a profibrotic β-galactoside-binding lectin that plays a key role in the pathogenesis of idiopathic pulmonary fibrosis (IPF) and IPF exacerbations. TD139 is a novel and potent small-molecule inhibitor of Gal-3.A randomised, double-blind, multicentre, placebo-controlled, phase 1/2a study was conducted to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of inhaled TD139 in 36 healthy subjects and 24 patients with IPF. Six dose cohorts of six healthy subjects were evaluated (4:2 TD139:placebo ratio) with single doses of TD139 (0.15-50 mg) and three dose cohorts of eight patients with IPF (5:3 TD139:placebo ratio) with once-daily doses of TD139 (0.3-10 mg) for 14 days.Inhaled TD139 was well tolerated with no significant treatment-related side-effects. TD139 was rapidly absorbed, with mean time taken to reach maximum plasma concentration (C max) values ranging from 0.6 to 3 h and a plasma half-life (T 1/2) of 8 h. The concentration of TD139 in the lung was >567-fold higher than in the blood, with systemic exposure predicting exposure in the target compartment. Gal-3 expression on alveolar macrophages was reduced in the 3 and 10 mg dose groups compared with placebo, with a concentration-dependent inhibition demonstrated. Inhibition of Gal-3 expression in the lung was associated with reductions in plasma biomarkers centrally relevant to IPF pathobiology (platelet-derived growth factor-BB, plasminogen activator inhibitor-1, Gal-3, CCL18 and YKL-40).TD139 is safe and well tolerated in healthy subjects and IPF patients. It was shown to suppress Gal-3 expression on bronchoalveolar lavage macrophages and, in a concerted fashion, decrease plasma biomarkers associated with IPF progression.

Trial registration: ClinicalTrials.gov NCT02257177.

Conflict of interest statement

Conflict of interest: N. Hirani reports grants from Galecto Biotech, during the conduct of the study. Conflict of interest: A.C. MacKinnon reports personal fees from Galecto Biotech, outside the submitted work; and has a patent CA2,794,066 issued, a patent US13/832,672 issued and a patent WO/2014/067986 pending (all patents are fully owned by Galecto Biotech). Conflict of interest: L. Nicol reports grants from Galecto Biotech, during the conduct of the study; personal fees for lectures from Boehringer Ingelheim, outside the submitted work. Conflict of interest: P. Ford reports personal fees and nonfinancial support from Galecto, during the conduct of the study; and has a patent TD139 issued. Conflict of interest: H. Schambye reports personal fees from Galecto Inc, outside the submitted work; and has a patent WO/2016/180483 pending (fully owned by Galecto Biotech). Conflict of interest: A. Pedersen reports personal fees from Galecto Biotech, outside the submitted work. Conflict of interest: U.J. Nilsson has a patent CA2,794,066 issued, a patent US13/832,672 issued, a patent WO/2014/067986 pending, a patent WO/2005/113569 pending and a patent WO/2009/139719 pending (all patents are fully owned by Galecto Biotech). Conflict of interest: H. Leffler has a patent CA2,794,066 issued, a patent US13/832,672 issued, a patent WO/2014/067986 pending and a patent WO/2005/113569 pending (all patents are fully owned by Galecto Biotech). Conflict of interest: T. Sethi reports personal fees from Galecto Biotech, outside the submitted work; and has a patent CA2,794,066 issued, a patent US13/832,672 issued and a patent WO/2014/067986 pending (patents are fully owned by Galecto Biotech). Conflict of interest: S. Tantawi reports personal fees from Galecto Biotech, outside the submitted work. Conflict of interest: L. Gravelle reports personal fees from Galecto Biotech, outside the submitted work; and has a patent WO/2017/103109 pending (fully owned by Galecto Biotech). Conflict of interest: R.J. Slack reports personal fees from Galecto Biotech, outside the submitted work. Conflict of interest: R. Mills has nothing to disclose. Conflict of interest: U. Karmakar has nothing to disclose. Conflict of interest: D. Humphries has nothing to disclose. Conflict of interest: F. Zetterberg reports personal fees from Galecto Biotech, outside the submitted work. Conflict of interest: L. Keeling has nothing to disclose. Conflict of interest: L. Paul has nothing to disclose. Conflict of interest: P.L. Molyneaux has, via his institution, received industry-academic funding from AstraZeneca and has received speaker and consultancy fees from Boehringer Ingelheim and Hoffman-La Roche, outside the submitted work. Conflict of interest: F. Li has nothing to disclose. Conflict of interest: W. Funston has nothing to disclose. Conflict of interest: I.A. Forrest reports personal fees for consultancy and meeting attendance from Boehringer Ingelheim, personal fees for lectures and meeting attendance from Roche Ltd, outside the submitted work. Conflict of interest: A.J. Simpson has nothing to disclose. Conflict of interest: M.A. Gibbons has nothing to disclose. Conflict of interest: T.M. Maher has, via his institution, received industry-academic funding from AstraZeneca and GlaxoSmithKline R&D, and has received consultancy or speaker fees from AstraZeneca, Bayer, Blade Therapeutics, Boehringer Ingelheim, Bristol-Myers Squibb, Galapagos, GlaxoSmithKline R&D, Indalo, Novartis, Pliant, Respivant, Roche and Samumed.

Copyright ©ERS 2021.

Figures

FIGURE 1
FIGURE 1
Schematic of the study design and CONSORT (Consolidated Standards of Reporting Trials) diagrams. PK: pharmacokinetics; BAL: bronchoalveolar lavage; PD: pharmacodynamics. a) Part 1 (healthy subjects). Each single-dose cohort consisted of six healthy subjects randomised 4:2 to active and placebo. A minimum of five patients’ data was required in each cohort prior to data review. b) CONSORT diagram for part 1. c) Part 2 (idiopathic pulmonary fibrosis patients). Each repeat-dose cohort consisted of eight subjects randomised 5:3 to active and placebo. A minimum of seven patients’ data was required in each cohort prior to data review. d) CONSORT diagram for part 2.
FIGURE 2
FIGURE 2
TD139 pharmacokinetics in healthy subjects and idiopathic pulmonary fibrosis (IPF) patients. ELF: epithelial lining fluid; AM: alveolar macrophage. a) Healthy subjects: log-linear mean plasma concentration of TD139 versus time over 48 h following a single dose of drug. b) IPF patients: log-linear mean plasma concentration of TD139 versus time over 24 h following the first dose of drug on day 1 or the last dose on day 14. c) Log-linear individual measured concentrations (median) in plasma (total), ELF and AMs at 2 h post-administration of 0.3, 3 and 10 mg TD139 on day 14. d) Correlation between concentrations of TD139 in ELF and AMs for all active cohorts on day 14 (r=0.89 (95% CI 0.70–0.96); p<0.0001). e) Correlation between concentrations of TD139 in plasma (total) and AMs for all active cohorts on day 14 (r=0.89 (95% CI 0.65–0.96); p<0.0001). Shading represents 95% CI of the linear fit.
FIGURE 3
FIGURE 3
Galectin (Gal)-3 changes in alveolar macrophages (AMs). a) Box plot of TD139 dose-dependent effect on percentage change from baseline in surface macrophage Gal-3 levels. Boxes indicate median and interquartile range; whiskers indicate minimum–maximum range. p-value from ANCOVA model adjusted for baseline. b) Concentration-dependent effect between TD139 in AMs and percentage of baseline in surface macrophage Gal-3, with maximum plasma concentration (Cmax) at 10 mg dose (dotted line) shown for comparison (r= −0.63 (95% CI −0.86– −0.18); p=0.011). Shading represents 95% CI of the linear fit. Individual subject data are labelled by dose. c) Fitted maximum effect model of surface macrophage Gal-3 (percentage of baseline) versus macrophage TD139 concentration. Maximum effect (Emax, AM surface Gal-3 percentage of baseline) and median inhibitory concentration (IC50) values are shown. Shading represents 95% CI of the fitted maximum effect model.
FIGURE 4
FIGURE 4
Biomarker changes in plasma. PDGF: platelet-derived growth factor; AM: alveolar macrophage; PAI: plasminogen activator inhibitor; Gal: galectin; CCL: chemokine (C-C motif) ligand. a, c, e, g, i) Percentage of baseline at day 14 for a) PDGF-BB, c) PAI-1, e) Gal-3, g) CCL18 and i) YKL-40 in the 0.3, 3 and 10 mg TD139 dose groups or placebo. Boxes indicate median and interquartile range; whiskers indicate minimum–maximum range. p-value from ANCOVA model adjusted for baseline. b, d, f, h, j) Correlation between percentage of baseline in b) PDGF-BB, d) PAI-1, f) Gal-3, h) CCL18 and j) YKL-40 versus the change in lung macrophage Gal-3 expression in the placebo and 10 mg TD139 groups. Dotted line shows no change from baseline. Fisher's exact test for percentage of baseline <100% for plasma Gal-3 and AM Gal-3 expression (yes/no) versus treatment group (placebo, 10 mg) p=0.031.

References

    1. Raghu G, Collard HR, Egan JJ, et al. . An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med 2011; 183: 788–824. doi:10.1164/rccm.2009-040GL
    1. King TE Jr, Pardo A, Selman M. Idiopathic pulmonary fibrosis. Lancet 2011; 378: 1949–1961. doi:10.1016/S0140-6736(11)60052-4
    1. King TE Jr, Bradford WZ, Castro-Bernardini S, et al. . A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med 2014; 370: 2083–2092. doi:10.1056/NEJMoa1402582
    1. Richeldi L, du Bois RM, Raghu G, et al. . Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014; 370: 2071–2082. doi:10.1056/NEJMoa1402584
    1. Nishi Y, Sano H, Kawashima T, et al. . Role of galectin-3 in human pulmonary fibrosis. Allergol Int 2007; 56: 57–65. doi:10.2332/allergolint.O-06-449
    1. Mackinnon AC, Gibbons MA, Farnworth SL, et al. . Regulation of transforming growth factor-β1-driven lung fibrosis by galectin-3. Am J Respir Crit Care Med 2012; 185: 537–546. doi:10.1164/rccm.201106-0965OC
    1. Partridge EA, Le Roy C, Di Guglielmo GM, et al. . Regulation of cytokine receptors by Golgi N-glycan processing and endocytosis. Science 2004; 306: 120–124. doi:10.1126/science.1102109
    1. Shen H, Wang J, Min J, et al. . Activation of TGF-β1/α-SMA/Col I profibrotic pathway in fibroblasts by galectin-3 contributes to atrial fibrosis in experimental models and patients. Cell Physiol Biochem 2018; 47: 851–863. doi:10.1159/000490077
    1. Chen WS, Cao Z, Leffler H, et al. . Galectin-3 inhibition by a small-molecule inhibitor reduces both pathological corneal neovascularization and fibrosis. Invest Ophthalmol Vis Sci 2017; 58: 9–20. doi:10.1167/iovs.16-20009
    1. Guo SM, Feng ZM. Galectin-3 mediates the effect of PDGF on pulmonary arterial hypertension. Int J Clin Exp Med 2015; 8: 15302–15307.
    1. Jastrzebski K, Zdzalik-Bielecka D, Maminska A, et al. . Multiple routes of endocytic internalization of PDGFRβ contribute to PDGF-induced STAT3 signaling. J Cell Sci 2017; 130: 577–589. doi:10.1242/jcs.191213
    1. Henderson NC, Mackinnon AC, Farnworth SL, et al. . Galectin-3 regulates myofibroblast activation and hepatic fibrosis. Proc Natl Acad Sci USA 2006; 103: 5060–5065. doi:10.1073/pnas.0511167103
    1. MacKinnon AC, Liu X, Hadoke PW, et al. . Inhibition of galectin-3 reduces atherosclerosis in apolipoprotein E-deficient mice. Glycobiology 2013; 23: 654–663. doi:10.1093/glycob/cwt006
    1. Henderson NC, Mackinnon AC, Farnworth SL, et al. . Galectin-3 expression and secretion links macrophages to the promotion of renal fibrosis. Am J Pathol 2008; 172: 288–298. doi:10.2353/ajpath.2008.070726
    1. Delaine T, Collins P, MacKinnon A, et al. . Galectin-3-binding glycomimetics that strongly reduce bleomycin-induced lung fibrosis and modulate intracellular glycan recognition. Chembiochem 2016; 17: 1759–1770. doi:10.1002/cbic.201600285
    1. MacKinnon AC, Farnworth SL, Hodkinson PS, et al. . Regulation of alternative macrophage activation by galectin-3. J Immunol 2008; 180: 2650–2658. doi:10.4049/jimmunol.180.4.2650
    1. Mackinnon AC, Humphries D, Mills R, et al. . Deletion of galectin-3 in myeloid cells inhibits bleomycin-induced lung fibrosis. Am J Respir Crit Care Med 2019; 199: A2422. doi:10.1164/ajrccm-conference.2019.199.1_MeetingAbstracts.A2422
    1. Parmar NR, Mackinnon AC, Zetterberg F, et al. . Role of galectin-3 in idiopathic pulmonary fibrosis development. Am J Respir Crit Care Med 2019; 199: A5328. doi:10.1164/ajrccm-conference.2019.199.1_MeetingAbstracts.A5328
    1. Hirani N, Mackinnon A, Nicol L, et al. . TD139, a novel inhaled galectin-3 inhibitor for the treatment of idiopathic pulmonary fibrosis (IPF). Results from the first in (IPF) patients study. Am J Respir Crit Care Med 2017; 195: A7560.
    1. Klech H, Pohl W. Technical recommendations and guidelines for bronchoalveolar lavage (BAL). Report of the European Society of Pneumology Task Group. Eur Respir J 1989; 2: 561–585.
    1. Naderer OJ, Rodvold KA, Jones LS, et al. . Penetration of GSK1322322 into epithelial lining fluid and alveolar macrophages as determined by bronchoalveolar lavage. Antimicrob Agents Chemother 2014; 58: 419–423. doi:10.1128/AAC.01836-13
    1. Rennard SI, Basset G, Lecossier D, et al. . Estimation of volume of epithelial lining fluid recovered by lavage using urea as marker of dilution. J Appl Physiol 1986; 60: 532–538. doi:10.1152/jappl.1986.60.2.532
    1. Molyneaux PL, Smith JJ, Saunders S, et al. . BAL is safe and well tolerated in individuals with idiopathic pulmonary fibrosis: an analysis of the PROFILE study. Am J Respir Crit Care Med 2021; 203: 136–139. doi:10.1164/rccm.202004-1138LE
    1. Sakamoto K, Taniguchi H, Kondoh Y, et al. . Acute exacerbation of IPF following diagnostic bronchoalveolar lavage procedures. Respir Med 2012; 106: 436–442. doi:10.1016/j.rmed.2011.11.006
    1. Lukey PT, Harrison SA, Yang S, et al. . A randomised, placebo-controlled study of omipalisib (PI3K/mTOR) in idiopathic pulmonary fibrosis. Eur Respir J 2019; 53: 1801992. doi:10.1183/13993003.01992-2018
    1. Maher TM, van der Aar EM, Van de Steen O, et al. . Safety, tolerability, pharmacokinetics, and pharmacodynamics of GLPG1690, a novel autotaxin inhibitor, to treat idiopathic pulmonary fibrosis (FLORA): a phase 2a randomised placebo-controlled trial. Lancet Respir Med 2018; 6: 627–635. doi:10.1016/S2213-2600(18)30181-4
    1. Wells AU, Hansell DM, Harrison NK, et al. . Clearance of inhaled 99mTc-DTPA predicts the clinical course of fibrosing alveolitis. Eur Respir J 1993; 6: 797–802.
    1. Bonner JC. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine Growth Factor Rev 2004; 15: 255–273. doi:10.1016/j.cytogfr.2004.03.006
    1. Martinet Y, Rom WN, Grotendorst GR, et al. . Exaggerated spontaneous release of platelet-derived growth factor by alveolar macrophages from patients with idiopathic pulmonary fibrosis. N Engl J Med 1987; 317: 202–209. doi:10.1056/NEJM198707233170404
    1. Ziora D, Jastrzebski D, Adamek M, et al. . Circulating concentration of markers of angiogenic activity in patients with sarcoidosis and idiopathic pulmonary fibrosis. BMC Pulm Med 2015; 15: 113. doi:10.1186/s12890-015-0110-3
    1. Zhu X, Fang X, Chen W, et al. . Lower expression of platelet derived growth factor is associated with better overall survival rate of patients with idiopathic nonspecific interstitial pneumonia. J Thorac Dis 2017; 9: 519–528. doi:10.21037/jtd.2017.02.50
    1. Pechkovsky DV, Prasse A, Kollert F, et al. . Alternatively activated alveolar macrophages in pulmonary fibrosis-mediator production and intracellular signal transduction. Clin Immunol 2010; 137: 89–101. doi:10.1016/j.clim.2010.06.017
    1. Prasse A, Pechkovsky DV, Toews GB, et al. . A vicious circle of alveolar macrophages and fibroblasts perpetuates pulmonary fibrosis via CCL18. Am J Respir Crit Care Med 2006; 173: 781–792. doi:10.1164/rccm.200509-1518OC
    1. Flevaris P, Vaughan D. The role of plasminogen activator inhibitor type-1 in fibrosis. Semin Thromb Hemost 2017; 43: 169–177. doi:10.1055/s-0036-1586228
    1. Eitzman DT, McCoy RD, Zheng X, et al. . Bleomycin-induced pulmonary fibrosis in transgenic mice that either lack or overexpress the murine plasminogen activator inhibitor-1 gene. J Clin Invest 1996; 97: 232–237. doi:10.1172/JCI118396
    1. Hattori N, Degen JL, Sisson TH, et al. . Bleomycin-induced pulmonary fibrosis in fibrinogen-null mice. J Clin Invest 2000; 106: 1341–1350. doi:10.1172/JCI10531
    1. Menou A, Duitman J, Crestani B. The impaired proteases and anti-proteases balance in idiopathic pulmonary fibrosis. Matrix Biol 2018; 68-69: 382–403. doi:10.1016/j.matbio.2018.03.001
    1. Kotani I, Sato A, Hayakawa H, et al. . Increased procoagulant and antifibrinolytic activities in the lungs with idiopathic pulmonary fibrosis. Thromb Res 1995; 77: 493–504. doi:10.1016/0049-3848(95)00025-9
    1. Korthagen NM, van Moorsel CHM, Zanen P, et al. . Evaluation of circulating YKL-40 levels in idiopathic interstitial pneumonias. Lung 2014; 192: 975–980. doi:10.1007/s00408-014-9647-9
    1. Korthagen NM, van Moorsel CHM, Barlo NP, et al. . Serum and BALF YKL-40 levels are predictors of survival in idiopathic pulmonary fibrosis. Respir Med 2011; 105: 106–113. doi:10.1016/j.rmed.2010.09.012
    1. Furuhashi K, Suda T, Nakamura Y, et al. . Increased expression of YKL-40, a chitinase-like protein, in serum and lung of patients with idiopathic pulmonary fibrosis. Respir Med 2010; 104: 1204–1210. doi:10.1016/j.rmed.2010.02.026
    1. Neighbors M, Cabanski CR, Ramalingam TR, et al. . Prognostic and predictive biomarkers for patients with idiopathic pulmonary fibrosis treated with pirfenidone: post-hoc assessment of the CAPACITY and ASCEND trials. Lancet Respir Med 2018; 6: 615–626. doi:10.1016/S2213-2600(18)30185-1
    1. Adegunsoye A, Alqalyoobi S, Linderholm A, et al. . Circulating plasma biomarkers of survival in anti-fibrotic treated patients with idiopathic pulmonary fibrosis. Chest 2020; 158: 1526–1534. doi:10.1016/j.chest.2020.04.066

Source: PubMed

3
Iratkozz fel