Rationale, design and objectives of two phase III, randomised, placebo-controlled studies of GLPG1690, a novel autotaxin inhibitor, in idiopathic pulmonary fibrosis (ISABELA 1 and 2)

Toby M Maher, Michael Kreuter, David J Lederer, Kevin K Brown, Wim Wuyts, Nadia Verbruggen, Simone Stutvoet, Ann Fieuw, Paul Ford, Walid Abi-Saab, Marlies Wijsenbeek, Toby M Maher, Michael Kreuter, David J Lederer, Kevin K Brown, Wim Wuyts, Nadia Verbruggen, Simone Stutvoet, Ann Fieuw, Paul Ford, Walid Abi-Saab, Marlies Wijsenbeek

Abstract

Introduction: While current standard of care (SOC) for idiopathic pulmonary fibrosis (IPF) slows disease progression, prognosis remains poor. Therefore, an unmet need exists for novel, well-tolerated agents that reduce lung function decline and improve quality of life. Here we report the design of two phase III studies of the novel IPF therapy, GLPG1690.

Methods and analysis: Two identically designed, phase III, international, randomised, double-blind, placebo-controlled, parallel-group, multicentre studies (ISABELA 1 and 2) were initiated in November 2018. It is planned that, in each study, 750 subjects with IPF will be randomised 1:1:1 to receive oral GLPG1690 600 mg, GLPG1690 200 mg or placebo, once daily, on top of local SOC, for at least 52 weeks. The primary endpoint is rate of decline of forced vital capacity (FVC) over 52 weeks. Key secondary endpoints are week 52 composite endpoint of disease progression or all-cause mortality (defined as composite endpoint of first occurrence of ≥10% absolute decline in per cent predicted FVC or all-cause mortality at week 52); time to first respiratory-related hospitalisation until end of study; and week 52 change from baseline in the St George's Respiratory Questionnaire total score (a quality-of-life measure).

Ethics and dissemination: Studies will be conducted in accordance with Good Clinical Practice guidelines, Declaration of Helsinki principles, and local ethical and legal requirements. Results will be reported in a peer-reviewed publication.

Trial registration numbers: NCT03711162; NCT03733444.

Keywords: interstitial fibrosis; rare lung diseases.

Conflict of interest statement

Competing interests: TMM has received personal fees from AstraZeneca, Bayer, Biogen, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Galapagos, GlaxoSmithKline, Indalo Therapeutics, Pliant Therapeutics, ProMetic, Roche, Samumed and UCB, and stock options from Apellis outside the submitted work. His institution received grants or research fees from AstraZeneca, GlaxoSmithKline and UCB outside the submitted work. MK reports grants and personal fees from Galapagos during the conduct of the study, and grants and personal fees from Boehringer Ingelheim and Roche outside the submitted work. DJL reports personal fees from Galapagos during the conduct of the study, and personal fees from Philips Respironics, Roche, Sanofi Genzyme and Veracyte, grants and personal fees from Boehringer Ingelheim, FibroGen and Global Blood Therapeutics, unpaid Steering Committee membership for Galecto, and institutional fees for consultancy work from the Pulmonary Fibrosis Foundation outside the submitted work. KKB reports personal fees from Galapagos during the conduct of the study, and conversation under CDA with Genoa, grants from National Heart, Lung, and Blood Institute, and personal fees from Aeolus, AstraZeneca, aTyr, Biogen, Boehringer Ingelheim, Galapagos, Galecto Biotech, MedImmune, Novartis, Roche/Genentech, ProMetic, Patara and Third Pole outside the submitted work. WW reports no personal fees; the University Hospitals Leuven received consultancy fees from Galapagos during the conduct of the study and grants from Boehringer Ingelheim and Roche outside the submitted work. NV, SS, PF and WA-S are employees of, and have received warrants from, Galapagos. AF is an employee of Galapagos. MW reports no personal fees; the Erasmus MC received an advisory board fee from Galapagos, and speaker and advisory board fees from Boehringer Ingelheim and Hoffmann-La Roche outside the submitted work.

Figures

Figure 1
Figure 1
Design of the ISABELA 1 and 2 studies. D, day; EoSA, end-of-study assessment; EoST, end-of-study treatment; FU, follow-up; qd, once daily; V, visit; W, week.

References

    1. Sharif R. Overview of idiopathic pulmonary fibrosis (IPF) and evidence-based guidelines. Am J Manag Care 2017;23(11 Suppl):S176–82.
    1. Martinez FJ, Collard HR, Pardo A, et al. . Idiopathic pulmonary fibrosis. Nat Rev Dis Primers 2017;3:17074 10.1038/nrdp.2017.74
    1. Wolters PJ, Collard HR, Jones KD. Pathogenesis of idiopathic pulmonary fibrosis. Annu Rev Pathol Mech Dis 2014;9:157–79. 10.1146/annurev-pathol-012513-104706
    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. 10.1164/rccm.2009-040GL
    1. Li S, Zhao J, Shang D, et al. . Ubiquitination and deubiquitination emerge as players in idiopathic pulmonary fibrosis pathogenesis and treatment. JCI Insight 2018;3:e120362 10.1172/jci.insight.120362
    1. Ley B, Collard HR, King TE. Clinical course and prediction of survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2011;183:431–40. 10.1164/rccm.201006-0894CI
    1. Spagnolo P, Tzouvelekis A, Bonella F. The management of patients with idiopathic pulmonary fibrosis. Front Med 2018;5:148 10.3389/fmed.2018.00148
    1. Chambers RC. Abnormal wound healing responses in pulmonary fibrosis: focus on coagulation signalling. Eur Respir Rev 2008;17:130–7. 10.1183/09059180.00010905
    1. Scelfo C, Caminati A, Harari S. Recent advances in managing idiopathic pulmonary fibrosis. F1000Res 2017;6:2052 10.12688/f1000research.10720.1
    1. Raghu G. Pharmacotherapy for idiopathic pulmonary fibrosis: Current landscape and future potential. Eur Respir Rev 2017;26:pii: 170071 10.1183/16000617.0071-2017
    1. Raghu G, Rochwerg B, Zhang Y, et al. . An official ATS/ERS/JRS/ALAT clinical practice guideline: treatment of idiopathic pulmonary fibrosis. An update of the 2011 clinical practice guideline. Am J Respir Crit Care Med 2015;192:e3–19. 10.1164/rccm.201506-1063ST
    1. Noble PW, Albera C, Bradford WZ, et al. . Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials. Lancet 2011;377:1760–9. 10.1016/S0140-6736(11)60405-4
    1. Richeldi L, du Bois RM, Raghu G, et al. . Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014;370:2071–82. 10.1056/NEJMoa1402584
    1. King TE, Bradford WZ, Castro-Bernardini S, et al. . A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med 2014;370:2083–92. 10.1056/NEJMoa1402582
    1. Richeldi L, Cottin V, du Bois RM, et al. . Nintedanib in patients with idiopathic pulmonary fibrosis: Combined evidence from the TOMORROW and INPULSIS® trials. Respir Med 2016;113:74–9. 10.1016/j.rmed.2016.02.001
    1. Jo HE, Glaspole I, Grainge C, et al. . Baseline characteristics of idiopathic pulmonary fibrosis: analysis from the Australian idiopathic pulmonary fibrosis registry. Eur Respir J 2017;49:1601592 10.1183/13993003.01592-2016
    1. Fisher M, Nathan SD, Hill C, et al. . Predicting life expectancy for pirfenidone in idiopathic pulmonary fibrosis. J Manag Care Spec Pharm 2017;23(3-b Suppl):S17–S24. 10.18553/jmcp.2017.23.3-b.s17
    1. Galli JA, Pandya A, Vega-Olivo M, et al. . Pirfenidone and nintedanib for pulmonary fibrosis in clinical practice: tolerability and adverse drug reactions. Respirology 2017;22:1171–8. 10.1111/resp.13024
    1. Kreuter M, Swigris J, Pittrow D, et al. . Health related quality of life in patients with idiopathic pulmonary fibrosis in clinical practice: insights-IPF registry. Respir Res 2017;18:139 10.1186/s12931-017-0621-y
    1. Loveman E, Copley VR, Scott DA, et al. . Comparing new treatments for idiopathic pulmonary fibrosis – a network meta-analysis. BMC Pulm Med 2015;15:37 10.1186/s12890-015-0034-y
    1. van Manen MJG, Geelhoed JJM, Tak NC, et al. . Optimizing quality of life in patients with idiopathic pulmonary fibrosis. Ther Adv Respir Dis 2017;11:157–69. 10.1177/1753465816686743
    1. Tager AM. Autotaxin emerges as a therapeutic target for idiopathic pulmonary fibrosis: limiting fibrosis by limiting lysophosphatidic acid synthesis. Am J Respir Cell Mol Biol 2012;47:563–5. 10.1165/rcmb.2012-0235ED
    1. Funke M, Zhao Z, Xu Y, et al. . The lysophosphatidic acid receptor LPA1 promotes epithelial cell apoptosis after lung injury. Am J Respir Cell Mol Biol 2012;46:355–64. 10.1165/rcmb.2010-0155OC
    1. Ninou I, Magkrioti C, Aidinis V. Autotaxin in pathophysiology and pulmonary fibrosis. Front Med 2018;5:180 10.3389/fmed.2018.00180
    1. Oikonomou N, Mouratis M-A, Tzouvelekis A, et al. . Pulmonary autotaxin expression contributes to the pathogenesis of pulmonary fibrosis. Am J Respir Cell Mol Biol 2012;47:566–74. 10.1165/rcmb.2012-0004OC
    1. Tager AM, LaCamera P, Shea BS, et al. . The lysophosphatidic acid receptor LPA1 links pulmonary fibrosis to lung injury by mediating fibroblast recruitment and vascular leak. Nat Med 2008;14:45–54. 10.1038/nm1685
    1. Desroy N, Housseman C, Bock X, et al. . Discovery of 2-[[2-Ethyl-6-[4-[2-(3-hydroxyazetidin-1-yl)-2-oxoethyl]piperazin-1-yl]-8-methylimidazo[1,2-a]pyridin-3-yl]methylamino]-4-(4-fluorophenyl)thiazole-5-carbonitrile (GLPG1690), a first-in-class autotaxin inhibitor undergoing clinical evaluation for the treatment of idiopathic pulmonary fibrosis. J Med Chem 2017;60:3580–90. 10.1021/acs.jmedchem.7b00032
    1. van der Aah EM, Fagard L, Desrivot J, et al. . Favorable human safety, pharmacokinetics and pharmacodynamics of the autotaxin inhibitor GLPG1690, a potential new treatment in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2016;193:A2701.
    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;6:627–35. 10.1016/S2213-2600(18)30181-4
    1. Raghu G, Remy-Jardin M, Myers JL, et al. . Diagnosis of idiopathic pulmonary fibrosis. An official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med 2018;198:e44–68. 10.1164/rccm.201807-1255ST
    1. Lynch DA, Sverzellati N, Travis WD, et al. . Diagnostic criteria for idiopathic pulmonary fibrosis: a Fleischner Society white paper. Lancet Respir Med 2018;6:138–53. 10.1016/S2213-2600(17)30433-2
    1. Jones PW, Quirk FH, Baveystock CM, et al. . A self-complete measure of health status for chronic airflow limitation. The St. George's Respiratory Questionnaire. Am Rev Respir Dis 1992;145:1321–7. 10.1164/ajrccm/145.6.1321
    1. Miller MR, Hankinson J, Brusasco V, et al. . Standardisation of spirometry. Eur Respir J 2005;26:319–38. 10.1183/09031936.05.00034805
    1. Quanjer PH, Stanojevic S, Cole TJ, et al. . Multi-ethnic reference values for spirometry for the 3-95-yr age range: the global lung function 2012 equations. Eur Respir J 2012;40:1324–43. 10.1183/09031936.00080312
    1. Rabin R, de Charro F. EQ-5D: a measure of health status from the EuroQol group. Ann Med 2001;33:337–43. 10.3109/07853890109002087
    1. Patel AS, Siegert RJ, Brignall K, et al. . The development and validation of the King's Brief Interstitial Lung Disease (K-BILD) health status questionnaire. Thorax 2012;67:804–10. 10.1136/thoraxjnl-2012-201581
    1. Birring SS, Prudon B, Carr AJ, et al. . Development of a symptom specific health status measure for patients with chronic cough: Leicester Cough Questionnaire (LCQ). Thorax 2003;58:339–43. 10.1136/thorax.58.4.339
    1. Brown AW, Nathan SD. The value and application of the 6-minute-walk test in idiopathic pulmonary fibrosis. Ann Am Thorac Soc 2018;15:3–10. 10.1513/AnnalsATS.201703-244FR
    1. American Association for Respiratory Care AARC clinical practice guideline. Exercise testing for evaluation of hypoxemia and/or desaturation. American Association for respiratory care. Respir Care 1992;37:907–12.
    1. ATS Committee on Proficiency Standards for Clinical Pulmonary Function Laboratories ATS statement: guidelines for the six-minute walk test. Am J Respir Crit Care Med 2002;166:111–7. 10.1164/ajrccm.166.1.at1102
    1. Maher TM, Molina-Molina M, Russell A-M, et al. . Unmet needs in the treatment of idiopathic pulmonary fibrosis—insights from patient chart review in five European countries. BMC Pulm Med 2017;17:124 10.1186/s12890-017-0468-5
    1. Maher TM, Swigris JJ, Kreuter M, et al. . Identifying barriers to idiopathic pulmonary fibrosis treatment: a survey of patient and physician views. Respiration 2018;96:514–24. 10.1159/000490667
    1. Lederer DJ, Bradford WZ, Fagan EA, et al. . Sensitivity analyses of the change in FVC in a phase 3 trial of pirfenidone for idiopathic pulmonary fibrosis. Chest 2015;148:196–201. 10.1378/chest.14-2817
    1. Maher TM. Combination therapy and the start of a new epoch for idiopathic pulmonary fibrosis? Am J Respir Crit Care Med 2018;197:283–4. 10.1164/rccm.201709-1939ED
    1. Wuyts WA, Antoniou KM, Borensztajn K, et al. . Combination therapy: the future of management for idiopathic pulmonary fibrosis? Lancet Respir Med 2014;2:933–42. 10.1016/S2213-2600(14)70232-2
    1. Kolb M, Raghu G, Wells AU, et al. . Nintedanib plus sildenafil in patients with idiopathic pulmonary fibrosis. N Engl J Med 2018;379:1722–31. 10.1056/NEJMoa1811737
    1. Collard HR, King TE, Bartelson BB, et al. . Changes in clinical and physiologic variables predict survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2003;168:538–42. 10.1164/rccm.200211-1311OC
    1. Flaherty KR, Mumford JA, Murray S, et al. . Prognostic implications of physiologic and radiographic changes in idiopathic interstitial pneumonia. Am J Respir Crit Care Med 2003;168:543–8. 10.1164/rccm.200209-1112OC
    1. Spagnolo P, Tonelli R, Cocconcelli E, et al. . Idiopathic pulmonary fibrosis: diagnostic pitfalls and therapeutic challenges. Multidiscip Respir Med 2012;7:42 10.1186/2049-6958-7-42

Source: PubMed

3
Subscribe