Mycophenolate mofetil versus oral cyclophosphamide in scleroderma-related interstitial lung disease (SLS II): a randomised controlled, double-blind, parallel group trial

Donald P Tashkin, Michael D Roth, Philip J Clements, Daniel E Furst, Dinesh Khanna, Eric C Kleerup, Jonathan Goldin, Edgar Arriola, Elizabeth R Volkmann, Suzanne Kafaja, Richard Silver, Virginia Steen, Charlie Strange, Robert Wise, Fredrick Wigley, Maureen Mayes, David J Riley, Sabiha Hussain, Shervin Assassi, Vivien M Hsu, Bela Patel, Kristine Phillips, Fernando Martinez, Jeffrey Golden, M Kari Connolly, John Varga, Jane Dematte, Monique E Hinchcliff, Aryeh Fischer, Jeffrey Swigris, Richard Meehan, Arthur Theodore, Robert Simms, Suncica Volkov, Dean E Schraufnagel, Mary Beth Scholand, Tracy Frech, Jerry A Molitor, Kristin Highland, Charles A Read, Marvin J Fritzler, Grace Hyun J Kim, Chi-Hong Tseng, Robert M Elashoff, Sclerodema Lung Study II Investigators, Donald P Tashkin, Michael D Roth, Philip J Clements, Daniel E Furst, Dinesh Khanna, Eric C Kleerup, Jonathan Goldin, Edgar Arriola, Elizabeth R Volkmann, Suzanne Kafaja, Richard Silver, Virginia Steen, Charlie Strange, Robert Wise, Fredrick Wigley, Maureen Mayes, David J Riley, Sabiha Hussain, Shervin Assassi, Vivien M Hsu, Bela Patel, Kristine Phillips, Fernando Martinez, Jeffrey Golden, M Kari Connolly, John Varga, Jane Dematte, Monique E Hinchcliff, Aryeh Fischer, Jeffrey Swigris, Richard Meehan, Arthur Theodore, Robert Simms, Suncica Volkov, Dean E Schraufnagel, Mary Beth Scholand, Tracy Frech, Jerry A Molitor, Kristin Highland, Charles A Read, Marvin J Fritzler, Grace Hyun J Kim, Chi-Hong Tseng, Robert M Elashoff, Sclerodema Lung Study II Investigators

Abstract

Background: 12 months of oral cyclophosphamide has been shown to alter the progression of scleroderma-related interstitial lung disease when compared with placebo. However, toxicity was a concern and without continued treatment the efficacy disappeared by 24 months. We hypothesised that a 2 year course of mycophenolate mofetil would be safer, better tolerated, and produce longer lasting improvements than cyclophosphamide.

Methods: This randomised, double-blind, parallel group trial enrolled patients from 14 US medical centres with scleroderma-related interstitial lung disease meeting defined dyspnoea, pulmonary function, and high-resolution CT (HRCT) criteria. The data coordinating centre at the University of California, Los Angeles (UCLA, CA, USA), randomly assigned patients using a double-blind, double-dummy, centre-blocked design to receive either mycophenolate mofetil (target dose 1500 mg twice daily) for 24 months or oral cyclophosphamide (target dose 2·0 mg/kg per day) for 12 months followed by placebo for 12 months. Drugs were given in matching 250 mg gel capsules. The primary endpoint, change in forced vital capacity as a percentage of the predicted normal value (FVC %) over the course of 24 months, was assessed in a modified intention-to-treat analysis using an inferential joint model combining a mixed-effects model for longitudinal outcomes and a survival model to handle non-ignorable missing data. The study was registered with ClinicalTrials.gov, number NCT00883129.

Findings: Between Sept 28, 2009, and Jan 14, 2013, 142 patients were randomly assigned to either mycophenolate mofetil (n=69) or cyclophosphamide (n=73). 126 patients (mycophenolate mofetil [n=63] and cyclophosphamide [n=63]) with acceptable baseline HRCT studies and at least one outcome measure were included in the primary analysis. The adjusted % predicted FVC improved from baseline to 24 months by 2·19 in the mycophenolate mofetil group (95% CI 0·53-3·84) and 2·88 in the cyclophosphamide group (1·19-4·58). The course of the % FVC did not differ significantly between the two treatment groups based on the prespecified primary analysis using a joint model (p=0·24), indicating that the trial was negative for the primary endpoint. However, in a post-hoc analysis of the primary endpoint, the within-treatment change from baseline to 24 months derived from the joint model showed that the % FVC improved significantly in both the mycophenolate mofetil and cyclophosphamide groups. 16 (11%) patients died (five [7%] mycophenolate mofetil and 11 [15%] cyclophosphamide), with most due to progressive interstitial lung disease. Leucopenia (30 patients vs four patients) and thrombocytopenia (four vs zero) occurred more often in patients given cyclophosphamide than mycophenolate mofetil. Fewer patients on mycophenolate mofetil than on cyclophosphamide prematurely withdrew from study drug (20 vs 32) or met prespecified criteria for treatment failure (zero vs two). The time to stopping treatment was shorter in the cyclophosphamide group (p=0·019).

Interpretation: Treatment of scleroderma-related interstitial lung disease with mycophenolate mofetil for 2 years or cyclophosphamide for 1 year both resulted in significant improvements in prespecified measures of lung function over the 2 year course of the study. Although mycophenolate mofetil was better tolerated and associated with less toxicity, the hypothesis that it would have greater efficacy at 24 months than cyclophosphamide was not confirmed. These findings support the potential clinical effectiveness of both cyclophosphamide and mycophenolate mofetil for progressive scleroderma-related interstitial lung disease, and the present preference for mycophenolate mofetil because of its better tolerability and toxicity profile.

Funding: National Heart, Lung and Blood Institute, National Institutes of Health; with drug supply provided by Hoffmann-La Roche and Genentech.

Copyright © 2016 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Disposition of study participants
Figure 2
Figure 2
Figure 2A. Primary Outcome; The course of %-predicted FVC from 3 through 24 months by treatment arm based on the joint model† †Adjustments for baseline FVC%-predicted, baseline HRCT lung fibrosis score (QLF) and non-ignorable missing data (time to premature discontinuation of study drug, deaths and treatment failure). The horizontal dotted line represents the average baseline FVC%-predicted for both treatment arms based on the joint model (baseline values did not differ between the two treatments). The vertical lines (dashed CYC; solid MMF) represent 95% confidence intervals. Of the 142 randomized participants, 126 (63 - CYC; 63- MMF) were included in the primary analysis. The 16 randomized subjects excluded from analysis did not have outcome assessments at ≥3 months or baseline HRCT scans that were suitable for quantitative assessment of the extent of lung fibrosis (a covariate in the joint model). Three and two participants assigned to CYC and MMF, respectively, who did not have a 3-month visit did have assessments at ≥6 month visits and were therefore included in the analysis. On the other hand, two and three participants assigned to CYC and MMF, respectively, who had a 3-month visit did not have a baseline HRCT scan that was suitable for quantitative assessment of the extent of fibrosis and were therefore excluded from the analysis. Figure 2B. Frequency distribution of changes from baseline to 24 months in FVC%-predicted by treatment arm and by whether participants completed the entire study treatment or not (all observed data, ITT). Figure 2C. Tolerability and Toxicity Assessment; Time to premature withdrawal from study medication or treatment failure by treatment arm. The protocol definition of treatment failure was an absolute decline in FVC% predicted of ≥15% that persisted for at least 1 month.
Figure 2
Figure 2
Figure 2A. Primary Outcome; The course of %-predicted FVC from 3 through 24 months by treatment arm based on the joint model† †Adjustments for baseline FVC%-predicted, baseline HRCT lung fibrosis score (QLF) and non-ignorable missing data (time to premature discontinuation of study drug, deaths and treatment failure). The horizontal dotted line represents the average baseline FVC%-predicted for both treatment arms based on the joint model (baseline values did not differ between the two treatments). The vertical lines (dashed CYC; solid MMF) represent 95% confidence intervals. Of the 142 randomized participants, 126 (63 - CYC; 63- MMF) were included in the primary analysis. The 16 randomized subjects excluded from analysis did not have outcome assessments at ≥3 months or baseline HRCT scans that were suitable for quantitative assessment of the extent of lung fibrosis (a covariate in the joint model). Three and two participants assigned to CYC and MMF, respectively, who did not have a 3-month visit did have assessments at ≥6 month visits and were therefore included in the analysis. On the other hand, two and three participants assigned to CYC and MMF, respectively, who had a 3-month visit did not have a baseline HRCT scan that was suitable for quantitative assessment of the extent of fibrosis and were therefore excluded from the analysis. Figure 2B. Frequency distribution of changes from baseline to 24 months in FVC%-predicted by treatment arm and by whether participants completed the entire study treatment or not (all observed data, ITT). Figure 2C. Tolerability and Toxicity Assessment; Time to premature withdrawal from study medication or treatment failure by treatment arm. The protocol definition of treatment failure was an absolute decline in FVC% predicted of ≥15% that persisted for at least 1 month.
Figure 2
Figure 2
Figure 2A. Primary Outcome; The course of %-predicted FVC from 3 through 24 months by treatment arm based on the joint model† †Adjustments for baseline FVC%-predicted, baseline HRCT lung fibrosis score (QLF) and non-ignorable missing data (time to premature discontinuation of study drug, deaths and treatment failure). The horizontal dotted line represents the average baseline FVC%-predicted for both treatment arms based on the joint model (baseline values did not differ between the two treatments). The vertical lines (dashed CYC; solid MMF) represent 95% confidence intervals. Of the 142 randomized participants, 126 (63 - CYC; 63- MMF) were included in the primary analysis. The 16 randomized subjects excluded from analysis did not have outcome assessments at ≥3 months or baseline HRCT scans that were suitable for quantitative assessment of the extent of lung fibrosis (a covariate in the joint model). Three and two participants assigned to CYC and MMF, respectively, who did not have a 3-month visit did have assessments at ≥6 month visits and were therefore included in the analysis. On the other hand, two and three participants assigned to CYC and MMF, respectively, who had a 3-month visit did not have a baseline HRCT scan that was suitable for quantitative assessment of the extent of fibrosis and were therefore excluded from the analysis. Figure 2B. Frequency distribution of changes from baseline to 24 months in FVC%-predicted by treatment arm and by whether participants completed the entire study treatment or not (all observed data, ITT). Figure 2C. Tolerability and Toxicity Assessment; Time to premature withdrawal from study medication or treatment failure by treatment arm. The protocol definition of treatment failure was an absolute decline in FVC% predicted of ≥15% that persisted for at least 1 month.
Figure 3
Figure 3
Figure 3A. Absolute change in modified Rodnan Skin Score (mRSS) from baseline by treatment arm based on the joint model† †Adjustments for baseline skin score (mRSS), baseline HRCT lung fibrosis score and non-ignorable missing data (time to premature discontinuation of study drug, deaths and treatment failure). Vertical lines represent 95% confidence intervals. Dotted horizontal line represents the average baseline skin score for both treatment arms based on the joint model (baseline values did not differ between the two treatments). Figure 3B. Frequency distribution of observed changes at 24 months from baseline in modified Rodnan Skin Score (mRSS). (observed data) Both limited and diffuse cutaneous SSc patients (N= 52 CYC; 53 MMF)
Figure 3
Figure 3
Figure 3A. Absolute change in modified Rodnan Skin Score (mRSS) from baseline by treatment arm based on the joint model† †Adjustments for baseline skin score (mRSS), baseline HRCT lung fibrosis score and non-ignorable missing data (time to premature discontinuation of study drug, deaths and treatment failure). Vertical lines represent 95% confidence intervals. Dotted horizontal line represents the average baseline skin score for both treatment arms based on the joint model (baseline values did not differ between the two treatments). Figure 3B. Frequency distribution of observed changes at 24 months from baseline in modified Rodnan Skin Score (mRSS). (observed data) Both limited and diffuse cutaneous SSc patients (N= 52 CYC; 53 MMF)

References

    1. Tyndall AJ, Bannert B, Vonk M, et al. Causes and risk factors for death in systemic sclerosis: a study from the EULAR Scleroderma Trials and Research (EUSTAR) database. Ann Rheum Dis. 2010;69:1809–15.
    1. Nihtyanova S, Schreiber BE, Ong VH, et al. Prediction of pulmonary complications and long-term survival in systemic sclerosis. Arthritis Rheumatol. 2014;66:1625–35.
    1. Tashkin DP, Elashoff R, Clements PJ, et al. Scleroderma Lung Study Research Group. Cyclophosphamide versus placebo in scleroderma lung disease. N Engl J Med. 2006;354:2655–66.
    1. Goldin J, Elashoff R, Kim HJ, et al. Treatment of scleroderma-interstitial lung disease with cyclophosphamide is associated with less progressive fibrosis on serial thoracic high-resolution CT scan than placebo: findings from the scleroderma lung study. Chest. 2009;136:1333–40.
    1. Kim HJ, Brown MS, Elashoff R, et al. Quantitative texture-based assessment of one-year changes in fibrotic reticular patterns on HRCT in scleroderma lung disease treated with oral cyclophosophamide. Eur Radiol. 2011;21:2455–65.
    1. Tashkin DP, Elashoff R, Clements PJ, et al. Scleroderma Lung Study Research Group. Effects of 1-year treatment with cyclophosphamide on outcomes at 2 years in scleroderma lung disease. Am J Respir Crit Care Med. 2007;176:1026–34.
    1. Furst DE, Tseng CH, Clements PJ, Strange C, Tashkin DP, Roth MD, Khanna D, Li N, Elashoff R, Schraufnagel DE Scleroderma Lung Study. Adverse events during the Scleroderma Lung Study. Am J Med. 2011;124:459–67.
    1. Martinez FJ, McCune JW. Cyclophosphamide for scleroderma lung disease. N Engl J Med. 2006;354:2707–9.
    1. Staatz CE, Tett SE. Pharmacology and toxicology of mycophenolate in organ transplant recipients: an update. Arch Toxicol. 2014;88:1351–89.
    1. Hahn BH, McMahon MA, Wilkinson A, et al. American college of rheumatology guidelines for screening, treatment and management of lupus nephritis. Arthritis Care Res. 2012;64:797–808.
    1. Nihtyanova SI, Brough GM, Black CM, Denton CP. Mycophenolate mofetil in diffuse cutaneous systemic sclerosis–a retrospective analysis. Rheumatology (Oxford) 2007;46:442–5.
    1. Gerbino AJ, Goss CH, Molitor JA. Effect of mycophenolate mofetil on pulmonary function in scleroderma-associated interstitial lung disease. Chest. 2008;133:455–60.
    1. Swigris JJ, Olson AL, Fischer A, Lynch DA, Cosgrove GP, Frankel SK, Meehan RT, Brown KK. Mycophenolate mofetil is safe, well tolerated and preserves lung function in patients with connective tissue disease-related interstitial lung disease. Chest. 2006;130:30–6.
    1. Zamora AC, Wolters PJ, Collard HR, et al. Use of mycophenolate mofetil to treat scleroderma-associated interstitial lung disease. Respir Med. 2008;102:150–55.
    1. Simeón-Aznar CP, Fonollosa-Plá V, Tolosa-Vilella C, et al. Effect of mycophenolate sodium in scleroderma-related interstitial lung disease. Clin Rheumatol. 2011;30:1393.
    1. Fischer A, Brown KK, DuBois RM, et al. Mycophenolate mofetil improves lung function in connective tissue disease-associated interstitial lung disease. J Rheumatol. 2013;40:640–6.
    1. Masi T Subcommittee for Scleroderma Criteria of the American Rheumatism Association Diagnostic and Therapeutic Criteria Committee. Preliminary criteria for the classification of systemic sclerosis (scleroderma) Arthritis Rheumatol. 1980;23:581–90.
    1. Mahler DA, Weinberg DH, Wells CK, Feinstein AR. The measurement of dyspnea. Contents, interobserver agreement and physiologic correlates of two new clinical indexes. Chest. 1984;85:751–758.
    1. Clements PJ, Lachenbruch PA, Seibold JR, et al. Skin thickness score in systemic sclerosis (SSc): An assessment of inter-observer variability in three independent studies. J Rheumatol. 1993;20:1892–6.
    1. Mahler DA, Ward J, Fierro-Carrion G, et al. Development of self-administered versions of modified baseline and transition dyspnea indexes in COPD. COPD: J of COPD. 2004;1:1–8.
    1. Kim HJ, Tashkin DP, Clements PJ, et al. A computer-aided diagnosis system for quantitative scoring of extent of lung fibrosis in scleroderma patients. Clin Exp Rheumatol. 2010;5:S26–35.
    1. Roth MD, Tseng CH, Clements PJ, et al. Scleroderma Lung Study Research Group. Predicting Lung function and treatment outcomes in scleroderma-related interstitial lung disease. Arthritis Rheumatism. 2011;63:2797–808.
    1. Li N, Elashoff RM, Li G, Tseng CH. Joint analysis of bivariate longitudinal ordinal outcomes and competing risks survival times with nonparametric distributions for random effects. Stat Med. 2012;31:1707–21.
    1. Elashoff RM, Li G, Li N. A joint model for longitudinal measurements and survival data in the presence of multiple failure types. Biometrics. 2008;64:762–771.
    1. Tashkin DP, Volkmann ER, Tseng CH, et al. Relationship between quantitative radiographic assessments of interstitial lung disease and physiological and clinical features of systemic sclerosis. Ann Rheum Dis. 2016;75:374–81.
    1. Khanna D, Furst DE, Hays RD, et al. Minimally important difference in diffuse systemic sclerosis: results of the D-penicillamine study. Ann Rheum Dis. 2006;65:1325–9.
    1. Khanna D, Tseng CH, Furst DE, et al. Minimally important differences in the Mahler’s Transition Dyspnoea Index in a large randomized controlled trial—results from the Scleroderma Lung Study. Rheumatology (Oxford) 2009;48:1537–40.
    1. Zheng Y, Li M, Zhang Y, Shi X, Li L, Jin M. The effects and mechanisms of mycophenolate mofetil on pulmonary arterial hypertension in rats. Rheumatol Int. 2010;30:341–8.
    1. Saketkoo LA, Lammi MR, Fischer A, Molitor J, Steen VD on behalf of the PHAROS Investigators. Observations from the pulmonary hypertension recognition and outcomes in scleroderma (Pharos) Cohort [Abstract] Ann Rheum Dis. 2015;74:820.
    1. Khanna D, Hayes RD, Maranian P, et al. Reliability and validity of the University of California, Los Angeles Scleroderma Clinical Trial Consortium Gastrointestinal Tract Instrument. Arthritis Rheum. 2009;61:1257–63.
    1. Bae S, Allanor Y, Furst DE, et al. Associations between a scleroderma-specific gastrointestinal instrument and objective tests of upper gastrointestinal involvements in systemic sclerosis. Clin Exp Rheumatol. 2013;31:57–63.

Source: PubMed

3
Předplatit