An open-label clinical trial of agalsidase alfa enzyme replacement therapy in children with Fabry disease who are naïve to enzyme replacement therapy

Ozlem Goker-Alpan, Nicola Longo, Marie McDonald, Suma P Shankar, Raphael Schiffmann, Peter Chang, Yinghua Shen, Arian Pano, Ozlem Goker-Alpan, Nicola Longo, Marie McDonald, Suma P Shankar, Raphael Schiffmann, Peter Chang, Yinghua Shen, Arian Pano

Abstract

Background: Following a drug manufacturing process change, safety/efficacy of agalsidase alfa were evaluated in enzyme replacement therapy (ERT)-naïve children with Fabry disease.

Methods: In an open-label, multicenter, Phase II study (HGT-REP-084; Shire), 14 children aged ≥7 years received 0.2 mg/kg agalsidase alfa every other week for 55 weeks. Primary endpoints: safety, changes in autonomic function (2-hour Holter monitoring). Secondary endpoints: estimated glomerular filtration rate, left ventricular mass index (LVMI), midwall fractional shortening, pharmacodynamic parameters, and patient-reported quality-of-life.

Results: Among five boys (median 10.2 [range 6.7, 14.4] years) and nine girls (14.8 [10.1, 15.9] years), eight patients experienced infusion-related adverse events (vomiting, n=4; nausea, n=3; dyspnea, n=3; chest discomfort, n=2; chills, n=2; dizziness, n=2; headache, n=2). One of these had several hypersensitivity episodes. However, no patient discontinued for safety reasons and no serious adverse events occurred. One boy developed immunoglobulin G (IgG) and neutralizing antidrug antibodies. Overall, no deterioration in cardiac function was observed in seven patients with low/abnormal SDNN (standard deviation of all filtered RR intervals; <100 ms) and no left ventricular hypertrophy: mean (SD) baseline SDNN, 81.6 (20.9) ms; mean (95% confidence interval [CI]) change from baseline to week 55, 17.4 (2.9, 31.9) ms. Changes in SDNN correlated with changes in LVMI (r=-0.975). No change occurred in secondary efficacy endpoints: mean (95% CI) change from baseline at week 55 in LVMI, 0.16 (-3.3, 3.7) g/m(2.7); midwall fractional shortening, -0.62% (-2.7%, 1.5%); estimated glomerular filtration rate, 0.15 (-11.4, 11.7) mL/min/1.73 m(2); urine protein, -1.8 (-6.0, 2.4) mg/dL; urine microalbumin, 0.6 (-0.5, 1.7) mg/dL; plasma globotriaosylceramide (Gb3), -5.71 (-10.8, -0.6) nmol/mL; urinary Gb3, -1,403.3 (-3,714.0, 907.4) nmol/g creatinine, or clinical quality-of-life outcomes.

Conclusion: Fifty-five weeks' agalsidase alfa ERT at 0.2 mg/kg every other week was well tolerated. Disease progression may be slowed when ERT is started prior to major organ dysfunction.

Trial registration: https://ClinicalTrials.gov identifier NCT01363492.

Keywords: Fabry disease; agalsidase alfa; efficacy; enzyme replacement therapy; pediatric study; safety.

Figures

Figure 1
Figure 1
Heart rate variability as assessed by SDNN: individual values at baseline and at 55 weeks. Notes: (A) Observed values of SDNN (ms). (B) SDNN Z-scores. SDNN represents the standard deviation of all filtered RR intervals for the length of the analysis. The different colored shapes within the figure show individual patient data at baseline and 55 weeks.
Figure 2
Figure 2
LVMI over time for the patient who had LVH at baseline. Note: SDNN represents the standard deviation of all filtered RR intervals for the length of the analysis. Abbreviations: LVH, left ventricular hypertrophy; LVMI, left ventricular mass index.
Figure 3
Figure 3
Correlation between change from baseline to week 55 in SDNN and LVMI in patients with baseline SDNN Notes: *r = Pearson correlation coefficient. SDNN represents the standard deviation of all filtered RR intervals for the length of the analysis. Abbreviation: LVMI, left ventricular mass index.
Figure 4
Figure 4
Mean (SE) change from baseline in eGFR. Note: The safety population comprises all enrolled patients who received at least one dose of agalsidase alfa. Abbreviations: eGFR, estimated glomerular filtration rate; SE, standard error; SP, safety population.
Figure 5
Figure 5
Mean plasma Gb3 over time for the overall population and by sex. Notes: (A) Observed values (nmol/mL). (B) Change from baseline. Abbreviation: Gb3, globotriaosylceramide.

References

    1. Garman SC, Garboczi DN. The molecular defect leading to Fabry disease: structure of human alpha-galactosidase. J Mol Biol. 2004;337(2):319–335.
    1. Germain DP. Fabry disease. Orphanet J Rare Dis. 2010;5:30.
    1. Schiffmann R, Kopp JB, Austin HA, 3rd, et al. Enzyme replacement therapy in Fabry disease: a randomized controlled trial. JAMA. 2001;285(21):2743–2749.
    1. Aerts JM, Groener JE, Kuiper S, et al. Elevated globotriaosylsphingosine is a hallmark of Fabry disease. Proc Natl Acad Sci U S A. 2008;105(8):2812–2817.
    1. Desnick RJ, Ioannou YA, Eng CM. α-Galactosidase A deficiency: Fabry disease. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The Metabolic and Molecular Basis of Inherited Disease. 8th ed. New York: McGraw-Hill; 2001. pp. 3733–3774.
    1. Wang RY, Lelis A, Mirocha J, Wilcox WR. Heterozygous Fabry women are not just carriers, but have a significant burden of disease and impaired quality of life. Genet Med. 2007;9(1):34–45.
    1. Wilcox WR, Oliveira JP, Hopkin RJ, et al. Females with Fabry disease frequently have major organ involvement: lessons from the Fabry Registry. Mol Genet Metab. 2008;93(2):112–128.
    1. Deegan PB, Baehner AF, Barba Romero MA, Hughes DA, Kampmann C, Beck M, European FOS Investigators Natural history of Fabry disease in females in the Fabry Outcome Survey. J Med Genet. 2006;43(4):347–352.
    1. MacDermot KD, Holmes A, Miners AH. Natural history of Fabry disease in affected males and obligate carrier females. J Inherit Metab Dis. 2001;24:13–14. discussion 11–12.
    1. Sestito S, Ceravolo F, Concolino D. Anderson-Fabry disease in children. Curr Pharm Des. 2013;19(33):6037–6045.
    1. Ries M, Gupta S, Moore DF, et al. Pediatric Fabry disease. Pediatrics. 2005;115(3):e344–e355.
    1. Ramaswami U, Whybra C, Parini R, et al. Clinical manifestations of Fabry disease in children: data from the Fabry Outcome Survey. Acta Paediatr. 2006;95(1):86–92.
    1. Kampmann C, Wiethoff CM, Whybra C, Baehner FA, Mengel E, Beck M. Cardiac manifestations of Anderson-Fabry disease in children and adolescents. Acta Paediatr. 2008;97(4):463–469.
    1. Ramaswami U, Wendt S, Pintos-Morell G, et al. Enzyme replacement therapy with agalsidase alfa in children with Fabry disease. Acta Paediatr. 2007;96(1):122–127.
    1. Schiffmann R, Martin RA, Reimschisel T, et al. Four-year prospective clinical trial of agalsidase alfa in children with Fabry disease. J Pediatr. 2010;156(5):832–837. e831.
    1. Ries M, Clarke JT, Whybra C, et al. Enzyme-replacement therapy with agalsidase alfa in children with Fabry disease. Pediatrics. 2006;118(3):924–932.
    1. Schiffmann R, Pastores GM, Lien YH, et al. Agalsidase alfa in pediatric patients with Fabry disease: a 6.5-year open-label follow-up study. Orphanet J Rare Dis. 2014;9(1):169.
    1. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use ICH Harmonised Tripartite Guideline: Comparability of Biotechnological/Biological Products Subject to Changes in their Manufacturing Process. 2004. [Accessed March 15, 2016]. Available from: .
    1. European Medicines Agency Replagal, agalsidase alfa. 2015. [Accessed February 22, 2016]. Available from: .
    1. World Health Organization Growth reference data for 5–19 years. 2007. [Accessed January 8, 2014]. Available from:
    1. Massin M, von Bernuth G. Normal ranges of heart rate variability during infancy and childhood. Pediatr Cardiol. 1997;18(4):297–302.
    1. Lang RM, Bierig M, Devereux RB, et al. Recommendations for chamber quantification: a report from the American Society of Echocardiography’s Guidelines and Standards Committee and the Chamber Quantification Writing Group, developed in conjunction with the European Association of Echocardiography, a branch of the European Society of Cardiology. J Am Soc Echocardiogr. 2005;18(12):1440–1463.
    1. National High Blood Pressure Education Program Working Group on High Blood Pressure in Children and Adolescents The fourth report on the diagnosis, evaluation, and treatment of high blood pressure in children and adolescents. Pediatrics. 2004;114(2 Suppl 4th Report):555–576.
    1. Kampmann C, Linhart A, Baehner F, et al. Onset and progression of the Anderson-Fabry disease related cardiomyopathy. Int J Cardiol. 2008;130(3):367–373.
    1. Charles S. Cleeland. The Brief Pain Inventory User Guide. 1991. [Accessed October 14, 2014]. Available from: .
    1. Furlong WJ, Feeny DH, Torrance GW, Health Utilities Inc Self-Complete Questionnaire Manual. 2012. [Accessed October 14, 2014]. Available from: .
    1. Healthactchq CHQ: Child Health Questionnaire™. 2014. [Accessed October 14, 2014]. Available from: .
    1. Crepaz R, Cemin R, Pedron C, Gentili L, Trevisan D, Pitscheider W. Age-related variations of left ventricular endocardial and midwall function in healthy infants, children, and adolescents. Ital Heart J. 2005;6(8):634–639.
    1. Morrissey RP, Philip KJ, Schwarz ER. Cardiac abnormalities in Anderson-Fabry disease and Fabry’s cardiomyopathy. Cardiovasc J Afr. 2011;22(1):38–44.
    1. Kleiger RE, Miller JP, Bigger JT, Jr, Moss AJ. Decreased heart rate variability and its association with increased mortality after acute myocardial infarction. Am J Cardiol. 1987;59(4):256–262.
    1. Zuanetti G, Neilson JM, Latini R, Santoro E, Maggioni AP, Ewing DJ. Prognostic significance of heart rate variability in post-myocardial infarction patients in the fibrinolytic era. The GISSI-2 results. Gruppo Italiano per lo Studio della Sopravvivenza nell’ Infarto Miocardico. Circulation. 1996;94(3):432–436.
    1. Mehta A, West ML, Pintos-Morell G, et al. Therapeutic goals in the treatment of Fabry disease. Genet Med. 2010;12(11):713–720.
    1. National Kidney Foundation I KDOQI Clinical Practice Guidelines and Clinical Practice Recommendations for Diabetes and Chronic Kidney Disease. 2007. [Accessed September 30, 2015]. Available from: .

Source: PubMed

3
Iratkozz fel