The validation of pharmacogenetics for the identification of Fabry patients to be treated with migalastat

Elfrida R Benjamin, Maria Cecilia Della Valle, Xiaoyang Wu, Evan Katz, Farhana Pruthi, Sarah Bond, Benjamin Bronfin, Hadis Williams, Julie Yu, Daniel G Bichet, Dominique P Germain, Roberto Giugliani, Derralynn Hughes, Raphael Schiffmann, William R Wilcox, Robert J Desnick, John Kirk, Jay Barth, Carrolee Barlow, Kenneth J Valenzano, Jeff Castelli, David J Lockhart, Elfrida R Benjamin, Maria Cecilia Della Valle, Xiaoyang Wu, Evan Katz, Farhana Pruthi, Sarah Bond, Benjamin Bronfin, Hadis Williams, Julie Yu, Daniel G Bichet, Dominique P Germain, Roberto Giugliani, Derralynn Hughes, Raphael Schiffmann, William R Wilcox, Robert J Desnick, John Kirk, Jay Barth, Carrolee Barlow, Kenneth J Valenzano, Jeff Castelli, David J Lockhart

Abstract

Purpose: Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the α-galactosidase A gene. Migalastat, a pharmacological chaperone, binds to specific mutant forms of α-galactosidase A to restore lysosomal activity.

Methods: A pharmacogenetic assay was used to identify the α-galactosidase A mutant forms amenable to migalastat. Six hundred Fabry disease-causing mutations were expressed in HEK-293 (HEK) cells; increases in α-galactosidase A activity were measured by a good laboratory practice (GLP)-validated assay (GLP HEK/Migalastat Amenability Assay). The predictive value of the assay was assessed based on pharmacodynamic responses to migalastat in phase II and III clinical studies.

Results: Comparison of the GLP HEK assay results in in vivo white blood cell α-galactosidase A responses to migalastat in male patients showed high sensitivity, specificity, and positive and negative predictive values (≥0.875). GLP HEK assay results were also predictive of decreases in kidney globotriaosylceramide in males and plasma globotriaosylsphingosine in males and females. The clinical study subset of amenable mutations (n = 51) was representative of all 268 amenable mutations identified by the GLP HEK assay.

Conclusion: The GLP HEK assay is a clinically validated method of identifying male and female Fabry patients for treatment with migalastat.Genet Med 19 4, 430-438.

Trial registration: ClinicalTrials.gov NCT00214500 NCT00283959 NCT00283933 NCT00304512 NCT00925301 NCT01218659.

Figures

Figure 1
Figure 1
Substrate changes following migalastat treatment by amenability category. (a) (top): Kidney interstitial capillary GL-3 absolute change after 6 months of treatment with migalastat grouped by amenability category. Study 011 combines male and female patients receiving 6 months of migalastat (baseline to month 6 for patients randomized to migalastat at baseline, plus month 6 to month 12 for male patients receiving placebo from baseline to month 6). IC GL-3: mean number of GL-3 inclusions per interstitial capillary. Baseline refers to the kidney IC GL-3 value during the study 011 baseline visit (visit 1) for patients treated with migalastat during stages 1 and 2; it refers to the kidney IC GL-3 value at month 6 for patients treated with placebo during stage 1 and migalastat during stage 2. The mean (minimum, maximum) baseline IC GL-3 values for patients with amenable and nonamenable mutations were 0.646 (0.013, 5.692; n = 42) and 1.350 (0.117, 5.248; n = 14), respectively. After 6 months of treatment, the mean changes (95% confidence interval) from baseline for patients with amenable and nonamenable mutations were −0.282 (−0.455, −0.109; n = 42) and 0.324 (−0.105, 0.752; n = 14), respectively. The mean difference (95% confidence interval) in the change from baseline after 6 months (amenable minus nonamenable) was −0.606 (−1.059, −0.153). (b) (bottom): Plasma lyso-Gb3 absolute change after 6 months of treatment with migalastat grouped by amenability category. Study 011 combines male and female patients receiving 6 months of migalastat (baseline to month 6 for patients randomized to migalastat at baseline, plus month 6 to month 12 for patients receiving placebo from baseline to month 6). Baseline refers to the plasma lyso-Gb3 value during the study 011 baseline visit (visit 1) for patients treated with migalastat during stage 1 and stage 2; it refers to the kidney IC GL-3 value at month 6 in patients treated with placebo during stage 1 and migalastat during stage 2. The mean (minimum, maximum) baseline plasma lyso-Gb3 values for patients with amenable and nonamenable mutations were 45.236 (1.187, 218.333; n = 31) and 74.396 (9.720, 249.333; n = 13), respectively. After 6 months of treatment, the mean changes (95% confidence interval) from baseline for patients with amenable and nonamenable mutations were −13.011 (−20.642, −5.380; n = 31) and 9.801 (−1.6478, 21.2498; n = 13), respectively. The mean difference (95% confidence interval) in the change from baseline after 6 months (amenable minus nonamenable) was −22.812 (−36.100, −9.524).
Figure 2
Figure 2
Plasma lyso-Gb3 absolute change from baseline after treatment with migalastat grouped by GLA mutation category. Study 012 changes from baseline in patients with amenable and nonamenable GLA mutations. Blue dotted lines represent zero change from baseline. Left: Data points represent the mean, error bars represent the standard deviation, and values in parentheses represent the number (n) of patients with amenable mutations. At month 18, in patients with amenable mutations, the mean changes (95% confidence interval) from baseline for migalastat and ERT were 1.728 (−0.301, 3.758) and −1.926 (−4.632, 0.781), respectively. Right: Data points are from individual patients with nonamenable mutations. Lines represent the mean, values in parentheses represent the number (n) of patients with nonamenable mutations. Due to small sample sizes, no statistical comparisons were made for patients with nonamenable mutations. Data are based on patients with available samples for this analysis.

References

    1. Brady RO, Gal AE, Bradley RM, Martensson E, Warshaw AL, Laster L. Enzymatic defect in Fabry's disease. Ceramidetrihexosidase deficiency. N Engl J Med 1967;276:1163–1167.
    1. Germain DP. Fabry disease. Orphanet J Rare Dis 2010;5:30.
    1. Gal A, Schäfer E, Rohard I. The genetic basis of Fabry disease. In: Mehta A, Beck M, Sunder-Plassmann G (eds). Fabry Disease: Perspectives From 5 Years of FOS. Oxford PharmaGenesis: Oxford, UK, 2006.
    1. Germain DP, Shabbeer J, Cotigny S, Desnick RJ. Fabry disease: twenty novel alpha-galactosidase A mutations and genotype-phenotype correlations in classical and variant phenotypes. Mol Med 2002;8:306–312.
    1. Filoni C, Caciotti A, Carraresi L, et al. Functional studies of new GLA gene mutations leading to conformational Fabry disease. Biochim Biophys Acta 2010;1802:247–252.
    1. Topaloglu AK, Ashley GA, Tong B, et al. Twenty novel mutations in the alpha-galactosidase A gene causing Fabry disease. Mol Med 1999;5:806–811.
    1. Shabbeer J, Yasuda M, Benson SD, Desnick RJ. Fabry disease: identification of 50 novel alpha-galactosidase A mutations causing the classic phenotype and three-dimensional structural analysis of 29 missense mutations. Hum Genomics 2006;2:297–309.
    1. Khanna R, Soska R, Lun Y, et al. The pharmacological chaperone 1-deoxygalactonojirimycin reduces tissue globotriaosylceramide levels in a mouse model of Fabry disease. Mol Ther 2010;18:23–33.
    1. Yam GH, Zuber C, Roth J. A synthetic chaperone corrects the trafficking defect and disease phenotype in a protein misfolding disorder. FASEB J 2005;19:12–18.
    1. Fan JQ, Ishii S, Asano N, Suzuki Y. Accelerated transport and maturation of lysosomal alpha-galactosidase A in Fabry lymphoblasts by an enzyme inhibitor. Nat Med 1999;5:112–115.
    1. Benjamin ER, Flanagan JJ, Schilling A, et al. The pharmacological chaperone 1-deoxygalactonojirimycin increases alpha-galactosidase A levels in Fabry patient cell lines. J Inherit Metab Dis 2009;32:424–440.
    1. Yam GH, Bosshard N, Zuber C, Steinmann B, Roth J. Pharmacological chaperone corrects lysosomal storage in Fabry disease caused by trafficking-incompetent variants. Am J Physiol Cell Physiol 2006;290:C1076–C1082.
    1. Lemansky P, Bishop DF, Desnick RJ, Hasilik A, von Figura K. Synthesis and processing of alpha-galactosidase A in human fibroblasts. Evidence for different mutations in Fabry disease. J Biol Chem 1987;262:2062–2065.
    1. Shin SH, Murray GJ, Kluepfel-Stahl S, et al. Screening for pharmacological chaperones in Fabry disease. Biochem Biophys Res Commun 2007;359:168–173.
    1. Ishii S, Kase R, Okumiya T, Sakuraba H, Suzuki Y. Aggregation of the inactive form of human alpha-galactosidase in the endoplasmic reticulum. Biochem Biophys Res Commun 1996;220:812–815.
    1. Wu X, Katz E, Della Valle MC, et al. A pharmacogenetic approach to identify mutant forms of α-galactosidase A that respond to a pharmacological chaperone for Fabry disease. Hum Mutat 2011;32:965–977.
    1. Lukas J, Giese AK, Markoff A, et al. Functional characterisation of alpha-galactosidase a mutations as a basis for a new classification system in fabry disease. PLoS Genet 2013;9:e1003632.
    1. Echevarria L, Benistan K, Toussaint A, et al. X-chromosome inactivation in female patients with Fabry disease. Clin Genet 2016;89:44–54.
    1. Johnson FK, Mudd PN Jr, Bragat A, Adera M, Boudes P. Pharmacokinetics and safety of migalastat HCl and effects on agalsidase activity in healthy volunteers. Clin Pharmacol Drug Dev 2013;2:120–132.
    1. Conzelmann E, Sandhoff K. Partial enzyme deficiencies: residual activities and the development of neurological disorders. Dev Neurosci 1983;6:58–71.
    1. Desnick RJ. Enzyme replacement and enhancement therapies for lysosomal diseases. J Inherit Metab Dis 2004;27:385–410.
    1. Germain DP, Giugliani R, Hughes DA, et al. Safety and pharmacodynamic effects of a pharmacological chaperone on α-galactosidase A activity and globotriaosylceramide clearance in Fabry disease: report from two phase 2 clinical studies. Orphanet J Rare Dis 2012;7:91.
    1. Giugliani R, Waldek S, Germain DP, et al. A Phase 2 study of migalastat hydrochloride in females with Fabry disease: selection of population, safety and pharmacodynamic effects. Mol Genet Metab 2013;109:86–92.
    1. Germain DP, Hughes DA, Nicholls K, et al. Treatment of Fabry's disease with the pharmacologic chaperone Migalastat. N Engl J Med 2016;375:545–555.
    1. Barisoni L, Jennette JC, Colvin R, et al. Novel quantitative method to evaluate globotriaosylceramide inclusions in renal peritubular capillaries by virtual microscopy in patients with fabry disease. Arch Pathol Lab Med 2012;136:816–824.
    1. Altman DG, Bland JM. Statistics Notes: diagnostic tests 1: sensitivity and specificity. BMJ 1994;308:1552.
    1. Altman DG, Bland JM. Statistics Notes: diagnostic tests 2: predictive values. BMJ 1994;309:102.
    1. Eng CM, Banikazemi M, Gordon RE, et al. A phase ½ clinical trial of enzyme replacement in fabry disease: pharmacokinetic, substrate clearance, and safety studies. Am J Hum Genet 2001;68:711–722.
    1. Rombach SM, Dekker N, Bouwman MG, et al. Plasma globotriaosylsphingosine: diagnostic value and relation to clinical manifestations of Fabry disease. Biochim Biophys Acta 2010;1802:741–748.
    1. van Breemen MJ, Rombach SM, Dekker N, et al. Reduction of elevated plasma globotriaosylsphingosine in patients with classic Fabry disease following enzyme replacement therapy. Biochim Biophys Acta 2011;1812:70–76.
    1. Ries M, Gal A. Genotype-phenotype correlation in Fabry disease. In: Mehta A, Beck M, Sunder-Plassmann G (eds). Fabry Disease: Perspectives From 5 Years of FOS. Oxford PharmaGenesis: Oxford, UK, 2006.
    1. Altarescu G, Moore DF, Schiffmann R. Effect of genetic modifiers on cerebral lesions in Fabry disease. Neurology 2005;64:2148–2150.
    1. Desnick RJ, Brady R, Barranger J, et al. Fabry disease, an under-recognized multisystemic disorder: expert recommendations for diagnosis, management, and enzyme replacement therapy. Ann Intern Med 2003;138:338–346.
    1. Wilcox WR, Oliveira JP, Hopkin RJ, et al.; Fabry Registry. Females with Fabry disease frequently have major organ involvement: lessons from the Fabry Registry. Mol Genet Metab 2008;93:112–128.

Source: PubMed

3
Subskrybuj