One-year results of a clinical trial of olipudase alfa enzyme replacement therapy in pediatric patients with acid sphingomyelinase deficiency

George A Diaz, Simon A Jones, Maurizio Scarpa, Karl Eugen Mengel, Roberto Giugliani, Nathalie Guffon, Isabela Batsu, Patricia A Fraser, Jing Li, Qi Zhang, Catherine Ortemann-Renon, George A Diaz, Simon A Jones, Maurizio Scarpa, Karl Eugen Mengel, Roberto Giugliani, Nathalie Guffon, Isabela Batsu, Patricia A Fraser, Jing Li, Qi Zhang, Catherine Ortemann-Renon

Abstract

Purpose: To assess olipudase alfa enzyme replacement therapy for non-central nervous system manifestations of acid sphingomyelinase deficiency (ASMD) in children.

Methods: This phase 1/2, international, multicenter, open-label trial (ASCEND-Peds/NCT02292654) administered intravenous olipudase alfa every 2 weeks with intrapatient dose escalation to 3 mg/kg. Primary outcome was safety through week 64. Secondary outcomes included pharmacokinetics, spleen and liver volumes, lung diffusing capacity (DLCO), lipid profiles, and height through week 52.

Results: Twenty patients were enrolled: four adolescents (12-17 years), nine children (6-11 years), and seven infants/early child (1-5 years). Most adverse events were mild or moderate, including infusion-associated reactions (primarily urticaria, pyrexia, and/or vomiting) in 11 patients. Three patients had serious treatment-related events: one with transient asymptomatic alanine aminotransferase increases, another with urticaria and rash (antidrug antibody positive [ADA+]), and a third with an anaphylactic reaction (ADA+) who underwent desensitization and reached the 3 mg/kg maintenance dose. Mean splenomegaly and hepatomegaly improved by >40% (p < 0.0001). Mean % predicted DLCO improved by 32.9% (p = 0.0053) in patients able to perform the test. Lipid profiles and elevated liver transaminase levels normalized. Mean height Z-scores improved by 0.56 (p < 0.0001).

Conclusion: In this study in children with chronic ASMD, olipudase alfa was generally well-tolerated with significant, comprehensive improvements in disease pathology across a range of clinically relevant endpoints.

Conflict of interest statement

George A. Diaz, Simon A. Jones, Maurizio Scarpa, K. Eugen Mengel, Roberto Giugliani, and Nathalie Guffon are principal investigators in Sanofi-Genzyme sponsored trials. George A. Diaz and Simon A. Jones have received honoraria and consulting fees from Sanofi Genzyme. K. Eugen Mengel has received research support, honoraria, and consulting fees from Sanofi Genzyme. Roberto Giugliani has received honoraria, consulting fees, speaker fees and travel reimbursement from Sanofi Genzyme. Nathalie Guffon has received honoraria and travel reimbursement from Sanofi Genzyme. Maurizio Scarpa has received honoraria, consulting fees, speaker fees and travel reimbursement from Sanofi Genzyme. Isabela Batsu, Patricia A. Fraser, Jing Li, Qi Zhang, and Catherine Ortemann-Renon are/were employees of Sanofi Genzyme.

© 2021. The Author(s).

Figures

Fig. 1. Patient Disposition in the Trial.
Fig. 1. Patient Disposition in the Trial.
Patient disposition.
Fig. 2. Safety Biomarker and Pharmacodynamic Parameters.
Fig. 2. Safety Biomarker and Pharmacodynamic Parameters.
Plasma levels of ceramide, lyso-sphingomyelin, and chitotriosidase activity during treatment with olipudase alfa. (a) Mean (±SD) plasma levels of ceramide for the overall patient population preinfusion, 24 hours and 48 hours after olipudase alfa infusions. Normal range for plasma ceramide was 1.3–3.3 mg/L. 24 hour postinfusion levels were assessed in the adolescent group only.  (b) Mean (±SD) plasma levels of lyso-sphingomyelin for the overall patient population preinfusion, 24 hours and 48 hours after olipudase alfa infusions. Upper limit of normal for lyso-sphingomyelin is 9.99 μg/L. Twenty-four hour postinfusion levels were assessed in the adolescent group only. (c) Mean (±SD) normalized plasma activity of chitotriosidase for the overall patient population. Upper limit of normal is 181 μmol/L/hr.
Fig. 3. Changes over time in efficacy…
Fig. 3. Changes over time in efficacy assessments (hepatosplenomegaly, liver transaminases, lung diffusion capacity, plasma lipids, and height) with olipudase alfa treatment.
(a, b) Individual patient responses for spleen volumes in multiples of normal (MN) and the percent change from baseline for spleen volumes in least square mean (LSM) ± standard error of the mean (SEM), respectively at 6 months and 1 year. Cutoffs of MN for severity categories are indicated by shading. (c,d) Individual patient responses for liver volumes in MN and the percent change from baseline for spleen and liver volumes in LSM ± SEM, respectively at 6 months and 1 year. Cutoffs of MN for severity categories are indicated by shading. (e,f) Mean (±SD) preinfusion plasma levels for aspartate aminotransferase (AST) and alanine aminotransferase (ALT), respectively, for the overall patient population and the means for each age group at baseline and throughout treatment with olipudase alfa. The AST and ALT upper limit of normal (ULN) for the for the adolescent, child, and infant/early child groups is 40, 59, and 69 IU/L and 43, 34, and 34 IU/L, respectively (values the same in U/L). Ranges of ULN are indicated by gray boxes. (g, h) individual patient responses and LSM ± SEM percent change from baseline for the percent predicted DLCO adjusted for hemoglobin. Cutoffs for gas exchange impairment are indicated. (i) Plasma lipid mean (±SD) for the overall population and means for each age group for total cholesterol, low density lipid cholesterol (LDL-C), high density lipid cholesterol (HDL-C), and triglycerides (TG). Low to high ranges of normal for total cholesterol: 4.4–5.15 mmol/L ( <170–199 mg/dL). Low to high ranges of normal for LDL-C: adolescent: 1.60 to 3.52 mmol/L, child: 1.63 to 3.63 mmol/L, infant/early child: 0.98 to 3.63 mmol/L (38–140 mg/dL); for TG: adolescent: 0.36 to 1.67 mmol/L, child: 0.34 to 1.48 mmol/L, infant/early child: 0.34 to 1.24 mmol/L (32–149 mg/dL); for HDL-C: adolescent: 0.78 to 1.92 mmol/L, child: 0.93 to 1.94 mmol/L, infant/early child: not reported (30–75 mg/dL). (j) Individual patient height Z-scores (graph) and the LSM changes from baseline with p values (inset table) over time.

References

    1. Bernadette B, Konrad S. Lysosomal glycosphingolipid storage diseases. Ann. Rev. Biochem. 2019;88:461–485. doi: 10.1146/annurev-biochem-013118-111518.
    1. Schuchman EH, Desnick RJ. Types A and B Niemann–Pick disease. Mol. Genet. Metab. 2017;120:27–33. doi: 10.1016/j.ymgme.2016.12.008.
    1. Wasserstein MP, et al. The natural history of type B Niemann–Pick disease: results from a 10-year longitudinal study. Pediatrics. 2004;114:e672–677. doi: 10.1542/peds.2004-0887.
    1. McGovern MM, et al. Lipid abnormalities in children with types A and B Niemann Pick disease. J. Pediatr. 2004;145:77–81. doi: 10.1016/j.jpeds.2004.02.048.
    1. Kingma SD, Bodamer OA, Wijburg FA. Epidemiology and diagnosis of lysosomal storage disorders; challenges of screening. Best Pract. Res. Clin. Endocrinol. Metab. 2015;29:145–157. doi: 10.1016/j.beem.2014.08.004.
    1. McGovern MM, Aron A, Brodie SE, Desnick RJ, Wasserstein MP. Natural history of type A Niemann–Pick disease: possible endpoints for therapeutic trials. Neurology. 2006;66:228–232. doi: 10.1212/01.wnl.0000194208.08904.0c.
    1. Wasserstein MP, Aron A, Brodie SE, Simonaro C, Desnick RJ, McGovern MM. Acid sphingomyelinase deficiency: prevalence and characterization of an intermediate phenotype of Niemann–Pick disease. J. Pediatr. 2006;149:554–559. doi: 10.1016/j.jpeds.2006.06.034.
    1. Cassiman D, et al. Cause of death in patients with chronic visceral and chronic neurovisceral acid sphingomyelinase deficiency (Niemann–Pick disease type B and B variant): literature review and report of new cases. Mol. Genet. Metab. 2016;118:206–213. doi: 10.1016/j.ymgme.2016.05.001.
    1. Barczykowski AL, Foss AH, Duffner PK, Yan L, Carter RL. Death rates in the U.S. due to Krabbe disease and related leukodystrophy and lysosomal storage diseases. Am. J. Med. Genet. A. 2012;158a:2835–2842. doi: 10.1002/ajmg.a.35624.
    1. McGovern MM, Lippa N, Bagiella E, Schuchman EH, Desnick RJ, Wasserstein MP. Morbidity and mortality in type B Niemann–Pick disease. Genet. Med. 2013;15:618–623. doi: 10.1038/gim.2013.4.
    1. Wasserstein, M. et al. Recommendations for clinical monitoring of patients with acid sphingomyelinase deficiency (ASMD). Mol. Genet. Metab.126, 98–105 (2019).
    1. Wasserstein MP, et al. Successful within-patient dose escalation of olipudase alfa in acid sphingomyelinase deficiency. Mol. Genet. Metab. 2015;116:88–97. doi: 10.1016/j.ymgme.2015.05.013.
    1. Wasserstein MP, et al. Olipudase alfa for treatment of acid sphingomyelinase deficiency (ASMD): safety and efficacy in adults treated for 30 months. J. Inherit. Metab. Dis. 2018;41:829–838. doi: 10.1007/s10545-017-0123-6.
    1. Mendelson DS, et al. Type B Niemann–Pick disease: findings at chest radiography, thin-section CT, and pulmonary function testing. Radiology. 2006;238:339–345. doi: 10.1148/radiol.2381041696.
    1. Miller MR, et al. Standardisation of spirometry. Eur. Respir. J. 2005;26:319–338. doi: 10.1183/09031936.05.00034805.
    1. Crapo RO, Morris AH. Standardized single breath normal values for carbon monoxide diffusing capacity. Am. Rev. Respir. Dis. 1981;123:185–189.
    1. NGuyen, L.-P., Harper, R. & Louie, S. Using and interpreting carbon monoxide diffusing capacity (DLco) correctly. Consultant. 56, 440–445 (2016).
    1. Pellegrino R, et al. Interpretative strategies for lung function tests. Eur. Respir. J. 2005;26:948–968. doi: 10.1183/09031936.05.00035205.
    1. Pastores GM, et al. Therapeutic goals in the treatment of Gaucher disease. Semin. Hematol. 2004;41:4–14. doi: 10.1053/j.seminhematol.2004.07.009.
    1. Chuang WL, et al. Lyso-sphingomyelin is elevated in dried blood spots of Niemann–Pick B patients. Mol. Genet. Metab. 2014;111:209–211. doi: 10.1016/j.ymgme.2013.11.012.
    1. McGovern MM, Avetisyan R, Sanson BJ, Lidove O. Disease manifestations and burden of illness in patients with acid sphingomyelinase deficiency (ASMD) Orphanet J. Rare Dis. 2017;12:41. doi: 10.1186/s13023-017-0572-x.
    1. von Ranke FM, et al. Pulmonary involvement in Niemann–Pick disease: a state-of-the-art review. Lung. 2016;194:511–518. doi: 10.1007/s00408-016-9893-0.
    1. Wasserstein MP, Larkin AE, Glass RB, Schuchman EH, Desnick RJ, McGovern MM. Growth restriction in children with type B Niemann–Pick disease. J. Pediatr. 2003;142:424–428. doi: 10.1067/mpd.2003.113.
    1. Labrune P, Bedossa P, Huguet P, Roset F, Vanier MT, Odievre M. Fatal liver failure in two children with Niemann–Pick disease type B. J. Pediatr. Gastroenterol. Nutr. 1991;13:104–109. doi: 10.1097/00005176-199107000-00020.
    1. McGovern MM, et al. A prospective, cross-sectional survey study of the natural history of Niemann–Pick disease type B. Pediatrics. 2008;122:e341–349. doi: 10.1542/peds.2007-3016.
    1. Richards SM. Immunologic considerations for enzyme replacement therapy in the treatment of lysosomal storage disorders. Clin. Appl. Immunol. Rev. 2002;2:241–253. doi: 10.1016/S1529-1049(02)00049-1.
    1. Kim MH, et al. Hepatic inflammatory cytokine production can be regulated by modulating sphingomyelinase and ceramide synthase 6. Int. J. Mol. Med. 2017;39:453–462. doi: 10.3892/ijmm.2016.2835.
    1. Maceyka M, Spiegel S. Sphingolipid metabolites in inflammatory disease. Nature. 2014;510:58–67. doi: 10.1038/nature13475.
    1. McGovern M, et al. Novel first-dose adverse drug reactions during a phase 1 trial of recombinant human acid sphingomyelinase (rhASM) in adults with Niemann–Pick disease type B (acid sphingomyelinase deficiency) Genet. Med. 2016;18:34–40. doi: 10.1038/gim.2015.24.
    1. Murray JM, et al. Nonclinical safety assessment of recombinant human acid sphingomyelinase (rhASM) for the treatment of acid sphingomyelinase deficiency: the utility of animal Models of disease in the toxicological evaluation of potential therapeutics. Mol. Genet. Metab. 2014;114:217–225. doi: 10.1016/j.ymgme.2014.07.005.
    1. Xue L, et al. Recommendations for the assessment and Management of pre-existing drug-reactive antibodies during biotherapeutic development. AAPS J. 2017;19:1576–1586. doi: 10.1208/s12248-017-0153-x.
    1. Xaubet A, et al. Erratum to "Guidelines for the medical treatment of idiopathic pulmonary fibrosis" <[Arch Bronconeumol. 53 (2017) 263-9] >. Arch. Bronconeumol. 2017;53:657–658.
    1. Cottin V, et al. French practical guidelines for the diagnosis and management of idiopathic pulmonary fibrosis—2017 update. . Rev. Mal. Respir. 2017;34:900–968. doi: 10.1016/j.rmr.2017.07.017.
    1. Khanna D, et al. Connective tissue disease-associated interstitial lung diseases (CTD-ILD)—report from OMERACT CTD-ILD Working Group. J. Rheumatol. 2015;42:2168–2171. doi: 10.3899/jrheum.141182.

Source: PubMed

3
Se inscrever