Clinical effect and safety profile of pegzilarginase in patients with arginase 1 deficiency

George A Diaz, Andreas Schulze, Markey C McNutt, Elisa Leão-Teles, J Lawrence Merritt 2nd, Gregory M Enns, Spyros Batzios, Allison Bannick, Roberto T Zori, Leslie S Sloan, Susan L Potts, Gillian Bubb, Anthony G Quinn, George A Diaz, Andreas Schulze, Markey C McNutt, Elisa Leão-Teles, J Lawrence Merritt 2nd, Gregory M Enns, Spyros Batzios, Allison Bannick, Roberto T Zori, Leslie S Sloan, Susan L Potts, Gillian Bubb, Anthony G Quinn

Abstract

Hyperargininemia in patients with arginase 1 deficiency (ARG1-D) is considered a key driver of disease manifestations, including spasticity, developmental delay, and seizures. Pegzilarginase (AEB1102) is an investigational enzyme therapy which is being developed as a novel arginine lowering approach. We report the safety and efficacy of intravenously (IV) administered pegzilarginase in pediatric and adult ARG1-D patients (n = 16) from a Phase 1/2 study (101A) and the first 12 weeks of an open-label extension study (102A). Substantial disease burden at baseline included lower-limb spasticity, developmental delay, and previous hyperammonemic episodes in 75%, 56%, and 44% of patients, respectively. Baseline plasma arginine (pArg) was elevated (median 389 μM, range 238-566) on standard disease management. Once weekly repeat dosing resulted in a median decrease of pArg of 277 μM after 20 cumulative doses (n = 14) with pArg in the normal range (40 to 115 μM) in 50% of patients at 168 hours post dose (mean pegzilarginase dose 0.10 mg/kg). Lowering pArg was accompanied by improvements in one or more key mobility assessments (6MWT, GMFM-D & E) in 79% of patients. In 101A, seven hypersensitivity reactions occurred in four patients (out of 162 infusions administered). Other common treatment-related adverse events (AEs) included vomiting, hyperammonemia, pruritus, and abdominal pain. Treatment-related serious AEs that occurred in five patients were all observed in 101A. Pegzilarginase was effective in lowering pArg levels with an accompanying clinical response in patients with ARG1-D. The improvements with pegzilarginase occurred in patients receiving standard treatment approaches, which suggests that pegzilarginase could offer benefit over existing disease management.

Trial registration: ClinicalTrials.gov NCT02488044 NCT03378531.

Keywords: ARG1-D; arginase 1 deficiency; human enzyme; hyperammonemia; hyperargininemia; pegzilarginase; spasticity.

Conflict of interest statement

G. A. D. has performed as a consultant and received travel and clinical study support from Aeglea. A. Schulze has received funds from Aeglea for the clinical study and for investigator‐initiated research. M. C. McNutt has received funds from Aeglea for clinical studies, honoraria, and travel reimbursement. E. Leão‐Teles has received financial support for educational activities and consulting and has received funds from Aeglea for the clinical study. J. L. Merritt II has received funds from Aeglea for the clinical study. G. M. Enns has received funds from Aeglea for the clinical study. S. Batzios has received funds from Aeglea for the clinical study, acted as a speaker on behalf of Aeglea. A. Bannick has received funds from Aeglea for the clinical study. R. T. Zori has performed as a consultant and received travel support and clinical study funding from Aeglea. L. S. Sloan, S. L. Potts, G. Bubb, and A. G. Quinn are employees of Aeglea BioTherapeutics Inc.

© 2020 The Authors. Journal of Inherited Metabolic Disease published by John Wiley & Sons Ltd on behalf of SSIEM.

Figures

FIGURE 1
FIGURE 1
Flowchart diagram of the 101A Phase 1/2 study and the 102A long‐term open label extension. *Two patients withdrew for personal reasons
FIGURE 2
FIGURE 2
Box plot demonstrating the minimum, first quartile, median, third quartile, and maximum values for, A, plasma arginine at baseline, 7 days after doses 1 and 8 in 101A part 2 and 7 days after doses 1 and 12 in 102A. B, Plasma GC levels for GAA (guanidinoacetic acid), NAA(N‐α‐acetyl‐L‐arginine), GVA (α‐keto‐δ‐guanidinovaleric acid), and ArgA (argininic acid) at baseline and dose 8 in 102A, and as median in open‐label extension study. Note: For arginine, Dose 20 (if available) or Dose 19 (if D20 was not available) was used for 102A. Ten patients had D20 and 4 patients had D19. For GC, the mean of all OLE visits from 102, that is, Doses 9 to 20 was used. ArgA, R,S‐argininic acid; BL, baseline; D, dose; GAA, guanidinoacetic acid; GVA, α‐keto‐δ‐guanidinovaleric acid; NAArg, Nα‐acetyl‐L‐arginine; OLE, mean of doses in 102A (open‐label extension); ULN, upper limit of normal, determined from study of GCs in healthy adults (n = 12), equal to 0.2, 3.7, 0.3, and 0.17 μM for ArgA, GAA, GVA, and NAArg, respectively
FIGURE 3
FIGURE 3
Heat map of baseline deficits in clinical outcomes assessments in all patients & overall clinical responses and individual assessment responses using three key complementary mobility assessments
FIGURE 4
FIGURE 4
Patient profiles outlining baseline deficits, therapeutic & disease history, and occurrence of hypersensitivity and ADA

References

    1. Cederbaum SD, Shaw KN, Spector EB, Verity MA, Snodgrass PJ, Sugarman GI. Hyperargininemia with arginase deficiency. Pediatr Res. 1979;13:827‐833.
    1. Schlune A, vom Dahl S, Häussinger D, Ensenauer R, Mayatepek E. Hyperargininemia due to arginase I deficiency: the original patients and their natural history, and a review of the literature. Amino Acids. 2015;47:1751‐1762.
    1. Sin YY, Baron G, Schulze A, Funk CD. Arginase‐1 deficiency. J Mol Med. 2015;93(12):1287‐1296.
    1. De Deyn P, Marescau B, Qureshi I, Mori A.Hyperargininemia: a treatable inborn error of metabolism? The Fourth International Symposium on Guanidino Compounds in Biology and Medicine: Guanidino Compounds in Biology & Medicine II; 1997: 53‐69.
    1. Scholl‐Bürgi S, Baumgartner Sigl S, Häberle J, et al. Amino acids in CSF and plasma in hyperammonaemic coma due to arginase1 deficiency. J Inherit Metab Dis. 2008;31(S2):S323‐S328.
    1. Carvalho DR, Brand GD, Brum JM, Takata RI, Speck‐Martins CE, Pratesi R. Analysis of novel ARG1 mutations causing hyperargininemia and correlation with arginase I activity in erythrocytes. Gene. 2012;509(1):124‐130.
    1. Waisbren SE, Cuthbertson D, Burgard P, Holbert A, McCarter R, Cederbaum S. Members of the urea cycle disorders consortium. Biochemical markers and neuropsychological functioning in distal urea cycle disorders. J Inherit Metab Dis. 2018;41(4):657‐667.
    1. Huemer M, Carvalho DR, Brum JM, et al. Clinical phenotype, biochemical profile, and treatment in 19 patients with arginase 1 deficiency. J Inherit Metab Dis. 2016;39(3):331‐340.
    1. Häberle J, Burlina A, Chakrapani A, et al. Suggested guidelines for the diagnosis and management of urea cycle disorders: first revision. J Inherit Metab Dis. 2019;42(6):1192‐1230.
    1. Cederbaum SD, Moedjono SJ, Shaw KN, Carter M, Naylor E, Walzer M. Treatment of hyperargininaemia due to arginase deficiency with a chemically defined diet. J Inherit Metab Dis. 1982;5:95‐99.
    1. Lambert MA, Marescau B, Desjardins M, et al. Hyperargininemia: intellectual and motor improvement related to changes in biochemical data. J Pediatr. 1991;118(3):421‐424.
    1. Desnick RJ, Schuchman EH. Enzyme replacement therapy for lysosomal diseases: lessons from 20 years of experience and remaining challenges. Annu Rev Genomics Hum Genet. 2012;13:307‐335.
    1. Burrage LC, Sun Q, Elsea SH, et al. Human recombinant arginase enzyme reduces plasma arginine in mouse models of arginase deficiency. Hum Mol Genet. 2015;24(22):6417‐6427.
    1. Haller C, Song W, Cimms T, et al. Individual heat map assessments demonstrate vestronidase alfa treatment response in a highly heterogeneous mucopolysaccharidosis VII study population. JIMD Rep. 2019;49(1):53‐62.
    1. NIH . ToolboxR Scoring and Interpretation Guide for the iPad; 2016.
    1. Schrover R, Evans K, Giugliani R, Noble I, Bhattacharya K. Minimal clinically important difference for the 6‐min walk test: literature review and application to Morquio a syndrome. Orphanet J Rare Dis. 2017;12(1):78.
    1. Oeffinger D, Bagley A, Rogers S, et al. Outcome tools used for ambulatory children with cerebral palsy: responsiveness and minimum clinically important differences. Dev Med Child Neurol. 2008;50(12):918‐925.
    1. Wu G, Morris SM. Arginine metabolism: nitric oxide and beyond. Biochem J. 1998;336(Pt 1):1‐17.
    1. Sakiyama T, Nakabayashi H, Shimizu H, Kondo W, Kodama S, Kitagawa T. A successful trial of enzyme replacement therapy in a case of argininemia. Tohoku J Exp Med. 1984;142(3):239‐248.
    1. Deshmukh DR, Sarnaik AP, Marescau B, et al. Guanidino compound metabolism in arginine‐free diet induced hyperammonemia. Enzyme. 1991;45(3):128‐136.
    1. Geiger R, Strasak A, Treml B, et al. Six‐minute walk test in children and adolescents. J Pediatr. 2007;150(4):395‐399.

Source: PubMed

3
Předplatit