[PDF][PDF] Fabry disease: preclinical studies demonstrate the effectiveness of α-galactosidase A replacement in enzyme-deficient mice

YA Ioannou, KM Zeidner, RE Gordon… - The American Journal of …, 2001 - cell.com
YA Ioannou, KM Zeidner, RE Gordon, RJ Desnick
The American Journal of Human Genetics, 2001cell.com
Preclinical studies of enzyme-replacement therapy for Fabry disease (deficient α-
galactosidase A [α-Gal A] activity) were performed in α-Gal A–deficient mice. The
pharmacokinetics and biodistributions were determined for four recombinant human α-Gal A
glycoforms, which differed in sialic acid and mannose-6-phosphate content. The plasma half-
lives of the glycoforms were∼ 2–5 min, with the more sialylated glycoforms circulating
longer. After intravenous doses of 1 or 10 mg/kg body weight were administered, each …
Preclinical studies of enzyme-replacement therapy for Fabry disease (deficient α-galactosidase A [α-Gal A] activity) were performed in α-Gal A–deficient mice. The pharmacokinetics and biodistributions were determined for four recombinant human α-Gal A glycoforms, which differed in sialic acid and mannose-6-phosphate content. The plasma half-lives of the glycoforms were ∼2–5 min, with the more sialylated glycoforms circulating longer. After intravenous doses of 1 or 10 mg/kg body weight were administered, each glycoform was primarily recovered in the liver, with detectable activity in other tissues but not in the brain. Normal or greater activity levels were reconstituted in various tissues after repeated doses (10 mg/kg every other day for eight doses) of the highly sialylated AGA-1 glycoform; 4 d later, enzyme activity was retained in the liver and spleen at levels that were, respectively, 30% and 10% of that recovered 1 h postinjection. Importantly, the globotriaosylceramide (GL-3) substrate was depleted in various tissues and plasma in a dose-dependent manner. A single or repeated doses (every 48 h for eight doses) of AGA-1 at 0.3–10.0 mg/kg cleared hepatic GL-3, whereas higher doses were required for depletion of GL-3 in other tissues. After a single dose of 3 mg/kg, hepatic GL-3 was cleared for ⩾4 wk, whereas cardiac and splenic GL-3 reaccumulated at 3 wk to ∼30% and ∼10% of pretreatment levels, respectively. Ultrastructural studies demonstrated reduced GL-3 storage posttreatment. These preclinical animal studies demonstrate the dose-dependent clearance of tissue and plasma GL-3 by administered α-Gal A, thereby providing the in vivo rationale—and the critical pharmacokinetic and pharmacodynamic data—for the design of enzyme-replacement trials in patients with Fabry disease.
cell.com