Browse > Article
http://dx.doi.org/10.5483/BMBRep.2015.48.8.101

Glyco-engineering strategies for the development of therapeutic enzymes with improved efficacy for the treatment of lysosomal storage diseases  

Oh, Doo-Byoung (Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), University of Science and Technology (UST))
Publication Information
BMB Reports / v.48, no.8, 2015 , pp. 438-444 More about this Journal
Abstract
Lysosomal storage diseases (LSDs) are a group of inherent diseases characterized by massive accumulation of undigested compounds in lysosomes, which is caused by genetic defects resulting in the deficiency of a lysosomal hydrolase. Currently, enzyme replacement therapy has been successfully used for treatment of 7 LSDs with 10 approved therapeutic enzymes whereas new approaches such as pharmacological chaperones and gene therapy still await evaluation in clinical trials. While therapeutic enzymes for Gaucher disease have N-glycans with terminal mannose residues for targeting to macrophages, the others require N-glycans containing mannose-6-phosphates that are recognized by mannose-6-phosphate receptors on the plasma membrane for cellular uptake and targeting to lysosomes. Due to the fact that efficient lysosomal delivery of therapeutic enzymes is essential for the clearance of accumulated compounds, the suitable glycan structure and its high content are key factors for efficient therapeutic efficacy. Therefore, glycan remodeling strategies to improve lysosomal targeting and tissue distribution have been highlighted. This review describes the glycan structures that are important for lysosomal targeting and provides information on recent glyco-engineering technologies for the development of therapeutic enzymes with improved efficacy. [BMB Reports 2015; 48(8): 438-444]
Keywords
Enzyme replacement therapy; Glcyan; Glyco-engineering; Lysosomal storage disease; Mannose-6-phosphate; Therapeutic enzymes;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Tiels P, Baranova E, Piens K et al (2012) A bacterial glycosidase enables mannose-6-phosphate modification and improved cellular uptake of yeast-produced recombinant human lysosomal enzymes. Nat Biotechnol 30, 1225-1231   DOI
2 Gil JY, Park JN, Lee KJ et al (2015) Increased mannosylphosphorylation of N-glycans by heterologous expression of YlMPO1 in glyco-engineered Saccharomyces cerevisiae for mannose-6-phosphate modification. J Biotechnol 206, 66-74   DOI
3 Zhu Y, Li X, Kyazike J et al (2004) Conjugation of mannose 6-phosphate-containing oligosaccharides to acid alpha-glucosidase improves the clearance of glycogen in pompe mice. J Biol Chem 279, 50336-50341   DOI
4 Zhu Y, Li X, McVie-Wylie A et al (2005) Carbohydrate-remodelled acid alpha-glucosidase with higher affinity for the cation-independent mannose 6-phosphate receptor demonstrates improved delivery to muscles of Pompe mice. Biochem J 389, 619-628   DOI
5 Zhu Y, Jiang JL, Gumlaw NK et al (2009) Glycoengineered acid alpha-glucosidase with improved efficacy at correcting the metabolic aberrations and motor function deficits in a mouse model of Pompe disease. Mol Ther 17, 954-963   DOI
6 Zhou Q, Stefano JE, Harrahy J et al (2011) Strategies for Neoglycan conjugation to human acid alpha-glucosidase. Bioconjug Chem 22, 741-751   DOI
7 Zhou Q, Avila LZ, Konowicz PA et al (2013) Glycan structure determinants for cation-independent mannose 6-phosphate receptor binding and cellular uptake of a recombinant protein. Bioconjug Chem 24, 2025-2035   DOI
8 Kishnani PS and Beckemeyer AA (2014) New therapeutic approaches for Pompe disease: enzyme replacement therapy and beyond. Pediatr Endocrinol Rev 12 Suppl 1, 114-124
9 Chiba Y, Sakuraba H, Kotani M et al (2002) Production in yeast of alpha-galactosidase A, a lysosomal enzyme applicable to enzyme replacement therapy for Fabry disease. Glycobiology 12, 821-828   DOI
10 Akeboshi H, Kasahara Y, Tsuji D et al (2009) Production of human beta-hexosaminidase A with highly phosphorylated N-glycans by the overexpression of the Ogataea minuta MNN4 gene. Glycobiology 19, 1002-1009   DOI
11 Tsukimura T, Kawashima I, Togawa T et al (2012) Efficient uptake of recombinant alpha-galactosidase A produced with a gene-manipulated yeast by Fabry mice kidneys. Mol Med 18, 76-82   DOI
12 Van den Hout JM, Kamphoven JH, Winkel LP et al (2004) Long-term intravenous treatment of Pompe disease with recombinant human alpha-glucosidase from milk. Pediatrics 113, e448-457   DOI
13 Kishnani PS, Nicolino M, Voit T et al (2006) Chinese hamster ovary cell-derived recombinant human acid alpha-glucosidase in infantile-onset Pompe disease. J Pediatr 149, 89-97   DOI
14 McVie-Wylie AJ, Lee KL, Qiu H et al (2008) Biochemical and pharmacological characterization of different recombinant acid alpha-glucosidase preparations evaluated for the treatment of Pompe disease. Mol Genet Metab 94, 448-455   DOI
15 Tekoah Y, Tzaban S, Kizhner T et al (2013) Glycosylation and functionality of recombinant beta-glucocerebrosidase from various production systems. Biosci Rep 33   DOI
16 Grabowski GA, Golembo M and Shaaltiel Y (2014) Taliglucerase alfa: an enzyme replacement therapy using plant cell expression technology. Mol Genet Metab 112, 1-8   DOI
17 Chavez CA, Bohnsack RN, Kudo M, Gotschall RR, Canfield WM and Dahms NM (2007) Domain 5 of the cation-independent mannose 6-phosphate receptor preferentially binds phosphodiesters (mannose 6-phosphate N-acetylglucosamine ester). Biochemistry 46, 12604-12617   DOI
18 Berger J, Stirnemann J, Bourgne C et al (2012) The uptake of recombinant glucocerebrosidases by blood monocytes from type 1 Gaucher disease patients is variable. Br J Haematol 157, 274-277   DOI
19 Van Patten SM, Hughes H, Huff MR et al (2007) Effect of mannose chain length on targeting of glucocerebrosidase for enzyme replacement therapy of Gaucher disease. Glycobiology 17, 467-478   DOI
20 Koeberl DD, Luo X, Sun B et al (2011) Enhanced efficacy of enzyme replacement therapy in Pompe disease through mannose-6-phosphate receptor expression in skeletal muscle. Mol Genet Metab 103, 107-112   DOI
21 Qian Y, Lee I, Lee WS et al (2010) Functions of the alpha, beta, and gamma subunits of UDP-GlcNAc:lysosomal enzyme N-acetylglucosamine-1-phosphotransferase. J Biol Chem 285, 3360-3370   DOI
22 Sommerlade HJ, Selmer T, Ingendoh A et al (1994) Glycosylation and phosphorylation of arylsulfatase A. J Biol Chem 269, 20977-20981
23 Lee K, Jin X, Zhang K et al (2003) A biochemical and pharmacological comparison of enzyme replacement therapies for the glycolipid storage disorder Fabry disease. Glycobiology 13, 305-313   DOI
24 Sakuraba H, Murata-Ohsawa M, Kawashima I et al (2006) Comparison of the effects of agalsidase alfa and agalsidase beta on cultured human Fabry fibroblasts and Fabry mice. J Hum Genet 51, 180-188   DOI
25 Lauer RM, Mascarinas T, Racela AS, Diehl AM and Brown BI (1968) Administration of a mixture of fungal glucosidases to a patient with type II glycogenosis (Pompe's disease). Pediatrics 42, 672-676
26 Sohn Y, Lee JM, Park HR, Jung SC, Park TH and Oh DB (2013) Enhanced sialylation and in vivo efficacy of recombinant human alpha-galactosidase through in vitro glycosylation. BMB Rep 46, 157-162   DOI
27 Vedder AC, Linthorst GE, Houge G et al (2007) Treatment of Fabry disease: outcome of a comparative trial with agalsidase alfa or beta at a dose of 0.2 mg/kg. PLoS One 2, e598   DOI
28 Togawa T, Takada M, Aizawa Y, Tsukimura T, Chiba Y and Sakuraba H (2014) Comparative study on mannose 6-phosphate residue contents of recombinant lysosomal enzymes. Mol Genet Metab 111, 369-373   DOI
29 de Barsy T, Jacquemin P, Van Hoof F and Hers HG (1973) Enzyme replacement in Pompe disease: an attempt with purified human acid alpha-glucosidase. Birth Defects Orig Artic Ser 9, 184-190
30 Ohashi T (2012) Enzyme replacement therapy for lysosomal storage diseases. Pediatr Endocrinol Rev 10 Suppl 1, 26-34
31 Ortolano S, Vieitez I, Navarro C and Spuch C (2014) Treatment of lysosomal storage diseases: recent patents and future strategies. Recent Pat Endocr Metab Immune Drug Discov 8, 9-25   DOI
32 Baldo BA (2015) Enzymes approved for human therapy: indications, mechanisms and adverse effects. BioDrugs 29, 31-55   DOI
33 Mechler K, Mountford WK, Hoffmann GF and Ries M (2015) Pressure for drug development in lysosomal storage disorders - a quantitative analysis thirty years beyond the US orphan drug act. Orphanet J Rare Dis 10, 46   DOI
34 Tiede S, Storch S, Lubke T et al (2005) Mucolipidosis II is caused by mutations in GNPTA encoding the alpha/beta GlcNAc-1-phosphotransferase. Nat Med 11, 1109-1112   DOI
35 Braulke T and Bonifacino JS (2009) Sorting of lysosomal proteins. Biochim Biophys Acta 1793, 605-614   DOI
36 Kim JJ, Olson LJ and Dahms NM (2009) Carbohydrate recognition by the mannose-6-phosphate receptors. Curr Opin Struct Biol 19, 534-542   DOI
37 Coutinho MF, Prata MJ and Alves S (2012) Mannose-6- phosphate pathway: a review on its role in lysosomal function and dysfunction. Mol Genet Metab 105, 542-550   DOI
38 Kudo M, Bao M, D'Souza A et al (2005) The alpha- and beta-subunits of the human UDP-N-acetylglucosamine: lysosomal enzyme N-acetylglucosamine-1-phosphotransferase [corrected] are encoded by a single cDNA. J Biol Chem 280, 36141-36149   DOI