Aktuelle Neurologie 2007; 34(5): 283-290
DOI: 10.1055/s-2007-970887
Übersicht
© Georg Thieme Verlag KG Stuttgart · New York

Glykogenspeichererkrankung Typ 2 - Morbus Pompe

Neue pathophysiologische Aspekte und aktueller Stand der Enzymersatztherapie mit Alglucosidase-alfaGlycogen Storage Disease Type 2 - Pompe DiseaseNew Pathophysiological Aspects and Current State of Enzyme Replacement Therapy with Alglucosidase AlfaB.  G. H.  Schoser1
  • 1Friedrich-Baur-Institut, Neurologische Klinik, Ludwig-Maximilians-Universität München
Weitere Informationen

Publikationsverlauf

Publikationsdatum:
06. Juni 2007 (online)

Zusammenfassung

Die autosomal-rezessive Glykogenspeichererkrankung Typ 2 (GSD2, Morbus Pompe) ist durch Mangel des Glykogens degradierenden lysosomalen Enzyms α-1,4-Glucosidase (saure Maltase, acid alpha-glucosidase GAA) verursacht. Seit 2006 ist als sog. Ophan Drug Alglucosidase-alfa in Europa und den USA zur Behandlung der GSD2 zugelassen. Klinische Studien mit Alglucosidase-alfa bei infantiler und juveniler Pompe-Erkrankung zeigten eine Verlängerung der Überlebenszeit, der statomotorischen Entwicklung, und Verbesserung der Kardiomyopathie. Daher wird die Enzymersatztherapie auch für adulte Patienten eingesetzt und es liegen Erfolg versprechende Kasuistiken für diese Altersgruppe vor. Seit 2005 wird eine weitere Studie für diese Altersgruppe durchgeführt. Es zeichnen sich aber pathophysiologisch begründbare Einschränkungen der Wirksamkeit bei den Erwachsenen ab, insbesondere da weder das Überleben noch die Kardiomyopathie in dieser Altersgruppe relevante Zielparameter sein können. Die Enzymersatztherapie beruht auf einer rezeptormediierten Endozytose von rekombinantem humanem GAA. Dies ist besonders im Herzmuskel erfolgreich, aber im Skelettmuskel ist der Glykogenabbau in Typ-2-Fasern eingeschränkt. Neueste Untersuchungen zur Pathogenese zeigten neben dem Enzymmangel eine profunde Störung des lysosomalen autophagischen Abbauweges. Es findet sich eine progressive altersabhängige Zunahme von autophagischen Vakuolen in Kombination mit erweiterten, glykogengefüllten Lysosomen im Muskelgewebe. Teile des substituierten Enzyms scheinen in diesen autophagischen Zonen gefangen und erreichen nicht ihr Zielorganell, die Lysosomen. Zusätzlich sind mitochondriale Veränderungen, Lipofuszinablagerungen und eine Zerstörung des kontraktilen Apparates nachweisbar. Diese Zellstrukturänderungen sind relevante Faktoren für die Muskelschwäche und Ausdauereinschränkung der GSD2 Patienten. In dieser Arbeit werden aktuellen Aspekte zur Pathophysiologie vorgestellt und eine Zusammenfassung der Ergebnisse der Enzymersatztherapie berichtet.

Abstract

Glycogen storage disease type 2 (GSD2, Pompe disease) is caused by a deficiency of glycogen-degrading lysosomal enzyme acid alpha-glucosidase (GAA). In 2006 alglucosidase alfa was authorized as orphan drug treatment option of all phenotypes of GSD2. Results of clinical trails in infantile and juvenile Pompe disease with alglucosidase alfa showed prolonged survival, motor gains, and reversal of cardiomyopathy. These principal disease improvements by enzyme replacement therapy (ERT) in children led to spread the use of this therapy to the adult patients. Although there are some promising reports of successful ERT in adults, there seems to be some shortcomings of ERT in adults. The therapy, which relies on receptor-mediated endocytosis of recombinant human GAA (rhGAA), appears to be successful in cardiac muscle, but to a lesser degree in skeletal muscle, especially in clearing glycogen stores from type 2 muscle fibers. Recent investigations demonstrated a profound disturbance of the lysosomal degradative autophagic pathway. A progressive age-dependent autophagic built-up in addition to enlargement of glycogen-filled lysosomes was found in muscle tissue. Part of the substituted enzyme is trapped in these autophagic areas instead of reaching its lysosomal target. Furthermore, asides swollen, glycogen packed lysosomes, pronounced autophagia, mitochondrial alterations, lipofuscin, and direct contractile texture disturbances, like Z-line thickening and smearing are relevant for muscle weakness and power endurance handicap, seen in men and mice. In this report, an update on the pathophysiology and a summary of the current state of ERT in GSD2 will be given.

Literatur

  • 1 OMIM 232 300 Glycogen storage disease type 2. 
  • 2 Reuser A J, Koster J F, Hoogeveen A, Galjaard H. Biochemical, immunological, and cell genetic studies in glycogenosis type II.  Am J Hum Genet. 1978;  30 132-43
  • 3 Raben N, Plotz P, Byrne B J. Acid alpha-glucosidase deficiency (glycogenosis type II, Pompe disease).  Curr Mol Med. 2002;  2 145-166
  • 4 Baethmann M, Straub V. Morbus Pompe - Grundlagen, Diagnose und Therapie. Bremen; Uni-Med Verlag 2006
  • 5 Hirschhorn R, Reuser A JJ. Glycogen storage disease type II: acid α-glucosidase (Acid maltase) deficiency. In: Scriver CR, Beaudet AL, Valle D, Sly WS (eds) The metabolic and molecular bases of inherited disease. 8th ed. New York; McGraw-Hill 2001: 3389-3420
  • 6 Schoser B GH, Müller-Höcker J, Gempel K. et al .Glycogen storage disease type 2: clinico-pathological phenotype revisited. Neuropathol Appl Neurol 2007, im Druck
  • 7 Boerkoel C F, Exelbert R, Nicastri C. et al . Leaky splicing mutation in the acid maltase gene is associated with delayed onset of glycogenosis type II.  Am J Hum Genet. 1995;  56 887-897
  • 8 Hermans M MP, Leenen D van, Kroos M A. et al . Twenty-two novel mutations in the lysosomal alpha-glucosidase gene (GAA) underscore the genotype-phenotype correlation in glycogen storage disease type II.  Human Mutation. 2004;  23 47-56
  • 9 Laforet P, Nicolino M, Eymard B. et al . Juvenile and adult-onset acid maltase deficiency in France.  Neurology. 2000;  55 1122-1128
  • 10 Ausems M G, Wokke J H, Reuser A J, Diggelen O P van. Juvenile and adult-onset acid maltase deficiency in France: genotype-phenotype correlation.  Neurology. 2001;  57 1938
  • 11 Vorgerd M, Burwinkel B, Reichmann H. et al . Adult-onset glycogen storage disease type II: phenotypic and allelic heterogeneity in German patients.  Neurogenetics. 1998;  3 205-211
  • 12 Martin J J, Barsy T de, Hoof F van, Palladini G. Pompe's disease: an inborn lysosomal disorder with storage of glycogen. A study of brain and striated muscle.  Acta Neuropathol. 1973;  23 229-244
  • 13 Swash M, Schwartz M S, Apps M C. Adult onset acid maltase deficiency. Distribution and progression of clinical and pathological abnormality in a family.  J Neurol Sci. 1985;  68 61-74
  • 14 Walt J D van der, Swash M, Leake J, Cox E L. The pattern of involvement of adult-onset acid maltase deficiency at autopsy.  Muscle Nerve. 1987;  10 272-281
  • 15 Bijvoet A G, Kamp E H van de, Kroos M A. et al . Generalized glycogen storage and cardiomegaly in a knockout mouse model of Pompe disease.  Hum Mol Genet. 1998;  7 53-62
  • 16 Bijvoet A G, Hirtum H Van, Vermey M. et al . Pathological features of glycogen storage disease type II highlighted in the knockout mouse model.  J Pathol. 1999;  189 416-424
  • 17 Hesselink R P, Gorselink M, Schaart G. et al . Impaired performance of skeletal muscle in alpha-glucosidase knockout mice.  Muscle Nerve. 2002;  25 873-883
  • 18 Hesselink R P, Wagenmakers A J, Drost M R, Vusse G J Van der. Lysosomal dysfunction in muscle with special reference to glycogen storage disease type II.  Biochim Biophys Acta. 2003;  1637 164-170
  • 19 Hesselink R P, Kranenburg G Van, Wagenmakers A J. et al . Age-related decline in muscle strength and power output in acid 1 - 4 alpha-glucosidase knockout mice.  Muscle Nerve. 2005;  31 374-381
  • 20 Hesselink R P, Schaart G, Wagenmakers A J. et al . Age-related morphological changes in skeletal muscle cells of acid alpha-glucosidase knockout mice.  Muscle Nerve. 2006;  33 505-513
  • 21 Fukuda T, Ewan L, Bauer M. et al . Dysfunction of endocytic and autophagic pathways in a lysosomal storage disease.  Ann Neurol. 2006;  59 700-708
  • 22 Pompe J-C. Over idiopatische hypertrophie van het hart.  Ned Tijdschr Geneeskd. 1932;  76 304
  • 23 Putschar M. Über angeborene Glykogenspeicher-Krankheit des Herzens. „Thesaurimosis glycogenica” (v. Gierke).  Beitr Pathol Anat Allg Pathol. 1932;  90 222
  • 24 Hout H MP van den, Hop W, Diggelen O P van. et al . The natural course of infantile Pompe's disease: 20 original cases compared with 133 cases from the literature.  Pediatrics. 2003;  112 332-340
  • 25 Hagemans M L, Winkel L P, Hop W C. et al . Disease severity in children and adults with Pompe disease related to age and disease duration.  Neurology. 2005;  64 2139-2141
  • 26 Winkel L P, Hagemans M L, Doorn P A van. et al . The natural course of non-classic Pompe's disease; a review of 225 published cases.  J Neurol. 2005;  252 875-884
  • 27 Hagemans M LC, Winkel L PF, Doorn P A van. et al . Clinical manifestation and natural course of late-onset Pompe's disease in 54 Dutch patients.  Brain. 2005;  128 671-677
  • 28 Hagemans M L, Janssens A C, Winkel L P. et al . Late-onset Pompe disease primarily affects quality of life in physical health domains.  Neurology. 2004;  63 1688-1692
  • 29 Anneser J M, Pongratz D E, Podskarbi T. et al . Mutations in the acid alpha-glucosidase gene (M. Pompe) in a patient with an unusual phenotype.  Neurology. 2005;  64 368-370
  • 30 Kishnani P S, Steiner R D, Bali D. et al . Pompe disease diagnosis and managment guideline.  Genet in Med. 2006;  8 267-288
  • 31 Filipe M I, Lake B D. Histochemistry in pathology. Edinburgh; Churchill Livingstone 1983
  • 32 Pichiecchio A, Uggetti C, Ravaglia S. et al . Muscle MRI in adult-onset acid maltase deficiency.  Neuromuscul Disord. 2004;  14 51-55
  • 33 Engel A G, Dale A J. Autophagic glycogenosis of late onset with mitochondrial abnormalities: light and electron microscopic observations.  Mayo Clin Proc. 1968;  43 233-279
  • 34 Hudgson P, Gardner-Medwin D, Worsfold M. et al . Adult myopathy from glycogen storage disease due to acid maltase deficiency.  Brain. 1968;  91 435-462
  • 35 Engel A G. Acid maltase deficiency in adults: studies in four cases of a syndrome which may mimic muscular dystrophy or other myopathies.  Brain. 1970;  93 599-616
  • 36 Engel A G, Gomez M R, Seybold M E, Lambert E H. The spectrum and diagnosis of acid maltase deficiency.  Neurology. 1973;  23 95-106
  • 37 Slonim A E, Bulone L, Goldberg T. et al . Modification of the natural history of adult-onset acid maltase deficiency by nutrition and exercise therapy.  Muscle Nerve. 2007;  35 70-77
  • 38 Bodamer O A, Haas D, Hermans M M. et al . L-alanine supplementaion in late infantile glycogen storage disease type 2.  Pedratr Neurol. 2002;  27 145-156
  • 39 Bodamer O A, Halliday D, Leonard J V. The effects of l-alanine supplementation in late-onset glycogen storage disease type 2.  Neurology. 2000;  55 710-712
  • 40 Desnick R J. Enzyme replacement and enhancement therapies for lysosomal disorders.  J Inherit Metab Dis. 2004;  27 385-410
  • 41 Mengel E, Musch A. Alglucosidase-alfa.  Arzneimitteltherapie. 2007;  25 40-44
  • 42 Fachinformation Alglucosidase-alfa (Myozyme™),. Stand 29.03.2006
  • 43 Kikuchi T, Yang H W, Pennybacker M. et al . Clinical and metabolic correction of Pompe disease by enzyme therapy in acid maltase-deficient quail.  J Clin Invest. 1998;  101 827-833
  • 44 Bijvoet A G, Hirtum H Van, Kroos M A. et al . Human acid alphaglucosidase from rabbit milk has therapeutic effect in mice with glycogen storage disease type 2.  Hum Mol Genet. 1999;  8 2145-2153
  • 45 Raben N, Danon M, Gilbert A L. et al . Enzyme replacement therapy in the mouse model of Pompe disease.  Mol Genet Metab. 2003;  80 159-169
  • 46 Hout J L Van den, Reuser A J, Vulto A G. et al . Recombinant human alpha-glucosidase from rabbit milk in Pompe patients.  Lancet. 2000;  356 397-398
  • 47 Amalfitano A, Bengur A R, Morse R P. et al . Recombinant human acid alpha-glucosidase enzyme therapy for infantile glycogen storage disease type II: results of a phase I/II clinical trial.  Genet Med. 2001;  3 132-138
  • 48 Kishnani P S, Nicolinoo M, Voit T. et al . Chinese hamster ovary cell-derived recombinant human acid alpha-glucosidase in infantile-onset Pompe disease.  J Pediatr. 2006;  149 89-97
  • 49 Klinge L, Straub V, Neudorf U. et al . Safety and efficacy of recombinant acid alpha-glucosidase (rhGAA) in patients with classical infantile Pompe disease: results of a phase II clinical trial.  Neuromuscl Disord. 2005;  15 24-31
  • 50 Kishnani P S, Corzo D, Nicolino M. et al . Recombinant human acid [alpha]-glucosidase: major clinical benefits in infantile-onset Pompe disease.  Neurology. 2007;  68 99-109
  • 51 Hout J MP Van den, Kamphoven J HJ, Winkel L PF. et al . Long-Term intravenous treatment f Pompe disease with recombinant human alpha-glucosidase from milk.  Pediatrics. 2004;  113 448-457
  • 52 Klinge L, Straub V, Neudorf U, Voit T. Enzyme replacement therapy in classic infantile Pompe disease: results of a ten-month follow-up study.  Neuropediatrics. 2005;  36 6-11
  • 53 Winkel L P, Hout J M Van den, Kamphoven J H. et al . Enzyme replacement therapy in late-onset Pompe's disease: a three-year follow-up.  Ann Neurol. 2004;  55 495-502
  • 54 Koeberl D D, Kishnani P S, Chen Y T. Glycogen storage disease types 1 and 2: treatment updates.  J Inherit Metab Dis. 2007;  30 159-164
  • 55 Hunley T E, Corzo D, Dudek M. et al . Nephrotic syndrome complicating alpha-glucosidase replacement therapy for Pompe disease.  Pediatrics. 2004;  114 532-535
  • 56 Raben N, Fukuda T, Gilbert A L. et al . Replacing acid alpha-glucosidase in Pompe disease: recombinant and transgenic enzymes are equipotent, but neither completely clears glycogen from type II muscle fibers.  Mol Ther. 2005;  11 48-56
  • 57 Winkel L P, Kamphoven J H, Hout H J van den. et al . Morphological changes in muscle tissue of patients with infantile Pompe's disease receiving enzyme replacement therapy.  Muscle Nerve. 2003;  27 743-751
  • 58 Zhu Y, Li X, Kyazike J. et al . Conjugation of mannose 6-phosphate-containing oligosaccharides to acid alfa-glucosidase improves the clearance of glycogen in Pompe mice.  J Biol Chem,. 2004;  279 50336-50341
  • 59 Sun B, Zhang H, Franco L M. et al . Efficacy of an adeno-associated virus 8-pseudotyped vector in glycogen storage disease type II.  Mol Ther. 2005;  11 57-65
  • 60 Mah C, Pacak C A, Cresawn K O. et al . Physiological correction of Pompe disease by systemic delivery of adeno-associated virus serotype 1 vector.  Mol Ther. 2007;  15 501-507

PD Dr. med. Benedikt G. H. Schoser

Friedrich-Baur-Institut, Neurologische Klinik, Ludwig-Maximilians-Universität München

Ziemssenstraße 1 a

80336 München

eMail: bschoser@med.uni-muenchen.de

    >