79
Views
0
CrossRef citations to date
0
Altmetric
Review

A review of treatment of Pompe disease in infants

&
Pages 195-201 | Published online: 07 Dec 2022

Abstract

The glycogen storage disease type II (GSD-II), or Pompe disease, is due to the deficit of lysosomal glycogen degradation enzyme acid α-glucosidase (GAA). In infants, Pompe disease is characterized by prominent hypotonia, muscle weakness, motor delay, feeding problems, and respiratory and cardiac insufficiency. In a retrospective study, the median age at death was 8.7 months. Enzyme replacement therapy with recombinant human GAA is recently used to treat patients with Pompe disease, and has been shown to prolong survival, reverse cardiomyopathy, and improve motor function. This article briefly reviews the history and manifestations of Pompe disease, and then focuses on the development of the drug for Pompe disease, alglucosidase alfa. Current status of treatment and future developments are also discussed.

History

Pompe disease was first described by Dr. Pompe almost 55 years ago in a 7-month-old girl with cardiomyopathy, in whom massive accumulation of glycogen in vacuoles was observed in all tissues examined (CitationPompe 1932). After identification of lysosome by Citationde Duve in 1963 (CitationDe Duve 1963, Citation2005), Pompe disease became the first recognized lysosomal storage disorder (LSD) when it was found that the disease was due to the deficit of lysosomal glycogen degradation enzyme acid α-glucosidase (GAA, EC 3.2.1.20) (CitationHers 1963). Pompe disease is formally named glycogen storage disease type II (GSD-II), but most other types of GSDs are caused by defects in the cytosolic glycogen cleavage or synthesis pathways (CitationShin 2006). Pompe disease is remotely related to Danon disease, also known as X-linked vacuolar cardiomyopathy and myopathy (CitationDanon et al 1981). In Danon disease, lysosomal glycogen storage is caused by mutation of the lysosome-associated membrane protein-2 (LAMP-2) gene, and there is no GAA deficiency (CitationNishino et al 2000).

Clinical manifestation

Pompe disease is usually classified into infantile-onset and late-onset for convenience, although there is clearly a continuum in disease severity ranging from the most severe classical infantile-onset Pompe disease (IOPD), to the mildest adult-onset Pompe disease (CitationHirschhorn and Reuser 2001; CitationRaben et al 2002). In the Netherlands, a combined frequency for all type of Pompe disease was estimated to be 1 in 50,000 (CitationPoorthuis et al 1999). After screening for seven mutations in an ethnically diverse population, a study in New York suggested an incidence of 1 in 40,000 (CitationMartiniuk et al 1998). The incidence of Pompe disease in certain ethnic groups, for example in Israel (CitationBashan et al 1988) and in Taiwan (CitationLin and Shieh 1996), may be higher. Patients of IOPD are usually recognized at the age of 3–5 months because of a respiratory infection (CitationHirschhorn and Reuser 2001; CitationMarsden 2005). A chest X-ray at that time may show cardiomegaly which leads to further check-up and diagnosis of IOPD. On physical examination, head lag during a traction from the supine position is usual prominent. Although weakness of extremities or trunk can usually be confirmed retrospectively at the age of 2–3 months (CitationMarsden 2005; CitationKishnani et al 2006a), only rarely do these signs trigger an investigation of affected children (CitationHowell et al 2006). The lack of power for early diagnosis for hypotonia can be explained both by parental lack of experience and by children’s large individual variation in normal development.

Because of the late diagnosis in most cases of IOPD, it is not clear how early signs of cardiomegaly can be detected. Pompe disease diagnosis at the newborn stage has been described, but was not found in a recent large-scale natural history study (CitationKishnani et al 2006a). However, recently a newborn baby was diagnosed in Hong Kong because of respiratory distress shortly after birth (personal communication to Dr. Grace Poon). Large QRS complexes and short PR interval have been the cardinal diagnostic criteria in EKG (CitationAnsong et al 2006), but the sensitivity of short PR interval is not high. In echocardiography, the thickening of the ventricular wall and the interventricular septum are marked, the ventricular cavities are very small and the outflow tract can be obliterated. Symptomatic outflow tract obstruction may occur shortly after the diagnosis of IOPD. A careful management of this condition is critical, and an adequate use of diuretics and β-blockers can usually help the affected baby to overcome this symptom, but acute death is also possible (CitationKishnani et al 2006c). As the disease advances, the heart becomes more dilated and obstruction sign can disappear. Occasionally, overt cardiac failure leads to death.

According to the recent natural history review, the progression of IOPD is very fast, and the gap between the median age at diagnosis and the median age of first use of ventilator was only 1.2 months (CitationKishnani et al 2006a), and between median age at diagnosis and death was 2.4 months in a Dutch study (Citationvan den Hout et al 2003). The major illness beyond cardiac obstruction would be respiratory failure. Weakness of the respiratory muscles and cough reflex are the major causes of respiratory problem, and once these children are intubated, they rapidly become ventilator dependent. If the lives of patients could be prolonged, for example by ventilator support or enzyme replacement (to be discussed later), symptoms related to other organ systems may present (CitationKishnani et al 2006c). Weakness of the diaphragm makes hygiene of the lower pulmonary segments difficult. Paradoxical respiration, that is, elevation of the diaphragm during inspiration, is also common. Dysfunction of the gastrointestinal tract is a serious complication. Dysphagia, gastro-esophageal reflux, delayed gastric empty time, and intestinal dysfunction are all likely. If swallowing is problematic, jejunostomy rather than gastrostomy will be necessary to achieve adequate feeding without causing aspiration (CitationKishnani et al 2006c). The involvements of other organ systems besides the muscle are not a surprise, since glycogen storage is universal throughout the body. Four cases with IOPD revealed a hearing loss which may be due to a problem in the cochlea or middle ear (CitationVan den Hout et al 2004). Brain development is also a concern, since excessive glycogen storage has been found in both neurons and glial cells (CitationGambetti et al 1971; CitationSakurai et al 1974). However, catch up in brain myelination after stabilization of the patients has also been reported (CitationChien et al 2006). In conclusion, without specific treatment, neither any of the above-mentioned efforts, nor rehabilitation or dietary management could alter the outcome of IOPD.

Molecular basis

The GAA gene is located on chromosome 17q25. The gene encodes a peptide of 952 amino acids with nonglycosylated weight of 105 kDa (CitationHirschhorn and Reuser 2001). Extensive modifications of the protein occurs afterwards, which include glycosylation and proteolytic processing, resulting in the 76 and 70 kDa mature GAA protein (CitationHirschhorn and Reuser 2001). More than 70 mutations have been found on the GAA gene (CitationRaben et al 2002; CitationHermans et al 2004). The mutations are spread over the gene. Several common mutations are found in different ethnic groups, including the IVS1 t-13g in Caucasian patients with adult-onset disease (CitationHuie et al 1994; CitationBoerkoel et al 1995; CitationKroos et al 1995), the Arg854X mutation in African Americans (CitationTsujino et al 2000), and the Asp645Glu mutation in Chinese patients from Taiwan (CitationLin and Shieh 1996; CitationKo et al 1999). A few of the mutations have been expressed in cultured cells and the GAA activities obtained can be correlated to the phenotypes or the severity of the disease (CitationHermans et al 2004).

Laboratory diagnosis

Laboratory diagnosis depends on the measurement of GAA activity in blood or tissues. The assay used to measure GAA activity usually employs an artificial fluorogenic substrate, 4-methylumbelliferyl-α-D-glucopyranoside (4-MUG), but several α-glucosidase (E.C. 3.2.1.20) isoenzymes especially maltase-glucoamylase (MGA) in blood and hematopoietic cells contribute to the observed reaction (CitationTaniguchi et al 1978; CitationShin et al 1985; CitationHirschhorn and Reuser 2001). Therefore the assay has to be done either in lymphocytes or in skin fibroblasts which have activity at pH 4.0 mainly due to GAA (CitationTaniguchi et al 1978; CitationShin et al 1985). However, lymphocyte preparations are often contaminated by leukocytes, and the resulting falsely high residual activity cannot be used to predict phenotype. Recently, immobilized antibodies have been used to remove the interfering enzymes (CitationSchram et al 1979; CitationUmapathysivam et al 2000; CitationUmapathysivam et al 2001). GAA specific activity measured in this way, together with data from skin fibrobasts, may make it possible to predict the age of onset of Pompe disease (CitationUmapathysivam et al 2005).

The start of enzyme replacement therapy

The first successful attempt towards a specific pharmaceutical treatment of LSD was in Gaucher disease. The enzyme deficient in Gaucher disease, the β-glucosidase, was first purified from placenta in 1974 (CitationBrady et al 1974; CitationFurbish et al 1977). It was discovered later that it was necessary to modify the oligosaccharide moieties on the enzyme to facilitate its macrophage targeting. The exogenous enzyme molecules are internalized and shipped in vacuoles through receptor-mediated mechanisms. These vacuoles then fuse with the lysosomes elegantly to correct their intrinsic defect. These processes make exogenous supplementation of lysosomal enzymes, or enzyme replacement therapy (ERT), an efficient way to treat LSD (CitationBrady 2006). Placental β-glucosidase was approved for this use by the US Food and Drug Administration (FDA) in 1991, and was later replaced by a Chinese hamster ovary (CHO) cell-produced recombinant β-glucosidase in 1994 (CitationBarranger and O’Rourke 2001). More than 4000 patients all over the world have been treated with this drug (CitationRosenbloom et al 2005). ERT have been developed for several other diseases, including Fabry disease, mucopolysaccharidosis (MPS) type I, MPS type II and MPS type IV (CitationBeck 2007).

Recombinant human GAA (rhGAA) production

Previously, there was no effective treatment for Pompe disease. Bone marrow transplantation seems not to work. A combined treatment including dietary management and physical therapy has not shown significant effects in IOPD patients (CitationBembi et al 2003). Recently, two groups of researchers followed different approaches to produce a recombinant GAA protein. One group used the innovative approach of transgenic production of the therapeutic enzyme in mammalian milk (CitationBijvoet et al 1996). The human GAA cDNA was placed under the control of the αS1-casein promoter and expressed in mice. The recombinant GAA purified from mice milk was internalized via the mannose 6-phosphate receptor and corrected the enzyme deficiency in fibroblasts from patients (CitationBijvoet et al 1996). The mouse transgenic construct was later modified for higher production, and the enzyme, when injected intravenously, corrects the GAA deficiency in heart and skeletal muscles of GSD-II knockout mice (CitationBijvoet et al 1998). This success in producing recombinant GAA from mice milk led to the industrial manufacturing of GAA from rabbit. In the GAA knockout mice, a single intravenous infusion of the enzyme gave a full correction of GAA deficiency in all mice tissues except brain (CitationBijvoet et al 1999). Weekly enzyme infusions over 6 months resulted in degradation of lysosomal glycogen in the heart and muscle. The results then led to a clinical trial with rabbit milk recombinant enzyme.

Another study group chose the more traditional approach of expressing the recombinant enzyme in CHO cells (CitationVan Hove et al 1996, Citation1997). This enzyme was effectively taken up by fibroblasts from Pompe patients (CitationVan Hove et al 1996), restoring normal levels of GAA and glycogen. By intravenous infusion, the enzyme can be targeted to the heart and liver efficiently in guinea pigs (CitationVan Hove et al 1996). Pre-clinical studies for CHO enzyme were conducted in GAA-deficient Japanese quails. Uptake and targeting of the enzyme were first tested in quail fibroblast (CitationYang et al 1998). The results of the treatment were very encouraging; besides improvements in muscle histology, there was clear gain in function (CitationKikuchi et al 1998). The quails were treated at the age of 4 weeks. After 7 injections over a 16-day period, the sham-treated quails exhibited progressive myopathy and could not lift their wings or right themselves from the supine position. However, treated birds could flap their wings, and one bird flew up more than 100 cm.

Dosage and frequency

However, these pre-clinical studies also disclosed one truth: high doses of the recombinant enzyme were required for a positive response. For example, in the quail study, the effect of high-dose treatment (14 mg/kg) was much better than low-dose treatment (4.2 mg/kg) (CitationKikuchi et al 1998). There was an additional experiment with intermediate doses (5.7–9 mg/kg) and extended treatment. Despite significantly improved histopathology of affected tissues, the quails in this group showed no clinical improvement in muscle strength and were not able to flap their wings or fly (CitationKikuchi et al 1998). The requirement in Pompe disease of large amount of enzyme to correct muscle pathology is in contrast to the small dose necessary to correct several other LSDs. For example, the regular dosage for Fabry disease is 1 mg/kg (CitationWilcox et al 2004). Currently, why a high dose of the enzyme is required in Pompe disease is not fully known. One possible explanation is that the paucity of mannose 6-phosphate receptor in the muscle tissues prevents efficient uptake of the enzyme (CitationWenk et al 1991; CitationRaben et al 2003). This large requirement of enzyme should have posed a challenge in both the production and the cost of the drug, but currently these issues have been solved. Problems, such as immunologic responses, arising from the infusion of large amounts of foreign protein, will be discussed later.

Clinical trials

Early clinical trials for rhGAA in IOPD patients revealed promising results. With the rabbit milk rhGAA, 4 patients were treated for 36 weeks and first reported in 2000 (CitationVan den Hout et al 2000). Doses started from 15–20 mg/kg to 40 mg/kg once weekly. The most prominent effect was on the heart: the left ventricular mass index decreased to less than 30% of baseline within 36 weeks. Skeletal muscle function and strength also improved in all patients. One patient treated at the age of 2.5 months was cross-reactive-immunological-material (CRIM) negative, but her improvement continued over the study period. Only one patient gained the normal major motor development. Therefore the researchers recommended that treatment be started early, before the destruction of muscle architecture, for successful outcome of treatment (CitationVan den Hout et al 2004). The long-term outcomes of 2 cases were further described, and survival time under therapy was 55 and 58 months (CitationKlinge et al 2005a; CitationKlinge et al 2005b). There were only mild infusion-associated reactions during the follow-up period. Both patients sat, and their mental development was normal. The production of milk rhGAA was later discontinued.

The first clinical trial (phase I/II) with CHO cell-produced rhGAA was done in 3 patients (CitationAmalfitano et al 2001). They received twice-weekly infusion of rhGAA at a dosage of 5 mg/kg for 14–17 months. Adverse reactions were mild and could be resolved by a pre-infusion dose of antihistamine. Steady decrease in heart size was universal. Improvements in skeletal muscle function were also noted. One patient walked independently from 12 months of age. A high titer of anti-rhGAA antibody developed in two patients who had no detectable GAA protein by Western blot analysis (CRIM negative). This promising preliminary result triggered a scaled-up production of the enzyme and the phase II trial in 8 patients with IOPD (CitationKishnani et al 2006b). Dosages were either 10–20 mg/kg weekly or 20 mg/kg every 2 weeks. Safety of the drug was first confirmed. Although IgG antibodies to rhGAA developed in all 8 patients by week 8 of treatment, after 52 weeks of treatment the antibody titers either decreased or remained unchanged. Six of 8 patients were alive after the study period. Clinical improvements included ameliorated cardiomyopathy, improved growth and cognition, and 3 patients walked independently. Even though 4 patients subsequently died, in view of the fact that this is a rapidly fatal disease if left untreated, rhGAA did change the clinical course of the disease.

Finally a phase III trial with the CHO cell rhGAA started. This trial enrolled 18 patients younger than 6 months of age worldwide (CitationKishnani et al 2007). The dosage was 20 or 40 mg/kg every other week. After treatment for 52 weeks, all patients survived to 18 months of age. There was no clear advantage of the 40 mg/kg dose. A Cox proportional hazards analysis demonstrated that the treatment decreased risk of death by 99%. This clinical trial led to FDA approval of Alglucosidase Alfa (Myozyme®, Genyzme Corporation). The recommended dose is 20 mg/kg every other week. For late-onset forms of Pompe disease, rhGAA both from rabbit milk or from CHO cells has shown preliminary therapeutic effects, although the number of reported cases is still small (CitationVan den Hout et al 2004; CitationWinkel et al 2004).

Disadvantages: high required dosage and antibody formation

The requirement of high dose of rhGAA to treat Pompe disease, almost 10 fold of other diseases, is also unique. The volume of muscle tissue to treat is large, as mentioned. Lower efficiency in enzyme uptake by the muscular system may be another factor. Both mediated by the mannose 6-phosphate receptor, the cardiac muscles are much more efficient in enzyme uptake than the skeletal muscles (CitationWenk et al 1991; CitationRaben et al 2003). Infusion of large amount of foreign proteins may causes problems. One case report described the development of nephrotic syndrome in a patient receiving a daily infusion of 10 mg/kg of rhGAA (CitationHunley et al 2004). Life-threatening anaphylactic reactions including anaphylactic shock have been observed in patients during alglucosidase alfa treatment. Antibody development is universal in treated patients, most within the first 3 months of treatment (CitationKishnani et al 2006b). Antibodies to the infused enzyme have been observed in other ERTs, but rarely cause trouble (CitationRosenberg et al 1999; CitationWilcox et al 2004). In IOPD, some patients were CRIM negative. Antibody titers seemed to be higher in those cases (CitationAmalfitano et al 2001). Although the surge of antibody titers in some patients has been suspected to be related to the loss of therapeutic effect (CitationAmalfitano et al 2001), the case number is too small to conclude whether CRIM status or the antibody titer will be an untoward prognostic factor in treatment of Pompe disease.

CRIM status may indeed reflect the severity of the disease. It is suggested that the better the clinical status before initiation of treatment, the better the outcome. With a milder or later-onset disease, application of alglucosidase alfa therapy promises a good likelihood of recovery. In a recent paper, the muscle pathologies of Pompe disease were classified into 5 stages (CitationThurberg et al 2006). Landmarks in the staging include appearance of intra-lysosomal or cytoplasmic glycogen, integrity of the mitochondria, and myofibril dissociation (CitationThurberg et al 2006). It is likely that muscle cells may be not able to recover when the damage is too advanced.

Early treatment

Another view in considering the relationship between treatment effect and disease severity is the timing of treatment. That is, treatment should be started before the threshold of muscle pathology been reached (CitationThurberg et al 2006). A more severe disease or more rapid disease progression requires earlier treatment. Clinical conditions like respiratory failure, mechanical ventilation, or immobilization are almost fatal regardless of treatment. However, from a recent IOPD natural history survey, the median age at diagnosis was 4.7 months, while the median age of first ventilator use was 5.9 months, only one month apart (Citationvan den Hout et al 2003; CitationKishnani et al 2006a). On the other hand, previous experience in Pompe disease treatment has demonstrated that drug effect takes 2–3 months to appear. For example, the decrease in heart size usually takes 3 months after initiation of treatment (CitationAmalfitano et al 2001). Therefore it is likely that a poor early clinical course will destroy the therapeutic effect. In the past few years, a number of IOPD patients have been treated in different clinical trials or by compassionate use. Because of the difficulty of getting into treatment, many cases had late treatment. Partial reversal of disease manifestations, including being bedridden or wheelchair bound, feeding problems, or even ventilator dependence were not uncommon. In those partially treated cases, complications included bone fracture, heart arrhythmia (preexisting ectopy including premature ventricular contractions, perhaps caused by fibrous tissue deposition in the myocardium), and problems in brain myelination (CitationCook et al 2006; CitationCase et al 2007). After the formal launch of alglucosidase alfa, the situation should improve. However, patients receiving ERT may need a follow-up to see the long-term safety and efficacy of alglucosidase alfa.

Currently accumulating evidence suggests that early treatment for IOPD should be beneficial. However, as discussed in previously sections, early diagnosis of IOPD is rarely achieved unless there is positive family history, because early signs are non-specific. Therefore, screening for IOPD in very early ages of babies is the best and only choice. Laboratory diagnosis of Pompe disease, as discussed in a previous section, has been difficult. But in the past few years there were significant progresses in this field. First is the employment of blood spot as the source for lysosomal enzyme assays (CitationChamoles et al 2001). With an extended enzyme reaction time, a tiny amount of blood lysate from 1/8 inch spots could serve several assays (CitationChamoles et al 2002a, b). The most critical invention is the use of inhibitors (maltose or acarbose) in the assay, so the assay could be performed without blood cell separation (CitationChamoles et al 2004; CitationLi et al 2004). Very important information will be generated from the screening programs, for example, what percentage of IOPD can have a satisfactory outcome if treated early, or whether CRIM-negative patients can be rescued if treated early.

Future aspects in ERT

Lastly, the failure of the rabbit milk product has shown the difficulty of developing transgenic methods to produce large molecule therapeutic agents. It is not clear why the company withdraw the project, since early clinical trials disclosed comparable treatment effect to the other product. There may be difficulties or high cost in maintaining the herd or in purification of the product from the large quantities of milk protein. However, the method of standardization in culturing cells does look more feasible than raising animals. Several recent enzyme products are produced from human fibroblasts. It has been argued whether human cell products will be better than animal cell products, for example the agalsidase beta from CHO cells (Fabrazyme®, Genzyme Corporation) and agalsidase alfa from human fibroblasts (Replagal®, Shire) (CitationBeck 2002). But the first and successful ERT product, the imiglucerase (Cerezyme®, Genzyme Corporation), is from CHO cells. The infusion reaction for imiglucerase is transient and mild, and rarely interferes with treatment. Therefore, contamination of animal cell proteins seems negligible. The question of the efficacy of fibroblasts enzymes cannot be easily answered because of the large differences in disease pathophysiology.

Future development in ERT therapy will need to overcome other difficulties especially in targeting the enzyme. For example, conjugation of mannose 6-phosphate-containing oligosaccharides to acid alpha-glucosidase improves the delivery to muscles and the clearance of glycogen in pompe mice (CitationZhu et al 2004, Citation2005). Also, a tag with an acidic oligopeptide may enhance drug delivery to bone (CitationNishioka et al 2006), and a phosphorylated rhGALNS can be delivered to multiple tissues, including bone, implying a potential enzyme replacement treatment for mucopolysaccharidosis type IVA (CitationTomatsu et al 2007).

References

  • AmalfitanoABengurARMorseRP2001Recombinant human acid alpha-glucosidase enzyme therapy for infantile glycogen storage disease type II: results of a phase I/II clinical trialGenet Med3132811286229
  • AnsongAKLiJSNozik-GrayckE2006Electrocardiographic response to enzyme replacement therapy for Pompe diseaseGenet Med829730116702879
  • BarrangerJAO’RourkeE2001Lessons learned from the development of enzyme therapy for Gaucher diseaseJ Inherit Metab Dis24Suppl 28996 discussion 87–811758684
  • BashanNPotashnikRBarashV1988Glycogen storage disease type II in IsraelIsrael J Med Sci2422473132435
  • BeckM2002Agalsidase alfa–a preparation for enzyme replacement therapy in Anderson-Fabry diseaseExpert Opin Investig Drugs118518
  • BeckM2007New therapeutic options for lysosomal storage disorders: enzyme replacement, small molecules and gene therapyHum Genet12112217089160
  • BembiBCianaGMartiniC2003Efficacy of multidisciplinary approach in the treatment of two cases of nonclassical infantile glycogenosis type IIJ Inherit Metab Dis266758114707516
  • BijvoetAGKroosMAPieperFR1996Expression of cDNA-encoded human acid alpha-glucosidase in milk of transgenic miceBiochimica et biophysica acta13089368764823
  • BijvoetAGKroosMAPieperFR1998Recombinant human acid alpha-glucosidase: high level production in mouse milk, biochemical characteristics, correction of enzyme deficiency in GSDII KO miceHum Mol Genet71815249736785
  • BijvoetAGVan HirtumHKroosMA1999Human acid alpha-glucosidase from rabbit milk has therapeutic effect in mice with glycogen storage disease type IIHum Mol Genet821455310545593
  • BoerkoelCFExelbertRNicastriC1995Leaky splicing mutation in the acid maltase gene is associated with delayed onset of glycogenosis type IIAm J Hum Genet56887977717400
  • BradyRO2006Enzyme replacement for lysosomal diseasesAnnu Rev Med572839616409150
  • BradyROPentchevPGGalAE1974Replacement therapy for inherited enzyme deficiency. Use of purified glucocerebrosidase in Gaucher’s diseaseN Engl J Med291989934415565
  • CaseLEHannaRFrushDP2007Fractures in children with Pompe disease: a potentiallong-term complicationPediatr Radiol374374517342521
  • ChamolesNABlancoMGaggioliD2001Diagnosis of alpha-L-iduronidase deficiency in dried blood spots on filter paper: the possibility of newborn diagnosisClin Chem47780111274042
  • ChamolesNABlancoMGaggioliD2002aGaucher and Niemann-Pick diseases–enzymatic diagnosis in dried blood spots on filter paper: retrospective diagnoses in newborn-screening cardsClin Chim Acta317191711814475
  • ChamolesNABlancoMGaggioliD2002bTay-Sachs and Sandhoff diseases: enzymatic diagnosis in dried blood spots on filter paper: retrospective diagnoses in newborn-screening cardsClin Chim Acta318133711880123
  • ChamolesNANiizawaGBlancoM2004Glycogen storage disease type II: enzymatic screening in dried blood spots on filter paperClin Chim Acta3479710215313146
  • ChienYHLeeNCPengSF2006Brain development in infantile-onset Pompe disease treated by enzyme replacement therapyPediatr Res603495216857770
  • CookALKishnaniPSCarboniMP2006Ambulatory electrocardiogram analysis in infants treated with recombinant human acid alpha-glucosidase enzyme replacement therapy for Pompe diseaseGenet Med83131716702882
  • DanonMJOhSJDiMauroS1981Lysosomal glycogen storage disease with normal acid maltaseNeurology315176450334
  • De DuveC1963The lysosomeSci Am208647214025755
  • de DuveC2005The lysosome turns fiftyNat Cell Biol7847916136179
  • FurbishFSBlairHEShiloachJ1977Enzyme replacement therapy in Gaucher’s disease: large-scale purification of glucocerebrosidase suitable for human administrationProc Nat Acad Sci USA7435603269414
  • GambettiPDiMauroSBakerL1971Nervous system in Pompe’s disease. Ultrastructure and biochemistryJ Neuropathol Exp Neurol30412305284681
  • HermansMMvan LeenenDKroosMA2004Twenty-two novel mutations in the lysosomal alpha-glucosidase gene (GAA) underscore the genotype-phenotype correlation in glycogen storage disease type IIHum Mutat23475614695532
  • HersHG1963Alpha-Glucosidase deficiency in generalized glycogenstorage disease (Pompe’s disease)Biochem J86111613954110
  • HirschhornRReuserAJJSchriverCRBeaudetALSlyWSValleD2001Glycogen storage disease type II: acid alpha-glucosidase (acid maltase) deficiencyThe Metabolic and Molecular Bases of Inherited DiseaseMcGraw-HillNew York3389420
  • HowellRRByrneBDarrasBT2006Diagnostic challenges for Pompe disease: an under-recognized cause of floppy baby syndromeGenet Med82899616702878
  • HuieMLHirschhornRChenAS1994Mutation at the catalytic site (M519V) in glycogen storage disease type II (Pompe disease)Hum Mutat429137866409
  • HunleyTECorzoDDudekM2004Nephrotic syndrome complicating alpha-glucosidase replacement therapy for Pompe diseasePediatrics114e532515466083
  • KikuchiTYangHWPennybackerM1998Clinical and metabolic correction of pompe disease by enzyme therapy in acid maltase-deficient quailJ Clin Investig101827339466978
  • KishnaniPSCorzoDNicolinoM2007Recombinant human acid [alpha]-glucosidase: major clinical benefits in infantile-onset Pompe diseaseNeurology689910917151339
  • KishnaniPSHwuWLMandelH2006aA retrospective, multinational, multicenter study on the natural history of infantile-onset Pompe diseaseJ Pediatr148671616737883
  • KishnaniPSNicolinoMVoitT2006bChinese hamster ovary cell-derived recombinant human acid alpha-glucosidase in infantile-onset Pompe diseaseJ Pediatr149899716860134
  • KishnaniPSSteinerRDBaliD2006cPompe disease diagnosis and management guidelineGenet Med82678816702877
  • KlingeLStraubVNeudorfU2005aSafety and efficacy of recombinant acid alpha-glucosidase (rhGAA) in patients with classical infantile Pompe disease: results of a phase II clinical trialNeuromuscul Disord15243115639117
  • KlingeLStraubVNeudorfU2005bEnzyme replacement therapy in classical infantile pompe disease: results of a ten-month follow-up studyNeuropediatrics3661115776317
  • KoTMHwuWLLinYW1999Molecular genetic study of Pompe disease in Chinese patients in TaiwanHum Mutat13380410338092
  • KroosMAVan der KraanMVan DiggelenOP1995Glycogen storage disease type II: frequency of three common mutant alleles and their associated clinical phenotypes studied in 121 patientsJ Med Genet32836378558570
  • LiYScottCRChamolesNA2004Direct multiplex assay of lysosomal enzymes in dried blood spots for newborn screeningClin Chem5017859615292070
  • LinCYShiehJJ1996Molecular study on the infantile form of Pompe disease in Chinese in TaiwanZhonghua Minguo xiao er ke yi xue hui za zhi [Journal]3711521
  • MarsdenD2005Infantile onset Pompe disease: a report of physician narratives from an epidemiologic studyGenet Med71475015714084
  • MartiniukFChenAMackA1998Carrier frequency for glycogen storage disease type II in New York and estimates of affected individuals born with the diseaseAm J Med Genet7969729738873
  • NishinoIFuJTanjiK2000Primary LAMP-2 deficiency causes X-linked vacuolar cardiomyopathy and myopathy (Danon disease)Nature4069061010972294
  • NishiokaTTomatsuSGutierrezMA2006Enhancement of drug delivery to bone: characterization of human tissue-nonspecific alkaline phosphatase tagged with an acidic oligopeptideMol Genet Metab882445516616566
  • PompeJC1932Over idiopatische hypertrophie van het hartNed Tijdshr Geneeskd76304
  • PoorthuisBJWeversRAKleijerWJ1999The frequency of lysosomal storage diseases in The NetherlandsHum Genet105151610480370
  • RabenNDanonMGilbertAL2003Enzyme replacement therapy in the mouse model of Pompe diseaseMolecular genetics and metabolism801596914567965
  • RabenNPlotzPByrneBJ2002Acid alpha-glucosidase deficiency (glycogenosis type II, Pompe disease)Curr Mol Med21456611949932
  • RosenbergMKingmaWFitzpatrickMA1999Immunosurveillance of alglucerase enzyme therapy for Gaucher patients: induction of humoral tolerance in seroconverted patients after repeat administrationBlood932081810068682
  • RosenbloomBEWeinrebNJZimranA2005Gaucher disease and cancer incidence: a study from the Gaucher RegistryBlood10545697215718419
  • SakuraiITosakaAMoriY1974Glycogenosis type II (Pompe). The fourth autopsy case in JapanActa Pathol Jpn24829464281990
  • SchramAWBrouwer-KelderBDonker-KoopmanWE1979Use of immobilized antibodies in investigating acid alpha-glucosidase in urine in relation to Pompe’s diseaseBiochimica et biophysica acta5673708336157
  • ShinYS2006Glycogen storage disease: clinical, biochemical, and molecular heterogeneitySemin Pediatr Neurol131152017027861
  • ShinYSEndresWUnterreithmeierJ1985Diagnosis of Pompe’s disease using leukocyte preparations. Kinetic and immunological studies of 1,4-alpha-glucosidase in human fetal and adult tissues and cultured cellsClin Chim Acta1489193891151
  • TaniguchiNKatoEYoshidaH1978alpha-glucosidase activity in human leucocytes: choice of lymphocytes for the diagnosis of Pompe’s disease and the carrier stateClin Chim Acta892939361294
  • ThurbergBLLynch MaloneyCVaccaroC2006Characterization of pre- and post-treatment pathology after enzyme replacement therapy for pompe diseaseLab Invest8612082017075580
  • TomatsuSMontanoAMGutierrezM2007Characterization and pharmacokinetic study of recombinant human N-acetylgalactosamine-6-sulfate sulfataseMol Genet Metab91697817336563
  • TsujinoSHuieMKanazawaN2000Frequent mutations in Japanese patients with acid maltase deficiencyNeuromuscul Disord1059960311053688
  • UmapathysivamKHopwoodJJMeiklePJ2001Determination of acid alpha-glucosidase activity in blood spots as a diagnostic test for Pompe diseaseClin Chem4713788311468225
  • UmapathysivamKHopwoodJJMeiklePJ2005Correlation of acid alpha-glucosidase and glycogen content in skin fibroblasts with age of onset in Pompe diseaseClin Chim Acta361191815993875
  • UmapathysivamKWhittleAMRanieriE2000Determination of acid alpha-glucosidase protein: evaluation as a screening marker for Pompe disease and other lysosomal storage disordersClin Chem4613182510973860
  • Van den HoutHReuserAJVultoAG2000Recombinant human alpha-glucosidase from rabbit milk in Pompe patientsLancet356397810972374
  • van den HoutHMHopWvan DiggelenOP2003The natural course of infantile Pompe’s disease: 20 original cases compared with 133 cases from the literaturePediatrics1123324012897283
  • Van den HoutJMKamphovenJHWinkelLP2004Long-term intravenous treatment of Pompe disease with recombinant human alpha-glucosidase from milkPediatrics113e4485715121988
  • Van HoveJLYangHWOliverLM1997Purification of recombinant human precursor acid alpha-glucosidaseBioch Mol Biol Int4361323
  • Van HoveJLYangHWWuJY1996High-level production of recombinant human lysosomal acid alpha-glucosidase in Chinese hamster ovary cells which targets to heart muscle and corrects glycogen accumulation in fibroblasts from patients with Pompe diseaseProc Nat Acad Sci USA9365708552676
  • WenkJHilleAvon FiguraK1991Quantitation of Mr 46000 and Mr 300000 mannose 6-phosphate receptors in human cells and tissuesBiochem Int23723311651728
  • WilcoxWRBanikazemiMGuffonN2004Long-term safety and efficacy of enzyme replacement therapy for Fabry diseaseAm J Hum Genet75657415154115
  • WinkelLPVan den HoutJMKamphovenJH2004Enzyme replacement therapy in late-onset Pompe’s disease: a three-year follow-upAnn Neurol5549550215048888
  • YangHWKikuchiTHagiwaraY1998Recombinant human acid alpha-glucosidase corrects acid alpha-glucosidase-deficient human fibroblasts, quail fibroblasts, and quail myoblastsPediatr Res43374809505277
  • ZhuYLiXKyazikeJ2004Conjugation of mannose 6-phosphate-containing oligosaccharides to acid alpha-glucosidase improves the clearance of glycogen in pompe miceJ Biol Chem279503364115383547
  • ZhuYLiXMcVie-WylieA2005Carbohydrate-remodelled acid alpha-glucosidase with higher affinity for the cation-independent mannose 6-phosphate receptor. demonstrates improved delivery to muscles of Pompe miceBiochem J3896192815839836

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.