Next Article in Journal
Features of Protein Unfolding Transitions and Their Relation to Domain Topology Probed by Single-Molecule FRET
Next Article in Special Issue
Advances in Dystrophinopathy Diagnosis and Therapy
Previous Article in Journal
Sequences of Alterations in Inflammation and Autophagy Processes in Rd1 Mice
Previous Article in Special Issue
Effects of Triheptanoin on Mitochondrial Respiration and Glycolysis in Cultured Fibroblasts from Neutral Lipid Storage Disease Type M (NLSD-M) Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comprehensive Update on Late-Onset Pompe Disease

1
Department of Clinical and Experimental Sciences, University of Brescia, 25100 Brescia, Italy
2
Unit of Neurology, ASST Spedali Civili, 25100 Brescia, Italy
3
NeMO-Brescia Clinical Center for Neuromuscular Diseases, 25064 Brescia, Italy
*
Author to whom correspondence should be addressed.
Submission received: 17 June 2023 / Revised: 10 August 2023 / Accepted: 21 August 2023 / Published: 22 August 2023

Abstract

:
Pompe disease (PD) is an autosomal recessive disorder caused by mutations in the GAA gene that lead to a deficiency in the acid alpha-glucosidase enzyme. Two clinical presentations are usually considered, named infantile-onset Pompe disease (IOPD) and late-onset Pompe disease (LOPD), which differ in age of onset, organ involvement, and severity of disease. Assessment of acid alpha-glucosidase activity on a dried blood spot is the first-line screening test, which needs to be confirmed by genetic analysis in case of suspected deficiency. LOPD is a multi-system disease, thus requiring a multidisciplinary approach for efficacious management. Enzyme replacement therapy (ERT), which was introduced over 15 years ago, changes the natural progression of the disease. However, it has limitations, including a reduction in efficacy over time and heterogeneous therapeutic responses among patients. Novel therapeutic approaches, such as gene therapy, are currently under study. We provide a comprehensive review of diagnostic advances in LOPD and a critical discussion about the advantages and limitations of current and future treatments.

1. Introduction

Glycogen storage disease type II (GSDII), also named Pompe disease (PD), is an autosomal recessive disorder caused by a deficiency in the acid alpha-glucosidase enzyme (GAA), which is responsible for the hydrolysis of glycogen to glucose in the lysosome [1,2].
In the past, the prevalence and expected frequency of PD were reported to be around 1 in 40,000 births [3,4,5,6]. However, thanks to the implementation of newborn screening (NBS), the prevalance seems to be much higher, ranging from 1.0 per 4447 to 1.0 per 37,094, with differences between countries related to ethnic factors and screening tools [7,8,9,10,11,12,13,14,15,16,17,18]. Data from the main epidemiological studies are detailed in Table 1.
The GAA gene (OMIM *606800) is located on chromosome 17q25 and it is the only gene associated with Pompe disease (OMIM #232300) in the online Mendelian Inheritance in Man Database [19,20]. It encodes a 110 kDa precursor polypeptide that undergoes several post-translational modifications in the rough endoplasmic reticulum (ER) [19,20,21]. After N-glycosylation and proper folding in the ER, the enzyme is transported to the Golgi, where it acquires mannose 6-phosphate (M6P), which is required to target the lysosome via the cation-independent mannose 6-phosphate receptor (CI-M6P) pathway [21,22,23,24,25,26]. CI-M6P receptors are transmembrane glycoproteins located mostly in the trans-Golgi and endosomal compartments, but also at low levels at the cell surface [21]. CI-M6P receptor cycling over the cell surface is crucial for enzyme replacement therapy (ERT) because it represents the key to enzyme access in the cell [21,27].
GAA, contained in a vesicle that pinches off from the Golgi to the endosomes, undergoes a series of proteolytic and N-glycan processing steps that are necessary for its activation [21,22,28,29,30]. In patients affected by PD, the defective enzyme is not able to catalyze hydrolysis of the α-1,4 and α-1,6-glucosidic bonds in glycogen, thus leading to increased glycogen storage in lysosomes, which is responsible for lysosomal dysfunction, enlargement, and finally rupture [28,31]. However, the pathogenesis of PD is not simply related to the presence of damaged lysosomes; defective autophagy, dysregulation of lysosome-based signaling pathways (i.e., the nutrient-sensitive mammalian target of rapamycin complex 1—mTORC1), oxidative stress, and mitochondrial abnormalities take part in the pathogenic cascade and contribute to altering muscle architecture with displacement of the myofibrils [19,28,31].
Table 1. Data from main epidemiological studies on Pompe disease.
Table 1. Data from main epidemiological studies on Pompe disease.
Methods and SourcePeriod
of Study
Expected Frequency of GSDII CasesPopulation and CountryReference
Estimation of the frequency of PD by determining carrier status in a randomly selected healthy population for seven of the most common mutations.19981.0 per 40,000N = 928 healthy individuals
Five different race groups.
Mostly aged between 20 and 50 y.o.
USA
Martiniuk F et al. Am J Med Genet. 1998 [4]
Calculation of the frequency of PD by screening a random sample of newborns for three frequent mutations in the acid alpha-glucosidase gene (IVS1(-13T®G), 525delT and delexon18). The screening was performed on DNA extracted from over 3000 Guthrie cards from Dutch neonates.19991.0 per 40,000 (1.0 per 138,000 for infantile GSDII; 1.0 per 57,000 for adult GSDII)N = over 3000 Guthrie cards from
Dutch neonates
Netherlands
Ausems M. et al. Eur J Hum Genet. 1999 [5]
Calculation of birth prevalence of lysosomal storage diseases (LSDs) based on records from the laboratories of the clinical genetic centers involved in the post- and prenatal diagnosis of LSDs.1970–1996Birth prevalence of 2.0 per 100,000
live births
N = 963 enzymatically confirmed
cases
Netherlands
Poorthuis BJ et al. Hum Genet. 1999 [3]
Calculation of birth prevalence of each LSD by dividing the total number of diagnosed cases (post- and prenatal diagnosis) by the total number of live births that occurred between the years of birth of the older and younger patients (birth period).1940–1999Birth prevalence of 0.17 per 100,000Data from total N of live births from Instituto Nacional de Estatistica, Porto
North Portugal
Pinto R et al. Eur J Hum Genet. 2004 [6]
Frequency of PD assessed by newborn screening program.20101.0 per 8684N = 34,736 newborns screened
Austria
Mechtler TP et al. Lancet. 2012 [15]
Frequency of PD assessed by newborn screening program.20111.0 per 4447N = 40,024 newborns screened
Hungary
Wittmann J et al. JIMD Rep. 2012 [12]
Frequency of PD assessed by newborn screening program.2013–20161.0 per 34,401N = 103,204 newborns screened
Japan
Momosaki K et al. Hum Genet. 2019 [18]
Frequency of PD assessed by newborn screening program.2016–20191.0 per 16,095N = 531,139 newborns screened
Pennsylvania, USA
Ficicioglu C et al. Int J Neonatal Screen. 2020 [8]
Frequency of PD assessed by newborn screening program.2014–20191.0 per 23,596N = 684,290 newborns screened
Illinois, USA
Burton BK et al. Int J Neonatal Screen. 2020 [14]
Frequency of PD assessed by newborn screening program.2018–20191.0 per 25,200N = 453,152 screened newborns
California, USA
Tang H et al. Int J Neonatal Screen. 2020 [9]
Frequency of PD assessed by newborn screening program.2013–20181.0 per 10,152N = 476,000 screened newborns
Missouri, USA
Klug TL et al. Int J Neonatal Screen. 2020 [10]
Frequency of PD assessed by newborn screening program.20191.0 per 19,777N = 59,332 screened newborns
Georgia, USA
Hall PL et al. Int J Neonatal Screen. 2020 [16]
Frequency of PD assessed by newborn screening program.2013–20201.0 per 37,094N = 296,759 newborns screened
Japan
Sawada T et al. Orphanet J Rare Dis. 2021 [7]
Frequency of PD assessed by newborn screening program.2015–20221.0 per 18,432N = 206,741 screened newborns
Italy
Gragnaniello V et al. Mol Genet Metab Rep. 2022 [11]

2. Phenotypes and Phenotypic Heterogeneity

PD represents a spectrum of phenotypes that differ in age of onset, organ involvement, severity of disease, and rate of progression [1,2]. PD phenotypes are named according to age at onset, with infantile, childhood, juvenile, and adult forms [32,33,34,35,36,37,38]. Infantile-onset Pompe disease encompasses classical and non-classical infantile forms [32,33]. The classical infantile form usually presents in the first days or weeks of life, and it is associated with cardiomegaly and hypertrophic cardiomyopathy [32,33]. Skeletal impairment manifests with feeding difficulties, hypotonia, and respiratory distress [32,33]. Death occurs within the first two years of life due to progressive cardiac and respiratory insufficiency [32,33]. The latter is mainly caused by weakness of the diaphragmatic and abdominal wall muscles, especially the internal obliques and rectus abdominis [32,33]. In this review, the acronym “IOPD” refers to the classical infantile form.
The term “non-classical infantile form” is applied to infants under 1 year presenting with less severe cardiac involvement and milder motor symptoms, allowing longer survival and better motor development [32,33].
The term “LOPD” is applied to patients with an onset between 12 months and adulthood (childhood, juvenile, and adult forms) [34,35,36,37,38,39]. The LOPD phenotype may be extremely variable, encompassing cases with mild muscle involvement to those with severe chronic respiratory failure and marked limb-girdle and axial weakness [34,35,36,37,38,39].
In common use, the term “LOPD” is applied also to asymptomatic patients who harbor LOPD mutations in the GAA gene diagnosed because of family history, elevated serum creatine kinase levels, or NBS, and whose clinical expression is not predictable at diagnosis [35,36,37,38].
The heterogeneous severity of the disease is related to the residual activity of the GAA enzyme in tissues [20,40]. Some GAA mutations are particularly deleterious and cause a complete lack of enzyme, while other less severe mutations allow a certain residual enzyme activity [36,41,42,43,44,45]. Known mutations in the GAA gene are reported in the “Pompe Disease GAA variant database” (http://www.pompevariantdatabase.nl, accessed on 1 May 2023). Details of the most common variants are provided in Table 2.
In the genotype-phenotype report from the Pompe Registry published in 2019, the non-sense variant c.2560C>T is frequently reported in IOPD, tracing its origins from Northern Africa and being diffuse in North and Latin America due to the slave trade [42]. The missense c.1935C>A variant is the most frequent variant in IOPD patients from the Asia-Pacific area and is frequently associated with the pseudo-deficiency variant c.1726G>A [42].
The c.-32–13T>G variant is typically associated with the LOPD phenotype spectrum [45,46,47]. Earlier onset in patients with the c.-32–13T>G variant is possibly related to the concomitant presence of more deleterious variants in GAA, such as c.510C>T [46,47].
Among the most frequent variants, the c.525delT variant and exon 18 deletion can be associated with either IOPD or LOPD, depending partially on the nature of the other allele [42]. The presence of a variant close to the splice site can affect splicing and may be predictive of LOPD, while splice site variants, non-sense variants, deletions, and insertions are often associated with IOPD [42].
However, genetics cannot alone determine the phenotypic variability in PD and cannot explain, for example, the variability in terms of age of onset, severity, and distribution often observed in individuals carrying the same mutations [48,49,50]. Variants in modifier genes have been proposed to influence these characteristics, such as ACE and ACTN3 polymorphisms that are known to affect fiber type composition and muscle properties [49]. Nevertheless, a recent clinical trial with 24 families did not show clear influence of ACE I/D polymorphism on clinical presentation or response to ERT, as patients with discordant ACE genotypes had similar disease courses while siblings with the same ACE genotype presented with different disease courses [51].
Indeed, more studies are needed to investigate the possible influence on clinical variability of epigenetic and trans-acting factors that may regulate lysosomal or skeletal muscle homeostasis [48,50].

3. Clinical Assessment and Diagnostic Tools

Family history of muscle diseases, age of onset, and course of disease should always be carefully assessed. Progressive limb-girdle weakness is one of the most frequent features reported by patients with adult-onset LOPD and often the main reason for clinician evaluation [52]. However, red flags include isolated respiratory muscle involvement and isolated elevated serum creatine kinase levels, found in around 2.5% of patients in population studies [52].
Neurological examination should include evaluation of trunk muscles that can be affected early in LOPD, even though their involvement is difficult to clinically assess because the ability to rise from the supine position or raise the trunk from the prone position requires activation of different muscle groups and may be influenced by osteoarticular conditions or respiratory dysfunction [53]. The presence of scoliosis may suggest weakness of trunk muscles [53].
Tongue weakness can be an early sign of disease and should always be evaluated, as it may be useful in differentiating LOPD from other forms of myopathy [54]. Clinical assessment of tongue weakness can be later confirmed through magnetic resonance imaging (MRI) using the so-called “bright tongue sign,” which unfortunately may be missed or unreported by non-expert radiologists [54,55,56,57].
Laboratory tests usually reveal a mild increase in creatine kinase (CK) levels in LOPD, although these can sometimes be normal [58,59,60,61,62]. Elevation of plasma cardiac troponin T (cTnT) levels, which reflects skeletal muscle damage, can also be found and may erroneously lead to the diagnosis of myocardial infarction or injury [63]. Commonly, elevated alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels are found and should be considered red flags for suspicion of metabolic myopathies [64]. However, all of these laboratory findings are nonspecific and have limited prognostic value.
Electromyography (EMG), other than showing a myopathic pattern with spontaneous activity, may detect complex repetitive discharges and electrical myotonia in axial muscles, which always raises suspicion of the LOPD phenotype spectrum [65,66]. Rare cases of neurogenic pattern are reported in the literature, leading to misdiagnosis as spinal muscular atrophy [67].
Muscle MRI represents a promising diagnostic tool for early diagnosis and follow-up in the LOPD phenotype spectrum [68,69,70]. The most typical MRI pattern is systematic axial muscle involvement, especially in trunk extensors [71]. These abnormalities may occur even in the pre-symptomatic stage of disease, possibly driving clinical suspicion [71]. Paravertebral atrophy is not specific and may be detected in other inherited or acquired myopathies, but its association with abdominal belt weakness is not commonly observed in other diseases [72,73,74]. Mild or moderate involvement of the adductor magnus muscle may be evident in the early stages of disease, followed by the semimembranosus, and later, the long head of the biceps femoris and the semitendinosus [75,76]. Fat replacement in knee extensors is usually detected in more advanced stages of disease, with selective sparing of the rectus femoris, sartorius, and gracilis muscles; this pattern can be useful in differential diagnosis from other muscular diseases, such as dystrophies [75,76]. In the upper limbs, involvement of the trapezius inferior, rhomboid, and subscapularis muscles with later moderate damage of the serratus anterior, deltoid, and supraspinatus muscles is usually described [37,71]. Diaphragmatic MRI abnormalities may be present, even when functional pulmonary tests are still within normal range [77,78,79].
Muscle biopsy may be performed using standard procedures in histological and histochemical studies, including Periodic acid-Schiff (PAS) staining for identifying glycogen accumulation and acid phosphatase staining as a marker of lysosomal abnormalities [80,81]. The typical pathological pattern is characterized by vacuolar myopathy with glycogen storage and increased acid phosphatase reactivity (Figure 1) [80,81]. Fiber atrophy is more evident in IOPD and childhood forms, with type 1 and type 2 fibers equally involved [80,81]. The presence of internal nuclei and necrosis is also reported in adult cases [81]. The number of vacuoles and amount of glycogen accumulation are variable and usually more prominent in IOPD than in adult forms, in which histological studies may also show mild involvement or, in some cases, normal appearance [81,82]. In a French study of 21 patients with LOPD, muscle biopsy was negative in 30%, despite confirmation by a positive blood-based enzyme assay [83]. Correct conservation of the sample and optimal fixation, embedding, and staining methods may play a role, and PAS on a semi-thin section has been found to be more reliable, allowing better visualization of glycogen-filled vacuoles, regardless of their size [81,84,85]. It has also been suggested that detection of large lipofuscin inclusions within the area of autophagic accumulation using high-resolution light microscopy can be suggestive of PD [86]. Genetic analysis is considered the gold standard for a definitive diagnosis and can be performed on blood, cheek swab, muscle tissue, or saliva samples [87,88]. While in the past the diagnostic process always included muscle biopsy and enzyme activity evaluation followed by targeted genetic studies, technological advances in genetics have greatly changed clinical practice [87,88]. The availability of NGS panels that allow rapid analysis of a large number of genes has meant that, often, genetic study has become the first diagnostic step following clinical suspicion [87,88]. However, if variants of undefined pathogenetic significance are found, muscle biopsy and biochemical assays remain of great importance in defining their role in causing disease. A biochemical assay of GAA activity can be performed in mixed leukocytes, fibroblasts, or muscle tissue [40,58,88,89,90]. In general, GAA activity is extremely reduced in IOPD patients (<1%), while LOPD patients have 2–30% of normal GAA activity [40,91].
In many centers, faced with suspicion of LOPD based on clinical red flags such as limb-girdle or axial weakness or elevated serum creatine kinase levels, screening is performed by assaying acid alpha-glucosidase activity on a dried blood spot (DBS) via tandem mass spectrometry or fluorometry [92,93]. It is frequently a first-line test because it has the advantages of being easy to perform, inexpensive, minimally invasive, and able to provide rapid results [92,93,94,95,96,97]. In the past, discrepancies in GAA activity could be related to isozymes (e.g., renal isozyme or isoenzyme maltase-glucoamylase) that showed activity at the (acidic) pH used to measure activity, interfering with the results of clinical assays [95,96]. For this reason, newer blood assays use acarbose to competitively inhibit maltase-glucoamylase [90,96,97,98]. Also, incorrect blood spotting and the combination of humidity and heat caused by insufficient drying or inappropriate shipping can interfere with enzyme stability and, consequently, the assessment of GAA levels [98]. Rarely, positive DBS with no pathogenic variant may also be related to pseudodeficiency alleles [40,99,100]. As a screening test for LOPD, quantification of PAS-positive lymphocytes in peripheral blood has recently demonstrated its usefulness [101]. Moreover, a decrease in the percentage of PAS-positive lymphocytes in a small subgroup of LOPD patients suggests that it could be useful as a therapy response biomarker [101]. Urinary glucose tetrasaccharide (Glc4) could be used as a second-tier test after positive DBS, and its concentration seems to correlate with age of symptom onset, with more elevated levels in IOPD patients [102,103,104,105]. However, elevated urinary tetrasaccharide glucose levels are not specific to PD and may also be related to urinary infections, acute pancreatitis, muscular trauma, and some cancers [102]. Defining reference intervals is still being debated, as some affected patients can have normal Glc4 excretion in the early phases of disease [102,104]. A summary of the main clinical findings in LOPD is reported in Table 3.

4. Multisystem Involvement and Indications for Follow-up and Management

A multidisciplinary approach for adequate management and follow-up is fundamental, as respiratory dysfunction and cardiac involvement impact the survival of patients [37]. Practical recommendations for pulmonary assessment include an anamnestic questionnaire, clinical examination, and pulmonary functioning tests [106]. If patients complain of morning headaches or excessive daily sleepiness, an overnight cardiorespiratory polysomnography is recommended to rule out nocturnal hypoventilation and the development of chronic daytime hypercapnia [107]. To assess respiratory failure, it is important to measure forced vital capacity (FVC) both in seated and supine positions: a drop of over 25% in the percentage of predicted FVC upon changing posture from the upright to the supine position is suggestive of diaphragmatic weakness [108,109,110]. As weakness of the diaphragm may be an early sign and evolves independently to skeletal muscle status, close follow-up is recommended, and pulmonary function should be monitored routinely once or twice per year [106,111]. Clinical indications for the introduction of non-invasive or invasive ventilation are detailed in the American Thoracic Society and European Respiratory Society guidelines and in the paper by Boentert et al. [106,112].
A standard cardiac assessment, including clinical history, physical examination, a 12-lead electrocardiogram, and echocardiography, is required for each patient and may be repeated every 1–2 years in case of normal findings [113]. Severe cardiac involvement is present in IOPD and may lead to left ventricular (LV) outflow obstruction resulting in fatal cardiac failure [114,115]. Right ventricular systolic function is usually preserved in LOPD, while diastolic LV dysfunction has been rarely reported [116,117,118,119,120]. Atrial enlargement may be detected as a consequence of poor ventricular compliance and diastolic dysfunction [119]. A Holter electrocardiogram should be performed in order to rule out rhythm disturbances such as Wolff–Parkinson–White syndrome, often in association with a short PR interval or second-degree atrioventricular block [120,121,122]. However, cardiac involvement in LOPD has been recently questioned, as prevalence may be biased by the lack of data on established cardiovascular risk factors (e.g., obesity, hypertension, dyslipidemia, diabetes mellitus) among patients in most cohort studies [123]. A periodic neurological follow-up in LOPD should include clinical evaluation by assessment of muscle strength and basic functional tests (e.g., Gowers’ maneuver), combined with laboratory tests (e.g., CK) and muscle MRI [124]. Cerebral MRI with sequences for intracranial and extracranial large vessel study should be included at baseline and repeated every two years, as cerebrovascular manifestations (e.g., aneurysms, vertebrobasilar dolichoectasia, dilatative arteriopathy) have been reported [125,126,127,128]. If an intracranial aneurysm is identified, the first step is assessing risk factor stratification for rupture depending on aneurysm-related (e.g., size or shape) and patient-related (e.g., age, cardiovascular risk factors) findings [129]. If a high-risk profile is defined, endovascular treatment is indicated [129]. Evidence of small vessel disease in LOPD is reported in the literature, although data corrected for baseline cardiovascular risk factors are few [130]. Differently, IOPD patients may present with diffuse leukoencephalopathy related to glycogen storage on the CNS, affecting mostly periventricular white matter and centrum semiovale areas, associated with cognitive decline [131,132]. The impact on cognitive profile seems to be less severe in LOPD, although functional disruption of neuronal networks involved in executive functioning, planning, and abstract reasoning was found even without grey matter atrophy in small cohort studies [130,133].
Other organ involvement is usually assessed according to the patient’s referred symptoms. For example, degeneration of bones and joints as a consequence of muscle weakness is a frequent finding, as scoliosis, kyphosis, and hyperlordosis are usually linked to abdominal and hip extensor weakness [37]. Rarely, more severe phenotypes may present with rigid spine syndrome, characterized by limited flexion of the cervical and dorsolumbar spines, or bent spine syndrome in which weakness of extensor spine muscles causes a pathological anterior curvature of the thoracolumbar tract [37,134]. Routine spine and femoral radiography with bone densitometry are recommended periodically because they may unveil asymptomatic and atraumatic vertebral fractures that are not always related to significant impairment of bone mass [37,134].

5. Future Perspectives in Monitoring LOPD Symptoms

While Glc4 was initially applied to monitor the response to ERT with only mild results, more recent studies have focused on serum microRNA profiles as potential biomarkers of disease [135,136]. Elevated levels of so-called dystromirs (miR-1-3p, miR-133a-3p, and miR-206), which are microRNAs involved in muscle regeneration, have been found in PD and other neuromuscular diseases [136]. In a sample of 35 patients with LOPD, miR-206 levels were significantly higher in serum samples of symptomatic patients compared to asymptomatic patients, suggesting that this microRNA could be used to follow up presymptomatic patients and guide the timing of treatment [136]. Correlations between serum levels and severity of disease were also observed in the plasma samples of 52 patients, where miR-133a levels were higher in infantile-onset patients than in late-onset subjects [137]. Although promising, these recent data are applied only in research studies for now and need further confirmation.
Meanwhile, in the past 10 years, muscle MRI techniques have developed and emerged as an interesting tool not only for diagnosis but also for follow-up in LOPD. A recent study performed with conventional T1-TSE sequences and assessment of fat replacement with Mercuri scoring suggested the stabilization of the fat fraction of the psoas muscle under ERT, yet several follow-up MRI studies were able to detect the slow progression of the skeletal muscle fat fraction by using more sensitive sequences such as water/fat (Dixon) [138,139,140]. As fatty muscle replacement can occur before clinical weakness, and mildly affected muscles seem to respond better to ERT than severely involved ones, MRI studies may help to decide when to start ERT in clinically asymptomatic patients [141].
Although muscle MRI is the gold standard in imaging evaluation in order to follow up and evaluate treatment response, ultrasonography could be an interesting alternative if muscle MRI is not available, as a recent study suggested it to be efficient in evaluating diaphragm dysfunction [142].

6. Treatment Approaches

The main current and future therapeutic approaches are summarized in Table 4.

6.1. Enzyme Replacement Therapy (ERT)

Disease severity increases with disease duration, and past observational studies have shown that, every year from diagnosis, the odds for wheelchair use increase by 13% and the odds for respiratory support increase by 8% [143]. In 2006, perspectives changed thanks to the approval of ERT based on the intravenous infusion of a recombinant human GAA (rhGAA) precursor enzyme for IOPD [21,144]. The uptake of rhGAA from the bloodstream into the cells is driven by the M6P receptors, which later target rhGAA to the lysosomes, where it can be turned into the active form [19,21,144].
In IOPD patients, rhGAA (alglucosidase alfa) clearly improved survival with a decreased risk of invasive ventilation and preserved cardiac function, yet a therapeutic problem emerged in the so-called IOPD CRIM-negative patients, who have two deleterious GAA mutations and are completely unable to make native enzyme [145]. IOPD CRIM-negative patients had poorer clinical outcomes because of their immunological response to rhGAA, so immunomodulatory therapy was recommended in order to prevent this side effect [145,146,147].
After promising results in IOPD patients, a randomized, placebo-controlled trial with alglucosidase alfa (LOTS trial) was conducted with 90 LOPD patients (8 years old or older; mean age at first infusion: 45.3 for the treated group and 42.6 for the placebo group) [148]. Patients received biweekly intravenous alglucosidase alfa (20 mg per kilogram of body weight) or placebo for 78 weeks at eight centers in the United States and Europe, and the primary endpoints (improvement of distance walked during a 6-min walk test and percentage of predicted FVC) were reached [148]. The open-label extension LOTS trial and several other studies confirmed the stabilization of motor and pulmonary function, which correlated with reduced lysosomal glycogen levels after ERT in the EMBASSY study [148,149,150,151,152,153,154].
Interestingly, the reduction in vacuolated fibers and PAS-positive material in muscle biopsies after ERT was more marked in less affected fibers and in smaller PAS-positive collections, thus underscoring that the efficacy of ERT was greater when initiated in the early stages of disease [155]. However, according to the European consensus, clinical or instrumental manifestation of PD is required to start treatment, which can be later stopped only in cases of severe infusion-associated reactions, immunogenicity, or loss of efficacy for more than two years [156]. Follow-up studies revealed significant variability in response to therapy [157,158,159,160,161,162,163,164]. Usually, LOPD patients very rarely develop high titers of neutralizing anti-rhGAA antibodies and, therefore, immunogenicity would not explain the variable therapeutic response [155,157,158,159,160,161,162,163]. A mild or moderate improvement in LOPD patients may be observed during the first 2 to 3 years of treatment, followed later by a plateau or slight decline [157,158,159]. A recent 10-year follow-up study confirmed this trend and underlined the variability in duration of the plateau phase among LOPD patients [164]. Younger age and better clinical status for supine FVC or muscle strength were initially identified as good prognostic factors for response to treatment, yet long-term follow-up in treated patients did not confirm this evidence [152,157,163]. The reasons for the progressive decline are still unclear. Several hypotheses have been proposed, including possible limited biodistribution of rhGAA and reduced intracellular uptake due to low expression of the CI-M6P receptors on skeletal muscle cells [165].
One of the first options to overcome limited therapeutic efficacy was to design novel rhGAAs, such as avalglucosidase, produced by chemical conjugation of an oligosaccharide harboring bis-M6P residues onto recombinant human GAA via oxime chemistry, thus increasing affinity for the CI-M6P receptors and achieving larger distribution in systemic tissues [166]. After the first positive results of avalglucosidase in the NEO1 trial, a recent randomized, double-blind trial tested its efficacy and safety compared to alglucosidase (COMET trial) [167,168,169]. Participants in the COMET trial received intravenous infusions of alglucosidase alfa or avalglucosidase alfa (20 mg/kg) every 2 weeks [169]. The primary objective for efficacy, non-inferiority in respiratory function (as measured by upright FVC% predicted), was met and far exceeded the predefined margin [169]. Although the primary objective was met and the safety profile seemed more favorable than that of alglucosidase alfa, superiority testing did not reach statistical significance [169]. Avalglucosidase was approved for treatment of LOPD in 2021 and the recommended dose is 20 mg/kg body weight, administered once every 2 weeks.
A weight-tiered mg/kg dosing regimen of avalglucosidase alfa in IOPD patients (40 mg/kg every 2 weeks <30 kg) has been defined, supported by data from a model-informed extrapolation approach [170]. In IOPD, a 6-month primary analysis of the mini-COMET study recently showed a trend toward improvement or stabilization of clinical outcomes and a good safety profile, with better results for patients treated with avalglucosidase alfa 40 mg/kg every 2 weeks [171]. The Baby-COMET trial to assess efficacy in naïve IOPD patients is still recruiting.
In order to optimize ERT efficacy, proper exercise training and nutrition should be also considered [172,173,174]. A small cohort of LOPD patients showed improvement in laboratory tests (e.g., creatine kinase, lactate dehydrogenase) and exercise tolerance after introduction of personalized sessions of training and/or a high-protein diet [173]. A randomized, controlled study evaluating lifestyle interventions in IOPD and childhood-onset LOPD (median age: 10 years old) showed improvement in muscle strength, core stability, muscle function, quality of life, and fatigue after a tailored 12-week program [174].

6.2. Pharmacological Chaperone Therapy

Pharmacological chaperone therapy (PCT), first experimented in treatment of Fabry disease, is based on the concept that small-molecule ligands may act by blocking conformational fluctuations of a partially misfolded protein, thereby facilitating its trafficking into organelles [175,176]. In 2007, the action of the chaperone deoxynojirimycin (DNJ) and its derivative, N-butyldeoxynojirimycin (NB-DNJ), on the fibroblasts of Pompe patients showed a significant increase in GAA activity in fibroblasts carrying particular mutations [177,178]. However, the responsiveness to PCT alone seemed to be influenced by the location and nature of the amino acid substitutions, as an in vitro study showed that DNJ had no effect on 17 variants among the 76 tested mutant forms of GAA [176].
While facing bioavailability and pharmacodynamics limits of rhGAA, one strategy to improve these aspects was to use enzyme stabilizers to modulate GAA enzymatic activity. In fact, co-incubation of Pompe fibroblasts with recombinant human α-glucosidase and NB-DNJ revealed increased activity of GAA, which was later confirmed in animal model studies [179,180]. The results of the first clinical trial with combination therapy were published in 2014, comparing rhGAA ERT plus NB-DNJ chaperone (miglustat) to ERT alone in 13 patients with PD (3 infantile-onset and 10 late-onset) [181]. After a 2-month assessment of ERT alone, patients were treated with combination therapy for 12 months, then switched again to ERT alone and reevaluated after 2 months [181]. Increased and prolonged GAA activity with combination therapy compared to ERT alone was assessed by DBS, yet no clinical outcomes were analyzed because of the limitations of the study protocol [181].
Later, an open-label trial with different doses of chaperone (from 50 to 600 mg) confirmed this evidence, showing a 1.2- to 2.8-fold increase in total GAA activity after administration of PCT in 25 patients previously treated with ERT alone [182]. Recently, a phase 3 trial combining cipaglucosidase alfa, a new rhGAA with enhanced glycosylation for improved cellular uptake and processing, plus miglustat (n = 85) compared to alglucosidase alfa plus placebo (PROPEL study) was published in 2022 [183]. Although the primary endpoint (changing 6MWT) did not reach statistical superiority over alglucosidase, a meaningful improvement in motor and respiratory functions at 52 weeks was observed with cipaglucosidase alfa plus miglustat compared to alglucosidase alfa plus placebo [183]. These promising results led to the approval on 27 March 2023 of cipaglucosidase alfa as a long-term ERT used in combination with miglustat for the treatment of adults with LOPD in the EU [184].
To date, the advantages of combination therapy include possible blood–brain barrier crossing and better bioavailability of rhGAA, yet further data from the open-label phase of the PROPEL trial are needed to better define therapy response and clinical outcomes.

6.3. Gene Therapy

The risk of immunogenicity complications and the pharmacodynamics problems related to ERT have stimulated the development of different therapeutic approaches. This is the reason why gene therapy, predominantly adeno-associated viral (AAV) gene therapy for in vivo applications and lentiviral (LV) gene therapy for ex vivo applications, has gained much interest in the last few years.

6.3.1. AAV Vector-Mediated Gene Therapy in Pompe Disease

AAV vectors contain an icosahedral capsid that is serotype- and tissue-specific, depending on which receptor or coreceptor is used to enter tissue cells [185,186]. Clearly, the first experimental studies focused on serotypes endowed with muscle tropism, such as AAV9, AAV6, and AAV1 [187,188]. In an animal model, injection of theAAV2/6 vector in the gastrocnemius led to an increase in GAA activity in the injected muscle, even though no significant cross-correction was observed in the contralateral gastrocnemius, liver, or heart [187]. Limited results were also reported after intramuscular injection of the AAV9 vector at different stages of disease in a murine model, which showed that, despite high levels of AAV9-GAA tissue transduction and clearance of glycogen from myofibers, advanced-stage mice showed no improvement in motor function [188]. As respiratory dysfunction is one of the most severe comorbidities in PD, local intradiaphragmatic AAV delivery was piloted in preclinical studies and in a phase I/II AAV trial conducted with five ventilator-dependent children [189]. Although mild improvement in ventilatory endurance was observed, no significant change in maximal inspiratory pressure was identified and all patients developed variable Ig antibody responses to the AAV capsid proteins [189]. As similar results were later reported with four other patients, researchers lost interest in intramuscular AAV administration due to its poor rate of correction and they looked for a possible systemic route of administration [190,191]. In a murine model, intravenous AAV vector encoding the human GAA protein under control of a muscle-restricted promoter/enhancer element (AT845) led to functional improvement and glycogen clearance in skeletal and cardiac muscles, with accumulation of active enzyme in the muscle tissue at supraphysiological levels [191]. However, the safety outcome showed dose-dependent multi-organ toxicity in cynomolgus macaques, probably related to a xenogeneic immune response [191].
To improve CNS glycogen clearance, preclinical and animal studies have explored spinal, intrathecal, or intracerebroventricular delivery of AAV-GAA [192,193,194]. Intracerebroventricular injection of AAV-GAA transduced in the cortex and cerebellum decreased astrogliosis and improved myelination in a Pompe mouse model, yet a poor muscle glycogen storage response was observed [194].
A more recent study with a murine model applied intravenous administration of an AAV vector containing a regulatory cassette to drive GAA expression (AAV-PHP.B- chicken b-actin [CB]-hGAA) in the heart, skeletal muscles, and CNS [195]. Although reduced efficacy in copy numbers compared to CNS tissues was observed, therapeutic levels of GAA activity were achieved in muscle tissues with no adverse immune effects [195]. Taking inspiration from studies aimed at treating hemophilia B, liver-AAV vectors have been evaluated to correct GAA activity across the entire body, with potential benefits also in inducing antigen-specific immunological tolerance [196,197,198]. Administration of an adenoviral vector containing the CMV enhancer/promoter to drive GAA liver expression showed increased hepatic production of the human GAA precursor enzyme with consequently higher levels of GAA activity in plasma [185]. This approach was based on the idea that saturation of the receptor-mediated lysosomal targeting mechanism by over-expression of GAA in the liver would lead to secretion of the enzyme precursor into the circulatory system and increase its distribution to other tissues [185]. Liver-AAV vector treatment was associated with great improvement in glycogen clearance in the heart and variable responses in muscles, suggesting the presence of a threshold of circulating GAA activity required to achieve glycogen clearance in different tissues [198,199].
In a novel GAA knock-out model with a severe respiratory phenotype, systemic early administration of the AAV-sec GAA vector using a liver muscle promoter not only cleared pathological glycogen storage in the spinal cord but also respiratory impairment, which was at least partially influenced by neuroinflammation [200]. To further increase secretion of GAA, the use of a chimeric enzyme containing an alternative signal peptide was also applied, with mildly positive results [201].
Combined gene and enzyme replacement therapy has been studied in a murine model, based on the immunomodulatory effects of liver-AAV gene therapy [202]. After a first phase of administration of ERT or the AAV vector to compare their efficacy in glycogen clearance showed similar results, groups were treated with the vector alone or combined with the ERT to define a minimum dose of the AAV vector needed to induce immunotolerance [202]. However, further studies are required to evaluate the long-term effects and determine if a satisfactory response could be induced in long-treated ERT patients. Unfortunately, immunogenicity is still a problem for AAV vector therapies [203,204,205]. Although they suggest possible immunotolerance, the results of preclinical studies of AAV-liver gene therapy are variable and influenced by serotype selection, vector composition, dosing, and choice of immune modulation [198]. Furthermore, a large quantity of AAV vector would be required to treat Pompe patients because of hepatocyte-restricted expression, possibly exposing them to risk of immune responses against the transgene product and/or AAV capsid.
A recent study tried to assess the minimum effective dose of the administered AAV8 vector, which was set in preclinical studies at around 2 × 1011 vector genomes (vg)/kg body weight [186]. Although a good liver response in GAA activity was seen at this dose, a 4-fold higher dose was required to significantly achieve GAA activity in the heart, diaphragm, and quadriceps [186]. It has also been noted that GAA biochemical correction in animal models differs according to the age of the treated animals, as shown by comparing the effects of gene therapy in mice with PD aged 2 weeks or 2 months, raising the need of evaluate dose adjustment [206]. Biochemical correction was far superior in older mice with decreased left ventricle mass, decreased breathing frequency, and increased wire hang latency [206]. The latter two improvements were greater in females than in males despite a lower trend of correction of GAA activity, suggesting unknown sex-related differences in response to therapy [206]. In this regard, a recent study evaluating gene therapy response in a female mouse model suggested improvement of GAA activity after administration of androgen hormones [207]. However, this evidence is limited to preclinical studies as improvements seemed to be poor compared to the systemic adverse effects of treating females with androgen hormone therapy.

6.3.2. Lentiviral-Mediated Gene Therapy in Pompe Disease

Ex vivo studies have applied hematopoietic stem cell (HSC) gene therapy. It consists of isolating CD34+ cells from mobilized peripheral blood and transducing them with a lentiviral vector containing the functional GAA gene [208]. Then, the modified cells are infused back into the patient who was previously conditioned with alkylating agents, such as busulfan, in order to achieve repopulation of the bone marrow with stem cells capable of secreting functional GAA enzyme [208]. The greatest advantages of HSC gene therapy are the possibility of single administration and the immune tolerance induction that has already been demonstrated after HSC gene transfer of other exogenous proteins. The first trial evaluating this approach was published in 2009 [209]. A partial restoration of GAA enzymatic activity was observed in bone marrow and peripheral blood cells, with less efficacy in reducing glycogen storage in the heart [209].
By using an LV vector having a stable mammalian promoter and a human beta-globin locus control region, PD mice showed great improvement in hypertrophic cardiomyopathy and an increase in motor function, although not statistically significant compared to untreated mice 6 months post-transplant [210]. LV gene therapy in murine models also seems to be effective in CNS involvement. Transplantation of HSCs expressing codon-optimized GAA reduced glycogen to near normal levels in brain tissue [211]. In this study, immunofluorescence staining of mouse brain sections 3.5 months post-transplantation showed GAA expression in a large proportion of microglia and virtually all astrocytes, probably because of their expression of M6P receptors [211].

6.4. Other Experimental Therapies

Additional experimental therapies have targeted oxidative stress, splicing dysfunction, and substrate reduction.
As oxidative stress is linked to dysfunctional autophagy in PD pathogenesis, a pre-clinical study recently evaluated the association between ERT and antioxidant agents, suggesting that they may improve GAA activity in rhGAA-treated cells [212]. The use of splice-switching antisense oligonucleotides (AOs) has also been investigated. Most AO studies have focused on the common c.-32-13T>G mutation known to cause skipping of exon 2 from GAA transcripts [213]. This exon contains the translation start codon, and its skipping promotes mRNA degradation [213]. In this case, the function of AOs may be to modulate splicing by blocking splicing regulatory sequences. The feasibility of AO-mediated exon correction was explored by Van der Wal et al. in Pompe patient-derived myotubes and fibroblasts, and an increase in GAA enzyme activity was observed above the 20% threshold [214]. A recent study showed dose-dependent improvement in GAA activity in myogenic differentiation 1-induced myogenic cells derived from nine patients with LOPD, although the responses were influenced by cell quality and the nature of the second mutation [215]. Evaluating response in clinical trials is necessary as genomic sequence homology in the exon 2 region of GAA/Gaa between humans and mice is only around 50% [215]. A substrate reduction-based therapy inhibiting glycogen synthesis could be used as an adjunctive strategy to ERT. Antisense oligonucleotide-induced exon 6 skipping of the glycogen synthase 1 (Gys1) gene, which encodes skeletal muscle glycogen synthase activity, has shown great reduction in skeletal enzyme levels in an animal model [216]. More recently, the development of small-molecule inhibitors of human skeletal muscle glycogen synthase, such as substituted pyrazoles, has led to a phase 1 clinical trial in healthy individuals (NCT05249621) [217].

7. Conclusions

The diagnosis of PD has considerably improved over the years thanks to increased awareness among clinicians, the spread of newborn screening and clinical use of DBS, and the application of novel diagnostic tools, such as NGS and muscle MRI. An interesting point of discussion is the best outcome measures that future clinical trials for PD should evaluate. In most studies, FVC and 6MWT are set as the primary outcomes, even though they have some limitations. The evaluation of novel biomarkers is needed to assess the progression of the disease and guide clinicians in the timing of treatment. ERT represents the first and only treatment approved for PD. Although the advent of ERT has changed the natural history of the disease and the quality of life of patients, many unresolved issues remain, and limitations of treatment include the variable response and reduced duration of efficacy, which tends to decrease after the first few years of treatment. Next-generation ERT could improve this trend and have longer lasting efficacy than the current recombinant enzyme. Since the timing of treatment may influence the response to therapy, ERT should be started early when clinical, pathological, or MRI signs of disease appear. The wide accessibility of newborn screening for PD around the world also raises the question of when to start treatment in those subjects who are found to carry LOPD-associated variants and may develop clinical signs of disease much later in life. This will be a pressing problem in the near future and clinicians will have to confront each other to address it unambiguously.
More recently, the application of gene therapy has been explored. Even though gene therapy has the advantage of single administration of treatment, more studies are required to assess its efficacy in glycogen clearance in different tissues and clinical response. A critical discussion of all of these issues is crucial when looking forward to the future prospects of more effective treatments that would further change the natural history of the disease.

Author Contributions

Conceptualization, B.L. and M.F; methodology, S.C.P., B.R., F.C., S.D. and E.B.; writing—original draft preparation, B.L., L.P., S.C.P., B.R., F.C., S.D. and E.B.; writing—review and editing, M.F. and A.P.; supervision, M.F., L.P. and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors are members of a healthcare provider as part of the European Reference Network for Neuromuscular Diseases (ERN Euro-NMD).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Meena, N.K.; Raben, N. Pompe Disease: New Developments in an Old Lysosomal Storage Disorder. Biomolecules 2020, 10, 1339. [Google Scholar] [CrossRef] [PubMed]
  2. Reuser, A.J.J.; Kroos, M.A.; Hermans, M.M.P.; Bijvoet, A.G.A.; Verbeet, M.P.; Van Diggelen, O.P.; Kleijer, W.J.; Van Der Ploeg, A.T. Glycogenosis type II (acid maltase deficiency). Muscle Nerve 1995, 18, S61–S69. [Google Scholar] [CrossRef] [PubMed]
  3. Poorthuis, B.; Wevers, R.; Kleijer, W.; Groener, J.; de Jong, J.; van Weely, S.; Niezen-Koning, K.; van Diggelen, O. The frequency of lysosomal storage diseases in The Netherlands. Hum. Genet. 1999, 105, 151. [Google Scholar] [CrossRef]
  4. Martiniuk, F.; Chen, A.; Mack, A.; Arvanitopoulos, E.; Chen, Y.; Rom, W.N.; Codd, W.J.; Hanna, B.; Alcabes, P.; Raben, N.; et al. Carrier frequency for glycogen storage disease type II in New York and estimates of affected individuals born with the disease. Am. J. Med. Genet. 1998, 79, 69–72. [Google Scholar] [CrossRef]
  5. Ausems, M.; Verbiest, J.; Hermans, M.M.P.; Kroos, M.A.; Beemer, F.A.; Wokke, J.H.; Sandkuijl, L.A.; Reuser, A.J.J.; Van der Ploeg, A.T. Frequency of glycogen storage disease type II in the Netherlands: Implications for diagnosis and genetic counselling. Eur. J. Hum. Genet. 1999, 7, 713–716. [Google Scholar] [CrossRef]
  6. Pinto, R.R.; Caseiro, C.; Lemos, M.; Lopes, L.; Fontes, A.; Ribeiro, H.; Pinto, E.; Silva, E.; Rocha, S.; Marcão, A.; et al. Prevalence of lysosomal storage diseases in Portugal. Eur. J. Hum. Genet. 2003, 12, 87–92. [Google Scholar] [CrossRef] [PubMed]
  7. Sawada, T.; Kido, J.; Sugawara, K.; Momosaki, K.; Yoshida, S.; Kojima-Ishii, K.; Inoue, T.; Matsumoto, S.; Endo, F.; Ohga, S.; et al. Current status of newborn screening for Pompe disease in Japan. Orphanet J. Rare Dis. 2021, 16, 516. [Google Scholar] [CrossRef]
  8. Ficicioglu, C.; Ahrens-Nicklas, R.C.; Barch, J.; Cuddapah, S.R.; DiBoscio, B.S.; DiPerna, J.C.; Gordon, P.L.; Henderson, N.; Menello, C.; Luongo, N.; et al. Newborn screening for Pompe Disease: Pennsylvania experience. Int. J. Neonatal Screen. 2020, 6, 89. [Google Scholar] [CrossRef]
  9. Tang, H.; Feuchtbaum, L.; Sciortino, S.; Matteson, J.; Mathur, D.; Bishop, T.; Olney, R.S. The First Year Experience of Newborn Screening for Pompe Disease in California. Int. J. Neonatal Screen. 2020, 6, 9. [Google Scholar] [CrossRef]
  10. Klug, T.L.; Swartz, L.B.; Washburn, J.; Brannen, C.; Kiesling, J.L. Lessons Learned from Pompe Disease Newborn Screening and Follow-up. Int. J. Neonatal Screen. 2020, 6, 11. [Google Scholar] [CrossRef]
  11. Gragnaniello, V.; Pijnappel, P.W.W.M.; Burlina, A.P.; In ‘t Groen, S.L.M.; Gueraldi, D.; Cazzorla, C.; Maines, E.; Polo, G.; Salviati, L.; Di Salvo, G.; et al. Newborn screening for Pompe Disease in Italy: Long-term results and future challenges. Mol. Genet. Metab. Rep. 2022, 33, 100929. [Google Scholar] [CrossRef]
  12. Wittmann, J.; Karg, E.; Turi, S.; Legnini, E.; Wittmann, G.; Giese, A.-K.; Lukas, J.; Gölnitz, U.; Klingenhäger, M.; Bodamer, O.; et al. Newborn Screening for Lysosomal Storage Disorders in Hungary. JIMD Rep. Case Res. Rep. 2012, 6, 117–125. [Google Scholar] [CrossRef]
  13. Chiang, S.C.; Hwu, W.; Lee, N.; Hsu, L.W.; Chien, Y.H. Algorithm for Pompe Disease newborn screening: Results from the Taiwan screening program. Mol. Genet. Metab. 2012, 106, 281–286. [Google Scholar] [CrossRef] [PubMed]
  14. Burton, B.K.; Charrow, J.; Hoganson, G.E.; Fleischer, J.; Grange, D.K.; Braddock, S.R.; Hitchins, L.; Hickey, R.; Christensen, K.M.; Groepper, D.; et al. Newborn Screening for Pompe Disease in Illinois: Experience with 684,290 Infants. Int. J. Neonatal Screen. 2020, 6, 4. [Google Scholar] [CrossRef] [PubMed]
  15. Mechtler, T.P.; Stary, S.; Metz, T.F.; De Jesús, V.R.; Greber-Platzer, S.; Pollak, A.; Herkner, K.R.; Streubel, B.; Kasper, D.C. Neonatal screening for lysosomal storage disorders: Feasibility and incidence from a nationwide study in Austria. Lancet 2012, 379, 335–341. [Google Scholar] [CrossRef] [PubMed]
  16. Hall, P.L.; Sanchez, R.; Hagar, A.F.; Jerris, S.C.; Wittenauer, A.; Wilcox, W.R. Two-Tiered Newborn Screening with Post-Analytical Tools for Pompe Disease and Mucopolysaccharidosis Type I Results in Performance Improvement and Future Direction. Int. J. Neonatal Screen. 2020, 6, 2. [Google Scholar] [CrossRef]
  17. Navarrete-Martínez, J.I.; Limón-Rojas, A.E.; Gaytán-García, M.d.J.; Reyna-Figueroa, J.; Wakida-Kusunoki, G.; Delgado-Calvillo, M.d.R.; Cantú-Reyna, C.; Cruz-Camino, H.; Cervantes-Barragán, D.E. Newborn screening for six lysosomal storage disorders in a cohort of Mexican patients: Three-year findings from a screening program in a closed Mexican health system. Mol. Genet. Metab. 2017, 121, 16–21. [Google Scholar] [CrossRef]
  18. Momosaki, K.; Kido, J.; Yoshida, S.; Sugawara, K.; Miyamoto, T.; Inoue, T.; Okumiya, T.; Matsumoto, S.; Endo, F.; Hirose, S.; et al. Newborn screening for Pompe disease in Japan: Report and literature review of mutations in the GAA gene in Japanese and Asian patients. J. Hum. Genet. 2019, 64, 741–755. [Google Scholar] [CrossRef]
  19. Kohler, L.; Puertollano, R.; Raben, N. Pompe Disease: From Basic Science to Therapy. Neurotherapeutics 2018, 15, 928–942. [Google Scholar] [CrossRef]
  20. Peruzzo, P.; Pavan, E.; Dardis, A. Molecular genetics of Pompe disease: A comprehensive overview. Ann. Transl. Med. 2019, 7, 278. [Google Scholar] [CrossRef]
  21. Wisselaar, H.A.; Kroos, M.A.; Hermans, M.P.; Van Beeumens, J.; Reusers, A.J. Structural and functional changes of lysosomal acid alpha-glucosidase during intracellular transport and maturation. J. Biol. Chem. 1993, 268, 2223–2231. [Google Scholar] [CrossRef] [PubMed]
  22. Roig-zamboni, V.; Cobucci-Ponzano, B.; Iacono, R.; Ferrara, M.C.; Germany, S.; Bourne, Y.; Parenti, G.; Moracci, M.; Sulzenbacher, G. Structure of human lysosomal acid α-glucosidase– a guide for the treatment of Pompe Disease. Nat. Commun. 2017, 8, 1111. [Google Scholar] [CrossRef]
  23. Park, H.; Kim, J.; Lee, Y.K.; Kim, W.; You, S.K.; Do, J.; Jang, Y.; Oh, D.B.; Kim, H.H. Four unreported types of glycans containing mannose-6-phosphate are heterogeneously attached at three sites (including newly found Asn 233) to recombinant human acid alpha-glucosidase that is the only approved treatment for Pompe Disease. Biochem. Biophys. Res. Commun. 2018, 495, 2418–2424. [Google Scholar] [CrossRef] [PubMed]
  24. Hermans, M.M.; Wisselaar, H.A.; Kroos, M.A.; Oostra, B.A.; Reuser, A.J. Human lysosomal alpha-glucosidase: Functional characterization of the glycosylation sites. Biochem. J. 1993, 289, 681–686. [Google Scholar] [CrossRef] [PubMed]
  25. Tong, P.Y.; Gregory, W.; Kornfelds, S. Ligand interactions of the cation-independent mannose 6-phosphate receptor. The stoichiometry of mannose 6-phosphate binding. J. Biol. Chem. 1989, 264, 7962–7969. [Google Scholar] [CrossRef]
  26. Kaplan, A.; Achord, D.T.; Sly, W.S. Phosphohexosyl components of a lysosomal enzyme are recognized by pinocytosis receptors on human fibroblasts. Proc. Natl. Acad. Sci. USA 1977, 74, 2026–2030. [Google Scholar] [CrossRef]
  27. Griffiths, G.; Hoflack, B.; Simons, K.; Mellman, I.; Kornfeld, S. The mannose 6-phosphate receptor and the biogenesis of lysosomes. Cell 1988, 52, 329–341. [Google Scholar] [CrossRef]
  28. Raben, N.; Roberts, A.; Plotz, P.H. Role of autophagy in the pathogenesis of Pompe disease. Acta Myol. Myopathies cardiomyopathies Off. J. Mediterr. Soc. Myol. 2007, 26, 45–48. [Google Scholar]
  29. Moreland, R.J.; Jin, X.; Zhang, X.K.; Decker, R.W.; Albee, K.L.; Lee, K.L.; Cauthron, R.D.; Brewer, K.; Edmunds, T.; Canfield, W.M. Lysosomal Acid α-Glucosidase Consists of Four Different Peptides Processed from a Single Chain Precursor. J. Biol. Chem. 2005, 280, 6780–6791. [Google Scholar] [CrossRef]
  30. Selvan, N.; Mehta, N.; Venkateswaran, S.; Brignol, N.; Graziano, M.; Sheikh, M.O.; McAnany, Y.; Hung, F.; Madrid, M.; Krampetz, R.; et al. Endolysosomal N-glycan processing is critical to attain the most active form of the enzyme acid alpha-glucosidase. J. Biol. Chem. 2021, 296, 100769. [Google Scholar] [CrossRef]
  31. Raben, N.; Wong, A.; Ralston, E.; Myerowitz, R. Autophagy and mitochondria in Pompe disease: Nothing is so new as what has long been forgotten. Am. J. Med. Genet. Part C Semin. Med. Genet. 2012, 160, 13–21. [Google Scholar] [CrossRef] [PubMed]
  32. Slonim, A.E.; Bulone, L.; Ritz, S.; Goldberg, T.; Chen, A.; Martiniuk, F. Identification of two subtypes of infantile acid maltase deficiency. J. Pediatr. 2000, 137, 283–285. [Google Scholar] [CrossRef] [PubMed]
  33. Kishnani, P.S.; Hwu, W.-L.; Mandel, H.; Nicolino, M.; Yong, F.; Corzo, D. Infantile-Onset Pompe Disease Natural History Study Group. A retrospective, multinational, multicenter study on the natural history of infantile-onset Pompe disease. J. Pediatr. 2006, 148, 671–676.e2. [Google Scholar] [CrossRef]
  34. Angelini, C.; Engel, A.G. Comparative study of acid maltase deficiency: Biochemical differences between infantile, childhood, and adult types. Arch. Neurol. 1972, 26, 344–349. [Google Scholar] [CrossRef] [PubMed]
  35. Holzwarth, J.; Minopoli, N.; Pfrimmer, C.; Smitka, M.; Borrel, S.; Kirschner, J.; Muschol, N.; Hartmann, H.; Hennermann, J.B.; Neubauer, B.A.; et al. Clinical and Genetic Aspects of Juvenile Onset Pompe Disease. Neuropediatrics 2021, 53, 39–45. [Google Scholar] [CrossRef] [PubMed]
  36. Semplicini, C.; Letard, P.; De Antonio, M.; Taouagh, N.; Perniconi, B.; Bouhour, F.; Echaniz-Laguna, A.; Orlikowski, D.; Sacconi, S.; Salort-Campana, E.; et al. Late-onset Pompe disease in France: Molecular features and epidemiology from a nationwide study. J. Inherit. Metab. Dis. 2018, 41, 937–946. [Google Scholar] [CrossRef] [PubMed]
  37. Toscano, A.; Rodolico, C.; Musumeci, O. Multisystem late onset Pompe disease (LOPD): An update on clinical aspects. Ann. Transl. Med. 2019, 7, 284. [Google Scholar] [CrossRef]
  38. Filosto, M.; Cotelli, M.; Vielmi, V.; Todeschini, A.; Rinaldi, F.; Rota, S.; Scarpelli, M.; Padovani, A. Late-onset Glycogen Storage Disease type 2. Curr. Mol. Med. 2014, 14, 971–978. [Google Scholar] [CrossRef]
  39. Wokke, J.H.J.; Escolar, D.M.; Pestronk, A.; Jaffe, K.M.; Carter, G.T.; Van Den Berg, L.H.; Florence, J.M.; Mayhew, J.; Skrinar, A.; Corzo, D.; et al. Clinical features of late-onset Pompe disease: A prospective cohort study. Muscle Nerve 2008, 38, 1236–1245. [Google Scholar] [CrossRef]
  40. Niño, M.Y.; Wijgerde, M.; De Faria, D.O.S.; Hoogeveen-Westerveld, M.; Bergsma, A.J.; Broeders, M.; Van der Beek, N.A.M.E.; Van den Hout, H.J.M.; Van der Ploeg, A.T.; Verheijen, F.W.; et al. Enzymatic diagnosis of Pompe Disease: Lessons from 28 years of experience. Eur. J. Hum. Genet. 2021, 29, 434–446. [Google Scholar] [CrossRef]
  41. Viamonte, M.A.; Filipp, S.L.; Zaidi, Z.; Gurka, M.J.; Byrne, B.J.; Kang, P.B. Phenotypic implications of pathogenic variant types in Pompe disease. J. Hum. Genet. 2021, 66, 1089–1099. [Google Scholar] [CrossRef] [PubMed]
  42. Reuser, A.J.J.; Ploeg, A.T.; Chien, Y.; Llerena, J.; Abbott, M.; Clemens, P.R.; Kimonis, V.E.; Leslie, N.; Maruti, S.S.; Sanson, B.; et al. GAA variants and phenotypes among 1079 patients with Pompe disease: Data from the Pompe Registry. Hum. Mutat. 2019, 40, 2146–2164. [Google Scholar] [CrossRef] [PubMed]
  43. Sampaolo, S.; Esposito, T.; Farina, O.; Formicola, D.; Diodato, D.; Gianfrancesco, F.; Cipullo, F.; Cremone, G.; Cirillo, M.; Del Viscovo, L.; et al. Distinct disease phenotypes linked to different combinations of GAA mutations in a large late-onset GSDII sibship. Orphanet J. Rare Dis. 2013, 8, 159. [Google Scholar] [CrossRef] [PubMed]
  44. Angelini, C.; Bembi, B.; Burlina, A.; Filosto, M.; Maioli, M.A.; Morandi, L.O.; Parini, R.; Pegoraro, E.; Ravaglia, S.; Servidei, S.; et al. Changing Characteristics of Late-Onset Pompe Disease Patients in Italy: Data from the Pompe Registry. J. Neuromuscul. Dis. 2015, 2, S36–S37. [Google Scholar] [CrossRef]
  45. Kroos, M.A.; Van der Kraan, M.; Van Diggelen, O.P.; Kleijer, W.J.; Reuser, A.J.; Boogaard, M.J.V.D.; Ausems, M.G.; van Amstel, H.K.P.; Poenaru, L.; Nicolino, M. Glycogen storage disease type II: Frequency of three common mutant alleles and their associated clinical phenotypes studied in 121 patients. J. Med. Genet. 1995, 32, 836–837. [Google Scholar] [CrossRef] [PubMed]
  46. Herbert, M.; Case, L.E.; Rairikar, M.; Cope, H.; Bailey, L.; Austin, S.L.; Kishnani, P.S. Early-onset of symptoms and clinical course of Pompe disease associated with the c.-32–13 T > G variant. Mol. Genet. Metab. 2018, 126, 106–116. [Google Scholar] [CrossRef] [PubMed]
  47. Bergsma, A.J.; In ‘t Groen, S.L.M.; Van den Dorpel, J.J.A.; Van den Hout, H.J.M.P.; Van der Beek, N.A.M.E.; Schoser, B.; Toscano, A.; Musumeci, O.; Bembi, B.; Dardis, A.; et al. A genetic modifier of symptom onset in Pompe Disease. eBioMedicine 2019, 43, 553–561. [Google Scholar] [CrossRef]
  48. Wens, S.; Van Gelder, C.; Kruijshaar, M.; De Vries, J.; Van der Beek, N.; Reuser, A.; Van Doorn, P.; Van der Ploeg, A.; Brusse, E.P. 17.1 Phenotypic variation within 22 families with Pompe disease. Neuromuscul. Disord. 2013, 23, 826. [Google Scholar] [CrossRef]
  49. De Filippi, P.; Saeidi, K.; Ravaglia, S.; Dardis, A.; Angelini, C.; Mongini, T.; Morandi, L.; Moggio, M.; Di Muzio, A.; Filosto, M.; et al. Genotype-phenotype correlation in Pompe disease, a step forward. Orphanet J. Rare Dis. 2014, 9, 102. [Google Scholar] [CrossRef]
  50. Kroos, M.A.; Pomponio, R.J.; Hagemans, M.L.; Keulemans, J.; Phipps, M.; DeRiso, M.; Palmer, R.E.; Ausems, M.G.; Van der Beek, N.A.; Van Diggelen, O.P.; et al. Broad spectrum of Pompe disease in patients with the same c.-32-13T->G haplotype. Neurology 2007, 68, 110–115. [Google Scholar] [CrossRef]
  51. Kuperus, E.; Van der Meijden, J.C.; In ‘t Groen, S.L.M.; Kroos, M.A.; Hoogeveen-Westerveld, M.; Rizopoulos, D.; Martinez, M.Y.N.; Kruijshaar, M.E.; Van Doorn, P.A.; Van der Beek, N.A.M.E.; et al. The ACE I/D polymorphism does not explain heterogeneity of natural course and response to enzyme replacement therapy in Pompe Disease. PLoS ONE. 2018, 13, e0208854. [Google Scholar] [CrossRef] [PubMed]
  52. Lukacs, Z.; Cobos, P.N.; Wenninger, S.; Willis, T.A.; Guglieri, M.; Roberts, M.; Quinlivan, R.; Hilton-Jones, D.; Evangelista, T.; Zierz, S.; et al. Prevalence of Pompe disease in 3076 patients with hyperCKemia and limb-girdle muscular weakness. Neurology 2016, 87, 295–298. [Google Scholar] [CrossRef] [PubMed]
  53. Alejaldre, A.; Diaz-Manera, J.; Ravaglia, S.; Tibaldi, E.C.; D’Amore, F.; Moris, G.; Muelas, N.; Vilchez, J.J.; García-Medina, A.; Usón, M.; et al. Trunk muscle involvement in late-onset Pompe disease: Study of thirty patients. Neuromuscul. Disord. 2012, 22 (Suppl. 2), S148–S154. [Google Scholar] [CrossRef] [PubMed]
  54. Jones, H.N.; Hobson-Webb, L.D.; Kuchibhatla, M.; Crisp, K.D.; Whyte-Rayson, A.; Batten, M.T.; Zwelling, P.J.; Kishnani, P.S. Tongue weakness and atrophy differentiates late-onset Pompe disease from other forms of acquired/hereditary myopathy. Mol. Genet. Metab. 2021, 133, 261–268. [Google Scholar] [CrossRef] [PubMed]
  55. Dubrovsky, A.; Corderi, J.; Lin, M.; Kishnani, P.S.; Jones, H.N. Expanding the phenotype of late-onset pompe disease: Tongue weakness: A new clinical observation. Muscle Nerve 2011, 44, 897–901. [Google Scholar] [CrossRef]
  56. Al-Hashel, J.; Ismail, I. Late-Onset Pompe Disease Presenting with Isolated Tongue Involvement. Case Rep. Neurol. 2022, 14, 98–103. [Google Scholar] [CrossRef]
  57. Karam, C.; Dimitrova, D.; Yutan, E.; Chahin, N. Bright tongue sign in patients with late-onset Pompe disease. J. Neurol. 2019, 266, 2518–2523. [Google Scholar] [CrossRef]
  58. Kishnani, P.S.; Steiner, R.D.; Bali, D.; Berger, K.; Byrne, B.J.; Case, L.E.; Crowley, J.F.; Downs, S.; Howell, R.R.; Kravitz, R.M.; et al. Pompe disease diagnosis and management guideline. Anesthesia Analg. 2006, 8, 267–288. [Google Scholar] [CrossRef]
  59. Wagner, M.; Chaouch, A.; Müller, J.S.; Polvikoski, T.; Willis, T.A.; Sarkozy, A.; Eagle, M.; Bushby, K.; Straub, V.; Lochmüller, H. Presymptomatic late-onset Pompe disease identified by the dried blood spot test. Neuromuscul. Disord. 2013, 23, 89–92. [Google Scholar] [CrossRef]
  60. Spada, M.; Porta, F.; Vercelli, L.; Pagliardini, V.; Chiadò-Piat, L.; Boffi, P.; Pagliardini, S.; Remiche, G.; Ronchi, D.; Comi, G.; et al. Screening for later-onset Pompe’s disease in patients with paucisymptomatic hyperCKemia. Mol. Genet. Metab. 2013, 109, 171–173. [Google Scholar] [CrossRef]
  61. Musumeci, O.; La Marca, G.; Spada, M.; Mondello, S.; Danesino, C.; Comi, G.P.; Pegoraro, E.; Antonini, G.; Marrosu, G.; Liguori, R.; et al. LOPED study: Looking for an early diagnosis in a late-onset Pompe Disease high-risk population. J. Neurol. Neurosurg. Psychiatry. 2016, 87, 5–11. [Google Scholar] [CrossRef] [PubMed]
  62. Gutiérrez-Rivas, E.; Bautista, J.; Vílchez, J.; Muelas, N.; Díaz-Manera, J.; Illa, I.; Martínez-Arroyo, A.; Olivé, M.; Sanz, I.; Arpa, J.; et al. Targeted screening for the detection of Pompe disease in patients with unclassified limb-girdle muscular dystrophy or asymptomatic hyperCKemia using dried blood: A Spanish cohort. Neuromuscul. Disord. 2015, 25, 548–553. [Google Scholar] [CrossRef] [PubMed]
  63. Wens, S.C.; Schaaf, G.J.; Michels, M.; Kruijshaar, M.E.; van Gestel, T.J.; Groen, S.I.; Pijnenburg, J.; Dekkers, D.H.; Demmers, J.A.; Verdijk, L.B.; et al. Elevated Plasma Cardiac Troponin T Levels Caused by Skeletal Muscle Damage in Pompe Disease. Circ. Cardiovasc. Genet. 2016, 9, 6–13. [Google Scholar] [CrossRef] [PubMed]
  64. Hoeksma, M.; Boon, M.; Niezen-Koning, K.E.; Van Overbeek-van Gils, L.; Van Spronsen, F.J. Isolated elevated serum transaminases leading to the diagnosis of asymptomatic Pompe Disease. Eur. J. Pediatr. 2007, 166, 871–874. [Google Scholar] [CrossRef]
  65. Müller-Felber, W.; Horvath, R.; Gempel, K.; Podskarbi, T.; Shin, Y.; Pongratz, D.; Walter, M.C.; Baethmann, M.; Schlotter-Weigel, B.; Lochmüller, H.; et al. Late onset Pompe disease: Clinical and neurophysiological spectrum of 38 patients including long-term follow-up in 18 patients. Neuromuscul. Disord. 2007, 17, 698–706. [Google Scholar] [CrossRef]
  66. Kassardjian, C.D.; Engel, A.G.; Sorenson, E.J. Electromyographic findings in 37 patients with adult-onset acid maltase deficiency. Muscle Nerve 2015, 51, 759–761. [Google Scholar] [CrossRef]
  67. Pongratz, D.; Kötzner, H.; Hübner, G.; Deufel, T.; Wieland, O.H. Adult form of acid maltase deficiency presenting as progressive spinal muscular atrophy. Deut Med. Wochenschr. 1984, 109, 537–541. [Google Scholar] [CrossRef]
  68. Khan, A.A.; Boggs, T.; Bowling, M.; Austin, S.; Stefanescu, M.; Case, L.; Kishnani, P.S. Whole-body magnetic resonance imaging in late-onset Pompe disease: Clinical utility and correlation with functional measures. J. Inherit. Metab. Dis. 2019, 43, 549–557. [Google Scholar] [CrossRef]
  69. Figueroa-Bonaparte, S.; Segovia, S.; Llauger, J.; Belmonte, I.; Pedrosa, I.; Alejaldre, A.; Mayos, M.; Suárez-Cuartín, G.; Gallardo, E.; Illa, I.; et al. Muscle MRI Findings in Childhood/Adult Onset Pompe Disease Correlate with Muscle Function. PLoS ONE 2016, 11, e0163493. [Google Scholar] [CrossRef]
  70. Alonso-Jiménez, A.; Nuñez-Peralta, C.; Montesinos, P.; Alonso-Pérez, J.; García, C.; Montiel, E.; Belmonte, I.; Pedrosa, I.; Segovia, S.; Llauger, J.; et al. Different Approaches to Analyze Muscle Fat Replacement With Dixon MRI in Pompe Disease. Front. Neurol. 2021, 12, 675781. [Google Scholar] [CrossRef]
  71. Carlier, R.-Y.; Laforet, P.; Wary, C.; Mompoint, D.; Laloui, K.; Pellegrini, N.; Annane, D.; Carlier, P.G.; Orlikowski, D. Whole-body muscle MRI in 20 patients suffering from late onset Pompe disease: Involvement patterns. Neuromuscul. Disord. 2011, 21, 791–799. [Google Scholar] [CrossRef]
  72. Mercuri, E.; Counsell, S.; Allsop, J.; Jungbluth, H.; Kinali, M.; Bonne, G.; Schwartz, K.; Bydder, G.; Dubowitz, V.; Muntoni, F. Selective Muscle Involvement on Magnetic Resonance Imaging in Autosomal Dominant Emery-Dreifuss Muscular Dystrophy. Neuropediatrics 2002, 33, 10–14. [Google Scholar] [CrossRef]
  73. Jordan, B.; Eger, K.; Koesling, S.; Zierz, S. Camptocormia phenotype of FSHD: A clinical and MRI study on six patients. J. Neurol. 2010, 258, 866–873. [Google Scholar] [CrossRef]
  74. Mercuri, E.; Clements, E.; Offiah, A.; Pichiecchio, A.; Vasco, G.; Bianco, F.; Berardinelli, A.; Manzur, A.; Pane, M.; Messina, S.; et al. Muscle magnetic resonance imaging involvement in muscular dystrophies with rigidity of the spine. Ann. Neurol. 2010, 67, 201–208. [Google Scholar] [CrossRef]
  75. Pichiecchio, A.; Rossi, M.; Cinnante, C.; Colafati, G.S.; Icco, R.; Parini, R.; Menni, F.; Furlan, F.; Burlina, A.; Sacchini, M.; et al. Muscle MRI of classic infantile pompe patients: Fatty substitution and edema-like changes. Muscle Nerve 2017, 55, 841–848. [Google Scholar] [CrossRef] [PubMed]
  76. Vaeggemose, M.; Mencagli, R.A.; Hansen, J.S.; Dräger, B.; Ringgaard, S.; Vissing, J.; Andersen, H. Function, structure and quality of striated muscles in the lower extremities in patients with late onset Pompe Disease—An MRI study. PeerJ 2021, 9, e10928. [Google Scholar] [CrossRef] [PubMed]
  77. Harlaar, L.; Ciet, P.; van Tulder, G.; Pittaro, A.; van Kooten, H.A.; van der Beek, N.A.M.E.; Brusse, E.; Wielopolski, P.A.; de Bruijne, M.; van der Ploeg, A.T.; et al. Chest MRI to diagnose early diaphragmatic weakness in Pompe disease. Orphanet J. Rare Dis. 2021, 16, 21. [Google Scholar] [CrossRef]
  78. Wens, S.C.A.; Ciet, P.; Perez-Rovira, A.; Logie, K.; Salamon, E.; Wielopolski, P.; De Bruijne, M.; Kruijshaar, M.E.; Tiddens, H.A.W.M.; Van Doorn, P.A.; et al. Lung MRI and impairment of diaphragmatic function in Pompe Disease. BMC Pulm. Med. 2015, 15, 54. [Google Scholar] [CrossRef] [PubMed]
  79. Harlaar, L.; Ciet, P.; Van Tulder, G.; Brusse, E.; Timmermans, R.G.M.; Janssen, W.G.M.; De Bruijine, M.; van der Ploeg, A.T.; Tiddens, H.A.W.M.; Van Doorn, P.A.; et al. Diaphragmatic dysfunction in neuromuscular disease, an MRI study. Neuromuscul. Disord. 2022, 32, 15–24. [Google Scholar] [CrossRef] [PubMed]
  80. Montagnese, F.; Barca, E.; Musumeci, O.; Mondello, S.; Migliorato, A.; Ciranni, A.; Rodolico, C.; De Filippi, P.; Danesino, C.; Toscano, A. Clinical and molecular aspects of 30 patients with late-onset Pompe disease (LOPD): Unusual features and response to treatment. J. Neurol. 2015, 262, 968–978. [Google Scholar] [CrossRef] [PubMed]
  81. Werneck, L.C.; Lorenzoni, P.J.; Kay, C.S.K.; Scola, R.H. Muscle biopsy in Pompe disease. Arq. de Neuro-Psiquiatria 2013, 71, 284–289. [Google Scholar] [CrossRef]
  82. Schoser, B.G.H.; Müller-Höcker, J.; Horvath, R.; Gempel, K.; Pongratz, D.; Lochmüller, H.; Müller-Felber, W. Adult-onset glycogen storage disease type 2: Clinico-pathological phenotype revisited. Neuropathol. Appl. Neurobiol. 2007, 33, 544–559. [Google Scholar] [CrossRef] [PubMed]
  83. Laforet, P.; Nicolino, M.; Eymard, B.; Puech, J.P.; Caillaud, C.; Poenaru, L.; Fardeau, M. Juvenile and adult-onset acid maltase deficiency in France: Genotype-phenotype correlation. Neurology 2000, 55, 1122–1128. [Google Scholar] [CrossRef] [PubMed]
  84. Lynch, C.M.; Johnson, J.; Vaccaro, C.; Thurberg, B.L. High-resolution Light Microscopy (HRLM) and Digital Analysis of Pompe Disease Pathology. J. Histochem. Cytochem. 2005, 53, 63–73. [Google Scholar] [CrossRef] [PubMed]
  85. Terracciano, C.; Rastelli, E.; Massa, R. Periodic acid-Schiff staining on resin muscle sections: Improvement in the histological diagnosis of late-onset Pompe disease. Muscle Nerve 2011, 45, 611–612. [Google Scholar] [CrossRef]
  86. Feeney, E.J.; Austin, S.; Chien, Y.-H.; Mandel, H.; Schoser, B.; Prater, S.; Hwu, W.-L.; Ralston, E.; Kishnani, P.S.; Raben, N. The value of muscle biopsies in Pompe disease: Identifying lipofuscin inclusions in juvenile- and adult-onset patients. Acta Neuropathol. Commun. 2014, 2, 2. [Google Scholar] [CrossRef]
  87. Taverna, S.; Cammarata, G.; Colomba, P.; Sciarrino, S.; Zizzo, C.; Francofonte, D.; Zora, M.; Scalia, S.; Brando, C.; Curto, A.L.o.; et al. Pompe Disease: Pathogenesis, molecular genetics and diagnosis. Aging 2020, 12, 15856–15874. [Google Scholar] [CrossRef]
  88. Savarese, M.; Torella, A.; Musumeci, O.; Angelini, C.; Astrea, G.; Bello, L.; Bruno, C.; Comi, G.P.; Di Fruscio, G.; Piluso, G.; et al. Targeted gene panel screening is an effective tool to identify undiagnosed late onset Pompe disease. Neuromuscul. Disord. 2018, 28, 586–591. [Google Scholar] [CrossRef]
  89. Whitaker, C.H.; Felice, K.J.; Natowicz, M. Biopsy-proven alpha-glucosidase deficiency with normal lymphocyte enzyme activity. Muscle Nerve 2004, 29, 440–442. [Google Scholar] [CrossRef]
  90. Okumiya, T.; Keulemans, J.L.; Kroos, M.A.; Van der Beek, N.M.; Boer, M.A.; Takeuchi, H.; Van Diggelen, O.P.; Reuser, A.J. A new diagnostic assay for glycogen storage disease type II in mixed leukocytes. Mol. Genet. Metab. 2006, 88, 22–28. [Google Scholar] [CrossRef]
  91. Van der Ploeg, A.T.; Reuser, A.J. Pompe’s disease. Lancet 2008, 372, 1342–1353. [Google Scholar] [CrossRef]
  92. Lukacs, Z.; Oliva, P.; Nieves Cobos, P.; Scott, J.; Mechtler, T.P.; Kasper, D.C. At-Risk Testing for Pompe Disease using dried bloodspots: Lessons learned for newborn screening. Int. J. Neonatal Screen. 2020, 6, 96. [Google Scholar] [CrossRef]
  93. Vissing, J.; Lukacs, Z.; Straub, V. Diagnosis of Pompe Disease: Muscle biopsy vs blood-based assays. JAMA Neurol. 2013, 70, 923–927. [Google Scholar] [CrossRef] [PubMed]
  94. Kishnani, P.S.; Amartino, H.M.; Lindberg, C.; Miller, T.M.; Wilson, A.; Keutzer, J. Methods of diagnosis of patients with Pompe disease: Data from the Pompe Registry. Mol. Genet. Metab. 2014, 113, 84–91. [Google Scholar] [CrossRef] [PubMed]
  95. Goldstein, J.L.; Young, S.P.; Changela, M.; Dickerson, G.H.; Zhang, H.; Dai, J.; Peterson, D.; Millington, D.S.; Kishnani, P.S.; Bali, D.S. Screening for pompe disease using a rapid dried blood spot method: Experience of a clinical diagnostic laboratory. Muscle Nerve 2009, 40, 32–36. [Google Scholar] [CrossRef] [PubMed]
  96. Winchester, B.; Bali, D.; Bodamer, O.; Caillaud, C.; Christensen, E.; Cooper, A.; Cupler, E.; Deschauer, M.; Fumić, K.; Jackson, M.; et al. Methods for a prompt and reliable laboratory diagnosis of Pompe disease: Report from an international consensus meeting. Mol. Genet. Metab. 2008, 93, 275–281. [Google Scholar] [CrossRef] [PubMed]
  97. Kallwass, H.; Carr, C.; Gerrein, J.; Titlow, M.; Pomponio, R.; Bali, D.; Dai, J.; Kishnani, P.; Skrinar, A.; Corzo, D.; et al. Rapid diagnosis of late-onset Pompe disease by fluorometric assay of α-glucosidase activities in dried blood spots. Mol. Genet. Metab. 2007, 90, 449–452. [Google Scholar] [CrossRef]
  98. Elbin, C.S.; Olivova, P.; Marashio, C.A.; Cooper, S.K.; Cullen, E.; Keutzer, J.M.; Zhang, X.K. The effect of preparation, storage and shipping of dried blood spots on the activity of five lysosomal enzymes. Clin. Chim. Acta 2011, 412, 1207–1212. [Google Scholar] [CrossRef]
  99. Thuriot, F.; Gravel, E.; Hodson, K.; Ganopolsky, J.; Rakic, B.; Waters, P.J.; Gravel, S.; Lévesque, S. Molecular Diagnosis of Pompe Disease in the Genomic Era: Correlation with Acid Alpha-Glucosidase Activity in Dried Blood Spots. J. Clin. Med. 2021, 10, 3868. [Google Scholar] [CrossRef]
  100. Goldstein, J.L.; Dickerson, G.; Kishnani, P.S.; Rehder, C.; Bali, D.S. Blood-based diagnostic testing for Pompe disease: Consistency between GAA enzyme activity in dried blood spots and GAA gene sequencing results. Muscle Nerve 2013, 49, 775–776. [Google Scholar] [CrossRef]
  101. Parisi, D.; Musumeci, O.; Mondello, S.; Brizzi, T.; Oteri, R.; Migliorato, A.; Ciranni, A.; Mongini, T.E.; Rodolico, C.; Vita, G.; et al. Vacuolated PAS-Positive Lymphocytes on Blood Smear: An Easy Screening Tool and a Possible Biomarker for Monitoring Therapeutic Responses in Late Onset Pompe Disease (LOPD). Front. Neurol. 2018, 9, 880. [Google Scholar] [CrossRef]
  102. Saville, J.T.; Fuller, M. Experience with the Urinary Tetrasaccharide Metabolite for Pompe Disease in the Diagnostic Laboratory. Metabolites 2021, 11, 446. [Google Scholar] [CrossRef]
  103. Chien, Y.-H.; Goldstein, J.L.; Hwu, W.-L.; Smith, P.B.; Lee, N.-C.; Chiang, S.-C.; Tolun, A.A.; Zhang, H.; Vaisnins, A.E.; Millington, D.S.; et al. Baseline urinary glucose tetrasaccharide concentrations in patients with infantile- and late-onset Pompe Disease identified by newborn screening. JIMD Rep. 2015, 19, 67–73. [Google Scholar]
  104. Piraud, M.; Pettazzoni, M.; De Antonio, M.; Vianey-Saban, C.; Froissart, R.; Chabrol, B.; Young, S.; Laforêt, P.; French Pompe study group. Urine glucose tetrasaccharide: S good biomarker for glycogenoses type II and III? A study of the French cohort. Mol. Genet. Metab. Rep. 2020, 23, 100583. [Google Scholar] [CrossRef] [PubMed]
  105. Young, S.P.; Piraud, M.; Goldstein, J.L.; Zhang, H.; Rehder, C.; Laforet, P.; Kishnani, P.S.; Millington, D.S.; Bashir, M.R.; Bali, D.S. Assessing disease severity in Pompe disease: The roles of a urinary glucose tetrasaccharide biomarker and imaging techniques. Am. J. Med. Genet. Part C Semin. Med. Genet. 2012, 160, 50–58. [Google Scholar] [CrossRef] [PubMed]
  106. Boentert, M.; Prigent, H.; Várdi, K.; Jones, H.N.; Mellies, U.; Simonds, A.K.; Wenninger, S.; Cortés, E.B.; Confalonieri, M. Practical Recommendations for Diagnosis and Management of Respiratory Muscle Weakness in Late-Onset Pompe Disease. Int. J. Mol. Sci. 2016, 17, 1735. [Google Scholar] [CrossRef] [PubMed]
  107. Boentert, M.; Karabul, N.; Wenninger, S.; Stubbe-Dräger, B.; Mengel, E.; Schoser, B.; Young, P. Sleep-related symptoms and sleep-disordered breathing in adult Pompe disease. Eur. J. Neurol. 2014, 22, 369.e27. [Google Scholar] [CrossRef]
  108. Gaeta, M.; Barca, E.; Ruggeri, P.; Minutoli, F.; Rodolico, C.; Mazziotti, S.; Milardi, D.; Musumeci, O.; Toscano, A. Late-onset Pompe disease (LOPD): Correlations between respiratory muscles CT and MRI features and pulmonary function. Mol. Genet. Metab. 2013, 110, 290–296. [Google Scholar] [CrossRef]
  109. Berger, K.I.; Chan, Y.; Rom, W.N.; Beno, W.; Oppenheimer, B.W.; Goldring, R.M. Progression from respiratory dysfunction to failure in late-onset Pompe Disease. Neuromuscul. Disord. 2016, 26, 481–489. [Google Scholar] [CrossRef]
  110. Fromageot, C.; Lofaso, F.; Annane, D.; Falaize, L.; Lejaille, M.; Clair, B.; Gajdos, P.; Raphaël, J.C. Supine fall in lung volumes in the assessment of diaphragmatic weakness in neuromuscular disorders. Arch. Phys. Med. Rehabil. 2001, 82, 123–128. [Google Scholar] [CrossRef]
  111. Van der Beek, N.A.; Van Capelle, C.I.; Van der Velden-van Etten, K.I.; Hop, W.C.; Van den Berg, B.; Reuser, A.J.; Van Doorn, P.A.; Van der Ploeg, A.T.; Stam, H. Rate of progression and predictive factors for pulmonary outcome in children and adults with Pompe Disease. Mol. Genet. Metab. 2011, 104, 129–136. [Google Scholar] [CrossRef] [PubMed]
  112. American Thoracic Society / European Respiratory Society ATS/ERS Statement on respiratory muscle testing. Am. J. Respir. Crit. Care Med. 2002, 166, 518–624. [CrossRef] [PubMed]
  113. Boentert, M.; Florian, A.; Dräger, B.; Young, P.; Yilmaz, A. Pattern and prognostic value of cardiac involvement in patients with late-onset pompe disease: A comprehensive cardiovascular magnetic resonance approach. J. Cardiovasc. Magn. Reson. 2016, 18, 91. [Google Scholar] [CrossRef] [PubMed]
  114. Seifert, B.L.; Snyder, M.S.; Klein, A.A.; O’Loughlin, J.E.; Magid, M.S.; Engle, M.A. Development of obstruction to ventricular outflow and impairment of inflow in glycogen storage disease of the heart: Serial echocardiographic studies from birth to death at 6 months. Am. Heart J. 1992, 123, 239–242. [Google Scholar] [CrossRef]
  115. van den Hout, H.M.; Hop, W.; van Diggelen, O.P.; Smeitink, J.A.M.; Smit, G.P.A.; Poll-The, B.-T.T.; Bakker, H.D.; Loonen, M.C.B.; De Klerk, J.B.C.; Reuser, A.J.J.; 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. [Google Scholar] [CrossRef]
  116. Morris, D.A.; Blaschke, D.; Krebs, A.; Canaan-Kühl, S.; Plöckinger, U.; Knobloch, G.; Walter, T.C.; Kühnle, Y.; Boldt, L.-H.; Kraigher-Krainer, E.; et al. Structural and functional cardiac analyses using modern and sensitive myocardial techniques in adult Pompe disease. Int. J. Cardiovasc. Imaging 2015, 31, 947–956. [Google Scholar] [CrossRef]
  117. Fayssoil, A.; Nardi, O.; Annane, D.; Orlikowski, D. Right ventricular function in late-onset Pompe disease. J. Clin. Monit. Comput. 2014, 28, 419–421. [Google Scholar] [CrossRef]
  118. Soliman, O.I.I.; Van der Beek, N.A.; Van Doorn, P.A.; Vletter, W.B.; Nemes, A.; Van Dalen, B.M.; Ten Cate, F.J.; Van der Ploeg, A.T.; Geleijnse, M.L. Cardiac involvement in adults with Pompe Disease. J. Intern. Med. 2008, 264, 333–339. [Google Scholar] [CrossRef]
  119. Forsha, D.; Li, J.S.; Smith, P.B.; Van der Ploeg, A.T.; Kishnani, P.; Pasquali, K.S. Late-Onset Treatment Study Investigators. Cardiovascular abnormalities in late onset Pompe Disease and response to enzyme replacement therapy. Genet. Med. 2011, 13, 625–631. [Google Scholar] [CrossRef]
  120. Herbert, M.; Cope, H.; Li, J.S.; Kishnani, P.S. Severe cardiac involvement is rare in patients with late-onset Pompe Disease and the common c.-32-13T>G Variant: Implications for newborn screening. J. Pediatr. 2018, 198, 308–312. [Google Scholar] [CrossRef]
  121. Bulkley, B.H.; Hutchins, G.M. Pompe’s disease presenting as hypertrophic myocardiopathy with Wolff-Parkinson-White syndrome. Am. Heart J. 1978, 96, 246–252. [Google Scholar] [CrossRef]
  122. Francesconi, M.; Auff, E. Cardiac arrhythmias and the adult form of type II glycogenosis. N. Engl. J. Med. 1982, 306, 937–938. [Google Scholar] [PubMed]
  123. Van Kooten, H.A.; Roelen, C.H.A.; Brusse, E.; Van der Beek, N.A.M.E.; Michels, M.; Van der Ploeg, A.T.; Wagenmakers, M.A.E.M.; Van Doorn, P.A. Cardiovascular disease in non-classic Pompe Disease: A systematic review. Neuromuscul. Disord. 2021, 31, 79–90. [Google Scholar] [CrossRef] [PubMed]
  124. Angelini, C.; Semplicini, C.; Ravaglia, S.; Moggio, M.; Comi, G.P.; Musumeci, O.; Pegoraro, E.; Tonin, P.; Filosto, M.; Servidei, S.; et al. New motor outcome function measures in evaluation of Late-Onset Pompe disease before and after enzyme replacement therapy. Muscle Nerve 2012, 45, 831–834. [Google Scholar] [CrossRef] [PubMed]
  125. Montagnese, F.; Granata, F.; Musumeci, O.; Rodolico, C.; Mondello, S.; Barca, E.; Cucinotta, M.; Ciranni, A.; Longo, M.; Toscano, A. Intracranial arterial abnormalities in patients with late onset Pompe disease (LOPD). J. Inherit. Metab. Dis. 2016, 39, 391–398. [Google Scholar] [CrossRef]
  126. Filosto, M.; Todeschini, A.; Cotelli, M.S.; Vielmi, V.; Rinaldi, F.; Rota, S.; Scarpelli, M.; Padovani, A. Non-muscle involvement in late-onset Glycogenosis II. Acta Myol. 2013, 32, 91–94. [Google Scholar]
  127. Korlimarla, A.; Lim, J.; Kishnani, P.S.; Sun, B. An emerging phenotype of central nervous system involvement in Pompe Disease: From bench to bedside and beyond. Ann. Transl. Med. 2019, 7, 289. [Google Scholar] [CrossRef]
  128. Garibaldi, M.; Sacconi, S.; Antonini, G.; Desnuelle, C. Long term follow-up of cerebrovascular abnormalities in late onset Pompe disease (LOPD). J. Neurol. 2017, 264, 589–590. [Google Scholar] [CrossRef]
  129. Mormina, E.; Musumeci, O.; Tessitore, A.; Ciranni, A.; Tavilla, G.; Pitrone, A.; Vinci, S.L.; Caragliano, A.A.; Longo, M.; Granata, F.; et al. Intracranial aneurysm management in patients with late-onset Pompe disease (LOPD). Neurol. Sci. 2020, 42, 2411–2419. [Google Scholar] [CrossRef]
  130. Musumeci, O.; Marino, S.; Granata, F.; Morabito, R.; Bonanno, L.; Brizzi, T.; Lo Buono, V.; Corallo, F.; Longo, M.; Toscano, A. Central nervous system involvement in late onset Pompe Disease (LOPD): Clues from neuroimaging and neuropsychological analysis. Eur. J. Neurol. 2019, 26, 442.e35. [Google Scholar] [CrossRef]
  131. Ebbink, B.J.; Poelman, E.; Aarsen, F.K.; Plug, I.; Régal, L.; Muentjes, C.; Beek, N.A.M.E.v.d.; Lequin, M.H.; Ploeg, A.T.; Hout, J.M.P.v.D. Classic infantile Pompe patients approaching adulthood: A cohort study on consequences for the brain. Dev. Med. Child. Neurol. 2018, 60, 579–586. [Google Scholar] [CrossRef]
  132. Korlimarla, A.; Spiridigliozzi, G.A.; Crisp, K.; Herbert, M.; Chen, S.; Malinzak, M.; Stefanescu, M.; Austin, S.L.; Cope, H.; Zimmerman, K.; et al. Novel approaches to quantify CNS involvement in children with Pompe disease. Neurology 2020, 95, e718–e732. [Google Scholar] [CrossRef]
  133. Borroni, B.; Cotelli, M.S.; Premi, E.; Gazzina, S.; Cosseddu, M.; Formenti, A.; Gasparotti, R.; Filosto, M.; Padovani, A. The brain in late-onset glycogenosis II: A structural and functional MRI study. J. Inherit. Metab. Dis. 2013, 36, 989–995. [Google Scholar] [CrossRef]
  134. Bertoldo, F.; Zappini, F.; Brigo, M.; Moggio, M.; Lucchini, V.; Angelini, C.; Semplicini, C.; Filosto, M.; Ravaglia, S.; Cotelli, S.; et al. Prevalence of asymptomatic vertebral fractures in late-onset Pompe Disease. J. Clin. Endocrinol. Metab. 2015, 100, 401–406. [Google Scholar] [CrossRef] [PubMed]
  135. An, Y.; Young, S.P.; Kishnani, P.S.; Millington, D.S.; Amal, A.; Corzo, D.; Chen, Y. Glucose tetrasaccharide as a biomarker for monitoring the therapeutic response to enzyme replacement therapy for Pompe Disease. Mol. Genet. Metab. 2005, 85, 247–254. [Google Scholar] [CrossRef] [PubMed]
  136. Carrasco-Rozas, A.; Fernández-Simón, E.; Lleixà, M.C.; Belmonte, I.; Pedrosa-Hernandez, I.; Montiel-Morillo, E.; Nuñez-Peralta, C.; Rossello, J.L.; Segovia, S.; De Luna, N.; et al. Identification of serum microRNAs as potential biomarkers in Pompe disease. Ann. Clin. Transl. Neurol. 2019, 6, 1214–1224. [Google Scholar] [CrossRef] [PubMed]
  137. Tarallo, A.; Carissimo, A.; Gatto, F.; Nusco, E.; Toscano, A.; Musumeci, O.; Coletta, M.; Karali, M.; Acampora, E.; Damiano, C.; et al. microRNAs as biomarkers in Pompe disease. Genet Med. 2019, 21, 591–600. [Google Scholar] [CrossRef] [PubMed]
  138. Lollert, A.; Stihl, C.; Hötker, A.M.; Mengel, E.; König, J.; Laudemann, K.; Gökce, S.; Düber, C.; Staatz, G. Quantification of intramuscular fat in patients with late-onset Pompe disease by conventional magnetic resonance imaging for the long-term follow-up of enzyme replacement therapy. PLoS ONE 2018, 13, e0190784. [Google Scholar] [CrossRef] [PubMed]
  139. Nuñez-Peralta, C.; Alonso-Pérez, J.; Llauger, J.; Segovia, S.; Montesinos, P.; Belmonte, I.; Pedrosa, I.; Montiel, E.; Alonso-Jiménez, A.; Sánchez-González, J.; et al. Follow-up of late-onset Pompe disease patients with muscle magnetic resonance imaging reveals increase in fat replacement in skeletal muscles. J. Cachexia Sarcopenia Muscle 2020, 11, 1032–1046. [Google Scholar] [CrossRef] [PubMed]
  140. Figueroa-Bonaparte, S.; Llauger, J.; Segovia, S.; Belmonte, I.; Pedrosa, I.; Montiel, E.; Montesinos, P.; Sánchez-González, J.; Alonso-Jiménez, A.; Gallardo, E.; et al. Quantitative muscle MRI to follow up late onset Pompe patients: A prospective study. Sci. Rep. 2018, 8, 10898. [Google Scholar] [CrossRef]
  141. Gruhn, K.M.; Heyer, C.M.; Güttsches, A.-K.; Rehmann, R.; Nicolas, V.; Schmidt-Wilcke, T.; Tegenthoff, M.; Vorgerd, M.; Kley, R.A. Muscle imaging data in late-onset Pompe disease reveal a correlation between the pre-existing degree of lipomatous muscle alterations and the efficacy of long-term enzyme replacement therapy. Mol. Genet. Metab. Rep. 2015, 3, 58–64. [Google Scholar] [CrossRef]
  142. Ruggeri, P.; Monaco, L.L.; Musumeci, O.; Tavilla, G.; Gaeta, M.; Caramori, G.; Toscano, A. Ultrasound assessment of diaphragm function in patients with late-onset Pompe disease. Neurol. Sci. 2020, 41, 2175–2184. [Google Scholar] [CrossRef] [PubMed]
  143. Hagemans, M.; Winkel, L.P.; Hop, W.C.; Reuser, A.J.; Van Doorn, P.A.; Van der Ploeg, A.T. Disease severity in children and adults with Pompe disease related to age and disease duration. Neurology 2005, 64, 2139–2141. [Google Scholar] [CrossRef] [PubMed]
  144. Angelini, C.; Nascimbeni, A.C.; Semplicini, C. Therapeutic advances in the management of Pompe disease and other metabolic myopathies. Ther. Adv. Neurol. Disord. 2013, 6, 311–321. [Google Scholar] [CrossRef] [PubMed]
  145. Kishnani, P.S.; Goldenberg, P.C.; DeArmey, S.L.; Heller, J.; Benjamin, D.; Young, S.; Bali, D.; Smith, S.A.; Li, J.S.; Mandel, H.; et al. Cross-reactive immunologic material status affects treatment outcomes in Pompe disease infants. Mol. Genet. Metab. 2010, 99, 26–33. [Google Scholar] [CrossRef] [PubMed]
  146. Doerfler, P.A.; Nayak, S.; Corti, M.; Morel, L.; Herzog, R.W.; Byrne, B.J. Targeted approaches to induce immune tolerance for Pompe disease therapy. Mol. Ther. Methods Clin. Dev. 2016, 3, 15053. [Google Scholar] [CrossRef]
  147. Poelman, E.; Hoogeveen-westerveld, M.; Kroos-de Haan, M.A.; Van den Hout, J.M.P.; Bronsema, K.J.; Van de Merbel, N.C.; Van der Ploeg, A.T.; Pijnappel, W.W.M.P. High sustained antibody titers in patients with classic infantile Pompe Disease following immunomodulation at start of rnzyme replacement therapy. J. Pediatr. 2018, 195, 236–243.e3. [Google Scholar] [CrossRef]
  148. van der Ploeg, A.T.; Clemens, P.R.; Corzo, D.; Escolar, D.M.; Florence, J.; Groeneveld, G.J.; Herson, S.; Kishnani, P.S.; Laforet, P.; Lake, S.L.; et al. A Randomized Study of Alglucosidase Alfa in Late-Onset Pompe’s Disease. N. Engl. J. Med. 2010, 362, 1396–1406. [Google Scholar] [CrossRef]
  149. Regnery, C.; Kornblum, C.; Hanisch, F.; Vielhaber, S.; Strigl-Pill, N.; Grunert, B.; Müller-Felber, W.; Glocker, F.X.; Spranger, M.; Deschauer, M.; et al. 36 months observational clinical study of 38 adult Pompe disease patients under alglucosidase alfa enzyme replacement therapy. J. Inherit. Metab. Dis. 2012, 35, 837–845. [Google Scholar] [CrossRef]
  150. Furusawa, Y.; Mori-Yoshimura, M.; Yamamoto, T.; Sakamoto, C.; Wakita, M.; Kobayashi, Y.; Fukumoto, Y.; Oya, Y.; Fukuda, T.; Sugie, H.; et al. Effects of enzyme replacement therapy on five patients with advanced late-onset glycogen storage disease type II: A 2-year follow-up study. J. Inherit. Metab. Dis. 2011, 35, 301–310. [Google Scholar] [CrossRef]
  151. Anderson, L.J.; Henley, W.; Wyatt, K.M.; Nikolaou, V.; Waldek, S.; Hughes, D.A.; Lachmann, R.H.; Logan, S. Effectiveness of enzyme replacement therapy in adults with late-onset Pompe disease: Results from the NCS-LSD cohort study. J. Inherit. Metab. Dis. 2014, 37, 945–952. [Google Scholar] [CrossRef] [PubMed]
  152. De Vries, J.M.; Van der Beek, N.A.; Hop, W.C.; Karstens, F.P.; Wokke, J.H.; De Visser, M.; Van Engelen, B.G.; Kuks, J.B.; Van der Kooi, A.J.; Notermans, N.C.; et al. Effect of enzyme therapy and prognostic factors in 69 adults with Pompe Disease: An open-label single-center study. Orphanet J. Rare Dis. 2012, 7, 73. [Google Scholar] [CrossRef] [PubMed]
  153. Orlikowski, D.; Pellegrini, N.; Prigent, H.; Laforêt, P.; Carlier, R.; Carlier, P.; Eymard, B.; Lofaso, F.; Annane, D. Recombinant human acid alpha-glucosidase (rhGAA) in adult patients with severe respiratory failure due to Pompe disease. Neuromuscul. Disord. 2011, 21, 477–482. [Google Scholar] [CrossRef] [PubMed]
  154. van der Ploeg, A.; Carlier, P.G.; Carlier, R.-Y.; Kissel, J.T.; Schoser, B.; Wenninger, S.; Pestronk, A.; Barohn, R.J.; Dimachkie, M.M.; Goker-Alpan, O.; et al. Prospective exploratory muscle biopsy, imaging, and functional assessment in patients with late-onset Pompe disease treated with alglucosidase alfa: The EMBASSY Study. Mol. Genet. Metab. 2016, 119, 115–123. [Google Scholar] [CrossRef] [PubMed]
  155. Ripolone, M.; Violano, R.; Ronchi, D.; Mondello, S.; Nascimbeni, A.; Colombo, I.; Fagiolari, G.; Bordoni, A.; Fortunato, F.; Lucchini, V.; et al. Effects of short-to-long term enzyme replacement therapy (ERT) on skeletal muscle tissue in late onset Pompe disease (LOPD). Neuropathol. Appl. Neurobiol. 2017, 44, 449–462. [Google Scholar] [CrossRef]
  156. van der Ploeg, A.T.; Kruijshaar, M.E.; Toscano, A.; Laforêt, P.; Angelini, C.; Lachmann, R.H.; Pascual, S.I.P.; Roberts, M.; Rösler, K.; Stulnig, T.; et al. European consensus for starting and stopping enzyme replacement therapy in adult patients with Pompe disease: A 10-year experience. Eur. J. Neurol. 2017, 24, 768-e31. [Google Scholar] [CrossRef]
  157. Stepien, K.M.; Hendriksz, C.J.; Roberts, M.; Sharma, R. Observational clinical study of 22 adult-onset Pompe disease patients undergoing enzyme replacement therapy over 5years. Mol. Genet. Metab. 2016, 117, 413–418. [Google Scholar] [CrossRef]
  158. Angelini, C.; The Italian GSDII Group; Semplicini, C.; Ravaglia, S.; Bembi, B.; Servidei, S.; Pegoraro, E.; Moggio, M.; Filosto, M.; Sette, E.; et al. Observational clinical study in juvenile-adult glycogenosis type 2 patients undergoing enzyme replacement therapy for up to 4 years. J. Neurol. 2011, 259, 952–958. [Google Scholar] [CrossRef]
  159. Strothotte, S.; Strigl-Pill, N.; Grunert, B.; Kornblum, C.; Eger, K.; Wessig, C.; Deschauer, M.; Breunig, F.; Glocker, F.X.; Vielhaber, S.; et al. Enzyme replacement therapy with alglucosidase alfa in 44 patients with late-onset glycogen storage disease type 2: 12-month results of an observational clinical trial. J. Neurol. 2009, 257, 91–97. [Google Scholar] [CrossRef]
  160. van der Ploeg, A.T.; Barohn, R.; Carlson, L.; Charrow, J.; Clemens, P.R.; Hopkin, R.J.; Kishnani, P.S.; Laforêt, P.; Morgan, C.; Nations, S.; et al. Open-label extension study following the Late-Onset Treatment Study (LOTS) of alglucosidase alfa. Mol. Genet. Metab. 2012, 107, 456–461. [Google Scholar] [CrossRef]
  161. Hahn, S.H.; Kronn, D.; Leslie, N.D.; Pena, L.D.; Tanpaiboon, P.; Gambello, M.J.; Gibson, J.B.; Hillman, R.; Stockton, D.W.; Day, J.W.; et al. Efficacy, safety profile, and immunogenicity of alglucosidase alfa produced at the 4000-liter scale in US children and adolescents with Pompe disease: ADVANCE, a phase IV, open-label, prospective study. Anesthesia Analg. 2018, 20, 1284–1294. [Google Scholar] [CrossRef]
  162. Filosto, M.; Piccinelli, S.C.; Ravaglia, S.; Servidei, S.; Moggio, M.; Musumeci, O.; Donati, M.A.; Pegoraro, E.; Di Muzio, A.; Maggi, L.; et al. Assessing the Role of Anti rh-GAA in Modulating Response to ERT in a Late-Onset Pompe Disease Cohort from the Italian GSDII Study Group. Adv. Ther. 2019, 36, 1177–1189. [Google Scholar] [CrossRef] [PubMed]
  163. Kuperus, E.; Kruijshaar, M.E.; Wens, S.C.A.; De Vries, J.M.; Favejee, M.M.; Van der Meijden, J.C.; Brusse, E.; Van Doorn, P.A.; Van der Ploeg, A.T.; Van der Beek, N.A.M.E. Long-term benefit of enzyme replacement therapy in Pompe Disease: A 5-year prospective study. Neurology 2017, 89, 2365–2373. [Google Scholar] [CrossRef]
  164. Harlaar, L.; Hogrel, J.-Y.; Perniconi, B.; Kruijshaar, M.E.; Rizopoulos, D.; Taouagh, N.; Canal, A.; Brusse, E.; van Doorn, P.A.; van der Ploeg, A.T.; et al. Large variation in effects during 10 years of enzyme therapy in adults with Pompe disease. Neurology 2019, 93, e1756–e1767. [Google Scholar] [CrossRef] [PubMed]
  165. Sun, B.; Li, S.; Bird, A.; Yi, H.; Kemper, A.; Thurberg, B.L.; Koeberl, D.D. Antibody formation and mannose-6-phosphate receptor expression impact the efficacy of muscle-specific transgene expression in murine Pompe Disease. J. Gene Med. 2010, 12, 881–891. [Google Scholar] [CrossRef]
  166. Zhu, Y.; Jiang, J.-L.; Gumlaw, N.K.; Zhang, J.; Bercury, S.D.; Ziegler, R.J.; Lee, K.; Kudo, M.; Canfield, W.M.; Edmunds, T.; et al. Glycoengineered Acid α-Glucosidase With Improved Efficacy at Correcting the Metabolic Aberrations and Motor Function Deficits in a Mouse Model of Pompe Disease. Mol. Ther. 2009, 17, 954–963. [Google Scholar] [CrossRef] [PubMed]
  167. Pena, L.D.M.; Barohn, R.J.; Byrne, B.J.; Desnuelle, C.; Goker-Alpan, O.; Ladha, S.; Laforêt, P.; Mengel, K.E.; Pestronk, A.; Pouget, J.; et al. Safety, tolerability, pharmacokinetics, pharmacodynamics, and exploratory efficacy of the novel enzyme replacement therapy avalglucosidase alfa (neoGAA) in treatment-naïve and alglucosidase alfa-treated patients with late-onset Pompe Disease: A phase 1, open-label, multicenter, multinational, ascending dose study. Neuromuscul. Disord. 2019, 29, 167–186. [Google Scholar]
  168. Dimachkie, M.M.; Barohn, R.J.; Byrne, B.; Goker-Alpan, O.; Kishnani, P.S.; Ladha, S.; Laforêt, P.; Mengel, K.E.; Pena, L.D.; Sacconi, S.; et al. NEO1 and NEO-EXT studies: Long-term safety and exploratory efficacy of repeat avalglucosidase alfa dosing for 5.5 years in late-onset Pompe disease patients. Mol. Genet. Metab. 2020, 129, S49. [Google Scholar] [CrossRef]
  169. Diaz-Manera, J.; Kishnani, P.S.; Kushlaf, H.; Ladha, S.; Mozaffar, T.; Straub, V.; Toscano, A.; van der Ploeg, A.T.; Berger, K.I.; Clemens, P.R.; et al. Safety and efficacy of avalglucosidase alfa versus alglucosidase alfa in patients with late-onset Pompe disease (COMET): A phase 3, randomised, multicentre trial. Lancet Neurol. 2021, 20, 1012–1026. [Google Scholar] [CrossRef]
  170. Li, R.J.; Ma, L.; Drozda, K.; Wang, J.; Punnose, A.R.; Jeng, L.J.B.; Maynard, J.W.; Zhu, H.; Pacanowski, M. Model—Informed approach supporting approval of Nexviazyme (Avalglucosidase Alfa—Ngpt) in pediatric patients with Late-Onset Pompe Disease. AAPS J. 2023, 25, 16. [Google Scholar] [CrossRef]
  171. Kishnani, P.S.; Kronn, D.; Brassier, A.; Broomfield, A.; Davison, J.; Hahn, S.H.; Kumada, S.; Labarthe, F.; Ohki, H.; Pichard, S.; et al. Safety and efficacy of avalglucosidase alfa in individuals with infantile-onset Pompe disease enrolled in the phase 2, open-label Mini-COMET study: The 6-month primary analysis report. Genet Med. 2022, 25, 100328. [Google Scholar] [CrossRef]
  172. Angelini, C. Exercise, nutrition and enzyme replacement therapy are efficacious in adult Pompe patients: Report from EPOC Consortium. Eur. J. Transl. Myol. 2021, 31, 9798. [Google Scholar] [CrossRef] [PubMed]
  173. Sechi, A.; Zuccarelli, L.; Grassi, B.; Frangiamore, R.; De Amicis, R.; Marzorati, M.; Porcelli, S.; Tullio, A.; Bacco, A.; Bertoli, S.; et al. Exercise training alone or in combination with high-protein diet in patients with late onset Pompe disease: Results of a cross over study. Orphanet J. Rare Dis. 2020, 15, 143. [Google Scholar] [CrossRef] [PubMed]
  174. Scheffers, L.E.; Somers, O.C.; Dulfer, K.; Dieleman, G.C.; Walet, S.; Van der Giessen, L.J.; Van der Ploeg, A.T.; Van den Hout, J.M.P.; Van den Berg, L.E.; exercise team. Physical training and high-protein diet improved muscle strength, parent-reported fatigue, and physical quality of life in children with Pompe Disease. J. Inherit. Metab. Dis. 2023, 46, 605–617. [Google Scholar] [CrossRef] [PubMed]
  175. Borie-Guichot, M.; Tran, M.L.; Génisson, Y.; Ballereau, S.; Dehoux, C. Pharmacological Chaperone Therapy for Pompe Disease. Molecules 2021, 26, 7223. [Google Scholar] [CrossRef] [PubMed]
  176. Flanagan, J.J.; Rossi, B.; Tang, K.; Wu, X.; Mascioli, K.; Donaudy, F.; Tuzzi, M.R.; Fontana, F.; Cubellis, M.V.; Porto, C.; et al. The pharmacological chaperone 1-deoxynojirimycin increases the activity and lysosomal trafficking of multiple mutant forms of acid alpha-glucosidase. Hum. Mutat. 2009, 30, 1683–1692. [Google Scholar] [CrossRef] [PubMed]
  177. Parenti, G.; Zuppaldi, A.; Pittis, M.G.; Tuzzi, M.R.; Annunziata, I.; Meroni, G.; Porto, C.; Donaudy, F.; Rossi, B.; Rossi, M.; et al. Pharmacological Enhancement of Mutated α-Glucosidase Activity in Fibroblasts from Patients with Pompe Disease. Mol. Ther. 2007, 15, 508–514. [Google Scholar] [CrossRef]
  178. Okumiya, T.; Kroos, M.A.; Van Vliet, L.; Takeuchi, H.; Van der Ploeg, A.T.; Reuser, A.J. Chemical chaperones improve transport and enhance stability of mutant α-glucosidases in glycogen storage disease type II. Mol. Genet. Metab. 2007, 90, 49–57. [Google Scholar] [CrossRef]
  179. Porto, C.; Cardone, M.; Fontana, F.; Rossi, B.; Tuzzi, M.R.; Tarallo, A.; Barone, M.V.; Andria, G.; Parenti, G. The Pharmacological Chaperone N-butyldeoxynojirimycin Enhances Enzyme Replacement Therapy in Pompe Disease Fibroblasts. Mol. Ther. 2009, 17, 964–971. [Google Scholar] [CrossRef]
  180. Khanna, R.; Flanagan, J.J.; Feng, J.; Soska, R.; Frascella, M.; Pellegrino, L.J.; Lun, Y.; Guillen, D.; Lockhart, D.J.; Valenzano, K.J. The pharmacological chaperone AT2220 increases recombinant human acid alfa-glucosidase uptake and glycogen reduction in a mouse model of Pompe Disease. PLoS ONE. 2012, 7, e40776. [Google Scholar] [CrossRef]
  181. Parenti, G.; Fecarotta, S.; la Marca, G.; Rossi, B.; Ascione, S.; Donati, M.A.; Morandi, L.O.; Ravaglia, S.; Pichiecchio, A.; Ombrone, D.; et al. A Chaperone Enhances Blood α-Glucosidase Activity in Pompe Disease Patients Treated With Enzyme Replacement Therapy. Mol. Ther. 2014, 22, 2004–2012. [Google Scholar] [CrossRef] [PubMed]
  182. Kishnani, P.; Tarnopolsky, M.; Roberts, M.; Sivakumar, K.; Dasouki, M.; Dimachkie, M.M.; Finanger, E.; Goker-alpan, O.; Guter, K.A.; Mozaffar, T.; et al. Duvoglustat HCl increases systemic and tissue exposure of active acid alfa -glucosidase in Pompe patients co-administered with alglucosidase alfa. Mol. Ther. 2017, 25, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
  183. Schoser, B.; Roberts, M.; Byrne, B.J.; Sitaraman, S.; Jiang, H.; Laforêt, P.; Toscano, A.; Castelli, J.; Díaz-Manera, J.; Goldman, M.; et al. Safety and efficacy of cipaglucosidase alfa plus miglustat versus alglucosidase alfa plus placebo in late-onset Pompe disease (PROPEL): An international, randomised, double-blind, parallel-group, phase 3 trial. Lancet Neurol. 2021, 20, 1027–1037. [Google Scholar] [CrossRef]
  184. Amicus Therapeutics. Amicus Therapeutics Announces European Commission Approval for Pombiliti™ in Patients with Late-onset Pompe Disease [Media Release]. Available online: https://ir.amicusrx.com/news-releases/news-release-details/amicus-therapeutics-announces-european-commission-approval-1 (accessed on 27 March 2023).
  185. Amalfitano, A.; McVie-Wylie, A.J.; Hu, H.; Dawson, T.L.; Raben, N.; Plotz, P.; Chen, Y.T. Systemic correction of the muscle disorder glycogen storage disease type II after hepatic targeting of a modified adenovirus vector encoding human acid-α-glucosidase. Proc. Natl. Acad. Sci. USA 1999, 96, 8861–8866. [Google Scholar] [CrossRef]
  186. Han, S.O.; Gheorghiu, D.; Li, S.; Kang, H.R.; Koelberg, D. Minimum effective dose to achieve biochemical correction with adeno-associated virus vector-mediated gene therapy in mice with Pompe Disease. Hum. Gene Ther. 2022, 33, 492–498. [Google Scholar] [CrossRef] [PubMed]
  187. Sun, B.; Zhang, H.; Franco, L.M.; Brown, T.; Bird, A.; Schneider, A.; Koeberl, D.D. Correction of glycogen storage disease type II by an adeno-associated virus vector containing a muscle-specific promoter. Mol. Ther. 2005, 11, 889–898. [Google Scholar] [CrossRef] [PubMed]
  188. Todd, A.G.; McElroy, J.A.; Grange, R.W.; Fuller, D.D.; Walter, G.A.; Byrne, B.J.; Falk, D.J. Correcting Neuromuscular Deficits With Gene Therapy in Pompe Disease. Ann. Neurol. 2015, 78, 222–234. [Google Scholar] [CrossRef]
  189. Smith, B.K.; Collins, S.W.; Conlon, T.J.; Mah, C.S.; Lawson, L.A.; Martin, A.D.; Fuller, D.D.; Cleaver, B.D.; Cle, N.; Phillips, D.; et al. Phase I/II trial of adeno-associated virus–mediated alpha-glucosidase gene therapy to the diaphragm for chronic respiratory failure in Pompe disease: Initial safety and ventilatory outcomes. Hum. Gene Ther. 2013, 24, 630–640. [Google Scholar] [CrossRef]
  190. Corti, M.; Liberati, C.; Smith, B.K.; Lawson, L.A.; Tuna, I.S.; Conlon, T.J.; Coleman, K.E.; Islam, S.; Herzog, R.W.; Fuller, D.D.; et al. Safety of Intradiaphragmatic Delivery of adeno-associated gene therapy in children affected by Pompe Disease. Hum. Gene Ther. 2017, 28, 208–218. [Google Scholar]
  191. Eggers, M.; Vannoy, C.H.; Huang, J.; Purushothaman, P.; Brassard, J.; Fonck, C.; Meng, H.; Prom, M.J.; Lawlor, M.W.; Cunningham, J.; et al. Muscle-directed gene therapy corrects Pompe disease and uncovers species-specific GAA immunogenicity. EMBO Mol. Med. 2021, 14, e13968. [Google Scholar] [CrossRef]
  192. Hordeaux, J.; Dubreil, L.; Robveille, C.; Deniaud, J.; Pascal, Q.; Dequéant, B.; Pailloux, J.; Lagalice, L.; Ledevin, M.; Babarit, C.; et al. Long-term neurologic and cardiac correction by intrathecal gene therapy in Pompe disease. Acta Neuropathol. Commun. 2017, 5, 66. [Google Scholar] [CrossRef] [PubMed]
  193. Qiu, K.; Falk, D.J.; Reier, P.J.; Byrne, B.J.; Fuller, D.D. Spinal Delivery of AAV Vector Restores Enzyme Activity and Increases Ventilation in Pompe Mice. Mol. Ther. 2012, 20, 21–27. [Google Scholar] [CrossRef] [PubMed]
  194. Lee, N.C.; Hwu, W.L.; Muramatsu, S.I.; Falk, D.J.; Byrne, B.J.; Cheng, C.H.; Shih, N.C.; Chang, K.L.; Tsai, L.K.; Chien, Y.H. A neuron-specific gene therapy relieves motor deficits in Pompe Disease Mice. Mol. Neurobiol. 2018, 55, 5299–5309. [Google Scholar] [CrossRef] [PubMed]
  195. Lim, J.A.; Yi, H.; Gao, F.; Raben, N.; Kishnani, P.S.; Sun, B. Intravenous injection of an AAV-PHP.B vector encoding human acid alfa-glucosidase rescues both muscle and CNS defects in murine Pompe Disease. Mol. Ther. Methods Clin. Dev. 2019, 12, 233–245. [Google Scholar] [CrossRef]
  196. Sidonio, R.F.; Pipe, S.W.; Callaghan, M.U.; Valentino, L.A.; Monahan, P.E.; Croteau, S.E. Discussing investigational AAV gene therapy with hemophilia patients: A guide. Blood Rev. 2020, 47, 100759. [Google Scholar] [CrossRef] [PubMed]
  197. Franco, L.M.; Sun, B.; Yang, X.; Bird, A.; Zhang, H.; Schneider, A.; Brown, T.; Young, S.P.; Clay, T.M.; Amalfitano, A.; et al. Evasion of immune responses to introduced human acid alfa-glucosidase by liver-restricted expression in glycogen storage disease type II. Mol. Ther. 2005, 12, 876–884. [Google Scholar] [CrossRef]
  198. Keeler, G.D.; Markusic, D.M.; Hoffman, B.E. Liver induced transgene tolerance with AAV vectors. Cell. Immunol. 2017, 342, 103728. [Google Scholar] [CrossRef]
  199. Puzzo, F.; Colella, P.; Biferi, M.G.; Bali, D.; Paulk, N.K.; Vidal, P.; Collaud, F.; Simon-Sola, M.; Charles, S.; Hardet, R.; et al. Rescue of Pompe disease in mice by AAV-mediated liver delivery of secretable acid α-glucosidase. Sci. Transl. Med. 2017, 9, 6375. [Google Scholar] [CrossRef]
  200. Colella, P.; Sellier, P.; Gomez, M.J.; Biferi, M.G.; Tanniou, G.; Guerchet, N.; Cohen-Tannoudji, M.; Moya-Nilges, M.; van Wittenberghe, L.; Daniele, N.; et al. Gene therapy with secreted acid alpha-glucosidase rescues Pompe disease in a novel mouse model with early-onset spinal cord and respiratory defects. eBiomedicine 2020, 61, 103052. [Google Scholar] [CrossRef]
  201. Sun, B.; Zhang, H.; Benjamin, D.K.J.r.; Brown, T.; Bird, A.; Young, S.P.; McVie-Wylie, A.; Chen, Y.; Koeberl, D.D. Enhanced efficacy of an AAV vector encoding chimeric, highlySecreted acid α-glucosidase in glycogen storage disease type II. Mol. Ther. 2006, 14, 822–830. [Google Scholar] [CrossRef]
  202. Sun, B.; Kulis, M.D.; Young, S.P.; Hobeika, A.C.; Li, S.; Bird, A.; Zhang, H.; Li, Y.; Clay, T.M.; Burks, W.; et al. Immunomodulatory Gene Therapy Prevents Antibody Formation and Lethal Hypersensitivity Reactions in Murine Pompe Disease. Mol. Ther. 2010, 18, 353–360. [Google Scholar] [CrossRef] [PubMed]
  203. Han, S.-O.; Ronzitti, G.; Arnson, B.; Leborgne, C.; Li, S.; Mingozzi, F.; Koeberl, D. Low-Dose Liver-Targeted Gene Therapy for Pompe Disease Enhances Therapeutic Efficacy of ERT via Immune Tolerance Induction. Mol. Ther. Methods Clin. Dev. 2017, 4, 126–136. [Google Scholar] [CrossRef] [PubMed]
  204. Xu, F.; Ding, E.; Liao, S.X.; Migone, F.; Dai, J.; Schneider, A.; Serra, D.; Chen, Y.T.; Amalfitano, A. Improved efficacy of gene therapy approaches for Pompe Disease using a new, immune-deficient GSD-II mouse model. Gene Ther. 2004, 11, 1590–1598. [Google Scholar] [CrossRef] [PubMed]
  205. Puppi, J.; Guillonneau, C.; Pichard, V.; Bellodi-Privato, M.; Cuturi, M.C.; Anegon, I.; Ferry, N. Long term transgene expression by hepatocytes transduced with retroviral vectors requires induction of immune tolerance to the transgene. J. Hepatol. 2004, 41, 222–228. [Google Scholar] [CrossRef] [PubMed]
  206. Han, S.-O.; Li, S.; McCall, A.; Arnson, B.; Everitt, J.I.; Zhang, H.; Young, S.P.; ElMallah, M.K.; Koeberl, D.D. Comparisons of Infant and Adult Mice Reveal Age Effects for Liver Depot Gene Therapy in Pompe Disease. Mol. Ther. Methods Clin. Dev. 2019, 17, 133–142. [Google Scholar] [CrossRef]
  207. Han, S.O.; Gheorghiu, D.; Chang, A.; Mapatano, S.H.; Li, S.; Brooks, E.; Koerbel, D. Efficacious androgen hormone administration in combination with adeno-associated virus vector-mediated gene therapy in female mice with Pompe Disease. Hum. Gene Ther. 2022, 33, 479–491. [Google Scholar] [CrossRef]
  208. Unnisa, Z.; Yoon, J.K.; Schindler, J.W.; Mason, C.; van Til, N.P. Gene Therapy Developments for Pompe Disease. Biomedicines 2022, 10, 302. [Google Scholar] [CrossRef]
  209. Douillard-Guilloux, G.; Richard, E.; Batista, L.; Caillaud, C. Partial phenotypic correction and immune tolerance induction to enzyme replacement therapy after hematopoietic stem cell gene transfer of α-glucosidase in Pompe disease. J. Gene Med. 2009, 11, 279–287. [Google Scholar] [CrossRef]
  210. Piras, G.; Montiel-Equihua, C.; Chan, Y.-K.A.; Wantuch, S.; Stuckey, D.; Burke, D.; Prunty, H.; Phadke, R.; Chambers, D.; Partida-Gaytan, A.; et al. Lentiviral Hematopoietic Stem Cell Gene Therapy Rescues Clinical Phenotypes in a Murine Model of Pompe Disease. Mol. Ther. Methods Clin. Dev. 2020, 18, 558–570. [Google Scholar] [CrossRef]
  211. Stok, M.; de Boer, H.; Huston, M.W.; Jacobs, E.H.; Roovers, O.; Visser, T.P.; Jahr, H.; Duncker, D.J.; van Deel, E.D.; Reuser, A.J.; et al. Lentiviral Hematopoietic Stem Cell Gene Therapy Corrects Murine Pompe Disease. Mol. Ther. Methods Clin. Dev. 2020, 17, 1014–1025. [Google Scholar] [CrossRef]
  212. Tarallo, A.; Damiano, C.; Strollo, S.; Minopoli, N.; Indrieri, A.; Polishchuk, E.; Zappa, F.; Nusco, E.; Fecarotta, S.; Porto, C.; et al. Correction of oxidative stress enhances enzyme replacement therapy in Pompe disease. EMBO Mol. Med. 2021, 13, 14434. [Google Scholar] [CrossRef] [PubMed]
  213. Van der Wal, E.; Bergsma, A.J.; Pijnenburg, J.M.; Van der Ploeg, A.T.; Pijnappel, W.W.M.P. Antisense oligonucleotides promote exon inclusion and correct the common c. -32-13T > G GAA splicing variant in Pompe Disease. Mol. Ther. Nucleic Acids 2019, 7, 90–100. [Google Scholar] [CrossRef]
  214. Van der Wal, E.; Bergsma, A.J.; Van Gestel, T.J.M.; In ‘t Groen, S.L.M.; Zaehres, H.; Araúzo-bravo, M.J.; Schöler, H.R.; Van der Ploeg, A.T.; Pijnappel, W.W.M.P. GAA deficiency in Pompe Disease is alleviated by exon inclusion in iPSC-Derived skeletal muscle cells. Mol. Ther. Nucleic Acid. 2017, 7, 101–115. [Google Scholar] [CrossRef]
  215. Aung-Htut, M.T.; Ham, K.A.; Tchan, M.; Johnsen, R.; Schnell, F.J.; Fletcher, S.; Wilton, S.D. Splice modulating antisense oligonucleotides restore some acid-alpha-glucosidase activity in cells derived from patients with late-onset Pompe disease. Sci. Rep. 2020, 10, 6702. [Google Scholar] [CrossRef]
  216. Clayton, N.P.; Nelson, C.A.; Weeden, T.; Taylor, K.M.; Moreland, R.J.; Scheule, R.K.; Phillips, L.; Leger, A.J.; Cheng, S.H.; Wentworth, B.M. Antisense Oligonucleotide-mediated Suppression of Muscle Glycogen Synthase 1 Synthesis as an Approach for Substrate Reduction Therapy of Pompe Disease. Mol. Ther. Nucleic Acids 2014, 3, e206. [Google Scholar] [CrossRef] [PubMed]
  217. Tang, B.; Frasinyuk, M.S.; Chikwana, V.M.; Mahalingan, K.K.; Morgan, C.A.; Segvich, D.M.; Bondarenko, S.P.; Mrug, G.P.; Wyrebek, P.; Watt, D.S.; et al. Discovery and Development of Small-Molecule Inhibitors of Glycogen Synthase. J. Med. Chem. 2020, 63, 3538–3551. [Google Scholar] [CrossRef]
Figure 1. Quadriceps muscle biopsy in patients with LOPD. (A) Hematoxylin-eosin staining showing the presence of intracytoplasmic vacuolization with a “rimmed” appearance (arrow). (B) Periodic acid-Schiff (PAS) staining on semi-fine section showing the presence of abnormal accumulation of intrafibral glycogen. (C,D) Acid phosphatase staining showing the presence of lysosomal activation in scattered fibers.
Figure 1. Quadriceps muscle biopsy in patients with LOPD. (A) Hematoxylin-eosin staining showing the presence of intracytoplasmic vacuolization with a “rimmed” appearance (arrow). (B) Periodic acid-Schiff (PAS) staining on semi-fine section showing the presence of abnormal accumulation of intrafibral glycogen. (C,D) Acid phosphatase staining showing the presence of lysosomal activation in scattered fibers.
Biomolecules 13 01279 g001
Table 2. Some of the most common variants in the GAA gene.
Table 2. Some of the most common variants in the GAA gene.
DNA
Nomenclature
SiteProtein NomenclatureType of VariantBiochemical
Evidence
Predicted
Severity
Predicted Phenotype
with Null Allele
PopulationGnomAD
Frequency
c.-32-13T>GIntron 1p.[=,0]Deletion (translation
initiation site)
No effect on splicing
Protein is expressed Variant causes skipping of exon 2Potentially
mild
LOPD
(childhood or adult onset)
Caucasian0.00309
c.2560C>TExon 18p.(Arg854*)Non-senseNo protein expressionVery severeIOPDAfrican Descendent0.00048
c.841C>TExon 4p.(Arg281Trp)MissenseProtein is expressedPotentially mildLOPDEuropean
(non-Finnish)
0.00033
c.525delExon 2p.(Glu176Argfs*45)FrameshiftNo protein expressionVery severeIOPDEuropean
(non-Finnish)
0.00013
c.1979G>AExon 14p.(Arg660His)MissenseProtein is expressedPotentially
mild
LOPD
(childhood
onset)
African0.00009
c.853C>TExon 4p.(Pro285Ser)MissenseProtein is expressedMildLOPDAfrican0.00007
c.655G>AExon 3p.(Gly219Arg)MissenseProtein is expressedPotentially mildIOPDAfrican0.00006
c.953T>CExon 5p.(Met318Thr)MissenseProtein is expressedPotentially mildIOPDAfrican0.00006
c.1935C>AExon 14p.(Asp645Glu)MissenseProtein is expressedPotentially mildIOPDSouth Asian0.00004
Table 3. Main clinical findings in LOPD.
Table 3. Main clinical findings in LOPD.
LOPD
Clinical
Examination
Limb-girdle and axial weakness
Tongue involvement
Respiratory insufficiency
Easily fatigued
Systemic
Involvement
Musculoskeletal complications (i.e., hyperlordosis, kyphosis, scoliosis, spine rigid syndrome)
Brain vascular abnormalities
Laboratory TestsMildly elevated or normal serum creatine kinase levels
Elevation of AST/ALT levels
EMGMyopathic pattern with spontaneous activity including myotonic discharges, especially in axial muscles
Muscle MRIParavertebral and abdominal muscle involvement
Tongue involvement
Hip extensor involvement (i.e., adductor magnus) with sparing of leg muscles
Muscle BiopsyVacuolar myopathy with glycogen storage and acid phosphatase reactivity
Table 4. Main current and future therapeutic approaches for Pompe disease.
Table 4. Main current and future therapeutic approaches for Pompe disease.
TherapyMechanism of ActionStatus of Approval
Alglucosidase alfaEnzyme replacement therapy by intravenous infusion of recombinant human GAA (rhGAA)First approved 28 April 2006 (FDA)
Avalglucosidase alfaEnzyme replacement therapy by intravenous infusion of recombinant human GAA (rhGAA)First approved 6 August 2021 (FDA)
Active study recruiting:
An Open-label, Multinational, Multicenter, Intravenous Infusion Study of the Efficacy, Safety, Pharmacokinetics, and Pharmacodynamics of Avalglucosidase Alfa in Treatment Naïve Pediatric Participants With Infantile-Onset Pompe Disease (IOPD)
Cipaglucosidase alfa +
miglustat
Enzyme replacement therapy by intravenous infusion of recombinant human GAA (rhGAA) + small-molecule ligands (chaperones) as enzyme stabilizersFirst approved 23 March 2023 for LOPD (EU)
Active study recruiting:
Open-label Study to Evaluate the Safety, Efficacy, Pharmacokinetics, Pharmacodynamics, and Immunogenicity of Cipaglucosidase Alfa/Miglustat in Both ERT-Experienced and ERT- Naïve Pediatric Subjects With Infantile-Onset Pompe Disease Aged 0 to < 18 Years
Open-label Study of the Safety, Pharmacokinetics, Efficacy, Pharmacodynamics, and Immunogenicity of Cipaglucosidase Alfa/Miglustat in Pediatric Subjects Aged 0 to < 18 Years With Late- Onset Pompe Disease
Gene TherapyProviding cells with a healthy copy of the GAA gene to restore functional GAA enzyme productionNot approved.
Active study recruiting:
Single Arm, Multicenter, Open and Dose-Escalation Clinical Study on Safety, Tolerance, and Efficacy of GC301, an AAV-Delivered Gene Transfer Therapy in Patients with Infantile-Onset Pompe Disease
Active study, not recruiting:
Phase 1 Study of the Safety of AAV2/8-LSPhGAA (ACTUS-101) in Late-Onset Pompe Disease
Phase 1/2, Dose-Escalation Study to Evaluate the Safety, Tolerability, and Efficacy of a Single Intravenous Infusion of SPK-3006 in Adults with Late-Onset Pompe Disease
Open-Label, Fixed-Sequence, Ascending-Dose, First-in-Human Study to Assess the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Efficacy of Intravenous Infusions of ATB200 Co-Administered with Oral AT2221 in Adult Subjects with Pompe Disease
Phase 3 Open-label Extension Study to Assess the Long-Term Safety and Efficacy of Intravenous ATB200 Co-Administered With Oral AT2221 in Adult Subjects with Late-Onset Pompe Disease
Substrate Reduction TherapySmall-molecule inhibitors of human skeletal muscle glycogen synthaseNot approved.
Active study, not recruiting:
Phase 1, Randomized, Double-Blind, Placebo-Controlled, Single and Multiple Ascending Dose Study of MZE001 to Evaluate the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics in Healthy Subjects
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Labella, B.; Cotti Piccinelli, S.; Risi, B.; Caria, F.; Damioli, S.; Bertella, E.; Poli, L.; Padovani, A.; Filosto, M. A Comprehensive Update on Late-Onset Pompe Disease. Biomolecules 2023, 13, 1279. https://0-doi-org.brum.beds.ac.uk/10.3390/biom13091279

AMA Style

Labella B, Cotti Piccinelli S, Risi B, Caria F, Damioli S, Bertella E, Poli L, Padovani A, Filosto M. A Comprehensive Update on Late-Onset Pompe Disease. Biomolecules. 2023; 13(9):1279. https://0-doi-org.brum.beds.ac.uk/10.3390/biom13091279

Chicago/Turabian Style

Labella, Beatrice, Stefano Cotti Piccinelli, Barbara Risi, Filomena Caria, Simona Damioli, Enrica Bertella, Loris Poli, Alessandro Padovani, and Massimiliano Filosto. 2023. "A Comprehensive Update on Late-Onset Pompe Disease" Biomolecules 13, no. 9: 1279. https://0-doi-org.brum.beds.ac.uk/10.3390/biom13091279

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop