Elsevier

Revue Neurologique

Volume 169, Issues 8–9, August–September 2013, Pages 595-602
Revue Neurologique

Muscular pathology
The French Pompe registry. Baseline characteristics of a cohort of 126 patients with adult Pompe diseaseRegistre Français de la maladie de Pompe. Caractérisation d’une cohorte de 126 patients adultes

https://doi.org/10.1016/j.neurol.2013.07.002Get rights and content

Abstract

Pompe disease is a rare autosomal recessive muscle lysosomal glycogenosis, characterised by limb-girdle muscle weakness and frequent respiratory involvement. The French Pompe registry was created in 2004 with the initial aim of studying the natural history of French patients with adult Pompe disease. Since the marketing in 2006 of enzyme replacement therapy (alglucosidase alfa, Myozyme®), the French Pompe registry has also been used to prospectively gather the biological and clinical follow-up data of all adult patients currently treated in France. This report describes the main clinical and molecular features, at the time of inclusion in the French registry, of 126 patients followed up in 21 hospital-based neuromuscular or metabolic centres. Sixty-five men and 61 women have been included in the registry. Median age at inclusion was 49 years, and the median age at onset of progressive limb weakness was 35 years. Fifty-five percent of the patients were walking without assistance, 24% were using a stick or a walking frame, and 21% were using a wheelchair. Forty-six percent of the patients needed ventilatory assistance, which was non-invasive in 35% of the cases. When performed, muscle biopsies showed specific features of Pompe disease in less than two-thirds of the cases, confirming the importance of acid alpha-glucosidase enzymatic assessment to establish the diagnosis. Molecular analysis detected the common c.-32-13T > G mutation, in at least one allele, in 90% of patients. The French Pompe registry is so far the largest country-based prospective study of patients with Pompe disease, and further analysis will be performed to study the impact of enzyme replacement therapy on the progression of the disease.

Résumé

La maladie de Pompe est une glycogénose musculaire rare, de transmission autosomique récessive, caractérisée par une faiblesse des ceintures, fréquemment associée à une insuffisance respiratoire. Le registre français de la maladie de Pompe a été créé en 2004, avec pour objectif initial d’étudier l’histoire naturelle des patients atteints de la forme adulte de la maladie de Pompe. Depuis la commercialisation de l’enzymothérapie substitutive par alglucosidase alfa (Myozyme®) en 2006, le registre français de la maladie de Pompe a aussi permis le recueil de données cliniques et biologiques pour l’ensemble des patients adultes actuellement traités en France. Ce travail décrit les principales caractéristiques cliniques et moléculaires des 126 patients adultes, qui sont suivis dans 21 centres de référence de maladies neuromusculaires ou métaboliques. Soixante-cinq hommes et 61 femmes ont été inclus dans le registre. L’âge médian à l’inclusion était de 49 ans ; l’âge médian du début de la faiblesse musculaire évolutive des ceintures était de 35 ans. Soixante-cinq pour cent des patients pouvaient marcher sans aide, 24 % avaient recours à l’aide d’une canne ou d’un déambulateur, et 21 % avaient recours au fauteuil roulant. Quarante-six pour cent des patients avaient recours à une ventilation assistée, avec une ventilation non invasive dans 35 % des cas. La biopsie musculaire montrait des anomalies caractéristiques de la maladie de Pompe dans moins de deux tiers des cas, lorsqu’elle avait été pratiquée, confirmant ainsi l’importance du dosage enzymatique de l’activité alpha-glucosidase acide pour établir le diagnostic. Les analyses moléculaires ont permis de détecter la présence de la mutation commune c.-32-13T > G sur au moins un des deux allèles chez 90 % des patients. Le registre français de la maladie de Pompe a permis de constituer la plus grande cohorte de patients adultes atteints de cette maladie, et suivis de façon prospective, à l’échelle d’un pays. Des analyses complémentaires sont en cours afin d’étudier les effets de l’enzymothérapie substitutive sur l’évolution de la maladie.

Introduction

Pompe disease, or glycogen storage disease type II, is an autosomal recessive disorder caused by a deficiency in the activity of the lysosomal enzyme acid α-glucosidase (GAA). The clinical spectrum of this disease is extremely broad, varying from a severe infantile form leading to cardiorespiratory failure in the first months of life to adult-onset muscular manifestations (limb-girdle weakness and diaphragmatic paralysis) occurring in the third or fourth decade. Despite the fact that the infantile form was the first to be described (Pompe, 1932), the adult form is the most common one (Byrne et al., 2011, van der Ploeg and Reuser, 2008), representing more than 90% of diagnosed patients in France. Adults with Pompe disease typically present with limb-girdle muscle and axial weakness associated with diaphragmatic involvement (Engel, 1970, Laforêt et al., 2000). Disease progression is variable, but generally occurs over decades, potentially confining some patients to a wheelchair or causing them to require respiratory assistance (Hagemans et al., 2005, Pellegrini et al., 2005). Mean age for wheelchair or respiratory assistance has been estimated at around 50 years (Hagemans et al., 2005), and the major cause of death is respiratory failure.

The pathophysiology of this disease is particularly complex, with major cell disturbances caused by lysosomal glycogen accumulation and increased autophagy. Glycogen accumulation per se is not the only cause of cell dysfunction, and autophagy probably also has a major role in the pathogenic process, with recent works showing an autophagic build-up due to dysfunction of the endocytic and autophagic pathways in the muscle fibres (Raben et al., 2010). Autophagy seems to be more prominent in the juvenile and adult forms than in the infantile form of Pompe disease (Raben et al., 2010).

The prognosis for infantile Pompe disease has been modified considerably by enzyme replacement therapy (ERT), introduced 10 years ago (Kishnani et al., 2007, Kishnani et al., 2009). This treatment (alglucosidase alfa, Myozyme®), administered intravenously every other week, may improve cardiac and motor function and prolong the survival of affected babies. However at least one third of children do not respond to ERT, and ultimately die or do not reach a normal motor milestone.

In the adult form, results obtained from a double-blind, placebo-controlled study (LOTS) has shown a moderate but significant improvement in walking distance, and a stabilisation in sitting vital capacity after 18 months of treatment with alglucosidase alfa (Myozyme®) (van der Ploeg et al., 2010). Several open studies on older children and adults, published in recent years, have confirmed that ERT may improve or stabilise limb muscle strength and respiratory function (Strothotte et al., 2010, Bembi et al., 2010, Orlikowski et al., 2011, van Capelle et al., 2008, van Capelle et al., 2010, Angelini et al., 2011, Furusawa et al., 2012, Regnery et al., 2012), but data is still needed in order to better predict the long-term benefit of ERT.

Several guidelines reflecting consensus recommendations for the management and treatment of Pompe disease in various countries have also been published (Bembi et al., 2008, Llerena et al., 2009, Cupler et al., 2012). Their purpose was to define criteria for the diagnosis and management of specific symptoms, pre-treatment assessment, indications and monitoring of ERT. However, there is still a lack of global consensus today concerning the precise clinical criteria for ERT initiation and interruption in adults. Continuation of data gathering across the entire spectrum of Pompe disease, via national or international patient registries, is therefore warranted in order to assess the long-term safety and efficacy of ERT, and to formulate more precise guidelines for treatment. An international Pompe registry developed by Genzyme (www.pomperegistry.com) enrolled patients all over the world, but no data has yet been published on the efficacy and tolerance of treatment for patients with long-term follow-up (Byrne et al., 2011).

In order to improve our knowledge of Pompe disease, a national (French) Pompe registry has been created, with the collaboration of all French reference centres for rare neuromuscular and metabolic diseases. Below are the main clinical, pathological and molecular features of patients with adult-onset Pompe disease who have been included in the French Pompe registry since it was first set up.

Section snippets

Objectives of the French Pompe registry

Since 2004, we have been developing a national registry, with the aim of prospectively gathering the clinical, functional and biological data of all French patients with a diagnosis of Pompe disease confirmed by enzymatic and/or molecular analysis, whether treated or not. This registry has been qualified since 2008 by the French National Committee for Rare Diseases Registries (CNR-MR) supported by Inserm and institut national de veille sanitaire (InVS). Funding for the running of the registry

Demography

One hundred and twenty-six patients were included in the French Pompe registry at the time of data extraction in September 2012 (Fig. 1). Sixty-one patients are women (48%) and 65 are men (52%). Parental consanguinity was noticed in only five patients. Forty-two patients (33%) reported affected siblings. Three siblings of three patients and eight siblings of two patients have been included in the registry so far. Several siblings were not included in the registry because they had died, were not

Muscle biopsy

Ninety-three muscle biopsies were performed in an overall number of 84 patients (nine patients underwent two muscle biopsies). Muscle biopsy was performed in the quadriceps in 49 cases, in the deltoid in 34 cases, and in another muscle in 10 cases. Vacuoles were observed in 63% of the muscle biopsies, and were most often observed in the quadriceps (67%) compared to the deltoid muscle (50%). An increase in PAS staining was observed in 54% of the muscle biopsies. Acid phosphatase activity was

Discussion

Several publications have reported, for nearly 20 years, the description of clinical and molecular features of large series of patients with the adult form of acid alpha-glucosidase deficiency, also named acid maltase deficiency or “late-onset Pompe disease” (Wokke et al., 1995, Laforêt et al., 2000, Hagemans et al., 2005). Worldwide collections of clinical data have also been published recently, based either on self-assessment questionnaires (Güngör et al., 2011), or on data collection from

Disclosure of interest

Pascal Laforêt reports having received honoraria and grants from Sanofi-Genzyme laboratory.

The other authors declare that they have no conflicts of interest concerning this article.

Acknowledgements

We thank Genzyme-Sanofi, the Association Française contre les myopathies (AFM), the Association Francophone des Glycogénoses (AFG), Inserm, InVS (Institut National de Veille Sanitaire) for funding the French Pompe registry.

References (27)

  • C. Angelini et al.

    Observational clinical study in juvenile-adult glycogenosis type 2 patients undergoing enzyme replacement therapy for up to 4 years

    J Neurol

    (2011)
  • B. Bembi et al.

    Management and treatment of glycogenosis type II

    Neurology

    (2008)
  • B. Bembi et al.

    Long-term observational, non-randomized study of enzyme replacement therapy in late-onset glycogenosis type II

    J Inherit Metab Dis

    (2010)
  • Cited by (44)

    • Early clinical phenotype of late onset Pompe disease: Lessons learned from newborn screening

      2022, Molecular Genetics and Metabolism
      Citation Excerpt :

      Data from other countries with successful NBS programs for Pompe disease, such as Taiwan, have limited application to patients in the US due to the overall lack of the IVS1 variant in non-Caucasian populations. Herein, we report on the phenotypic spectrum that is observed among patients with LOPD under 2 years of age, most of which possess at least one copy of the IVS1 variant, the most common LOPD variant in Caucasians [7,12,16,17,26]. Patients in our cohort with this variant demonstrated a spectrum of clinical manifestations of disease, similar to what has been documented in the literature [9,22].

    • Gene therapy with secreted acid alpha-glucosidase rescues Pompe disease in a novel mouse model with early-onset spinal cord and respiratory defects

      2020, EBioMedicine
      Citation Excerpt :

      The respiratory phenotype is particularly relevant to PD, since currently available Gaa KO mice present very mild respiratory defects [18, 21–27]. Respiratory problems instead manifest in all forms of the disease, representing one of the leading causes of morbidity, in addition to IOPD-specific cardiomyopathy [5, 28–32]. While cardiac problems have been widely reported to be effectively treated by ERT [5], the effective treatment of the PD respiratory phenotype still represents a medical need [14, 32].

    View all citing articles on Scopus
    1

    The French Pompe Study Group : Dr A.-L. Bedat-Millet (Centre de compétence de pathologie neuromusculaire, CHU Charles Nicolle, Rouen), Dr A. Behin, Pr B. Eymard, Dr T. Stojkovic, A. Canal, Dr V. Decostre, G. Ollivier, Dr P. Carlier, Pr R.-Y. Carlier, Dr C. Wary (Centre de référence de pathologie neuromusculaire Paris-Est, institut de Myologie, groupe hospitalier Pitié-Salpêtrière, Paris), Pr F. Boyer (Service de médecine physique et de réadaptation, CHU de Reims), Dr C. Caillaud (Laboratoire de biochimie génétique, faculté de médecine Cochin, Paris), Dr Y. Castaing (Service de réanimation, CHU de Bordeaux, Bordeaux), Dr P. Cintas (Centre SLA et maladies neuromusculaires, CHU de Toulouse-Rangueil, Toulouse, France), Pr I. Durieu (Service de médecine interne, centre hospitalier Lyon Sud, Pierre-Bénit), Dr A. Echaniz-Laguna (Département de neurologie, hôpitaux universitaires de Strasbourg, hôpital de Hautepierre, Strasbourg) , Dr L. Feasson, Dr A. Furby (Centre de référence des maladies neuromusculaires rares Rhône-Alpes, Hôpital Nord, CHU de Saint-Étienne), Dr X. Ferrer, Dr. G .Solé (Centre de référence des maladies neuromusculaires, CHU de Bordeaux-GH Sud - hôpital Haut-Lévêque, Pessac), Dr R. Froissart, Dr M. Piraud (Centre de biologie et pathologie Est, hospices civils de Lyon, Bron), Dr N. Guffon-Fouilhoux (Unité des maladies métaboliques, département de pédiatrie, CHU de Lyon-GH Est - hôpital Femme-Mère-Enfant, Bron), Dr H. Journel (Génétique médicale, centre hospitalier Bretagne-Atlantique, Vannes), Dr P. Kaminsky (Centre de référence des maladies héréditaires du métabolisme de Nancy, centre hospitalier universitaire de Nancy, hôpitaux de Brabois, Vandœuvre), Pr P Labauge (Département de Neurologie, CHU de Montpellier, Montpellier), Dr A. Levy (Service de pneumologie, centre hospitalier Jacques-Cœur, Bourges), Dr S. Louis (Service de neurologie, hôpital Central, Nancy), Dr A. Magot (Centre de référence des maladies neuromusculaires Nantes-Angers, Hôtel Dieu, Nantes), Dr M.-C. Minot-Myhié (service neurologie, CHU de Rennes, Rennes), Dr A. Nadaj-Pakleza, Anne-Catherine Aubé-Nathier (Centre de référence des maladies neuromusculaires Nantes/Angers, Service de neurologie, CHU d’Angers, Angers), Dr N. Pellegrini (Service de soins de suite et de réadaptation neurologie, GHI du Vexin, Aincourt), Dr P. Petiot (Centre de référence maladies neuromusculaires de la région Rhône-Alpes, hôpital de la Croix-Rousse, Lyon), Dr J. Praline (Centre de compétences maladies neuromusculaires, CHRU de Tours, Tours), Dr H. Prigent (Service de physiologie et explorations fonctionnelles, hôpital Raymond Poincaré, Garches), Dr D. Renard (CHU de Nîmes, hôpital Caremeau, Nîmes), Dr V. Tiffreau (Centre de référence des maladies neuromusculaires, CHRU de Lille, Lille), Dr D. Vincent (Service de neurologie, groupe hospitalier La Rochelle – Ré – Aunis, La Rochelle).

    View full text