ArticlePDF Available

Enzyme replacement therapy for mucopolysaccharidosis VI: Evaluation of long-term pulmonary function in patients treated with recombinant human N-acetylgalactosamine 4-sulfatase

Authors:

Abstract

Pulmonary function is impaired in untreated mucopolysaccharidosis type VI (MPS VI). Pulmonary function was studied in patients during long-term enzyme replacement therapy (ERT) with recombinant human arylsulfatase B (rhASB; rhN-acetylgalactosamine 4-sulfatase). Pulmonary function tests prior to and for up to 240 weeks of weekly infusions of rhASB at 1 mg/kg were completed in 56 patients during Phase 1/2, Phase 2, Phase 3 and Phase 3 Extension trials of rhASB and the Survey Study. Forced vital capacity (FVC), forced expiratory volume in 1 s (FEV1) and, in a subset of patients, maximum voluntary ventilation (MVV), were analyzed as absolute volume in liters. FEV1 and FVC showed little change from baseline during the first 24 weeks of ERT, but after 96 weeks, these parameters increased over baseline by 11% and 17%, respectively. This positive trend compared with baseline continued beyond 96 weeks of treatment. Improvements from baseline in pulmonary function occurred along with gains in height in the younger group (5.5% change) and in the older patient group (2.4% change) at 96 weeks. Changes in MVV occurred earlier within 24 weeks of treatment to approximately 15% over baseline. Model results based on data from all trials showed significant improvements in the rate of change in pulmonary function during 96 weeks on ERT, whereas little or no improvement was observed for the same time period prior to ERT. Thus, analysis of mean percent change data and longitudinal modeling both indicate that long-term ERT resulted in improvement in pulmonary function in MPS VI patients.
ORIGINAL ARTICLE
Enzyme replacement therapy for mucopolysaccharidosis
VI: evaluation of long-term pulmonary function in patients
treated with recombinant human N-acetylgalactosamine
4-sulfatase
Paul Harmatz &Zi-Fan Yu &Roberto Giugliani &Ida Vanessa D. Schwartz &
Nathalie Guffon &Elisa Leão Teles &M. Clara Miranda &J. Edmond Wraith &
Michael Beck &Laila Arash &Maurizio Scarpa &David Ketteridge &John J. Hopwood &
Barbara Plecko &Robert Steiner &Chester B Whitley &Paige Kaplan &
Stuart J. Swiedler &Karen Hardy &Kenneth I. Berger &Celeste Decker
Received: 22 June 2009 / Revised: 4 November 2009 / Accepted: 9 November 2009 / Published online: 6 February 2010
#The Author(s) 2010. This article is published with open access at Springerlink.com
Abstract Pulmonary function is impaired in untreated
mucopolysaccharidosis type VI (MPS VI). Pulmonary
function was studied in patients during long-term enzyme
replacement therapy (ERT) with recombinant human
arylsulfatase B (rhASB; rhN-acetylgalactosamine 4-
sulfatase). Pulmonary function tests prior to and for up
to 240 weeks of weekly infusions of rhASB at 1 mg/kg
were completed in 56 patients during Phase 1/2, Phase 2,
Phase 3 and Phase 3 Extension trials of rhASB and the
Survey Study. Forced vital capacity (FVC), forced
expiratory volume in 1 s (FEV1) and, in a subset of
patients, maximum voluntary ventilation (MVV), were
Communicated by: Frits Wijburg
References to electronic databases: MPS VI: OMIM 253200.
The authors are reporting for the MPS VI Study Group; see
Acknowledgments for list of co-investigators.
P. Harmatz (*):K. Hardy
Childrens Hospital & Research Center Oakland,
747 52nd Street,
Oakland, CA 94609, USA
e-mail: Pharmatz@mail.cho.org
Z.-F. Yu
Statistics Collaborative, Inc,
Washington, DC, USA
R. Giugliani :I. V. D. Schwartz
Medical Genetics Service/ HCPA and Department of Genetics/
UFRGS,
Porto Alegre, Brazil
N. Guffon
Hôpital Femme Mère Enfant, Service Maladies Métaboliques,
Bron, France
E. L. Teles
Departamento Pediatria, Hospital de Sao João,
Unidade de Doenças Metabólicas,
Porto, Portugal
M. C. S. Miranda
Instituto de Biologia Molecular e Celular,
Unidade de Biologia do Lisossoma e Peroxisoma,
Porto, Portugal
J. E. Wraith
Royal Manchester Childrens Hosp,
Manchester, UK
M. Beck :L. Arash
Childrens Hosp, University of Mainz,
Mainz, Germany
M. Scarpa
Department of Pediatrics,
University of Padova,
Padova, Italy
D. Ketteridge
SA Pathology at Womens and Childrens Hospital,
North Adelaide, SA, Australia
J Inherit Metab Dis (2010) 33:5160
DOI 10.1007/s10545-009-9007-8
analyzed as absolute volume in liters. FEV1 and FVC
showed little change from baseline during the first
24 weeks of ERT, but after 96 weeks, these parameters
increased over baseline by 11% and 17%, respectively.
This positive trend compared with baseline continued
beyond 96 weeks of treatment. Improvements from
baseline in pulmonary function occurred along with gains
in height in the younger group (5.5% change) and in the
older patient group (2.4% change) at 96 weeks. Changes
in MVV occurred earlier within 24 weeks of treatment to
approximately 15% over baseline. Model results based on
data from all trials showed significant improvements in the
rate of change in pulmonary function during 96 weeks on
ERT, whereas little or no improvement was observed for
the same time period prior to ERT. Thus, analysis of mean
percent change data and longitudinal modeling both
indicate that long-term ERT resulted in improvement in
pulmonary function in MPS VI patients.
Abbreviations
MPS VI Mucopolysaccharidosis VI
rhASB Recombinant human arylsulfatase B
ERT Enzyme replacement therapy
GAG Glycosaminoglycans
FVC Forced vital capacity
FEV1 Forced expiratory volume in 1 s
MVV Maximum voluntary ventilation
LME Longitudinal linear mixed-effects model
Introduction
Mucopolysaccharidosis type VI (MPS VI; Maroteaux-
Lamy syndrome) is a lysosomal storage disease in which
deficient activity of the enzyme N-acetylgalactosamine 4-
sulfatase (arylsulfatase B, or ASB; E.C # 3.1.6.12) impairs
the stepwise degradation of the glycosaminoglycan (GAG)
dermatan sulfate (DS) (Giugliani et al. 2007). Partially
degraded GAG accumulates in lysosomes and in a wide
range of tissues, causing a chronic progressive disorder
characterized by significant functional impairment and a
shortened lifespan.
A survey of 121 MPS VI affected individuals found that
high urinary GAG values (>200 μg/mg creatinine) were
associated with an accelerated clinical course including
reduced endurance, greater pulmonary function impairment,
and lower height values for age (Swiedler et al. 2005).
Impairment in endurance based on a 6-min walk test was
observed across all age groups and levels of GAG
accumulation.
Three enzyme replacement therapy (ERT) studies using
recombinant human ASB (known as rhASB ; recombinant
human N-acetylgalactosamine 4-sulfatase; galsulfase;
Naglazyme®) to treat patients with MPS VI have been
reported. A Phase 1/2 study and a Phase 2 study both
demonstrated that weekly infusions of 1 mg/kg rhASB were
well tolerated, produced a rapid reduction in urinary GAG
levels, and improved endurance in patients with rapidly
advancing disease (Harmatz et al. 2004; Harmatz et al.
2005). A Phase 3 double-blind, placebo-controlled study
demonstrated greater improvement in endurance on the 12-
min walk test (12MWT) in patients treated with rhASB for
24 weeks compared with patients receiving placebo
(Harmatz et al. 2006). In all studies, improvement in
endurance was maintained during the open-label extension
phase for up to 240 weeks, with an acceptable safety
profile (Harmatz et al. 2008). In addition to endurance
measures, all three studies included pulmonary function
assessments. The mechanism for this improved endurance
is unknown but may relate to an impact of ERT on
pulmonary function.
The purpose of this paper is to evaluate pooled long-term
data from the clinical ERT trials and the Survey Study to
determine the impact of ERT on pulmonary function in
patients with MPS VI.
Methods
Study design
Detailed study design and evaluation criteria have been
reported in previous publications of the MPS VI Survey
J. J. Hopwood
Department of Genetic Medicine,
Womens and Childrens Hospital Adelaide,
North Adelaide, SA, Australia
B. Plecko
Univ. Klinik für Kinder und Jugendheilkunde,
Graz, Austria
R. Steiner
Departments of Pediatrics and Molecular and Medical Genetics,
Oregon Health & Science University,
Portland, OR, USA
C. B. Whitley
University of Minnesota Medical School,
Minneapolis, MN, USA
P. Kaplan
Childrens Hospital of Philadelphia,
Philadelphia, PA, USA
S. J. Swiedler :C. Decker
BioMarin Pharmaceutical Inc,
Novato, CA, USA
K. I. Berger
Departments of Medicine, Physiology and Neuroscience,
New York University School of Medicine,
New York, NY, USA
52 J Inherit Metab Dis (2010) 33:5160
Study and the Phase 1/2, Phase 2, and Phase 3 clinical
studies of rhASB (galsulfase, Naglazyme) treatment in
MPS VI, which reported results of 48 weeks of treatment
(Harmatz et al. 2004; Harmatz et al. 2005; Harmatz et al.
2006). Collection of efficacy data continued for up to
240 weeks during the extension phase of these studies.
Pretreatment data were specifically collected from patients in
the Survey Study and placebo-treated patients in the first
24 weeks of the Phase 3 clinical trial. These studies are
summarized in Table 1. An Institutional Review Board (IRB)
or Ethics Committee (EC) at each participating clinical site
approved each study. All adult patients and parent/guardians
gave written consent; patients younger than 18 years old
gave written assent according to local IRB regulations.
All patients received rhASB at 1 mg/kg/week infused over
a 4-h period, except three patients in the Phase 1/2 study who
received 0.2 mg/kg per week for the initial part of that study
and 19 patients who completed the Phase 3 study. These 19
patients received placebo during the blinded portion of the
Phase 3 study and received rhASB for the open-label
remainder of the study (weeks 2496). These patients
underwent evaluations following 24 and 72 weeks of active
therapy, i.e., at weeks 48 and 96 of the study. Assessments
were completed for each study group as shown in Table 2.
Pulmonaryfunction parameters examined during all studies
included forced vital capacity (FVC) and forced expiratory
volume in 1 s (FEV1). Data were obtained in accord with the
American Thoracic Society guidelines (1995). The Phase 3
study also measured the maximum voluntary ventilation
(MVV), which was defined as the maximum volume of air
that can be breathed in 1 min. Data were collected while the
patients breathed as deeply and quickly as possible for 15 s
and then were extrapolated to 1 min (American Thoracic
Society guidelines 1991).
Analysis methods
This analysis focused on long-term pulmonary function
outcomes in patients receiving ERT over an extended
period. In addition, the pooled data were analyzed to
determine: (1) the importance of height change on the mean
improvement in pulmonary function during ERT, and (2)
the mean rate of increase in pulmonary function parameters
prior to and following ERT initiation. To evaluate the
improvement in pulmonary function relative to growth,
pooled data were stratified into two groups: patients
<12 years versus patients 12 years at treatment initiation.
The age of 12 years was chosen to approximate the
midpoint of normal pubertal development, and it is
assumed that patients who started ERT after this age are
less likely to experience significant growth. In each age
group, mean percent change in FVC, FEV1, and height
were analyzed.
Table 1 Summary of study populations
Study Study design Study time
period (years)
Duration
of efficacy
evaluations
(weeks)
Number of
patients enrolled/
completed
Dose of
rhASB
mg/kg
Age (years)
mean±SD
(range years)
Sex
(M/F)
Height (cm)
mean±SD
Number of patients
withdrawn: time
of withdrawal
Baseline urinary
GAG (µg/mg
creatinine)
mean±SD
Phase 1/2 Double-blind, randomized,
dose comparison/
open-label extension
20012005 240 7/5 0.2
1.0
12.0± 3.8 (716 ) 4/3 107.5±21.5 1 (0.2 mg/kg)
after week 3;
1 (0.2 mg/kg)
after week 32
365± 148
Survey study
(patients not
on ERT)
Cross-sectional study
of patients not on ERT
20022003 123/121 None 13.9± 10 (456) 58/63 115± 26 321± 200
Phase 2 Open-label, nonrandomized 20022006 144 10/10 1.0 12.1± 5.3 (621) 7/3 103.7±14.4 0 336± 116
Phase 3 Double-blind, placebo-
controlled, randomized/
open-label extension
20032006 96 39/38 1.0 13.7± 6.5 (rhASB)
10.7± 4.4 (placebo)
(529)
13/26 104.4± 2.9
(rhASB)
100.3± 13.5
(placebo)
1 (placebo group)
after week 5
346± 128 (rhASB)
330± 114 (placebo)
J Inherit Metab Dis (2010) 33:5160 53
To determine the mean rate of increase in pulmonary
function parameters prior to and following initiation of ERT,
a longitudinal linear mixed-effects model (LME) was
constructed using pooled data. The model incorporated both
pre-ERT and post-ERT data by including a linear spline for
time with a knot at treatment week 0. This formulation
allows different slopes of the mean trend before and after
ERT initiation. A random intercept gives all individuals their
own regression lines with separate intercepts that deviate
from the population line. The longitudinal model includes
repeated measures over time and allows observations within
a patient to be correlated. The model uses empirical estimates
for the standard error, which in large samples correctsfor
misspecifying the correlation structure. The model also
includes baseline height as a covariate.
Timerefers to time from ERT initiation. Because data
were also obtained before the start of treatment, time includes
negative values. The length of follow-up for each trial phase
differs; most patients have at least 7296 weeks of follow-up
after ERT initiation, and the LME method is flexible in that
patients do not have to have all measurements at all time
points. The availability of pretreatment data is also limited;
analysis therefore restricts the length of time to approximately
a 2-year window on either side of ERT initiation.
Results
Baseline data
The age of patients at time of enrollment in a clinical
therapy trial ranged from 5 to 29 years; the mean age was
approximately 12 years. An overview of baseline height
and pulmonary function data is presented in Table 3.As
expected, patients <12 years were shorter on average than
patients in the older age group, with a mean height of
99.4 cm versus 105.4 cm, respectively. Pulmonary function
parameters were significantly impaired for age in compar-
ison with a healthy population (Rosenthal et al. 1993). The
younger group showed mean values for FVC and FEV1 of
0.56 L and 0.52 L, respectively, whereas the older group
showed similar mean values of 0.55 L and 0.48 L,
respectively, for the same measures.
Mean observed improvement in pulmonary function
(FEV1, FVC, and MVV) during ERT
Data showing mean percent change in FEV1, FVC, and
MVV during ERT are presented in Fig. 1. Changes from
baseline in FEV1 and FVC were minimal up to 24 weeks of
Study Evaluation Treatment week
0 24 48 72 96 144 192 240
Phase 1/2 FVC X X X X X X X
FEV1 X X X X X X X
Height X X X X X X X
Phase 2 FVC X X X X X X PSC PSC
FEV1 X X X X X X PSC PSC
Height X X X X X X PSC PSC
Phase 3 rhASB/rhASB FVC X X X X PSC PSC PSC
FEV1 X X X X PSC PSC PSC
MVV X X X X PSC PSC PSC
Height X X X X X PSC PSC PSC
Phase 3 placebo/rhASB FVC X X X PSC PSC PSC PSC
FEV1 X X X PSC PSC PSC PSC
MVV X X X PSC PSC PSC PSC
Height X X X X PSC PSC PSC PSC
Table 2 Schedule of assessments
Phase 1/2 had no week 72 but
had week 84 PFT height
assessments. PSC, post-study
completion
Table 3 Combined Phase 1, Phase 2, and Phase 3 data: baseline height and pulmonary function
All patients; mean (range) Number Age <12years; mean (range) Number Age 12years; mean (range) Number
Height (cm) 101.9 (81.5, 136) 54 99.4 (81.5, 133) 32 105.4 (86, 136) 22
FVC (L) 0.56 (0.16, 1.74) 53 0.56 (0.16, 1.64) 32 0.55 (0.28, 1.74) 21
FEV1 (L) 0.50 (0.16, 1.67) 53 0.52 (0.16, 1.42) 32 0.48 (0.25, 1.67) 21
54 J Inherit Metab Dis (2010) 33:5160
treatment with rhASB but increased thereafter through
96 weeks of treatment. For FVC and FEV1, those on
treatment for 72 weeks improved 14% from baseline on
average (p<0.001) for both outcomes. Those on treatment
for 96 weeks improved approximately 17% (p= 0.009) and
11% (p= 0.014), respectively, relative to baseline. Changes
in MVV occurred earlier. At 24 weeks of treatment, MVV
increased approximately 15% over baseline (p=0.021).
Although sample sizes beyond 144 weeks of treatment were
too small to make valid inferences, this trend of pulmonary
function improvement compared with baseline appears to
continue through 240 weeks of treatment.
Pulmonary function improvement from baseline relative
to growth
In order to examine whether the observed change in
pulmonary function could be attributed to growth, we
examined pulmonary function after dividing the population
into two groups based on age at time of treatment initiation:
older or younger than 12 years (Table 4; Fig. 2). The left
panel of Fig. 2graphs data obtained in patients <12 years.
Both FEV1 and FVC showed little change from baseline
during the first 24 weeks of ERT. By 96 weeks of treatment,
these parameters showed meaningful improvement, with
increases in FEV1 and FVC averaging approximately 10%
and 13%, respectively, with respect to baseline. Height
increased concomitantly with increases in FEV1 and FVC in
the younger age group.
The right panel of Fig. 2graphs similar data in older
patients (age12 years). As with the younger patients,
FEV1 and FVC did not improve relative to baseline in the
first 24 weeks but showed meaningful improvement in
subsequent weeks. For those on 96 weeks of treatment,
FEV1 and FVC improved from baseline by approximately
13% and 23%, respectively. However, in contrast to the
younger patient group, improvements in FEV1 and FVC
seen in older patients occurred despite a smaller percent
increase in height.
Mean rate of increase in pulmonary function parameters
(FVC, FEV1) prior to and following ERT initiation
The observed increases over time during ERT are described
using longitudinal modeling of the absolute changes in lung
function relative to baseline, defined as the week prior to
ERT initiation. The longitudinal models used all available
pre-ERT and post-ERT data.
Regression analyses
Model results showed improvements for all patients on
ERT compared with before ERT (Table 5;Fig.3). For
FEV1, the estimated mean value for all patients at baseline
was 0.49 L. For approximately 2 years prior to ERT, mean
FEV1 increased only 0.01 L on average. In contrast, over
2 years on ERT, mean FEV1 increased 0.06 L on average
(p<0.001) (Table 5). This improvement in FEV1
corresponds to an increase of approximately 12% relative
to baseline value. When patients were subdivided into
those <12 years versus 12 years, the change in FEV1
post-ERT still remained, corresponding to approximately
Weeks on ERT
Percent change
0 24 48 72 96 144 192 240
n*=53 53 34 28 33 14 5 5
*N refers to the number of patients for whom data were available for that particular timepoint;
sample sizes at each timepoint do not necessaril
y
include the same patients
0
10
20
30
40
50
Changes from baseline in FVC and FEV1 -
All ERT-Treated Patients (Phase 1, 2, and 3 Data)
FVC
FEV1
Weeks on ERT
Percent change
024487296
n=34 30 16 17 14
0
10
20
30
40
50
Changes from baseline in MVV -
ERT-Treated Phase 3 Data
Fig. 1 Mean percent change in FVC, FEV1, and MVV by treatment week over all available patient data
J Inherit Metab Dis (2010) 33:5160 55
14% in the younger group and 11% in the older group
(refer to Table 5; small differences in percentages
between text and Table 5are related to actual data versus
modeled data).
For FVC, the estimated mean for all patients at baseline
was 0.54 L. In the 2 years prior to ERT, mean FVC increased
approximately 0.01 L on average. In contrast, over 2 years on
ERT, mean FVC increased 0.10 L on average (p<0.001)
(Table 5). This improvement in FVC corresponds to an
increase of approximately 19% relative to baseline value.
Both younger and older groups demonstrated improvement
in FVC after treatment (approximately 14% and 25%,
respectively) compared with lesser improvements before
treatment (refer to Table 5). For all parameters, the younger
group showed minimal or no improvement prior to treat-
ment, with significant improvement after ERT initiation.
Older patients demonstrated minor improvement in lung
function pre-ERT, with significant improvement after ERT
(Table 5).
Discussion
Studies have demonstrated that ERT with rhASB leads to a
sustained improvement in endurance in the MPS VI patient
population (Harmatz et al. 2008). An important factor
contributing to improved endurance is likely to be
pulmonary function. In this study, analysis of pooled
pulmonary function data from rhASB clinical studies shows
that MPS VI patients on ERT demonstrated improvement
from baseline in pulmonary function that was sustained
over long-term treatment and occurred independent of age.
Whereas the improvement in pulmonary function may be in
part related to growth in the younger patients, the
pulmonary function improvement seen in older patients
occurred with smaller change in height and may be
attributed to other mechanical, anatomical, or physiological
factors influencing lung function.
Consistent with previously reported findings in individual
MPS VI clinical trials, analysis of combined data did not show
mean percent improvement in FVC and FEV1 over the short
term (24 weeks). However, by 72 or 96 weeks of treatment,
both FVC and FEV1 showed improvement from baseline of at
least 11%. For individuals with normal lung function, a 15%
relative increase in FEV1 year to year is considered a
clinically meaningful change according to the American
Thoracic Society guidelines (1991) (Pellegrino et al. 2005).
It is important to note that we examined improvement in
pulmonary function in terms of absolute volume, not percent
predicted. These gains could not be expressed in terms of
percent predicted ([actual result/predicted result ] x 100%),
as a standard curve does not exist for this population, which
Table 4 Combined Phase 1, Phase 2, and Phase 3 data: percent change height and pulmonary function from start of ERT by age group
Treatment
week
Number
a
FEV1 (L) FVC (L) Height (cm)
Observed
mean (SD)
% Change
mean (SD)
Observed
mean (SD)
% Change
mean (SD)
n Observed
mean (SD)
% Change
mean (SD)
Age <12 years
0 32 0.52 (0.26) 0.56 (0.31) 32 99.4 (11.1)
24 32 0.52 (0.31) 0.96 (21.0) 0.57 (0.37) 1.5 (21.7) 32 101.0 (11.2) 1.6 (1.1)
48 17 0.62 (0.41) 7.1 (32.5) 0.69 (0.48) 6.8 (32.0) 30 102.9 (11.4) 3.6 (1.6)
72 20 0.55 (0.30) 15.3 (18.1) 0.57 (0.21) 17.5 (20.0) 29 103.4 (11.2) 4.9 (1.7)
96 17 0.64 (0.44) 9.6 (28.8) 0.73 (0.51) 12.5 (32.9) 27 105.7 (11.8) 5.5 (2.1)
144 7 0.75 (0.41) 11.1 (15.4) 0.91 (0.48) 17.6 (11.6) 9 110.6 (13.6) 8.6 (2.4)
192 3 0.73 (0.35) 16.4 (21.6) 0.99 (0.49) 39.6 (14.7) 3 116.2 (16.0) 8.6 (3.9)
240 3 0.70 (0.30) 13.6 (12.1) 0.98 (0.48) 39.8 (12.0) 3 120.2 (17.0) 12.2 (3.1)
Age 12 years
0 21 0.48 (0.30) 0.55 (0.32) 22 105.4 (12.8)
24 21 0.49 (0.29) 2.6 (14.6) 0.54 (0.32) 2.0 (9.6) 22 106.3 (12.5) 0.9 (1.4)
48 17 0.54 (0.32) 8.2 (16.0) 0.60 (0.22) 5.2 (17.6) 20 109.1 (11.8) 1.8 (1.7)
72 8 0.47 (0.19) 9.7 (9.6) 0.52 (0.22) 6.5 (7.3) 17 109.9 (12.6) 2.7 (2.1)
96 16 0.57 (0.32) 12.9 (20.4) 0.74 (0.49) 22.6 (39.0) 19 110.2 (12.4) 2.4 (2.9)
144 7 0.49 (0.22) 17.0 (27.4) 0.57 (0.21) 20.2 (20.9) 10 107.9 (10.3) 1.2 (2.1)
192 2 0.47 (0.02) 35.9 (3.6) 0.52 (0.06) 49.0 (22.2) 2 96.5 (3.5) -1.5 (1.5)
240 2 0.39 (0.03) 14.5 (16.5) 0.46 (0.08) 30.6 (27.5) 2 100.1 (6.3) 2.0 (2.4)
Patient population will not necessarily include exactly the same patients at each timepoint.
a
Percent change FEV1 and FVC have same n
56 J Inherit Metab Dis (2010) 33:5160
is similar to the achondroplasia patient population in which
small stature and dysplastic bone changes confound calcu-
lation of these percentages (Stokes et al. 1988;1990).
In contrast to the delayed improvement in traditional
pulmonary function measures of FVC and FEV1, the MVV
showed rapid improvement relative to baseline over
24 weeks. The MVV maneuver of rapid respiration is
thought to replicate maximal ventilation during exercise
(Stein et al. 2003). Although MVV is generally well
correlated with FEV1 (Fulton et al. 1995; Stein et al.
2003), a disproportionate decrease in MVV relative to
FEV1hasbeenreportedinneuromusculardisorders
(Serisier et al. 1982; Braun et al. 1983) and upper airway
obstruction (Engstroem et al. 1964), and therefore, im-
Model Predicted mean
at baseline
Time period N Predicted change
over 2 years (SE)
Pvalue
FEV1 (L) All patients 0.49 Pre-ERT 35 0.01±0.03 0.84
Post-ERT 53 0.06 ± 0.02 <0.001
Age <12 years 0.51 Pre-ERT 22 0.00 ± 0.02 0.93
Post-ERT 32 0.07 ± 0.02 0.005
Age 12 years 0.46 Pre-ERT 13 0.02 ±0.07 0.81
Post-ERT 21 0.05 ± 0.02 0.027
FVC (L) All patients 0.54 Pre-ERT 35 0.01 ± 0.03 0.71
Post-ERT 53 0.10 ± 0.03 <0.001
Age <12 years 0.56 Pre-ERT 22 -0.02 ± 0.02 0.47
Post-ERT 32 0.08 ± 0.02 0.001
Age 12 years 0.51 Pre-ERT 13 0.07± 0.09 0.41
Post-ERT 21 0.13 ± 0.06 0.036
Table 5 Predicted changes
over 2 years based on
longitudinal model results
Percent change
0 24 48 72 96 144 192 240
n=32 32 17 20 17 7 3 3
n=32 32 30 29 27 9 3 3
0
10
20
30
40
50
Weeks on ERT
FVC, FEV1
Height
Patient age < 12 years
FVC
FEV1
Height
0 24 48 72 96 144 192 240
n=21 21 17 8 16 7 2 2
n=22 22 20 17 19 10 2 2
0
10
20
30
40
50
Weeks on ERT
FVC, FEV1
Height
Patient age >= 12 years
FVC
FEV1
Height
Fig. 2 Mean percent change in height and pulmonary function by treatment week and age group over all available patient data
J Inherit Metab Dis (2010) 33:5160 57
provement in these areas with ERT may contribute to the
earlier response on the MVV assessment.
The mechanism for the observed improvement in lung
function during ERT and its relationship to growth is of
interest. In this study, lung function improved relative to
baseline to a similar extent in younger and older age
groups, suggesting height did not determine this improve-
ment. Observations in other MPS disorders during ERT
suggest that the improvement in lung function in older
patients may be due to multiple mechanisms, including
decreased upper airway obstruction as evidenced by
improvement in sleep apnea severity, increased chest wall
compliance as evidenced by improved joint mobility, and
improved respiratory muscle strength and endurance as well
as improved diaphragmatic excursion as evidenced by
reduction in liver size (Wraith et al. 2004; Clarke et al.
2009). In younger patients, all of these mechanisms may
apply, and height/thoracic enlargement may have an
additive effect on FVC and FEV1.
A limitation of examining mean percent change data by
treatment week is that the number of patients does not
remain consistent across all data points due to variations in
design and length of the three clinical trials, potentially
distorting the magnitude of changes over time. To minimize
this effect, longitudinal modeling was chosen to estimate
improvement trends at 96 weeks pre-ERT and post-ERT
initiation. Modeled results in Table 5do not reflect
significant improvement pre-ERT but do show significant
improvement post-ERT. In general, the magnitude of the
changes did not differ greatly in the younger and older age
groups, demonstrating that pulmonary function improve-
ment occurs in ERT-treated patients regardless of age at
treatment initiation.
There are several factors that may have influenced the
results of the longitudinal modeling. In the pre-ERT data,
comprising data that were collected in the Survey Study
and from placebo patients in the Phase 3 study, some
individuals had only one or two observations within the 2-
year period prior to ERT initiation. Because these data
tend to be variable, additional observations over time may
have given a more accurate estimate of lung function
during this time period. It is reassuring, then, that these
data were collected in a controlled clinical setting and that
the standard errors for pre-ERT versus post-ERT estimates
of lung function are similar in both time periods. In
addition, the number of patients with data beyond
96 weeks was limited, and thus, the observed trends
should not be extrapolated beyond the range of data
presented. Because the observed data showed a gradual
improvement over time, a linear trend for modeling was
chosen as a simple way to see whether the rate of
improvement differed during the 2 years pre-ERT and
post-ERT initiation. Individual patients may deviate from
this trend, especially if growth and/or puberty occurred
during treatment. In addition, longer-term follow-up
(>2 years) may suggest more distinct nonlinear trends,
but in this study, these would be difficult to detect or
differentiate from random variation.
In this study, we cannot rule out the effect of growth on
FVC. Whereas we considered including growthi.e., time-
varying heightin our model, several issues limited this
possibility. Treatment with ERT may affect lung function
through several causal pathways: it may have a direct and
independent effect on lung function, or increase height,
which in turn changes lung function. In the second
scenario, height may be an intermediate variable in the
causal pathway for lung function, particularly for FVC. As
a result, a statistical model that controls for time-varying
height may be inappropriate; it would likely obscure any
effect of treatment that was mediated by height. Accord-
ingly, we considered only baseline height in our model
rather than height over time (i.e., growth).
In conclusion, progressive impairment in pulmonary
function is characteristic of MPS VI disease, and a
FVC (L)
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1.0
1.1
1.2
1.3
1.4
1.5
1.6
1.7
1.8
1.9
2.0
2.1
2.2
Weeks prior to ERT Weeks on ERT
-96 -72 -48 -24 0 24 48 72 96
Fig. 3 Observed FVC (L) and modeled regression line. Dots show the
scatter of all patientsFVC measurements over time
58 J Inherit Metab Dis (2010) 33:5160
significant amount of morbidity and mortality is attributable
to respiratory complications (Simmons et al. 2005). The
study presented here suggests by multiple statistical
techniques that this trend toward decline in pulmonary
function can be halted and partially reversed during ERT
with N-acetylgalactosamine 4-sulfatase (rhASB, galsulfase,
Naglazyme) over a period of 96 weeks of therapy. It is
likely that this improvement is one factor underlying the
increase in endurance documented in the 6-min and 12-min
walk tests, although changes in pulmonary function appear
to be delayed relative to improvement in endurance that is
evident by 24 weeks of ERT (Harmatz et al. 2008). This
improvement in respiratory function relative to baseline
may lead to a decrease in the severity of respiratory
illnesses and number of hospitalizations, and an overall
improvement in the quality of life of MPS VI patients.
Acknowledgments We acknowledge the participation of study
patients and their families and the expert assistance of all study-site
coordinators and personnel. We also acknowledge the key contribu-
tions of our colleagues Dr. Ann Lowe and Ms. Mary Newman, as well
as the many other BioMarin employees and consultants who
performed important roles during the studies. Dr. Helen Nicely of
BioMarin contributed to the editing of this document. This study was
sponsored by BioMarin Pharmaceutical Inc., and supported, in part,
with funds provided by the National Center for Research Resources,
5 M01 RR-01271 (Dr. Harmatz), 5 M01 RR-00400 (Dr. Whitley),
M01 RR-00334 (Dr. Steiner), and UL1-RR-024134 (Dr. Kaplan). The
content is solely the responsibility of the authors and does not
necessarily represent the official views of the National Center for
Research Resources or the National Institutes of Health.
The MPS VI Study Group co-investigators are: John Waterson,MD,
PhD and Elio Gizzi, MD, Childrens Hospital & Research Center
Oakland, Oakland, California; Yasmina Amraoui, MD, Childrens
Hosp, University of Mainz, Germany; Bonito Victor, MD, Unidade de
Doenças Metabólicas, Departamento Pediatria, Hospital de Sao João,
Porto, Portugal; Javier Arroyo, MD, Hospital San Pedro de Alcantara,
Hospital de a de Pediatría, Caceres, Spain; D.N. Bennett-Jones,MD,
Consultant General & Renal Physician, Whitehaven, UK; Philippe
Bernard, MD, Centre Hospitalier dArras, Arras, France; Prof. Billette
de Villemeur, Hôpital Trousseau, Paris, France; Raquel Boy,MD,
Hospital Universitário Pedro Ernesto, Rio de Janeiro, Brazil; Eduardo
Coopman, MD, Hospital del Cobre De. Salvador, Calama, Chile; Prof.
Rudolf Korinthenberg, Universitätsklinikum Freiburg, Zentrum für
Kinderheilkunde und Jugendmedizin, Klinik II Neuropädiatrie und
Muskelerkrankungen, Freiburg, Germany; Michel Kretz, MD, Hôpital
Civil de Colmar, Le Parc Centre de la Mère et de lEnfant, Colmar,
France; Shuan-Pei Lin, MD, MacKay Memorial Hospital, Department
of Genetics, Taipei, Taiwan; Ana Maria Martins, MD, UNIFESP,
Instituto de Oncologia Pediátrica, GRAACC/UNIFESP, Departamento
de Pediatria, São Paulo, Brazil; Anne OMeara, MD, Our Ladys
Hospital for Sick Children, Dublin, Ireland; Gregory Pastores,MD,
PhD, NYU Medical Center, Rusk Institute, New York, New York;
Lorenzo Pavone,MD,Rita Barone,MD,Agata Fiumara, MD, and
Prof. Giovanni Sorge, Department of Pediatrics, University of Catania,
Catania, Italy; Silvio Pozzi, MD, Ospedale Vito Fazzi, UO Pediatria,
Lecce, Italy; Uwe Preiss, MD, Universitätsklinik und Poliklinik fűr
Kinder, Halle, Germany; Emerson Santana Santos, MD, Fundação
Universidade de Ciências da Saúde de Alagoas Governador, Departa-
mento de Pediatria, Maceió, Brazil; Isabel Cristina Neves de Souza,
MD and Luiz Carlos Santana da Silva,PhD,UniversidadeFederaldo
Pará, Centro de Ciências Biológicas, Hospital Universitário João de
Barros Barreto, Belém, Brazil; Eugênia Ribeiro Valadares, MD, PhD,
Hospital das Clínicas, Faculdade de Medicina da Universidade Federal
de Minas Gerais-UFMG, Avenida Professor Alfredo Balena, Belo
Horizonte-Minas Gerais, Brazil; Laura Keppen, MD, Department of
Pediatrics, University of South Dakota School of Medicine, Sioux Falls,
SD; David Sillence, MD, Childrens Hospital, Westmead, Australia;
Lionel Lubitz, MD, Royal Childrens Hospital, Melbourne, Australia;
William Frischman, MD, The Townsville Hospital, Townsville,
Australia; Julie Simon, RN, Childrens Hospital & Research Center
Oakland, Oakland, California; Claudia Lee,MPH,Childrens Hospital
& Research Center Oakland, Oakland, California; Stephanie Oates,RN
Department of Genetic Medicine, Womens and Childrens Hospital
Adelaide, North Adelaide, Australia; Lewis Waber, MD, PhD, Pediatric
Genetics and Metabolism, University of Texas Southwest Medical
Center, Dallas, TX; Ray Pais, MD, Pediatric Hematology/Oncology,
East Tennessee Childrens Hospital, Knoxville, TN
Conflict of interest Drs. Harmatz, Beck, Giugliani, Berger, Steiner,
and Yu have provided consulting support to BioMarin Pharmaceutical
Inc, Novato, CA, USA. Dr. Hopwood has received commercial
research project funding to assist the development of enzyme
replacement therapy for MPS VI patients. Drs. Harmatz and Arash
each report receiving a speakers honorarium and travel support from
BioMarin. BioMarin is a supporter of the Lysosomal Disease Networks
WORLD Symposium organized by Dr. Whitley. Drs. Swiedler and
Decker are former and current employees of BioMarin Pharmaceutical,
Inc., respectively; both are stockholders.
Open Access This article is distributed under the terms of the
Creative Commons Attribution Noncommercial License which per-
mits any noncommercial use, distribution, and reproduction in any
medium, provided the original author(s) and source are credited.
References
American Thoracic Society (1991) Lung function testing: selection of
reference values and interpretative strategies. Am Rev Respir Dis
144(5):12021218
American Thoracic Society (1995) Standardization of Spirometry,
1994 Update. Am J Respir Crit Care Med 152(3):11071136
Braun NM, Arora NS et al (1983) Respiratory muscle and pulmonary
function in polymyositis and other proximal myopathies. Thorax
38(8):616623
Clarke LA, Wraith JE et al (2009) Long-term efficacy and safety of
laronidase in the treatment of mucopolysaccharidosis I. Pediatrics
123(1):229240
Engstroem H, Grimby G et al (1964) Dynamic spirometry in patients
with tracheal stenosis. Acta Med Scand 176:329334
Fulton JE, Pivarnik JM et al (1995) Prediction of maximum voluntary
ventilation (MVV) in African-American adolescent girls. Pediatr
Pulmonol 20(4):225233
Giugliani R, Harmatz P et al (2007) Management guidelines for
mucopolysaccharidosis VI. Pediatrics 120(2):405418
Harmatz P, Whitley CB et al (2004) Enzyme replacement therapy in
mucopolysaccharidosis VI (Maroteaux-Lamy syndrome). J
Pediatr 144(5):574580
Harmatz P, Ketteridge D et al (2005) Direct comparison of measures
of endurance, mobility, and joint function during enzyme-
replacement therapy of mucopolysaccharidosis VI (Maroteaux-
Lamy syndrome): results after 48 weeks in a phase 2 open-label
J Inherit Metab Dis (2010) 33:5160 59
clinical study of recombinant human N-acetylgalactosamine 4-
sulfatase. Pediatrics 115(6):e681689
Harmatz P, Giugliani R et al (2006) Enzyme replacement therapy for
mucopolysaccharidosis VI: a phase 3, randomized, double-blind,
placebo-controlled, multinational study of recombinant human
N-acetylgalactosamine 4-sulfatase (recombinant human arylsul-
fatase B or rhASB) and follow-on, open-label extension study. J
Pediatr 148(4):533539
Harmatz P, Giugliani R et al (2008) Long-term follow-up of endurance
and safety outcomes during enzyme replacement therapy for
mucopolysaccharidosis VI: final results of three clinical studies
of recombinant human N-acetylgalactosamine 4-sulfatase. Mol
Genet Metab 94(4):469475
Pellegrino R, Viegi G et al (2005) Interpretative strategies for lung
function tests. Eur Respir J 26(5):948968
Rosenthal M, Bain SH et al (1993) Lung function in white
children aged 4 to 19 years: ISpirometry. Thorax 48(8):794802
Serisier DE, Mastaglia FL et al (1982) Respiratory muscle function and
ventilatory control. I in patients with motor neurone disease. II in
patients with myotonic dystrophy. Q J Med 51(202):205226
Simmons MA, Bruce IA et al (2005) Otorhinolaryngological
manifestations of the mucopolysaccharidoses. Int J Pediatr
Otorhinolaryngol 69(5):589595
Stein R, Selvadurai H et al (2003) Determination of maximal voluntary
ventilation in children with cystic fibrosis. Pediatr Pulmonol 35
(6):467471
Stokes DC, Pyeritz RE et al (1988) Spirometry and chest wall
dimensions in achondroplasia. Chest 93(2):364369
Stokes DC, Wohl ME et al (1990) The lungs and airways in achondroplasia.
Do little people have little lungs? Chest 98(1):145152
Swiedler SJ, Beck M et al (2005) Threshold effect of urinary
glycosaminoglycans and the walk test as indicators of disease
progression in a survey of subjects with Mucopolysaccharidosis
VI (Maroteaux-Lamy syndrome). Am J Med Genet 134A(2):
144150
Wraith JE, Clarke LA et al (2004) Enzyme replacement therapy
for mucopolysaccharidosis I: a randomized, double-blinded,
placebo-controlled, multinational study of recombinant hu-
man alpha-L-iduronidase (laronidase). J Pediatr 144(5):581
588
60 J Inherit Metab Dis (2010) 33:5160
... Galsulfase has been approved by the Food and Drug Administration in 2005 and by the European Medicines Agency in 2006 [1,7,8]. The pivotal clinical trials of galsulfase have shown rapid and sustained reductions in urinary GAGs (uGAG) and significant and sustained improvements in endurance in the 6-min walk test (6MWT) and 3-min stair climb test (3MWCT), and in pulmonary function in treated patients, as well as an acceptable safety profile [9][10][11][12][13]. Glasulfase has been available to MPS VI patients in Turkey since 2006. ...
... Only case 3 retained height within 2 SDs from normal values at last follow-up. Comparison of height at treatment initiation and last follow-up with MPS VI-specific growth curves [18] showed a shift towards a higher height percentile in one patient (case 2), no change in six patients (cases 4,5,6,9,10,14), and a shift towards a lower percentile in seven patients (cases 1, 3, 7, 8, 11, 12, 13) (Additional file 1: S2). All patients showed an increase in weight after initiation of ERT. ...
... Bone abnormalities did not resolve during follow-up in any of the patients. Additional abnormalities on radiography, mostly kyphosis or scoliosis, were reported for nine cases (1,4,7,8,9,10,11,12, and 13) before initiation of ERT and at last follow-up. Bone age remained normal in all patients throughout the study, with deviations from chronological age being smaller than 1.5 years for all cases. ...
Article
Full-text available
Background The objective of this study was to describe clinical manifestations and events of patients with mucopolysaccharidosis (MPS) VI in Turkey who are treated with galsulfase enzyme replacement therapy (ERT). Clinical data of 14 children with MPS VI who were followed up at the Department of Pediatrics of the Gazi University Faculty of Medicine in Ankara, Turkey were retrospectively collected from the patients’ medical records. Patients were selected based on availability of a pre-ERT baseline and follow-up clinical data for a similar period of time (1.9–3.2 years). Event data (occurrence of acute clinical events, onset of chronic events, surgeries) collected during hospital visits and telemedicine were available for up to 10 years after initiation of ERT (2.5–10 years). Results Age at initiation of ERT ranged from 2.8 to 15.8 years (mean age 7.5 years). All patients presented with reduced endurance and skeletal abnormalities (dysostosis multiplex) on radiography. Other common clinical manifestations were cardiac valve disease (N = 13), short stature (N = 11), cranial abnormalities on MRI (N = 10), spinal abnormalities on MRI (N = 7), and mild cognitive impairment (N = 6). School attendance was generally poor, and several patients had urinary incontinence. After 1.9 to 3.2 years of ERT, most patients showed improvements in endurance in the 6-min walk test and 3-min stair climb tests; the frequency of urinary incontinence decreased. ERT did not seem to prevent progression of cardiac valve disease, eye disorders, hearing loss, or bone disease. Long-term event-based data showed a high incidence of respiratory tract infections, adenotonsillectomy/adenoidectomy, reduced sleep quality, sleep apnea, and depression before initiation of ERT. The number of events tended to remain stable or decrease in all patients over 2.5–10 years follow-up. However, the nature of the events shifted over time, with a reduction in the frequency of respiratory tract infections and sleep problems and an increase in ophthalmologic events, ear tube insertions, and depression. Conclusions This case series shows the high disease burden of the MPS VI population in Turkey and provides a unique insight into their clinical journey based on real-life clinical and event-based data collected before and after initiation of ERT.
... Although respiratory and cardiac complications are key drivers of mortality, and daily respiratory physiotherapy is recommended in these patients [3][4][5][6], current knowledge regarding respiratory function in MPS is based upon limited lung function and polysomnography data [5,6]. Although hepatosplenomegaly is suggested to elevate the diaphragm and alter its mechanical properties, diaphragm mobility is unkown in this population [6, [11][12][13][14]. Therefore, we evaluated respiratory muscle strength, diaphragm mobility, functional capacity, and quality of life of MPS patients and compared them with healthy individuals paired by age and body mass index. ...
... A restrictive ventilatory pattern is also common in patients with MPS and worsens with increasing age [39]. This was expected due to ribcage deformity, causing mechanical disadvantage of inspiratory and expiratory muscles and also contributing to reduced respiratory muscle strength [7, 14,40,41]. The FEV 1 /FVC ratio was not altered in MPS children, probably due to an absence of macroglossia and GAG infiltration in the oropharynx and trachea soft tissues. ...
Article
Full-text available
Purpose: We investigated respiratory muscle strength, diaphragm mobility, lung function, functional capacity, quality of life, body composition, breathing pattern, and chest wall (VT,CW) and compartmental volumes of Mucopolysaccharidosis (MPS) patients and compared these variables with matched healthy individuals. Methods: A cross-sectional study with data analyzed separately according to age group. A total of 68 individuals (34 MPS and 34 matched-healthy subjects) were included. Six-minute walking test assessed functional capacity and ultrasound assessed diaphragm mobility during quiet spontaneous breathing (QB). Optoelectronic plethysmography assessed VT,CW and breathing pattern during QB in two different positions: seated and supine (45° trunk inclination). Results: Body composition, lung function, respiratory muscle strength, and functional capacity were reduced in MPS (all p < 0.01). Diaphragm mobility was only reduced in adolescents (p = 0.01) and correlated with body composition and breathing pattern. Upper chest wall compartmental volumes were significantly lower in MPS, while abdominal volume only differed significantly in adolescents. Percentage contribution (%) of upper ribcage compartments to tidal volume was reduced in MPS children, whereas %AB was significantly increased compared with healthy subjects. Conclusion: Lung function, respiratory muscle strength, functional capacity, diaphragm mobility, and quality of life are reduced in MPS compared with matched healthy subjects. VT,CW was mainly reduced due to pulmonary and abdominal ribcage impairment. Implications for RehabilitationReduction in respiratory muscle strength, functional capacity, diaphragm excursion and low lung volumes were found in individuals with Mucopolysaccharidoses (MPS).Chest wall volumes and the upper chest wall compartmental volumes during quiet spontaneous breathing are reduced in MPS.Assessment and monitoring of the respiratory system for individuals with MPS should be performed periodically through standardized assessments to enable identification of changes and early intervention by rehabilitation protocols.This study may provide the necessary basis for carrying out respiratoty rehabilitation protocols that can improving chest wall mechanics with breathing exercise in this group.
... As MPS I is a disease caused by progressive accumulation of GAG, early treatment is of great importance to prevent irreversible pathologies and substantially improve a patient's life expectancy [4][5][6][7][8][9]. Non-specific symptoms, variable presentations, and lack of disease awareness are some factors that prevent early and accurate diagnosis of the syndrome [10]. ...
Article
Full-text available
Hurler syndrome is a rare autosomal recessive disorder of deficiency in the metabolism of glycosaminoglycans (GAGs), including heparan sulfate and dermatan sulfate, which consequently accumulate in the different organs of the body, resulting from deficiency of an enzyme named Alpha-L-iduronidase. Here, we present an interesting case of a young female patient who presented with a combination of skeletal, oro-facial, ophthalmologic, neurological, and radiological findings of this disease. A diagnosis of Hurler syndrome (Mucopolysaccharidosis Type I) was made late in the disease due to lack of facilities, and the patient was ultimately managed supportively.
... evaluando la respuesta a TRE con medición de VEF 1 y CVF previo y posterior a la terapia, observándose una mejoría del 14% en ambos parámetros a las 72 semanas de terapia y 17% para CVF y 11% para VEF 1 a las 96 semanas posterior al inicio de tratamiento. No se conoce si esto es debido a una disminución en la inflamación o infiltración de la vía aérea, como mejoría de los volúmenes pulmonares al disminuir la visceromegalia que presentan, mejorando así los volúmenes y capacidades pulmonares o la asociación de una mejoría propia del crecimiento corporal de los niños (29) . ...
Article
Full-text available
Las Mucopoliscaridosis (MPS) son parte de las denominadas enfermedades lisosomales. El depósito de los distintos glicosaminoglicanos comprometidos, dependiendo del déficit enzimático, genera manifestaciones multisistémicas, en donde el sistema respiratorio es uno de los principales afectados y que se asocia con morbilidad y mortalidad significativa. Los diferentes tipos de MPS presentan un grado variable de compromiso desde etapas precoces de la vida, síntomas de obstrucción de vía aérea superior de grado variable, rinorrea persistente, otitis media, patología obstructiva de vía aérea periférica, neumonías o infecciones asociadas a un mal drenaje mucociliar son las principales manifestaciones que los pacientes presentan. El compromiso neurológico y musculo esquelético, trae consigo además el compromiso de la bomba respiratoria. Desde esa perspectiva el enfoque debe ser multidisciplinario, ya que el compromiso abarca varios órganos y sistemas. Las actuales terapias están dirigidas a reemplazar la enzima deficitaria, disponibles sólo para algunas de ellas, esto trae consigo el retardo de la evolución de la enfermedad pero no lo evita, considerando que más aun no tiene ningún efecto sobre el sistema nervioso central, por lo que el compromiso cognitivo es inevitable. El trasplante de médula es una terapia no exenta de complicaciones, pero que es capaz de cambiar la progresión de la enfermedad en las etapas precoces de ella. El enfoque terapéutico se basa en terapia de sostén y el manejo de las distintas complicaciones que se van dando, siendo éstos los ejes del siguiente artículo.
... [1][2] Due to genetic mutations, lysosomes in patients with MPS are deficient in enzymes necessary to break down GAGs. 3 As a result, GAGs accumulate causing swelling and interference with cell function, which leads to deleterious effects on various organ systems associated with MPS. 3 MPS VI is caused by a decreased activity of N-acetylgalactosamine-4-sulfatase (arylsulfatase B [ASB]) and is estimated to affect between 1 in 230 000 and 1 in 1 300 000 individuals. 4 Current treatment guidelines recommend the enzyme replacement therapy (ERT), galsulfase, to improve walking ability, endurance, and pulmonary function and to reduce urinary GAGs (uGAGs). [5][6][7][8][9] Galsulfase has limited effects on cardiovascular, ophthalmological, or skeletal manifestations of MPS VI. 5,[10][11][12][13] Odiparcil is an orally available small molecule previously studied at doses up to 1000 mg/day for prevention of venous thrombosis with no safety findings observed in >1900 subjects/patients. 14,15 By acting as a substrate for galactosyltransferase I, odiparcil diverts the synthesis of endogenous and soluble dermatan sulfate (DS) and chondroitin sulfate (CS) GAG bound to odiparcil, excreted from the cells, bypassing lysosomal degradation, reducing GAG load, and eliminated via the urine as shown in in vitro and in vivo models. ...
Article
Mucopolysaccharidosis (MPS) disorders are a group of rare, progressive lysosomal storage diseases characterized by the accumulation of glycosaminoglycans (GAGs) and classified according to the deficient enzyme. Enzyme replacement therapy of MPS VI has limited effects on ophthalmic, cardiovascular, and skeletal systems. Odiparcil is an orally-available small molecule that results in the synthesis of odiparcil linked GAGs facilitating their excretion and reducing cellular and tissue GAG accumulation. iMProveS (improve MPS treatment) was a Phase 2a study of the safety, pharmacokinetics/pharmacodynamics (PK/PD), and efficacy of two doses of odiparcil in patients with MPS VI. The core study was a 26-week, randomized, double-blind, placebo-controlled trial in patients receiving ERT and an open-label, non-comparative, single dose cohort not receiving ERT. Patients aged >16 years receiving ERT were randomized to odiparcil 250 mg or 500 mg twice daily or placebo. Patients without ERT received odiparcil 500 mg twice daily. Of 20 patients enrolled, 13 (65.0%) completed the study. Odiparcil increased total urine GAGs (uGAG), chondroitin sulfate (CS), and dermatan sulfate (DS) concentrations. A linear increase in uGAG levels and odiparcil exposure occurred with increased odiparcil dose. Odiparcil demonstrated a good safety and tolerability profile. Individual analyses found more improvements on pain, corneal clouding, cardiac, vascular, and respiratory functions in the odiparcil groups vs. placebo. This study confirmed the mechanism of action and established the safety of odiparcil with clinical beneficial effects after only a short treatment duration in an advanced stage of disease. Further assessment of odiparcil in younger patients is needed. This article is protected by copyright. All rights reserved.
Preprint
Full-text available
Background Mucopolysaccharidosis type I (MPS I) is an autosomal recessive multisystem lysosomal storage disorder. Methods Herein, we report the Egyptian experience of enzyme replacement therapy (ERT) for MPS type I patients and the faced challenges. Thirty-eight MPS-I patients were examined at presentation and throughout ERT to evaluate its effect on different body systems. Clinical and radiological examination of the patients confirmed the characteristic manifestations. Results Follow up after one year of ERT initiation revealed improvement of respiratory function tests, significant decrease in the size of liver and spleen, a stationary course of cardiac problems and a decrease of total urinary glycosaminoglycans (GAGs) levels. We experienced the problems of late presentation, time consuming procedures to get approval for ERT and receiving the treatment thus, leading to delayed ERT initiation in addition to irregular interrupted ERT courses due to delay in treatment renewal and difficulties in patient’s transportation from far governorates. Laronidase was generally well tolerated apart from mild infusion-related adverse reactions. Conclusion ERT is an effective treatment in the management of MPS-I patients. Early diagnosis, less complicated process for treatment approval, effecient multidisciplinary centers that are aware of the disease manifestations and able to provide ERT are recommended.
Article
Background: Mucopolysaccharidosis type VI (MPS VI) or Maroteaux-Lamy syndrome is a rare genetic disorder caused by the deficiency of arylsulphatase B. The resultant accumulation of dermatan sulphate causes lysosomal damage. The clinical symptoms are related to skeletal dysplasia (i.e. short stature and degenerative joint disease). Other manifestations include cardiac disease, impaired pulmonary function, ophthalmological complications, hepatosplenomegaly, sinusitis, otitis, hearing loss and sleep apnea. Intellectual impairment is generally absent. Clinical manifestation is typically by two or three years of age; however, slowly progressive cases may not present until adulthood. Enzyme replacement therapy (ERT) with galsulfase is considered a new approach for treating MPS VI. Objectives: To evaluate the effectiveness and safety of treating MPS VI by ERT with galsulfase compared to other interventions, placebo or no intervention. Search methods: Eletronic searches were performed on the Cystic Fibrosis and Genetic Disorders Group's Inborn Errors of Metabolism Trials Register. Date of the latest search: 09 June 2021. Further searches of the following databases were also performed: CENTRAL, MEDLINE, LILACS, the Journal of Inherited Metabolic Disease, the World Health Organization International Clinical Trials Registry Platform and ClinicalTrials.gov. Date of the latest search: 20 August 2021. Selection criteria: Randomized and quasi-randomized controlled clinical studies of ERT with galsulfase compared to other interventions or placebo. Data collection and analysis: Two authors independently screened the studies, assessed the risk of bias, extracted data and assessed the certainty of the the evidence using the GRADE criteria. Main results: One study was included involving 39 participants who received either ERT with galsulfase (recombinant human arylsulphatase B) or placebo. This small study was considered overall to have an unclear risk of bias in relation to the design and implementation of the study, since the authors did not report how both the allocation generation and concealment were performed. Given the very low certainty of the evidence, we are uncertain whether at 24 weeks there was a difference between groups in relation to the 12-minute walk test, mean difference (MD) of 92.00 meters (95% confidence interval (CI) 11.00 to 172.00), or the three-minute stair climb, MD 5.70 (95% CI -0.10 to 11.50). In relation to respiratory tests, we are uncertain whether galsulfase makes any difference as compared to placebo in forced vital capacity in litres (FVC (L) (absolute change in baseline), given the very low certainty of the evidence. Cardiac function was not reported in the included study. We found that galsulfase, as compared to placebo, may decrease urinary glycosaminoglycan levels at 24 weeks, MD -227.00 (95% CI -264.00 to -190.00) (low-certainty evidence). We are uncertain whether there are differences between the galsulfase and placebo groups in relation to adverse events (very low-certainty evidence). In general, the dose of galsulfase was well tolerated and there were no differences between groups. These events include drug-related adverse events, serious and severe adverse events, those during infusion, drug-related adverse events during infusion, and deaths. More infusion-related reactions were observed in the galsulfase group and were managed with interruption or slowing of infusion rate or administration of antihistamines or corticosteroids drugs. No deaths occurred during the study. AUTHORS' CONCLUSIONS: The results of this review are based only on one small study (a 24-week randomised phase of the study and prior to the open-label extension). We are uncertain whether galsulfase is more effective than placebo, for treating people with MPS VI, in relation to the 12-minute walk test or the three-minute stair climb, as the certainty of the evidence has been assessed as very low. We found that galsulfase may reduce urinary glycosaminoglycans levels. We are also uncertain whether there are any differences between treatment groups in relation to cardiac or pulmonary functions, liver or spleen volume, overnight apnea-hypopnea, height and weight, quality of life and adverse effects. Further studies are needed to obtain more information on the long-term effectiveness and safety of ERT with galsulfase.
Article
Achondroplasia is a unique model of the effects of skeletal dysplasia and dwarfism on the respiratory system. We measured chest dimensions, spirometry, lung volumes, maximal expiratory flow volume curves, nasal and airways resistance, closing volume, maximal inspiratory/expiratory pressures, and tracheal area by acoustic reflection in 12 healthy subjects with achondroplasia. Anterior-posterior thoracic diameter was mildly reduced in men. Vital capacity for all subjects was 108 percent +/- 18.6 percent (SD) of that predicted for achondroplastic subjects, but was reduced when compared with values for people of average stature that were predicted, based on either sitting height or thoracic height. The reduction was relatively greater in male than in female subjects. The RV/TLC and FRC/TLC ratios were normal. Other measurements were similar to those in average-statured adults. We conclude that achondroplasia results in a reduction in vital capacity out of proportion to what would be expected if these subjects had normal limb size. Although the lungs may be small, they are functionally normal, as are the airways.
Article
Maximal voluntary ventilation (MVV) may be determined directly by the sprint method or calculated from pulmonary function data, using the functions MVV = forced expired volume in 1 sec (FEV1) × 35 or MVV = FEV1 × 40. The purpose of this paper was to test the validity of the equation over a wide range of lung function in children. Cystic fibrosis (CF), a chronic lung disease where children typically have a wide range of pulmonary function, was chosen as the study requirement. Spirometric data from 332 children with CF who underwent pulmonary function testing between 1987–2000 were stratified according to disease severity, and box-plots comparing the ratio of MVV to FEV1 for each category were generated. As results indicated that the equation underestimates true MVV proportionally to the degree of airflow limitation, a new function to predict MVV for this population was derived and tested. The new equation was derived using data from patients who were tested on odd-numbered days (group A). The validity of the new equation was then tested on the patients tested on even-numbered days (group B). To test its validity, the results were compared to the “gold standard” sprint values using a Bland and Altman plot. MVV was expressed as a function of FEV1 and predicted FEV1: MVV = 27.7(FEV1) + 8.8(PredFEV1) (R2 = 0.98, P < 0.05). In this way, the accuracy of the new equation was confirmed. Whenever possible, we recommend MVV be determined by the sprint method in accordance with ATS guidelines. If this is not feasible, we recommend considering the new prediction equation. Pediatr Pulmonol. 2003; 35:467–471. © 2003 Wiley-Liss, Inc.
Article
Our goal was to evaluate the long-term safety and efficacy of recombinant human alpha-l-iduronidase (laronidase) in patients with mucopolysaccharidosis I. All 45 patients who completed a 26-week, double-blind, placebo-controlled trial of laronidase were enrolled in a 3.5-year open-label extension study. Mean patient age at baseline was 16 (range: 6-43) years. All patients had attenuated disease (84% Hurler-Scheie, 16% Scheie phenotypes). Clinical, biochemical, and health outcomes measures were evaluated through the extension phase. Changes are presented as the mean +/- SEM. All 40 patients (89%) who completed the trial received at least 80% of scheduled infusions. As shown in earlier trials, urinary glycosaminoglycan levels decreased within the first 12 weeks and liver volume decreased within the first year. Percent predicted forced vital capacity remained stable, with a linear slope of -0.78 percentage points per year. The 6-minute walk distance increased 31.7 +/- 10.2 m in the first 2 years, with a final gain of 17.1 +/- 16.8 m. Improvements in the apnea/hypopnea index (decrease of 7.6 +/- 4.5 events per hour among the patients with significant baseline sleep apnea) and shoulder flexion (increase of 17.4 degrees +/- 3.6 degrees) were most rapid during the first 2 years. Improvements in the Child Health Assessment Questionnaire/Health Assessment Questionnaire disability index (decrease of 0.31 +/- 0.11, signifying a clinically meaningful improvement in activities of daily living) were gradual and sustained over the treatment period. Laronidase infusions were generally well tolerated except in 1 patient who experienced an anaphylactic reaction. Infusion-associated reactions, which occurred in 53% of the patients, were mostly mild, easily managed, and decreased markedly after 6 months. One patient died as a result of an upper respiratory infection unrelated to treatment. Antibodies to laronidase developed in 93% of the patients; 29% of the patients were seronegative at their last assessment. This trial demonstrates the long-term clinical benefit and safety of laronidase in attenuated patients with mucopolysaccharidosis I and highlights the magnitude and chronology of treatment effects. Prompt diagnosis and early treatment will maximize treatment outcomes.