Download as pdf or txt
Download as pdf or txt
You are on page 1of 47

ARTICLE IN PRESS

Mucopolysaccharidosis type II
(Hunter syndrome): Clinical and
biochemical aspects of the disease
and approaches to its diagnosis
and treatment
Shifaza Mohamed, Qi Qi He, Arti A. Singh, Vito Ferro*
School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
*Corresponding author: e-mail address: v.ferro@uq.edu.au

Contents
1. Introduction 2
1.1 Lysosomal storage diseases (LSDs) 2
1.2 Mucopolysaccharidoses (MPS) and glycosaminoglycans (GAGs) 3
2. Mucopolysaccharidosis type II (MPS II) 4
2.1 History and incidence of MPS II 4
2.2 Genetics of MPS II 8
2.3 Clinical aspects of MPS II 8
3. Biochemical basis of disease 13
3.1 Iduronate-2-sulfatase (IDS): Structure, substrate specificity, and enzyme
mechanism 13
3.2 Mutational analysis of IDS in MPS II 16
4. Diagnostic methods for MPS II 18
4.1 Overview 18
4.2 Development of IDS enzyme activity assays 19
4.3 Assays based on biomarkers of MPS II 27
5. Management and treatment of MPS II 30
5.1 Overview 30
5.2 Enzyme replacement therapy 30
5.3 Substrate reduction therapy 32
5.4 Pharmacological chaperone therapy 33
5.5 Other treatments 34
6. Conclusions 35
Acknowledgments 36
References 37

Advances in Carbohydrate Chemistry and Biochemistry # 2019 Elsevier Inc. 1


ISSN 0065-2318 All rights reserved.
https://doi.org/10.1016/bs.accb.2019.09.001
ARTICLE IN PRESS

2 Shifaza Mohamed et al.

1. Introduction
1.1 Lysosomal storage diseases (LSDs)
Lysosomes are the organelles responsible for the intracellular degradation of
macromolecules such as oligosaccharides, glycolipids, proteins and glyco-
proteins; their optimal functioning is crucial for cellular homeostasis. Lyso-
somal storage diseases (LSDs) are hereditary disorders caused by specific
mutations in genes encoding lysosomal enzymes, with approximately 50 dif-
ferent LSDs currently known.1 In several cases, the mutated enzymes are
catalytically competent but are misfolded and are thus more rapidly degraded
by cellular proteases within the endoplasmic reticulum-associated degrada-
tion machinery, leading to reduced lysosomal trafficking. This results in the
progressive intra-lysosomal accumulation of undegraded or partially
degraded substrate(s) of the defective enzymes, giving rise to severe clinical
phenotypes that ultimately lead to cell and tissue damage. LSDs are generally
classified according to the major storage material(s) or the nature of the
underlying lysosomal defect(s) (Table 1).1–4

Table 1 General classification of LSDs.


Category Major storage material(s)
Mucopolysaccharidoses Mucopolysaccharides or glycosaminoglycans (GAGs),
(MPS) oligosaccharides
Glycoproteinoses Glycopeptides, oligosaccharides
Oligosaccharidoses Oligosaccharides
Sphingolipidoses Sphingolipids, glycolipids
Mucolipidoses Lipids, oligosaccharides, GAGs
Lipidoses Lipids
Glycogenoses Glycogen
Neuronal ceroid Lipofuscin
lipofuscinoses
Lysosomal membrane protein disorders
Lysosomal transport defects
Multiple enzyme deficiencies
Other lysosomal protein disorders
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 3

The majority of LSDs are inherited in an autosomal recessive manner and


are rare genetic diseases individually, prevailing in the range of one per 4.2
million to one per 60,000 live births.5 However, the total prevalence of
LSDs is quite significant and has been estimated to be as high as one per
7700 live births, depending on factors such as geographic location and eth-
nicity.6,7 The current worldwide estimate is approximately one per 5000
live births.1 Signs and symptoms of LSDs vary greatly from very severe infan-
tile variants to attenuated variants in adolescence and adulthood. The various
types of LSDs and their impact on cell biology is well reviewed by Boustany1
and Vellodi.2
Classically, the treatments for LSDs have been limited and focused on
symptomatic care of disease manifestations. However, recent decades have
seen the development and continual improvement of therapies such as hema-
topoietic stem-cell transplantation and enzyme-replacement therapy.1,2,5,8–14
The multisystemic nature of LSDs require healthcare professionals of different
expertise, such as cardiologists, dermatologists, pediatricians, nephrologists
and general practitioners, to be involved in providing medical assistance for
patients throughout their lifetime. Advances in these fields have led to signif-
icant clinical improvement and enhanced quality of life for patients suffering
from various LSDs.

1.2 Mucopolysaccharidoses (MPS) and


glycosaminoglycans (GAGs)
Mucopolysaccharidoses (MPS) are LSDs dominated by glycosaminoglycan
(GAG) storage15 and have been reported to affect between one in 19,000
and one in 43,000 individuals, depending on the geographic region.16 GAGs,
also known as mucopolysaccharides, are linear heteropolysaccharides com-
posed of characteristic repeating disaccharide units with a typical unit length
of 50–150 disaccharide units and extensive modification by N-acetylation,
N-sulfation, O-sulfation and epimerization.17,18 These modifications result
in a large variety of structurally diverse and information-dense biological
molecules; it is thus unsurprising that defects in GAG degradation result
in complex multisystem diseases. Advances in biochemistry and genetics
over the past six decades have resulted in the identification of the key
enzymes underlying the MPSs, paving the way for the isolation and char-
acterization of the genes involved. The diverse history of these disorders is
well reviewed.19–21
GAGs are categorized into subtypes depending on the individual disac-
charide units that comprise them. These repeating units are composed of a
ARTICLE IN PRESS

4 Shifaza Mohamed et al.

uronic acid residue, either D-glucuronic acid or L-iduronic acid (IdoA) and
an amino sugar, either D-glucosamine or D-galactosamine.17,22 There are
four classes of sulfated GAGs: heparan sulfate (HS), dermatan sulfate (DS),
chondroitin sulfate (CS) and keratan sulfate (KS). Hyaluronic acid, or
hyaluronan, is a unique non-sulfated GAG.17,22
GAGs destined for lysosomal degradation travel through the endosomal
network after endocytosis, following which GAG degradation occurs
through enzyme-mediated exoglycosidic cleavage of specific terminal sugars
and desulfation of sulfated residues (Figs. 1 and 2). The resulting monosac-
charides and inorganic sulfate generated from complete degradation are then
actively transported out of the lysosome. In the case of MPS-affected indi-
viduals, different substrates are accumulated in their lysosomes depending on
the specific enzyme deficiency.
Eleven known lysosomal deficiencies result in seven distinct forms of
MPS (Table 2).5,22–24 With the exception of MPS IIIC, which is caused
by the deficiency of a transferase, these disorders are caused by deficiency
in one of the glycosidase or sulfatase enzymes.
Some of the physiological processes that may be affected by defective
GAG degradation and GAG-filled lysosomes include additional lysosomal
storage by interference of lysosomal hydrolases; altered plasma-membrane
receptor assembly; altered sequestration of growth factors; altered sequestra-
tion, recruitment and presentation to signaling receptors of cytokines;
abnormal extracellular matrix crosslinks; altered cell attachment and inter-
ference with cellular trafficking; and macrophage dysfunction.22 Each
MPS disease is characterized by progressive multisystem involvement with
key morbid manifestations involving the skeleton, joints, somatic tissues,
heart and, in some disorders, the central nervous system (CNS).22–26 Fur-
thermore, each of the disorders is characterized by a spectrum of disease
manifestations and clinical severity, which ranges from early onset with rapid
progression to attenuated forms with later onset and slow progression. The
disease spectrum seen in MPSs can be attributed to the differences in residual
enzyme activity.

2. Mucopolysaccharidosis type II (MPS II)


2.1 History and incidence of MPS II
Mucopolysaccharidosis type II (MPS II), also known as Hunter syndrome, is
a rare X-linked, recessively-inherited LSD caused by mutations of the gene
encoding the lysosomal enzyme iduronate-2-sulfatase (IDS).27,28 Affecting
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 5

Fig. 1 Degradation of heparan sulfate (HS). Large HS chains are first degraded
into smaller fragments by an endo-β-glucuronidase (heparanase), followed by a
well-ordered sequential degradation, one monosaccharide unit at a time from the
non-reducing end. Reproduced with permission from Varki, A., et al., Eds. Essentials
of Glycobiology, 2nd ed.; Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press,
2009 by The Consortium of Glycobiology Editors, La Jolla, California.

mostly males, its incidence has been reported to range from one per 500,000
to one per 92,000 live births, depending on the population studied.6,16,27
The disease that now bears his name was first documented by Charles
Hunter in 1917,29 when he described its physical features in two brothers.
ARTICLE IN PRESS

6 Shifaza Mohamed et al.

Fig. 2 Degradation of dermatan sulfate (DS) and chondroitin sulfate (CS). Reproduced
with permission from Varki, A., et al., Eds. Essentials of Glycobiology, 2nd ed.; Cold Spring
Harbor (NY): Cold Spring Harbor Laboratory Press, 2009 by The Consortium of Glycobiology
Editors, La Jolla, California.

Hunter described the brothers as being undersized with large heads,


although of normal intelligence, and having dysmorphic facial features,
protruding abdomens and a limited range of motion in all joints of the
extremities. Hearing was impaired in both brothers, and their respiration
was described as being noisy, becoming labored and uneasy during sleep
that generally featured heavy snoring. One of the boys also had an enlarged
heart with clear diastolic and systolic murmurs.29 In 1920, pediatrician
Gertrud Hurler30 reported on two unrelated boys who showed similar
manifestations accompanied by corneal clouding, spinal deformities, and
mental impairment, in what was the first report of the related but now
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 7

Table 2 Classification of MPS disorders.5,22–24


Primary
Gene storage
Disorder Deficient enzyme location material
MPS I (Hurler/Scheie α-L-Iduronidase 4p16.3 DS, HS
syndrome)
MPS II (Hunter Iduronate-2-sulfatase Xq28 DS, HS
syndrome) (IDS)
MPS IIIA (Sanfilippo-A Heparan-N-sulfatase 17q25.3 HS
syndrome)
MPS IIIB (Sanfilippo-B N-Acetyl-α-glucosaminidase 17q21 HS
syndrome)
MPS IIIC (Sanfilippo-C Acetyl-CoA:α-glucosaminide 8p11.1 HS
syndrome) N-acetyltransferase
MPS IIID (Sanfilippo-D N-Acetylglucosamine 12q14 HS
syndrome) 6-sulfatase
MPS IVA (Morquio-A N-Acetylgalactosamine- 16q24.3 CS, KS
syndrome) 6-sulfatase
MPS IVB (Morquio-B β-Galactosidase 3p21.33 KS
syndrome)
MPS VI (Maroteaux- N-Acetylgalactosamine- 5q11-q13 CS, DS
Lamy syndrome) 4-sulfatase
MPS VII (Sly syndrome) β-D-Glucuronidase 7q21 CS, DS, HS
MPS IX (Natowicz Hyaluronidase I 3p21 Hyaluronan
disease)

known to be distinct disease MPS I (Hurler syndrome). MPS I and MPS II


were initially considered to be one disease, known by various names includ-
ing Hunter–Hurler syndrome,31 until the biochemical basis of each was
determined decades later.
The term “mucopolysaccharidosis” was first coined in 1952, when Brante32
identified chondroitin sulfate-like material from the livers of MPS I patients.
This was followed in 1957 by Dorfman and Lorincz’s33 demonstration of the
excess urinary excretion of DS and HS in another MPS I patient. This excess
GAG accumulation was originally thought to be the result of overproduction,
but it was demonstrated in 1968 by Fratantoni et al.34 that the excess was due to
ARTICLE IN PRESS

8 Shifaza Mohamed et al.

reduced degradation rather than excess production or decreased cellular


secretion, in both MPS I and II. Further investigations by the Neufeld
group35–37 showed that the metabolic defects in fibroblasts cultured from
MPS II patients could be corrected by a fibroblast-secreted factor from
MPS I patients, and vice versa. This significant discovery led to the isolation
of the “Hunter corrective factor” from normal human urine38 and its sub-
sequent identification as a protein, initially named sulfoiduronate sulfatase39
and now known as IDS.40

2.2 Genetics of MPS II


MPS II is an X-linked, recessively inherited disease. The IDS gene located
on chromosome Xq28 consists of nine exons spread over 24 kb, encoding
the 550-amino-acid IDS protein.40–43 The wide spectrum of clinical man-
ifestations of MPS II can be associated with the high level of molecular het-
erogeneity at the IDS gene locus. According to the Human Gene Mutation
Database,44,45 more than 640 different IDS mutations have been reported to
March 2019, including point mutations, small insertions, deletion, missense
or nonsense mutations, and major structural alterations, such as complex
rearrangements and gross insertions or deletions.46–57 In general, gross
structural changes are associated with the severe phenotype of the disease,
whereas small gene mutations may result in a moderate, attenuated
phenotype.
Although MPS II is an X-linked recessive disorder, it has been observed
in a small number of females.58–60 Phenotypic expression of MPS II in
females may be the result of homozygosity for disease causing mutations,
monosomy or structural abnormalities of the X-chromosome, or skewed
X-chromosome inactivation.58–60 Most females with the disorder inherit
the IDS gene mutation from the mother with preferential inactivation of
the non-mutant paternal alleles. The clinical course in female patients is sim-
ilar to the male clinical phenotype.48,58

2.3 Clinical aspects of MPS II


2.3.1 Overview
The loss of IDS enzyme activity in MPS II leads to progressive lysosomal
storage of undegraded HS and DS in tissues and organs such as the liver,
spleen, heart, bone, joints, and airways. These materials disturb cellular func-
tion through several activities including the activation of signal transduction
by non-physiological substances, alteration of intracellular calcium
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 9

homeostasis, impairment of autophagy, and inflammation.1–3 Conse-


quently, this leads to the characteristic multisystemic disease manifestations
of MPS II, with involvement of the musculoskeletal, respiratory and nervous
systems, among others.61,62
The phenotypic expression of MPS II displays variable rates of onset
and progression spanning a wide spectrum of clinical severity, typically
involving some level of skeletal and joint deformity, cardiopulmonary
and respiratory compromise, and neurodegeneration.61,62 Two clinical
forms of MPS II have historically been recognized, “attenuated” (formerly
“mild”) and “severe,” which were clinically distinguished by the involve-
ment of the central nervous system (CNS) and depend on the level of
IDS enzyme deficiency.62 However, as both forms significantly impact
quality of life,63 it is more appropriate to describe MPS II as a heteroge-
neous disorder with a broad spectrum of clinical features. The terms
“slowly progressive” and “early progressive” have been suggested to better
reflect the phenotypic continuum rather than “attenuated” and “severe,”
respectively.61
Patients with the severe, or early progressive form of the disorder suffer
from progressive neurological involvement resulting in severe mental
impairment, with the onset of symptoms by 2–4 years of age.62–66 In these
cases, death occurs within the first two decades of life, typically by
10–15 years of age.64,65 Patients with the attenuated, or slowly progressive
form do not show significant cognitive involvement and tend to have nor-
mal intelligence.62–64,66–68 They also typically display a slightly later onset of
clinical signs and symptoms: a 1982 study of 55 male MPS II patients indi-
cated a mean age of onset of 4.30 years for the attenuated form (n ¼ 16) vs
2.47 years for the severe form (n ¼ 39).67 While the disease is still chronic,
these patients can often survive into adulthood, sometimes even into and
beyond their 50s.27,64,67,68

2.3.2 Development
Infants with MPS II appear normal at birth and may achieve early develop-
mental milestones, even in the presence of acute somatic disease. Develop-
mental delays generally become apparent by 18–24 months of age, with very
slow progress thereafter; most patients hit a developmental plateau between
3 and 5 years of age.27 The disease has been found to considerably impact
both the physical and psychological aspects of the quality of life of both
patients and their family members,63,69 with some affected children dis-
playing aggression and hyperactivity, and teenagers and young adults
ARTICLE IN PRESS

10 Shifaza Mohamed et al.

suffering from significant psychosocial problems.70 In the most severe cases,


patients are severely mentally handicapped and completely dependent on
caregivers by the time of death, generally in their second decade.27,64

2.3.3 General appearance and skeletal abnormalities


Regardless of the clinical severity of the disorder, the appearance and skeletal
abnormalities of MPS II patients are quite similar. Patients generally appear
normal at birth with coarsening of facial features becoming apparent by
2–4 years of age,27 most likely due to a combination of bone dysostosis
and GAG storage in the soft tissues of the orofacial region. In addition,
patients tend to be of less than average height throughout life and have a large
head, broad noses with flared nostrils, prominent supraorbital ridges, large
jowls, thickened ear lobules and lips, and an enlarged, protruding tongue.27
Collectively known as dysostosis multiplex, the skeletal deformities of
the MPS disorders were originally described by Hunter in his first report
on MPS II.29 The skeletal deformities of MPS II patients include abnormal
thickness of all bones, irregular epiphyseal ossification in the hand, shoulder
and elbow joints, which results in joint stiffness and a claw-like appearance of
the hands, enlarged clavicles, thickened and misshapen ribs, and vertebral
irregularities.27,29,62,71 These skeletal deformities result in profound loss of
joint motion and adversely affect patients’ quality of life. Furthermore,
patients are at an increased risk of carpal tunnel syndrome,72 which in com-
bination with claw hands can lead to the loss of hand function, and they often
walk on their toes because of tight heel cords and joint stiffness,62 which
gives them an unsteady gait. Most patients also suffer from progressive
arthropathy that especially affects the hip joints,71 with many patients
becoming wheelchair-bound because of the resulting pain.

2.3.4 Eyes and vision


The ocular system is not largely affected in MPS II, but retinal degeneration
has been demonstrated in some patients who reported some loss of
vision.73–76 Corneal clouding may also be present to some degree but is
not itself a major feature of MPS II.75 Thickening of the sclera due to
GAG accumulation may also cause compression of the optic nerve,74,75
although this is relatively less common in MPS II than in some of the other
MPS disorders. Bilateral, asymmetric, vitreous bodies have also been
reported in a young MPS II patient who developed maculopathy.77 Loss
of vision is under-appreciated in MPS II and it is suggested that this be
screened for yearly.62,78
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 11

2.3.5 Ears and hearing


Recurrent ear infection and progressive hearing loss is common in MPS II
patients, often from within the first year of life, and correlates with the sever-
ity of somatic disease.27,62,78 Hearing loss can be attributed to both conduc-
tive and sensorineural deficits,65,68 including dysostosis of the middle ear
ossicles, Eustachian tube dysfunction, and damage to the vestibulocochlear
nerve and tympanic membrane.27 In addition, the middle ear mucosa tends
to be thick and edematous, with the cells being large and foamy due to GAG
storage.78 Regular 6- to 12-monthly audiologic and otologic screening is
recommended78 to aid in the provision of appropriate auditory aids to
patients,79–81 and to assist in preventing hearing loss-related learning diffi-
culties and behavioral problems.

2.3.6 Mouth and throat


Speech delays and swallowing disorders are common in MPS II patients due
to the structural and musculoskeletal changes to the jaw and throat.78 These
problems may potentially be intensified by neural involvement and cogni-
tive impairment in severe disease. Poor jaw mobility limits the patient’s abil-
ity to open the mouth for chewing and speaking, and factors such as an
enlarged tongue, and potentially enlarged tonsils and adenoids can interfere
with swallowing.78 Chewing can also be affected by dental abnormalities,
including delayed tooth eruption, widely-spaced and peg-shaped teeth,
and enlarged gums due to gingival hypertrophy and hyperplasia.82,83 Rou-
tine dental procedures may be difficult due to the poor jaw mobility and may
require general rather than local anesthetic,78 although this itself can pose
greater risks for patients due to their anatomical predisposition to airway
obstruction and compromise.

2.3.7 Respiratory and upper airway manifestations


A number of anatomical features predispose MPS II patients to airway com-
promise, which is a major contributor to their morbidity and mortality.62,78
These include an enlarged tongue, epiglottis, tonsils and adenoids, narrow
and abnormally-shaped trachea and bronchi with flaccid supporting carti-
lage, thick tracheal and nasal secretions, thickened vocal cords, redundant
tissue in the upper airway that is prone to swelling, a small chest cavity
with stiff joints and chest wall, and thickened and misshapen ribs.62,78,84–87
These factors can lead to complications such as labored and noisy breathing,
frequent pneumonia and upper respiratory infections, collapse of the
ARTICLE IN PRESS

12 Shifaza Mohamed et al.

trachea, pharynx and bronchi, and acute airway obstruction.62,78,84–88


Airway obstruction is particularly likely to occur during mechanical stimu-
lation, such as during tracheal intubation for general anesthesia. Obstructive
sleep apnea87,89,90 is common in later stages of the disease and can lead to
behavioral problems due to a lack of restful sleep.

2.3.8 Gastrointestinal involvement


GAG storage often leads to enlarged livers and spleens in MPS II patients.
Several studies have reported a prevalence of between 58% and 90% for
either organ being enlarged in their participants.65,68,91–94 Abdominal dis-
tention, umbilical hernias (reported in 76% and 95% of male participants
in two studies),65,68 and inguinal hernias (reported in approximately 60%
of male participants in both studies)65,68 are thus commonly observed,
although hepatic or splenic dysfunction is generally not. Chronic diarrhea
is commonly observed in patients with neurological involvement;65–67
however, this is uncommon in patients with the attenuated disease
phenotype.

2.3.9 Cardiac involvement


Cardiac involvement has been described in all forms of MPS,95 with almost
all MPS II patients suffering from some form of cardiac disease. Signs and
symptoms may present as early as 5 years of age.68 Cardiac disease is progres-
sive in MPS II and is thought to be a major cause of death.96–99 Valvular
involvement is commonly reported, with thickening and stiffening of valve
leaflets often resulting in ventricular hypertrophy and heart failure.95,100 For
example, a study of 27 male MPS II patients aged between 2 and 11 years
revealed morphological changes in the mitral and aortic valves in 19 and 5
patients, respectively.97 Clinical signs of heart disease were detected in
10 patients, with only 5 having echocardiogram results considered normal.97
Another such study found that 11 of 18 patients had abnormal mitral
valves,100 and the 2008 Hunter Outcome Survey98 found 57% of the 202
surveyed MPS II patients with available cardiac data to have cardiac valve
disease and 6% to suffer from hypertension. Nodular thickening of the valves
has been observed from autopsy examinations,95,100,101 and storage material
has been detected within the valves and the myocardium through histolog-
ical and electron-micrographic examination.102 Valve replacement has been
reported but is uncommon,95,100 while hypertension is under-appreciated in
MPS II and should be treated as necessary.78
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 13

2.3.10 Neurological involvement


Neurological involvement is more commonly seen in patients with the
severe form of MPS II and is initially suspected when patients fail to meet
developmental milestones, such as the abilities to sit unsupported, walk
and speak.65,68 Severe CNS involvement results in profound and progressive
mental impairment, with developmental regression reported to begin
between 6 and 8 years of age.65 Behavioral problems, such as aggression,
hyperactivity and obstinacy are commonly observed in severely affected
patients by the age of 2 years and have been reported to continue until lim-
ited by the onset of neurodegeneration, generally at around age eight or
nine.67,70,103 Progressive compression of the spinal cord with resulting cer-
vical myelopathy is also common in these patients. Caused by dural thick-
ening or by instability of the atlantoaxial joint, this leads to symptoms such as
muscle weakness, abnormal gait, bladder dysfunction, and clumsiness with
fine motor skills.104–106 Communicating hydrocephalus, which is a distur-
bance in the reabsorption of cerebrospinal fluid (CSF), may also contribute
to neurological deterioration in MPS II. Symptoms may include behavioral
changes, headaches, and vision disturbances.107,108 Seizures are also fre-
quently observed in severely affected patients but are much less common
in patients with the attenuated form of the disease. The frequency of seizures
reportedly increases in parallel with cognitive deterioration.65,68 A 2009
study of historical data by Jones et al.64 stated that none of the studied
patients with reported cognitive involvement survived beyond 23 years of
age. In contrast, some of the attenuated patients reportedly survived into
their 40s, highlighting the significant impact of neurological involvement
on lifespan in MPS II.

3. Biochemical basis of disease


3.1 Iduronate-2-sulfatase (IDS): Structure, substrate
specificity, and enzyme mechanism
As part of lysosomal GAG degradation, the biological function of IDS is to
hydrolyze O-linked sulfate groups at the C-2 position of the terminal non-
reducing end of L-iduronic acid (IdoA) residues in dermatan sulfate (DS) and
heparan sulfate (HS) (Fig. 3).27,28
IDS belongs to the sulfatase family of enzymes and is responsible for cat-
alyzing the hydrolytic desulfonation of sulfate esters and sulfamates.109 Sul-
fatases are a heterogeneous group of enzymes that have been found to have
remarkable similarities, including: (a) 20%–60% sequence homology over
ARTICLE IN PRESS

14 Shifaza Mohamed et al.

Fig. 3 IDS cleaves the C-2 sulfo group of terminal IdoA residues in HS (top) and DS (bottom).

the entire protein length, (b) a highly conserved N-terminal region con-
taining the sulfatase motif, and (c) a unique active-site aldehyde residue,
α-formylglycine (FGly).110 The IDS enzyme has recently been crystallized
and its X-ray crystal structure solved to 2.3Å resolution110a; and some
homology models have also been constructed (see below). Various studies
have been conducted on the topography of the active site,111 as well as
on enzyme kinetics and physical properties of the enzyme.112 The catalytic
pocket of the enzyme is believed to contain a divalent metal cation and to be
lined with several positively charged residues that facilitate the anionic sul-
fate binding, with the FGly residue resting at the bottom of this pocket as the
aldehyde hydrate. The FGly residue is essential for IDS catalytic activity and
is generated post-translationally by oxidation of a cysteine precursor found
in the encoded peptide sequence of the enzyme. The minimum consensus
motif that allows the conversion of cysteine to FGly is formed by 11 amino
acids that form the core motif C(X/T)P(X/S)R, which is highly conserved
among most sulfatases from all species.113,114 It has been shown that the oxi-
dation event occurs during later co- or post-translation, after translocation to
the endoplasmic reticulum and before protein folding. Mutations in the
active site or in the flanking regions lead to decreased substrate affinity or
enzyme stability.51
Fig. 4 shows the catalytic mechanism for the cleavage of the sulfate ester
bond of the terminal IdoA residues of HS and DS by IDS. The aldehyde
hydrate catalytic form of the FGly residue in the active site is formed by
the addition of a water molecule to the formyl group. Transesterification
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 15

Fig. 4 Mechanism of sulfate ester cleavage by sulfatases such as IDS.115

of the sulfate group of the substrate on to the enzyme then results in an FGly
sulfate adduct (left). Subsequent elimination of the sulfate group by the reac-
tion of the second geminal hydroxyl group of the intermediate and the
cleavage of the C–O bond regenerates the aldehyde (top).115
In 1990, Hopwood and co-workers substantially purified IDS more than
500,000-fold in 5% yield from human liver, lung, kidney and placenta, and
identified two major forms of IDS, form A and form B.112 Form A and
form B had almost the same recovered enzyme activities toward a series
of substrates derived from heparin, HS and DS, but with different molecular
masses of 42 kDa and 14 kDa, respectively. Kinetic parameters of form A
were determined with a variety of substrates derived from HS, heparin
and DS. It was found that the structure of the residue next to the non-
reducing end of the IdoA-2-sulfate (IdoA2S) residue significantly affected
the IDS enzyme’s catalytic efficiency and binding affinity. Heparin-derived
tetrasaccharide substrates with a 6-O-sulfo group on the adjacent glucos-
amine residue were hydrolyzed with up to 200  greater efficiency than
those without sulfation at this position. A GlcNS substituent also increased
the binding affinity by up to twofold compared with GlcNAc or GlcNH.
The simplest disaccharide substrate tested for IDS activity was
IdoA2S-anM (1, Fig. 5). The related substrate IdoA2S-anM6S (2) with
an additional sulfate on the anhydromannitol residue was hydrolyzed by
IDS with a 63-fold increase in catalytic efficiency and a fivefold increase
in binding affinity.114 There are only a few known competitive inhibitors
of IDS (see also Section 5.4), including compounds derived from degrada-
tion of heparin (3–7) and ascorbic acid sulfate 8.111,112 The most potent
ARTICLE IN PRESS

16 Shifaza Mohamed et al.

Fig. 5 Structures of substrates (1–2) and inhibitors (3–8) of IDS with Ki values at pH 4.5.

inhibitor to date is 2,5-anhydromannitol-1-sulfate (7) with Ki ¼ 0.25 μM at


pH 4.5.

3.2 Mutational analysis of IDS in MPS II


Mutational analysis of the IDS gene is potentially a useful tool for the diagnosis
of MPS II with regard to carrier and prenatal diagnosis, and may allow for
testing of IDS mutations present in families. These variants potentially have
considerable impact on the protein structure, protein processing and traffick-
ing, and enzyme–substrate interactions. In general, gross structural changes are
associated with the severe phenotype of the disease whereas small gene muta-
tions may result in a moderate to mild phenotype.48,49,51,52,54,57,116 Before the
crystal structure of IDS was available, mutations of the IDS gene and their
effects on protein structure and enzyme activity were extensively studied in
an attempt to establish the genotype–phenotype correlations.45–47,49,51–57,116
In 2003, Kim et al.49 studied the mutations of the IDS gene in 25 unrelated
Korean MPS II patients using a three-dimensional model of the IDS protein
developed based on the known X-ray structures of several lysosomal sulfatase
enzymes, including arylsulfatase A and N-acetylgalactosamine-4-sulfatase.
From this model, 20 mutations of the IDS gene were identified. Thirteen
of these mutations were novel, including six small deletions, two insertions,
two nonsense and three missense mutations.49 It was suggested that most of
these mutations were likely to affect the refolding of the protein in terms
of protein structure, and that the nonsense and missense mutations potentially
interfered with the IDS enzyme function.49 However, the molecular mecha-
nisms of these genotype–phenotype relationships were not elucidated. Chkioua
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 17

et al.54 utilized the same homology model to investigate IDS mutations and
polymorphisms in a Tunisian MPS II-affected family.
Kato et al. (2005)51 then investigated the genotype–phenotype relation-
ships of IDS gene mutations in 18 MPS II patients in Japan by carrying out a
mutational and structural analysis of the IDS gene using an IDS homology
model created based on the structure of arylsulfatase A. A total of 16 muta-
tions were identified, with seven of these being novel.51 Mutations identi-
fied from the severe clinical phenotype included four missense mutations
and one nonsense mutation. Mutations from the attenuated phenotype
included four missense mutations, two nonsense mutations, three frame shift
mutations, one splice site mutation, and one amino acid deletion. IDS muta-
tions in severely affected individuals had significant influences on the tertiary
structure of the IDS protein, resulting in loss of IDS enzyme activity,51 while
most mutations in the attenuated patients only partially affected the protein
structure with preservation of residual enzyme activity, leading to the
observed milder clinical phenotype.51
Further investigation of genotype–phenotype correlations by Sukegawa-
Hayasaka et al.52 in 2006 focused on 11 common IDS missense mutations
identified in MPS II patients. Mutant IDS proteins with these mutations
were produced in CHO cells, and enzyme activity, protein processing,
and structural analyses were conducted using an engineered reference
IDS protein and a homology model based on the human arylsulfatases
A and B.52 Mutant proteins in the attenuated MPS II phenotype were found
to have residual enzyme activity (0.2%–2.5% of wildtype), while mutants in
the severe clinical phenotype had none.52 In addition, the mutations con-
siderably changed the structural conformation of the IDS protein, leading
to its degradation and/or insufficiency in protein processing.52 These find-
ings about the distribution of IDS mutations and their effects on the pro-
tein structure have considerable impacts on the selection of the available
therapies for MPS II patients, as well as for the design and development of
new MPS II treatments (see Section 5.4). Saenz et al.116 have also reported
the construction of a homology model of IDS based on arylsulfatases
A and B.
Galvis et al. carried out protein modeling and molecular docking simu-
lations on wildtype and mutant forms of IDS to investigate the effects of
mutations on enzyme structure and functions, and to identify correlations
between genotype and phenotype among Colombian MPS II patients.57
In this study, a homology model was constructed using arylsulfatase A as
the template. Molecular docking was carried out on IDS mutants with
ARTICLE IN PRESS

18 Shifaza Mohamed et al.

HS and DS ligands, although it was not clear what size fragments were used.
These studies identified putative active site residues and showed that muta-
tions affected protein conformation and substrate–protein interactions,
suggesting implications for protein stability, processing and trafficking to
lysosomes.57

4. Diagnostic methods for MPS II


4.1 Overview
Timely diagnosis is the key to improving outcomes for MPS II patients, and
diagnosis involves the examination of different clinical factors, biochemical
parameters, and molecular characterization. However, the signs and symp-
toms of MPS II are not specific to the disorder and not all symptoms will be
present in all patients. Patients typically have normal appearance at birth, and
will only begin to show symptoms after several years. Thus, in the absence of
family history of the disease, the time between presentation of symptoms and
diagnosis may be quite substantial. The initial suspicion of MPS II is often
based on facial features and is made by the health care provider while exam-
ining for other issues. Laboratory diagnostic tests must be carried out to con-
firm the diagnosis.117,118
Analysis of urinary GAG levels can be used as an initial diagnostic tool
for MPS, because in almost all cases of MPS, the total urinary GAG levels
are elevated. The presence of excess DS and HS in urine is indicative of
MPS I, MPS II or MPS VII.117,118 Urinary GAG analysis is a useful initial
screening test for MPSs and may also be helpful for monitoring treatment
efficacy. Over the years, numerous methods for quantitative and qualitative
analysis of urinary GAGs have been developed. Several of the dye-based
procedures reported are qualitative screening “spot” tests, which include
the Toluidine Blue O Spot Test (Berry Spot Test)119 and the Alcian Blue
Spot Test. Furthermore, quantitative colorimetric methodologies using
1,9-dimethylmethylene blue on liquid urine samples to detect GAGs in
older children have been reported.120–122 However, these tests are not diag-
nostic of MPS II, thus additional tests are required to confirm the diagnosis.
Furthermore, a negative urinary GAG test does not necessarily rule out
MPS. False negative results may occur due to dilute samples, variation in
GAG excretion over time, and overlap in ranges between affected and unaf-
fected patients. Thus, when the patient has a family history of MPS, addi-
tional diagnostic testing should be carried out.117,118
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 19

In cases where urinary GAG levels are elevated, or if there is a strong


clinical suspicion of MPS II, enzyme activity testing should be followed.
Assays based on cultured fibroblasts, leukocytes, plasma, or serum are com-
monly used to measure the enzyme activity of IDS.117,118,123 Enzyme assays
based on the analysis of dried blood spots have also been used more recently
and are useful in areas where it is difficult to collect and transport liquid
samples.124,125 Absent or low IDS activity in males is diagnostic of MPS II
provided that another sulfatase is measured and its activity is normal. However,
enzyme activity cannot be used to measure the severity of the disease.126 In
females suspected of carrying MPS II, mutation analysis is required, since
IDS levels in female carriers and unaffected individuals show a considerable
overlap.127
Furthermore, genetic testing allows the identification of the mutation
in the IDS gene, which inevitably leads to the decrease or absence of
enzyme activity. Thus, genetic analysis can be used to confirm the diagnosis
of MPS II. In addition, genetic testing also enables the identification of the
carrier status of female relatives, which is a critical factor in family planning
decisions. The nature of IDS gene mutations and genetic heterogeneity
associated with MPS II presents major challenges to genotype and pheno-
type correlation. However, it is known that the severe form of the disease is
the result of complete deletion or rearrangement of the IDS gene.117,118

4.2 Development of IDS enzyme activity assays


4.2.1 Radiometric assays
One of the first assays used for the diagnosis of MPS II was developed by
Neufeld and co-workers. The biochemical criteria used for this assay is based
on excessive accumulation of [35S]mucopolysaccharide in skin fibroblasts
and correction of such abnormal accumulation by Hunter corrective
factor.38,39 The Hunter corrective factor is a culture medium containing
IDS obtained from fibroblast secretions or urine, derived from individuals
who do not have MPS II. This accumulation-correction test is an indirect
way to detect a total or near total deficiency of IDS. Although reliable, this
assay was not widely adopted as it requires highly purified Hunter corrective
factor and specialized techniques. Thus, Neufeld and co-workers went on to
develop a simple reliable assay for MPS II using the radioactively labeled sub-
strate 10 (Scheme 1).128 Substrate 10 is composed of a sulfated IdoA glyco-
sidically linked to a sulfated anhydromannitol that has been labeled with
tritium. Substrate 10 was obtained by sodium borotritide (NaB3H4) reduction
of disaccharide 9, derived from the nitrous acid degradation of heparin.129
ARTICLE IN PRESS

20 Shifaza Mohamed et al.

Scheme 1 Degradation of 3H-labeled substrate by a combination of IDS and α-L-


iduronidase.

Scheme 2 Reaction scheme for fluorometric assay for IDS enzyme activity.

The principle of the assay is shown in Scheme 2. Treatment of 10 with IDS


present in the Hunter corrective factor cleaves the sulfate group from the
IdoA residue, leaving a radioactive monosulfated disaccharide 11, which in
turn serves as substrate for any α-L-iduronidase that might be present. This
liberates unlabeled iduronic acid (12) and radioactive anhydromannitol sulfate
(13). Radioactive products are then separated electrophoretically from each
other and unreacted substrate, and are quantified by scintillation counting.
The use of radiolabeled substrate for estimation of IDS activity makes the
enzymatic diagnosis of MPS II accurate and reliable, but it has several draw-
backs. Labor-intensity and expensiveness are some of the limitations of the
assay, because it requires separation of products from substrate, and the han-
dling of radio-hazardous substrates. Chamoles and co-workers developed the
first dried blood spot (DBS) on filter paper assay for IDS activity using sub-
strate 10, which also demonstrated that many lysosomal enzymes are active
in rehydrated DBS.130 DBS analysis offers several advantages over whole
blood samples in terms of cost, ease of transportation, and the suitability of
the sample collection method for neonates. Furthermore, DBS methods
could be used to diagnose potential patients from areas of the world that lack
specialized laboratories.
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 21

4.2.2 Fluorometric assays


Fluorometric assays have proven to be more sensitive, specific and convenient
than the conventional radiolabeled disaccharide assays for MPS II diagnosis.
4-Methylumbelliferyl-α-L-iduronate-2-sulfate (14) is a commercially available
fluorogenic substrate for IDS (Scheme 2). It has been used in assays to detect
both MPS I and II. For MPS II, the initial enzymatic product 15 can be mea-
sured by mass spectrometry (see Section 4.2.3) or it can be hydrolyzed with α-L-
iduronidase to liberate the fluorophore 4-MU 16. Van Diggelen and
co-workers developed the first fluorogenic assay for measuring IDS enzyme
activity, which has demonstrated utility in leucocytes, plasma and skin fibro-
blasts.131 The enzymatic liberation of the fluorochrome from 15 requires the
sequential action of IDS and α-L-iduronidase. The fluorochrome 16 is detected
using a fluorometer to directly measure the kinetics of IDS activity.131 This is a
simple and rapid assay for the analysis of IDS enzyme activity. The same group
later described the use of this assay in retrospective and prospective analyses
of chorionic villi, amniotic fluid cells and cell-free amniotic fluid. They also
demonstrated its suitability for prenatal diagnosis of MPS II, as it gave early
(12th week), rapid (2–3 days) and reliable results, allowing early detection of
MPS II.132 Some of the drawbacks of this assay include long incubation times
and the need for excess purified α-L-iduronidase. Recently, this assay has been
optimized and can now be completed in less than 6 h.133
In 2011, Sista et al.134 developed an assay which improved upon van
Diggelen and co-workers’ assay by using digital microfluidics. The micro-
fluidics assay is a rapid, single-step newborn screening assay for MPS II dis-
ease in DBS samples on a digital microfluidic platform using the substrate 14
to measure IDS enzyme activity.134 This assay is also homogenous as both
the enzyme reactions are combined into a single reaction mix. The digital
microfluidic platform comprises of a disposable, self-contained microfluidics
cartridge and an instrument having an electronic fluidic control and detec-
tion capabilities. The microfluidic cartridge, in which the enzymatic reac-
tion occurs, is incorporated into the instrument, which automatically
carries out all assay manipulations. The fluorescent 4-MU product 16 is then
detected by a fluorometer within the instrument. This method can signifi-
cantly decrease the incubation time (around 90 min from extraction to
result) and automates all the liquid handling procedures by avoiding all
the discrete sample processing steps. Using this assay Sista et al. were able
to discriminate 6 MPS II patients from 105 normal newborns.
Miekle and co-workers125 drew inspiration from the work of Chamoles
et al. to develop a sensitive immunoassay to quantify IDS protein from DBS
ARTICLE IN PRESS

22 Shifaza Mohamed et al.

and plasma samples using the commercially available fluorogenic substrate


4-methylumbelliferyl sulfate (4-MU sulfate). This immunoassay uses a poly-
clonal antibody to capture IDS. The 4-MU sulfate is then added to detect
enzyme activity. IDS does not efficiently hydrolyze the substrate under
uncaptured assay conditions. This fluorogenic immune-captured IDS assay
has the advantage of allowing specific determination of IDS protein and
enzyme activity without the interfering effect of other lysosomal enzymes,
and thus, enabling the differentiation between affected and unaffected indi-
viduals. The assay is simple to perform, requires a small amount of sample
and uses commercially available fluorogenic substrate, allowing the possibil-
ity of it being adapted to large-scale, high-throughput screening programs.
However, this immunoassay requires several overnight incubation steps, and
it cannot quantify IDS protein in patients with low enzyme activity but with
normal protein levels; thus, this assay will miss those patients with significant
concentrations of inactive protein.

4.2.3 ESI-MS/MS assays


In 2007, Turecek, Gelb and co-workers developed a novel electrospray
ionization-tandem mass spectrometry (ESIMS/MS) assay for newborn
screening for IDS enzyme activity in DBS.135 This method involves addi-
tion of a designed, synthetic substrate for the selected enzyme to a buffer
rehydrated punch from DBS. After incubation, the amount of enzyme gen-
erated product is quantified, along with an isotope-labeled internal standard
by selective detection with ESI-MS/MS. MS detection is advantageous as it
offers analytical sensitivity and selectivity, and is set up for quantifying mul-
tiple enzymes during a single infusion into the instrument. The synthetic
substrate for the assay was designed to closely mimic the carbohydrate struc-
tural moieties in natural GAGs. Furthermore, the enzymatic products and
internal standards were designed to have mutually exclusive molecular
masses. The synthetic IDS substrate 17 (Scheme 3) was derived from the
nitrous acid degradation of commercially available heparin. Substrate 17
contains a hydrophobic carbon chain allowing purification of the enzymatic
product 18a by reversed-phase C18 chromatography. Furthermore, pres-
ence of the benzoyl group on 17 gave a practical and inexpensive heavy iso-
tope tag.135 In this assay, 17 is desulfated by IDS to produce 18a. The
amount of 18a was determined by comparing ion peak intensities of 18a
and the internal standard 18b.135 The isotope-labeled internal standard
18b was designed to be chemically identical to the enzymatic product
18a but is 5 Da heavier due to the presence of five deuterium atoms in
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 23

Scheme 3 IDS enzyme product of substrate 17 and its CID ion products.

the benzoyl group. ESI-MS/MS can then detect and quantify product ions
19a and 19b, after collision-induced dissociation (CID) of the 6-sulfate
group present in the 2,5-anhydromannosyl residues (80 Da mass difference).
This ESI-MS/MS assay proved to be sensitive, only required a small amount
of sample, and had the capability for multiplexing to assay several different
lysosomal enzymes for LSDs. However, a potential limitation of this assay is
the interfering effect of other lysosomal or non-lysosomal enzymes present
in DBS. In addition, it was impractical to attain the amount of substrate 17
needed to support worldwide newborn screening for MPS II due to the dif-
ficulties with scale-up preparation of 17 using the nitrous acid degradation of
heparin.
Thus, Gelb and co-workers developed a new method for large-scale
preparation (tens of grams per year) of appropriate substrates for
IDS.136,137 The common structural features of the substrates are (1) a group
that is specifically recognized by the enzyme, (2) a hydrophobic carbon
chain as part of the enzyme-generated product that allows chromatographic
separation, and (3) a readily fragmentable functional group that leads to a
dominant fragmentation pathway in the mass spectrometer (improving assay
sensitivity). The target substrate α-L-iduronidate-2-sulfate glycoside 23
(Scheme 4) consists of an umbelliferyl group bearing a hydrophobic linker
with a terminal tert-butyloxycarbamate group. The substrates were synthe-
sized starting from α-L-iduronate glycoside methyl ester 20, prepared pre-
viously by the Gelb group using a nine-step synthetic sequence.123
Amide coupling with a Boc-protected diamine to give 21 was followed
by selective sulfation at the C-2 position via the stannylene acetal 22 and
ARTICLE IN PRESS

24 Shifaza Mohamed et al.

Scheme 4 Synthesis of IDS substrates and internal standard for ESIMS/MS (A) and the
resulting CID ion products (B).

deprotection of the methyl ester to give target substrates 23a and 23b.136
Substrate 23b has a longer hydrophobic carbon chain than that of 23a,
which results in better purification by HPLC, and is thus the preferred sub-
strate. Substrate 23b can be used to assay IDS enzyme activity using either a
fluorometric assay (see Section 4.2.2) or an ESI-MS/MS assay (this section).
The former is made possible by the presence of the umbelliferyl moiety,
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 25

which when supplemented with the enzyme α-L-iduronidase cleaves to


release the fluorescent coumarin. For the latter assay, the desulfated α-L-
iduronidate glycoside 24 can be detected directly by ESI-MS/MS with
quantification using a deuterium-labeled internal standard 24b.137 The
internal standard 24b consists of a t-Boc group with nine deuterium atoms
and is designed to be chemically identical to the enzymatic product 24.
Later, substrate 23b along with the internal standard 24b was utilized in
an UPLC-MS/MS multiplexing assay of nine enzymes in DBS samples suit-
able for newborn screening of LSDs.138 The sample preparation for this assay
required only four liquid transfers before injection into a UPLC system with
a dual column: an analytical column was used to carry out sample separa-
tions, while the guard column was being equilibrated with solvent for the
injection of the next sample. This UPLC-MS/MS assay can automatically
process the samples efficiently, with an inject-to-inject time of only
1.8 min, thus emphasizing the multiplexing capacity of the tandem mass
spectrometry (MS/MS) method and its compatibility for the large workflow
of newborn screening laboratories.
Song and co-workers developed a MS/MS-based assay for IDS activity
which utilized commercially available (fluorogenic) substrate 14 and
UPLC-MS/MS;139 the IDS enzymatic product 15 was measured directly
by UPLC-MS/MS using commercially available 4-methylumbelliferyl
α-L-idopyranoside as internal standard. Using this assay, Song and
co-workers analyzed IDS enzyme activities in DBS of normal newborns
and MPS II patients and reported a significant decrease in enzyme activity
for the three patient DBS samples as compared to 110 normal DBS sam-
ples. However, additional analyses with a larger number of DBSs are nec-
essary to further evaluate and validate the method.
Gelb and co-workers then went on to design and synthesize a superior
IDS substrate 30 (Scheme 5) for use in newborn screening of MPS II. This
substrate eliminates the unnecessary fluorescent aglycone and contains a
bisamide unit that is hypothesized to readily protonate in the gas phase, thus
improving detection sensitivity by ESI-MS/MS. It also contains a benzoyl
group that provides a site for inexpensive deuteration, thus facilitating the
preparation of internal standards for accurate quantification of enzymatic
products.140 The lipophilic side chain also facilitates desalting prior to MS
analysis by improving the extraction efficiency into organic solvent (ethyl
acetate). Substrate 30 was prepared from aglycone 28 by glycosylation with
IdoA donor 27, followed by deprotection and subsequent selective sulfation
of the 2-OH group. This synthetic protocol is amenable for scale-up as it
ARTICLE IN PRESS

26 Shifaza Mohamed et al.

Scheme 5 (A) Synthesis of IDS substrate 30 for ESIMS/MS assay; (B) diagnostic fragment
ion from CID of product 31 following cleavage of 30 by IDS; (C) diagnostic fragment ion
from CID of aglycone 34 following cleavage of 31 by α-L-iduronidase.

does not require the installation of a 7-hydroxycoumarin unit as with sub-


strate 23. LC-MS/MS detection of product 31 following cleavage of the
substrate by IDS resulted in a 5.8-fold improvement in sensitivity. The tan-
dem mass spectrometry of product 31 gives rise to a major fragment ion 33
due to a specific cleavage pathway (Scheme 5B). This improves the sensitiv-
ity of product detection as only a single product ion is monitored in the assay.
Formation of multiple products would have resulted in a decrease in signal
intensity thus reducing the sensitivity of the assay. Due to the increased sen-
sitivity of this ESI-MS/MS assay for determining IDS enzyme activity, it
gives a better differentiation between healthy individuals and disease-
affected newborns, compared to the original IDS substrate 23b.
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 27

In 2015, Gelb, Spacil and co-workers further enhanced the ESI-MS/MS


assay for IDS resulting in a higher analytical range assay.141 This assay is based
on the hypothesis that the aglycone of substrate 30 (i.e., 34) would give a
stronger MS/MS response per mole than that for the immediate sulfatase
product 31.141 Upon IDS treatment of substrate 30, the initial product 31
contains the sugar, which is hydrophilic and thus reduces the solvent extrac-
tion yield. Therefore, conversion to the aglycone will increase the transfer of
analyte to the organic layer, resulting in higher sensitivity. In this improved
assay, substrate 30 was initially reacted with IDS from the DBS to remove
the 2-sulfate group. This was followed by treatment with α-L-iduronidase
resulting in the cleavage of the glycoside to generate free aglycone 34
(Scheme 5C). The amount of product aglycone 34 was then quantified
by ESI-MS/MS using the deuterated internal standard aglycone, after
CID of the precursor ions in MS/MS.141 As predicted by Gelb and
co-workers, the free aglycone 34 gave a stronger MS/MS assay response
due to the enhanced transfer of aglycone analyte to the organic layer.141
The analytical range of this ESI-MS/MS assay was compared to the fluoro-
metric assay using 4-methylumbelliferyl substrate 14 and was found to give a
much higher analytical range (34-fold higher) than the fluorometric assay.141
Due to its increased sensitivity, this assay leads to a lower rate of false pos-
itives in newborn screening, a more accurate diagnosis of MPS II disease, a
better differentiation between healthy and MPS II samples, and a better pre-
diction of disease severity.

4.3 Assays based on biomarkers of MPS II


MPS disorders including MPS II are characterized by the lysosomal storage
of partially degraded GAGs in tissues resulting in elevated concentration of
these compounds in body fluids, urine, plasma and blood. These metabolic
markers therefore have potential application in diagnosis, phenotype predic-
tion, and monitoring of therapies. A number of assays for measuring GAGs
in patient samples have been reported in the literature and have been
reviewed.15 The strategy most commonly used for quantification of GAGs
is to reduce the complexity of the analytical problem by quantifying smaller
oligosaccharides by MS. This has been achieved mostly by depolymerization
of polysaccharides to disaccharides, which can be accomplished either
chemically or by enzymatic cleavage. In both these processes the native state
of the analyte is lost; however, the molar amount of disaccharides is much
higher than that of the starting material. Furthermore, the number of
ARTICLE IN PRESS

28 Shifaza Mohamed et al.

different chemical species is greatly reduced, and the size of the molecules is
much smaller. This is crucial for MS analysis, as small molecules are more
amenable to standard reversed-phase separation and subsequent MS analysis.
Tomatsu et al. demonstrated that for MPS I, II, III, and VI the characteristic
accumulation of GAGs is represented by increased levels of HS and DS
derived disaccharides in plasma.142,143
The function of IDS is to hydrolyze the C2-sulfate of IdoA residues at
the non-reducing end of HS and DS, and thus, the HS- and DS-derived oli-
gosaccharides from MPS II patients have non-reducing end iduronate-
2-sulfate. Fuller et al. used ESI-MS/MS to identify di- to pentasaccharides
isolated from urine samples of MPS II patients.144 Oguma et al. reported the
development of an HPLC-ESI-MS/MS method capable of detecting
nanomolar amounts of HS- and DS-derived disaccharides in serum and
plasma and showed that these disaccharides are elevated approximately
5- to 10-fold in MPS II patients compared to unaffected controls.143 This
method requires digestion of sample GAGs to disaccharides with bacterial
endo-enzymes prior to analysis. Pan et al. developed an LC-MS/MS
assay to quantify the elevated DS levels in the CSF of MPS II patients
compared with controls. DS was quantified by ion-pairing LC-MS/MS
analysis of the major disaccharides, DS46 and DS6S, from digestion with
chondroitinase B.145 In 2010, Fuller and co-workers developed an
HPLC-MS/MS assay for determining intact HS- and DS-derived di- to
pentasaccharides that they previously identified in MPS II urine, as
3-methyl-1-phenyl-5-pyrazolone derivatives.146 It was found that the ele-
vated levels of each of the oligosaccharides enabled a complete differenti-
ation of the MPS II patients from unaffected controls.146 Furthermore, a
number of oligosaccharides were more abundant in MPS II patients with
CNS involvement compared with patients without CNS disease. Recently,
following technological improvements in HPLC, Fuller and co-workers
have expanded this method to diagnose 10 MPS subtypes with 100% spec-
ificity and sensitivity.147 Identification of MPS II is reliant on the presence
of a diagnostic sulfated disaccharide UA-HNAc (1S).
In 2012, Lawrence et al. developed an approach for diagnosis of MPS
disorders including MPS II by determining the characteristic non-
reducing end structures of GAGs as diagnostic biomarkers.148 Lysosomal
degradation of GAGs occurs in an ordered manner from the non-reducing
end of the chains. Thus, absence of any one enzyme in the pathway results
in the accumulation of characteristic non-reducing terminal carbohydrate
structures. The approach involved liberating sets of disease-specific
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 29

Scheme 6 Depolymerization of HS from MPS II patient samples or animal model


sources with heparin lyase, followed by derivatization with aniline, results is a diagnostic
biomarker 37 readily distinguished by LC-MS/MS.148

biomarkers derived from the non-reducing ends of the GAGs that accu-
mulate in MPS patients. These biomarkers were then readily distinguished
from internal segments of the chain by LC-MS and were then quantified.
For the diagnosis of MPS II, HS was depolymerized by the bacterial
enzyme heparin lyase, releasing the non-reducing end disaccharide
2-sulfoiduronic acid-N-sulfoglucosamine-6-sulfate (35) along with unsat-
urated disaccharides from internal cleavage (e.g., 36, Scheme 6).148 The
digestion products were derivatized with isotope-labeled aniline by reduc-
tive amination to facilitate improved LC resolution and quantification of
recovery with available standards. The non-reducing end biomarker (37)
for MPS II was readily distinguished from internal disaccharides (38) by its
greater mass in the LC-MS/MS.148
De Ruijter et al. also reported a similar assay to analyze levels of HS- and
DS-derived disaccharides in newborn DBS.149 The DBS were enzymati-
cally treated using heparinase I, II, III and chondroitinase B to liberate disac-
charides that were then quantified by LC-MS/MS. D0A0,150 the most
abundant disaccharide in HS, and D0a4, which makes up 94% of DS, were
the only two disaccharides that could be detected. The levels of both disac-
charides were significantly elevated in MPS patients compared with con-
trols, although the method is not specific for MPS II. Similarly, various
other assays have been described that depolymerize HS into disaccharides,
for example, by methanolysis151–154 or butanolysis155,156 degradation.
ARTICLE IN PRESS

30 Shifaza Mohamed et al.

These methods are suitable for detecting elevated HS levels indicative of


MPS from a variety of biological samples, e.g., tissue, urine, and CSF; how-
ever, they are not specific for MPS II, and further work is needed to identify
specific disaccharide signatures157 in order to diagnose the particular form
of MPS.
In summary, newborn screening assays for the diagnosis of MPS II disease
that give early and accurate detection involve fluorometric enzymatic assays,
immunoassays, MS/MS assays and biomarker analysis. ESI-MS/MS based
enzyme activity assays have several advantages over fluorometric assays, such
as low false-negative rates, the capability of performing multiple lysosomal
enzyme assays on individual dried blood spots, and providing a larger
analytical range leading to a more accurate determination of enzyme activity,
and better differentiation between disease-affected and non-affected indi-
viduals.158 Biomarker quantification methods are promising but will likely
be used for second-tier analysis and monitoring of therapies given their more
involved and lengthy procedures. The development of diagnostic assay
options for MPS II disease provides a degree of flexibility that meets the dif-
ferent preferences among the large number of laboratories worldwide that
screen for newborns.

5. Management and treatment of MPS II


5.1 Overview
The management of MPS II requires lifelong attention since none of the
therapeutic options currently available result in complete resolution of mor-
bidity. The wide range of clinical symptoms of MPS II on the body and their
severity require substantial multidisciplinary medical and surgical support for
the patients (see Section 2.3).28,78 Most of the treatments available for MPS
II are symptomatic; however, in recent years much progress has been made
in developing efficient therapeutic options for managing the disease. This
section of the review will give a brief outline of the existing treatments
and their effectiveness.

5.2 Enzyme replacement therapy


The treatment of MPS II was palliative prior to the introduction of enzyme
replacement therapy (ERT), which is the current standard of care. The
concept of ERT is reconstitution of the missing or defective enzymes by
intravenous administration of a recombinant human enzyme.8,159
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 31

Currently, there are two recombinant IDS enzymes available for ERT for
MPS II patients: idursulfase (Elaprase®)160 or idursulfase beta (Hunterase®).161
Both idursulfase and idursulfase beta are derived from the human IDS gene via
recombinant DNA technology and thus have identical amino acid sequences.
However, idursulfase is produced from an HT-1080 cell line, while
idursulfase beta is produced from a CHO cell line. This accounts for some
differences in their post-translational modifications, such as formylglycine
content, which results in differences in specific enzyme activity. The enzymes
are heavily glycosylated with mannose-6-phosphate (M6P) and sialylated gly-
cans. The former allows selective binding to M6P receptors on the surface of
the cells, resulting in subsequent cellular internalization. The levels of M6P
and sialic acid are similar in both products but there is a difference in their
cellular uptake.161 A recent comparative study indicated that idursulfase beta
contained less high-mannose type and hybrid-type glycans compared with
idursulfase, which could account for its lower immunogenicity.162
ERT has been shown to benefit a majority of individuals with MPS II by
reducing urinary GAG excretion levels, decreasing the volumes of their liver
and spleen, and increasing their cardiopulmonary function and average
walking distance.8,160,163,164 ERT with idursulfase or idursulfase beta is gen-
erally well tolerated, and the adverse infusion-related events that commonly
occur during therapy can be easily managed by the temporary pausing of infu-
sion, or by providing antihistamines and/or steroid medications before subse-
quent infusions.163,164 However, ERT is not a cure and has several limitations.
ERT using idursulfase is an expensive therapy with an estimated average cost
of US$490,000 in a 30 kg child.163 Moreover, the recombinant enzyme
does not cross the blood–brain barrier (BBB) and hence does not alter the pro-
gression of CNS disease among severely affected patients.27 In order to address
the issues with lack of BBB penetration, ERT with delivery by the intrathecal
or intracerebroventricular route is being investigated in several clinical trials.165
Another approach under investigation is the development of IDS-fusion
proteins specifically engineered to cross the BBB. For example, AGT-182 is
a fusion protein containing IDS and an anti-human insulin receptor mono-
clonal antibody (HIRMAb).166 The HIRMAb domain of the fusion protein
acts as a molecular Trojan horse to ferry the fused IDS across the BBB
via receptor-mediated transport on the insulin receptor, and across the cell
plasma membrane by receptor-mediated endocytosis. Brain uptake of AGT-
182 as well as safety and pharmacokinetics were demonstrated in Rhesus
monkeys following intravenous administration.166,167 JR-141 is another
fusion protein that consists of an IDS enzyme linked to an anti-transferrin
ARTICLE IN PRESS

32 Shifaza Mohamed et al.

receptor antibody. It has been reported to cross the BBB, reduce GAG accu-
mulation in the brain, and maintain cognitive functions in the MPS II
mouse model.168 Both AGT-182 and JR-141 are undergoing clinical
evaluation.165

5.3 Substrate reduction therapy


Substrate reduction therapy (SRT) aims to use small-molecule inhibitors of
GAG biosynthesis to reduce the concentration of accumulating substrate to
a level where the residual degradative enzymes can maintain homeostasis.169
SRT is a promising approach to treatment and is not specific to MPS II but
rather is applicable to all MPS disorders. Many small molecules have been
identified as inhibitors of GAG biosynthesis,170 but so far only a few com-
pounds have been evaluated as treatments for MPS. Genistein (39, Fig. 6),
a non-toxic isoflavone, has been shown to reduce GAG levels in various
organs, including the brain, in MPS II and MPS IIIB mice, and also corrects
neuroinflammation and behavior of these animals.171 In a study conducted
with seven MPS II patients over 26 weeks of treatment, genistein was well
tolerated and resulted in improved joint mobility.172 Although the results with
genistein are promising, long-term efficacy still needs to be demonstrated. The
results from clinical trials in other MPS disorders have been inconclu-
sive.173,174 The dye rhodamine B (40) has also shown some potential when
investigated as a GAG biosynthesis inhibitor for SRT in MPS I and
IIIA,175,176 but it has not been tested in MPS II to date. In contrast, rather
than inhibiting biosynthesis, the orally available 5-thio-β-D-xyloside odiparcil,
(41)177 reduces cellular GAG levels by competing with endogenous xyloside-
containing core proteins for GAG assembly.178 This results in reduced
endogenous proteoglycan-bound GAGs while increasing odiparcil-bound
GAGs, which are excreted in the urine instead of building up in the lysosome.
Originally developed as an anticoagulant,179 odiparcil is currently in clinical
development for MPS VI,180 but should also be applicable to MPS II.

Fig. 6 Structures of compounds evaluated for substrate reduction therapy for MPS
disorders.
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 33

5.4 Pharmacological chaperone therapy


The genetic mutations in LSDs often result in the formation of misfolded
enzymes. Such misfolded enzymes are prematurely degraded by cellular pro-
teases within the endoplasmic reticulum-associated degradation machinery
of the cell before they are transported to their intended location, the lyso-
some. Pharmacological chaperone therapy (PCT) relies on small molecules
(typically active-site-directed, reversible, enzyme inhibitors) that interact
with the enzyme to enhance folding, stability and trafficking efficiency to
the lysosome, thus increasing lysosomal enzyme concentration and restoring
partial enzyme activity.181,182 Moreover, as small molecules, PCs have the
potential to be bio-distributed widely and to penetrate the BBB to amelio-
rate CNS symptoms. Furthermore, although PCT does not have an impact
on missense or deletion mutations, there is some evidence that PCs can
enhance the effectiveness of ERT by protecting recombinant wild type
enzyme against misfolding and degradation.183 PCT has been intensively
investigated for many LSDs and the concept was recently validated with
the approval of the first PCT drug migalastat (Galafold®) for the treatment
of Fabry disease.184
Hoshina et al. recently tested the commercially available unsaturated disac-
charide D2S0 (42, Fig. 7) as a chaperone for MPS II.185 D2S0 is derived from
enzymatic cleavage of heparin and resembles the natural substrate of IDS.

Fig. 7 Structures of pharmacological chaperone DS20 (42) and multivalent IDS inhibi-
tors with putative pharmacological chaperone activity (43–45).
ARTICLE IN PRESS

34 Shifaza Mohamed et al.

When D2S0 was incubated with recombinant IDS in vitro, it enhanced the
stability of the enzyme toward thermal degradation in a dose-dependent
manner. In addition, D2S0 increased the residual activity of IDS in MPS II
patient fibroblasts and in HEK293T cells expressing mutated IDS. Cardona
and co-workers synthesized a nonavalent trihydroxypiperidine dendrimer
43 as a glycosidase inhibitor. Interestingly, 43 was also evaluated against
IDS and found to have modest inhibitory activity (69% at 1 mM),186
thus identifying it as a potential PC for MPS II. Replacement of the piper-
idine core of the dendrimer with a pyrrolidine resulted in more potent
inhibitors 44 (IC50 ¼ 140 μM) and 45 (IC50 ¼ 31 μM).187 Multivalent pre-
sentation of the same pyrrolidine as in 44 onto gold nanoparticles resulted
in a poor inhibitor for IDS,188 although it did show good inhibition of
N-acetylgalactosamine-6-sulfatase, whose deficiency leads to MPS IVA.
The evaluation of the chaperone activity of these compounds for IDS
has yet not been reported.

5.5 Other treatments


Hematopoietic stem cell transplantation (HSCT) via umbilical cord blood
transplantation or bone marrow transplantation has been used as a way of
providing sufficient enzyme activity to slow or stop the progression in cer-
tain LSDs. HSCT is considered as a mainstay treatment for severe cases of
MPS I and has shown promising results in other LSDs.189 Even though
HSCT has been shown to be beneficial in patients suffering from MPS I,
the outcomes in patients with MPS II has not been very promising.23,190
In some cases individuals have shown improvement of visceral and skeletal
manifestations, including decreased urinary GAG levels, decreased liver and
spleen volume, diminished facial coarsening, improved respiratory function,
and increased joint mobility. However, HSCT has not been shown to stop
the progression of neurodegenerative manifestations of MPS II.190,191 This is
perhaps because, unlike MPS I, MPS II cannot be diagnosed in infancy and
thus transplant usually occurs past 2 years of age after the onset of CNS dis-
ease.189 Long-term follow-up of MPS II patients who have undergone
HSCT showed little or no neurological improvement.190,192 Furthermore,
the significantly high morbidity and mortality associated with HSCT makes
it a less attractive form of treatment for MPS II.
Gene therapy is an attractive approach to LSDs because it could be a
one-time, permanent therapy that repairs the cause of the enzyme defi-
ciency. Gene therapy for MPS disorders have not yet been approved;
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 35

however, clinical trials are either in progress or are scheduled, including for
MPS II (e.g., RGX-121 and SB-913).193 In preclinical studies, intracisternal
gene therapy with an adeno-associated virus serotype 9 (AAV9) vector
encoding murine IDS was reported to correct neurological and systemic
symptoms in MPS II mice.194 In other studies, an AAV9 vector carrying
the human IDS gene was administered to MPS II mice via the intra-
cerebroventricular route.195,196 The treatments showed reduced GAG accu-
mulation in the brain and improved neurological and behavioral responses in
the mice. These studies indicate that intra-CSF administration of AAV9
gene therapy should be useful for MPS II patients. RGX-121 is a gene ther-
apy product in clinical development that consists of an AAV9 vector con-
taining the human IDS gene administered by intrathecal injection.193
SB-913 contains a zinc finger nuclease (ZFN) in an AAV6 vector delivered
by intravenous infusion and is undergoing clinical trials.193 The ZFN is a
genetic engineering tool that modifies DNA sequences, mediating the
site-specific insertion of the corrective gene.

6. Conclusions
MPS II is a rare, inherited lysosomal storage disease caused by muta-
tions in the enzyme IDS. The function of IDS is to cleave the 2-O-sulfate
groups on terminal, non-reducing end IdoA residues on HS and DS in the
lysosome. Deficiency of IDS activity results in accumulation of HS and DS
in the lysosome leading to pathological changes in multiple organs. MPS II is
a progressive disease with a wide range of symptoms. Severely affected indi-
viduals have profound neurological impairment and a significantly short-
ened lifespan. An understanding of the function of IDS has led to
significant progress in the development of various assays, particularly those
based on LC-MS/MS, for enzyme activity or detecting the presence of bio-
markers for diagnosis/newborn screening and for monitoring of new ther-
apies. The current standard of care is ERT; however, the recombinant
enzyme does not cross the BBB and thus has little impact on neurological
symptoms. The past decade has seen significant progress toward the devel-
opment of new treatment options, and some of these have progressed to
clinical trials. These include efforts to develop IDS-fusion proteins specifi-
cally engineered to cross the BBB, small-molecule therapies based on sub-
strate reduction or pharmacological chaperones, and gene therapy
approaches, with the latter offering hope of a one-time permanent treat-
ment. Continued research and the combination of multiple effective
ARTICLE IN PRESS

36 Shifaza Mohamed et al.

therapies with early diagnosis could result in improved prospects for MPS II
patients in the future.

Acknowledgments
We thank the University of Queensland, the National MPS Society (USA), and the
Australian Research Council (DP170104431 to V.F.) for financial support.

Abbreviations
AAV adeno-associated virus
anM 2,5-anhydro-D-mannitol
BBB blood–brain barrier
CID collision-induced dissociation
CNS central nervous system
CS chondroitin sulfate
CSF cerebrospinal fluid
DBS dried blood spot
DNA deoxyribonucleic acid
DS dermatan sulfate
ERT enzyme replacement therapy
ESI-MS/MS electrospray ionization tandem mass spectrometry
FGly α-formylglycine
GAG glycosaminoglycan
HIRMAb human insulin receptor monoclonal antibody
HPLC high performance liquid chromatography
HPLC-MS/MS high performance liquid chromatography-tandem mass spectrometry
HS heparan sulfate
HSCT hematopoietic stem-cell transplant
IdoA L-iduronic acid
IDS iduronate-2-sulfatase
KS keratan sulfate
LC liquid chromatography
LC-MS/MS liquid chromatography-tandem mass spectrometry
LSD lysosomal storage disease
M6P mannose-6-phosphate
MPS mucopolysaccharidosis
MS/MS tandem mass spectrometry
MU 4-methylumbelliferyl
NBS newborn screening
PC pharmacological chaperone
PCT pharmacological chaperone therapy
SRT substrate reduction therapy
UPLC ultra-performance liquid chromatography
UPLC-MS/MS ultra-performance liquid chromatography-tandem mass spectrometry
ZFN zinc finger nuclease
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 37

References
1. Boustany, R.-M. N. Lysosomal Storage Diseases—The Horizon Expands. Nat. Rev.
Neurol. 2013, 9, 583–598.
2. Vellodi, A. Lysosomal Storage Disorders. Br. J. Haematol. 2005, 128, 413–431.
3. Ballabio, A.; Gieselmann, V. Lysosomal Disorders: From Storage to Cellular Damage.
Biochim. Biophys. Acta 2009, 1793, 684–696.
4. Winchester, B. Classification of Lysosomal Storage Diseases. In: Lysosomal Storage
Disorders; Mehta, A., Winchester, B., Eds.; John Wiley & Sons, Ltd, 2012, pp 37–46.
5. Beck, M. Emerging Drugs for Lysosomal Storage Diseases. Expert Opin. Emerg. Drugs
2010, 15, 495–507.
6. Poorthuis, B. J. H. M.; Wevers, R. A.; Kleijer, W. J.; Groener, J. E. M.; de
Jong, J. G. N.; van Weely, S.; Niezen-Koning, K. E.; van Diggelen, O. P. The Fre-
quency of Lysosomal Storage Diseases in The Netherlands. Hum. Genet. 1999, 105,
151–156.
7. Meikle, P. J.; Hopwood, J. J.; Clague, A. E.; Carey, W. F. Prevalence of Lysosomal
Storage Disorders. J. Am. Med. Soc. 1999, 281, 249–254.
8. Burrow, T. A.; Hopkin, R. J.; Leslie, N. D.; Tinkle, B. T.; Grabowski, G. A. Enzyme
Reconstitution/Replacement Therapy for Lysosomal Storage Diseases. Curr. Opin.
Pediatr. 2007, 19, 628–635.
9. Heese, B. A. Current Strategies in the Management of Lysosomal Storage Diseases.
Semin. Pediatr. Neurol. 2008, 15, 119–126.
10. Platt, F. M.; Lachmann, R. H. Treating Lysosomal Storage Disorders: Current Practice
and Future Prospects. Biochim. Biophys. Acta 2009, 1793, 737–745.
11. Lachmann, R. Treatments for Lysosomal Storage Disorders. Biochem. Soc. Trans. 2010,
38, 1465–1468.
12. Murthy, T. E. G. K.; Nagarjuna, S.; Vali, P. S.; Saritha, T.; Rao, G. M. S. Lysosomal
Storage Disorders and Treatment. Int. J. PharmTech Res. 2010, 2, 1082–1091.
13. Pastores, G. M. Therapeutic Approaches for Lysosomal Storage Diseases. Ther. Adv.
Endocrinol. Metab. 2010, 1, 177–188.
14. Ortolano, S.; Vieitez, I.; Navarro, C.; Spuch, C. Treatment of Lysosomal Storage Dis-
eases: Recent Patents and Future Strategies. Recent Pat. Endocr. Metab. Immune Drug
Discov. 2014, 8, 9–25.
15. Tomatsu, S.; Fujii, T.; Fukushi, M.; Oguma, T.; Shimada, T.; Maeda, M.; Kida, K.;
Shibata, Y.; Futatsumori, H.; Montaño, A. M.; Mason, R. W.; Yamaguchi, S.;
Suzuki, Y.; Orii, T. Newborn Screening and Diagnosis of Mucopolysaccharidoses.
Mol. Genet. Metab. 2013, 110, 42–53.
16. Kingma, S. D. K.; Bodamer, O. A.; Wijburg, F. A. Epidemiology and Diagnosis of
Lysosomal Storage Disorders; Challenges of Screening. Best Pract. Res. Clin. Endocrinol.
Metab. 2015, 29, 145–157.
17. Gandhi, N. S.; Mancera, R. L. The Structure of Glycosaminoglycans and Their Inter-
actions With Proteins. Chem. Biol. Drug. Des. 2008, 72, 455–482.
18. Rabenstein, D. L. Heparin and Heparan Sulfate: Structure and Function. Nat. Prod.
Rep. 2002, 19, 312–331.
19. McKusick, V. A.; Kaplan, D.; Wise, D.; Hanley, W. B.; Suddarth, S. B.; Sevick, M. E.;
Maumanee, A. E. The Genetic Mucopolysaccharidoses. Medicine 1965, 44, 445–483.
20. Dorfman, A.; Matalon, R. The Mucopolysaccharidoses (A Review). Proc. Natl. Acad.
Sci. U. S. A. 1976, 73, 630–637.
21. Coutinho, M. F.; Lacerda, L.; Alves, S. Glycosaminoglycan Storage Disorders:
A Review. Biochem. Res. Int. 2012, 2012 471325.
22. Clarke, L. A. The Mucopolysaccharidoses: A Success of Molecular Medicine. Expert
Rev. Mol. Med. 2008, 10, e1.
ARTICLE IN PRESS

38 Shifaza Mohamed et al.

23. Muenzer, J. Overview of the Mucopolysaccharidoses. Rheumatology 2011, 50, v4–v12.


24. Cimaz, R.; La Torre, F. Mucopolysaccharidoses. Curr. Rheumatol. Rep. 2014, 16, 389.
25. Giugliani, R. Mucopolysacccharidoses: From Understanding to Treatment, a Century
of Discoveries. Genet. Mol. Biol. 2012, 35, 924–931.
26. Muenzer, J. The Mucopolysaccharidoses: A Heterogeneous Group of Disorders With
Variable Pediatric Presentations. J. Pediatr. 2004, 144, S27–S34.
27. Wraith, J. E.; Scarpa, M.; Beck, M.; Bodamer, O. A.; De Meirleir, L.; Guffon, N.;
Meldgaard Lund, A.; Malm, G.; Van der Ploeg, A. T.; Zeman, J. Mucopolysaccharidosis
Type II (Hunter Syndrome): A Clinical Review and Recommendations for Treatment
in the Era of Enzyme Replacement Therapy. Eur. J. Pediatr. 2008, 167, 267–277.
28. Beck, M. Mucopolysaccharidosis Type II (Hunter Syndrome): Clinical Picture and
Treatment. Curr. Pharm. Biotechnol. 2011, 12, 861–866.
29. Hunter, C. A Rare Disease in Two Brothers. Proc. R. Soc. Med. 1917, 10, 104–116.

30. Hurler, G. Uber Einen Typ Multipler Abartungen, Vorwiegend am Skelettsystem.
Z. Kinderheilkd. 1920, 24, 220–234.
31. Lorincz, A. E. The Mucopolysaccharidoses: Advances in Understanding and Treat-
ment. Pediatr. Ann. 1978, 7, 104–122.
32. Brante, G. Gargoylism—A Mucopolysaccharidosis. Scand. J. Clin. Lab. Invest. 1952, 4,
43–46.
33. Dorfman, A.; Lorincz, A. E. Occurrence of Urinary Acid Mucopolysaccharides in the
Hurler Syndrome. Proc. Natl. Acad. Sci. U. S. A. 1957, 43, 443–446.
34. Fratantoni, J. C.; Hall, C. W.; Neufeld, E. F. The Defect in Hurler’s and Hunter’s Syn-
dromes: Faulty Degradation of Mucopolysaccharide. Proc. Natl. Acad. Sci. U. S. A.
1968, 60, 699–706.
35. Fratantoni, J. C.; Hall, C. W.; Neufeld, E. F. The Defect in Hurler and Hunter Syn-
dromes, II. Deficiency of Specific Factors Involved in Mucopolysaccharide Degrada-
tion. Proc. Natl. Acad. Sci. U. S. A. 1969, 64, 360–366.
36. Neufeld, E. F.; Cantz, M. J. Corrective Factors for Inborn Errors of Mucopolysaccha-
ride Metabolism. Ann. N. Y. Acad. Sci. 1971, 179, 580–586.
37. Cantz, M.; Chrambach, A.; Neufeld, E. F. Characterization of the Factor Deficient in
the Hunter Syndrome by Polyacrylamide Gel Electrophoresis. Biochem. Biophys. Res.
Commun. 1970, 39, 936–942.
38. Cantz, M.; Chrambach, A.; Bach, G.; Neufeld, E. F. The Hunter Corrective Factor:
Purification and Preliminary Characterization. J. Biol. Chem. 1972, 247, 5456–5462.
39. Bach, G.; Eisenberg, F.; Cantz, M.; Neufeld, E. F. The Defect in the Hunter
Syndrome: Deficiency of Sulfoiduronate Sulfatase. Proc. Natl. Acad. Sci. U. S. A.
1973, 70, 2134–2138.
40. Wilson, P. J.; Morris, C. P.; Anson, D. S.; Occhiodoro, T.; Bielicki, J.;
Clements, P. R.; Hopwood, J. J. Hunter Syndrome: Isolation of an Iduronate-2-
Sulfatase cDNA Clone and Analysis of Patient DNA. Proc. Natl. Acad. Sci. U. S. A.
1990, 87, 8531–8535.
41. Wilson, P. J.; Suthers, G. K.; Callen, D. F.; Baker, E.; Nelson, P. V.; Cooper, A.;
Wraith, J. E.; Sutherland, G. R.; Morris, C. P.; Hopwood, J. J. Frequent Deletions
at Xq28 Indicate Genetic Heterogeneity in Hunter Syndrome. Hum. Genet. 1991,
86, 505–508.
42. Flomen, R. H.; Green, E. P.; Green, P. M.; Bentley, D. R.; Giannelli, F. Determina-
tion of the Organisation of Coding Sequences Within the Iduronate Sulphate Sul-
phatase (IDS) Gene. Hum. Mol. Genet. 1993, 2, 5–10.
43. Wilson, P. J.; Meaney, C. A.; Hopwood, J. J.; Morris, C. P. Sequence of the Human
Iduronate 2-Sulfatase (IDS) Gene. Genomics 1993, 17, 773–775.
44. Human Gene Mutation Database. http://www.hgmd.cf.ac.uk/ac/index.php; 2019.
(accessed March 2019).
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 39

45. Stenson, P. D.; Ball, E. V.; Mort, M.; Phillips, A. D.; Shiel, J. A.; Thomas, N. S. T.;
Abeysinghe, S.; Krawczak, M.; Cooper, D. N. Human Gene Mutation Database
(HGMD®): 2003 Update. Hum. Mutat. 2003, 21, 577–581.
46. Rathmann, M.; Bunge, S.; Beck, M.; Kresse, H.; Tylki-Szymanska, A.; Gal, A. Muco-
polysaccharidosis Type II (Hunter Syndrome): Mutation "Hot Spots" in the Iduronate-
2-Sulfatase Gene. Am. J. Hum. Genet. 1996, 59, 1202–1209.
47. Filocamo, M.; Bonuccelli, G.; Corsolini, F.; Mazzotti, R.; Cusano, R.; Gatti, R.
Molecular Analysis of 40 Italian Patients With Mucopolysaccharidosis Type II: New
Mutations in the Iduronate-2-Sulfatase (IDS) Gene. Hum. Mutat. 2001, 18, 164–165.
48. Froissart, R.; da Silva, I. M.; Guffon, N.; Bozon, D.; Maire, I. Mucopolysaccharidosis
Type II—Genotype/Phenotype Aspects. Acta Paediatr. 2002, 91, 82–87.
49. Kim, C. H.; Hwang, H. Z.; Song, S. M.; Paik, K. H.; Kwon, E. K.; Moon, K. B.;
Yoon, J. H.; Han, C. K.; Jin, D.-K. Mutational Spectrum of the Iduronate-2-Sulfatase
Gene in 25 Unrelated Korean Hunter Syndrome Patients: Identification of 13 Novel
Mutations. Hum. Mutat. 2003, 21, 449–450.
50. Ricci, V.; Filocamo, M.; Regis, S.; Corsolini, F.; Stroppiano, M.; Duca, M. D.;
Gatti, R. Expression Studies of Two Novel in CIS-mutations Identified in an Interme-
diate Case of Hunter Syndrome. Am. J. Med. Genet. A 2003, 120A, 84–87.
51. Kato, T.; Kato, Z.; Kuratsubo, I.; Tanaka, N.; Ishigami, T.; Kajihara, J.-i.; Sukegawa-
Hayasaka, K.; Orii, K.; Isogai, K.; Fukao, T.; Shimozawa, N.; Orii, T.; Kondo, N.;
Suzuki, Y. Mutational and Structural Analysis of Japanese Patients With Muco-
polysaccharidosis Type II. J. Hum. Genet. 2005, 50, 395–402.
52. Sukegawa-Hayasaka, K.; Kato, Z.; Nakamura, H.; Tomatsu, S.; Fukao, T.; Kuwata, K.;
Orii, T.; Kondo, N. Effect of Hunter Disease (Mucopolysaccharidosis Type II) Muta-
tions on Molecular Phenotypes of Iduronate-2-Sulfatase: Enzymatic Activity, Protein
Processing and Structural Analysis. J. Inherit. Metab. Dis. 2006, 29, 755–761.
53. Froissart, R.; Da Silva, I. M.; Maire, I. Mucopolysaccharidosis Type II: An Update on
Mutation Spectrum. Acta Paediatr. 2007, 96, 71–77.
54. Chkioua, L.; Khedhiri, S.; Ferchichi, S.; Tcheng, R.; Chahed, H.; Froissart, R.; Vianey-
Saban, C.; Laradi, S.; Miled, A. Molecular Analysis of Iduronate-2-Sulfatase Gene in
Tunisian Patients With Mucopolysaccharidosis Type II. Diagn. Pathol. 2011, 6, 42.
55. Jin, P.; Hao, J.-W.; Chen, K.; Dong, C.-s.; Yang, Y.-B.; Mo, Z.-H. A 30 Splice Site
Mutation of IDS Gene in a Chinese Family With Mucopolysaccharidosis Type II. Gene
2013, 528, 236–240.
56. Brusius-Facchin, A. C.; Schwartz, I. V. D.; Zimmer, C.; Ribeiro, M. G.; Acosta, A. X.;
Horovitz, D.; Monlleó, I. L.; Fontes, M. I. B.; Fett-Conte, A.; Sobrinho, R. P. O.;
Duarte, A. R.; Boy, R.; Mabe, P.; Ascurra, M.; de Michelena, M.; Tylee, K. L.;
Besley, G. T. N.; Garreton, M. C. V.; Giugliani, R.; Leistner-Segal, S. Muco-
polysaccharidosis Type II: Identification of 30 Novel Mutations Among Latin
American Patients. Mol. Genet. Metab. 2014, 111, 133–138.
57. Galvis, J.; González, J.; Torrente, D.; Velasco, H.; Barreto, G. E. In Silico Analysis of
Iduronate 2 Sulfatase Mutations in Colombian Patients With Hunter Syndrome
(MPSII). In: Advances in Computational Biology. Advances in Intelligent Systems and Com-
puting; Castillo, L. F., Cristancho, M., Isaza, G., Pinzón, A., Rodrı́guez, J. M. C., Eds.;
Vol. 232, ;Springer: Cham, 2014; pp 205–212.
58. Tuschl, K.; Gal, A.; Paschke, E.; Kircher, S.; Bodamer, O. A. Mucopolysaccharidosis
Type II in Females: Case Report and Review of Literature. Pediatr. Neurol. 2005, 32,
270–272.
59. Jurecka, A.; Krumina, Z.; Żuber, Z.; Różdży nska-Świa˛tkowska, A.; Kłoska, A.;
Czartoryska, B.; Tylki-Szyma nska, A. Mucopolysaccharidosis Type II in Females
and Response to Enzyme Replacement Therapy. Am. J. Med. Genet. A 2012,
158A, 450–454.
ARTICLE IN PRESS

40 Shifaza Mohamed et al.

60. Lonardo, F.; Di Natale, P.; Lualdi, S.; Acquaviva, F.; Cuoco, C.; Scarano, F.;
Maioli, M.; Pavone, L. M.; Di Gregorio, G.; Filocamo, M.; Scarano, G. Muco-
polysaccharidosis Type II in a Female Patient With a Reciprocal X;9 Translocation
and Skewed X Chromosome Inactivation. Am. J. Med. Genet. A 2014, 164,
2627–2632.
61. Scarpa, M. Mucopolysaccharidosis Type II. In: GeneReviews® [Internet]; Adam, M. P.,
Ardinger, H. H., Pagon, R. A., Wallace, S. E., Bean, L. J. H., Stephens, K.,
Amemiya, A., Eds.; University of Washington: Seattle, WA, 2007.
62. Martin, R.; Beck, M.; Eng, C.; Giugliani, R.; Harmatz, P.; Muñoz, V.; Muenzer, J.
Recognition and Diagnosis of Mucopolysaccharidosis II (Hunter Syndrome).
Pediatrics 2008, 121, e377–e386.
63. Raluy-Callado, M.; Chen, W.-H.; Whiteman, D. A. H.; Fang, J.; Wiklund, I. The
Impact of Hunter Syndrome (Mucopolysaccharidosis Type II) on Health-Related
Quality of Life. Orphanet J. Rare Dis. 2013, 8, 101.
64. Jones, S. A.; Almássy, Z.; Beck, M.; Burt, K.; Clarke, J. T.; Giugliani, R.;
Hendriksz, C.; Kroepfl, T.; Lavery, L.; Lin, S.-P.; Malm, G.; Ramaswami, U.;
Tincheva, R.; Wraith, J. E. Mortality and Cause of Death in Mucopolysaccharidosis
Type II—A Historical Review Based on Data From the Hunter Outcome Survey
(HOS). J. Inherit. Metab. Dis. 2009, 32, 534–543.
65. Young, I. D.; Harper, P. S. The Natural History of the Severe Form of Hunter’s Syn-
drome: A Study Based on 52 Cases. Dev. Med. Child Neurol. 1983, 25, 481–489.
66. Scarpa, M.; Almássy, Z.; Beck, M.; Bodamer, O.; Bruce, I. A.; De Meirleir, L.;
Guffon, N.; Guillen-Navarro, E.; Hensman, P.; Jones, S.; Kamin, W.;
Kampmann, C.; Lampe, C.; Lavery, C. A.; Leão Teles, E.; Link, B.; Lund, A. M.;
Malm, G.; Pitz, S.; Rothera, M.; Stewart, C.; Tylki-Szyma nska, A.; van der
Ploeg, A.; Walker, R.; Zeman, J.; Wraith, J. E. Mucopolysaccharidosis Type II:
European Recommendations for the Diagnosis and Multidisciplinary Management
of a Rare Disease. Orphanet J. Rare Dis. 2011, 6, 72.
67. Young, I. D.; Harper, P. S.; Newcombe, R. G.; Archer, I. M. A Clinical and Genetic
Study of Hunter’s Syndrome. 2 Differences Between the Mild and Severe Forms.
J. Med. Genet. 1982, 19, 408–411.
68. Young, I. D.; Harper, P. S. Mild Form of Hunter’s Syndrome: Clinical Delineation
Based on 31 Cases. Arch. Dis. Child. 1982, 57, 828–836.
69. Guffon, N.; Heron, B.; Chabrol, B.; Feillet, F.; Montauban, V.; Valayannopoulos, V.
Diagnosis, Quality of Life, and Treatment of Patients With Hunter Syndrome in the
French Healthcare System: A Retrospective Observational Study. Orphanet J. Rare
Dis. 2015, 10, 43.
70. Young, I. D.; Harper, P. S. Psychosocial Problems in Hunter’s Syndrome. Child Care
Health Dev. 1981, 7, 201–209.
71. Link, B.; de Camargo Pinto, L. L.; Giugliani, R.; Wraith, J. E.; Guffon, N.; Eich, E.;
Beck, M. Orthopedic Manifestations in Patients With Mucopolysaccharidosis Type II
(Hunter Syndrome) Enrolled in the Hunter Outcome Survey. Orthop. Rev. 2010, 2, e16.
72. Norman-Tyler, F.; Fixsen, J. A.; Sharrad, W. J. Hunter’s Syndrome as a Cause of
Childhood Carpal Tunnel Syndrome: A Report of Three Cases. J. Pediatr. Orthop.
B 1995, 4, 106–109.
73. Goldberg, M. F.; Duke, J. R. Ocular Histopathology in Hunter’s Syndrome: Systemic
Mucopolysaccharidosis Type II. Archiv. Ophthalmol. 1967, 77, 503–512.
74. Topping, T. M.; Kenyon, K. R.; Goldberg, M. F.; Maumenee, A. Ultrastructural Ocu-
lar Pathology of Hunter’s Syndrome: Systemic Mucopolysaccharidosis Type II. Archiv.
Ophthalmol. 1971, 86, 164–177.
75. Ashworth, J. L.; Biswas, S.; Wraith, E.; Lloyd, I. C. Mucopolysaccharidoses and the
Eye. Surv. Ophthalmol. 2006, 51, 1–17.
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 41

76. Kim, S.; Yoo, Y. J.; Woo, S. J.; Yang, H. K. Early Retinal Changes in Hunter Syn-
drome According to Spectral Domain Optical Coherence Tomography. Korean J.
Ophthalmol. 2016, 30, 151–153.
77. Anawis, M. A. Hunter Syndrome (MPS II-B): A Report of Bilateral Vitreous Floaters
and Maculopathy. Ophthalmic Genet. 2006, 27, 71–72.
78. Muenzer, J.; Beck, M.; Eng, C. M.; Escolar, M. L.; Giugliani, R.; Guffon, N. H.;
Harmatz, P.; Kamin, W.; Kampmann, C.; Koseoglu, S. T.; Link, B.; Martin, R. A.;
Molter, D. W.; Muñoz Rojas, M. V.; Ogilvie, J. W.; Parini, R.; Ramaswami, U.;
Scarpa, M.; Schwartz, I. V.; Wood, R. E.; Wraith, E. Multidisciplinary Management
of Hunter Syndrome. Pediatrics 2009, 124, e1228–e1239.
79. Peck, J. E. Hearing Loss in Hunter’s Syndrome—Mucopolysaccharidosis II. Ear Hear.
1984, 5, 243–246.
80. Keilmann, A.; Nakarat, T.; Bruce, I. A.; Molter, D.; Malm, G. Hearing Loss in Patients
with Mucopolysaccharidosis II: Data From HOS—The Hunter Outcome Survey.
J. Inherit. Metab. Dis. 2012, 35, 343–353.
81. Lin, H.-Y.; Shih, S.-C.; Chuang, C.-K.; Lee, K.-S.; Chen, M.-R.; Lin, H.-C.;
Chiu, P. C.; Niu, D.-M.; Lin, S.-P. Assessment of Hearing Loss by Pure-tone Audiom-
etry in Patients With Mucopolysaccharidoses. Mol. Genet. Metab. 2014, 111, 533–538.
82. Hopkins, R.; Watson, J. A.; Jones, J. H.; Walker, M. Two Cases of Hunter’s
Syndrome—The Anaesthetic and Operative Difficulties in Oral Surgery. Br. J. Oral
Surg. 1973, 10, 286–299.
83. Downs, A. T.; Crisp, T.; Ferretti, G. Hunter’s Syndrome and Oral Manifestations:
A Review. Pediatr. Dent. 1995, 17, 98–100.
84. Brama, I.; Gay, I.; Feinmesser, R.; Springer, C. Upper Airway Obstruction in Hunter
Syndrome. Int. J. Pediatr. Otorhinolaryngol. 1986, 11, 229–235.
85. Sasaki, C. T.; Ruiz, R.; Kirchner, J. A.; Gaito, R. J.; Seshi, B. Hunter’s Syndrome:
A Study in Airway Obstruction. Laryngoscope 1987, 97, 280–285.
86. Yoskovitch, A.; Tewfik, T. L.; Brouillette, R. T.; Schloss, M. D.; Der
Kaloustian, V. M. Acute Airway Obstruction in Hunter Syndrome. Int. J. Pediatr.
Otorhinolaryngol. 1998, 44, 273–278.
87. Leighton, S. E. J.; Papsin, B.; Vellodi, A.; Dinwiddie, R.; Lane, R. Disordered Breath-
ing During Sleep in Patients With Mucopolysaccharidoses. Int. J. Pediatr.
Otorhinolaryngol. 2001, 58, 127–138.
88. Morehead, J. M.; Parsons, D. S. Tracheobronchomalacia in Hunter’s Syndrome. Int. J.
Pediatr. Otorhinolaryngol. 1993, 26, 255–261.
89. Shapiro, J.; Strome, M.; Crocker, A. C. Airway Obstruction and Sleep Apnea in Hurler
and Hunter Syndromes. Ann. Otol. Rhinol. Laryngol. 1985, 94, 458–461.
90. Orliaguet, O.; Pepin, J. L.; Veale, D.; Kelkel, E.; Pinel, N.; Levy, P. Hunter0 s Syn-
drome and Associated Sleep Apnoea Cured by CPAP and Surgery. Eur. Respir. J.
1999, 13, 1195–1197.
91. Muenzer, J.; Wraith, J. E.; Beck, M.; Giugliani, R.; Harmatz, P.; Eng, C. M.; Vellodi, A.;
Martin, R.; Ramaswami, U.; Gucsavas-Calikoglu, M.; Vijayaraghavan, S.; Wendt, S.;
Puga, A.; Ulbrich, B.; Shinawi, M.; Cleary, M.; Piper, D.; Conway, A. M.; Kimura, A.
A Phase II/III Clinical Study of Enzyme Replacement Therapy With Idursulfase in
Mucopolysaccharidosis II (Hunter Syndrome). Genet. Med. 2006, 8, 465–473.
92. Muenzer, J.; Gucsavas-Calikoglu, M.; McCandless, S. E.; Schuetz, T. J.; Kimura, A.
A Phase I/II Clinical Trial of Enzyme Replacement Therapy in Mucopolysaccharidosis
II (Hunter Syndrome). Mol. Genet. Metab. 2007, 90, 329–337.
93. Okuyama, T.; Tanaka, A.; Suzuki, Y.; Ida, H.; Tanaka, T.; Cox, G. F.; Eto, Y.; Orii, T.
Japan Elaprase® Treatment (JET) Study: Idursulfase Enzyme Replacement Therapy in
Adult Patients with Attenuated Hunter Syndrome (Mucopolysaccharidosis II, MPS II).
Mol. Genet. Metab. 2010, 99, 18–25.
ARTICLE IN PRESS

42 Shifaza Mohamed et al.

94. Muenzer, J.; Beck, M.; Eng, C. M.; Giugliani, R.; Harmatz, P.; Martin, R.;
Ramaswami, U.; Vellodi, A.; Wraith, J. E.; Cleary, M.; Gucsavas-Calikoglu, M.;
Puga, A. C.; Shinawi, M.; Ulbrich, B.; Vijayaraghavan, S.; Wendt, S.;
Conway, A. M.; Rossi, A.; Whiteman, D. A. H.; Kimura, A. Long-term, Open-labeled
Extension Study of Idursulfase in the Treatment of Hunter Syndrome. Genet. Med.
2011, 13, 95–101.
95. Dangel, J. H. Cardiovascular Changes in Children with Mucopolysaccharide Storage
Diseases and Related Disorders—Clinical and Echocardiographic Findings in
64 Patients. Eur. J. Pediatr. 1998, 157, 534–538.
96. Gross, D. M.; Williams, J. C.; Caprioli, C.; Dominguez, B.; Howell, R. R. Echocar-
diographic Abnormalities in the Mucopolysaccharide Storage Diseases. Am. J. Cardiol.
1988, 61, 170–176.
97. Rigante, D.; Segni, G. Cardiac Structural Involvement in Mucopolysaccharidoses.
Cardiology 2002, 98, 18–20.
98. Wraith, J. E.; Beck, M.; Giugliani, R.; Clarke, J.; Martin, R.; Muenzer, J. Initial
Report From the Hunter Outcome Survey. Genet. Med. 2008, 10, 508–516.
99. Kampmann, C.; Beck, M.; Morin, I.; Loehr, J. P. Prevalence and Characterization of
Cardiac Involvement in Hunter Syndrome. J. Pediatr. 2011, 159, 327–331.
100. Bhattacharya, K.; Gibson, S. C.; Pathi, V. L. Mitral Valve Replacement for
Mitral Stenosis Secondary to Hunter’s Syndrome. Ann. Thorac. Surg. 2005, 80,
1911–1912.
101. Kettles, D. I.; Sheppard, M.; Liebmann, R. D.; Davidson, C. Left Ventricular
Aneurysm, Aortic Valve Disease and Coronary Narrowing in a Patient With Hunter’s
Syndrome. Cardiovasc. Pathol. 2002, 11, 94–96.
102. Oda, H.; Sasaki, Y.; Nakatani, Y.; Maesaka, H.; Suwa, S. Hunter’s Syndrome. An
Ultrastructural Study of an Autopsy Case. Acta Pathol. Jpn. 1988, 38, 1175–1190.
103. Holt, J. B.; Poe, M. D.; Escolar, M. L. Natural Progression of Neurological Disease in
Mucopolysaccharidosis Type II. Pediatrics 2011, 127, e1258–e1265.
104. Ballenger, C. E.; Swift, T. R.; Leshner, R. T.; El Gammal, T. A.; McDonald, T. F.
Myelopathy in Mucopolysaccharidosis Type II (Hunter Syndrome). Ann. Neurol.
1980, 7, 382–385.
105. Koyama, K.; Moda, Y.; Sone, A.; Tanaka, H.; Hino, Y. Neurogenic Bladder in
Hunter’s Syndrome. J. Med. Genet. 1994, 31, 257–258.
106. Vinchon, M.; Cotten, A.; Clarisse, J.; Chiki, R.; Christiaens, J. L. Cervical Myelop-
athy Secondary to Hunter Syndrome in an Adult. Am. J. Neuroradiol. 1995, 16,
1402–1403.
107. Van Aerde, J.; Plets, C.; Van der Hauwaert, L. Hydrocephalus in Hunter Syndrome.
Acta Paediatr. Belg. 1981, 34, 93–96.
108. O’Brien, D. P.; Cowie, R. A.; Wraith, J. E. Cervical Decompression in Mild Muco-
polysaccharidosis Type II (Hunter Syndrome). Child. Nerv. Syst. 1997, 13, 87–90.
109. Parenti, G.; Meroni, G.; Ballabio, A. The Sulfatase Gene Family. Curr. Opin. Genet.
Dev. 1997, 7, 386–391.
110. Hanson, S. R.; Best, M. D.; Wong, C. H. Sulfatases: Structure, Mechanism, Biological
Activity, Inhibition, and Synthetic Utility. Angew. Chem. Int. Ed. 2004, 43,
5736–5763; (a) Demydchuk, M.; Hill, C. H.; Zhou, A.; Bunkóczi, G.; Stein, P. E.;
Marchesan, D.; Deane, J. E.; Read, R. J. Insights into Hunter Syndrome from the
Structure of Iduronate-2-sulfatase. Nat. Commun. 2017, 8, 15786.
111. Daniele, A.; Di Natale, P. The Active Site of the Enzyme Iduronate 2-Sulfate Sulfatase.
In Perspectives in Inherited Metabolic Diseases, vol. 7, Ermes, 1987; pp 85–93.
112. Bielicki, J.; Freeman, C.; Clements, P. R.; Hopwood, J. J. Human Liver Iduronate-2-
Sulphatase. Purification, Characterization and Catalytic Properties. Biochem. J. 1990,
271, 75–86.
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 43

113. von Figura, K.; Schmidt, B.; Selmer, T.; Dierks, T. A Novel Protein Modification
Generating an Aldehyde Group in Sulfatases: Its Role in Catalysis and Disease.
BioEssays 1998, 20, 505–510.
114. Appel, M. J.; Bertozzi, C. R. Formylglycine, A Post-Translationally Generated Res-
idue With Unique Catalytic Capabilities and Biotechnology Applications. ACS Chem.
Biol. 2015, 10, 72–84.
115. Boltes, I.; Czapinska, H.; Kahnert, A.; von B€ ulow, R.; Dierks, T.; Schmidt, B.; von
Figura, K.; Kertesz, M. A.; Usón, I. 1.3 Å Structure of Arylsulfatase from Pseudomonas
aeruginosa Establishes the Catalytic Mechanism of Sulfate Ester Cleavage in the Sulfatase
Family. Structure 2001, 9, 483–491.
116. Sáenz, H.; Lareo, L.; Poutou, R. A.; Sosa, Á. C.; Barrera, L. A. Computational Pre-
diction of the Tertiary Structure of the Human Iduronate 2-Sulfate Sulfatase. Biomedica
2007, 27, 7–20.
117. Burton, B. K.; Giugliani, R. Diagnosing Hunter Syndrome in Pediatric Practice: Prac-
tical Considerations and Common Pitfalls. Eur. J. Pediatr. 2012, 171, 631–639.
118. Giugliani, R.; Brusius-Facchin, A. C.; Moura de Souza, C. F.; Civallero, G.; Burin, M.;
Leistner-Segal, S.; Baldo, G.; Vairo, F. Diagnosis and Therapy Options in Muco-
polysaccharidosis II (Hunter Syndrome). Expert Opin. Orphan Drugs 2015, 3, 141–150.
119. Berry, H. K. Screening for Mucopolysaccharide Disorders With the Berry Spot Test.
Clin. Biochem. 1987, 20, 365–371.
120. Humbel, R.; Etringer, S. Colorimetric Method for the Determination of Sulfated Gly-
cosaminoglycans. Rev. Roum. Biochim. 1974, 11, 21–24.
121. Whitley, C. B.; Draper, K. A.; Dutton, C. M.; Brown, P. A.; Severson, S. L.;
France, L. A. Diagnostic Test for Mucopolysaccharidosis. II. Rapid Quantification
of Glycosaminoglycan in Urine Samples Collected on a Paper Matrix. Clin. Chem.
1989, 35, 2074–2081.
122. De Jong, J. G. N.; Wevers, R. A.; Laarakkers, C.; Poorthuis, B. J. H. M.
Dimethylmethylene Blue-Based Spectrophotometry of Glycosaminoglycans in
Untreated Urine: A Rapid Screening Procedure for Mucopolysaccharidoses. Clin.
Chem. 1989, 35, 1472–1477.
123. Blanchard, S.; Sadilek, M.; Scott, C. R.; Turecek, F.; Gelb, M. H. Tandem Mass Spec-
trometry for the Direct Assay of Lysosomal Enzymes in Dried Blood Spots: Application to
Screening Newborns for Mucopolysaccharidosis I. Clin. Chem. 2008, 54, 2067–2070.
124. Civallero, G.; Michelin, K.; de Mari, J.; Viapiana, M.; Burin, M.; Coelho, J. C.;
Giugliani, R. Twelve Different Enzyme Assays on Dried-blood Filter Paper Samples
for Detection of Patients with Selected Inherited Lysosomal Storage Diseases. Clin.
Chim. Acta 2006, 372, 98–102.
125. Dean, C. J.; Bockmann, M. R.; Hopwood, J. J.; Brooks, D. A.; Meikle, P. J. Detection
of Mucopolysaccharidosis Type II by Measurement of Iduronate-2-Sulfatase in Dried
Blood Spots and Plasma Samples. Clin. Chem. 2006, 52, 643–649.
126. Parkinson, E. J.; Muller, V.; Hopwood, J. J.; Brooks, D. A. Iduronate-2-Sulphatase
Protein Detection in Plasma From Mucopolysaccharidosis Type II Patients. Mol. Genet.
Metab. 2004, 81, 58–64.
127. Lin, S.-P.; Chang, J.-H.; Lee-Chen, G.-J.; Lin, D.-S.; Lin, H.-Y.; Chuang, C.-K.
Detection of Hunter Syndrome (Mucopolysaccharidosis Type II) in Taiwanese: Bio-
chemical and Linkage Studies of the Iduronate-2-Sulfatase Gene Defects in MPS II
Patients and Carriers. Clin. Chim. Acta 2006, 369, 29–34.
128. Lim, T. W.; Leder, I. G.; Bach, G.; Neufeld, E. F. An Assay for Iduronate Sulfatase
(Hunter Corrective Factor). Carbohydr. Res. 1974, 37, 103–109.
129. Hopwood, J. J. α-L-Iduronidase, β-D-Glucuronidase, and 2-Sulfo-L-iduronate
2-Sulfatase: Preparation and Characterization of Radioactive Substrates From Heparin.
Carbohydr. Res. 1979, 69, 203–216.
ARTICLE IN PRESS

44 Shifaza Mohamed et al.

130. Chamoles, N. A.; Blanco, M. B.; Gaggioli, D.; Casentini, C. Hurler-like Phenotype:
Enzymatic Diagnosis in Dried Blood Spots on Filter Paper. Clin. Chem. 2001, 47,
2098–2102.
131. Voznyi, Y. V.; Keulemans, J. L. M.; van Diggelen, O. P. A Fluorimetric Enzyme Assay
for the Diagnosis of MPS II (Hunter Disease). J. Inherit. Metab. Dis. 2001, 24, 675–680.
132. Keulemans, J. L. M.; Sinigerska, I.; Garritsen, V. H.; Huijmans, J. G. M.; Voznyi, Y. V.;
van Diggelen, O. P.; Kleijer, W. J. Prenatal Diagnosis of the Hunter Syndrome and the
Introduction of a New Fluorimetric Enzyme Assay. Prenatal Diagn. 2002, 22, 1016–1021.
133. Azadeh, M.; Pan, L.; Qiu, Y.; Boado, R. A Rapid Two-Step Iduronate-2-Sulfatatse
Enzymatic Activity Assay for MPSII Pharmacokinetic Assessment. JIMD Rep. 2018,
38, 89–95.
134. Sista, R.; Eckhardt, A. E.; Wang, T.; Sellos-Moura, M.; Pamula, V. K. Rapid, Single-
Step Assay for Hunter Syndrome in Dried Blood Spots Using Digital Microfluidics.
Clin. Chim. Acta 2011, 412, 1895–1897.
135. Wang, D.; Wood, T.; Sadilek, M.; Scott, C. R.; Turecek, F.; Gelb, M. H. Tandem
Mass Spectrometry for the Direct Assay of Enzymes in Dried Blood Spots: Application
to Newborn Screening for Mucopolysaccharidosis II (Hunter Disease). Clin. Chem.
2007, 53, 137–140.
136. Blanchard, S.; Turecek, F.; Gelb, M. H. Short Synthetic Sequence for 2-Sulfation of
α-L-Iduronate Glycosides. Carbohydr. Res. 2009, 344, 1032–1033.
137. Wolfe, B. J.; Blanchard, S.; Sadilek, M.; Scott, C. R.; Turecek, F.; Gelb, M. H. Tan-
dem Mass Spectrometry for the Direct Assay of Lysosomal Enzymes in Dried Blood
Spots: Application to Screening Newborns for Mucopolysaccharidosis II (Hunter Syn-
drome). Anal. Chem. 2011, 83, 1152–1156.
138. Spacil, Z.; Tatipaka, H.; Barcenas, M.; Scott, C. R.; Turecek, F.; Gelb, M. H. High-
Throughput Assay of 9 Lysosomal Enzymes for Newborn Screening. Clin. Chem. 2013,
59, 502–511.
139. Lee, K.; Jun, S.-H.; Song, S. H.; Park, H.-D.; Park, K. U.; Song, J. Direct Assay of
Iduronate-2-Sulfatase for Hunter Disease Using UPLC-Tandem Mass Spectrometry
and Fluorogenic Substrate. Clin. Biochem. 2015, 48, 1350–1353.
140. Chennamaneni, N. K.; Kumar, A. B.; Barcenas, M.; Spácil, Z.; Scott, C. R.;
Turecek, F.; Gelb, M. H. Improved Reagents for Newborn Screening of Muco-
polysaccharidosis Types I, II, and VI by Tandem Mass Spectrometry. Anal. Chem.
2014, 86, 4508–4514.
141. Kumar, A. B.; Masi, S.; Ghomashchi, F.; Chennamaneni, N. K.; Ito, M.; Scott, C. R.;
Turecek, F.; Gelb, M. H.; Spacil, Z. Tandem Mass Spectrometry Has a Larger Analyt-
ical Range than Fluorescence Assays of Lysosomal Enzymes: Application to Newborn
Screening and Diagnosis of Mucopolysaccharidoses Types II, IVA, and VI. Clin. Chem.
2015, 61, 1363–1371.
142. Tomatsu, S.; Montano, A. M.; Oguma, T.; Dung, V. C.; Oikawa, H.; Gutierrez, M. L.;
Yamaguchi, S.; Suzuki, Y.; Fukushi, M.; Barrera, L. A.; Kida, K.; Kubota, M.; Orii, T.
Validation of Disaccharide Compositions Derived From Dermatan Sulfate and Heparan
Sulfate in Mucopolysaccharidoses and Mucolipidoses II and III by Tandem Mass Spec-
trometry. Mol. Genet. Metab. 2010, 99, 124–131.
143. Oguma, T.; Tomatsu, S.; Montano, A. M.; Okazaki, O. Analytical Method for the
Determination of Disaccharides Derived From Keratan, Heparan, and Dermatan Sul-
fates in Human Serum and Plasma by High-Performance Liquid Chromatography/
Turbo Ionspray Ionization Tandem Mass Spectrometry. Anal. Biochem. 2007, 368,
79–86.
144. Fuller, M.; Chau, A.; Nowak, R. C.; Hopwood, J. J.; Meikle, P. J. A Defect in
Exodegradative Pathways Provides Insight Into Endodegradation of Heparan and
Dermatan Sulfates. Glycobiology 2006, 16, 318–325.
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 45

145. Pan, P.; Chen, M.; Zhang, Z.; Corte, A. D.; Souza, C.; Giugliani, R.; Pan, L.; Qiu, Y.;
Amaravadi, L.; Wu, J. A Novel LC-MS/MS Assay to Quantify Dermatan Sulfate in
Cerebrospinal Fluid as a Biomarker for Mucopolysaccharidosis II. Bioanalysis 2018,
10, 825–838.
146. Nielsen, T. C.; Rozek, T.; Hopwood, J. J.; Fuller, M. Determination of Urinary
Oligosaccharides by High-Performance Liquid Chromatography/Electrospray
Ionization-Tandem Mass Spectrometry: Application to Hunter Syndrome. Anal. Bio-
chem. 2010, 402, 113–120.
147. Saville, J. T.; McDermott, B. K.; Fletcher, J. M.; Fuller, M. Disease and Subtype Spe-
cific Signatures Enable Precise Diagnosis of the Mucopolysaccharidoses. Genet. Med.
2019, 21, 753–757.
148. Lawrence, R.; Brown, J. R.; Al-Mafraji, K.; Lamanna, W. C.; Beitel, J. R.;
Boons, G.-J.; Esko, J. D.; Crawford, B. E. Disease-Specific Non–Reducing End Car-
bohydrate Biomarkers for Mucopolysaccharidoses. Nat. Chem. Biol. 2012, 8, 197–204.
149. de Ruijter, J.; de Ru, M. H.; Wagemans, T.; Ijlst, L.; Lund, A. M.; Orchard, P. J.;
Schaefer, G. B.; Wijburg, F. A.; van Vlies, N. Heparan Sulfate and Dermatan Sulfate
Derived Disaccharides are Sensitive Markers for Newborn Screening for
Mucopolysaccharidoses Types I, II and III. Mol. Genet. Metab. 2012, 107, 705–710.
150. Lawrence, R.; Lu, H.; Rosenberg, R. D.; Esko, J. D.; Zhang, L. Disaccharide Structure
Code for the Easy Representation of Constituent Oligosaccharides From Glycosami-
noglycans. Nat. Methods 2008, 5, 291–292.
151. Trim, P. J.; Lau, A. A.; Hopwood, J. J.; Snel, M. F. A Simple Method for Early Age
Phenotype Confirmation Using Toe Tissue From a Mouse Model of MPS IIIA. Rapid
Commun. Mass Spectrom. 2014, 28, 933–938.
152. Auray-Blais, C.; Bherer, P.; Gagnon, R.; Young, S. P.; Zhang, H. H.; An, Y.;
Clarke, J. T.; Millington, D. S. Efficient Analysis of Urinary Glycosaminoglycans by
LC-MS/MS in Mucopolysaccharidoses Type I, II and VI. Mol. Genet. Metab. 2011,
102, 49–56.
153. Auray-Blais, C.; Lavoie, P.; Zhang, H.; Gagnon, R.; Clarke, J. T.; Maranda, B.;
Young, S. P.; An, Y.; Millington, D. S. An Improved Method for Glycosaminoglycan
Analysis by LC-MS/MS of Urine Samples Collected on Filter Paper. Clin. Chim. Acta
2012, 413, 771–778.
154. Zhang, H.; Young, S. P.; Auray-Blais, C.; Orchard, P. J.; Tolar, J.; Millington, D. S.
Analysis of Glycosaminoglycans in Cerebrospinal Fluid From Patients With Muco-
polysaccharidoses by Isotope-Dilution Ultra-Performance Liquid Chromatography-
Tandem Mass Spectrometry. Clin. Chem. 2011, 57, 1005–1012.
155. Trim, P. J.; Hopwood, J. J.; Snel, M. F. Butanolysis Derivatization: Improved
Sensitivity in LC-MS/MS Quantitation of Heparan Sulfate in Urine From Muco-
polysaccharidosis Patients. Anal. Chem. 2015, 87, 9243–9250.
156. Forni, G.; Malvagia, S.; Funghini, S.; Scolamiero, E.; Mura, M.; Della Bona, M.;
Villanelli, F.; Damiano, R.; la Marca, G. LC-MS/MS Method for Simultaneous Quan-
tification of Heparan Sulfate and Dermatan Sulfate in Urine by Butanolysis Derivati-
zation. Clin. Chim. Acta 2019, 488, 98–103.
157. He, Q. Q.; Trim, P. J.; Snel, M. F.; Hopwood, J. J.; Ferro, V. Synthesis and Mass Spec-
trometric Analysis of Disaccharides From Methanolysis of Heparan Sulfate. Org. Biomol.
Chem. 2018, 16, 8791–8803.
158. Gelb, M. H.; Scott, C. R.; Turecek, F. Newborn Screening for Lysosomal Storage Dis-
eases. Clin. Chem. 2015, 61, 335–346.
159. Kirkegaard, T. Emerging Therapies and Therapeutic Concepts for Lysosomal Storage
Diseases. Expert Opin. Orphan Drugs 2013, 1, 385–404.
160. Scarpa, M. Evaluation of Idursulfase for the Treatment of Mucopolysaccharidosis II
(Hunter Syndrome). Expert Opin. Orphan Drugs 2013, 1, 89–98.
ARTICLE IN PRESS

46 Shifaza Mohamed et al.

161. Chung, Y. K.; Sohn, Y. B.; Sohn, J. M.; Lee, J.; Chang, M. S.; Kwun, Y.; Kim, C. H.;
Lee, J. Y.; Yook, Y. J.; Ko, A.-R.; Jin, D.-K. A Biochemical and Physicochemical
Comparison of Two Recombinant Enzymes Used for Enzyme Replacement Therapies
of Hunter Syndrome. Glycoconjugate J. 2014, 31, 309–315.
162. Kim, C.; Seo, J.; Chung, Y.; Ji, H. J.; Lee, J.; Sohn, J.; Lee, B.; Jo, E. C. Comparative Study
of Idursulfase Beta and Idursulfase In Vitro and In Vivo. J. Hum. Genet. 2017, 62, 167–174.
163. Burrow, T. A.; Leslie, N. D. Review of the Use of Idursulfase in the Treatment of
Mucopolysaccharidosis II. Biol. Targets Ther. 2008, 2, 311–320.
164. Sohn, Y. B.; Cho, S. Y.; Park, S. W.; Kim, S. J.; Ko, A.-R.; Kwon, E.-K.; Han, S. J.;
Jin, D.-K. Phase I/II Clinical Trial of Enzyme Replacement Therapy With Idursulfase
Beta in Patients With Mucopolysaccharidosis II (Hunter Syndrome). Orphanet J. Rare
Dis. 2013, 8, 42.
165. Poswar, F.; Baldo, G.; Giugliani, R. Phase I and II Clinical Trials for the
Mucopolysaccharidoses. Expert. Opin. Investig. Drugs 2017, 26, 1331–1340.
166. Lu, J. Z.; Boado, R. J.; Hui, E. K. W.; Zhou, Q.-H.; Pardridge, W. M. Expression in
CHO Cells and Pharmacokinetics and Brain Uptake in the Rhesus Monkey of an IgG-
Iduronate-2-Sulfatase Fusion Protein. Biotechnol. Bioeng. 2011, 108, 1954–1964.
167. Boado, R. J.; Ka-Wai Hui, E.; Zhiqiang Lu, J.; Pardridge, W. M. Insulin Receptor
Antibody-Iduronate 2-Sulfatase Fusion Protein: Pharmacokinetics, Anti-drug Antibody,
and Safety Pharmacology in Rhesus Monkeys. Biotechnol. Bioeng. 2014, 111, 2317–2325.
168. Sonoda, H.; Morimoto, H.; Koshimura, Y.; Kinoshita, M. Correction of Central Nervous
System Deficits in the Mouse Model of Hunter Syndrome by Recombinant Iduronate
2-Sulfatase Crossing the Blood-Brain Barrier. Mol. Genet. Metab. 2017, 120, S125–S126.
169. Coutinho, M. F.; Santos, J. I.; Alves, S. Less Is More: Substrate Reduction Therapy for
Lysosomal Storage Disorders. Int. J. Mol. Sci. 2016, 17, 1065.
170. Ghiselli, G. Drug-Mediated Regulation of Glycosaminoglycan Biosynthesis. Med. Res.
Rev. 2017, 37, 1051–1094.
171. Friso, A.; Tomanin, R.; Salvalaio, M.; Scarpa, M. Genistein Reduces Glycosaminogly-
can Levels in a Mouse Model of Mucopolysaccharidosis Type II. Br. J. Pharmacol. 2010,
159, 1082–1091.
172. Marucha, J.; Tylki-Szyma nska, A.; Jakóbkiewicz-Banecka, J.; Piotrowska, E.; Kloska, A.;
Czartoryska, B.; Węgrzyn, G. Improvement in the Range of Joint Motion in Seven Patients
with Mucopolysaccharidosis Type II During Experimental Gene Expression-Targeted Iso-
flavone Therapy (GET IT). Am. J. Med. Genet. A 2011, 155, 2257–2262.
173. Delgadillo, V.; O’Callaghan Mdel, M.; Artuch, R.; Montero, R.; Pineda, M. Genistein
Supplementation in Patients Affected by Sanfilippo Disease. J. Inherit. Metab. Dis. 2011,
34, 1039–1044.
174. de Ruijter, J.; Valstar, M. J.; Narajczyk, M.; Wegrzyn, G.; Kulik, W.; Ijlst, L.;
Wagemans, T.; van der Wal, W. M.; Wijburg, F. A. Genistein in Sanfilippo Disease:
A Randomized Controlled Crossover Trial. Ann. Neurol. 2012, 71, 110–120.
175. Derrick-Roberts, A. L. K.; Jackson, M. R.; Pyragius, C. E.; Byers, S. Substrate Dep-
rivation Therapy to Reduce Glycosaminoglycan Synthesis Improves Aspects of Neu-
rological and Skeletal Pathology in MPS I Mice. Diseases 2017, 5, 5.
176. Roberts, A. L.; Rees, M. H.; Klebe, S.; Fletcher, J. M.; Byers, S. Improvement in
Behaviour After Substrate Deprivation Therapy with Rhodamine B in a Mouse Model
of MPS IIIA. Mol. Genet. Metab. 2007, 92, 115–121.
177. Masson, P.; Junien, J.-L. Use of Odiparcil in the Treatment of a Mucopolysacchardosis.
US Patent 9,561,246 B2 2017.
178. Chua, J. S.; Kuberan, B. Synthetic Xylosides: Probing the Glycosaminoglycan Biosyn-
thetic Machinery for Biomedical Applications. Acc. Chem. Res. 2017, 50, 2693–2705.
179. Toomey, J. R.; Abboud, M. A.; Valocik, R. E.; Koster, P. F.; Burns-Kurtis, C. L.;
Pillarisetti, K.; Danoff, T. M.; Erhardt, J. A. A Comparison of the β-D-Xyloside,
Odiparcil, to Warfarin in a Rat Model of Venous Thrombosis. J. Thromb. Haemost.
2006, 4, 1989–1996.
ARTICLE IN PRESS

Mucopolysaccharidosis type II (Hunter syndrome) 47

180. ClinicalTrials.gov NCT03370653. A Study in MPS VI to Assess Safety and Efficacy of


Odiparcil (iMProveS).
181. Boyd, R. E.; Lee, G.; Rybczynski, P.; Benjamin, E. R.; Khanna, R.; Wustman, B. A.;
Valenzano, K. J. Pharmacological Chaperones as Therapeutics for Lysosomal Storage
Diseases. J. Med. Chem. 2013, 56, 2705–2725.
182. St€utz, A. E.; Wrodnigg, T. M. Carbohydrate-Processing Enzymes of the Lysosome:
Diseases Caused by Misfolded Mutants and Sugar Mimetics as Correcting Pharmaco-
logical Chaperones. Adv. Carbohydr. Chem. Biochem. 2016, 73, 225–302.
183. 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.
184. Markham, A. Migalastat: First Global Approval. Drugs 2016, 76, 1147–1152.
185. Hoshina, H.; Shimada, Y.; Higuchi, T.; Kobayashi, H.; Ida, H.; Ohashi, T. Chaperone
Effect of Sulfated Disaccharide from Heparin on Mutant Iduronate-2-Sulfatase in
Mucopolysaccharidosis Type II. Mol. Genet. Metab. 2018, 123, 118–122.
186. Matassini, C.; Mirabella, S.; Goti, A.; Robina, I.; Moreno-Vargas, A. J.; Cardona, F.
Exploring Architectures Displaying Multimeric Presentations of a Trihydroxypiperidine
Iminosugar. Beilstein J. Org. Chem. 2015, 11, 2631–2640.
187. D’Adamio, G.; Matassini, C.; Parmeggiani, C.; Catarzi, S.; Morrone, A.; Goti, A.;
Paoli, P.; Cardona, F. Evidence for a Multivalent Effect in Inhibition of Sulfatases
Involved in Lysosomal Storage Disorders (LSDs). RSC Adv. 2016, 6, 64847–64851.
188. Matassini, C.; Vanni, C.; Goti, A.; Morrone, A.; Marradi, M.; Cardona, F. Multi-
merization of DAB-1 Onto Au GNPs Affords New Potent and Selective
N-Acetylgalactosamine-6-Sulfatase (GALNS) Inhibitors. Org. Biomol. Chem. 2018,
16, 8604–8612.
189. Vellodi, A. Bone Marrow Transplantation for Lysosomal Storage Disorders. Expert Rev.
Endocrinol. Metab. 2006, 1, 425–438.
190. Vellodi, A.; Young, E.; Cooper, A.; Lidchi, V.; Winchester, B.; Wraith, J. E. Long-
Term Follow-Up Following Bone Marrow Transplantation for Hunter Disease.
J. Inherit. Metab. Dis. 1999, 22, 638–648.
191. McKinnis, E. J. R.; Sulzbacher, S.; Rutledge, J. C.; Sanders, J.; Scott, C. R. Bone Mar-
row Transplantation in Hunter Syndrome. J. Pediatr. 1996, 129, 145–148.
192. Guffon, N.; Bertrand, Y.; Forest, I.; Fouilhoux, A.; Froissart, R. Bone Marrow Trans-
plantation in Children with Hunter Syndrome: Outcome After 7 to 17 Years. J. Pediatr.
2009, 154, 733–737.
193. Sawamoto, K.; Chen, H. H.; Almeciga-Diaz, C. J.; Mason, R. W.; Tomatsu, S. Gene
Therapy for Mucopolysaccharidoses. Mol. Genet. Metab. 2018, 123, 59–68.
194. Motas, S.; Haurigot, V.; Garcia, M.; Marco, S.; Ribera, A.; Roca, C.; Sanchez, X.;
Sanchez, V.; Molas, M.; Bertolin, J.; Maggioni, L.; Leon, X.; Ruberte, J.; Bosch, F.
CNS-Directed Gene Therapy for the Treatment of Neurologic and Somatic Muco-
polysaccharidosis Type II (Hunter Syndrome). JCI Insight 2016, 1 e86696.
195. Hinderer, C.; Katz, N.; Louboutin, J. P.; Bell, P.; Yu, H.; Nayal, M.; Kozarsky, K.;
O’Brien, W. T.; Goode, T.; Wilson, J. M. Delivery of an Adeno-Associated Virus Vec-
tor into Cerebrospinal Fluid Attenuates Central Nervous System Disease in Muco-
polysaccharidosis Type II Mice. Hum. Gene Ther. 2016, 27, 906–915.
196. Laoharawee, K.; Podetz-Pedersen, K. M.; Nguyen, T. T.; Evenstar, L. B.; Kitto, K. F.;
Nan, Z.; Fairbanks, C. A.; Low, W. C.; Kozarsky, K. F.; McIvor, R. S. Prevention of
Neurocognitive Deficiency in Mucopolysaccharidosis Type II Mice by Central Ner-
vous System-Directed, AAV9-Mediated Iduronate Sulfatase Gene Transfer. Hum.
Gene Ther. 2017, 28, 626–638.

You might also like