International Journal of Clinical Biochemistry and Research

Print ISSN: 2394-6369

Online ISSN: 2394-6377

CODEN : IJCBK6

International Journal of Clinical Biochemistry and Research (IJCBR) open access, peer-reviewed quarterly journal publishing since 2014 and is published under auspices of the Innovative Education and Scientific Research Foundation (IESRF), aim to uplift researchers, scholars, academicians, and professionals in all academic and scientific disciplines. IESRF is dedicated to the transfer of technology and research by publishing scientific journals, research content, providing professional’s membership, and conducting conferences, seminars, and award more...

  • Article highlights
  • Article tables
  • Article images

Article statistics

Viewed: 396

PDF Downloaded: 234


Get Permission Keerthiga M, Chakraborty, Silvia CR, Atre, and Shivakumar: Mucopolysaccharidoses: An overview and new treatment modalities


Introduction

Mucopolysaccharides also known as Glycosaminoglycans (GAGs), are negatively-charged polysaccharide compounds

Mucopolysaccharides or Glycosaminoglycans, are composed of uronic acid and amino sugar to form heteropolysaccharides which are unbranched. They were first secluded from mucin hence called mucopolysaccharides. They are predominantly present in extracellular compartment.1 Their functions within the body are widespread and determined by their molecular structure. GAGs play a key role in cell signaling, which regulates many biochemical processes. Some of these processes include regulation of cell growth and proliferation, promotion of cell adhesion, anticoagulation, and wound repair. The four primary groups of GAGs are classified based on their core disaccharide units and include heparin/heparan sulfate, chondroitin sulfate/dermatan sulfate, keratan sulfate, and hyaluronic acid.

Properties of glycosaminoglycans

The slippery nature of the mucous and synovial fluid is due to the presence of numerous negative charges on heteropolysaccharide chains making them repulse each other. This is responsible for them to revert to a hydrated state when the compression force is released, hence the pliant nature of synovial fluid and vitreous humor.1

Enzymes involved in glycosaminoglycans degradation

The enzymes are 3 exo – glycosidase (Alpha L- Iduronidase, beta – glucuronidase and beta hexosaminidase) and 2 sulfatase (iduronate -2- sulfatase and N – acetyl galactosamine -4- sulfatase).

Dermatan Sulfate Catabolism

Dermatan sulfate is made up of sulfated N- acetyl galactosamine alternating with uronic acid. Degradation of dermatan sulfate starts from a non-reducing end by the successive action of 3 exo – glycosidase (Alpha L- Iduronidase, beta–glucuronidase and beta-hexosaminidase) and 2 sulfatase (iduronate -2- sulfatase and N – acetyl galactosamine -4- sulfatase). Alpha – L- iduronidase also known as Hurler's corrective factor degrades Alpha – L – iduronic acid residues of dermatan and Heparan sulfate, a deficiency of which is seen in Hurler's syndrome.2 Alpha – L – Iduronidase is present in precursor form, which undergoes intracellular modification to a shorter form by losing 10 amino acids and contains Mannose – 6- phosphate marker for targeting lysosomes.3 Beta – glucuronidase detaches glucuronic acid in dermatan, chondroitin, and heparan sulfate. A deficiency of this enzyme is seen in Mucopolysaccharidosis VII (MPS VII). Beta – hexosaminidase present as 3 isoenzymes A, B, and S removes N- acetyl galactosamine from dermatan sulfate. The absence of isoenzymes A and B results only in a milder form of Sanfilippo syndrome, whereas frank Mucopolysaccharidosis is seen only when all 3 isoenzymes are absent. This suggests that isoenzyme S is involved in the degradation of dermatan sulfate in the absence of isoenzymes A and B.4 The enzyme deficient in MPS II, Iduronate sulfatase also known as Hunter corrective factor cleaves the sulfate group from L – iduronic acid present in dermatan sulfate. Removal of sulfur from N – acetlygalactosamine -4- sulfate is carried out by arylsulfatase B, the enzyme that is absent or deficient in Maroteaux – Lamy syndrome.

Heparan Sulfate Catabolism

Heparan sulfate is made up of glucuronic acid and L – iduronic acid, some of which are sulfated. Alpha – Iduronidase and beta–glucuronidase cleaves terminal L – iduronic acid and glucuronic acid respectively.5 Alpha – N – acetylglucosaminidase, the enzyme that is scarce in MPS III B, is required for the removal of N- acetyl glucosamine that is present in heparan sulfate.6 Iduronate sulfatase causes desulfation of 2- sulfated iduronic acid in heparan sulfate. Therefore, patients with alpha – L – Iduronidase deficiency (MPS I), beta – glucuronidase deficiency (MPS VII), or iduronate sulfatase deficiency (MPS II) will have defects in the degradation of both heparan sulfate and dermatan sulfate.

Keratan Sulfate Catabolism

Glycosaminoglycans that do not contain uronic acid are keratan sulfate. Instead of uronic acid, sulfated galactose alternates with sulfated N- acetyl glucosamine. Any alternations in the pathway of degradation of keratan sulfate lead to MPS IV. The restricted distribution of keratan sulfate in cartilage and cornea is responsible for its distinctive clinical features, compared to dermatan and heparan sulfate which have wide distribution. Beta–galactosidase removes galactose from non-reducing end of keratan sulfate. Complete absence of beta–galactosidase leads to GM 1 gangliosidosis whereas Morquio syndrome is due to mutations that mainly affect the catabolism of heparan sulfate. Beta – hexosaminidase cleaves terminal N- acetylglucosamine and its deficiency leads to Tay – Sach's and Sandhoff syndrome.7 Sulfatase removes 6- sulfate from galactose, its deficiency is seen in MPS IV. Apart from glycosaminoglycans, gangliosides GM2 and GM3 also concentrate in the brain parenchyma of MPS patients. Glycosaminoglycans accumulation leads to secondary inhibition of enzymes of gangliosides metabolism. Gangliosides accumulation is seen in the brains of patients with MPS I, MPS II (severe) and MPS III A, MPS III B, MPS III D and all of them show mental retardation, but gangliosides are not accumulated in the brain of patients with MPS I whose intelligence quotient is normal.

Mucopolysaccharidoses

Mucopolysaccharidoses (MPS) is a group of metabolic disorders caused due to deficiency or malfunctioning of lysosomal enzymes required for the degradation of glycosaminoglycans.1 Lysosome is a cell organelle containing various enzymes, which will process and degrade excess heteropolysaccharide chains to their simpler forms, therefore popularly known as a suicide bag. Inadequate or impaired production of these enzymes from lysosomes results in the accumulation of long heteropolysaccharide chains in various tissues. Such large accumulation will eventually cause cellular injury which hampers cell or organ function and its appearance and physical abilities. This condition which is caused due to decreased production or malfunctioning or absence of release of enzymes from lysosome is called lysosomal storage disorder. Based on the deficient enzyme, the catabolism of dermatan sulfate, keratan sulfate, chondroitin sulfate, or hyaluronate may be blocked. There is activation of an alternative pathway and products formed in this alternative pathway are excreted through urine. Each enzyme has unique genes and cDNA encoded which can be used for the production of recombinant enzymes and to identify mutation-causing diseases. Mutations of Mucopolysaccharidosis are heterogeneous, but specific populations have one / a few mutant alleles which are predominant. Except for MPS II which is X-linked, all other Mucopolysaccharidosis are Autosomal Recessive. Most Mucopolysaccharidosis have either point mutations or small changes in genes, whereas large deletion and major rearrangement occur in MPS II.8

MPS I (Hurler, Hurler – Scheie syndrome, Scheie)

MPS I is caused due to deficiency of alpha – L – Iduronidase and is characterized by a wide spectrum of manifestations with Hurler and Scheie syndrome representing extreme ends, whereas Hurler – Scheie syndrome represents intermediate severity. Classification of MPS I into 3 subtypes is based on clinical features, as these subtypes cannot be distinguished based on the diagnostic procedure (Table 1).

MPS I H (Hurler syndrome)

Hurler syndrome occupies the severe end of the clinical spectrum with multisystem involvement in a progressive manner. Diagnosis of a case of Hurler syndrome is usually made between 4-8 months, only umbilical hernia or inguinal hernia may be present at birth and as an infant, they are usually normal as it takes time for accumulation of glycosaminoglycans and cause cell injury. Deceleration of growth is seen between 6-8 months. Developmental delay begins in 12-24 months and is obvious by 2-4 years. Macroglossia first clues about medical recognition. Most children with this syndrome have limited language skills due to hearing loss, developmental delay, and macroglossia. Other features include recurrent ear, nose, and throat infections, coarse facial features, hepatosplenomegaly, and skeletal abnormalities. Most children with Hurler have recurrent upper respiratory tract infections, noisy breathing, and profuse nasal discharge. By the first year of age, corneal clouding becomes apparent. Cardiac complications and respiratory infections are common causes of death. Radiological features include a large skull, with untimely closure of sagittal and lambdoid sutures, thickening of the calvarium, shallow orbit, large – J – shaped sella, and atypical spacing of teeth with a dentigerous cyst, kyphosis with anterior hypoplasia of lumbar vertebrae, diaphyseal enlargement of long bones with irregular metaphyses. Coxa Volga, poorly formed pelvis with the short head of the femur. The ribs have characteristic oar – shape, flat at the sternal end and narrow at the vertebral end. Short phalanges are also seen.

MPS I H/S (Hurler/Scheie syndrome)

The features are intermediate between Hurler and Scheie syndrome. Symptoms start between 3-8 years. Deafness, corneal clouding, and skeletal abnormalities begin early to mid–teens. Micrognathia is a characteristic facial feature in patients with H-S syndrome. Accumulation of glycosaminoglycans in the spinal sheath leads to compression of the cervical cord, but intelligence is normal.

MPS I S (Scheie syndrome)

Scheie syndrome is the mildest form with onset of symptoms after 5 years of age and diagnosis is usually made between 10- 20 years of age. Corneal clouding, skeletal abnormalities, and coarse facial features are seen but stature and intelligence are normal. Skeletal abnormalities are more confined to hands causing claw–hand and carpal tunnel syndrome limiting daily functions. Apart from corneal clouding, glaucoma, and retinal degeneration are also seen causing visual impairment. Accumulation of glycosaminoglycans on valves and chordae tendinae leads to aortic valve stenosis and/or regurgitation.

MPS II (Hunter’s syndrome)

Based on clinical features they are recognized in milder and more severe form. MPS II is transmitted in an X – linked Recessive manner, therefore affected females are rare to find, however, there are few female patients with milder forms and these females could have some additional mutations, preventing the expression of normal alleles (Table 1).

MPS II (Severe form)

The onset of disease is between 2-4 years with progressive neurological deficit. Communicating hydrocephalus exacerbates neurological deficit with increased intracranial pressure after 7 years of age and is apparent by 10- 15 years. Corneas are characteristically clear but retinal detachment can be seen. Involvement of autonomic nervous system causing persistent diarrhea is troublesome to younger patients. Neurological complications are very severe in these patients with deterioration of central nervous system after 7-10 years of age. Cardiac failure due to myocardial thickening, valvular dysfunction, pulmonary hypertension, coronary narrowing is the common cause of death. Other atypical features include ptosis and early onset of seizures.

MPS II (Mild form)

Patients with a milder form of Hunter syndrome have normal intelligence and will survive into adulthood. Clinical features are similar to severe forms of MPS II but at a reduced rate of progression. Corneal papilledema is evident due to the deposition of glycosaminoglycans in the sclera, causing optic nerve compression.

MPS III (Sanfilippo syndrome)

MPS III is divided into four types based on enzyme deficiency which are required for the degradation of heparan sulfate. The enzyme deficiency in type A is N – sulfatase, alpha – N- acetlyglucosaminidase in type B, alpha – glucosaminide acetyltransferase in type C and N – acetylglucosamine – 6 – sulfatase in type D. Clinical features manifest by 2-6 years of age. Hyperactivity with wild behavior, developmental delay, hirsutism, coarse hair, and insomnia is presenting features. Coarse facial features are not prominent and some patients have normal features as adults. Stature is normal and skeletal deformities are mild. Due to poor articulation development of language skills is poor. Severe hearing loss is common. By 16 years of age, there is cortical atrophy leading to severe neurological deficit. Dementia, insomnia, poor attention span, hyperactivity, aggressive behavior, temper tantrums, and profound mental retardation is seen (Table 1).

MPS IV (Morquio syndrome)

The two types of MPS IV TYPE A and TYPE B are caused due to deficiency of enzymes of keratan sulfate metabolism, N- acetlygalactosamine-6–sulfatase and beta–galactosidase respectively. Both types are characterized by skeletal deformities resulting in dwarfism (short stature), with normal intelligence making them unique from other forms of MPS. Patients appear normal at birth. Short stature, genu valgum, kyphosis, and waddling gait with a tendency to fall frequently are early symptoms. Other skeletal abnormalities include platyspondylia, odontoid hypoplasia, hyperlordosis, kyphoscoliosis, ovoid deformities of vertebrae, ulnar deviation at the wrist joint, cubitus valgus, short Phalanges, and osteoporosis. Atlantoaxial subluxation is a unique feature of Morquio syndrome. Extra-skeletal manifestations include recurrent upper respiratory tract infection, small teeth, valvular disease, hearing impairment, and facial features like prognathism, and broad mouth (Table 1).

MPS VI (MaroteauxLamy syndrome)

N – acetylgalactosamine deficiency is responsible for MPS VI. Neurological development is normal whereas physical development is impaired. Presentation at birth would be macrocephaly, inguinal hernia, or Umbilical hernia. An obvious clouding of the cornea is seen. Growth is normal for the first few years of life but slowly deteriorate after 6 or 8 years. Restriction of movement at large joints results in a crouched stance. Hepatosplenomegaly is always present. The typical presentation of MPS VI includes patients with short stature, protuberant abdomen, and lumbar lordosis. Radiological changes are macrocephaly, ovoid shape vertebrae, acetabulum hypoplasia, and small flared iliac wings (Table 1).

MPS VII (Sly syndrome)

Patients with the sly syndrome have coarse facial features, protruding sternum, hepatosplenomegaly, umbilical/ inguinal hernia, thoracolumbar gibbus, and mild–moderate mental retardation. Granulocytes showing coarse metachromatic granules are striking. The severe forms of the Sly syndrome are characterized by hydrops foetalis, dysostosis multiplex and features of lysosomal storage disorder. The neonatal form itself is heterogeneous, ranging from death in utero to mild or no hydrops at birth. A review of other pregnancies among mothers of MPS VII patients suggests an increase number of spontaneous abortions. Most patients presenting beyond the neonatal period have increased urinary excretion of glycosaminoglycans. MPS VII patients who present as infants or young children have hepatosplenomegaly, Umbilical/inguinal hernia, short stature, repeated upper respiratory tract infection, pneumonia, and developmental delay.

Table 1

Recognition pattern of Mucopolysaccharidoses1

Clinical features

MPS lH

MPS lS

MPS ll

MPS lll

MPS IV

MPS Vl

MPS Vll

Common name

Hurler’s syndrome

Scheie syndrome

Hunter syndrome

Sanfilippo syndrome

Morquio syndrome

Maroteaux-Lamy syndrome

Sly syndrome

Mental deficiency

+

-

+

+

-

-

?

Coarse facial features

+

+

+

-

-

+

?

Corneal clouding

+

+

-

-

+

+

?

Visceromegaly

+

+

+

-

-

+

+

Short stature

+

+

+

+

+

+

+

Joint contractures

+

+

+

-

-

+

+

Dysostosis multiplex

+

+

+

+

+

+

+

Leucocyte inclusions

+

+

+

+

-

+

+

Mucopolysacchariduria

+

+

+

+

+

+

+

[i] (+): Clinical feature is present

[ii] (-): Clinical feature is absent

[iii] (?): Status not known

Diagnosis

Diagnosis is based on clinical examination, Mucopolysaccharides excreted in urine can also be detected in urine analysis. Deficient enzymes can be detected by enzyme assay which aids in definitive diagnosis. Nowadays prenatal tests like chorionic villus sampling and amniocentesis are becoming popular which identify the defective or mutated gene in the fetus. Genetic counseling plays a pivotal role in backstopping parents with a family history of Mucopolysaccharidosis.

Treatment

Symptomatic management

Early initiation of daily exercise, physiotherapy, and counseling helps in improving the quality of life. Dietary modification like restricting sugar, milk, and dairy products will not prevent the progression of disease but has helped people with excessive mucus. Polysomnography can be done to assess the requirements of nighttime oxygen. Tonsillectomy and adenoidectomy can also help to relieve obstructive airway pressure in individuals with obstructive sleep apnea. Shunting surgery helps to drain excess cerebrospinal fluid. Corneal transplantation can be used to tackle corneal clouding to improve vision.9

Table 2

Enzyme therapy with indication with their dilution and dosage

Enzyme

Indication for Administration

Dilution

Dosage

Children

Adult

Aldurazym

Mucopolysaccharidosis type I

0.9% sodium chloride containing 0.1% albumin

> 5 years - 0.58mg/ kg once weekly

Adults - 0.58 mg / kg once weekly

Elaprase

Mucopolysaccharidosis type II

0.9% sodium chloride.

>16 months – 0.5 mg/kg once a week

0.5mg/ kg once a week.

Naglazyme

Maroteaux-Lamy syndrome (MPSlV),

0.9% normal saline.

Infants: 1 or 2 mg / kg once weekly. Children and adolescents – 1 mg/ kg once weekly.

Bone marrow therapy / Hematopoietic stem cell therapy/ Umbilical cord blood transfusion

Although these procedures are risky with increased mortality and morbidity, if they are done at the right time before significant accumulation in neurons, then the benefits outweigh the risk of causing drastic changes in the natural history of disease and increasing life expectancy.10 Hematopoietic stem cell therapy consists of transferring stem cells from a healthy donor to a patient affected, if the procedure is successful, the donated stem cells begin to multiply and produce all the enzyme which was deficient or malfunctioning in the recipient. Except for skeletal and eye abnormalities, rest all physical abnormalities can be rectified. But coming to neurological outcomes it always shows varied results. Since these procedures are risky extensive counseling of family members is essential before initiation of the procedure.

Enzyme therapy

Nowadays enzyme replacement therapy is gaining more popularity amongst the treatment options available for Mucopolysaccharidosis, due to its ability to alleviate somatic symptoms. Enzyme replacement therapy is based on the principle of the production of recombinant enzymes in continuous human or animal cell lines. These specific enzymes produced by recombinant DNA technology will have mannose – 6- phosphate residue, which will bind to their respective mannose – 6- phosphate receptor present on the cell surface and gain entry into the cell, subsequently executing their action of catabolism on accumulated Glycosaminoglycans. This novel therapy is approved in various countries for patients with moderate to severe grades or for those with complications (Table 7). However, hematopoietic stem cell therapy remained the best choice for patients less than 2 years of age if a suitable donor is available. In such cases, enzyme replacement therapy is usually initiated in the period between diagnosis and transplantation to reduce the mortality and morbidity associated with transplantation.11, 12, 13, 14

Aldurazym

Table 3

Aldurazym infusion volume 100 ml for weight < 20 kg

ml/hour

Rate (mcg / kg/hour)

Duration in minutes

2

10

15

4

20

15

8

50

15

16

100

15

32

200

15

Table 4

Aldurazym nfusion volume 250 ml for weight > 20 kg

ml/hour

Rate (mcg/ kg/hour)

Duration in minutes

w 5

10

15

10

20

15

20

50

15

40

100

15

80

200

15

Infusion-related reactions are very common therefore pretreatment with antihistamines and antipyretics is very essential. If in case patients develop any Infusion-related reactions like rashes, fever, pruritis, angioedema, respiratory distress, or urticaria then immediately the rate of infusion should be decreased and antihistamines should be administered.15, 16, 17

Elaprase

Table 5

Total volume 100 ml

ml/ hour

Duration

8

15

16

15

32

15

64

15

Total volume should be infused over 3 hours starting at a rate of 8 ml/hour and incrementing every 15 minutes, but the rate should not exceed 100 ml/hour. Infusion-related reactions are common therefore pretreatment with antihistamines and antipyretics should be given.18

Naglazyme

Table 6

Total volume 250 ml

Rate (ml/hour)

Duration (hours)

6

1

80

1

80

1

80

1

A total volume of 250 ml should be infused over at least 4 hours, starting at a rate of 6 ml/hour for the first hour and if there are no infusion-related reactions, then infuse at 80 ml/hour for the next 3 hours. The total volume can be reduced to 100 ml if patients require fluid restrictions.19

Intervention to ease penetration of enzymes across blood-brain barrier

Trojan horse strategy

An alternate approach to facilitate enzymes to cross blood-brain barrier is to combine the enzyme with a genetically engineered molecule like an antibody, that binds to receptors present on blood-brain barrier and allows enzymes to enter the CNS through receptor-mediated transcytosis. This is known as the Trojan horse strategy.20 Clinical trials conducted on mice have been successful when Alpha – L- Iduronidase is fused with transferrin or apolipoprotein to cross blood-brain barrier. But for humans, it's still in phase 1 of clinical trials.21

Direct delivery of enzymes in cerebrospinal fluid

ERT can be delivered directly through intracerebroventricular injection, where the enzyme is injected into the lateral ventricle, or can also be injected intrathecally into subarachnoid space. Clinical trials in mice are found to be safe and effective, while study on humans is still going on.22

Gene therapy

Gene therapy has derived much attention due to its ability to provide a constant source of deficient enzymes. cDNA of the enzyme prepared by recombinant technique can be given either in vivo or ex vivo.23

The in vivo method includes the injection of a vector containing cDNA systemically or locally, resulting in the expression of enzymes by cells in the target tissue.

The viral vectors used in this technique were procured from adeno- associated viruses (AAV), lentiviruses, or retroviruses (Table 7).

Table 7

Routes of administration of vectors for Mucopolysaccharidosis

Vector

Route

Animal model

Outcomes

AAV2

Intrathecal

Adult mouse with MPSIIIB

• Enzyme activity increased • Glycosaminoglycans storage decreased • Improvement in behavioral deficits is seen 24

AAV2

Intracerebral

Adult mouse with MPS IIIB

• Enzyme activity increased • Glycosaminoglycans storage decreased. • Improvement in behavioral deficits is seen

AAV2/5

Intracerebral

Neonatal mouse with MPSIIIB

• Enzyme activity increased • Glycosaminoglycans storage decreased. • Improvement in behavioral deficits is not determined 25

AAV2/5

Intra-cerebroventrical

Neonate mouse with MPSIIIA

• Enzyme activity increased • Glycosaminoglycans storage decreased • Decrease in inflammation and neurodegeneration • Improvement in behavioral deficits is seen 25

AAV2/8

Intra-cerebroventrical

Adult mouse with MPSIIIA

•Enzyme activity increased • Glycosaminoglycans storage decreased • Decrease in inflammation and neurodegeneration • Improvement in behavioral deficits is seen 25

AAV4

Intra-cerebroventrical

Adult mouse with MPSVII

• Enzyme activity increased • Glycosaminoglycans storage decreased • Improvement in behavioral deficits is not determined[36]

AAV5

Intracerebral and intracerebroventrical

Adult mouse with MPS I H

• Enzyme activity increased • Glycosaminoglycans storage decreased • Improvement in behavioral deficits is not determined 25

AAV9

Intrathecal

Adolescent feline with MPS I

• Enzyme activity increased • Glycosaminoglycans storage decreased. • Improvement in behavioral deficits is not determined25

AAV9

Intracisternal

Adult mouse with MPS IIIA, IIIB, IIID

• Enzyme activity increased • Glycosaminoglycans storage decreased •Decrease inflammation and neurodegeneration seen • Improvement in behavioral deficits is seen 25

AAV9

Intravenous

Adult mouse with MPS I

• Enzyme activity increased • Glycosaminoglycans storage decreased • Improvement in behavioral deficits is not determined 25

AAV9/rh10

Intrathecal

Neonatal canine with MPS VII

• Enzyme activity increased • Glycosaminoglycans storage decreased • Decrease inflammation and neurodegeneration seen • Improvement in behavioral deficits is not determined 25

AAVrh10

Intracerebral

Adult mouse with MPS IIIA

• Enzyme activity increased • Glycosaminoglycans storage decreased •Decrease inflammation and neurodegeneration seen • Improvement in behavioral deficits is not determined 26

Lentiviral

Intra-cerebroventrical

Adult mouse with MPSIIIA

• Enzyme activity increased • Glycosaminoglycans storage decreased • Improvement in behavioral deficits is seen 27

The ex vivo technique involves the transfer of modified autologous cells, which will express the new cDNA deficit of early steps of the lysosomal degradation pathway. Genistein and Rhodamin B are the molecules used for substrate reduction therapies. Genistein is a tyrosine kinase inhibitor that inhibits signal transduction mediated by epidermal growth factor, responsible for expressing glycosaminoglycans synthesizing genes. The antioxidant and anti-inflammatory properties of genistein provide an additional advantage in decreasing proinflammatory cytokines.27 Rhodamin B is a nonspecific inhibitor of glycosaminoglycan synthesis.28

All these new modalities of treatment like direct delivery of enzymes into the cerebrovascular system, gene therapy, substrate reduction therapies, Nano-enabled therapy have shown improvement in neural symptoms to various extents, but all these studies have been conducted on animal models, while studies in human are still in progress.

Conclusion

Mucopolysaccharidoses is a group of disorders caused due to deficiency of lysosomal enzymes which are required for glycosaminoglycans breakdown. These glycosaminoglycans which are not degraded accumulate in various tissues and cause characteristic features. Bone marrow transplant became the first treatment to be successful. Over the last few decades, various other approaches have developed in addressing this problem. Hematopoietic stem cell therapy has gained popularity among all the new approaches because of its promising effects on deteriorating the natural history of disease and improving the quality of life. Enzyme replacement therapy which involves regular infusion of deficient enzymes is gradually becoming successful. It has shown improvement in pulmonary function, cardiovascular function, and organomegaly. However neurological improvement is poor due to the inability of enzymes to cross the blood brain barrier. This paved way for other new modalities trials like direct delivery of enzymes into the cerebrospinal fluid, gene therapy, substrate reduction therapy, and nanotherapy that succeeded in an animal model.

Source of Funding

None.

Conflict of Interest

None.

References

1 

J Casale JS Crane Biochemistry, Glycosaminoglycans [Internet]StatPearls PublishingTreasure Island (FL)2023

2 

W Wang L Shi Y Qin F Li Research and Application of Chondroitin Sulfate/Dermatan Sulfate-Degrading EnzymesFront Cell Dev Biol20208560442

3 

Y Zeng X He T Danyukova S Pohl AR Kermode Toward Engineering the Mannose 6-Phosphate Elaboration Pathway in Plants for Enzyme Replacement Therapy of Lysosomal Storage DisordersJ Clin Med20198122190

4 

P Awolade N Cele N Kerru L Gummidi E Oluwakemi P Singh Therapeutic significance of β-glucuronidase activity and its inhibitors: A reviewEur J Med Chem202018711192110.1016/j.ejmech.2019.111921

5 

Z Shriver I Capila G Venkataraman R Sasisekharan Heparin and heparan sulfate: analyzing structure and microheterogeneityHandb Exp Pharmacol201220715976PMCID

6 

G Birrane AL Dassier A Romashko D Lundberg K Holmes T Cottle Structural characterization of the α-N-acetylglucosaminidase, a key enzyme in the pathogenesis of Sanfilippo syndrome BJ Struct Biol201920536571

7 

AF Leal E Benincore-Flórez E Rintz AM Herreño-Pachón B Celik Y Ago Cellular Consequences of Glycosaminoglycans Accumulation and Potential TargetsInt J Mol Sci2022241477

8 

YF Huang S Mizumoto M Fujita Novel Insight Into Glycosaminoglycan Biosynthesis Based on Gene Expression ProfilesFront Cell Dev Biol2021970901810.3389/fcell.2021.709018

9 

P Hegerová Z Dědková L Sobotka Early nutritional support and physiotherapy improved long-term self-sufficiency in acutely ill older patientsNutrition201531116670

10 

M Taylor S Khan M Stapleton J Wang J Chen R Wynn Hematopoietic stem cell transplantation for mucopolysaccharidoses: past, present, and futureBiol Blood Marrow Transplant201925722646

11 

MD Poe SL Chagnon ML Escolar Early treatment is associated with improved cognition in Hurler syndromeAnn Neurol201476574753

12 

LA Clarke JE Wraith M Beck EH Kolodny GM Pastores J Muenzer Long-term efficacy and safety of laronidase in the treatment of mucopolysaccharidosis IPediatrics2009123122940

13 

E Jameson S Jones T Remmington Enzyme replacement therapy with laronidase (Aldurazyme®) for treating mucopolysaccharidosis type ICochrane Database Syst Rev201966CD00935410.1002/14651858.CD009354.pub5

14 

D Concolino F Deodato R Parini Enzyme replacement therapy: efficacy and limitationsItal J Pediatr201844Suppl 2120

15 

MC Cáceres J Guerrero-Martín D Pérez-Civantos P Palomo-López JI Delgado-Mingorance N Durán-Gómez The importance of early identification of infusion-related reactions to monoclonal antibodiesTher Clin Risk Manag20191596577

16 

J Muenzer O Bodamer B Burton L Clarke GS Frenking R Giugliani The role of enzyme replacement therapy in severe Hunter syndrome-an expert panel consensusEur J Pediatr201217111818

17 

EMK daSilva MWL Strufaldi RB Andriolo LA Silva Enzyme replacement therapy with idursulfase for mucopolysaccharidosis type II (Hunter syndrome)Cochrane Database Syst Rev201620162CD008185

18 

Naglazyme FDA Approval History https://www.drugs.com/history/naglazyme.html[Cited 2023 Jul 1]

19 

P Harmatz C B Whitley L Waber R Pais R Steiner B Plecko Enzyme replacement therapy in mucopolysaccharidosis VI (Maroteaux-Lamy syndrome)J Pediatr2004144557480

20 

DJ Begley CC Pontikis M Scarpa Lysosomal storage diseases and the blood-brain barrierCurr Pharm Des20081416156680

21 

P Dickson M Mcentee C Vogler S Le B Levy M Peinovich Intrathecal enzyme replacement therapy: successful treatment of brain disease via the cerebrospinal fluidMol Genet Metab2007911618

22 

EL Aronovich PB Hackett Lysosomal storage disease: Gene therapy on both sides of the blood-brain barrierMol Genet Metab201511428393

23 

CD Heldermon KK Ohlemiller ED Herzog C Vogler E Qin DF Wozniak Therapeutic Efficacy of Bone Marrow Transplant, Intracranial AAV-mediated Gene Therapy, or Both in the Mouse Model of MPS IIIBMol Ther201018587380

24 

LK Winner H Beard S Hassiotis AA Lau AJ Luck JJ Hopwood A preclinical study evaluating AAVrh10-based gene therapy for Sanfilippo syndromeHum Gene Ther201627536375

25 

C Mcintyre AL Derrick-Roberts S Byers DS Anson Correction of murine mucopolysaccharidosis type IIIA central nervous system pathology by intracerebroventricular lentiviral-mediated gene deliveryJ Gene Med20141611-1237487

26 

PJ Gaillard CC Visser AG deBoer Targeted delivery across the blood-brain barrierExpert Opin Drug Deliv200522299309

27 

G Liu I Martins JA Wemmie JA Chiorini BL Davidson Functional correction of CNS phenotypes in a lysosomal storage disease model using adeno-associated virus type 4 vectorsJ Neurosci2005254193217

28 

M Salvalaio L Rigon D Belletti F D ’avanzo F Pederzoli F Ruozi Targeted polymeric nanoparticles for brain delivery of high molecular weight molecules in lysosomal storage disordersPloS One2016115e0156452



jats-html.xsl


This is an Open Access (OA) journal, and articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.

Article type

Review Article


Article page

101-109


Authors Details

Keerthiga M, Arpita Chakraborty, Wilma Delphine Silvia CR*, Akshay Atre, Preksha Shivakumar


Article History

Received : 19-06-2023

Accepted : 03-07-2023


Article Metrics


View Article As

 


Downlaod Files