48
Views
1
CrossRef citations to date
0
Altmetric
Review

α-L-iduronidase therapy for mucopolysaccharidosis type I

&
Pages 743-751 | Published online: 05 Dec 2008

Abstract

More than 500 patients with mucopolysaccharidosis type IH (MPS IH; Hurler syndrome) have been treated with hematopoietic cell transplantation (HCT) throughout the world since the introduction of transplantation as therapy almost 30 years ago. More recently, the availability of recombinant α-L-iduronidase (IDUA) has resulted in the widespread treatment of less severe forms of MPS I with enzyme replacement therapy (ERT). In addition, over 50 MPS IH patients have been treated with a combination of ERT and HCT. The rationale for both ERT and HCT stems from the pivotal experiments performed 4 decades ago that showed α-L-iduronidase supplied in the environment can correct the accumulation of substrate in MPS I cells. Our purpose is to address the multiple applications associated with the therapeutic delivery of IDUA: intermittent delivery of recombinant protein (ERT), continuous administration through cellular therapy (HCT), the use of other stem cells or, potentially, correction of the enzyme defect itself through gene therapy approaches. Even though gene therapy and non-hematopoietic stem cell approaches, have yet to be tested in a clinical setting, it is possible that all these approaches will in the near future be a part of a paradigm shift from unimodal to multimodal therapy for MPS I.

Enzyme replacement therapy

Mucopolysaccharidoses are autosomal recessive disorders characterized by deficiencies of lysosomal hydrolases needed for the step-wise catabolism of complex carbohydrates termed glycosaminoglycans (GAG) (CitationNeufeld 1991). For each disease, the specific enzyme deficiency defines which substrates accumulate, and the tissues which are primarily affected. In mucopolysaccharidosis type I (MPS I), the deficiency in α-L-iduronidase (IDUA) results in lysosomal accumulation of GAG heparan and dermatan sulfate (CitationBach et al 1972). Abnormal accumulation of these GAG leads to progressive cellular and multi-organ dysfunction.

The clinical phenotype of children with severe (ie, complete) deficiency of IDUA may be apparent at birth, but in the majority of cases these initial symptoms and signs may not be evident until at 6 to 8 months of age, and the diagnosis may not be made until some months later. Typically, the clinical findings may include hepatosplenomegaly, umbilical or inguinal hernia, evidence of cardiac disease (coronary artery disease or valvular abnormalities), obstructive airway disease, corneal clouding and retinal degeneration, chronic rhinitis and otitis, hydrocephalus, progressive neurocognitive deterioration, and multiple musculoskeletal abnormalities. Without treatment, early death is observed, usually between 5 and 10 years of age (CitationNeufeld 1991; CitationOrchard et al 2007).

In addition to patients with severe MPS I described above, two other clinical syndromes exist that are characterized by less severe deficiencies of IDUA; Hurler-Scheie syndrome with an intermediate phenotype and Scheie syndrome with a mild phenotype. In contrast to patients with Hurler syndrome, the patients with Hurler-Scheie and Scheie syndromes have mild or no cognitive impairment. While generally the signs and symptoms of these more attenuated disorders occur later and are less severe than in patients affected with Hurler syndrome, significant medical problems may be seen paralleling what is seen in the Hurler patients.

While traditionally labeled as three distinct entities, Hurler (frequency 1:100,000 live births), Hurler-Scheie (1:115,000 live births), and Scheie (1:500,000 live births) syndromes represent a progressive clinical continuum, with the three phenotypes not necessarily clearly delineated (CitationRoubicek et al 1985; CitationScott et al 1995; CitationMeikle et al 1999). This reflects the substantial heterogeneity in the genotypes encountered in the IDUA gene, and may reflect other modifying genes affecting the severity of disease (CitationPastores et al 2007).

The IDUA gene is localized to chromosomal band 4p16.3. The total length of the gene is 19 kilobases and includes 14 exons. In wild type form, it codes for 653 amino acids and leads to the production of a 82 kDa protein (CitationSchuchman et al 1984; CitationUnger et al 1994). Many IDUA mutations are private but some exhibit a significant bias in different populations (CitationLee-Chen et al 1999; CitationLee-Chen and Wang 1997; CitationRempel et al 2005). For example, approximately 70% of patients with European ancestry carry one of the two severe mutations, W402X or Q70X, yet the spectrum of the MPS I syndromes presents a significant challenge in defining treatment (CitationScott et al 1992, Citation1993; CitationSugawara et al 2008).

Since the 1980s HCT has proven to be a life-saving measure for the severe form of MPS IH (CitationHobbs et al 1981; CitationOrchard et al 2007). The significant toxicities associated with HCT, however, have made its application in less severe forms (Hurler-Scheie and Scheie syndrome) difficult to justify.

The availability of recombinant IDUA (laronidase) for utilization as enzyme replacement therapy (ERT) has been a major advancement in the field. ERT has become a mainstay of therapy for patients with Hurler-Scheie and Scheie, the attenuated forms of IDUA deficiency that do not have central nervous system manifestations of their disease. As the intravenous administration of the enzyme has been shown to not effectively cross the blood brain barrier, the treatment of severe MPS I deficiency with enzyme replacement alone has not been accepted; in the Hurler population, HCT remains the standard of care. However, one major consequence of the success of ERT in MPS I has been the potential addition of ERT prior to transplantation in, patients with Hurler syndrome.

As a prerequisite for the human studies, the infusion of enzyme as therapy has been evaluated in animal models of MPS I (CitationKakkis et al 1996, Citation2001b; CitationClarke et al 1997; CitationOhmi et al 2003). Two murine models with genetically engineered IDUA deficiency, as well as feline and canine MPS I models, exist. Even though they are not full phenocopies of human disease, all mimic the human MPS I disorder biochemically (increased GAG levels in tissues and in urine) and in the development of visceral and central nervous system pathology. Studies in these animal models showed that both accumulation of GAG and GAG-mediated pathological features can be alleviated by treatment with systemic recombinant enzyme, and suggested that a benefit could be expected by ERT in humans (CitationShull et al 1994; CitationKakkis et al 1996, Citation2001b).

The recombinant IDUA, available for clinical use since 2001, is a polymorphic variant of the human enzyme produced by recombinant DNA technology in the Chinese hamster ovary cell line (CitationKakkis et al 1994). In the study that defined the ERT field, CitationKakkis et al (2001a) treated 10 patients from 5 to 22 years of age with MPS I with recombinant human IDUA at a dose of 125,000 units per kg of body weight given intravenously once weekly for 52 weeks. The authors concluded that the therapy reduced lysosomal storage of GAG and ameliorated some of the clinical manifestations of MPS I, with effects on growth, restrictions of range of motion at the shoulder and elbow, and sleep apnea. In addition, urinary GAG, a rough measure of total body GAG load, decreased after 3 to 4 weeks of IDUA treatment. Approximately half the patients had urticaria during IDUA infusion, but in general the ERT was tolerated well. Importantly, serum antibodies to IDUA were detected in 4 out of 10 patients, but this did not appear to affect the response to therapy (CitationKakkis et al 2001a). A follow-up study concluded that the patients continued to exhibit an improved ability to perform normal daily activities (CitationSifuentes et al 2007). As such, these data demonstrate in a compelling fashion an opportunity for long-term alteration of morbidity associated with the attenuated forms of MPS I.

Since that time, additional information has been provided in regards to the treatment of MPS I with enzyme therapy (CitationWraith 2001). Foremost among these is a placebo-controlled, randomized, double-blind study of CitationWraith et al (2004). Fortyfive MPS I patients were evaluated, 22 of whom received 0.58 mg/kg (100 units/kg) of intravenous IDUA weekly for 26 weeks, and 23 of whom received a placebo. It was shown that IDUA was well tolerated, and reduced urinary GAG. Critically, statistically significant improvement was documented in the ability to perform physical tasks and in the respiratory capacity in the IDUA-treated group compared to placebo-treated patients (CitationWraith et al 2004).

The same group has recently published a large, multinational, open-label study of 20 patients with MPS I who were less than 5 years old (CitationWraith et al 2007). Consistent with the previous report, ERT was again shown to be well tolerated, with a decrease in GAG storage (as evidenced by decreased hepatomegaly and GAG urinary clearance), and to provide clinical benefits such as improvement in sleep apnea and hypopnea to patients with severe MPS I in this early age group. Interestingly, a more robust decrease in GAG total body load was observed in patients who were receiving a higher dose of IDUA (200 units/kg vs 100 units/kg), and those with low anti-IDUA antibody levels.

These observations are relevant since IDUA therapy may be optimal at higher doses than those currently used. In addition, there is a suggestion that the immunologic response to the drug can be a formidable barrier to both ERT and HCT. Therefore, a central question in MPS I therapy is how completely IDUA will correct the deficiency in various organ systems, and how generation of anti-IDUA antibodies in immunocompetent hosts will impact the efficacy of serial, indeed life-long, IDUA infusions.

Intuitively, one would expect that IDUA naïve (ie, IDUAnull MPS I/Hurler syndrome) recipients would develop an immune response to systemically infused enzyme. In fact, this has been observed in both animal and human recipients of recombinant enzyme (CitationKakkis et al 1996; CitationShull et al 1996; CitationTurner et al 2000; CitationGlaros et al 2002). In an effort to minimize the antibody response, induction of immune tolerance in canine MPS I model with a regimen consisting of cyclosporine and azathioprine has been tested and proven to be successful (CitationKakkis et al 2004a).

Even more relevant to human disease has been the study of CitationKakavanos et al (2003) who observed that the patients who had an IDUA-specific immune reaction initially, developed immune tolerance after 2 years of treatment with IDUA. As accumulating data indicate that almost all ERT-treated patients develop anti-IDUA antibodies, it is important to recognize that in the majority of cases IDUA-directed antibodies appear to be non-neutralizing, and as such would not be expected to affect the active site of the enzyme on which the beneficial effects of ERT depend.

Even though most available studies have demonstrated that ERT is generally safe and efficacious, ultimately the long-term impact of ERT is unknown (CitationComan et al 2008). This is in part because of immune responses that may prove important over long periods of enzyme administration. It is also because the effects of intermittent ERT on different tissues are likely to be heterogeneous in response to therapy (eg, brain, bone, and heart valves are likely to be more resistant to ERT), and because how these responses are maintained over years of therapy is unknown. Based on these limitations, plus the high cost of ERT, the need for weekly infusions over a lifetime, and the possibility of immune-based reactions that may complicate therapy, the critical question is: should cellular therapy be used alone or integrated as multimodal therapy with ERT to best provide optimal correction of IDUA deficiency?

Cellular therapy

The rationale for HCT originated from studies of Neufeld et al which showed that “cross correction” of IDUAnull cells with externally supplied IDUA is possible (CitationFratantoni et al 1968a, Citationb; CitationBach et al 1972; CitationNeufeld 1991). IDUA secreted by donor cells is modified by the cellular mechanisms with the addition of mannose-6-phosphate residue. After secretion, glycosylated IDUA is taken up by recipient IDUAnull cells via mannose-6-phosphate receptors present on the cellular and subcellular membranes of the recipient cells (CitationHasilik and Neufeld 1980). After internalization, the enzyme is channeled into lysosomes where it takes part in degradation of GAG (CitationFischer et al 1980). Therefore, as functional IDUA is provided by donor cells, HCT has the ability to provide the enzyme in a manner akin to ERT, except on a continuous rather than intermittent basis.

Allogeneic stem cell transplantation for MPS I was first performed by Hobbs et al and was quickly followed by others as a life-saving measure for children with severe MPS I (CitationHobbs et al 1981; CitationPeters et al 1996, Citation1998a, Citation1998b; CitationVellodi et al 1997; CitationSouillet et al 2003; CitationHansen et al 2008). Heterozygous to normal serum levels of IDUA are routinely achieved after HCT, and the progression of this debilitating disease is arrested in most, but not all, organ systems. Initially, only human leukocyte antigen (HLA) matched related donors were used in order to minimize the immune-related complications of HCT, specifically graft-versus-host disease (GVHD). However, as most affected children do not have an HLA-matched related donor, we and others have explored the potential of alternative donor transplantation using unrelated HLA-matched donors or cord blood grafts. Over the last 3 decades, conditioning regimens, HLA typing techniques and the ability to identify well matched stem cell grafts for HCT has been improved greatly, and HCT remains the treatment method of choice for the severe form of MPS I (CitationStaba et al 2004; CitationBoelens et al 2007; CitationOrchard et al 2007).

Several important concerns, however, remain. First, graft failure has been observed in as many as one third of MPS I patients treated with HCT (CitationGrewal et al 2002). In a large recent European study of outcomes of HCT for Hurler syndrome, CitationBoelens et al (2007) reported more graft failure with T-cell depletion and reduced intensity conditioning, and less graft failure when cord blood versus bone marrow has been used as a graft source. This is consistent with findings of CitationStaba et al (2004) who reported favorable outcomes of MPS IH (MPS I Hurler syndrome) in recipients of cord blood HCT. Second, the conditioning regimen has been an important determinant of HCT-related complications, which include infection, immune injury such as GVHD, or direct injury to tissues such as the lung from chemotherapy and radiation. Furthermore, late effects after HCT include increased risks of cancer, sterility, effects on growth, and chronic GVHD.

Combination therapy

Unfortunately, some complications of HCT appear more common in MPS IH patients. In addition to graft failure discussed above, respiratory difficulties have been frequently observed, and many MPS IH children require mechanic ventilation in the peri-transplant period. These complications include lower respiratory infections, idiopathic pulmonary syndrome and, one of the most difficult complications to effectively treat, pulmonary hemorrhage (CitationGassas et al 2003; CitationTolar et al 2008). As the upper airway is typically compromised in MPS IH children due to GAG deposition in oral, nasal and laryngeal tissues, these patients may be at greater risk for intubation and prolonged ventilation, which in turn increases the risk of multiorgan failure and poor outcome of HCT.

Thus, we and others reasoned that ERT in peri-transplant period has a potential to decrease GAG accumulation in lung and other visceral tissues prior to transplantation, which in turn may decrease the risk of life-threatening complications during HCT in MPS IH patients (CitationTolar et al 2007; CitationCox-Brinkman et al 2006).

Among the first communications of this approach was a multi-institutional report of 12 MPS IH recipients of combined therapy (ERT and HCT) by CitationGrewal et al (2005). The median duration of ERT was 12 weeks before HCT and 7 weeks after HCT. Eight patients had complete donor engraftment and 11 of 12 survived at a median time of follow-up of 3 months (1 child died of pulmonary hemorrhage). Based on these case studies, it was concluded that the ERT and HCT combination therapy is safe and feasible, but prospective studies were needed to further evaluate the outcomes of co-modality therapy (CitationGrewal et al 2005).

The European study lead by CitationCox-Brinkman et al (2006) also observed that ERT with HCT has been well tolerated. Nineteen of 22 patients studied were surviving (2 patients died after HCT repeated for graft failure). The generation of anti-IDUA antibodies has not been correlated with development of graft failure, but overall no benefit of ERT for this population of patients could be documented. Thus, these authors concluded that only the patients in poor clinical condition would benefit from ERT before HCT.

Of note, the patient group described in this study was heterogeneous: patients came from multiple institutions, were recipients of various stem cell grafts (bone marrow, peripheral blood stem cells, cord blood, matched and mismatched family donors, and unrelated donors), were transplanted after variable conditioning regimens (4 different myeloablative regimens, 1 reduced intensity regimen), and received a varied number of stem cell grafts (approximately one third received second transplants and about 10% received a third HCT) (CitationCox-Brinkman et al 2006).

In light of this, we chose to investigate prospectively the combination of ERT and HCT, utilizing a single conditioning regimen, receiving a similar number of enzyme infusions, and providing consistent supportive care in a single transplant center (CitationTolar et al 2007). Seven patients were studied at a median age of 1.5 years at the time of myeloablative HCT. Before HCT, 5 had pulmonary complications (pneumonia with hospitalization, abnormal sleep study, reactive early disease, and oxygen needs), which would be expected to increase their risk of complex HCT-related complications significantly. The patients were treated with either cord blood transplants or related-donor bone marrow. All survived and their outcomes appeared superior to those of patients with similar number of pulmonary risk factors who were treated with HCT alone.

Importantly, we confirmed the findings of others who showed that anti-IDUA antibodies (which develop in most if not in all of the patients) did not appear to compromise donor engraftment. Theoretically, it is possible that pre-HCT clearance of GAG by ERT from bone marrow stroma could make the recipients’ bone marrow niche a more permissive environment for the engraftment of donor cells. Therefore, we favor the interpretation that all patients with severe MPS I, regardless of their clinical condition prior to transplant, could benefit from combination therapy with ERT and HCT, and this co-modality therapy is offered to all families undergoing transplantation for MPS IH at our institution (CitationTolar et al 2007).

Taken together, the cumulative data reflecting the results of ERT and HCT co-modality therapy indicate that it is a safe and effective regimen which is now gaining wide acceptance. Despite the apparent benefits of ERT + HCT therapy, however, little is known about its long-term efficacy. In addition, there are several obstacles that hinder the advancement of this approach. One is the high cost and the limited availability of ERT in some countries. The second relates to the degree of correction on a cellular level in various tissues achieved in a defined period of time prior to transplantation, and whether this can be correlated with long-term sustained benefits for the MPS IH patients. Thirdly and significantly, intravenously administered IDUA does not appear to have the capacity to penetrate several organ systems – most notably brain, bone, and the heart valves. In addition, it is unclear at present whether the additive benefit of ERT and HCT observed early after co-modality therapy will translate into improvement (over and above HCT itself) of mental, musculoskeletal and cardiac disabilities in MPS I patients now surviving long term.

Brain

The major obstacle to translocation of enzyme delivered intravenously across the blood-brain barrier could in theory be overcome by infusion of IDUA into the spinal canal or lateral ventricles of the brain. This approach has been modeled successfully in the canine model of MPS I (CitationKakkis et al 2004b, CitationDickson et al 2007). Based on the animal data, Dickson et al (Dr Dickson, pers comm, May 2008) are testing intermittent intrathecal ERT in patients with attenuated MPS I disease to treat progressive narrowing of cervical spinal canal. While promising, these therapies are still in the early stages and, because of the dynamics of spinal cord compression in MPS I (CitationKennedy et al 1973; CitationKaufman et al 1982), data documenting outcomes with intrathecal delivery will not be available for several years.

Even more importantly, data from canine MPS I model suggest that intrathecal ERT has the potential to treat central nervous system and mental decline in humans (CitationDickson et al 2007). Even though some treated animals developed meningeal inflammation, it is encouraging that with improved pharmaceutical formulation the side effects of intrathecal administration in patients were minimal (Dr. Dickson, pers comm, May 2008). The experience with intravenous ERT has shown that, for a beneficial effect, frequent administration is required; this would suggest that the effects of intermittently dosed intrathecal ERT will be also be transient, although the frequency of administration may be less than what is required via the intravenous route (CitationDickson et al 2007). This would of course pose difficulties for the life-long treatment of IDUA deficiency in the central nervous system.

It is plausible that intrathecal ERT will “bridge” the time between diagnosis and IDUA production by donor cells in the brain of the recipient following donor hematopoietic engraftment. The benefits of HCT in the brain appear to stem from the ability of the donor hematopoietic cells to migrate from systemic vessels to the brain parenchyma after HCT (CitationShapiro et al 1995; CitationKennedy and Abkowitz 1997; CitationPeters et al 1998c; CitationKrivit et al 1999). The cells implicated in this process are microglia, hematopoietic cells of monocytic lineage (CitationKrivit et al 1995). The dynamics of this engraftment are not well understood, as it is difficult to determine in a clinical setting. However, several lines of evidence indicate that after HCT, donor microglia home to and engraft in the brain in sufficient numbers in both mice and humans to arrest the anticipated ongoing neurologic deterioration.

An alternative, “Trojan horse” approach to crossing the blood-brain barrier has been tested by CitationPardridge et al (2005a, Citationb, Citationc) who used fusion proteins to transport IDUA across blood-brain barrier via the endocytosis-exocytosis pathway. Similarly, we have chosen a transferrin-IDUA fusion gene product to provide a proof of the concept that transferrin receptor-mediated transcytosis across blood brain barrier endothelium can be used as a functionally efficient pathway for an uptake of IDUA in brain cells, including neurons (CitationOsborn et al 2008).

Lastly, based on the assumption that microglial engraftment after HCT or intermittent IDUA intrathecal dosing both have sufficient drawbacks to make them alternative strategies worth exploring, continuous IDUA infusion into the spinal fluid by a semi-permanent pump within the central nervous system or externally by catheter delivery is theoretically feasible.

Bone

One of the most prevalent and difficult to manage long-term difficulties in otherwise successfully treated Hurler patients include musculoskeletal manifestations from progressive dysostosis multiplex (CitationOestreich 1985; CitationSchmidt et al 1987; CitationMasterson et al 1996; CitationOdunusi et al 1999; CitationWeisstein et al 2004). The patients exhibit numerous orthopedic complications despite HCT, including kyphosis, scoliosis, cervical spine instability, genu valgum, and carpal tunnel syndrome. Despite full donor engraftment after “successful” HCT (CitationField et al 1994; CitationWeisstein et al 2004; CitationKhanna et al 2007; CitationTaylor et al 2008; CitationPolgreen et al 2008), many patients require multiple surgeries, and, as a result of the combined effect of the primary MPS I disease and the impact of the conditioning therapy on endocrine and bone functions, are affected with musculoskeletal limitations for the rest of their lives.

Heart

Cardiac pathology is prominent in patients with Hurler syndrome. While coronary artery disease and cardiomyopathy appear to be treated by successful HCT (and presumably ERT), heart valve abnormalities do not appear to be corrected by ERT or HCT, although it is possible that these interventions may affect progression of valvular disease (CitationRenteria et al 1976; CitationVinallonga et al 1992; CitationDangel 1998; CitationBraunlin et al 2003, Citation2006; CitationHirth et al 2007). Murine model of MPS I (CitationClarke et al 1997; CitationRussell et al 1998; CitationOhmi et al 2003) represents a platform for testing various interventions including targeted cell therapy or attenuation of thickening of heart valves by secondary means such as decreasing the GAG-mediated inflammation (CitationTaylor and Gallo 2006).

Gene therapy

Yet another source of investigations likely to be relevant in the future are data generated by gene therapy experimentation in animal models of MPS I (CitationLutzko et al 1999; CitationZheng et al 2003; CitationEllinwood et al 2004; CitationHartung et al 2004; CitationDi Domenico et al 2005; CitationKobayashi et al 2005; CitationLiu et al 2005; CitationPonder and Haskins 2007; CitationTraas et al 2007; CitationChung et al 2007; CitationWatson et al 2006; CitationMa et al 2007). Despite significant complications and side effects observed in the viral-mediated gene therapy clinical trials for X-linked severe combined immune deficiency and chronic granulomatous disease (CitationBaum et al 2003; CitationHacein-Bey-Abina et al 2003a, Citationb; CitationMcCormack et al 2003; CitationFischer et al 2004; Citationvon Kalle et al 2004), optimism remains that improved viral and non-viral vectors will prove to be effective for delivery of enzymes such as IDUA in a sufficiently high local concentration to alter the sites (eg, brain, bones, heart) that are not fully corrected by the currently available therapies. Aside from immune reactions to the viral proteins, most lethal complications of gene therapy trials were caused by disruption of gene integrity and regulation in the recipient. For this reason, tremendous amounts of research have focused on the mechanism and risk quantification of insertional mutagenesis. The probability of uncovering a relatively low-frequency/high-risk side effect event (such as the development of leukemias occurring in the X-linked severe combined immune deficiency trial) will be difficult in a surrogate organism due to the prolonged latency of these events. Alternatively, cancer-prone animals or cell-based assays are likely to be used to define risk-benefit ratio assessments prior to future viral-based clinical trials. Improved vector design, enabling for example gene insertion in a specific genome location, or insulation of the gene of interest from the rest of the genome, or gene correction by homologous recombination to avoid insertional mutagenesis, would represent important advances in gene therapy (CitationPorteus et al 2006).

Stem cell gene therapy

The possibility of using gene therapy to correct hematopoietic cells of the recipient ex vivo followed by re-infusion with or without conditioning therapy, would avoid the substantial toxicity of allogenic transplantation, including GVHD, sustained immune suppression, and graft rejection. In addition, non-hematopoietic stem cells isolated from bone marrow can be corrected ex vivo and, because of their multi-lineage potential, could be harnessed for organ–specific delivery of IDUA-producing cells. It is of interest that donor mesenchymal stem cells (MSC) can home to sites of tissue injury (CitationKunter et al 2006; CitationProckop 2007). MSC have been shown to be safe in the first clinical trials for treatment for GVHD (CitationLe Blanc et al 2004; CitationAggarwal and Pittenger 2005; CitationLe Blanc and Ringden 2005a, Citationb; CitationProckop and Olson 2007).

The options for non-hematopoietic stem cells in therapy of MPS I are multiple (CitationMuller et al 2006). First, allogenic MSC have been used in metachromatic leukodystrophy and appeared to improve nerve conduction velocities in several patients (CitationKoc et al 2002). Therefore, they can perhaps be used as a “depot” of cells able to produce the IDUA continuously and potentially at sites other than those targeted by hematopoietic cells. Second, allogenic or gene-corrected autologous MSC can be considered as treatment of neurologic, bone, and heart valve disease not readily accessible by freely diffusible IDUA after HCT. In theory, they could be infused intravenously (to correct visceral GAG storage) or intrathecally (to correct brain pathology). Lastly, when used at the time of hematopoietic cell infusion, MSC have a potential to improve engraftment as well as a potential to prevent acute GVHD, as has been shown already in patients with malignant disorders receiving HCT.

Summary

Multiple agents and interventions are the mainstay of therapy for malignancies and infectious disease, and clearly seem to be the future trend in treatment of enzymopathies such as MPS I as well. As detailed above, the multiple tools available to us today for IDUA delivery include delivery of protein systemically and intermittently by ERT. Alternatively, enzyme may be delivered by a cellular approach using either allogenic cells producing enzyme such as HCT, or by gene therapy vectors to correct IDUA deficient cells, or to overexpress the gene product. These strategies will be available to researchers and clinicians interested in defining more efficacious and less toxic therapy for MPS I patients in the future.

We believe that the best available approach for newly diagnosed MPS IH patients at the current time is combination therapy of ERT and HCT. The use of ERT can prepare the patient for the transplant process by decreasing the GAG burden in the viscera, thereby providing an opportunity to limit the significant toxic effects of HCT. This is of key importance since MPS IH patients with pulmonary disease are at higher risk for HCT complication than MPS IH children without pulmonary symptoms before HCT (unpublished data).

This pre-emptive attempt to decrease morbidity associated with HCT by using ERT needs to be defined better by prospective long-term and short-term quantitative metrics, such as urinary GAG, determination of organomegaly, evidence of airway obstruction, and neuropsychological evaluations. Organized, multi-institutional efforts spanning experiences and practices throughout the world will provide a unified collection of data and evidence-based approaches, allowing conclusions to be drawn more expediently as greater numbers of patients can be evaluated (CitationPastores et al 2007). For rare disorders such as MPS I, collaborative studies will be important in moving the field forward and achieving optimal outcomes for these patients and their families.

Acknowledgements

Supported by: Children’s Cancer Research Fund in Minneapolis, Minnesota.

Disclosures

The authors report no conflicts of interest.

References

  • AggarwalSPittengerMF2005Human mesenchymal stem cells modulate allogeneic immune cell responsesBlood10518152215494428
  • BachGFriedmanRWeissmannB1972The defect in the Hurler and Scheie syndromes: deficiency of -L-iduronidaseProc Natl Acad Sci USA692048514262258
  • BaumCDullmannJLiZ2003Side effects of retroviral gene transfer into hematopoietic stem cellsBlood101209911412511419
  • BoelensJJWynnRFO’MearaA2007Outcomes of hematopoietic stem cell transplantation for Hurler’s syndrome in Europe: a risk factor analysis for graft failureBone Marrow Transplant402253317529997
  • BraunlinEMackey-BojackSPanoskaltsis-MortariA2006Cardiac functional and histopathologic findings in humans and mice with mucopolysaccharidosis type I: implications for assessment of therapeutic interventions in hurler syndromePediatr Res59273216326988
  • BraunlinEAStaufferNRPetersCH2003Usefulness of bone marrow transplantation in the Hurler syndromeAm J Cardiol92882614516901
  • ChungSMaXLiuY2007Effect of neonatal administration of a retroviral vector expressing alpha-L-iduronidase upon lysosomal storage in brain and other organs in mucopolysaccharidosis I miceMol Genet Metab901819216979922
  • ClarkeLARussellCSPownallS1997Murine mucopolysaccharidosis type I: targeted disruption of the murine alpha-L-iduronidase geneHum Mol Genet6503119097952
  • ComanDJHayesIMCollinsV2008Enzyme replacement therapy for mucopolysaccharidoses: opinions of patients and familiesJ Pediatr152723718410781
  • Cox-BrinkmanJBoelensJJWraithJE2006Haematopoietic cell transplantation (HCT) in combination with enzyme replacement therapy (ERT) in patients with Hurler syndromeBone Marrow Transplant38172116715104
  • DangelJH1998Cardiovascular changes in children with mucopolysaccharide storage diseases and related disorders–clinical and echocardiographic findings in 64 patientsEur J Pediatr15753489686810
  • Di DomenicoCVillaniGRDi NapoliD2005Gene therapy for a mucopolysaccharidosis type I murine model with lentiviral-IDUA vectorHum Gene Ther16819015703491
  • DicksonPMcEnteeMVoglerC2007Intrathecal enzyme replacement therapy: successful treatment of brain disease via the cerebrospinal fluidMol Genet Metab9161817321776
  • EllinwoodNMViteCHHaskinsME2004Gene therapy for lysosomal storage diseases: the lessons and promise of animal modelsJ Gene Med648150615133760
  • FieldREBuchananJACopplemansMG1994Bone-marrow transplantation in Hurler’s syndrome. Effect on skeletal developmentJ Bone Joint Surg Br76975817983131
  • FischerAAbinaSHThrasherA2004LMO2 and gene therapy for severe combined immunodeficiencyN Engl J Med35025267author reply 2526–715190153
  • FischerHDGonzalez-NoriegaASlyWS1980Phosphomannosylenzyme receptors in rat liver. Subcellular distribution and role in intracellular transport of lysosomal enzymesJ Biol Chem2559608156253448
  • FratantoniJCHallCWNeufeldEF1968aThe defect in Hurler’s and Hunter’s syndromes: faulty degradation of mucopolysaccharideProc Natl Acad Sci USA606997064236091
  • FratantoniJCHallCWNeufeldEF1968bHurler and Hunter syndromes: mutual correction of the defect in cultured fibroblastsScience16257024236721
  • GassasASungLDoyleJJ2003Life-threatening pulmonary hemorrhages post bone marrow transplantation in Hurler syndrome. Report of three cases and review of the literatureBone Marrow Transplant32213512838287
  • GlarosENTurnerCTParkinsonEJ2002Immune response to enzyme replacement therapy: single epitope control of antigen distribution from circulationMol Genet Metab771273512359140
  • GrewalSSKrivitWDeforTE2002Outcome of second hematopoietic cell transplantation in Hurler syndromeBone Marrow Transplant29491611960268
  • GrewalSSWynnRAbdenurJE2005Safety and efficacy of enzyme replacement therapy in combination with hematopoietic stem cell transplantation in Hurler syndromeGenet Med7143615714083
  • Hacein-Bey-AbinaSVon KalleCSchmidtM2003aA serious adverse event after successful gene therapy for X-linked severe combined immunodeficiencyN Engl J Med348255612529469
  • Hacein-Bey-AbinaSVon KalleCSchmidtM2003bLMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1Science302415914564000
  • HansenMDFilipovichAHDaviesSM2008Allogeneic hematopoietic cell transplantation (HCT) in Hurler’s syndrome using a reduced intensity preparative regimenBone Marrow Transplant413495318026148
  • HartungSDFrandsenJLPanD2004Correction of metabolic, craniofacial, and neurologic abnormalities in MPS I mice treated at birth with adeno-associated virus vector transducing the human alpha-L-iduronidase geneMol Ther98667515194053
  • HasilikANeufeldEF1980Biosynthesis of lysosomal enzymes in fibroblasts. Phosphorylation of mannose residuesJ Biol Chem2554946506989822
  • HirthABergAGreveG2007Successful treatment of severe heart failure in an infant with Hurler syndromeJ Inherit Metab Dis3082017768668
  • HobbsJRHugh-JonesKBarrettAJ1981Reversal of clinical features of Hurler’s disease and biochemical improvement after treatment by bone-marrow transplantationLancet2709126116856
  • KakavanosRTurnerCTHopwoodJJ2003Immune tolerance after long-term enzyme-replacement therapy among patients who have mucopolysaccharidosis ILancet36116081312747881
  • KakkisELesterTYangR2004aSuccessful induction of immune tolerance to enzyme replacement therapy in canine mucopolysaccharidosis IProc Natl Acad Sci USA1018293414715900
  • KakkisEMcEnteeMVoglerC2004bIntrathecal enzyme replacement therapy reduces lysosomal storage in the brain and meninges of the canine model of MPS IMol Genet Metab831637415464431
  • KakkisEDMatyniaAJonasAJ1994Overexpression of the human lysosomal enzyme alpha-L-iduronidase in Chinese hamster ovary cellsProtein Expr Purif5225327950365
  • KakkisEDMcEnteeMFSchmidtchenA1996Long-term and high-dose trials of enzyme replacement therapy in the canine model of mucopolysaccharidosis IBiochem Mol Med58156678812735
  • KakkisEDMuenzerJTillerGE2001aEnzyme-replacement therapy in mucopolysaccharidosis IN Engl J Med344182811172140
  • KakkisEDSchuchmanEHeX2001bEnzyme replacement therapy in feline mucopolysaccharidosis IMol Genet Metab7219920811243725
  • KaufmanHHRosenbergHSScottCI1982Cervical myelopathy due to dural compression in mucopolysaccharidosisSurg Neurol17404106810485
  • KennedyDWAbkowitzJL1997Kinetics of central nervous system microglial and macrophage engraftment: analysis using a transgenic bone marrow transplantation modelBlood90986939242527
  • KennedyPSwashMDeanMF1973Cervical cord compression in mucopolysaccharidosisDev Med Child Neurol1519494266829
  • KhannaGVan HeestAEAgelJ2007Analysis of factors affecting development of carpal tunnel syndrome in patients with Hurler syndrome after hematopoietic cell transplantationBone Marrow Transplant39331417277793
  • KobayashiHCarbonaroDPepperK2005Neonatal gene therapy of MPS I mice by intravenous injection of a lentiviral vectorMol Ther117768915851016
  • KocONDayJNiederM2002Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH)Bone Marrow Transplant302152212203137
  • KrivitWPetersCShapiroEG1999Bone marrow transplantation as effective treatment of central nervous system disease in globoid cell leukodystrophy, metachromatic leukodystrophy, adrenoleukodystrophy, mannosidosis, fucosidosis, aspartylglucosaminuria, Hurler, Maroteaux-Lamy, and Sly syndromes, and Gaucher disease type IIICurr Opin Neurol121677610226749
  • KrivitWSungJHShapiroEG1995Microglia: the effector cell for reconstitution of the central nervous system following bone marrow transplantation for lysosomal and peroxisomal storage diseasesCell Transplant4385927582569
  • KunterURongSDjuricZ2006Transplanted mesenchymal stem cells accelerate glomerular healing in experimental glomerulonephritisJ Am Soc Nephrol1720212
  • Le BlancKRasmussonISundbergB2004Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cellsLancet3634394114962524
  • Le BlancKRingdenO2005aImmunobiology of human mesenchymal stem cells and future use in hematopoietic stem cell transplantationBiol Blood Marrow Transplant112134
  • Le BlancKRingdenO2005bUse of mesenchymal stem cells for the prevention of immune complications of hematopoietic stem cell transplantationHaematologica9038a
  • Lee-ChenGJLinSPTangYF1999Mucopolysaccharidosis type I: characterization of novel mutations affecting alpha-L-iduronidase activityClin Genet56667010466419
  • Lee-ChenGJWangTR1997Mucopolysaccharidosis type I: identification of novel mutations that cause Hurler/Scheie syndrome in Chinese familiesJ Med Genet3439419032648
  • LiuYXuLHennigAK2005Liver-directed neonatal gene therapy prevents cardiac, bone, ear, and eye disease in mucopolysaccharidosis I miceMol Ther11354715585404
  • LutzkoCOmoriFAbrams-OggAC1999Gene therapy for canine alpha-L-iduronidase deficiency: in utero adoptive transfer of genetically corrected hematopoietic progenitors results in engraftment but not amelioration of diseaseHum Gene Ther1052132
  • MaXLiuYTittigerM2007Improvements in mucopolysaccharidosis I mice after adult retroviral vector-mediated gene therapy with immunomodulationMol Ther1588990217311010
  • MastersonELMurphyPGO’MearaA1996Hip dysplasia in Hurler’s syndrome: orthopaedic management after bone marrow transplantationJ Pediatr Orthop16313
  • McCormackMPForsterADrynanL2003The LMO2 T-cell oncogene is activated via chromosomal translocations or retroviral insertion during gene therapy but has no mandatory role in normal T-cell developmentMol Cell Biol2390031314645513
  • MeiklePJHopwoodJJClagueAE1999Prevalence of lysosomal storage disordersJAMA2814954
  • MullerIKustermann-KuhnBHolzwarthC2006In vitro analysis of multipotent mesenchymal stromal cells as potential cellular therapeutics in neurometabolic diseases in pediatric patientsExp Hematol344139
  • NeufeldEF1991Lysosomal storage diseasesAnnu Rev Biochem605780
  • OdunusiEPetersCKrivitW1999Genu valgum deformity in Hurler syndrome after hematopoietic stem cell transplantation: correction by surgical interventionJ Pediatr Orthop197049890291
  • OestreichAE1985The stylohyoid ligament in Hurler syndrome and related conditions: comparison with normal childrenRadiology15466563918332
  • OhmiKGreenbergDSRajavelKS2003Activated microglia in cortex of mouse models of mucopolysaccharidoses I and IIIBProc Natl Acad Sci USA1001902712576554
  • OrchardPJBlazarBRWagnerJ2007Hematopoietic cell therapy for metabolic diseaseJ Pediatr151340617889065
  • OsbornMJMcElmurryRTPeacockB2008Targeting of the CNS in MPS-IH Using a Nonviral Transferrin-alpha-L-iduronidase Fusion Gene ProductMol TherIn press
  • PardridgeWM2005aThe blood-brain barrier and neurotherapeuticsNeuroRx21215717052
  • PardridgeWM2005bThe blood-brain barrier: bottleneck in brain drug developmentNeuroRx231415717053
  • PardridgeWM2005cMolecular biology of the blood-brain barrierMol Biotechnol30577015805577
  • PastoresGMArnPBeckM2007The MPS I registry: design, methodology, and early findings of a global disease registry for monitoring patients with Mucopolysaccharidosis Type IMol Genet Metab91374717336562
  • PetersCBalthazorMShapiroEG1996Outcome of unrelated donor bone marrow transplantation in 40 children with Hurler syndromeBlood8748949028639864
  • PetersCShapiroEGAndersonJ1998aHurler syndrome: II. Outcome of HLA-genotypically identical sibling and HLA-haploidentical related donor bone marrow transplantation in fifty-four children. The Storage Disease Collaborative Study GroupBlood91260189516162
  • PetersCShapiroEGKrivitW1998bHurler syndrome: past, present, and futureJ Pediatr133799672503
  • PetersCShapiroEGKrivitW1998cNeuropsychological development in children with Hurler syndrome following hematopoietic stem cell transplantationPediatr Transplant2250310084724
  • PolgreenLETolarJPlogM2008Growth and endocrine function in patients with Hurler syndrome after hematopoietic stem cell transplantationBone Marrow Transplant4110051118278070
  • PonderKPHaskinsME2007Gene therapy for mucopolysaccharidosisExpert Opin Biol Ther713334517727324
  • PorteusMHConnellyJPPruettSM2006A look to future directions in gene therapy research for monogenic diseasesPLoS Genet2e13317009872
  • ProckopDJ2007“Stemness” does not explain the repair of many tissues by mesenchymal stem/multipotent stromal cells (MSCs)”Clin Pharmacol Ther82241317700588
  • ProckopDJOlsonSD2007Clinical trials with adult stem/progenitor cells for tissue repair: let’s not overlook some essential precautionsBlood10931475117170129
  • RempelBPClarkeLAWithersSG2005A homology model for human alpha-l-iduronidase: insights into human diseaseMol Genet Metab85283715862278
  • RenteriaVGFerransVJRobertsWC1976The heart in the Hurler syndrome: gross, histologic and ultrastructural observations in five necropsy casesAm J Cardiol38487501823811
  • RoubicekMGehlerJSprangerJ1985The clinical spectrum of alpha-L-iduronidase deficiencyAm J Med Genet20471813922223
  • RussellCHendsonGJevonG1998Murine MPS I: insights into the pathogenesis of Hurler syndromeClin Genet53349619660052
  • SchmidtHUllrichKVon LengerkeHJ1987Radiological findings in patients with mucopolysaccharidosis I H/S (Hurler-Scheie syndrome)Pediatr Radiol17409143114705
  • SchuchmanEHGuzmanNADesnickRJ1984Human alpha-L-iduronidase. I. Purification and properties of the high uptake (higher molecular weight) and the low uptake (processed) formsJ Biol Chem2593132406421818
  • ScottHSBungeSGalA1995Molecular genetics of mucopolysaccharidosis type I: diagnostic, clinical, and biological implicationsHum Mutat62883028680403
  • ScottHSLitjensTNelsonPV1992alpha-L-iduronidase mutations (Q70X and P533R) associate with a severe Hurler phenotypeHum Mutat133391301941
  • ScottHSLitjensTNelsonPV1993Identification of mutations in the alpha-L-iduronidase gene (IDUA) that cause Hurler and Scheie syndromesAm J Hum Genet53973868213840
  • ShapiroEGLockmanLABalthazorM1995Neuropsychological outcomes of several storage diseases with and without bone marrow transplantationJ Inherit Metab Dis18413297494400
  • ShullRLuXDubeI1996Humoral immune response limits gene therapy in anine MPS IBlood8837798704199
  • ShullRMKakkisEDMcEnteeMF1994Enzyme replacement in a canine model of Hurler syndromeProc Natl Acad Sci USA9112937417809150
  • SifuentesMDoroshowRHoftR2007A follow-up study of MPS I patients treated with laronidase enzyme replacement therapy for 6 yearsMol Genet Metab901718017011223
  • SouilletGGuffonNMaireI2003Outcome of 27 patients with Hurler’s syndrome transplanted from either related or unrelated haematopoietic stem cell sourcesBone Marrow Transplant3111051712796790
  • StabaSLEscolarMLPoeM2004Cord-blood transplants from unrelated donors in patients with Hurler’s syndromeN Engl J Med3501960915128896
  • SugawaraKSaitoSOhnoK2008Structural study on mutant alpha-L-iduronidases: insight into mucopolysaccharidosis type IJ Hum Genet534677418340403
  • TaylorCBradyPO’MearaA2008Mobility in Hurler syndromeJ Pediatr Orthop28163818388709
  • TaylorKRGalloRL2006Glycosaminoglycans and their proteoglycans: host-associated molecular patterns for initiation and modulation of inflammationFaseb J2092216394262
  • TolarJGrewalSSBjorakerKJ2007Combination of enzyme replacement and hematopoietic stem cell transplantation as therapy for Hurler syndromeBone Marrow TransplantIn press
  • TolarJOrchardPJKeyNS2008Circulating anticoagulant glycosaminoglycans in mucopolysaccharidosis type IJ Thromb Haemost6893518315551
  • TraasAMWangPMaX2007Correction of clinical manifestations of canine mucopolysaccharidosis I with neonatal retroviral vector gene therapyMol Ther1514233117519893
  • TurnerCTHopwoodJJBrooksDA2000Enzyme replacement therapy in mucopolysaccharidosis I: altered distribution and targeting of alpha-L-iduronidase in immunized ratsMol Genet Metab692778510870845
  • UngerEGDurrantJAnsonDS1994Recombinant alpha-L-iduronidase: characterization of the purified enzyme and correction of mucopolysaccharidosis type I fibroblastsBiochem J3044397998955
  • VellodiAYoungEPCooperA1997Bone marrow transplantation for mucopolysaccharidosis type I: experience of two British centresArch Dis Child769299068295
  • VinallongaXSanzNBalaguerA1992Hypertrophic cardiomyopathy in mucopolysaccharidoses: regression after bone marrow transplantationPediatr Cardiol1310791614914
  • Von KalleCFehseBLayh-SchmittG2004Stem cell clonality and genotoxicity in hematopoietic cells: gene activation side effects should be avoidableSemin Hematol413031815508116
  • WatsonGBastackyJBelichenkoP2006Intrathecal administration of AAV vectors for the treatment of lysosomal storage in the brains of MPS I miceGene Ther139172516482204
  • WeissteinJSDelgadoESteinbachLS2004Musculoskeletal manifestations of Hurler syndrome: long-term follow-up after bone marrow transplantationJ Pediatr Orthop249710114676543
  • WraithJE2001Enzyme replacement therapy in mucopolysaccharidosis type I: progress and emerging difficultiesJ Inherit Metab Dis242455011405343
  • WraithJEBeckMLaneR2007Enzyme replacement therapy in patients who have mucopolysaccharidosis I and are younger than 5 years: results of a multinational study of recombinant human alpha-L-iduronidase (laronidase)Pediatrics120e374617606547
  • WraithJEClarkeLABeckM2004Enzyme replacement therapy for mucopolysaccharidosis I: a randomized, double-blinded, placebo-controlled, multinational study of recombinant human alpha-L-iduronidase (laronidase)J Pediatr144581815126990
  • ZhengYRozengurtNRyazantsevS2003Treatment of the mouse model of mucopolysaccharidosis I with retrovirally transduced bone marrowMol Genet Metab792334412948739

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.