Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Jul 1.
Published in final edited form as: Exp Neurol. 2009 Apr 8;218(1):5–8. doi: 10.1016/j.expneurol.2009.03.040

Promising CNS-Directed Enzyme Replacement Therapy for Lysosomal Storage Diseases

Shannon L Macauley 1, Mark S Sands 1,*
PMCID: PMC2701189  NIHMSID: NIHMS108720  PMID: 19361502

Lysosomal storage diseases (LSD) are a group of inherited metabolic disorders usually resulting from a deficiency in a single acid hydrolase (Neufeld, 1991). The remaining subsets of LSDs are caused by defects in key lysosomal membrane proteins, proteins involved in lysosomal enzyme trafficking, or lysosomal enzyme activator proteins (Neufeld, 1991). Regardless of the gene defect, most LSDs result in an accumulation of undegraded or partially degraded substrates that leads to the hallmark of LSDs, microscopically demonstrable lysosomal distension. These diseases are usually autosomal recessive in nature but several have an X-linked inheritance pattern. Individually, LSDs are rare but as a group they encompass as many as 50 distinct disorders with a frequency of approximately 1 in 5,000 live births (Meikle et al., 1999). This makes the LSDs one of the most common childhood genetic diseases. In many instances, children are asymptomatic at birth. However, LSDs are progressive in nature and invariably fatal. Since lysosomal enzymes are typically ubiquitously expressed, LSDs can present with a broad spectrum of clinical signs including some or all of the following: organomegally, visual and hearing deficits, cardiac insufficiencies, skeletal defects, immunologic abnormalities, and central nervous system (CNS) involvement (Neufeld, 1991, Marodi et al., 1995, Daly et al., 1999, Gadola et al., 2006). Approximately 75% of LSDs have CNS involvement that manifest as mental retardation, seizures, profound neurodegeneration, behavioral abnormalities, and psycho-motor defects. Due to the debilitating nature of the associated clinical signs and the protective nature of blood brain barrier, the CNS disease remains one of the major challenges when developing therapies for this class of inherited disorder.

Enzyme replacement therapy (ERT) for LSDs is one of the true success stories in modern molecular medicine. The seminal discovery that lysosomal enzymes can be secreted from one cell and taken up by an adjacent cell (originally referred to as “cross-correction) opened up the possibility that this process could be exploited for therapeutic purposes (Neufeld and Fratantoni, 1970). However, the successful application of this process for the treatment of LSDs had to wait until the molecular mechanisms involved in “cross-correction” were dissected. The discoveries of the mannose (Achord et al., 1978) and mannose 6-phosphate (Kornfeld, 1992) recognition systems provided a mechanistic framework by which lysosomal enzymes could be modified for targeted or widespread delivery, respectively. The mannose receptor is localized to the plasma membrane and its expression is limited primarily to cells of the reticuloendothelial (RE) system. In contrast, the mannose 6-phosphate receptor is a ubiquitously expressed protein localized to both the endoplasmic reticulum and the plasma membrane.

Once the uptake mechanisms and their respective receptors were determined, properly modified enzyme could be produced and tested clinically. The first successful clinical trial of ERT for a LSD was performed in patients with type I Gaucher disease (Barton et al., 1991, Beutler et al., 1991). Type I Gaucher disease is a LSD caused by the deficiency of acid β-glucocerebrosidase activity and primarily affects cells of the RE system (Beutler and Grabowski, 2001). β-Glucocerebrosidase was modified to expose terminal mannose residues in order to target the enzyme to cells of the RE system. This therapy improves several hematologic parameters, decreases splenomegally, and improves skeletal abnormalities, albeit at a slower rate than the hematologic and splenic improvements. Enzyme replacement therapy for type I Gaucher disease is very effective and is now considered the standard-of-care.

Once ERT for type I Gaucher disease was shown to be safe and efficacious, the field moved quickly to develop ERT for some of the more common LSDs. The major difference between ERT for type I Gaucher disease and other LSDs is that most of the LSDs affect multiple cell types in addition to cells of the RE system. This required enzyme that can be taken up by diverse cell types. It was shown that preparations produced in mammalian cells contained a high proportion of enzyme that contained the mannose 6-phosphate recognition marker. As mentioned above, the mannose 6-phosphate receptor is expressed on most cell types of the body (Kaplan et al., 1977, Sando and Neufeld, 1977, Robbins, 1979, Ullrich et al., 1979, Rome and Miller, 1980). Clinical trials were performed nearly simultaneously for mucopolysaccharidosis type I (MPS I, α-L-iduronidase deficiency) and Fabry disease (α-galactosidase A deficiency) (Kakkis et al., 2001, Eng et al., 2001). Both diseases respond to weekly or bi-weekly intravenous infusions of recombinant enzyme. Enzyme replacement therapy is now available clinically for mucopolysaccharidosis types II (Hunter Syndrome, iduronate sulfatase deficiency) and VI (Maroteaux-Lamy Syndrome, arylsulfatase B deficiency) and Pompe Disease (acid maltase deficiency). Clinical trials are currently underway for Niemann-Pick B (NP-B) disease and mucopolysaccharidosis type IV (MPS IV). Pre-clinical experiments in small animals have also shown promise for ERT in Aspartylglucosaminuria (Kelo et al., 2005), Metachromatic Leukodystrophy (Matzner et al., 2009), Krabbe disease (Lee et al., 2005), Wolman disease (Du et al., 2008), and Sly Syndrome (Sands et al., 1994). One common aspect of all the clinical ERT protocols is that they are approved for mild, non-neuropathic forms of disease. Although ERT can be very effective at treating the systemic disease, lysosomal enzymes do not readily cross the blood brain barrier. Therefore, alternate means for delivering enzyme to the brain will have to be developed.

The article by Dodge et al., in the February issue of Experiment Neurology addresses the problem of CNS delivery for lysosomal enzymes (Dodge et al., 2009). In this paper, the investigators sought to treat the CNS manifestations of Niemann-Pick A (NP-A). NP-A is a rare, autosommal recessive LSD that results from a deficiency in the soluble lysosomal enzyme, acid sphingomyelinase (ASM; Schuchman and Desnick, 2001). Acid sphingomyelinase is responsible for the hydrolosis of spingomyelin (SPM; Elleder, 1989; Schuchman and Desnick, 2001). The absence of ASM results in the accumulation of SPM throughout the CNS and viscera. In addition to lysosomal distension resulting in hepatosplenomegaly, NP-A patients suffer from severe cognitive deficits, motor dysfunction, and blindness due to sphingomyelin accumulation in both neurons and glia. Patients are normally diagnosed with NP-A in the first year of life with death occurring by age 3. A less severe form of Niemann-Pick Disease, type B (NP-B), occurs when greater than 10% of residual ASM activity remains causing the viscera to be affected but sparing the CNS.

The creation of mouse models of NP-A (Horinouchi et al., 1995; Otterbach and Stoffel, 1995) has aided in the development of therapies for NP-A and NP-B. The ASM-deficient mouse has been extensively characterized (Horinouchi et al., 1995; Macauley et al., 2008) and recapitulates the clinical course of the disease, especially within the CNS (Sarna et al., 2001; Macauley et al., 2008). Most therapies aimed at treating the systemic aspects of NP-B or NP-A have largely been successful. Notably, considerable success has been achieved with ERT (Miranda et al., 2000) and, as mentioned above, this approach is currently in Phase I clinical trials.

As with other LSDs, treating the CNS manifestations of NP-A continues to be problematic. Previous work in the ASM-deficient mouse utilizing cell-based therapies, such as the intracerebral transplantation of mesenchymal stem cells (Jin et al., 2002) or adult-derived neural progenitors (Shihabuddin et al., 2004), led to improvements in the CNS disease. In both cases, the authors demonstrated that the cells were viable and migrated away from the injection site, providing the CNS with ASM activity and a delay in the onset of disease. However, in both cases, these therapies were partially efficacious, resulted in focal areas of correction, and were not a long-term cure of NP-A. Perhaps the most promising approach has been the use of CNS-directed gene therapy (Passini et al., 2005, 2007; Dodge et al., 2005). These studies demonstrate that various serotypes of AAV vectors expressing ASM are appropriate therapeutic delivery systems for enzymes to the CNS. The vectors showed long-term expression of ASM, axonal transport of both virus and enzyme, reversal of SPM accumulation, decreased neuronal loss, and improvement in motor deficits. Although gene therapy treats the focal areas of CNS pathology (i.e. cerebellum), to date, no therapy has successfully treated the entire CNS. Furthermore, the use of both cell-based therapies and viral vectors are often the subjects of debate due to ethical and safety concerns surrounding their use (see below).

Dodge et al., (2009) utilized the principles of ERT and adapted them for delivery to the CNS. In an attempt to globally treat the CNS in the ASM-deficient mouse, the authors implanted a cannula into the lateral ventricle and slowly infused human ASM (hASM) over the course of 6 hours. This single infusion resulted in detectable levels of hASM in the brains for up to 3 weeks. The spread of hASM throughout the neuroaxis was impressive with areas proximal to the injection site displaying the highest levels of hASM (i.e. forebrain) and a decreasing gradient of enzyme to sites more distal to the infusion site (i.e. cerebellum). Concurrent with the presence of hASM, the authors demonstrate a decrease in the amount of sequestered SPM and cholesterol; most notably in the forebrain. Interestingly, hASM was also detected in the serum. Consistent with this observation, levels of SPM in systemic organs like liver and lung were significantly decreased compared to untreated ASM-deficient animals. After determining that a single infusion successfully lowered SPM levels for 3 weeks post-injection, the authors performed hASM infusions every 2 weeks for 8 weeks. Repeated infusions successfully decreased the SPM load in the brains of ASM-deficient mice for an extended period of time. Furthermore, the treated ASM-deficient mice showed moderate improvements on foot fault tests and gait analysis.

The work by Dodge et al., (2009) provides an alternate approach to the treatment of CNS pathology associated with LSDs. CNS-directed ERT has several advantages. First, unlike other therapeutic strategies, this approach does not require the injection of live cells or viral vectors. Thus, ethical concerns surrounding the use of stem cells or viral vectors are circumvented. There are no concerns about placing a viable cell in the brain without a “kill switch” if an unexpected adverse event occurs. Similarly, in certain cases the use of gene transfer vectors, such as γ-retroviruses (Hacein-Bey-Abina et al., 2003) and adeno-associated viruses (Donsante et al., 2007), have been linked to malignant transformation of transduced cells. One advantage of ERT is that if there is an adverse event, such as a severe immune response, the infusion can simply be stopped. This gives a patient the option to modify or abandon therapy if the enzyme infusions become problematic.

Second, the authors demonstrate the feasibility of repeated infusions. Although CNS-directed ERT using a single injection has been described previously in the ASM-deficient mouse (Yang et al., 2007) and other models of LSD (Lee et al., 2007; Lonser et al., 2005; Chang et al., 2008), a single injection will not lead to persistent therapeutic levels of enzyme. Dodge et al., (2009) clearly show that the enzyme levels wane after a single injection and SPM begins to reaccumulate relatively rapidly, therefore creating a need for multiple infusions. The data suggest that long-term CNS-directed ERT could be accomplished by performing multiple infusions via cannulation or alternatively, using an implantable pump.

Third, in contrast to other ERT approaches directed to the CNS, Dodge et al., (2009) utilized a slow infusion rate coupled with a relatively long infusion time and large injection volume. This injection protocol is similar to convection-enhanced delivery that uses bulk flow to maximize the spread of therapeutic agents during direct CNS injections (Bobo et al., 1994; Bankiewicz et al., 2000). The authors adapted this approach for an intraventricular infusion, which resulted in enzyme delivery to a greater portion of the brain. The widespread delivery of enzyme to the brain has stymied the field and the approach described by Dodge et al., (2009) gives hope for delivering therapeutic agents to the entire brain and spinal cord.

Fourth, one surprising benefit of CNS-directed infusions was the presence of circulating levels of hASM in the blood as well as clearance of SPM from both the liver and lungs. This novel finding demonstrates that therapies aimed at treating the CNS might be beneficial in mitigating the visceral disease as well. Although the mechanism underlying this finding is unclear, it represents an important observation that warrants further study. Future studies thoroughly investigating the effects of CNS-directed infusions on lysosomal distension and functional deficits associated with the systemic disease will help determine whether this finding has clinical relevance.

Lastly and not surprisingly, Dodge et al., (2009) show that the highest levels of hASM and the greatest decrease in SPM occur near the injection site. This approach might be ideally suited for other LSDs, like Infantile Neuronal Ceroid Lipofuscinosis (INCL, Infantile Batten Disease), where the disease manifests first within the thalamus and cortex (Kielar et al., 2007). Although there is also significant pathology in the INCL cerebellum (Macauley et al., 2009), treatment of the forebrain by this approach could dramatically improve the outcome for these patients. Thus, although enzyme spread throughout the neuroaxis is essential, targeting therapies to the most severely affected brain structures is of paramount importance.

Although there are numerous benefits to the therapeutic approach described by Dodge et al., (2009) there are also questions that remain to be answered before this can be translated into the clinic. Although the authors demonstrate that elevated hASM levels in the CNS correlate with a decrease in SPM and cholesterol, it is not clear whether this correlates with a decrease in neuronal loss, especially in regions that are distant from the injection site. For example, ASM-deficient mice have a severe, time-dependent loss of Purkinje cells in the cerebellum and associated motor deficits as measured in the rotarod behavioral test. Does this approach spare Purkinje cells and improve the motor deficits that have been previously reported in this model (Sarna et al., 2001; Dodge et al., 2005; Macauley et al., 2008)?

Another concern for repeated enzyme infusions is the feasibility of this approach in a larger brain. Since a single cannulation into a mouse brain was not sufficient to completely normalize the SPM levels in the most distal sites (i.e. the cerebellum), the question remains as to whether this approach can be translated to the human brain. Infusions of other lysosomal enzymes both intraventricularly and intraparenchymally are reported in other LSD models (Lee et al., 2007; Yang et al., 2007; Chang et al., 2008) as well is in human clinical trials for Gaucher (Lonser et al.,2005), however, reports suggest that these therapies are only partially corrective. Thus, for this approach to be successful a second or possibly multiple cannulation sites will be needed when translated to human patients. This obviously increases the risk of complications and possibility of infection.

Alternatively, performing combination therapies might enhance the efficacy of this singular approach. In addition to CNS-directed ERT, a number of labs are exploring other treatments for the CNS disease, which has led to the development of cell-, viral vector-, bone marrow transplant- and small molecule-based treatments (for reviews, see Jeyakumar et al., 2005; Sands and Davidson, 2006). Although no single treatment has “cured” the CNS disease, many approaches yield promising results. Therefore, several labs are using a combination of treatments in an attempt to develop a more synergistic, complete therapeutic strategy. The additive benefit of combination therapy was clearly demonstrated in the MPSVII mouse model when ERT was coupled with bone marrow transplantation (BMT; Sands et al., 1997). Subsequent studies in Sandhoff mice combined substrate deprivation therapy with either BMT (Jeyakumar et al., 2001) or neural stem cells (Lee et al., 2007), which yielded a synergistic benefit in several parameters including lifespan. Additional combination therapies utilizing bone marrow transplantation with either a small molecule (Biswas et al., 2002) or CNS-directed gene therapy (Lin et al., 2007) led to the greatest increase in lifespan to date in a mouse model of Krabbe’s disease. Furthermore, work in NP-A demonstrated that the intravenous and intracerebral injections of either bone marrow-derived cells (Jin and Schuchman, 2003) or gene therapy (Passini et al., 2007) also produces an additive effect. Thus, the efficacy of CNS-directed ERT as described by Dodge et al might be increased with periodic intrathecal enzyme administration (Dickson et al., 2007), systemic or CNS-directed gene therapy (Sands and Davidson, 2006), or the use of small molecules (Schuchman, 2007) that are currently in development.

The work by Dodge et al., (2009) identifies a promising approach and represents an important milestone for the treatment of the CNS disease associated with NP-A disease. It will be important to determine if this approach is equally efficacious in other forms of LSD that have a significant CNS component. This is particularly important when considering diseases where preexisting storage material can be reversed, such as NP-AB (for review, see Schuchman, 2007) and MPS VII (Brooks et al., 2002), as compared to diseases that are more refractory to late intervention, such as Krabbe disease (Lin et al., 2007) and Neuronal Ceroid Lipofuscinosis (Griffey et al., 2004, 2006; Passini et al., 2006; Cabrera-Salazar et al., 2007). Although there are important questions that remain unanswered regarding the ability to translate this approach to humans, chronic or repeated intracerebroventricular infusion of a lysosomal enzyme could be an important component of an overall strategy that combines several modalities.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Achord DA, Brot FE, Bell CE, Sly WS. Human β-glucuronidase: In vivo clearance and in vitro uptake by a glycoprotein recognition system on reticuloendothelial cells. Cell. 1978;15:269–278. doi: 10.1016/0092-8674(78)90102-2. [DOI] [PubMed] [Google Scholar]
  2. Bankiewicz KS, Eberling JL, Kohutnicka M, Jagust W, Pivirotto P, Bringas J, Cunningham J, Budinger TF, Harvey-White J. Convection-enhanced delivery of AAV vector in parkinsonian monkeys; in vivo detection of gene expression and restoration of dopaminergic function using pro-drug approach. Exp Neurol. 2000;164:2–14. doi: 10.1006/exnr.2000.7408. [DOI] [PubMed] [Google Scholar]
  3. Barton NW, Brady RO, Dambrosia JM, Di Bisceglie AM, Doppelt SH, Hill SC, Mankin HJ, Murray GJ, Parker RI, Argoff CE, Grewel RP, Yu KT Collaborators. Replacement therapy for inherited enzyme deficiency – macrophage-targeted glucocerebrosidase for Gaucher’s disease. N Eng J Med. 1991;324:1464–1470. doi: 10.1056/NEJM199105233242104. [DOI] [PubMed] [Google Scholar]
  4. Beutler E, Kay A, Saven A, Garver P, Thurston D, Dawson A, Rosenbloom B. Enzyme replacement therapy for Gaucher disease. Blood. 1991;78:1183–1189. [PubMed] [Google Scholar]
  5. Beutler E, Grabowski GA. Gaucher disease. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The Metabolic and Molecular Bases of Inherited Disease. McGraw Hill; New York: 2001. pp. 3635–3668. [Google Scholar]
  6. Bobo RH, Laske DW, Akbasak A, Morrison PF, Dedrick RL, Oldfield EH. Convection-enhanced delivery of macromolecules in the brain. Proc Natl Acad Sci U S A. 1994;91:2076–2080. doi: 10.1073/pnas.91.6.2076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Biswas S, LeVine SM. Substrate-reduction therapy enhances the benefits of bone marrow transplantation in young mice with globoid cell leukodystrophy. Pediatr Res. 2002;51:40–47. doi: 10.1203/00006450-200201000-00009. [DOI] [PubMed] [Google Scholar]
  8. Brooks AI, Stein CS, Hughes SM, Heth J, McCray PM, Sauter SL, Johnston JC, Cory-Slechta DA, Federoff HJ, Davidson BL. Functional correction of established central nervous system deficits in an animal model of lysosomal storage disease with feline immunodeficiency virus-based vectors. Proc Natl Acad Sci U S A. 2002;99:6216–6221. doi: 10.1073/pnas.082011999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Cabrera-Salazar MA, Roskelley EM, Bu J, Hodges BL, Yew N, Dodge JC, Shihabuddin LS, Sohar I, Sleat DE, Scheule RK, Davidson BL, Cheng SH, Lobel P, Passini MA. Timing of therapeutic intervention determines functional and survival outcomes in a mouse model of late infantile batten disease. Mol Ther. 2007;15:1782–1788. doi: 10.1038/sj.mt.6300249. [DOI] [PubMed] [Google Scholar]
  10. Chang M, Cooper JD, Sleat DE, Cheng SH, Dodge JC, Passini MA, Lobel P, Davidson BL. Intraventricular enzyme replacement improves disease phenotypes in a mouse model of late infantile neuronal ceroid lipofuscinosis. Mol Ther. 2008;16:649–656. doi: 10.1038/mt.2008.9. [DOI] [PubMed] [Google Scholar]
  11. Daly TM, Lorenz RG, Sands MS. Abnormal Immune function in vivo in a murine model of lysosomal storage disease. Ped Res. 1999;47:757–762. doi: 10.1203/00006450-200006000-00012. [DOI] [PubMed] [Google Scholar]
  12. Dickson P, McEntee M, Vogler C, Le S, Levy B, Peinovich M, Hanson S, Passage M, Kakkis E. Intrathecal enzyme replacement therapy: Successful treatment of brain disease via the cerebrospinal fluid. Mol Genet Metab. 2007;91:61–68. doi: 10.1016/j.ymgme.2006.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Dodge JC, Clarke J, Song A, Bu J, Yang W, Taksir TV, Griffiths D, Zhao MA, Schuchman EH, Cheng SH, O’Riordan CR, Shihabuddin LS, Passini MA, Stewart GR. Gene transfer of human acid sphingomyelinase corrects neuropathology and motor deficits in a mouse model of Niemann-Pick type A disease. Proc Natl Acad Sci U S A. 2005;102:17822–17827. doi: 10.1073/pnas.0509062102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Dodge JC, Clarke J, Treleaven CM, Taksir TV, Griffiths DA, Yang W, Fidler JA, Passini MA, Karey KP, Schuchman EH, Cheng SH, Shihabuddin LS. Intracerebroventricular infusion of acid sphingomyelinase corrects CNS manifestations in a mouse model of Niemann-Pick A disease. Exp Neurol. 2009;215:349–357. doi: 10.1016/j.expneurol.2008.10.021. [DOI] [PubMed] [Google Scholar]
  15. Donsante A, Levy B, Vogler C, Sands MS. Clinical response to persistent, low-level β-glucuronidase expression in the murine model of mucopolysaccharidosis type VII. J Inherit Metab Dis. 2007;30:227–238. doi: 10.1007/s10545-007-0483-4. [DOI] [PubMed] [Google Scholar]
  16. Du H, Cameron TL, Garger SJ, Pogue GP, Hamm LA, White E, Hanley KM, Grabowski GA. Wolman disease/cholesterol ester storage disease: efficacy of plant-produced human lysosomal acid lipase in mice. J Lipid Res. 2008;49:1646–1657. doi: 10.1194/jlr.M700482-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Elleder M. Niemann-Pick disease. Pathol Res Pract. 1989;185:293–328. doi: 10.1016/S0344-0338(89)80006-8. [DOI] [PubMed] [Google Scholar]
  18. Eng CM, Guffon N, Wilcox WR, Germain DP, Lee P, Waldek S, Caplan L, Linthorst GE, Desnick RJ. Safety and efficacy of recombinant human α-galactosidase A replacement therapy in Fabry’s disease. N Eng J Med. 2001;345:9–32. doi: 10.1056/NEJM200107053450102. [DOI] [PubMed] [Google Scholar]
  19. Gadola SD, Silk JD, Jeans A, Illarionov PA, Salio M, Besra GS, Dwek R, Butters TB, Platt FM, Cerundolo V. Impaired selection of invariant natural killer T cells in diverse mouse models of glycosphingolipid lysosomal storage diseases. J Exp Med. 2006;203:2293–2303. doi: 10.1084/jem.20060921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Griffey M, Bible E, Vogler C, Levy B, Gupta P, Cooper J, Sands MS. Adeno associated virus 2-mediated gene therapy decreases autofluorescent storage material and increases brain mass in a murine model of infantile neuronal ceroid lipofuscinosis. Neurobiol Dis. 2004;16:360–369. doi: 10.1016/j.nbd.2004.03.005. [DOI] [PubMed] [Google Scholar]
  21. Griffey MA, Wozniak D, Wong M, Bible E, Johnson K, Rothman SM, Wentz AE, Cooper JD, Sands MS. CNS-directed AAV2-mediated gene therapy ameliorates functional deficits in a murine model of infantile neuronal ceroid lipofuscinosis. Mol Ther. 2006;13:538–547. doi: 10.1016/j.ymthe.2005.11.008. [DOI] [PubMed] [Google Scholar]
  22. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch P, Lim A, Osborne CS, Pawliuk R, Morillon E, Sorenson R, Forster A, Fraser P, Cohen JI, de Saint Basile G, Alexander I, Wintergerst U, Frebourg T, Aurias A, Stoppa-Lyonett D, Romana S, Radford-Weiss I, Gross F, Valensi F, Delabesse E, Macintyre E, Sigaux F, Soulier J, Leiva LE, Wissler M, Prinz C, Rabbitts TH, Le Deist F, Fisher A, Cavazzana-Calvo M. LMO2-Associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003;302:415–419. doi: 10.1126/science.1088547. [DOI] [PubMed] [Google Scholar]
  23. Horinouchi K, Erlich S, Perl D, Ferlinz K, Bisgaier C, Sandhoff K, Desnick R, Stewart C, Schuchman E. Acid sphingomyelinase deficient mice: a model of types A and B Niemann-Pick disease. Nature Genetics. 1995;10:288–293. doi: 10.1038/ng0795-288. [DOI] [PubMed] [Google Scholar]
  24. Jeyakumar M, Norflus F, Tifft CJ, Cortina-Borja M, Butters TD, Proia RL, Perry VH, Dwek RA, Platt FM. Enhanced survival in Sandhoff disease mice receiving a combination of substrate deprivation therapy and bone marrow transplantation. Blood. 2001;97:327–329. doi: 10.1182/blood.v97.1.327. [DOI] [PubMed] [Google Scholar]
  25. Jeyakumar M, Dwek RA, Butters TD, Platt FM. Storage solutions: treating lysosomal disorders of the brain. Nat Rev Neurosci. 2005;6:713–25. doi: 10.1038/nrn1725. [DOI] [PubMed] [Google Scholar]
  26. Jin HK, Carter JE, Huntley GW, Schuchman EH. Intracerebral transplantation of mesenchymal stem cells into acid sphingomyelinase-deficient mice delays the onset of neurological abnormalities and extends their life span. J Clin Invest. 2002;109:1183–1191. doi: 10.1172/JCI14862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Jin HK, Schuchman EH. Ex vivo gene therapy using bone marrow-derived cells: combined effects of intracerebral and intravenous transplantation in a mouse model of Niemann-Pick disease. Mol Ther. 2003;8:876–885. doi: 10.1016/j.ymthe.2003.07.008. [DOI] [PubMed] [Google Scholar]
  28. Kakkis ED, Muenzer J, Tiller GE, Waber L, Belmont J, Passage M, Izykowski B, Phillips J, Doroshow R, Walot I, Hoft R, Neufeld EF. Enzyme replacement therapy in mucopolysaccharidosis I. N Eng J Med. 2001;344:182–188. doi: 10.1056/NEJM200101183440304. [DOI] [PubMed] [Google Scholar]
  29. Kaplan A, Achord DT, Sly WS. Phosphohexosyl components of a lysosomal enzyme are recognized by pinocytosis receptors on human fibroblasts. Proc Natl Acad Sci USA. 1977;74:2026–2030. doi: 10.1073/pnas.74.5.2026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Kelo E, Dunder U, Mononen I. Massive accumulation of Man2GlcNAc2-Asn in nonneuronal tissues of glycosylasparaginase-deficient mice and its removal by enzyme replacement therapy. Glycobiol. 2005;15:79–85. doi: 10.1093/glycob/cwh145. [DOI] [PubMed] [Google Scholar]
  31. Kieler CL, Maddox L, Bible E, Macauley SL, Griffey MA, Wong M, Sands MS, Cooper JD. Loss of thalamic relay neurons precedes pathological effects upon the cortex of PPT1 null mutant mice. Neurobiol Dis. 2007;25:150–162. doi: 10.1016/j.nbd.2006.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Kornfeld S. Structure and function of the mannose 6-phosphate/insulin like growth factor II receptors. Ann Rev Biochem. 1992;61:307–330. doi: 10.1146/annurev.bi.61.070192.001515. [DOI] [PubMed] [Google Scholar]
  33. Lee WC, Courtney A, Troendle FJ, Stallings-Mann ML, Dickey CA, DeLucia MW, Dickson DW, Eckman CB. Enzyme replacement therapy results in substantial improvements in early clinical phenotype in a mouse model of globoid cell leukodystrophy. FASEB J. 2005;19:1549–1551. doi: 10.1096/fj.05-3826fje. [DOI] [PubMed] [Google Scholar]
  34. Lee JP, Jeyakumar M, Gonzalez R, Takahashi H, Lee PJ, Baek RC, Clark D, Rose H, Fu G, Clarke J, McKercher S, Meerloo J, Muller FJ, Park KI, Butters TD, Dwek RA, Schwartz P, Tong G, Wenger D, Lipton SA, Seyfried TN, Platt FM, Snyder EY. Stem cells act through multiple mechanisms to benefit mice with neurodegenerative metabolic disease. Nat Med. 2007;13:439–447. doi: 10.1038/nm1548. [DOI] [PubMed] [Google Scholar]
  35. Lee WC, Tsoi YK, Troendle FJ, DeLucia MW, Ahmed Z, Dicky CA, Dickson DW, Eckman CB. Single-dose intracerebroventricular administration of galactocerebrosidase improves survival in a mouse model of globoid cell leukodystrophy. FASEB J. 2007;21:2520–2527. doi: 10.1096/fj.06-6169com. [DOI] [PubMed] [Google Scholar]
  36. Lin DS, Macauley SL, Donsante A, Win Y, Levy B, Vogler C, Sands MS. Central nervous system-directed AAV2/5-mediated gene therapy synergizes with bone marrow transplantation in the murine model of globoid cell leukodystrophy. Mol Ther. 2007;15:44–52. doi: 10.1038/sj.mt.6300026. [DOI] [PubMed] [Google Scholar]
  37. Lonser RR, Walbridge S, Murray GJ, Aizenberg MR, Vortmeyer AO, Aerts JM, Brady RO, Oldfield EH. Convection perfusion of glucocerebrosidase for neuronopathic Gaucher’s disease. Ann Neurol. 2005;57:542–548. doi: 10.1002/ana.20444. [DOI] [PubMed] [Google Scholar]
  38. Macauley SL, Sidman RL, Schuchman EH, Taksir T, Stewart GR. Neuropathology of the acid sphingomyelinase knockout mouse model of Niemann-Pick A disease including structure-function studies associated with cerebellar Purkinje cell degeneration. Exp Neurol. 2008;214:181–192. doi: 10.1016/j.expneurol.2008.07.026. [DOI] [PubMed] [Google Scholar]
  39. Macauley SL, Wozniak DF, Kielar C, Tan Y, Cooper JD, Sands MS. Cerebellar pathology and motor deficits in the palmitoyl protein thioesterase 1-deficient mouse. Exp Neurol. 2009 doi: 10.1016/j.expneurol.2009.01.022. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Marodi L, Kaposzta R, Toth J, Laszlo A. Impaired microbicidal capacity of mononuclear phagocytes from patients with type I Gaucher disease: partial correction by enzyme replacement therapy. Blood. 1995;86:4645–4649. [PubMed] [Google Scholar]
  41. Matzner U, Lüllmann-Rauch R, Stroobants S, Andersson C, Weigelt C, Eistrup C, Fogh J, D’Hooge R, Gieselmann V. Enzyme Replacement Improves Ataxic Gait and Central Nervous System Histopathology in a Mouse Model of Metachromatic Leukodystrophy. Mol Ther. 2009 doi: 10.1038/mt.2008.305. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Meikle PJ, Hopwood JJ, Claque AE, Carey WF. Prevalence of lysosomal storage disorders. JAMA. 1999;281:249–254. doi: 10.1001/jama.281.3.249. [DOI] [PubMed] [Google Scholar]
  43. Miranda SR, He X, Simonaro CM, Gatt S, Dagan A, Desnick RJ, Schuchman EH. Infusion of recombinant human acid sphingomyelinase into niemann-pick disease mice leads to visceral, but not neurological, correction of the pathophysiology. FASEB J. 2000;14:1988–1995. doi: 10.1096/fj.00-0014com. [DOI] [PubMed] [Google Scholar]
  44. Neufeld EF, Fratantoni JC. Inborn errors of mucopolysaccharide metabolism. Science. 1970;169:141–146. doi: 10.1126/science.169.3941.141. [DOI] [PubMed] [Google Scholar]
  45. Neufeld EF. Lysosomal storage diseases. Ann Rev Biochem. 1991;60:257–280. doi: 10.1146/annurev.bi.60.070191.001353. [DOI] [PubMed] [Google Scholar]
  46. Otterbach B, Stoffel W. Acid sphingomyelinase-deficient mice mimic the neurovisceral form of human lysosomal storage disease (Niemann-Pick disease) Cell. 1995;81:1053–1061. doi: 10.1016/s0092-8674(05)80010-8. [DOI] [PubMed] [Google Scholar]
  47. Passini MA, Dodge JC, Bu J, Yang W, Zhao Q, Sondhi D, Hackett NR, Kaminsky SM, Mao Q, Shihabuddin LS, Cheng SH, Sleat DE, Stewart GR, Davidson BL, Lobel P, Crystal RG. Intracranial delivery of CLN2 reduces brain pathology in a mouse model of classical late infantile neuronal ceroid lipofuscinosis. J Neurosci. 2006;26:1334–1342. doi: 10.1523/JNEUROSCI.2676-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Passini MA, Macauley SL, Huff MR, Taksir TV, Bu J, Wu IH, Piepenhagen PA, Dodge JC, Shihabuddin LS, O’Riordan CR, Schuchman EH, Stewart GR. AAV vector-mediated correction of brain pathology in a mouse model of Niemann-Pick A disease. Mol Ther. 2005;11:754–762. doi: 10.1016/j.ymthe.2005.01.011. [DOI] [PubMed] [Google Scholar]
  49. Passini MA, Bu J, Fidler JA, Ziegler RJ, Foley JW, Dodge JC, Yang WW, Clarke J, Taksir TV, Griffiths DA, Zhao MA, O’Riordan CR, Schuchman EH, Shihabuddin LS, Cheng SH. Combination brain and systemic injections of AAV provide maximal functional and survival benefits in the Niemann-Pick mouse. Proc Natl Acad Sci U S A. 2007;104:9505–9510. doi: 10.1073/pnas.0703509104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Robbins AR. Isolation of lysosomal α-mannosidase mutants of Chinese hamster ovary cells. Proc Natl Acad Sci USA. 1979;76:1911–1915. doi: 10.1073/pnas.76.4.1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Rome LH, Miller J. Butanedione treatment reduces receptor binding of a lysosomal enzyme to cells and membranes. Biochem Biophys Res Comm. 1980;92:986–993. doi: 10.1016/0006-291x(80)90799-8. [DOI] [PubMed] [Google Scholar]
  52. Sando GN, Neufeld EF. Recognition and receptor-mediated uptake of a lysosomal enzyme, α-L-iduronidase, by cultured human fibroblasts. Cell. 1977;12:619–627. doi: 10.1016/0092-8674(77)90262-8. [DOI] [PubMed] [Google Scholar]
  53. Sands MS, Vogler C, Kyle JW, Grubb JH, Levy B, Galvin N, Sly WS, Birkenmeier EH. Enzyme replacement therapy for murine mucopolysaccharidosis type VII. J Clin Invest. 1994;93:2324–2331. doi: 10.1172/JCI117237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Sands MS, Vogler C, Torrey A, Grubb J, Levy B, Sly WS, Birkenmeier EH. Murine mucopolysaccharidosis type VI: long term therapeutic effects of enzyme replacement and enzyme replacement followed by bone marrow transplantation. J Clin Invest. 1997;99:1596–1605. doi: 10.1172/JCI119322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Sands MS, Davidson BL. Gene therapy for lysosomal storage diseases. Mol Ther. 2006;13:839–849. doi: 10.1016/j.ymthe.2006.01.006. [DOI] [PubMed] [Google Scholar]
  56. Sarna J, Miranda SR, Schuchman EH, Hawkes R. Patterned cerebellar Purkinje cell death in a transgenic mouse model of Niemann Pick type A/B disease. Eur J Neurosci. 2001;13:1873–1880. doi: 10.1046/j.0953-816x.2001.01564.x. [DOI] [PubMed] [Google Scholar]
  57. Schuchman EH, Desnick RJ. Niemann-Pick disease types A and B: acid sphingomyelinase deficiencies. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The Metabolic and Molecular Bases of Inherited Disease. McGraw Hill; New York: 2001. pp. 3589–3610. [Google Scholar]
  58. Schuchman EH. The pathogenesis and treatment of acid sphingomyelinase-deficient Niemann-Pick disease. J Inherit Metab Dis. 2007;30:654–663. doi: 10.1007/s10545-007-0632-9. [DOI] [PubMed] [Google Scholar]
  59. Shihabuddin LS, Numan S, Huff MR, Dodge JC, Clarke J, Macauley SL, Yang W, Taksir TV, Parsons G, Passini MA, Gage FH, Stewart GR. Intracerebral transplantation of adult mouse neural progenitor cells into the Niemann-Pick-A mouse leads to a marked decrease in lysosomal storage pathology. J Neurosci. 2004;24:10642–10651. doi: 10.1523/JNEUROSCI.3584-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Ullrich K, Basner R, Gieselmann V, Von Figura K. Recognition of human urine a-N-acetylglucosaminidase by rat hepatocytes. Biochem J. 1979;180:413–419. doi: 10.1042/bj1800413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Yang WW, Dodge JC, Passini MA, Taksir TV, Griffiths D, Schuchman EH, Cheng SH, Shihabuddin LS. Intraparenchymal injections of acid sphingomyelinase results in regional correction of lysosomal storage pathology in the Niemann-Pick A mouse. Exp Neurol. 2007;207:258–266. doi: 10.1016/j.expneurol.2007.06.017. [DOI] [PubMed] [Google Scholar]

RESOURCES