71. RegistryNXT!: New technology to enhance data collection and reporting for the LSD registries

71. RegistryNXT!: New technology to enhance data collection and reporting for the LSD registries

S22 Abstracts/Molecular Genetics and Metabolism 99 (2010) S8–S41 known disease-causing mutation. ERT was initiated. A monitoring sample was sent mis...

54KB Sizes 2 Downloads 32 Views

S22

Abstracts/Molecular Genetics and Metabolism 99 (2010) S8–S41

known disease-causing mutation. ERT was initiated. A monitoring sample was sent mistakenly to Lab A. This showed a small band of HS with trace DS and was interpreted as ‘‘significance uncertain’’. This case demonstrates problematic variability in testing for urinary GAG. doi:10.1016/j.ymgme.2009.10.083

67. Immune cell phenotypes and cytokine response in a mouse model of sialidase deficiency Suleiman Igdourab, Elizabeth Whitea, Abraham Yanga, Bernardo Trigattic, Suleiman Igdouraa, aMcMaster University, Department of Biology, Hamilton, Canada, bMcMaster University, Department of Pathology & Molecular Medicine, Hamilton, Canada, cMcMaster University, Department of Biochemistry & Biomedical Sciences, Hamilton, Canada Sialidase (Neu1) deficiency leads to the accumulation of sialylconjugates within lysosomes of neurons resulting in a fatal progressive neurodegeneration. The interplay between neuroinflammation and neuronal cell death has been poorly understood in Sialidosis patients. In this study, we aim to examine the effect of sialidase deficiency on a model of systemic inflammation, i.e. ApoE knockout mice. The double mutant mice share sialidase deficiency, due to a mutation within the neu1 promoter, with the systemic inflammation associated with deficiency of ApoE lipoprotein. Specifically, we aim to determine the impact of sialidase deficiency on immune cell phenotypes and cytokine milieu in the presence of a pro-inflammatory background. We hypothesize neu1 deficiency in ApoE knockout mice reduces proportions of innate cell and phagocytic immune cell phenotypes and reduces stimulated pro-inflammatory cytokine production. Peripheral blood was collected from sialidase deficient and control mice and immunostained with directly conjugated antibodies for immune cell markers and analyzed by flow cytometry. In addition, serum samples were analyzed for IL-2, IL-4, IL-10 and IFN-gamma by ELISA after invivo lipopolysaccharide or anti-CD3 stimulation. In comparison to controls, the sialidase deficient mice had reduced proportions of CD3+CD8+ cytotoxic T cells, CD3+NK1.1+ NK T cells, CD3 NK1.1+ NK cells, CD11b+Gr 1hi granulocytes, but increased CD3+CD4+ T cells and CD19+ B cells. The sialidase deficient mice produced significantly less IFN-gamma at 4 h and 8 h after anti-CD3 administration. Sialidase deficiency reduces the proportion of early responder immunophenotypes and reduces stimulated IFN-gamma release in ApoE knockout mice. The present data highlights the role of sialidase in the immune system and explains the immune-compromised state of Sialidosis patients. doi:10.1016/j.ymgme.2009.10.084

68. Impact of tumor necrosis factor-alpha deletion on astrolgliosis and microgliosis in the CNS of Sandhoff mice Suleiman Igdouraa,b, Alex Hoopera, Hatim Abou-oufa, Michael Randazzoa, Bernardo Trigattic, aDepartment of Biology, McMaster University, Hamilton, Canada, bDepartment of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada, c Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada Sandhoff disease (SD) is caused by mutations in the HEXB gene, leading to accumulation of the ganglioside GM2 in the CNS of patients, who generally present with poor motor, mental and visual abilities and suffer early mortality. Tumor necrosis factor- (TNF) plays a major role during in neuroinflammation and neuronal death in SD. We have previously generated a double knockout of TNF and HexB in which we demonstrated that TNF deficiency attenuates the neurological deficits in Sandhoff mice. Since TNF is both autocrine and paracrine effector molecule for CNS glia, we investigated and quantified reactive astrogliosis and microgliosis in the CNS of hexb / and hexb / tnf / mice. Paraffin Sections from the brain and spinal cord were immunostained and quantified for the astroglial marker, GFAP and the macrophage marker, MAC3. B When compared to control wild type mice, both hexb / and hexb / tnf / mice show significantly higher numbers of microglia and astrocytes within various regions of the CNS. However, the double knockout mice show significantly lower numbers of astrocytes and microglia in the cerebral cortex and lumbar region of the spinal cord than in observed hexb / mice. These observations were also confirmed by Western blots and qRT-PCR. We propose that down-regulation of TNF expression decreases reactive microgliosis and astrolgliosis in the CNS of Sandhoff mice and as a result improves the neurological outcome of the double knockout mice. Our results suggest that TNF may be a potential therapeutic target for the treatment of Sandhoff disease. doi:10.1016/j.ymgme.2009.10.085

69. The therapeutic potential of small pharmacological molecules in the treatment of Sialidosis Erin M. OLeary, Suleiman A. Igdoura, Department of Biology, McMaster University, Hamilton, Ont., Canada, Department of Pathology & Molecular Medicien, McMaster University, Hamilton, Ont., Canada Sialidosis is an autosomal recessive disorder caused by a dysfunctional sialidase enzyme. Categorized into two phenotypes, Sialidosis type I and II, Sialidosis is a highly heterogeneous disorder with varying ages of onset and pathologies. Currently there is no viable therapy for the treatment of Sialidosis patients. At the molecular level, cells from Sialidosis patients with compound heterozygous mutations show improper enzyme folding, loss of sialidase enzyme activity and subsequent accumulation of sialylconjugates within lysosomes. One promising treatment option is the use of small pharmacological molecules as chaperones, and proteosome inhibitors. In this study, we examined the efficacy of a pharmacological chaperone (Celastrol) as well as a proteosomal inhibitor (MG-132) in the rescue of mutant enzymes with altered conformation. We have assessed the impact of the treatments on Sialidosis patient cell lines using sialidase enzymatic assays and lysosomal localization. Our data show that incubation with Celastrol, MG-132 or a combination of the two drugs results in a significant 3.2, 1.3 or 8.1 increase in sialidase enzyme activity, respectively. Our results also revealed a significant impact of the drugs on lysosomal targeting and ganglioside hydrolysis. This promising study presents a novel biological criteria to assess the efficacy of small molecules and points to a potential therapeutic strategy for the treatment of Sialidosis. doi:10.1016/j.ymgme.2009.10.086

70. Review of 11 patients with NPC1 treated with miglustat Elizabeth Jacklin, Jackie Imrie, Simon Jones, Ed Wraith, Royal Manchester Childrens Hospital, Manchester, England, UK Abstract: Review of 11 patients with NPC1 treated with miglustat. Introduction: Niemann–Pick disease type C (NP-C) is an autosomal recessive disorder characterized by progressive neurological deterioration leading to premature death. The disease is caused by mutations in one of two genes, NPC1 or NPC2, leading to impaired intracellular lipid transport and build-up of lipids in various tissues, particularly the brain. Miglustat, a reversible inhibitor of glycosphingolipid synthesis, has recently been authorized in the European Union, Brazil and South Korea for the treatment of progressive neurological symptoms in adult and pediatric patients, and represents the first specific treatment for NP-C. Methods: This is a review of the patients with NPC 1 that have been treated on Miglustat, at our centre. It shows our clinical findings on 11 patients, all were reviewed 6 monthly. The age they commenced miglustat varied from aged 7 yrs to aged 29 yrs. They have been on treatment for between 2 yrs 8 months and 7 yrs 7 months. The patients are now aged between 12 yrs and 32 yrs. The patients were scored using the clinical scoring system used in the OGT-918 study. Results: The results show an initial stabilization of the disease progression. However this stabilization is not maintained over time and a clinical deterioration was noted in all patients. Summary: Our results suggest that following an initial response, to miglustat the patients will decline in keeping with the suggestion that miglustat can slow the progression of the neurological abnormalities but not prevent them. doi:10.1016/j.ymgme.2009.10.087

71. RegistryNXT!: New technology to enhance data collection and reporting for the LSD registries Christopher Jonesb, J. Alexander Coleb, Jodi Cutlerb, Kathleen Fahertya, Catherine Koeppera, Marcel van Kuijcka, Andy Siegelb, Hemant Virkarc, John Yeea, aGenzyme Corporation, Cambridge, MA, USA, bIBM Interactive, Cambridge, MA, USA, cDigital Infuzion, Gaithersburg, MD, USA The LSD Registries program was initiated in 1991, sponsored by Genzyme Corporation. This program includes global registries for Gaucher disease, Fabry disease, MPS I, and Pompe disease. Rare disease registry design involves many scientific and technological challenges. In the last ten years, the LSD Registries have successfully transitioned from paper case report forms to electronic data collection. Now, a multi-year, multi-registry technology and design initiative, known as RegistryNXT!, is underway to redesign the data collection technologies and data reporting systems that underlie the LSD Registries. RegistryNXT! completed a multi-registry planning phase in 2008 and a project to define the Gaucher Reg-

Abstracts/Molecular Genetics and Metabolism 99 (2010) S8–S41 istry reporting requirements and system design in March 2009. A build phase began in 2009 and a launch of the Gaucher Registry on the new RegistryNXT! platform is planned for late 2010. These efforts have identified key needs for Registry participants, including more immediate access to patient data and more flexible, easy-to-use reporting tools. New features, such as an interactive patient case report, will enable near real-time disease management support or the option to share reports with other members of the care team or with patients. The RegistryNXT! platform will also support more rapid analyses of aggregate Registry data for research purposes. A prototype of the RegistryNXT! platform will be available for demonstration. doi:10.1016/j.ymgme.2009.10.088

72. Transforming the development of treatments for lysosomal storage disorders Emil Kakkis, Kakkis EveryLife Foundation, Novato, CA, USA The last 10 years have produced new treatments for many lysosomal storage disorders. Despite the successes, many lysosomal and other rare disorders remain untreated. For some disorders, there are technical challenges that block or delay progress, such as distributing enzymes to the brain. For those diseases that have been successfully treated, substantial delays and increased costs occurred due to the difficult regulatory pathway for these rare disorders where there is no precedent for treatment. While we can be happy with the great progress and the successes, the more difficult to treat disorders that are either more rare or have more ‘‘inconvenient biology’’ may never get treated given the increasing cost and complexity of development. For this reason, a great deal of the good science will not be translated to treatments. Transforming the regulatory process could allow for more rational, scientifically-sound strategies that could provide the opportunity to treat more disorders. Our foundation is supporting an effort to establish a rare biochemical and genetic disease office of drug evaluation at FDA that would establish new guidances and policy on the better ways to design and analyze data from clinical studies of these heterogeneous disorders, as well as ways to qualify appropriate biochemical surrogate endpoints where the science supports their use. These changes would concentrate the right people and policies in one place and lead to more efficient development and more approvals based on clinical endpoints or accelerated approvals based on qualified surrogates. doi:10.1016/j.ymgme.2009.10.089

73. One year follow-up on chaperone therapy for two siblings with Adult TaySachs Disease John (Jack) Keimel, University of Minnesota Background: Adult Tay-Sachs Disease (ATSD) is caused by inadequate HexA activity resulting in GM2 ganglioside accumulation. The most prevalent ATSD mutation, alphaG269S, does not impact HexA enzyme function but instead causes post-translation misfolding and reduced enzyme stability. In-vitro studies have demonstrated that pyrimethamine (PYR) improves alphaG269S mutated HexA stability and transport to lysosomes. Case Report: Two brothers confirmed with ATSD and the alphaG269S mutation had been taking a substrate reduction therapy, miglustat (200mg, tid), for >4 years, but at age 24 and 25 years, showed accelerated decline in coordination and leg muscle strength resulting in inability to rise unassisted from a sitting position or climb stairs. PYR (75 mg qd) at dinner was begun in combination with folic acid (5 mg, qd). Repeat leukocyte HexA assays and hematology tests (CBC) were conducted. A prior short-term case study of the older brother reported leukocyte HexA activity increased (3) for up to 8 weeks following start of daily PYR (Keimel and Charnas, WORLD 2009). Results: Additional follow-up on the older brother showed decline of leukocyte HexA to baseline values after 13 weeks. The younger brother also showed an increase (2) in leukocyte HexA activity, but repeatedly encountered anorexia and vomiting after being on PYR for >4 weeks. Hematologic results remained normal for both. PYR (75 mg, qd) was subsequently cycled 3 weeks ON followed by 3 weeks OFF for both siblings. Additional follow-up will be presented. doi:10.1016/j.ymgme.2009.10.090

74. Immunological aspects of treatment of Pompe disease Priya Kishnania, Suhrad Banugariaa, Stephanie DeArmeya, Joanne Mackeya, Sarah Younga, Deeksha Balia, Dwight Koeberla, Amy Rosenbergb, Yoav Messingera,b, Nancy

S23

Mendelsohna,b, William Rheada,b, Y.T. Chena, aDepartment of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, NC, USA, bDivision of Therapeutic Proteins, Office of Biotechnology Products, Center for Drug Evaluation and Research With the advent of enzyme replacement therapy (ERT) with alglucosidase alpha for Pompe disease, the immunological aspects of treated disease is being uncovered. Several factors impact outcome including extent of skeletal muscle damage, rate of disease progression, Cross Reacting Immunological material (CRIM) status, sustained high antibody titers, defective autophagy and muscle fiber type. Antibody titers in the majority of CRIM positive patients diminish over time and do not appear to neutralize uptake or activity. There are some CRIM positive patients infantile and late onset) with high, sustained antibody titers. We retrospectively analysed 10 CRIM-positive infantile Pompe patients with persistently elevated titers at >1:51,200 after 6 months on ERT. The median age at start of ERT was 7 months (range 5–13 months). 2 patients, ages 6 and 7 months respectively were invasively ventilated at start of ERT. The outcome in all 10 patients was poor. Eight out of 10 patients died (median 29.1 months (range 18–40.7 months). This included the 2 patients who were vented at baseline. The 2 surviving patients became vent dependent at ages 18.4 and 26.5 months. These data suggest that patients with high and sustained antibody titers (irrespective of CRIM status) show an attenuated response to ERT. Given this poor outcome, there is a need for immune modulation in patients at risk for development of high sustained titers. Lessons learned from Pompe disease will shed light on treatment strategies for conditions where a foreign protein elicits robust antibody responses that interfere with product efficacy. doi:10.1016/j.ymgme.2009.10.091

75. Fabry International Network – Fabry patient survey Adrian (Ed) Koninga, Rune Sedala, Erica Schenkb, Kees Bosmanc, Lut De Baered, Marlene Koninge, aFabry International Network, Bergen, Norway, bFabry International Network, Oosterwolde, Netherlands, cFabry International Network, Drachten, Netherlands, d Fabry International Network, Melsele, Belgium, eFabry International Network, Edmonton, Canada Fabry disease is a very rare inherited life threatening disease that affects only about 5000–10,000 people worldwide and is caused by a lack of an enzyme that breaks down fatty deposits, resulting in debilitating pain, organ failure, and premature death. The only effective treatment is enzyme replacement therapy (ERT). In 2005 the Fabry International Network (FIN) was incorporated in the Netherlands as non profit organizations. To date there are 23 member organizations in 21 countries in North and South America, Europe and the South Pacific. FIN has developed an electronic online survey. The objective of this survey is to obtain input and feedback from the current 23 FIN member organizations and their respective leader(s) as well as the FIN Medical Advisory Board. This tool is one method to improve two way communications, with the goal to reach consensus on FINs priorities and action plan for the next 12–18 months. In addition the survey will:  Better help FIN determine its long term goals and objectives.  Enhance development of a data base of all Fabry organizations, their leadership contacts as well as their particular areas of interest/skills that could one day be utilized and shared around the world.  Provide useful data on the needs of Fabry patients and the organizations that represent patients as well as to understand key concerns, issues and unmet needs. The hope and plan is that data collected can be shared internationally.

doi:10.1016/j.ymgme.2009.10.092

76. Fabry International NetworkWorldwide Fabry patient nonprofit organization Adrian (Ed) Koninga, Rune Sedala, Erica Schenkb, Kees Bosmanc, Lut De Baered, Marlene Koninge, aFabry International Network, Bergen, Norway, bFabry International Network, Oosterwolde, The Netherlands, cFabry International Network, Drachten, The Netherlands, dFabry International Network, Melsele, Belgium, eFabry International Network, Edmonton, Canada Fabry disease is a very rare inherited life threatening disease that affects only about 10,000 people worldwide. It takes on average of 17 years to diagnose this metabolic storage disorder. Patients lack sufficient enzyme to break down fatty deposits,