Are you newly diagnosed?
Learn More
Children's Hospital of Philadelphia

#MSDCon2024

Natural History Study of Multiple Sulfatase Deficiency: updates

Laura Adang

Attending Physician

Children’s Hospital of Philadelphia

Laura A Adang

Multiple sulfatase deficiency (MSD) is an ultra-rare, fatal neurodegenerative disorder with emerging therapeutic options. This includes an AAV9-based approach recently funded through the Foundation for the NIH Bespoke Gene Therapy Program (PIs Adang, Ahrens-Nicklas) as well as a drug repurposing strategy in Europe (PI: Schlotawa, Remedi4All). MSD is the result of dysfunctional sulfatase activation by SUMF1-encoded formylglycine-generating enzyme (FGE). The resulting phenotype arises from the combined of loss of activity of all sulfatases, although the primary neurologic impact is from loss of the enzymes associated with mucopolysaccharidosis III (MPS IIIA/D) and metachromatic leukodystrophy (MLD). The spectrum of neurologic disease includes a common subtype with stereotyped regression during the toddler years, similar to late infantile MLD. Children also experience an accumulating burden of systemic disease, which can include ichthyosis, hepatosplenomegaly, and dysostosis multiplex. With several investigational drug candidates undergoing clinical planning, there is an urgent unmet need to identify optimal patient-centric clinical endpoints.

Our prior work defined distinct phenotypes of MSD (severe and attenuated), which correlate with genotype and a novel biomarker, glucosaminoglycan non-reducing ends (GAG-NREs). Despite its rarity, we have enrolled more than 40 children in our ongoing natural history study, which represents a large proportion of the known global population. This ongoing study includes collection of historical medical records, longitudinal administration of clinical outcome assessments (COA) in parallel with the collection of biospecimens. This work leverages the resources of the Rare Disease Clinical Research Network (RDCRN)-funded Global Leukodystrophy Initiative Clinical Trials Network (GLIA-CTN), which supports the regulatory infrastructure of the MSD natural history study. The disease severity and complexity of MSD requires a patient-centric approaches for clinical trial design, utilizing concepts of interest most meaningful to patients and families.

Biochemical signatures of disease severity in Multiple Sulfatase Deficiency

Rebecca Ahrens-Nicklas

Assistant Professor

Children’s Hospital of Philadelphia

Laura A Adang, Samar Mowafy, Zackary M Herbst, Zitao Zhou, Lars Schlotawa, Karthikeyan Radhakrishnan, Brenna Bentley, Vi Pham, Emily Yu, Nishitha R Pillai, Paul J Orchard, Mauricio De Castro, Adeline Vanderver, Marzia Pasquali, Michael H Gelb, Rebecca C Ahrens-Nicklas

Sulfatases catalyze essential cellular reactions, including degradation of glycosaminoglycans (GAGs). All sulfatases are post-translationally activated by the formylglycine generating enzyme (FGE) which is deficient in Multiple Sulfatase Deficiency (MSD), a neurodegenerative lysosomal storage disease. Historically, patients were presumed to be deficient of all sulfatase activities; however, a more nuanced relationship is emerging. Each sulfatase may differ in their degree of post-translational modification by FGE, which may influence the phenotypic spectrum of MSD. Here, we evaluate if residual sulfatase activity and accumulating GAG patterns distinguish cases from controls and stratify clinical severity groups in MSD. We quantify sulfatase activities and GAG accumulation using three complementary methods in MSD participants. Sulfatases differed greatly in their tolerance of reduction in FGE-mediated activation. Enzymes that degrade heparan sulfate (HS) demonstrated lower residual activities than those that act on other GAGs. Similarly, HS-derived urinary GAG subspecies preferentially accumulated, distinguished cases from controls, and correlated with disease severity. Accumulation patterns of specific sulfatase substrates in MSD provide fundamental insights into sulfatase regulation and will serve as much-needed biomakers for upcoming clinical trials. This work highlights that biomarker investigation of an ultra-rare disease can simultaneously inform our understanding of fundamental biology and advance clinical trial readiness efforts.

Recent Progress on the Identification of Small Molecule Reactivators of Sulfatases

Matthias Baud

Dr/Associate Professor

University of Southampton

Matthias Baud

Encoded by the SUMF1 gene, the Formylglycine Generating Enzyme (FGE) is inactivated by mutation in virtually all Multiple Sulfatase Deficiency (MSD) cases. These mutations generally reduce the stability of FGE, preventing it to adopt a functional fold and perform its normal cellular function, that is the downstream activation of the sulfatases enzymes by post-translational modification. FGE inactivation is a hallmark of MSD.

Our work focuses on the search of ligands binding at the surface of FGE. We hypothesise that these ligands, when optimised, will act as “pharmacological chaperones” or “molecular plasters”, stabilising the fold and function of FGE mutants found in MSD, and reactivating their cellular function. With our collaborators, we have combined compounds screening assays to identify hit molecules as starting points, molecular modelling and chemical synthesis for synthetic optimisation, and cellular assays to determine the cellular mode of action and therapeutic potential of the new molecules/compounds identified. We will present our progress and recent results in our search of small molecule reactivators of FGE, along with the challenges encountered and future opportunities. We will disclose our recent findings on the “druggability” of FGE using fragment screening and X-ray crystallography, and our latest efforts to use this knowledge for the assembly of higher affinity ligands to stabilise FGE. Finally, we will also discuss the design/preparation/evaluation of Tazarotene and Bexarotene derivatives, two previously FDA approved drugs which were recently shown to upregulate FGE activity (though via a distinct/indirect cellular mechanism) an structure-activity relationship (SAR) data gathered to date.

Collaboration is Everything: Newborn Screening Advocacy for Rare Diseases

Lesa Brackbill

Director of Advocacy

Leukodystrophy Newborn Screening Action Network

Lesa Brackbill

The Newborn Screening system is complex with many stakeholders involved, and it can be difficult to know where families can make a difference. Learn more about Newborn Screening (including details about how the system works), why coalition building with the Metachromatic Leukodystrophy community will be critical for MSD, and how a collaborative approach can change everything.

Patient-derived NGN2-induced neurons recapitulate biochemical phenotypes of Multiple Sulfatase Deficiency in vitro.

Margaret M. Cassidy

Graduate Student

Children’s Hospital of Philadelphia/University of Pennsylvania

Margaret M. Cassidy, Vi Pham, Livia Sertori Finoti, Kaitlyn King, Parith Wongkittichote, Xinying Hong, Rebecca C. Ahrens-Nicklas

Multiple Sulfatase Deficiency (MSD) is an ultra-rare lysosomal storage disorder (LSD)caused by mutations in the SUMF1 gene. The SUMF1 gene encodes formylglycine-generated enzyme (FGE), the protein responsible for activating all sulfatases in the cell. The reduction or complete loss of FGE causes detrimental accumulation of sulfated substrates in the lysosome, including glycosaminoglycans (GAGs), sulfolipids, and steroid sulfates. Neurological regression is a primary clinical feature of MSD, which suggests that neural cell types are particularly intolerant to dysfunctional lysosomal flux. Therapeutics targeting MSD are currently limited, and there is an urgent need for a reproducible in vitro model of MSD that recapitulates biochemical phenotypes seen in the patient population. To fulfill this need, we recently generated NGN2-induced neurons (NGN2-iN) from induced pluripotent stemcells (iPSC) derived from a patient with MSD. We use a battery of biochemical and molecular techniques to demonstrate that our model reproduces biochemical phenotypes associated with MSD. Patient NGN2-iN display hallmark pathology of MSD seen in humans, including reduced activity of multiple sulfatases and lysosomal accumulation as compared to isogenic controls. Over time, these neuronsalso demonstrate progressive accumulation of GAG subspecies associated with mucopolysaccharidosis type III. Interestingly, these phenotypes worsen in more mature (Day 11) MSD NGN2-iN, as compared to immature (Day 3) MSD NGN2-iN andiPSCs. This observation suggests that neurons are selectively vulnerable to the loss of FGE, and that dysfunction worsens over neuronal maturation. We anticipate that our model will be utilized to compare the selective vulnerability of individual CNS cell types, further our current understanding of MSD pathophysiology, and ultimately aid the development of therapeutics.

The Accelerating Medicines Partnerships® (AMP®) Bespoke Gene Therapy Consortium (BGTC) Regulatory Playbook

Brad Garrison

Sr. Project Manager

Foundation for the National Institutes of Health

Brad Garrison

Over 10,000 known diseases are characterized as “rare” affecting fewer than 200,000 people in the United States. Collectively, these diseases affect about 30 million people in the United States – a significant proportion of the population. About 80% of rare diseases are monogenic where known alterations in a single gene are responsible for the clinical manifestations that result in significant morbidity and mortality. Monogenic diseases are potential candidates for gene therapies because the defective gene can be corrected or replaced thereby restoring gene function and stopping or reversing the disease state. Gene therapies have the potential to provide long-term benefit for patients where there currently is no treatment – a prospect that would be transformational in the treatment of many serious and life-threatening rare disorders.

The Bespoke Gene Therapy Consortium (BGTC) was formed by the Foundation for the National Institutes of Health (FNIH) as part of the Accelerating Medicines Partnership® programs to address the challenge of developing AAV gene therapies for diseases of no commercial interest. It brings together 35 member organizations, including the NIH, FDA, life sciences / biopharmaceutical companies, nonprofits, and patient advocacy groups to collaborate in a pre-competitive space. These partners possess extensive experience in designing, developing and manufacturing AAV-based gene therapies for use in clinical trials.

One objective of the BGTC is to streamline navigation of the regulatory pathway for AAV gene therapies.

To achieve this objective, BGTC has developed a Regulatory Playbook, which is a collection of resources and best practices that reflect a sponsor’s typical regulatory journey from research and development (R&D) to pre-clinical through clinical development and ID preparation/submission. BGTC released playbook version 1.0 on February 6, 2024 to provide an initial framework for gene therapy developers. Subsequent playbook versions will incorporate learnings from up to eight rare disease clinical trials being that are part of the BGTC portfolio. With the BGTC leading the charge and drawing upon the collective experience, expertise, and passion of the scientific community, this playbook aims to serve as a guiding light, enabling researchers and developers to bring safe, effective, and transformative gene therapies to patients in need.

The impact of Multiple Sulfatase Deficiency on children & families: caregiver perspectives

Francesco Gavazzi

Research Staff Scientist II

Division of Neurology, Children’s Hospital of Philadelphia

Emily Yu BA, Francesco Gavazzi MD, PhD, Sarah Woidill BS, MS, Vivian Breitling MD, Lars Schlotawa MD, Rebecca Ahrens-Nicklas MD, PhD, Laura Adang MD, PhD, MSTR

Multiple Sulfatase Deficiency (MSD) is an ultra-rare lysosomal storage disorder associated with pathogenic variants in the SUMF1 gene, which is required for sulfatase activation. Despite the known severity of disease, the impact of MSD on affected children, caregivers, and their family is uncharacterized.

Nineteen families affected by MSD participated in our assessments. The Vineland Adaptive Behavior Scales (VABS-3) was administered by a child neurologist to caregivers in the context of an affected individual’s in-person or remote research encounter. Health-related quality of life (HRQOL) surveys were collected electronically, including the Caregiver Priorities and Child Health Index of Life with Disabilities (CPCHILD), Pediatric Quality of Life Inventory Generic Core Scales (PedsQL GC), PedsQL Family Impact Module (FIM), and the Traumatic Brain Injury Caregiver Quality of Life (CareQOL) questionnaires.

All clinical outcome measures demonstrated a range of performance. Most caregivers assessed their children as severely affected (VABS-3 <40 Standard Score across all Domains and Adaptive Behavior Composite Score: n=10/19; 52.63%), with the lowest median scores in the Communication and Motor Skills domains. The Communication domain’s mean performance was significantly lower than the Socialization and Daily Living Skills domains. The Motor Skills domain was significantly lower than the Socialization domain only. The CPCHILD (n=12) identified items within the Comfort and Emotions and Overall Health domains as top priorities when preserving affected individuals’ quality of life. The Comfort and Emotions domain’s mean performance was significantly higher than all other domains. The CPCHILD overall HRQOL remained high (median 80%, IQR 50%). The PedsQL GC (n=14) demonstrated higher mean performance in the Emotional than the School/Work domain only. The CareQOL (n=15) identified caregivers’ significantly higher frequency of a sense of loss for the affected individual and feelings of entrapment. The PedsQL FIM (n=13) demonstrated that there was no significant difference between family functioning and parent HRQOL performance.

In this study, we demonstrated that MSD has profound, global effects on both affected individuals and their caregivers as measured by the VABS-3 and HRQOL surveys. This data will be integral to identifying Health Concepts and related Concept of Interests to inform future clinical trial design.

Newborn Screening for Lysosomal Storage Disorders and Beyond (including MSD)

Michael Gelb

Professor

University of Washington

Michael Gelb

The assay for newborn screening of MLD is well developed. A pilot study on 30,000 newborns in Washington state found 1 case of high prognosis for MLD, no false positives. Data shows that 40 out of 40 newborn blood spots from patients who were diagnosed with MLD tested positive in this assay suggesting that the false negative rate is essentially zero. Subsequently, pilot studies have started in UK, France, Italy, Germany, and Austria. In Germany, 4 cases with high prognosis for MLD were identified, those with early onset disease proceeded toward treatment channels.

Newborn screening for MLD can be readily multiplexed using mass spectrometry with other lysosomal diseases that have been recently added to expanded newborn screening panels. Reagents including quality control materials are available for use by newborn screening laboratories.

Limited data suggest that MSD patients will be picked up during newborn screening of MLD, but additional data using newborn blood spots is important to confirm this observation.

Efficacy of a scAAV9/SUMF1 viral vector for the treatment of Multiple Sulfatase Deficiency

Steven Gray

Associate Professor

University of Texas Southwestern

Maximiliano Presa, Rachel M Bailey, Somdatta Ray, Lauren Bailey, Saurabh Tata, Tara Murphy, Harold Combs, Steven J Gray, Cathleen Lutz

Multiple Sulfatase Deficiency (MSD) is a rare autosomal recessive disorder characterized by deficient enzymatic activity of all known sulfatase. MSD patients frequently carried two loss of function mutations in the SUMF1 gene, encoding a sulfatase modifying factor which activates 17 different sulfatases. MSD patients shown common features of mucopolysaccharidosis, and metachromatic leukodystrophy, including neurologic impairments, developmental delay, and visceromegaly. There are currently no approved therapies for MSD patients. Gene therapy may provide a meaningful and long-term therapeutic benefit for MSD patients by delivering a functional copy of the SUMF1 gene to patient cells. We developed a self-complementary vector encoding a codon-optimized human SUMF1 gene (scAAV9/SUMF1), the unaltered design of which could be appropriate for human use. Using a severe Sumf1 knock-out (KO) mouse model of MSD, we tested the benefits of treatment with our vector in mice shortly after birth and later in the disease course by administering virus into the cerebrospinal fluid (CSF) alone or in combination with an intravenous injection. CSF administration of scAAV9/SUMF1 was sufficient to rescue early lethality, normalize behavior and improve newly identified visual and cardiac dysfunctions in KO mice, even when given at an advanced disease stage. We found dose-dependent and long-term restoration of sulfatase activity in tissues, which is accompanied by decreased accumulation of glycosaminoglycans, lysosomal defects and neuroinflammation in the brain. Even at the highest dose tested, scAAV9/SUMF1 was well-tolerated without toxicity from high transgenic SUMF1 expression in KO and wild-type mice or in rats that were treated as part of a GLP-toxicology study. Overall, our preclinical results attest to the safety of scAAV9/SUMF1 delivery and predict a benefit of this treatment to MSD patients.

GLP toxicology findings for AAV9/SUMF1 intrathecal or intracisternal magna administration in WT rats

Steven Gray

Associate Professor

UT Southwestern Medical Center

Steven J. Gray

AAV9/SUMF1 gene therapy is being considered as a treatment for Multiple Sulfatase Deficiency. A GLP toxicology study to evaluate the safety of AAV9/SUMF1 in rats, following lumbar intrathecal (IT) or cisterna magna (ICM) administration, was carried out by Charles River Laboratories. Cohorts consisted of 5 males and 5 females, injected with AAV9/SUMF1 (5E11, 2E12, and 4.8E12 vg/rat IT, as well as 2E12 vg/rat ICM), and animals were examined twice daily for mortality/moribundity along with a detailed clinical examination and weight acquisition performed weekly. Cohorts were sacrificed at 29 days to evaluate tolerability of AAV9/SUMF1 at the peak of transgene expression for acute toxicities. A long-term cohort was sacrificed at 181 days post-administration to evaluate signs of long-term adverse effects and/or possible resolution of short-term findings. Overall, the study conclusion was that AAV9/SUMF1 was well-tolerated across both time points and all doses. No difference was seen in the findings of the IT versus ICM groups. Findings are as follows for each dose group at the day 29 time point: 5E11 vg: low-grade spinal nerve root degeneration. 2E12 vg: Minimal severity inflammation/degeneration of lumbar and thoracic dorsal root ganglia (DRG), occasional spinal nerve root degeneration, and low-grade white matter degeneration. 4.8E12 vg: Similar histology findings as with the 2×1012 vg IT group, but trending slightly higher incidence/severity (occasional moderate severity findings); also minor maturation delays in peripheral sensory nerve conduction were observed, but graded as negligible and not neuropathic. All findings were partially or fully resolved by day 181.

United Multiple Sulfatase Deficiency Foundation Research Strategy Map

Matthew Jarpe

Project Lead, Research Strategy Map

United MSD Foundation

Matthew Jarpe

With the announcement of the Bespoke Grant in May 2023, which will fund Phase I/II clinical trials for AAV9 gene therapy, the United MSD Foundation has an opportunity to examine its funding strategy for MSD research projects. Inspired by the Sanfilippo Children’s Foundation’s Global Research Roadmap, the Foundation has begun drafting its own Research Strategy Map to guide its funding over the next 5 years. The first draft of this Research Strategy Map will be presented and the patient family and research communities will have an opportunity to respond and shape the final document.

The REMEDi4ALL drug repurposing consortium

Donald Lo

Director of Medicines Development

EATRIS

Donald Lo, Martin de Kort and Alicia Soler Canton

REMEDi4ALL (repurposing medicines for all) is an European Union-funded research initiative to aimed to create a sustainable and globally connected platform and drive forward the repurposing of medicines in Europe. The REMEDi4ALL consortium (https://remedi4all.org/) brings together a unique combination of expertise to address the complexities of drug repurposing with capacities accessible to the research community. Led by EATRIS, the European infrastructure for translational medicine (Ussi et al., 2017), (Gilliland et al., 2016), (https://eatris.eu/), 24 organizations have joined forces to build a alleviate systemic bottlenecks in drug repurposing. It aims to construct an integrated platform encompassing advanced drug research facilities with clinical and translational research expertise, clinical operations, patient engagement methodologies, education & training, regulatory experts, health technology assessment, building a globally connected community of practice that will also include a network of funders.

As a disease-agnostic and patient-centric platform, REMEDi4ALL has the potential to boost drug repurposing or repositioning of already approved, discontinued, shelved or investigational therapeutics by improving the ecosystem in Europe as well as globally. Its ultimate aim is to unlock the full potential drug repurposing by reducing development times and costs for new therapeutic opportunities for conditions with high unmet need.

REMEDi4ALL launched in late 2022 with 4 initial drug repurposing projects, including one focused on MSD, in various development stages and addressing different challenges in drug repurposing (e.g., drug screening, reformulation, combination therapy and multinational clinical trials) and in different disease areas (e.g., oncology, infectious disease, ultra-rare genetic diseases). The philosophy of the platform is to work with multidisciplinary teams in a targeted and patient-centric manner and for each project focus on the critical path to bring an existing drug substance or drug product through the approval process and into clinical practice.

Gilliland, C. T., Zuk, D., Kocis, P., Johnson, M., Hay, S., Hajduch, M., et al. (2016). Putting translational science on to a global stage. Nat Rev Drug Discov 15, 217–8. doi: 10.1038/nrd.2016.33.

Ussi, A. E., de Kort, M., Coussens, N. P., Aittokallio, T., and Hajduch, M. (2017). In Search of System-Wide Productivity Gains – The Role of Global Collaborations in Preclinical Translation. Clin Transl Sci 10, 423–425. doi: 10.1111/cts.12498.

Finding starting points for structure-based drug design

Hartmut Niemann

Prof. Dr.

Bielefeld University

Hartmut H. Niemann, Julia L. Kowal, Niama Ezzaidi, Jan Wollenhaupt, Karthikeyan Radhakrishnan, Tatjana Pietrowski, Manisha Thangarasah, Willy Keller, Sarfaraz Alam, Manfred S. Weiss, Matthias G. J. Baud

Background:
The key defect in Multiple Sulfatase Deficiency (MSD) is the loss of function of an essential protein called formylglycine-generating enzyme (FGE). In MSD patients, mutations in the gene coding for FGE compromise FGE’s function, often due to incorrect protein folding and low intracellular protein stability. We want to develop drug candidates that stabilize FGE in cells and thereby rescue its function. The binding of a small molecule to a protein often increases the protein’s stability. Hence, we plan to develop small molecule drugs that bind to FGE and stabilize its structure. Such molecules are called pharmacological chaperones. Pharmacological chaperones have recently become a treatment option for other lysosomal storage diseases. For example, Migalastat was approved for treatment of Fabry’s disease.

Results:
We use purified FGE protein to search for very small molecules, so-called fragments, which snuggly bind to grooves on the surface of FGE. So far, we have tested around 600 fragments and found more than two dozen fragments that bind to FGE. Fragments are smaller than usual drugs and their binding to FGE is generally not strong enough to result in sufficient stabilization of FGE. Therefore, the small fragments serve as starting point for optimization into larger and tighter-binding drug-like molecules. Our collaborators from medicinal chemistry perform this optimization. For efficient optimization, structural information about the interaction between the fragments and FGE is essential. X-ray crystallography, the method that we use to search for FGE-binding fragments, does reveal where on FGE a small molecule binds and which atoms of the molecule are presumably important for the binding. Our experiments showed that most of the fragments bind to one of four specific sites on FGE. Three of these sites are different from the active site, suggesting that their binding will not impair FGE’s normal function, i.e. the activation of its sulfatase substrates. Our results suggest that FGE should be druggable, i.e. the development of pharmacological chaperones should be possible in principle. Moreover, the almost 30 fragments that we found to bind to FGE and the detailed visualization of their binding mode is a treasure trove for medicinal chemists to rationally design larger optimized molecules that will eventually be tested for their therapeutic effect in cell culture or in animal models for MSD.

Single vs. dual transgene ex vivo gene therapy for multiple sulfatase deficiency

Vi Pham

PhD Candidate

University of Pennsylvania, Children’s Hospital of Philadelphia

Vi Pham, Lucas Tricoli, Parith Wongkittichote, Xinying Hong, Lars Schlotawa, Stefano Rivella, Rebecca C. Ahrens-Nicklas

Background:
Multiple sulfatase deficiency (MSD) is an ultra-rare, inherited lysosomal storage disorder characterized by the functional deficiency of all sulfatase enzymes. MSD is caused by mutations in the gene SUMF1, encoding a sulfatase modifying factor that activates 17 downstream sulfatases. MSD patients present with combined features of single-sulfatase disorders such as the mucopolysaccharidoses and metachromatic leukodystrophy (MLD). Currently, there are no disease-modifying therapies for MSD. We aimed to develop ex vivo gene therapy with hematopoietic stem cell transplant for MSD, as this approach has proven successful for related disorders.

Methods:
We designed lentiviral vectors encoding either 1) SUMF1 or 2) a downstream sulfatase ARSA that is greatly implicated in the disease. ARSA ex vivo gene therapy is approved by the EMA for MLD and has the potential to be repurposed for patients with MSD.

Results:
In vitro, MSD patient fibroblasts transduced with the lentiviral vectors demonstrated improved biochemical phenotypes including rescue of sulfatase activities and glycosaminoglycan (GAG) accumulation. In vivo, both lentiviral vectors proved efficacious in rescuing biochemical and behavioral phenotypes in MSD mice that underwent hematopoietic stem cell transplant with ex vivo gene therapy. To further augment outcomes, we co-transduced MSD patient fibroblasts with both SUMF1 and ARSA vectors and found that co-expressing these transgenes greatly enhanced biochemical rescue in vitro. We will present the results of our ongoing work evaluating the efficacy of this innovative dual-transgene approach in MSD mice after ex vivo gene therapy.

Tracking Clinical and Biochemical Changes Post Bone Marrow Transplant in Multiple Sulfatase Deficiency – A 3-Year Follow-Up

Nishitha Pillai

Assistant Professor

University of Minnesota

Nishitha R Pillai, Ning Liu, Rebecca Ahrens Nicklas, Laura Adang, Julie B Eisengart, Grace Bronken, Ashish Gupta, Troy Lund, Chester B Whitley, Sarah H Elsea and Paul J Orchard

Background:
Multiple sulfatase deficiency (MSD) is an exceptionally rare neurodegenerative disorder due to the absence or deficiency of 17 known cellular sulfatases. Two siblings with attenuated MSD underwent hematopoietic cell transplantation (HCT), evaluating the possibility of lysosomal enzymatic cross-correction from the donor cells.

Methods:
A retrospective data analysis was done on the two siblings who underwent HCT. The data collected included biomarkers including urine glycosaminoglycans, urine sulfatides, untargeted metabolomics in plasma and enzyme assays. The data from neuropsychology evaluation was also collected up to 3 years after HCT.

Results:
Biochemical correction of all the previously deficient enzyme activity in the leukocytes was evident within 3 months post HCT. Urine heparan and dermatan sulfate along with urine sulfatides were normal in the pre and post HCT samples due to the milder phenotype. While single, unique biomarkers were not identified for MSD, multiple consistent metabolic perturbations across patients were noted. Specifically, glycerophospholipids constituted the predominant category of perturbed sub-pathways in both siblings. Arylsulfates and fatty acids and derivatives, exhibited substantial perturbations, representing the second or third largest proportion of perturbed metabolites across the siblings. Additionally, energy-related sub-pathways such as bile acids, cholesterols, sterols, nucleotides, and various amino acids were disrupted. Notably, the relative levels of disruption across these sub-pathways between the siblings are consistent. These results indicate that MSD induces a broad state of metabolic dysregulation. The impact of HCT were monitored by comparing metabolomic profiles pre- and post-HCT. An almost instant neutralization effect introduced by HCT was observed. The correction of abnormalities was further quantitatively analyzed in every sequential post-HCT samples, while the initial post-HCT samples collected at 3 months showed a significant correction, with more than 70% of abnormal concentration levels falling within the normal range. This correction persisted in subsequent post-HCT samples, demonstrating the effectiveness of HCT. These finding align with the biochemical enzyme corrections observed in the patients.

Conclusion:
These data suggest biochemical benefit post-transplant, although the clinical implication of these corrections are unknown, particularly in the context of the siblings’ attenuated phenotype. Long-term follow-up is necessary to evaluate the therapeutic benefit of HCT in MSD.

Repurposing of tazarotene– update on preparations for the first clinical trial for MSD

Lars Schlotawa

MD/Leading Consultant

University Medical Centre Goettingen, Department of Paediatrics and Adolescent Medicine

Lars Schlotawa, Donald Lo, Martin de Kort, Rok Dreu, Alicia Soler, Aimo Kannt, Tanja Rossmanith, Thomas Asendorf, Stephanie Dauth, Sabrina Zierof, Alan Finglas, Christin Jonetzko, Rebecca C Ahrens-Nicklas, Laura A Adang, Robert Gurke, Jutta Gärtner

Despite growing efforts Multiple Sulfatase Deficiency (MSD, MIM #272200) is still an untreatable disease with a high unmet need for a therapy. Among different approaches like gene therapy and haematopoietic stem cell transplantation identification of small molecules has been a part of the MSD research strategy for therapy development. The fastest way towards a therapy for a yet untreatable condition can be repurposing or repositioning of existing drugs, originally designed and licensed for a different disease entity. Knowledge on formulation, toxicity, dose, safety, and unwanted effects saves time and costs on lengthy processes of pharmacological, pre-clinical, and early clinical development and sometimes post marketing observations.

A high throughput drug screening on MSD patient derived cells identified tazarotene, a third generation retinoic acid derivative, to reverse disease pathology in-vitro. Tazarotene is currently licensed for topical treatment of plaque psoriasis and acne but has been investigated as an oral treatment for the same indications until 2004 in phase I-III trials with approximately 1500 participants. Tazarotene in its oral formulation has been proven and effective for the given indication. Nevertheless it has not benn licensed by the FDA because of likely severe unwanted effects inert to the pharmacological group of retinoids and the lack of trials showing benefit over already existing, licensed retinoids.

Tazarotene for its use in MSD has been chosen as a demonstrator projects by a consortium funded by the European Union, REMEDI4ALL, consisting of 24 members from academia, industry, patient organisations, and regulators. REMEDI4ALL promotes drug re-purposing for various diseases including rare diseases. With the help of REMEDI4ALL a crucial target product profile and gap analysis paved the way for a reformulation, evaluation and production of oral tazarotene for its use in MSD patients, collaboration with pharma industry for access on existing toxicity, pre-clinical, and clinical data on oral tazarotene for reference and regulatory purposes, trial design and study protocol. The efforts cumulated in a first scientific advice meeting with regulators ultimately leading to the first clinical trial in MSD patients supposed to start by the end of 2024.

Development of a rigorous approach to performing Real-World Evidence- based natural history studies in leukodystrophies

Anjana Sevagamoorthy

Clinical Research Associate II

Children’s Hospital of Philadelphia

Anjana Sevagamoorthy MBBS, MPH, Francesco Gavazzi M.D., Ph.D., Omar Sherbini MPH, Ariel Vincent B.S., Russel D’Aiello M.S., Nicholson Modesti, B.S., Sylvia Mutua B.A., Emily Yu B.A., Sarah Woidill M.S., Johanna L. Schmidt MPH, MGC, LCGC, Amy Pizzino M.S., LCGC, Justine Shults Ph.D., Adeline Vanderver M.D., Laura Adang M.D., Ph.D., MSTR

Background:
Leukodystrophies are rare genetic disorders affecting myelination. Emerging therapeutics primarily target the pre-symptomatic population, but there is a knowledge gap on the impact of disease across the lifespan. To address this, we propose a standardized approach to performing retrospective natural history studies using existing medical records.

Methods:
As part of Global Leukodystrophy Initiative Clinical Trials Network (GLIA-CTN), we developed Standard Operating Procedures (SOPs) that delineated all study processes related to staff training, source documentation, and data sharing. Additionally, the SOP detailed the standardized approach to data extraction including diagnosis, clinical presentation, and medical events, such as age at gastrostomy tube placement. The key variables for extraction were selected through face validity, and common electronic case report forms (eCRF) across leukodystrophies were created to collect analyzable data. To enhance the depth of the data, clinical notes are extracted into “original” and “imputed” encounters, with imputed encounter referring to a historic event (e.g., loss of ambulation 3 months prior). Retrospective Functional Assessments are assigned by child neurologists, using a blinded dual-rater approach and any discrepancies are adjudicated by a third rater. Upon completion of extraction, data source verification is performed. Data missingness was evaluated using statistics.

Results:
To date, the records from 922 subjects have undergone this process across the GLIA-CTN database and this includes 300 subjects with Metachromatic leukodystrophy, 220 with Aicardi Goutières Syndrome, 167 with Pelizaeus-Merzbacher Disease, 25 with Multiple Sulfatase Deficiency, and 210 with Hypomyelination with Atrophy of Basal Ganglia and Cerebellum.

Conclusion:
The proposed methodology will enable us to leverage existing medical records to accrue subjects and capture clinical information, specifically pre-symptomatic data, on a large cohort of subjects in a shorter duration. This robust data collection will enable— (i) greater understanding of clinical course across leukodystrophies, and (ii) robust, regulatory-standard data applicable as historic controls in clinical trials.

Patient-derived NGN2-induced neurons recapitulate biochemical phenotypes of Multiple Sulfatase Deficiency in vitro.

Margaret M. Cassidy

Graduate Student

Children’s Hospital of Philadelphia/University of Pennsylvania

Margaret M. Cassidy, Vi Pham, Livia Sertori Finoti, Kaitlyn King, Parith Wongkittichote, Xinying Hong, Rebecca C. Ahrens-Nicklas

Multiple Sulfatase Deficiency (MSD) is an ultra-rare lysosomal storage disorder (LSD)caused by mutations in the SUMF1 gene. The SUMF1 gene encodes formylglycine-generated enzyme (FGE), the protein responsible for activating all sulfatases in the cell. The reduction or complete loss of FGE causes detrimental accumulation of sulfated substrates in the lysosome, including glycosaminoglycans (GAGs), sulfolipids, and steroid sulfates. Neurological regression is a primary clinical feature of MSD, which suggests that neural cell types are particularly intolerant to dysfunctional lysosomal flux. Therapeutics targeting MSD are currently limited, and there is an urgent need for a reproducible in vitro model of MSD that recapitulates biochemical phenotypes seen in the patient population. To fulfill this need, we recently generated NGN2-induced neurons (NGN2-iN) from induced pluripotent stemcells (iPSC) derived from a patient with MSD. We use a battery of biochemical and molecular techniques to demonstrate that our model reproduces biochemical phenotypes associated with MSD. Patient NGN2-iN display hallmark pathology of MSD seen in humans, including reduced activity of multiple sulfatases and lysosomal accumulation as compared to isogenic controls. Over time, these neuronsalso demonstrate progressive accumulation of GAG subspecies associated with mucopolysaccharidosis type III. Interestingly, these phenotypes worsen in more mature (Day 11) MSD NGN2-iN, as compared to immature (Day 3) MSD NGN2-iN andiPSCs. This observation suggests that neurons are selectively vulnerable to the loss of FGE, and that dysfunction worsens over neuronal maturation. We anticipate that our model will be utilized to compare the selective vulnerability of individual CNS cell types, further our current understanding of MSD pathophysiology, and ultimately aid the development of therapeutics.

Identification of the cellular mode of action of tazarotene and bexarotene reveals alternative treatment options in Multiple Sulfatase Deficiency

Alisa DeGrave

PhD/PostDoc

Fraunhofer Institute for Tranlational Medicine and Pharmacology – Translational Neuroinflammation and Automated Microscopy

Alisa DeGrave, Lina Schmidt, Karolina Matysiak, Lilly Hutzschenreuter, Lea Keufen, Karthikeyan Radhakrishnan, Jutta Gärtner, Lars Schlotawa

High throughput drug screening identified 3rd generation retinoids tazarotene and bexarotene to restore sulfatase activities, lysosomal positioning and reduction of glycosaminoglycan storage in Multiple Sulfatase Deficiency (MSD, MIM#272200) patient derived fibroblasts. MSD is an ultra-rare inherited lysosomal disorder caused by mutations in the Sulfatase-Modifying-Factor 1 gene (SUMF1) resulting in misfolding, accelerated degradation, and reduced functionality of variants of the Formylglycine-generating enzyme (FGE). Tazarotene is currently investigated for its clinical application as an oral treatment for MSD. The potential of unwanted effects trough tazarotene treatment in children requires the timely identification of alternative druggable targets in MSD cells.

Tazarotene and bexarotene bind to retinoic acid receptors (RARs) and retinoid-X-receptors (RXRs), respectively, in MSD fibroblasts. Binding of retinoic acid derivatives to RARs and RXRs initiates formation of homo- and heterodimers that bind to DNA binding motifs thereby initiating transcription of genes. Ultimately, treatment of cells leads to sulfatase activity increase by decelerated degradation of variant FGE. Blocking of RAR and RXR receptors, respectively, is able to abrogate tazarotene and bexarotene function in MSD fibroblasts. Detailed mechanism of retinoid function downstream of RAR and RXR receptors and upstream of FGE stabilization remains unknown.

In recent work we identified tazarotene and bexarotene to induce endoplasmic reticulum stress mediated by the activation of proteins of the unfolded protein response as one putative mode of action of tazarotene and bexarotene response in MSD cells. Treatment of cells with alternative drug substances and compounds targeting downstream signals of retinoic acid signalling mimicked tazarotene and bexarotene treatment in MSD fibroblasts thereby providing first evidence of effective in-vitro MSD treatment with small molecules as alternatives to retinoids.

Lead Optimization of Small Molecule Sulfatase Reactivators for Multiple Sulfatase Deficiency

Niama Ezzaidi

PhD

University of Southampton

Niama Ezzaidi

Multiple Sulfatase Deficiency (MSD) is a rare lysosomal autosomal recessive disease caused by the loss of function of Formylglycine generating enzyme (FGE) and characterised by almost complete lack of all sulfatase activities. An in-house drug repurposing screening assay of circa 800 FDA approved drugs, carried out in the Schlotawa laboratory in Gottingen, identified a family of structurally and functionally related molecules which are able to upregulate sulfatases activity, reducing glycosaminoglycan storage, and reducing lysosomal size in MSD patient derived cell lines.

In this session we will discuss the development of new small molecule sulfatase reactivators for MSD, following-up and building on these recent preliminary findings. We report the design, synthesise and biologically evaluation of a library of retinoid derivatives targeting the RAR and RXR receptors as well as a fragment-based drug discovery approach which builds on the recent crystallographic fragment screenings (CFS) carried out by the Hartmut laboratory in Bielefeld. We have identified the first small molecule to bind the Formylglycine generating enzyme, a key milestone towards the development of a pharmacological chaperone-based drug for MSD and the discovery of the first FGE stabilizer.

Generation and characterization of Multiple Sulfatase Deficiency iPSC-line and neurogenin-2-induced neurons

Livia Sertori Finoti

Research Associate

Childres’s Hospital of Philadelphia

Livia Sertori Finoti, Vi Pham, Margaret M. Cassidy, Jean Ann Maguire, Alyssa L. Gagne, Elisa A. Waxman, Deborah L. French, Rebecca C. Ahrens-Nicklas

Induced pluripotent stem cells (iPSCs) are reprogrammed from patient somatic cells, thereby harboring natural disease-causing variants within the patient’s genetic background. iPSCs can be differentiated into cellular lineages that are difficult to obtain from patients, such as neurons. Multiple sulfatase deficiency (MSD) is an ultra-rare, inherited lysosomal storage disease caused by mutations in the gene sulfatase modifying factor 1 (SUMF1). MSD patients present neurologic symptoms and neurological disability is universal. However, the specific impact of lysosomal storage on neuronal function and degeneration remains unclear. Here we purposed differentiated MSD and isogenic control iPSCs into neurogenin-2-induced neurons (NGN2-iNs) and to characterized and compare them. To generate an iPSC model of MSD, peripheral blood mononuclear cells were isolated from the whole blood of an MSD patient that was homozygous for the recurrent pathogenic variant SUMF1 c.836C>T, p.A279V. Cells were reprogrammed using non-integrating Sendai viral vectors expressing the human factors OCT3/4, SOX2, KLF4, and cMYC. An isogenic control iPSC line was generated in order to reduce variation in the genetic background between groups. We used CRISPR/Cas9 to correct the pathogenic c.836C>T in the MSD iPSC parental line. We next generated a MSD and isogenic control NGN2-iNs by overexpressing NGN2 transcription factor. NGN2-iNs were collected for analyses at Days 3 (D3 NGN2-iN) and 11 (D11 NGN2-iN) of differentiation to compare phenotypes during neuronal development. iPSCs demonstrated neurite outgrowth by Day 3 and had elaborate processes by Day 11. These findings indicate that MSD iPSCs can successfully differentiate into NPCs and NGN2-iNs to produce neural models. In summary, we successfully differentiated iPSC lines into NGN2-induced neurons (NGN2-iN) to model the neuropathology of MSD. These cell models can be used as tools to further elucidate the mechanisms of MSD pathology and for the development of therapeutics.

Identification of Health Concepts in Aicardi Goutières Syndrome

Francesco Gavazzi

Research Staff Scientist II

Division of Neurology, Children’s Hospital of Philadelphia

Francesco Gavazzi, MD, PhD, Ashley Martin, MPH, Anjana Sevagamoorthy, MBPS, MPH, Ylenia Vaia, MD, Ariel Vincent, BS, Sarah Woidill, MS, Russell D’Aiello III, MS, Sara B. DeMauro, MD, MSCE, Scott A. Lorch, MD, MSCE, Adeline Vanderver, MD, Laura Adang, MD, PhD, MSTR

Background:
Aicardi Goutières Syndrome (AGS) is a common and phenotypically heterogenous leukodystrophy. In general, neurodegenerative disorders of childhood are known to strongly affect the quality-of-life (QoL) and perspective of the whole family and caregiver network. This study explores AGS-related QoL impact in affected children and their caregivers through qualitative interviews and non-concurrently administered validated quality-of-life (QoL) surveys. The goal of this project is to identify disease-specific Health Concepts, to appropriately inform development of disease-specific prospective natural history studies focused on identification of Concept of Interest (COI) and selection of clinical outcome assessments (COAs) able to capture them.

Methods:
The study was designed as mixed-methods. Qualitative interviews (N= 33) and QoL questionnaires (N= 38 to 53) were administered to caregivers of AGS-affected children. The questionnaires included Pediatric Quality of Life-Generic Core (PedsQL-GC) and Family Impact (PedsQL-FI) modules, Caregiver Priorities and Child Health Index of Life with Disabilities (CPCHILD), and Caregiver TBI-CareQoL. The open-ended interviews with guided questions addressed caregiver’s perception of AGS and determinants of QoL. Interviews were conducted until thematic saturation, and qualitative analyses generated salient themes.

Results:
The CPCHILD (n=46), PedsQL-GC (n=53), CareQol (n=38), and PedsQL-FI (n=49) showed impairment of gross and fine motor abilities. activities of daily living (including personal care and independence in the context of domestic and community environments), and overall health and comfort. CPCHILD and PedsQL-FI showed better performance in our cohort, avoiding floor effect (defined as <20%) in each section and demonstrating high compliance to completion.
In the context of qualitative interviews, there was overall alignment between the emerging themes and the most impacted domains of the QoL questionnaires. Families reported stress surrounding symptoms of AGS, poor neurologic development with communication and motor impairment, and the overall impact of the disease on the family. Small improvements in any of these areas was described as important and meaningful.

Discussion:
The study showed the profound effects of AGS on families with global effects on family life. Gross and fine motor abilities, independence in activities of daily living, and overall health and comfort emerged as relevant Health Concepts, that will be used to develop COI and select COAs. This methodology can be replicated in other rare disorders, to design more patient-centric natural history study protocols and inform design of future clinical trials.

Preservation of cognitive skills in Aicardi Goutières Syndrome

Francesco Gavazzi

Research Staff Scientist

Children’s Hospital of Philadelphia

Francesco Gavazzi, MD, PhD, Ylenia Vaia, MD, Sarah Woidill, MS, Brielle Formanowski, MS, Isabella Peixoto de Barcelos, MD, Anjana Sevagamoorthy, MBBS, MPH, Nicholson B. Modesti, BS, Lauren Charlton, BS, Stacy V. Cusack, MSOT, Ariel Vincent, BS, Russell D’Aiello III, MS, Abbas Jawad,PhD, Jessica Galli, MD, PhD, Costanza Varesio, MD, Elisa Fazzi, MD, PhD, Simona Orcesi, MD, PhD, Allan M. Glanzman, PT, Scott Lorch, MD MSCE, Sara B. DeMauro, MD MSCE, Danielle Guez-Barber, MD, PhD, Amy Waldman, MD, Adeline Vanderver, MD, Laura A. Adang, MD, PhD, MSTR

Background:
Aicardi Goutières Syndrome (AGS) is a leukodystrophy characterized by systemic inflammation and severe neurologic impairment. While many children with AGS demonstrate severe motor and expressive language deficits, the magnitude of receptive language impairment is uncharacterized. One challenge is that most cognitive tests require intact motor function. We sought to characterize cognitive function in AGS-affected children using assessment tools with reduced dependence on motor abilities and compare cognitive testing outcomes with overall severity and parental assessment of adaptive behavior.

Methods:
Individuals with a confirmed clinical and molecular diagnosis of AGS were included. Demographic information and AGS Severity Scale Scores were derived from medical records. The Leiter International Performance Scale 3rd edition (Leiter-3) was administered to subjects and parental assessment of function was captured by the Vineland Adaptive Behavior Scale (VABS) 3rd edition. Motor skills were categorized based on mobility levels captured using the AGS Severity Scale.

Results:
Cognitive and adaptive behavior performance was captured in 57 subjects. The mean age at test administration was 8.51 years (SD=5.15). The median score recorded using Leiter-3 was 51 (IQR=60). On the VABS, the Motor Domain was significantly more impacted than the Communication, Daily Living Skills, and Social Domains (p<0.0001). Performance on the AGS scale strongly correlated with VABS-3 and Leiter-3 scores (Spearman’s rank correlation coefficient: r=0.86, p<0.0001; r=0.87, p<0.0001, respectively). There was strong correlation between domains of the VABS-3 with performance on the Leiter-3 (Spearman’s rank correlation coefficient: range=0.83-0.97). Gross motor and fine motor categorization correlated with VABS-3 and Leiter-3 performance (gross motor: Kruskal-Wallis test, both p<0.0001; fine motor: Mann-Whitney U test, both p<0.0001). Within each gross motor and fine motor category of the AGS Severity Scale, a subset of children scored within the normal IQ range.

Discussion:
Parental assessment of function by the VABS-3 strongly correlated with the directly assessed performance measures (physician-reported AGS scale and Leiter-3). Our data underscores the potential value of the VABS-3 and Leiter-3 as tools to assess psychometric function in AGS. With a deeper understanding of our patients’ abilities, we can better guide clinicians and families to provide appropriate support and personalized interventions to empower children with leukodystrophies to maximize their communication and educational potential.

The Search for FGE Stabilizing Molecules: From Fragment Screen Hits to Potential Lead(s)

Hartmut Niemann

Prof. Dr.

Bielefeld University

Julia L. Kowal, Niama Ezzaidi, Jan Wollenhaupt, Karthikeyan Radhakrishnan, Tatjana Pietrowski, Manisha Thangarasah, Willy Keller, Sarfaraz Alam, Manfred S. Weiss, Matthias G. J. Baud, Hartmut H. Niemann

Formylglycine-generating enzyme (FGE) modifies the active site of all known human sulfatases post-translationally, by conversion of a cysteine to formylglycine. Missense mutations in the gene encoding FGE lead to catalytically impaired or unstable protein variants that are prone to degradation. The resulting lack of sulfatase modification causes the rare disease multiple sulfatase deficiency (MSD). With X-ray protein crystallography, we search for small molecules (fragments) that bind to FGE. In a structure-based approach, the found fragments can be further developed into pharmacological chaperones, that potentially stabilize mutated FGE variants and rescue their activity.

Using the F2X-Entry Screen, a subset of the F2X-Universal library, and additional compounds selected from previous experiments, we soaked preformed crystals of human FGE with fragments to identify hits and interesting binding pockets on the enzyme surface. Data collection at BESSY BL 14.2 and multi data set analysis revealed several unique fragment hits in four interaction sites, including the active site of FGE.
Based on the results of the first fragment screen, two compounds, with overlapping binding positions in an allosteric site, were used for merging and synthesis of several follow-ups. Data collection at DESY P13 revealed Fo-Fc density for one of these follow-ups in a data set from a single crystal which suggests stronger interaction with the enzyme. To evaluate whether the discovered hits and follow-up compounds stabilize the protein or not, we obtained first results from differential scanning fluorimetry.

Additionally, we have started a bigger crystallographic screen with the full F2X-Universal library at BESSY. In this ongoing project, we already found further compounds that interact with FGE in three major binding sites, the results of this experiment complement to the ongoing drug design process.

Case Report of Hydrocephalus in Multiple Sulfatase Deficiency Treated with Ventriculoperitoneal Shunt Placement

Isabella Panse

MD/PhD Rotation Student

Division of Human Genetics, The Children’s Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania

Isabella Panse, Emily Yu, Laura Adang, and Rebecca Ahrens-Nicklas

Multiple sulfatase deficiency (MSD) is an ultra-rare neurometabolic disease caused by insufficient formyl glycine-generating enzyme (FGE) function, which is required for sulfatase activation, and thus results in a deficiency of all cellular sulfatases. MSD is clinically characterized by early developmental delays and severe, progressive neurologic regression that can additionally present with ichthyosis, hepatosplenomegaly, and hearing loss. Furthermore, it has been noted that hydrocephalus can be a feature of MSD, which can be visualized by changes on MRI. Here we report the case of a child with MSD and chronic marked communicative hydrocephalus with MRI brain findings of severe enlargement of the ventricular system and extra-axial CSF spaces, as well as increased lumbar CSF opening pressure. To treat the hydrocephalus, a right-sided ventriculoperitoneal (VP) shunt was placed with laparoscopic assistance.

Single vs. dual transgene ex vivo gene therapy for multiple sulfatase deficiency

Vi Pham

PhD Candidate

University of Pennsylvania, Children’s Hospital of Philadelphia

Vi Pham, Lucas Tricoli, Parith Wongkittichote, Xinying Hong, Lars Schlotawa, Stefano Rivella, Rebecca C. Ahrens-Nicklas

Background:
Multiple sulfatase deficiency (MSD) is an ultra-rare, inherited lysosomal storage disorder characterized by the functional deficiency of all sulfatase enzymes. MSD is caused by mutations in the gene SUMF1, encoding a sulfatase modifying factor that activates 17 downstream sulfatases. MSD patients present with combined features of single-sulfatase disorders such as the mucopolysaccharidoses and metachromatic leukodystrophy (MLD). Currently, there are no disease-modifying therapies for MSD. We aimed to develop ex vivo gene therapy with hematopoietic stem cell transplant for MSD, as this approach has proven successful for related disorders.

Methods:
We designed lentiviral vectors encoding either 1) SUMF1 or 2) a downstream sulfatase ARSA that is greatly implicated in the disease. ARSA ex vivo gene therapy is approved by the EMA for MLD and has the potential to be repurposed for patients with MSD.

Results:
In vitro, MSD patient fibroblasts transduced with the lentiviral vectors demonstrated improved biochemical phenotypes including rescue of sulfatase activities and glycosaminoglycan (GAG) accumulation. In vivo, both lentiviral vectors proved efficacious in rescuing biochemical and behavioral phenotypes in MSD mice that underwent hematopoietic stem cell transplant with ex vivo gene therapy. To further augment outcomes, we co-transduced MSD patient fibroblasts with both SUMF1 and ARSA vectors and found that co-expressing these transgenes greatly enhanced biochemical rescue in vitro. We will present the results of our ongoing work evaluating the efficacy of this innovative dual-transgene approach in MSD mice after ex vivo gene therapy.

Efficacy of a scAAV9/SUMF1 viral vector for the treatment of Multiple Sulfatase Deficiency

Pierre-Alexandre Piec

Research Assistant

Jackson Laboratory

Maximiliano Presa, Rachel M Bailey, Somdatta Ray, Lauren Bailey, Saurabh Tata, Tara Murphy, Harold Combs, Steven J Gray, Cathleen Lutz

Multiple Sulfatase Deficiency (MSD) is a rare autosomal recessive disorder characterized by deficient enzymatic activity of all known sulfatase. MSD patients frequently carried two loss of function mutations in the SUMF1 gene, encoding a sulfatase modifying factor which activates 17 different sulfatases. MSD patients shown common features of mucopolysaccharidosis, and metachromatic leukodystrophy, including neurologic impairments, developmental delay, and visceromegaly. There are currently no approved therapies for MSD patients. Gene therapy may provide a meaningful and long-term therapeutic benefit for MSD patients by delivering a functional copy of the SUMF1 gene to patient cells. We developed a self-complementary vector encoding a codon-optimized human SUMF1 gene (scAAV9/SUMF1), the unaltered design of which could be appropriate for human use. Using a severe Sumf1 knock-out (KO) mouse model of MSD, we tested the benefits of treatment with our vector in mice shortly after birth and later in the disease course by administering virus into the cerebrospinal fluid (CSF) alone or in combination with an intravenous injection. CSF administration of scAAV9/SUMF1 was sufficient to rescue early lethality, normalize behavior and improve newly identified visual and cardiac dysfunctions in KO mice, even when given at an advanced disease stage. We found dose-dependent and long-term restoration of sulfatase activity in tissues, which is accompanied by decreased accumulation of glycosaminoglycans, lysosomal defects and neuroinflammation in the brain. Even at the highest dose tested, scAAV9/SUMF1 was well-tolerated without toxicity from high transgenic SUMF1 expression in KO and wild-type mice or in rats that were treated as part of a GLP-toxicology study. Overall, our preclinical results attest to the safety of scAAV9/SUMF1 delivery and predict a benefit of this treatment to MSD patients.

Tracking Clinical and Biochemical Changes Post Bone Marrow Transplant in Multiple Sulfatase Deficiency – A 3-Year Follow-Up

Nishitha Pillai

Assistant Professor

University of Minnesota

Nishitha R Pillai, Ning Liu, Rebecca Ahrens Nicklas, Laura Adang, Julie B Eisengart, Grace Bronken, Ashish Gupta, Troy Lund, Chester B Whitley, Sarah H Elsea and Paul J Orchard

Background:
Multiple sulfatase deficiency (MSD) is an exceptionally rare neurodegenerative disorder due to the absence or deficiency of 17 known cellular sulfatases. Two siblings with attenuated MSD underwent hematopoietic cell transplantation (HCT), evaluating the possibility of lysosomal enzymatic cross-correction from the donor cells.

Methods:
A retrospective data analysis was done on the two siblings who underwent HCT. The data collected included biomarkers including urine glycosaminoglycans, urine sulfatides, untargeted metabolomics in plasma and enzyme assays. The data from neuropsychology evaluation was also collected up to 3 years after HCT.

Results:
Biochemical correction of all the previously deficient enzyme activity in the leukocytes was evident within 3 months post HCT. Urine heparan and dermatan sulfate along with urine sulfatides were normal in the pre and post HCT samples due to the milder phenotype. While single, unique biomarkers were not identified for MSD, multiple consistent metabolic perturbations across patients were noted. Specifically, glycerophospholipids constituted the predominant category of perturbed sub-pathways in both siblings. Arylsulfates and fatty acids and derivatives, exhibited substantial perturbations, representing the second or third largest proportion of perturbed metabolites across the siblings. Additionally, energy-related sub-pathways such as bile acids, cholesterols, sterols, nucleotides, and various amino acids were disrupted. Notably, the relative levels of disruption across these sub-pathways between the siblings are consistent. These results indicate that MSD induces a broad state of metabolic dysregulation. The impact of HCT were monitored by comparing metabolomic profiles pre- and post-HCT. An almost instant neutralization effect introduced by HCT was observed. The correction of abnormalities was further quantitatively analyzed in every sequential post-HCT samples, while the initial post-HCT samples collected at 3 months showed a significant correction, with more than 70% of abnormal concentration levels falling within the normal range. This correction persisted in subsequent post-HCT samples, demonstrating the effectiveness of HCT. These finding align with the biochemical enzyme corrections observed in the patients.

Conclusion:
These data suggest biochemical benefit post-transplant, although the clinical implication of these corrections are unknown, particularly in the context of the siblings’ attenuated phenotype. Long-term follow-up is necessary to evaluate the therapeutic benefit of HCT in MSD.

Clinical Trial Readiness for Rare Diseases: An innovative approach

Anjana Sevagamoorthy

Clinical Research Associate II

Children’s Hospital of Philadelphia

Anjana Sevagamoorthy1 MBBS, MPH, Francesco Gavazzi1 M.D., Ph.D., Laura Adang1 M.D., Ph.D., MSTR, Adeline Vanderver1,2. M.D. 1. Division of Neurology, Children’s Hospital of Philadelphia, PA, USA 2. Department of Neurology, University of Pennsylvania, PA, USA

Background:
Leukodystrophies are rare genetic disorders affecting myelination. The enactment of the Food and Drug Administration (FDA) 1983 Orphan Drug Development Act, in addition to advancements in clinical characterization and diagnostic tools, has led to increasing interest in rare disease drug development. The needs of rare disease drug development and approval is rapidly evolving in the context of the 2022 four-part FDA guidance “Patient-Focused Drug Development” (PFDD), necessitating its adoption in the context of clinical trial readiness. In this study, we propose a multi-component standardized patient-centric approach to the identification of clinical outcome assessments (COAs) for use in the context of clinical trials.

Methods:
We developed a multi-component approach based on FDA’s conceptual model. First, patient caregiver-identified Health Concepts (HC) and derived priorities (Concepts of Interest [COI]) were identified through standardized impact-of-disease questionnaires. This was followed by consensus building on COIs for use in the context of clinical trials (Context of Use [COU]), among a panel of disease experts, through modified eDelphi approach, a multi-round iterative virtual process of anonymized voting on pre-specified themes by an expert panel. Disease experts are identified based on contribution to the field and/or clinical expertise. Identified experts are solicited to vote on statements describing “importance” and “changeability within a pre-defined time frame”, in the COU, of the COIs. COIs are qualified upon reaching 70% agreement on both its importance and changeability. Upon failure to qualify, the statements are clarified and deployed for subsequent voting. Upon eDelphi completion, the caregiver- and expert-identified COIs within COU are aligned. Subsequently, selection of Clinical Outcome Assessments (COAs) capturing COI is performed through similar consensus building among outcome experts, followed by determination of COA fit-for-purpose for application in the COU. A study protocol delineating COA performance is then developed for prospective application in a cohort of affected individuals for validation of the COA and feasibility assessment.

Results:
The results of the consensus building approach to identification of COIs within COU for Aicardi-Goutieres Syndrome is summarized here. A total of 4-rounds of voting was performed among 10 experts each in rounds 1 & 3, and 12 in rounds 2 & 4. The following COIs achieved overall consensus: (i) Motor- postural function, floor mobility, coordination, endurance in physical activity, use of electronic devices, fine motor performance speed and pointing; (ii) Adaptive behavior- use of adaptive/alternative communication, complexity of verbal language, endurance and quality in completion of cognitive tasks, independence in completion of activities of daily living, imitation of activities, preferential looking and communication through behavior; (iii) Neurologic function- visual function and neurologic signs; (iv) skin lesions.

Conclusion:
The proposed approach is the first step towards clinical trial readiness adopting patient-centricity in rare diseases. The outcome is a rigorous COA protocol for prospective application and a panel of validated COAs applicable in clinical trials. This is critical to (i) identify clinical endpoints and determine meaningful changes in the endpoints as required by the FDA guidelines on PFDD, and (ii) inform clinical trial design.

Development of a rigorous approach to performing Real-World Evidence- based natural history studies in leukodystrophies

Anjana Sevagamoorthy

Clinical Research Associate II

Children’s Hospital of Philadelphia

Anjana Sevagamoorthy MBBS, MPH, Francesco Gavazzi M.D., Ph.D., Omar Sherbini MPH, Ariel Vincent B.S., Russel D’Aiello M.S., Nicholson Modesti, B.S., Sylvia Mutua B.A., Emily Yu B.A., Sarah Woidill M.S., Johanna L. Schmidt MPH, MGC, LCGC, Amy Pizzino M.S., LCGC, Justine Shults Ph.D., Adeline Vanderver M.D., Laura Adang M.D., Ph.D., MSTR

Background:
Leukodystrophies are rare genetic disorders affecting myelination. Emerging therapeutics primarily target the pre-symptomatic population, but there is a knowledge gap on the impact of disease across the lifespan. To address this, we propose a standardized approach to performing retrospective natural history studies using existing medical records.

Methods:
As part of Global Leukodystrophy Initiative Clinical Trials Network (GLIA-CTN), we developed Standard Operating Procedures (SOPs) that delineated all study processes related to staff training, source documentation, and data sharing. Additionally, the SOP detailed the standardized approach to data extraction including diagnosis, clinical presentation, and medical events, such as age at gastrostomy tube placement. The key variables for extraction were selected through face validity, and common electronic case report forms (eCRF) across leukodystrophies were created to collect analyzable data. To enhance the depth of the data, clinical notes are extracted into “original” and “imputed” encounters, with imputed encounter referring to a historic event (e.g., loss of ambulation 3 months prior). Retrospective Functional Assessments are assigned by child neurologists, using a blinded dual-rater approach and any discrepancies are adjudicated by a third rater. Upon completion of extraction, data source verification is performed. Data missingness was evaluated using statistics.

Results:
To date, the records from 922 subjects have undergone this process across the GLIA-CTN database and this includes 300 subjects with Metachromatic leukodystrophy, 220 with Aicardi Goutières Syndrome, 167 with Pelizaeus-Merzbacher Disease, 25 with Multiple Sulfatase Deficiency, and 210 with Hypomyelination with Atrophy of Basal Ganglia and Cerebellum.

Conclusion:
The proposed methodology will enable us to leverage existing medical records to accrue subjects and capture clinical information, specifically pre-symptomatic data, on a large cohort of subjects in a shorter duration. This robust data collection will enable— (i) greater understanding of clinical course across leukodystrophies, and (ii) robust, regulatory-standard data applicable as historic controls in clinical trials.

miRNAs in Multiple Sulfatase Deficiency as a Signature of Neurodegeneration

Eden Teferedegn

Postdoc Fellow

Children’s Hospital of Philadelphia

Eden Teferedegn,Livia Sertori Finoti , Vi Pham, Elizabeth Bohj & Rebecca C Ahrens-nicklas

Introduction:
MicroRNAs (miRNAs) are small RNA molecules that play a crucial role in translation regulation, influencing major cellular processes such as cell fate and development. miRNAs have been implicated in MSD, an ultra-rare neurodegenerative disorder characterized by progressive neurodegeneration leading to the loss of sensory and motor skills.

Methods:
In this study, we present miRNAs that are dysregulated in neurons derived from patient-derived induced pluripotent stem cell and isogenic controls. Next-generation sequencing (NGS) miRNA sequencing was conducted to profile their differential expression, followed by downstream analysis. Initial quality control and annotation were performed on the Illumina BaseSpace platform.

Result and Discussion:
Downstream analysis using Ingenuity Pathway Analysis (IPA) revealed significant upregulation of hsa-miR-1269b, hsa-miR-4516, and hsa-miR-4761-3p in the MSD neurons (FDR p-value < 0.0001). EN2 and HNRNPU emerged as the two most significantly enriched predictive targets for these miRNAs. EN2 plays a role in controlling the development of the central nervous system and dopaminergic neurogenesis, with dysregulation linked to cerebellar hypoplasia and other neurodegenerative disorders. Similarly, HNRNPU, involved in the formation of ribonucleoprotein complexes, is associated with HNRNPU-related neurodevelopmental disorder (HNRNPU-NDD) characterized by seizures, early onset epilepsy, hypotonia, autistic features, and intellectual disability. Notably, our analysis identified the KEAP1:NFE2L2 pathway as significantly enriched in our miRNA dataset. This pathway plays a central role in protecting cells against various homeostatic responses, including adaptation to oxidative, inflammatory and metabolic stresses. The NFE2L2 transcriptome has been implicated in protection against numerous chronic diseases, including metabolic and neurodegenerative disorders. In conclusion, our data demonstrates how miRNAs can serve as a signature for neurodegenerative diseases, shedding light on potential molecular mechanisms and therapeutic targets.

The impact of Multiple Sulfatase Deficiency on children & families: caregiver perspectives

Emily Yu

MSD Clinical Research Coordinator

Children’s Hospital of Philadelphia

Emily Yu BA, Francesco Gavazzi MD, PhD, Sarah Woidill BS, MS, Vivian Breitling MD, Lars Schlotawa MD, Rebecca Ahrens-Nicklas MD, PhD, Laura Adang MD, PhD, MSTR

Multiple Sulfatase Deficiency (MSD) is an ultra-rare lysosomal storage disorder associated with pathogenic variants in the SUMF1 gene, which is required for sulfatase activation. Despite the known severity of disease, the impact of MSD on affected children, caregivers, and their family is uncharacterized.

Nineteen families affected by MSD participated in our assessments. The Vineland Adaptive Behavior Scales (VABS-3) was administered by a child neurologist to caregivers in the context of an affected individual’s in-person or remote research encounter. Health-related quality of life (HRQOL) surveys were collected electronically, including the Caregiver Priorities and Child Health Index of Life with Disabilities (CPCHILD), Pediatric Quality of Life Inventory Generic Core Scales (PedsQL GC), PedsQL Family Impact Module (FIM), and the Traumatic Brain Injury Caregiver Quality of Life (CareQOL) questionnaires.

All clinical outcome measures demonstrated a range of performance. Most caregivers assessed their children as severely affected (VABS-3 <40 Standard Score across all Domains and Adaptive Behavior Composite Score: n=10/19; 52.63%), with the lowest median scores in the Communication and Motor Skills domains. The Communication domain’s mean performance was significantly lower than the Socialization and Daily Living Skills domains. The Motor Skills domain was significantly lower than the Socialization domain only. The CPCHILD (n=12) identified items within the Comfort and Emotions and Overall Health domains as top priorities when preserving affected individuals’ quality of life. The Comfort and Emotions domain’s mean performance was significantly higher than all other domains. The CPCHILD overall HRQOL remained high (median 80%, IQR 50%). The PedsQL GC (n=14) demonstrated higher mean performance in the Emotional than the School/Work domain only. The CareQOL (n=15) identified caregivers’ significantly higher frequency of a sense of loss for the affected individual and feelings of entrapment. The PedsQL FIM (n=13) demonstrated that there was no significant difference between family functioning and parent HRQOL performance.

In this study, we demonstrated that MSD has profound, global effects on both affected individuals and their caregivers as measured by the VABS-3 and HRQOL surveys. This data will be integral to identifying Health Concepts and related Concept of Interests to inform future clinical trial design.

Let’s Connect

Connect with MSD families and get valuable information from medical researchers and doctors.