ARSA Mouse

Arylsulfatase A Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Relevance of ARSA Mouse Models

ARSA Mouse refers to murine models genetically engineered to lack functional Arylsulfatase A (ARSA), an enzyme critical for sulfatide degradation. These models are used to study metachromatic leukodystrophy (MLD), a lysosomal storage disorder characterized by sulfatide accumulation, demyelination, and neurodegeneration. ARSA deficiency leads to progressive neurological symptoms, mirroring human MLD pathology .

Gene Structure and Expression

  • Gene length: 3.8 kb with 8 exons .

  • Transcript variants:

    • Primary mRNA: 3.2 kb (5' UTR: 638 nt, 3' UTR: ~700 nt) .

    • Alternative polyadenylation: Rare 4.0 kb transcript in liver .

  • Protein: 506 amino acids (53.2 kDa), glycosylated, lysosomal localization .

Recombinant ARSA Production

ParameterValueSource
Host systemSf9 insect cells (Baculovirus)
Tag6–9 amino acid His-tag (C-terminus)
Molecular weight53.2 kDa
Purity>95% (SDS-PAGE)

Disease Pathology in ARSA KO Mice

  • Key features:

    • Sulfatide accumulation in CNS and peripheral organs .

    • Astrogliosis (GFAP+ cells) and microgliosis (Iba1+ cells) .

    • Motor deficits (e.g., rotarod performance) emerging at 12–18 months .

Gene Therapy Approaches

VectorAdministration RouteKey OutcomesSource
AAVPHP.eB-hARSA-HAIntravenous3–5× ARSA activity in CNS; reduced neuroinflammation
HSC15/ARSAIntravenousHigher ARSA activity vs AAV9; serum ARSA detectable
AAV.GMU01-ARSAIntracerebroventricular84% cross-correction in brain; NHP CNS transduction

Efficacy and Biodistribution

MetricAAVPHP.eB-hARSA-HAHSC15/ARSAAAV.GMU01-ARSA
CNS ARSA activity3–5× WT levels4–6× WT4× WT
Peripheral organ uptakeMinimalLowLow
Species testedMouseMouse/NHPsMouse/NHPs

Mechanistic Insights

  • Lysosomal targeting: Co-localization of ARSA with Lamp1 confirmed in treated mice .

  • Cross-correction: Widespread ARSA expression in neurons, astrocytes, and microglia .

Therapeutic Milestones

  1. Complete CNS correction: AAVPHP.eB-hARSA-HA reversed sulfatide storage and neuroinflammation in 9-month-old KO mice .

  2. Dose-dependent efficacy: AAV.GMU01-ARSA showed sustained ARSA expression for 13 months in pre-neuropathic mice .

  3. NHP translation: HSC15/ARSA and AAV.GMU01-ARSA achieved therapeutic ARSA levels in cynomolgus macaques .

Challenges and Future Directions

  • Vector optimization: Improving BBB penetration and reducing peripheral organ transduction .

  • Biomarker validation: Correlating ARSA activity with motor recovery and sulfatide clearance .

Product Specs

Introduction
Arylsulfatase A (ARSA) is an enzyme that breaks down cerebrosidesulfate into cerebroside and sulfate. This process is hindered by phosphate, which forms a covalent bond with the enzyme's active site. Genetic defects affecting the ARSA gene lead to metachromatic leucodystrophy (MLD), a progressive neurological disorder characterized by demyelination, a range of neurological issues, and ultimately, death.
Description
Recombinant ARSA Mouse, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain consisting of 498 amino acids (specifically, amino acids 18-506). It has a molecular mass of 53.2kDa. However, on SDS-PAGE, the molecular size appears to be between 50-70kDa. This ARSA protein is expressed with a 6 amino acid His tag at its C-Terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
ARSA protein solution at a concentration of 0.5mg/ml. It is supplied in Phosphate Buffered Saline with a pH of 7.4 and contains 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For longer storage, it should be frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. It is crucial to avoid repeated freezing and thawing of the product.
Purity
The purity of the product is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Arylsulfatase A, ASA, Cerebroside-sulfatase.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
ADPSPPNILL IFADDLGYGD LGSYGHPSST TPNLDQLAEG GLRFTDFYVP VSLCTPSRAA LLTGRLPVRS GMYPGVLGPS SQGGLPLEEV TLAEVLAARG YLTGMAGKWH LGVGPEGAFL PPHQGFHRFL GIPYSHDQGP CQNLTCFPPD IPCKGGCDQG LVPIPLLANL TVEAQPPWLP GLEARYVSFS RDLMADAQRQ GRPFFLYYAS HHTHYPQFSG QSFTKRSGRG PFGDSLMELD GAVGALMTTV GDLGLLEETL VIFTADNGPE LMRMSNGGCS GLLRCGKGTT FEGGVREPAL VYWPGHITPG VTHELASSLD LLPTLAALTG APLPNVTLDG VDISPLLLGT GKSPRKSVFF YPPYPDEIHG VFAVRNGKYK AHFFTQGSAH SDTTSDPACH AANRLTAHEP PLLYDLSQDP GENYNVLESI EGVSPEALQA LKHIQLLKAQ YDAAMTFGPS QIAKGEDPAL QICCQPSCTP HPVCCHCPGS QSHHHHHH.

Q&A

What is the ARSA mouse model and how does it mimic human MLD?

The ARSA knockout (KO) mouse model, also referred to as Arsa-/- mice, replicates key aspects of metachromatic leukodystrophy. This model was created through disruption of the Arsa gene, specifically in Exon 4 near the HindIII site, using a loxP-hUBp-em7-Neo-polyA-loxP cassette in C57BL6 embryonic stem cells . The resulting model is listed at Jackson Labs as B6N.129P2(CBA)-Arsatm1Gie/J and is genetically validated through a Loss of Native Allele (LOA) assay .

The Arsa KO model exhibits progressive accumulation of sulfatides in both the central nervous system (CNS) and visceral organs, mirroring the human pathology. These mice develop gliosis, microglial activation, histopathological abnormalities including degeneration and vacuolation in brain parenchyma, and an auditory phenotype that correlates with the loss of neurons in the ventral cochlear nucleus . This pathology closely resembles the original Gieselmann model, which is another established MLD mouse model.

While the mouse model doesn't fully recapitulate the severe demyelination seen in human patients, it does provide a valuable platform for studying sulfatide metabolism dysregulation and testing potential therapeutic interventions for MLD.

What phenotypic and biochemical characteristics define ARSA knockout mice?

ARSA knockout mice display several key phenotypic and biochemical characteristics that make them valuable for MLD research:

  • Sulfatide accumulation: The most defining characteristic is the progressive accumulation of sulfatides in the CNS and visceral organs due to the absence of ARSA enzyme activity. This accumulation is detectable through methods such as mass spectrometry and histochemical staining .

  • Neuroinflammation: Evidence of modest gliosis and microglial activation throughout the brain, which can be quantified through immunohistochemical staining for markers such as GFAP and Iba1 .

  • Auditory dysfunction: A hallmark functional deficit in these models is hearing impairment, which mirrors a common symptom in human MLD patients. This can be assessed through auditory brainstem response testing .

  • Age-dependent progression: The pathology worsens with age, with sulfatide accumulation becoming more pronounced and widespread over time, similar to the human disease progression .

  • Absence of ARSA activity: Biochemically, these mice show undetectable levels of ARSA enzyme activity in all tissues, which can be confirmed through enzymatic assays .

  • Normal lifespan with subtle motor deficits: Unlike human patients, these mice generally have a normal lifespan but do develop subtle motor coordination deficits that can be measured through behavioral testing .

These characteristics provide multiple endpoints for assessing therapeutic interventions, from biochemical markers like sulfatide levels to functional outcomes like hearing ability, making the model particularly useful for preclinical testing of potential MLD treatments.

How are ARSA mouse models utilized in different stages of therapeutic development?

ARSA mouse models serve critical roles across the therapeutic development pipeline:

  • Target validation: These models confirm that ARSA deficiency leads to sulfatide accumulation and subsequent pathology, validating ARSA replacement as a therapeutic approach. Studies have demonstrated that restoring ARSA activity through gene therapy leads to reduction in toxic sulfatides and phenotypic reversal .

  • Proof-of-concept studies: Initial testing of novel therapeutic approaches, such as AAV-mediated gene therapy, is conducted in these models to establish feasibility. For example, studies have shown that delivery of AAV-GMU01-ARSA into the CNS of Arsa knockout mice results in significant therapeutic benefit .

  • Dose-finding studies: ARSA mice are used in dose-response studies to determine optimal therapeutic dosing. Research has demonstrated dose-dependent increases in sulfatase activity and concomitant decreases in sulfatide deposits following treatment with AAV vectors at different doses .

  • Administration route optimization: These models help determine the most effective delivery routes for therapies. Studies have compared different administration approaches, with intracerebroventricular (ICV) injection showing reproducibility and efficient AAV vector delivery into the CSF of mice .

  • Long-term efficacy evaluation: ARSA mice enable assessment of therapeutic durability, with studies establishing transgene expression in neonate and adult mice out to 12 and 52 weeks, respectively .

  • Translational biomarker development: These models facilitate identification and validation of biomarkers that could translate to clinical use. Research has supported "the applicability of CSF and plasma biomarkers for monitoring therapeutic efficacy" .

By enabling these various applications, ARSA mouse models form the foundation for advancing potential therapies toward clinical testing for MLD patients.

How does AAV-mediated gene therapy restore ARSA function in mouse models?

AAV-mediated gene therapy for ARSA restoration operates through several sophisticated mechanisms:

These mechanisms collectively restore ARSA enzyme activity throughout the CNS, leading to normalization of sulfatide metabolism, reduction in accumulated sulfatides, and reversal of functional deficits in ARSA knockout mice.

What are the comparative advantages of different AAV capsids for CNS delivery in ARSA mouse models?

Different AAV capsids demonstrate varying efficacy in CNS delivery for ARSA gene therapy, with significant implications for treatment outcomes:

  • Novel engineered capsids vs. traditional serotypes: Novel capsids like AAV.GMU01 demonstrate superior biodistribution and transgene expression in the CNS compared to traditional serotypes. When compared with intravenously administered AAV9/ARSA, significant increases in brain ARSA activity, transcript levels, and vector genomes were observed with HSC15/ARSA .

  • Transduction efficiency and cell tropism: In wild-type mice treated with AAV.GMU01-eGFP at a dose of 1.6e11 VG per mouse, single-cell RNA sequencing revealed transduction of approximately 12% of cells in the forebrain and 3% in the hindbrain, demonstrating region-specific differences in transduction efficiency . Different capsids show varying tropism for neuronal versus glial cell populations, which impacts therapeutic outcomes.

  • Blood-brain barrier penetration: Some capsids, like HSC15, demonstrate superior blood-nerve, blood-spinal, and blood-brain barrier crossing capabilities. Research has shown that "intravenous administration of HSC15/ARSA restored the endogenous murine biodistribution of the corresponding enzyme," allowing for systemic administration rather than direct CNS delivery .

  • Immune profile considerations: Novel capsids may offer advantages in terms of pre-existing immunity in the human population, an important consideration for clinical translation.

  • Tissue distribution beyond CNS: Different capsids show varying propensities for transducing peripheral tissues. Some studies noted a "dose-dependent increase in vector biodistribution in liver, spleen, and cervical lymph node," which may be advantageous or detrimental depending on the therapeutic goals .

The selection of an appropriate capsid depends on the intended route of administration, target cell populations, and desired biodistribution profile. Comprehensive comparative studies in both mice and non-human primates have been essential to optimize these parameters for potential clinical translation.

How can researchers optimize therapeutic efficacy assessment in ARSA mouse models?

Optimizing therapeutic efficacy assessment in ARSA mouse models requires a comprehensive, multi-dimensional approach:

By implementing these approaches, researchers can more accurately characterize therapeutic efficacy, establish dose-response relationships, and generate translatable findings that support clinical development of ARSA gene therapies for MLD.

What are the optimal administration routes for AAV-ARSA delivery in mouse models?

The selection of administration routes for AAV-ARSA delivery in mouse models depends on research objectives and the stage of disease being targeted. Several routes have been investigated, each with distinct advantages and technical considerations:

  • Intracerebroventricular (ICV) injection:

    • Advantages: Provides reproducibility and efficient AAV vector delivery directly into the CSF of mice, bypassing the blood-brain barrier.

    • Technical approach: Bilateral ICV injections of AAV.GMU01-ARSA at doses ranging from 1e10 VG to 3.3e11 VG/gm brain weight.

    • Research applications: Selected for efficacy studies in MLD mice due to its reliability in delivering vector to the CNS .

    • Limitations: Requires specialized surgical skills and is more invasive than systemic delivery.

  • Intravenous (IV) administration:

    • Advantages: Less invasive, potential for widespread distribution including both CNS and peripheral organs.

    • Research findings: "Intravenous administration of HSC15/ARSA restored the endogenous murine biodistribution of the corresponding enzyme" .

    • Best applications: Particularly effective with capsids that can efficiently cross the blood-brain barrier, such as HSC15 .

    • Limitations: May result in higher vector distribution to peripheral organs with only a fraction reaching the CNS.

  • Intracisternal magna (ICM) injection:

    • Technical approach: Animals are placed in Trendelenburg position throughout dosing and for up to 15 minutes following dosing completion. A needle is inserted into the cisterna magna, and vector is infused at controlled rates.

    • Applications: Used primarily in larger animal models like non-human primates but can be adapted for mice.

    • Advantages: Provides direct access to CSF circulation with less invasiveness than ICV .

    • Considerations: Requires careful positioning and imaging guidance for accurate delivery.

  • Combined approaches:

    • Rationale: Multiple administration routes can be combined to achieve optimal biodistribution.

    • Examples: Sequential ICV and IV administration to target both CNS and peripheral organs.

For mouse studies focused primarily on CNS effects, the ICV route has been particularly well-validated, demonstrating dose-dependent vector biodistribution and robust increases in sulfatase activity in the brain, with levels surpassing those of wild-type mice at all tested doses .

How should researchers design dose-response studies for ARSA gene therapy?

Designing robust dose-response studies for ARSA gene therapy requires careful consideration of multiple experimental parameters:

  • Dose range selection:

    • Coverage span: Employ a wide dose range that extends from sub-therapeutic to potentially supra-therapeutic levels.

    • Dose intervals: Use logarithmic spacing between doses (e.g., 3-10 fold increments) to efficiently characterize the dose-response curve.

    • Example design: Studies have utilized dose ranges from 1e10 VG to 3.3e11 VG/gm brain weight across 3-4 dose levels .

  • Age-appropriate intervention timing:

    • Pre-symptomatic model: Dosing at P0 (postnatal day 0) to model early intervention before disease manifestation.

    • Early-symptomatic model: Dosing at 6 months of age to represent treatment after disease initiation but before extensive pathology.

    • Late-stage model: Dosing in older mice with established pathology to assess reversal potential .

  • Control group selection:

    • Positive controls: Age-matched wild-type mice providing normal ARSA activity benchmarks.

    • Negative controls: Untreated age-matched Arsa KO mice representing disease progression without intervention.

    • Vehicle controls: Arsa KO mice receiving formulation buffer to account for procedural effects .

  • Comprehensive endpoint measurements:

    • Vector biodistribution: Quantification of vector genomes across brain regions and peripheral tissues.

    • ARSA expression: Measurement of ARSA mRNA and protein levels in various tissues.

    • Enzyme activity: Quantification of ARSA-mediated sulfatase activity.

    • Biochemical efficacy: Assessment of sulfatide reduction in central and peripheral tissues.

    • Functional outcomes: Evaluation of phenotypic improvements such as auditory function .

  • Temporal assessment strategy:

    • Short-term evaluations: 2-3 months post-treatment to assess initial efficacy.

    • Long-term follow-up: 6-12 months post-treatment to determine durability of effect.

    • Example timepoints: Neonatally treated animals assessed 6 months post-dose; early-neuronopathic mice assessed 3 months post-dose .

  • Statistical considerations:

    • Group sizes: Minimum of 8-10 animals per dose group for adequate statistical power.

    • Analysis methods: Non-linear regression to establish EC50 values and dose-response curves.

    • Correlation analyses: Relationships between biomarker changes and functional improvements.

This systematic approach enables identification of optimal therapeutic doses, characterization of dose-dependent effects, and establishment of biomarker-outcome relationships that can guide subsequent translational studies.

What techniques are most effective for quantifying sulfatide reduction in ARSA mouse models?

Accurate quantification of sulfatide reduction is crucial for assessing therapeutic efficacy in ARSA mouse models. The most effective techniques include:

  • Liquid Chromatography-Mass Spectrometry (LC-MS/MS):

    • Methodology: Tissue homogenates are subjected to lipid extraction, followed by chromatographic separation and mass spectrometry detection.

    • Advantages: Provides precise quantification of specific sulfatide species and their molecular variants, enabling differential analysis of various sulfatide species that may respond differently to treatment.

    • Applications: Allows for sensitive detection of changes in sulfatide levels across different brain regions and peripheral tissues.

    • Sensitivity: Can detect subtle changes in sulfatide levels even with partial therapeutic response .

  • Immunohistochemistry with anti-sulfatide antibodies:

    • Methodology: Tissue sections are stained with specific antibodies that recognize sulfatides, followed by visualization with secondary detection systems.

    • Advantages: Provides spatial information about sulfatide distribution and clearance patterns across different anatomical regions and cell types.

    • Applications: Useful for regional and cellular analysis of sulfatide accumulation and clearance.

    • Quantification: Can be semi-quantitative when coupled with digital image analysis and standardized controls .

  • Histochemical staining techniques:

    • Methodology: Alcian blue or other histochemical stains that interact with sulfatides can visualize their presence in tissue sections.

    • Applications: Particularly useful for qualitative assessment of sulfatide deposits in different tissues.

    • Advantages: Relatively simple technique that can be implemented in most histology laboratories .

  • Normalized quantification approaches:

    • Reference standards: Inclusion of age-matched wild-type tissue (representing normal sulfatide levels) and untreated Arsa KO tissue (representing maximal accumulation).

    • Data expression: Results presented as percentage of wild-type levels or percentage reduction compared to untreated controls.

    • Statistical analysis: Application of appropriate statistical tests to determine significance of observed reductions .

These complementary approaches provide comprehensive assessment of therapeutic efficacy. Research has demonstrated that following ARSA gene therapy, mice show dose-dependent reduction in sulfatide levels, with neonatally treated animals showing significant decreases in brain sulfatide levels six months post-treatment, while early-neuronopathic mice showed substantial reduction three months post-treatment .

How can biomarker changes be correlated with functional improvements in ARSA mouse models?

Establishing correlations between biomarker changes and functional improvements in ARSA mouse models requires sophisticated experimental design and analytical approaches:

  • Integrated assessment timeline:

    • Sequential measurements: Perform biomarker assessments prior to functional testing to establish temporal relationships.

    • Longitudinal design: Track both biomarkers and functional outcomes in the same animals over time to establish within-subject correlations.

    • Multiple timepoints: Evaluate at early (1-2 months), intermediate (3-6 months), and late (6-12 months) post-treatment phases to capture dynamic relationships .

  • Hierarchical biomarker framework:

    • Primary biomarkers: ARSA enzyme activity in brain tissue, CSF and plasma.

    • Secondary biomarkers: Sulfatide levels in CNS and peripheral tissues.

    • Tertiary biomarkers: Neuroinflammatory markers, myelin integrity measures.

    • Functional outcomes: Auditory brainstem responses, motor coordination tests .

  • Quantitative correlation analysis:

    • Regression analysis: Establish mathematical relationships between biomarker levels and functional parameters.

    • Threshold determination: Identify minimum levels of ARSA activity required for functional improvement.

    • Multivariate modeling: Account for multiple predictors and their interactions.

    • Research has defined "levels and correlation between changes in biomarkers and ARSA activity required to achieve functional motor benefit" .

  • Regional correlation approaches:

    • Brain region-specific analysis: Correlate regional biomarker improvements with functions mediated by those regions.

    • Cell type-specific effects: Analyze relationships between cell-specific biomarker changes and related functional outcomes.

    • Pathway analysis: Correlate molecular pathway normalization with specific functional domains.

  • Translational biomarker development:

    • Accessible biomarkers: Focus on CSF and plasma biomarkers that can be readily measured in clinical settings.

    • Predictive relationships: Establish how early biomarker changes predict later functional improvements.

    • Research supports "the applicability of CSF and plasma biomarkers for monitoring therapeutic efficacy" .

By implementing these approaches, researchers can establish causal relationships between biochemical normalization and functional improvement, identify predictive biomarkers for therapeutic response, and develop clinically relevant monitoring strategies for translation to human studies.

How do findings in ARSA mouse models translate to non-human primate studies for MLD gene therapy?

The translation from ARSA mouse models to non-human primate (NHP) studies involves several important considerations and comparative analyses:

  • Comparative AAV capsid performance:

    • Cross-species evaluation: Capsids showing efficacy in mice undergo comparative testing in NHPs to assess translation.

    • Specific comparisons: Studies have evaluated AAV.GMU01 versus AAV.rh10 in cynomolgus monkeys to directly compare their performance in larger animals.

    • Translational findings: Results demonstrated that successful capsids in mouse models (like AAV.GMU01) also showed promising biodistribution and expression patterns in NHPs, but with species-specific differences in regional distribution .

  • Administration route optimization:

    • Technical adaptations: Routes successful in mice require technical adaptation for NHPs.

    • Comparative assessment: Different administration techniques are evaluated in NHPs, including intrathecal catheter insertion, bilateral intracerebroventricular injection (ICV), and direct cisterna magna injection (ICM).

    • Translational outcomes: Research has shown that direct ICM injection results in widespread ARSA expression in the CNS of cynomolgus monkeys, providing a clinically feasible approach .

  • Dose scaling considerations:

    • Allometric scaling: NHP studies test multiple dose levels to determine appropriate scaling factors from mice to larger animals.

    • Dose-response assessment: Four doses ranging from 7.5e11 to 2.5e13 VG/animal were tested in cynomolgus NHPs.

    • Comparative findings: Dose-dependent increases in vector biodistribution, ARSA mRNA, and protein levels were observed at the 7.5e12 and 2.5e13 VG/animal doses in NHPs, establishing translational dose-response relationships .

  • Biodistribution patterns:

    • Regional assessment: Vector distribution is compared across species for regional similarities and differences.

    • Cross-species findings: "Uniform, dose-dependent vector biodistribution and ARSA expression observed in DRGs and spinal cord along the rostral-caudal axis in NHPs" demonstrated some conservation of distribution patterns .

  • Safety and tolerability evaluation:

    • Comprehensive assessment: NHP studies include neurological and behavioral tests pre-dose, 7-days post-dose, and at necropsy.

    • Outcome measures: Tissue, CSF, and plasma are collected for comprehensive safety analysis.

    • Translational implications: Research has demonstrated that AAV.GMU01-ARSA treatment is well tolerated in NHPs, supporting potential clinical safety .

These translational studies are crucial for establishing that therapeutic approaches developed in mouse models have potential clinical applicability, providing the foundation for human clinical trials while accounting for species-specific differences in CNS anatomy, immune responses, and vector tropism.

What methodological approaches enable translation of ARSA mouse findings to clinical applications?

Translating findings from ARSA mouse models to clinical applications requires specialized methodological approaches:

  • Clinically relevant administration routes:

    • Adaptation of delivery methods: While ICV administration is common in mice, studies in NHPs explore clinically feasible routes like ICM injection.

    • Technical refinement: Development of "minimally invasive and clinically feasible administration route" that can be implemented in human patients .

    • Outcome evaluation: Assessment of whether clinically relevant routes achieve sufficient vector distribution and ARSA expression for therapeutic benefit.

  • Cross-species biomarker validation:

    • Biomarker conservation: Identification of biomarkers that show similar relationships to disease and treatment across species.

    • Non-invasive monitoring: Validation of CSF and plasma biomarkers that can be readily assessed in human patients.

    • Predictive relationships: Establishment of how biomarker changes predict functional outcomes across species .

  • Therapeutic threshold determination:

    • Minimum effective dose: Determination of ARSA activity levels required for functional improvement in mice.

    • Cross-species extrapolation: Assessment of whether similar relative increases in ARSA activity are needed in larger animals.

    • Studies have defined "levels and correlation between changes in biomarkers and ARSA activity required to achieve functional motor benefit" .

  • Long-term efficacy prediction:

    • Duration modeling: Development of mathematical models to predict long-term efficacy from shorter-term NHP studies.

    • Surrogate endpoints: Identification of early markers that reliably predict sustained therapeutic benefit.

    • Mouse research established "durability of transgene expression in neonate and adult mice out to 12 and 52 weeks," providing benchmarks for translation .

  • Immune response management:

    • Cross-species immune profiling: Characterization of immune responses to vector and transgene across species.

    • Immunomodulation strategies: Development of approaches to manage potential immune reactions in clinical settings.

    • NHP studies include screening for "AAV neutralizing antibodies, and seronegative animals were selected for the studies" .

  • Regulatory-compatible study design:

    • GLP-compliant protocols: Implementation of Good Laboratory Practice standards in pivotal NHP studies.

    • FDA-aligned endpoints: Selection of outcome measures acceptable to regulatory agencies.

    • Manufacturing considerations: Vector production using methods suitable for clinical application .

These methodological approaches facilitate the translation of promising findings from mouse models through NHP studies to eventual clinical applications, addressing species-specific considerations while maintaining focus on the fundamental therapeutic mechanisms.

How can combined biomarker and functional assessments optimize therapeutic development using ARSA mouse models?

Integrating biomarker and functional assessments creates a powerful approach to optimize therapeutic development using ARSA mouse models:

  • Integrated assessment framework:

    • Triangulation strategy: Combining biochemical, histological, and functional measures provides comprehensive evaluation of therapeutic efficacy.

    • Translational confidence: Research has emphasized "the importance of triangulating multiple end points to increase the translation into higher species" .

    • Complementary insights: Different assessment modalities can reveal aspects of therapeutic response not captured by single measures.

  • Biomarker-to-function correlation analysis:

    • Threshold determination: Identifying minimal levels of ARSA activity needed for functional improvement.

    • Predictive relationships: Establishing how early biomarker changes predict later functional outcomes.

    • Studies have defined "levels and correlation between changes in biomarkers and ARSA activity required to achieve functional motor benefit" .

  • Staged assessment hierarchy:

    • Primary endpoints: Direct measures of target engagement (ARSA activity, vector biodistribution).

    • Secondary endpoints: Biochemical efficacy (sulfatide reduction).

    • Tertiary endpoints: Functional improvements (auditory function, motor performance).

    • This staged approach provides clear evidence of therapeutic mechanism and functional relevance .

  • Temporal relationship mapping:

    • Dynamic profiling: Tracking how biomarker changes precede, coincide with, or follow functional improvements.

    • Intervention timing optimization: Determining optimal treatment windows based on combined biomarker-function relationships.

    • Durability assessment: Evaluation of whether biomarker stability predicts sustained functional benefit .

  • Clinically translatable assessments:

    • Non-invasive biomarkers: Focus on biomarkers accessible in CSF or plasma for potential clinical monitoring.

    • Functional measures with human equivalents: Selection of mouse functional tests that have clear human correlates.

    • Research supports "the applicability of CSF and plasma biomarkers for monitoring therapeutic efficacy" .

  • Personalized therapy considerations:

    • Response variation analysis: Examination of factors influencing variable responses across individual animals.

    • Predictive biomarkers: Identification of baseline characteristics that predict optimal response.

    • Adaptive design support: Development of biomarker-guided approaches for potential personalized dosing in clinical settings.

This comprehensive assessment approach enables more effective identification of promising therapeutic candidates, determination of optimal dosing regimens, and establishment of clinically relevant monitoring strategies to increase the likelihood of successful translation to MLD patients.

Product Science Overview

Introduction

Arylsulfatase A (ARSA) is an enzyme that belongs to the sulfatase family. It is encoded by the ARSA gene and plays a crucial role in the lysosomal degradation of cerebroside-3-sulfate, a sphingolipid sulfate ester and a major constituent of the myelin sheath . The recombinant form of this enzyme, derived from mouse sources, is used in various research and therapeutic applications.

Structure and Function

The deduced amino acid sequence of mouse ARSA consists of a signal peptide (residues 1-17) and a mature chain (residues 18-506) . The enzyme’s activity is measured by its ability to hydrolyze the substrate 4-Nitrocatechol Sulfate (PNCS), with a specific activity of over 100 pmol/min/µg . The recombinant mouse ARSA protein is typically produced in a mouse myeloma cell line (NS0) and includes a C-terminal 10-His tag for purification purposes .

Biological Importance

ARSA is essential for the degradation of cerebroside-3-sulfate in lysosomes. Deficiency in ARSA activity leads to a lysosomal storage disease known as metachromatic leukodystrophy (MLD), which affects the central and peripheral nervous systems and results in severe and progressive neurological symptoms . The recombinant form of ARSA is used to study the enzyme’s function and to develop potential therapies for MLD.

Applications

Recombinant mouse ARSA is used in various research applications, including enzyme activity assays, structural studies, and therapeutic research. It is supplied as a 0.2 μm filtered solution in Tris and NaCl and is stable for up to six months when stored at -20 to -70 °C .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.