Consulting Services

Expert guidance for IND applications in gene therapy and vaccine development tailored to your needs.

two white tablets
two white tablets
IND Application Support

We assist with IND submissions and regulatory compliance for gene therapy projects.

Gene Therapy Consulting

Specialized consulting for gene therapy development, ensuring successful regulatory navigation and project execution.

Vaccine Development Aid

Comprehensive support for vaccine development, from concept to regulatory submission and approval.

  • Development of Analytical Methods
    • Purpose: Design and optimize analytical methods to assess critical quality attributes (CQAs) such as identity, purity, potency, and stability.

    • Activities:

      • Development of assays for product release, characterization, and process development support.

      • Use of orthogonal techniques (e.g., qPCR, ddPCR, analytical ultracentrifugation [AUC], capillary electrophoresis, cryo-TEM) to ensure comprehensive characterization.

      • Customization for specific modalities, such as viral vectors (e.g., AAV, lentivirus), plasmid DNA, or mRNA-based vaccines.

      • Incorporation of bioassays, including cell-based potency assays, to confirm biological activity.

    • Regulatory Alignment: Methods are developed to meet FDA and EMA requirements, including ICH guidelines for validation (e.g., ICH Q2 for analytical method validation).

  • Characterization of Product and Components
    • Purpose: Provide detailed data on the physicochemical and biological properties of the drug substance and drug product.

    • Activities:

      • Characterization of viral vectors (e.g., AAV capsid content, full/empty capsid ratios, genome titer, infectious titer).

      • Analysis of mRNA therapeutics for sequence identity, integrity, and impurities (e.g., truncated mRNA, aggregates).

      • Assessment of delivery systems, such as lipid nanoparticles (LNPs) for mRNA vaccines, including lipid content, encapsulation efficiency, and release profiles.

      • Testing of raw materials, starting materials (e.g., master/working cell banks, viral banks), and excipients for purity, identity, and safety.

      • Use of advanced techniques like next-generation sequencing (NGS), bio-layer interferometry (BLI), or mass spectrometry for structural and functional analysis.

  • Release Testing
    • Purpose: Ensure the final product meets predefined specifications for identity, purity, potency, and safety before clinical use.

    • Activities:

      • Development and validation of release assays, including:

        • Identity Tests: Confirm product composition (e.g., PCR for vector identity, gel electrophoresis for viral proteins).

        • Purity Tests: Quantify product-related impurities (e.g., empty capsids, host cell proteins) and process-related impurities (e.g., residual DNA, benzonase).

        • Potency Tests: Measure biological activity (e.g., cell-based assays for gene expression or immune response).

        • Safety Tests: Screen for adventitious agents (e.g., mycoplasma, viral contaminants, bioburden) in bulk harvest and purified drug substance.

      • Establishment of product specifications based on robust laboratory data to support IND submission.

      • Validation of release methods to ensure reproducibility and compliance with cGMP requirements.

  • Stability Testing
    • Purpose: Assess the stability of the drug substance and drug product under various conditions to establish shelf life and storage requirements.

    • Activities:

      • Design and execution of real-time and accelerated stability studies to evaluate product degradation (e.g., vector integrity, mRNA stability).

      • Analysis of stability-indicating parameters, such as genome titer, infectious titer, and aggregation.

      • Assessment of delivery systems (e.g., LNPs) for stability under storage and transport conditions.

      • Development of stability protocols and reports for inclusion in the IND application.

      • Guidance on container closure systems (e.g., extractables and leachables testing) to ensure compatibility.

  • Impurity Analysis
    • Purpose: Identify and quantify process- and product-related impurities to ensure safety and quality.

    • Activities:

      • Detection of residual host cell DNA, proteins, and reagents (e.g., benzonase, Triton X-100) using qPCR, ELISA, or other sensitive methods.

      • Quantification of empty or partial capsids in viral vectors (e.g., via AUC, TEM, or A260/280 ratio).

      • Analysis of mRNA-specific impurities, such as truncated or aggregated mRNA, using capillary gel electrophoresis (CGE) or HPLC.

      • Establishment of impurity thresholds compliant with regulatory guidelines.

  • Potency Assay Development
    • Purpose: Demonstrate the biological activity of the gene therapy or vaccine product to support clinical efficacy.

    • Activities:

      • Development of cell-based potency assays to measure gene expression, protein production, or immune response (e.g., neutralizing antibody assays for vaccines).

      • Validation of potency assays to ensure sensitivity, specificity, and linearity as per FDA guidance (e.g., “Potency Tests for Cellular and Gene Therapy Products,” 2011).

      • Alignment with regulatory expectations for phase-appropriate potency testing (e.g., qualification for early-phase trials, full validation for later phases).

  • Validation of Analytical Methods
    • Purpose: Ensure analytical methods are robust, reproducible, and suitable for regulatory submission.

    • Activities:

      • Validation of methods for accuracy, precision, specificity, linearity, and range as per ICH Q2 guidelines.

      • Preparation of validation protocols and reports for inclusion in the IND application.

      • Troubleshooting and optimization of methods to address variability or sensitivity issues.

      • Ensuring methods are phase-appropriate (e.g., qualified for early-phase INDs, fully validated for later stages).

  • Regulatory Compliance and Documentation
    • Purpose: Ensure the analytical section meets FDA, EMA, and other global regulatory requirements for IND submission.

    • Activities:

      • Preparation of the Chemistry, Manufacturing, and Controls (CMC) section of the IND, including analytical data and method descriptions.

      • Alignment with FDA guidance, such as “Chemistry, Manufacturing, and Control (CMC) Information for Human Gene Therapy INDs” (2020) and “Potency Assurance for Cellular and Gene Therapy Products” (2023).

      • Support for pre-IND meetings with regulatory agencies to discuss analytical strategies and obtain feedback.

      • Authoring and reviewing analytical sections of IND applications, including protocols, reports, and justifications for specifications.

      • Ensuring compliance with cGMP, ICH Q7 (for raw materials), and other relevant guidelines.

  • Risk Assessment and Quality Control
    • Purpose: Identify and mitigate risks associated with analytical methods and product quality.

    • Activities:

      • Conducting risk assessments for critical analytical steps to ensure robust quality control.

      • Development of quality management systems (QMS) to support inspection readiness.

      • Monitoring of critical quality attributes (CQAs) throughout the manufacturing process.

      • Implementation of orthogonal techniques to enhance confidence in analytical results.

  • Support for Novel Technologies
    • Purpose: Address analytical challenges specific to emerging modalities, such as mRNA vaccines or gene editing therapies.

    • Activities:

      • Development of analytical methods for mRNA therapeutics, including characterization of lipid nanoparticles and mRNA integrity.

      • Support for gene editing therapies (e.g., CRISPR-based) with assays for on-target/off-target effects and genome editing efficiency.

      • Customization of analytical approaches for novel vectors (e.g., non-viral vectors, microbial vectors) or vaccine adjuvants.

  • Technology Transfer and Scale-Up Support
    • Purpose: Ensure analytical methods are transferable and scalable for clinical and commercial manufacturing.

    • Activities:

      • Support for technology transfer of analytical methods to contract manufacturing organizations (CMOs) or in-house facilities.

      • Validation of methods at scale to ensure consistency during process scale-up or scale-out.

      • Guidance on analytical method robustness for large-scale production of viral vectors or mRNA vaccines.

  • Bioanalytical Support for Preclinical and Clinical Studies

    • Purpose: Provide analytical data to support preclinical and clinical development, bridging the analytical section with nonclinical/clinical data.

    • Activities:

      • Development of bioassays to support pharmacokinetic (PK) and pharmacodynamic (PD) studies.

      • Analysis of biodistribution, shedding, and persistence for gene therapies (e.g., viral vector shedding studies).

      • Support for long-term follow-up (LTFU) studies, including analytical testing for safety and efficacy endpoints.

      • Integration of real-world evidence (RWE) to inform analytical strategies for clinical trials.