PK and ADA Assays: ICH M10, FDA & EMA Guidance Explained

    Navigating Regulatory Expectations for PK and ADA Assays in Biologics (ICH M10 and Beyond)

    PK and ADA Assays in Biologics ICH M10

    In biologics development, two critical analytical pillars are pharmacokinetic (PK) assays – which measure drug concentrations – and anti-drug antibody (ADA) assays – which detect immune responses against the biologic (immunogenicity). Regulatory agencies have stringent expectations for these assays because they generate data essential for evaluating safety, efficacy, and comparability of biologic therapies. Navigating these expectations can be challenging, especially with evolving guidelines like ICH M10 and region-specific FDA/EMA guidances. This article provides a deep dive into current bioanalytical validation standards and immunogenicity testing regulations, summarizing key points from ICH M10, FDA, and EMA, and highlighting best practices to ensure PK assay compliance and ADA assayregulatory readiness at every development stage.

    ICH M10 Guidance for PK Assay Validation

    The ICH M10 guidance (finalized in 2022) is a harmonized international standard for bioanalytical method validation, particularly relevant to PK assays used in support of drug development. ICH M10 outlines the requirements for validating analytical methods that quantify drugs (and their metabolites) in biological samples across nonclinical and clinical studies. It covers both chromatographic methods (e.g. LC-MS/MS) and ligand-binding assays (LBAs such as ELISA or MSD) – the latter being commonly used for large-molecule biologics. The goal is to ensure assays are sensitive, accurate, and reliable for their intended purpose, thereby yielding data that can support regulatory submissions globally.

    Key validation parameters covered by ICH M10 include accuracy, precision, selectivity, sensitivity (LLOQ), calibration curve range, carry-over, dilution integrity, and stability. Acceptance criteria largely mirror past standards (typically ±15% for accuracy and precision, and 20% at the LLOQ). The guideline also addresses certain considerations specific to LBAs (e.g. avoiding high-dose “hook” effects).

    It’s important to note that ICH M10 explicitly excludes immunogenicity assays for anti-drug antibodies from its scope. In other words, ADA assays are governed by separate immunogenicity guidance (discussed below) rather than the general bioanalytical validation framework in M10. Nonetheless, ICH M10 has become the cornerstone for PK assay validation globally, replacing or aligning previously disparate regional guidelines. By following ICH M10, sponsors ensure their PK concentration data (e.g. from pharmacokinetic or toxicokinetic studies) meet the harmonized expectations of regulators across ICH regions and beyond.

    PK and ADA Assays in Biologics ICH M10

    FDA and EMA Guidance on ADA Assay (Immunogenicity) Testing

    While ICH M10 governs PK assay validation, ADA assays (which assess immunogenicity) fall under specific guidances issued by agencies such as the FDA and EMA. Both agencies stress a tiered approach to immunogenicity testing and the need to validate ADA assays to ensure they can detect anti-drug antibodies reliably.

    FDA Guidance: The FDA’s guidance titled “Immunogenicity Testing of Therapeutic Protein Products: Developing and Validating Assays for Anti-Drug Antibody Detection” (2019) provides detailed recommendations for ADA assay development and validation. The FDA endorses a multi-tiered testing approach: first, a highly sensitive screening assayis used to detect any anti-drug antibodies; any screen-positive samples are then subjected to a confirmatory assay to verify the antibodies are truly directed against the therapeutic. Confirmed positive samples may also be evaluated for their antibody levels (titer) or neutralizing antibodies that block the drug’s activity.

    For validation, the FDA expects parameters similar to PK assay validation, but tailored to ADA assays. Key elements include establishing the assay cut-point, sensitivity, specificity, selectivity, precision, drug tolerance, and robustness. Determining a statistically sound cut-point – the threshold distinguishing positive from negative samples – is a fundamental step. Guidelines recommend using robust statistical methods on an adequate number of samples to derive the cut-point, and confirming it with target population samples if initial estimates used surrogate matrices. For drug tolerance, FDA recommends testing the assay’s ability to detect ADA across a range of drug concentrations to identify the highest drug level at which ADA can still be detected. Techniques like acid dissociation can be employed to improve drug tolerance if significant interference is observed.

    EMA Guidance: The European Medicines Agency’s Guideline on immunogenicity assessment of biotechnology-derived therapeutic proteins (rev. 2017) similarly mandates a thorough immunogenicity evaluation for biologics. EMA’s guideline recommends the same basic three-tier ADA testing approach (screening, confirmatory, and characterization assays) and requires validated assays for reliable immunogenicity data. However, it is less detailed on validation specifics compared to the FDA’s guidance, instead emphasizing risk-based principles and noting that assay validation is an ongoing process throughout development. By the time of marketing authorization in Europe, assays used for pivotal clinical trial samples should be fully validated for their intended use. Early-phase immunogenicity assessments might be performed with methods still in development (qualified assays), but pivotal studies must employ fully validated ADA assays to satisfy regulators.

    Despite some differences in detail, both regulators ultimately demand sensitive, specific ADA assays with appropriate controls. In practice, many sponsors follow the FDA’s detailed recommendations as a de facto standard to satisfy both FDA and EMA expectations. The bottom line is that a scientifically sound ADA assay validation – with a well-justified cut-point, demonstrated sensitivity, adequate drug tolerance, and thorough documentation – will meet the immunogenicity testing standards of both agencies and withstand regulatory scrutiny.

    PK and ADA Assays in Biologics ICH M10

    Best Practices for Compliance Across Development Stages

    Ensuring compliance with PK and ADA assay expectations is not a one-time task – it spans the entire drug development timeline. Here are some best practices to meet regulatory standards from early development through licensure:

    • Fit-for-Purpose Validation: In early-stage development (discovery, preclinical, and Phase 1 trials), employ a fit-for-purpose approach. This means that the extent of validation can be scaled to the development stage and risk. For example, FDA suggests that a partial validation focusing on key parameters like precision, cut-point, and drug tolerance (with less emphasis on long-term stability or robustness) may suffice for first-in-human or Phase 1 studies. Similarly, for PK assays in early toxicology or exploratory clinical trials, you might perform limited validation (e.g. accuracy and precision in a few matrices) to ensure the method works, then expand to full validation later. Always document what was validated and any limitations.

    • Full Validation by Pivotal Phase: Before pivotal Phase 3 trials and certainly before filing a Biologics License Application (BLA) or Marketing Authorisation Application (MAA), ensure all assays are fully validated to meet regulatory requirements. ICH M10 and EMA guidelines concur that assays used for pivotal clinical data must be fully validated for their intended purpose. For PK assays, this means completing the full battery of validation tests (accuracy, inter/intra-assay precision, range, stability under sample handling conditions, etc.). For ADA assays, it means you have an established cut-point using samples from the target population, known sensitivity (lowest ADA level detectable), specificity confirmed (e.g. by drug competition in the confirmatory assay), and so on, all documented in a validation report.
    • Reassess Cut-Points: If the initial ADA assay cut-point was established using healthy donor samples, re-evaluate it with samples from the target patient population. Confirming or adjusting the cut-point in-study ensures the threshold is appropriate for that population’s background and reduces false results. Additionally, include quality controls in each assay run (e.g. a low positive control near the cut-point) to monitor any drift in assay sensitivity over time.
    • Optimize Drug Tolerance: Determine the assay’s drug tolerance – the highest drug concentration at which ADA can still be detected. Use methods like acid dissociation or alternative assay formats to increase drug tolerance if necessary, so that ADA detection is not hindered by circulating drug. Document the drug tolerance in validation reports, as it indicates up to what drug level the assay remains effective.

    Frequently Asked Questions (FAQs)

    Q: What is ICH M10 and why is it important for PK assays?
    A: ICH M10 is a harmonized guidance for bioanalytical method validation issued by the International Council for Harmonisation. It standardizes how to validate PK assays (and other bioanalytical assays) across all ICH regions, ensuring that data from drug concentration measurements meet consistent quality criteria. ICH M10 is crucial because it aligns the expectations of FDA, EMA, and other regulators, making it easier to prepare globally acceptable validation packages. By following ICH M10, you demonstrate that your pharmacokinetic assays for biologics are accurate, precise, specific, and robust enough to support regulatory decisions.

    Q: What is a cut-point in an ADA assay, and how is it determined?
    A: The cut-point is the assay threshold that differentiates a positive ADA result from a negative result. In other words, if a sample’s signal is above the cut-point, it’s considered ADA-positive. It involves testing many ADA-negative samples to determine the normal background signal, then statistically defining a threshold that separates positives from negatives (often set to yield ~5% false positives). Guidances recommend using a sufficient number of samples (e.g. ~50) and robust statistics to set the cut-point. The initial cut-point, established in validation, may be adjusted with in-study data if the patient population shows different background reactivity. Because the cut-point directly impacts how sensitive your ADA detection is, it’s a critical part of assay validation and must be justified to regulators.

    Q: What does “drug tolerance” mean in an ADA assay?
    A: “Drug tolerance” refers to the ability of an ADA assay to detect antibodies in the presence of the drug. High levels of circulating drug can interfere with ADA detection by binding the antibodies and preventing them from binding the assay reagents. An assay with good drug tolerance can still detect a positive control ADA even when a substantial concentration of the drug is present in the sample. Improving drug tolerance might involve modifying the assay protocol (for example, adding an acid dissociation step to break antibody-drug complexes before detection). It’s important to determine and report drug tolerance during validation, since it indicates at what drug level the assay may start missing ADA signals.

    Conclusion

    Navigating the regulatory landscape for PK and ADA assays in biologics requires understanding both global standards like ICH M10 and regional guidances from FDA and EMA. By staying abreast of these guidelines and implementing best practices in assay validation, sponsors can ensure that their bioanalytical data stand up to scrutiny. The effort invested in robust PK and immunogenicity assays pays off in de-risking clinical development and smoothing the path to regulatory approval. As the field continues to evolve (with ongoing discussions, Q&A documents, and refinements to guidelines), it’s wise to continually update your strategies and train your teams on the latest expectations.

    If you’re interested in a deeper dive into PK and ADA assay requirements and practical tips for implementation, consider the upcoming training course on “PK and ADA Assays for Biologics”. This course offers expert insights into assay development, validation, and compliance, helping you stay ahead in the biologics arena.

    Where to go from here?

    Basket