You have 3 free guides left 😟
Unlock your guides
You have 3 free guides left 😟
Unlock your guides

11.4 Regulatory considerations in biomarker development

3 min readjuly 25, 2024

Biomarker development is a complex process overseen by regulatory agencies like the FDA and EMA. These bodies ensure the safety and effectiveness of biomarkers through specialized centers and programs, while international organizations work to harmonize requirements across regions.

Biomarkers serve various purposes, from predicting clinical outcomes to guiding treatment selection. Validation is crucial, involving analytical, clinical, and utility assessments. Regulatory approval hinges on meeting specific requirements, navigating challenges like varying performance standards and ethical considerations.

Regulatory Framework for Biomarker Development

Regulatory landscape for biomarker development

Top images from around the web for Regulatory landscape for biomarker development
Top images from around the web for Regulatory landscape for biomarker development
  • (FDA) oversees biomarker development in US through specialized centers
    • Center for Drug Evaluation and Research (CDER) regulates prescription and over-the-counter drugs
    • Center for Biologics Evaluation and Research (CBER) focuses on biological products (vaccines, blood products)
    • Center for Devices and Radiological Health (CDRH) regulates medical devices and radiation-emitting products
  • (EMA) regulates biomarkers in European Union provides scientific advice and protocol assistance
  • (ICH) harmonizes regulatory requirements across regions (US, EU, Japan)
  • (CLIA) ensures quality standards for laboratory testing
  • evaluates biomarkers for use in drug development
  • Regulatory pathways determine approval process based on risk and novelty
    • for devices substantially equivalent to existing products
    • (PMA) for high-risk devices requiring clinical trials
    • for novel low to moderate risk devices

Types of biomarker qualifications

  • Prognostic biomarkers predict clinical outcome regardless of treatment (tumor stage, gene expression profiles)
  • Predictive biomarkers identify patients likely to respond to specific treatment guide therapy selection (HER2 for breast cancer)
  • measure biological response to treatment assess drug mechanism of action (blood glucose levels for diabetes medications)
  • detect or confirm presence of disease or condition (PSA for prostate cancer)
  • assess status of disease or medical condition over time (HbA1c for diabetes management)
  • indicate potential for toxicity or adverse events (liver enzymes for drug-induced liver injury)

Biomarker Validation and Regulatory Approval

Requirements for biomarker validation

  • ensures biomarker measurement is reliable and reproducible
    1. Accuracy measures closeness of test results to true value
    2. Precision evaluates reproducibility of results
    3. Sensitivity determines lowest detectable concentration
    4. Specificity assesses ability to distinguish target from similar molecules
    5. Reproducibility examines consistency across different labs or instruments
    6. Robustness evaluates performance under varying conditions
  • assesses biomarker performance in target population
    • Sensitivity and specificity in target population determine true positive and true negative rates
    • Positive and negative predictive values indicate probability of disease given test result
    • Receiver operating characteristic (ROC) curve analysis evaluates overall test performance
  • demonstrates biomarker's impact on patient care
    • Improvement in patient outcomes (survival rates, quality of life)
    • Cost-effectiveness analysis compares biomarker use to standard of care
    • Impact on clinical decision-making evaluates changes in treatment strategies
  • tailors validation process to intended use of biomarker
  • (COU) documentation defines specific application and interpretation of biomarker
  • vary based on biomarker type and intended use
    • provide highest level of evidence
    • offer real-world data on biomarker performance
    • combine results from multiple studies to increase statistical power

Challenges in biomarker regulatory approval

  • Analytical performance requirements vary based on biomarker type and intended use
  • Clinical performance requirements depend on and intended use
  • Risk classification determines level of regulatory scrutiny (Class I, II, III)
  • Intended use and indications for use define specific applications and patient populations
  • communicate proper use and limitations of biomarker test
  • monitors long-term safety and effectiveness
  • requires coordinated regulatory strategy
  • for multiple platforms ensure consistent results across different testing methods
  • Harmonization of global regulatory requirements facilitates international development and approval
  • and enable data sharing and meta-analyses
  • Ethical considerations impact study design and implementation
    • ensures participants understand risks and benefits
    • Privacy and data protection safeguard sensitive genetic information
  • Reimbursement and health economic considerations affect market adoption and access
© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.


© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.

© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.
Glossary
Glossary