regulatory updates

The Future of GCP is Here
What you need to know about ICH E6(R3)

Updated January 2025

As of January 6, 2025, the ICH E6(R3) Good Clinical Practice (GCP) guideline has been finalized and adopted by the ICH Assembly at Step 4 of the ICH process.

This significant update reflects the culmination of extensive collaboration among global regulatory authorities and industry stakeholders to modernize and enhance the standards for clinical trial conduct.

At QACV Consulting, we believe staying ahead of regulatory changes is essential to achieving compliance, quality, and patient safety. This edition of our newsletter brings you key insights and practical takeaways on ICH E6(R3).

Key Updates in R3:
ICH E6(R3) is structured into two major sections.

1. Principles Section:

  • Introduces 12 GCP principles (revised from the original 13).
  • Emphasizes participant safety, data integrity, and proportionality.
  • Promotes risk-based approaches and quality by design (QbD).

2. Annexes:

  • Annex 1: GCP expectations for traditional interventional trials.
  • Annex 2 (in development): Will address non-traditional designs such as decentralized, pragmatic, or real-world evidence-based trials.

What This Means for Sponsors, CROs, and Investigators:

  • Early Integration of Quality by Design (QbD): Focus on identifying critical-to-quality factors early in trial design.
  • Risk-Based Monitoring (RBM): Continued emphasis on centralized monitoring, data analytics, and remote oversight.
  • Fit-for-Purpose Documentation: Encourage lean documentation practices aligned with the complexity and risk of the trial.
  • Technology and Innovation: Supports electronic systems, digital health technologies, and decentralized trial models while ensuring validation, security, and data integrity.

What is ICH E6(R3)?

ICH E6(R3) is the latest revision of the GCP guideline originally adopted in 1996. This third revision reflects
over two decades of advancements in clinical trial design, technology, and regulatory expectations. It aims to:

  • Modernize the GCP principles
  • Embrace a risk-based, flexible, and proportional approach
  • Support innovations such as decentralized trials and real-world data.

Implications for Quality and Compliance

  • Organizations must reassess SOPs, training, and vendor oversight strategies.
  • Quality teams should prepare for regulatory scrutiny on risk assessment practices and critical data/process documentation.
  • Auditors will need to adjust checklists and risk models to evaluate compliance with the new principles and approaches.

For detailed information, you can access
the final ICH E6(R3) Good Clinical Practice (GCP) Final Guideline

How QACV Consulting
Can Help

As global experts in quality and compliance, QACV Consulting offers:

  • Gap Assesments against ICH E6(R3) principles and Annex 1.
  • Customized training programs for QA teams, clinical operations, and vendor partners.
  • Audit tools and checklists aligned with the new guidelines.
  • Strategic consulting to align QbD and RBM with your organizational practices

The transition to ICH E6(R3) represents a pivotal shift in clinical research—one that emphasizes agility, innovation, and a renewed focus on what matters most: PARTICIPANT SAFETY and RELIABLE DATA. Now is the time to get ready. QACV Consulting is here to help you lead the change.

The finalized guideline is scheduled to become effective in July 2025, providing a transitional period for sponsors, investigators, and regulatory bodies to align their practices with the updated standards. This period allows for necessary adjustments in trial design, conduct, oversight, and documentation to ensure compliance with the new requirements.

Additionally, the FDA has provided resources and guidance to facilitate the implementation of ICH E6(R3) within
the United States. These resources offer insights into the FDA’s expectations and recommendations
for adopting the updated GCP standards.

updated
april 2025

On April 10, 2025, the U.S. Food and Drug Administration (FDA) announced a significant shift in drug development practices by initiating a phase-out of traditional animal testing requirements for monoclonal antibodies and other drugs. This move aims to enhance drug safety, reduce research and development costs, and ultimately lower drug prices.

The FDA plans to replace Animal Testing with New Approach Methodologies (NAMs), which include:​

AI-Based Computational Models:

Utilizing Artificial Intelligence to predict a drug’s behavior and potential side effects.​

Human-Based Lab Models:

Employing lab-grown human organoids and organ-on-a-chip systems that mimic human organs to test drug safety.​

To facilitate this transition, the FDA will launch a pilot program allowing select developers of monoclonal antibodies to use primarily non-animal-based testing strategies. Companies that submit strong safety data from these alternative methods may receive streamlined reviews, incentivizing investment in modern testing platforms.

This initiative aligns with the FDA Modernization Act 2.0, signed into law in December 2022, which removed the requirement for animal testing in drug development, making it optional. The Act enables the use of alternative testing methods such as computer models and cell-based assessments.

While this marks a transformative change in drug evaluation, the FDA acknowledges that further development and validation of these alternative methods are necessary to ensure they can effectively replace animal testing across all areas of drug development.​

Updated
January 2025

The FDA’s Guidance for Industry on Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment continues to serve as a cornerstone for ensuring the safety of marketed pharmaceutical products. The guidance outlines critical recommendations for identifying, evaluating, and mitigating risks associated with drug and biological product use in the post-marketing setting.

1

Adverse Event Reporting

Emphasizes timely, accurate, and complete adverse event reporting. Sponsors are encouraged to implement robust systems to ensure compliance with 21 CFR 314.80 and 600.80 regulations.

2

Risk Assesment

Recommends systematic approaches for signal detection and risk evaluation, including active surveillance and periodic aggregate data analysis.

3

Pharmacoepidemiologic Studies

Encourages the use of real-world data, registries, and observational studies to understand adverse event trends and rare outcomes, supporting proactive risk management decisions.

4

Risk Minimization Action Plans (RiskMAPs)

Describes strategies to communicate and mitigate identified risks—ranging from labeling changes to more complex interventions such as restricted distribution programs.

5

Periodic Safety Update Reporting

Aligns safety reporting practices with international standards (e.g., ICH E2E), enabling greater harmonization for global sponsors.

QACV Consulting Logo

PERSPECTIVE

As pharmacovigilance practices evolve in complexity, QACV Consulting supports clients by strengthening their safety surveillance systems and ensuring inspection readiness. From designing signal detection frameworks to conducting pharmacovigilance audits and compliance training, our team delivers scalable solutions tailored to regulatory expectations.

Our commitment is to help sponsors integrate pharmacovigilance with real-time data monitoring and analytics to safeguard patient safety and ensure regulatory alignment throughout the product lifecycle.

You can access the full FDA guidance here:

Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment | FDA