๐‡๐จ๐ฐ ๐ญ๐ก๐ž ๐…๐ƒ๐€’๐ฌ ๐๐ž๐ฐ ๐‚๐จ๐ฆ๐ฆ๐ฎ๐ง๐ข๐œ๐š๐ญ๐ข๐จ๐ง ๐Œ๐ž๐š๐ฌ๐ฎ๐ซ๐ž๐ฌ ๐‚๐š๐ง ๐…๐š๐ฌ๐ญ-๐“๐ซ๐š๐œ๐ค ๐๐ข๐จ๐ฉ๐ก๐š๐ซ๐ฆ๐š๐œ๐ž๐ฎ๐ญ๐ข๐œ๐š๐ฅ ๐ƒ๐ž๐ฏ๐ž๐ฅ๐จ๐ฉ๐ฆ๐ž๐ง๐ญ โ€“ (Biopharma Regulatory Affairs)

Authored by Lital Israeli Yagev

The Food and Drug Administration (FDA) has always been pivotal in drug and biopharmaceutical development. With the enactment of the Prescription Drug User Fee Act (PDUFA) VII for the period 2023-2027, the FDA has introduced innovative measures that promise to revolutionize the way biopharmaceutical developers communicate with the agency, potentially fast-tracking the development process.

One of the main introductions under PDUFA VII is the expansion of the INTERACT meeting program. Originally, this program was designed to offer early advice on various aspects such as toxicology, proof-of-concept, biodistribution study design, Chemistry, Manufacturing, and Controls (more on CMC regulatory affairs) issues, and first-in-human trials, primarily in collaboration with the Center for Biologics Evaluation and Research (CBER). Now, this program has extended its reach to include similar engagements with the Center for Drug Evaluation and Research (CDER). This expansion is a significant boon for developers of complex pharmaceuticals, particularly at stages where clear guideline documents may not be readily available. By engaging with the FDA in these early stages, developers can gain invaluable insights and guidance, smoothing the path for their innovative products and the opportunity for another free consultation with the FDA, as part of the Pre-IND meeting for a later stage.

Adres operates as a regulatory affairs consultancy specializing in regulatory affairs, Medical QA and CMC. Navigating the intricate crossroads of innovation and regulation can be daunting, and securing professional advice is not merely advantageousโ€”it is imperative.
We encourage you to seek assistance without reservation. We are here for you incase you have questions. Come pick our brains!

In addition to the INTERACT meetings, the FDA has introduced a new Type D meeting. This meeting format is designed to accelerate feedback from the agency on a narrow set of issues, limited to two focused topics. The unique aspect of this meeting type is its efficiency and specificity, involving only a few associated questions and the participation of no more than three disciplines within the FDA. What makes Type D meetings particularly appealing is the FDA’s  commitment to providing response to meeting request within 14 days and written response within 50 calendar days. This rapid turnaround  enables developers to receive timely feedback and make quicker decisions in their development process.

These new communication routes under PDUFA VII represent a significant step forward in the FDA’s approach to supporting drug and biopharmaceutical development. By offering more structured and timely interactions, the FDA acknowledges the complexities of modern pharmaceutical development and actively works to facilitate and expedite these processes.


The FDA’s new communication measures under PDUFA VII mark a pivotal shift in the landscape of biopharmaceutical development. By embracing these opportunities, developers can navigate the regulatory process more efficiently, bringing their groundbreaking treatments to the market faster than ever before.

Donโ€™t hesitate to reach out for support. Remember, in this journey, youโ€™re not alone. Feel free to contact us and pick our minds.

What about the Third Stage? Continuous Process Validation (CPV)ย 

Authored by Or Degany

In the intricate world of pharmaceutical and biopharmaceutical manufacturing, pharma regulatory consultants emphasize the importance of ensuring consistent quality and regulatory compliance. Continuous Process Validation (CPV) is a critical player in this arena, often recognized as the third stage in the process validation lifecycle. CPV’s primary aim is to demonstrate that a commercial pharmaceutical and biopharmaceutical manufacturing process remains in a state of control post-approval.

Understanding CPV Implementation Challenges

Implementing a CPV program, especially for legacy biopharmaceutical products with a long manufacturing history, poses significant challenges. These products, which have been licensed and manufactured over extended periods, may not initially align with contemporary CPV methodologies. Their process designs may be less rigorous, control strategies less developed, and process parameter classifications not fully accomplished. This gap between older manufacturing processes and current regulatory expectations requires careful navigation.

ADRES: Your Partner in CPV Implementation

At ADRES, we understand the intricacies of implementing CPV in the biopharmaceutical sector. Implementing CPV can be daunting, especially when dealing with legacy products. Understanding the nuances of process control, regulatory compliance, and quality assurance within the CPV framework requires expertise and experience. 

If you’re navigating these complex waters, reaching out for expert assistance can make a significant difference.

Whether you are just beginning to explore the world of CPV or want to enhance your existing program, our team at ADRES is here to offer guidance and support. We are committed to helping you understand and implement CPV effectively, ensuring your pharmaceutical and biopharmaceutical manufacturing processes meet the highest quality and control standards.

If you have any questions or need further assistance, please feel free to reach out to us at https://adres.bio/contact-us.

Unraveling the Intricacies of PRIME: Your Pathway to Accelerated Medicine Approval (Biotech Regulatory Affairs)


Authored by Dr. Liron Gibbs-Bar

Embarking on the regulatory affairs journey of bringing a new medicinal product to the market is no small feat. The road is long and filled with regulatory hurdles, especially when aiming to meet the high standards required for regulatory approval and market access. However, there’s a beacon of hope for applicants, particularly those in the academic sector and small to medium-sized enterprises (SMEs): the PRIME scheme by the European Medicines Agency (EMA).

Launched in 2016, the PRIority MEdicines (PRIME) scheme represents a pivotal shift in the EMA’s approach to medicinal regulation. Its primary goal is straightforward yet ambitious: to offer continuous scientific and regulatory guidance, coupled with an accelerated assessment process for new medicines that target unmet medical needs and hold significant public interest.

For researchers and companies working on groundbreaking treatments, PRIME offers a unique opportunity. Early Entry PRIME status can be a game-changer, especially for SMEs and academic applicants. This status is granted when proof of principle is demonstrated, opening doors to invaluable guidance from regulators early in the development process.

The EMA’s recent revision to the PRIME guidelines introduces several key features to enhance the scheme’s effectiveness:

  1. Pre-submission meetings to assist applicants in planning their PRIME application.
  2. Expedited follow-up scientific advice with shortened timelines, providing quicker access to essential guidance.
  3. A submission readiness meeting about a year before the marketing authorization application (MAA) filing date, offering a platform to discuss the development status and readiness of the marketing application dossier.
  4. A revamped approach involving the submission and maintenance of a regulatory roadmap and product development tracker, taking the place of the PRIME annual update.

These enhancements are designed to deepen knowledge, and support accelerated assessments, significantly benefiting those navigating the complex waters of medicinal product development.

At ADRES, we understand the intricacies of the regulatory landscape, including the PRIME designation. Our team has extensive regulatory submissions experience, catering to SME and non-SME companies. 

We recognize the challenges and opportunities of seeking PRIME status and are equipped to guide you every step of the way.

Navigating the PRIME pathway can be daunting, but with proper support and guidance, it’s a journey you can embark on with confidence. 

You’ve got this!

If you have any questions or need further assistance, please feel free to reach out to us at https://adres.bio/contact-us.

Phase-appropriate validation – Biotech Regulatory Affairsย 

By Inbal Apel

Are you a biotech or biopharma company embarking on the challenging journey of clinical trials? The road ahead, particularly in the early stages, is often fraught with complexities and uncertainties. One key area that usually raises questions and concerns is validating testing methods and manufacturing processes. If thoughts about CMC regulatory affairs keep you up at night, you’re not alone.

In the nascent phases of clinical trials, it’s typical for manufacturing methods and processes to be in a state of evolution. They are often still being refined, adjusted, and understood. This fluidity can make full validation not just challenging but sometimes unnecessary and even counterproductive. This is especially true for companies taking their first steps in clinical research and development.

To provide some perspective, let’s consider the guidelines laid out by the European Medicines Agency (EMA). Specifically, their guideline on “strategies to identify and mitigate risks for first-in-human and early clinical trials with investigational medicinal products.” This guideline offers a critical insight: it points out that “the methods used for determination of the strength and/or the potency of the product need to be relevant for the intended mechanism of action, reliable, and qualified.” Note the emphasis on being ‘qualified’ rather than ‘validated.’ This distinction is crucial and underscores a more tailored approach to validation that is phase-appropriate.

Understanding the nuances of these requirements is vital for navigating the regulatory landscape successfully. It’s not just about ticking boxes; it’s about ensuring that your methods are robust, reliable, and appropriate for the stage of development your product is in. This phase-appropriate validation approach allows for a more flexible, adaptive, and ultimately more effective and cheap pathway through the early stages of clinical development.

But how do you translate these guidelines into actionable steps? How do you plan and align your company’s Gantt chart for each stage of clinical development? The task can seem daunting, especially when juggling the myriad other responsibilities that come with running a biotech or biopharma company.

This is where expert guidance on regulatory affairs can make a world of difference. Seeking advice from those who have navigated these waters before can provide clarity and direction. It can transform a confusing maze of regulations and best practices into a clear roadmap towards successful clinical trial progression.

regulatory affairs consulting

Adres is a regulatory affairs consultancy. For those who find themselves at this complex intersection of innovation and regulation, seeking expert guidance is not just helpful โ€“ it’s essential. Don’t hesitate to reach out for support. Remember, in this journey, you’re not alone.

Feel free to contact us and pick our minds https://adres.bio/contact-us.

Implementing the New GCP R3 Guidelines.

Authored byย Carmel Dafna

The updated ICH E6 R3 introduces key improvements from Version 2. Here are fundamental changes, focusing on how they directly impact the sponsor role:


Increased focus on Sponsor Oversight:
The update focuses on the responsibility of the sponsors to have a greater level of oversight. ICH E6 Version 3 emphasizes a more robust and proactive approach to ensure that the sponsors will play a critical role in protecting the participant’s safety and the integrity of clinical trials. This includes increasing the highlight on risk management and quality management systems.
 
Enhanced Sponsor Quality Assurance:
Version 3 emphasizes a proactive approach to quality management for sponsors and suggests tools and strategies to ensure the highest standards of data integrity, compliance, and overall trial success. This includes recommendations for sponsors to adopt Quality Management Systems, indicating that Sponsor Quality Assurance is adapting and evolving with the clinical research.
 
Risk-Based Monitoring Integration:
Significant change, allowing sponsors to evaluate how to allocate resources where they are most needed. This ensures sponsors can cultivate a more dynamic and responsive approach to managing study conduct and identifying risks.
 
Data Integrity and Security:
With an increased focus on data integrity and security, sponsors should be able to protect sensitive information throughout the trial. This not only addresses compliance concerns but also improves the reliability of the data generated.
 
Continuous Improvement Mindset:
ICH E6 Version 3 asks for continuous improvement from sponsors, expecting them to proactively assess and improve their processes to ensure that sponsors will optimize trial conduct and outcomes.

If you have any questions or need further assistance, please feel free to reach out to us at https://adres.bio/contact-us.

Skip to content