Silence Gets You Nowhere – FDA Layoffs

Add me to the list of people who are disheartened y the silence of the Pharmaceutical Research and Manufacturers of America and the Biotechnology Innovation Organization to the cuts at the FDA. In an interest to write something that should be coming loud and clear from our industry groups, I give you…

The Impact of Recent FDA Layoffs on Agency Capacity and Public Health

The recent wave of layoffs at the U.S. Food and Drug Administration (FDA), enacted as part of broader illegal federal workforce reductions under the Trump administration, has exacerbated long-standing staffing challenges at the agency. By targeting probationary employees—recent hires and those promoted within the past two years—the cuts have disproportionately affected early-career professionals with cutting-edge technical expertise, disrupted workforce development pipelines, and weakened oversight in critical areas such as medical device regulation, food safety, veterinary medicine, and emerging technologies. These reductions come at a time when the FDA is already grappling with recruitment challenges, inspection backlogs, and increasing demands for regulatory innovation.

Scope and Targets of the Layoffs

The Department of Health and Human Services (HHS), under Secretary Robert F. Kennedy Jr., terminated approximately 5,200 probationary employees across its agencies in mid-February 2025, including hundreds at the FDA. While the agency’s drug review divisions were largely spared, layoffs hit staff in the Center for Devices and Radiological Health (CDRH), the Center for Food Safety and Applied Nutrition (CFSAN), the Center for Veterinary Medicine (CVM), and the Center for Tobacco Products (CTP).

Medical Devices and Digital Health

In CDRH, at least 230 employees were dismissed, including specialists in artificial intelligence (AI) and digital health—fields undergoing rapid technological advancement. These roles are critical for evaluating AI-driven diagnostic tools, wearable devices, and software-as-a-medical-device (SaMD) products. The loss of early-career researchers and engineers threatens the FDA’s ability to keep pace with industry innovation, potentially delaying approvals for technologies like neural interfaces and AI-powered imaging systems.

Food Safety and Additives

CFSAN lost staff responsible for reviewing new food additives, colorings, and ingredients—a priority area for Kennedy, who has advocated for stricter chemical regulations. With approximately 2,000 uninspected food and drug facilities globally, the FDA’s inspection backlog is now likely to grow further, raising risks of contamination incidents similar to recent outbreaks linked to infant formula and baby food.

Veterinary Medicine

The Center for Veterinary Medicine (CVM) saw cuts to reviewers assessing the safety of pharmaceuticals for pets and livestock. These roles ensure that medications for animals are effective and that residues in products like milk and eggs remain safe for human consumption. Reductions here could delay approvals for veterinary drugs and weaken monitoring of antimicrobial resistance.

Exacerbating Existing Staffing Challenges

The FDA has historically struggled to recruit and retain specialized staff due to competition from higher-paying private-sector roles. The layoffs worsen these issues by destabilizing workforce development and eroding institutional knowledge.

Loss of Early-Career Talent

Probationary employees—often younger professionals with advanced degrees in fields like data science, bioengineering, and toxicology—represent the FDA’s pipeline for replacing retiring staff. By targeting this group, the cuts disrupt the “learning chain” essential for maintaining expertise. As Kenneth Kaitin, a Tufts University professor, noted: “You’re eliminating the learning chain of people who come into the agency. There’s a long learning curve, and you’re eliminating people at the early stage” (https://www.biopharmadive.com/news/fda-layoffs-trump-doge-hhs-cuts-impact/740499/).

Increased Workloads and Burnout

Remaining staff now face expanded responsibilities. For example, CDRH’s device reviewers, already managing a surge in AI and digital health submissions, must absorb the work of dismissed colleagues without additional support. Similarly, food safety inspectors—many of whom were hired post-pandemic to address backlogs—are now stretched thinner, increasing the likelihood of oversights.

Recruitment and Morale

The layoffs have demoralized the workforce and damaged the FDA’s reputation as a stable employer. As Mitch Zeller, former FDA tobacco director, stated: “The combined effect of what they’re trying to do is going to destroy the ability to recruit and retain talent” (https://www.startribune.com/trump-administration-cuts-reach-fda-employees-in-food-safety-medical-devices-and-tobacco-products/601223844). With hiring frozen under an executive order requiring agencies to replace only one employee for every four departures, the FDA cannot easily rebuild capacity.

The Training Bottleneck

The probationary period at the FDA (1–2 years for new hires) is designed to provide hands-on training in complex regulatory science. Dismissing employees during this phase wastes significant investments in onboarding and delays the development of proficiency.

Specialized Skill Development

Reviewers in areas like AI-driven medical devices or gene therapies require months of training to evaluate technical dossiers, assess clinical data, and understand regulatory precedents. Losing these employees resets progress, forcing the FDA to restart the training process once hiring resumes.

Cross-Departmental Collaboration

New hires often rotate through multiple divisions to build interdisciplinary expertise. For instance, a food additive reviewer might collaborate with toxicologists and epidemiologists to assess long-term health risks. Disrupting these rotations limits opportunities for knowledge-sharing, weakening the agency’s ability to address novel public health challenges.

Long-Term Consequences for Public Health

Slower Product Reviews

User fee-funded positions—which account for nearly half of the FDA’s $6.9 billion budget—were not spared from cuts. Since these roles are financed by industry to expedite reviews, their elimination could delay approvals for new drugs, devices, and food ingredients without reducing federal spending.

Weakened Outbreak Response

The FDA collaborates with the CDC to trace contamination sources during foodborne illness outbreaks. With fewer inspectors and scientists, the agency’s capacity to identify pathogens like Salmonella or Listeria will diminish, prolonging outbreaks and increasing hospitalization risks.

Erosion of Global Leadership

The FDA’s regulatory standards influence global markets. Slower reviews and outdated technical capacity could push companies to seek approvals in regions with more predictable oversight, such as the EU or Singapore, undermining U.S. competitiveness.

Conclusion

The FDA layoffs represent a shortsighted approach to government efficiency that prioritizes short-term spending cuts over long-term public health. By targeting probationary employees, the administration has exacerbated recruitment challenges, disrupted workforce development, and weakened oversight in critical areas. Rebuilding the FDA’s capacity will require reversing hiring freezes, increasing salaries to compete with the private sector, and safeguarding user fee funds from political interference. Without these steps, the agency’s ability to ensure food safety, evaluate emerging technologies, and respond to health crises will continue to erode—with dire consequences for consumers, industry, and global health security.

“The cuts at FDA will be terribly harmful for the American people. Indiscriminately firing people because they are new to the agency makes no sense.”
– Patti Zettler, former HHS Deputy General Counsel (https://www.biopharmadive.com/news/fda-layoffs-trump-doge-hhs-cuts-impact/740499/)

The Role of the HACCP

Reading Strukmyer LLC’s recent FDA Warning Letter, and reflecting back to last year’s Colgate-Palmolive/Tom’s of Maine, Inc. Warning Letter, has me thinking of common language In both warning letters where the FDA asks for “A comprehensive, independent assessment of the design and control of your firm’s manufacturing operations, with a detailed and thorough review of all microbiological hazards.”

It is hard to read that as anything else than a clarion call to use a HACCP.

If that isn’t a HACCP, I don’t know what is. Given the FDA’s rich history and connection to the tool, it is difficult to imagine them thinking of any other tool. Sure, I can invent about 7 other ways to do that, but why bother when there is a great tool, full of powerful uses, waiting to be used that the regulators pretty much have in their DNA.

The Evolution of HACCP in FDA Regulation: A Journey to Enhanced Food Safety

The Hazard Analysis and Critical Control Points (HACCP) system has a fascinating history that is deeply intertwined with FDA regulations. Initially developed in the 1960s by NASA, the Pillsbury Company, and the U.S. Army, HACCP was designed to ensure safe food for space missions. This pioneering collaboration aimed to prevent food safety issues by identifying and controlling critical points in food processing. The success of HACCP in space missions soon led to its application in commercial food production.

In the 1970s, Pillsbury applied HACCP to its commercial operations, driven by incidents such as the contamination of farina with glass. This prompted Pillsbury to adopt HACCP more widely across its production lines. A significant event in 1971 was a panel discussion at the National Conference on Food Protection, which led to the FDA’s involvement in promoting HACCP for food safety inspections. The FDA recognized the potential of HACCP to enhance food safety standards and began to integrate it into its regulatory framework.

As HACCP gained prominence as a food safety standard in the 1980s and 1990s, the National Advisory Committee on Microbiological Criteria for Foods (NACMCF) refined its principles. The committee added preliminary steps and solidified the seven core principles of HACCP, which include hazard analysis, critical control points identification, establishing critical limits, monitoring procedures, corrective actions, verification procedures, and record-keeping. This structured approach helped standardize HACCP implementation across different sectors of the food industry.

A major milestone in the history of HACCP was the implementation of the Pathogen Reduction/HACCP Systems rule by the USDA’s Food Safety and Inspection Service (FSIS) in 1996. This rule mandated HACCP in meat and poultry processing facilities, marking a significant shift towards preventive food safety measures. By the late 1990s, HACCP became a requirement for all food businesses, with some exceptions for smaller operations. This widespread adoption underscored the importance of proactive food safety management.

The Food Safety Modernization Act (FSMA) of 2011 further emphasized preventive controls, including HACCP, to enhance food safety across the industry. FSMA shifted the focus from responding to food safety issues to preventing them, aligning with the core principles of HACCP. Today, HACCP remains a cornerstone of food safety management globally, with ongoing training and certification programs available to ensure compliance with evolving regulations. The FDA continues to support HACCP as part of its broader efforts to protect public health through safe food production and processing practices. As the food industry continues to evolve, the principles of HACCP remain essential for maintaining high standards of food safety and quality.

Why is a HACCP Useful in Biotech Manufacturing

The HACCP seeks to map a process – the manufacturing process, one cleanroom, a series of interlinked cleanrooms, or the water system – and identifies hazards (a point of contamination) by understanding the personnel, material, waste, and other parts of the operational flow. These hazards are assessed at each step in the process for their likelihood and severity. Mitigations are taken to reduce the risk the hazard presents (“a contamination control point”). Where a risk cannot be adequately minimized (either in terms of its likelihood of occurrence, the severity of its nature, or both), this “contamination control point” should be subject to a form of detection so that the facility has an understanding of whether the microbial hazard was potentially present at a given time, for a given operation. In other words, the “critical control point” provides a reasoned area for selecting a monitoring location. For aseptic processing, for example, the target is elimination, even if this cannot be absolutely demonstrated.

The HACCP approach can easily be applied to pharmaceutical manufacturing where it proves very useful for microbial control. Although alternative risk tools exist, such as Failure Modes and Effects Analysis, the HACCP approach is better for microbial control.

The HACCP is a core part of an effective layers of control analysis.

Conducting a HACCP

HACCP provides a systematic approach to identifying and controlling potential hazards throughout the production process.

Step 1: Conduct a Hazard Analysis

  1. List All Process Steps: Begin by detailing every step involved in your biotech manufacturing process, from raw material sourcing to final product packaging. Make sure to walk down the process thoroughly.
  2. Identify Potential Hazards: At each step, identify potential biological, chemical, and physical hazards. Biological hazards might include microbial contamination, while chemical hazards could involve chemical impurities or inappropriate reagents. Physical hazards might include particulates or inappropriate packaging materials.
  3. Evaluate Severity and Likelihood: Assess the severity and likelihood of each identified hazard. This evaluation helps prioritize which hazards require immediate attention.
  4. Determine Preventive Measures: Develop strategies to control significant hazards. This might involve adjusting process conditions, improving cleaning protocols, or enhancing monitoring systems.
  5. Document Justifications: Record the rationale behind including or excluding hazards from your analysis. This documentation is essential for transparency and regulatory compliance.

Step 2: Determine Critical Control Points (CCPs)

  1. Identify Control Points: Any step where biological, chemical, or physical factors can be controlled is considered a control point.
  2. Determine CCPs: Use a decision tree to identify which control points are critical. A CCP is a step at which control can be applied and is essential to prevent or eliminate a hazard or reduce it to an acceptable level.
  3. Establish Critical Limits: For each CCP, define the maximum or minimum values to which parameters must be controlled. These limits ensure that hazards are effectively managed.
Control PointsCritical Control Points
Process steps where a control measure (mitigation activity) is necessary to prevent the hazard from occurringProcess steps where both control and monitoring are necessary to assure product quality and patient safety
Are not necessarily critical control points (CCPs)Are also control points
Determined from the risk associated with the hazardDetermined through a decision tree

Step 3: Establish Monitoring Procedures

  1. Develop Monitoring Plans: Create detailed plans for monitoring each CCP. This includes specifying what to monitor, how often, and who is responsible.
  2. Implement Monitoring Tools: Use appropriate tools and equipment to monitor CCPs effectively. This might include temperature sensors, microbial testing kits, or chemical analyzers.
  3. Record Monitoring Data: Ensure that all monitoring data is accurately recorded and stored for future reference.

Step 4: Establish Corrective Actions

  1. Define Corrective Actions: Develop procedures for when monitoring indicates that a CCP is not within its critical limits. These actions should restore control and prevent hazards.
  2. Proceduralize: You are establishing alternative control strategies here so make sure they are appropriately verified and controlled by process/procedure in the quality system.
  3. Train Staff: Ensure that all personnel understand and can implement corrective actions promptly.

Step 5: Establish Verification Procedures

  1. Regular Audits: Conduct regular audits to verify that the HACCP system is functioning correctly. This includes reviewing monitoring data and observing process operations.
  2. Validation Studies: Perform validation studies to confirm that CCPs are effective in controlling hazards.
  3. Continuous Improvement: Use audit findings to improve the HACCP system over time.

Step 6: Establish Documentation and Record-Keeping

  1. Maintain Detailed Records: Keep comprehensive records of all aspects of the HACCP system, including hazard analyses, CCPs, monitoring data, corrective actions, and verification activities.
  2. Ensure Traceability: Use documentation to ensure traceability throughout the production process, facilitating quick responses to any safety issues.

Step 7: Implement and Review the HACCP Plan

  1. Implement the Plan: Ensure that all personnel involved in biotech manufacturing understand and follow the HACCP plan.
  2. Regular Review: Regularly review and update the HACCP plan to reflect changes in processes, new hazards, or lessons learned from audits and incidents.

Safecor Health Warning Letter Closeout

I got a post on my RSS feed today from the FDA for a closeout letter to Safecor Health in response to the 2023 Warning Letter. Always happy to see an actual closeout letter.

The main takeaways from the FDA warning letter:

Inadequate Line Clearance and Packaging Controls:

    • Safecor failed to properly inspect packaging and labeling facilities before use, leading to potential mix-ups of drug products. This was evidenced by the presence of unrelated tablets and capsules during the packaging of a specific product.
    • The company has a history of product mix-ups, including instances where a vitamin was found in a drug meant to prevent organ transplant rejection and mislabeled blood clot prevention medication.

    Insufficient Cleaning and Maintenance Procedures:

      • The firm lacked adequate procedures for cleaning and maintaining equipment, with unidentified residues found on supposedly clean equipment. This poses a risk of cross-contamination.
      • The company’s cleaning validation program was deemed inadequate, particularly in addressing worst-case scenarios.

      Failure to Test Components:

        • Safecor did not adequately test incoming components, such as water used in drug manufacturing, for purity, strength, and quality.
        • The company relied on visual inspections without performing necessary chemical and microbiological tests.

        Quality Control Unit Deficiencies:

          • The quality control unit failed to ensure compliance with CGMP regulations, including inadequate document control and data integrity issues.
          • Manufacturing records were not properly controlled, and corrections were made using correction fluid, raising concerns about data authenticity.

          Understanding the Distinctions Between 503B Outsourcing Facilities and Compounding Pharmacies

          I get really confused on the differences between compounding pharmacies and outsourcing facilities. I’ve never worked at either, but see a lot of 483s and warning letters. So today I spent some snow day time doing some reading. I then wrote this up as a reminder to myself.

          The Drug Quality and Security Act (DQSA) of 2013 introduced significant changes by distinguishing between compounding pharmacies under Section 503A and outsourcing facilities under Section 503B of the Federal Food, Drug, and Cosmetic Act (FDCA). This distinction is crucial for ensuring the safety and quality of compounded drugs.

          Compounding Pharmacies (503A)

          Definition and Purpose: Compounding pharmacies are licensed by state boards of pharmacy and primarily focus on creating customized medications for individual patients when commercially available drugs do not meet their needs. These pharmacies must adhere to standards set by the United States Pharmacopeia (USP), such as USP 797 for sterile compounding and USP 800 for hazardous drugs.

          Regulatory Framework: Compounding pharmacies operate under the supervision of a licensed pharmacist and require a prescription for each compounded product. They are generally limited to small batches and are not allowed to engage in office-use compounding without a prescription.

          Outsourcing Facilities (503B)

          Definition and Purpose: Outsourcing facilities, on the other hand, are registered with the FDA and specialize in producing large batches of sterile drugs, often without the need for individual prescriptions. These facilities are designed to address drug shortages and provide complex or rarely compounded preparations to healthcare organizations.

          Regulatory Framework: Unlike 503A pharmacies, 503B facilities must comply with FDA’s Current Good Manufacturing Practices (CGMP) to ensure the quality and safety of their products. They are subject to regular FDA inspections and must report on their compounded products.

          Recent Regulatory Actions: The Case of ProRx, LLC

          This research came about because I was reading a recent warning letter issued to ProRx, LLC, which basically stated they were failing to comply with CGMP regulations for 503B outsourcing facilities. The FDA identified serious deficiencies in sterile drug production practices, posing significant patient safety risks.

          Implications for 503B Facilities

          The warning letter to ProRx, LLC, serves as a reminder of the high regulatory bar set for 503B outsourcing facilities. Key implications include:

          • Enhanced Oversight: The FDA’s ability to inspect and enforce cGMP compliance means that 503B facilities must maintain meticulous quality control and production standards.
          • Patient Safety: The primary concern is ensuring that compounded drugs are safe for use. Facilities must address any deficiencies promptly to avoid recalls and protect patient health.
          • Partnerships and Supply Chain: The ability of 503B facilities to supply compounded drugs to healthcare organizations and pharmacies relies on their compliance with FDA regulations. Non-compliance can disrupt supply chains and impact patient access to necessary medications.

          What I take away from my research is that 503B outsourcing facilities are GMP facilities, and are held to the same standard. Good to know as I evaluate their regulatory actions in the future. I think I’ve tended to dismiss them as not being in the same class of regulatory expectations.

          Also, this is the second time this month where I really wonder what regulatory agencies fascination with pharmacists are in GMP facilities. Seems pretty clear to me that being a pharmacist is no indication of any capability around GMP activities.

          Leaks in Single-Use Manufacturing: A Critical Challenge in Bioprocessing

          The recent FDA warning letter to Sanofi highlights a critical issue in biopharmaceutical manufacturing: the integrity of single-use systems (SUS) and the prevention of leaks. This incident serves as a stark reminder of the importance of robust control strategies in bioprocessing, particularly when it comes to high-pressure events and product leakage.

          The Sanofi Case: A Cautionary Tale

          In January 2025, the FDA issued a warning letter to Sanofi regarding their Genzyme facility in Framingham, Massachusetts. The letter cited significant deviations from Current Good Manufacturing Practice (CGMP) for active pharmaceutical ingredients (APIs). One of the key issues highlighted was the company’s failure to address high-pressure events that resulted in in-process product leakage.

          Sanofi had been using an unapproved workaround, replacing shipping bags to control the frequency of high-pressure and in-process leaking events. This deviation was not properly documented or the solution validated.

          A proper control strategy in this context would likely involve:

          1. A validated process modification to prevent or mitigate high-pressure events
          2. Engineering controls or equipment upgrades to handle pressure fluctuations safely
          3. Improved monitoring and alarm systems to detect potential high-pressure situations
          4. Validated procedures for responding to high-pressure events if they occur
          5. A comprehensive risk assessment and mitigation plan related to pressure control in the manufacturing process

          The Importance of Leak Prevention in Single-Use Systems

          Single-use technologies have become increasingly prevalent in biopharmaceutical manufacturing due to their numerous advantages, including reduced risk of cross-contamination and increased flexibility. For all this to work, the integrity of these systems is paramount to ensure product quality and patient safety.

          Leaks in single-use bags can lead to:

          1. Product loss
          2. Contamination risks
          3. Costly production delays
          4. Regulatory non-compliance

          Strategies for Leak Prevention and Detection

          To address the challenges posed by leaks in single-use systems, manufacturers need to consider implementing a comprehensive control strategy. Here are some key approaches:

          1. Integrity Testing

          Implementing robust integrity testing protocols is crucial. Two non-destructive testing methods are particularly suitable for single-use systems:

          • Pressure-based tests: These tests can detect leaks by inflating components with air to a defined pressure. They can identify defects as small as 10 µm in flat bags and 100 µm in large-volume 3D systems.
          • Trace-gas-based tests: Typically using helium, these tests offer the highest level of sterility assurance and can detect even smaller defects.

          2. Risk-Based Quality by Design (QbD) Approach

          Single-use components and the manufacturing process must be established and maintained using a risk-based QbD approach that can help identify potential failure points and implement appropriate controls. This should include:

          • Comprehensive risk assessments
          • Validated procedures for responding to high-pressure events
          • Improved monitoring and alarm systems

          Validated Process Modifications

          Instead of using unapproved workarounds, companies need to develop and validate process modifications to prevent or mitigate high-pressure events. One thing to be extra cautious about is the worry of a temporary solution becoming a permanent one.

          Conclusion

          The Sanofi warning letter serves as a crucial reminder of the importance of maintaining the integrity of single-use systems in biopharmaceutical manufacturing. By implementing comprehensive control strategies, including robust integrity testing, risk-based approaches, and validated process modifications, manufacturers can significantly reduce the risk of leaks and ensure compliance with cGMP standards.

          As the industry continues to embrace single-use technologies, it’s imperative that we remain vigilant in addressing these challenges to maintain product quality, patient safety, and regulatory compliance.