The Challenge of Cleanroom Classification Harmonization

In the world of pharmaceutical manufacturing, cleanroom classifications play a crucial role in ensuring product quality and patient safety. However, a significant hurdle in the global harmonization of regulations has been a pain in our sides for a long time, that highlights the persistent differences between major regulatory bodies, including the FDA, EMA, and others, despite efforts to align through organizations like the World Health Organization (WHO) and the Pharmaceutical Inspection Co-operation Scheme (PIC/S).

The Current Landscape

United States Approach

In the United States, cleanroom classifications are primarily governed by two key documents:

  1. The FDA’s “Sterile Drug Products Produced by Aseptic Processing” guidance
  2. ISO 14644-1 standard for cleanroom classifications

The ISO 14644-1 standard is particularly noteworthy as it’s a general standard applicable across various industries utilizing cleanrooms, not just pharmaceuticals.

European Union Approach

The European Union takes a different stance, employing a grading system outlined in the EU GMP guide:

  • Grades A through D are used for normal cleanroom operation
  • ISO 14644 is still utilized, but primarily for validation purposes

World Health Organization Alignment

The World Health Organization (WHO) aligns with the European approach, adopting the same A to D grading system in its GMP guidelines.

The Implications of Disharmony

This lack of harmonization in cleanroom classifications presents several challenges:

  1. Regulatory Complexity: Companies operating globally must navigate different classification systems, potentially leading to confusion and increased compliance costs.
  2. Technology Transfer Issues: Transferring manufacturing processes between regions becomes more complicated when cleanroom requirements differ.
  3. Inspection Inconsistencies: Differences in classification systems can lead to varying interpretations during inspections by different regulatory bodies.

The Missed Opportunity in Annex 1

The recent update to Annex 1, a key document in GMP regulations, could have been a prime opportunity to address this disharmony. However, despite involvement from WHO and PIC/S (and through them the FDA), the update failed to bring about the hoped-for alignment in cleanroom classifications.

Moving Forward

As the pharmaceutical industry continues to globalize, the need for harmonized regulations continues to be central. I would love to see future efforts towards harmonization here that would:

  1. Prioritize alignment on fundamental technical specifications like cleanroom classifications
  2. Consider the practical implications for manufacturers operating across multiple jurisdictions

While the journey towards full regulatory harmonization may be long and challenging, addressing key discrepancies like cleanroom classifications would represent a significant step forward for the global pharmaceutical industry.

EMA GMP Plans for Regulation Updates

Like one does, I watch upcoming regulations like a hawk. Here are a few of the forthcoming GMP changes coming from the 3-year work plan for the Inspectors Working Group.

DocumentIntended ChangesWhenMy Thoughts
GMP Guide: Chapter 4 (Documentation)Assure data integrity in the context of GMP. This would be in parallel with similar consideration of Annex 11 (Computerised Systems).Q1 2026An update is needed to align with current thinking. Data Integrity has advanced significantly in the last five years, and Chapter 4 could benefit from alignment with the PIC/S guidance.
GMP Guide: Annex 11 (Computerised Systems)Assure data integrity in the context of GMP. This would be in parallel with similar consideration of Chapter 4 (Documentation).Q1 2026A necessary update. Will be curious to see how it aligns with the FDA’s CSA approach (which isn’t really all that new).

We pretty much know what will be in it from the concept paper. At least it will solidify this requirement for cloud systems “Regulated users should
26 have access to the complete documentation for validation and safe operation of a system and be able to present this during regulatory inspections, e.g. with the help of the service provider.”
Guidelines on GMP specific to ATMPSReview the Guidelines in collaboration with CAT and the European Commission
following the publication of a new regulation on standards of quality and safety for substances of human origin intended for human application and need to update legal references and definitions.
Review the Guidelines in the light of new Annex 1 Manufacture of Sterile Medicinal Products and consider whether any updates are necessary.
Q4 2026This is a fast area of change, and this update is called for.

Aligning to Annex 1 is overdue.
GMP Guide: Annex 3 Manufacture of RadiopharmaceuticalsA review and update of the Annex to reflect current state of the art.Q4 2026I’ve never worked in radiopharmaceuticals. Maybe someday.
GMP Guide: Annex 15 Qualification and ValidationIn the context of new technology in facilities, products and processes and following
up on LLE recommendations, and extend the scope to APIs.
Q4 2025LLE is the EMA’s lessons learnt report (LLE) on Nitrosamines.

I’d love to see significant changes to finally align with ATSM E2500 and other recent challenges in validation.
GMP Guide: Annex 16 Certification by a Qualified Person and Batch ReleaseFollowing up on LLE recommendations.Q4 2025I’m not a massive fan of QPs as structured. Not expecting that to change.
GMP and Marketing Authorisation HoldersTo revise the paper in line with recommendations from the Nitrosamines LLE, to strengthen guidance for MAHs in terms of having adequate quality agreement with manufactures.Q4 2025Anything to strengthen quality agreements is probably a good thing.

Anytime we see a major chapter update in the Eudralex Volume 4 is an exciting year, and the next few promise to be big. Maybe not Annex 1 big, but maybe the EMA and PIC/S will surprise us.

Leveraging Inspection Manuals for GMP Inspection Readiness

The various agency inspection manuals are critical tools for inspection readiness. I want to lay out where to find some of these manuals and then go deep into pre-approval inspections, focusing on data integrity.

European Medicines Agency

The European Medicines Agency (EMA) has established detailed procedures and work instructions for coordinating and conducting Good Clinical Practice (GCP), Good Manufacturing Practice (GMP), and pharmacovigilance inspections. Here are the key points regarding EMA’s inspection procedures:

GCP Inspection Procedures

  • EMA identifies applications for GCP inspections based on risk assessment criteria and exchanges information on shared applications with the FDA.
  • Inspections can be joint (conducted concurrently by EMA and FDA inspectors) or sequential (conducted separately by each agency).
  • EMA notifies the applicant/marketing authorization holder (MAH) and inspects sites about upcoming inspections through the IRIS industry portal instead of formal letters.
  • Applicants/MAHs must provide a signed statement accepting the inspection and granting direct access to documents and medical records.
  • Requested documents should be provided directly to inspectors in electronic format after consulting the reporting inspector.
  • After the inspection, EMA receives the draft inspection report, finalizes it with the inspectee’s responses, and publishes it in IRIS.

GMP Inspection Procedures

  • EMA coordinates GMP inspections based on risk assessment for marketing authorization applications, variations, and routine re-inspections.
  • Work instructions cover areas such as inspection announcement, fee calculation, product sampling/testing, and report circulation.

Pharmacovigilance Inspection Procedures

  • EMA has specific procedures for coordinating pharmacovigilance inspections and managing non-compliance notifications from MAHs.
  • Work instructions detail the inspection program creation, data entry in databases, and interactions with third-country inspectorates.

The EMA aims to harmonize inspection processes with the FDA and other regulatory bodies to streamline collaboration and information sharing while ensuring clinical trial subject protection and product quality.

FDA

The FDA Investigations Operations Manual (IOM) is the primary inspection manual used by FDA personnel when performing inspections and investigations.

The key points about the IOM are:

  • It provides comprehensive instructions, procedures, and policies for FDA investigators and inspectors to follow when conducting inspections, surveys, and investigations.
  • It covers inspectional activities for foods, drugs, medical devices, biologics, cosmetics, and other FDA-regulated products.
  • The manual details procedures for inspections of manufacturing facilities, sampling, import operations, recalls, consumer complaints, and other compliance activities.
  • It aims to ensure inspections are conducted consistently across FDA field offices and provide clear guidance to the industry on the FDA’s inspection approach.
  • The IOM is updated periodically to incorporate new laws, regulations, policies, and technological changes impacting FDA’s operations.
  • While not legally binding, the IOM represents the FDA’s current thinking and policies on inspections and investigations.

The FDA Investigations Operations Manual serves as the comprehensive inspection reference and procedure manual for FDA field staff carrying out the agency’s oversight and enforcement activities across all regulated product areas.

Pre-Approval Inspections

For new facilities, CPGM 7346.832, the FDA’s Compliance Program Guidance Manual for Pre-Approval Inspections (PAIs) of drug manufacturing facilities, is critical to spend time with. It outlines the objectives and procedures for FDA inspectors to evaluate a facility’s readiness for commercial manufacturing before approving a new drug application.

The key objectives of CPGM 7346.832 are:

  1. Assess if the facility has a quality system capable of controlling commercial manufacturing operations.
  2. Verify that the manufacturing processes, formulation, and analytical methods conform to the application details.
  3. Audit raw data integrity to authenticate the data submitted in the application.
  4. Evaluate the facility’s commitment to quality in pharmaceutical development (new objective added in 2022 revision).

The guidance instructs inspectors on evaluating the firm’s quality systems, process validation, data integrity, laboratory controls, change management, investigations, batch release procedures, and compliance with current Good Manufacturing Practices (cGMPs). It aims to ensure the facility can reliably produce the drug product described in the application.

Data Integrity

CPGM 7346.832 has specific requirements for data integrity audits during drug manufacturing facility pre-approval inspections (PAIs). Utilizing this document is an excellent way to evaluate your data integrity program.

The key points are:

  1. Objective 3 of the guidance is “Data Integrity Audit”—auditing and verifying raw data associated with the product to authenticate the data submitted in the application.
  2. Inspectors must audit the accuracy and completeness of data reported by the facility for the product. This involves verifying the factual integrity (data matches what was submitted) and contextual integrity (supporting data is complete).
  3. Inspectors should examine raw data, such as chromatograms, analyst notebooks, electronic data, etc., and compare it to the summary data in the application’s Chemistry, Manufacturing, and Controls (CMC) section.
  4. The data integrity audit should focus on finished product stability, dissolution, content uniformity, API impurities, etc.
  5. Inspectors must identify any unreported relevant data, data falsification, improper invalidation of results, or unexplained data discrepancies.
  6. Indications of data integrity issues include altered raw data, references to failing studies, discrepancies between samples, and missing records.

The data integrity audit aims to ensure the CMC data submitted to FDA is complete, reliable, and can be fully authenticated from the raw data at the manufacturing site. Robust data integrity is critical for the FDA to decide on the application’s approval.

European Guideline on Data Integrity in GCP Studies

The EMA has published “Guideline on computerised systems and electronic data in clinical trials.”

Anyone familiar with Annex 11 of Eudralex Annex 4 won’t be surprised by the content, but frankly I expect a lot of folks who have primarily experience on the clinical side will be scratching their heads. The fact that the authors felt the need to have an entire paragraph dedicated to unique user names is telling.

This is a great resource for sponsors who need to figure out just what to evaluate at investigators sites, a requirement this guideline repeats multiple times.

I’ll be very curious how effective sponsors are in ensuring this requirement is met “The investigator should receive an introduction on how to navigate the audit trail of their own data in order to be able to review changes.”

EMA Publishes 2021 GCP Compliance Report

The EMA has published the Annual Report of the Good Clinical Practice (GCP) Inspectors Working Group (IWG) 2021.

Beyond wishing for an 11 month cycle of writing and approval on my annual reports, there is some valuable information there.

In 2021, three CHMP GCP inspections were conducted entirely remotely, and three inspections were conducted in a hybrid setting. A total of 286 deficiencies, comprising 24 critical, 152 major and 110 minor findings were recorded for the 27 CHMP requested inspections conducted in 2021. This represents an average of 10-11 findings per site inspected. The three top categories were: “General”, “Trial Management” and “Computer System”. An increase in findings related to computer systems (e. g. Audit Trail and Authorized Access, Computer Validation, Physical Security System and Backup) is noted compared to the last reports.

More information is available at EMA´s Good Clinical Practice Inspectors Working Group website.

Under organisation and personel we see “Delegation of tasks to inappropriate team members.” This reinforces the needs for strong cv and job descriptions, and linking to both hiring and personnel qualification.

The computer systems observations are the greatest hits of data integrity, and should be a wakeup call to any company that treats GCP and GMP computer systems differently.

Let the 2022 annual GCP training development begin. And make sure you get that training done on time!