Before pharmaceutical products and medical devices reach the market, they must undergo rigorous regulatory oversight and multi-layered review processes. Among these processes, the Pre-Approval Inspection (PAI) stands as an exceptionally critical milestone. This article provides a comprehensive explanation of the purpose and scope of PAI, the importance of commercial manufacturing readiness, and the significance of data integrity audits—written in a manner accessible to beginners while maintaining professional rigor.
What is Pre-Approval Inspection (PAI)?
PAI is an on-site inspection conducted by regulatory authorities such as the U.S. Food and Drug Administration (FDA) or Japan’s Pharmaceuticals and Medical Devices Agency (PMDA) at manufacturing facilities prior to product approval. The primary objective is to verify that the manufacturing methods and quality control systems described in the application are actually being implemented appropriately at the facility. This inspection serves as the final confirmation that the product can be consistently manufactured with appropriate quality.
According to FDA guidance documents and the Compliance Program Guidance Manual (CPGM 7346.832), PAI has evolved into a comprehensive, risk-based evaluation system. The FDA’s PAI program was established around 1999 to transform what had been an ad hoc approach into a well-defined, systematic program that ensures consistent and thorough assessments.
Facilities Subject to PAI
Any manufacturing site named for the first time in the following applications is subject to PAI:
- New Drug Application (NDA) and Abbreviated New Drug Application (ANDA) for both human and animal drugs
- Biologics License Application (BLA)
- Premarket Approval (PMA) for medical devices
PAI may also be conducted at supporting vendors and service providers listed in the application, including active pharmaceutical ingredient (API) manufacturers, control testing laboratories, and packaging facilities.
Three Primary Objectives of PAI
According to FDA guidance and industry best practices, PAI has three clearly defined primary objectives:
Objective 1: Readiness for Commercial Manufacturing
The first objective is to determine whether the establishment has a pharmaceutical quality system designed to achieve sufficient control over the facility and commercial manufacturing operations.
New drug or new product applications often involve test production conducted at reduced scale. However, following approval, production must transition to large-scale commercial manufacturing, making readiness assessment critically important. During PAI, inspectors scrutinize a wide range of elements, including equipment, process controls, personnel training status, and raw material receiving systems. This comprehensive evaluation confirms whether the facility has established a system capable of consistently maintaining production quality.
The assessment extends beyond merely confirming that equipment exists; it evaluates whether standard operating procedures (SOPs) are properly documented, whether personnel are adequately trained to execute those procedures, and whether quality control systems function effectively in practice.
Objective 2: Conformance to Application
The second objective is to verify that the formulation, manufacturing or processing methods, and analytical (or examination) methods are consistent with descriptions contained in the Chemistry, Manufacturing, and Controls (CMC) section of the application.
This verification applies to the bio-batch (and other pivotal clinical batches, when applicable), the proposed commercial scale batch, and the active pharmaceutical ingredient(s). Inspectors compare the actual manufacturing process observed during the PAI against what was documented in the application to ensure consistency and verify that the product can be reliably reproduced according to the submitted specifications.
During this phase, inspectors examine complete batch records, deviation reports, and out-of-specification (OOS) investigations related to conformance batches. Process validation reports are also reviewed to confirm that conclusions align with raw data observed on the manufacturing floor.
Objective 3: Data Integrity Audit
The third objective is to audit the raw data—whether hardcopy or electronic—to authenticate the data submitted in the CMC section of the application.
In recent years, data integrity has become a particularly emphasized area of focus during PAI inspections.
The Heightened Importance of Data Integrity Audits
Data integrity refers to ensuring that “records remain accurate and unaltered at any point in time.” The accuracy, truthfulness, and completeness of data submitted during the approval application process and recorded at manufacturing sites form the foundation of product quality assurance.
ALCOA and ALCOA+ Principles
Data integrity is typically evaluated according to the ALCOA principles, which have evolved into ALCOA+ and even ALCOA++ frameworks:
ALCOA (Core Principles):
- Attributable: Every action can be traced back to a specific individual
- Legible: Data is clear, readable, and understandable
- Contemporaneous: Data is recorded promptly and in real-time
- Original: Documents must be original or certified “true copies”
- Accurate: Data must reflect exactly what was observed and recorded
ALCOA+ (Additional Criteria):
- Complete: Data must be documented fully, with nothing omitted
- Consistent: Documentation should be chronological and orderly
- Enduring: Records must be maintained for the duration specified by regulatory authorities
- Available: Documents must be accessible when needed for reference or audit purposes
ALCOA++ (Further Enhanced): Additional principles include Integrity, Robustness, Transparency, Accountability, and Reliability. These enhanced frameworks strengthen data reliability and traceability, further aligning with the rigorous standards of highly regulated industries.
Data Integrity in PAI Context
During PAI, inspectors rigorously verify whether records and data are appropriately managed and preserved, and whether audit trails enable complete traceability of all operations. This extends to both paper-based and electronic records. Modern electronic systems must include comprehensive audit trail capabilities, electronic signature functionality, data verification at input and output stages, and robust backup, recovery, and data transfer processes.
If fraud or record tampering is discovered, approval itself may be denied. According to 2024 FDA inspection data, data integrity issues remained among the most frequently cited deficiencies, with problems including:
- Poor or absent attribution of activities (lack of segregated roles, use of generic accounts)
- Poor or absent legibility (overwritten or altered records without justification, disabled audit trails)
- Questionable accuracy (uncontrolled data transformations, manual calculations without independent oversight)
- Governance issues with physical records (discarded original cGMP records, inadequate document controls)
The FDA and other regulatory authorities have emphasized that data integrity is not merely an IT issue but a fundamental quality system requirement that must be embedded throughout the organization’s culture and processes.
Integration with ICH Q10 Pharmaceutical Quality System
The importance of PAI extends beyond a single inspection event—it reflects the organization’s overall pharmaceutical quality system maturity. The International Council for Harmonisation (ICH) Q10 guideline on Pharmaceutical Quality Systems provides a harmonized, lifecycle approach to quality management that complements PAI objectives.
ICH Q10 emphasizes:
- Management responsibilities and quality culture throughout the product lifecycle
- Process performance and product quality monitoring systems
- Corrective and Preventive Action (CAPA) systems
- Change management systems
- Management review and continuous improvement
The FDA’s Quality Management Maturity (QMM) program, launched in 2022, evaluates pharmaceutical companies on Q10-aligned attributes including culture, governance, and continual improvement. A 2024 workshop report described QMM as “the FDA’s blueprint for pharmaceutical excellence.” Achieving high QMM scores is increasingly becoming a competitive advantage and regulatory expectation.
Organizations that implement robust ICH Q10-compliant systems demonstrate measurability through quality metrics such as deviation rates, on-time CAPA closure, and batch failure rates. Some companies have reported measurable improvements—for example, reducing deviations by 30% within two years through strengthened CAPA and trend analysis aligned with Q10 principles.
Remote Assessment and Alternative Tools
The COVID-19 pandemic accelerated regulatory adoption of alternative assessment tools. In September 2025, the FDA issued final guidance on “Alternative Tools: Assessing Drug Manufacturing Facilities Identified in Pending Applications,” which formalizes the continued use of:
- Remote Interactive Evaluations (RIEs)—real-time remote inspections using video technology
- Review of foreign regulatory inspection reports
- Records and information requests under 704(a)(4) authority
These alternative tools may be used in advance of, in lieu of, or to supplement traditional on-site PAI. However, facilities must respond to information requests within specified timeframes (typically 15-30 U.S. business days) and provide complete, accurate information. Insufficient responses may delay application action or result in complete response letters.
Implications for Industry
When problems are identified during PAI on-site inspections, approval review may be suspended or significantly delayed. Therefore, organizations must maintain high standards in both production systems and data management from early development stages. For companies pursuing global expansion, compliance with regulatory standards across multiple jurisdictions becomes an unavoidable challenge.
Preparation Best Practices
Industry experts recommend the following preparation strategies:
- Conduct Mock PAIs: Perform mock inspections 6-12 months before anticipated PAI to allow sufficient time to address identified issues
- Establish PAI Readiness Teams: Designate cross-functional teams responsible for inspection preparation and coordination
- Implement Document Freeze Protocols: Establish temporary change control freezes on critical PAI-related documents 30-45 days before the expected inspection window
- Train Personnel: Provide comprehensive training to all individuals who could be involved in the inspection, focusing on expectations, inspector interactions, and proper documentation practices
- Maintain Centralized Documentation: Ensure all critical documents, batch records, validation reports, and supporting data are readily accessible and organized
- Review Quality Metrics: Regularly monitor quality key performance indicators (KPIs) to demonstrate system effectiveness
The PAI represents the starting point for all future inspections. Once approval is granted, facilities become subject to routine inspections. While risk assessment determines inspection frequency, domestic facilities can typically expect inspections every two years, while facilities located outside the United States can expect periodic notifications of impending inspections.
Conclusion
Pre-Approval Inspection (PAI) is an exceptionally important process through which regulatory authorities directly verify the reality of pharmaceutical and medical device manufacturing sites. The inspection focuses on commercial manufacturing readiness and data integrity assurance; deficiencies in these areas prevent products from reaching the market.
Conversely, successfully navigating PAI enables companies to gain trust in their ability to safely deliver high-quality products to patients. Understanding the significance of PAI and the importance of appropriate preparation has become increasingly essential as regulatory expectations continue to evolve.
In today’s environment, where data integrity is emphasized through frameworks like ALCOA++, pharmaceutical quality systems are evaluated through ICH Q10 principles, and regulatory authorities employ risk-based, lifecycle approaches to oversight—organizations must view PAI not as a hurdle to overcome but as an opportunity to demonstrate quality system maturity and commitment to patient safety.
The integration of emerging technologies, remote assessment tools, and enhanced data governance requirements means that PAI preparation is no longer a discrete event but rather an ongoing commitment to quality excellence embedded throughout the organization’s culture and operations.
Comment