Introduction to Cleaning Validation


Introduction to Cleaning Validation

Cleaning validation is an essential part of Good Manufacturing Practices (GMPs), crucial for avoiding cross-contamination in pharmaceutical products. While cleaning is fundamentally simple, the validation process has become complex and resource-intensive due to regulatory expectations. The validation of cleaning processes in cGMP environments is traditionally time-consuming, particularly in facilities handling multiple products and cleaning protocols. The effort spans across method development, protocol drafting, laboratory testing, and comprehensive report writing. Efforts to streamline this process, including equipment dedication and strategic grouping, often fall short due to insufficient justification and the intricate nature of cleaning validations. The industry's approach to cleaning has been heavily influenced by regulatory expectations, particularly associating cleaning with process validation.

Science-Based Cleaning Process Development

The traditional validation approach, characterized by a preapproved protocol and a fixed number of validation runs, has faced criticism for not necessarily aligning with the actual needs of effective cleaning validation. The industry has struggled with setting appropriate acceptance criteria, often defaulting to conventional standards without questioning their applicability.

Resultantly, there has been an industry shift towards advocating for a more science-based, risk-based, and statistics-based methodology. Initiatives like "GMPs for the 21st Century," Quality by Design (QbD), and Process Analytical Technology (PAT) have driven a move towards more efficient and rational approaches. The introduction of Acceptable Daily Exposure (ADE) standards has further enabled the establishment of more scientifically grounded acceptance criteria, promoting safety while potentially reducing validation efforts in lower-risk contexts. Such  modernized approaches to cleaning validation, leveraging risk-based and statistical methods, can lead to more reliable and safer cleaning procedures, contributing to better patient safety and product quality. Incorporating these methods can streamline the validation process, reduce costs, and enhance operational efficiency, aligning with the industry's move towards a more rational and scientifically grounded framework.

ADE-Derived Cross-Contamination Risk Scale 

The move towards a more systematic approach began significantly with the collaboration between pharmaceutical toxicologists, industrial hygienists, and regulatory representatives. This collaboration led to the development of the International Society for Pharmaceutical Engineering's (ISPE) Risk-Based Manufacturing of Pharmaceutical Products (Risk-MaPP) Baseline Guide. This guide recommends managing cross-contamination risks using a science-based, risk-based approach, aligning with the International Conference on Harmonisation's Quality Risk Management Guideline (ICH Q9). It emphasizes understanding the toxicity of drugs and employing risk management models to ensure compliance with regulations like 21 CFR 211.42(c).

A pivotal concept introduced by Risk-MaPP is the ADE, which serves as a metric for assessing risk in cleaning validation. The ADE, based on comprehensive clinical and preclinical data, helps quantify the toxicity and exposure levels, guiding the necessary controls and documentation efforts according to the risk levels.

Implementation of ADE has transformed cleaning validation by providing a measurable, science-based criterion for setting acceptance limits and determining the severity of potential contamination. This shift encourages a continuum perspective on drug hazards, moving away from the binary classification of compounds as either highly hazardous or non-hazardous. Moreover, Risk-MaPP and ADE utilization have influenced cleaning validation practices significantly. They enable a clearer, quantitative assessment of risks associated with product cross-contamination in shared facilities, considering factors like mix-up, retention, mechanical transfer, and airborne transfer. This approach allows for tailored risk control strategies, ensuring patient safety while assuring product quality.

Process Capability for Compound Carryover Risk

In cleaning validation, managing the risk of compound carryover in shared facilities is crucial for product safety. The process capability (Cp) scale, rooted in the principles of the International Conference on Harmonisation (ICH) Q9 guideline, serves as an innovative method to assess this risk. It complements the Acceptable Daily Exposure (ADE) approach by evaluating the likelihood of residues exceeding safe limits post-cleaning.

Process capability, or Cp, compares a process's variability to its specification limits. In cleaning validation, the focus shifts to the process capability index (Cpk), particularly the Cpu for upper limits, to determine the effectiveness of cleaning processes against set specifications. The Cpu value, derived from cleaning validation data, quantifies the risk of residue levels surpassing the ADE. Higher Cpu values indicate better process capability and lower risk of cross-contamination.

This risk assessment tool aids in making informed decisions regarding the introduction of new products into existing facilities. By calculating the expected process capability for a new product, manufacturers can predict how well their current cleaning processes will remove residues to safe levels. A lab-scale cleanability test can further validate this.

TOC Analysis Detectability Scale

Selection of analytical methods for cleaning validation is critical in pharmaceutical manufacturing. These methods range from specific to nonspecific, and the choice depends on a science-based, risk-based approach. The detectability scale, informed by detection limits (DLs) and health-based exposure limits (HBELs), aids in determining the suitability of analytical methods like Total Organic Carbon (TOC) for cleaning validation.

Detection limits are pivotal; typically, for HPLC, it's based on the signal-to-noise ratio, while for methods like TOC, it's often derived from the standard deviation of the blank. The lower the DL, the more sensitive the method, making it crucial for method validation in ensuring cleaning effectiveness. TOC has gained acceptance for cleaning validation, particularly for organic residues. The DL for TOC varies significantly across studies, impacting its applicability. A low DL is beneficial as it allows for the detection of minute residues, ensuring the safety and cleanliness of manufacturing equipment.

The Detectability Scale, similar to the previously discussed toxicity and process capability scales, is derived by comparing the DL of an analytical method to the swab limit calculated from the HBEL. A logarithmic scale, such as the Carbon Detection Index (CDI), can quantify this relationship. A CDI greater than zero indicates the method's detection limit is not sufficiently sensitive for the swab limit, questioning its suitability for cleaning validation.

Visual Inspection

Visual inspection is a foundational method in cleaning validation, serving as a primary assessment tool for equipment cleanliness in pharmaceutical manufacturing. It operates alongside analytical methods, scaling from non-specific methods like Total Organic Carbon (TOC) to specific methods for high-risk contaminants. The process hinges on a risk-based approach, with the severity of potential contamination informing the choice of inspection and analytical techniques.

Historically, the standard for visual residue limits (VRLs) has been debated, with figures ranging from 1 to 10μg/cm², depending on the substance, surface, and inspection conditions. This variability underscores the need for clear, science-based criteria for visual inspection as a validation tool. The use of overly conservative limits like the 1/1,000th dose or 10 ppm could unnecessarily restrict the utility of visual inspection. Instead, employing Acceptable Daily Exposure (ADE)-based limits for MSSRs can make visual inspection a feasible and efficient validation method for a wider range of compounds.

Historically, studies on VI like those by Fourman and Mullen, and subsequent works by Jenkins, Vanderwielen, and others have set varying benchmarks for visual residue limits (VDLs), ranging from 0.4 to 4 μg/cm². These studies primarily used the "spotting" method on surrogate materials to determine the lowest visible residue level, which has raised questions about VI's reliability due to lack of standardization and validation. Recent investigations conducted at the Stevens Pharmaceutical Research Center aimed to validate VI as a cleaning validation method. The studies involved trained graduate students examining evenly coated stainless steel coupons under standardized conditions to identify the lowest residue levels detectable by the human eye. These studies found VDLs ranging between 1 and 10 μg/cm², varying with factors such as the residue's nature, inspection methodology, and the analysts' training.

Measuring Cleaning Risk: FMEAs and Dashboards

The International Council on Harmonization (ICH) Q9 provides a framework for risk management, emphasizing the importance of scientific knowledge and patient protection. The FMEA tool assesses potential cleaning failures, considering the severity, occurrence, and detectability of each failure mode. Traditional FMEA methods use subjective ordinal scales, leading to non-meaningful Risk Priority Numbers (RPNs). Instead, applying specific, data-driven scales based on scientific principles, such as HBEL-derived toxicity scores and Cpu-derived occurrence scores, provides a clearer, more objective analysis. Incorporating scientifically justified scales into FMEAs enhances the pharmaceutical industry's approach to risk management. For example, using HBEL-derived toxicity and Cpu-derived occurrence scales helps objectively evaluate the risk associated with specific cleaning failure modes.

Measuring and managing cleaning risk through a structured, data-driven approach ensures effective resource allocation and patient safety. By adopting scientifically based scales for FMEA and implementing comprehensive risk dashboards, pharmaceutical companies can enhance their quality risk management practices, aligning efforts with the actual risk to patients.

Download Checklist

Connect with a Leucine expert and become a facility of future

Book a demo with us now
Schedule demo

Leucine's AI based solutions for

cleaning Validation.

Learn More

Heading 1

Heading 2

Heading 3

Heading 4

Heading 5
Heading 6

Lorem ipsum dolor sit amet, consectetur adipiscing elit, sed do eiusmod tempor incididunt ut labore et dolore magna aliqua. Ut enim ad minim veniam, quis nostrud exercitation ullamco laboris nisi ut aliquip ex ea commodo consequat. Duis aute irure dolor in reprehenderit in voluptate velit esse cillum dolore eu fugiat nulla pariatur.

Block quote

Ordered list

  1. Item 1
  2. Item 2
  3. Item 3

Unordered list

Text link

Bold text

Emphasis

Superscript

Subscript