ICH Q8, Q9, and Q10 - Questions and Answers?

0
Q. Is the minimal approach accepted by regulators?

Ans: Yes. The minimal approach as defined in Q8(R2) (sometime also called “baseline” or “traditional” approach) is the expectation that is to be achieved for a fully acceptable submission. However, the “enhanced” approach as described in ICH Q8(R2) is encouraged.

Q. What is an appropriate approach for process validation using ICH Q8, Q9, and Q10?

Ans: The objectives of process validation are unchanged when using ICH Q8, Q9, and Q10. The main objective of process validation remains that a process design yields a product meeting its predefined quality criteria. ICH Q8, Q9, and Q10 provide a structured way to define product critical quality attributes, design space, the manufacturing process, and the control strategy. This information can be used to identify the type and focus of studies to be performed prior to and on initial commercial production batches. As an alternative to the traditional process validation, continuous process verification can be utilized in process validation protocols for the initial commercial production and for manufacturing process changes for the continual improvement throughout the remainder of the product life-cycle.

Q. How can information from risk management and continuous process verification provide for a robust continual improvement approach under ICH Q8, Q9 and Q10?

Ans: Like the product itself, process validation also has a lifecycle (process design, process qualification and ongoing process verification). A risk assessment conducted prior to initial commercial validation batches can highlight the areas where particular focus and data collection could demonstrate the desired high level of assurance of commercial process robustness. Continual monitoring (e.g., via continuous process verification) can further demonstrate the actual level of assurance of process consistency and provide the basis for continual improvement of the product. Quality Risk Management methodologies of ICH Q9 can be applied throughout the product lifecycle to maintain a state of process control.

Quality by Design (QbD):

Q. Is it always necessary to have a design space (DS) or real-time release (RTR) testing to implement QbD?

Ans:  Under Quality by Design, establishing a design space or using real-time release testing is not necessarily expected.

Q. Is it necessary to study multivariate interactions of all parameters to develop a design space?

Ans:  No, the applicant should justify the choice of material attributes and parameters for multivariate experimentation based on risk assessment and desired operational flexibility.

Q. Can a design space be applicable to a site change?

Ans:  Yes, it is possible to justify a site change using a site independent design space based on a demonstrated understanding of the robustness of the process and an in depth consideration of site specific factors (e.g., equipment, personnel, utilities, manufacturing environment, and equipment). There are region specific regulatory requirements associated with site changes that need to be followed.

Q. Can a design space be developed for single and/or multiple unit operations?

Ans: Yes, it is possible to develop a design space for single unit operations or across a series of unit operations.

Q. Is it possible to develop a design space for existing products?

Ans: Yes, it is possible. Manufacturing data and process knowledge can be used to support a design space for existing products. Relevant information should be utilized from e.g., commercial scale manufacturing, process improvement, corrective and preventive action (CAPA), and development data. For manufacturing operations run under narrow operational ranges in fixed equipment, an expanded region of operation and an understanding of multiparameter interactions may not be achievable from existing manufacturing data alone and additional studies may provide the information to develop a design space. Sufficient knowledge should be demonstrated, and the design space should be supported experimentally to investigate interactions and establish parameter/attribute ranges.

Q. Is there a regulatory expectation to develop a design space for an existing product?

Ans: No, development of design space for existing products is not necessary unless the applicant has a specific need and desires to use a design space as a means to achieve a higher degree of product and process understanding. This may increase manufacturing flexibility and/or robustness.

Q. Can a design space be applicable to formulation?

Ans: Yes, it may be possible to develop formulation (not component but rather composition) design space consisting of the ranges of excipient amount and its physicochemical properties (e.g., particle size distribution, substitution degree of polymer) based on an enhanced knowledge over a wider range of material attributes. The applicant should justify the rationale for establishing the design space with respect to quality attributes such as bioequivalence, stability, manufacturing robustness etc. Formulation adjustment within the design space depending on material attributes does not need a submission in a regulatory post-approval change.

Q. Does a set of proven acceptable ranges alone constitute a design space?

Ans: No, a combination of proven acceptable ranges (PARs) developed from univariate experimentation does not constitute a design space. Proven acceptable ranges from only univariate experimentation may lack an understanding of interactions between the process parameters and/or material attributes. However proven acceptable ranges continue to be acceptable from the regulatory perspective but are not considered a design space.

Q. Should the outer limits of the design space be evaluated during process validation studies at the commercial scale?

Ans: No. There is no need to run the qualification batches at the outer limits of the design space during process validation studies at commercial scale. The design space should be sufficiently explored earlier during development studies.

Q. How is batch release affected by employing real-time release testing?

Ans: Batch release is the final decision to release the product to the market regardless of whether RTR testing or end-product testing is employed. End-product testing involves performance of specific analytical procedures on a defined sample size of the final product after completion of all processing for a given batch of that product. Results of real-time release testing are handled in the same manner as end-product testing results in the batch release decision. Batch release involves an independent review of batch conformance to predefined criteria through review of testing results and manufacturing records together with appropriate good manufacturing practice (GMP) compliance and quality system, regardless of which approach is used.

Q. Does real-time release testing mean elimination of end-product testing?

Ans: Real-time release testing does not necessarily eliminate all end-product testing. For example, an applicant can propose RTR testing for some attributes only or not all. If all critical quality attributes (CQAs) (relevant for real-time release testing) are assured by in-process monitoring of parameters and/or testing of materials, then end-product testing might not be needed for batch release. Some product testing will be expected for certain regulatory processes such as stability studies or regional requirements.

Q. Is a product specification still necessary in the case of RTR testing?

 Ans: Yes, product specifications still need to be established and met, when tested.

Q. When using RTR testing, is there a need for stability test methods?

Ans: Even where RTR testing is applied, a stability monitoring protocol that uses stability indicating methods is required4 for all products regardless of the means of release testing.

Q. What is the relationship between control strategy and RTR testing?

Ans: RTR testing, if utilized, is an element of the control strategy in which tests and/or monitoring can be performed as in-process testing (in-line, on-line, at-line) rather than tested on the end product.

Q. Do traditional sampling approaches apply to RTR testing?

Ans: No, traditional sampling plans for in-process and end-product testing involve a discrete sample size that represents the minimal sampling expectations. Generally, the use of RTR testing will include more extensive on-line/in-line measurement. A scientifically sound sampling approach should be developed, justified, and implemented.

Q. If RTR testing results fail or trending toward failure, can end-product testing be used to release the batch?

Ans: No, in principle the RTR testing results should be routinely used for the batch release decisions and not be substituted by end-product testing. Any failure should be investigated and trending should be followed up appropriately. However, batch release decisions should be made based on the results of the investigations. In the case of failure of the testing equipment, please refer to the previous question. The batch release decision should comply with the content of the marketing authorization and GMP compliance.

Q. What is the relationship between in-process testing and RTR testing?

Ans: In-process testing includes any testing that occurs during the manufacturing process of drug substance and/or finished product. Real-time release testing includes those in-process tests that have a direct impact on the decision for batch release through evaluation of critical quality attributes.

Q. What is the difference between “real time release” and “real-time release testing”?

Ans: The definition of real-time release testing in Q8(R2) is “the ability to evaluate and ensure the acceptable quality of in-process and/or final product based on process data, which typically includes a valid combination of measured material attributes and process controls.” The term real time release in the Q8(R2), step 2 document was revised to “realtime release testing” in the final Q8(R2) Annex to fit the definition more accurately and thus avoid confusion with batch release.

Q. Can surrogate measurement be used for RTR testing?

Ans: Yes, RTR testing can be based on measurement of a surrogate (e.g., process parameter, material attribute) that has been demonstrated to correlate with an in-process or end-product specification.

Q. What is the relationship between RTR testing and parametric release?

Ans: Parametric release is one type of RTR testing. Parametric release is based on process data (e.g., temperature, pressure, time for terminal sterilization, physicochemical indicator) rather than the testing of material and/or a sample for a specific attribute.

Q. What is the difference in a control strategy for products developed using the minimal approach vs. “quality-by-design” approach?

Ans: Control strategies are expected irrespective of the development approach. Control strategy includes different types of control proposed by the applicant to assure product quality such as in-process testing and end-product testing. For products developed following the minimal approach, the control strategy is usually derived empirically and typically relies more on discrete sampling and end-product testing. Under QbD, the control strategy is derived using a systematic science and risk-based approach. Testing, monitoring, or controlling is often shifted earlier into the process and conducted in-line, online, or at-line testing.

Q. Are GMP requirements different for batch release under QbD?

Ans: No, the same GMP requirements apply for batch release under minimal and QbD approaches.

Q. What is the relationship between a design space and a control strategy?

Ans: A control strategy is required for all products. If a design space is developed and approved, the control strategy provides the mechanism to ensure that the manufacturing process is maintained within the boundaries described by the design space.

Q. What approaches can be taken in the event of on-line/in-line/at-line testing or monitoring equipment breakdown?

Ans: The control strategy provided in the application should include a proposal for use of alternative testing or monitoring approaches in cases of equipment failure. The alternative approach could involve use of end-product testing or other options, while maintaining an acceptable level of quality. Testing or monitoring equipment breakdown should be managed in the context of a deviation under the quality system and can be covered by GMP inspection.

Q. Are product specifications different for minimal versus QbD approaches?

Ans: In principle no, product specifications are the same for minimal and QbD approaches. For a QbD approach, the control strategy can facilitate achieving the end product specifications via real time release testing approaches. Product must meet specification, when tested.

Pharmaceutical Quality System

Q. What are the benefits of implementing a pharmaceutical quality system (PQS) (in accordance with ICH Q10)?

Ans:  The benefits are:

Facilitated robustness of the manufacturing process, through facilitation of continual improvement through science and risk-based post-approval change processesConsistency in the global pharmaceutical environment across regionsEnable transparency of systems, processes, and organizational and management responsibilityClearer understanding of the application of a quality system throughout product lifecycleFurther reducing risk of product failure and incidence of complaints and recalls, thereby providing greater assurance of pharmaceutical product consistency and availability (supply) to the patientBetter process performanceOpportunity to increase understanding between industry and regulators and more optimal use of industry and regulatory resources; enhance manufacturer’s and regulators’ confidence in product qualityIncreased compliance with GMPs, which builds confidence in the regulators and may result in shorter inspections

Q. How does a company demonstrate implementation of PQS in accordance with ICH Q10?

Ans: When implemented, a company will demonstrate the use of an effective PQS through its documentation (e.g., policies, standards), its processes, its training/qualification, its management, its continual improvement efforts, and its performance against pre-defined key performance indicators. A mechanism should be established to demonstrate at a site how the PQS operates across the product lifecycle, in an easily understandable way for management, staff, and regulatory inspectors, e.g., a quality manual, documentation, flowcharts, procedures. Companies can implement a program in which the PQS is routinely audited in-house (i.e., internal audit program) to ensure that the system is functioning at a high level.

Q. Is it necessary to describe the PQS in a regulatory submission?

Ans: No, however relevant elements of the PQS (such as quality monitoring system, change control, and deviation management) can be referenced as part of the control strategy as supporting information.

Q. Will there be certification that the PQS is in accordance with ICH Q10?

Ans: No. There will not be a specific ICH Q10 certification program.

Q. How should the implementation of the design space be evaluated during inspection of the manufacturing site?

Ans: Inspection should verify/assess that manufacturing operations are appropriately carried out within the design space. The inspector in collaboration with the assessor, where appropriate, should also verify successful manufacturing operations under the design space and that movement within the design space is managed within the company’s change management system

Q. What should be done if manufacturing operations run inadvertently outside of the design space?

Ans: This should be handled as a deviation under GMP. For example, unplanned “oneoff” excursions occurring as a result of unexpected events, such as operator error or equipment failure, would be investigated, documented, and dealt with as a deviation in the usual way. The results of the investigation could contribute to the process knowledge, preventive actions, and continual improvement of the product.

Q. What information and documentation of the development studies should be available at a manufacturing site?

Ans: Pharmaceutical development information (e.g., supporting information on design space, chemo-metric model, risk management) is available at the development site. Pharmaceutical development information that is useful to ensure the understanding of the basis for the manufacturing process and control strategy, including the rationale for selection of critical process parameters and critical quality attributes, should be available at the manufacturing site. Scientific collaboration and knowledge sharing between pharmaceutical development and manufacturing is essential to ensure the successful transfer to production.

Q. Can process parameters be adjusted throughout the product lifecycle?

Ans: Process parameters are studied and selected during pharmaceutical development and monitored during commercial manufacturing. Knowledge gained could be utilized for adjustment of the parameters as part of continual improvement of the process throughout the lifecycle of the drug product.

Q. How will product-related inspections differ in an ICH Q8, Q9 and Q10 environment?

Ans: In the case of product-related inspection (in particular, preauthorization) depending on the complexity of the product and/or process, greater collaboration between inspectors and assessors could be helpful (for example, for the assessment of development data). The inspection would normally occur at the proposed commercial manufacturing site, and there is likely to be greater focus on enhanced process understanding and understanding relationships, e.g., critical quality attributes (CQAs), critical process parameters (CPPs). The inspection might also focus on the application and implementation of quality risk management principles, as supported by the pharmaceutical quality system (PQS).

Q. How will system-related inspections differ in an ICH Q8, Q9, and Q10 environment?

Ans: The inspection process will remain similar. However, upon the implementation of ICH Q8, Q9, and Q10, inspections will have greater focus on (but not only focus on) how the PQS facilitates the use of e.g., quality risk management methods, implementation of design space, and change management.

Q. How is control strategy approved in the application and evaluated during inspection?

Ans: Elements of control strategy submitted in the application will be reviewed and approved by the regulatory agency. However, additional elements are subject to inspection.

Knowledge Management

Q. How has the implementation of ICH Q8, Q9, and Q10 changed the significance and use of knowledge management?

Ans: Q10 defines knowledge management as: “Systematic approach to acquiring, analyzing, storing, and disseminating information related to products, manufacturing processes and components.” Knowledge management is not a system; it enables the implementation of the concepts described in ICH Q8, Q9 and Q10.

Knowledge management is not a new concept. It is always important regardless of the development approach. Q10 highlights knowledge management because it is expected that more complex information generated by appropriate approaches (e.g., QbD, process analytical technology (PAT), real-time data generation, and control monitoring systems) should be better captured, managed, and shared during product life-cycle.

In conjunction with quality risk management, knowledge management can facilitate the use of concepts such as prior knowledge (including from other similar products), development of design space, control strategy, technology transfer, and continual improvement across the product life cycle.

Q. Does Q10 suggest an ideal way to manage knowledge?

Ans: No. Q10 provides a framework and does not prescribe how to implement knowledge management. Each company decides how to manage knowledge, including the depth and extent of information assessment based on its specific needs.

Q. What are potential sources of information for knowledge management?

Ans: Some examples of knowledge sources are:

Prior knowledge based on experience obtained from similar processes (internal knowledge, industry scientific and technical publications) and published information (external knowledge: literature and peer-reviewed publications)Pharmaceutical development studiesMechanism of actionStructure/function relationshipsTechnology transfer activitiesProcess validation studiesManufacturing experience, e.g., — Internal and vendor audits — Raw material testing dataInnovationContinual improvementChange management activitiesStability reportsProduct quality reviews/annual product reviewsComplaint reportsAdverse event reports (patient safety)Deviation reports, recall InformationTechnical investigations and/or CAPA reportsSuppliers and contractorsProduct history and /or manufacturing historyOngoing manufacturing processes information.

Information from the above can be sourced and shared across a site or company, between companies and suppliers/contractors, products, and across different disciplines (e.g., development, manufacturing, engineering, quality units).

Q. Is a specific dedicated, computerized information management system required for the implementation of knowledge management with respect to ICH Q8, Q9, and Q10?

Ans: No, but such computerized information management systems can be invaluable in capturing, managing, assessing, and sharing complex data and information.

Q. Will regulatory agencies expect to see a formal knowledge management approach during inspections?

Ans: No. There is no added regulatory requirement for a formal knowledge management system. However, it is expected that knowledge from different processes and systems will be appropriately utilized. Note: “formal” means: it is a structured approach using a recognized methodology or information technology (IT) tool, executing and documenting something in a transparent and detailed manner.

Post a Comment

0 Comments
* Please Don't Spam Here. All the Comments are Reviewed by Admin.
Post a Comment (0)

#buttons=(Accept !) #days=(20)

Our website uses cookies to enhance your experience. Learn More
Accept !
To Top