ICH Q8/Q9/Q10 Questions and Answers

ICH Q8/Q9/Q10

1.1 For General Clarification

Date of Approval Questions Answers
1.0 June
2009
Is the minimal approach accepted by regulators? Yes. The minimal approach as defined in Q8(R2) (sometimes 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 (Ref. Q8(R2) Appendix 1).
2.0 Oct.
2009
What is an appropriate approach for process validation using ICH
Q8, Q9, and Q10?
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 [see definition in ICH Q8(R2) glossary] can be utilized in process validation protocols for the initial commercial production and for manufacturing process changes for the continual
3.0 Oct.
2009
How can information from risk management and continuous
process verification provides for a robust continual improvement
approach under ICH Q8, Q9, and Q10?
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 are needed to 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.

2. QUALITY BY DESIGN TOPICS

Date of Approval Questions Answers
01 April
2009
Is it always necessary to have a Design Space (DS) or Real-Time Release (RTR) testing to implement QbD? Under Quality by Design, establishing a design space or using
real-time release testing is not necessarily expected [ICH Q8(R2), Step 4].

2.1 Design Space

Date of Approval Questions Answers
01 April
2009
Is it necessary to study multivariate interactions of all parameters to develop a design space? No, the applicant will need to justify the choice of material
attributes and parameters for multivariate experimentation
based on risk assessment and desired operational flexibility.
02 April
2009
Can a design space be applicable to scale-up? Yes, when appropriately justified. An example of scale-independent design space is provided in the EFPIA Mock P2 document [EFPIA Mock P2  submission on “Explain”: Chris Potter, Rafael Beerbohm, Alastair Coupe, Fritz Erni, Gerd Fischer, Staffan Folestad, Gordon Muirhead, Stephan Roenninger, Alistair Swanson, A guide to EFPIA’s “Mock P.2” Document, Pharm. Tech. (Europe), 18, December 2006, 39-44].
This example may not reflect the full regulatory requirements
for a scale-up.
03 April
2009
Can a design space be applicable to a site change? 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 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.
04 April
2009
Can a design space be developed for single and/or multiple
unit operations?
Yes, it is possible to develop a design space for a single unit
operations or across a series of unit operations [see Q8(R2)
Section 2.4.3].
05 April
2009
Is it possible to develop a design space for existing products? 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, CAPA, and development data.
For manufacturing operations run under narrow operational ranges in fixed equipment, an expanded region of operation and an understanding of multi-parameter interactions may not be achievable from existing manufacturing data alone and additional studies may be needed 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.
06 April
2009
Is there a regulatory expectation to develop a design space for
an existing product?
No, the 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.
07 June
2009
Can a design space be applicable to formulation? Yes, it may be possible to develop formulation (not the 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.
08 June
2009
Does a set of proven acceptable ranges alone constitute a
design space?
No, a combination of proven acceptable ranges (PARs) developed from univariate experimentation does not constitute a design space [see Q8(R2), Section 2.4.5.]. 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 [see ICH Q8(R2) Section 2.4.5].
The applicant may elect to use proven acceptable ranges or
design space for different aspects of the manufacturing process.
09 Nov.2010 Should the outer limits of the Design Space be evaluated during process validation studies at the commercial scale? No, there is no need to run the qualification batches at the outer limits of the design space during process validation studies at a commercial scale.

The design space must be sufficiently explored earlier during development studies (for scale-up see also Chapter 2.1 Design Space Question 2; for life cycle approach see Chapter 1.1 for general clarification Question 3).

2.2 Real-Time Release Testing

Date of Approval Questions Answers
01 April
2009
How is batch release affected by employing real-time release
testing?
The 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 the 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 a review of testing results and manufacturing records together with appropriate GMP compliance and quality system, regardless of which approach is used.

02 April
2009
Does real-time release testing mean the elimination of end
product testing?
Real-time release testing does not necessarily eliminate all end-product testing.

For example, an applicant may propose RTR testing for some attributes only or not all. If all CQAs (relevant for real-time release testing) is 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.

03 April
2009
Is a product specification still necessary in the case of RTR testing? Yes, product specifications [see ICH Q6A and Q6B] still need
to be established and met, when tested
04 April
2009
When using RTR testing, is there a need for a stability test
methods?
Even where RTR testing is applied, a stability monitoring
protocol that uses stability-indicating methods is required for
all products regardless of the means of release testing. [see
ICH Q1A and ICH Q5C].
05 April
2009
What is the relationship between Control Strategy and RTR testing? 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.
06 April
2009
Do traditional sampling approaches apply to RTR testing? No, traditionally sampling plans for in-process and endproduct 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.

07 April
2009
If RTR testing results fail or trending toward failure, can endproduct testing be used to release the batch? 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 will need to be made based on the results of the investigations.

The batch release decision needs to comply with the content of the marketing authorization and GMP compliance.

08 June
2009
What is the relationship between in-process testing and RTR
testing?
In-process testing includes any testing that occurs during the
the manufacturing process of drug substance and/or finished
product.Real-time release testing includes those in-process tests that directly impact the decision for batch release through the evaluation of Critical Quality Attributes.
09 June
2009
What is the difference between ‘real-time release’ and ‘real
time release testing’?
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 ‘Real-time release testing’ in the final Q8(R2)
Part II document to fit the definition more accurately and thus
avoid confusion with batch release.
10 June
2009
Can a surrogate measurement be used for RTR testing? 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
11 Oct.
2009
What is the relationship between RTR testing and Parametric
Release?
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.

2.3 Control Strategy
Refer to the definition of control strategy provided in the ICH Q10 glossary: Q10 Control Strategy definition: ‘a planned set of controls, derived from current product and process understanding that assures process performance and product quality.

The controls can include parameters and attributes related to drug substance and drug product materials and components, facility and equipment operating conditions, in-process controls, finished product specifications, and the associated methods and frequency of monitoring and control.’

Date of Approval Questions Answers
01 April
2009
What is the difference in a control strategy for products
developed using the minimal approach vs. ‘quality-by-design’
approach?
Control strategies are expected irrespective of the development approach. Control strategy includes different types of control proposed by the applicant to assure product quality (Section 3.2.1 ICH Q10), 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, on-line or at-line testing.
02 April
2009
Are GMP requirements different for batch release under QbD? No, the same GMP requirements apply for batch release
under minimal and QbD approaches.
03 April
2009
What is the relationship between a Design Space and a Control Strategy? A control strategy is required for all products. If a Design Space is developed and approved, the Control Strategy [see ICH Q8(R2), Part II, Section 4] provides the mechanism to ensure that the manufacturing process is maintained within the boundaries described by the Design Space.
04 June
2009
What approaches can be taken in the event of on-line/inline/at-line testing or monitoring equipment breakdown? 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 needs to be managed in the context of a deviation under the Quality System and can be covered by GMP inspection.

05 Oct.
2009
Are product specifications different for minimal versus QbD
approaches?
In principle no, the same product specifications are needed for minimal and QbD approaches. For a QbD approach, the control strategy may allow achieving the end product specifications via real-time release testing approaches [see ICH Q8(R2), Appendix 1]. Product must meet specifications when tested.

3. PHARMACEUTICAL QUALITY SYSTEM

Date of Approval Questions Answers
01 April
2009
What are the benefits of implementing a Pharmaceutical
Quality System (in accordance with ICH Q10)?
The benefits are:
• Facilitated robustness of the manufacturing process, through facilitation of continual improvement through science and risk-based post-approval change processes;
• Consistency in the global pharmaceutical environment across regions;
• Enable transparency of systems, processes, organizational
and management responsibility;
• Clearer understanding of the application of a Quality System throughout product lifecycle;
• Further reducing risk of product failure and incidence of
complaints and recalls thereby providing greater assurance of pharmaceutical product consistency and availability (supply) to the patient;
• Better process performance;
• Opportunity to increase understanding between industry and regulators and more optimal use of industry and regulatory resources. Enhance manufacturer’s and regulators’ confidence in product quality;
• Increased compliance with GMPs, which builds confidence
in the regulators and may result in shorter inspections.
02 April
2009
How does a company demonstrate the implementation of PQS in accordance with ICH Q10? 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 [see ICH Q10 glossary on ‘Performance indicator’].
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.
03 April
2009
Is it necessary to describe the PQS in a regulatory
submission?
No, however relevant elements of the PQS, such as quality
monitoring system, change control, and deviation management may be referenced as part of the control strategy as supporting information.
04 April
2009
Will, there be certification that the PQS is in accordance with
ICH Q10?
No. There will not be a specific ICH Q10 certification programme.
05 April
2009
How should the implementation of the design space be
evaluated during inspection of the manufacturing site?
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 [see ICH Q10,
Section 3.2. Table III].
06 April
2009
What should be done if manufacturing operations run
inadvertently outside of the Design Space?
This should be handled as a deviation under GMP. For example unplanned ‘one-off‘ 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 may contribute to the process knowledge, preventive actions and continual improvement of the product.
07 June
2009
What information and documentation of the development
studies should be available at a manufacturing site?
Pharmaceutical development information (e.g., supporting
information on design space, chemometric model, risk management) is available at the development site.
Pharmaceutical development information which 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.
08 June
2009
Can process parameters be adjusted throughout the product
lifecycle?
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 (see ICH Q10, Section 3.).

4. ICH NEW QUALITY GUIDELINES’ IMPACT ON GMP INSPECTION PRACTICES

Date of Approval Questions Answers
01 April
2009
How will product-related inspections differ in an ICH Q8, Q9
and Q10 environment?
In the case of product-related inspection (in particular preauthorisation) depending on the complexity of the product
and/or process, there could be a need for greater collaboration
between inspectors and assessors 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 Attribute (CQAs), Critical Process
Parameters (CPPs).

It will also extend into the application and implementation of quality risk management principles,as supported by the Pharmaceutical Quality System (PQS).

02 April
2009
How will system-related inspections differ in an ICH Q8, Q9
and Q10 environment?
The inspection process will remain similar. However upon
the implementation of ICH Q8, Q9 and Q10, inspections will
have greater focus (but not only) on how the PQS facilitates
the use of e.g., Quality Risk Management methods, implementation of design space and change management [see ICH Q10].
03 Oct.
2009
How is control strategy approved in the application and
evaluated during inspection?
Elements of control strategy submitted in the application will
be reviewed and approved by the regulatory agency.
However, additional elements are subject to inspection (as described in Q10).

5. KNOWLEDGE MANAGEMENT

Date of Approval Questions Answers
01 April
2009
How has the implementation of ICH Q8, Q9, and Q10 changed
the significance and use of knowledge management?
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, PAT, real-time data generation and
control monitoring systems) will need to 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.

02 April
2009
Does Q10 suggest an ideal way to manage knowledge? 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 their specific needs.
03 April
2009
What are potential sources of information for Knowledge
Management?
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 studies;
• Mechanism of action;
• Structure/function relationships;
• Technology transfer activities;
• Process validation studies;
• Manufacturing experience e.g.: – Internal and Vendor audits;
– Raw material testing data;
• Innovation;
• Continual improvement;
• Change management activities;
• Stability reports;
• Product Quality Reviews/Annual Product Reviews;
• Complaint Reports;
• Adverse event reports (Patient safety);
• Deviation Reports, Recall Information;
• Technical investigations and/or CAPA reports;
• Suppliers and Contractors;
• Product history and /or manufacturing history;
Ongoing manufacturing processes information (e.g., trends).
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).
04 April
2009
Is a specific dedicated computerized information management
system required for the implementation of knowledge
management with respect to ICH Q8, Q9, and Q10?
No, but such computerized information management systems
can be invaluable in capturing, managing, assessing and sharing complex data and information.
05 June
2009
Will regulatory agencies expect to see a formal knowledge
management approach during inspections?
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 utilised.
Note: ‘formal’ means: it is a structured approach using a recognized methodology or (IT-) tool, executing and documenting something in a transparent and detailed manner.

6. SOFTWARE SOLUTIONS

Date of Approval Questions Answers
01 April
2009
With the rapid growth of the new science and risk-based quality paradigm coupled with the IWG efforts to facilitate
globally consistent implementation of Q8, Q9, and Q10, a number of commercial vendors are now offering  products that are being marketed as ‘ICH compliant solutions’ or ICH Q8, 9 & 10 Implementation software, etc.Is it necessary for a
pharmaceutical firm to purchase these products to achieve a successful implementation of these ICH guidelines within
their companies?
No. The ICH Implementation Working Group has not endorsed any commercial products and does not intend to do so. ICH is not a regulatory agency with reviewing authority and thus does not have a role in determining or defining ‘ICH compliance’ for any commercial products.

While there will likely be a continuous proliferation of new products targeting the implementation of these ICH guidelines, firms will need
to carry out their own evaluation of these products relative to
their business needs.

Reference: Quality Implementation Working Group on Q8, Q9, and Q10 Questions & Answers (R4)

Technology Transfer in pharmaceutical manufacturing (WHO)

About Pharmaceutical Guidanace

Ms. Abha Maurya is the Author and founder of pharmaceutical guidance, he is a pharmaceutical Professional from India having more than 18 years of rich experience in pharmaceutical field. During his career, he work in quality assurance department with multinational company’s i.e Zydus Cadila Ltd, Unichem Laboratories Ltd, Indoco remedies Ltd, Panacea Biotec Ltd, Nectar life Science Ltd. During his experience, he face may regulatory Audit i.e. USFDA, MHRA, ANVISA, MCC, TGA, EU –GMP, WHO –Geneva, ISO 9001-2008 and many ROW Regularities Audit i.e.Uganda,Kenya, Tanzania, Zimbabwe. He is currently leading a regulatory pharmaceutical company as a head Quality. You can join him by Email, Facebook, Google+, Twitter and YouTube

Check Also

Temperature mapping of storage areas: Ensuring Reliable Environmental Control and Compliance

Temperature mapping of storage areas: Ensuring Reliable Environmental Control and Compliance In various industries, maintaining …