Guide To Validation - Drugs and Supporting Activities: June 29, 2021
Guide To Validation - Drugs and Supporting Activities: June 29, 2021
supporting activities
Health Canada
Address Locator 0900C2, Ottawa, ON K1A 0K9
Tel.: 613-957-2991
Toll free: 1-866-225-0709
Fax: 613-941-5366
TTY: 1-800-465-7735
Email: [email protected]
© Her Majesty the Queen in Right of Canada, as represented by the Minister of Health, 2021
This publication may be reproduced for personal or internal use only without permission provided
the source is fully acknowledged.
Cat.: H139-7/2021E-PDF
ISBN: 978-0-660-33546-9
Pub.: 200033
Table of contents
About this document............................................................................................................................. 5
1. Purpose .............................................................................................................................. 5
2. Scope .................................................................................................................................. 5
3. Introduction ....................................................................................................................... 6
Guidance ................................................................................................................................................ 8
4. Principles ............................................................................................................................ 8
5. General validation requirements ...................................................................................... 9
6. Lifecycle approach to process validation........................................................................ 10
6.1 Phase 1 – Process design...................................................................................... 11
6.2 Phase 2 – Process performance qualification (PPQ) ........................................ 14
6.2.1 Protocol ....................................................................................................... 15
6.2.2 Number of process qualification batches.................................................. 16
6.2.3 Challenge plans .......................................................................................... 17
6.2.4 Distinct manufacturing step requirements ............................................... 18
6.2.5 Sampling and testing plan requirements .................................................. 18
6.2.6 Acceptance Criteria .................................................................................... 19
6.2.7 Final report .................................................................................................. 20
6.2.8 Continuous process verification................................................................. 21
6.3 Phase 3 – Ongoing process verification ............................................................. 23
7. Change review .................................................................................................................. 24
8. Guidance for validation or qualification of supporting activities .................................. 24
8.1 Facility, equipment and utility qualification ...................................................... 25
8.2 Analytical Method Validation ............................................................................. 27
8.3 Computer System Validation ............................................................................ 27
8.4 Packaging Validation ........................................................................................... 28
8.5 Cleaning Validation ........................................................................................... 29
Appendices .......................................................................................................................................... 30
Appendix A – Special concerns for solid dose products ..................................................... 30
Uniformity .............................................................................................................. 30
Dissolution .............................................................................................................. 32
Appendix B – Glossary .......................................................................................................... 34
Acronyms..................................................................................................................... 34
Terms ........................................................................................................................... 35
The following are the three types of icons used in this document, and the way they are intended
to be used.
It describes how to properly qualify and validate drug manufacturing processes, facilities,
equipment, utilities and analytical methods. The application of this document will vary
depending on the nature of your operations.
This document will help you to comply with Part C, Division 2 of the Food and Drug Regulations,
(the Regulations).
It has been revised with a view to harmonize with current guidance from other regulatory
agencies and organizations such as the United States Food and Drug Administration (US FDA),
European Medicines Agency (EMA), Pharmaceutical Inspection Cooperation Scheme (PIC/S),
International Council for Harmonisation (ICH) and World Health Organization (WHO).
2. Scope
These guidelines apply to the following types of drugs:
pharmaceutical
radiopharmaceutical
biological
veterinary
This document outlines general validation requirements. Information for more specific topics
(e.g. sterile products) can be found in these Health Canada guidelines:
Additional guidance on validation related activities for certain lower risk veterinary drugs and
Category IV monograph drugs can be found in these guidance documents;
Annex 7 to Good manufacturing practices guide for drug products - Selected non-
prescription drugs (GUI-0066)
Health Canada has adopted the Guideline ICH Q7A: Good Manufacturing
Practice for Active Pharmaceutical Ingredients. This document may be used as
supplementary optional guidance for validation activities for active
pharmaceutical ingredients (APIs).
3. Introduction
These guidelines interpret the requirements for good manufacturing practices (GMP) in Part C,
Division 2 of the Regulations. They were developed by Health Canada in consultation with
stakeholders.
Guidance documents like this one are meant to help industry and health care professionals
understand how to comply with regulations. They also provide guidance to Health Canada staff,
so that the rules are enforced in a fair, consistent and effective way across Canada.
Health Canada inspects establishments to assess their compliance with the Food and Drugs Act
(the Act) and associated regulations. When we conduct an inspection, we will use this document
as a guide in assessing your compliance with GMP requirements.
These guidelines are not the only way GMP regulations can be interpreted, and are not intended
to cover every possible case. Other ways of complying with GMP regulations will be considered
with proper scientific justification. Also, as new technologies emerge, different approaches may
be called for.
Process validation – The collection and evaluation of data, from the process
design stage through commercial production, which establishes scientific
evidence that a process is capable of consistently delivering quality products.
Process Validation: General Principles and Practices, US FDA.
Validation is not a single study—it represents the cumulative knowledge gained during product
development and manufacture. Process validation should incorporate a lifecycle approach,
including:
Ensure you apply quality risk management (QRM) principles when developing your validation
program and individual validation studies. Use these principles to determine the scope and
extent of study requirements. Document and justify all decisions made.
You may wish to consider the following ICH guidance documents for additional information as
you develop your approach to process validation:
2. For large and complex projects you may want to generate a project specific Validation
Master Plan to ensure:
such projects are appropriately planned and controlled
the inter-relationship between various documents and studies for such projects is
clearly defined
4. Ensure personnel performing qualification and validation studies are trained and
qualified when needed, and that they follow procedures.
5. Ensure all documents generated during qualification and validation are approved and
authorized by appropriate personnel and by the quality department.
8. Record any failure to meet predefined acceptance criteria as a deviation within your
quality system. Any implications to validation studies should be discussed in the final
report.
9. Any significant changes to the protocol during execution will need to be fully explained
and justified. Such changes should be handled in accordance with a company’s quality
management system.
10. Incorporate checks into qualification and validation work, in order to ensure the
integrity of data obtained.
2. The QTPP is normally defined at the beginning of Phase 1 as it represents the objective
of the development process. It should be referenced throughout the product lifecycle
and may need to be revisited as product and process understanding improve.
Examples of sources of variation and potential control strategies are provided in Table 1.
Starting the PPQ study represents the end of development and beginning of commercial
manufacture.
1. Conduct a readiness assessment to verify that you are ready to start PPQ batch
manufacture. Items to verify in a readiness assessment include the following:
adequate manufacturing understanding/controls have been established from the
process design phase
manufacturing instructions are sufficiently detailed to ensure batches are
manufactured in a reproducible manner
2. Manufacture PPQ batches at commercial scale, according to GMP requirements and the
manufacturing process/parameter ranges established during Phase 1.
3. Consider whether qualification batches should be enrolled into the ongoing stability
program. This will depend on the extent of stability monitoring conducted during the
design phase and the extent of process variability (such as maximum hold times,
different raw material suppliers or lots) incorporated into the qualification batches.
6.2.1 Protocol
Conduct all PPQ studies according to a preapproved protocol. This protocol document
should include:
a brief description of the process with reference to the master batch record, and
other relevant supporting documents
validation team membership including roles and responsibilities
summaries of CQAs to be investigated CPPs with associated limits and any other
non-critical attributes and parameters which will be monitored
intermediate and finished product sampling and testing requirements; sampling
plans and acceptance criteria should be justified
in-process testing controls and acceptance criteria
proposed number of PPQ batches to be made (see section 6.2.2 Number of
process qualification batches)
a description of any challenge plans to be included (see section 6.2.3
Challenge plans)
a summary of the evaluation of the site’s readiness for the qualification study as
described above
2. Based on the results of your risk assessment, a grouping strategy to the process
qualification study may also be acceptable. For example, a bracketing approach might
evaluate the extremes of a single variable, such as multiple strengths of a tablet using
common granulation. Such a strategy could involve additional batches of the highest and
lowest strength tablets and reduced evaluation of all intermediate strengths.
3. It is generally expected that you will conduct PPQ studies in a prospective manner. The
studies should be completed, evaluated and deemed to be successful before
commercial distribution of any batches.
a. Concurrent validation may be acceptable in exceptional circumstances, for example,
where there is a strong benefit–risk ratio for the patient. Concurrent validation may
be considered appropriate when involving drugs for which there may be a limited
demand (e.g. orphan drugs, or veterinary drugs with limited uses) and drugs with
short expiry dates (e.g. radiopharmaceuticals, including positron emission
tomography drugs). Concurrent validation may also be appropriate, in coordination
with Health Canada, to alleviate shortages of medically necessary drugs.
Other items to consider challenging during the process qualification study execution include
the following (if practical and relevant).
Different API and critical excipient lots. This can account for lot to lot differences.
Note: APIs and critical excipients sourced from different vendors are a potential
source of variability and may require additional evaluation and/or testing.
Interchangeable equipment. Qualification studies may be used in demonstrating that
the use of interchangeable equipment will not impact quality. In such cases, there
should be documented justification as to why equipment may be considered
interchangeable and why separate process qualification studies are not required.
Different personnel. Attempts should be made to ensure that qualification batches
are produced by different personnel and on different shifts.
Sampling and testing plans should focus on the quality attributes most likely to
demonstrate intra and inter batch variability.
Sampling locations should be chosen to include those that represent the highest
potential for variability. This is of special concern for processes that produce unit
dose products, such as tablets or capsules or those with the potential of material
segregation or non-uniformity, such as filling suspensions into tubes.
Sampling and testing plans should be tailored to the type of material being produced
at that stage. For example, if a unit manufacturing step is producing a true solution,
sampling may be reduced compared to making a potentially less uniform powder or
suspension mixture.
2. Ensure your Process Performance Qualification (PPQ) protocol clearly outlines and
justifies all sampling and testing requirements (including test methods).
Highly variable inter or intra batch testing results, or results barely meeting
specifications, may indicate an inadequately controlled process. This may
mean the manufacturing process is not adequately understood and requires
additional development work.
Acceptance criteria should consider the expectation that drugs be formulated
to provide 100% of label claim as outlined in Good manufacturing practices for
drug products (GUI-0001). Additional requirements on limits of variability can
be found in Sections C.01.061 and C.01.062 of the Food and Drug Regulations.
Your report should also include a conclusion that clearly states that the manufacturing
process is considered to be one of the following:
It is also generally not acceptable to change protocol acceptance criteria in order to justify
the successful completion of a qualification study, unless it can be shown that the change
does not impact patient safety or product quality. In these cases, it would be important to
discuss why the original criteria were set as they were.
An option you may use when planning for process performance qualification is continuous
process verification, as introduced in ICH Q8(R2): Pharmaceutical Development. It is a
science and risk based approach to ensuring a manufacturing process operates within the
required parameters and consistently produces material meeting specifications. Continuous
process verification can make it easier to evaluate the manufacturing process, by providing
more information about process variability and control.
Ensure the rationale to use continuous process verification is clearly stated and justified
within development reports, submission documents and qualification/monitoring protocols.
You should determine and justify the number of batches to be assessed using continuous
process verification using risk based principles. Clearly define the point at which a product
will be considered to be qualified and the basis on which that decision will be made before
release of batches.
Many legacy products will not have been developed and qualified in the
manner outlined in this guidance. Applying the principles of Phase 3 to these
products will facilitate both ensuring the process is in an adequate state of
control and that process understanding increases as additional batches are
manufactured.
2. Depending on the risk to the patient, level of variability in CQAs and confidence in the
process, it may be desirable to monitor post qualification batches at an enhanced level.
Testing requirements may then be re-evaluated once sufficient data has been obtained
to determine estimates of process variability. Such information should be used in the
determination of on-going batch monitoring, sampling and testing requirements.
4. The extent and frequency of review is relative to the established level of process
knowledge and confidence (i.e. newer products may require a more frequent review).
7. Changes in results and result trending should be investigated and actioned, where
warranted. A shift in results may be indicative of a source of variability that was not
previously identified, an unintended change to the process, or a need to revise control
strategies.
7. Change review
A change control system is a key GMP requirement. Use this system to evaluate changes that
can potentially impact the validation status of a process/product (e.g. changes to process
components, equipment, manufacturing parameters and test methods). From a validation
perspective, implementing such changes could result in requirements to update product risk
assessments/control strategies and/or requalify the manufacturing process.
Note: A series of minor changes over time may represent a major change requiring additional
qualification.
c. Ensuring that clear information is available to install, operate and maintain the
facility/utility/equipment. Examples of this include manuals, drawings and
standard operating procedures.
d. Ensuring that requirements are met for proper ongoing operation of the
unit/facility (e.g. calibration, preventative maintenance and training).
e. Ensuring operation in accordance with the manner in which the system was
designed including process and procedural requirements. The qualification
effort should clearly demonstrate that the equipment/systems are operating in a
manner that meets requirements as outlined in the user and functional
requirement specifications. Operating ranges and/or worst case conditions
should be challenged.
1. Design Qualification
2. Installation Qualification
3. Operational Qualification
4. Performance Qualification
3. Qualification studies should demonstrate that the systems are suitable for intended use.
Manufacturer supplied qualification documents may not fully meet these requirements
as the company using the equipment will be more knowledgeable of the intended use of
the system.
4. Tests conducted during prequalification testing (e.g. factory acceptance testing (FAT))
may not need to be repeated during the qualification study execution if justified.
a) studies are generally required for direct contact systems (e.g. heating/ventilation/air
conditioning (HVAC) or water systems) or high-risk non-contact systems
b) ensure that the studies account for potential impact of seasonal variation
7. Maintenance events and equipment changes should be reviewed to ensure they don’t
impact equipment functionality or product quality.
3. The ongoing operation and configuration of the system should be monitored and
reviewed at a defined frequency according to a procedure e.g. reviewing significant
events/alarm logs, or user access. Use QRM principles to determine the extent and
nature of effort to be required at each stage. Risk for a computerized system comes
from two predominant aspects.
The intended use of the system.
4. Qualification stage testing should demonstrate that systems are appropriate for their
intended use. This requires verification that the system has been installed, set-up and
operates in accordance with pre-defined requirements. Ensure that system
configuration and parameters are clearly documented.
5. Ensure that data control requirements are included in the computer system validation
studies. The necessary system and procedural controls must be established to ensure
data is maintained and protected from inappropriate modification or deletion.
2. In some cases, packaging operations are essential in ensuring that a product is uniform
(e.g. liquid, powder and semi-solid products). Process performance qualification studies
should fully assess packaging functions for such products.
3. Packaging validation needs also to confirm container integrity, and that essential
labelling information such as imprinting of lot and expiry date, correct fill volume or
count, leaflet insertion etc. are able to be performed correctly.
4. Sampling plans for package integrity and other critical packaging attributes should be
justified based on known sources of variation in the process.
Solid dose manufacturing processes should be developed in accordance with the life-cycle
approach. Proper attention should be paid to ensuring that parameters impacting the product
quality are understood and appropriately controlled.
Validation efforts and process performance qualification studies should generally focus on
ensuring that product is uniform and meets dissolution testing requirements. This is in addition
to other specific critical quality attributes that may be of concern for the product in question.
Uniformity
Ensuring that a manufacturing process produces a uniform blend is a key consideration for a
process validation study. Non-uniform blends can be caused by inadequate mixing and/or
component segregation.
1. The powder-blend characteristics that can affect uniformity should be understood and
controlled. Examples of potential control strategies for ensuring robust powder blend
characteristics include:
The assessment of blend uniformity normally includes two steps: a blend uniformity assessment
and a content uniformity assessment. Sampling from bins may also be required after the
discharge step has been completed.
1. Blend uniformity assessment: Obtain samples from the blender at the completion of the
mixing process. Evaluate results to ensure the blend is well mixed and that no significant
difference exists between locations in the blender that could adversely affect product
quality. Criteria to be evaluated include:
a. Blend uniformity sample size: Blend uniformity samples should be 1 to 3 times the
minimum dosage size being evaluated. You may use larger samples with justification.
b. Sampling locations: Sampling locations should enable full assessment of the blender
and include potential problem areas (e.g. dead spots). Use statistical tools to
determine the number of replicates to be obtained and the number of sampling
locations within the blender
c. Sampling method: Sample thieves remain widely used and can—in many cases—
present acceptable results. However, they do have drawbacks because they cause
bed disturbance so sampling errors are possible. Evaluate the reliability of sampling
as part of the analytical method development.
d. Testing requirements: Determine the number of replicate samples to be analyzed
from each location. Testing more than one replicate sample has the benefit of
helping to determine whether you have location to location variation or within
location variation. Regardless of how many samples are taken, each sample should
be tested separately. Combining doses as a composite for a single analytical
determination is not appropriate.
e. Acceptance criteria: Pre-establish acceptance criteria and base it on a statistical
analysis.
If you decide to implement PAT or other process-monitoring and control techniques for a
powder blend homogeneity assessment, your decision should be supported with
appropriate data and rationale, using both a science and risk-based approach.
2. Content uniformity assessment: Content uniformity involves obtaining samples from the
tablet press (or dosing machine). The process performance qualification protocol should
define:
sampling times, ensuring that worst case locations such as first and last
accepted tablets are included
number of tablets or capsules to be sampled at each location
acceptance criteria
It is expected that no significant differences would exist between in-process locations
that could affect finished product quality. Between- and within-location variability is a
critical component of the finished product quality, so should be evaluated. All such
evaluations should be statistically based and scientifically justified. Examples of
acceptable statistical methods are outlined in ASTM E2709 and ASTM E2810.
Dissolution
It is important to understand the factors that will control the overall dissolution rate before
beginning the process performance qualification study. For example:
a. lubrication mix time can impact dissolution rates, for example for immediate release
tablets
b. tablet hardness can impact the tablet dissolution rate. Conduct hardness studies
(where tablet hardness and dissolution rates are compared/correlated) as part of the
development process. Establish hardness specification and guideline tablet press
settings (e.g. feeder type/speed, compression speed, pre-compression force and
main compression force) before starting the process qualification effort.
The impact of any formulation and manufacturing parameter variability on dissolution rates
should be understood during Phase 1 and appropriate controls should be established.
You may be required to demonstrate that the dissolution profile of commercial lots is similar to
that of the bio lot or submission profile. Any approach used to demonstrate similarity should be
statistically justified. One such example is the use of a similarity factor (f2), in which an f2 value
of between 50 and 100 is considered to be indicative of a similar dissolution profile.
These definitions explain how terms are used in this document (unless
otherwise specified). Definitions cited directly from other documents are noted
in brackets at the end of the definition.
If there is a conflict with a definition in the Food and Drugs Act or Food and
Drug Regulations, the definition in the Act/Regulations prevails.
Change control – A written procedure that describes the action to be taken if a change is
proposed (a) to facilities, materials, equipment, and/or processes used in the fabrication,
packaging, and testing of drugs or (b) that may affect the operation of a quality or support
system. (GUI-0001)
Concurrent Validation - Validation carried out in exceptional circumstances, justified on the basis of
significant patient benefit, where the validation protocol is executed concurrently with commercialisation
of the validation batches. (PIC/S Annex 15 – PE 009-14 (Annexes))
Control strategy – A planned set of controls, derived from current product and process
understanding that ensures 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. (ICH Q10)
Critical process parameter – A process parameter whose variability has an impact on a critical
quality attribute and therefore should be monitored or controlled to ensure the process
produces the desired quality. (ICH Q8)
Design Qualification – The documented verification that the proposed design of the facilities,
systems and equipment is suitable for the intended purpose. (PIC/S Annex 15 – PE 009-14
(Annexes))
Drug – Includes any substance or mixture of substances manufactured, sold or represented for
use in:
Installation Qualification – The documented verification that the facilities, systems and
equipment, as installed or modified, comply with the approved design and the manufacturer’s
recommendations. (PIC/S Annex 15 – PE 009-14 (Annexes))
Lifecycle – All phases in the life of a product from the initial development through marketing
until the product’s discontinuation. (ICH Q8)
Operational Qualification – The documented verification that the facilities, systems and
equipment, as installed or modified, perform as intended throughout the anticipated operating
ranges. (PIC/S Annex 15 – PE 009-14 (Annexes))
Performance Qualification – The documented verification that systems and equipment can
perform effectively and reproducibly based on the approved process method and product
specification. (PIC/S Annex 15 – PE 009-14 (Annexes))
Process analytical technology (PAT) – A system for designing, analyzing and controlling
manufacturing through timely measurements (i.e. during processing) of critical quality and
performance attributes of raw and in-process materials and processes, with the goal of ensuring
final product quality. (ICH Q8)
Prospective Validation – Validation carried out before routine production of products intended for sale.
(PIC/S Annex 15 – PE 009-14 (Annexes))
Quality target product profile (QTPP) – A prospective summary of the quality characteristics of a
drug product that ideally will be achieved to ensure the desired quality, taking into account
safety and efficacy of the drug product . (ICH Q8)
Risk – The combination of the probability of occurrence of harm and the severity of that harm.
(ICH Q9)
Risk management – The systematic application of quality management policies, procedures and
practices to the tasks of assessing, controlling, communicating and reviewing risk. (ICH Q9)
State of control – A condition in which the set of controls consistently provides assurance of
acceptable process performance and product quality. (ICH Q10
Trend – A statistical term referring to the direction or rate of change of a variable(s). (ICH Q9)
User requirements specifications – The set of owner, user, and engineering requirements
necessary and sufficient to create a feasible design meeting the intended purpose of the system.
(PIC/S Annex 15 – PE 009-14 (Annexes))
Validation – A documented program that provides a high degree of assurance that a specific
process, method, or system will consistently produce a result meeting pre-determined
acceptance criteria. (ICH Q7)
Worst-case – A condition or set of conditions encompassing upper and lower processing limits
and circumstances, within standard operating procedures, which pose the greatest chance of
product or process failure when compared to ideal conditions. Such conditions do not
necessarily induce product or process failure.
Guidance Document: Quality (Chemistry and Manufacturing) Guidance: New Drug Submissions
(NDSs) and Abbreviated New Drug Submissions (ANDSs)
www.canada.ca/en/health-canada/services/drugs-health-products/drug-products/applications-
submissions/guidance-documents/chemical-entity-products-quality/guidance-document-quality-
chemistry-manufacturing-guidance-new-drug-submissions-ndss-abbreviated-new-drug-
submissions.html
US FDA guidance
FDA Guidance for Industry - Process Validation, General Principles and Practices
https://www.fda.gov/regulatory-information/search-fda-guidance-documents/process-
validation-general-principles-and-practices
FDA Guidance for Industry: SUPAC-IR: Immediate-Release Solid Oral Dosage Forms: Scale-Up and
Post-Approval Changes: Chemistry, Manufacturing and Controls, In Vitro Dissolution Testing, and
In Vivo Bioequivalence Documentation (November 1995)
www.fda.gov/regulatory-information/search-fda-guidance-documents/supac-ir-immediate-
release-solid-oral-dosage-forms-scale-and-post-approval-changes-chemistry
FDA Guidance for Industry: SUPAC-MR: Modified Release Solid Oral Dosage Forms Scale-Up and
Postapproval Changes: Chemistry, Manufacturing, and Controls; In Vitro Dissolution Testing and
In Vivo Bioequivalence Documentation (October 1997)
www.fda.gov/regulatory-information/search-fda-guidance-documents/supac-mr-modified-
release-solid-oral-dosage-forms-scale-and-postapproval-changes-chemistry
ICH guidance
ICH Q7: Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients
https://ich.org/page/quality-guidelines
ICH Q11: Development and Manufacture of Drug Substances (Chemical Entities and
Biotechnological/Biological Entities)
www.canada.ca/en/health-canada/services/drugs-health-products/drug-products/applications-
submissions/guidance-documents/international-conference-
harmonisation/quality/development-manufacture-drug-substances-chemical-entities-
biotechnological-biological-entities-topic.html
Other guidance
ASTM E2500 - 13 Standard Guide for Specification, Design, and Verification of Pharmaceutical
and Biopharmaceutical Manufacturing Systems and Equipment
www.astm.org/Standards/E2500.htm
ASTM International: ASTM E2709 Standard Practice for Demonstrating Capability to Comply with
an Acceptance Procedure
www.astm.org/Standards/E2709.htm
ASTM International: ASTM E2810: Standard Practice for Demonstrating Capability to Comply
with the Test for Uniformity of Dosage Units
www.astm.org/Standards/E2810.htm
Glossary and Tables for Statistical Quality Control, Fourth Edition, American Society for Quality
Control, Statistics Division, ASQ Quality Press, 2004.
Parenteral Drug Association Technical Report 60 Process Validation: A Lifecycle Approach (2013)
www.pda.org/publications/pda-publications/pda-technical-reports
Pharmaceutical Inspection Cooperation Scheme Annex 15: Qualification and Validation (PE 009-
14)
www.picscheme.org/en/publications