GMP Inspection East Africa
GMP Inspection East Africa
GMP Inspection East Africa
STEPS DATE
Development of draft harmonized technical 30th July 2012 to 28th June 2013
documents for Good Manufacturing Practices
(GMP) for the East African Community
Medicines Regulatory Harmonization Initiative
Draft harmonized technical documents 3rd September 2013
approved by the Steering Committee
Release of draft harmonized technical 9th September 2013
documents for Public Consultation
End of National Consultation 28th February 2014
(deadline for comments)
Incorporation of national stakeholders inputs 13th to 17th January 2014
by EAC Secretariat in collaboration with EAC
Partner States
Release of revised technical documents for 20th January 2014
regional and international consultation
Draft technical documents reviewed and 12th March 2014
adopted by EAC Technical Working Group on
Medicines and Food Safety
Draft technical documents adopted by the 14th March 2014
18th EAC Sectoral Committee on Health
Draft technical documents finalized by 1st to 4th April 2014
EAC Secretariat in collaboration with
EAC Partner States
Final technical documents approved by the 17th April 2014
9th EAC Sectoral Council on Health
Date for coming into effect 17th April 2014
i
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
ii
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
iii
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
iv
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
FOREWORD
v
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
PREFACE
vi
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
The EAC Treaty Article 118, Chapter 21 on regional cooperation on health provides for harmonization
of medicines regulation among the EAC Partner States. Therefore, this Compendium will be
implemented by National Medicines Regulatory Authorities (NMRAs) in accordance with the
relevant policies, laws, cooperation, guidelines, manuals and procedures existing at national and
regional level.
The 9th EAC Sectoral Council of Ministers of Health has officially approved the operationalization
of this Compendium among the East African Community Partner States’ National Medicines
Regulatory Authorities (NMRAs).
Signed on this ………. Day of ………..2014 by the Honourable Ministers responsible for East
African Community Affairs as here-in:
vii
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
TABLE OF CONTENTS
AUTHORS/CONTRIBUTORS
FOREWORD
PREFACE
PART ONE: INSPECTION MANUAL FOR GOOD MANUFACTURING PRACTICE
1. ABBREVIATIONS AND ACRONYMS
2. GLOSSARY
3. INTRODUCTION
4. SCOPE
5. TYPES OF INSPECTIONS
5.1. Routine inspection
5.2. Concise inspection
5.3. Follow-up inspection
5.4. Special inspection
6. Frequency of inspections
6.1. LOCAL PHARMACEUTICAL MANUFACTURERS
6.2. FOREIGN PHARMACEUTICAL MANUFACTURERS
7. Planning for GMP Inspections
8. Preparation for GMP inspection
9. JOINT GMP INSPECTION
10. QUALIFICATION OF GMP INSPECTOR
11. CODE OF ETHICS AND CONDUCT FOR GMP INSPECTORS
12. DECLARATION OF CONFLICT OF INTERESTS
13. CONDUCTING GMP INSPECTION
14. SAMPLE COLLECTION AND TESTING
15. INSPECTION REPORT
16. Classification of GMP Inspection Observations
17. RECOMMENDED REGULATORY ACTION(S)
18. PRODUCT RECALL
19. Appeal
20. REFERENCES
21. REVISION HISTORY
22. LIST OF COMPLEMENTARY DOCUMENTS
PART TWO: GUIDELINES ON GOOD MANUFACTURING PRACTICE FOR MEDICINAL PRODUCTS FOR
USE IN EAC
ABBREVIATIONS AND ACRONYMS
GLOSSARY
1. INTRODUCTION
2. SCOPE
CHAPTER 1: QUALITY MANAGEMENT
Principle
Quality assurance
Good manufacturing practices (GMP)
Quality control
Product Quality Review
Quality Risk Management
Sanitation and hygiene
CHAPTER 2: PERSONNEL
Principle
General
Key personnel
Training
01
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Personal hygiene
CHAPTER 3: PREMISES
Principle
General
Production Area
Storage areas
Quality control Areas
Ancillary areas
CHAPTER 4: EQUIPMENT
Principle
General
CHAPTER 5: DOCUMENTATION
Principle
General
Labels
Documents required
Specifications and testing procedures
Specifications for starting and packaging materials
Specifications for intermediate and bulk products
Specifications for finished products
Master formulae and Processing instructions
Packaging instructions
Batch processing records
Batch packaging records
Procedures (SOPs) and records
Receipts
Sampling
Testing
Others
CHAPTER 6: GOOD PRACTICES IN PRODUCTION
Principle
General
Prevention of cross-contamination and bacterial contamination in production
Validation
Starting materials
Processing operations: intermediate and bulk products
Packaging materials
Packaging operations
Finished products
Rejected, Recovered Reprocessed and Returned materials
Waste materials
Miscellaneous
CHAPTER 7: GOOD PRACTICES IN QUALITY CONTROL
Principle
General
Documentation
Sampling
Control of starting materials and intermediate, bulk products
Test requirements
In-process control
Finished products
Batch record review
Stability studies
02
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
03
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Design qualification
Installation qualification
Operational qualification
Performance qualification
Qualification of established (in-use) facilities, systems and equipment
Process validation
Cleaning validation
Change control
Revalidation
Glossary
04
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
ANNEX 16: MODEL PROCEDURE FOR PREPARING AND REVIEWING GMP INSPECTION REPORT
5.0 PROCEDURES
6.0 RECORD KEEPING
8.0 REVISION HISTORY
9.0 ATTACHMENTS
ANNEX 17: MODEL PROCEDURE FOR RISK CLASSICATION OF GMP DEFECIENCIES
ANNEX 18: MODEL PROCEDURE FOR FOLLOW UP ON NON COMPLIANCES AFTER GMP INSPECTION
8. DOCUMENT REVISION HISTORY AND AUTHORIZATION
ANNEX 19: MODEL PROCEDURE FOR HANDLING PRODUCT RECALL
PART THREE: GUIDELINES FOR PREPARATION OF SITE MASTER FILE FOR PHARMACEUTICAL
MANUFACTURING FACILITIES
2. GLOSSARY
4. SCOPE
5. LAY OUT OF THE SITE MASTER FILE:
6. CONTENT OF SITE MASTER FILE
6.1 GENERAL INFORMATION
6.1.1 Contact information on the manufacturer
6.1.2 Authorized pharmaceutical manufacturing activities of the site
6.1.3 Any other manufacturing activities carried out on the site.
6.2 QUALITY MANAGEMENT
6.2.1 The quality management system of the manufacturer
6.2.1.1 Brief description of the quality management systems run by the company and reference to the
standards used.
6.2.1.2 Responsibilities related to the maintaining of the quality system including senior
management
6.2.1.3 Information on activities for which the site is accredited and certified, including dates and contents of
accreditations, and names of accrediting bodies.
6.2.2 Release procedure of finished products
6.3 MANAGEMENT OF SUPPLIERS AND CONTRACTORS
6.4 PRODUCT QUALITY REVIEWS
6.4.2 Brief description of methodologies used.
6.5 PERSONNEL
6.5.2 Qualifications, experience, and responsibilities of technical personnel should be included
as Annex 5.
6.5.3 Outline of arrangements for basic and in-service training and how records are maintained.
6.5.5 Personnel hygiene requirements, including clothing.
6.6 PREMISES AND EQUIPMENT
6.6.1 Premises
6.6.1.2 Nature of construction and finishes
6.6.1.6 Brief description of planned preventive maintenance programmes for premises and of the
recording system.
6.6.1.7 Brief description of other relevant utilities, such as steam, compressed air, nitrogen, etc. Schematic
diagrams should be added in annex 9.
6.6.1.8 Availability of written specifications and procedures for cleaning manufacturing areas
6.6.2 Equipment
6.6.2.2 Brief description of the procedures used for cleaning major equipment.
6.6.2.3 Brief description of planned preventive maintenance programmes for equipment and of the
recording system.
05
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
6.6.2.4 Brief description of the company’s Qualification and calibration policy, including the recording
system. Reference should be made to the Validation master plan.
6.7 DOCUMENTATION
6.7.1 Arrangements for the preparation, revision, distribution and archiving of necessary documentation
for manufacture should be stated.
6.7.2 Brief description of the validation master plan
6.7.3 Brief description of the change control procedure
9.7.4 Any other documentation related to product quality that is not mentioned elsewhere (e.g.,
microbiological controls on air and water).
6.8 PRODUCTION
6.8.1 Type of products
6.8.2 Process validation
6.8.3 Material management and warehousing
6.8.3.2 Arrangements for the handling of rejected materials and products.
6.9 QUALITY CONTROL
6.10 DISTRIBUTION, COMPLAINTS, PRODUCTS DEFECT AND RECALL
6.11 SELF-INSPECTION
6.12 SHELF LIFE / STABILITY DETERMINATION PROGRAM
6.12.1 General policy for the determination of the shelf-life and stability of products manufactured at
the site.
7. REFERENCES:
8. REVISION HISTORY
06
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
PART ONE:
07
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
08
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
09
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
10
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
11
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
12
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
13
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
14
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
15
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
16
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
17
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
18
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
1. PURPOSE 5. PROCEDURES
The purpose of this SOPis to ensure that GMP 5.1 Selection of Companies to
Inspectorates follow a standardized procedure be Inspected
when planning for routine GMP inspections.
This should assist with ensuring a consistent 5.1.1 Selection of facilities to be inspected
approach in conducting inspections. is considered an initial and crucial
step in planning for an inspection.
2. SCOPE
5.1.2 The concerned department shall
The scope of thisSOP applies for planning undertake the selection and ensure
GMP inspections of manufacturers of FPPs that:
and of APIs applied within the EAC Partner
States, including joint GMP inspection. 5.1.2.1 The local technical representative or
manufacturer should have filled in
the details of the inspection on a
3. RESPONSIBILITY form with details of actual site to
be inspected, lines and contact
3.1 Head of NMRAs
details of the responsible persons.
3.2 Head GMP department
5.1.2.2 The local technical representative/
manufacturer will also have to pay
3.3 LeadInspectors
the prescribed fee for the GMP
inspection
3.4 GMPinspectors
5.1.2.3 Selection shall ideally be based on
3.6 Procurement department
first application first inspected basis.
3.7 Human Resource Unit
5.1.3 Without unduly contravening the
provision 5.1.5 above, facilities may
4. DISTRIBUTION LIST be selected based on;
19
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.2.1.2 Anticipated duration of inspection 5.3.4 Verify the objective of the inspection
based on plant size, number of that is to be carried out.
blocks/production lines and activities
5.3.5 Determine what the scope and
5.2.1.3 A combination of all, or some of the depth of the inspection will be to
factors for selection as appropriate. enable to prepare properly for
the inspection.
5.2.2 Scheduling to be carried out within
a period of six months and allocate 5.3.6 Scrutinize the relevant documents
tentative dates and will be checked as indicated in SOP for Preparing
and reviewed regularly within the for inspection.
specified period.
20
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
21
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Name________________________________________________________________
Physical Address_______________________________________________________
Country____________________Telephone__________________________________
Fax________________________E-mail_____________________________________
Name of site___________________________________________________________
Country_____________________Tel_______________________________________
Fax____________________E-mail:________________________________________
Tel: __________________________________Fax:____________________________
E-mail:_______________________________________________________________
Tel: __________________________________________________________________
5. TYPE OF MEDICINES
6. REGISTRATION OF PRODUCTS
………………………………………………………………………….....................................
22
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
8 DECLARATION
Commitment from manufacturers to welcome inspectors for inspection any time
I hereby certify that the above information is correct and apply for Good Manufacturing Practice
inspection of the above-named site(s).
Notes:
1. Please submit a copy of the Site Master File (not more than 25 pages) together with
this application (refer to Guideline on preparation of a Site Master File)
2. This application must be submitted together with the appropriate fee to the Head
of NMRA
23
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
24
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
The scope of this SOP applies for preparation 5.2.2 Verify the objective of the inspection
of GMP inspections of manufacturers of FPPs that is to be carried out.
and of APIs applied within the EAC Partner
States, including joint GMP inspection. 5.2.3 Verify whether the inspection will
cover the entire factory or just part
of it.
3. RESPONSIBILITY
5.2.4 Determine what the scope and
3.5 Head of NMRAs depth of the inspection will be to
prepare properly for the inspection.
3.6 Head GMP department
5.2.5 Scrutinize the product dossiers for
3.7 Lead Inspectors the products manufactured in the
respective manufacturing site.
3.8 GMP inspectors
5.2.6 Decide what products will be
4. DISTRIBUTION LIST covered during the inspection.
25
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.2.9 Confirm the amount of time that will 5.2.11 If desired, prepare a checklist of
be required to carry out the points to be verified during the
inspection and plan the date when inspection. Prepare notes for
the inspection will take place. verification in the aide memoire
specific to site to be verified during
A routine inspection for one site the inspection.
can be performed over a period of
at least two to five working days. 5.2.12 Prepare a Tentative Inspection
The length of an inspection is Plan (Annex I) which can be used
determined by a number of as a template that can be modified.
factors, including the type of Indicate in the programme which
inspection to be performed, the sections or departments will be
number of inspectors, the size of the inspected, and when.
company and the purpose of the 5.2.13 Distribute the Plan to the team
inspection or visit. Annex: criteria for members for comments and after
deciding the duration. finalization, to the company
approximately 2 weeks before
5.2.9 Study the Site Master File and the inspection.
make notes to be followed up during
the inspection (e.g. available 5.4 Re-Inspection
equipment, SOPs, records).Study
the layout and design of the facility 5.4.1 During preparation for GMP
to get a better understanding of the inspection, the CAPA and/or
flow of material, personnel and previous GMP inspection reports
processes in the facility. Study some should be reviewed.
of the systems the organization has
in place (e.g. HVAC and Water). 6. REFERENCE
5.2.10 If a current SMF does not exist, WHO PQP SOP 402.1;
request for an updated copy from PREPARING FOR AN INSPECTION
the company.
7. REVISION HISTORY
26
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
27
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Notes:
• The inspection will start at approx. 8.30am and finish at approximately 5pm each day
• At the end of each day if need be a brief meeting will be held to review the findings and
discuss the plan for the next day
28
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.1 The Inspectors should identify 5.8 Maintain notes during the inspection
themselves at the entrance of the and keep this record for filing on
site before entering the site the company files after completion
of the inspection.
29
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
8. REVISION HISTORY
30
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Annex III: Record of documents requested iv. Explain that the closing meeting
during an inspection (optional) allows providing a summary of the
observations made-and the intention
OPENING MEETING is not to list each observation
Opening the meeting should at least include (Note: You should have
but not limited to the following; discussed the observations made
at the end of each day or at some
i. Introduction of the inspectors point during the inspection. No
surprises in the inspection
ii. Ask the company to introduce report!)
the people present and to make a
brief presentation v. Provide a summary of issues of
concern under different areas such
iii. Explain how the inspection is to be as:
conducted
• Quality Assurance
iv. Scope of the inspection • Documentation
• Personnel
v. Inspection plan • Premises
• Equipment
vi. Discuss Inspection Time Table • Materials
• Cleaning/sanitation/Hygiene
vii. How the feedback will be given e.g. • Production
end of each day • Quality control
• Validation
viii. Which are the standards that will be • Utilities
applied(EAC- GMP)
vi. Mention ,if relevant, whether there
are any critical or major deficiencies
CLOSING MEETING
vii. Ask if the company needs
i. Thank the company for their clarification on any point
cooperation
31
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
32
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
33
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
34
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
35
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
36
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.3.1 The GMP NMRAs Joint Inspection 5.3.4 In case of a request, the decision
coordinators shall share information made is forwarded to the
on manufacturing sites that have manufacturer. If the contacted or
expressed interest in participating in concerned agencies agree to
a joint EAC inspection. conduct a joint inspection, the
applicant should receive an
5.3.2 The GMP Inspection coordinators electronic mail message
shall set up a communication acknowledging such agreement.
platform (e.g teleconference, email, The message should state the
skype call) to agree that a joint EAC primary contact person at each
inspection is warranted, determine agency for the specific inspection.
timelines and identify the inspection See Clause 2.17 of document
team and lead inspector. number EAC/TF-MED/GMP/FD/
FOM/N6R0
5.3.3 Based on the information/response
received and the common areas
of interest, the concerned authorities
37
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.3.5 In the other cases, the letter sent in 5.5.1.4 At least one meeting (VC, Skype,
advance of the inspection will inform Google hangout, etc) should
officially of the joint inspection. be organized by the lead inspector
for discussing the preparation
5.4 PLANNING FOR THE JOINT Works of the joint inspection, which
INSPECTION includes verification of the
scope, the inspection plan,
5.4.1 The joint inspection planned should composition of the team and
be integrated in the planning of allocation of responsibilities.
each NMRAs.
5.6 CONDUCTING THE JOINT
5.4.2 The plan of jointed inspection INSPECTION
is updated by a chair of the
coordination committee. 5.6.1 The joint inspection should be
conducted following the “EAC SOP
5.5 PREPARATION FOR THE JOINT for Conducting GMP Inspection”
INSPECTION with the following particulars;
38
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.8.1 Each participating authority 6.4 EAC SOP for preparing and
is responsible for any follow-up reviewing GMP inspection reports
actions according to their own
regulations and procedures. 6.5 EAC SOP for follow up on
non-compliances.
39
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
7. REVISION HISTORY
40
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
1.2.3 Extending mutual confidence, and 2.7 All the EAC Partner States NMRAs
the opportunity to optimize remain committed to meeting
resources and avoid unnecessary domestic process and review goals
duplication of inspections. and timeframes. The joint inspection
shall not adversely affect either
2.0 PRINCIPLES NMRA’s ability to meet its formal
domestic performance expectations.
2.1 Concerned EAC authorities shall
be responsible for ensuring that 2.8 All NMRAs commit to be cognizant
appropriate confidentiality of the other’s formal domestic
arrangements are in place to allow performance expectations and
them to conduct joint EAC NMRAs to exhibit as much flexibility
GMP inspections. as possible in scheduling meetings
and accommodating the different
2.2 All participating companies shall timeframes for the inspection.
be expected to permit unrestricted
and comprehensive exchange of 2.9 All NMRAs within the EAC will
information between authorities. make these “General Principles”
public on their websites in order to
2.3 The normal rules for national make the joint GMP inspections
coordination of inspections will apply programme, procedures and
goals more transparent and to
41
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
help answer many questions about 2.12 If an applicant’s request for a joint
the joint inspections programme that inspection within the EAC is
my exist in the general public. declined, this will in no way affect
the processing of any submission
2.10 Exchange of information on FPP/ which will proceed with each agency
APIs, manufacturing sites, individually, following each agency’s
inspection reports and other normal procedures.
detailed information shall be
subject to specific confidentiality 2.12.1 Each authority will follow the SOP
agreements with concerned for GMP inspections in the EAC.
authorities and companies
concerned, as necessary. All 2.12.2 The agencies will assure that
the NMRAs in the EAC will maintain records are maintained to facilitate
the confidentiality of all such the monitoring and evaluation of
information. the program and for the assessment
of the benefits and detriments of
2.11 A request for a joint inspection is the program.
no guarantee that a joint inspection
will be performed. For a variety of
reasons, including scheduling
conflicts and available resources
at any specific time, any of the
NMRAs contacted for joint
inspections may decline to
participate. In which case, the
remaining NMRAs contacted may
proceed with the joint inspection.
42
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Name______________________________________________________________________
Physical Address_____________________________________________________________
Country____________________Telephone________________________________________
Fax________________________E-mail__________________________________________
Name of site________________________________________________________________
Country_____________________Tel_____________________________________________
Fax____________________E-mail:______________________________________________
43
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Tel:__________________________________Fax:___________________________________
E-mail:______________________________________________________________________
Tel:________________________________________________________________________
6. TYPE OF MEDICINES
7. REGISTRATION OF PRODUCTS
…………………………………………………………………………..................................................
8. LINES TO BE INSPECTED
44
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Notes:
1. Please submit a copy of the Site Master File (not more than 25 pages) together with
this application (refer to Guideline on preparation of a Site Master File)
2. This application must be submitted together with the appropriate fee to the Head
of NMRAs
45
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
46
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
6.6 Assign and mark each sample 6.11 Samples should be collected in
collected with a number from their original package. In case a
the respective Sample Receipt product which is not on its
Form. The following sample original package will need to
numbering system is be sampled, then Inspector should
recommended - date, month, year, make sure that all information
country, region, name of required should be recorded on the
manufacturing facility Sampling Form
6.7 Inspector should use appropriate 6.12 Inspector should complete a Sample
type of materials for sealing Receipt FormEAC/TF-MED/GMP/
samples or for embargoing FD/FOM/N8R0 for each
of products sample collected.
47
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
7.0 ATTACHMENTS
7.1 Sample Receipt Form (SRF)- EAC/TF-MED/GMP/FD/FOM/N8R0
Physical Address:…………………………………………………………………………........
…………………………………………………………………………………………………....
48
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
1. PURPOSE 5. PROCEDURES
The purpose of this procedure is to guide 5.1 PREPARING A GMP INSPECTION
GMP inspectors and Peer review committees REPORT
on how to prepare and review an inspection
report respectively. 5.1.1 The GMP inspection report shall be
in Times Roman 12; line spacing 1.5
2. SCOPE and in the format attached herewith.
It shall comprise the sections
The scope of this SOP applies to the indicated in form EAC/TF-MED/
preparation and review of the GMP GMP/FD/FOM/N9R0. The
inspections reports of manufacturers of FPPs inspection reports should be written
and of APIs applied within the EAC Partner in 3rd person passive style and I
States, including joint GMP inspection. past tense.
49
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
inspection exit report, and any other review technical team within seven
document as may be necessary (7) days before discussion of report.
5.1.6 The GMP inspection report should 5.2 REVIEWS AND APPROVAL OF
be balanced, unbiased and factual. THE GMP INSPECTION REPORT
It shall be detailed enough to enable
GMP peer review technical 5.2.1 The GMP peer review technical
team make an informed opinion of team, procedurally selected, shall
the recommendation made by read the reports, assess their text,
the inspectors. The observations context and facts and agree or
should be referenced to the relevant disagree with the recommendations
applicable clause in the EAC of the GMP audit team. The head
GMP guideline. An observation that of inspection to convene a meeting
can’t be reasonably referenced to discuss and approve the
should not be listed as inspection report.
an observation.
5.2.2 The signed report must then
5.1.7 Where more than one observation be scanned and emailed by the
relate to the same basic quality Head of the NMRA to the applicant
system failure, they should be and/ or contact person in the
grouped and listed as a single manufacturing facility within
observation, under a heading that forty five (45) days from the last
reflects the basic system failure. day of the inspection. As necessary,
the local technical representative
5.1.8 The observations identified shall be may have a copy of the report.
classified according to standard
procedure number EAC/TF-MED/ 5.2.3 In case the facility complies
GMP/FD/SOP/N7R0. with current EAC GMP
requirements, the respective
5.1.9 The inspection team should prepare NMRAs will issue Certificate of
finalize, sign and submit the final Compliance as per form EAC/TF-
GMP Inspection report within MED/GMP/FD/FOM/N12R0.
fourteen (14) working days after the
date of return to office. 6.0 RECORD KEEPING
5.1.10 All inspection team members should 6.1 The GMP department/Unit/Division
sign the inspection report. shall keep both an electronic copy
and hard copy of the reports in PDF
5.1.11 The Lead Inspector shall submit or any other protected format in a
the inspection reports to the Head designated folder and prepare the
GMP Inspectorate /division certificates or cover letters of
non-compliance or approval as per
5.1.12 The Head of GMP Inspectorate shall the GMP Inspection
circulate copies of the report to
every member of the GMP peer
50
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
9.0 ATTACHMENTS
9.1 Template for FPP/API Inspection Report: EAC/TF-MED/GMP/FD/N6RO
51
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
COVER PAGE
The page should bear the name of the Regulatory Authority
1.0 INTRODUCTION
1.1 General information
Name of Manufacturer:
Physical address:
Unit number:
Production Block:
Contact person and email address (full Postal address, Tel numbers, faxes and email address)
Manufacturing license:
GMP certificate:
All the abbreviations used in the report should be included in this section.
Name of the GMP Inspectors involved. The team leader should be specified.
1.6 Names, titles and qualifications of key personnel of the facility that participated
Other people met during the inspection should also be mentioned. Key personnel
52
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
PART 2: SUMMARY
2.1 General information about the company and site
Where applicable, State major changes made on the facility, equipment, products, and senior
personnel since the previous inspection should be stated.
Brief description of the objective/reason for the Inspection. State the type of inspection and
whether it was product related inspection.
What you planned to do, areas inspected.
QUALITY ASSURANCE
GOOD MANUFACTURING PRACTICES (GMPs) FOR PHARMACEUTICAL PRODUCTS
COMPLAINTS
PRODUCT RECALLS
53
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
PERSONNEL
TRAINING
PERSONAL HYGIENE
PREMISES
EQUIPMENT
MATERIALS
DOCUMENTATION
Conclusion
Make conclusion of your assessment of the acceptability of the facility’s GMP status for the range
of products manufactured.
Based on the areas inspected, the people met and the documents reviewed, and considering
the findings of the inspection, including the observations listed in the inspection report, were
considered to be operating at an acceptable level of compliance with EAC GMP guidelines.
54
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
However, the observations (non-compliances with guidelines) listed below must be addressed in a
timely manner. The manufacturer is expected to respond to all observations and for each include
a description of the corrective action implemented or planned to be implemented, and the date of
completion or target date for completion. The acceptability of corrective actions will be assessed
through evaluation of the response to each observation and will be followed up during the next
inspection.
or
Based on the areas inspected, the people met and the documents reviewed, and considering the
findings of the inspection, including the observations listed in the Inspection Report, a decision
on the compliance of with WHO GMP will be made after the manufacturer’s response to the
observations has been assessed.
The manufacturer is expected to respond to all observations and for each include a description
of the corrective action implemented or planned to be implemented, and the date of completion
or target date for completion. In addition, for observations classified as “major”, supporting
documentation should be submitted with the response as objective evidence of completion of
corrective actions. The acceptability of corrective actions will be assessed through evaluation of
the response to each observation and will be followed up during the next inspection. If considered
necessary, an on-site follow up inspection may be conducted to verify effective implementation of
corrective actions.
or
Based on the areas inspected, the people met and the documents reviewed, and considering
the findings of the inspection, including the observations listed in the Inspection report,
was considered to be operating at an unacceptable level of compliance with EACGMP
guidelines.
Another inspection will be required to verify the implementation of corrective actions before the
manufacturer’s level of GMP compliance can be reconsidered.
5.0 APPENDIXES
Attach the necessary Appendixes to the report, chronologically numbered in Roman numerals.
The following Appendixes should appear:
Manufacturing certificate
GMP certificate of the local regulatory authority
55
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Date: ………………………..
56
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Attached please find the inspection report. Attention should be given to the noted deficiencies and
corrective action taken on the major and minor defiencies observed in due course. Please send
the corrective action schedule and documentation of what has been done.
Please note:
1. That each product must be registered with NMRA (mention the name) before export to
………............. (Country name)
2. NMRA (mention name) can inspect your facility at any time as long as your product is
on the……………. (country name) market.
3. Approval for GMP compliance is valid for three years from the date of inspection.
The company has to apply for re-inspection six months prior to the expiry date if interested in
maintaining products on the NMRA (mention name of NMRA) register.
…………………………………………………...
Head of NMRA (name & Signature)
57
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
On the basis of the inspection carried out on [date] ................. we certify that the site indicated
on this certificate complies with Good Manufacturing Practices for dosage forms, categories and
activities listed in Table 1.
Table 1:
Dosage form (s) Categories of medicines Activity i.e. Packaging, manufacture of finished
pharmaceutical products
The responsibility for the quality of the individual batches of the pharmaceutical products
manufactured lies with the manufacturer/applicant.
This certificate remains valid until [date] .............. It becomes invalid if the dosage forms, activities
and/or categories certified herewith are changed or if the site is no longer considered to be in
compliance with GMP.
.................................... ................................
Signature Stamp and date:
Note: 1.This certificate certifies the status of the site listed in point 1 of the certificate
2. This certificate shall remain valid for a period of 3 years from the date of issue, but
can be revoked at any time if there is evidence that the facility is no longer complies
with then current EAC GMP regulations.
58
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
An observation describing a situation that is a product that does not meet the
may have an impact on the product but is manufacturer’s specifications or those in the
not as significant as a critical observation. It authorized pharmacopeia
may have an indirect impact in the strength,
identity, purity or safety of the product. There 3. GUIDE
is reduced usability of the product without a
probability of causing harm to the consumer. Whereas it is recognized that it is impossible
Observation of a major deficiency puts a to encompass every situation that may
question mark on the reliability of the firm’s generate a risk, the following principles should
quality assurance system. be considered:
59
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
60
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
61
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
62
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
3.6.2.3 Clean in place (CIP) equipment not 3.6.3.3 Insufficient training for personnel
validated. involved in production and QC
resulting in related GMP violations.
3.6.2.4 Tanks for manufacturing of liquids
and ointments not equipped with 3.6.3.4 No medical check-ups for personnel
sanitary clamps. involved in critical areas of
production and quality control
3.6.2.5 Stored equipment not protected
from contaminations.* 3.6.4 Sanitation
3.6.2.6 Inappropriate equipment for 3.6.4.1 Sanitation program not in writing but
production: surfaces porous and premises in acceptable state of
non-cleanable/material to shed cleanliness.
particles.*
3.6.4.2 No Standard Operating Procedure
3.6.2.7 No covers for tanks, hoppers or (SOP) for microbial/environmental
similar manufacturing equipment. monitoring, no action limits for areas
where susceptible non-sterile
products are manufactured.
63
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
3.6.4.3 Cleaning procedure for production 3.6.6.4 Line clearance between productions
equipment not validated (including of different products not covered by
analytical methods). SOP and not documented.
3.6.5.9 No SOP for conditions of 3.6.6.11 Raw materials dispensing not done
transportation and storage. by qualified persons, according
to SOP.
3.6.6 Manufacturing Control
3.6.6.12 Master Formulae incomplete or
3.6.6.1 Master Formulae prepared/verified showing inaccuracies in the
by unqualified personnel. processing operations.
3.6.6.2 Deviations from instructions during 3.6.6.13 Changes in batch size not prepared/
production not documented and not verified by qualified personnel
approved.
3.6.6.14 Inaccurate/incomplete information in
3.6.6.3 Discrepancies in yield or manufacturing/packaging batch
reconciliation following production document.
not investigated.
64
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
3.6.6.15 Although documented, combination 3.6.8.3 No SOP approved and available for
of batches done without QC sampling, inspection and testing of
approval/not covered by SOP. materials.
3.6.6.16 No written procedures for packaging 3.6.8.4 Products made available for sale
operations. without approval of QC department.*
65
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
3.6.13.2 No action taken when data 3.6.14.8 Inadequate gowning practices for
shows that the products do not meet clean and aseptic areas.
their specifications prior to the
expiry date. 3.6.14.9 Inadequate practices/precautions to
minimize contamination or prevent
mix-ups.
66
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
3.6.14.19 Purified water is not used as the 3.7.2.1 Insufficient space between
feed water for water for injection equipment and walls to permit
system and the clean steam cleaning.
generator.
3.7.2.2 Base of immovable equipment not
3.6.14.20 The water for injection used in the adequately sealed at points of
preparation of parenterals is not contact.
tested for endotoxins.
3.7.2.3 Use of temporary means or devices
3.6.14.21 The water for injection used for final for repair.
rinsing of containers and
components used for parenteral 3.7.2.4 Defective or unused equipment
drugs is not tested for endotoxins used for non-critical products not
when those containers and qualified.
67
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
68
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
69
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
70
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.2 A site shall be considered non- 5.8 Manufacturing facilities that fail
compliant if it has: to comply with GMP shall have:
5.2.1 One or more critical non 5.8.1 their products removed from the
compliances national medicine register thus
halting further manufacture or
5.2.2 Several major non compliances that importation of the products
imply a failure in the quality
assurance system 5.8.2 Their products recalled from the
market depending on the criticality
5.3 The NMRA shall issue a GMP of the findings (see SOP on recall of
certificate and /or a products from the market annex
manufacturing license where VI H).
applicable for a site that is
compliant i.e has no critical or 5.9 The NMRA may decide to close
has minor observations down the whole site by
withdrawing the GMP certificate
5.4 The NMRA shall demand for a and/or manufacturing license or a
corrective and preventive action section of the site depending on
report for review and where the critical observations
possible a follow up inspection identified by the inspection team
for a site that has major non
compliances may be done prior 5.10 Upon careful consideration of
to issue of a GMP certificate and the findings in the inspection
close out of the inspection. report and where these affect the
health of patients in a critical
5.5 The NMRA shall not issue manner; the NMRA may consider
a GMP certificate to a non- raising a rapid alert to EAC
compliant manufacturing facility NMRAs, health care workers
that has critical or several major and patients
non compliances
6. RECORDS
5.6 Local manufacturers shall require
physical re-inspection for a 6.1 The quality manuals, master
site that has critical or several distribution list file, obsolete
major non compliances until a documents file and general list of
satisfactory report is achieved documents shall be kept and
maintained by HQM for a period
5.7 The re-inspection of a non- specified in the respective
compliant facility shall be after document
submission of corrective action
report and an application
together with payment of the
inspection fee.
71
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
72
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
1. PURPOSE 6. PROCEDURE
To outline the procedure for classification and 6.1 OCCASIONS UNDER WHICH
communications involved in a product recall or A PRODUCT MAY BE RECALLED
withdrawal OR WITHDRAWN
73
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
6.2.3. Situation in which the use of or Action: Recall Notification to all distribution
exposure to a suspect product points plus Media release.
is not likely to cause any adverse
health consequences 6.3.2. Type B
A type B recall is designed to reach
The following classification criterion is wholesalers throughout the country,
recommended: directors of hospital services
(private as well as state hospitals),
Class I retail outlets, doctors, nurses,
pharmacists, authorized prescribers
Class I is for defective/dangerous/potentially and dispensers.
life threatening medicines that predictably or
probably could result into serious health risk/ Action: Recall letter to all distribution points.
adverse events or even death.
6.3.3. Type C
Class II A type C recall is designed to reach
wholesale level and other
Class II is for medicines that possibly could distribution points (e.g. pharmacies,
cause temporary or medically reversible doctors, hospitals). This can be
adverse health problem or mistreatment. achieved by means of
representatives calling on
Class III wholesalers and/or retail outlets. If
it is known where the product in
Class III is for medicine that is defective and is question had been distributed to,
unlikely to cause any adverse health reaction specific telephone calls or recalls
or which do not comply with the requirements letters to arrange for the return of
of the NMRA Laws of the individual partner the product could be made.
States and regional bidding laws and
regulations of the EAC Action: Specific telephone calls, recall letters/
representatives calling at distribution points
6.3. TYPES OF RECALL if known where the medicines have been
distributed.
6.3.1. Type A
A type A recall is designed to reach 6.4. RECALL NOTIFICATION
all suppliers of medicines (all
distribution points) i.e. wholesalers It is imperative that before or upon initiating a
throughout the country, directors recall, the company immediately on becoming
of hospital services (private as well aware of the problem, notifies the head NMRA
as state hospitals), retail outlets, or, in his absence, his designate
doctors, nurses, pharmacists,
authorized prescribers and If the notification fails and there is urgent
dispensers and individual customers need to recall the product then the company
or patients through media release may proceed according to their discretion
(radio, television, internet, regional and follow up contact with the NMRA to be
and national press). pursued in the process.
74
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
6.5. BASIC INFORMATION REQUIRED or considered for recall will also be conducted
FOR RECALL by the NMRA and will take into account, but
need not be limited to, assessment of the
6.5.1. Name, strength, pack size, batch/ following factors:
lot number and means of
identification of the recalled product 6.6.1. Whether any disease or injuries
have already occurred from the use
6.5.2. Total quantity of the product being of the product
re called originally in possession of
the company 6.6.2. Hazard to various segments of the
population e.g. children, surgical
6.5.3. The date distribution of the product patients etc, who are expected to
began be exposed to the product, with
particular attention to those
6.5.4. The total quantity of the product individuals who may be at greatest
being re called that had been risk
distributed up to the time of the
recall should be indicated. 6.6.3. The degree of seriousness of the
health hazard to which the
6.5.5. Area of distribution of the product population at greatest risk would
and, if exported, the country to be exposed.
where it was exported.
6.6.4. The likelihood of occurrence of
6.5.6. List of customers to whom product that hazard
was issued
6.6.5. The consequences (immediate
6.5.7. The quantity of the recalled product or long-term) of occurrence of
still in their possession the hazard. The recalling company
will be given every opportunity
6.5.8. The reason for initiating the recall; to contribute to the information
nature of defect on which the health hazard
evaluation is made by the NMRA,
6.5.9. Suggested action to be taken and who, on the basis of this
its urgency determination, classifies it based on
the relative degree of health hazard
6.5.10. Indication of the health risk together posed by the product being recalled
with reasons or considered for recall.
Before initiating a recall, the company will In formulating a recall strategy, the following
gather, correlate and evaluate all known should be taken into consideration:
information on the nature and extent of the
reputed health risk. An evaluation of the health
hazard presented by a product being recalled
75
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
6.7.1. Result of health hazard evaluation with the hazard of the product and the
strategy developed for that recall. Recall
6.7.2. Ease in identifying the product communication should convey:
6.7.3. Extent to which the product 6.9.1. That the product in question is
deficiency is obvious to the subject to recall
consumer/user
6.9.2. That further distribution or use
6.7.4. Continued availability of essential of any remaining product should
products (risk: benefit) cease immediately
76
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
NMRA to decide if necessary and who to issue 6.12. POST RECALL PROCEDURES
such a warning and the type of public warning
should be specified in the recall strategy for The NMRA must be furnished with a report
the product e.g. within a specified period (2 weeks) of the
recall or withdrawal being instituted. The
General public warning in the general media report should contain the following information:
as appropriate and a public warning through
specialized news media to professionals or 6.12.1. Name of the product
to specific segments of the population like
physicians, hospitals etc. 6.12.2. Strength of the product
6.11.3. Indicate nature of the defect 6.12.10. Indication of the health risk and
reported clinical problems
6.11.4. Specify urgency of the action
6.12.11. Copies of all the recall
6.11.5. Indicate reason for the action correspondence; and
6.11.6. Indicate the health risk; and 6.12.12. Steps taken to prevent re-
occurrence of the problem
6.11.7. Provide specific instructions on what
should be done with the recalled 6.12.13. After termination of a recall and
product. not later than 90 days after a recall
has been instituted, a full
Note: Where necessary, follow-up reconciliation must be submitted.
communication should be sent to those
who fail to respond to the initial recall A recall will be terminated when the NMRA
communication. This should be done within a and the recalling company are in agreement
reasonable time depending on the urgency of that the non-compliant product has been
the recall. removed and proper disposal or correction
has been made.
77
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
78
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
79
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
80
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
81
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
PART TWO:
82
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
83
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
MA - Marketing Authorization
MAL - Material Air Lock
MRA - Medicines Regulatory Authority
MTC - Manufacturing Technology Committee
NEPAD - New Partnership for Africa’s Development
NMRA - National Medicines Regulatory Authority
OQ - Operational Qualification
OSD - Oral Solid Dosage
PAL - Personnel Air Lock
pH - Power of Hydrogen
Ph. Eur - European Pharmacopoeia
Ph. Int. - International Pharmacopoeia
PIC/S - Pharmaceutical Inspection Cooperation Scheme
PP - Process Parameter
PQ - Performance Qualification
PQRI - Product Quality Research Institute
PQS - Pharmaceutical Quality System
PW - Purified Water
QA - Quality Assurance
QC - Quality Control
QRM - Quality Risk Management
QTPP - Quality Target Product Profile
Requirements for Registration of Pharmaceuticals for Human Use
RMP - Risk Management Plan
SCADA - System Control and Data Acquisition
SOP - Standard Operating Procedure
TWG - Technical Working Group
UDAF - Unidirectional Air Flow
USP - United State Pharmacopoeia
VMP - Validation Master Plan
WFI - Water for Injection
WHO - World Health Organization
84
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
85
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
86
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
87
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Reprocessing: The reworking of all or part of Validation protocol (or plan): A document
a batch of product of an unacceptable quality describing the activities to be performed in a
from a defined stage of production so that its validation, including the acceptance criteria for
quality may be rendered acceptable by one or the approval of a manufacturing process - or a
more additional operations. part thereof - for routine use.
Returned product: Finished product sent Validation report: A document in which the
back to the manufacturer. records, results and evaluation of a completed
Revalidation: Repeated validation of an validation program are assembled. It may
approved process (or a part thereof) to also contain proposals for the improvement of
ensure continued compliance with established processes and/or equipment.
requirements.
88
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
1. INTRODUCTION 2. SCOPE
The quality of medicinal products in the EAC This guideline and its annexes shall be used
region has always been a concern of National as a basis for the inspection of medicinal
Medicine Regulatory Authorities. EAC NMRAs products manufacturing facilities and as a
strive to ensure quality, safety and efficacy standard to justify GMP status during the
of human and veterinary medicines and assessment of applications for manufacturing
other health care products through regulation authorizations. The annexes provide details
and control of their production, importation, on specific areas which include; sterile
distribution and use. preparations, biological medicinal products
and vaccines for human use, computerized
Through the medicine regulation systems, water for pharmaceutical use,
harmonization initiative in the EAC region, heating ventilation and air conditioning
the GMP guidelines have been developed systems, qualification & validation, GMP
to ensure that medicinal products marketed for manufacture of active pharmaceutical
in the Partner States meet uniform and ingredients, waste management for
acceptable quality, safety and efficacy. medicinal product manufacturers, quality risk
management and authorized persons.
The EAC Guide on Good Manufacturing
Practices (GMP) is based on the World Health This guideline is applicable to all
Organization (WHO) Good Manufacturing manufacturers (local and foreign) of finished
Practices guidelines in the WHO Technical pharmaceutical product formulations
Report Series No. 961, 2011. This guide and active pharmaceutical ingredients
also refers to the Pharmaceutical Inspection manufactured and marketed in the EAC region
Convention Scheme (PIC/S) guide PE 009-9. for human use.
89
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
90
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
(g) Medicinal products are not sold and as required by the marketing
or supplied before the authorized authorization and product
persons have certified that each specifications. GMP rules are
production batch has been directed primarily to diminishing
produced and controlled in the risks, inherent in any
accordance with the requirements pharmaceutical production that
of the marketing authorization cannot be prevented completely
and any other regulations relevant through the testing of final
to the production, control and products. Such risks are essentially
release of medicinal products; of two types: cross-contamination
(h) Satisfactory arrangements exist (in particular by unexpected
to ensure, as far as possible, that contaminants) and mix-ups
the medicinal products are stored, (confusion) caused by false
distributed, and subsequently labels being put on containers. The
handled so that quality is maintained basic requirements of GMP are that:
throughout their shelf-life;
(i) There is a procedure for self- (a) all manufacturing processes are
inspection and/or quality audit that clearly defined, systematically
regularly appraises the reviewed in the light of experience,
effectiveness and applicability of the and shown to be capable of
quality assurance system; consistently manufacturing
(j) Deviations are reported, medicinal products of the required
investigated and recorded; quality that comply with their
(k) There is a system for approving specifications;
changes that may have an impact (b) critical steps of manufacturing
on product quality; processes and any significant
(l) Regular evaluation of the quality of changes made to the processes
pharmaceutical products should be are validated;
conducted with the objective of (c) all necessary facilities are provided,
verifying the consistency of including:
the process and ensuring its
continuous improvement; (i) appropriately qualified
(m) And there is a system for quality risk and trained personnel;
management (QRM); (ii) adequate premises
and space;
GOOD MANUFACTURING PRACTICES (iii) suitable equipment
(GMP) and services;
(iv) correct materials,
1.4 Good manufacturing practice is containers,
that part of quality assurance and labels;
which ensures that products (v) approved procedures
are consistently produced and and instructions;
controlled to the quality standards (vi) suitable storage and
appropriate to their intended use transport, and;
91
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
92
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
93
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
94
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
95
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
96
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
97
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
98
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
99
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
risk of errors and permit effective cleaning 3.6 Premises should be designed
and maintenance in order to avoid cross- and equipped so as to provide
contamination, build-up of dust or dirt, and, in maximum protection against the
general, any adverse effect on the quality of entry of insects, birds or other
products. animals. There should be a
procedure for rodent and pest
GENERAL control.
100
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
3.9 The adequacy of the working and to the products handled, to the
in-process storage space should operations undertaken, and to the
permit the orderly and logical external environment. These
positioning of equipment and areas should be regularly monitored
materials so as to minimize the risk during production and non-
of confusion between different production periods to ensure
pharmaceutical products or their compliance with their design
components, to avoid cross- specifications.
contamination, and to minimize the
risk of omission or wrong application 3.14 Where dust is generated (e.g.
of any of the manufacturing or during sampling, weighing, mixing,
control steps. processing operations and
packaging of powders) measures
3.10 Where starting and primary should be taken to avoid cross
packaging materials and contamination and facilitate
intermediate or bulk products are cleaning.
exposed to the environment, interior
surfaces (walls, floors, and ceilings) 3.15 Premises for the packaging of
should be smooth and free from medicinal products should be
cracks and open joints, should not specifically designed and laid out so
shed particulate matter, and should as to avoid mix-ups or cross-
permit easy and effective cleaning contamination.
and, if necessary, disinfection.
3.16 Production areas should be well lit,
3.11 Pipe work, light fittings, ventilation particularly where visual on-line
points, and other services should controls are carried out.
be designed and sited to avoid the
creation of recesses that are difficult STORAGE AREAS
to clean. As far as possible, for
maintenance purposes, they should 3.17 Storage areas should be of
be accessible from outside the sufficient capacity to allow
manufacturing areas. orderly storage of the various
categories of materials and
3.12 Drains should be of adequate size products: starting and packaging
and equipped to prevent back-flow. materials, intermediates, bulk and
Open channels should be avoided finished products, products in
where possible, but if they are quarantine, and released, rejected,
necessary they should be shallow to returned, or recalled products.
facilitate cleaning and disinfection.
3.18 Storage areas should be
3.13 Production areas should be designed or adapted to ensure good
effectively ventilated, with air-control storage conditions. In particular,
facilities (including control of they should be clean and
temperature and, where necessary, dry, sufficiently lit and maintained
humidity and filtration) appropriate within acceptable temperature limits.
101
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
102
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
103
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
104
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
105
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
106
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
107
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.22 The master formula should include: (e) where necessary, the requirements
for storage of the products, including
(a) the name of the product, with the container, the labelling, and any
a product reference code relating to special storage conditions;
its specification; (f) any special precautions to be
(b) a description of the dosage form, observed.
strength of the product, and batch
size; PACKAGING INSTRUCTIONS
(c) a list of all starting materials to
be used (if applicable, with the 5.24 There should be formally approved
International Nonproprietary packaging instructions for each
Names), with the amount of each, product pack size and type. These
described using the designated should normally include, or make
name and a reference that is unique reference to the following:
to that material (mention should be
made of any substance that may (a) the name of the product;
disappear in the course of (b) a description of its pharmaceutical
processing); form, strength, and method of
(d) a statement of the expected final application where applicable;
yield with the acceptable limits, (c) the pack size expressed in terms of
and of relevant intermediate yields, the number, weight, or volume of
where applicable. the product in the final container;
(d) a complete list of all the packaging
5.23 The processing Instructions should materials required for a standard
include: batch size, including quantities,
sizes, and types, with the code or
(a) a statement of the processing reference number relating to the
location and the principal equipment specifications for each packaging
to be used; material;
(b) the methods, or reference to the (e) where appropriate, an example or
methods, to be used for preparing reproduction of the relevant printed
the critical equipment, e.g., cleaning packaging materials and specimens,
(especially after a change in indicating where the batch number
product), assembling, calibrating, and expiry date of the product have
sterilizing; been marked;
(c) detailed stepwise processing (f) special precautions to be observed,
instructions (e.g., checks on including a careful examination of
materials, pretreatments, the packaging area and equipment
sequence for adding materials, in order to ascertain the line
mixing times, temperatures); clearance before and after
(d) the instructions for any in-process operations ;
controls with their limits; (g) a description of the packaging
operation, including any significant
subsidiary operations, and
equipment to be used;
108
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
109
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
is clean and suitable for use. These (h) notes on any special problems,
checks should be recorded. including details of any deviation
from the packaging instructions, with
5.30 The following information should written authorization by an
be recorded at the time each action appropriate person;
is taken and, after completion, the (i) the quantities and reference number
date and the person responsible or identification of all printed
should be clearly identified by packaging materials and bulk
signature or electronic password: product issued, used, destroyed, or
returned to stock and the quantities
(a) the name of the product, the batch of product obtained to permit an
number, and the quantity of bulk adequate reconciliation.
product to be packed, as well as
the batch number and the planned PROCEDURES (SOPS) AND
quantity of finished product that will RECORDS RECEIPTS
be obtained, the quantity actually
obtained, and the reconciliation; 5.31 There should be written standard
(b) the date(s) and time(s) of the procedures and records for the
packaging operations; receipt of each delivery of each
(c) the name of the responsible person starting material and primary and
carrying out the packaging printed packaging material.
operation;
(d) the initials of the operators of the 5.32 The records of the receipts should
different significant steps; include:
(e) the checks made for identity and
conformity with the packaging (a) the name of the material on the
instructions, including the results of delivery note and the containers;
in-process controls; (b) the “in-house” name and/or code of
(f) details of the packaging operations material if different from (a);
carried out, including references (c) the date of receipt;
to equipment and the packaging (d) the supplier’s name and, if possible,
lines used, and, when necessary, manufacturer’s name;
the instructions for keeping the (e) the manufacturer’s batch or
product unpacked or a record of reference number;
returning product that has not been (f) the total quantity, and number of
packaged to the storage area; containers received;
(g) whenever possible, samples of the (g) the batch number assigned after
printed packaging materials used, receipt;
including specimens bearing (h) any relevant comment (e.g., state of
approval of the printing, the batch the containers).
number, expiry date, and any
additional overprinting;
110
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.33 There should be written standard 5.37 Analysis records should include at
operating procedures for the internal least the following data:
labeling, quarantine, and storage of
starting materials, packaging (a) the name of the material or product
materials, and other materials, as and, where applicable, dosage form;
appropriate. (b) the batch number and, where
appropriate, the manufacturer and/
SAMPLING or supplier;
(c) references to the relevant
5.34 There should be standard operating specifications and testing
procedures for sampling, which procedures;
specify the person(s) authorized to (d) test results, including observations
take samples. and calculations, and reference to
any specifications (limits);
5.35 The sampling instructions should (e) dates and reference number of
include: testing;
(f) the initials of the persons who
(a) the method of sampling and the performed the testing;
sampling plan; (g) the dates and initials of the persons
(b) the equipment to be used; who verified the testing and the
(c) any precautions to be observed to calculations, where appropriate;
avoid contamination of the material (h) a clear statement of release or
or any deterioration in its quality; rejection (or other status decision)
(d) the amount(s) of sample(s) to be and the dated signature of the
taken; designated responsible person.
(e) instructions for any required
subdivision of the sample; OTHERS
(f) the type of sample container(s) to
be used, and whether they are for 5.38 There should be a standard
aseptic sampling or for normal operating procedure describing the
sampling; details of the batch (lot) numbering
(g) any specific precautions to be system, with the objective of
observed, especially in regard to the ensuring that each batch of
sampling of sterile or noxious intermediate, bulk, or finished
material. product is identified with a specific
batch number.
TESTING
The standard operating procedures
5.36 There should be written procedures for batch numbering that are applied
for testing materials and products at to the processing stage and to the
different stages of manufacture, respective packaging stage should
describing the methods and be related to each other.
equipment to be used. The tests
performed should be recorded.
111
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.39 The standard operating procedure 5.44 Logbooks should be kept with major
for batch numbering should and critical equipment and should
assure that the same batch record, as appropriate, any
numbers will not be repeatedly validations, calibrations,
used; this applies also to maintenance, cleaning, or repair
reprocessing. operations, including dates and
the identity of the people who
5.40 Batch-number allocation should carried these operations out.
be immediately recorded, e.g., in
a logbook. The record should 5.45 Clear standard operating
include date of allocation, product procedures should be available
identity, and size of batch. for major items of manufacturing
and test equipment and placed in
5.41 Written release and rejection close proximity to the equipment.
procedures should be available for
materials and products, and in 5.46 The use of major and critical
particular for the release for sale of equipment and the areas where
the finished product by an products have been processed
authorized person. should be appropriately recorded in
chronological order.
5.42 Records should be maintained for
the distribution of each batch of a 5.47 There should be written procedures
product in order to facilitate the assigning responsibility for cleaning
recall of the batch if necessary. and sanitation and describing
in sufficient detail the cleaning
5.43 Standard operating procedures and schedules, methods, equipment,
associated records of actions taken and materials to be used and
or, where appropriate, conclusions facilities to be cleaned. Such written
reached should be available for: procedures should be followed.
112
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
113
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
114
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
115
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
labeled. Labels should bear at least 6.32 Materials dispensed for each batch
the following information: of the final product should be kept
together and conspicuously labeled
(a) the designated name of the product as such.
and the internal code reference
where applicable; 6.33 All incoming materials and finished
(b) the batch number(s) given by the products should be quarantined
supplier and on receipt by the immediately after receipt or
manufacturer, if any; processing, until they are released
(c) where appropriate, the status of for use or distribution.
the contents (e.g., on quarantine, on
test, released, rejected, returned, 6.34 All materials and products should
recalled); be stored under the appropriate
(d) where appropriate, an expiry date or conditions established by the
a date beyond which retesting is manufacturer and in an orderly
necessary. fashion to permit batch segregation
and stock rotation by a first-expiry,
When fully computerized storage first-out rule.
systems are used, not all of the
above information need be in a PROCESSING OPERATIONS:
legible form on the label. INTERMEDIATE AND BULK
PRODUCTS
6.28 There should be appropriate
procedures or measures to ensure 6.35 Before any processing operation is
the identity of the contents of each started, steps should be taken to
container of starting material. Bulk ensure that the work area and
containers from which samples have equipment are clean and free from
been drawn should be identified. any starting materials, products,
product residues, labels, or
6.29 Only starting materials released by documents not required for the
the quality control department and current operation.
within their shelf-life should be used.
6.36 Intermediate and bulk products
6.30 Starting materials should be should be kept under appropriate
dispensed only by designated conditions.
persons, following a written
procedure, to ensure that the correct 6.37 Intermediate and bulk products
materials are accurately weighed purchased as such should be
or measured into clean and properly handled on receipt as though they
labeled containers. were starting materials.
6.31 Each dispensed material and its 6.38 Any necessary in-process controls
weight or volume should be and environmental controls should
independently checked and the be carried out and recorded.
check recorded.
116
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
117
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
6.50 The name and batch number of the to ensure that any electronic code
product being handled should be readers, label counters, or similar
displayed at each packaging station devices are operating correctly.
or line. When labels are attached manually,
in-process control checksshould be
6.51 All products and packaging performed more frequently.
materials to be used should
be checked on delivery to 6.56 Checks should be made to ensure
the packaging department for that any electronic code readers,
quantity , identity and conformity label counters, or similar devices
with the packaging instructions. are operating correctly.
6.52 Containers for filling should be clean 6.57 Printed and embossed information
before filling. Attention should on packaging materials should be
be given to avoiding and removing distinct and resistant to fading
any contaminants such as glass or erasing.
fragments and metal particles.
6.58 On-line control of the product during
6.53 Normally, filling and sealing should packaging should include at least
be followed as quickly as possible checks on:
by labeling. If labeling is delayed,
appropriate procedures should (a) the general appearance of the
be applied to ensure that no mix-ups packages;
or mislabeling can occur. (b) whether the packages are complete;
(c) whether the correct products and
6.54 The correct performance of any packaging materials are used;
printing (for example of code (d) whether any overprinting is correct;
numbers or expiry dates) done (e) the correct functioning of line
separately or in the course of the monitors.
packaging should be checked and
recorded. Attention should be Samples taken away from the
paid to printing by hand, which packaging line should not be
should be rechecked at regular returned.
intervals.
6.59 Products that have been involved in
6.55 Special care should be taken when an unusual event during packaging
cut labels are used and when should be reintroduced into
overprinting is carried out off-line, the process only after special
and in hand-packaging operations. inspection, investigation, and
Roll-feed labels are normally approval by authorized personnel.
preferable to cut labels in helping to A detailed record should be kept of
avoid mix-ups. Online verification of this operation.
all labels by automated electronic
means can be helpful in preventing
mix ups, but checks should be made
118
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
119
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
120
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
121
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
122
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
123
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
124
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
7.32 A summary of all the data 7.36 Stability should be determined prior
generated, including any interim to marketing and following any
conclusions on the programme, significant changes in processes,
should be written and maintained. equipment, packaging materials,
This summary should be subjected etc.
to periodic review.
REAGENTS AND CULTURE MEDIA
7.33 The quality control department
should evaluate the quality and 7.37 All reagents and culture media
stability of finished pharmaceutical should be recorded upon receipt or
products and, when necessary, of preparation.
starting materials and intermediate
products. 7.38 Reagents made up in the laboratory
should be prepared according to
7.34 The quality control department written procedures and
should establish expiry dates and appropriately labeled. The
shelf-life specifications on the basis label should indicate the
of stability tests related to storage concentration, standardization
conditions. factor, shelf-life, the date when
re-standardization is due, and
7.35 A written programme for ongoing the storage conditions. The label
stability determination should should be signed and dated by the
be developed and implemented to person preparing the reagent.
include elements such as:
125
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
126
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
127
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
aspects of the contract should be 8.15 The contract should describe the
drawn up by competent persons handling of starting materials,
suitably knowledgeable in intermediate and bulk products, and
pharmaceutical technology, finished products if they are
analysis, and GMP. All rejected. It should also describe
arrangements for production and the processing of information if
analysis must be in accordance with the contract analysis shows that the
the marketing authorization and tested product must be rejected.
agreed by both parties.
8.16 Technical aspects of the contract
8.12 The contract should specify the should be drawn up by
way in which the authorized person competentpersons suitably
releasing the batch for sale ensures knowledgeable in pharmaceutical
that each batch has been technology, analysis and GMP.
manufactured in, and checked
for, compliance with the 8.17 All arrangements for production
requirements of the marketing and analysis must be in
authorization. accordance with the marketing
authorization and agreed by both
8.13 The contract should describe parties.
clearly who is responsible for
purchasing, testing, and releasing CHAPTER 9: COMPLAINTS
materials and for undertaking
production and quality controls, HANDLING AND PRODUCT
including in-process controls, and RECALL
who has responsibility for sampling
and analysis. In the case of contract PRINCIPLE
analysis, the contract should
state whether or not the contract All complaints and other information
accepter should take samples at the concerning potentially defective products
premises of the manufacturer. must be carefully reviewed according to
written procedures. In order to provide for all
8.14 Manufacturing, analytical, and contingencies, a system should be designed
distribution records and reference to recall, if necessary, promptly and effectively
samples should be kept by, or be products known or suspected to be defective
available to, the contract giver. from the market complaints.
Any records relevant to assessing
the quality of a product in the event 9.1 A person responsible for handling
of complaints or a suspected defect the complaints and deciding the
must be accessible and specified measures to be taken should be
in the defect/recall procedures of designated, together with sufficient
the contract giver. supporting staff to assist him or her.
If this person is different from the
authorized person, the latter
128
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
129
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
130
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
131
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
7. REVISION HISTORY
8. LIST OF ANNEXES
132
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
133
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Grade B: For aseptic preparation and filling, this is the background environment for grade A zone.
Grade C and D: Clean areas for carrying out less critical stages in the manufacture of sterile
products.
The airborne particulate classification for these grades is given in the following table.
Notes:
(a) Particle measurement based on the use of a discrete airborne particle counter to
measure the concentration of particles at designated sizes equal to or greater than
the threshold stated. A continuous measurement system should be used for monitoring
the concentration of particles in the grade A zone, and is recommended for the
surrounding grade B areas. For routine testing the total sample volume should not be
less than 1 m³ for grade A and B areas and preferably also in grade C areas.
(b) The particulate conditions given in the table for the “at rest” state should be achieved
after a short “clean up” period of 15-20 minutes (guidance value) in an unmanned state
after completion of operations. The particulate conditions for grade A “in operation”
given in the table should be maintained in the zone immediately surrounding the product
whenever the product or open container is exposed to the environment. It is accepted
that it may not always be possible to demonstrate conformity with particulate standard
at the point of fill when filling is in progress, due to the generation of particles or droplets
from the product itself.
(c) In order to reach the B, C and D air grades, the number of air changes should be
related to the size of the room and the equipment and personnel present in the room.
The air system should be provided with appropriate terminal filters such as HEPA for
grades A, B and C.
(d) The guidance given for the maximum permitted number of particles in the “at rest”
and “in operation” conditions correspond approximately to the cleanliness classes in the
EN/ISO 14644-1 at a particle size of 0.5 µm.
134
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
(e) These areas are expected to be completely free from particles of size greater than
5 micrometer. As it is impossible to demonstrate the absence of particles with any
statistical significance, the limits are set to 1 particle /m3. During the clean room
qualification it should be shown that the areas can be maintained within the defined
limits.
(f) The requirements and limits will depend on the nature of the operations carried out.
Examples of operations to be carried out in the various grades are given in the table below
(see also para. 11 and 12):
4. The areas should be monitored during operation in order to control the particulate
cleanliness of the various grades.
5. Where aseptic operations are performed monitoring should be frequent using methods
such as settle plates, volumetric air and surface sampling (e.g. swabs and contact
plates). Sampling methods used in operation should not interfere with zone protection.
Results from monitoring should be considered when reviewing batch documentation for
finished product release. Surfaces and personnel should be monitored after critical
operations.
135
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Notes:
(a) These are average values.
(b) Individual settle plates may be exposed for less than 4 hours.
6. Appropriate alert and action limits should be set for the results of particulate and
microbiological monitoring. If these limits are exceeded operating procedures should
prescribe corrective action.
ISOLATOR TECHNOLOGY
The transfer of materials into and out of the unit is one of the greatest potential
sources of contamination. In general the area inside the isolator is the local zone for
high risk manipulations, although it is recognized that laminar air flow may not exist
in the working zone of all such devices. The air classification required for the
background environment depends on the design of the isolator and its application. It
should be controlled and for aseptic processing be at least grade D.
8. Isolators should be introduced only after appropriate validation. Validation should take
into account all critical factors of isolator technology, for example the quality of the air
inside and outside (background) the isolator, sanitation of the isolator, the transfer
process and isolator integrity.
136
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
137
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
138
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
139
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
140
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
141
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
142
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
143
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
144
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
64. Sufficient time must be allowed for 67. The items to be sterilized, other
the whole of the load to reach the than products in sealed containers,
required temperature before should be wrapped in a material
measurement of the sterilizing which allows removal of air and
time-period is commenced. This penetration of steam but which
time must be determined for each prevents recontamination after
type of load to be processed. sterilization. All parts of the load
should be in contact with the
65. After the high temperature phase of sterilizing agent at the required
a heat sterilization cycle, temperature for the required time.
precautions should be taken against
contamination of a sterilized load 68. Care should be taken to ensure
during cooling. Any cooling fluid that steam used for sterilization is of
or gas in contact with the product suitable quality and does not
should be sterilized, unless it can be contain additives at a level which
shown that any leaking container could cause contamination of
would not be approved for use. product or equipment.
66. Both temperature and pressure 69. The process used should include
should be used to monitor the air circulation within the chamber
process. Control instrumentation and the maintenance of a positive
should normally be independent pressure to prevent the entry of non-
of monitoring instrumentation and sterile air. Any air admitted should
recording charts. Where automated be passed through a HEPA filter.
control and monitoring systems Where this process is also intended
are used for these applications they to remove pyrogens, challenge tests
should be validated to ensure using endotoxins should be used as
that critical process requirements part of the validation.
are met. System and cycle faults
should be registered by the system STERILIZATION BY RADIATION
and observed by the operator.
The reading of the independent 70. Radiation sterilization is used mainly
temperature indicator should be for the sterilization of heat sensitive
routinely checked against the chart materials and products. Many
recorder during the sterilization medicinal products and some
period. For sterilizers fitted with a packaging materials are radiation-
drain at the bottom of the chamber, sensitive, so this method is
it may also be necessary to record permissible only when the absence
the temperature at this position, of deleterious effects on the product
throughout the sterilization period. has been confirmed experimentally.
There should be frequent leak tests
on the chamber when a vacuum
phase is part of the cycle.
145
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
146
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
80. For each sterilization cycle, records the filtration process with some
should be made of the time taken degree of heat treatment.
to complete the cycle, of the
pressure, temperature and humidity 83. Due to the potential additional risks
within the chamber during the of the filtration method as compared
process and of the gas with other sterilization processes,
concentration and of the total a second filtration via a further
amount of gas used. The pressure sterilized microorganism retaining
and temperature should be recorded filter, immediately prior to filling, may
throughout the cycle on a chart. The be advisable. The final sterile
record(s) should form part of the filtration should be carried out as
batch record. close as possible to the filling point.
81. After sterilization, the load should be 84. Fibre shedding characteristics of
stored in a controlled manner under filters should be minimal.
ventilated conditions to allow
residual gas and reaction products 85. The integrity of the sterilized filter
to reduce to the defined level. This should be verified before use and
process should be validated. should be confirmed immediately
after use by an appropriate method
FILTRATION OF MEDICINAL such as a bubble point, diffusive
PRODUCTS WHICH CANNOT flow or pressure hold test. The time
BE STERILIZED IN THEIR FINAL taken to filter a known volume of
bulk solution and the pressure
CONTAINER
difference to be used across the
filter should be determined during
82. Filtration alone is not considered
validation and any significant
sufficient when sterilization in
differences during routine
the final container is possible. With
manufacturing from this should
regard to methods currently
be noted and investigated. Results
available, steam sterilization is
of these checks should be included
to be preferred. If the product
in the batch record. The integrity
cannot be sterilized in the final
of critical gas and air vent filters
container, solutions or liquids can
should be confirmed after use. The
be filtered through a sterile filter of
integrity of other filters should be
nominal pore size of 0.22 micron
confirmed at appropriate intervals.
(or less), or with at least equivalent
micro-organism retaining
86. The same filter should not be used
properties, into a previously
for more than one working day
sterilized container. Such filters can
unless such use has been validated.
remove most bacteria and
moulds, but not all viruses or
87. The filter should not affect the
mycoplasma’s. Consideration
product by removal of ingredients
should be given to complementing
from it or by release of substances
into it.
147
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
88. Containers should be closed by 91. The sterility test applied to the
appropriately validated methods. finished product should only be
Containers closed by fusion, e.g. regarded as the last in a series of
glass or plastic ampoules should control measures by which sterility
be subject to 100% integrity testing. is assured. The test should be
Samples of other containers validated for the product(s)
should be checked for integrity concerned.
according to appropriate
procedures. 92. In those cases where parametric
release has been authorized,
89. Containers sealed under vacuum special attention should be paid to
should be tested for maintenance the validation and the monitoring of
of that vacuum after an appropriate, the entire manufacturing process.
pre-determined period.
93. Samples taken for sterility testing
90. Filled containers of parenteral should be representative of the
products should be inspected whole of the batch, but should in
individually for extraneous particular include samples taken
contamination or other defects. from parts of the batch considered
When inspection is done visually, it to be most at risk of contamination,
should be done under suitable e.g.:
and controlled conditions of
illumination and background. a) for products which have been filled
Operators doing the inspection aseptically, samples should include
should pass regular eye-sight containers filled at the beginning
checks, with spectacles if worn, and and end of the batch and after any
be allowed frequent breaks from significant intervention;
inspection. Where other methods b) for products which have been
of inspection are used, the process heat sterilized in their final
should be validated and the containers, consideration should be
performance of the equipment given to taking samples from the
checked at intervals. Results should potentially coolest part of the load.
be recorded.
148
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
149
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
150
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Apart from the obvious problem of cleanable traps and with breaks
exposure of staff to infectious to prevent back-flow. The traps may
agents, potent toxins or allergens, it contain electrically operated heating
is necessary to avoid the risk of devices or other means for
contamination of a production batch disinfection. Any floor channels
with these agents. should be open, shallow and easily
cleanable and be connected to
10. Where BCG vaccines are being drains outside the area in a
manufactured, access to production manner that prevents ingress of
areas shall be restricted to staff microbial contaminants.
who are carefully monitored
by regular health checks. In the 13. Sinks shall be excluded from
case of manufacture of products aseptic areas. Any sink installed in
derived from human blood or other clean areas shall be of
plasma, vaccination of workers suitable material such as
against hepatitis is recommended. stainless steel, without an overflow,
and be supplied with water of
PREMISES AND EQUIPMENT potable quality. Adequate
precautions shall be taken to avoid
11. As a general principle, buildings contamination of the drainage
must be located, designed system with dangerous effluents.
constructed, adapted and Airborne dissemination of
maintained to suit the operations pathogenic microorganisms and
to be carried out within them. viruses used for production and the
Laboratories, operating rooms and possibility of contamination by
all other rooms and buildings other types of viruses or substances
(including those for animals) that during the production process,
are used for the manufacture including those from personnel,
of biological products shall be shall be avoided.
designed and constructed of
materials of the highest standard so 14. Lighting, heating, ventilation
that cleanliness, especially freedom and, if necessary, air-conditioning
form dust, insects and vermin, can should be designed to maintain a
be maintained. satisfactory temperature and relative
humidity, to minimize contamination
12. Interior surfaces (walls, floors and and to take account of the comfort
ceilings) shall be smooth and free of personnel working in protective
from cracks; they shall not shed clothing. Buildings shall be in a good
matter and shall permit easy state of repair. The condition of the
cleaning and disinfection. Drains buildings should be reviewed
shall be avoided whenever regularly and repairs carried out
possible and, unless essential, when and where necessary. Special
should be excluded from aseptic care should be exercised to ensure
areas. Where installed they should that building repair or maintenance
be fitted with effective, easily operations do not compromise
151
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
16. Seed lots and cell banks used for 21. All containers of biological
the production of biological substances, regardless of the stage
products should be stored of manufacture, shall be
separately from other material. identified by securely attached
Access should be restricted to labels. Cross-contamination should
authorized personnel. be prevented by adoption of some
or all of the following measures:
17. Live organisms shall be handled
in equipment that ensures that • processing and filling in segregated
cultures are maintained in a pure areas;
state and are not contaminated • avoiding manufacture of different
during processing. products at the same time, unless
they are effectively segregated;
152
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
153
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
154
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
155
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
27. The information given on the label Lot Processing Records (Protocols) And
on the container and the label on Distribution Records
the package shall be approved by
the national control authority. 30. Processing records of regular
production lots must provide a
28. The label on the container shall complete account of the
show:- manufacturing history of each lot of
a biological preparation, showing
• the name of the drug product; that it has been manufactured,
• a list of the active ingredients and tested, dispensed into containers
the amount of each present, with a and distributed in accordance with
statement of the net contents, e.g. the licensed procedures.
number of dosage units, weight
or volume; 31. A separate processing record
• the batch or final lot number should be prepared for each lot of
assigned by the manufacturer; biological product, and should
• the expiry date; include the following information:
• recommended storage conditions or
handling precautions that may • the name and dosage of
be necessary; the product;
• directions for use, and warnings and • the date of manufacture;
precautions that may be necessary; • the lot identification number;
• the nature and amount of any • the complete formulation of the
156
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
157
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
158
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
159
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
160
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
161
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
conditions under which they 33. The source of data for this validation
are produced should comply fully should include, but not be limited
with the requirements of Good to batch processing and packaging
Manufacturing Practice, including records, process control charts,
the satisfactory outcome of the maintenance log books, records of
validation exercise, and (where personnel changes, process
applicable) the marketing capability studies, finished product
authorization. data, including trend cards and
storage stability results.
Concurrent validation
34. Batches selected for retrospective
28. In exceptional circumstances it may validation should be representative
be acceptable not to complete of all batches made during the
a validation programme before review period, including any batches
routine production starts. that failed to meet specifications,
and should be sufficient in number
29. The decision to carry out concurrent to demonstrate process consistency.
validation must be justified, Additional testing of retained
documented and approved by samples may be needed to obtain
authorized personnel. the necessary amount or type of
data to retrospectively validate
30. Documentation requirements the process.
for concurrent validation are the
same as specified for prospective 35. For retrospective validation,
validation. generally data from ten to thirty
consecutive batches should be
Retrospective validation examined to assess process
consistency, but fewer batches may
31. Retrospective validation is only be examined if justified.
acceptable for well-established
processes and will be inappropriate CLEANING VALIDATION
where there have been recent
changes in the composition of the 36. Cleaning validation should be
product, operating procedures performed in order to confirm the
or equipment. effectiveness of a cleaning
procedure. The rationale for
32. Validation of such processes selecting limits of carryover of
should be based on historical data. product residues, cleaning agents
The steps involved require the and microbial contamination should
preparation of a specific protocol be logically based on the materials
and the reporting of the results of involved. The limits should be
the data review, leading to a achievable and verifiable.
conclusion and a recommendation.
37. Validated analytical methods having
sensitivity to detect residues or
162
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
163
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Definitions of terms relating to qualification The documented verification that the facilities,
and validation which are not given in the systems and equipment, as installed or
glossary of the current EAC-GMP Guide, but modified, comply with the approved design
which are used in this Annex, are given below. and the manufacturer’s recommendations.
The documented verification that the proposed Validation carried out before routine
design of the facilities, systems and equipment production of products intended for sale.
is suitable for the intended purpose.
164
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Retrospective Validation
Validation of a process for a product which has been marketed based upon accumulated
manufacturing, testing and control batch data.
Re-Validation
A repeat of the process validation to provide an assurance that changes in the process/equipment
introduced in accordance with change control procedures do not adversely affect process
characteristics and product quality.
Risk analysis
Method to assess and characterize the critical parameters in the functionality of an equipment or
process.
Simulated Product
A material that closely approximates the physical and, where practical, the chemical
characteristics (e.g. viscosity, particle size, pH etc.) of the product under validation. In many
cases, these characteristics may be satisfied by a placebo product batch.
System
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.
165
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
166
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
167
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
For example, information required 18. When outside agencies are used to
to effect a recall must be available provide a computer service, there
at short notice. should be a formal agreement
including a clear statement of the
16. The procedures to be followed if the responsibilities of that outside
system fails or breaks down should agency. (see chapter 8)
be defined and validated. Any
failures and remedial action taken 19. When the release of batches
should be recorded. for sale or supply is carried out
using a computerized system,
17. A procedure should be established the system should recognize that
to record and analyze errors and to only an Authorized Person can
enable corrective action to be taken. release the batches and it should
clearly identify and record the
person releasing the batches.
168
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
169
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
170
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
171
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
The chosen water purification method, or The design, configuration and layout of the
sequence of purification steps, must be water purification equipment, storage and
appropriate to the application in question. The distribution systems should also take into
following should be considered when selecting account the following physical considerations:
the water treatment method:
• the space available for the
• the water quality specification; installation;
• the yield or efficiency of the • structural loadings on buildings;
purification system; • the provision of adequate access for
• feed-water quality and the variation maintenance; and
over time (seasonal changes); • the ability to safely handle
• the reliability and robustness of the regeneration and sanitization
water-treatment equipment chemicals.
in operation;
• the availability of water-treatment 5.2 Production of drinking-water
equipment on the market;
• the ability to adequately support and Drinking-water is derived from a raw water
maintain the water purification source such as a well, river or reservoir. There
equipment; and are no prescribed methods for the treatment
• the operation costs. of raw water to produce potable drinking-water
from a specific raw water source.
The specifications for water purification
equipment, storage and distribution systems Typical processes employed at a user plant or
should take into account the following: by a water supply authority include:
172
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
The drinking-water quality should be with appropriately protected vents, allow for
monitored routinely. Additional testing should visual inspection and for being drained and
be considered if there is any change in the sanitized. Distribution pipe work should be
raw-water source, treatment techniques or able to be drained, or flushed, and sanitized.
system configuration. If the drinking water
quality changes significantly, the direct use Special care should be taken to control
of this water as a WPU, or as the feed-water microbiological contamination of sand filters,
to downstream treatment stages, should carbon beds and water softeners. Once
be reviewed and the result of the review microorganisms have infected a system, the
documented. contamination can rapidly form biofilms and
spread throughout the system. Techniques
Where drinking-water is derived from an for controlling contamination such as back-
“in-house” system for the treatment of raw flushing, chemical or thermal sanitization and
water, the water-treatment steps used and the frequent regeneration should be considered.
system configuration should be documented. Additionally, all water-treatment components
Changes to the system or its operation should be maintained with continuous water
should not be made until a review has been flow to inhibit microbial growth.
completed and the change approved by the
QA department. 5.3 Production of purified water
Where drinking-water is stored and distributed There are no prescribed methods for the
by the user, the storage systems must not production of PW in the pharmacopoeias. Any
allow degradation of the water quality before appropriate qualified purification technique
use. After any such storage, testing should or sequence of techniques may be used to
be carried out routinely in accordance with a prepare PW. Typically ion exchange, ultra
defined method. Where water is stored, its use filtration and/or reverse osmosis processes
should ensure a turnover of the stored water are used. Distillation can also be used.
sufficient to prevent stagnation. The drinking-
water system is usually considered to be an The following should be considered when
“indirect impact system” and does not need to configuring a water purification system:
be qualified.
• the feed-water quality and its
Drinking-water purchased in bulk and variation over seasons;
transported to the user by tanker presents • the required water-quality
special problems and risks not associated with specification;
potable water delivered by pipeline. Vendor • the sequence of purification stages
assessment and authorized certification required;
activities, including confirmation of the • the energy consumption;
acceptability of the delivery vehicle, should be • the extent of pretreatment required
undertaken in a similar way to that used for to protect the final purification steps;
any other starting material. • performance optimization, including
yield and efficiency of unit
Equipment and systems used to produce treatment- process steps;
drinking-water should be able to be drained
and sanitized. Storage tanks should be closed
173
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
There are no prescribed methods for The storage and distribution system should be
the production of HPW in any major considered as a key part of the whole system,
pharmacopoeia, including the European and should be designed to be fully integrated
Pharmacopoeia. Any appropriate qualified with the water purification components of the
purification technique or sequence of system.
techniques may be used to prepare HPW.
Typically ion exchange, ultrafiltration and/
or reverse osmosis processes are used. The
guidance provided in section 5.3 for PW is
equally applicable to HPW.
174
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Once water has been purified using an be compatible with the pipe work
appropriate method, it can either be used used. Appropriate sanitary-
directly or, more frequently, it will be fed into specification plastics and stainless
a storage vessel for subsequent distribution steel materials are acceptable for
to points of use. The following text describes WPU systems. When stainless
the requirements for storage and distribution steel is used it should be at
systems. least grade 316 L. The system
should be passivated after initial
The storage and distribution system should be installation or after modification.
configured to prevent recontamination of the When accelerated passivation is
water after treatment and be subjected to a undertaken, the system should be
combination of online and offline monitoring to thoroughly cleaned first, and the
ensure that the appropriate water specification passivation process should be
is maintained. under- taken in accordance with a
clearly defined documented
6.2 Materials that come into contact with procedure.
systems for water for pharmaceutical use
• Smooth internal finish. Once
This section applies to generation equipment water has been purified it is
for PW, HPW and WFI, and the associated susceptible to microbiological
storage and distribution systems. contamination, and the system is
subject to the formation of
The materials that come into contact with biofilms when cold storage and
WPU, including pipe work, valves and fittings, distribution is employed. Smooth
seals, diaphragms and instruments, should be internal surfaces help to avoid
selected to satisfy the following objectives. roughness and crevices within the
WPU system. Crevices are
• Compatibility. All materials used frequently sites where corrosion can
should be compatible with the commence. The internal finish
temperature and chemicals used by should have an arithmetical average
or in the system. surface roughness of not greater
than 0.8 micrometre arithmetical
• Prevention of leaching. All mean roughness (Ra). When
materials that come into contact stainless steel is used, mechanical
with WPU should be non-leaching at and electropolishing techniques
the range of working temperatures. may be employed. Electropolishing
improves the resistance of the
• Corrosion resistance. PW, HPW stainless steel material to surface
and WFI are highly corrosive. To corrosion.
prevent failure of the system and
contamination of the water, the • Jointing. The selected system
materials selected must be materials should be able to be
appropriate, the method of jointing easily jointed by welding in a
must be carefully controlled, and all controlled manner. The control of
fittings and components must the process should include as
175
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
176
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
177
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
an alternative approach whereby the utility • The system design should ensure
is main- trained and monitored at a lower the shortest possible length
pressure than the WPU may be considered. of pipework.
Where heat exchangers are used they should • For ambient temperature systems,
be arranged in continually circulating loops or pipework should be isolated from
sub-loops of the system to avoid unacceptable adjacent hot pipes.
static water in systems. • Deadlegs in the pipework
installation greater than 1.5 times
When the temperature is reduced for the branch diameter should
processing purposes, the reduction should be avoided.
occur for the minimum necessary time. The • Pressure gauges should be
cooling cycles and their duration should be separated from the system by
proven satisfactory during the qualification of membranes.
the system. • Hygienic pattern diaphragm valves
should be used.
6.5.2 Circulation pumps • Pipework should be laid to falls to
allow drainage.
Circulation pumps should be of a sanitary • The growth of microorganisms can
design with appropriate seals that prevent be inhibited by:
contamination of the system. Where stand-
by pumps are provided, they should be 1. ultraviolet radiation sources
configured or managed to avoid dead zones in pipework;
trapped within the system. 2. maintaining the system heated
(guidance temperature 70–80°C);
6.5.3 Biocontamination control techniques 3. sanitizing the system periodically
using hot water (guidance
The following control techniques may be used temperature >70°C);
alone or more commonly in combination. 4. sterilizing or sanitizing the system
periodically using superheated hot
• Maintenance of continuous turbulent water or clean steam; and
flow circulation within water distribu- 5. routine chemical sanitization using
tion systems reduces the ozone or other suitable chemical
propensity for the formation of agents. When chemical sanitization
biofilms. The main- tenance of the is used, it is essential to prove that
design velocity for a specific the agent has been removed prior
system should be proven during the to using the water. Ozone can
system qualification and the be effectively removed by using
maintenance of satisfactory ultraviolet radiation.
performance should be monitored.
During the operation of a distribution
system, short- term fluctuations
in the flow velocity are unlikely to
cause contamination problems
provided that cessation of flow, flow
reversal or pressure loss does
not occur.
178
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
179
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
180
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
181
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
182
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Figure 1: The guidelines address the various system criteria according to the
sequence set out in this diagram
183
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
3. Glossary As-built
The definitions given below apply to terms Condition where the installation is complete
used in these guidelines. They may have with all services connected and functioning
different meanings in other contexts but with no production equipment, materials or
personnel present.
Acceptance criteria
At-rest
Measurable terms under which a test result
will be considered acceptable. Condition where the installation is complete
with equipment installed and operating in a
Action limit manner agreed upon by the customer and
supplier, but with no personnel present.
The action limit is reached when the
acceptance criteria of a critical parameter Central air-conditioning unit (see air-
have been exceeded. Results outside handling unit)
these limits will require specified action and
investigation.
Change control
Air-handling unit (AHU)
A formal system by which qualified
The air-handling unit serves to condition the representatives of appropriate disciplines
air and provide the required air movement review proposed or actual changes that
within a facility. might affect a validated status. The intent is
to determine the need for action that would
Airlock ensure that the system is maintained in a
validated state.
An enclosed space with two or more doors,
which is interposed between two or more Clean area (clean room)
rooms, e.g. of differing classes of cleanliness,
for the purpose of controlling the airflow An area (or room) with defined environmental
between those rooms when they need to be control of particulate and micro- bial
entered. An airlock is designed for and used contamination, constructed and used in such a
by either people or goods (PAL, personnel way as to reduce the intro- duction, generation
airlock; MAL, material airlock). and retention of contaminants within the area.
The alert limit is reached when the normal Commissioning is the documented process of
operating range of a critical para- meter has verifying that the equipment and systems are
been exceeded, indicating that corrective installed according to specifications, placing
measures may need to be taken to prevent the equipment into active service and verifying
the action limit being reached. its proper action. Commissioning takes place
at the conclusion of project construction but
prior to validation.
184
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
A process or device to contain product, dust or A system that is expected to have a direct
contaminants in one zone, preventing it from impact on product quality. These systems are
escaping to another zone. designed and commissioned in line with good
engineering practice (GEP) and, in addition,
Contamination are subject to qualification practices.
185
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
186
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
The ratio of the actual water vapour pressure The documented act of proving that any
of the air to the saturated water vapour procedure, process, equipment, material,
pressure of the air at the same temperature activity or system actually leads to the
expressed as a percentage. More simply put, expected results.
it is the ratio of the mass of moisture in the
air, relative to the mass at 100% moisture Validation master plan (VMP)
saturation, at a given temperature.
VMP is a high-level document which
Standard operating procedure (SOP) establishes an umbrella validation plan for
the entire project, and is used as guidance
An authorized written procedure, giving by the project team for resource and
instructions for performing operations, not technical planning (also referred to as master
necessarily specific to a given product or qualification plan).
material, but of a more general nature (e.g.
operation of equipment, maintenance and 4. Protection
cleaning, validation, cleaning of premises
and environmental control, sampling and 4.1 Product and personnel
inspection). Certain SOPs may be used to
supplement product-specific master and batch 4.1.1 Areas for the manufacture of
production documentation. pharmaceuticals, where
pharmaceutical starting materials
Turbulent flow and products, utensils and
equipment are exposed to the
Turbulent flow, or non-unidirectional airflow, environment, should be classified as
is air distribution that is intro- duced into the “clean areas”.
controlled space and then mixes with room air
by means of induction. 4.1.2 The achievement of a particular
clean area classification depends on
Unidirectional airflow (UDAF) a number of criteria that should
be addressed at the design and
Unidirectional airflow is a rectified airflow over qualification stages. A suitable
the entire cross-sectional area of a clean balance between the different
zone with a steady velocity and approximately criteria will be required in order to
parallel streamlines(see also turbulent flow). create an efficient clean area.
(Modern standards no longer refer to laminar
flow, but have adopted the term unidirectional 4.1.3 Some of the basic criteria to be
airflow.) considered should include:
187
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
• location of air terminals and 4.1.6 Air change rates normally vary
directional airflow between 6 and 20 air changes per
• temperature hour and are normally determined
• humidity by the following considerations:
• material flow
• personnel flow • level of protection required
• equipment movement • the quality and filtration of the
• process being carried out supply air
• outside air conditions • particulates generated by the
• occupancy manufacturing process
• type of product. • particulates generated by
the operators
4.1.4 Air filtration and air change rates • configuration of the room and air
should ensure that the defined clean supply and extract locations
area classification is attained. • sufficient air to achieve containment
effect
4.1.5 The air change rates should • sufficient air to cope with the room
be determined by the manufacturer heat load
and designer, taking into account • sufficient air to maintain the required
the various critical parameters. room pressure.
Primarily the air change rate should
be set to a level that will achieve the 4.1.7 In classifying the environment, the
required clean area classification. manufacturer should state whether
this is achieved under “as-built” (Fig.
2), “at-rest” (Fig. 3) or “operational”
(Fig. 4) conditions.
188
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
189
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.1.8 Room classification tests in the “as- 4.1.11 Materials and products should
built” condition should be carried be protected from contamination
out on the bare room, in the and cross-contamination during
absence of any equipment all stages of manufacture (see
or personnel. also section 5.5 for cross-
contamination control). Note:
4.1.9 Room classification tests in the contaminants may result from
“at-rest” condition should be carried inappropriate premises (e.g. poor
out with the equipment operating design, layout or finishing), poor
where relevant, but without any cleaning procedures, contaminants
operators. Because of the amounts brought in by personnel, and a poor
of dust usually generated in a solid HVAC system.
dosage facility most clean area
classifications are rated for the “at- 4.1.12 Airborne contaminants should be
rest” condition. controlled through effective
ventilation.
4.1.10 Room classification tests in the
“operational” condition should be 4.1.13 External contaminants should be
carried out during the normal removed by effective filtration of
production process with equipment the supply air. (See Fig. 5 for an
operating, and the normal number example of a shell-like building
of personnel present in the layout to enhance containment and
room. Generally a room that is protection from external
tested for an “operational” condition contaminants.)
should be able to be cleaned up to
the “at-rest” clean area classification
after a short clean-up time. The
clean-up time should be determined
through validation and is generally
of the order of 20 minutes.
190
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Note: The process core is regarded as the most stringently controlled clean zone which is
protected by being surrounded by clean areas of a lower classification.
191
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
192
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Level 2 Protected Area in which steps are taken to protect the exposed
pharmaceutical starting material or product from
contamination or degradation, e.g. manufacturing,
primary packing, dispensing
193
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Note: The filter classifications referred to above relate to the EN1822 and EN779 test standards
(EN 779 relates to filter classes G1 to F9 and EN 1822 relates to filter classes H10 to U16).
4.2.2 Filter classes should always be linked to the standard test method because
referring to actual filter efficiencies can be very misleading (as different test
methods each result in a different value for the same filter) (Fig. 8).
4.2.3 In selecting filters, the manufacturer should have considered other factors,
such as particularly contaminated ambient conditions, local regulations
and specific product requirements. Good pre-filtration extends the life of the
more expensive filters downstream.
4.2.4 Materials for components of an HVAC system should be selected with care
so that they do not become the source of contamination. Any component with
the potential for liberating particulate or microbial contamination into the air
stream should be located upstream of the final filters.
4.2.5 Ventilation dampers, filters and other services should be designed and
positioned so that they are accessible from outside the manufacturing areas
(service voids or service corridors) for maintenance purposes.
194
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
This figure gives a rough comparison between the different filter standards (filter classes should
always be connected to the standard test method).
195
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
196
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
197
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.3.3 Sampling of materials such 4.3.6 The source of the dust and the
as starting materials, primary position in which the operator
packaging materials and normally stands should be
products, should be carried out determined before deciding on the
in the same environmental direction of unidirectional flow.
conditions that are required for the
further processing of the product.
Example: In Fig. 12 the dust generated at
4.3.4 In a weighing booth situation, the the weighing station is immediately extracted
aim of the design using UDAF through the perforated worktop, thus
should be to provide dust protecting the operator from dust inhalation,
containment. but at the same time protecting the product
from contamination by the operator by means
of the vertical unidirectional airflow stream.
198
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.3.7 The unidirectional flow velocity Fig. 16 shows that a solid worktop can
should be such that it does not sometimes cause deflection of the vertical
disrupt the sensitivity of balances in unidirectional airflow resulting in a flow
weighing areas. Where necessary reversal. A possible solution would be to have
the velocity may be reduced a 100 mm gap between the back of the table
to prevent inaccuracies during and the wall, with the air being extracted here.
weighing, provided that sufficient
airflow is maintained to provide 4.3.11 The manufacturer should select
containment. either vertical or horizontal
unidirectional flow (Fig. 17) and an
4.3.8 The position in which the operator appropriate airflow pattern to
stands relative to the source of dust provide the best protection for the
liberation and airflow should be particular application.
determined to ensure that the
operator is not in the path of an 4.4 Infiltration
airflow that could lead to
contamination of the product 4.4.1 Air infiltration of unfiltered air into a
(Fig. 13). pharmaceutical plant should not be
the source of contamination.
4.3.9 Once the system has been
designed and qualified with a 4.4.2 Manufacturing facilities should
specific layout for operators and be maintained at a positive pressure
processes, this should be relative to the outside, to limit the
maintained in accordance with ingress of contaminants. Where
an SOP. facilities are to be maintained at
negative pressures relative to the
4.3.10 There should be no obstructions in ambient pressure to prevent the
the path of a unidirectional flow air escape of harmful products to the
stream that may cause the operator outside (such as penicillin and
to be exposed to dust. hormones), special precautions
should be taken.
Fig. 14 illustrates the incorrect use of a
weighing scale which has a solid back. The 4.4.3 The location of the negative
back of the weighing scale should not block pressure facility should be carefully
the return air path as this causes air to rise considered with reference to the
vertically, resulting in a hazardous situation for areas surrounding it, particular
the operator. attention being given to ensuring
that the building structure is
Fig. 15 illustrates a situation where an open well sealed.
bin is placed below a vertical unidirectional
flow distributor. The downward airflow should
be prevented from entering the bin, and then
being forced to rise again, as this would carry
dust up towards the operator’s face.
199
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
200
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
201
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
202
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.4.4 Negative pressure zones should, 4.5.6 Highly potent products should be
as far as possible, be encapsulated manufactured under a pressure
by sur- rounding areas with clean cascade regime that is negative
air supplies, so that only clean air relative to atmospheric pressure.
can infiltrate into the controlled
zone. 4.5.7 The pressure cascade for each
facility should be individually
4.5 Cross-contamination assessed according to the product
handled and level of protection
4.5.1 Where different products are required.
manufactured at the same time, in
different areas or cubicles, in 4.5.8 Building structure should be
a multiproduct OSD manufacturing given special attention to
site, measures should be taken accommodate the pressure cascade
to ensure that dust cannot move design.
from one cubicle to another.
4.5.9 Airtight ceilings and walls, close
4.5.2 Correct directional air movement fitting doors and sealed light fittings
and a pressure cascade system should be in place.
can assist in preventing cross-
contamination. The pressure Displacement concept (low pressure
cascade should be such that differential, high airflow)
the direction of airflow is from the
clean corridor into the cubicles, Note: This method of containment is not
resulting in dust containment. the preferred method, as the measurement
and monitoring of airflow velocities in
4.5.3 The corridor should be maintained doorways is difficult. This concept should
at a higher pressure than the ideally be applied in production processes
cubicles, and the cubicles at a where large amounts of dust are generated.
higher pressure than atmospheric
pressure. 4.5.10 Under this concept the air should
be supplied to the corridor, flow
4.5.4 Containment can normally be through the doorway, and be
achieved by application of extracted from the back of the
the displacement concept (low cubicle. Normally the cubicle door
pressure differential, high airflow), should be closed and the air
or the pressure differential concept should enter the cubicle through
(high pressure differential, low a door grille, although the concept
airflow), or the physical barrier can be applied to an opening
concept. without a door.
4.5.5 The pressure cascade regime and 4.5.11 The velocity should be high enough
the direction of airflow should be to prevent turbulence within the
appropriate to the product and doorway resulting in dust escaping.
processing method used.
203
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.5.12 This displacement airflow should be 4.5.16 The pressure differential between
calculated as the product of the door adjacent rooms could be considered
area and the velocity, which a critical parameter, depending on
generally results in fairly large the outcome of risk analysis. The
air quantities. limits for the pressure differential
between adjacent areas should be
Pressure differential concept (high such that there is no risk of overlap,
pressure differential, low airflow) e.g. 5 Pa to 15 Pa in one room and
15 Pa to 30 Pa in an adjacent room,
Note: The pressure differential concept may resulting in no pressure cascade, if
normally be used in zones where little or no the first room is at the maximum
dust is being generated. It may be used alone tolerance and the second room is
or in combination with other containment at the minimum tolerance.
control techniques and concepts, such as a
double door airlock. 4.5.17 Low pressure differentials may be
acceptable when airlocks (pressure
4.5.13 The high pressure differential sinks or pressure bubbles) are used.
between the clean and less clean
zones should be generated by 4.5.18 The effect of room pressure
leakage through the gaps of the tolerances are illustrated in Fig. 18.
closed doors to the cubicle.
4.5.19 The pressure control and monitoring
4.5.14 The pressure differential should be devices used should be calibrated
of sufficient magnitude to ensure and qualified. Compliance with
containment and prevention of specifications should be regularly
flow reversal, but should not be so verified and the results recorded.
high as to create turbulence Pressure control devices should be
problems. linked to an alarm system set
according to the levels determined
4.5.15 In considering room pressure by a risk analysis.
differentials, transient variations,
such as machine extract systems,
should be taken into consideration.
204
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.5.20 Manual control systems, where used, should be set up during commissioning and
should not change unless other system conditions change.
4.5.21 Airlocks can be important components in setting up and maintaining pressure cascade
systems.
4.5.22 Airlocks with different pressure cascade regimes include the cascade airlock, sink
airlock and bubble airlock (Figs 19–21).
• Cascade airlock: high pressure on one side of the airlock and low pressure on the other.
• Sink airlock: low pressure inside the airlock and high pressure on both outer sides.
• Bubble airlock: high pressure inside the airlock and low pressure on both outer sides.
4.5.23 Doors should open to the high pressure side, and be provided with self- closers. Door
closer springs, if used, should be designed to hold the door closed and prevent the
pressure differential from pushing the door open. Sliding doors are not recommended.
4.5.24 Central dust extraction systems should be interlocked with the appropriate air handling
systems, to ensure that they operate simultaneously.
205
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
206
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.5.25 Room pressure imbalance between 4.5.28 Spot ventilation or capture hoods
adjacent cubicles which are linked may be used as appropriate.
by common dust extraction ducting
should be prevented. 4.6 Temperature and relative
humidity
4.5.26 Air should not flow from the room
with the higher pressure to the room 4.6.1 Temperature and relative humidity
with the lower pressure, via the dust should be controlled, monitored
extract ducting (this would normally and recorded, where relevant,
occur only if the dust extraction to ensure compliance with
system was inoperative). requirements pertinent to the
materials and products, and to
Physical barrier concept provide a comfortable environment
for the operator where necessary.
4.5.27 Where appropriate, an impervious
barrier to prevent cross- 4.6.2 Maximum and minimum room
contamination between two zones, temperatures and relative humidity
such as barrier isolators or pumped should be appropriate.
transfer of materials, should
be used. 4.6.3 Temperature conditions should be
207
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
208
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
209
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
5.10 When planning the system for 6.1 Dust in exhaust air
the extraction of vapours, the
density of the vapour should be 6.1.1 Exhaust air discharge points on
taken into account. If the vapour is pharmaceutical equipment and
lighter than air, the extract grilles facilities, such as from fluid bed
should be at a high level, or possibly driers and tablet-coating equipment,
at both high and low levels. and exhaust air from dust extraction
systems, carry heavy dust loads and
5.11 When dealing with particularly should be provided with adequate
harmful products, additional steps, filtration to prevent contamination of
such as handling the products the ambient air.
in glove boxes or using barrier
isolator technology, should be used.
210
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
211
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
6.1.14 When wet scrubbers are used, the 6.2.5 The type and quantity of the
dust-slurry should be removed by a vapours to be removed should be
suitable drainage system. known to enable the appropriate
filter media, as well as the volume of
6.1.15 The quality of the exhaust air should media required to be determined.
be determined to see whether the
filtration efficiency is adequate with 7. HVAC systems and components
all types of dust collectors and
wet scrubbers. Note: The required degree of air cleanliness
in most OSD manufacturing facilities can
6.1.16 Where necessary, additional normally be achieved without the use of
filtration may be provided high-efficiency particulate air (HEPA) filters
downstream of the dust collector. provided the air is not re-circulated. Many
open product zones of OSD form facilities
6.2 Fume removal are capable of meeting ISO 14644-1 Class 8,
“at-rest” condition, measured against particle
6.2.1 The systems for fume, dust and sizes of 0.5 cm and 5 cm, but cleanliness may
effluent control should be designed, not be classified as such by manufacturers.
installed and operated in such a
manner that they do not become
possible sources of contamination
or cross- contamination, e.g. an
exhaust-air discharge point located
close to the HVAC system fresh
air inlet.
212
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
213
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
7.1.5 Possible additional components The airflow schematics of the two systems
that may be required should (Figs 24 and 25) indicate air-handling unit
be considered depending on the with return air or re-circulated air, having a
climatic conditions and locations. percentage of fresh air added. Fig. 25 is a
These may include items such as: schematic diagram of an air-handling system
serving rooms with horizontal unidirectional
• frost coils on fresh air inlets in very flow, vertical unidirectional flow and turbulent
cold climates to preheat the air; flow, for rooms A, B and C, respectively.
• snow eliminators to prevent snow
entering air inlets and blocking The airflow diagram in Fig. 24 is an example
airflow; of a typical system with a lower clean area
• dust eliminators on air inlets in arid classification.
and dusty locations;
• moisture eliminators in humid areas Note: There are two basic concepts of air
with high rainfall; and delivery to pharmaceutical production facilities:
• fresh air pre-cooling coils for very a recirculation system, and a full fresh air
hot or humid climates. system (100% outside air supply).
214
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Figure 24. Air-handling system with high-efficiency particulate air filters in air-
handling unit
215
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
7.2.3 HEPA filters may not be required with toxic products, where
where the air-handling system is recirculation of air with contaminants
serving a single product facility and should be avoided.
there is evidence that cross-
contamination would not 7.2.8 The required degree of filtration
be possible. of the exhaust air depends on
the exhaust air contaminants and
7.2.4 Recirculation of air from areas local environmental regulations.
where pharmaceutical dust is not
generated such as secondary 7.2.9 Energy-recovery wheels should
packing may not require HEPA normally not be used in multiproduct
filters in the system. facilities. When such wheels are
used they should not become
7.2.5 HEPA filters may be located in the a source of possible contamination
air-handling unit or placed (see Fig. 27).
terminally.
Note: Alternatives to the energy-
7.2.6 Air containing dust from highly toxic recovery wheels, such as crossover
processes should never be re- plate heat exchangers and
circulated to the HVAC system. water-coil heat exchangers, may be
used in multiproduct facilities.
7.2.7 Full fresh-air systems
Fig. 26 indicates a system operating
on 100% fresh air and would
normally be used in a facility dealing
216
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
7.2.10 The potential for air leakage in the user requirement specification
between the supply air and exhaust (URS), and capacities as specified
air as it passes through the wheel by the designer or developer.
should be prevented. The relative
pressures between supply and 8.1.2 The installation records of the
exhaust air systems should be such system should provide documented
that the exhaust air system operates evidence of all measured capacities
at a lower pressure than the supply of the system.
system.
8.1 Commissioning
217
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
218
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
219
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
8.2.14 Out-of-limit results (e.g. action limit 8.2.16 A narrow range of relative humidities
deviations) should be recorded and coupled with a wide range of
form part of the batch manufacturing temperatures is unacceptable as
records. changes in temperature will
automatically give rise to variations
8.2.15 The relationships between design in the relative humidity.
conditions, operating range and
qualified acceptance criteria are
given in Fig. 29.
220
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
221
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
8.2.18 The maximum time interval between 8.2.21 Clean-up or recovery times
tests should be defined by the normally relate to the time it takes
manufacturer. The type of facility to “clean up” the room from one
under test and the product level of condition to another, e.g.
protection should be considered. the relationship between “at- rest”
and “operational” conditions in the
Note: Table 3 gives intervals clean area may be used as the
for reference purposes only. The criteria for clean-up tests. Therefore,
actual test periods may be more the clean-up time can be expressed
frequent or less frequent, depending as the time taken to change from an
on the product and process. “operational” condition to an “at
rest” condition.
8.2.19 Periodic requalification of
parameters should be done at
regular intervals, e.g. annually.
222
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
223
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
224
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
225
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
7.6 The drug regulatory authority should all relevant requirements. Therefore
approve the authorized person on it is required that the manufacturer
the basis of his or her professional establishes and maintains a
curriculum vitae. Authorized persons comprehensive quality management
have duties not only to their system, covering all aspects
employer but also to the competent of GMP.
authorities such as the drug
regulatory authority. They should 7.9 The Authorized person must ensure
establish good working relations that there is a quality manual
with inspectors and as far as describing the quality policy and
possible provide information on objectives (commitment to quality)
request during site inspections. of the company, the organizational
structure, responsibilities and
7.7 The authorized person depends authorities, together with a
upon many working colleagues for description of or references to
the achievement of quality documented quality system
objectives, and may delegate some procedures.
duties to appropriately trained staff
while remaining the overall quality 7.10 The Authorized person must
controller. It is therefore of ensure that Research and
paramount importance that he or development activities and the
she establish and maintain a good transfer of results of the
working relationship with other developmental work to routine
persons in positions of responsibility, manufacture, including original
especially those responsible for product design, formulation,
production and quality control. processes development and
validation, should observe GMP
Implementation of the quality system principles as guidance. Batches
produced for clinical trials must
7.8 Authorized persons have a personal follow applicable GMP. It is of
and professional responsibility for vital importance that the quality of
ensuring that each batch of routine production batches
finished products has been corresponds to a specification
manufactured in accordance with derived from the composition of
the marketing authorization, GMP development batches. The quality
rules and all related legal and and safety of a pharmaceutical
administrative provisions. This product depend on the application
does not necessarily mean that of appropriate procedures, based
they must have directly supervised on GMP, leading to a product
all manufacturing and quality control within the recognized specification.
operations. They must be Standard procedures and
satisfied either directly or, more recognized specifications cannot
usually, by proper operation of be separated.
quality systems that manufacturing
and testing have complied with
226
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
227
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
228
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
229
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
230
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
231
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Taken from reference 6: ICH Q9: Quality Risk Management. This figure is
also available on the ICH website www.ich.org
Decision points are not shown in the diagram The approach described in this guideline
above because decisions can occur at any should be used to:
point in the process. These decisions might be
to return to the previous step and seek further • systematically analyze products
information, to adjust the risk models or even and processes to ensure the best
to terminate the risk management process scientific rationale is in place to
based upon information that supports such improve the probability of success;
a decision.
• identify important knowledge gaps
Note: “unacceptable” in the flowchart does not associated with processes that need
only refer to statutory, legislative or regulatory to be understood to properly identify
requirements, but also indicates that the risk risks;
assessment process should be revisited.
232
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
233
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Possible steps used to initiate and plan a The objectives and scope of the QRM
QRM process might include the following (Ref. activities should be clearly defined. The scope
ICH Q9): should describe the segment of the process
involved.
• define the problem and/or risk
question, including pertinent 2.3 Knowledge of the product
assumptions identifying the potential and process
for risk;
• assemble background information Any activity of QRM would need to be based
and/or data on the potential hazard, on knowledge of the product or processes
harm or human health impact concerned, according to the stage of the
relevant to the risk assessment; product life-cycle.
• identify a leader and necessary
resources; and Where necessary, a flow diagram may be
• specify a timeline, deliverables and helpful, covering all operations and controls
appropriate level of decision-making in the process under evaluation. When
for the risk management process. applying QRM to a given operation, the steps
preceding and following that operation should
2.2 Personnel involved in QRM also be considered. A block-type diagram may
be sufficiently descriptive. Amendments to the
The implementing party, i.e. pharmaceutical flow diagram may be made where appropriate,
manufacturer or regulatory authority, should and should be documented.
assure that personnel with appropriate
product-specific knowledge and expertise 2.4 Risk assessment
are available to ensure effective planning
and completion of QRM activities. This may When risk assessment is conducted safety
be best accomplished by assembling a and efficacy need to be considered in addition
multidisciplinary team according to guidance to the quality concerns.
in section 3.2.
During the assessment all the risks that may
The personnel should be able to: be reasonably expected to occur in the activity
under evaluation should be listed. This is
(a) conduct a risk analysis; usually applied during its initiation when there
(b) identify and analyse potential risks; is a change or a concern and may also be
(c) identify, evaluate risks and applied to existing processes. An analysis
determine which ones should should be conducted to identify which risks
be controlled and which ones can are of such a nature that their elimination or
be accepted; reduction to acceptable levels is essential.
(d) recommend and implement
adequate risk control measures; A thorough risk analysis is required to
(e) devise procedures for risk review, ensure an effective risk control. It should
monitoring and verification. review the materials, activities, equipment,
storage, distribution and intended use of the
product. Typically a list of the potential risks
(biological, chemical and physical) which
234
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Risk assessment can be facilitated by the use The expectation of QRM is to assess risks to
of a decision-tree, which facilitates a logical the product quality and to the patient and then
approach. The way that a decision-tree is manage these risks to an acceptable level. It
used will depend on the operation concerned, is appropriate for companies to assess their
e.g. production, packing, reprocessing, control systems to implement the optimum
storage or distribution. The best use of QRM controls to ensure product quality and patient
tools is discussed further in section 5 of this safety. Risk assessment and mitigation in
guidance. order to achieve cost savings but which
could be to the detriment of the patient is an
Normally, potential risks in relation to the unacceptable practice (10).
following should be considered:
2.5 Risk control
• materials and ingredients;
• physical characteristics and Risk control is a decision-making activity
composition of the product; designed to reduce and/or accept risks. It
• processing procedures; usually occurs after risk assessment, and at a
235
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
fundamental level its purpose is to reduce the occurs, who is responsible for implementing
risk to an acceptable level. the corrective actions, and that a record will be
kept and maintained of the actions taken.
During risk control activities the following key
questions should be asked: 2.6 Risk review
236
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
(c) confirmation that identified risks is Individuals doing verification should have
kept under control. appropriate technical expertise to perform this
function.
Initial verification of the planned QRM
activities is necessary to determine whether it 2.8 Risk communication and
is scientifically and technically sound, that all documentation
risks have been identified and that, if the QRM
activities are properly completed, these risks Communication of the QRM process should
will be effectively controlled. include key stakeholders. By ensuring that key
stakeholders are engaged in both the data
Information reviewed to verify the QRM collection process for the risk assessment
process should include: and the decision-making for risk control,
this will ensure commitment and support for
(a) expert advice and scientific studies; the QRM. The output of the QRM process
(b) in-plant observations, and associated risk analysis justifying
measurements and evaluations. the approach should be documented and
endorsed by the organization’s quality unit and
Subsequent verifications should be performed management. Additionally, this information
and documented by a QRM team or an should be communicated to stakeholders for
independent expert, as needed. For example, their information and to ensure their support.
verifications may be conducted when there is
an unexplained system failure, a significant There should be a report for every risk
change in product, process or packaging assessment, but the level of effort, formality
occurs or new risks are recognized. Where and documentation will commensurate with
possible verification should include actions the level of risk (2).
to confirm the efficacy of all elements of the
QRM activities. Regarding conclusions to a risk assessment
the mitigation controls should minimize
In addition, a comprehensive review of the the likelihood of risk to patient safety to
QRM process and specific instances of an acceptable level of assurance, on the
QRM application by an independent third understanding that no risk whatsoever is
party may be useful. This would include a unlikely in reality. The degree of risk tolerated
technical evaluation of the risk analysis and very much depends on the circumstances,
each element of the QRM process and its the proximity to the patient and other controls
application as well as an on-site review of that might follow the process being assessed
all flow diagrams and appropriate records before the product reaches the patient (2).
of the operation of the QRM activity. Such a
comprehensive verification is independent of It is expected that risk mitigation plans are
other verification procedures and should be identified and implemented where any risk to
performed in order to ensure that the QRM patient safety is posed. Companies should
process is resulting in the control of the risks. take the holistic view and be mindful that
If the results of the comprehensive verification critical issues often arise where multiple
identify deficiencies the QRM process should failures in systems occur together so
be modified as necessary. mitigation plans should be sufficiently robust
to cover this scenario. Inspectors will assess
237
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
238
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
expertise or knowledge. Their role should be complete. For this stage of a project there may
justifiable and clearly defined and resultant be significant gaps in knowledge. Therefore,
accountability must be understood. A technical it will be important to apply risk tools that are
agreement or other equivalent document with appropriate for such a situation.
the consultant may be appropriate where a
GMP responsibility is assumed. These might include:
Similarly, contract staff may become involved • cause and effect diagrams (also
to lead or participate in risk assessments, known as Ishikawa or Fishbone
e.g. a contract authorized person. The diagrams);
extent of involvement and responsibility/ • flowcharts (e.g. input-process-output
accountability must be documented in a (IPO));
technical agreement or other equivalent • decision-trees;
document between the individual and the • fault-tree analysis; and
pharmaceutical company. Regarding the • relationship matrices.
authorized person it is important that a
company’s internal procedures are clear on As the product progresses to later stage
where the responsibility lies for final approval development, a more detailed analysis of
of risk acceptance documents. the risks associated with both the API and
FPP becomes a requirement. Risks would
Effective matrix team leadership is required cover concerns associated with stability,
to take responsibility for coordinating QRM bioavailability and patient safety including
across various functions and departments of any challenges to these resulting from
their organization and ensuring that the QRM the manufacturing process (including, for
activities are adequately defined, planned, example, API form conversion under certain
resourced, deployed and reviewed. The conditions of processing).
leader and team will need to identify critical
resources to progress the QRM activities, As product knowledge advances more
and also specify a timeline, deliverables and detailed QRM exercises can be considered,
appropriate levels of decision-making for the concentrating on areas considered to be
QRM process. higher priority risk. As the product’s critical
quality attributes (CQAs) become defined,
3.3 QRM application during product the potential risks arising from each input
development material (API, excipients, any device or pack
components) and each secondary product unit
The application of QRM procedures evolves operation can be investigated.
through the various stages in development of
a product. Eventually, for the developed FPP the
increasingly comprehensive risk assessment
It is important, where possible, to identify risks will support a thorough understanding of the
in the early phases of product development product and will enable all key variables to be
that could challenge the achievement of the identified, understood and controlled.
QTPP. The first QRM exercise should be
performed once the QTPP is defined and
preformulation work on the drug candidate is
239
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
3.4 QRM application during that needs to be made depends on the depth
validation and qualification of knowledge about the process.
In keeping with the principles of QRM, For very low-volume products, e.g. orphan
this guideline recommends that process drugs, this may preclude the need to
validation embraces the product life cycle manufacture multiple batches. It would
concept already mentioned. Accordingly, be beneficial for decisions of this nature
process validation activities should involve the regarding conformance batches to have an
generation and evaluation of data throughout effective company/MRA dialogue to agree
the development process into full-scale on requirements for a regulatory submission.
production that will provide a science-based Until new approaches to demonstrate
assurance of consistent delivery of quality validation mature and become widely used,
product in the production operation the traditional three-batch approach to validate
(10, 11, 12). a process is still acceptable. When applicable
the principles of QRM should also be applied
An important emphasis is that the building for qualification activities.
of scientific assurance begins early in
development. It is obtained through rational Qualification includes four stages (design
design of experiments and robust evaluation qualification (DQ), installation qualification
of data during product/process development (IQ), operational qualification (OQ) and
through to the commercial production phase performance qualification (PQ)) but most
at which time the API and drug product frequently, only IQ, OQ and PQ are performed
CQAs are well understood and controlled. by manufacturers. QRM principles can be
In this scenario, validation or (perhaps used to narrow the scope of IQ, OQ and
more appropriately termed) conformance PQ to cover only the essential elements
batches just serve to reinforce the science- or that can affect product quality. It can also
risk-based decisions that have been made be used to determine the optimal schedule
as product development has advanced for maintenance, monitoring, calibration and
and should demonstrate good control of requalification.
all identified critical sources of variability.
Any unplanned variations within a batch Most importantly, by the time that a
or between batches should be evaluated product is ready for commercialization, the
accordingly, employing suitable statistical manufacturing company will be expected
tools, e.g. trend analysis, to check on process to have derived sufficient knowledge of the
control. commercial production process to support that
commercialization to the optimized benefit of
A potential advantage of this approach is and minimized risk to the patient.
that there can be flexibility in the number of
validation or conformance batches required 3.5 QRM application during
for regulatory scrutiny prior to approval. The commercial manufacturing
traditional number of batches required for
validation has been three but, with QRM QRM principles applied as a process supports
embedded in a product’s development science-based and practical decisions when
process, the number of conformance batches integrated into commercial manufacturing.
In general implementing QRM should not
240
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
obviate a manufacturer’s obligation to comply Risk review may also include evaluation of,
with regulatory expectations (e.g. regulatory e.g:
requirements, regulatory filings, inspection
commitments, etc.). All QRM activities should • effectiveness of risk control activities
be structured in a way that allows escalation and actions;
of risks to the appropriate management level • changes in observed risk levels or
within the organization. existing controls.
Special focus can be on the risk assessment In principal there are two focuses when
and risk control of, e.g: implementing QRM in commercial
manufacturing: a system focus and a
• product quality risks; product focus.
• adverse impact to patient health
based on product quality defects; 3.5.1 QRM integration with key quality
• product supply interruption to system elements
patients;
• GMP and regulatory compliance Effective QRM can facilitate the “What to
risks; do?” and, therefore, support better and more
• multisite risks; informed decisions. QRM should be integrated
• multiproduct risks; into existing quality system elements and
• new facility and changes to existing related business processes and documented
facility, e.g. start-ups, new appropriately.
commercial manufacturing
processes, technology transfers and Situations in which the use of the QRM
product discontinuation. process might provide information are
beneficial in a variety of operations, e.g:
After completion of the risk assessment and
risk control activities the outcomes must be • integrated quality
summarized and communicated. The results management:documentation;
may be documented in a new or existing training and education; quality
report or they may be included as part of defects; auditing/inspection; change
another document approved by appropriate management/change control
decision-makers (e.g. site or functional (includes equipment, facilities,
management, system owner, quality unit, utilities, control and IT systems);
etc.). A risk review is important if new risks or continual improvement/corrective
changes to existing risk levels are identified and preventive actions (CAPA);
through planned or unplanned events such
as routine operation, changes, complaints, • facilities, equipment and utilities:
product returns, discrepancies/deviations, e.g. design; qualification;
data monitoring, trends, inspections/audits, maintenance and decommissioning
changes in regulatory environment, etc. of facility/equipment; hygiene
aspects; cleaning of equipment and
environmental control; calibration/
preventive maintenance; computer
241
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
242
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
including (but not limited to) inspection Various factors should be considered in
planning, review of corrective and preventive the risk assessment exercise, and may
actions after inspections and complaint be different for the different types of GXP
handling and investigation. Where appropriate, inspections.
the procedures and activities during inspection
should be in line with the principles of QRM. Risk factors to be considered depend on the
type of inspection, and may include:
The unit should have a risk management
plan (RMP) that describes the philosophy, (a) outcome of inspection by another
approach, procedures and implementation of regulatory authority;
risk management. The risk management plan (b) outcome of the previous inspection;
should be reviewed and updated on a rolling (c) complexity of the site (e.g. buildings,
basis, or at least annually and should cover utilities);
all types of inspections (including GMP, GCP, (d) complexity of the product(e.g.
GLP) and other activities. sterile, non-sterile);
(e) type of product (e.g. biological,
Appropriate risk assessment tools should be low dose);
used in the process, and the risk assessment (f) complaints and recalls;
for a site to be inspected should be (g) significance of changes (e.g.
documented in a risk assessment worksheet. equipment, key personnel);
Records should be maintained. (h) results of product testing;
(i) risk to the patient;
A metric system should be used for risk (j) complex route of synthesis (API);
ratings, e.g. on a scale from 1 to 3. (k) polymorphism (API);
(l) biopharmaceutical classification of
4.2.2 Inspection planning and conduct the product;
The frequency and scope of inspections The number of inspectors and number of days
should be determined based on risk required for the inspection, as well as the
assessment that covers product risk and scope of the inspection, should be determined
patient risk. based on the risk rating of the site inspection.
Risk rating should normally be done only for Inspection reports should contain findings and
sites that had been previously inspected. observations. Departures from GXP should be
The risk assessment worksheet should be classified where appropriate, as “critical, major
completed after every inspection. Inspection or minor”.
of a site that had not been inspected
previously may be waived only in cases The unit should have an SOP that describes
where a recognition procedure exists between the classification process. Classification
regulatory inspection units, and where should be based on risk assessment. The
in addition appropriate evidence of GXP level of risk assigned should be in relation to
compliance is available that indicates that the nature of the observation as well as the
there is no or acceptable low risk to products number of occurrences.
and patients.
243
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.2.3 Corrective action and preventive The company’s QRM procedure should
action review, and scheduling of be appropriately detailed and should be
routine inspections integrated into the company’s quality
management system.
Corrective actions and preventive actions
(CAPA) should be requested from a site, It should cover at least the following areas:
following an inspection. The CAPAs should
address the observations included in an (a) general approach to both planned
inspection report. and unplanned risk assessment –
and include scope, responsibilities,
Based on the inspection outcome and the controls, approvals, management
acceptability of the CAPA, the risk rating of the systems, applicability and
site should be reviewed and recorded. exclusions;
(b) personnel with appropriate
Inspection frequency should be defined based qualifications, experience and
on the risk rating. For example, a frequency training. Their responsibilities with
can be defined as every 6, 12, 18 or 24 regard to QRM should be clearly
months. (The maximum time interval should defined;
be no more than every 36 months.) (c) senior management should
be involved in the identification and
4.2.4 Complaint handling and implementation of QRM principles
investigation within the company;
(d) the risk management procedure(s)
Handling and investigation of quality for each area of application should
complaints should be done in accordance with be clearly defined;
a written SOP. (e) quality assurance principles should
be applied to QRM-related
The scope and depth of the investigation documentation, e.g. review,
(including whether a desk review or on-site approval, implementation and
inspection will be done) should be based on archiving.
risk assessment.
QRM policies and procedures should be clear
4.3 Inspection of QRM at a and the workflow should be systematic and
manufacturing site conducted in a logical order.
The procedure for risk management should be
Note. During inspections, inspectors implemented. Manufacturers should identify
should assess whether a manufacturer has significant risks and consider all the relevant
appropriate skills, scientific knowledge as well data from reliable sources.
as product and process knowledge for the
QRM procedure being inspected. This is also Personnel should be trained and assessed in
relevant where a company has made use of the principles of QRM.
contracted parties.
Where appropriate, a team of members of
personnel should participate in the QRM
processes.
244
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
The level of effort and resources used in risk the prioritization of dossiers, the screening
assessment should be appropriate to the process, identification of the specific risk
importance of the identified problem. Critical factors inherent for a given dossier or dosage
issues should be addressed with appropriate form, and allocation of resources to the
urgency and formality. various sections of a dossier for a given
product. In addition, product-related risk
There should be a logical selection of tools factors must be managed throughout the
for risk assessment. Risk acceptance criteria lifecycle of the product, for example through
should be appropriate. Risk assessments effective communication between assessors
should not underrate the severity, nor and inspectors, and by establishing systems
overrate detection of occurrences resulting in for dealing with the products after approval.
underestimating patient risk.
The prioritization of dossiers should consider
The risk acceptance criteria should be the therapeutic needs of the regional
appropriate for the specific situation in population (disease occurrence, the need for
question. paediatric formulations, combination products,
etc.) and the availability of medicines on
Risk controls should be effective. The the market. Prioritization should be a
company should have a review programme to dynamic process in order to accommodate
measure the efficiency of the measures taken. emerging issues such as pandemics. Other
considerations related to prioritization based
Risk-based decision(s) should be science- on medical need may include fixed-dose
based and concordant with the predefined combinations versus single-ingredient or
acceptance criteria. co-packaged products, extended release
products versus twice or thrice daily dosing
All documentation related to the QRM products, second-line versus first-line
activities should be completed in a reasonable products, flexible dosage forms such as
time frame and should be accessible. dispersible tablets and variable dose products
such as oral liquids.
Risk assessments performed should be
reviewed when appropriate, and additional The screening process examines the
controls implemented when required. completeness of a dossier. Screening
ensures that only those dossiers that meet
4.4 QRM applied to dossier review minimum standards for completeness are
(assessment) entered into the full assessment process.
Insufficient screening processes allow a lower
NMRA assessment processes rely on QRM standard of quality of dossiers to be accepted
principles in the management of resources for review, significantly increasing assessment
(time and assessors), as well as in the time.
management of product-related risk factors.
Efficient management of resources minimizes Identification of dossier related and product
the risk that limited resources are not used related risk factors allows for the allocation
to best effect, and ultimately ensures that of proper resources to specific dossiers.
important products are available in a timely Possible risk factors include the experience
manner. Key factors to be considered include and track record of the manufacturer, narrow
245
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
246
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
The important criterion for acceptability is that One aspect worth highlighting is the
the tool or tools are used effectively to support development of a risk matrix to facilitate
the key attributes of a good risk assessment. categorization of identified risks during the risk
assessment phase. In order to prioritize a risk,
The Product Quality Research Institute (PQRI) it is essential to agree upon its significance.
Manufacturing Technology Committee (MTC) The risk associated with any situation or event
has produced a summary (10) of common RM can be represented as the impact of that event
principles and best practices, several working multiplied by the probability of its occurrence;
tools to foster consistency in the use of ICH in other words, how likely is it to happen and
Q9 (6) in day-to-day RM decision-making, how severe would it be if it did happen. Impact
and a series of examples of RM applications and probability can each be classified, e.g.
currently in use by major pharmaceutical firms. into 5 levels (1-5) or with a weighting towards
They have also produced very helpful risk tool the higher probability and impact ratings (e.g.
training modules for risk ranking and filtering, 1,3,5,7,10, etc.), so that a grid or matrix can
failure modes effects analysis (FMEA) (14, 15, be constructed.
16), hazard operability analysis (HAZOP) (17)
and HACCP (3)
The shading in the table represents an example of how the risk values (sometimes called
composite risk indices or risk index values) can be assigned a high, medium or low status. The
definition for each status should be predetermined in the QRM process after consideration of the
specific consequences for the process undergoing risk assessment. These consequences can be
split according to the probability and impact scores, as exemplified in Table 2.
247
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
This table is just a very basic example and would need to be customized for the specific process
in question to enable better and practical definition of the consequence categories. It should be
cautioned that the value of a risk matrix does very much rely upon input information and should
only be used by staff with a good understanding of the embedded judgments and, as such, the
resolution of low/medium/high categorization.
As a summary of the common, well-recognized QRM tool options available for the purposes of
this guideline, the following table has been based on the one from the PQRI-MTC report (10).
The list is not comprehensive but it does include some of the more frequently used approaches.
248
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Basic tools
Risk ranking and • Method to compare and rank risks • Prioritize operating
filtering areas or sites for audit/
• Typically involves evaluation of assessment
multiple diverse quantitative and
qualitative factors for each risk, • Useful for situations
• and weighting factors and risk when the risks and
• score underlying
consequences are
diverse and difficult
to compare using a
single tool
Advanced tools
Fault tree analysis • Method used to identify all root causes of • Investigate product
(FTA) an assumed failure or problem complaints
249
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Hazard operability • Tool assumes that risk events are • Access manufacturing
analysis (HAZOP) caused by deviations from the processes, suppliers,
design and operating intentions facilities and equipment
Hazards analysis • Identify and implement process controls • Better for preventative
and critical control that consistently and effectively prevent applications rather than
points (HACCP) hazard conditions from occurring reactive
250
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
251
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Stakeholder
252
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
253
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
14. Failure Mode and Effect Analysis. Annex 5 (WHO Technical Report Series, No.
FMEA from Theory to Execution. 823); Quality assurance of pharmaceuticals.
2ndEdition 2003. D. H. Stamatis, A compendium of guidelines and related
ISBN 0873895983. materials. Vol. 2,Secondupdated edition.
Good manufacturing practices and inspection.
15. Guidelines for Failure Modes and Geneva, World Health Organization, 2007;
Effects Analysis (FMEA) for Quality assurance of pharmaceuticals.
Medical Devices. 2003 Dyadem A compendium of guidelines and related
Press, ISBN 0849319102. materials. World Health Organization, 2011
(CD-ROM) (http://apps.who.int/medicinedocs/
16. McDermott R et al (1996) The en/q/).
Basics of FMEA. ISBN 0527763209.
254
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
255
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
that its application in this case, although The guidance in this document would normally
recommended, is not required in PIC/S be applied to the steps showing ray in Table 1.
countries. It does not imply that all steps shown should
be completed. The stringency of GMP in API
An“ API Starting Material” is a raw material, manufacturing should increase as the process
intermediate, or an API that is used in the proceeds from early API steps to final steps,
production of an API and that is incorporated purification, and packaging.
as a significant structural fragment into the Physical processing of APIs, such as
structure of the API. An API Starting Material granulation, coating or physical manipulation
can be an article of commerce, a material of particle size (e.g. milling, micronizing),
purchased from one or more suppliers should be conducted at least to the standards
under contract or commercial agreement, or of this Guide.
produced in-house. API Starting Materials
normally have defined chemical properties This GMP Guide does not apply to steps prior
and structure. to the introduction of the defined “API Starting
Material”.
The manufacturer should designate and
document the rationale for the point at which
production of the API begins. For synthetic
processes, this is known as the point at
which” API Starting Materials” are entered
into the process. For other processes (e.g.
fermentation, extraction, purification, etc), this
rationale should be established on a case-by-
case basis. Table1givesguidanceon the point
at which the API Starting Material is normally
introduced into the process.
256
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
257
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
2.14 The persons authorized to release 2.22 The main responsibilities of the
intermediates and APIs should independent quality unit(s) should
be specified. not be delegated. These
responsibilities should be described
2.15 All quality related activities should in writing and should include but not
be recorded at the time they necessarily be limited to:
are performed.
258
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
1. Releasing or rejecting all APIs. 13. Making sure that materials are
Releasing or rejecting intermediates appropriately tested and the results
for use outside the control of the are reported;
manufacturing company;
14. Making sure that there is stability
2. Establishing a system to release or data to support retest or expiry
reject raw materials, intermediates, dates and storage conditions on
packaging and labelling materials; APIs and/or intermediates where
appropriate; and
3. Reviewing completed batch
production and laboratory control 15. Performing product quality reviews
records of critical process steps (as defined in Section 2.5)
before release of the API
for distribution; 2.3 Responsibility for Production
Activities
4. Making sure that critical deviations
are investigated and resolved; The responsibility for production activities
should be described in writing, and should
5. Approving all specifications and include but not necessarily be limited to:
master production Instructions;
1. Preparing, reviewing, approving and
6. Approving all procedures impacting distributing the instructions for the
the quality of Intermediates or APIs; production of intermediates or APIs
according to written procedures;
7. Making sure that internal audits
(self-inspections) are Performed; 2. Producing APIs and, when
appropriate, intermediates
8. Approving intermediate and API according to pre- approved
contract manufacturers; instructions;
12. making sure those effective systems 5. making sure that production facilities
are used for maintaining and are clean and when appropriate
calibrating critical equipment; disinfected;
259
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
260
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
261
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
262
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
4.23 Permanently installed pipe work 4.34 Where the manufacturer of anon-
should be appropriately identified. sterile API either intends or claims
This can be accomplished by that it is suitable for use in
identifying individual lines, further processing to produce a
documentation, computer control sterile drug (medicinal) product,
systems, or alternative means. water used in the final isolation
Pipe work should be located and purification steps should be
to avoid risks of contamination of monitored and controlled for total
the intermediate or API. microbial counts, objectionable
organisms, and endotoxins.
4.24 Drains should be of adequate size
and should be provided with an air 4.4 Containment
break or a suitable device to prevent
back-siphonage, when appropriate. 4.40 Dedicated production areas,
which can include facilities,
4.3 Water air handling equipment and/or
process equipment, should
4.30 Water used in the manufacture of be employed in the production of
APIs should be demonstrated to be highly sensitizing materials, such as
suitable for its intended use. penicillins or cephalosporins.
4.31 Unless otherwise justified, process 4.41 Dedicated production areas should
water should, at a minimum, also be considered when material of
Meet World Health Organization an infectious nature or high
(WHO) guidelines for drinking pharmacological activity or toxicity
(potable) water quality. is involved (e.g., certain steroids or
cytotoxic anti-cancer agents)
4.32 If drinking (potable) water is unless validated in activation and/
insufficient to assure API quality, or cleaning procedures are
and tighter chemical and/or established and maintained.
263
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
264
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
265
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
266
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
data change made, the previous 5.49 Data can be recorded by a second
entry, who made the change, and means in addition to the computer
when the change was made. system.
267
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
6.14 When entries are made in records, established and documented for
these should be made indelibly in in-process controls.
spaces provided for such entries,
directly after performing the 6.18 If electronic signatures are used on
activities, and should identify the documents, they should be
person making the entry. authenticated and secure.
Corrections to entries should be
dated and signed and leave the 6.2 Equipment Cleaning and
original entry still readable. Use Record
6.15 During the retention period, originals 6.20 Records of major equipment use,
or copies of records should be cleaning, sanitization and/or
readily available at the sterilization and maintenance should
establishment where the activities show the date, time(if appropriate),
described in such records occurred. product, and batch number of
Records that can be promptly each batch processed in the
retrieved from another location by equipment, and the person who
electronic or other means performed the cleaning and
are acceptable. maintenance.
268
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
269
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
• the instructions for storage of the • Dates and, when appropriate, times;
intermediate or API to assure its
suitability for use, including the • Identity of major equipment (e.g.,
labelling and packaging materials reactors, driers, mills, etc.) used;
and special storage conditions with
time limits, where appropriate. • Specific identification of each batch,
including weights, measures,
6.5 Batch Production Records (Batch and batch numbers of raw materials,
Production and Control Records) intermediates, or any reprocessed
Materials used during
6.50 Batch production records should be manufacturing;
prepared for each intermediate and
API and should include complete • Actual results recorded for critical
information relating to the process parameters;
production and control of each
batch. The batch production record • any sampling performed;
should be checked before issuance
to assure that it is the correct • Signatures of the persons
version and a legible accurate performing and directly supervising
reproduction of the appropriate or checking each critical step in
master production instruction. If the the operation;
batch production record is produced
from a separate part of the master • In-process and laboratory test
document, that document should results;
include a reference to the current
master production instruction • Actual yield at appropriate phases
being used. or times;
270
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
271
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
272
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
273
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
274
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
275
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
8.32 Critical in-process controls (and 8.40 For the purpose of this document,
critical process monitoring), blending is defined as the process
including the control points and of combining materials with in the
methods, should best put in writing same specification to produce a
and approved by the quality unit(s). homogeneous intermediate or API.
In-process mixing of fractions
from single batches (e.g., collecting
8.33 In-process controls can be several centrifuge loads from a
performed by qualified production single crystallization batch)or
department personnel and the combining fractions from several
process adjusted without prior batches for further processing
quality unit(s) approval if the is considered to be part of the
adjustments are made with in production process and is not
pre-established limits approved by considered to be blending.
the quality unit(s). All tests and
results should be fully documented 8.41 Out-Of-Specification batches should
as part of the batch record. not be blended with other batches
for the purpose of meeting
8.34 Written procedures should specifications. Each batch
describe the sampling methods incorporated into the blend should
for in-process materials, have been manufactured using
intermediates, and APIs. Sampling an established process and should
plans and procedures should have been individually tested and
be based on scientifically sound found to meet appropriate
sampling practices. specifications prior to blending.
276
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
277
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
9.30 Access to the label storage areas 9.4 Packaging and Labelling
should be limited to authorized Operations
personnel.
9.40 There should be documented
9.31 Procedures should be used to procedures designed to ensure that
reconcile the quantities of labels correct packaging materials and
is sued, used, and returned and to labels are used.
evaluate discrepancies found
between the number of containers 9.41 Labelling operations should be
labelled and the number of labels designed to prevent mix-ups. There
issued. Such discrepancies should should be physical or spatial
be investigated, and the separation from operations involving
investigation should be approved by other intermediates or APIs.
the quality unit (s).
9.42 Labels used on containers of
9.32 All excess labels bearing batch intermediates or APIs should
numbers or other batch-related indicate the name or identifying
printing should be destroyed. code, the batch number of the
278
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
279
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
280
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
281
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
necessary for APIs from herbal 11.42 The Certificate should list each test
or animal tissue origin. performed in accordance with
Biotechnology considerations are compendial or customer
covered in ICH Guideline Q6B. requirements, including the
acceptance limits, and the numerical
11.22 The impurity profile should be results obtained (if test results
compared at appropriate intervals are numerical).
against the impurity profile in the
regulatory submission or 11.43 Certificates should be dated
compared against historical data in and signed by authorized personnel
order to detect changes to the API of the quality unit (s) and should
resulting from modifications in raw show the name, address
materials, equipment operating and telephone number of the
parameters, or the production original manufacturer. Where
process. the analysis has been carried out by
a re-packer or re-processor, the
11.23 Appropriate microbiological tests Certificate of Analysis should show
should be conducted on each batch the name, address and telephone
of intermediate and API where number of the re-packer/reprocess
microbial quality is specified. or and a reference to the name of
the original manufacturer.
11.3 Validation of Analytical
Procedures-see Section 12. 11.44 If new Certificates are issued by or
on behalf of re-packers/re-
11.4 Certificates of Analysis processors, agents’ or brokers,
these Certificates should show the
11.40 Authentic Certificates of Analysis name, address and telephone
should be issued for each batch of number of the laboratory that
intermediate or API on request. performed the analysis. They should
also contain a reference to the
11.41 Information on the name of name and address of the original
the intermediate or API including manufacturer and to the original
where appropriate its grade, the batch Certificate, a copy of which
batch number, and the date of should be attached.
release should be provided on the
Certificate of Analysis. For 11.5 Stability Monitoring of APIs
intermediates or APIs with an expiry
date, the expiry date should be 11.50 A documented, on-going testing
provided on the label and Certificate program should be designed to
of Analysis. For intermediates or monitor the stability characteristics
APIs with a retest date, the retest of APIs, and the results should be
date should be indicated on the used to confirm appropriate storage
label and/or Certificate of Analysis. conditions and retest or expiry
dates.
282
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
11.51 The test procedures used in stability specific test intervals (e.g.9 month
testing should be validated and be testing) can be considered.
stability indicating.
11.56 Where appropriate, the stability
11.52 Stability samples should be stored storage conditions should be
in containers that simulate the consistent with the ICH guidelines
market container. For example, if on stability.
the API is marketed in bags
within fiber drums, stability samples 11.6 Expiry and Retest Dating
can be packaged in bags of the
same material and in smaller- 11.60 When an intermediate is intended
scale drums of similar or identical to be transferred outside the control
material composition to the market of the manufacturer’s material
drums. management system and an expiry
or retest date is assigned,
11.53 Normally the first three commercial supporting stability information
production batches should be should be available (e.g. published
placed on the stability monitoring data, test results).
program to confirm there test or
expiry date. However, where data 11.61 An API expiry or retest date should
from previous studies show that the be based on an evaluation of data
API is expected to remain stable for derived from stability studies.
at least two years, fewer than three Common practice is to use a retest
batches can be used. date, not an expiration date.
11.54 Thereafter, at least one batch per 11.62 Preliminary API expiry or retest
year of API manufactured (unless dates can be based on pilot scale
none is produced that year) should batches if (1) the pilot batches
be added to the stability monitoring employ a method of manufacture
program and tested at least annually and procedure that simulates the
to confirm the stability. final process to be used on a
commercial manufacturing scale;
11.55 For APIs with short shelf-lives, and (2) the quality of the API
testing should be done more represents the material to be made
frequently. For example, for those on a commercial scale.
biotechnological/biologic and other
APIs with shelf-lives of one year or 11.63 A representative sample should be
less, stability samples should be taken for the purpose of performing
obtained and should be tested a retest.
monthly for the first three months
and at three month intervals after 11.7 Reserve/Retention Samples
that. When data exist that
confirm that the stability of the API 11.70 The packaging and holding of
is not compromised, elimination of reserve samples is for the purpose
of potential future evaluation of the
283
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
For APIs with retest dates, similar • defining the API in terms of its
reserve samples should be retained critical product attributes;
for three years after the batch is • identifying process parameters that
completely distributed by the could affect the critical quality
manufacturer. attributes of the API;
• determining the range for each
11.72 The reserve sample should be critical process parameter expected
stored in the same packaging to be used during routine
system in which the API is stored or manufacturing and process control.
in one that is equivalent to
or more protective than the
marketed packaging system. 12.12 Validation should extend to those
operations determined to be critical
Sufficient quantities should to the quality and purity of the API.
be retained to conduct at least
two full compendial analyses or, 12.2 Validation Documentation
when there is no pharmacopoeial
monograph, two full specification 12.20 Written validation protocols should
analyses. be established that specifies how
validation of a particular process
12. VALIDATION will be conducted. The protocol
should be reviewed and approved
12.1 Validation Policy by the quality unit (s) and other
designated units.
12.10 The company’s overall policy,
intentions, and approach to 12.21 The validation protocol should
validation, including the validation of specify critical process steps and
production processes, cleaning acceptance criteria as well as
procedures, analytical methods, in- the type of validation to be
process control test procedures, conducted (e.g. retrospective,
computerized systems, and persons prospective, concurrent) and the
responsible for design, review, number of process runs.
approval and documentation of each
284
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
285
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
286
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
for batches used for pivotal clinical and the calculation of residue limits
and toxicological studies. based on potency, toxicity, and
stability.
12.6 Periodic Review of
Validated Systems 12.72 The cleaning validation protocol
should describe the equipment to
12.60 Systems and processes should be cleaned, procedures, materials,
be periodically evaluated to verify acceptable cleaning levels,
that they are still operating in a parameters to be monitored and
valid manner. Where no significant controlled, and analytical methods.
changes have been made to the
system or process, and a quality The protocol should also indicate
review confirms that the system or the type of samples to be obtained
process is consistently producing and how they are collected
material meeting its specifications, and labelled.
there is normally no need for
revalidation. 12.73 Sampling should include swabbing,
rinsing, or alternative methods (e.g.,
12.7 Cleaning Validation direct extraction), as appropriate,
to detect both insoluble and soluble
12.70 Cleaning procedures should residues. The sampling methods
normally be validated. In general, used should be capable of
cleaning validation should be quantitatively measuring levels of
directed to situations or process residues remaining on the
steps where contamination or equipment surfaces after cleaning.
carryover of materials poses the Swab sampling may be impractical
greatest risk to API quality. For when product contact surfaces are
example, in early production it not easily accessible due to
may be un necessary to validate equipment design and/or process
equipment cleaning procedures limitations (e.g., inner surfaces of
where residues are removed by hoses, transfer pipes, reactor tanks
subsequent purification steps. with small ports or handling toxic
materials, and small intricate
12.71 Validation of cleaning procedures equipment such as micronizers
should reflect actual equipment and microfluidizers).
usage patterns. If various APIs or
intermediates are manufactured 12.74 Validated analytical methods having
in the same equipment and the sensitivity to detect residues or
equipment is cleaned by the same contaminants should be used.
process, a representative The detection limit for each
intermediate or API can be selected analytical method should be
for cleaning validation. This sufficiently sensitive to detect
selection should be based on the the established acceptable
solubility and difficulty of cleaning level of the residue or contaminant.
The method’s attainable recovery
287
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
288
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
289
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
290
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
14.43 The use of recovered solvents, 15.11 Complaint records should include:
mother liquors, and other recovered
materials should be adequately • Name and address of complainant;
documented.
• Name (and, where appropriate, title)
14.5 Returns and phone number of person
submitting the complaint;
14.50 Returned intermediates or APIs
should be identified as such • Complaint nature (including name
and quarantined. and batch number of the API);
291
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
and how the recalled material pass to a third party any of the work
should be treated. entrusted to him under the contract
without the contract giver’s prior
15.15 In the event of a serious or evaluation and approval of the
potentially life-threatening situation, arrangements.
local, national, and/or international
authorities should be informed and 16.15 Manufacturing and laboratory
their advices ought. records should be kept at the site
where the activity occurs and be
16. CONTRACT readily available.
MANUFACTURERS 16.16 Changes in the process, equipment,
(INCLUDING test methods, specifications, or
LABORATORIES) other contractual requirements
should not be made unless the
16.10 All contract manufacturers (including contract giver is informed and
laboratories) should comply with approves the changes.
the GMP defined in this Guide.
Special consideration should be 17. AGENTS, BROKERS,
given to the prevention of cross-
contamination and to maintaining
TRADERS,
traceability. DISTRIBUTORS,
REPACKERS AND
16.11 Contract manufacturers (including RELABELLERS
laboratories) should be evaluated by
the contract giver to ensure 17.1 Applicability
GMP compliance of the specific
operations occurring at the contract 17.10 This section applies to any party
sites. other than the original manufacturer
who may trade and/or take
16.12 There should be a written and possession, repack, relabel,
approved contract or formal manipulate, distribute or store an
agreement between the contract API or intermediate.
giver and the contract accept or
that defines in detail the GMP 17.11 All agents, brokers, traders,
responsibilities, including the quality distributors, repackers, and
measures, of each party. relabellers should comply with GMP
as defined in this Guide.
16.13 The contract should permit the
contract giver to audit the contract 17.2 Traceability of Distributed APIs
acceptor’s facilities for compliance and Intermediates
with GMP.
17.20 Agents, brokers, traders,
16.14 Where subcontracting is allowed, distributors, repackers, or relabellers
the contract acceptor should not
292
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
293
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
17.71 If the situation warrants, the agents, 18.10 Section 18 is intended to address
brokers, traders, distributors, specific controls for APIs or
repackers, or relabellers should intermediates manufactured by
review the complaint with the cell culture or fermentation using
original API or intermediate natural or recombinant organisms
manufacturer in order to determine and that have not been covered
whether any further action, either adequately in the previous sections.
with other customers who may It is not intended to beast and-alone
have received this API or Section. In general, the GMP
intermediate or with the regulatory principles in the other sections of
authority, or both, should be this document apply. Note that
initiated. The investigation in to the the principles of fermentation
cause for the complaint or recall for “classical” processes for
should be conducted and production of small molecules
documented by the appropriate and for processes using
party. recombinant and non-recombinant
organisms for production of
17.72 Where a complaint is referred to proteins and/or polypeptides are the
the original API or intermediate same, although the degree of
manufacturer, the record maintained control will differ. Where practical,
by the agents, brokers, traders, this section will address these
distributors, repackers, or relabellers differences. In general, the degree
should include any response of control for bio technological
received from the original API or processes used to produce proteins
intermediate manufacturer and polypeptides is greater than
(including date and information that for classical fermentation
provided). processes.
294
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
295
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
• Harvest and purification procedures 18.22 Records of the use of the vials from
that remove cells, cellular debris the cell banks and storage
and media components while conditions should be maintained.
protecting the intermediate or
API from contamination (particularly 18.23 Where appropriate, cell banks
of microbiological nature) and from should be periodically monitored to
loss of quality; determine suitability for use.
296
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
18.35 Culture media should be sterilized 18.41 Harvest and purification procedures
before use when appropriate to that remove or inactivate the
protect the quality of the API. producing organism, cellular debris
and media components (while
18.36 There should be appropriate minimizing degradation,
procedures in place to detect contamination, and loss of
contamination and determines the quality) should be adequate
course of action to be taken. This to ensure that the intermediate or
should include procedures to API is recovered with consistent
determine the impact of the quality.
contamination on the product
and those to decontaminate the
equipment and return it to a
condition to be used in subsequent
batches. Foreign organisms
observed during fermentation
297
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
18.42 All equipment should be properly 18.53 The same equipment is not normally
cleaned and, as appropriate, used for different purification steps.
sanitized after use. Multiple However, if the same equipment
successive batching without is to be used, the equipment should
cleaning can be used if intermediate be appropriately cleaned and
or API quality is not compromised. sanitized before reuse. Appropriate
precautions should be taken to
18.43 If open systems are used; prevent potential virus carry-over
purification should be performed (e.g. through equipment or
under environmental conditions environment) from previous steps.
appropriate for the preservation of
product quality. 19. APIs FOR USE IN
18.44 Additional controls, such as the
CLINICAL TRIALS
use of dedicated chromatography
19.1 General
resins or additional testing, may
be appropriate if equipment is to be
19.10 Not all the controls in the previous
used for multiple products.
sections of this Guide are
appropriate for the manufacture of
18.5 Viral Removal/Inactivation Steps
a new API for investigational
use during its development. Section
18.50 See the ICH Guideline Q5A Quality
19 provides specific guidance
of Biotechnological Products: Viral
unique to these circumstances.
Safety Evaluation of Biotechnology
Products Derived from Cell Lines
19.11 The controls used in the
of Human or Animal Origin for more
manufacture of APIs for use in
specific information.
clinical trials should be consistent
with the stage of development of the
18.51 Viral removal and viral inactivation
drug product incorporating the
steps are critical processing steps
API. Process and test procedures
for some processes and should be
should be flexible to provide
performed within their validated
for changes as knowledge of the
parameters.
process increases and clinical
testing of a drug product progresses
18.52 Appropriate precautions should be
from pre-clinical stages through
taken to prevent potential viral
clinical stages. Once drug
contamination from pre-viral to post-
development reaches the stage
viral removal/in-activation steps.
where the API is produced for use
in drug products intended for clinical
Therefore, open processing should
trials, manufacturers should ensure
be performed in areas that are
that APIs are manufactured in
separate from other processing
suitable facilities using appropriate
activities and have separate air
production and control procedures
handling units.
to ensure the quality of the API.
298
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
19.21 A quality unit(s) independent from 19.40 Raw materials used in production
production should be established for of APIs for use in clinical trials
the approval or rejection of each should be evaluated by testing,
batch of API for use in clinical trials. or received with a supplier’s
analysis and subjected to identity
19.22 Some of the testing functions testing. When a material is
commonly performed by the considered hazardous, a supplier’s
quality unit (s) can be performed analysis should suffice.
within other organizational units.
19.41 In some instances, the suitability of
19.23 Quality measures should include a a raw material can be determined
system for testing of raw materials, before use based on acceptability in
packaging materials, intermediates, small-scale reactions (i.e., use
and APIs. testing) rather than on analytical
testing alone.
19.24 Process and quality problems
should be evaluated. 19.5 Production
19.25 Labelling for APIs intended for 19.50 The production of APIs for use
use in clinical trials should be in clinical trials should be
appropriately controlled and documented in laboratory
should identify the material as being notebooks, batch records, or by
for investigational use. other appropriate means. These
documents should include
19.3 Equipment and Facilities information on the use of production
materials, equipment, processing,
19.30 During all phases of clinical and scientific observations.
development, including the use
of small-scale facilities or 19.51 Expected yields can be more
laboratories to manufacture batches variable and less defined than
of APIs for use in clinical trials, the expected yields used in
procedures should be in place to commercial processes.
ensure that equipment is calibrated, Investigations in to yield variations
clean and suitable for its intended are not expected.
use.
299
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
300
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
301
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
302
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Material Qualification
A general term used to denote raw materials Action of proving and documenting that
(starting materials, reagents, and solvents), equipment or ancillary systems are properly
process aids, intermediates, APIs and installed, work correctly, and actually lead to
packaging and labelling materials. the expected results. Qualification is part of
validation, but the individual qualification steps
Mother Liquor alone do not constitute process validation.
303
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
A substance that has been shown by an Subjecting an intermediate or API that does
extensive set of analytical tests to be authentic not conform to standards or specifications
material that should be of high purity. This to one or more processing steps that are
standard can be: (1) obtained from an different from the established manufacturing
officially recognized source, or (2) prepared by process to obtain acceptable quality
independent synthesis, or (3) obtained from intermediate or API (e.g., recrystallizing with a
existing production material of high purity, or different solvent).
(4) prepared by further purification of existing
production material. Signature (signed)
304
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
A documented program that provides a high The quantity of material or the percentage of
degree of assurance that a specific process, theoretical yield anticipated at any appropriate
method, or system will consistently produce phase of production based on previous
a result meeting pre-determined acceptance laboratory, pilot scale, or manufacturing data.
criteria.
Yield, Theoretical
Validation Protocol
The quantity that would be produced at any
A written plan stating how validation will be appropriate phase of production, based
conducted and defining acceptance criteria. upon the quantity of material to be used, in
For example, the protocol for a manufacturing the absence of any loss or error in actual
process identifies processing equipment, production.
critical process parameters/operating ranges,
product characteristics, sampling, test data to
be collected, number of validation runs, and
acceptable test results.
305
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
306
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
307
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
308
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
PART THREE:
309
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
310
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
Validation: The documented act of proving 3.4 The Site Master File should be a
that any procedure, process, equipment, part of documentation belonging
material, activity, or system actually leads to to the quality management system
the expected results. of the manufacturer and kept
updated accordingly. The Site
Master File should have an edition
3. INTRODUCTION number, the date it becomes
effective and the date by which it
3.1 The Site Master File is prepared has to be reviewed. It should be
by the pharmaceutical manufacturer subject to regular review to ensure
and should contain specific that it is up to date and
information about the quality representative of current activities.
management policies and activities Each Appendix can have an
of the site, the production and/ individual effective date, allowing for
or quality control of pharmaceutical independent updating.
311
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
312
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
313
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
314
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
315
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
316
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
PART FOUR:
GUILDELINES ON
TRAINING AND QUALIFICATIONS
317
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
318
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
GMP Inspector means a GMP Inspector is Senior GMP inspector is an officer who
an officer appointed by the NMRA of Partner by virtue of experience and competence
states in accordance with national regulations is appointed as such to conduct GMP
and the provisions of the NMRA to conduct inspections and train junior officers in
an inspection or assessment in order to verify inspections after evaluation by the NMRA as
GMP compliance of a manufacturing site on by the criteria outlined in the assessment form.
behalf of the NMRA.
Specialized GMP inspector is a GMP
Lead GMP inspector is a Senior GMP inspector who possesses specialized
Inspector who is charged with the knowledge and experience in conducting
responsibility for leading a GMP inspection GMP inspections for specialized areas e.g
team to undertake inspection of a specified Microbiology, HVAC, Biologicals, API
pharmaceutical manufacturing site(s).
319
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
The main objective of GMP is to control and Inspectors should have previous training and
enforce general standards of production and practical experience in the manufacture and/
to provide authorization for the manufacture of or quality control of pharmaceutical products.
specific pharmaceutical products. Graduate pharmacists and chemists or
scientists with an industrial background in
Specifically GMP inspection involves a pharmaceutical production, would qualify for
sequential examination of production and consideration.
control activities on the basis of the GMP
guidelines issued by EAC NMRAs. In The guideline provides information on minimal
addition requires verification that production requirements. They cover inspection of the
and quality control procedures employed production and control of final dosage forms
in the manufacture of specific products are of pharmaceutical products destined for
performed correctly and that they accord human use and of drug substances (active
with data supplied in the relevant licensing pharmaceutical ingredients or bulk drug
applications. substances) employed in their manufacture.
Inspection and licensing of pharmaceutical
Taking into account the paramount importance manufacturing facilities on the basis of
of the management of inspection services, compliance with GMP are a vital element of
this guidance establishes some requirements drug control.
concerning experience, training, assessment
and qualifications of GMP inspectors.
320
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
321
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
322
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
323
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
324
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
325
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
326
DOCUMENT NO: EAC/TF-MED/GMP/FD/COM/N1R0
EAC CLOSE
P.O.BOX 1096
ARUSHA, TANZANIA