Вы находитесь на странице: 1из 16

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/340351376

EU GMP Annex 1 - The new draft and implications for sterile product
manufacturers

Technical Report · April 2020


DOI: 10.13140/RG.2.2.14468.68487

CITATIONS READS

0 4,671

1 author:

Tim Sandle
The University of Manchester
558 PUBLICATIONS   779 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Medicine View project

Microbiological testing View project

All content following this page was uploaded by Tim Sandle on 01 April 2020.

The user has requested enhancement of the downloaded file.


EU GMP Annex 1: The New Draft and the
Implications for Sterile Products Manufacturing

White Paper

Author:
Dr Tim Sandle

1
Abstract
EU GMP Annex 1 is the primary document governing the manufacture, control and release of
sterile pharmaceutical products (both terminally sterilised and aseptically filled medicines).
The Annex will shortly undergo a comprehensive update, as signalled by a new draft
issued in February 2020. This white paper assesses many of the key points within the draft,
focusing on those areas that have a direct impact upon sterile product manufacturing.

Contents
Introduction 3

What is EU GMP Annex 1? 3

Key Changes 4

New Structure 4

Contamination Control Strategy 4

Centrality of the Pharmaceutical Quality System 5

Premises 5

Personnel 8

Microbiological Environmental Monitoring 9

Quality Control 10

Filtration 10

Product Inspection 10

Cleaning and Disinfection 11

Utilities 12

Summary 12

References 13

2
Introduction
Annex 1 of EudraLex “The Rules Governing Medicinal Products in the European Union” forms part of Volume 4 of the
European guidelines covering the manufacture and release of sterile products, which are subject to special
requirements (1).

The Annex is going through a revision process, with the final version expected in 2020. This review process began with
the issuing of a draft in December 2017. This draft was subject to over 6,000 comments submitted by professional
organisations. After a review of these, across a two-year period, the European Medicines Agency (EMA), issued a
new draft (described as ‘version 12’) (2). The new draft addresses some of the issues in the first version and contains
some additional requirements. The scope for the review of version 12 is more limited, with specific areas marked for
consultation and the group of professional bodies invited to comment has been narrowed. This signals that much of the
draft content will be in the final version, and that the final version will be issued within a relatively short timeframe.

The trigger points for the revision to the Annex included emergence of new technologies, and what regulatory
inspectors have highlighted as inadequate root cause analysis and ineffective CAPA. Other points of regulatory concern
included poor implementation of ICHQ9 (which relates to quality risk management) (3). A further reason for revision
was to address various points of ambiguity in the interpretation of the Annex 1 requirements.

This white paper provides an overview of the draft Annex 1 and considers the focal points for sterile products
manufacture, control and release.

What is EU GMP Annex 1?

There are two major, global guidance documents for The scope of Annex 1 relates to pharmaceutical
sterile products manufacture: the FDA guidance, last companies who manufacture products within the
revised in 2004 (4), and Annex 1 of EU GMP. Annex 1 of European Union and those companies who import
EudraLex “The Rules Governing Medicinal Products in the products into the European Union (including a post-
European Union” forms part of Volume 4 of the European Brexit United Kingdom).
guidelines (1) (there is also a WHO guidance. The purpose
Annex 1 of EU GMP has undergone no major revision
of these guidances, is to detail the controls required
since 2007 and no change whatsoever since 2009
around the manufacture of sterile products. Special
(where there was a minor point of clarification about the
requirements are needed in order to minimise risks of
required air supply grading for oversealing - Grade A air
microbiological, particulate and pyrogen contamination
supply). The lack of an update through the intervening
of sterile products; and also, to provide guidance as to
years was especially notable in the context of updates to
how sterile products are best protected. Annex 1 includes
cleanroom technology and the appearance of new types
key areas like personnel training, equipment qualification,
of rapid microbiological methods. As part of the revision
cleanroom design and environmental monitoring.
process, the current title of “Annex 1: Manufacture of
Sterile Medicinal Products” is now tweaked to read
“Annex 1: Manufacture of Sterile Products”.

3
Key Changes Contamination Control
There are five broad areas of change to the draft, in terms Strategy
of tone and emphasis. These are reflected at various
intervals in the document (often through repeated A substantial part of the draft Annex is given over to
occurrences). These areas are: each organisation having a detailed, facility-specific
contamination control strategy (abbreviated to CCS). To
1. The global acceptance and implementation of ICH Q9 be effective this needs to be an approach that can assess
(Quality Risk Management) and Q10 (Pharmaceutical seemingly isolated contamination events holistically and
Quality System), is not reflected in the current Annex. which is capable of putting appropriate corrective and
The new draft contains many references to Quality Risk preventive actions (CAPA) in place. This requirement
Management (QRM) in particular, emphasising that is intended to signal a new paradigm in terms of
QRM should be used as a proactive tool. contamination control, shifting the risk review process to
2. There is the requirement for a formal, holistic one that assesses the impact of a contamination event in
contamination control strategy. The expectation is for a a far wider context.
formal document which reflects the site-wide strategy The main elements of such a control strategy are:
for minimising contamination control with respect to
sterile manufacturing. Such a strategy, as outlined below, • Design of both the plant and process
is for each organisation to fully-understand and review • Equipment and facilities
design, procedural, technical and organisational controls. • Personnel
3. There have been advances in sterile manufacturing • Utilities
technology, especially with RABS and isolators. There • Raw materials control – including in-process controls
have also been advances with rapid microbiological
• Product containers and closures
methods, which the draft Annex acknowledges
• Vendor approval – such as key component suppliers,
4. There was some ambiguity with the current version sterilisation of components and single use systems,
and these needed correction or clarification and services
5. Annex 1 is often beyond sterile manufacturing, • For outsourced services, such as sterilisation,
including aspects of non-sterile manufacturing. sufficient evidence should be provided to the
The scope of the new draft has been modified and contract giver to ensure the process is operating
broadened to reflect this. correctly

6. In particular the references to Quality Risk Management • Process risk assessment


are more extensive, with requirement to use QRM to • Process validation
review existing products and processes; new products • Preventative maintenance – maintaining equipment
and processes (where risk assessment needs to link and premises (planned and unplanned maintenance)
to Quality by Design); and to address problems with to a standard that will not add significant risk of
procedures and processes, with the expectation that contamination
QRM becomes a key part of deviation management. • Cleaning and disinfection
This reflects a significant shift in regulatory thinking
• Monitoring systems - including an assessment of the
towards a more risk-centric approach.
feasibility of the introduction of scientifically sound,

New Structure modern methods that optimise the detection of


environmental contamination
The new draft is divided into eleven parts, which are • Prevention – trending, investigations, corrective and
presented across 52 pages (which means that it is two preventive actions (CAPA), root cause determination
pages longer than the 2017 version and considerably and the need for more robust investigational tools
more than the current Annex’s 16 pages). • Continuous improvement based on information from
1. Scope the above systems
2. Principle With each of these different elements it is notable
3. Pharmaceutical Quality System (PQS) that they are not confined to biocontamination, since
4. Premises reference is also made to subvisible particles (the classic
5. Equipment appearance tests) and the overall appearance of the
6. Utilities pharmaceutical product.
7. Personnel
8. Production and Specific Technologies
9. Viable and Non-viable Environmental and
Process Monitoring
10. Quality Control (QC)
11. Glossary
4
Centrality of the Pharmaceutical Quality System
The first main section of the draft refers to the Pharmaceutical Quality System, a quality system that each manufacturer
should have in place. While general matters are covered in Chapter 1 of EU GMP, the draft Annex makes reference to
specific aspects for sterile products manufacture. These include:

• The proactive use of risk management

• Regular review of risk assessments

• Rationales in place to address different categories of risk arising from risk assessments

• Employing staff with sufficient expertise to undertake risk assessments

• Use of effective root cause and CAPA

• Those tasked with releasing products must be fully conversant with risks and quality issues

• The first two bullet points signal the continuing reference to risk management and risk assessment throughout the
document

The reference to risk assessment should not simply confined to manufacturing, it needs to extend to packaging (primary
and secondary) and to the distribution of the finished medicinal product, thereby embracing the requirements of Good
Distribution Practice (GDP).

Premises

The main changes under the premises section relate to cleanroom design and operation. With the overall design of
cleanrooms, the draft stresses the importance of ‘quality-by-design’, with particular reference made to lay-out and
process flow, where the process steps are reflective of an intent to maintain contamination control. This design concept
extends to the necessity of conducting all non-essential processes outside clean areas. Reference is made to Annex 15 of
the EU GMP guide (“Qualification and Validation”) in relation to ensuring that equipment has been suitably qualified.

The text makes reference to the specific series of tests required when a cleanroom is qualified. These tests are (where
relevant to the design):

• Installed filter leakage and integrity testing • Microbial airborne and surface contamination

• Airflow measurement - Volume and velocity • Temperature measurement

• Air pressure difference measurement (with the new • Relative humidity measurement
draft the specification changes from 10 to 15 pascals
• Recovery testing
to a minimum of 10 Pascals)
• Containment leak testing
• Airflow direction and visualisation

5
Notably here is the task of microbiological monitoring. part of the acceptance of the adjusted volume includes an
This is something that has been discussed in relation to airflow visualisation to be conducted. For isolators that are
the update of the ISO 14698 biocontamination standards, ‘closed’ the Annex states that the air direction need not
about adding a microbiological monitoring aspect to the necessarily be unidirectional.
assessment of cleanrooms.
The revised Annex has added information relating to
With particle assessments, the Annex keeps the requirement environmental controls in relation to pressure. In addition
to count airborne particulates equal to or greater than 0.5 to revising the limit for pressure (as indicated above), there
and 5.0 μm (which continues to create a difference with the is a requirement that all areas with differential pressure
FDA aseptic processing guidance, which refers to particles have warning alarms in place, where the warning signals a
of ≥0.5 μm only). However, for Grade A zone and Grade B potential problem with air supply.
at rest, classification only needs to be for particles equal
Barrier Technology
to or greater than 0.5 μm. The cleanroom classification
expectations apply to the ‘at rest’ and ‘in operation’ states
(with the ‘in operation’ state there is a suggestion that for
aseptic processing areas that the exercise is undertaken
during media fills, in order to represent worst-case). For
selecting particle locations the draft Annex states that the
ISO 14644 methodology is to be followed, along with the
additional expectation that, for aseptic processing areas,
sample locations are positioned so that all critical processing
zones like the point of fill and stopper bowls are included and
based on a documented risk assessment. The Annex does not go as far as mandating the use of
barrier technology; however, there is a recommendation
Whilst there are no changes to requalification intervals
that manufacturers consider adopting “appropriate
(Grades A and B six-monthly and Grade C and D annually)
technologies”, such as Restricted Access Barriers Systems
additional text has been added stipulating that re-
(RABS) or isolators. The recommendation extends to
qualifications should be undertaken following any remedial
consideration of robotic systems, which will reduce the
works needed on equipment or where the facility requires
need for human intervention. With the emphasis upon
work or where new equipment is added to the cleanroom,
barrier technology the Annex requires that any alternative
as assessed through change control. Other reasons for
must be robustly risk assessed.
undertaking a re-qualification include change of room use
and to reassess areas following a loss of power. With isolator technology used for aseptic processing, the
text states that pass through of items should be minimised
Outside of classified cleanrooms, reference is made to
and ideally go through a rapid decontamination chamber.
‘controlled but not classified areas’. Here the movement
Gloves are recognised as the weakest point with an isolator
of material from controlled but not classified to Grade C
system, so consequently there is the requirement for glove
needs to be based on risk management principles, with
leak testing at a minimum interval of before and after
means that the level of cleaning and disinfection and the
each batch (the leak assessment also extends to the main
control of materials needs to be commensurate with the
isolator unit as well). In the text, the frequency of glove leak
level of risk assessed.
testing is detailed as “a minimum at the beginning and end
For the first time the draft Annex provides guidance for of each batch.” Also, with gloves, there is reference to the
Grade D particles, albeit for ‘at rest’ rather than for ‘in importance of selecting the correct isolator gloves; those
operation’. Here a value of 29,000 particles for the ≥0.5 with good mechanical and chemical resistance. The period
μm size particle is provided as the limit. The current Annex of time between the replacement of gloves now requires
simply lists Grade D particles as ‘separately determined’, justifying in the contamination control strategy.
with no further guidance supplied.
For the preparation of isolators for filling, the new draft
In terms of other critical parameters, the draft Annex stresses the importance of cleaning prior to disinfection
offers more scope with unidirectional airflow velocities. (since the presence of residues can inhibit the ability of
With air speed, while the range of 0.36 – 0.54 m/s disinfectants to traverse microbial cell walls, which is
remains the general requirement at working height; necessary for the killing of microorganisms). In keeping
however, there is scope for companies to operate clean with cleaning, the ‘clean hold time’ also requires qualifying.
air devices outside of this range should be designed, There is a requirement to verify that the disinfectant used,
provided that this is scientifically justified and detailed in relation to any remaining residues, does not have an
in the contamination control strategy. With the related adverse impact upon the product.
aspect of airflow visualisation (‘smoke’) studies these
A further change, with the set-up of filling line contained
now need to be conducted in both the ’at rest’ and ‘in
within barrier technology, is that a sporicidal disinfection
operational’ states (previously the reference was just to
process is used following set-up, as mechanism to
‘in operation’). The draft Annex requires that following
reduce any microbial contamination that may have been
an adjustment to air velocities for qualified devices that
presented during the assembly process.

6
Specific points relating to aseptic For transfer of items and equipment into aseptic
processing processing areas, there is added emphasis
upon unidirectional airflow and sterilisation or
The current Annex requires all connections for aseptic decontamination. Where possible, items should be
processing (such as vessel to manifold) to be performed sterilised and passed into the area through double-ended
under Grade A. The draft acknowledges advancements sterilisers (such as a through a double-door autoclave
with sterile processing technology, permitting aseptic or depyrogenation oven/tunnel). Where sterilisation
connections that use intrinsic sterile connection devices, on transfer of the items is not possible, pass through
designed to minimise any potential contamination from hatches equipped with unidirectional airflow and with a
the immediate environment, to be performed in lower method of transfer disinfection, in place should be used.
classified environments provided that the connection With equipment sterilisation, as required for aseptic
device has been appropriately validated to show no processing, the text requires all direct and indirect
ingress of microbial contamination. contact parts to be sterilised.

The control of operation time is discussed in several Single-use systems are technologies used in manufacture
places, perhaps with the view that times outside what is of sterile products as alternatives to reusable equipment.
ordinarily seen can lead to an increased contamination The systems are designed to maintain integrity
risk. Time is referred to with: throughout processing under the intended operational
conditions and their application can help to reduce
• Pre-fill time should be assessed as part of media fills
contamination risk, e.g. aseptic connections. The use of
• A time limit required for aseptic assembly single-use systems and technologies continues to be
• Maximum exposure time of sterilised containers and encouraged within the draft Annex, albeit with the caveat
closures prior to closure. This infers oversealing times that the interaction between the product and product
need to be set. This has an impact with regards to contact surface (notably adsorption, leachable and
ensuring crimping is conducted expediently extractables) is carefully understood.

• For items sterilised “in house” (such as by Other revisions to the Annex which impact on
autoclaving), these need to be stored in Grade A operations conducted within cleanrooms include
or B, using appropriately sealed packaging and a additional information about media fills (aseptic process
maximum hold period must be established simulations), where more ‘time based’ criteria have been
added (such as assessing filling machine hold times and
• The Annex sets out the main elements for Grade A sterilised equipment hold times as part of the exercise).
processing, which allows manufacturers to risk assess The importance of simulating all events remains (given
the essential elements. These are: that events, especially interventions are more important
• Maintaining the critical processing zone for the success or failure of a media fill than simply run
time). There has also been a change to acceptable failure
• The aseptic assembly of filling equipment
limits in media fill studies, based on filling run size. The
• Aseptic connections (these should be sterilised by formally specified statistical approach of >99.9% at 95%
steam-in-place whenever feasible). confidence interval is no longer applicable to determine
acceptable failure limits. Instead the expectation is
• Special focus on aseptic compounding and mixing
zero, irrespective of the media fill size. This will create
• The risks around the replenishment of sterile product, tighter limits for those manufacturers who have yet to
containers and closures adopt more stringent, especially for large batch sizes.
• Concerns around the removal and cooling of items A further point with media fill failures is that, following
from heat sterilisers investigation and root cause analysis, the previous note
about running one media fill has changed to a near
• Staging and conveying of sterile primary packaging requirement to run three media fills.
components
Greater detail is provided for assessing the success of
• Aseptic filling, sealing, transfer of open or partially autoclave operations, such as the requirement to inspect
stoppered vials, including interventions sterilised packaging for its integrity and dryness. Such
• Loading and unloading of a lyophiliser changes are designed to strengthen controls around
sterile products manufacture.
These elements could be cross-referred back to
the contamination control strategy and implied risk
assessment.

7
Personnel

The biggest section within the draft is dedicated to changing areas for access into Grade B and C areas have
personnel. The section includes a requirement to restrict been strengthened. The draft states that outdoor clothing
the numbers of personnel permitted to enter cleanrooms. (other than personal underwear) cannot be brought into
The recommendation is that personnel numbers, changing rooms. In terms of what is not permitted to be
especially in sterile manufacturing areas, is minimised taken into clean areas, the Annex now calls out mobile
based on risk assessment. This acknowledges that the devices (reflecting the ubiquity of smartphones and the
biggest risk of microbial contamination in cleanrooms like) unless these have been supplied by the organisation
derives from people, principally through shedding and by and are shown to be suitable for cleanroom used, and
touching critical surface. covered by a cleaning and disinfection process. With
entry into Grade B changing areas, there needs to be a
In relation to controlling access to cleanrooms there
separate way in and a separate way out (the requirement
is reference made to the importance of on-going
to add this to Grade C areas has been dropped). This is in
quality assurance oversight. To minimise additional
order to reduce cross-contamination.
risk from people, there is a reference to such activities
being completed outside of the cleanroom, such as by With gowns, the text also requires that:
inspection windows or through the use of CCTV.
• Gowns must be suitable to prevent shedding and
As expected, training plays a significant part in the the environment in which operators work and the
personnel section. This extends to everyone entering the movements they may be undertaking needs to be
cleanroom, including cleaning and maintenance staff. taken into account
The minimum basis for a training program is set out as
• The gown worn needs to be of a suitable size
to include: hygiene, cleanroom practices, contamination
control, aseptic techniques, and potential safety • Gowns must not affect the product (this is a
implications to the patient of a loss of product sterility reference to the risk of fibres being released from
and in the basic elements of microbiology. For access gowns and ending up in the product)
into the higher-grade cleanrooms (Grades A and B) to • That gowns are assessed for their suitability (e.g. no
work on aseptic processing lines, personnel are expected signs of damage) prior to be being worn and once
to have completed aseptic process qualifications (such donned, prior to entering the cleanroom. There is a
as through a media fill). An exception is made for comment that visual inspection may not be sufficient
maintenance staff under exceptional circumstances, to assess gown integrity. However, no further advice
although procedures need to be in place to describe how is afforded, and this is something for which there is
this will be controlled. no obvious solution for, prior to entering a cleanroom
With qualifying operators, the language of the 2020 draft • Gowns must be processed in dedicated laundry
refers to ‘qualified’ and ‘unqualified’ operators (it is also facilities and the process of cleaning, evaluating and
possible for qualified operators to become disqualified sterilising gowns must be qualified
if there are concerns around microbial contamination).
Part of the qualification is a gown test and the movement
through a well-designed cleanroom system that seeks to
minimise contamination. The requirements for entering

8
For cleanroom operators entering aseptic processing safety implications to the patient of a loss of product
areas a gowning test is required (which is a combination sterility and in the basic elements of microbiology. As
of visual assessment and microbiological monitoring). well as requiring that personnel are suitably qualified
The new draft expands the list of recommended locations to work in cleanrooms, the new draft of the Annex
on an operator’s gown that require monitoring as part states that each facility must have staff who are
of the gowning qualification: hands, arms, chest and specifically experienced in microbiology, environmental
forehead. Each one of these locations presents a different monitoring regime and with conducting microbiological
microbial contamination risk, in terms of the types of investigations.
organisms and the route of contamination transfer. In
With aseptic operations specifically, the revised draft
addition, microbiological limits are presented for gown
places strong emphasis upon how operators are trained,
plates for the first time (these are afforded the same
especially in relation to any interventions with Grade A.
maximal values as finger plates). Further with gowning,
It is stressed that practices must not disrupt Grade A
the new draft now requires the maximum time that a
unidirectional airflow in terms of movement or with the
gown can be worn for to be defined.
placing of objects that might cause air disruption. To
All staff working in cleanrooms are expected to aid with operator training a recommendation is made
have knowledge of hygiene, cleanroom practices, that airflow visualisation studies constitute part of the
contamination control, aseptic techniques, and potential operator training program.

Microbiological Environmental Monitoring


The Annex also sets out the expectation for continuous
monitoring in relation to aseptic processing. This is
mandatory for Grade A and recommended for Grade B.
By continuous monitoring this means air samples (either
settle plates or volumetric air samplers).

Trending is an important consideration and the Annex


provides information about setting alert and action
levels and for looking for data patterns. Where action
level excursions occur, there is a necessity to investigate,
determine root cause and to set alert and action levels.
Trending is not only with numbers of colony forming
units; trending also applies to microbial speciation. For
microbial identification the draft Annex stipulates that all
isolates from Grade A and B areas should be identified,
and recommends that those isolated from Grade C and
D areas are identified. The Annex does not appear to
acknowledge that it is not always possible to obtain
an accurate identification result although the intent is
presumably for the microbiologist to try to obtain a
genus or species level outcome.
For viable monitoring expanded information is provided
in relation to sample site selection, stating that this needs To strengthen monitoring, a reference is made to
to be risk based and, where applicable, determined qualifying some aspects of the technologies used,
through a review of airflow visualisation studies. including verifying that contact plates are not providing
false counts due to the presence of disinfectant residues.
A change has been made to the EU GMP Grade A limit; Most users overcome this through the use of a neutraliser
which changes from 1 CFU to ‘no growth’. This change is being added to the agar.
both a reflection of the expectation that microorganisms
are not typically recovered from Grade A environments A limitation is placed on monitoring (and testing) being
(and that every recovery requires an investigation) and no substitute for a robust sterility assurance system. With
with the different types of techniques that could be this there is an emphasis that testing is too imprecise to
applied as replacements to the classic culture-based detect a weakness with the sterility assurance system.
methods, such as the use of rapid and automated As the draft states: “Exclusively monitoring or testing
microbiological methods are permitted provided the does not give assurance of sterility.” Instead, design and
facility has demonstrated their equivalency or superiority. control matter far more for patient safety.

9
Quality Control
Microbiological tests also feature in an expanded ‘quality on sampling from the beginning, middle and end of the
control’ section. These tests include the examination of batch, plus following any significant intervention. For
raw materials, where the specification applied should terminally sterilised products the worst case locations
be based on the required microbial quality as defined relate to areas like cold spots; and with lyophilised
in the contamination control strategy. Further tests products, samples need to be taken from different freeze-
include assessing intermediate bioburden of intermediate dryer loads.
product and a bioburden test conducted immediately
before the sterilising filter (for aseptically filled products)
or prior to terminal sterilisation. In both cases, the
samples should be representative of the worst-case
conditions, such as at the end of a process hold time.

The quality control section contains a discussion


around the sterility test. While full limitations with the
sterility test are not discussed in detail, the Annex does
emphasise that the value of the sterility test is diminished
if sterilisation and environmental controls are not in place.
To strengthen the sterility test, in terms of making the
test more representative, the draft text provides guidance

Filtration
Sterile filtration is a critical step for aseptically filled of contract sterilisation facilities; modes of transport;
products, where products are passed through a packaging of the sterilised filter, designed to prevent
μ0.22 μm filter, with pressure and time controlled damage to the filter during transportation and storage.
(following validation of the filter, where a bacterial Further controls around filtration include process
challenge has been used in the presence of product). A knowledge including the specific product type, including
key measure is an assessment of the product bioburden microbial challenge and whether there exists any risk of
prior to filtration. impact on filter integrity values, such as the potential to
alter integrity testing values and therefore prevent the
The issue of pre-use post sterilisation integrity testing
detection of a non-integral filter during a post-use filter
(PUPSIT) is set to apply to all large volume products.
integrity test. Finally, pre-filtration and processing steps,
For small volume products, a risk assessment can be
prior to the sterilising filter, must be in place to remove a
used, assessing the filter sterilisation process in terms of
microbial challenge and clarify the product prior to the
having controls in place ensuring that the risk of damage
sterile filtration.
to the filter is minimised. There is also a requirement to
understand the supply chain, such as having oversight

Product Inspection
Sterile, medicinal pharmaceutical products need to be impact of transportation, a further reference to Good
sterile, apyrogenic and particle free. This latter point Distribution Practices (GDP). The third point is that
relates to visual inspection, a subject that has not been microbial ingress studies (or alternative methods) should
covered in any great detail by the Annex before. Each be utilised to determine the acceptable stopper height
facility is required to have a list of critical deficiencies displacement.
– such as particle, hairs and turbidity – and to subject
operators to regular assessment. The assessment should
be under practical conditions, with control of inspection
time, line speed, and component size. To capture
operator fatigue the test should be executed at the end
of the shift.

There are several references made to container integrity.


The first is that containers sealed under vacuum
should be tested for maintenance of vacuum after an
appropriate, pre-determined period and during shelf life
(which relates to a stability studies). The second is that
container closure integrity testing needs to consider the

10
Cleaning and Disinfection

The references to disinfection have been expanded. The need to rotate between two different disinfectants remains,
although the fact that one of these should be a sporicidal agent is a new one. Reference is also made to disinfectant
qualification, for both cleanrooms and for transfer disinfection (the act of introducing items into cleanrooms or between
cleanrooms of different grades). Not only is disinfectant efficacy testing described as important the Annex infers that this
is a type of testing that needs to be carried out by a facility independently. One reason for this is because the different
types of surfaces found in different facilities, together with regional variations with the microbial flora. With the discussions
about disinfectants in section 5, references are made to the need to assess the bioburden of non-sterile disinfectants and
to assign expiry dates.

Further requirements are that:

• Validation studies relate to the specific manner in • Monitoring the effectiveness of disinfectants through
which disinfectants are use (by this there will be environmental monitoring does appear in the earlier
differences between sprays, sprays followed by draft; however, reference to “spore-resistant strains”
wiping, and pre-saturated wipes, for example) has been replaced with “organisms resistant to the
disinfection regime currently in use”, which is more
• Validation needs to be extended to the in-use expiry
scientifically accurate since a microorganism does
periods of prepared solutions
not always need to enter into a spore state to be
• It remains that disinfectants used in Grade A and difficult to kill. By resistance this does not refer to
B areas needs to be sterile; to this point the text ‘acquired resistance’ (for which there is little scientific
has been revised to state that disinfectants used in support) but to the fact of innate resistance, where
Grade C and D areas may need to be sterile (this some microorganisms are more resistant (such as
generalisation is not especially helpful) due to a factor of their cell wall, for example) to a
• There is a requirement that disinfectants that are not given disinfectant
purchased ready-made and which are thus made up
in-house as assessed for microbial contamination
(the time point is not specified, but presumably
this means end-of-use to represent ‘worst case’).
For ready-made disinfectants, assessment can be
undertaken by a review of certificates of analysis

11
Utilities

The section on utilities has a reference to heating systems As part of design controls the Annex requires that water
for the first time. There is also an added requirement remains in a state of turbulent flow through distribution
for the trending of critical utility parameters. This systems (this minimises the risk of microbial adhesion
is expressed as an important step so alarms can be and hence biofilm development) and that a sanitisation
reviewed to determine if they are specific or common method is used. As an example, holding water at 70°C or
causes, thereby enabling the appropriate engineering above is provided (although there are other forms water
action to be taken. With alarm-based monitoring control, such as chemical treatments). Another control
particular emphasis is placed on Water-for-Injections measure for water systems is with filtration. In section
systems where is stated technology for monitoring Total 6.11 the new text states: “Where WFI storage tanks are
Organic Carbon and conductivity is put in place, at equipped with hydrophobic bacteria retentive vent
locations determined by risk. This is because this type of filters, the filters should be sterilised and the integrity
process analytical technology that permits continuous of the filter tested before installation and after removal
monitoring is seen as superior to the type of discrete following use.”
monitoring that might be conducted by a chemistry
Where microbial counts occur, especially in response
department.
to an upward trend, a normal recourse is to disinfect
The main change with water systems follows on from the the system (by heat or by chemicals). In relation to this,
update to the European Pharmacopeia, permitting the the draft Annex states: “To minimise the risk of biofilm
production of WFI through methods other than distillation. formation, sterilisation or disinfection or regeneration
Due to unspecified concerns with biofilm growth on of water systems should be carried out according to
filters and the potential risk of endotoxin, the draft Annex a predetermined schedule and when microbial counts
makes a recommendation that further techniques such exceed action limits.”
as nanofiltration and ultra-filtration are considered in
conjunction with operating reverse osmosis membranes.

Summary
The latest draft Annex 1 expands considerably upon the current text. This white paper has considered many of the key
aspects, focusing on the changes that impact sterile products manufacture. No review can cover every aspect, and
readers are encouraged to read the proposed Annex in detail for themselves.

In summary, the key takeaways from the latest draft are:

• The expectation for each facility to have in place a formal, holistic contamination control strategy, focused on
minimising contamination control with respect to sterile manufacturing

• Additional requirements for cleanroom classification (beyond ISO requirements)

• A major focus on risk-based approaches

• Recommendations for the wider use of barrier technology

• A strong focus on personnel controls, such as gowning, and training

Such stipulations are unlikely to alter when the final version appears and it is recommended that sterile product
manufacturers invest some time in considering the impact of these changes upon their procedures and processes.

12
References
1. Eudralex The Rules Governing Medicinal Products in the European Union, Volume 4 EU Guidelines to Good
Manufacturing Practice Medicinal Products for Human and Veterinary Use. Annex 1 Manufacture of Sterile Medicinal
Products, European Commission, Brussels, Belgium, 2009:
https://ec.europa.eu/health/sites/health/files/files/eudralex/vol-4/2008_11_25_gmp-an1_en.pdf

2. EMA (2020) Second targeted stakeholders’ consultation on the revision of Annex 1, on manufacturing of sterile
medicinal products, of Eudralex volume 4, at: https://ec.europa.eu/health/medicinal_products/consultations/2020_
sterile_medicinal_products_en

3. ICH Q9 Quality Risk Management. ICH Harmonised Tripartite Guideline, Step 4, Geneva, Switzerland. 2005.

About the author


Dr Tim Sandle
Dr. Tim Sandle has over twenty-five years’ experience of microbiological research and
biopharmaceutical processing. He is a member of several editorials boards and he has
written over six-hundred book chapters, peer reviewed papers and technical articles
relating to microbiology. Dr. Sandle works for a pharmaceutical manufacturer in the UK,
and is a visiting tutor at both the University of Manchester and UCL.

13
Excellence in Science and Service

Reading Scientific Services Limited (RSSL), is an award-winning Contract Research Organisation (CRO) and winner of
Best CRO at 2019 OBN awards, we pride ourselves on our excellence in science, quality and service.
For over 30 years, we have been providing support to
the Pharmaceutical Sterile Manufacturing Industry and
recently launched Sterility Testing (membrane filtration
and direct inoculation), with Mycoplasma Testing to be
offered soon. Our expert team can also support with raw
material, vial and stopper testing to microbial analysis
such as TAMC/TYMC and endotoxin (LAL).

We work in partnership with our clients to ensure that


they meet the regulatory requirements both with routine
testing as well as more complex projects such as cleaning
validation and environmental monitoring, using the
wealth of experience from our multi-disciplinary team of
technical experts and consultants.

Sterile Manufacturing Support Services:

• Sterility Testing
• Endotoxin Testing
• Environmental Monitoring
• Raw Materials
• Vial and Stopper Testing
• Mycoplasma (coming soon)
• Investigative Problem Solving
• 24/7 Emergency Response Service
• Training and Consultancy
To find out more about how we can support your Sterile

Manufacturing or to discuss your needs further, please contact us on:


+44 (0)118 918 4076, email enquiries@rssl.com, or visit www.rssl.com

14
About Reading Scientific Services Ltd (RSSL)
RSSL is an award-winning Contract are wide ranging, and provide support
Research Organisation (CRO), through the full product lifecycle.
providing analytical, investigational,
RSSL is routinely inspected by the MHRA,
consultancy and training services to
FDA and UKAS which ensures that our
clients in the global pharmaceutical,
analytical services meet the needs of
biopharmaceutical and healthcare
industry. We are trusted by industry
sectors.
to provide a solution with scientific
Our chemical, physical, biochemical, excellence, outstanding customer service
biological and microbiological services and professionalism.

Contact us to find out more about our


expertise and how we can support you:
Tel: +44 (0)118 918 4076 Email: enquiries@rssl.com Web: www.rssl.com

© 2020 RSSL. All rights reserved.

Reading Scientific Services Ltd


The Reading Science Centre, Whiteknights Campus,
Pepper Lane, Reading, Berkshire RG6 6LA

15
View publication stats

Вам также может понравиться