ECA Good Practice Guide –
GMP Auditors Reference Handbook
– Attachment 3: Audit Areas and Criteria –
A guidance document by the ECA Foundation
Version 3.0; May 2025
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Contents
A. Pharmaceutical Quality System .................................................. 2
B. Equipment / Utilities / Systems .................................................. 6
C. Warehouse and Logistics ........................................................... 7
D. Premises ................................................................................. 7
E. Environmental Conditions .......................................................... 9
F. Handling of Goods .................................................................... 9
G. Processes .............................................................................. 10
H. Quality Control ....................................................................... 11
I. Microbiology........................................................................... 13
J. Evaluating Testing Process & Methods ....................................... 15
K. Evaluating major Equipment and Instruments ............................ 16
L. Utilities.................................................................................. 17
M. Maintenance .......................................................................... 18
N. Non-sterile and sterile Solid Dosage Forms Production Area ......... 18
O. Particulars for sterile Dosage Forms .......................................... 20
P. Water and Sanitisers ............................................................... 23
Q. How to audit a medical Cannabis Grower ................................... 23
R. Preparing Audits for Cell and Gene Therapies (CGT) based Advanced
Therapy Medicinal Products (ATMPs) ......................................... 26
1 ATMP-specific Challenges .........................................................................27
2 How to audit ATMPs - managing the Complexities of allogenic CGT
Manufacturing ........................................................................................27
2.1. The Challenges of the Supply Chain ........................................................................................... 27
2.2. Where to start at the Manufacturer .......................................................................................... 28
2.3. ATMP OOS Batches being used .................................................................................................. 28
2.4. QP Certification of ATMP and Import......................................................................................... 29
2.5. Transport and Storage of ATMPs ............................................................................................... 30
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 1
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
A. Pharmaceutical Quality System
A company’s pharmaceutical quality system (PQS) should describe the procedures and
controls in place to ensure reliable operations across the organisation.
The PQS should ensure all operations follow current GMP requirements and local
regulatory expectations and comply with licensing requirements.
Audits of the PQS are detailed in various guidelines, but especially in chapter one of the
EU-GMP guidelines. Examples of what to look for when auditing the PQS are indicated in
this chapter.
Aspect Possible details to evaluate
Structure of the PQS Consistency across the process?
Are all processes clearly defined?
Are elements of the PQS reviewed and approved by
management and the Quality Unit?
Are responsibilities clearly defined?
Regulatory status Is the company operating within its authorised licences?
Company organisation Is an organisational diagram in place illustrating roles and
responsibilities of key roles?
Is the company staffed appropriately?
Is the Quality Control Unit operating independently of
production?
Release system Who is responsible for batch confirmation, certification and
release?
Who actually performs batch confirmation, certification, and
release, and can decisions be made independently of other
management functions?
Is all relevant documentation and information related to each
batch available at the point of review, confirmation,
certification and release?
Are the functions involved appropriately resourced to ensure
a full overview?
Deviation handling Is there a clear definition of a deviation and how deviations
are classified (e.g. incident, minor, major, critical)? Note:
different organisations may use different terms when
referring to these processes, and this is acceptable as long as
the definitions are clear and all processes are covered.
Who is responsible for investigating and reporting deviations?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 2
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Who is responsible for reviewing and approving the
investigation report?
Who assesses the impact of the deviation on the final batch
before confirmation, certification and release?
Are flow charts available for associated processes?
What actions are taken when serious deviations are reported,
and how are actions justified and documented?
Are all personnel trained in the processes?
How many deviations are overdue, and how are they treated
(deviation, extension, etc.)?
Corrective and Preventive Are there clear definitions of the terms used (e.g. corrections,
Action (CAPA) corrective actions and preventive actions)?
Are CAPAs defined when needed? And if no CAPA is opened,
how is this justified / documented?
How are effectiveness checks realised?
Is the Quality Unit involved in defining, closing and approving
CAPAs?
Are all personnel trained in the processes?
Root cause analysis (RCA) Is the RCA process clearly defined in a procedure?
Has the organisation defined and provided useful RCA tools?
Are RCA processes logical and scientifically sound, and have
the potential root causes and contributing factors been
identified?
Are the defined CAPAs based on root causes?
How is the Quality Unit involved in defining, closing and
approving an RCA?
Are all personnel trained in the processes?
Are there any repeated RCAs (e.g. ‘human error’) reported
with no clear corrective action to resolve the issue?
Change Control Are types of changes and relevant responsibilities defined?
Is the process / are further batches halted until the change is
fully implemented (when appropriate)?
Is an impact analysis available for the change and each
action that leads to the change, and is there a requirement
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 3
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
for an effectiveness check for each change / action? If not,
how is this justified?
How many changes are overdue? How are they handled
(deviation or extension, etc.)?
Is the Quality Unit involved in closing and approving a
change?
Are all personnel trained in the processes?
Supplier management Does the audit programme define the frequency of audits? A
company may use a risk-based approach, as described in
chapter 3 of this guide. An auditor should check this approach
and the associated documentation.
Are the audits performed as defined in the programme?
Are the audits performed by trained auditors?
How is supplier performance monitored and linked to the
qualification?
Do the responsible positions (QP, Purchasing) have access to
relevant data and audit reports? Are they aware of possible
deficiencies and the corrective actions agreed upon?
Is there a formal assessment of the impact on product quality
when serious deficiencies are reported?
Are quality agreements in place for every outsourced
activity? Is the Quality Unit involved (e.g. in review and
approval)?
Training system Is there a GMP / GDP training programme for new
employees?
Is training recorded, and is it possible to track staff training
to the tasks they performed?
How is training realised (online, classroom, shop floor)?
Who trains, and what are the requirements to become a
trainer?
How are effectiveness checks done and documented?
How are personnel disqualified for processes, if necessary?
How is disqualification recorded and communicated?
Internal audits Is there an SOP in place to describe the IA process?
Is there an annual plan / schedule in place?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 4
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Does the company adhere to the schedule, and how are
overdue IAs addressed?
Who is performing IAs and how are the auditors trained?
Are the actions arising incorporated into the Corrective and
Preventive Action (CAPA) system?
Document management Is there a version control for each GMP-relevant document
including unique number and version number?
Are documents distributed and tracked appropriately?
Is the documentation hierarchy clear, and are documents
structured?
Are the documents reviewed periodically as defined in the
Document Management procedure, or earlier if required?
Is there a link between the Change Control and Document
Management procedures to ensure documents are reviewed
and updated if impacted by a change?
Quality Risk Management Is there a QRM procedure in place, and is it applied
(QRM) appropriately?
Which QRM tools are used, and are they appropriate?
Are all personnel trained in the processes?
Management review Does the management review process follow ICH Q10
requirements?
Is the management review conducted regularly based on a
pre-defined agenda covering all relevant quality-related
activities of the organisation?
Are key personnel involved?
Is the protocol / report signed off by management?
Are the actions arising incorporated into the CAPA system?
Product Quality Review Are Quality Reviews in place, and are they conducted
(PQR)/ Annual Product regularly?
Review (APR)
How is the QP involved?
Is the protocol / report signed off by management?
Are the actions arising incorporated into the corrective and
preventive action (CAPA) system?
Recall system Is there an SOP in place to describe the recall system?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 5
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Are roles and responsibilities in a recall process clearly
defined?
Who makes the final decision on a recall?
In case of a recall, who is informed, including the local
regulatory authority?
How are the activities documented?
Who makes the final decision on a recall?
Complaint handling How are complaints received by the company?
Are they investigated appropriately (see deviations, RCA,
CAPA, etc.)?
Are appropriate actions taken (CAPA)?
How is feedback provided to the complaint provider?
B. Equipment / Utilities / Systems
Aspect Possible details to evaluate
Qualification How is equipment designed, qualified, monitored and
maintained?
Is equipment qualified before use and re-qualified after any
significant changes or repairs?
Is equipment identified unambiguously?
How are cleaning processes defined, and are they validated?
How is equipment labelled?
Maintenance Is a maintenance programme in place?
Are maintenance plans (for individual equipment) in place?
Are pre-defined frequencies complied with?
Logbooks Are logbooks available?
Are they regularly checked by personnel who can draw
conclusions and, for example, adapt maintenance plans?
Outsourcing Are maintenance / calibration activities outsourced, and are
contracts in place?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 6
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Do external partners have to comply with the auditees’ SOPs,
and, if so, how are they kept up-to-date?
Status labelling Is the status (clean / to be cleaned) clearly identified?
Are holding times for clean / dirty equipment defined?
C. Warehouse and Logistics
Aspect Possible details to evaluate
Storage How are the product and its components stored before,
during and after the process?
What are the storage time limits and conditions for each
stage?
How are the storage areas controlled and monitored?
Transport How are the personnel, material, waste and product flows
managed in each area?
What are the transportation methods and routes used for
each item (unidirectional flow)?
How are the transportation conditions controlled and
monitored?
D. Premises
Aspect Possible details to evaluate
Warehouse How is access controlled?
What is the status and maintenance condition of the facility
(holes and cracks in walls and floors, broken windows, rusty
equipment, etc.)?
Are there water or solvent stains that might be caused by
incoming water during the rainy season or leakages in pipes /
vessels?
What is the overall quality of starting materials (missing
labels, rusty or damaged drums, insufficient sampling and
quality control tests)?
Is a status labelling of materials defined and adhered to?
Are cleaning processes in place and controlled?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 7
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Are there API drums from third parties which might just be
re-labelled?
Are areas, rooms, and key equipment qualified and key
processes validated?
Are medicinal products stored directly on the ground (making
sufficient cleaning impossible)?
Is there adequate separation between the receipt, dispatch
and storage areas?
Is there a defined room capacity for cool rooms, and is this
qualified and controlled?
Outer premises Is there a fence or other protective measures against
unauthorised access?
How are visitors registered?
Is there CCTV coverage of the outer area?
How are incoming and outgoing trucks registered?
Is there any outside storage or stacking of materials, and if
so, how is it justified and controlled?
External (contracted) Do contracted premises exist?
premises
Do contracted premises have their own certification?
How are these premises qualified and monitored?
Are transportation activities between sites validated and
monitored?
Pest control Is there an adequate system in place?
Is there an unregulated usage of biocides?
What happens if animals are caught?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 8
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
E. Environmental Conditions
Aspect Possible details to evaluate
Storage conditions How are storage conditions maintained in different climatic
conditions (heat in summer, cold periods in winter, rain
periods)
Are seasonal variations taken into consideration during
qualification and validation activities?
Is the equipment used to control or monitor environmental
conditions calibrated at defined intervals based on a risk
assessment?
How are (temperature) deviations managed?
Mapping Was an initial temperature mapping carried out before the
Note: For small premises consisting warehouse was put into use?
of a few square meters, an
assessment of potential risks (e.g. Was temperature mapping performed under representative
heaters) can be conducted and conditions?
temperature monitors positioned
accordingly. Are temperature monitoring devices located according to the
results of the mapping exercise (positioned in the areas that
experience extreme fluctuations)?
Is the mapping exercise repeated according to the results of
a risk assessment or whenever significant modifications are
made to the facility or the temperature controlling
equipment?
Have all relevant areas been mapped?
F. Handling of Goods
Aspect Possible details to evaluate
Returned goods If returned to stock, has it been demonstrated by the
Note: for medicinal products customer that products have been transported, stored and
requiring specific temperature handled in compliance with their specific storage
storage conditions such as low requirements?
temperature, returns to saleable
stock can only be made if there is If returned to stock, have they been examined and assessed
documented evidence that the
product has been stored under the
by a sufficiently trained and competent person authorised to
authorised conditions throughout the do so?
entire storage period. If any
deviation has occurred, a risk
assessment has to be performed to
determine the integrity of the
product.
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 9
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Dispensing and intermediate Are there multi-purpose drums for internal transport and
storage areas storage?
Is there a risk of mix-ups (labelling of raw materials and
intermediates)?
How is (cross-)contamination of drums avoided (dust,
multiple use, cleaning)?
Storage of solvents Which solvents are used and how are they stored (focus on
toxic solvents)?
How are tanks and pipes identified / labelled?
How is the recovery and testing of solvents performed?
How is (cross-)contamination of storage tanks avoided?
G. Processes
Aspect Possible details to evaluate
Qualification and validation Is there a list of critical equipment?
Is there a validation master plan (VMP)?
Is the company's approach to qualification and validation
correctly described?
Are there proper documentation requirements for the risk
assessment, validation plan, protocol, report?
How often is re-qualification realised, and how is it justified?
Are worst-case and bracketing approaches applied and
justified?
Are deviations treated appropriately in the various systems?
Is there a process in place to disqualify equipment, etc.?
Is there a possibility to use non-validated equipment /
processes?
How are the qualification and validation results recorded and
analysed?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 10
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
H. Quality Control
Auditing a quality control laboratory involves evaluating various areas such as, but not
limited to, the laboratory facility, including stability chambers, sample management,
equipment calibration and maintenance, result creation and reporting, OOS/OOE/OOT
investigations and storage of reagents and reference substances.
Aspect Possible details to evaluate
Laboratories How are the rooms classified, qualified and maintained?
How is the equipment classified, qualified, calibrated and
maintained?
Is there sufficient space for the preparation of samples and
reagent with good labelling control / documentation
practices?
How are waste and leftovers dealt with?
Are equipment logbooks available, and do they include
entries on at least the following: operations performed
(including product), maintenance (planned / breakdown /
calibration), checks and verifications, and cleaning?
Maintenance of analytical equipment
Equipment calibration In-house calibration:
Are personnel trained (training plan and records should be
clear)?
Traceability of the standard used.
External calibration:
• Is the service provider qualified / approved?
• How is equipment transported?
• How does the contractor transport and store the
standards?
• Traceability of the standard used.
Does the calibration certificate include the following?
• Equipment identification
• Results (pass / fail) – incl. specification
• Uncertainty measurements
• Traceability to international standard of measurement
• Limitation on use (if any)
• Date of calibration
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 11
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
• Signature of the accepting calibration company, if
outsourced
• Next due date for calibration
Does the calibration label include the following?
Equipment identifier
Date of calibration performed
Date of expiry / Date of next calibration
Performed by
Are out of limit / specification results investigated according
to an SOP, including notification / information and an impact
assessment on prior tests / results?
Reference substances / Are reference substances / standards correctly labelled, have
standards (RS) an expiry date and indicate purity on the label or CoA?
From where do RS originate?
Is there a certificate for each RS?
Are storage conditions for the RS adhered to?
Is there a usage log for each RS?
How are expiry dates handled?
Out of specification, out of Do processes comply with GMP requirements and instructions
trend and out of expectation / SOPs?
results (OOS, OOT, OOE)
How are such results investigated, recorded and reported?
Was all glassware and instrumentation retained in the state it
was used until results were deemed acceptable to report?
Is re-measurement only allowed using the approved protocol
which specify the purpose and intent of the test? It should
not be used to “fix” the result.
Is re-testing only allowed after both laboratory and
production investigations have been completed and both
have been deemed inconclusive?
Is re-sampling only allowed if evidence is available indicating
that the original sample was not taken correctly or
compromised before analytical testing started? This should be
done following the same procedure as the original sample.
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 12
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
I. Microbiology
When auditing microbiological laboratories, it is essential to ensure that processes and
methods comply with marketing authorisation and meet necessary standards and
requirements.
By following a systematic approach and thoroughly examining key areas such as
laboratory facilities, documentation, personnel, sample management, culture media,
reference cultures, sterilisation indicators, method validation, major equipment,
environmental monitoring and record keeping, auditors can ensure the laboratory's
quality management system is well-established and meets necessary requirements.
Note: this is a highly specialised subject, therefore the auditor conducting a full audit of
these facilities should have adequate knowledge of microbiology and microbiological test
methods or consult with / bring an expert.
Aspect Possible details to evaluate
Sample receipt Is there a dedicated area assigned for receipt of samples?
How does the company prevent sample mix-ups?
Are storage conditions observed where necessary? Sample
receipt requires adherence to defined storage requirements
and may include frozen (-80°C) or cold (5°C), a defined time
before cold storage and adherence to correct labelling.
Do sampling processes comply with GMP requirements and
instructions / SOPs? How is it documented (at minimum are
the lot number, expiry date, storage conditions and the
test(s) required)?
How are the samples labelled?
How is traceability ensured?
Where are the samples stored?
Sample preparation Do processes comply with GMP requirements and instructions
/ SOPs? How are they documented?
Are balances checked regularly within appropriate ranges and
masses, ensuring that they are situated on stable platforms,
are clean and unaffected by environmental conditions?
Is volumetric glassware of the correct grade used, washed
and stored?
Documentation Are laboratory notebooks assigned to specific individuals,
version controlled and appropriately stored and archived?
Are results sheets pre-printed, and what is the procedure for
this? What is the reconciliation process of pre-printed sheets?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 13
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Are specifications consistent with the dossier and / or quality
agreement?
Data and reporting How is raw data defined?
Are test results independently checked to ensure there are no
errors?
Is system suitability testing performed before the start of the
test?
How are reports reviewed and approved?
Who has the authority to approve reports, and how is this
defined?
How is data transferred to the certificate of analysis (CoA)?
How is it ensured that CoAs meet customer requirements /
specifications?
Are IT, computer system validation and data integrity
requirements being met?
Microbiology laboratory tour How are access, gowning and changing rooms managed and
checked?
Is the size, housekeeping and cleanliness of the laboratory
adequate?
Are there dedicated areas for sample and culture media
preparation, incubation, reference microbial cultures, bio-
hazardous waste collection, endotoxin and sterility testing,
sampling and material storage (e.g. freezers)?
Is equipment labelled, qualified and maintained? Are
logbooks available, and do they confirm these activities?
Personnel How is protective gowning practised, including hygienic hand
washing (e.g. the use of antimicrobial soap and duration),
disinfection and drying?
Are garments for the laboratories dedicated to the micro lab
fresh and sterilised, non-shedding and with no external
pockets? Do garments for sterility testing (sterile zone)
include closed footwear, a hair net, gloves, a mask and
goggles?
Sample management How are contaminated biohazardous samples, used culture
media, containers, plates and tubes decontaminated?
How is it ensured that the correct media is used for testing?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 14
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Culture media, including Is there an SOP for media preparation, and how are the
reference and working processes validated?
cultures
Have heat / cold shock studies for assessment of media
transportation been performed?
Which tests are run for identity and purity (genotypic
analysis)?
How is identity and purity confirmed for incoming cultures?
How long can the working culture be used under the defined
storage condition, and how was this defined?
Indicators for sterilisation / Which BI is used, and from where was it purchased?
biological indicators (BI)
Is the supplier qualified and regularly audited, and did the
supplier audit cover the verification of the D-value claimed by
the supplier?
Is the bacterial population (and purity) confirmed for every BI
lot?
Review procedure on Are standard solutions of strains suitable, available and
microbial identification controlled?
Is the database suitable (with representative organisms used
within the lab facility)?
J. Evaluating Testing Process & Methods
Aspect Possible details to evaluate
Sterility testing Are procedures for testing and transferring sterility samples
and media into the test environment in place (including
media usage, incubation time, closure integrity), and do they
comply with requirements?
What actions are taken after a sterility failure?
Bacterial endotoxin testing Are there procedures in place for preparation of a standard
endotoxin stock solution and for preparation of test solutions?
Is the test method validated appropriately, and how often is
validation / re-validation checked?
How is test sensitivity confirmed with a new LAL reagent lot?
Microbial limit testing Is there a procedure in place for a growth promotion test?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 15
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Is media use effective (media should not only recover the
target microorganism, but also other inhibitors)?
Is the incubation process sufficient (correct temperature,
duration and conditions)? How are the conditions defined?
K. Evaluating major Equipment and Instruments
Aspect Possible details to evaluate
Autoclave Is the sterilisation autoclave separated from the
decontaminating autoclave?
Is the autoclave qualified, including calibration of sensors, a
timer, type of thermocouples, etc.?
Are sterilisation processes validated?
Incubator Is air equally distributed (incl. during the heat-up phase)?
How are details like door opening (causing a temperature
drop) handled and documented?
Is there a log for temperature / humidity excursions?
Is the incubator qualified, including temperature / humidity
sensors, a timer, a gas supply and an alarm?
Was mapping performed for the location of the probes?
Water bath How is the water level monitored?
What quality of water is used (only distilled water should be
acceptable)?
How is closure integrity of samples controlled?
How are samples labelled?
How are samples cleaned after removing them from the
water bath and before testing?
How are the samples dried?
Biosafety cabinet Where is it placed, and how is cabinet seal integrity
controlled?
How are filter integrity tests and glove integrity tests
performed?
Is the decontamination process appropriate (including
frequency, monitoring, verification)?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 16
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
How is the decontamination agent chosen, controlled and
verified?
L. Utilities
Aspect Possible details to evaluate
Air and gas quality How are specifications for the quality of the gases used
defined?
How is the air and gas quality ensured in each area?
Air supply Do air supply systems provide adequate filtration, ventilation,
humidity, temperature control, air speed, number of air
exchanges per hour and unidirectional flow (in laminar flow
compartments)?
Do they meet the microbial and particulate limits for each
grade?
Do they have regular testing and (re-)validation?
Compressed air How is the compressed air quality ensured for pneumatic
purposes?
Does it meet the requirements for purity, dryness, pressure
and flow rate?
Does it have regular testing and validation? How were the
specifications set?
Steam How is the steam quality ensured for sterilisation purposes?
Does it meet the requirements for purity, dryness, pressure
and temperature?
Does it have regular testing and validation? How were the
specifications set?
Other gases (N2, etc.) How are other gases used in the process quality controlled?
Do they meet the requirements for purity, dryness, pressure
and flow rate?
Do they have regular testing and validation? How were the
specifications set?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 17
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
M. Maintenance
Aspect Possible details to evaluate
Maintenance and repair Are rooms maintained properly, employing suitable cleaning
and disinfection agents and procedures?
Are rooms qualified before use and re-qualified after any
significant changes or repairs?
Equipment maintenance Is there a procedure in place describing what the company
expects from preventative maintenance?
Who is authorised to realise it, and how is it documented?
Is there a clear definition for replacement parts and final
assessment / review of the impact of any parts? Is there a
handover process from an engineer to the laboratory?
How is it ensured that the equipment is performing as before
and if its validated status is maintained?
Is there a clear description of what parts are in the
programme and how often?
Are personnel qualified to perform the tasks in question?
How is the glassware washer maintained (drains check, sieve,
signs of wear, cleaning of trolleys and jet nozzles)?
N. Non-sterile and sterile Solid Dosage Forms Production Area
Solid dosage forms come in various styles such as powders, granules, tablets, capsules
and suppositories. Each form has its own unique characteristics and administration
methods. Therefore, it is important to consider these characteristics when auditing
facilities involved in the manufacture and testing of such products.
Sterile dosage forms are products that are intended to be administered bypassing the
patient’s digestion system, making them a potentially life-threatening source of infections
if they are contaminated with microorganisms and pyrogens. Usually they are
administered by injection, infusion, implantation or inhalation. To ensure the quality and
safety of these products (sterility), auditors need to ask key questions and focus on
certain areas during an audit.
When auditing the production of these dosage forms, it is important to pay attention to
the specific requirements relating to the product. This includes ensuring that the raw
materials used in production meet the required specifications, the processes follow the
established validated processes and that the finished product is fully produced (and
tested) in line with the Marketing Authorisation (MA) and cGMP requirements.
If an auditor has access to the relevant technical sections (Module 2, 3) of the product
Marketing Authorisation (MA), the auditor can check the manufacturing documentation
against these documents to identify any non-compliance issues.
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 18
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
To review the manufacturing process, the auditor may select a batch based on one of the
following:
o A batch that was reported in a non-compliance investigation or a deviation /
complaint / OOS, etc.
o A batch that was impacted by significant change (equipment, procedures, layout,
etc.)
o A re-processed or reworked batch
o A rejected batch
o Other considerations (new product, volume, specificity, etc.)
o Or simply selection of one of the last three batches manufactured recently.
Alternatively, auditors can review a proposed batch by the auditee, but this may not
guarantee a representative document. Auditors should check if there is a system
controlling the 'blank' batch records and check if that is controlled by the change control
procedure. The auditor can then check the compliance of this template with the
validation documentation or MA. Then the selected batch record should be checked to
ensure the process is followed correctly.
When auditing without having access to the MA, auditors can use process validation as a
starting point. If there is no access to the MA and the process validation, an auditor can
review the manufacturing process as per the evaluation focus areas.
Aspect Possible details to evaluate
Audit with Marketing Does process validation (batch size, scale up, yield) follow
Authorisation (MA) the defined master formula?
Do executed batch records correspond to the master batch
records (including packaging)?
Do critical IPC test results and acceptance limits match with
pre-defined IPC checks and acceptance limits?
Do QC test methods, incl. IPC, comply with validated
methods?
Do QC test results comply with product specifications?
Is the holding time of bulk product(s) documented and
justified?
Was the equipment used for manufacturing also used during
validation (type, model, including packaging)?
Are listed suppliers of starting materials qualified?
Audit without marketing Process validation in comparison with master batch records
authorisation (MA)
• Manufacturing process flow
• Equipment and rooms
• Starting / Raw materials
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 19
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
• Sampling
• IPC checks
• Acceptance criteria
• Environmental conditions
• Cleaning process
Audit without Marketing Check overall GMP compliance
Authorisation (MA) or
process validation
O. Particulars for sterile Dosage Forms
Aspect Possible details to evaluate
Room design Are rooms designed to follow a clean room cascade,
preventing contamination from lower-grade areas to higher-
grade areas?
How are the (air over-) pressure cascades established and
controlled? Do they prevent the ingress of contaminants from
lower-grade areas to higher-grade areas? Do they account for
the different activities and operations in each area?
How are the rooms classified, qualified and maintained?
Do they comply with the relevant standards and regulations,
such as Annex 1 and the US FDA aseptic guide?
Are rooms easy to clean and disinfect?
Are the rooms free from drains in grade A and B areas (as
they can be a source of microbial contamination)?
Monitoring Are the rooms monitored regularly for microbial and
particulate counts using appropriate methods and locations?
How are rooms, environment, surfaces monitored?
What methods and instruments are used for monitoring?
How often and where are the samples taken?
How are the results recorded and analysed?
Contamination control How is contamination and degradation of the product and its
components prevented?
Are there adequate cleaning and sterilisation processes and
procedures in place?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 20
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Are cleaning and sterilisation processes validated?
Personal hygiene How are personnel adequately monitored and controlled?
Do personnel follow strict hygiene and gowning requirements
for each area and perform their tasks correctly and
consistently following standard operating procedures (SOPs)
based on applicable GMPs? How is this ensured?
Do personnel undergo regular media fill tests to demonstrate
their aseptic skills?
When (i.e. based on which criteria) are qualified personnel
dis-qualified to continue working in the aseptic environment?
How is it ensured that personnel minimise their interventions
and movements in critical areas where aseptic operations
take place?
Aseptic process simulation How are media fills performed to simulate the aseptic filling
process according to specifications and instructions?
What are the media fill methods, materials and frequency
used for each product type and container size?
How are the media fill results recorded and analysed?
Filling How is the product filled into the final containers, and does it
comply with GMP requirements and instructions / SOPs?
Are filling processes validated appropriately and re-validated
after any significant changes or modifications?
What are the filling methods, equipment and parameters
used for each product type and container size? Do they
comply with GMP requirements, process validation and MA?
How are the filling accuracy, uniformity and sterility ensured
and verified?
Are filling processes simulated using media fills to
demonstrate aseptic performance?
Lyophilisation How is the product lyophilised, and does it comply with GMP
requirements and instructions / SOPs?
What are the lyophilisation methods, equipment, and
parameters used for each product type and container size?
Do they comply with GMP requirements, process validation,
and MA?
How are the lyophilisation cycle, temperature, pressure, and
duration controlled and measured?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 21
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
How are the lyophilised product characteristics, such as
appearance, moisture content and reconstitution time,
ensured and verified?
Crimping and container Do the processes employed comply with GMP requirements
closure integrity (CCI) and instructions / SOPs?
What are the crimping methods, equipment and parameters
used for each product type and container size? Do they
comply with GMP requirements, process validation and MA?
How are the crimping quality, uniformity and integrity
ensured and verified?
How is the container closure integrity (CCI) tested and
validated for each product type and container size?
Sterile filtration Do filtration processes comply with GMP requirements and
instructions / SOPs?
What are the filtration methods, equipment and materials
used for each product type and container size?
How are the filter integrity, efficiency and compatibility
ensured and verified?
Sterilisation Do sterilisation processes comply with GMP requirements and
instructions / SOPs?
What are the sterilisation methods, equipment and materials
used for each product type and equipment?
How are the sterilisation efficacy, uniformity and compatibility
ensured and verified?
Sterilisation of containers Do the sterilisation processes used comply with GMP
Note: Most companies speak about requirements and instructions / SOPs?
“Depyrogenation”, when
“Sterilisation” of containers is the What are the sterilisation methods, equipment and materials
target. Depyrogenation is something used for each item?
that happens when glass containers
undergo thermal treatment for How are the sterilisation efficacy, uniformity and compatibility
sterilisation. Containers may be
“free from pyrogens” – but this does
ensured and verified?
not necessarily mean that they are
sterile!
Visual inspection Do visual inspection processes comply with GMP
requirements and instructions / SOPs?
What are the visual inspection methods, equipment and
materials used for each product type and container size?
How are the visual inspection results recorded and analysed?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 22
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
P. Water and Sanitisers
Aspect Possible details to evaluate
Water What quality of water is used for which purpose?
How are the water samples taken (where and when;
rationale)?
Where are the sampling points (rationale)?
Which sample containers are used (glass, sterilised, labelled,
identified; rationale)?
What is the max. time of storage prior to testing? Is it
controlled?
Sanitiser What sanitisers are used and for what purpose (type and
temperature)?
How are sanitising solutions prepared, approved and stored?
How is effectiveness checked and verified?
How are contact time and drying method and time controlled?
Q. How to audit a medical Cannabis Grower
Principles and Background
Auditing a medical cannabis grower introduces several new concepts at the edges of GMP
such as hydroponic cultivation systems, good agricultural collection practice (GACP) and
increased demand for site and product security measures. Some authorities inspect both
GACP and GMP, but others solely inspect GMP. The scope here is mainly limited to GMP
considerations.
Some authorities regard cannabis flowers as APIs, whereas others treat them as
intermediate products and some as finished products. This is an evolving area and the
inspector needs to know the intended markets current requirements.
Laboratory testing of cannabis products frequently needs several test laboratories, and
these are often contractors. Testing facilities at growing sites are often absent or very
basic.
Relevant sections of Eudralex Vol 4 should be followed and include non-sterile production
topics plus relevant annexes such as Annex 7 (Herbals), Annex 11 (Computers) and
Annex 15 (Validation).
Most sites have sophisticated plant monitoring measures to satisfy narcotics regulations.
However there is still a potential for mix-ups, as some sites grow plants from seed,
others may purchase plants and others may clone plants. Normal GMP principles of
segregation, identification and labelling should be included in the inspection.
Annex 7 refers to the Guideline on Good Agricultural and collection practice for starting
materials of herbal origin. The inspector is recommended to read this document before a
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 23
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
GMP inspection. Annex 7 provides advice on the borderlines between GACP and Part I
and Part II of Eudralex Vol 4. The document reference is EMEA/HMPC/246816/2005.
Some products may be irradiated, with current examples being some products exported
from Canada. Practices differ within the EC. The inspector may have to examine results
and certification from a contracted irradiation facility. Annex 12 is useful in this case. The
effects of irradiation on the multiple components of cannabis, such as terpenes, is the
subject of debate. If products are irradiated, the inspector should ensure that the
recipient is notified of this.
Products may not be the subject of QP release due to their current status. In addition,
QC staff at these sites may be less experienced than those in a mainstream
pharmaceutical factory. Cannabis products can present substantial patient risks from
aflatoxins, ochratoxins and microbial contaminants. Inspectors should ensure that the
finished product specification is appropriate and results are being adequately monitored.
Prior to inspection of these sites, and especially for inspectors new to this topic, it is
recommended that preparation is started early so that any questions can be answered
before commencing the inspection.
Aspect Possible details to evaluate
Preparation What is the sites determined cut-off between GACP and GMP?
Is there a formal QMS for GACP?
Are plants bought in, or grown at the site?
Is the site activity limited to flower growing, or are there
additional activities such as extraction and formulation?
What narcotics licences are held or applied for?
Does the company use hydroponics or soil growing?
What laboratories are used for chemical and microbial
testing?
Is there 24/7 security with manned presence and a security
plan?
Goods received and storage Are growth media received with suitable certification?
Are chemical nutrients suitably labelled and stored to prevent
degradation?
Do the company adequately address TSE risks in all potential
product-contact items?
Are any printed materials received and suitably checked and
securely stored?
Facility design Are rooms classified to expected GMP standards for non-
steriles?
Are suitable pressure cascades maintained and monitored?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 24
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
Are air inlets and outlets located to achieve suitable sweeping
and absence of stagnant areas?
What filtration standards are applied to both incoming and
recirculated air?
Is a suitable area provided for cleaning equipment and
solutions?
Clothing Is a suitable changing area provided with adequate space and
washing facility?
What measures are in place to protect workers and inspectors
from light radiation - eye protection, special clothing and
sunscreen?
Are any external laundry facilities audited and approved?
Fertigation controls Is there an automated system controlling key parameters
such as solution preparation, irrigation, temperatures, light
and gas levels?
Has the system been formally validated and approved?
Request and examination calibration records of key sensors.
What controls are in place to stop unauthorised changes or
optimisation?
Pest controls and fungus What pest control measures are in place and how is their
risks efficacy challenged?
What measures are in place to reduce/discourage fungal
growth?
Harvesting, drying, trimming How is flowering monitored to determine a suitable and
and curing consistent point for harvesting?
Are machines used for bucking and trimming?
Is there sufficient instrumentation on automatic machines?
Is there an adequate procedure for cleaning and sanitising
machinery?
Packaging and labelling Is the packing process manual or is machinery required?
What containers are used for the product – are these bought
in or is a local pouch produced?
Is there any use of nitrogen in the packing process and, if so,
is this subject to point-of-use filtration?
Are suitable in-process checks conducted during packing?
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 25
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
Aspect Possible details to evaluate
If pouches are used, what checks are carried out for pinholes
and integrity of the seal?
Are product labels printed fully in house, or just variable data
such as the batch number and expiry date added locally?
QC testing Are the water sources evaluated for pesticides, heavy metals
and micro levels?
Is there an adequate environmental monitoring programme
in place?
Does the company check for the absence of aspergillus in the
finished product?
Does the company monitor and trend levels of aflatoxins and
ochratoxins in the finished product?
Is there a requirement for the absence of salmonella and
E.coli in the product?
Are cannabinol levels in the finished product controlled and
evaluated?
Are the qualifications and experience of the releasing officer
or QP adequate?
R. Preparing Audits for Cell and Gene Therapies (CGT) based
Advanced Therapy Medicinal Products (ATMPs)
Background
The European Commission has published a separate regulation for Advanced Therapeutic
Medicinal Products (ATMPs) which lays down specific rules concerning the authorisation,
supervision and pharmacovigilance of ATMPs (Regulation 1394/2007). The regulation
defines three types of ATMPs: gene therapy medicines, somatic cell medicines and tissue
engineered medicines.
This article is mainly focused on ATMP based cell and gene therapies (CGT). CGTs can be
described as therapies that offer personalised and targeted treatments for a number of
rare and chronic diseases. These therapies are classified into two main types: autologous
and allogenic.
Autologous CGTs involve using the patient’s own cells, which are collected, modified and
then reintroduced into the patient. This personalised approach minimizes the risk of
immune rejection.
Allogenic CGTs, on the other hand, use cells from a donor. These therapies can be
produced in larger quantities and administered to multiple patients, but they carry the
risk of immune response and require careful matching of donor and recipient.
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 26
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
1 ATMP-specific Challenges
Due to the specific nature of ATMPs, new challenges need to be addressed. These new
challenges and difficulties regarding sterility, out-of-specification handling, batch release
and EU import testing are situated throughout the entire manufacturing process.
One of the major challenges is the aseptic risk associated with the ATMP manufacturing.
ATMP administration requires injection, infusion or transplantation of the medicinal
product. Therefore, the ATMP product needs to be manufactured under aseptic
conditions. Unlike traditional small molecule medicinal products, sterile filtration or
terminal sterilisation is in most cases impossible, either because these methods impede
the product’s structure or function, or because cells are simply too large to pass through
the pore size of the filter. As stated in Part IV of the EU GMPs (Good Manufacturing
Practice), all processes related to aseptic processing need to be validated before entering
clinical phase I trials. The combination of gowning requirements, clean room qualification
and validation of these aseptic processes by media fill and a validated sterility test
method needs to ensure the sterility of the product and, therefore, requires specific
attention during an audit according to Annex 1, as applicable.
2 How to audit ATMPs - managing the Complexities of allogenic
CGT Manufacturing
2.1. The Challenges of the Supply Chain
The creation of a CGT based ATMP involves the collection or “donation” from an
individual. The donated material is then generally modified, engineered or manufactured
into a therapy product for delivery to the patient. This complex manufacturing and
delivery model is known as the “vein-to-vein supply chain”.
CGTs have not yet been adopted on a large scale, due to a combination of clinical,
regulatory and cost-related hurdles. At present, supply chains are necessarily bespoke
and costly. However, given the potential presented by CGTs and the current focus on
developing them, it seems likely that they will form an integral part of future therapies.
As they become more prevalent, ensuring the ongoing integrity of their supply chains will
be of paramount importance. This also includes the storage and transport of them.
Right from cell collection to the delivery of the therapy product, each step involves
several processes which bring their own risks and require controls.
During the collection process (via apheresis, etc.), preparing and monitoring the patient
and equipment to yield the appropriate quality raw material is essential.
• The raw material and drug products can get affected by factors such as timing,
temperature, microbiological basic load, packaging, etc. and therefore need to be
handled, packed and transported with care. Obtaining a sterile starting material
can be a challenge.
• Certain complex manipulations and activities are performed during the
manufacturing process and are crucial if the end product is to meet specifications.
These are often performed manually and require great skill and consistency.
• Understanding the GMP requirements, equipping the team with the necessary
skills and rigorous training is critical.
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 27
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
• Lot genealogy, including chain of identity (COI) and chain of custody (COC), are
two critical data sets that maintain the record of all the materials, processes,
testing and outputs of a batch to ensure that patients are treated using the
specific product meant for them.
• It must be ensured that the qualification and performance of vendors meet the
required standards, especially since many of them do not operate in a GMP
environment.
• Standardised processes and controls ensure that the products are safe and fit for
the intended use and comply with regulatory requirements.
• Risks have to be identified, and mitigation measures established using an
integrated process for both quality and risk management.
• Continuous improvement has to be set up through monitoring of processes and
performance, track and trend investigations, root cause and CAPA analysing of
supply chain performance and managing risks.
A robust audit process is critical for ensuring patient safety, mitigating risks, reducing
failure and increasing operational efficiency.
2.2. Where to start at the Manufacturer
When manufacturing an ATMP, it is important to pay attention to the choice of raw
materials used. These materials influence the entire process, from clinical development
to market authorisation. GMP guidelines contain important considerations.
This requires specialist expertise, with careful consideration and impact assessment
undertaken, which is not routinely available for the dense supplier management needed.
Agreements with suitable suppliers such as blood centres are required to facilitate
delivery of these specialist materials to be of suitable specified quality. Audits and
continuing oversight arrangements should confirm the technical quality and suitability of
the services documented in a quality and technical agreement.
Therefore, when auditing manufacturers, an essential focus should be on raw material
supplier management.
2.3. ATMP OOS Batches being used
Due to a higher variability and the quality of raw materials, a higher level of deviations
and OOS results is more likely. In case of an OOS result, there is the possibility to
release the batch and administrate it to the patient. This contrasts with traditional
medicinal products, where a product which does not comply with the specifications
cannot be certified and released.
During an audit the following should be checked with regard to OOS documentation:
• A description of the OOS with a risk analysis by the manufacturer on the impact
on the following:
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 28
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
• The quality of the product, including the percentage of batches that is OOS on
the total of commenced productions and a reflection on the production process
(root cause analysis and CAPAs (Corrective Action and Preventive Action)),
• Role of the QP
• The safety and efficacy of the product in relation to the patient;
• There should also be a feedback loop in place to the critical quality attributes
(CQAs), which have been identified during the quality target product profile
(QTTP) process to define the quality, safety and efficacy attributes of the
desired final product during the risk-based process development (e.g. quality
by design)
• A confirmation of the physician to the manufacturer to accept the product;
• A declaration from the MAH (marketing authorisation holder) as to whether or not
the product has been administered.
A Q&A document on how to handle an OOS for authorised batches was first published by
the EMA in April 2019.
2.4. QP Certification of ATMP and Import
According to EU directive 2001/83/EC, a registered qualified person (QP) is responsible
for the certification of medicinal products prior to their release. This and the applicable
responsibilities of a QP do not differ for ATMPs. However, the difficulties which might be
faced by a QP are different throughout the entire manufacturing process.
• A QP cannot certify an OOS ATMP batch, the release of these batches is based on
a documented request by a treating physician.
The QP needs to assure that verification according to GMP Part IV – 11.27 has
been performed. Prerequisites to release the OOS batch are mentioned in GMP
Part IV – 11.5.
As a reminder, QP certification is a written signed statement that the manufacture
and testing of a batch has been conducted in accordance with GMP and the
relevant approved marketing authorisation or clinical trial application before the
batch can be released.
• The obligation to perform import testing in case a medicinal product is
manufactured in a third country, does not always apply to ATMPs (e.g. for
ATIMPs, import waiver). There are exemptions from EU batch retesting laid down
in paragraph 11.17 of GMP Part IV. In case of exemptions, the manufacturing of
the ATMP needs to be performed according to EU GMP. This implies that an audit
prior to first importation is required. As the QP takes on the responsibility of
certifying this batch from a third country, the audit is preferably performed by the
QP. A Q&A document on the exemption from batch controls carried out on ATMPs
imported into the European Union from a third country was published first by the
EMA in July 2019
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 29
ECA Good Practice Guide –
GMP Auditors Reference Handbook
Attachment 3 – Audit Areas and Criteria
2.5. Transport and Storage of ATMPs
Much of the discussion in the ATMP industry today focuses on the complexity of
manufacturing. However, the ultimate success of an ATMP rests on the ability to deliver a
viable, potent product to the patient.
In order to ensure that this “living” drug is delivered to the right patient at the right time,
location and temperature is essential to patient safety and product effectiveness and
therefore should not be forgotten when auditing.
The logistics of these types of medicines are complex, due to specific requirements
regarding temperature, reliability and traceability. For example:
• Frozen storage requirements: Cryogenic storage and distribution capabilities are
established for many ATMPs. Distribution in deep-frozen or cryogenic conditions
needs specialist shippers, to keep the temperature stable and track the location
and conditions during the shipment.
• Short shelf lives —ATMP or their starting materials may have an extremely short
shelf life. This could even mean that not all test results are available at the time
points of release or even administration. A risk assessment, risk mitigation and
processes (e.g. what happens if a specification is not met) need to be in place.
Any deviation from storage, distribution or traceability requirements has the potential to
impact the quality of the ATMP and, therefore, the safety of patients. Storage and
distribution under GMP and good distribution practice (GDP) guidelines are applicable for
CGTs, too.
The challenges of CGT for pharma logistics are as followed:
• Delays due to inspection and compliance checks
• Complexity calls for experienced logistics partners
• Limited visibility in logistics because of low quantity
• Scalability of transport methods
• Unforeseen disruptions in supply chains
© Copyright: ECA Foundation, Germany – Copying, editing and distributing require the written agreement of the ECA Foundation 30