Wednesday, November 25, 2009

General Guidance for Process Validation Sampling

2
􀃭 Sensitive/labile products (light, temperature, time, oxygen, moisture).
􀃭 Mixtures prone to segregation/separation.
b) Number of Samples, Sample Size and Population: Samples should be
representative of the population. Some materials may not homogenous due to
segregation that occurs during transport, or handling; variability occurring during
the manufacturing process; and a variety of other factors that might impact a
representative sample. Samples or sampling plans are often based on statistical
criteria and the use of an appropriate statistically-based sampling plan can be
important to ensure the sample is representative of the population. Each sampling
plan should be developed to consider the specific attributes being measured and
the risks associated with accepting a defective lot.
· Samples should relate to evaluation of critical quality attributes and the
critical process parameters.
· Use sampling plans designed to obtain a representative sample from the
product being evaluated or cite reference to an established sampling plan (e.g.
site procedure, or ANSI/ASQ standards for inspection of attributes in
packaging validation for example).
· Ensure that an adequate amount of sample is available to complete all tests.
The number of tests per sample should be determined in consultation with the
laboratory performing the testing and specified in the sampling plan or
protocol. One test per sample may be preferable when sample material is
inexpensive or there is a risk of cross-contamination when performing
multiple tests on the same sample.
· Collect sufficient reserve sample, when possible, (e.g. >2x normal amount) to
support potential investigation, except for sterility testing.
c) Sample collection, handling, and identification: Proper sample collection,
handling and identification are vital to the success of the validation and the
guidance provided by Sampling of Production Materials and Finished Goods
should be followed.

Inspection Attributes in Packaging Validation of Non-Sterile Drug Products

An important aspect of AQLs and UQLs is the continuous learning and possible
adjustments of defect descriptions and levels. Failure to meet established defect limits is
investigated to determine the impact on validation. As events and history of the packaged
product and process are gained, changes may be warranted. Re-evaluation of the attribute
description (e.g. quantitative measurements enhanced or more specific description of the
defect) and acceptability by the Quality Unit of that defect and corresponding acceptance
criteria may be beneficial. Trending, quality incidents and investigations and statistical
treatment of inspection data are a means to review the defects. Quality risk management
tools may also be used to provide a basis for evaluating the potential impact of package
defects.
Definitions of classifications:
Common defect classification criteria for critical, major and minor and its impact on the
safety, regulations, use, consumer relations and company are shown in Appendix I.
Sampling Plans:
Typical sampling plans that can be used are General Inspection Level II (ANSI/ASQC
Z1.4-1993), with Single, Normal Sampling Plan or ISO 2859-4 (10). Other sampling
plans may be appropriate depending on administrative difficulty of the ensuing sample
size, desired or given AQL, sample size of the available plan, packaging history and
routine monitoring intentions.
Sample size of multiple plans is less than double sampling plans, which in turn is less
than the single sample plans. Once determined, the total sample size is divided by the
number of sampling intervals to determine the number of samples per interval (e.g. 200
bottles (sample size) /24 intervals = 9 bottles/interval).

Matrices and Bracketing in Process Validation

Matrixing across different products may be applied to the packaging validation of the
final dosage form, for example to evaluate the packaging of different products in a
common packaging presentation. As with other uses of bracketing and matrixing, the risk
of using this strategy for the potential products encompassed by the matrixing plan
should be considered, documented and approved.
The use of bracketing/matrixing for the validation of a manufacturing process across
different products should be approached with caution because of the risk of overlooking
other possible affects of the change. Use of this type of bracketing/matrixing requires a
good understanding of the processes involved and the risks being assumed. For example,
in the evaluation of a change of a critical material for different products, the excipient
interactions, critical process parameters and critical quality attributes (CQAs) are not
necessarily the same for each product. The effect of the change in the CQAs may be
different for each product. A product sensitive to the change may experience a failure in a
CQA (e.g. dissolution) while in a case of a product not sensitive to
the change, it may experience no effect at all in its CQAs.
To obtain the maximum benefit with minimum risk from bracketing and matrices, it is
necessary to have a well-developed understanding of the impact of critical process
parameters on critical quality attributes. There should be a documented and justified
rationale that explains why one set of test conditions (e.g., manufacturing process,
product presentation, etc.) is representative of one or more related test conditions.
Typically, the rationale is addressed by selecting parameters and/or products that
represent the edges of a range or 􀂳worst case􀂴 of allowable conditions. The rationale and
justification for the bracketing/matrixing strategy to be used in validating a process
should be provided in the validation protocol, or in another document referenced in the
protocol.
Depending on the circumstances, prospective and concurrent validation approaches may
be used for validating a process using bracketing or matrixing. If a concurrent approach is
used, an interim report provides a summary of the results obtained for a product batch, in
order to justify the validation and release of one of the product presentations within the
bracket/matrix. This approach may also assist in approving the manufacturing and/or
release of additional batches of a particular presentation. At the completion of the
validation, the validation report will address all batches.
At present, some regulatory authorities may not accept the use of bracketing or matrixing
for validation. Japan, for example, currently requires that all combinations be validated

Performance Qualification versus Process Validation

Robustness is the ability of a process to consistently produce the same product while
remaining unaffected by small variations in the process. It is possible to have a repeatable
process and not have a robust process, but it is unlikely to have a robust process that is
not repeatable.
One approach to evaluating robustness of a process is to evaluate challenge conditions
during a separate PQ study. In this approach, challenging the validity of the Normal
Operating Range limits is considered Performance Qualification (PQ). This
type of PQ is considered 􀂵worst-case􀂶 or 􀂵most appropriate challenge􀂶 of the parameter
limits.
The number of PQ batches may vary depending on the number and range of critical
parameters to be qualified.
Another (more common) approach is to obtain information on challenge conditions
during process development. The PQ or process development studies are then followed
by Process Validation (PV). In the PV, all process parameters are set to target and
typically 3 consecutive batches are produced to satisfy the requirement for
reproducibility.
The ranges of the critical process parameters must be defined. They may be defined
either during performance qualification or during process development.
Example 1 (PQ for a process)
It has been demonstrated in the pilot plant that lubricant blending for 7 minutes leads to
acceptable product. Seven minutes is chosen as the target lubricant blend time for the
regulatory filing. The production blender with an automated timer is qualified for +/-15
seconds (0.25 minute) and 7 minutes is target blend time. In the PQ, blend time should be
qualified at times above and below the target (e.g. 5 and 9 minutes). Then, in PV, the
three validation lots would be manufactured at target (NOR range would be 6.75 -7.25
minutes).
The PQ has confirmed the limits and the PV have been performed in triplicate at target.
Therefore, data has been obtained demonstrating the robustness of the NOR/regulatory
range and reproducibility of the process.
Example 2 (PQ for a system)
A reactor is controlled by an automated DCS (distributed control system) that is a direct
impact system as defined in the project plan. During OQ portions of the DCS and the
reactor are tested individually. The Performance Qualification (PQ) portion of the system
qualification is defined to execute critical functional testing to demonstrate that the
systems perform reproducibly in the normal manufacturing environment. This would
typically include using water and/or solvent runs in the reactor while evaluating the SOPs
(Standard Operating Procedures). The PV of product processes in the reactor will follow
the successful reactor PQ.

Test Deviations during Validation

Document the deviation
The deviation should be documented according to the applicable procedure or protocol.
This should include assignment of a reference number to the deviation, the test section
(and run number, where applicable), the test step (where applicable), a description of the
deviation and the signature and date of the person recording the deviation.
Although errors may be grouped by type, repetitive protocol errors are the most
practically grouped in this way. When deviations are grouped, care should be taken to
ensure that the link between each individual issue identified, its investigation, and
corrective actions is clear. In addition, all issues will need to be resolved (or transferred
to a tracking system) to allow closure of the deviation.
Investigation
An investigation should be conducted to determine the root cause of the deviation. In
many cases, this will not require any in-depth analysis. However, for system or process
failures, a formal investigation including appropriate technical representatives may be
required. Such a detailed investigation should be conducted according to applicable
procedures.
The investigation may be used to identify the type of deviation; this can be documented
descriptively on the form or by using check boxes. Examples of types of deviations could
include:
- System/Process Error -An actual problem with the functionality of the system
or a technical problem with the process, whereby the system or process does
not meet the acceptance criteria when the test step is executed.
- Protocol Error -The test step instructions require addition, deletion,
modification and/or clarification due to missing/incorrect/ambiguous test
instructions. Or the acceptance criteria require addition, deletion, modification
and/or clarification due to missing/incorrect/ambiguous acceptance criteria.
- Other -A deviation occurs that is not related to the test method, acceptance
criteria or system, for example, operator error.
Protocol errors that require correction in order to allow the test to be executed as intended
may include missing test instructions or incorrect acceptance criteria.

Validation Documentation

Testing Documentation
Documentation, such as protocols or test scripts, should be developed that specifies how
the validation study will be conducted. Testing documentation should contain or
reference the following information, as applicable:
o Title and unique identification number;
o References to related documents such as the validation planning document
and SOPs;
o Objectives and scope of the study;
o Prerequisites (e.g. qualified equipment for process validation or
Installation Qualification with no major deviations prior to Operational
Qualification)
o Clear, precise definition, or reference to same, of the system or process to
be validated, for example:
o Summary and/or process flow diagram of critical processing steps
included in the study;
o The Master Manufacturing instructions or Device Master Record to be
validated (i.e., that to be used in preparation of validation lots or batches);
o The critical process parameters (CPPs) for the process steps being
validated;

Matrices and Bracketing in Process Validation

2
􀂇 Product packaging, such as where only a minor adjustment in packaging
parameters is required to accommodate different bottle heights or dosage
counts.
Matrixing across different products may be applied to the packaging validation of the
final dosage form, for example to evaluate the packaging of different products in a
common packaging presentation. As with other uses of bracketing and matrixing, the risk
of using this strategy for the potential products encompassed by the matrixing plan
should be considered, documented and approved.
The use of bracketing/matrixing for the validation of a manufacturing process across
different products should be approached with caution because of the risk of overlooking
other possible affects of the change. Use of this type of bracketing/matrixing requires a
good understanding of the processes involved and the risks being assumed. For example,
in the evaluation of a change of a critical material for different products, the excipient
interactions, critical process parameters and critical quality attributes (CQAs) are not
necessarily the same for each product. The effect of the change in the CQAs may be
different for each product. A product sensitive to the change may experience a failure in a
CQA (e.g. dissolution) while in a case of a product not sensitive to
the change, it may experience no effect at all in its CQAs.
To obtain the maximum benefit with minimum risk from bracketing and matrices, it is
necessary to have a well-developed understanding of the impact of critical process
parameters on critical quality attributes. There should be a documented and justified
rationale that explains why one set of test conditions (e.g., manufacturing process,
product presentation, etc.) is representative of one or more related test conditions.
Typically, the rationale is addressed by selecting parameters and/or products that
represent the edges of a range or 􀂳worst case􀂴 of allowable conditions. The rationale and
justification for the bracketing/matrixing strategy to be used in validating a process
should be provided in the validation protocol, or in another document referenced in the
protocol.
Depending on the circumstances, prospective and concurrent validation approaches may
be used for validating a process using bracketing or matrixing. If a concurrent approach is
used, an interim report provides a summary of the results obtained for a product batch, in
order to justify the validation and release of one of the product presentations within the
bracket/matrix. This approach may also assist in approving the manufacturing and/or
release of additional batches of a particular presentation. At the completion of the
validation, the validation report will address all batches.
At present, some regulatory authorities may not accept the use of bracketing or matrixing
for validation. Japan, for example, currently requires that all combinations be validated.
The following examples include possible matrixing/bracketing approaches. There may be
other acceptable approaches.

Equivalence Criteria of Impurities for API Process Validation

Recommendations and Rationales
For validation of a process to prepare a new API, the impurity profile should be comparable
to or better than the profile determined during process development, or for batches used for
clinical or toxicological studies. For evaluation of a newly developed or modified process to
prepare an API that is already commercially distributed, the comparison provides assurance that
the process produces material that is equivalent to (or better than) acceptable material prepared in
the past by an existing process, with respect to impurities.
The need to evaluate equivalence for isolated process intermediates should be considered on
a case-by-case basis.
For some validations, insufficient reference batches are available for a meaningful comparison.
Meeting established limits is considered adequate for the equivalence comparison in these
situations. For other validations, the availability of adequate reference batch data makes the use
of statistical acceptance criteria more desirable because it enables comparison of the validation
batches to established process capability data.
A. Selection of appropriate reference batches:
1. New Products (new API or intermediate process at first manufacturing site)
Batches prepared during process development should be selected. These may include
batches prepared in Production equipment, and those prepared at laboratory and pilot
scale. They should be batches made by the same process and may include pivotal
clinical batches, and those used for toxicological and/or stability studies. In many
cases, the number of acceptable development batches may be relatively small.
2. Existing APIs
a) Major changes undergoing revalidation, or first-time validation 􀂱 Reference
batches should be selected from plant batches prepared prior to the validation to
be performed.

Equipment Cleaning Validation For Active Pharmaceutical Ingredients

2
· Stability of material(s) being cleaned under the proposed cleaning conditions;
· Suitability of the cleaning agent(s) for the materials of construction of the
equipment;
· Equipment surface finish (e.g., stainless steel, glass, polypropylene);
· Rationales for decisions on which materials to test, the limits for testing, and the method
of verification (see Table 1); and
· Evaluation of campaign length.
2.
Equipment with the Same Design and Operating Principle may be grouped for the
purpose of validation. These groupings should be documented and justified. The
documentation should be approved by Site Quality Team and Production
Team. If equipment grouping is used, cleaning validation should be performed
using three executions of the same cleaning procedure using any combination of
equipment within a group.
3.
Where Equipment is Used to Produce Only Early Intermediates (i.e., intermediates
produced prior to the introduction of the API starting materials), cleaning verification
is required. Validation of the cleaning procedures for these cases is not required (see
Table 1).
4.
Selection of The Most Difficult To Clean Product or Process requires consideration
of, at least, the following:
• Solubility of residues in cleaning agents (including cleaning and rinse solvents);
• Potential for polymers, or other side products to form during or prior to the
cleaning operations;
• The Residue Acceptability Limit (RAL) required for cleaning;
• Processing Parameters (e.g., high temperature, use of carbon); and
• Cleaning history.
Selection of the most difficult to clean product or process should be documented in
the validation protocol or a Cleaning Evaluation Report.
5.
Rinsate Method - if a rinsate method is used as the sampling method, a measured
volume of solvent used for the final rinse should thoroughly wet all product contact
surfaces, and should be circulated through all product contact lines before the rinsate
is tested in the laboratory for residues.
6.
Swabbing Method - if swabbing is used as the sampling method, swabbing of
product contact surfaces should be performed in locations from which there is a
likelihood of transfer of residue to a subsequent product and from most difficult to
clean areas (i.e., dead-legs, bottom valves, overheads, tank domes and inlets).

Process Validation for Drug Products and Medical Devices

Brief description of product, including product name, dosage form, and strength
where applicable;
· Master manufacturing instructions or Device Master Record (DMR) to be
validated;
· Brief description of process with a summary and/or process flow diagram of
critical processing steps to be evaluated and critical parameters to be monitored;
Acceptance criteria for the following:
- Acceptability (meeting established critical quality attributes and
specifications);
- The number of consecutive successful validation batches/lots needed to show
consistent control of the process.
- Equivalency to existing drug products (where applicable) by comparison to
previously produced batches/lots (commercial, development, or biobatches).
- Requirements to conduct homogeneity and hold time studies, if applicable;
􀂇 Sampling plan, including type, amount, and number of samples, together with any
special sampling or handling requirements.
􀂇 Critical process parameters and operating ranges, including justification for these
Ranges.
􀂇 Calibration of any critical equipment used specifically for the validation studies
(e.g., one-time studies on validation batches/lots using portable equipment, measuring
equipment);
􀂇 Plan for the number of batches/lots to be put on stability, if any; and
􀂇 Methods for recording and evaluating results (e.g., statistical analysis).
3. On-Line or In-Line Monitoring may be used, in lieu of discrete sampling (e.g., to
demonstrate homogeneity or acceptance criteria).
4. Homogeneity should be demonstrated throughout the batch, when required by the
validation protocol.
A sampling plan for the homogeneity study should be provided that justifies the
number of individual locations and the number of samples to be taken from the
product batch [e.g., for blend uniformity]. The bulk should be representatively
sampled based on product type (e.g., aqueous solution or solid dosage), mixing
container geometry and process (e.g., mixing mechanism) on completion of process
step. Additional sampling on completion of discrete critical steps may also be
6
􀂇 In-process assay of bulk tank and bioburden testing, where applicable;
􀂇 Environmental conditions (e.g., temperature and humidity control, air
classification, pressure differentials), where applicable;
􀂇 Removal of oxygen, where applicable;
􀂇 Dose and/or content uniformity;
􀂇 Fill weights or volume controls;
􀂇 Moisture content, where applicable; and
􀂇 Foreign matter including particulates.
If validation is being carried out as a result of a change to an existing process,
documented justification should be provided in the validation protocol if any of the
above applicable parameters are not to be assessed.
Media fill, environmental monitoring, and moist heat terminal sterilization process
studies that support the process being validated should be referenced in the validation
protocol.
14. Dry Powder Inhalers -validation should include evaluation of the mixing process
(where applicable) and filling process.
In addition to the general protocol requirements, the process validation protocol for dry
powder inhalers should include assessment of, at least, the following:
􀂇 Emitted dose uniformity; this should assess both inter and intra inhaler dose
uniformity;
􀂇 Fill weight or volume and number of deliveries from the container;
􀂇 Airflow resistance;
􀂇 Aerodynamic assessment of fine particles using a multi-stage impactor;
􀂇 Compliance with finished product specification, including any microbiological
requirements; and
􀂇 Comparison with the previously produced lots (e.g., commercial, clinical,
development, or biobatches).
Device Parameters:
􀂇 Robustness of Process Capability of component manufacturing and finished
device assembly processes demonstrated; and
􀂇 Compliance of component(s) to specification including extractables data for
components in the drug/airway path and in intimate mucosal contact. Validation
should be initiated as the result of a component change.
Drug/Device Combination Parameters:
􀂇 Respirable Fraction of delivered dose.
If validation is being carried out as a result of a change to an existing process,
documented justification should be provided in the validation protocol if any of the

Cleaning Validation – Visual Inspection and Quantitation

Consideration of the following is suggested as part of the development of a
manual cleaning methodology:
- The inspection of major equipment following or during manual
cleaning should take place prior to the analytical rinsing or swabbing
to provide a greater probability that target residues are removed prior
to sampling.
- The inspection should not substitute for the final visual inspection that
would typically take place following analytical sampling. The final
visual inspection determines the success or failure of the validation
execution.
- For those areas that will be inspected again for final determination of
visual cleanliness, this in-process inspection may be less stringent than
the final visual inspection. For instance, flashlights and mirrors might
not be necessary, complete absence of visible residues might not be
required, and complete disassembly of equipment might not be
justifiable. The justification for this approach being that this type of
inspection is not the final inspection and once a residue has been acted
upon in some manual manner (e.g., scrubbing, power washing) it is
more likely to be effectively removed subsequently by the CIP system.
- The inspection is at the discretion of the process designer(s) and is not
required. The purpose of inspection after manual cleaning is to
measure the effectiveness of the manual methodology before resuming
CIP cleaning. It might not be justified for example, if the manual
methodology has been shown to be rugged in the past, or is simply a
precautionary measure to provide a greater probability of passing
acceptance criteria at the conclusion of the cleaning process.
Inspection of equipment that is cleaned manually and can be 100% visually
inspected prior to release back to production (e.g., mills and minor equipment)
is not the subject of this specific section. Rather, the inspection of these
examples should follow the guidance of 􀂳final visual inspection􀂴 detailed
below.
2 (a). Visual Inspection of Dedicated Equipment 􀂱 Interval Cleaning:
Interval cleaning, or cleaning processes that take place within a campaign of
the same product, are appropriate when an evaluation of the material being
cleaned has been completed and there are no quality concerns (e.g.
degradation of material) about carryover of some amount of one batch into the
next batch.
Although the intent of this section of the procedure is focused primarily on
dedicated equipment it may also be applied to interval cleaning that takes
place between batches within a campaign using multi-purpose equipment.

Cleaning Validation – Rinsate and Swab Sample,Test Method Development and Validation

If a specific method is being validated, then specificity studies need to be performed for
the analyte of interest. The potential for interference from the following should be
considered:
o Swab extractables
o Cleaning agents
o Sample containers and lids
o Excipients and other compounds potential present
􀂇 If a specific method is being validated for a cleaning agent, the only specificity
experiment typically executed is specificity from swab extractables.
Range
The equipment cleaning analytical method should be validated around the calculated RAL for the
material. The method is considered valid for any RAL within the validated recovery range. If the
RAL falls outside the validated recovery range, the method should be revalidated with respect to
the affected elements (e.g. range, linearity).
Linearity
Linearity should at a minimum cover the expected analyte RAL and encompass the levels
included in repeatability and recovery studies. The lower end of the linearity study shall take into
consideration the correction factor for sampling recovery, if applicable (e.g. if the RALs have a
range of 4-6 ug/cm2 and the recovery is 50 percent, the linearity study should include levels of
2-6 ug/cm2 ).
Intermediate Precision
Intermediate precision is the study of the effects of random events (e.g. days, analysts, equipment
etc.) on the precision of the analytical procedure. A method intermediate precision experiment
should be conducted unless there is a documented rationale otherwise (e.g. a reliable and robust
swabbing verification program is implemented). Method intermediate precision should include
use of a second Lab Analyst, on a different day, using different solutions and different analytical
equipment, if possible.
System Suitability
System suitability should be conducted for systems such as HPLC and TOC. Although nonspecific
methods like UV, pH etc. may be used; the ability of the selected method to detect the
residue shall be demonstrated (for example UV absorbance at the residue maximum wavelength
and non-interference of the rinse solution).
Recovery Studies
Analyte residue recovery shall be challenged as part of the analytical method validation. The
recoveries of each material (product or cleaning agent) from the different process-contact surfaces
that constitute the major portions of equipment􀂶s surface area are typically demonstrated.
Alternatively, the recovery value of a worst case material could be substituted for all the materials
sampled with the same rinse solvent. Typical surfaces may include hastelloy, stainless steel,
glass/glass lined carbon steel and PTFE (polytetrafluoroethylene).
The solvent used in the recovery study should be the same as is used for routine sampling.

Cleaning Validation - Product and Equipment Grouping and Worst - Case Product Selection

Product Grouping
For the purpose of cleaning validation a group of related products to be identified and a single
product selected as 􀂵worst-case􀂶 or representative of the product family The rationale for the
grouping must be documented. Types and examples of product grouping include:
Figure 1: Types and examples of product grouping
Once products are appropriately grouped, the worst-case product or products can be selected
from among the group for the purpose of executing the cleaning validation protocol. A number
of scenarios are possible:
· Within a group, two or more products may be determined to provide an equivalent
􀂵worst-case􀂶 challenge to the cleaning procedure. Once the rationale for equivalency
has been documented and approved by the Quality Authority, the equivalent products
are used to demonstrate the effectiveness of the cleaning procedure during validation.
· Example: Product A and Product C are established as equivalent worst-case
challenge products for the cleaning procedure used for products A, B, C, D and E.
During validation, any lot combination of Products A and C are used to fulfill the 3 validation
cleanup requirement (e.g. 3 of A or C, 2 of A and 1 of C or1 of A and 2 of C).
􀀀 Within a group, two or more products are determined to be 􀂵worst-case􀂶
challenges, but are not equivalent. Each worst-case product should be subjected to
the 3 validation cleanup requirement.
􀀀 The same cleaning procedure is used for two or more groups of products. Each
worst-case product within each group should be subjected to the 3 validation
cleanup requirement, unless a rationale is documented and approved by the
Quality Authority that the worst-case product of one particular group is clearly a
worst-case product for all groups.
􀀀 Other scenarios may be possible and each product, or any new product introduced
to the site, should be evaluated on a case-by-case basis.
The criteria that should be considered when selecting a worst-case product or products include:
􀀀 Solubility of the residues in the cleaning agent, including cleaning and rinse
solvents - the least soluble residue among a group is the most common approach.
􀀀 Ease of removal by a detergent, if applicable.

Cleaning Validation - Calculations of Residue Limits For Drug Products for Equipment Cleaning

Cleaning Validation - Calculations of Residue Limits for Drug Product Therapeutic and
Non-therapeutic Materials for Equipment Cleaning
Introduction
This guideline provides equations and examples for calculating the Maximum Allowable Residue (MAR),
and Residue Acceptability Limits (RAL) for Drug Products and Non-Therapeutics.
Examples are provided for determining the acceptable equipment cleaning residue limits for
therapeutic drug products (MART and RALT) and for non-therapeutic ingredients (MARN and
RALN). For the therapeutic drug products both single product combination (Product A to B)
and multiple products combination examples are given.
Recommendations & Rationale
The attached Appendices give equations and example calculations for therapeutic drug
product cleaning limits for a solid oral dosage form (tablet), creams/ointment and ophthalmic
product. In Case-1 therapeutic example it is included the calculation of Maximum Allowable
Residue (MAR) limits using two formulas; dose MART and toxicity MAR. It also includes an
example for determining the worst case limit for shared equipment using multiple products.
Example for residue limits calculation of CIP® 100 detergent, a non-therapeutic ingredient is
also given.
Background Information and Equations:
The Maximum Allowable Residue for Therapeutics (MART) and Residue Acceptability Limit for
Therapeutic Dose (RALT) should be calculated based on each product that is to be processed in a specific
equipment train and determined by the formulas and equations provided in Appendix A. A common
default MART is not more than ten (10) ppm.
The calculated MART and the default MART should be compared and the lower of the two (2)
used. A Toxicity limit may also be calculated and compared to the default MART and the
NMT 10 ppm, and the lower of the three limits chosen.
The Maximum Allowable Residue for Non-Therapeutics (MARN) and Residue Acceptability Limit for
Non-Therapeutic Materials (RALN), such as materials used for equipment cleaning,
is a function of toxicity, and should be determined by use of the formulas and equations provided in
Appendix B. A default MARN is not more than one hundred (100) ppm. The calculated MARN and the
default MARN should be compared and the lower of the two (2) used. Residue

Analytical Test Method Validation - Robustness

2
The following example approaches may be considered. The acceptance criteria should be based on the
limits􀂶 range. The extent of change should not significantly affect the final result. The change should
also not affect the decisions made from the data.
Approach 1:
As directed by the test method, prepare standard and sample aliquots and analyze them. The test samples
are allowed to stand, under normal conditions of test (e.g., at room temperature), for a minimum length of
time equivalent to the maximum expected use time, (typically 24 hours to one week). Sample and/or
standard stability are demonstrated for more than 24 hours if applicable. If possible, analyte stability is
demonstrated over a time period that slightly exceeds the stability time period indicated in the test
method. During this study, solutions are analyzed against freshly prepared solutions. For acceptance a
minimum discernible trend in analyte response from initial and final analyses is observed and analyses
should agree within reproducibility found for the system precision.
Approach 2:
For standard stability for a low level impurity method, two different stock preparations of equal
concentration are prepared (a1 and b1) and diluted separately to the same solution concentration (a2 and
b2). Six (6) injections of standard check solution 􀂳a2􀂴 and three (3) injections of standard check solution
􀂳b2􀂴 are performed. From each set of injections calculate the mean peak area response for the analyte
main peak then calculate the standard check using the following equation.
Check = Mean Area STD 􀂳a2􀂴 x Concentration STD 􀂳b2􀂴(μg/ml) x 100
Mean Area Std 􀂳b2􀂴 x Concentration Std 􀂳a2􀂴(μg/ml)
Approximately 50ml of standard check solution A is decanted into a flask clearly labeled and stored in a
refrigerator (+2 to +8 degrees C). The remaining volume is stored at room temperature. A fresh standard
check solution is prepared on the day of analysis and the standard check procedure is repeated for each of
the stored standard A solutions against the freshly prepared check solution after a period of 24, 48, 72
hours and 7 days storage. For acceptance criteria, the standard check is between 95% and 105% (any
acceptance criteria applied must consider the concentration of the standard solutions under test, for
example the acceptance range may vary from a 10ppm solution (0.001%) to a 0.1% solution). Standard
stability may be performed over a longer period if necessary.
Approach 3:
For a chiral HPLC method, solution stability is assessed using an injection and analysis of the sample of
the appropriate test material at the following times after preparation.
- 0 hours (i.e., within 1 hour of preparation)
- 24 hours
- 48 hours
Use one of the samples prepared for the precision or robustness studies for the sample stability study, i.e.,
the first sample injected for the precision study may be used to obtain the t = 0 data-point. Extra time
points may be added and some of the tests may fail the expected criteria. However, this is recommended
to be explained in the protocol ahead of time. These extra time points are examined to determine at which
point the solutions are no longer stable.
Approach 4:
For TLC where the sample is required to be analyzed immediately, the standard only is analyzed and the
intensity should be the same as at t=0 and the plate should not have new spots.

Analytical Test Method Validation - Linearity, Range and Specificity

Recommended Linearity Acceptance Criteria:
The assay need not give results that are directly proportional to the concentration (amount) of
analyte in the sample for the test method to be valid. However, the desire to have a linear
relationship reflects a practical consideration, since a linear relationship should be accurately
described with fewer standards.
A validated method may be sufficiently linear to meet accuracy requirements in the concentration
range in which it is intended to be used. When inferring accuracy from a linearity study, linearity
could be considered acceptable if results, as compared to a standard, meet the accuracy criteria. A
plot of the data should visually appear to be linear. Suggested acceptance criteria (for API Raw
Material, In Process Control, and early intermediate material tests) for an acceptable linear
relationship may be a test method having a minimum correlation coefficient (r) of > 0.95.
Range:
The range is the interval between the upper and lower levels of analyte concentration for which
acceptable linearity, accuracy (recovery), and precision are obtained. It is recommended that the
range be established to include all specification limits for a method and the expected results. The
range should include at least five points to establish linearity. Values outside of the validated
range can be reported as estimates. Range should be established by summarizing the accuracy
(where appropriate), the linearity, and the precision data.
The following minimum specified ranges are taken from ICH and may be considered as minimum
start points for test methods within the scope of this document.
O For the assay, the ICH range is normally from 80% to 120% of the test
concentration. If assay and purity are performed together as one test and only a
100% standard is used, linearity should cover the range from the reporting level
of the impurities to 120% of the assay specification.
o For determination of an impurity; the range of concentrations used to evaluate the
linearity should consist of the quantitation limit and at least 120% greater than
the concentration that would be the impurity specification limit.
o For example, if the concentration at the specification limit was 0.2% w/w,
and the limit of quantitation was 0.08% w/w then the range should span
0.08% (w/w) to 0.24% w/w. For example, the concentrations for the
linearity experiment might be 0.08%, 0.12%, 0.16% 0.20% and 0.24%.
More solutions may be evaluated if the linearity range must be extended.
o In cases where specified impurities/degradation products are not available a
surrogate material such as a compound with similar structure or API may be used
to demonstrate linearity. In these cases, a rationale for the use of a surrogate
should be given.

Analytical Test Method Validation - Quantitation and Detection Limit

2
with known concentrations of analyte and by establishing the minimum level at which the analyte
can be quantified with acceptable accuracy and precision. The quantitation limit must not be
greater than 50% of the specification, where technically feasible.
· Based on Signal-to-Noise Approach
This approach can only be applied to analytical procedures that exhibit baseline noise.
Determination of the signal-to-noise ratio is performed by comparing measured signals from
samples with known low concentrations of analyte with those of blank samples, and by
establishing the minimum concentration at which the analyte can be reliably quantified. A typical
signal-to-noise ratio is 10:1.
· Based on Capability of the Instrument
In some cases the instrument itself is the limiting factor for the analysis regardless of the sample.
An example of this is an LOD test using an analytical balance. In this case a discussion of the
quantitation limit may be constructed in the validation documentation based on the calibration
tolerance of the equipment rather than analysis of actual samples. The actual limit of quantitation
would still be presented in numerical terms relevant to the assay method based on the discussion.
Another example of this may be for KF titration assays where the ability of the instrument to
deliver a minimum amount of titrant would be the limiting factor. It is recommended that
experiments to determine this minimum amount of sample should be conducted for the specific
instrument model if this approach is taken. The experiment(s) could then be referred to in any
validation that utilizes the same model of equipment.
· Based on the Standard Deviation of the Response and the Slope
The quantitation limit (QL) may be expressed as: QL = 10 􀄱/ S where, 􀄱= the deviation of the
response; S = the slope of the calibration curve. The slope S may be estimated from the
calibration curve of the analyte. The estimate of 􀄱 is carried out in a variety of ways including:
o Based on the Standard Deviation of the Blank:
Analyzing an appropriate number of blank samples and calculating the standard
deviation of these responses and perform measurement of the magnitude of
analytical background response.
o Based on the Calibration Curve:
A specific calibration curve should be studied using samples containing an
analyte in the range of the QL. The residual standard deviation of a regression
line or the standard deviation of y-intercepts of regression lines may be used as
the standard deviation.
In all cases, the quantitation limit can be subsequently validated by the analysis
of a suitable number of samples known to be near or prepared at the quantitation
limit or reporting level.
Two possible approaches include:
A)
Three replicate preparations of a spiked sample are prepared at the quantitation level or
reporting level and analyzed. Calculate the % recovery. Calculate the average of the
replicates and % RSD.

Analytical Test Method Validation - Precision and Accuracy

demonstrated by performing 3 replicates each of three separate sample concentrations (9
determinations) covering the specified range of these procedures.
- Recommended Repeatability Data:
Calculate the result for each replicate. The % RSD for each level should meet the recommended
criteria.
- uggested Repeatability Criteria:
Several factors should be considered when selecting criteria: The intended purpose of the test and
the expected specification range are important parameters. It is recommended that acceptance
criteria be established as recommended in Table 1 and Table 2.
Intermediate Precision:
Intermediate precision expresses 􀂳within laboratories􀂴 variations (e.g., different days, different
analysts and different equipment).
The extent to which intermediate precision may be established depends on the circumstances
under which the procedure is intended to be used. It is suggested that sites establish the effects of
critical random events on the precision of the analytical test procedure.
Typical variations to be studied include days, analysts, equipment, etc. ICH does not consider it
necessary to study these effects individually and this is endorsed by this guideline.
The use of an experimental design (matrix) is considered useful. Certain markets (i.e. Japan)
have more specific requirements for intermediate precision. To meet intermediate precision
requirements for Japan for assay and quantitative impurity procedures, it is recommended that
the analyses be carried out as prescribed by the method over a minimum of six occasions with at
least three analyses per occasion.
An example of such a matrix for Japanese markets is provided in the following table:
Occurrence
#1 Occur #2 Occur#3 Occur #4 Occur.#5 Occur
#6
Day 1 1 2 2 3 3
Analyst 1 2 2 1 1 2
Instrument 1 2 1 2 1 2
Column 1 2 1 2 2 1
Analyses can be carried out using either samples spiked at a suitable level(s) and/or representative
lots containing a representative amount of impurities. If the representative lots do not contain
specified impurities/degradation products, spike studies should be performed. In cases where
specified impurities/degradation products are not available a surrogate material such as a
compound with similar structure or API may be used to demonstrate precision. In these cases, a
rationale for the use of a surrogate should be given.
Recommended Intermediate Precision Data:
Intermediate Precision: Calculate overall % RSD of the multiple occasions. The overall SD or
RSD of the multiple occasions should meet the recommended criteria.

Analytical Test Method Validation - System Suitability

o Peak symmetry, as measured by the tailing factor, may be of importance to report, especially in
impurity testing. If tailing is too extensive, it may mask other impurities. When the tailing factor
increases, integration becomes less reliable and precision can be compromised. Peak fronting may
also cause integration problems and may become a factor as columns age.
o It is considered that repeatability is normally used as an essential criterion for system suitability
testing but this may not be possible for all types of IPC test methods. For example, Area %
methods do not require repeatability. System repeatability is regarded as the contribution of the
instrumental variability to the precision.
o Demonstration of specificity may be required for certain applications and may involve resolution
between two significant peaks, peak efficiency by theoretical plates or peak symmetry by tailing
factor. It is recommended that the specificity be demonstrated as part of the SST criteria where
variability of sample make up is possible (e.g. for a chromatographic method or TLC method, the
sample diluent is prepared on day of analysis or may be of a different batch/lot of solvent to that
used during validation). It may be of benefit to demonstrate adequate specificity between the
diluent (blank solvent) and the sample solution. This is particularly useful when investigating
low-level impurities as the detection and attribution of a novel impurity to the sample may be
discounted by its presence in the sample diluent.
o For non-chromatographic testing, the use of control samples or day/ time of use calibration may
sometimes be appropriate for some technologies. Examples include but are not limited to KF,
LOD (day/time of use calibration), and particle size (control samples).
Other pharmacopoeias should be consulted if required, however, the US Pharmacopoeia recommends that
to determine system suitability % RSD, 5 replicate injections if the % RSD is 2.0 or less and if the %
RSD is greater than 2.0, six replicate injections are recommended. While the USP recommends the above
% RSDs, these criteria may not be adequately low to assure method performance (e.g. when the %RSD of
the assay is 1% or when the specification is tight such as 99.0 􀂱 101.0%). Therefore, it may be relevant to
consider using the EP Pharmacopoeia recommendation for a tighter % RSD of 1/3 of the specification
range to have a 95% confidence that the result is within the limit. The EP recommends that system
suitability for repeatability is based on the limit range and number of standards used in the test, where n
can vary from three to six.
Recommended SST Criteria:
The acceptance criteria used should assure adequate precision and specificity for the intended use. One
approach is to set chromatographic SST criteria based on data collected during a validation exercise.
Equivalency may be demonstrated as follows,
· If resolution is no less than 0.8 times of the average typical value; tailing is no greater than 1.3 of
typical value; and efficiency is no less than 0.8 times of the average typical value. It is
recommended that a sensitivity standard at the 0.05% level for API impurity and degradation
methods be utilized.
Appendix 1: In-Process Control Test Types 􀂱 System Suitability Testing Examples

Analytical Test Method Validation - Risk Assessment and Prioritization

Risk Assessment and Prioritization
To ascertain which raw material, in-process control (IPC) and intermediate test methods require
validation, it is suggested that a documented Risk Assessment be carried out on test methods
currently utilized at an API site. This risk assessment approach can be used to evaluate legacy or
new test methods.
This risk assessment evaluation may be based upon the following suggested principles:

• Regulatory expectations at the time of product filing or re-registration
• Methods may generally be grouped from low risk to high risk. This risk grouping
helps to prioritize those areas that need validation based on a science based
evaluation of impact to final API product quality. The following table (Table 1)
gives an example grouping strategy for the analytical methods within the scope
of this procedure:
Table 1: M ethods, types and their recommended risk levels

Evaluation Process:
Conduct a cross functional and documented analytical test method evaluation based upon an
understanding of the test data utilization by the Site Quality Authority and other site functions.
This cross functional review might be conducted by colleagues drawn from the Site Quality
Authority, Site Production Operations, development support and other site based or center
technical functions as necessary.
Methods may be grouped for this evaluation, such as in the above table to set prioritization for the
team.
During this evaluation phase, a number of contributing factors tend to determine impact to
quality. Downstream effects may also need to be considered. For example, once the material is
isolated, tested, and discharged from the equipment, one may have to reprocess/ rework material
because of potentially inaccurate data from the earlier IPC test method.
The use of what-if scenarios can assist in the risk analysis. For example, consider the following:
o Stage in the API Process: Where does this test method􀂶s result lie within the
overall quality 􀂵control􀂶 or 􀂵assurance􀂶 strategy in producing a quality API?
o Critical Quality Attribute: Can analytical data gleaned from this early stage of
the step highlight where an impurity or its precursor is forming?
o Critical Quality Parameter: Could the process step be adjusted within allowable
parameters to marginalize or purge unwanted impurities before isolation?

Analytical Test Method Validation - General Guidence

pre-determined in order to properly select the validation experiment parameters before the test
method is developed and the validation exercise is begun.
To begin the validation exercise, individual sites can determine from Appendix 1 and 2 what
analytical elements should be evaluated for the particular test and sample of interest.
In the case that a limited set of elements are included in the validation, it is recommended that the
Method Validation Summary or protocol should address the reasons for limiting the validation to
the chosen elements.
Validation of the test procedures should take into account the reactivity of the sample. If the
sample can change over time, the validating scientist should attempt to find a means to quench
the reaction before analysis. While this is normally a method development activity, this
consideration should be evaluated by the site if method validation should occur at the sites. Once
execution of the method validation has begun, deviations and experimental failures should be
documented in the method validation report.
In some instances a set of experiments may be able to satisfy several validation parameters i.e.
robustness, intermediate precision and repeatability could be combined and studied together
instead of as isolated effects.
· For example, compendial physical tests such as LOD/ROI/pH where the analyst a
sample amount are primary contributors to variation and the method is well
defined in general chapters are ideal for this situation.
· An experiment could be constructed such that variations in sample size, analyst
and equipment could all be studied at once.
Documentation
It is recommended that sites have local site procedures for performing test method validation. For
methods in the scope of this guideline, sites may choose to prepare a protocol and obtain approval
prior to commencing validation. Alternatively an SOP containing pre-approved templates or
acceptance criteria guidelines could be used Because these test methods are generally lower risk
than those used to test final APIs, more flexibility in documentation of validation information
(e.g. use of an SOP versus a protocol) is considered acceptable.
Suggested criteria are included in this guideline on this topic; however criteria should be
directly linked to the method􀂶s intended use.
Method Validation Summary Report:
It is recommended to analyse the experimental results and prepare a Method Validation Summary
of the findings.
These method validation summaries may include but are not limited to:
􀂇 The performance results against criteria listed within this guideline, site SOP, or separate
pre- approved protocol.
􀂇 For higher risk methods, at least two reviewer signatures are recommended to be obtained
for the Method Validation Summary to be approved.

Gel Clot Validation Method

Standard Operating Procedure
Title: Gel Clot Validation Method
______________________________________________________________________________________

Document Owner
Micro Laboratory Manager
Affected Parties
All Microbiology Laboratory colleagues
Purpose
To describe the method of Gel-
Clot validation to be used in the Micro . Lab.
Scope
The procedures outlined in this SOP are to be followed by the Micro . Lab. staff.
Definition
BET
Bacterial Endotoxin Test : A test used to detect or quantify endo toxins
Endotoxin Toxic molecules originating from the outer cell wall of gram -negative bacteria
Endotoxin
Limit
The maximum amount of endotoxin allowed in a sterile product or on a medical device .
Maximum
Valid Dilution
A figure that shows how much a pa renteral product or raw material may be diluted without
losing the ability to detect endotoxin at the limit concentration
Related Documents
MICLAB 070 Identification of Micro organisms to Genus and Species Level
MICLAB 080
Bacterial Endo Toxin Testing (
LAL) - Gel Clot Method
Form 590 Verification Assay Results Sheet
Form 595 Bacterial Endotoxin Test Data
Form 600 Maximum Valid Dilution and Endotoxin Limit Calculations
Form 605 Bacterial Endotoxin Gel Clot Validation - Final inhibition and Enhancement Test
Form 610 Bacterial Endotoxin Gel Clot Validation - Preliminary inhibition and Enhancement test
EHS Statement
The reagents used in testing must be disposed of into the Biohazard Bin along with all the disposable
equipment. Safety glasses must be w
orn if using IPA/solvent.
Table of Contents
1. General ................................ ................................ ................................ ................................ ............... 2
Department Micro Laboratory Document no
MICLAB 105
Prepared by:
Date:
Supersedes:
Checked by:
Date:
Date Issued:
Approved by:
Date:
Review Date:
Standard Operating Procedure
Title: Gel Clot Validation Method

Preparation of the Endotoxin Working Standard , see
MICLAB 080.
2.2.
Performing the Preliminary Inhibition / Enhancement Test:
a)
Adjust the pH of the product (if necessary) to within the range of pH 6.0-
7.5 with 0.1N pyrogen
free HCL or 0.1N pyrogen
free NaOH.
Note: Do not adjust the pH of the unbuffered saline or water
. Add Pyrosperse so that the
concentration of Pyrosperse in the product is 2% i.e. 0.1ml Pyrosperse to 5ml of product.
b) Product Control Dilution :
ïò Prepare a dilution series of the pr
oduct in 2 fold increments to a dilution level of 1 in 32.
Further serial dilution of the product may be necessary , i.e.
1ml product + 1ml WFI = 2 fold dilution.
îò
Prepare two (2) sets of six 10 x 75 mm test tubes (appropriately labelled) of the above
produ
ct dilution series , i.e. from zero dilution to 1 in 32 dilutions, by pipetting 0.1ml of the
appropriate dilution into 2 test tubes.
c) Product Compatibility :
Prepare a dilution series of the product in 2 fold increments with endotoxin spiked WFI +2%
Pyrosp
erse to a product dilution level of 1 in 32 such that the concentration of endotoxin
throughout the dilution series remains the same . Further serial dilution of the product may be
necessary. The level of endotoxin spike should be equal to twice the sensi
tivity of Lysate used,
e.g.
If the Lysate sensitivity is 0.06EU/ml the level of endotoxin spiked per dilution tube should be
0.125EU/ml. Hence the endotoxin spike WFI must contain 0.25 EU/ml and 0.125 EU/ml
The product compatibility dilution series for a
Lysate sensitivity of 0.06 EU/ml will therefore be
as follows:
Tube Dilution
No Factor Contents of tube
Endotoxin Conc.
0
8ml product
+ 0.05ml endotoxin *
(0.20EU/ml)
1 - 2
1ml product
+ 1ml endotoxin WFI 0.25EU/ml
0.125EU/ml
2 4
1ml product
+ 1ml e
ndotoxin WFI 0.125EU/ml
0.125EU/ml
3 8
1ml product
+ 1ml endotoxin WFI 0.125EU/ml
0.125EU/ml
4 - 16
1ml product
+ 1ml endotoxin WFI 0.125EU/ml
0.125EU/ml
5 32
1ml product
+ 1ml endotoxin WFI 0.125EU/ml
0.125EU/ml
* Endotoxin diluted in WFI
1. Prepa
re two (2) sets of six 10 x 75 mm test tubes (appropriately labelled) of the above
product/endotoxin dilution series , i.e. from a zero to a 1 in 32 dilution, by pipetting 100³l of
the appropriate dilution into 2 test tubes.
i Negative controls
Prepare 2 nega
tive controls by pipetting 0.1ml of pyrogen free WFI into two
10 x 75mm test tubes.
i Positive controls
Prepare two sets of four 10 x 75mm test tubes (appropriately labelled) of the
endotoxin standards dilution series to bracket the concentration of the Lysa te used in
the test, e.g. if the Lysate sensitivity is 0.125EU/ml the endotoxin standards to be
employed are tubes No 4, 5, 6, 7, i.e. concentration of 0.25EU/ml, 0.125EU/ml,
0.06EU/ml and 0.03EU/ml.
i
Pipette 0.1ml of the reconstituted Pyrogent into each of
the above test tubes, i.e.
i
12 tubes Product control dilution
i
12 tubes Product compatibility
Standard Operating Procedure
Title: Gel Clot Validation Method

K = Maximum allowable endotoxin exposure
5EU/Kg/Hour for intramuscular
0.2 EU/Kg/Hour for intrathecal
D = Maximum human dose
Potency
= drug concentration (This is not required if the dose is expressed in mLs )
An average human weight for the purpose of MVD calculation is regarded as 70kg (or 60Kg
for Japan).
Examples of Calculation for Endotoxin Limits :
Endotoxin Limits
Example 1 Pro
duct 1
Example 2
Product 2
Dose = 2 mg/Kg
Dose = 10mL/Kg
Potency = 100mg/mL
Potency = not applicable
Endotoxin Limit =
5EU/Kg x 100mg/mL
2 mg/Kg
= 250EU/mL
Endotoxin Limit =
5EU/Kg
10mL/Kg
= 0.5EU/mL
Note: The product dilution level required to overcome inhibition as established by the
inhibition /enhancement must be compared with the MVD calculated to ensure that the
Maximum Valid Dilution f
or that product has not been exceeded.
3.2.
Final Inhibition /Enhancement Test
The Inhibition/Enhancement test is then to be repeated using the diluted product
concentration in 4(b) containing varying concentrations of endotoxin that bracket the lysate
sensitivity and comparing this product series with a series of the same endotoxin
concentration in water alone .
Method:
Preparation of Pyrogent, see
MICLAB 080.
Preparation of the Endotoxin Standard ,
Preparation of the Endotoxin Working Standard ,
ìò л®º±®³·²¹ ¬¸» Ú·²¿´ ײ¸·¾·¬·±²ñÛ²¸¿²½»³»²¬ Ì»­¬æ
a)
Adjust the pH of the product (if necessary) to within the range of pH 6.0
7.5 with 0.1N
pyrogenfree
HCL or 0.1N pyrogenfree
NaOH.
Note: Do not adjust the pH of the unbuffered saline or water
. Add Pyrosperse so that
the concentration of Pyrosperse in the product is 2%, i.e. 0.1ml Pyrosperse to 5ml of
product.
Product endotoxin dilutions:
i)
Prepare 20ml of Product diluted with pyrogen -
free WFI .
ii)
Pipette 10ml of this diluted product into a pyrogen -free test tube and spike with
endotoxin to give the most concentrated endotoxin level required for an endotoxin
series to bracket the Lysate sensitivity , e.g. if the Lysate sensitivity is 0.125 EU/ml the
endotoxin series will be 0.25 EU/ml, 0.125 EU/ml, 0.06 EU/ml and 0.03 EU/ml.
Therefore to spike the diluted product with a endotoxin concentration of 0.25 EU/ml use:
10ml diluted product +0.1ml (25 EU/ml) endotoxin
= Tube No.1.
= Endotoxin conc. 0.25EU/ml
iii)
Prepare a 2 fold dilution series of the endotoxin spiked diluted product using the
additional 10ml of diluted product prepared in (i) as the diluent such that the product
concentration remains constant throughout the dilution series whereas the endotoxin
Form 590
Issue date:
Verification Assay Result Sheet
Verification Assay For Microbi ology Laboratory Technicians
(Ref. MICLAB 105)
File Location:
Date Printed: Page 7 of 15
Verification Assay Date:
Na
me of Technician:
Test Reagents
Reagents
Lot No. Reconstitution Date Expiry date
Pyrogent
EU/mL sensitivity
Endotoxin
EU/mL potency
Pyrosperse NA
2% working concentration
Test kit NA
L.A.L, Endotoxin & Endotoxin Working Standards dilu
ent.
Any sterile batch (WFI) (Tested to be L.A.L. negative) Batch No.: Expiry:
Test Session Standards - Results
Key: (+) firm gel, (-
) no gel or viscous gel.
Endotoxin Concentration & Gelation Res
Replicate ults (EU/mL) Endpoint
Assay
Number 1
0.5
0.25
0.125
0.06
0.03
0.015
EU/mL Log
Negative Controls
Key: (+) firm gel, (-
) no gel or viscous gel.
Replicate
Assay No.
Control
Results
1
2
Form 590
Issue date:
Verification Assay Result Sheet
Verification Assay For Microbi ology Laboratory Technicians
(Ref. MICLAB 105)
File Location:
Date Printed: Page 9 of 15
Calculations:
GMx = 10
Where =
ø ÷
=
GMx =10
=
=
Standard:
GM sensitivity value should fall within the range 0.03
1.25 EU/mL.
GMx = _____________ EU/mL
Has the standard been met ? YES/NO
Signature of Technician :
Approved by:
Date:
Date:
Comments about Session :
Where =
ø ÷
n
y i
n
1
;
Form 600
Issue Date:
Maximum Valid Dilution & Endotoxin Limit Calculations
(Ref. MICLAB 105 & MICLAB 085)
File Location:
Date Printed: Page 11 of 15
Maximum Valid Dilution (MVD) and Endotoxin Limits
for
Sterile finished Products Tested by LAL Gel Clot and KCA Test methods
Product:
D = Maximum human dose/Kg
mg/Kg
ml/Kg
Potency of Product
This is not required if the dose
is expressed in ml /kg
mg/ml
´
= Sensitivity of Lysate
EU/ml
K = 5.0 EU/Kg for parenterals except intrathecal drugs where k = 0.2
Endotoxin Li mit
EU/ml
MVD = Endotoxin Limit
´
Therefore: MVD for Product = _______________
Calculation for Endotoxin Limit if required
Endotoxin Limit = K x Potency
D
Therefore: Endotoxin Limit for Product = _______________
Form 605
Issue date:
Bacterial Endotoxin Gel Clot Validation
Final Inhibition and Enhancement Test
(Ref. MICLAB 105)
File
Location:
Date Printed: Page 13 of 15
Result sheet
Bacterial Endotoxin test (U.S.P.) Pyrogent
Interpretation of Results :
Result Acceptance Levels
A2
Product Endotoxin Dilution series
endpoint, Part 1 (Highest dilution
positive)
=______________EU/mL
(0.5
2 x Lysate sensitivity)
B2
Positive Controls, Part 2 (Highest
dilution positive)
=______________EU/mL
(0.5
2 x Lysate sensitivity)
C2 Negative contro
ls, Part 3
Pos / Neg
Must be negative.
D2
Lowest dilution giving positive Lysate in
Preliminary test. (Product Compatibility
Test, B1 result)
Less than MVD.
E2
Comparison of Endotoxin
determinations in Product A2and Water
B2. (ie Dilution level they di
ffer by)
Must not differ by more than
plus or minus a 2 fold dilution.
Have all the acceptance levels been met ?
YES / NO

Validation of Aseptic Gowning Procedures

Standard Operating Procedure
Title: Validation of Aseptic Gowning Procedures

Document Owner
Micro Laboratory Manager
Affected Parties
All Validation and Microbiology Labora tory colleagues
Purpose
Aseptic gowning is the ability to complete the gowning procedure without compromising the sterility of the
garment. This SOP outlines the sterile gowning validation procedure as required for the final sign off for the
initial steri
le training and the revalidation of currently trained Operators , Fitters, Electricians and Cleaners
and all organization staff who are authorised to enter Sterile areas .
Responsibilities
i Each Functional Area Manager is responsible for ensuring that all st aff who are required to enter Sterile
areas have successfully demonstrated their gowning competency through passing their gowning
validation.
i The Microbiology Laboratory staffs are responsible for carrying out the review of the gowning
competency and asses
sing the results.
i
The Training department is responsible for recording and storing (filing) the results in the individuals
training files and keeping the individuals /teams updated with this information .
i The Training Department is responsible for the scheduling and booking of all sterile training and
retraining sessions.
Definition
cfu Colony Forming Unit representing one micro -organism
Aseptic Free from contamination
Contact plates Sterile dish of microbiological media used to monitor surfaces such as st erile gowns
Related Documents
MICLAB 005 Entry Procedure of Sterile Filling Area
Form 655 Validation Record For Sterile Gowning Procedure
EHS Statement
i Safety glasses and gloves
must be worn when using IPA .
i
Personnel who are ill , e.g. with a cold, flu
symptoms, stomach disorder, open lesions, any type of skin
disorder such as sunburn must check with their Process Manager / Microlab Department before they
can enter the sterile areas.
Department Micro Laboratory Document no
MICLAB 010
Prepared by:
Date:
Supersedes:
Checked by:
Date:
Date Issued:
Approved by:
Date:
Review Date:
Standard Operating Procedure
Title: Validation of Aseptic Gowning Procedures
______________________________________________________________________________________
Ú·´» Ô±½¿¬·±²æ Ü¿¬» Ю·²¬»¼æ п¹» í ±º ê
3. Areas of the gown to be monitored at validation
i
Gloves (without the addition of Hexifoam ).
i
Gown sleeves (at cuff)
i
Gown chest (
at zipper top)
i
Hood (near forehead)
i
Hood (side or back)
i
Gown (at top of overshoes at knee)
i
Overshoes (at top of foot)
3.1.
At the time of the Validation session , the Microbiology Laboratory observer will label all
plates with the candidate
s name, area monitore
d and the date.
3.2. The plates will then be brought to the Microbiology Laboratory together with
Form 655 for
incubation at 32p
C for 48 hours. The plates will be then transferred into the 25
C for 72
hours. After the incubation the results will be read and rec
orded in the registrar book. The
form is to be signed by the Microbiology Laboratory observer and photocopied and the copy
sent to Training Dept . for filing in the Operator
s Training File. The original document will be
kept in the Microbiology Laboratory
Validated Sterile Operators folder .
The individual should not enter the sterile area until a gowning validation has been
successfully completed.
3.3.
Individuals will be notified with their results and copied to their Process Manager . Successful
trainee shoul
d be grant access to the sterile areas.
4. Method of Re-Validation
4.1. Annual re-validation is required for all validated personnel and will consist of the sterile re -
training sessions followed by the aseptic gowning procedure validation . For personnel being
retrained a Re-validation needs to be completed within
1 month.
4.2. The revalidation
procedure is the same as the procedure outlined in Section 3
5. Acceptance Criteria
5.1.
The acceptance criteria are less than or equal to 1 cfu per plate for finger dabs and no more
than a total of 3 cfus / set of 6 contact plates or per single contact plate .
5.2.
If the acceptance criteria are not met then a second validation is permitted . In this event
notification will be sent to the Process Manager and the individual concerned .
6. Actions in the Event of a Failure
6.1. In the event of a failure the Microbiology Laboratory will inform the relevant individual
concerned and their Process Manager to ensure follow up steps are appropriate . This may
include counselling or re -training if deemed neces
sary.
6.2.
The objective of the counselling session is to :
6.2.1.
Highlight the results to the operator .
6.2.2.
Discuss potential causes; provide advice to help prevent recurrence.
6.2.3.
Outline the remedial action , which will be taken .
7.
Remedial Actions Initial Validation (After F
ull Sterile Training)
7.1. In the event of a failure of the initial validation a re -
validation is permitted .
Form 655
Issue date:
Validation Record For Sterile Gowning Procedure
(Ref. MICLAB 010)
Ú·´» Ô±½¿¬·±²æ Ü¿¬» Ю·²¬»¼æ п¹» ë ±º ê
Participant
Date
Position
Validation
Attempt
Team/Work Area
Reason for Validation Please
Tick
Reports to New Sterile Operator
Annual Re-validation
Three Months
Comments
Other
(Please Specify)
GOWNING PROCEDURE CHECKLIST
CONFORMS (Y/N) / COMMENTS
1. Dirty
side procedures, hands and nails free
of excess dirt, fresh band aids on cuts.
2.
Undergarments and hairnets, correct use.
3. Putting on Sterile socks while crossing barrier
between dirty and clean
sides.
4. Putting on slip -
on safety shoes.
5.
1st wash -
hands & forearms including area
between fingers. Contact time of Biocide
solution
at least 60 sec.
6. Nailbrush scrub with Biocide solution
-
Top surface of nails, under fingernails rinsing
procedure.
7.
Drying with sterile wipe (picked up from the
top of the pile )
forearms, hands.
8. Putting on Hood
technique, not touching
external surface, hair net coverage.
9. Putting on Face mask
technique, position.
10.
2nd wash - h
ands & forearms including area
between fingers. Contact time of Biocide
solution
at least 60 sec.
11.
Drying with sterile wipe (picked up from the
top of the pile ) -
forearms, hands
12. Putting on Overalls uniform not touching
floor, not touching external su
rface, covering
hood flap.
13. Putting on Overshoes not touching external
surface, foot straps and clips.
14. Putting on Gloves
technique, not touching
external surfaces with bare hands.
15. Pulling up Overshoes
leg straps and clips.
Validated Microbiolo gy Print Sign
Technician

Cleaning Validation – Comparative Analysis

1. Test Description
This test is to be conducted to document the validation of a TOC analysis method for use in measuring
samples for cleaning validation. A parallel analysis of samples will be carried out and compared using TOC
and HPLC analysis.
2. Test Objective
To determine the linearity and precision of a series of standards measured by TOC and HPLC over a
known concentration range.
• To determine the accuracy and recovery of a series of standards measured by TOC and HPLC
over a known concentration range.
• To determine the limit of quantitation and limit of detection of a series of standards measured by
TOC and HPLC over a known concentration range.
3. Acceptance Criteria
Test Objective Measured Response Acceptance Criteria
3.1. Linearity The correlation coefficient
(R2) for the linear
concentration range.
3.1.1. HPLC: equal to or greater than 0.997
3.1.2. TOC: equal to or greater than 0.980
3.2. Accuracy % recovery of the theoretical
amount at all concentrations
tested.
3.2.1. HPLC: 90-110%
3.2.2. TOC: 85-115%
3.3. Limit of
Quantitation
% RSD and % recovery 3.3.1. HPLC: lowest concentration of the “active”
residue at which: The area %RSD of six replicate
injections is less than or equal to 10 and recovery
between 90-110% of the theoretical amount.
3.3.2. TOC: lowest concentration at which: the
area % RSD from four replicate injections is less
than or equal to 15.0% and recovery of the
“active” residue must be between 85-115% of the
theoretical amount.
3.4. Limit of
Detection
Lowest detectable standard
concentration.
3.4.1. HPLC: The lowest concentration of active
residue that can be consistently detected for four
consecutive replicate injections.
3.4.2. TOC:
• The lowest concentration of “active” residue
that can be readily detected by the instrument
in four consecutive injections.
• The determined LOD must be equal to or
greater than that carbon concentration obtained
by multiplying the standard deviation of six
injections of Type I water (18 megohms-cm

Cleaning Validation – Swab Test

SWABBING RECOVERY STUDIES
1. Test Description
This test is to be conducted to document the validation of a TOC analysis method for use
in measuring samples for cleaning validation. A parallel analysis of swab samples will be
carried out and compared using TOC and HPLC analysis. Standard solutions will be
applied to stainless steel plates, dried and the residue removed by swabbing using the
Texwipe TX761 swab. Assessment of Linearity, Accuracy (recovery), LOQ, LOD and
precision of the swabbing method will be determined.
2. Test Objective
1. Determine the linearity and precision of a series of standards swabbed from a
stainless steel plate and measured by TOC and HPLC over a known concentration
range.
2. Determine the accuracy (recovery) of a series of standards swabbed from a stainless
steel plate and measured by TOC and HPLC over a known concentration range.
3. Determine the Limit of quantitation and limit of detection of a series of standards
measured by TOC and HPLC over a known concentration range.
4. Determine correlation between HPLC and TOC analysis.
5. Determine the visually clean limit.
6. Determine the final HPLC and TOC swabbing limits based on recovery studies.

Cleaning Validation – Rinsing Test

Test Description
This test is to be conducted to document the validation of Total Organic Carbon (TOC)
analysis method for use in measuring samples for cleaning validation. A parallel analysis
of rinse samples will be carried out and compared using TOC and High Performance
Liquid Chromatography (HPLC) analysis. Standard solutions will be applied to stainless
steel plates, dried and the residue removed by rinsing using an appropriate solvent
(usually water). Assessment of Linearity, Accuracy (recovery), LOQ, LOD and precision
of the rinsing method will be determined.
2. Test Objective
1. Determine the linearity and precision of a series of standards rinsed from a stainless
steel plate or container and measured by TOC and HPLC over a known concentration
range.
2. Determine the accuracy (recovery) of a series of standards rinsed from a stainless
steel plate or container and measured by TOC and HPLC over a known concentration
range.
3. Determine the Limit of quantitation and limit of detection of a series of standards
measured by TOC and HPLC over a known concentration range.
4. Determine correlation between HPLC and TOC analysis.
5. Determine the final HPLC and TOC rinsing limits based on recovery studies.

Auditing a Validation System

Auditing a Validation System
3
on the approved process method and specifications. This phase qualifies process and
procedures and demonstrates that the equipment and ancillary systems do what they
claim to do.
Piping and installation drawings (P&ID): Mechanical drawing or blueprints of the
required piping system for installation of equipment.
Quality risk management: A systematic process for the assessment, control,
communication and review of risks to the quality of the drug (medicinal) product across
the product lifecycle.
Worst Case: Conditions within normal parameters most likely to give failure. For
processing purposes, worst case means those values of normal operating parameters
most likely to cause process failure. For sampling locations, worst case means those
equipment locations most likely to have higher levels of residues after cleaning. For
sampling recovery, worst case means those procedures, within normal sampling
parameters, most likely to give poorer percentage recovery.
Explanation of Topic
Introduction
Validation is the action of proving, in accordance with the principles of GMP, that any
procedure, process, equipment, material activity or system consistently leads to the
expected results. Documented evidence provides a high degree of assurance that a
specific system, equipment or process will consistently produce a product meeting its
pre-determined specifications and quality attributes. To put it simply, validation is
nothing more than proving that a process actually works.
What should be valida
ted?
We use the terms validation and qualification to cover the documented verification of a
spectrum of GMP activities including
· Facilities
· Equipment used in manufacturing
· Equipment used to control the environment(s) where product is manufactured or
stored
· Utilities with product contact (e.g., water systems, compressed gases, air)
· HVAC systems
· Alarm systems that monitor utilities and air handling for process and storage areas
· Analytical methods
· Analytical instruments
· Computerized systems (e.g., computerized training system, documentation control
system)
· Cleaning processes
· Manufacturing processes
Auditing a Validation System
5
likelihood of detection of the failure. Appropriate systems are implemented to ensure that
the theoretical hazards do not present a hazard to the process. The appropriate limits are
set for these systems.
If the risk assessment has been properly carried out and documented, parts of a process
that are low risk may be eliminated or paid minimal attention during validation.
Benefits of validation
Validation assures that the entire manufacturing process and support systems work
properly before actual production begins.
Validation ensures that safe, quality products are consistently manufactured. The
validation process gives the manufacturer a further understanding of the process, possibly
improved operational efficiency, more robust processes, reduced risk of failure and
improved compliance.
General Validation guidelines
The first step to the overall validation process is to define the key requirements of the
product/process, e.g. by documenting the appropriate parameters in a User Requirement
Specification (URS) and a Functional Specification describing what is needed and how
the final result is to be achieved. A design qualification (DQ) is performed to collect
documented verification that the proposed design of facilities is suitable for the intended
purpose.
The total validation process involves DQ, IQ, OQ, PQ, process
validation,
maintenance, change control and revalidation.
Rationales used as basis for validation strategies should be documented.
Validation Stages
Establishing Qualification Process Monitoring and
key (IQ, OQ, Validation Change Control Revalidation
requirements PQ)
Since validation consists of different stages, critical sections of each stage must be
completed before moving to the next stage. Some of these critical sections include
complete testing, investigation of critical deviations and/or exceptions, and repai r (e.g.
wiring connected incorrectly). Deviations must be closed and interim approval of the
stage should be obtained prior to beginning the next stage. The protocol for the next
Auditing a Validation System
7
criteria to be tested against, etc. This directing document is the Validation Plan (VP).
The VP is a strategic document which identifies the elements to be validated, the
approach to be taken for validation of each element, the organizational responsibilities
and the documentation to be produced to ensure full consideration is given to product
quality aspects. It shows how the separate validation activities are organized and
interlinked. Overall it provides the details and relative time scales for the validation work
to be performed.
Specific elements of the VP are qualification/validation methodology, qualification
activities, personnel responsibilities, schedule, preventive maintenance, change control,
relevant SOPs and documentation requirements.
The VP is required to be prepared and approved at an early stage of the project. This plan
may or may not include design installation and operational performance qualification
protocols as individual documents. Not all projects need to be included in a VP. This
document is required when the coordination of many validation activities is necessary.
Examples are:
Ø Construction of a new manufacturing facility
Ø Purchase and installation of a new utility, (e.g. a new purified water system)
Ø Installation of a new packaging line
Ø Significant refurbishment to existing facilities, utilities and equipment
Equipment Qualification
To ensure that a manufacturing or testing process will work properly, the equipment used
in the process must perform reliably and within specifications. To achieve this, the
equipment must be qualified.
Design, installation, operation and performance qualification (DQ/IQ/OQ/PQ) are phases
of validation that support the startup of new, modified, or retrofitted equipment.
DQ/IQ/OQ/PQ studies establish confidence that the equipment and ancillary systems are
capable of consistently operating within the established design and operating limits and
tolerances.
Design Qualification (DQ)
Design qualification (DQ) may be established as a separate process with its own protocol
or may be combined in the Validation Plan. The purpose of this qualification is to assure
that a proposed new or modified facility, system or equipment meets GMP requirements,
is suitable for its intended purpose and defines how the Users Requirement Specification
(URS) are to be met.
The URS establishes agreed and properly defined facilities/utilities/equipment functionality
requirements. Variables should be specified in terms of expected reliability, consistency and
capability.
Auditing a Validation System
9
The protocol should include the roles and responsibilities for IQ development, execution,
assurance of completion and maintenance. It should identify functional groups required
to review and approve the protocol. Functional groups should include those who can
provide a technical evaluation (e.g., Engineering, Maintenance) and QA. QA should
always be an approver to ensure that all GMP and regulatory requirements have been
considered, met, and documented as appropriate. If all critical items in the protocol have
been addressed and approved, but there are some remaining items, the next phase of
qualification may be executed provided that there is written documentation to justify it.
The IQ protocol should include piping and instrumentation diagrams (P & ID), HVAC
drawings and equipment drawings.
Installation Qualification Execution
Installation qualification (IQ) occurs during the equipment installation phase. Equipment
is compared to the original design and installation plans, found either in the Purchase
Order or design specification. This phase ensures that the manufacturer s and the
company s engineering specifications agree. Equipment type and "as - built" conditions
are verified against the design specifications. During this phase, materials of construction,
manufacturer s identification (serial number) are verified against the purchase
specifications and the manufacturer s specifications.
During the installation qualification, utilities completion and calibration take place. The
utilities, (electrical, water, steam, vacuum, compressed gases and HVAC) are checked to
determine if they are connected properly. Wiring installation of electrical lines (loop
checks) is conducted. Software that is used in a computer is checked for the correct
version and that it is installed correctly.
Instruments are also calibrated to verify that indicators for temperature, pressure, flow,
weight, volume, etc. are accurate. Piping systems are reviewed to verify that they have
been correctly installed and that the composition of materials is in agreement with the
design specifications. Piping systems are also reviewed to ensure that the lines have been
degreased, and passivated, if the water systems use stainless steel piping. Piping should
also be identified and tagged.
The installation qualification should include a review of pertinent maintenance
procedures; repair parts lists, and calibration methods for each piece of equipment. The
objective is to assure that all repairs can be performed in such a way that will not affect
the characteristics of material processed after the repair. In addition, special post -repair
cleaning and calibration requirements should be developed to prevent inadvertent
manufacture a of non-conforming product
Information from the installation qualification should be used to:
Ø develop written procedures on calibration
Ø establish a preventative maintenance program
Ø determine which calibration, maintenance and adjustment requirements could
affect the process and product.
Auditing a Validation System
11
Performance Qualification Execution
Performance qualification is a bridge between Operational Qualification and Process
Validation. In Performance Qualification, production materials, qualified substitutes, or
simulated product may be used to test the upper and lower operation limits or worst
case conditions. Performance Qualification is a test of the overall system, with
equipment and ancillary systems tested together.
Process Validation
Process Validation includes establishing documented evidence, which provides a high
degree of assurance that a specific process will consistently produce a product meeting its
pre-determined specifications and quality characteristics.
A predetermined number of validation batches should be manufactured to demonstrate
that, under normal conditions and defined ranges of operating parameters, the
commercial scale process appears to make an acceptable product. It should normally
cover the manufacture of at least three consecutive batches of material.
Validation should be performed under conditions to be used for routine manufacture. The
batch size should be the same as or representative of the intended commercial scale
batches. Sampling and testing should be carried out to ensure compliance with the most
stringent requirements.
If the validation batches are intended for commercial use, the conditions under which
they are prepared and manufactured must comply with the GMP requirements.
Prospective process validation should normally be applied to API and to medicinal
products. The general principle is to validate a manufacturing process and the same
process can typically be used for several related products. Rather than to develop a plan
for each product manufactured by a process, it can be possible to develop a plan for that
process instead. There are two general principles that could be applied. Matrix
approach generally means a plan to conduct process validation on different strengths of
the same product. Family approach describes a plan to conduct process validation on
different, but similar products. Either approach must demonstrate that the process is
consistent for all the strengths or products involved. The plan should be designed to
evaluate all likely sources of variation in the products manufactured by the process.
Process validation must be completed, evaluated, documented and approved before
commercial distribution.
Worst Case and Challenge Tests
The process should be challenged by making deliberate changes to demonstrate its
robustness and to define its limits of tolerance. In challenging a process to assess its
adequacy, the conditions should simulate those that could be encountered during actual
production. These tests should be repeated enough times to assure that the results are
meaningful and consistent.
Auditing a Validation System
13
equipment is considered clean prior to use. For modern, highly automated, equipment the
logical sequencing of cleaning activities should be critically reviewed for adherence to
validated procedures. Maximum permitted time for campaign manufacturing should be
included in cleaning validation studies.
Normally an ongoing monitoring program is set up to confirm satisfactory operation
of the established cleaning process. Key process parameters may be reviewed for
automated systems, in other cases monitoring based on swabs/rinse samples or visual
examination may be conducted.
Validation of Analytical (Chemical, Physical and Microbiological) Test
Methods
Validation of methods is performed to confirm that the performance characteristics of a
method meet the requirements for the intended application. An assessment of validation
studies demonstrating that the method is suitable for its intended purpose shall be
documented appropriately.
Test methods described in any submission for manufacturing authorization, including
development pharmaceuticals, in-process control during manufacture, control tests on
intermediate products, control tests on finished products and stability testing should be
appropriately validated for the phase of development before use. Methods used in testing
of commercial finished products, raw material or packaging component must be validated
before use.
Appropriate level of revalidation of methods may be required by regulatory authori ties
when methods are transferred from one site to another, methods are transferred from one
laboratory to another, if significant changes are made in the manufacturing of major
starting materials or if the composition of the finished product is changed .
Methods and equipment described in pharmacopoeias may be considered validated.
Suitability for use of pharmacopeial methods should be established by testing/technical
review and documented. When claiming compliance with pharmacopoeias, unless the
exact method is being used, a cross validation should be performed.
A number of parameters should be considered in validation of quantitative methods.
These parameters include tolerance limits, specificity, accuracy, precision, detection
limit, quantification limit, linearity and range.
Maintenance of Validated Status/Change Control
The suitability of a facility, utility, process or equipment must be maintained through its
operating life. Systems and documentation need to be in place to support this. Such
monitoring systems should include SOPs, performance monitoring, calibration,
maintenance and cleaning, change control, and periodic review by internal audits. Where
no significant changes have been made to the system or process, and a quality review
confirms that the system or process is consistently producing material meeting its
specifications, there is normally no need for revalidation.
Auditing a Validation System
15
· Verify that a Validation Protocol exists for each validation project, which identifies
qualification activities for DQ/IQ/OQ/PQ and their locations.
· If DQ is performed as a separate qualification,
Ø Verify that there is a system to manage the setup of a preventive maintenance
and change control program for new equipment. This management system
may be part of the IQ/OQ process or part of the site s change control system.
· Ensure that the site has performed the following checks during the IQ phase.
Ø For electrical and instrumentation completion,
i Verify that there is documentation of loop checks.
i Verify that the software version installed is documented.
Ø For installation and instrumentation,
i Verify that the delivered equipment and instrumentation meet the defined
specifications of equipment order.
Ø For utilities completion,
i Verify that there is documentation (e.g., a utilities checkli st) of the
connection of utilities to equipment.
Ø For piping systems completion,
i Verify that the system "walk down" was completed.
i Verify that any red line changes to the P&ID's have been submitted.
Ø Verify that the following documentation is correct:
i As built engineering and installation drawings
i calibration documentation
i weld documentation
i material of construction certifications
· Ensure that the Operational Qualification is complete.
Ø Verify that the valve sequencing (e.g. API tanks) is correct by comparing it
against the piping and installation drawing.
Ø Verify that ranges for process and safety alarms have been set according to
process parameters.
Ø Verify that critical alarms are present by comparing the alarms against a list of
critical parameters.
Ø Verify that the range tested in the protocol encompasses the current alarm
condition.
Ø Verify that worst case conditions have been tested for the equipment and
ancillary systems.
· Ensure that the documentation system is operating in full compliance.
Ø Verify that when any protocol has been approved, any changes to the protocol
followed the site s change control process.
· Ensure that the Performance Qualification is complete.
Ø Verify that the testing performed is within established acceptance criteria.
· Ensure that appropriate SOPs and batch documentation are developed for the
equipment and systems being qualified. Ensure that any restrictions to use are
implemented.
·

Pharmaceutical Validation Documentation Requirements

Pharmaceutical validation is a critical process that ensures that pharmaceutical products meet the desired quality standards and are safe fo...