Pharmaceutical Cleaning & Cleaning Validation

Download as pdf or txt
Download as pdf or txt
You are on page 1of 31

RedLotus Pharmtech Private Limited

Along with its identified and qualified associates and


partners RedLotus, "Pharma Technical Services
Company" brings in years of collective experience in the
Date: 18 May 2018
areas of a drug substance as well as a drug product
Document No. GD009
development, analytical method development & transfer,
Version: 00
technology transfer, manufacturing, quality assurance &
control, regulatory affairs, pharmaceutical engineering
qualification & validation and supply chain.

A Pharma Technical Services Company

F-702, Goodwill Paradise,


Plot No. 24, Sector 15,
Kharghar, Navi Mumbai,
PIN – 410210
MAHARASHTRA
+91 9930 912 577
Guidance Document
Cleaning & Cleaning Validation
RedLotus Pharmtech Private Limited

Table of Contents

1.0 INTRODUCTION – RedLotus Pharmtech ................................................................................ 4

2.0 PURPOSE ................................................................................................................................. 5

3.0 BACKGROUND ......................................................................................................................... 5

4.0 SCOPE....................................................................................................................................... 6

5.0 INTRODUCTION ....................................................................................................................... 6

6.0 REGULATORY REQUIREMENT .............................................................................................. 7

7.0 PROCEDURE ............................................................................................................................ 8

7.1 Multiple Levels of Cleaning (API) ........................................................................................ 8

7.1.1 Level 1 Cleaning (Type A) ................................................................................................. 9

7.1.2 Level 2 Cleaning (Type B) ................................................................................................. 9

7.1.3 Level 3 Cleaning (Type C) ................................................................................................. 9

7.1.4 Level 4 Cleaning (Type D) ................................................................................................. 9

7.2 Multiple Levels of Cleaning (Drug Products) .................................................................... 10

7.2.1 Level 1 Cleaning (Type A) ............................................................................................... 10

7.2.2 Level 2 Cleaning (Type B) ............................................................................................... 10

7.3 Development of Cleaning Program ................................................................................... 11

7.3.1 Nature of Contaminant .................................................................................................... 11

7.3.2 Product Grouping and Worst-Case Selection ................................................................. 11

7.4 Selection & Development of Cleaning Method ................................................................. 13

7.4.1 Clean-In-Place (CIP) Method .......................................................................................... 13

7.4.2 Clean-Out-Of-Place (COP) Method................................................................................. 13

7.4.3 Manual Cleaning Method ................................................................................................ 13

7.5 Selection of Sampling Method ........................................................................................... 15

7.5.1 Direct Surface (Swab) Sampling Method ........................................................................ 15

7.5.2 Rinse Sampling Method .................................................................................................. 15

7.5.3 Placebo Method............................................................................................................... 15

7.5.4 Immersion Method .................................................................................................... 15

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 2
RedLotus Pharmtech Private Limited

7.6 Selection of Analytical Method .......................................................................................... 16

7.6.1 Non-Specific Analytical Test Methods ............................................................................. 17

7.6.2 Specific Analytical Test Methods..................................................................................... 17

7.7 Establishment of Limits and Acceptance Criteria ............................................................. 17

7.7.1 Visible Cleanliness Limit (Limit Visible)............................................................................... 18

7.7.2 Limit calculation on the basis of Smallest Therapeutic Dose (Limit STD) ......................... 20

7.7.3 Limit calculation on the basis of Toxicity (Limit Toxicity) ..................................................... 20

7.7.4 Limit calculation on the basis of Equipment Surface Area (Limit ESA) ............................. 22

7.7.5 Limit calculation on the 10 – ppm basis (Limit 10 ppm) ...................................................... 23

7.7.6 Limit Calculation for Allergenic & Highly Potent Products (Limit Potent) ............................ 23

7.7.7 Microbiological Limits (Limit Micro) .................................................................................... 23

7.7.8 Selection of the appropriate acceptance criteria ............................................................. 24

7.7.9 Recovery Factor and Limits............................................................................................. 25

7.8 Cleaning Agents ................................................................................................................. 26

7.9 Hold Time Limits ................................................................................................................. 26

7.10 Planning & Organization of Cleaning Validation Study .................................................... 26

7.11 Documentation for Cleaning Validation............................................................................. 27

7.12 COMPLIANCE AND RE-VALIDATION .............................................................................. 28

8.0 ABBREVIATIONS AND DEFINITIONS................................................................................... 29

9.0 APPENDIX ............................................................................................................................... 30

10.0 REFERENCES ........................................................................................................................ 30

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 3
RedLotus Pharmtech Private Limited

1.0 INTRODUCTION – RedLotus Pharmtech


In the last two years, import ban orders have been issued by the USFDA and C anada’s
Health Canada to more than 40 API (active pharmaceutical ingredients) and formulations
manufacturers in Asia, primarily in India and China. GMP/ Quality non-compliance could
be considered as a grave threat to trip-up the Asian pharmaceutical industry,
demonstrating a steady double-digit growth rate in the last few years.
Leading pharmaceutical companies seek to achieve and maintain world-class excellence
in all aspects of pharmaceutical quality from research and development through
manufacturing. In pursuit of this goal, savvy companies continuously strive to upgrade
and improve the development and manufacturing set-up to match the standing and
business performance of the company and ensuring compliance with the dynamic
regulations. Usually these efforts work for the companies up to a certain limit, drying
research pipeline and constant market pressures of product cost reduction without
compromising the product quality often results in a decision of making these highly
specialized resources redundant. This is contradicting but true to the findings of these
two industry reports.
This quality staffing and structures that support the full spectrum of various functions,
including research & development, vendor qualification/ development, manufacturing
process development, technology transfer, manufacturing facility & equipment design,
qualification & validation, process validation, trouble-shooting, related computer systems
validation, failure investigations, and continuous product/ process improvements is
important for any company to develop & manufacture a quality product. As all of these
are cross-functional activities, developing such skill-set takes enormous time and usually
this staff, become liability for an organization after completion of a research/ development
project(s).
With an intention to fill in this critical competence/ performance gaps RedLotus, “ A
Pharma Technical Services Company” has been formed. Along with its identified and
qualified national as well as international associates and partners (including ex-regulatory
officials) it can help pharmaceutical companies in responding to resolve with guarantee
any GMP/ Quality issue arising from a regulatory inspection, such as the USFDA warning
letter and/ or prospectively work with the companies in developing, establishing and
continuously improving each element/ system (Quality, Materials, Equipment & Facility,
Production, Laboratory, Labeling & Packaging) that constitutes a reliable and robust
GMP/ Quality compliance system.
According to a conservative estimate, about 25 percent of the costs of bringing a
pharmaceutical product to market comes from manufacturing expenses. By adopting
optimal structures and strategies for manufacturing technical services, companies can
maximize resources while positioning the function as an improvement arm for processes
across the supply chain. RedLotus Pharmtech can play a critical role in ensuring safe
and efficacious products reaching the market, by using the most cost-effective methods
possible, and in guiding the companies at different stages of project, as explained below:
(a) Design & Construction:
During the manufacturing process/ product development stage in designing and
selection of right facility, equipment & technology, their qualification, process
validations, data handling, computer system & critical support systems validation,
and establishing robust quality assurance and management systems/ processes.
(b) Operation, Maintenance & Compliance:
During the regulatory approval phase in preparing for the regulatory approval
inspections by performing technology transfer, mock inspections, identification &
resolution of GMP compliance risk/ gaps, staff training, resolution of regulatory
inspectional findings/ observations.
(c) Continuous Improvement:
During the commercial manufacturing phase in trouble-shooting, failure
investigations, continuous process and quality systems performance

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 4
RedLotus Pharmtech Private Limited

verification and improvement by performing general as well as specific GMP


audits/ gap-analysis.
All of the above – if done well – can avoid regulatory challenges, costly product recalls
and facility shutdowns.
For further details on RedLotus Pharmtech & our services, please refer Annex – 1, and
also visit the website www.redlotuspharma.com
2.0 PURPOSE
While guidelines are available from many regulatory agencies on almost every GMP
topic/ requirement and expectation, it’s effective implementation in a real-life operation/
application is entirely dependent upon correct interpretation and accurate correlation with
own operation/ application. Manufacturers often fail to interpret the guidelines, thus
resulting in a deficiency/ gap which could adversely impact a regulatory GMP inspection
outcome. An adverse outcome of a regulatory GMP inspection has far reaching impact
on the entire organization business, profits and business continuity.
For the benefit of our clients and customers, we consolidate our knowledge, experience,
learning, and interpretation of various guidelines on the subject in this, “ RedLotus
Guidance Document”. This document includes lots of examples, explanations, references
and visuals for a better understanding on this subject.
We believe, this document would be helpful in understanding this topic and thus in correct
implementation of appropriate, adequate and efficient system, process and/ or control for
GMP compliance on this topic.
3.0 BACKGROUND
The regulatory requirement of equipment cleaning prior to the use is not new. USFDA
1963 GMP Regulations (Part 133.4) stated as follows "Equipment *** shall be maintained
in a clean and orderly manner ***." A very similar section on equipment cleaning (211.67)
was included in the 1978 cGMP regulations. Of course, the main rationale for requiring
clean equipment is to prevent contamination or adulteration of drug products.
Historically, regulatory investigators have looked for gross insanitation due to inadequate
cleaning and maintenance of equipment and / or poor dust control systems. Also,
historically speaking, the regulatory authorities were more concerned about the
contamination of non-penicillin drug products with penicillins or the cross-contamination
of drug products with potent steroids or hormones. A number of products have been
recalled over the past decade due to actual or potential penicillin cross-contamination.
One event which increased awareness of the potential for cross contamination due to
inadequate procedures was the 1988, US recall of a finished drug product,
Cholestyramine Resin USP. The bulk pharmaceutical chemical used to produce the
product had become contaminated with low levels of intermediates and degradants from
the production of agricultural pesticides. The cross-contamination in that case is believed
to have been due to the reuse of recovered solvents. The recovered solvents had been
contaminated because of a lack of control over the reuse of solvent drums. Drums that
had been used to store recovered solvents from a pesticide production process were later
used to store recovered solvents used for the resin manufacturing process. The firm did
not have adequate controls over these solvent drums, did not do adequate testing of
drummed solvents, and did not have validated cleaning procedures for the drums. Some
shipments of this pesticide contaminated bulk pharmaceutical were supplied to a second
facility at a different location for finishing. This resulted in the contamination of the bags
used in that facility's fluid bed dryers with pesticide contamination. This in turn led to cross
contamination of lots produced at that site, a site where no pesticides were normally
produced.
USFDA instituted an import alert in 1992 on a foreign bulk pharmaceutical manufacturer,
which manufactured potent steroid products as well as non-steroidal products using
common equipment. This firm was a multi-use bulk pharmaceutical facility. USFDA
considered the potential for cross-contamination to be significant and to pose a
serious health risk to the public. The firm had only recently started a cleaning

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 5
RedLotus Pharmtech Private Limited

validation program at the time of the inspection and it was considered inadequate by
USFDA. One of the reasons it was considered inadequate was that the firm was only
looking for evidence of the absence of the previous compound. The firm had evidence,
from TLC tests on the rinse water, of the presence of residues of reaction byproducts and
degradants from the previous process.
Following figure shows the break-up of cGMP deficiencies cited by USFDA in the FY
2010 in the inspection of India pharmaceutical companies. Equipment cleaning and
cleaning validation constitutes a major deficiency area.

4.0 SCOPE
This guidance document is applicable for designing, implementing, and continuously
improving the cleaning, cleaning validation concept and approach in a drug substance
and a drug product manufacturing set-up. This document is intended to address special
considerations and issues pertaining to validation of cleaning procedures for equipment
used in the manufacture of pharmaceutical products, active pharmaceutical ingredients,
and biological drugs. The document is also intended to establish inspection consistency
and uniformity with respect to equipment cleaning procedures.
Principles incorporated in the corporate policy, international guidance(s) and current
industry trends in cleaning & cleaning validation have been taken into consideration while
preparing this guideline. This guideline is intended to cover validation of equipment
cleaning for the removal of contaminants associated with previous products,
intermediates, residues of cleaning agents as well as the control of potential microbial
contaminants.
This document does not cover the cleaning and cleaning validation of manufacturing
plant, facility, premises, laboratory equipment / apparatus / glassware, apparels used by
the manufacturing personnel, dedicated piece of equipment / change parts and utilities
such as water / gas systems etc. However, the principles and concepts can be applied
for cleaning / validation as it is or after modification according to the application or nature
of the equipment / system.
5.0 INTRODUCTION
In context to the application of cleaning in the pharmaceutical industry, cleaning is defined
as, “the process of removing contaminants from process equipment, such that the
equipment can be safely used for subsequent product manufacture”.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 6
RedLotus Pharmtech Private Limited

The objective of cleaning validation of equipment, utensils and components is to establish


sufficient documented evidence to assure that, the cleaning procedure can reproducibly
remove residue of the previous material / product below the established acceptance limit.
The quantity of residue of the previous material / product (within the acceptance limit) will
not affect quality and safety of the subsequent product manufactured using the same
manufacturing equipment.
The cleaning Validation requirements not only apply to the drug product manufacturing
equipment but also apply to the API manufacturing equipment as well. Cleaning of the
equipment train, individual equipment, utensil and / or component should be cleaned
separately and clubbed with a pre-wash and inspection program. Usually, the cleaning
procedure comprise of thorough cleaning with detergents, neutralizing agents, chelants
or solvents alone or in suitable combination followed with final rinsing with high quality
pharmaceutical grade water such as Purified Water or Water for Injection also called as
Placebo. The final rinse water is then tested for the pH, TOC and conductivity in
conformance with the Pharmacopoeial &/or in-house standard of acceptance.
Cleaning process for the API can be considered more complex as compared to the
cleaning procedure for drug product manufacturing equipment due to the complicated
nature of manufacturing process and equipment itself. The API manufacturing process
may be of multiple steps involving chemical and / or physical transformation at each step
thus involving potential for generation of more residues and contaminants. The API
manufacturing Equipment such as material / product transfer piping, pumps, valves,
elbows / bents may prove to be, difficult for cleaning.
During development of cleaning procedure and its validation, due importance should be
paid to the residue and contaminants. The residue and contaminants may include but not
limited to the precursors, by-products, desired actives, solvents, microbes, bacterial
endotoxin and equipment related materials such as equipment linings, gaskets, filter
agents and / or lubricants.
The following basic differences in manufacturing process of API and drug product
influence the cleaning program and procedures.
• Due to the nature of manufacturing process, API may contain precursor and by-
products along with the actives and residual solvents i.e. more potential residues
and contaminants than the drug product manufacturing.
• The precursor and by-products often have clinical or toxicological activity, thus
making them more serious contaminants than a harmless excipient residue from a
dosage form manufacturing process.
• A typical manufacturing process of API involves chemical reactions in the early
steps and purification in the terminal / final step, on the other hand usually there is
no further purification step involved in the manufacturing of Drug Product. The
terminal / final steps are combination of re-crystallization, filtration and / or other
purification steps.
• In view of the above differences between the manufacturing process of API and
drug product following points should be considered while developing the cleaning
procedure or cleaning validation study.
- Identification of each potential contaminant present and there clinical and
toxicological activities.
- Residue from the early steps of API manufacturing may be subsequently removed
in the final purification steps; hence cleaning requirement should be more flexible
in the early steps and more stringent in the final steps of manufacturing.
6.0 REGULATORY REQUIREMENT
Truly the basis of complex cleaning program and procedures has actually arrived from
the regulatory requirement for cleaning.
(I) As per the cGMP regulations, Section 211.67 of the CFR, “Equipment cleaning
and maintenance”

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 7
RedLotus Pharmtech Private Limited

(a) Equipment and utensils shall be cleaned, maintained, and sanitized at


appropriate intervals to prevent malfunctions or contamination that would alter
the safety, identity, strength, quality or purity of the drug product beyond the
official or established requirements.
(b) Written procedures shall be established and followed for cleaning and
maintenance of equipment including utensils used in the manufacture,
processing, packing, or holding of a drug product. These procedures shall
include, but are not limited to, the following:
(1) Assignment of responsibility for cleaning and maintaining equipment;
(2) Maintenance and cleaning schedules, including, where appropriate,
sanitizing schedules;
(3) A description in sufficient detail of the methods, equipment and materials
used in cleaning and maintenance operations, and the method of
disassembling and reassembling equipment as necessary to assure proper
cleaning and maintenance;
(4) Removal or obliteration of previous batch identification,
(5) Protection of clean equipment from contamination prior to use;
(6) Inspection of equipment for cleanliness immediately before use.
(7) Records shall be kept of maintenance, cleaning, sanitizing and inspection.
(II) The “Guide to Inspections of Bulk Pharmaceutical Chemicals”, instruct the inspector:
(a) To evaluate the API facility for the “potential for cross-contamination from any
source and relative ease and thoroughness of clean up”.
(b) To focus on the cleaning of product contact surfaces, “Cleaning of multiple use
equipment is an area where validation must be carried out”.
(c) To give specific inspectional coverage for, (1) Detailed cleaning procedure, (2)
Sampling plan & (3) Analytical method/Cleaning Limits.
(III) Another Agency document “Biotechnology Inspection Guide” bears remarkable
similarity in conceptual approach to cleaning validation.
(IV) As per the MHRA, “Rules and Guidance for Pharmaceutical Manufacturers and
Distributors 2007”.
(a) Section II, “Guidance on Good Manufacturing Practice (GMP)”, Sub-section 3.36,
“Manufacturing equipment should be designed so that it can be easily and
thoroughly cleaned. It should be cleaned according to detailed and written
procedures and stored only in a clean and dry condition”.
(b) Section II, “Guidance on Good Manufacturing Practice (GMP)”, Sub-section 5.19
(e), “using cleaning and decontamination procedures of known effectiveness, as
ineffective cleaning of equipment is a common source of cross-contamination”.
(c) Section II, “Guidance on Good Manufacturing Practice (GMP)”, Sub-section 5.20,
“Measures to prevent cross-contamination and their effectiveness should be
checked periodically according to set procedures”.
In the pre-approval Inspection (PAI) process, cleaning validation is the most important
issue, i.e. a company may be denied approval of their New Drug Application (NDA) or
Abbreviated New Drug Application (ANDA) for lack of satisfactory cleaning validation
package.
7.0 PROCEDURE
7.1 Multiple Levels of Cleaning (API)
In an API manufacturing process different cleaning situation may arise and each situation
may be quite unique such as;

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 8
RedLotus Pharmtech Private Limited

• Batch to batch changeover cleaning


• Changeover from early steps to intermediate within a given product sequence
• Changeover from intermediate of one product to intermediate of another product
• Changeover from intermediate of one product to final product of anot her product
• Changeover from one final product to another final product
In a non-dedicated / multiple products manufacturing facility different cleaning procedure
may evolve depending upon the manufacturing step and nature of the next manufacturing
step to occur in the manufacturing equipment. This might result in four different levels of
cleaning.
7.1.1 Level 1 Cleaning (Type A)
Used only between steps in the same manufacturing process.
Example – For manufacturing of product X, there are 4 steps as shown in the following
flow diagram.
Step A  Step B  Step C  Step D
If, for a particular equipment, step A of the first batch in the campaign is to be followed
by manufacturing step A of the second batch in the campaign, then a level 1 cleaning is
required.
7.1.2 Level 2 Cleaning (Type B)
Used when cleaning between steps in the same manufacturing process.
Example – For the above example level 2 cleaning would be used when step B is to be
performed immediately after completion of step A for the same product line (Product X)
or step D is to be carried immediately after step C and so on. In all these conditions level
2 cleaning will be used when no other product is manufactured in the equipment between
the two steps involved.
7.1.3 Level 3 Cleaning (Type C)
Used when cleaning between an intermediate or final product step of one product and an
intermediate step of another product
Example – For manufacturing of product X and product Y, there are 4 steps as shown in
the following flow diagrams.
PRODUCT X Step A  Step B  Step C  Step D
PRODUCT Y Step A  Step B  Step C  Step D
If Step C of Product Y is run immediately after Step B of Product X, then a level 3 cleaning
is required.
7.1.4 Level 4 Cleaning (Type D)
Used when cleaning between an intermediate or final product step of one p roduct and
final step of another product
If Step D of Product Y is run immediately after Step B of Product X, then a level 4 cleaning
is required. The only difference between the Level 3 and Level 4 cleaning is that in the
Level 4 cleaning, the next production will be a final product step.
The four levels of cleaning may differ in thoroughness of the cleaning process, cleaning
conditions, the level of verification of cleaning and degree of risk associated with
contamination, as shown in the following order.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 9
RedLotus Pharmtech Private Limited

Cleaning 
Level 1 Level 2 Level 3 Level 4

Risk Lowest ….. ….. Highest

Acceptance Limit Highest ….. ….. Lowest

Less
Extent of Cleaning ….. ….. More Intensive
Extensive
Visual Analytical
Cleaning Qualification ….. …..
Inspection Testing

This is a general principle followed for developing and / or implementation of a cleaning


procedure i.e. less efforts for early stages where less risk is involved, but this may not be
true for every cleaning situation. For example, cleaning program for a cytotoxic /
biologically active agent manufacturing facility cannot be developed based on this
approach.
7.2 Multiple Levels of Cleaning (Drug Products)
In a drug product manufacturing process two types of cleaning situations as mentioned
below may arise;
• Batch to batch changeover cleaning
• Product to product changeover cleaning
In a non-dedicated drug product manufacturing facility two different types of cleaning
procedures may evolve depending upon the changeover to occur in the same
manufacturing equipment.
7.2.1 Level 1 Cleaning (Type A)
Used only between batch to batch changeover of same product.
Example – If an equipment is used to manufacture batch A followed by batch B of a
product X in a manner as shown below;
Batch A  Batch B
Then, a level 1 cleaning is required between the changeover.
7.2.2 Level 2 Cleaning (Type B)
Example – If an equipment is used to manufacture a batch of product X followed by
another batch of product Y (containing different API’s) in a manner as shown below;
Product X  Product Y
Then, a level 2 cleaning is required between the changeover.
The two levels of cleaning may differ in thoroughness of the cleaning process, cleaning
conditions, the level of verification of cleaning and degree of risk associated with
contamination, as shown in the following order.

Cleaning 
Level 1 (Type A) Level 2 (Type B)
Risk Lowest Highest
Acceptance Limit Highest Lowest
Extent of Cleaning Less Extensive More Intensive
Cleaning Qualification Visual Inspection Analytical Testing

This is a general principle followed for developing and / or implementation of a cleaning


procedure i.e. less efforts for same product batch to batch changeover where less

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 10
RedLotus Pharmtech Private Limited

risk is involved and more efforts for the product to product (containing different API’s)
changeovers.
7.3 Development of Cleaning Program
A systematic approach should be followed to develop a cleaning program either for drug
product or API manufacturing equipment / system / facility. Various aspects related to the
factors affecting the cleaning procedure should be considered, critically reviewed and
evaluated accordingly prior to the development. Some of the factors, that could affect the
development of cleaning program, are described in the following section.
7.3.1 Nature of Contaminant
An efficient cleaning program cannot be developed without understanding the nature of
potential contaminants involved in manufacturing process. The potential contaminants in
a drug product manufacturing could be residue of the previous product, colour, excipients
etc. Similarly, in API manufacturing the potential contaminants could be unreacted
starting materials (precursors / reactants), by-products of the various chemical synthesis
reaction. However, cleaning agents, lubricants, solvents, micro-organisms, bacterial
endotoxin, filter media, gasket materials, tank and transfer pipe linings etc. could also be
potential contaminants that could contaminate the drug product and / or the API’s.
To evaluate cleanliness with respect to the variety of contaminants, thorough physical,
chemical and biological testing is required. As these are highly specific methods there
are chances that a contaminant may not be detected if a wrong method is selected, hence
the testing method should be based on the nature of all potential contaminants.
Additionally, nature of contaminant will determine the specific requirements and extent of
cleaning verification / validation. Following figure-1 (a decision tree) would be useful in
determination of the cleaning verification / validation requirements and extent depending
upon the nature of the contaminant. Additionally, this decision tree would be also useful
in making a decision to validate the cleaning process on a prospective or a concurrent
basis.
Traditionally, it is considered that any process is considered validated based on
successful achievement of the validation criteria on three consecutive runs / trials /
batches / replicates. However, in a real life manufacturing scenario it could be difficult in
some circumstances to manufacture three batches just to simulate three changeover
conditions only for the cleaning validation studies, such as trial product, clinical batch,
exhibit / demonstration batch and products with very low market demand. In such
conditions cleaning verification of each cleaning cycle should be performed following the
same controls and criteria for each cleaning cycle until enough confidence is gained. The
cleaning process is considered validated based on retrospective review of each cleaning
verification results meeting the acceptance criteria.
7.3.2 Product Grouping and Worst-Case Selection
During development of a cleaning program it is a normal practice to group similar
products into a single group (such as all tablet or oral liquid formulations in drug product
manufacturing facility) or group based on combination of the equipment used and the
cleaning process employed (Usually in API manufacturing facility). Few examples of
equipment grouping based on application / use of the equipment is shown in the following
figure-2.
Once the groups are formed then one or more, worst case condition may be selected to
evaluate the subject cleaning process. The worst-case selection is based on potency,
toxicity, solubility, stability and difficult to clean product / equipment or combination of
these.
The criteria for product grouping and selection of worst case condition should be based
on a written scientific rationale describing selection process. In case of multiple level of
cleaning, at least one suitable product should be validated for each level of cleaning viz.
one worst case for Level 1, one worst case for level 2 cleaning and so on.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 11
RedLotus Pharmtech Private Limited

Figure – 1: Decision tree for determination of cleaning validation/ verification requirement & extent
based on the contaminant nature
Cleaning
Assessment

Identify all potential


contaminants (such as API,
Potential Document rationale in the Cleaning verification /
starting materials, reactants, YES NO
Residue? cleaning assessment / CVP validation not required
degredants, cleaning agents,
endotoxin, microbes etc)

Are residue
harmful to next product, NO
batch or patient?

Is it practical
to verify cleaning after Document rationale in the
NO (Validation)
each cleaning cleaning assessment / CVP
cycle?

YES (Verification)

Identify all potential


contaminants (such as API, Perform cleaning validation
starting materials, reactants, study as per the defined
degredants, cleaning agents, CVP
endotoxin, microbes etc)

Perform cleaning verification


Compile all cleaning
as per the defined CVP
verification results, evaluate
concurrently with Cleaning Validation Report
as per the instructions in
changeover for no of times
CVP
as defined in the CVP

Figure – 2: Examples of equipment grouping


F
A
API - Step 1 B DF - Step 1 DF - Step 1 DF - Step 1
R
I
I C
N A
T T
E API - Step 2 I DF - Step 2 DF - Step 2 DF - Step 2
R O
M N
E
D
I T
API - Step 3 A DF - Step 3
A
T B
E DF - Step 3
C
API - Step 4 O
M
P

I
F N
I API - Step 5 S DF - Step 4
N P
A E
L C
T
I
API - Step 6 DF - Step 5
P O
R N
O
D &
U
C API - Step 7 P DF - Step 6
T K
G

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 12
RedLotus Pharmtech Private Limited

7.4 Selection & Development of Cleaning Method


There are following three types of cleaning methods utilized in the drug product as well
as the API manufacturing facilities.
7.4.1 Clean-In-Place (CIP) Method
• Cleaning of the equipment is performed in place without disassemb ling.
• Cleaning process may be controlled manually or by an automated program.
• Very consistent and reproducible cleaning method.
• Can be validated readily.
• Being a closed system visual inspection of all components is difficult.
7.4.2 Clean-Out-Of-Place (COP) Method
• Cleaning of the disassembled equipment is performed in a central washing
machine.
• The washing machine also requires validation such as the temperature, ultrasonic
activity, cycle time & sequence, detergent quantity dispensed etc.
• Equipment can be visually inspected during the reassembly.
7.4.3 Manual Cleaning Method
• Difficult to validate.
• Most extensive and elaborate cleaning procedures are required.
• A High quality and extensive training program is required.
For any type of cleaning method as described above the cleaning process or sequence
usually remain similar, the only difference is about the controls and extent of human
involvement. Usually, any cleaning process / procedure / sequence generall y include
three steps (a) Pre-cleaning, (b) Cleaning and (c) Post-cleaning.
Following table-1 provides guidance on development of a cleaning process including the
appropriate process controls and step / process end point.
For any type of cleaning method used, cleaning validation is a basic requirement,
samples should be taken, analyzed and results documented. Both the hardware and
software involved in the cleaning process should be validated. After validation of the
cleaning method, suitable documents / records should be maintained for each cleaning
cycle to demonstrate that cleaning process parameters such as the temperature, cycle
time and sequence, quantity of detergent dispensed etc. are still being achieved and / or
followed on a routine basis. These records may be incorporated in the Batch Production
Record.
Once the cleaning method is validated, it should be ensured that the method is not
changed. If any change is required in the validated cleaning method, then the change
should be governed by the company’s change control policy / SOP and the need for re -
validation should be evaluated critically.
Development of a cleaning process is equally important to the selection of cleaning
method. Development of suitable cleaning method is usually done during the
development and / or the equipment qualification process. These development studies
should be properly documented as these often becomes the basis for the development
of cleaning SOP’s and the cleaning validation protocols. The cleaning process
development activity generally should cover the selection of cleaning agents and cleaning
cycle / process parameters, such as time, temperature, pH, flow rate, agitation /
scrubbing, (re)circulation etc. The cleaning process development studies are particularly
very useful in establishment of the process control parameters, identification of the
difficult to clean area and sampling locations for cleaning verification / validation / routine
sampling. Use of placebo or an inert fluorescent marker’s such as riboflavin is very
useful in development and / or establishment of the cleaning procedure / process.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 13
RedLotus Pharmtech Private Limited

Following pictures show the use of photography to help document hard-to-clean areas
while designing cleaning procedures.
Table – 1: Guidance for Development of Cleaning Process
Process Process
Sub-step /
Step / Description Control
options End-point Termination
Sequence Parameters
Powder
Volume, time, sucked &
Removal of residue at
turbidity / disposed as
gross level by using one or
Pre- color / per disposal
-- more of the following (a) Flow, pressure
cleaning conductivity / procedure.
scraping, (b) vacuum, (c)
pH of drain Drained to the
air jet, (d) water jet
water kill / effluent
tank
Removal of the residue
below the acceptable
residue limit by washing or
combination of soaking &
washing depending upon
Soaking
the nature of the residue
and / or the type of
Flow,
equipment. Usually a Volume, time,
pressure,
cleaning agent such as turbidity /
temperature, Drained to the
detergent, acid or alkali is color /
Cleaning pH, kill / effluent
used to remove the conductivity /
concentration tank
residue. The cleaning pH of drain
of cleaning
agent is usually water
agent
(re)circulated multiple
Cleaning / times and then drained for
washing better results.
Sometimes temperature of
the cleaning agent also
helps in achieving better
results.
Removal of the residue of
Rinse & drain cleaning agent below the
acceptable limits by rinsing Volume, time,
Neutralization with water and / or solvent pH,
Drained to the
& drain normally used in the next conductivity,
Post- kill / effluent
product planned for Flow TOC, specific
cleaning Final rinse & tank /
manufacturing. The rinsing method,
drain recycling tank
solutions are generally not visual
(re)circulated. inspection.
Drying Usually, rinse solvents /
solution is not heated.
Figure – 3: A UV-active placebo highlighting residues before cleaning and after cleaning

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 14
RedLotus Pharmtech Private Limited

Such studies and pictures are very helpful in assessing the cleaning process capability
and / or identification of the process limitation, difficult to clean spots, thus helping in
improving the cleaning process / cycle, augmenting the cleaning process with additional
cleaning step / requirement and identification of the sampling location(s) for cleaning
verification / validation studies.
7.5 Selection of Sampling Method
There are many types of sampling methods for cleaning purpose, being followed in the
industry, such as swab sampling, solvent rinse sampling, water rinse sampling, placebo
sampling, sampling of the following product, direct surface monitoring etc. Only the
following are most acceptable methods for cleaning purpose by the regulatory authorities.
7.5.1 Direct Surface (Swab) Sampling Method
• Strongly preferred method, as some residues may need a mechanical or physical
action to remove from the surface.
• Not suitable for the equipment which are difficult to access, such as inner surface
of the long hoses, transfer lines, small intricate instruments (as micronizers),
sieves / screens and brushes.
• The sampling medium and solvent used for extraction of residue (fr om the
sampling medium) may interfere with the analytical test.
7.5.2 Rinse Sampling Method
• Large surface area can be sampled.
• Strongly preferred method for difficult to access equipment.
• May indicate false result when, the residue need mechanical or physical ac tion to
remove from the surface. For example, when the contaminant is not soluble or
occluded in the equipment.
• The residue can be diluted below the level of detection, if large rinse volumes are
used.
7.5.3 Placebo Method
The placebo sampling method involves manufacturing a batch of placebo (the product
minus the active ingredient or contaminant of interest) in the cleaned equipment. The
final placebo is then tested for the excluded component. There are several potential
problems with this method. The first results when contamination occurs at one point in
the process, so the placebo products that are formed first are likely to be more
contaminated than those that follow later. Secondly a lot of material is wasted in
manufacturing the placebo. Thirdly, the levels of contamination in the placebo may be
difficult to detect.
7.5.4 Immersion Method
Certain machines require complete / partial disassembly for complete and effective
cleaning, such as tablet compression machine, size reduction mills, liquid / powder
filling machines etc.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 15
RedLotus Pharmtech Private Limited

Some of the components that are disassembled for cleaning are small and could be very
intricate to enable adequate and effective sampling by one of the above sampling
methods. In such conditions, the small components which are difficult to sample could be
immersed completely into a suitable immersion agent, such as water or solvent. If
required certain level of agitation, such as compressed air or ultrasonic could be used to
enhance removal of the residue from component surface. Complete immersion agent is
then recovered and sample is analyzed to determine the residue levels using a suitable
validated analytical method.
With this method it is possible that, the residue can be diluted below the level of detection,
if large volumes of immersion agent are used.
Irrespective of the sampling method a sampling procedure / protocol should be prepared
to demonstrate the following.
• Rationale for selecting a specific sampling method.
• Most difficult to clean location(s) is selected for sampling.
• Sampling location(s) is clearly defined and / or indicated on an actual photograph
or a schematic diagram.
• Detail of the sampling procedure including the volume of rinse sample, sampled
surface area (usually 25 cm 2 to 100 cm 2), swab specification (manufacturer,
catalogue number), number of swabs, condition of swab (wet / dry), number of
strokes of the swab & swabbing direction (swab pattern), blanks / controls, sample
labeling and transportation of the sample to the analytical laboratory.
Figure – 4: Swab pattern example

7.6 Selection of Analytical Method


It is equally important to select an appropriate method for detection of the residue in the
cleaning sample. The method must be selected carefully for the specific situation; a non -
specific analytical method may lead to false analytical results. The most common errors
associated in reporting the residue in cleaning samples occur due to:
• Selection of a non-specific analytical method
• The analytical method is not sensitive enough. Sensitivity is related to the limit,
normally it is assumed that “not detected” is zero, in fact “not detected” is equal to
or less than the sensitivity of analytical test method. Sensitivity of the analytical
test method is related to the limit of detection (LOD) and limit of quantitation (LOQ)
• Analytical test methods used for testing of cleaning samples are not validated
• Recovery of the expected residue from the equipment surface is not studied
• Interference of the sampling medium and solvent (used for extraction from the
medium) with the analytical test method is not studied

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 16
RedLotus Pharmtech Private Limited

There are various analytical test methods (specific as well as non-specific), which may
be employed for the detection of residue in the cleaning sample. The following are some
of the common analytical test methods.
7.6.1 Non-Specific Analytical Test Methods
Although it is expected to follow specific analytical test methods for the cleaning samples,
but many of the non-specific methods such as visual, pH, conductivity and TOC are
simple, fast and still provide valuable information related to the level of cleaning and
presence of any contaminant. Due to these properties these methods can be effectively
used for evaluation of cleaning and on-line monitoring application. Following are some of
the non-specific analytical methods, which are commonly used for cleaning validation.
• Visual Examination
• Gravimetric analysis
• pH
• Conductivity
• Microscopy
• Titration
• Total Organic Carbon (TOC)
7.6.2 Specific Analytical Test Methods
Several regulatory guidelines refer to use specific analytical test methods. The product
specific analytical test methods are specific and accurate but they take more time and
hence limited application in the routine monitoring. Following are some of the specific
analytical methods, which are commonly used for cleaning validation.
• High Performance Liquid Chromatography (HPLC)
• Thin Layer Chromatography (TLC)
• Fourier-Transform Infrared (FT-IR)
• Near Infra-red (NIR)
• Ion Chromatography
• Microbial/Endotoxin test
• Capillary zone electrophoresis (for Biotechnology products)
• Lowry protein (for Biotechnology products)
• ELISA - Enzyme Linked Immunosorbent Assay (for Biotechnology products)
• SDS-PAGE - Sodium Dodecyl sulphate-polyacrylamide gel electrophoresis (for
Biotechnology products)
Many times, during the analytical test method development a non-specific method is
utilized as a screening method to determine the level of cleanliness and the most difficult
portion to clean, followed by a product specific analytical test method with particular focus
on hard to clean location in the equipment.
7.7 Establishment of Limits and Acceptance Criteria
There cannot be a standard fixed limit to determine the effectiveness of a cleaning
procedure, due the reason that variety of equipment and products are used throughout
the API and drug product manufacturing industries. Rationale for the residue limit
established should be scientific, logical and based upon the knowledge of the material.
As per the Guide to inspections of Validation of Cleaning Processes the limits should be
“practical, achievable and verifiable”. Some limits prevalent in the industry are analytical
detection level such as 10 PPM, biological activity level such as 1/1000 of the normal

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 17
RedLotus Pharmtech Private Limited

therapeutic dose and organoleptic levels such as no visible residue. However, these limits
may not be applicable to each and every product.
To arrive at the quantitative acceptance limit for cleaning validation of particular
equipment there has to be a good scientific and logical rationale. A quantitative limit
should be based on one or more of the following:
• Therapeutic dose
• Toxicity of the material
• Solubility of the potential residue
• Difficulty of cleaning
• Use/Application of the product
• Nature of other products manufactured in the same equipment
• Batch size of other product manufactured in the same equipment
The limit is often based on allowing not more than a fraction of a therapeutic dose to be
present in a subsequent product. The fraction in this case is called as a “Safety factor”.
For example, quantitative limit for a contaminant in an ophthalmic product is 1/5000 th
fraction of smallest therapeutic dose. 1/5000 in this case is a “Safety factor”. The Safety
Factor is a measure of degree of risk for a particular situation. The degree of risk may be
different for API’s and drug product manufacturing again it will be dif ferent for different
dosage forms such as tablets and parenteral preparations. Normally accepted Safety
Factor for different dosage forms are given in the following table.
Table – 2: Safety factors
Dosage Form Safety Factor
Research Compound 1/100,000 – 1/10,000
Parenteral Products 1/10,000 – 1/5,000
Ophthalmic Products 1/5,000
Oral Dosage Forms (tablets, Capsules etc.) 1/1000
Topical Products 1/100 – 1/10
Before finalizing limit on the basis of these widely accepted safety factor limits, it must be
related to product use / application, therapeutic dose, toxicity and should be adjusted
accordingly. By using these factors, the acceptance limit may be calculated as described
in the following subsections.
7.7.1 Visible Cleanliness Limit (Limit Visible )

When the formal cleaning validation programs did not exist, visual inspection was the
primary means of determining equipment cleanliness. The use of visual inspection is still
typically a component of a cleaning validation program and for routine inspections of
cleaning effectiveness. The FDA, in their “Guide to Inspection of Validation of Cleaning
Processes,” limited the potential acceptability of “visually clean criteria” for use between
lots of the same product.
The Acceptable Residue Limit for drug residue is often determined on a health based and
/ or adulteration-based criteria and the limit used for the cleaning validation is usually
lowest of these two limits. A health-based limit is generated from toxicity data, which is
expressed as the Acceptable Daily Intake (ADI). The health-based limit is calculated
using the ADI or lowest therapeutic dose and the parameters of the equipment used to
manufacture the formulation. For the adulteration limit, a carry-over limit of 10 ppm or a
baseline limit of 100 μg/swab is often used in the industry. Health-based limit calculation
is explained in the subsequent sections of this guideline.
An established visible residue limit, which is below the Acceptable Residue Limit, is a
reasonable criterion for cleaning validation. Visible cleanliness is the absence of any
visible residue after cleaning, but a number of factors as described below influence
visual determination:

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 18
RedLotus Pharmtech Private Limited

• Observer visual inspector: Not only the observer’s visual acuity, but also training
on what to observe, influences the outcome of a visual inspection.
• Illumination: Level of illumination in the inspection areas and shadows caused by
the equipment influence what is seen.
• Distance and the angle of inspection: Distance and angle of the visual inspector /
observer from the equipment surface also have an effect.
• Nature of residue: The individual residues that comprise a given formulation affect
the overall visible residue limit.
A study of viewing parameters, including viewing distance, viewing angle, light inte nsity,
residue composition, and observer subjectivity help establishing optimal viewing
conditions to detect visible residues. In different published studies, various residues have
been observed down to 1.0 μg/cm 2 with the aid of a light source (Jenkins and
Vanderwielen). Fourman and Mullen qualitatively determined a visible limit at
approximately 100 μg per 2 × 2-inch swab area or about 4 μg/cm 2.
A general guidance for developing a study to establish Visible Residue Level (VRL) is
briefly described below:
• A solution or suspension of the API is applied at different concentrations to the
stainless-steel coupons.
• Multiple observers determine VRL levels under controlled viewing conditions (light
intensity, viewing distance & angle).
• The VRL is established at the lowest residue concentration, which all observers
successfully detected under standardized viewing condition.
Following figure-5 illustrates the visible residue appearance according to the
concentration of the API solution applied on SS coupon.
Figure – 5: Effect of API concentration on the residue appearance

Figure – 6: Effect of volume (fixed concentration API solution) on visible residue


appearance

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 19
RedLotus Pharmtech Private Limited

7.7.2 Limit calculation on the basis of Smallest Therapeutic Dose (Limit STD )

Let’s assume an example of an equipment used for manufacturing different tablet


products A, B, C & D all with different batch size (100, 200, 300 & 400 Kg respectively),
therapeutic dose (100, 200, 300 and 400 mg respectively) and maximum daily dose
(1000, 2000 3000 & 4000 mg respectively). As all are tablet dosage forms the safety
factor (SF) of 1/1000 is applied for calculation of acceptance limit (refer table-2).
Now, there are two methods to calculate the acceptance limit by using the smallest
therapeutic dose concept.
The smallest therapeutic dose (STD) amongst all the products is 100 mg for product A,
so the maximum allowable residue (MAR) limit can be calculated as follows:

𝑀𝐴𝑅 = 𝑆𝑇𝐷 × 𝑆𝐹
1
100 𝑚𝑔 × = 0.1 𝑚𝑔 𝑝𝑒𝑟 𝑑𝑎𝑖𝑙𝑦 𝑑𝑜𝑠𝑒
1000

Assume a situation of product change over from Product A to Product B. The maximum
daily dose (MDD) of Next Product B is 2000 mg (500 mg tablet q.i.d.) and the batch size
(BS) is 200 Kg. Maximum allowable limit of product A residue in 200 Kg batch of product
B can be calculated as follows:

𝑀𝐴𝑅 × 𝐵𝑆
𝑀𝐴𝑅𝑃𝑟𝑜𝑑𝑢𝑐𝑡 𝐴 =
𝑀𝐷𝐷

(0.1 𝑚𝑔) × (200 × 1000 × 1000 𝑚𝑔)


𝑀𝐴𝑅𝑃𝑟𝑜𝑑𝑢𝑐𝑡 𝐴 = = 10,000 𝑚𝑔
2000 𝑚𝑔

Hence, 10,000mg (10g) product A residue is allowed collectively for all manufacturing
and filling / packaging equipment. Using this method, a separate calculation would be
required for each product followed after product A, similarly separate calculation would
be required for each combination of product changeover, thus resulting in generation of
different acceptance limits. Having so many acceptance limits is difficult to handle and
could create confusion.
By assuming same example, a common acceptance limit for any combination of
changeover may be calculated by considering the smallest therapeutic dose (Product A,
100 Kg batch size) and highest daily dose among all the products (Product D, 4000 mg)
manufactured in the subject equipment.
A common acceptance limit for the residue of previous product in the subsequent product,
can be calculated as follows:
In this case a common limit for this particular example is calculated 2500 mg (2.5 g)
collectively for all manufacturing and filling/packaging equipment. Normally this method
of calculation is suitable for the drug product manufacturing facility where therapeutic
doses are established for almost each potential residue in the subject dosage form.

(0.1 𝑚𝑔) × (100 × 1000 × 1000 𝑚𝑔)


𝑀𝐴𝑅 𝐶𝑜𝑚𝑚𝑜𝑛 = = 2,5000 𝑚𝑔
4000 𝑚𝑔

7.7.3 Limit calculation on the basis of Toxicity (Limit Toxicity )

In case of API manufacturing scenario therapeutic doses for precursors, by-products,


cleaning agents etc. are not always available, hence in such situation the limit can be
calculated on the basis of different materials used in the manufacturing process. This

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 20
RedLotus Pharmtech Private Limited

method is based on the concept of acceptable daily intake (ADI) and no observed effect
level (NOEL).
The level of ADI can be calculated from the toxicity of the subject m aterial expressed as
LD50. This data is readily available in the references where toxicology data can be
obtained or in the Material Safety Data Sheet (MSDS). NOEL can be calculated from the
LD50 using the following equation.

𝑁𝑂𝐸𝐿 = 𝐿𝐷50 × 0.0005

Where, 0.0005 is a constant derived from a large toxicology database. NOEL can be then
used for calculation of ADI, by using the following equation.

𝑁𝑂𝐸𝐿
𝐴𝐷𝐼 =
𝑆𝐹

Where, SF is the Safety Factor. The maximum Allowable Residue (MAR) can be
calculated from the following equation.

𝐵
𝑀𝐴𝑅 = ADI ×
𝑅

Where, B is the smallest batch size of any of the product manufactured in particular
equipment / equipment train and R is the largest daily dose of any of the product
manufactured in the same equipment.
Example – Assume a condition where a chemical C is handled in equipment followed by
another product P in the same equipment. If the following information is known then MAR
can be calculated as follows.
• Toxicity of chemical C (LD50) – 100 mg/Kg (Oral) and 50 mg/Kg (IV)
• Smallest Batch Size manufactured in the equipment (B) – 10 Kg
• Max. daily dose of next product P manufactured in same equipment (R) – 2000 mg
(500 mg q.i.d.)

𝑁𝑂𝐸𝐿(𝑂𝑟𝑎𝑙) = 100 × 0.0005 = 0.05 𝑚𝑔⁄𝐾𝑔 𝑝𝑒𝑟 𝑑𝑎𝑦

𝑁𝑂𝐸𝐿(𝐼𝑉) = 50 × 0.0005 = 0.025 𝑚𝑔⁄𝐾𝑔 𝑝𝑒𝑟 𝑑𝑎𝑦

For an adult of 60 Kg,

𝑁𝑂𝐸𝐿
𝐴𝐷𝐼 =
𝑆𝐹

𝑁𝑂𝐸𝐿(𝑂𝑟𝑎𝑙) = 0.05 𝑚𝑔⁄𝐾𝑔 𝑝𝑒𝑟 𝑑𝑎𝑦 × 60 = 3.0 𝑚𝑔

𝑁𝑂𝐸𝐿(𝐼𝑉) = 0.025 𝑚𝑔⁄𝐾𝑔 𝑝𝑒𝑟 𝑑𝑎𝑦 × 60 = 1.5 𝑚𝑔

3.0
𝐴𝐷𝐼(𝑂𝑟𝑎𝑙) = = 0.003 𝑚𝑔
1000

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 21
RedLotus Pharmtech Private Limited

1.5
𝐴𝐷𝐼(𝑂𝑟𝑎𝑙) = = 0.00015 𝑚𝑔
10,000

Maximum Allowable Residue (MAR) limit can be calculated as follows.

(10 × 1000 × 1000 𝑚𝑔)


𝑀𝐴𝑅 (𝑂𝑟𝑎𝑙) = 0.003 𝑚𝑔 × = 𝟏𝟓 𝒎𝒈
2000 𝑚𝑔

(10 × 1000 × 1000 𝑚𝑔)


𝑀𝐴𝑅 (𝐼𝑉) = 0.00015 𝑚𝑔 × = 𝟎. 𝟕𝟓 𝒎𝒈
2000 𝑚𝑔

7.7.4 Limit calculation on the basis of Equipment Surface Area (Limit ESA )

The above sections 0 and 0 calculate the limit for Maximum Allowable Residue (MAR) on
possible manufacturing and packing equipment utilized in the manufacturing process.
Once the Maximum Allowable Residue limit is calculated it is practical and logical to
break-up the limit for individual equipment in an equipment train proportionately with
reference to the respective equipment surface area.
Example – In a manufacturing process following set of equipment are required,
considering the example of section 0 and assuming that the same equipment will be used
for manufacturing of API for Oral as well as Parenteral (IV) application and the surface
area of each equipment is known. Then MAR limit can be divided for individual equipment
as illustrated in the following table-3.
Table – 3: An example of limit calculation based on equipment surface area
Proportion of
Surface Area Limit (Oral) Limit (IV)
Equipment Total Surface
(Ft²) (mg) (mg)
Area (%)
Reactor 30 13.9535 2.0930 0.1047
Holding Tank 45 20.9302 3.1395 0.1570
Centrifuge 40 18.6047 2.7907 0.1395
Dryer 50 23.2558 3.4884 0.1744
Filters 10 4.6512 0.6977 0.0349
Product transfer lines 40 18.6047 2.7907 0.1395
Total 215 100 15 0.75

Again, depending upon the sampling method, the limits can be further subdivided. If rinse
sampling method, is followed and the complete equipment surface is rinsed then the
same limits as calculated in the above table can be used.
On the other hand, if Swab Sampling method is followed then the MAR limits calculated
for individual equipment must be further subdivided into the total surface area sampled
by swab method (or per swab). Assuming that 6 inch by 6-inch (0.25 Ft2) surface area is
sampled per swab and different numbers of swab samples were taken from different
equipment, in this condition the MAR limit for the total area sampled collectively can be
calculated as follows.

(𝑁𝑜. 𝑜𝑓 𝑆𝑤𝑎𝑏 𝑆𝑎𝑚𝑝𝑙𝑒𝑠 × 0.25 𝐹𝑡 2 )


𝑀𝐴𝑅 (𝐿𝑖𝑚𝑖𝑡 𝑓𝑜𝑟 𝑡ℎ𝑒 𝑠𝑎𝑚𝑝𝑙𝑒𝑑 𝑠𝑢𝑟𝑓𝑎𝑐𝑒) = × 𝑇𝑜𝑡𝑎𝑙 𝑀𝐴𝑅 𝐿𝑖𝑚𝑖𝑡
𝐸𝑞𝑢𝑖𝑝𝑚𝑒𝑛𝑡 𝑆𝑢𝑟𝑓𝑎𝑐𝑒 𝐴𝑟𝑒𝑎

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 22
RedLotus Pharmtech Private Limited

Table – 4: An example of limit calculation based on the sampled surface area


Total MAR MAR Limit for
No. of Equipment Limit Sampled Surface
Equipment Swab Surface Area (mg)**
(mg)
samples (Ft²)
Oral IV Oral IV
Reactor 6 30 2.0930 0.1047 0.1047 0.0052
Holding Tank 6 45 3.1395 0.1570 0.1047 0.0052
Centrifuge 4 40 2.7907 0.1395 0.0698 0.0035
Dryer 6 50 3.4884 0.1744 0.1047 0.0052
Filters 4 10 0.6977 0.0349 0.0698 0.0035
Product
4 40 2.7907 0.1395 0.0698 0.0035
transfer lines
Total 30 215 15 0.75 0.5233 0.0262
** Limit can be calculated in terms of the total swabbed area or per Swab.

7.7.5 Limit calculation on the 10 – ppm basis (Limit 10 ppm )

The maximum acceptable residue (MAR) limit in the subsequent product is calculated
using the ppm criteria in accordance with the following formula.

mg
MAR (mg) = 10 ppm ( ) × Batch Size (Kg)
Kg

The ppm criterion has the advantage that, it can be used regardless of the previous
product / subsequent product combination, which makes the limit calculation within a
complex validation project considerably easier. The worst case for the limit calculation
can be considered by using the smallest possible batch size for the subsequent product
as the basis for calculation.
On the other hand, this method of limit calculation has the disadvantage that it is
substance-independent, in other words, the therapeutic dose of the active
pharmaceutical ingredient is not included in the limit calculation. This means that the
various pharmacological properties of the different active pharmaceutical ingredients are
not taken into consideration. For highly potent drug substances, the limit calculated
according to the ppm criteria may therefore not be acceptable from a pharmacological
point of view.
7.7.6 Limit Calculation for Allergenic & Highly Potent Products (Limit Potent )

For certain allergenic ingredients, penicillins, cephalosporins or potent steroids and


cytotoxics, the limit should be below the limit of detection by best available analytical
methods. In practice this may mean that dedicated plants are used for these products.
However, in case of changeover from one product to another product within the same or
dedicated manufacturing facility, limit for maximum acceptable residue level can be
calculated according to the above sections. Due to the extremely small dosage, MAR
calculated in accordance with the guidance provided in above sections could sometimes
be so low that it cannot be detected by the analytical method. In such cases, the
acceptance criteria should be set on either the safety threshold derived from the
pharmacological data or below detection limit of the validated analytical method.
7.7.7 Microbiological Limits (Limit Micro )

One cannot expect equipment free from all micro-organisms, particularly when the final
step in cleaning does not involve sterilization and/or final rinsing with sterile water for
injection. Setting of the microbial limits is difficult, on the other hand it is important and
required because a microbial contamination of the equipment surface could possibly
result in product contamination.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 23
RedLotus Pharmtech Private Limited

There are no clear guidelines for the acceptable microbial contamination limit for the
process equipment and / or the environment used in the manufacturing of non-sterile
dosage forms or API. However, guidance on surface microbial contamination limits for
different clean room grades (A, B, C & D) used in manufacturing of sterile products does
exist. For the determination of limits, the "Supplementary and reviewed guidelines for
the manufacture of sterile medicinal products" in the appendix of the EU GMP Guideline
can be used. Limits (CFU/plate) for cleanroom classes A to D are specified in this
guidance document. In a recently published overview article, Seyfarth derived proposals
from this for implementing the requirements of the "Supplementary guideline for the
manufacture of sterile medicinal products" for the microbial status of surfaces. Alert and
action limits accepted by the FDA were also taken into consideration in this proposal. In
this proposal values given in this regulatory guidance document were graded as guideline
values or recommended limits which should be reached on average for a larger number
of measurements. This only concerns guideline values for sterile production (clean room
grades A-D). Data about the microbiological status of surfaces in “Controlled but Not
Monitored” so called “GMP areas” are not included in the EU Guideline. The guideline
value of 50 CFU / plate specified for product contact surfaces in areas of clean room
grade D can be used for product contact surfaces in the “GMP area”. According to
experience, it is not a problem to keep this value for freshly-cleaned equipment surfaces
presuming correct and hygienic execution of cleaning and drying.
The very first acceptance criteria are absence of pathogenic organisms such as E. coli,
Enterococcus, Salmonella etc. from the sampled equipment surface. For total viable
count limits in the following table could be used for guidance.
Table – 5: Suggested microbiological limits
Recommended limits for microbial Guidance for setting limits for table or
contamination of surface (Average values) equipment surface (CFU/ 25 cm2)
Contact Plates Guidance Action
Grade Alert Limit
Dia. 55 mm (CFU/ Plate) Value** Limit
A (Class 100) <1 <1 2 3
B (Class 100) 5 5 2 5
C (Class 10,000) 25 25 25 50
D (Class 100,000) 50 50 100 200
** Guideline values or recommended limits which should be reached “on average for a larger number
of measurements”

7.7.8 Selection of the appropriate acceptance criteria


With view to these considerations, the recommendation from the PIC/S guideline to use
the most stringent criteria to determine the limits is comprehensible. In the planning phase
of the cleaning validation, it is therefore necessary after identifying the worst-case
products to calculate the acceptance limits using the dose as well as the ppm criteria.
Based on this calculation, limits should be derived to define the corresponding
acceptance criteria for each critical active pharmaceutical ingredient, finally based on this
comparative calculation the above-mentioned rationale can be established.
The following example (see table) shows the limit calculation for the critical products A,
E and F.
Table – 6: An example of MAR limit comparison when calculated by dose & ppm methods

Next product MAR Limit (mg)


Lowest dose
Product Batch size Max daily dose Dose PPM
(mg)
(Kg) (mg) Method Method
A 500 100 5 x 1000 10,000 1,000
E 10 100 5 x 1000 200 1,000

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 24
RedLotus Pharmtech Private Limited

F 75 100 5 x 1000 1,500 1,000

In this example, it is assumed that the smallest batch size manufactured in a


manufacturing facility is 100 kg. According to the ppm criteria, the maximum acceptable
residue in the subsequent product is 1,000 mg for all three critical products.
For calculation in accordance with the dose criteria, the therapeutic dose of the critical
product and the daily dose of the subsequent product are still required. The latter is
calculated from the intake frequency and maximum daily dose of the individual products.
Following the solution approach described above, a fictitious subsequent product is used
as the basis for calculation, whose batch size is 100 kg and whose maximum daily dose
is 5 x 1 g. The resulting limits in the subsequent product lie between 200 and 10,000 mg.
A comparison of the limits shows that the dose criteria must be used for product E,
whereas the ppm criteria delivers the stricter limit for products A and F.
7.7.9 Recovery Factor and Limits
The limits calculated in the above are the acceptable residue limits in the analysed
sample. These limits should be adjusted with the established “Recovery Factor”, failure
to do this might result in a false result.
The recovery studies should be done as a part of the cleaning validation studies.
Typically, a recovery study comprises of the following.
• Deliberately contaminating the subjected equipment surface and/or similar coup on
surface (predefined area) with known concentration of the molecule of interest.
• Recover the interested molecule residue from the same surface by using the same
sampling techniques, which would be used during the validation studies.
• Determine the recovered quantity by using the same validated analytical methods,
which would be used during the validation studies.
• Establish a recovery factor based on the statistical evaluation of the results.
Example – An equipment surface of area 10 cm2 is deliberately contaminated with 10 µg
of the molecule of interest at five different locations. The contaminated surface is sampled
and subsequently analyzed by using the same methods, which would be used during the
actual validation studies to determine the actual recovered quantity. The recovery results
are as per the following table-7.
Table – 7: An Example for Recovery Factor Calculation
Initial Quantity (contamination) in µg (A) 10.00
8.92
9.09
Actual Recovered Quantity in µg 8.54
8.21
8.68
Average (B) 8.69
Standard Deviation 0.34
Recovery Factor = (B) / (A) 0.87

The simplest method to adjust the calculated limits based on the recovery factor is by
dividing the analytical result (sample) by the established recovery factor. For example, if
analytical result for a swab sample collected during a particular cleaning validation study
is 9 ppm, then the actual analytical result should be adjusted by the recovery factor 0.87
(for this example).

Actual Analytical Result (9 𝑝𝑝𝑚)


𝐴𝑑𝑗𝑢𝑠𝑡𝑒𝑑 𝑅𝑒𝑠𝑖𝑑𝑢𝑒 𝐶𝑜𝑛𝑡𝑒𝑛𝑡 = = 𝟏𝟎. 𝟑𝟓 𝒑𝒑𝒎
𝑅𝑒𝑐𝑜𝑣𝑒𝑟𝑦 𝐹𝑎𝑐𝑡𝑜𝑟 (0.87)

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 25
RedLotus Pharmtech Private Limited

7.8 Cleaning Agents


A cleaning procedure of an equipment / system may include the use of a cleaning agent
such as soaps, detergents, surfactants, acids, alkalis & organic solvents also. The
cleaning program should be designed to demonstrate the removal of cleaning agent(s)
below an acceptable level. Usually composition of the commercially available cleaning
agents is not provided by the manufacturer, this creates difficulty in testing the residue of
cleaning agents. Unlike the product residues, it is expected by the regulatory authorities
that no detergent level should remain after cleaning and same methods shall be followed
for determination of cleaning agents as were followed for determination of the product
residue.
7.9 Hold Time Limits
Establishment of time limits allowable between the process end & cleaning of the
equipment and maximum storage period of cleaned equipment before next use is very
important to demonstrate assurance to control the bio-burden through adequate cleaning
and storage of the cleaned equipment.
Sufficient documented evidence should be established to demonstrate that the routine
cleaning and storage of the cleaned equipment does not allow microbial gr owth. Any
equipment cleaned, sanitized and/or sterilized should be dried and adequately protected
to ensure that, there will be no (re)contamination, dust build-up and / or microbial
proliferation and microbial contamination of the cleaned equipment.
7.10 Planning & Organization of Cleaning Validation Study
Once the cleaning method / procedure is developed and acceptance criteria established
as described in previous sections, it is important to club all studies and / or derivations
together and prepare a comprehensive cleaning validation master plan and cleaning
validation protocols to avoid any kind of ambiguity in subsequent execution of the
cleaning validation studies.
Cleaning validation master plan is a high-level document, which establishes a broad plan
for a complete cleaning validation project and is used as a guidance document to the
cleaning validation project execution team for general guidance, resource and technical
planning. There are certain activities, which are not necessarily performed under the
purview of cleaning validation, are performed independently, but are very important for
designing the cleaning validation master plan and / or the cleaning validation protocols.
Few examples of such activities are analytical method development / validation / transfer,
cleaning method / procedure development, equipment qualification (IQ / OQ / PQ),
product development report, sampling procedure etc.
All such studies / documents are very much important and should become part of the
cleaning validation studies. Following figure-7, “Flow sequence of cleaning validation
activities”, provide an overview of all such activities and their integration with the primary
activity of cleaning validation. The figure also provides guidance on defining scope of
various different study protocols.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 26
RedLotus Pharmtech Private Limited

Figure – 7: Flow sequence of cleaning validation activities


Equipment OQ / PQ Activity Analytical Method Dev. / Validation Activity
Support
Activities for
the
Cleaning Process Mapping Recovery Studies
Validation Cleaning Procedures Analytical Method
Training AMV / AMT

Cleaning Validation Cleaning Cleaning Validation


Cleaning Validation Change Control
Master Plan (CVMP) Assessment Strategy

Residue & Cleaning Product Contact


Characterization Surface Area
Primary Design & Development Sampling locations &
Cleaning of Cleanability strategy
Validation
Activity Product list & details,
such as batch size,
dose / toxicity etc.

CVMP Cleaning Validation Protocol

CVMP Scope

RedLotus Guidance Document # GD001 & GD002, “SOP for Organization & Planning of
Qualification & Validation Studies” provides clear and specific instructions on planning,
execution and contents of the cleaning validation master plan.
7.11 Documentation for Cleaning Validation
To demonstrate that, the cleaning procedure developed for cleaning of the manufacturing
equipment is capable of reducing the potential residue or contaminants below the
maximum allowable residue limit. On the basis of the sections discussed earlier in this
document on selection of analytical test method, sampling method, sampling locations,
finalization of limits etc. should be done in the validation protocol.
Protocol will form an experimental plan to be followed to demonstrate the capability of
the cleaning procedure. The protocol should contain following heads but may not be
limited to this. However, RedLotus Guidance Document # GD001 & GD002, “SOP for
Organization & Planning of Qualification & Validation Studies” provides clear and specific
instructions on contents, preparation and execution of the cleaning validation protocol.
• Objective
• Scope
• Description of the process
• Identification of critical monitoring parameters (for the cleaning procedure)
• Test Functions (Tests to be utilized for validation of cleaning procedure)
• Sampling Procedures
• Analytical test methods
• Limits and Acceptance criteria
• Analytical Observation and results
• Conclusion
• Approval
Some heads such as sampling procedures, Analytical test methods and results either
should be written in the protocol or appropriately referred. The validation report should
be accompanied with the sampling verification, original / copy of the analytical test
reports or test data. Once the cleaning procedure is validated by following either of

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 27
RedLotus Pharmtech Private Limited

the approaches as described in sections 7.3.1 & 7.3.2, a comprehensive cleaning


validation report should be prepared and personnel involved in performing, sampling and
testing should be trained for the relevant operation and training records should be
maintained. RedLotus Guidance Document # GD001 & GD002, “SOP for Organization &
Planning of Qualification & Validation Studies” provides clear and specific instructions on
preparation and contents of the cleaning validation report.
Finally, typical steps as described in previous sections of this document necessary to be
followed in development of a cleaning validation for a particular product / equipment are
represented in the following figure-8.
Figure – 8: Typical steps in development of a cleaning validation program

A validated cleaning procedure is not


subject to change, if required change
must be implemented following a
change control process

7.12 Compliance and Re-validation


Once the cleaning method / process is qualified / validated it is very impor tant to ensure
that, same process is followed every time in each cleaning cycle. This should be done by
making sure that the cleaning process control parameters are locked and verified by the
user and QA after completion of each cleaning cycle.
In case of automatic cleaning processes, the process controller must be locked and
actual cleaning process control parameters on each cleaning cycle print-outs should

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 28
RedLotus Pharmtech Private Limited

be verified against the validated cleaning process control parameters. However, in case
of manual cleaning processes a detailed check-list must be prepared and actual cleaning
process control parameters must be recorded and verified against the validated cleaning
process control parameters for each cleaning cycle.
Deviations / failure observed, if any should be investigated as per the applicable
management procedure and if required the cleaning process should be improved and
(re)validated.
In order to ensure that, the cleaning process once validated remain in a state of control
a re-validation should be performed in one or more of the following conditions:
• Re-validation frequency defined in the cleaning validation protocol / report.
• Change in cleaning validation procedure.
• Introduction or deletion of a product from the original product list.
• Changes in product details, such as dosage, batch size.
• Change in equipment.
Decision tree in the following figure-9 provides guidance on the extent of re-validation
studies to be conducted in different circumstances.
Figure – 9: Decision Tree to Determine Re-Validation Requirements & Extent
Periodic Re-Validation & Failure / Repeated Deviation Initiated Change Initiated

Review, assessment & Change in:


evaluation of: (a) Introduction or deletion of a
(a) trend data of process control product from the original product list,
Change in cleaning procedure
values, (b) cleaning verification results, (b) changes in product details, such as
(c) trend of failure, OOS & OOT dosage, batch size, (c) change in
results equipment.

Rework / review the worst case


Write a review report based on validation matrix in changed / revised
assessment of data and trends Consistency in data? condition (Simulation of worst case
concluding that, the cleaning process Yes (Like CpK > 2 / no deviations / condition in revised / changed
continues to remain in a state of No OOS / OOT results) condition)
validation

No

Due to failure & based


Detailed Failure Investigation & Change results in a new
on the failure investigation & Yes
implementation of appropriate CAPA worst case?
associated CAPA?

Yes
No (Improvement) No

Abbreviated (re)validation:(a) on worst Write a review report based on


Complete & detailed (re)validation as assessment of revised information
case condition / sampling area or (b)
per the revised CVP concluding that, subject change falls
based on risk assessment
within the established worst case
brackets and does not require full (re)
validation

Cleaning Validation Report

8.0 ABBREVIATIONS AND DEFINITIONS


API Bulk Pharmaceutical Chemical
TOC Total Organic Carbon
CGMP Current Good Manufacturing Practice
CFR Code of Federal Regulation
PAI Pre-Approval Inspection
NDA New Drug Application

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 29
RedLotus Pharmtech Private Limited

ANDA Abbreviated New Drug Application


CIP Clean-In-Place
COP Clean-Out-of-Place
LOD Limit of Detection
LOQ Limit of Quantitation
HPLC High Performance Liquid Chromatography
TLC Thin Layer Chromatography
FT-IR Fourier Transform Infra-Red
NIR Near Infra-Red
ELISA Enzyme Linked Immunosorbent Assay
SDS-PAGE Sodium Dodecyl Polyacrylamide Gel Electrophoresis
ADI Acceptable Daily Intake
NOEL No Observed Effect Level
MSDS Material Safety Data Sheet
MAR Maximum Allowable Residue
IV Intravenous

9.0 APPENDIX
Appendix – 1: An example of a typical Cleaning Validation Master Plan
Appendix – 2: An example of a typical Cleaning Validation Protocol
Appendix – 3: An example of a typical Cleaning Validation Report
Appendix – 4: An example of a typical equipment Cleaning SOP & Cleaning Check -list
10.0 REFERENCES
• ICH Q7A, “Good manufacturing practices for Active Pharmaceutical Ingredient”.
2000.
• Code of Federal Regulations, Title 21, Part 211, Washington DC, US Government
printing office, “Current Good Manufacturing practices for finished
Pharmaceuticals”.
• US FDA, 1993,” Guide to Inspection of Validation of Cleaning Processes”.
• US FDA, 1994,” Guide to Inspection of Bulk Pharmaceutical Chemicals”.
• MHRA,” Rules and Guidance for Pharmaceutical Manufacturers and Distributors
2007”.
• “Validation of Bulk Pharmaceutical Chemicals”, By Daniel Harpaz & Ira R Berry,
Interpharm Press, Inc. Illinois, 1997.
• D. A. Le Blanc, “Rinse sampling for Cleaning Validation Studies”, Pharmaceutical
Technology, 22(5), 66-74, 1998.
• D. A. Le Blanc, “Establishing scientifically justified Acceptance Criteria for
Cleaning Validation of Finished Drug Products”, Pharmaceutical Technology, 136 -
148, October 1998.
• Active Pharmaceutical Ingredient Committee, “Guide to Cleaning Validation in API
Plants”, Sep. 1999
• José A. Morales Sánchez, “Equipment Cleaning Validation Within a Multi -Product
Manufacturing Facility”, BioPharm, Feb. 2006
• Pei Yang, Kim Burson, Debra Feder, and Fraser Macdonald, “Method
Development of Swab Sampling for Cleaning Validation of a Residual Active
Pharmaceutical Ingredient”, Pharmaceutical Engineering, Jan. 2005.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 30
RedLotus Pharmtech Private Limited

• “Cleaning Procedures as the FDA See It….”, International Journal of Generic


Drugs.
• “Cleaning Validation”, Health Sciences Regulatory Authority, Regulatory
Guideline, Dec. 2008
• Richard J. Forsyth, “Using Visible Residue Limits for Cleaning”, Pharmaceutical
Engineering, Jan. 2009
• “Acceptance Criteria and Limit Calculation”,
http://www.gmpua.com/World/GMPManual/daten/autorenteil/kapitel_08/08_e.htm
• Discussion and practical implementation of the requirements of ICH Guideline
Q7A, “Justification of Limits for Cleaning Validation in the Manufacture of Active
Pharmaceutical Ingredients”, GMP News, 14 May 2007
• Richard J. Forsyth, Vincent Van Nostrand, and Gregory P. Martin, “Visible-Residue
Limit for Cleaning Validation and its Potential Application in a Pharmaceutical
Research Facility”.

Guidance Document – Cleaning & Cleaning Validation


Document No – GD009 Ι Version – 00 Ι Date – 18 May 2018 31

You might also like