You are on page 1of 43

World Journal of Pharmaceutical Research

Mulik et al. SJIF Impact


World Journal of Pharmaceutical Factor 8.084
Research
Volume 8, Issue 13, 355-397. Review Article ISSN 2277– 7105

A FUNDAMENTAL REVIEW ON ‘CLEANING VALIDATION &


CLEANING PROCEDURE’

Dr. Ravindra B. Saudagar* and Mr. Santoshkumar R. Mulik

Department of Quality Assurance Techniques, R. G. Sapkal College of Pharmacy, Anjaneri,


Nashik – 422213, Maharashtra, India.

ABSTRACT
Article Received on
27 Sept. 2019, Cleaning Validation and Cleaning Procedure are two separate activities
Revised on 17 Oct. 2019, during implementation, but both are supportive to each other and in
Accepted on 07 Nov. 2019,
DOI: 10.20959/wjpr201913-15874
complete without others existence. Cleaning Validation and Cleaning
Procedure have the largest opportunity / probabilities to prevent patient
risk by assuring the no cross-contamination can occur. Ineffective
*Corresponding Author
Dr. Ravindra B. Saudagar cleaning can lead to adulterated product, which may be the traces of
Department of Quality previous product, cleaning agent &/or other extraneous material
Assurance Techniques, introduced &/or generated during the process. To establish the
R. G. Sapkal College of
validated cleaning procedure is becoming more and very important, as
Pharmacy, Anjaneri, Nashik
we deals with potent, complicated drug substances and complex
– 422213, Maharashtra,
India. biotechnology products. This article will over all the elements of
cleaning validation & main focus one the current gaps in cleaning
validation study & in establishment of cleaning procedures with respect to quantity used for
rinse, selection of batch size for cleaning validation & dirt cleaning study.

KEYWORDS: Validation, Cleaning Validation, Cleaning Procedures, Cleaning agent,


Pharmaceuticals Products, CIP.

1. INTRODUCTION
Cleaning validation: It is documented evidence with a high degree of assurance about
procedure that one can consistently clean a system or a piece of equipment or chain of
equipments to predetermined limit and at acceptable level.

The prime purpose of validating a cleaning process / procedure is to ensure product free from
cross-contamination, patient safety and compliance with regulations & GMP standards.

www.wjpr.net Vol 8, Issue 13, 2019. 1


Mulik et al. World Journal of Pharmaceutical Research

The most important benefit of conducting the validation work is to establish the cleaning
procedure that identify and correct the potential problems, Gaps in previously unsuspected,
which could compromise the safety, efficacy or quality drug product.

HISTORY
Cleaning validation has come a long way since the days of Barr Laboratories Court Case and
related to FDA guidelines. Subject of cleaning validation were published in 1991. At that
time, the requirements for cleaning validation barely filled a single page of the Bulk
Pharmaceutical Chemical and Biopharmaceutical guidance documents. Those documents
were then expanded to create the Guide to Inspection of Cleaning Validations by FDA (first
published in 1992 as a Mid-Atlantic Inspection Guidance, then reissued as an FDA guidance
document in 1993). GMP regulations have their basis in cleaning validation.

Beginning in 1906 with Upton Sinclair‟s “The Jungle,” the people demanded that the
government improve cleanliness practices in the processing of food giving rise to what we
know of today as the cGMPs for both food and drugs. While cleaning has always been part of
the GMP regulations. The GMPs that we follow today were predominantly written in 1978.

References to cleaning and related documentation associated with cleaning can be found
throughout. As with many other areas of validation, however, there is no explicit reference to
cleaning as a process to be validated. The GMPs that was challenged in the Barr Laboratories
court case. In that decision, Judge Wolin ruled that cleaning did require treatment as a
process and therefore required validation.

In 1996 proposed revisions to the GMPs were drafted by the FDA; although not adopted,
these revisions proposed to re-define the manufacturing process as beginning with a cleaning
operation.

When the FDA published “Pharmaceutical cGMPs for the 21st Century: A Risk-Based
Approach” in August of 2002, and reported on their progress in September 2004 and
validation was reinforced in pharmaceutical manufacturing. Although risk-based decision-
making in the establishment of scientific rationales was always a cornerstone of cleaning
validation requirements, efforts have been renewed to ensure the incorporation of risk
analysis documentation in cleaning programs.

www.wjpr.net Vol 8, Issue 13, 2019. 2


Mulik et al. World Journal of Pharmaceutical Research

OBJECTIVE
The objective of the cleaning validation is to verify the effectiveness of the cleaning
procedure for removal of product residues, degradation products, preservatives, Excipients, or
cleaning agents as well as the control of potential microbial contaminants. It is necessary to
Validate Cleaning procedures for the following reasons:
 Pharmaceutical products and API can be contaminated by other pharmaceutical products,
cleaning agent & microbial contamination.
 It is regulatory requirement in pharmaceutical product manufacture the concern is the
same-assurance that equipment is clean and that product quality and safety are
maintained.
 It assure as an internal control and compliance in view of quality at manufacturer end.
 To protect product integrity
 To reuse the equipment
 It will save cost due to rejection of contaminated product & regulatory non-compliance.

Why Cleaning Validation Required


To verify the effectiveness of cleaning procedures and to ensure no risks are associated with
cross contamination of active ingredient or cleaning aids.

Where Cleaning Validation Needed


 Initial qualification of process/equipment.
 Critical change in a cleaning procedure.
 Critical change in formulation.
 Significant change in formulation.
 Change in a cleaning process.
 Change in a cleaning agent/aids.
 Modification in equipment / chain of equipment of system.

When Cleaning Validation Required to Do:


1. Initial qualification of process/ equipment.
2. Critical change in a cleaning procedure.
3. Critical change in formulation.
4. Significant change in formulation.
5. Change in a cleaning process.

www.wjpr.net Vol 8, Issue 13, 2019. 3


Mulik et al. World Journal of Pharmaceutical Research

6. Change in a cleaning agent

Advantages of Cleaning Validation


1. Safety: Validation can also result in increased operator safety. Properly calibrated,
validated instruments and gauge used to reduce accident and results in safety.

2. Better Customer quality: Through proper validation, market recall is avoided which
results in better customer care and quality of the product.

3. Contamination & Cross Contamination


Generally cross contamination and contamination by a foreign material are two types of
contamination. Cross contamination is usually through an active ingredient from one product
carrying over into subsequent manufactured product. However, carryover of other product
component such as Excipients can also be problematic and may degrade and final quality of
product. Contamination of one batch of product with significant level of residual active
ingredient from a previous batch may pose obvious problem to consumer or patients from
unintended contaminants.

Potential clinically significant synergistic interaction between pharmacologically active


chemical is a real concern. Inert ingredients used in drug product are generally recognized as
safe for human consumption and for routine use also. Maintenance and cleaning of equipment
provide the potential for contamination with items such as equipment parts and lubricant.
Chemical cleaning agent and piece of cleaning tools can cause problems ranging from poor
pharmaceutical elegance to exceeding acceptable levels of particulate matter in parenteral
products to inadvertent inclusion of toxic compounds in the product. In addition, some
activities are adversely affected by trace contaminants and may exhibit change in stability or
bioavailability if exposed to such contamination.

The second type of contamination is by foreign material these may be bacterial in nature or
could represent part of the equipment. Maintenance, cleaning, and storage condition may
provide adventitious microorganisms with the opportunity to proliferate with in processing
equipment. This could pose obvious problems for sterile products manufacture (generation of
high level of pyrogens, decreasing the assurance of sterile achieved by equipment
sterilization procedures etc.) It also possess serious problem for the manufacture on non-
sterile dosage form particularly unpreserved products which support microbial growth.

www.wjpr.net Vol 8, Issue 13, 2019. 4


Mulik et al. World Journal of Pharmaceutical Research

Mechanism of Contamination
1. Cross contamination with active ingredient
One of the real dangers in cross contamination of active ingredients is that by being
contaminated results in a multiple ingredient product instead of single active ingredient.
Depending on medical effects, the contamination may enhance the action or negate the action
or contaminant may have an entirely different medical effects.
2. Microbiological contamination: This form of contamination is particularly insidious
because the contamination may develop at any time, even after cleaning. A major
contributing factor is the storage of equipment in a wet condition. This provides a natural
medium in which bacteria can grow.
3. Contamination by cleaning or sanitizing agents: Some pharmaceutical operations may
find it necessary to use fairly toxic materials for cleaning purpose for stubborn residues. This
is particularly true in the manufacture of active pharmaceutical ingredients (APIs). As such,
these materials represent a potential threat as contaminants. It seems obvious that effective
way of dealing with this potential problem is to use cleaning agents with the lowest toxicity
that will still be effective in removing the residue in the given cleaning situation. The same
factors also apply to sanitizing agents used to wipe down cleaned equipment.
4. Contamination by miscellaneous other materials: In addition to the usual expected or
anticipated list of potential contamination in a pharmaceutical operation, many other less
likely materials can also contaminate products. A partial list includes equipment parts such as
excipients, bristles from brushes used in packaging filling equipment, paper filters, micron
filters, fibers and rubber particles from gloves, cleaning aids such as brush bristles, cloth, and
cotton fibers from rags and wiping materials, lubricants.

1. The nature of the potential contaminants (toxicity, solubility etc)


The CEFIC-APIC guide to cleaning validation recommends three levels of cleaning that may
be implemented. This approach is outlined in the table below, however it should be
mentioned that additional levels might be necessary depending on the nature of the process
and requirement.

Level Thorough of Cleaning Cleaning validation


Carryover of the previous product is critical.
2 Cleaning required until predetermined stringent Essential
carryover limits are met.
Carryover of the previous product is less critical. Increase from not required
1
Cleaning should reduce the potential carryover to a to necessary (Lower

www.wjpr.net Vol 8, Issue 13, 2019. 5


Mulik et al. World Journal of Pharmaceutical Research

less stringent limit as required for level 2. acceptable carryover limits)


Only gross cleaning if carryover of the previous
0 Not required
product is not critical

Essential Programs that maintain the validated state and their required elements
In case, introduction of new parts or repaired or modification in equipment, after
completion of cleaning validation; monitor & test /challenge the respective pre-validated
cleaning procedure.
initiatives on site that maintain quality and will affect the
company‟s ability to maintain in the validated state.

entive maintenance

and Cleaning Validation


especially for manual cleaning operations)
Equipment cleaning and use logs
Equipment sampling procedures for cleaning assessments (e.g., swab, rinse, etc.).
Visual inspection requirements for cleaned equipment

Equipment Storage area / environment & Storage period

Level of Cleaning: The level or degree of cleaning and validation required for the
manufacturing process of drug substances mainly depends on:
equipment or not)
intermediate or final step)
(solubility, toxicity, categories, physiochemical
properties of molecule to be handled etc.)
Equipment Storage Period or in-use period / campaign.

In case of Drug Products: Different cleaning situation may arise during the manufacturing
of drug products, such as;
a. Batch-to-Batch changeover cleaning.
b. Product-to-Product changeover cleaning

www.wjpr.net Vol 8, Issue 13, 2019. 6


Mulik et al. World Journal of Pharmaceutical Research

c. Over the number of batches in manufactured in single campaign


d. Over the period of storage of equipment

In case of non-dedicated drug product manufacturing facility, different cleaning procedures


may exist depending on the manufacturing step and nature of the next manufacturing step to
be followed in the same equipment. This results in two different levels of cleaning as
explained below.

Level-1 Cleaning: [Type-A]


Cleaning procedure followed between the manufacturing of different batches of the same
product.
Example – In a manufacturing Campaign for Product-A, there are 3 Batches to be
manufactured as shown below.
Batch-1, Batch-2, Batch-3
For a given equipment &/or equipment train, if Batch-1 in the campaign is to be followed by
Batch-2 in the campaign, then Level-1 cleaning is required.
Also in case of different strength of same molecule / same API formulation with same
preparation with lower to higher change over.

Level-2 Cleaning: [Type-B]


Cleaning procedure used between the manufacturing of Batches of different Product and / or
at the end of manufacturing campaign even if same product is planned for the next campaign
and / or new / modified / out of service equipment to be used for product.

Different Product: It can be the difference in term of API used, Difference in formulation
with different in-active materials / proportion / different strength of same molecule. i. e.
higher to lower change over.

Level-3 Cleaning: [Type-C]


Cleaning procedure to be used, after validity of ' Clean-Storage-Period' of the clean
equipment stored in controlled area, before usage to product-process.

The above three degree or level of cleaning differs from each other in terms of the degree of
risk associated with it, acceptance limit, degree of cleaning & method of verification of the
cleaning process.

www.wjpr.net Vol 8, Issue 13, 2019. 7


Mulik et al. World Journal of Pharmaceutical Research

Comparison between Cleaning Levels.


Factor Level - 1 Level - 2 Level - 3
Risk Lowest Highest Lowest
Acceptance
Highest Lowest Highest
Limit
Degree of Less More
Moderate
Cleaning Extensive Extensive
Verification of Visual Analytical Visual
Cleaning Inspection Testing Inspection

In case of Drug Substance


Different cleaning situation may arise during the manufacturing of drug products, such as:
-to-Batch changeover cleaning
intermediate of same product.
product to intermediate of another product.
product to final stage of another product.
another final product
-dedicated drug substance manufacture.

Figure No. 1: Cleaning validation process flow.

www.wjpr.net Vol 8, Issue 13, 2019. 8


Mulik et al. World Journal of Pharmaceutical Research

Figure No. 2: Cleaning Procedure.

CLEANING VALIDATON MASTER PLAN


Master Plan should.
site/facility/area that is governed by the Master Plan
An overview of the typical manufacturing process to be performed for the product in area.
cleaning that are to be used (e.g., automated Clean-In-
Place or Clean-Out of-Place, semi-automated cleaning or manual cleaning)
various departments having a role in cleaning validation
activities
the cleaning validation program, including:

www.wjpr.net Vol 8, Issue 13, 2019. 9


Mulik et al. World Journal of Pharmaceutical Research

Elements of Cleaning Validation


1. Residue selection
2. Equipment characterization
3. Cleaning agent selection
4. Limits calculation
5. Product grouping
6. Equipment grouping
7. Cleaning procedure
8. Sampling
9. Analytical methods
10. Validation protocol
11. Validation report
12. Changes & change control procedures

Potential Residues
a. Precursors of the drug substance.
b. By-products and/or degradation products of the drug substance.
c. Product from previous batch.
d. Solvents and other Excipients employed during manufacturing process.
e. Microorganism
f. Cleaning agents and lubricants.

Residue Identification: When performing cleaning validation there are a number of residues
that must be considered:
1. API
2. Constituents of the cleaning agent
3. Preservatives
4. Precursors or starting materials
5. Intermediates
6. Processing aids
7. Media
8. Buffer
9. Cellular debris or metabolites
10. Particulate

www.wjpr.net Vol 8, Issue 13, 2019. 10


Mulik et al. World Journal of Pharmaceutical Research

11. Bioburden
12. Endotoxin
13. Viral particles
14. TSE
15. Excipients
16. Colorants, dyes, flavors or fragrances
17. And many more.

If we have the advantage of using a nonspecific method for cleaning assessment (e.g., TOC,
pH, conductivity), we may be able to use a single analytical method to look for all (or most)
types of residues. In yet other instances, it is desirable to use a specific analytical method
(e.g., HPLC IMS, UV, GC and FTIR), which, by definition, requires that we select the
residue(s) of interest to the cleaning validation.

Sampling and Testing Methodologies


This section should typically include a step-by-step explanation of sampling techniques and
requirements, as well as the specific analytical procedures to be used in the analysis of those
samples. It should specify which laboratories are to be involved in the testing and any
precautions to be taken throughout the validation exercise.

A visual check should be incorporated into the cleaning assessment. The sampling technique
chosen to evaluate the effectiveness of the cleaning procedure should be swabbing, the fluid
rinse of samples, or a combination of both methods. The following sampling methods provide
various levels of assurance concerning cleaning:

• Visual inspection
✓Active product contact parts of the equipment are individually examined (wherever
possible) for cleanliness. This visual inspection allows the early localization and
identification of any inadequacies in the cleaning procedure.
✓ Qualitative – dependent upon inspector and item sampled.
✓ Subjective – dependent upon inspector and item sampled.

• Rinse water sampling and analysis

www.wjpr.net Vol 8, Issue 13, 2019. 11


Mulik et al. World Journal of Pharmaceutical Research

✓ According to 2004 FDA “Guide to Inspections Validation of Cleaning Processes:” “Two


advantages of using rinse samples are that a larger surface area may be sampled, and
inaccessible systems or ones that cannot be routinely disassembled can be sampled and
evaluated.”
✓ Analysis can be quantitative, using pH, conductivity, particle count, microbial count, Total
Organic Carbon (TOC) determination, spectrophotometry, bioassays, or limulus amebocyte
lysate for pyrogens.
✓ Recovery factor is uncertain; it involves dilution

• Surface sampling and analysis


✓ Removes adherent materials.
✓ Analysis can be quantitative.
✓ Precise definition of the area sampled is required.

• Surface sampling from coupons


✓ Quantitative.
✓ Depends on whether coupons are equivalent to the surface of interest.
✓ Requires removing coupons from the system.

• Method Selection
Whenever possible, each piece of equipment should be dismantled into its individual
components after cleaning and each part should be individually tested for cleanliness. In this
manner, any inadequacies in the cleaning process will be more readily identified and
localized.

It may not be practical or desirable to dismantle large or Clean–In–Place (CIP) equipment.


Regardless, validation sampling and testing should commence as soon as possible after the
cleaning process is complete to reduce the chance for contamination by outside sources.

Equipment that has just been cleaned should be covered immediately by appropriate means to
protect it from any contamination.

www.wjpr.net Vol 8, Issue 13, 2019. 12


Mulik et al. World Journal of Pharmaceutical Research

• Solvents
Aqueous or organic solvents used in the cleaning procedure, should be sufficient to remove
residues, and at the same time, should be minimized to reduce the risk of reaction with or
damage to the equipment, or the over-dilution of the residue and the resultant loss of
analytical sensitivity.

Samples should be collected in clean or sterile containers. Sterile containers are suitable for
this intended use. All validation samples must be properly labeled with complete information
regarding the source of the sample, sampler‟s name, sampling date, reference number,
product name, and the part of equipment from which the sample has been collected.

A sample of the rinse or swabbing solvent should always be included with the actual test
samples to serve as a reagent blank for any chemical or microbiological determination when
required.

All types of samples, physical, chemical, or microbiological, should be collected according to


a written procedure and using techniques, reagents, equipment, and containers appropriate to
the type of testing to be performed. Only trained personnel should perform the collection of
these samples.

The environmental effectiveness of cleaning procedures should be assessed by surface


sampling of non-product contact surfaces (e.g.: floors, walls, air ducts, exterior equipment
surfaces, etc.) Samples should be collected and analyzed for potential contamination.

• Sampling Methods
The sampling method selection for cleaners, involves choosing between rinse water
sampling, swabbing surfaces, coupon sampling, or placebo sampling. Rinse water sampling
involves taking a sample of an equilibrated post-final rinse that has been re-circulated over all
surfaces. Rinse samples should be correlated to a direct measuring technique such as
swabbing.

Swabbing involves using a wipe or swab that is moistened with high purity water, such as
Water-for-Injection (WFI) that is typically wiped over a defined area in a systematic multi-
pass way always going from clean to dirty areas to avoid recontamination (e.g.: 10 cm side by
side strokes vertically, 10 cm horizontally, and 10 cm each with the flip side of the swab in
each diagonal direction). For TOC analysis, very clean swabs or wipes and sample vials

www.wjpr.net Vol 8, Issue 13, 2019. 13


Mulik et al. World Journal of Pharmaceutical Research

should be used. (All of these are commercially available). The amount of residue is known to
be uniformly distributed on the smooth surfaces of equipment parts. Also, the most difficult
to clean or “worst-case” areas of the equipment should be identified and specifically targeted
for sampling whenever possible.

Residue Detection
Selecting a method to detect cleaner residues can involve specific methods for specific
cleaner ingredients such as: High Performance Liquid Chromatography (HPLC), ion selective
electrodes, flame photometry, derivative Ultra Violet (UV) spectroscopy, Thin Layer
Chromatography, enzymatic detection, and titration. It can also involve non-specific methods
that detect the presence of a blend of ingredients such as: TOC, pH, and conductivity. The
FDA prefers specific methods, but will accept non-specific methods with adequate rationales
for their use. For investigations of failures or action levels, a specific method is usually
preferable.

• Analytical Evaluation
Analytical validation of the cleaning procedure should be performed after the approval of
visual inspection (absence of stains or any materiel residue). The specificity, sensitivity, and
percentage of recovery of the test method should be adequate to meet acceptance criteria.

For the swab method it may be necessary to determine


✓ The percentage recovery of the swab extraction procedure.
✓ The effectiveness of the swab at recovering residues from equipment parts surface.
✓ The interference of swab materials in the analysis.

For the rinse solution method it may be necessary to determine


✓ The percentage recovery of the rinse solution extraction procedure.
✓ The effectiveness of the rinse solution at recovering residues from equipment parts
surfaces.
✓ The interference of the rinse solution in the cleaning procedure and analysis.
✓ A correction for recovery efficiency in calculations for acceptable residue levels.
Percentage Recovery = 100 x Sample Concentration /Standard Concentration

www.wjpr.net Vol 8, Issue 13, 2019. 14


Mulik et al. World Journal of Pharmaceutical Research

The percentage recovery is important because it will be applied when evaluating the final
residual concentration according to the relation:

Percentage of actual amount of residual = Calculated Amount x Percentage Recovery

It is very difficult to establish acceptable fixed limits for recovery percentage due to the
individual difference in solubility of residues, the solvent used, and the nature of the
manufacturing surfaces.

The following three factors contribute to the difficulty of establishing fixed limits for the
recovery percentage.
1. The residues behavior toward the solvent used.
2. The solvent used.
3. The nature of the manufacturing surfaces.

Some products such as proteins, for example, have a very low solubility, so the percentage
recovery may be as low as 10–20%. For soluble residue, a higher percentage recovery should
be expected. In general we can expect an ideal percentage recovery that falls between 60%
and 90%. It is very important to continuously develop the sampling and swabbing methods
and reproducibility to improve percentage recovery values.

• Microbiological Cleaning Considerations: All equipment that comes in contact directly


with raw material intermediate as well as final product - must be considered for inclusion,
because of its potential to act as a possible source for microbiological contamination. In
addition, the facilities must be considered for the level of microbiological contamination
appropriate to the area classification.

Microbiological samples should be collected prior to and throughout the cleaning procedure
to assist in selection and confirmation of the efficacy of disinfectants and detergents.
Microbiological cleanliness is assessed as < 200 cfu / 100 cm2 for non-sterile production.

It is important to determine the type of organism present. It is necessary to demonstrate the


absence of pollution indicator organisms such as, Escherichia coli, Salmonella spp and
Pseudomonas aeroginosa, from all locations monitored. It is necessary, as well, to ensure that
high levels of other microbial flora do not mask these organisms.

www.wjpr.net Vol 8, Issue 13, 2019. 15


Mulik et al. World Journal of Pharmaceutical Research

Within sterile production, attention must be paid to the number of organisms present rather
than their type. The level of microbiological contamination of the rinse water should be 10
cfu / 100 ml. Sampling is repeated three times during the validation.

• Worst-Case Determination
Worst-case determination of cleaning validation is a crucial step in defining contamination
limits and in cleaning procedure efficacy. A worst-case determination study should be based
on: active product solubility; active product toxicity; smallest batch size that can be
manufactured using the equipment concerned; the maximum daily dose of this product; the
number of dosages that can be made from next batch (contaminated); the product in its
largest available tables mass, or in case of ampoules or vials, the largest available filling
volume, and in both cases, the highest daily dose; the total area with which the product comes
into contact; the area of one tablet or the volume of one individual fill; and the total amount
of residual contaminant.

After completing the worst-case determination table, we can easily identify the product
representing this case (A, B, or C). The table should list all products to be manufactured in
the same equipment whatever the chemical and bioactivity types of actives.

Equipment characterization
Cleaning validation involves not only the removal of residues but also gives assurance that
each and every piece of equipment associated with the process has been cleaned to acceptable
levels. It is typically referred as train based approach. The equipment train is series of
equipments, through which the product or products move as they progress through the
manufacturing process. In order to asses that the equipment is cleanable or not; it should be
characterized in such a way that its design features are well known. Equipment
characterization can assist cleaning validation initiatives in many ways.
procedure by identifying cleaning challenges and
ensuring that they are addressed in the cleaning methods employed.
identifying hard to clean locations and high risk locations in equipment for the purpose of
sampling site selection.
Materials of construction of targeted equipment‟s that will be included in sampling
recovery studies and those that will not be included.
at the end of a production process and/or will be
dedicated to a single product.

www.wjpr.net Vol 8, Issue 13, 2019. 16


Mulik et al. World Journal of Pharmaceutical Research

are compatible with the selected cleaning agents


and temperature that will be used with the cleaning process.
surface areas for the purpose of calculating limits
and results.
and use of process equipment for the purpose of
grouping that equipment.

Equipment characterization: Cleaning validation involves not only the removal of residues
but also gives assurance that each and every piece of equipment associated with the process
has been cleaned to acceptable levels. It is typically referred as train based approach. The
equipment train is series of equipment through which the product or products move as they
progress through the manufacturing process. In order to asses that the equipment is cleanable
or not it should be characterized in such a way that its design features are well known.

Equipment characterization can assist cleaning validation initiatives in many ways:


1. Promote more effective cleaning procedure by identifying cleaning challenges and
ensuring that they are addressed in the cleaning methods employed.
2. Identifying hard to clean locations and high risk locations in equipment for the purpose of
sampling site selection.
3. Target materials of construction that will be included in sampling recovery studies and
those that will not be included.
4. Isolate materials that will be disposed of at the end of a production process and/or will be
dedicated to a single product.
5. Verify that all materials of construction are compatible with the selected cleaning agents
and temperature that will be used with the cleaning process.
6. Collect product contact and sample site surface areas for the purpose of calculating limits
and results.
7. Confirm similar geometries, capacities, and use of process equipment for the purpose of
grouping that equipment

Note: When performing correctly, equipment characterization is the process whereby it


catalogues the features and attributes of equipment, thereby ensuring that equipment can be
cleaned reliably and reproducibly.

www.wjpr.net Vol 8, Issue 13, 2019. 17


Mulik et al. World Journal of Pharmaceutical Research

Cleaning Solvent Selection


Solubility of the API: Product having least solubility in water and higher strength can be
considered as worst case product on basis of solubility
Appropriate quantities of Solvent by volume
Group Included descriptive terms
for 1 part of solute by weight
Very soluble Less than 1 Part
1
Freely soluble From 1 to 10 Parts
Soluble From 10 to 30 parts
2
Sparingly soluble From 30 to 100 Parts
Slightly soluble From 100 to 1000 parts
3 Very slightly soluble From 1000 to 10000 parts
Practically insoluble More than 10000 Parts

Cleaning Agent Selection: All cleaning processes rely on the principle of TACT and WINS.

TACT
Time, Action, Concentration/Chemistry & Temperature or TACT are the process parameters
that are required to be controlled in any cleaning process, whether manual, semi-automated or
automated. Changes in one TACT parameter will cause a commensurate increase or decrease
in the other parameters.

In all cases, however, the correct balancing of the TACT parameters. Requires proper
knowledge and understanding of WINS:

WINS
Water, Individual, Nature of the Soil / solvent, Surface. WINS represents the parameters that
affect the soil‟s removal from the surface and each parameter can affect your ability to apply
TACT in a given situation. Cleaning chemistries fall into several broad categories.
Water
Solvents
Commodity chemicals
Formulated cleaning agents

Current approaches in determining the acceptance limits for cleaning validation.


Below different type of approaches can be used in combinations with each other suitable for
better assurance of cleaning.

www.wjpr.net Vol 8, Issue 13, 2019. 18


Mulik et al. World Journal of Pharmaceutical Research

Approach - I (Dose Criteria)


Not more than 0.001 mg/day of minimum daily dose of any Product-A will appear in the
maximum daily dose of another Product-B.

Milligrams of active ingredient =


=I x K x M of Product- A permitted per J x L 4 inch2 swab area
I = 0.001 of the smallest strength of Product-A manufactured per day expressed as mg/day
and based on the number of milligrams of active ingredient.
J = Maximum number of dosage units of Product-B per day
K = Number of dosage units per batch of final mixture of Product-B
L = Common equipment surface area between Product-A & Product-B expressed as sq.
inches.
M= 4 inch2/swab.

2. Approach - II (10 ppm Criteria)


Any active ingredient can be present in a subsequently manufactured product at a maximum
level of 10 ppm.
Milligrams of active ingredient = R x S x M of Product - A permitted per L 4 inch2 swab
area.
R = 10mg active ingredient of Product - A in one kg of Product - B
S = Number of kilograms per batch of final mixture of Product - B
L = Common equipment surface area between Product-A & Product-B expressed as sq.
inches.
M = 4 inch2/swab.

3. Approach - III (Visually Clean Criteria)


No quantity of residue should be visible on the equipment after cleaning procedures are
performed.

Conditions to Apply Approach - III


 Validated, reproducible cleaning procedure / method.
 Equipment / all surface area of equipment shall be visible at suitable light intensity.
 Description of product residual / cleaning agent residues should be of different physical
characteristic & easily identifiable, distinguishable by normal eye sight from equipment
surfaces that to be clean.

www.wjpr.net Vol 8, Issue 13, 2019. 19


Mulik et al. World Journal of Pharmaceutical Research

 Riboflavin - UV challenge test can be adopted to ensure the assurance / effectiveness of


cleaning by visible method.

3. Approach - IV
(Physico-Chemical Test Criteria)
 Rinse Clarity & Colour / Description with respect to cleaning solvent used.
 Rinse Visual & Sub - Visual particulate load.
 Chemical Trace analysis of materials / content used in previous product
 Chemical Trace analysis of cleaning against or / & solvent used for cleaning
 Chemical Trace analysis of decomposed chemicals / materials / impurities during
cleaning process.
 Physical Test like TOC, Conductivity, pH etc. / cleaning.

Grouping of Equipment
All equipment must be:
 Used to produce products from the same product group i.e. different strengths, slight
variation in in-put material proportion or difference few in-active materials.
 Cleaned with the same cleaning agent
 Cleaned with the same cleaning method
 Equivalent in terms of position or role in the manufacturing process
 Similar functionality with similar design (e.g., different size of vessels with similar
shape.)

Product Grouping and Equipment Grouping


It is a method by which products or equipment is considered to be similar or equivalent for
the purpose of cleaning validation. When considering similar, a worst case area of the
instrument or Site is selected for demonstrating cleaning validation. When considering
equivalent, any area of the instrument or site may be selected as representative of any other
area of the instrument or site. Bracketing, a term that appear in EU GMP Annex on cleaning
validation, has an equivalent meaning to grouping, although it may include an added burden
for testing the extremes of population. Grouping may be used to simply prioritize cleaning
validation studies or may be used to eliminate some of the numerous possible combinations
of product and equipment studies that might otherwise need to be performed.

www.wjpr.net Vol 8, Issue 13, 2019. 20


Mulik et al. World Journal of Pharmaceutical Research

Product Grouping and Equipment Grouping


Grouping, sometimes also called as a family approach. It is a method by which products or
equipment is considered to be similar or equivalent for the purpose of cleaning validation.
When considering similar, a worst case member of the family is selected for demonstrating
cleaning validation. When considering equivalent, any member of the family may be selected
as representative of any other member.

Bracketing, a term that appear in EU GMP Annex on cleaning validation, has an equivalent
meaning to grouping, although it may include an added burden for testing the extremes of
population.

Grouping may also be used to simply prioritize cleaning validation studies or may be used to
eliminate some of the numerous possible combinations of product and equipment studies that
might otherwise need to perform.

When grouping products, all products must be


1. Manufacture on the same equipment group.
2. Cleaned with the same cleaning agent.
3. Cleaned with the same cleaning procedure.

Grouping considerations for products include


1. Similar patient risk levels (e.g. therapeutic indication, patient population route of
administration, potency, toxicity for drugs/devices/ nutraceuticals/ cosmetics or in case of in-
vitro diagnostics used, such as so called health and safety products)
2. Similar formulations.
3. Similar manufacturing process.

Cleaning validation must always be carried out to meet lowest limit of the entire product
group.
When grouping equipment, all equipment must be
1. Used to produce products from the same product group.
2. Cleaned with the same cleaning agent.
3. Cleaned with the same cleaning method.

www.wjpr.net Vol 8, Issue 13, 2019. 21


Mulik et al. World Journal of Pharmaceutical Research

Grouping considerations for equipment include


1. Equivalent in terms of position or role in the manufacturing process.
2. Similar functionality.
3. Similar design.

Grouping for products


All products must be
ufactured on the same equipment group cleaned with the same cleaning agent
procedure

Grouping considerations for products include.


Similar patient risk levels (e.g., therapeutic indication, potency, toxicity for drugs/devices /
neutraceuticals/ cosmetics).
Similar formulations
Similar manufacturing processes cleaning validation must always be carried out to meet
the lowest limit of the entire product group.
Geometry, materials of construction, capacity.

Worst Case Rating


 Solubility in subjected solvent
 Maximum Toxicity
 Minimum Therapeutic Dose / Potent Drug
 Difficult to Clean
 MACO: Maximum carryover of previous product.
 Maximum batch size in Kg or Liter of worst selected product to be identify to execute the
cleaning validation study, in which maximum active ingredients handles for maximum
run time.
 PDE : Permitted Daily exposure calculated based on NOAEL(No-Observed-Adverse-
Effect Level), OEL (Occupational Exposure Limit) , ADE [Accepted Daily Dose] , TTC
(Threshold of Toxicological Concerns) & LD50 (Median Lethal Dose) .
 Lowest Limit based on therapeutic dose / toxic data, batch sizes, surface areas, etc

www.wjpr.net Vol 8, Issue 13, 2019. 22


Mulik et al. World Journal of Pharmaceutical Research

Acceptance Criteria
Companies must demonstrate during validation that the cleaning procedure routinely
employed for a piece of equipment limits potential carryover to an acceptable level. That
limit established must be calculated based on sound scientific rational.

Methods of Calculating Acceptance Criteria


1. Based on Therapeutic Daily Dose
The principle for the requirement is that the standard Therapeutic Daily Dose (TDD) of the
following substance („contaminated‟ substance, in this case called "next") may be
contaminated by no more than a certain proportion (usually 1/1000 part) of the TDD of the
substance investigated in the cleaning validation (contaminating substance, in this case called
"previous"). This method only applies when the therapeutic daily dose is known. It is
generally used for final product changeover API Process “A” to API Process “B.

Procedure
Establish the limit for Maximum Allowable carryover (MACO) according to the following
equation.
TDD Previous X MBS
MACO = -------------------------------
SF x TDD next

MACO: Maximum Allowable Carryover: Acceptable transferred amount from the


investigated product ("previous").

TDD previous
Standard therapeutic dose of the investigated product (in the same dosage form as TDD next)

TDD next
Standard therapeutic dose of the daily dose for the next product.

MBS: Minimum batch size for the next product(s)


(Where MACO can end up)

SF: Safety factor (normally 1000 is used in calculations based on TDD next)

www.wjpr.net Vol 8, Issue 13, 2019. 23


Mulik et al. World Journal of Pharmaceutical Research

Example 1
Product - A will be cleaned out. The product has a standard daily dose of 10 mg and the batch
size is 200 kg. The next Product-B has standard a daily dose of 250 mg and the batch size is
50 kg. Both A and B are administrated orally and SF is set to 1000. Calculate the MACO for
A in B.
250 (mg) x 50000000 (mg)
MACO = ------------------------------------- = 2000(mg)
1000 x 250 (mg)

Result: MACO is 2 g (2000 mg)

Example 2
Now Product-B in example 1 will be cleaned out. The following product is product A in
example 1.
Calculate the MACO for B in A.
250 (mg) x 200000000 (mg)
MACO = ------------------------------------- = 5000000(mg)
1000 x 10 (mg)

Result: MACO is 5 kg (5 000 000 mg)


In API manufacture it is possible to obtain a very high MACO figure. In example 2, the
figure obtained is clearly unacceptable. Although there would be no effects expected, the
equipment would be obviously dirty and a general GMP limit should be chosen.

Instead of calculating each potential product change situation, the worst case scenario can be
chosen. Then a case with most active API (lowest TDD) is chosen to end up in the following
API with the smallest ratio of batch size divided with TDD (MBS/TDD ratio). This could be
done if the safety factor is the same for all products (otherwise the lowest MBS/(TDD x SF)
ratio should be chosen).

2. Based on Toxicological Data


In cases in which a therapeutic dose is not known (e.g. for intermediates and detergents),
toxicity data may be used for calculating MACO.

Procedure
Calculate the so called NOEL number (No Observable Effect Level) according to the
following equation and use the result for the establishment of MACO.

www.wjpr.net Vol 8, Issue 13, 2019. 24


Mulik et al. World Journal of Pharmaceutical Research

LD 50 g/Kg x 70 (Kg a person)


NOEL = --------------------------------------------
2000

From the NOEL number a MACO can then be calculated according to:
NOEL x MBS
MACO = --------------------------------------------
SF x TDD next

MACO: Maximum Allowable Carryover: acceptable transferred amount from the


investigated product ("previous")

NOEL: No Observed Effect Level.


LD 50: Lethal Dose 50 in g/kg animal. The identification of the animal (mouse, rat etc.) and
the way of entry (IV, oral etc.) is important.
70 kg: 70 kg is the weight of an average adult
2000: 2000 is an empirical constant
TDD next: Largest normal daily dose for the next product
MBS: Minimum batch size for the next product(s)
(Where MACO can end up)

SF: Safety factor


The safety factor (SF) varies depending on the route of administration. Generally a factor of
200 is employed when manufacturing APIs to be administered in oral dosage forms. SF can
vary depending on substance/dosage form according to (suppose tox values from oral
administration) as for example as presented on the next page.

Safety factors
Topical : 10 – 100
Oral products: 100 – 1000
Parenteral : 1000 – 10 000

Remarks: API`s in development may require higher safety factors due to lack of knowledge.

Calculation of MACO values from toxicological data is frequently done when therapeutic
dosage data is not available or not relevant. It is generally employed if the previous product is
an intermediate and the following product an API.

www.wjpr.net Vol 8, Issue 13, 2019. 25


Mulik et al. World Journal of Pharmaceutical Research

3. General Limit
If the calculation methods based on therapeutic doses or toxicological data result in
unacceptably high or irrelevant carryover figures, or toxicological data for intermediates are
not known, the approach of a general limit may be suitable. Companies may chose to have
such an upper limit as a policy. The general limit is often set as an upper limit for the
maximum concentration (MAXCONC) of a contaminating substance in a subsequent batch.

The concentration (CONC) of the investigated substance which can be accepted in the next
batch according to dose related calculations, is.

MACO
CONC = ------------------------------
MBS

MACO: Maximum Allowable Carryover: acceptable transferred amount from the


investigated product ("previous"). Calculated from therapeutic doses.

MACO ppm
Maximum Allowable Carryover: acceptable transferred amount from the investigated product
("previous"). Calculated from general ppm limit.

CONC
Concentration (kg/kg or ppm) of "previous" substance in the next batch. Based on MACO
calculated from therapeutic doses and/or tox data.

MAXCONC
General limit for maximum allowed concentration (kg/kg or ppm) of "previous" substance in
the next batch.

MBS
Minimum batch size for the next product(s) (where MACO can end up).

A general upper limit for the maximum concentration of a contaminating substance in a


subsequent batch (MAXCONC) is often set to 5-100 ppm depending on the nature of
products produced from the individual company (e.g. toxicity, pharmacological activity, 10
ppm in APIs is very frequent).

www.wjpr.net Vol 8, Issue 13, 2019. 26


Mulik et al. World Journal of Pharmaceutical Research

If the calculated concentration (CONC) of the previous product (based on MACO calculated
from therapeutic doses/tox data) exceeds the general upper limit (MAXCONC), then
MAXCONC level will be the limit.

Procedure
Establish MACO ppm, based on a general limit, using the following equations.
MACO ppm = MAXCONC x MBS
E.g. for a general limit of 100ppm: MACO = 0.01% of the minimum batch size (MBS), and for
a general limit of 10ppm: MACO = 0.001% of the minimum batch size (MBS).

Remarks: The ICH impurity document (Q 3) indicates that up to 0.1% of an individual


unknown or 0.5% total unknowns may be present in the product being tested.

Example 3
A product B will be cleaned out. The product has a standard daily dose of 250 mg and the
batch size is 50 kg. The next product A has a standard daily dose of 10 mg and the batch size
is 200 kg. The general limit of the company is 10ppm. Calculate the MACO ppm for B in A!

MACO ppm= 0.00001(mg/mg) x 200000000(mg) = 2000(mg)

Result: MACO ppm is 2 g (2000 mg)


In the worst case a maximum of 2 g of B may appear in API A. This is more reasonable than
the limit 5kg calculated in example 2.

4. Swab Limits
If homogeneous distribution is assumed on all surfaces, a recommended value can be set for
the content in a swab. This can be used as basic information for preparation of a method of
analysis and detection limit.

Procedure
Establish the target value for swab limit for the whole equipment train, using the following
equation:
MACO [µ g]
Target value [µ g/dm2]= -----------------------------
Total surface [dm2]

www.wjpr.net Vol 8, Issue 13, 2019. 27


Mulik et al. World Journal of Pharmaceutical Research

Also other methods with different swab limits for different surfaces in a piece of equipment
and/ or equipment train can be used. Using this approach, the total amount found on the
equipment train has to be below the MACO.

5. Rinse Limits
The residue amount in the equipment can be assumed to be equal to the amount of residue in
the last wash /boil) or rinse solvent portion. The assumption is based on the worst case
consideration that a further washing or rinsing run (or any reaction) would not wash more
than the same amount of residue out of the equipment as the analyzed solvent portion did.

The MACO is usually calculated on each individual product change over scenario and
individual acceptance criteria are established using the following equation.

For quantization a solvent sample is taken, the residue in the sample is determined by a
suitable analytical method and the residue in the whole equipment is calculated according to
the following equation:
MACO [µ g]
Target value [µ g/l]= ----------------------------------------
Volume of rinse or boil [l]
M = V x (C - CB)

Where,
M - Amount of residue in the cleaned equipment in mg
V - Volume of the last rinse or wash solvent portion in l
C - Concentration of impurities in the sample in mg/l.
CB - Blank of the cleaning or rinsing solvent in mg/l. If several samples are taken during one
run, one and the same blank can be used for all samples provided the same solvent lot was
used for the whole run.

Requirement: M < Target value.

Documentation
A Cleaning Validation Protocol is required laying down the procedure on how the cleaning
process will be validated. It should include the following: -
 The objective of the validation process
 Responsibilities for performing and approving the validation study

www.wjpr.net Vol 8, Issue 13, 2019. 28


Mulik et al. World Journal of Pharmaceutical Research

 Description of the equipment to be use


 The interval between the end of production and the beginning of the cleaning procedures
 Cleaning procedures to be used for each product, each manufacturing system or each
piece of equipment.
 The number of cleaning cycles to be performed consecutively.
 Sampling procedures, including the rationale for why a certain sampling method is used
 Any routine monitoring equipment
 Clearly defined sampling locations
 Data on recovery studies where appropriate
 Analytical methods including the limit of detection and the limit of quantization of those
methods.
 The acceptance criteria, including the rationale for setting the specific limits.
 Other products, processes, and equipment for which the planned validation is valid
according to the “bracketing” concept; and - When Re-validation will be required.

Cleaning Procedures
Standard cleaning procedure for each part of equipment and process should be prepared.

It is important that the equipment design is evaluated in detail to remove the product residues.
Cleaning procedure should establish in line with cleaning validation study with respect to
cleaning approach, cleaning method, cleaning agent usage & related concentration, Cleaning
steps, cleaning solvents, cleaning time, trace analysis / evaluations, process of cleaning
efficacy verifications after completion of cleaning & before usage, storage time of
equipment‟s in dirt & clean condition.

Following parameters are to be considered during cleaning procedures.

A. Equipment Parameters to be evaluated


1. Identification of the equipment to be cleaned
2. 'Difficult to clean' areas
3. Property of materials
4. Ease of disassembly
5. Mobility

www.wjpr.net Vol 8, Issue 13, 2019. 29


Mulik et al. World Journal of Pharmaceutical Research

B. Residues to be cleaned
1. Cleaning limits
2. Solubility of the residues
3. Length of campaigns

C. Cleaning agent parameters to be evaluated


1. Preferable materials that are normally used in the process.
2. Detergents available (as a general guide, minimal use of detergents recommended unless
absolutely required)
3. Solubility properties
4. Environmental considerations
5. Health and safety considerations

Cleaning Agent selection: Cleaning chemistries fall into several broad categories;
1. Water
2. Solvents
3. Commodity chemicals
4. Formulated cleaning agents

D. Cleaning techniques to be evaluated:


1. Manual cleaning
2. CIP (Clean-in-place)
3. COP (Clean-out-of-place)
4. Semi automatic procedures
5. Automatic procedures / GMP washers
6. Time considerations
7. Number of cleaning cycles

Selection of batch size cleaning validation:


To establish the robust cleaning procedure, study needs to perform considering below factors:
1. Maximum batch size in term of quantity of worst selected API/ ingredients used in batch
size.
2. Maximum Surface area covered by worst selected products.
3. Maximum run time of batch on line i.e. maximum contact time.
4. Maximum Time interval between the completion of batch activities & start of cleaning.

www.wjpr.net Vol 8, Issue 13, 2019. 30


Mulik et al. World Journal of Pharmaceutical Research

5. If worst selected product is more than one, then study shall carried out on each worst
selected product for same chain of equipment‟s.
6. On basis of cleaning procedure evaluated for different worse product more than one, then
all studied cleaning procedure shall compared to each other with matrix for each step and
select the final cleaning procedure that can take care of all worst molecule.
7. As per cleaning procedure matrix, if cleaning stages / methods are contraindicating to
each other for same equipment‟s / chain of process equipment‟s e.g. different solvent or
different cleaning agent etc. Then SOP / cleaning procedure shall prepare including
provision of multiple cleaning procedures as pre previous product for equipment utilized.
8. Same product but different change of equipment‟s. E.g. Common blend for Dry Syrup,
Capsule & sachet. Packing mechanism & related equipment chains are different.

Dirt Equipment Cleaning Procedure


As per property & characteristic of the molecule and MOC & design equipment, approach to
adherence of product material with equipment surface area are different due to different
internal surface tenses. This approach of molecule towards equipment is impacting with the
time period. E.g. un-clean Tea cup dried after tea completion. It will easily clean immediate
of tea finishing, while after few hrs, it may require additional efforts & water to clean.

Its needs to ensure that cleaning procedure is suitable to clean the stored dirt equipment‟s for
worst molecule. Also along with chemical trace free study, microbial load study is important
to be evaluated.

During production, equipment chain shall monitored against the validated dirt equipment
cleaning time period.

Clean Equipment Storage Period:


After cleaning of equipment & chain of equipment‟s, its necessary to hold the same in non-
contaminated area / controlled environments. So that equipment remains clean & ready to
use. However its recommended to validate &, evaluate storage period for equipment as to
particulate free / microbial load. Its should be monitored as line clearance part before to
usage of such 'Ready To clean' equipment‟s.

www.wjpr.net Vol 8, Issue 13, 2019. 31


Mulik et al. World Journal of Pharmaceutical Research

Sampling Technique
Sampling sites was selected based on the difficult clean geometries of the equipment and
these locations are inaccessible i.e. their inaccessibility makes them difficult to clean
therefore, before choosing for sampling sites one must be conscious in selecting the desired
sampling locations.

Equipment is characterized into hot spots and critical sites. Hot spot is the location that is
likely to become dirty during the manufacturing process and it is difficult to clean.

Critical sites are those locations if remain dirty will certainly show disproportionate level of
contamination to the next exhibit batch. The common sampling method employed in cleaning
validation is rinse sampling, Direct surface sampling and swab sampling.

Direct Surface Sampling


It involves the determination of the type of sampling material used and its impact on the test
data to check the interference of the sampling material with the test. Therefore, early in the
validation programme, it is crucial to assure the sampling medium and solvent if they are
satisfactory and be readily used.

It is done by using FTIR or photoelectron emission techniques. By employing these


techniques, specific spectra obtained from residue remaining on the surface will directly
measure the quality of the surface.

Advantages
hardest to clean and which are reasonably accessible can be evaluated,
insoluble can be sampled by physical removal.
place in one step and there will be no real loss of
sampling system.

Swab Sampling
It usually requires materials which are absorptive & to physically wipe the surface and
recover the analyst. Swabs used should be compatible with the active ingredients and should
not interfere with the assay.

They should not cause any degradation of the compound. The solvent used for swabbing
should provide good solubility for the compound and should not encourage degradation.

www.wjpr.net Vol 8, Issue 13, 2019. 32


Mulik et al. World Journal of Pharmaceutical Research

Advantages

and cleaning agent residues.

Limitation
que that may introduce fibers.
material and design may inhibit recovery and
specificity of the method.
reach areas difficult.

Figure No. 3: Recommended directions and motions of swabbing.

Rinse Sampling
Generally till date understanding about rinse sampling is, it does not employ mechanical
action on the surface to collect sample, However it not true. Rinsing of equipment may be
carried out by force solvent, mostly water / purified water / WFI jet. or under pressure
recirculation of system solvent, mostly water / purified water / WFI.

www.wjpr.net Vol 8, Issue 13, 2019. 33


Mulik et al. World Journal of Pharmaceutical Research

Rinse sample will give most accurate/appropriate assurance about cleaning activities
performed.

The sample is collected as a final rinse or rinse applied specifically for collecting a validation
sample. Sampling and testing of rinse samples for residual active ingredient is a commonly
adopted method to evaluate cleanliness. This is a fairly convenient method in many cases and
requires control over the solvent used for rinsing,
 The contact time
 Quantity of solvent used during rinse. Quantity used solvent should be at least ml/cm2 for
open equipments / accessories. COP system or full volume solvent in case of closed loop
circulation / CIP system.
 Mixing force & pressure involved
 The solvent used should be selected based on the solubility of the active ingredient and
other excipients. Simulate a subsequent batch of product or at least provide adequate
solubility.

Advantages
to on-line monitoring

-intrusive
le for actives, cleaning agents and Excipients

Limitation

sitivity.

usually used
for rinsing an entire piece of equipment, such as vessel.

Placebo Sampling
Placebo is recognized as both potential cleaning techniques and potential sampling
techniques. Placebo material comprises of all typical excipients, but not the active ingredient.

www.wjpr.net Vol 8, Issue 13, 2019. 34


Mulik et al. World Journal of Pharmaceutical Research

And the placebo batches were passed through a same line so that it will have possibility to
scrub of the clean system.

The principle involved in placebo is that it is passed through the same pathway as the product
therefore; it will have the possibility to scrub off residual product along those pathways. And
it usually employed for measuring system cleanliness. It majorly depends on
1. Excipients solubility in placebo.
2. Appropriate contact time of the placebo for collecting representative sample.
3. Coverage of the placebo in-process pathways ensures removal of the placebo from all
equipment location.
4. Quantity of the placebo and residue being matched should be detectable range and the
distribution of residue uniformly in the placebo ensures the detection of sample at any portion
of the placebo.

Advantages
ntacts the same surfaces as the product.
-to-reach surfaces

Limitations
(contaminants may not be evenly distributed in the
placebo)
ytical specificity and inhibits detect ability
equipment must be cleaned after the placebo run.
potential product
esidues may not be homogenously distributed
rect measurement of residues on product contact surfaces The preferred sampling
method and the one considered as the most acceptable be regulatory authorities is the
swabbing method.

www.wjpr.net Vol 8, Issue 13, 2019. 35


Mulik et al. World Journal of Pharmaceutical Research

Table No. 1: Major Sampling Techniques and Their Attributes.

4. Coupon sampling: It involves the use of a coupon sampling or an actually removable


piece of pipe that is dipped into high purity water to extract residues for analysis.

ANALYTICAL TECHNIQUES
Choosing the appropriate analytical TECHNIQUE depends on a variety of factors. The most
Important factor is to determine the specifications or parameters to be measured. The limit
should always be established prior to the selection of the analytical tool.

There are two methods:

Non-specific method

Table No: 2: Comparison of Features of Typical Cleaning Validation Assay Methods.

www.wjpr.net Vol 8, Issue 13, 2019. 36


Mulik et al. World Journal of Pharmaceutical Research

A specific method detects unique compounds in the presence of potential contaminants. Ex:
HPLC. Non-specific methods are those methods that detect any compound that produces a
certain response Ex: Total Organic Carbon TOC), pH and conductivity.

Additional techniques
Apart from the above mentioned techniques the biopharmaceutical industry utilizes a wide
variety of techniques. TLC is widely used for the qualitative determination of surfactants.
Atomic absorption spectroscopy is used for the determination of inorganic contaminants. Bio
luminescence is useful for biologicals. This type of analysis usually uses ATP
bioluminescence.

It also include Enzyme Linked Immuno Sorbent Assay (ELISA) and Limulus.

Testing Methods: The basic requirements of the analytical methods should have the
following criteria.
1. Testing method should have the ability to detect target substances at levels consistent
with the acceptance criteria.
2. Testing method should have the ability to detect target substances in the presence of other
materials that may also be present in the sample.
3. The testing analytical method should include a calculation to convert the amount of
residue detected in the sample to 100%, if the recovery data generated indicates a
recovery outside the allowed range.

Analyzing cleaning Validation samples: There are many analytical techniques available in
cleaning validation. But choosing the appropriate analytical tool depends on a variety of
factors. The most important factor is to determine the specifications or parameters to be
measured. The limit should always be established prior to the selection of the analytical tool.

Specific and non-specific methods


A specific method detects unique compounds in the presence of potential contaminants e.g.
HPLC.

Nonspecific methods are those methods that detect any compounds that products a certain
response e.g. Total organic carbon, pH and conductivity.

www.wjpr.net Vol 8, Issue 13, 2019. 37


Mulik et al. World Journal of Pharmaceutical Research

1. High Performance Liquid Chromatography


Almost every pharmaceutical company has an HPLC instrument, utilizing a variety of
detector. These include UV, Fluorescence, Electrochemical, Refractive Index, Conductivity,
Evaporate Light Scattering Detector and many others. The vast majority of techniques
described in the literature are for the determination of surfactants in concentrated products.

Therefore, the limits of quantitation and the limit of detection are rather high. Analysis of
anionic and cationic surfactants is done by HPLC and Capillary electrophoresis whereas
amphoteric surfactants are analyzed by HPLC.

2. Capillary Electrophoresis
Capillary electrophoresis can be used for many different types of analysis, viz., separation,
detection and determination of sodium lauryl sulphate in cationic, anionic and non-ionic
surfactants. Another technique known as Micellar electro kinetic capillary chromatography is
used for the separation of non-ionic alkyl phenol polyoxy ethylene type surfactants.

3. Total organic carbon: It is used widely in the pharmaceutical industries for various
purposes. TOC is determined by the oxidation of an organic compound into carbon dioxide.
The oxidation can occur through a number of mechanisms depending on the instrument being
used. TOC is used for the analysis of detergents, endotoxins, biological media and poly
ethylene glycol.

4. Ion Chromatography: Ion chromatography can be used for the analysis of inorganic,
organic and surfactants present in the cleaners. Most cleaners contain sodium and/or
potassium. The ion chromatography detection technique of suppressed conductivity is more
sensitive to potassium ions than to sodium ions. Very low levels of cleaning agents can be
detected by using this technique.

5. Others
 Thin layer chromatography: TLC is widely used for the qualitative determination of
surfactants.
 Atomic absorption spectroscopy: AAS is used for the determination of inorganic
contaminants.
 Bio luminescence: It is useful for biologicals. This type of analysis usually uses ATP-
bioluminescence.

www.wjpr.net Vol 8, Issue 13, 2019. 38


Mulik et al. World Journal of Pharmaceutical Research

6. Optically simulated electron emission: In some cases the limits of residue are very less
that they can‟t be detected by conventional methods. OSEE is a very sensitive method that
can be used for both qualitative and quantitative manner in this regard.
7. Portable mass spectrometer: Portable mass spectrometer can be used to detect ultra-
sensitive measurements and identification of the residue.
8. Additional techniques: Apart from the above mentioned techniques, Biopharmaceutical
industry utilizes a wide variety of techniques include ELISA and LAL technique.
9. Method Validation: It is very important to scientifically establish the residue limit prior to
choosing the method of analysis. This includes the limit in the analytical sample and the limit
in the next product. This will ensure the ability of the chosen method to detect and
quantitated the limit present.

Once the technique for analysis has been chosen, it is very important to validate the method
used. The validation of a method is very different from validation of recovery. A validated
method is one that is rugged and robust enough to measure the residual limit established,
whereas, the validation of a recovery helps to determine the amount that can be recovered
from a surface.

Data analysis for estimating possible contamination: To support the cleaning validation
study, an appropriate analytical method must be developed to product at a sensitivity level, at
least equal to that of the acceptable residual level. For each analytical method, values defined
as „minimum quantifiable quantity‟ (MQQ) and non-detectable (ND) are applied. A test
result greater than or equal to the MQQ is considered reliable, whereas if it lies between ND
and MQQ it is considered unreliable. Therefore values reported as ND or between ND and
MQQ can be manipulated to apply for the possible contamination.

Difficulty in cleaning the equipment: The most difficult to clean pieces of equipment
require the most intensive monitoring schedule. Easier to clean pieces require a moderate
monitoring schedule.

Difficulty in cleaning the product and equipment


It is divided into three groups based on the degree of difficulty in cleaning the product and
equipment.
1. Most difficult to clean product and equipment‟s requires the most intensive monitoring
schedule.

www.wjpr.net Vol 8, Issue 13, 2019. 39


Mulik et al. World Journal of Pharmaceutical Research

2. Easier to clean product and equipment that requires a moderate monitoring schedule.
3. Easier to clean product and equipment that requires only periodic monitoring.
The monitoring program provides a mechanism to verify the capability of the cleaning
procedures, the efficiency of the training program and the effectiveness of the equipment
maintenance program.

VALIDATION PROTOCOLS
A Validation Protocol is necessary to define the specific items and activities that will
constitute a cleaning validation study. It is advisable for companies to have drawn up a
master validation plan indicating the overall cleaning Validation strategy for the product
range / equipment type / entire site. The protocol must be prepared prior to the initiation of
the study and must either include or reference the documentation required to provide the
following information.

validation study
Sampling procedure to be used

before the study

VALIDATION REPORTS
A validation report is necessary to present the results and conclusions and secure approval of
the study. The report should include the following.
Summary of or reference to the procedures used to clean, sample and test
references for same, as well as any pertinent
observations.
ons regarding the acceptability of the results, and the status of the procedure(s)
being validated.
the results or relevant information obtained during the
study including revalidation practices if applicable.

www.wjpr.net Vol 8, Issue 13, 2019. 40


Mulik et al. World Journal of Pharmaceutical Research

that occurred.
batches of the product will be manufactured for a
period of time it is advisable to generate interim reports on a batch by batch basis until such
time as the cleaning validation study has been completed.
appropriate level of verification subsequent to validation.

An effective cleaning validation maintenance programme. When a minimum of three


cleaning validation runs get completed and if the results meet the acceptance criteria, then the
cleaning procedures would be demonstrated sufficiently and consistently to remove chemical
and detergent residues from equipment surfaces during the study in order to meet the pre-
established criteria. However, overtime and certain other factors can decrease the efficiency
and consistency of the cleaning program.

They are
1. Operator variability
2. Equipment aging and repair
3. Potential non representative results and monitoring programmes.
4. Changes to the product, equipment and process.

CONCLUSION
Cleaning is directly related to safety and purity of the pharmaceutical product therefore it
becomes most important and primary activity. So, It is necessary to have effective cleaning
program in place because of the regulatory requirement.

Form this review article it can be concluded that cleaning validation is a process of attaining
and documenting sufficient evidence to prove the effectiveness of cleaning process. this
article covers all aspects related to cleaning validation like Residue selection, acceptance
criteria for the validation, different levels of cleaning, cleaning procedure, sampling
procedure, product grouping and equipment characterization, cleaning agent selection &
current gaps in cleaning validation study & in its establishment through cleaning procedures
with respect to quantity used for rinse, selection of batch size for cleaning validation & dirt
cleaning study.

www.wjpr.net Vol 8, Issue 13, 2019. 41


Mulik et al. World Journal of Pharmaceutical Research

Cleaning validation provides a means of proving that the contamination levels of proving that
the contamination levels have been reduced below contamination acceptable limits.

Cleaning validation programme should be based on detailed cleaning procedures, a validation


protocol, validated methods, a change control programme, and a validation report.

REFERENCES
1. Rebecca Brewer Dober Group, Midlothian, Illino: Cleaning Validation for the
Pharmaceutical, Biopharmaceutical, Cosmetic, Nutraceutical, Medical Device, and
Diagnostic Industries Validation of Pharmaceutical Processes Third Edition, 491-518.
2. Sharma A, Vaghela J.S, Sharma P, Sharma B, Agrawal M, Jagiwala S: cleaning
validation in pharmaceutical industry: a comprehensive approach; pharmatutor-art, 1755.
3. Parenteral Drug Association. Points to Consider for Cleaning Validation .Technical
Report No, 29: 1998.
4. Robert Fredrick: The Basic Facts of Cleaning Validation. Canada: Pharmainfo.net; 2004.
Available from.
5. Fourman GL, Mullen MV. Determining cleaning validation acceptance limits for 37
Babita Lodhi et al, JIPBS, 1(1): 27-38, 2014 Pharmaceutical manufacturing operations.
Pharm Tech, 1993; 54-60.
6. Vitale KM. Cleaning validation acceptance criteria, 15th Annual Pharm Tech conference,
East Brunswi New jersey, September 1995.
7. Brewer R. Regulatory aspects of cleaning validation, ISPE seminar, Rockville, Maryland,
March 1996.
8. D. Narayana Murthy and K. Chitra Fourman GL, Mullen MV. Determining cleaning
validation acceptance limits for Pharmaceutical manufacturing operations. Pharm Tech,
1993; 54-60.
9. Bodavula Samba Siva Rao: Cleaning validation of albendazole tablet 400 mg. Pharma
Innovations, 2012; 1(4): 76-94.
10. Kumar S: A review on concept of cleaning validation in pharmaceutical Industry.
International Research Journal of Pharmaceutical Sciences, 2012; 3(7): 17-19.
11. Kumar V S, Sanjeev T: Overview of cleaning validation in pharmaceutical manufacturing
unit. International Journal of Advanced Research in Pharmaceutical & Biosciences, 2012;
1(2): 154-164.

www.wjpr.net Vol 8, Issue 13, 2019. 42


Mulik et al. World Journal of Pharmaceutical Research

12. D. Narayana Murthy and K. Chitra: a review article on cleaning validation International
Journal of Pharmaceutical Sciences and Research, 2013; 4(9): 3317-3327.
13. Kaiser HJ, Tirey JF, LeBlanc DA: Measurement of Organic and Inorganic Residues
Recovered from Surfaces. J Valn Tech, 1999; 6(1): 424-436.
14. LeBlanc DA. Establishing Scientifically Justified Acceptance Criteria for Cleaning
Validation of Finished Drug Products. Pharm Tech, 1998; 22(10): 136-148.
15. Fourman GL, Mullen MV. Determining Cleaning Validation Acceptance Limits for
Pharmaceutical Manufacturing Operations. Pharm Tech, 1993; 17: 54-60.
16. Inampudi P, Lombardo S, Ruezinsky G. An Integrated Approach for Validating Cleaning
Procedures in Biopharmaceutical Manufacturing Facilities. Annals of the New York
Academy of Sciences, 1996; 782: 363374.
17. Rowell FJ, Miao ZF, Neeve RN. Pharmaceutical Analysis Nearer the Sampling Point,
Use of Simple, Rapid On-Site Immunoassays for Cleaning Validation, Health and Safety,
and Environmental. Release Applications. J Pharm & Pharmacology, 1998; 50: 47.
18. Hwang RC. How to establish an effective Maintenance program for cleaning validation.
Pharm Tech, 2000; 24(1): 62-72.
19. McCormick PY, Cullen LF. Cleaning validation In: Berry, I.R., and Nash, R.A. editor.
Pharmaceutical Process Validation. 2nd ed. New York: Marcel Dekker; 1993; 319-349.
20. Lakshmana P: Cleaning validation and its importance in pharmaceutical Industry. Pharma
Times, 2010; 42(7): 21--25.
21. James A. Points to consider in the validation of equipment cleaning procedures. J Parental
Sci Tech, 1992; 46(5): 163-168.
22. Phillips GF. The need for impurity control. Die Pharm Ind, 1989; 51(11): 1282-1286.

www.wjpr.net Vol 8, Issue 13, 2019. 43

You might also like