Sei sulla pagina 1di 87

SPECIAL EDITION

CLEANING
VALIDATION

Published by:
CLEANING VALIDATIoN
Validation Case Study: Erroneous Negative Cleaning Validation Results | IVT ...................................................... 1

Experimental Parameters for Small-scale Cleaning Characterization


Part I: Dilution of Process Fluids During Cleaning | IVT ....................................................................................... 7

Experimental Parameters for Small-Scale Cleaning Characterization.


Part II: Effect of Fluid Velocity on the Kinetics of Cleaning | IVT ........................................................................ 11

Methodology for Assessing Product Inactivation during Cleaning


Part I: Experimental Approach and Analytical Methods | IVT ............................................................................. 16

Methodology for Assessing Product Inactivation During Cleaning


Part II: Setting Acceptance Limits of Biopharmaceutical Product Carryover for Equipment Cleaning | IVT ........... 20

Aseptic Transfer Risk Assessment: A Case Study | IVT ...................................................................................... 27

People in Cleanrooms: Understanding and Monitoring the Personnel Factor | IVT .............................................. 32

PAT: Using PAT to Support the Transition from Cleaning Process Validation to Continued Cleaning Process
Verification | IVT ............................................................................................................................................. 39

Translating Laboratory-Developed Visual Residue Limits to Process Area Applications | IVT .............................. 45

New Perspectives on Cleaning: Cleaning Validation of Multiproduct EquipmentAcceptance Limits for Inactivated
Product | IVT ................................................................................................................................................... 51

Biopharmaceutical Cleaning Validation: Acceptance Limits for Inactivated Product Based on Gelatin as a
Reference Impurity | IVT .................................................................................................................................. 60

Multiproduct Cleaning Validation: Acceptance Limits for the Carryover of Inactivated API Part IThe Comparable
Quality Approach ............................................................................................................................................. 67

Validation of a Cleaning Process for Medical Devices | IVT ............................................................................... 72

Ensuring Sterility: Autoclaves, Wet Loads, and Sterility Failures | IVT ............................................................... 79
2015 Advanstar Communications Inc. All rights reserved.
Reproduction in whole or part is prohibited without prior written permission of the publisher.
CLVQ4005
Paul L. Pluta

Validation Case Study:


Erroneous Negative Cleaning
Validation Results | IVT
Paul L. Pluta, Ph.D.

Validation Case Studies provides a forum for validation practitioners to share


information about actual validation experiences. Previous discussions ad-
dressed a wide range of activities. Previous case study titles discussed in this
series include the following:

1. Visual Observations, Journal of Validation technology ( JVT), Volume


16, #1.
2. Equipment Qualification, JVT, Volume 16, #1.
3. Identical mixing Tanks, JVT, Volume 16, #3.
4. Cleaning HPLC Peaks, JVT, Volume 16, #4.
5. Documentation Practices, JVT, Volume 17, #1.
6. Yield, JVT, Volume 17, #2.
7. Like-for-Like Changes, JVT, Volume 17, #2.

Readers are invited to participate and contribute manuscripts for this


series -- we encourage sharing successful practices with others. Please contact
journal editor-in-chief Paul Pluta at paul.pluta@comcast.net or content spe-
cialist Dustin Henderson at dustin.henderson@cbinet.com with comments or
submissions for publication.

ABSTRACT
This case study describes a cleaning validation event in which failing
results for API residue from a small molecule extended release tablet
dosage form were observed. The initial two lots in the cleaning valida-
tion were successful. The third lot failed acceptable residue limits.
Investigation of the failure comprised cleaning process development
and performance; residue sampling, sample handling, sample analy-
sis, and evaluation of the analytical method. Investigation of this
event initially involved interviews of relevant personnel and reviews
of associated documentation. Two areas were identified for further
evaluation residue sampling and the cleaning process. Regarding
sampling, a newly trained technician, working alone, sampled the first
two acceptable lots, while an experienced technician working with a
colleague sampled the third failing lot. Evaporation of sampling sol-
vent occurred causing residue to be insufficiently recovered from the
equipment surface resulting in erroneous false negative test results.
Regarding the cleaning process, manufacturing operators commented
that the new extended release formulation was more difficult to clean
than the original immediate release formulation although the same
cleaning procedure was utilized for both products. Evaluation of the
cleaning process indicated that the process parameters were not opti-
mal to clean the new extended release product. An improved cleaning
process with increased cleaning agent concentration, increased clean-
ing time, and higher temperatures was developed, implemented, and
ultimately validated.

Special edition: Cleaning Validation 1


Paul L. Pluta

Cleaning validation sampling personnel must BACKGROUND


have good technical understanding of their work, The process of validation typically comprise the fol-
and must know the technical reasons for the pro- lowing sequence of activities:
cedures they perform, and potential problems if
procedures are not correctly performed. Sampling 1. Change desired. An equipment or process
personnel training for cleaning validation should change is needed or required. This may be a
include a quantitative demonstration of acceptable necessary change or a desirable improvement.
cleaning by means of analytical testing. Training 2. Development work. Appropriate Stage 1 devel-
exercises must also include worst-case sampling opment work is completed in support of the
such as with volatile solvents, multiple equipment, change.
and other potential variations in sampling. SOPs 3. Validation plan. A formal request to initiate the
must be carefully written to describe potential validation process is submitted to the valida-
problems and include performance constraints to tion approval committee (VAC). Development
minimize variation and risks. There is an inherent reports may be included in the request in sup-
danger when variation is not deliberately intro- port of the change. The change request includes
duced into a validation project material variation, a proposed level of work to confirm the accept-
manufacturing operator variation, and in this case ability of the change. The level of work is based
study, sampling personnel variation. Sampling by on risk to the patient and to the organization.
two different technicians enabled erroneous results The VAC approves the change request. The
to be discovered. Regarding the cleaning process, approved change request document is stored in
inactive ingredients in a formulation may have very the validation library.
significant effects on cleaning processes. Cleaning 4. Protocol. A protocol is written specifying
of residues does not depend solely on the proper- detailed sampling and testing to confirm the
ties of the API. acceptability of the change. The VAC approves
the protocol. The approved protocol is stored in
INTRODUCTION the validation library.
The compliance event involved cleaning valida- 5. Validation work. Stage 2 PPQ validation work
tion for cleaning of residue from a new small is performed according to the protocol. Sam-
molecule extended release tablet dosage form. pling and testing are completed. Data and other
The active ingredient in the tablet was a potent results are generated and recorded.
drug dosage of the active ingredient was low 6. Validation report. A report containing all
and cleaning was expected to be successful. The test results with discussion and conclusions
site already had a successful history of cleaning is prepared and submitted to the VAC for
a marketed immediate-release tablet containing approval. The report is approved and the pro-
a lower dose of the same active drug. No changes cess or equipment change is implemented. The
in the cleaning method were required for the new approved validation report is stored in the vali-
product. After cleaning, the target residue level dation library.
was below visually clean and required the resi- 7. Validation closure. If no other work is needed,
due level to be determined by swab sampling and the validation project initiated by the change
chemical analysis. Three lots were required for request is closed.
cleaning validation. 8. Continued verification. Stage 3 post-validation
The following are discussed: monitoring confirming acceptability of the
change continues throughout the product / pro-
Compliance event. A description of the cleaning cess lifecycle.
validation event
Investigation. Interviews and actions conducted The issue addressed in this case study occurs
to investigate the event in #5 and #6 above. The actual work conducted to
Discussion. Key information, activities, and anal- confirm the acceptability of the validation project is
ysis performed by technical people. In this case study,
CAPA. Actions and improvements implemented samples were removed and tested for residue content.
in the cleaning process, sampling process for
cleaning validation, and training of sampling per- VALIDATION EVENT
sonnel A small molecule pharmaceutical company conducted
Cleaning validation of modified cleaning process. initial cleaning validation on a new extended release
Implementation of the new cleaning process and tablet product containing a water-insoluble API as
subsequent validation. active ingredient. The new product was a line exten-

2 Special edition: Cleaning Validation


Paul L. Pluta

sion -- an extended release formulation of a marketed 4. Residue samples. Was the integrity of residue
immediate-release tablet product. The new product samples adequately protected during transport
contained a polymeric matrix to enable prolonged to the lab? Could samples have become contam-
release and once-daily dosage to patients. inated causing the test failures? Were samples
The cleaning validation exercise was expected correctly and quickly transferred to the lab for
to be successful. Although the product contained a analysis? Were samples handled during trans-
highly potent active drug which required low residue port and storage according to procedures? Were
levels on cleaned equipment, the company had ex- samples exposed to high temperatures during
tensive experience with the cleaning procedure over transport and storage?
several years. The original immediate-release product 5. Analytical laboratory. Was the analysis correctly
cleaning was relatively easy and had a long success- performed? Which technician performed the
ful history of performance. Several previous cleaning analysis? Were laboratory technicians adequate-
validations had been successfully accomplished. The ly trained? Was analytical equipment qualified
analytical method for residual API from swab samples for use for the API analysis? Was system suit-
was easily performed and very reliable. ability below required limits?
Sampling of three lots of new product was planned 6. Analytical R&D. Were there any problems
for cleaning validation. The manufacturing process with the analytical method? Was the analytical
comprised several unit operations. Sampling of unit method correctly developed? Was the analytical
operations for cleaning validation was performed on method validated?
multiple days for each lot. The first lot was manu-
factured and cleaning completed on all equipment. INVESTIGATION
Equipment was visually clean. Swab sampling was Investigation of this compliance event initially in-
done by the sampling technician. Cleaning validation volved interviews of relevant personnel and reviews
analytical test data indicated no active drug present of associated documentation. Personnel related to the
in all swab samples all acceptable results. A second compliance event included manufacturing personnel,
lot was manufactured. Cleaning was completed. Swab QC personnel, cleaning sampling technicians, and
sampling was done. Cleaning validation analytical the technical personnel responsible for product for-
test data again indicated no levels of residual drug mulation and process, cleaning method development,
in all swab samples. A third lot was manufactured. technical support, and analytical testing. There were
Cleaning was completed. Swab sampling was done. many details and variables that needed to be inves-
Cleaning validation analytical test data indicated tigated and/or confirmed. Personnel from all groups
extremely high residue levels significantly above interacted to address the above issues.
the required acceptance criteria. Test results on the Documentation reviews included manufacturing
third lot indicated a significant failure of the cleaning documentation, cleaning documentation, equipment
process. inspection records, laboratory records, analytical
method development reports, validation reports, and
This event prompted multiple questions to be investi- other records. All applicable manufacturing SOPs
gated and answered. and analytical SOPs were reviewed.
1. Cleaning process performance. Did manufac-
turing personnel correctly perform the cleaning Cleaning Process Performance
process in the third (failing) lot? Which opera- Manufacturing personnel correctly performed the
tor cleaned the equipment? Were manufactur- cleaning process in all three lots. Cleaning procedure
ing personnel adequately trained in the cleaning documentation for all lots was reviewed and found to
procedure? What was past history with use of be perfectly executed. No deviations were issued. Dif-
this cleaning process? Were repeat cleanings ferent operators executed cleaning of multiple equip-
required in past cleaning? Were deviations ment in the validation lots. An experienced operator
required? executed cleaning in the third (failing) lot. Training
2. Cleaning process development. How were the records for all operators were reviewed and found to
cleaning process parameters developed? What be acceptable. Operators commented that the new
was the history of this method with the imme- extended release product was more difficult to clean
diate release product? We any changes made for than the original immediate release product. The
cleaning the extended release product? extended release polymer made removal of the prod-
3. Sampling. Did the sampling technician cor- uct residue more difficult that was typical with the
rectly sample the recommended equipment original immediate-release product. This observation
surfaces? Were sampling personnel adequately reflected operator experience with manual cleaning
trained? of small parts. Product was able to be removed from

Special edition: Cleaning Validation 3


Paul L. Pluta

equipment surfaces and yield visually-clean surfaces. performed cleaning as required by procedure. Equip-
No repeat cleanings were required. No deviations ment was cleaned by automated methods wherever
were issued. possible. All associated small parts were manually
cleaned was cleaned according to procedure. The
Cleaning Process Development manufacturing supervisor verified that procedures
The cleaning process for the immediate release were followed and that the equipment was visu-
product had been previously developed. An alkaline ally clean. Quality unit personnel who inspected
cleaning agent that was used on several other prod- the equipment also verified that all equipment and
ucts in the plant was used. Because the API in the small parts were visually clean. All inspections were
new extended release product was the same as in the conducted after the equipment was dry. Samples
immediate release product, no changes were made were transported to the lab quickly and according to
to the cleaning method. Technical personnel were procedure. Samples were also quickly stored in the
unaware of any difficulty in cleaning the extended laboratory upon receipt and under specified security
release product. conditions. Laboratory personnel confirmed accept-
able performance of analytical procedures. Analytical
Sampling standards over a range of concentrations tested along
Two different sampling technicians performed sam- with the actual cleaning validation samples yielded
pling in the three lots. A newly-trained technician accurate results. Analytical R&D scientists confirmed
sampled lots #1 and #2, both of which had accept- acceptable performance of the validated test method.
able low residue levels. The newly-trained techni- Two areas were identified for further investigation.
cian worked alone to accomplish sampling since These included:
no other sampling technicians were available. Lot
#3 was sampled by an experienced technician. The 1. Swab sampling. A newly trained technician,
experienced technician worded with a colleague who working alone, sampled the first two acceptable
helped sample the recommended equipment surfaces lots. All samples in these lots were acceptable.
and complete required documentation. Both sam- An experienced technician, working with a
pling personnel were adequately trained as evidenced colleague, sampled the third failing. Was some-
by training documentation. thing different about the third lot, or was the
failing data due to the difference in sampling
Residue samples personnel?
Residue samples were packaged in protective wrap- 2. Cleaning process. Manufacturing operators
ping for transfer to the lab. Samples were immediately commented that the new extended release for-
closed and not contaminated. Transport to the lab mulation was more difficult to clean than the
was rapid and without exposure to unusual environ- original immediate release formulation. The
mental conditions or excessive heat. same cleaning procedure was utilized for both
products.
Analytical laboratory
The laboratory analysis was correctly performed. Swab Samplingy
Experienced technicians performed the analysis. Swab sampling for the three lots was done by two
All technicians were adequately trained. Analytical different sampling technicians. The first two lots were
equipment was qualified for use for the API analysis. sampled by a newly-trained person. Data for these
Lab documentation indicated acceptable execution of lots indicated minimal or no residual soil acceptable
the analytical procedure. results. The third lot with failing data was sampled
by an experienced technician who worked with a
Analytical R&D colleague.
The same analytical method was used for the original The sampling method required wetting of the
irradiate-release product and for the new extended- swab with organic solvent to dissolve residue from
release product. Analytical R&D verified that the test the equipment surface. The new technician did all
method performed acceptably for the new product. sampling alone. The experienced technician per-
There were no problems with the analytical method. formed sampling with a colleague to accomplish the
The analytical method was validated. sampling procedure in minimum time. She explained
the necessity of the rapid sampling technique because
DISCUSSION evaporation of the sampling solvent must be mini-
Interviews and discussion of the above questions did mized. The new technician was not aware of the time
not clearly indicate an obvious cause for the prob- limitation in sampling. Although not conclusively
lem. Manufacturing personnel confirmed that they proven, it was suspected that evaporation of solvent

4 Special edition: Cleaning Validation


Paul L. Pluta

occurred causing residue to not be adequately recov- was not performed quickly, solvent evaporated and
ered from the equipment surface. The new technician surface residue was not able to be dissolved. Analyti-
worked slowly and carefully, and completed all neces- cal results on evaporated swab samples indicated
sary steps. However, the time required for perfor- extremely low or no levels of residue which errone-
mance, especially since she worked alone, may have ously passed cleaning validation acceptance criteria
caused residue recovery to be incomplete or minimal. a false negative due to solvent evaporation.
The analytical lab confirmed that if sufficient solvent Future training of swab sampling technicians
was not present on the swab, residue recovery would included new test procedures to require rapid
be unsuccessful. performance of sampling procedures. The previous
qualification test did not utilize a volatile solvent and
Cleaning Process did not require rapid performance. The new qualifi-
Technical personnel responsible for the cleaning cation test required technicians to demonstrate rapid
process had no previous experience with the clean- sampling in order to become a qualified sampling
ing method. The cleaning method for the original technician. Sampling teams (two technicians) were
product had been established many years ago and required when volatile solvents were used in sam-
never required new technical evaluation. Manufactur- pling. Technicians were required to quantitatively
ing management decided to use the well-established recover residue in training to be qualified for residue
cleaning method without involvement of technical sampling. Training was repeated on an annual basis.
personnel. Managements rationale was that since the SOPs describing cleaning sampling methods using
API in the original product had been reliably cleaned volatile solvents were strengthened to require rapid
for many years, there was no need to evaluate the sampling and working in teams. The combined em-
cleaning process for the new product. Technical per- phasis of new training and new procedures that both
sonnel had not been requested to evaluate the clean- underscored the risks and potential variation in resi-
ing process used in the failed cleaning validation. due sampling strongly addressed the issues described
In light of the cleaning failure, technical personnel in this case study.
recommended laboratory studies to evaluate available
cleaning agents, cleaning process parameters, and Modified Cleaning Process for Extended Release
related factors in a systematic way. Evaluation of the product
cleaning process indicated that the process param- Technical personnel evaluated the cleaning process
eters were insufficient to clean the new product. The and determined that process parameters were not op-
polymeric matrix in the new product (methylcellulose timal to reliably clean process residues. The cleaning
mixture) was much more difficult to clean than the agent concentration was increased, the temperature
original immediate release product. Technical per- was increased, and the cleaning time was increased
sonnel conducted studies to establish new cleaning in the new procedure. These parameters enhanced
process parameters suitable for the extended release the cleaning process to more effectively and more ef-
product. A new cleaning method with increased ficiently remove the polymeric residue.
cleaning agent concentration, increased cleaning
time, and higher temperatures was developed. CLEANING VALIDATION OF MODIFIED CLEANING
PROCESS
CORRECTIVE ACTION / PREVENTIVE ACTION (CAPA) The new cleaning process was implemented. Manu-
Two CAPA activities corrected the problems expe- facturing operators confirmed that new cleaning
rienced in the original cleaning validation. These process parameters significantly improved the clean-
involved new training of swab sampling personnel ing process. Three product lots were manufactured.
and a modified cleaning process for the extended Cleaning was performed on required equipment in
release product. three lots. Worst-case locations on equipment were
swab sampled by two-person teams of sampling per-
Swab Sampling Training sonnel. Two-person teams ensured minimal solvent
Personnel who perform cleaning residue sampling evaporation and rapid sampling procedures. All test
using swabs wetted with volatile solvents were taught results passed the acceptance criteria.
the importance of rapidly performing swab sampling.
Many of the swab sampling technicians did not have SUMMARY AND FINAL THOUGHTS
a technical background and did not understand A case study describing a compliance event in which
solvent volatility and the consequences for swab erroneous false negative analytical data was generated
sampling. Studies confirmed that the new techni- in cleaning validation. These data caused a mistaken
cian, who worked alone in the sampling activity, did conclusion that a cleaning process for a new modi-
not perform swab sampling quickly. When sampling fied release dosage form was acceptable. The cause

Special edition: Cleaning Validation 5


Paul L. Pluta

of the problem was not easily determined all test the procedures they perform. They must know
data were acceptable. Initial investigation of potential potential problems if procedures are not correctly
problem areas indicated that everything was done performed.
according to procedure nothing was done incor- New procedures. SOPs describing cleaning
rectly. It was ultimately determined that the sampling sampling methods using volatile solvents were
process for product residue was not sufficiently con- strengthened to require rapid sampling and
trolled, and that the equipment cleaning process was working in teams. Time constraints were added
not adequate for the modified release formulation. to all affected procedures. SOPs must be carefully
The sampling error, i.e., loss of solvent in sampling, written to identify potential risks and minimize
had a major effect on cleaning validation. The sam- variation.
pling technician did not understand the importance Sampling personnel training. Training of
of working quickly to minimize solvent loss. This cleaning validation sampling technicians is a
lack of understanding resulted in a false negative test critical activity. Training exercises must include
result and an erroneous conclusion that the cleaning a quantitative demonstration of acceptable
process was acceptable. Fortunately the error was cleaning by means of analytical testing. Training
discovered when a different technician correctly and exercises must also include worst-case sampling
rapidly sampled the equipment surfaces. The com- such as with volatile solvents, multiple sampling
bined emphasis of new training and new procedures equipment, and other potential variations used
that both emphasized the risks and potential varia- in sampling. Retaining of technicians at some
tion of sampling strongly addressed the sampling defined and reasonable frequency should be
issues described in this case study. Observations by considered.
manufacturing personnel caused the cleaning process Inactive ingredients effects on cleaning. Inac-
to be evaluated by technical personnel, and a new tive ingredients may have very significant effects
cleaning process with optimized process parameters on cleaning processes. Cleaning of residues
was developed. The new cleaning process was ulti- does not depend solely on the properties of the
mately validated. API. Formulation ingredients may significantly
affect the cleaning process. In this case study,
Lessons Learned an extended release polymer in the formulation
Several important lessons may be learned from this caused difficulty in the cleaning process. Inactive
case study. ingredients such as dyes and flavors may also
Sampling personnel understanding of sam- greatly influence cleaning, and may actually be
pling process and training. Sampling person- the most difficult ingredients to clean in a formu-
nel must have good technical understanding lation. All components in a formulation must be
of their work. They must know the reasons for considered when developing a cleaning process.

6 Special edition: Cleaning Validation


Rizwan Sharnez

Experimental Parameters
for Small-scale Cleaning
Characterization Part I:
Dilution of Process Fluids
During Cleaning | IVT
Rizwan Sharnez, Ph.d., Angela to, Arun tholudur, Ph.d.

CLEANING VALIdAtIoN
Methodologies for estimating experimental parameters for small-scale cleaning
characterization studies are described in this series: dilution of process fluids (e.g.,
process soil, cleaning solution, or rinse water) during cleaning is discussed in this
paper; worst-case fluid velocity and soil load will be discussed in subsequent parts.
Dilution of the process fluid during cleaning was estimated to be on the order of
1016 for a typical cleaning cycle and 105 for intermediate cleaning steps. These
dilution factors are used to estimate the concentration of impurities in the final
rinse and to simulate worst-case cleaning conditions for cleanability and inactiva-
tion studies.

INtRodUCtIoN
Small-scale cleaning characterization (CLC) studies are used to identify suit-
able cleaning chemistries (1, 2), optimize cleaning parameters and processes
(3, 4), establish cleaning times for manufacturing equipment (5, 6), and
streamline validation requirements for multiproduct equipment (7, 8).
Experimental models for small-scale CLC have been described in the
literature (9-12). In these experiments, the kinetics of soil removal (mass
transfer) from a surface is measured under simulated cleaning conditions. A
critical step in the development of these models is to identify and scale down
the hardest-to-clean (worst-case) location in the equipment (13). Note that
it is not necessary to simulate the entire cleaning process; instead, it is only
necessary to simulate the location within the equipment that is the hardest
to clean. If the process soil can be adequately removed from the worst-case
location, it follows that it can also be adequately removed from the other
locations in the equipment. Further, with this approach, the cleaning times
obtained at small-scale would be indicative of those at full scale assuming
that there is adequate coverage at the surfaces that need to be cleaned.
The scalability of small-scale CLC data depends on the accuracy with
which the experimental parameters are estimated. Experimental parameters
for simulating the worst-case location fall into two categories (14):

Parameters that can be readily determined from process data such as:
o Material of construction and surface smoothness of coupons or parts
used to simulate large-scale equipment
o Post-soiling parameters such as hold time, temperature, and humidity
o Cleaning parameters such as rinse or wash time, temperature of rinse
solvent, and temperature and concentration of cleaning solution.
Parameters that typically need to be determined from first principles
such as:
o Dilution of the process fluid during cleaning

Special edition: Cleaning Validation 7


Rizwan Sharnez

o Velocity of rinse solvent or cleaning solution at the worst-case process soil from the standpoint of drainage.
worst-case location Also, residual volumes obtained with curved surfaces
o Soil load at the worst-case location. (pipes) were significantly greater than that for flat sur-
faces (plates). Consequently, pipes were used to estimate
Residual Process Fluid the worst-case residual volumes.
The volume of residual process fluid (VR) can be esti-
mated from the surface area of the equipment that makes
contact with the process fluid (SA) and the residual
volume of the process fluid per unit surface area (R):

VR = SA R [Equation 1a]

R can be determined experimentally by measuring the


amount of process fluid that remains on a surface after it
is drained. An experimental method for estimating R is
described in the next section.
VR for a CIP circuit can be estimated by dividing the
equipment into the following sections: vessel walls (W);
bottom dish of vessel (D); and associated piping, filter Figure 1: Experimental Setup for Estimating Residual Process Fluid.
housings, and other miscellaneous parts (P). Thus,
The experimentally determined residual volumes for the
VR = SAw Rw + SAD RD + SAP RP [Equation 1b] configurations that were tested are given in Table I.
Note that impellers, filter housings, and other com-
For a tank, the surface area of the sidewall that makes ponents associated with the vessel may contain surfaces
contact with the process fluid is at a range of angles, typically between 5 and 90. The
residual volume for these components may be set to the
SAw = dh [Equation 2] R values for the smaller angle of repose (5) because it
represents a worse case from the standpoint of drainage.
Where d is the diameter of the tank and h is the height
up to which the process fluid wets the sidewall.
The bottom dish surface area (SAD) is estimated from
ASME engineering tables (15). The surface area of piping,
filter housings, and other miscellaneous components
(SAP) is estimated with a 15% overage factor. Thus,

SAP = 0.15 (SAw + SAD) [Equation 3]


table I: Residual Volumes per Unit Surface Area (R) for Deionized Water (<1
Estimation of Residual Process Fluid S/cm) and Process Soil.
The experimental method for estimating the residual
volume of a process fluid (R) is described in this section. dilution of Process Fluid during Cleaning
Two flanged stainless steel sections of piping and an end For a given step in the cleaning process, the dilution of
cap were connected as shown in the Figure. The pipes the process fluid (DS) is determined by the ratio of the
were one inch in diameter and four inches in length. volume of the cleaning solvent or solution (VS) to the
The bottom section of piping was used to model the volume of the residual process fluid on the surface that is
continuous nature of surfaces at full scale; it was not part being cleaned (VR):
of the surface area that was used to estimate the residual
volume. The piping assembly was filled with the process DS = VS / VR [Equation 4]
fluid, turned to the appropriate angle (5 or 90), and
then drained by removing the end cap. The amount of VS can be obtained from the cleaning cycle param-
residual process fluid in the top section was measured by eters, and VR can be estimated as described in the previ-
gravimetry. ous section.
Residual volumes were estimated for rinse water and Equipment cleaning cycles typically consist of mul-
process soil. These fluids were simulated with deion- tiple steps such as a pre-rinse, alkaline wash, post-alka-
ized (DI) water (< 1 S/cm) and a 56% glucose solution line rinse, acidic wash, post-acidic rinse, and final rinse.
(w/w), respectively. Due to its very high concentration The residual process fluid is serially diluted during each
and viscosity, the glucose solution was representative of a step of the cleaning cycle. The cumulative dilution of the

8 Special edition: Cleaning Validation


Rizwan Sharnez

process fluid (DC) is the product of the DS values for all DS,PR = VS,PR / VR = 125 L / 0.345 L = 362
of the subsequent cleaning steps to which the process DC,PR = DS,PR = 362
fluid is subjected:
Alkaline wash (AW):
[Equation 5] DS,AW = VS,AW / VR = 312 L / 0.345 L = 904

Where i denotes the ith step and n is the total number DC,AW = DS,PR x DS,AW = 362 x 904 = 3.27 x 105
of subsequent cleaning steps.
Note that the above equations do not account for any
change in the volume of the residual process fluid (VR)
due to drying. Thus, this approach is only applicable
when VR is negligible.

Estimation of dilution during CIP


In this section, the dilution of a process fluid is estimated
for a CIP circuit for a 10,000 liter (L) vessel. The vessel
has a diameter (d) of 84 inches (2.14 m), and the process
fluid wets the side wall of the vessel up to a height (h) table II: Dilution of Process Fluid During Cleaning.
of 120 inches (3.04 m). The cleaning steps and solution
volumes for the circuit are given in Table II. Assuming that an impurity has negligible affinity for
The surface area of the vessel in contact with the pro- the equipment surfaces and is miscible, solubilized, or
cess fluid is estimated from Equations 2 and 3 and the otherwise homogeneously distributed in the process
engineering table for ASME domed heads (15): fluid, the cumulative dilution factors in Table II can be
used to estimate the concentration in the final rinse. For
SAW = dh = 20.4 m2 example, a miscible impurity such as hydrogen peroxide,
an oxidizing cleaning agent,would be diluted by a factor
SAD = 4.24 m2 of 2 x 1016 during the six steps of the above cleaning
cycle. Thus, if the concentration of peroxide before the
SAP = 0.15 (SAD+SAW) = 3.70 m2 pre-rinse is 10,000 ppm, its concentration in the final
rinse would be <1 part per quintillion (i.e., 1 part per
The residual volume per unit area (R) for the above 1018).
surfaces is obtained from the R-values for 56% glucose The step dilution factors for intermediate steps of the
(representative of the worst-case process soil) in Table cleaning cycle are summarized in Table III.
I. The R value for a 90 angle of repose (6.60 mL/m2) is
used to estimate the residual volume for the side walls
(RW), and the R value for a 5 angle of repose (26.5 mL/
m2) is used to estimate the residual volumes for the ves-
sel dish and miscellaneous parts (RD and RP).
The total residual volume of process fluid in the circuit
(VR) is calculated by substituting the above surface areas
and R values into Equation 1b:

VR = SAW RW + SAD RD + SAP RP


table III: Cumulative Dilution of the Process Fluid for Intermediate Cleaning
= (20.4 m2) (6.60 mL/m2) + (4.24 m2) (26.5 mL/m2) + Steps.
(3.7 m2) (26.5 mL/m2)
The above step dilution factors for intermediate steps
= 345 mL can be used to simulate intermediate cleaning steps at
small scale. For example, if the pre-rinse and alkaline
The dilution of the process soil for each cleaning step wash steps are to be simulated for a product inactivation
(DS) and the cumulative dilution (DC) are calculated from study (16, 17), the experimental parameters should be set
the residual volume of process fluid calculated above and to limit the dilution of the product to less than 3.8 x 105
the volumes of cleaning solution given in Table II. DS and for these steps. This would ensure that the inactivation
DC for the pre-rinse and alkaline wash steps are calcu- rate of the product at small scale for these steps repre-
lated as follows: sents a worst-case condition relative to that at full scale.
Pre-rinse (PR): This is because for a given pH, temperature, and shear

Special edition: Cleaning Validation 9


Rizwan Sharnez

rate, the inactivation rate of the product is determined by SA Surface area of equipment that comes into contact
the concentration of cleaning agent and the concentra- with process fluid
tion of the product, both of which depend on cumulative SAD Surface area of bottom dish of vessel
dilution. SAP Surface area of vessel piping, filter housings, and
Note that the above dilutions are based on the residual other miscellaneous components
volumes for 56% glucose, a relatively viscous process SAW Surface area of vessel side wall that is wetted by
soil. This represents a worst-case condition from the the process fluid
standpoint of estimating the residual volume (VR). Since VR Residual volume of process fluid in equipment
cleaning is typically performed with relatively dilute VS Volume of cleaning solution
aqueous solutions, VR for most cleaning solutions would
be substantially smaller, and therefore the dilution of the REFERENCES
process fluids during cleaning would be commensurate- 1. R. Sharnez., et al, Industry Comes Clean at PDA Annual Meet-
ly higher. Thus, the step and cumulative dilution factors ing, PDA Letter XLVIII (7), 28-32, July/Aug 2012.
2. B. Hoist, Developing a Cleaning Process: Cleaning in Develop-
for most CIP systems are likely to be much higher than
ment, Journal of GXP Compliance 10 (3), 2006.
those estimated in this example. 3. R. Sharnez, Strategies for Developing a Robust Cleaning Process
Part I: Application of Quality by Design to Cleaning, American
Conclusion Pharmaceutical Review 13 (5), 77-80, 2010.
A methodology for estimating the residual volume and 4. R. Sharnez, Validating for the Long Haul, Journal of Validation
dilution of the process fluid during cleaning was de- Technology 14 (5), 2008.
5. R. Sharnez, and M. VanTrieste, Quality-by-Design for Cleaning
scribed. Depending on the viscosity of the process fluid
Validation, in Cleaning and Cleaning Validation 1, (2009) Davis
and the angle of repose, the residual volume of a process Healthcare International & PDA.
fluid (R) that remains on the equipment surface after 6. R. Sharnez and L. Klewer, Strategies for Developing a Robust
drainage was estimated to be between 4.3 and 26.5 mL/ Cleaning Process Part II: Demonstrating Cycle Effectiveness,
m2. These estimates are applicable only to systems with American Pharmaceutical Review - Digital Edition 15 (3), 2012.
negligible pooling of the process fluid. Systems without 7. R. Sharnez, Taking the Guesswork out of Validation, Journal of
Validation Technology 14 (3), 2008.
dead-legs and with surfaces sloped at an angle of at least
8. R. Sharnez, Master Soils for Cleaning Cycle Development and
5 generally meet this criterion. Validation: A Case Study, Cleaning and Cleaning Validation 2(2013)
Davis Healthcare & PDA.
Based on the worst-case R value of 26.5 mL/m2 and 9. A. Canhoto, A Novel Bench Scale Apparatus to Model and
typical rinse and wash volumes, the cumulative and step Develop Biopharmaceutical Cleaning Procedures, Journal of
dilution of the process fluid during cleaning were esti- Validation Technology 11 (4), 2004.
10. R. Sharnez et. al, In Situ Monitoring of Soil Dissolution Dynam-
mated to be on the order of 1016 and 105, respectively.
ics: A Rapid and Simple Method for Determining Worst-case Soils
The cumulative dilution factor is used to estimate the for Cleaning Validation, PDA Journal of Pharm. Sc. & Tech.; 58 (4),
concentration of an impurity in the final rinse. The step 203-214, 2004.
dilution factors are used to simulate worst-case cleaning 11. P. Pluta, Laboratory Studies in Cleaning Validation, Journal of
conditions for intermediate cleaning steps. Validation Technology 13 (4), 2007.
Methodologies for estimating fluid velocity and soil 12. R. Sharnez, Leveraging Small-Scale Models to Streamline Valida-
tion, Journal of Validation Technology 14 (4), 2008.
load for small-scale cleaning characterization studies will
13. R. Sharnez and L. Klewer, Parametric Release for Cleaning, Part
be described in subsequent parts of this series. I: Process Characterization, Journal of Validation Technology (14) 8,
30, 2009.
ARtICLE ACRoNYMS LIStING 14. R. Sharnez and M. Monk, Strategies for Enhancing the Perfor-
CLC Cleaning characterization mance of Cleaning Processes Part I: A Framework for Assessing
d Diameter of vessel Performance, Journal of Validation Technology 17 (1), 36-39, 2011.
15. Surface Area of Tank Heads, Mid-states Mechanical Services, Inc.,
DC Cumulative dilution of process fluid for multiple
available at: http://www.mid-statesmechanical.com/pdf/Tank%20
cleaning steps Heads%20Surface%20Area.pdf.
DS Step dilution of process fluid for an intermediate 16. R. Sharnez, et al, Methodology for Assessing Product Inacti-
cleaning step vation during Cleaning Part I: Experimental Approach and
DI Deionized Analytical Methods, Journal of Validation Technology 18 (4), 42-45,
h Height up to which the process fluid wets the side 2012.
17. R. Sharnez, J. Bussiere, D. Mytych, A. Spencer, A. To, and A.
wall
Tholudur, Acceptance Limits for Inactivated Product based on
R Residual process fluid per unit surface area Gelatin as a Reference Impurity, Journal of Validation Technology
RD R for vessel bottom dish 19 (1), 2013, available at: http://www.ivtnetwork.com/article/
RP R for miscellaneous parts of vessel biopharmaceutical-cleaning-validation-acceptance-limits-inacti-
ppq Parts per quintillion (one part per 1018) vated-product-based-gelatin-re.

Originally published in the Spring 2011 issue of Journal of GXP Compliance

10 Special edition: Cleaning Validation


Rizwan Sharnez

Experimental Parameters
for Small-Scale Cleaning
Characterization. Part II:
Effect of Fluid Velocity on
the Kinetics of Cleaning | IVT
Rizwan Sharnez, Ph.d., Angela to, S. Ravi Annapragada, Phd

ABStRACt
Methodologies for estimating experimental parameters for small-scale clean-
ing characterization are described in this series: dilution of process fluids
during clean-in-place (CIP) operations was discussed in Part I (1); the effect of
fluid velocity on the kinetics of cleaning is described in this part; the effect of
humidity, hold time and soil load on cleanability will be discussed in Part III.
The kinetics of cleaning under worst-case conditions is modeled from first
principles. The model is based on diffusion-controlled mass transfer in a
laminar falling film, which typifies worst-case cleaning conditions for CIP
operations. The effect of flowrate per unit width (Q/W) and fluid velocity
(V) on mass transfer rate in film flow is characterized. An experimental ap-
proach for optimizing Q/W and V for identifying worst-case soils for clean-
ing validation is described. The model is also used to estimate fluid velocity,
film thickness and Reynolds Number for a range of values of Q/W and angle
of inclination ().
The results indicate that Q/W and V have a relatively weak effect on the
kinetics of cleaning. For instance, when these parameters are doubled, the
mass transfer rate increases only by a factor of 8% and 12%, respectively.
The results also indicate that for 5 < < 90 and Q/W < ~1 gpm/ft (~2
mL/s/cm), the flow would be laminar and the thickness of the film would
be < ~1 mm. Further, under these worst-case conditions, V would be < ~52
cm/sec, which is substantially less than the design criterion for minimum
fluid velocity in pipes and hoses viz. 150 cm/sec (5 ft/s).

INtRodUCtIoN
Small-scale cleaning characterization data can be used to streamline valida-
tion requirements for multiproduct equipment i.e. equipment that is used
to manufacture or clean more than one product. This is accomplished by
ranking process soils associated with a given cleaning circuit based on the
relative cleanability of the soils. The hardest-to-clean or worst-case soil for
the circuit is then used to validate that circuit. This approach obviates the
need to validate the cleaning of every soil associated with a circuit. It also
facilitates the introduction of a new product into an existing multiproduct
facility. If it can be shown that the process soils associated with the new
product are easier to clean than the corresponding soils of the previously
validated product, the new product can be introduced into the facility with-
out revalidating the cleaning procedures (2).
In addition to streamlining validation requirements for multiproduct
equipment (3, 4), small-scale cleaning characterization studies can also be
used to identify suitable cleaning chemistries (5, 6), optimize cleaning pa-
rameters and processes (7, 8) and estimate cleaning times at full scale (9, 10).

Special edition: Cleaning Validation 11


Rizwan Sharnez

Experimental models for small-scale cleaning charac- These criteria are designed to ensure turbulent flow dur-
terization have been described in the literature (11-14). ing CIP; the 5 ft/s criterion for VMIN is also designed to
In these studies, the rate at which the process residue is ensure flooding in pipes and hoses.
removed from the surface i.e., the mass transfer rate For pipes and hoses, the 5 ft/s criterion for VMIN can
is measured under simulated cleaning conditions. A be readily met at all locations and is therefore relatively
critical step in the development of these models is to straightforward to simulate at small scale. For film flows,
identify and scale down the hardest-to-clean (worst-case) however, the flow rate per unit width at the worst-case
location in the equipment (15). The worst-case loca- location (Q/W)WCL such as the underside of an impel-
tion is typically an area with poor circulation, such as a ler blade or a magnetically coupled bottom-mounted
shadowed or occluded area. Note that it is not necessary impeller is likely to be substantially less than (Q/W)
to simulate the entire cleaning process at small scale; MIN. Design and operational variables that contribute to
instead, it is sufficient to simulate the worst-case location (Q/W)WCL being less than (Q/W)MIN are summarized in
within the equipment. If the process residue can be ad- Table 1.
equately removed from the worst-case location, it follows
that it can also be adequately removed from other loca-
tions in the equipment. Thus, with this approach, the
cleaning times obtained at small scale would be indica-
tive of those at full scale, provided that there is adequate
spray coverage at all surfaces that need to be cleaned,
and the worst-case location is appropriately identified table I: Design and operational variables that contribute to the flow rate
and simulated at small scale. per unit width at the worst-case location (Q/W)WCL being substantially less
The scalability of small-scale cleaning characteriza- than the recommended minimum operating value (Q/W)MIN. For vessels, the
recommended value for (Q/W)MIN is 2.5 gpm per foot of
tion data depends on the accuracy with which relevant vessel circumference.
experimental parameters are estimated. Experimental
parameters for simulating the worst-case location fall into Effect of Flowrate and Fluid Velocity on Kinetics and
two distinct categories (1): Cleaning
Parameters that can be readily determined from pro- The mass transfer rate from a stationary surface into a
cess data such as: laminar falling film of a Newtonian fluid was investi-
Parameters that can be readily determined from gated by Kramers and Kreyger for a flat rectangular layer
equipment and process data, such as: (17, 18) and by Blount for viscous drops (19, 20). Their
o Material of construction and surface characteristics results indicate that the mass or molar flux (NAX) of the
such as roughness and curvature of coupons or solute (A) into the fluid (X) is a function of the solubil-
parts used to simulate large-scale equipment; ity (SAX) and diffusivity (DAX) of the solute in the fluid;
o Post-soiling parameters such as hold time, and the density (), dynamic viscosity () and flowrate per
ambient temperature and humidity; and, unit width (Q/W) of the fluid; and the length (L) of the
o Cleaning parameters such as rinse or wash time, surface along the direction of the flow (Figure 1):
temperature of rinse solvent, and temperature and
concentration of cleaning solution. NAX = k SAX (DAX L)2/3 (Q/W)1/9 ()-2/9 Equation [1a]

Parameters that typically need to be determined from where is /, the kinematic viscosity; and k is a con-
first principles, such as: stant that includes the acceleration due to gravity (g), the
o Dilution of the process fluid during cleaning (i.e. angle of inclination (), and the width (W).
soil to rinse solvent or cleaning solution ratio).
o Velocity of rinse solvent or cleaning solution at the
worst-case location the subject of this paper.
o Soil load.

Flowrate and Fluid Velocity at Worst-Case Location


Engineering standards provide guidelines for setting the
minimum average velocity or minimum volumetric flow
rate of rinse solvent or cleaning solution in various sec-
tions of a CIP circuit. For instance, it is recommended
that for pipes and hoses the minimum fluid velocity
VMIN be 5 ft/s (1.5 m/s), and for film flows in vessels Figure 1: Mass transfer in gravity-driven film flow: Solute A diffusing
the minimum flow rate per unit width (Q/W)MIN be into a laminar falling film of fluid X, moving with a fully developed para-
2.5 gpm per ft of vessel circumference (31 L/min/m) (16). bolic velocity profile.

12 Special edition: Cleaning Validation


Rizwan Sharnez

For a surface of given length, and a given fluid viscosity An experimental approach for optimizing flowrate
and angle of inclination, and fluid velocity for evaluating relative cleanability at
small scale is described in the next section.
NAX (SAX DAX2/3) (Q/W)1/9 Equation [1b]
Flow Rate and Fluid Velocity for Evaluating Relative
Further, for laminar falling films, Cleanability
Consider a system that is validated to manufacture and
Q/W V3/2 clean product A. A new product (B) needs to be manu-
factured and cleaned in the same equipment. A small-
where V is the average fluid velocity (21); thus, scale study is performed to evaluate the cleanability of
A relative to that of B. If A is harder to clean than B, the
NAX (SAX DAX2/3) V1/6 Equation [1c] new product could be introduced without revalidation.
The objective is to determine the optimum flowrate per
The flux (NAX) of the solute into the fluid is the rate at unit width (Q/W) and fluid velocity (V) for the small-
which the solute is removed from the surface. Thus, NAX scale study.
is effectively a measure of the kinetics of cleaning. Note For products A and B, Equation 1b can be written as
that the effect of Q/W and V on the mass transfer rate follows:
and therefore the kinetics of cleaning is relatively weak.
For example, when Q/W or V is doubled, the mass NAX (SAX DAX2/3) (Q/W)1/9 Equation [2a]
transfer rate increases only by a factor of 21/9 (8%), and
21/6 (12%), respectively. NBX (SBX DBX2/3) (Q/W)1/9 Equation [2b]

Equation 1 is valid when (1) the Reynolds Number Re Thus,


= 4(Q/W)/ < 1500, the criterion for laminar flow in a NAX / NBX = (SAX DAX2/3) / (SBX DBX2/3) Equation [2c]
falling film; (2) the velocity profile in the falling film is
fully developed, a condition that holds when L >> , the Since the cleaning time (t) is inversely proportional to
thickness of the film; and (3) the distance over which the the mass transfer rate (N), the cleanability of B (tB) rela-
solute diffuses into the film (d) is << , and as a result, tive to that of A (tA) can be expressed as
the velocity profile for 0 < y < d can be approximated
as a linear function of distance from the surface being tB / tA = NAX / NBX = (SAX DAX2/3) / (SBX DBX2/3) Eq [2d]
cleaned (y) (Figure 1). In terms of the parameters in
Equation 1, the third condition is satisfied when the Equation 2d indicates that relative cleanability (tB/
solubility (SAX) and/or diffusivity (DAX) of the solute in tA) depends on the physical properties of A and B (viz.
the fluid are low enough for the mass transfer to be diffu- the solubility and diffusivity of A and B in the fluid).
sion controlled. Further, relative cleanability is independent of Q/W, and
The above conditions would be satisfied at the therefore of V. Thus, if the objective of the study is to
worst-case location in the equipment for a process soil rank A and B based on cleanability, Q/W and V can be
that is difficult to clean, as this represents a worst-case set to any reasonable value, provided that the resulting
scenario from the standpoint of cleaning viz. diffusion- flow is laminar in this case Re = 4(Q/W)/ < ~1000.
controlled mass transfer in a laminar falling film. Thus, In practice, however, a lower value of Q/W is preferable
Equation 1 can be used to characterize the effect of because the absolute difference between tA and tB (tAB)
process parameters such as Q/W and V on the kinetics is amplified, which in turn makes it commensurately
of cleaning under worst-case conditions and for design easier to differentiate between the two soils based on the
purposes. It should also be noted that since Equation larger magnitude of tAB. If necessary, tAB can be am-
1 is derived for mass transfer of a single component (A) plified by reducing Q/W up to the point where the film
into a pure solvent (X), its applicability to complex multi- is still intact and uniform, i.e. it does not disintegrate into
component process soils or solvents containing formulat- slower-moving unsteady drops.
ed cleaning agents would require the use of an effective An equation for estimating fluid velocity of a laminar
solubility and diffusivity in the cleaning solution. Con- falling film from Q/W and is derived in the next sec-
sequently, Equation 1 cannot be readily used to predict tion.
the cleanability of multicomponent soils; nonetheless, it
can be used to characterize the effect of process param- Estimation of Fluid Velocity in a Laminar Falling Film
eters on the kinetics of cleaning, and to thereby identify A laminar falling film of a Newtonian fluid flowing pri-
and establish meaningful operating ranges for critical marily under the influence of gravity is shown in Figure
process parameters. Further, this equation can also be 1. The flow is delineated as a thin sheet of liquid flowing
used to develop experimental models for cleaning (2). down an inclined flat plate of length L and width W. As

Special edition: Cleaning Validation 13


Rizwan Sharnez

the liquid flows down the plate, it forms a film of thick- mm, if the film were stable (i.e. if it did not disintegrate
ness and develops a parabolic velocity profile, with the into unsteady drops). Further, under these conditions,
maximum velocity at the film surface. For this type of the average velocity of the fluid would be < ~52 cm/sec,
flow, the average fluid velocity (V) and film thickness () which is substantially less than the design criterion for
can be expressed as follows (21): VMIN in pipes and hoses viz. 150 cm/sec (5 ft/s).

Equation 3

Equation 4

Where is the density of the liquid, g is acceleration due


to gravity, is the angle of, is the dynamic viscosity of
the fluid, is the mass flow rate, and W is the width of
the film.
Equations 3 and 4 can be combined to eliminate and
express V in terms of measurable parameters:

Equation 5

Where Q is /, the volumetric flow rate, and is /,


the kinematic viscosity of the fluid.
table II: Average velocity, film thickness, Reynolds Number and relative
Equation 5 is valid under the following conditions: (1)
mass transfer rate in a laminar falling film for a range of values of flowrate
when edge effects are negligible, a condition that is valid and angle of inclination.
when L and W are >> ; and (2) when viscous forces are
large enough to prevent continued acceleration of the
liquid along the length of the plate i.e., at a low Reyn- CoNCLUSIoN
olds Number, when the flow is laminar. Under these Small-scale experimental models are used to determine
conditions, V is independent of the distance traversed worst-case soils for cleaning validation, and estimate
along the incline (L). Note that at the worst-case location cleaning times and other performance parameters. A
the above conditions would be satisfied because (a) the critical step in the development of these models is to
surfaces being cleaned are relatively large, and thus L and identify and scale down the hardest-to-clean or worst-
W would be >> ; and (b) the flow would be laminar. case location in the equipment. For cleaning operations,
The Reynolds number (Re) is used to classify a fall- the worst-case location is typically an area within the
ing film into three flow regimes: (a) laminar flow with equipment with poor circulation, such as a shadowed or
negligible rippling (Re < 20); (b) laminar flow with occluded area. Examples of such locations include the
pronounced rippling (20 < Re < 1500); and (c) turbulent underside of a probe or an impeller blade where there
flow (Re > 1500). When Re is less than 20, the ripples is no direct or indirect impingement of the cleaning
are very long and grow slowly down the surface of solution during CIP operations. Instead, the flow of the
the film. As Re increases above 20, the ripple growth fluid at the worst-case location is in the form of a laminar
increases rapidly. Because of the assumptions made falling film.
in developing the above model (21), the error in using A mathematical model for diffusion-controlled mass
Equation 5 to estimate velocity increases with ripple transfer in a laminar falling film was used to character-
growth and Re. The velocities estimated using Equa- ize the effect of flowrate per unit width (Q/W) and fluid
tion 5 have been shown to be in good agreement with velocity (V) on the kinetics of cleaning under worst-case
experimentally observed velocities when Re is less than conditions. The results indicate that the effect of these
1000 (22, 23). parameters on the rate of mass transfer and therefore
The average velocity of the cleaning solution is esti- the kinetics of cleaning is relatively weak. The mass
mated from the flowrate per unit width (Q/W) and the transfer rate increases only by a factor of 8% and 12%,
angle of inclination () using Equation 5. The estimates respectively, when Q/W or V is doubled.
are based on the kinematic viscosity () of water at 20C An experimental approach for optimizing Q/W and
(0.01 cm2/s), acceleration due to gravity (g) of 981 cm/ V for evaluating relative cleanability at small scale was
s2, and a range of values of Q/W and . The results, described. Relative cleanability was shown to depend on
summarized in Table 2, indicate that for 5 < < 90 the solubility and diffusivity of the soils being compared.
and Q/W < ~1 gpm/ft (~2 mL/s/cm), the flow would be Further, for diffusion-controlled mass transfer which
laminar and the thickness of the film ( would be < ~1 typifies worst-case cleaning conditions for CIP opera-

14 Special edition: Cleaning Validation


Rizwan Sharnez

tions relative cleanability was found to be independent REFERENCES


of Q/W and V. Thus, if the objective of the study is to 1. R. Sharnez, A. To, and A. Tholudur, Experimental Parameters for
rank coils based on cleanability, Q/W and V can be set Small-scale Cleaning Characterization Part I: Dilution of Process
Fluids during Cleaning, Journal of Validation Technology 19 (3),
to any reasonable value, provided that the resulting flow
2013.
is laminar. In practice, however, a lower value of Q/W 2. R. Sharnez, Leveraging Small-Scale Models to Streamline Valida-
is preferable because the absolute difference between the tion, Journal of Validation Technology 14 (4), 2008.
cleaning times of the soils (tAB) is amplified, and as a 3. R. Sharnez, Taking the Guesswork out of Validation, Journal of
result, the ability to differentiate between the soils based Validation Technology 14 (3), 2008.
on the larger magnitude of tAB is enhanced commen- 4. R. Sharnez, Master Soils for Cleaning Cycle Development and
Validation: A Case Study, Cleaning and Cleaning Validation 2(2013)
surately. If necessary, tAB can be amplified by reducing
Davis Healthcare & PDA.
Q/W up to the point where the film is still intact and 5. R. Sharnez, et al., Industry Comes Clean at PDA Annual Meet-
uniform i.e. it does not disintegrate into slower-moving ing, PDA Letter XLVIII (7), 28-32, July/Aug 2012.
unsteady drops. 6. B. Hoist, Developing a Cleaning Process: Cleaning in Develop-
The laminar falling film model was also used to esti- ment, Journal of GXP Compliance 10 (3), 2006.
mate fluid velocity (V), film thickness () and Reynolds 7. R. Sharnez, Strategies for Developing a Robust Cleaning Process
Part I: Application of Quality by Design to Cleaning, American
Number (Re) from Q/W and the angle of inclination
Pharmaceutical Review 13 (5), 77-80, 2010.
(). The results indicate that for 5 < < 90 and Q/W 8. R. Sharnez, Validating for the Long Haul, Journal of Validation
< ~1 gpm/ft (~2 mL/s/cm), the flow would be laminar Technology 14 (5), 2008.
and would be < ~1 mm, if the flow was stable. Further, 9. R. Sharnez and M. VanTrieste, Quality-by-Design for Cleaning
under these conditions, V would be < ~52 cm/sec, which Validation, in Cleaning and Cleaning Validation 1, (2009) Davis
is substantially less than the design criterion for VMIN in Healthcare International & PDA.
10. R. Sharnez and L. Klewer, Strategies for Developing a Robust
pipes and hoses viz. 150 cm/sec (5 ft/s). The calculated
Cleaning Process Part II: Demonstrating Cycle Effectiveness,
values of V have been shown to be in good agreement American Pharmaceutical Review - Digital Edition 15 (3), 2012.
with experimentally observed velocities when Re is less 11. Canhoto, A Novel Bench Scale Apparatus to Model and Develop
than 1000. Biopharmaceutical Cleaning Procedures, Journal of Validation
Technology 11 (4), 2004.
SYMBoLS ANd ACRoNYMS 12. R. Sharnez et al, In Situ Monitoring of Soil Dissolution Dynam-
ics: A Rapid and Simple Method for Determining Worst-case Soils
CIP Clean-in-place
for Cleaning Validation, PDA Journal of Pharm. Sc. & Tech. 58 (4),
D Diffusivity 203-214, 2004.
g Acceleration due to gravity 13. P. Pluta, Laboratory Studies in Cleaning Validation, Journal of
L Length of object being cleaned Validation Technology 13 (4), 2007.
N Mass or molar flux 14. R. Sharnez and L. Klewer, Parametric Release for Cleaning, Part
Q Volumetric flow rate I: Process Characterization, Journal of Validation Technology (14) 8,
30, 2009.
Re Reynolds number
15. R. Sharnez and M. Monk, Strategies for Enhancing the Perfor-
S Solubility mance of Cleaning Processes Part I: A Framework for Assessing
t Time Performance, Journal of Validation Technology 17 (1), 36-39, 2011.
V Average velocity of cleaning solution 16. ASME Bioprocessing Equipment (BPE) Standard, 2012.
W Width of laminar falling film 17. Kramers, H., and P. J. Kreyger, Mass Transfer between a Flat
Angle of inclination (slope) Surface and a Falling Film, Chemical Engineering Science, Vol. 6,
pp. 42-48 (1956).
Film thickness
18. R. Bird, W. Stewart, and E. Lightfoot, Transport Phenomena, 2nd
Dynamic Viscosity Ed., 2007. p. 562-563.
Kinematic viscosity 19. M. Blount, Aspects of advection-diffusion-reaction flows of
Density relevance to decontamination, KTN Internship Report, (2010).
Mass flow rate 20. J. Landel, H. McEvoy, and S. Dalziel, Cleaning of Viscous Drops
on a Flat Inclined Surface Using Gravity-Driven Film Flows,
Food and Bioproducts Processing, Vol 93, p. 310-317 (2015).
SUBSCRIPtS
21. R. Bird, W. Stewart, and E. Lightfoot, Transport Phenomena. 2nd
A Product A Ed., 2007. pg.46.
B Product B 22. West and Cole, Surface velocities of thin liquid films, Chemical
MIN Minimum Engineering Science, 22, 1388-1389, 1967.
WCL Worst-case location 23. S. Portalski, Velocities in film flow of liquids on vertical plates,
X Fluid X Chemical Engineering Science 19, 575-582, 1964.

Special edition: Cleaning Validation 15


Rizwan Sharnez

Methodology for Assessing


Product Inactivation during
Cleaning Part I: Experimental
Approach and Analytical
Methods | IVT
Rizwan Sharnez, Ph.D

ABSTRACT
For multiproduct cleaning validation, the conventional approach for setting
an acceptance limit for the process residue is based on the maximum allow-
able carryover (MAC) of the active pharmaceutical ingredient (API) (de-
pending on the process soil, API refers to the active pharmaceutical ingredi-
ent in the drug product, drug substance, or drug substance intermediate).
However, if the API becomes pharmacologically inactive during cleaning
the acceptance limit does not need to be based on active product. This is
an important consideration in biopharmaceutical manufacturing because
the cleaning conditions are generally aggressive enough to inactivate the
product.
The experimental approach and analytical methods for assessing
inactivation of the API during cleaning are described in Part I. A rational
approach for setting safety-based acceptance limits for inactivated product
and process residuals is described in Part II. The scope of this paper is
limited to biopharmaceutical cleaning processes; nonetheless, the under-
lying concepts may be useful in designing inactivation studies and setting
acceptance limits for other types of pharmaceutical cleaning processes.

INTRODUCTION
An important regulatory expectation for multiproduct cleaning validation is
to demonstrate that potential carryover of the previously manufactured API
(Product A) into the subsequently manufactured product (Product B) is be-
low an acceptable level. This criterion is often assessed through a maximum
allowable carryover (MAC) calculation for the previously manufactured API
(1-5). The MAC calculation is typically based either on the minimum thera-
peutic dose (1), or the acceptable daily exposure (ADE) (2) of the previously
manufactured API.

Limitations of the MAC Approach


A limitation of the conventional MAC approach is that it is based on the
assumption that the product is active after the cleaning. This has important
implications for biopharmaceutical manufacturing because the API is often
inactivated by the cleaning process (6, 7).
Another limitation of the MAC approach is that the calculated accep-
tance limits are often below the limit of quantitation (LOQ) of non-
specific analytical methods, such as total organic carbon (TOC). The
LOQ of TOC- based methods is typically between 0.05 and 0.2 ppm. The
large surface areas and small batch sizes involved in biopharmaceutical
manufacturing further exacerbate this issue. Product specific immunoas-

16 Special edition: Cleaning Validation


Rizwan Sharnez

says (PSIA) such as ELISA and EIA have been used worst-case cleaning conditions at bench scale (10, 11).
to address this issue; the LOQ of most PSAs is on The results of the bench-scale studies can justifiably be
the order of 10 ppb. PSIAs detect activity indirectly extrapolated to the full-scale cleaning process. That is
by recognizing specific epitopes (short sequences of because under worst-case cleaning conditions of laminar
amino acids ) in the API; however, epitopes are known flow and low shear rate fragmentation and inactivation
to degrade during cleaning, and thus the results can be are independent of scale (i.e., they depend on cleaning
misleading (8, 9). parameters that are not a function of the of spatial coor-
Other limitations of the MAC approach are dis- dinates of the system, such as time, temperature, concen-
cussed in the literature (9). tration, and the ratio of cleaning solution to process soil).
The bench-scale experiments are typically per-
PRODUCT INACTIVATION APPROACH formed in a vial or dialysis cassette, and are designed
With the product inactivation approach, if the API is in- to simulate full-scale cleaning conditions that are least
activated during cleaning, the acceptance limits may be conducive (worst-case) for inactivation. For example,
set based on the inactivated product instead of the API. for a chemical wash, the lowest applicable concentra-
The product inactivation approach is therefore more re- tion of cleaning agent, temperature, duration, and ratio
flective of the phenomenological aspects of the cleaning of cleaning solution to residual process soil should be
process. Additionally, the acceptance limits based on in- considered in simulating the cleaning cycle at bench
activated product are very unlikely to be below the LOQ scale. Other operating parameters that can contribute
of TOC (refer to Part II). Thus, the product inactivation to product inactivation include dirty hold time (DHT)
approach alleviates the limitations of the MAC approach and associated drying conditions (humidity and air
described in the previous section. circulation rate), and shear rate due to impingement
The methodology described in Part I includes and turbulence.
experimental simulation of the cleaning processes at An operating parameter or step can be eliminated
small scale and analytical methods to evaluate inacti- from the experimental design if its elimination repre-
vation of the API during cleaning. A rational approach sents a worst-case scenario from the standpoint of in-
for setting safety-based acceptance limits is described activation. This approach can be leveraged to simplify
in Part II. the bench-scale studies. For instance, if it is reasonable
to assume that product inactivation increases with
PROPOSED METHODOLOGY shear rate, then it can be eliminated from the experi-
Inactivation of the product during cleaning has impor- mental design (i.e., the shear rate need not be simu-
tant implications for cleaning validation of multiproduct lated in the experiment). Similarly, the ratio of cleaning
equipment. If it can be demonstrated that the product solution to process soil can be reduced, and the acid
becomes pharmacologically inactive during cleaning, wash and rinse steps can be eliminated to minimize
there is limited value in verifying removal of the active dilution of the process soil, and facilitate detection of
ingredient. Instead, it is more appropriate to demonstrate the process residue in the sample. When making such
that the inactivated product has been removed below a changes, unexpected effects such as aggregation of the
predefined acceptance limit. This is consistent with the API can occur. It is therefore important to make sure
expectation that the carryover of an extrinsic impurity that the modifications do not result in experimental
into a subsequent batch should be justified from the artifacts.
standpoint of the safety and efficacy of the product. It If the cleaning cycles are being developed or modi-
also obviates the need to develop PSIAs for cleaning fied, the inactivation study should be designed to
validation. evaluate the effect of key operating parameters on the
Biopharmaceutical cleaning cycles are generally fragmentation and inactivation rate of the API. This in-
designed to expose product contact equipment to formation, together with data from cleanability studies
extremes of pH (<2 and >13) and temperature (60- (12, 13), can be used to identify cycle parameters that
80C) for several minutes. Under these conditions are effective in inactivating the API.
monoclonal antibodies, therapeutic proteins, and For existing cleaning cycles, the cleaning conditions
other biopharmaceuticals are known to degrade and for the inactivation study should be based on worst-
denature rapidly, and are therefore likely to become case operating parameters for all systems involved. For
pharmacologically inactive (6, 7). The product inac- instance, if different systems are cleaned with differ-
tivation approach should therefore be considered for ent cleaning solutions and at different temperatures,
biopharmaceutical cleaning validation. then the study should be performed with the mildest
cleaning solution, at the lowest cleaning agent concen-
GUIDANCE FOR DESIGNING INACTIVATION STUDIES tration, and the lowest temperature, if these conditions
Fragmentation and inactivation of an API during clean- are least conducive for inactivation. Further, for clean-
ing can be assessed by exposing the process soil to in-place (CIP) systems with multiple toggle paths, the

Special edition: Cleaning Validation 17


Rizwan Sharnez

duration of cleaning should be based on the toggle (binding sites that are functionally intact), such as a
path with the shortest cleaning time. bioassay. These methods measure the relative amount
After exposing the process soil to worst-case clean- of biologically active product. Thus, they can be used
ing conditions, the samples are titrated to a neutral to evaluate the degree of inactivation of the API during
pH, and cooled to 4C to minimize further fragmen- cleaning.
tation and inactivation of the API. The samples are A non-specific method such as total organic carbon
then subjected to analytical testing as described in the (TOC) can be used to detect target impurities in clean-
next section. If the API is inactivated when exposed to ing validation samples. TOC has an LOQ of approxi-
worst-case cleaning conditions, then the acceptance mately 0.2 ppm, which is sufficient for most cleaning
limit for the inactivated product can be set based on applications. The use of a non-specific method allows
the approach described in Part II. for the detection of both intact API and inactivated
If the results indicate that the API is not inactivated product.
during cleaning, then the acceptance limits should be With the above methods, appropriate standards and
set based on the acceptable carryover of the API (1-2). untreated controls should be included to provide a
If the API is partially inactivated, then the acceptance basis for comparison, and to assess the impact of any
limits should be determined for the API, as well as for experimental artifacts and potential matrix effects.
the inactivated product, and the lower of the two limits For SDS-PAGE, appropriate MW markers should be
should be used. Alternatively, the cleaning parameters included to facilitate comparison of the fragments to
can be modified to ensure inactivation of the API. This the untreated controls.
can be facilitated by running additional studies to
characterize the effect of specific cleaning parameters CONCLUSION
on the API. An important consideration in multiproduct cleaning
validation is to demonstrate that the carryover of the
RECOMMENDED ANALYTICAL METHODS previously manufactured API into a batch of the subse-
Analytical methods commonly used to evaluate the effect quently manufactured product is below an acceptable
of cleaning parameters on the previously manufactured limit. This criterion is often met through a MAC assess-
API are described in this section. These methods are ment for the API; however, if the previously manufac-
used to evaluate fragmentation and inactivation of the tured API becomes pharmacologically inactive during
API at bench scale, and to detect target impurities (in this cleaning, the acceptance limit does not need to be based
case, the previously manufactured API and/or inactivated on active product. This is an important consideration in
product in the process residue) in cleaning validation biopharmaceutical manufacturing because the cleaning
samples. The analytical results are used to understand conditions are generally aggressive enough to inactivate
the impact of the cleaning conditions on the process soil, the product.
and to set rational safety-based acceptance limits for the Fragmentation and inactivation of an API during
target impurities (Part II). The detection methods are cleaning can be assessed by exposing the process
used to verify that the concentrations of target impurities soil to simulated cleaning conditions at bench scale
in cleaning validation samples are below their respective (10, 11). The bench scale experiments are typically
acceptance limits. designed to simulate full-scale cleaning conditions that
Sodium dodecyl sulphate polyacrylamide gel elec- are least conducive (worst case) for inactivation. The
trophoresis (SDS-PAGE) or capillary electrophoresis degree of inactivation is evaluated by subjecting the
(CE) are generally used to characterize fragmentation sample and untreated controls to the appropriate as-
of the API during cleaning. For SDS-PAGE, a 4 to 20 says, (e.g., SDS-PAGE and bioassay are used to evaluate
percent gradient corresponds to a molecular weight fragmentation and biological activity, respectively). The
range of 4 to 250 kDa, which is sufficient for most results of the study are used to set appropriate accep-
biological APIs. While CE provides greater sensitivity, tance limits for cleaning validation.
lower variability due to the absence of staining, and If the API is inactivated during cleaning, the accep-
high throughput capability as compared to SDS-PAGE, tance limits for the process residuals can be set based
both methods are adequate for demonstrating distinct, on the approach described in Part II.
size-based separation of fragmented protein. Size
exclusion high-pressure liquid chromatography (SE- ACKNOWLEDGEMENTS
HPLC) can also be utilized for size-based separation of We thank Arun Tholudur, Aine Hanley, and Sam Guhan
protein fragments; however, it can be difficult to obtain of Amgen; Rich Kemmer and Paul Whetstone of Bayer;
a distinct size-based separation across a wide range of James Crawford and Michael Maurer of GlaxoSmith-
fragment sizes. Kline; John Krayer of Janssen; Josh Getchell of Lonza;
Inactivation of the API can be evaluated by meth- David Barabani and Michael Parks of Pfizer; and Markus
ods that measure loss of biological activity or function Bluemel of Novartis for their help and support.

18 Special edition: Cleaning Validation


Rizwan Sharnez

REFERENCES parable Quality Approach to Intrasite Assessments, Journal of


1. G.L. Fourman and M.V. Mullen, Determining Cleaning Valida- Validation Technology 18 (2), 17-25, 2012.
tion Acceptance Limits for Pharmaceutical Manufacturing 8. Health Canada, Cleaning Validation Guidelines (GUIDE-0028);
Operations, Pharmaceutical Technology 17 (4), 54-60, 1983. Section 8.3, Health Products and Food Branch Inspectorate,
2. ISPE, Risk-Based Manufacture of Pharmaceutical Products: 2008, available at: http://www.hc-sc.gc.ca/dhp-mps/compli-
A Guide to Managing Risks Associated with Cross-Contami- conform/gmp-bpf/validation/index-e....
nation 7, International Society for Pharmaceutical Engineers 9. R. Sharnez, M. Horner, A. Spencer, and A. Tholudur, Leverag-
(ISPE). Tampa, FL, First ed., 2010. ing Acceptable Exposure of Host Cell Protein to Set Acceptance
3. R. Sharnez, Strategies for Setting Rational MAC-based Limits Limits for Inactivated Product, Journal of Validation Technology
Part I: Reassessing the Carryover Criterion, Journal of Valida- 18 (3), 38-44, 2012.
tion Technology 16 (1), 71-74, 2010. 10. K. Kendrick, A. Canhoto, and M. Kreuze, Analysis of Degrada-
4. R. Sharnez, A. To, and L. Klewer, Strategies for Setting Ratio- tion Properties of Biopharmaceutical Active Ingredients as
nal MAC-based Limits Part II: Application to Rinse Samples, Caused by Various Process Cleaning Agents and Temperature,
Journal of Validation Technology 17 (2), 43-46, 2011. Journal of Validation Technology 15 (3), 69, 2009.
5. R. Sharnez, and A. To, Strategies for Setting Rational MAC- 11. N. Rathore, W. Qi, C. Chen, and W. Ji, Bench-scale character-
based Limits Part III: Leveraging Toxicology and Cleanability ization of cleaning process design space for biopharmaceuti-
Data, Journal of Validation Technology 17 (3), 24-28, 2011. cals, Biopharm International 22 (3), 2009.
6. R. Sharnez, and A. To, Cleaning Validation of Multiproduct 12. R. Sharnez, Dont Bet on Quality-by-Chance: Part II Lever-
Equipment: Acceptance Limits for Inactivated Product, Part aging Small-Scale Models to Streamline Validation, Journal of
I The Comparable Quality Approach, Journal of Validation Validation Technology 14 (4), 2008.
Technology 17 (4), 32-36, 2011. 13. R. Sharnez and L. Klewer, Strategies for Developing a Robust
7. R. Sharnez, E. Aisenbrey, J. Bercu, D. Binkley, and A. Tholudur, Cleaning Process Part II: Demonstrating Cycle Effectiveness,
Cleaning Validation of Multiproduct Equipment: Acceptance American Pharmaceutical Review - Digital Edition 15 (3), 2012.
Limits for Inactivated Product, Part II- Application of the Com- JVT

Special edition: Cleaning Validation 19


Rizwan Sharnez

Methodology for Assessing


Product Inactivation During
Cleaning Part II: Setting
Acceptance Limits of
Biopharmaceutical Product
Carryover for Equipment
Cleaning | IVT
By Adam Mott, Bill Henry, Edward Wyman, Greg Randall, Kathleen Bellorado, Markus Blmel, Mary
Ellen Clark, Michael Parks, Ronan Hayes, Scott Runkle, Wendy Luo

ABSTRACT
For multi-product biopharmaceutical facilities, setting the acceptable level of
process residues following equipment cleaning is an important regulatory,
business, product quality, and patient safety consideration. Conventional
approaches for setting an acceptance limit for process residues have been
based on the assumption that the active pharmaceutical ingredient (API)
(depending on the process soil, API refers to the active pharmaceutical in-
gredient in the drug product, drug substance, or drug substance intermedi-
ate) is chemically or functionally intact following the cleaning process. These
approaches include Maximum Allowable Carryover (MAC) Health Based
Exposure Limits and other dose or Permissible Daily Exposure (PDE)-
based limits. The concept for cleaning acceptance limits based on intact
product originated from the manufacturing of small molecule pharmaceu-
ticals (1). In contrast to pharmaceutical small molecules, biopharmaceutical
products are large molecules that are likely to degrade and become inactive
when exposed to cleaning conditions. Therefore, an alternative approach to
setting cleaning acceptance limits for biopharmaceutical products based on
the actual process residues that could potentially be present on production
equipment should be considered.
Part I described the methodology to assess and verify API inactivation
during cleaning (2). In Part II, alternative approaches for setting accept-
able levels of process residue will be described building upon the basis
that API inactivation by the cleaning process has been demonstrated.

INTRODUCTION
When multiple products are manufactured using the same equipment, it is
important to ensure that potential product or process residues from the pre-
viously manufactured batch are removed to an acceptable level to ensure the
subsequently manufactured product will not be impacted. The acceptable
level of carryover has often been based on the active, intact API. However,
for biopharmaceutical products, the API typically degrades and becomes
pharmacologically inactive during cleaning, and therefore the cleaning ac-
ceptance criteria do not need to be based on the concept of intact and active
product. Rather, the cleaning acceptance limit should be based on potential
process residues that have a greater carryover potential founded on phe-
nomenological aspects of the cleaning process. The scope of this paper tar-

20 Special edition: Cleaning Validation


Rizwan Sharnez

gets biopharmaceutical APIs; nonetheless, the underlying final rinse of higher grade water quality, typically Water
concepts may be useful in setting acceptance limits for for Injection (WFI). The volume and flow rate of this
other types of pharmaceutical products where inactiva- rinse are designed to be sufficient to remove all residual
tion during the cleaning process can be demonstrated. cleaning agent(s) to a conductivity level approaching the
This paper will include a review of product inacti- WFI source water. The inactivated product that results
vation, information on product detection using total from exposure to the cleaning conditions is likely to be
organic carbon (TOC), and alternative approaches more water-soluble than the intact protein due to its de-
for setting acceptance limits for equipment cleaning. creased size (3) and, therefore, should be readily rinsed
The intention of this paper is to propose acceptable from equipment surfaces in the last step of the cleaning
approaches for setting cleaning limits for biopharma- process. The rinsibility or ease of removal of inacti-
ceutical process equipment that may be considered. vated product/product fragments may be evaluated in a
However, it should not be considered prescriptive for rinsibility study, where the inactivated product material
what approach is most appropriate or should be used is spiked onto representative coupons and exposed to a
since every production facility, processes, and products worst-case (e.g., no impingement, lower flow rate, etc.)
manufactured are unique. water rinse in comparison to full scale cleaning cycles.
If the worst-case rinse removes the product spike from
PRODUCT INACTIVATION the coupon, it demonstrates that the inactivated product
Biopharmaceuticals are large molecule drug products fragments are not a carryover concern.
(e.g., monoclonal antibodies, therapeutic proteins, etc.) The Product Inactivation Study demonstrates the
that are made in processes using living organisms rather product is not active after exposure to cleaning condi-
than extracted from a native source or by synthesizing tions. The rinsibility study demonstrates that the
compounds. The equipment cleaning cycles are designed potential product fragments created from exposure of
to expose product contact areas to cleaning detergents the product to cleaning conditions are not a carryover
that include alkaline and acidic chemicals. Under these risk. Therefore, setting acceptance limits for equip-
exposure conditions, the high pH in alkaline chemi- ment cleanliness based upon intact product activity or
cals (typically pH >11) and low pH in acidic chemicals potential product fragments would not be reflective of
(typically pH <2) are efficient in hydrolyzing biological the actual residuals that are most likely to be present
peptide bonds, rendering biopharmaceutical products on equipment after cleaning based upon the phenom-
biologically inactive by degradation and denaturation. enological effects of the cleaning process.
If it is demonstrated that the product becomes pharma-
cologically inactive during cleaning, there is no longer a DETECTION OF PRODUCT OR PROCESS RESIDUES
risk of active product carryover and, furthermore, a lim- Most biopharmaceutical process components (e.g.,
ited value in verification of the removal of active product API, host cell proteins, media, and cleaning detergents)
from equipment surfaces. include organic carbon within their composition. The ap-
It should be noted that an antibody-drug-conjugate plication of TOC as the post-cleaning detection method
(ADC) is considered a biopharmaceutical product, for product carryover is considered more stringent than
but it contains an extremely toxic small molecule that a product-specific method as it would detect all process/
attaches to a protein through organic linkers. Due to cleaning residuals containing carbon, including poten-
the functional and toxicological behavior of an ADC tially difficult to remove materials. The TOC analysis
product, specifically the toxic small molecules attached method is relatively sensitive (scale of ppb limits of detec-
to the large molecule, PDE limits should be established tion and quantitation) that can be used for swab samples,
for ADC products based on the toxicity of the conju- rinse samples, and inline monitoring.
gate; therefore, they are not in the scope of this paper. The approaches included in this paper for assessing
equipment cleanliness are based on TOC, but they can
Part I discussed experimental approaches and ana- be adapted to product specific methods if required.
lytical methods that can be used to evaluate product
inactivation by the cleaning detergents. This important SETTING THE ACCEPTANCE LIMITS
first step characterizes the biological activity of the API Four different approaches for setting cleaning acceptance
and may also be used to gain a further understanding limits will be discussed. Each limit setting approach
of remaining product fragments. (Cleaning Process Capability, Safety Factor, Toxicology
Threshold, and Performance Control) ensures patient
Inactivated Product Rinsibility/Removal safety and no impact to subsequent product quality. The
The inactivated product and/or product fragments may assumption inherent in each approach is that prod-
be further evaluated to better understand the effect of uct inactivation from the cleaning process conditions
the cleaning process and the potential for carryover. The has been demonstrated, which provides the scientific
final step in most, if not all, cleaning procedures is a rationale and assurance of no active product carryover.

Special edition: Cleaning Validation 21


Rizwan Sharnez

Every facility has unique characteristics, products, and suite. The worst-case equipment will be the unit with
operational variables to consider. The following ap- the largest surface area to volume ratio.
proaches are not intended to be inclusive of all acceptable The following equation is used to calculate the
approaches to determine cleaning limits. The following limit of TOC contribution on production equipment
approaches may be considered as an alternative to the surfaces that could be from the final WFI rinse. This
MAC approach, which may have limited applicability for limit is determined by calculating the amount of TOC
biopharmaceutical products. on the equipment surface that would not result in TOC
concentration in minimum working volume allowed in
Cleaning Process Capability Approach the equipment that would be greater than the accept-
The cleaning process capability approach sets the accep- able TOC limit of WFI (the final rinse source water):
tance limits for equipment cleaning based on demonstra-
tion that all carbon containing process materials have
been removed to the level that the cleaning process is Maximum Surface Residual TOC (ng TOC/cm2) =
capable. The basis for the cleaning process capability
limit is that equipment surfaces cannot be cleaner than
the potential residual contribution from the last solution
of the cleaning process to contact equipment surfaces. To convert the Maximum Surface Residual TOC
If TOC is used as the most suitable measure to demon- limit into the limit for a swab sample, the following
strate removal of process material, the limit of process equation is applied:
capability of the cleaning process to measure cleanliness Residual TOC Swab Limit = Maximum Surface
would be based on the potential TOC contribution of the Residual TOC (ng TOC/cm2) x SSA (cm2/swab) x 1 g
final WFI rinse. /1000 ng
TOC results from surfaces that are below the clean-
ing process capability limit that cannot be differentiated Where:
from TOC intrinsic to the final WFI rinse or from po-
tentially low levels of residual cleaning agent or process Maximum Surface Residual TOC (ng TOC/cm2):
material. TOC results that are above the cleaning process The maximum amount of residual material that is al-
capability limit would be as a result of residual clean- lowed per square centimeter of production equipment.
ing agent or process material and not from the final SSA (cm2): Swabbed Surface Area, the area which
water rinse. It should be noted that this is a conserva- his swabbed for sample analysis. For example, 5 cm x
tive approach to setting limits for equipment cleaning 5 cm (2 inches x 2 inches) equals 25 cm2.
verification and calculated limits are relatively low. Unit Conversion (ng to g): converts units of ng to
To calculate the TOC surface limit, the following g where 1 g equals 1000 ng.
variables are required: equipment surface area, small-
est volume that the equipment could process (e.g.,
working volume), final rinse (WFI) TOC limit (source Figure 1: Determination of Carryover Limit based
of potential TOC contribution), and swab surface area. on Cleaning Process Capability.
It should be noted that the surface area and volume
are specific to the equipment to be cleaned and not to An actual example of cleaning limit calculations
the entire production train. When the MAC approach using the approach describe above is presented below
is used, there is a concern of a cumulative carryover of (Note: worst-case [tightest] limits will be calculated
active product; which would not be removed through where the production equipment surface area relative
common purification steps of subsequent product
production, which is the reason total surface area of all
equipment in the production train is used (1). Howev-
er, active product is not a concern once the product in-
activation and rinsibility are completed because active,
intact product would not be present after cleaning;
product fragments, just as other non-product proteins
(e.g., HCPs), would be removed during purification,
and product fragments created after cleaning are free
rinsing and easily removed from equipment surfaces.
The cleaning process capability limit may be deter-
mined for each piece of equipment, or the worst-case
piece of equipment in each production suite may be Figure 1: illustrates the approach described above to calculate the residu-
used to set a limit to be used for all equipment in the al TOC limit as measured by a swab sample.

22 Special edition: Cleaning Validation


Rizwan Sharnez

to working volume is large as is typically observed in reduction) is used to set the acceptance limit for a
smaller equipment). product with a concentration of 100 mg/mL and a
molecular makeup of 53% carbon:
Example: 200 L Reactor (150 L minimum working
volume):

Maximum Surface Once the initial acceptance limit has been set based
Residual TOC (ng TOC/cm2) = on the safety factor, the surface area limit can be cal-
culated:
= 2625 ng TOC/cm2
Residual TOC Swab Limit = 2625 ng TOC/cm2 x 25 cm2/
swab x 1 g /1000 ng
= 66 g TOC/swab

Acceptance limits for cleaning equipment set using


the Cleaning Process Capability approach is a conser- Where:
vative limit that ensures removal of all carbon contain- The TOC acceptance limit is in ppm.
ing process residuals and cleaning agents to safe levels. Volume is the amount of desorption solution used in
mL.
SAFETY FACTOR APPROACH The surface area swabbed in cm2.
Continuing from the example above, the calculation
SAFETY FACTOR APPROACH is shown below with a 5 ppm acceptance limit, 25 cm2
This approach is to determine the safety factor involved; swab surface area, and 30 mL desorption solution:
that is to calculate the reduction of the inactivated prod-
uct at the acceptance criteria level as an organic impurity
in the drug substance. This organic impurity limit is
0.10% (4), which is the equivalent to a Safety Factor of
1,000.

Note: The Residual TOC Swab Limit is adjusted, as


necessary, based on surface area sampled where it is
not practical or possible to swab 25 cm2.

Concentration is the amount of active ingredient in TOxICOLOGY THRESHOLD APPROACH


the drug substance/drug product. If it can be demonstrated that the biological products
Fifty-percent represents the approximate amount becomes degraded and inactivated, application of a
of carbon in protein (5). This may also be calculated toxicological threshold of concern (TTC) may be applied
based on the molecular makeup of the API if available. in order to mitigate the risk of process residues (degraded
The initial cleaning acceptance limits are typically and inactivated fragments) affecting the next biophar-
in the range of 1-10 ppm TOC for swab and rinse maceutical produced (6-9). Once an appropriate TTC
samples. An example calculation is shown below; a 2 has been determined based on structural class of process
ppm acceptance limit with a product concentration of residuals, a calculation such as the one below can be
100 mg/mL yields a Safety Factor of 25,000. Since this applied.
is greater than a 1,000 Safety Factor, the 2 ppm accep-
tance limit has been appropriately set to demonstrate
adequate removal of residual active ingredient.

Where:
ARL = Acceptable Residual Limit = g/cm2
TTC = Toxilogical Threshold of Concern = g/day
Another example calculation is shown below, MBS = Minimum Batch Size for Subsequently Manu-
wherein a targeted Safety Factor of 10,000 (i.e., a 4-log factured Product= g

Special edition: Cleaning Validation 23


Rizwan Sharnez

MDD = Maximum Daily Dose for Subsequently Manu- tive of the performance of the cleaning process. The
factured Product = g/day Performance Control Limit, sometimes referred to as an
SA = Surface Area (SSA) = cm2 Alert Limit, enables detection of a change in the perfor-
For example, degraded biopharmaceutical product mance of the cleaning process and allows for a proactive
fragments may be considered to be Class I chemicals investigation into a potential cleaning process issue.
with a residual soil threshold of 100 g/day. A 200 L The Performance Control Limit approach discussed
Final Product Vessel may have a surface area of 28,573 below is based on the TOC data collected during on-
cm2: minimum batch size is 400 g, and maximum going cleaning studies. The evaluation of data should
daily dose is 50,000 g/day. be statistically based and strike an appropriate balance
between sensitivity to data shifts and excessive false
signals. Many standard statistical methods are based
on the assumption of normality and independence of
the data population. The setting of a control limit at
To calculate the TOC limit of a swab sample using three standard deviations from the mean is an appro-
the ARL determined above, the following equation priate approach for setting a control (or performance)
would be used: limit, but it assumes a normally distributed dataset.
Residual TOC Swab Limit = Acceptable Residual A control limit at three standard deviations from the
Limit (g/cm2) x SSA (cm2/swab) x 50% mean ensures a false out-of-tolerance (OOT) rate of
Where: 0.27%. This 0.27% value is referred to as the alpha
Acceptable Residual Limit (g TOC/cm2): The maxi- rate. The problem with the data typically generated
mum amount of residual material that is allowed per from effective cleaning processes is that the data are
square centimeter of production equipment. not normally distributed, as shown in the following
SSA (cm2): Swabbed Surface Area, the area which his example in Figure 2.
swabbed for sample analysis. For example, 5 cm x 5
cm (2 inches x 2 inches) equals 25 cm2.
50%: Represents the approximate amount of carbon in Because the data are not normally distributed, data
protein/protein fragments. transformation techniques such as Box-Cox, mean
Continuing with example above to calculate the scores, reciprocal, negative binomial, etc. are to be
ARL, the following is an example limit for Residual used to normalize data to apply appropriate statisti-
TOC on a swab from production equipment: cal tools to establish an appropriate Performance limit
Residual TOC Swab Limit = 28 g/cm2 x 25 cm x 2 (10). The Box-Cox method is a log transformation that
(g TOC/swab) 50% TOC optimizes the normality of the data set and was used
= 350 g TOC/swab to transform the dataset presented above.
If swab is desorbed in 40 mL of Low TOC Water, The Box-Cox method computes the lambda value to
the measured TOC limit as measured in ppb TOC optimize normality using the following equation:
would be determined by dividing the Residual TOC
Swab Limit by 40 mL and converted to ppb using
the unit conversion of 1000 mL/L as described in the
equation below:
Where Yoriginal is each TOC value, which must be > 0

PERFORMANCE CONTROL LIMIT APPROACH


Performance Control Limits may be considered once
the cleaning validation studies have been completed and
routine cleaning consistently demonstrates the equip-
ment cleaning process removes process residue below
the acceptance limits, especially if the data is consider-
ably lower than the acceptance limit. The Performance
Control Limit approach does not change the level of
carryover that has previously been determined to be
acceptable, but it will establish a limit that is more reflec- Figure 2: Example Histogram of Cleaning Verification Data.

24 Special edition: Cleaning Validation


Rizwan Sharnez

number of 0 results relative to the total number of


results as described in the equation below:

After the review and adjustment for the 0 data


results, the Box-Cox transformation is performed us-
ing the adjusted alpha rate (in the example dataset, the
adjusted alpha rate is 0.0046). Figures 3 and 4, below,
depict the example TOC data that have been trans-
formed using the Box-Cox method.

Figure 3: Box-Cox Transformation.


The top-left histogram describes the distribution of
the original TOC dataset. This dataset is non-normal,
being truncated at zero. The same non-normal phe-
nomenon is displayed in the associated normal prob-
ability plot in the lower-left. The top-right histogram
describes the same data after applying the Box-Cox
transformation. In this case, the data are normally
distributed as evidenced with the normal probability
plot in the lower-right.

The Performance Limits are then back-calculated to


the original scale using the transformed dataset and
the equation below:

Yoriginal = (Ytransformed * lambda + 1)(1/lambda)

Figure 4: Effect of Using the Box-Cox Transformation. The Box-Cox transformed Performance Limit from
the example data is 4876 ppb TOC and is shown in
Figure 5.

Finally, as further cleaning studies are conducted,


additional TOC data will be collected. An appropri-
ate review of the overall dataset should be conducted,
and the performance limits adjusted if performance
changes for reasons that should be well understood.

CONCLUSION
Setting acceptable limits for process residue following
equipment cleaning in multiproduct biopharmaceutical
facilities requires an understanding of each products
composition and the effects of the cleaning process on
the API. The degrading and denaturing effects of chemi-
Figure 5: Performance Control Limit from Example Dataset. cal detergents should be studied for each product manu-
factured within the facility. Setting acceptance limits for
If the dataset contains an excessive number of zero product carryover based on TOC can be accomplished
values, the 0 values should be removed and the alpha with the Cleaning Process Capability, Safety Factor, or
rate (e.g., 0.27% or 0.0027) adjusted accordingly prior Toxicology Threshold approaches. As on-going cleaning
to transforming the data with the Box-Cox method. In studies collect TOC data, these data can be evaluated
the example dataset, 428 of 1034 results are 0. The with the Performance Control Limit approach to ensure
alpha rate (0.027) is therefore adjusted according to the control of the equipment cleaning process is maintained.

Special edition: Cleaning Validation 25


Rizwan Sharnez

ACKNOWLEDGEMENTS amino acid residues. Oligopeptides have 10


We thank Kristina Conroy, Mariann Neverovitch, or fewer amino acids. Molecules consisting
Michael Hausladen of Bristol Myers Squibb, Rob Lynch from 10 to 50 amino acids are called pep-
of GlaxoSmithKline, Jim Heimbach and Ben Locwin of tides. The term protein describes molecules
Lonza, Stephanie Donat, Gareth Sanderson of Novar- with more than 50 amino acids.
tis, Martin Hammarstrm of Pfizer and David Bain of TOC Total Organic Carbon
BioPhorum Operations Group for their help and support. TTC Toxicological Threshold of Concern

ACRONYMS AND DEFINITIONS REFERENCES


Action Limit An empirical limit that the cleaning process 1. G.L. Fourman and M.V. Mullen, Determining Cleaning Valida-
cannot exceed without potential impact to tion Acceptance Limits for Pharmaceutical Manufacturing
product quality or patient safety. Operations, Pharmaceutical Technology 17 (4), 54-60, 1993.
PDE Permissible Daily Exposure (also called ADE, 2. Methodology for Assessing Product Inactivation during Clean-
Acceptable Daily Exposure) which represents ing, Part I: Experimental Approach and Analytical Methods,
a dose of a drug to which a human may be Journal of Validation Technology 17 (4), 2012.
exposed per day or per dose (for biologics) 3. Solubility of Proteins, Journal of Protein Chemistry 5 (6), 1986.
without any anticipated pharmacologic or 4. ICH, Q3A Impurities in New Drug Substances, 2008.
toxicological effects, so in the event of po- 5. AR.C Beavis, Chemical mass of carbon in proteins, Analytical
tential carry-over of one API to another, there Chemistry 65, 496-497, 1993.
would be no risk to the patient. 6. Kroes et al., Structure-based thresholds of toxicological con-
Alert Limit An empirical limit, statistically established cern (TTC): Guidance for Application to Substances Present
from study data, which is used to monitor at Low Levels in the Diet, Food and Chemical Toxicology 42
the quality of the cleaning process. 65-83, 2004.
API Active Pharmaceutical Ingredient 7. D.G. Dolan, B.D. Naumann, E.V. Sargent, A. Maier, and M.
Degrade To cause the cleavage and hydrolysis of Dourson, Application of the Threshold of Toxicological Con-
chemical bonds within peptides and amino cern Concept to Pharmaceutical Manufacturing Operations,
acid strings, such that the biological activity Regulatory Toxicology and Pharmacology 43, 19, 2005.
is diminished or eliminated. 8. J.P. Bercu and D.G. Dolan, Application of the Threshold of
Denature To cause the tertiary structure of a biologi- Toxicological Concern Concept When Applied to Pharma-
cal product to unfold, as with heat, alkali, or ceutical Manufacturing Operations Intended for Short-term
acid, so that some of its original properties, Clinical Trials, Regulatory Toxicology and Pharmacology 65,
especially its biological activity, are dimin- 162167, 2013.
ished or eliminated. 9. ICH, Assessment and control of DNA reactive (mutagenic) impurities
MAC Maximum Allowable Carryover in pharmaceuticals to limit potential carcinogenic risk. M7 Step 2
Peptides A chemical compound containing two or version (2013).
more amino acids (amino acid polymers) that 10. Statistics for Experimenters: Design, Innovation, and Discovery, 2nd
are coupled by a peptide bond. Peptides are ed., 2005.
often classified according to the number of

26 Special edition: Cleaning Validation


Tim Sandle

Aseptic Transfer Risk


Assessment:
A Case Study | IVT
Tim Sandle, Ph.D.

ABSTRACT
This paper uses one example of a risk assessment approach to illustrate
how risk assessment can be incorporated into good manufacturing
controls. The specific activity assessed involves transferring a set of
sterilised stoppers from an autoclave to a filling machine within a ster-
ile manufacturing facility. The risk assessment approach adopted is a
form of HACCP (Hazard Analysis Critical Control Points). Approaches
to risk assessments are discussed. Key activities are described. Docu-
mentation is critical. There is no such thing as zero risk. A deci-
sion is thus required as to what is acceptable risk. Key components
of HACCP including hazard analysis and critical control points. The
seven pillars of HACCP are described. Stepwise activities needed to
accomplish the risk assessment are discussed. This case study de-
scribed a low risk activity. Rationale for the assessment is described.
This discussion described a risk assessment approach using a relatively
simple case to illustrate its potential for other applications.

INTRODUCTION
Risk analysis and risk management are significant considerations in
current pharmaceutical manufacturing practices. All areas and func-
tions in the pharmaceutical manufacturing plant assess level of risk to a
process and then take steps to eliminate that risk. This paper uses one
example of a risk assessment approach to illustrate how risk assessment
can be incorporated into good manufacturing controls.
Risk assessment involves identifying risk scenarios either prospectively
or retrospectively. The former involves determining what can go wrong
in the system and all the associated consequences and likelihoods; the
latter this looks at what has gone wrong. Risk assessment is then used to
assess the process, product, or environmental risk and to aid in formulat-
ing the appropriate actions to prevent the incident from re-occurring (1).
The case examined here is a prospective risk assessment.
The case study involves transferring a set of sterilised stoppers from
an autoclave to a filling machine within a sterile manufacturing facility.
The risk assessment approach adopted is a form of HACCP (Hazard
Analysis Critical Control Points).

APPROACHES TO RISK ASSESSMENT


There are various approaches to risk assessment being used in the phar-
maceutical industry. Each has their respective merits. The use of risk
assessment is commonly expected by regulatory authorities (2).
There are various tools that can be drawn upon for conducting risk
assessments. Some common methods include FMEA (Failure Mode
and Effects Analysis), FTA (Fault Tree Analysis), and HACCP (Hazard
Analysis Critical Control Points). Many of these employ a scoring ap-
proach. No definitive method exists for all applications and different

Special edition: Cleaning Validation 27


Tim Sandle

approaches are useful for different situations (3). Before embarking upon risk assessment it is impor-
The various approaches differ in their structure tant to establish and to define:
and with the degree of complexity involved. None- Develop and agree on the risk question. A clearly
theless, there are some broad similarities. The differ- defining the risk question facilitates selection
ent analytical tools are similar in that they generally of the appropriate risk assessment tool, identi-
involve: fies relevant data, information and assumptions;
assists in the identification of resources, respon-
Constructing diagrams of work flows, sibilities and accountabilities; and ensures that
Pin-pointing areas of greatest risk, appropriate focus on the business objective is
Examining potential sources of contamination, maintained.
Deciding on the most appropriate sample meth- Select the most appropriate risk assessment tool.
ods, The selected risk assessment method or tool will
Helping to establish alert and action levels, be used to organise collected data, understand
Taking into account changes to the work process what steps can be taken to reduce or control risk,
/ seasonal activities. and to help make appropriate decisions.

Documenting risk assessments is important. APPLICATIONS TO CLEANROOM ENVIRONMENTS AND


When a risk assessment is presented to an auditor, PHARMACEUTICAL PROCESSES
it is likely that the reviewer will be concerned with The primary risk assessment approaches can al-
whether the risk assessment is traceable to a risk low for a complete review of operations within the
methodology and that the process is clear, under- cleanroom ensuring those facilities, operations and
standable, and that it has been performed consistent- practices are also satisfactory. The approaches recog-
ly. In this sense, the reviewer will want to under- nise a risk, rate the level of the risk, and then develop
stand how the decisions were made and that the risk a plan to minimise, control, and monitor the risk.
(or problem) was defined upfront. With the question For example, the monitoring of the risk will help
established, the reviewer will also wish to see if the to determine the frequency, locations, and level for
process performed actually answered the question in environmental monitoring (4).
a meaningful way using scientific principles. This case study outlined in this paper uses a
Important points to remember for any risk assess- modification of the HACCP method, which is some-
ment are the following: times called the Lifecycle Approach. The approach
adopted has been used for similar assessments of
There is no such thing as zero risk. A decision pharmaceutical processes (5).
is thus required as to what is acceptable risk. There are numerous risk factors to be considered
Risk Assessment is not an exact science - differ- when analysing aseptic processing. The HACCP
ent people will have different perspectives on the approach can help to identify these risk factors. The
same hazard. following factors shown in Figure 1 are useful to note

Risk assessments should always:

Be based on systematic identifications of possible


risk factors
Take full account of current scientific knowledge
Be conducted by people with experience in the
risk assessment process and the process being
risk assessed
Use factual evidence supported by expert assess-
ment to reach conclusions
Do not include any unjustified assumptions
Identify all reasonably expected risks-simply and
clearly, along with a factual assessment and miti-
gation where required
Be documented to an appropriate level and con-
trolled/approved
Ultimately be linked to the protection of the
patient
Should contain objective risk mitigation plan. Figure 1: ASEPTIC PROCESSING RISKS

28 Special edition: Cleaning Validation


Tim Sandle

when undertaking HACCP analysis in pharmaceuti- primary disadvantage of HACCP is that, unlike
cal manufacturing. FMEA, HACCP cannot be used to rank or priori-
tize risks.
HACCP
HACCP was developed during the 1960s from DECONSTRUCTING THE PROCESS
collaboration between NASA, a food company (the Four stepwise activities are needed to accomplish the
Pillsbury Company), and the US Army Natick Labo- risk assessment:
ratories. The objective was to provide a zero-defect
food supply for astronauts. HACCP was derived from A route map. The facility is drawn and the route
Failure Mode Analysis (6). There are two key compo- indicated
nents of HACCP: Identification of hazards, which can be divided
into biological; physical; equipment; transport
Hazard Analysis: Determining what microbio- and chemical. This will allow an assessment of
logical, physical, or chemical risks are associated existing control measures
with a process. Process flow
Critical Control Point: A point, step, or procedure Assessment of environmental monitoring. This
at which control can be applied. will determine if the activity is safe to proceed (7).

HACCP generally involves an assessment of the fol- Route Map


lowing conditions known as the seven pillars. The first step was to outline a route map. This
focused on transporting the stoppers by the shortest
1. Conducting a hazard analysis. List all potential and safest route possible as illustrated in figure 2.
hazards associated with each step, conduct a
hazard analysis, and consider any measures to
control identified hazards.
2. Determining the Critical Control Points (CCPs).
Critical control points have to be defined as
processing steps at which necessary action can
be applied to ensure and maintain compliance
with specified conditions. Those points are
identified with appropriate measures that can
be applied to control each hazard. If no such
critical control points can be established, prod-
uct specific validation should eliminate poten-
tial risks of a certain process step.
3. Establishing critical limit(s). Establish critical
limits for each CCP. Figure 2: ROUTE MAP. Arrows indicate stopper transfer route from auto-
4. Establishing a system to monitor control of the claves to filling room
CCP.
5. Establishing the corrective action to be taken Identification of Hazards
when monitoring indicates that a particular The second step was to understand the hazards and
CCP is not under control. the process. The primary hazards with an aseptic
6. Establishing procedures for verification to con- process are biological, physical, and equipment re-
firm that the HACCP system is working effec- lated (8) as displayed in Table 1.
tively.
7. Establishing documentation and record keep- Process flow
ing. The third step was to outline the process flow as
shown in Figure 3.
Aspects of these have been utilized in the case study.
Environmental monitoring
In general, the advantages of the HACCP ap- The fourth step involved assessing dynamic state
proach are that it allows for a systematic overview viable microbiological environmental monitoring
of the process for the evaluation of each process- and particle counts. If data have been satisfactory
ing step, allows each step to be examined the and there are no indications of adverse trends, the
possible risks, and allows for the specification of proposal can be advanced. If there is a level of
measures required for controlling each risk. The risk, this must be addressed first using appropriate

Special edition: Cleaning Validation 29


Tim Sandle

Table 1a: IDENTIFICATION OF HAZARDS

Figure 3a: Process flow with CCPs (critical control points) marked.
Process flow identifies hazards.

Table 1b: IDENTIFICATION OF HAZARDS

corrective and preventative action. Undertaking an


unusual event in a high-risk situation will com- Figure 3b: Process flow with CCPs (critical control points) marked.
pound the problem. Process flow identifies hazards.

30 Special edition: Cleaning Validation


Tim Sandle

RISK ASSESSMENT PERFORM A SIMULATION REFERENCES


Before undertaking the activity, a simulation should 1. Sandle, T. (2011): Risk Management in Pharmaceutical
be performed so that any previously unforeseen prob- Microbiology. In Saghee, M.R., Sandle, T. and Tidswell, E.C.
lems can be noted and further preventative measures (Eds.) Microbiology and Sterility Assurance in Pharmaceuticals
taken. If any variability is expected, the simulation and Medical Devices, New Delhi: Business Horizons, pp553-
should be repeated. 588
The simulation should be timed and the number 2. Sandle, T. and Lamba, S. S. Effectively Incorporating Quality
of staff required noted. The focus should remain on Risk Management into Quality Systems. In Saghee, M.R.
the ease of transit. In this case study, the duration (2012) Achieving Quality and Compliance Excellence in Pharma-
of the activity is five minutes and the number of staff ceuticals: A Master Class GMP Guide, New Delhi: Business
involved is two. Horizons, pp89-128
3. Sandle, T. (2013). Contamination Control Risk Assessment.
CONCLUSION In Masden, R.E. and Moldenhauer, J. (Eds.) Contamination
From three possible options -- high risk, medium risk Control in Healthcare Product Manufacturing, Volume 1, DHI
or low risk -- the conclusion of this risk analysis was Publishing, River Grove: USA, pp423-474
that the risk level was low. This evaluation is based 4. Sandle, T. (2003): The use of a risk assessment in the phar-
on the successful identification of the hazards and maceutical industry the application of FMEA to a sterility
control points. The strongest evidence for this came testing isolator: a case study, European Journal of Parenteral and
from the following: Pharmaceutical Sciences, 8(2): 43-49
5. Munro, M. J., Millar, B. W. and Radley. A. S. (2003):
The process being performed by trained clean A Risk Assessment of the Preparation of Parenteral
room personnel operating in an aseptic manner Medicines in Clinical Areas, Hospital Pharmacist, Vol. 10,
The autoclaved stoppers being contained within pp303-305
a sealed box. The seal remains intact throughout 6. Gavin, A., and Weddig, L.M. (Eds.). (1995): Canned Foods:
the transfer Principles of Thermal Process Control, Acidification and Con-
The outlet at the bottom of the box has an integ- tainer Closure Evaluation, Food Processors Institute, Washing-
rity tested vent filter ton, D.C., pp5-10
An additional vent filter is placed on the top of 7. Sandle, T. (2006) Environmental Monitoring Risk Assess-
the box ment, Journal of GXP Compliance, 10(2): 54-73
The stoppers are wheeled on a trolley which 8. Whyte, W. and Eaton, T. (2004): Assessing microbial risk
minimises human intervention to patients from aseptically manufactured pharmaceuticals,
The microbiological environmental monitoring European Journal of Parenteral and Pharmaceutical Sciences, 9
for the rooms is satisfactory. (3): 71-79
9. Whyte, W. and Eaton, T. (2004): Microbiological contamina-
It is important to note the impact of change and tion models for use in risk assessment during pharmaceutical
to review any risk assessments on a regular basis in production, European Journal of Parenteral and Pharmaceutical
developing any risk assessment. Sciences, 9 (1): 11-15
This case study may or may not be typical. It may 10. Sandle, T. (2004): General Considerations for the Risk Assess-
or may not be wise for an organisation to undertake ment of Isolators used for Aseptic Processes, Pharmaceutical
such an activity. The purpose of this discussion was Manufacturing and Packaging Sourcer, Samedan Ltd, Winter
to demonstrate an example of the risk assessment ap- 2004, pp43-47
proach using a relatively simple case so that the wider
application can be appreciated.

Special edition: Cleaning Validation 31


Tim Sandle

People in Cleanrooms:
Understanding and Monitoring
the Personnel Factor | IVT
Tim Sandle, Ph.D.

ABSTRACT
Cleanroom contamination can arise from a number of sources. Most
contamination within the pharmaceutical facility can be traced to
humans working in cleanrooms. The paper discusses staff gowning and
personnel behavior in pharmaceutical cleanrooms, and how cleanroom
risk can be minimized. The human skin ecosystem is discussed. The
Human Microbiome Project (HMP) from the US NIH characterized
microorganisms found in association with both healthy and diseased
humans. Information from this project has great impact on cleanroom
activities including gowning practices. Topics associated with clean-
room garments are discussed including fabric types, garment lifespan,
recycling, laundering, human changing procedures, training, behavior,
hand sanitization, ongoing assessments, and associated topics.

INTRODUCTION
Cleanroom contamination can arise from a number of sources. These
may vary depending upon the type of cleanroom, its geographic loca-
tion, the types of products processed, and so on. Nevertheless, these
sources can generally be divided into the following groups1:

People
Water
Air and ventilation
Surfaces
Transport of items in and out of clean areas

Most contamination within the pharmaceutical facility can be traced to


humans working in cleanrooms2. This is, in some way, evidenced from
the association of microorganisms transient or residential to skin being
the primary isolates from environmental monitoring in controlled envi-
ronments3. Human personnel shed high numbers of skin cells mostly as
skin flakes. The cleanroom garments worn by personnel cannot contain
all human detritus.
The paper discusses staff gowning and personnel behavior in phar-
maceutical cleanrooms. Further, it considers how cleanroom risk can
be minimized. Basic training for all cleanroom staff including activities
such as cleanroom entry and gowning practices is examined.

HUMAN SKIN
Before proceeding to look at gowning, it is worthwhile to consider
the human skin ecosystem. The human body is an intricate system
that hosts trillions of microbial cells across the epithelial surface and
within the mouth and gut. These microorganisms play a role in human
physiology and organ function including digestion and immunity. The
microorganisms also affect the outside environment as they are shed

32 Special edition: Cleaning Validation


Tim Sandle

AREA NUMBER OF MICRO- affected by available nutrients as well as by several


ORGANISMS/cm2 non-nutritional factors such as pH, humidity, and
temperature. With the skin, however, as epithelial
Scalp 1 million
cells are continually being shed, many microbial
Saliva and nasal fluid 10 million/gram communities on the external surface are rarely
Back 100 stable6.
Groin 1 20 million The outcomes of the HMP research have shown
Forehead 100 1000 that there is a high population on, and a considerable
Hand 10,000 100,000 diversity of microbial species across, the outer layer
Feet 1 million of the skin. There are approximately 1000 species
Table 1: HUMAN BODY SITES AND TYPICAL NUMBERS OF of bacteria from 19 phyla on human skin. Of these,
MICROORGANISMS WITHIN AREA most bacteria can be categorized into four phyla:

Actinobacteria (51.8%). Actinobacteria are a


from the skin or deposited through different orifices. group of Gram-positive bacteria with high gua-
This latter issue has important implications for clean- nine and cytosine content, such as Micrococcus,
rooms. The outer layer of human skin can host up to Corynebacteria and Propionibacteria.
one million microorganisms per square centimeter4. Firmicutes (24.4%). This includes the genera
The population, as well as the diversity, varies ac- Clostridia and Bacillus.
cording to anatomical locale. Proteobacteria (16.5%). This is a major phylum of
Research suggests that a typical person sheds bacteria that includes a wide variety of pathogens
1,000,000,000 skin cells per day of a size 33 m x such as Escherichia, Salmonella, Vibrio, Helico-
44 m x 4 m -- equivalent to a rate of 30,000 to bacter, and many other notable genera.
40,000 dead skin cells shed from the surface of the Bacteroidetes (6.3%). The phylum Bacteroidetes
skin every minute. Of these, Whyte indicates that ap- is composed of three large classes of Gram-
proximately 10% of particles carry microorganisms5. negative, nonspore-forming, anaerobic, and rod-
There are, on average, four microorganisms per skin shaped bacteria7.
cell. A term commonly used to describe skin flakes
with adhered microorganisms is microbial carrying Reasons for the topographical variations relate to
particles. the physicochemical properties of the skin. This is
The significance of this is that people are not only especially so for temperature, pH, amounts of oil, and
a source of contamination, but also are an agent for moisture8. From this, there are three main ecological
transferring contamination possibly to locations that areas of the skin: sebaceous, moist, and dry. Exam-
could pose a product risk. Microorganisms are spread ples of microbial divergence include Propionibacteria
from sneezing, coughing, and touching. While micro- and Staphylococci species dominating the sebaceous
organisms suspended in the air are less of a concern, areas (with a high oil content). Dry, calloused areas
should such organisms gravitate towards a product or (arms and legs), Gram-positive cocci (primarily the
critical location, they may present a significant risk. Micrococcaceae) are found on the arms and legs and
Gram-positive rods are found in high numbers on the
HUMAN MICROBIOMe PROjeCT torso. Staphylococci and Corynebacteria are found
Our understanding of the risk the people pose to together with some Gram-negative bacteria in moist
cleanrooms has been advanced by the findings from areas9. These types of microorganisms generally
the Human Microbiome Project. The Human Microbi- reflect the types recovered from cleanrooms10.
ome Project (HMP) is a United States National Insti- The reason that Gram-positive bacteria predomi-
tutes of Health initiative. The project goal is to iden- nate across the skin is because the skin is generally a
tify and characterize the microorganisms found in dry environment, and any fluids present on the sur-
association with both healthy and diseased humans face generally have a high osmotic pressure11. Thus
(the human microbiome). The human microbiome Gram-positive bacteria (especially the Staphylococci
describes the aggregate of microorganisms and their and Micrococci) are better adapted for such environ-
genetic interactions that reside on the surface and in ments, not least to being resistant to desiccation.
deep layers of skin, in the saliva and oral mucosa, in Where other species occur this is due to variations in
the conjunctiva, and in the gastrointestinal tract. temperature and with areas of higher sweat produc-
Some of the most illuminating HMP research has tion12. For example, this can lead to higher levels
been with the human skin. The skin is a complex of fungi on the feet13. In relation to pharmaceutical
ecosystem, supporting a range of microbial commu- manufacturing, the presence of any such organisms
nities that live in distinct niches. These niches are remains problematic.

Special edition: Cleaning Validation 33


Tim Sandle

A further observation is that the ratio of the micro- PROTeCTING PRODUCTS FROM PeOPLe
organisms recovered from the skin is relatively evenly Despite some advances with automation and robotics,
divided between the aerobic and the anaerobic. The in most situations people cannot be eliminated from
aerobic microorganisms tend to live on the outermost cleanrooms. Control of contamination from people in
layers of the skin and the anaerobic microorganisms cleanrooms is achieved by application of two prin-
live in the deeper layers of the skin and hair follicles14. ciples:
The information from the human microbiome proj-
ect about the rich depth of variety of microorganisms We wrap the people to minimize the amount of
on the skin introduces several implications for clean- shedding of microorganisms.
room environmental monitoring. The most important We put localized protection around the prod-
question of whether gowning practices are adequate uct to minimize the amount of contact with the
to exclude all microorganisms from the richest areas people.
of the skin microbiome. This is a pertinent point
given that most bacteria free-floating in cleanroom air The localized protection issue is achieved through
current are not free-living but are instead the result local air protection, such as unidirectional airflow
of direct particle shedding of desquamated skin cells cabinets and isolators. With clothing, personnel
and subsequent re-suspension of skin detritus in the working in cleanrooms are required to wear special
air stream. clothing designed for the clean environments. Such
clothing is necessary, as indicated above, because the
The answer to this question should lead to a consid- human body creates its own micro-environment of
eration of: potentially damaging particulate contamination.

1. The types of cleanroom undergarments used To be effective, cleanroom clothing must:


and an examination as to whether these pro-
vide an effective barrier, especially for the more Form a particulate barrier for the human micro-
moist parts of the body. environment.
2. The importance of the outer gown covering all Allow freedom of movement and be comfortable.
parts of the body, including the forehead. Address any specialist requirement, e.g. static
3. The quality of cleanroom certified undergar- dissipation.
ments. Avoid being a significant particulate contributor
4. The level of training required for operators in in itself.
relation to gowning and the way that gowning
qualification as conducted. Cleanroom garments must meet specific protection
5. How long a cleanroom suit should be worn for criteria. Not all cleanroom garments are of the same
in relation to material integrity against operator quality. This involves manufacturing the garments
perspiration. from special materials, following particular construc-
6. The environment in which gowns are donned, tion methods, and then tailored for individual styling.
where higher air-change rates might prove effec- The gowns must be comfortable, easy to apply and
tive. practical in use. Some gowns are disposable and
7. How often gowns should be recycled which others are made to be re-laundered and sterilized
involves washing and irradiation. At some point depending on the cleanroom grade.
the material fibers will weaken, thereby reduc-
ing the bacteria filter efficiency of the gown. CLeANROOM GARMeNTS AND FABRICS
The wearer of the gown should know what Fabrics used in the manufacture of cleanroom gar-
types of testing is conducted on recycled gowns ments must have the following features15:
and what the procedures are for rejecting gowns
where a loss of integrity is detected. Be low in particulate shedding.
Permit the body to breathe while trapping parti-
Cleanroom microbiologists may wish to consider cles within the garment. The contaminant should
how concerned they are with each of the items listed be retained within the garment and not released
in excluding microorganisms found on all regions of into the surrounding atmosphere.
the skin. There must be good understanding of the Be flexible enough for comfortable wearing.
environmental monitoring methods used to assess Withstand repeated cleaning and sterilization
cleanrooms. These may not show how good or bad cycles.
gown changing and gown wearing is. These concerns Meet any specific requirements such as control of
are best addressed through good gowning practices. static.

34 Special edition: Cleaning Validation


Tim Sandle

Meet opacity requirements. worn only for the length of the shift (normally four
Look and feel as good as possible. hour periods to enable operators to take breaks). In
Be cost-effective. lower grade cleanrooms, a gown might be worn for
several sessions during the course of the working day.
Fabric Categories Other factors affecting the lifespan of the gowns
There are three broad categories of fabric used in the that are subject to recycling are repairs and the num-
construction of cleanroom garments: ber of permitted washing cycles. With repairs, it is
prudent to have a repair policy. This will vary across
Woven fabrics. Woven or re-usable fabrics are facilities, and again, it will be affected by the clean-
the most commonly used fabrics in cleanroom room class. With aseptic areas, if a gown becomes
environments. Such garments are woven on torn it is normally discarded. In other grades of
sophisticated looms from yarns of continuous cleanroom, a gown can be repaired depending upon
filaments of polyester. The thickness of the yarn the size of the hole and the impact on the material.
and filaments is important -- the finer the yarn, Some organizations set a maximum size for any hole
the tighter the weave can be made, and the bet- or tear and for the number of times a gown can be
ter the filtration. The pattern and tightness of the repaired.
weave is important to reduce the pore size to a Gowns that are recycled are subject to laundering.
minimum. The use of continuous filament poly- Gowns are washed by special washing machines with
ester means that there are few loose ends from suitable detergents, dried, folded, and then wrapped
which particles may be shed. in cleanroom packaging. For gowns that are to be
Laminated or membrane fabrics. Laminated used in aseptic areas, such gowns are irradiated. A
fabrics are favored for some high-grade micro- policy should be in place outlining how often a gown
electronic environments. These types of garments can be processed -- typical times range between 20
are not commonly used in the pharmaceutical and 40 times. To make the tracking task easier, many
sector. gowns sterilized by irradiation or gassing are fitted
Disposable or limited life materials. The most with barcodes and scanned. It is further important
common of these non-woven fabrics are from to establish the extent that the sterilization process
spun bonded olefin and polypropylene. Compris- affects the integrity of the gown material.
ing a densely interlinked matt of fibers, these In order to assess the contamination risks from
fabrics can provide good results for a limited re-laundering, gowns are subject to particle count-
period. Garments from such materials need to be ing. There are different ways to do this, although the
processed and decontaminated before use in the most common means is the Helmke Drum particle
cleanroom. Disposable or limited use garments emission test. With this, the test method simulates
are mainly used in those environments where particle shedding of clothing under movement. The
protection of the wearer against potentially haz- garment under test is tumbled in a rotating drum
ardous products is required. (approximately 10 revolutions per minute) to release
particles from the surface of the cleanroom garment
Garment Considerations in a controlled manner. An automatic particle counter
Garments are designed to provide protection for the is used to sample the air within the drum to deter-
head, body, hands and feet. In establishing a system mine the average particle concentration of the air
for garment selection, it is important to consider the during the initial ten minutes of the test. The com-
broader aspects of cleanroom use: suitability of fabric, mon standard is the IEST Recommended Practice
garment style, layers, the nature of the tasks involved, RP-CC003.3: Garment System Considerations for
costs, regulatory requirements, and any specific cus- Cleanrooms and Other Controlled Environments.
tomer requirements. The classification of the clean- An alternative measure is the Body Box test. This
room will inevitably be the major factor in determin- method simulates particle filtration and release under
ing the degree of personnel protection required and real wear conditions. As a consequence it measures
the fundamental choice of garments. the contamination of the cleanroom by the cloth-
One important issue with gowns is the maximum ing/wearer. For this, particle counters determine the
length of time that a gown can be worn. As people quantity of particles generated by the wearer/garment
perspire, the integrity of the gown will weaken. Com- that are emitted into the chamber.
plicating factors are the temperature and humidity
of the cleanroom and the variations between people. CHANGe PROCeDURe
The length of time will also depend upon the grade Cosmetics, such as powder, rouge, eye liner, mascara,
of the cleanroom. In aseptic areas, such as ISO 14644 and lipstick must be banned in cleanroom environ-
class 7 / EU GMP Grade B areas, gowns are typically ments. Jewelry, such as rings, watches, necklaces,

Special edition: Cleaning Validation 35


Tim Sandle

bracelets, earrings and other items, together with all Training must be documented and regularly
forms of visible piercing, are commonly not allowed reviewed. Training must be effective. Actual perfor-
in cleanrooms. mance of personnel competency in gowning should
The best method of changing into cleanroom gar- be demonstrated on a regular basis.
ments is one that minimizes contamination getting
onto the outside of the garments. Change areas can CLeANROOM PeRSONNeL BeHAVIOR
vary in design, but it is common to find them divided Working in clean environments demands knowledge,
into three zones: discipline, motivation as well as a thorough under-
standing of contamination risks among all personnel
1. Pre-change zone. Outside of changing rooms involved. Each individual cleanroom should have its
tacky mats or polymeric flooring can be posi- own documented rules and procedures.
tioned to help reduce the level of particles car- Training includes reminding personnel that they
ried on footwear16. must not be allowed to touch critical products and
2. Changing zone. The changing room design equipment with their naked hands. All critical work
contributes to the assurance of appropriate must be undertaken wearing gloves. Critical ac-
personnel access and microbial contamination tivities requiring personnel contact such as aseptic
control. The changing room should be provided processing or sampling must be done through the
with filtered air. Intermediate (bag) filters will use of clean utensils such as tweezers, forceps, and
typically be suitable for this purpose, though the equivalent. All devices and gloves used must
High Efficiency Particulate Air (HEPA) filtration fully comply with the cleanliness demands of the
may be used. The air pressure should be nega- cleanroom and work undertaken in the cleanroom.
tive with regards to the manufacturing area cor- They must be cleaned, disinfected, or sterilized as
ridor, but positive relative to external adjacent appropriate for the criticality or activity and risk of
areas. contamination.
3. Cleanroom entrance zone. This must be of the Another aspect of best practice is in instruct-
same grade or class as the main cleanroom into ing personnel in the appropriate behaviors within
which the area leads. the cleanroom. The generation of contamination is
proportional to activity conducted. A person with
Ideally there should be separate routes through head, arms, and body moving can generate about
airlocks for material required in cleanrooms. Tak- 1,000,000 particles 0.5 m/min. A person who is
ing items through personnel change areas should be walking can generate about 5,000,000 particles 0.5
discouraged. m/min. However a person in motionless position
can generate only 100,000 particles 0.5 m/min.
TRAINING In addition, personnel should reduce activities like
Personnel training in gowning is an important func- talking, singing, whistling, coughing, sneezing etc.,
tion. Gowning practices must be assessed periodi- especially when being close to the handled products
cally and monitored frequently. Training programs and production equipment.
should ideally include visual assessment and microbi- People working in cleanrooms and other forms of
ological assessment. The microbiological assessment controlled environments must be physically healthy.
varies, but can include the exposure of settle plates Diseases in the upper respiratory tract as well as
during the change process and the assessment of stomach disorders can create problem in hygienic
gown cleanliness through post-use suit contact plates. applications.
The results of the cleanroom sampling should not ex- Another factor that can impact upon the environ-
ceed those of the room class. If results are exceeded, ment is the number of people in the cleanroom. Only
the individual may be an unusually high shedder of necessary and limited number of persons should be
skin particles. allowed in a cleanroom at the same time. The more
Training required for staff who work in cleanrooms persons simultaneously present in a cleanroom, then
should include: the higher the contamination level will be, i.e., the
higher concentration of particles in the air). This is
Introduction to micro-organisms and microbio- particularly important in relation to changing rooms.
logical contamination control.
Entry and exit of production facilities (including HAND SANITIZATION
gowning). Good personal hygiene is a requirement of all phar-
Personal hygiene training. maceutical cleanroom activities. However, studies
Microbiological risks associated with specific show poor compliance is common in relation to basic
production tasks. hand washing technique. Hand hygiene and glove

36 Special edition: Cleaning Validation


Tim Sandle

Thus, ethanol is more often applied to bare skin


(often in a denatured form) whereas IPA is more often
applied to gloves.
These sanitizers have bactericidal action against
vegetative cells but not spores. The concentration of
alcohol to water varies, although the optimal range is
60 to 90% (volume/volume). Below 60%, bactericidal
action drops, and above 90% there is insufficient wa-
ter for the bacterial cell to absorb water. The alcohol
does not enter the cell and is unable to denature the
bacterial proteins21. Most preparatory concentrations
are 70%.
While most bacteria are killed after ten seconds of
contact with alcohol22, contact times in practice are
Figure 1: HAND SANITIZATION (IMAGE: TIM SANDLE) longer due to the variability of hand rubbing. Typical
contact times are thirty seconds.
hygiene are important given the high numbers of It is important that the selection of a hand sani-
microorganisms found on the human body including tizer is qualified. There are different approaches that
the hands and the risks posed by hands as a means can be taken for qualification. Most of these require
of contamination transfer. Microorganisms associ- individuals to wear gloves and to place their hands
ated with hands are found mainly on the surface of into broth containing a high concentration of a non-
the skin and under the superficial cells of the stratum pathogenic microorganism. Disinfectant is then ap-
corneum. The dominant species is Staphylococcus plied, and the bacterial reduction is assessed through
epidermidis that is found on almost every hand, placing the treated hands into broth and performing
together with other species of Staphylococcus and dilutions.
species of the genera Micrococcus17.
Hands must be washed with soap and water prior ON-GOING ASSeSSMeNT
to entry to the cleanroom. Hand washing facilities In higher-grade cleanrooms such as those used for
should not be located in an actual cleanroom, but aseptic processing, it is common practice to assess the
rather in an area leading to the cleanroom changing risk of personnel to the process by taking suit contact
room. As an alternative, a hygienic handrub can be plates of the gown as worn by the person as they
used. leave the aseptic area. The gown must be discarded
Where gloves are required these should be put on after the plates have been taken due to the potential
using a method designed to prevent the ungloved effect on the gown integrity when an agar plate con-
hand from touching the clean or sterile outer part tacts the gown.
of the glove. Once in the cleanroom, gloved hands It is good practice to begin suit sampling with
should be subject to periodic hand sanitization18. a higher number of samples. These can then be
When decontaminating hands with an alcohol- reduced over time. Some facilities perform more
based antiseptic hand rub, apply product to palm samples from the gown during gowning test qualifi-
of one hand and rub hands together, covering all cations compared with routine sampling. Sites con-
surfaces of hands, fingers and wrists, until hands are sidered for selection include the top of the head, the
dry (alcohol-based hand rubs are not to be used with face mask, both arms, middle torso, and both legs.
water). The process typically takes between thirty In terms of limits, for EU GMP Grade B/ISO class 7
seconds and one minute. Follow the manufacturers areas, the aim is often to adopt the same limits as per
recommendations regarding the volume of product to the limits applied to finger plates. The action level for
use. gowns is ordinarily 5 CU/25cm 2.
The technique for applying alcohol to gloved hands
is similar to applying a handrub to skin. It is im- Experience has shown that higher counts are
portant to ensure that all surfaces are covered. With obtained from the top of the head, perhaps because
glove sanitization, there are two alcohols of choice: this is the warmest region of the body. Care must be
ethyl alcohol (ethanol) and isopropyl alcohol (IPA). undertaken when sampling as so not to break the
Other alcohols, such as methyl alcohol (methanol) are integrity of the gown.
unsuitable19. Of the two alcohol forms, IPA is slightly In addition to gowning control, a procedure should
more bactericidal than ethanol, although ethanol has be in place for the notification of health conditions
better viricidal properties20. Another factor is appli- by staff. Staff who are ill (coughs, colds, and so on)
cation to the skin, and here IPA can be quite harsh. should not enter cleanrooms. This is because the

Special edition: Cleaning Validation 37


Tim Sandle

illness may affect product quality. It is important to 8. Costello, E.K., Lauber, C. L., Hamady, M., Fierer, N., Gordon,
control the potential risks from personnel carrying J.I., Knight, R. (2009). Bacterial community variation in
human body habitats across space and time, Science, 326:
Infectious disease. 16941697
Open lesions on any exposed part of body. 9. Chen YE, Tsao H. (2013). The skin microbiome: current
Shedding skin conditions, such as eczema or pso- perspectives and future challenges, J Am Acad Dermatol.
riasis, dermatitis, and dandruff (skin scales may 69(1):143-55
harbor objectionable micro-organisms that may 10. Grice, E.A., Kong, H.H., Conlan, S. et al (2009). Topographi-
impact pharmaceutical products and patients). cal and Temporal Diversity of the Human Skin Microbiome,
Gastric upsets. Science, 324: 1190 1192
11. Gao, Z., Tseng, C.H., Pei, Z., and Blaser, M.J. (2007). Molecu-
Personnel with any of the above conditions must be lar analysis of human forearm superficial skin bacterial biota.
excluded from working within cleanrooms for the Proc. Natl. Acad. Sci. 104: 29272932
duration of their illness. 12. Kong, H.H. and Segre, J.A. (2012). Skin Microbiome: Looking
Back to Move Forward, Journal of Investigative Dermatology,
CONCLUSION 132: 933939
This paper has considered the personnel factor and 13. Findley, K., Oh, J. Yang, J., Conlan, S. et al (2013). Topo-
the relationship between people and cleanrooms. It graphic diversity of fungal and bacterial communities in
addressed why people are a risk in relation to the human skin, Nature, 498(7454):367-70. doi:10.1038/na-
skin microbiome, and how good gowning practices ture12171
can help to minimize that risk. The paper has also 14. Cogen A.L, Nizet, V. and Gallo, R.L. (2008): Skin Microbiota:
considered other factors that can affect contamination A Source of Disease or Defence?, Br. J. Dermatol., 158 (3): 442-
risks from people, including the importance of good 55
behaviors and the necessity of hand sanitization. 15. Ramstorp, M. (2011) Microbial Contamination Control in
Capturing these various issues through procedures Pharmaceutical Manufacturing. In Saghee, M.R., Sandle, T.
and imparting the key concepts through training are and Tidswell, E. (Eds.) Microbiology and Sterility Assurance in
a necessary part of cleanroom management. Pharmaceuticals and Medical Devices, Business Horizons: New
Delhi, pp615-701
ReFeReNCeS 16. Sandle, T. (2006) The use of polymeric flooring to reduce
1. Reinmller, B. (2001). People as a Contamination Source contamination in a cleanroom changing area, European Jour-
- Clothing Systems. In: Dispersion and Risk Assessment of nal of Parenteral and Pharmaceutical Sciences, 11 (3): 7
Airborne Contaminants in Pharmaceutical Cleanrooms. Royal In- 17. Kampf, G., Kramer, A. (2004). Epidemiologic Background of
stitute of Technology, Building Services Engineering, Bulletin Hand Hygiene and Evaluation of the Most Important Agents
No. 56, Stockholm, pp. 54-77 for Scrubs and Rubs, Clinical Microbiology Review, p. 863893
2. Hyde, W. (1998). Origin of bacteria in the clean room and 18. Sutton. S., (2009). Hand Washing-A Critical Aspect of Per-
their growth requirements. PDA J Sci Technol; 52:154164 sonal Hygiene in Pharma, Journal of Validation Technology, 15
3. Sandle, T. (2011). A Review of Cleanroom Microflora: Types, (4): 50-55
Trends, and Patterns, PDA Journal of Pharmaceutical Science 19. Spaudling, E.H. (1964) Alcohol as a surgical disinfectant,
and Technology, 65 (4): 392-403 AORN J., 2: 67-71
4. Proksch E.; Brandner J.M.; Jensen J.M. (2008) The Skin: An 20. Klein, M. and DeForest, A. (1963) The inactivation of viruses
Indispensable Barrier, Exp. Dermatol. 17 (12): 106372 by germicides, Chem. Specialist Manufact. Assoc. proc., 49: 116-
5. Whyte, W. (1981) Setting and impaction of particles into con- 118
tainers in manufacturing pharmacies, J. Paren. Sci. Technol., 21. Morton, H.E. (1950) The relationship of concentration and
36: 255-268 germicidal efficiency of ethyl alcohol, Ann N.Y Acad. Sci, 50:
6. Roth, R.R. and James, W.D. (1988): Microbial Ecology of the 191-196
Skin. Annu. Rev. Microbiol. Vol. 42, pp. 44164 22. Morton, H.E. (1983) Alcohols. In Block, S.S. (Ed.) Disinfection,
7. Grice, E.A., Kong, H.H., Renaud, G., Young, A.C. (2008). Sterilisation and Preservation, Philadelphia: Lea and Febiger,
A diversity profile of the human skin microbiota. Genome pp225-239
Research.18:1043-50. (PMID:18502944)

38 Special edition: Cleaning Validation


Xxxxxxx

PAT: Using PAT to


Support the Transition
from Cleaning Process
Validation to Continued
Cleaning Process
Verification | IVT
XXXXXX

ABSTRACT
In accordance with the 2011 FDA guidance for industry, Process Valida-
tion General Principles and Practices, there is a requirement to continue to
verify equipment cleaning processes. This continued verification would
reduce the risk of cross contamination, and consequently, the accept-
able residue limits could be increased to reflect this reduced risk. A
combination of visual residue limits and rapid analytical technologies
for the quantification of residues may lead to a reduced risk of cross
contamination for the patient while at the same time result in signifi-
cantly reduced manufacturing cycle times. This paper discusses the
current cleaning paradigm and the changes continued cleaning process
verification could bring for small molecule pharmaceutical manufactur-
ers. Technologies that offer potential for increased automation of clean-
ing validation are introduced.

INTRODUCTION
Cleaning can generally be defined as the removal of unwanted contami-
nants. The objective of a cleaning process is primarily to ensure safety,
efficacy, and quality of product subsequently manufactured in the same
equipment. Cleaning validation is the documented evidence demon-
strating the effectiveness of a cleaning procedure based on pre-deter-
mined acceptance criteria. It is performed to assure that production ma-
terials that come into contact with manufacturing equipment surfaces
are not contaminated or adulterated. A graphical representation of the
typical cleaning process development and validation process is detailed
in Figure 1. It summarizes the typical process and demonstrates the
extensive workload associated with cleaning process development and
validation.
Figures 2 and 3 summarize the current cleaning procedure develop-
ment and validation approach. This approach is time-consuming and
costly. It can significantly inhibit clinical trial manufacture activities
because of once-off swabbing requirements following each mini-batch
manufactured. It can also inhibit on-going commercial manufacturing
activities due to the burden associated with cleaning procedure valida-
tion and routine monitoring.
Within the FDA guidance document on process validation (1),
process validation is defined as the collection and evaluation of data,
from the process design stage throughout production, which establishes
scientific evidence that a process is capable of consistently delivering
quality products. Process validation involves a series of activities taking

Special edition: Cleaning Validation 39


Xxxxxxx

Figure 3: Current cleaning procedure validation process flow.

Figure 1: Typical cleaning process development and validation process. of risk of cross-contamination of subsequent formu-
lations would reduce accordingly. As a result, the
acceptable residue limits (ARLs) currently associated
with cleaning validation may no longer be appropri-
ate. This is because the safety factors built into such
calculations may have been included as a result of
the snapshot-in-time nature of a cleaning validation
activity.

VISIBLE-RESIDUE LIMITS DURING CLEANING


PROCEDURE DEVELOPMENT AND VALIDATION
A visible-residue limit (VRL) is the quantity of target
residue (usually API) remaining on manufacturing
equipment surfaces when it has reached a visually de-
tectable level. An ARL is the amount of target residue
(usually API) that remains on manufacturing equip-
ment surfaces and carry over to the next formulation
with no pharmacological or adulteration risk. This is
typically calculated using 10ppm or 1/1000th mini-
mum daily dose as outlined in Figure 1.
With a shift to continued verification of a clean-
Figure 2: Current cleaning procedure development process flow.
ing procedure following every clean and the reduc-
tion of cross-contamination risk, the associated
place over the lifecycle of the product and process. ARLs will increase, leading to greater numbers
The guidance describes the process validation activi- of ARLs exceeding VRLs. This may subsequently
ties in three stages: process design, process qualifica- enable greater use of a VRL approach to continued
tion, and continued process verification. verification. The use of visual inspection as an as-
While the greatest focus of this guidance docu- sessment criterion for equipment cleaning effec-
mentation has been related to manufacturing process tiveness has always been a component of cleaning
validation, the same three stages of validation have programs. Mendenhall proposed the use of only
relevance to cleaning process validation. It is likely visual examination to assess equipment cleanliness
that the demonstration of continued cleaning process in 1989 (2). He summarised that visible cleanliness
verification is expected by regulatory bodies in order criteria were more rigid than quantitative calcula-
to demonstrate sufficient control of the cleaning tions and, therefore, adequate. FDA acknowledges
process. the use of visually clean criteria for product-dedi-
With the increased frequency of verification associ- cated equipment. LeBlanc has also reviewed the use
ated with such a philosophy, it is logical that the level of visual examination as the sole acceptance criteria

40 Special edition: Cleaning Validation


Xxxxxxx

development, process qualification, and continued


verification. Considerable quality assurance, time and
resource, and financial benefits may be realized from
adopting such an approach.

ALTERNATIVE TECHNOLOGIES FOR CONTINUED


VERIFICATION OF
Some examples of alternative technologies currently
employed or that show potential for continued verifi-
cation of equipment cleaning processes either as lab,
Table 1: Advantages and limitations of visible-residue limits (VRLs) during at-line, or in-line approaches are detailed as follows.
cleaning procedure development and validation.
Lab-based Technologies
Lab-based technologies include ion-mobility spec-
trometry (IMS), total organic carbon (TOC), and
liquid chromatography-mass spectrometry-mass
spectrometry (LC-MS-MS).

Ion-mobility spectrometry. IMS is a separation


technique, similar to time-of-flight mass spectrom-
etry, that distinguishes ions of a given compound
based on their velocities through a drift tube under
the influence of a weak electric field. To perform the
IMS analysis, a sample solution containing a com-
pound of interest is injected by an autosampler onto a
polytetrafluoroethylene (PTFE) substrate and allowed
to dry. The sample is introduced by heating the
substrate to circa 290C, which results in desorption
or vaporization of the sample into an inlet tube (6).
Primary ion formation occurs through atmospheric
pressure chemical ionisation (APCI) using nickel-63
Figure 4: A process flow of proposed cleaning procedure development as the radioactive source. Following many collisions,
and validation activities. product ions are formed and gated into the drift tube
(6). These ions based on size, shape, and charge travel
for cleaning validation (3). through the drift tube toward the detector at dif-
Forsyth et al. (4, 5) propose that VRLs could be ferent velocities. In contrast to mass spectrometers,
adopted as the primary acceptance criteria during separation of ions is based on a size-charge relation-
cleaning evaluation activities if the VRL is calculated ship rather than a mass-charge ration. It is a fast and
to be less than the ARL. They cited the following specific off-line tool for verifying the cleanliness of
advantages and limitations detailed in the Table. pharmaceutical equipment. Recovery percentages and
As is outlined in the Table, there are still vulnera- standard deviations for IMS samples are consistent
bilities of such an approach, in particular the person- with those obtained with HPLC analysis, but sample
to-person variability of visually clean equipment throughput of IMS is about 50 times faster (6). The
and the variable lighting conditions associated with disadvantages associated with such a method are that
commercial-scale equipment. Even with considerable it is generally an off-line analytical tool that requires
training and experience these risks may remain. sample swabbing and preparation activities. In addi-
There is an opportunity to utilize rapid at-line and tion, significant method development time is required
on-line automated technologies to compensate for the prior to implementation. However online, real time
deficiencies within the VRL approach. A combination applications are becoming available.
of both approaches (i.e., VRLs and automated residue
detection systems) could support the pharmaceutical Total organic carbon. TOC analysis is a method
manufacturing sector to realize a significantly robust that has been used successfully for monitoring water
cleaning process aligned with reduced manufactur- quality, in particular the quality of water for injection
ing cycle times. This proposed paradigm is outlined used in pharmaceutical manufacturing. It offers a
in Figure 4. Figure 4 provides an overview of the number of distinct advantages over other commonly
proposed alternative approach to cleaning procedure used residual testing methods. A typical method may

Special edition: Cleaning Validation 41


Xxxxxxx

involve acidification of a swab sample followed by are not sufficiently rigorous (9-11).
sparging with purified air to remove inorganic carbon 0n-line Technologies
from solution as carbon dioxide gas. The organic
compounds remaining in solution are then oxidized UV-Vis spectrophotometry. UV-Vis technologies
to carbon dioxide in, for example, a combustion monitor rinse effluent to quantify the level of API
tube packed with a catalyst at 680C (7). The carbon contained. UV-Vis spectroscopy is the measure-
dioxide produced in this manner passes through a ment of the wavelength and intensity of absorption
dehumidifier and halogen scrubber before it reaches of near-ultraviolet and visible light by a sample.
a cell where it is quantified by a non-dispersive in- Ultraviolet and visible light are energetic enough
frared detector tuned to the highly specific carbonyl to promote outer electrons to higher energy levels.
frequency in the 1-700cm-1 range (7). The signal UV-Vis spectroscopy is usually applied to molecules
response is compared with a calibration curve gener- and inorganic ions or complexes in solution. The
ated by analysis of solutions of potassium biphthalate UV-Vis spectra have broad features that are of lim-
corresponding to known levels of organic carbon. ited use for sample identification but are useful for
The disadvantages associated with such a method are quantitative measurements. The concentration of an
that it is non-specific and an at-line analytical tool analyte in solution can be determined by measur-
that requires sample swabbing and preparation activi- ing the absorbance at some wavelength and applying
ties (7). the Beer-Lambert Law. The light source is usually a
hydrogen or deuterium lamp for UV measurements
LC-MS-MS. For low-dose compounds, equipment and a tungsten lamp for visible measurements. The
requiring low residue limits, and compounds lacking wavelengths of these continuous light sources are
strong chromophores, the sensitivity and selectivity selected with a wavelength separator such as a prism
of LC-MS-MS facilitates rapid detection of low levels or grating monochromator. Spectra are obtained by
of residues of active pharmaceutical ingredients (8). scanning the wavelength separator and quantitative
Like IMS, the disadvantages associated with LC- measurements can be made from a spectrum or at a
MS-MS are that it is an off-line analytical tool that single wavelength. When the level of API in the rinse
requires significant method development, sample effluent reaches a pre-determined acceptable low
swabbing, and preparation activities. level, the cleaning process end-point is reached. This
is a novel approach that has significant benefits for
At-line Technologies identification of cleaning process endpoint.

ATP luminescence. Adenosine triphosphate (ATP) One disadvantage identified would be the challenges
luminescence-based technologies may be used to of wavelength accuracy. Wavelength accuracy is de-
evaluate the level of microbial contamination follow- fined as the deviation of the wavelength reading at an
ing the completion of an equipment cleaning process. absorption band or emission band from the known
ATP is a marker for cell viability because it is present wavelength of the band. The wavelength deviation
in all metabolically active cells where the concentra- can cause errors in the qualitative and quantitative
tion declines rapidly when the cells undergo necrosis results of the UV-Vis measurement, thereby affecting
or apoptosis. The approach is based on the produc- the accuracy and the sensitivity of the method (12). A
tion of light caused by the reaction of ATP with second disadvantage, similar to other rinse studies, is
added luciferase and D-luciferin (9-11). The emitted that it may not detect API if it is adhered to the vessel
light is proportional to the ATP concentration within wall.
certain limits. The limitation associated with com-
mon luciferase assay technology is the short half-life Single point near infrared (NIR). In situ infrared
of the light emission. This flash-type signal requires reflection-absorption spectroscopy (IRRAS) has the
luminometers with reagent injectors to measure the potential to provide a more rapid analysis than the
quick reaction. Prior to addition of luciferase, there existing methods. Modern Fourier Transform-Infra-
is a requirement to lyse the cells so that the ATP is red (FT-IR) instruments are quite stable and have
released. sufficient sensitivity. Infrared fibreoptics based on
chalcogenide glasses can allow sampling up to several
This is a rapid method of swab analysis that is ac- meters from the spectrometer (13-16). Multivariate
cepted within the food and beverage industries. chemometric tools, such as partial least squares (PLS)
It may have application within sterile and aseptic regression, allow implicit modelling of unquantified
operations in particular. However, it is a non-specific interfering species and instrumental artifacts. For
method and has the potential for contamination by cleaning validation, the physical presentation of the
human somatic cells if sample-handling procedures contamination will depend on the solvent, the chemi-

42 Special edition: Cleaning Validation


Xxxxxxx

cal nature and quantity of the residual material, and processing of the hyperspectral data cube (18).
the substrate surface. Mehta et al. (14), Hamilton et al. The large number of individual spectra acquired
(14), and Teelucksingh et al. (16) demonstrated that across the spatial dimension of heterogeneous com-
IRRAS calibrations can be established for individual pounds provides a basis from which relative concen-
surfactants and APIs on metallic and glass surfaces, trations can be determined for each spatial location.
for an API in the presence of a surfactant on a glass Alternatively, these individual concentrations may
surface, and for an API in the presence of excipients be added together to give the total concentration of a
on a stainless steel surface (13-16). Single point NIR specific material within the sample area.
has potential applications for cleaning verification Chemical images are made up of hundreds of
purposes. Its main advantage over traditional swab- contiguous wavebands for each spatial position of a
HPLC techniques is that it samples those contami- target studied. Consequently each pixel in a chemical
nants that are present on the surface, rather than image contains the spectrum of that specific posi-
those that can be removed by the swab coupled with tion. The resulting spectrum acts like a fingerprint
its speed of analysis. The primary limitation associ- that can be used to characterize the composition of
ated with the method is the restricted field of analysis that particular pixel. There are two basic methods to
and whether it is sufficiently large and representative construct the chemical image. One method involves
to assess the success of the cleaning activity. acquisition of simultaneous spectral positions. The
object is moved underneath an imaging spectro-
NIR-chemical imaging. Chemical imaging (CI) is an graphthis is termed pushbroom acquisition. The
emerging platform technology that integrates conven- other method involves keeping the image field of
tional imaging and spectroscopy to attain both spatial view fixed and obtaining images one wavelength after
and spectral information from an object (17). It has anotherthis is termed staring imager configura-
not to date been evaluated as a technology to quantify tion (17). This method is of particular interest within
residual levels of active ingredients and detergents the pharmaceutical industry for sample analysis and
from equipment surfaces. However, based on its would be considered to pose the greatest potential for
broad applicability and sensitivity, it has potential for use as a cleaning verification device. This is primar-
cleaning validation. Near infrared-chemical imaging ily because the sample can remain stationary and the
(NIR-CI) is the fusion of near-infrared spectroscopy field of view would be comparable with that currently
and image analysis. It can be used to visualize the used during conventional swabbing techniques (circa
spatial distribution of the chemical compounds in a 25cm2).
sample providing a chemical map of a region (Figure Point-source spectroscopic assessments do not
5). Each sample measurement generates a hyper- provide information on spatial distribution of dif-
spectral data cube containing thousands of spectra. ferent constituents. In other words, NIR and Raman
An important part of a NIR-CI analysis is the data spectroscopy can only provide information on a very
narrow sample site. NIR-CI can provide informa-
tion on a substantial sample site thereby making it
applicable to analysis of equipment surfaces dur-
ing cleaning validation. Challenges with regards to
method sensitivity for such residual levels will need
to be overcome before this technology becomes com-
mercially viable.

CONCLUSIONS
Rapid technologies would remove some significant
variables identified with VRLs and enable an analyti-
cal technique that could significantly reduce the time
and cost associated with the cleaning procedure.
They could also support the transition away from
once-off cleaning validation towards continued clean-
ing process verification. This would be in-line with
regulatory expectations and pharmaceutical manu-
facturing needs.
The increased frequency of equipment cleaning
process verification will reduce the risk of active and
Figure 5: Schematic representation of hyperspectral imaging hypercube
showing the relationship between spectral () and spatial (X, Y) dimen- detergent cross-contamination and thereby enable
sions (17). higher acceptance criteria for active and detergent

Special edition: Cleaning Validation 43


Xxxxxxx

carryover. As the pharmaceutical manufacturing 12. Lam, Herman, Performance of UV-Vis Spectrophotometers,
industry transitions towards these continued verifica- Laboratory FocusGazette Edition, Issue, 8, April 2000.
tion philosophies, the requirement for rapid mobile 13. Hamilton et al., Grazing-angle fiber-optic IR reflection-ab-
analytical technologies has become essential in order sorption spectrometry (IRRAS) for in situ cleaning valida-
to sustain robust and lean equipment cleaning pro- tion, Org. Process Res. & Devel., 9(3), 337-343, 2005.
cesses. 14. Mehta, N.K.; Goenaga-Polo, J.; Hernandez-Rivera, S.P.; Her-
nandez, D.; Thomson, M.A.; Melling, P.J. Biopharmacology, 15,
REFERENCES 36-71, 2002.
1. FDA, Guidance for Industry, Process Validation General Prin- 15. Hamilton, M.L.; Persoton, B.B.; Harland, P.W.; Williamson,
ciples and Practices, Current Good Manufacturing Practices, B.E.; Thomson, M.A.; Melling, P.J.; Org Process Res. Dev., 9,
January 2011. 337-343, 2005.
2. D.W. Mendenhall, Cleaning Validation, Drug Development 16. Teelucksingh, N.; Reddy, K.B., Spectroscopy, 20, 16-20, 2005.
and Industrial Pharmacy 15 (13), 2105 2114, 1989. 17. Gowen.A, ODonnell.C.P, Cullen.P.J, Bell.S.E.J, Recent appli-
3. D.A. Le Blanc, Visually Clean as a Sole Acceptance Criteria cations of Chemical Imaging to pharmaceutical process moni-
for Cleaning Validation Protocols, PDA J. Pharm Sci. And Tech- toring and quality control, European Journal of Pharmaceutics
nology, 56 (1), 31-36, 2002. and Biopharmaceutics 69, 1022, 2008.
4. Richard J. Forsyth et al., Risk-Management Assessment of 18. Ravn,C. Skibsted,E. Bro, R. Near-infrared chemical Imaging
Visible-Residue Limits in Cleaning Validation, Pharm. Tech- (NIR-CI) on pharmaceutical solid dosage forms-Comparing
nol. 30, 104114 September 2006. Common calibration approaches, Journal of Pharmaceutical
5. Richard J. Forsyth, Ruggedness of Visible Residue Limits for and Biomedical Analysis 48 554561, 2008. JVT
Cleaning Validation, Pharm. Technol. 33, 102 111 March
2009. ARTICLE ACRONYM LISTING
6. Elizabeth Galella et al., Cleaning Verification: Method De- APCI Atmospheric Pressure Chemical Ionisation
velopment and Validation using Ion Mobility Spectrometry, API Active Pharmaceutical Ingredient
Pharm. Technol. 33, 60 63 July 2009. ARLs Acceptable Residue Limits
7. A. Strege, Terry L Stinger, Brett T. Farell and Avinash L Lagu, ATP Adenosine triphosphate
Biopharm International, April 1996. CI Chemical Imaging
8. Kevin J. Kolodsick et al., Enhancing Drug Develoment by FT-IR Fourier Transform-Infrared
Applying LC-MS-MS for Cleaning Validation in Manufactur- IMS Ion-Mobility Spectrometry
ing Equipment, Pharm. Technol. 30, 56 71 February 2006. IRRAS Infrared Reflection-Absorption Spectroscopy
9. Kangas L., Grnroos M. and Nieminen A.L., Biolumines- LC-MS-MS Liquid Chromatography-Mass
cence of cellular ATP: a new method for evaluating agents in Spectrometry-Mass Spectrometry
vitro, Medical Biology, 62,338 343, 1984. NIR Near Infrared
10. Lundin A., Hasenson M., Persson J. and Pousette A., Estima- NIR-CI Near Infrared-Chemical Imaging
tion of biomass in growing cell lines by ATP assay, Methods PTFE Polytetrafluoroethylene
Enzymol. 133, 27 42, 1986. TOC Total Organic Carbon
11. Crouch S.P.M., Kozlowski R., Slater K.J. and Fletcher J., The UV Ultraviolet
use of ATP bioluminescence as a measure of cell proliferation VRL Visible Residue Limit
and cytotoxicity, J. Immunol. Methods, 160, 81 88, 1993.

44 Special edition: Cleaning Validation


Keith Bader & Kelly Scalva

Translating Laboratory-
Developed Visual Residue
Limits to Process Area
Applications | IVT
Keith Bader and Kelly Scalva

ABStrAct
While considerable attention has been given to the development of
visual residue limits (VRLs) in a laboratory setting, translating the
bench scale values to the assessment of process surfaces has not yet
been thoroughly assessed. However, knowledge of both the critical
parameters that impact the determination of VRLs and the influence of
those parameters on visual inspection can provide a framework for the
development of a robust visual inspection program. Development of
such a program first entails the determination of constraints imposed
by equipment geometries and facility lighting. VRLs can then be deter-
mined for post-productions residues of concern, which, of course, car-
ries its own specific challenges. Once VRLs have been determined, they
cannot be immediately applied without considering certain strategic
cleaning program approaches and potential sources of variability.
Many factors influence how visual inspection will be conducted in a
manufacturing facility. Among the most critical are inspection condi-
tions in the facility, the condition of existing equipment surfaces, and
the physical characteristics of post-production residues deposited on
product contact surfaces. While many in industry embrace the im-
portance of visible residue limits (VRLs), few have a clear pathway to
translate VRLs determined in the laboratory to the manufacturing floor.
The intent of this paper is to provide the background and informa-
tion that will allow industry to formulate a plan of attack to practically
integrate VRLs into the visual inspection program. To begin, the best
starting place is to determine the constraints imposed by the manufac-
turing floor.

EquipmEnt inSpEction conditionS


Conducting a visual residue limit (VRL) determination study in the
laboratory must be completed such that important process equipment
parameters from the manufacturing floor are captured. To wit, the first
step is characterization of the conditions on the manufacturing floor
where visual inspection is put into practice. For any residue, there are
three parameters that must be considered for proper translation and
laboratory analysis: viewing angle, distance, and light intensity at the
product contact surface. The most critical variable for visual inspection
is, of course, the room lighting, so it is important to understand and
characterize lighting in the facility to properly later simulate it at the
bench scale.
The interior task lighting generally conforms to standards set by a
professional organization such as the Illuminating Engineering Society
of North America (IESNA). In the past, over-lighting was the norm
with standard luminance regularly reaching 1200 lux (1). Lighting was

Special edition: Cleaning Validation 45


Keith Bader & Kelly Scalva

light fixtures and whether the color temperature of


the lamp closely replicates sunlight. Lighting at the
product contact surfaces within process vessels is a
unique and involved set of measurements to acquire,
as the distances and angles from which operators
view the product contact surface is dependent upon
available viewing ports and the differences in tank
configuration, shown in Figure 1. Measurement of
the distances and angles is most reliably performed in
the field using a laser distance meter and clinometers;
however, this information can also be determined
from mechanical drawings of the equipment.
As process equipment is surveyed, it is advisable
to develop a baseline record of known blemishes,
marks, imperfections, and visual obstructions to
ensure that they can be distinguished from post-pro-
duction residues. Establishing this baseline for each
process tank and training inspectors on the location
and appearance of these fingerprint imperfections
are critical to prevent the development of a viewing
bias that may impact the care used when inspecting
process equipment surfaces.
Characterized and appropriately restricted view-
ing angles developed during the survey will allow for
better translation of the VRL from the laboratory to
figure 1: Determination of Viewing Distance and Angles. the floor.
Conditions in areas where manually cleaned
equipment or small parts are inspected after they
also primarily done with less efficient incandescent are cleaned out of place are important in that many
and full-spectrum lamps that inherently provided pieces of manufacturing equipment used for closed
good color rendering. However, the advent of current processing operations do not allow for reliance on
sustainable design, legislated energy conservation ambient lighting and require the specification of an
requirements, and project engineers that target seem- independent light source for visual inspection. Ac-
ingly non-critical lighting designs in value engineer- cordingly, it is important to determine the maximum
ing exercises means that manufacturers can no longer inspection distance based on the size and configura-
assume that room lighting conditions will be consis- tion of vessels and other closed processing equip-
tent or even adequate for visual inspection. Specifi- ment. Once this is determined, it can be used not
cally, current legislation limits energy use for lighting only as a selection criterion for a light source but also
to 1.12.0 w/ft, depending on the space function, as one of the constraints for conducting laboratory
forcing less experienced lighting designers to select VRL studies.
options that may not provide equivalent performance
to historical lighting choices. thE SElEction of A light SourcE for ViSuAl
Indeed, Forsyth et al noted that lighting levels can inSpEction
vary from 400 to 1200 lux in a facility; such a range Before conducting the laboratory study or send-
is representative of that found in most manufactur- ing technicians into the field to inspect equipment,
ing facilities (2). Such variability is often unavoidable it is important to specify a hand-held light source.
as facility and process layout can change over time Specifying a hand-held or tank-mounted light source
to meet manufacturing needs, leading to shadows or carries some additional considerations. This requires
reflections unaccounted for in the original design. understanding some lighting design terminology and
Accordingly, it is recommended that the lighting be heuristics. The hand-held light should be selected
assessed and recorded through the use of commonly with three primary characteristics taken into con-
available light meters to acquire custom-tailored sideration: color temperature, color rendering index
ranges for developmental VRL studies. (CRI), and light output or luminous flux.
The most convenient way of recording these Color temperature, or chromaticity, refers to the
numbers is to record them on a layout drawing of the color of the light emitted from the light source. Colors
facility. Furthermore, it is useful to note the style of similar to the warm yellow orange of a candle flame

46 Special edition: Cleaning Validation


Keith Bader & Kelly Scalva

Source CRI Chromaticity (K)


Incandescent 100 2800K
Halogen 100 2900K
Fluorescent 6290 27006000K
Mercury 15 5700K
High Pressure Sodium 22 2000K
Metal Halide 6570 27004000K
Pulse Start Metal Halide 6575 30004000K figure 2: Light Intensity vs. Distance.

are assigned a temperature of 1800 Kelvin; up the


scale is the bluish light cast by metal halide and the lamps as shown in the Table. Furthermore, a flash-
fluorescent light sources that are assigned a color light with an adjustable spot size can also be used to
temperature of 4200 Kelvin (3). The ability of observ- vary the intensity and provide greater flexibility over
ers to accurately perceive colors is also somewhat de- a range of distances.
pendent on the chromaticity of the light source based
on the second characteristic noted above, CRI. lABorAtory Vrl dEtErminAtion StudiES
CRI is quantified on a scale that assigns the light Translating the angle, distance, and lighting param-
source a rating from 0 to 100, with the best rendition eters from the onsite equipment to the confines of
of color achieved with a CRI of 100. For detail-orient- the laboratory is best examined within a range of
ed physical inspection work and accurate evaluation values. Most commonly tested in the lab are three
of residues on process equipment surfaces, the CRI angles (30, 45, 90), two to three distances (clos-
of a light source should, at a minimum, be at a value est and farthest away), a minimum of two different
of 65 or greater (4). The general relationship between lighting intensities (standard room task lighting and
color temperature, CRI, and various light sources is the intensity from a hand-held light source), and a
shown in the Table. variety of materials of construction (MOC). Assum-
Finally, the light output of the source should be ing the viewing variables were tightly and accurately
considered. Many are also tempted to get the bright- measured at the manufacturing site, the only other
est light they can find; however, this can lead to test variables to be investigated in the laboratory
problems with detecting residues because excessive are the process soils (in varying concentrations),
glare can actually inhibit detection. The inspection materials of construction, and the assessment opera-
of process vessels is thus facilitated by selecting a tors. As the potential variability in technicians is
tank light or handheld light source that replicates the most difficult parameter to control and replicate,
levels of lighting equivalent to the recommended for they are discussed in greater detail in a subsequent
ambient task lighting within the vessel. Most light section.
manufacturers, however, do not provide the luminous
intensity at various distances from the source. Hand- ExpErimEntAl dEtAilS And logiSticS
held light output, when rated, is often quantified First, coupons composed of materials of construc-
using lumens out-of-the-front (OTF). On occasion, a tion representative of process equipment should be
specific intensity may be provided in Lux (lumen/m 2) obtained (see Figure 3) and cleaned thoroughly to
for some lamps at a specific distance. remove any possible residue on the coupons; such
To convert intensity at a given distance to one as dust, mechanical cutting oils; or, if the coupons
matching the distances determined for the produc- are not new, residue from previous tests. Typically,
tion equipment in the field, it is important to know a cleaning regimen consistent with United States
that light intensity follows an inverse square relation- Pharmacopeia (USP) <1051> for cleaning glassware
ship relative to distance, as shown in Figure 2. As employed for total organic carbon analysis will effec-
the distance from the light source increases, the light tively remove materials that can potentially confound
must diffuse over a larger surface thereby reducing results. Cleanliness can then be confirmed using the
the surface brightness in accordance with a one over American Society for Testing and Materials (ASTM)
d squared relationship. Alternatively, an inspec- water break free test (5, 6). Clean coupons are then
tion light of appropriate intensity can be determined rigorously inspected for visual cleanliness, surface
empirically in the laboratory by using a light meter imperfections, water spots, discoloration, or oxidation
at the appropriate distance. For the most part, an ac- before use in the VRL Study. Coupons that appear to
ceptable CRI and chromaticity are provided in hand- have any noticeable imperfections are rejected from
held light sources by either incandescent or halogen the study.

Special edition: Cleaning Validation 47


Keith Bader & Kelly Scalva

figure 4: Determining the Visibility Inflection Point.

includes a mixture of residue, purified water, and


isopropyl alcohol (IPA) to reduce the solution surface
tension and more uniformly distribute the residue
over the spiking area as the liquids dry, thereby
preventing concentration around the outer perimeter
or center of the spiking area. The exact ratios of IPA
to water must be determined on a per soil basis as
well as the solubility because each residue is differ-
ent. Furthermore, chemical interactions or protein
denaturation that alters the visual characteristics of
the soil should also be considered.
Once drying effects have been effectively contend-
ed with, the threshold concentration of each process
soil must be determined before multiple technicians
are screened. Specifically, the approximate concentra-
tion range visibility inflection point, when a residue
goes from visible to not visible, can be determined, as
shown in Figure 4.
Creating the initial spiking solutions in order of
magnitude intervals works well to initially narrow
the range. For example, a serial dilution of 1 ml resi-
due solution in 10 ml solvent followed by 1 mL of the
second solution in 10 mL of water, and so on, allows
a simple way to determine the range of concentra-
tions to be used for the VRL intermediate precision
figure 3: Coupons of Representative MOCs
testing assessment.
To prepare the coupons for testing, a consistent
Following a visual inspection of the coupons to volume of spiking solution is then spiked onto cou-
ensure their cleanliness, the process soils of inter- pons over a reproducible surface area. The concentra-
est are diluted into a range of concentrations. These tions applied to the coupons span the range of Vis-
concentration ranges are then spiked with equiva- ible, Slightly Visible, and Not Visible as determined
lent volumes onto the center of the coupon surface in the initial bulk VRL screening exercise described
and allowed to dry. Coupons commonly are dried at above. At least three replicates of each concentration
ambient conditions overnight to ensure dryness, but per material of construction should be used to ensure
they can be dried at elevated temperatures depending statistical significance within the results. Four opera-
on process conditions. A visual target range is then tors with 20/20 or corrected 20/20 vision are then
determined to find the appropriate loading density selected to establish the VRL intermediate precision
and loading solvent for use in VRL testing. testing at different viewing angles and distances un-
Purified water is the common solvent used to make der ambient and additional hand-held lighting.
the residue dilutions; however, surface tension ef- To better simulate visual inspection conditions
fects combined with some process soils can create a on the manufacturing floor, it is important that the
very visible ring around the periphery of the original coupons are placed on a background made of visually
spiking area. As water dries at the edges of the spiked similar material. The visually similar material reduc-
solutions, it concentrates the residue, making them es operator bias during the laboratory study so that
more visible and quantification of the mass per unit contrast from the viewing background will not be
surface area more difficult. This concentrates the an issue. Thus, when testing the viewing of different
residue to a greater degree in the laboratory setting materials of construction, the background for these
than that found on process equipment. Thus, it is assessments must either be constructed of the same
sometimes nessary to create a dilution mixture that material as the coupon or must be consistent with the

48 Special edition: Cleaning Validation


Keith Bader & Kelly Scalva

in the lab will not have the geometric restrictions


imposed by ancillary equipment, viewing ports, and
manways, thereby necessitating artificial limitations
on the amount of movement they may instinctively
employ as they try to observe spotted coupons. A
representative arrangement of coupons is shown in
Figure 5.

rEcommEndAtionS for tESting


In any quality system, the integration of quality-
by-design principles starts with the education of all
personnel involved in the production of a therapeutic
substance. Specifically, when developing training ma-
terials for visual inspection of the process equipment,
operators should be informed of the importance of
the activity as an important orthogonal method for
the prevention cross contamination. This will help
prevent technicians from treating the activity as just
figure 5: VRL Determination Conditions Based on Manufacturing Area
another task and rushing to complete it at the poten-
Assessment.
tial peril of patients receiving a particular therapeutic
appropriate viewing parameters employed for the pro- compound.
cess equipment. For example, if the major material of In the laboratory setting, the study was most influ-
construction is 316L SS EP20Ra with minor materi- enced by the level of training of the visual assessment
als such as borosilicate glass (commonly site glasses) operators. For example, operators who had experi-
and/or PTFE (primarily tested to simulate valves), the ence cleaning coupons in the lab were much more
minor materials should have viewing backgrounds adept at dismissing imperfections on the surface of
equivalent or comparable to the major material of coupons that may have otherwise confused those
construction. For example, 316L, 316, and 304 stain- who had never before conducted visual inspection or
less steel would be visually comparable materials. If VRL testing with residues. Not only is it important
glass were the major material of construction, match- for operators to know and understand why they are
ing the background could also be somewhat complex completing a task, but it is just as important for the
since interactions between process equipments and operators to have the chance to study a dirty vessel
the manufacturing area floors or walls could lead to a directly after a process run and at the end of the dirty
more difficult situation to emulate in the laboratory. hold time in addition to having historic areas of con-
Other variables to consider are the impact of personal cern and vessel imperfections pointed out to them.
protective equipment (PPE) worn in the manufactur- A well-educated operator trained to recognize the
ing space, such as goggles, safety glasses, laboratory appearance of process equipment at both its worst
coats, coveralls, or scrubs. The spiked coupons are and best relative to cleanliness is much more likely
then arranged in a viewing area with controlled light- to properly ascertain the presence of residues on
ing and background. product contact areas. Specifically, operators should
To control for variations in visual acuity, operators also have the opportunity during training to visually
were screened to ensure that they had either correct- inspect the vessel directly after the cleaning cycle.
ed or uncorrected 20/20 vision. Those without 20/20 If possible, they should also be able to observe the
vision were excluded from the study. Screening was equipment in a clean, wet state and a completely dry
conducted by first asking each operator if they had state since residue is typically better seen on dry pro-
had a professional eye exam within the last twelve cess surfaces. Knowledge of how the vessel appears
months. If so, the operator was asked to view two dif- when the surfaces are covered in process residues
ferent Snellen eye charts from distances of 6 ft. and typically makes the cleanliness easier to assess if an
10 ft., respectively. A passing result for this portion idea of the range of cleanliness is known.
of the test required that the operator could correctly Bias can also be avoided in the laboratory evalu-
recite the line on the chart corresponding to 20/20 ation by arranging the coupons both in a random
without reading any of the letters incorrectly. This loading concentration order and in rows of, or greater
practice is recommended to qualify technicians both than, five coupons to prevent operators from memo-
in the laboratory and on the manufacturing floor. rizing which coupons they identified as soiled under
When transferring the measured angles and distances a previous lighting, distance, and viewing angle
to the laboratory, keep in mind that technicians combination. Based on the authors experience, more

Special edition: Cleaning Validation 49


Keith Bader & Kelly Scalva

consistent results are also obtained if the operators ing agents that degrade post-production residues. As
verbally relay whether the residue is visible rather with any chemical change to a compound, the physi-
than be required to fill out protocol worksheets. cal properties can change, including visibility of the
Overall, it was found that slight movement of the compound on process surfaces. Future studies will
flashlight was necessary for the operators to attempt investigate this and other similar considerations.
to see the residues; directing the light towards the
coupon led to excessive glare that hindered the ability rEfErEncES
of the operator to view residue. Residues were typi- 1. E. Teicholz, LIGHTING in Facility Design and Management
cally noted when they were on the periphery of the Handbook, (McGraw-Hill Professional, 2001), AccessEngineer-
hand-held light source spot. It was also noted that ing.
operators who viewed the surfaces longer generally 2. R.J. Forsyth, V. Van Nostrand, and G. Martin, Visible-Resi-
saw more residue than operators who quickly glanced due Limit for Cleaning Validation and its Potential Applica-
at the coupons. However, with the use of the flash- tion in a Pharmaceutical Research Facility, Pharmaceutical
light, operator-viewing duration increased by about Technology 28 (10), 5872, 2004.
50%. Interestingly enough, visibility and detection 3. T. Croft, W.I. Summers, F.P. Hartwell, PRINCIPLES AND
of dirty coupons decreased when operators used the UNITS in American Electricians Handbook, Fifteenth Edition,
hand-held light over diffuse ambient lighting. (McGraw-Hill Professional, 15th ed., 2009 2002 1996 1992
While the procedures described above may seem 1987 1981 1970 1961).
fairly complete and extensive, there are still many re- 3. R.C. Rosaler, Lighting in Standard Handbook of Plant Engi-
lated topical permutations warranting further inves- neering, Third Edition (McGraw-Hill Professional, 3rd ed.,
tigation. For example, most studies, to date, focus on 2002 1995 1983)
the intact API; however, cleaning processes employed 4. USP29NF24 <1051>, Cleaning Glass Apparatus, 2896
for biopharmaceuticals often employ alkaline clean-

50 Special edition: Cleaning Validation


Rizwan Sharnez

Cleaning Validation of
Multiproduct Equipment
Acceptance Limits for
Inactivated Product
Part IIApplication of the Comparable
Quality Approach to Intrasite Assessments

Rizwan Sharnez, Elizabeth Aisenbrey, Joel Bercu, David Binkley, and Arun Tholudur

New Perspectives on Cleaning is an ongoing series of articles dedicated


to cleaning-process development, validation, and monitoring. This column
addresses scientific principles, strategies, and approaches associated with
cleaning that are relevant in everyday work situations.
Reader questions, comments, and suggestions are requested for future
discussion topics. These can be submitted to Rizwan Sharnez at rsharnez@
amgen.com.

SUMMARY
For multiproduct cleaning validation, acceptable carryover of the previ-
ously manufactured active pharmaceutical ingredient (API) (Product A) into
the subsequently manufactured API (Product B) is determined through a
maximum allowable carryover (MAC) calculation (1). A limitation of the
conventional MAC approach is that if the previously manufactured API
becomes therapeutically inactive during cleaning, then there is limited value
in verifying removal of the API. Instead, it is more appropriate to demon-
strate that the carryover of the inactivated product between lots of different
products (i.e., A B, or B A) is acceptable.
If the API is inactivated when exposed to worst-case cleaning condi-
tions (i.e., conditions that are least conducive for inactivation), the compa-
rable quality (CQ) approach can be used to set acceptance limits for car-
ryover of the inactivated product (IP) (2). The CQ approach is designed to
ensure that the amount of inactivated Product A in the largest dose (LD)
of Product B (M LDIP AB ) is less than or equal to the amount of inactivated
Product A in the largest dose of Product A (M LD IP AA),

M LD
IP AB M LD
IP AA [Equation 1]

To apply the CQ criterion to cleaning validation, the above inequality


must be expressed in terms of measureable equipment and product pa-
rameters. An approach for expressing this criterion in terms of analytical
data (e.g., total organic carbon [TOC] swab or rinse data), product contact
surface area, and batch and dose sizes is described in this paper. The
resulting expression for the CQ criterion for intrasite assessments (i.e.,
where Product A is being manufactured at the same site where Product
B is being manufactured) is given by Equation 2. When this inequality is
satisfied, the amount of inactivated Product A in the largest dose of Prod-
uct B will be limited to the amount of inactivated Product A in the largest
dose of Product A.

Special edition: Cleaning Validation 51


Rizwan Sharnez

If the characterization data indicate that the previ- because the concentration of the degradants in clean-
ously manufactured API (i.e., API in Product A) is in-place (CIP) samples is generally well below the limit
inactivated when exposed to cleaning conditions, and of detection (LOD) of SDS-PAGE and most bioassays.
the CQ criterion is met, then the acceptance limits In the small-scale studies, this issue is addressed by
for the process residue can be set based on the lowest using a soil to solution ratio (R) that is high enough
of the following two limits: process capability and 10 to ensure that the concentration of degraded product
ppm limit for carryover of process residue. in the sample is above the LOD of the assay. Note
A flowchart summarizing the methodology de- that a higher R represents a milder condition from the
scribed in this paper is shown in Figure 1. The CQ cri- standpoint of degradation (i.e., higher R implies that a
terion is derived in Appendix I and a worked example smaller fraction of the total soil would degrade).
is given in Appendix II.
The CQ approach was described in Part I of this Implications for Cleaning Validation
series. In Part II, the CQ approach is applied to intra- The inactivation of the API during cleaning has impor-
site assessments; application of the CQ approach to tant implications for cleaning validation of multiproduct
intersite transfers is described in Part III. equipment. Demonstrating that the product is inactivat-
ed during cleaning obviates the need to set limits based
INTRODUCTION on a conventional MAC assessment for the API (5). It also
Biopharmaceutical cleaning cycles are generally designed eliminates the need to develop product specific assays for
to expose product contact equipment to extremes of cleaning validation (6).
pH (pH 213) and temperature (60-80C) for several
minutes. The equipment may also be steam sterilized or The Comparable Quality Approach
sanitized. Under these conditions, monoclonal antibod- If the API is inactivated when exposed to worst-case
ies, therapeutic proteins, and other biological APIs are cleaning conditions, the CQ approach can be used to
known to degrade and denature rapidly, and become set acceptance limits for cleaning (2). The CQ approach
therapeutically inactive (2-4). ensures that the amount of inactivated Product A in
the largest dose of Product B is limited to the amount
Inactivation Studies of inactivated Product A in the largest dose of Product
Inactivation of the API during cleaning can be assessed A. Thus, the CQ approach provides assurance that in a
by exposing the process soil to simulated cleaning single exposure the amount of inactivated Product A that
conditions at bench scale. The inactivation studies are a patient taking Product B receives is less than or equal
designed to simulate full-scale cleaning conditions that to the amount of inactivated Product A that a patient tak-
are least conducive (i.e., worst case) for inactivation (e.g., ing Product A receives. The rationale for this approach is
lowest ratio of cleaning solution to protein and shortest that if one set of patients (i.e., those taking Product A) are
exposure). The objective of these studies is to ascertain being exposed to a certain amount of inactivated Product
whether the API in the process sample is inactivated A, then it is acceptable for another set of patients (i.e.,
when exposed to worst-case cleaning conditions. those taking Product B) to be exposed to a lesser or equal
After exposure to the worst-case cleaning condi- amount of inactivated Product A.
tions, the samples are titrated to a neutral pH and The CQ approach is based on the premise that
cooled to 4C to minimize further degradation and in- an appropriate product quality assessment has been
activation. Appropriate untreated controls are included completed on Product A to show that Product A is of
to assess the impact of any experimental artifacts and acceptable quality.
potential matrix effects. The samples are then sub-
jected to SDS-PAGE and bioassay to determine the Differences in Patient Subpopulations and Frequency
degree of degradation and inactivation, respectively. or Duration of Dosing
The samples may also be subjected to TOC analysis For pharmacologically-active substances, therapeutic ef-
to determine whether the inactivated product can be fects and toxicity that occur from a single peak exposure
adequately recovered by TOC. or continually over time are important considerations for
The results of the small-scale studies can justifi- setting acceptable limits for exposure. In order to study
ably be extrapolated to the full-scale cleaning process, the effects of key variables such as dosing frequency,
because degradation and inactivation are essentially length of exposure, and sensitivity, active substances are
scale-independent phenomena (i.e., they depend on extensively tested in the patient subpopulations tak-
cleaning parameters that are effectively independent ing the drug. While these endpoints are important for
of spatial coordinates, namely time, temperature, con- the study of active substances, they are not relevant to
centration, and the ratio of cleaning solution to process degradants that are pharmacologically inactive. Instead,
soil). It should be noted that degradation and inactiva- the comparable CQ approach is more appropriate for
tion studies are typically not performed at full scale establishing limits for inactive degradants.

52 Special edition: Cleaning Validation


Rizwan Sharnez

Perform
inactivation
study a

Is API in process sample


inactivated after exposure
to cleaning conditions? b
Yes No

If CQ criterion is Use conventional MAC


satisfiedc set acceptance approach to
limit (AL) for inactivated set acceptance limit (AL)
product (IP) d per Eqn. 2 for API e

Can IP be Can API be


recovered by recovered by
TOC? TOC?
Yes No Yes No

Is AL LOQ No Is AL LOQ No
of TOC? of TOC?

Use TOC to Develop Use TOC to Develop


demonstrate alternative assay demonstrate alternative assay
removal of to demonstrate removal of to demonstrate
IP removal of IP API removal of API
a
Inputs: process sample, reference standard, cleaning cycle, and equipment parameters
b
Based on bioassay analysis
c
If CQ criterion is not satisfied, the approach described in this paper is not applicable
d
Inputs: dose and batch sizes of product, surface area of dedicated and shared equipment,
and surface concentration of IP (refer to Appendix 1.2)
e
Inputs: dose and batch sizes of product, surface area and rinse parameters of shared equipments [8]

Figure 1: Flowchart for setting acceptance limits for multiproduct cleaning validation.

Effect of Cleaning Process results of the bioassay), then the CQ approach is used
Note that for a given cleaning process for Product A, to set acceptance limits (AL) for the inactivated product
the molecular weight distribution (MWD) of Product A (left side of Figure 1). However, if the sample is shown to
that is carried over into a subsequent lot of Product A have biologically active product, the conventional MAC
(intracampaign cleaning) will be similar to the MWD approach is used to set the AL for the previously manu-
of Product A that is carried over into a subsequent lot of factured product (APIA) (right side of Figure 1). If the
Product B (intercampaign cleaning). It is also important process residue (inactivated product or API, as the case
to note that no additional inactivated product is intro- may be) can be recovered by TOC and the correspond-
duced into either product as a result of implementing the ing AL the LOQ of TOC, TOC may be used to verify
CQ approach. removal of the process residue at full scale; otherwise, an
A literature search did not uncover methods for cal- alternative assay may need to be developed.
culating cleaning validation acceptance limits for the
carryover of inactivated product. COMPARABLE QUALITY CRITERION IN TERMS OF
MEASURABLE PARAMETERS
Setting Acceptance Limits If the product becomes therapeutically inactive during
Based on Protein Inactivation cleaning, then the acceptance limit for the inactivated
A methodology for setting acceptance limits for multi- degradants of the previously manufactured product
product equipment is summarized in Figure 1. Degrada- (Product A) in the subsequently manufactured product
tion and inactivation studies are first performed under (Product B) can be set based on the CQ criterion. In
simulated worst-case cleaning conditions. If the sample terms of the carryover of inactivated product, the CQ
is shown to have no detectable activity (based on the criterion can be stated as follows.

Special edition: Cleaning Validation 53


Rizwan Sharnez

The amount of inactivated


LD
Product A in the largest and the CQ criterion per Equation 2 is met, then the
dose (LD) of Product B (M IP AB ) should be less than acceptance limit for the process residue can be set based
or equal to the amount of inactivated
LD
Product A in the on the lowest of the following two limits:
largest dose of Product A (M IP AA) (2). Thus, Process capability. This is based on either the
capability of the equipment or the capability of
M LD
IP AB M LD
IP AA [Equation 1] the CIP circuit to clean a specific process resi-
due. Note that the CQ approach is based on the
To apply this criterion to set acceptance limits for use of historical cleaning data to support the
inactivated product, the above inequality must be baseline amount of inactivated Product A in a
expressed in terms of measureable equipment and dose of Product A. Thus, the use of historical
product parameters. An equation for expressing this cleaning data to set acceptance limits for clean-
criterion in terms of analytical data, product contact ing is justified. If the historical data are below
surface area, and batch and dose sizes is derived in the limit of quantitation (LOQ) of the assay, the
Appendix I. For intrasite assessments, where the process capability limit can be set to the LOQ of
intracampaign cleaning cycle (AA) is similar to the the assay. This approach is consistent with indus-
intercampaign cleaning cycle (AB), the result is as try practices. It is justified because the cleaning
follows (Equation AI-5): cycle is validated and continually monitored, and
has appropriate controls in place. Additionally,
Ni=1 [ CAA,i SA AA,i] SBS A LD B
[Equation 2] adopting this approach does not impact the rela-
Mj=1 [CAB,j SA AB,j ] SBS B LD A
tive amount of degraded product that is carried
LD
where, over (i.e., M LD
IP AA vs. M IP AB), which is consis-
SAAA,i is the total product-contact surface area tent with the CQ criterion.
for the ith system (CIP circuit) of the equipment 10 ppm limit for carryover of process residue.
train used to manufacture Product A This is a widely used default limit (8) that is
N is the total number of systems that are used to based on the criterion that the cumulative car-
manufacture Product A ryover of process residue from the previously
SAAB,j is the product-contact surface area manufactured batch into the subsequently manu-
for the jth system of the equipment train that is factured batch should be 10 ppm. Note that
shared between Products A and B the raw data from the analytical instrument
M is the number of systems that are shared should be divided by the appropriate swab or
between the two products rinse recovery factor when estimating the residue
CAA,i and CAB,j are average surface concen- on the surface. Additionally, if TOC is used to
tration of inactivated Product A on SAAA,i and analyze the sample, the default limit should be
SAAB,j. The average surface concentrations multiplied by the fraction of carbon in the pro-
can be estimated from swab and rinse data as cess soil to account for the amount of carbon in
described in Section AI.2 of Appendix I. the residue.
SBSA and SBSB are the smallest integral (7) batch
size of Product A and B, respectively CONCLUSION
LDB and LDA are the largest doses of Products A An important consideration in multiproduct clean-
and B, respectively. ing validation is to demonstrate that the carryover of
If inactivated Product A can be recovered by TOC the previously manufactured API into a batch of the
assay, then TOC is used to verify removal of the subsequently manufactured product is below an accept-
inactivated product; otherwise, an alternative assay is able limit. If, however, the previously manufactured API
developed to demonstrate removal of the inactivated becomes pharmacologically inactive during cleaning,
product at full scale. then there is limited value in verifying removal of the
The CQ criterion can be applied to smaller sections API. Instead, from a patient safety standpoint, it is more
of the total equipment train. For instance, if the drug meaningful to demonstrate that the inactivated product
substance (DS) and drug product (DP) are manufac- is adequately removed.
tured in different facilities, it may be more efficient Inactivation of the API during cleaning can be
from a change management perspective to decouple the assessed by exposing the process soil to simulated
CQ assessments for the DS and DP equipment trains. cleaning conditions at bench scale. The degree of
inactivation is evaluated by subjecting the sample and
SETTING ACCEPTANCE LIMITS untreated controls to the appropriate assays (e.g., SDS-
FOR PROCESS RESIDUE PAGE and bioassay for evaluation of degradation and
If the characterization data indicate that the API de- biological activity, respectively).
grades when exposed to worst-case cleaning conditions,

54 Special edition: Cleaning Validation


Rizwan Sharnez

If the protein is inactivated during cleaning, then Active Ingredients as Caused by Various Process
the acceptance limit for the inactivated degradants of Cleaning Agents and Temperature, Journal of Vali-
the previously manufactured product (Product A) in dation Technology, Vol 15, No. 3, p. 69, 2009.
the subsequently manufactured product (Product B) 4. Rathore, N., Qi, W., Chen, C., Ji, W., Bench-scale
can be set based on the CQ criterion. The CQ ap- characterization of cleaning process design space
proach provides assurance that the maximum amount for biopharmaceuticals, Biopharm Int, Vol 22, No.
of inactivated Product A that a patient taking Prod- 3, 2009.
uct B receives (M LD
IP AB) is limited to the maximum 5. Parental Drug Association, Inc., PDA Technical Report
amount of inactivated Product A that a patient taking 49 Points to Consider for Biotechnology Cleaning Vali-
Product A receives (M LD IP AA). The rationale for this dation, July 2010.
approach is that if one set of patients (i.e., those taking 6. Health Canada, Cleaning Validation Guidelines
Product A) are being exposed to a certain amount of (GUIDE-0028); Section 8.3, Health Products and
inactivated Product A, then it is acceptable for an- Food Branch Inspectorate, 2008.
other set of patients (i.e., those taking Product B) to 7. Note: i.e., before the original manufacturing batch is
be exposed to a lesser or equal amount of inactivated subdivided into smaller batches for processing (e.g.,
Product A. filling).
In terms of the mass of the inactivated product, the 8. Note: i.e., if the calculated acceptance limit is above
CQ criterion is given by Equation 1. the default limit, then the default limit is used; con-
In order to use this criterion to set acceptance limits versely, if the calculated acceptance limit is below
for inactivated product, the Equation 1 inequality the default limit, then the acceptance limit is used.
was expressed in terms of measureable equipment
and product parameters (Appendix I). The resulting ARTICLE ACRONYM LISTING
expression for the CQ criterion for intrasite assess- A Product A Previously manufactured prod-
ments, where the intracampaign cleaning cycle (AA) uct
is similar to the intercampaign cleaning cycle (AB), is AA Between batches of the same product (in-
given by Equation 2. tracampaign)
When this inequality is satisfied, the amount of AB Between batches of different products (inter-
inactivated Product A in the largest dose of Product B campaign)
will be less than the amount of inactivated Product A ADE Acceptable Daily Exposure
in the largest dose of Product A. API Active Pharmaceutical Ingredient
If inactivated Product A can be adequately recovered B Product B Subsequently manufactured
by TOC, then TOC is used to verify removal of the product
inactivated product; otherwise, an alternative assay is CQ Comparable Quality
developed to demonstrate removal of the inactivated DP Drug Product
product at full scale. DS Drug Substance
If the characterization data indicate that the API is LD Largest Dose
inactivated when exposed to cleaning conditions, and LOQ Limit of Quantitation
the CQ criterion is met, then the acceptance limits M LD
IP AA Mass of inactivated Product A in largest dose
for the process residue can be set based on the lowest of Product A
of the following two limits: process capability and 10 M LD
IP AB Mass of inactivated Product A in largest dose
ppm limit for carryover of process residue. of Product B
Application of the CQ criterion to intersite transfers MAC Maximum Allowable Carryover
will be described in Part III. MWD Molecular Weight Distribution
SDS PAGE Sodium Dodecyl Sulfate Polyacrylamide Gel
REFERENCES Electrophoresis
1. Fourman, Gary L. and Michael V. Mullen, Deter- SBS Smallest Batch Size
mining Cleaning Validation Acceptance Limits for TOC Total Organic Carbon
Pharmaceutical Manufacturing Operations, Phar-
maceutical Technology, 17 (4), 54-60, 1993. ACKNOWLEDGEMENTS
2. Sharnez, R., To, A., Cleaning Validation of Multi- The authors thank Aine Hanly, Donna Corvese, Lillian
product Equipment: Acceptance Limits for Inac- Colon, Sam Guhan and Steve Hatke for their help and
tivated Product, Part IThe Comparable Quality support.
Approach, Journal of Validation Technology, Autumn
2011, p. 32-36, 2011.
3. Kendrick, K., Canhoto, A., Kreuze, M., Analysis
of Degradation Properties of Biopharmaceutical

Special edition: Cleaning Validation 55


Rizwan Sharnez

APPENDIX I facture Product A (Figure AI.2). The surface area that


both products are exposed to, also known as the
Derivation of CQ Criterion shared surface area, is SA AB.
Similarly, the carryover of residual inactivated
Product A (that remains on the equipment surface after
batch
Derivation of the comparable quality (CQ) criterion in cleaning), into a batch of Product B (M IP,AB) can be
terms of measureable equipment and product parameters expressed as:
is described below.
In terms of the carryover of inactivated product, the M

[C
batch
CQ criterion states that: M IP,AB = AB,j
SA AB,j] [Equation AI-2b]
The amount of inactivated Product A in the largest j=1
dose (LD) of Product B (M LD
IP AB) should be less than
or equal to the amount of inactivated Product A in the where, SA AB,j is the product contact surface area for
largest dose of Product A (M LD
IP AA) (2). Thus, the jth system of the equipment train (cm2), M is the
number of systems that are shared between the two
products, and CAB,j is the average surface concentration
M LD
IP AB M LD
IP AA [Equation AI-1] of inactivated Product A on SA AB,j (g/cm2).
Thus, the carryover of inactivated Product A into
AI.1 CQ criterion in terms largest dose of Product B (M LD
IP AB) can be expressed
of measureable parameters as:
N


In order to apply this criterion to set acceptance limits LDB
for inactivated product, the above inequality must be ex- M IP,AB=
LD
[CAB,j SA AB,j]
pressed in terms of measureable equipment and product j=1 SBSB
parameters. This is demonstrated in Figure AI.1 with an [Equation AI-3b]
example in which two products, A and B, are subse-
quently manufactured in a shared equipment train. where, LDB and SBSB are the largest dose and smallest
The equipment train shown in Figure AI.1 is used batch size of Product B, respectively.
to manufacture Product A and has a total surface area Substituting for the left-hand and right-hand sides
of SA AA. of Equation AI-1 with the right-hand side of Equations
The carryover of the residual inactivated Product A AI-3b and AI-3a, respectively, gives:
(that remains on the equipment surface after batchclean- M N

[C
LDB LDA
ing), into the next batch of Product A (M IP,AA) can be [CAB,j SA AB,j] AA,i
SA AA,i ]
SBSB SBSA
expressed as: j=1 i=1

[Equation AI-4]
N

[C
batch
M IP,AA = AA,i
SA AA,i] [Equation AI-2a] Rearranging the terms in Equation AI-4 gives:
i=1


N
[CAA,i SA AA,i ] SBSA LDB
where SA AA,i is the product contact surface area for the i=1


ith system (CIP circuit) of the equipment train (cm2), N M SBSB LDA
is the total number of systems that are used to manufac- j=1
[CAB,j SA AB,j ]
ture Product A, and CAA,i is the average surface concen- [Equation AI-5]
tration of inactivated Product A on SA AA,i (g/cm2).
Thus, the carryover of inactivated Product A into the Equation AI-5 gives the CQ criterion for intrasite
largest dose of Product A (M LD
IP AA) can be expressed as: manufacturing (i.e., when Product A is already being
manufactured at the same site where Product B is man-
N


LDA ufactured). Note that N is the total number of systems
M IP,AA=
LD
[CAA,i SA AA,i] used in the manufacture of Product A, and M is the
i=1 SBSA number of systems that are used in the manufacture of
both Product A and Product B (i.e., shared systems).
[Equation AI-3a]
AI.2. Surface concentration in terms of swab or rinse
where LDA and SBSA are the largest dose and smallest data
batch size of Product A, respectively. The surface concentration terms in Equation AI-5
Part or all of the equipment train that is used to (CAA,j and CAB,j) can be expressed in terms of TOC
manufacture Product B will also be used to manu- (or other analytical) results as follows:

56 Special edition: Cleaning Validation


Rizwan Sharnez

Upstream Downstream Fill/Finish

Figure AI.1: Equipment train used to manufacture Product A has a total surface area of SA AA

Upstream Downstream Fill/Finish

Figure AI.2: The surface area that both products will be exposed to, also known as the shared surface area, is SAAB
S


1 swab recovery factor and carbon fraction for the process
CAA,i= CAA,k,i [Equation AI-6] soil in the ith system.
S k= 1 Similarly, CAA,k,i can be estimated from TOC rinse
data as follows:
where S is the number of data points (samples) for the
ith system (CIP circuit) and CAA,k,i are the measured
CAA,k,i =rTOCk,i
surface concentrations of inactivated product for each of [Equation AI-7b]
the S data points. rRFi cfi
The CAA,k,i for the equipment train used to manu-
facture Product A can be estimated from TOC swab where, rTOCk,i is the TOC rinse data for the kth rinse
data as follows: sample result for the ith system, and rRFi and cfi are the
rinse recovery factor and carbon fraction for the process
CAA,k,i =sTOCk,i soil in the ith system.
[Equation AI-7a] Similarly, CAB,j can be estimated from TOC swab
sRFi cfi or rinse data, recovery factors and carbon fractions for
the shared equipment.
where, sTOCk,i is the TOC swab data for the kth swab
sample result for the ith system, and sRFi and cfi are the

Special edition: Cleaning Validation 57


Rizwan Sharnez

APPENDIX II

N
i=1
[CAA,i SA AA,i ] SBSA LDB
Worked Example for Intrasite
SBSB LDA
M
Assessment
j=1
[CAB,j SA AB,j ]
Is the CQ criterion satisfied for the following scenario?:
Product A and Product B are currently being The calculated parameters for this example are as
manufactured at the same site. The total surface area given in Table AII-4.
associated with the manufacture of Product A and
3

[C
relevant process parameters are given in Table AII-1.
SA AA,i ]= (3.7112.6)+(2.1410.2)
TOC rinse data for the above circuits are listed in i=1
AA,i

Table AII-2. In this case, for each circuit there is one +(3.1511.6)=105.1
TOC rinse sample. 2

Part of the equipment train used to manufacture


Product A is also used to manufacture Product B. Cir-
[C
j=1
AB,j
SA AB,j] = (2.1410.2)+(3.1511.6)=58.4

cuits 2 and 3 are shared for the manufacture of Prod- SBSA 4.5
uct A and Product B. The shared surface area SA AB,j = = 0.9
SBSB 5
and relevant process parameters for the manufacturing
Product B are as given in Table AII-3. LDB 75
= = 1.5
Because the cleaning cycles for AA are the same as LDA 50
the cleaning cycles for AB, CAA,i = CAB,j for Cir-
cuits 2 and 3, and the TOC rinse data listed in Table Substituting the above results into the left and right
AII-2 can be used for both surface concentrations. hand sides of Equation AI-5 gives:
Left hand side = 1.80
AII. Solution Right hand side= 1.35
For this scenario (intrasite assessment), the CQ criterion Thus, the CQ criterion is satisfied for the above
is given in Equation AI-5: intrasite assessment. JVT

Table AII-3: Shared surface area for Products A and B and relevant process parameters.

Description of Parameter Abbreviation Value


Number of shared CIP circuits M 2
The shared surface area of circuit 2 (j=1) that is used to manufacture Prod-
SA A B,1 10.2 m 2
uct A and Product B
The shared surface area of circuit 3 (j=2) that is used to manufacture Prod-
SA A B,2 11.6 m 2
uct A and Product B
Smallest batch size of Product B SBSB 5 kg
Largest dose size of Product B LDB 75 mg

Table AII-4: Calculated parameters for intrasite assessment.

Calculated Parameter Equation Result Worked Example for i=1


C A A,1 AI-7b 3.71 ppm
C A A,2 AI-7b 2.14 ppm CAA,k,i =rTOCk,i
rRFi cfi
C A A,3 AI-7b 3.15 ppm
CAA,1,1=0.65 =3.71
C A B,1 C A B,1= C A A,2 2.14 ppm (0.70x0.25)

C A B,2 C A B,2=C A A,3 3.15 ppm

58 Special edition: Cleaning Validation


Rizwan Sharnez

Table AII-1: Total surface area associated with the manufacture of Product A and relevant process parameters.

Description of Parameter Abbreviation Value


Number of CIP circuits N 3
Surface area of circuit 1 (i=1) that is used to manufacture Product A SA A A,1 12.6 m 2
Surface area of circuit 2 (i=2) that is used to manufacture Product A SA A A,2 10.2 m 2
Surface area of circuit 3 (i=3) that is used to manufacture Product A SA A A,3 11.6 m 2
Smallest batch size of Product A SBSA 4.5 kg
Largest dose size of Product A LDA 50 mg

Table AII-2: TOC rinse data.

Description of Parameter Abbreviation Circuit 1 (j=1) Circuit 2 (j=2) Circuit 3 (j=3)


TOC rinse sample data for the i circuit th
rTOCi,j 0.65 ppm 0.2 ppm 1.1 ppm
Rinse recovery factor for the i circuit
th
rRFj 0.70 0.55 0.92
Carbon fraction for the i circuit
th
cfj 0.25 0.17 0.38

Special edition: Cleaning Validation 59


Rizwan Sharnez

Biopharmaceutical
Cleaning Validation:
Acceptance Limits for
Inactivated Product
Based on Gelatin as a
Reference Impurity | IVT
Rizwan Sharnez, Ph.D.,Abby Spencer, Jeanine Bussiere, Ph.D., Dan Mytych, Ph.D., Angela
To, and Arun Tholudur, Ph.D.

Biopharmaceutical cleaning and sterilization processes denature and degrade


the active pharmaceutical ingredient (API)i into fragments that are pharmaco-
logically inactive. A rational approach for setting safety-based acceptance lim-
its for inactive fragments is described. The approach is based on the use of
gelatin as a reference impurity. It is designed to ensure that the carryover of
inactive fragments between batches of different products is acceptable from a
predictive safety standpoint.
The scope of this paper is limited to process residues of non-conjugated
human therapeutic proteins. Nonetheless, the underlying principles may be
useful in setting acceptance limits for other types of inactive impurities.

INTRODUCTION
An important regulatory expectation for multiproduct cleaning validation
is that the potential carryover of the previously manufactured API into the
subsequently manufactured product should be below an acceptable level.
This criterion is often met through a maximum allowable carryover (MAC)
assessment for the previously manufactured API (1-5). The MAC assessment
is typically based either on the minimum therapeutic dose (1), or the accept-
able daily exposure (ADE) (2) of the previously manufactured API.
A limitation of the conventional MAC approach is that it does not
provide appropriate acceptance limits for pharmacologically inactive
product. This has important implications for biopharmaceutical manu-
facturing because the API is inactivated during cleaning and sterilization.
Another limitation of this approach is that the acceptance limit for the
API is often below the process capability limit (PCL) of the cleaning pro-
cesses and, in some instances, below the limit of quantitation (LOQ) of
non-specific assays, such as total organic carbon (TOC). Other limitations
of the conventional MAC approach have been discussed previously (6).
Fragmentation and inactivation of an API during cleaning and steriliza-
tion can be assessed by exposing the process soil to simulated operating
conditions at bench scale (7). The bench scale experiments are designed
to simulate full-scale operating conditions that are least conducive
(worst-case) for inactivation. The degree of inactivation is evaluated by
subjecting the sample and untreated controls to the appropriate assays
(e.g., sodium dodecyl sulfate polyacrylamide gel electrophoresis [SDS-
PAGE] and bioassay can be used to evaluate fragmentation and biological
activity, respectively). The results of the study are used to set appropriate

60 Special edition: Cleaning Validation


Rizwan Sharnez

acceptance limits for the process residue. The product ments of Product B (IFB). Thus, the IFA that are carried
inactivation approach is therefore more science-based over during changeover into Product B do not present
and reflective of the phenomenological aspects of a new or unknown risk from a safety standpoint. This
cleaning processes. implies that the equipment train can be used to manu-
Inactivation of the product during cleaning and facture multiple products without introducing a new or
sterilization has important implications for clean- unknown class of impurities into any of the products.
ing validation of multiproduct equipment. If the API Further, the carryover of IFA into Product B is significant
degrades into pharmacologically inactive fragments, only for the first lot of Product B that is manufactured
the acceptance limit for the process residue can be set following changeover.
based on the inactive fragments instead of the active
ingredient. This is consistent with the expectation that COMPARABLE QUALITY APPROACH
the carryover of an impurityin this case the inac- The acceptance limits for inactive fragments in the pro-
tive fragmentsinto the subsequently manufactured cess residue can be set based on the Comparable Quality
product should be justified from the standpoint of the (CQ) approach (6, 10-11). With the CQ approach, the
safety and efficacy of the product. amount of the target impurityin this case inactive frag-
The experimental approach and analytical methods ments of Product Athat is carried over into the largest
for assessing fragmentation and inactivation of the API dose (i.e., largest dose that is administered to a patient
have been discussed in the literature (6-11). This paper in a day) of the subsequently manufactured product
describes a rational approach for setting safety-based (Product B) is limited to the acceptable exposure per
acceptance limits for the inactive fragments. The pro- dose of a reference impurity. The reference impurity must
posed methodology builds upon previously published be comparable to or worse than the target impurity from
approaches (6, 10-11). a predictive safety standpoint.
Predictive safety for inactive fragments is evaluated
ASSESSING THE SAFETY OF in terms of the key factors that determine toxicity and
INACTIVE FRAGMENTS OF HTPS immunogenicity. For HTPs, toxicity is determined by
The safety of inactive fragments of human therapeutic pharmacological activity (13); thus, toxicity is generally
proteins (HTPs) is assessed qualitatively in this section. not a concern for inactive fragments of HTPs. Im-
Consider an equipment train that is used to manufac- munogenicity is primarily determined by foreignness
ture Product A. The cleaning and sterilization cycles are and chemical complexity (14). Chemical complexity
known to denature and degrade any residual product in increases with molecular weight (MW); thus, larger
the equipment into fragments that are pharmacologically molecules tend to be more immunogenic (15). The
inactive. The inactive fragments of Product A are carried most active immunogens tend to have a MW greater
over into the subsequent batch of Product A. Thus, as than 100 kilo Daltons (kDa) (14). HTP fragments with
a class of molecules, inactive fragments of HTPs do not MWs less than 10 kDa are generally weak immuno-
present a new or unknown risk from a safety standpoint. gens (14). Small polypeptides under 10 kDa usually
In fact, these types of fragments have been present in need to be conjugated to large immunogenic carrier
biopharmaceutical products for decades. Further, a com- proteins or administered with adjuvants to ensure an
prehensive literature search did not reveal any evidence antibody response (16).
of safety or efficacy issues directly attributable to the The suitability of gelatin as a reference impurity for
presence of inactive fragments in parenteral drugs (12). setting acceptance limits for inactive HTP fragments is
evaluated in the next section.
Implications for Multiproduct Cleaning Validation
Consider the introduction of a new product (Product B) GELATIN AS A REFERENCE IMPURITY FOR
into the facility. Part of the equipment train is now used INACTIVE HTP FRAGMENTS
to process both products. The cleaning and sterilization The use of gelatin as a reference impurity for inactive
cycles between batches of different products (A B or B HTP fragments is justified for the following reasons:
A, [i.e., intercampaign processing or changeover]) are Gelatin consists of a mixture of animal protein frag-
the same as those between batches of the same product ments derived from the hydrolysis of collagen, a pro-
(A A or B B, [i.e.,intracampaign processing]). Thus, tein that is commonly found in connective tissues
for a given set of cleaning and sterilization cycles, the (17). The collagen is hydrolyzed by exposing the
molecular weight distribution (MWD) of inactive frag- connective tissues to pH and temperature extremes
ments of Product A (IFA) that are carried over into a sub- (18). HTPs in the process residue are exposed to
sequent batch of Product B (intercampaign processing) is similar operating conditions during cleaning and
the same as the MWD of IFA that are carried over into a sterilization. Thus, in terms of chemical composi-
subsequent batch of Product A (intracampaign process- tion, the protein fragments in gelatin are comparable
ing). The same is true for the MWD of inactive frag- to the HTP fragments in the process residue.

Special edition: Cleaning Validation 61


Rizwan Sharnez

To elicit an immune response, a molecule must with MWs less than 10 kDa are generally weak
be recognized as nonself by the immune system immunogens (14). Protein fragments in gelatin
(14). The protein fragments in gelatin are of ani- range from 15 kDa to 400 kDa (18), which is
mal origin whereas the HTP fragments in the substantially higher than the 10 kDa threshold
process residue are of human origin. Thus, the for weak immunogens.
peptide sequences in the HTP fragments are more
likely to be recognized by human immune sys- CQ APPROACH BASED ON GELATIN
tems than the peptide sequences in the protein This section describes the application of the CQ ap-
fragments in gelatin. Consequently, as compared proach based on the use of gelatin as a reference impu-
to the protein fragments in gelatin, the HTP frag- rity to inactive HTP fragments.
ments in the process residue are less likely to Gelatin is used as a stabilizer in many parenteral
elicit an immunogenic response in humans. products. The amount of gelatin in common parenteral
The molecular weights of most of the HTP frag- products ranges from several hundred micrograms to
ments are less than 10 kDa (19). HTP fragments over 15,000 g per dose (refer to table).

Table: Gelatin Content of Common Parenteral Products.

Product Trade Name Gelatin Content Route of Dosing Schedule


per dose Administration
DTaP (Sanofi Pasteur)1, 2 Tripedia 28 g in 0.5 mL Intramuscular Five dose series at two, four,
injection six, 15 months, and four to six
years of age
Influenza (Sanofi Pasteur)2 Fluzone 250 g in 0.5 mL Intramuscular Yearly
injection
Japanese Encephalitis JE-VAX 500 g in 1.0 mL Subcutaneous Three doses on days zero,
(Sanofi Pasteur)2 injection seven, and 30. Booster dose of
1 mL can be given after two
years
Leuprolide acetate for Lupron Depot, 650 g in 3.75 mg Intramuscular Monthly
depot suspension (Abbot)3 3.75 mg

Measles, Mumps, Rubella ATTENUVAX 14,500 g in 0.5 mL Subcutaneous Two doses: one at 12 months of
(Merck)2 injection age and one at four years of age
Measles, Mumps, Rubella, ProQuad 11,000 g in 0.5 mL Subcutaneous Two doses: one at 12 months of
Varicella (Merck)2 injection age and one at four years of age
Rabies (Novartis)2 RabAvert 12,000 g in 1.0 mL Intramuscular Post-exposure dosage: 1 mL
injection doses on days zero, three,
seven, 14, and 28
Varicella (Oka/Merck)2 VARIVAX 12,500 g in 0.5 mL Subcutaneous Two doses each given four
(frozen) injection weeks apart
Yellow Fever (Sanofi YF-VAX 7,500 g in 0.5 mL Subcutaneous One dose every 10 years
Pasteur)1, 2 injection
Zoster (Oka/Merck)2 ZOSTAVAX 15,580 g in Subcutaneous Single dose
0.65 mL injection
Isoplex Solution for ISOPLEX 4% w/v (20g in Intravenous
Given for blood loss, 500 mL
Infusion4 500 mL bag) can be given in as little time as
five minutes in the case of rapid
blood loss
1
Kelso, John M., Li, James T., Adverse reactions to vaccines; Ann Allergy Asthma Immunol., 2009 Oct;103(4 Suppl
2):S1-14.
2
Package inserts via www.fda.gov/BiologicsBloodVaccines/Vaccines/default.htm
3
Package insert via http://www.rxabbvie.com
4
Package insert via http://www.mhra.gov.uk/Safetyinformation/Medicinesinformation/SPCandPILs/index.htm

62 Special edition: Cleaning Validation


Rizwan Sharnez

Lupron serves as a good model for the CQ approach SC SSA /


IFA
[6b]
because it is administered in multiple doses over an
extended period. Note that from the standpoint of Rearranging Equation 6b gives
immunogenicity, repeated dosing of an immunogen
is important; however, dosing frequency is generally SC 0.65 mg (
IFA
/ )/ SSA [6c]
not critical provided that the time interval between
exposures is relatively short (e.g., weekly, bi-weekly, The above inequality can be used to set an ac-
or monthly). Each dose of Lupron contains 0.65 mg of ceptance limit forSC in terms of three measureable
IFA

gelatin; thus, the CQ Criterion based on the gelatin in parameters:


this product is , , and SSA.

0.65 mg[1a] Solved Example


Calculate the acceptance limit for TOC in the swab ex-
Where re
is the mass of inactive frag- tractant for the following scenario: = 20 L, =
ments of Product A (IFA) that is carried over into the 10 mL, SSA = 105 cm2, swab recovery factor for the pro-
largest dose (LD) of the subsequently manufactured cess residue (SRF) = 0.5, carbon fraction in the process
Product (B). residue (CF) = 0.5, area swabbed (A) = 25 cm2, and mass
The concentration of the inactive fragments of Prod- of extractant (m) = 25 g.
uct A (CIFA) in the largest dose of Product B ( ) is, Substituting for , ,and SSA into Equation
6c gives:
CIFA=
[2a]

SC 650 (g) 20,000 (mL) / 10 mL / 105 cm2 =
IFA

13 g/cm2
Substituting for from Equation 1a gives Based on the worst-case assumption that all the
the acceptance limit (AL) for CIFA, carbon in the process residue is from the IFA, the ac-
ceptance limit for TOC (ALTOC) in the swab extractant
[2b]
is
ALTOC = SC A SRF CF / m
IFA

A worst-case estimate of CIFA ( ) is obtained by ALTOC = 13 g/cm2 25 cm2 0.5 0.5 / 25 g


assuming that all of the residual IFA on the surface = 3.25 ppm
of the shared equipment train after cleaning (RIFA) is Thus, the cleaning is effective if the TOC in the
transferred into the smallest batch of Product B ( swab extractant is 3.25 ppm above the baseline TOC
) that is manufactured in the equipment trainii. Thus, of the control sample.
Note that the above acceptance limit is based on
=R / [3] relatively unfavorable system parameters (small batch
size, large dose and shared surface area, and low
Also, recovery). Further, the acceptance limit is considerably
higher than the process capability limit (PCL) of most
RIFA = SSA SCIFA [4] cleaning processes, which is typically on the order of 1
ppm Carbon for TOC swab samples. It is also substan-
Where SSA is the shared surface area of the equip- tially higher than the LOQ of TOC, which is typically
ment train, andSC is the average surface concentra-
IFA
between 0.05 and 0.2 ppm Carbon. It is therefore un-
tion of IFA on the shared surface area. Thus, likely that the acceptance limit based on this approach
would be below the PCL of the cleaning process or the
=SC SSA/
IFA
[5] LOQ of TOC.

Note that any splits of the batch into smaller batches METHODOLOGY FOR SETTING ACCEPTANCE LIMITS
for filling or other operations should be appropriately The proposed methodology for setting acceptance
accounted for in estimating SSA and . limits for multiproduct equipment is summarized in
Based on the definitions of andC ALIFA, the the flowchart. Inactivation studies are first performed
cleaning is effective if under simulated worst-case operating conditions (7). If
the sample is shown to have no detectable activity, the
[6a] CQ approach can be used to set acceptance limits for the
inactivated product (right side of flowchart). However,
Substituting for and from Equation 2b if there is no detectable loss in activity, the conventional
and Equation 5, respectively, gives MAC approach can be used to set the acceptance limit

Special edition: Cleaning Validation 63


Rizwan Sharnez

Figure 1: Flowchart for Setting Acceptance Limits (AL) for the Process Residue.
a
Based on results of bioassay
for the previously manufactured product (1-2). If the cal method. This approach is consistent with industry
results indicate that the API is partially inactivated, the practices and is justified because the cleaning cycle is
acceptance limit should be determined for the API as validated and continually monitored, and has appro-
well as the inactivated product, and the lower of the priate controls in place. Additionally, adopting this ap-
two limits should be used. Alternatively, the operating proach does not impact the relative amount and MWD
parameters can be modified to ensure inactivation of the of the target impurity that is carried over between
API. This can be facilitated by running additional studies batches of different products, which is consistent with
to characterize the effect of the operating parameters on the CQ criterion. Thus, setting the acceptance limit to
the API. the PCL ensures that the MWD of the target impurity
that is carried over into a batch of Product B is com-
Default Limits parable to the MWD of that target impurity that was
If the carryover of the target impurity into the subse- historically present in a batch of Product A.
quent batch based on the above acceptance limit (AL) is
above 10 ppm, then the AL should be reduced to limit CONCLUSION
the carryover to 10 ppm. The 10 ppm carryover limit is a Biopharmaceutical cleaning and sterilization cycles are
widely used default limit (20). It is based on the empiri- designed to expose product contact equipment to pH
cal observation that raw materials and intermediates and temperature extremes for several minutes. Under
commonly found in biopharmaceutical manufacturing these conditions biological APIs degrade and denature
process residues are safe to ingest at concentrations up to rapidly thereby becoming pharmacologically inactive.
0.1% (1000 ppm). In order to extrapolate this oral limit Inactivation of the product during cleaning and
to other routes of administration, it is reduced by a factor sterilization has important implications for cleaning
of 100 to 10 ppm. validation of multiproduct equipment. If the API is in-
If sufficient historical cleaning data are available to activated, the acceptance limits for the process residue
establish a statistically sound process capability limit can be set based on the inactive product instead of the
(PCL), then the AL should be set to the PCL. If the API.
historical data are below the LOQ of the analytical A comprehensive literature search did not reveal any
method, the PCL can be set to the LOQ of the analyti- evidence of safety or efficacy issues directly attribut-

64 Special edition: Cleaning Validation


Rizwan Sharnez

able to the presence of inactive fragments in parenteral 0.05 and 0.2 ppm Carbon. It also compares favorably
drugs (12). Further, these types of fragments have been to the process capability limit (PCL) of most cleaning
present in biopharmaceutical products for decades. processes, which is typically on the order of 1 ppm
Thus, as a class of molecules, inactive fragments of Carbon for TOC swab samples. Thus, with the CQ ap-
HTPs do not present a new or unknown risk from a proach based on gelatin, it is unlikely that the accep-
safety standpoint. tance limit for the process residue would be below the
The Comparable Quality (CQ) approach based on PCL of the cleaning process or the LOQ of TOC.
gelatin can be used to set acceptance limits for inactive The methodology described in this paper is not
fragments of non-conjugated HTPs. With this ap- applicable to allergenic ingredients, penicillin, cepha-
proach, the carryover of inactive fragments into the losporin, potent steroids, and cytotoxic compounds.
largest dose of the subsequently manufactured product Acceptance limits for process residues associated with
is limited to the acceptable exposure of an appropri- these types of APIs are typically set to the limit of
ate reference impurity. The reference impurityin this detection (LOD) of the best available analytical method
case gelatinwas shown to be comparable to or worse (21).
than the inactive fragments from a predictive safety
standpoint. i
Depending on the process soil, API refers to the
If the product is not inactivated during cleaning active pharmaceutical ingredient in the drug product,
and sterilization, the acceptance limit for the process drug substance, or drug substance intermediate.
residue should be set based on the acceptable carry- ii
i.e., surface area of equipment train that is subjected
over of the API (1-2). However, if the results indicate to cleaning and that comes into contact with both
that the API is partially inactivated, the acceptance Product A and Product B. If inactive fragments of
limits should be determined for the API as well as for Product A are removed by the purification steps
the inactive fragments, and the lower of the two limits for Product B, then the part of the equipment train
should be used for cleaning validation (refer to Figure). upstream of those purification steps can be excluded
The acceptance limit for inactive fragments based from the shared surface area.
on gelatin as a reference impurity was ascertained to iii
Note that the use of a non-specific method such as
be 0.65 mg per dose. At 0.65 mg of inactive fragments TOC also allows for the detection of intact protein.
per dose, the acceptance limit for TOC swab samples
was shown to be 3.25 ppm Carbon.iii This estimate
was based on relatively unfavorable system parameters. ACKNOWLEDGEMENTS
Note that this acceptance limit is substantially higher We thank Vijay Chiruvolu, Sam Guhan, Aine Hanly and
than the LOQ of TOC, which is typically between Anthony Mire-Sluis for their help and support.

Special edition: Cleaning Validation 65


Rizwan Sharnez

ACRONYMS
A Area swabbed
AA Between batches of the same product (intracampaign)
AB Between batches of different products (intercampaign)
ADE Acceptable Daily Exposure
AL Acceptance limit
ALCQ Acceptance limit based on comparable quality approach
ALMAC Acceptance limit based on MAC
ALTOC Acceptance limit for TOC swab sample
API Active pharmaceutical ingredient
Acceptable limit for concentration of inactivated fragments of Product A
CIFA Concentration of inactivated fragments of Product A
Worst-case estimate of CIFA
CF Carbon fraction in process residue
CQ Comparable quality
HTP Human therapeutic proteins
IFA Inactive fragments of Product A
IFB Inactive fragments of Product B
kDa Kilo Dalton
LD Largest dose
LOD Limit of detection
LOQ Limit of quantitation
m Mass of extractant
MAC Maximum allowable carryover
Mass of inactive fragments of Product A in largest dose of Product B
MW Molecular weight
MWD Molecular weight distribution
PCL Process capability limit
PR Process residue
Product A Previously manufactured product
Product B Subsequently manufactured product
RIFA Residual IFA on the surface of the shared equipment train after cleaning
SC IFA
Average surface concentration of IFA on the shared surface area
SDS PAGE Sodium dodecyl sulphate polyacrylamide gel electrophoresis
SRF Swab recovery factor
Shared surface area (i.e., surface area of the equipment train that comes into contact with
SSA
both Product A and Product B)
TOC Total organic carbon
Largest dose of Product B

Minimum volume of the final batch of Product B (i.e., smallest batch size of Product B)

66 Special edition: Cleaning Validation


Rizwan Sharnez and Angela To

Multiproduct Cleaning
Validation: Acceptance
Limits for the Carryover
of Inactivated API
Part IThe Comparable
Quality Approach
Rizwan Sharnez and Angela To

New Perspectives on Cleaning is an ongoing series of articles dedicated


to cleaning process development, validation, and monitoring. This column
addresses scientific principles, strategies, and approaches associated with
cleaning that are faced in everyday work situations.
Reader questions, comments, and suggestions are requested for future
discussion topics. These can be submitted to the column coordinator Rizwan
Sharnez at rsharnez@amgen.com.

SUMMARY
An important consideration in multiproduct cleaning validation is to dem-
onstrate that the carryover of the previously manufactured active pharma-
ceutical ingredient (API) into a batch of the subsequently manufactured
product is below an acceptable limit. If, however, the previously manufac-
tured API becomes therapeutically inactive during cleaning, then there is
limited value in verifying clearance of the API. Instead, it is more appropri-
ate to demonstrate clearance of inactivated product. This approach is gaining
acceptance in the industry.
A methodology for evaluating the degree of inactivation of a product
during cleaning and setting acceptance limits for the carryover of inacti-
vated product in multiproduct equipment is described. A new approach
for justifying acceptance limits for inactivated product, known as the
comparable quality (CQ) approach, is described in Part I; the application
of this approach to biopharmaceutical cleaning will be described in Part
II. The general principles of the CQ approach are applicable to most ac-
tive pharmaceutical ingredients (APIs).

INTRODUCTION
For multiproduct cleaning validation, acceptable carryover of the previously
manufactured API (Product A) into the subsequently manufactured API
(Product B) is determined through a maximum allowable carryover (MAC)
calculation (1-3). A limitation of the conventional MAC approach is that
it does not account for the carryover of the inactivated molecule between
lots of different products (i.e., A B, or B A). This is an important factor
to consider when aggressive cleaning conditions are used. For example,
biopharmaceutical cleaning cycles are generally designed to expose prod-
uct contact equipment to extremes of pH (i.e., 2-13) and temperature (i.e.,
60-80C) for several minutes. The equipment may also be steam sterilized
or sanitized after cleaning. Under these conditions, monoclonal antibod-

Special edition: Cleaning Validation 67


Rizwan Sharnez and Angela To

ies, therapeutic proteins, and other biological APIs are Similarly, it is possible for the API to be inactivated even
known to degrade and denature rapidly (4, 5) and are, though the epitopes are not completely destroyed, and
therefore, likely to become therapeutically inactive (6). this can lead to false positives. Another issue with PSAs
Inactivation of the product during cleaning has is that it is difficult to get an accurate recovery factor.
important implications for cleaning validation of mul- That is because the experimental conditions of the
tiproduct equipment. If it can be demonstrated that recovery study (e.g., direct spotting of coupons with the
the product becomes therapeutically inactive during API) do not represent the actual sample matrix (i.e., a
cleaning, a MAC assessment for the API would not be small amount of API in the presence of degradants and
required. It also obviates the need to develop product cleaning agent residues). For the above reasons, PSAs
specific assays (PSA) for cleaning validation. should be used judiciously for verifying clearance of
Inactivation of the product during cleaning can be biological APIs after cleaning (7).
assessed by exposing the process soil to simulated Another issue with the MAC approach is that every
cleaning conditions at bench scale (4, 5). The bench time a new product is introduced into a facility there is
scale studies are designed to simulate the conditions a risk that one or more of the new MAC limits for the
that are least conducive (worst-case) for inactivation. For previously validated products could be below the exist-
example, for alkaline washes, the lowest applicable pH, ing acceptance limits for cleaning validation.
temperature, duration, and ratio of cleaning solution
to process soil is used to simulate the cleaning cycle at PROTEIN DEgRADATION
bench scale. The sample is then neutralized and cooled AND INACTIvATION APPROACH
to minimize any further inactivation. The degree of in- Performing Degradation and Inactivation Studies
activation is evaluated by subjecting the sample and an A bench-scale approach for evaluating the bioactivity of
untreated control to the appropriate assays (e.g., Sodium the residual API and the molecular weight distribution of
Dodecyl Sulfate Polyacrylamide Gel Electrophoresis the degradants after exposure to cleaning conditions is
[SDS PAGE] and bioassay for biological products). described in this section.
A literature search did not uncover any scientific Full-scale cleaning conditions of shared product
approaches or regulatory guidances for setting accept- contact equipment are evaluated to determine the con-
able limits for the carryover of inactivated product for ditions that are least conducive for inactivation (e.g.,
cleaning validation. lowest ratio of wash volume to protein, lowest cleaning
agent concentration, lowest temperature, and shortest
LIMITATIONS OF THE MAC APPROACH duration of exposure). The product is exposed to these
The MAC approach is often used to set cleaning valida- conditions at bench scale. The objective of the study is
tion acceptance criteria for the carryover of the previous- to ascertain whether the API in the process sample is
ly manufactured API (Product A) into the subsequently inactivated when exposed to cleaning conditions.
manufactured API (Product B) (1-3). A limitation of this Product is spiked into tubes containing alkaline
approach is that it does not account for the carryover of cleaning solution, heated to the appropriate tempera-
the inactivated molecule between lots of different prod- ture, and allowed to incubate for the duration of the
ucts (i.e., A B, or B A). alkaline wash. Samples may also be titrated with the
Another limitation of the MAC approach is that the acidic cleaning solution to the pH of the acidic wash
acceptance limits for cleaning validation are often below and held for the duration of the acidic wash. Samples
process capability limits and/or below the limit of quan- are then titrated to a neutral pH and cooled to 4C to
titation (LOQ) of non-specific assays (e.g., total organic minimize further degradation and inactivation. The
carbon [TOC]; the LOQ of TOC is typically between samples are then subjected to SDS-PAGE and bioas-
0.05 and 0.2 ppm). This issue is further exacerbated say to determine the degree of degradation and TOC
by the low recovery of APIs from process soils. PSAs analysis to determine whether the degradants can be
such as enzyme-linked immunosorbent assay (ELISA) adequately recovered by TOC.
and enzyme immunoassay (EIA) are sometimes used
to address this issue because they have very low LOQs Assays
(typically below 10 ppb). However, PSAs are difficult SDS-PAGE and bioassays are used to evaluate protein
and laborious to qualify, and can give inaccurate results degradation and inactivation, respectively. The product
if the API degrades during cleaning (7). That is because is exposed to cleaning conditions at small scale and then
PSAs detect activity indirectly, by recognizing specific analyzed with the above assays to determine degree of
epitopes (i.e., short amino acid sequences that PSAs are degradation (i.e., molecular weight distribution) and
designed to detect). The epitopes can be destroyed by bioactivity. Additionally, samples are analyzed for TOC
buffer and cleaning agent components. However, a bio- to determine the recovery of the inactivated protein and
logical API can be therapeutically active even if the epi- the applicability of the TOC assay for demonstrating
topes are destroyed, and this can lead to false negatives. clearance of the inactivated product at full scale.

68 Special edition: Cleaning Validation


Rizwan Sharnez and Angela To

SDS-PAGE solubilizes aggregated and degraded product, the MAC approach is used to limit carryover of
proteins and separates them based on molecular previous product to an acceptable level (1-3). If the MAC
weight (MW). The inclusion of MW standards allows limit is higher than the LOQ of TOC, TOC is used to
for estimation of the MWs of the degradants and any verify clearance of previous product at full scale. If the
aggregates, and the inclusion of control samples at a MAC limit is below the LOQ of TOC, an alternate assay
defined protein load allows for the estimated quantita- may need to be developed to verify clearance of the API.
tion of protein concentration by densitometry. Staining The MAC approach limits the amount of API of the
of gels is sensitive to 5-10 ng for Silver Staining and previously manufactured product (A) in a dose of the
100 ng for Coomassie Staining. SDS-PAGE has the subsequently manufactured product (B) to the accept-
advantage of providing a wide range of specificity for able daily exposure (ADE) of A (8), or to 1/1000th of
detecting proteins with unknown primary structures, the minimum dose of A (9).
size, charge, and hydrophobic states. This feature is
particularly useful for protein degradation analysis COMPARABLE QUALITY APPROACH
because the level of protein degradation due to clean- If the protein becomes therapeutically inactive during
ing is highly unpredictable and can extend over a wide cleaning, then the acceptance limit for the inactivated
range of MW. SDS-PAGE also provides high sensitivity molecule of the previously manufactured product
for detecting trace amounts of protein. (Product A) in the subsequently manufactured product
Bioassays measure the relative amount of biological- (Product B) can be set based on the CQ approach. With
ly active product present in a sample. Thus, bioassays the CQ approach, the amount of inactivated Product A in
can be used to determine the effect of cleaning condi- a dose of Product B is limited to the amount of inacti-
tions on the inactivation of biologicals. vated API of Product A in a dose of Product A. Appropri-
ate adjustments are made to account for differences in
SETTINg ACCEPTANCE LIMITS process parameters of the two products. If inactivated
BASED ON PROTEIN INACTIvATION Product A can be recovered and detected by TOC, and
The methodology for setting acceptance limits is summa- its acceptance limit is greater than the LOQ of TOC, then
rized in the Figure. Degradation and inactivation studies TOC is used to verify clearance of the inactivated API.
are first performed under simulated cleaning conditions. Otherwise, an alternative assay is developed to demon-
If the sample is shown to have therapeutically active strate clearance of the inactivated API at full scale.

Figure: Methodology for setting acceptance limits flowchart. Continued on page 70

Inputs Perform
Process samples and inactivation
reference standard study
Cleaning cycle and
equipment parameters

Is API in process
sample inactivated after
1 Based on exposure to cleaning
results of conditions?1
bioassay
Yes No

Use CQ approach to Use MAC


set acceptance limit approach to
(AL) for inactivated set acceptance
API limit (AL) for API
continued continued

Special edition: Cleaning Validation 69


Rizwan Sharnez and Angela To

Continued from page 69

Inputs
Dose and batch sizes of
Use MAC
products
Surface area and rinse
approach to
parameters of shared set acceptance
equipment limit (AL) for API
Recovery study for API
Carbon content of API

Is AL LOQ
of TOC?
Yes No

Develop alternative
Use TOC to
assay to
demonstrate demonstrate
clearance of API
clearance of API

Inputs
Dose and batch sizes of products Use CQ approach to set
Surface area of dedicated and
acceptance limit (AL) for
shared equipment
Recovery study for inactivated API inactivated API
Carbon content of inactivated API

Can
inactivated API
be recovered
by TOC?
Yes No

2 Thisis
No done on a
Is AL LOQ
case-by-
of TOC?
case basis
Yes
Use TOC to
Develop alternative assay
demonstrate
to demonstrate clearance
clearance of
of inactivated API2
inactivated API

70 Special Edition: Cleaning Validation


Rizwan Sharnez and Angela To

CONCLUSION ACKNOWLEDgEMENTS
The inactivation of a product during cleaning and steam- The authors are grateful to Arun Tholudur and Joel
ing has important implications for cleaning validation Bercu for their helpful suggestions and support.
of multiproduct equipment. Demonstrating that the
product becomes inactivated during these operations ob- ARTICLE ACRONYM LISTINg
viates the need to perform arduos MAC assessments for A Product Apreviously manufactured product
the API. It also eliminates the need to develop PSAs for ADE Acceptable Daily Exposure
cleaning validation. PSAs are designed to detect specific AL Acceptance Limit
epitopes; thus, if the API degrades, the result from the as- API Active Pharmaceutical Ingredient
say may not necessarily correlate to therapeutic activity. B Product Bsubsequently manufactured
The CQ approach can be used to set acceptance lim- product
its for the carryover of inactivated product for multi- CQ Comparable Quality
product cleaning validation. This approach is designed EIA Enzyme Immunoassay
to ensure that the amount of inactivated Product A in ELISA Enzyme-Linked Immunosorbent Assay
a dose of Product B is less than the amount of inacti- LOQ Limit of Quantitation
vated Product A in a dose of Product A. Application of MAC Maximum Allowable Carryover
the CQ approach to biopharmaceutical cleaning will MW Molecular Weight
be described in Part II of this series. PSA Product Specific Assay
SDS PAgE Sodium Dodecyl Sulfate Polyacrylamide Gel
REFERENCES Electrophoresis
1. Sharnez, R., Strategies for Setting Rational MAC- TOC Total Organic Carbon
based LimitsPart I: Reassessing the Carryover
Criterion, Journal of Validation Technology, Vol 16,
No. 1, p. 71-74, 2010.
2. Sharnez, R., To, A., Klewer, L., Strategies for
Setting Rational MAC-based LimitsPart II: Ap-
plication to Rinse Samples, Journal of Validation
Technology, Vol 17, No. 2, p. 43-46, 2011.
3. Sharnez, R., To, A., Strategies for Setting Rational
MAC-based LimitsPart III: Leveraging Toxicol-
ogy and Cleanability Data, Journal of Validation
Technology, Vol 17, No. 3, p. 24-28, 2011.
4. Kendrick, K., Canhoto, A., Kreuze, M., Analysis
of Degradation Properties of Biopharmaceutical
Active Ingredients as Caused by Various Process
Cleaning Agents and Temperature, Journal of Vali-
dation Technology, Vol 15, No. 3, p. 69, 2009.
5. Rathore, N., Qi, W., Chen, C., Ji, W., Bench-scale
characterization of cleaning process design space
for biopharmaceuticals, Biopharm Int, Vol 22,
No. 3, 2009.
6. Martinez, J.E., Immunogenic Potential of Thera-
peutic Protein Residues after Cleaning, Bioprocess
International, Vol. 9, P. 38-44, 2004.
7. Health Canada, Cleaning Validation Guidelines
(GUIDE-0028); Section 8.3, Health Products and
Food Branch Inspectorate, 2008. http://www.hc-sc.
gc.ca/dhp-mps/compli-conform/gmp-bpf/valida-
tion/index-eng.php
8. ISPE, Risk-Based Manufacture of Pharmaceutical
Products: A Guide to Managing Risks Associated with
Cross-Contamination, 1st Edition, Vol. 7, ISPE, 2010.
9. Fourman, Gary L. and Michael V. Mullen, Deter-
mining Cleaning Validation Acceptance Limits for
Pharmaceutical Manufacturing Operations, Phar-
maceutical Technology, 17 (4), 54-60, 1993. JVT

Special edition: Cleaning Validation 71


Sebastian Clerkin

Validation of a Cleaning
Process for Medical
Devices | IVT
Sebastian Clerkin, Ph.D.

AbStrACt
Many medical device manufacturers find it a considerable challenge
to plan and conduct a cleaning validation. The main challenges are
an establishment of the cleanliness limits and an identification of the
challenge conditions to be assessed during the process validation. This
paper describes a logical risk-based approach to overcome these chal-
lenges.
It begins with assessing the complete manufacturing process and
identifying the manufacturing agents. It discusses risk tools to deter-
mine which manufacturing agents need to have cleanliness limits. It
describes the manufacturing conditions to be considered when con-
ducting the cleaning validation.
The concepts described within this paper can be utilized by a medi-
cal device manufacturer to establish a cleaning process that will con-
sistently provide clean medical devices and comply with the relevant
regulations.

INtrODUCtION
Contamination of a medical device can have serious implications. Medi-
cal device manufacturers must ensure they have correctly identified all
potential contaminants and have established controls.
The United States Food and Drug Administration captures this re-
quirement within the Quality System Regulations (QSR) by stating that
each manufacturer shall (1, 2):
Establish and maintain procedures to prevent contamination of product by
substances that could be expected to have an adverse effect on product quality
Establish and maintain procedures for the use and removal of manufactur-
ing materials to ensure that it is removed or limited to an amount that does
not adversely affect the devices quality (2).
International Organization for Standardization (ISO) 13485 requires
that a medical device manufacturer establish documented requirements
for the cleanliness of a medical device in the following circumstances
(3):

Product is cleaned by the organization prior to sterilization and/or


its use, or
Product is supplied non-sterile to be subjected to a cleaning process
prior to sterilization and/or its use, or
Product is supplied to be used non-sterile and its cleanliness is of
significance in use, or
Process agents are to be removed from product during manufac-
ture.

Therefore, to comply with the QSR and ISO 13485, a medical device
manufacturer must establish documented cleanliness requirements.

72 Special edition: Cleaning Validation


Sebastian Clerkin

Figure 1: Example of a Manufacturing Process Flow.

However, these regulations do not explicitly state An organic residual; these are mostly insoluble in
that a cleaning process validation must be completed. water and include greases and oils
The Global Harmonization Task Force (GHTF) Study An inorganic residual; these are water soluble,
Group 3 does provide guidance on the requirement and examples include metal ions
for a cleaning process validation. Their process vali- Particulate; an example would be metallic par-
dation guidance (4); which was written by regula- ticles left over from a cutting process.
tors in the US, Europe, Japan, Australia and Canada;
states a cleaning process may be validated or may Hazardous components of manufacturing materials
be satisfactorily covered by verification. ISO 14696, can be obtained from the Material Safety Data Sheet
which provides guidance on the application of ISO (MSDS). The MSDS should be available for all the
13485, states that a cleaning process needs consid- manufacturing materials used in the process.
eration of use and the controls in place to determine It must be noted, however, that the MSDS gener-
whether some or all of the elements of validation are ally only lists the main components that are pres-
required (5). Therefore, a decision to conduct a clean- ent in a mixture. For example, the United States
ing process validation is dependent on the outputs Department of Occupational Safety and Health
of the cleaning process and the controls in place. For Administration (OSHA) only requires that hazard-
example, Figure 1 shows a manufacturing process ous constituents in excess of 1% be disclosed on a
flow that has an intermediate cleaning step. The sole MSDS (6). For carcinogens it is 0.1%. This is due to
output of this intermediate clean step may be that the the fact that MSDS are designed to protect the work-
parts are visibly clean. In this instance, verification ers, not identify potential hazardous contaminants
may be sufficient with no requirement for process on a medical device.
validation. On the other hand, the manufacturer There is also a risk that multiple constituents
taking a risk-based approach has specified a level of below the 1% disclosure threshold could have a cu-
acceptable residues after intermediate cleaning. In mulative effect on the intended use of the device.
this instance, verification would not be sufficient and Even though the MSDS does have these shortcom-
process validation would be necessary. ings, it is still a very useful initial tool in identifying
In the above manufacturing process flow (Figure potentially hazardous manufacturing agents.
1), the final cleaning step is the more critical cleaning When dealing with complex manufacturing
step of the two, as this is the last step in the process materials such as cutting fluids, it is a considerable
to make sure that the medical devices are sufficiently challenge to identify the individual components
clean prior to packaging. The cleaning validation ap- as they contain many different chemicals. These
proach described within this paper is more applicable chemicals may not be identified by the supplier or
to this final clean step. It consists of a number of may be considered proprietary. In these instances,
logical steps from identifying the risks to establishing it is imperative to liaise directly with the manufac-
limits and from validation to process monitoring. turing material supplier to identify any potential
contaminants that could impact the intended use of
revIew PrOCeSS FlOw AND IDeNtIFy the MANU- the device.
FACtUrINg MAterIAlS In addition, any process steps completed by an ex-
In order to determine the cleanliness limits for manu- ternal supplier must also be evaluated. For example,
facturing agents after the final cleaning process, it is manufacturing material may be present on a supplied
imperative to first evaluate the complete manufactur- component. These must be considered. This is why it
ing process. An overall process flowchart should be is crucial that the medical device manufacturer has a
created to demonstrate that the process has been ade- written agreement in place with their supplier that no
quately assessed, and it should contain information on changes are made to their manufacturing materials
the manufacturing materials that come in contact with without prior agreement.
the product at each process step. Examples of manu- Only after the overall process flow has been com-
facturing materials that must be considered include: pletely reviewed and all the potential contaminants
lubricants, detergents, wipes used during inspection, on the device identified can the manufacturer start
and polishing agents. This manufacturing material to consider their impact. Their impact is assessed by
maybe attributable to one of the following groups: conducting a risk analysis.

Special edition: Cleaning Validation 73


Sebastian Clerkin

Process Manufacturing Potential Potential Potential S L R Mitigation S L R


Step Material Hazards Effect Cause

Passivation Passivation Sodium Car- Residual 5 2 10 Use a passiv- 5 1 5


solution dichromate cinogen; Sodium ation solution
(CAS#: Irritant; dichromate that contains
7789-12-0) Mutagen present on no Sodium
the finished dichromate
device

Grinding Grinding wheel Metallic Third Metallic 4 2 8 Establish a 4 1 4


particulates body wear particulates particulate
of bearing present on specification
surface finished for finished
device device
table 1: Hazard Analysis. Severity (S) is scored1 to 5, 5 being the most severe; Likelihood is scored 1 to 5, 5 being the most likely; Risk index (R) is cal-
culated by multiplying the Severity score by the Likelihood score.

risk Analysis and Identification of Materials calculate limits for the pre-identified manufacturing
of Concern materials.
For the risk analysis, the impact of the contaminants However, in many instances, the NOAEL is not
from a hazardous perspective and from an intended known, and so the manufacturer must rely on using the
functionality perspective must be considered. LD50 values. LD50 is the median lethal dose. In other
A useful tool in identifying which contaminants words, the amount of a particular toxin that will kill 50%
are of the most concern is to use a hazard analysis of the population over specified time duration. These
and to ask the following questions: LD50 values can be readily obtained from the MSDS.
The LD50 values are then used to calculate the Accept-
Will too much of this contaminant be harmful to able Daily intake (ADI) using the following equation:
the patient?
Will too much of this contaminant impact the ADI = LD50 x mB/CF
proper functioning of the device?
Where:
An example of a hazard analysis is shown in Table
I; the process steps, manufacturing material and par- LD50 = median lethal dose
ticular agents are listed, and then the risk is consid- mB = is the body mass of the patient population and is
ered. The mitigation from the hazard analysis can be generally defaulted to 70kg
used to establish a particular cleaning limit or use of CF= conversion factor
an alternative manufacturing material.
The conversion Factor (CF) is typically a factor be-
eStAblIShINg CleANINg lIMItS tween 100 and a 1000 and is derived to incorporate
Now that the potential hazardous contaminants have uncertainty factors (UF) such as:
been identified, the acceptable level of contamination
on the medical device must be determined. These Extrapolation from animal to human tolerances
levels or limits must be documented and scientifically (typically defaulted to a factor of 10)
justified by the medical device manufacturer. Inter-human variability (typically defaulted to a
For toxic contaminants where there is known factor of 10)
toxicity data, ISO 10993-17 is very useful. It de- Additional UFs can be based on the type of
scribes a method to determine the acceptable levels medical device (i.e., medical device class) and the
of leachable material from a medical device using the duration of exposure.
No Observed Adverse Effect Level (NOAEL) (7). The
NOAEL is the highest concentration of a material that The weighting of each UF should be documented
causes no significant adverse effects in the exposed and justified (8). The UFs are then used to calculate
population. The standard takes this value and uses CF:
it to calculate the tolerable intake (TI) for a specific
leachable substance. This approach can be used to CF = UF1 x UF2 x UF3

74 Special edition: Cleaning Validation


Sebastian Clerkin

In most cases, a conversion factor between 100 and contaminants where the LD50 value is known but
1,000 is sufficient; however, there may be instances the manufacturer has identified additional risks.
where significant risks have been identified and a CF as For example, if a carcinogen has been identified as
high as 10,000 may be appropriate (9). Refer to Kramer a potential contaminant, a spiking study may be
et al.for further information on conversion factors (10). instigated with a genotoxicity and/or carcinogenicity
Consider a real world example of the above ap- study as the endpoint.
proach using an alkaline cleaner: Working out the cleanliness limits for non-toxic
contaminants, which have the potential to interfere
The CF has been established as 1,000 to account with the proper functioning of the device, needs to be
for no human toxicity data (factor of 10), inter-human established by reviewing historical data or by spiking
variability (factor of 10), and a short exposure time of experiments. The spiking study is conducted in a
the device (factor of 10). similar manner as above, but instead of a biocompati-
The LD50of the alkaline cleaner is 365 mg/kgrat. bility study being the endpoint, the levels of contami-
The average human body weight is 70 kg. nation are now evaluated against a specific functional
This would give: requirement for the device. For example, for a device
ADI/device = 365 mg/kg x 70 kg/1000 =25.55 mg/ that has a bearing surface and device failure attribut-
device ed to particulate contamination, a spiking study may
Therefore, the cleanliness limit for this chemical be developed to determine the level of particulate that
would be 25.55 mg per device. will induce the failure.
Using this approach, a cleanliness limit can be
calculated for each specific toxin that was identified eStAblIShINg CleANINg teSt MethOD
during the risk analysis. Now that the cleaning limits have been identified
Obviously, this approach only identifies a cleanli- for specific manufacturing agents, the next step is to
ness limit for known toxins. It is not suitable for decide on a method to quantify the levels. There are
calculating the cleanliness limit where there is a lack two types of tests that can be developed:
of toxicological data available or the contaminants
have no associated toxicity but will impact the proper A specific analytical test can be developed to
functioning of the device. quantify a contaminant (refer to Table II for
For potential toxins where there is no readily avail- examples).
able toxicological data, a series of spiking studies can A non-specific test can be developed to quantify
be completed. This is where the device is artificially many different contaminants at the same time
contaminated with known amounts of the potential (refer to Table II for examples).
toxin. Biocompatible studies can then be completed
to determine the point of failure. The suite of ISO- There are pros and cons for both these approaches.
10993 standards provide a wealth of information that With respect to a specific test method, an accurate
can be used to define the biocompatibility studies measurement of a particular residue can be evaluated.
needed in establishing the failure point. Cytotoxic- This can be very important when this residue has
ity, sensitization, systemic toxicity, and genotoxicity been identified as being highly hazardous. However,
studies are examples of biocompatibility studies that these specific methods are more difficult to imple-
could be considered. The established failure point ment and are more expensive; therefore, they are re-
can then be used to derive the cleanliness limit. ally only used when a specific risk has been identified
Another approach to spiking studies is to approach during the risk analysis.
the issue from the opposite end. In other words, in- Non-specific methods are more commonly used in
stead of finding the failure point, the device is spiked validating cleaning lines; they are less expensive and
with a known amount of the contaminant that is easier to develop. However, due to their non-specific
above the level expected to be observed after clean- nature, they do not give an accurate quantification
ing. If this higher level is established as safe for the of any individual contamination, only a total level
patient, it can be defined as the cleanliness limit. of a group of contaminants. Generally, this can be
Spiking studies can also be useful if the risk analysis sufficient where the requirement is to demonstrate a
has identified a potential cumulative effect of various certain level of overall cleanliness.
contaminants. In other words, if each contaminant is When developing any test method, the following
treated independently of each other, a cleanliness limit factors should be considered:
may be established that does not take into account a
potential cumulative effect. In this instance, the patient Detection Limit; for example, the test method
may be exposed to unacceptable risk. must be sensitive enough to detect relevant levels
Spiking studies should also be considered for of the contaminants.

Special edition: Cleaning Validation 75


Sebastian Clerkin

Specific/Non- Test Method Description


Specific
Specific Infrared Spectroscopy A spectroscopic technique. Useful to identify and quantify
specific molecules
Grinding wheel Metallic particulates
Specific Grinding wheel Metallic particulates
Gas Liquid Partition Chro- Useful for quantifying a specific contaminant that can be
matography (GLPC) vaporized without composition
Specific High-Performance Liquid A method that separates a mixture allowing individual
Chromatography (HPLC) components to be quantified
Non-Specific Conductivity Measure of ionic compounds
Non-Specific Total Organic Carbon Quantitative measure of carbon contained within an or-
(TOC) ganic compound. A very good method of showing overall
cleanliness.
Non-Specific Visual Visual Assessment of cleanliness
Non-Specific Gravimetric Analysis Quantitative measure of mass of a solid within a solution
table 2: List of Test Methods. Each could potentially be used to assess outputs of a cleaning process.

Percentage recovery; the amount of contaminants To adequately challenge a cleaning process during OQ,
that can be recovered from the device must be the worst-case product should be used. For medical
determined. device manufacturers that clean multiple different
Reproducibility and Repeatability devices, identification of the worst-case product can
Linearity be a challenge. To aid in this identification, a worst-
Specificity. case product matrix can be used to help determine the
worst-case product. Each product is scored according
Once the test methods have been developed, they to predefined criteria. These predefined criteria would
must be qualified prior to being used in a cleaning have a potential impact on the cleaning ability of the
process validation. The test method validation must process. Examples include the following:
demonstrate that the analytical method and the extrac-
tion and/or sampling method is repeatable. With re- Device geometry; more complex geometry is
spect to the extraction method, it must be demonstrat- potentially more difficult to clean
ed that the contaminant can be consistently recovered Surface area; greater surface area could be a
from the medical device. There is no point in having greater cleaning challenge.
a repeatable analytical method if the contaminant of Up-stream process flow; this could have an
interest cannot be consistently extracted off the device. impact on the cleanliness levels of the parts pre-
cleaning. A greater level of contamination on
Cleaning equipment Qualification (IQ) the parts pre-clean would obviously be a greater
The cleaning equipment must be qualified prior to challenge to the process.
commencing the process validation. This will dem-
onstrate that the equipment is installed correctly and The scores against these criteria can then be tabu-
functions as intended. lated in the form of a matrix and used to calculate a
total risk score. See Table IV for an example.
Operational Qualification (OQ)Challenge Condi- Once one has established the critical process
tions inputs, these should be challenged within the OQ to
As part of the OQ phase, the critical process inputs prove that product can be cleaned that meets the pre-
should be identified. It must be established how these determined cleaning specifications under all antici-
process inputs impact the cleanliness outputs. Brain- pated manufacturing conditions.
storming and tools such as fishbone diagrams can be
very useful in identifying the critical process inputs. PerFOrMANCe QUAlIFICAtION
Some examples of potential process inputs that could PQ means establishing by objective evidence that the pro-
be considered during a brainstorming are listed in cess, under anticipated conditions, consistently produces
Table III. a product that meets all predetermined requirements (11).

76 Special edition: Cleaning Validation


Sebastian Clerkin

Cleaning Process Inputs Comment


Tank Temperature Higher temperatures are generally more effective while lower temperatures are less
effective.
Cleaning Time Longer times are generally more effective while lower times are less effective.
Concentration Usually the higher the concentration of cleaning agent the better; however, at
higher concentrations there may be higher level of cleaning residual present on the
parts post-cleaning.
Cleaning Solution As parts are processed through the cleaning solution, it will become contaminated.
Contamination There is a risk that as the cleaning tank becomes dirtier, its cleaning effectivity will
decrease. To assess this, the change-out frequency of the tank, the level of con-
tamination on the parts, and the number of parts processed between change-outs
must be considered.
Flow Rate of Cleaning Higher flow rates are generally more effective while lower flow rates are less effec-
Agent tive.
Agitation Agitation should improve the cleaning effectiveness.
Ultrasonic Ultrasonic should improve the cleaning effectiveness.
Load A higher number of parts or dirtier parts may represent a greater challenge to the
cleaning process.
Rinse Parameters Rinse times, temperatures and volume are important in removing the cleaning
agent from the product.
Drying Parameters Time, temperature and airflow are critical parameters that can impact the dryness
of the parts. The geometry of some parts, such as the internal diameter of a narrow
tube, may make them difficult to dry. Another consideration is the temperature
used for drying. Too high a temperature could have an impact on the material
properties of the product.
table 3: Cleaning Process Inputs.

Worse Case Product Matrix


Product Number Surface Area Device Geometry Up-Stream Pro- Total Risk Score
cess Flow
1001 10 10 5 500
1002 7 5 10 350
table 4: Worst-Case Product Matrix. The product is scored between 1 and 10 for each criterion. These values are then multiplied together to give the
total risk score. The product with the highest risk score is then considered the worse case product.

To establish this, the medical device manufacturer must PrOCeSS MONItOrINg


demonstrate that the process in anticipated manufactur- After the cleaning process is validated, it is desir-
ing conditions is stable and can consistently produce able to implement process monitoring of key process
product to the predefined cleaning specifications. The parameters and product outputs to maintain a state of
manufacturer can use tools such as control charts to control. The action levels can be established during
demonstrate that the process is stable as well as capability the process validation.
analysis to show that the process is consistent. ISO 14969 specifically states that the process pa-
With respect to identifying the anticipated con- rameters used for cleaning should be routinely moni-
ditions of a cleaning process, factors that could be tored in accordance with documented procedures
considered are the following: (12). Examples of critical process inputs that could be
monitored would be conductivity on a rinse tank or
Material lots, for example, batches of cleaning agents concentration of cleaning agent in a cleaning tank.
Environmental conditions The product outputs that would be monitored are
Multiple Shifts risk-based and would relate to the cleaning specifica-
Operators tions established previously. For example, the medical
Process operating windows defined in the OQ; device manufacturer may consider monitoring the TOC
for examples, see above. of the product on a regular basis to maintain confidence

Special edition: Cleaning Validation 77


Sebastian Clerkin

that the cleaning process is still in control. For newly de- facturing process step, establish cleanliness limits,
veloped cleaning processes, this monitoring can initially and then validate the cleanliness test methods and
be at a higher frequency and reduced as the manufac- the cleaning process itself (Figure 2).
turer gains more confidence in their process. Cleaning validation does not need to be difficult.
If medical device manufacturers take a methodical
revAlIDAtION approach and base each decision on sound scientific
Obviously, changes to the cleaning system and process rational, they will be able to establish a cleaning
should be assessed for their impact to the validation. In process that will consistently provide clean medical
addition, any changes to the manufacturing materi- devices to the market.
als used in the up-stream manufacturing processes
may have an impact on the cleaning validation. This reFereNCeS
includes manufacturing steps completed by an external 1. Code of Federal Regulations, Title 21, Quality System Regula-
vendor. If the change is deemed to have an impact, the tions, Part 820.70(e), 2013
cleaning validation should be either repeated in part or 2. Code of Federal Regulations, Title 21, Quality System Regula-
in full. Again, this needs to be a risk-based decision. tions, Part 820.70(h), 2013
The addition of new products to the clean line should 3. ISO 13485:2003, Medical devices -- Quality management sys-
be assessed to determine if they are a new worst-case. tems -- Requirements for regulatory purposes, section 7.5.1.2.1.
If they are considered a new worst-case product, the 4. Global Harmonisation Task Force Study Group 3, Quality
cleaning validation should be repeated. A recommended Management Systems Process Validation Guidance, January 2004
approach to determining if revalidation is required with 5. ISO 14969:2004, Medical devices -- Quality management
a new product is to update the worst-case product ma- systems -- Guidance on the application of ISO 13485: 2003, section
trix described earlier with the new product. If the new 7.5.2.1.1.5.
addition scores higher than the original worst-case, then 6. Code of Federal Regulations, Title 29, Part 1910 - Occupational
the cleaning validation must be repeated. Safety and Health Standards, Subpart Z Toxic and Hazardous
Substances, 2013.
CONClUSIONS 7. ISO 10993-17:2009, Biological evaluation of medical devices
The main steps in conducting a cleaning validation is -- Part 17: Establishment of allowable limits for leachable sub-
to assess the manufacturing material for each manu- stances.
8. ISO 10993-17:2009, Biological evaluation of medical devices
-- Part 17: Establishment of allowable limits for leachable sub-
stances.
9. ISO 10993-17:2009, Biological evaluation of medical devices
-- Part 17: Establishment of allowable limits for leachable sub-
stances.
10. H.J. Kramer, W.A Van Den Ham, W. Slob, and M.N. Pieters,
Conversion Factors Estimating Indicative Chronic No-Ob-
served-Adverse-Effect Levels from Short-Term Toxicity Data,
Regulatory Toxicology Pharmacology 23, 249-255, 1996.
11. Global Harmonisation Task Force, Quality Management Sys-
tems - Process Validation Guidance Edition 2, January 2004
12. ISO 14969:2004, Medical devices -- Quality management sys-
tems -- Guidance on the application of ISO 13485: 2003, Section
7.5.2.1.1.6.

AbOUt the AUthOr


Sebastian Clerkin, Ph.D., B.Sc., is the Founder of GMP Advisory
Services Ltd. and has more than 10 years experience in labora-
tory, pharmaceutical, and medical device manufacturing environ-
ments. He currently provides validation and regulatory expertise
to the medical device industry. He has direct experience across a
broad range of medical device types and process technologies. He
has a keen understanding of FDA regulatory requirements, and
has a strong background in process development and validation.
He has a Ph.D. from the University of Bristol, which involved
studying the toxicological and biological effects of prosthetic im-
plants. He holds a B.Sc. in Biochemistry from University College
Dublin, and is a member of AAMI and ASTM. E-mail: gmpadviso-
ryservices@gmail.com
Figure 2: Overall Process Flow to a Cleaning Validation.

78 Special edition: Cleaning Validation


Tim Sandle

Ensuring Sterility: Autoclaves,


Wet Loads, and Sterility
Failures | IVT
Tim Sandle, Ph.D.

ABSTRACT
Steam sterilization is a critical process in the pharmaceutical and
related industries. Modern autoclaves are computer-controlled and
reliably provide a defined sterilization cycle. When steam enters the
autoclave chamber and contacts with the item to be sterilized, steam
collapses (condenses). Water formed must be discharged through
condensate management or re-vaporized in order to prevent wet loads.
Repeated occurrences of wet loads are indicative of a major fault with
the sterilizer, potential non-sterilized materials, and other problems.
This paper considers some of the potential causes for wet loads and ad-
dresses some of the measures that can be taken to address occurrences.
Topics discussed include reasons for wet loads, causes including wet
steam, inadequate condensate removal, steam trams, pressure control,
and other causes; diagnosing problems by information collection, and
corrective actions. Corrective actions may include vacuum drying, heat-
ing the load before steam introduction, an air in-bleed phase, and other
approaches. Problems identified may be caused by a combination of
factors requiring a multidisciplinary team to evaluate potential causes.

INTRODUCTION
The most widely used sterilization method in the pharmaceutical in-
dustry remains steam sterilization in autoclaves (moist heat in the form
of saturated steam under pressure). This method is primarily appli-
cable to the terminal sterilization of products, stainless steel items, and
equipment not intended for single-use. With this method, sterilization
occurs as the latent heat of condensation is transferred to the load caus-
ing it to heat rapidly (1).
Modern autoclaves are computer-controlled and are generally very re-
liable. The autoclave acts as a pressure-cooker: Water boils at 100C at
atmospheric pressure; water boils at lower temperatures at lower pres-
sures, and water boils at higher temperatures at higher pressures. At a
steam over-pressure of one bar (a non-SI unit of pressure, exactly equal
to 100,000 Pascals), water boils at approximately 121C. This allows
the autoclave to produce temperatures above those that can ordinarily
be achieved. For sufficient time and with the correct conditions, such
temperatures can destroy bacterial endospores (2). More importantly,
the required temperature must be maintained for the required time in
order for the sterilization cycle to be successful (3).
Retrieving sterilized items from autoclaves only to discover that they
are wet results in a need to repeat the autoclave cycle. A wet item is
evidence of non-sterility. This is time consuming and expensive. This
forms part of the general rule with packages removed from autoclaves:
Any item that has been sterilized should not be used after the expira-
tion date has been exceeded or if the sterilized package is wet, torn, or
punctured (4).

Special edition: Cleaning Validation 79


Tim Sandle

When steam enters the autoclave chamber and that all condensate that gets separated from its energy
contacts with the product, it is important that the finds its way to the drain. Thus it is critical that the
steam collapses (condenses) on the product. This is in condensate made by each load item (including load-
order for the heat to be released to the load. However, ing furniture and wraps) must not migrate to other
the formation of water must be discharged through objects.
condensate management or re-vaporized in order to There are three possible scenarios for wet loads.
prevent contamination of the product. Removal of the These are:
excess water is important to prevent insulation of the
load from the steam. Visible moisture on outside of packs
Repeated occurrences of wet loads are indicative Moisture inside pack (e.g. moist towel)
of a major fault with the sterilizer. Water or damp Visible water inside the tray containing the load.
spots on the load insulate the intended product from
achieving temperature. This means that the load has Each of these is of equal seriousness.
not been subjected to the required lethality and it is
therefore not sterile. During sterilization, the wetness POTENTIAL CAUSES OF WET LOADS
in the steam clogs the pores of packed loads and pre- In examining the causes of wet loads there are several
vents the steam from properly penetrating wrapped areas to consider. These are discussed below:
loads or sealed pouches.
Furthermore, there is a post-contamination risk. Steam Supply
If the instruments or products absorb too much Wet steam can also be associated with wet loads;
humidity, resulting in wet loads at process termina- therefore steam needs to have a certain level pf dry-
tion, this dampness is an optimal habitat for bacteria ness (5). The moisture content of the steam (dryness
to thrive. Sterile items that become wet are consid- fraction) is measured as the weight of dry steam pres-
ered contaminated because moisture brings with it ent in a mixture of dry saturated steam and entrained
microorganisms from the air and surfaces. Moreover, water. Poor steam quality (insufficient dryness) can
if the sterile barrier system is still wet, it has lost its lead to excessive condensate formation within the
bacterial barrier properties and there is a major risk autoclave. This can arise when excessive demands are
of contamination. placed on the steam supply (essentially the boiler).
In addition to issues of sterility, wetness can cause Wet steam is steam at saturation temperature con-
corrosion or spotting on the instrument being steril- taining more than 5% water (6). Wet steam lowers the
ized. This can cause irreparable damage. heat transfer efficiency of steam, which results in an
Wet loads present a problem of non-sterility; a inefficient sterilization procedure.
problem in terms of recontamination of a load; and Steam quality can be assessed by measuring the
damage can occur to equipment. This paper consid- condensate discharge just ahead of the sterilizer. The
ers some of the potential causes for wet loads and measurement of steam quality is known as the dry-
addresses some of the measures that can be taken to ness fraction and this is dependent upon the steam
address the occurrence of wet loads. flow rate (7) where

REASONS FOR WET LOADS = dryness fraction


Water is heated up to the boiling point to generate
steam. As water reaches its boiling point at approxi- ww = mass of water flow (kg, or lb / unit time)
mately 100C, the temperature will no longer rise;
any energy added to the water will lead to evapora- ws = mass of steam (kg or lb / unit time).
tion of water thus creating steam. If steam touches
a material or gas with a lower temperature than the For example, if the water content of the steam is
temperature of the steam itself, the steam will give 5% by mass, then the steam is said to be 95% dry and
off energy and will condense and water is formed. In has a dryness fraction of 0.95. In this example, the
order to be able to evaporate the condensate energy is steam quality would be considered unsatisfactory. A
needed. The way to obtain sufficient energy is from satisfactory dryness fraction is generally regarded to
its environment. be 0.97 (8).
The reason why wet loads occur is that the con- Non-condensable gasses can also affect steam qual-
densate becomes separated from the condensation ity. Noncondensable gases should be removed before
energy and therefore cannot re-evaporate. To avoid the steam leaves the boiler. When they are not, they
wet loads, autoclave cycles must ensure that the modify the steam from being pure water vapor to a
condensate remains in contact with the materials it mixture of steam and gas. These become an unwant-
condensed on, or, alternatively, the process ensures ed contaminant that does not allow steam to make

80 Special edition: Cleaning Validation


Tim Sandle

contact with the item. Further, there is the issue of port, to ensure that they are clear and functioning
superheated steam (discussed below), properly.
Too much steam at too fast a rate can result in
Areas of Condensate Formation excessive water formation that might overwhelm or
Insufficient condensate being removed from the dis- swamp steam traps. Therefore steam traps need to
tribution system can result in in water lying around be properly sized during the autoclave design phase.
in the system and being drawn into the sterilizer
when the cycle demands it (pulsing). Signs of con- Pressure Control
densation can sometimes be seen from an examina- Given that autoclaves destroy microorganisms by
tion of the inside of the autoclave chamber. This may direct steam contact at the required temperature
resemble water lines or circles of water droplets (9). and pressure for a specified time, pressure control is
When this occurs, the steam circuit and supply important. Poor pressure control can cause variations
should be examined to determine where condensate in steam velocities. This often arises because mainte-
formation might occur. As steam travels, there are nance of the pressure-regulating valves (PRV) in the
many places and reasons why steam cools down and system is not always reliable. PRVs can change the
condenses. Reasons include dead legs in pipework condition of the steam and, in theory, can actually
or improperly trapped or insulated piping. When dry the steam as it passes through the valve.
steam comes into contact with piping connections it
can condense; moreover, poorly insolated piping will Autoclave Loads
cause steam to condense. Some condensate is natural What is being loaded into the autoclave can be a con-
and is taken care of by steam traps which are placed tributing factor. One mistake that some organizations
throughout the piping system. This can also be due make is to pack the chamber full of product or con-
to inadequate maintenance of the system and steam sumables in order to meet demand. However, dense
traps (see below). packing is frequently a cause of excessive condensa-
The further away the steam line is from the heating tion. This cannot always be satisfactorily flashed off
source/medium, then the more likely condensate is to by subsequent steam injections. Furthermore, large
form. This is connected to insulation, for the further quantities of materials and complex packaging can
away the steam line is then the greater the need for make effective steam circulation a challenge.
insulation. It is also worth considering:

Other Issues with Pipes and Steam Supply The assembly of instruments in the set. For
There are other factors relating to pipework and the instance, are the instruments well distributed in
supply of steam, which may need to be considered. the set? Is there too much metal mass in the set?
These are: High density items wrapped in absorbent mate-
rial can help to reduce condensate (10).
Where steam pipes are not properly insulated How the set was packaged. It is important to con-
steam will condense in the pipe sider if the wrapper is too large (traps condensate
The water separator should be close to the auto- inside set), whether the package was taped too
clave tightly (traps condensate inside package), the
If pipes run down towards the sterilizer, this can size of peel pouches for load contents (too much
cause steam to condense metal mass in the pouch). In addition, before
The steam generator should be located close to using wrapping material, it should be held at
autoclave, minimizing pipework length room temperature (20oC to 25oC) and at a rela-
If a steam generator does not have sufficient tive humidity of 30 to 60%.
capacity for the autoclave, pressure drop may Loading techniques. Consider reducing the metal
occur at times of peak demand for steam and a mass in the load and check whether linen items on
carry over of water can happen the top rack and metal items on the bottom rack.
If chemicals are added during the steam produc- Rigid containers. Containers should be spaced
tion, operational issues may occur. about one inch apart from each other. Stacking
should not be performed unless the container
Steam Traps manufacturer gives specific information on this
Steam traps are intended to discharge condensate process. Place containers beneath other items
while not allowing the escape of live steam. Problems since they produce condensate.
can arise if the steam trap is not functioning cor- Sterilizers should never be overloaded. There
rectly. To guard against this, the jacket steam trap should be sufficient space between items to allow
should be inspected, along with the valve at the drain for steam to permeate around the package.

Special edition: Cleaning Validation 81


Tim Sandle

Packaging Materials The materials that make up the load, for exam-
Packaging materials, used to wrap the items to be ple heavy metal mass can be a cause of wet
sterilized, are probably one of the most important packs (11). Further, some loads are erroneously
parts of the sterilization process. Packaging provides designed where they become water traps or they
a protective barrier for the sterile item and prevents are positioned in such a way the orientation leads
it from becoming re-contaminated. When used in them to retain water.
aseptic processing, the packaging should be of low Condense drains can become blocked. It is
particle generation. important to clean these at least twice a year.
Packaging needs to be well designed and of suffi- If they become saturated they will not take the
cient porosity so that steam can pass through packag- water out of the steam.
ing. In addition, packaging should be water repellent Sometimes the wet loads follow a pattern and
to provide a sterile field. Furthermore, the packag- may occur if the autoclave has not been used for
ing material must not change significantly during a few days. Here the cause may be the build-up
sterilization or release any substances that might of condensate in the supply pipework.
interfere with the action of the steam. When steam pipework is of an incorrect diam-
eter.
Other Factors If the drain valve is not closed, leading to drain
There are other factors that are linked with wet loads, water being returned.
although these are less common with a well-designed Consistency of operator control of the sterilizers
system. These are: and loading patterns.

Autoclaves are equipped with heated jackets to There are some actions that mask the wet load
assist in drying of the load and to ensure that issue rather than address it. One example is with the
condensate does not form on the chamber walls use of tray liners. Tray-liners do not solve the problem
during the hold period. If the autoclave jacket but disguises it. Tray-liners have a high absorption
has a different temperature than the chamber, capacity and are placed in the tray under the items to
wet loads may occur. To avoid this, larger steam be sterilized. This way the condense is absorbed and
jackets contain more heat and assist in the drying dispersed; however, non-sterility remains an ever-
process. present risk.
The autoclave carriage can be a factor. A stainless
steel carriage shelf creates more condensate than DIAGNOSIS
an equivalent aluminum carriage. To add to this, Collecting meaningful information is important for
the shape and size of the chamber can greatly establishing why wet loads occur. Whenever wet
affect the outcome of the load. packs occur, some of the information that should be
The preheat process. Here the problems range collected includes:
from poor steam penetration, superheat, dam-
aged sterile barrier systems and other problems. Date of wet pack(s)
When the autoclave doors have a different tem- Time of day
perature than the chamber or walls. Or where Load configuration (number of trays)
the jacket temperature is not uniform or lower Number and description of trays that were
than intended. reported as wet
When the item being sterilized touches the wall Selected cycle time (gravity or dynamic air
of the autoclave. removal)
Stacking two trays for holding loads within the Cycle temperature and exposure time
autoclave. The lower tray does not have sufficient Packaging material used for set(s) that were wet
energy to dry the condensation. Type of containment device used (e.g. Mayo tray,
Items being loaded in wet (anything going in metal mesh basket)
wet will come out wet.) When the steam makes Person(s) who prepared/wrapped sets
contact with the wet instruments additional con- Inventory list for set (to consider set configura-
densate and cause dripping on other instruments tion and weight of set)
that may not dry. Vacuum integrity
Overloading the autoclave. Large quantities of Vacuum performance
hard goods or even complex packaging can make Temperature/pressure calibration
proper steam circulation a challenge. Other Chamber level
parameters that may influence drying are the Chamber/jacket trap performance
density of the wraps and the design of the set Chamber valves (solenoid and check)

82 Special edition: Cleaning Validation


Tim Sandle

Jacket insulation integrity Superheated steam occurs when the temperature


Sterilizer exposure time of the steam is higher than its saturation pressure.
Sterilizer dry time. This usually happens when the pressure of the steam
drops as it moves through the steam supply system.
Analyzing such data can help to establish patterns This dries the steam and once there is a reduction in
and these can feed into the root cause analysis. moisture, less energy is required to raise the tempera-
ture of the steam. An autoclave fed with superheated
CORRECTIVE ACTIONS steam will function like a dry heat sterilizer, in which
In addition to running a diagnostic to address some or the killing of microorganisms is far less efficient than
all of the above areas, there are other actions that can the optimal saturated steam required for sterilization.
be considered in order to reduce the possibility of wet A further cause of superheated steam is if the re-
loads occurring. These include: duction valve is too close to the autoclave. This valve
should be at least 10 meters away. This is because
Vacuum Drying where the distance is too short the temperature will
In order to dry the load after the steam sterilization not be reduced. A further reason relates to steam
step, some users are tempted to employ excessively pressure, where this is too high. Although fundamen-
long deep vacuums at the end of the cycle. Because the tal to the laws of physics, it should not be forgotten
chamber is heated via the jacketing and the fact that that temperature and pressure are related to each
water will flash off into steam at a lower temperature other. Here it follows that the higher the pressure
under vacuum, this technique can be relatively suc- then the higher the temperature.
cessful. However, it will lead to an unnecessarily long
cycle time, and a potentially a non-sterile load can still Charge Rate Control
result since water can collect in certain areas. Some autoclaves do not have a proportional control
valve. This provides the control system capability to
Reducing Condensate Formation control the ramp up (charge) rate of steam injec-
A strong preventative measure to prevent the forma- tion during the cycle. If a chamber does not have the
tion of condensate is to ensure that the load itself is means to control the rate that steam that is added to
brought up to a certain temperature prior to the in- the chamber, this can lead to flooding of the chamber
troduction of steam. This can be accomplished by es- with steam. This, in turn, results in instant condensate
tablishing a load heat up phase at the beginning of formation. If the steam injection can be controlled so
the cycle. The phase functions at the dictate of jacket that it as it is ramped up more slowly, the amount of
set point, chamber drain (or load thermocouple) set steam (and subsequent condensate) can be more easily
point and a phase timer. The phase is not advanced controlled.
until the set points are achieved and the phase timer
elapses. From here it would then pass to a purge (or Filter Sterilization
pre-vacuum) condition. Some autoclaves are fitted with an air in-bleed filter
casing that can be sterilized in situ. Some manufacturers
Air-in Bleed Phase and the Problem of Superheated of autoclaves will sterilize this filter during each cycle,
Steam whereas other types require a special cycle to do this.
An alternative approach is to add a heated air in- This second approach reduces the wear on the filter ele-
bleed phase at the beginning of the sterilization cycle. ment, resulting in a much longer life-time and prevents
For this, a heat exchanger can be installed to the air repeatedly saturating the filter with steam. This latter
in-bleed assembly to enable a combination of vacuum factor would result in excessive moisture being brought
and air in-bleed pulses. These would bring the cham- into the chamber at the completion of the cycle. Thus
ber load up to temperature more quickly, reducing avoiding the in situ sterilization of the filter can be effec-
the condensate formation at the beginning of the tive in avoiding excessive moisture.
cycle. While a heat exchanger is not absolutely neces-
sary, it can be useful for particularly large chambers CONCLUSION
or dense loads. This paper has addressed the issue of wet loads from
As an alternative to the heat exchanger, increasing steam sterilization through an autoclave. Although
the jacket temperature set point can be considered. the probability of wet loads occurring can be avoided
Care must be taken to ensure that this does not in- through good validation and effective cycle develop-
terfere with maintaining the temperature uniformity ment, even autoclaves that have been in service for
within the chamber during the sterilize dwell phase. years can be subject to wet loads. This paper has
Merely increasing the jacket temperature will result outlined why wet loads are a major concern (non-
in a super-heating of the steam. sterility, the risk of re-contamination and damage to

Special edition: Cleaning Validation 83


Tim Sandle

equipment) and it has presented some of the reasons 5. Baker, J. & Godfrey, G. (2009). Water quality systems in sterile
why wet loads can occur. The paper has also out- processing. Healthcare Purchasing News, January 2009, 3235
lined, in addition to addressing the potential causes 6. Agalloco J. (2000). Steam sterilization and steam quality. Com-
of wet loads, some preventative measures that can be mentary. PDA J Pharm Sci. Technol.; 54(1):5962
adopted. 7. Shuttleworth K. (2000). The application of steam quality test
Humidity problems are a complex matter and are limits, Eur J Parenter Pharm Sci.; 5(4): 10914
in the majority of cases created by a combination 8. Association for the Advancement of Medical Instrumentation.
of different factors. Often wet load issues cannot be Steam sterilization and sterility assurance in health care facilities.
solved immediately and time is required, together ANSI/AAMI ST46. Arlington, VA, 2002:ANSI/AAMI ST46:2002
with a multidisciplinary team, in order to address the 9. Brown, J.M. & Bliley, J. (2008). How to solve wet packs and
problem. evaluate water issues. Materials Management in Healthcare, July
2008, (5052).
REFERENCES 10. Rutala WA, Weber DJ. (2000) Choosing a sterilization wrap. Infect.
1. Lemieux, P. (2006). Destruction of Spores on Building Decon- Control Today; 4:64,70
tamination Residue in a Commercial Autoclave, Appl. Environ 11. Chobin N, Furr D, Nuyttens A. (2004) Wet packs and plastic ac-
Microbial, 72(12): 7687-7693 cessory cases. Infect Control Today, 24, 28-30
2. Sandle, T. (2013). Sterility, Sterilisation and Sterility Assurance for
Pharmaceuticals: Technology, Validation and Current Regulations, ABOUT THE AUTHORS
Woodhead Publishing Ltd.: Cambridge, UK, pp93-110 Tim Sandle, Ph.D., Tim Sandle, Ph.D., is the head of the microbi-
ology department at Bio Products Laboratory Limited, a pharma-
3. Agalloco JP, Akers JE, Madsen RE. (1998) Moist heat sterilization-
ceutical organization owned by the UK Department of Health. Dr.
-myths and realities. PDA J. Pharmaceutical Sci. Technol. 52:346-50 Sandle is, additionally, a visiting tutor at the School of Pharmacy,
4. Dunkelberg H, Fleitmann-Glende F. (2006) Measurement of the Manchester University.
microbial barrier effectiveness of sterilization containers in terms
of the log reduction value for prevention of nosocomial infections.
Am. J. Infect. Control 34:285-9.

84 Special edition: Cleaning Validation

Potrebbero piacerti anche