You are on page 1of 9

BIOTECH

TRENDS
Viral Clearance Strategies for
Biopharmaceutical Safety
Part II: A Multifaceted Approach to Process Validation

Hazel Aranha* and Sharlene Forbes

P
rocess validation is an integral part of any in plasma products) and to estimate the robust-
manufacturing process. It assures a consis- ness of the process to clear potential adventitious
tent outcome during key manufacturing viral contaminants (that may have gained access
processes. FDA’s Center for Drug Evaluation and to the product) by characterizing the ability of the
Research, Center for Biologics Evaluation and Re- process to clear nonspecific “model” viruses.
search, and Center for Devices and Radiological Process analysis and evaluation. Ideally, strategic
Health define process validation as “establishing planning for process validation begins early in
documented evidence that provides a high de- product development. The first steps in the vali-
gree of assurance that a spe- dation process involve a critical analysis of the
In addition to fulfilling a regulatory cific process will consistently bioprocess to determine likely sources of viral
requirement, validation studies produce a product meeting contamination, including the pathogenic poten-
its predetermined specifica- tial of those contaminants, followed by process
maximize productivity and tions and quality attributes” characterization to identify which steps in the
minimize production failures by (1). Viral clearance valida- manufacturing process have the potential for viral
setting operational parameters tion studies do not meet the clearance.
requirement for conforming Each process step to be tested should be evalu-
and ensuring product consistency to a “predetermined specifi- ated for the viral clearance mechanism; that is,
and safety. Viral validation studies cation.” Nevertheless, the ob- whether clearance will be by inactivation, removal,
should be designed to justify the jective of the clearance study or a combination of both. A robust step is one in
is to document product which the effectiveness of the viral inactivation
selected operating conditions and quality and process speci- or removal is independent of variability in pro-
to document their adequacy in ficity to assure viral safety. duction parameters (4). Both serendipitous meth-
achieving expected process Viral clearance studies also ods (steps already in the manufacturing process,
may be called qualification such as chromatography and low pH-buffer elu-
performance. (This article was studies or clearance evalua- tion steps, that coincidentally clear viruses) and
published previously in BioPharm tion studies, terms used syn- methods deliberately incorporated for viral clear-
14 [5], 43–54, 90 [2001].) onymously with virus vali- ance (such as filtration and heat inactivation) are
dation studies in this article. usually validated.
Earlier articles in this se- What needs validating? Guidance in identifying
ries addressed general considerations related to which processes to validate comes from regula-
Hazel Aranha viral safety and pragmatic approaches to ensur- tory guidelines, industry trends, and other re-
is a senior staff scientist ing virological safety of biologicals (2,3). This ar- sources. Excellent resources include vendor in-
at Pall Corporation, 25 ticle highlights considerations in designing virus formation (such as that from manufacturers of
Harbor Park Dr., Port safety studies and cautions when developing ap- chromatographic resins, filtration systems, and
Washington, NY 11050,
propriate virus contamination control programs membranes) and the experience of others, espe-
tel. 516.484.3600, fax
516.484.3628, e-mail to be integrated into the manufacturing processes cially contract testing laboratories, who have ex-
hazel_aranha@pall.com, for biological products. Clearance validation case tensive experience in study design. Other sources
www.pall.com. studies help to illustrate these points. of information include published literature and
Sharlene Forbes is a seminar proceedings. A cautionary note is im-
senior associate at IDEC
Pharmaceuticals,
Study design portant here: Data reported in the literature may
www.idec.com. Analytical limitations make it impossible to demon- reflect results of a study rather than actual process
strate absolute viral absence. Viral validation stud- validation data and therefore may not address
*To whom all correspondence
should be addressed.
ies are, therefore, conducted both to document validation-related considerations at the manu-
clearance of viruses known to be associated with facturing scale. Additionally, details related to
the product (such as HIV, hepatitis, and parvovirus process specifics (flow rates and product concen-
26 Pharmaceutical Technology JUNE 2001 www.phar maportal.com
Biotech Trends

Choosing the challenge site


Three options exist for choosing a challenge site when conducting viral clearance studies.
Advantages Disadvantages
Option 1: All processes and samples are tested Immediate testing of samples Costs associated with using the vendor site
at the vendor site. Reduced data turnaround time Scheduling subject to vendor availability
Viral containment area on site Possible need to ship special equipment
No titer loss from additional freezing and thawing of samples Possible need to send trained personnel for
Access to troubleshooting experts extended times for larger studies
Extension of lab time may be unavailable if
studies do not go as planned
Option 2: Some processes and all samples are tested Control over more complicated processes Logistics of shipping virus and samples
at the vendor site while others are performed in-house. Inactivation studies completed by vendor Potential titer loss from additional freezing
(overlapping timeline for study completion) and thawing cycles
Testing easier to schedule with vendor Additional cost of shipping,and freezing and
Potential price break with frozen samples thawing controls
Potentially faster processing with adequate in-house staff Viral containment area required in-house
Access to troubleshooting experts
Option 3: All processes are performed in-house. Control over all processes and testing Viral containment area required in-house
Testing easier to schedule Viral vendors usually do not supply viruses
Potentially faster processing with adequate in-house staff without a testing agreement
Immediate testing of samples Need viral experts for study design,virus stock
No titer loss from additional freezing and thawing of samples supply,and troubleshooting

tration, for instance), which could poten- clearance steps that can be claimed and approach is to perform a small-scale run
tially affect virus clearance, may be vague. reduces the overall claim that can be es- and compare the purity and contaminant
Clearance data reported at these forums tablished for the entire process. The best profiles with the large-scale or develop-
should be taken as an indication of pos- compromise is to evaluate each of the in- ment history. Other process parameters
sible clearance and not as the expected dividual orthogonal steps separately and should be evaluated for their possible im-
value across different processes. then sum the amount of clearance ob- pact on viral clearance to determine
Multiple methods. Regulatory guidelines tained for the entire process. Although this whether they should be included in the
recommend incorporating multiple or- method has some limitations and intro- scaled-down study model.
thogonal methods for viral clearance; that duces errors by overestimating the clear-
is, methods that have independent (unre- ance, it is the only practical approach to a Worst-case conditions
lated) clearance mechanisms (5,6). complex problem. Regulatory guidelines recommend using
One misconception is that an entire Scaling considerations and identifying virus validation data to set in-process lim-
manufacturing process that includes, for critical parameters. Ideally, a process vali- its for critical process parameters (7). In
example, ion-exchange chromatography, dation is conducted at pilot or full scale, general, validations are usually conducted
pH inactivation, and detergent inactiva- but logistic limitations preclude that ap- at both process extremes. However, be-
tion can be tested by challenging with a proach for virus clearance validations. cause viral studies are costly and time con-
large spike of virus during the first step Evaluation of viral clearance strategies re- suming, testing at both process extremes
and sampling during subsequent steps. quires demonstration of the equivalence is seldom done. Instead, testing is per-
Logistically, this is impossible for two rea- of scalability from bench to manufactur- formed under worst-case conditions to
sons: Based on the product and possible ing scale and vice versa. The scale-down demonstrate the minimum clearance a
contaminants, most processes require a must truly represent what occurs in the step can provide. Worst-case conditions
demonstration of greater than 12–15 log manufacturing process; that is, the process will vary depending on the method used
of clearance for individual viruses, and it modeling must be accurate. Depending and are determined by those factors that
is impossible to grow mammalian viruses on the process, critical operating para- influence the clearance mechanism.
to such high concentration. Using a low meters to be conserved in scaled-down Virus removal methods. In chromato-
level of viral challenge will result in an ini- studies include volume, flow rates, con- graphic processes, depending on the resin
tial low viral load, with each successive tact time, and product and contaminant and binding mode, critical variables might
step in the bioprocess being challenged by load. The test material composition should include product and contaminant con-
fewer viral particles (assuming the previ- be similar in protein concentration, pH, centration, buffers, flow rates, wash vol-
ous steps are effective at inactivation or and ionic strength. Product generated by umes, and temperature, among others. Be-
removal of viruses). That type of study de- large- and small-scale processes should be cause of the competitive binding for
sign also restricts the number of viral similar in purity, potency, and yield. One interactive sites in product-binding mode
28 Pharmaceutical Technology JUNE 2001 www.phar maportal.com
Biotech Trends
chromatography, depending on the resin
being evaluated, the kinetics of virus bind-
Auditing a contract laboratory
ing is enhanced by the lowest product con- A quality system review is needed when choosing a contract testing laboratory.Several aspects of the lab
centration, thereby constituting the worst need to be audited.
case. Using the minimum wash volume
before elution encourages virus to elute Sample-related review
● Confirm log-in and receipt procedures (may be provided in writing if requested in advance).
with the product. As with all chromato-
● Check that the storage offers a controlled environment that meets customer specifications.
graphic processes, flow rate influences
● Make sure the identification procedure includes proper segregation between customers’samples to ensure
kinetics.
For contaminant- or impurity-binding lack of cross-contamination (microbial or otherwise).
● Ensure that samples and test solutions are tracked throughout the testing process.
mode chromatography, the worst-case
contaminant conditions can be achieved Test procedures and assay validation review
by either increasing the contaminant-to- ● Review the laboratory’s procedures relating to challenge viruses,cell lines,media,and reagents.Confirm that
product ratio or by loading the column the acquisition of viruses and cell lines are documented; tests are performed to confirm identity and purity;
with a larger volume of product than is SOPs are available for preparation,storage,and cultivation,and the acceptance criteria are defined for the
processed during manufacturing. This viruses and cell lines; and shelf life (stability) studies have been performed on the media and reagents.
provides competition between the virus ● Check that equipment is calibrated; that the installation qualification (IQ),operational qualification (OQ),and

and the expected contaminants and im- performance qualification (PQ) have been documented for the equipment; and that SOPs and acceptance
purities. Using the largest postload wash criteria have been established for the calibrated equipment.
volume anticipated in the manufacturing ● Determine whether summaries of protocols can be provided by the vendor (usually on request),and review

process (before the first cleaning step) will the protocols (available during the audit).
remove the maximum amount of virus ● Confirm data-recording procedures:whether SOPs are available for general record keeping and data

from the resin along with the product and processing,and all project-related items can be traced (lot numbers on cell lines,virus stocks,and reagents
thus constitutes the worst case. and equipment part number,for instance).
In filtration studies, depending on the ● Ensure that computer processes are validated:Software is validated; computer SOPs are available; the IQ,OQ,

filtration mode (direct or tangential and PQ for the computer systems are documented,and acceptance criteria have been established; data tables
flow), variables include the composition are generated and confirmed appropriately; and procedures are available for control of software changes with
of the solution to be filtered (the nature revalidations.
of the protein, the protein concentration, Technician training review
and other solution characteristics such as ● Ensure that training records are adequate for documenting an extensive training program for the technicians
pH and ionic strength); the process- before they perform critical validation studies.
associated factors such as the differential ● Monitor vendor safety training program.
pressure and the flux; and the appropri- ● Confirm vendor CGLP training program.
ateness of down-scaling, that is, the ratio
of filter volume to filter area (3). Investigation program review
Inactivation methods. All viral inactiva-
● Evaluate vendor SOPs for documenting evidence of investigations,conclusions,and corrective or preventative
tion methods should result in irreversible actions.
loss of viral activity. Viral inactivation ki- Internal auditing program review
netics are rarely linear, and sometimes a ● Determine whether the vendor’s quality assurance department reviews process during critical steps.

small residual fraction of the viral conta-


minant, resistant to the inactivation strat-
egy, can persist. The rate of inactivation, values of 3.9 or below but can be effective gent concentration combined with the
and thus the potential margin of safety in at different ranges for different lengths of shortest time is the worst-case condition.
the production process, can be assessed time. Choosing a pH value closest to neu- Temperature can be an important factor
by using kinetic inactivation experiments tral within the range tested provides a and may need to be evaluated at the ex-
at several points in the process. worst-case challenge, as does the shortest tremes during development to determine
Variables in inactivation studies include time. High protein concentrations gener- its effect. In general, the lowest tempera-
exposure time, temperature, product con- ally have a protective effect, so during viral ture provides the slowest kinetics.
centration, volumes, flow rates, the pres- clearance studies, product (protein) con- For heat inactivation studies, temperature
ence or absence of contaminant proteins, centration should be maximized (within distribution must be uniform, and timing
and container equivalence. Samples must process ranges) to ensure worst-case must begin only when steady state is
be homogenous before the treatment conditions. reached. “Worst case” in heat inactivation
strategy, and equipment (such as timers Variables in detergent inactivation stud- studies would constitute the highest sta-
and chart recorders) must be calibrated ies include concentration, exposure time, bilizer concentration used, the highest
and qualified. and exposure temperature. Additionally, product concentration, and the lowest
In pH inactivation studies, low pH inac- because detergents are viscous, samples temperature. If scaled-down studies are
tivation is generally considered robust at must be homogenous. The lowest deter- conducted, container equivalence must be
30 Pharmaceutical Technology JUNE 2001 www.phar maportal.com
Biotech Trends
demonstrated. Appropriately calibrated
equipment such as timers and chart
Table I: Viruses commonly used in viral clearance evaluation studies.
recorders must be used; equipment quali- Virus Family Genome Envelope Size (nm) Shape
fication is mandatory. Parvovirus (canine, parvoviridae DNA no 18–24 icosahedral
murine, and porcine)
Study design Encephalomyo- picornaviridae RNA no 25–30 icosahedral
Testing locations. Once the proposed manu- carditis virus
facturing process has been evaluated and Hepatitis A virus picornaviridae RNA no 25–30 icosahedral
the potential viral clearance steps identi- Poliovirus picornaviridae RNA no 25–30 icosahedral
fied, it is necessary to determine whether SV40 virus papovaviridae DNA no 40–50 icosahedral
the viral testing is to be done in-house or Bovine viral diarrhea togaviridae RNA yes 50–70 pleomorphic/
at a contract testing lab. We refer to the virus (BVDV) spherical
company scheduling viral validation stud- Sindbis virus togaviridae RNA yes 60–70 spherical
ies as the client and the provider of sup- Reovirus 3 reoviridae RNA no 60–80 spherical
plies or services as the vendor. Human immuno- retroviridae RNA yes 80–100 spherical
Three options exist for the location of deficiency virus (HIV)
the tests: The viral clearance studies can Murine leukemia retroviridae RNA yes 80–110 spherical
be conducted at a contract lab; depend- virus (MuLV)
ing on the specific processes and their Herpes simplex herpesviridae DNA yes 120–200 spherical
complexity, some of the testing can be virus
conducted in-house and the rest con- Pseudorabies virus herpesviridae DNA yes 120–200 spherical
tracted out; or all test processes can be Parainfluenza virus paramyxoviridae RNA yes 150–300 pleomorphic/
performed in-house. The “Choosing the spherical
challenge site” sidebar lists some of the Vesicular stomatitis rhabdoviridae RNA yes 70  175 bullet
advantages and disadvantages of each virus
option.
Virus stock availability comes into play of the documentation file, and the justifi- Study design technical features
when scheduling the evaluation of mul- cation for negating the unexpected results Choosing a panel of test viruses. No single
tiple process steps in a short time frame, must be included. indicator species can be used for virus vali-
more so if the viral stocks are purchased It is intuitively obvious that cost should dations — a panel of viruses must be cho-
from a vendor and the viral clearance not be the primary consideration in the sen. Choosing the appropriate panel of
studies are done in-house by the client. choice of a vendor. Vendor evaluation viruses depends on the source material
Before beginning studies that require large often begins with obtaining quotes. Nested (whether derived from plasma or from a
volumes of virus, consult the vendor to in the quote, the vendor usually recom- cell line) and on the clinical trial phase (1,
determine the availability of viral stock mends a study design. Such designs can 2, or 3) when the viral clearance will be
and the lag time. differ significantly between vendors and tested.
Contract lab considerations. Coordinat- often are not directly comparable. Cost- In general, the panel of test viruses
ing a multistep validation is logistically ing can vary and be based either on the should include relevant viruses (known
challenging and becomes more so when number of dilutions to be tested or on the or suspected viral contaminants) and
working with outside vendors. When eval- expected clearances, with the cost of ad- model viruses. Examples of relevant
uating a potential vendor, consider its tech- ditional dilutions built into the quote. viruses are HIV and hepatitis B and C
nical capabilities, customer service, and Customer service is an integral but often viruses, which are known blood product
cost. Ultimately, the quality of the final a neglected aspect of vendor selection. Un- contaminants. Some relevant viruses such
product (the viral clearance claim) is in- foreseen test-related factors can sometimes as hepatitis B and C viruses are difficult
fluenced by the entire package: its sound necessitate changing the strategy or re- to propagate in vitro. In those cases spe-
study design, the appropriateness of its peating a test. Customer-friendly contacts cific model viruses can be used. Specific
testing regimens, and correct interpreta- can smooth the way when midstudy model viruses are viruses that resemble
tion of its data. Although contract labs can changes are required or questions about known viral contaminants. For example,
offer guidance and assistance in study de- billing surface. The time it takes for report bovine viral diarrhea virus (BVDV) and
sign and data interpretation, it is the turnaround or to receive a “quality re- the Sindbis virus have been used as mod-
purview of the client to agree, acknowl- viewed” report also can play a part in last- els for the hepatitis C virus. Similarly,
edge, and justify the scientific data. In- minute evaluations. murine leukemia virus (MuLV) often is
consistent or anomalous virus clearance Before commissioning a virus valida- used as a model for noninfectious endo-
data, which may be a consequence of poor tion study, the vendor should be audited, genous retroviruses associated with ro-
study design or badly conducted tests, can- if possible. The “Auditing a contract lab- dent cell lines. Additionally, nonspecific
not just be ignored. The results must be oratory” sidebar lists some of the criteria model viruses also are included in the test
presented to regulatory authorities as part to use when evaluating a vendor. panel to characterize the theoretical clear-
32 Pharmaceutical Technology JUNE 2001 www.phar maportal.com
Biotech Trends
ance capability of the manufacturing
process — the robustness of the process.
Table II: Examples of reduction in Table III: Loss of virus titer because
Nonspecific model viruses come in dif- viral clearance claim due to viral of prefiltration of spiked test
ferent sizes and varied physicochemical spike–related factors during a filter material and its effect on the viral
and biophysical characteristics. They are validation study.* clearance claim.*
not expected to be associated with the Expected** Actual Sample Sample
product but are included to address the- Sample titer/mL 4.9  104 4.9  104 One** Two†
oretical safety concerns and to increase Volume (mL) 300 82.1 Input titer/mL
confidence in the ability of the process to Total challenge (log10) 6.3  104 3.85  102
handle unknown or undetected viruses. (log10) 7.17 6.60 Volume (mL) 127 137‡
Table I lists examples of viruses that have Total recovery Total input
been used in virus validation studies. (log10) 2.16 2.16 concentration
Because of the high costs associated Log titer (log10) 6.90 4.72
with an entire virus validation package, reduction 5.01 4.44 Total recovery
preliminary testing with surrogates such (log10) 4.69 1.88
* Test virus used was pseudorabies virus
as bacteriophages can be undertaken in Log titer
(PRV), the product was a 4.1 mg/mL
some cases. Such testing is, of course, rele- monoclonal antibody. reduction 2.21 2.84§
vant only if removal is size-based, as in fil- **“Expected” refers to the initial spiked
tration; if clearance depends on a partic- * Challenge virus used was
volume and anticipated throughput.
xenotropicmurine leukemia virus (X-
ular physicochemical or other surface MuLV); the filter was a Ultipor VF grade
characteristic of the virus, surrogate test- visable to work with high titer virus stocks, DV50 (Pall BioPharmaceuticals).
ing cannot be used. The applicability of care must be exercised to ensure that the **Sample one is the prefiltered sample
bacteriophages as surrogates for mam- methods used to concentrate the virus and used product filtered through a
malian viruses in filter validation studies 0.2-m filter.
stock and achieve high stock titers are not
† Sample two was virus-reduction filtered,
has been discussed (8). conducive to aggregation. using 0.2 m of effluent.
Different clinical trial phases may re- The quality of the virus stocks, as mea- ‡ Includes wash volume (to maximize
quire different virus choices. Before phase sured by the presence of viral aggregates, product recovery).
1, the clearance of known viral contami- cell debris, or other particulates, can in- § Calculated based on input concentration
nants (HIV in the case of plasma-derived in the 0.2-m prefiltered load.
fluence results by falsely enhancing or re-
products) or specific model viruses is usu- ducing viral clearance. For example, extra
ally assessed. During phases 2 and 3, viral cell debris during a contaminant-binding membrane particulates. Virus-stock solu-
clearance studies should include both spe- chromatography process may compete tions often contain stabilizers (such as
cific model and nonspecific model viruses. with the virus for binding sites on the resin, bovine serum albumin) and other addi-
The entire virus panel evaluated for phase causing a decreased clearance value. In a tives (such as serum), and those can in-
2 or 3 products should be evaluated again tangential-flow filtration process, a virus terfere with evaluating the clearance
if final manufacturing conditions change stock containing high amounts of cell de- process. Suggested remedies include cen-
or if significant scale-up occurs during or bris would enhance virus retention by po- trifugation of the virus stock and buffer
after phase 3 trials. larizing the membrane. In direct-flow fil- exchange just before use. Additional ma-
Virus stock considerations. The quality of tration, a membrane prematurely clogged nipulation of the stock can lead to in-
the stock preparation and of the virus by cell debris cannot filter the entire load creased aggregation. A small-scale run
spike titer will significantly influence the volume and, therefore, full log clearance using storage solution for a virus spike
test results and the ability to make a viral cannot be claimed. Table II shows the clear- (mock spike without the virus) provides
clearance claim. Unfortunately, no stan- ance expected from a filtration study when useful information and should be done
dardized methodologies are available for using the expected load and filtrate vol- when possible.
preparing and purifying virus stocks. umes (300 mL) compared with the actual Lot-to-lot differences can occur even
Those methods will vary from vendor to volume (82.1 mL) collected as filtrate. Be- when the lots are produced by the same
vendor and must therefore be discussed cause less than one-third of the desired vendor. For example, in one validation
with the vendor when designing the study. volume was filtered, the overall clearance study under identical challenge conditions,
In general, starting with a high viral load claim made for that filtration step was we challenged two aliquots from the same
to challenge a process step will maximize decreased. batch of test material using a viral spike
the potential viral clearance claim. The vol- Variables associated with virus prepa- from two different virus stocks purchased
ume of virus spiked into the challenge ma- ration (such as stabilizers in stock, aggre- from the same vendor. Each filter mem-
terial and the virus stock titer combine to gation, and debris) can depend on the brane was theoretically challenged with
determine the total virus titer in the spiked virus and the vendor. Some vendors take approximately the same number of parti-
product. The virus density depends pri- precautions to reduce cell debris, whereas cles as were contained in the certified titer
marily on the biology of the virus and can other viral stocks are minimally purified provided by the vendor. Test material
vary from virus to virus. Although it is ad- and can contain significant amounts of spiked with virus lot 1 behaved similarly
34 Pharmaceutical Technology JUNE 2001 www.phar maportal.com
Biotech Trends
and mass-to-surface area specification. If
Table IV: Virus inactivation by a low-pH product shows the importance of the viral spike contains a large number of
“media control.”* aggregates (which consequently increase
the viral size), the prefilter can effectively
Input Concentration Volume Total Viral Load Log10 Titer clear a significant amount of virus, re-
Sample (TCID50/mL) (mL) (Log10TCID50) Reduction** ducing (by a few log) the viral load to the
T  0 min 1.12  105 19.5 7.52  0.32 viral removal filter and decreasing the log-
T  5 min 6.76  100 21.1 3.33 5.82 reduction claim that can be made for that
T  30 min 6.76  100 21.1 3.33 5.82 step.
T  60 min 6.76  100 21.1 3.33 5.82 In our experiments, prefiltration using
Media a 0.2 m-rated filter resulted in a 2.2 log
T  0 min 4.79  106 20 7.98  0.24 reduction (see Table III) and thus resulted
in a decreased claim — a log titer reduc-
Media
tion (LTR) greater than or equal to 2.84
T  60 min 2.00  106 20 7.60  0.37 0.38
— because of viral clearance by the pre-
* Challenge virus used was xenotropic murine leukemia virus (X-MuLV). filter. We are still investigating whether the
**Log10 titer reduction was calculated based on the starting media control titer (T  0)
loss from using the prefilter was from cell
because of the virus inactivation by the test product.
debris or aggregation.
Viral spike volumes will affect clearance
Table V: Detection of low levels of virus using large-volume sampling studies (especially in cases of large
amounts of debris) and should generally
assessment.* be maintained at 10% or less of the final
Inverse Volume Log10 Log10 Titer volume to keep the feed stream represen-
Sample Log10TCID50/mL (mL) Adjusted Titer Reduction** tative of the manufacturing process (7).
Spiked load 4.79  103 108.4 5.72  0.40 Using a 5% instead of a 10% virus spike
Adjusted spiked load †
6.31  105 108.4 7.84  0.35 reduces the number of particles only by
Flow-through/wash 6.76  101 168 4.06 3.78  0.35 half, or approximately 0.5 log. For exam-
Eluate 6.76  101 109 3.87 3.97  0.35 ple, in one of our filter validation studies,
Eluate 2.57  101 109 3.45  0.15 4.39  0.38 using a 10% viral spike resulted in clog-
Large-Volume Sampling ging of the virus filter (in spite of pre-
Hold control 1.12  104 108.4 6.08  0.32 1.76  0.47 filtering the spiked input volume through
Media control (start) 6.31  105 a 0.2 m-rated prefilter). Because filter-
Media control (end) 4.79  105 0.12  0.53 ability tests at the bench and pilot scale
had indicated appropriate sizing, filter
* Xenotropic murine leukemia virus (X-MuLV) was removed by phenyl-sepharose
chromatography.
plugging was probably associated with the
**Log10 titer reduction was calculated based on the media control titer (start) because of virus spike. When the viral spike volume
virus inactivation by the test product. was decreased to 5%, the equivalent vol-

Spiked load was adjusted based on the media start control. ume of batch throughput was readily fil-
tered. (Anecdotally, using a 5% spike al-
to development work done without virus. crease clearance. Some titer loss can be lowed even greater volumes to pass
However, test material spiked with virus expected with a prefilter, which will re- through similar virus-removal filters.)
lot 2 showed a dramatic decrease in flow duce the amount of virus contacting the
rate, and the test had to be aborted pre- test membrane. The amount of loss will The importance of controls
maturely because of flow decay. Differences depend on the method of virus stock The importance of controls in the virus
between those two virus stock lots meant preparation and on the aggregating effect study design cannot be overemphasized.
that the challenge level (particles/cm2) was of the test material on the virus. The ef- Controls allow clearance effects to be at-
different in those tests: Virus lot 1 had a fect of using a prefilter, both on the prod- tributed to the treatment procedures
particle load/cm2 of 9.61 log, whereas the uct and on the level of viral spike, must rather than to test design artifacts or
particle load for virus lot 2 was 9.80 log. be acknowledged. Testing must be per- methodology. Several controls should be
Considering previous experience with this formed in advance to determine the included in virus clearance evaluations.
antibody, we concluded that virus lot 2 amount of product loss and dilution from Before viral clearance assays are con-
probably had a higher level of cell debris. any filter wash and the volume retained ducted, the product must be shown to
The visible cloudiness of the virus stock from passage over the prefilter. If the have no inhibitory effect on either the in-
lent credence to that conclusion. product is lost or diluted, that loss and dicator cell line (generalized cytotoxicity
Prefilters are sometimes used before dilution must be compensated for so that control) or the test virus (viral interfer-
virus removal filtration to remove virus the starting material loaded on the test ence studies). Cytotoxicity and viral in-
aggregates or debris that can falsely in- filter meets the minimum concentration terference controls are often conducted
36 Pharmaceutical Technology JUNE 2001 www.phar maportal.com
Biotech Trends
considerably in advance of the
validation study to ensure that
Risk calculations
the clearance capacity has not The required level of clearance is assessed in relation to the perceived hazard to the target population and guided by a
been overestimated because of risk–benefit analysis like the one below,which uses xenotropic murine leukemia virus (X-MuLV) as the challenge virus.
test-related considerations. Retrovirus-like particles per mL 1.62  107 particles/mL
The cytotoxicity control is in- Antibody titer 0.274 mg/mL
cluded to ensure that any indi- Weight of average person 80 kg
cator cell cytopathology ob- Dose per mass 15 mg/kg
served during the study is due One dose 1200 mg
to the virus alone. The cells are Viral clearance factor unknown
exposed to process components The total amount of retrovirus-like particles in one dose 
(product intermediates and
buffers) in the absence of virus {(1.62  10 particles/mL)(1200 mg/dose  0.274 mg/mL)}  10 particles/dose  7.09  10
7 6 16

for the length of time that the or 16.85 log minimum clearance required to achieve a clearance value of one particle per million doses.
test material will be in contact
with the cells. A cytopathic or
morphological effect relative to the un- effects alone did not affect titers within same clearance step. Hold controls are
exposed control cells is an indication of the assay variability. The probable cause therefore essential when evaluating viral
cytotoxicity. of the decreased input titer was inactiva- clearance.
Viral interference control determines tion from the starting material (product) A freeze–thaw control is defined differ-
whether process components interfere components. Because this is an inactiva- ently by different vendors. One definition
with the capacity of the test virus to infect tion step, the media control titer (T  0) is an aliquot of virus stock thawed and
the indicator cell line. Essentially, follow- can be substituted as the starting titer, with held unopened at process temperature and
ing exposure of the indicator cell line to the final result claimed as inactivation time. That test provides information about
the process component, the cells are ex- under those process conditions. Media the thawing of the virus in concentrated
posed to the virus and evaluated to deter- control titers are often substituted as an form compared with the effects of thaw-
mine any loss of infectivity and thus viral estimate for the initial titer when the test ing on a diluted form of the virus from
interference by the product. If either the material is virucidal. the media controls. Another approach is
cytotoxicity or the viral interference con- The hold control ensures that the test using the media control to verify that the
trols demonstrate positive results, the test virus is stable in the presence of test ma- freeze–thaw cycle does not affect the stock
material can be diluted (to determine a terials throughout the duration of the test. titer. Enveloped viruses especially can be
noninhibitory concentration), or the test The hold control involves virus-spiking affected by a slow transition between
solution can be neutralized or otherwise the starting material, then holding the frozen and thawed states.
adjusted. starting material at the process tempera- Stability and storage controls are primar-
A media control consists of virus spiked ture for the length of the process time. This ily a concern if process challenges are per-
into the virus cultivation medium at the control essentially demonstrates inactiva- formed somewhere other than at the virus
same ratio as the test material and helps tion effects that are a consequence of the vendor’s site. If virus stocks are to be
to determine inactivation by the test ma- product (the starting material). Loss shipped to another location, the stocks are
terial. Media “start” and “end” controls demonstrated by a hold control is unre- thawed, processed for the manufacturing
demonstrate the stability of the test virus lated to the clearance strategy under study step to be challenged, and frozen for later
under the test conditions. and should be evaluated accordingly. shipping. These controls may differ from
Table IV demonstrates the importance In one of our chromatography valida- challenges performed at the vendor site
of the media control. To validate a pH in- tion studies, a monoclonal antibody so- because many vendors assay the test ma-
activation step, the test material at neutral lution in 150 mM NaCl, 50 mM Tris pH terial immediately. A reduction in this test
pH was spiked with virus (X-MuLV), ag- 7.7  0.1 buffer was spiked with pseudo- can affect the final clearance claim, so the
itated, and then sampled. The remaining rabies virus (PRV). This buffer was not freeze–thaw stability should be reviewed.
volume was then adjusted to below pH 4. expected to affect the stability of PRV. Shipping controls determine whether
An immediate drop in virus titer was ob- However, the test material spiked with the temperature changes that may occur when
served: The expected titer at T  0 min virus and held for the process time (that the virus is shipped to a different site
was 6.52 log; the actual titer in the sam- is, the hold control) did reduce the titer would affect the titers.
ple was 5.05 log, a viral titer drop of 1.63 by 1.21 log, suggesting that the decrease
log immediately after spiking. No virus in the titer could be attributed to inacti- Pitfalls and cautions
was detectable at 5 or 60 min after virus vation. The media controls confirmed that As mentioned, a “good” viral clearance
spiking. The media controls demonstrate conclusion. Data interpretation for chro- validation study is detailed and well de-
that the number of virus particles expected matography steps can sometimes be com- signed. Scaled-down studies are, at best,
was achievable (no reduction in titer for plicated by the combination of inactiva- approximations of the conditions that will
the stock virus). Time and temperature tion and removal occurring during the be achieved during manufacturing, and
38 Pharmaceutical Technology JUNE 2001 www.phar maportal.com
Biotech Trends
the validity of the clearance data reflects available limits of detection. Virus quanti-
the accuracy of the process modeling and tation methods can be modified to enhance
Example of the calculation of viral
study design. Several pitfalls can be asso- sensitivity by using additional replicates clearance (log titer reduction) for
ciated with small-scale validation studies. and increasing inoculation volumes. the overal process for a monoclonal
Virus-related considerations. Viral spike- It is important to avoid overestimating antibody product.*
related perturbations can make a process the effectiveness of a viral clearance
Log Reduction Factor
nonrepresentative of the actual manufac- process that fails to detect low levels of
Ion-exchange
turing conditions. Also, model viruses residual virus. Large-volume assessments
chromatography 5.39
used in process validation studies are at can be used as supplements to conven-
Nanofiltration 5.14
best just that — models — and a wild- tional titration methods to increase the
Low-pH inactivation 5.85
type strain may not behave the same way probability of detection for extremely low
Detergent inactivation 6.06
as a laboratory strain. virus concentrations. Table V demon-
Total clearance 22.44
Inaccurate process modeling. Conditions strates that when a routine sampling pro-
in small-scale validations may be incon- tocol (volume assayed 0.4 mL) was used, *Challenge virus used was xenotropic
gruent with the process-scale conditions. no virus was detected in the eluate from murine leukemia virus (X-MuLV).
For instance, columns used only once for a Phenyl Sepharose chromatography col-
a validation study may not reflect the abil- umn. However, large-volume sampling assay variability, report the lowest clear-
ity of columns used repeatedly (during (total sample volume of 3.2 mL) allowed ance value as the conservative approach.
manufacture) to remove virus consis- detection of low levels of the challenge Other approaches, if justified, may be ac-
tently: Certain sites on resins can become virus (2.5  101/mL). ceptable. The procedure chosen should be
blocked with repeated use, reducing the logical, defendable, and defined.
effectiveness of virus removal over the Estimating viral clearance Achieving the goal. A key factor affecting
resin lifetime. Establishing clearance for an entire process viral clearance for the overall process is the
Sample-related considerations include the (the overall clearance value) requires at amount of product required to produce a
use of nonrepresentative samples in viral least two orthogonal, robust methods of single dose. The required level of clearance
validations. For example, the proper in- viral clearance. The individual steps must is assessed in relation to the perceived haz-
termediate or the actual product sample possess fundamentally different mecha- ard to the target population and is guided
may not have been used; the sample may nisms of virus removal or inactivation for by risk–benefit analysis. For example,
not be representative of the protein con- the values to be considered cumulative. murine cell lines are frequently contami-
centration, the pH, or other solution char- Only data for the same model virus can nated with endogenous retrovirus-like par-
acteristics such as ionic strength; and sam- be cumulative because viruses vary greatly ticles that pose, primarily, a theoretical
ples may be nonhomogenous because of in their inactivation or removal profiles. safety concern (10–12). The putative risk
inadequate mixing. Clearance estimates and their variances stems from the cell lines’ morphological
Assay-related considerations include fail- are calculated for each orthogonal unit and biochemical resemblance to tumori-
ure to evaluate buffer toxicity, poor model operation; total virus reduction is the sum genic retroviruses. Chinese hamster ovary
virus selection, lack of appropriate con- of individual log reduction factors. In cases (CHO) cell lines containing endogenous
trols, and poor standardization of viral as- of complete clearance, a theoretical titer retroviruses at levels of 106–109 parti-
says. Critical criteria for assay performance value is based on a statistical distribution cles/mL (as seen by electron microscopy)
are accuracy, reproducibility, repeatabil- (the Poisson distribution). Table VI pro- are deemed acceptable if adequate retro-
ity, linearity of range, limit of detection vides cumulative virus clearance values virus clearance is demonstrated in the
(LOD), and limit of quantification (LOQ), for MuLV. manufacturing process.
and all must be validated (9). Few guidelines describe how to perform Risk calculations to determine retro-
Product dilution steps (because of viral final data interpretation. All existing guide- viral load per dose are shown in the “Risk
interference or other toxicity-related con- lines state that clearance steps yielding one calculations” sidebar. This example as-
siderations) will affect assay results and the log or less of clearance are considered neg- sumes a one-time dose of 1200 mg to the
ability to make a high viral clearance claim. ligible and are not to be included in the patient. Using the conservative goal of as-
For example, high salt concentrations, pH overall clearance for a process. The FDA– suming a probability of viral contamina-
extremes, or other sample conditions can ICH guidelines recommend running repli- tion of one particle per million doses of
interfere with virus titration. Decreasing cate challenges presumably to increase product, and assuming a retroviral load
the volume assayed (because the sample is confidence in the data (9). However, no of 1.62  107 particles/mL in the starting
diluted) will result in decreased sensitivity specific guidance is provided on how to material, the purification process for this
and is especially important when no virus report replicate data, nor on how data in- product would have to demonstrate a
is detected and a theoretical limit titer for terpretation can be made consistent for minimum log clearance of 16.85 log to
the sample is calculated. each process step and across all processes. achieve the stated goal of one viral parti-
In general, greater viral reduction can be One suggestion is to average the replicates cle per 106 doses. The clearance goal is usu-
claimed with more observations from using if each value is within the assay variabil- ally chosen based on product use and the
larger volumes and from testing the lowest ity. If replicates differ by more than the risk to the patient population. The extent
40 Pharmaceutical Technology JUNE 2001 www.phar maportal.com
Biotech Trends
of necessary product testing depends on References
the source and nature of the product, the 1. Center for Biologics Evaluation and Re-
stage of product development, and the search, Guideline on General Principles of
Process Validation (FDA, Rockville, MD), May
clinical indication. Serious or immediately 1987, pp. 5–9.
life-threatening conditions for which no 2. H. Aranha, “Viral Clearance Strategies for
effective alternative treatment exists can Biopharmaceutical Safety, Part 1: General
justify abbreviated testing. Considerations,” BioPharm 14 (1), 28–35
(2001).
3. H. Aranha, “Viral Clearance Strategies for
Offering assurance Biopharmaceutical Safety, Part 2: Filtration
Virus clearance studies are an integral For Viral Clearance,” BioPharm 14 (2), 32–43
component of the multifaceted approach (2001).
recommended to ensure the safety of 4. H. Willkommen, I. Schmidt, and J. Lower,
biologicals and biopharmaceuticals. Ulti- “Safety Issues for Plasma Derivatives and
Benefit from NAT Testing,” Biologicals 27,
mately, the quality of the final product (the 325–331 (1999).
viral clearance claim) is influenced by the 5. Committee for Proprietary Medicinal
entire package, by sound study design, ap- Products (CPMP), Rev. 2: Note for Guid-
propriate testing regimens, and correct ance on Plasma-Derived Medicinal Products
data interpretation. (CPMP/BWP/269/95), July 1998.
6. “Guidance on Viral Safety Evaluation of
The study design is critical to viral vali- Biotechnology Products Derived from Cell
dations. Process modeling must be accu- Lines of Human or Animal Origin,” Federal
rate. Careful and comprehensive studies Register 63 (185), 51074–51084 (1998).
should be conducted to ensure scientifi- 7. Committee for Proprietary Medicinal Prod-
cally and statistically sound results. Virus ucts (CPMP), Note for Guidance on Virus
Validation Studies: The Design, Contribution,
quantitation methods can be adapted to and Interpretation of Studies Validating
suit specific study needs by including ad- the Inactivation and Removal of Viruses
ditional replicates, increasing inoculation (CPMP/BWP/268/95), February 1996.
volumes, and testing at multiple sampling 8. H. Aranha-Creado and H. Brandwein, “Ap-
periods. Factors such as test volumes, batch plication of Bacteriophages as Surrogates for
Mammalian Viruses: A Case for Use in Fil-
volumes, and test sensitivity determine the ter Validation Based on Precedents and Cur-
probability of detecting low-level virus rent Practices in Medical and Environmen-
concentrations. tal Virology,” PDA J. Pharm. Sci. Technol. 53,
Process validation coexists with and sup- 75–82 (1999).
plements in-process testing. In addition to 9. A.J. Darling, J.A. Boose, and J. Spaltro, “Virus
Assay Methods: Accuracy and Validation,”
fulfilling a regulatory requirement, it maxi- Biologicals 26, 105–110 (1998).
mizes productivity by setting operational 10. K.P. Anderson et al., “Defective Endogenous
limit parameters, minimizing production Retrovirus-Like Sequences and Particles of
failures, and providing a high assurance of Chinese Hamster Ovary Cells,” Dev. Biol.
product consistency and safety. Viral vali- Stand. 75, 123–132 (1991).
11. C. Liptrot and K. Gull, “Detection of Viruses
dation studies should be designed to jus- in Recombinant Cells by Electron Micro-
tify the selected operating conditions and scopy,” Animal Cell Technology: Devel-
to document their adequacy in achieving opments, Processes, and Products, R.E.
expected process performances. Although Spier, J.B. Griffiths, and C. MacDonald, Eds.
any validation study only approximates the (Butterworth-Heinemann Ltd., Oxford, UK,
1991), pp. 653–656.
real situation, it identifies critical parame- 12. K.K. Lueders, “Genomic Organization and
ters affecting viral clearance and provides Expression of Endogenous Retrovirus-Like
a framework for setting operational limits Elements in Cultured Rodent Cells,” Biolog-
and worst-case conditions. icals 19, 1–7 (1991). PT
This article has highlighted the various
cautions and considerations to be borne
in mind when designing and developing
appropriate virus contamination control
strategies into manufacturing processes for
biological products and is designed to pro-
vide suggestions and highlight pitfalls
based on personal experience and indus-
try trends.

Circle/eINFO 31
www.phar maportal.com

You might also like