You are on page 1of 6

554612

research-article2014
JLAXXX10.1177/2211068214554612Journal of Laboratory AutomationPeris-Vicente et al.

Review Article
Journal of Laboratory Automation

Validation of Rapid Microbiological Methods


2015, Vol. 20(3) 259­–264
© 2014 Society for Laboratory
Automation and Screening
DOI: 10.1177/2211068214554612
jala.sagepub.com

Juan Peris-Vicente1, Samuel Carda-Broch1, and


Josep Esteve-Romero1

Abstract
Classical microbiological methods currently have unacceptably long cycle times. Rapid microbiological methods have
been available on the market for decades and have been applied by the clinical and food industries. However, their
implementation in the pharmaceutical industry has been hampered by stringent regulations on validation and comparison
with classical methods. To encourage the implementation of these methodologies, they must be validated to assess that the
results are straightforward. A comparison with traditional methods should be also performed. In this review, information
about the validation of rapid microbiological methods reported in the literature is provided as well as an explanation of
the difficulty of validation of these methods. A comparison with traditional methods is also discussed. This information is
useful for industries and laboratories that can potentially implement these methods.

Keywords
analysis, guideline, laboratory, microorganism, validation

Introduction growth. These limitations have motivated sponsors and


manufacturers to develop rapid microbiological methods
Gene therapy products and human somatic cell therapy are (RMMs) based on techniques that reportedly yield accurate
emerging and present multiple challenges to safety, purity, and reliable test results in less time and often with less oper-
and potency. Gene therapy products include vectors (such ator intervention. Therefore, the use of RMMs for cell-
as nucleic acid, virus, or genetically modified microorgan- based products might provide rapid results that could be
isms) that are directly administered to patients and cells that applied for product release.5
are transduced with a vector ex vivo prior to administration RMMs are methods designed to provide performance
to the patient. Certain genetically modified microorganisms equivalent to the sterility traditional methods while provid-
(bacteria and yeast), cellular therapy products, and cells ing results in significantly less time. In general, RMMs
transduced with a gene therapy vector present similar chal- involve technologies that can be growth-based, viability-
lenges to sterility assurance and can be considered cell- based, or surrogate-based cellular markers for a microor-
based products. These products cannot be cryopreserved or ganism (e.g., nucleic acid based, fatty acid based). RMMs
otherwise stored without affecting viability and potency. are usually automated, and many have been used in clinical
Most cell-based products are manufactured using aseptic laboratories to detect viable microorganisms in patient
manipulations because they can undergo sterile filtration or specimens. These methods show improved sensitivity in
sterilization.1 However, the incubation times needed for detecting changes in the sample matrix (e.g., by-products of
these methods is usually too long.2 Thus, rapid and effective microbial metabolism), under conditions that favor the
testing is needed because many cell-based products have a growth of microorganisms. The RMMs can be applied in
potential short dating period, which often necessitates the manufacturing of cell-based products to test the compo-
administration of the final product to a patient before steril- nents (raw material, excipients), for in-process testing, for
ity test results. Because of the challenges associated with drug substance testing, and to test the drug product in its
cell-based products, there is a significant need to develop, final container.6
validate, and implement sterility test methods.3
Traditional methods, described by Title 21 of the Code
of Federal Regulations, 610.12 (21 CFR 610.12),4 are used 1
QFA, ESTCE, Universitat Jaume I, 12071, Castelló, Spain
for the isolation of microorganisms. However, the present Received May 13, 2014.
limitations related to cell-based products include low pro-
Corresponding Author:
duction volumes, limited manufacturing time, short dating Juan Peris-Vicente, Universitat Jaume I, QFA, ESTCE, UJI, Campus del
periods, requirement for large sample volumes, and the Riu Sec, Av/Sos Banyat s/n, Castelló, 12071, Spain.
need for manual and visual examination of cultures to detect Email: vicentej@uji.es
260 Journal of Laboratory Automation 20(3)

To apply these methods, the operator must prove that Biologics Evaluation and Research/FDA, from the U.S.
RMMs provide the same assurance of effectiveness of the bio- Department of Health and Human Services. FDA guidance
logical product as traditional methods do. A validation guide exposes a recommendation to test the suitability of the vali-
should be followed to indicate the protocol is needed to assess dation of the RMMs, but it has not law level.3
the usefulness of the method. Several guides have been sug- This guide proposes recommendations for manufactur-
gested and can be applied to validation of RMMs, such as the ers and users for validation and sterility testing of therapy
Guides for Industry: Validation of Growth-Based Rapid and gene therapy products, as well as growth-based RMMs.
Microbiological Methods for Sterility Testing of Cellular and However, the guideline is not directly applicable to human
Gene Therapy Products, described by the Food and Drug cells, tissues, or cellular and tissue products.3
Administration (FDA)3; Guidance for Industry PAT—A The guidance applies to the following products.
Framework for Innovative Pharmaceutical Development,
Manufacturing, and Quality Assurance, September 20046; and 1. Somatic cell therapy products: Autologous or allo-
Presidential Task Force on Best Practices in Microbiological geneic cells that have been propagated, expanded,
Methodology performed by AOAC International, 2006.7 These selected, pharmacologically treated, or otherwise
guides require detailed information on all of the validation altered in biological characteristics ex vivo, to be
parameters: accuracy, precision, ruggedness, specificity, limit administered to humans and applicable to the pre-
of quantification (LOQ), limit of detection (LOD), linearity, vention, treatment, cure, diagnosis, or mitigation of
range, equivalence, and quality by design. An analytical meth- disease or injuries.8
odology is accepted if the validation parameters are under 2. Gene therapy products: All products that mediate
those guideline requirements. their effects by transcription and/or translation of
Although RMMs are now routinely used in limited set- transferred genetic material and/or by integrating
tings, such as clinical testing, they have not been compre- into the host genome and that are administered as
hensively validated for use in a variety of manufacturing nucleic acids, viruses, or genetically engineered
settings. Cell-based products present especially challenging microorganisms. The products may be used to mod-
manufacturing issues, as these products often introduce ify cells in vivo or transferred to cells ex vivo prior
additional product or process variables that would likely to administration to the recipient.9
affect the test outcome. These variables should be consid-
ered in a validation study to demonstrate that the method is Guidance for Industry PAT—A Framework for Innovative
suitable for its intended product and application. Pharmaceutical Development, Manufacturing, and Quality
The aim of this work is to discuss the two main guides Assurance was developed by the U.S. Department of Health
proposed to validate RMMs: the Guidance for Industry: and Human Services, FDA, in September 2004. The guide
Validation of Growth-Based RMMs for Sterility Testing and is intended to describe a regulatory framework (process
Gene Therapy Products and the Guidance for Industry analytical technology [PAT]) that will encourage industry
PAT—A Framework for Innovative Pharmaceutical and research laboratories to develop and implement innova-
Development, Manufacturing, and Quality Assurance focus- tive pharmaceutical development, manufacturing, and qual-
ing on the description of the applied validation protocol. The ity assurance. Working with existing regulations, the agency
ability to recover and detect organisms from the product and has developed an innovative approach for helping the phar-
demonstrate their viability by multiplication in liquid media maceutical industry address anticipated technical and regu-
must be evaluated. The guidance has been developed for latory issues and questions.6
RMMs with qualitative results (e.g., detection of microor- The Presidential Task Force on Best Practices in
ganisms). Reliance on validated sterility testing methods is a Microbiological Methodology was performed by AOAC
critical element in assuring the safety of a product. Validation International in September 2006.7 This guide is devoted to
of most methods for final product testing, including those the validation microbiological methods for the industry and
assessing sterility, is required under 21 CFR 211.165(e) and refers to laboratories working in the field of food safety,
21 CFR 11.194(a).4 Validation of critical methods proves that quality assurance, clinical diagnostics, veterinary diagnos-
the methods are suitable for their intended purpose, to mea- tics, and engineering.7
sure the quantity of microorganisms in the sample, and pro-
vides assurance about the accuracy and reproducibility.
Validation Procedure
The final objective of validation of an analytical method is
Description of the Guides to demonstrate that it is suitable for its intended purpose.
Guidance for Industry:Validation of Growth-Based Rapid Thus, RMMs must be compared with traditional methods to
Microbiological Methods for Sterility Testing and Gene prove that they are equivalent or even that they provide less
Therapy Products has been developed by the Center for uncertainty.10
Peris-Vicente et al. 261

Contamination Studies •• Specificity: The ability of the test method to detect a


panel of organisms within the sample matrix estab-
During the production of cell-based products and microor- lished in the validation protocol. Specificity is demon-
ganisms, several mistakes, such as incorrectly controlled strated by challenging the RMMs to detect variations in
process streams and failures in aseptic processing tech- microbial growth characteristics and concentrations.
niques, can cause the introduction of microorganisms. Thus, Challenge testing is performed with organisms, includ-
a serious assessment of the production must be performed ing those recovered in the manufacturing environment
to identify and, eventually, avoid potential routes of micro- and from sterility test positives, as well as common
bial contamination8. technical and operator contaminants. Known quantities
The main risk of contamination comes from the follow- of organisms inoculated into product samples need to
ing: source material, level manipulation, processing path, be detected within the detection limit (see LOD), recov-
cell culture, incubation duration, and cell culture vessel.11 ered, and identity confirmed.
•• Ruggedness: Degree of reproducibility of results
obtained by analysis of the same sample under a variety
Validation Parameters
of normal test conditions, such as different analysts, dif-
The validation parameters that must be tested during valida- ferent instruments, and different reagent lots.
tion are described below.10,12 Ruggedness is the lack of influence on the test results of
operational and environmental variables of the micro-
•• Limit of detection (LOD): The lowest number of biological method and should be assessed by being pre-
microorganisms detectable in the sample matrix. pared by different analysts, as specified in the
LOD is essential to define what is considered to be procedures. Positive and negative controls should be
contaminated. The LOD is determined by inoculat- included and should test true to their characteristics.
ing samples with serial dilutions of viable contami- Ruggedness is evaluated by tabulating the mean time to
nants, with colony-forming units (CFUs) confirmed detection for each inoculate and determining the differ-
at inoculation by plate counts. Challenge testing is ence between the means for each analyst.
performed with each challenge microorganism using •• Robustness: Capacity of a method to remain unaf-
an amount between 10 and 100 CFU. Any growth is fected by slight, but deliberate, variations in method
recovered and identified to confirm identity. parameters.
•• Incubation time robustness: Time range required for
detection of microorganisms at a determinate level.
•• Accuracy: Closeness between measured value and
Selection of Microorganisms for Validation
true value of microorganism amount. The RMMs must be tested using the microorganism more rel-
•• Precision: Agreement among individual test results evant to the product and manufacturing method. The guide
for multiple samplings from a homogeneous sample. suggests the inclusion of microorganisms belonging to several
Repeatability refers to using the test procedure categories: gram-negative bacteria, gram-positive bacteria,
within a laboratory over a short time period. aerobic bacteria, anaerobic bacteria, yeast, fungi, isolates
•• Linearity: The linearity is the ability (within a given detected in starting materials, isolates detected by in-process
range) to obtain a detection signal that is directly testing or during preliminary product testing, isolates detected
proportional to the CFU of a microorganism in a by environmental monitoring of your manufacturing facility,
sample, with suitable accuracy and precision. The isolates from production areas that represent low-nutrient and
evaluation of linearity is assessed by the determina- high-stress environments, and microorganisms from commer-
tion coefficient (r2), which should be greater than cial sources that have continually been exposed to high-nutri-
0.95 to consider that linearity is adequate. ent growth media and slow-growing bacteria.3
•• Limit of quantification (LOQ): The lowest CFU of The same microorganism can show strong differences in
microorganisms quantifiable in cell culture with ade- growth-rate kinetics, depending on the source, significantly
quate accuracy and precision. affecting the needed incubation time and the temperature to
•• Calibration range: Minimal and maximal amount of detect growth. Thus, the validation should include the study
CFU, which could be reliably quantified. of growth under aerobic and anaerobic conditions, espe-
•• Recovery: Ratio between the measured CFU and the cially for products manufactured in closed systems.3
CFU really incubated in the cell culture. The accep-
tance criterion is more than 70%.
•• Viability recovery: Ratio between the CFU count
Quality by Design of Method Validation
after reincubation and traditional method count. The The composition of the test sample should be representative of
acceptance criterion is more than 70%. the product samples that RMMs would test. The parameters to
262 Journal of Laboratory Automation 20(3)

consider are cell concentration, media, and additives, as well as (e.g., worst case, matrix approach). Revalidation of the
preservation and antimicrobial agents, with a bacteriostatic or RMM should be performed whenever there are changes in
fungistatic effect. Several matrix compositions should be the process that could potentially inhibit (e.g., the addition
tested to validate the method for multiple sample types of the of antimicrobials) or enhance detection of viable microor-
same cellular product. ganisms. Verification of critical parameters of the test
The application of RMMs must be validated for in-pro- method postprocess, or product changes using the microor-
cess and final product in a cell culture production. The cell ganisms most difficult to detect, can serve as an indication
culture medium can contain antibiotics in different stages of of the need to revalidate the method. Validation of the steril-
the in-process, which might contain a preservation agent as ity test should be performed on all new products.3
well as a higher cell concentration. Thus, the different steps
of cell production must be separately validated because of
their different compositions.
Advantages of RMMs
To validate the performance of a commercially available The implementation of RMMs also provides interesting
testing system, studies using characteristic organisms under economic features, because of the significant reductions in
prescribed assay conditions should be performed, and the time to result over conventional methods. Normally, the
results should be compared with those provided by the production cycle is stopped or a product is kept until
supplier. checking the absence of contamination. If the time to
In the validation study design, the potential for the materi- result is shortened, the product would be more rapidly
als being tested to generate false-positive or false-negative released. Moreover, RMMs allow continuous sampling
results should be evaluated using the appropriate controls. and data collection and provide time-sensitive outputs and
This will depend on the product matrix, additives and preser- real-time data analysis. Therefore, more screenings and
vatives, and unique characteristics of the product. For exam- repeat tests could be performed in all stages of production,
ple, if a freshly isolated cellular product is tested for sterility improving the surveillance process.13 Real-time control
using a detection method based on CO2 production, then the and early warning of contamination are essential before
same freshly isolated cellular product should serve as a con- introducing raw materials and other product components
trol to determine whether uncontaminated cells generate lev- to the process, validating manufacturing, or distributing
els of CO2 that would produce a false-positive result. The use finished products.14
of additional positive and negative controls containing prod- RMMs involves an easy-to-handle protocol, with a neg-
uct components, but not cells, is also recommended.3 ligible production of waste, minimal operator variability,
reduced staff training requirement, and possibility of auto-
mation. They also facilitate research through generating
Comparison between RMMs and Traditional earlier results. Moreover, the shorter analysis time reduces
Methods the probability of contamination, improving the robustness.
To compare RMMs and traditional methods, the same amount A shorter time to result facilitates replication of an analysis
of potential microbial contaminants as microorganisms indi- if a contamination or a strange result occurs. Thus, RMMs
cated in the previous section are inoculated in cell cultures can improve manufacturing consistency by permitting
(measured in CFU/volume). The ability of both methods to faster implementation of corrective actions and fostering
identify the microorganisms is tested, together with recovery, opportunities to improve the safety of the process. Moreover,
detection limit, and assay performance. Several dilutions of RMMs also show improved sensitivity, efficiency, selectiv-
microorganisms are tested (10–99 CFU/sample), in order to ity, accuracy, high throughput, and low false-positive rate,
evaluate the usefulness of RMMs at the main working con- leading to higher efficiency.13 These features of RMMs pro-
centration levels of microorganisms.3 vide an opportunity to assess the significance of viable but
nonculturable or stressed microorganisms.15,16
Revalidation Method
Any changes in product manufacturing, including formula- Application of Validation of RMMs
tion, or changes in the RMMs that can potentially inhibit or The Milliflex Quantum method is an RMM based in fluo-
enhance detection of viable microorganisms need to revali- rescence detection for the quantification of microorgan-
date the RMMs. Changes in cell types, harvesting proce- isms. The culture media are prefilled tryptic soy agar plates,
dures, culture media, additives, critical processing steps, or R2A plates, and Sabouraud dextrose agarplates. The authors
postprocessing handling can potentially affect the detection have validated this methodology for the analysis of contami-
of viable microorganisms. Initial validation can be designed nants in water. The tested microorganisms were the following
to minimize the effect of some changes by designing assay American Type Culture Collection (ATCC) strains (TM):
conditions that encompass known or proposed changes Candida albicans (10231D-5), Aspergillus brasiliensis
Peris-Vicente et al. 263

(16404D-2), Staphylococcus epidermidis (12228D-5), work has been supported by a project from the University Jaume I
Ralstonia pickettii (27511), Brevundimonas diminuta (19146), (project P1.1B2012-36).
Staphylococcus aureus (700699D-5), Pseudomonas aerugi-
nosa (47085D-5), Bacillus subtilis (23857D-5), and Escherichia References
coli (10798D-5). The experimental procedure was described 1. Food and Drug Administration. Guidance for Industry:
Meder et al.12 This method was compared with a compendial Sterile Drug Products Produced by Aseptic Processing—
method. Current Good Manufacturing Practice (September 2004).
http://www.fda.gov/cber/guidelines.html (accessed May 14,
•• Incubation time robustness was tested at 10 CFU and 2014).
was found to be between 8 and 28 h, depending on 2. Verdonk, G. P. H. T.; Willemse, M. J.; Hoefs, S. G. G.;
et al. The Most Probable Limit of Detection (MPL) for Rapid
the microorganism.
Microbiological Methods. J. Microbiol. Meth. 2010, 82,
•• Ruggedness was evaluated using different media
193–197.
lots, membrane lots, and reagent lots and treating the 3. Food and Drug Administration. Guidance for Industry: Validation
results by analysis of variance. No significant differ- of Growth-Based Rapid Microbiological Methods for Sterility
ences were detected in fluorescence or viability Testing of Cellular and Gene Therapy Products (February 2008).
recovery. http://www.fda.gov/downloads/BiologicsBloodVaccines/
•• Fluorescence recovery was 98% to 102%, and vali- GuidanceComplianceRegulatoryInformation/Guidances/
dation recovery was 97% to 102%. Thus, no signifi- CellularandGeneTherapy/ucm078696.pdf (accessed May 14,
cant differences were found between the Milliflex 2014).
Quantum and traditional method. 4. United States Pharmacopeia 28 and National Formulary 23
•• Accuracy was 90% to 112%. (USP 28 NF 23), Sterility Tests, General Chapter 71, 2005;
pp 1–13.
•• Linearity was found at r2 > 0.96 between 4 to 10
5. Miller, M. J. Case Study of a New Growth-Based Rapid
CFU and 100 CFU.
Microbiological Method (RMM) That Detects the Presence
•• The LOD was 1 CFU. of Specific Organisms and Provides an Estimation of Viable
•• Specificity: the challenged microorganisms were Cell Count. Am. Pharmaceut. Rev. 2012, 15(2).
correctly identified. 6. Food and Drug Administration. Guidance for Industry
PAT—A Framework for Innovative Pharmaceutical
The validated RMM and the compendial method were Development, Manufacturing, and Quality Assurance
applied to the detection of microorganisms in purified water (September 2004). http://www.fda.gov/downloads/Drugs/
sampled in pharmaceutical plants. Milliflex Quantum pro- Guidances/ucm070305.pdf (accessed May 14, 2014).
vides results in 24 to 40 h, whereas the compendial method 7. Food and Drug Administration; AOAC International. Final
takes 5 to 7 d. The obtained results were similar in both Report and Executive Summaries, Presidential Task Force
on Best Practices in Microbiological Methodology (August
cases.12
2006). http://www.fda.gov/Food/FoodScienceResearch/Labora
RMMs offer evident improvements in comparison with
toryMethods/ucm124900.htm (accessed May 14, 2014).
traditional methods, and they especially provided results in 8. Food and Drug Administration. Application of Current
a shorter time. This review outlines the basic principles of Statutory Authorities to Human Somatic Cell Therapy Products
validation and verification in an implementation process for and Gene Therapy Products (58 FR 53248) (October 1993).
microbiological methods. The main validation parameters http://www.fda.gov/downloads/BiologicsBloodVaccines/
have been described. Thus, the implementation of these SafetyAvailability/UCM148113.pdf (accessed May 14,
methodologies is encouraged. According to the literature, 2014).
RMMs provide significant improvements with the same 9. Food and Drug Administration. Guidance for Industry:
results as traditional methods. This encourages their imple- Gene Therapy Clinical Trials—Observing Participants for
mentation in laboratories carrying out microbiological stud- Delayed Adverse Events (November 2006). http://www
.fda.gov/BiologicsBloodVaccines/GuidanceCompliance
ies for clinical or food safety purposes.
RegulatoryInformation/Guidances/CellularandGeneTherapy/
ucm072957.htm (accessed May 14, 2014).
10. ICH Expert Working Group. The International Conference
Declaration of Conflicting Interests
on Harmonisation (ICH) Guidance; Q2(R1), Validation of
The authors declared no potential conflicts of interest with respect Analytical Procedures: Text and Methodology (November
to the research, authorship, and/or publication of this article. 2005). http://www.ich.org/fileadmin/Public_Web_Site/ICH_
Products/Guidelines/Quality/Q2_R1/Step4/Q2_R1__
Guideline.pdf (accessed May 14, 2014).
Funding 11. ICH Expert Working Group. The International Conference
The authors disclosed receipt of the following financial support on Harmonisation (ICH) Guidance; Q9, Quality Risk
for the research, authorship, and/or publication of this article: This Management (June 2006). http://www.ich.org/fileadmin/
264 Journal of Laboratory Automation 20(3)

Public_Web_Site/ICH_Products/Guidelines/Quality/Q9/ 14. Cundell, A. M. Opportunities for Rapid Microbial Methods.


Step4/Q9_Guideline.pdf (accessed May 14, 2014). Eur. Pharm. Rev. 2006, 1, 64–70.
12. Meder, H.; Baumstummler, A.; Chollet, R.; et al. Fluorescence- 15. Newby, P. The Significance and Detection of VBNC
Based Rapid Detection of Microbiological Contaminants in Microorganisms. Eur. Pharm. Rev. 2007, 3, 87–92.
Water Samples. Sci. World J. 2012, 234858. 16. Sandle, T. A Review of Cleanroom Microflora: Types,
13. Miller, M. J., , ed. Encyclopedia of Rapid Microbiological Trends, and Patterns. PDA J. Pharm. Sci. Technol. 2011, 65,
Methods; DHI Publishing, LLC: River Grove, IL; 2005. 392–403.

You might also like