Usp 1042
Usp 1042
Usp 1042
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
1
This general chapter is intended to provide users with practical guidelines regarding the derivation, production, storage,
characterization, and qualification of bacterial and mammalian cell banks used in the manufacture of biological drug products.
The chapter is primarily focused on the characterization of cell banks used to produce biological products that are subject to
approval by the US FDA. Cell banking characterization and qualification should be based on sound scientific principles and
current regulatory guidance. The relevant requirements and guidances for generating cell banks for biotherapeutics seeking
FDA approval include the US Code of Federal Regulations (21 CFR Part 610.18) (1), FDA Points to Consider documents (2–4),
FDA Guidance for Industry documents, and International Council for Harmonisation (ICH) guidelines (5–8). These guidances
state that the final biological product and production should be consistent, uniform, and free from cellular and adventitious
contaminants. [NOTE—It is recognized that there are also relevant guidances from other international organizations such as the
World Health Organization (WHO) and European Medicines Agency (EMA); however, this chapter focuses on US FDA, USP, and
ICH regulations and guidances.]
Therefore, the intent of this chapter is to elaborate on principles noted in FDA and ICH guidances and in USP general chapters
for the establishment and characterization of mammalian and bacterial cell banks used in the manufacture of biological
products. This chapter represents current best practices for the production, characterization, and maintenance of controlled
and consistent cell sources using risk-based and product-phase-appropriate quality considerations.
al
The evaluation of the risk to quality (9) should be based on scientific knowledge and ultimately link to the protection of the
patient; the level of effort, formality, and documentation of the quality risk management process should be commensurate with
the level of risk. Regarding good cell line banking practice, risk analysis1 should be conducted on the basis of existing experience
with the type of cell line and should be updated throughout the cell line’s product life cycle as further knowledge is gained to
characterize the associated risks. The risk criteria for good cell line banking practices should include such quality risk factors as:
• Cell line history and origin
ci
• Cell line growth and characteristics (relevant phenotypic and genotypic markers)
• Cell count
• Viability
ffi
• Authentication
• Absence of adventitious agents (e.g., microbial, fungal, viral, and mycoplasma contaminants)
• Genetic drift (e.g., transgene stability)
• Loss of cell line characteristics of interest (such as the host cell transformations, surface antigens, or monoclonal antibody
expression)
• Process validation and capability
O
• In-process controls
• Change controls
• Cross contamination
• Testing
• Stability
• Storage
• Product and/or process trending that demonstrates the original cell line characteristics and resultant quality characteristics
of the final product that are retained throughout the cell line and product’s life cycle
Over time and experience—through feedback from the quality systems, trending, post-marketing surveillance, and
pharmacovigilance systems—the risk assessment should continue to evolve to minimize the risks associated with the cell line’s
performance during the product’s life cycle.1
1.1 Scope
Cell banks are the starting materials for manufacturing biological drug substances and therefore have a direct impact on the
characteristics and safety of the resultant biological products. Cell banks preserve characteristics in a contamination-free process
and ensure consistency of the quality attributes of the final drug substance and drug product (DP). This chapter covers cell
banking for biotechnology products for human use that are derived from cell lines of mammalian and microbial origin. In many
instances, biological product development involves developing new recombinant prokaryote [e.g., Escherichia coli (E. coli)],
eukaryote [e.g., Chinese hamster ovary (CHO)] or transgenic cell lines, and requirements may differ depending on the strain
or cell line. Each new strain/cell line needs to be evaluated with respect to its own particular cellular characteristics and its
associated master cell banks (MCBs) and working cell banks (WCBs) need to be manufactured under conditions consistent with
good manufacturing practices (GMPs). Each GMP cell bank should be tested and characterized to satisfy quality standards for
identity, purity, and genetic stability and ensured to be free from adventitious contaminants for use in drug substances for
commercial applications. If possible, storage stability should also be performed. A risk-based approach should be applied for
1 See ICH and appropriate FDA guidelines for more specific information.
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 1/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
2
manufacturing. The bank can be conditionally released at risk before it is fully tested to start drug substance manufacturing;
however, the bank has to be fully tested, especially for safety prior to a drug substance’s release for clinical applications.
This chapter does not cover cell banking for antibiotics, viral seed banks, viruses that express vaccine antigens, bacterial seed
banks, avian or insect cells, or cell banks for cell and gene therapies. For specific information on cell banking for vaccines, see
Vaccines for Human Use—Bacterial Vaccines á1238ñ and Vaccines for Human Use—General Considerations á1235ñ.
This chapter primarily focuses on cell banks derived from recombinant E. coli strains and CHO cell lines as they are the most
common substrates for production of therapeutic proteins and centers on the cell line or strain history, GMP manufacturing
and testing, and qualification of cell banks. Most, if not all, of the techniques and practices discussed are also applicable to other
cell strains besides E. coli and CHO cell substrates. Based on the organism, suitable approaches can be selected to cover the cell
line/strain history, GMP manufacturing and testing, as well as qualification of cell banks.
Cell banks used for manufacturing biological drug substances are usually prepared from research cell banks (RCBs) and use a
two-tiered bank system, MCB and WCB or working seed lot. The MCB is generated as the source for the WCB, which is derived
by expanding 1 or more vials of the MCB. Thus, the MCB is considered the starting raw material for the GMP manufacturing
of drug substances. RCBs are usually generated under non-GMP conditions, whereas MCBs and WCBs are required to be
produced in GMP facilities. Documentation of suitability of the RCB for GMP manufacture is needed. Figure 1 outlines the general
practices and considerations of cell banking processes and how they evolve through development into GMP manufacturing.
The critical steps shown in the figure are applicable to recombinant and non-recombinant cell lines. More detailed best practices
and recommendations are described in the sections that follow. For more detailed recommendations about preparation and
preservation of cell banks, see ICH Q5D (7).
al
ci
ffi
O
The generation of an RCB starts with choosing a suitable expression system that consists of host cells and expression
constructs carrying the genes of interest (GOI).
Any facility including a research laboratory should be regarded as a potential source of contamination by viruses, bacteria,
fungi, and mycoplasma, so the introduction of a new cell line should be handled under quarantine conditions until proven
negative for such contaminants. The history, identity, and purity, as well as associated testing of the host cells, should be well
documented (10).
An expression construct consists of the GOI and an expression vector carrying the genetic elements such as a promoter,
signal sequences, selection markers, and other elements needed for the stable expression of the GOI in the host cells. Detailed
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 2/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
3
descriptions of the GOI and those genetic elements carried by the vectors, as well as the steps in the assembly of the expression
construct, should be documented.
The next step for RCB is the generation of the candidate production cell lines/strains. It is critical to document the details of
the cell-line generation history, including:
• Description of the transfection/transformation of the GOI
• Screening and selection of the production cell lines/strains
• Demonstration of clonal derivation
• Information about the raw materials used for the generation of candidate production cell lines
The source, origin, generation, propagation, and selection of the cell line must be documented and described in detail.
Multiple RCBs may be generated in relatively small quantities during the screening/selection processes. Only the final, chosen
production cell lines/strains and backup cell lines/strains are made in larger quantities. The RCBs chosen for the production cell
lines/strains and backup cell lines/strains are sometimes referred to as premaster research cell banks (pmRCBs), which need to
go through purity testing prior to the production of MCBs and subsequent WCBs.
Cell substrate for manufacturing biological drug substances, MCBs, and WCBs must be produced in compliance with current
GMPs (cGMPs). As illustrated in Figure 2, MCBs are often used for manufacturing drug substances for early clinical studies and
as the source of the WCBs, which may not be available during the early phase of the development life cycle. Drug substances
used for late-phase development and commercial applications are usually manufactured from WCBs. To ensure a consistent
supply, it is generally recommended that additional replacement WCBs be produced as needed once a biologic is approved for
marketing. Section 5. Cell Bank Characterization describes in detail cell bank characterization, testing and qualifications, as well
as general considerations on raw materials, cell bank cryopreservation, storage and stability, and transportation.
al
ci
ffi
O
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 3/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
4
al
proteins for human use and since then have remained the most common cell line for the production of therapeutic glycoproteins
for the following reasons:
1. CHO cells pose a lower adventitious virus safety risk as only a few human pathogenic viruses are able to propagate in
CHO cells (11).
2. CHO cells grow well in animal-component-free, chemically defined media, which further minimizes adventitious virus
safety concerns.
ci
3. As immortal cells, CHO cells can be propagated for a long time in vitro. This is an important characteristic for commercial
production of recombinant proteins because it takes many cell passages to progress from cell line generation through
cell banking to the final, large-scale commercial production process. A non-immortalized cell line cannot be propagated
long enough to generate a profitable and sustained commercial production process.
ffi
4. CHO cells are able to adapt and grow in suspension culture, which is ideal for large-scale manufacturing of biologics.
5. CHO cells have the capacity for efficient, post-translational modifications, such as glycosylation, which enable the
CHO-derived therapeutic proteins to be both compatible with and bioactive in humans.
6. CHO cells are amenable to genetic manipulation, which allows for easy introduction of recombinant DNA resulting in
expression/secretion of large quantities of the desired protein.
O
See Table 1 for a comparison of two common cell substrates for recombinant therapeutic products.
Table 1. Comparison of Two Common Cell Substrates for Recombinant Therapeutic Products
Cell Substrate Mammalian Cell Lines (e.g., CHO Cell Lines) Microbial Strains (e.g., E. coli Strains)
• Recombinant proteins are soluble and are secreted in cell • Fast growing
culture medium • High protein yield/low cost
• Secreted recombinant proteins are properly folded and are • Easy transformation and fermentation
Advantages identical in higher order structure to the natural human • Do not harbor mammalian viruses
protein
• Simpler cell bank characterization
• Conformational and post-translational modifications in
these proteins are similar to the types found in humans
Because CHO cells are immortalized, they can pose a significant challenge, though, for the cell-line development process
with respect to finding genetically stable cell lines needed for consistent commercial manufacturing processes. CHO cells have
varied ploidy, and a culture of any CHO cell line, with or without any transgenes, consists of a population of cells that exhibit
both genetic and phenotypic diversity and instability under continuous cultivation. Random integration of recombinant genes
into the CHO genome adds new genetic variability to the CHO cell population. After transfection, implementation of a single-cell
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 4/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
5
cloning step helps to minimize cell population heterogeneity by removing genetic variations that either existed prior to or were
introduced during transfection.
All CHO cell substrates used for commercial recombinant protein production are expected to be single-cell derived. However,
incorporation of the single-cell cloning step alone cannot prevent genetic and phenotypic heterogeneity within the population
of cells that arises post-cloning, as these events are inherent to the use of immortalized mammalian cell lines (12). Thus,
identifying an appropriate production cell line ultimately depends on the genotypic and phenotypic characterization of potential
cell lines post-cloning at the limit of in vitro cell growth and the evaluation of the expressed product.
Common CHO cell lines: Some commonly used CHO cell lines include: CHO-DXB11, CHO-DG44, CHO-K1, and CHO-K1SV.
These cell lines are transfected with expression plasmids carrying the GOI and corresponding selection markers specific to the
CHO host cells. Their characteristics are briefly summarized below.
CHO-DXB11 and CHO-DG44: CHO-DXB11 (also known as CHO-DUK-XB11 or CHO-DUKX) and CHO-DG44 are dihydrofolate
reductase (DHFR)-deficient mutants established in the early 1980s. CHO-DXB11 cells carry a deletion of one locus for DHFR
and a missense mutation (T137R) of the second DHFR locus, rendering the cells incapable of reducing folate, a precursor for
thymidine and hypoxanthine synthesis; CHO-DG44 cells carry the full deletion of both DHFR loci.
The DHFR-deficient cells are triple auxotrophs for glycine, hypoxanthine (a proline derivative), and thymidine. Introduction
of the GOI into CHO cells can be accomplished by cotransfection with a functional DHFR gene, which obviates the need for
these nutrients. Recombinant cell lines carrying the integrated GOI are selected in media devoid of those three nutrients. The
GOI expression level can be further enhanced by gene amplification with methotrexate (MTX), an antagonist of DHFR. Selection
of recombinant cell lines can be performed using stepwise increases in the media concentration of MTX, which results in
amplification of the transfected DHFR gene together with the GOI, possibly increasing the productivity of the GOI.
Sometimes MTX-induced gene amplification can result in large genomic reorganizations. New chromosomal structures,
known as homogeneously staining regions (HSRs), can be found in the metaphase chromosome of MTX-selected CHO cells.
These regions show multiple repetitions—up to thousands of smaller chromosomal regions (amplicons)—all containing DNA
al
encoding at least in part sequences with DHFR activity as well as hundreds and even up to thousands of copies of the GOI.
Gene amplification and high-level expression of a transgene product can challenge the cells with potential toxicity and metabolic
burden. Additional DNA rearrangements within the GOI region may also occur during long-term subculturing in the absence
of MTX, which may eventually lead to instability of the recombinant protein production. Therefore, genetic stability evaluation
of the chosen CHO production cell line is important to ensure that the desired characteristics for the commercial production
are acquired.
ci
CHO-K1 and CHO-K1SV: CHO-K1 was derived as a sub-clone of a parental CHO cell line lacking the proline synthesis gene.
CHO-K1 was adapted to grow in suspension culture in protein-free medium, resulting in the cell line CHO-K1SV. Introduction
of a GOI into the CHO-K1SV cells can be accomplished by cotransfection of the GOI with a functional glutamine synthetase
(GS) gene followed by selection in the absence of glutamine. In addition, application of a GS inhibitor [methionine sulfoximine
ffi
(MSX)] allows an increase in the stringency of selection. Although the copy number of the GOI and GS is significantly lower in
the GS-based selection system compared to that seen in the DHFR-based system, instability of the GS-based CHO cell lines is
still often observed during long-term cultivation. Therefore, it is important to evaluate genetic stability of the chosen CHO
production cell line over the proposed in vitro cell age needed for commercial manufacturing.
Expression vectors for CHO cell substrate: Expression vectors used for CHO-based cell substrates consist of multiple
components that contribute to the successful expression of the therapeutic proteins. These components include:
O
• The GOI and associated transcriptional and translational genetic elements (e.g., promoters, enhancers, Kozak translation
initiation sequences, leader sequences, and polyadenylation signals)
• Selection markers such as the commonly used DHFR or GS for recombinant cell line selection
• Selection markers such as the β-lactamase gene (bla) and replicons such as ColE1 for propagation of the plasmid in E. coli
Expression plasmids are often linearized prior to transfection and are generally integrated into the CHO host cell genome by
random integration. Resulting transfectants are a mixture of CHO host cells carrying variable copy numbers of expression
plasmids randomly inserted in single or multiple loci in the host genome. A single-cell cloning process enables the clonal
derivation of future production cell lines. Significant effort is spent on screening the clonally derived transfectants to identify
candidates for final production cell lines that possess high productivity. Furthermore, the identified cell lines must be capable
of maintaining the integrity and stability of the expression construct integrated into the host genome in order to be used for
large-scale manufacturing processes.
To reduce the heterogeneity associated with traditional random integration approaches, new expression systems have been
developed to facilitate targeted integration of the GOI into a specific desirable locus within the CHO genome. Targeted
integration systems described in the literature include site-specific recombinases, integrases (ØC31), transposases (piggyBac),
zinc finger nucleases, transcription activator-like effector nucleases (TALENs), and CRISPR-Cas RNA-guided nucleases. Detailed
descriptions of the genetic components carried by the expression plasmids, the specific integration method, and identification
of the preferred production cell line should be included in the history of the final production cell line.
CHO host cell engineering: In addition to expressing the GOI, recombinant CHO cell lines can be engineered to further
enhance productivity and quality of the recombinant product as well as the robustness of the cell culture performance. Host
engineering includes overexpression or knock-out of specific genes that may impact the desired recombinant product
characteristics and thereby the performance of the production cell line. Examples of overexpressed genes are those involved
in:
• Glycosylation [e.g., α-2,6-sialyltransferase (ST6Gal)]
• Cellular metabolism [e.g., vitreoscilla hemoglobin (VHb)]
• Anti-apoptotic pathways [e.g., B-cell lymphoma 2 (BCL2)]
• Pro-proliferative pathways [e.g., valosin-containing protein (VCP)], as well as genes that may enhance cellular protein
biosynthesis machinery and secretion [e.g., transcription factor ATF4, heat shock 70 kDa protein 5, binding
immunoglobulin protein (BiP), etc.]
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 5/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
6
Alternatively, there are some host cell proteins that can decrease the quality or yield of recombinant proteins [e.g., α-
1,6-fucosyltransferase (FUT8) and peptidylglycine α-amidating monooxygenase]. In these cases, genomic knockouts of the host
cell genes can yield a more homogeneous product.
Although the overexpressed or knocked out genes do not encode sequences for the final therapeutic protein, they play critical
roles in the productivity and quality of the expressed therapeutic proteins. Therefore, evaluation of the integrity and stability
of the overexpressed genes for the intended purposes should be included as a part of the cell substrate genetic stability
characterization. In addition, knocked out genes should be characterized.
ESCHERICHIA COLI
General information: E. coli is one of the best-understood organisms in terms of genetics, physiology, and biochemistry.
The key advantages of the E. coli system include:
• The GOI can be easily inserted into an expression plasmid using recombinant DNA technology. The resulting recombinant
plasmid, which is usually small and simple, can be inserted into the E. coli host by transformation and maintained as an
extrachromosomal segment of the host DNA or a piece of the host genome after integration. Numerous E. coli expression
systems are commercially available, making this organism easy to test for production of a new GOI.
• E. coli is well-characterized genetically with versatile comprehensive engineering tools.
• E. coli strains are capable of rapid growth in inexpensive media in small shake flasks as well as in large-scale fermenters
used to produce therapeutics with high yield and quality. E. coli expression systems continue to evolve, and the advances
in biotechnology and bioengineering as well as high-density fermentation technology have further enabled E. coli systems
to produce a variety of complex proteins, including antibody fragments and Fc-fusion proteins.
However, E. coli strains also have limitations as cell substrates for therapeutics production, including their inability to perform
al
certain post-translational modifications. In addition, endotoxins need to be thoroughly removed during purification of the
recombinant protein produced by an E. coli host (also see Table 1).
Sometimes the recombinant protein itself can be an obstacle for production in E. coli systems because it can be toxic to the
host. In such cases, a well-designed but more sophisticated expression system may be required, for instance, an inducible
promoter to produce the recombinant protein at the end of cell expansions. In the following sections, the characteristics of
ci
some E. coli-based cell substrates consisting of E. coli host strains and vectors carrying the genes encoded for the therapeutic
proteins are briefly summarized.
E. coli host strains: The most commonly utilized strains are the K12 strain and its derivatives including E. coli RV308 and W3110,
which are all genetically modified so they cannot colonize a human host or produce toxins. These strains may qualify for an
exempt status according to the requirements of the National Institutes of Health (NIH) guidelines for research involving
ffi
recombinant DNA molecules.
The other commonly used E. coli hosts are the B strain and its derivatives, such as BL21, C41, and C43. Although these strains
do not qualify for exempt status and require full Institutional Biosafety Committee (IBC) review, they are still desirable hosts for
therapeutic protein production in the industry because they are able to express various types of proteins. Advantages of the B
strain and its derivatives are the low levels of acetate accumulation under high glucose concentrations, the specific protease
deficiencies, and the high outer membrane permeability.
O
Expression vectors for E. coli strains: The expression vectors often used are the result of multiple combinations of replicons,
promoters, selection markers, and other genetic elements that are essential or beneficial for the desired efficiency of
transcription, translation, and protein secretion, as well as the stability of the plasmid.
A replicon in a vector consists of one origin of replication together with its associated cis-acting control elements. It controls
the copy number of a plasmid replicated in an E. coli host. Commonly used replicons include:
• pMB1 and its derivative origins used in pUC-based vectors (500–700 copies/genome)
• pBR322-based vectors (10–20 copies/genome)
• p15A origin used in pACYC and pSC101, which have low copy numbers (about 5 copies/genome)
For large-scale therapeutic protein production, an optimal copy number should be identified because too few gene copies may
result in low levels of mRNA leading to low protein productivity while too many copies may impose metabolic burdens on cells.
In addition, high-copy-number plasmids have been shown to possess higher segregation instability, especially in the absence
of selective markers. This can lead to the overgrowth of plasmid-free cells, resulting in a significant loss of protein productivity.
Therefore, it is usually not desirable to use high-copy-number plasmids, and it is critical to monitor plasmid retention during
large-scale production. Testing to qualify the microbial cell bank includes testing to demonstrate purity and freedom from
phage contamination and to confirm strain identity. Additional testing includes viability, plasmid copy number, plasmid
retention, and restriction enzyme mapping.
Promoters control and regulate the efficiency of transcription. Commonly used promoters include:
• lac and its derivatives such as lac UV5, tat and trc, which are induced by isopropyl-β-D-thiogalactopyranoside (IPTG)
• Bacteriophage λ pL and/or pR promoters, which are induced by temperature shift via the regulation of the
temperature-sensitive cI857 repressor
• T7 promoter, specifically recognized by T7 bacteriophage RNA polymerase from T7 bacteriophage
• phoA, trp, and ara PBAD, which are nutritionally inducible promoters
Selection markers, usually antibiotic-resistant genes such as TetR, are included in the plasmid to determine the growth of
plasmid-free cells. Therefore, antibiotics are usually included in the growth media. Some common examples include neomycin,
polymyxin B, streptomycin, and gentamicin. However, the use of antibiotics in clinical manufacturing can lead to adverse events
in some patients and raises concerns over the development of drug-resistant pathogens, so it is desirable to develop
antibiotic-free plasmid systems, if possible.
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 6/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
7
In addition to the coding sequence encoding the protein, genetic elements in expression vectors that impact protein
expression include the sequence and structure of the translation initiation region (TIR) at the 5′ end of each mRNA and the
stability (half-life) of mRNA.
Another genetic element used in some expression vectors is the signal sequence fused to the N-terminus of the protein,
which leads the protein to the periplasmic space of the host or the cell culture medium. Without signal sequences, recombinant
proteins mainly accumulate in the bacterial cytoplasm. Due to the lack of glycosylation and disulfide-bond formation, many
human therapeutic proteins expressed in E. coli tend to aggregate and form inclusion bodies due to misfolding. Correct folding
of eukaryotic proteins containing multiple disulfide bonds is more likely to occur in the oxidative environment of the periplasm.
The secretory process allows removal of the N-terminus signal peptide and yield of the authentic N-terminus of the expressed
protein. Co-expression of periplasmic chaperones, fusion with secretory partners, and modification of the outer membrane
structure by host engineering may further promote release of periplasmic proteins.
Overall, E. coli expression systems have been preferred choices as cell substrates for the expression of non-glycosylated
therapeutic proteins due to their well-characterized genetics, rapid growth, and high-yield production capability. Both the host
strains and the genetic elements carried by the expression vectors should be well described in the strain development histories
and documented for the cGMP cell bank generation. Together with the therapeutic protein-coding sequence, the key genetic
elements that may potentially impact the drug substance quality and the consistency of drug substance production should be
evaluated during cell bank characterization and qualification.
Clonal selection of E. coli host strains: As with CHO-derived cell lines, it is expected that E. coli strains used for therapeutics
production be clonally derived. Microbial clonality is easily achieved and accepted by picking transformants from well-isolated
single colonies grown on solid media. Additional rounds (usually two) of single-colony isolation and phenotypic confirmation
may be performed to ensure the clonality.
Table 1 compares two commonly used mammalian cell and microbial strain substrates for generating cell banks for biological
products.
al
BACILLUS
Other useful bacterial systems are Gram-positive bacilli, including Bacillus megaterium, B. subtilis, B. licheniformis, and B.
brevis. Bacillus is very often considered an attractive alternative to E. coli for the following three reasons:
1. Bacillus strains usually secrete proteins
ci
2. Bacillus strains are suitable for high-density cultures in fermenters
3. Most Bacillus strains lack membrane proteases
In addition, the genomes of B. subtilis and B. licheniformis have been sequenced, and there is no production of harmful exotoxins
ffi
or endotoxins. With these bacterial systems, secretion of the desired proteins into the fermentation medium results in easy
downstream processing, thereby eliminating the need for cell disruption or chemical processing techniques. Thus, the recovery
is relatively efficient and cost effective. Host strains used for successful expression of recombinant proteins are often deleted for
the genes amyE, aprE, nprE, spoIIAC, and srfC and are easily transformed. These bacteria are often preferred for homologous
expression of enzymes such as proteases and amylases.
O
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 7/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
8
The steps in the GOI assembly from the original sources to the final expression plasmid should be described in detail and are
often summarized using a flow diagram. In addition, the source and function of all genetic components carried by the expression
plasmid should be provided. The components include:
• The GOI and associated transcriptional and translational genetic elements (promoters, enhancers, Kozak translation
initiation sequences, leader sequences, and polyadenylation signals)
• Selection markers such as DHFR or GS for recombinant cell line selection, or antibiotic resistance genes for recombinant
strain selection
• Selection markers such as the bla gene and the replicon such as ColE1 for propagation of the plasmid in E. coli
• Any other expressed proteins encoded by the plasmid
Furthermore, a well-annotated plasmid map clearly indicating the size of the plasmid, all genetic components described above,
and the restriction enzymes used for major cloning steps and for genetic stability characterization should be provided. The DNA
sequence of the whole construct/plasmid, or at least the sequences encoding the recombinant protein that have been confirmed
by DNA sequencing, should be included.
al
2. Any exposure of the parental cells to materials of human or animal origin such as serum, enzymes, hydrolysates, or other
living cells
3. Genetic and phenotypic characteristics of the parental cells that may enhance the evaluation of the safety and
characteristics of the subsequently derived cell banks
The lineage of the parental cells traced back to the original source or related literature references should be provided. If the
ci
parental cells undergo additional genetic modifications (e.g., targeted gene knockout or overexpression) that are aimed at
impacting the quality of the future recombinant proteins, then the methodology and characterization results should be
described in detail.
Characterization of the initial cell line should take risk into consideration. Use of low-risk cells (e.g., cells shown to be sterile
and phage- or mycoplasma-free) is recommended. Cell line strain ID can be assessed with polymerase chain reaction (PCR) or
using short tandem repeat (STR) profiling or DNA fingerprinting to verify the identity of the host cells. When higher-risk cells
ffi
(e.g., a mixture of cells with varying degrees of genetic instability) are used, a more thorough screening is necessary, unless the
supplier of the cells can provide documentation that minimizes the associated risk.
1. Transferring the expression plasmid to the parental cells and selecting recombinant strains/cell lines
2. Single-cell cloning to produce clonally derived recombinant strains/cell lines
3. Choosing the production strains/cell lines through screening/characterization
Recommendations for documentation to include in regulatory submissions are as follows:
• Details on transformation/transfection conditions
• Manipulations of the plasmid DNA prior to the transfer (e.g., linearization by restriction enzymes often applied prior to
transfection)
• Procedures/methods used for the selection of recombinant strains/cell lines
• Detailed description demonstrating the single-cell derivation of the production strain/cell line
○ For an E. coli strain, 2 or more rounds of single-colony streaking are commonly applied to ensure the clonal derivation
of the production strains
○ For CHO cell lines, numerous approaches can be used to achieve clonal derivation (see 4. Mammalian Cell Line
Clonality)
• Description of how the production strains/cell lines were screened and identified including the following three parts:
1. Detailed cell culture conditions
2. Selection criteria
3. All raw materials and reagents used throughout the generation process
It is helpful to provide a flow diagram (see Figure 3) of the production strain/cell line generation process, which includes the
above three parts and ends with the MCB and WCB production.
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 8/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
9
al
ci
ffi
O
al
Post-banking controls after thawing include measuring cell number and viability. Because these parameters vary between
individual cell banks, appropriate post-banking quality control for viability can be established and justified for each bank.
After the production of an MCB or WCB, each bank must be tested against specific acceptance criteria before that specific
cell bank can be released for use. Mammalian cell banks should be stored in multiple geographical locations. Figure 4 shows an
example of a cell process for a mammalian (CHO) cell bank. ci
ffi
O
Similarly, for bacterial cell lines used for therapeutic protein manufacturing, the chosen clone is subjected to growth
propagation experiments to ensure that the best growth conditions are chosen for this specific clone. The growth conditions
for an E. coli clone can be evaluated using shake-flask culturing. The optimal cell density is established by following growth
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 10/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
11
curves in order to harvest the cells at an exponential growth. The cells are harvested in vials containing sterile 20%–30% glycerol
under sterile conditions. E. coli cells harvested at a living cell density of 106 to 1010 are adequate for production; however, this
depends on the specific strains and selection pressure chosen.
All viability tests should be done with care, keeping in mind the actual selection system as well as how the harvest of any
produced protein will be performed (e.g., from inclusion bodies). This is not a concern for the frozen E. coli cells after thoroughly
mixing with glycerol; however, without adequate knowledge of the optimal growth point for harvesting and freezing as well
as the actual needed cell density, the viability and expected growth rate can create problems at the production site.
The optimization process shall be described and followed in detail each time a new growth of the strain is necessary. From
these optimized conditions, the first cell bank will be produced, which can be used for research purposes, followed by the
production of an MCB. For the general process flow or production of a microbial WCB, see Figure 5.
al
ci
ffi
O
As illustrated in Figure 5, in-process control testing is performed by checking the culture broth under a microscope before
the filling of containers. After filling, the suspension of culture broth and glycerol (or DMSO) is checked for contamination by
monitoring growth on appropriate plates. The containers are labeled individually with batch number, cell bank identification,
and container number and then filled. The WCB containers are kept frozen under well-defined and controlled conditions.
ADDITIONAL CONSIDERATIONS
During development, the production strains/cell lines used for manufacturing clinical materials may continue to evolve to
meet the commercial manufacturing needs. For example, new expression plasmids or new parental cells (or both) may be used
to generate higher-yielding production strains/cell lines for commercial applications. Additional testing considerations may be
needed when retroviruses (e.g., lentiviruses) are used to insert the gene of interest. The new cell lines may be from a new species
(e.g., a change from NS0 to CHO) or from the same species with additional genetic modifications to improve the cell culture
performance and/or the quality/productivity of the therapeutic proteins. In such cases, it may be helpful to provide a summary
of the linkage among the expression plasmids/parental cells used throughout product development. In addition, comparability
and other product impacts should be evaluated. To avoid any unexpected loss of the cell bank, the containers are typically kept
in at least two different geographical locations. During transportation, the containers are shipped and are continuously
monitored for temperature to confirm that they remain acceptably frozen.
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 11/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
12
4.1 Overview
This section discusses clonality of production cell lines derived from mammalian cells. Obtaining a stable cell line that
maintains high levels of recombinant therapeutic protein expression and consistent product quality is critical for commercial
development. Typically, production cell lines are derived from immortalized cells, such as CHO cells, which exhibit a relatively
high degree of genetic plasticity (6). Therefore, the addition of a cloning step is important to the overall cell line generation
process. Incorporation of a cloning step serves two core purposes:
1. Minimizing cell population heterogeneity
2. Isolating a highly productive, stable cell population
It is recognized that incorporation of a cloning step does not prevent genetic and phenotypic heterogeneity within the
population of cells that arises post-cloning due to genetic features of immortalized mammalian cell lines. However, non-clonally
derived cell lines have been used for toxicology studies. It is important to demonstrate that critical quality attributes (CQAs) of
the preclinical and clinical proteins are similar enough to assure safety based upon the toxicology studies.
This section describes relevant regulatory guidances regarding clonality as well as practices to achieve clonally derived
production cell lines. These practices include limiting dilution, fluorescence-activated cell sorting (FACS), semisolid plating, and
single-cell printing (see Table 2). In addition, this section describes principles and types of data used to support clonal derivation
of a cell line.
The concept of clonality is recognized in multiple regulatory guidance documents. For instance, ICH Q5D (7) states: “For
recombinant protein products, the cell substrate is the transfected cell containing the desired sequences, which has been cloned
al
from a single cell progenitor.” ICH Q5B (6) defines the MCB as: “An aliquot of a single pool of cells which generally has been
prepared from the selected cell clone.” The guidance goes on to indicate that “In addition, methods used to amplify the
expression construct and criteria used to select the cell clone for production should be described in detail.” Likewise, the FDA’s
Points to Consider in the Manufacture and Testing of Monoclonal Antibody Products for Human Use (4) indicates that a “description
of the cell cloning procedures” should be captured in documentation.
ci
The aforementioned guidance documents indicate a general expectation that a clonally derived cell line be used to produce
recombinant therapeutic products. Using a cell line derived from a single progenitor minimizes the variability of the starting
cryopreserved cell population, which would be anticipated to have the effect of minimizing product heterogeneity. However,
sustained culturing of immortalized cells can result in genetic alterations, which may be further exacerbated by amplification
procedures. Therefore, it is not feasible to term a population as truly genetically homogeneous following sustained culture.
Despite this, the reason for generating a clonally derived cell line relates to the ability of a controlled process to produce a
ffi
consistent product with minimal heterogeneity. Thus, for these reasons, any adaptations (e.g., switching to serum-free
conditions) should be considered prior to cloning. In contrast, use of an entirely non-clonal cell population as a starting point
may give rise to outgrowth of a subpopulation of cells that generate products with different CQAs. For instance, this could
affect glycosylation, which could then impact the mechanism of action if the product is an antibody that functions by
antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC). Likewise, a different
population with a different integration site might have altered expression levels, growth metrics, and stability, which could have
O
the potential to lead to drug shortages if a cell bank is no longer performing as expected. Such adverse end points could be
exacerbated in conditions where cell culture parameters or raw materials have been altered in a way that places selective pressure
on the system.
4.2 Methodology
Several methodologies used to generate clonally derived cell lines are described below.
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 12/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
13
al
protein is secreted into the medium, a halo of fluorescence builds up around the growing clone of interest. The system is able
to image the cells under both white and fluorescent light; thus, both the cell colony that is growing and the amount of
fluorescence that is being produced can be monitored. Up to a threshold level, mean exterior fluorescence has been shown to
correlate with antibody productivity; however, this relationship was not maintained at levels above the threshold. On the basis
of preset thresholds, the machine will automatically pick clones of interest and place them into microtiter plates for expansion.
Optimization of the parameters used may be necessary. As with FACS, it is important to verify that the apparatus has been
ci
sanitized since its last use to avoid cross-contamination. Also, potential adventitious agents arising from the antibody used in
the selection process are a concern; therefore, appropriate testing should be performed. As with other methods, it is important
to keep in mind cellular behavior such as clumping. In addition, the plating density can impact the ability of the machine to
select only 1 colony. Thus, such machines are often used in combination with a single round of LDC to support assurance of
clonality.
ffi
OTHER AUTOMATED CLONE SELECTION TECHNOLOGY
There are many methods that can be used for clone selection, and other methods may be developed. One example of clone
selection methods is to combine high-speed image scanning and quantitation with high-speed laser manipulation, which allows
for in situ measurement of recombinant protein secretion of single cells, followed by laser-mediated destruction of the
O
low-producing cells in a population. This can enable higher throughput by using 384-well plates instead of 96-well plates.
Image documentation of a clone begins on day 0, immediately after plating, with the aid of a fluorescent viable cell dye and
continues throughout the entire expansion process. Multicolor live cell imaging is also possible through the simultaneous use
of fluorescence detection reagents that associate specifically with either the cell clones or the expressed recombinant protein.
The sample wells are coated with capture matrix to mediate in situ capture of expressed recombinant protein as most of the
recombinant proteins are secreted out of the cell. By measuring the fluorescent intensity of the respective detection reagents,
each cell in the same well is ranked based on the amount of secreted recombinant protein. Other criteria such as cell growth
rate, area, and proximity to other cells also affect the ranking. The highest-ranking cell in a well is identified as the most suitable
high-producer cell clone. Subsequently, a laser beam is directed to the lower-ranking cell clones in the same well to induce
photomechanical lysis. This allows the high-producing cell remaining in the well to grow and then be transferred to a larger
well for expansion, clone characterization, and cell banking. The platform could be used alone or in combination with a single
round of LDC.
See Table 2 for a comparison of the above methods.
Table 2
Method Comparison Advantages Disadvantages
• Statistically powered with two rounds • One round insufficient to provide monoclonal-
• Least expensive ity assurance; requires additional supplemen-
Limiting dilution cloning tal data
• Labor and time intensive
• Requires accurate initial cell counting
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 13/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
14
Table 2 (continued)
Method Comparison Advantages Disadvantages
• Identification of isolated individual cells • Use of semisolid media may result in stacked
• Can be coupled with LDC cells in z-axis
• Can be used to identify high-producing cells • May require an additional round of LDC
Automated clone picking from semisolid medium • Does not allow imaging of single cells
• Requires expensive equipment
• Requires equipment qualification
• Requires highly trained operators
al
When probability calculations are included, they should be done so prospectively. Back calculation from the number of wells
in which the colonies grew does not take into account the number of wells in which cells failed to grow, resulting in an
overestimate of monoclonality.
ci IMAGERS
Imagers that can document the contents of an entire microplate well2 from day 0 through colony growth can be powerful
tools. An image of a single expanded colony would not be enough evidence of monoclonality, but a time lapse showing the
development of the colony from a single cell would provide supportive evidence. In this case, it would be important that the
entire well was captured to prove that only 1 cell was in the well from the beginning. Furthermore, day 0 fluorescence imaging
as applicable allows for the detection of living cells and for differentiation from cell-like artifacts that can be observed in the
ffi
brightfield image.
GENETIC SUPPORT
For clones with random (non-site-specific) integration, genetic information supporting the location of the integration site
provides evidence that the cell line was clonal. Information about the sites of integration can help to identify the number and
O
location of genomic integration sites. Integration site identification does not prove that the cell line was clonally derived.
Change to read:
5. CELL BANK CHARACTERIZATION
Depending on the type of cells used, the biological production processes for cell banking can be variable, which has important
consequences for the safety and efficacy of the resulting product. In order to introduce these cell-derived products into routine
clinical use, the manufacturer must ensure that robust cell banking procedures are developed on the basis of process
understanding and characterization, which should result in consistent production and quality of cell banks. Process
understanding and consistency are critical because slight changes in cell growth conditions can occasionally lead to changes
(e.g., in glycosylation) that can have major adverse effects later on (e.g., changes in immunogenicity), which may be a critical
attribute for the expressed protein or polysaccharide. Validated and/or appropriately qualified-for-use test methods for
characterization of the cell bank (e.g., host cell quality, purity, freedom from adventitious agents, in-process testing) are quality
recommendations for any MCB or WCB.
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 14/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
15
a
Table 3. Microbial Cell Bank Characterization
Assay Type MCB WCB EOPb
Specification Analysis
Microbial purity + + —
Phage contamination + + —
DNA sequencing + + —
Strain identity + − —
Characterization Tests
Viability + + +
Microbial purity c — — +
DNA sequence — — +
Phage contamination — — +
al
Nucleic acid fingerprinting + + +
a ICH Q5A was under revision during development of this chapter. The table is aligned with the current R1 version of ICH Q5A. For more information see current
version of ICH Q5A.
b EOP, end of production.
c Purity needs to be performed on EOP if not done in MCB and WCB.
ci
d This test may be more appropriate to perform on the end-of-growth sample.
Microbial Contaminants
Sterility: USP/European Pharmacopoeia/ICH requirements + + +
+ ▲ f
−▲ +▲▲
In vitro assayd, e (ERR 1-May-2023) (ERR 1-May-2023)
In vivo assay j + — e +
Electron microscopy + — +
Infectivity + − +
a ICH Q5A was under revision during development of this chapter. The table is aligned with the current R1 version of ICH Q5A. For more information see current
version of ICH Q5A.
b EOP, end of production.
c The strategy for testing extraneous agents should be developed based on a risk assessment following the principles of viral contamination risk (see Viral Safety
Evaluation of Biotechnology Products Derived from Cell Lines of Human or Animal Origin á1050ñ).
d The nature of the assay and choice of cells used are governed by the species of origin of the cell bank and the type of virus that may be present based on the
handling of the cells. The assay should include a human and/or a non-human primate cell line susceptible to human viruses. Both cytopathic and hemadsorbing
viruses should be sought.
e The strategy should include suitable tests that are able to detect different families of extraneous agents that may infect the source materials, including cell substrates
and raw materials of animal or plant origin. The strategy also considers the capacity of the manufacturing process to remove or inactivate viruses (see Design,
Evaluation, and Characterization of Viral Clearance Procedures á1050.1ñ).
f Performed on cells generated from the first WCB at the limit of in vitro cell age; for subsequent WCBs, a single in vitro and in vivo test can be done either directly
on the WCB or on the cells at the limit of in vitro cell age (LIVCA). On the basis of a risk assessment, these tests may not be required.
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 15/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
16
g Species-specific viruses present in rodent cell lines may be detected by mouse, rat, or hamster antibody production tests (MAP or HAP).
h Alternative validated method (e.g., NAT) agreed upon with the regulatory authority may be used.
i Selection of viruses to be screened should take into account cell lines that are known to have been infected by such agents and the source from which the cell line
was derived [e.g., mouse minute virus (MMV), Vesivirus].
j Includes adult and suckling mice and embyronated eggs. Additional animal species may be used depending on the nature and source of the cell lines being tested.
k When bovine and porcine raw materials are used, these tests should be performed. Bovine and porcine virus in vitro assay can be omitted for MCB, WCB, and
EOP if the parental cell line used to generate MCB had been tested negative for bovine and porcine viruses and no animal-derived raw material were used in
transfection and subsequent steps leading to the generation of MCB and WCB as well as in the production of EOP.
l Not required if infectivity assay is positive.
al
Used in the Production of Viral Vaccines for Infections Disease Indications (14). If the cell substrate under evaluation is found to be
tumorigenic, it must be evaluated for oncogenicity in suitable animal models (newborn nude mice, newborn hamsters, and
newborn rats) using an appropriate positive control.
For an engineered cell line, the inserted GOI should remain intact and the copy number should be determined to support
consistent production. ci
The cell bank should comply with the appropriate test for sterility given in Sterility Tests á71ñ and carried out using an
appropriate volume from cell cultures. The test should be carried out on 1% of containers with a minimum of 2 containers. The
cell bank should also comply with the test for mycoplasma in Mycoplasma Tests á63ñ performed on 1 or more containers.
Electron microscopy (EM) of the MCB should be done to detect the presence of any extraneous agents. The MCB should
also be assessed by the vitro virus assay with the two endpoints of cytopathic effect and hemadsorption. The end of production
(EOP) cells are also characterized to demonstrate absence of extraneous agents that might be present in the MCB.
ffi
Additionally, the cell bank should be tested for possible virus contaminants, depending on the origin of the cells, using
appropriate specific tests (e.g., mouse, rat, or hamster antibody production tests or a nucleic acid amplification test may be
used for specific contaminants). See Viral Safety Evaluation of Biotechnology Products Derived from Cell Lines of Human or Animal
Origin á1050ñ and Design, Evaluation, and Characterization of Viral Clearance Procedures á1050.1ñ for more details.
The MCB should be examined for retroviruses using:
O
• Infectivity assay
• Transmission electron microscopy (TEM)
• Reverse transcriptase (RT) or other appropriate assays should be performed to detect retroviruses that may be noninfectious
if infectivity is not detected and no retrovirus or retrovirus-like particles have been observed by EM
Detection for retroviruses in the MCB is governed by the host species and can include detection methods such as reverse
transcriptase activity, infectivity in sensitive cell cultures or electron microscopy. It is recommended to continue to follow
advances in new technology to supplement cell bank characterization where appropriate.
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 16/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
17
the cell banks without further cloning. For cell banks carrying chromosomal copies of the GOI, the sequence encoding the
product can be verified. This is commonly performed by sequencing of the coding regions amplified by the PCR from cDNA.
The nucleic acid sequence should be identical, within the limits of detection method, to the sequence determined for the
expression construct and should correspond to the expected protein sequence.
The most common methodology for the assessment of gross structure of the expression construct is restriction endonuclease
(RE) mapping analysis. New technologies [e.g., next generation DNA sequencing (NGS)] are currently being explored for this
purpose. For extrachromosomal expression systems, the RE analysis can be viewed through gel electrophoresis; for
chromosomally integrated expression systems, the RE analysis is usually accomplished by Southern blotting or full sequencing
by NGS. The RE digested patterns as well as the DNA sequence should be consistent for DNA derived from either the cell banks
or the cells at the LIVCA.
Although not required by ICH Q5B, transcript analysis of GOI can be performed if needed to further evaluate the integrity
of the GOI expression. A number of techniques can be used for transcript analysis, including Northern blotting, reverse
transcription–polymerase chain reaction (RT–PCR) and transcriptome analysis by NGS.
The most common methodology for the evaluation of GOI copy number is quantitative PCR (qPCR). GOI-specific qPCR assays
and a normalizer qPCR assay targeting a host genome region are usually applied. In some cases, there is no strong correlation
between the stability and integrity of the therapeutic product. The GOI copy number analysis could be considered for the cell
bank characterization purposes only. The genetic determination is part of the quality control. New DNA sequencing technology
such as NGS may further enhance the capability for genetic characterization.
The impact of a change in cell culture process after the commercial process has been defined should be evaluated to
determine if retesting of the cells at LIVCA is necessary. Changes in cell culture scale and/or manufacturing site may not require
retesting of cells at LIVCA if the defined cell age covers any increase in population doubling resulting from the change and if
cell culture performance meets the predetermined acceptance criteria. However, changes in media components (especially if
animal-derived components are used) and growth conditions often result in changes in cell culture profiles, so one should
al
consider retesting the integrity of the expression construct at LIVCA to ensure suitability of the cell line for commercial
production.
Although a two-tier cell-banking system (MCB and WCB) is commonly established for the commercial manufacturing of a
drug substance, the characterization of the expression construct for each WCB is considered unnecessary if the following criteria
are met:
ci
• The MCB has been fully characterized and the expression construct is confirmed to be stable
• The cell age of the current and future WCB, as well as the EOP cells derived from the future WCB, will be controlled within
the previously determined LIVCA
If the characterization cannot be carried out on the MCB, it should be carried out on each WCB.
ffi
5.4 General Considerations for Qualification of Replacement WCBs
Replacement WCBs prepared using procedures equivalent (as described in the license) to those used to generate the
previously approved WCB must meet all specified requirements [e.g., certificate of analysis (CoA) testing] but require no further
evaluation under a validation protocol. When the new WCB is a “like-for-like” replacement, the WCB can be implemented after
O
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 17/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
18
appropriately labeled, and cryolabels must be able to withstand cryopreservation. Cryology considerations for cells are
addressed in á1044ñ. Additional considerations for cell banking are as follows:
• Cells are frozen at early passage (MCB), intermediate passage (WCB), or late passage (EOP).
• Frozen cell volumes can vary from 0.5 to >100 mL depending on the number of cells required upon thawing.
• Cell banks can be stored in cryovials or cryobags.
• Cell density can range from 106 to 108 cells/mL.
• Cell bank storage containers should be filled with an excess to accommodate loss of cells upon thaw.
• For MCB or WCB, cells should be banked at a density that allows for direct inoculation into vessels for expansion upon
thawing.
• Mammalian cells should be preserved in GMP-grade cryopreservation solution with appropriate cryoprotectant (e.g.,
DMSO or glycerol) to support cell recovery consistent with established in-process viability limits.
• Post-bank thaw testing with assessment of cell viability should be performed and documented. A viability limit should be
set and justified to demonstrate that the cells survived the cryopreservation process. The viability limit could be set based
on historical data.
• Each cell bank vial or bag should be labeled in advance with the appropriate information (e.g., cell bank lot number, tube
number, and production date) on liquid nitrogen-resistant cryolabels; use appropriate controls and limits.
If different cell banks are stored in the same facility, appropriate measures and controls (for example segregation or cryolabels)
should be in place to prevent errors due to switching and cross contamination. Different freezers or storage areas within freezers
should be used to separate the cell banks prior to release testing from the cell banks to be used for production.
al
A cell bank is considered stable if, upon thaw and subsequent passage, it can consistently meet growth and viability criteria
based on historical performance. Bank-specific limits for viability and growth rates can be set after thaw studies have been
completed. Cell bank stability can be equally ensured through trending of thaw performance data across a more substantive
data set such as WCB vial thaw and/or batch. ci
Stability limits for mammalian cell banks should be site independent, product specific, cell bank specific, and determined on
the basis of thaw performance. Stability limits can also be vial-size specific due to the operational differences between seed
strain sources from different-sized vials. Data used to establish final viability and specific growth rate limits should be
documented.
Data for MCB stability monitoring should include data from when 1 or more vials of the cryopreserved MCB are thawed for:
ffi
• Initial characterization
• Production of clinical or commercial material
• Preparation of a new WCB
Data for WCB stability monitoring can include data from when 1 or more vials of the cryopreserved WCB are thawed for a
clinical or commercial production campaign.
Data for WCB stability monitoring may also include data from appropriately documented scale-down or pilot-scale processes,
O
such as those performed during process characterization and validation studies. WCB stability should be periodically monitored
unless the bank is no longer in use for commercial production or the product has been removed from the market.
RAW MATERIALS
Control of raw materials is necessary to reduce the risk of contamination due to adventitious agents and possible cross
contamination from incoming raw materials. Contamination risk for all raw materials should be assessed; the assessment can
then be used to support product exposure history.
Qualification of raw materials: Raw materials should be qualified and characterized as to their identity, purity, functionality,
and safety. Appropriate testing for raw materials should be performed; examples include testing for endotoxins, mycoplasma,
adventitious agents, and/or sterility (see Ancillary Materials for Cell, Gene, and Tissue-Engineered Products á1043ñ). Any raw
material used in the manufacture of a cell substrate (including media components) that is obtained from suppliers should be
controlled appropriately by the cell substrate manufacturer (see á1043ñ).
The qualification program should evaluate the source, identity, purity, and safety of the raw material relative to the established
specifications for raw materials. Identification and selection of raw materials should be performed on the basis of suitability for
use in the intended process and evaluation of toxicity, availability, consistency, traceability, and potential to be sources of
contamination. If the raw materials are human derived, the certificate of origin (CoO) and donor infectious disease status should
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 18/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
19
be evaluated. If the raw materials are animal-derived, the herd qualification and country of origin certificates, should be
evaluated.
For some human- and animal-derived raw materials used in cell culture medium (e.g., insulin, transferrin), a validated
production process used for elimination of viruses may substitute for virus detection tests, when justified. The evaluation of the
suitability for use should encompass any effects on safety, potency, and purity. It should also include assessment of the potential
risk of toxicity and adventitious agents and their removal if such raw materials are used in large amounts in manufacturing.
Drug manufacturers should develop a raw materials management system as part of their quality systems that would include a
raw materials qualification program. This program should include:
1. Incoming receipt, segregation, inspection, and quality release of the raw materials prior to use
2. Vendor audits and certification
3. CoA verification testing or identity testing, as applicable
4. CoO
5. Formal procedures and policies for out of specification results
6. Stability testing
7. Archival sample storage
Any raw material supporting the cell bank growth will be an ancillary product; therefore, supplier material should be
controlled by the manufacturer. Usually a CoA and/or a risk statement addressing the likelihood of the presence of
animal-derived materials [e.g., bovine material, which carries the risk of bovine spongiform encephalopathy (BSE)] should be
provided for all media components. In addition, contracts ensuring a steady supply of a material of the proper purity level
should be secured.
al
6. CONCLUSION
The production of cell banks needs to satisfy quality standards for identity, purity, clonality, stability, and the suitability of
cells for use in manufacturing. In addition to the appropriate testing for identity, purity, and adventitious agents, the
homogeneity, reproducibility, history, cell bank generation and qualification, genetic stability, storage, raw materials, and
ci
reagent qualification, as well as good manufacturing practices, must be demonstrated.
GLOSSARY
Adventitious agent: A microorganism [including bacteria, fungi, Mycoplasma/Spiroplasma, Mycobacteria, Rickettsia,
viruses, protozoa, parasites, or transmissible spongiform encephalopathy (TSE) agents] that is inadvertently introduced into the
ffi
production of a biological product (15).
Cell bank: A collection of appropriate containers, whose contents are of uniform composition, stored under defined
conditions. Each container represents an aliquot of a single pool of cells (7).
Cell bank qualification: Any testing deemed appropriate to demonstrate fitness of use of the bank for its intended purpose
and the environmental conditions where appropriate.
Cell line: Type of cell population that originates by serial subculture of a primary cell population that can be banked (7).
O
Cell substrate: Microbial cells or cell lines derived from human or animal sources that possess the full potential for
generation of the desired biotechnological/biological products for human in vivo or ex vivo use (7).
Clonality: A group of cells derived from a single progenitor sharing common ancestry (7).
Diploid cell line: Type of cell population having a finite in vitro lifespan in which the chromosomes are paired (euploid)
and are structurally identical to those of the species from which they were derived (ICH Q5D).
End of production cells (EOP cells): Cells cultured (under conditions comparable to those used in production) from the
MCB or WCB to a passage level or population doubling level comparable to or beyond the highest level reached in production
(15).
Expression construct: The expression vector that contains the coding sequence of the recombinant protein and the
elements necessary for its expression (6).
Host cells: See Parental cells below (7).
Genetic integrity: Test to establish that the gene of interest is intact, sequence of coding sequence is correct, and there
are no insertions or deletions.
Integration site: The site where 1 or more copies of the expression construct are integrated into the host cell genome (6).
Limit of in vitro cell age (LIVCA): Time between thaw of the MCB vial(s) and harvest of the production vessel as measured
by elapsed chronological time, by population doubling level of the cells, or by passage level of the cells when subcultivated
by a defined procedure for dilution of the culture (6–7).
Master cell bank (MCB): An aliquot of a single pool of cells that generally has been prepared from the selected cell clone
under defined conditions, dispensed into multiple containers, and stored under defined conditions (6–7, 16).
Parental cells: Cells to be manipulated to give rise to a cell substrate or an intermediate cell line. For microbial expression
systems, it is typical to also describe the parental cells as the host cell (7).
Passage level: The number of times that a cell population has been removed from the culture vessel and has undergone a
subculture (passage) process (15).
Pilot plant scale: The production of a recombinant protein by using a procedure fully representative of and simulating
that to be applied on a full commercial manufacturing scale. The methods of cell expansion, harvest, and product purification
should be identical except for the scale of production (6).
Research cell bank (RCB)/Premaster research cell bank (pmRCB): Cell bank(s) developed as a precursor to the master
cell bank.
Recombinant biologic: Biological substance produced by genetic engineering techniques.
Tumorigenicity: Process by which immortalized cells form tumors when inoculated into animals (15).
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 19/20
Printed on: Fri Jun 14 2024, 02:32:40 PM(EST) Status: Currently Official on 14-Jun-2024 DocId: GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US
Printed by: Muhannad Qasem Official Date: Official as of 01-May-2023 Document Type: GENERAL CHAPTER @2024 USPC
Do Not Distribute DOI Ref: arf9u DOI: https://doi.org/10.31003/USPNF_M12415_03_01
20
Viral clearance: Elimination of target virus by removal of viral particles or inactivation of viral infectivity (5; á1050ñ,
á1050.1ñ).
Virus-like particles: Structures visible by electron microscopy that morphologically appear to be related to known viruses
(5).
Working cell bank (WCB): A collection of cells prepared from aliquots of a homogeneous suspension of cells obtained by
culturing the MCB under defined culture conditions (6–7).
REFERENCES
1. Food and Drug Administration. US Code of Federal Regulations. Title 21 CFR 610.18 Cultures.
2. Food and Drug Administration. Supplement to the points to consider in the production and testing of new drugs and
biologics produced by recombinant DNA technology: nucleic acid characterization and genetic stability, Rockville, MD:
Food and Drug Administration; April 1992.
3. Food and Drug Administration. Points to consider in the characterization of cell lines used to produce biologics. Rockville,
MD: Food and Drug Administration; July 1993.
4. Food and Drug Administration. Points to consider in the manufacture and testing of monoclonal antibody products for
human use. Rockville, MD: Food and Drug Administration; February 1997.
5. International Council for Harmonisation. Q5A(R1) Viral safety evaluation of biotechnology products derived from cell
lines of human or animal origin. 1998. https://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/ucm073454.pdf.
6. International Council for Harmonisation. Q5B Quality of biotechnological products: analysis of the expression construct
al
in cells used for the production of rDNA derived protein products. 1995. https://www.ich.org/page/quality-guidelines.
7. International Council for Harmonisation. Q5D Derivation and characterization of cell substrates used for production of
biotechnological/biological products. 1997. https://www.ich.org/page/quality-guidelines.
8. International Council for Harmonisation. Q7 Good manufacturing practice guide for active pharmaceutical ingredients.
2000. http://academy.gmp-compliance.org/guidemgr/files/3-1-18.pdf.
ci
9. International Council for Harmonisation. Q9 Quality risk management. 2005.
10. Xu X, Nagarajan H, Lewis NE, et al. The Genomic Sequence of the Chinese Hamster Ovary (CHO) K1 cell line. Nat
Biotechnol. 29(8), 735–741; 2011.
11. Berting A, Faucet M, Kreil T. Biotechnology, Bioengineering. Vol. 106, 598–607; July 1, 2010.
ffi
12. Frye C, Deshpande R, Estes S, Francissen K, Joly J, Lubiniecki A, et al. Industry view on the relative importance of “clonality”
of biopharmaceutical-producing cell lines. Biologicals. 2016;44(2):117–122.
13. European Medicines Agency. Guideline on development, production, characterisation and specification for monoclonal
antibodies and related product (3AB4A); July 2016.
14. World Health Organization. WHO Technical Report Series No. 978, Annex-3. Recommendations for the evaluation of
animal cell cultures as substrates for the manufacture of biological medicinal products and for the characterization of cell
O
banks; 2013.
15. Food and Drug Administration. Guidance for Industry: Characterization and qualification of cell substrates and other
biological starting materials used in the production of viral vaccines for infectious disease indications. Rockville, MD: Food
and Drug Administration; February 2010.
16. European Pharmacopoeia (Ph. Eur.). 9th edition. 2.6.7 Mycoplasmas.
17. Food and Drug Administration. US Code of Federal Regulations. Title 9—Animals and animal products; 2000.
▲ (USP 1-May-2023)
https://online.uspnf.com/uspnf/document/1_GUID-B6F19A67-1501-4AE1-8FB0-1843B39AF9BA_3_en-US 20/20