Fact-checked by Grok 2 weeks ago

Cell bank

A cell bank is a of standardized, reproducible lines—such as primary s, immortalized lines, or s—derived from a single source and preserved under controlled conditions, typically through , to maintain their viability, genetic stability, and functionality for applications in biomedical research and production. Cell banks play a critical role in by providing a consistent, reliable source of cells that ensures in experiments, supports large-scale of biologics like and monoclonal antibodies, and facilitates advancements in , , , , and . The process begins with the establishment of a master cell bank (MCB), which consists of a well-characterized initial stock of cells frozen in multiple aliquots after rigorous testing for identity, purity, potency, and contaminants such as or adventitious agents. From the MCB, a working cell bank (WCB) is derived under defined conditions to generate genetically identical cells for routine use in production or assays, minimizing variability and extending the lifespan of the original cell line. Cryopreservation is the cornerstone of cell banking, involving the suspension of cells in a protective medium with cryoprotectants like (DMSO) before rapid freezing to -196°C in vapor phase storage systems, which prevents formation and cellular damage. Storage occurs in secure, monitored biorepositories compliant with good manufacturing practices (GMP), with regular viability assessments and quality controls to ensure long-term stability, often spanning decades. Regulatory standards, such as the ICH Q5D guideline for cell line derivation and ISO 20387 for biobanking, along with region-specific regulations like the FDA's points to consider for cell substrates and the Directive 2004/23/EC for cells, govern cell banking to uphold , , and ethical considerations, such as material transfer agreements (MTAs) for proprietary lines. These frameworks are essential for clinical and commercial applications, where cell banks from authoritative institutions like the American Type Culture Collection (ATCC) provide authenticated materials that underpin global research and therapeutic development.

Overview

Definition and Purpose

A cell bank is a systematically organized collection of cryopreserved viable cells derived from a single cell line or primary cell source, maintained under controlled conditions to preserve their long-term stability and availability. This repository ensures that the cells retain their genetic and functional integrity, serving as a foundational resource in biotechnology. The primary purpose of a cell bank is to provide a reliable, consistent supply of well-characterized cells for reproducible scientific experiments, manufacturing, and therapeutic development, thereby minimizing variability and risks. By maintaining genetic over extended periods, cell banks support applications such as production, , and , where uniformity in cell populations is critical for safety and efficacy. This approach also facilitates the prevention of or adventitious agent introduction that could occur in continuous cultures. Basic components of a cell bank include comprehensive of the cells' origin, such as derivation history and donor screening if applicable, alongside data encompassing viability assessments, genetic profiling, and purity testing. Storage protocols, tailored to the specific cell line, are also integral to ensure traceability and compliance with regulatory standards.

Importance and Applications

Cell banks play a pivotal role in by enabling standardization through the provision of uniform populations that ensure consistent experimental and production conditions across studies and runs. This uniformity is essential for reproducible results in , as it minimizes discrepancies arising from differences in cell sources or handling protocols. By preserving cells at defined levels, cell banks reduce variability in research outcomes, allowing scientists to focus on biological insights rather than artifacts from inconsistent materials. Furthermore, they support in production by serving as a reliable for expanding cultures to meet large-scale demands, such as in therapeutic . In addition to and , cell banks act as a critical against the loss of valuable lines due to , equipment failure, or over time. This preservation strategy facilitates rapid recovery and continuity in operations, mitigating disruptions that could otherwise delay or timelines. Cell banks also address key challenges in , such as batch-to-batch variability, by enabling the use of well-characterized, frozen aliquots that maintain genetic and phenotypic stability. They further promote ethical sourcing by documenting the origins of cells from donors or immortalized lines, ensuring compliance with and standards that uphold scientific . Applications of cell banks span diverse fields, particularly in vaccine development, where they provide stable substrates for propagating viruses like , allowing for efficient seasonal and vaccine production. For instance, during the , cell banks supported the rapid scaling of vaccine manufacturing using established cell lines to meet global demands. In monoclonal antibody production, cell banks derived from optimized clones, such as Chinese hamster ovary () cells, ensure consistent high-yield expression for therapeutics targeting diseases like cancer and autoimmune disorders. Cell banks are equally vital in , where they store lines for therapies aimed at repairing damaged tissues, such as in cardiac or neurological conditions. In for CAR-T cell therapies, banked T cells from healthy donors enable the creation of off-the-shelf products that enhance accessibility and reduce manufacturing time for treating refractory cancers. Overall, these applications underscore the role of cell banks in bolstering reliability for clinical trials, ensuring a steady supply of quality-controlled cells to advance therapeutic development without interruptions.

Types

Master Cell Bank

The master cell bank (MCB) is defined as an of a single pool of s derived from a selected or , prepared under controlled conditions, dispensed into multiple containers, and stored under specified conditions to serve as the primary source for deriving all subsequent working cell banks (WCBs). It acts as the foundational, fully characterized repository ensuring consistency in biotechnological and biological product manufacturing, such as and cell therapies, by providing a uniform starting material for production lots. This role minimizes variability and supports in processes like propagation and development. The creation of an MCB involves initial cell selection from a research or preliminary , followed by controlled expansion in culture, pooling of cells, and using cryoprotectants in sealed containers, typically stored in vapor phase. At establishment, extensive testing is conducted to verify identity through methods like DNA analysis or isoenzyme assays, purity via , , and adventitious agent screening, potency by assessing growth and production consistency, and sterility according to pharmacopeial standards. To reduce handling risks and contamination potential, only a limited number of aliquots—often exceeding 200 —are produced, with each containing sufficient cells for downstream applications. Designed for long-term archival storage at temperatures below -130°C, the MCB is not intended for direct use in routine production but rather to generate WCBs, thereby preserving the original cell line's integrity over decades. Comprehensive genetic and phenotypic profiling, including karyotyping, short analysis, and studies, accompanies the MCB to document its characteristics and enable . In contrast to WCBs, which are derived from the MCB for operational use, the MCB emphasizes initial, exhaustive characterization to safeguard the production lineage.

Working Cell Bank

The working cell bank (WCB) is a collection of cryopreserved cells derived from the master cell bank (MCB) through controlled expansion and serves as the for initiating routine processes in , thereby preserving the MCB for reference and replacement purposes. Unlike the MCB, the WCB is designed for practical, expendable use in generating working cultures for direct manufacturing of biologics, such as or recombinant proteins, ensuring a consistent and stable cell population without depleting the original stock. This derivative role allows the WCB to support repeated experimental or runs while maintaining genetic and phenotypic uniformity inherited from the validated MCB. The creation of a WCB involves thawing one or more vials from the MCB, expanding the cells through limited serial subcultures under defined conditions to achieve the desired volume, pooling the expanded cells, and then aliquoting them into multiple sterile vials with a cryopreservation medium before freezing, typically in liquid nitrogen vapor phase. This process emphasizes controlled propagation to produce sufficient vials—often hundreds—for operational demands, with fewer characterization tests required compared to the MCB, as it inherits most validated properties; testing is limited to confirming identity, purity (e.g., absence of adventitious agents introduced during expansion), and basic viability, performed once per WCB batch. Such abbreviated qualification ensures efficiency while relying on the MCB's extensive prior validation to mitigate risks. In usage, the WCB supports ongoing bioprocessing by providing cells to inoculate production bioreactors for generating drug substance lots, as seen in the manufacture of monoclonal antibodies or viral vectors, where multiple vials may be pooled to start a single run. To monitor , passage numbers or doubling levels are tracked from the initial MCB thaw through WCB-derived cultures, ensuring they do not exceed predefined limits that could compromise product consistency or yield. This tracking is critical for maintaining process reliability across production campaigns, with new WCBs generated from the MCB as needed to replenish stocks.

Preparation

Cell Selection and Expansion

Cell selection for banking begins with evaluating the origin and type of the cell line, which may include primary cells derived directly from tissues, immortalized cell lines engineered for indefinite propagation, or stem cells such as mesenchymal or induced pluripotent stem cells, depending on the intended therapeutic or application. Criteria emphasize high post-thaw viability, typically targeting greater than 90% to ensure reliable recovery and functionality upon use, alongside confirmation of suitability through assessments of growth characteristics like rates and adherence to or suspension culture formats. Essential authentication methods include short tandem repeat () profiling for genetic identity verification in human cell lines and karyotyping or cytogenetic analysis to detect chromosomal abnormalities, ensuring the cells match the documented source and maintain genetic stability. Absence of contaminants is a core prerequisite, with rigorous testing required for sterility, , and adventitious agents such as viruses, achieved through documented sourcing from screened donors or tissues and comprehensive qualification of the master cell bank (MCB). These selections prioritize genetic homogeneity via low-passage cultures and scalability potential, minimizing risks of phenotypic drift or heterogeneity that could compromise downstream applications. Once selected, cell expansion involves controlled culturing to generate sufficient , often aiming for 10^8 to 10^9 cells total across multiple vials in the MCB, using static flasks for small-scale initial growth or dynamic bioreactors like stirred-tank or hollow-fiber systems for larger volumes to support uniform nutrient distribution and oxygenation. Protocols optimize culture media composition, incorporating basal nutrients, sera or serum alternatives, and specific growth factors (e.g., for stem cells) to sustain while preserving phenotypic markers such as marker expression and potential. Passage limits are strictly enforced, typically 10-20 subcultures from the original source, to avoid genetic instability or loss of functionality, with monitoring of doubling times and morphology throughout to confirm homogeneity before proceeding to banking. This process ensures the expanded population remains representative of the selected cells, facilitating scalable production without introducing variability.

Freezing Methods

Cryopreservation is a critical step in establishing cell banks, where expanded populations are preserved in a viable state for long-term storage following initial culturing and selection. This process involves the controlled and cooling of cells to prevent damage from formation, ensuring high rates upon future use. Standard methods emphasize the use of cryoprotective agents and precise to maintain cellular . Cryoprotectants are essential additives that mitigate intracellular formation by altering water's physical properties, such as depressing the freezing point and promoting glass-like . (DMSO) is widely used at concentrations of 5-10% (v/v), while serves as an alternative at 5-15% (v/v), with selection depending on sensitivity. These agents are introduced stepwise or gradually, often at 0-4°C over 10-30 minutes, to minimize osmotic shock and toxicity from rapid exposure. Freezing protocols typically begin with equilibration of the cell suspension in cryoprotectant-supplemented medium under sterile conditions, followed by dispensing into pre-labeled cryogenic vials (e.g., 1-2 mL volumes in 2-mL vials) within a to prevent . The vials are then subjected to slow-rate freezing using programmable freezers, cooling at -1 to -3°C per minute from down to -80°C, which allows extracellular formation and solute concentration without excessive intracellular damage. This is often followed by immediate transfer to vapor phase for deeper cooling. Optimization of these methods focuses on parameters like cell density, typically 10^6 to 10^7 viable cells per mL, to balance packing efficiency and post-thaw recovery, with tailored cooling curves monitored via temperature probes to achieve greater than 80% viability. Factors such as or supplementation in the freezing medium further enhance protection, and protocols are validated for specific cell lines to ensure consistent outcomes.

Storage and Maintenance

Storage Conditions

Cell banks are typically stored in the vapor phase of at -196°C to maintain ultra-low temperatures while minimizing the risk of cross-contamination, which can occur in the liquid phase due to microbial transfer through the nitrogen. Alternatively, mechanical ultra-low temperature freezers operating between -130°C and -150°C provide a reliable option for long-term preservation, ensuring cell viability without the hazards associated with immersion. These conditions halt metabolic activity, preserving cellular integrity by preventing reformation or enzymatic degradation. Storage containers consist of sterile, cryogenic-grade cryovials or ampoules, which are sealed to withstand extreme temperatures and placed in organized racks within the storage units for efficient access and tracking. Barcoding systems, often including codes etched on vials, facilitate and , ensuring precise during retrieval. To enhance security, cell banks are distributed across multiple backup sites, providing redundancy against equipment failure or environmental disasters. These storage protocols are designed for long-term stability, with studies demonstrating viability retention for up to 20 years or more in certain cryopreserved biological materials. Regular validation confirms that cells retain functionality upon thawing, supporting their use in manufacturing without degradation over extended periods.

Quality Control and Monitoring

Quality control and monitoring of cell banks during storage are essential to maintain cell integrity, prevent degradation, and ensure with regulatory standards. These protocols involve ongoing oversight to detect any deviations that could compromise the bank's usability for . Applied to master and working cell banks stored under controlled cryogenic conditions, such measures help preserve viability and purity over extended periods. Routine checks form the foundation of storage monitoring, encompassing periodic viability testing on retained vials to assess recovery rates post-thaw, typically using exclusion or automated cell counters. Temperature logging is conducted continuously via data loggers in storage units like vapor-phase systems, with alarm systems triggered for deviations exceeding set thresholds (e.g., above -130°C) to enable immediate corrective actions. Additionally, screening is performed routinely on representative samples using methods such as and or assays, while endotoxin testing via ensures absence of bacterial contaminants that could arise from storage breaches. Stability assessments evaluate long-term cell bank performance through structured studies that track genetic and phenotypic consistency. These include genetic stability monitoring via next-generation sequencing to detect mutations or insertions in expression constructs, or to analyze marker expression and levels in thawed samples. Such evaluations are conducted at intervals determined by and stability data. Documentation is meticulously maintained to support and regulatory audits, including certificates of analysis that detail viability, sterility, and results from each monitoring event. Chain-of-custody track , locations, and handling history, ensuring from deposition to retrieval. These are required in regulatory submissions and updated with each check to provide a verifiable history of bank integrity.

Usage

Thawing Procedures

Thawing procedures for cryopreserved cell banks involve retrieving vials from liquid nitrogen storage and rapidly warming them to revive cellular metabolism while minimizing damage from ice recrystallization and cryoprotectant toxicity. The standard method employs a 37°C water bath for quick thawing, typically lasting 1 to 2 minutes with gentle swirling until only a small ice crystal remains, to achieve warming rates of approximately 45–70°C per minute and limit exposure to potentially harmful intracellular ice formation. Immediately following thawing, the cell suspension is diluted in pre-warmed recovery medium to reduce osmotic shock from cryoprotectants like DMSO, which can otherwise cause cellular stress if exposure persists. Post-thaw handling prioritizes swift removal of cryoprotectants to further decrease risks. The diluted is transferred dropwise into a tube containing 9–10 mL of complete , then centrifuged at 125–200 × g for 5–10 minutes to pellet the s and discard the supernatant containing the cryoprotectant. The pellet is gently resuspended in 1–2 mL of fresh and transferred to a vessel for , with the entire process from thaw initiation to medium addition targeted to complete in under 5 minutes to optimize recovery. Best practices emphasize maintaining sterility throughout to prevent contamination, including performing all steps within a (laminar flow hood) under aseptic conditions, decontaminating the vial exterior with 70% prior to opening, and using pre-warmed media and equipment to avoid temperature shocks. , such as gloves and , is recommended due to the explosion risk from pressure buildup in cryovials stored in liquid phase.

Viability and Recovery Assessment

Viability assessment of thawed cells from a cell bank is a critical step to ensure the integrity and functionality of the cell population prior to use in manufacturing or research. This evaluation typically occurs immediately after thawing and focuses on quantifying the proportion of live cells using established assays. The trypan blue exclusion method, considered a gold standard, involves staining cells with trypan blue dye, which is excluded by intact cell membranes of viable cells, allowing manual or automated counting under a microscope to determine the percentage of live cells. Alternative assays, such as the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) colorimetric test, measure metabolic activity in viable cells by assessing the reduction of MTT to formazan, providing a complementary readout of cell health that correlates with proliferation potential. For more detailed analysis, flow cytometry can detect apoptosis markers, such as annexin V binding to phosphatidylserine on early apoptotic cells or propidium iodide uptake in late apoptotic or necrotic cells, enabling precise quantification of cell death mechanisms. A common acceptance threshold for viability is greater than 70%, as lower levels may compromise downstream applications unless supported by additional data demonstrating no impact on safety or efficacy. Beyond initial viability, recovery assessment evaluates the cells' ability to resume normal function post-thaw, typically over a short culture period. Growth rate monitoring involves seeding thawed cells and measuring population doubling times or total cell yields over 24-48 hours using techniques like or automated to confirm proliferative capacity comparable to pre-freeze conditions. For adherent cell types, attachment is assessed by calculating the percentage of cells that adhere to the culture substrate within 1-24 hours post-seeding, often via or dye-based quantification, as this metric strongly correlates with long-term colony formation and expansion success. Functional assays, such as generating proliferation curves through serial passaging or specific activity tests (e.g., marker expression via ), further verify that recovered cells maintain their intended biological properties. Acceptance criteria for viability and recovery ensure that thawed cells from the or working cell exhibit characteristics matching the original bank, including viability above 70%, stable growth kinetics, and absence of aberrant phenotypes, thereby validating the process's efficacy before proceeding to production or experimentation. This post-thaw evaluation directly follows the thawing procedure to confirm cell bank stability under storage conditions.

Regulatory Framework

Standards and Guidelines

Cell banks are governed by several key international and industry frameworks to ensure safety, quality, and consistency in their use for biotechnological and medicinal products. In the United States, the (FDA) regulates biological products, including cell banks, through 21 CFR Part 610, which outlines general standards for potency, sterility, purity, and safety to prevent risks such as contamination or loss of viability. In the , the (EMA) issues guidelines on human cell-based medicinal products, covering aspects of , , and to support the authorization of advanced therapy medicinal products derived from cells. Additionally, the Council for Harmonisation (ICH) Q5D guideline provides harmonized standards for the derivation and characterization of cell substrates, including requirements for genetic stability, identity verification, and adventitious agent testing in cell lines used for biotechnological production. Core requirements across these frameworks prioritize (GMP) compliance to maintain controlled conditions during cell banking processes, ensuring reproducibility and minimizing variability. is mandated from the origin of the cells through all stages of banking, including documentation of donor information, processing steps, and storage details, to facilitate accountability and . Risk-based approaches to control are emphasized, involving assessments for microbial, , and genetic contaminants, with measures such as validated procedures and integrated into operations. Global variations exist to address specific applications, such as the (WHO) recommendations for vaccine seed lots, which require the establishment of master and working seed lots with rigorous testing for identity, purity, and stability to support consistent vaccine manufacturing worldwide. For stem cell banks, the International Society for Cell & (ISCT) endorses standards like ISO/TS 22859 and ISO 24651, focusing on biobanking protocols for mesenchymal stromal cells from sources such as and , including detailed characterization for potency, immunophenotype, and sterility to enable therapeutic applications. These standards apply broadly to all cell bank types, promoting in global research and clinical use.

Validation and Testing Requirements

Validation and testing requirements for cell banks ensure the integrity, safety, and consistency of cells used in biopharmaceutical production and therapeutic applications. These processes are governed by international standards such as the International Council for Harmonisation (ICH) Q5D guideline, which outlines comprehensive testing to qualify master cell banks (MCBs) and working cell banks (WCBs). Testing is typically performed on aliquots of the bank or derived cultures, with the goal of verifying that the cells meet predefined specifications before release for use. The testing suite encompasses several key categories to assess the bank's quality attributes. Identity testing confirms the cells' origin and uniqueness, often employing DNA fingerprinting techniques such as (RFLP) or variable number tandem repeats (VNTR) analysis for metazoan cells, alongside cytogenetic methods like karyotyping to detect genetic stability. Purity evaluations address contamination risks, including sterility tests for and fungi compliant with pharmacopeial standards (e.g., Ph. Eur., USP), detection via or culture methods, and screening for adventitious agents such as viruses through / assays and molecular techniques like . Potency assessments verify the cells' functional capability, utilizing assays to confirm consistent product expression or stability, particularly at the limits of cell age. Safety testing includes evaluations, such as limits on residual DNA (e.g., <10 ng per dose for parenteral products), and tumorigenicity studies in animal models like nude mice for continuous cell lines to rule out oncogenic potential. Validation steps are integral to establishing the reliability of cell bank processes and long-term usability. Process validation for freezing and thawing involves viability assessments post-reconstitution to ensure recovery rates meet acceptance criteria, which vary depending on cell type. Stability studies evaluate the bank's integrity over its shelf-life under defined storage conditions, testing at multiple time points to confirm no degradation in identity, purity, or potency. Comparability protocols are required for bank extensions or modifications, comparing the new bank's characteristics to the original via side-by-side testing to demonstrate equivalence in performance and safety. Documentation is a critical component to support and . Comprehensive batch records detail the cell , banking procedures, and all testing outcomes, while audit trails maintain electronic or paper logs of all manipulations and decisions. Release criteria are predefined specifications for each quality attribute, such as viability thresholds and absence of contaminants, ensuring only qualified banks are approved for downstream use; failure to meet these triggers rejection and investigation per good manufacturing practices (GMP).

History

Early Development

The concept of cell banking emerged in the mid-20th century, driven by advances in techniques that enabled the long-term storage of viable cells for and production purposes. In 1949, researchers Christopher Polge, Audrey U. , and Alan S. Parkes accidentally discovered the cryoprotective properties of while attempting to preserve spermatozoa, allowing revival after freezing to -79°C. This breakthrough was quickly extended to mammalian cells; by 1950, Polge and colleagues successfully cryopreserved bull spermatozoa using , demonstrating its efficacy for preserving cellular integrity during freeze-thaw cycles. further applied these methods to human red blood cells, confirming glycerol's role in preventing haemolysis at low temperatures. These early cryopreservation innovations were pivotal for virus propagation in cell cultures, as they addressed the challenge of maintaining consistent cell lines for virological studies. In the 1950s, as tissue culture techniques advanced, researchers began storing frozen aliquots of cell lines to ensure reproducibility in experiments, marking the initial shift toward formalized cell banking systems. This was particularly relevant in virology, where stable cell substrates were needed to grow viruses without repeated isolation from animal tissues, reducing variability and contamination risks. The World Health Organization (WHO) formalized early guidelines for cell substrates in 1959, specifically for inactivated polio vaccine production using primary monkey kidney cells, emphasizing the need for standardized, testable cell sources. Initial applications of cell banking focused on for production, with short-term storage in research laboratories to support consistent virus yields. In the 1960s, the development of human diploid cell strains, such as by in 1962, revolutionized this field by providing safer alternatives to primary animal cells for virus propagation. cells were banked in frozen vials to serve as a reliable source for manufacturing, enabling the production of vaccines free from simian viruses like that had contaminated earlier monkey kidney-based lots. These seed banks of cell lines facilitated the global rollout of vaccines, prioritizing controlled storage over ad hoc culturing. Foundational techniques relied on as the primary cryoprotectant, added to suspensions at concentrations of 10-15% before slow freezing to -70°C in mechanical freezers, followed by transfer to for indefinite storage. This basic protocol minimized formation and osmotic damage, achieving post-thaw viabilities of 50-80% in early mammalian experiments. These methods laid the groundwork for broader applications in .

Key Milestones and Advances

In the 1980s, the advent of technology prompted the (WHO) to introduce the master cell bank (MCB) and working cell bank (WCB) system as a standardized framework for ensuring the consistency and safety of cell substrates used in producing biologicals, particularly recombinant proteins and . This two-tiered banking approach, formalized in WHO's 1987 requirements for animal cell substrates, minimized variability by deriving the WCB from the MCB and requiring extensive characterization for contaminants, genetic stability, and productivity prior to use. By the 1990s, the U.S. (FDA) extended these principles through (GMP) guidelines tailored to , emphasizing rigorous validation of cell banks to support viral vector production and somatic cell modifications in clinical applications. The marked significant technological progress in cell banking, with the widespread adoption of automated controlled-rate freezing systems to enhance reproducibility and cell viability during large-scale production. These systems, such as programmable freezers from manufacturers like Planer and Thermo Fisher, allowed precise cooling rates (typically 1–2°C/min) to prevent formation, reducing batch-to-batch variability in master and working banks for mammalian cell lines like . Concurrently, advances in genomic stability assessment tools, including (FISH) and (CGH), became integral to bank qualification, enabling detection of chromosomal aberrations and ensuring long-term genetic integrity over multiple passages. From the onward, cell banking evolved to support , with the establishment of (iPSC) banks facilitating personalized therapies by providing HLA-matched lines for disease modeling and transplantation. Institutions like Japan's Center for iPS Cell Research and Application (CiRA) developed GMP-compliant iPSC banks in the mid-2010s, storing thousands of lines to enable off-the-shelf applications in conditions such as . This period also saw the integration of CRISPR-Cas9-edited cell lines into banking protocols, allowing precise genomic modifications for therapeutic enhancement, such as knockouts for improved safety in CAR-T cells, with banks now routinely incorporating next-generation sequencing (NGS) for edit verification and stability monitoring. Cell banks played a pivotal role in the rapid response to the , exemplified by the accelerated establishment of bacterial cell banks (e.g., E. coli) for DNA production in manufacturing. In 2020, companies like and / quickly generated and qualified MCBs and WCBs under emergency GMP protocols to scale up yields, enabling billions of doses within months while maintaining quality through expedited sterility and sequence integrity testing.

References

  1. [1]
    Unlocking Potential: A Comprehensive Overview of Cell Culture ...
    Nov 10, 2024 · A cell bank is expected to provide a reliable and standardized repository of biological materials, including various types of cell lines such as ...
  2. [2]
    Cell Bank Production | ATCC
    The cell bank can only be as good as the cells from which it was developed, especially for primary cell and stem cell storage banks. ATCC has a robust process, ...
  3. [3]
    Cell Banking - Eurofins Scientific
    A cell bank is a repository or collection of cells that are derived from a single cell source, such as a cell line or primary cell culture.
  4. [4]
    Cell banking: Definition, Process & Manufacturing
    Aug 1, 2023 · Cell banking is the process of preserving and storing cells under controlled conditions to ensure their viability and genetic stability for ...
  5. [5]
    What is the Difference Between Master Cell Bank and Working Cell ...
    May 24, 2022 · Cell banking provides a characterized, common starting source for each batch and ensure that there is an adequate supply of equivalent cells for ...
  6. [6]
    The purpose of cell banking: Uses & Applications - Single Use Support
    Aug 1, 2023 · Cell banking's purpose is to preserve and store cell lines, ensuring a stable, consistent source for research and biopharmaceutical production.Cell Banking In Brief · Stem Cell Research And... · Cell Therapies
  7. [7]
    Cell bank system, establishment, and application in the virus ... - NIH
    Aug 15, 2025 · It provides sufficient quantities of cells for research, development, and manufacturing processes.
  8. [8]
    [PDF] Guidance for Industry- Characterization and Qualification of Cell ...
    As with cell banks, you should document passage history and derivation history of viral seeds. Your description should include donor screening, testing and ...
  9. [9]
    Cell Bank Preparation and Characterization - BioProcess International
    Jun 7, 2021 · Living cells are at the heart of biotechnology, and cell lines for production and testing of biopharmaceuticals are highly valuable assets.
  10. [10]
    Advantages of cell banking - Single Use Support
    Aug 1, 2023 · Cell banking plays a crucial role in maintaining the integrity of cell lines over time. By freezing and storing cells at specific passage levels ...Missing: variability scalability
  11. [11]
    Developing Stable Cell Banks for Commercial Production
    Oct 9, 2025 · Building a stable cell bank requires that every vial from the MCB is genetically, phenotypically, and functionally identical to every other vial ...<|control11|><|separator|>
  12. [12]
    5 Ways Mammalian Cell Banking Protects Against Production Risks
    Sep 22, 2025 · Unexpected events like contamination or equipment failure can halt production. With cell banks, recovery is faster and less disruptive to supply ...
  13. [13]
    Cell culture processes for monoclonal antibody production - PMC
    This review provides an overview of the state-of-the art technology in key aspects of cell culture, eg, generation of highly productive cell lines and ...Cell Line Development · Cell Line Engineering · Bioreactor Optimization And...
  14. [14]
    Cell culture-based influenza vaccines: A necessary and ... - NIH
    Cell culture-based influenza vaccine platform can be used both for production of seasonal vaccine, and to mitigate vaccine shortages in pandemic situations.
  15. [15]
    Cell banks and stability of antibody production - PubMed
    A pool of cells derived from a single clone is frozen in a number of vials (say 100) to create a master cell bank (MCB). This MCB is carefully stored in liquid ...
  16. [16]
    Archive: Stem Cell Banks Envisioned for Regenerative Medicine
    Nov 29, 2012 · Stem cell “banks” could serve as a valuable resource for emerging treatments in the field of regenerative medicine, though challenges remain ...
  17. [17]
    Off-the-shelf CAR T-cells: how do we get there? - PubMed Central
    Banked CAR T-cells immediately available for off-the-shelf (OTS) application can solve key limitations of patient-specific CAR T-cell products while retaining ...
  18. [18]
    None
    ### Summary of Master Cell Bank (MCB) from ICH Q5D Guideline
  19. [19]
    Guidelines for the use of cell lines in biomedical research - PMC
    Cell lines can be developed in-house, acquired from other laboratories (if there is no more reliable source) or purchased from a cell bank. Whatever the source ...
  20. [20]
    Recommendations for Cell Banks Used in GXP Assays
    Jan 1, 2012 · Contaminated or unstable cell lines can have a major impact on patient safety through changes to expressed proteins, so it is reasonable to ...
  21. [21]
    Authentication of Human and Mouse Cell Lines by Short Tandem ...
    Jan 17, 2023 · The simple molecular genotyping technique Short Tandem Repeat (STR) DNA genotyping of human and mouse cell lines is available.
  22. [22]
    cGMP Cell Banking Services - Applied StemCell
    Master Cell Bank. Master Cell Bank. Number of Vials: 50-200; Cell Count: 1×10^6 cells/vial; Full QC to standard or custom parameters with Certificate of ...
  23. [23]
    Human Pluripotent Stem Cell Culture - NIH
    In this review, we dissect these growth components by comparing cell culture methods and identifying the benefits and pitfalls associated with each one.Human Pluripotent Stem Cell... · Cell Culture Platforms For... · Emerging Coculture Systems...
  24. [24]
    Cell line passage numbers explained | Culture Collections
    The passage number of a cell culture is a record of the number of times the culture has been subcultured, ie harvested and reseeded into multiple 'daughter' ...
  25. [25]
    Biological Considerations in Scaling Up Therapeutic Cell ...
    This review outlines our current understanding of MSC properties and immunomodulatory function, expansion in commercial manufacturing systems, and gaps in our ...
  26. [26]
    Cryopreservation: An Overview of Principles and Cell-Specific ... - NIH
    Mar 24, 2021 · Glycerol or DMSO can be used as CPAs for long-term storage of islet cells at subzero temperatures (typically less than −100°C). Chandravanshi et ...
  27. [27]
    Cryogenic Storage of Animal Cells - ATCC
    DMSO (ATCC 4-X, 5 × 5 mL) is most often used at a concentration of 5 to 10% (v/v) in freezing media; the optimum concentration varies with the cell line.
  28. [28]
    [PDF] gc-1044-cryopreservation-of-cells.pdf - US Pharmacopeia (USP)
    Sep 27, 2018 · This chapter presents best practices for cryopreservation, maintenance, and use of a wide range of cells, cell therapy products, and cell banks ...
  29. [29]
    [PDF] Q5D Guideline - ICH
    Cell banks used to prepare gene therapy products or vaccines should follow the recommendations presented in this document. Some biological products, such as ...<|control11|><|separator|>
  30. [30]
  31. [31]
    Genetic Stability Testing Using NGS and MAM: Optimizing CHO Cell ...
    Sep 11, 2025 · Genetic stability testing is the process of verifying that a cell line retains its genetic composition over time, without accumulating mutations ...Missing: flow cytometry
  32. [32]
    Use of flow-cytometric analysis to optimize cell banking strategies for ...
    Here, we describe a novel strategy employing flow-cytometric (FC) analysis of Annexin V-stained cells for high-throughput characterization of cell banks. Our ...Missing: sequencing | Show results with:sequencing
  33. [33]
    ATCC Animal Cell Culture Guide
    The recovery of cryopreserved cells is straightforward: Cells are thawed rapidly in a water bath at 37°C, removed from the freeze-medium by gentle ...
  34. [34]
    Thawing Cells | Thermo Fisher Scientific - US
    Remove the cryovial containing frozen cells from the liquid nitrogen storage and immediately place it into a 37°C water bath. · Quickly thaw the cells (<1 minute) ...
  35. [35]
    Guidelines for the use of flow cytometry and cell sorting in ...
    Testing for cell viability. The “gold standard” of viability determination is the Trypan blue staining technique, which is carried out with a hemocytometer ...
  36. [36]
    a comparison with MTT, NR, Trypan blue and cloning efficiency assays
    In this paper we compared high-throughput real-time measurements through Electric Cell-substrate Impedance-Sensing (ECIS®) with endpoint measurements of ...
  37. [37]
    Viable Mononuclear Cell Stability Study for Implementation in a ...
    Our data suggest flow cytometry as an analytical method of choice, because of its higher specificity relative to Trypan Blue assessment and to CASY cell ...Missing: MTT | Show results with:MTT
  38. [38]
    [PDF] Guidance for FDA Reviewers and Sponsors
    You should establish minimum release criteria for viability. For somatic cellular therapies, the minimum acceptable viability specification is generally set at ...
  39. [39]
    Post-Thaw Culture and Measurement of Total Cell Recovery Is ...
    Jun 5, 2020 · We critically study post-thaw cellular outcomes upon addition of macromolecular cryoprotectants to provide unambiguous evidence that post-thaw culturing time ...Scheme 1 · Cryopreservation Of Cell... · Apoptosis Assay
  40. [40]
    The effect of Me2SO overexposure during cryopreservation on HOS ...
    Notably, a correlation of attachment to long term growth of 0.9704 was observed (Fig. 2E), indicating that a 1 h attachment efficiency assay reflected the ...The Effect Of Meso... · 2. Materials And Methods · 3. Results And Discussion
  41. [41]
    Efficient and reproducible generation of human iPSC-derived ...
    Jul 15, 2024 · Our freeze/thaw protocol yielded ~94% viable cells (trypan blue negative) after cryo-recovery, with plating efficiencies of ~51% for bCMs and ~ ...
  42. [42]
    21 CFR Part 610 -- General Biological Products Standards - eCFR
    No lot of any licensed product shall be released by the manufacturer prior to the completion of tests for conformity with standards applicable to such product.Title 21 · 21 CFR 610.12 -- Sterility. · 21 CFR 610.18 -- Cultures. · 610.39 – 610.48
  43. [43]
    [PDF] GUIDELINE ON HUMAN CELL-BASED MEDICINAL PRODUCTS
    May 30, 2008 · This guideline replaces the Points to Consider on the Manufacture and Quality Control of Human. Somatic Cell Therapy Medicinal Products ...
  44. [44]
    [PDF] Annex 2 - World Health Organization (WHO)
    Annex 2 provides guidelines for independent lot release of vaccines, involving assessment before market release, and confirmation each lot meets specifications.
  45. [45]
    Christopher Polge and Lionel Edward Aston Rowson's Experiments ...
    Dec 19, 2017 · In 1950, Polge and Rowson began a series of two experiments in which they attempted to freeze mammalian sperm that can be used to successfully ...Missing: origins banking
  46. [46]
    Cryopreservation: An emerging paradigm change - PMC
    In 1949 Polge, Parks and Smith reported on the “chance” discovery of glycerol's cryoprotective function during their efforts to preserve avian spermatozoa in ...Missing: origins | Show results with:origins
  47. [47]
    Cell bank system, establishment, and application in the virus ...
    The two primary types of cell banks in biotechnology manufacturing are the master cell bank and the working cell bank, each of which serves different ...<|control11|><|separator|>
  48. [48]
    Cell substrates - World Health Organization (WHO)
    The first WHO requirements for cell cultures used in the production of biologicals were formulated in 1959 as part of the production of inactivated ...
  49. [49]
    The Role of the WI-38 Cell Strain in Saving Lives and Reducing ...
    Mar 2, 2017 · Here we illustrate the unique contribution of the WI-38 cell strain to all human virus vaccines administered globally since the strains' origin ...
  50. [50]
    The controversial cells that saved 10 million lives - BBC
    Nov 4, 2020 · For decades, the polio vaccine had been made in cells taken from monkey kidneys, some of which – it was later discovered – were infected ...
  51. [51]
    [PDF] Annex 3 - World Health Organization (WHO)
    The concept of a master cell bank (MCB) and working cell bank. (WCB) system and the characterization of the cell substrate were introduced during that period ...
  52. [52]
    WHO requirements for the use of animal cells as in vitro substrates ...
    WHO requirements for the use of animal cells as in vitro substrates for the production of biologicals (Requirements for biological susbstances no. 50).
  53. [53]
    [PDF] Recommendations for the evaluation of animal cell cultures as ...
    The first requirements for cell substrates were published by WHO in 1959 for the production of inactivated poliomyelitis vaccine in PCCs derived from the ...
  54. [54]
    Human iPSC banking: barriers and opportunities
    Oct 28, 2019 · The introduction of induced pluripotent stem cells (iPSCs) has opened up the potential for personalized cell therapies and ushered in new ...
  55. [55]
    Induced pluripotent stem cells (iPSCs): molecular mechanisms of ...
    Apr 26, 2024 · iPSCs have been widely applied to model human development and diseases, perform drug screening, and develop cell therapies.
  56. [56]
    [PDF] assessment report: COVID-19 vaccine Moderna
    Jan 4, 2021 · The applicant requested the active substance CX-024414 (single-stranded, 5'-capped messenger RNA. (mRNA) produced using a cell-free in vitro ...