Fact-checked by Grok 2 weeks ago

Hybridoma technology

Hybridoma technology is a biotechnological method for producing monoclonal antibodies, involving the fusion of antibody-secreting B lymphocytes from an immunized animal with immortal myeloma cells to create stable hybridoma cell lines that continuously produce antibodies of a single specificity. Developed in 1975 by Georges Köhler and at the Laboratory of Molecular Biology in , , this technique revolutionized by enabling the large-scale, reproducible generation of highly specific antibodies, earning its inventors the 1984 in or . The process begins with the of an animal, typically a , using a target antigen to stimulate production of specific , followed by isolation of spleen and their fusion with myeloma cells—often using () or electrofusion—to form . These cells are then selected in hypoxanthine-aminopterin-thymidine (, which eliminates unfused myeloma cells and non-fused , allowing only hybridomas to survive and proliferate. Subsequent screening identifies clones secreting the desired , which can be expanded through or via fluid production in , yielding high-purity monoclonal antibodies suitable for research, diagnostics, and therapeutics. Key advantages of hybridoma technology include its ability to produce unlimited quantities of identical antibodies with high specificity and , reducing variability compared to polyclonal antibody production, and its cost-effectiveness for long-term applications once established. However, limitations persist, such as the time-intensive nature of the process (typically 6–9 months), low efficiency (less than 1% viability), potential for genetic instability in hybridomas, and immunogenicity issues arising from murine-derived in human therapies, which have spurred advancements like humanization techniques. In clinical and research contexts, hybridoma-derived monoclonal antibodies have transformed fields like (e.g., rituximab for ), infectious disease diagnostics (e.g., pregnancy tests and ), and , with over 200 approved therapeutics worldwide as of 2025, underscoring its enduring impact despite competition from recombinant methods. Ongoing innovations, including in screening and , continue to enhance its efficiency and applicability in .

History and Development

Discovery and Early Work

Hybridoma technology was pioneered by Georges Köhler and in 1975 at the Medical Research Council (MRC) Laboratory of Molecular Biology in , . Working to address the limitations of polyclonal antibody production, which yielded heterogeneous mixtures lacking specificity, they sought a method to generate pure, monoclonal antibodies in continuous culture. Their approach involved somatic cell hybridization, fusing antibody-producing B lymphocytes with immortal myeloma cells to create stable hybridomas capable of indefinite proliferation while retaining the desired antibody-secreting function. In their foundational experiments, Köhler and Milstein fused mouse B cells isolated from the spleens of immunized mice with mouse myeloma cell lines, such as the X63-Ag8 variant, to produce hybrid cells. These fusions, often induced using inactivated Sendai virus, demonstrated the potential for hybrid cells to express immunoglobulin chains from both parental origins without , a key insight into . Building on earlier preparatory work involving mouse-rat myeloma fusions, they achieved stable hybrids that secreted antibodies of predefined specificity. The breakthrough came in early 1975 when Köhler demonstrated the production of monoclonal antibodies specifically targeting sheep red blood s (SRBC), verified through a plaque assay that confirmed the hybridomas' ability to lyse SRBC in the presence of complement. This marked the first successful generation of continuous lines secreting homogeneous antibodies, as detailed in their seminal . Early efforts were hampered by challenges including unstable hybrid fusions, where cells often reverted to non-secretory states, and low fusion efficiency, with most attempts yielding non-viable or unproductive clones. Köhler and Milstein overcame these hurdles through meticulous selection of myeloma lines deficient in enzymes like (HGPRT), enabling to selectively propagate only the fused hybrids, and by optimizing fusion conditions to improve yield. These innovations laid the groundwork for reliable production.

Key Milestones and Nobel Recognition

Following the foundational experiments reported in 1975, which demonstrated the feasibility of fusing antibody-producing cells with myeloma cells to generate stable hybridomas secreting monoclonal antibodies of predefined specificity, Köhler and Milstein published a follow-up study in that established the technique's and broad applicability across different antigens. This work detailed the derivation of specific antibody-producing and tumor lines through , confirming the method's reliability for producing homogeneous antibodies with high specificity, thereby paving the way for widespread adoption in immunological research. Notably, Köhler and Milstein chose not to the hybridoma technique, facilitating its free dissemination and accelerating its integration into scientific and commercial practices worldwide. The profound impact of hybridoma technology was internationally recognized in 1984, when Georges J.F. Köhler and were awarded the in Physiology or Medicine, jointly with Niels K. Jerne, for the discovery of the principle for production of monoclonal antibodies. The Nobel Committee highlighted how the hybridoma technique revolutionized the ability to produce unlimited quantities of identical antibodies, enabling precise tools for studying immune responses and disease mechanisms. In the 1980s, hybridoma technology transitioned from research to commercial applications, with the first monoclonal antibody produced via this method receiving regulatory approval. Muromonab-CD3 (Orthoclone OKT3), a murine monoclonal antibody targeting the CD3 receptor on T cells, was approved by the U.S. Food and Drug Administration in 1986 as the inaugural therapeutic monoclonal antibody for preventing acute kidney transplant rejection. This milestone underscored the technology's clinical potential, despite challenges like immunogenicity from murine origins. To address immunogenicity issues associated with fully murine antibodies, researchers in the developed human-mouse heterohybridomas by fusing human B lymphocytes with mouse myeloma cells, enabling the production of monoclonal antibodies with reduced risk of immune reactions in patients. These advancements, exemplified by early stable heterohybridoma lines secreting antibodies against specific antigens like or proteins, marked a critical step toward more tolerable therapeutic options.

Principles

Core Concept of Cell Fusion

Hybridoma technology centers on the creation of a stable hybrid cell line through the fusion of an antigen-specific B , which secretes targeting a particular , with an immortal myeloma that does not produce immunoglobulins. This yields a hybridoma that inherits the B 's capacity for specific antibody production while acquiring the myeloma 's indefinite proliferative potential, enabling continuous and large-scale antibody secretion. The approach, developed by Georges Köhler and , revolutionized the production of monoclonal antibodies by overcoming the finite lifespan of primary B cells. The core of involves inducing the merging of the two distinct s' plasma membranes to form a single entity. (PEG) is a widely used chemical agent that promotes fusion by creating a dehydrating environment around the s, which draws the membranes into close contact and facilitates the intermixing of lipid bilayers through volume exclusion effects. Electrofusion represents an alternative physical method, where brief electric pulses generate transient pores in the membranes, allowing cytoplasmic contents to blend while preserving viability. Both techniques ensure efficient formation, typically at rates of 1 in 10^4 to 10^5 s, though optimization varies by . At the genetic level, the resulting hybridoma is initially near-tetraploid, combining the diploid genomes of the parental cells, but it frequently experiences chromosomal instability, leading to selective loss and stabilization at a pseudodiploid state. Crucially, the immunoglobulin loci from the —encoding the heavy and light chains for antigen-specific antibodies—are preferentially retained and actively transcribed, while non-essential B cell chromosomes may be eliminated. This selective retention, coupled with the B cell's metabolic contributions like functional HGPRT for HAT resistance and the myeloma's proliferative potential and growth resilience, underpins the hybridoma's ability to perpetually express monoclonal antibodies without losing specificity.

Mechanism of Antibody Production

In hybridoma cells, the activation of -derived genes ensures the continuous production of specific heavy and light immunoglobulin chains. The immunized contributes its , which includes rearranged variable region genes for a single specificity, allowing transcription and of the corresponding heavy and light chain mRNAs in the hybridoma . This genetic contribution from the overrides the myeloma cell's non-productive immunoglobulin loci, leading to the exclusive synthesis of monoclonal antibodies with predefined recognition. The synthesized heavy and light chains are produced independently on polysomes attached to the rough (), where the polypeptide backbones are assembled into nascent immunoglobulin molecules. These chains pair through bonds to form complete tetramers (two heavy and two light chains), a process facilitated by chaperone proteins like BiP to ensure proper folding. The assembled immunoglobulins then undergo post-translational modifications, including N-linked in the Golgi apparatus, where carbohydrate moieties such as and are added to enhance stability and effector functions. Mature monoclonal antibodies are transported from the Golgi to secretory vesicles and released via at the plasma membrane, resulting in the continuous secretion of identical molecules specific to a single without the need for further antigenic stimulation. This process yields high-titer production, typically 10-100 μg per 10^6 cells per day in . The hybridoma's is maintained through initial selection in HAT (hypoxanthine--thymidine) medium, which exploits the HGPRT deficiency in myeloma parent cells; blocks de novo synthesis, while only hybridomas, inheriting functional HGPRT from B cells, utilize the salvage pathway to survive and proliferate.

Methods and Procedures

Immunization and Cell Preparation

The production of monoclonal antibodies via hybridoma technology begins with the of animals to elicit a robust humoral against a specific target . Typically, female mice aged 6-8 weeks are selected due to their syngeneic compatibility with common myeloma cell lines. The initial involves subcutaneous injection of the (e.g., 100-200 µg) emulsified in complete (CFA) to enhance by stimulating a strong T-cell dependent response and depot effect at the injection site. Subsequent booster injections are administered to amplify the antibody response and increase the frequency of antigen-specific B cells. A second booster, typically 4-6 weeks after the initial dose, uses the antigen mixed with incomplete (IFA) for continued stimulation without the intense of CFA. Additional boosters (third and fourth) may employ the antigen alone, either subcutaneously or intravenously, with intervals of 2-4 weeks. Serum titers are monitored 7-10 days post-booster via enzyme-linked immunosorbent assay (ELISA), where antigen-coated plates detect specific IgG levels, aiming for titers exceeding 1:10,000 to confirm adequate immune activation before proceeding. Three to four days following the final booster, the is harvested from the immunized mouse to isolate activated B s at peak plasmablast frequency. The mouse is euthanized humanely, the excised aseptically, and dissociated into a single- suspension using a cell strainer and RPMI-1640 medium, yielding approximately 1-2.5 × 10^8 viable splenocytes per after and erythrocyte . These splenocytes, enriched for antigen-specific B lymphocytes, are then ready for . Parallel to B cell preparation, myeloma cells are cultured to serve as immortal fusion partners. The SP2/0-Ag14 cell line, derived from BALB/c mice and deficient in hypoxanthine-guanine phosphoribosyltransferase (HGPRT), is commonly used and maintained in HAT-sensitive medium (RPMI-1640 supplemented with 10-20% fetal calf serum, L-glutamine, and antibiotics) to ensure selective survival post-fusion, as unfused myeloma cells cannot grow in HAT due to their metabolic deficiency. Cells are expanded in logarithmic phase, harvested by centrifugation, and counted to achieve a 5:1 to 10:1 splenocyte-to-myeloma ratio for optimal fusion efficiency.

Fusion, Selection, and Screening

The process of creating hybridoma cells begins with the of antigen-specific B s, typically derived from the of an immunized animal, and immortal myeloma s. Two primary methods are employed for this : chemical using () and electrical via electrofusion. In PEG-mediated , a concentrated of (usually 40-50% w/v) is added to the mixed suspension, inducing of head groups and transient asymmetry that promotes -to- contact and merger; this method is straightforward and widely used but can be cytotoxic and lead to non-specific fusions. Electrofusion, on the other hand, applies high-intensity electric pulses (typically 1-2 kV/cm) to create transient pores in membranes, facilitating controlled with higher efficiency (up to 10-fold greater than in some protocols) and reduced , though it requires specialized . Both techniques are typically performed at a B -to-myeloma of 5:1 to 10:1, with efficiencies ranging from 1 in 10^4 to 1 in 10^5 s depending on viability and conditions. Following fusion, hybridoma cells are selectively cultured in hypoxanthine-aminopterin-thymidine (HAT) medium to eliminate unfused parental cells. Aminopterin in HAT blocks the de novo synthesis of purines and pyrimidines by inhibiting dihydrofolate reductase, forcing cells to rely on the salvage pathway enzymes hypoxanthine-guanine phosphoribosyltransferase (HGPRT) and thymidine kinase (TK). Myeloma cells, engineered to lack HGPRT (e.g., via 8-azaguanine selection), cannot utilize the salvage pathway and thus die within 7-10 days, while non-fused B cells, though possessing HGPRT, fail to proliferate indefinitely due to their finite lifespan. Only hybridomas, inheriting HGPRT from B cells and immortality from myeloma cells, survive and expand, typically achieving visible growth in 10-14 days post-fusion. Surviving hybridomas are then cloned and screened to isolate those secreting antibodies of desired specificity. Limiting dilution is the standard cloning method, where cells from HAT-selected cultures are serially diluted (e.g., to 0.5-1 per well in 96-well plates) and cultured until single colonies form, ensuring monoclonality; this process is repeated 2-3 times for stability. Screening for specificity is commonly performed using enzyme-linked immunosorbent (ELISA), where hybridoma supernatants are tested against the immunizing coated on plates; positive clones show binding detected via secondary antibodies conjugated to enzymes like , with absorbance measured at 450 nm. This step identifies -specific secretors amid the low yield of functional hybridomas (often <1% of fusions). Finally, positive clones undergo isotyping to determine the antibody class (e.g., IgG1, IgM) and subclass, typically via isotype-specific ELISA kits or flow cytometry with fluorescent anti-isotype antibodies, which informs potential applications based on effector functions. Productivity assessment evaluates antibody secretion rates by quantifying titers in supernatants, often using quantitative ELISA or bio-layer interferometry, to select high-yield clones (e.g., >10 μg/mL/day) for further development.

Cloning and Scale-Up

Once positive hybridomas have been identified through screening, they undergo to establish stable monoclonal lines that produce a single antibody specificity. The primary methods for achieving monoclonality include limiting dilution and soft agar cloning, often repeated over multiple rounds to verify stability and eliminate non-producing or heterogeneous subpopulations. In the limiting dilution technique, hybridoma cells are serially diluted and plated in multi-well plates at densities aiming for approximately one cell per well, typically 0.3 to 1 cell per well, allowing individual clones to proliferate without interference from neighboring cells. This probabilistic approach ensures that resulting colonies derive from progenitor cells, with at lower densities (e.g., 0.3 cells per well) used for confirmation. The soft method, an alternative, embeds cells in a semi-solid medium overlaid on a base, where only anchored hybridomas form visible from cells, facilitating their based on colony . These cloning steps typically take 2-4 weeks per round, with stability assessed by consistent antibody production over passages. Following cloning, selected hybridoma lines are cryopreserved to create secure cell banks for long-term viability and . A master (MCB) is established from the initial clonal population, serving as the definitive source material frozen in multiple aliquots using cryopreservatives like 10% (DMSO) in , stored at -196°C in vapor phase. Working s (WCBs) are then derived from the MCB through limited expansion and similarly cryopreserved, providing the immediate supply for production while minimizing ; each bank undergoes viability testing (e.g., >80% post-thaw recovery) and characterization for identity, purity, and productivity. This banking strategy ensures reproducible antibody production and supports good manufacturing practices for therapeutic applications. Scale-up of antibody production occurs either in vitro or in vivo to meet demand, with in vitro methods preferred for purity and scalability in modern applications. In vitro expansion involves culturing hybridomas in , such as hollow-fiber systems that mimic networks for high-density growth (up to 10^8 cells/mL) or stirred-tank fermenters for larger volumes (liters to thousands), often in serum-free media to enhance yield and reduce contaminants; these systems can produce 1-10 g/L of antibody over 2-4 weeks. In vivo scale-up, historically common, entails of 10^6 to 10^7 hybridoma cells into syngeneic mice, leading to ascites tumor formation and fluid accumulation (5-20 mL per mouse) containing 1-10 mg/mL antibody after 10-21 days, harvested via tapping. approaches are favored for therapeutic-grade production due to higher consistency and avoidance of animal variability. Purification of monoclonal antibodies from hybridoma-derived supernatants or fluid primarily relies on using or protein G resins, which specifically bind the region of (IgG) subclasses with high affinity (Kd ~10^-8 M). The process involves loading the clarified harvest onto a column equilibrated in neutral buffer ( 7-8), washing to remove unbound proteins, and eluting the antibody with low- ( 2.5-3.0), followed by immediate neutralization to 7-8 to prevent aggregation; yields typically exceed 80-95% with purity >95%. is effective for most human and mouse IgG1-3, while protein G offers broader binding for other subclasses, often combined as protein A/G for hybridoma products. Additional polishing steps, such as ion-exchange chromatography, may follow if needed, but affinity capture establishes the core purification platform.

Applications

In Biomedical Research

Hybridoma-derived monoclonal antibodies have played a pivotal role in identifying cell surface markers, particularly in , where they enabled the classification of leukocyte subsets through the (CD) system. For instance, the OKT4 antibody, produced via hybridoma technology, specifically recognizes the CD4 antigen on helper T cells, facilitating the separation of functional T cell subsets and advancing understanding of immune responses. Similarly, the OKT8 antibody targets the CD8 antigen on cytotoxic T cells, allowing precise delineation of T cell populations essential for studying cellular immunity. These antibodies, generated by fusing immunized B cells with myeloma cells, have been instrumental in workshops that standardized CD nomenclature, transforming immunological research by providing tools for and functional assays. In protein purification and analysis, hybridoma-derived monoclonal antibodies are widely employed in immunoprecipitation (IP) techniques to isolate specific proteins from complex mixtures, enabling downstream studies of protein interactions and structures. Complementing this, monoclonal antibodies facilitate , which identifies precise antigenic determinants on proteins, as demonstrated in studies characterizing monoclonal antibodies against proteins where epitope specificity distinguishes intra- from extracellular binding sites. Such applications have been crucial for dissecting protein function without , providing researchers with reliable reagents for biochemical assays. Hybridoma technology has contributed to developing animal models for diseases by producing neutralizing monoclonal antibodies that block pathogen entry or modulate immune responses in vivo. Neutralizing antibodies against filoviruses, such as those targeting virus glycoproteins, have protected from lethal challenges, mimicking progression and evaluating intervention strategies. These models have informed studies, revealing mechanisms of and immunity in controlled settings. By providing antigen-specific reagents, hybridoma-derived antibodies enhance the fidelity of such models, bridging and translational insights. Furthermore, in vaccine design, hybridoma-derived monoclonal antibodies aid characterization by mapping immunodominant epitopes, guiding the selection of candidates that elicit protective responses. For virus, monoclonal antibodies specific to the E2 have defined conformational epitopes critical for viral neutralization, informing the development of differentiating infected from vaccinated animals () strategies. This epitope-focused approach ensures vaccines target conserved regions, improving efficacy against variants and accelerating preclinical evaluation.

In Diagnostics and Therapeutics

Hybridoma technology has revolutionized diagnostics by providing monoclonal antibodies (mAbs) essential for (IHC), where they detect tumor markers such as HER2 in tissues, enabling precise pathological classification and guiding treatment decisions. These mAbs bind specifically to antigens on fixed tissue sections, allowing visualization of abnormal cell populations that indicate . In the 1980s, early applications of hybridoma-derived mAbs in marked a shift toward more reliable tumor identification. In flow cytometry diagnostics, hybridoma-produced mAbs form multi-color panels that target cell surface markers like and CD45, facilitating the rapid of leukemias and lymphomas in clinical samples. This technique analyzes thousands of cells per second, distinguishing malignant from normal populations based on expression patterns, which is critical for initial diagnosis and monitoring . For , standard IHC staining protocols using these mAbs involve deparaffinizing tissue sections, blocking non-specific sites, incubating with the primary mAb, followed by a secondary enzyme-linked and chromogenic substrate to produce visible signals for localization in tumors. Therapeutically, hybridoma technology laid the foundation for mAbs like rituximab, a chimeric antibody targeting on B cells, approved for and , where it induces and . Similarly, infliximab, a chimeric anti-TNF-α antibody derived from hybridoma-generated murine sequences, treats autoimmune conditions such as and by neutralizing inflammatory cytokines. To mitigate immune reactions against murine components, such as responses, early hybridoma mAbs evolved into chimeric constructs (retaining murine variable regions with human constant regions) and further into humanized versions with minimized murine content, improving and in long-term therapies.

Advantages, Limitations, and Advances

Advantages and Challenges

Hybridoma technology offers several key advantages in the production of monoclonal (mAbs), primarily stemming from its ability to generate highly specific and consistent antibody clones. The method produces mAbs that target a single with high specificity, enabling precise applications in research and diagnostics. Once stable hybridoma cell lines are established, they provide batch-to-batch consistency through reproducible antibody production, minimizing variability compared to polyclonal sources. Additionally, for initial discovery, the technology is cost-effective, as immortalized hybridomas can be cryopreserved and scaled up indefinitely without repeated animal immunizations. Despite these benefits, hybridoma technology faces significant challenges that limit its efficiency and applicability. The process is time-intensive, often requiring 6 to 9 months for , fusion, selection—such as screening—and cloning to isolate viable hybridomas. It relies heavily on animal models, typically mice or rats, for B-cell , raising ethical concerns over and the need for their sacrifice. This animal dependence also introduces risks of disease transmission from animals to production lines. Further drawbacks include the potential for generating low-affinity antibodies, particularly against small peptides or complex antigens, due to screening biases that favor high-secretion but lower-quality clones. Murine-derived mAbs pose risks in human therapeutic use, as they can elicit responses, reducing efficacy and causing adverse reactions. Hybridoma stability is another issue, with fused cells often exhibiting genetic instability that leads to loss of over time, and fusion efficiency remains low, with over 99% of cells typically dying post-fusion. These factors contribute to variable rates and overall reduced yields in production.

Recent Developments and Alternatives

In the , hybridoma has seen significant enhancements to overcome traditional limitations such as low yield and . Single B cell cloning represents a key advance, allowing direct isolation and immortalization of individual antigen-specific B cells without initial , enabling faster discovery of high-affinity monoclonal antibodies with superior functional properties compared to classical hybridomas. CRISPR/Cas9 editing has further revolutionized hybridoma production by enabling precise genetic modifications, such as introducing mutations for increased antibody yields or functional diversification, including site-specific conjugation for enhanced therapeutic applications. Additionally, trioma , involving the of heteromyeloma cells with human lymphoid cells, has facilitated the generation of fully lines, reducing risks associated with murine hybrids and supporting clinical translation. Integration of next-generation sequencing (NGS) with hybridoma workflows has accelerated discovery by enabling high-throughput sequencing of immunoglobulin genes from hybridoma clones, allowing rapid identification and optimization of sequences even from unstable cell lines. This approach, often combined with barcoding, supports the digitization of diverse repertoires, streamlining screening and recombinant expression while preserving the natural maturation of B cell-derived antibodies. As of 2025, further innovations include targeted techniques for antibody-secreting cells, which improve hybridoma by selecting and fusing terminally differentiated cells, potentially shortening production timelines and increasing yields. and AI-driven screening have also advanced hybridoma workflows, enabling higher throughput and reduced animal dependency. Emerging alternatives to hybridoma technology, such as phage display and single-cell RNA sequencing (RNA-seq), have gained prominence for generating recombinant antibodies without relying on animal immunization, thereby reducing ethical concerns and accelerating development timelines. Phage display libraries express antibody fragments on bacteriophage surfaces for high-throughput selection, offering scalability and ease of engineering that surpass hybridoma's labor-intensive fusion and cloning steps. Single-cell RNA-seq, paired with microfluidic isolation, captures full-length antibody sequences from individual B cells, enabling de novo design of human-like antibodies with minimal animal use and enhanced diversity screening. Despite these recombinant methods dominating routine discovery, hybridoma technology retains an enduring role in complex glycoengineering, where its mammalian expression system naturally produces antibodies with precise glycosylation patterns critical for effector functions like antibody-dependent cellular cytotoxicity, which are challenging to replicate in non-mammalian platforms.

References

  1. [1]
    Continuous cultures of fused cells secreting antibody of predefined ...
    Aug 1, 1975 · Continuous cultures of fused cells secreting antibody of predefined specificity. G. KÖHLER &; C. MILSTEIN. Nature volume 256, pages 495– ...Missing: original | Show results with:original
  2. [2]
    Hybridoma technology; advancements, clinical significance, and ...
    Oct 18, 2021 · Hybridoma technology is one of the most common methods used to produce monoclonal antibodies. In this process, antibody-producing B lymphocytes are isolated ...
  3. [3]
  4. [4]
  5. [5]
    The Nobel Prize in Physiology or Medicine 1984 - Press release
    Köhler and César Milstein in 1975 introduced the so-called hybridoma technology for production of monclonal antibodies. The principle features of the ...Missing: paper | Show results with:paper
  6. [6]
    The Story of César Milstein and Monoclonal Antibodies
    Milstein and Köhler wondered whether one way to resolve some of the difficulties encountered by previous researchers was to fuse a normal B cell from the spleen ...Missing: challenges | Show results with:challenges
  7. [7]
    The Nobel Prize in Physiology or Medicine 1984 - NobelPrize.org
    The Nobel Prize in Physiology or Medicine 1984 was awarded jointly to Niels K. Jerne, Georges JF Köhler and César Milstein for theories concerning the ...
  8. [8]
    Muromonab-CD3 - an overview | ScienceDirect Topics
    Muromonab-CD3 (Orthoclone OKT3®), the first murine antibody to be approved in 1986, was a mouse hybridoma MAb that was identified, developed, and manufactured ...
  9. [9]
    A human-mouse hybridoma producing monoclonal antibody against ...
    The establishment of human-mouse hybridomas, which produced anti-semen antibodies as produced in sterile women, became essential.Missing: immunogenicity | Show results with:immunogenicity
  10. [10]
    Long-term production of human monoclonal antibodies by human ...
    We have stabilized two such heterohybridomas over one year period and induced the production of κ-bearing and λ-bearing human mAb, respectively. Increased ...Research Report · Abstract · AbbreviationsMissing: immunogenicity | Show results with:immunogenicity
  11. [11]
    Use of Human Hybridoma Technology To Isolate Human ... - NIH
    This chapter reviews recent efforts to develop and employ novel methods for the efficient generation of human hybridomas secreting human mAbs for clinical use.
  12. [12]
    Specific suppressive factors produced by hybridomas derived from ...
    Aug 1, 1978 · In parallel with the losses was an alteration in chromosome number from near-tetraploid, soon after cell fusion, to near- diploid. Preliminary ...
  13. [13]
    INDEPENDENT SYNTHESIS OF LIGHT AND HEAVY CHAINS - NIH
    The ratio of heavy and light chains in the two cell lines was approximately the same, and the size of the intracellular pool of immunoglobulin was proportioned ...
  14. [14]
    IMMUNOGLOBULIN SYNTHESIS AND SECRETION - NIH
    The subcellular sites of synthesis and route of intracellular transfer of immunoglobulin G (IgG) have been investigated by electron microscope radioautography.
  15. [15]
    Generation of Hybridomas: Permanent Cell Lines Secreting ... - NCBI
    It has also been possible to genetically replace much of the mouse mAb-producing genes with human sequences, reducing the immunogenicity of mAb destined for ...
  16. [16]
    Hybridoma Technology: Steps, Protocol, and Application
    Hybridoma technology is a well-established, cost-effective, and widely used method that produces highly specific and remarkably uniform monoclonal antibodies.
  17. [17]
    Generation of Murine Monoclonal Antibodies by Hybridoma ... - NIH
    Jan 2, 2017 · Step 1 is an appropriate immunization of the animal and step 2 is the fusion of B lymphocytes with immortal myeloma cells in order to generate ...Missing: mechanism | Show results with:mechanism
  18. [18]
    Statistical Assessment of Hybridoma Monoclonality After Subcloning ...
    Thus, many laboratories still use subcloning by the technique of limiting dilution (2,3) for establishing the monoclonality of their hybridomas. Although the ...
  19. [19]
    Single-Cell Cloning of Hybridoma Cells by Limiting Dilution
    Limiting dilution is a highly preferred technique for the identification of individual hybridoma clones, where a set of candidate hybrid cells are plated in a ...
  20. [20]
    Antibody Hybridoma Core - Cloning, Subcloning, Isotyping and ...
    Cloning and subcloning are performed using the limiting dilution technique; hybrids are cloned at one cell per well and subcloned at 0.3 cell per well in one ...Missing: soft agar
  21. [21]
    Monoclonal Antibodies: Production, Advantages and Limitations
    Two techniques are commonly employed for cloning hybrid cells-limiting dilution method and soft agar method. Limiting dilution method: In this procedure, the ...
  22. [22]
    Selective cloning of hybridoma cells for enhanced immunoglobulin ...
    The dilutionistatistical approaches (limiting dilution, soft agar) provide a rel- atively simple approach for producing multiple num- bers of cloned cultures ...
  23. [23]
    Hybridoma Technology: Monoclonal Antibody Production
    Hybridoma technology is used to produce monoclonal antibodies (mAbs). It involves fusing B cells, which produce antibodies, with myeloma cells.
  24. [24]
    Master Cell Banking - Leinco Technologies
    The master cell bank is created from a single cell source optimized for monoclonal antibody production. Cells are expanded to a level required to freeze an ...<|separator|>
  25. [25]
    [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 ...
  26. [26]
    Large-Scale Production of Monoclonal Antibodies - NCBI - NIH
    Biologic behavior of a hybridoma cell line is very important in determining whether in vitro culture will be successful or the ascites method must be used.
  27. [27]
    Evaluation of hollow fiber bioreactors as an alternative to murine ...
    The objective of this study was to compare monoclonal antibody production in hollow fiber bioreactor systems and murine ascites to determine the feasibility ...
  28. [28]
    Overcome ascites production headaches: in vitro bioreactors deliver ...
    Hybridoma cells are introduced and grow to become an ascites tumor, which releases mAbs into the ascites fluid. The mAb-enriched ascites fluid is periodically ...
  29. [29]
    Antibody Purification Using Protein A/G | Thermo Fisher Scientific - ES
    Antibody purification involves isolation of antibody from serum (polyclonal antibody), ascites fluid, or from the culture supernatant of a hybridoma cell ...
  30. [30]
    Protein A/G Affinity | Bio-Rad
    Protein A and G chromatography media are used in antibody purification due to the high binding affinity and specificity with the antibody Fc region.
  31. [31]
    Purification of Monoclonal Antibody from Hybridoma Supernatant ...
    Monoclonal antibodies obtained from hybridoma cell supernatant were purified using a protein A/G agarose resin bead affinity chromatography column.
  32. [32]
    Purification of Monoclonal Antibodies Using Chromatographic ... - NIH
    A 2020 study by Akhtar evaluated protein A affinity chromatography for purifying a recombinant monoclonal antibody from cell culture using methods like SDS-PAGE ...
  33. [33]
  34. [34]
    Monoclonal Antibody Purification: Process, Methods | Sino Biological
    Protein A affinity chromatography remains the gold standard for this step, leveraging its high specificity for the Fc region of IgG antibodies to capture most ...
  35. [35]
    A toolbox of immunoprecipitation-grade monoclonal antibodies to ...
    The critical steps involved in the generation and distribution of high-quality PCRP mAbs include antigen production; hybridoma production; primary validation ...
  36. [36]
    Production and characterization of monoclonal antibodies sensitive ...
    In terms of epitope mapping, the most obvious distinction for an mAb that recognizes a membrane protein is whether it binds to intra- or extracellular regions ...Results · Antibody Screening · Epitope Mapping
  37. [37]
    Novel neutralizing monoclonal antibodies protect rodents against ...
    In this study, we characterized 2 novel neutralizing MAbs against EBOV and MARV, and show that both MAbs protect against lethal EBOV or MARV challenge in mice ...
  38. [38]
    Characterization of monoclonal antibodies that specifically ...
    Jul 18, 2022 · This study provides new insights into the antigenic structure of CSFV E2 and E rns and the differentiating mAbs will contribute to the development of a ...
  39. [39]
    The unique monoclonal antibodies and immunochemical assay for ...
    Feb 17, 2024 · New anti-HER2 antibodies were generated using the hybridoma cells technology utilizing the previously described method. The selection of cells ...
  40. [40]
    Laboratory Evaluation of Tumor Biomarkers - StatPearls - NCBI - NIH
    Oct 28, 2023 · [22] Immunohistochemistry: Immunohistochemistry is a type of immunoassay employed when tumor biomarkers must be detected in solid tissue ...
  41. [41]
    Newer developments in immunohistology - PMC - PubMed Central
    Immunohistochemistry has traditionally been used to demonstrate tissue proteins, largely as markers of a particular cell or tissue type. Increased specificity ...
  42. [42]
    Flow Cytometry in the Diagnosis of Leukemias - NCBI
    Flow cytometry can quickly identify the abnormal cell population, characterize its phenotype, give lineage classification, make the diagnosis, or narrow down ...Abstract · INTRODUCTION · PRINCIPLES AND KEY...
  43. [43]
    Advances and issues in flow cytometric detection of ... - NIH
    Leukemia cells can be recognized by virtue of unique cell marker combinations, visualized with monoclonal antibodies conjugated and detected by flow cytometry.
  44. [44]
    Immunohistochemistry for Pathologists: Protocols, Pitfalls, and Tips
    On the other hand, monoclonal antibodies react to a single epitope in an antigen. They are obtained from a single clone of hybridoma, which produces antibodies.
  45. [45]
    How the discovery of rituximab impacted the treatment of B-cell non ...
    MoAb development began in 1975 by Köhler and Milstein with the use of hybridoma technology ... rituximab and also assigned the trade names Rituxan and Mabthera.
  46. [46]
    Understanding How Monoclonal Antibodies Work - StatPearls - NCBI
    Jun 26, 2023 · [5] Examples of chimeric mAbs include infliximab and rituximab.[1] Chimeric clones were followed by developing "humanization," a process ...
  47. [47]
    Therapeutic Antibodies in Medicine - PMC - PubMed Central - NIH
    Production of therapeutic antibody drugs has evolved from the use of murine antibodies, first created via hybridoma technology, to the latest humanized or human ...
  48. [48]
    Hybridoma technology a versatile method for isolation of ...
    May 27, 2020 · The antibodies isolated through hybridoma methodology preserves the native pairing of variable and constant regions gene combination, which ...Missing: retention | Show results with:retention
  49. [49]
    Hybridoma technology: is it still useful? - PMC - PubMed Central
    Mar 22, 2021 · Despite being an old technique with low complexity, hybridoma-based strategies have limitations that include the low efficiency on B lymphocyte- ...
  50. [50]
    Review Single B cell technologies for monoclonal antibody discovery
    For selection of immortalized HGPRT+, ASC-myeloma fusions are placed in medium containing hypoxanthine, aminopterin and thymidine (HAT medium); a first single ...<|control11|><|separator|>
  51. [51]
    Functional diversification of hybridoma-produced antibodies by ...
    We demonstrate a versatile CRISPR/HDR platform to rapidly engineer the constant immunoglobulin domains to obtain recombinant hybridomas.
  52. [52]
    High-volume hybridoma sequencing on the NeuroMabSeq platform ...
    Sep 27, 2023 · NeuroMabSeq is a concerted effort to determine and make publicly available hybridoma-derived sequences of monoclonal antibodies (mAbs) valuable to neuroscience ...Results · Bioinformatics Processing · Methods
  53. [53]
    Barcoded sequencing workflow for high throughput digitization of ...
    Thus, we present in this manuscript a barcoded sequencing workflow that utilizes NGS technology for the identification of antibody sequences in the hybridoma ...Technical Note · 2. Materials And Methods · 3. Results And DiscussionMissing: integration | Show results with:integration
  54. [54]
    High-throughput strategies for monoclonal antibody screening
    May 8, 2025 · New high-throughput technologies like antibody library display, single B cell antibody technologies, and single-cell sequencing have ...
  55. [55]
    Advancements in mammalian display technology for therapeutic ...
    Single B cell technologies have emerged as powerful tools for therapeutic antibody discovery, offering significant advantages over hybridoma technology in speed ...
  56. [56]
    Glycoengineering of Therapeutic Antibodies with Small Molecule ...
    Nov 4, 2021 · We review selected examples for the use of small molecule inhibitors of glycan biosynthesis for mAb glycoengineering, we discuss the potential advantages and ...