Skip to main content

High-throughput strategies for monoclonal antibody screening: advances and challenges

Abstract

Antibodies characterized by high affinity and specificity, developed through high-throughput screening and rapid preparation, are crucial to contemporary biomedical industry. Traditional antibody preparation via the hybridoma strategy faces challenges like low efficiency, long manufacturing cycles, batch variability and labor intensity. Advances in molecular biology and gene editing technologies offer revolutionary improvements in antibody production. New high-throughput technologies like antibody library display, single B cell antibody technologies, and single-cell sequencing have significantly cut costs and boosted the efficiency of antibody development. These innovations accelerate commercial applications of antibodies, meeting the biopharmaceutical industry's evolving demands. This review explores recent advancements in high-throughput development of antibody, highlighting their potential advantages over traditional methods and their promising future.

Introduction

Initially, antibodies were produced by stimulating animal immune systems with target antigens, which suffered from limitations such as inconsistent specificity and significant batch-to-batch variability [1]. Hybridoma technology addresses these challenges by fusing host spleen cells with myeloma cells to create hybridoma cells. which can continuously proliferate and produce large amounts of specific, high-affinity monoclonal antibodies in vitro. Monoclonal antibodies (mAb) are highly stable, reproducible, and specific, making them valuable for research, drug development, and diagnostics [2]. However, this technology is limited by low-throughput mAb production due to the complex and labor-intensive hybridoma cell fusion process, along with time-consuming screening and dilution steps [3, 4].

High-throughput mAb production technologies utilize advanced biotechnologies, including high-throughput screening, single-cell sequencing, and antibody library display, among others, to quickly obtain coding sequences for target antibody variable regions [5, 6]. Furthermore, these technologies enable the precise optimization, modification, and expression of antibodies through genetic engineering techniques. Through these methods, high-affinity and highly specific antibodies can be rapidly and massively produced [7]. With the deepening of biomedical research and clinical applications, high-throughput mAb production technologies are expected to become the mainstream method for antibody development, bringing further breakthroughs and progress in the fields of disease diagnosis, treatment, and drug discovery [8, 9]. This review summarizes recent advancements in high-throughput monoclonal antibody screening, highlighting their advantages over traditional methods and discussing future applications in antibody production.

Antibody library display technology

Due to somatic DNA recombination, the mammalian B-cell antibody repertoire can generate a diversity of 1012 to 1018 unique antibodies to recognize and combat a wide range of microbial pathogens [10]. The antibody library display technology presents antibody fragments from all variable regions of the antibody repertoire on the surfaces of vectors (such as phages, cells, or ribosomes, etc.), enabling the generation of an antibody library. Through multiple rounds of specific selection and screening parameter optimization, it facilitates the rapid and efficient production of antibodies targeting specific antigen conformations or epitopes [11, 12]. The high-throughput screening process for antibody library display technology is shown in Fig. 1

Fig. 1
figure 1

The high-throughput antibody production scheme of the antibody library display technology. Antibody libraries from immunized animals and humans are displayed on a variety of vectors (e.g. phage, yeast, ribosomes, etc.) to construct diverse antibody libraries, and high-affinity binding vectors are isolated through multiple rounds of iterative screening. The gene sequences of the variable regions of the antibodies were determined by ELISA identification and sequencing analysis. The gene sequences of the variable regions are integrated into the designated expression vectors for antibody production, followed by downstream characterization and analysis

Phage display antibody library technology

Phage display antibody library technology is one of the most widely used methods for antibody development. Clackson et al. first demonstrated that the variable regions of antibodies could be displayed on the phage surface [13]. Additionally, Barbas III and Lerner enabled the construction of large combinatorial antibody libraries by fusing antibody gene fragments to phage coat proteins, allowing antibodies to be displayed on the phage surface [14]. By incubating the phage library with antigens and performing panning, phages with specific binding affinities are selected. Finally, high-affinity antibody genes are obtained through positive phage clone identification and DNA sequencing, providing a basis for further genetic engineering and antibody expression [15, 16].

Initially, antigen presentation and selection were carried out using solid-phase carriers such as ELISA plates and magnetic beads. With the development of technology, the introduction of automated microplate-based screening, magnetic bead processors, and robotic workstations has optimized the workflow and enhanced throughput [17,18,19]. Additionally, array-based formats further improve screening efficiency [20]. For instance, Pérez-Gamarra et al. reported an in-well array assay for multiparametric screening of phage display antibody libraries [21]. Fluorescence-activated cell sorting (FACS) and microfluidics integrated with phage display antibody libraries have further enhanced automation and high-throughput screening [22, 23]. Hemadou et al. employed multiplex flow cytometry for high-throughput screening of scFvs with precise binding specificity, reducing the number of screening rounds and simultaneously obtaining a broader diversity of clones [24]. Philpott et al. designed the μCellect platform, which utilizes microfluidic selection to identify picomolar-affinity antibodies within just two rounds of screening [25]. Additionally, studies have reported that combining phage antibody display with FACS-based yeast surface display not only enables the generation and screening of large libraries but also allows for flexible quantification and efficient enrichment of antibodies with specific binding properties [26, 27]. To reduce multiple rounds of screening and improve efficiency. Keyser et al. employed biosensors to present antigens to nanobody-displaying phages within wells, leveraging an automated Octet bio-layer interferometry sensor for high-throughput selection with precise control over each step [28]. Hornsby described an optimized automated phage display pipeline that generated approximately 3,000 sequenced antigen-binding domains with high affinity, significantly increasing the throughput of antibody discovery [29].

To enhance parallel analysis capacity and increase screening diversity and precision, next-generation sequencing (NGS) has been integrated into phage display selection. Lee et al. constructed a single-domain antibody (sdAb) phage display library and employed NGS for high-throughput analysis, validating library diversity and identifying multiple high-affinity sdAbs [30]. Sasso et al. co-incubated phages with activated lymphocytes to select clones binding to CD27, BTLA, and TIGIT, followed by NGS-based VH region sequencing, enabling large-scale parallel screening [31]. Nakada-Masuta et al. integrated selection pressure with NGS analysis to effectively reduce expression bias and maintain antibody library diversity [32]. Furthermore, Barreto et al. developed an NGS-assisted antibody discovery platform, demonstrating that rare clones identified by NGS exhibited superior affinity and specificity compared to high-abundance clones detected by Sanger sequencing [33]. These advancements have significantly enhanced the efficiency of phage display selection, driving its widespread application in antibody development for cancer, viral infections, and autoimmune diseases [34,35,36,37,38].

Cell display antibody library technology

Cell display antibody library technology utilizes engineered cell surface display systems to present antibody fragments, providing an alternative to phage display. The major cell display platforms include yeast display, bacterial cell display, and mammalian cell display, each offering distinct advantages in terms of expression efficiency and screening capabilities [39].

Yeast display antibody library technology

Yeast display antibody library technology is a well-established method for displaying antibody fragments on the yeast cell surface and constructing antibody libraries. It typically employs high-throughput screening methods, such as FACS or microfluidics, to isolate high-affinity antibody clones for further analysis or subsequent applications [40, 41]. After multiple rounds of screening and amplification, high-affinity antibody clones can be obtained. The automation of high-affinity antibody characterization, sequencing, and expression, along with engineering advancements, has significantly accelerated the antibody production process [42]. Yeast’s eukaryotic nature provides an ideal environment for antibody display, facilitating proper folding and post-translational modifications like glycosylation, which enhances the solubility and expression of disulfide-bonded and natively glycosylated antibodies or antibody fragments [43, 44]. Bowley et al. directly compared the same HIV-1 immune scFv cDNA library expressed in both phage and yeast display vectors, finding that yeast display yielded three times more specific scFv clones than phage display, and included all the clones recovered via phage [45]. This result demonstrates that yeast display can capture a broader and more functional diversity of antibodies. In addition, this technology based on cell surface antibody library display (including the mammalian cell surface display antibody library discussed below) integrates high-throughput screening and analysis methods, allowing for the high-throughput screening and characterization of tens of thousands of candidates [46]. This capability offers significant benefits in affinity, discovery of rare antibody clones, and improved cost-effectiveness and efficiency. Holliger et al. recently utilized the Illumina HiSeq platform to sequence yeast antibody libraries, which greatly accelerated the identification of high-affinity candidate sequences, screening for 108 antibody-antigen interactions within 3 days [47]. It is important to note, however, that while the identification process using NGS data can be accomplished quickly, the subsequent steps (e.g., gene synthesis, cloning, expression, and functional validation) are still time- and resource-intensive. The entire process of physically synthesizing and validating high-affinity antibodies typically takes several weeks, and cloning for synthesis and expression is costly [48]. This is largely due to the lack of cost-effective, high-throughput gene synthesis and expression solutions, which limits the scalability of the antibody discovery pipeline.

Mammalian cell display antibody library technology

Mammalian cell display antibody library technology involves inserting the target antibody gene into an expression vector and introducing it into mammalian cells (like CHO or HEK293) via transfection or viral infection. This allows the surface expression of the exogenous proteins, creating display libraries [49]. The high-throughput antibody screening of mammalian cell display following library construction are similar to yeast display. The mammalian cell display technology, owing to its endogenous eukaryotic secretion mechanism, partially mitigates the issues of low effective activity and misfolding that arise from the absence of post-translational modifications inherent in phage display. Theoretically, complex and highly stable antibodies (including Fab, scFv, and full-length IgG antibodies) can be displayed directly on the cell surface using mammalian cell surface display systems [50]. Robertson et al. developed an antibody discovery platform based on mammalian cell display technology for the natural conformation of membrane proteins, thereby increasing the opportunities to obtain high-affinity antibodies [51]. Furthermore, mammalian cell antibody display libraries can be designed in a secreted antibody format to enhance the efficiency of antibody screening. The secreted antibody repertoire has been integrated with various high-throughput auxiliary screening techniques, including gel microdroplet-fluorescence-activated cell sorting, droplet microfluidics, nanopores, and microarrays, among others [52]. Recently, Doerner et al. created a display-secretion switch system for pre-enriching highly manufacturable antibodies and conducting functional screening [53]. In the aforementioned context, antibody therapeutics utilizing mammalian cell display technology have undergone significant development in recent years [54,55,56]. However, it should be noted that conventional mammalian cell display antibody libraries struggle to meet all expectations of an ideal library. Researchers may need to employ targeted strategies to enhance library quality, thereby increasing the likelihood of identifying ideal antibodies during the screening process. For instance, enhancing the library's capacity by utilizing immunized animals, and achieving stable integration and transcriptional normalization of library mutations through programmable nucleases such as CRISPR/Cas9 and TALENs [57, 58]. Furthermore, this technology involves cell transfection and transmembrane secretion during library construction, which still requires further optimization to enhance the overall efficiency of antibody development.

Bacterial cell display antibody library technology

Bacterial cell display antibody library technology provides a simple and rapid alternative for the efficient transformation and functional screening of large peptide libraries. However, in bacterial display systems, proteins must traverse two membranes or contend with a thick peptidoglycan cell wall, which poses significant challenges for the display of large proteins or antibodies [59]. Although Francisco and Lee et al. demonstrated that scFv fragments can be functionally displayed on the surface of E. coli and sorted using flow cytometry, the outer membrane remains a barrier [60, 61]. As a result, only a few bacterial display systems can effectively present antibody fragments, limiting yield and complicating large-scale production. Mazor and Lombana et al. innovatively described a bacterial cell antibody library method in which IgG antibodies were immobilized on the periplasm-facing inner membrane. Using FACS, they successfully selected specific antigen binders, obtaining a diverse set of high-affinity IgG clones within 3–4 weeks [62, 63]. The dominant screening method for bacterial cell display antibody libraries is FACS, as it enables the assessment of expression levels, antigen binding, and estimation of antibody affinity [64]. While FACS excels in throughput and high-affinity binder isolation, it presents technical challenges when screening highly diverse libraries. To address this, researchers have integrated phage display as a preselection step to reduce library size, allowing subsequent rounds of FACS screening in E. coli. This approach facilitates the selection of a highly diverse repertoire of binders while enabling real-time monitoring and optimization of the screening process [65, 66]. In addition to the high-throughput FACS screening method, MACS is also widely used for bacterial cell display antibody libraries due to its faster library processing speed and the absence of expensive laboratory equipment [67, 68]. Furthermore, by incubating the bacterial antibody library with cells expressing the target antigen, bacteria can adhere to the target cells, enabling specific clone selection based on target cell binding [69]. This live-cell screening is particularly suitable for the development of nanobodies targeting tumor surfaces [70]. Overall, bacterial cell display antibody libraries are emerging as an efficient, cost-effective, and scalable antibody screening platform, overcoming technical bottlenecks and expanding into broader applications.

Ribosome display antibody library technology

Ribosome display technology enables the screening and identification of functional proteins by forming"protein-ribosome-mRNA"(PRM) ternary complexes in vitro, which links newly synthesized proteins to their corresponding mRNA molecules [71]. Ribosome display Antibody libraries are typically screened using antigen-functionalized magnetic beads. After the screening process, the eluted mRNA undergoes RT-PCR to synthesize cDNA, which could serve as the template for subsequent antibody expression [72].

Library display technology, with its advantages in high-throughput screening and the rapid generation of high-affinity antibodies, has greatly advanced the development of therapeutic antibodies. However, display techniques that present antibody fragments on the surface of living cells or viruses face the challenge of low efficiency in transfecting large DNA libraries [73]. Ribosome display libraries overcome these limitations by constructing the library in a cell-free system and introducing methods such as error-prone PCR and DNA recombination. This approach significantly enhances molecular genetic diversity and facilitates the affinity maturation and evolution of antibodies in vitro [74]. Despite its advantages, the effective function of ribosome display technology is heavily reliant on the integrity of the PRM complex. One significant challenge is the susceptibility of mRNA within the PRM complex to degradation in vitro, which can lead to decreased stability of the complexes, thereby affecting antibody selection efficiency, since incomplete or unstable complexes may not be effectively selected [75]. To address this issue, Ohashi et al. invented the PURE system for protein synthesis in vitro. This optimized system, which contains fewer nucleases and proteases, improves mRNA recovery and produces more stable ternary complexes [76]. Additionally, Wagner et al. developed the PolyMap high-throughput platform, which combines bulk binding of ribosome display libraries with scRNA-seq for pairwise mapping of protein–protein interactions, characterizing unique antibody clones without the need for mRNA recovery, thereby improving the efficiency of antibody development [77]. However, ribosome display technology is considered less optimal compared to cell- or phage-based antibody library display technologies due to reduced fidelity in cell-free reactions. For instance, cell-based display technologies leverage the natural mechanisms of living cells, offering higher expression efficiency and more reliable screening outcomes. In contrast, ribosome display often faces challenges related to stability and fidelity in vitro, which can negatively impact antibody selection and optimization processes [78, 79].

Single B cell antibody technology

Mammalian B-lymphocyte are extremely diverse and can produce as many as 1012 antibody clonotypes [80]. Unlike traditional antibody display technologies (e.g. phage display), which rely on iterative screening within a vast antibody library, single B-cell antibody technology allows for the direct isolation of individual antigen-specific B cells from animal tissues or peripheral blood [81]. The specific process of this technology(outlined in Fig. 2) includes antigen-specific B cell isolation, antibody gene amplification, and the expression and purification of recombinant antibodies. Compared with traditional antibody production techniques, single B cell antibody production technology offers the advantage of rapidly obtaining naturally paired light and heavy chain variable regions through high-throughput screening of antigen-specific B cells from animals or humans [82]. With the progression of technology, downstream expression methodologies in single B-cell antibody technology, including the utilization of expression plasmids such as the pFUSE series, as well as various engineered cell lines like CHO-K1 and HEK293, have become well-established [83,84,85]. In addition, matured nested PCR enables high-fidelity amplification of antibody variable region genes, thereby serving as a crucial tool for the generation of single-cell antibodies [86, 87]. However, the cell sorting method plays a crucial role in isolating individual B cells with high antigen affinity and specificity from heterogeneous cell populations, which significantly influences the specificity and affinity of the antibodies produced. In this section, we provide a comprehensive review of various single B-cell sorting techniques and their applications in antibody production.

Fig. 2
figure 2

The high-throughput antibody production scheme of the single B cell antibody technology. In the context of isolating PBMCs or splenocytes from immunized animals, as well as PBMCs from convalescent patients, various techniques such as MACS can be employed to enhance the concentration of target B cells. Subsequently, antigen-specific single B cells be isolated using methods such as FACS and microfluidics. The antibody variable region genes can be cloned through RT-PCR to facilitate antibody expression and subsequent analysis

Single B-cell antibody preparation based on fluorescence activated cell sorting

FACS is a widely recognized and highly efficient technique that employs a flow cytometer for the multiparametric detection and classification of cells. This method enables selective separation of target B cells through the establishment of screening"gates."The sorting principle is as follows. Initially, fluorescently labeled antigens and antibodies targeting various cell surface markers are utilized to immunolabel the antigen-specific B cells in the single-cell suspension. Then, the labeled cellular samples are introduced into a flow chamber operating under high-pressure conditions, wherein the sheath fluid arranges the cells into a single row. The cells then traverse a fluorescence detection channel, where they are exposed to ultrahigh-frequency vibrations to generate single-cell droplets. These droplets are subsequently sorted into individual cells utilizing an electrostatic deflection system [88]. The individual cells are then isolated into distinct wells of cell culture plates. Following cell lysis for RNA extraction, cDNA synthesis and amplification of antibody variable region genes can be performed. In fact, FACS technology is capable of screening individual cells at rates of up to 10,000 cells per second and can precisely isolate specific B cells [89]. The principal advantage of FACS technology in single B-cell antibody preparation is its ability to swiftly and accurately identify individual antigen-specific B cells through multicolor fluorescence. This capability has significantly facilitated the development of a wide array of antibodies in research. Carbonetti et al. rapidly and efficiently isolated, cloned, and produced monoclonal antibodies from immunized mice, which was achieved through the utilization of multichannel screening techniques employing markers such as B220-PacBlue, CD38-APC, and IgM-FITC, among others [90]. Additionally, Fan et al. employed multicolor FACS to generate humanized monoclonal antibodies targeting seven distinct subtypes of BoNT/F, resulting in antibodies that could neutralize multiple antigenic variants [91]. This approach exemplifies how FACS technology can streamline the production of highly specific and versatile monoclonal antibodies.

Nonetheless, the low sensitivity and high detection noise of FACS often lead to challenges associated with false positives (FP) and false negatives (FN), which in turn increase the workload of subsequent antibody characterization and validation processes [92]. To improve the specificity of sorting individual B cell, the integration of MACS with FACS is increasingly employed. Zhou et al. utilized magnetic beads to selectively enrich specific B cell populations before dual fluorescent dye sorting, which markedly diminished the interference from other cell types within the sample [93]. This antibody development strategy not only improves sorting specificity but also reduces the subsequent workload associated with antibody expression and characterization. Moreover, to further reduce the impact of FN and FP, it is crucial to enhance the identification of positive clones through single B cell culture and the analysis of culture supernatants [94]. Moreover, antibody-secreting cells (ASCs) not only exhibit higher antibody affinity compared to memory B cells but are also present in significantly larger quantities in vivo [95]. However, their low expression of surface markers poses substantial challenges for isolation and identification using FACS, as this method heavily relies on marker expression for cell sorting. These technical difficulties have greatly hindered the widespread application of FACS in studying ASCs [96]. Although the hetero-functional particles developed by Ramirez et al. integrated with the FACS platform enable high-throughput isolation of specific ASCs and significantly enhance the enrichment efficiency of antibody-associated cells, this method is relatively complex to design and requires skilled operation to fully optimize the platform's capabilities [97]. Moreover, the application of FACS screening may be limited by unclear or absent cell surface markers, which can lead to the loss of ideal antibody clones. For instance, rabbit-derived lymphocytes are currently constrained by the lack of useful surface markers on their cell membranes for effective screening. While Starkie et al. introduced a two-color antigen staining method for the recognition of antigen-specific rabbit memory B cells. the intricate design of the screening process led to challenges in ensuring the specificity of the identified positive cells, necessitating extensive downstream validation efforts, including a substantial amount of antibody cloning and validation work in subsequent stages [98]. In addition, FACS often requires a large starting number of cells for the screening of antigen-specific B cells, and manipulations in the pre-treatment may affect cell viability, resulting in difficulties in the identification of downstream antibody affinity and antibody activity.

Microfluidic sorting-based single B-cell antibody preparation technology

To address the limitations of traditional FACS methods in sorting single B cells for antibody development, microfluidics-based sorting technologies offer significant advancements in both efficiency and precision. Microfluidics technology is categorized into microdroplet and microwell systems according to their operational principles [99]. These microfluidic platforms enable the detection and isolation of specific B cells at the single-cell level and support the cultivation of positive clones within a controlled nutrient environment [100]. Compared to traditional antibody production methods, microfluidics-based single-cell antibody production technologies offer superior cell sorting precision, throughput, and integration capabilities. These advantages make microfluidics a key tool for large-scale antibody screening and optimization, underscoring its potential to advance the fields.

Based on droplet-based microfluidics

Microdroplet microfluidics utilizes an aqueous phase containing cells and analytical reagents, in conjunction with an oil phase that isolates individual cell droplets. Within the flow channel, these droplets are swiftly analyzed and identified using a high-frequency laser, while an electric field classifies them based on their unique characteristics [101]. A notable advantage of this technique for antibody screening is that each droplet not only functions as an independent microreactor but also has an extremely small volume (in the picoliter range), effectively preventing cross-contamination between droplets and ensuring that the cells and reagents within each droplet can undergo efficient and precise reactions in a confined environment [102]. Additionally, precise manipulation of droplets through merging, splitting, and sorting enables high-throughput separation and recovery of cells. This technique allows for the generation of droplets in the order of 107 per experiment, processing thousands per second. Consequently, it significantly reduces the screening cycle from months to a single day, greatly enhancing assay efficiency [103].

Microfluidic platforms, celebrated for their efficient sorting capabilities, have significantly propelled the advancement of antibody development. In the realm of scientific research, Eyer et al. have elucidated a droplet-based microfluidic technique known as DropMap. This technique facilitates massively parallel kinetic analysis of individual ASCs by integrating immobilized picoliter-sized droplets with highly sensitive assays. DropMap enables precise assessment of antibody secretion rates, specificity, and affinity for antigens, thereby enhancing the capabilities of antibody evaluation and characterization [104]. For commercial applications, the Cyto-Mine® platform developed by Sphere Fluidics Co., Ltd. offers a significant advantage in antibody screening. This system utilizes fluorophore-labeled donor and acceptor probes to enable fluorescence resonance energy transfer (FRET) for the detection of cellular secretions. Antibodies secreted by cells within droplets are captured by these probes; one fluorophore is excited by a laser, transferring energy to the second fluorophore. This technique provides precise quantification of target antibodies and facilitates the efficient production of recombinant homologous paired antibody candidates for secondary screening [105]. According to Doerner et al., the Cyto-Mine® platform provides several key advantages: 1) It enables the rapid identification of antigen-specific positive cells through FRET technology, with direct applicability to full IgG formats. 2) It can efficiently analyze up to one million single antibody-secreting cells, achieving a screening rate of 250 drops per second. 3) The microdroplet technology minimizes mechanical damage to cells, thereby maintaining cell viability for subsequent antibody performance evaluations. 4) The entire process, from cell preparation to recombinant antibody confirmation, is completed within a four-week timeframe [106]. Similarly, Brenan et al. introduced the Celli GO technique, which combines fluorescence-based single-cell bioanalysis with single-cell barcoding in droplets to visualize specific ASCs. This technique allows sorting of droplets at velocities up to 600 per second, resulting in the generation of 77 recombinant antibodies from identified sequences [107]. The MTX-COVAB antibody drug developed by Hillenbrand et al. DROPZYLLA® platform has demonstrated significant efficacy in vivo, further highlighting the impact of microdroplet microfluidic technologies in antibody development [108]. Additionally, Adler et al. introduced an emulsion droplet microfluidic approach that enables the isolation of millions of single B cells while preserving the natural pairing of heavy and light chains, facilitating the affinity screening of rare antibodies from murine libraries [109]. Furthermore, DeKosky et al. developed a single-cell, emulsion-based microfluidic technology capable of generating VH-VL amplicons for NGS [110]. These advancements further expand the applications of droplet microfluidics in antibody discovery and lay the foundation for single-cell immune repertoire sequencing.

Recent advancements in microdroplet microfluidic platforms have significantly enhanced the capabilities for antibody development, yet several critical challenges remain. Key issues include the need for greater simplification of technology and the integration of automated systems. Addressing these challenges is crucial for the continued progress of droplet microfluidic antibody discovery platforms. In this context, Wippold et al. have introduced a rapid assessment platform that leverages integrated microfluidic technology to enable high-throughput, single-cell resolution identification of neutralizing antibody libraries in response to viral infections [111]. Wang et al. have developed a sophisticated signal capture system that addresses cell viability issues arising from iterative enrichment processes and overcomes some limitations associated with existing commercial plasma cell enrichment reagents [112]. In a related development, Wang et al. have successfully combined an autocrine-based lentiviral transduction system with microfluidic droplet technology, enhancing both efficiency and capacity. This innovation facilitates the screening of low-frequency functional antibodies and the characterization of bispecific antibodies [113]. Mazutis et al. have employed FRET-based droplet microfluidics to screen individual antibody-secreting cells, achieving sorting efficiencies of up to 97.5% and an 800-fold enrichment of target cells in a single sorting round, thus significantly accelerating the antibody discovery process [114]. Additionally, Ahmadi et al. have pioneered a novel method for on-demand co-encapsulation of cells, providing new insights into the automation of droplet microfluidic antibody discovery techniques [115]. These advancements highlight the rapid progress in droplet microfluidic platforms for antibody discovery, showcasing their transformative potential in high throughput antibody research. High-throughput microdroplet technologies can now generate and screen approximately ten million picoliter-scale microdroplet reaction units in a single experimental iteration. This unprecedented capacity not only accelerates throughput but also shortens development timelines and reduces operational costs, making these platforms highly attractive for large-scale antibody screening. However, significant challenges persist that must be addressed to unlock the full potential of this technology. A major limitation is the occurrence of false-negative results, often attributed to the inefficient encapsulation of ASCs within the defined microdroplet volumes. Furthermore, the intrinsic constraints of picoliter-sized droplets, such as limited reagent availability and dilution effects, can compromise detection sensitivity and accuracy. Overcoming these barriers will require innovations in droplet design, cell encapsulation efficiency, and signal amplification strategies. Future advancements in these areas are essential for enhancing the reliability and scalability of droplet microfluidics, paving the way for its broader application in antibody discovery and beyond.

Based on microwell-based microfluidics

Antibody discovery and development have made significant strides with the advent of microfluidic technologies. Single-Cell Antibody Nanopores (SCANs) represent a synthesis of microwell chips devices and high-throughput screening techniques, offering a novel approach to isolating and analyzing individual B cells. The original microengraving and immunospot array assay on a chip (ISAAC) is represents a sophisticated technique for SCAN applications. Among these, microengraving technology enables the precise separation and isolation of individual cells in microfluidic chip chambers by exploiting variations in cell size, shape, or surface markers, ensuring that a single cell is present in each chamber. These miniature chambers are meticulously designed to offer an optimal cell culture environment, encompassing essential nutrients, growth factors, and extracellular matrix components, thereby facilitating the growth and maturation of individual B cells. Antibodies secreted by individual B cells are transferred to a protein microarray for antigen-specific screening and are mapped to the corresponding target cells. Subsequently, the target cells are isolated from the wells through micromanipulation techniques, facilitating high-throughput cell sorting [116]. ISAAC technique utilizes anti-IgG or specific antigens immobilized on the surface of a microarray. Antibodies secreted by the cells bind to the immobilized anti-IgG or specific antigens, resulting in the formation of distinct circular spots that are readily distinguishable from non-specific signals [117]. Using this technique, ASCs are identified within 3–4 days and subsequently analyzed through clonal expansion or RT-PCR [118].

SCANs can screen up to 100,000 polyclonal B cells simultaneously, thereby providing remarkable high-throughput capacity, sensitivity, and specificity [119]. The use of fluorescently labeled antigens for screening and identification reduces extensive validation of antibody clones [120, 121]. Additionally, the multi-nanopore design of SCANs technology enables the simultaneous study of specific antibodies targeting a diverse array of distinct antigens on a single chip. Esfandiary et al. employed SCAN screening to identify high-frequency cell clones producing specific IgG isotypes targeting anti-SSA/Ro60 and anti-SSB/La antigens, illustrating the capability of SCAN to isolate multiple antigen-specific B cells on a single chip [122]. Currently, researchers have enhanced the microwell system to improving the efficiency of antibody development. For instance, Li et al. introduced CSMN, a nanopore-based, semi-antigen-specific rabbit ASC selection method, enabling the acquisition of target antibody gene sequences within a 5-day screening period [123]. Abali et al. employed a nanopore platform for screening, enabling the generation of high-titer antibodies. This process, from the introduction of single cell and the measurement of antibody yield to the production of fully amplified clones, was completed within a timeframe of 4 to 6 weeks [124]. However, this technique necessitates intricate and precise experimental procedures, including the extraction of cells from microwells and the application of antibody coatings on the chip surface. Furthermore, the design and fabrication of these chips involve substantial costs. As a result, the technology platform has limited applicability. Another important consideration is the high cell density within the miniature device, which may result in cross-contamination between cells and subsequently impact the accuracy of the experimental outcomes.

In contrast to SCANs, the commercial Beacon® single B-cell screening platform exhibits superior automation capabilities. Introduced in 2016 by Berkeley Lights, Co., Ltd., the Beacon® platform offers fully automated and high-throughput functionality, establishing itself as a pivotal tool for high-throughput antibody screening [125]. This platform integrates advanced opto-electro-positioning (OEP) technology, nanoscale microreactor systems, and innovative microfluidic design. This combination enables precise single B-cell sorting, nanoscale single B-cell culture, and validation of positive clonal antibody secretion—all within a compact, centimeter-scale chip [126]. The experimental procedure for this technology platform consists of four sequential stages. In the first stage, 10,000 to 20,000 B cells are introduced onto the Beacon® chip via microfluidics. Next, using OEP technology, individual B cells are isolated and transferred to NanoPen™ culture chambers for cultivation. Each NanoPen™ chamber can only accommodate one cell, ensuring a monoclonality rate of 99% or higher while maintaining high cell viability. In the third stage, the platform performs real-time detection of antibody binding strength, specificity, and affinity within the NanoPen™ chambers to identify effective clones. Finally, specific B cells are selected based on culture and real-time analysis results, extracted using OEP technology, and recovered into multi-well plates for further expansion [127].

The Beacon® single B cell OEP platform enables full automation of the entire single-cell experimental workflow. Winters et al. employed this platform to generate antibodies targeting unique reagents [128]. Remarkably, the entire workflow, encompassing chip-based cell import, antibody screening, and data export for discovery, was completed in less than 5 h. Furthermore, the complete discovery process, encompassing immunization to recombinant antibody expression, can be accomplished in as few as 40 days. Douet et al. utilized the Beacon platform to generate efficient CHO clones in a reduced timeframe, thereby enhancing both the throughput of the Beacon® system and the identification of high-producing clones [129]. Compared to traditional single-cell antibody preparation platforms, the Beacon® single B-cell screening platform enables automated cell sorting, cultivation, and positive clone identification, significantly reducing manual intervention associated with traditional methods. This advancement not only dramatically accelerates antibody development but also improves stability and reproducibility. Additionally, the Beacon® single B-cell screening platform can simultaneously process tens of thousands of cells, providing exceptional scalability and significantly enhancing antibody development efficiency. Given these advantages, the platform has become a pivotal tool in the development and industrialization of therapeutic and diagnostic antibodies [130, 131].

Notably, while both microwell-based microfluidics and the commercial Beacon® System facilitate the cultivation and characterization of individual specific ASCs within microwell or NanoPen™ chamber, amplification and sequencing of variable region genes is still performed off-chip, this separation increases the risk of ASC loss and cross-contamination. To address these issues, Zhang et al. introduced an innovative modular design for the SSMA chip, which allows for the simultaneous acquisition of paired variable region genes from a single ASC within a single SSMA chip, thereby reducing the risks of cell loss and cross-contamination [132]. Nevertheless, microwell-based microfluidics and commercial beacon® systems remain prohibitively expensive and complex for antibody development. Future advancements in user-friendliness, cost efficiency, and increased throughput may offer substantial opportunities for the field of single cell antibody development.

Antibody production technology for high-throughput single-cell sequencing

The complex processes of V(D)J gene rearrangement, somatic hypermutation (SHM), and class switching generate a vast repertoire of variable region sequences in antibody heavy and light chains, contributing to the huge capacity of the B cell immune repertoire [133]. Generally, the antibody preparation technique based on high-throughput single-cell sequencing includes the following processes: 1) Isolation and enrichment of specific B cells from peripheral blood mononuclear cells (PBMCs) or splenocytes obtained from animal models. 2) In water-in-oil microdroplets, gel beads or magnetic beads with tagged sequences are used to capture single B cells, followed by the extraction of RNA from each cell, which is then reverse-transcribed into cDNA for sequencing library construction. 3) Sequencing of B cells by single-cell NGS technology to obtain large-scale single-cell resolution data. 4) Bioinformatics tools are used to analyze the sequencing data from individual B cells to obtain the paired IgG heavy and light chain variable region genes [134]. Figure 3 depicts the technical workflow of high-throughput antibodies preparation based on high-throughput single-cell sequencing. This technology exemplifies the synergy between state-of-the-art sequencing techniques and advanced bioinformatic methodologies. Such integration underscores the sophistication and depth of modern antibody repertoire research. By offering a comprehensive framework for analyzing the B cell antibody repertoire, this approach markedly advances the development of high-throughput antibody solutions.

Fig. 3
figure 3

The high-throughput antibody production scheme of the single-cell sequencing. In the context of isolating PBMCs or splenocytes from immunized animals, as well as PBMCs from convalescent patients. Target B cells were enriched using MACS or FACS. Single-cell sequencing was conducted on the enriched cells to obtain 5'-end transcriptome data and immune repertoire data. Bioinformatics analysis was then employed to identify high-affinity antibody gene sequences from a vast amount of single-cell sequencing data. Specific vectors were subsequently incorporated for antibody expression and downstream characterization

The introduction of Cellular Indexing of Transcriptomes and Epitopes by sequencing (CITE-seq) in 2017 marked a significant breakthrough in the field of single-cell sequencing [135]. Building on the success of CITE-seq, Setliff et al. proposed linking B-cell receptors to antigen specificity through sequencing (LIBRA-seq). The core of LIBRA-seq is the addition of fluorescent modifications and oligonucleotide labeling (including PCR processing, barcode sequences, and capture sequences) onto antigens. By incubating B cells with different antigens and using FACS to sort antigen-specific B cells. Subsequently, microfluidic technology is employed to encapsulate individual B cells in droplets containing magnetic beads, followed by single cell RNA sequencing (scRNA-seq). The magnetic beads are coated with universal cell barcodes, which facilitate the linking of antigen-specific barcodes and B cell receptor (BCR) transcripts, enabling the identification of both the BCR sequences and their corresponding antigen specificity [136]. This method offers several advantages, including rapid, high-throughput antibody screening, the establishment of direct correlations with antigen specificity, and the simultaneous identification of broad-spectrum neutralizing antibodies. These broad-spectrum antibodies are capable of recognizing and neutralizing a number of different antigens or antigenic variants, which is particularly valuable against different strains of pathogens or against multiple epitopes of a single antigen. Kramer et al. used LIBRA-seq technology to isolate a monoclonal antibody, 54,042–4, from COVID-19 convalescents that was effective in neutralizing SARS-CoV-2 viruses including multiple SARS-CoV-2 variants [137]. Additionally, Pilewski et al. employed LIBRA-seq to study the antibody repertoire of individuals co-infected with chronic HIV-1 and HCV, identifying five antibodies with broad neutralization activity and potent functional effects against both HIV-1 and HCV [138]. Recently, substantial progress has been made in LIBRA-seq. Shiakolas et al. incorporated barcode-labeled antigenic ligands into the LIBRA-seq platform, thereby establishing a comprehensive three-dimensional correlation among antibody sequences, antigens, and neutralization effects. This advancement has significantly improved the efficiency of neutralizing antibody screenings [139]. Additionally, Walker et al. employed a panel of antigens with point mutations to map the relationship between antigenic epitopes and antibody sequences. This approach has refined the antigenic epitopes of thousands of B cell receptors simultaneously, thereby elevating the functionality and utility of LIBRA-seq [140]. LIBRA-seq has now achieved remarkable success in the development of antibodies against a diverse array of viruses [141,142,143]. Overall, LIBRA-seq represents a powerful and innovative method for antibody development. However, its application may be constrained in certain scenarios due to the complexity of recombinant antigen design and the discontinuous nature of its experimental workflow.

In contrast, the 10 × Genomics single-cell sequencing platform has shown great potential in simplifying experimental workflows and providing broader and more comprehensive antibody discovery. As a premier platform for single-cell antibody library sequencing, it utilizes sophisticated microfluidic technology and efficient molecular identification methods to support large-scale antibody development and optimization, thereby offering a dependable solution for high-throughput antibody screening [144]. The platform leverages microfluidic technology to encapsulate single cell, gel beads, and reaction reagents within a"water-in-oil"droplet microenvironment, each tagged with molecular identifiers (UMIs) and cellular barcodes. Upon cell lysis within the droplets, mRNA containing Poly(dA) sequences are released, subsequently binding and being reverse-transcribed into cDNA that carries both the cellular barcode and UMI. After oil dissolution, a portion of the resulting cDNA is used for the construction of a 5'transcriptome library, while the remainder undergoes PCR amplification for V(D)J gene analysis, facilitating the construction of an immune repertoire library [145, 146]. The 10 × Genomics platform facilitates high-throughput single-cell V(D)J sequencing, providing the capability to accurately capture and pair the complete variable region sequences of both light and heavy chains with high precision. This technology not only facilitates detailed single cell analysis but also provides comprehensive expression profiling information. With its significant advantages, including high resolution and efficiency, the platform is widely used by researchers to develop antibodies with high affinity and specificity, significantly advancing the field of antibody discovery and development [147, 148].

Compared to other antibody development techniques, high-throughput single-cell sequencing offers a faster and more efficient solution by bypassing intermediate steps and directly obtaining high-quality antibody data [149]. However, the vast volumes of sequencing data generated by high-throughput single-cell sequencing technologies can be overwhelming. A critical challenge in identifying and isolating high-efficiency antibodies lies in enhancing the proportion of target-specific or high-value information within these datasets. Developing efficient strategies to prioritize and refine these datasets is essential for advancing antibody discovery. To address this challenge, researchers often focus on enriching target B cells specific to the antigen before sequencing. This approach helps reduce interference from low-affinity B cells and other cellular components, thereby enhancing the quality and pertinence of the data. For instance, Cao et al. improved the success rates for high-efficiency neutralizing antibody screening by using magnetic bead-specific enrichment of patient sera followed by 10 × Genomics single-cell sequencing, identifying 14 high-efficiency neutralizing antibodies within two weeks [150]. Similarly, Wang et al. employed FACS to isolate memory B cells bound to recombinant Staphylococcus aureus antigen, followed by 10 × Genomics single-cell sequencing to identify 10 distinct IgG antibodies [151]. These studies highlight the effectiveness of pre-sequencing enrichment strategies in reducing non-informative noise from low-affinity B cells and other contaminants. By selectively enriching target cells prior to sequencing, this approach significantly enhances the likelihood of identifying high-performance antibodies, thus advancing the development of potent therapeutic antibodies.

To fully leverage the advantages of high-throughput single-cell sequencing technology, implementing pre-sequencing enrichment strategies is crucial for improving data quality and relevance. However, these strategies alone are inadequate. Equally important is the bioinformatics analysis of the downstream data produced by single-cell sequencing. Generally, data from single-cell immune repertoire sequencing must be processed to infer B-cell population structures and quantify detailed features [152]. Various tools have been developed for BCR analysis, including IgBLAST and IMGT/HighV-QUEST for V(D)J gene annotation, as well as AbNum and AbRSA for antibody numbering [153,154,155]. Research indicates that various tools exhibit differing levels of performance. For example, a study by Smakaj et al. identified MiXCR as the most efficient tool for sequence processing when compared to alternatives such as IMGT/HighV-QUEST and IgBLAST [156]. The large volume of sequences poses a significant processing challenge. Zong et al. recently introduced Abalign (http://cao.labshare.cn/abalign/), based on AbRSA, which significantly reduces the time required for high-throughput BCR data analysis from weeks to hours [157]. Additionally, machine learning and deep learning have extensive applications for antibody data analysis. Researchers are developing artificial intelligence models that integrate these techniques with traditional bioinformatics methods to extract valuable information from immunome libraries [158]. Bai et al. has reviewed major machine learning techniques and their applications, including computational predictions of antibody structures, antigen interfaces, and interactions [159]. These advanced methods enhance data processing efficiency and support high-throughput antibody production and screening. By leveraging these tools, researchers can rapidly identify and optimize therapeutically valuable antibodies, thus accelerating the antibody development process and advancing the discovery of novel antibody drugs. The application of these technologies is ushering in a new era in high-throughput antibody screening, making the transition from discovery to product more efficient and precise.

Summary and outlook

High-throughput antibody screening has gained widespread attention for its rapid, efficient, and scalable capabilities in antibody preparation. With continuous advancements in bioinformatics, structural biology, antibody engineering, artificial intelligence, and automation, these technologies are increasingly focusing on integration, automation, and intelligence. In this systematic review, we summarize current high-throughput antibody screening technologies, including antibody library display-based screening strategies, single-cell technologies, and single-cell sequencing. While several recent reviews have delved into various strategies for monoclonal antibody discovery, such as antibody display technologies, microfluidics, and single B cell technologies, they typically focus on individual strategies without offering a comprehensive comparison within a high-throughput framework [160,161,162]. In contrast, our review not only summarizes these strategies but also emphasizes how they integrate and work synergistically within modern high-throughput antibody discovery pipelines.

Table 1 provides a summary of the characteristics of antibody library technology, single B cell antibody production, and single-cell sequencing in the context of antibody development. Overall, compared to the initial complex procedures that required multiple rounds of antibody library screening, the integration of automation and higher-throughput methods (such as microfluidics and NGS) has transformed antibody library display technology into a powerful platform for innovative drug discovery. However, it is important to note that the quality of the antibody display library has a significant impact on screening outcomes. In contrast, single-cell antibody preparation technology offers high-throughput screening capabilities, enabling the generation of naturally paired antibodies with both light and heavy chain variable regions. This technology is known for its speed and high affinity, making it one of the primary methods for industrial-scale antibody production. Despite these advantages, there are still challenges, such as ensuring the accuracy of positive clone screening, as well as the complexity and cost of the required instruments. Among these techniques, high-throughput single-cell sequencing stands out as the most efficient method for antibody preparation. It can provide gene expression profiles and immune repertoire data from a large number of single cells in a single run. However, the complexity and volume of immune repertoire sequencing data present significant challenges. While existing bioinformatics tools, along with the integration of machine learning and deep learning techniques, offer promising solutions for data analysis and mining, there are still limitations. Further improvements and enhancements to algorithms, scoring functions, databases, and benchmarking tools are necessary to better address these challenges.

Table 1 Characteristics of different antibody high-throughput preparation techniques

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

mAb:

Monoclonal antibodies

NGS:

next-generation sequencing

HAMA:

human anti-mouse antibody

scFv:

single chain antibody fragment

SdAb:

single-domain antibodies

FACS:

fluorescence activated cell sorting

MCS:

multiple cloning site

PCR:

polymerase chain reaction

PRM:

protein-ribosome-mRNA

cDNA:

complementary DNA

FP:

false positives

FN:

false negatives

MACS:

Magnetic Activated Cell Sorting

ASCs:

antibody-secreting cells

FRET:

fluorescence resonance energy transfer

SCANs:

Single-Cell Antibody Nanopores

ISAAC:

immunospot array assay on a chip

OEP:

opto-electro-positioning

SHM:

somatic hypermutation

LIBRA-seq:

linking B-cell receptors to antigen specificity through sequencing

CITE-seq:

Cellular Indexing of Transcriptomes and Epitopes by sequencing

scRNA-seq:

single cell RNA sequencing

References

  1. Bradbury A, Plückthun A. Reproducibility: Standardize antibodies used in research. Nature. 2015;518(7537):27–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/518027a.

    Article  Google Scholar 

  2. Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256(5517):495–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/256495a0.

    Article  Google Scholar 

  3. Basu K, Green EM, Cheng Y, Craik CS. Why recombinant antibodies - benefits and applications. Curr Opin Biotechnol. 2019;60:153–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.copbio.2019.01.012.

    Article  Google Scholar 

  4. Li Y, Yang L, Yu W, Yu X, Wen K, Shao B, et al. Highly efficient and precise two-step cell selection method for tetramethylenedisulfotetramine-specific monoclonal antibody production. J Hazard Mater. 2022;424(Pt D): 127689. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jhazmat.2021.127689.

    Article  Google Scholar 

  5. Fischer K, Lulla A, So TY, Pereyra-Gerber P, Raybould MIJ, Kohler TN, et al. Rapid discovery of monoclonal antibodies by microfluidics-enabled FACS of single pathogen-specific antibody-secreting cells. Nat Biotechnol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41587-024-02346-5.

    Article  Google Scholar 

  6. Segaliny AI, Jayaraman J, Chen X, Chong J, Luxon R, Fung A, et al. A high throughput bispecific antibody discovery pipeline. Commun Biol. 2023;6(1):380. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42003-023-04746-w.

    Article  Google Scholar 

  7. Tiller T, Meffre E, Yurasov S, Tsuiji M, Nussenzweig MC, Wardemann H. Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. J Immunol Methods. 2008;329(1–2):112–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jim.2007.09.017.

    Article  Google Scholar 

  8. Dumontet C, Reichert JM, Senter PD, Lambert JM, Beck A. Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov. 2023;22(8):641–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41573-023-00709-2.

    Article  Google Scholar 

  9. Rogers TF, Zhao F, Huang D, Beutler N, Burns A, He WT, et al. Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model. Science. 2020;369(6506):956–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.abc7520.

    Article  Google Scholar 

  10. Elhanati Y, Sethna Z, Marcou Q, Callan CG, Mora T, Walczak AM. Inferring processes underlying B-cell repertoire diversity. Philosophical Transactions of the Royal Society B: Biological Sciences. 2015;370(1676). https://doiorg.publicaciones.saludcastillayleon.es/10.1098/rstb.2014.0243.

  11. Wellner A, McMahon C, Gilman MSA, Clements JR, Clark S, Nguyen KM, et al. Rapid generation of potent antibodies by autonomous hypermutation in yeast. Nat Chem Biol. 2021;17(10):1057–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41589-021-00832-4.

    Article  Google Scholar 

  12. Huang CY, Lok YY, Lin CH, Lai SL, Wu YY, Hu CY, et al. Highly reliable GIGA-sized synthetic human therapeutic antibody library construction. Front Immunol. 2023;14:1089395. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1089395.

    Article  Google Scholar 

  13. Clackson T, Hoogenboom HR, Griffiths AD, Winter G. Making antibody fragments using phage display libraries. Nature. 1991;352(6336):624–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/352624a0.

    Article  Google Scholar 

  14. Barbas CF 3rd, Kang AS, Lerner RA, Benkovic SJ. Assembly of combinatorial antibody libraries on phage surfaces: the gene III site. Proc Natl Acad Sci U S A. 1991;88(18):7978–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.88.18.7978.

    Article  Google Scholar 

  15. Tohidkia MR, Sepehri M, Khajeh S, Barar J, Omidi Y. Improved Soluble ScFv ELISA Screening Approach for Antibody Discovery Using Phage Display Technology. SLAS Discov. 2017;22(8):1026–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/2472555217701059.

    Article  Google Scholar 

  16. McGowan EC, Flower RL, Jones ML, Irving DO, Barnard RT, Hyland CA, et al. Aggregates in blood filter chambers used from the plasma donations of anti-D donors: evaluation for monoclonal antibody discovery using phage display. Blood Transfus. 2021;19(1):64–72. https://doiorg.publicaciones.saludcastillayleon.es/10.2450/2020.0093-20.

    Article  Google Scholar 

  17. Krebs B, Rauchenberger R, Reiffert S, Rothe C, Tesar M, Thomassen E, et al. High-throughput generation and engineering of recombinant human antibodies. J Immunol Methods. 2001;254(1–2):67–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0022-1759(01)00398-2.

    Article  Google Scholar 

  18. Turunen L, Takkinen K, Söderlund H, Pulli T. Automated panning and screening procedure on microplates for antibody generation from phage display libraries. J Biomol Screen. 2009;14(3):282–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1087057108330113.

    Article  Google Scholar 

  19. Min B, Yoo M, Kim H, Cho M, Nam DH, Yoon Y. Semi-Automated Cell Panning for Efficient Isolation of FGFR3-Targeting Antibody. Int J Mol Sci. 2021;22(12). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms22126240.

  20. de Wildt RM, Mundy CR, Gorick BD, Tomlinson IM. Antibody arrays for high-throughput screening of antibody-antigen interactions. Nat Biotechnol. 2000;18(9):989–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/79494.

    Article  Google Scholar 

  21. Pérez-Gamarra S, Hattara L, Batra G, Saviranta P, Lamminmäki U. Array-in-well binding assay for multiparameter screening of phage displayed antibodies. Methods. 2017;116:43–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ymeth.2016.12.004.

    Article  Google Scholar 

  22. de Kruif J, Terstappen L, Boel E, Logtenberg T. Rapid selection of cell subpopulation-specific human monoclonal antibodies from a synthetic phage antibody library. Proc Natl Acad Sci U S A. 1995;92(9):3938–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.92.9.3938.

    Article  Google Scholar 

  23. Hsiao YH, Huang CY, Hu CY, Wu YY, Wu CH, Hsu CH, et al. Continuous microfluidic assortment of interactive ligands (CMAIL). Sci Rep. 2016;6:32454. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep32454.

    Article  Google Scholar 

  24. Hemadou A, Laroche-Traineau J, Antoine S, Mondon P, Fontayne A, Le Priol Y, et al. An innovative flow cytometry method to screen human scFv-phages selected by in vivo phage-display in an animal model of atherosclerosis. Sci Rep. 2018;8(1):15016. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-018-33382-2.

    Article  Google Scholar 

  25. Philpott DN, Gomis S, Wang H, Atwal R, Kelil A, Sack T, et al. Rapid On-Cell Selection of High-Performance Human Antibodies. ACS Cent Sci. 2022;8(1):102–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acscentsci.1c01205.

    Article  Google Scholar 

  26. Bidlingmaier S, Su Y, Liu B. Combining Phage and Yeast Cell Surface Antibody Display to Identify Novel Cell Type-Selective Internalizing Human Monoclonal Antibodies. Methods Mol Biol. 2015;1319:51–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-1-4939-2748-7_3.

    Article  Google Scholar 

  27. Azevedo Reis Teixeira A, Erasmus MF, D'Angelo S, Naranjo L, Ferrara F, Leal-Lopes C, et al. Drug-like antibodies with high affinity, diversity and developability directly from next-generation antibody libraries. MAbs. 2021;13(1):1980942. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/19420862.2021.1980942.

  28. De Keyser P, Kalichuk V, Zögg T, Wohlkönig A, Schenck S, Brunner J, et al. A biosensor-based phage display selection method for automated, high-throughput Nanobody discovery. Biosens Bioelectron. 2025;271: 116951. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bios.2024.116951.

    Article  Google Scholar 

  29. Hornsby M, Paduch M, Miersch S, Sääf A, Matsuguchi T, Lee B, et al. A High Through-put Platform for Recombinant Antibodies to Folded Proteins. Mol Cell Proteomics. 2015;14(10):2833–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/mcp.O115.052209.

    Article  Google Scholar 

  30. Lee HE, Cho AH, Hwang JH, Kim JW, Yang HR, Ryu T, et al. Development, High-Throughput Profiling, and Biopanning of a Large Phage Display Single-Domain Antibody Library. Int J Mol Sci. 2024;25(9). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms25094791.

  31. Sasso E, D’Avino C, Passariello M, D’Alise AM, Siciliano D, Esposito ML, et al. Massive parallel screening of phage libraries for the generation of repertoires of human immunomodulatory monoclonal antibodies. MAbs. 2018;10(7):1060–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/19420862.2018.1496772.

    Article  Google Scholar 

  32. Nakada-Masuta T, Takeda H, Uchida K. Novel Approach for Obtaining Variable Domain of New Antigen Receptor with Different Physicochemical Properties from Japanese Topeshark (Hemitriakis japanica). Mar Drugs. 2023;21(11). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/md21110550.

  33. Barreto K, Maruthachalam BV, Hill W, Hogan D, Sutherland AR, Kusalik A, et al. Next-generation sequencing-guided identification and reconstruction of antibody CDR combinations from phage selection outputs. Nucleic Acids Res. 2019;47(9): e50. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gkz131.

    Article  Google Scholar 

  34. Ballmann R, Hotop SK, Bertoglio F, Steinke S, Heine PA, Chaudhry MZ, et al. ORFeome Phage Display Reveals a Major Immunogenic Epitope on the S2 Subdomain of SARS-CoV-2 Spike Protein. Viruses. 2022;14(6). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/v14061326.

  35. Krohn S, Holtrop T, Brandsma AM, Moerer P, Nederend M, Darzentas N, et al. Combining Cellular Immunization and Phage Display Screening Results in Novel, FcγRI-Specific Antibodies. Viruses. 2024;16(4). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/v16040596.

  36. Su Y, Zhang X, Bidlingmaier S, Behrens CR, Lee NK, Liu B. ALPPL2 Is a Highly Specific and Targetable Tumor Cell Surface Antigen. Cancer Res. 2020;80(20):4552–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.Can-20-1418.

    Article  Google Scholar 

  37. Chen F, Liu Z, Jiang F. Prospects of Neutralizing Nanobodies Against SARS-CoV-2. Front Immunol. 2021;12: 690742. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.690742.

    Article  Google Scholar 

  38. Liu X, Wang Y, Sun L, Xiao G, Hou N, Chen J, et al. Screening and optimization of shark nanobodies against SARS-CoV-2 spike RBD. Antiviral Res. 2024;226: 105898. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.antiviral.2024.105898.

    Article  Google Scholar 

  39. Spadiut O, Capone S, Krainer F, Glieder A, Herwig C. Microbials for the production of monoclonal antibodies and antibody fragments. Trends Biotechnol. 2014;32(1):54–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tibtech.2013.10.002.

    Article  Google Scholar 

  40. Feldhaus MJ, Siegel RW, Opresko LK, Coleman JR, Feldhaus JM, Yeung YA, et al. Flow-cytometric isolation of human antibodies from a nonimmune Saccharomyces cerevisiae surface display library. Nat Biotechnol. 2003;21(2):163–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nbt785.

    Article  Google Scholar 

  41. Lebrun E, Shenshin V, Plaire C, Vigneres V, Pizette T, Dumas B, et al. Efficient full-length IgG secretion and sorting from single yeast clones in droplet picoreactors. Lab Chip. 2023;23(15):3487–500. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/d3lc00403a.

    Article  Google Scholar 

  42. Grzeschik J, Yanakieva D, Roth L, Krah S, Hinz SC, Elter A, et al. Yeast Surface Display in Combination with Fluorescence-activated Cell Sorting Enables the Rapid Isolation of Antibody Fragments Derived from Immunized Chickens. Biotechnol J. 2019;14(4): e1800466. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/biot.201800466.

    Article  Google Scholar 

  43. Benatuil L, Perez JM, Belk J, Hsieh C-M. An improved yeast transformation method for the generation of very large human antibody libraries. Protein Eng Des Sel. 2010;23(4):155–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/protein/gzq002.

    Article  Google Scholar 

  44. Mei M, Li J, Wang S, Lee KB, Iverson BL, Zhang G, et al. Prompting Fab Yeast Surface Display Efficiency by ER Retention and Molecular Chaperon Co-expression. Front Bioeng Biotechnol. 2019;7:362. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fbioe.2019.00362.

    Article  Google Scholar 

  45. Bowley DR, Labrijn AF, Zwick MB, Burton DR. Antigen selection from an HIV-1 immune antibody library displayed on yeast yields many novel antibodies compared to selection from the same library displayed on phage. Protein Eng Des Sel. 2007;20(2):81–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/protein/gzl057.

    Article  Google Scholar 

  46. Chao G, Lau WL, Hackel BJ, Sazinsky SL, Lippow SM, Wittrup KD. Isolating and engineering human antibodies using yeast surface display. Nat Protoc. 2006;1(2):755–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nprot.2006.94.

    Article  Google Scholar 

  47. Porebski BT, Balmforth M, Browne G, Riley A, Jamali K, Fürst M, et al. Rapid discovery of high-affinity antibodies via massively parallel sequencing, ribosome display and affinity screening. Nat Biomed Eng. 2024;8(3):214–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41551-023-01093-3.

    Article  Google Scholar 

  48. Choi HL, Yang HR, Shin HG, Hwang K, Kim JW, Lee JH, et al. Generation and Next-Generation Sequencing-Based Characterization of a Large Human Combinatorial Antibody Library. Int J Mol Sci. 2023;24(6). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms24066011.

  49. Mottershead DG, Alfthan K, Ojala K, Takkinen K, Oker-Blom C. Baculoviral Display of Functional scFv and Synthetic IgG-Binding Domains. Biochem Biophys Res Commun. 2000;275(1):84–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1006/bbrc.2000.3264.

    Article  Google Scholar 

  50. Mahdavi SZB, Oroojalian F, Eyvazi S, Hejazi M, Baradaran B, Pouladi N, et al. An overview on display systems (phage, bacterial, and yeast display) for production of anticancer antibodies; advantages and disadvantages. Int J Biol Macromol. 2022;208:421–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijbiomac.2022.03.113.

    Article  Google Scholar 

  51. Robertson N, Lopez-Anton N, Gurjar SA, Khalique H, Khalaf Z, Clerkin S, et al. Development of a novel mammalian display system for selection of antibodies against membrane proteins. J Biol Chem. 2020;295(52):18436–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.RA120.015053.

    Article  Google Scholar 

  52. Yang Y, Liu S, Luo Y, Wang B, Wang J, Li J, et al. High-throughput saturation mutagenesis generates a high-affinity antibody against SARS-CoV-2 variants using protein surface display assay on a human cell. PLoS Pathog. 2023;19(2): e1011119. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.ppat.1011119.

    Article  Google Scholar 

  53. Gaa R, Mayer HM, Noack D, Kumari K, Guenther R, Tsai SP, et al. Mammalian display to secretion switchable libraries for antibody preselection and high throughput functional screening. MAbs. 2023;15(1):2251190. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/19420862.2023.2251190.

    Article  Google Scholar 

  54. Fahad AS, Timm MR, Madan B, Burgomaster KE, Dowd KA, Normandin E, et al. Functional Profiling of Antibody Immune Repertoires in Convalescent Zika Virus Disease Patients. Front Immunol. 2021;12: 615102. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.615102.

    Article  Google Scholar 

  55. Walker LM, Bowley DR, Burton DR. Efficient Recovery of High-Affinity Antibodies from a Single-Chain Fab Yeast Display Library. J Mol Biol. 2009;389(2):365–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jmb.2009.04.019.

    Article  Google Scholar 

  56. Schoof M, Faust B, Saunders RA, Sangwan S, Rezelj V, Hoppe N, et al. An ultrapotent synthetic nanobody neutralizes SARS-CoV-2 by stabilizing inactive Spike. Science. 2020;370(6523):1473–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.abe3255.

    Article  Google Scholar 

  57. Mason DM, Weber CR, Parola C, Meng SM, Greiff V, Kelton WJ, et al. High-throughput antibody engineering in mammalian cells by CRISPR/Cas9-mediated homology-directed mutagenesis. Nucleic Acids Res. 2018;46(14):7436–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gky550.

    Article  Google Scholar 

  58. Chan KF, Shahreel W, Wan C, Teo G, Hayati N, Tay SJ, et al. Inactivation of GDP-fucose transporter gene (Slc35c1) in CHO cells by ZFNs, TALENs and CRISPR-Cas9 for production of fucose-free antibodies. Biotechnol J. 2016;11(3):399–414. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/biot.201500331.

    Article  Google Scholar 

  59. Rockberg J, Löfblom J, Hjelm B, Uhlén M, Ståhl S. Epitope mapping of antibodies using bacterial surface display. Nat Methods. 2008;5(12):1039–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nmeth.1272.

    Article  Google Scholar 

  60. Lee SJ, Bong JH, Jung J, Sung JS, Kang MJ, Jose J, et al. Screening of biotin-binding F(V)-antibodies from autodisplayed F(V)-library on E. coli outer membrane. Anal Chim Acta. 2021;1169:338627. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aca.2021.338627.

  61. Francisco JA, Campbell R, Iverson BL, Georgiou G. Production and fluorescence-activated cell sorting of Escherichia coli expressing a functional antibody fragment on the external surface. Proc Natl Acad Sci U S A. 1993;90(22):10444–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.90.22.10444.

    Article  Google Scholar 

  62. Mazor Y, Van Blarcom T, Iverson BL, Georgiou G. E-clonal antibodies: selection of full-length IgG antibodies using bacterial periplasmic display. Nat Protoc. 2008;3(11):1766–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nprot.2008.176.

    Article  Google Scholar 

  63. Lombana TN, Dillon M, Bevers J 3rd, Spiess C. Optimizing antibody expression by using the naturally occurring framework diversity in a live bacterial antibody display system. Sci Rep. 2015;5:17488. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep17488.

    Article  Google Scholar 

  64. Guo X, Cao H, Wang Y, Liu Y, Chen Y, Wang N, et al. Screening scFv antibodies against infectious bursal disease virus by co-expression of antigen and antibody in the bacteria display system. Vet Immunol Immunopathol. 2016;180:45–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.vetimm.2016.09.004.

    Article  Google Scholar 

  65. Mazor Y, Van Blarcom T, Carroll S, Georgiou G. Selection of full-length IgGs by tandem display on filamentous phage particles and Escherichia coli fluorescence-activated cell sorting screening. Febs j. 2010;277(10):2291–303. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1742-4658.2010.07645.x.

    Article  Google Scholar 

  66. Hu FJ, Volk AL, Persson H, Säll A, Borrebaeck C, Uhlen M, et al. Combination of phage and Gram-positive bacterial display of human antibody repertoires enables isolation of functional high affinity binders. N Biotechnol. 2018;45:80–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbt.2017.07.011.

    Article  Google Scholar 

  67. Salema V, Mañas C, Cerdán L, Piñero-Lambea C, Marín E, Roovers RC, et al. High affinity nanobodies against human epidermal growth factor receptor selected on cells by E. coli display. MAbs. 2016;8(7):1286–301. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/19420862.2016.1216742.

  68. Salema V, Marín E, Martínez-Arteaga R, Ruano-Gallego D, Fraile S, Margolles Y, et al. Selection of single domain antibodies from immune libraries displayed on the surface of E. coli cells with two β-domains of opposite topologies. PLoS One. 2013;8(9):e75126. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0075126.

  69. Chen PY, Chen YC, Chen PP, Lin KT, Sargsyan K, Hsu CP, et al. A whole-cell platform for discovering synthetic cell adhesion molecules in bacteria. Nat Commun. 2024;15(1):6568. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-024-51017-1.

    Article  Google Scholar 

  70. Salema V, Fernández L. Escherichia coli surface display for the selection of nanobodies. Microb Biotechnol. 2017;10(6):1468–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/1751-7915.12819.

    Article  Google Scholar 

  71. Hanes J, Jermutus L, Weber-Bornhauser S, Bosshard HR, Plückthun A. Ribosome display efficiently selects and evolves high-affinity antibodies in vitro from immune libraries. Proc Natl Acad Sci U S A. 1998;95(24):14130–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.95.24.14130.

    Article  Google Scholar 

  72. Kunamneni A, Ogaugwu C, Bradfute S, Durvasula R. Ribosome Display Technology: Applications in Disease Diagnosis and Control. Antibodies. 2020;9(3). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antib9030028.

  73. Thom G, Cockroft AC, Buchanan AG, Candotti CJ, Cohen ES, Lowne D, et al. Probing a protein-protein interaction by in vitro evolution. Proc Natl Acad Sci U S A. 2006;103(20):7619–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.0602341103.

    Article  Google Scholar 

  74. Hu M, Kang G, Cheng X, Wang J, Li R, Bai Z, et al. In vitro affinity maturation to improve the efficacy of a hypoxia-inducible factor 1α single-domain intrabody. Biochem Biophys Res Commun. 2020;529(4):936–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbrc.2020.06.097.

    Article  Google Scholar 

  75. Zimmermann I, Egloff P, Hutter CAJ, Kuhn BT, Bräuer P, Newstead S, et al. Generation of synthetic nanobodies against delicate proteins. Nat Protoc. 2020;15(5):1707–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41596-020-0304-x.

    Article  Google Scholar 

  76. Ohashi H, Kanamori T, Osada E, Akbar BK, Ueda T. Peptide screening using PURE ribosome display. Methods Mol Biol. 2012;805:251–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-1-61779-379-0_14.

    Article  Google Scholar 

  77. Wagner EK, Carter KP, Lim YW, Chau GJ, Enstrom A, Wayham NP, et al. High-throughput specificity profiling of antibody libraries using ribosome display and microfluidics. Cell Rep Methods. 2024;4(12): 100934. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.crmeth.2024.100934.

    Article  Google Scholar 

  78. Hanes J, Plückthun A. In vitro selection and evolution of functional proteins by using ribosome display. Proc Natl Acad Sci U S A. 1997;94(10):4937–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.94.10.4937.

    Article  Google Scholar 

  79. Zahnd C, Amstutz P, Plückthun A. Ribosome display: selecting and evolving proteins in vitro that specifically bind to a target. Nat Methods. 2007;4(3):269–79. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nmeth1003.

    Article  Google Scholar 

  80. Seifert M, Küppers R. Human memory B cells. Leukemia. 2016;30(12):2283–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/leu.2016.226.

    Article  Google Scholar 

  81. Attaf N, Baaklini S, Binet L, Milpied P. Heterogeneity of germinal center B cells: New insights from single-cell studies. Eur J Immunol. 2021;51(11):2555–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/eji.202149235.

    Article  Google Scholar 

  82. Lightwood DJ, Carrington B, Henry AJ, McKnight AJ, Crook K, Cromie K, et al. Antibody generation through B cell panning on antigen followed by in situ culture and direct RT-PCR on cells harvested en masse from antigen-positive wells. J Immunol Methods. 2006;316(1–2):133–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jim.2006.08.010.

    Article  Google Scholar 

  83. Yang Y, Li Z, Li Q, Ma K, Lin Y, Feng H, et al. Increase recombinant antibody yields through optimizing vector design and production process in CHO cells. Appl Microbiol Biotechnol. 2022;106(13–16):4963–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00253-022-12051-5.

    Article  Google Scholar 

  84. Ayyar BV, Arora S, Ravi SS. Optimizing antibody expression: The nuts and bolts. Methods. 2017;116:51–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ymeth.2017.01.009.

    Article  Google Scholar 

  85. Shi W, Hao H, Li M, Niu J, Hu Y, Zhao X, et al. Expression and Purification of a PEDV-Neutralizing Antibody and Its Functional Verification. Viruses. 2021;13(3). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/v13030472.

  86. Sblattero D, Bradbury A. A definitive set of oligonucleotide primers for amplifying human V regions. Immunotechnology. 1998;3(4):271–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s1380-2933(97)10004-5.

    Article  Google Scholar 

  87. Tiller T, Busse CE, Wardemann H. Cloning and expression of murine Ig genes from single B cells. J Immunol Methods. 2009;350(1–2):183–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jim.2009.08.009.

    Article  Google Scholar 

  88. Wang L, Madera R, Li Y, Gladue DP, Borca MV, McIntosh MT, et al. Development of Porcine Monoclonal Antibodies with In Vitro Neutralizing Activity against Classical Swine Fever Virus from C-Strain E2-Specific Single B Cells. Viruses. 2023;15(4). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/v15040863.

  89. Bautista D, Vásquez C, Ayala-Ramírez P, Téllez-Sosa J, Godoy-Lozano E, Martínez-Barnetche J, et al. Differential Expression of IgM and IgD Discriminates Two Subpopulations of Human Circulating IgM+IgD+CD27+ B Cells That Differ Phenotypically, Functionally, and Genetically. Frontiers in Immunology. 2020;11. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2020.00736.

  90. Carbonetti S, Oliver BG, Vigdorovich V, Dambrauskas N, Sack B, Bergl E, et al. A method for the isolation and characterization of functional murine monoclonal antibodies by single B cell cloning. J Immunol Methods. 2017;448:66–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jim.2017.05.010.

    Article  Google Scholar 

  91. Fan Y, Sun Z, Conrad F, Wen W, Zhao L, Lou J, et al. Multicolor fluorescence activated cell sorting to generate humanized monoclonal antibody binding seven subtypes of BoNT/F. PLoS ONE. 2022;17(9): e0273512. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0273512.

    Article  Google Scholar 

  92. Strassberger V, Gutbrodt KL, Krall N, Roesli C, Takizawa H, Manz MG, et al. A comprehensive surface proteome analysis of myeloid leukemia cell lines for therapeutic antibody development. J Proteomics. 2014;99:138–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jprot.2014.01.022.

    Article  Google Scholar 

  93. Zhou Y, Liu Z, Wang Z, Zhang Q, Mayer CT, Schoofs T, et al. Single-Cell Sorting of HBsAg-Binding Memory B Cells from Human Peripheral Blood Mononuclear Cells and Antibody Cloning. STAR Protocols. 2020;1(3). https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.xpro.2020.100129.

  94. Dippong M, Carl P, Lenz C, Schenk JA, Hoffmann K, Schwaar T, et al. Hapten-Specific Single-Cell Selection of Hybridoma Clones by Fluorescence-Activated Cell Sorting for the Generation of Monoclonal Antibodies. Anal Chem. 2017;89(7):4007–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.analchem.6b04569.

    Article  Google Scholar 

  95. Nutt SL, Hodgkin PD, Tarlinton DM, Corcoran LM. The generation of antibody-secreting plasma cells. Nat Rev Immunol. 2015;15(3):160–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri3795.

    Article  Google Scholar 

  96. Mikelov A, Alekseeva EI, Komech EA, Staroverov DB, Turchaninova MA, Shugay M, et al. Memory persistence and differentiation into antibody-secreting cells accompanied by positive selection in longitudinal BCR repertoires. Elife. 2022;11. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.79254.

  97. Ramirez K, Kyu S, Nguyen D, Han SY, Lee YL, Bradley J, et al. Heterofunctional Particles as Single Cell Sensors to Capture Secreted Immunoglobulins and Isolate Antigen-Specific Antibody Secreting Cells. Adv Healthc Mater. 2021;10(15): e2001947. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/adhm.202001947.

    Article  Google Scholar 

  98. Starkie DO, Compson JE, Rapecki S, Lightwood DJ. Generation of Recombinant Monoclonal Antibodies from Immunised Mice and Rabbits via Flow Cytometry and Sorting of Antigen-Specific IgG+ Memory B Cells. PLoS ONE. 2016;11(3): e0152282. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0152282.

    Article  Google Scholar 

  99. Broketa M, Bruhns P. Single-Cell Technologies for the Study of Antibody-Secreting Cells. Front Immunol. 2021;12: 821729. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.821729.

    Article  Google Scholar 

  100. Sun H, Hu N, Wang J. Application of microfluidic technology in antibody screening. Biotechnol J. 2022;17(8): e2100623. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/biot.202100623.

    Article  Google Scholar 

  101. Guo MT, Rotem A, Heyman JA, Weitz DA. Droplet microfluidics for high-throughput biological assays. Lab Chip. 2012;12(12):2146–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/c2lc21147e.

    Article  Google Scholar 

  102. Shembekar N, Hu H, Eustace D, Merten CA. Single-Cell Droplet Microfluidic Screening for Antibodies Specifically Binding to Target Cells. Cell Rep. 2018;22(8):2206–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2018.01.071.

    Article  Google Scholar 

  103. Mazutis L, Gilbert J, Ung WL, Weitz DA, Griffiths AD, Heyman JA. Single-cell analysis and sorting using droplet-based microfluidics. Nat Protoc. 2013;8(5):870–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nprot.2013.046.

    Article  Google Scholar 

  104. Eyer K, Doineau RCL, Castrillon CE, Briseño-Roa L, Menrath V, Mottet G, et al. Single-cell deep phenotyping of IgG-secreting cells for high-resolution immune monitoring. Nat Biotechnol. 2017;35(10):977–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nbt.3964.

    Article  Google Scholar 

  105. Josephides D, Davoli S, Whitley W, Ruis R, Salter R, Gokkaya S, et al. Cyto-Mine: An Integrated, Picodroplet System for High-Throughput Single-Cell Analysis, Sorting, Dispensing, and Monoclonality Assurance. SLAS Technol. 2020;25(2):177–89. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/2472630319892571.

    Article  Google Scholar 

  106. Gaa R, Menang-Ndi E, Pratapa S, Nguyen C, Kumar S, Doerner A. Versatile and rapid microfluidics-assisted antibody discovery. MAbs. 2021;13(1):1978130. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/19420862.2021.1978130.

    Article  Google Scholar 

  107. Gérard A, Woolfe A, Mottet G, Reichen M, Castrillon C, Menrath V, et al. High-throughput single-cell activity-based screening and sequencing of antibodies using droplet microfluidics. Nat Biotechnol. 2020;38(6):715–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41587-020-0466-7.

    Article  Google Scholar 

  108. Hillenbrand M, Esslinger C, Seidenberg J, Weber M, Zingg A, Townsend C, et al. Fast-Track Discovery of SARS-CoV-2-Neutralizing Antibodies from Human B Cells by Direct Functional Screening. Viruses. 2024;16(3). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/v16030339.

  109. Adler AS, Mizrahi RA, Spindler MJ, Adams MS, Asensio MA, Edgar RC, et al. Rare, high-affinity anti-pathogen antibodies from human repertoires, discovered using microfluidics and molecular genomics. MAbs. 2017;9(8):1282–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/19420862.2017.1371383.

    Article  Google Scholar 

  110. DeKosky BJ, Kojima T, Rodin A, Charab W, Ippolito GC, Ellington AD, et al. In-depth determination and analysis of the human paired heavy- and light-chain antibody repertoire. Nat Med. 2015;21(1):86–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nm.3743.

    Article  Google Scholar 

  111. Wippold JA, Wang H, Tingling J, Leibowitz JL, de Figueiredo P, Han A. PRESCIENT: platform for the rapid evaluation of antibody success using integrated microfluidics enabled technology. Lab Chip. 2020;20(9):1628–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/c9lc01165j.

    Article  Google Scholar 

  112. Lyu M, Shi X, Liu X, Liu Y, Zhu X, Liao L, et al. Generation and Screening of Antigen-Specific Nanobodies from Mammalian Cells Expressing the BCR Repertoire Library Using Droplet-Based Microfluidics. Anal Chem. 2022;94(22):7970–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.analchem.2c00865.

    Article  Google Scholar 

  113. Wang Y, Jin R, Shen B, Li N, Zhou H, Wang W, et al. High-throughput functional screening for next-generation cancer immunotherapy using droplet-based microfluidics. Sci Adv. 2021;7(24). https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.abe3839.

  114. Rutkauskaite J, Berger S, Stavrakis S, Dressler O, Heyman J, Casadevall ISX, et al. High-throughput single-cell antibody secretion quantification and enrichment using droplet microfluidics-based FRET assay. iScience. 2022;25(7):104515. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.isci.2022.104515.

  115. Ahmadi F, Tran H, Letourneau N, Little SR, Fortin A, Moraitis AN, et al. An Automated Single-Cell Droplet-Digital Microfluidic Platform for Monoclonal Antibody Discovery. Small. 2024;20(26): e2308950. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/smll.202308950.

    Article  Google Scholar 

  116. Love JC, Ronan JL, Grotenbreg GM, van der Veen AG, Ploegh HL. A microengraving method for rapid selection of single cells producing antigen-specific antibodies. Nat Biotechnol. 2006;24(6):703–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nbt1210.

    Article  Google Scholar 

  117. Ozawa T, Piao X, Kobayashi E, Zhou Y, Sakurai H, Andoh T, et al. A novel rabbit immunospot array assay on a chip allows for the rapid generation of rabbit monoclonal antibodies with high affinity. PLoS ONE. 2012;7(12): e52383. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0052383.

    Article  Google Scholar 

  118. Sendra VG, Lie A, Romain G, Agarwal SK, Varadarajan N. Detection and isolation of auto-reactive human antibodies from primary B cells. Methods. 2013;64(2):153–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ymeth.2013.06.018.

    Article  Google Scholar 

  119. Ogunniyi AO, Story CM, Papa E, Guillen E, Love JC. Screening individual hybridomas by microengraving to discover monoclonal antibodies. Nat Protoc. 2009;4(5):767–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nprot.2009.40.

    Article  Google Scholar 

  120. Horta S, Neumann F, Yeh SH, Langseth CM, Kangro K, Breukers J, et al. Evaluation of Immuno-Rolling Circle Amplification for Multiplex Detection and Profiling of Antigen-Specific Antibody Isotypes. Anal Chem. 2021;93(15):6169–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.analchem.1c00172.

    Article  Google Scholar 

  121. Ogunniyi AO, Thomas BA, Politano TJ, Varadarajan N, Landais E, Poignard P, et al. Profiling human antibody responses by integrated single-cell analysis. Vaccine. 2014;32(24):2866–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.vaccine.2014.02.020.

    Article  Google Scholar 

  122. Esfandiary L, Gupta N, Voigt A, Wanchoo A, Chan EK, Sukumaran S, et al. Single-cell antibody nanowells: a novel technology in detecting anti-SSA/Ro60- and anti-SSB/La autoantibody-producing cells in peripheral blood of rheumatic disease patients. Arthritis Res Ther. 2016;18(1):107. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-016-1010-5.

    Article  Google Scholar 

  123. Li Y, Li P, Ke Y, Yu X, Yu W, Wen K, et al. A rare monoclonal antibody discovery based on indirect competitive screening of a single hapten-specific rabbit antibody secreting cell. Analyst. 2022;147(13):2942–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/d2an00678b.

    Article  Google Scholar 

  124. Abali F, Schasfoort R, Nijland S, Wittenberns J, Tibbe AGJ, den Hartog M, et al. A nanowell platform to identify, sort and expand high antibody-producing cells. Sci Rep. 2024;14(1):9457. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-60054-1.

    Article  Google Scholar 

  125. Jorgolli M, Nevill T, Winters A, Chen I, Chong S, Lin FF, et al. Nanoscale integration of single cell biologics discovery processes using optofluidic manipulation and monitoring. Biotechnol Bioeng. 2019;116(9):2393–411. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/bit.27024.

    Article  Google Scholar 

  126. Hsu H-y, Ohta AT, Chiou P-Y, Jamshidi A, Neale SL, Wu MC. Phototransistor-based optoelectronic tweezers for dynamic cell manipulation in cell culture media. Lab Chip. 2010;10(2):165–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/b906593h.

  127. Mullen TE, Abdullah R, Boucher J, Brousseau AS, Dasuri NK, Ditto NT, et al. Accelerated antibody discovery targeting the SARS-CoV-2 spike protein for COVID-19 therapeutic potential. Antibody Therapeutics. 2021;4(3):185–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/abt/tbab018.

    Article  Google Scholar 

  128. Winters A, McFadden K, Bergen J, Landas J, Berry KA, Gonzalez A, et al. Rapid single B cell antibody discovery using nanopens and structured light. mAbs. 2019;11(6):1025–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/19420862.2019.1624126.

  129. Desmurget C, Frentzel J, Strembitska A, Sobkowiak K, Perilleux A, Souquet J, et al. Combined approach of selective and accelerated cloning for microfluidic chip-based system increases clone specific productivity. Biotechnol J. 2024;19(5): e2300488. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/biot.202300488.

    Article  Google Scholar 

  130. Shapiro MB, Boucher J, Brousseau A, Dehkharghani A, Gabriel J, Kamat V, et al. Alpaca single B cell interrogation and heavy-chain-only antibody discovery on an optofluidic platform. Antib Ther. 2023;6(3):211–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/abt/tbad018.

    Article  Google Scholar 

  131. Zost SJ, Gilchuk P, Chen RE, Case JB, Reidy JX, Trivette A, et al. Rapid isolation and profiling of a diverse panel of human monoclonal antibodies targeting the SARS-CoV-2 spike protein. Nat Med. 2020;26(9):1422–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41591-020-0998-x.

    Article  Google Scholar 

  132. Zhang W, Li Q, Jia F, Hu Z, Wei Z. A Microfluidic Chip for Screening and Sequencing of Monoclonal Antibody at a Single-Cell Level. Anal Chem. 2021;93(29):10099–105. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.analchem.1c00918.

    Article  Google Scholar 

  133. Watson CT, Breden F. The immunoglobulin heavy chain locus: genetic variation, missing data, and implications for human disease. Genes Immun. 2012;13(5):363–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/gene.2012.12.

    Article  Google Scholar 

  134. Kivioja T, Vähärautio A, Karlsson K, Bonke M, Enge M, Linnarsson S, et al. Counting absolute numbers of molecules using unique molecular identifiers. Nat Methods. 2011;9(1):72–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nmeth.1778.

    Article  Google Scholar 

  135. Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H, et al. Simultaneous epitope and transcriptome measurement in single cells. Nat Methods. 2017;14(9):865–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nmeth.4380.

    Article  Google Scholar 

  136. Setliff I, Shiakolas AR, Pilewski KA, Murji AA, Mapengo RE, Janowska K, et al. High-Throughput Mapping of B Cell Receptor Sequences to Antigen Specificity. Cell. 2019;179(7):1636-46.e15. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2019.11.003.

    Article  Google Scholar 

  137. Kramer KJ, Johnson NV, Shiakolas AR, Suryadevara N, Periasamy S, Raju N, et al. Potent neutralization of SARS-CoV-2 variants of concern by an antibody with an uncommon genetic signature and structural mode of spike recognition. Cell Rep. 2021;37(1): 109784. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2021.109784.

    Article  Google Scholar 

  138. Pilewski KA, Wall S, Richardson SI, Manamela NP, Clark K, Hermanus T, et al. Functional HIV-1/HCV cross-reactive antibodies isolated from a chronically co-infected donor. Cell Rep. 2023;42(2): 112044. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2023.112044.

    Article  Google Scholar 

  139. Shiakolas AR, Kramer KJ, Johnson NV, Wall SC, Suryadevara N, Wrapp D, et al. Efficient discovery of SARS-CoV-2-neutralizing antibodies via B cell receptor sequencing and ligand blocking. Nat Biotechnol. 2022;40(8):1270–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41587-022-01232-2.

    Article  Google Scholar 

  140. Walker LM, Shiakolas AR, Venkat R, Liu ZA, Wall S, Raju N, et al. High-Throughput B Cell Epitope Determination by Next-Generation Sequencing. Front Immunol. 2022;13: 855772. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.855772.

    Article  Google Scholar 

  141. Vukovich MJ, Raju N, Kgagudi P, Manamela NP, Abu-Shmais AA, Gripenstraw KR, et al. Development of LIBRA-seq for the guinea pig model system as a tool for the evaluation of antibody responses to multivalent HIV-1 vaccines. J Virol. 2024;98(1): e0147823. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/jvi.01478-23.

    Article  Google Scholar 

  142. Vukovich MJ, Shiakolas AR, Lindenberger J, Richardson RA, Bass LE, Barr M, et al. Isolation and characterization of IgG3 glycan-targeting antibodies with exceptional cross-reactivity for diverse viral families. PLoS Pathog. 2024;20(9): e1012499. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.ppat.1012499.

    Article  Google Scholar 

  143. Nguyen NK, Devilder MC, Gautreau-Rolland L, Fourgeux C, Sinha D, Poschmann J, et al. A cluster of broadly neutralizing IgG against BK polyomavirus in a repertoire dominated by IgM. Life Sci Alliance. 2023;6(4). https://doiorg.publicaciones.saludcastillayleon.es/10.26508/lsa.202201567.

  144. Gao C, Zhang M, Chen L. The Comparison of Two Single-cell Sequencing Platforms: BD Rhapsody and 10x Genomics Chromium. Curr Genomics. 2020;21(8):602–9. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1389202921999200625220812.

    Article  Google Scholar 

  145. Goldstein LD, Chen Y-JJ, Wu J, Chaudhuri S, Hsiao Y-C, Schneider K, et al. Massively parallel single-cell B-cell receptor sequencing enables rapid discovery of diverse antigen-reactive antibodies. Commun Biol. 2019;2(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42003-019-0551-y.

  146. Klein Allon M, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, et al. Droplet Barcoding for Single-Cell Transcriptomics Applied to Embryonic Stem Cells. Cell. 2015;161(5):1187–201. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2015.04.044.

    Article  Google Scholar 

  147. Li F, Luo M, Zhou W, Li J, Jin X, Xu Z, et al. Single cell RNA and immune repertoire profiling of COVID-19 patients reveal novel neutralizing antibody. Protein Cell. 2021;12(10):751–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13238-020-00807-6.

    Article  Google Scholar 

  148. Cao Y, Yisimayi A, Bai Y, Huang W, Li X, Zhang Z, et al. Humoral immune response to circulating SARS-CoV-2 variants elicited by inactivated and RBD-subunit vaccines. Cell Res. 2021;31(7):732–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41422-021-00514-9.

    Article  Google Scholar 

  149. Yang X, Chi H, Wu M, Wang Z, Lang Q, Han Q, et al. Discovery and characterization of SARS-CoV-2 reactive and neutralizing antibodies from humanized CAMouse(HG) mice through rapid hybridoma screening and high-throughput single-cell V(D)J sequencing. Front Immunol. 2022;13: 992787. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.992787.

    Article  Google Scholar 

  150. Cao Y, Su B, Guo X, Sun W, Deng Y, Bao L, et al. Potent Neutralizing Antibodies against SARS-CoV-2 Identified by High-Throughput Single-Cell Sequencing of Convalescent Patients’ B Cells. Cell. 2020;182(1):73-84.e16. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2020.05.025.

    Article  Google Scholar 

  151. Wang W, Gu Y, Ou Y, Zhou J, Liu B, Zuo H, et al. Human monoclonal antibodies against Staphylococcus aureus A protein identified by high-throughput single-cell sequencing of phase I clinical volunteers’ B cells. Clin Immunol. 2023;257: 109843. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.clim.2023.109843.

    Article  Google Scholar 

  152. Irac SE, Soon MSF, Borcherding N, Tuong ZK. Single-cell immune repertoire analysis. Nat Methods. 2024;21(5):777–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41592-024-02243-4.

    Article  Google Scholar 

  153. Ye J, Ma N, Madden TL, Ostell JM. IgBLAST: an immunoglobulin variable domain sequence analysis tool. Nucleic Acids Res. 2013;41(Web Server issue):W34–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gkt382.

  154. Alamyar E, Duroux P, Lefranc MP, Giudicelli V. IMGT(®) tools for the nucleotide analysis of immunoglobulin (IG) and T cell receptor (TR) V-(D)-J repertoires, polymorphisms, and IG mutations: IMGT/V-QUEST and IMGT/HighV-QUEST for NGS. Methods Mol Biol. 2012;882:569–604. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-1-61779-842-9_32.

    Article  Google Scholar 

  155. Abhinandan KR, Martin AC. Analysis and improvements to Kabat and structurally correct numbering of antibody variable domains. Mol Immunol. 2008;45(14):3832–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molimm.2008.05.022.

    Article  Google Scholar 

  156. Smakaj E, Babrak L, Ohlin M, Shugay M, Briney B, Tosoni D, et al. Benchmarking immunoinformatic tools for the analysis of antibody repertoire sequences. Bioinformatics. 2020;36(6):1731–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/btz845.

    Article  Google Scholar 

  157. Zong F, Long C, Hu W, Chen S, Dai W, Xiao ZX, et al. Abalign: a comprehensive multiple sequence alignment platform for B-cell receptor immune repertoires. Nucleic Acids Res. 2023;51(W1):W17-w24. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gkad400.

    Article  Google Scholar 

  158. Shouval DS, Nagar M, Salamon N, Dor C, Werner L. T and B Cell Receptor Immune Repertoire Analysis using Next-generation Sequencing. J Visual Exper. 2021(167). https://doiorg.publicaciones.saludcastillayleon.es/10.3791/61792.

  159. Bai G, Sun C, Guo Z, Wang Y, Zeng X, Su Y, et al. Accelerating antibody discovery and design with artificial intelligence: Recent advances and prospects. Semin Cancer Biol. 2023;95:13–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcancer.2023.06.005.

    Article  Google Scholar 

  160. Ledsgaard L, Ljungars A, Rimbault C, Sørensen CV, Tulika T, Wade J, et al. Advances in antibody phage display technology. Drug Discovery Today. 2022;27(8):2151–69. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.drudis.2022.05.002.

    Article  Google Scholar 

  161. Pedrioli A, Oxenius A. Single B cell technologies for monoclonal antibody discovery. Trends Immunol. 2021;42(12):1143–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.it.2021.10.008.

    Article  Google Scholar 

  162. Omidfar K, Kashanian S. A mini review on recent progress of microfluidic systems for antibody development. J Diabetes Metab Disord. 2024;23(1):323–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40200-024-01386-7.

    Article  Google Scholar 

Download references

Acknowledgements

N/A.

Funding

This work was supported by the National Natural Science Foundation of China (82270013, 81870016), Guangdong Basic and Applied Basic Research Foundation (2022A1515010525), Science and Technology Project of Dongguan (20211800905072), Medical Research Foundation of Guangdong Province (A2024616), Innovation Team Project of Universities in Guangdong Province (2022KCXTD010), Talent Development Foundation of The First Dongguan Affiliated Hospital of Guangdong Medical University (PU2023003) , Talent Development Foundation of The First Dongguan Affiliated Hospital of Guangdong Medical University & Foundation of State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia (SKL-HIDCA-2024- GD6B), Project of Songshan Lake Innovation Center of Medicine & Engineering of Guangdong Medical University (4SG22317P) and Discipline construction project of Guangdong Medical University (4SG24228G, 4SG23030G, 4SG24025G, 4SG24044G, 4SG24129G, 4SG23143G and 4SG23077G).

Author information

Authors and Affiliations

Authors

Contributions

X.W. and P.Z. conceptualized the manuscript and wrote the draft. B.M. prepared the figures (Figures 1–3) and the table. All authors reviewed and edited the manuscript. X.W. and P.Z. supervised the project. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Yan-Guang Cong, Jun-Fa Xu or Peng-Fei Zhang.

Ethics declarations

Ethics approval and consent to participate

N/A.

Consent for publication

N/A.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, XD., Ma, BY., Lai, SY. et al. High-throughput strategies for monoclonal antibody screening: advances and challenges. J Biol Eng 19, 41 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13036-025-00513-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13036-025-00513-z

Keywords