Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.224
Filtrer
1.
MAbs ; 16(1): 2379903, 2024.
Article de Anglais | MEDLINE | ID: mdl-39077932

RÉSUMÉ

A sensitive and specific bioanalytical method was required to measure the exposure of a LAGA-mutated surrogate mouse IgG2a monoclonal antibody in mouse plasma, but the lack of highly specific reagents for the LAGA mutant hindered the development of a ligand-binding assay. Equally problematic is that no sensitive unique tryptic peptides suitable for quantitative mass spectrometric analysis could be identified in the mIgG2a complementarity-determining regions. To overcome these challenges, a trypsin alternative pepsin, an aspartic protease, was systematically investigated for its use in digesting the mutated mIgG2a antibody to allow generation of signature peptides for the bioanalytical quantification purpose. After a series of evaluations, a rapid one-hour pepsin digestion protocol was established for the mutated Fc backbone. Consequently, a new pepsin digestion-based liquid chromatography-tandem mass spectrometry (LC/MS/MS) method was successfully developed to support the mouse pharmacokinetic (PK) sample analysis. In brief, robust and reproducible C-terminal cleavage of both leucine and phenylalanine near the double mutation site of the mutated mIgG2a was accomplished at pH ≤2 and 37°C. Combined with a commercially available rat anti-mIgG2a heavy-chain antibody, the established immunoaffinity LC/MS/MS assay achieved a limit of quantitation of 20 ng/mL in the dynamic range of interest with satisfactory assay precision and accuracy. The successful implementation of this novel approach in discovery PK studies eliminates the need for tedious and costly generation of specific immunocapturing reagents for the LAGA mutants. The approach should be widely applicable for developing popular LAGA mutant-based biological therapeutics.


Sujet(s)
Immunoglobuline G , Pepsine A , Spectrométrie de masse en tandem , Animaux , Immunoglobuline G/génétique , Immunoglobuline G/immunologie , Spectrométrie de masse en tandem/méthodes , Souris , Chromatographie en phase liquide/méthodes , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/génétique , Rats , Mutation , Chromatographie d'affinité/méthodes
2.
Methods Mol Biol ; 2829: 175-183, 2024.
Article de Anglais | MEDLINE | ID: mdl-38951333

RÉSUMÉ

Monoclonal antibodies have widespread applications in disease treatment and antigen detection. They are traditionally produced using mammalian cell expression system, which is not able to satisfy the increasing demand of these proteins at large scale. Baculovirus expression vector system (BEVS) is an attractive alternative platform for the production of biologically active monoclonal antibodies. In this chapter, we demonstrate the production of an HIV-1 broadly neutralizing antibody b12 in BEVS. The processes including transfer vector construction, recombinant baculovirus generation, and antibody production and detection are described.


Sujet(s)
Baculoviridae , Vecteurs génétiques , Baculoviridae/génétique , Vecteurs génétiques/génétique , Animaux , Humains , Expression des gènes , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Protéines recombinantes/génétique , Protéines recombinantes/biosynthèse , Anticorps monoclonaux/génétique , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Test ELISA , Anticorps anti-VIH/immunologie , Anticorps anti-VIH/génétique , Cellules Sf9
3.
Methods Mol Biol ; 2826: 231-249, 2024.
Article de Anglais | MEDLINE | ID: mdl-39017897

RÉSUMÉ

The hybridoma method for production of monoclonal antibodies has been a cornerstone of biomedical research for several decades. Here we convert the monoclonal antibody sequence from mouse-derived hybridomas into a "devilized" recombinant antibody with devil IgG heavy chain and IgK light chain. The chimeric recombinant antibody can be used in functional assays, immunotherapy, and to improve understanding of antibodies and Fc receptors in Tasmanian devils. The process can be readily modified for other species.


Sujet(s)
Hybridomes , Immunoglobuline G , Marsupialia , Animaux , Souris , Immunoglobuline G/génétique , Immunoglobuline G/immunologie , Hybridomes/immunologie , Marsupialia/immunologie , Marsupialia/génétique , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/génétique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie
4.
Monoclon Antib Immunodiagn Immunother ; 43(4): 119-126, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39034896

RÉSUMÉ

Mammalian cell line stability is an important consideration when establishing a biologics manufacturing process in the biopharmaceutical and in vitro diagnostics (IVD) industries. Traditional Chinese hamster ovary (CHO) cell line development methods use a random integration approach that requires transfection, selection, optional amplification, screenings, and single-cell cloning to select clones with acceptable productivity, product quality, and genetic stability. Site-specific integration reduces these disadvantages, and new technologies have been developed to mitigate risks associated with genetic instability. In this study, we applied the Leap-In® transposase-mediated expression system from ATUM to generate stable CHOK1 pools for the production of four recombinant antibody reagents for IVD immunoassays. CHO cell line stability is defined by consistent antibody production over time. Three of the CHOK1 pools maintained productivity suitable for manufacturing, with high antibody yields. The productivity of the remaining CHOK1 pool decreased over time; however, derivative clones showed acceptable stability. l-glutamine had variable effects on CHOK1 cell line or stable pool stability and significantly affected antibody product titer. Compared with traditional random integration methods, the ATUM Leap-In system can reduce the time needed to develop new immunoassays by using semi site-specific integration to generate high-yield stable pools that meet manufacturing stability requirements.


Sujet(s)
Cricetulus , Protéines recombinantes , Cellules CHO , Animaux , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , Protéines recombinantes/biosynthèse , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/génétique , Cricetinae , Humains , Transposases/génétique , Transposases/métabolisme
5.
Sci Rep ; 14(1): 14587, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918509

RÉSUMÉ

Engineered mammalian cells are key for biotechnology by enabling broad applications ranging from in vitro model systems to therapeutic biofactories. Engineered cell lines exist as a population containing sub-lineages of cell clones that exhibit substantial genetic and phenotypic heterogeneity. There is still a limited understanding of the source of this inter-clonal heterogeneity as well as its implications for biotechnological applications. Here, we developed a genomic barcoding strategy for a targeted integration (TI)-based CHO antibody producer cell line development process. This technology provided novel insights about clone diversity during stable cell line selection on pool level, enabled an imaging-independent monoclonality assessment after single cell cloning, and eventually improved hit-picking of antibody producer clones by monitoring of cellular lineages during the cell line development (CLD) process. Specifically, we observed that CHO producer pools generated by TI of two plasmids at a single genomic site displayed a low diversity (< 0.1% RMCE efficiency), which further depends on the expressed molecules, and underwent rapid population skewing towards dominant clones during routine cultivation. Clonal cell lines from one individual TI event demonstrated a significantly lower variance regarding production-relevant and phenotypic parameters as compared to cell lines from distinct TI events. This implies that the observed cellular diversity lies within pre-existing cell-intrinsic factors and that the majority of clonal variation did not develop during the CLD process, especially during single cell cloning. Using cellular barcodes as a proxy for cellular diversity, we improved our CLD screening workflow and enriched diversity of production-relevant parameters substantially. This work, by enabling clonal diversity monitoring and control, paves the way for an economically valuable and data-driven CLD process.


Sujet(s)
Clones cellulaires , Cricetulus , Codage à barres de l'ADN pour la taxonomie , Cellules CHO , Animaux , Codage à barres de l'ADN pour la taxonomie/méthodes , Génomique/méthodes , Anticorps monoclonaux/génétique
6.
J Biosci Bioeng ; 138(2): 171-180, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38806389

RÉSUMÉ

Chinese hamster ovary (CHO) cells are the most widely used for therapeutic antibody production. In cell line development, engineering secretion processes such as folding-related protein upregulation is an effective way of constructing cell lines with high recombinant protein productivity. However, there have been few studies on the transport of recombinant proteins between the endoplasmic reticulum (ER) and the Golgi apparatus. In this study, Sar1A, a protein involved in COPII vesicle formation, was focused on to improve antibody productivity by enhancing COPII vesicle-mediated antibody transport from the ER to the Golgi apparatus, and to clarify its effect on the secretion process. The constructed Sar1A-overexpressing CHO cell lines were batch-cultured, in which they showed an increased specific antibody production rate. The intracellular antibody accumulation and the specific localization of the intracellular antibodies were investigated by chase assay using a translation inhibitor and observed by immunofluorescence-based imaging analysis. The results showed that Sar1A overexpression reduced intracellular antibody accumulation, especially in the ER. The effects of the engineered antibody transport on the antibody's glycosylation profile and the unfolded protein response (UPR) pathway were analyzed by liquid chromatography-mass spectrometry and UPR-related gene expression evaluation, respectively. Sar1A overexpression lowered glycan galactosylation and induced a stronger UPR at the end of the batch culture. Sar1A overexpression enhanced the antibody productivity of CHO cells by modifying their secretion process. This approach could also contribute to the production of not only monoclonal antibodies but also other therapeutic proteins that require transport by COPII vesicles.


Sujet(s)
Cricetulus , Réticulum endoplasmique , Appareil de Golgi , Protéines recombinantes , Cellules CHO , Animaux , Réticulum endoplasmique/métabolisme , Protéines recombinantes/métabolisme , Protéines recombinantes/génétique , Appareil de Golgi/métabolisme , Glycosylation , Cricetinae , Réponse aux protéines mal repliées , Vésicules COP/métabolisme , Anticorps monoclonaux/métabolisme , Anticorps monoclonaux/génétique , Transport des protéines , Techniques de culture cellulaire en batch/méthodes
7.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1536-1547, 2024 May 25.
Article de Chinois | MEDLINE | ID: mdl-38783814

RÉSUMÉ

The aim of this study was to prepare a mouse monoclonal antibody against the nonstructural protein 1 (NS1) of respiratory syncytial virus (RSV) to analyze its expression and distribution during transfection and infection. Additionally, we aimed to evaluate the antibody's application in immunoprecipitation assay. Firstly, the NS1 gene fragment was cloned into a prokaryotic plasmid and expressed in Escherichia coli. The resulting NS1 protein was then purified by affinity chromatography, and used to immunize the BALB/c mice. Subsequently, hybridoma cells capable of stably secreting the NS1 monoclonal antibody were selected using indirect enzyme linked immunosorbent assay (ELISA). This monoclonal antibody was employed in both indirect immunofluorescence assay (IFA) and Western blotting to analyze the expression and distribution of RSV NS1 in overexpressed and infected cells. Finally, the reliability of this monoclonal antibody was evaluated through the immunoprecipitation assay. The results showed that the RSV NS1 protein was successfully expressed and purified. Following immunization of mice with this protein, we obtained a highly specific RSV NS1 monoclonal antibody, which belonged to the IgG1 subtype with an antibody titer of 1:15 360 000. Using this monoclonal antibody, the RSV NS1 protein was identified in both transfected and infected cells. The IFA results revealed predominant distribution of NS1 in the cytoplasm and nucleus. Moreover, we confirmed that this monoclonal antibody could effectively bind specifically to NS1 protein in cell lysates, making it suitable as a capture antibody in immunoprecipitation assay. In conclusion, our study successfully achieved production of the RSV NS1 protein through a prokaryotic expression system and prepared a specific monoclonal antibody against NS1. This antibody demonstrates the ability to specifically identify the NS1 protein and can be used in the immunoprecipitation assay, thereby laying a foundation for the functional studies of the NS1 protein.


Sujet(s)
Anticorps monoclonaux , Protéines virales non structurales , Animaux , Femelle , Souris , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/génétique , Anticorps antiviraux/immunologie , Escherichia coli/génétique , Escherichia coli/métabolisme , Hybridomes/immunologie , Souris de lignée BALB C , Virus respiratoires syncytiaux/immunologie , Virus respiratoires syncytiaux/génétique , Protéines virales non structurales/immunologie , Protéines virales non structurales/génétique
8.
Hum Antibodies ; 32(3): 107-120, 2024.
Article de Anglais | MEDLINE | ID: mdl-38788063

RÉSUMÉ

Monoclonal antibody biologics have significantly transformed the therapeutic landscape within the biopharmaceutical industry, partly due to the utilisation of discovery technologies such as the hybridoma method and phage display. While these established platforms have streamlined the development process to date, their reliance on cell transformation for antibody identification faces limitations related to library diversification and the constraints of host cell physiology. Cell-free systems like ribosome display offer a complementary approach, enabling antibody selection in a completely in vitro setting while harnessing enriched cellular molecular machinery. This review aims to provide an overview of the fundamental principles underlying the ribosome display method and its potential for advancing antibody discovery and development.


Sujet(s)
Anticorps monoclonaux , Banque de peptides , Ribosomes , Ribosomes/immunologie , Humains , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/génétique , Animaux , Techniques d'exposition à la surface cellulaire , Découverte de médicament , Eucaryotes/immunologie , Eucaryotes/génétique
9.
Protein Expr Purif ; 220: 106499, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38703798

RÉSUMÉ

Monoclonal antibodies (mAbs) are a driving force in the biopharmaceutical industry. Therapeutic mAbs are usually produced in mammalian cells, but there has been a push towards the use of alternative production hosts, such as Escherichia coli. When the genes encoding for a mAb heavy and light chains are codon-optimized for E. coli expression, a truncated form of the heavy chain can form along with the full-length product. In this work, the role of codon optimization in the formation of a truncated product was investigated. This study used the amino acid sequences of several therapeutic mAbs and multiple optimization algorithms. It was found that several algorithms incorporate sequences that lead to a truncated product. Approaches to avoid this truncated form are discussed.


Sujet(s)
Anticorps monoclonaux , Escherichia coli , Anticorps monoclonaux/génétique , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/composition chimique , Escherichia coli/génétique , Escherichia coli/métabolisme , Codon/génétique , Algorithmes , Séquence d'acides aminés , Protéines recombinantes/génétique , Protéines recombinantes/biosynthèse , Protéines recombinantes/composition chimique , Humains , Expression des gènes , Chaines lourdes des immunoglobulines/génétique , Chaines lourdes des immunoglobulines/composition chimique
10.
Front Immunol ; 15: 1341389, 2024.
Article de Anglais | MEDLINE | ID: mdl-38698845

RÉSUMÉ

Monoclonal antibodies (mAbs) are one of the most important classes of biologics with high therapeutic and diagnostic value, but traditional methods for mAbs generation, such as hybridoma screening and phage display, have limitations, including low efficiency and loss of natural chain pairing. To overcome these challenges, novel single B cell antibody technologies have emerged, but they also have limitations such as in vitro differentiation of memory B cells and expensive cell sorters. In this study, we present a rapid and efficient workflow for obtaining human recombinant monoclonal antibodies directly from single antigen-specific antibody secreting cells (ASCs) in the peripheral blood of convalescent COVID-19 patients using ferrofluid technology. This process allows the identification and expression of recombinant antigen-specific mAbs in less than 10 days, using RT-PCR to generate linear Ig heavy and light chain gene expression cassettes, called "minigenes", for rapid expression of recombinant antibodies without cloning procedures. This approach has several advantages. First, it saves time and resources by eliminating the need for in vitro differentiation. It also allows individual antigen-specific ASCs to be screened for effector function prior to recombinant antibody cloning, enabling the selection of mAbs with desired characteristics and functional activity. In addition, the method allows comprehensive analysis of variable region repertoires in combination with functional assays to evaluate the specificity and function of the generated antigen-specific antibodies. Our approach, which rapidly generates recombinant monoclonal antibodies from single antigen-specific ASCs, could help to identify functional antibodies and deepen our understanding of antibody dynamics in the immune response through combined antibody repertoire sequence analysis and functional reactivity testing.


Sujet(s)
Anticorps monoclonaux , Cellules productrices d'anticorps , COVID-19 , Protéines recombinantes , SARS-CoV-2 , Humains , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/génétique , Anticorps monoclonaux/biosynthèse , Protéines recombinantes/immunologie , Protéines recombinantes/génétique , Cellules productrices d'anticorps/immunologie , SARS-CoV-2/immunologie , COVID-19/immunologie , Anticorps antiviraux/immunologie , Femelle
11.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38732011

RÉSUMÉ

Immunoglobulin G-based monoclonal antibodies (mAbs) have been effective in treating various diseases, but their large molecular size can limit their penetration of tissue and efficacy in multifactorial diseases, necessitating the exploration of alternative forms. In this study, we constructed a phage display library comprising single-domain antibodies (sdAbs; or "VHHs"), known for their small size and remarkable stability, using a total of 1.6 × 109 lymphocytes collected from 20 different alpacas, resulting in approximately 7.16 × 1010 colonies. To assess the quality of the constructed library, next-generation sequencing-based high-throughput profiling was performed, analyzing approximately 5.65 × 106 full-length VHH sequences, revealing 92% uniqueness and confirming the library's diverse composition. Systematic characterization of the library revealed multiple sdAbs with high affinity for three therapeutically relevant antigens. In conclusion, our alpaca sdAb phage display library provides a versatile resource for diagnostics and therapeutics. Furthermore, the library's vast natural VHH antibody repertoire offers insights for generating humanized synthetic sdAb libraries, further advancing sdAb-based therapeutics.


Sujet(s)
Camélidés du Nouveau Monde , Banque de peptides , Anticorps à domaine unique , Anticorps à domaine unique/génétique , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/composition chimique , Animaux , Camélidés du Nouveau Monde/immunologie , Séquençage nucléotidique à haut débit , Humains , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/génétique , Tests de criblage à haut débit/méthodes , Affinité des anticorps , Techniques d'exposition à la surface cellulaire/méthodes
12.
J Biotechnol ; 389: 30-42, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38685416

RÉSUMÉ

Ichnos has developed a multi-specific antibody platform based on the BEAT® (Bispecific engagement by antibodies based on the T-cell receptor) interface. The increased complexity of the bi- and multi-specific formats generated with this platform makes these molecules difficult-to-express proteins compared to standard monoclonal antibodies (mAbs). This report describes how expression limitations of a bi-specific bi-paratopic BEAT antibody were improved in a holistic approach. An initial investigation allowed identification of a misbalance in the subunits composing the BEAT antibody as the potential root cause. This misbalance was then addressed by a signal peptide optimization, and the overall expression level was increased by the combination of two vector design elements on a single gene vector. Further improvements were made in the selection of cell populations and an upstream (USP) platform process was applied in combination with a cell culture temperature shift. This allowed titer levels of up to 6 g/L to be reached with these difficult-to-express proteins. Furthermore, a high-density seeding process was developed that allowed titers of around 11 g/L for the BEAT antibody, increasing the initial titer by a factor of 10. The approach was successfully applied to a tri-specific antibody with titer levels reaching 10 g/L. In summary, a platform process for difficult-to-express proteins was developed using molecular biology tools, cell line development, upstream process optimization and process intensification.


Sujet(s)
Anticorps monoclonaux , Animaux , Anticorps monoclonaux/génétique , Anticorps monoclonaux/biosynthèse , Cellules CHO , Cricetulus , Humains
13.
Viruses ; 16(4)2024 04 01.
Article de Anglais | MEDLINE | ID: mdl-38675896

RÉSUMÉ

Neutralizing antibodies (NtAbs) against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) are indicators of vaccine efficacy that enable immunity surveillance. However, the rapid mutation of SARS-CoV-2 variants prevents the timely establishment of standards required for effective XBB vaccine evaluation. Therefore, we prepared four candidate standards (No. 11, No. 44, No. 22, and No. 33) using plasma, purified immunoglobulin, and a broad-spectrum neutralizing monoclonal antibody. Collaborative calibration was conducted across nine Chinese laboratories using neutralization methods against 11 strains containing the XBB and BA.2.86 sublineages. This study demonstrated the reduced neutralization potency of the first International Standard antibodies to SARS-CoV-2 variants of concern against XBB variants. No. 44 displayed broad-spectrum neutralizing activity against XBB sublineages, effectively reduced interlaboratory variability for nearly all XBB variants, and effectively minimized the geometric mean titer (GMT) difference between the live and pseudotyped virus. No. 22 showed a broader spectrum and higher neutralizing activity against all strains but failed to reduce interlaboratory variability. Thus, No. 44 was approved as a National Standard for NtAbs against XBB variants, providing a unified NtAb measurement standard for XBB variants for the first time. Moreover, No. 22 was approved as a national reference reagent for NtAbs against SARS-CoV-2, offering a broad-spectrum activity reference for current and potentially emerging variants.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Tests de neutralisation , SARS-CoV-2 , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Humains , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , COVID-19/immunologie , COVID-19/virologie , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/génétique , Vaccins contre la COVID-19/immunologie , Chine , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique
14.
Int J Biol Macromol ; 266(Pt 2): 131379, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38580014

RÉSUMÉ

Monoclonal antibodies (mAbs) are laboratory-based engineered protein molecules with a monovalent affinity or multivalent avidity towards a specific target or antigen, which can mimic natural antibodies that are produced in the human immune systems to fight against detrimental pathogens. The recombinant mAb is one of the most effective classes of biopharmaceuticals produced in vitro by cloning and expressing synthetic antibody genes in a suitable host. Yeast is one of the potential hosts among others for the successful production of recombinant mAbs. However, there are very few yeast-derived mAbs that got the approval of the regulatory agencies for direct use for treatment purposes. Certain challenges encountered by yeasts for recombinant antibody productions need to be overcome and a few considerations related to antibody structure, host engineering, and culturing strategies should be followed for the improved production of mAbs in yeasts. In this review, the drawbacks related to the metabolic burden of the host, culturing conditions including induction mechanism and secretion efficiency, solubility and stability, downstream processing, and the pharmacokinetic behavior of the antibody are discussed, which will help in developing the yeast hosts for the efficient production of recombinant mAbs.


Sujet(s)
Anticorps monoclonaux , Protéines recombinantes , Levures , Animaux , Humains , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/génétique , Protéines recombinantes/immunologie , Protéines recombinantes/génétique , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Levures/métabolisme , Levures/génétique
15.
MAbs ; 16(1): 2342243, 2024.
Article de Anglais | MEDLINE | ID: mdl-38650451

RÉSUMÉ

The controlled expression of two or more proteins at a defined and stable ratio remains a substantial challenge, particularly in the bi- and multispecific antibody field. Achieving an optimal ratio of protein subunits can facilitate the assembly of multimeric proteins with high efficiency and minimize the production of by-products. In this study, we propose a solution based on alternative splicing, enabling the expression of a tunable and predefined ratio of two distinct polypeptide chains from the same pre-mRNA under the control of a single promoter. The pre-mRNA used in this study contains two open reading frames situated on separate exons. The first exon is flanked by two copies of the chicken troponin intron 4 (cTNT-I4) and is susceptible to excision from the pre-mRNA by means of alternative splicing. This specific design enables the modulation of the splice ratio by adjusting the strength of the splice acceptor. To illustrate this approach, we developed constructs expressing varying ratios of GFP and dsRED and extended their application to multimeric proteins such as monoclonal antibodies, achieving industrially relevant expression levels (>1 g/L) in a 14-day fed-batch process. The stability of the splice ratio was confirmed by droplet digital PCR in a stable pool cultivated over a 28-day period, while product quality was assessed via intact mass analysis, demonstrating absence of product-related impurities resulting from undesired splice events. Furthermore, we showcased the versatility of the construct by expressing two subunits of a bispecific antibody of the BEAT® type, which contains three distinct subunits in total.


Sujet(s)
Épissage alternatif , Animaux , Sous-unités de protéines/génétique , Humains , Poulets , Anticorps bispécifiques/génétique , Anticorps bispécifiques/biosynthèse , Cellules CHO , Exons/génétique , Cricetulus , Protéines à fluorescence verte/génétique , Anticorps monoclonaux/génétique , Anticorps monoclonaux/biosynthèse , Précurseurs des ARN/génétique
16.
Biotechnol J ; 19(4): e2300505, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38651269

RÉSUMÉ

Chinese hamster ovary (CHO) cells are the commonly used mammalian host system to manufacture recombinant proteins including monoclonal antibodies. However unfavorable non-human glycoprofile displayed on CHO-produced monoclonal antibodies have negative impacts on product quality, pharmacokinetics, and therapeutic efficiency. Glycoengineering such as genetic elimination of genes involved in glycosylation pathway in CHO cells is a viable solution but constrained due to longer timeline and laborious workflow. Here, in this proof-of-concept (PoC) study, we present a novel approach coined CellEDIT to engineer CHO cells by intranuclear delivery of the CRISPR components to single cells using the FluidFM technology. Co-injection of CRISPR system targeting BAX, DHFR, and FUT8 directly into the nucleus of single cells, enabled us to generate triple knockout CHO-K1 cell lines within a short time frame. The proposed technique assures the origin of monoclonality without the requirement of limiting dilution, cell sorting or positive selection. Furthermore, the approach is compatible to develop both single and multiple knockout clones (FUT8, BAX, and DHFR) in CHO cells. Further analyses on single and multiple knockout clones confirmed the targeted genetic disruption and altered protein expression. The knockout CHO-K1 clones showed the persistence of gene editing during the subsequent passages, compatible with serum free chemically defined media and showed equivalent transgene expression like parental clone.


Sujet(s)
Systèmes CRISPR-Cas , Cricetulus , Édition de gène , Cellules CHO , Animaux , Systèmes CRISPR-Cas/génétique , Édition de gène/méthodes , Anticorps monoclonaux/génétique , Protéines recombinantes/génétique , Techniques de knock-out de gènes/méthodes , Protéine Bax/génétique , Protéine Bax/métabolisme , Dihydrofolate reductase/génétique , Dihydrofolate reductase/métabolisme , Fucosyltransferases/génétique , Fucosyltransferases/métabolisme , Cricetinae , Génie génétique/méthodes
17.
Methods Mol Biol ; 2768: 211-239, 2024.
Article de Anglais | MEDLINE | ID: mdl-38502396

RÉSUMÉ

The affinity distribution of the antigen-specific memory B cell (Bmem) repertoire in the body is a critical variable that defines an individual's ability to rapidly generate high-affinity protective antibody specificities. Detailed measurement of antibody affinity so far has largely been confined to studies of monoclonal antibodies (mAbs) and are laborious since each individual mAb needs to be evaluated in isolation. Here, we introduce two variants of the B cell ImmunoSpot® assay that are suitable for simultaneously assessing the affinity distribution of hundreds of individual B cells within a test sample at single-cell resolution using relatively little labor and with high-throughput capacity. First, we experimentally validated that both ImmunoSpot® assay variants are suitable for establishing functional affinity hierarchies using B cell hybridoma lines as model antibody-secreting cells (ASC), each producing mAb with known affinity for a defined antigen. We then leveraged both ImmunoSpot® variants for characterizing the affinity distribution of SARS-CoV-2 Spike-specific ASC in PBMC following COVID-19 mRNA vaccination. Such ImmunoSpot® assays promise to offer tremendous value for future B cell immune monitoring efforts, owing to their ease of implementation, applicability to essentially any antigenic system, economy of PBMC utilization, high-throughput capacity, and suitability for regulated testing.


Sujet(s)
Lymphocytes B , Agranulocytes , Agranulocytes/métabolisme , Test ELISpot , Antigènes , Cellules productrices d'anticorps , Anticorps monoclonaux/génétique , Anticorps monoclonaux/métabolisme
18.
Adv Sci (Weinh) ; 11(21): e2308763, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38552157

RÉSUMÉ

The CRISPR/Cas system has been introduced as an innovative tool for therapy, however achieving specific delivery to the target has been a major challenge. Here, an antibody-CRISPR/Cas conjugate platform that enables specific delivery and target gene editing in HER2-positive cancer is introduced. The CRISPR/Cas system by replacing specific residues of Cas9 with an unnatural amino acid is engineered, that can be complexed with a nanocarrier and bioorthogonally functionalized with a monoclonal antibody targeting HER2. The resultant antibody-conjugated CRISPR/Cas nanocomplexes can be specifically delivered and induce gene editing in HER2-positive cancer cells in vitro. It is demonstrated that the in vivo delivery of the antibody-CRISPR/Cas nanocomplexes can effectively disrupt the plk1 gene in HER2-positive ovarian cancer, resulting in substantial suppression of tumor growth. The current study presents a useful therapeutic platform for antibody-mediated delivery of CRISPR/Cas for the treatment of various cancers and genetic diseases.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Édition de gène/méthodes , Systèmes CRISPR-Cas/génétique , Humains , Souris , Animaux , Femelle , Lignée cellulaire tumorale , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/thérapie , Modèles animaux de maladie humaine , Techniques de transfert de gènes , Anticorps monoclonaux/génétique , Tumeurs/thérapie , Tumeurs/génétique , Récepteur ErbB-2/génétique
19.
Antiviral Res ; 225: 105867, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38521465

RÉSUMÉ

Monoclonal antibody-based therapeutics have achieved remarkable success in treating a wide range of human diseases. However, conventional systemic delivery methods have limitations in insufficient target tissue permeability, high costs, repeated administrations, etc. Novel technologies have been developed to address these limitations and further enhance antibody therapy. Local antibody delivery via respiratory tract, gastrointestinal tract, eye and blood-brain barrier have shown promising results in increasing local concentrations and overcoming barriers. Nucleic acid-encoded antibodies expressed from plasmid DNA, viral vectors or mRNA delivery platforms also offer advantages over recombinant proteins such as sustained expression, rapid onset, and lower costs. This review summarizes recent advances in antibody delivery methods and highlights innovative technologies that have potential to expand therapeutic applications of antibodies.


Sujet(s)
Vecteurs génétiques , Facteurs immunologiques , Humains , Plasmides , Anticorps monoclonaux/génétique , ARN messager , Systèmes de délivrance de médicaments/méthodes
20.
Biotechnol J ; 19(2): e2300407, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38403434

RÉSUMÉ

Monitoring the stability of recombinant Chinese Hamster Ovary (CHO) cell lines is essential to ensure the selection of production cell lines suitable for biomanufacturing. It has been frequently observed that recombinant CHO cell lines develop phenotypic changes upon aging, such as accelerated cell growth in late generation cultures. However, the mechanism responsible for age-correlated changes is poorly understood. In this study, we investigated the molecular mechanisms underlying the age-correlated cell growth improvement in Pfizer's platform fed-batch production process, by examining multiple cell lines derived from different CHO expression systems, expressing a variety of monoclonal antibodies (mAbs). Comprehensive whole-genome resequencing analysis revealed duplication of a continuous 50.2 Mbp segment in chromosome 2 (Chr2) specific to clones that showed age-correlated growth change as compared to clones that did not exhibit age-correlated growth change. Moreover, such age- and growth-related Chr2 duplication was independent of the presence or type of recombinant monoclonal antibody expression. When we compared transcriptome profiles from low-growth and high-growth cell lines, we found that >95% of the genes overexpressed in high-growth cell lines were in the duplicated Chr2 segment. To the best of our knowledge, this is the first report of large genomic duplication, specific to Chr2, being associated with age-correlated growth change. Investigation of the cause-and-effect relationship between the genes identified in the duplicated regions and age-correlated growth change is underway. We are confident that this effort will lead to improved cell line screening and targeted rational cell line engineering efforts to develop cell lines with improved stability performance.


Sujet(s)
Anticorps monoclonaux , Chromosomes humains de la paire 2 , Cricetinae , Humains , Animaux , Cricetulus , Cellules CHO , Chromosomes humains de la paire 2/métabolisme , Protéines recombinantes/métabolisme , Anticorps monoclonaux/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE