Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.467
Filtrer
1.
Cancer Immunol Immunother ; 73(10): 195, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39105809

RÉSUMÉ

BACKGROUND: The efficacy of antibody-targeted therapy of solid cancers is limited by the lack of consistent tumour-associated antigen expression. However, tumour-associated antigens shared with non-malignant cells may still be targeted using conditionally activated-antibodies, or by chimeric antigen receptor (CAR) T cells or CAR NK cells activated either by the tumour microenvironment or following 'unlocking' via multiple antigen-recognition. In this study, we have focused on tissue factor (TF; CD142), a type I membrane protein present on a range of solid tumours as a basis for future development of conditionally-activated BiTE or CAR T cells. TF is frequently upregulated on multiple solid tumours providing a selective advantage for growth, immune evasion and metastasis, as well as contributing to the pathology of thrombosis via the extrinsic coagulation pathway. METHODS: Two well-characterised anti-TF monoclonal antibodies (mAb) were cloned into expression or transposon vectors to produce single chain (scFv) BiTE for assessment as CAR and CD28-CD3-based CAR or CD3-based BiTE. The affinities of both scFv formats for TF were determined by surface plasmon resonance. Jurkat cell line-based assays were used to confirm the activity of the BiTE or CAR constructs. RESULTS: The anti-TF mAb hATR-5 and TF8-5G9 mAb were shown to maintain their nanomolar affinities following conversion into a single chain (scFv) format and could be utilised as CD28-CD3-based CAR or CD3-based BiTE format. CONCLUSION: Because of the broad expression of TF on a range of solid cancers, anti-TF antibody formats provide a useful addition for the development of conditionally activated biologics for antibody and cellular-based therapy.


Sujet(s)
Récepteurs chimériques pour l'antigène , Lymphocytes T , Thromboplastine , Humains , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Thromboplastine/immunologie , Thromboplastine/métabolisme , Lymphocytes T/immunologie , Immunothérapie adoptive/méthodes , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Tumeurs/immunologie , Tumeurs/thérapie , Cellules Jurkat
2.
Biotechnol J ; 19(7): e2300745, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39014926

RÉSUMÉ

We developed a method to produce a soluble form of a single-chain fragment variable (scFv) targeting human epithelial growth factor receptor 2 (HER2) in Escherichia coli. By optimizing the orientations of the variable heavy (VH) and variable light (VL) domains and the His-tag, we identified the HL-His type antibody with the highest HER2-binding activity. Purification of HL-His yielded 40.7 mg from a 1 L culture, achieving >99% purity. The limit of detection was determined to be 2.9 ng, demonstrating high production yield, purity, and sensitivity. Moreover, we successfully labeled HER2+ cell lines with fluorescent dye-conjugated scFv, resulting in a significantly higher observed signal-to-background ratio, compared to that of HER2- cell lines. This highlights the potential of these fluorescent scFvs as valuable probes for HER2+ breast cancer diagnostics. Notably, the process for the complete scFv production was streamlined and required only 4-5 days. Additionally, the product maintained its activity after freeze storage, allowing for large-scale production and a wide range of practical applications.


Sujet(s)
Escherichia coli , Récepteur ErbB-2 , Protéines recombinantes , Anticorps à chaîne unique , Récepteur ErbB-2/immunologie , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/isolement et purification , Humains , Escherichia coli/génétique , Escherichia coli/métabolisme , Lignée cellulaire tumorale , Tumeurs du sein/immunologie
3.
Sci Rep ; 14(1): 15864, 2024 07 09.
Article de Anglais | MEDLINE | ID: mdl-38982108

RÉSUMÉ

In 2019, the novel SARS-CoV-2 coronavirus emerged in China, causing the pneumonia named COVID-19. At the beginning, all research efforts were focused on the spike (S) glycoprotein. However, it became evident that the nucleocapsid (N) protein is pivotal in viral replication, genome packaging and evasion of the immune system, is highly immunogenic, which makes it another compelling target for antibody development alongside the spike protein. This study focused on the construction of single chain fragments variable (scFvs) libraries from SARS-CoV-2-infected patients to establish a valuable, immortalized and extensive antibodies source. We used the Intracellular Antibody Capture Technology to select a panel of scFvs against the SARS-CoV-2 N protein. The whole panel of scFv was expressed and characterized both as intrabodies and recombinant proteins. ScFvs were then divided into 2 subgroups: those that exhibited high binding activity to N protein when expressed in yeast or in mammalian cells as intrabodies, and those purified as recombinant proteins, displaying affinity for recombinant N protein in the nanomolar range. This panel of scFvs against the N protein represents a novel platform for research and potential diagnostic applications.


Sujet(s)
Anticorps antiviraux , COVID-19 , Protéines de la nucléocapside des coronavirus , SARS-CoV-2 , Anticorps à chaîne unique , Humains , SARS-CoV-2/immunologie , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , COVID-19/immunologie , COVID-19/virologie , Anticorps antiviraux/immunologie , Protéines de la nucléocapside des coronavirus/immunologie , Phosphoprotéines/immunologie , Protéines recombinantes/immunologie , Protéines recombinantes/génétique , Banque de peptides
4.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2258-2269, 2024 Jul 25.
Article de Chinois | MEDLINE | ID: mdl-39044589

RÉSUMÉ

This study aims to prepare bacterial outer membrane vesicles (OMVs) with anti-glypican-3 (GPC3) single-chain antibody and analyze their targeting effects on Hep G2 hepatocellular carcinoma (HCC) cells and tissue. The recombinant plasmid pET28a-Hbp-hGC 33-scFv was constructed by ligating Hbp-hGC 33-scFv to pET28a. Western blotting was employed to determine the prokaryotic expression of the fusion protein Hbp-hGC 33-scFv, on the basis of which the optimal induction conditions were determined. Hbp-hGC 33-OMVs secreted from the recombinant expressing strains were collected by ultrafiltration concentration and then characterized. The localization of Hbp-hGC 33-scFv in bacteria and Hbp-hGC 33-OMVs was analyzed by immune electron microscopy. The binding of Hbp-hGC 33-scFv to Hep G2 cells was observed by immunofluorescence. The Hep G2 tumor-bearing mouse model was established, and the targeted retention of Hbp-hGC 33-OMVs in the tumor site of mice was observed by a fluorescence imaging system in vivo. The results showed that the actual molecular weight of the fusion protein was 175.3 kDa, and the optimal induction conditions were as follows: OD600=0.5, IPTG added at a final concentration of 0.5 mmol/L, and overnight induction at 16 ℃. The prepared Hbp-hGC 33-OMVs were irregular spherical structures with an average particle size of (112.3±4.6) nm, expressing OmpC, OmpA, and the fusion protein Hbp-hGC 33-scFv. The Hbp-hGC 33-OMVs prepared in this study demonstrated stronger ability of binding to Hep G2 cells than the wild-type OMVs (P=0.008). All the data indicated that Hbp-hGC 33-OMVs with anti-GPC3 single-chain antibody were successfully prepared and could be used for research on the targeted therapy of hepatocellular carcinoma.


Sujet(s)
Membrane bactérienne externe , Carcinome hépatocellulaire , Glypicanes , Tumeurs du foie , Anticorps à chaîne unique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/composition chimique , Animaux , Souris , Humains , Tumeurs du foie/immunologie , Tumeurs du foie/métabolisme , Membrane bactérienne externe/métabolisme , Membrane bactérienne externe/immunologie , Cellules HepG2 , Glypicanes/immunologie , Glypicanes/métabolisme , Glypicanes/génétique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/biosynthèse , Escherichia coli/génétique , Escherichia coli/métabolisme , Systèmes de délivrance de médicaments , Souris nude
5.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39000338

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells represent a revolutionary immunotherapy that allows specific tumor recognition by a unique single-chain fragment variable (scFv) derived from monoclonal antibodies (mAbs). scFv selection is consequently a fundamental step for CAR construction, to ensure accurate and effective CAR signaling toward tumor antigen binding. However, conventional in vitro and in vivo biological approaches to compare different scFv-derived CARs are expensive and labor-intensive. With the aim to predict the finest scFv binding before CAR-T cell engineering, we performed artificial intelligence (AI)-guided molecular docking and steered molecular dynamics analysis of different anti-CD30 mAb clones. Virtual computational scFv screening showed comparable results to surface plasmon resonance (SPR) and functional CAR-T cell in vitro and in vivo assays, respectively, in terms of binding capacity and anti-tumor efficacy. The proposed fast and low-cost in silico analysis has the potential to advance the development of novel CAR constructs, with a substantial impact on reducing time, costs, and the need for laboratory animal use.


Sujet(s)
Intelligence artificielle , Antigènes CD30 , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Récepteurs chimériques pour l'antigène , Anticorps à chaîne unique , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/génétique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/génétique , Humains , Antigènes CD30/immunologie , Antigènes CD30/métabolisme , Animaux , Souris , Liaison aux protéines , Résonance plasmonique de surface
6.
Commun Biol ; 7(1): 922, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39085379

RÉSUMÉ

Designing effective monoclonal antibody (mAb) therapeutics faces a multi-parameter optimization challenge known as "developability", which reflects an antibody's ability to progress through development stages based on its physicochemical properties. While natural antibodies may provide valuable guidance for mAb selection, we lack a comprehensive understanding of natural developability parameter (DP) plasticity (redundancy, predictability, sensitivity) and how the DP landscapes of human-engineered and natural antibodies relate to one another. These gaps hinder fundamental developability profile cartography. To chart natural and engineered DP landscapes, we computed 40 sequence- and 46 structure-based DPs of over two million native and human-engineered single-chain antibody sequences. We find lower redundancy among structure-based compared to sequence-based DPs. Sequence DP sensitivity to single amino acid substitutions varied by antibody region and DP, and structure DP values varied across the conformational ensemble of antibody structures. We show that sequence DPs are more predictable than structure-based ones across different machine-learning tasks and embeddings, indicating a constrained sequence-based design space. Human-engineered antibodies localize within the developability and sequence landscapes of natural antibodies, suggesting that human-engineered antibodies explore mere subspaces of the natural one. Our work quantifies the plasticity of antibody developability, providing a fundamental resource for multi-parameter therapeutic mAb design.


Sujet(s)
Anticorps monoclonaux , Ingénierie des protéines , Humains , Ingénierie des protéines/méthodes , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux/immunologie , Conformation des protéines , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie
7.
Glycobiology ; 34(8)2024 06 22.
Article de Anglais | MEDLINE | ID: mdl-38963938

RÉSUMÉ

Heparan sulfate (HS) is a linear polysaccharide with high structural and functional diversity. Detection and localization of HS in tissues can be performed using single chain variable fragment (scFv) antibodies. Although several anti-HS antibodies recognizing different sulfation motifs have been identified, little is known about their interaction with HS. In this study the interaction between the scFv antibody HS4C3 and heparin was investigated. Heparin-binding lysine and arginine residues were identified using a protect and label methodology. Site-directed mutagenesis was applied to further identify critical heparin-binding lysine/arginine residues using immunohistochemical and biochemical assays. In addition, computational docking of a heparin tetrasaccharide towards a 3-D homology model of HS4C3 was applied to identify potential heparin-binding sites. Of the 12 lysine and 15 arginine residues within the HS4C3 antibody, 6 and 9, respectively, were identified as heparin-binding. Most of these residues are located within one of the complementarity determining regions (CDR) or in their proximity. All basic amino acid residues in the CDR3 region of the heavy chain were involved in binding. Computational docking showed a heparin tetrasaccharide close to these regions. Mutagenesis of heparin-binding residues reduced or altered reactivity towards HS and heparin. Identification of heparin-binding arginine and lysine residues in HS4C3 allows for better understanding of the interaction with HS and creates a framework to rationally design antibodies targeting specific HS motifs.


Sujet(s)
Héparine , Héparitine sulfate , Héparitine sulfate/composition chimique , Héparitine sulfate/immunologie , Héparitine sulfate/métabolisme , Héparine/composition chimique , Héparine/métabolisme , Simulation de docking moléculaire , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Humains , Animaux , Mutagenèse dirigée , Sites de fixation , Séquence d'acides aminés
8.
Sci Rep ; 14(1): 14419, 2024 06 22.
Article de Anglais | MEDLINE | ID: mdl-38909102

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has prolonged the duration of the pandemic because of the continuous emergence of new variant strains. The emergence of these mutant strains makes it difficult to detect the virus with the existing antibodies; thus, the development of novel antibodies that can target both the variants as well as the original strain is necessary. In this study, we generated a high-affinity monoclonal antibody (5G2) against the highly conserved region of the SARS-CoV-2 spike protein to detect the protein variants. Moreover, we generated its single-chain variable antibody fragment (sc5G2). The sc5G2 expressed in mammalian and bacterial cells detected the spike protein of the original SARS-CoV-2 and variant strains. The resulting sc5G2 will be a useful tool to detect the original SARS-CoV-2 and variant strains.


Sujet(s)
Anticorps antiviraux , SARS-CoV-2 , Anticorps à chaîne unique , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , SARS-CoV-2/immunologie , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Humains , Anticorps antiviraux/immunologie , COVID-19/immunologie , COVID-19/virologie , Animaux , Anticorps monoclonaux/immunologie , Séquence conservée
9.
Sci Rep ; 14(1): 14558, 2024 06 24.
Article de Anglais | MEDLINE | ID: mdl-38914666

RÉSUMÉ

Plants offer a cost-effective and scalable pharmaceutical platform devoid of host-derived contamination risks. However, their medical application is complicated by the potential for acute allergic reactions to external proteins. Developing plant-based protein therapeutics for localized diseases with non-invasive treatment modalities may capitalize on the benefits of plant proteins while avoiding their inherent risks. Dupilumab, which is effective against a variety of allergic and autoimmune diseases but has systemic responses and injection-related side effects, may be more beneficial if delivered locally using a small biological form. In this study, we engineered a single-chain variable fragment (scFv) of dupilumab, termed Dup-scFv produced by Nicotiana benthamiana, and evaluated its tissue permeability and anti-inflammatory efficacy in air-liquid interface cultured human nasal epithelial cells (HNECs). Despite showing 3.67- and 17-fold lower binding affinity for IL-4Ra in surface plasmon resonance assays and cell binding assays, respectively, Dup-scFv retained most of the affinity of dupilumab, which was originally high, with a dissociation constant (KD) of 4.76 pM. In HNECs cultured at the air-liquid interface, Dup-scFv administered on the air side inhibited the inflammatory marker CCL26 in hard-to-reach basal cells more effectively than dupilumab. In addition, Dup-scFv had an overall permeability of 0.8% across cell layers compared to undetectable levels of dupilumab. These findings suggest that plant-produced Dup-scFv can be delivered non-invasively to cultured HNESc to alleviate inflammatory signaling, providing a practical approach to utilize plant-based proteins for topical therapeutic applications.


Sujet(s)
Anticorps monoclonaux humanisés , Cellules épithéliales , Nicotiana , Anticorps à chaîne unique , Humains , Nicotiana/métabolisme , Anticorps monoclonaux humanisés/pharmacologie , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Anticorps à chaîne unique/pharmacologie , Anticorps à chaîne unique/génétique , Chimiokines CC/métabolisme , Sous-unité alpha du récepteur à l'interleukine-4/métabolisme , Cellules cultivées , Muqueuse nasale/métabolisme , Muqueuse nasale/cytologie , Muqueuse nasale/immunologie
10.
Arch Biochem Biophys ; 758: 110068, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38909835

RÉSUMÉ

Affinity maturation increases antigen-binding affinity and specificity of antibodies by somatic hypermutation. Various monoclonal antibodies against (4-hydroxy-3-nitrophenyl)acetyl (NP) were obtained during affinity maturation. Among them, highly matured anti-NP antibodies, such as E11 and E3, possess Cys96H and Cys100H in the complementarity-determining region 3 of the heavy chain, which would form a disulfide bond. In this study, we evaluated the effects of disulfide bonds on antigen binding by generating single-chain Fv (scFv) antibodies of E11 and its mutants, E11_C96KH/C100EH and E11_C96KH/C100QH, and determined their antigen-binding thermodynamics and kinetics. The binding affinities of the Cys mutants were lower than that of E11 scFv, indicating that the disulfide bond contributed to antigen binding, especially for stable complex formation. This was also supported by the decreased affinity of E11 scFv in the presence of a reducing agent. The crystal structures of NP-free and NP-bound E11 scFvs were determined at high resolution, showing the existence of a disulfide bond between Cys96H and Cys100H, and the antigen recognition mechanism, which could be compared with those of other anti-NP antibodies, such as germline-type N1G9 and matured-type C6, as reported previously. These structures could explain the molecular basis of changes in antigen-binding affinity and thermal stability in the absence or presence of antigens. Small-angle X-ray scattering further showed a local conformational change in E11 scFv upon antigen binding in solution.


Sujet(s)
Affinité des anticorps , Régions déterminant la complémentarité , Disulfures , Anticorps à chaîne unique , Disulfures/composition chimique , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Régions déterminant la complémentarité/composition chimique , Humains , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux/immunologie , Animaux , Thermodynamique , Cinétique , Cristallographie aux rayons X , Modèles moléculaires
11.
ACS Synth Biol ; 13(6): 1679-1693, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38819389

RÉSUMÉ

Immune-checkpoint blockade (ICB) reinvigorates T cells from exhaustion and potentiates T-cell responses to tumors. However, most patients do not respond to ICB therapy, and only a limited response can be achieved in a "cold" tumor with few infiltrated lymphocytes. Synthetic biology can be used to engineer bacteria as controllable bioreactors to synthesize biotherapeutics in situ. We engineered attenuated Salmonella VNP20009 with synthetic gene circuits to produce PD-1 and Tim-3 scFv to block immunosuppressive receptors on exhausted T cells to reinvigorate their antitumor response. Secreted PD-1 and Tim-3 scFv bound PD-1+ Tim-3+ T cells through their targeting receptors in vitro and potentiated the T-cell secretion of IFN-γ. Engineered bacteria colonized the hypoxic core of the tumor and synthesized PD-1 and Tim-3 scFv in situ, reviving CD4+ T cells and CD8+ T cells to execute an antitumor response. The bacteria also triggered a strong innate immune response, which stimulated the expansion of IFN-γ+ CD4+ T cells within the tumors to induce direct and indirect antitumor immunity.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Salmonella , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Animaux , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/immunologie , Souris , Salmonella/immunologie , Salmonella/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Récepteur cellulaire-2 du virus de l'hépatite A/génétique , Lignée cellulaire tumorale , Lymphocytes T CD8+/immunologie , Humains , Interféron gamma/métabolisme , Interféron gamma/immunologie , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/pharmacologie , Souris de lignée C57BL , Biologie synthétique/méthodes , Lymphocytes T CD4+/immunologie , Immunothérapie/méthodes
12.
Protein Sci ; 33(6): e5017, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38747382

RÉSUMÉ

Biparatopic antibodies (bpAbs) are engineered antibodies that bind to multiple different epitopes within the same antigens. bpAbs comprise diverse formats, including fragment-based formats, and choosing the appropriate molecular format for a desired function against a target molecule is a challenging task. Moreover, optimizing the design of constructs requires selecting appropriate antibody modalities and adjusting linker length for individual bpAbs. Therefore, it is crucial to understand the characteristics of bpAbs at the molecular level. In this study, we first obtained single-chain variable fragments and camelid heavy-chain variable domains targeting distinct epitopes of the metal binding protein MtsA and then developed a novel format single-chain bpAb connecting these fragment antibodies with various linkers. The physicochemical properties, binding activities, complex formation states with antigen, and functions of the bpAb were analyzed using multiple approaches. Notably, we found that the assembly state of the complexes was controlled by a linker and that longer linkers tended to form more compact complexes. These observations provide detailed molecular information that should be considered in the design of bpAbs.


Sujet(s)
Anticorps à chaîne unique , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie , Animaux , Humains , Ingénierie des protéines/méthodes , Épitopes/composition chimique , Épitopes/immunologie , Chaines lourdes des immunoglobulines/composition chimique , Chaines lourdes des immunoglobulines/génétique , Chaines lourdes des immunoglobulines/immunologie
13.
J Virol ; 98(6): e0003824, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38767356

RÉSUMÉ

Recent progress on chimeric antigen receptor (CAR)-NK cells has shown promising results in treating CD19-positive lymphoid tumors with minimal toxicities [including graft versus host disease (GvHD) and cytokine release syndrome (CRS) in clinical trials. Nevertheless, the use of CAR-NK cells in combating viral infections has not yet been fully explored. Previous studies have shown that CAR-NK cells expressing S309 single-chain fragment variable (scFv), hereinafter S309-CAR-NK cells, can bind to SARS-CoV-2 wildtype pseudotyped virus (PV) and effectively kill cells expressing wild-type spike protein in vitro. In this study, we further demonstrate that the S309-CAR-NK cells can bind to different SARS-CoV-2 variants, including the B.1.617.2 (Delta), B.1.621 (Mu), and B.1.1.529 (Omicron) variants in vitro. We also show that S309-CAR-NK cells reduce virus loads in the NOD/SCID gamma (NSG) mice expressing the human angiotensin-converting enzyme 2 (hACE2) receptor challenged with SARS-CoV-2 wild-type (strain USA/WA1/2020). Our study demonstrates the potential use of S309-CAR-NK cells for inhibiting infection by SARS-CoV-2 and for the potential treatment of COVID-19 patients unresponsive to otherwise currently available therapeutics. IMPORTANCE: Chimeric antigen receptor (CAR)-NK cells can be "off-the-shelf" products that treat various diseases, including cancer, infections, and autoimmune diseases. In this study, we engineered natural killer (NK) cells to express S309 single-chain fragment variable (scFv), to target the Spike protein of SARS-CoV-2, hereinafter S309-CAR-NK cells. Our study shows that S309-CAR-NK cells are effective against different SARS-CoV-2 variants, including the B.1.617.2 (Delta), B.1.621 (Mu), and B.1.1.529 (Omicron) variants. The S309-CAR-NK cells can (i) directly bind to SARS-CoV-2 pseudotyped virus (PV), (ii) competitively bind to SARS-CoV-2 PV with 293T cells expressing the human angiotensin-converting enzyme 2 (hACE2) receptor (293T-hACE2 cells), (iii) specifically target and lyse A549 cells expressing the spike protein, and (iv) significantly reduce the viral loads of SARS-CoV-2 wild-type (strain USA/WA1/2020) in the lungs of NOD/SCID gamma (NSG) mice expressing hACE2 (hACE2-NSG mice). Altogether, the current study demonstrates the potential use of S309-CAR-NK immunotherapy as an alternative treatment for COVID-19 patients.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Cellules tueuses naturelles , Récepteurs chimériques pour l'antigène , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Charge virale , Animaux , SARS-CoV-2/immunologie , Cellules tueuses naturelles/immunologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/immunologie , Souris , Humains , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , COVID-19/immunologie , COVID-19/virologie , COVID-19/thérapie , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Souris SCID , Souris de lignée NOD
14.
J Biol Chem ; 300(6): 107397, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38763332

RÉSUMÉ

Constant domains in antibody molecules at the level of the Fab (CH1 and CL) have long been considered to be simple scaffolding elements that physically separate the paratope-defining variable (V) region from the effector function-mediating constant (C) regions. However, due to recent findings that C domains of different isotypes can modulate the fine specificity encoded in the V region, elucidating the role of C domains in shaping the paratope and influencing specificity is a critical area of interest. To dissect the relative contributions of each C domain to this phenomenon, we generated antibody fragments with different C regions omitted, using a set of antibodies targeting capsular polysaccharides from the fungal pathogen, Cryptococcus neoformans. Antigen specificity mapping and functional activity measurements revealed that V region-only antibody fragments exhibited poly-specificity to antigenic variants and extended to recognition of self-antigens, while measurable hydrolytic activity of the capsule was greatly attenuated. To better understand the mechanistic origins of the remarkable loss of specificity that accompanies the removal of C domains from identical paratopes, we performed molecular dynamics simulations which revealed increased paratope plasticity in the scFv relative to the corresponding Fab. Together, our results provide insight into how the remarkable specificity of immunoglobulins is governed and maintained at the level of the Fab through the enforcement of structural restrictions on the paratope by CH1 domains.


Sujet(s)
Cryptococcus neoformans , Épitopes , Cryptococcus neoformans/immunologie , Cryptococcus neoformans/composition chimique , Épitopes/composition chimique , Épitopes/immunologie , Régions constantes des immunoglobulines/composition chimique , Régions constantes des immunoglobulines/génétique , Simulation de dynamique moléculaire , Fragments Fab d'immunoglobuline/composition chimique , Fragments Fab d'immunoglobuline/immunologie , Fragments Fab d'immunoglobuline/métabolisme , Humains , Spécificité des anticorps , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Animaux , Anticorps antifongiques/immunologie , Anticorps antifongiques/composition chimique
15.
J Immunoassay Immunochem ; 45(4): 307-324, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38776466

RÉSUMÉ

Single Chain Variable Fragment (scFv), a small fragment of antibody can be used to substitute the monoclonal antibody for diagnostic purposes. Production of scFv in Escherichia coli host has been a challenge due to the potential miss-folding and formation of inclusion bodies. This study aimed to express anti-CHIKV E2 scFv which previously designed specifically for Asian strains by co-expression of three chaperones that play a role in increasing protein solubility; GroEL, GroES, and Trigger Factor. The scFv and chaperones were expressed in Origami B E. coli host under the control of the T7 promoter, and purified using a Ni-NTA column. Functional assay of anti-CHIKV-E2 scFv was examined by electrochemical immunosensor using gold modified Screen Printed Carbon Electrode (SPCE), and characterized by differential pulses voltammetry (DPV) using K3[Fe(CN)6] redox system and scanning microscope electron (SEM). The experimental condition was optimized using the Box-Behnken design. The results showed that co-expression of chaperone increased the soluble scFv yield from 54.405 µg/mL to 220.097 µg/mL (~5×). Furthermore, scFv can be used to detect CHIKV-E2 in immunosensor electrochemistry with a detection limit of 0.74048 ng/mL and a quantification limit of 2,24388 ng/mL. Thus, the scFv-anti-CHIKV-E2 can be applied as a bioreceptor in another immunoassay method.


Sujet(s)
Techniques de biocapteur , Techniques électrochimiques , Escherichia coli , Chaperons moléculaires , Anticorps à chaîne unique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/génétique , Escherichia coli/métabolisme , Escherichia coli/génétique , Chaperons moléculaires/immunologie , Dosage immunologique/méthodes
16.
Int Immunopharmacol ; 136: 112273, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38810311

RÉSUMÉ

Cholangiocarcinoma (CCA) presents a significant clinical challenge which is often identified in advanced stages, therby restricting the effectiveness of surgical interventions for most patients. The high incidence of cancer recurrence and resistance to chemotherapy further contribute to a bleak prognosis and low survival rates. To address this pressing need for effective therapeutic strategies, our study focuses on the development of an innovative cellular immunotherapy, specifically utilizing chimeric antigen receptor (CAR)-engineered natural killer (NK) cells designed to target the cMET receptor tyrosine kinase. In this investigation, we initiated the screening of a phage library displaying human single-chain variable fragment (ScFv) to identify novel ScFv molecules with specificity for cMET. Remarkably, ScFv11, ScFv72, and ScFv114 demonstrated exceptional binding affinity, confirmed by molecular docking analysis. These selected ScFvs, in addition to the well-established anti-cMET ScFvA, were integrated into a CAR cassette harboring CD28 transmembrane region-41BB-CD3ζ domains. The resulting anti-cMET CAR constructs were transduced into NK-92 cells, generating potent anti-cMET CAR-NK-92 cells. To assess the specificity and efficacy of these engineered cells, we employed KKU213A cells with high cMET expression and KKU055 cells with low cMET levels. Notably, co-culture of anti-cMET CAR-NK-92 cells with KKU213A cells resulted in significantly increased cell death, whereas no such effect was observed with KKU055 cells. In summary, our study identified cMET as a promising therapeutic target for CCA. The NK-92 cells, armed with the anti-cMET CAR molecule, have shown strong ability to kill cancer cells specifically, indicating their potential as a promising treatment for CCA in the future.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Cellules tueuses naturelles , Protéines proto-oncogènes c-met , Récepteurs chimériques pour l'antigène , Anticorps à chaîne unique , Humains , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/usage thérapeutique , Anticorps à chaîne unique/immunologie , Cholangiocarcinome/thérapie , Cholangiocarcinome/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Cellules tueuses naturelles/immunologie , Lignée cellulaire tumorale , Tumeurs des canaux biliaires/thérapie , Tumeurs des canaux biliaires/immunologie , Protéines proto-oncogènes c-met/métabolisme , Protéines proto-oncogènes c-met/immunologie , Immunothérapie adoptive/méthodes , Immunothérapie/méthodes , Médecine de précision
17.
Immunol Lett ; 268: 106881, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38810886

RÉSUMÉ

Wilms' tumor (WT1), a transcription factor highly expressed in various leukemias and solid tumors, is a highly specific intracellular tumor antigen, requiring presentation through complexation with HLA-restricted peptides.. WT1-derived epitopes are able to assemble with MHC-I and thereby be recognized by T cell receptors (TCR). Identification of new targetable epitopes derived from WT1 on solid tumors is a challenge, but meaningful for the development of therapeutics that could in this way target intracellular oncogenic proteins. In this study, we developed and comprehensively describe methods to validate the formation of the complex of WT1126-134 and HLA-A2. Subsequently, we developed an antibody fragment able to recognize the extracellular complex on the surface of cancer cells. The single chain variable fragment (scFv) of an established TCR-mimic antibody, specifically recognizing the WT1-derived peptide presented by the HLA-A2 complex, was expressed, purified, and functionally validated using a T2 cell antigen presentation model. Furthermore, we evaluated the potential of the WT1-derived peptide as a targetable extracellular antigen in multiple solid tumor cell lines. Our study describes methodology for the evaluation of WT1-derived peptides as tumor-specific antigen on solid tumors, and may facilitate the selection of potential candidates for future immunotherapy targeting WT1 epitopes.


Sujet(s)
Antigène HLA-A2 , Tumeurs , Liaison aux protéines , Protéines WT1 , Humains , Protéines WT1/immunologie , Protéines WT1/métabolisme , Antigène HLA-A2/immunologie , Antigène HLA-A2/métabolisme , Lignée cellulaire tumorale , Tumeurs/immunologie , Tumeurs/métabolisme , Tumeurs/thérapie , Antigènes néoplasiques/immunologie , Antigènes néoplasiques/métabolisme , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/métabolisme , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/génétique , Présentation d'antigène/immunologie , Épitopes/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Déterminants antigéniques des lymphocytes T/métabolisme , Peptides/immunologie , Peptides/composition chimique , Peptides/métabolisme
18.
J Biotechnol ; 387: 69-78, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38582406

RÉSUMÉ

Banana, a globally popular fruit, is widely cultivated in tropical and sub-tropical regions. After fruit harvest, remaining banana plant materials are low-value byproducts, mostly composted or used as fibre or for food packaging. As an aim to potentially increase farmer income, this study explored underutilised banana biomass as a novel plant tissue for production of a high-value product. Protein scFvTG130 used in this study, is an anti-toxoplasma single chain variable fragment antibody that can be used in diagnostics and neutralising the Toxoplasma gondii pathogen. Using detached banana leaves, we investigated the factors influencing the efficacy of a transient expression system using reporter genes and recombinant protein, scFvTG130. Transient expression was optimal at 2 days after detached banana leaves were vacuum infiltrated at 0.08 MPa vacuum pressure for a duration of 3 min with 0.01% (v/v) Tween20 using Agrobacterium strain GV3101 harbouring disarmed virus-based vector pIR-GFPscFvTG130. The highest concentration of anti-toxoplasma scFvTG130 antibody obtained using detached banana leaves was 22.8 µg/g fresh leaf tissue. This first study using detached banana leaf tissue for the transient expression of a recombinant protein, successfully demonstrated anti-toxoplasma scFvTG130 antibody expression, supporting the potential application for other related proteins using an underutilised detached banana leaf tissue.


Sujet(s)
Musa , Feuilles de plante , Anticorps à chaîne unique , Musa/génétique , Musa/immunologie , Feuilles de plante/métabolisme , Feuilles de plante/génétique , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie , Protéines recombinantes/génétique , Toxoplasma/génétique , Agrobacterium/génétique , Végétaux génétiquement modifiés/génétique , Agriculture/méthodes
19.
Biotechnol Lett ; 46(3): 385-398, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38607601

RÉSUMÉ

BACKGROUND: Diphtheria can be prevented by vaccination, but some epidemics occur in several places, and diphtheria's threat is considerable. Administration of diphtheria antitoxin (DAT) produced from hyperimmunized animals is the most common treatment. Recombinant human antibody fragments such as single-chain variable fragments (scFv) produced by phage display library may introduce an interesting approach to overcome the limitations of the traditional antibody therapy. In the present study, B cells of immunized volunteers were used to construct a human single-chain fragment (HuscFv) library. MATERIALS AND METHODS: The library was constructed with the maximum combination of heavy and light chains. As an antigen, Diphtheria toxoid (DTd) was used in four-round phage bio-panning to select phage clones that display DTd bound HuscFv from the library. After panning, individual scFv clones were selected. Clones that were able to detect DTd in an initial screening assay were transferred to Escherichia coli HB2151 to express the scFvs and purification was followed by Ni metal ion affinity chromatography. Toxin neutralization test was performed on Vero cells. The reactivity of the soluble scFv with diphtheria toxin were done and affinity calculation based on Beatty method was calculated. RESULTS: The size of the constructed scFv library was calculated to be 1.3 × 106 members. Following four rounds of selection, 40 antibody clones were isolated which showed positive reactivity with DTd in an ELISA assay. Five clones were able to neutralize DTd in Vero cell assay. These neutralizing clones were used for soluble expression and purification of scFv fragments. Some of these soluble scFv fragments show neutralizing activity ranging from 0.6 to 1.2 µg against twofold cytotoxic dose of diphtheria toxin. The affinity constant of the selected scFv antibody was determined almost 107 M-1. CONCLUSION: This study describes the prosperous construction and isolation of scFv from the immune library, which specifically neutralizes diphtheria toxin. The HuscFv produced in this study can be a potential candidate to substitute the animal antibody for treating diphtheria and detecting toxins.


Sujet(s)
Anticorps neutralisants , Toxine diphtérique , Anticorps à chaîne unique , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/isolement et purification , Animaux , Humains , Cellules Vero , Toxine diphtérique/immunologie , Toxine diphtérique/génétique , Anticorps neutralisants/immunologie , Techniques d'exposition à la surface cellulaire , Banque de peptides , Chlorocebus aethiops , Escherichia coli/génétique , Escherichia coli/métabolisme
20.
Viruses ; 16(4)2024 04 12.
Article de Anglais | MEDLINE | ID: mdl-38675937

RÉSUMÉ

Antibodies that specifically bind to individual human fragment crystallizable γ receptors (FcγRs) are of interest as research tools in studying immune cell functions, as well as components in bispecific antibodies for immune cell engagement in cancer therapy. Monoclonal antibodies for human low-affinity FcγRs have been successfully generated by hybridoma technology and are widely used in pre-clinical research. However, the generation of monoclonal antibodies by hybridoma technology that specifically bind to the high-affinity receptor FcγRI is challenging. Monomeric mouse IgG2a, IgG2b, and IgG3 bind human FcγRI with high affinity via the Fc part, leading to an Fc-mediated rather than a fragment for antigen binding (Fab)-mediated selection of monoclonal antibodies. Blocking the Fc-binding site of FcγRI with an excess of human IgG or Fc during screening decreases the risk of Fc-mediated interactions but can also block the potential epitopes of new antibody candidates. Therefore, we replaced hybridoma technology with phage display of a single-chain fragment variable (scFv) antibody library that was generated from mice immunized with FcγRI-positive cells and screened it with a cellular panning approach assisted by next-generation sequencing (NGS). Seven new FcγRI-specific antibody sequences were selected with this methodology, which were produced as Fc-silent antibodies showing FcγRI-restricted specificity.


Sujet(s)
Anticorps monoclonaux , Récepteurs du fragment Fc des IgG , Récepteurs du fragment Fc des IgG/immunologie , Récepteurs du fragment Fc des IgG/métabolisme , Animaux , Souris , Humains , Anticorps monoclonaux/immunologie , Immunoglobuline G/immunologie , Immunisation , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/génétique , Banque de peptides , Techniques d'exposition à la surface cellulaire , Hybridomes , Spécificité des anticorps , Femelle , Souris de lignée BALB C
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE