Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.413
Filter
1.
Sci Rep ; 14(1): 15864, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982108

ABSTRACT

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.


Subject(s)
Antibodies, Viral , COVID-19 , Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , Single-Chain Antibodies , Humans , SARS-CoV-2/immunology , Single-Chain Antibodies/immunology , Single-Chain Antibodies/genetics , COVID-19/immunology , COVID-19/virology , Antibodies, Viral/immunology , Coronavirus Nucleocapsid Proteins/immunology , Phosphoproteins/immunology , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Peptide Library
2.
Glycobiology ; 34(8)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38963938

ABSTRACT

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.


Subject(s)
Heparin , Heparitin Sulfate , Heparitin Sulfate/chemistry , Heparitin Sulfate/immunology , Heparitin Sulfate/metabolism , Heparin/chemistry , Heparin/metabolism , Molecular Docking Simulation , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Single-Chain Antibodies/genetics , Humans , Animals , Mutagenesis, Site-Directed , Binding Sites , Amino Acid Sequence
3.
Anal Chim Acta ; 1317: 342887, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39030019

ABSTRACT

BACKGROUND: Procollagen type III N-terminal peptide (P-III-NP) is a fibrosis biomarker associated with liver and cardiac fibrosis. Despite the value of P-III-NP as a biomarker, its analysis currently relies on enzyme-linked immunosorbent assays (ELISA) and radioimmunoassays (RIA), which require more than 3 h. To facilitate early diagnosis and treatment through rapid biomarker testing, we developed a one-step immunoassay for P-III-NP using a quenchbody, which is a fluorescence-labeled immunosensor for immediate signal generation. RESULTS: To create quenchbodies, the total mRNA of P-III-NP antibodies was extracted from early-developed hybridoma cells, and genes of variable regions were obtained through cDNA synthesis, inverse PCR, and sequencing. A single-chain variable fragment (scFv) with an N-terminal Cys-tag was expressed in E. coli Shuffle T7, resulting in a final yield of 9.8 mg L-1. The fluorescent dye was labeled on the Cys-tag of the anti-P-III-NP scFv using maleimide-thiol click chemistry, and the spacer arm lengths between the maleimide-fluorescent dyes were compared. Consequently, a TAMRA-C6-labeled quenchbody exhibited antigen-dependent fluorescence signals and demonstrated its ability to detect P-III-NP at concentrations as low as 0.46 ng mL-1 for buffer samples, 1.0 ng mL-1 for 2 % human serum samples. SIGNIFICANCE: This one-step P-III-NP detection method provides both qualitative and quantitative outcomes within a concise 5-min timeframe. Furthermore, its application can be expanded using a 96-well platform and human serum, making it a high-throughput and sensitive method for testing fibrotic biomarkers.


Subject(s)
Biomarkers , Fibrosis , Fluorescent Dyes , Peptide Fragments , Procollagen , Biomarkers/blood , Biomarkers/analysis , Humans , Fluorescent Dyes/chemistry , Procollagen/blood , Procollagen/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Biosensing Techniques , Immunoassay/methods
4.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39000338

ABSTRACT

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.


Subject(s)
Artificial Intelligence , Ki-1 Antigen , Molecular Docking Simulation , Molecular Dynamics Simulation , Receptors, Chimeric Antigen , Single-Chain Antibodies , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Humans , Ki-1 Antigen/immunology , Ki-1 Antigen/metabolism , Animals , Mice , Protein Binding , Surface Plasmon Resonance
5.
Biotechnol J ; 19(7): e2300745, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39014926

ABSTRACT

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.


Subject(s)
Escherichia coli , Receptor, ErbB-2 , Recombinant Proteins , Single-Chain Antibodies , Receptor, ErbB-2/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/isolation & purification , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Cell Line, Tumor , Breast Neoplasms/immunology
6.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 549-555, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38952095

ABSTRACT

Objective To verify the anti-tumor effect of the mesenchymal-epithelial transition single-chain antibody (Met scFv) on subcutaneously transplanted tumors in nude mice. Methods A tumor model was established in nude mice by subcutaneous injection of A549 lung adenocarcinoma cells. Once the tumors were formed, IRDye680 LT N-hydroxysuccinimide (NHS) ester-labeled Met scFv was administered intraperitoneally. Real-time monitoring was conducted using a small animal imager to observe the dynamic distribution of the antibody in tumor-bearing mice. The affinity between c-Met and the antibody in tumor cells was detected. Tumor volume changes were observed and the tumor growth curve were plotted following regular tail vein injections of Met scFv. Immunohistochemical staining was employed to determine whether Met scFv could effectively bind to the c-Met antigen in tumor tissues. Results The distribution of Met scFv in nude mice showed that it was primarily located in the peritoneal cavity within the first 3 hours. After approximately 48 hours, fluorescent signals began to accumulate in the tumor tissue. Immunohistochemical staining of the tumors revealed high expression of c-Met in the tumor tissues; regular tail vein injections of Met scFv significantly slowed down the growth of tumors in mice. Conclusion Met scFv specifically recognizes tumor cells in vivo and exhibites significant anti-tumor activity.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Mice, Nude , Proto-Oncogene Proteins c-met , Single-Chain Antibodies , Animals , Humans , Proto-Oncogene Proteins c-met/immunology , Proto-Oncogene Proteins c-met/metabolism , Single-Chain Antibodies/immunology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/pharmacology , Lung Neoplasms/immunology , A549 Cells , Mice , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Injections, Intraperitoneal , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Xenograft Model Antitumor Assays , Mice, Inbred BALB C , Cell Line, Tumor
7.
PLoS One ; 19(6): e0301223, 2024.
Article in English | MEDLINE | ID: mdl-38837964

ABSTRACT

New immune checkpoints are emerging in a bid to improve response rates to immunotherapeutic drugs. The adenosine A2A receptor (A2AR) has been proposed as a target for immunotherapeutic development due to its participation in immunosuppression of the tumor microenvironment. Blockade of A2AR could restore tumor immunity and, consequently, improve patient outcomes. Here, we describe the discovery of a potent, selective, and tumor-suppressing antibody antagonist of human A2AR (hA2AR) by phage display. We constructed and screened four single-chain variable fragment (scFv) libraries-two synthetic and two immunized-against hA2AR and antagonist-stabilized hA2AR. After biopanning and ELISA screening, scFv hits were reformatted to human IgG and triaged in a series of cellular binding and functional assays to identify a lead candidate. Lead candidate TB206-001 displayed nanomolar binding of hA2AR-overexpressing HEK293 cells; cross-reactivity with mouse and cynomolgus A2AR but not human A1, A2B, or A3 receptors; functional antagonism of hA2AR in hA2AR-overexpressing HEK293 cells and peripheral blood mononuclear cells (PBMCs); and tumor-suppressing activity in colon tumor-bearing HuCD34-NCG mice. Given its therapeutic properties, TB206-001 is a good candidate for incorporation into next-generation bispecific immunotherapeutics.


Subject(s)
Adenosine A2 Receptor Antagonists , Receptor, Adenosine A2A , Humans , Animals , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/immunology , HEK293 Cells , Mice , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/therapeutic use , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology , Macaca fascicularis , Peptide Library
8.
Biosens Bioelectron ; 261: 116511, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38917513

ABSTRACT

Single-chain fragment variables (scFvs), composed of variable heavy and light chains joined together by a peptide linker, can be produced using a cost-effective bacterial expression system, making them promising candidates for pharmaceutical applications. However, a versatile method for monitoring recombinant-protein production has not yet been developed. Herein, we report a novel anti-scFv aptamer-based biosensing system with high specificity and versatility. First, anti-scFv aptamers were screened using the competitive systematic evolution of ligands by exponential enrichment, focusing on a unique scFv-specific peptide linker. We selected two aptamers, P1-12 and P2-63, with KD = 2.1 µM or KD = 1.6 µM toward anti-human epidermal growth factor receptor (EGFR) scFv, respectively. These two aptamers can selectively bind to scFv but not to anti-EGFR Fv. Furthermore, the selected aptamers recognized various scFvs with different CDRs, such as anti-4-1BB and anti-hemoglobin scFv, indicating that they recognized a unique peptide linker region. An electrochemical sensor for anti-EGFR scFv was developed using anti-scFv aptamers based on square wave voltammetry. Thus, the constructed sensor could monitor anti-EGFR scFv concentrations in the range of 10-500 nM in a diluted medium for bacterial cultivation, which covered the expected concentration range for the recombinant production of scFvs. These achievements promise the realization of continuous monitoring sensors for pharmaceutical scFv, which will enable the real-time and versatile monitoring of large-scale scFv production.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , ErbB Receptors , Single-Chain Antibodies , Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Humans , Recombinant Proteins/genetics , SELEX Aptamer Technique/methods , Electrochemical Techniques/methods
9.
Sci Rep ; 14(1): 14419, 2024 06 22.
Article in English | MEDLINE | ID: mdl-38909102

ABSTRACT

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.


Subject(s)
Antibodies, Viral , SARS-CoV-2 , Single-Chain Antibodies , Spike Glycoprotein, Coronavirus , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , SARS-CoV-2/immunology , Single-Chain Antibodies/immunology , Single-Chain Antibodies/genetics , Humans , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/virology , Animals , Antibodies, Monoclonal/immunology , Conserved Sequence
10.
Int J Mol Sci ; 25(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891821

ABSTRACT

CAR-T cell therapy is at the forefront of next-generation multiple myeloma (MM) management, with two B-cell maturation antigen (BCMA)-targeted products recently approved. However, these products are incapable of breaking the infamous pattern of patient relapse. Two contributing factors are the use of BCMA as a target molecule and the artificial scFv format that is responsible for antigen recognition. Tackling both points of improvement in the present study, we used previously characterized VHHs that specifically target the idiotype of murine 5T33 MM cells. This idiotype represents one of the most promising yet challenging MM target antigens, as it is highly cancer- but also patient-specific. These VHHs were incorporated into VHH-based CAR modules, the format of which has advantages compared to scFv-based CARs. This allowed a side-by-side comparison of the influence of the targeting domain on T cell activation. Surprisingly, VHHs previously selected as lead compounds for targeted MM radiotherapy are not the best (CAR-) T cell activators. Moreover, the majority of the evaluated VHHs are incapable of inducing any T cell activation. As such, we highlight the importance of specific VHH selection, depending on its intended use, and thereby raise an important shortcoming of current common CAR development approaches.


Subject(s)
Immunotherapy, Adoptive , Multiple Myeloma , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Humans , Animals , Immunotherapy, Adoptive/methods , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Antibodies, Anti-Idiotypic/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , B-Cell Maturation Antigen/immunology , B-Cell Maturation Antigen/metabolism , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/chemistry , Single-Chain Antibodies/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Lymphocyte Activation/immunology
11.
Nat Commun ; 15(1): 5255, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898050

ABSTRACT

GPRC5D is an atypical Class C orphan G protein-coupled receptor. Its high expression on the surface of multiple myeloma cells has rendered it an attractive target for therapeutic interventions, including monoclonal antibodies, CAR-T cells, and T-cell engagers. Despite its therapeutic potential, the insufficient understanding regarding of the receptor's structure and antibody recognition mechanism has impeded the progress of effective therapeutic development. Here, we present the structure of GPRC5D in complex with a preclinical-stage single-chain antibody (scFv). Our structural analysis reveals that the GPRC5D presents a close resemblance to the typical Class C GPCRs in the transmembrane region. We identify a distinct head-to-head homodimer arrangement and interface mainly involving TM4, setting it apart from other Class C homo- or hetero-dimers. Furthermore, we elucidate the binding site engaging a sizable extracellular domain on GPRC5D for scFv recognition. These insights not only unveil the distinctive dimer organization of this unconventional Class C GPCR but also hold the potential to advance drug development targeting GPRC5D for the treatment of multiple myeloma.


Subject(s)
Multiple Myeloma , Protein Multimerization , Receptors, G-Protein-Coupled , Single-Chain Antibodies , Humans , Multiple Myeloma/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Single-Chain Antibodies/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Protein Binding , Binding Sites , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry
12.
Front Immunol ; 15: 1407398, 2024.
Article in English | MEDLINE | ID: mdl-38933276

ABSTRACT

Poisoning by widow-spider (genus Latrodectus) bites occurs worldwide. The illness, termed latrodectism, can cause severe and persistent pain and can lead to muscle rigidity, respiratory complications, and cardiac problems. It is a global health challenge especially in developing countries. Equine serum-derived polyclonal anti-sera are commercially available as a medication for patients with latrodectism, but the use of sera imposes potential inherent risks related to its animal origin. The treatment may cause allergic reactions in humans (serum sickness), including anaphylactic shock. Furthermore, equine-derived antivenom is observed to have batch-to-batch variability and poor specificity, as it is always an undefined mix of antibodies. Because latrodectism can be extremely painful but is rarely fatal, the use of antivenom is controversial and only a small fraction of patients is treated. In this work, recombinant human antibodies were selected against alpha-latrotoxin of the European black widow (Latrodectus tredecimguttatus) by phage display from a naïve antibody gene library. Alpha-Latrotoxin (α-LTX) binding scFv were recloned and produced as fully human IgG. A novel alamarBlue assay for venom neutralization was developed and used to select neutralizing IgGs. The human antibodies showed in vitro neutralization efficacy both as single antibodies and antibody combinations. This was also confirmed by electrophysiological measurements of neuronal activity in cell culture. The best neutralizing antibodies showed nanomolar affinities. Antibody MRU44-4-A1 showed outstanding neutralization efficacy and affinity to L. tredecimguttatus α-LTX. Interestingly, only two of the neutralizing antibodies showed cross-neutralization of the venom of the Southern black widow (Latrodectus mactans). This was unexpected, because in the current literature the alpha-latrotoxins are described as highly conserved. The here-engineered antibodies are candidates for future development as potential therapeutics and diagnostic tools, as they for the first time would provide unlimited supply of a chemically completely defined drug of constant quality and efficacy, which is also made without the use of animals.


Subject(s)
Antibodies, Neutralizing , Antivenins , Black Widow Spider , Spider Venoms , Humans , Animals , Black Widow Spider/immunology , Antibodies, Neutralizing/immunology , Spider Venoms/immunology , Antivenins/immunology , Single-Chain Antibodies/immunology , Spider Bites/immunology , Immunoglobulin G/immunology
13.
Sci Rep ; 14(1): 14558, 2024 06 24.
Article in English | MEDLINE | ID: mdl-38914666

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized , Epithelial Cells , Nicotiana , Single-Chain Antibodies , Humans , Nicotiana/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Single-Chain Antibodies/pharmacology , Single-Chain Antibodies/genetics , Chemokines, CC/metabolism , Interleukin-4 Receptor alpha Subunit/metabolism , Cells, Cultured , Nasal Mucosa/metabolism , Nasal Mucosa/cytology , Nasal Mucosa/immunology
14.
Arch Biochem Biophys ; 758: 110068, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38909835

ABSTRACT

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.


Subject(s)
Antibody Affinity , Complementarity Determining Regions , Disulfides , Single-Chain Antibodies , Disulfides/chemistry , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Single-Chain Antibodies/genetics , Complementarity Determining Regions/chemistry , Humans , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Animals , Thermodynamics , Kinetics , Crystallography, X-Ray , Models, Molecular
15.
ACS Appl Bio Mater ; 7(7): 4486-4496, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38886921

ABSTRACT

Human tissue kallikrein-related peptidase 7 (KLK7) is a serine protease implicated in the physiology of skin desquamation, and its uncontrolled activity can lead to chronic diseases such as psoriasis, atopic dermatitis, and Netherton syndrome. For this reason, kallikrein 7 has been identified as a potential therapeutic target. This work aimed to evaluate Pluronic (PL) hydrogels as topical carriers of four specific scFv-Fc antibodies to inhibit KLK7. The hydrogels comprised PL F127 (30% w/v) alone and a binary F127/P123 (28-2% w/v) system. Each formulation was loaded with 1 µg/mL of each antibody and characterized by physicochemical and pharmaceutical techniques, considering antibody-micelle interactions and hydrogel behavior as smart delivery systems. Results showed that the antibodies were successfully loaded into the PL-based systems, and the sol-gel transition temperature was shifted to high values after the P123 addition. The antibodies released from the gels preserved their rheological properties (G' > G'', 35- to 41-fold) and inhibitory activity against KLK7, even after 24 h. This work presented potential agents targeting KLK7 that may provide strategies for treating skin abnormalities.


Subject(s)
Hydrogels , Kallikreins , Hydrogels/chemistry , Hydrogels/pharmacology , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , Humans , Materials Testing , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Particle Size , Poloxamer/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/administration & dosage , Temperature , Skin Diseases/drug therapy , Skin Diseases/pathology
16.
Cells ; 13(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38891106

ABSTRACT

Toxoplasma gondii holds significant therapeutic potential; however, its nonspecific invasiveness results in off-target effects. The purpose of this study is to evaluate whether T. gondii specificity can be improved by surface display of scFv directed against dendritic cells' endocytic receptor, DEC205, and immune checkpoint PD-L1. Anti-DEC205 scFv was anchored to the T. gondii surface either directly via glycosylphosphatidylinositol (GPI) or by fusion with the SAG1 protein. Both constructs were successfully expressed, but the binding results suggested that the anti-DEC-SAG1 scFv had more reliable functionality towards recombinant DEC protein and DEC205-expressing MutuDC cells. Two anti-PD-L1 scFv constructs were developed that differed in the localization of the HA tag. Both constructs were adequately expressed, but the localization of the HA tag determined the functionality by binding to PD-L1 protein. Co-incubation of T. gondii displaying anti-PD-L1 scFv with tumor cells expressing/displaying different levels of PD-L1 showed strong binding depending on the level of available biomarker. Neutralization assays confirmed that binding was due to the specific interaction between anti-PD-L1 scFv and its ligand. A mixed-cell assay showed that T. gondii expressing anti-PD-L1 scFv predominately targets the PD-L1-positive cells, with negligible off-target binding. The recombinant RH-PD-L1-C strain showed increased killing ability on PD-L1+ tumor cell lines compared to the parental strain. Moreover, a co-culture assay of target tumor cells and effector CD8+ T cells showed that our model could inhibit PD1/PD-L1 interaction and potentiate T-cell immune response. These findings highlight surface display of antibody fragments as a promising strategy of targeting replicative T. gondii strains while minimizing nonspecific binding.


Subject(s)
B7-H1 Antigen , Single-Chain Antibodies , Toxoplasma , Toxoplasma/metabolism , Toxoplasma/immunology , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Cell Line, Tumor , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism
17.
Theranostics ; 14(8): 3267-3281, 2024.
Article in English | MEDLINE | ID: mdl-38855181

ABSTRACT

Background: Myocardial infarction (MI) as a consequence of atherosclerosis-associated acute thrombosis is a leading cause of death and disability globally. Antiplatelet and anticoagulant drugs are standard therapies in preventing and treating MI. However, all clinically used drugs are associated with bleeding complications, which ultimately limits their use in patients with a high risk of bleeding. We have developed a new recombinant drug, targ-HSA-TAP, that combines targeting and specific inhibition of activated platelets as well as anticoagulation. This drug is designed and tested for a prolonged circulating half-life, enabling unique thromboprophylaxis without bleeding complications. Methods: Targ-HSA-TAP combines a single-chain antibody (scFv) that targets activated glycoprotein IIb/IIIa on activated platelets, human serum albumin (HSA) for prolonged circulation, and tick anticoagulant peptide (TAP) for coagulation FX inhibition. A non-binding scFv is employed as a non-targeting control (non-targ-HSA-TAP). Its efficacy was investigated in vivo using murine models of acute thrombosis and cardiac ischemia-reperfusion (I/R) injury. Results: Our experiments confirmed the targeting specificity of targ-HSA-TAP to activated platelets and demonstrated effective prevention of platelet aggregation and thrombus formation, as well as FXa inhibition in vitro. Thromboprophylactic administration of targ-HSA-TAP subcutaneously in mice prevented occlusion of the carotid artery after ferric chloride injury as compared to non-targ-HSA-TAP and PBS-control treated mice. By comparing the therapeutic outcomes between targ-TAP and targ-HSA-TAP, we demonstrate the significant improvements brought by the HSA fusion in extending the drug's half-life and enhancing its therapeutic window for up to 16 h post-administration. Importantly, tail bleeding time was not prolonged with targ-HSA-TAP in contrast to the clinically used anticoagulant enoxaparin. Furthermore, in a murine model of cardiac I/R injury, mice administered targ-HSA-TAP 10 h before injury demonstrated preserved cardiac function, with significantly higher ejection fraction and fractional shortening, as compared to the non-targ-HSA-TAP and PBS control groups. Advanced strain analysis revealed reduced myocardial deformation and histology confirmed a reduced infarct size in targ-HSA-TAP treated mice compared to control groups. Conclusion: The inclusion of HSA represents a significant advancement in the design of targeted therapeutic agents for thromboprophylaxis. Our activated platelet-targeted targ-HSA-TAP is a highly effective antithrombotic drug with both anticoagulant and antiplatelet effects while retaining normal hemostasis. The long half-life of targ-HSA-TAP provides the unique opportunity to use this antithrombotic drug for more effective, long-lasting and safer anti-thrombotic prophylaxis. In cases where MI occurs, this prophylactic strategy reduces thrombus burden and effectively reduces cardiac I/R injury.


Subject(s)
Blood Platelets , Hemorrhage , Serum Albumin, Human , Thrombosis , Animals , Mice , Thrombosis/prevention & control , Thrombosis/drug therapy , Humans , Hemorrhage/prevention & control , Blood Platelets/drug effects , Blood Platelets/metabolism , Disease Models, Animal , Male , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Single-Chain Antibodies/pharmacology , Single-Chain Antibodies/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/drug therapy , Myocardial Infarction/drug therapy , Mice, Inbred C57BL , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
18.
Protein Sci ; 33(7): e5084, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38923711

ABSTRACT

Small antibody fragments have recently been used as alternatives to full-length monoclonal antibodies in therapeutic applications. One of the most popular fragment antibodies is single-chain fragment variables (scFvs), consisting of variable heavy (VH) and variable light (VL) domains linked by a flexible peptide linker. scFvs have small molecular sizes, which enables good tissue penetration and low immunogenicity. Despite these advantages, the use of scFvs, especially for therapeutic purpose, is still limited because of the difficulty to regulate the binding activity and conformational stability. In this study, we constructed and analyzed 10 scFv fragments derived from 10 representatives of FDA-approved mAbs to evaluate their physicochemical properties. Differential scanning calorimetry analysis showed that scFvs exhibited relatively high but varied thermostability, from 50 to 70°C of melting temperatures, and different unfolding cooperativity. Surface plasmon resonance analysis revealed that scFvs fragments that exhibit high stability and cooperative unfolding likely tend to maintain antigen binding. This study demonstrated the comprehensive physicochemical properties of scFvs derived from FDA-approved antibodies, providing insights into antibody design and development.


Subject(s)
Protein Stability , Single-Chain Antibodies , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Humans , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Calorimetry, Differential Scanning , Protein Binding
19.
Int Immunopharmacol ; 136: 112273, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38810311

ABSTRACT

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.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Killer Cells, Natural , Proto-Oncogene Proteins c-met , Receptors, Chimeric Antigen , Single-Chain Antibodies , Humans , Single-Chain Antibodies/genetics , Single-Chain Antibodies/therapeutic use , Single-Chain Antibodies/immunology , Cholangiocarcinoma/therapy , Cholangiocarcinoma/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Killer Cells, Natural/immunology , Cell Line, Tumor , Bile Duct Neoplasms/therapy , Bile Duct Neoplasms/immunology , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/immunology , Immunotherapy, Adoptive/methods , Immunotherapy/methods , Precision Medicine
20.
Microbiol Res ; 285: 127749, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38761490

ABSTRACT

Among 5 types of the Christie-Atkins-Munch-Petersen factor (CAMP) of Cutibacterium acnes, CAMP1 is highly expressed in phylotype II as well as IB, and thought to be a virulence factor of opportunistic but fatal blood, soft tissue, and implant-related infections. The target of a human single-chain variable antibody fragment (scFv), recently isolated from a phage display library, has been identified as CAMP1 of phylotype II, using immunoprecipitation followed by mass spectrometry, phage display peptide biopanning, 3D-modelling, and ELISA. The IgG1 format of the antibody could enhance phagocytosis of C. acnes DMST 14916 by THP-1 human monocytes. Our results suggest that the antibody-dependent phagocytosis process is mediated by the caveolae membrane system and involves the induction of IL-1ß. This is the first report on the study of a human antibody against CAMP1 of C. acnes phylotype II, of which a potential use as therapeutic antibody against virulence C. acnes infection is postulated.


Subject(s)
Immunoglobulin G , Macrophages , Phagocytosis , Humans , Macrophages/immunology , Macrophages/microbiology , Immunoglobulin G/immunology , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , THP-1 Cells , Virulence Factors/immunology , Antibodies, Bacterial/immunology , Monocytes/immunology , Monocytes/microbiology , Single-Chain Antibodies/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Propionibacteriaceae/immunology
SELECTION OF CITATIONS
SEARCH DETAIL