Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.352
Filter
1.
MAbs ; 14(1): 2021601, 2022.
Article in English | MEDLINE | ID: mdl-35030983

ABSTRACT

Coronavirus disease 2019, caused by SARS-CoV-2, remains an on-going pandemic, partly due to the emergence of variant viruses that can "break-through" the protection of the current vaccines and neutralizing antibodies (nAbs), highlighting the needs for broadly nAbs and next-generation vaccines. We report an antibody that exhibits breadth and potency in binding the receptor-binding domain (RBD) of the virus spike glycoprotein across SARS coronaviruses. Initially, a lead antibody was computationally discovered and crystallographically validated that binds to a highly conserved surface of the RBD of wild-type SARS-CoV-2. Subsequently, through experimental affinity enhancement and computational affinity maturation, it was further developed to bind the RBD of all concerning SARS-CoV-2 variants, SARS-CoV-1 and pangolin coronavirus with pico-molar binding affinities, consistently exhibited strong neutralization activity against wild-type SARS-CoV-2 and the Alpha and Delta variants. These results identify a vulnerable target site on coronaviruses for development of pan-sarbecovirus nAbs and vaccines.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Broadly Neutralizing Antibodies/immunology , COVID-19/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Antibodies, Viral/genetics , Antibodies, Viral/metabolism , Antibody Affinity , Antibody Specificity , Antigen-Antibody Reactions , Antigens, Viral/chemistry , Antigens, Viral/genetics , Broadly Neutralizing Antibodies/genetics , Broadly Neutralizing Antibodies/metabolism , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/immunology , Humans , Immunoglobulin Fragments/immunology , Molecular Docking Simulation , Monte Carlo Method , Neutralization Tests , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Domains , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics
2.
J Immunol ; 208(3): 772-779, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35022271

ABSTRACT

Human IgE is useful for immunological assays, such as sensitization of FcεRI-positive cells and IgE measurement. In this study, we report the development of a recombinant Ig fragment, designated IgCw-γεκ, as an alternative reagent to human IgE. IgCw-γεκ (∼130 kDa) comprises two hybrid constant H chain regions (Cγ1-Cε2-4, each ∼53 kDa) and two constant κ L chains (Cκ, each ∼12 kDa) and lacks a V domain. The presence of Cγ1 instead of Cε1 within the H chain increased the production yield and facilitated assembly of the H and L chains. IgCw-γεκ was produced in cultured human embryonic kidney 293F cells, with a yield of ∼27 mg/l. IgCw-γεκ bound to human FcεRIαRs expressed on the surface of rat basophilic leukemia-2H3 cells. A ß-hexosaminidase release assay revealed that the biological activity of IgCw-γεκ was comparable with that of IgE. The IgE concentration measured using IgCw-γεκ as a standard was similar to that measured using IgE as a standard. These results suggest that the IgCw-γεκ molecule retains the basic characteristics of IgE, but does not cross-react with Ags, making it an alternative to the IgE isotype references used in a variety of immunological assays.


Subject(s)
Immunoglobulin E/immunology , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin kappa-Chains/immunology , Animals , Cell Line , HEK293 Cells , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin kappa-Chains/genetics , Indicators and Reagents , Rats , Recombinant Proteins/genetics , Recombinant Proteins/immunology
3.
Bull Cancer ; 108(10S): S168-S180, 2021 Oct.
Article in French | MEDLINE | ID: mdl-34920800

ABSTRACT

CAR-T cells originate from two different approaches, cellular immunotherapy based on tumor immunosurveillance by T lymphocytes, combined with molecular engineering of bispecific antibodies and antibody fragments. The latter makes it possible to retarget immune effector cytotoxic cells (such as NK cells and T lymphocytes) to tumor cells through the binding to tumor-associated antigens. We present herein the history of bispecific antibodies, highlighting how such antibodies played a major role in CAR-T cell development. We will first evoke how antibody engineering led to the construction of various bispecific formats, in particular using the single chain Fv fragment (scFv) which has been used as the initial building block to generate chimeric bi-, tri- or multifunctional molecules. We will also describe how bispecific antibodies, either full IgG or as scFv or F(ab')2 format, directed against Fcγ receptors or CD3ɛ and against tumor-associated antigens, induce a potent anti-tumor cytotoxicity following the recruitment and activation of immune effector cells, including CD3+ T lymphocytes. These anti-tumor effects have been translated into the clinics, especially to treat malignant hemopathies. At last, recently generated bispecific CAR-T cells suggest that the embrace between cell therapy and bispecific antibodies is not over and that we are yet to witness further discoveries enabling these cells to be even more efficient.


Subject(s)
Antibodies, Bispecific/therapeutic use , Immunotherapy, Adoptive/trends , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Antibodies, Bispecific/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Clinical Trials as Topic , Forecasting , Genetic Engineering , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, IgG/immunology
4.
Front Immunol ; 12: 731845, 2021.
Article in English | MEDLINE | ID: mdl-34616400

ABSTRACT

Superantigens are unconventional antigens which recognise immune receptors outside their usual recognition sites e.g. complementary determining regions (CDRs), to elicit a response within the target cell. T-cell superantigens crosslink T-cell receptors and MHC Class II molecules on antigen-presenting cells, leading to lymphocyte recruitment, induction of cytokine storms and T-cell anergy or apoptosis among many other effects. B-cell superantigens, on the other hand, bind immunoglobulins on B-cells, affecting opsonisation, IgG-mediated phagocytosis, and driving apoptosis. Here, through a review of the structural basis for recognition of immune receptors by superantigens, we show that their binding interfaces share specific physicochemical characteristics when compared with other protein-protein interaction complexes. Given that antibody-binding superantigens have been exploited extensively in industrial antibody purification, these observations could facilitate further protein engineering to optimize the use of superantigens in this and other areas of biotechnology.


Subject(s)
Antigen-Presenting Cells/metabolism , B-Lymphocytes/metabolism , Histocompatibility Antigens Class II/metabolism , Immunoglobulin Fragments/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , Superantigens/metabolism , T-Lymphocytes/metabolism , Animals , Antibodies/isolation & purification , Antigen-Presenting Cells/immunology , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Clonal Anergy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/metabolism , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin Fragments/immunology , Protein Engineering , Receptors, Antigen, T-Cell/immunology , Signal Transduction , Superantigens/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
5.
Nat Commun ; 12(1): 469, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33473140

ABSTRACT

Antibody cocktails represent a promising approach to prevent SARS-CoV-2 escape. The determinants for selecting antibody combinations and the mechanism that antibody cocktails prevent viral escape remain unclear. We compared the critical residues in the receptor-binding domain (RBD) used by multiple neutralizing antibodies and cocktails and identified a combination of two antibodies CoV2-06 and CoV2-14 for preventing viral escape. The two antibodies simultaneously bind to non-overlapping epitopes and independently compete for receptor binding. SARS-CoV-2 rapidly escapes from individual antibodies by generating resistant mutations in vitro, but it doesn't escape from the cocktail due to stronger mutational constraints on RBD-ACE2 interaction and RBD protein folding requirements. We also identified a conserved neutralizing epitope shared between SARS-CoV-2 and SARS-CoV for antibody CoV2-12. Treatments with CoV2-06 and CoV2-14 individually and in combination confer protection in mice. These findings provide insights for rational selection and mechanistic understanding of antibody cocktails as candidates for treating COVID-19.


Subject(s)
Antibodies, Monoclonal/pharmacology , COVID-19 Drug Treatment , SARS-CoV-2/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , COVID-19/virology , Chlorocebus aethiops , Disease Models, Animal , Female , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Models, Molecular , Mutation , Protein Binding , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vero Cells
6.
Anal Biochem ; 616: 114090, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33383001

ABSTRACT

The free sulfhydryl groups of the hinge region of monovalent antibody fragments (rIgG) allow the orientation of rIgG on functionalized surfaces in immunosensors. To evaluate the contribution of reduction and orientation on signal enhancement we compared the performance of whole antibodies and their rIgG in ELISA performed on polystyrene or maleimide-functionalized microplates. Monoclonal anti-horseradish peroxidase (anti-HRP) and monoclonal anti-fPSA antibodies (1 mg/mL) were reduced with 2-mercaptoethylamine (53 mM). Western blot confirmed the presence of rIgG as a band at 75 kDa, detectable only by anti-heavy chain but not by anti-light chain antibodies, suggesting a possible folding rearrangement. Using anti-HRP we confirmed the retention of the antigen binding capacity of rIgG. Moreover, we observed a signal enhancement for rIgG even if randomly absorbed on polystyrene [linear regression slope (95%CI): rIgG 0.524 (0.434-0.614), IgG 0.370 (0.430-0.399); P = 0.0016] suggesting that chemical reduction might affect the antigen binding capacity of antibodies. ELISA with anti-fPSA rIgG coated on polystyrene confirmed these observations. Oriented anti-fPSA rIgG on a maleimide surface showed comparable signals to the assay performed on polystyrene for each analyzed concentration of antigen (PANOVA = 0.1980), anyway, with a significant improvement of the repeatability likely providing a more homogeneous capturing surface (SD rIgGmaleimide-rIgGpolystirene: fPSA 0.725 ng/mL:0.74-2.89; 1.45 ng/mL:1.56-8.69; 3.625 ng/mL:3.52-15.03; 7.25 ng/mL:7.78-18.44).


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Enzyme-Linked Immunosorbent Assay/methods , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/immunology , Animals , Binding Sites, Antibody , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Maleimides/chemistry , Oxidation-Reduction , Polystyrenes/chemistry , Reproducibility of Results , Sulfhydryl Compounds/chemistry
7.
Front Immunol ; 12: 670561, 2021.
Article in English | MEDLINE | ID: mdl-35003053

ABSTRACT

Broadly neutralizing antibodies (bNAbs), known to mediate immune control of HIV-1 infection, only develop in a small subset of HIV-1 infected individuals. Despite being traditionally associated with patients with high viral loads, bNAbs have also been observed in therapy naïve HIV-1+ patients naturally controlling virus replication [Virus Controllers (VCs)]. Thus, dissecting the bNAb response in VCs will provide key information about what constitutes an effective humoral response to natural HIV-1 infection. In this study, we identified a polyclonal bNAb response to natural HIV-1 infection targeting CD4 binding site (CD4bs), V3-glycan, gp120-gp41 interface and membrane-proximal external region (MPER) epitopes on the HIV-1 envelope (Env). The polyclonal antiviral antibody (Ab) response also included antibody-dependent cellular phagocytosis of clade AE, B and C viruses, consistent with both the Fv and Fc domain contributing to function. Sequence analysis of envs from one of the VCs revealed features consistent with potential immune pressure and virus escape from V3-glycan targeting bNAbs. Epitope mapping of the polyclonal bNAb response in VCs with bNAb activity highlighted the presence of gp120-gp41 interface and CD4bs antibody classes with similar binding profiles to known potent bNAbs. Thus, these findings reveal the induction of a broad and polyfunctional humoral response in VCs in response to natural HIV-1 infection.


Subject(s)
Broadly Neutralizing Antibodies/immunology , CD4 Antigens/immunology , Epitopes/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV-1/immunology , Peptide Fragments/immunology , Survivors , Viremia/immunology , Antibody Specificity , Binding Sites, Antibody , CD4 Antigens/metabolism , CD4 Lymphocyte Count , Epitope Mapping , Female , Genes, env , HLA-B Antigens/immunology , Humans , Immune Evasion , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fragments/immunology , Male , Models, Molecular , Phagocytosis , Protein Domains , Recombinant Proteins/immunology , Viral Load
8.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255488

ABSTRACT

The accumulation of amyloid protein aggregates in tissues is the basis for the onset of diseases known as amyloidoses. Intriguingly, many amyloidoses impact the central nervous system (CNS) and usually are devastating diseases. It is increasingly apparent that neurotoxic soluble oligomers formed by amyloidogenic proteins are the primary molecular drivers of these diseases, making them lucrative diagnostic and therapeutic targets. One promising diagnostic/therapeutic strategy has been the development of antibody fragments against amyloid oligomers. Antibody fragments, such as fragment antigen-binding (Fab), scFv (single chain variable fragments), and VHH (heavy chain variable domain or single-domain antibodies) are an alternative to full-length IgGs as diagnostics and therapeutics for a variety of diseases, mainly because of their increased tissue penetration (lower MW compared to IgG), decreased inflammatory potential (lack of Fc domain), and facile production (low structural complexity). Furthermore, through the use of in vitro-based ligand selection, it has been possible to identify antibody fragments presenting marked conformational selectivity. In this review, we summarize significant reports on antibody fragments selective for oligomers associated with prevalent CNS amyloidoses. We discuss promising results obtained using antibody fragments as both diagnostic and therapeutic agents against these diseases. In addition, the use of antibody fragments, particularly scFv and VHH, in the isolation of unique oligomeric assemblies is discussed as a strategy to unravel conformational moieties responsible for neurotoxicity. We envision that advances in this field may lead to the development of novel oligomer-selective antibody fragments with superior selectivity and, hopefully, good clinical outcomes.


Subject(s)
Amyloid/immunology , Amyloidosis/diagnosis , Neurotoxicity Syndromes/diagnosis , Protein Aggregation, Pathological/diagnosis , Amyloid/antagonists & inhibitors , Amyloidosis/immunology , Amyloidosis/pathology , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fragments/immunology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Peptide Fragments/immunology , Protein Aggregation, Pathological/immunology , Single-Domain Antibodies , Structure-Activity Relationship
9.
Expert Opin Ther Pat ; 30(12): 907-909, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32970473

ABSTRACT

INTRODUCTION: To date, microbial infections are also difficult to eradicate due to the increasing capability of bacteria to form a biofilm. In the era of antibiotic resistance, antibody-based approaches represent great promise in curing infective pathogens. The authors of US2019338018 patent propose a method for the treatment of biofilm-related disorders by using specific antibody fragments. AREAS COVERED: The US2019338018 patent reports antibody fragments, pharmaceutical composition that contains it, and their application for the treatment of biofilm-linked disorders. Proof concept and preclinical results show that mAb mIhfB4NTHI Fab caused robust eradication of the biofilm in the middle ear lumen of chinchillas affected by Hemophilus influenzae infection. EXPERT OPINION: Fab fragments of the US2019338018 patent are new in a general concept to treat bacterial biofilms and biofilm-linked disorders. However, pre-clinical data are only shown for the treatment with Fab fragments of infections caused by H. influenzae in the middle ear of chinchillas. There are no clinical trials that demonstrate that the treatment with Fab fragments may induce a disruption of biofilm produced by H. influenzae or other pathogens and an anti-inflammatory response in infected patients.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Haemophilus Infections/drug therapy , Immunoglobulin Fragments/immunology , Animals , Chinchilla , Ear, Middle/drug effects , Ear, Middle/microbiology , Haemophilus Infections/immunology , Haemophilus influenzae/drug effects , Haemophilus influenzae/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Patents as Topic
10.
J Drug Target ; 28(10): 1018-1033, 2020 12.
Article in English | MEDLINE | ID: mdl-32434407

ABSTRACT

Monoclonal antibodies (mAbs) are a swiftly growing class of targeted therapeutics for malignancies. After their first advent, the antibody (Ab) engineering trail has shown an evolutionary trajectory - from the rodent-derived Abs to the chimeric, humanised and fully human Abs with higher efficacy and lower/no immunotoxicity. Despite possessing great clinical potentials, several reports have highlighted that monospecific mAbs, even with high-affinity, often fail to induce sufficient immunologic responses. The full activation of the immune system demands cooperative interactions of immunotherapies with target antigen (Ag) towards functional avidity. Although the monospecific mAbs show affinity to a target Ag, they often fail to render sufficient avidity necessary for the activation of intracellular signalling mechanisms and the provocation of the immune system. Thus, various Ab/non-Ab scaffolds with much greater therapeutic impacts have been engineered based on the adjustment of their affinity and avidity balance. Novel multivalent Ab scaffolds (e.g. MDX-447, MT110, CD20Bi, TF2 and FBTA05) and mimetic Abs (e.g. adnectin, DARPins and ecallantide) offer improved pharmacokinetic and pharmacodynamic properties. Here, we discuss the avidity and multivalency and provide comprehensive insights into advanced Ab scaffolds used for immunotargeting and therapy of cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Immunotherapy/methods , Neoplasms/drug therapy , Antibodies, Monoclonal/administration & dosage , Antibody Affinity/immunology , Antigens/immunology , Antineoplastic Agents, Immunological/administration & dosage , Humans , Immunoglobulin Fragments/immunology , Tumor Microenvironment/immunology
11.
Methods Mol Biol ; 2136: 255-268, 2020.
Article in English | MEDLINE | ID: mdl-32430827

ABSTRACT

High-affinity monoclonal antibodies are valuable tools for studying the humoral immune response to Group A Streptococcus (GAS) antigens. This protocol describes a method for the selection of monoclonal antibody fragments that bind to GAS antigens using either naïve or immune repertoires displayed on the surface of M13 bacteriophage. Clones that specifically bind to GAS antigens are enriched for during the biopanning process, in which antibody-phage clones bind to an immobilized GAS antigen and are then washed, eluted, and amplified for subsequent rounds of selection. After the final round of biopanning, individual clones are screened by phage enzyme-linked immunosorbent assay (ELISA), and unique clones are identified by DNA fingerprinting and sequencing. The isolated monoclonal antibodies can be used to explore antibody-antigen interactions in molecular detail and provide insight into the protective mechanisms from GAS infection.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Cell Surface Display Techniques/methods , Streptococcus pyogenes/immunology , Antibodies, Monoclonal/immunology , Antigens , Bacteriophage M13/genetics , Bacteriophage M13/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunity, Humoral/immunology , Immunoglobulin Fragments/immunology , Peptide Library
12.
MAbs ; 12(1): 1752529, 2020.
Article in English | MEDLINE | ID: mdl-32316838

ABSTRACT

The success of therapeutic antibodies is largely attributed for their exquisite specificity, homogeneity, and functionality. There is, however, a need to engineer antibodies to extend and enhance their potency. One parameter is functional affinity augmentation, since antibodies matured in vivo have a natural affinity threshold. Generation of multivalent antibodies is one option capable of surpassing this affinity threshold through increased avidity. In this study, we present a novel platform consisting of an array of multivalent antibody formats, termed Quads, generated using the self-assembling tetramerization domain from p53. We demonstrate the versatility of this tetramerization domain by engineering anti-tumor necrosis factor (TNF) Quads that exhibit major increases in binding potency and in neutralizing TNF-mediated cytotoxicity compared to parental anti-TNF molecules. Further, Quads are amenable to fusion with different binding domains, allowing generation of novel multivalent monospecific and bispecific formats. Quads are thus a novel group of molecules that can be engineered to yield potential therapeutics with novel modalities and potencies.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibody Affinity/immunology , Protein Engineering/methods , Protein Multimerization/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antigens, CD20/genetics , Antigens, CD20/immunology , Antigens, CD20/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , HEK293 Cells , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Mice , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
13.
Food Chem ; 321: 126685, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32240918

ABSTRACT

In this study, we demonstrated the feasibility of isolating recombinant phage-antibodies against gluten from a non-immunized library of human single-domain antibodies (dAbs). Phage display technology enabled the selection of affinity probes by successive rounds of biopanning against a biotinylated synthetic peptide comprising repetitive immunogenic gluten motifs. The analysis of a wide representation of heterologous plant species corroborated that two of the isolated clones were specific to wheat, barley and rye proteins. The phage antibody selected as the most appropriate clone for the detection of gluten in foods (dAb8E-phage) was further applied in an indirect ELISA to the analysis of 50 commercial food samples. Although the limit of detection achieved did not improve those of current immunoassays, the proposed methodology could provide promising new pathways for the generation of recombinant antibodies that allow a comprehensive determination of gluten in foods, whilst replacing the need for animal immunization.


Subject(s)
Allergens/immunology , Food Analysis , Glutens/immunology , Hordeum/immunology , Plant Proteins/immunology , Secale/immunology , Triticum/immunology , Enzyme-Linked Immunosorbent Assay , Food , Glutens/analysis , Hordeum/chemistry , Humans , Immunoglobulin Fragments/immunology , Peptide Library , Plant Proteins/chemistry , Secale/chemistry , Triticum/chemistry
14.
J Infect Dis ; 222(3): 470-478, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32211769

ABSTRACT

Human noroviruses cause an estimated 685 million infections and 200 000 deaths annually worldwide. Although vaccines against GII.4 and GI.1 genotypes are under development, no information is available regarding vaccines or monoclonal antibodies to other noroviral genotypes. Here, we developed 2 variable-domain llama heavy-chain antibody fragment (VHHs) clones, 7C6 and 1E4, against GII.4 and GII.17 human noroviruses, respectively. Although 7C6 cross-reacted with virus-like particles (VLPs) of GII.17, GII.6, GII.3, and GII.4, it neutralized only GII.4 norovirus. In contrast, 1E4 reacted with and neutralized only GII.17 VLPs. Both VHHs blocked VLP binding to human induced pluripotent stem cell-derived intestinal epithelial cells and carbohydrate attachment factors. Using these 2 VHHs, we produced a heterodimeric VHH fragment that neutralized both GII.4 and GII.17 noroviruses. Because VHH fragments are heat- and acid-stable recombinant monoclonal antibodies, the heterodimer likely will be useful for oral immunotherapy and prophylaxis against GII.4 and GII.17 noroviruses in young, elderly, or immunocompromised persons.


Subject(s)
Antibodies, Monoclonal/immunology , Caliciviridae Infections/prevention & control , Capsid Proteins/immunology , Immunization, Passive/methods , Immunoglobulin Fragments/immunology , Antibodies, Monoclonal/administration & dosage , Antibodies, Viral/immunology , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Capsid Proteins/genetics , Cross Reactions , Epitopes/immunology , Humans , Immunoglobulin Fragments/administration & dosage , Induced Pluripotent Stem Cells/immunology , Norovirus/drug effects , Norovirus/genetics , Norovirus/immunology , Recombinant Proteins/immunology
15.
Antiviral Res ; 174: 104666, 2020 02.
Article in English | MEDLINE | ID: mdl-31760108

ABSTRACT

Argentine haemorrhagic fever (AHF) is a rodent-borne disease with a lethality as high as ~30%, which is caused by the New World arenavirus, Junín virus (JUNV). It was once a major epidemic in South America and puts millions of people in Argentina at risk. Here, we aimed to develop horse antibodies or antibody fragments against JUNV. Before preparing the horse antibodies, a strategy to efficiently generate horse antisera was established based on comparisons among immunogens and immunization methods in both mice and horses. Antisera against JUNV were finally obtained by vaccinating horses with vesicular stomatitis virus pseudotypes bearing JUNV GP. The horse antibodies IgG and F(ab')2 were subsequently demonstrated to effectively neutralize vesicular stomatitis virus pseudotypes bearing JUNV GP and to show some cross-neutralization against pathogenic New World arenaviruses. Further research revealed that Asp123 on GP1 is an important site for the binding of antibodies targeting mainly JUNV GP1 for neutralization. Collectively, this study presents an efficient strategy to develop horse antisera against JUNV and provides GP1-specific horse antibodies as potential therapeutics for AHF.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemorrhagic Fever, American/veterinary , Horses/immunology , Immunoglobulin Fragments/immunology , Junin virus/immunology , Animals , Cross Reactions , Female , Hemorrhagic Fever, American/immunology , Mice , Mice, Inbred BALB C , Neutralization Tests
16.
Sci Rep ; 9(1): 14907, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624303

ABSTRACT

Carbonic anhydrase IX (CAIX) is a cancer-associated molecular target for several classes of therapeutics. CAIX is overexpressed in a large fraction of renal cell carcinomas (RCC). Radionuclide molecular imaging of CAIX-expression might offer a non-invasive methodology for stratification of patients with disseminated RCC for CAIX-targeting therapeutics. Radiolabeled monoclonal antibodies and their fragments are actively investigated for imaging of CAIX expression. Promising alternatives are small non-immunoglobulin scaffold proteins, such as affibody molecules. A CAIX-targeting affibody ZCAIX:2 was re-designed with the aim to decrease off-target interactions and increase imaging contrast. The new tracer, DOTA-HE3-ZCAIX:2, was labeled with 111In and characterized in vitro. Tumor-targeting properties of [111In]In-DOTA-HE3-ZCAIX:2 were compared head-to-head with properties of the parental variant, [99mTc]Tc(CO)3-HE3-ZCAIX:2, and the most promising antibody fragment-based tracer, [111In]In-DTPA-G250(Fab')2, in the same batch of nude mice bearing CAIX-expressing RCC xenografts. Compared to the 99mTc-labeled parental variant, [111In]In-DOTA-HE3-ZCAIX:2 provides significantly higher tumor-to-lung, tumor-to-bone and tumor-to-liver ratios, which is essential for imaging of CAIX expression in the major metastatic sites of RCC. [111In]In-DOTA-HE3-ZCAIX:2 offers significantly higher tumor-to-organ ratios compared with [111In]In-G250(Fab')2. In conclusion, [111In]In-DOTA-HE3-ZCAIX:2 can be considered as a highly promising tracer for imaging of CAIX expression in RCC metastases based on our results and literature data.


Subject(s)
Carbonic Anhydrase IX/antagonists & inhibitors , Carcinoma, Renal Cell/diagnostic imaging , Kidney Neoplasms/diagnostic imaging , Molecular Imaging/methods , Molecular Probes/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX/immunology , Carbonic Anhydrase IX/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Humans , Immunoglobulin Fragments/administration & dosage , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/pharmacology , Indium Radioisotopes , Kidney Neoplasms/pathology , Mice , Molecular Probes/chemistry , Molecular Probes/immunology , Molecular Probes/pharmacology , Radioactive Tracers , Single Photon Emission Computed Tomography Computed Tomography/methods , Tissue Distribution , Xenograft Model Antitumor Assays
17.
Bioconjug Chem ; 30(10): 2624-2633, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31498995

ABSTRACT

Regulatory T (Treg) cells play a major role in immune suppression permitting tumors to evade immune surveillance. Depletion of intratumoral Treg cells can result in tumor regression. However, systemic depletion of Tregs may also induce autoimmune adverse events. Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed cell-specific cancer therapy that locally kills specific cells in the tumor. Antibody-photoabsorber (IRDye700DX) conjugates (APC) are injected and bind to the tumor, and subsequent administration of NIR light to the tumor results in rapid cell death only in targeted cells. CD25-targeted NIR-PIT has been shown to induce spatially selective depletion of tumor-associated Treg cells. In this study, we compared the efficacy of an antibody fragment, anti-CD25-F(ab')2, and a full antibody, anti-CD25-IgG, as agents for NIR-PIT. Tumor-bearing mice were divided into four groups: (1) no treatment; (2) anti-CD25-IgG-IR700 i.v. only; (3) anti-CD25-F(ab')2-IR700 i.v. with NIR light exposure; and (4) anti-CD25-IgG-IR700 i.v. with NIR light exposure. Although both CD25-targeted NIR-PITs resulted in significant tumor growth inhibition, the anti-CD25-F(ab')2-IR700 based NIR-PIT was superior to the anti-CD25-IgG-IR700 NIR-PIT. The anti-CD25-F(ab')2-IR700 demonstrated faster clearance from the body than the anti-CD25-IgG-IR700. Sustained circulation of anti-CD25-IgG-IR700 may block IL-2 binding on the activated effector T-cells decreasing immune response. In conclusion, anti-CD25-F(ab')2 based NIR-PIT was more effective in reducing tumor growth than anti-CD25-IgG based NIR-PIT. Absence of the Fc portion of the APC leads to faster clearance and therefore promotes a superior activated T cell response in tumors.


Subject(s)
Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/therapeutic use , Immunotherapy/methods , Interleukin-2 Receptor alpha Subunit/immunology , Phototherapy/methods , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Mice , Optical Imaging
18.
Artif Cells Nanomed Biotechnol ; 47(1): 3621-3630, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31468992

ABSTRACT

With the arrival of the precision medicine and personalized treatment era, targeted therapy that improves efficacy and reduces side effects has become the mainstream approach of cancer treatment. Antibody fragments that further enhance penetration and retain the most critical antigen-specific binding functions are considered the focus of research targeting cancer imaging and therapy. Thanks to the superior penetration and rapid blood clearance of antibody fragments, antibody fragment-based imaging agents enable efficient and sensitive imaging of tumour sites. In tumour-targeted therapy, antibody fragments can directly inhibit tumour proliferation and growth, serve as an ideal carrier for delivery of anti-tumour drugs, or manipulate the immune system to eliminate tumour cells. In this review, the excellent physicochemical properties and the basic structure of antibody fragments are expressly depicted depicted, the progress of antibody fragments in cancer therapy and imaging are thoroughly summarized, and the future development of antibody fragments is predicted.


Subject(s)
Diagnostic Imaging/methods , Immunoglobulin Fragments/therapeutic use , Molecular Targeted Therapy/methods , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Humans , Immunoglobulin Fragments/immunology , Neoplasms/immunology
19.
PLoS One ; 14(5): e0217793, 2019.
Article in English | MEDLINE | ID: mdl-31150495

ABSTRACT

The success of the targeting of amyloid-ß (Aß) oligomers through immunotherapy in Alzheimer's disease (AD) mouse models has not been translated into the clinics. The use of single-chain variable fragments (scFvs) has been proposed to prevent the potential severe effects of full-length mAbs by precluding crystallizable fraction-mediated microglia activation. The efficacy of scFv-h3D6, a bapineuzumab-derived anti-Aß scFv, has been extensively proven. In this work, we compared scFv-h3D6-EL, an elongated variant of the scFv-h3D6, with its original version to assess whether its characteristic higher thermodynamic stability improved its pharmacokinetic parameters. Although scFv-h3D6-EL had a longer half-life than its original version, its absorption from the peritoneal cavity into the systemic compartment was lower than that of the original version. Moreover, we attempted to determine the mechanism underlying the protective effect of scFv-h3D6. We found that scFv-h3D6 showed compartmental distribution and more interestingly crossed the blood-brain barrier. In the brain, scFv-h3D6 was engulfed by glial cells or internalized by Aß peptide-containing neurons in the early phase post-injection, and was colocalized with the Aß peptide almost exclusively in glial cells in the late phase post-injection. Aß peptide levels in the brain decreased simultaneously with an increase in scFv-h3D6 levels. This observation in addition to the increased tumor necrosis factor-α levels in the late phase post-injection suggested that the engulfment of Aß peptide/scFv-h3D6 complex extruded from large neurons by phagocytic cells was the mechanism underlying Aß peptide withdrawal. The mechanism of action of scFv-h3D6 demonstrates the effectivity of Aß-immunotherapy and lays the background for other studies focused on the finding of a treatment for AD.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Immunoglobulin Fragments/immunology , Single-Chain Antibodies/immunology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacokinetics , Amyloid beta-Peptides/pharmacology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Disease Models, Animal , Humans , Immunoglobulin Fragments/pharmacology , Mice , Neurons/metabolism , Protein Sorting Signals/genetics , Protein Stability , Single-Chain Antibodies/pharmacokinetics , Single-Chain Antibodies/pharmacology , Thermodynamics
20.
J Neurochem ; 150(5): 612-625, 2019 09.
Article in English | MEDLINE | ID: mdl-31055836

ABSTRACT

Synucleinopathies including Parkinson's disease, dementia with Lewy bodies and multiple system atrophy are characterized by the abnormal accumulation and propagation of α-synuclein (α-syn) pathology in the central and peripheral nervous system as Lewy bodies or glial cytoplasmic inclusions. Several antibodies against α-syn have been developed since it was first detected as the major component of Lewy bodies and glial cytoplasmic inclusions. Over the years, researchers have generated specific antibodies that alleviate the accumulation of intracellular aggregated α-syn and associated pathology in cellular and preclinical models of synucleinopathies. So far, antibodies have been the first choice as tools for research and diagnosis and currently, a wide variety of antibody fragments have been developed as an alternative to full-length antibodies for increasing its therapeutic usefulness. Recently, conformation specific antibody-based approaches have been found to be promising as therapeutic strategies, both to block α-syn aggregation and ameliorate the resultant cytotoxicity, and as diagnostic tools. In this review, we summarize different α-syn specific antibodies and provide their usefulness in tackling synucleinopathies. This article is part of the Special Issue "Synuclein".


Subject(s)
Antibodies/immunology , Synucleinopathies/therapy , alpha-Synuclein/immunology , Antibodies/therapeutic use , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Biomarkers , Delayed Diagnosis , Epitopes/immunology , Humans , Immunoglobulin Fragments/immunology , Immunologic Tests/methods , Parkinson Disease/diagnosis , Parkinson Disease/immunology , Parkinson Disease/therapy , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/prevention & control , Protein Conformation , Protein Engineering , Recombinant Proteins/immunology , Single-Domain Antibodies/immunology , Synucleinopathies/diagnosis , Synucleinopathies/immunology , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...