Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Methods Mol Biol ; 2350: 105-123, 2021.
Article in English | MEDLINE | ID: mdl-34331282

ABSTRACT

Early detection of malignant tumors, micrometastases, and disseminated tumor cells is one of the effective way of fighting cancer. Among the many existing imaging methods like computed tomography (CT), ultrasound (US), magnetic resonance imaging (MRI), positron emission tomography (PET), and single-photon emission computed tomography (SPECT), optical imaging with fluorescent probes is one of the most promising alternatives because it is fast, inexpensive, safe, sensitive, and specific. However, traditional fluorescent probes, based on organic fluorescent dyes, suffer from the low signal-to-noise ratio. Furthermore, conventional organic fluorescent dyes are unsuitable for deep tissue imaging because of the strong visible light absorption by biological tissues. The use of fluorescent semiconductor nanocrystals, or quantum dots (QDs), may overcome this limitation due to their large multiphoton cross section, which ensures efficient imaging of thick tissue sections inaccessible with conventional fluorescent probes. Moreover, the lower photobleaching and higher brightness of fluorescence signals from QDs ensures a much better discrimination of positive signals from the background. The use of fluorescent nanoprobes based on QDs conjugated to uniformly oriented high-affinity single-domain antibodies (sdAbs) may significantly increase the sensitivity and specificity due to better recognition of analytes and deeper penetration into tissues due to small size of such nanoprobes.Here, we describe a protocol for the fabrication of nanoprobes based on sdAbs and QDs, preparation of experimental xenograft mouse models for quality control, and multiphoton imaging of deep-tissue solid tumors, micrometastases, and disseminated tumor cells.


Subject(s)
Fluorescent Antibody Technique/methods , Microscopy, Fluorescence, Multiphoton/methods , Neoplasms/diagnostic imaging , Neoplasms/pathology , Quantum Dots , Single-Domain Antibodies , Cell Line, Tumor , Fluorescent Antibody Technique/standards , Humans , Immunoconjugates/chemistry , Immunohistochemistry/methods , Molecular Probes , Multimodal Imaging/methods , Nanoparticles , Neoplasm Micrometastasis , Optical Imaging/methods
2.
Oncoimmunology ; 10(1): 1854529, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33457075

ABSTRACT

The natural killer group 2 member D (NKG2D) receptor is a C-type lectin-like activating receptor mainly expressed by cytotoxic immune cells including NK, CD8+ T, γδ T and NKT cells and in some pathological conditions by a subset of CD4+ T cells. It binds a variety of ligands (NKG2DL) whose expressions is finely regulated by stress-related conditions. The NKG2DL/NKG2D axis plays a central and complex role in the regulation of immune responses against diverse cellular threats such as oncogene-mediated transformations or infections. We generated a panel of seven highly specific anti-human NKG2D single-domain antibodies targeting various epitopes. These single-domain antibodies were integrated into bivalent and bispecific antibodies using a versatile plug-and-play Fab-like format. Depending on the context, these Fab-like antibodies exhibited activating or inhibitory effects on the immune response mediated by the NKG2DL/NKG2D axis. In solution, the bivalent anti-NKG2D antibodies that compete with NKG2DL potently blocked the activation of NK cells seeded on immobilized MICA, thus constituting antagonizing candidates. Bispecific anti-NKG2DxHER2 antibodies that concomitantly engage HER2 on tumor cells and NKG2D on NK cells elicited cytotoxicity of unstimulated NK in a tumor-specific manner, regardless of their apparent affinities and epitopes. Importantly, the bispecific antibodies that do not compete with ligands binding retained their full cytotoxic activity in the presence of ligands, a valuable property to circumvent immunosuppressive effects induced by soluble ligands in the microenvironment.


Subject(s)
Neoplasms , Single-Domain Antibodies , Humans , Immunity , Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily K , Tumor Microenvironment
3.
Front Immunol ; 10: 1593, 2019.
Article in English | MEDLINE | ID: mdl-31354732

ABSTRACT

Triple negative breast cancers (TNBC) remain a major medical challenge due to poor prognosis and limited treatment options. Mesothelin is a glycosyl-phosphatidyl inositol-linked membrane protein with restricted normal expression and high level expression in a large proportion of TNBC, thus qualifying as an attractive target. Its overexpression in breast tumors has been recently correlated with a decreased disease-free survival and an increase of distant metastases. The objective of the study was to investigate the relevance of a bispecific antibody-based immunotherapy approach through mesothelin targeting and CD16 engagement using a Fab-like bispecific format (MesobsFab). Using two TNBC cell lines with different level of surface mesothelin and epithelial/mesenchymal phenotypes, we showed that, in vitro, MesobsFab promotes the recruitment and penetration of NK cells into tumor spheroids, induces potent dose-dependent cell-mediated cytotoxicity of mesothelin-positive tumor cells, cytokine secretion, and decreases cell invasiveness. MesobsFab was able to induce cytotoxicity in resting human peripheral blood mononuclear cells (PBMC), mainly through its NK cells-mediated antibody dependent cell cytotoxicity (ADCC) activity. In vivo, the anti-tumor effect of MesobsFab depends upon a threshold of MSLN density on target cells. Collectively our data support mesothelin as a relevant therapeutic target for the subset of TNBC that overexpresses mesothelin characterized by a low overall and disease-free survival as well as the potential of MesobsFab as antibody-based immunotherapeutics.


Subject(s)
Antibodies, Bispecific/therapeutic use , Breast Neoplasms/therapy , GPI-Linked Proteins/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Receptors, IgG/immunology , Triple Negative Breast Neoplasms/therapy , Antibody-Dependent Cell Cytotoxicity , Breast Neoplasms/immunology , Cell Line, Tumor , Epitopes , Female , Humans , Mesothelin , Triple Negative Breast Neoplasms/immunology
4.
Biophys J ; 116(8): 1516-1526, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30979550

ABSTRACT

Antibodies are key tools in biomedical research and medicine. Their binding properties are classically measured in solution and characterized by an affinity. However, in physiological conditions, antibodies can bridge an immune effector cell and an antigen-presenting cell, implying that mechanical forces may apply to the bonds. For example, in antibody-dependent cell cytotoxicity-a major mode of action of therapeutic monoclonal antibodies-the Fab domains bind the antigens on the target cell, whereas the Fc domain binds to the activating receptor CD16 (also known as FcgRIII) of an immune effector cell, in a quasi-bidimensional environment (2D). Therefore, there is a strong need to investigate antigen/antibody binding under force (2D) to better understand and predict antibody activity in vivo. We used two anti-CD16 nanobodies targeting two different epitopes and laminar flow chamber assay to measure the association and dissociation of single bonds formed between microsphere-bound CD16 antigens and surface-bound anti-CD16 nanobodies (or single-domain antibodies), simulating 2D encounters. The two nanobodies exhibit similar 2D association kinetics, characterized by a strong dependence on the molecular encounter duration. However, their 2D dissociation kinetics strongly differ as a function of applied force: one exhibits a slip bond behavior in which off rate increases with force, and the other exhibits a catch-bond behavior in which off rate decreases with force. This is the first time, to our knowledge, that catch-bond behavior was reported for antigen-antibody bond. Quantification of natural killer cells spreading on surfaces coated with the nanobodies provides a comparison between 2D and three-dimensional adhesion in a cellular context, supporting the hypothesis of natural killer cell mechanosensitivity. Our results may also have strong implications for the design of efficient bispecific antibodies for therapeutic applications.


Subject(s)
Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mechanical Phenomena , Single-Domain Antibodies/immunology , Biomechanical Phenomena , Cell Adhesion , Cell Line , Humans
5.
Sci Rep ; 8(1): 4595, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545609

ABSTRACT

Early detection of malignant tumours and, especially, micrometastases and disseminated tumour cells is still a challenge. In order to implement highly sensitive diagnostic tools we demonstrate the use of nanoprobes engineered from nanobodies (single-domain antibodies, sdAbs) and fluorescent quantum dots (QDs) for single- and two-photon detection and imaging of human micrometastases and disseminated tumour cells in ex vivo biological samples of breast and pancreatic metastatic tumour mouse models expressing human epidermal growth factor receptor 2 (HER2) or carcinoembryonic antigen (CEA). By staining thin (5-10 µm) paraffin and thick (50 µm) agarose tissue sections, we detected HER2- and CEA-positive human tumour cells infiltrating the surrounding tissues or metastasizing to different organs, including the brain, testis, lung, liver, and lymph nodes. Compared to conventional fluorescently labelled antibodies the sdAb-HER2-QD and sdAb-CEA-QD nanoprobes are superior in detecting micrometastases in tissue sections by lower photobleaching and higher brightness of fluorescence signals ensuring much better discrimination of positive signals versus background. Very high two-photon absorption cross-sections of QDs and small size of the nanoprobes ensure efficient imaging of thick tissue sections unattainable with conventional fluorescent probes. The nanobody-QD probes will help to improve early cancer diagnosis and prognosis of progression by assessing metastasis.


Subject(s)
Breast Neoplasms/pathology , Quantum Dots/chemistry , Single-Domain Antibodies/immunology , Animals , Breast Neoplasms/metabolism , Carcinoembryonic Antigen/immunology , Cell Line, Tumor , Female , Fluorescent Dyes/chemistry , Humans , Mice , Mice, Nude , Microscopy, Confocal , Microscopy, Fluorescence, Multiphoton , Neoplasm Micrometastasis , Receptor, ErbB-2/immunology , Single-Domain Antibodies/chemistry , Transplantation, Heterologous
6.
J Nucl Med ; 59(7): 1056-1062, 2018 07.
Article in English | MEDLINE | ID: mdl-29572256

ABSTRACT

Mesothelin is a cell-surface glycoprotein restricted to mesothelial cells overexpressed in several types of cancer, including triple-negative breast cancer not responding to trastuzumab or hormone-based therapies. Mesothelin-targeting therapies are currently being developed. However, the identification of patients potentially eligible for such a therapeutic strategy remains challenging. The objective of this study was to perform the radiolabeling and preclinical evaluation of 99mTc-A1 and 99mTc-C6, two antimesothelin single-domain antibody (sdAb)-derived imaging agents. Methods: A1 and C6 were radiolabeled with 99mTc and evaluated in vitro on recombinant protein and cells, as well as in vivo in xenograft mouse models of the triple-negative breast cancer cell lines HCC70 (mesothelin-positive) and MDA-MB-231 (mesothelin-negative). Results: Both 99mTc-A1 and 99mTc-C6 bound mesothelin with high affinity in vitro, with 99mTc-A1 affinity being 2.4-fold higher than that of 99mTc-C6 (dissociation constant, 43.9 ± 4.0 vs. 107 ± 16 nM, P < 0.05). 99mTc-A1 and 99mTc-C6 remained stable in vivo in murine blood (>80% at 2 h) and ex vivo in human blood (>90% at 6 h). In vivo 99mTc-A1 uptake (percentage injected dose) in HCC70 tumors was 5-fold higher than in MDA-MB-231 tumors and 1.5-fold higher than that of 99mTc-C6 (2.34% ± 0.36% vs. 0.48% ± 0.18% and 1.56% ± 0.43%, respectively, P < 0.01) and resulted in elevated tumor-to-background ratios. In vivo competition experiments demonstrated the specificity of 99mTc-A1 uptake in HCC70 tumors. Conclusion: Mesothelin-positive tumors were successfully identified by SPECT using 99mTc-A1 and 99mTc-C6. Considering its superior characteristics, 99mTc-A1 was selected as the most suitable tool for further clinical translation.


Subject(s)
GPI-Linked Proteins/metabolism , Tomography, Emission-Computed, Single-Photon/methods , Triple Negative Breast Neoplasms/diagnostic imaging , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Humans , Hydrophobic and Hydrophilic Interactions , Isotope Labeling , Ligands , Mesothelin , Mice , Organotechnetium Compounds/blood , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/pharmacokinetics , Tissue Distribution , Triple Negative Breast Neoplasms/pathology
7.
Nat Commun ; 8(1): 1967, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29213077

ABSTRACT

Antibodies have enormous therapeutic and biotechnology potential. G protein-coupled receptors (GPCRs), the main targets in drug development, are of major interest in antibody development programs. Metabotropic glutamate receptors are dimeric GPCRs that can control synaptic activity in a multitude of ways. Here we identify llama nanobodies that specifically recognize mGlu2 receptors, among the eight subtypes of mGluR subunits. Among these nanobodies, DN10 and 13 are positive allosteric modulators (PAM) on homodimeric mGlu2, while DN10 displays also a significant partial agonist activity. DN10 and DN13 have no effect on mGlu2-3 and mGlu2-4 heterodimers. These PAMs enhance the inhibitory action of the orthosteric mGlu2/mGlu3 agonist, DCG-IV, at mossy fiber terminals in the CA3 region of hippocampal slices. DN13 also impairs contextual fear memory when injected in the CA3 region of hippocampal region. These data highlight the potential of developing antibodies with allosteric actions on GPCRs to better define their roles in vivo.


Subject(s)
Fear/physiology , Hippocampus/metabolism , Receptors, Metabotropic Glutamate/metabolism , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Binding Sites , Camelids, New World , Cyclic AMP/metabolism , Cyclopropanes , Glutamic Acid/blood , Glutamic Acid/metabolism , Glycine/analogs & derivatives , HEK293 Cells , Hippocampus/drug effects , Humans , Inositol Phosphates/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Neurons/physiology , Receptors, Opioid
8.
Sci Rep ; 6: 21834, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26912069

ABSTRACT

The detection of tumours in an early phase of tumour development in combination with the knowledge of expression of tumour markers such as epidermal growth factor receptor (EGFR) is an important prerequisite for clinical decisions. In this study we applied the anti-EGFR nanobody (99m)Tc-D10 for visualizing small tumour lesions with volumes below 100 mm(3) by targeting EGFR in orthotopic human mammary MDA-MB-468 and MDA-MB-231 and subcutaneous human epidermoid A431 carcinoma mouse models. Use of nanobody (99m)Tc-D10 of a size as small as 15.5 kDa enables detection of tumours by single photon emission computed tomography (SPECT) imaging already 45 min post intravenous administration with high tumour uptake (>3% ID/g) in small MDA-MB-468 and A431 tumours, with tumour volumes of 52.5 mm(3) ± 21.2 and 26.6 mm(3) ± 16.7, respectively. Fast blood clearance with a serum half-life of 4.9 min resulted in high in vivo contrast and ex vivo tumour to blood and tissue ratios. In contrast, no accumulation of (99m)Tc-D10 in MDA-MB-231 tumours characterized by a very low expression of EGFR was observed. Here we present specific and high contrast in vivo visualization of small human tumours overexpressing EGFR by preclinical multi-pinhole SPECT shortly after administration of anti-EGFR nanobody (99m)Tc-D10.


Subject(s)
ErbB Receptors/immunology , Organotechnetium Compounds/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology , Tomography, Emission-Computed, Single-Photon , Animals , Blotting, Western , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cetuximab/chemistry , Cetuximab/metabolism , Disease Models, Animal , ErbB Receptors/metabolism , Female , Flow Cytometry , Fluorescent Dyes/chemistry , Half-Life , Humans , Isotope Labeling , Mice , Mice, Nude , Microscopy, Fluorescence , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/immunology , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Technetium/chemistry , Tissue Distribution , Tomography, X-Ray Computed
9.
J Acquir Immune Defic Syndr ; 71(3): 237-45, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26413851

ABSTRACT

BACKGROUND: Highly potent broadly neutralizing monoclonal antibodies (bNAbs) have been obtained from individuals infected by HIV-1 group M variants. We analyzed the cross-group neutralization potency of these bNAbs toward non-M primary isolates (PI). MATERIAL AND METHODS: The sensitivity to neutralization was analyzed in a neutralization assay using TZM-bl cells. Twenty-three bNAbs were used, including reagents targeting the CD4-binding site, the N160 glycan-V1/V2 site, the N332 glycan-V3 site, the membrane proximal external region of gp41, and complex epitopes spanning both env subunits. Two bispecific antibodies that combine the inhibitory activity of an anti-CD4 with that of PG9 or PG16 bNAbs were included in the study (PG9-iMab and PG16-iMab). RESULTS: Cross-group neutralization was observed only with the bNAbs targeting the N160 glycan-V1/V2 site. Four group O PIs, 1 group N PI, and the group P PI were neutralized by PG9 and/or PG16 or PGT145 at low concentrations (0.04-9.39 µg/mL). None of the non-M PIs was neutralized by the bNAbs targeting other regions at the highest concentration tested, except 10E8 that neutralized weakly 2 group N PIs and 35O22 that neutralized 1 group O PI. The bispecific bNAbs neutralized very efficiently all the non-M PIs with IC50 below 1 µg/mL, except 2 group O strains. CONCLUSION: The N160 glycan-V1/V2 site is the most conserved neutralizing site within the 4 groups of HIV-1. This makes it an interesting target for the development of HIV vaccine immunogens. The corresponding bNAbs may be useful for immunotherapeutic strategies in patients infected by non-M variants.


Subject(s)
Antibodies, Neutralizing/immunology , Conserved Sequence , Epitopes/genetics , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Amino Acid Sequence , Antibodies, Monoclonal/immunology , CD4 Antigens , CD4 Lymphocyte Count , Chlorofluorocarbons, Methane , Epitopes/immunology , Gene Expression Regulation, Viral/physiology , HIV Envelope Protein gp120/immunology , HIV Infections/virology , HIV-1/classification , HIV-1/genetics , Humans , Molecular Sequence Data , Phylogeny , Recombinant Proteins , env Gene Products, Human Immunodeficiency Virus/immunology
10.
J Biomed Nanotechnol ; 11(7): 1201-12, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26307843

ABSTRACT

Mesothelin, a cancer biomarker overexpressed in tumors of epithelial origin, is a target for nanotechnology-based diagnostic, therapeutic, and prognostic applications. The currently available anti-mesothelin antibodies present limitations, including low penetration due to large size and/or lack of in vivo stability. Single domain antibodies (sdAbs) or nanobodies (Nbs) provide powerful solutions to these specific problems. We generated a phage-display library of Nbs that were amplified from B cells of a llama that was immunized with human recombinant mesothelin. Two nanobodies (Nb A1 and Nb C6) were selected on the basis of affinity (K(D) = 15 and 30 nM, respectively). Nb A1 was further modified by adding either a cysteine to permit maleimide-based bioconjugations or a sequence for the site-specific metabolic addition of a biotin in vivo. Both systems of conjugation (thiol-maleimide and streptavidin/biotin) were used to characterize and validate Nb A1 and to functionalize nanoparticles. We showed that anti-mesothelin Nb A1 could detect native and denatured mesothelin in various diagnostic applications, including flow cytometry, western blotting, immunofluorescence, and optical imaging. In conclusion, anti-mesothelin Nbs are novel, cost-effective, small, and single domain reagents with high affinity and specificity for the tumor-associated antigen mesothelin, which can be simply bioengineered for attachment to nanoparticles or modified surfaces using multiple bioconjugation strategies. These anti-mesothelin Nbs can be useful in both conventional and nanotechnology-based diagnostic, therapeutic and prognostic biomedical applications.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , GPI-Linked Proteins/immunology , Nanoparticles/therapeutic use , Subcellular Fractions/immunology , Antibodies, Monoclonal/genetics , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , HeLa Cells , Humans , Mesothelin , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Protein Engineering/methods , Subcellular Fractions/pathology
11.
Anal Biochem ; 478: 26-32, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25766579

ABSTRACT

Compact single-domain antibodies (sdAbs) are nearly 13 times smaller than full-size monoclonal antibodies (mAbs) and have a number of advantages for biotechnological applications, such as small size, high specificity, solubility, stability, and great refolding capacity. Carcinoembryonic antigen (CEA) is a tumor-associated glycoprotein expressed in a variety of cancers. Detection of CEA on the tumor cell surface may be carried out using anti-CEA antibodies and conventional fluorescent dyes. Semiconductor quantum dots (QDs) are brighter and more photostable than organic dyes; they provide the possibility for labeling of different recognition molecules with QDs of different colors but excitable with the same wavelength of excitation. In this study, the abilities for specific detection of CEA expressed by tumor cells with anti-CEA sdAbs biotinylated in vitro and in vivo, as well as with anti-CEA mAbs biotinylated in vitro, were compared using flow cytometry and the conjugates of streptavidin with QDs (SA-QDs). The results demonstrated that either in vitro or in vivo biotinylated anti-CEA sdAbs are more sensitive for cell staining compared to biotinylated anti-CEA mAbs. The data also show that simultaneous use of biotinylated sdAbs with highly fluorescent SA-QDs can considerably improve the sensitivity of detection of CEA on tumor cell surfaces.


Subject(s)
Carcinoembryonic Antigen/analysis , Quantum Dots/chemistry , Single-Domain Antibodies/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Biotinylation , Cell Line , Flow Cytometry/methods , Humans , Mice , Molecular Sequence Data
12.
ACS Nano ; 9(2): 1388-99, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25603171

ABSTRACT

The epidermal growth factor receptor (EGFR) is a cell-surface receptor with a single transmembrane domain and tyrosine kinase activity carried by the intracellular domain. This receptor is one of the four members of the ErbB family including ErbB2, ErbB3, and ErbB4. Ligand binding, like EGF binding, induces a conformational rearrangement of the receptor and induces a homo/hetero dimerization essentially with ErbB family receptors that leads to the phosphorylation of the kinase domain, triggering a signaling cascade. EGFR can also form inactive dimers in a ligand-independent way through interactions between cytoplasmic domains. To date, the conformation of EGFR extracellular domain engaged in these inactive dimers remains unclear. In this study, we describe the successful selection and characterization of llama anti-EGFR nanobodies and their use as innovative conformational sensors. We isolated three different specific anti-EGFR clones binding to three distinct epitopes. Interestingly, the binding of all three nanobodies was found highly sensitive to ligand stimulation. Two nanobodies, D10 and E10, can only bind the ligand-free EGFR conformation characterized by an intramolecular tether between domains II and IV, whereas nanobody G10 binds both ligand-free and ligand activated EGFR, with an 8-fold higher affinity for the extended conformation in the presence of ligand. Here we took advantage of these conformational probes to reveal the existence of tethered EGFR in EGFR/ErbB2 predimers. These biosensors represent important tools allowing the determination of EGFR conformations and should help the design of relevant inhibitors.


Subject(s)
Biosensing Techniques , ErbB Receptors/chemistry , ErbB Receptors/immunology , Protein Multimerization , Receptor, ErbB-2/chemistry , Single-Domain Antibodies/immunology , Animals , Antibody Specificity , Binding Sites , Camelids, New World , Cell Line , Epitopes/immunology , Humans , Mice , Protein Structure, Quaternary
13.
Antibodies (Basel) ; 5(1)2015 Dec 26.
Article in English | MEDLINE | ID: mdl-31557983

ABSTRACT

As evidenced by the recent approvals of Removab (EU, Trion Pharma) in 2009 and of Blincyto (US, Amgen) in 2014, the high potential of bispecific antibodies in the field of immuno-oncology is eliciting a renewed interest from pharmaceutical companies. Supported by rapid advances in antibody engineering and the development of several technological platforms such as Triomab or bispecific T cell engagers (BiTEs), the "bispecifics" market has increased significantly over the past decade and may occupy a pivotal space in the future. Over 30 bispecific molecules are currently in different stages of clinical trials and more than 70 in preclinical phase. This review focuses on the clinical potential of bispecific antibodies as immune effector cell engagers in the onco-immunotherapy field. We summarize current strategies targeting various immune cells and their clinical interests. Furthermore, perspectives of bispecific antibodies in future clinical developments are addressed.

14.
PLoS One ; 9(12): e113729, 2014.
Article in English | MEDLINE | ID: mdl-25436999

ABSTRACT

The targeting of HIV-1 using antibodies is of high interest as molecular tools to better understand the biology of the virus or as a first step toward the design of new inhibitors targeting critical viral intracellular proteins. Small and highly stable llama-derived single-domain antibodies can often be functionally expressed as intracellular antibodies in the cytoplasm of eukaryotic cells. Using a selection method based on the Sos Recruitment System, a cytoplasmic yeast two-hybrid approach, we have isolated single-domain antibodies able to bind HIV-1 Vpr and Capside proteins in the yeast cytoplasm. One anti-Vpr single domain antibody was able to bind the HIV-1 regulatory Vpr protein in the cytoplasm of eukaryotic cells, leading to its delocalization from the nucleus to the cytoplasm. To our knowledge, this is the first description of a functional single-domain intrabody targeting HIV-1 Vpr, isolated using an in vivo cytoplasmic selection method that alleviates some limitations of the conventional yeast two-hybrid system.


Subject(s)
Cytoplasm/metabolism , Single-Domain Antibodies/metabolism , Two-Hybrid System Techniques , vpr Gene Products, Human Immunodeficiency Virus/immunology , Cell Nucleus/metabolism , HIV-1/metabolism , HeLa Cells , Humans , Protein Binding , Saccharomyces cerevisiae/metabolism , Transfection
15.
Oncotarget ; 5(14): 5304-19, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-24979648

ABSTRACT

Trastuzumab is established as treatment of HER2high metastatic breast cancers but many limitations impair its efficacy. Here, we report the design of a Fab-like bispecific antibody (HER2bsFab) that displays a moderate affinity for HER2 and a unique, specific and high affinity for FcγRIII. In vitro characterization showed that ADCC was the major mechanism of action of HER2bsFab as no significant HER2-driven effect was observed. HER2bsFab mediated ADCC at picomolar concentration against HER2high, HER2low as well as trastuzumab-refractive cell lines. In vivo HER2bsFab potently inhibited HER2high tumor growth by recruitment of mouse FcγRIII and IV-positive resident effector cells and more importantly, exhibited a net superiority over trastuzumab at inhibiting HER2low tumor growth. Moreover, FcγRIIIA-engagement by HER2bsFab was independent of V/F158 polymorphism and induced a stronger NK cells activation in response to target cell recognition. Thus, taking advantage of its epitope specificity and affinity for HER2 and FcγRIIIA, HER2bsFab exhibits potent anti-tumor activity against HER2low tumors while evading most of trastuzumab Fc-linked limitations thereby potentially enlarging the number of patients eligible for breast cancer immunotherapy.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/therapy , Receptor, ErbB-2/immunology , Receptors, IgG/immunology , Animals , Antibodies, Bispecific/immunology , Breast Neoplasms/enzymology , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Humans , Jurkat Cells , Killer Cells, Natural/immunology , Mice , Mice, Nude , Random Allocation , Receptor, ErbB-2/biosynthesis , Trastuzumab , Xenograft Model Antitumor Assays
16.
Nanomedicine ; 10(8): 1701-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24941465

ABSTRACT

An ideal multiphoton fluorescent nanoprobe should combine a nanocrystal with the largest possible two-photon absorption cross section (TPACS) and the smallest highly specific recognition molecules bound in an oriented manner. CdSe/ZnS quantum dots (QDs) conjugated to 13-kDa single-domain antibodies (sdAbs) derived from camelid IgG or streptavidin have been used as efficient two-photon excitation (TPE) probes for carcinoembryonic antigen (CEA) imaging on normal human appendix and colon carcinoma tissue. The TPACS for some conjugates was higher than 49,000 GM (Goeppert-Mayer units), considerably exceeding that of organic dyes being close to the theoretical value of 50,000 GM calculated for CdSe QDs. The ratio of sdAb-QD emission to the autofluorescence for 800 nm TPE was 40 times higher than that for 457.9 nm one-photon excitation. TPE ensures a clear discrimination of CEA-overexpressing tumor areas from normal tissue. Oriented sdAb-QD conjugates are bright specific labels for detecting low concentrations of antigens using multiphoton microscopy. FROM THE CLINICAL EDITOR: This study demonstrates carcinoembryonic antigen imaging on normal human appendix and colon carcinoma tissue utilizing CdSe/ZnS quantum dots conjugated to streptavidin or to 13-kDa single-domain antibodies as efficient two-photon excitation probes.


Subject(s)
Diagnostic Imaging/methods , Quantum Dots , Single-Domain Antibodies/chemistry , Animals , Biomarkers, Tumor , In Vitro Techniques
17.
ACS Nano ; 8(6): 5682-95, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24873349

ABSTRACT

Despite the widespread availability of immunohistochemical and other methodologies for screening and early detection of lung and breast cancer biomarkers, diagnosis of the early stage of cancers can be difficult and prone to error. The identification and validation of early biomarkers specific to lung and breast cancers, which would permit the development of more sensitive methods for detection of early disease onset, is urgently needed. In this paper, ultra-small and bright nanoprobes based on quantum dots (QDs) conjugated to single domain anti-HER2 (human epidermal growth factor receptor 2) antibodies (sdAbs) were applied for immunolabeling of breast and lung cancer cell lines, and their performance was compared to that of anti-HER2 monoclonal antibodies conjugated to conventional organic dyes Alexa Fluor 488 and Alexa Fluor 568. The sdAbs-QD conjugates achieved superior staining in a panel of lung cancer cell lines with differential HER2 expression. This shows their outstanding potential for the development of more sensitive assays for early detection of cancer biomarkers.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Lung Neoplasms/metabolism , Quantum Dots , Receptor, ErbB-2/metabolism , Antibodies/chemistry , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Coculture Techniques , Flow Cytometry , Fluorescent Dyes/chemistry , Humans , Immunohistochemistry , Macrophages/metabolism , Microscopy, Confocal
18.
Mol Cell Proteomics ; 13(2): 653-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24361863

ABSTRACT

Phage display is a well-established procedure to isolate binders against a wide variety of antigens that can be performed on purified antigens, but also on intact cells. As selection steps are performed in vitro, it is possible to focus the outcome of the selection on relevant epitopes by performing some additional steps, such as depletion or competitive elutions. However in practice, the efficiency of these steps is often limited and can lead to inconsistent results. We have designed a new selection method named masked selection, based on the blockade of unwanted epitopes to favor the targeting of relevant ones. We demonstrate the efficiency and flexibility of this method by selecting single-domain antibodies against a specific portion of a fusion protein, by selecting binders against several members of the seven transmembrane receptor family using transfected HEK cells, or by selecting binders against unknown breast cancer markers not expressed on normal samples. The relevance of this approach for antibody-based therapies was further validated by the identification of four of these markers, Epithelial cell adhesion molecule, Transferrin receptor 1, Metastasis cell adhesion molecule, and Sushi containing domain 2, using immunoprecipitation and mass spectrometry. This new phage display strategy can be applied to any type of antibody fragments or alternative scaffolds, and is especially suited for the rapid discovery and identification of cell surface markers.


Subject(s)
Cell Surface Display Techniques , Epitope Mapping/methods , Epitopes/metabolism , Proteins/metabolism , Antigens/immunology , Antigens/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Immunoprecipitation , Jurkat Cells , MCF-7 Cells , Protein Binding , Proteins/immunology , Proteomics/methods
19.
J Virol ; 87(18): 10173-81, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23843638

ABSTRACT

The conserved HIV-1 site of coreceptor binding is protected from antibody-directed neutralization by conformational and steric restrictions. While inaccessible to most human antibodies, the coreceptor site has been shown to be accessed by antibody fragments. In this study, we used X-ray crystallography, surface plasmon resonance, and pseudovirus neutralization to characterize the gp120-envelope glycoprotein recognition and HIV-1 neutralization of a heavy chain-only llama antibody, named JM4. We describe full-length IgG2b and IgG3 versions of JM4 that target the coreceptor-binding site and potently neutralize over 95% of circulating HIV-1 isolates. Contrary to established trends that show improved access to the coreceptor-binding region by smaller antibody fragments, the single-domain (VHH) version of JM4 neutralized less well than the full-length IgG2b version of JM4. The crystal structure at 2.1-Å resolution of VHH JM4 bound to HIV-1 YU2 gp120 stabilized in the CD4-bound state by the CD4-mimetic miniprotein, M48U1, revealed a JM4 epitope that combined regions of coreceptor recognition (including the gp120 bridging sheet, V3 loop, and ß19 strand) with gp120 structural elements involved in recognition of CD4 such as the CD4-binding loop. The structure of JM4 with gp120 thus defines a novel CD4-induced site of vulnerability involving elements of both coreceptor- and CD4-binding sites. The potently neutralizing JM4 IgG2b antibody that targets this newly defined site of vulnerability adds to the expanding repertoire of broadly neutralizing antibodies that effectively neutralize HIV-1 and thereby potentially provides a new template for vaccine development and target for HIV-1 therapy.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Immunoglobulin G/immunology , Animals , Antibodies, Neutralizing/chemistry , Binding Sites/immunology , Camelids, New World , Crystallography, X-Ray , HIV Antibodies/chemistry , HIV Envelope Protein gp120/chemistry , HIV-1/chemistry , Immunoglobulin G/chemistry , Neutralization Tests , Surface Plasmon Resonance
20.
Mol Cancer Ther ; 12(8): 1481-91, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23757164

ABSTRACT

Antibody-dependent cell-mediated cytotoxicity, one of the most prominent modes of action of antitumor antibodies, suffers from important limitations due to the need for optimal interactions with Fcγ receptors. In this work, we report the design of a new bispecific antibody format, compact and linker-free, based on the use of llama single-domain antibodies that are capable of circumventing most of these limitations. This bispecific antibody format was created by fusing single-domain antibodies directed against the carcinoembryonic antigen and the activating FcγRIIIa receptor to human Cκ and CH1 immunoglobulin G1 domains, acting as a natural dimerization motif. In vitro and in vivo characterization of these Fab-like bispecific molecules revealed favorable features for further development as a therapeutic molecule. They are easy to produce in Escherichia coli, very stable, and elicit potent lysis of tumor cells by human natural killer cells at picomolar concentrations. Unlike conventional antibodies, they do not engage inhibitory FcγRIIb receptor, do not compete with serum immunoglobulins G for receptor binding, and their cytotoxic activity is independent of Fc glycosylation and FcγRIIIa polymorphism. As opposed to anti-CD3 bispecific antitumor antibodies, they do not engage regulatory T cells as these latter cells do not express FcγRIII. Studies in nonobese diabetic/severe combined immunodeficient gamma mice xenografted with carcinoembryonic antigen-positive tumor cells showed that Fab-like bispecific molecules in the presence of human peripheral blood mononuclear cells significantly slow down tumor growth. This new compact, linker-free bispecific antibody format offers a promising approach for optimizing antibody-based therapies.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents/pharmacology , Receptors, IgG/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/chemistry , Carcinoembryonic Antigen/immunology , Cell Line, Tumor , Cytokines/biosynthesis , Drug Stability , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Killer Cells, Natural/immunology , Mice , Neoplasms/drug therapy , Neoplasms/immunology , Polymorphism, Genetic , Protein Binding/immunology , Protein Stability , Receptors, IgG/genetics , Receptors, IgG/metabolism , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Tissue Distribution , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...