Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Front Immunol ; 13: 838406, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35651607

RESUMEN

CD38 is a target for immunotherapy of multiple myeloma. Llama-derived CD38-specific nanobodies allow easy reformatting into mono-, bi- and multispecific proteins. To evaluate the utility of nanobodies for constructing CD38-specific nanobody-based killer cell engagers (nano-BiKEs), we generated half-life extended nano-BiKEs (HLE-nano-BiKEs) by fusing a CD38-specific nanobody to a CD16-specific nanobody for binding to the Fc-receptor on NK cells and further to an albumin-specific nanobody to extend the half-life in vivo. HLE-nano-BiKEs targeting three different epitopes (E1, E2, E3) of CD38 were expressed in transiently transfected HEK-6E cells. We verified specific and simultaneous binding to CD38 on myeloma cells, CD16 on NK cells, and to albumin. We tested the capacity of these HLE-nano-BiKEs to mediate cytotoxicity against CD38-expressing multiple myeloma cell lines and primary myeloma cells from human bone marrow biopsies in bioluminescence and flowcytometry assays with NK92 cells as effector cells. The results revealed specific time- and dose-dependent cytolysis of CD38+ myeloma cell lines and effective depletion of CD38-expressing multiple myeloma cells from primary human bone marrow samples. Our results demonstrate the efficacy of CD38-specific HLE-nano-BiKEs in vitro and ex vivo, warranting further preclinical evaluation in vivo of their therapeutic potential for the treatment of multiple myeloma.


Asunto(s)
Mieloma Múltiple , Anticuerpos de Dominio Único , ADP-Ribosil Ciclasa 1/metabolismo , Albúminas/uso terapéutico , Línea Celular Tumoral , Semivida , Humanos , Mieloma Múltiple/tratamiento farmacológico , Anticuerpos de Dominio Único/farmacología , Anticuerpos de Dominio Único/uso terapéutico
2.
Theranostics ; 10(6): 2645-2658, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194826

RESUMEN

Rationale: CD38 is a target for the therapy of multiple myeloma (MM) with monoclonal antibodies such as daratumumab and isatuximab. Since MM patients exhibit a high rate of relapse, the development of new biologics targeting alternative CD38 epitopes is desirable. The discovery of single-domain antibodies (nanobodies) has opened the way for a new generation of antitumor therapeutics. We report the generation of nanobody-based humanized IgG1 heavy chain antibodies (hcAbs) with a high specificity and affinity that recognize three different and non-overlapping epitopes of CD38 and compare their cytotoxicity against CD38-expressing hematological cancer cells in vitro, ex vivo and in vivo. Methods: We generated three humanized hcAbs (WF211-hcAb, MU1067-hcAb, JK36-hcAb) that recognize three different non-overlapping epitopes (E1, E2, E3) of CD38 by fusion of llama-derived nanobodies to the hinge- and Fc-domains of human IgG1. WF211-hcAb shares the binding epitope E1 with daratumumab. We compared the capacity of these CD38-specific hcAbs and daratumumab to induce CDC and ADCC in CD38-expressing tumor cell lines in vitro and in patient MM cells ex vivo as well as effects on xenograft tumor growth and survival in vivo. Results: CD38-specific heavy chain antibodies (WF211-hcAb, MU1067-hcAb, JK36-hcAb) potently induced antibody-dependent cellular cytotoxicity (ADCC) in CD38-expressing tumor cell lines and in primary patient MM cells, but only little if any complement-dependent cytotoxicity (CDC). In vivo, CD38-specific heavy chain antibodies significantly reduced the growth of systemic lymphomas and prolonged survival of tumor bearing SCID mice. Conclusions: CD38-specific nanobody-based humanized IgG1 heavy chain antibodies mediate cytotoxicity against CD38-expressing hematological cancer cells in vitro, ex vivo and in vivo. These promising results of our study indicate that CD38-specific hcAbs warrant further clinical development as therapeutics for multiple myeloma and other hematological malignancies.


Asunto(s)
ADP-Ribosil Ciclasa 1/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Cadenas Pesadas de Inmunoglobulina/uso terapéutico , Glicoproteínas de Membrana/inmunología , Mieloma Múltiple/tratamiento farmacológico , Anticuerpos de Dominio Único/uso terapéutico , Anciano , Animales , Línea Celular Tumoral , Epítopos/inmunología , Femenino , Humanos , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad
3.
Mol Cancer Ther ; 18(4): 823-833, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30824613

RESUMEN

Epidermal growth factor receptor (EGFR) ectodomain variants mediating primary resistance or secondary treatment failure in cancer patients treated with cetuximab or panitumumab support the need for more resistance-preventive or personalized ways of targeting this essential pathway. Here, we tested the hypothesis that the EGFR nanobody 7D12 fused to an IgG1 Fc portion (7D12-hcAb) would overcome EGFR ectodomain-mediated resistance because it targets a very small binding epitope within domain III of EGFR. Indeed, we found that 7D12-hcAb bound and inhibited all tested cell lines expressing common resistance-mediating EGFR ectodomain variants. Moreover, we assessed receptor functionality and binding properties in synthetic mutants of the 7D12-hcAb epitope to model resistance to 7D12-hcAb. Because the 7D12-hcAb epitope almost completely overlaps with the EGF-binding site, only position R377 could be mutated without simultaneous loss of receptor functionality, suggesting a low risk of developing secondary resistance toward 7D12-hcAb. Our binding data indicated that if 7D12-hcAb resistance mutations occurred in position R377, which is located within the cetuximab and panitumumab epitope, cells expressing these receptor variants would retain sensitivity to these antibodies. However, 7D12-hcAb was equally ineffective as cetuximab in killing cells expressing the cetuximab/panitumumab-resistant aberrantly N-glycosylated EGFR R521K variant. Yet, this resistance could be overcome by introducing mutations into the Fc portion of 7D12-hcAb, which enhanced immune effector functions and thereby allowed killing of cells expressing this variant. Taken together, our data demonstrate a broad range of activity of 7D12-hcAb across cells expressing different EGFR variants involved in primary and secondary EGFR antibody resistance.


Asunto(s)
Cetuximab/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Panitumumab/farmacología , Dominios Proteicos/genética , Anticuerpos de Dominio Único/farmacología , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cetuximab/inmunología , Cetuximab/uso terapéutico , Epítopos/química , Epítopos/inmunología , Receptores ErbB/química , Receptores ErbB/genética , Receptores ErbB/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/química , Inmunoglobulina G/genética , Mutación , Panitumumab/inmunología , Panitumumab/uso terapéutico , Polimorfismo de Nucleótido Simple/genética , Dominios Proteicos/inmunología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Transducción Genética
4.
Front Immunol ; 9: 2553, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30524421

RESUMEN

CD38 is overexpressed by multiple myeloma cells and has emerged as a target for therapeutic antibodies. Nanobodies are soluble single domain antibody fragments derived from the VHH variable domain of heavy chain antibodies naturally occurring in camelids. We previously identified distinct llama nanobodies that recognize three non-overlapping epitopes of the extracellular domain of CD38. Here, we fused these VHH domains to the hinge, CH2, and CH3 domains of human IgG1, yielding highly soluble chimeric llama/human heavy chain antibodies (hcAbs). We analyzed the capacity of these hcAbs to mediate complement-dependent cytotoxicity (CDC) to CD38-expressing human multiple myeloma and Burkitt lymphoma cell lines. Combinations of two hcAbs that recognize distinct, non-overlapping epitopes of CD38 mediated potent CDC, in contrast to the hcAb monotherapy with only weak CDC capacity. Similarly, combining daratumumab with a hcAb that recognizes a non-overlapping epitope resulted in dramatically enhanced CDC. Further, introducing the E345R HexaBody mutation into the CH3 domain strongly enhanced the CDC potency of hcAbs to CD38-expressing cells. Exploiting their high solubility, we genetically fused two distinct nanobodies into heteromeric dimers via a flexible peptide linker and then fused these nanobody dimers to the hinge, CH2 and CH3 domains of human IgG1, yielding highly soluble, biparatopic hcAbs. These biparatopic hcAbs elicited CDC toward CD38-expressing myeloma cells more effectively than daratumumab. Our results underscore the advantage of nanobodies vs. pairs of VH and VL domains for constructing bispecific antibodies. Moreover, the CD38-specific biparatopic heavy chain antibodies described here represent potential new powerful therapeutics for treatment of multiple myeloma.


Asunto(s)
ADP-Ribosil Ciclasa 1/inmunología , Anticuerpos Biespecíficos/farmacología , Antígenos de Neoplasias/inmunología , Antineoplásicos/farmacología , Epítopos de Linfocito B/inmunología , Inmunoterapia/métodos , Mieloma Múltiple/inmunología , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Camélidos del Nuevo Mundo , Línea Celular Tumoral , Proteínas del Sistema Complemento/metabolismo , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Mieloma Múltiple/terapia , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Dominio Único/genética
5.
Sci Rep ; 7(1): 14289, 2017 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-29084989

RESUMEN

The cell surface ecto-enzyme CD38 is a promising target antigen for the treatment of hematological malignancies, as illustrated by the recent approval of daratumumab for the treatment of multiple myeloma. Our aim was to evaluate the potential of CD38-specific nanobodies as novel diagnostics for hematological malignancies. We successfully identified 22 CD38-specific nanobody families using phage display technology from immunized llamas. Crossblockade analyses and in-tandem epitope binning revealed that the nanobodies recognize three different non-overlapping epitopes, with four nanobody families binding complementary to daratumumab. Three nanobody families inhibit the enzymatic activity of CD38 in vitro, while two others were found to act as enhancers. In vivo, fluorochrome-conjugated CD38 nanobodies efficiently reach CD38 expressing tumors in a rodent model within 2 hours after intravenous injection, thereby allowing for convenient same day in vivo tumor imaging. These nanobodies represent highly specific tools for modulating the enzymatic activity of CD38 and for diagnostic monitoring CD38-expressing tumors.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Glicoproteínas de Membrana/metabolismo , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/tratamiento farmacológico , Anticuerpos de Dominio Único/inmunología , ADP-Ribosil Ciclasa 1/inmunología , Animales , Camélidos del Nuevo Mundo , Línea Celular Tumoral , Técnicas de Visualización de Superficie Celular , Modelos Animales de Enfermedad , Epítopos/inmunología , Colorantes Fluorescentes , Humanos , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Desnudos , Mieloma Múltiple/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Vis Exp ; (98): e52462, 2015 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-25867711

RESUMEN

This protocol outlines the steps required to perform ex vivo validation of in vivo near-infrared fluorescence (NIRF) xenograft imaging experiments in mice using fluorophore labelled nanobodies and conventional antibodies. First we describe how to generate subcutaneous tumors in mice, using antigen-negative cell lines as negative controls and antigen-positive cells as positive controls in the same mice for intraindividual comparison. We outline how to administer intravenously near-infrared fluorophore labelled (AlexaFluor680) antigen-specific nanobodies and conventional antibodies. In vivo imaging was performed with a small-animal NIRF-Imaging system. After the in vivo imaging experiments the mice were sacrificed. We then describe how to prepare the tumors for parallel ex vivo analyses by flow cytometry and fluorescence microscopy to validate in vivo imaging results. The use of the near-infrared fluorophore labelled nanobodies allows for non-invasive same day imaging in vivo. Our protocols describe the ex vivo quantification of the specific labeling efficiency of tumor cells by flow cytometry and analysis of the distribution of the antibody constructs within the tumors by fluorescence microscopy. Using near-infrared fluorophore labelled probes allows for non-invasive, economical in vivo imaging with the unique ability to exploit the same probe without further secondary labelling for ex vivo validation experiments using flow cytometry and fluorescence microscopy.


Asunto(s)
Anticuerpos/química , Colorantes Fluorescentes/química , Xenoinjertos/patología , Linfoma/patología , Anticuerpos de Dominio Único/química , Animales , Línea Celular Tumoral , Citometría de Flujo/métodos , Colorantes Fluorescentes/análisis , Humanos , Ratones , Ratones Desnudos , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Imagen Molecular/métodos , Trasplante de Neoplasias , Reproducibilidad de los Resultados , Anticuerpos de Dominio Único/análisis , Trasplante Heterólogo
7.
Contrast Media Mol Imaging ; 10(5): 367-78, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25925493

RESUMEN

The utility of nanobodies and conventional antibodies for in vivo imaging is well known, but optimum dosing and timing schedules for one versus the other have not been established. We aimed to improve specific tumor imaging in vivo with nanobodies and conventional antibodies using near-infrared fluorescence (NIRF) imaging. We used ARTC2 expressed on lymphoma cells as a model target antigen. ARTC2-specific nanobody s+16a and conventional antibody Nika102 were labeled with NIRF-dye AF680. In vivo NIRF-imaging of ARTC2-positive and ARTC2-negative xenografts was performed over 24 h post-injection of 5, 10, 25, or 50 µg of each conjugate. Specific target-binding and tissue-penetration were verified by NIRF imaging ex vivo, flow cytometry and fluorescence microscopy. NIRF-imaging of s+16a(680) in vivo revealed a six times faster tumor accumulation than of Nika102(680). Using 50 µg of s+16a(680) increased the specific signals of ARTC2-positive tumors without increasing background signals, allowing a tumor-to-background (T/B) ratio of 12.4 ± 4.2 within 6 h post-injection. Fifty micrograms of Nika102(680) increased specific signals of ARTC2-positive tumors but also of ARTC2-negative tumors and background, thereby limiting the T/B ratio to 6.1 ± 2.0. Ten micrograms of Nika102(680) only slightly reduced specific tumor signals but dramatically reduced background signals. Ex vivo analyses confirmed a faster and deeper tumor penetration with s+16a(680). Using nanobody s+16a allowed same-day imaging with a high T/B ratio, whereas antibody Nika102 gave optimal imaging results only 24 h post injection. Nanobody s+16a required a high dose, whereas antibody Nika102 had the best T/B-ratio at a low dose. Therefore, timing and dosage should be addressed when comparing nanobodies and conventional antibodies for molecular imaging purposes.


Asunto(s)
Anticuerpos/análisis , Microscopía Fluorescente/métodos , Anticuerpos de Dominio Único/análisis , Animales , Línea Celular Tumoral , Citometría de Flujo , Humanos , Ratones , Imagen Molecular/métodos , Espectroscopía Infrarroja Corta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA