Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Theranostics ; 10(6): 2645-2658, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194826

RESUMO

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.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias Hematológicas/tratamento farmacológico , Imunoglobulina G/uso terapêutico , Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Glicoproteínas de Membrana/imunologia , Mieloma Múltiplo/tratamento farmacológico , Anticorpos de Domínio Único/uso terapêutico , Idoso , Animais , Linhagem Celular Tumoral , Epitopos/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade
2.
Cells ; 9(2)2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-32013131

RESUMO

The NAD-hydrolyzing ecto-enzyme CD38 is overexpressed by multiple myeloma and other hematological malignancies. We recently generated CD38-specific nanobodies, single immunoglobulin variable domains derived from heavy-chain antibodies naturally occurring in llamas. Nanobodies exhibit high solubility and stability, allowing easy reformatting into recombinant fusion proteins. Here we explore the utility of CD38-specific nanobodies as ligands for nanobody-based chimeric antigen receptors (Nb-CARs). We cloned retroviral expression vectors for CD38-specific Nb-CARs. The human natural killer cell line NK-92 was transduced to stably express these Nb-CARs. As target cells we used CD38-expressing as well as CRISPR/Cas9-generated CD38-deficient tumor cell lines (CA-46, LP-1, and Daudi) transduced with firefly luciferase. With these effector and target cells we established luminescence and flow-cytometry CAR-dependent cellular cytotoxicity assays (CARDCCs). Finally, the cytotoxic efficacy of Nb-CAR NK-92 cells was tested on primary patient-derived CD38-expressing multiple myeloma cells. NK-92 cells expressing CD38-specific Nb-CARs specifically lysed CD38-expressing but not CD38-deficient tumor cell lines. Moreover, the Nb-CAR-NK cells effectively depleted CD38-expressing multiple myeloma cells in primary human bone marrow samples. Our results demonstrate efficacy of Nb-CARs in vitro. The potential clinical efficacy of Nb-CARs in vivo remains to be evaluated.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Linfoma de Burkitt/metabolismo , Mieloma Múltiplo/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Anticorpos de Domínio Único/metabolismo , Medula Óssea/patologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Humanos , Células Matadoras Naturais/metabolismo , Lentivirus/metabolismo , Luciferases/metabolismo , Luminescência , Modelos Moleculares
3.
Front Immunol ; 9: 2553, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524421

RESUMO

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.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Anticorpos Biespecíficos/farmacologia , Antígenos de Neoplasias/imunologia , Antineoplásicos/farmacologia , Epitopos de Linfócito B/imunologia , Imunoterapia/métodos , Mieloma Múltiplo/imunologia , Animais , Anticorpos Biespecíficos/genética , Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos , Camelídeos Americanos , Linhagem Celular Tumoral , Proteínas do Sistema Complemento/metabolismo , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Mieloma Múltiplo/terapia , Proteínas Recombinantes de Fusão/genética , Anticorpos de Domínio Único/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...