Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Eixos temáticos
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Front Oncol ; 12: 1017612, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263202

RESUMO

DNA-encoded delivery and in vivo expression of antibody therapeutics presents an innovative alternative to conventional protein production and administration, including for cancer treatment. To support clinical translation, we evaluated this approach in 18 40-45 kg sheep, using a clinical-matched intramuscular electroporation (IM EP) and hyaluronidase-plasmid DNA (pDNA) coformulation setup. Two cohorts of eight sheep received either 1 or 4 mg pDNA encoding an ovine anti-cancer embryonic antigen (CEA) monoclonal antibody (mAb; OVAC). Results showed a dose-response with average maximum serum concentrations of respectively 0.3 and 0.7 µg/ml OVAC, 4-6 weeks after IM EP. OVAC was detected in all 16 sheep throughout the 6-week follow-up, and no anti-OVAC antibodies were observed. Another, more exploratory, cohort of two sheep received a 12 mg pOVAC dose. Both animals displayed a similar dose-dependent mAb increase and expression profile in the first two weeks. However, in one animal, an anti-OVAC antibody response led to loss of mAb detection four weeks after IM EP. In the other animal, no anti-drug antibodies were observed. Serum OVAC concentrations peaked at 4.9 µg/ml 6 weeks after IM EP, after which levels gradually decreased but remained detectable around 0.2 to 0.3 µg/ml throughout a 13-month follow-up. In conclusion, using a delivery protocol that is currently employed in clinical Phase 1 studies of DNA-based antibodies, we achieved robust and prolonged in vivo production of anti-cancer DNA-encoded antibody therapeutics in sheep. The learnings from this large-animal model regarding the impact of pDNA dose and host immune response on the expressed mAb pharmacokinetics can contribute to advancing clinical translation.

2.
Hum Gene Ther ; 32(19-20): 1200-1209, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34482757

RESUMO

DNA-encoded delivery of antibodies presents a labor- and cost-effective alternative to conventional antibody therapeutics. This study aims to improve the potency and safety of this approach by evaluating various plasmid backbones and expression cassettes. In vitro, antibody levels consistently improved with decreasing sizes of backbone, ranging from conventional to minimal. In vivo, following intramuscular electrotransfer in mice, the correlation was less consistent. While the largest conventional plasmid (10.2 kb) gave the lowest monoclonal antibody (mAb) levels, a regular conventional plasmid (8.6 kb) demonstrated similar levels as a minimal Nanoplasmid (6.8 kb). A reduction in size beyond a standard conventional backbone thus did not improve mAb levels in vivo. Cassette modifications, such as swapping antibody chain order or use of two versus a single encoding plasmid, significantly increased antibody expression in vitro, but failed to translate in vivo. Conversely, a significant improvement in vivo but not in vitro was found with a set of muscle-specific promoters, of which a newly engineered variant gave roughly 1.5- to 2-fold higher plasma antibody concentrations than the ubiquitous CAG promoter. In conclusion, despite the limited translation between in vitro and in vivo, we identified various clinically relevant improvements to our DNA-based antibody platform, both in potency and biosafety.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Animais , DNA , Eletroporação , Camundongos , Plasmídeos/genética , Regiões Promotoras Genéticas
3.
Cancer Gene Ther ; 28(7-8): 828-838, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-32733055

RESUMO

Nanobodies present an appealing class of potential cancer therapeutics. The current study explores the in vivo expression of these molecules through DNA-encoded delivery. We hypothesized that this approach could address the rapid clearance of Nanobodies and, through half-life modulation, increase the produced levels in circulation. We therefore evaluated pharmacokinetics and efficacy of variants of an anti-death receptor 5 Nanobody (NbDR5), either monovalent or multivalent with half-life extension properties, after DNA-based administration. Intramuscular electrotransfer of a monovalent NbDR5-encoding plasmid (pNbDR5) did not result in detectable plasma levels in BALB/c mice. A tetravalent NbDR5-encoding plasmid (pNbDR54) provided peak concentrations of 54 ng/mL, which remained above 24 ng/mL during a 12-week follow-up. DNA-based delivery of these Nanobody formats fused to a Nanobody binding to serum albumin (NbSA), pNbDR5-NbSA and pNbDR54-NbSA, resulted in significantly higher plasma levels, with peak titers of 5.2 and 7.7 µg/mL, respectively. In an athymic-nude mice COLO 205 colon-cancer model, a quadrupled intramuscular DNA dose led to peak plasma levels of 270 ng/mL for pNbDR54 and 38 µg/mL for pNbDR54-NbSA. Potent anti-tumor responses were only observed for pNbDR54, following either intramuscular or intratumoral delivery. Despite comparable in vitro activity and superior plasma exposure, NbDR54-NbSA was less effective than NbDR54 in vivo, regardless of whether delivered as DNA or protein. Overall, DNA-based Nanobody delivery resulted in more potent and durable anti-tumor responses than protein-based Nanobody delivery. In conclusion, this study demonstrates pre-clinical proof of concept for DNA-based Nanobodies in oncology and highlights the improved outcome over conventional protein administration.


Assuntos
DNA/genética , Proteínas/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Anticorpos de Domínio Único/genética , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus
4.
Hum Gene Ther ; 30(11): 1431-1443, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31382777

RESUMO

Clinical translation of DNA-based administration of monoclonal antibodies (mAbs) is uncertain due to lack of large animal data. To bridge the clinical gap, we evaluated a panel of novel plasmid DNA (pDNA)-encoded mAbs in 40-70 kg sheep with a clinical intramuscular electroporation protocol. Injection of 4.8 mg of pDNA, encoding ovine anti-human CEA mAb (OVAC), led to peak plasma mAb titers of 300 ng/mL. OVAC remained detectable for 3 months and was boosted by a second pOVAC administration. Hyaluronidase muscle pretreatment increased OVAC concentrations up to 10-fold. These higher plasma titers, however, led to anti-drug antibodies (ADAs) toward the OVAC variable regions, resulting in loss of mAb detection and of adequate redosing. Transient immune suppression avoided ADA formation, with OVAC peaking at 3.5 µg/mL and remaining detectable for 11 months after pOVAC injection. DNA-based delivery of ovine anti-human EGFR mAb (OVAE), identical to OVAC except for the variable regions, preceded by hyaluronidase, allowed for at least three consecutive administrations in an immune-competent sheep, without ADA response. When tripling the pOVAE dose to 15 mg, transient ADAs of limited impact were observed; plasma OVAE peaked at 2.6 µg/mL and was detected up to 7 months. DNA-based anti-HER2 trastuzumab in sheep gave no detectable mAb concentrations despite previous validation in mice, highlighting the limitations of relying on small-rodent data only. In conclusion, our results highlight the potential and caveats of clinical DNA-based antibody therapy, can expedite preclinical and clinical development, and benefit the field of gene transfer as a whole.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Terapia Genética , Pesquisa Translacional Biomédica , Animais , Anticorpos Monoclonais/farmacocinética , Feminino , Técnicas de Transferência de Genes , Terapia de Imunossupressão , Camundongos , Ovinos , Trastuzumab/sangue , Trastuzumab/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa