Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Nucl Med Mol Imaging ; 43(3): 489-98, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26341366

ABSTRACT

PURPOSE: Dual-targeted therapy has been shown to be a promising treatment option in recurrent and/or refractory B-cell non-Hodgkin's lymphoma (B-NHL). We generated radioimmunoconjugates (RICs) comprising either a novel humanized anti-CD22 monoclonal antibody, huRFB4, or rituximab, and the low-energy ß-emitter (177)Lu. Both RICs were evaluated as single agents in a human Burkitt's lymphoma xenograft mouse model. To increase the therapeutic efficacy of the anti-CD22 RIC, combination therapy with unlabelled anti-CD20 rituximab was explored. METHODS: The binding activity of CHX-A″-DTPA-conjugated antibodies to target cells was analysed by flow cytometry. To assess tumour targeting of (177)Lu-labelled antibodies, in vivo biodistribution experiments were performed. For radioimmunotherapy (RIT) studies, non-obese diabetic recombination activating gene-1 (NOD-Rag1 (null) ) interleukin-2 receptor common gamma chain (IL2rγ (null) ) null mice (NRG mice) were xenografted subcutaneously with Raji Burkitt's lymphoma cells. (177)Lu-conjugated antibodies were administered at a single dose of 9.5 MBq per mouse. For dual-targeted therapy, rituximab was injected at weekly intervals (0.5 - 1.0 mg). Tumour accumulation of RICs was monitored by planar scintigraphy. RESULTS: Conjugation of CHX-A"-DTPA resulted in highly stable RICs with excellent antigen-binding properties. Biodistribution experiments revealed higher tumour uptake of the (177)Lu-labelled anti-CD22 IgG than of (177)Lu-labelled rituximab. Treatment with (177)Lu-conjugated huRFB4 resulted in increased tumour growth inhibition and significantly longer survival than treatment with (177)Lu-conjugated rituximab. The therapeutic efficacy of the anti-CD22 RIC could be markedly enhanced by combination with unlabelled rituximab. CONCLUSION: These findings suggest that dual targeting with (177)Lu-based CD22-specific RIT in combination with rituximab is a promising new treatment option for refractory B-NHL.


Subject(s)
Burkitt Lymphoma/therapy , Immunoconjugates/therapeutic use , Lutetium/chemistry , Radioimmunotherapy/methods , Rituximab/therapeutic use , Sialic Acid Binding Ig-like Lectin 2/chemistry , Animals , Burkitt Lymphoma/radiotherapy , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Immunoglobulin G/therapeutic use , Maximum Tolerated Dose , Mice , Mice, Inbred NOD , Xenograft Model Antitumor Assays
2.
Cancer Lett ; 357(1): 364-373, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25434798

ABSTRACT

Cytotoxic ribonucleases such as the leopard frog derivative Ranpirnase (Onconase(®)) have emerged as a valuable new class of cancer therapeutics. Clinical trials employing single agent Ranpirnase in cancer patients have demonstrated significant clinical activity and surprisingly low immunogenicity. However, dose-limiting toxicity due to unspecific uptake of the RNase into non-cancerous cells is reached at relatively low concentrations of > 1 mg/m(2). We have in the present study generated a dimeric anti-EGFR Ranpirnase-diabody fusion protein capable to deliver two Ranpirnase moieties per molecule to EGFR-positive tumour cells. We show that this compound mediated far superior efficacy for killing EGFR-positive tumour cells than a monomeric counterpart. Most importantly, cell killing was restricted to EGFR-positive target cells and no dose-limiting toxicity of Ranpirnase-diabody was observed in mice. These data indicate that by targeted delivery of Ranpirnase non-selective toxicity can be abolished and suggests Ranpirnase-diabody as a promising new drug for therapeutic interventions in EGFR-positive cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/metabolism , Head and Neck Neoplasms/drug therapy , Recombinant Fusion Proteins/pharmacology , Ribonucleases/pharmacology , Animals , Carcinoma, Squamous Cell/enzymology , Cell Line, Tumor , Disease-Free Survival , Female , Head and Neck Neoplasms/enzymology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ribonucleases/genetics , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
3.
MAbs ; 6(1): 130-42, 2014.
Article in English | MEDLINE | ID: mdl-24256717

ABSTRACT

The development of efficient strategies for generating fully human monoclonal antibodies with unique functional properties that are exploitable for tailored therapeutic interventions remains a major challenge in the antibody technology field. Here, we present a methodology for recovering such antibodies from antigen-encountered human B cell repertoires. As the source for variable antibody genes, we cloned immunoglobulin G (IgG)-derived B cell repertoires from lymph nodes of 20 individuals undergoing surgery for head and neck cancer. Sequence analysis of unselected "LYmph Node Derived Antibody Libraries" (LYNDAL) revealed a naturally occurring distribution pattern of rearranged antibody sequences, representing all known variable gene families and most functional germline sequences. To demonstrate the feasibility for selecting antibodies with therapeutic potential from these repertoires, seven LYNDAL from donors with high serum titers against herpes simplex virus (HSV) were panned on recombinant glycoprotein B of HSV-1. Screening for specific binders delivered 34 single-chain variable fragments (scFvs) with unique sequences. Sequence analysis revealed extensive somatic hypermutation of enriched clones as a result of affinity maturation. Binding of scFvs to common glycoprotein B variants from HSV-1 and HSV-2 strains was highly specific, and the majority of analyzed antibody fragments bound to the target antigen with nanomolar affinity. From eight scFvs with HSV-neutralizing capacity in vitro,the most potent antibody neutralized 50% HSV-2 at 4.5 nM as a dimeric (scFv)2. We anticipate our approach to be useful for recovering fully human antibodies with therapeutic potential.


Subject(s)
Antibodies, Viral , Cloning, Molecular , Gene Library , Herpesvirus 1, Human/immunology , Herpesvirus 2, Human/immunology , Immunoglobulin G , Immunoglobulin Variable Region , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Female , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Male , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...