Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Drug Metab Dispos ; 40(9): 1677-85, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22627466

ABSTRACT

Although it is widely accepted that one can extend the pharmacokinetic half-life of a therapeutic protein by covalent conjugation with polyethylene glycol (PEG), the disposition properties of such biologics have not yet been fully evaluated. Therefore, a novel [¹4C]-labeling method was developed that can be applied to a biologic conjugated with PEG through a maleimide-cysteine reaction. The method was used to study the tissue and tumor distribution of a PEGylated Adnectin, a recombinant protein derived from the 10th type III domain of fibronectin, in nude mice bearing human xenograft tumors. The PEGylated Adnectin contained a 40-kDa branched PEG (P40B) that was labeled with [¹4C] at the linker region between the PEG and Adnectin, without compromising cellular activity and plasma half-life in mice. After a single intravenous or intraperitoneal dose (33 mg/kg, 1.7 µCi per mouse) of [¹4C]-P40B-Adnectin, quantitative whole-body autoradiography analysis revealed that the liver had the highest uptake of the radioactivity among nontumor tissues, followed by the kidneys and lung. The muscle and brain showed the least penetration of the radioactivity among all tissues examined. In addition, the [¹4C]-P40B-EI-tandem penetrated into the tumor tissue, although the extent of accumulation was largely dependent on tumor type. Therefore, it was possible to assess the tissue distribution of a PEGylated biologic after it had been [¹4C] labeled using the novel method described herein.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Biological Products/pharmacokinetics , Carbon Radioisotopes/pharmacokinetics , Fibronectins/pharmacokinetics , Isotope Labeling/methods , Neoplasms/metabolism , Polyethylene Glycols/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Autoradiography , Biological Products/administration & dosage , Biological Products/chemical synthesis , Carbon Radioisotopes/administration & dosage , Carbon Radioisotopes/chemistry , Cell Line, Tumor , ErbB Receptors/metabolism , Female , Fibronectins/administration & dosage , Fibronectins/chemical synthesis , Fibronectins/genetics , Humans , Injections, Intraperitoneal , Injections, Intravenous , Mice , Mice, Nude , Mutation , Neoplasms/pathology , Phosphorylation , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemical synthesis , Protein Engineering , Receptor, IGF Type 1/metabolism , Recombinant Proteins/pharmacokinetics , Tissue Distribution , Tumor Burden , Whole Body Imaging
2.
MAbs ; 3(1): 38-48, 2011.
Article in English | MEDLINE | ID: mdl-21099371

ABSTRACT

Engineered domains of human fibronectin (Adnectins™) were used to generate a bispecific Adnectin targeting epidermal growth factor receptor (EGFR) and insulin-like growth factor-I receptor (IGF-IR), two transmembrane receptors that mediate proliferative and survival cell signaling in cancer. Single-domain Adnectins that specifically bind EGFR or IGF-IR were generated using mRNA display with a library containing as many as 10 ( 13) Adnectin variants. mRNA display was also used to optimize lead Adnectin affinities, resulting in clones that inhibited EGFR phosphorylation at 7 to 38 nM compared to 2.6 µM for the parental clone. Individual, optimized, Adnectins specific for blocking either EGFR or IGF-IR signaling were engineered into a single protein (EI-Tandem Adnectin). The EI-Tandems inhibited phosphorylation of EGFR and IGF-IR, induced receptor degradation, and inhibited down-stream cell signaling and proliferation of human cancer cell lines (A431, H292, BxPC3 and RH41) with IC 50 values ranging from 0.1 to 113 nM. Although Adnectins bound to EGFR at a site distinct from those of anti-EGFR antibodies cetuximab, panitumumab and nimotuzumab, like the antibodies, the anti-EGFR Adnectins blocked the binding of EGF to EGFR. PEGylated EI-Tandem inhibited the growth of both EGFR and IGF-IR driven human tumor xenografts, induced degradation of EGFR, and reduced EGFR phosphorylation in tumors. These results demonstrate efficient engineering of bispecific Adnectins with high potency and desired specificity. The bispecificity may improve biological activity compared to monospecific biologics as tumor growth is driven by multiple growth factors. Our results illustrate a technological advancement for constructing multi-specific biologics in cancer therapy.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Fibronectins/chemistry , Peptide Fragments/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Female , Humans , Immunoblotting , Kinetics , Mice , Mice, Nude , Molecular Sequence Data , Panitumumab , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protein Binding , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...