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.
Surg Oncol ; 28: 1-8, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30851880

ABSTRACT

Tumor-specific fluorescent imaging agents are moving towards the clinic, supporting surgeons with real-time intraoperative feedback about tumor locations. The epithelial cell adhesion molecule (EpCAM) is considered as one of the most promising tumor-specific proteins due its high overexpression on epithelial-derived cancers. This study describes the development and evaluation of EpCAM-F800, a novel fluorescent anti-EpCAM antibody fragment, for intraoperative tumor imaging. Fab production, conjugation to the fluorophore IRDye 800CW, and binding capacities were determined and validated using HPLC, spectrophotometry and cell-based assays. In vivo, dose escalation-, blocking-, pharmacokinetic- and biodistribution studies (using both fluorescence and radioactivity) were performed, next to imaging of clinically relevant orthotopic xenografts for breast and colorectal cancer. EpCAM-F800 targets EpCAM with high specificity in vitro, which was validated using in vivo blocking experiments with a 10x higher dose of unlabeled Fab. The optimal dose range for fluorescence tumor detection in mice was 1-5 nmol (52-260 µg), which corresponds to a human equivalent dose of 0.2-0.8 mg/kg. Biodistribution showed high accumulation of EpCAM-F800 in tumors and metabolizing organs. Breast and colorectal tumors could clearly be visualized within 8 h post-injection and up to 96 h, while the agent already showed homogenous tumor distribution within 4 h. The blood half-life was 4.5 h. This study describes the development and evaluation of a novel EpCAM-targeting agent and the feasibility to visualize breast and colorectal tumors by fluorescence imaging during resections. EpCAM-F800 will be translated for clinical use, considering its abundance in a broad range of tumor types.


Subject(s)
Benzenesulfonates/pharmacokinetics , Breast Neoplasms/diagnosis , Colorectal Neoplasms/diagnosis , Epithelial Cell Adhesion Molecule/immunology , Immunoglobulin Fragments/immunology , Indoles/pharmacokinetics , Optical Imaging/methods , Adult , Aged , Aged, 80 and over , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Female , Humans , Middle Aged , Spectroscopy, Near-Infrared , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Oncotarget ; 8(13): 21054-21066, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28416744

ABSTRACT

Incomplete resections and damage to critical structures increase morbidity and mortality of patients with cancer. Targeted intraoperative fluorescence imaging aids surgeons by providing real-time visualization of tumors and vital structures. This study evaluated the tumor-targeted zwitterionic near-infrared fluorescent peptide cRGD-ZW800-1 as tracer for intraoperative imaging of multiple cancer types. cRGD-ZW800-1 was validated in vitro on glioblastoma (U-87 MG) and colorectal (HT-29) cell lines. Subsequently, the tracer was tested in orthotopic mouse models with HT-29, breast (MCF-7), pancreatic (BxPC-3), and oral (OSC-19) tumors. Dose-ranging studies, including doses of 0.25, 1.0, 10, and 30 nmol, in xenograft tumor models suggest an optimal dose of 10 nmol, corresponding to a human equivalent dose of 63 µg/kg, and an optimal imaging window between 2 and 24 h post-injection. The mean half-life of cRGD-ZW800-1 in blood was 25 min. Biodistribution at 4 h showed the highest fluorescence signals in tumors and kidneys. In vitro and in vivo competition experiments showed significantly lower fluorescence signals when U-87 MG cells (minus 36%, p = 0.02) or HT-29 tumor bearing mice (TBR at 4 h 3.2 ± 0.5 vs 1.8 ± 0.4, p = 0.03) were simultaneously treated with unlabeled cRGD. cRGD-ZW800-1 visualized in vivo all colorectal, breast, pancreatic, and oral tumor xenografts in mice. Screening for off-target interactions, cRGD-ZW800-1 showed only inhibition of COX-2, likely due to binding of cRGD-ZW800-1 to integrin αVß3. Due to its recognition of various integrins, which are expressed on malignant and neoangiogenic cells, it is expected that cRGD-ZW800-1 will provide a sensitive and generic tool to visualize cancer during surgery.


Subject(s)
Neoplasms/diagnostic imaging , Optical Imaging/methods , Peptides, Cyclic/pharmacokinetics , Quaternary Ammonium Compounds/pharmacokinetics , Sulfonic Acids/pharmacokinetics , Animals , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Disease Models, Animal , Feasibility Studies , Female , HT29 Cells , Half-Life , Humans , Integrin alphaVbeta3/metabolism , Intraoperative Period , MCF-7 Cells , Mice , Mice, Nude , Neoplasms/surgery , Optical Imaging/instrumentation , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/adverse effects , Quaternary Ammonium Compounds/administration & dosage , Quaternary Ammonium Compounds/adverse effects , Sulfonic Acids/administration & dosage , Sulfonic Acids/adverse effects , Time Factors , Tissue Distribution , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...