Nanotracing and cavity-ring down spectroscopy: A new ultrasensitive approach in large molecule drug disposition studies

Authors
Kratochwil, Nicole A.Dueker, Stephen R.Muri, DieterSenn, ClaudiaYoon, HyeJinYu, Byung-YongLee, Gwan-HoDong, FengOtteneder, Michael B.
Issue Date
2018-10-17
Publisher
PUBLIC LIBRARY SCIENCE
Citation
PLOS ONE, v.13, no.10
Abstract
New therapeutic biological entities such as bispecific antibodies targeting tissue or specific cell populations form an increasingly important part of the drug development portfolio. However, these biopharmaceutical agents bear the risk of extensive target-mediated drug disposition or atypical pharmacokinetic properties as compared to canonical antibodies. Pharmacokinetics and bio-distribution studies become therefore more and more important during lead optimization. Biologics present, however, greater analytical challenges than small molecule drugs due to the mass and selectivity limitation of mass spectrometry and ligand-binding assay, respectively. Radiocarbon (C-14) and its detection methods, such as the emerging C-14 cavity ring down spectroscopy (CRDS), thus can play an important role in the large molecule quantitation where a C-14-tag is covalently bound through a stable linker. CRDS has the advantage of a simplified sample preparation and introduction system as compared to accelerator mass spectrometry (AMS) and can be accommodated within an ordinary research laboratory. In this study, we report on the labeling of an anti-IL17 IgG 1 model antibody with C-14 propionate tag and its detection by CRDS using it as nanotracer (2.1 nCi or 77.7 Bq blended with the therapeutic dose) in a pharmacokinetics study in a pre-clinical species. We compare these data to data generated by AMS in parallel processed samples. The derived concentration time profiles for anti-IL17 by CRDS were in concordance with the ones derived by AMS and gamma-counting of an I-125-labeled anti-IL17 radiotracer and were well described by a 2-compartment population pharmacokinetic model. In addition, antibody tissue distribution coefficients for anti-IL17 were determined by CRDS, which proved to be a direct and sensitive measurement of the extravascular tissue concentration of the antibody when tissue perfusion was applied. Thus, this proof-of-concept study demonstrates that trace C-14-radiolabels and CRDS are an ultrasensitive approach in (pre)clinical pharmacokinetics and bio-distribution studies of new therapeutic entities.
Keywords
ACCELERATOR MASS-SPECTROMETRY; LASER SPECTROSCOPY; RADIOCARBON; ANTIBODIES; PHARMACOKINETICS; C-14; PARAMETERS; DIOXIDE; SAMPLES; ADME; ACCELERATOR MASS-SPECTROMETRY; LASER SPECTROSCOPY; RADIOCARBON; ANTIBODIES; PHARMACOKINETICS; C-14; PARAMETERS; DIOXIDE; SAMPLES; ADME; Cavity ring-down spectroscopy; disposition; large molecule; accelerator mass spectrometry
ISSN
1932-6203
URI
https://pubs.kist.re.kr/handle/201004/120783
DOI
10.1371/journal.pone.0205435
Appears in Collections:
KIST Article > 2018
Files in This Item:
There are no files associated with this item.
Export
RIS (EndNote)
XLS (Excel)
XML

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.

BROWSE