Targeting a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) for RAS-driven cancers

Extracellular proteolysis is frequently dysregulated in disease and can generate proteoforms with unique neoepitopes not found in healthy tissue. Here, we demonstrate that Abs that selectively recognize a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) could enable more effective a...

Full description

Saved in:
Bibliographic Details
Published inThe Journal of clinical investigation Vol. 132; no. 4
Main Authors Lim, Shion A, Zhou, Jie, Martinko, Alexander J, Wang, Yung-Hua, Filippova, Ekaterina V, Steri, Veronica, Wang, Donghui, Remesh, Soumya G, Liu, Jia, Hann, Byron, Kossiakoff, Anthony A, Evans, Michael J, Leung, Kevin K, Wells, James A
Format Journal Article
LanguageEnglish
Published United States American Society for Clinical Investigation 15.02.2022
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Extracellular proteolysis is frequently dysregulated in disease and can generate proteoforms with unique neoepitopes not found in healthy tissue. Here, we demonstrate that Abs that selectively recognize a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) could enable more effective and safer treatments for solid tumors. CDCP1 is highly overexpressed in RAS-driven cancers, and its ectodomain is cleaved by extracellular proteases. Biochemical, biophysical, and structural characterization revealed that the 2 cleaved fragments of CDCP1 remain tightly associated with minimal proteolysis-induced conformational change. Using differential phage display, we generated recombinant Abs that are exquisitely selective to cleaved CDCP1 with no detectable binding to the uncleaved form. These Abs potently targeted cleaved CDCP1-expressing cancer cells as an Ab-drug conjugate, an Ab-radionuclide conjugate, and a bispecific T cell engager. In a syngeneic pancreatic tumor model, these cleaved-specific Abs showed tumor-specific localization and antitumor activity with superior safety profiles compared with a pan-CDCP1 approach. Targeting proteolytic neoepitopes could provide an orthogonal "AND" gate for improving the therapeutic index.
Bibliography:U.S. Department of Defense (USDOD)
Prostate Cancer Foundation
AC02-05CH11231; F32 5F32CA236151; PC200407; R01EB025207; 1P41CA196276; CA191018; GM097316; P30 GM124169; S10OD018483
Bristol Myers Squibb
National Institutes of Health (NIH)
USDOE Office of Science (SC), Biological and Environmental Research (BER)
Authorship note: SAL and JZ are co–first authors and contributed equally to the study.
ISSN:1558-8238
0021-9738
1558-8238
DOI:10.1172/JCI154604