Your browser doesn't support javascript.
loading
PROTAC-mediated dual degradation of BCL-xL and BCL-2 is a highly effective therapeutic strategy in small-cell lung cancer.
Khan, Sajid; Cao, Lin; Wiegand, Janet; Zhang, Peiyi; Zajac-Kaye, Maria; Kaye, Frederic J; Zheng, Guangrong; Zhou, Daohong.
  • Khan S; Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
  • Cao L; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
  • Wiegand J; Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
  • Zhang P; Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA.
  • Zajac-Kaye M; Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida, USA.
  • Kaye FJ; Department of Anatomy & Cell Biology, College of Medicine, University of Florida, Gainesville, Florida, USA.
  • Zheng G; Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA.
  • Zhou D; Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida, USA.
bioRxiv ; 2024 Mar 01.
Article en En | MEDLINE | ID: mdl-38464204
ABSTRACT
BCL-xL and BCL-2 are validated therapeutic targets in small-cell lung cancer (SCLC). Targeting these proteins with navitoclax (formerly ABT263, a dual BCL-xL/2 inhibitor) induces dose-limiting thrombocytopenia through on-target BCL-xL inhibition in platelets. Therefore, platelet toxicity poses a barrier in advancing the clinical translation of navitoclax. We have developed a strategy to selectively target BCL-xL in tumors, while sparing platelets, by utilizing proteolysis-targeting chimeras (PROTACs) that hijack the cellular ubiquitin proteasome system for target ubiquitination and subsequent degradation. In our previous study, the first-in-class BCL-xL PROTAC, called DT2216, was shown to have synergistic antitumor activities when combined with venetoclax (formerly ABT199, BCL-2-selective inhibitor) in a BCL-xL/2 co-dependent SCLC cell line, NCI-H146 (hereafter referred to as H146), in vitro and in a xenograft model. Guided by these findings, we evaluated our newly developed BCL-xL/2 dual degrader, called 753b, in three BCL-xL/2 co-dependent SCLC cell lines and the H146 xenograft models. 753b was found to degrade both BCL-xL and BCL-2 in these cell lines. Importantly, it was considerably more potent than DT2216, navitoclax, or DT2216+venetoclax to reduce the viability of BCL-xL/2 co-dependent SCLC cell lines in cell culture. In vivo, 5 mg/kg weekly dosing of 753b leads to significant tumor growth delay similar to the DT2216+venetoclax combination in H146 xenografts by degrading both BCL-xL and BCL-2. Additionally, 753b administration at 5 mg/kg every four days induced tumor regressions. 753b at this dosage was well tolerated in mice without induction of severe thrombocytopenia as seen with navitoclax nor induced significant changes in mouse body weights. These results suggest that the BCL-xL/2 dual degrader could be an effective and safe therapeutic for a subset of SCLC patients warranting clinical trials in future.
Palabras clave