학술논문

Rationally designed BCL6 inhibitors target activated B cell diffuse large B cell lymphoma
Document Type
article
Source
Journal of Clinical Investigation. 126(9)
Subject
Biomedical and Clinical Sciences
Cardiovascular Medicine and Haematology
Oncology and Carcinogenesis
Orphan Drug
Cancer
Hematology
Genetics
Lymphoma
Biotechnology
Rare Diseases
Development of treatments and therapeutic interventions
5.1 Pharmaceuticals
Animals
Antineoplastic Agents
Cell Line
Tumor
Doxorubicin
Drug Design
Drug Screening Assays
Antitumor
Gene Expression Regulation
Neoplastic
HEK293 Cells
Humans
Indoles
Ligands
Lymphoma
Large B-Cell
Diffuse
Magnetic Resonance Spectroscopy
Male
Mice
Mice
SCID
Neoplasm Transplantation
Protein Binding
Proto-Oncogene Proteins c-bcl-6
Thiazolidinediones
Translocation
Genetic
Medical and Health Sciences
Immunology
Biological sciences
Biomedical and clinical sciences
Health sciences
Language
Abstract
Diffuse large B cell lymphomas (DLBCLs) arise from proliferating B cells transiting different stages of the germinal center reaction. In activated B cell DLBCLs (ABC-DLBCLs), a class of DLBCLs that respond poorly to current therapies, chromosomal translocations and amplification lead to constitutive expression of the B cell lymphoma 6 (BCL6) oncogene. The role of BCL6 in maintaining these lymphomas has not been investigated. Here, we designed small-molecule inhibitors that display higher affinity for BCL6 than its endogenous corepressor ligands to evaluate their therapeutic efficacy for targeting ABC-DLBCL. We used an in silico drug design functional-group mapping approach called SILCS to create a specific BCL6 inhibitor called FX1 that has 10-fold greater potency than endogenous corepressors and binds an essential region of the BCL6 lateral groove. FX1 disrupted formation of the BCL6 repression complex, reactivated BCL6 target genes, and mimicked the phenotype of mice engineered to express BCL6 with corepressor binding site mutations. Low doses of FX1 induced regression of established tumors in mice bearing DLBCL xenografts. Furthermore, FX1 suppressed ABC-DLBCL cells in vitro and in vivo, as well as primary human ABC-DLBCL specimens ex vivo. These findings indicate that ABC-DLBCL is a BCL6-dependent disease that can be targeted by rationally designed inhibitors that exceed the binding affinity of natural BCL6 ligands.