Phenyl Dihydrouracil: An Alternative Cereblon Binder for PROTAC Design
Use of proteolysis targeting chimeras (PROTACs) is an innovative approach developed to silence a specific target from the biological environment. PROTACs are heterobifunctional conjugates of a ligand that binds to the target protein of interest with a second ligand for an E3 ligase system. Once formed, a ternary complex between an E3 ubiquitin ligase polyubiquitinates the target protein, causing its degradation by the proteasome.
In this issue, Jarusiewicz
et al. (DOI: 10.1021/acsmedchemlett.2c00436) prepared and analyzed novel cereblon ligands endowed with favorable
pharmacokinetic (PK) and stability properties. Due to the chemical
instability of thalidomide derivatives in in vitro assays, including imide hydrolysis and racemization, the authors
previously synthesized a family of phenyl glutarimide (PG)-containing
compounds which demonstrated a high affinity for E3 ligase and improved
stability. Nevertheless, the drawback of the PG series is their propensity
to still racemize, resulting in a reduction in potency due to the
inactivity of the S-enantiomer. To overcome the racemization
issue, the authors replaced the PG component with an achiral phenyl
dihydrouracil (PD)-based moiety. Three different PROTAC systems were
synthesized while maintaining dasatinib as the LCK ligand and different
E3 ligands and linkers for comparative studies. Cereblon affinity,
chemical stability, target degradation, and cytotoxicity were assessed.
PD-PROTACs exhibited only a modest reduction in cereblon affinity
but displayed improved chemical stability compared to PG analogs.
The good stability values increased the cellular degradation of LCK
and the antiproliferative activity on KOPT-K1 cells, leading to the
most potent LCK degrader to date. ADME analysis revealed acceptable
active permeability but low microsomal stability. High metabolic clearance
remains a liability for this compound class, including parent molecule
dasatinib. This study provides more design principles on cereblon
ligands and offers new insights for improving PROTACs.
Discovery of OICR12694: A Novel, Potent, Selective, and Orally Bioavailable BCL6 BTB Inhibitor
Diffuse large B-cell lymphoma (DLBCL) is the most common type of malignant non-Hodgkin lymphoma (NHL) that affects B-lymphocytes. Although DLBCLs can develop at any age, they are more frequent in older people. Despite the success of frontline therapy, due to tumor heterogeneity, novel targeted therapies are required.
In this issue, Mamai
and co-workers (DOI: 10.1021/acsmedchemlett.2c00502) disclose the identification of OICR12694/JNJ-65234637,
a BCL6-BTB inhibitor that exhibits potent antiproliferative activity
against DLCBL cells and excellent oral bioavailability. Starting from
a series of previously reported BCL6 inhibitors, the authors efficiently
performed a virtual screen and tested the top-scoring compounds in
a surface plasmon resonance assay. The screening campaign led to the
identification of a novel pyrrolopyridone hit structure with
inhibitory activity in the high micromolar range. Further optimization
and co-crystal structures of analogs with the BCL6 BTB domain resulted
in the synthesis of a series of tricyclic C3-substituted pyrrolopyrimidinones
with potent biochemical and nanomolar cellular activity. ADME analysis
revealed good microsomal stability and high active permeability. Pharmacokinetic
studies in both mouse and dog models confirmed good oral bioavailability.
The absence of genotoxicity and the minimal inhibition of the hERG
channel highlighted the success of the optimization campaign.
Pyrazole Ureas as Low Dose, CNS Penetrant Glucosylceramide Synthase Inhibitors for the Treatment of Parkinson’s Disease
Parkinson’s disease (PD) is a complex neurological pathology with a multifactorial origin. It is marked by tremors and bradykinesia. While the exact molecular causes of PD are still unknown, various cellular processes and genes have been linked to its progression. One process includes the loss of function of the glucosylcerebrosidase (GBA) gene, leading to central nervous system (CNS) accumulation of glucosylceramide and glucosylsphingosine.
In this issue, Roecker
and co-workers from Merck (DOI: 10.1021/acsmedchemlett.2c00441) describe the identification
of novel small molecules that block glucosylceramide synthase (GCS).
Starting from a hit previously disclosed from a high-throughput screening
campaign and characterized by suboptimal potency but excellent CNS
permeability, the authors carried out extensive structure–activity
relationship studies. To enrich the possibility of CNS penetration,
the internal library was filtered with a CNS multiparameter score
function, and permeability and P-gp efflux were taken into consideration
from the first phases of analysis. The optimization protocol resulted
in a novel series of potent compounds. The absence of CYP and hERG
inhibition, coupled with good in vitro ADME, led
the authors to perform in vivo pharmacokinetic and
pharmacodynamic studies. The lead compound demonstrated a statistically
relevant dose-dependent reduction of GlcCer in C57 black mice, reaching
the highest effect at a dose of 100 mg/kg. Overall, this paper described
a novel scaffold that inhibits GCS in mice and introduced new molecular
tools to better understand GSC biology in in vivo and ex vivo models.
