The Hippo pathway is an evolutionary conserved signaling network that regulates essential processes such as organ size, cell proliferation, migration, stemness and apoptosis. Alterations in this pathway are commonly found in solid tumors and can lead to hyperproliferation, resistance to chemotherapy, compensation for mKRAS and tumor immune evasion. As the terminal effectors of the Hippo pathway, the transcriptional coactivators YAP1/TAZ and the transcription factors TEAD1–4 present exciting opportunities to pharmacologically modulate the Hippo biology in cancer settings, inflammation and regenerative medicine. This review will provide an overview of the progress and current strategies to directly and indirectly target the YAP1/TAZ protein–protein interaction (PPI) with TEAD1–4 across multiple modalities, with focus on recent small molecules able to selectively bind to TEAD, block its autopalmitoylation and inhibit YAP1/TAZ–TEAD-dependent transcription in cancer.
The
leucine-rich repeat kinase 2 (LRRK2) protein has been genetically
and functionally linked to Parkinson’s disease (PD), a disabling
and progressive neurodegenerative disorder whose current therapies
are limited in scope and efficacy. In this report, we describe a rigorous
hit-to-lead optimization campaign supported by structural enablement,
which culminated in the discovery of brain-penetrant, candidate-quality
molecules as represented by compounds 22 and 24. These compounds exhibit remarkable selectivity against the kinome
and offer good oral bioavailability and low projected human doses.
Furthermore, they showcase the implementation of stereochemical design
elements that serve to enable a potency- and selectivity-enhancing
increase in polarity and hydrogen bond donor (HBD) count while maintaining
a central nervous system-friendly profile typified by low levels of
transporter-mediated efflux and encouraging brain penetration in preclinical
models.
The
clinical success of anti-IL-17 monoclonal antibodies (i.e.,
Cosentyx and Taltz) has validated Th17 pathway modulation for the
treatment of autoimmune diseases. The nuclear hormone receptor RORγt
is a master regulator of Th17 cells and affects the production of
a host of cytokines, including IL-17A, IL-17F, IL-22, IL-26, and GM-CSF.
Substantial interest has been spurred across both academia and industry
to seek small molecules suitable for RORγt inhibition. A variety
of RORγt inhibitors have been reported in the past few years,
the majority of which are orthosteric binders. Here we disclose the
discovery and optimization of a class of inhibitors, which bind differently
to an allosteric binding pocket. Starting from a weakly active hit 1, a tool compound 14 was quickly identified
that demonstrated superior potency, selectivity, and off-target profile.
Further optimization focused on improving metabolic stability. Replacing
the benzoic acid moiety with piperidinyl carboxylate, modifying the
4-aza-indazole core in 14 to 4-F-indazole, and incorporating
a key hydroxyl group led to the discovery of 25, which
possesses exquisite potency and selectivity, as well as an improved
pharmacokinetic profile suitable for oral dosing.
4,5-Diacetamidoacridine-9(10H)-one was prepared, and its interactions with halide and benzoate anions were studied using a combination of NMR, fluorescence, and isothermal titration calorimetry experiments. Whereas chloride and bromide exhibited simple association, both fluoride and benzoate exhibited initial entropy-driven association followed by an enthalpically favorable deprotonation of the receptor by a second equivalent of the anion.
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.