Signal Transducer and Activator of Transcription 3 (STAT3) plays important roles in the transduction of signals from growth factors and cytokines in both normal and malignant cells. Upon activation, STAT3 controls expression of genes that regulate cell growth, survival, differentiation, stemness and cell-cell interactions. Aberrant activation of STAT3 has been observed in many cancers including lymphoma and leukemias through activating mutations, hyper-signaling through upstream regulators or loss of negative feedback regulation. Additionally, STAT3-mediated cross-talk in the tumor microenvironment results in suppression of immune surveillance compromising anti-tumor immunity.

STAT3 has been historically considered "undruggable". Heterobifunctional degraders that recruit endogenous E3 ligases to ubiquitinate substrate proteins leading to their degradation by the proteosome have emerged as a novel therapeutic modality with great potentials to drug undrugged protein targets. Here we introduce a first-in-class, potent and selective STAT3 degrader KT-333 that is being developed for the treatment of hematologic malignancies and solid tumors.

KT-333 is a small molecule targeted protein degrader of STAT3 and was discovered by an iterative medicinal chemistry SAR campaign improving parameters such as potency, selectivity and ADME profile. It has high solubility in PBS buffer (103 mg/mL, pH7.4). low to moderate clearance in rat, dog and monkey, and moderate plasma half lives across pre-clinical species.

KT-333 exhibits potent and selective degradation of STAT3 with DC 50 of 2.5 -11.8 nM in four anaplastic T cell lymphoma (ALCL) lines. Selective degradation of STAT3 over nearly 9000 proteins, including other STAT family members, was demonstrated by mass spectrometry in human PBMCs . Tumor cells expressing NPM-ALK fusion oncoproteins have activated STAT3 and dependency on STAT3 signaling for survival KT-333 treatment led to rapid apoptosis of these ALK+ALCL cells. To better understand the cellular responses to STAT3 degradation, time-course transcriptomic and proteomic analyses were performed using SU-DHL-1 cells treated with KT-333. Time-dependent changes in both mRNA and protein levels of STAT3-regulated genes and pathways were observed: at 8 h only STAT3 showed significant depletion, at 24 h canonical STAT3 downstream genes including SOCS3, IL-2RA and GRZMB showed modulation and at 48 h, pathways including interferon response and cell cycle were significantly enriched. A comparison of the transcriptomic and proteomic data sets showed high correlation. Furthermore, changes in mRNA and protein levels of bona fide STAT3 regulated targets showed a high degree of concordance between SU-DHL-1 cells and SU-DHL-1 xenograft tumors.

Preclinical in vitro experiments have demonstrated that approximately 90% degradation of STAT3 for 48 h was sufficient to drive ALK+ ALCL lines into irreversible cell growth inhibition and death. In vivo xenograft studies of SU-DHL-1 and SUP-M2 ALCL models demonstrated dose-dependent tumor growth suppression. An indirect response model that linked exposure to target degradation was established to describe the relationship between exposure and target degradation in the tumor. Consistent with the invitro data, PK-PD-efficacy analysis in the SU-DHL-1 xenograft model showed that tumor regression could be achieved with intermittent dosing schedules that achieved ~90% degradation of STAT3 for about 48 h over a weekly or bi-weekly dosing interval. . Based on preclinical ADME and PK/PD/efficacy relationship, KT-333 is predicted to drive tumor regression using weekly doses.

In conclusion, we have identified a potent STAT3 degrader as development candidate that can be administered parenterally in the clinic on an intermittent dosing regimen that is predicted to be both efficacious and tolerated in STAT3-dependent hematologic malignancies.

Disclosures

Liu:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Dixit:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Mayo:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Dey:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Yuan:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Karnik:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Walther:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Shi:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Sharma:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Rong:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Yang:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Gollerkeri:BEAM Therapeutics: Current Employment, Current equity holder in publicly-traded company; Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. Gollob:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company. DeSavi:Kymera Therapeutics: Current Employment, Current equity holder in publicly-traded company.

Sign in via your Institution