Regulated Induced Proximity Targeting Chimeras (RIPTACs): a Novel Heterobifunctional Small Molecule Therapeutic Strategy for Killing Cancer Cells Selectively

Kanak Raina, Chris D. Forbes, Rebecca Stronk, Jonathan P. Rappi,Kyle J. Eastman, Samuel W. Gerritz,Xinheng Yu, Hao Li,Amit Bhardwaj, Mia Forgione, Abigail Hundt, Madeline P. King,Zoe M. Posner, Allison Denny,Andrew McGovern, David E. Puleo, Ethan Garvin, Rebekka Chenard,Nilesh Zaware,James J. Mousseau,Jennifer Macaluso, Michael Martin, Kyle Bassoli,Kelli Jones,Marco Garcia, Katia Howard, Levi M. Smith, Jinshan M. Chen, Cesar A. De Leon,John Hines,Katherine J. Kayser-Bricker,Craig M. Crews

biorxiv(2023)

引用 6|浏览7
暂无评分
摘要
While specific cell signaling pathway inhibitors have yielded great success in oncology, directly triggering cancer cell death is one of the great drug discovery challenges facing biomedical research in the era of precision oncology. Attempts to eradicate cancer cells expressing unique target proteins, such as antibody-drug conjugates (ADCs), T-cell engaging therapies, and radiopharmaceuticals have been successful in the clinic, but they are limited by the number of targets given the inability to target intracellular proteins. More recently, heterobifunctional small molecules such as Proteolysis Targeting Chimera (PROTACs) have paved the way for protein proximity inducing therapeutic modalities. Here, we describe a proof-of-concept study using novel heterobifunctional small molecules called R egulated I nduced P roximity Ta rgeting C himeras or RIPTACs, which elicit a stable ternary complex between a target protein selectively expressed in cancer tissue and a pan-expressed protein essential for cell survival. The resulting cooperative protein:protein interaction (PPI) abrogates the function of the essential protein, thus leading to cell death selectively in cells expressing the target protein. This approach not only opens new target space by leveraging differentially expressed intracellular proteins but also has the advantage of not requiring the target to be a driver of disease. Thus, RIPTACs can address non-target mechanisms of resistance given that cell killing is driven by inactivation of the essential protein. Using the HaloTag7-FKBP model system as a target protein, we describe RIPTACs that incorporate a covalent or non-covalent target ligand connected via a linker to effector ligands such as JQ1 (BRD4), BI2536 (PLK1), or multi-CDK inhibitors such as TMX3013 or dinaciclib. We show that these RIPTACs exhibit positive co-operativity, accumulate selectively in cells expressing HaloTag7-FKBP, form stable target:RIPTAC:effector trimers in cells, and induce an anti-proliferative response in target-expressing cells. We propose that RIPTACs are a novel heterobifunctional therapeutic modality to treat cancers that are known to selectively express a specific intracellular protein. ### Competing Interest Statement C.M.C. is a shareholder and consultant to Halda Therapeutics, which supports research in his laboratory.
更多
查看译文
关键词
induced proximity targeting chimeras,killing cancer cells,riptacs
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要