Arvinas Operations, Inc., New Haven, Connecticut.
Thermo Fisher Scientific, Materials and Structural Analysis, Eindhoven, Netherlands.
Clin Cancer Res. 2024 Aug 15;30(16):3549-3563. doi: 10.1158/1078-0432.CCR-23-3465.
PURPOSE: Estrogen receptor (ER) alpha signaling is a known driver of ER-positive (ER+)/human epidermal growth factor receptor 2 negative (HER2-) breast cancer. Combining endocrine therapy (ET) such as fulvestrant with CDK4/6, mTOR, or PI3K inhibitors has become a central strategy in the treatment of ER+ advanced breast cancer. However, suboptimal ER inhibition and resistance resulting from the ESR1 mutation dictates that new therapies are needed. EXPERIMENTAL DESIGN: A medicinal chemistry campaign identified vepdegestrant (ARV-471), a selective, orally bioavailable, and potent small molecule PROteolysis-TArgeting Chimera (PROTAC) degrader of ER. We used biochemical and intracellular target engagement assays to demonstrate the mechanism of action of vepdegestrant, and ESR1 wild-type (WT) and mutant ER+ preclinical breast cancer models to demonstrate ER degradation-mediated tumor growth inhibition (TGI). RESULTS: Vepdegestrant induced ≥90% degradation of wild-type and mutant ER, inhibited ER-dependent breast cancer cell line proliferation in vitro, and achieved substantial TGI (87%-123%) in MCF7 orthotopic xenograft models, better than those of the ET agent fulvestrant (31%-80% TGI). In the hormone independent (HI) mutant ER Y537S patient-derived xenograft (PDX) breast cancer model ST941/HI, vepdegestrant achieved tumor regression and was similarly efficacious in the ST941/HI/PBR palbociclib-resistant model (102% TGI). Vepdegestrant-induced robust tumor regressions in combination with each of the CDK4/6 inhibitors palbociclib, abemaciclib, and ribociclib; the mTOR inhibitor everolimus; and the PI3K inhibitors alpelisib and inavolisib. CONCLUSIONS: Vepdegestrant achieved greater ER degradation in vivo compared with fulvestrant, which correlated with improved TGI, suggesting vepdegestrant could be a more effective backbone ET for patients with ER+/HER2- breast cancer.
目的:雌激素受体(ER)α信号是 ER 阳性(ER+)/人表皮生长因子受体 2 阴性(HER2-)乳腺癌的已知驱动因素。将内分泌治疗(ET)药物,如氟维司群,与 CDK4/6、mTOR 或 PI3K 抑制剂联合使用已成为治疗 ER+晚期乳腺癌的主要策略。然而,由于 ESR1 突变导致的 ER 抑制不足和耐药性,需要新的治疗方法。
实验设计:一项药物化学研究发现了 vepdegestrant(ARV-471),这是一种选择性、口服生物可利用的、有效的小分子 PROteolysis-TArgeting Chimera(PROTAC)降解剂,可降解 ER。我们使用生化和细胞内靶标结合测定来证明 vepdegestrant 的作用机制,并使用 ESR1 野生型(WT)和突变型 ER+的临床前乳腺癌模型来证明 ER 降解介导的肿瘤生长抑制(TGI)。
结果:Vepdegestrant 诱导野生型和突变型 ER 降解超过 90%,在体外抑制依赖 ER 的乳腺癌细胞系增殖,并在 MCF7 原位异种移植模型中实现了实质性的 TGI(87%-123%),优于 ET 药物氟维司群(31%-80%TGI)。在激素非依赖性(HI)突变型 ER Y537S 患者来源的异种移植(PDX)乳腺癌模型 ST941/HI 中,Vepdegestrant 实现了肿瘤消退,在 ST941/HI/PBR 帕博西尼耐药模型中同样有效(TGI 为 102%)。Vepdegestrant 与 CDK4/6 抑制剂帕博西尼、阿贝西利和瑞博西尼、mTOR 抑制剂依维莫司以及 PI3K 抑制剂阿培利司和因伐利斯布联合使用,可导致肿瘤明显消退。
结论:Vepdegestrant 在体内实现了比氟维司群更高的 ER 降解,这与改善的 TGI 相关,表明 Vepdegestrant 可能是一种更有效的 ER+/HER2-乳腺癌患者的基础 ET 药物。
Clin Cancer Res. 2021-3-15
Pharmaceutics. 2025-6-5
MedComm (2020). 2025-6-25
Mol Biomed. 2025-6-19
Front Pharmacol. 2025-6-4
Commun Biol. 2025-6-18
Cancers (Basel). 2025-6-3
Science. 2022-11-4
Front Cell Dev Biol. 2022-4-25
Nat Rev Drug Discov. 2022-3
Breast Cancer Res. 2021-5-12