Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.
Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
Clin Cancer Res. 2024 Jul 15;30(14):2917-2924. doi: 10.1158/1078-0432.CCR-24-0428.
The antibody-drug conjugate (ADC) sacituzumab govitecan (SG) comprises the topoisomerase 1 (TOP1) inhibitor (TOP1i) SN-38, coupled to a monoclonal antibody targeting trophoblast cell surface antigen 2 (TROP-2). Poly(ADP-ribose) polymerase (PARP) inhibition may synergize with TOP1i and SG, but previous studies combining systemic PARP and TOP1 inhibitors failed due to dose-limiting myelosuppression. Here, we assess the proof-of-mechanism and clinical feasibility for SG and talazoparib (TZP) employing an innovative sequential dosing schedule.
In vitro models tested pharmacodynamic endpoints, and in a phase 1b clinical trial (NCT04039230), 30 patients with metastatic triple-negative breast cancer (mTNBC) received SG and TZP in a concurrent (N = 7) or sequential (N = 23) schedule. Outcome measures included safety, tolerability, preliminary efficacy, and establishment of a recommended phase 2 dose.
We hypothesized that tumor-selective delivery of TOP1i via SG would reduce nontumor toxicity and create a temporal window, enabling sequential dosing of SG and PARP inhibition. In vitro, sequential SG followed by TZP delayed TOP1 cleavage complex clearance, increased DNA damage, and promoted apoptosis. In the clinical trial, sequential SG/TZP successfully met primary objectives and demonstrated median progression-free survival (PFS) of 7.6 months without dose-limiting toxicities (DLT), while concurrent dosing yielded 2.3 months PFS and multiple DLTs including severe myelosuppression.
While SG dosed concurrently with TZP is not tolerated clinically due to an insufficient therapeutic window, sequential dosing of SG followed by TZP proved a viable strategy. These findings support further clinical development of the combination and suggest that ADC-based therapy may facilitate novel, mechanism-based dosing strategies.
抗体药物偶联物(ADC)Sacituzumab govitecan(SG)包含拓扑异构酶 1(TOP1)抑制剂(TOP1i)SN-38,与靶向滋养细胞表面抗原 2(TROP-2)的单克隆抗体偶联。聚(ADP-核糖)聚合酶(PARP)抑制可能与 TOP1i 和 SG 协同作用,但由于剂量限制的骨髓抑制,以前联合使用系统 PARP 和 TOP1 抑制剂的研究均以失败告终。在这里,我们采用创新的序贯给药方案评估 SG 和他拉唑帕尼(TZP)的作用机制和临床可行性。
体外模型测试了药效学终点,在一项 1b 期临床试验(NCT04039230)中,30 名转移性三阴性乳腺癌(mTNBC)患者接受了 SG 和 TZP 的同时(N=7)或序贯(N=23)给药方案。主要终点包括安全性、耐受性、初步疗效和推荐的 2 期剂量的确定。
我们假设通过 SG 进行的 TOP1i 的肿瘤选择性递送将减少非肿瘤毒性并产生一个时间窗口,从而能够进行 SG 和 PARP 抑制的序贯给药。在体外,SG 序贯 TZP 延迟了 TOP1 切割复合物的清除,增加了 DNA 损伤,并促进了细胞凋亡。在临床试验中,序贯 SG/TZP 成功达到了主要目标,且未出现剂量限制毒性(DLT)的情况下,中位无进展生存期(PFS)为 7.6 个月,而同时给药的 PFS 为 2.3 个月,且发生了包括严重骨髓抑制在内的多个 DLT。
虽然由于治疗窗口不足,SG 与 TZP 同时给药在临床上无法耐受,但 SG 序贯 TZP 给药是可行的策略。这些发现支持该联合用药的进一步临床开发,并表明 ADC 为基础的治疗可能促进新的、基于机制的给药策略。