Kwan Edmond M, Ng Sarah W S, Tolmeijer Sofie H, Emmett Louise, Sandhu Shahneen, Buteau James P, Iravani Amir, Joshua Anthony M, Francis Roslyn J, Subhash Vinod, Lee Sze-Ting, Scott Andrew M, Martin Andrew J, Stockler Martin R, Donnellan Gráinne, Annala Matti, Herberts Cameron, Davis Ian D, Hofman Michael S, Azad Arun A, Wyatt Alexander W
Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia.
Nat Med. 2025 May 27. doi: 10.1038/s41591-025-03704-9.
The prostate-specific membrane antigen (PSMA)-targeted radioligand [¹⁷⁷Lu]Lu-PSMA-617 is a new standard treatment for metastatic castration-resistant prostate cancer (mCRPC), but predictive genomic biomarkers informing its rational use are unknown. We performed detailed dissection of prostate cancer driver genes across 290 serial plasma cell-free DNA samples from 180 molecular imaging-selected patients with mCRPC from the randomized TheraP trial of [¹⁷⁷Lu]Lu-PSMA-617 (n = 97) versus cabazitaxel chemotherapy (n = 83). The primary endpoint was PSA50 biochemical response, with secondary endpoints of progression-free survival (PFS) and overall survival (OS). In this post-hoc biomarker analysis, a low pretreatment circulating tumor DNA (ctDNA) fraction predicted a superior biochemical response (100% versus 58%, P = 0.0067) and PFS (median 14.7 versus 6.0 months; hazard ratio 0.12, P = 2.5 × 10) on [¹⁷⁷Lu]Lu-PSMA-617 independent of predictive PSMA-positron emission tomography imaging parameters, although this benefit did not extend to OS. Deleterious PTEN alterations were associated with worse PFS and OS on cabazitaxel, whereas ATM defects were observed in select patients with favorable [¹⁷⁷Lu]Lu-PSMA-617 outcomes. Comparing pretreatment and progression ctDNA revealed population flux but no evidence that alterations in individual mCRPC genes (or FOLH1) are dominant causes of acquired [¹⁷⁷Lu]Lu-PSMA-617 or cabazitaxel resistance. Our results nominate new candidate biomarkers for [¹⁷⁷Lu]Lu-PSMA-617 selection and ultimately expand the mCRPC predictive biomarker repertoire. We anticipate our ctDNA fraction-aware analytical framework will aid future precision management strategies for [¹⁷⁷Lu]Lu-PSMA-617 and other PSMA-targeted therapeutics. ClinicalTrials.gov identifier: NCT03392428 .
前列腺特异性膜抗原(PSMA)靶向放射性配体[¹⁷⁷Lu]Lu-PSMA-617是转移性去势抵抗性前列腺癌(mCRPC)的一种新的标准治疗方法,但目前尚不清楚可指导其合理使用的预测性基因组生物标志物。我们对来自180名经分子成像选择的mCRPC患者的290份连续血浆游离DNA样本中的前列腺癌驱动基因进行了详细剖析,这些患者来自[¹⁷⁷Lu]Lu-PSMA-617(n = 97)与卡巴他赛化疗(n = 83)的随机TheraP试验。主要终点是PSA50生化反应,次要终点是无进展生存期(PFS)和总生存期(OS)。在这项事后生物标志物分析中,低预处理循环肿瘤DNA(ctDNA)分数预测[¹⁷⁷Lu]Lu-PSMA-617具有更好的生化反应(100%对58%,P = 0.0067)和PFS(中位值14.7对6.0个月;风险比0.12,P = 2.5×10),且独立于预测性PSMA正电子发射断层扫描成像参数,尽管这种益处未延伸至OS。有害的PTEN改变与卡巴他赛治疗时较差的PFS和OS相关,而在部分[¹⁷⁷Lu]Lu-PSMA-617治疗效果良好的患者中观察到ATM缺陷。比较预处理和疾病进展时的ctDNA显示了群体通量,但没有证据表明单个mCRPC基因(或FOLH1)的改变是获得性[¹⁷⁷Lu]Lu-PSMA-617或卡巴他赛耐药的主要原因。我们的结果为[¹⁷⁷Lu]Lu-PSMA-617的选择提名了新的候选生物标志物,并最终扩展了mCRPC预测性生物标志物的范围。我们预计我们的ctDNA分数感知分析框架将有助于未来对[¹⁷⁷Lu]Lu-PSMA-617和其他PSMA靶向治疗药物的精准管理策略。临床试验.gov标识符:NCT03392428 。