Edmond M. Kwan, Sarah W. S. Ng, Sofie H. Tolmeijer, Louise Emmett, Shahneen Sandhu, James P. Buteau, Amir Iravani, Anthony M. Joshua, Roslyn J. Francis, Vinod Subhash, Sze-Ting Lee, Andrew M. Scott, Andrew J. Martin, Martin R. Stockler, Gráinne Donnellan, Matti Annala, Cameron Herberts, Ian D. Davis, Michael S. Hofman, Arun A. Azad, Alexander W. Wyatt
{"title":"Lutetium-177–PSMA-617 or cabazitaxel in metastatic prostate cancer: circulating tumor DNA analysis of the randomized phase 2 TheraP trial","authors":"Edmond M. Kwan, Sarah W. S. Ng, Sofie H. Tolmeijer, Louise Emmett, Shahneen Sandhu, James P. Buteau, Amir Iravani, Anthony M. Joshua, Roslyn J. Francis, Vinod Subhash, Sze-Ting Lee, Andrew M. Scott, Andrew J. Martin, Martin R. Stockler, Gráinne Donnellan, Matti Annala, Cameron Herberts, Ian D. Davis, Michael S. Hofman, Arun A. Azad, Alexander W. Wyatt","doi":"10.1038/s41591-025-03704-9","DOIUrl":null,"url":null,"abstract":"<p>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 (<i>n</i> = 97) versus cabazitaxel chemotherapy (<i>n</i> = 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%, <i>P</i> = 0.0067) and PFS (median 14.7 versus 6.0 months; hazard ratio 0.12, <i>P</i> = 2.5 × 10<sup>−4</sup>) on [¹⁷⁷Lu]Lu–PSMA-617 independent of predictive PSMA–positron emission tomography imaging parameters, although this benefit did not extend to OS. Deleterious <i>PTEN</i> alterations were associated with worse PFS and OS on cabazitaxel, whereas <i>ATM</i> 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 <i>FOLH1</i>) 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.</p>","PeriodicalId":19037,"journal":{"name":"Nature Medicine","volume":"133 1","pages":""},"PeriodicalIF":58.7000,"publicationDate":"2025-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Nature Medicine","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1038/s41591-025-03704-9","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOCHEMISTRY & MOLECULAR BIOLOGY","Score":null,"Total":0}
引用次数: 0
Abstract
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 × 10−4) 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.
期刊介绍:
Nature Medicine is a monthly journal publishing original peer-reviewed research in all areas of medicine. The publication focuses on originality, timeliness, interdisciplinary interest, and the impact on improving human health. In addition to research articles, Nature Medicine also publishes commissioned content such as News, Reviews, and Perspectives. This content aims to provide context for the latest advances in translational and clinical research, reaching a wide audience of M.D. and Ph.D. readers. All editorial decisions for the journal are made by a team of full-time professional editors.
Nature Medicine consider all types of clinical research, including:
-Case-reports and small case series
-Clinical trials, whether phase 1, 2, 3 or 4
-Observational studies
-Meta-analyses
-Biomarker studies
-Public and global health studies
Nature Medicine is also committed to facilitating communication between translational and clinical researchers. As such, we consider “hybrid” studies with preclinical and translational findings reported alongside data from clinical studies.