Giovanni Trovato, Francesco Schietroma, Laura Chiofalo, Giulia Caira, Annaluisa Bologna, Davide De Sio, Sergio Alfieri, Filippo Lococo, Dania Nachira, Stefano Margaritora, Fabio Pacelli, Francesco Ardito, Felice Giuliante, Brunella Barbaro, Antonia Strippoli, Maria Alessandra Calegari, Lisa Salvatore, Michele Basso, Giampaolo Tortora, Carmelo Pozzo
Background: Metastatic colorectal cancer (mCRC) remains a leading cause of cancer-related mortality. While metastasectomy could improve survival in selected patients, the role of repeated resections in oligorecurrent disease is less defined.
Methods: We retrospectively analyzed patients with mCRC treated at Fondazione Policlinico Universitario Agostino Gemelli-IRCCS (Rome, Italy) between 2010 and 2024. Eligible patients underwent ≥2 resections of metastatic lesions. Disease-free survival after second metastasectomy (DFS2) and overall survival (OS) were the coprimary end points. Prognostic factors were tested with Cox regression, and a composite risk score (Fondazione Policlinico Gemelli Risk Score [FPGRiskScore]) was developed.
Results: Among 1586 patients with mCRC, 396 (24.9%) received at least one metastatic surgery and 143 (9%) underwent ≥2 metastasectomies. Median DFS2 was 8.2 months (95% confidence interval [CI]: 7.3-13.1), and 5-years OS rate was 73.1% (95% CI: 64.5-83.0); after a median follow-up from the last metastasectomy of 34.3 months, 49/143 patients (34.2%) were relapse-free and median DFS after the second surgery of the metastases from the last surgery was 13.1 months (95% CI: 9.1-17.5). Patients stratified by FPGRiskScore (disease-free interval [DFI] from the first metastasectomy ≤ vs. >12 months, metastatic burden ≤ vs >5 cm, RAS/BRAF status, and Eastern Cooperative Oncology Group Performance Status [ECOG PS] scale 0 vs. ≥1) showed distinct outcomes: low-risk patients achieved a median DFS2 of 18.4 months and 5-years OS of 87.6%, compared with 7.8 months/72.0% in intermediate-risk and 4.9 months/55.3% in high-risk group.
Conclusions: Repeated metastasectomy offers substantial survival benefit in selected patients with oligorecurrent mCRC, with long-term disease control achievable in a subset. Prognostic stratification incorporating clinical and molecular features (FPGRiskScore) may refine patient selection and guide multidisciplinary management.
{"title":"Long-term survival and prognostic factors after repeated metastasectomies in metastatic colorectal cancer: a 15-years retrospective study.","authors":"Giovanni Trovato, Francesco Schietroma, Laura Chiofalo, Giulia Caira, Annaluisa Bologna, Davide De Sio, Sergio Alfieri, Filippo Lococo, Dania Nachira, Stefano Margaritora, Fabio Pacelli, Francesco Ardito, Felice Giuliante, Brunella Barbaro, Antonia Strippoli, Maria Alessandra Calegari, Lisa Salvatore, Michele Basso, Giampaolo Tortora, Carmelo Pozzo","doi":"10.1093/oncolo/oyaf384","DOIUrl":"10.1093/oncolo/oyaf384","url":null,"abstract":"<p><strong>Background: </strong>Metastatic colorectal cancer (mCRC) remains a leading cause of cancer-related mortality. While metastasectomy could improve survival in selected patients, the role of repeated resections in oligorecurrent disease is less defined.</p><p><strong>Methods: </strong>We retrospectively analyzed patients with mCRC treated at Fondazione Policlinico Universitario Agostino Gemelli-IRCCS (Rome, Italy) between 2010 and 2024. Eligible patients underwent ≥2 resections of metastatic lesions. Disease-free survival after second metastasectomy (DFS2) and overall survival (OS) were the coprimary end points. Prognostic factors were tested with Cox regression, and a composite risk score (Fondazione Policlinico Gemelli Risk Score [FPGRiskScore]) was developed.</p><p><strong>Results: </strong>Among 1586 patients with mCRC, 396 (24.9%) received at least one metastatic surgery and 143 (9%) underwent ≥2 metastasectomies. Median DFS2 was 8.2 months (95% confidence interval [CI]: 7.3-13.1), and 5-years OS rate was 73.1% (95% CI: 64.5-83.0); after a median follow-up from the last metastasectomy of 34.3 months, 49/143 patients (34.2%) were relapse-free and median DFS after the second surgery of the metastases from the last surgery was 13.1 months (95% CI: 9.1-17.5). Patients stratified by FPGRiskScore (disease-free interval [DFI] from the first metastasectomy ≤ vs. >12 months, metastatic burden ≤ vs >5 cm, RAS/BRAF status, and Eastern Cooperative Oncology Group Performance Status [ECOG PS] scale 0 vs. ≥1) showed distinct outcomes: low-risk patients achieved a median DFS2 of 18.4 months and 5-years OS of 87.6%, compared with 7.8 months/72.0% in intermediate-risk and 4.9 months/55.3% in high-risk group.</p><p><strong>Conclusions: </strong>Repeated metastasectomy offers substantial survival benefit in selected patients with oligorecurrent mCRC, with long-term disease control achievable in a subset. Prognostic stratification incorporating clinical and molecular features (FPGRiskScore) may refine patient selection and guide multidisciplinary management.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12668679/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145544001","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shuqin Dai, Yanling Wen, Xingping Wu, Xi Wang, Zhongyu Yuan, Fei Xu, Xiaoming Xie, Wen Xia, Shusen Wang, Yanxia Shi, Peng Liu, Qiufan Zheng, Jun Tang, Xin An, Jiajia Huang, Meiting Chen, Yongyi Zhong, Xiwen Bi, Yi Yang, Cong Xue
Background: Limited data are available on estradiol (E2) levels during fulvestrant treatment in women with hormone receptor-positive breast cancer.
Methods: We measured plasma E2 levels in women receiving fulvestrant using liquid chromatography-tandem mass spectrometry. Patient characteristics and treatment efficacy were assessed in relation to E2 levels. A cutoff of 2.72 pg/mL was used because it defines E2 suppression and postmenopausal status.
Results: A total of 69 women were enrolled, with a median age of 48 years. The median duration of fulvestrant treatment was 11.6 months. The median E2 level across the cohort was 3.60 pg/mL, with considerable interindividual variability (range: 1.11-526.13 pg/mL), and 49 women (71.0%) had E2 levels above 2.72 pg/mL. Eleven women (15.9%) had premenopausal E2 levels (>10 pg/mL). During a median follow-up period of 8.4 months, there was no statistically significant difference in progression-free survival (PFS) between women with E2 levels >2.72 pg/mL and those with E2 levels ≤2.72 pg/mL (P = .391). However, among women who benefited from first- or second-line fulvestrant therapy (PFS > 6 months), those with E2 levels >2.72 pg/mL exhibited significantly poorer PFS compared to those with E2 levels ≤2.72 pg/mL (P = .043).
Conclusions: These findings support the need for E2 monitoring in women receiving fulvestrant therapy to better assess E2 status and its association with treatment efficacy.
{"title":"Estradiol levels in women with hormone receptor-positive advanced breast cancer on fulvestrant therapy.","authors":"Shuqin Dai, Yanling Wen, Xingping Wu, Xi Wang, Zhongyu Yuan, Fei Xu, Xiaoming Xie, Wen Xia, Shusen Wang, Yanxia Shi, Peng Liu, Qiufan Zheng, Jun Tang, Xin An, Jiajia Huang, Meiting Chen, Yongyi Zhong, Xiwen Bi, Yi Yang, Cong Xue","doi":"10.1093/oncolo/oyaf403","DOIUrl":"10.1093/oncolo/oyaf403","url":null,"abstract":"<p><strong>Background: </strong>Limited data are available on estradiol (E2) levels during fulvestrant treatment in women with hormone receptor-positive breast cancer.</p><p><strong>Methods: </strong>We measured plasma E2 levels in women receiving fulvestrant using liquid chromatography-tandem mass spectrometry. Patient characteristics and treatment efficacy were assessed in relation to E2 levels. A cutoff of 2.72 pg/mL was used because it defines E2 suppression and postmenopausal status.</p><p><strong>Results: </strong>A total of 69 women were enrolled, with a median age of 48 years. The median duration of fulvestrant treatment was 11.6 months. The median E2 level across the cohort was 3.60 pg/mL, with considerable interindividual variability (range: 1.11-526.13 pg/mL), and 49 women (71.0%) had E2 levels above 2.72 pg/mL. Eleven women (15.9%) had premenopausal E2 levels (>10 pg/mL). During a median follow-up period of 8.4 months, there was no statistically significant difference in progression-free survival (PFS) between women with E2 levels >2.72 pg/mL and those with E2 levels ≤2.72 pg/mL (P = .391). However, among women who benefited from first- or second-line fulvestrant therapy (PFS > 6 months), those with E2 levels >2.72 pg/mL exhibited significantly poorer PFS compared to those with E2 levels ≤2.72 pg/mL (P = .043).</p><p><strong>Conclusions: </strong>These findings support the need for E2 monitoring in women receiving fulvestrant therapy to better assess E2 status and its association with treatment efficacy.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12715405/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145687964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Giandomenico Roviello, Elisabetta Gambale, Irene De Gennaro Aquino, Marco Maruzzo, Carlo Messina, Ismaela Anna Vascotto, Virginia Rossi, Davide Bimbatti, Elisa Erbetta, Marco Messina, Alessia Mennitto, Sara Elena Rebuzzi, Cecilia Nasso, Chiara Mercinelli, Brigida Anna Maiorano, Martina Fanelli, Mariella Sorarù, Federico Scolari, Marinella Micol Mela, Luca Galli, Alessia Salfi, Mimma Rizzo, Silvia Puglisi, Valentina Orlando, Giuseppe Fornarini, Alessandro Rametta, Patrizia Giannatempo, Linda Cerbone, Laura Doni, Serena Pillozzi, Lorenzo Antonuzzo
Importance: Avelumab maintenance therapy improves survival in patients with advanced urothelial carcinoma who respond to first-line platinum-based chemotherapy. However, real-world evidence on its effectiveness and on the prognostic value of baseline clinical factors remains limited.
Objective: To evaluate the real-world effectiveness of Avelumab maintenance therapy and to identify baseline prognostic factors associated with clinical outcomes.
Design: Multicenter prospective observational study including a retrospective cohort of patients treated before study initiation.
Setting: Several Italian oncology centers participating between 2021 and 2023.
Participants: A total of 251 patients with advanced or metastatic urothelial carcinoma who received Avelumab maintenance therapy after achieving disease control with first-line platinum-based chemotherapy.
Intervention(s) or exposure(s): Avelumab maintenance therapy administered according to clinical practice. Baseline clinical variables, including ECOG performance status, metastatic sites, and corticosteroid use, were assessed for prognostic significance.
Main outcome(s) and measure(s): Primary outcomes were overall survival (OS) and progression-free survival (PFS). Secondary outcomes included overall response rate (ORR) and disease control rate (DCR). Prognostic factors were evaluated using multivariate Cox regression.
Results: Median OS was 22.4 months, and median PFS was 7 months. The ORR and DCR were 26.75% and 69.30%, respectively. Independent predictors of worse OS were ECOG ≥1 (HR 1.57), bone metastases (HR 1.88), brain metastases (HR 7.02), and baseline corticosteroid use (HR 2.42). An exploratory prognostic score integrating ECOG status, bone metastases, and steroid use stratified patients into three groups with significantly different outcomes: median OS was 39.9 months (no adverse factors), 14.1 months (one factor), and 8.4 months (two or more factors), with corresponding ORRs of 33.6%, 28.0%, and 6.9%.
Conclusions and relevance: In this large real-world cohort, Avelumab maintenance confirmed its effectiveness and safety in advanced urothelial carcinoma. A simple exploratory prognostic score based on ECOG performance status, bone metastases, and baseline corticosteroid use successfully stratified patients by survival outcomes and may support personalized treatment strategies in clinical practice.
背景:Avelumab维持治疗已证明对一线铂基化疗有反应的晚期尿路上皮癌患者的生存获益。然而,关于其有效性和基线临床因素的预后价值的实际数据仍然有限。方法:这是一项多中心前瞻性观察性研究,包括在研究开始前接受治疗的患者的回顾性队列,包括251例晚期或转移性尿路上皮癌患者,这些患者在2021年至2023年间接受Avelumab作为维持治疗。主要终点是总生存期(OS)和无进展生存期(PFS);次要终点包括总缓解率(ORR)和疾病控制率(DCR)。结果:中位OS为22.4个月,中位PFS为7个月。ORR和DCR分别为26.75%和69.30%。多因素分析发现ECOG≥1 (HR 1.57)、骨转移(HR 1.88)、脑转移(HR 7.02)和基线类固醇使用(HR 2.42)是较差OS的独立预测因素。ECOG表现状态≥1、骨转移的存在和基线时皮质类固醇的使用被确定为独立的预后因素,并被用于构建三个预后组的探索性分层。无、1个或2个或更多负面因素的患者有显著不同的OS(中位数:39.9 vs. 14.1 vs. 8.4个月)和ORR (33.6% vs. 28.0% vs. 6.9%)。结论:在这个真实世界的队列中,Avelumab维持证实了其治疗晚期尿路上皮癌的有效性和安全性。基于ECOG状态、骨转移和基线类固醇使用的简单探索性预后分层评分有效地根据预后对患者进行分层,并可能支持个性化治疗策略。
{"title":"Real-world effectiveness of Avelumab maintenance in advanced urothelial carcinoma: results from the Italian multicenter MALVA study (Meet-URO 25).","authors":"Giandomenico Roviello, Elisabetta Gambale, Irene De Gennaro Aquino, Marco Maruzzo, Carlo Messina, Ismaela Anna Vascotto, Virginia Rossi, Davide Bimbatti, Elisa Erbetta, Marco Messina, Alessia Mennitto, Sara Elena Rebuzzi, Cecilia Nasso, Chiara Mercinelli, Brigida Anna Maiorano, Martina Fanelli, Mariella Sorarù, Federico Scolari, Marinella Micol Mela, Luca Galli, Alessia Salfi, Mimma Rizzo, Silvia Puglisi, Valentina Orlando, Giuseppe Fornarini, Alessandro Rametta, Patrizia Giannatempo, Linda Cerbone, Laura Doni, Serena Pillozzi, Lorenzo Antonuzzo","doi":"10.1093/oncolo/oyaf388","DOIUrl":"10.1093/oncolo/oyaf388","url":null,"abstract":"<p><strong>Importance: </strong>Avelumab maintenance therapy improves survival in patients with advanced urothelial carcinoma who respond to first-line platinum-based chemotherapy. However, real-world evidence on its effectiveness and on the prognostic value of baseline clinical factors remains limited.</p><p><strong>Objective: </strong>To evaluate the real-world effectiveness of Avelumab maintenance therapy and to identify baseline prognostic factors associated with clinical outcomes.</p><p><strong>Design: </strong>Multicenter prospective observational study including a retrospective cohort of patients treated before study initiation.</p><p><strong>Setting: </strong>Several Italian oncology centers participating between 2021 and 2023.</p><p><strong>Participants: </strong>A total of 251 patients with advanced or metastatic urothelial carcinoma who received Avelumab maintenance therapy after achieving disease control with first-line platinum-based chemotherapy.</p><p><strong>Intervention(s) or exposure(s): </strong>Avelumab maintenance therapy administered according to clinical practice. Baseline clinical variables, including ECOG performance status, metastatic sites, and corticosteroid use, were assessed for prognostic significance.</p><p><strong>Main outcome(s) and measure(s): </strong>Primary outcomes were overall survival (OS) and progression-free survival (PFS). Secondary outcomes included overall response rate (ORR) and disease control rate (DCR). Prognostic factors were evaluated using multivariate Cox regression.</p><p><strong>Results: </strong>Median OS was 22.4 months, and median PFS was 7 months. The ORR and DCR were 26.75% and 69.30%, respectively. Independent predictors of worse OS were ECOG ≥1 (HR 1.57), bone metastases (HR 1.88), brain metastases (HR 7.02), and baseline corticosteroid use (HR 2.42). An exploratory prognostic score integrating ECOG status, bone metastases, and steroid use stratified patients into three groups with significantly different outcomes: median OS was 39.9 months (no adverse factors), 14.1 months (one factor), and 8.4 months (two or more factors), with corresponding ORRs of 33.6%, 28.0%, and 6.9%.</p><p><strong>Conclusions and relevance: </strong>In this large real-world cohort, Avelumab maintenance confirmed its effectiveness and safety in advanced urothelial carcinoma. A simple exploratory prognostic score based on ECOG performance status, bone metastases, and baseline corticosteroid use successfully stratified patients by survival outcomes and may support personalized treatment strategies in clinical practice.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12680435/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145566423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kuei-Ting Chen, Vidyalakshmi Sethunath, Ericka Ebot, Brennan Decker, Amaya Gasco, Jeffrey S Ross, Andreas M Heilmann, Serenedy Smith, Katherine Lofgren, Richard S P Huang
Introduction: FoundationOne®CDx (F1CDx) and Oncomine Dx target test (ODxTT) are commercially available oncology biomarker assays that apply different next-generation sequencing methodologies used to identify predictive markers for therapy selection in non-small cell lung cancer (NSCLC). We hypothesized that F1CDx can detect a higher number of actionable alterations compared with the ODxTT assay.
Materials and methods: We compared the prevalence of genomic alterations (GA) in a real-world NSCLC cohort tested by F1CDx to the theoretical detection by ODxTT, based on publicly available coverage specifications. The clinical actionability of GA was assessed based on NCCN treatment guidelines and FDA oncology drug approvals.
Results: KRAS G12C, EGFR mutations, and MET mutations were among the most frequent actionable alterations detected in a large genomic dataset of NSCLC data assayed by the F1CDx test. The ODxTT specifications indicate that ODxTT detects all BRAF and KRAS FDA-approved alterations detected by F1CDx and subsets of the following clinically significant markers: EGFR 83%, RET 76%, ROS1 74%, and ERBB2 38%. In addition, ODxTT had no coverage for actionable genomic findings in ALK, MET, NTRK1/2/3, TMB, or MSI-High status. In this study, the ODxTT assay may fail to report at least one of the clinically actionable biomarkers in 42% of tissue samples compared to F1CDx.
Discussion: In summary, in advanced NSCLC, hybrid capture-based NGS, such as F1CDx, may capture more actionable genomic alterations as compared with ODxTT, an amplicon-based NGS.
{"title":"Coverage of actionable alterations in non-small cell lung cancer by hybrid capture-based FoundationOne®CDx compared with amplicon-based OncomineDX.","authors":"Kuei-Ting Chen, Vidyalakshmi Sethunath, Ericka Ebot, Brennan Decker, Amaya Gasco, Jeffrey S Ross, Andreas M Heilmann, Serenedy Smith, Katherine Lofgren, Richard S P Huang","doi":"10.1093/oncolo/oyaf390","DOIUrl":"10.1093/oncolo/oyaf390","url":null,"abstract":"<p><strong>Introduction: </strong>FoundationOne®CDx (F1CDx) and Oncomine Dx target test (ODxTT) are commercially available oncology biomarker assays that apply different next-generation sequencing methodologies used to identify predictive markers for therapy selection in non-small cell lung cancer (NSCLC). We hypothesized that F1CDx can detect a higher number of actionable alterations compared with the ODxTT assay.</p><p><strong>Materials and methods: </strong>We compared the prevalence of genomic alterations (GA) in a real-world NSCLC cohort tested by F1CDx to the theoretical detection by ODxTT, based on publicly available coverage specifications. The clinical actionability of GA was assessed based on NCCN treatment guidelines and FDA oncology drug approvals.</p><p><strong>Results: </strong>KRAS G12C, EGFR mutations, and MET mutations were among the most frequent actionable alterations detected in a large genomic dataset of NSCLC data assayed by the F1CDx test. The ODxTT specifications indicate that ODxTT detects all BRAF and KRAS FDA-approved alterations detected by F1CDx and subsets of the following clinically significant markers: EGFR 83%, RET 76%, ROS1 74%, and ERBB2 38%. In addition, ODxTT had no coverage for actionable genomic findings in ALK, MET, NTRK1/2/3, TMB, or MSI-High status. In this study, the ODxTT assay may fail to report at least one of the clinically actionable biomarkers in 42% of tissue samples compared to F1CDx.</p><p><strong>Discussion: </strong>In summary, in advanced NSCLC, hybrid capture-based NGS, such as F1CDx, may capture more actionable genomic alterations as compared with ODxTT, an amplicon-based NGS.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12687587/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145566299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fares Jamal, Oudai Sahvan, Pedro Luiz Serrano Uson Junior, Jeremy C Jones, Fang-Shu Ou, Tanios Bekaii-Saab, Mohamad Bassam Sonbol
Background: The increasing reliance on accelerated approvals and surrogate endpoints for Food and Drug Administration (FDA) approvals of gastrointestinal (GI) cancer therapies raises concerns about their clinical benefit and long-term patient outcomes. The shift toward single-arm trials in regulatory decisions further complicates treatment evaluation.
Material and methods: This retrospective observational study evaluated all FDA approvals for GI cancer therapies from January 2006 through January 2025. Data were extracted from FDA archives, ClinicalTrials.gov, and PubMed. Approvals were categorized by regulatory pathway (accelerated vs regular), trial design (single-arm vs randomized), and primary endpoint (surrogate vs overall survival [OS]). Clinical benefit was assessed based on OS improvement and the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). The primary outcome was the proportion of approvals based on surrogate versus OS endpoints. Secondary outcomes included use of single-arm designs, frequency of accelerated approvals, OS gain, and the proportion meeting ESMO-MCBS substantial benefit (score ≥ 4).
Results: The FDA granted 60 GI cancer drug approvals from 67 trials. Approvals rose from 15 (25%) in 2006-2014 to 45 (75%) in 2015-2025. Single-arm trials increased to 24%, and surrogate endpoints were used in 41.8% (ORR 20.9%, PFS 19.4%). Median OS improvement was 2.1 months (IQR: 1.6-2.65). Only 24.3% of trials met ESMO-MCBS substantial benefit. Of 15 accelerated approvals, 66.7% remained pending, 13.3% received full approval, and 20% were withdrawn.
Conclusion: Expedited approvals have improved drug access in GI oncology, but modest benefits highlight the need to balance speed with outcomes that truly matter to patients.
{"title":"Evaluating Food and Drug Administration approved gastrointestinal cancer drugs: clinical benefit and trial endpoints over the past two decades.","authors":"Fares Jamal, Oudai Sahvan, Pedro Luiz Serrano Uson Junior, Jeremy C Jones, Fang-Shu Ou, Tanios Bekaii-Saab, Mohamad Bassam Sonbol","doi":"10.1093/oncolo/oyaf381","DOIUrl":"10.1093/oncolo/oyaf381","url":null,"abstract":"<p><strong>Background: </strong>The increasing reliance on accelerated approvals and surrogate endpoints for Food and Drug Administration (FDA) approvals of gastrointestinal (GI) cancer therapies raises concerns about their clinical benefit and long-term patient outcomes. The shift toward single-arm trials in regulatory decisions further complicates treatment evaluation.</p><p><strong>Material and methods: </strong>This retrospective observational study evaluated all FDA approvals for GI cancer therapies from January 2006 through January 2025. Data were extracted from FDA archives, ClinicalTrials.gov, and PubMed. Approvals were categorized by regulatory pathway (accelerated vs regular), trial design (single-arm vs randomized), and primary endpoint (surrogate vs overall survival [OS]). Clinical benefit was assessed based on OS improvement and the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). The primary outcome was the proportion of approvals based on surrogate versus OS endpoints. Secondary outcomes included use of single-arm designs, frequency of accelerated approvals, OS gain, and the proportion meeting ESMO-MCBS substantial benefit (score ≥ 4).</p><p><strong>Results: </strong>The FDA granted 60 GI cancer drug approvals from 67 trials. Approvals rose from 15 (25%) in 2006-2014 to 45 (75%) in 2015-2025. Single-arm trials increased to 24%, and surrogate endpoints were used in 41.8% (ORR 20.9%, PFS 19.4%). Median OS improvement was 2.1 months (IQR: 1.6-2.65). Only 24.3% of trials met ESMO-MCBS substantial benefit. Of 15 accelerated approvals, 66.7% remained pending, 13.3% received full approval, and 20% were withdrawn.</p><p><strong>Conclusion: </strong>Expedited approvals have improved drug access in GI oncology, but modest benefits highlight the need to balance speed with outcomes that truly matter to patients.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12693570/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145702831","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fabrizio Di Costanzo, Chiara Mercinelli, Alessio Signori, Carlo Messina, Vincenza Conteduca, Giovanni Dima, Matteo Santoni, Luigi Formisano, Christoph Oing, Giuseppe Fornarini, Sara Elena Rebuzzi, Orazio Caffo, Giuseppe Luigi Banna, Ugo De Giorgi, Giuseppe Procopio, Francesco Montorsi, Alberto Briganti, Luca Galli, Massimo Di Maio, Andrea Necchi, Brigida Anna Maiorano, Pasquale Rescigno
Context: With new treatment strategies approved in metastatic hormone-sensitive prostate cancer (mHSPC), heterogeneity across trials hinders the physicians' choice for first-line treatment.
Objective: We conducted a systematic review and network meta-analysis to assess the efficacy of currently approved treatments for mHSPC stratifying patients according to their disease burden (high- vs. low-volume as per CHAARTED criteria) and onset of metastatic disease (synchronous vs. metachronous).
Intervention: Eleven randomized controlled trials (RCTs) published until October 30th, 2024 were included. Treatment regimens were grouped as Triplets for combinations of docetaxel, androgen receptor pathway inhibitors (ARPIs) and androgen-deprivation therapy (ADT), separate Doublets for docetaxel plus ADT, ARPI plus ADT, or Monotherapy for ADT alone.
Outcome measurements and statistical analysis: Overall survival (OS) and radiographic progression-free survival (rPFS) outcomes were collected. OS as primary endpoint, and rPFS as secondary endpoint, were analyzed separately in high-volume and low-volume patients. Additional subgroup analyses accounted for timing of metastases categorized as high-volume/synchronous, high-volume/metachronous, low-volume/synchronous and low-volume/metachronous disease.
Evidence synthesis: Triplet combinations prolonged significantly OS and rPFS in high-volume disease (P-score 0.99), and high-volume/synchronous disease (P-score 0.99). ARPI/ADT doublets performed best in low-volume patients (P-score 0.94), and low-volume/metachronous (P-score 0.99). In the high-volume/metachronous population, triplets and doublets were equally effective.
Conclusions: The results provide collective evidence for treatment selection based on disease volume and timing of metastasis with strongest survival benefits of triplets for high-volume/synchronous mHSPC patients and of ARPI doublets for low-volume disease.
{"title":"Best therapeutic approach in metastatic hormone-sensitive prostate cancer based on disease volume: a systematic review and network meta-analysis.","authors":"Fabrizio Di Costanzo, Chiara Mercinelli, Alessio Signori, Carlo Messina, Vincenza Conteduca, Giovanni Dima, Matteo Santoni, Luigi Formisano, Christoph Oing, Giuseppe Fornarini, Sara Elena Rebuzzi, Orazio Caffo, Giuseppe Luigi Banna, Ugo De Giorgi, Giuseppe Procopio, Francesco Montorsi, Alberto Briganti, Luca Galli, Massimo Di Maio, Andrea Necchi, Brigida Anna Maiorano, Pasquale Rescigno","doi":"10.1093/oncolo/oyaf386","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf386","url":null,"abstract":"<p><strong>Context: </strong>With new treatment strategies approved in metastatic hormone-sensitive prostate cancer (mHSPC), heterogeneity across trials hinders the physicians' choice for first-line treatment.</p><p><strong>Objective: </strong>We conducted a systematic review and network meta-analysis to assess the efficacy of currently approved treatments for mHSPC stratifying patients according to their disease burden (high- vs. low-volume as per CHAARTED criteria) and onset of metastatic disease (synchronous vs. metachronous).</p><p><strong>Intervention: </strong>Eleven randomized controlled trials (RCTs) published until October 30th, 2024 were included. Treatment regimens were grouped as Triplets for combinations of docetaxel, androgen receptor pathway inhibitors (ARPIs) and androgen-deprivation therapy (ADT), separate Doublets for docetaxel plus ADT, ARPI plus ADT, or Monotherapy for ADT alone.</p><p><strong>Outcome measurements and statistical analysis: </strong>Overall survival (OS) and radiographic progression-free survival (rPFS) outcomes were collected. OS as primary endpoint, and rPFS as secondary endpoint, were analyzed separately in high-volume and low-volume patients. Additional subgroup analyses accounted for timing of metastases categorized as high-volume/synchronous, high-volume/metachronous, low-volume/synchronous and low-volume/metachronous disease.</p><p><strong>Evidence synthesis: </strong>Triplet combinations prolonged significantly OS and rPFS in high-volume disease (P-score 0.99), and high-volume/synchronous disease (P-score 0.99). ARPI/ADT doublets performed best in low-volume patients (P-score 0.94), and low-volume/metachronous (P-score 0.99). In the high-volume/metachronous population, triplets and doublets were equally effective.</p><p><strong>Conclusions: </strong>The results provide collective evidence for treatment selection based on disease volume and timing of metastasis with strongest survival benefits of triplets for high-volume/synchronous mHSPC patients and of ARPI doublets for low-volume disease.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145543931","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Brandon DaSilva, Alfred I Neugut, Tito Fojo, Susan E Bates
{"title":"Highlights of ASCO 2025.","authors":"Brandon DaSilva, Alfred I Neugut, Tito Fojo, Susan E Bates","doi":"10.1093/oncolo/oyaf336","DOIUrl":"10.1093/oncolo/oyaf336","url":null,"abstract":"","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605717/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145245629","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hermioni L Amonoo, Joely Centracchio, Claire Greydanus, Mitchell W Lavoie, Emma P Keane, Joseph A Greer, Elizabeth C Lindenberger, Keri Brenner, Nneka N Ufere, Lara Traeger, Jennifer S Temel, Elyse R Park, Vicki Jackson, Areej El-Jawahri
Introduction: Prior studies have shown mixed findings regarding the relationship between prognostic awareness with quality of life (QOL) and psychological distress in patients with advanced cancer. Prognostic awareness is a multidimensional construct including a cognitive component (ie, the ability to understand one's life-limiting illness) and an emotional coping component (ie, the capacity to process terminal prognosis). Yet, it remains unclear which domains of prognostic awareness are associated with QOL and psychological distress.
Materials and methods: We conducted a cross-sectional study of adults with metastatic solid tumors treated with noncurative intent at a single academic center from 11/2019 to 6/2022. We used the Prognostic Awareness Impact Scale (PAIS) to measure components of prognostic awareness, including the cognitive and emotional coping components. We used the Functional Assessment of Cancer Therapy-G and Hospital Anxiety and Depression Scale to measure QOL and psychological distress, respectively. Linear regression models were used to examine the relationship between the PAIS domains and patient-reported outcomes.
Results: We enrolled 61.9% (632/1021) of eligible patients. Cognitive understanding of prognosis was not associated with QOL (B = -2.3, P = .114), anxiety (B = 0.7, P = .057), or depression symptoms (B = 0.4, P = .293). However, higher emotional coping with prognosis was associated with better QOL (B = 1.7; P < .001), lower anxiety (B = -0.6; P < .001), and depression (B = -0.3; P < .001).
Conclusion: Patients' emotional coping with their prognosis, rather than cognitive acknowledgment of their incurable cancer, was associated with QOL and psychological distress. Our findings underscore patients' ability to tolerate an accurate understanding of their prognosis and the critical need to incorporate effective coping strategies during prognostic discussions.
先前的研究显示,关于预后意识与晚期癌症患者生活质量(QOL)和心理困扰之间的关系,研究结果好坏参半。预后意识是一个多维度的结构,包括认知成分(即了解限制生命的疾病的能力)和情感应对成分(即处理临终预后的能力)。然而,目前尚不清楚哪些领域的预后意识与生活质量和心理困扰有关。材料和方法:我们于2019年11月至2022年6月在一个学术中心对非治愈性转移性实体瘤进行了一项横断面研究。我们使用预后意识影响量表(PAIS)测量预后意识的组成部分,包括认知和情绪应对组成部分。我们采用肿瘤治疗功能评估- g和医院焦虑抑郁量表分别测量生活质量和心理困扰。线性回归模型用于检验PAIS域与患者报告结果之间的关系。结果:61.9%(632/1021)的符合条件的患者入组。对预后的认知认知与生活质量(B=-2.3, P = 0.114)、焦虑(B= 0.7, P = 0.057)、抑郁症状(B= 0.4, P = 0.293)无关。结论:患者对预后的情绪应对与生活质量和心理困扰相关,而非对无法治愈癌症的认知认知与生活质量和心理困扰相关。我们的研究结果强调了患者对其预后的准确理解以及在预后讨论中纳入有效应对策略的关键需要。
{"title":"Domains of prognostic awareness, quality of life, and psychological distress in patients with advanced cancer.","authors":"Hermioni L Amonoo, Joely Centracchio, Claire Greydanus, Mitchell W Lavoie, Emma P Keane, Joseph A Greer, Elizabeth C Lindenberger, Keri Brenner, Nneka N Ufere, Lara Traeger, Jennifer S Temel, Elyse R Park, Vicki Jackson, Areej El-Jawahri","doi":"10.1093/oncolo/oyaf344","DOIUrl":"10.1093/oncolo/oyaf344","url":null,"abstract":"<p><strong>Introduction: </strong>Prior studies have shown mixed findings regarding the relationship between prognostic awareness with quality of life (QOL) and psychological distress in patients with advanced cancer. Prognostic awareness is a multidimensional construct including a cognitive component (ie, the ability to understand one's life-limiting illness) and an emotional coping component (ie, the capacity to process terminal prognosis). Yet, it remains unclear which domains of prognostic awareness are associated with QOL and psychological distress.</p><p><strong>Materials and methods: </strong>We conducted a cross-sectional study of adults with metastatic solid tumors treated with noncurative intent at a single academic center from 11/2019 to 6/2022. We used the Prognostic Awareness Impact Scale (PAIS) to measure components of prognostic awareness, including the cognitive and emotional coping components. We used the Functional Assessment of Cancer Therapy-G and Hospital Anxiety and Depression Scale to measure QOL and psychological distress, respectively. Linear regression models were used to examine the relationship between the PAIS domains and patient-reported outcomes.</p><p><strong>Results: </strong>We enrolled 61.9% (632/1021) of eligible patients. Cognitive understanding of prognosis was not associated with QOL (B = -2.3, P = .114), anxiety (B = 0.7, P = .057), or depression symptoms (B = 0.4, P = .293). However, higher emotional coping with prognosis was associated with better QOL (B = 1.7; P < .001), lower anxiety (B = -0.6; P < .001), and depression (B = -0.3; P < .001).</p><p><strong>Conclusion: </strong>Patients' emotional coping with their prognosis, rather than cognitive acknowledgment of their incurable cancer, was associated with QOL and psychological distress. Our findings underscore patients' ability to tolerate an accurate understanding of their prognosis and the critical need to incorporate effective coping strategies during prognostic discussions.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605813/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145276787","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cindy Y Jiang, Hyunsoo Hwang, Ilana Y Epstein, Dimitra Rafailia Bakaloudi, Rafee Talukder, Amy K Taylor, Amanda Nizam, Tanya Jindal, Michael J Glover, Ali Raza Khaki, Pedro C Barata, Charles B Nguyen, Eugene Oh, Nancy B Davis, Hannah Mabey, Christopher J Hoimes, Sean T Evans, Bashar Abuqayas, Emily Lemke, Irene Tsung, Wei Qiao, Deepak Kilari, Yousef Zakharia, Mehmet A Bilen, Matthew I Milowsky, Sumit A Shah, Shilpa Gupta, Hamid Emamekhoo, Joaquim Bellmunt, Ajjai S Alva, Petros Grivas, Pavlos Msaouel, Vadim S Koshkin, Matthew T Campbell, Omar Alhalabi
Background: Erdafitinib is approved for locally advanced/metastatic urothelial cancer (LA/mUC). As enfortumab vedotin (EV) plus pembrolizumab enters frontline management, outcomes with erdafitinib pre- and post-EV are clinically relevant but not specifically evaluated in clinical trials.
Methods: UNITE is a multi-institutional retrospective study of patients with LA/mUC treated with novel targeted agents. All patients with FGFR3 alterations treated with EV only, erdafitinib then EV (Erda->EV), and EV then erdafitinib (EV->Erda) were included. Sequential treatment with EV and Erda was not required. Primary endpoints were observed response rates (ORR) and progression-free survival (PFS); secondary endpoint was overall survival (OS).
Results: We identified 83 patients with FGFR3 alterations and separated them into three cohorts: EV only (n = 44), Erda->EV (n = 24), and EV->Erda (n = 15). Most (72%) received ≥2 lines of therapy before erdafitinib (checkpoint inhibitor [87%], platinum-based chemotherapy [64%]). Median PFS with erdafitinib for EV-naïve cohort was 7.5 months and in EV-treated cohort 4.0 months (HR 0.78; 95% CI 0.35-1.7). ORR with erdafitinib for EV-naïve was 33% and EV-treated 31% (OR 1.1; 95% CI 0.29-4.1). Median PFS with EV for patients who were erdafitinib-naïve was 6 months and in erdafitinib-treated 5.3 months (HR 0.61; 95% CI 0.34-1.09). ORR with EV was 54% in erdafitinib-naïve cohort and 32% in erdafitinib-treated cohort (OR 2.5; 95% CI 0.87-6.3).
Conclusion: In patients with FGFR3-altered LA/mUC, erdafitinib is active pre- and post-EV. Outcomes with erdafitinib were consistent with clinical trial data generated prior to broader frontline use of EV. Findings are hypothesis-generating and given small sample size should be interpreted with caution.
{"title":"Efficacy of erdafitinib before or after enfortumab vedotin in FGFR3-altered advanced urothelial cancer: analysis of the UNITE collaborative study.","authors":"Cindy Y Jiang, Hyunsoo Hwang, Ilana Y Epstein, Dimitra Rafailia Bakaloudi, Rafee Talukder, Amy K Taylor, Amanda Nizam, Tanya Jindal, Michael J Glover, Ali Raza Khaki, Pedro C Barata, Charles B Nguyen, Eugene Oh, Nancy B Davis, Hannah Mabey, Christopher J Hoimes, Sean T Evans, Bashar Abuqayas, Emily Lemke, Irene Tsung, Wei Qiao, Deepak Kilari, Yousef Zakharia, Mehmet A Bilen, Matthew I Milowsky, Sumit A Shah, Shilpa Gupta, Hamid Emamekhoo, Joaquim Bellmunt, Ajjai S Alva, Petros Grivas, Pavlos Msaouel, Vadim S Koshkin, Matthew T Campbell, Omar Alhalabi","doi":"10.1093/oncolo/oyaf342","DOIUrl":"10.1093/oncolo/oyaf342","url":null,"abstract":"<p><strong>Background: </strong>Erdafitinib is approved for locally advanced/metastatic urothelial cancer (LA/mUC). As enfortumab vedotin (EV) plus pembrolizumab enters frontline management, outcomes with erdafitinib pre- and post-EV are clinically relevant but not specifically evaluated in clinical trials.</p><p><strong>Methods: </strong>UNITE is a multi-institutional retrospective study of patients with LA/mUC treated with novel targeted agents. All patients with FGFR3 alterations treated with EV only, erdafitinib then EV (Erda->EV), and EV then erdafitinib (EV->Erda) were included. Sequential treatment with EV and Erda was not required. Primary endpoints were observed response rates (ORR) and progression-free survival (PFS); secondary endpoint was overall survival (OS).</p><p><strong>Results: </strong>We identified 83 patients with FGFR3 alterations and separated them into three cohorts: EV only (n = 44), Erda->EV (n = 24), and EV->Erda (n = 15). Most (72%) received ≥2 lines of therapy before erdafitinib (checkpoint inhibitor [87%], platinum-based chemotherapy [64%]). Median PFS with erdafitinib for EV-naïve cohort was 7.5 months and in EV-treated cohort 4.0 months (HR 0.78; 95% CI 0.35-1.7). ORR with erdafitinib for EV-naïve was 33% and EV-treated 31% (OR 1.1; 95% CI 0.29-4.1). Median PFS with EV for patients who were erdafitinib-naïve was 6 months and in erdafitinib-treated 5.3 months (HR 0.61; 95% CI 0.34-1.09). ORR with EV was 54% in erdafitinib-naïve cohort and 32% in erdafitinib-treated cohort (OR 2.5; 95% CI 0.87-6.3).</p><p><strong>Conclusion: </strong>In patients with FGFR3-altered LA/mUC, erdafitinib is active pre- and post-EV. Outcomes with erdafitinib were consistent with clinical trial data generated prior to broader frontline use of EV. Findings are hypothesis-generating and given small sample size should be interpreted with caution.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605803/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145245590","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Justine Panian, Nicholas C Henderson, Daniel Herchenhorn, Pedro C Barata, Mehmet Asim Bilen, Laura Graham, Elisabeth Heath, Clara Hwang, Avery Supernois, Deepak Kilari, Bicky Thapa, Vadim S Koshkin, Tanya Jindal, Jones T Nauseef, Alexandra Sokolova, Taylor Amery, Yousef Zakharia, Michael T Schweizer, Ruben Raychaudhuri, Zachery R Reichert, Tanya Dorff, Andrew J Armstrong, John Wang, Ajjai Alva, Rana R McKay
Background: 177Lu-PSMA-617 is approved for patients with metastatic castration-resistant prostate cancer (mCRPC). Although treatment is associated with improved outcomes, not all patients benefit and response is heterogeneous. We aim to characterize genomic alterations associated with benefit to 177Lu-PSMA-617.
Materials and methods: This study used the Prostate Cancer Precision Medicine Multi-Institutional Collaborative Effort (PROMISE) clinical-genomic database (n = 2445). The primary endpoint was ≥50% PSA decline (PSA50) from baseline with 177Lu-PSMA-617 in molecular subgroups. Secondary endpoints included 90% PSA decline (PSA90). Associations were assessed using Fisher's exact test and Cox regression in multivariable analysis.
Results: We identified 183 mCRPC patients treated with 177Lu-PSMA-617. Median number of prior lines of mCRPC therapy was 3. Overall, PSA50 was 49%, median progression-free survival was 7.6 months, and median overall survival was 13.9 months. NF1 (n = 8) and FOXA1 alterations (n = 5) were associated with increased PSA50 (88% vs 47%, P = .03 for NF1; 100% vs 47%, P = .03 for FOXA1). Among CRPC sequenced tumors (n = 119), androgen receptor (AR) alterations (n = 58) were associated with lower PSA50 (38% vs 60%, P = .03). While any tumor suppressor genes (TSG) (PTEN, TP53, RB1) (n = 109) or TP53 (n = 83) alteration were associated with lower PSA90 (P = .02 for both), NF1 (n = 8), and FOXA1 alterations were associated with higher PSA90 (P = .03 and P = .003, respectively).
Conclusions: This analysis identifies potential genomic predictors of response to 177Lu-PSMA-617, with NF1 and FOXA1 alterations associated with favorable outcomes and AR and TSG alterations with diminished response. These hypothesis-generating findings suggest genomic profiling may inform selection for PSMA-targeted therapy and warrant prospective validation in larger cohorts.
{"title":"Genomic alterations and associated outcomes in patients with PSMA-positive metastatic castration-resistant prostate cancer treated with 177Lu-PSMA-617.","authors":"Justine Panian, Nicholas C Henderson, Daniel Herchenhorn, Pedro C Barata, Mehmet Asim Bilen, Laura Graham, Elisabeth Heath, Clara Hwang, Avery Supernois, Deepak Kilari, Bicky Thapa, Vadim S Koshkin, Tanya Jindal, Jones T Nauseef, Alexandra Sokolova, Taylor Amery, Yousef Zakharia, Michael T Schweizer, Ruben Raychaudhuri, Zachery R Reichert, Tanya Dorff, Andrew J Armstrong, John Wang, Ajjai Alva, Rana R McKay","doi":"10.1093/oncolo/oyaf358","DOIUrl":"10.1093/oncolo/oyaf358","url":null,"abstract":"<p><strong>Background: </strong>177Lu-PSMA-617 is approved for patients with metastatic castration-resistant prostate cancer (mCRPC). Although treatment is associated with improved outcomes, not all patients benefit and response is heterogeneous. We aim to characterize genomic alterations associated with benefit to 177Lu-PSMA-617.</p><p><strong>Materials and methods: </strong>This study used the Prostate Cancer Precision Medicine Multi-Institutional Collaborative Effort (PROMISE) clinical-genomic database (n = 2445). The primary endpoint was ≥50% PSA decline (PSA50) from baseline with 177Lu-PSMA-617 in molecular subgroups. Secondary endpoints included 90% PSA decline (PSA90). Associations were assessed using Fisher's exact test and Cox regression in multivariable analysis.</p><p><strong>Results: </strong>We identified 183 mCRPC patients treated with 177Lu-PSMA-617. Median number of prior lines of mCRPC therapy was 3. Overall, PSA50 was 49%, median progression-free survival was 7.6 months, and median overall survival was 13.9 months. NF1 (n = 8) and FOXA1 alterations (n = 5) were associated with increased PSA50 (88% vs 47%, P = .03 for NF1; 100% vs 47%, P = .03 for FOXA1). Among CRPC sequenced tumors (n = 119), androgen receptor (AR) alterations (n = 58) were associated with lower PSA50 (38% vs 60%, P = .03). While any tumor suppressor genes (TSG) (PTEN, TP53, RB1) (n = 109) or TP53 (n = 83) alteration were associated with lower PSA90 (P = .02 for both), NF1 (n = 8), and FOXA1 alterations were associated with higher PSA90 (P = .03 and P = .003, respectively).</p><p><strong>Conclusions: </strong>This analysis identifies potential genomic predictors of response to 177Lu-PSMA-617, with NF1 and FOXA1 alterations associated with favorable outcomes and AR and TSG alterations with diminished response. These hypothesis-generating findings suggest genomic profiling may inform selection for PSMA-targeted therapy and warrant prospective validation in larger cohorts.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12622373/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145349998","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}