OncologistPub Date : 2025-10-07DOI: 10.1093/oncolo/oyaf342
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
{"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":"https://doi.org/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><p><strong>Implications for practice: </strong>Non-trial outcomes of erdafitinib in FGFR3-altered locally advanced/metastatic urothelial cancer are consistent with reported clinical trial data. Erdafitinib therapy is effective in the pre- and post- EV setting.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","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":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncologistPub Date : 2025-10-07DOI: 10.1093/oncolo/oyaf336
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":"https://doi.org/10.1093/oncolo/oyaf336","url":null,"abstract":"","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","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":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncologistPub Date : 2025-10-07DOI: 10.1093/oncolo/oyaf339
Fabio Salomone, Giorgia Novero, Oriana Ciani, Roberto Ferrara, Alberto Servetto, Narjust Florez, Massimo Di Maio, Gabriella Pravettoni, Cecilia Pompili
{"title":"Health-related Quality of Life assessment in trials testing Tyrosine Kinase Inhibitors or Immune Checkpoints Inhibitors in early-stage NSCLC.","authors":"Fabio Salomone, Giorgia Novero, Oriana Ciani, Roberto Ferrara, Alberto Servetto, Narjust Florez, Massimo Di Maio, Gabriella Pravettoni, Cecilia Pompili","doi":"10.1093/oncolo/oyaf339","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf339","url":null,"abstract":"<p><strong>Background: </strong>Health-related quality of life (HRQoL) remains underassessed and underreporting in randomized clinical trials (RCTs) evaluating new therapies in metastatic non-small cell lung cancer (NSCLC). However, evaluation and preservation of favorable HRQoL are critically important in trials including patients in early-stage settings, in which the primary objective is cure. Herein, we evaluated whether HRQoL was adequately evaluated and reported in trials including immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) in resectable NSCLC.</p><p><strong>Methods: </strong>A systematic search was performed on Embase and PubMed to identify RCTs testing TKIs or ICIs in resectable NSCLC. We selected full articles and abstracts from major meetings. Risk of bias and reporting assessment of HRQoL were collected.</p><p><strong>Results: </strong>As of October 2024, we identified 25 RCTs. The primary endpoint was overall survival for 2 RCTs, while 21 and 7 RCTs evaluated risk of recurrence and tumour response as (co)-primary endpoints, respectively. Twelve RCTs (48%) did not assess HRQoL as an endpoint, while 13 (52%) included HRQoL evaluation as a secondary or exploratory endpoint. The most common tools utilized were FACT-L (6/13; 46%), EORTC-QLQ30/LC13 (4/13; 30%) and SF-36 (2/13; 15%). Phase II (33%) and adjuvant (44%) trials evaluated HRQoL in a lower rate than phase III (62%) and neoadjuvant/perioperative (66%) RCTs. Three out of 22 RCTs (14%) with available full-texts reported HRQoL results in the primary publication. Two out of the 19 remaining RCTs reported HRQoL in an indipendent publications, and 2 of them presented data in meeting abstracts. Remarkably, for 15 (68%) RCTs HRQoL evaluation is not available.</p><p><strong>Conclusions: </strong>Our systematic evaluation revealed suboptimal evaluation and underreporting of HRQoL in patients treated with novel agents and combinations in resectable NSCLC. Systematic evaluation and reporting of HRQoL should be prioritized in future trials.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145245632","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}
OncologistPub Date : 2025-10-07DOI: 10.1093/oncolo/oyaf338
Rana R McKay, Shayan S Nazari, Andrew Elliott, Jennifer R Ribeiro, Brent S Rose, Pedro C Barata, Deepak Kilari, Rohan Garje, Neeraj Agarwal, Norm Smith, Emmanuel S Antonarakis, Aditya Bagrodia, Himisha Beltran
{"title":"Molecular and clinical correlates of high FOLH1 (PSMA) RNA expression in primary and metastatic prostate cancer.","authors":"Rana R McKay, Shayan S Nazari, Andrew Elliott, Jennifer R Ribeiro, Brent S Rose, Pedro C Barata, Deepak Kilari, Rohan Garje, Neeraj Agarwal, Norm Smith, Emmanuel S Antonarakis, Aditya Bagrodia, Himisha Beltran","doi":"10.1093/oncolo/oyaf338","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf338","url":null,"abstract":"<p><strong>Background: </strong>Prostate-specific membrane antigen (PSMA; FOLH1) is a cell-surface target for diagnostics and treatment in prostate cancer. We utilized a database of molecularly-profiled prostate tumors to evaluate clinical, genomic, and immunologic correlates of high FOLH1 RNA expression.</p><p><strong>Patients and methods: </strong>Prostate cancer specimens (N = 7,082) underwent DNA/RNA sequencing and immunohistochemistry at Caris Life Sciences. FOLH1-High/Low expression was defined by upper/lower quartiles or median transcripts per million (TPM). Overall survival (OS) was calculated from insurance claims.</p><p><strong>Results: </strong>Prostate adenocarcinoma had 2.97-fold higher FOLH1 expression compared to high grade neuroendocrine prostate cancer (q < 0.0001). Of 7,020 adenocarcinomas, 4,464 were primary prostate samples, 828 were lymph node metastases, and 1,686 were distant metastases. FOLH1 expression varied across metastatic sites (highest in lymph node and lowest in liver; 1.2-fold difference, q < 0.05). FOLH1-High tumors were enriched in AR-V7 variants (10.1% vs. 4.5%, q < 0.05) and associated with higher androgen receptor (AR) signaling (0.82 vs 0.78, q < 0.05). Conversely, FOLH1-Low tumors were enriched with FOXA1 (12.2% vs. 6.4%), APC (11.4% vs. 3.0%), PIK3CA (5.8% vs. 2.7%) and PIK3R1 (2.6% vs. 0.48%) mutations (q < 0.05). FOLH1-High tumors had a lower M1: M2 ratio, fewer Tregs and CD8+ T cells, higher immune checkpoint expression, and lower interferon signature scores. FOLH1-High primary tumors were more frequently microsatellite instability (MSI)-High, tumor mutational burden (TMB)-High, and PD-L1-positive. Among patients with metastatic tumors, median OS was improved in the FOLH1-High group (31.9 vs 23.3 months, p < 0.001).</p><p><strong>Conclusions: </strong>This enhanced understanding of the distinct molecular and clinical profiles of FOLH1-expressing prostate cancers may inform optimization of PSMA-directed treatments.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145245768","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}
OncologistPub Date : 2025-10-07DOI: 10.1093/oncolo/oyaf341
Haojie Zhou, Minzhi Lv, Wei Li, Yan Wang, Jing Wu, Qing Liu, Tianshu Liu, Qian Li
{"title":"Efficacy and safety of pyrotinib in patients with previously treated HER2-positive non-breast solid tumors: a phase 2, open-label basket trial.","authors":"Haojie Zhou, Minzhi Lv, Wei Li, Yan Wang, Jing Wu, Qing Liu, Tianshu Liu, Qian Li","doi":"10.1093/oncolo/oyaf341","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf341","url":null,"abstract":"<p><strong>Background: </strong>Amplification/overexpression of the human epidermal growth factor receptor 2 (HER2) gene drives cell proliferation, differentiation, and migration of breast cancer. However, HER2-targeted therapies are not standard treatment for HER2-positive non-breast solid tumors currently. This phase II open-label basket trial evaluated pyrotinib's efficacy and safety in patients with non-breast solid tumors.</p><p><strong>Methods: </strong>Patients with previously treated HER2-positive advanced non-breast solid tumors were enrolled at Zhongshan Hospital, Fudan University. All participants received pyrotinib in 21-day cycles. Primary endpoint was objective response rate (ORR) at 6 weeks, assessed per RECIST version 1.1. Circulating tumor DNA (ctDNA) analysis was conducted to identify biomarkers of efficiacy.</p><p><strong>Results: </strong>Fifty-three patients were enrolled and 51 evaluable for efficiacy analysis. The median follow-up was 32.2 months. ORR was 18.9% (95% CI: 10.2-31.7%) and disease control rate was 73.6% (95% CI: 60.1-84.2%). Median progression-free survival (mPFS) was 5.1 (95% CI: 4.2-8.1) months and median overall survival (mOS) was 17.2 (95% CI: 16.2-33.2) months. Patients with colorectal cancer had the highest ORR and the longest mOS (33.2 months, 95% CI: 14.9-51.5). Among patients with HER2 overexpression, those with immunohistochemistry 3+ had longer mPFS and mOS. Sixteen patients required dose reductions due to grade 3 adverse events (AEs); no ≥grade 4 AEs occurred. Analysis of ctDNA revealed that patients with progression disease (PD) had higher mutation frequency, more diverse mutational profiles, and higher copy number burden.</p><p><strong>Conclusion: </strong>Pyrotinib demonstrated a favorable safety profile and modest efficacy in patients with previously treated HER2-positive advanced non-breast solid tumors.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145245494","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}
{"title":"Pathomic Immune Biomarkers Define Recurrence Risk in Early-Stage Melanoma.","authors":"Thazin Nwe Aung, Ajay Singh, Gerardo Espinoza, Chenxin Zhang, Tianyun Jiang, Divya Kenchappa, Yadriel Bracero, Swanand Rakhade, Sharmin Sultana, Suryansh Shukla, Emily Nghiem, Matteo Abbruzzese, Chaoyuan Kuang, Larisa Geskin, David Entenberg, Robyn Gartrell, Tammie Ferringer, Lawrence Leung, Jee-Young Moon, Basil Horst, Kent Nastiuk, David Rimm, Rui Chang, Yvonne Saenger","doi":"10.1093/oncolo/oyaf327","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf327","url":null,"abstract":"<p><strong>Background: </strong>There is an urgent need for biomarkers in early-stage melanoma because the benefit of adjuvant immunotherapy is marginal while the toxicity is considerable. Immune surveillance is a key mechanism limiting melanoma progression, but no immune biomarkers have yet been sufficiently validated for clinical use.</p><p><strong>Methods: </strong>We validate three digital pathology biomarkers, all previously trained in published cohorts and testable in formalin fixed paraffin embedded (FFPE) specimens, for correlation with recurrence free survival (RFS) and distant metastatic free survival (DMFS): 1) tumor infiltrating lymphocytes identified using artificial intelligence in digital images (eTILs), 2) nanoString Melanoma Immune Profile (MIP), and 3) quantitative immunofluorescence (qIF) of CD8+ cells.</p><p><strong>Results: </strong>Three immune biomarkers were validated and found to correlate with recurrence at 36 months by receiver operating curve (ROC) analysis (eTILs AUC= 0.706, p = 0.002; MIP AUC= 0.775, p < 0.001; and CD8% AUC= 0.682, p = 0.009) and define high and low risk groups using Kaplan Meier (KM) curves within the IIA-IIID population (p = 0.007 for eTILs and p < 0.001 for MIP and CD8%) and within the difficult to treat IIB-IIIA subset (p = 0.005, p < 0.001, p < 0.001 respectively). Immune biomarkers enhance clinical predictors (p = 0.012, p < 0.001, p = 0.004), and correlate with distant metastatic recurrence (p = 0.031, p = 0.047, p = 0.014 respectively). Network analysis shows that stage and depth combined with pathomic immune features yields the highest correlation with recurrence (AUC=0.875, p < 0.001).</p><p><strong>Conclusions: </strong>Immune biomarkers should be prospectively validated in stage II-III melanoma for the purpose of avoiding overtreatment of low-risk patients and to stratify high-risk patients for clinical trials.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145240245","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}
OncologistPub Date : 2025-10-06DOI: 10.1093/oncolo/oyaf333
Enrique Grande, Joaquim Bellmunt, Syed A Hussain, Mubarak M Al Mansour, Aristotle Bamias, Philippe Barthélémy, David J Benjamin, Normand Blais, Maria T Bourlon, Daniel Castellano, Pongwut Danchaivijitr, Mauricio Fernandez Lazzaro, Patrizia Giannatempo, Félix Guerrero-Ramos, Roberto Iacovelli, Philipp Ivanyi, Eun Hee Jung, Ravindran Kanesvaran, Ray Manneh, Joana C Marinho, Nobuaki Matsubara, Axel S Merseburger, Deborah Mukherji, Chandler H Park, Ben Tran, Karine Martins da Trindade, Yüksel Ürün, Ashish M Kamat, Alison J Birtle
{"title":"Guiding First-Line Treatment Decisions In Advanced Urothelial Carcinoma: A Global Survey.","authors":"Enrique Grande, Joaquim Bellmunt, Syed A Hussain, Mubarak M Al Mansour, Aristotle Bamias, Philippe Barthélémy, David J Benjamin, Normand Blais, Maria T Bourlon, Daniel Castellano, Pongwut Danchaivijitr, Mauricio Fernandez Lazzaro, Patrizia Giannatempo, Félix Guerrero-Ramos, Roberto Iacovelli, Philipp Ivanyi, Eun Hee Jung, Ravindran Kanesvaran, Ray Manneh, Joana C Marinho, Nobuaki Matsubara, Axel S Merseburger, Deborah Mukherji, Chandler H Park, Ben Tran, Karine Martins da Trindade, Yüksel Ürün, Ashish M Kamat, Alison J Birtle","doi":"10.1093/oncolo/oyaf333","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf333","url":null,"abstract":"<p><strong>Background: </strong>Enfortumab vedotin combined with pembrolizumab (EV-P) has become the new standard first-line therapy for patients with advanced urothelial carcinoma (aUC), based on its superior efficacy over platinum-based chemotherapy. As this regimen is increasingly adopted in routine care, treatment decisions may often occur in sites without dedicated genitourinary oncology expertise. This global survey aimed to explore how physicians perceive clinical factors that may influence the safe and effective use of EV-P in daily practice.</p><p><strong>Material and methods: </strong>A panel of international physicians with experience in treating patients with genitourinary cancers developed a 17-question survey addressing practice settings, experience in managing aUC, and clinical considerations relevant to the use of EV-P. The participants were recruited through a network-based convenience sampling method. The responses were descriptively analyzed.</p><p><strong>Results: </strong>201 genitourinary physicians from 32 countries completed the questionnaire. The most frequently cited potential absolute contraindications were sensory or motor neuropathy grade ≥2 (64.2%), ECOG-PS ≥3 (59.2%), and non-urothelial component >50% of the tissue sample (59.2%). Other notable concerns included severe corneal/retinal abnormalities, HbA1c >11%, severe skin comorbidities, liver impairment grade ≥2, and dialysis dependence.</p><p><strong>Conclusions and relevance: </strong>This survey provides practical insights into real-world physician perspectives on patient selection for EV-P. The findings highlight the need for guidance to support personalized risk assessment, facilitate early identification of patients who may require enhanced monitoring, and optimize safe integration of EV-P into clinical practice.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145240233","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}
OncologistPub Date : 2025-10-06DOI: 10.1093/oncolo/oyaf331
Arjun Syal, May-Lucie Meyer, Kenneth Angelino, Noah Osei, Jorge E Gomez, Triparna Sen, Fred R Hirsch
{"title":"Antibody Drugs Conjugates in Non-Small Cell Lung Cancer: Current Status and Challenges.","authors":"Arjun Syal, May-Lucie Meyer, Kenneth Angelino, Noah Osei, Jorge E Gomez, Triparna Sen, Fred R Hirsch","doi":"10.1093/oncolo/oyaf331","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf331","url":null,"abstract":"<p><strong>Background: </strong>Antibody-drug conjugates (ADCs) are an emerging class of therapeutics that combine the specificity of monoclonal antibodies with cytotoxic or immune-stimulatory payloads. In non-small cell lung cancer (NSCLC), they offer a novel strategy with potential in both first line therapy and in cases to overcome resistance to existing targeted and immune-based therapies.</p><p><strong>Objective: </strong>To review the clinical development, efficacy, safety, biomarker strategies, and emerging targets of ADCs in NSCLC, with a focus on implications for practice and ongoing challenges.</p><p><strong>Methods: </strong>We conducted a comprehensive literature review of published trials, conference abstracts, and press releases evaluating ADCs in NSCLC, with attention to target antigens, clinical trial outcomes, and biomarker approaches.</p><p><strong>Results: </strong>ADCs targeting HER2, TROP2, and c-MET have received regulatory approval in NSCLC, with demonstrated efficacy-particularly in biomarker-selected populations. Bispecific HER3/EGFR-directed ADCs have shown encouraging activity in early-phase studies, with ongoing trials expected to clarify durability and optimal patient selection. Other targets such as ITGB6, B7-H3, and AXL have shown early signals of efficacy. Predictive biomarkers vary in reliability, and mutation, amplification, or protein expression do not uniformly predict response. Toxicity and acquired resistance remain key challenges; improved diagnostics may enhance patient selection.</p><p><strong>Conclusion: </strong>ADCs are poised to reshape the therapeutic landscape of NSCLC. Their success will hinge on refining biomarker strategies, managing toxicity, and integrating resistance-mitigating approaches such as bispecific constructs or rational combinations. As research advances, ADCs may become essential components of personalized therapy across a range of molecular and histologic NSCLC subtypes.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145240302","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}
OncologistPub Date : 2025-10-06DOI: 10.1093/oncolo/oyaf330
Anthea Peters, Grzegorz S Nowakowski, Rosy Dabas, Theresa Amoloja, Zhenyi Xue, Caroline Koch, Eva E Waltl, Isabelle Fleury
{"title":"Management of Canadian Patients With Refractory or Relapsed Diffuse Large B-Cell Lymphoma in the Real World: A Subanalysis of the RE-MIND2 Study.","authors":"Anthea Peters, Grzegorz S Nowakowski, Rosy Dabas, Theresa Amoloja, Zhenyi Xue, Caroline Koch, Eva E Waltl, Isabelle Fleury","doi":"10.1093/oncolo/oyaf330","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf330","url":null,"abstract":"<p><strong>Background: </strong>In the current Canadian treatment landscape for relapsed or refractory diffuse large B-cell lymphoma (R/R DLBCL), eligibility for autologous stem cell transplantation (ASCT) guides the choice of salvage treatment. CD19 chimeric antigen receptor T-cell (CAR-T) therapies have improved outcomes in patients with chemorefractory DLBCL, but access is limited to eligible patients. This subanalysis of the RE-MIND2 observational retrospective cohort study investigated treatment patterns for R/R DLBCL in Canada.</p><p><strong>Patients and methods: </strong>Data from patients enrolled in RE-MIND2 treated between 2010 and 2020 at 2 Canadian centers were retrospectively collected from health records. Descriptive statistics were used to analyze baseline characteristics, treatment initiated, and duration of treatment by line of therapy.</p><p><strong>Results: </strong>One hundred and nine patients were included; 74.2% of patients were eligible for ASCT as 2 L therapy and 45.4% received transplants. ASCT eligibility for third- (3 L) and fourth-line (4 L) therapy declined to 17.1% and 5.9%, respectively. Patients received a wide variety of treatments in 3 L and 4 L. CAR-T therapy became available in 3 L and 4 L by the end of 2019. Median durations of treatment were <2.6 months in all lines of therapy; median time to next treatment ranged from 3.4 months in 4 L to 5.3 months in 2 L.</p><p><strong>Conclusion: </strong>Results of our study support that ASCT-ineligible patients have a poor prognosis with conventional salvage chemotherapy. Prior to the availability of novel immunotherapies, no apparent standard of care was observed for Canadian patients with R/R DLBCL who were ineligible for or did not receive ASCT, especially after 2 L treatment.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145240243","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}
OncologistPub Date : 2025-10-06DOI: 10.1093/oncolo/oyaf332
Arjun Syal, May-Lucie Meyer, Kenneth Angelino, Noah Osei, Jorge E Gomez, Fred R Hirsch, Triparna Sen
{"title":"Antibody Drugs Conjugates in Small-Cell Lung Cancer: Present-Day Status and Promises.","authors":"Arjun Syal, May-Lucie Meyer, Kenneth Angelino, Noah Osei, Jorge E Gomez, Fred R Hirsch, Triparna Sen","doi":"10.1093/oncolo/oyaf332","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf332","url":null,"abstract":"<p><strong>Introduction: </strong>Small cell lung cancer (SCLC) accounts for 15% of lung cancers and is characterized by an aggressive disease course and historically poor prognosis. Although tumors in most patients respond to initial chemotherapy, relapse is nearly universal and treatment options remain limited. Antibody-drug conjugates (ADCs) have emerged as a novel therapeutic class with potential to address this unmet need.</p><p><strong>Methods: </strong>ClinicalTrials.gov, PubMed, and their associated references and press releases were queried for the search terms \"Antibody-drug conjugates\" and \"SCLC.\" Only English-language sources were included.</p><p><strong>Results: </strong>Multiple ADCs targeting diverse antigens have been evaluated in relapsed or refractory SCLC. Topoisomerase I inhibitor payloads have generated the most consistent activity across DLL3, TROP2, B7-H3, and SEZ6 targets, while microtubule and pyrrolobenzodiazepine (PBD)- based constructs have not demonstrated durable benefit. Despite encouraging response rates, progression-free survival has remained short, reflecting intrinsic resistance, antigen heterogeneity, and toxicity-related dose limitations.</p><p><strong>Conclusion: </strong>While ADCs have generated encouraging response rates in SCLC, durability has remained limited. Future development will require optimization of payloads, linkers, and trial design to determine whether ADCs can achieve a sustained role in this disease.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145240290","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}