JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-17DOI: 10.1200/PO-24-00668
Claire Ducos, Brice Fresneau, Filippo Rosselli, Giao Vu-Bezin, Boris Schwartz, Rodrigue S Allodji, Gaelle Marenne, Thomas E Ludwig, Anne Boland-Augé, Jean-François Deleuze, Helene Blanché, Robert Olaso, Julie Nys, Sarah Winter, Franck Bourdeaut, Chiraz El-Fayech, Carole Rubino, Ibrahima Diallo, Florent de Vathaire, Simone Benhamou, Nadia Haddy
{"title":"Rare Variants Associated With Pediatric Cancer Treatment-Related Second Malignant Neoplasm Risk.","authors":"Claire Ducos, Brice Fresneau, Filippo Rosselli, Giao Vu-Bezin, Boris Schwartz, Rodrigue S Allodji, Gaelle Marenne, Thomas E Ludwig, Anne Boland-Augé, Jean-François Deleuze, Helene Blanché, Robert Olaso, Julie Nys, Sarah Winter, Franck Bourdeaut, Chiraz El-Fayech, Carole Rubino, Ibrahima Diallo, Florent de Vathaire, Simone Benhamou, Nadia Haddy","doi":"10.1200/PO-24-00668","DOIUrl":"https://doi.org/10.1200/PO-24-00668","url":null,"abstract":"<p><strong>Purpose: </strong>Survivors of childhood cancers are at risk of second malignant neoplasms (SMNs). Exposure to radiation therapy and different chemotherapy agents are known risk factors for SMN development. However, there remains interindividual variability that could be attributed to genetic variations. Our study aims to identify rare genetic variants associated with SMN risk and assess the influence of treatment on the associated risk of these variants.</p><p><strong>Materials and methods: </strong>We conducted a whole-exome sequencing in a nested case-control study within the French Childhood Cancer Survivors Study for 450 survivors including 163 cases and 287 controls. Rare variants in genes within replication, recombination, and repair pathways were selected. Gene-based association tests were conducted and a logistic regression analysis was used to estimate the effect of selected genes on SMN risk, adjusting for relevant clinical variables. Specific analysis restricted to breast and thyroid SMNs were also performed. Stratified analyses on the basis of the radiation doses received by the bone marrow and specific organs were also conducted.</p><p><strong>Results: </strong>We have identified several genetic associations: the <i>RNASEL</i> and <i>APOBEC3F</i> genes (odds ratio [OR], 5.43 [95% CI, 1.76 to 20.44]; <i>P</i> = .0055 and OR, 4.8 [95% CI, 1.28 to 22.95]; <i>P</i> = .027, respectively) are associated with the risk of SMN, while the <i>FANCM</i> gene (OR, 4.65 [95% CI, 1.06 to 21.56]; <i>P</i> = .041) is associated with the risk of developing breast SMN.</p><p><strong>Conclusion: </strong>This study provides new evidence regarding the involvement of genetics in the development of SMN. Additional research is needed to confirm these results and uncover the underlying mechanisms behind these associations. If replicated, these findings will help identify survivors at higher risk of developing SMN, enabling tailored treatment and follow-up strategies.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400668"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145080750","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}
JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-26DOI: 10.1200/PO-25-00048
Manlu Liu, Rachel L Minne, Saahil Javeri, D Bryan Johnson, Scott A Tomlins, Randall J Kimple, Andrew M Baschnagel
{"title":"Immune and Genomic Heterogeneity of MET-Altered Non-Small Cell Lung Cancer.","authors":"Manlu Liu, Rachel L Minne, Saahil Javeri, D Bryan Johnson, Scott A Tomlins, Randall J Kimple, Andrew M Baschnagel","doi":"10.1200/PO-25-00048","DOIUrl":"https://doi.org/10.1200/PO-25-00048","url":null,"abstract":"<p><strong>Purpose: </strong>Patients with non-small cell lung cancer (NSCLC) harboring <i>MET</i> exon 14 skipping mutations (<i>MET</i>ex14) or <i>MET</i> amplifications (<i>MET</i>amp) have demonstrated varied responses to immunotherapy. This study aimed to better understand the genomic and immune characteristics of <i>MET-</i>altered NSCLC.</p><p><strong>Materials and methods: </strong>The study included 3,841 patients with NSCLC sequenced using the Strata Select assay on the Strata Oncology Platform. Genomic alterations, tumor mutational burden (TMB), PD-L1 expression, and immune gene expression were compared between high <i>MET</i>amp (copy number gain [CNG] ≥10), low <i>MET</i>amp (CNG 6-9), <i>MET</i>ex14, other <i>MET</i> mutations, and <i>MET</i> wild-type (<i>MET</i>wt) patients. Immune-related gene expression was also analyzed in adenocarcinomas (n = 2,708) with targetable oncogenic drivers.</p><p><strong>Results: </strong>The most common genomic alterations were <i>TP53</i> mutations and <i>MDM2</i> amplification in <i>MET</i>ex14 and <i>TP53</i> and <i>CDKN2A</i> in <i>MET</i>amp tumors. TMB was lowest in patients with <i>MET</i>ex14 and highest in patients with other <i>MET</i> mutations. PD-L1 expression was high in <i>MET</i>ex14, high in <i>MET</i>amp, and low in <i>MET</i>amp. Tumors with both <i>MET</i>amp and <i>EGFR</i> mutations had higher PD-L1 expression compared with tumors with only EGFR mutations. <i>MET</i>ex14 and low <i>MET</i>amp had higher receptor tyrosine kinase <i>AXL</i> gene expression relative to <i>MET</i>wt. Comparisons across oncogene-driven lung adenocarcinomas revealed that <i>MET</i>ex14 had an enriched immune landscape, whereas <i>MET</i>amp harbored an immunosuppressive environment.</p><p><strong>Conclusion: </strong><i>MET</i>ex14 and <i>MET</i>amp differed in genomic coalterations, TMB, and immune gene expression. These variations provide insight for the inconsistent response to immunotherapy in NSCLC with <i>MET</i> alterations, warranting further investigation.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500048"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145175574","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}
JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-16DOI: 10.1200/PO-25-00027
Diantha Terlouw, Maartje Nielsen, Jurjen J Boonstra, Monique E van Leerdam, Tom van Wezel, Hans Morreau, Alexandra M J Langers
{"title":"Exploring <i>APC</i> Mosaicism in Upper Intestinal Tract Adenomas.","authors":"Diantha Terlouw, Maartje Nielsen, Jurjen J Boonstra, Monique E van Leerdam, Tom van Wezel, Hans Morreau, Alexandra M J Langers","doi":"10.1200/PO-25-00027","DOIUrl":"10.1200/PO-25-00027","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500027"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12442778/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145075206","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}
{"title":"Interpretable Machine Learning Models for Predicting Lateral Pelvic Lymph Node Metastasis in Rectal Cancer: A Chinese Multicenter Retrospective Study.","authors":"Tixian Xiao, Wei Zhao, Zhen Sun, Fangze Wei, Fuqiang Zhao, Fei Huang, Zeyu Wu, Junge Bai, Xin Wang, Qian Liu","doi":"10.1200/PO-25-00192","DOIUrl":"10.1200/PO-25-00192","url":null,"abstract":"<p><strong>Purpose: </strong>Internal iliac and obturator lymph nodes are common sites of metastasis in rectal cancer. This study developed a machine learning (ML) model using clinical data to predict lymph node metastasis and applied the Shapley Additive explanations (SHAP) method for interpretation.</p><p><strong>Materials and methods: </strong>Retrospectively, data from patients with rectal cancer at four Chinese centers-who underwent total mesorectal excision and lateral pelvic lymph node dissection without neoadjuvant therapy-were collected. Two centers provided training/test sets (3:1 ratio) and two centers supplied external validation. Lymph node enlargement was determined by imaging and confirmed by pathology. Five ML models were evaluated by AUC, accuracy, and F1 score. Key features included demographics, tumor stage, tumor-to-anal verge distance, imaging measurements, tumor histological differentiation, preoperative carcinoembryonic antigen, and carbohydrate antigen 19-9. SHAP was used to assess feature importance.</p><p><strong>Results: </strong>Of the 411 cases (174 positives) in the training/test sets and 109 cases (43 positives) in external validation, the random forest (RF) model ranked second in terms of AUC and accuracy in the training set (0.999, 0.995), whereas it achieved the highest AUC and accuracy (0.877 and 0.788) in the test set. In the external validation, the RF model outperformed all other ML models (AUC of 0.899, accuracy of 0.827). Overall, the RF model demonstrates the superior overall performance. According to the SHAP analysis, the most important predictors of internal iliac and obturator lymph node metastasis were, in descending order, the short-axis diameter of enlarged lymph nodes, regional lymph node metastasis, and tumor-to-anal verge distance. At the individual patient level, SHAP force plots provided explanations of the RF model predictions for internal iliac and obturator lymph node metastasis.</p><p><strong>Conclusion: </strong>An interpretable ML model was developed that accurately predicts internal iliac and obturator lymph node metastasis using clinical data. SHAP analysis enhances understanding of feature contributions, supporting personalized treatment planning.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500192"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12445183/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145080716","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}
JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-17DOI: 10.1200/PO-25-00365
Spencer Gibson, Farah Shah, Ritesh Parajuli
{"title":"Complete Response With Alpelisib and Trastuzumab in a Patient With Estrogen Receptor-/Progesterone Receptor-Negative, Human Epidermal Growth Factor Receptor 2-Positive Refractory Metastatic Breast Cancer.","authors":"Spencer Gibson, Farah Shah, Ritesh Parajuli","doi":"10.1200/PO-25-00365","DOIUrl":"10.1200/PO-25-00365","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500365"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12445173/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145080748","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}
JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-04DOI: 10.1200/PO-25-00295
Annalice Gandini, Hélène Blons, Pierre Laurent-Puig, Chloé Broudin, Simon Garinet, Julien Taieb, Claire Gallois
{"title":"Targeting <i>EGFR</i> Fusion in Metastatic Colorectal Cancer: A Case Report of a Lazarus Response in a 19-Year-Old Patient.","authors":"Annalice Gandini, Hélène Blons, Pierre Laurent-Puig, Chloé Broudin, Simon Garinet, Julien Taieb, Claire Gallois","doi":"10.1200/PO-25-00295","DOIUrl":"https://doi.org/10.1200/PO-25-00295","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500295"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145000621","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}
JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-24DOI: 10.1200/PO-25-00636
Sanam Loghavi, Hiam Abdel-Salam, Danielle Hammond, Kelly Chien, Keyur P Patel, Banu K Arun, Courtney D DiNardo
{"title":"Unveiling the Origins of Pathogenic <i>TP53</i> Variants in Clinical Context: A Molecular Tumor Board Case Discussion.","authors":"Sanam Loghavi, Hiam Abdel-Salam, Danielle Hammond, Kelly Chien, Keyur P Patel, Banu K Arun, Courtney D DiNardo","doi":"10.1200/PO-25-00636","DOIUrl":"https://doi.org/10.1200/PO-25-00636","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500636"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145137723","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}
JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-19DOI: 10.1200/PO-25-00159
Carsten Bokemeyer, Keith R Abrams, Jesus Garcia-Foncillas, Antoine Italiano, Ulrik Lassen, Louise Linsell, Marisca Marian, Noman Paracha, Sean D Sullivan, Nicoletta Brega, Juliette Thompson
{"title":"Clinical Benefit From Molecularly Guided Cancer Care: A Matching-Adjusted Indirect Comparison of Larotrectinib Versus Standard of Care.","authors":"Carsten Bokemeyer, Keith R Abrams, Jesus Garcia-Foncillas, Antoine Italiano, Ulrik Lassen, Louise Linsell, Marisca Marian, Noman Paracha, Sean D Sullivan, Nicoletta Brega, Juliette Thompson","doi":"10.1200/PO-25-00159","DOIUrl":"10.1200/PO-25-00159","url":null,"abstract":"<p><strong>Purpose: </strong>Widespread adoption of the tropomyosin receptor kinase (TRK) inhibitor larotrectinib has been hampered by limited comparisons against non-TRK inhibitor standard of care (SoC) regimens because of the rarity of TRK fusions. Cross-trial comparisons may be facilitated using matching-adjusted indirect comparison (MAIC), a validated methodology that balances baseline population characteristics.</p><p><strong>Materials and methods: </strong>MAIC was conducted using individual patient-level data from three larotrectinib trials, compared with aggregate real-world data (RWD) from adult patients with locally advanced/metastatic TRK fusion-positive cancer in the Hartwig Medical Foundation database. Patient populations were matched on available clinical characteristics. Estimates of overall survival (OS; defined as the time from start of first postbiopsy treatment [or larotrectinib] to death) were compared between larotrectinib and non-TRK inhibitor SoC.</p><p><strong>Results: </strong>The analysis included 24 patients receiving nontargeted TRK inhibitor SoC and 120 receiving larotrectinib. After matching, median OS was 50.3 months (IQR, 23.3-not estimable) for larotrectinib versus 13 months (IQR, 6.4-18.3) for SoC; larotrectinib was associated with an 84% risk reduction of death (adjusted hazard ratio, 0.16 [95% CI, 0.07 to 0.36]). To account for the longer follow-up time in the larotrectinib group than in the SoC group (median: 56.7 <i>v</i> 23.2 months), a restricted mean survival analysis was conducted up to 26.2 months (largest observed event time in SoC arm), which showed median survival of 22.6 months for larotrectinib and 12.8 months for SoC (mean difference: 9.8 months [95% CI, 5.6 to 14.0]).</p><p><strong>Conclusion: </strong>In patients with TRK fusion-positive cancer, larotrectinib was associated with a mean survival advantage of 9.8 months compared with non-TRK inhibitor SoC, corroborating favorable findings for larotrectinib from a previous analysis that used a different set of RWD.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500159"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12462675/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145091825","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}
JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-26DOI: 10.1200/PO-24-00832
Chiara Maura Ciniselli, Elena Conca, Michele Bartoletti, Nicoletta Colombo, Umberto Malapelle, Salvatore Lopez, Sandro Pignata, Francesco Raspagliesi, Paolo Verderio, Claudio Zamagni, Valentina Sara Barbiero, Silvia Castellani, Emilia Sicari, Luca Agnelli, Federica Perrone, Giancarlo Pruneri
{"title":"Leveraging Real-World Data From a Clinicogenomic Database Addresses the Treatment Gap in Patients With High-Grade Serous Ovarian Cancer.","authors":"Chiara Maura Ciniselli, Elena Conca, Michele Bartoletti, Nicoletta Colombo, Umberto Malapelle, Salvatore Lopez, Sandro Pignata, Francesco Raspagliesi, Paolo Verderio, Claudio Zamagni, Valentina Sara Barbiero, Silvia Castellani, Emilia Sicari, Luca Agnelli, Federica Perrone, Giancarlo Pruneri","doi":"10.1200/PO-24-00832","DOIUrl":"10.1200/PO-24-00832","url":null,"abstract":"<p><strong>Purpose: </strong>This study used a deidentified nationwide (US-based) high-grade serous ovarian cancer (HGSOC) clinicogenomic database (CGDB) to enrich our understanding of HGSOC's genomic heterogeneity and assess the utility of comprehensive genomic profiling (CGP) in clinical settings.</p><p><strong>Patients and methods: </strong>We conducted a retrospective observational analysis on 856 patients with HGSOC profiled with CGP genomic tests, retrieved from CGDB from January 2011 to September 2023.</p><p><strong>Results: </strong>In addition to <i>BRCA1</i> (11.7%) and <i>BRCA2</i> (6.5%) variants, CGP revealed further potentially actionable alterations (amplifications and/or mutations) in <i>CCNE1</i> (16%), <i>FGFR1/2/3/4</i> (6.5%), <i>PIK3CA</i> (3.9%), <i>TP53</i><sup><i>Y220C</i></sup> (3.7%), <i>ERBB2</i> (3.5%), <i>CDK12</i> (2.3%), <i>ARID1A</i> (2.2%), <i>KRAS</i> (2.1%), and <i>BRAF</i> (1%) genes. Then, 439 patients were selected, presenting both CGP test performed on specimen collected at the time of surgery and initiation of first-line therapy within ±8 months from surgery, and categorized into no (NS, n = 74), interval (IS, n = 157), and upfront surgery (n = 208) groups, each comparable by clinical features. The CGP revealed <i>BRCA</i> mutations, at similar frequency in the three groups, in 54/439 patients (12.3%). Patients with pathogenic <i>BRCA</i> mutations had better event-free survival (EFS) compared with those with <i>BRCA</i><sup><i>wt</i></sup>. Loss-of-heterozygosity (LOH) ≥16 (LOH-positive patients) was found in 142/433 (32.8%) patients, with different prevalence across treatment groups (12.8% NS; 9% IS; 8.8% upfront surgery). Patients treated with poly (ADP-ribose) polymerase inhibitors (PARPi) had improved EFS (hazard ratio for other drugs <i>v</i> PARPi 1.77 [95% CI, 1.21 to 2.58]). Interestingly, in 206 <i>BRCA</i><sup><i>wt</i></sup> and LOH-negative patients, not eligible for PARPi, CGP detected potentially targetable alterations in 99 of them (48%).</p><p><strong>Conclusion: </strong>Overall, our study provides evidence that CGP significantly improves the identification of molecular targets in HGSOC, supporting its importance in the clinical practice to provide patients with more therapeutic options.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400832"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145175597","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}
JCO precision oncologyPub Date : 2025-09-01Epub Date: 2025-09-24DOI: 10.1200/PO-25-00177
Javier Perez, Keith M Kerr, Brian Baker, Fang Fang, Jason Li, Jannine McDonald, Siyu Li, Bo Gao, Jean-Francois Pouliot, Frank Seebach, Israel Lowy, Giuseppe Gullo, Gary Herman, Jennifer Hamilton, Petra Rietschel, Kristina McGuire
{"title":"Clinical Interchangeability of PD-L1 Immunohistochemistry Assays in First-Line Non-Small Cell Lung Cancer Management With Cemiplimab.","authors":"Javier Perez, Keith M Kerr, Brian Baker, Fang Fang, Jason Li, Jannine McDonald, Siyu Li, Bo Gao, Jean-Francois Pouliot, Frank Seebach, Israel Lowy, Giuseppe Gullo, Gary Herman, Jennifer Hamilton, Petra Rietschel, Kristina McGuire","doi":"10.1200/PO-25-00177","DOIUrl":"10.1200/PO-25-00177","url":null,"abstract":"<p><strong>Purpose: </strong>Multiple immunohistochemistry (IHC) assays targeting PD-L1 have been developed independently, and a lack of harmonization creates undue complexity for clinicians who use PD-L1 tests to guide treatment decisions. This novel bridging study demonstrates the clinical interchangeability of two PD-L1 IHC assays in first-line treatment of non-small cell lung cancer (NSCLC) with PD-L1 ≥50%.</p><p><strong>Methods: </strong>In the phase III EMPOWER-Lung 1 study (ClinicalTrials.gov identifier: NCT03088540), 710 patients were randomly assigned 1:1 to first-line cemiplimab or platinum-doublet chemotherapy for advanced NSCLC with PD-L1 ≥50%, selected by PD-L1 IHC 22C3 pharmDx (Agilent Technologies, Santa Clara, CA).</p><p><strong>Results: </strong>A total of 871 patient samples were retrospectively tested using the VENTANA PD-L1 (SP263) assay (Roche Diagnostics, Indianapolis, IN), including 481 enrolled patients and 390 patients who did not pass screening (22C3 PD-L1 <50%). Concordance of 22C3 and SP263 was evaluated in 768 patient samples, with an overall percent agreement (concordance) of 88% between the two assays. Overall survival (OS) and progression-free survival (PFS) were estimated in the PD-L1 ≥50% SP263+ population (including the 22C3+/SP263+ and 22C3-/SP263+ subpopulations). In the primary analysis, the clinical efficacy of cemiplimab versus chemotherapy in the 22C3+/SP263+ population (n = 324; OS hazard ratio [HR], 0.52 [95% CI, 0.34 to 0.80]; PFS HR, 0.43 [95% CI, 0.32 to 0.59]) was similar to that in the 22C3+ population (n = 563). A sensitivity analysis of the overall SP263+ population showed consistent results with the primary analysis.</p><p><strong>Conclusion: </strong>Similar efficacy (OS and PFS) was observed with the 22C3+ and SP263+ populations, demonstrating the interchangeability of these PD-L1 IHC assays for selecting patients with PD-L1 ≥50% for first-line cemiplimab monotherapy for advanced NSCLC.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500177"},"PeriodicalIF":5.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12487655/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145137762","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}