JCO precision oncologyPub Date : 2025-05-01Epub Date: 2025-05-22DOI: 10.1200/PO-25-00369
Thomas J George, Ji-Hyun Lee, David L DeRemer, Peter J Hosein, Steven Staal, Merry Jennifer Markham, Dennie Jones, Karen C Daily, Jonathan A Chatzkel, Brian H Ramnaraign, Julia L Close, Nkiruka Ezenwajiaku, Martina C Murphy, Carmen J Allegra, Sherise Rogers, Zhongyue Zhang, Derek Li, Gayathri Srinivasan, Montaser Shaheen, Robert Hromas
{"title":"Erratum: Phase II Trial of the PARP Inhibitor, Niraparib, in BAP1 and Other DNA Damage Response Pathway-Deficient Neoplasms.","authors":"Thomas J George, Ji-Hyun Lee, David L DeRemer, Peter J Hosein, Steven Staal, Merry Jennifer Markham, Dennie Jones, Karen C Daily, Jonathan A Chatzkel, Brian H Ramnaraign, Julia L Close, Nkiruka Ezenwajiaku, Martina C Murphy, Carmen J Allegra, Sherise Rogers, Zhongyue Zhang, Derek Li, Gayathri Srinivasan, Montaser Shaheen, Robert Hromas","doi":"10.1200/PO-25-00369","DOIUrl":"https://doi.org/10.1200/PO-25-00369","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500369"},"PeriodicalIF":5.3,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144127591","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 : 2025-05-01Epub Date: 2025-05-29DOI: 10.1200/PO-24-00898
Nicholas Mai, Miriam M Klar Lieberman, Emanuela Ferraro, Maria Bromberg, Yuan Chen, Pedram Razavi, Shanu Modi, Sarat Chandarlapaty, Elaine M Walsh, Joshua Z Drago
{"title":"Sequential Antibody-Drug Conjugate Therapy in Patients With Metastatic Breast Cancer Treated With Sacituzumab Govitecan and Trastuzumab Deruxtecan.","authors":"Nicholas Mai, Miriam M Klar Lieberman, Emanuela Ferraro, Maria Bromberg, Yuan Chen, Pedram Razavi, Shanu Modi, Sarat Chandarlapaty, Elaine M Walsh, Joshua Z Drago","doi":"10.1200/PO-24-00898","DOIUrl":"https://doi.org/10.1200/PO-24-00898","url":null,"abstract":"<p><strong>Purpose: </strong>Sacituzumab govitecan (SG) and trastuzumab deruxtecan (T-DXd) are antibody-drug conjugates (ADCs) approved for the treatment of human epidermal growth factor receptor 2 (HER2)-low metastatic breast cancer (MBC). Both carry topoisomerase-1-inhibiting payloads, and it is unknown whether these drugs retain activity when used sequentially.</p><p><strong>Methods: </strong>Patients who received both T-DXd and SG for treatment of MBC were eligible. The primary objective was to describe clinical outcomes and clinicogenomic characteristics associated with improved real-world progression-free survival (<sup>rw</sup>PFS) of both T-DXd and SG.</p><p><strong>Results: </strong>Eighty-five patients were eligible. Regardless of which ADC was deployed first (<sup>rw</sup>PFS1), median <sup>rw</sup>PFS with the second ADC (<sup>rw</sup>PFS2) was shorter in 75.2% of patients, with 14 patients remaining on treatment at the data cutoff. Individual patient cases of prolonged benefit were however observed with both T-DXd and SG when used second. In multivariate analyses, predictors of better <sup>rw</sup>PFS2 included longer <sup>rw</sup>PFS1 (hazard ratio [HR], 0.94 [95% CI, 0.89 to 1.00]; <i>P</i> = .04) and earlier overall treatment line (HR, 1.10 [95% CI, 1.01 to 1.21]; <i>P</i> = .03). Genomic analysis of pretreatment tissue samples revealed that PTEN loss is associated with de novo resistance to T-DXd (HR, 3.20 [95% CI, 1.47 to 6.97]; <i>P</i> = .003) but not SG (HR, 1.18 [95% CI, 0.54 to 2.56]; <i>P</i> = .68). There were no significant associations between estrogen receptor or HER2 status and <sup>rw</sup>PFS2.</p><p><strong>Conclusion: </strong>Sequential ADC therapy with topoisomerase-1-inhibiting payloads is a viable treatment strategy in HER2-low MBC. These results have hypothesis-generating clinical and translational implications. Further studies are needed to better understand ADC cross-resistance as more of these agents enter our clinical armamentarium.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400898"},"PeriodicalIF":5.3,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144180951","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 : 2025-05-01Epub Date: 2025-05-29DOI: 10.1200/PO-24-00830
Edward G Hughes, Drew T Bergman, Donald C Green, Shrey S Sukhadia, Joel A Lefferts, Jeremiah X Karrs, Rutu S Vyas, Zofia Cieslak, Jennifer N Barbuto, Nevena B Ognjenovic, Brianna E Palisoul, Gregory J Tsongalis, Laura J Tafe, Parth S Shah
{"title":"Clinical Implementation of Expanded Solid Tumor Fusion Detection With Whole-Transcriptome Sequencing at an Academic Cancer Center.","authors":"Edward G Hughes, Drew T Bergman, Donald C Green, Shrey S Sukhadia, Joel A Lefferts, Jeremiah X Karrs, Rutu S Vyas, Zofia Cieslak, Jennifer N Barbuto, Nevena B Ognjenovic, Brianna E Palisoul, Gregory J Tsongalis, Laura J Tafe, Parth S Shah","doi":"10.1200/PO-24-00830","DOIUrl":"https://doi.org/10.1200/PO-24-00830","url":null,"abstract":"<p><strong>Purpose: </strong>Chromosomal rearrangements drive tumorigenesis through fusion transcripts. Clinical whole-transcriptome sequencing (WTS) enables comprehensive fusion detection, overcoming limitations of targeted methods, but its clinical application has been hindered by technical and cost barriers.</p><p><strong>Methods: </strong>We validated a clinical bait-capture WTS assay using 78 solid tumor samples across diverse tissue types, including 59 with known fusions or oncogenic splice variants. Sensitivity was assessed against clinically reported fusions from targeted next-generation sequencing or fluorescence in situ hybridization. Analytical performance was evaluated for sensitivity, specificity, limit of detection, and reproducibility across varying RNA inputs and sequencing conditions. Clinical implementation data were also analyzed.</p><p><strong>Results: </strong>WTS demonstrated a sensitivity of 97.1% and a specificity of 100% for detecting clinically relevant fusions. Detection was robust across tumor types, with RNA input as low as 1 ng. Optimized sequencing thresholds of 60 million reads and 130-bp read length improved performance. The assay was highly reproducible and resistant to common contaminants. Over 6 months of clinical use (410 specimens), WTS identified 29 fusions (7% detection rate), including six clinically relevant fusions undetectable by our institution's previous targeted panel, increasing the fusion yield by 26%.</p><p><strong>Conclusion: </strong>WTS provides high sensitivity, specificity, and expanded fusion detection, supporting its routine clinical adoption. Future work will explore its broader role in integrated genomic profiling and therapeutic guidance.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400830"},"PeriodicalIF":5.3,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144183630","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 : 2025-05-01Epub Date: 2025-05-22DOI: 10.1200/PO-24-00779
Marta Brunetti, Valeria Vitelli, Anca Mihaela Naas, Ane Gerda Zahl Eriksson, Hans Kristian Haugland, Camilla Krakstad, Francesca Micci
{"title":"Molecular Landscape of Endometrial Stromal Tumors.","authors":"Marta Brunetti, Valeria Vitelli, Anca Mihaela Naas, Ane Gerda Zahl Eriksson, Hans Kristian Haugland, Camilla Krakstad, Francesca Micci","doi":"10.1200/PO-24-00779","DOIUrl":"10.1200/PO-24-00779","url":null,"abstract":"<p><strong>Purpose: </strong>The molecular heterogeneity of endometrial stromal tumors (ESTs) is demonstrated by the presence of the same fusion gene in distinct pathologic entities, such as endometrial nodules and low-grade endometrial stromal sarcoma, both exhibiting the <i>JAZ1::SUZ12</i> chimeric transcript. Given the limited knowledge on these tumors, which is based on a small number of cases studied with a restricted range of techniques, we analyzed 47 ESTs to explore their methylation and transcriptomic landscapes.</p><p><strong>Materials and methods: </strong>Tumor methylation and transcriptomes profiles were investigated.</p><p><strong>Results: </strong>The methylation profile showed distinct clusters, which correlated with established histopathologic and molecular subtypes. The highest methylation value was reported for nuclear factor of activated T cytoplasmic 1, and the lowest was detected for miR34C. Two different 5'-C-phosphate-G-3' (CpG) sites of <i>LMX1B</i> (<i>LMX1B-cg04996334</i> and <i>LMX1B</i>), along with <i>miR34C</i>, showed the same methylation pattern in both low-grade and high-grade endometrial stromal sarcoma (HG-ESS). Similarly, <i>CFAP45</i>, <i>HDAC4</i>, <i>ACY3</i>, <i>MOB3A</i>, and <i>XXYLT1</i> showed identical methylation patterns in HG-ESS and undifferentiated uterine sarcomas, highlighting the similarities between these tumors within the EST spectrum. We identified 13 novel fusion transcripts involving several genes that are active in transcriptional regulation.</p><p><strong>Conclusion: </strong>In ESTs, the genes involved in chromosomal rearrangements function as transcription regulators, either directly through the formation of zinc finger motifs or indirectly through epigenetic regulation. The methylation signature is different for distinct subgroups of the EST spectrum, with more aggressive tumors, HG-ESS, and undifferentiated uterine sarcoma, clustering together. Some genes showed similar methylation levels in different entities, highlighting the presence of a continuum in the tumor profile. Methylation levels of CpG sites at specific gene loci may serve as valuable biomarkers for these tumors.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400779"},"PeriodicalIF":5.3,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12122098/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144127604","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 : 2025-05-01Epub Date: 2025-05-22DOI: 10.1200/PO-24-00790
Maria Vittoria Chiaruttini, Claudia Proto, Giuseppe Lo Russo, Arsela Prelaj, Miriam Segale, Anna Zanghì, Francesca Galli, Francesca G Greco, Diego Signorelli, Marta Brambilla, Mario Occhipinti, Filippo De Braud, Marina C Garassino, Gabriella Sozzi, Eliana Rulli, Mattia Boeri
{"title":"Tracking the Response to Immunotherapy: Blood microRNA Dynamics in Patients With Advanced Non-Small Cell Lung Cancer.","authors":"Maria Vittoria Chiaruttini, Claudia Proto, Giuseppe Lo Russo, Arsela Prelaj, Miriam Segale, Anna Zanghì, Francesca Galli, Francesca G Greco, Diego Signorelli, Marta Brambilla, Mario Occhipinti, Filippo De Braud, Marina C Garassino, Gabriella Sozzi, Eliana Rulli, Mattia Boeri","doi":"10.1200/PO-24-00790","DOIUrl":"10.1200/PO-24-00790","url":null,"abstract":"<p><strong>Purpose: </strong>Despite the significant improvement in outcomes for patients with advanced non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors (ICIs), resistance, whether primary or secondary, remains a substantial challenge. Currently, reliable biomarkers to monitor ICI response are lacking, highlighting the need for minimally invasive tools like liquid biopsy to track treatment efficacy. This study aimed to identify circulating microRNAs (miRNAs) as potential biomarkers to track ICI response in patients with NSCLC.</p><p><strong>Materials and methods: </strong>The Apollo longitudinal study enrolled patients with advanced NSCLC receiving ICI in first or subsequent lines. Plasma samples were collected at baseline and follow-up to prospectively assess miRNA profiles until progressive disease (PD). Using a custom reverse transcription-quantitative polymerase chain reaction platform, 276 ratios among 24 lung cancer-related miRNAs were analyzed. The generalized estimating equation and joint models were applied to select the miRNA ratios most associated with PD over time. To control for multiple testing, the Benjamini-Yekutieli method was applied setting a 10% false discovery rate threshold.</p><p><strong>Results: </strong>From the 211 patients, a total of 454 plasma samples were analyzed. Clinical and biochemical variables had little effect on miRNAs' profile. The analysis identified nine miRNA ratios, all involving miR-145-5p, as significant biomarkers for monitoring treatment response, even after adjustment for the line of therapy. These ratios exhibited a longitudinal modulation pattern consistent with radiologic response, particularly in patients who initially benefited from ICI treatment. In addition, in an independent set of 32 plasma samples from 10 patients receiving ICI as maintenance therapy, the same trends were observed.</p><p><strong>Conclusion: </strong>A focused panel of miRNA ratios, driven by miR-145-5p, effectively reflects response to ICI therapy in patients with advanced NSCLC, highlighting their potential as biomarkers for treatment monitoring.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400790"},"PeriodicalIF":5.3,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12122099/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144127610","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 : 2025-05-01Epub Date: 2025-05-29DOI: 10.1200/PO-24-00855
Roelof van Ewijk, Laura S Hiemcke-Jiwa, Jayne Y Hehir-Kwa, Nathalie Gaspar, Lianne M Haveman, Uta E Flucke, Rana Dandis, Marc van Tuil, Claudia Y Janda, Michiel A J van de Sande, Lizz van der Heijden, Marco J Koudijs, Johannes H M Merks, Bastiaan B J Tops, Antonin Marchais, Lennart A Kester
{"title":"Prognostic Value of the G2 Expression Signature and MYC Overexpression in Childhood High-Grade Osteosarcoma.","authors":"Roelof van Ewijk, Laura S Hiemcke-Jiwa, Jayne Y Hehir-Kwa, Nathalie Gaspar, Lianne M Haveman, Uta E Flucke, Rana Dandis, Marc van Tuil, Claudia Y Janda, Michiel A J van de Sande, Lizz van der Heijden, Marco J Koudijs, Johannes H M Merks, Bastiaan B J Tops, Antonin Marchais, Lennart A Kester","doi":"10.1200/PO-24-00855","DOIUrl":"https://doi.org/10.1200/PO-24-00855","url":null,"abstract":"<p><strong>Purpose: </strong>In high-grade osteosarcoma, prognostic factors at diagnosis are insufficient for stratifying patients into relevant subgroups. Recently, a transcriptomic study developed the G1/G2 gene expression signature, in which the G2 signature was associated with unfavorable survival. An orthogonal study identified <i>MYC</i> amplification as an unfavorable prognostic factor using targeted next-generation sequencing. The purpose of this study was to validate the independent prognostic value and to investigate the combined prognostic value of the G1/G2 signature with <i>MYC</i> amplification and/or <i>MYC</i> expression for survival prediction.</p><p><strong>Materials and methods: </strong>This study included pediatric and adolescent patients with high-grade osteosarcoma. RNA-seq was performed in 48 patients. Whole-exome sequencing was performed in 40 patients. Gene expression signature scores, <i>MYC</i> amplification (defined as >seven copies), and <i>MYC</i> expression levels were calculated. Multivariable Cox proportional hazards analysis was performed for event-free survival (EFS; primary end point) and overall survival (OS; secondary end point).</p><p><strong>Results: </strong>In the full cohort, the 3-year EFS rate was 37%. In multivariable Cox regression analysis with metastatic disease stage (n = 21, 44%) as covariate, the G2 signature and <i>MYC</i> expression were independently associated with worse outcomes in terms of EFS (hazard ratio [HR], 3.32 [95% CI, 1.34 to 8.21] and HR, 3.38 [95% CI, 1.71 to 6.66], respectively) and OS (HR, 4.07 [95% CI, 1.19 to 13.9] and HR, 2.88 [95% CI, 1.22 to 6.76], respectively). MYC amplification was not associated with EFS or OS in univariable analysis (HR, 1.88 [95% CI, 0.74 to 4.77] and HR, 0.79 [95% CI, 0.21 to 3.05], respectively).</p><p><strong>Conclusion: </strong>The G2 gene expression signature and <i>MYC</i> expression were independently associated with unfavorable outcomes in a pediatric cohort of patients with high-grade osteosarcoma. The combined prognostic value warrants further prospective validation and could potentially serve as a stratification marker for future osteosarcoma treatment protocols.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400855"},"PeriodicalIF":5.3,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144181872","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 : 2025-05-01Epub Date: 2025-05-29DOI: 10.1200/PO-24-00475
Juan J Juarez-Vignon Whaley, Soravis Osataphan, Ben Ponvilawan, Nipith Charoenngam, Mary Linton Peters
{"title":"PD-L1 Expression in Biliary Tract Cancer: Comparison Across Antibody Clones and Role as a Predictor of Response to Chemoimmunotherapy: A Meta-Analysis.","authors":"Juan J Juarez-Vignon Whaley, Soravis Osataphan, Ben Ponvilawan, Nipith Charoenngam, Mary Linton Peters","doi":"10.1200/PO-24-00475","DOIUrl":"https://doi.org/10.1200/PO-24-00475","url":null,"abstract":"<p><strong>Purpose: </strong>PD-L1 positivity in biliary tract cancers (BTCs) is reported from 4% to 76%. BTC clinical trials have not demonstrated PD-L1 expression as a predictor of response to chemotherapy combined with immune checkpoint inhibitor (chemo-ICI). This meta-analysis examines PD-L1 positivity rates in BTC and association between PD-L1 expression and outcomes in patients treated with chemo-ICI.</p><p><strong>Materials and methods: </strong>Observational studies or clinical trials reporting tissue-based PD-L1 expression by Tumor Proportional Score/Combined Positive Score using immunohistochemistry were included. Clinical trials of BTC treated with chemo-ICI were included to assess PD-L1 expression on treatment response. PubMed, Embase, Web of Science, and Cochrane Library were searched for relevant studies before November 15, 2023. Methods of PD-L1 assessment, including antibody clone, cutoff for PD-L1 positivity, and anatomical subtype, were analyzed. Overall survival (OS) and objective response rates (ORRs) were the main outcomes. The generic inverse variance method and random-effect model were used to assess pooled effect sizes.</p><p><strong>Results: </strong>Fifty-six studies met eligibility criteria. Among 7,768 patients, pooled PD-L1 positivity was 30%. Positivity rates varied significantly by antibody clone (5H1, 58% <i>v</i> SP142, 17%; <i>P</i> = .02). Clinical trials reported a higher positivity rate compared with observational studies (48% <i>v</i> 26%; <i>P</i> < .01). Across five phase I/II clinical trials (194 patients), PD-L1 ≥1% patients tended to have a better ORR than PD-L1 <1% patients (64% <i>v</i> 46%; <i>P</i> = .08). In two randomized controlled trials (874 patients), PD-L1 ≥1% had a statistically significant improvement in OS (hazard ratio [HR], 0.83; <i>P</i> < .01), while PD-L1 <1% did not (HR, 0.85; <i>P</i> = .2).</p><p><strong>Conclusion: </strong>Given the high PD-L1 positivity rate seen in this study, as well as a possible signal of predictive response for chemo-ICI treatment, PD-L1 expression should be further explored as a predictive biomarker.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400475"},"PeriodicalIF":5.3,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144181615","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 : 2025-05-01Epub Date: 2025-05-15DOI: 10.1200/PO-24-00658
Kevin B Kim, Pierre-Yves Desprez, David de Semir, Rinette W L Woo, Anima Sharma, Robyn Jones, Chongshan Caressi, Mehdi Nosrati, Emilia Janiczek, Julia Rivera Penafiel, Mohammed Kashani-Sabet
{"title":"Phase II Study of Niraparib in Patients With Advanced Melanoma With Homologous Recombination Pathway Gene Mutations.","authors":"Kevin B Kim, Pierre-Yves Desprez, David de Semir, Rinette W L Woo, Anima Sharma, Robyn Jones, Chongshan Caressi, Mehdi Nosrati, Emilia Janiczek, Julia Rivera Penafiel, Mohammed Kashani-Sabet","doi":"10.1200/PO-24-00658","DOIUrl":"https://doi.org/10.1200/PO-24-00658","url":null,"abstract":"<p><strong>Purpose: </strong>Patients with metastatic melanoma who progress on checkpoint inhibitors and BRAF-targeting drugs have limited therapeutic options. Up to one third of melanomas harbor at least one molecular aberration in the homologous recombination (HR) pathway, leading to HR deficiency.</p><p><strong>Patients and methods: </strong>In this single-arm trial, we assessed the overall response rate to niraparib in patients with metastatic melanoma, harboring a genetic alteration in the HR pathway (<i>ARID1A/B</i>, <i>ARID2</i>, <i>ATM</i>, <i>ATR</i>, <i>ATRX</i>, <i>BARD1</i>, <i>BRCA1/2</i>, <i>BAP1</i>, <i>BRIP1</i>, <i>CHEK2</i>, <i>FANCD2</i>, <i>MRE11A</i>, <i>RAD50</i>, <i>RAD51</i>, <i>RAD54B</i>, or <i>PALB2</i>) who had disease progression after PD-1 blockade or BRAF/MEK inhibition if <i>BRAF</i>-mutant. Niraparib was administered orally at 300 mg or 200 mg daily, based on body weight and platelet count.</p><p><strong>Results: </strong>Fourteen patients were accrued to the trial, which was discontinued because of slow accrual. The median age was 71 years. Nine patients had an Eastern Cooperative Oncology Group performance status of 1. Eleven patients had elevated lactate dehydrogenase levels. Ten patients had nonuveal melanoma and four had uveal melanoma. Two (14%) had a partial response and seven (50%) had stable disease, with a disease control rate of 64%. The median progression-free survival was 16 weeks. Among the patients with nonuveal melanoma, two (20%) achieved partial response with a time to progression of 32 and 24 weeks, while five (50%) had stable disease lasting 16-98 weeks. None of the four patients with uveal melanoma responded. There were no unexpected adverse events related to niraparib treatment. Notably, one responder with an <i>ARID1A</i> mutation had detectable circulating tumor DNA at baseline, which became undetectable during treatment.</p><p><strong>Conclusion: </strong>Despite the small sample size, our results indicate a promising signal for single agent niraparib in patients with pretreated nonuveal metastatic melanoma with HR gene mutations.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2400658"},"PeriodicalIF":5.3,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144077879","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}