Pub Date : 2025-10-01Epub Date: 2025-10-23DOI: 10.1200/PO-25-00402
Pratik Chandra, Yi Song, Esther Drill, Alice C Wei, Nancy Kemeny, Andrea Cercek, Louise Connell, James Harding, Ghassan Abou-Alfa, Wungki Park, T Peter Kingham, Kevin Soares, Vinod Balachandran, Jeffrey Drebin, Michael D'Angelica, Eileen O'Reilly, Bas Groot Koerkamp, William R Jarnagin
Purpose: The impact of first recurrence site on outcome after resection of intrahepatic cholangiocarcinoma (IHC) is ill-defined, as are the genomic underpinnings of recurrence and outcomes.
Methods: Resected patients with IHC at two institutions with genomic data were included. Sites of first recurrence (SOFR) were classified as liver only (LO), extrahepatic (EH) only, or simultaneous liver and extrahepatic (SIM). Overall survival (OS) was calculated from the time of recurrence.
Results: Between 1993 and 2021, 318 patients met inclusion criteria; 232 (73%) recurred. SOFR were LO = 93 (40%), EH = 80 (34%), and SIM = 59 (26%). Median OS from recurrence was similar in the LO (33 [26, 42] months) and EH groups (33 [23, 46] months) but much lower in SIM (12 [9.8, 18] months; P < .001). Moderate/poor tumor differentiation, lymphovascular invasion, N1 disease, perineural invasion, and time to recurrence (all P < .05) were associated with SIM; only positive resection margin predicted LO (P = .007). No individual genomic or pathway alterations predicted SOFR; however, for all recurrers, TP53mut (n = 51, 22%; hazard ratio [HR], 2.0 [1.4 to 2.9]; P = .002), CDKN2Adel (n = 34, 15%; HR, 3.4 [95% CI, 2.2 to 5.3]; P < .001), CDKN2B (n = 25, 11%; HR, 3.2 [95% CI, 2.0 to 5.0]; P < .001), and KRASmut (n = 25, 11%; HR, 2.5 [95% CI, 1.6 to 4.0]; P = .002) were associated with worse OS. On multivariable analysis, SIM (HR, 2.5 [95% CI, 1.7 to 3.5]; P < .001), N1 status (HR, 1.8 [95% CI, 1.2 to 2.6]; P = .004), and alterations in the high-risk genotype (TP53mut, CDKN2Adel or KRASmut; n = 84, 36%; HR, 2.4 [95% CI, 1.7 to 3.3]; P < .001) were independent predictors of poor OS.
Conclusion: Recurrence after resection of IHC is common, and the liver was the most common site (66%). Clinicopathologic and genomic factors had limited ability to predict SOFR. Although LO and EH were associated with similar OS, SIM recurrences had dramatically worse OS. Adjuvant strategies targeting liver recurrence may improve outcomes after resection of IHC.
{"title":"Intrahepatic Cholangiocarcinoma: Recurrence Patterns, Genomics, and Survival.","authors":"Pratik Chandra, Yi Song, Esther Drill, Alice C Wei, Nancy Kemeny, Andrea Cercek, Louise Connell, James Harding, Ghassan Abou-Alfa, Wungki Park, T Peter Kingham, Kevin Soares, Vinod Balachandran, Jeffrey Drebin, Michael D'Angelica, Eileen O'Reilly, Bas Groot Koerkamp, William R Jarnagin","doi":"10.1200/PO-25-00402","DOIUrl":"10.1200/PO-25-00402","url":null,"abstract":"<p><strong>Purpose: </strong>The impact of first recurrence site on outcome after resection of intrahepatic cholangiocarcinoma (IHC) is ill-defined, as are the genomic underpinnings of recurrence and outcomes.</p><p><strong>Methods: </strong>Resected patients with IHC at two institutions with genomic data were included. Sites of first recurrence (SOFR) were classified as liver only (LO), extrahepatic (EH) only, or simultaneous liver and extrahepatic (SIM). Overall survival (OS) was calculated from the time of recurrence.</p><p><strong>Results: </strong>Between 1993 and 2021, 318 patients met inclusion criteria; 232 (73%) recurred. SOFR were LO = 93 (40%), EH = 80 (34%), and SIM = 59 (26%). Median OS from recurrence was similar in the LO (33 [26, 42] months) and EH groups (33 [23, 46] months) but much lower in SIM (12 [9.8, 18] months; <i>P</i> < .001). Moderate/poor tumor differentiation, lymphovascular invasion, N1 disease, perineural invasion, and time to recurrence (all <i>P</i> < .05) were associated with SIM; only positive resection margin predicted LO (<i>P</i> = .007). No individual genomic or pathway alterations predicted SOFR; however, for all recurrers, TP53<i>mut</i> (n = 51, 22%; hazard ratio [HR], 2.0 [1.4 to 2.9]; <i>P</i> = .002), CDKN2A<i>del</i> (n = 34, 15%; HR, 3.4 [95% CI, 2.2 to 5.3]; <i>P</i> < .001), CDKN2B (n = 25, 11%; HR, 3.2 [95% CI, 2.0 to 5.0]; <i>P</i> < .001), and KRAS<i>mut</i> (n = 25, 11%; HR, 2.5 [95% CI, 1.6 to 4.0]; <i>P</i> = .002) were associated with worse OS. On multivariable analysis, SIM (HR, 2.5 [95% CI, 1.7 to 3.5]; <i>P</i> < .001), N1 status (HR, 1.8 [95% CI, 1.2 to 2.6]; <i>P</i> = .004), and alterations in the high-risk genotype (TP53<i>mut</i>, CDKN2A<i>del</i> or KRAS<i>mut</i>; n = 84, 36%; HR, 2.4 [95% CI, 1.7 to 3.3]; <i>P</i> < .001) were independent predictors of poor OS.</p><p><strong>Conclusion: </strong>Recurrence after resection of IHC is common, and the liver was the most common site (66%). Clinicopathologic and genomic factors had limited ability to predict SOFR. Although LO and EH were associated with similar OS, SIM recurrences had dramatically worse OS. Adjuvant strategies targeting liver recurrence may improve outcomes after resection of IHC.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500402"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12614773/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145354903","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}
Pub Date : 2025-10-01Epub Date: 2025-10-23DOI: 10.1200/PO-25-00911
Maen Abdelrahim, Abdullah Esmail
{"title":"Reply to: Beyond Detection: Addressing Threshold Definition and Clinical Integration of Circulating Tumor DNA in Post-Curative Hepatocellular Carcinoma.","authors":"Maen Abdelrahim, Abdullah Esmail","doi":"10.1200/PO-25-00911","DOIUrl":"https://doi.org/10.1200/PO-25-00911","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500911"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145354827","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}
Pub Date : 2025-10-01Epub Date: 2025-10-30DOI: 10.1200/PO-25-00603
Hsin-Yi Chang, William Tap, Emily Slotkin, Leonard Wexler, Damon Reed, Meera Hameed, Chad Vanderbilt, Cristina R Antonescu
Purpose: Translocation-associated sarcomas (TASs) encompass a wide spectrum of pathologic entities and clinical behavior. Driven by the oncogenic fusion, TASs typically show a stable genome and a low tumor mutational burden (TMB), with infrequent secondary genetic alterations (SGAs). Although oncogenic/likely oncogenic SGAs have been associated with an aggressive clinical course in certain histotypes, a comprehensive comparative study of their clinical impact across TAS histotypes has not been performed to date.
Materials and methods: Herein, we investigate the genomic landscape of 632 TASs, spanning the most common histotypes: Ewing sarcoma (ES, n = 196), desmoplastic small round cell sarcoma (DSRCT, n = 115), solitary fibrous tumor (SFT, n = 87), synovial sarcoma (SS, n = 75), etc. All tumors were tested on a clinically validated, matched tumor-normal DNA-targeted next-generation sequencing panel, with confirmed gene fusion partners. Both gene-level and arm-level copy number alterations were assessed and correlated with survival.
Results: Overall, oncogenic SGAs were detected in 51% of patients, with the highest incidence in myxoid liposarcoma (MLS, 88%). Patients with oncogenic SGA had a higher TMB and were associated with distant metastasis and/or progression-free survival (PFS) in ES, SFT, SS, and MLS in the localized group. TP53 mutations were the most common oncogenic SGAs across histotypes, with SFT and ES showing the highest rate (26% and 11%) and associated with worse PFS and disease-specific survival (P < .01). TP53 mutations in ES (P < .01) and SFT (P = .045) and TERT promoter mutations in SFT (P < .001) and MLS (P = .049) correlated with metastasis in the localized group.
Conclusion: Overall, oncogenic SGA correlated with a worse survival in certain TAS types.
目的:易位相关性肉瘤(TASs)包含广泛的病理实体和临床行为。在致癌融合的驱动下,TASs通常表现出稳定的基因组和低肿瘤突变负担(TMB),并伴有罕见的继发性遗传改变(SGAs)。尽管在某些组织类型中,致癌/可能致癌的SGAs与侵袭性临床病程相关,但迄今为止尚未对其在TAS组织类型中的临床影响进行全面的比较研究。材料和方法:本文研究了632例TASs的基因组图谱,涵盖了最常见的组织类型:Ewing肉瘤(ES, n = 196)、结缔组织增生小圆细胞肉瘤(DSRCT, n = 115)、孤立性纤维瘤(SFT, n = 87)、滑膜肉瘤(SS, n = 75)等。所有肿瘤均在临床验证的、匹配的肿瘤正常dna靶向下一代测序面板上进行测试,并确认基因融合伙伴。评估了基因水平和手臂水平拷贝数的改变,并将其与生存率相关。结果:总体而言,51%的患者检测到致癌性SGAs,其中黏液样脂肪肉瘤(MLS)的发病率最高(88%)。在ES、SFT、SS和MLS的局部组中,癌性SGA患者有较高的TMB,并与远处转移和/或无进展生存(PFS)相关。TP53突变是所有组织类型中最常见的致癌SGAs,其中SFT和ES的发生率最高(26%和11%),并与较差的PFS和疾病特异性生存率相关(P < 0.01)。ES和SFT的TP53突变(P < 0.01)和MLS的TERT启动子突变(P < 0.001)与局部组转移相关(P = 0.049)。结论:总体而言,在某些TAS类型中,癌性SGA与较差的生存率相关。
{"title":"Spectrum and Clinical Impact of Secondary Genetic Alterations in Translocation-Associated Sarcomas.","authors":"Hsin-Yi Chang, William Tap, Emily Slotkin, Leonard Wexler, Damon Reed, Meera Hameed, Chad Vanderbilt, Cristina R Antonescu","doi":"10.1200/PO-25-00603","DOIUrl":"https://doi.org/10.1200/PO-25-00603","url":null,"abstract":"<p><strong>Purpose: </strong>Translocation-associated sarcomas (TASs) encompass a wide spectrum of pathologic entities and clinical behavior. Driven by the oncogenic fusion, TASs typically show a stable genome and a low tumor mutational burden (TMB), with infrequent secondary genetic alterations (SGAs). Although oncogenic/likely oncogenic SGAs have been associated with an aggressive clinical course in certain histotypes, a comprehensive comparative study of their clinical impact across TAS histotypes has not been performed to date.</p><p><strong>Materials and methods: </strong>Herein, we investigate the genomic landscape of 632 TASs, spanning the most common histotypes: Ewing sarcoma (ES, n = 196), desmoplastic small round cell sarcoma (DSRCT, n = 115), solitary fibrous tumor (SFT, n = 87), synovial sarcoma (SS, n = 75), etc. All tumors were tested on a clinically validated, matched tumor-normal DNA-targeted next-generation sequencing panel, with confirmed gene fusion partners. Both gene-level and arm-level copy number alterations were assessed and correlated with survival.</p><p><strong>Results: </strong>Overall, oncogenic SGAs were detected in 51% of patients, with the highest incidence in myxoid liposarcoma (MLS, 88%). Patients with oncogenic SGA had a higher TMB and were associated with distant metastasis and/or progression-free survival (PFS) in ES, SFT, SS, and MLS in the localized group. <i>TP53</i> mutations were the most common oncogenic SGAs across histotypes, with SFT and ES showing the highest rate (26% and 11%) and associated with worse PFS and disease-specific survival (<i>P</i> < .01). <i>TP53</i> mutations in ES (<i>P</i> < .01) and SFT (<i>P</i> = .045) and <i>TERT</i> promoter mutations in SFT (<i>P</i> < .001) and MLS (<i>P</i> = .049) correlated with metastasis in the localized group.</p><p><strong>Conclusion: </strong>Overall, oncogenic SGA correlated with a worse survival in certain TAS types.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500603"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145409229","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}
Pub Date : 2025-10-01Epub Date: 2025-10-09DOI: 10.1200/PO-25-00108
Seong-Eun Kim, Yongjae Kim, Hye Hyeon Moon, Soo Jeong Nam, Kang-Seo Park, Chang-Ki Hong, Ho-Su Lee, Shinkyo Yoon, Ji Eun Park
{"title":"Vabametkib Treatment in Refractory Glioblastoma: A Case Report.","authors":"Seong-Eun Kim, Yongjae Kim, Hye Hyeon Moon, Soo Jeong Nam, Kang-Seo Park, Chang-Ki Hong, Ho-Su Lee, Shinkyo Yoon, Ji Eun Park","doi":"10.1200/PO-25-00108","DOIUrl":"10.1200/PO-25-00108","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500108"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12513043/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145258360","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}
Pub Date : 2025-10-01Epub Date: 2025-12-18DOI: 10.1200/PO-25-00574
Abhishek Ajay, Reza Ferdousi, Peronne L Joseph, Seren Durer, Ali Rezvani, Zhengyi Chen, Gary M Wildey, Minh Lam, Pingfu Fu, Afshin Dowlati
Purpose: Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy associated with an exceptionally poor prognosis. A limitation in its standard-of-care management is the absence of practical molecular tools for monitoring disease progression. Circulating tumor DNA (ctDNA) has emerged as a promising biomarker with potential applications in prognostication, early detection of cancer recurrence, refined evaluation of treatment response, and improved disease surveillance.
Materials and methods: We analyzed serial plasma samples from 81 patients with SCLC using a 105-gene hybrid capture-based next-generation sequencing liquid biopsy assay at three key time points: diagnosis, postchemotherapy, and clinical relapse.
Results: Extensive-stage (ES) patients demonstrated significantly higher median baseline maximum variant allele frequency (VAFmax) compared with limited-stage cases. Notably, limited-stage patients with VAFmax >40% experienced outcomes similar to those with ES disease. Across the cohort, median VAFmax values declined from 53.30% at baseline to 0.15% during remission and then rose to 38.65% at relapse, reflecting initial therapeutic sensitivity followed by disease recurrence that remained unremitting. Baseline VAFmax correlated with the overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). Moreover, disease-free survival (DFS), defined as the time to relapse from the date of clinical response (evaluated post-4-cycle chemotherapy), was significantly associated with baseline VAFmax and a decline in VAFmax of <99.89% at remission. Relapse genomic profiles largely mirrored baseline alterations, although some patients showed novel mutations.
Conclusion: Median ctDNA VAFmax was strongly associated with OS, PFS, DFS, and ORR. Importantly, patients with detectable residual alterations at completion of prescribed chemotherapy exhibited a markedly shorter DFS, less than that of patients with lower detectable residual alterations, highlighting the potential utility of ctDNA in risk stratification and the value of early trial enrollment for high-risk subgroups.
{"title":"Comprehensive Analysis of the Value of Dynamic Changes in Circulating Tumor DNA in Small Cell Lung Cancer.","authors":"Abhishek Ajay, Reza Ferdousi, Peronne L Joseph, Seren Durer, Ali Rezvani, Zhengyi Chen, Gary M Wildey, Minh Lam, Pingfu Fu, Afshin Dowlati","doi":"10.1200/PO-25-00574","DOIUrl":"10.1200/PO-25-00574","url":null,"abstract":"<p><strong>Purpose: </strong>Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy associated with an exceptionally poor prognosis. A limitation in its standard-of-care management is the absence of practical molecular tools for monitoring disease progression. Circulating tumor DNA (ctDNA) has emerged as a promising biomarker with potential applications in prognostication, early detection of cancer recurrence, refined evaluation of treatment response, and improved disease surveillance.</p><p><strong>Materials and methods: </strong>We analyzed serial plasma samples from 81 patients with SCLC using a 105-gene hybrid capture-based next-generation sequencing liquid biopsy assay at three key time points: diagnosis, postchemotherapy, and clinical relapse.</p><p><strong>Results: </strong>Extensive-stage (ES) patients demonstrated significantly higher median baseline maximum variant allele frequency (VAF<sub>max</sub>) compared with limited-stage cases. Notably, limited-stage patients with VAF<sub>max</sub> >40% experienced outcomes similar to those with ES disease. Across the cohort, median VAF<sub>max</sub> values declined from 53.30% at baseline to 0.15% during remission and then rose to 38.65% at relapse, reflecting initial therapeutic sensitivity followed by disease recurrence that remained unremitting. Baseline VAF<sub>max</sub> correlated with the overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). Moreover, disease-free survival (DFS), defined as the time to relapse from the date of clinical response (evaluated post-4-cycle chemotherapy), was significantly associated with baseline VAF<sub>max</sub> and a decline in VAF<sub>max</sub> of <99.89% at remission. Relapse genomic profiles largely mirrored baseline alterations, although some patients showed novel mutations.</p><p><strong>Conclusion: </strong>Median ctDNA VAF<sub>max</sub> was strongly associated with OS, PFS, DFS, and ORR. Importantly, patients with detectable residual alterations at completion of prescribed chemotherapy exhibited a markedly shorter DFS, less than that of patients with lower detectable residual alterations, highlighting the potential utility of ctDNA in risk stratification and the value of early trial enrollment for high-risk subgroups.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500574"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12721678/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145781178","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}
Pub Date : 2025-10-01Epub Date: 2025-10-02DOI: 10.1200/PO-25-00510
Hongsik Kim, Chiyoon Oum, Soo Ick Cho, Wonkyung Jung, Hong Jae Chon, Myung Ah Lee, Hyeon-Su Im, Min Hwan Kim, Taekjin Nam, Chan-Young Ock, Hye Jin Choi, Choong-Kun Lee
Purpose: Despite recent advances in anti-human epidermal growth factor receptor 2 (HER2) treatments for HER2-positive biliary tract cancer (BTC), current guidelines lack clear thresholds for defining HER2 positivity in BTC. This study investigated the use of artificial intelligence (AI) to analyze HER2 expression and immune phenotypes (IP) in patients with HER2-positive BTC treated with anti-HER2 therapy.
Materials and methods: We conducted a post hoc analysis of a phase II trial (KCSG HB19-14) of trastuzumab plus folinic acid, fluorouracil, and oxaliplatin (FOLFOX) for HER2-positive BTC. AI-powered HER2 quantification and IP analyses were performed on whole-slide images of pretreatment samples. Clinical outcomes were analyzed on the basis of HER2 positivity using a continuous AI-based HER2 immunohistochemistry scoring system. Additionally, we evaluated the spatial distribution of tumor-infiltrating lymphocytes using AI-based IP analysis.
Results: Among 29 patients, the overall concordance rate between pathologists and the HER2-AI analyzer was 79.1%. AI-defined HER2-positivity status, characterized by a ≥30% H3 tumor cell proportion threshold, significantly predicted improved outcomes with trastuzumab plus FOLFOX (progression-free survival: 6.7 v 4.9 months, P = .039; overall survival: not reached v 8.4 months, P = .018). By contrast, traditional pathologist-based scoring did not stratify outcomes. AI-powered immune profiling revealed that HER2 3+ tumors predominantly exhibited immune-desert phenotypes, whereas HER2 2+ tumors displayed more inflamed phenotypes, potentially limiting the efficacy of current immunotherapy regimens for HER2 3+ BTC.
Conclusion: AI-powered HER2 quantification provides a refined biomarker for predicting the response to HER2-targeted therapies in BTC, proposing a ≥30% HER2 3+ tumor cell proportion threshold. Our findings highlight the potential of combining anti-HER2 therapy with immune checkpoint inhibitors on the basis of IP profiles.
{"title":"Artificial Intelligence-Powered Human Epidermal Growth Factor Receptor 2 Quantification and Clinical Outcomes in Human Epidermal Growth Factor Receptor 2-Positive Biliary Tract Cancer Treated With Trastuzumab Plus Folinic Acid, Fluorouracil, and Oxaliplatin.","authors":"Hongsik Kim, Chiyoon Oum, Soo Ick Cho, Wonkyung Jung, Hong Jae Chon, Myung Ah Lee, Hyeon-Su Im, Min Hwan Kim, Taekjin Nam, Chan-Young Ock, Hye Jin Choi, Choong-Kun Lee","doi":"10.1200/PO-25-00510","DOIUrl":"10.1200/PO-25-00510","url":null,"abstract":"<p><strong>Purpose: </strong>Despite recent advances in anti-human epidermal growth factor receptor 2 (HER2) treatments for HER2-positive biliary tract cancer (BTC), current guidelines lack clear thresholds for defining HER2 positivity in BTC. This study investigated the use of artificial intelligence (AI) to analyze HER2 expression and immune phenotypes (IP) in patients with HER2-positive BTC treated with anti-HER2 therapy.</p><p><strong>Materials and methods: </strong>We conducted a post hoc analysis of a phase II trial (KCSG HB19-14) of trastuzumab plus folinic acid, fluorouracil, and oxaliplatin (FOLFOX) for HER2-positive BTC. AI-powered HER2 quantification and IP analyses were performed on whole-slide images of pretreatment samples. Clinical outcomes were analyzed on the basis of HER2 positivity using a continuous AI-based HER2 immunohistochemistry scoring system. Additionally, we evaluated the spatial distribution of tumor-infiltrating lymphocytes using AI-based IP analysis.</p><p><strong>Results: </strong>Among 29 patients, the overall concordance rate between pathologists and the HER2-AI analyzer was 79.1%. AI-defined HER2-positivity status, characterized by a ≥30% H3 tumor cell proportion threshold, significantly predicted improved outcomes with trastuzumab plus FOLFOX (progression-free survival: 6.7 <i>v</i> 4.9 months, <i>P</i> = .039; overall survival: not reached <i>v</i> 8.4 months, <i>P</i> = .018). By contrast, traditional pathologist-based scoring did not stratify outcomes. AI-powered immune profiling revealed that HER2 3+ tumors predominantly exhibited immune-desert phenotypes, whereas HER2 2+ tumors displayed more inflamed phenotypes, potentially limiting the efficacy of current immunotherapy regimens for HER2 3+ BTC.</p><p><strong>Conclusion: </strong>AI-powered HER2 quantification provides a refined biomarker for predicting the response to HER2-targeted therapies in BTC, proposing a ≥30% HER2 3+ tumor cell proportion threshold. Our findings highlight the potential of combining anti-HER2 therapy with immune checkpoint inhibitors on the basis of IP profiles.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500510"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12506691/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145212728","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}
Pub Date : 2025-10-01Epub Date: 2025-12-18DOI: 10.1200/PO-25-00998
Wee Lee Chan, Masturah Bte Mohd Abdul Rashid, Rui Xue Lee, Sanjay de Mel, Edward Kai-Hua Chow, Anand D Jeyasekharan
{"title":"Reply to: Improving the Clinical Interpretability of Functional Drug Screens: A Suggestion for Standardized Clinical Decision Thresholds in Quadratic Phenotypic Optimization Platform.","authors":"Wee Lee Chan, Masturah Bte Mohd Abdul Rashid, Rui Xue Lee, Sanjay de Mel, Edward Kai-Hua Chow, Anand D Jeyasekharan","doi":"10.1200/PO-25-00998","DOIUrl":"https://doi.org/10.1200/PO-25-00998","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500998"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145781323","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}
Pub Date : 2025-10-01Epub Date: 2025-10-11DOI: 10.1200/PO-25-00696
Anjeli Manam, Jessica N Everett, Sigurdis Haraldsdottir, Phil A Hart, Vivek Kaul, Kelsey Klute, Kasmintan A Schrader, Daniel A Sussman, Bryson W Katona
Purpose: Pancreatic cancer (PC) surveillance is increasingly recommended for high-risk individuals, but there are remaining areas of uncertainty that are not consistently addressed by guidelines contributing to heterogeneity in clinical practice. Herein, we compare PC surveillance practices across sites in the international Pancreatic Cancer Early Detection (PRECEDE) Consortium to understand the application of eligibility criteria and testing strategies among experienced PC surveillance centers.
Methods: This analysis represents a cross-sectional survey administered in 2024 to PRECEDE institutions. The site principal investigator (or designee) completed the survey reflecting PC surveillance practices of their site, with one response per institution. Survey questions were related to surveillance eligibility for carriers of BRCA1/2, ATM, PALB2, and Lynch syndrome along with imaging approaches.
Results: Of the 57 PRECEDE sites, 54 (95%) completed the survey. For high-risk gene carriers, there was heterogeneity among sites with respect to whether family history of PC was used when assessing eligibility for surveillance. In the absence of family history, 44.4% and 35.2% of sites would offer PC surveillance to BRCA2 and BRCA1 carriers, respectively, whereas 31.5% and 13% would offer surveillance to PALB2/ATM and Lynch syndrome carriers, respectively. There was general consensus that surveillance should start at age 50 for men and women across all included genes. Magnetic resonance imaging with magnetic resonance cholangiopancreatography was used by 64.8% of sites as index imaging. The majority recommended an alternative modality if index imaging was unremarkable, and recommended annual surveillance imaging.
Conclusion: This is the largest assessment of global PC surveillance practices to date, showing variability in practice patterns. Additional investigation is needed to refine risk stratification for gene carriers without a family history of PC and address optimal imaging strategies.
{"title":"Areas of Uncertainty in Pancreatic Cancer Surveillance: A Survey Across the International Pancreatic Cancer Early Detection (PRECEDE) Consortium.","authors":"Anjeli Manam, Jessica N Everett, Sigurdis Haraldsdottir, Phil A Hart, Vivek Kaul, Kelsey Klute, Kasmintan A Schrader, Daniel A Sussman, Bryson W Katona","doi":"10.1200/PO-25-00696","DOIUrl":"https://doi.org/10.1200/PO-25-00696","url":null,"abstract":"<p><strong>Purpose: </strong>Pancreatic cancer (PC) surveillance is increasingly recommended for high-risk individuals, but there are remaining areas of uncertainty that are not consistently addressed by guidelines contributing to heterogeneity in clinical practice. Herein, we compare PC surveillance practices across sites in the international Pancreatic Cancer Early Detection (PRECEDE) Consortium to understand the application of eligibility criteria and testing strategies among experienced PC surveillance centers.</p><p><strong>Methods: </strong>This analysis represents a cross-sectional survey administered in 2024 to PRECEDE institutions. The site principal investigator (or designee) completed the survey reflecting PC surveillance practices of their site, with one response per institution. Survey questions were related to surveillance eligibility for carriers of <i>BRCA1/2</i>, <i>ATM</i>, <i>PALB2</i>, and Lynch syndrome along with imaging approaches.</p><p><strong>Results: </strong>Of the 57 PRECEDE sites, 54 (95%) completed the survey. For high-risk gene carriers, there was heterogeneity among sites with respect to whether family history of PC was used when assessing eligibility for surveillance. In the absence of family history, 44.4% and 35.2% of sites would offer PC surveillance to <i>BRCA2</i> and <i>BRCA1</i> carriers, respectively, whereas 31.5% and 13% would offer surveillance to <i>PALB2/ATM</i> and Lynch syndrome carriers, respectively. There was general consensus that surveillance should start at age 50 for men and women across all included genes. Magnetic resonance imaging with magnetic resonance cholangiopancreatography was used by 64.8% of sites as index imaging. The majority recommended an alternative modality if index imaging was unremarkable, and recommended annual surveillance imaging.</p><p><strong>Conclusion: </strong>This is the largest assessment of global PC surveillance practices to date, showing variability in practice patterns. Additional investigation is needed to refine risk stratification for gene carriers without a family history of PC and address optimal imaging strategies.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500696"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145274556","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}
Pub Date : 2025-10-01Epub Date: 2025-10-11DOI: 10.1200/PO-25-00700
Asaf Maoz, Leah Biller, Marios Giannakis, Douglas Rubinson, Thejus Jayakrishnan, Miki Horiguchi, Chinedu Ukaegbu, Alyson Caruso, Sapna Syngal, Matthew B Yurgelun
Purpose: Individuals with Lynch syndrome (LS) predominantly develop mismatch repair-deficient/microsatellite-unstable (MMR-D/MSI-H) cancer, for which immunotherapy can yield long-term disease control, even in the metastatic setting, leading to tissue-agnostic US Food and Drug Administration approval for pembrolizumab for any advanced MMR-D/MSI-H cancer in May 2017. The primary aim of this study was to assess the causes of death in individuals with LS in the era of immunotherapy.
Methods: We performed a retrospective cohort study of individuals with LS seen at a single academic cancer center for oncology-/genetics-related care, who died between May 30, 2017, and December 20, 2024. An expert panel adjudicated the cause of death after a detailed review of the electronic medical record (EMR). Data regarding personal cancer history, germline and somatic testing, and treatment were obtained from the EMR.
Results: Fifty-four individuals with LS were known to have died since May 2017 (52% female; median age at death 64 years; IQR, 51-75), of whom 44 were determined to have died of cancer and/or cancer treatment toxicities/complications. Of these, 19 of 44 (43%) died of MMR-proficient/MS-stable cancer (five because of pancreatic cancer, four because of colorectal cancer), 20 of 44 (46%) died of MMR-D/MSI-H cancer (six because of colorectal cancer, four because of pancreatic cancer), and 5 of 44 (11%) died of cancer with unknown/indeterminate MMR/MSI status. Among those who died with MMR-D/MSI-H cancer, 14 of 20 (70%) had disease progression on immunotherapy, 2 of 20 (10%) died of immunotherapy toxicity, and 3 of 20 (15%) died of other treatment complications. Deaths from MMR-proficient/MS-stable cancer were seen among patients with MSH2-, MSH6-, and PMS2-associated LS, but not MLH1-associated LS.
Conclusion: In this single-center analysis, MMR-proficient/MS-stable cancers were a frequent cause of death among individuals with LS. MMR-D/MSI-H cancers resistant to immunotherapy and treatment-associated deaths were other significant contributors to mortality. Larger, multicenter studies are warranted to validate these findings.
目的:Lynch综合征(LS)患者主要发展为错配修复缺陷/微卫星不稳定(MMR-D/MSI-H)癌症,免疫治疗可以产生长期疾病控制,即使在转移性情况下,导致美国食品和药物管理局于2017年5月批准派姆单抗用于任何晚期MMR-D/MSI-H癌症。本研究的主要目的是评估免疫治疗时代LS患者的死亡原因。方法:我们对2017年5月30日至2024年12月20日期间在单一学术癌症中心接受肿瘤/遗传学相关治疗的LS患者进行了回顾性队列研究。一个专家小组在详细审查了电子医疗记录(EMR)后,裁定了死因。从EMR中获得有关个人癌症病史、生殖系和体细胞检测以及治疗的数据。结果:自2017年5月以来,已知有54名LS患者死亡(52%为女性;死亡时中位年龄64岁;IQR, 51-75岁),其中44人被确定死于癌症和/或癌症治疗毒性/并发症。其中,44人中有19人(43%)死于MMR熟练/MSI稳定的癌症(5人死于胰腺癌,4人死于结直肠癌),44人中有20人(46%)死于MMR- d /MSI- h癌症(6人死于结直肠癌,4人死于胰腺癌),44人中有5人(11%)死于MMR/MSI状态未知/不确定的癌症。在死于MMR-D/MSI-H癌症的患者中,20人中有14人(70%)在免疫治疗中出现疾病进展,20人中有2人(10%)死于免疫治疗毒性,20人中有3人(15%)死于其他治疗并发症。MSH2-、MSH6-和pms2相关的LS患者死于mmr熟练/ ms稳定的癌症,而mlh1相关的LS患者则没有。结论:在这个单中心分析中,mmr熟练/ ms稳定的癌症是LS患者死亡的常见原因。免疫治疗耐药的MMR-D/MSI-H癌症和治疗相关死亡是死亡率的其他重要因素。需要更大规模的多中心研究来验证这些发现。
{"title":"Causes of Death Among Individuals With Lynch Syndrome in the Immunotherapy Era.","authors":"Asaf Maoz, Leah Biller, Marios Giannakis, Douglas Rubinson, Thejus Jayakrishnan, Miki Horiguchi, Chinedu Ukaegbu, Alyson Caruso, Sapna Syngal, Matthew B Yurgelun","doi":"10.1200/PO-25-00700","DOIUrl":"https://doi.org/10.1200/PO-25-00700","url":null,"abstract":"<p><strong>Purpose: </strong>Individuals with Lynch syndrome (LS) predominantly develop mismatch repair-deficient/microsatellite-unstable (MMR-D/MSI-H) cancer, for which immunotherapy can yield long-term disease control, even in the metastatic setting, leading to tissue-agnostic US Food and Drug Administration approval for pembrolizumab for any advanced MMR-D/MSI-H cancer in May 2017. The primary aim of this study was to assess the causes of death in individuals with LS in the era of immunotherapy.</p><p><strong>Methods: </strong>We performed a retrospective cohort study of individuals with LS seen at a single academic cancer center for oncology-/genetics-related care, who died between May 30, 2017, and December 20, 2024. An expert panel adjudicated the cause of death after a detailed review of the electronic medical record (EMR). Data regarding personal cancer history, germline and somatic testing, and treatment were obtained from the EMR.</p><p><strong>Results: </strong>Fifty-four individuals with LS were known to have died since May 2017 (52% female; median age at death 64 years; IQR, 51-75), of whom 44 were determined to have died of cancer and/or cancer treatment toxicities/complications. Of these, 19 of 44 (43%) died of MMR-proficient/MS-stable cancer (five because of pancreatic cancer, four because of colorectal cancer), 20 of 44 (46%) died of MMR-D/MSI-H cancer (six because of colorectal cancer, four because of pancreatic cancer), and 5 of 44 (11%) died of cancer with unknown/indeterminate MMR/MSI status. Among those who died with MMR-D/MSI-H cancer, 14 of 20 (70%) had disease progression on immunotherapy, 2 of 20 (10%) died of immunotherapy toxicity, and 3 of 20 (15%) died of other treatment complications. Deaths from MMR-proficient/MS-stable cancer were seen among patients with <i>MSH2</i>-, <i>MSH6</i>-, and <i>PMS2</i>-associated LS, but not <i>MLH1</i>-associated LS.</p><p><strong>Conclusion: </strong>In this single-center analysis, MMR-proficient/MS-stable cancers were a frequent cause of death among individuals with LS. MMR-D/MSI-H cancers resistant to immunotherapy and treatment-associated deaths were other significant contributors to mortality. Larger, multicenter studies are warranted to validate these findings.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"9 ","pages":"e2500700"},"PeriodicalIF":5.6,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145274592","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}