Kortnye Smith, Sophie O'Haire, Benjamin Markman, Hui K Gan, Kenneth O'Byrne, Michael Millward, Ben Tran, Benjamin J Solomon, Clare Scott, Damien Kee, Grant McArthur, Andrew Fellowes, Dong Anh K Khoung-Quang, Paul Ekert, Paul James, Huiling Xu, Melissa Martyn, Elly Lynch, Rona Weerasuriya, Clara Gaff, Stephen B Fox, Jayesh Desai
Purpose: Despite increasing evidence of benefit supporting complex genomic sequencing (CGS) in personalizing cancer therapy, its widespread uptake remains limited.
Methods: This mixed-methods, prospective cross-institutional demonstration study was designed to evaluate implementation of CGS in the care of patients with advanced cancer. DNA sequencing was undertaken on formalin-fixed paraffin-embedded tumor and matched blood was completed with the Peter MacCallum Cancer Centre Comprehensive Cancer Panel; 391 genes via central laboratory. Oncologists performed consent and result delivery. Patients completed pre- and post-test surveys, including validated and study-specific questions and, if eligible, semistructured interviews. Qualitative interviews were undertaken with study clinicians to evaluate processes.
Results: One hundred ninety-nine (63%) had ≥1 finding with the potential to affect management, including 172 (55%) whose finding could affect their treatment options, 25 (8%) whose test led to the resolution of diagnostic ambiguity, and 49 (16%) with a pathogenic germline variant. In 6-month follow-up, 50 (16%) participants had their subsequent therapy changed on the basis of their CGS results. Two hundred ninety-three (88% of adult patients) completed surveys at three time points. At consent, patients cited multifaceted value in testing, showed good understanding of basic concepts, but most (69%) overestimated the likelihood of result-led change. Post-test patients remained consistently satisfied with accessing CGS. 21% struggled with understanding results but there were low levels of decisional regret after participation (89% had nil/mild regret). Clinicians cited collaboration and communication as critical to delivery.
Conclusion: Patients undergoing CGS are generally satisfied and place value on its use beyond potential therapeutic benefit. Our results suggest that to improve test utility and delivery of CGS with value to patients and investing institutions, focus must be placed on addressing the additional barriers to its wider implications including efforts to improve process efficiencies, clinician genomic literacy, and decision-making support.
目的:尽管有越来越多的证据表明复杂基因组测序(CGS)有利于癌症治疗的个性化,但其广泛应用仍然有限:这项混合方法的前瞻性跨机构示范研究旨在评估 CGS 在晚期癌症患者治疗中的应用情况。对经福尔马林固定的石蜡包埋的肿瘤进行 DNA 测序,并通过中心实验室用 Peter MacCallum 癌症中心癌症综合样本(Peter MacCallum Cancer Centre Comprehensive Cancer Panel; 391 genes)完成配血。由肿瘤学家签署同意书并提供结果。患者完成测试前和测试后的调查,包括经过验证的特定研究问题,如果符合条件,还可进行半结构化访谈。对研究临床医生进行了定性访谈,以评估流程:199人(63%)有≥1项可能影响治疗的结果,其中172人(55%)的结果可能会影响他们的治疗选择,25人(8%)通过检测解决了诊断不明确的问题,49人(16%)有致病性种系变异。在 6 个月的随访中,有 50 人(16%)根据 CGS 结果改变了后续治疗方案。293名患者(占成年患者的88%)在三个时间点完成了调查。在同意测试时,患者认为测试具有多方面的价值,对基本概念有很好的理解,但大多数患者(69%)高估了由结果导致改变的可能性。测试后,患者仍对使用 CGS 表示满意。21%的患者在理解结果方面遇到困难,但参与测试后对决定的后悔程度较低(89%的患者无/轻微后悔)。临床医生认为合作与沟通对治疗至关重要:结论:接受 CGS 检查的患者普遍感到满意,他们对 CGS 的使用价值超出了潜在的治疗效果。我们的研究结果表明,要提高 CGS 的测试效用并为患者和投资机构带来价值,就必须重点解决影响 CGS 广泛应用的其他障碍,包括努力提高流程效率、临床医生基因组知识和决策支持。
{"title":"Patient Experience of Complex Genomic Sequencing Exploring Patient Preference, Barriers, and Enablers for Delivery.","authors":"Kortnye Smith, Sophie O'Haire, Benjamin Markman, Hui K Gan, Kenneth O'Byrne, Michael Millward, Ben Tran, Benjamin J Solomon, Clare Scott, Damien Kee, Grant McArthur, Andrew Fellowes, Dong Anh K Khoung-Quang, Paul Ekert, Paul James, Huiling Xu, Melissa Martyn, Elly Lynch, Rona Weerasuriya, Clara Gaff, Stephen B Fox, Jayesh Desai","doi":"10.1200/PO.23.00247","DOIUrl":"https://doi.org/10.1200/PO.23.00247","url":null,"abstract":"<p><strong>Purpose: </strong>Despite increasing evidence of benefit supporting complex genomic sequencing (CGS) in personalizing cancer therapy, its widespread uptake remains limited.</p><p><strong>Methods: </strong>This mixed-methods, prospective cross-institutional demonstration study was designed to evaluate implementation of CGS in the care of patients with advanced cancer. DNA sequencing was undertaken on formalin-fixed paraffin-embedded tumor and matched blood was completed with the Peter MacCallum Cancer Centre Comprehensive Cancer Panel; 391 genes via central laboratory. Oncologists performed consent and result delivery. Patients completed pre- and post-test surveys, including validated and study-specific questions and, if eligible, semistructured interviews. Qualitative interviews were undertaken with study clinicians to evaluate processes.</p><p><strong>Results: </strong>One hundred ninety-nine (63%) had ≥1 finding with the potential to affect management, including 172 (55%) whose finding could affect their treatment options, 25 (8%) whose test led to the resolution of diagnostic ambiguity, and 49 (16%) with a pathogenic germline variant. In 6-month follow-up, 50 (16%) participants had their subsequent therapy changed on the basis of their CGS results. Two hundred ninety-three (88% of adult patients) completed surveys at three time points. At consent, patients cited multifaceted value in testing, showed good understanding of basic concepts, but most (69%) overestimated the likelihood of result-led change. Post-test patients remained consistently satisfied with accessing CGS. 21% struggled with understanding results but there were low levels of decisional regret after participation (89% had nil/mild regret). Clinicians cited collaboration and communication as critical to delivery.</p><p><strong>Conclusion: </strong>Patients undergoing CGS are generally satisfied and place value on its use beyond potential therapeutic benefit. Our results suggest that to improve test utility and delivery of CGS with value to patients and investing institutions, focus must be placed on addressing the additional barriers to its wider implications including efforts to improve process efficiencies, clinician genomic literacy, and decision-making support.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2300247"},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142107585","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}
Purpose: Panel-based comprehensive genomic profiling (CGP) is used in clinical practice worldwide; however, large real-world data (RWD) of patients with advanced small intestine cancer have not been characterized. We investigated differences in the prevalence of clinically relevant alterations across molecularly defined or age-stratified subgroups.
Patients and methods: This was a collaborative biomarker study of RWD from CGP testing (Foundation Medicine, Inc). Hybrid capture was conducted on at least 324 cancer-related genes and select introns from up to 31 genes frequently rearranged in cancer. Overall, 1,364 patients with advanced small intestine cancer were available for analyses and were stratified by age (≥40 years/<40 years), microsatellite instability (MSI) status, tumor mutational burden (TMB) status (high ≥10/low <10 Muts/Mb), and select gene alterations. The frequency of alterations was analyzed using a chi-square test with Yate's correction.
Results: Genes with frequent alterations included TP53 (59.8%), KRAS (54.8%), APC (27.7%), and CDKN2A (22.4%). Frequent genes with amplifications were MYC (6.7%), MDM2 (5.9%), GATA6 (5.5%), and CCND1 (3.4%). Patients younger than 40 years had significantly lower frequency of APC mutations than those 40 years and older (10.4% v 28.7%; P = .0008). Druggable genomic alterations were detected in 22.3% of patients: BRAF V600E (1.2%), BRCA1 (1.8%), BRCA2 (3.2%), ERBB2 amplification (3.2%), KRAS G12C (3.3%), NTRK1/2/3 fusion (0.07%), MSI-high (7.0%), and TMB-high (12.2%), with no significant differences in the frequency according to age (<40 years v ≥40 years; 22.1% v 22.3%). TMB of 10-20 Mut/Mb was observed in 4.8% of patients, and TMB ≥20 Mut/Mb was seen in 7.3% of the cohort.
Conclusion: RWD from clinical panel testing revealed the genomic landscape in small intestine cancer by subgroup. These findings provide insights for the future development of treatments in advanced small intestine cancer.
{"title":"Genomic Profiling of Small Intestine Cancers From a Real-World Data Set Identifies Subgroups With Actionable Alterations.","authors":"Hiroyuki Takeda, Hiroyuki Yamamoto, Ritsuko Oikawa, Kumiko Umemoto, Hiroyuki Arai, Takuro Mizukami, Kazuki Ogawa, Yoshiyasu Uchida, Yusuke Nagata, Yohei Kubota, Ayako Doi, Yoshiki Horie, Takashi Ogura, Naoki Izawa, Jay A Moore, Ethan S Sokol, Yu Sunakawa","doi":"10.1200/PO.23.00425","DOIUrl":"https://doi.org/10.1200/PO.23.00425","url":null,"abstract":"<p><strong>Purpose: </strong>Panel-based comprehensive genomic profiling (CGP) is used in clinical practice worldwide; however, large real-world data (RWD) of patients with advanced small intestine cancer have not been characterized. We investigated differences in the prevalence of clinically relevant alterations across molecularly defined or age-stratified subgroups.</p><p><strong>Patients and methods: </strong>This was a collaborative biomarker study of RWD from CGP testing (Foundation Medicine, Inc). Hybrid capture was conducted on at least 324 cancer-related genes and select introns from up to 31 genes frequently rearranged in cancer. Overall, 1,364 patients with advanced small intestine cancer were available for analyses and were stratified by age (≥40 years/<40 years), microsatellite instability (MSI) status, tumor mutational burden (TMB) status (high ≥10/low <10 Muts/Mb), and select gene alterations. The frequency of alterations was analyzed using a chi-square test with Yate's correction.</p><p><strong>Results: </strong>Genes with frequent alterations included <i>TP53</i> (59.8%), <i>KRAS</i> (54.8%), <i>APC</i> (27.7%), and <i>CDKN2A</i> (22.4%). Frequent genes with amplifications were <i>MYC</i> (6.7%), <i>MDM2</i> (5.9%), <i>GATA6</i> (5.5%), and <i>CCND1</i> (3.4%). Patients younger than 40 years had significantly lower frequency of <i>APC</i> mutations than those 40 years and older (10.4% <i>v</i> 28.7%; <i>P</i> = .0008). Druggable genomic alterations were detected in 22.3% of patients: <i>BRAF</i> V600E (1.2%), <i>BRCA1</i> (1.8%), <i>BRCA2</i> (3.2%), <i>ERBB2</i> amplification (3.2%), <i>KRAS</i> G12C (3.3%), <i>NTRK1/2/3</i> fusion (0.07%), MSI-high (7.0%), and TMB-high (12.2%), with no significant differences in the frequency according to age (<40 years <i>v</i> ≥40 years; 22.1% <i>v</i> 22.3%). TMB of 10-20 Mut/Mb was observed in 4.8% of patients, and TMB ≥20 Mut/Mb was seen in 7.3% of the cohort.</p><p><strong>Conclusion: </strong>RWD from clinical panel testing revealed the genomic landscape in small intestine cancer by subgroup. These findings provide insights for the future development of treatments in advanced small intestine cancer.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2300425"},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906691","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}
Alberto Hernando-Calvo, S Y Cindy Yang, Maria Vila-Casadesús, Ming Han, Zhihui Amy Liu, A Hal K Berman, Anna Spreafico, Albiruni Abdul Razak, Stephanie Lheureux, Aaron R Hansen, Deborah Lo Giacco, Farnoosh Abbas-Aghababazadeh, Judith Matito, Benjamin Haibe-Kains, Trevor J Pugh, Scott V Bratman, Alexey Aleshin, Roger Berche, Omar Saavedra, Elena Garralda, Sawako Elston, Lillian L Siu, Pamela S Ohashi, Ana Vivancos, Philippe L Bedard
Purpose: Immune gene expression signatures are emerging as potential biomarkers for immunotherapy (IO). VIGex is a 12-gene expression classifier developed in both nCounter (Nanostring) and RNA sequencing (RNA-seq) assays and analytically validated across laboratories. VIGex classifies tumor samples into hot, intermediate-cold (I-Cold), and cold subgroups. VIGex-Hot has been associated with better IO treatment outcomes. Here, we investigated the performance of VIGex and other IO biomarkers in an independent data set of patients treated with pembrolizumab in the INSPIRE phase II clinical trial (ClinicalTrials.gov identifier: NCT02644369).
Materials and methods: Patients with advanced solid tumors were treated with pembrolizumab 200 mg IV once every 3 weeks. Tumor RNA-seq data from baseline tumor samples were classified by the VIGex algorithm. Circulating tumor DNA (ctDNA) was measured at baseline and start of cycle 3 using the bespoke Signatera assay. VIGex-Hot was compared with VIGex I-Cold + Cold and four groups were defined on the basis of the combination of VIGex subgroups and the change in ctDNA at cycle 3 from baseline (ΔctDNA).
Results: Seventy-six patients were enrolled, including 16 ovarian, 12 breast, 12 head and neck cancers, 10 melanoma, and 26 other tumor types. Objective response rate was 24% in VIGex-Hot and 10% in I-Cold/Cold. VIGex-Hot subgroup was associated with higher overall survival (OS) and progression-free survival (PFS) when included in a multivariable model adjusted for tumor type, tumor mutation burden, and PD-L1 immunohistochemistry. The addition of ΔctDNA improved the predictive performance of the baseline VIGex classification for both OS and PFS.
Conclusion: Our data indicate that the addition of ΔctDNA to baseline VIGex may refine prediction for IO.
{"title":"Combined Transcriptome and Circulating Tumor DNA Longitudinal Biomarker Analysis Associates With Clinical Outcomes in Advanced Solid Tumors Treated With Pembrolizumab.","authors":"Alberto Hernando-Calvo, S Y Cindy Yang, Maria Vila-Casadesús, Ming Han, Zhihui Amy Liu, A Hal K Berman, Anna Spreafico, Albiruni Abdul Razak, Stephanie Lheureux, Aaron R Hansen, Deborah Lo Giacco, Farnoosh Abbas-Aghababazadeh, Judith Matito, Benjamin Haibe-Kains, Trevor J Pugh, Scott V Bratman, Alexey Aleshin, Roger Berche, Omar Saavedra, Elena Garralda, Sawako Elston, Lillian L Siu, Pamela S Ohashi, Ana Vivancos, Philippe L Bedard","doi":"10.1200/PO.24.00100","DOIUrl":"10.1200/PO.24.00100","url":null,"abstract":"<p><strong>Purpose: </strong>Immune gene expression signatures are emerging as potential biomarkers for immunotherapy (IO). VIGex is a 12-gene expression classifier developed in both nCounter (Nanostring) and RNA sequencing (RNA-seq) assays and analytically validated across laboratories. VIGex classifies tumor samples into hot, intermediate-cold (I-Cold), and cold subgroups. VIGex-Hot has been associated with better IO treatment outcomes. Here, we investigated the performance of VIGex and other IO biomarkers in an independent data set of patients treated with pembrolizumab in the INSPIRE phase II clinical trial (ClinicalTrials.gov identifier: NCT02644369).</p><p><strong>Materials and methods: </strong>Patients with advanced solid tumors were treated with pembrolizumab 200 mg IV once every 3 weeks. Tumor RNA-seq data from baseline tumor samples were classified by the VIGex algorithm. Circulating tumor DNA (ctDNA) was measured at baseline and start of cycle 3 using the bespoke Signatera assay. VIGex-Hot was compared with VIGex I-Cold + Cold and four groups were defined on the basis of the combination of VIGex subgroups and the change in ctDNA at cycle 3 from baseline (ΔctDNA).</p><p><strong>Results: </strong>Seventy-six patients were enrolled, including 16 ovarian, 12 breast, 12 head and neck cancers, 10 melanoma, and 26 other tumor types. Objective response rate was 24% in VIGex-Hot and 10% in I-Cold/Cold. VIGex-Hot subgroup was associated with higher overall survival (OS) and progression-free survival (PFS) when included in a multivariable model adjusted for tumor type, tumor mutation burden, and PD-L1 immunohistochemistry. The addition of ΔctDNA improved the predictive performance of the baseline VIGex classification for both OS and PFS.</p><p><strong>Conclusion: </strong>Our data indicate that the addition of ΔctDNA to baseline VIGex may refine prediction for IO.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2400100"},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11371115/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142043912","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}
Lilie L Lin, Franklin Wong, Ruitao Lin, Timothy Yap, Jennifer K Litton
Purpose: We tested the ability of [18F] fluorthanatrace (FTT), a radiolabeled analog of poly(ADP-ribose) polymerase (PARP)-1 inhibitors, to demonstrate target engagement on positron emission tomography (PET) scans from patients with newly diagnosed primary breast cancer receiving the PARP inhibitor (PARPi) talazoparib.
Methods: Seven patients with germline BRCA1/2 pathogenic variants underwent [18F]FTT PET-computed tomography scanning at baseline, and five underwent repeat scanning 14 days after talazoparib initiation. Maximum uptake on PET was quantified in the primary tumor, involved nodes, contralateral pectoralis muscle, and lumbar vertebra body level 3, and compared between the two time points.
Results: Blocking of [18F]FTT was observed on the second scan. Potentially strong but nonsignificant correlations were found between changes in tumor volume (on ultrasound at 1 month v baseline) and percentage changes in tumor-to-muscle uptake ratio at 14 days from baseline (Spearman rank correlation coefficient r = 1; P = .083); and between the highest-grade hematologic toxicity and baseline bone marrow-to-muscle (B/M) uptake ratio (r = 0.72; P = .068) and percentage change in B/M ratio at 14 days from baseline (r = 0.87; P = .058).
Conclusion: We conclude that [18F]FTT can image target engagement by PARPi, but larger studies are needed to determine whether [18F]FTT uptake can predict response to PARPi and whether uptake of [18F]FTT in bone marrow may be an early predictor of hematologic toxicity.
{"title":"Pharmacodynamic Activity of [<sup>18</sup>F]-Fluorthanatrace Poly(ADP-ribose) Polymerase Positron Emission Tomography in Patients With <i>BRCA1/2</i>-Mutated Breast Cancer Receiving Talazoparib.","authors":"Lilie L Lin, Franklin Wong, Ruitao Lin, Timothy Yap, Jennifer K Litton","doi":"10.1200/PO.24.00303","DOIUrl":"10.1200/PO.24.00303","url":null,"abstract":"<p><strong>Purpose: </strong>We tested the ability of [<sup>18</sup>F] fluorthanatrace (FTT), a radiolabeled analog of poly(ADP-ribose) polymerase (PARP)-1 inhibitors, to demonstrate target engagement on positron emission tomography (PET) scans from patients with newly diagnosed primary breast cancer receiving the PARP inhibitor (PARPi) talazoparib.</p><p><strong>Methods: </strong>Seven patients with germline <i>BRCA1/2</i> pathogenic variants underwent [<sup>18</sup>F]FTT PET-computed tomography scanning at baseline, and five underwent repeat scanning 14 days after talazoparib initiation. Maximum uptake on PET was quantified in the primary tumor, involved nodes, contralateral pectoralis muscle, and lumbar vertebra body level 3, and compared between the two time points.</p><p><strong>Results: </strong>Blocking of [<sup>18</sup>F]FTT was observed on the second scan. Potentially strong but nonsignificant correlations were found between changes in tumor volume (on ultrasound at 1 month <i>v</i> baseline) and percentage changes in tumor-to-muscle uptake ratio at 14 days from baseline (Spearman rank correlation coefficient <i>r</i> = 1; <i>P</i> = .083); and between the highest-grade hematologic toxicity and baseline bone marrow-to-muscle (B/M) uptake ratio (<i>r</i> = 0.72; <i>P</i> = .068) and percentage change in B/M ratio at 14 days from baseline (<i>r</i> = 0.87; <i>P</i> = .058).</p><p><strong>Conclusion: </strong>We conclude that [<sup>18</sup>F]FTT can image target engagement by PARPi, but larger studies are needed to determine whether [<sup>18</sup>F]FTT uptake can predict response to PARPi and whether uptake of [<sup>18</sup>F]FTT in bone marrow may be an early predictor of hematologic toxicity.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2400303"},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142107586","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}
Jason M Baron, Sarrah Widatalla, Matthew A Gubens, Farah Khalil
Purpose: Patients with metastatic or advanced non-small cell lung cancer (NSCLC) need biomarker testing, including, in most cases, anaplastic lymphoma kinase (ALK), epidermal growth factor receptor (EGFR), and PD-L1, to identify options for targeted therapies and to optimally incorporate immune checkpoint inhibitors into therapeutic regimens. We sought to examine real-world patterns of biomarker testing, quantify interphysician practice variation, and correlate testing with clinical outcomes.
Methods: We extracted real-world data from a nationwide electronic health record-derived deidentified database from 17,165 patients diagnosed with advanced NSCLC between 2018 and 2021 and receiving care in the community setting. We analyzed data using descriptive analyses, fixed- and mixed-effects logistic regression models, and proportional hazard models.
Results: Only 67% of all 17,165 patients and 77% of patients with nonsquamous, metastatic NSCLC had ALK, EGFR, and PD-L1 testing within 90 days of diagnosis. Later diagnosis year (2019-2021 compared with 2018) was associated with higher rates of ALK, EGFR, and PD-L1 testing; stage IIIB/C disease (compared with stage IV), squamous histology, and Black or African American race were associated with lower rates. Interphysician variation was substantial with a median odds ratio between physicians (adjusted for patient factors) of 1.78 for ALK, EGFR, and PD-L1 testing. Patients with nonsquamous, metastatic NSCLC had significantly prolonged survival if tested with all three biomarkers (median, 364 days for all three v 180 for none of the three; hazard ratio, 0.67; P < .001).
Conclusion: Rates of biomarker testing appear suboptimal with substantial interphysician variation. Testing correlates with improved survival, although causality cannot be proven from this study. Additional work is needed to address the underlying causes of suboptimal test ordering.
{"title":"Real-World Biomarker Test Ordering Practices in Non-Small Cell Lung Cancer: Interphysician Variation and Association With Clinical Outcomes.","authors":"Jason M Baron, Sarrah Widatalla, Matthew A Gubens, Farah Khalil","doi":"10.1200/PO.24.00039","DOIUrl":"10.1200/PO.24.00039","url":null,"abstract":"<p><strong>Purpose: </strong>Patients with metastatic or advanced non-small cell lung cancer (NSCLC) need biomarker testing, including, in most cases, anaplastic lymphoma kinase (ALK), epidermal growth factor receptor (EGFR), and PD-L1, to identify options for targeted therapies and to optimally incorporate immune checkpoint inhibitors into therapeutic regimens. We sought to examine real-world patterns of biomarker testing, quantify interphysician practice variation, and correlate testing with clinical outcomes.</p><p><strong>Methods: </strong>We extracted real-world data from a nationwide electronic health record-derived deidentified database from 17,165 patients diagnosed with advanced NSCLC between 2018 and 2021 and receiving care in the community setting. We analyzed data using descriptive analyses, fixed- and mixed-effects logistic regression models, and proportional hazard models.</p><p><strong>Results: </strong>Only 67% of all 17,165 patients and 77% of patients with nonsquamous, metastatic NSCLC had ALK, EGFR, and PD-L1 testing within 90 days of diagnosis. Later diagnosis year (2019-2021 compared with 2018) was associated with higher rates of ALK, EGFR, and PD-L1 testing; stage IIIB/C disease (compared with stage IV), squamous histology, and Black or African American race were associated with lower rates. Interphysician variation was substantial with a median odds ratio between physicians (adjusted for patient factors) of 1.78 for ALK, EGFR, and PD-L1 testing. Patients with nonsquamous, metastatic NSCLC had significantly prolonged survival if tested with all three biomarkers (median, 364 days for all three <i>v</i> 180 for none of the three; hazard ratio, 0.67; <i>P</i> < .001).</p><p><strong>Conclusion: </strong>Rates of biomarker testing appear suboptimal with substantial interphysician variation. Testing correlates with improved survival, although causality cannot be proven from this study. Additional work is needed to address the underlying causes of suboptimal test ordering.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2400039"},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11371103/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142107587","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}
Samantha Pollard, Morgan Ehman, Anna Hermansen, Deirdre Weymann, Emanuel Krebs, Cheryl Ho, Howard J Lim, Steven Jones, Yvonne Bombard, Timothy P Hanna, Chiquita Hessels, Holly Longstaff, Robert Cook-Deegan, Tania Bubela, Dean A Regier
Purpose: In Canada, health data are siloed, slowing bioinnovation and evidence generation for personalized cancer care. Secured data-sharing platforms (SDSPs) can enable data analysis across silos through rapid concatenation across trial and real-world settings and timely researcher access. To motivate patient participation and trust in research, it is critical to ensure that SDSP design and oversight align with patients' values and address their concerns. We sought to qualitatively characterize patient preferences for the design of a pan-Canadian SDSP.
Methods: Between January 2022 and July 2023, we conducted pan-Canadian virtual focus groups with individuals who had a personal history of cancer. Following each focus group, participants were invited to provide feedback on early-phase analysis results via a member-checking survey. Three trained qualitative researchers analyzed data using thematic analysis.
Results: Twenty-eight individuals participated across five focus groups. Four focus groups were conducted in English and one in French. Thematic analysis generated two major and five minor themes. Analytic themes spanned personal and population implications of data sharing and willingness to manage perceived risks. Participants were supportive of increasing access to health data for precision oncology research, while voicing concerns about unintended data use, reidentification, and inequitable access to costly therapeutics. To mitigate perceived risks, participants highlighted the value of data access oversight and governance and informational transparency.
Conclusion: Strategies for secured data sharing should anticipate and mitigate the risks that patients perceive. Participants supported enhancing timely research capability while ensuring safeguards to protect patient autonomy and privacy. Our study informs the development of data-governance and data-sharing frameworks that integrate real-world and trial data, informed by evidence from direct patient input.
{"title":"\"I Just Assumed This Was Already Being Done\": Canadian Patient Preferences for Enhanced Data Sharing for Precision Oncology.","authors":"Samantha Pollard, Morgan Ehman, Anna Hermansen, Deirdre Weymann, Emanuel Krebs, Cheryl Ho, Howard J Lim, Steven Jones, Yvonne Bombard, Timothy P Hanna, Chiquita Hessels, Holly Longstaff, Robert Cook-Deegan, Tania Bubela, Dean A Regier","doi":"10.1200/PO.24.00184","DOIUrl":"10.1200/PO.24.00184","url":null,"abstract":"<p><strong>Purpose: </strong>In Canada, health data are siloed, slowing bioinnovation and evidence generation for personalized cancer care. Secured data-sharing platforms (SDSPs) can enable data analysis across silos through rapid concatenation across trial and real-world settings and timely researcher access. To motivate patient participation and trust in research, it is critical to ensure that SDSP design and oversight align with patients' values and address their concerns. We sought to qualitatively characterize patient preferences for the design of a pan-Canadian SDSP.</p><p><strong>Methods: </strong>Between January 2022 and July 2023, we conducted pan-Canadian virtual focus groups with individuals who had a personal history of cancer. Following each focus group, participants were invited to provide feedback on early-phase analysis results via a member-checking survey. Three trained qualitative researchers analyzed data using thematic analysis.</p><p><strong>Results: </strong>Twenty-eight individuals participated across five focus groups. Four focus groups were conducted in English and one in French. Thematic analysis generated two major and five minor themes. Analytic themes spanned personal and population implications of data sharing and willingness to manage perceived risks. Participants were supportive of increasing access to health data for precision oncology research, while voicing concerns about unintended data use, reidentification, and inequitable access to costly therapeutics. To mitigate perceived risks, participants highlighted the value of data access oversight and governance and informational transparency.</p><p><strong>Conclusion: </strong>Strategies for secured data sharing should anticipate and mitigate the risks that patients perceive. Participants supported enhancing timely research capability while ensuring safeguards to protect patient autonomy and privacy. Our study informs the development of data-governance and data-sharing frameworks that integrate real-world and trial data, informed by evidence from direct patient input.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2400184"},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11371116/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906689","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}
Laura S Graham, Nicholas C Henderson, Olesia Kellezi, Clara Hwang, Pedro C Barata, Mehmet A Bilen, Deepak Kilari, Michael Pierro, Bicky Thapa, Abhishek Tripathi, George Mo, Matthew Labriola, Joseph J Park, Shoshana Rothstein, Rohan Garje, Vadim S Koshkin, Vaibhav G Patel, Tanya Dorff, Andrew J Armstrong, Rana R McKay, Ajjai Alva, Michael T Schweizer
Purpose: Outcomes data for DNA-damaging therapeutics for men with prostate cancer (PC) and non-BRCA1/2 homologous recombination repair (HRR) mutations are limited. We evaluated outcomes by HRR alteration in men with PC treated with poly(ADP-ribose)polymerase inhibitors (PARPi) and/or platinum chemotherapy.
Methods: Retrospective data from the PROMISE consortium were used. Clinical outcomes differences were assessed between patients with BRCA1/2 mutations (cohort A) and those with HRR mutations without direct BRCA complex interaction (cohort B: ATM, CDK12, CHEK1, CHEK2, and FANCL). Outcomes in patients with HRR mutations with direct BRCA complex interaction were also explored (cohort C: RAD51B/C/D, RAD54L2, BARD1, GEN1, PALB2, FANCA, and BRIP1).
Results: One hundred and forty-six patients received PARPi (cohort A: 94, cohort B: 45, cohort C: 7) and 104 received platinum chemotherapy (cohort A: 48, cohort B: 44, cohort C: 10). PSA50 response rate to PARPi was higher in cohort A (61%) than cohort B (5%), P < .001. Median clinical/radiographic progression-free survival (crPFS) with PARPi in cohort A was significantly longer than in cohort B: 15.9 versus 8.7 months, P = .005. PSA50 response rate to platinum therapy was higher in cohort A (62%) than in cohort B (32%), P = .024, although crPFS was not significantly different. PSA50 response rate to PARPi and platinum was 40% and 32%, respectively, in cohort C. In multivariable analysis, cohort A had significantly improved overall survival and crPFS compared with cohort B with PARPi but not platinum chemotherapy.
Conclusion: Patients with BRCA1/2-mutated PC had significantly improved outcomes to PARPi but not platinum chemotherapy compared with those with HRR mutations without direct BRCA complex interaction.
{"title":"DNA-Damaging Therapies in Patients With Prostate Cancer and Pathogenic Alterations in Homologous Recombination Repair Genes.","authors":"Laura S Graham, Nicholas C Henderson, Olesia Kellezi, Clara Hwang, Pedro C Barata, Mehmet A Bilen, Deepak Kilari, Michael Pierro, Bicky Thapa, Abhishek Tripathi, George Mo, Matthew Labriola, Joseph J Park, Shoshana Rothstein, Rohan Garje, Vadim S Koshkin, Vaibhav G Patel, Tanya Dorff, Andrew J Armstrong, Rana R McKay, Ajjai Alva, Michael T Schweizer","doi":"10.1200/PO.24.00014","DOIUrl":"10.1200/PO.24.00014","url":null,"abstract":"<p><strong>Purpose: </strong>Outcomes data for DNA-damaging therapeutics for men with prostate cancer (PC) and non-<i>BRCA1/2</i> homologous recombination repair (HRR) mutations are limited. We evaluated outcomes by HRR alteration in men with PC treated with poly(ADP-ribose)polymerase inhibitors (PARPi) and/or platinum chemotherapy.</p><p><strong>Methods: </strong>Retrospective data from the PROMISE consortium were used. Clinical outcomes differences were assessed between patients with <i>BRCA1</i>/<i>2</i> mutations (cohort A) and those with HRR mutations without direct BRCA complex interaction (cohort B: <i>ATM</i>, <i>CDK12</i>, <i>CHEK1</i>, <i>CHEK2</i>, and <i>FANCL</i>). Outcomes in patients with HRR mutations with direct BRCA complex interaction were also explored (cohort C: <i>RAD51B/C/D</i>, <i>RAD54L2</i>, <i>BARD1</i>, <i>GEN1</i>, <i>PALB2</i>, <i>FANCA</i>, and <i>BRIP1</i>).</p><p><strong>Results: </strong>One hundred and forty-six patients received PARPi (cohort A: 94, cohort B: 45, cohort C: 7) and 104 received platinum chemotherapy (cohort A: 48, cohort B: 44, cohort C: 10). PSA50 response rate to PARPi was higher in cohort A (61%) than cohort B (5%), <i>P</i> < .001. Median clinical/radiographic progression-free survival (crPFS) with PARPi in cohort A was significantly longer than in cohort B: 15.9 versus 8.7 months, <i>P</i> = .005. PSA50 response rate to platinum therapy was higher in cohort A (62%) than in cohort B (32%), <i>P</i> = .024, although crPFS was not significantly different. PSA50 response rate to PARPi and platinum was 40% and 32%, respectively, in cohort C. In multivariable analysis, cohort A had significantly improved overall survival and crPFS compared with cohort B with PARPi but not platinum chemotherapy.</p><p><strong>Conclusion: </strong>Patients with <i>BRCA1/2</i>-mutated PC had significantly improved outcomes to PARPi but not platinum chemotherapy compared with those with HRR mutations without direct BRCA complex interaction.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2400014"},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11346579/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142043913","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":"Erratum: Unexpected Durable Complete Response With Anti-PD-L1 Blockade in Metastatic Undifferentiated Pleomorphic Sarcoma: A Case Report With Host and Tumor Biomarker Analysis.","authors":"","doi":"10.1200/PO-24-00427","DOIUrl":"https://doi.org/10.1200/PO-24-00427","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2400427"},"PeriodicalIF":5.3,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141748253","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}
Nicholas P Giustini, Colin C Pritchard, Nikhil V Kamat, Manoj P Menon
Up-front osimertinib leads to clinical benefit in EGFR V834L and L858R comutated NSCLC.
奥希替尼可使表皮生长因子受体 V834L 和 L858R 组合型 NSCLC 患者获得临床获益。
{"title":"<i>EGFR</i> V834L and L858R Comutation Is Associated With Response to Osimertinib in Non-Small-Cell Lung Cancer.","authors":"Nicholas P Giustini, Colin C Pritchard, Nikhil V Kamat, Manoj P Menon","doi":"10.1200/PO.23.00215","DOIUrl":"10.1200/PO.23.00215","url":null,"abstract":"<p><p>Up-front osimertinib leads to clinical benefit in EGFR V834L and L858R comutated NSCLC.</p>","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2300215"},"PeriodicalIF":5.3,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141590327","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":"Deciphering Disease Profiles in the Era of Prostate-Specific Membrane Antigen Positron Emission Tomography: Aggressive or Indolent?","authors":"Peter D Zang, Salvador Jaime-Casas, Wesley Yip","doi":"10.1200/PO-24-00377","DOIUrl":"10.1200/PO-24-00377","url":null,"abstract":"","PeriodicalId":14797,"journal":{"name":"JCO precision oncology","volume":"8 ","pages":"e2400377"},"PeriodicalIF":5.3,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141626805","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}