首页 > 最新文献

JAMA Oncology最新文献

英文 中文
Gastroenteropancreatic Neuroendocrine Tumor Incidence by Sex and Age in the US 美国按性别和年龄划分的胃肠胰神经内分泌肿瘤发病率
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-16 DOI: 10.1001/jamaoncol.2024.5937
Yazan Abboud, Anand Shah, Riya Sutariya, Vraj P. Shah, Ahmed Al-Khazraji, Paul J. Gaglio, Kaveh Hajifathalian
This observational study reports on a comprehensive nationwide evaluation of rising gastroenteropancreatic neuroendocrine tumor incidence in the US from 2001 to 2020.
本观察性研究报告了2001年至2020年美国胃肠胰腺神经内分泌肿瘤发病率上升的综合全国评估。
{"title":"Gastroenteropancreatic Neuroendocrine Tumor Incidence by Sex and Age in the US","authors":"Yazan Abboud, Anand Shah, Riya Sutariya, Vraj P. Shah, Ahmed Al-Khazraji, Paul J. Gaglio, Kaveh Hajifathalian","doi":"10.1001/jamaoncol.2024.5937","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.5937","url":null,"abstract":"This observational study reports on a comprehensive nationwide evaluation of rising gastroenteropancreatic neuroendocrine tumor incidence in the US from 2001 to 2020.","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"68 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142986885","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Intratumoral Injection of mRNA-2752 and Pembrolizumab for High-Risk Ductal Carcinoma In Situ 肿瘤内注射mRNA-2752和派姆单抗治疗高危导管原位癌
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-16 DOI: 10.1001/jamaoncol.2024.5927
Kirithiga Ramalingam, Rachel Woody, Alexa Glencer, Christopher J. Schwartz, Hidetoshi Mori, Jasmine Wong, Gillian Hirst, Jennifer Rosenbluth, Natsuko Onishi, Jessica Gibbs, Nola Hylton, Alexander D. Borowsky, Michael Campbell, Laura J. Esserman
ImportanceIntratumoral immunotherapy that leverages the biological characteristics of high-risk ductal carcinoma in situ (DCIS) may be able to reduce the extent of surgical treatment and provide an alternative approach to improve patient outcomes.ObjectiveTo determine if combination intratumoral immunotherapy can activate immune cells to shrink or eliminate high-risk DCIS.Design, Setting, and ParticipantsThis phase 1 open-label nonrandomized clinical trial at a single academic center tested the safety and efficacy of intratumoral immunotherapy in patients with high-risk DCIS, defined as at least 2 of the following present: younger than 45 years, tumor size greater than 5 cm, high-grade, palpable mass, hormone receptor (HR)–negative, or ERBB2-positive. Patients were enrolled between June 8, 2021, and December 13, 2022.InterventionPembrolizumab (anti–programmed cell death protein 1), dose ranging from 2 mg to 8 mg, and mRNA-2752 (a combination of interleukin [IL]-23, IL-36γ, and OX40L mRNAs), dose ranging from 1 mg to 4 mg, delivered intratumorally, with 2 to 4 doses given 2 to 3 weeks apart.Main Outcomes and MeasuresThe primary objective was to evaluate the safety and tolerability of intratumoral injections of pembrolizumab and mRNA-2752. The secondary objectives were to assess radiologic and pathological responses and immunological and histological differences in the posttreatment tumor microenvironment.ResultsTen female patients with high-risk DCIS (median [range] age, 46 [35-80] years) were enrolled. The median (range) tumor size was 5.3 (1.0-10.0) cm. Five tumors were HR-negative ERBB2-positive; 2 HR-negative ERBB2-negative; 2 HR-positive ERBB2-negative; and 1 HR-positive ERBB2-positive. Of all treated patients, 8 of 10 responded to treatment, and all 8 patients had ERBB2-positive or HR-negative DCIS. Three patients had complete responses. Three patients with negative posttreatment core biopsy results declined surgery and remained disease-free after 1 to 2 years. Multiplex immunofluorescence staining demonstrated that high baseline levels of tumor-infiltrating lymphocytes and programmed cell death ligand 1–positive cells (immune or tumor) were associated with a better treatment response. All patients experienced up to 1 week of fever, malaise, flulike symptoms, axillary adenopathy, erythema, injection site swelling, and swelling in the breast. One patient had intermittent urticaria for 3 months. The dose was serially reduced from 8 mg to 2 mg for pembrolizumab and 4 mg to 1 mg for mRNA-2752 to improve tolerability. The final recommended combination dose is pembrolizumab, 4 mg, with mRNA-2752, 1 mg.Conclusions and RelevanceIn this phase 1 nonrandomized clinical trial, the results suggest that intratumoral injections of pembrolizumab and mRNA-2752 are safe and may induce rapid regression of high-risk DCIS with high immune infiltrates. These findings warrant additional investigation, and studies are ongoing.Trial RegistrationClinicalTrials.gov Id
重要性利用高危导管原位癌(DCIS)生物学特性的瘤内免疫治疗可能能够减少手术治疗的范围,并提供一种改善患者预后的替代方法。目的探讨肿瘤内联合免疫治疗是否能激活免疫细胞缩小或消除高危DCIS。设计、环境和参与者:在一个学术中心进行的1期开放标签非随机临床试验测试了肿瘤内免疫治疗对高危DCIS患者的安全性和有效性,定义为以下至少2项:小于45岁、肿瘤大小大于5cm、高级别、可触及肿块、激素受体(HR)阴性或erbb2阳性。患者在2021年6月8日至2022年12月13日期间入组。干预:pembrolizumab(抗程序性细胞死亡蛋白1),剂量范围为2mg至8mg, mRNA-2752(白介素[IL]-23, IL-36γ和OX40L mrna的组合),剂量范围为1mg至4mg,瘤内给药,间隔2至3周给予2至4次剂量。主要结局和措施主要目的是评估肿瘤内注射派姆单抗和mRNA-2752的安全性和耐受性。次要目的是评估治疗后肿瘤微环境的放射学和病理反应以及免疫学和组织学差异。结果入选10例高危DCIS女性患者,中位年龄46岁[35-80]岁。中位(范围)肿瘤大小为5.3 (1.0-10.0)cm。5例hr阴性,erbb2阳性;2例hr阴性erbb2阴性;2例hr阳性erbb2阴性;1例hr阳性,erbb2阳性。在所有接受治疗的患者中,10名患者中有8名对治疗有反应,所有8名患者都有erbb2阳性或hr阴性的DCIS。3例患者完全缓解。3例治疗后核心活检结果阴性的患者拒绝手术,并在1至2年后保持无病状态。多重免疫荧光染色显示,高基线水平的肿瘤浸润淋巴细胞和程序性细胞死亡配体1阳性细胞(免疫或肿瘤)与更好的治疗反应相关。所有患者都经历了长达1周的发热、不适、流感样症状、腋窝腺病、红斑、注射部位肿胀和乳房肿胀。1例患者间歇性荨麻疹3个月。为了提高耐受性,派姆单抗的剂量从8mg降至2mg, mRNA-2752的剂量从4mg降至1mg。最终推荐的联合剂量是派姆单抗4mg和mRNA-2752 1mg。结论和相关性在这项1期非随机临床试验中,结果表明瘤内注射派姆单抗和mRNA-2752是安全的,并且可以诱导高免疫浸润的高危DCIS快速消退。这些发现值得进一步调查,研究仍在进行中。临床试验注册号:NCT02872025
{"title":"Intratumoral Injection of mRNA-2752 and Pembrolizumab for High-Risk Ductal Carcinoma In Situ","authors":"Kirithiga Ramalingam, Rachel Woody, Alexa Glencer, Christopher J. Schwartz, Hidetoshi Mori, Jasmine Wong, Gillian Hirst, Jennifer Rosenbluth, Natsuko Onishi, Jessica Gibbs, Nola Hylton, Alexander D. Borowsky, Michael Campbell, Laura J. Esserman","doi":"10.1001/jamaoncol.2024.5927","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.5927","url":null,"abstract":"ImportanceIntratumoral immunotherapy that leverages the biological characteristics of high-risk ductal carcinoma in situ (DCIS) may be able to reduce the extent of surgical treatment and provide an alternative approach to improve patient outcomes.ObjectiveTo determine if combination intratumoral immunotherapy can activate immune cells to shrink or eliminate high-risk DCIS.Design, Setting, and ParticipantsThis phase 1 open-label nonrandomized clinical trial at a single academic center tested the safety and efficacy of intratumoral immunotherapy in patients with high-risk DCIS, defined as at least 2 of the following present: younger than 45 years, tumor size greater than 5 cm, high-grade, palpable mass, hormone receptor (HR)–negative, or ERBB2-positive. Patients were enrolled between June 8, 2021, and December 13, 2022.InterventionPembrolizumab (anti–programmed cell death protein 1), dose ranging from 2 mg to 8 mg, and mRNA-2752 (a combination of interleukin [IL]-23, IL-36γ, and OX40L mRNAs), dose ranging from 1 mg to 4 mg, delivered intratumorally, with 2 to 4 doses given 2 to 3 weeks apart.Main Outcomes and MeasuresThe primary objective was to evaluate the safety and tolerability of intratumoral injections of pembrolizumab and mRNA-2752. The secondary objectives were to assess radiologic and pathological responses and immunological and histological differences in the posttreatment tumor microenvironment.ResultsTen female patients with high-risk DCIS (median [range] age, 46 [35-80] years) were enrolled. The median (range) tumor size was 5.3 (1.0-10.0) cm. Five tumors were HR-negative ERBB2-positive; 2 HR-negative ERBB2-negative; 2 HR-positive ERBB2-negative; and 1 HR-positive ERBB2-positive. Of all treated patients, 8 of 10 responded to treatment, and all 8 patients had ERBB2-positive or HR-negative DCIS. Three patients had complete responses. Three patients with negative posttreatment core biopsy results declined surgery and remained disease-free after 1 to 2 years. Multiplex immunofluorescence staining demonstrated that high baseline levels of tumor-infiltrating lymphocytes and programmed cell death ligand 1–positive cells (immune or tumor) were associated with a better treatment response. All patients experienced up to 1 week of fever, malaise, flulike symptoms, axillary adenopathy, erythema, injection site swelling, and swelling in the breast. One patient had intermittent urticaria for 3 months. The dose was serially reduced from 8 mg to 2 mg for pembrolizumab and 4 mg to 1 mg for mRNA-2752 to improve tolerability. The final recommended combination dose is pembrolizumab, 4 mg, with mRNA-2752, 1 mg.Conclusions and RelevanceIn this phase 1 nonrandomized clinical trial, the results suggest that intratumoral injections of pembrolizumab and mRNA-2752 are safe and may induce rapid regression of high-risk DCIS with high immune infiltrates. These findings warrant additional investigation, and studies are ongoing.Trial RegistrationClinicalTrials.gov Id","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"245 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142986884","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Biomarker-Directed Radiotherapy in Breast Cancer: A Narrative Review. 生物标志物定向放疗在乳腺癌中的应用综述
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-16 DOI: 10.1001/jamaoncol.2024.5780
Icro Meattini,Charlotte E Coles,Trine Tramm,Simona Borghesi,David Krug,Angel Montero,Valerio Nardone,Viola Salvestrini,Marianna Valzano,Vincenzo Valentini,Cynthia Aristei,Philip Poortmans,
ImportanceIntegration of molecular biomarker information into systemic therapy has become standard practice in breast cancer care. However, its implementation in guiding radiotherapy (RT) is slower. Although postoperative RT is recommended for most patients after breast-conserving surgery and, depending on risk factors, following mastectomy, emerging evidence has indicated that patients with low scores on gene expression signatures or selected clinical-pathological features may have very low local recurrence rates. This narrative review explored the potential of biomarker-directed personalized RT approaches, which may optimize treatment strategies and be associated with improved patient outcomes and experiences.ObservationsDistinctions between prognostic and predictive biomarkers were highlighted, emphasizing the importance of analytical and clinical validity in biomarker-based studies. Findings from studies investigating the prognostic and predictive value of various genomic signatures and immunohistochemical markers for guiding breast RT were presented. These included the Adjuvant Radiotherapy Intensification Classifier and the Profile for the Omission of Local Adjuvant Radiation, which have shown potential in predicting RT benefits. The genomic-adjusted radiation dose and role of tumor-infiltrating lymphocytes were also discussed. Ongoing clinical trials exploring the use of biomarkers in ductal carcinoma in situ and invasive breast cancer to refine RT decision-making were illustrated.Conclusions and RelevanceThe results of this narrative review suggest that evidence-based shared decision-making is crucial to optimize treatment according to the individual's predicted benefits and risks along with their personal preferences. Incorporation of biomarker-directed approaches in RT for breast cancer may hold promise for personalized treatment, potentially facilitating omission of RT for patients at low risk of recurrence, while identifying those who may benefit from intensified therapy. This personalized RT approach may be associated with improved clinical outcomes and quality of life and facilitate decision-making for people with breast cancer. However, there remains a need for robust clinical and analytical validation of biomarkers to ensure reliability and clinical utility for RT optimization.
重要性将分子生物标记信息纳入全身治疗已成为乳腺癌治疗的标准做法。然而,其在指导放射治疗(RT)中的应用却较为缓慢。虽然大多数患者在接受保乳手术后都建议进行术后放疗,而且根据风险因素,乳房切除术后也建议进行术后放疗,但新出现的证据表明,基因表达特征或选定临床病理特征得分较低的患者的局部复发率可能很低。本综述探讨了以生物标志物为导向的个性化 RT 方法的潜力,这种方法可能会优化治疗策略并改善患者的预后和体验。会上介绍了对指导乳腺 RT 的各种基因组特征和免疫组化标记物的预后和预测价值的研究结果。其中包括 "辅助放疗强化分类器"(Adjuvant Radiotherapy Intensification Classifier)和 "局部辅助放疗遗漏概况"(Profile for the Omission of Local Adjuvant Radiation),它们在预测乳腺放射治疗的获益方面已显示出潜力。会上还讨论了基因组调整放射剂量和肿瘤浸润淋巴细胞的作用。本综述的结果表明,以证据为基础的共同决策对于根据个人预测的获益和风险以及个人偏好优化治疗至关重要。在乳腺癌的 RT 治疗中纳入生物标志物导向方法可能会为个性化治疗带来希望,有可能帮助低复发风险患者省去 RT 治疗,同时识别那些可能从强化治疗中获益的患者。这种个性化 RT 方法可能会改善临床疗效和生活质量,并有助于乳腺癌患者做出决策。然而,生物标记物仍需经过严格的临床和分析验证,以确保其在 RT 优化方面的可靠性和临床实用性。
{"title":"Biomarker-Directed Radiotherapy in Breast Cancer: A Narrative Review.","authors":"Icro Meattini,Charlotte E Coles,Trine Tramm,Simona Borghesi,David Krug,Angel Montero,Valerio Nardone,Viola Salvestrini,Marianna Valzano,Vincenzo Valentini,Cynthia Aristei,Philip Poortmans,","doi":"10.1001/jamaoncol.2024.5780","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.5780","url":null,"abstract":"ImportanceIntegration of molecular biomarker information into systemic therapy has become standard practice in breast cancer care. However, its implementation in guiding radiotherapy (RT) is slower. Although postoperative RT is recommended for most patients after breast-conserving surgery and, depending on risk factors, following mastectomy, emerging evidence has indicated that patients with low scores on gene expression signatures or selected clinical-pathological features may have very low local recurrence rates. This narrative review explored the potential of biomarker-directed personalized RT approaches, which may optimize treatment strategies and be associated with improved patient outcomes and experiences.ObservationsDistinctions between prognostic and predictive biomarkers were highlighted, emphasizing the importance of analytical and clinical validity in biomarker-based studies. Findings from studies investigating the prognostic and predictive value of various genomic signatures and immunohistochemical markers for guiding breast RT were presented. These included the Adjuvant Radiotherapy Intensification Classifier and the Profile for the Omission of Local Adjuvant Radiation, which have shown potential in predicting RT benefits. The genomic-adjusted radiation dose and role of tumor-infiltrating lymphocytes were also discussed. Ongoing clinical trials exploring the use of biomarkers in ductal carcinoma in situ and invasive breast cancer to refine RT decision-making were illustrated.Conclusions and RelevanceThe results of this narrative review suggest that evidence-based shared decision-making is crucial to optimize treatment according to the individual's predicted benefits and risks along with their personal preferences. Incorporation of biomarker-directed approaches in RT for breast cancer may hold promise for personalized treatment, potentially facilitating omission of RT for patients at low risk of recurrence, while identifying those who may benefit from intensified therapy. This personalized RT approach may be associated with improved clinical outcomes and quality of life and facilitate decision-making for people with breast cancer. However, there remains a need for robust clinical and analytical validation of biomarkers to ensure reliability and clinical utility for RT optimization.","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"45 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142988591","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Overcoming Barriers to Make Patient-Reported Outcome Collection the Standard of Care in Oncology. 克服障碍,使患者报告结果收集成为肿瘤学的标准护理。
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-16 DOI: 10.1001/jamaoncol.2024.6157
James A Colbert,Louis Potters
{"title":"Overcoming Barriers to Make Patient-Reported Outcome Collection the Standard of Care in Oncology.","authors":"James A Colbert,Louis Potters","doi":"10.1001/jamaoncol.2024.6157","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.6157","url":null,"abstract":"","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"66 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142988523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Genetic Ancestry–Based Differences in Biomarker-Based Eligibility for Precision Oncology Therapies 基于生物标志物的精准肿瘤治疗资格的遗传血统差异
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-09 DOI: 10.1001/jamaoncol.2024.5794
Kanika Arora, Sarah P. Suehnholz, Hongxin Zhang, Irina Ostrovnaya, Ritika Kundra, Subhiksha Nandakumar, Moriah H. Nissan, A. Rose Brannon, Chaitanya Bandlamudi, Marc Ladanyi, Alexander Drilon, Carol L. Brown, David B. Solit, Nikolaus Schultz, Michael F. Berger, Debyani Chakravarty
ImportanceAlthough differences in the prevalence of key cancer-specific somatic mutations as a function of genetic ancestry among patients with cancer has been well-established, few studies have addressed the practical clinical implications of these differences for the growing number of biomarker-driven treatments.ObjectiveTo determine if the approval of precision oncology therapies has benefited patients with cancer from various ancestral backgrounds equally over time.Design, Setting, and ParticipantsA retrospective analysis of samples from patients with solid cancers who underwent clinical sequencing using the integrated mutation profiling of actionable cancer targets (MSK-IMPACT) assay between January 2014 and December 2022 was carried out. The annual fraction of patients per ancestral group with at least 1 level 1 biomarker was calculated for FDA drug approvals from January 1998 to December 2023. Analysis began in January 2024.Main Outcomes and MeasuresFor each patient, genetic ancestry was quantitatively inferred, and patients were grouped based on predominant reference ancestry. OncoKB was used to identify all Food and Drug Administration (FDA)–recognized somatic biomarkers associated with FDA-approved therapies (level 1 biomarkers) in each tumor sample.ResultsOverall, the study included 59 433 patients. The approval of the EGFR-tyrosine kinase inhibitor erlotinib for patients with EGFR-mutant lung cancers in 2013 disproportionately benefited patients of East Asian and South Asian ancestries, leading to higher patient fractions with level 1 biomarkers in these ancestral groups compared with other populations. Although the increase in precision oncology drug approvals from 2019 to 2020 had a notable positive impact on clinical actionability for patients of European ancestry, patients of African ancestry had the lowest fraction of level 1 biomarkers compared with other groups from 2019 onward.Conclusion and RelevanceThis study systematically assessed and compared temporal changes in genomic biomarker-based eligibility for precision oncology therapies as a function of inferred genetic ancestry derived from DNA sequencing data. Despite the accelerated rate of FDA approvals for precision oncology therapies over the past decade, measurable differences in biomarker-based drug eligibility among patient ancestral groups exist. These differences may exacerbate the systemic disparities in clinical outcomes in patients of African ancestry due to existing deficiencies in their access to cancer care.
尽管癌症患者中关键癌症特异性体细胞突变的患病率差异作为遗传祖先的功能已经得到证实,但很少有研究解决这些差异对越来越多的生物标志物驱动治疗的实际临床意义。目的确定精确肿瘤疗法的批准是否随着时间的推移使不同祖先背景的癌症患者同样受益。设计、环境和参与者对2014年1月至2022年12月期间使用可操作癌症靶点综合突变谱(MSK-IMPACT)测定法进行临床测序的实体癌患者样本进行回顾性分析。计算1998年1月至2023年12月FDA药物批准的每个祖先组中至少有1个1级生物标志物的患者的年比例。分析开始于2024年1月。对于每位患者,定量推断遗传血统,并根据主要参考血统对患者进行分组。OncoKB用于识别每个肿瘤样本中所有FDA认可的与FDA批准的治疗相关的体细胞生物标志物(1级生物标志物)。结果共纳入59 433例患者。2013年批准用于egfr突变型肺癌患者的egfr -酪氨酸激酶抑制剂厄洛替尼(erlotinib)对东亚和南亚血统的患者格外有利,与其他人群相比,这些祖先群体中具有1级生物标志物的患者比例更高。尽管从2019年到2020年,精准肿瘤药物批准的增加对欧洲血统患者的临床可操作性产生了显著的积极影响,但从2019年起,与其他群体相比,非洲血统患者的1级生物标志物比例最低。结论和相关性本研究系统地评估和比较了基于基因组生物标志物的精确肿瘤治疗资格的时间变化,作为从DNA测序数据推断的遗传祖先的功能。尽管在过去十年中,FDA对精确肿瘤治疗的批准速度加快,但在患者祖先群体中,基于生物标志物的药物资格存在可测量的差异。这些差异可能会加剧非洲裔患者临床结果的系统性差异,因为他们在获得癌症治疗方面存在缺陷。
{"title":"Genetic Ancestry–Based Differences in Biomarker-Based Eligibility for Precision Oncology Therapies","authors":"Kanika Arora, Sarah P. Suehnholz, Hongxin Zhang, Irina Ostrovnaya, Ritika Kundra, Subhiksha Nandakumar, Moriah H. Nissan, A. Rose Brannon, Chaitanya Bandlamudi, Marc Ladanyi, Alexander Drilon, Carol L. Brown, David B. Solit, Nikolaus Schultz, Michael F. Berger, Debyani Chakravarty","doi":"10.1001/jamaoncol.2024.5794","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.5794","url":null,"abstract":"ImportanceAlthough differences in the prevalence of key cancer-specific somatic mutations as a function of genetic ancestry among patients with cancer has been well-established, few studies have addressed the practical clinical implications of these differences for the growing number of biomarker-driven treatments.ObjectiveTo determine if the approval of precision oncology therapies has benefited patients with cancer from various ancestral backgrounds equally over time.Design, Setting, and ParticipantsA retrospective analysis of samples from patients with solid cancers who underwent clinical sequencing using the integrated mutation profiling of actionable cancer targets (MSK-IMPACT) assay between January 2014 and December 2022 was carried out. The annual fraction of patients per ancestral group with at least 1 level 1 biomarker was calculated for FDA drug approvals from January 1998 to December 2023. Analysis began in January 2024.Main Outcomes and MeasuresFor each patient, genetic ancestry was quantitatively inferred, and patients were grouped based on predominant reference ancestry. OncoKB was used to identify all Food and Drug Administration (FDA)–recognized somatic biomarkers associated with FDA-approved therapies (level 1 biomarkers) in each tumor sample.ResultsOverall, the study included 59 433 patients. The approval of the <jats:italic>EGFR</jats:italic>-tyrosine kinase inhibitor erlotinib for patients with <jats:italic>EGFR</jats:italic>-mutant lung cancers in 2013 disproportionately benefited patients of East Asian and South Asian ancestries, leading to higher patient fractions with level 1 biomarkers in these ancestral groups compared with other populations. Although the increase in precision oncology drug approvals from 2019 to 2020 had a notable positive impact on clinical actionability for patients of European ancestry, patients of African ancestry had the lowest fraction of level 1 biomarkers compared with other groups from 2019 onward.Conclusion and RelevanceThis study systematically assessed and compared temporal changes in genomic biomarker-based eligibility for precision oncology therapies as a function of inferred genetic ancestry derived from DNA sequencing data. Despite the accelerated rate of FDA approvals for precision oncology therapies over the past decade, measurable differences in biomarker-based drug eligibility among patient ancestral groups exist. These differences may exacerbate the systemic disparities in clinical outcomes in patients of African ancestry due to existing deficiencies in their access to cancer care.","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"2 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142939823","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
What We Can Learn About Patient Safety While Driving to Work 开车上班时我们能学到的关于病人安全的知识
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-09 DOI: 10.1001/jamaoncol.2024.6151
Lawrence B. Marks, Caprice C. Greenberg, Lukasz M. Mazur
This Viewpoint discusses strategies used with driving that can be applied to health care to promote consistent and predictable physician and patient actions.
本观点讨论了可应用于卫生保健的与驾驶一起使用的策略,以促进一致和可预测的医生和患者行动。
{"title":"What We Can Learn About Patient Safety While Driving to Work","authors":"Lawrence B. Marks, Caprice C. Greenberg, Lukasz M. Mazur","doi":"10.1001/jamaoncol.2024.6151","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.6151","url":null,"abstract":"This Viewpoint discusses strategies used with driving that can be applied to health care to promote consistent and predictable physician and patient actions.","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"20 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142939874","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Shared Local Oncology Care After Allogeneic Hematopoietic Cell Transplantation 同种异体造血细胞移植术后的局部肿瘤护理
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-09 DOI: 10.1001/jamaoncol.2024.5786
Gregory A. Abel, Haesook T. Kim, Ira Zackon, Edwin T. Alyea, Alexandra S. Bailey, John P. Winters, Kenneth R. Meehan, John L. Reagan, Jeanna H. Walsh, Thomas P. Walsh, Alexandra Ivanov, Meredith A. Faggen, Sarah Sinclair, Amy C. Joyce, Sara D. Close, Amy Emmert, Jon Koreth, Joseph H. Antin, Corey S. Cutler, Vincent T. Ho, Robert J. Soiffer
ImportanceAlthough sharing care with local oncologists after allogeneic hematopoietic cell transplantation (HCT) has been proposed for patients living far from HCT centers, it is not known whether a shared strategy is safe or improves patient quality of life (QOL).ObjectiveTo determine the efficacy and safety of sharing follow-up care after HCT between the HCT specialty center and local oncologists.Design, Setting, and ParticipantsThis was a multicenter collaborative randomized clinical trial of patients undergoing HCT at Dana-Farber Cancer Institute (DFCI)—a high volume HCT center in Boston (Massachusetts)—and 8 local oncology practices. Eligible patients were enrolled from December 2017 to December 2021 and were randomized 1:1 to shared vs usual care after neutrophil engraftment, stratified by local sites in Massachusetts, Rhode Island, New Hampshire, New York, and Maine. Data analyses were performed in January 2024.InterventionShared care involved alternating post-HCT visits at DFCI and local oncology practices through day 100; for usual care, all post-HCT visits occurred only at DFCI.Main Outcomes and MeasuresCoprimary outcomes were nonrelapse mortality (NRM) at day 100, and QOL measured by the FACT-BMT (Functional Assessment of Cancer Therapy–Bone Marrow Transplantation) instrument and the QLQ-C30 (European Organization for Research and Treatment of Cancer’s Quality of Life Questionnaire) at day 180. Prespecified secondary outcomes included day 100 QOL and 1-year overall survival.ResultsA total of 302 participants (median [range] age, 63 [20-79] years; 117 [38.7%] females; 185 [61.3%] males) were included in the analysis; 152 were randomized to shared care and 150 to usual care. Day 100 NRM was noninferior for shared vs usual care (2.6% [95% CI, 0.7% to 6.6%] vs 2.7% [95% CI, 0.7% to 6.7%]; P = .98). There were no differences at day 180 for the FACT-BMT total score (mean difference, 3.8; 95% CI, −2.1 to 9.6; P = .20) or QLQ-C30 global score (1.9; 95% CI, −4.9 to 8.8; P = .58). At day 100, the FACT-BMT total score was better for shared care (mean difference, 6.6; 95% CI, 1.0 to 12.1; P = .02) as was the QLQ-C30 global score (8.8; 95% CI, 1.8 to 15.7; P = .02).Conclusions and RelevanceThis randomized clinical trial found that shared care resulted in noninferior NRM at day 100 but similar QOL at day 180, with improved QOL at day 100. These data suggest that shared care is safe, improves QOL early on, and has the potential to become a routine model for post-HCT care.Trial RegistrationClinicalTrials.gov Identifier: NCT03244826
尽管在异基因造血细胞移植(HCT)后与当地肿瘤学家共享护理已被提议用于远离HCT中心的患者,但尚不清楚共享策略是否安全或是否能提高患者的生活质量(QOL)。目的探讨HCT专科中心与当地肿瘤医师共享HCT术后随访护理的有效性和安全性。设计、环境和参与者这是一项多中心协作随机临床试验,在丹娜-法伯癌症研究所(DFCI) -波士顿(马萨诸塞州)的高容量HCT中心-和8个当地肿瘤诊所接受HCT的患者。符合条件的患者于2017年12月至2021年12月入组,中性粒细胞植入后按1:1随机分为共享治疗和常规治疗,按马萨诸塞州、罗德岛州、新罕布什尔州、纽约州和缅因州的当地地点分层。数据分析于2024年1月进行。干预:共享护理包括在第100天在DFCI和当地肿瘤诊所交替进行hct后访问;对于常规护理,所有hct后访问仅发生在DFCI。主要结果和测量主要结果为第100天的非复发死亡率(NRM),以及第180天用FACT-BMT(肿瘤治疗-骨髓移植功能评估)仪器和QLQ-C30(欧洲癌症研究和治疗组织生活质量问卷)测量的生活质量。预先指定的次要结局包括100天的生活质量和1年的总生存期。结果共302例受试者(年龄中位数[范围]为63[20-79]岁;女性117例(38.7%);185例(61.3%)男性纳入分析;152例随机分配到共同护理组,150例随机分配到常规护理组。第100天的NRM与常规护理相比并不逊色(2.6% [95% CI, 0.7%至6.6%]vs 2.7% [95% CI, 0.7%至6.7%];P = .98)。在第180天,FACT-BMT总分无差异(平均差异3.8;95% CI,−2.1 ~ 9.6;P = 0.20)或QLQ-C30整体评分(1.9;95% CI,−4.9 ~ 8.8;P = .58)。在第100天,共同护理组的FACT-BMT总分更好(平均差异为6.6;95% CI, 1.0 ~ 12.1;P = .02), QLQ-C30整体评分(8.8;95% CI, 1.8 ~ 15.7;P = .02)。结论和相关性这项随机临床试验发现,共享护理在第100天的NRM不差,但在第180天的生活质量相似,在第100天的生活质量有所改善。这些数据表明,共享护理是安全的,可以在早期改善生活质量,并有可能成为hct后护理的常规模式。临床试验注册号:NCT03244826
{"title":"Shared Local Oncology Care After Allogeneic Hematopoietic Cell Transplantation","authors":"Gregory A. Abel, Haesook T. Kim, Ira Zackon, Edwin T. Alyea, Alexandra S. Bailey, John P. Winters, Kenneth R. Meehan, John L. Reagan, Jeanna H. Walsh, Thomas P. Walsh, Alexandra Ivanov, Meredith A. Faggen, Sarah Sinclair, Amy C. Joyce, Sara D. Close, Amy Emmert, Jon Koreth, Joseph H. Antin, Corey S. Cutler, Vincent T. Ho, Robert J. Soiffer","doi":"10.1001/jamaoncol.2024.5786","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.5786","url":null,"abstract":"ImportanceAlthough sharing care with local oncologists after allogeneic hematopoietic cell transplantation (HCT) has been proposed for patients living far from HCT centers, it is not known whether a shared strategy is safe or improves patient quality of life (QOL).ObjectiveTo determine the efficacy and safety of sharing follow-up care after HCT between the HCT specialty center and local oncologists.Design, Setting, and ParticipantsThis was a multicenter collaborative randomized clinical trial of patients undergoing HCT at Dana-Farber Cancer Institute (DFCI)—a high volume HCT center in Boston (Massachusetts)—and 8 local oncology practices. Eligible patients were enrolled from December 2017 to December 2021 and were randomized 1:1 to shared vs usual care after neutrophil engraftment, stratified by local sites in Massachusetts, Rhode Island, New Hampshire, New York, and Maine. Data analyses were performed in January 2024.InterventionShared care involved alternating post-HCT visits at DFCI and local oncology practices through day 100; for usual care, all post-HCT visits occurred only at DFCI.Main Outcomes and MeasuresCoprimary outcomes were nonrelapse mortality (NRM) at day 100, and QOL measured by the FACT-BMT (Functional Assessment of Cancer Therapy–Bone Marrow Transplantation) instrument and the QLQ-C30 (European Organization for Research and Treatment of Cancer’s Quality of Life Questionnaire) at day 180. Prespecified secondary outcomes included day 100 QOL and 1-year overall survival.ResultsA total of 302 participants (median [range] age, 63 [20-79] years; 117 [38.7%] females; 185 [61.3%] males) were included in the analysis; 152 were randomized to shared care and 150 to usual care. Day 100 NRM was noninferior for shared vs usual care (2.6% [95% CI, 0.7% to 6.6%] vs 2.7% [95% CI, 0.7% to 6.7%]; <jats:italic>P</jats:italic> = .98). There were no differences at day 180 for the FACT-BMT total score (mean difference, 3.8; 95% CI, −2.1 to 9.6; <jats:italic>P</jats:italic> = .20) or QLQ-C30 global score (1.9; 95% CI, −4.9 to 8.8; <jats:italic>P</jats:italic> = .58). At day 100, the FACT-BMT total score was better for shared care (mean difference, 6.6; 95% CI, 1.0 to 12.1; <jats:italic>P</jats:italic> = .02) as was the QLQ-C30 global score (8.8; 95% CI, 1.8 to 15.7; <jats:italic>P</jats:italic> = .02).Conclusions and RelevanceThis randomized clinical trial found that shared care resulted in noninferior NRM at day 100 but similar QOL at day 180, with improved QOL at day 100. These data suggest that shared care is safe, improves QOL early on, and has the potential to become a routine model for post-HCT care.Trial RegistrationClinicalTrials.gov Identifier: <jats:ext-link xmlns:xlink=\"http://www.w3.org/1999/xlink\" ext-link-type=\"uri\" xlink:href=\"https://clinicaltrials.gov/study/NCT03244826\">NCT03244826</jats:ext-link>","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"22 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142939875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immunotherapy Benefit Over Best Supportive Care in Hepatocellular Cancer With Child-Pugh B Dysfunction. 免疫治疗对Child-Pugh B功能障碍肝细胞癌的疗效优于最佳支持治疗。
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-26 DOI: 10.1001/jamaoncol.2024.5816
Manuel David Gil-Sierra,María Del Pilar Briceño-Casado,Cristina Moreno-Ramos
{"title":"Immunotherapy Benefit Over Best Supportive Care in Hepatocellular Cancer With Child-Pugh B Dysfunction.","authors":"Manuel David Gil-Sierra,María Del Pilar Briceño-Casado,Cristina Moreno-Ramos","doi":"10.1001/jamaoncol.2024.5816","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.5816","url":null,"abstract":"","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"306 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142887556","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular Testing for the World Health Organization Classification of Central Nervous System Tumors 世界卫生组织中枢神经系统肿瘤分类的分子检测
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-26 DOI: 10.1001/jamaoncol.2024.5506
Craig Horbinski, David A. Solomon, Rimas V. Lukas, Roger J. Packer, Priscilla Brastianos, Patrick Y. Wen, Matija Snuderl, Mitchel S. Berger, Susan Chang, Maryam Fouladi, Joanna J. Phillips, Burt Nabors, Daniel J. Brat, Jason T. Huse, Kenneth Aldape, Jann N. Sarkaria, Matthias Holdhoff, Terry C. Burns, Katherine B. Peters, Ingo K. Mellinghoff, David Arons, Evanthia Galanis
ImportanceMolecular techniques, including next-generation sequencing, genomic copy number profiling, fusion transcript detection, and genomic DNA methylation arrays, are now indispensable tools for the workup of central nervous system (CNS) tumors. Yet there remains a great deal of heterogeneity in using such biomarker testing across institutions and hospital systems. This is in large part because there is a persistent reluctance among third-party payers to cover molecular testing. The objective of this Review is to describe why comprehensive molecular biomarker testing is now required for the accurate diagnosis and grading and prognostication of CNS tumors and, in so doing, to justify more widespread use by clinicians and coverage by third-party payers.ObservationsThe 5th edition of the World Health Organization (WHO) classification system for CNS tumors incorporates specific molecular signatures into the essential diagnostic criteria for most tumor entities. Many CNS tumor types cannot be reliably diagnosed according to current WHO guidelines without molecular testing. The National Comprehensive Cancer Network also incorporates molecular testing into their guidelines for CNS tumors. Both sets of guidelines are maximally effective if they are implemented routinely for all patients with CNS tumors. Moreover, the cost of these tests is less than 5% of the overall average cost of caring for patients with CNS tumors and consistently improves management. This includes more accurate diagnosis and prognostication, clinical trial eligibility, and prediction of response to specific treatments. Each major group of CNS tumors in the WHO classification is evaluated and how molecular diagnostics enhances patient care is described.Conclusions and RelevanceRoutine advanced multidimensional molecular profiling is now required to provide optimal standard of care for patients with CNS tumors.
分子技术,包括下一代测序、基因组拷贝数分析、融合转录检测和基因组DNA甲基化阵列,现在是中枢神经系统(CNS)肿瘤检查不可或缺的工具。然而,在各机构和医院系统中使用这种生物标志物测试仍然存在很大的异质性。这在很大程度上是因为第三方支付者一直不愿支付分子检测费用。本综述的目的是描述为什么现在需要全面的分子生物标志物检测来准确诊断、分级和预测中枢神经系统肿瘤,并以此证明临床医生和第三方付款人更广泛地使用该检测。世界卫生组织(WHO)第五版中枢神经系统肿瘤分类系统将特异性分子特征纳入大多数肿瘤实体的基本诊断标准。根据目前的世卫组织指南,如果没有分子检测,许多中枢神经系统肿瘤类型无法得到可靠诊断。国家综合癌症网络也将分子检测纳入其中枢神经系统肿瘤的指南中。如果对所有中枢神经系统肿瘤患者常规实施这两套指南,它们将是最有效的。此外,这些检测的费用不到照顾中枢神经系统肿瘤患者总体平均费用的5%,并不断改善管理。这包括更准确的诊断和预测,临床试验资格,以及对特定治疗反应的预测。评估了世界卫生组织分类中的每一主要中枢神经系统肿瘤组,并描述了分子诊断如何提高患者护理。结论和相关性:目前需要常规的高级多维分子谱分析来为中枢神经系统肿瘤患者提供最佳的护理标准。
{"title":"Molecular Testing for the World Health Organization Classification of Central Nervous System Tumors","authors":"Craig Horbinski, David A. Solomon, Rimas V. Lukas, Roger J. Packer, Priscilla Brastianos, Patrick Y. Wen, Matija Snuderl, Mitchel S. Berger, Susan Chang, Maryam Fouladi, Joanna J. Phillips, Burt Nabors, Daniel J. Brat, Jason T. Huse, Kenneth Aldape, Jann N. Sarkaria, Matthias Holdhoff, Terry C. Burns, Katherine B. Peters, Ingo K. Mellinghoff, David Arons, Evanthia Galanis","doi":"10.1001/jamaoncol.2024.5506","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.5506","url":null,"abstract":"ImportanceMolecular techniques, including next-generation sequencing, genomic copy number profiling, fusion transcript detection, and genomic DNA methylation arrays, are now indispensable tools for the workup of central nervous system (CNS) tumors. Yet there remains a great deal of heterogeneity in using such biomarker testing across institutions and hospital systems. This is in large part because there is a persistent reluctance among third-party payers to cover molecular testing. The objective of this Review is to describe why comprehensive molecular biomarker testing is now required for the accurate diagnosis and grading and prognostication of CNS tumors and, in so doing, to justify more widespread use by clinicians and coverage by third-party payers.ObservationsThe 5th edition of the World Health Organization (WHO) classification system for CNS tumors incorporates specific molecular signatures into the essential diagnostic criteria for most tumor entities. Many CNS tumor types cannot be reliably diagnosed according to current WHO guidelines without molecular testing. The National Comprehensive Cancer Network also incorporates molecular testing into their guidelines for CNS tumors. Both sets of guidelines are maximally effective if they are implemented routinely for all patients with CNS tumors. Moreover, the cost of these tests is less than 5% of the overall average cost of caring for patients with CNS tumors and consistently improves management. This includes more accurate diagnosis and prognostication, clinical trial eligibility, and prediction of response to specific treatments. Each major group of CNS tumors in the WHO classification is evaluated and how molecular diagnostics enhances patient care is described.Conclusions and RelevanceRoutine advanced multidimensional molecular profiling is now required to provide optimal standard of care for patients with CNS tumors.","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"32 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142887225","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Deep Learning Model for Predicting Immunotherapy Response in Advanced Non−Small Cell Lung Cancer 预测晚期非小细胞肺癌免疫治疗反应的深度学习模型
IF 28.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-26 DOI: 10.1001/jamaoncol.2024.5356
Mehrdad Rakaee, Masoud Tafavvoghi, Biagio Ricciuti, Joao V. Alessi, Alessio Cortellini, Fabrizio Citarella, Lorenzo Nibid, Giuseppe Perrone, Elio Adib, Claudia A. M. Fulgenzi, Cassio Murilo Hidalgo Filho, Alessandro Di Federico, Falah Jabar, Sayed Hashemi, Ilias Houda, Elin Richardsen, Lill-Tove Rasmussen Busund, Tom Donnem, Idris Bahce, David J. Pinato, Åslaug Helland, Lynette M. Sholl, Mark M. Awad, David J. Kwiatkowski
ImportanceOnly a small fraction of patients with advanced non−small cell lung cancer (NSCLC) respond to immune checkpoint inhibitor (ICI) treatment. For optimal personalized NSCLC care, it is imperative to identify patients who are most likely to benefit from immunotherapy.ObjectiveTo develop a supervised deep learning−based ICI response prediction method; evaluate its performance alongside other known predictive biomarkers; and assess its association with clinical outcomes in patients with advanced NSCLC.Design, Setting, and ParticipantsThis multicenter cohort study developed and independently validated a deep learning−based response stratification model for predicting ICI treatment outcome in patients with advanced NSCLC from whole slide hematoxylin and eosin–stained images. Images for model development and validation were obtained from 1 participating center in the US and 3 in the European Union (EU) from August 2014 to December 2022. Data analyses were performed from September 2022 to May 2024.ExposureMonotherapy with ICIs.Main Outcomes and MeasuresModel performance measured by clinical end points and objective response rate (ORR) differentiation power vs other predictive biomarkers, ie, programmed death-ligand 1 (PD-L1), tumor mutational burden (TMB), and tumor-infiltrating lymphocytes (TILs).ResultsA total of 295 581 image tiles from 958 patients (mean [SD] age, 66.0 [10.6] years; 456 [48%] females and 502 [52%] males) treated with ICI for NSCLC were included in the analysis. The US-based development cohort consisted of 614 patients with median (IQR) follow-up time of 54.5 (38.2-68.1) months, and the EU-based validation cohort, 344 patients with 43.3 (27.4-53.9) months of follow-up. The ORR to ICI was 26% in the developmental cohort and 28% in the validation cohort. The deep learning model’s area under the receiver operating characteristic curve (AUC) for ORR was 0.75 (95% CI, 0.64-0.85) in the internal test set and 0.66 (95% CI, 0.60-0.72) in the validation cohort. In a multivariable analysis, the deep learning model’s score was an independent predictor of ICI response in the validation cohort for both progression-free (hazard ratio, 0.56; 95% CI, 0.42-0.76; <jats:italic>P</jats:italic> &amp;lt; .001) and overall survival (hazard ratio, 0.53; 95% CI, 0.39-0.73; <jats:italic>P</jats:italic> &amp;lt; .001). The tuned deep learning model achieved a higher AUC than TMB, TILs, and PD-L1 in the internal set; in the validation cohort, it was superior to TILs and comparable with PD-L1 (AUC, 0.67; 95% CI, 0.60-0.74), with a 10-percentage point improvement in specificity. In the validation cohort, combining the deep learning model with PD-L1 scores achieved an AUC of 0.70 (95% CI, 0.63-0.76), outperforming either marker alone, with a response rate of 51% compared to 41% for PD-L1 (≥50%) alone.Conclusions and RelevanceThe findings of this cohort study demonstrate a strong and independent deep learning−based feature associated with ICI respons
只有一小部分晚期非小细胞肺癌(NSCLC)患者对免疫检查点抑制剂(ICI)治疗有反应。为了获得最佳的个性化非小细胞肺癌治疗,必须确定最有可能从免疫治疗中获益的患者。目的建立一种基于监督深度学习的ICI响应预测方法;与其他已知的预测性生物标志物一起评估其性能;并评估其与晚期非小细胞肺癌患者临床结果的关系。设计、环境和参与者这项多中心队列研究开发并独立验证了基于深度学习的反应分层模型,该模型可通过苏木精和伊红染色全切片图像预测晚期NSCLC患者ICI治疗结果。用于模型开发和验证的图像从2014年8月至2022年12月从美国的1个参与中心和欧盟的3个参与中心获得。数据分析时间为2022年9月至2024年5月。暴露单药治疗。主要结果和测量方法通过临床终点和客观缓解率(ORR)分化能力与其他预测性生物标志物(即程序性死亡配体1 (PD-L1)、肿瘤突变负荷(TMB)和肿瘤浸润淋巴细胞(TILs))来衡量模型的性能。结果958例患者共获得295 581张图像贴片(平均[SD]年龄66.0[10.6]岁;456例(48%)女性和502例(52%)男性接受了非小细胞肺癌的ICI治疗。美国开发队列包括614例患者,中位(IQR)随访时间为54.5(38.2-68.1)个月,欧盟验证队列包括344例患者,随访时间为43.3(27.4-53.9)个月。发展组到ICI的ORR为26%,验证组为28%。深度学习模型的接受者工作特征曲线下面积(AUC)在内部测试集中为0.75 (95% CI, 0.64-0.85),在验证队列中为0.66 (95% CI, 0.60-0.72)。在多变量分析中,深度学习模型的评分是验证队列中ICI反应的独立预测因子,无进展(风险比,0.56;95% ci, 0.42-0.76;P, amp;肝移植;.001)和总生存率(风险比,0.53;95% ci, 0.39-0.73;P, amp;肝移植;措施)。调整后的深度学习模型在内部集的AUC高于TMB、TILs和PD-L1;在验证队列中,它优于TILs,与PD-L1相当(AUC, 0.67;95% CI, 0.60-0.74),特异性提高了10个百分点。在验证队列中,将深度学习模型与PD-L1评分相结合的AUC为0.70 (95% CI, 0.63-0.76),优于单独使用任何一种标记物,反应率为51%,而单独使用PD-L1(≥50%)的反应率为41%。结论和相关性这项队列研究的结果表明,在不同队列的NSCLC患者中,ICI反应具有强大且独立的基于深度学习的特征。该深度学习模型的临床应用可以提高治疗精度,更好地识别可能受益于ICI治疗晚期NSCLC的患者。
{"title":"Deep Learning Model for Predicting Immunotherapy Response in Advanced Non−Small Cell Lung Cancer","authors":"Mehrdad Rakaee, Masoud Tafavvoghi, Biagio Ricciuti, Joao V. Alessi, Alessio Cortellini, Fabrizio Citarella, Lorenzo Nibid, Giuseppe Perrone, Elio Adib, Claudia A. M. Fulgenzi, Cassio Murilo Hidalgo Filho, Alessandro Di Federico, Falah Jabar, Sayed Hashemi, Ilias Houda, Elin Richardsen, Lill-Tove Rasmussen Busund, Tom Donnem, Idris Bahce, David J. Pinato, Åslaug Helland, Lynette M. Sholl, Mark M. Awad, David J. Kwiatkowski","doi":"10.1001/jamaoncol.2024.5356","DOIUrl":"https://doi.org/10.1001/jamaoncol.2024.5356","url":null,"abstract":"ImportanceOnly a small fraction of patients with advanced non−small cell lung cancer (NSCLC) respond to immune checkpoint inhibitor (ICI) treatment. For optimal personalized NSCLC care, it is imperative to identify patients who are most likely to benefit from immunotherapy.ObjectiveTo develop a supervised deep learning−based ICI response prediction method; evaluate its performance alongside other known predictive biomarkers; and assess its association with clinical outcomes in patients with advanced NSCLC.Design, Setting, and ParticipantsThis multicenter cohort study developed and independently validated a deep learning−based response stratification model for predicting ICI treatment outcome in patients with advanced NSCLC from whole slide hematoxylin and eosin–stained images. Images for model development and validation were obtained from 1 participating center in the US and 3 in the European Union (EU) from August 2014 to December 2022. Data analyses were performed from September 2022 to May 2024.ExposureMonotherapy with ICIs.Main Outcomes and MeasuresModel performance measured by clinical end points and objective response rate (ORR) differentiation power vs other predictive biomarkers, ie, programmed death-ligand 1 (PD-L1), tumor mutational burden (TMB), and tumor-infiltrating lymphocytes (TILs).ResultsA total of 295 581 image tiles from 958 patients (mean [SD] age, 66.0 [10.6] years; 456 [48%] females and 502 [52%] males) treated with ICI for NSCLC were included in the analysis. The US-based development cohort consisted of 614 patients with median (IQR) follow-up time of 54.5 (38.2-68.1) months, and the EU-based validation cohort, 344 patients with 43.3 (27.4-53.9) months of follow-up. The ORR to ICI was 26% in the developmental cohort and 28% in the validation cohort. The deep learning model’s area under the receiver operating characteristic curve (AUC) for ORR was 0.75 (95% CI, 0.64-0.85) in the internal test set and 0.66 (95% CI, 0.60-0.72) in the validation cohort. In a multivariable analysis, the deep learning model’s score was an independent predictor of ICI response in the validation cohort for both progression-free (hazard ratio, 0.56; 95% CI, 0.42-0.76; &lt;jats:italic&gt;P&lt;/jats:italic&gt; &amp;amp;lt; .001) and overall survival (hazard ratio, 0.53; 95% CI, 0.39-0.73; &lt;jats:italic&gt;P&lt;/jats:italic&gt; &amp;amp;lt; .001). The tuned deep learning model achieved a higher AUC than TMB, TILs, and PD-L1 in the internal set; in the validation cohort, it was superior to TILs and comparable with PD-L1 (AUC, 0.67; 95% CI, 0.60-0.74), with a 10-percentage point improvement in specificity. In the validation cohort, combining the deep learning model with PD-L1 scores achieved an AUC of 0.70 (95% CI, 0.63-0.76), outperforming either marker alone, with a response rate of 51% compared to 41% for PD-L1 (≥50%) alone.Conclusions and RelevanceThe findings of this cohort study demonstrate a strong and independent deep learning−based feature associated with ICI respons","PeriodicalId":14850,"journal":{"name":"JAMA Oncology","volume":"304 1","pages":""},"PeriodicalIF":28.4,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142887224","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
JAMA Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1