首页 > 最新文献

ESMO Gastrointestinal Oncology最新文献

英文 中文
Gastric cancer hospital-based registry: real-world gastric cancer data from Latin America and Europe 胃癌医院登记:拉丁美洲和欧洲的真实胃癌数据
Pub Date : 2024-09-18 DOI: 10.1016/j.esmogo.2024.100088

Background

Gastric cancer has a high incidence and mortality rate worldwide. Epidemiological, clinical, and molecular features significantly impact patient outcomes. In regions lacking a national gastric cancer registry, hospital-based registries can provide crucial data that may aid in planning therapeutic strategies for the disease.

Methods

A retrospective observational cohort design was carried out in European Union (EU) and Latin American (LATAM) countries participating in the LEGACy project. Survival estimates were determined using actuarial Kaplan–Meier curves. Comparison was carried out with the log-rank test, and differences were considered statistically significant for P values ≤0.05.

Results

A total of 689 patients diagnosed with gastric cancer from November 2018 to November 2019 were included. Both cohorts had the body as the most common site reported (34.4% for EU and 51% for LATAM). The most used method for staging was computed tomography for both cohorts, although 6.9% of the LATAM population had positron emission tomography/computed tomography instead. Intestinal histological subtype was the most common (41.9% and 46.3% reported by EU and LATAM), while diffuse subtype was 44.9% for the LATAM and 21.3% for the EU population. Among patients tested for human epidermal growth factor receptor 2 (HER2), 12.5% were positive in the EU cohort and 13.8% in the LATAM cohort. For both cohorts, the most common site of human epidermal growth factor receptor 2 positivity was the gastroesophageal junction. Systemic treatment with curative intention was indicated in 50.7% in the EU cohort and 46.4% of the LATAM cohort. The most frequent scheme indicated for both the localized and the advanced setting was platinum-based (42.6% and 84.8% for EU and LATAM). Considering both cohorts, only 14.4% of the patients received second-line treatment, and 3% received a third-line treatment. After using Cox regression analysis, no difference in overall survival was reported, with a median of 10.9 months.

Conclusions

Despite the limitations of hospital-based registry analysis, our study has provided valuable insights into clinical characteristics and treatment approaches of EU and LATAM populations.

背景胃癌在全世界的发病率和死亡率都很高。流行病学、临床和分子特征对患者的预后有重大影响。在缺乏国家胃癌登记处的地区,以医院为基础的登记处可以提供重要的数据,从而帮助规划该疾病的治疗策略。方法 在参与 LEGACy 项目的欧盟(EU)和拉丁美洲(LATAM)国家开展了回顾性观察队列设计。采用精算卡普兰-梅耶曲线确定生存期估计值。比较采用对数秩检验,P值≤0.05则认为差异具有统计学意义。结果共纳入2018年11月至2019年11月期间确诊的689名胃癌患者。两个队列报告的最常见部位均为身体(欧盟为 34.4%,拉丁美洲和加勒比海地区为 51%)。两个队列中最常用的分期方法都是计算机断层扫描,但拉丁美洲和加勒比海地区有 6.9% 的人使用正电子发射断层扫描/计算机断层扫描。肠组织学亚型最常见(欧盟和拉美及加勒比地区报告的比例分别为 41.9% 和 46.3%),而弥漫亚型在拉美及加勒比地区为 44.9%,在欧盟人群中为 21.3%。在接受人类表皮生长因子受体 2(HER2)检测的患者中,欧盟组有 12.5%呈阳性,拉美和加勒比海地区组有 13.8%呈阳性。在这两个队列中,人表皮生长因子受体 2 阳性最常见的部位是胃食管交界处。在欧盟国家组中,50.7%的患者接受了以治愈为目的的全身治疗,而在拉丁美洲和加勒比海国家组中,46.4%的患者接受了以治愈为目的的全身治疗。无论是局部治疗还是晚期治疗,最常用的方案都是以铂为基础的治疗(欧盟和拉丁美洲分别为42.6%和84.8%)。在两个队列中,只有 14.4% 的患者接受了二线治疗,3% 的患者接受了三线治疗。结论尽管基于医院的登记分析存在局限性,但我们的研究为了解欧盟和拉丁美洲人口的临床特征和治疗方法提供了宝贵的见解。
{"title":"Gastric cancer hospital-based registry: real-world gastric cancer data from Latin America and Europe","authors":"","doi":"10.1016/j.esmogo.2024.100088","DOIUrl":"10.1016/j.esmogo.2024.100088","url":null,"abstract":"<div><h3>Background</h3><p>Gastric cancer has a high incidence and mortality rate worldwide. Epidemiological, clinical, and molecular features significantly impact patient outcomes. In regions lacking a national gastric cancer registry, hospital-based registries can provide crucial data that may aid in planning therapeutic strategies for the disease.</p></div><div><h3>Methods</h3><p>A retrospective observational cohort design was carried out in European Union (EU) and Latin American (LATAM) countries participating in the LEGACy project. Survival estimates were determined using actuarial Kaplan–Meier curves. Comparison was carried out with the log-rank test, and differences were considered statistically significant for <em>P</em> values ≤0.05.</p></div><div><h3>Results</h3><p>A total of 689 patients diagnosed with gastric cancer from November 2018 to November 2019 were included. Both cohorts had the body as the most common site reported (34.4% for EU and 51% for LATAM). The most used method for staging was computed tomography for both cohorts, although 6.9% of the LATAM population had positron emission tomography/computed tomography instead. Intestinal histological subtype was the most common (41.9% and 46.3% reported by EU and LATAM), while diffuse subtype was 44.9% for the LATAM and 21.3% for the EU population. Among patients tested for human epidermal growth factor receptor 2 (HER2), 12.5% were positive in the EU cohort and 13.8% in the LATAM cohort. For both cohorts, the most common site of human epidermal growth factor receptor 2 positivity was the gastroesophageal junction. Systemic treatment with curative intention was indicated in 50.7% in the EU cohort and 46.4% of the LATAM cohort. The most frequent scheme indicated for both the localized and the advanced setting was platinum-based (42.6% and 84.8% for EU and LATAM). Considering both cohorts, only 14.4% of the patients received second-line treatment, and 3% received a third-line treatment. After using Cox regression analysis, no difference in overall survival was reported, with a median of 10.9 months.</p></div><div><h3>Conclusions</h3><p>Despite the limitations of hospital-based registry analysis, our study has provided valuable insights into clinical characteristics and treatment approaches of EU and LATAM populations.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000499/pdfft?md5=5402a74d1083f8a4a81cbaad9c35b6ba&pid=1-s2.0-S2949819824000499-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142242232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Geriatric Assessment-guided therapy modification and outcomes in patients with non-metastatic gastroesophageal cancer: a retrospective cohort study☆ 老年病学评估指导下的非转移性胃食管癌患者治疗调整与疗效:一项回顾性队列研究☆。
Pub Date : 2024-09-12 DOI: 10.1016/j.esmogo.2024.100093

Background

Despite aggressive multimodal treatment for locally advanced esophagogastric cancer (LA-EGC), many patients experience early disease progression/death. We aimed to explore the role of Geriatric Assessment (GA) in optimizing patient care in older patients with LA-EGC.

Materials and methods

A retrospective cohort study was conducted in patients aged ≥60 years with LA-EGC referred to the geriatric oncology clinic at our institute between June 2018 and November 2022, who were planned for curative treatment. We explored the role of GA-guided therapy modifications on survival, identification of factors predicting potential ‘overtreatment’ (arbitrarily defined as patients in whom disease recurrence or death occurred within 6 months of treatment completion), and utility of the GA in identification of this patient subset.

Results

We enrolled 199 patients. The median age was 68 years (interquartile range 64-73 years). There were 131 (65.8%) males and 157 patients (78.9%) had a performance status of 0-1. Based on the GA, 110 (55.3%) patients were deemed fit (≤2 domains affected). Therapy modification (primarily de-intensification) occurred in 72 (36.2%) patients. At a median follow-up of 34.1 months [95% confidence interval (CI) 31.5-36.7 months], median event-free survival with de-intensified treatment was 12.2 months (95% CI 9.1-15.3 months) versus 18.8 months (95% CI 14.7-22.9 months) with standard treatment; P = 0.113. Median overall survival was 15.4 months (95% CI 9.3-21.5 months) with de-intensified treatment versus 21.1 months (95% CI 16.1-26.1 months) with standard treatment, P = 0.116. Six months following treatment completion, 79 (39.7%) patients were potentially overtreated. Initial GA failed to identify patients who were potentially overtreated (P = 0.923).

Conclusion

GA-tailored treatment de-escalation does not impair survival in older patients with LA-EGC but fails to identify the patient cohort at risk for overtreatment.

背景尽管对局部晚期食管胃癌(LA-EGC)进行了积极的多模式治疗,但许多患者仍经历了早期疾病进展/死亡。我们旨在探索老年病学评估(GA)在优化老年食管胃癌患者护理中的作用。材料与方法对2018年6月至2022年11月期间转诊至我院老年肿瘤门诊、计划接受根治性治疗的年龄≥60岁的食管胃癌患者进行了一项回顾性队列研究。我们探讨了GA指导下的治疗调整对生存期的影响,确定了预测潜在 "过度治疗"(任意定义为治疗结束后6个月内疾病复发或死亡的患者)的因素,以及GA在识别该患者亚群方面的效用。中位年龄为 68 岁(四分位数区间为 64-73 岁)。男性患者有 131 名(65.8%),157 名患者(78.9%)的表现状态为 0-1。根据GA,110名患者(55.3%)被认为适合治疗(受影响的领域≤2个)。72名患者(36.2%)接受了治疗调整(主要是降低强度)。中位随访时间为 34.1 个月[95% 置信区间 (CI) 31.5-36.7 个月],去强化治疗的中位无事件生存期为 12.2 个月 (95% CI 9.1-15.3 个月),而标准治疗的中位无事件生存期为 18.8 个月 (95% CI 14.7-22.9 个月);P = 0.113。去强化治疗的中位总生存期为 15.4 个月(95% CI 9.3-21.5 个月),而标准治疗为 21.1 个月(95% CI 16.1-26.1 个月);P = 0.116。治疗结束后六个月,79 名(39.7%)患者可能治疗过度。最初的GA未能识别出可能过度治疗的患者(P = 0.923)。结论GA定制的治疗降级不会影响LA-EGC老年患者的生存,但未能识别出有过度治疗风险的患者群体。
{"title":"Geriatric Assessment-guided therapy modification and outcomes in patients with non-metastatic gastroesophageal cancer: a retrospective cohort study☆","authors":"","doi":"10.1016/j.esmogo.2024.100093","DOIUrl":"10.1016/j.esmogo.2024.100093","url":null,"abstract":"<div><h3>Background</h3><p>Despite aggressive multimodal treatment for locally advanced esophagogastric cancer (LA-EGC), many patients experience early disease progression/death. We aimed to explore the role of Geriatric Assessment (GA) in optimizing patient care in older patients with LA-EGC.</p></div><div><h3>Materials and methods</h3><p>A retrospective cohort study was conducted in patients aged ≥60 years with LA-EGC referred to the geriatric oncology clinic at our institute between June 2018 and November 2022, who were planned for curative treatment. We explored the role of GA-guided therapy modifications on survival, identification of factors predicting potential ‘overtreatment’ (arbitrarily defined as patients in whom disease recurrence or death occurred within 6 months of treatment completion), and utility of the GA in identification of this patient subset.</p></div><div><h3>Results</h3><p>We enrolled 199 patients. The median age was 68 years (interquartile range 64-73 years). There were 131 (65.8%) males and 157 patients (78.9%) had a performance status of 0-1. Based on the GA, 110 (55.3%) patients were deemed fit (≤2 domains affected). Therapy modification (primarily de-intensification) occurred in 72 (36.2%) patients. At a median follow-up of 34.1 months [95% confidence interval (CI) 31.5-36.7 months], median event-free survival with de-intensified treatment was 12.2 months (95% CI 9.1-15.3 months) versus 18.8 months (95% CI 14.7-22.9 months) with standard treatment; <em>P</em> = 0.113. Median overall survival was 15.4 months (95% CI 9.3-21.5 months) with de-intensified treatment versus 21.1 months (95% CI 16.1-26.1 months) with standard treatment, <em>P</em> = 0.116. Six months following treatment completion, 79 (39.7%) patients were potentially overtreated. Initial GA failed to identify patients who were potentially overtreated (<em>P</em> = 0.923).</p></div><div><h3>Conclusion</h3><p>GA-tailored treatment de-escalation does not impair survival in older patients with LA-EGC but fails to identify the patient cohort at risk for overtreatment.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000542/pdfft?md5=91ec064e979d0034844d3c1e7188b953&pid=1-s2.0-S2949819824000542-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142172971","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PRABITAS study design: a pragmatic, randomized phase III trial of bi-weekly versus conventional trifluridine/tipiracil plus bevacizumab for metastatic colorectal cancer PRABITAS 研究设计:转移性结直肠癌双周疗法与常规三氟啶/替比西嘧啶加贝伐单抗疗法的务实、随机 III 期试验
Pub Date : 2024-09-01 DOI: 10.1016/j.esmogo.2024.100090

Trifluridine/tipiracil (FTD/TPI) plus bevacizumab (BEV) is the established therapy for refractory metastatic colorectal cancer, but there are concerns regarding the regimen’s complexity and hematotoxic effects, especially for patients with organ dysfunction, comorbidities, or a reduced performance status—groups often excluded from conventional clinical trials. Preliminary studies demonstrated that bi-weekly FTD/TPI + BEV may mitigate these hematotoxic effects compared with the conventional schedule without compromising efficacy. No clinical trials, however, have directly compared these two regimens. Therefore, we initiated the PRABITAS trial, a multicenter, randomized, phase III non-inferiority trial, to evaluate the efficacy and safety of bi-weekly FTD/TPI + BEV compared with conventional FTD/TPI + BEV. This was designed as a pragmatic trial, a novel approach in clinical trials aiming to aid decision-making in daily practice by mimicking real-world clinical settings. The PRABITAS trial incorporates minimal eligibility criteria to include a more representative patient population, allows flexibility in intervention adherence and assessment, and employs streamlined data collection to reduce the burden on both patients and healthcare providers. The primary endpoint is overall survival in the intention-to-treat population. Launched in December 2023, the trial aimed to enroll a total of 890 patients.

曲氟尿苷/替比拉西(FTD/TPI)+贝伐珠单抗(BEV)是治疗难治性转移性结直肠癌的成熟疗法,但该疗法的复杂性和血液毒性效应令人担忧,尤其是对于器官功能障碍、合并症或表现状态较差的患者--这些患者通常被排除在常规临床试验之外。初步研究表明,与常规治疗方案相比,每两周一次的 FTD/TPI + BEV 可减轻这些血液毒性反应,同时不影响疗效。然而,目前还没有临床试验对这两种治疗方案进行直接比较。因此,我们启动了 PRABITAS 试验,这是一项多中心、随机、III 期非劣效试验,旨在评估双周 FTD/TPI + BEV 与常规 FTD/TPI + BEV 相比的疗效和安全性。这是一项实用性试验,是临床试验中的一种新方法,旨在通过模拟真实的临床环境来帮助日常实践中的决策。PRABITAS 试验采用了最低资格标准,以纳入更具代表性的患者人群,允许灵活地坚持干预和评估,并采用简化的数据收集方法以减轻患者和医疗服务提供者的负担。主要终点是意向治疗人群的总生存期。该试验于2023年12月启动,旨在招募890名患者。
{"title":"PRABITAS study design: a pragmatic, randomized phase III trial of bi-weekly versus conventional trifluridine/tipiracil plus bevacizumab for metastatic colorectal cancer","authors":"","doi":"10.1016/j.esmogo.2024.100090","DOIUrl":"10.1016/j.esmogo.2024.100090","url":null,"abstract":"<div><p>Trifluridine/tipiracil (FTD/TPI) plus bevacizumab (BEV) is the established therapy for refractory metastatic colorectal cancer, but there are concerns regarding the regimen’s complexity and hematotoxic effects, especially for patients with organ dysfunction, comorbidities, or a reduced performance status—groups often excluded from conventional clinical trials. Preliminary studies demonstrated that bi-weekly FTD/TPI + BEV may mitigate these hematotoxic effects compared with the conventional schedule without compromising efficacy. No clinical trials, however, have directly compared these two regimens. Therefore, we initiated the PRABITAS trial, a multicenter, randomized, phase III non-inferiority trial, to evaluate the efficacy and safety of bi-weekly FTD/TPI + BEV compared with conventional FTD/TPI + BEV. This was designed as a pragmatic trial, a novel approach in clinical trials aiming to aid decision-making in daily practice by mimicking real-world clinical settings. The PRABITAS trial incorporates minimal eligibility criteria to include a more representative patient population, allows flexibility in intervention adherence and assessment, and employs streamlined data collection to reduce the burden on both patients and healthcare providers. The primary endpoint is overall survival in the intention-to-treat population. Launched in December 2023, the trial aimed to enroll a total of 890 patients.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000517/pdfft?md5=c62af2a7047f63261d89de04561562d4&pid=1-s2.0-S2949819824000517-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142096601","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
New perspectives in biliary tract cancers 胆道癌症的新视角
Pub Date : 2024-09-01 DOI: 10.1016/j.esmogo.2024.100092

Biliary tract cancer (BTC) is a rare but highly lethal malignancy. Despite recent advances in diagnosis and treatment, the overall prognosis remains dismal, with a median survival of <1 year in most cases. This highlights an urgent medical need for better treatment options, especially in the area of systematic treatments.

This review aims to give a concise overview of the current available treatment options for BTC, including a short summary of longstanding therapeutic approaches such as surgery, interventional techniques, radiotherapy, chemotherapy, and chemoradiotherapy. Special emphasis is placed on genetic alterations and treatment advances with immunotherapy in combination with chemotherapy, however, including current trials on new immunotherapeutic drugs. Furthermore, the recent recommendation by international guidelines to use durvalumab plus the combination of gemcitabine and cisplatin as a first-line treatment in the advanced setting is highlighted and the evidence supporting this recommendation is explored. Moreover, this review looks at genetic alterations which can be used as targets for immunotherapy, especially isocitrate dehydrogenase 1/2 (IDH1/2), fibroblast growth factor receptor 2 (FGFR2), and human epidermal growth factor receptor 2 (HER2/neu). Upcoming biomarkers such as microRNAs (miRNAs and especially miR-221) can possibly facilitate the choice of the appropriate treatment regimen in the future.

We conclude that there is a lot of recent development in the area of biomarker-driven targeted therapies and immunotherapies for BTC, which could consequently bring major benefits to patients’ treatment outcomes and quality of life.

胆道癌(BTC)是一种罕见但致死率极高的恶性肿瘤。尽管最近在诊断和治疗方面取得了进展,但总体预后仍然不容乐观,大多数病例的中位生存期仅为 1 年。本综述旨在简明扼要地概述目前可用的 BTC 治疗方案,包括对手术、介入技术、放疗、化疗和化学放疗等长期治疗方法的简短总结。然而,本文特别强调了基因改变和免疫疗法与化疗相结合的治疗进展,包括当前对新型免疫治疗药物的试验。此外,本综述还重点介绍了国际指南最近提出的建议,即在晚期患者中使用杜伐单抗联合吉西他滨和顺铂作为一线治疗,并探讨了支持这一建议的证据。此外,本综述还探讨了可作为免疫疗法靶点的基因改变,尤其是异柠檬酸脱氢酶1/2(IDH1/2)、成纤维细胞生长因子受体2(FGFR2)和人表皮生长因子受体2(HER2/neu)。我们的结论是,生物标记物驱动的 BTC 靶向疗法和免疫疗法领域近来取得了长足的发展,这将为患者的治疗效果和生活质量带来重大益处。
{"title":"New perspectives in biliary tract cancers","authors":"","doi":"10.1016/j.esmogo.2024.100092","DOIUrl":"10.1016/j.esmogo.2024.100092","url":null,"abstract":"<div><p>Biliary tract cancer (BTC) is a rare but highly lethal malignancy. Despite recent advances in diagnosis and treatment, the overall prognosis remains dismal, with a median survival of &lt;1 year in most cases. This highlights an urgent medical need for better treatment options, especially in the area of systematic treatments.</p><p>This review aims to give a concise overview of the current available treatment options for BTC, including a short summary of longstanding therapeutic approaches such as surgery, interventional techniques, radiotherapy, chemotherapy, and chemoradiotherapy. Special emphasis is placed on genetic alterations and treatment advances with immunotherapy in combination with chemotherapy, however, including current trials on new immunotherapeutic drugs. Furthermore, the recent recommendation by international guidelines to use durvalumab plus the combination of gemcitabine and cisplatin as a first-line treatment in the advanced setting is highlighted and the evidence supporting this recommendation is explored. Moreover, this review looks at genetic alterations which can be used as targets for immunotherapy, especially isocitrate dehydrogenase 1/2 (<em>IDH1/2</em>), fibroblast growth factor receptor 2 (<em>FGFR2</em>), and human epidermal growth factor receptor 2 (<em>HER2/neu</em>). Upcoming biomarkers such as microRNAs (miRNAs and especially miR-221) can possibly facilitate the choice of the appropriate treatment regimen in the future.</p><p>We conclude that there is a lot of recent development in the area of biomarker-driven targeted therapies and immunotherapies for BTC, which could consequently bring major benefits to patients’ treatment outcomes and quality of life.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000530/pdfft?md5=f3580b29dcc2c0311d1469921ce03659&pid=1-s2.0-S2949819824000530-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142096603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hepatic chemosaturation with melphalan in patients with primary or secondary liver tumors with or without extrahepatic tumor manifestation 在有或无肝外肿瘤表现的原发性或继发性肝肿瘤患者中使用美法仑进行肝化疗
Pub Date : 2024-08-22 DOI: 10.1016/j.esmogo.2024.100082

Background

Hepatic chemosaturation by isolated percutaneous liver perfusion (CS-PHP) with melphalan controls hepatic tumor growth. However, optimal treatment frequency and the prognostic relevance of extrahepatic tumor manifestation remain unclear. We analyzed response and tolerability of repeated treatment in regular cycles of CS-PHP.

Materials and methods

CS-PHP was administered to patients with primary or secondary liver tumors. Overall survival (OS) and hepatic disease control rate (hDCR) were assessed retrospectively by modified RECIST after at least one response assessment, and toxicity by Common Terminology Criteria for Adverse Events v4.03.

Results

A total of 97 CS-PHP treatments were carried out in 33 patients between 2016 and 2023. Patients had unresectable intrahepatic metastases of uveal melanoma (n = 19), intrahepatic cholangiocarcinoma (n = 8), hepatocellular carcinoma (n = 2), ciliary body melanoma (n = 1), acinar cell carcinoma (n = 1), pancreatic cancer (n = 1) or tonsil cancer (n = 1). CS-PHP was carried out seven times in 1 patient, six times in 5, five times in 3, four times in 2, three times in 4, twice in 7 and once in 11 patients. The median OS was 65 weeks (standard error 13.6, 95% confidence interval 38.2-91.5 weeks). hDCR was 91% (30 of 33 patients) at last observation time point. Extrahepatic tumor manifestations were not associated with OS. CS-PHP was well tolerated. Grade III or IV pancytopenia occurred in two patients.

Conclusion

CS-PHP induced hepatic disease control in the majority of patients. Extrahepatic tumor manifestation had no significant impact on OS. The relevance of CS-PHP as long-term treatment needs to be validated in future studies.

背景通过孤立经皮肝灌注(CS-PHP)与美啡仑进行肝化疗可控制肝肿瘤的生长。然而,最佳治疗频率和肝外肿瘤表现的预后相关性仍不清楚。我们对CS-PHP常规周期重复治疗的反应和耐受性进行了分析。结果2016年至2023年间,33名患者共接受了97次CS-PHP治疗。患者的肝内转移灶为不可切除的葡萄膜黑色素瘤(19 例)、肝内胆管癌(8 例)、肝细胞癌(2 例)、睫状体黑色素瘤(1 例)、尖细胞癌(1 例)、胰腺癌(1 例)或扁桃体癌(1 例)。1名患者进行了7次CS-PHP,5名患者进行了6次,3名患者进行了5次,2名患者进行了4次,4名患者进行了3次,7名患者进行了2次,11名患者进行了1次。中位生存期为 65 周(标准误差为 13.6,95% 置信区间为 38.2-91.5 周),最后观察时间点的 hDCR 为 91%(33 例患者中的 30 例)。肝外肿瘤表现与OS无关。CS-PHP 的耐受性良好。结论CS-PHP可使大多数患者的肝病得到控制。肝外肿瘤表现对 OS 无明显影响。CS-PHP 作为长期治疗的相关性还需要在未来的研究中加以验证。
{"title":"Hepatic chemosaturation with melphalan in patients with primary or secondary liver tumors with or without extrahepatic tumor manifestation","authors":"","doi":"10.1016/j.esmogo.2024.100082","DOIUrl":"10.1016/j.esmogo.2024.100082","url":null,"abstract":"<div><h3>Background</h3><p>Hepatic chemosaturation by isolated percutaneous liver perfusion (CS-PHP) with melphalan controls hepatic tumor growth. However, optimal treatment frequency and the prognostic relevance of extrahepatic tumor manifestation remain unclear. We analyzed response and tolerability of repeated treatment in regular cycles of CS-PHP.</p></div><div><h3>Materials and methods</h3><p>CS-PHP was administered to patients with primary or secondary liver tumors. Overall survival (OS) and hepatic disease control rate (hDCR) were assessed retrospectively by modified RECIST after at least one response assessment, and toxicity by Common Terminology Criteria for Adverse Events v4.03.</p></div><div><h3>Results</h3><p>A total of 97 CS-PHP treatments were carried out in 33 patients between 2016 and 2023. Patients had unresectable intrahepatic metastases of uveal melanoma (<em>n</em> = 19), intrahepatic cholangiocarcinoma (<em>n</em> = 8), hepatocellular carcinoma (<em>n</em> = 2), ciliary body melanoma (<em>n</em> = 1), acinar cell carcinoma (<em>n</em> = 1), pancreatic cancer (<em>n</em> = 1) or tonsil cancer (<em>n</em> = 1). CS-PHP was carried out seven times in 1 patient, six times in 5, five times in 3, four times in 2, three times in 4, twice in 7 and once in 11 patients. The median OS was 65 weeks (standard error 13.6, 95% confidence interval 38.2-91.5 weeks). hDCR was 91% (30 of 33 patients) at last observation time point. Extrahepatic tumor manifestations were not associated with OS. CS-PHP was well tolerated. Grade III or IV pancytopenia occurred in two patients.</p></div><div><h3>Conclusion</h3><p>CS-PHP induced hepatic disease control in the majority of patients. Extrahepatic tumor manifestation had no significant impact on OS. The relevance of CS-PHP as long-term treatment needs to be validated in future studies.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000438/pdfft?md5=b7686ff207b1ad5a4ea8b627711b8bc7&pid=1-s2.0-S2949819824000438-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142040795","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Concordance of PD-L1 status in primary gastroesophageal adenocarcinoma and matched peritoneal metastases: a single institution study 原发性胃食管腺癌和匹配的腹膜转移瘤中 PD-L1 状态的一致性:一项单机构研究
Pub Date : 2024-08-19 DOI: 10.1016/j.esmogo.2024.100089

Background

Programmed death-ligand 1 (PD-L1) expression serves as a predictive biomarker for response to immune checkpoint inhibitors (ICIs) in metastatic gastroesophageal adenocarcinoma (mGEA). The peritoneum is one of the most common metastatic sites and is associated with a poor prognosis and apparently lower clinical efficacy of ICIs.

Patients and methods

We investigated the heterogeneity of PD-L1 expression in mGEA by examining its concordance between primary tumors and matched peritoneal metastases (PMs), and before and after systemic treatment. PD-L1 expression was assessed using combined positive score (CPS) by immunohistochemistry with VENTANA PD-L1 (SP263) assays on formalin-fixed paraffin-embedded tumor tissues. Results were reported using CPS cut-offs of 1 and 5.

Results

Twenty-two primary tumor and matched PM specimens from patients with mGEA were analyzed. The concordance of PD-L1 CPS was 54.5% with a CPS cut-off of 1 and 72.7% with a CPS cut-off of 5, highlighting spatial heterogeneity. Notably, none of the PD-L1-positive primary tumor samples tested positive in the matched PM specimens using the CPS ≥5 cut-off. Potential temporal heterogeneity of PD-L1 expression related to chemo(immuno)therapy administration was also observed, with a 55.6% concordance rate using the CPS ≥5 cut-off.

Conclusions

PD-L1 expression in PMs from mGEA is characterized by both spatial and potentially temporal heterogeneity, with the variability being more pronounced at lower CPS cut-off values. This variability complicates its role as a predictive biomarker for ICI outcomes. The high intrapatient discordance rate in PD-L1 CPS expression between positive primary tumor samples and matched PM specimens suggests that peritoneal specimens should not be used as the only source for PD-L1 assessment if representative tissue from other disease sites is available.

背景程序性死亡配体1(PD-L1)表达是转移性胃食管腺癌(mGEA)对免疫检查点抑制剂(ICIs)反应的预测性生物标志物。腹膜是最常见的转移部位之一,与不良预后和ICIs明显较低的临床疗效有关。患者和方法我们通过研究原发肿瘤和匹配的腹膜转移瘤(PMs)之间以及系统治疗前后PD-L1表达的一致性,研究了PD-L1在mGEA中表达的异质性。在福尔马林固定的石蜡包埋肿瘤组织上使用 VENTANA PD-L1 (SP263) 检测试剂盒进行免疫组化,以联合阳性评分(CPS)评估 PD-L1 的表达。结果分析了 22 例 mGEA 患者的原发肿瘤和匹配的 PM 标本。当 CPS 临界值为 1 时,PD-L1 CPS 的一致性为 54.5%,而当 CPS 临界值为 5 时,一致性为 72.7%,突出了空间异质性。值得注意的是,使用CPS≥5截断值时,没有一个PD-L1阳性的原发肿瘤样本在匹配的PM标本中检测出阳性。结论PD-L1在mGEA肿瘤标本中的表达具有空间异质性和潜在的时间异质性,在CPS截断值较低时,其变异性更为明显。这种变异性使其作为 ICI 结果预测生物标志物的作用变得更加复杂。阳性原发肿瘤样本与匹配的 PM 标本之间在 PD-L1 CPS 表达上的高患者间不一致性表明,如果有其他疾病部位的代表性组织,腹膜标本不应作为 PD-L1 评估的唯一来源。
{"title":"Concordance of PD-L1 status in primary gastroesophageal adenocarcinoma and matched peritoneal metastases: a single institution study","authors":"","doi":"10.1016/j.esmogo.2024.100089","DOIUrl":"10.1016/j.esmogo.2024.100089","url":null,"abstract":"<div><h3>Background</h3><p>Programmed death-ligand 1 (PD-L1) expression serves as a predictive biomarker for response to immune checkpoint inhibitors (ICIs) in metastatic gastroesophageal adenocarcinoma (mGEA). The peritoneum is one of the most common metastatic sites and is associated with a poor prognosis and apparently lower clinical efficacy of ICIs.</p></div><div><h3>Patients and methods</h3><p>We investigated the heterogeneity of PD-L1 expression in mGEA by examining its concordance between primary tumors and matched peritoneal metastases (PMs), and before and after systemic treatment. PD-L1 expression was assessed using combined positive score (CPS) by immunohistochemistry with VENTANA PD-L1 (SP263) assays on formalin-fixed paraffin-embedded tumor tissues. Results were reported using CPS cut-offs of 1 and 5.</p></div><div><h3>Results</h3><p>Twenty-two primary tumor and matched PM specimens from patients with mGEA were analyzed. The concordance of PD-L1 CPS was 54.5% with a CPS cut-off of 1 and 72.7% with a CPS cut-off of 5, highlighting spatial heterogeneity. Notably, none of the PD-L1-positive primary tumor samples tested positive in the matched PM specimens using the CPS ≥5 cut-off. Potential temporal heterogeneity of PD-L1 expression related to chemo(immuno)therapy administration was also observed, with a 55.6% concordance rate using the CPS ≥5 cut-off.</p></div><div><h3>Conclusions</h3><p>PD-L1 expression in PMs from mGEA is characterized by both spatial and potentially temporal heterogeneity, with the variability being more pronounced at lower CPS cut-off values. This variability complicates its role as a predictive biomarker for ICI outcomes. The high intrapatient discordance rate in PD-L1 CPS expression between positive primary tumor samples and matched PM specimens suggests that peritoneal specimens should not be used as the only source for PD-L1 assessment if representative tissue from other disease sites is available.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000505/pdfft?md5=6d36295a9ac2218ba01822ea0f63acaf&pid=1-s2.0-S2949819824000505-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142006917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Real-world uptake and effectiveness of triplet versus doublet chemotherapy with bevacizumab in patients with metastatic colorectal cancer in the United States 美国转移性结直肠癌患者接受三联化疗和贝伐单抗双联化疗的实际情况和效果
Pub Date : 2024-08-14 DOI: 10.1016/j.esmogo.2024.100087

Introduction

Triplet chemotherapy + bevacizumab (TripletBev) demonstrated an overall survival (OS) benefit for patients with newly diagnosed metastatic colorectal cancer in randomized trials. We aimed to evaluate the uptake and estimate the effectiveness of TripletBev versus doublet chemotherapy + bevacizumab (DoubletBev) in a real-world population in the United States.

Methods

We carried out a retrospective cohort study of patients initiating first-line treatment of metastatic colorectal cancer between 23 October 2014 and 31 October 2022 using the Flatiron Health nationwide electronic health record (EHR)-derived de-identified database. The data originated from ∼280 cancer clinics (∼800 sites of care) in the USA. The primary analysis compared OS between patients receiving TripletBev or DoubletBev using a Cox proportional hazards model with adjustment for pre-specified covariates using stabilized inverse probability of treatment weighting. This analysis was also carried out within pre-specified and post hoc subgroups. A secondary analysis used Stürmer-trimming of the propensity score distribution to include patients most likely to be eligible to receive either treatment.

Results

Some 391 patients received TripletBev and 9625 received DoubletBev. There was no association between TripletBev and OS in the primary analysis [hazard ratio (HR) 0.92; 95% confidence interval (CI) 0.75-1.13]. TripletBev was associated with lower hazard of death in patients with synchronous metastases (HR 0.79; 95% CI 0.64-0.98; statistically significant) and in the secondary analysis of patients most likely to be eligible to receive either treatment (HR 0.80; 95% CI 0.63-1.02; non-statistically significant).

Conclusion

Uptake of TripletBev remains low and the primary analysis did not demonstrate effectiveness in a real-world population in the United States. Patients with synchronous metastases and those most likely to be eligible to receive either treatment may be most likely to benefit from TripletBev.

导言在随机试验中,三联化疗+贝伐单抗(TripletBev)为新诊断的转移性结直肠癌患者带来了总生存期(OS)的益处。我们的目的是评估美国真实世界人群中三联化疗+贝伐单抗(DoubletBev)与双联化疗+贝伐单抗(DoubletBev)的使用率,并估算其有效性。方法我们利用Flatiron Health全国电子健康记录(EHR)衍生的去身份化数据库,对2014年10月23日至2022年10月31日期间开始一线治疗的转移性结直肠癌患者进行了一项回顾性队列研究。数据来自美国 280 家癌症诊所(800 个医疗点)。主要分析采用 Cox 比例危险模型,使用稳定的逆概率治疗加权法对预先指定的协变量进行调整,比较了接受 TripletBev 或 DoubletBev 治疗的患者的 OS。该分析还在预先指定的亚组和事后亚组中进行。二次分析使用了倾向得分分布的Stürmer-trimming方法,以纳入最有可能接受两种治疗方法的患者。结果约有391名患者接受了TripletBev治疗,9625名患者接受了DoubletBev治疗。在主要分析中,TripletBev 和 OS 之间没有关联[危险比 (HR) 0.92; 95% 置信区间 (CI) 0.75-1.13]。TripletBev与同步转移患者较低的死亡风险有关(HR 0.79;95% CI 0.64-0.98;有统计学意义),在最有可能接受两种治疗的患者的二次分析中也是如此(HR 0.80;95% CI 0.63-1.02;无统计学意义)。同步转移患者和最有可能接受两种治疗的患者可能最有可能从 TripletBev 中获益。
{"title":"Real-world uptake and effectiveness of triplet versus doublet chemotherapy with bevacizumab in patients with metastatic colorectal cancer in the United States","authors":"","doi":"10.1016/j.esmogo.2024.100087","DOIUrl":"10.1016/j.esmogo.2024.100087","url":null,"abstract":"<div><h3>Introduction</h3><p>Triplet chemotherapy + bevacizumab (TripletBev) demonstrated an overall survival (OS) benefit for patients with newly diagnosed metastatic colorectal cancer in randomized trials. We aimed to evaluate the uptake and estimate the effectiveness of TripletBev versus doublet chemotherapy + bevacizumab (DoubletBev) in a real-world population in the United States.</p></div><div><h3>Methods</h3><p>We carried out a retrospective cohort study of patients initiating first-line treatment of metastatic colorectal cancer between 23 October 2014 and 31 October 2022 using the Flatiron Health nationwide electronic health record (EHR)-derived de-identified database. The data originated from ∼280 cancer clinics (∼800 sites of care) in the USA. The primary analysis compared OS between patients receiving TripletBev or DoubletBev using a Cox proportional hazards model with adjustment for pre-specified covariates using stabilized inverse probability of treatment weighting. This analysis was also carried out within pre-specified and <em>post hoc</em> subgroups. A secondary analysis used Stürmer-trimming of the propensity score distribution to include patients most likely to be eligible to receive either treatment.</p></div><div><h3>Results</h3><p>Some 391 patients received TripletBev and 9625 received DoubletBev. There was no association between TripletBev and OS in the primary analysis [hazard ratio (HR) 0.92; 95% confidence interval (CI) 0.75-1.13]. TripletBev was associated with lower hazard of death in patients with synchronous metastases (HR 0.79; 95% CI 0.64-0.98; statistically significant) and in the secondary analysis of patients most likely to be eligible to receive either treatment (HR 0.80; 95% CI 0.63-1.02; non-statistically significant).</p></div><div><h3>Conclusion</h3><p>Uptake of TripletBev remains low and the primary analysis did not demonstrate effectiveness in a real-world population in the United States. Patients with synchronous metastases and those most likely to be eligible to receive either treatment may be most likely to benefit from TripletBev.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000487/pdfft?md5=401a17b322bd7cb0a0afa3d07ee7fcc1&pid=1-s2.0-S2949819824000487-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141990518","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeted therapies in biliary tract cancer—when precision becomes imprecise 胆道癌的靶向治疗--当精确变得不精确时
Pub Date : 2024-08-13 DOI: 10.1016/j.esmogo.2024.100085

Advanced biliary tract cancers (BTCs) have gained notoriety among gastrointestinal tumours for their comparatively high incidence of actionable alterations and their compelling benefit from targeted therapies matched to these alterations. Such successes are exemplified by BTC-specific approvals of fibroblast growth factor receptor (FGFR) inhibitors for tumours with FGFR2 rearrangements, as well as mutant isocitrate dehydrogenase 1 inhibitors. Nevertheless, there is a clear absence of therapeutic benefit in a subset of patients despite their tumours fulfilling the current molecular criteria for treatment with these drugs. This results in inefficient management of patients with otherwise bleak prognosis, as well as considerable financial burden. Even among responders, the duration of response is limited, a clinical observation that could be considered unusual as these inhibitors typically target driver genes hypothesised to be responsible for tumour formation. However, BTCs exhibit oncogenic addiction to signalling networks rather than individual genes, and by extension, therapeutic response is dependent on these signalling networks rather than simply the status of the specific target gene. Primary resistance is mediated by co-occurring genetic (DNA) and non-genetic (transcriptional, translational, post-translational) alterations in members of signalling networks that are upstream, downstream, or in parallel pathways to the target alteration. Refining the molecular criteria to select patients is a necessary next step, by incorporating co-occurrence of resistance biomarkers as individual parameters or into predictors of treatment benefit. Characterising the molecular bases of resistance to targeted therapies will fuel next-generation combination treatments, maximising the catchment of responders and enhancing the duration of response.

在胃肠道肿瘤中,晚期胆道癌(BTC)因其相对较高的可操作性改变发生率和与这些改变相匹配的靶向疗法而声名鹊起。针对成纤维细胞生长因子受体(FGFR)抑制剂和突变型异柠檬酸脱氢酶1抑制剂获批用于治疗FGFR2重排的肿瘤,就是这些疗法取得成功的例证。然而,尽管一部分患者的肿瘤符合目前使用这些药物治疗的分子标准,但其治疗效果明显不佳。这导致对预后不佳的患者的管理效率低下,并造成巨大的经济负担。即使在有反应的患者中,反应的持续时间也很有限,这种临床观察结果可能被认为是不寻常的,因为这些抑制剂通常针对的是假定导致肿瘤形成的驱动基因。然而,BTCs 表现出对信号网络而非单个基因的致癌成瘾性,推而广之,治疗反应取决于这些信号网络,而不仅仅是特定靶基因的状态。原发性耐药性是由信号网络成员中同时发生的遗传(DNA)和非遗传(转录、翻译、翻译后)改变介导的,这些改变是靶点改变的上游、下游或平行途径。下一步必须完善选择患者的分子标准,将抗药性生物标志物的共同出现作为单独参数或纳入治疗获益预测指标。确定靶向疗法耐药性的分子基础将有助于下一代联合疗法的开发,最大限度地扩大耐药患者的范围并延长耐药时间。
{"title":"Targeted therapies in biliary tract cancer—when precision becomes imprecise","authors":"","doi":"10.1016/j.esmogo.2024.100085","DOIUrl":"10.1016/j.esmogo.2024.100085","url":null,"abstract":"<div><p>Advanced biliary tract cancers (BTCs) have gained notoriety among gastrointestinal tumours for their comparatively high incidence of actionable alterations and their compelling benefit from targeted therapies matched to these alterations. Such successes are exemplified by BTC-specific approvals of fibroblast growth factor receptor (FGFR) inhibitors for tumours with <em>FGFR2</em> rearrangements, as well as mutant isocitrate dehydrogenase 1 inhibitors. Nevertheless, there is a clear absence of therapeutic benefit in a subset of patients despite their tumours fulfilling the current molecular criteria for treatment with these drugs. This results in inefficient management of patients with otherwise bleak prognosis, as well as considerable financial burden. Even among responders, the duration of response is limited, a clinical observation that could be considered unusual as these inhibitors typically target driver genes hypothesised to be responsible for tumour formation. However, BTCs exhibit oncogenic addiction to signalling networks rather than individual genes, and by extension, therapeutic response is dependent on these signalling networks rather than simply the status of the specific target gene. Primary resistance is mediated by co-occurring genetic (DNA) and non-genetic (transcriptional, translational, post-translational) alterations in members of signalling networks that are upstream, downstream, or in parallel pathways to the target alteration. Refining the molecular criteria to select patients is a necessary next step, by incorporating co-occurrence of resistance biomarkers as individual parameters or into predictors of treatment benefit. Characterising the molecular bases of resistance to targeted therapies will fuel next-generation combination treatments, maximising the catchment of responders and enhancing the duration of response.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000463/pdfft?md5=f000b02106c406f79be99af0aa5a9a09&pid=1-s2.0-S2949819824000463-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141978984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Current practices and challenges in implementing precision medicine for upper gastrointestinal cancers in European academic centers: an EORTC survey 欧洲学术中心针对上消化道癌症实施精准医疗的当前做法和挑战:EORTC 调查
Pub Date : 2024-08-08 DOI: 10.1016/j.esmogo.2024.100074

Background

Precision oncology is gaining momentum in managing patients with gastrointestinal cancers. This study examines the implementation of personalized medicine technologies in upper gastrointestinal (UGI) oncology across European academic centers.

Material and methods

Forty-five oncology specialists from 41 European institutions completed a survey designed by the Personalized Medicine Task Force of the European Organization of Research and Treatment of Cancer (EORTC) Gastrointestinal Tract Cancer Working Group, providing insights into molecular testing, timing, availability of targeted therapies, funding sources, and utilization of molecular tumor boards (MTBs) for patients with UGI cancers. Frequencies and percentages were calculated.

Results

Routine testing for human epidermal growth factor receptor 2 (HER2, 100%), programmed death-ligand 1 (PD-L1, 89%), and DNA mismatch repair (MMR, 91%) is implemented in most centers. Comprehensive gene panels on tumor tissue are frequently utilized, especially in biliary tract cancer, with almost all centers incorporating them into routine practice. Blood-based sequencing is increasingly employed, and half of centers carry out comprehensive gene panels for circulating tumor DNA analyses. MTBs are regularly held in 76% of centers, predominantly utilizing ESMO Scale for Clinical Actionability of molecular Targets (ESCAT)-based recommendations for tissue molecular alterations. The translation of genomic information into prescribed treatments remains limited, however, with the majority of centers reporting ∼25% of molecularly stratified treatment decisions following comprehensive genetic testing.

Conclusion

This survey provides important insights into current personalized medicine practice in European academic clinical centers for UGI oncology. Despite widespread adoption of molecular testing and implementation of MTBs, further efforts are needed to optimize the integration of personalized medicine into clinical practice.

背景精准肿瘤学在管理胃肠道癌症患者方面的发展势头日益强劲。材料与方法来自欧洲 41 家机构的 45 名肿瘤学专家完成了由欧洲癌症研究和治疗组织(EORTC)胃肠道癌症工作组个性化医学工作组设计的调查,提供了有关胃肠道癌症患者的分子检测、时机、靶向治疗的可用性、资金来源以及分子肿瘤委员会(MTB)的使用情况。结果大多数中心都对人类表皮生长因子受体 2(HER2,100%)、程序性死亡配体 1(PD-L1,89%)和 DNA 错配修复(MMR,91%)进行了常规检测。肿瘤组织上的综合基因面板经常被使用,尤其是在胆道癌中,几乎所有中心都将其纳入了常规实践。血液测序的应用越来越广泛,半数中心在循环肿瘤DNA分析中采用了综合基因检测。76%的中心定期进行MTB,主要采用基于ESMO分子靶点临床可操作性量表(ESCAT)的组织分子改变建议。然而,将基因组信息转化为处方治疗的工作仍然有限,大多数中心报告的综合基因检测后分子分层治疗决策的比例为 25%。尽管分子检测已被广泛采用并实施了 MTB,但仍需进一步努力将个性化医疗优化整合到临床实践中。
{"title":"Current practices and challenges in implementing precision medicine for upper gastrointestinal cancers in European academic centers: an EORTC survey","authors":"","doi":"10.1016/j.esmogo.2024.100074","DOIUrl":"10.1016/j.esmogo.2024.100074","url":null,"abstract":"<div><h3>Background</h3><p>Precision oncology is gaining momentum in managing patients with gastrointestinal cancers. This study examines the implementation of personalized medicine technologies in upper gastrointestinal (UGI) oncology across European academic centers.</p></div><div><h3>Material and methods</h3><p>Forty-five oncology specialists from 41 European institutions completed a survey designed by the Personalized Medicine Task Force of the European Organization of Research and Treatment of Cancer (EORTC) Gastrointestinal Tract Cancer Working Group, providing insights into molecular testing, timing, availability of targeted therapies, funding sources, and utilization of molecular tumor boards (MTBs) for patients with UGI cancers. Frequencies and percentages were calculated.</p></div><div><h3>Results</h3><p>Routine testing for human epidermal growth factor receptor 2 (HER2, 100%), programmed death-ligand 1 (PD-L1, 89%), and DNA mismatch repair (MMR, 91%) is implemented in most centers. Comprehensive gene panels on tumor tissue are frequently utilized, especially in biliary tract cancer, with almost all centers incorporating them into routine practice. Blood-based sequencing is increasingly employed, and half of centers carry out comprehensive gene panels for circulating tumor DNA analyses. MTBs are regularly held in 76% of centers, predominantly utilizing ESMO Scale for Clinical Actionability of molecular Targets (ESCAT)-based recommendations for tissue molecular alterations. The translation of genomic information into prescribed treatments remains limited, however, with the majority of centers reporting ∼25% of molecularly stratified treatment decisions following comprehensive genetic testing.</p></div><div><h3>Conclusion</h3><p>This survey provides important insights into current personalized medicine practice in European academic clinical centers for UGI oncology. Despite widespread adoption of molecular testing and implementation of MTBs, further efforts are needed to optimize the integration of personalized medicine into clinical practice.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000359/pdfft?md5=6b82e65311ef292351b3cf2cc906e667&pid=1-s2.0-S2949819824000359-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141953155","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Clinical implementation of simultaneous multiple biomarkers testing for metastatic or recurrent gastroesophageal adenocarcinoma: a single-institutional experience 转移性或复发性胃食管腺癌多种生物标记物同步检测的临床实施:单个机构的经验
Pub Date : 2024-08-02 DOI: 10.1016/j.esmogo.2024.100086

Background

Evaluating multiple biomarkers including human epidermal growth factor receptor 2 (HER2), mismatch repair (MMR) status, programed death-ligand 1 (PD-L1), and claudin 18.2 (CLDN18.2) is essential for selecting appropriate first-line therapy of metastatic gastroesophageal adenocarcinoma (mGEA). However, this can be challenging if tumor tissue amount is limited and may cause delays in the initiation of chemotherapy. Therefore, we assessed the feasibility of multiple biomarkers testing in routine clinical practice.

Materials and methods

We reviewed the medical records of treatment-naïve patients with mGEA who underwent prospective multiple biomarkers testing between April 2022 and October 2023 in our institution. Eight biomarker status including HER2, MMR, PD-L1, CLDN18.2, Epstein–Barr virus, fibroblast growth factor receptor 2, epidermal growth factor receptor and mesenchymal epithelial transition expressions were simultaneously examined using biopsy or surgical specimens.

Results

A total of 203 patients with mGEA were analyzed. Most patients underwent gastroendoscopy and tumor biopsy shortly after referral to our institution. Biomarkers testing was successfully completed on the first attempt in 198 patients (97.5%). With additional steps including additional biopsy or asking remaining tumor samples from the referring hospital, the biomarker results were eventually available in all cases. The median turnaround time from sample collection to reporting results was 7 days. Consequently, 178 patients (87.7%) could receive first-line treatment after obtaining the biomarker status.

Conclusions

Multiple biomarkers testing for patients with mGEA was feasible in clinical practice. Establishment of a testing infrastructure based on the collaboration with multiple departments is essential for implementing biomarker-driven precision treatment.

背景评估多种生物标志物,包括人表皮生长因子受体 2(HER2)、错配修复(MMR)状态、程序性死亡配体 1(PD-L1)和克劳丁 18.2(CLDN18.2),对于选择合适的转移性胃食管腺癌(mGEA)一线疗法至关重要。然而,如果肿瘤组织数量有限,这可能具有挑战性,并可能导致化疗延迟开始。因此,我们评估了多种生物标记物检测在常规临床实践中的可行性。材料与方法我们回顾了2022年4月至2023年10月期间在我院接受前瞻性多种生物标记物检测的治疗无效的mGEA患者的病历。使用活检或手术标本同时检测了包括HER2、MMR、PD-L1、CLDN18.2、Epstein-Barr病毒、成纤维细胞生长因子受体2、表皮生长因子受体和间质上皮转化表达在内的八种生物标记物状态。大多数患者在转诊至我院后不久即接受了胃内镜检查和肿瘤活检。198名患者(97.5%)首次尝试就成功完成了生物标记物检测。通过额外的步骤,包括额外的活检或向转诊医院索要剩余的肿瘤样本,最终所有病例都获得了生物标记物结果。从样本采集到报告结果的中位周转时间为 7 天。结论对 mGEA 患者进行多种生物标记物检测在临床实践中是可行的。建立基于多部门合作的检测基础设施对于实施生物标志物驱动的精准治疗至关重要。
{"title":"Clinical implementation of simultaneous multiple biomarkers testing for metastatic or recurrent gastroesophageal adenocarcinoma: a single-institutional experience","authors":"","doi":"10.1016/j.esmogo.2024.100086","DOIUrl":"10.1016/j.esmogo.2024.100086","url":null,"abstract":"<div><h3>Background</h3><p>Evaluating multiple biomarkers including human epidermal growth factor receptor 2 (HER2), mismatch repair (MMR) status, programed death-ligand 1 (PD-L1), and claudin 18.2 (CLDN18.2) is essential for selecting appropriate first-line therapy of metastatic gastroesophageal adenocarcinoma (mGEA). However, this can be challenging if tumor tissue amount is limited and may cause delays in the initiation of chemotherapy. Therefore, we assessed the feasibility of multiple biomarkers testing in routine clinical practice.</p></div><div><h3>Materials and methods</h3><p>We reviewed the medical records of treatment-naïve patients with mGEA who underwent prospective multiple biomarkers testing between April 2022 and October 2023 in our institution. Eight biomarker status including HER2, MMR, PD-L1, CLDN18.2, Epstein–Barr virus, fibroblast growth factor receptor 2, epidermal growth factor receptor and mesenchymal epithelial transition expressions were simultaneously examined using biopsy or surgical specimens.</p></div><div><h3>Results</h3><p>A total of 203 patients with mGEA were analyzed. Most patients underwent gastroendoscopy and tumor biopsy shortly after referral to our institution. Biomarkers testing was successfully completed on the first attempt in 198 patients (97.5%). With additional steps including additional biopsy or asking remaining tumor samples from the referring hospital, the biomarker results were eventually available in all cases. The median turnaround time from sample collection to reporting results was 7 days. Consequently, 178 patients (87.7%) could receive first-line treatment after obtaining the biomarker status.</p></div><div><h3>Conclusions</h3><p>Multiple biomarkers testing for patients with mGEA was feasible in clinical practice. Establishment of a testing infrastructure based on the collaboration with multiple departments is essential for implementing biomarker-driven precision treatment.</p></div>","PeriodicalId":100490,"journal":{"name":"ESMO Gastrointestinal Oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949819824000475/pdfft?md5=a23d806cb63844b639f8cda46e54b777&pid=1-s2.0-S2949819824000475-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141961732","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
ESMO Gastrointestinal 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