gastroesophageal junction adenocarcinoma

胃食管结合部腺癌
  • 文章类型: Journal Article
    背景:一线zolbetuximab加化疗(SPOTLIGHT,mFOLFOX6;GLOW,CAPOX)显着改善了人表皮生长因子受体2阴性患者的无进展生存期(PFS)和总生存期(OS)与安慰剂加化疗相比,在III期SPOTLIGHT(NCT03504397)和GLOW(NCT03653507)研究中,肿瘤为claudin18亚型2阳性的局部晚期不可切除或转移性胃或胃食管交界腺癌.我们介绍了这些研究的患者报告结果(PRO)。
    方法:使用欧洲癌症研究和治疗组织癌症患者生活质量核心问卷(QLQ-C30)和食管-胃模块(QLQ-OG25)在完整分析集中测量健康相关生活质量(HRQoL)。全球痛苦,和5级EQ-5D(EQ-5D-5L)问卷。分析侧重于关键PRO领域:全球健康状况(GHS)/QoL,身体机能,腹痛和不适,恶心/呕吐。在SPOTLIGHT和GLOW以及个体研究中,评估了从基线和时间到首次明确恶化(TTDD)的最小二乘均值(LSM)变化。独立评估SPOTLIGHT和GLOW的确认恶化时间(TTCD)。
    结果:合并分析集包括1072例患者(佐贝昔单抗加化疗,537;安慰剂加化疗,535).治疗组之间的依从率相似。与安慰剂组相比,唑贝妥昔单抗在关键PRO域中从基线的LSM变化中观察到了类似的趋势。没有临床意义的恶化。在最初的几个唑贝妥昔单抗周期中,恶心/呕吐恶化,但后来恢复到基线水平。在两项研究中,两组之间的总体TTCD和TTDD结果相似。
    结论:SPOTLIGHT和GLOW患者在接受一线佐贝妥昔单抗联合化疗治疗时相对于基线维持测量的HRQoL。与安慰剂加化疗相比,唑贝妥昔单抗加化疗改善了关键PRO领域的PFS和OS,而对HRQoL无负面影响。
    BACKGROUND: First-line zolbetuximab plus chemotherapy (SPOTLIGHT, mFOLFOX6; GLOW, CAPOX) significantly improved progression-free survival (PFS) and overall survival (OS) versus placebo plus chemotherapy in patients with human epidermal growth factor receptor 2-negative, locally advanced unresectable or metastatic gastric or gastroesophageal junction adenocarcinoma whose tumors were claudin 18 isoform 2-positive in the phase III SPOTLIGHT (NCT03504397) and GLOW (NCT03653507) studies. We present patient-reported outcomes (PROs) from these studies.
    METHODS: Health-related quality of life (HRQoL) was measured in the full analysis sets using the European Organisation for Research and Treatment of Cancer Quality of Life of Cancer Patients Core Questionnaire (QLQ-C30) and Oesophago-Gastric Module (QLQ-OG25), Global Pain, and 5-level EQ-5D (EQ-5D-5L) questionnaires. Analyses focused on key PRO domains: global health status (GHS)/QoL, physical functioning, abdominal pain and discomfort, and nausea/vomiting. Least squares mean (LSM) changes from baseline and time to first definitive deterioration (TTDD) were evaluated combined across SPOTLIGHT and GLOW and for individual studies. Time to confirmed deterioration (TTCD) was evaluated independently for SPOTLIGHT and GLOW.
    RESULTS: The combined analysis set included 1072 patients (zolbetuximab plus chemotherapy, 537; placebo plus chemotherapy, 535). Compliance rates were similar between treatment arms. Similar trends were observed in the zolbetuximab versus placebo arms for LSM changes from baseline in key PRO domains, with no clinically meaningful deterioration. Nausea/vomiting worsened during the first few zolbetuximab cycles but later returned to baseline levels. Overall TTCD and TTDD results were similar between arms in both studies.
    CONCLUSIONS: Patients in SPOTLIGHT and GLOW maintained measured HRQoL relative to baseline when treated with first-line zolbetuximab added to chemotherapy. Zolbetuximab plus chemotherapy improved PFS and OS without negatively affecting HRQoL in key PRO domains compared with placebo plus chemotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    本研究旨在开发和验证生存预测模型和列线图,以预测接受抗程序性细胞死亡1受体(PD-1)治疗的晚期胃或胃食管交界(G/GEJ)腺癌患者的生存。该模型结合了免疫相关的不良事件(irAEs)以及常见的临床特征作为预测因子。
    收集了一个包含255名诊断为晚期G/GEJ腺癌的成年患者的数据集。识别并整合影响总生存期(OS)的irAE作为候选变量,以及其他12个候选变量。这些包括性别,年龄,东部肿瘤协作组表现状况(ECOGPS)评分,肿瘤分期,人表皮生长因子受体2(HER2)表达状态,存在腹膜和肝转移,抗PD-1治疗的年份和线,中性粒细胞与淋巴细胞比率(NLR),控制营养状况(CONUT)评分,和Charlson合并症指数(CCI)。为了减轻与ERP相关的时序偏差,采用了里程碑分析。使用最小绝对收缩和选择算子(LASSO)回归进行变量选择,以确定重要的预测因子,并应用方差膨胀因子来解决多重共线性问题。随后,利用前向似然比方法进行Cox回归分析,以建立生存预测模型,排除不满足比例风险(PH)假设的变量。该模型是使用整个数据集开发的,然后通过引导重新采样进行内部验证,并与另一家医院的队列进行外部验证。此外,创建列线图来描绘预测模型.
    将来自皮肤和内分泌系统的irAE整合为单一的保护性irAE类别并应用界标分析后,我们对预后预测模型进行了变量选择以及其他候选变量.最终的模型包括七个变量:ECOGPS得分,肿瘤分期,HER2在肿瘤组织中的表达状态,一线抗PD-1治疗,腹膜转移,CONUT得分,和保护性IRAE。模型的总体一致性指数为0.66。校准分析验证了模型在将预测结果与实际结果对齐方面的准确性。临床决策曲线分析表明,利用该模型进行治疗决策可以提高患者1年和2年生存率的净收益。
    这项研究通过整合irAEs和G/GEJ腺癌的共同临床特征,建立了预后预测模型。该模型具有良好的临床实用性,对晚期G/GEJ腺癌患者的总生存期具有准确的预测能力。
    UNASSIGNED: This study aimed to develop and validate a survival prediction model and nomogram to predict survival in patients with advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma undergoing treatment with anti-programmed cell death 1 receptor (PD-1). This model incorporates immune-related adverse events (irAEs) alongside common clinical characteristics as predictive factors.
    UNASSIGNED: A dataset comprising 255 adult patients diagnosed with advanced G/GEJ adenocarcinoma was assembled. The irAEs affecting overall survival (OS) to a significant degree were identified and integrated as a candidate variable, together with 12 other candidate variables. These included gender, age, Eastern cooperative oncology group performance status (ECOG PS) score, tumor stage, human epidermal growth factor receptor 2 (HER2) expression status, presence of peritoneal and liver metastases, year and line of anti-PD-1 treatment, neutrophil-to-lymphocyte ratio (NLR), controlling nutritional status (CONUT) score, and Charlson comorbidity index (CCI). To mitigate timing bias related to irAEs, landmark analysis was employed. Variable selection was performed using the least absolute shrinkage and selection operator (LASSO) regression to pinpoint significant predictors, and the variance inflation factor was applied to address multicollinearity. Subsequently, a Cox regression analysis utilizing the forward likelihood ratio method was conducted to develop a survival prediction model, excluding variables that failed to satisfy the proportional hazards (PH) assumption. The model was developed using the entire dataset, then internally validated through bootstrap resampling and externally validated with a cohort from another Hospital. Furthermore, a nomogram was created to delineate the predictive model.
    UNASSIGNED: After consolidating irAEs from the skin and endocrine systems into a single protective irAE category and applying landmark analysis, variable selection was conducted for the prognostic prediction model along with other candidate variables. The finalized model comprised seven variables: ECOG PS score, tumor stage, HER2 expression status in tumor tissue, first-line anti-PD-1 treatment, peritoneal metastasis, CONUT score, and protective irAE. The overall concordance index for the model was 0.66. Calibration analysis verified the model\'s accuracy in aligning predicted outcomes with actual results. Clinical decision curve analysis indicated that utilizing this model for treatment decisions could enhance the net benefit regarding 1- and 2-year survival rates for patients.
    UNASSIGNED: This study developed a prognostic prediction model by integrating common clinical characteristics of irAEs and G/GEJ adenocarcinoma. This model exhibits good clinical practicality and possesses accurate predictive ability for overall survival OS in patients with advanced G/GEJ adenocarcinoma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Systematic Review
    背景:免疫疗法联合化疗已被批准为不可切除的晚期胃癌(GC)的初始治疗策略。然而,在局部晚期可切除的胃或胃食管交界部腺癌(GC/GEJC)的围手术期化疗中加入免疫治疗的疗效仍不确定.因此,我们对随机对照试验(RCTs)进行了荟萃分析,比较了围手术期免疫检查点抑制剂(ICIs)加化疗与单纯化疗在局部晚期可切除GC/GEJC患者中的有效性.
    方法:对在线数据库进行了全面搜索,以确定直到2023年11月30日发布的RCT。计算主要结局的几率(OR)和95%置信区间(CI),包括R0切除率,D2淋巴结清扫术,病理完全缓解(pCR),和治疗相关不良事件(TRAEs)。
    结果:共纳入5份RCTs(6份报告)的2718例患者纳入分析。R0切除率和D2淋巴结清扫术的合并OR表明,与ICIs联合治疗与单独化疗相比没有显着差异。然而,ICIs的添加显著提高了pCR率(OR=3.43,95%CI2.61-4.50,p<0.0001)。在任何等级的TRAE和3-4等级的TRAE的发生率中没有观察到显著差异。然而,ICIs联合治疗与任何等级irAE的发生率显着升高相关(OR=4.03,95%CI:2.70-6.00,p<0.0001),以及3-4级IRAE(OR=4.51,95%CI:2.27-8.97,p<0.0001)。
    结论:本研究是首次荟萃分析,证明与化疗相比,局部晚期GC/GEJC患者围手术期联合治疗ICIs可产生更高的pCR率。
    BACKGROUND: Immunotherapy in combination with chemotherapy has been approved as an initial treatment strategy for unresectable advanced gastric cancer (GC). However, the efficacy of adding immunotherapy to perioperative chemotherapy in locally advanced resectable gastric or gastroesophageal junction adenocarcinoma (GC/GEJC) remains uncertain. Therefore, a meta-analysis of randomized controlled trials (RCTs) was performed to compare the effectiveness of perioperative immune checkpoint inhibitors (ICIs) plus chemotherapy versus chemotherapy alone in patients with locally advanced resectable GC/GEJC.
    METHODS: A comprehensive search of online databases was conducted to identify RCTs published until November 30, 2023. Odds ratios (ORs) with 95% confidence interval (CI) were calculated for primary outcomes, including R0 resection rate, D2 lymphadenectomy, pathologic complete response (pCR), and treatment-related adverse events (TRAEs).
    RESULTS: A total of 2718 patients from five RCTs (six reports) were included in the analysis. The pooled ORs of R0 resection rate and D2 lymphadenectomy demonstrated that combination therapy with ICIs showed no significant difference compared to chemotherapy alone. However, the addition of ICIs significantly improved pCR rates (OR = 3.43, 95 % CI 2.61-4.50, p < 0.0001). There were no significant differences observed in the incidence of any grade TRAEs and grade 3-4 TRAEs. However, ICIs combination therapy was associated with significantly higher incidences of any grade irAEs (OR = 4.03, 95 % CI: 2.70-6.00, p < 0.0001), as well as grade 3-4 irAEs (OR = 4.51, 95 % CI: 2.27-8.97, p < 0.0001).
    CONCLUSIONS: This study represents the first meta-analysis to demonstrate that perioperative combination therapy with ICIs yields superior pCR rates for patients with locally advanced GC/GEJC compared to chemotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    胃和胃食管交界处腺癌(GA/GEJA)与不良预后相关,主要是由于晚期疾病诊断。人表皮生长因子受体2(HER2)过表达和程序性死亡配体1(PD-L1)表达是局部晚期不可切除和转移性GA/GEJA治疗选择的重要生物标志物。人们对它们在疾病早期阶段的作用越来越感兴趣。在这项研究中,我们的目的是评估治疗性GA/GEJA队列中HER2和PD-L1的表达,以描述其表达模式,并分析HER2表达与临床病理特征之间的关联.在手术和内镜黏膜下剥离术肿瘤样本中评估HER2表达,在HER2阳性病例中评估PD-L1。临床队列包括107例患者,8.4%的HER2检测呈阳性(其中7人也表现出PD-L1联合阳性评分≥1分.HER2状态与生存结果无显著相关。病理学家指导,区域特异性分析显示,PD-L1表达很少与HER2阳性肿瘤区域重叠.虽然这些观察的治疗意义仍然未知,这些研究结果表明,靶向HER2和PD-L1的联合治疗策略可能针对不同的肿瘤亚克隆.本文公开的区域特异性生物标志物表达模式可能具有重要的治疗和预后影响。值得进一步评估。
    Gastric and gastroesophageal junction adenocarcinomas (GA/GEJA) are associated with a poor prognosis, primarily due to late disease diagnosis. Human Epidermal Growth Factor Receptor 2 (HER2) overexpression and programmed death-ligand 1 (PD-L1) expression are important biomarkers for treatment selection in locally advanced unresectable and metastatic GA/GEJA, and there is increasing interest in their role in earlier stages of disease. In this study, we aimed to evaluate HER2 and PD-L1 expression in a curative-intent GA/GEJA cohort to describe their expression patterns and analyze the association between HER2 expression and clinicopathological features. HER2 expression was evaluated in surgical and endoscopic submucosal dissection tumor samples, and PD-L1 was evaluated in HER2-positive cases. The clinical cohort included 107 patients, with 8.4% testing positive for HER2 (seven of whom also exhibited a PD-L1 combined positive score of ≥1. HER2 status was not significantly associated with survival outcomes. A pathologist-guided, region-specific analysis revealed that PD-L1 expression rarely overlaps with HER2-positive tumor areas. While the therapeutic implications of these observations remain unknown, these findings suggest that combination strategies targeting HER2 and PD-L1 might be directed toward distinct tumor subclones. The herein disclosed region-specific biomarker expression patterns may have important therapeutic and prognostic impacts, warranting further evaluation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胃食管癌在解剖和分子特征方面是高度多样化的肿瘤,使药物开发具有挑战性。在理解这些癌症的分子谱方面的最新进展导致了几种新的生物标志物的鉴定。正在进行的临床试验正在研究具有可喜结果的新靶向药物。CLDN18.2已成为具有相关靶向疗法的既定活性的生物标志物。其他目标特工,例如bemarituzumab和DKN-01,正在积极调查中。随着新药物被纳入治疗连续体,生物标志物重叠的问题,肿瘤异质性,和毒性管理将需要解决。
    Gastroesophageal cancers are highly diverse tumors in terms of their anatomic and molecular characteristics, making drug development challenging. Recent advancements in understanding the molecular profiles of these cancers have led to the identification of several new biomarkers. Ongoing clinical trials are investigating new targeted agents with promising results. CLDN18.2 has emerged as a biomarker with established activity of associated targeted therapies. Other targeted agents, such as bemarituzumab and DKN-01, are under active investigation. As new agents are incorporated into the treatment continuum, the questions of biomarker overlap, tumor heterogeneity, and toxicity management will need to be addressed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:基于生物标志物的治疗在不同类型的癌症中显示出改善的患者预后。这篇综述的目的是总结我们对食管胃腺癌(EGA)当前和未来生物标志物的认识。
    方法:在本出版物中,我们将回顾目前在上消化道肿瘤患者中的标准生物标志物.我们还将讨论正在研究中的新型生物标志物及其目前正在临床试验中的相关疗法。
    结果:EGAa是一组异质性疾病,在解剖学和分子上。有几种已确定的生物标志物(HER2,PD-L1,微卫星不稳定性或错配修复蛋白表达)允许对这些癌症患者进行个性化治疗。还有一些新兴的EGA生物标志物,其中一些有临床相关的治疗。Claudin18.2是其中最远的。抗claudin抗体,zolbetuximab,在两项3期研究中,生物标志物选择晚期GEA患者的总生存期得到改善.其他新型生物标志物,如FGFR2b和DKN01,也在验证过程中,基于这些生物标志物的治疗目前正在临床研究中。
    结论:在EGA中鉴定新的生物标志物的持续努力导致了上消化道肿瘤的亚分类增强。这些进步,加上靶向治疗和免疫疗法的战略应用,有希望进一步改善患者的预后。
    OBJECTIVE: Biomarker-based therapies have shown improved patient outcomes across various cancer types. The purpose of this review to summarize our knowledge of current and future biomarkers in esophagogastric adenocarcinoma (EGA).
    METHODS: In this publication, we will review current standard biomarkers in patients with upper GI cancers. We will also discuss novel biomarkers that are under investigations and their associated therapies that are currently in clinical trials.
    RESULTS: EGAa are a group of heterogeneous diseases, both anatomically and molecularly. There are several established biomarkers (HER2, PD-L1, microsattelite instability or mismatch repair protein expression) that allow for individualized treatments for patients with these cancers. There are also several emerging biomarkers for EGA, some of which have clinically relevant associated therapies. Claudin 18.2 is the furthest along among these. Anti-claudin antibody, zolbetuximab, improved overall survival in biomarker select patients with advanced GEA in two phase 3 studies. Other novel biomarkers, such as FGFR2b and DKN01, are also in the process of validation, and treatments based on the presence of these biomarkers are currently in clinical studies.
    CONCLUSIONS: Ongoing efforts to identify novel biomarkers in EGA have led to enhanced subclassification of upper GI cancers. These advances, coupled with the strategic application of targeted therapies and immunotherapy when appropriate, hold promise to further improve patients outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    卡博替尼被批准用于先前治疗的晚期肝细胞癌(aHCC),并已在胃癌(GC)和胃食管结合部腺癌(GEJ)中进行了研究。阿替珠单抗加贝伐单抗被批准用于不可切除或转移性HCC,未经事先全身治疗。我们评估了卡博替尼联合阿特珠单抗在先前未经全身抗癌治疗或先前治疗的GC/GEJ的aHCC中的疗效和安全性。
    COSMIC-021(ClinicalTrials.gov,NCT03170960)是一个开放标签,1b期实体瘤研究,剂量递增阶段,随后是肿瘤特异性扩增队列,包括aHCC(队列14)和GC/GEJ(队列15)。符合条件的患者年龄≥18岁,可测量局部晚期,转移性,或根据RECIST1.1版的复发性疾病。患者每天口服卡博替尼40mg,每3周一次静脉注射阿特珠单抗1200mg,直至疾病进展或不可接受的毒性。主要终点是研究者根据RECIST1.1版评估的客观缓解率。
    患者在2019年2月14日至2020年5月7日和61之间进行了筛查(30aHCC,31GC/GEJ)被登记并接受至少一个剂量的研究治疗。aHCC的中位随访时间为31.2个月(IQR28.5-32.7),GC/GEJ的中位随访时间为30.4个月(28.7-31.9)。aHCC的客观反应率为13%(4/30,95%CI4-31),GC/GEJ的客观反应率为0%(95%CI0-11)。六名(20%)aHCC患者和三名(10%)GC/GEJ患者有治疗相关的不良事件,导致停止任一研究药物。
    卡博替尼联合阿特珠单抗在先前未经全身抗癌治疗的肝癌中具有临床活性,安全性可控。在先前治疗的GC/GEJ中,卡博替尼联合阿特珠单抗的临床活性最小。
    Exelixis,Inc.,阿拉米达,CA,美国。
    UNASSIGNED: Cabozantinib is approved for previously treated advanced hepatocellular carcinoma (aHCC) and has been investigated in gastric cancer (GC) and gastroesophageal junction adenocarcinoma (GEJ). Atezolizumab plus bevacizumab is approved for unresectable or metastatic HCC untreated with prior systemic therapy. We evaluated efficacy and safety of cabozantinib plus atezolizumab in aHCC previously untreated with systemic anticancer therapy or previously treated GC/GEJ.
    UNASSIGNED: COSMIC-021 (ClinicalTrials.gov, NCT03170960) is an open-label, phase 1b study in solid tumours with a dose-escalation stage followed by tumour-specific expansion cohorts, including aHCC (cohort 14) and GC/GEJ (cohort 15). Eligible patients were aged ≥18 years with measurable locally advanced, metastatic, or recurrent disease per RECIST version 1.1. Patients received oral cabozantinib 40 mg daily and intravenous atezolizumab 1200 mg once every 3 weeks until progressive disease or unacceptable toxicity. The primary endpoint was investigator-assessed objective response rate per RECIST version 1.1.
    UNASSIGNED: Patients were screened between February 14, 2019, and May 7, 2020, and 61 (30 aHCC, 31 GC/GEJ) were enrolled and received at least one dose of study treatment. Median duration of follow-up was 31.2 months (IQR 28.5-32.7) for aHCC and 30.4 months (28.7-31.9) for GC/GEJ. Objective response rate was 13% (4/30, 95% CI 4-31) for aHCC and 0% (95% CI 0-11) for GC/GEJ. Six (20%) aHCC patients and three (10%) GC/GEJ patients had treatment-related adverse events resulting in discontinuation of either study drug.
    UNASSIGNED: Cabozantinib plus atezolizumab had clinical activity with a manageable safety profile in aHCC previously untreated with systemic anticancer therapy. Clinical activity of cabozantinib plus atezolizumab was minimal in previously treated GC/GEJ.
    UNASSIGNED: Exelixis, Inc., Alameda, CA, USA.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    As a result of the high approval dynamics and the growing number of immuno-oncological therapy concepts, the complexity of therapy decisions and control in the area of carcinomas of the esophagus, gastroesophageal junction and stomach is constantly increasing. Since the treatment indication for PD‑1 inhibitors that are currently approved in the European Union is often linked to the expression of PD-L1 (programmed cell death-ligand 1), the evaluation of tissue-based predictive markers by the pathologist is of crucial importance for treatment stratification. Even though the immunohistochemical analysis of the PD-L1 expression status is one of the best studied, therapy-relevant biomarkers for an immuno-oncological treatment, due to the high heterogeneity of carcinomas of the upper gastrointestinal tract, there are challenges in daily clinical diagnostic work with regard to implementation, standardization and interpretation of testing. An interdisciplinary group of experts from Germany has taken a position on relevant questions from daily pathological and clinical practice, which concern the starting material, quality-assured testing and the interpretation of pathological findings, and has developed recommendations for structured reporting.
    UNASSIGNED: Infolge der hohen Zulassungsdynamik sowie der wachsenden Anzahl an immunonkologischen Therapiekonzepten nimmt die Komplexität der Therapieentscheidung und -steuerung im Bereich der Karzinome des Ösophagus, gastroösophagealen Übergangs und Magens stetig zu. Da die Indikationsstellung bei den derzeit in der Europäischen Union zugelassenen PD-1-Inhibitoren häufig an die Expression von PD-L1 (Programmed Cell Death Ligand 1) gekoppelt ist, ist die Bestimmung dieses gewebebasierten prädiktiven Markers durch die Pathologie für die Stratifizierung der Behandlung von maßgeblicher Bedeutung. Auch wenn die immunhistochemische Bestimmung des PD-L1-Expressionsstatus zu den am besten untersuchten, therapierelevanten Biomarkern für eine immunonkologische Behandlung gehört, ergeben sich aufgrund der hohen Heterogenität der Karzinome des oberen Gastrointestinaltrakts im klinisch-diagnostischen Alltag Herausforderungen in Bezug auf die Implementierung, Standardisierung und Interpretation der Testung. Eine interdisziplinäre Expertengruppe aus Deutschland hat zu relevanten Fragen aus dem klinisch-pathologischen Alltag Stellung bezogen, die das Ausgangsmaterial, die qualitätsgesicherte Testung und die Befundinterpretation betreffen und Empfehlungen für eine strukturierte Befunderstellung erarbeitet.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    全球范围内,第五大最常见的癌症和第四大癌症相关死亡原因是胃癌(GC)。最近关于实体瘤的临床试验招募了具有药物遗传改变的患者,蛋白质表达,和免疫特性。在胃或胃食管交界处(GEJ)癌症中,曲妥珠单抗联合人表皮生长因子受体2(HER2)阳性患者的一线化疗和雷莫珠单抗联合二线紫杉醇与单纯化疗相比显著延长总生存期(OS),根据3期试验结果。最近,免疫检查点抑制剂(ICI)单药治疗被批准为三线或后期治疗.根据Checkmate649试验结果,在HER2阴性患者中,与单纯化疗相比,化疗加ICIs作为一线治疗可改善生存率。相反,全身化疗预后仍然较差。尽管某些患者在晚期GC中可能会对治疗产生持久反应并延长生存期。最近,一流的,靶向并结合claudin18同工型2(CLDN18.2)的嵌合免疫球蛋白G1单克隆抗体(zolbetuximab)已成为GC治疗中一种新的靶向疗法。全球Ⅲ期试验显示,在CLDN18.2阳性和HER2阴性GC患者中,在一线化疗中加入唑贝昔单抗可延长OS。这篇综述总结了CLDN18.2靶向治疗的最新临床试验。
    Globally, the fifth most common cancer and the fourth leading cause of cancer-related mortality is gastric cancer (GC). Recent clinical trials on solid tumors enrolled patients who possess druggable genetic alterations, protein expression, and immune characteristics. In gastric or gastroesophageal junction (GEJ) cancers, trastuzumab combined with first-line chemotherapy in human epidermal growth factor receptor 2 (HER2)-positive patients and ramucirumab combined with second-line paclitaxel remarkably prolonged overall survival (OS) compared with chemotherapy alone, according to phase 3 trial results. Recently, immune checkpoint inhibitor (ICI) monotherapy was approved as third- or later-line treatment. Chemotherapy plus ICIs as first-line treatment exhibited improved survival compared with chemotherapy alone in HER2-negative patients according to Checkmate 649 trial results. Conversely, systemic chemotherapy prognosis remains poor. although some patients may achieve durable response to treatment and prolonged survival in advanced GC. Recently, a first-in-class, chimeric immunoglobulin G1 monoclonal antibody (zolbetuximab) that targets and binds to claudin 18 isoform 2 (CLDN18.2) has emerged as a new target therapy in GC treatment. Global phase Ⅲ trials revealed that the addition of zolbetuximab to first-line chemotherapy prolonged OS in CLDN18.2-positive and HER2-negative GC patients. This review summarizes recent clinical trials of CLDN18.2-targeted therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    胃癌是全球第五大流行癌症,也是癌症相关死亡率的第三大原因。在相当比例的患者中,原发性胃肿瘤患者可以发生腹膜癌病,估计范围从14%到43%。对于来自胃癌的腹膜癌患者,治疗选择有限。腹膜内化疗,联合热疗(HIPEC),在腹膜腔内提供局部剂量增强,有效地靶向腹膜癌与最小的全身暴露。我们报告了一例胃食管连接部腺癌,并通过细胞减灭术和HIPEC成功治疗了腹膜转移,然后是手术和全身化疗,强调HIPEC是可行的选择之一,以及多式联运方法对处理此案的重要性。
    Gastric cancer is the fifth most prevalent cancer worldwide and the third leading cause of cancer- related mortality. Peritoneal carcinomatosis can develop in patients with a primary gastric tumor in a substantial proportion of patients, with estimates ranging from 14% to 43%. For patients with peritoneal carcinomatosis from gastric cancer, therapeutic options are limited. Intraperitoneal chemotherapy, combined with hyperthermia (HIPEC), provides regional dose intensification within the peritoneal cavity, effectively targeting peritoneal carcinomatosis with minimal systemic exposure. We report a case of gastroesophageal junction adenocarcinoma with peritoneal metastasis successfully treated with of cytoreductive surgery and HIPEC, and followed by surgery and systemic chemotherapy, highlighting HIPEC as one of the viable options and the importance of a multimodal approach for the treatment of this case.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号