molecular oncology

  • 文章类型: Journal Article
    Sacituzumabgovitecan(品牌名称:TRODELVY®)是一种正在研究的膀胱癌患者的新疗法,叫做尿路上皮癌,已经进展到局部晚期或转移阶段。局部晚期和转移性尿路上皮癌通常用铂类化疗治疗。转移性尿路上皮癌也用免疫检查点抑制剂治疗。对于接受这些治疗后癌症恶化的人,几乎没有治疗选择。Sacituzumabgovitecan是大多数尿路上皮癌患者的合适治疗选择,因为它旨在直接向癌症提供抗癌药物,以限制对健康细胞的潜在有害副作用。这是一项名为TROPHY-U-01的临床研究的摘要,重点是第一组参与者,称为队列1。队列1中的所有参与者都接受了sacituzumabgovitecan。
    所有参与者都曾接受过转移性尿路上皮癌的治疗,包括铂类化疗和检查点抑制剂。113名参与者中有31名的肿瘤在sacituzumabgovitecan治疗后变得明显变小或无法在扫描中看到;效果持续了7.2个月的中位数。一半的参与者在开始治疗后5.4个月仍然活着,没有他们的肿瘤变得更大或进一步扩散。无论肿瘤大小如何变化,其中一半在开始治疗后10.9个月仍然存活。大多数参与者都有副作用。这些副作用包括某些类型的血细胞水平降低,有时发烧,和松散或水样的粪便(腹泻)。副作用导致113名参与者中有7名停止服用sacituzumabgovitecan。
    研究表明,sacituzumabgovitecan具有显着的抗癌活性。尽管大多数接受sacituzumabgovitecan的参与者都有副作用,这些通常并不能阻止参与者继续服用sacituzumabgovitecan.医生可以使用治疗指南帮助控制这些副作用,但是这些副作用可能很严重。临床试验注册:NCT03547973(ClinicalTrials.gov)(TROPHY-U-1)。
    UNASSIGNED: Sacituzumab govitecan (brand name: TRODELVY®) is a new treatment being studied for people with a type of bladder cancer, called urothelial cancer, that has progressed to a locally advanced or metastatic stage. Locally advanced and metastatic urothelial cancer are usually treated with platinum-based chemotherapy. Metastatic urothelial cancer is also treated with immune checkpoint inhibitors. There are few treatment options for people whose cancer gets worse after receiving these treatments. Sacituzumab govitecan is a suitable treatment option for most people with urothelial cancer because it aims to deliver an anti-cancer drug directly to the cancer in an attempt to limit the potential harmful side effects on healthy cells. This is a summary of a clinical study called TROPHY-U-01, focusing on the first group of participants, referred to as Cohort 1. All participants in Cohort 1 received sacituzumab govitecan.
    UNASSIGNED: All participants received previous treatments for their metastatic urothelial cancer, including a platinum-based chemotherapy and a checkpoint inhibitor. The tumor in 31 of 113 participants became significantly smaller or could not be seen on scans after sacituzumab govitecan treatment; an effect that lasted for a median of 7.2 months. Half of the participants were still alive 5.4 months after starting treatment, without their tumor getting bigger or spreading further. Half of them were still alive 10.9 months after starting treatment regardless of tumor size changes. Most participants experienced side effects. These side effects included lower levels of certain types of blood cells, sometimes with a fever, and loose or watery stools (diarrhea). Side effects led 7 of 113 participants to stop taking sacituzumab govitecan.
    UNASSIGNED: The study showed that sacituzumab govitecan had significant anti-cancer activity. Though most participants who received sacituzumab govitecan experienced side effects, these did not usually stop participants from continuing sacituzumab govitecan. Doctors can help control these side effects using treatment guidelines, but these side effects can be serious.Clinical Trial Registration: NCT03547973 (ClinicalTrials.gov) (TROPHY-U-1).
    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
    儿童加速治愈和公平研究(RACE)法案规定,当分子靶标与儿科癌症相关时,新开发的靶向肿瘤药物在儿童中进行测试。第一年,RACEforChildren法案在为癌症儿童创造新药开发机会方面是有效的;然而,临床试验注册的重大障碍依然存在.儿科癌症临床试验受到物流挑战的影响,复杂性,和访问。因此,有可能采用网络化和集中的研究方法来解决这些障碍。在这里,我们讨论为成人调整及时的临床试验方法以服务于儿科肿瘤人群。通过创新的患者匹配解决方案,利用大型、具有高计算能力的真实世界数据集,Tempus儿童综合分子评估(TIME)计划旨在解决新疗法开发中的障碍。这篇评论探讨了在开发新的儿科疗法中减少挑战的潜力,推进基因组生物标志物测试的公平性,以实现精准定制治疗,并改善儿科肿瘤患者的预后。
    The Research to Accelerate Cures and Equity (RACE) for Children Act mandates that newly developed targeted oncology drugs be tested in children when molecular targets are relevant to pediatric cancers. In its first year, the RACE for Children Act was effective in creating novel drug development opportunities for children with cancer; however, significant barriers to clinical trial enrollment persist. Pediatric cancer clinical trials are impacted by challenges surrounding logistics, complexity, and access. As such, there is potential for a networked and centralized study approach to address these barriers. Here we discuss adapting a just-in-time clinical trial approach for adults to serve the pediatric oncology population. Through innovative patient matching solutions leveraging large, real-world datasets with high computational power, the Tempus Integrated Molecular Evaluation (TIME) for Kids Program aims to address barriers in the development of new therapies. This commentary explores the potential for reducing challenges in developing novel pediatric therapeutics, advancing equity in genomic biomarker testing for precision tailored treatment, and improving outcomes for pediatric oncology patients.
    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:肾上腺皮质癌(ACC)是一种罕见的侵袭性恶性肿瘤,具有异质性的临床结局。最近的研究提出了临床/组织病理学参数(S-GRAS评分)或分子生物标志物(BM)的组合来改善预后。我们通过评估其对S-GRAS评分的附加预后价值,对基于DNA的BMS进行了比较分析。
    方法:194福尔马林固定,分析石蜡包埋(FFPE)ACC样品,包括回顾性训练队列(n=107)和前瞻性验证队列(n=87).靶向DNA测序和焦磷酸测序用于检测ACC特异性基因的体细胞单核苷酸变异和PAX5启动子区域的甲基化。ENSAT肿瘤分期,年龄,出现时的症状,切除状态,和Ki-67合并计算S-GRAS。终点是整体(OS),无进展(PFS),无病生存率(DFS)。通过多变量生存分析评估预后作用,并通过HarrellC指数比较其表现。
    结果:在培训队列中,在多变量分析中证实了两个基于DNA的BMs的独立预后作用:Wnt/β-catenin和Rb/p53通路的改变,和高甲基化的PAX5(PFS和DFS均P<0.05,HR1.47-2.33)。将这些组合到S-GRAS以获得组合评分(COMBI)。在比较分析中,最佳判别预后模型是两个队列中所有终点的COMBI评分,其次是S-GRAS评分(C指数分别为OS0.724和0.765,PFS0.717和0.670,DFS0.699和0.644).
    结论:在常规可用的FFPE样本上评估的基于靶向DNA的BM改善了ACC的预后,超出了常规可用的临床和组织病理学参数。这种方法可能有助于更好地个性化患者的管理。
    OBJECTIVE: Adrenocortical carcinoma (ACC) is a rare aggressive malignancy with heterogeneous clinical outcomes. Recent studies proposed a combination of clinical/histopathological parameters (S-GRAS score) or molecular biomarkers (BMs) to improve prognostication. We performed a comparative analysis of DNA-based BMs by evaluating their added prognostic value to the S-GRAS score.
    METHODS: A total of 194 formalin-fixed, paraffin-embedded (FFPE) ACC samples were analysed, including a retrospective training cohort (n = 107) and a prospective validation cohort (n = 87). Targeted DNA sequencing and pyrosequencing were used to detect somatic single-nucleotide variations in ACC-specific genes and methylation in the promoter region of paired box 5 (PAX5). The European Network for the Study of Adrenocortical Tumors (ENSAT) tumour stage, age, symptoms at presentation, resection status, and Ki-67 were combined to calculate S-GRAS. Endpoints were overall (OS), progression-free (PFS), and disease-free survival (DFS). Prognostic role was evaluated by multivariable survival analysis and their performance compared by Harrell\'s concordance index (C index).
    RESULTS: In training cohort, an independent prognostic role was confirmed at multivariate analysis for two DNA-based BMs: alterations in Wnt/β-catenin and Rb/p53 pathways and hypermethylated PAX5 (both P< .05 for PFS and DFS, hazard ratio [HR] 1.47-2.33). These were combined to S-GRAS to obtain a combined (COMBI) score. At comparative analysis, the best discriminative prognostic model was COMBI score in both cohorts for all endpoints, followed by S-GRAS score (C index for OS 0.724 and 0.765, PFS 0.717 and 0.670, and DFS 0.699 and 0.644, respectively).
    CONCLUSIONS: Targeted DNA-based BM evaluated on routinely available FFPE samples improves prognostication of ACC beyond routinely available clinical and histopathological parameters. This approach may help to better individualise patient\'s management.
    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: Randomized Controlled Trial
    暂无摘要。
    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:表皮生长因子受体(EGFR)抑制可能对晚期胃食管腺癌(aGEA)患者的生物标志物选择人群有效。这里,我们测试了接受aGEA(NCT00824785)的化疗或化疗+抗EGFR单克隆抗体帕尼单抗的随机一线III期临床试验中纳入患者的治疗前组织中的EGFR拷贝数(CN)和无浆细胞DNA(cfDNA)与结局之间的相关性.
    方法:通过组织中的荧光原位杂交(n=114)或数字液滴PCR(n=250)和血浆cfDNA(n=354)可用于意向治疗(ITT)人群中的474(86%)患者。组织和血浆低通全基因组测序用于筛选受体酪氨酸激酶中的共扩增。在来自aGEA患者的患者来源的类器官(PDO)中模拟了化疗和EGFR抑制剂之间的相互作用。
    结果:cfDNA中的EGFR扩增与ITT群体中的低存活率相关,并且当通过治疗臂在组织和血浆中进行分析时观察到类似的趋势。EGFR抑制联合化疗与生存率改善无关,即使在EGFRCN显著增加的患者中。在PDO中的化疗剂表柔比星中添加抗EGFR抑制剂,导致细胞活力的矛盾增加和细胞周期的加速进展,与p21和细胞周期蛋白B1下调和细胞周期蛋白E1上调相关,选择性地在EGFR扩增的CGEA的类器官中。
    结论:EGFRCN可以在组织和液体活检中准确测量,可用于选择aGEA患者。EGFR抑制剂可能拮抗蒽环类抗生素的抗肿瘤作用,对未来组合试验的设计具有重要意义。
    OBJECTIVE: Epidermal growth factor receptor (EGFR) inhibition may be effective in biomarker-selected populations of advanced gastro-oesophageal adenocarcinoma (aGEA) patients. Here, we tested the association between outcome and EGFR copy number (CN) in pretreatment tissue and plasma cell-free DNA (cfDNA) of patients enrolled in a randomised first-line phase III clinical trial of chemotherapy or chemotherapy plus the anti-EGFR monoclonal antibody panitumumab in aGEA (NCT00824785).
    METHODS: EGFR CN by either fluorescence in situ hybridisation (n=114) or digital-droplet PCR in tissues (n=250) and plasma cfDNAs (n=354) was available for 474 (86%) patients in the intention-to-treat (ITT) population. Tissue and plasma low-pass whole-genome sequencing was used to screen for coamplifications in receptor tyrosine kinases. Interaction between chemotherapy and EGFR inhibitors was modelled in patient-derived organoids (PDOs) from aGEA patients.
    RESULTS: EGFR amplification in cfDNA correlated with poor survival in the ITT population and similar trends were observed when the analysis was conducted in tissue and plasma by treatment arm. EGFR inhibition in combination with chemotherapy did not correlate with improved survival, even in patients with significant EGFR CN gains. Addition of anti-EGFR inhibitors to the chemotherapy agent epirubicin in PDOs, resulted in a paradoxical increase in viability and accelerated progression through the cell cycle, associated with p21 and cyclin B1 downregulation and cyclin E1 upregulation, selectively in organoids from EGFR-amplified aGEA.
    CONCLUSIONS: EGFR CN can be accurately measured in tissue and liquid biopsies and may be used for the selection of aGEA patients. EGFR inhibitors may antagonise the antitumour effect of anthracyclines with important implications for the design of future combinatorial trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase III
    索拉非尼是晚期肝细胞癌(HCC)的标准全身疗法。早期HCC的切除/局部消融的生存益处受到70%的5年复发率的损害。比较索拉非尼与安慰剂作为辅助治疗的3期STORM试验未达到改善无复发生存率(RFS)的主要终点。生物标志物伴随研究BIOSTORM旨在定义(A)索拉非尼预防复发的预测因子和(B)具有B级证据的预后因素。
    收集了在STORM试验中随机接受索拉非尼(83)或安慰剂(105)的188名患者的肿瘤组织。分析包括基因表达谱分析,靶向外显子组测序(19个已知的致癌因子),免疫组织化学(pERK,pVEGFR2,Ki67),荧光原位杂交(VEGFA)和免疫组。在索拉非尼治疗的患者中产生捕获改善的RFS的基因签名。所有70例RFS事件均为复发,因此,复发时间等于RFS。使用Cox回归模型和交互检验评估预测和预后价值。
    BIOSTORM概括了STORM的临床病理特征。没有测试的生物标志物(与血管生成和增殖有关)或先前提出的基因特征,或突变预测索拉非尼获益或复发。新产生的146个基因标签鉴定了30%的患者在RFS方面获得了索拉非尼的益处(相互作用的p=0.04)。这些索拉非尼RFS应答者在CD4+T中显著富集,B和溶细胞性自然杀伤细胞,缺乏激活的适应性免疫成分。肝细胞pERK(HR=2.41;p=0.012)和微血管侵犯(HR=2.09;p=0.017)是独立的预后因素。
    在生物风暴中,只有肝细胞pERK和微血管浸润预测RFS较差。没有突变,基因扩增或先前提出的基因特征预测索拉非尼的益处。与索拉非尼的改进RFS相关的新生成的多基因签名值得进一步验证。
    NCT00692770。
    Sorafenib is the standard systemic therapy for advanced hepatocellular carcinoma (HCC). Survival benefits of resection/local ablation for early HCC are compromised by 70% 5-year recurrence rates. The phase 3 STORM trial comparing sorafenib with placebo as adjuvant treatment did not achieve its primary endpoint of improving recurrence-free survival (RFS). The biomarker companion study BIOSTORM aims to define (A) predictors of recurrence prevention with sorafenib and (B) prognostic factors with B level of evidence.
    Tumour tissue from 188 patients randomised to receive sorafenib (83) or placebo (105) in the STORM trial was collected. Analyses included gene expression profiling, targeted exome sequencing (19 known oncodrivers), immunohistochemistry (pERK, pVEGFR2, Ki67), fluorescence in situ hybridisation (VEGFA) and immunome. A gene signature capturing improved RFS in sorafenib-treated patients was generated. All 70 RFS events were recurrences, thus time to recurrence equalled RFS. Predictive and prognostic value was assessed using Cox regression models and interaction test.
    BIOSTORM recapitulates clinicopathological characteristics of STORM. None of the biomarkers tested (related to angiogenesis and proliferation) or previously proposed gene signatures, or mutations predicted sorafenib benefit or recurrence. A newly generated 146-gene signature identifying 30% of patients captured benefit to sorafenib in terms of RFS (p of interaction=0.04). These sorafenib RFS responders were significantly enriched in CD4+ T, B and cytolytic natural killer cells, and lacked activated adaptive immune components. Hepatocytic pERK (HR=2.41; p=0.012) and microvascular invasion (HR=2.09; p=0.017) were independent prognostic factors.
    In BIOSTORM, only hepatocytic pERK and microvascular invasion predicted poor RFS. No mutation, gene amplification or previously proposed gene signatures predicted sorafenib benefit. A newly generated multigene signature associated with improved RFS on sorafenib warrants further validation.
    NCT00692770.
    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
    For patients with locally advanced rectal cancer (LARC), adjuvant chemotherapy selection following surgery remains a major clinical dilemma. Here, we investigated the ability of circulating tumour DNA (ctDNA) to improve risk stratification in patients with LARC.
    We enrolled patients with LARC (T3/T4 and/or N+) planned for neoadjuvant chemoradiotherapy. Plasma samples were collected pretreatment, postchemoradiotherapy and 4-10 weeks after surgery. Somatic mutations in individual patient\'s tumour were identified via massively parallel sequencing of 15 genes commonly mutated in colorectal cancer. We then designed personalised assays to quantify ctDNA in plasma samples. Patients received adjuvant therapy at clinician discretion, blinded to the ctDNA results.
    We analysed 462 serial plasma samples from 159 patients. ctDNA was detectable in 77%, 8.3% and 12% of pretreatment, postchemoradiotherapy and postsurgery plasma samples. Significantly worse recurrence-free survival was seen if ctDNA was detectable after chemoradiotherapy (HR 6.6; P<0.001) or after surgery (HR 13.0; P<0.001). The estimated 3-year recurrence-free survival was 33% for the postoperative ctDNA-positive patients and 87% for the postoperative ctDNA-negative patients. Postoperative ctDNA detection was predictive of recurrence irrespective of adjuvant chemotherapy use (chemotherapy: HR 10.0; P<0.001; without chemotherapy: HR 22.0; P<0.001). Postoperative ctDNA status remained an independent predictor of recurrence-free survival after adjusting for known clinicopathological risk factors (HR 6.0; P<0.001).
    Postoperative ctDNA analysis stratifies patients with LARC into subsets that are either at very high or at low risk of recurrence, independent of conventional clinicopathological risk factors. ctDNA analysis could potentially be used to guide patient selection for adjuvant 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: Comparative Study
    Despite its successful use in academic research, next-generation sequencing (NGS) still represents many challenges for routine clinical adoption due to its inherent complexity and specialised expertise typically required to set-up, test and operate a complete workflow.This study aims to evaluate QIAGEN\'s newly launched GeneReader NGS System solution in a pathology laboratory setting by assessing the system\'s ease of use, sequencing accuracy and data reproducibility. Our laboratory was able to implement the system and validate its performance using clinical samples in direct comparison to an approved Sanger sequencing platform and to an alternative in-house NGS technology. The QIAGEN workflow focuses on clinically actionable hotspots maximising testing efficiency. Combined with automated upstream sample processing and integrated downstream bioinformatics, it offers a realistic solution for pathology laboratories with limited prior experience in NGS technology.
    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
    OBJECTIVE: The biological heterogeneity of hepatocellular carcinoma (HCC) makes prognosis difficult. We translate the results of a genome-wide high-throughput analysis into a tool that accurately predicts at presentation tumour growth and survival of patients with HCC.
    METHODS: Ultrasound surveillance identified HCC in 78 (training set) and 54 (validation set) consecutive patients with cirrhosis. Patients underwent two CT scans 6 weeks apart (no treatment in-between) to determine tumour volumes (V0 and V1) and calculate HCC doubling time. Baseline-paired HCC and surrounding tissue biopsies for microarray study (Agilent Whole Human Genome Oligo Microarrays) were also obtained. Predictors of survival were assessed by multivariate Cox model.
    RESULTS: Calculated tumour doubling times ranged from 30 to 621 days (mean, 107±91 days; median, 83 days) and were divided into quartiles: ≤53 days (n=19), 54-82 days (n=20), 83-110 days (n=20) and ≥111 days (n=19). Median survival according to doubling time was significantly lower for the first quartile versus the others (11 vs 41 months, 42, and 47 months, respectively) (p<0.0001). A five-gene transcriptomic hepatic signature including angiopoietin-2 (ANGPT2), delta-like ligand 4 (DLL4), neuropilin (NRP)/tolloid (TLL)-like 2 (NETO2), endothelial cell-specific molecule-1 (ESM1), and nuclear receptor subfamily 4, group A, member 1 (NR4A1) was found to accurately identify rapidly growing HCCs of the first quartile (ROC AUC: 0.961; 95% CI 0.919 to 1.000; p<0.0001) and to be an independent factor for mortality (HR: 3.987; 95% CI 1.941 to 8.193, p<0.0001).
    CONCLUSIONS: The hepatic five-gene signature was able to predict HCC growth in individual patient and the consequent risk of death. This implies a role of this molecular tool in the future therapeutic management of patients with HCC.
    BACKGROUND: ClinicalTrials.gov Identifier: NCT01657695.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号