molecular tumor board

分子肿瘤委员会
  • 文章类型: Journal Article
    通过液体活检监测ctDNA似乎代表了精确肿瘤学及其基础临床框架的完美匹配:分子肿瘤板(MTB)。检测和仔细检查靶向治疗或跟踪的成功,就此而言,用肿瘤的进化性质解决治疗是人们认为对MTB重要的一些主要进步。一个挑战是将通过分析ctDNA测序结果确定的每个已识别遗传改变的估计等位基因频率相关联,并将其与合适药物的范围相匹配。这可能会限制所有肿瘤变异的同时治疗。出现这种限制是因为通常需要新的活检来评估对治疗的反应。因此,评估MTB建议的成功依赖于传统的分期方法,突出了现有的诊断差距。因此,优化液体活检技术可以提高MTB治疗建议和随之而来的定制疗法的疗效.在这里,我们讨论了ctDNA分析在分子肿瘤委员会中的前景。
    Monitoring ctDNA by liquid biopsies seems to represent the perfect match for precision oncology and its cornerstone clinical framework: the molecular tumour board (MTB). Detecting and scrutinising the success of targeted therapies or tracking and, for that matter, addressing the therapy with the evolutive nature of a tumour are some of the main advancements one considers to be important for the MTB. One challenge is correlating the estimated allele frequency of each identified genetic alteration determined by analysing the ctDNA sequencing results and matching these with the range of suitable drugs, which may limit the simultaneous treatment of all tumour variations. This limitation arises because a new biopsy would typically be required to evaluate the response to treatment. As a result, evaluating the success of MTB recommendations relies on traditional staging methods, highlighting an existing diagnostic gap. Thus, optimising liquid biopsy technology could enhance the efficacy of MTB treatment recommendations and ensuing tailored therapies. Herein, we discuss the prospect of ctDNA analyses in the molecular tumour board.
    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
    背景:自2017年以来,下一代测序(NGS)已被引入许多韩国机构,以支持癌症的分子诊断,当时它有资格获得国民健康保险服务的报销。然而,基于NGS结果的分子指导治疗(MGT)的吸收受到限制,因为有关批准适应症之外的处方的严格规定,缺乏临床试验机会,以及大多数机构对分子肿瘤委员会(MTB)的访问权限有限。KOSMOS-II研究旨在证明MGT的可行性和有效性,由MTB通知,使用全国精准医疗平台。
    方法:KOSMOS-II试验是一项大规模的全国性主观察性研究。它涉及一个框架,用于根据局部NGS测试筛选转移性实体瘤患者的可行遗传改变。它建议MGT通过每两周举行一次远程和集中的MTB会议。MGT可以包括以下选项之一:Tier1,针对ALK等遗传改变的研究药物的治疗用途,EGFR,ERBB2,BRAF,FH,ROS1和RET,或具有高肿瘤突变负担的药物;第2级,包括具有批准适应症的药物或韩国健康保险审查和评估服务批准的适应症之外的允许治疗的药物;第3级,涉及与MTB推荐的遗传改变相匹配的临床试验。考虑到接受MGT的患者的预期比例在50%±3.25%的范围内,这项研究旨在招募1,000名患者.患者必须已经进展到一个或多个治疗线并且在登记之前经历NGS。
    结论:这个实用的主协议为罕见的遗传改变和高质量的真实世界数据提供了一个大规模筛查平台。附属临床试验,翻译研究,和临床基因组数据库将有助于为药物重新定位和开发新的生物标志物提供证据。
    背景:NCT05525858。
    BACKGROUND: Next-generation sequencing (NGS) has been introduced to many Korean institutions to support molecular diagnostics in cancer since 2017, when it became eligible for reimbursement by the National Health Insurance Service. However, the uptake of molecularly guided treatment (MGT) based on NGS results has been limited because of stringent regulations regarding prescriptions outside of approved indications, a lack of clinical trial opportunities, and limited access to molecular tumor boards (MTB) at most institutions. The KOSMOS-II study was designed to demonstrate the feasibility and effectiveness of MGT, informed by MTBs, using a nationwide precision medicine platform.
    METHODS: The KOSMOS-II trial is a large-scale nationwide master observational study. It involves a framework for screening patients with metastatic solid tumors for actionable genetic alterations based on local NGS testing. It recommends MGT through a remote and centralized MTB meeting held biweekly. MGT can include one of the following options: Tier 1, the therapeutic use of investigational drugs targeting genetic alterations such as ALK, EGFR, ERBB2, BRAF, FH, ROS1, and RET, or those with high tumor mutational burden; Tier 2, comprising drugs with approved indications or those permitted for treatment outside of the indications approved by the Health Insurance Review and Assessment Service of Korea; Tier 3, involving clinical trials matching the genetic alterations recommended by the MTB. Given the anticipated proportion of patients receiving MGT in the range of 50% ± 3.25%, this study aims to enroll 1,000 patients. Patients must have progressed to one or more lines of therapy and undergone NGS before enrollment.
    CONCLUSIONS: This pragmatic master protocol provides a mass-screening platform for rare genetic alterations and high-quality real-world data. Collateral clinical trials, translational studies, and clinico-genomic databases will contribute to generating evidence for drug repositioning and the development of new biomarkers.
    BACKGROUND: NCT05525858.
    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
    精准医学的新兴时代的特点是靶向抗癌疗法的可用性越来越高,并且并行开发技术以获得更精细的分子数据。他的解释可能并不总是直截了当的。分子肿瘤委员会聚集了各种专业人士,以便利用分子数据分析并为临床医生提供预后和预测性见解。除了医疗保健发展,它们也可以成为促进知识和研究传播的工具。越来越多的证据表明,分子肿瘤委员会在临床实践中的应用正在形成,积极的信号正在出现,虽然存在一定程度的异质性。这项工作分析了分子肿瘤委员会的潜在工作流程,涉及的数字,数据源,样本矩阵和符合条件的患者,以及现有的证据和学习的例子。新兴的多机构概念,疾病特异性分子肿瘤委员会也通过提供两个正在进行的全国经验来考虑.
    The emerging era of precision medicine is characterized by an increasing availability of targeted anticancer therapies and by the parallel development of techniques to obtain more refined molecular data, whose interpretation may not always be straightforward. Molecular tumor boards gather various professional figures, in order to leverage the analysis of molecular data and provide prognostic and predictive insights for clinicians. In addition to healthcare development, they could also become a tool to promote knowledge and research spreading. A growing body of evidence on the application of molecular tumor boards to clinical practice is forming and positive signals are emerging, although a certain degree of heterogeneity exists. This work analyzes molecular tumor boards\' potential workflows, figures involved, data sources, sample matrices and eligible patients, as well as available evidence and learning examples. The emerging concept of multi-institutional, disease-specific molecular tumor boards is also considered by presenting two ongoing nationwide experiences.
    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:综合分子肿瘤谱在癌症患者的治疗中被广泛使用。分子肿瘤委员会根据测试结果设计治疗策略。在此设置中,跨部门分子肿瘤委员会交流平台Deutschland(TEAM-D)旨在推动点对点交流,以连接该领域的专家。
    方法:在第一次虚拟TEAM-D会议期间,来自16所德国大学和5所非学术机构的参与者讨论了5例PIK3CA热点突变病例.此外,提出了一个说明性的案例插图。
    结果:总体而言,德国护理人员在施用标签外PIK3CA抑制剂方面表现出克制,并支持在这种情况下进行临床试验。
    结论:在精准肿瘤学的背景下,TEAM-D可在德国医疗保健系统的不同部门进行虚拟病例讨论。以PIK3CA热点突变为例,TEAM-D展示了整合来自不同医疗保健专业人员的知识的价值。
    BACKGROUND: Comprehensive molecular tumor profiling is widely used in the management of patients with cancer. Molecular tumor boards devise treatment strategies based on testing results. In this setting, the Transsectoral Molecular Tumor Board exchange platform Deutschland (TEAM-D) aims to drive peer-to-peer exchange to connect experts in the field.
    METHODS: During the first virtual TEAM-D meeting, participants from 16 German universities and 5 nonacademic institutions discussed five cases with PIK3CA hotspot mutations. Furthermore, an illustrative case vignette was presented.
    RESULTS: Overall, German caregivers show restraint in administering off-label PIK3CA inhibitor and favor clinical trials in this setting.
    CONCLUSIONS: In the setting of precision oncology, TEAM-D enables virtual case discussion across the different sectors of the German healthcare system. Based on the example of PIK3CA hotspot mutations, TEAM-D demonstrated the value of integrating knowledge from different healthcare professionals.
    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
    背景:通过实施分子肿瘤委员会(MTB),精确肿瘤治疗在乳腺和妇科肿瘤中的应用越来越普遍。但真实世界的临床结果数据仍然有限.方法:回顾性分析2018-2023年我中心收治的乳腺癌(BC)和妇科恶性肿瘤患者的MTB。分析涵盖了患者特征,下一代测序(NGS)结果,MTB建议,接受治疗,和临床结果。结果:63例患者(77.8%)有转移性疾病,44例(54.3%)患者之前接受过3行或更多行全身治疗.向50名患者(63.3%)提供个性化治疗建议,29人(36.7%)没有可行的目标。最终,23例患者(29.1%)接受了分子匹配治疗(MMT)。泛健身房肿瘤(BC和妇科恶性肿瘤)患者中常见的基因改变包括TP53(n=42/81,51.9%),PIK3CA(n=18/81,22.2%),BRCA1/2(n=10/81,12.3%),和ARID1A(n=9/81,11.1%)。接受MMT治疗的患者无进展生存期显着延长(中位PFS5.5vs.3.5个月,p=0.0014)。在所有接受分子谱分析的患者中,13.6%的患者通过精确肿瘤学获得了主要的临床益处(PFSr≥1.3和PR/SD≥6个月)。结论:NGS引导的精准肿瘤学在妇科和乳腺癌患者亚组中显示出改善的临床结果。
    Background: Precision oncology treatments are being applied more commonly in breast and gynecological oncology through the implementation of Molecular Tumor Boards (MTBs), but real-world clinical outcome data remain limited. Methods: A retrospective analysis was conducted in patients with breast cancer (BC) and gynecological malignancies referred to our center\'s MTB from 2018 to 2023. The analysis covered patient characteristics, next-generation sequencing (NGS) results, MTB recommendations, therapy received, and clinical outcomes. Results: Sixty-three patients (77.8%) had metastatic disease, and forty-four patients (54.3%) had previously undergone three or more lines of systemic treatment. Personalized treatment recommendations were provided to 50 patients (63.3%), while 29 (36.7%) had no actionable target. Ultimately, 23 patients (29.1%) underwent molecular-matched treatment (MMT). Commonly altered genes in patients with pan-gyn tumors (BC and gynecological malignancies) included TP53 (n = 42/81, 51.9%), PIK3CA (n = 18/81, 22.2%), BRCA1/2 (n = 10/81, 12.3%), and ARID1A (n = 9/81, 11.1%). Patients treated with MMT showed significantly prolonged progression-free survival (median PFS 5.5 vs. 3.5 months, p = 0.0014). Of all patients who underwent molecular profiling, 13.6% experienced a major clinical benefit (PFSr ≥ 1.3 and PR/SD ≥ 6 months) through precision oncology. Conclusions: NGS-guided precision oncology demonstrated improved clinical outcomes in a subgroup of patients with gynecological and breast cancers.
    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
    目的:个性化医疗带来了巨大的机遇和挑战。虽然治疗前景明显扩大,关于状态的描述,精确肿瘤学和分子肿瘤委员会(MTB)的临床实施和现实世界的好处仍然很少,特别是在泌尿生殖系统(GU)癌症领域。因此,本研究以泌尿系MTB病例为特征,以更好地了解MTB在泌尿系肿瘤学中的潜在作用.
    方法:我们分析了2019年1月至2022年10月在MTB审查的完整数据集的患者,重点是分子分析和治疗建议的结果。
    结果:我们评估了102例GU癌症患者,患者平均年龄为61.7岁。前列腺癌(PCa)是最常见的实体,占52.9%(54/102),其次是膀胱癌(18.6%,19/102)和肾细胞癌(14.7%,15/102)。平均而言,MTB的病例报告发生在初次诊断后54.9个月和之前2.7行治疗后.在研究期间49.0%(50/102)的患者死亡。68.6%(70/102)的患者获得了额外的基于MTB的治疗建议,其中64.3%(45/70)的患者推荐靶向治疗.只有6.7%(3/45)的患者-由于不同的原因-接受推荐的基于MTB的治疗艰难,33%(1/3)的患者达到疾病控制。在整个MTB研究期间,GU癌症病例报告和治疗建议增加,而初始治疗和最终治疗建议之间的时间间隔随着时间的推移而减少.
    结论:在MTB中介绍泌尿肿瘤患者是临床决策的非常有价值的措施。Prospective,在MTB中更早地介绍患者,以及在全面分子检测和靶向治疗批准方面不断变化的立法问题,可能会进一步改善患者从全面分子诊断中获益.
    BACKGROUND: Personalized medicine poses great opportunities and challenges. While the therapeutic landscape markedly expands, descriptions about status, clinical implementation and real-world benefits of precision oncology and molecular tumor boards (MTB) remain sparse, particularly in the field of genitourinary (GU) cancer. Hence, this study characterized urological MTB cases to better understand the potential role of MTB in uro-oncology.
    METHODS: We analyzed patients with complete data sets being reviewed at an MTB from January 2019 to October 2022, focusing on results of molecular analysis and treatment recommendations.
    RESULTS: We evaluated 102 patients with GU cancer with a mean patient age of 61.7 years. Prostate cancer (PCa) was the most frequent entity with 52.9% (54/102), followed by bladder cancer (18.6%, 19/102) and renal cell carcinoma (14.7%, 15/102). On average, case presentation at MTB took place 54.9 months after initial diagnosis and after 2.7 previous lines of therapy. During the study period, 49.0% (50/102) of patients deceased. Additional MTB-based treatment recommendations were achieved in a majority of 68.6% (70/102) of patients, with a recommendation for targeted therapy in 64.3% (45/70) of these patients. Only 6.7% (3/45) of patients - due to different reasons - received the recommended MTB-based therapy though, with 33% (1/3) of patients reaching disease control. Throughout the MTB study period, GU cancer case presentations and treatment recommendations increased, while the time interval between initial presentation and final therapy recommendation were decreasing over time.
    CONCLUSIONS: Presentation of uro-oncological patients at the MTB is a highly valuable measure for clinical decision-making. Prospectively, earlier presentation of patients at the MTB and changing legislative issues regarding comprehensive molecular testing and targeted treatment approval might further improve patients\' benefits from comprehensive molecular diagnostics.
    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
    背景:个性化药物为癌症治疗提供了靶向治疗选择。然而,是否将患者纳入下一代测序(NGS)检测的决策尚未标准化.这可能导致一些患者接受不必要的测试,而其他可能从中受益的患者则没有进行测试。通常,已经用尽常规治疗方案的患者在分子靶向治疗中值得考虑.协助临床医生决策,我们利用精准肿瘤学项目的常规数据开发了一种决策支持工具.
    方法:我们在临床数据上训练了一个机器学习模型,以确定是否应该对患者进行分子谱分析。要验证模型,将该模型的预测结果与分子肿瘤委员会(MTB)使用具有其特征的多个患者病例样本做出的决策进行比较.
    结果:预测模型包括440名分子谱分析患者和13,587名未经测试的患者。高曲线下面积(AUC)分数表明工程特征在决定分子谱分析中的重要性。患者年龄,身体状况,肿瘤类型,转移,和以前的治疗是最重要的特征。在验证过程中,MTB专家做出了相同的决定,仅在15例以前的病例中有10例推荐患者进行分子谱分析,但在15例病例中有9例专家与模型之间达成了共识。
    结论:基于历史队列,我们的预测模型有可能帮助临床医生决定是否进行分子谱分析.
    BACKGROUND: Personalized medicine offers targeted therapy options for cancer treatment. However, the decision whether to include a patient into next-generation sequencing (NGS) testing is not standardized. This may result in some patients receiving unnecessary testing while others who could benefit from it are not tested. Typically, patients who have exhausted conventional treatment options are of interest for consideration in molecularly targeted therapy. To assist clinicians in decision-making, we developed a decision support tool using routine data from a precision oncology program.
    METHODS: We trained a machine learning model on clinical data to determine whether molecular profiling should be performed for a patient. To validate the model, the model\'s predictions were compared with decisions made by a molecular tumor board (MTB) using multiple patient case vignettes with their characteristics.
    RESULTS: The prediction model included 440 patients with molecular profiling and 13,587 patients without testing. High area under the curve (AUC) scores indicated the importance of engineered features in deciding on molecular profiling. Patient age, physical condition, tumor type, metastases, and previous therapies were the most important features. During the validation MTB experts made the same decision of recommending a patient for molecular profiling only in 10 out of 15 of their previous cases but there was agreement between the experts and the model in 9 out of 15 cases.
    CONCLUSIONS: Based on a historical cohort, our predictive model has the potential to assist clinicians in deciding whether to perform molecular profiling.
    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
    背景:临床样本是不可替代的,他们转变为可搜索和可重复使用的数字生物库对于进行统计授权的回顾性和综合研究至关重要。目前,主要采用独立于数据的采集策略来全面数字化临床样本队列。然而,DIA的灵敏度是有限的,这就是为什么选择的标记候选物通常通过平行反应监测来额外测量的原因。
    方法:这里,我们将最近共同开发的hybrid-PRM/DIA技术作为一种新的智能数据采集策略应用,该策略允许在蛋白质型水平对罕见临床样本进行全面数字化.混合PRM/DIA通过多重并行反应监测(MSxPRM)的智能触发与使用DIA的临床生物样本的发现驱动数字化相结合,实现对当前临床感兴趣的特定分析物组的增强测量灵敏度。使用重标记的参考肽作为MSxPRM和内源性肽监测的触发物。
    结果:我们首先在临床环境中评估了来自64个注释人蛋白质组的185个选择的肿瘤相关抗原的蛋白型肽库的杂合PRM/DIA。我们证明了内源性肽检测的重复性和灵敏度的提高,即使在较低的浓度接近检测极限。显示多达179次MSxPRM扫描不会影响整体DIA性能。接下来,我们应用hybrid-PRM/DIA对生物学标记的黑色素瘤样本进行整合数字化,使用一组30种AQUA肽对28种生物标志物候选物进行整合,这与黑色素瘤患者的分子肿瘤委员会评估相关.在DIA检测到的大约6500个蛋白质组中,选定的候选标记,如UFO,可以使用MSxPRM扫描对CDK4、NF1和PMEL进行一致和定量的监测,为支持未来的临床决策提供额外的信心。
    结论:结合PRM和DIA测量为目前正在分子肿瘤委员会中讨论的患者的蛋白质标志物的灵敏和可重复检测提供了一种新策略,同时也有机会发现新的候选生物标志物。
    BACKGROUND: Clinical samples are irreplaceable, and their transformation into searchable and reusable digital biobanks is critical for conducting statistically empowered retrospective and integrative research studies. Currently, mainly data-independent acquisition strategies are employed to digitize clinical sample cohorts comprehensively. However, the sensitivity of DIA is limited, which is why selected marker candidates are often additionally measured targeted by parallel reaction monitoring.
    METHODS: Here, we applied the recently co-developed hybrid-PRM/DIA technology as a new intelligent data acquisition strategy that allows for the comprehensive digitization of rare clinical samples at the proteotype level. Hybrid-PRM/DIA enables enhanced measurement sensitivity for a specific set of analytes of current clinical interest by the intelligent triggering of multiplexed parallel reaction monitoring (MSxPRM) in combination with the discovery-driven digitization of the clinical biospecimen using DIA. Heavy-labeled reference peptides were utilized as triggers for MSxPRM and monitoring of endogenous peptides.
    RESULTS: We first evaluated hybrid-PRM/DIA in a clinical context on a pool of 185 selected proteotypic peptides for tumor-associated antigens derived from 64 annotated human protein groups. We demonstrated improved reproducibility and sensitivity for the detection of endogenous peptides, even at lower concentrations near the detection limit. Up to 179 MSxPRM scans were shown not to affect the overall DIA performance. Next, we applied hybrid-PRM/DIA for the integrated digitization of biobanked melanoma samples using a set of 30 AQUA peptides against 28 biomarker candidates with relevance in molecular tumor board evaluations of melanoma patients. Within the DIA-detected approximately 6500 protein groups, the selected marker candidates such as UFO, CDK4, NF1, and PMEL could be monitored consistently and quantitatively using MSxPRM scans, providing additional confidence for supporting future clinical decision-making.
    CONCLUSIONS: Combining PRM and DIA measurements provides a new strategy for the sensitive and reproducible detection of protein markers from patients currently being discussed in molecular tumor boards in combination with the opportunity to discover new biomarker candidates.
    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
    背景:目的是通过全外显子组和RNA测序(WES/RNA-Seq)确定晚期和难治性癌症患者的综合分子谱的附加值,这些患者没有基于更有限的靶向基因组(TGP)和基于阵列的比较基因组杂交(aCGH)的分子治疗建议(MBTR)。
    方法:在本回顾性分析中,我们选择了50例先前纳入PROFILER试验(NCT01774409)的患者,根据靶向的90基因组和aCGH,这些患者不推荐MBT.对于每个病人来说,对用于TGP/aCGH分析的反映FFPE样品的冷冻肿瘤样品进行WES和RNA-Seq处理。来自TGP/aCGH的数据进行了重新分析,以及WES/RNA-Seq,在一个新的分子肿瘤委员会(MTB)同时讨论了这些发现。
    结果:排除未知意义的变体后,在50例患者中,共发现167例体细胞分子改变(中位数:3[1-10]).在这167个相关的分子改变中,51(31%)是TGP/aCGH和WES/RNA-Seq共有的,19(11%)仅由TGP/aCGH鉴定,97(58%)仅由WES/RNA-Seq鉴定,包括两名患者的两个融合转录本。使用TGP/aCGH的信息在4/50(8%)患者中提供了MBTR,而使用WES/RNA-Seq发现的患者为9/50(18%)。三名患者基于TGP/aCGH和WES/RNA-Seq有类似的建议。
    结论:在TGP/aCGH不推荐MBTR的晚期和难治性癌症患者中,WES/RNA-Seq允许识别更多的改变,这些改变可能反过来,在有限的患者中,导致新的MBTR。
    BACKGROUND: The objective was to determine the added value of comprehensive molecular profile by whole-exome and RNA sequencing (WES/RNA-Seq) in advanced and refractory cancer patients who had no molecular-based treatment recommendation (MBTR) based on a more limited targeted gene panel (TGP) plus array-based comparative genomic hybridization (aCGH).
    METHODS: In this retrospective analysis, we selected 50 patients previously included in the PROFILER trial (NCT01774409) for which no MBT could be recommended based on a targeted 90-gene panel and aCGH. For each patient, the frozen tumor sample mirroring the FFPE sample used for TGP/aCGH analysis were processed for WES and RNA-Seq. Data from TGP/aCGH were reanalyzed, and together with WES/RNA-Seq, findings were simultaneously discussed at a new molecular tumor board (MTB).
    RESULTS: After exclusion of variants of unknown significance, a total of 167 somatic molecular alterations were identified in 50 patients (median: 3 [1-10]). Out of these 167 relevant molecular alterations, 51 (31%) were common to both TGP/aCGH and WES/RNA-Seq, 19 (11%) were identified by the TGP/aCGH only and 97 (58%) were identified by WES/RNA-Seq only, including two fusion transcripts in two patients. A MBTR was provided in 4/50 (8%) patients using the information from TGP/aCGH versus 9/50 (18%) patients using WES/RNA-Seq findings. Three patients had similar recommendations based on TGP/aCGH and WES/RNA-Seq.
    CONCLUSIONS: In advanced and refractory cancer patients in whom no MBTR was recommended from TGP/aCGH, WES/RNA-Seq allowed to identify more alterations which may in turn, in a limited fraction of patients, lead to new MBTR.
    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
    分子肿瘤诊断的进展已经改变了癌症治疗。然而,目前尚不清楚精准肿瘤学是否在所有恶性肿瘤中具有相同的影响和转化性质.我们对与人乳头瘤病毒(HPV)相关的妇科恶性肿瘤患者进行了回顾性分析,并在大学医院的跨学科分子肿瘤委员会(MTB)进行了全面的分子分析和随后的讨论。LMU慕尼黑,在11/2017和06/2022之间。我们确定了31例诊断为宫颈(CC)的患者的总队列,阴道或外阴癌。22例患者(分数:0.71)至少有一个突变。15名患者(0.48)具有可操作的突变,14名患者(0.45)接受了MTB内靶向治疗的建议。一名CC患者接受了MTB推荐的生物标志物指导治疗,并在mTOR抑制剂坦西罗莫司治疗8个月后病情稳定。导致其他患者不遵守MTB建议的因素包括患者知情拒绝,迅速恶化,疾病稳定,或使用替代性靶向但与生物标志物无关的治疗方法,例如抗体-药物缀合物或检查点抑制剂。尽管在我们的机构中,HPV相关妇科恶性肿瘤的可操作突变率显着,生物标志物指导的靶向治疗建议的立即实施仍然很低,MTB讨论后获得有针对性的治疗方案仍然是一个主要挑战。
    Advances in molecular tumor diagnostics have transformed cancer care. However, it remains unclear whether precision oncology has the same impact and transformative nature across all malignancies. We conducted a retrospective analysis of patients with human papillomavirus (HPV)-related gynecologic malignancies who underwent comprehensive molecular profiling and subsequent discussion at the interdisciplinary Molecular Tumor Board (MTB) of the University Hospital, LMU Munich, between 11/2017 and 06/2022. We identified a total cohort of 31 patients diagnosed with cervical (CC), vaginal or vulvar cancer. Twenty-two patients (fraction: 0.71) harbored at least one mutation. Fifteen patients (0.48) had an actionable mutation and fourteen (0.45) received a recommendation for a targeted treatment within the MTB. One CC patient received a biomarker-guided treatment recommended by the MTB and achieved stable disease on the mTOR inhibitor temsirolimus for eight months. Factors leading to non-adherence to MTB recommendations in other patient cases included informed patient refusal, rapid deterioration, stable disease, or use of alternative targeted but biomarker-agnostic treatments such as antibody-drug conjugates or checkpoint inhibitors. Despite a remarkable rate of actionable mutations in HPV-related gynecologic malignancies at our institution, immediate implementation of biomarker-guided targeted treatment recommendations remained low, and access to targeted treatment options after MTB discussion remained a major challenge.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号