Tumor-agnostic therapy

  • 文章类型: Journal Article
    综合基因组分析(CGP)有望通过确定参与致癌作用的基因来扩大癌症药物选择的范围。然而,少数患者可以在CGP后获得推荐的治疗。在这里,我们报告了一个病例,其中pemigatinib,选择性成纤维细胞生长因子受体(FGFR)抑制剂,被用作最后一线治疗,以治疗表现出FGFR2基因组改变的晚期胃癌患者,由CGP测试确定。患者(男性,52岁)被诊断为晚期胃癌(cIV期,cT4aN3M1[LYM],波尔,HER20,微卫星稳定),并接受多西他赛+顺铂+S-1(7个周期),伊立替康+雷莫珠单抗(11个周期),和nivolumab(3个周期),但经历了进行性疾病(PD)。随后,进行了一次液体CDx测试,揭示FGFR2重排和扩增;然而,目前尚无针对FGFR2改变的基因型匹配疗法的临床试验.在TAS-102的三个周期后,患者经历了PD,并同意在标签外使用培米加替尼。我院肿瘤基因组学医学委员会批准自费治疗。患者在治疗开始后CEA和CA19-9水平明显下降,但经历了五门课程后的PD。在治疗过程中,观察到1级高磷血症和甲癣。据我们所知,这是报道的首例Pemigatinib治疗出现FGFR2基因改变的晚期胃癌患者.该病例可以作为肿瘤不可知疗法的一个值得注意的例子,以扩大具有罕见遗传改变的胃癌患者的治疗选择。
    Comprehensive genome profiling (CGP) is expected to widen the scope of cancer drug options by identifying the genes involved in carcinogenesis. However, a few patients can access recommended treatments following CGP. Herein, we report a case in which pemigatinib, a selective fibroblast growth factor receptor (FGFR) inhibitor, was used as last-line therapy to treat a patient with advanced gastric cancer exhibiting FGFR2 genomic alterations, as determined by CGP testing. The patient (male, 52 years old) was diagnosed with advanced gastric cancer (cStage IV, cT4aN3M1 [LYM], por, HER2 0, microsatellite stable) and received docetaxel + cisplatin + S-1 (7 cycles), irinotecan + ramucirumab (11 cycles), and nivolumab (3 cycles), but experienced progressive disease (PD). Subsequently, FoundationOne Liquid CDx testing was conducted, revealing FGFR2 rearrangement and amplification; however, no clinical trials on genotype-matched therapies for FGFR2 alterations were available. After three cycles of TAS-102, the patient experienced PD and provided consent for the off-label use of pemigatinib. The Cancer Genomics Medical Committee of our hospital approved the self-funded treatment. The patient had markedly decreased CEA and CA19-9 levels after treatment initiation, but experienced PD after five courses. Over the treatment course, grade 1 hyperphosphatemia and onychomadesis were observed. To the best of our knowledge, this is the first reported case of pemigatinib therapy employed in a patient with advanced gastric cancer exhibiting FGFR2 gene alterations. This case could serve as a notable example of tumor-agnostic therapy to broaden treatment options for gastric cancer patients with rare genetic alterations.
    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
    Due to the considerable technological progress in molecular and genetic diagnostics as well as increasing insights into the molecular pathogenesis of diseases, there has been a fundamental paradigm shift in the past two decades from a \"one-size-fits-all approach\" to personalized, molecularly informed treatment strategies. Personalized medicine or precision medicine focuses on the genetic, physiological, molecular, and biochemical differences between individuals and considers their effects on the development, prevention, and treatment of diseases. As a pioneer of personalized medicine, the field of oncology is particularly noteworthy, where personalized diagnostics and treatment have led to lasting change in the treatment of cancer patients in recent years. In this article, the significant change towards personalized treatment concepts, especially in the field of personalized oncology, will be discussed and examined in more detail.
    UNASSIGNED: Aufgrund des erheblichen technologischen Fortschritts in der molekularen und genetischen Diagnostik sowie zunehmender Erkenntnisse über die molekulare Pathogenese von Krankheiten hat in den letzten zwei Jahrzehnten ein grundlegender Paradigmenwechsel stattgefunden – von einem „One-size-fits-all-Ansatz“ hin zu personalisierten, molekular informierten Therapiestrategien. Die personalisierte Medizin oder Präzisionsmedizin konzentriert sich auf die genetischen, physiologischen, molekularen und biochemischen Unterschiede zwischen Individuen und berücksichtigt deren Auswirkungen auf die Entwicklung, Prävention und Behandlung von Krankheiten. Als Vorreiter der personalisierten Medizin ist v. a. der Bereich der Onkologie zu nennen, wo personalisierte Diagnostik und Therapie die Behandlung von Krebspatient:innen in den letzten Jahren nachhaltig verändert haben. In diesem Artikel wird der signifikante Wandel hin zu personalisierten Therapiekonzepten, insbesondere im Bereich der personalisierten Onkologie, ausführlich diskutiert und näher beleuchtet.
    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:Pembrolizumab,抗癌免疫治疗剂,自2014年首次获得美国食品和药物管理局(FDA)批准以来,已获得多次批准。关于其实际使用的数据有限,并且在2017年肿瘤不可知批准后转移,用于治疗任何微卫星不稳定性高/错配修复缺陷(MSI-H/dMMR)实体瘤的患者。这项研究分析了美国老年人中pembrolizumab在肿瘤不可知批准之前和之后的使用,揭示其在肿瘤学实践中不断发展的作用。
    方法:使用监视,流行病学和最终结果(SEER)-医疗保险数据(2014-2019年),我们检查了pembrolizumab受者在肿瘤不可知批准前后的癌症部位.根据位点特异性批准的时间(肿瘤不可知批准之前/之后)或无位点特异性批准对癌症位点进行分类。并纳入MSI-H/dMMR临床试验。
    结果:pembrolizumab受者的总人数从不可知前的4221人增加到不可知后的20479人。Pembrolizumab用于广泛的癌症类型,包括在使用时没有FDA批准的位点特异性适应症的癌症(在诊断前25.8%,在诊断后24.6%).在肿瘤不可知批准之前,接受pembrolizumab治疗癌症的pembrolizumab接受者的比例从77.3%降至70.8%。接受pembrolizumab治疗的pembrolizumab患者在肿瘤不可知批准后获得位点特异性批准的癌症的比例几乎翻了一番(6.8%至13.0%)。在肿瘤不可知批准后,MSI-H/dMMR试验中包括的pembrolizumab受体的癌症比例也翻了一番(12.3%至25.5%)。
    结论:随着时间的推移,Pembrolizumab在患有癌症的老年人中的使用有所增加,超出了FDA批准的特定部位适应症。
    BACKGROUND: Pembrolizumab, an anticancer immunotherapy agent, has received multiple approvals since its first approval by the U.S. Food and Drug Administration (FDA) in 2014. Limited data exist on its real-world use and shifts post tumor-agnostic approval in 2017 for the treatment of patients with any microsatellite instability-high/mismatch repair deficient (MSI-H/dMMR) solid tumors. This study analyzes pembrolizumab\'s pre and post-tumor-agnostic approval use among older U.S. adults, revealing its evolving role in oncology practice.
    METHODS: Using the Surveillance, Epidemiology and End Results (SEER)-Medicare data (2014-2019), we examined the cancer sites of pembrolizumab recipients before and after tumor-agnostic approval. Cancer sites were classified based on the timing of site-specific approvals (before/after tumor-agnostic approval) or no site-specific approval, and inclusion in MSI-H/dMMR clinical trials.
    RESULTS: The total number of pembrolizumab recipients increased from 4221 in the pre-agnostic period to 20 479 in the post-agnostic period. Pembrolizumab was used for a broad range of cancer types, including cancers that had no FDA-approved site-specific indications at the time of use (25.8% in pre- and 24.6% in post-agnostic periods). The proportion of pembrolizumab recipients receiving pembrolizumab for cancers with site-specific approvals before tumor-agnostic approval decreased from 77.3% to 70.8%. The proportion of pembrolizumab recipients receiving pembrolizumab for cancers that gained site-specific approvals following tumor-agnostic approval almost doubled (6.8% to 13.0%). The proportion of pembrolizumab recipients with cancers included in MSI-H/dMMR trials also doubled (12.3% to 25.5%) following tumor-agnostic approval.
    CONCLUSIONS: Pembrolizumab use has expanded over time among older adults with cancer, extending beyond those with FDA-approved site-specific indications.
    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
    转染期间重排(RET)是受体蛋白酪氨酸激酶。致癌RET融合或突变最常见于非小细胞肺癌(NSCLC)和甲状腺癌,而且在各种类型的低发病率癌症中也越来越多。在过去的几年里,两种有效和选择性的RET蛋白酪氨酸激酶抑制剂(TKIs),普拉塞替尼(BLU-667)和selpercatinib(LOXO-292,LY3527723)被开发并获得监管部门批准。尽管普雷替尼和selpercatinib的总体反应率(ORR)很高,<10%的患者达到完全缓解(CR)。RETTKI耐受的残留肿瘤不可避免地通过次级靶突变产生耐药性,获得的替代癌基因,或MET扩增。位于激酶溶剂前沿位点的RETG810突变被确定为对selpercatinib和pralsetinib的获得性耐药的主要目标机制。能够抑制selpercatinib/pralsetinib抗性RET突变体的几种下一代RETTKIs已经进展到临床试验。然而,很可能出现新的TKI适应性RET突变,从而对这些下一代RETTKIs产生抗性.解决该问题需要更好地理解支持RETTKI耐受的持久性的多种机制,以确定脆弱的汇合点,以设计有效的共同治疗来消除残留肿瘤。
    The rearranged during transfection (RET) is a receptor protein tyrosine kinase. Oncogenic RET fusions or mutations are found most often in non-small cell lung cancer (NSCLC) and in thyroid cancer, but also increasingly in various types of cancers at low rates. In the last few years, two potent and selective RET protein tyrosine kinase inhibitors (TKIs), pralsetinib (BLU-667) and selpercatinib (LOXO-292, LY3527723) were developed and received regulatory approval. Although pralsetinib and selpercatinib gave high overall response rates (ORRs), < 10% of patients achieved a complete response (CR). The RET TKI-tolerated residual tumors inevitably develop resistance by secondary target mutations, acquired alternative oncogenes, or MET amplification. RET G810 mutations located at the kinase solvent front site were identified as the major on-target mechanism of acquired resistance to both selpercatinib and pralsetinib. Several next-generation of RET TKIs capable of inhibiting the selpercatinib/pralsetinib-resistant RET mutants have progressed to clinical trials. However, it is likely that new TKI-adapted RET mutations will emerge to cause resistance to these next-generation of RET TKIs. Solving the problem requires a better understanding of the multiple mechanisms that support the RET TKI-tolerated persisters to identify a converging point of vulnerability to devise an effective co-treatment to eliminate the residual tumors.
    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: Clinical Trial, Phase II
    背景:越来越多的试验表明,当靶向治疗与个体患者(pts)的药物基因组改变相匹配时,患有转移性疾病的癌症患者的治疗结果得到改善。估计30-80%的晚期实体瘤具有可操作的基因组改变。然而,个性化癌症治疗的疗效仍然很少在更大的研究,由于不同组织学肿瘤类型之间的可药物改变的低频率和异质性分布,因此进行了对照试验。因此,靶向癌症治疗对临床结局的总体影响仍需研究.
    方法:ProTarget是一个国家,非随机化,多种药物,开放标签,泛癌症2期试验旨在研究目前市售13种的抗肿瘤活性和毒性,EMA批准的靶向治疗晚期恶性疾病的标记适应症之外,具有特定的可操作的基因组改变。该试验涉及丹麦国家分子肿瘤委员会,以确认药物变体匹配。主要纳入标准包括a)可测量的疾病(RECISTv.1.1),b)ECOG性能状态0-2,和c)与研究药物之一匹配的可操作的基因组改变。关键排除标准包括a)所选药物的EMA批准标签中的癌症类型,和b)已知赋予药物抗性的基因组改变。初始药物剂量,时间表和剂量修改是根据EMA批准的标签。主要终点是16周时的客观反应或疾病稳定。将Pts分配给所选药物定义的队列,基因组改变,和肿瘤组织学类型。队列根据Simon的两阶段设计进行监控。前24周每8周评估一次反应,然后每12周。该试验的设计类似于荷兰DRUP和ASCOTAPUR试验,是北欧精准癌症医学试验网络的合作伙伴。在ProTarget中,连续新鲜肿瘤和液体活检是强制性的,并收集用于广泛的翻译研究,包括全基因组测序,阵列分析,和RNA测序。
    结论:ProTarget试验将为靶向治疗确定新的预测性生物标志物,并提供新的数据和有关分子途径的必要见解,例如,耐药机制,从而有可能发展和扩大个性化癌症治疗策略。
    方法:2022年9月16日。
    背景:ClinicalTrials.gov标识符:NCT04341181。二级识别号:ML41742。欧盟编号:2019-004771-40。
    BACKGROUND: An increasing number of trials indicate that treatment outcomes in cancer patients with metastatic disease are improved when targeted treatments are matched with druggable genomic alterations in individual patients (pts). An estimated 30-80% of advanced solid tumors harbor actionable genomic alterations. However, the efficacy of personalized cancer treatment is still scarcely investigated in larger, controlled trials due to the low frequency and heterogenous distribution of druggable alterations among different histologic tumor types. Therefore, the overall effect of targeted cancer treatment on clinical outcomes still needs investigation.
    METHODS: ProTarget is a national, non-randomized, multi-drug, open-label, pan-cancer phase 2 trial aiming to investigate the anti-tumor activity and toxicity of currently 13 commercially available, EMA-approved targeted therapies outside the labeled indication for treatment of advanced malignant diseases, harboring specific actionable genomic alterations. The trial involves the Danish National Molecular Tumor Board for confirmation of drug-variant matches. Key inclusion criteria include a) measurable disease (RECIST v.1.1), b) ECOG performance status 0-2, and c) an actionable genomic alteration matching one of the study drugs. Key exclusion criteria include a) cancer type within the EMA-approved label of the selected drug, and b) genomic alterations known to confer drug resistance. Initial drug dose, schedule and dose modifications are according to the EMA-approved label. The primary endpoint is objective response or stable disease at 16 weeks. Pts are assigned to cohorts defined by the selected drug, genomic alteration, and tumor histology type. Cohorts are monitored according to a Simon\'s two-stage-based design. Response is assessed every 8 weeks for the first 24 weeks, then every 12 weeks. The trial is designed similar to the Dutch DRUP and the ASCO TAPUR trials and is a partner in the Nordic Precision Cancer Medicine Trial Network. In ProTarget, serial fresh tumor and liquid biopsies are mandatory and collected for extensive translational research including whole genome sequencing, array analysis, and RNA sequencing.
    CONCLUSIONS: The ProTarget trial will identify new predictive biomarkers for targeted treatments and provide new data and essential insights in molecular pathways involved in e.g., resistance mechanisms and thereby potentially evolve and expand the personalized cancer treatment strategy.
    METHODS: 16, 09-MAY-2022.
    BACKGROUND: ClinicalTrials.gov Identifier: NCT04341181. Secondary Identifying No: ML41742. EudraCT No: 2019-004771-40.
    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:精准医学在癌症治疗中越来越重要。无论肿瘤实体如何,都使用与肿瘤无关的疗法,因为它们靶向肿瘤中的特定生物标志物。在德国,传统上,肿瘤药物的获益评估是针对特定实体的.因此,对肿瘤不可知疗法的评估给利益相关者带来了各种挑战.我们的目标是使用两步顺序定性方法系统地确定挑战和可能的解决方案,以评估肿瘤不可知适应症中治疗的益处。
    方法:为了确定相关挑战,我们对德国以前参与肿瘤无关治疗获益评估的不同利益相关方进行了定性访谈.为了确定这些挑战的可能解决方案,我们系统地搜索了MEDLINE,Embase和欧洲HTA机构的网站提供相关文献。
    结果:我们确定了九类挑战,其中以下几点被认为特别相关:缺乏直接的比较研究,关于使用篮子研究和间接比较的挑战,在确定与肿瘤无关的适应症中适当的比较治疗方面的挑战,以及系统方面的挑战。确定了七类解决方案,包括越来越多地使用真实世界的证据,在系统重新评估的背景下做出有条件的决定,拆分应用领域,并找到(新的)方法来设计和分析篮子研究。
    结论:一系列可能的解决方案,这有助于应对德国确定的挑战,已经找到了。未来的研究应该调查这些解决方案的接受度和可行性。
    Precision medicine is increasingly important in cancer treatment. Tumor-agnostic therapies are used regardless of tumor entity because they target specific biomarkers in tumors. In Germany, the benefit assessment of oncological pharmaceuticals has traditionally been entity specific. Thus, the assessment of tumor-agnostic therapies leaves stakeholders with various challenges. Our aim was to systematically identify challenges and possible solutions for the benefit assessment of therapies in tumor-agnostic indications using a 2-step sequential qualitative approach.
    To identify relevant challenges, we conducted qualitative interviews with different stakeholders who were involved in previous benefit assessments of tumor-agnostic therapies in Germany. To identify possible solutions for these challenges, we systematically searched MEDLINE, Embase, and the websites of European health technology assessment bodies for relevant literature.
    We identified 9 categories of challenges of which the following were deemed particularly relevant: the absence of direct comparative studies, challenges regarding the use of basket studies and indirect comparisons, challenges in determining the appropriate comparative therapy in a tumor-agnostic indication, and challenges on the system side. Seven categories of solutions were identified, including an increased use of real-world evidence, making conditional decisions in the context of systematic reassessments, splitting the field of application, and finding (new) ways to design and analyze basket studies.
    A range of possible solutions, which can help to meet the identified challenges in Germany, have been found. Future research should investigate the acceptance and feasibility of these solutions.
    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: Clinical Trial, Phase II
    背景:先前的临床试验已经证明了聚(ADP-核糖)聚合酶(PARP)抑制剂(PARP)在涉及同源重组修复(HRR)基因突变的癌症患者中的潜在功效。此外,随着肿瘤突变负担的增加,用免疫检查点抑制剂(ICI)有效治疗HRR基因突变的癌症。我们建议进行多中心,单臂II期试验(IMAGENE试验),用于评估尼拉帕尼(PARPi)联合程序性细胞死亡-1抑制剂联合治疗对ICIs治疗难治性HRR基因突变癌症患者的疗效和安全性,使用基于下一代测序的循环肿瘤DNA(ctDNA)和肿瘤组织分析.
    方法:本试验的关键合格标准包括通过任何癌基因检测确定的HRR基因突变肿瘤;之前ICI治疗后的进展;以及东部肿瘤协作组表现状态≤1。主要终点是所有患者的确认客观反应率(ORR)。次要终点包括使用CarisAssure(CARIS,美国)。IMAGENE试验的目标样本量为57名患者。生物标志物分析将使用CarisAssure并行进行,蛋白质组分析,和T细胞库分析以揭示外周血中的肿瘤免疫监视。
    UASSIGNED:我们的试验旨在证实PARPi加ICI联合治疗对ICI耐药患者的临床获益。此外,通过转化研究,我们的试验将揭示即使在ICI失败后,对于HRR基因突变的肿瘤患者,哪些患者将受益于靶向联合治疗.
    背景:IMAGENE试验:jRCT,临床试验编号:jRCT2051210120,注册日期:2021年11月9日。
    BACKGROUND: Previous clinical trials have demonstrated the potential efficacy of poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis) in patients with cancer involving homologous recombination repair (HRR) gene-mutation. Moreover, HRR gene-mutated cancers are effectively treated with immune checkpoint inhibitors (ICIs) with the increase in tumor mutation burden. We have proposed to conduct a multicenter, single-arm phase II trial (IMAGENE trial) for evaluating the efficacy and safety of niraparib (PARPi) plus programmed cell death-1 inhibitor combination therapy in patients with HRR gene-mutated cancers who are refractory to ICIs therapy using a next generation sequencing-based circulating tumor DNA (ctDNA) and tumor tissue analysis.
    METHODS: Key eligibility criteria for this trial includes HRR gene-mutated tumor determined by any cancer gene tests; progression after previous ICI treatment; and Eastern Cooperative Oncology Group Performance Status ≤ 1. The primary endpoint is the confirmed objective response rate (ORR) in all patients. The secondary endpoints include the confirmed ORR in patients with HRR gene-mutation of ctDNA using the Caris Assure (CARIS, USA). The target sample size of the IMAGENE trial is 57 patients. Biomarker analyses will be performed in parallel using the Caris Assure, proteome analysis, and T cell repertoire analysis to reveal tumor immunosurveillance in peripheral blood.
    UNASSIGNED: Our trial aims to confirm the clinical benefit of PARPi plus ICI combination therapy in ICI-resistant patients. Furthermore, through translational research, our trial will shed light on which patients would benefit from the targeted combination therapy for patients with HRR gene-mutated tumor even after the failure of ICIs.
    BACKGROUND: The IMAGENE trial: jRCT, Clinical trial no.: jRCT2051210120, Registered date: November 9, 2021.
    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
    DNA修复机制对于维持细胞稳态至关重要。这些修复机制的功能障碍导致细胞DNA突变,致癌转化,细胞死亡。这些相同的缺陷也会产生相对特定于癌细胞的漏洞,并且可能被利用来增加抗癌治疗的治疗指数,从而改善患者的预后。基于抑制DNA损伤反应(DDR)的靶向治疗为DDR缺陷患者开辟了新的治疗前景。目前有两种DNA修复机制用作分子疗法的靶标:线粒体修复(MMR)和同源重组修复(HRR)。这些分子靶标允许在不同实体瘤中基于“检查点抑制剂”(ICIs)药物和“PARP抑制剂”(PARPi)药物的免疫治疗。在这篇综述中,我们将描述这种有趣机制的最新技术,并解释基于这些改变的治疗方案。此外,目前,许多临床试验正在为患有不同实体瘤的dMMR和HRD患者探索更好的治疗方案.
    DNA repair mechanisms are essential for maintaining cellular homeostasis. Malfunction of these repair mechanisms leads to cellular DNA mutations, carcinogenic transformation, and cell death. These same defects also create vulnerabilities that are relatively specific to cancer cells, and which could potentially be exploited to increase the therapeutic index of anticancer treatments and thereby improve patient outcomes. The targeted therapy based on inhibiting the DNA damage response (DDR) opens a new therapeutic landscape for patients with deficient DDR. Currently there are two DNA repair mechanisms that are used as targets for molecular therapies: Mitsmach Repair (MMR) and Homologous Recombination Repair (HRR). These molecular targets allow for immunotherapy treatments based on \"checkpoint inhibitors\" (ICIs) drugs and \"PARP inhibitor\" (PARPi) drugs in different solid tumors. In this review we will describe the state of the art of this interesting mechanism and explain the options for treatment based on these alterations. Moreover, many clinical trials are currently underway exploring better treatment options for dMMR and HRD patients with different solid tumours.
    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
    This study tackles several challenges of evaluating histology-independent treatments using entrectinib as an example. Histology-independent treatments are provided based on genetic marker(s) of tumors, regardless of the tumor type. We evaluated the lifetime cost-effectiveness of testing all patients for NTRK fusions and treating the positive cases with entrectinib compared with no testing and standard of care (SoC) for all patients.
    The health economic model consisted of a decision tree reflecting the NTRK testing phase followed by a microsimulation model reflecting treatment with either entrectinib or SoC. Efficacy of entrectinib was based on data from basket trials, whereas historical data from NTRK-negative patients were corrected for the prognostic value of NTRK fusions to model SoC.
    \"Testing\" (testing for NTRK fusions, with subsequent entrectinib treatment in NTRK-positive patients and SoC in NTRK-negative patients) had higher per-patient quality-adjusted life-years (QALYs) and costs than \"No testing\" (SoC for all patients), with a difference of 0.0043 and €732, respectively. This corresponded to an incremental cost-effectiveness ratio (ICER) of €169 957/QALY and, using a cost-effectiveness threshold of €80 000/QALY, an incremental net monetary benefit of -€388. When excluding the costs of genetic testing for NTRK fusions, the ICER was reduced to €36 290/QALY and the incremental net monetary benefit increased to €188.
    When treatment requires the identification of a genetic marker, the associated costs and effects need to be accounted for. Because of the low prevalence of NTRK fusions, the number needed-to-test to identify patients eligible for entrectinib is large. Excluding the testing phase reduces the ICER substantially.
    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
    短重复DNA序列的改变,被称为微卫星不稳定性(MSI),可以反映不匹配修复(MMR)系统的缺陷,该系统代表了DNA完整性维护的主要参与者。MSI-H/dMMR的发生率已显示为根据肿瘤类型而变化。几项研究证实,DMMR/MSI状态,尽管比PD-L1表达频率低,可以更好地预测实体瘤患者对免疫检查点抑制剂(ICIs)的反应。2016年10月,FDA批准pembrolizumab作为非CRC治疗的突破性疗法。MSI-H/dMMR肿瘤,提供,第一次,肿瘤不可知指征。在下一个未来,MSI-H/dMMR的组织不可知评估可能成为不同晚期实体瘤患者免疫治疗的共同点,为了选择可能受益于该疗法的患者亚组。在这篇综述中,我们提供了描述MSI-H/dMMR肿瘤与免疫治疗反应之间关联的主要临床研究的概述。
    Alterations in short-repetitive DNA sequences, known as microsatellite instability (MSI), can reflect deficiencies in Mismatch Repair (MMR) system which represents a major player in DNA integrity maintenance. The incidence of MSI-H/dMMR has been shown to be variable depending on the tumor type. Several studies confirmed that dMMR/MSI status, although less frequent than PD-L1 expression, may better predict response to immune-checkpoint inhibitors (ICIs) in patients with solid tumors. In October 2016, the FDA granted pembrolizumab as breakthrough therapy for the treatment of non-CRC, MSI-H/dMMR tumors, providing, for the first time, a tumor-agnostic indication. In the next future, the tissue-agnostic evaluation of MSI-H/dMMR could become the common denominator for the immunotherapy treatment of patients with different advanced solid tumors, in order to select patient subgroups which may benefit from this therapy. In this Review we provided an overview of the main clinical studies describing the association between MSI-H/dMMR tumors and immunotherapy response.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号