Precision Oncology

精准肿瘤学
  • 文章类型: Journal Article
    胰腺肉瘤样癌(SCP)是胰腺未分化导管腺癌的一种罕见的侵袭性亚型,预后普遍较差,全球仅报告散发病例。组织学上,SCP最显着的特征是肿瘤中存在大量的类间质梭形肿瘤细胞,缺乏腺体分化。免疫组织化学,SCP的特征在于间质和上皮标记的表达。只有少数病例报告,对其分子和临床病理特征的了解有限。因此,本研究进行了文献检索,以确定所有相关已发表的研究.本综述概述了组织发生,诊断,遗传特征,SCP的预后和治疗,特别关注分子改变。此外,据报道,单中心经验,这增加了文献中可用的有限证据。
    Sarcomatoid carcinoma of the pancreas (SCP) is a rare and aggressive subtype of undifferentiated pancreatic ductal adenocarcinoma, with a generally poor prognosis and only sporadic cases reported worldwide. Histologically, the most notable feature of SCP is the presence of abundant of mesenchymatoid spindle tumor cells in the tumor, which lack glandular differentiation. Immunohistochemically, SCP is characterized by the expression of both mesenchymal and epithelial markers. With only a few reported cases, there is limited knowledge about its molecular and clinicopathological characteristics. Therefore, the present study performed a literature search to identify all relevant published studies. The present review provides an overview of the histogenesis, diagnosis, genetic features, prognosis and treatment of SCP, specifically focusing on the molecular alterations. Furthermore, a single-center experience is reported, which adds to the limited evidence available in the literature.
    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
    精准肿瘤学承诺为癌症患者提供个性化的药物和临床护理:也就是说,“正确的药物,为了正确的病人,在正确的剂量下,在正确的时间。“虽然治疗耐药性和毒性的风险分层是精确肿瘤学的关键,有多种方法可以实现这种分层,例如,遗传,基于功能途径,在其他人中。在急性淋巴细胞白血病(ALL)的情况下,非常需要向精确肿瘤学发展,其中成年患者的存活率为30%至70%。本研究报道了成人B-ALL的通路活性特征,着眼于精确的肿瘤学。来自三个不同表达数据集的转录组概况,包括346名青少年或成人B-ALL患者,用于确定B-ALL中通常被破坏的信号通路的活性。通路活性分析显示,Ph样ALL与Ph阳性ALL非常相似。尽管在平均途径活性水平上是这种情况,B-ALL中的途径活性模式与遗传亚型不同。重要的是,聚类分析显示,基于通路活性,B-ALL患者存在五个不同的簇,每个集群都显示出独特的通路激活模式。因此,识别基于途径的亚型似乎至关重要,考虑到具有相同遗传亚型的患者之间固有的异质性。总之,基于途径的B-ALL分层可能允许同时靶向每个ALL亚型中的高活性途径,因此,可能为这种癌症的个性化/精准医疗开辟新的创新途径,这种癌症在成年患者中的预后与儿童相比仍然较差。
    Precision oncology promises individually tailored drugs and clinical care for patients with cancer: That is, \"the right drug, for the right patient, at the right dose, and at the right time.\" Although stratification of the risk for treatment resistance and toxicity is key to precision oncology, there are multiple ways in which such stratification can be achieved, for example, genetic, functional pathway based, among others. Moving toward precision oncology is sorely needed in the case of acute lymphoblastic leukemia (ALL) wherein adult patients display survival rates ranging from 30% to 70%. The present study reports on the pathway activity signature of adult B-ALL, with an eye to precision oncology. Transcriptome profiles from three different expression datasets, comprising 346 patients who were adolescents or adults with B-ALL, were harnessed to determine the activity of signaling pathways commonly disrupted in B-ALL. Pathway activity analyses revealed that Ph-like ALL closely resembles Ph-positive ALL. Although this was the case at the average pathway activity level, the pathway activity patterns in B-ALL differ from genetic subtypes. Importantly, clustering analysis revealed that five distinct clusters exist in B-ALL patients based on pathway activity, with each cluster displaying a unique pattern of pathway activation. Identifying pathway-based subtypes thus appears to be crucial, considering the inherent heterogeneity among patients with the same genetic subtype. In conclusion, a pathway-based stratification of the B-ALL could potentially allow for simultaneously targeting highly active pathways within each ALL subtype, and thus might open up new avenues of innovation for personalized/precision medicine in this cancer that continues to have poor prognosis in adult patients compared with the children.
    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:高通量测序技术已经彻底改变了肿瘤学。配对的种系-肿瘤DNA分析已成为一种全面的策略,可发现参加精确肿瘤学试验的晚期癌症患者(ACP)的可操作改变。然而,在变异解释和处理偶然的种系发现方面仍然存在挑战.
    方法:我们进行了一项涉及MOSCATO(NCT01566019)和MATCHR(NCT02517892)试验的288个ACP的研究,以评估影响癌症相关基因的种系变异。使用一组250个癌症相关基因进行种系DNA测序,结果在肿瘤分子委员会会议期间进行了讨论。
    结果:根据ESCAT分类对种系致病变异体(PV)进行分类。肺癌(36.8%),其次是前列腺癌(18.4%)和乳腺癌(17.7%),包括最常见的肿瘤类型。12.5%的患者发现PVs。大多数PV被归类为ESCATX(63.9%),突出有限的治疗可操作性。值得注意的是,2%的患者有可操作的变异(ESCATI-A/II-A)。偶然发现包括7.3%的PVs患者存在癌症易感性基因,2.4%的人有非常高风险的潜力,需要强制性的致癌咨询。近五分之一的患者(21.9%)至少有一个VUS。
    结论:我们的研究强调了种系测序在识别可操作的改变方面的重要性,以及在精确肿瘤学试验中需要改进的变异解释和预测试咨询计划。
    BACKGROUND: High-throughput sequencing techniques have revolutionized oncology. Paired germline-tumor DNA analysis has emerged as a comprehensive strategy to uncover actionable alterations in advanced cancer patients (ACP) enrolled in precision oncology trials. However, challenges persist in variant interpretation and managing incidental germline findings.
    METHODS: We conducted a study involving 288 ACP from MOSCATO (NCT01566019) and MATCHR (NCT02517892) trials to assess germline variants impacting cancer-related genes. Germline DNA sequencing was performed using a panel of 250 cancer-related genes, and the results were discussed during tumor molecular board sessions.
    RESULTS: Germline pathogenic variants (PV) were classified according to the ESCAT classification. Lung cancer (36.8 %), followed by prostate (18.4 %) and breast cancer (17.7 %), comprised the most prevalent tumor types. PVs were found in 12.5 % of patients. Most PVs were classified as ESCAT X (63.9 %), highlighting limited therapeutic actionability. Notably,2 % of patients had actionable variants (ESCAT I-A/II-A). Incidental findings included 7.3 % of patients with PVs in cancer-predisposition genes, with 2.4 % having very high-risk potential, necessitating mandatory oncogenetic counseling. Nearly one in five patients (21.9 %) had at least one VUS.
    CONCLUSIONS: Our study underscores the significance of germline sequencing in identifying actionable alterations and the need for improved variant interpretation as well as pretest counseling plans in precision oncology trials.
    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
    癌症仍然是一个巨大的全球健康挑战。其发病率和死亡率持续反映了其重大影响。精确肿瘤学的出现为靶向以前被认为是常规疗法和限制脱靶细胞毒性的“不可药用”的致癌驱动因素提供了突破。已经彻底改变了精确肿瘤学领域的两种突破性技术主要是CRISPR-Cas9基因编辑和最近的PROTAC(PROteolidTArgetingChimeras)靶向蛋白质降解技术。特别是CRISPR-Cas9,由于其精确修饰DNA序列的卓越能力,获得了广泛的认可和赞誉。而不是编辑遗传密码,PROTACs利用泛素蛋白酶体降解机制来选择性地降解感兴趣的蛋白质。尽管CRISPR-Cas9和PROTAC技术的原理不同,他们的共同目标是推进精准肿瘤学,这两种方法在临床前和临床试验中都显示出巨大的潜力.CRISPR-Cas9已经证明了其在这一领域的临床潜力,由于其能够直接和间接地修饰基因,高效,可逆,适应性强,和组织特异性方式,以及它作为诊断工具的潜力。另一方面,口服低剂量给药的能力,广泛瞄准,组织特异性,和可控性增强了PROTAC的临床潜力。因此,在精准肿瘤学领域,使用CRISPR技术的基因编辑彻底改变了针对性的干预措施,虽然PROTACs的出现通过实现选择性蛋白质降解进一步扩大了治疗领域。与其将它们视为精确肿瘤学领域中相互排斥或竞争的方法,它们的使用是上下文相关的(即,基于疾病的分子机制),它们可能可以协同使用,以补充CRISPR的优势,反之亦然。在这里,我们回顾了CRISPR和PROTAC设计的现状,以及它们在精确肿瘤学领域的临床潜力,临床试验数据,局限性,并比较它们在精确临床肿瘤学中的意义。
    Cancer continues to present a substantial global health challenge, with its incidence and mortality rates persistently reflecting its significant impact. The emergence of precision oncology has provided a breakthrough in targeting oncogenic drivers previously deemed \"undruggable\" by conventional therapeutics and by limiting off-target cytotoxicity. Two groundbreaking technologies that have revolutionized the field of precision oncology are primarily CRISPR-Cas9 gene editing and more recently PROTAC (PROteolysis TArgeting Chimeras) targeted protein degradation technology. CRISPR-Cas9, in particular, has gained widespread recognition and acclaim due to its remarkable ability to modify DNA sequences precisely. Rather than editing the genetic code, PROTACs harness the ubiquitin proteasome degradation machinery to degrade proteins of interest selectively. Even though CRISPR-Cas9 and PROTAC technologies operate on different principles, they share a common goal of advancing precision oncology whereby both approaches have demonstrated remarkable potential in preclinical and promising data in clinical trials. CRISPR-Cas9 has demonstrated its clinical potential in this field due to its ability to modify genes directly and indirectly in a precise, efficient, reversible, adaptable, and tissue-specific manner, and its potential as a diagnostic tool. On the other hand, the ability to administer in low doses orally, broad targeting, tissue specificity, and controllability have reinforced the clinical potential of PROTAC. Thus, in the field of precision oncology, gene editing using CRISPR technology has revolutionized targeted interventions, while the emergence of PROTACs has further expanded the therapeutic landscape by enabling selective protein degradation. Rather than viewing them as mutually exclusive or competing methods in the field of precision oncology, their use is context-dependent (i.e., based on the molecular mechanisms of the disease) and they potentially could be used synergistically complementing the strengths of CRISPR and vice versa. Herein, we review the current status of CRISPR and PROTAC designs and their implications in the field of precision oncology in terms of clinical potential, clinical trial data, limitations, and compare their implications in precision clinical oncology.
    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
    皮肤癌占所有诊断癌症病例的三分之一,仍然是一个主要的健康问题。遗传和环境参数是与皮肤癌发展相关的两个主要危险因素,紫外线辐射是最常见的环境危险因素。研究还发现肤色白皙,砷毒性,室内鞣制,和家族病史是皮肤癌的主要原因。预防和早期诊断在减少皮肤癌的发生和确保有效治疗中起着至关重要的作用。最近的研究集中在探索微创或非侵入性诊断技术以及人工智能,以促进快速准确的诊断。皮肤癌的治疗范围从传统的手术切除到各种先进的方法,如光疗,放射治疗,免疫疗法,靶向治疗,和联合治疗。最近的研究集中在免疫疗法,随着新的检查点抑制剂和个性化免疫治疗的引入,增强了治疗效果。多组学的进步,纳米技术,和人工智能进一步加深了对肿瘤生长及其与治疗效果相互作用的机制的理解,这为精准肿瘤学铺平了道路。这篇综述旨在强调皮肤癌的理解和管理的最新进展,并提供现有和新兴诊断的概述,预后,和治疗方式,同时强调需要进一步研究以弥合现有知识差距的领域。
    Skin cancer comprises one-third of all diagnosed cancer cases and remains a major health concern. Genetic and environmental parameters serve as the two main risk factors associated with the development of skin cancer, with ultraviolet radiation being the most common environmental risk factor. Studies have also found fair complexion, arsenic toxicity, indoor tanning, and family history among the prevailing causes of skin cancer. Prevention and early diagnosis play a crucial role in reducing the frequency and ensuring effective management of skin cancer. Recent studies have focused on exploring minimally invasive or non-invasive diagnostic technologies along with artificial intelligence to facilitate rapid and accurate diagnosis. The treatment of skin cancer ranges from traditional surgical excision to various advanced methods such as phototherapy, radiotherapy, immunotherapy, targeted therapy, and combination therapy. Recent studies have focused on immunotherapy, with the introduction of new checkpoint inhibitors and personalized immunotherapy enhancing treatment efficacy. Advancements in multi-omics, nanotechnology, and artificial intelligence have further deepened the understanding of the mechanisms underlying tumoral growth and their interaction with therapeutic effects, which has paved the way for precision oncology. This review aims to highlight the recent advancements in the understanding and management of skin cancer, and provide an overview of existing and emerging diagnostic, prognostic, and therapeutic modalities, while highlighting areas that require further research to bridge the existing knowledge gaps.
    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
    (1)背景:胰腺导管腺癌(PDAC)虽然治疗进展,但生存率较低。目的:本研究旨在展示分子谱分析如何可能指导个性化治疗策略,这可能有助于改善PDAC患者的生存结局。(2)材料与方法:对某学术中心142例PDAC患者进行回顾性分析。患者接受化疗和下一代测序进行分子谱分析。使用Reactome途径数据库鉴定关键的致癌途径。使用Kaplan-Meier曲线和Cox比例风险回归进行生存分析。(3)结果:患者主要接受FOLFIRINOX(n=62)或吉西他滨nab-紫杉醇(n=62)作为初始化疗。中位OS为13.6个月。NOTCH患者的中位OS更长(15vs.12.3个月,p=0.007)和KIT通路突变(21.3vs.12.12个月,p=0.04)。组合途径分析表明对存活的潜在协同作用。在PFS中,PI3K通路(6.6与5.7个月,p=0.03)和KIT途径(10.3vs.6.2个月,p=0.03)突变与吉西他滨nab-紫杉醇亚组PFS改善相关。(4)结论:分子谱分析可能在PDAC中发挥作用,用于预测结果和对FOLFIRINOX和吉西他滨nab-紫杉醇等疗法的反应。将基因组数据整合到临床决策中可以使PDAC治疗受益,尽管需要进一步验证才能在PDAC管理中充分利用精确肿瘤学。
    (1) Background: Pancreatic ductal adenocarcinoma (PDAC) has low survival rates despite treatment advancements. Aim: This study aims to show how molecular profiling could possibly guide personalized treatment strategies, which may help improve survival outcomes in patients with PDAC. (2) Materials and Methods: A retrospective analysis of 142 PDAC patients from a single academic center was conducted. Patients underwent chemotherapy and next-generation sequencing for molecular profiling. Key oncogenic pathways were identified using the Reactome pathway database. Survival analysis was performed using Kaplan-Meier curves and Cox Proportional Hazards Regression. (3) Results: Patients mainly received FOLFIRINOX (n = 62) or gemcitabine nab-paclitaxel (n = 62) as initial chemotherapy. The median OS was 13.6 months. Longer median OS was noted in patients with NOTCH (15 vs. 12.3 months, p = 0.007) and KIT pathway mutations (21.3 vs. 12.12 months, p = 0.04). Combinatorial pathway analysis indicated potential synergistic effects on survival. In the PFS, PI3K pathway (6.6 vs. 5.7 months, p = 0.03) and KIT pathway (10.3 vs. 6.2 months, p = 0.03) mutations correlated with improved PFS within the gemcitabine nab-paclitaxel subgroup. (4) Conclusions: Molecular profiling could play a role in PDAC for predicting outcomes and responses to therapies like FOLFIRINOX and gemcitabine nab-paclitaxel. Integrating genomic data into clinical decision-making can benefit PDAC treatment, though further validation is needed to fully utilize precision oncology in PDAC management.
    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
    集群定期间隔短回文重复(CRISPR)/Cas9系统,基因工程的突破性创新,彻底改变了我们克服复杂疾病的方法,最终获得CASGEVY™批准用于镰状细胞性贫血。来源于微生物免疫防御机制,CRISPR/Cas9,以精确度为特征,基因编辑的可操作性和普遍性,已被用作精确操纵哺乳动物DNA的通用工具。在将其应用于实践的过程中,新的直系同源物和变体的连续开发永远不会停止。它有助于了解疾病的本质,特别是癌症,这对诊断至关重要,预防,和治疗。CRISPR/Cas9不仅用于研究肿瘤基因的功能,还用于模拟不同的癌症。提供对肿瘤生物学有价值的见解,阻力,和免疫逃避。在癌症治疗中,CRISPR/Cas9有助于开发单独和精确的癌症疗法,可以选择性地激活或失活肿瘤细胞内的基因。旨在削弱肿瘤的生长和侵袭,并使癌细胞对治疗敏感。此外,它促进了创新治疗的发展,提高重编程免疫细胞的靶向效率,以CAR-T方案的进展为例。除了治疗,它是筛选易感基因的有力工具,提供了在肿瘤主动或进展之前进行干预的可能性。然而,尽管潜力巨大,CRISPR/Cas9在癌症研究和治疗中的应用伴随着显著的疗效,效率,技术,和安全考虑。需要不断升级的技术创新来解决这些问题。CRISPR/Cas9系统正在彻底改变癌症研究和治疗,为我们对癌症的理解和管理开辟了新的途径。将这种不断发展的技术整合到临床实践中,有望开创精准肿瘤学的新时代。有针对性的,个性化,和癌症患者的潜在治愈疗法。
    The Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/Cas9 system, a groundbreaking innovation in genetic engineering, has revolutionized our approach to surmounting complex diseases, culminating in CASGEVY™ approved for sickle cell anemia. Derived from a microbial immune defense mechanism, CRISPR/Cas9, characterized as precision, maneuverability and universality in gene editing, has been harnessed as a versatile tool for precisely manipulating DNA in mammals. In the process of applying it to practice, the consecutive exploitation of novel orthologs and variants never ceases. It\'s conducive to understanding the essentialities of diseases, particularly cancer, which is crucial for diagnosis, prevention, and treatment. CRISPR/Cas9 is used not only to investigate tumorous genes functioning but also to model disparate cancers, providing valuable insights into tumor biology, resistance, and immune evasion. Upon cancer therapy, CRISPR/Cas9 is instrumental in developing individual and precise cancer therapies that can selectively activate or deactivate genes within tumor cells, aiming to cripple tumor growth and invasion and sensitize cancer cells to treatments. Furthermore, it facilitates the development of innovative treatments, enhancing the targeting efficiency of reprogrammed immune cells, exemplified by advancements in CAR-T regimen. Beyond therapy, it is a potent tool for screening susceptible genes, offering the possibility of intervening before the tumor initiative or progresses. However, despite its vast potential, the application of CRISPR/Cas9 in cancer research and therapy is accompanied by significant efficacy, efficiency, technical, and safety considerations. Escalating technology innovations are warranted to address these issues. The CRISPR/Cas9 system is revolutionizing cancer research and treatment, opening up new avenues for advancements in our understanding and management of cancers. The integration of this evolving technology into clinical practice promises a new era of precision oncology, with targeted, personalized, and potentially curative therapies for cancer patients.
    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
    肝脏是晚期胃癌(GC)的常见转移部位。尽管诊断和治疗技术取得了重大进展,胃癌肝转移(GCLM)患者的总生存率仍然很低。精确肿瘤学在识别治疗靶标和通过基因组测序和分子表征增强我们对转移机制的理解方面取得了重大进展。因此,全面了解GCLM涉及的各种分子过程和全身治疗的基本原理,以开发新的治疗方法至关重要.本文旨在回顾有关诊断的最新发现,潜在的生物标志物,以及针对GCLM多分子过程的疗法,目的是改善GCLM患者的治疗策略。
    The liver is a frequent site of metastasis in advanced gastric cancer (GC). Despite significant advancements in diagnostic and therapeutic techniques, the overall survival rate for patients afflicted with gastric cancer liver metastasis (GCLM) remains dismally low. Precision oncology has made significant progress in identifying therapeutic targets and enhancing our understanding of metastasis mechanisms through genome sequencing and molecular characterization. Therefore, it is crucial to have a comprehensive understanding of the various molecular processes involved in GCLM and the fundamental principles of systemic therapy to develop new treatment approaches. This paper aims to review recent findings on the diagnosis, potential biomarkers, and therapies targeting the multiple molecular processes of GCLM, with the goal of improving treatment strategies for patients with GCLM.
    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
    下一代测序(NGS)可识别遗传变异以告知个性化治疗计划。成本效益的证据不足阻碍了NGS整合到常规癌症治疗中。个性化治疗的复杂性对传统的经济评价提出了挑战。清楚地描述挑战为未来的成本效益分析提供了信息,以更好地评估和了解健康,偏好-,和基于公平的结果。
    我们进行了范围审查,以描述NGS在肿瘤学中的应用方法和经济评估结果,并确定存在的挑战。我们纳入了自2016年以来发布的27篇文章,来自PubMed的搜索,Embase,和WebofScience。确定的挑战包括定义评估范围,管理证据限制,包括缺乏因果证据,结合基于偏好的效用,并评估基于分配和公平的影响。这些挑战反映了为NGS指导的干预措施产生高质量临床有效性和现实世界证据(RWE)的困难。
    使用RWE调整方法学方法和开发生命周期健康技术评估(HTA)指南对于在肿瘤学中实施NGS至关重要。医疗保健系统,决策者,和HTA组织正面临一个关键的机会,以适应不断发展的临床范式,并创建创新的监管和报销流程,公平,和以病人为导向的医疗保健。
    UNASSIGNED: Next-generation sequencing (NGS) identifies genetic variants to inform personalized treatment plans. Insufficient evidence of cost-effectiveness impedes the integration of NGS into routine cancer care. The complexity of personalized treatment challenges conventional economic evaluation. Clearly delineating challenges informs future cost-effectiveness analyses to better value and contextualize health, preference-, and equity-based outcomes.
    UNASSIGNED: We conducted a scoping review to characterize the applied methods and outcomes of economic evaluations of NGS in oncology and identify existing challenges. We included 27 articles published since 2016 from a search of PubMed, Embase, and Web of Science. Identified challenges included defining the evaluative scope, managing evidentiary limitations including lack of causal evidence, incorporating preference-based utility, and assessing distributional and equity-based impacts. These challenges reflect the difficulty of generating high-quality clinical effectiveness and real-world evidence (RWE) for NGS-guided interventions.
    UNASSIGNED: Adapting methodological approaches and developing life-cycle health technology assessment (HTA) guidance using RWE is crucial for implementing NGS in oncology. Healthcare systems, decision-makers, and HTA organizations are facing a pivotal opportunity to adapt to an evolving clinical paradigm and create innovative regulatory and reimbursement processes that will enable more sustainable, equitable, and patient-oriented healthcare.
    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
    尽管在治疗方面取得了一些进展,晚期胆道癌(BTC)患者在确诊后存活5年的比例仍然很低.对BTC中可靶向遗传改变的日益认识开启了治疗这些患者的新纪元。新的治疗剂靶向突变,如异柠檬酸脱氢酶(IDH),成纤维细胞生长因子受体(FGFR),人表皮生长因子受体(HER),等。在有这些突变的疾病患者的一线治疗进展时,已经建立了新的治疗标准。
    本综述旨在简要总结BTC各种靶向治疗方案的进展。我们还简要讨论了临床和转化研究的未来方向,以采用个性化方法治疗不可切除或晚期BTC。
    一些新的药物继续出现,作为具有靶向突变的晚期BTC患者的可行治疗选择。越来越需要确定克服对这些试剂的原发性和获得性抗性的机制。识别预测对靶向治疗的反应的潜在生物标志物可能有助于采用更量身定制的方法。所有接受晚期BTC治疗的患者应在诊断时进行组织基因组分析。
    UNASSIGNED: Despite several therapeutic advancements, the proportion of patients with advanced biliary tract cancers (BTC) surviving 5 years from diagnosis remains dismal. The increasing recognition of targetable genetic alterations in BTCs has ushered in a new era in the treatment of these patients. Newer therapeutic agents targeting mutations such as isocitrate dehydrogenase (IDH), fibroblastic growth factor receptor (FGFR), human epidermal growth factor receptor (HER), and so on have established a new standard of care for treatment upon progression on frontline therapy in patients with disease harboring these mutations.
    UNASSIGNED: The current review aims to concisely summarize progress with various targeted therapy options for BTC. We also briefly discuss future directions in clinical and translational research for the adoption of a personalized approach for the treatment of unresectable or advanced BTC.
    UNASSIGNED: Several new agents continue to emerge as feasible treatment options for patients with advanced BTC harboring targetable mutations. There is a growing need to identify mechanisms to conquer primary and acquired resistance to these agents. The identification of potential biomarkers that predict response to targeted therapy may be helpful in adopting a more tailored approach. All patients receiving treatment for advanced BTC should undergo tissue genomic profiling at diagnosis.
    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