patient-derived organoid

患者来源的类器官
  • 文章类型: Journal Article
    胰腺神经内分泌肿瘤是胰腺第二常见的肿瘤,大约一半的患者被诊断为肝转移。目前,相关治疗方法的疗效改善仍然有限。因此,目前迫切需要对胰腺神经内分泌肿瘤的分子生物学机制进行深入研究。然而,由于它们相对惰性的生物学,临床前模型极其稀缺。这里,患者来源的类器官,并成功构建了患者来源的异种移植物。这两个模型和先前构建的名为SPNE1的细胞系都来自患有3级非功能性胰腺神经内分泌肿瘤的同一患者,提供新的肿瘤建模平台,并使用免疫组织化学进行表征,全外显子组测序,和单细胞转录组测序。结合免疫缺陷小鼠的肿瘤形成实验,我们选择了最接近概括亲本肿瘤的模型.总的来说,患者来源的异种移植模型最类似于人类肿瘤组织.
    Pancreatic neuroendocrine tumors are the second most common tumors of the pancreas, and approximately half of patients are diagnosed with liver metastases. Currently, the improvement in the efficacy of relevant treatment methods is still limited. Therefore, there is an urgent need for in-depth research on the molecular biological mechanism of pancreatic neuroendocrine tumors. However, due to their relatively inert biology, preclinical models are extremely scarce. Here, the patient-derived organoid, and patient-derived xenograft were successfully constructed. These two models and the previously constructed cell line named SPNE1 all derived from the same patient with a grade 3 non-functional pancreatic neuroendocrine tumor, providing new tumor modeling platforms, and characterized using immunohistochemistry, whole-exome sequencing, and single-cell transcriptome sequencing. Combined with a tumor formation experiment in immunodeficient mice, we selected the model that most closely recapitulated the parental tumor. Overall, the patient-derived xenograft model most closely resembled human tumor tissue.
    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
    推进基础癌症研究和开发新癌症疗法的主要瓶颈之一是缺乏忠实地概括患者肿瘤特性的体外临床前模型。癌细胞系的单层培养物通常会失去亲本肿瘤的异质性,而患者来源的异种移植物(PDX)则具有时间和资源密集型的性质。类器官培养系统的出现及其在癌症研究中的应用为开发新的体外癌症临床前模型提供了独特的机会。在这里,我们回顾了利用类器官培养系统和其他相关三维培养系统研究癌症生物学的最新进展。进行药物筛选,开发癌症疗法。特别是,我们讨论了在基础癌症研究和精准医学中应用从患者来源的类器官(PDO)开始的异种移植作为忠实的癌症临床前模型的优势。
    One of the major bottlenecks in advancing basic cancer research and developing novel cancer therapies is the lack of in vitro pre-clinical models that faithfully recapitulate tumor properties in the patients. Monolayer cultures of cancer cell lines usually lose the heterogeneity of the parental tumors, while patient-derived xenograft (PDX) suffers from its time- and resource-intensive nature. The emergence of organoid culture system and its application in cancer research provides a unique opportunity to develop novel in vitro cancer pre-clinical models. Here we review the recent advances in utilizing organoids culture system and other related three-dimensional culture systems in studying cancer biology, performing drug screening, and developing cancer therapies. In particular, we discuss the advantages of applying xenograft initiated from patient-derived organoids (PDOs) as a faithful cancer pre-clinical model in basic cancer research and precision medicine.
    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
    小分子被认为是靶向致癌细胞内途径(包括表皮生长因子受体(EGFR)信号传导)的新型药物的来源。该研究的主要目标是评估LHT-17-19是否可以被认为是针对EGFR表达肿瘤细胞的有效靶分子。在体外,和体内。这是体内的,离体,和体内实验研究。通过配体的分子对接评估LHT-17-19对EGFR激酶结构域的亲和力。表达EGFR的Hs746T人胃癌细胞培养物和EGFR阳性乳腺癌(BC)的患者来源的类器官(PDO)模型用于分子抗癌特性的体外评估。IC50和GI50指数使用基于MTT和MTS的测试来估计,分别。在10只人源化BALB/c雄性小鼠中建立的患者源性异种移植(PDX)模型上研究了LHT-17-19对EGFR表达突变肺癌的抗癌活性。连续变量表示为平均值±标准偏差。通过双向t检验评估组间差异。Kaplan-Meier曲线用于生存分析。LHT-17-19对EGFR激酶结构域的高亲和力,dG评分为-7.9kcal/mol,EDoc-5.45kcal/mol,Ki101.24uM是由于分子间π-σ键的形成和配体分子内的转化。LHT-17-19诱导抗EGFR表达胃癌细胞的细胞毒性,IC50为0.32µM(95%置信区间[CI]0.11-0.54µM)。该衍生物抑制表达EGFR的BCPDO的生长,GI50为16.25µM(95%CI4.44-28.04µM)。2mg/kgLHT-17-19在7天内每天静脉注射抑制PDX肿瘤生长和转移活性,延长动物的生存时间,并消除残留肿瘤淋巴结中的EGFR突变肺癌细胞。LHT-17-19可以被认为是进一步寻找有希望的分子的分子平台,表达EGFR的癌细胞抑制剂。
    Small molecules are considered a source of novel medicines targeting carcinogenic intracellular pathways including epidermal growth factor receptor (EGFR) signaling. The main goal of the study is to assess whether LHT-17-19 could be considered an effective target molecule against EGFR-expressing tumor cells in silico, in vitro, and in vivo. This was an in vivo, ex vivo, and in vivo experimental study. LHT-17-19 affinity to EGFR\'s kinase domain was assessed by the ligand\'s molecular docking. EGFR-expressing Hs746T human gastric cancer cell culture and patient-derived organoid (PDO) model of EGFR-positive breast cancer (BC) were used for in vitro assessment of the molecule anticancer property. IC50 and GI50 indexes were estimated using MTT- and MTS-based tests, respectively. Anticancer activity of LHT-17-19 against EGFR-expressing mutant lung carcinoma was studied on patient-derived xenograft (PDX) model established in 10 humanized BALB/c male mice. Continuous variables were presented as a mean ± standard deviation. Intergroup differences were assessed by two-way t-test. Kaplan-Meier\'s curves were used for survival analysis. High affinity of LHT-17-19 for the EGFR kinase domain with dG score -7.9 kcal/mol, EDoc-5.45 kcal/mol, and Ki 101.24 uM was due to intermolecular π-σ bonds formation and the ligand intramolecular transformation. LHT-17-19 induced anti-EGFR-expressing gastric cancer cells cytotoxicity with IC50 0.32 µM (95% confidence interval [CI] 0.11-0.54 µM). The derivative inhibited growth of EGFR-expressing BC PDO with GI50 16.25 µM (95% CI 4.44-28.04 µM). 2 mg/kg LHT-17-19 intravenously daily during 7 days inhibited PDX tumor growth and metastatic activity, prolonged animals\' survival, and eliminated EGFR-mutant lung cancer cells from residual tumor\'s node. LHT-17-19 may be considered a molecular platform for further search of promising molecules, EGFR-expressing cancer cell inhibitors.
    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
    纤维发育不良(FD)是一种罕见的骨骼疾病,其特征是用良性纤维骨组织代替正常骨骼。缺乏合适的研究模型阻碍了我们对病理生理学和治疗选择的理解的发展。在这项研究中,我们开发了一种体外器官型模型,能够概括FD的关键内在和表型特性。最初,从患者病变组织中分离的单个细胞的转录组学分析揭示了病变内分子和细胞异质性。利用这些见解,我们使用从患者FD病变获得的原代细胞建立了患者来源的类器官(PDO).PDO的评估证明了在FD病变中观察到的纤维化相关组成细胞类型和转录特征的保留。此外,PDO保留了FD特有的基因组和代谢改变的不同星座。组织学评估进一步证实了PDO的保真度,以概括FD的重要表型特征,强调了其病理生理相关性。我们的发现代表了该领域的有意义的进展,因为它们为三维背景下罕见骨病变的体外建模开辟了可能性,并且可能标志着为研究和治疗研究创建个性化平台的第一步。
    Fibrous dysplasia (FD) is a rare bone disorder characterized by the replacement of normal bone with benign fibro-osseous tissue. Developments in our understanding of the pathophysiology and treatment options are impeded by the lack of suitable research models. In this study, we developed an in vitro organotypic model capable of recapitulating key intrinsic and phenotypic properties of FD. Initially, transcriptomic profiling of individual cells isolated from patient lesional tissues unveiled intralesional molecular and cellular heterogeneity. Leveraging these insights, we established patient-derived organoids (PDOs) using primary cells obtained from patient FD lesions. Evaluation of PDOs demonstrated preservation of fibrosis-associated constituent cell types and transcriptional signatures observed in FD lesions. Additionally, PDOs retained distinct constellations of genomic and metabolic alterations characteristic of FD. Histological evaluation further corroborated the fidelity of PDOs in recapitulating important phenotypic features of FD that underscore their pathophysiological relevance. Our findings represent meaningful progress in the field, as they open up the possibility for in vitro modeling of rare bone lesions in a three-dimensional context and may signify the first step towards creating a personalized platform for research and therapeutic studies.
    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
    尽管在确定人类肝癌基因组改变方面做出了相当大的努力,这些改变可能揭示了药物靶标,多组学数据的系统翻译仍然具有挑战性.这里,我们报道了来自中国人群的64例患者来源的肝胆肿瘤类器官(PDHOs)的长期培养成功.观察到对265种代谢和表观遗传学相关化学物质和36种抗癌药物的不同反应。整个基因组的整合,转录组,染色质可及性概况,64种临床相关药物的药物敏感性结果定义了超过32,000种基因组-药物相互作用。RUNX1启动子突变与染色质可及性增加和伴随的基因表达增加有关,促进一组在肝胆肿瘤中优先敏感的药物。这些结果不仅提供了人类肝癌的带注释的PDHO生物样本库,而且还提出了一种系统的方法,可以全面了解肝癌的基因调控网络。推进潜在个性化医疗的应用。
    Despite considerable efforts to identify human liver cancer genomic alterations that might unveil druggable targets, the systematic translation of multiomics data remains challenging. Here, we report success in long-term culture of 64 patient-derived hepatobiliary tumor organoids (PDHOs) from a Chinese population. A divergent response to 265 metabolism- and epigenetics-related chemicals and 36 anti-cancer drugs is observed. Integration of the whole genome, transcriptome, chromatin accessibility profiles, and drug sensitivity results of 64 clinically relevant drugs defines over 32,000 genome-drug interactions. RUNX1 promoter mutation is associated with an increase in chromatin accessibility and a concomitant gene expression increase, promoting a cluster of drugs preferentially sensitive in hepatobiliary tumors. These results not only provide an annotated PDHO biobank of human liver cancer but also suggest a systematic approach for obtaining a comprehensive understanding of the gene-regulatory network of liver cancer, advancing the applications of potential personalized medicine.
    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:细胞肿瘤蛋白p53(TP53)是一种肿瘤抑制基因,在人类癌症中经常发生突变。在各种癌症类型中,非常侵袭性的高级别浆液性卵巢癌(HGSOC)表现出TP53突变的最高患病率,>96%的病例。尽管努力重新激活p53,但尚未批准临床药物来挽救p53功能。在这项研究中,我们的主要目标是将体外转录(IVT)野生型(WT)p53-mRNA施用于HGSOC细胞系,原代细胞,和原位小鼠模型,为了探索其对抑制肿瘤生长和扩散的影响,在体外和体内。
    方法:为了恢复p53的活性,使用哺乳动物载体系统在HGSOC细胞系中外源表达WTp53。此外,使用脂质体将IVTWTp53mRNA递送到不同的HGSOC模型系统(原代细胞和患者来源的类器官)中,并研究其增殖,细胞周期进程,凋亡,菌落形成,和染色体不稳定。使用OVCAR-8和原代HGSOC细胞中的RNA测序分析p53mRNA诱导的转录组改变,其次是独创性途径分析。使用原位异种移植物和转移性腹膜内小鼠模型研究了对肿瘤生长和转移的体内作用。
    结果:使用新设计的基于IVTmRNA的方法,在不同的HGSOC模型系统中探索了TP53抑癌基因的再激活。WTp53mRNA的引入引发剂量依赖性细胞凋亡,细胞周期停滞,和有效的持久抑制HGSOC细胞增殖。基于mRNA的p53再激活后OVCAR-8细胞的转录组分析揭示了与p53信号相关的基因表达的显著改变。如细胞凋亡,细胞周期调节,和DNA损伤。同时恢复p53功能可减少HGSOC细胞内的染色体不稳定性,强调其在保护基因组完整性方面的关键贡献,通过缓和由复制应激引起的双链断裂的基线发生。此外,在各种老鼠模型中,p53mRNA治疗以剂量依赖性方式减少了肿瘤生长并抑制了肿瘤细胞在腹膜腔中的传播。
    结论:基于IVTmRNA的p53再激活有望成为HGSOC的潜在治疗策略,为p53功能的分子机制及其在卵巢癌治疗中的相关性提供有价值的见解。
    The cellular tumor protein p53 (TP53) is a tumor suppressor gene that is frequently mutated in human cancers. Among various cancer types, the very aggressive high-grade serous ovarian carcinoma (HGSOC) exhibits the highest prevalence of TP53 mutations, present in >96% of cases. Despite intensive efforts to reactivate p53, no clinical drug has been approved to rescue p53 function. In this study, our primary objective was to administer in vitro-transcribed (IVT) wild-type (WT) p53-mRNA to HGSOC cell lines, primary cells, and orthotopic mouse models, with the aim of exploring its impact on inhibiting tumor growth and dissemination, both in vitro and in vivo.
    To restore the activity of p53, WT p53 was exogenously expressed in HGSOC cell lines using a mammalian vector system. Moreover, IVT WT p53 mRNA was delivered into different HGSOC model systems (primary cells and patient-derived organoids) using liposomes and studied for proliferation, cell cycle progression, apoptosis, colony formation, and chromosomal instability. Transcriptomic alterations induced by p53 mRNA were analyzed using RNA sequencing in OVCAR-8 and primary HGSOC cells, followed by ingenuity pathway analysis. In vivo effects on tumor growth and metastasis were studied using orthotopic xenografts and metastatic intraperitoneal mouse models.
    Reactivation of the TP53 tumor suppressor gene was explored in different HGSOC model systems using newly designed IVT mRNA-based methods. The introduction of WT p53 mRNA triggered dose-dependent apoptosis, cell cycle arrest, and potent long-lasting inhibition of HGSOC cell proliferation. Transcriptome analysis of OVCAR-8 cells upon mRNA-based p53 reactivation revealed significant alterations in gene expression related to p53 signaling, such as apoptosis, cell cycle regulation, and DNA damage. Restoring p53 function concurrently reduces chromosomal instability within the HGSOC cells, underscoring its crucial contribution in safeguarding genomic integrity by moderating the baseline occurrence of double-strand breaks arising from replication stress. Furthermore, in various mouse models, treatment with p53 mRNA reduced tumor growth and inhibited tumor cell dissemination in the peritoneal cavity in a dose-dependent manner.
    The IVT mRNA-based reactivation of p53 holds promise as a potential therapeutic strategy for HGSOC, providing valuable insights into the molecular mechanisms underlying p53 function and its relevance in ovarian cancer treatment.
    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
    2022年12月,美国食品和药物管理局(FDA)取消了开发中的药物在临床评估之前必须经过动物测试的要求。该声明现在要求建立和验证与肿瘤复杂性密切相关并可以预测治疗反应的体外临床前模型。幸运的是,患者来源的类器官(PDO)培养物的出现使得能够通过重组组织特异性特征来体外模拟人类肿瘤的病理生理学.这些特征包括组织病理学变异性,分子表达谱,亲本组织的遗传和细胞异质性,此外,越来越多的证据表明,预测患者治疗反应的能力。专注于高致死性和异质性胃肠道(GI)肿瘤,本文介绍PDO的最新技术和当前方法。我们强调潜在的补充,改进和测试需要允许离体研究肿瘤微环境,以及对化疗和免疫治疗等治疗的临床反应的预测价值提供评论。
    In December 2022 the US Food and Drug Administration (FDA) removed the requirement that drugs in development must undergo animal testing before clinical evaluation, a declaration that now demands the establishment and verification of ex vivo preclinical models that closely represent tumor complexity and that can predict therapeutic response. Fortunately, the emergence of patient-derived organoid (PDOs) culture has enabled the ex vivo mimicking of the pathophysiology of human tumors with the reassembly of tissue-specific features. These features include histopathological variability, molecular expression profiles, genetic and cellular heterogeneity of parental tissue, and furthermore growing evidence suggests the ability to predict patient therapeutic response. Concentrating on the highly lethal and heterogeneous gastrointestinal (GI) tumors, herein we present the state-of-the-art and the current methodology of PDOs. We highlight the potential additions, improvements and testing required to allow the ex vivo of study the tumor microenvironment, as well as offering commentary on the predictive value of clinical response to treatments such as chemotherapy and immunotherapy.
    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
    患者来源的癌症类器官(PDO)在个性化治疗选择和改善患者预后方面具有广阔的前景。然而,在标准培养平台中生成足够数量的PDO来测试治疗是具有挑战性的。这种挑战对于胰腺导管腺癌(PDAC)尤其严重,其中大多数患者在晚期被诊断为不可切除的肿瘤,并且患者组织呈针状活检的形式。这里描述了使用可从PDAC活检获得的有限量的组织或PDO来测试治疗的微流体装置的开发和表征。已证明,微流体PDO在表型和基因型上与金标准Matrigel类器官相似,具有1)球体均匀性的优点,2)最小的细胞数量要求,和3)不依赖Matrigel。通过测试PDO对几种化疗的反应,证明了微流体PDO的实用性。包括糖原合成酶激酶(GSKI)的抑制剂。此外,微流控类器官培养物用于测试由NK细胞与新型生物制剂组合组成的免疫疗法的有效性。总之,我们的微流控设备提供了相当大的好处,用于基于癌症活检的个性化肿瘤学,在未来,被开发成用于化学疗法或免疫疗法治疗的伴随诊断。
    Patient-derived cancer organoids (PDOs) hold considerable promise for personalizing therapy selection and improving patient outcomes. However, it is challenging to generate PDOs in sufficient numbers to test therapies in standard culture platforms. This challenge is particularly acute for pancreatic ductal adenocarcinoma (PDAC) where most patients are diagnosed at an advanced stage with non-resectable tumors and where patient tissue is in the form of needle biopsies. Here the development and characterization of microfluidic devices for testing therapies using a limited amount of tissue or PDOs available from PDAC biopsies is described. It is demonstrated that microfluidic PDOs are phenotypically and genotypically similar to the gold-standard Matrigel organoids with the advantages of 1) spheroid uniformity, 2) minimal cell number requirement, and 3) not relying on Matrigel. The utility of microfluidic PDOs is proven by testing PDO responses to several chemotherapies, including an inhibitor of glycogen synthase kinase (GSKI). In addition, microfluidic organoid cultures are used to test effectiveness of immunotherapy comprised of NK cells in combination with a novel biologic. In summary, our microfluidic device offers considerable benefits for personalizing oncology based on cancer biopsies and may, in the future, be developed into a companion diagnostic for chemotherapy or immunotherapy treatments.
    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
    患者来源的肿瘤组织和细胞的使用导致了个性化癌症治疗和精准医学的重大进展。基因组测序技术的出现使肿瘤特征的综合分析成为可能。来自自组织癌症干细胞的三维肿瘤类器官是有价值的离体模型,忠实地复制结构,独特的功能,和肿瘤的遗传特征。这些肿瘤类器官已经成为广泛用于药物测试的创新工具,基因组编辑,和移植指导临床个性化治疗。然而,这项新兴技术的一个主要限制是缺乏包括免疫细胞和基质细胞的肿瘤微环境。免疫检查点抑制剂的治疗效果强调了免疫细胞的重要性。特别是渗透到肿瘤附近的细胞毒性T细胞,在患者预后中。为了解决这个限制,已经开发了结合肿瘤类器官和T细胞的共培养技术,为研究个体化药物反应性提供了多种途径。通过整合肿瘤微环境的细胞成分,包括T细胞,进入肿瘤类器官培养物,免疫肿瘤学已经接受了这项技术,正在迅速推进。肿瘤类器官共培养模型的最新进展使人们能够更好地理解这种新型模型的优点和局限性。探索其全部潜力。这篇综述集中在类器官-T细胞共培养模型在癌症研究中的当前应用,并强调了其在抗癌治疗中更广泛实施需要解决的剩余挑战。
    The use of patient-derived tumor tissues and cells has led to significant advances in personalized cancer therapy and precision medicine. The advent of genomic sequencing technologies has enabled the comprehensive analysis of tumor characteristics. The three-dimensional tumor organoids derived from self-organizing cancer stem cells are valuable ex vivo models that faithfully replicate the structure, unique features, and genetic characteristics of tumors. These tumor organoids have emerged as innovative tools that are extensively employed in drug testing, genome editing, and transplantation to guide personalized therapy in clinical settings. However, a major limitation of this emerging technology is the absence of a tumor microenvironment that includes immune and stromal cells. The therapeutic efficacy of immune checkpoint inhibitors has underscored the importance of immune cells, particularly cytotoxic T cells that infiltrate the vicinity of tumors, in patient prognosis. To address this limitation, co-culture techniques combining tumor organoids and T cells have been developed, offering diverse avenues for studying individualized drug responsiveness. By integrating cellular components of the tumor microenvironment, including T cells, into tumor organoid cultures, immuno-oncology has embraced this technology, which is rapidly advancing. Recent progress in co-culture models of tumor organoids has allowed for a better understanding of the advantages and limitations of this novel model, thereby exploring its full potential. This review focuses on the current applications of organoid-T cell co-culture models in cancer research and highlights the remaining challenges that need to be addressed for its broader implementation in anti-cancer 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: Journal Article
    肺肿瘤类器官(LTO)作为体外临床前模型引起了人们的关注;然而,其临床和实验应用尚未完全确立。
    我们试图从接受肺切除术的肺癌患者的切除标本中建立LTO。评估了与LTO建立相关的临床病理特征。对LTO及其亲本肿瘤进行组织学评估和遗传分析。在免疫活性小鼠中产生类器官来源的异种移植物。使用细胞增殖试验评估药物敏感性。
    我们从79个肺癌样本中建立了53个LTO,包括10种长期文化模式。鳞状细胞癌的建立率显着低于其他组织学类型(48%对75%,p=0.034)。在LTO之间证实了组织学相似性,亲本肿瘤,和类器官来源的异种移植物。七个突变,包括两个EGFRL858R和一个EGFR外显子20H773delinsYNPY突变,在LTO和亲本肿瘤中均检测到;在LTO或亲本肿瘤中检测到其他四种突变。LTO的广泛培养能力(传代>10次)与患者预后不良相关。携带EGFRH773delinsYNPY的LTO9细胞对奥希替尼敏感。父母的病人,有新的转移性病变的人,接受奥希替尼治疗并表现出显著的反应。
    LTO的建立和生长速率与组织学亚型和肿瘤大小有关。来自切除标本的LTO已成为临床前模型,可用于预测药物反应并加速罕见突变患者治疗策略的开发。
    UNASSIGNED: Lung tumor organoids (LTOs) have attracted attention as in vitro preclinical models; however, their clinical and experimental applications have not been fully established.
    UNASSIGNED: We attempted to establish LTOs from resected specimens of patients with lung cancer who underwent lung resection. Clinicopathologic characteristics related to the establishment of LTOs were evaluated. Histologic assessment and genetic analysis were conducted for both LTOs and their parental tumors. Organoid-derived xenografts were generated in immunocompetent mice. Drug sensitivity was assessed using cell proliferation assays.
    UNASSIGNED: We established 53 LTOs from 79 lung cancer samples, including 10 long-term culture models. The establishment rate was significantly lower in squamous cell carcinomas than in other histologic types (48% versus 75%, p = 0.034). Histologic similarities were confirmed among LTOs, the parental tumors, and organoid-derived xenografts. Seven mutations, including two EGFR L858R and one EGFR exon 20 H773delinsYNPY mutations, were detected in both LTO and parental tumors; the other four mutations were detected in either LTO or parental tumors. The extensive culture ability of LTO (passaged >10 times) correlated with poor patient prognosis. LTO9 cells harboring EGFR H773delinsYNPY were sensitive to osimertinib. The parental patient, who had new metastatic lesions, was treated with osimertinib and exhibited a remarkable response.
    UNASSIGNED: The establishment and growth rates of LTOs were associated with the histologic subtype and tumor size. LTOs derived from resected specimens have become preclinical models that can be used to predict drug responses and accelerate the development of treatment strategies for patients with rare mutations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号