Patient-derived xenografts

患者来源的异种移植物
  • 文章类型: Journal Article
    胰腺癌是全球癌症相关死亡的第七大原因。检查点免疫疗法尚未在胰腺癌中显示出令人鼓舞的结果,可能是因为免疫原性差和/或免疫抑制微环境。这项研究的目的是开发患者来源的异种移植(PDX)模型,将他们的遗传学与原始活检进行比较,并评估自体肿瘤浸润淋巴细胞(TIL)在胰腺癌中是否具有抗肿瘤活性。
    我们将29名患者的肿瘤皮下移植到NOG小鼠中以产生PDX模型。我们建立了TIL培养物并将它们注射到PDX小鼠中。我们分析了活检和PDX肿瘤的组织学和遗传学。
    29例移植中有11例证实了肿瘤生长。PDX肿瘤在组织学上与原始活检相似,但是因为PDX模型肿瘤中的基质细胞来自小鼠,他们的基因表达与原始活检不同。除程序性死亡配体-1(PD-L1)外的免疫检查点配体在胰腺癌中表达,但PD-L1很少表达。当它被表达时,它与PDX模型中的肿瘤摄取相关。3种表达PD-L1的肿瘤之一是腺鳞癌,另一个有错配修复缺陷。将TIL从6个肿瘤扩增并注射到携带PDX肿瘤的NOG或人白介素-2转基因-NOG小鼠中。在用自体TIL治疗的6个PDX模型中的3个中,可以在人白介素2转基因NOG小鼠中验证肿瘤的消退。包括腺鳞状PDX模型。
    胰腺癌的PDX模型可用于了解更多有关肿瘤特征和生物标志物的信息,并评估对过继性细胞疗法和联合疗法的反应。该模型的主要益处是可以在自体人源化小鼠模型中测试T细胞的修饰以获得临床前数据以支持临床试验的启动。
    UNASSIGNED: Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide. Checkpoint immunotherapy has not yet shown encouraging results in pancreatic cancer possibly because of a poor immunogenicity and/or an immune suppressive microenvironment. The aim of this study was to develop patient-derived xenograft (PDX) models, compare their genetics to the original biopsies, and assess if autologous tumor-infiltrating lymphocytes (TILs) would have antitumoral activity in pancreatic cancer.
    UNASSIGNED: We subcutaneously transplanted tumors from 29 patients into NOG mice to generate PDX models. We established TIL cultures and injected them into PDX mice. We analyzed histology and genetics of biopsies and PDX tumors.
    UNASSIGNED: Tumor growths were confirmed in 11 of 29 transplantations. The PDX tumors histologically resembled their original biopsies, but because stromal cells in the PDX model tumors were from mouse, their gene expression differed from the original biopsies. Immune checkpoint ligands other than programmed death ligand-1 (PD-L1) were expressed in pancreatic cancers, but PD-L1 was rarely expressed. When it was expressed, it correlated with tumor take in PDX models. One of the 3 tumors that expressed PD-L1 was an adenosquamous cancer, and another had a mismatch repair deficiency. TILs were expanded from 6 tumors and were injected into NOG or human interleukin-2 transgenic-NOG mice carrying PDX tumors. Regression of tumors could be verified in human interleukin-2 transgenic-NOG mice in 3 of the 6 PDX models treated with autologous TILs, including the adenosquamous PDX model.
    UNASSIGNED: PDX models of pancreatic cancer can be used to learn more about tumor characteristics and biomarkers and to evaluate responses to adoptive cell therapy and combination therapies. The major benefit of the model is that modifications of T cells can be tested in an autologous humanized mouse model to gain preclinical data to support the initiation of a clinical trial.
    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:稳定同位素示踪剂已越来越多地用于临床前癌症模型系统,包括细胞培养和小鼠异种移植,探测各种癌症的代谢改变,如加速糖酵解和谷氨酰胺分解和生成糖代谢产物。关于不同临床前模型系统在概括肿瘤异常代谢方面的保真度的报道相对较少。
    目的:我们一直在开发几种不同的实验模型系统,用于使用患者来源的组织对非小细胞肺癌(NSCLC1)进行系统生化分析,以评估代谢和表型分析的适当模型。
    方法:为了解决保真度问题,我们对新鲜切除的组织切片进行了详细的稳定同位素分解代谢组学研究,小鼠患者来源的异种移植物(PDX),和使用13C6-葡萄糖和13C5,15N2-谷氨酰胺示踪剂从单个患者获得的细胞。
    结果:尽管我们在三个模型中发现了相似的葡萄糖代谢,与原发性癌组织或直接组织异种移植物(PDX)相比,分离的细胞培养物和源自细胞培养物的异种移植物中的谷氨酰胺利用率明显更高。
    结论:这表明在体外或异种移植物中使用患者来源的癌细胞解释癌症生物化学时需要谨慎。即使在很早的时候,并且对患者来源的组织切片的直接分析为离体代谢组学提供了最佳模型。需要进一步的研究来确定这些观察的一般性。
    BACKGROUND: Stable isotope tracers have been increasingly used in preclinical cancer model systems, including cell culture and mouse xenografts, to probe the altered metabolism of a variety of cancers, such as accelerated glycolysis and glutaminolysis and generation of oncometabolites. Comparatively little has been reported on the fidelity of the different preclinical model systems in recapitulating the aberrant metabolism of tumors.
    OBJECTIVE: We have been developing several different experimental model systems for systems biochemistry analyses of non-small cell lung cancer (NSCLC1) using patient-derived tissues to evaluate appropriate models for metabolic and phenotypic analyses.
    METHODS: To address the issue of fidelity, we have carried out a detailed Stable Isotope-Resolved Metabolomics study of freshly resected tissue slices, mouse patient derived xenografts (PDXs), and cells derived from a single patient using both 13C6-glucose and 13C5,15N2-glutamine tracers.
    RESULTS: Although we found similar glucose metabolism in the three models, glutamine utilization was markedly higher in the isolated cell culture and in cell culture-derived xenografts compared with the primary cancer tissue or direct tissue xenografts (PDX).
    CONCLUSIONS: This suggests that caution is needed in interpreting cancer biochemistry using patient-derived cancer cells in vitro or in xenografts, even at very early passage, and that direct analysis of patient derived tissue slices provides the optimal model for ex vivo metabolomics. Further research is needed to determine the generality of these observations.
    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
    患有未知原发性癌症(CUP)的患者承担着侵袭性疾病的双重负担,并减少了获得治疗的机会。实验模型对于CUP生物学研究和药物测试至关重要。我们得到了两个CUP细胞系(CUP#55和#96),和相应的患者来源的异种移植物(PDX),来自腹水肿瘤细胞。CUP细胞系和PDX进行了组织学检查,免疫表型,分子,和基因组特征证实了原始肿瘤的特征。从肿瘤microRNA表达谱获得起源组织预测,并通过单细胞转录组学证实。基因组测试和FISH分析确定了两个模型中的FGFR2基因扩增,在CUP#55中以均匀染色区域(HSR)的形式和在CUP#96中以双倍分钟的形式。FGFR2被认为是主要的致癌驱动因子和治疗靶点。FGFR2靶向药物BGJ-398(infigratinib)与MEK抑制剂trametinib的组合被证明具有协同作用和异常活性,在体外和体内。通过单细胞基因表达分析联合治疗的效果揭示了肿瘤细胞的显着可塑性和具有上皮表型的细胞的更高敏感性。这项研究使个性化治疗更接近CUP患者,并为FGFR2和MEK靶向转移肿瘤的FGFR2途径激活提供了理论基础。
    Patients with cancer of unknown primary (CUP) carry the double burden of an aggressive disease and reduced access to therapies. Experimental models are pivotal for CUP biology investigation and drug testing. We derived two CUP cell lines (CUP#55 and #96) and corresponding patient-derived xenografts (PDXs), from ascites tumor cells. CUP cell lines and PDXs underwent histological, immune-phenotypical, molecular, and genomic characterization confirming the features of the original tumor. The tissue-of-origin prediction was obtained from the tumor microRNA expression profile and confirmed by single-cell transcriptomics. Genomic testing and fluorescence in situ hybridization analysis identified FGFR2 gene amplification in both models, in the form of homogeneously staining region (HSR) in CUP#55 and double minutes in CUP#96. FGFR2 was recognized as the main oncogenic driver and therapeutic target. FGFR2-targeting drug BGJ398 (infigratinib) in combination with the MEK inhibitor trametinib proved to be synergic and exceptionally active, both in vitro and in vivo. The effects of the combined treatment by single-cell gene expression analysis revealed a remarkable plasticity of tumor cells and the greater sensitivity of cells with epithelial phenotype. This study brings personalized therapy closer to CUP patients and provides the rationale for FGFR2 and MEK targeting in metastatic tumors with FGFR2 pathway activation.
    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:小儿胃肠胰腺神经内分泌肿瘤极为罕见,导致大多数儿科治疗建议基于来自成人的数据。曲美替尼是一种靶向MEK1/2的激酶抑制剂,已用于治疗Ras途径中存在突变的癌症。
    方法:我们利用已建立的具有已知NRAS突变的人类小儿胃肠胰腺神经内分泌样肿瘤患者异种移植物(PDX)来研究MEK抑制作用。我们评估了曲美替尼对增殖的影响,运动性,和体内肿瘤生长。我们建立了PDX的腹膜内转移模型,表征了转移性PDX的表型和基因型,研究了MEK抑制作用。
    结果:我们发现在曲美替尼治疗下,ERK1/2磷酸化降低的靶参与。曲美替尼导致体外细胞生长和运动减少,在小鼠侧腹肿瘤模型中,肿瘤生长减少,动物存活率增加。最后,我们证明曲美替尼能够显著减少胃肠胰腺神经内分泌腹膜内肿瘤转移.
    结论:这些研究的结果支持MEK抑制在小儿NRAS突变实体瘤中的进一步研究。
    BACKGROUND: Pediatric gastroenteropancreatic neuroendocrine tumors are exceedingly rare, resulting in most pediatric treatment recommendations being based on data derived from adults. Trametinib is a kinase inhibitor that targets MEK1/2 and has been employed in the treatment of cancers harboring mutations in the Ras pathway.
    METHODS: We utilized an established human pediatric gastroenteropancreatic neuroendocrine-like tumor patient-derived xenograft (PDX) with a known NRAS mutation to study the effects of MEK inhibition. We evaluated the effects of trametinib on proliferation, motility, and tumor growth in vivo. We created an intraperitoneal metastatic model of this PDX, characterized both the phenotype and the genotype of the metastatic PDX and again, investigated the effects of MEK inhibition.
    RESULTS: We found target engagement with decreased ERK1/2 phosphorylation with trametinib treatment. Trametinib led to decreased in vitro cell growth and motility, and decreased tumor growth and increased animal survival in a murine flank tumor model. Finally, we demonstrated that trametinib was able to significantly decrease gastroenteropancreatic neuroendocrine intraperitoneal tumor metastasis.
    CONCLUSIONS: The results of these studies support the further investigation of MEK inhibition in pediatric NRAS mutated solid tumors.
    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
    前列腺癌主要是激素依赖性的,和医学治疗集中于通过各种方法抑制雄激素生物合成或信号传导。尽管在引入雄激素受体信号抑制剂(ARSI)方面取得了重大进展,患者继续发展为去势抵抗前列腺癌(CRPC),强调需要超越激素阻断的靶向治疗。嵌合抗原受体(CAR)T细胞和其他工程化免疫细胞代表了新一代过继性细胞疗法。虽然这些疗法显著提高了血液系统恶性肿瘤患者的预后,正在进行的研究正在探索CAR-T疗法在实体肿瘤中的更广泛使用,包括晚期前列腺癌.总的来说,CART细胞疗法对实体癌的疗效较差,免疫抑制性肿瘤微环境阻碍T细胞浸润,抗原识别后的活化和细胞毒性。此外,在晚期前列腺癌患者中存在固有的肿瘤异质性,这可能会阻止使用单靶点药物的持久治疗反应.必须克服这些障碍,以告知临床试验设计并提高治疗效果。在这次审查中,我们讨论了正在研究中的创新和合理设计的策略,以改善前列腺癌细胞免疫治疗的临床翻译,并使这些患者的治疗结果最大化。
    Prostate cancer is primarily hormone-dependent, and medical treatments have focused on inhibiting androgen biosynthesis or signaling through various approaches. Despite significant advances with the introduction of androgen receptor signalling inhibitors (ARSIs), patients continue to progress to castration-resistant prostate cancer (CRPC), highlighting the need for targeted therapies that extend beyond hormonal blockade. Chimeric Antigen Receptor (CAR) T cells and other engineered immune cells represent a new generation of adoptive cellular therapies. While these therapies have significantly enhanced outcomes for patients with hematological malignancies, ongoing research is exploring the broader use of CAR T therapy in solid tumors, including advanced prostate cancer. In general, CAR T cell therapies are less effective against solid cancers with the immunosuppressive tumor microenvironment hindering T cell infiltration, activation and cytotoxicity following antigen recognition. In addition, inherent tumor heterogeneity exists in patients with advanced prostate cancer that may prevent durable therapeutic responses using single-target agents. These barriers must be overcome to inform clinical trial design and improve treatment efficacy. In this review, we discuss the innovative and rationally designed strategies under investigation to improve the clinical translation of cellular immunotherapy in prostate cancer and maximise therapeutic outcomes for these 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
    目标:在卵巢癌(OvCa)中,肿瘤细胞高糖皮质激素受体(GR)与患者预后不良有关。体外,GR激活抑制与编码抗凋亡蛋白的基因转录上调相关的化疗诱导的OvCa细胞死亡。最近的一项随机II期研究表明,与单独化疗相比,随机接受选择性GR调节剂(SGRM)加化疗的重度预处理OvCa患者的无进展生存期(PFS)改善。我们假设SGRM治疗可以改善OvCa患者来源的异种移植物(PDX)的卡铂反应。
    方法:用化疗+/-SGRM治疗6个表达GRmRNA(NR3C1)和蛋白的高级别浆液性(HGS)OvCaPDX模型。通过腹膜经皮超声检查纵向测量肿瘤大小。
    结果:6个GR阳性PDX模型之一在添加SGRM的情况下显示出PFS的显着改善。有趣的是,PFS改善的单一模型对卡铂敏感性最低.适度的SGRM活性的可能解释包括5种PDX肿瘤的高卡铂敏感性和SGRM激活患者中的肿瘤侵入性免疫细胞的潜力(在免疫受损小鼠中不存在)。单独的肿瘤GR蛋白表达水平似乎不足以预测SGRM反应。
    结论:在SGRM加化疗治疗后,6个模型中的1个模型显示PFS的显着改善强调需要确定HGSOvCa中SGRM治疗的预测性生物标志物,并更好地识别最有可能从化疗中添加GR调节中受益的患者亚组。
    OBJECTIVE: In ovarian cancer (OvCa), tumor cell high glucocorticoid receptor (GR) has been associated with poor patient prognosis. In vitro, GR activation inhibits chemotherapy-induced OvCa cell death in association with transcriptional upregulation of genes encoding anti-apoptotic proteins. A recent randomized phase II study demonstrated improvement in progression-free survival (PFS) for heavily pre-treated OvCa patients randomized to receive therapy with a selective GR modulator (SGRM) plus chemotherapy compared to chemotherapy alone. We hypothesized that SGRM therapy would improve carboplatin response in OvCa patient-derived xenograft (PDX).
    METHODS: Six high-grade serous (HGS) OvCa PDX models expressing GR mRNA (NR3C1) and protein were treated with chemotherapy +/- SGRM. Tumor size was measured longitudinally by peritoneal transcutaneous ultrasonography.
    RESULTS: One of the 6 GR-positive PDX models showed a significant improvement in PFS with the addition of a SGRM. Interestingly, the single model with an improved PFS was least carboplatin sensitive. Possible explanations for the modest SGRM activity include the high carboplatin sensitivity of 5 of the PDX tumors and the potential that SGRMs activate the tumor invasive immune cells in patients (absent from immunocompromised mice). The level of tumor GR protein expression alone appears insufficient for predicting SGRM response.
    CONCLUSIONS: The significant improvement in PFS shown in 1 of the 6 models after treatment with a SGRM plus chemotherapy underscores the need to determine predictive biomarkers for SGRM therapy in HGS OvCa and to better identify patient subgroups that are most likely to benefit from adding GR modulation to chemotherapy.
    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
    神经母细胞瘤是儿童中最具破坏性的颅外实体恶性肿瘤。尽管有严格的治疗方案,高危神经母细胞瘤患者的预后仍然很差,存活率不到40%。到目前为止,MYCN扩增状态被认为是最预后的因素,但仅对应于~25%的神经母细胞瘤患者。因此,在神经母细胞瘤患者中确定更好的预后和治疗反应标志物至关重要.我们应用了强大的生物信息学数据挖掘工具,如加权基因共表达网络分析,cisTarget,以及两个神经母细胞瘤患者数据集上的单细胞调控网络推断和聚类。我们发现Sin3A相关蛋白30(SAP30),与高风险呈正相关的驱动转录因子,programming,第4阶段,以及神经母细胞瘤患者队列中的低生存率。高风险神经母细胞瘤患者和复发特异性患者来源的异种移植物的肿瘤显示较高的SAP30水平。先进的药物基因组学分析和CRISPR-Cas9筛选表明SAP30的必要性与顺铂耐药性相关,并且在顺铂耐药性患者来源的异种移植肿瘤细胞系中进一步显示出更高的水平。SAP30的沉默在体外诱导细胞死亡,并导致体内肿瘤负荷和大小降低。总之,这些结果表明,SAP30是更好的预后和顺铂耐药标志物,因此是高危神经母细胞瘤的潜在药物靶点.
    Neuroblastoma is the most devastating extracranial solid malignancy in children. Despite an intense treatment regimen, the prognosis for high-risk neuroblastoma patients remains poor, with less than 40% survival. So far, MYCN amplification status is considered the most prognostic factor but corresponds to only ∼25% of neuroblastoma patients. Therefore, it is essential to identify a better prognosis and therapy response marker in neuroblastoma patients. We applied robust bioinformatic data mining tools, such as weighted gene co-expression network analysis, cisTarget, and single-cell regulatory network inference and clustering on two neuroblastoma patient datasets. We found Sin3A-associated protein 30 (SAP30), a driver transcription factor positively associated with high-risk, progression, stage 4, and poor survival in neuroblastoma patient cohorts. Tumors of high-risk neuroblastoma patients and relapse-specific patient-derived xenografts showed higher SAP30 levels. The advanced pharmacogenomic analysis and CRISPR-Cas9 screens indicated that SAP30 essentiality is associated with cisplatin resistance and further showed higher levels in cisplatin-resistant patient-derived xenograft tumor cell lines. Silencing of SAP30 induced cell death in vitro and led to a reduced tumor burden and size in vivo. Altogether, these results indicate that SAP30 is a better prognostic and cisplatin-resistance marker and thus a potential drug target in high-risk neuroblastoma.
    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
    化疗仍然是晚期食管癌的一线治疗方法。然而,由于难以捉摸的化学耐药性,只有有限的一部分个体才能获得持久的益处。这里,我们利用食管鳞状细胞癌患者来源的异种移植物(PDXs)研究临床前环境中的化疗耐药机制.我们观察到活化的癌症相关成纤维细胞(CAF)在对化学疗法具有抗性的PDX的肿瘤微环境中富集。机械上,我们发现癌细胞来源的S100A8通过与CAFs的CD147受体结合触发细胞内RhoA-ROCK-MLC2-MRTF-A途径,诱导CAF极化并导致化学抗性。治疗学上,我们证明阻断S100A8-CD147通路可以提高化疗效率.预后方面,我们发现外周血S100A8水平可以作为化疗反应性的指标.总的来说,我们的研究提供了对食管癌化疗耐药的分子机制的全面了解,并强调了S100A8在食管癌临床治疗中的潜在价值.
    Chemotherapy remains the first-line treatment for advanced esophageal cancer. However, durable benefits are achieved by only a limited subset of individuals due to the elusive chemoresistance. Here, we utilize patient-derived xenografts (PDXs) from esophageal squamous-cell carcinoma to investigate chemoresistance mechanisms in preclinical settings. We observe that activated cancer-associated fibroblasts (CAFs) are enriched in the tumor microenvironment of PDXs resistant to chemotherapy. Mechanistically, we reveal that cancer-cell-derived S100A8 triggers the intracellular RhoA-ROCK-MLC2-MRTF-A pathway by binding to the CD147 receptor of CAFs, inducing CAF polarization and leading to chemoresistance. Therapeutically, we demonstrate that blocking the S100A8-CD147 pathway can improve chemotherapy efficiency. Prognostically, we found the S100A8 levels in peripheral blood can serve as an indicator of chemotherapy responsiveness. Collectively, our study offers a comprehensive understanding of the molecular mechanisms underlying chemoresistance in esophageal cancer and highlights the potential value of S100A8 in the clinical management of esophageal cancer.
    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
    患者来源的异种移植物(PDX)在癌症研究中的使用正在增加,因为它们能够紧密地模拟患者肿瘤的特征。快速且稳健地测量这些组织中的蛋白质表达水平的能力是广泛的实验设计中所需的关键方法。Westernblotting(WB)是一种经济有效且简单的工具,对于检测和定量单个蛋白质具有高度特异性和敏感性。翻译后修饰和异常信号通路。这里,我们描述了一种使用WB检测参与细胞生长信号通路的蛋白质来评估PDX组织中蛋白质表达的方法.
    The use of patient-derived xenografts (PDXs) in cancer research is increasing due to their ability to closely mimic the features of patient tumors. The ability to quickly and robustly measure protein expression levels in these tissues is a key methodology required in a broad range of experimental designs. Western blotting (WB) is a cost effective and simple tool that is highly specific and sensitive for detecting and quantifying individual proteins, posttranslational modifications and aberrant signaling pathways. Here, we described a method to assess protein expression in PDX tissues using WB to detect proteins involved in cell growth signaling pathways.
    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:为了追求为肺癌患者创造个性化和更有效的治疗策略,患者来源的异种移植物(PDX)已被引入作为临床前平台,可以在体内模型中概括特定患者的肿瘤。我们研究了PDX模型在不同世代中如何很好地保留肿瘤的临床和分子特征。
    方法:在NSG-SGM3小鼠中建立非小细胞肺癌(NSCLC)PDX模型,并在随后的传代中评估临床和临床前因素。我们的队列包括40例NSCLC患者,用于在NSG-SGM3小鼠中创建20个患者特异性PDX模型。进行组织病理学染色和全外显子组测序(WES)分析以了解整个连续传代的肿瘤异质性。
    结果:导致小鼠移植PDX生长的主要因素是较高的疾病等级或阶段,与长期的化疗相反,与PDX传播呈负相关。成功的PDX生长也与较差的预后和总体生存率有关。虽然生长模式变异性受到肿瘤侵袭性的影响,主要影响第一段。病理分析显示保留了组织学类型和等级;然而,WES分析揭示了高级通道中的基因组不稳定性,导致PDX和活检之间的临床相关改变不一致。
    结论:我们的研究强调了多种临床和临床前因素对植入成功的影响,生长动力学,和患者特异性NSCLCPDX的肿瘤稳定性,并强调了在这些模型中指导和评估NSCLC患者的长期个性化治疗研究时考虑这些因素的重要性,以及表明必须进行其他研究以确定该技术的全部临床潜力。
    OBJECTIVE: In the pursuit of creating personalized and more effective treatment strategies for lung cancer patients, Patient-Derived Xenografts (PDXs) have been introduced as preclinical platforms that can recapitulate the specific patient\'s tumor in an in vivo model. We investigated how well PDX models can preserve the tumor\'s clinical and molecular characteristics across different generations.
    METHODS: A Non-Small Cell Lung Cancer (NSCLC) PDX model was established in NSG-SGM3 mice and clinical and preclinical factors were assessed throughout subsequent passages. Our cohort consisted of 40 NSCLC patients, which were used to create 20 patient-specific PDX models in NSG-SGM3 mice. Histopathological staining and Whole Exome Sequencing (WES) analysis were preformed to understand tumor heterogeneity throughout serial passages.
    RESULTS: The main factors that contributed to the growth of the engrafted PDX in mice were a higher grade or stage of disease, in contrast to the long duration of chemotherapy treatment, which was negatively correlated with PDX propagation. Successful PDX growth was also linked to poorer prognosis and overall survival, while growth pattern variability was affected by the tumor aggressiveness, primarily affecting the first passage. Pathology analysis showed preservation of the histological type and grade; however, WES analysis revealed genomic instability in advanced passages, leading to the inconsistencies in clinically relevant alterations between the PDXs and biopsies.
    CONCLUSIONS: Our study highlights the impact of multiple clinical and preclinical factors on the engraftment success, growth kinetics, and tumor stability of patient-specific NSCLC PDXs, and underscores the importance of considering these factors when guiding and evaluating prolonged personalized treatment studies for NSCLC patients in these models, as well as signaling the imperative for additional investigations to determine the full clinical potential of this technique.
    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