关键词: Germ cell tumour Non-gestational choriocarcinoma Ovarian cancer Patient-derived xenograft model RNA sequencing

Mesh : Female Humans Mice Animals Xenograft Model Antitumor Assays Cisplatin Methotrexate Heterografts Mice, Nude Choriocarcinoma, Non-gestational Mice, Inbred NOD Disease Models, Animal Chorionic Gonadotropin Mice, SCID Tumor Microenvironment

来  源:   DOI:10.1186/s12885-023-11626-3   PDF(Pubmed)

Abstract:
BACKGROUND: Non-gestational choriocarcinoma (NGC) is a rare subtype of malignant germ cell tumour and there is no consensus on its treatment. The lack of suitable preclinical models for NGC is a challenge in drug discovery research. Patient-derived xenograft (PDX) models recapitulate the tumour microenvironment of the original cancer tissue. Therefore, they have received considerable attention for studies on rare cancer. Here, we aimed to establish a PDX model from a patient with recurrent NGC.
METHODS: Fresh NGC tumour tissue was immediately transplanted into a severely immune-deficient mouse (NOD.Cg-Prkdcscid1l2rgtm1Wjl/SzJ) and maintained for more than three in vivo passages. Subsequently, we evaluated the molecular characteristics of the PDX model using immunohistochemistry, polymerase chain reaction, and RNA sequencing. Moreover, the PDX tumours were transplanted into BALB/c nude mice, and we evaluated their sensitivity for cisplatin and methotrexate.
RESULTS: The PDX tumour maintained the morphological features of NGC. Moreover, Immunohistochemistry revealed that the human chorionic gonadotropin, cytokeratin 7, and EpCAM expression levels were similar to those in the primary tumour. Furthermore, serum human chorionic gonadotropin levels were elevated in both the primary tumour and the PDX models. Additionally, using PCR analysis with species-specific primers, we confirmed that the PDX tumour contained human genes and was derived from human tissue. Moreover, the gene expression profile of the NGC was compared with that of epithelial ovarian cancer samples and cell lines, and 568 dysregulated genes in the NGC were extracted. The expression of the dysregulated genes in PDX was significantly correlated with that in the primary tumour (R2 = 0.873, P < 0.001). Finally, we demonstrated that the PDX tumour was sensitive to cisplatin and methotrexate; therefore, its clinical response to the agents was similar to that of the primary tumour.
CONCLUSIONS: We successfully established a PDX model of NGC, to the best of our knowledge, for the first time. The established PDX retained the molecular and transcriptome characteristics of the primary tumour and can be used to predict drug effects. It may facilitate further research and the development of novel therapeutic agents for NGC.
摘要:
背景:非妊娠绒毛膜癌(NGC)是一种罕见的恶性生殖细胞肿瘤亚型,对其治疗尚无共识。缺乏合适的NGC临床前模型是药物发现研究中的一个挑战。患者来源的异种移植物(PDX)模型概括了原始癌组织的肿瘤微环境。因此,他们在罕见癌症的研究中受到了相当大的关注。这里,我们旨在建立一个复发性NGC患者的PDX模型。
方法:将新鲜的NGC肿瘤组织立即移植到严重免疫缺陷小鼠中(NOD。Cg-Prkdcscid1l2rgtm1Wjl/SzJ)并维持超过三次体内传代。随后,我们使用免疫组织化学评估了PDX模型的分子特征,聚合酶链反应,和RNA测序。此外,将PDX肿瘤移植到BALB/c裸鼠中,我们评估了它们对顺铂和甲氨蝶呤的敏感性。
结果:PDX肿瘤保持了NGC的形态学特征。此外,免疫组织化学显示,人绒毛膜促性腺激素,细胞角蛋白7和EpCAM表达水平与原发性肿瘤相似。此外,血清人绒毛膜促性腺激素水平在原发性肿瘤和PDX模型中均升高。此外,使用物种特异性引物进行PCR分析,我们证实PDX肿瘤含有人类基因,来源于人体组织。此外,将NGC的基因表达谱与上皮性卵巢癌样品和细胞系进行比较,并提取了NGC中的568个失调基因。PDX中失调基因的表达与原发性肿瘤中的表达显着相关(R2=0.873,P<0.001)。最后,我们证明PDX肿瘤对顺铂和甲氨蝶呤敏感;因此,它对这些药物的临床反应与原发性肿瘤相似。
结论:我们成功建立了NGC的PDX模型,据我们所知,第一次。建立的PDX保留了原发性肿瘤的分子和转录组特征,可用于预测药物作用。它可能有助于进一步研究和开发新的NGC治疗剂。
公众号