Neuroendocrine differentiation

神经内分泌分化
  • 文章类型: Journal Article
    高分化神经内分泌肿瘤(NET)和低分化神经内分泌癌(NEC)是具有不同生物学行为的独特实体。然而,在一些器官中已经报道了表现出模棱两可的形态的疑难病例。在这里,我们报告了一例原发性肝神经内分泌肿瘤(NEN),其组织病理学特征不明确,并伴有常规肝细胞癌(HCC)。由于两个偶然的肝脏肿块,一名未经治疗的70岁慢性乙型肝炎患者接受了左内侧部分切除术。在病理检查中,切除的肿瘤之一具有混合的NEN和HCC成分。NEN成分由在小梁中增殖的相对均匀的肿瘤细胞组成,绳状,或具有外围核栅栏的固体图案。肿瘤细胞对突触素免疫阳性,嗜铬粒蛋白A,分化簇56(CD56),和局灶性肝细胞石蜡1.p53显示野生型表达。热点地区Ki-67标记指数为27%。手术后11个月,他死于脑出血,没有复发肝癌的证据。中等程度的分化和适度的增殖活性可以挑战NEC和NETG3之间的区别。虽然共存的HCC从发病的角度来看是NEC而不是NET,这种模棱两可的肿瘤可能不如典型的NEC具有侵袭性。
    Well-differentiated neuroendocrine tumor (NET) and poorly differentiated neuroendocrine carcinoma (NEC) are distinct entities with different biological behavior. However, difficult cases showing equivocal morphology have been reported in some organs. Herein, we report a case of primary hepatic neuroendocrine neoplasm (NEN) with ambiguous histopathological features admixed with conventional hepatocellular carcinoma (HCC). A 70-year-old man with untreated chronic hepatitis B underwent left medial sectionectomy because of two incidental liver masses. On pathological examination, one of the resected tumors had intermingling NEN and HCC components. The NEN component consisted of relatively uniform tumor cells proliferating in trabecular, cord-like, or solid patterns with peripheral nuclear palisading. The tumor cells were immunopositive for synaptophysin, chromogranin A, cluster of differentiation 56 (CD56), and focally hepatocyte paraffin 1. p53 showed wild-type expression. The Ki-67 labeling index was 27% at the hot spot. Eleven months after the surgery, he died of a cerebral hemorrhage without evidence of recurrent liver cancer. The intermediate degree of differentiation and the modest proliferative activity can challenge the distinction between NEC and NET G3. While the coexisting HCC indicates NEC rather than NET in a pathogenetic viewpoint, such ambiguous tumor may not be as aggressive as typical NECs.
    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:显示神经内分泌(NE)分化的特定类型的前列腺癌(PC)被称为NEPC。NEPC对雄激素剥夺治疗几乎没有反应,并且与转移性去势抗性PC(CRPC)的发展有关,预后极差。我们对NEPC中的遗传驱动因素和激活途径的理解是有限的,这阻碍了精准医学的方法。已知L1细胞粘附分子(L1CAM)在转移性癌症中起致癌作用,包括CRPC。然而,L1CAM对NEPC进展的影响仍然难以捉摸。
    方法:使用PC队列和患者来源的异种移植模型的公共基因表达数据库研究了L1CAM表达水平。在不同的PC细胞中进行L1CAM敲低以研究体外细胞功能。长期暴露于阿比特龙诱导NE分化后,建立了CRPC细胞系CWR22Rv1的亚系。在肿瘤球体形成条件下培养雄激素受体阴性细胞系PC3以丰富癌症干性特征。在PC细胞上测试了几种氧化应激诱导剂,以观察L1CAM介导的基因表达和细胞死亡。
    结果:与CRPC或腺癌肿瘤相比,L1CAM在NEPC中的表达明显较高。L1CAM还与NE标记表达相关,并与基因表达数据库和NE分化的CRPC细胞中腺癌到NEPC的进展相关。L1CAM还促进PC3细胞在癌干性富集下的癌干性和NE表型。L1CAM也被鉴定为活性氧诱导基因,通过这种方法,L1CAM可以抵消由电离辐射引发的CRPC细胞死亡。
    结论:我们的结果揭示了L1CAM在PC中NE表型获得中的新作用,有助于CRPC的NE分化相关治疗抗性。
    BACKGROUND: A specific type of prostate cancer (PC) that exhibits neuroendocrine (NE) differentiation is known as NEPC. NEPC has little to no response to androgen deprivation therapy and is associated with the development of metastatic castration-resistant PC (CRPC), which has an extremely poor prognosis. Our understanding of genetic drivers and activated pathways in NEPC is limited, which hinders precision medicine approaches. L1 cell adhesion molecule (L1CAM) is known to play an oncogenic role in metastatic cancers, including CRPC. However, the impact of L1CAM on NEPC progression remains elusive.
    METHODS: L1CAM expression level was investigated using public gene expression databases of PC cohorts and patient-derived xenograft models. L1CAM knockdown was performed in different PC cells to study in vitro cell functions. A subline of CRPC cell line CWR22Rv1 was established after long-term exposure to abiraterone to induce NE differentiation. The androgen receptor-negative cell line PC3 was cultured under the tumor sphere-forming condition to enrich cancer stemness features. Several oxidative stress inducers were tested on PC cells to observe L1CAM-mediated gene expression and cell death.
    RESULTS: L1CAM expression was remarkably high in NEPC compared to CRPC or adenocarcinoma tumors. L1CAM was also correlated with NE marker expressions and associated with the adenocarcinoma-to-NEPC progression in gene expression databases and CRPC cells with NE differentiation. L1CAM also promoted cancer stemness and NE phenotypes in PC3 cells under cancer stemness enrichment. L1CAM was also identified as a reactive oxygen species-induced gene, by which L1CAM counteracted CRPC cell death triggered by ionizing radiation.
    CONCLUSIONS: Our results unveiled a new role of L1CAM in the acquisition of the NE phenotype in PC, contributing to the NE differentiation-related therapeutic resistance of CRPC.
    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
    肺神经内分泌肿瘤(NENs)是一类以神经内分泌(NE)分化为特征的多样化肿瘤。在肺NENs中,肺类癌(LC)是一种罕见的肿瘤,具有独特的特征。最近的研究强调了转录因子(TFs)在建立肺NENs如小细胞肺癌基因表达程序中的重要性。然而,控制LC基因表达的TFs在很大程度上是未知的。在这项研究中,我们报告了称为Prospero同源盒蛋白1(PROX1)的TF在LC中的表达和潜在功能。公开可用的转录组数据表明PROX1在LC组织中高度表达,通过组织微阵列上的免疫组织化学分析证实了这一点。PROX1的敲低不影响LC衍生细胞系的细胞活力,NCI-H727。同时,转录组分析显示,PROX1敲低改变了NE分化相关基因的表达。ASCL1,CHGA,CALCA,和LINC00261被认为是PROX1的下游基因。这些发现表明,PROX1可能通过调节关键靶基因的表达在LC的NE身份中起重要作用。
    Lung neuroendocrine neoplasms (NENs) are a diverse group of tumors characterized by neuroendocrine (NE) differentiation. Among lung NENs, lung carcinoid (LC) is a rare tumor with unique characteristics. Recent research has highlighted the importance of transcription factors (TFs) in establishing gene expression programs in lung NENs such as small cell lung carcinoma. However, the TFs that control the gene expression of LC are largely unknown. In this study, we report the expression and potential function of a TF called Prospero homeobox protein1 (PROX1) in LC. Publicly available transcriptome data suggested that PROX1 was highly expressed in LC tissues, which was confirmed by immunohistochemical analysis on a tissue microarray. Knockdown of PROX1 did not impact the cellular viability of an LC-derived cell line, NCI-H727. Meanwhile, transcriptome analysis revealed that PROX1 knockdown altered the expression of genes involved in NE differentiation. ASCL1, CHGA, CALCA, and LINC00261 were suggested as downstream genes of PROX1. These findings indicate that PROX1 may play an important role in the NE identity of LC by regulating the expression of key target genes.
    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
    膀胱癌本质上是异质性的,显示不同的分子谱和组织病理学特征,这给诊断和治疗带来了挑战。然而,理解这种异质性的分子基础仍然难以捉摸。本研究旨在阐明神经内分泌样膀胱肿瘤的分子景观,重点关注β-catenin定位的参与。分析转录组数据并受益于分子分类工具,我们对肌层浸润性膀胱癌进行了深入分析,以揭示神经内分泌样分化的分子特征.该研究探讨了转录因子和染色质重塑复合物对膀胱癌神经内分泌分化的贡献。这项研究揭示了肌层浸润性膀胱肿瘤中β-catenin定位和神经内分泌分化之间的显著相关性。强调神经内分泌样肿瘤的分子复杂性。YY1转录因子的富集,E2F家族成员,β-catenin阳性肿瘤中的Polycomb阻抑复杂成分表明它们对神经内分泌表型的潜在贡献。我们的发现为神经内分泌样膀胱肿瘤的分子复杂性提供了有价值的见解。通过识别潜在的治疗靶点和完善诊断策略,这项研究促进了我们对膀胱癌内分泌学的理解.需要进一步研究这些分子关系的功能含义,以增强我们的知识并指导未来的治疗干预。
    Bladder cancers are heterogeneous in nature, showing diverse molecular profiles and histopathological characteristics, which pose challenges for diagnosis and treatment. However, understanding the molecular basis of such heterogeneity has remained elusive. This study aimed to elucidate the molecular landscape of neuroendocrine-like bladder tumors, focusing on the involvement of β-catenin localization. Analyzing the transcriptome data and benefiting from the molecular classification tool, we undertook an in-depth analysis of muscle-invasive bladder cancers to uncover the molecular characteristics of the neuroendocrine-like differentiation. The study explored the contribution of transcription factors and chromatin remodeling complexes to neuroendocrine differentiation in bladder cancer. The study revealed a significant correlation between β-catenin localization and neuroendocrine differentiation in muscle-invasive bladder tumors, highlighting the molecular complexity of neuroendocrine-like tumors. Enrichment of YY1 transcription factor, E2F family members, and Polycomb repressive complex components in β-catenin-positive tumors suggest their potential contribution to neuroendocrine phenotypes. Our findings contribute valuable insights into the molecular complexity of neuroendocrine-like bladder tumors. By identifying potential therapeutic targets and refining diagnostic strategies, this study advances our understanding of endocrinology in the context of bladder cancer. Further investigations into the functional implications of these molecular relationships are warranted to enhance our knowledge and guide future therapeutic interventions.
    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
    一名71岁的激素敏感型前列腺癌骨转移患者接受了雄激素剥夺疗法和阿帕鲁胺治疗。镭-223和放射治疗在其变得去势抵抗后进行。尽管前列腺特异性抗原水平仍然很低,观察到神经内分泌前列腺癌的多个皮下转移。对治疗前前列腺穿刺活检的回顾显示,一小部分具有提示神经内分泌分化的特征。观察到磷酸酶和tensine同源物的丢失和肿瘤蛋白p53的过度表达,确认侵袭性变异型前列腺癌的诊断。给予以铂为基础的化疗;然而,患者在诊断后28个月死亡.在这种情况下,如果侵袭性变异型前列腺癌的诊断是在较早的时间通过活检标本做出的,较早引入以铂类药物为基础的治疗方案可能会改善预后.
    在线版本包含补充材料,可在10.1007/s13691-024-00673-7获得。
    A 71-year-old man with bone metastasis of hormone-sensitive prostate cancer was treated with androgen deprivation therapy and apalutamide. Radium-223 and radiation therapy were administered after it become castration resistant. Although prostate-specific antigen levels remained low, multiple subcutaneous metastases of neuroendocrine prostate cancer were observed. A review of the pre-treatment prostate needle biopsy revealed a small component with features suggestive of neuroendocrine differentiation. Phosphatase and tensine homolog loss and tumor protein p53 overexpression were observed, confirming the diagnosis of aggressive variant prostate cancer. Platinum-based chemotherapy was administered; however, the patient died 28 months after diagnosis. In this case, if the diagnosis of aggressive variant prostate cancer had been made at an earlier time by biopsy specimens, there might have been a possibility to improve the prognosis by the earlier introduction of the platinum-based regimen.
    UNASSIGNED: The online version contains supplementary material available at 10.1007/s13691-024-00673-7.
    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(PCa)的肿瘤微环境(TME)的特征是高水平的免疫抑制分子,包括细胞因子和趋化因子。这产生了阻碍有效免疫反应的敌对免疫景观。白细胞介素-1(IL-1)受体拮抗剂(IL1RN),一个关键的抗炎分子,在抑制IL-1相关的免疫和炎症反应中起重要作用。我们的研究调查了IL1RN在PCa中的致癌作用,特别是它与毒蕈碱乙酰胆碱受体4(CHRM4)的相互作用,及其参与驱动PCaTME内的免疫抑制途径和M2样巨噬细胞极化。我们证明在雄激素剥夺治疗(ADT)之后,PCa中IL1RN-CHRM4相互作用激活MAPK/AKT信号通路。这种激活上调转录因子E2F1和MYCN,刺激IL1RN的产生,并创建一个正反馈回路,增加PCa细胞和M2样巨噬细胞中CHRM4的丰度。这种ADT驱动的IL1RN/CHRM4轴显着增强与神经内分泌分化和治疗抗性结果相关的免疫检查点标记。在晚期PCa患者中,较高的血清IL1RN水平与疾病侵袭性和M2样巨噬细胞标志物增加相关。此外,IL1RN水平升高与免疫治疗后更好的临床结局相关.在晚期PCa患者和神经内分泌PCa类器官模型中IL1RN和CHRM4表达之间的临床相关性突出了它们作为治疗靶标的潜力。我们的数据表明,靶向IL1RN/CHRM4信号可能是管理PCa进展和增强治疗反应的有希望的策略。
    The tumor microenvironment (TME) of prostate cancer (PCa) is characterized by high levels of immunosuppressive molecules, including cytokines and chemokines. This creates a hostile immune landscape that impedes effective immune responses. The interleukin-1 (IL-1) receptor antagonist (IL1RN), a key anti-inflammatory molecule, plays a significant role in suppressing IL-1-related immune and inflammatory responses. Our research investigates the oncogenic role of IL1RN in PCa, particularly its interactions with muscarinic acetylcholine receptor 4 (CHRM4), and its involvement in driving immunosuppressive pathways and M2-like macrophage polarization within the PCa TME. We demonstrate that following androgen deprivation therapy (ADT), the IL1RN-CHRM4 interaction in PCa activates the MAPK/AKT signaling pathway. This activation upregulates the transcription factors E2F1 and MYCN, stimulating IL1RN production and creating a positive feedback loop that increases CHRM4 abundance in both PCa cells and M2-like macrophages. This ADT-driven IL1RN/CHRM4 axis significantly enhances immune checkpoint markers associated with neuroendocrine differentiation and treatment-resistant outcomes. Higher serum IL1RN levels are associated with increased disease aggressiveness and M2-like macrophage markers in advanced PCa patients. Additionally, elevated IL1RN levels correlate with better clinical outcomes following immunotherapy. Clinical correlations between IL1RN and CHRM4 expression in advanced PCa patients and neuroendocrine PCa organoid models highlight their potential as therapeutic targets. Our data suggest that targeting the IL1RN/CHRM4 signaling could be a promising strategy for managing PCa progression and enhancing treatment responses.
    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
    雄激素受体信号调节前列腺的正常和病理生长。特别是,前列腺癌细胞的生长和存活最初依赖于雄激素受体信号传导。暴露于雄激素剥夺治疗导致去势抵抗性前列腺癌的发展。在前列腺肿瘤细胞中发生了大量的分子和细胞变化,包括神经内分泌特征和各种生物标志物的表达,这促进了癌细胞向雄激素非依赖性生长的转变。这些生物标志物包括转录因子(TP53,REST,BRN2,INSM1,c-Myc),信号分子(PTEN,极光激酶,视网膜母细胞瘤肿瘤抑制因子,钙结合蛋白),和受体(糖皮质激素,雄激素受体变体7),在其他人中。人们认为,基因改造,治疗,和肿瘤微环境的变化是导致前列腺癌进展的因素,其表型特征具有显着的异质性。然而,目前尚不清楚这些表型特征和分子修饰是如何在特定治疗条件下产生的。在这项工作中,我们总结了与前列腺癌进展相关的一些最重要的分子变化,并描述了这些细胞过程中涉及的一些因素。
    Androgen receptor signaling regulates the normal and pathological growth of the prostate. In particular, the growth and survival of prostate cancer cells is initially dependent on androgen receptor signaling. Exposure to androgen deprivation therapy leads to the development of castration-resistant prostate cancer. There is a multitude of molecular and cellular changes that occur in prostate tumor cells, including the expression of neuroendocrine features and various biomarkers, which promotes the switch of cancer cells to androgen-independent growth. These biomarkers include transcription factors (TP53, REST, BRN2, INSM1, c-Myc), signaling molecules (PTEN, Aurora kinases, retinoblastoma tumor suppressor, calcium-binding proteins), and receptors (glucocorticoid, androgen receptor-variant 7), among others. It is believed that genetic modifications, therapeutic treatments, and changes in the tumor microenvironment are contributing factors to the progression of prostate cancers with significant heterogeneity in their phenotypic characteristics. However, it is not well understood how these phenotypic characteristics and molecular modifications arise under specific treatment conditions. In this work, we summarize some of the most important molecular changes associated with the progression of prostate cancers and we describe some of the factors involved in these cellular processes.
    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
    肺神经内分泌(NE)细胞是罕见的气道上皮细胞。Achaete-scute复合物同源物1(ASCL1)与Notch信号通路的靶分子之一split1的毛状和增强子之间的平衡,对NE分化至关重要。小细胞肺癌(SCLC)是一种高度侵袭性的肺癌,以快速细胞增殖为特征,高转移潜能,以及对治疗的抵抗力。SCLC的亚型由NE细胞谱系转录因子的表达状态定义。例如ASCL1,其作用由SRY-box2,胰岛素瘤相关蛋白1,NK2同源盒1和无翼相关的整合位点信号支持。该网络增强了NE分化,并可能诱导SCLC的特征性形态和化学敏感性。Notch信号介导细胞命运决定,导致NE到非NE的命运转变。NE分化的抑制可能会将SCLC的组织学类型改变为非SCLC形态。在具有NE分化的SCLC中,Notch信号传导通常是无活性的并且是遗传或表观遗传调节的。然而,Notch信号可能在化疗后被激活,and,与Yes相关蛋白信号和RE1沉默转录因子一致,抑制NE分化,产生肿瘤内异质性和化学耐药性。关于SCLC分子机制的累积信息将有助于控制这种顽固性癌症的进一步进展。
    Pulmonary neuroendocrine (NE) cells are rare airway epithelial cells. The balance between Achaete-scute complex homolog 1 (ASCL1) and hairy and enhancer of split 1, one of the target molecules of the Notch signaling pathway, is crucial for NE differentiation. Small cell lung cancer (SCLC) is a highly aggressive lung tumor, characterized by rapid cell proliferation, a high metastatic potential, and the acquisition of resistance to treatment. The subtypes of SCLC are defined by the expression status of NE cell-lineage transcription factors, such as ASCL1, which roles are supported by SRY-box 2, insulinoma-associated protein 1, NK2 homeobox 1, and wingless-related integration site signaling. This network reinforces NE differentiation and may induce the characteristic morphology and chemosensitivity of SCLC. Notch signaling mediates cell-fate decisions, resulting in an NE to non-NE fate switch. The suppression of NE differentiation may change the histological type of SCLC to a non-SCLC morphology. In SCLC with NE differentiation, Notch signaling is typically inactive and genetically or epigenetically regulated. However, Notch signaling may be activated after chemotherapy, and, in concert with Yes-associated protein signaling and RE1-silencing transcription factor, suppresses NE differentiation, producing intratumor heterogeneity and chemoresistance. Accumulated information on the molecular mechanisms of SCLC will contribute to further advances in the control of this recalcitrant cancer.
    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
    前列腺癌是一种异质性疾病,具有广泛的病理特征,临床,和分子特征。随着新数据的结合,前列腺癌的诊断和分类一直在不断进行修改。世界卫生组织(WHO)第5版《泌尿和生殖器肿瘤分类》在第4版之后的6年最近出版。在这个新版本中,前列腺癌的分类在诊断标准中得到了完善,分级,命名法,和基因组学。本文回顾了WHO前列腺癌新分类的重要更新,包括高级别前列腺上皮内瘤,腺泡腺癌,导管内癌,导管癌,和神经内分泌肿瘤。讨论了格里森分级中的有争议的问题,如导管内癌和三级癌。我们还强调了前列腺癌中不同的遗传和表观遗传改变,这可能有助于其不同的临床病理特征。总的来说,世界卫生组织第5版的分类提供了一个全面的评估前列腺癌的形态学,免疫组织化学,基因组,和临床数据,这可能是诊断和治疗前列腺癌的最佳范例。
    Prostate cancer is a heterogeneous disease with a wide spectrum of pathological, clinical, and molecular features. The diagnosis and classification of prostate cancer have been constantly modified with the incorporation of new data. The 5th edition of the World Health Organization (WHO) Classification of Urinary and Genital Tumors was recently published six years after the 4th edition. In this new edition, the classification of prostate cancer has been refined in the diagnostic criteria, grading, nomenclature, and genomics. This paper reviews significant updates to the new WHO classification of prostate cancer, including high-grade prostatic intraepithelial neoplasia, acinar adenocarcinoma, intraductal carcinoma, ductal carcinoma, and neuroendocrine tumors. Controversial issues in the Gleason grading are discussed, such as intraductal carcinoma and tertiary grade. We also highlight distinct genetic and epigenetic alterations in prostate cancer that may contribute to its diverse clinicopathologic features. Overall, the 5th edition of the WHO classification provides a comprehensive assessment of prostate cancer with morphologic, immunohistochemical, genomic, and clinical data, which may represent an optimal paradigm for diagnosing and treating prostate 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
    上皮性卵巢癌(EOC)是最致命的妇科恶性肿瘤,预后和患者生存率较差。尽管开发了几种治疗干预措施,如聚ADP核糖聚合酶(PARP)抑制剂,EOC仍然难以管理,并且非常有必要发现新的早期检测生物标志物和治疗靶标。尽管神经内分泌分化(NED)涉及不同的人类癌症,包括前列腺癌和肺癌,缺乏关于上皮性卵巢癌NED的机制研究。我们报道Aurora激酶A以ERK1/2依赖性方式驱动上皮性卵巢癌的NED,Aurora激酶A的药理和遗传抑制抑制卵巢癌的NED。此外,我们证明蛋白激酶D2正调节极光激酶A驱动NED。催化活性PKD2的过表达驱动NED和集体,PKD2与Aurora激酶A/ERK1/2信号轴交叉对话以积极调节EOC的NED。PKD2/Aurora激酶A/ERK1/2信号轴是针对神经内分泌分化型EOC的新型治疗靶标。
    Epithelial ovarian cancer (EOC) is deadliest gynecological malignancy with poor prognosis and patient survival. Despite development of several therapeutic interventions such as poly-ADP ribose polymerase (PARP) inhibitors, EOC remains unmanageable and discovery of novel early detection biomarkers and treatment targets are highly warranted. Although neuroendocrine differentiation (NED) is implicated in different human cancers including prostate adenocarcinoma and lung cancer, mechanistic studies concerning NED of epithelial ovarian cancer are lacking. We report that Aurora kinase A drives NED of epithelial ovarian cancer in an ERK1/2-dependent manner and pharmacological and genetic inhibition of Aurora kinase A suppress NED of ovarian cancer. Moreover, we demonstrate that protein kinase D2 positively regulated Aurora kinase A to drive NED. Overexpression of catalytically active PKD2 drives NED and collectively, PKD2 cross talks with Aurora kinase A/ERK1/2 signalling axis to positively regulate NED of EOC. PKD2/Aurora kinase A/ERK1/2 signalling axis is a novel therapeutic target against neuroendocrine differentiated EOC.
    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