国际妇产科学杂志, 2022, 49(3): 286-290 doi: 10.12280/gjfckx.20220089

妇科肿瘤研究:综述

CCNE1基因在卵巢高级别浆液性癌中的研究进展

刘艳, 卞伟, 肖虹,

030001 太原,山西医科大学第一临床医学院(刘艳);山西医科大学基础医学院(卞伟);山西医科大学第一医院病理科(肖虹)

LIU Yan, BIAN Wei, XIAO Hong

LIU Yan, BIAN Wei, XIAO Hong,

The First Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China (LIU Yan);School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China (BIAN Wei);Department of Pathology, The First Hospital of Shanxi Medical University, Taiyuan 030001, China (XIAO Hong)

通讯作者: 肖虹,E-mail:xiaohh9999@163.com

审校者

责任编辑: 秦娟

收稿日期: 2022-01-30  

基金资助: 山西省自然科学基金(201901D111210)

Corresponding authors: xiaohh9999@163.com

Received: 2022-01-30  

摘要

细胞周期蛋白E1(Cyclin E1,CCNE1)基因扩增是卵巢高级别浆液性癌(high-grade serous ovarian carcinoma,HGSOC)最常见的拷贝数变异之一,扩增率为20%左右。CCNE1基因扩增不仅参与HGSOC的早期癌前病变,促进恶性肿瘤的发生、发展,还可引起细胞周期紊乱和染色体不稳定,从而增加HGSOC的铂类耐药性和复发率,最终导致患者的化疗效果不佳和预后较差。为改善CCNE1扩增的HGSOC患者的预后和生活质量,在暂时没有CCNE1扩增的HGSOC的靶向治疗药物的情况下,使用细胞周期蛋白依赖性激酶2(cyclin-dependent kinases 2,CDK2)小分子抑制剂或者多腺苷二磷酸核糖聚合酶[poly (ADP-ribose) polymerase,PARP]抑制剂与其他药物联合治疗,一定程度上能有效地抑制肿瘤细胞的增殖活性,促进肿瘤细胞死亡。因此,从CCNE1扩增对HGSOC的早期发病机制的影响和产生铂类耐药的角度上来说,CCNE1基因有望成为HGSOC的潜在治疗靶标和预后评价生物学指标,在改善患者的治疗和预后方面具有重要意义。

关键词: 卵巢肿瘤; 细胞周期蛋白E; 基因扩增; 药物疗法; 预后; 卵巢高级别浆液性癌

Abstract

CyclinE1(CCNE1) gene amplification is one of the most common copy number variations in high-grade serous ovarian carcinoma (HGSOC), with an amplification rate of about 20%. CCNE1 gene amplification not only participates in the early precancerous lesions of HGSOC and promotes the occurrence and development of malignant tumors, but also causes cell cycle disorder and chromosome instability, thus increasing platinum resistance and recurrence rate of HGSOC, and ultimately leading to poor chemotherapy effect and poor prognosis of patients. In order to improve the prognosis and quality of life of patients with CCNE1-amplified HGSOC, cyclin-dependent kinases 2(CDK2) small molecule inhibitors or poly (ADP-ribose) polymerase (PARP) inhibitors in combination with other drugs are used to treat CCNE1-amplified HGSOC in the absence of targeted drugs for CCNE1-amplified HGSOC. To a certain extent, it can effectively inhibit the proliferation of tumor cells and promote the death of tumor cells. Therefore, CCNE1 gene is expected to become a potential therapeutic target and prognostic biological indicator of HGSOC from the perspective of the influence of CCNE1 amplification on the early pathogenesis of HGSOC and the generation of platinum resistance, which is of great significance to improve the treatment and prognosis of patients.

Keywords: Ovarian neoplasms; Cyclin E; Gene amplification; Drug therapy; Prognosis; High-grade serous ovarian carcinoma

PDF (700KB) 元数据 多维度评价 相关文章 导出 EndNote| Ris| Bibtex  收藏本文

本文引用格式

刘艳, 卞伟, 肖虹. CCNE1基因在卵巢高级别浆液性癌中的研究进展[J]. 国际妇产科学杂志, 2022, 49(3): 286-290 doi:10.12280/gjfckx.20220089

LIU Yan, BIAN Wei, XIAO Hong. LIU Yan, BIAN Wei, XIAO Hong[J]. Journal of International Obstetrics and Gynecology, 2022, 49(3): 286-290 doi:10.12280/gjfckx.20220089

卵巢高级别浆液性癌(high-grade serous ovarian carcinoma,HGSOC)是上皮性卵巢癌(epithelial ovarian cancer,EOC)最常见的组织学亚型之一,约占所有EOC的75%,具有基因组不稳定性、高度侵袭性和预后较差等特征[1]。目前,HGSOC的标准治疗方式是手术切除后接受铂类联合紫杉烷类药物化疗,但80%的患者易出现复发,约40%~60%的患者对化疗敏感,25%的患者会对铂类产生耐药性[2]。可见,这些化疗药物对部分HGSOC患者具有一定的敏感性,但大多数HGSOC患者经化疗后易出现复发,这在一定程度上影响了HGSOC患者的生活质量。因此,为了提高HGSOC的诊断水平和改善HGSOC患者的生活质量,亟需探索和开发具有临床应用价值的治疗靶标。

细胞周期蛋白E1(Cyclin E1,CCNE1)基因扩增是HGSOC最常见的基因拷贝数变异之一,约20%的HGSOC患者存在CCNE1扩增[3]CCNE1基因常在卵巢癌[4]、子宫内膜癌[5]、骨肉瘤[6]和乳腺癌[7]等恶性肿瘤中发生扩增,扩增率为2%~40%。同时,CCNE1基因扩增不仅是这些恶性肿瘤的致癌和预后不良的因素,还与HGSOC耐药密切相关。因此,现就CCNE1基因与HGSOC发生、发展、耐药和预后不良的关系以及HGSOC的靶向治疗药物等方面进行综述。

1 CCNE1扩增与HGSOC发生、发展的关系

HGSOC是一种高度异质性和基因组不稳定的疾病,约20%的HGSOC出现CCNE1扩增,CCNE1扩增对HGSOC的发生、发展具有重要意义[3,8]。近年研究表明HGSOC大多起源于输卵管上皮分泌型细胞,而不是卵巢表面上皮[9-10]。输卵管上皮分泌型细胞对DNA损伤较为敏感,当DNA受到各种毒性因素刺激时,分泌型上皮细胞无法及时修复DNA,会造成大量DNA损伤蓄积,使细胞发生TP53基因突变,促进输卵管上皮分泌型细胞异型增生,逐渐形成浆液性输卵管上皮内癌(serous tubal intraepithelial carcinoma,STIC),随后癌细胞直接种植到卵巢表面后进展成HGSOC。TP53突变是发生癌变的一个初始事件,参与肿瘤的发生、发展,但只是TP53突变不足以使细胞恶变,还需要其他分子生物学的改变来共同促使早期STIC发展至HGSOC。CCNE1基因和TP53基因之间在体内外实验中都存在协同致癌作用,CCNE1基因去调控后会使TP53蛋白缺陷的输卵管分泌型上皮细胞获得恶性表型,从而促进肿瘤不受控制地生长[11]。然而,CCNE1扩增一般发生在TP53突变的早期,这种早期扩增可能提示HGSOC的癌前病变存在DNA拷贝数异常改变[12]。Kuhn等[13]比较STIC和HGSOC的CCNE1扩增率发现两者无显著差异,这表明发生癌前病变时已出现CCNE1扩增。由此可见,CCNE1扩增对癌前病变发生恶性转化至关重要,是HGSOC发生、发展的重要分子事件,可作为早期筛查的潜在靶点。

2 CCNE1扩增与HGSOC耐药的关系

2.1 细胞周期

细胞周期是指细胞从一次分裂完成后开始至下一次分裂结束的全过程。正常细胞周期是由细胞周期蛋白和CDKs构成,共同完成细胞分裂与增殖[14]。CCNE1基因编码的Cyclin E1是视网膜母细胞瘤(retinoblastoma,RB)蛋白抑制通路的关键调节因子,主要参与细胞周期的调控。Cyclin E1主要通过打开三磷酸腺苷结合域,促使三磷酸腺苷和底物进入CDKs活性位点,与CDKs家族成员中亲和力最高的CDK2结合后形成Cyclin E1/CDK2复合体,并磷酸化下游RB靶点使RB蛋白失活,从而释放出早期转录因子2(early 2 transcription factor,E2F),使细胞从G1期过渡到S期,促进细胞进行正常的DNA复制和细胞增殖[11]。细胞周期紊乱是人类发生恶性肿瘤和产生铂类耐药的原因之一。一般而言,细胞周期蛋白表达量的改变会引起肿瘤细胞异常增殖和分裂,从而导致基因组不稳定和化疗耐药性[15-16]。当CCNE1发生扩增时会促进Cyclin E1表达量增多,加快细胞分裂与增殖的同时可引发DNA不受控制地进行复制,从而破坏胞质分裂稳态,导致肿瘤细胞发生异常增殖和耐药性[17-19];CCNE1扩增的肿瘤不但本身可以增加化疗耐药性,还可以赋予具有高增殖和低凋亡活性的肿瘤细胞一定的生长优势,增强肿瘤细胞的增殖能力,加快有丝分裂,促进肿瘤细胞的生长[20]。如上所述,肿瘤细胞异常增殖与HGSOC铂类耐药性密切相关,也是间接造成HGSOC患者复发和预后不良的原因之一。

2.2 染色体不稳定(chromosome instability)

染色体不稳定是指增加或丢失整个或大部分染色体的速率,是基因组不稳定的主要形式之一。在一定程度上,CCNE1基因拷贝数变异意味着会发生染色体不稳定性,其中Cyclin E1/CDK2复合体与染色体不稳定密切相关。Cyclin E1/CDK2复合体主要通过2种方式来调控染色体不稳定:①参与中心体扩增。Cyclin E1/CDK2复合体的激活与中心体复制有关,主要通过磷酸化核磷酸蛋白(nucleophosmin,NPM),促使NPM从中心体内释放,启动中心体复制;但当NPM无法进行去磷酸化时,会形成2个以上的中心体,即中心体扩增,这不仅对胞质分裂造成严重干扰,还会导致染色体发生分离错误[21]。②参与有丝分裂G1/S期检查点的损伤。正常情况下,Cyclin E1受转录水平和蛋白酶体活性的严格调控。当Cyclin E1/CDK2复合体变得活跃时,Cyclin E1主要依赖G1后期完成E2F转录和S期泛素介导的蛋白水解。然而,Cyclin E1过表达会引起G1/S期检查点失控,无法激活有丝分裂蛋白水解酶,使细胞延迟进入有丝分裂后期,这可能会导致染色体错配和非整倍性[22]。值得注意的是,染色体不稳定不仅与肿瘤的恶变和异质性有关,还与HGSOC的复发、耐药和预后不良有关。Penner-Goeke等[23]研究发现在PEO4和A2780cp这2种EOC的耐药模型中均出现染色体不稳定,并与EOC的耐药性和复发性有关。最新一项研究也证实了染色体不稳定的存在与促进肿瘤的发生、发展、转移和耐药性有关[24]

3 CCNE1扩增与HGSOC预后不良的关系

鉴于HGSOC常出现铂类耐药和复发等情况,有学者提出CCNE1扩增联合Cyclin E1高表达可作为HGSOC预后不良的预测生物标志物[25-26]。Filippova等[27]研究表明患有HGSOC的年轻女性易发生乳腺癌易感基因1/2(breast cancer 1 and 2,BRCA1/2)突变,而CCNE1扩增常见于HGSOC的中老年女性,并可能与其预后不良有关。CCNE1扩增和同源重组修复基因BRCA1/2突变在HGSOC中是相互排斥的,BRCA1/2突变的HGSOC对铂类药物较敏感,但发生CCNE1扩增的HGSOC对铂类药物易耐药,对多腺苷二磷酸核糖聚合酶[poly (ADP-ribose) polymerase,PARP]抑制剂的反应有限,因此,患有HGSOC的中老年患者接受铂类及PARP抑制剂治疗效果较差。另有研究利用干扰RNA敲低CCNE1基因表达水平后,发现CCNE1基因下调会导致原先Cyclin E1蛋白过表达的OVCAR3和OVCAR5等4种卵巢癌细胞系出现细胞生长率下降65%~90%[28]。此外,CCNE1扩增不仅促进HGSOC细胞的生长,还能评估患者的疗效和预后情况。Nakayama等[29]对接受铂类和紫杉烷类化疗的卵巢癌患者进行研究发现,CCNE1扩增与总生存期缩短、预后不良有关,但Cyclin E1高表达与总体存活率无关。由此可见,Cyclin E1高表达不能作为HGSOC患者新辅助治疗或铂类联合紫杉烷类化疗产生耐药性的预测标志物,并且CCNE1无扩增合并Cyclin E1高表达的HGSOC患者较CCNE1扩增合并Cyclin E1高表达的患者预后较好[3,30]。这是因为CCNE1无扩增合并Cyclin E1高表达的HGSOC患者可能存在同源重组缺陷(homologous recombination deficiency,HRD),所以使用铂类化疗药物治疗较为敏感。总之。HGSOC患者不能以Cyclin E1高表达作为预后指标,但以CCNE1基因为治疗靶点可能有利于改善预后和提高总体生存率。

4 CCNE1扩增与HGSOC的靶向治疗药物

4.1 CDK2小分子抑制剂

CDK2小分子抑制剂主要通过阻碍CDK2与Cyclin E1形成Cyclin E1/CDK2复合体来抑制细胞异常增殖,从而改善HGSOC患者的不良预后和降低其铂类耐药性。CCNE1扩增的HGSOC细胞系对干扰RNA敲低的CDK2具有选择性敏感,提示CDK2可能是CCNE1扩增的HGSOC的潜在治疗药物[31]。Yang等[28]发现CCNE1蛋白过表达的卵巢癌细胞对CDK2抑制剂SNS-032的敏感性是没有Cyclin E1蛋白过表达的卵巢癌细胞的40倍,SNS-032可通过抑制Cyclin E1蛋白过表达的卵巢癌细胞的转移,从而延长Cyclin E1蛋白过表达的卵巢癌小鼠的总体生存期。因此,该研究表明CDK2抑制剂可以有效地抑制Cyclin E1蛋白过表达的卵巢癌细胞的生长。然而,CCNE1扩增的卵巢癌也会对CDK2抑制剂产生耐药性,使其对CDK2抑制剂的敏感性显著降低,这可能存在2种旁路耐药机制:一是CDK2蛋白的上调,二是与选择多倍体细胞有关[32]。目前,考虑到单用CDK2抑制剂治疗CCNE1扩增的卵巢癌的敏感性下降,有学者提出联合其他协同药物来减少耐药性和不良反应。Au-Yeung等[31]通过美国的癌症基因组图谱(The Cancer Genome Atlas,TCGA)证实了CCNE1和蛋白激酶B2(protein kinase B2,AKT2)在HGSOC中存在共扩增,随后利用高通量药物筛选出CDK2抑制剂(Dinaciclib)和AKT抑制剂(MK-2206)具有药物协同增效作用,该研究表明Dinaciclib和MK-2206的新型组合可以选择性靶向治疗CCNE1扩增的HGSOC。由此可见,CDK2抑制剂联合其他协同药物为治疗CCNE1扩增的HGSOC提供了一种崭新思路。

4.2 PARP抑制剂

近年来,已批准的PARP抑制剂药物分别有奥拉帕尼(Olaparib)、尼拉帕尼(Niraparib)和鲁卡帕尼(Rucaparib)。该类药物主要用于铂类化疗后复发性与铂类耐药性的EOC患者的维持治疗[33-34]。一项Meta分析结果表明,PARP抑制剂已被用作晚期HGSOC患者一线治疗后的维持治疗,PARP抑制剂维持治疗可使HGSOC患者的死亡风险降低46%[35]。然而,PARP抑制剂主要是针对治疗BRCA1/2突变的HGSOC具有高度敏感性,但对CCNE1扩增的HGSOC进行长期治疗的效果欠佳,甚至可能会产生耐药。有研究表明抗血管生成单克隆抗体(Bevacizumab)联合PARP抑制剂可降低耐药性,并推荐作为晚期卵巢癌的一线治疗药物[36]。因此,有学者提出PARP抑制剂与抗血管生成药物或其他协同药物联合应用可以有效地提高疗效。Kim等[37]研究表明PARP抑制剂联合共济失调毛细血管扩张症突变和 Rad3相关(ataxia telangiectasia mutated and Rad3-related,ATR)抑制剂(ATRi)能显著地抑制肿瘤快速生长和延长总体生存期。ATRi 联合 PARP抑制剂在新发性或获得性的铂类耐药且发生CCNE1扩增的卵巢癌模型中具有药物协同作用,ATRi(AZD6738)不仅能使HGSOC的肿瘤细胞重新对Olaparib敏感,还能使DNA复制叉停滞和细胞凋亡。最近,Gupta等[38]研究发现Olaparib联合选择性HDAC1/2抑制剂(Entinostat)治疗CCNE1扩增的HGSOC小鼠的耐受性良好,并能明显提高小鼠的存活率。Entinostat不仅能通过延缓DNA复制叉的进展来导致不可逆转的DNA损伤和细胞死亡,还能增强Olaparib药效来大幅度降低卵巢癌的细胞活性。目前PARP抑制剂治疗卵巢癌患者取得了显著的疗效,同时PARP抑制剂联合其他协同药物也为治疗CCNE1扩增的HGSOC提供了一种科学合理的策略。

5 结语与展望

CCNE1扩增与HGSOC的发生、发展、化疗耐药和预后不良密切相关,被认为是HGSOC化疗耐药的预测标志物和潜在的治疗靶标。CDK2小分子抑制剂或PARP抑制剂联合其他药物在治疗HGSOC方面已取得了一定的成效,但长期使用此类药物治疗CCNE1扩增的HGSOC可能会产生耐药性,严重影响患者预后。然而,CCNE1扩增的HGSOC产生耐药的作用机制尚未十分明确,暂时没有以CCNE1为靶向治疗的特效药物,因此有必要对发生CCNE1扩增的HGSOC的耐药机制进行深入探讨。随着研究的不断深入,未来以CCNE1为治疗靶标可能有助于改善HGSOC患者的预后和提高患者的生活质量。

参考文献

Kroeger PT Jr, Drapkin R.

Pathogenesis and heterogeneity of ovarian cancer

[J]. Curr Opin Obstet Gynecol, 2017, 29(1):26-34. doi: 10.1097/GCO.0000000000000340.

DOI:10.1097/GCO.0000000000000340      URL     [本文引用: 1]

Gorski JW, Ueland FR, Kolesar JM.

CCNE1 Amplification as a Predictive Biomarker of Chemotherapy Resistance in Epithelial Ovarian Cancer

[J]. Diagnostics (Basel), 2020, 10(5):279. doi: 10.3390/diagnostics10050279.

DOI:10.3390/diagnostics10050279      [本文引用: 1]

Sapoznik S, Aviel-Ronen S, Bahar-Shany K, et al.

CCNE1 expression in high grade serous carcinoma does not correlate with chemoresistance

[J]. Oncotarget, 2017, 8(37):62240-62247. doi: 10.18632/oncotarget.19272.

DOI:10.18632/oncotarget.19272      PMID:28977941      [本文引用: 3]

Delayed diagnosis of ovarian cancer, as well as high recurrence rates and lack of personalized therapy options, are among the causes for poor survival figures. Much effort is made towards developing new therapeutic possibilities, however predictive biomarkers are still unavailable. CCNE1 amplification, occurring in ∼20% of the high grade serous ovarian tumors, was previously proposed as a marker for platinum resistance and poor prognosis as well as for CDK2 inhibition. The current study aimed to examine the role of CCNE1 positive-immunostain as a predictor of first-line taxane-platinum chemoresistance. We evaluated matched pre- vs. post-neoadjuvant chemotherapy tumor samples and correlated the degree of pathological response to treatment with CCNE1 expression levels. Our results indicate that CCNE1 immunohistochemistry does not predict taxane-platinum chemoresistance in ovarian cancer patients. Further research is required in order to enable personalized adjuvant treatment, in cases where poor pathological response is achieved after the neoadjuvant phase.

Patch AM, Christie EL, Etemadmoghadam D, et al.

Whole-genome characterization of chemoresistant ovarian cancer

[J]. Nature, 2015, 521(7553):489-494. doi: 10.1038/nature14410.

DOI:10.1038/nature14410      URL     [本文引用: 2]

Li Y, Li L.

Bioinformatic screening for candidate biomarkers and their prognostic values in endometrial cancer

[J]. BMC Genet, 2020, 21(1):113. doi: 10.1186/s12863-020-00898-4.

DOI:10.1186/s12863-020-00898-4      URL     [本文引用: 2]

Schott C, Shah AT, Sweet-Cordero EA.

Genomic Complexity of Osteosarcoma and Its Implication for Preclinical and Clinical Targeted Therapies

[J]. Adv Exp Med Biol, 2020, 1258:1-19. doi: 10.1007/978-3-030-43085-6_1.

DOI:10.1007/978-3-030-43085-6_1      [本文引用: 2]

Zhao ZM, Yost SE, Hutchinson KE, et al.

CCNE1 amplification is associated with poor prognosis in patients with triple negative breast cancer

[J]. BMC Cancer, 2019, 19(1):96. doi: 10.1186/s12885-019-5290-4.

DOI:10.1186/s12885-019-5290-4      [本文引用: 2]

Integrated genomic analyses of ovarian carcinoma[J]. Nature, 2011, 474(7353):609-615. doi: 10.1038/nature10166.

DOI:10.1038/nature10166      URL     [本文引用: 1]

Li J, Fadare O, Xiang L, et al.

Ovarian serous carcinoma: recent concepts on its origin and carcinogenesis

[J]. J Hematol Oncol, 2012, 5:8. doi: 10.1186/1756-8722-5-8.

DOI:10.1186/1756-8722-5-8      URL     [本文引用: 1]

Piek JM, van Diest PJ, Zweemer RP, et al.

Dysplastic changes in prophylactically removed Fallopian tubes of women predisposed to developing ovarian cancer

[J]. J Pathol, 2001, 195(4):451-456. doi: 10.1002/path.1000.

DOI:10.1002/path.1000      PMID:11745677      [本文引用: 1]

The aim of this study was to investigate the occurrence of (pre)neoplastic lesions in overtly normal Fallopian tubes from women predisposed to developing ovarian carcinoma. The presence of (pre)neoplastic lesions was scored in histological specimens from 12 women with a genetically determined predisposition for ovarian cancer, of whom seven tested positive for a germline BRCA1 mutation. A control group included 13 women. Immunohistochemistry was used to determine the expression of p21, p27, p53, cyclin A, cyclin D1, bcl-2, Ki67, HER-2/neu, and the oestrogen and progesterone receptors. Loss of heterozygosity (LOH) analysis on the BRCA1 locus was also assessed on dysplastic tissue by PCR studies. Of the 12 women with a predisposition for ovarian cancer, six showed dysplasia, including one case of severe dysplasia. Five harboured hyperplastic lesions and in one woman no histological aberrations were found in the Fallopian tube. No hyperplastic, dysplastic or neoplastic lesions were detected in the Fallopian tubes of control subjects. In the cases studied, morphologically normal tubal epithelium contained a higher proportion of Ki67-expressing cells (p=0.005) and lower fractions of cells expressing p21 (p<0.0001) and p27 (p=0.006) than in the control group. Even higher fractions of proliferating cells were found in dysplastic areas (p=0.07) and accumulation of p53 was observed in the severely dysplastic lesion. Expression patterns of other proteins studied, including the hormone receptors, were similar in cases and controls. One subject, a germline BRCA1 mutation carrier, showed loss of the wild-type BRCA1 allele in the severely dysplastic lesion. In conclusion, the Fallopian tubes of women predisposed to developing ovarian cancer frequently harbour dysplastic changes, accompanied by changes in cell-cycle and apoptosis-related proteins, indicating an increased risk of developing tubal cancer.Copyright 2001 John Wiley & Sons, Ltd.

Karst AM, Jones PM, Vena N, et al.

Cyclin E1 deregulation occurs early in secretory cell transformation to promote formation of fallopian tube-derived high-grade serous ovarian cancers

[J]. Cancer Res, 2014, 74(4):1141-1152. doi: 10.1158/0008-5472.CAN-13-2247.

DOI:10.1158/0008-5472.CAN-13-2247      URL     [本文引用: 2]

Mei J, Tian H, Huang HS, et al.

Cellular models of development of ovarian high-grade serous carcinoma: A review of cell of origin and mechanisms of carcinogenesis

[J]. Cell Prolif, 2021, 54(5):e13029. doi: 10.1111/cpr.13029.

DOI:10.1111/cpr.13029      [本文引用: 1]

Kuhn E, Wang TL, Doberstein K, et al.

CCNE1 amplification and centrosome number abnormality in serous tubal intraepithelial carcinoma: further evidence supporting its role as a precursor of ovarian high-grade serous carcinoma

[J]. Mod Pathol, 2016, 29(10):1254-1261. doi: 10.1038/modpathol.2016.101.

DOI:10.1038/modpathol.2016.101      URL     [本文引用: 1]

Venuto S, Merla G.

E3 Ubiquitin Ligase TRIM Proteins, Cell Cycle and Mitosis

[J]. Cells, 2019, 8(5):510. doi: 10.3390/cells8050510.

DOI:10.3390/cells8050510      [本文引用: 1]

Malumbres M, Barbacid M.

Cell cycle, CDKs and cancer: a changing paradigm

[J]. Nat Rev Cancer, 2009, 9(3):153-166. doi: 10.1038/nrc2602.

DOI:10.1038/nrc2602      PMID:19238148      [本文引用: 1]

Tumour-associated cell cycle defects are often mediated by alterations in cyclin-dependent kinase (CDK) activity. Misregulated CDKs induce unscheduled proliferation as well as genomic and chromosomal instability. According to current models, mammalian CDKs are essential for driving each cell cycle phase, so therapeutic strategies that block CDK activity are unlikely to selectively target tumour cells. However, recent genetic evidence has revealed that, whereas CDK1 is required for the cell cycle, interphase CDKs are only essential for proliferation of specialized cells. Emerging evidence suggests that tumour cells may also require specific interphase CDKs for proliferation. Thus, selective CDK inhibition may provide therapeutic benefit against certain human neoplasias.

Pang W, Li Y, Guo W, et al.

Cyclin E: a potential treatment target to reverse cancer chemoresistance by regulating the cell cycle

[J]. Am J Transl Res, 2020, 12(9):5170-5187.

PMID:33042412      [本文引用: 1]

The cyclin family plays important roles in regulating the proliferative cycle of mammalian cells. Among the members of this family, cyclin E regulates multiple downstream molecules, such as the retinoblastoma susceptibility gene (RB1) and the transcription factor E2F, by interacting with cyclin-dependent kinases (CDKs) and plays an important role in the cell cycle transition from G1 to S phase. Over the years, studies have shown that cyclin E is closely related to the chemotherapy resistance of tumor cells and that its expression in tumor cells is closely related to prognosis. The dysregulated expression of cyclin E has a definite effect not only on the cell cycle regulation of tumor cells but also on the presence of low-molecular-weight cyclin E (LMW-E) and other cyclins that render tumor cells resistant. In addition, many studies in recent years have confirmed that chemotherapy resistance mediated by cyclin E can be reversed. For example, the combination of a cyclin-dependent kinase inhibitor (CKI) with anticancer drugs or the therapeutic targeting of related genes improves chemotherapy resistance by reducing the level or activity of cyclin E in tumor cells. This review summarizes the specific processes by which cyclin E regulates the cell cycle, its relationship to chemotherapy resistance in cancer, and its potential as a clinical therapeutic target to reverse chemotherapy resistance.AJTR Copyright © 2020.

Maloney SM, Hoover CA, Morejon-Lasso LV, et al.

Mechanisms of Taxane Resistance

[J]. Cancers (Basel), 2020, 12(11):3323. doi: 10.3390/cancers12113323.

DOI:10.3390/cancers12113323      URL     [本文引用: 1]

Otsuka I.

Mechanisms of High-Grade Serous Carcinogenesis in the Fallopian Tube and Ovary: Current Hypotheses, Etiologic Factors, and Molecular Alterations

[J]. Int J Mol Sci, 2021, 22(9):4409. doi: 10.3390/ijms22094409.

DOI:10.3390/ijms22094409      URL     [本文引用: 1]

Hanahan D, Weinberg RA.

The hallmarks of cancer

[J]. Cell, 2000, 100(1):57-70. doi: 10.1016/s0092-8674(00)81683-9.

DOI:10.1016/s0092-8674(00)81683-9      PMID:10647931      [本文引用: 1]

Etemadmoghadam D, George J, Cowin PA, et al.

Amplicon-dependent CCNE1 expression is critical for clonogenic survival after cisplatin treatment and is correlated with 20q11 gain in ovarian cancer

[J]. PLoS One, 2010, 5(11):e15498. doi: 10.1371/journal.pone.0015498.

DOI:10.1371/journal.pone.0015498      [本文引用: 1]

Hwang HC, Clurman BE.

Cyclin E in normal and neoplastic cell cycles

[J]. Oncogene, 2005, 24(17):2776-2786. doi: 10.1038/sj.onc.1208613.

DOI:10.1038/sj.onc.1208613      URL     [本文引用: 1]

Keck JM, Summers MK, Tedesco D, et al.

Cyclin E overexpression impairs progression through mitosis by inhibiting APC(Cdh1)

[J]. J Cell Biol, 2007, 178(3):371-385. doi: 10.1083/jcb.200703202.

DOI:10.1083/jcb.200703202      URL     [本文引用: 1]

Penner-Goeke S, Lichtensztejn Z, Neufeld M, et al.

The temporal dynamics of chromosome instability in ovarian cancer cell lines and primary patient samples

[J]. PLoS Genet, 2017, 13(4):e1006707. doi: 10.1371/journal.pgen.1006707.

DOI:10.1371/journal.pgen.1006707      URL     [本文引用: 1]

Vishwakarma R, McManus KJ.

Chromosome Instability; Implications in Cancer Development, Progression, and Clinical Outcomes

[J]. Cancers (Basel), 2020, 12(4):824. doi: 10.3390/cancers12040824.

DOI:10.3390/cancers12040824      URL     [本文引用: 1]

Chan AM, Enwere E, McIntyre JB, et al.

Combined CCNE1 high-level amplification and overexpression is associated with unfavourable outcome in tubo-ovarian high-grade serous carcinoma

[J]. J Pathol Clin Res, 2020, 6(4):252-262. doi: 10.1002/cjp2.168.

DOI:10.1002/cjp2.168      URL     [本文引用: 1]

Petersen S, Wilson AJ, Hirst J, et al.

CCNE1 and BRD4 co-amplification in high-grade serous ovarian cancer is associated with poor clinical outcomes

[J]. Gynecol Oncol, 2020, 157(2):405-410. doi: 10.1016/j.ygyno.2020.01.038.

DOI:10.1016/j.ygyno.2020.01.038      URL     [本文引用: 1]

Filippova OT, Selenica P, Pareja F, et al.

Molecular characterization of high-grade serous ovarian cancers occurring in younger and older women

[J]. Gynecol Oncol, 2021, 161(2):545-552. doi: 10.1016/j.ygyno.2021.02.028.

DOI:10.1016/j.ygyno.2021.02.028      PMID:33674143      [本文引用: 1]

To determine if the mutational landscapes and genomic features of homologous recombination DNA repair defects (HRD) vary between younger and older patients with high-grade serous ovarian cancer (HGSOC).Younger and older women were defined as bottom and top age quartiles, respectively. HGSOCs from 15 younger (median 49 years, range 35-53) and 15 older women (median 72 years, range 70-87) were subjected to whole-exome sequencing (WES). For validation, HGSOC WES data were obtained from The Cancer Genome Atlas (TCGA), including 38 younger (median 45 years, range 34-50) and 30 older women (median 74 years, range 68-84). Mutational profiles, BRCA1/2 status, genomic HRD features, and for TCGA cases RNA-sequencing-based HRD transcriptomic signatures were assessed.In the institutional cohort, pathogenic germline BRCA1/2 mutations were more frequent in younger (5/15) than older women (0/15, p = 0.042). No somatic BRCA1/2 mutations were identified. HGSOCs from older patients preferentially displayed aging-related mutational signatures and, in contrast to younger patients, harbored CCNE1 amplifications (3/15, 20%). In the TCGA cohort, pathogenic germline BRCA1 (younger 8/38, older 0/30, p = 0.007) but not BRCA2 mutations (young 3/38, older 4/30, p = 0.691) were more frequent in younger patients. Again, no somatic BRCA1/2 mutations were identified. HGSOCs from younger women more frequently displayed genomic features of HRD (all, p < 0.05), a significant HRD gene-signature enrichment, but less frequently CCNE1 amplification (p = 0.05). Immunoreactive CLOVAR subtypes were more common in HGSOCs from younger women, and proliferative subtypes in HGSOCs from older women (p = 0.041).HGSOC patients diagnosed at an older age less frequently harbor pathogenic BRCA1 germline mutations and genomic features of HRD than younger women. Individualized treatment options, particularly pertaining to use of PARP inhibitors, in older women may be warranted.Copyright © 2021 Elsevier Inc. All rights reserved.

Yang L, Fang D, Chen H, et al.

Cyclin-dependent kinase 2 is an ideal target for ovary tumors with elevated cyclin E1 expression

[J]. Oncotarget, 2015, 6(25):20801-20812. doi: 10.18632/oncotarget.4600.

DOI:10.18632/oncotarget.4600      PMID:26204491      [本文引用: 2]

CCNE1 gene amplification is present in 15-20% ovary tumor specimens. Here, we showed that Cyclin E1 (CCNE1) was overexpressed in 30% of established ovarian cancer cell lines. We also showed that CCNE1 was stained positive in over 40% of primary ovary tumor specimens regardless of their histological types while CCNE1 staining was either negative or low in normal ovary and benign ovary tumor tissues. However, the status of CCNE1 overexpression was not associated with the tumorigenic potential of ovarian cancer cell lines and also did not correlate with pathological grades of ovary tumor specimens. Subsequent experiments with CCNE1 siRNAs showed that knockdown of CCNE1 reduced cell growth only in cells with inherent CCNE1 overexpression, indicating that these cells may have developed an addiction to CCNE1 for growth/survival. As CCNE1 is a regulatory factor of cyclin-dependent kinase 2 (Cdk2), we investigated the effect of Cdk2 inhibitor on ovary tumorigenecity. Ovarian cancer cells with elevated CCNE1 expression were 40 times more sensitive to Cdk2 inhibitorSNS-032 than those without inherent CCNE1 overexpression. Moreover, SNS-032 greatly prolonged the survival of mice bearing ovary tumors with inherent CCNE1 overexpression. This study suggests that ovary tumors with elevated CCNE1 expression may be staged for Cdk2-targeted therapy.

Nakayama N, Nakayama K, Shamima Y, et al.

Gene amplification CCNE1 is related to poor survival and potential therapeutic target in ovarian cancer

[J]. Cancer, 2010, 116(11):2621-2634. doi: 10.1002/cncr.24987.

DOI:10.1002/cncr.24987      PMID:20336784      [本文引用: 1]

This study examined the clinical significance of CCNE1 (Cyclin E1) amplification and assessed whether CCNE1 is a potential therapeutic target in ovarian cancer.CCNE1 expression and amplification in ovarian cancer was assessed by immunohistochemistry, fluorescence in situ hybridization and clinical data collected by retrospective chart review. CCNE1 gene knockdown using silencing RNA and a CCNE1 gene transfection system were used to asses CCNE1 function in tissue samples of ovarian cancer.Gene amplification was identified in 18 (20.4%) of 88 ovarian carcinomas. CCNE1 copy number significantly correlated with CCNE1 protein expression (r = 0.522, P <.0001). CCNE1 amplification significantly correlated with shorter disease-free survival and overall survival (P <.001). There were nonsignificant trends between high protein expression and poor disease-free survival (P =.2865) and overall survival (P =.1248). Multivariate analysis showed gene amplification was an independent prognostic factor for disease-free survival and overall survival after standard platinum-taxane chemotherapy (P =.0274, P =.0023). Profound growth inhibition and apoptosis were observed in silencing RNA-treated cancer cells with gene amplification compared with results in cancer cells with CCNE1 moderate expression without gene amplification or with low CCNE1 expression. CCNE1 overexpression stimulated proliferation in ovarian cancer cell lines ES2 and TOV-21G, which have lower endogenous CCNE1 expression.These findings indicate that CCNE1 overexpression is critical to growth and survival of ovarian cancer tumors with CCNE1 gene amplification. Furthermore, they suggest that CCNE1 silencing RNA-induced phenotypes depend on amplification status of ovarian cancers. Therefore, CCNE1-targeted therapy may benefit ovarian cancer patients with CCNE1 amplification.(c) 2010 American Cancer Society.

Aziz D, Etemadmoghadam D, Caldon CE, et al. 19q12 amplified and non-amplified subsets of high grade serous ovarian cancer with overexpression of cyclin E1

differ in their molecular drivers and clinical outcomes

[J]. Gynecol Oncol, 2018, 151(2):327-336. doi: 10.1016/j.ygyno.2018.08.039.

DOI:10.1016/j.ygyno.2018.08.039      [本文引用: 1]

Au-Yeung G, Lang F, Azar WJ, et al.

Selective Targeting of Cyclin E1-Amplified High-Grade Serous Ovarian Cancer by Cyclin-Dependent Kinase 2 and AKT Inhibition

[J]. Clin Cancer Res, 2017, 23(7):1862-1874. doi: 10.1158/1078-0432.CCR-16-0620.

DOI:10.1158/1078-0432.CCR-16-0620      PMID:27663592      [本文引用: 2]

Cyclin E1 () amplification is associated with primary treatment resistance and poor outcome in high-grade serous ovarian cancer (HGSC). Here, we explore approaches to target -amplified cancers and potential strategies to overcome resistance to targeted agents. To examine dependency on in -amplified HGSC, we utilized siRNA and conditional shRNA gene suppression, and chemical inhibition using dinaciclib, a small-molecule CDK2 inhibitor. High-throughput compound screening was used to identify selective synergistic drug combinations, as well as combinations that may overcome drug resistance. An observed relationship between and the AKT pathway was further explored in genomic data from primary tumors, and functional studies in fallopian tube secretory cells. We validate as a therapeutic target by demonstrating selective sensitivity to gene suppression. However, we found that dinaciclib did not trigger amplicon-dependent sensitivity in a panel of HGSC cell lines. A high-throughput compound screen identified synergistic combinations in -amplified HGSC, including dinaciclib and AKT inhibitors. Analysis of genomic data from TCGA demonstrated coamplification of and Overexpression of Cyclin E1 and AKT isoforms, in addition to mutant, imparted malignant characteristics in untransformed fallopian tube secretory cells, the dominant site of origin of HGSC. These findings suggest a specific dependency of -amplified tumors for AKT activity, and point to a novel combination of dinaciclib and AKT inhibitors that may selectively target patients with -amplified HGSC..©2016 American Association for Cancer Research.

Etemadmoghadam D, Au-Yeung G, Wall M, et al.

Resistance to CDK2 inhibitors is associated with selection of polyploid cells in CCNE1-amplified ovarian cancer

[J]. Clin Cancer Res, 2013, 19(21):5960-5971. doi: 10.1158/1078-0432.CCR-13-1337.

DOI:10.1158/1078-0432.CCR-13-1337      PMID:24004674      [本文引用: 1]

Amplification of cyclin E1 (CCNE1) is associated with poor outcome in breast, lung, and other solid cancers, and is the most prominent structural variant associated with primary treatment failure in high-grade serous ovarian cancer (HGSC). We have previously shown that CCNE1-amplified tumors show amplicon-dependent sensitivity to CCNE1 suppression. Here, we explore targeting CDK2 as a novel therapeutic strategy in CCNE1-amplified cancers and mechanisms of resistance.We examined the effect of CDK2 suppression using RNA interference and small-molecule inhibitors in SK-OV-3, OVCAR-4, and OVCAR-3 ovarian cancer cell lines. To identify mechanisms of resistance, we derived multiple, independent resistant sublines of OVCAR-3 to CDK2 inhibitors. Resistant cells were extensively characterized by gene expression and copy number analysis, fluorescence-activated cell sorting profiling and conventional karyotyping. In addition, we explored the relationship between CCNE1 amplification and polyploidy using data from primary tumors.We validate CDK2 as a therapeutic target in CCNE1-amplified cells by showing selective sensitivity to suppression, either by gene knockdown or using small-molecule inhibitors. In addition, we identified two resistance mechanisms, one involving upregulation of CDK2 and another novel mechanism involving selection of polyploid cells from the pretreatment tumor population. Our analysis of genomic data shows that polyploidy is a feature of cancer genomes with CCNE1 amplification.These findings suggest that cyclinE1/CDK2 is an important therapeutic target in HGSC, but that resistance to CDK2 inhibitors may emerge due to upregulation of CDK2 target protein and through preexisting cellular polyploidy.

Mittica G, Ghisoni E, Giannone G, et al.

PARP Inhibitors in Ovarian Cancer

[J]. Recent Pat Anticancer Drug Discov, 2018, 13(4):392-410. doi: 10.2174/1574892813666180305165256.

DOI:10.2174/1574892813666180305165256      PMID:29512470      [本文引用: 1]

Treatment of Epithelial Ovarian Cancer (EOC), historically based on surgery and platinum doublet chemotherapy, is associated with high risk of relapse and poor prognosis for recurrent disease. In this landscape, the innovative treatment with PARP inhibitors (PARPis) demonstrated an outstanding activity in EOC, and is currently changing clinical practice in BRCA mutant patients.The study aimed to highlight the mechanism of action, pharmacokinetics, clinical activity, indications and current strategies of development of Olaparib, Niraparib, Rucaparib, Talazoparib and Veliparib, the 5 most relevant PARPis.We performed a review on Pubmed using 'ovarian cancer' and the name of each PARPi (PARP inhibitor) discussed in the review as Medical Subject Headings (MeSH) keywords. The same search was performed on "clinicaltrial.gov" to identify ongoing clinical trials and on "google. com/patents" and "uspto.gov" for recent patents exploring PARPIs in ovarian cancer.Olaparib, Niraparib and Rucaparib are already approved for the treatment of recurrent EOC and their indications are partially overlapping. Talazoparib and Veliparib are promising PARPis, but currently under investigation in early phase trials. Several studies are evaluating PARPis in monotherapy or in associations, in a wide range of settings (i.e. first line, neoadjuvant, platinum-sensitive and resistant disease).PARPis are valuable options in patients with recurrent ovarian cancer with promising activity in different stages of this disease. Further studies are required to better define optimal clinical settings, predictors of response beyond BRCA mutations and strategies to overcome secondary resistance of PARPis therapy in EOC.Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.

Mirza MR, Coleman RL, González-Martín A, et al.

The forefront of ovarian cancer therapy: update on PARP inhibitors

[J]. Ann Oncol, 2020, 31(9):1148-1159. doi: 10.1016/j.annonc.2020.06.004.

DOI:S0923-7534(20)39891-4      PMID:32569725      [本文引用: 1]

In recurrent ovarian cancer, poly(ADP-ribose) polymerase (PARP)-inhibiting agents have transformed the treatment of platinum-sensitive disease. New data support use of PARP inhibitors earlier in the treatment algorithm.We review results from recent phase III trials evaluating PARP inhibitors as treatment and/or maintenance therapy for patients with newly diagnosed ovarian cancer. We discuss the efficacy and safety of these agents in the all-comer and biomarker-selected populations studied in clinical trials, and compare the strengths and limitations of the various trial designs. We also consider priorities for future research, with a particular focus on patient selection and future regimens for populations with high unmet need.Four phase III trials (SOLO-1, PAOLA-1/ENGOT-OV25, PRIMA/ENGOT-OV26 and VELIA/GOG-3005) demonstrated remarkable improvements in progression-free survival with PARP inhibitor therapy (olaparib, niraparib or veliparib) for newly diagnosed ovarian cancer. Differences in trial design (treatment and/or maintenance setting; single agent or combination; bevacizumab or no bevacizumab), patient selection (surgical outcome, biomarker eligibility, prognosis) and primary analysis population (intention-to-treat, BRCA mutated or homologous recombination deficiency positive) affect the conclusions that can be drawn from these trials. Overall survival data are pending and there is limited experience regarding long-term safety.PARP inhibitors play a pivotal role in the management of newly diagnosed ovarian cancer, which will affect subsequent treatment choices. Refinement of testing for patient selection and identification of regimens to treat populations that appear to benefit less from PARP inhibitors are a priority.Copyright © 2020 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.

Ibrahim EM, Refae AA, Bayer AM, et al.

Poly(ADP-ribose) polymerase inhibitors as maintenance treatment in patients with newly diagnosed advanced ovarian cancer: a meta-analysis

[J]. Future Oncol, 2020, 16(10):585-596. doi: 10.2217/fon-2020-0057.

DOI:10.2217/fon-2020-0057      URL     [本文引用: 1]

Onstad M, Coleman RL, Westin SN.

Movement of Poly-ADP Ribose (PARP) Inhibition into Frontline Treatment of Ovarian Cancer

[J]. Drugs, 2020, 80(15):1525-1535. doi: 10.1007/s40265-020-01382-0.

DOI:10.1007/s40265-020-01382-0      URL     [本文引用: 1]

Kim H, Xu H, George E, et al.

Combining PARP with ATR inhibition overcomes PARP inhibitor and platinum resistance in ovarian cancer models

[J]. Nat Commun, 2020, 11(1):3726. doi: 10.1038/s41467-020-17127-2.

DOI:10.1038/s41467-020-17127-2      URL     [本文引用: 1]

Gupta VG, Hirst J, Petersen S, et al.

Entinostat, a selective HDAC1/2 inhibitor, potentiates the effects of olaparib in homologous recombination proficient ovarian cancer

[J]. Gynecol Oncol, 2021, 162(1):163-172. doi: 10.1016/j.ygyno.2021.04.015.

DOI:10.1016/j.ygyno.2021.04.015      URL     [本文引用: 1]

/

津ICP备05004273
版权所有 © 天津市医学科学技术信息研究所
地址:天津市和平区贵州路94号 邮编:300070 电话:022-23337521 E-mail:fuchanfence@gjfckx.ac.cn
本系统由北京玛格泰克科技发展有限公司设计开发