循环肿瘤细胞在卵巢癌预后中的研究进展

时间:2022-08-14 04:03:24

循环肿瘤细胞在卵巢癌预后中的研究进展

摘要:卵巢癌作为最常见妇科恶性肿瘤之一,其死亡率位居女性生殖系统肿瘤第一位。循环肿瘤细胞(circulating tumor cells,CTC)是由原发肿瘤病灶脱落进入循环中的肿瘤细胞,近年来有研究证实CTC可作为卵巢癌预后的独立因素,可能成为一种新型肿瘤标志物来预测卵巢癌的预后、指导治疗决策的制定甚至成为靶向治疗的靶向位点。本文简要综述了CTC的常用检测方法以及其对卵巢癌预后的预测价值的研究进展。

关键词:卵巢癌;循环肿瘤细胞;预后;肿瘤标志物

Advance on the Predict Value of Circulating Tumor Cells on Ovarian Cancer

ZENG Li-qiong,YANG Zhu

(Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University,Chongqing 400010,China)

Abstract:Ovarian cancer as one of the most common gynecological m alignant tumors, it played the first mortality in the female reproductive system. Recent years, Circulating tumor cells (CTC) which were fell off into circulatory system from the primary tumor lesions have been confirmed that could be an independent prognostic factor for ovarian cancer, and thus to become a new tumor marker to predict the prognosis of ovarian carcinoma, to guide the decision of treatment strategies, and even as a target spot of targeted therapy. In this paper, we briefly summarized the current methods of CTC-detection and its progress of the predictive value of prognosis for ovarian neoplasm.

Key words:Ovarian cancer; Circulating tumor cells (CTC); Prognosis; Biomarker

据全球癌症数据统计,2008年在全世界范围内,约有225500例新发病例,大约140200例女性死于卵巢癌,分别占女性癌症发病率第8位及死亡率第7位。在发展中国家,新发病例及死亡病例分别为125200例与75700例,居女性癌症发病率及死亡率的第9及第8位[1]。据我国癌症数据统计,2010年卵巢癌在女性肿瘤中全国发病率为6.47/10万,占女性肿瘤发病的第9位[2]。早期卵巢癌在经过规范治疗后,绝大部分可以达到治愈或者完全缓解,5年生存率可达90%。但由于缺乏有效的早期检测手段,70%卵巢癌发现时已属晚 期[3]。晚期卵巢癌,推荐治疗为满意肿瘤减灭术后,辅以化学治疗。对于不能手术或有明确手术禁忌患者,推荐先给予新辅助化疗后,再次评估手术治疗。复发卵巢癌,治疗方案选择主要依据复发时间而定。为提高卵巢癌疗效,靶向治疗药物尤其是贝伐珠单抗是目前研究的热点药物。目前对于卵巢癌疗效的评估手段有限,主要为CA-125和影像学观察等措施。CA-125作为上皮性卵巢癌的诊断及疗效观测常用标志物,但其对于卵巢癌治疗效果的评估缺乏足够的特异性。近年来有研究显示CTC可能成为一种新的肿瘤标志物来预测卵巢癌的预后。本文就近年来关于CTC的检测方法及其与卵巢癌预后关系的文章做一综述。

1 CTC的概述

Ashworth首次报道在肿瘤患者死后的血液中发现类似于肿瘤细胞的细 胞[4]。有研究证实CTC存在于肿瘤患者循环系统中,罕见于健康人群[5]。在多种肿瘤中,已证实CTC阳性患者整体生存期(OS)及无进展生存期(DFS)明显短于阴性患者。Cain等首次在卵巢癌患者骨髓(BM)样本中检测到CTC,其阳性率为23%[6]。许多研究证实CTC可作为一种新型的肿瘤标志物,预测卵巢癌的治疗效果[7-16]。肿瘤细胞到达远隔器官形成转移灶通常包括3个过程:内渗、外渗、定居[23]。通常情况下原发病灶的肿瘤细胞通过自分泌多种细胞因子溶解与基底膜之间的连接或者上皮间质转换(EMT)而获得侵袭能力[24],从而侵犯到临近基质组织,进入到循环系统中。每克肿瘤组织每天大约释放106个肿瘤细胞入血[25],这些细胞在循环中的存活率极低,绝大多数发生失巢凋亡, 只有极少部分具有较强侵袭能力的肿瘤细胞通过表达凋亡抑制因子存活下来[26]。在适宜的条件下最终在远隔器官生长成肉眼可见的病灶。同时CTC也可以向骨髓归巢,进入骨髓储备池并维持静息状态,一定条件下再次释放入外周血循环,活化增殖形成转移灶[27]。由于CTC在骨髓及血液中的量极少,接近于1 CTC/mL血液或者每106~107白细胞中有一个CTC[28],故CTC的检测有一定难度。

2 CTC的检测

2.1 CTC的富集 CTC的检测通常需两个步骤:富集和分离纯化。CTC富集主要根据肿瘤细胞的物理和生物学特性来完成[29]。根据肿瘤细胞的物理特性富集CTC主要通过细胞大小及密度来检测。有密度梯度离心法(FICOLL、Onco-Quick);特殊滤过膜过滤法(ISET[30],3D-滤过膜过滤法[31],MEMS[32])、微流体控制系统(捏流耦合切变惯性微流体法[33];MOFF and DEP[34])、非生物标志芯片法[35]。根据肿瘤生物学特性检测CTC则是结合肿瘤细胞表达肿瘤相关抗原来筛选细胞,分为阳性选择和阴性选择。上皮细胞粘附分子(EpCAM)是一种最常见的上皮性肿瘤表面检测靶点。Cellsearch 系统、CTC-chip和 Herringbone-chip法均为以EpCAM作为免疫标志[5,36-37]。现以EpCAM作为标志的纳米技术的运用使得肿瘤细胞检测的限制性明显降低(标本量),同时可提高测定的敏感性[38]。肿瘤细胞具有强侵袭性, 肿瘤在向周围组织侵袭的过程中会摄取大量细胞粘附基质(CAM),通过检测细胞摄取CAM的情况亦可富集肿瘤细胞[8]。

2.2肿瘤细胞的分离纯化 CTC分离纯化常用方法为免疫细胞组化法(Immunocytochemistry analysis,ICC),其荧光染色的靶点包括上皮性肿瘤特异性抗原如细胞角蛋白(CK)、上皮特异性粘附分子如EpCAM和E-钙黏蛋白、肿瘤特异性蛋白如雌孕激素受体等。其主要作为基础检测方式,现多种CTC检测方法均以ICC为基础。如流式细胞术(flow cytometry,FCM)、激光扫描细胞仪(LSC)[39]。Cellsearch作为最具代表性的ICC检测方法,其检测过程结合了EpCAM、CK-8、CK-18、CK-19、CD45,以及核染色DAPI(4,6-diamino-2-phenylindole)[5]。这些标准同样被运用于CTC-chip、HB-chip以及纳米检测技术中。基于ICC的逆转录聚合酶链反应(RT-PCR)将肿瘤特异性mRNA逆转录扩增后检测CTC,其灵敏度极高,但缺乏足够特异性,代表方法有Adnagen。近年来有研究通过PCR法检测肿瘤细胞基因有望取代单纯检测CTC本身(Cyclophilin C, PICC)[13]。EPISPOT亦是通过细胞免疫组化法检测CTC,其将离体肿瘤细胞培养24~48 h后提取肿瘤相关分泌蛋白,进而检测CTC。其靶向的细胞蛋白有CK-19,MUC1 (mucin-1),PSA(prostate-specific antigen), FGF-2(fibroblast growth factor-2)[40]。由于在肿瘤细胞富集过程中往往存在偏倚,因而光导纤维阵列技术[41]及数字化自动扫描荧光显微镜技术被开发出来[42]。

2.3 Cellsearch系统 目前市面上CTC检测方法众多,但唯一作为FDA通过的用于检测CTC的方法只有Cellsearch系统[5]。美国FDA分别在2004年、2007年和2008年批准CellSearch系统用于转移性乳腺癌、结直肠癌及前列腺癌的CTC检测。2012年CellSearch系统作为CTC检测与分析系统获得中国FDA批准用于乳腺癌患者的预后分析。该系统将循环肿瘤细胞定义为EpCAM+/CK+/DAPI+/CD45-表达细胞。CellSearch 系统是目前自动化程度最高的CTC 检测技术,受人为因素影响较小,具有较高的特异性、敏感性及可重复性[5]。目前各种检测方法测得CTC阳性率高低不同,Cellsearch系统作为唯一被FDA允许的CTC检测方法,但其对上皮性标志物如EpCAM/CK表达阴性细胞,存在检测困难,可能有漏诊。肿瘤细胞与EMT的关系已被众多研究证 实[24],众多经过EMT的高侵袭性肿瘤细胞可能不再表达EpCAM/CK从而逃避传统的检测方法[43]。有研究开始采用新的方法以检测那些因为缺乏或低表达上皮标记的CTC,Pecot等采用微流体控制系统来检测CK阴性细胞,可用于检测那些经过EMT上皮表达缺失的CTC[44]。近来,CTC的检测已不仅仅局限于细胞学的检测,有研究开始着眼于CTC相关基因的检测,Obermayr等采用RT-PCR法检测11个与CTC相关的基因及EpCAM,结果显示PPIC基因在CTC中阳性率为68%,而EpCAM仅为8%。且结果显示PPIC基因阳性是卵巢癌独立预后不良因子[13]。其提示有将PPIC替代CTC作为检测卵巢癌预后的指标可能。

3 CTCs在卵巢癌中的运用现状

早期腹膜扩散是卵巢癌预后不佳的重要原因,卵巢癌主要播散方式为腹腔内播散,较少通过血液循环传播[45],远处转移在卵巢癌是一种迟发型并发症,大约只发生在1/3的患者中,一旦卵巢癌发生远处转移,患者治疗效果十分有限[46]。CTC在乳腺癌和结直肠癌以及前列腺中的应用价值已被证实。有关CTC在卵巢癌运用中的研究已较多[15-30]。有研究显示CTC可作为一种新的肿瘤标志物[15-24]。CTC的检测可以通过PB和BM样本进行,阳性检出率为14.35%~60.6%,目前检测方法多为ICC法,其中Behbakht,Liu以及Poveda等采用了目前公认的Cellsearch系统。本文就近年来关于CTC检测与卵巢癌预后的文献做一总结,见表1。

4结论与展望

目前的研究数据多为治疗前的CTC数值与卵巢癌患者预后的关系,较少涉及随访过程中的CTC数目变化与患者预后的关系,Obermayr等研究了基础水平及随访过程中CTC水平与卵巢癌预后的关系,结果显示DFS及OS与CTCs基线水平无关,而与随访中CTCs仍保持阳性有关(P=0.001),显示出持续检测CTC的意义[13]。由于CTC检测价格高昂,故随访中CTC检测失访率较高,因而限制其运用。若能开发出敏感性和特异性更高以及成本价格更合理的CTC检测方法,CTC检测可能可以纳入到卵巢癌常规检测项目中。目前对CTC数目与肿瘤预后的研究已经相当多,但关于CTC与个体化治疗的研究仍不多,通过检测患者肿瘤组织中基因表达/突变来筛选相应抗肿瘤药物在众多研究中被证实。但所有的检测均建立在能得到肿瘤组织的基础上,由于肿瘤组织存在异质性,随着药物的使用,肿瘤的基因表达/突变可能发生改变,导致最初有效的药物变得无效。因此为了更好地对肿瘤患者实施个体化选药, 最好对肿瘤的性质进行实时监测。CTC为术后肿瘤标本的唯一来源,通过对其进行基因及表面抗位的检查,可以明确肿瘤的表型,使得其成为一个良好的肿瘤实时监测指标,从而更好的指导肿瘤的个体化治疗。根据得到的CTC监测中基因表达情况来选择相应治疗药物,达到事半功倍的效果。近年来靶向治疗已成为肿瘤治疗的热点,随着对CTC研究的成熟,以CTC作为靶向治疗药物位点可能成为现实。

参考文献:

[1]Jemal A.Global Cancer Statistics[J].CA Cancer J Clin,2011,2: 69-90.

[2]陈万青等. 中国2010年恶性肿瘤发病和死亡分析.中国肿瘤,2014,1,1-10.

[3]Badgwell D. Early detection of ovarian cancer[J].Dis Markers,2007,5-6:397-341.

[4]Ashworth T. R. A case of cancer in which cells similar to those in the tumours were seen in the blood after death[J].Australian Med,1869,14:146.

[5]Allard WJ.Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonm alignant diseases[J].Clin Cancer Res,2004,10:6897-6904.

[6]Cain JM.Bone marrow involvement in epithelial ovarian cancer by immunocytochemical assessment[J].Gynecol Oncol,1990,38(3):442-445.

[7]Poveda A.Circulating tumor cells Predict progression free survival and overall survival in patients with relapsed/recurrent advanced ovarian cancer[J].Gynecol Oncol,2011,3:567-572.

[8]Fan T.Clinical significance of circulating tumor cells detected by an invasion assay in peripheral blood of patients with ovarian cancer[J].Gynecol Oncol,2009,1:185-191.

[9]Banys M.Disseminated tumor cells in bone marrow may affect prognosis of patients with gynecologic m alignancies[J].Int J Gynecol Cancer,2009,5:948-952.

[10]Aktas B.Molecular profiling and prognostic relevance of circulating tumor cells in the blood of ovarian cancer patients at primary diagnosis and after platinum-based chemotherapy[J].Int J Gynecol Cancer,2011,5:822-830.

[11]Braun S.Occult tumor cells in bone marrow of patients with locoregionally restricted ovarian cancer predict early distant metastatic relapse[J].J Clin Oncol,2001,2:368-375.

[12]Wimberger P.Influence of platinum-based chemotherapy on disseminated tumor cells in blood and bone marrow of patients with ovarian cancer[J].Gynecol Oncol,2007,2:331-338.

[13]Obermayr E.Molecular characterization of circulating tumor cells in patients with ovarian cancer improves their prognostic significance[J].Gynecol Oncol,2013,1:15-21.

[14]Wimberger P.Impact of platinum -based chemotherapy on circulating nucleic acid levels, protease activities in blood and disseminated tumor cells in bone marrow of ovarian cancer patients[J].Int J Cancer,2011,11:2572-2580.

[15]Schindlbeck C.Prognostic impact of KI67, p53, human epithelial growth factor receptor 2,Topoisomerase IIalpha, epidermal growth factor receptor and nm23 expression of ovarian carcinomas anddisseminated tumor cells in the bone marrow[J].Int J Gynecol Cancer,2007,5:1047-1055.

[16]Fehm T.Pooled analysis of the prognostic relevance of disseminated tumor cells in the bone marrow of patients with ovarian cancer[J].Int J Gynecol Cancer,2013,23:839-845.

[17]Judson PL.Preoperative detection of peripherally circulating cancer cells and its prognostic significance in ovarian cancer[J].Gynecol Oncol,2003,2:389-394.

[18]Fehm T.Detection of disseminated tumor cells in patients with gynecological cancers[J]. Gynecol Oncol,2006,3:942-947.

[19]Behbakht K,.Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent andrecurrent epithelial ovarian and primary peritoneal m alignancies[J].Gynecol Oncol,2011,1:19-26.

[20]Liu JF.Predictive value of circulating tumor cells (CTCs) in newly-diagnosed and recurrent ovarian cancer patients[J].Gynecol Oncol,2013, 2:352-356.

[21]Kuhlmann JDLoss of Heterozygosity proximal to the M6P/IGF2R locus is predictive for the presence of disseminated tumor cells in the bone marrow of ovarian cancer patients before and after chemotherapy[J].Genes Chromosomes Cancer.2011,8:598-605.

[22]Marth C.irculating tumor cells in the peripheral blood and bone marrow of patients with ovarian carcinoma do not predict prognosis[J].Cancer,2002, 3:707-712.

[23]Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited[J].Nat Rev Cancer.2003,6:453-8.

[24]Joyce JA,et al. Microenvironmental regulation of metastasis[J].Nat Rev Cancer,2009,9:239-352.

[25]Chang YS.Mosaic blood vessels in tumors: frequency of cancer cells in contact with flowing blood. Proc[J].Natl Acad Sci, 2000,26:14608-13.

[26]Berezovskaya O.Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells[J].Cancer Re,6:2378-2386.

[27]Pantel K.Detection, clinical relevance and specific biological properties of disseminating tumour cells[J].Nat Rev Cancer,2008, 5:329-40.

[28]Franklin WA.Incidence of tumor-cell contamination in leukapheresis products of breast cancer patients mobilized with stem cell factor and granulocyte colony-stimulating factor (G-CSF)or with G-CSF alone[J].Blood,1999,94:340-347.

[29]Alix C.Circulating tumor cells and circulating tumor DNA[J].Annu Rev Med,2012,63:199-215.

[30]Vona G, Sabile A, Louha M,et al. Isolation by size of epithelial tumor cells: a new method for the immunomorphological and molecular characterization of circulating tumor cells[J].Am J Pathol,2000,1:57-63.

[31]Zheng S.3D-microfilter device for viable circulating tumor cell (CTC) enrichment from blood[J].Biomed Microdevices.2011,1:203-13.

[32]Zheng S.Membrane microfilter device for selective capture, electrolysis and genomic analysis of human circulating tumor cells[J].Chromatogr A,2007,2:154-161.

[33]Bhagat AA.Pinched flow coupled shear-modulated inertial microfluidics for high-throughput rare blood cell separation[J].Lab Chip,2011,11:1870-1878.

[34]Moon HS.Continuous separation of breast cancer cells from blood samples using multi-orifice flow fractionation (MOFF)and dielectrophoresis (DEP)[J].Lab Chip, 2011,11:1118-25.

[35]Tan SJ.Versatile label free biochip for the detection of circulating tumor cells from peripheral blood in cancer patients[J].Biosens. Bioelectron.2010.26:1701-1705.

[36]Nagrath S.Isolation of rare circulating tumour cells in cancer patients by microchip technology[J].Nature 2007;450:1235-1239.

[37]Stott SL.Isolation of circulating tumor cells using a microvortex-generating herringbone-chip.Proc Natl Acad Sci USA.43:18392-18397.

[38]Parkinson DR.Considerations in the development of circulating tumor cell technology for clinical use[J].J TranMed,2010,1:138.

[39]Pachmann K.An increase in cell number at completion of therapy may develop as all indicator of early relapse.J Cancer Res Clin Oncol,2008,1: 59-65.

[40]Alix- C, Vendrell JP, Slijper M, et al. Full-length cytokeratin-19 is released by human tumor cells: a potential role in metastatic progression of breast cancer. Breast Cancer Res.2009.11:R39.

[41]Krivacic RT.A rare-cell detector for cancer. Proc[J].Natl Acad Sci 2004.101:10501-10504.

[42]Ntouroupi TG, et al.Detection of circulating tumour cells in peripheral blood with an automated scanning fluorescence microscope[J].Br.J.Cancer, 2008.99:789-95.

[43]Ledford H. Cancer theory faces doubts. Nature.2011,472:273.

[44]Pecot CV. A novel platform for detection of CK+ and CK- CTCs[J].Cancer Discov.2011,7:580-6.

[45]Eccles SA &Welch DR. Metastasis: recent discoveries and novel treatment strategies[J]. Lancet,2007,1742-57.

[46]Cormio G.Distant metastases in ovarian carcinoma[J].Int J Gynecol Cancer.2003,2:125-9.

上一篇:关于乳腺囊性增生病 下一篇:小儿脊髓栓系综合症研究进展