王曉楠,程民,胡世蓮
(安徽醫(yī)科大學(xué)附屬安徽省立醫(yī)院、安徽省立醫(yī)院,安徽省老年醫(yī)學(xué)研究所,腫瘤免疫與營(yíng)養(yǎng)治療安徽省重點(diǎn)實(shí)驗(yàn)室,安徽省循證醫(yī)學(xué)中心,合肥 230001)
·綜述·
循環(huán)腫瘤DNA在胃癌發(fā)生發(fā)展中的作用
王曉楠,程民,胡世蓮
(安徽醫(yī)科大學(xué)附屬安徽省立醫(yī)院、安徽省立醫(yī)院,安徽省老年醫(yī)學(xué)研究所,腫瘤免疫與營(yíng)養(yǎng)治療安徽省重點(diǎn)實(shí)驗(yàn)室,安徽省循證醫(yī)學(xué)中心,合肥 230001)
近年來(lái),胃癌的發(fā)病率和死亡率居高不下,早期發(fā)現(xiàn)、早期診斷、早期治療是防治胃癌的關(guān)鍵。隨著現(xiàn)代醫(yī)學(xué)發(fā)展,腫瘤標(biāo)志物已應(yīng)用于臨床診斷,但由于腫瘤標(biāo)志物靈敏度和特異度較低,臨床應(yīng)用局限,難以滿足"精準(zhǔn)醫(yī)療"要求。研究表明,循環(huán)腫瘤DNA(ctDNA)檢測(cè)技術(shù)是一種快速、高效、相對(duì)無(wú)創(chuàng)性的新型"液體活檢"技術(shù),能夠?qū)崟r(shí)監(jiān)測(cè)機(jī)體腫瘤狀態(tài)、轉(zhuǎn)移、復(fù)發(fā)及預(yù)后評(píng)估。本文闡述ctDNA與胃癌的關(guān)系,介紹目前常用的ctDNA檢測(cè)技術(shù),探討ctDNA在胃癌臨床診療中的應(yīng)用價(jià)值。
胃腫瘤;腫瘤細(xì)胞,循環(huán);基因檢測(cè);分子靶向治療
胃癌是人類(lèi)最常見(jiàn)的惡性腫瘤之一[1],嚴(yán)重威脅人們的生命健康。近年來(lái),腫瘤的個(gè)體化診療已經(jīng)取得很好的效果,改善了患者的預(yù)后,但由于缺乏早期診斷的特異性標(biāo)志物和有效的治療手段,胃癌患者的復(fù)發(fā)和轉(zhuǎn)移率仍居高不下。因此,胃癌的早診、早治是臨床亟待解決的問(wèn)題。研究顯示,在腫瘤形成的早期階段,腫瘤組織可釋放腫瘤細(xì)胞和DNA至外周血,形成循環(huán)腫瘤細(xì)胞(CTC)及循環(huán)腫瘤DNA(ctDNA)。ctDNA的水平與腫瘤發(fā)生發(fā)展具有相關(guān)性,檢測(cè)腫瘤患者血液中ctDNA可以提供快速、高效、相對(duì)無(wú)創(chuàng)性的“液體活檢”[2],這為腫瘤的早期診斷和治療提供新的研究方向。
ctDNA與胃癌、乳腺癌、肺癌等多種腫瘤的發(fā)生發(fā)展關(guān)系密切[3-5],其與胃癌的關(guān)系倍受關(guān)注,Sai等[6]采用實(shí)時(shí)熒光定量PCR(RTQ-PCR)對(duì)53例胃癌患者血漿ctDNA濃度檢測(cè)發(fā)現(xiàn)胃癌組血漿DNA濃度明顯高于健康組,晚期胃癌患者的血漿DNA濃度高于早期胃癌患者,說(shuō)明ctDNA濃度與胃癌的進(jìn)展有關(guān),與臨床病理類(lèi)型無(wú)關(guān)。胃癌的發(fā)生與基因改變有關(guān),對(duì)胃癌患者的腫瘤組織和相應(yīng)血漿中的ctDNA進(jìn)行基因啟動(dòng)子區(qū)的甲基化檢測(cè),發(fā)現(xiàn)多數(shù)胃癌患者都存在一些基因啟動(dòng)子區(qū)高甲基化的現(xiàn)象。有研究[7]檢測(cè)73例胃癌患者血漿中腫瘤細(xì)胞TFPI2甲基化,采用實(shí)時(shí)熒光定量甲基化特異PCR(RTQ-MSP)檢出7例TFPI2甲基化,提示TFPI2甲基化在胃癌患者中存在,在健康對(duì)照組中并未檢出。再對(duì)樣本進(jìn)行RTQ-MSP法分析,發(fā)現(xiàn)淋巴結(jié)轉(zhuǎn)移和遠(yuǎn)處轉(zhuǎn)移的患者中TFPI2甲基化程度更高,提示其與胃癌的轉(zhuǎn)移有關(guān)。RUNX3是一種腫瘤抑制因子,RUNX3啟動(dòng)子異常甲基化與包括胃癌在內(nèi)的很多腫瘤的發(fā)生有關(guān)。Sakakura等[8]采用RTQ-MSP法檢測(cè)胃癌患者血漿ctDNA的RUNX3甲基化,發(fā)現(xiàn)29%(19/65)的患者外周血中檢測(cè)到RUNX3高甲基化,經(jīng)手術(shù)治療后患者血漿的RUNX3甲基化水平比術(shù)前降低12倍。對(duì)所有的術(shù)前和術(shù)后胃癌患者血漿RUNX3甲基化水平進(jìn)行前瞻性研究發(fā)現(xiàn),術(shù)后的胃癌患者RUNX3甲基化水平比術(shù)前下降83%,并且RUNX3甲基化程度與腫瘤的分期、淋巴結(jié)轉(zhuǎn)移、血管侵襲有關(guān),敏感性高于CEA。Kolesnikova[9]和Tani[10]采用甲基化特異性聚合酶鏈?zhǔn)椒磻?yīng)法(MSP)檢測(cè)MGMT,p15,和Hm-LH1的甲基化程度,發(fā)現(xiàn)20例胃癌患者M(jìn)GMT,p15,和Hm-LH1的甲基化程度分別為50%、70%、25%,而晚期胃癌患者中MGMT,p15,和Hm-LH1甲基化程度分別為90%、90%、60%,在健康對(duì)照組中并未檢測(cè)到異常甲基化。核苷酸切除修復(fù)交叉互補(bǔ)基因1(ERCC1)是一種DNA損傷修復(fù)基因,王紅兵等[11]采用甲基化特異性PCR技術(shù)(MSP法)檢測(cè)30例胃癌患者外周血、胃癌組織中ERCC1基因啟動(dòng)子CpG島甲基化狀態(tài)。結(jié)果顯示胃癌組織中ERCC1基因啟動(dòng)子CpG島甲基化率為76.7%,外周血ctDNA的ERCC1基因啟動(dòng)子CpG島甲基化率為63.3%,提示胃癌患者外周血中的ERCC1基因啟動(dòng)子CpG島甲基化率與胃癌組織中相似,同時(shí)發(fā)現(xiàn)ctDNA與原發(fā)腫瘤基因組DNA具有相同的遺傳變異。檢測(cè)外周血ctDNA可發(fā)現(xiàn)腫瘤相關(guān)基因的變異,從而反映腫瘤組織的特征性改變。
1999年,García-Olmo在結(jié)腸癌腫瘤小鼠模型中發(fā)現(xiàn)腫瘤小鼠的血漿可改變?nèi)斯づ囵B(yǎng)的細(xì)胞,因此提出:原發(fā)腫瘤病灶的致癌基因通過(guò)轉(zhuǎn)染使易感細(xì)胞經(jīng)過(guò)血液循環(huán)轉(zhuǎn)移到遠(yuǎn)處的靶器官,這個(gè)假設(shè)稱(chēng)為“基因轉(zhuǎn)移”[12]。Holmgren用實(shí)驗(yàn)證明凋亡小體基因中的DNA能夠轉(zhuǎn)移到吞噬細(xì)胞內(nèi),這種轉(zhuǎn)移DNA可以一直在巨噬細(xì)胞內(nèi)存在。由此推論,位于血液循環(huán)中的腫瘤細(xì)胞的凋亡小體被吞噬細(xì)胞攝取后,釋放發(fā)生變異的ctDNA重新進(jìn)入其他細(xì)胞轉(zhuǎn)染基因,并修改受體細(xì)胞的基因組成,即具有轉(zhuǎn)化潛能的腫瘤DNA發(fā)生遷移,造成遠(yuǎn)處組織器官轉(zhuǎn)移的機(jī)制稱(chēng)為“基因轉(zhuǎn)移”[13]。這種機(jī)制與腫瘤的擴(kuò)散有關(guān),機(jī)體內(nèi)此類(lèi)轉(zhuǎn)化細(xì)胞應(yīng)該具有干細(xì)胞潛能,因?yàn)樗麄兺┘?xì)胞相類(lèi)似,都具有自我更新能力、分化及轉(zhuǎn)移到不同組織內(nèi)等特點(diǎn)。ctDNA可能通過(guò)“基因轉(zhuǎn)移”機(jī)制參與了結(jié)腸癌、胃癌等的腫瘤復(fù)發(fā)與轉(zhuǎn)移[14]。
血清中ctDNA的量比血漿中高3~24倍,但血漿更適合用來(lái)分析ctDNA,可能是血清受溶解細(xì)胞DNA的污染而影響ctDNA的相對(duì)水平[15]。目前常用檢測(cè)ctDNA方法有熒光染料法、實(shí)時(shí)熒光定量PCR(RTQ-PCR)、液滴式數(shù)字PCR(ddPCR)和新一代靶向測(cè)序(NGS)等。由于所檢測(cè)DNA靶標(biāo)的不同,造成檢測(cè)結(jié)果亦存在一定的差異,每種方法都有優(yōu)缺點(diǎn)[16]。熒光染料檢測(cè)法敏感性高、重復(fù)性好,適合于大規(guī)模人群篩查和臨床診斷,但是特異性不強(qiáng)[17-18]。RTQ-PCR可以定量研究,應(yīng)用范圍廣,但是不同的DNA片段,檢測(cè)條件也不相同,需要反復(fù)實(shí)驗(yàn),而且依賴(lài)于熒光信號(hào)和標(biāo)準(zhǔn)曲線,只能分析較少且已經(jīng)確定的突變點(diǎn)[19]。ddPCR是將樣本通過(guò)微滴發(fā)生器進(jìn)行微滴化處理后進(jìn)行PCR反應(yīng),再用微滴檢測(cè)器對(duì)每個(gè)微滴逐個(gè)檢測(cè),最終經(jīng)分析軟件計(jì)算出目標(biāo)DNA分子的濃度,有以下優(yōu)點(diǎn):第一,ddPCR采用終點(diǎn)檢測(cè),直接計(jì)算目的序列的拷貝數(shù),無(wú)需依賴(lài)于Ct值和標(biāo)準(zhǔn)曲線就可以進(jìn)行精確的定量檢測(cè)。第二,ddPCR敏感度和特異度更高,可以篩查0.001%的突變頻率,較qPCR的敏感性增加了1000倍,廣泛地應(yīng)用于稀有突變檢測(cè)、拷貝數(shù)變異分析和復(fù)雜樣本基因表達(dá)檢測(cè)等方面[20-21]。NGS可在全基因組進(jìn)行高通量的檢測(cè),速度快、檢測(cè)范圍更廣,并且提高了檢測(cè)的準(zhǔn)確性,但存在一定的假陽(yáng)性比率,需進(jìn)一步驗(yàn)證。NGS主要用于復(fù)發(fā)或病情惡化前進(jìn)行早期預(yù)測(cè),以ctDNA-NGS為基礎(chǔ)的分析方法可通過(guò)獲取新興突變片段或克隆選擇從而制定一個(gè)新的治療方案[22]。
3.1 早期診斷 腫瘤早期診斷是提高腫瘤治愈率,降低腫瘤患者病死率的重要途徑。有些惡性腫瘤沒(méi)有可靠的蛋白質(zhì)生物標(biāo)志物,有些缺乏特異性,而且大多數(shù)的蛋白質(zhì)生物標(biāo)志物在循環(huán)中持續(xù)存在數(shù)周,難以準(zhǔn)確評(píng)估病情。ctDNA的半衰期短(約2h),特異性強(qiáng),可動(dòng)態(tài)評(píng)估癌癥情況[23]。Kim等[24]研究30例胃癌患者和34例健康對(duì)照組血漿ctDNA的水平,觀察胃癌患者術(shù)前和手術(shù)24 h后的ctDNA,發(fā)現(xiàn)晚期胃癌患者ctDNA濃度高于早期胃癌組,早期胃癌組高于健康對(duì)照組;相比于術(shù)前,胃癌術(shù)后患者的ctDNA的含量明顯下降。Park等[25]對(duì)54例胃癌患者和59例健康對(duì)照組研究發(fā)現(xiàn),胃癌組的ctDNA濃度是對(duì)照組的2.4倍,提示血漿ctDNA可作為胃癌早期診斷的腫瘤標(biāo)志物。腫瘤形成的早期,在外周血中檢測(cè)到脫落的ctDNA,有助于腫瘤的早期發(fā)現(xiàn)、早期診斷,并針對(duì)性治療。
3.2 療效評(píng)估 隨著治療手段的多樣化,選擇安全可靠的療效評(píng)估手段以驗(yàn)證新療法的有效性。目前臨床常用療效監(jiān)測(cè)方法有血清學(xué)指標(biāo)(如腫瘤標(biāo)志物)和影像學(xué)檢查(如B超、CT、MRI)均存在很多不足。Bai等[26]檢測(cè)非小細(xì)胞肺癌患者化療前ctDNA的EGFR突變比例為34.5%。但經(jīng)化療治療后,下降為23.1%。而這些由EGFR突變陽(yáng)性轉(zhuǎn)變?yōu)殛幮缘幕颊邔?duì)化療藥物表現(xiàn)出較好的反應(yīng)性和敏感性。研究對(duì)比ctDNA、CTC、CA153和影像學(xué)檢查監(jiān)測(cè)轉(zhuǎn)移性乳腺癌疾病進(jìn)展,發(fā)現(xiàn)ctDNA比其他指標(biāo)能更早的反映疾病進(jìn)展和藥物治療反應(yīng)[27]。用血漿標(biāo)本追蹤腫瘤特異性的突變基因的動(dòng)力學(xué)變化,ctDNA能夠反映疾病的臨床進(jìn)程[28]。
3.3 預(yù)后判斷 腫瘤的分期及細(xì)胞的分化程度是胃癌生存期預(yù)測(cè)的主要依據(jù),其中TNM臨床分期與疾病診斷、治療方案、患者預(yù)后息息相關(guān)。觀察結(jié)直腸癌患者術(shù)后2~5年ctDNA的水平,結(jié)果顯示術(shù)后檢測(cè)到ctDNA的患者病情復(fù)發(fā),而陰性的患者沒(méi)有復(fù)發(fā)[29]。將ctDNA與TNM分期結(jié)合,在治療前后動(dòng)態(tài)監(jiān)測(cè)ctDNA水平,便于臨床醫(yī)師評(píng)估療效及更改治療方案。
3.4 靶向治療 ctDNA最重要的臨床應(yīng)用之一就是根據(jù)腫瘤特定基因改變采用特定相應(yīng)的分子靶向治療,靶向治療不僅對(duì)患者身心的傷害小而且對(duì)腫瘤的治療效果將遠(yuǎn)比傳統(tǒng)方法明顯[30]。甲基化的基因治療可以預(yù)防腫瘤的發(fā)生,Qu等[31]研究發(fā)現(xiàn)5-氮-2′-脫氧胞苷可以逆轉(zhuǎn)人乳腺癌細(xì)胞PDLIM2啟動(dòng)子的甲基化狀態(tài),恢復(fù)PDLIM2表達(dá),并能抑制乳腺癌細(xì)胞的致瘤作用。未來(lái)幾年以基因治療為基礎(chǔ)的分子靶向治療將會(huì)是臨床研究應(yīng)用的熱點(diǎn)[32]。
ctDNA攜帶的腫瘤基因組信息與腫瘤組織具有良好的一致性,能克服常規(guī)腫瘤組織活檢所無(wú)法突破的腫瘤異質(zhì)性問(wèn)題。ctDNA檢測(cè)簡(jiǎn)單無(wú)創(chuàng),被稱(chēng)為“液體活檢”。但是目前仍存在以下問(wèn)題:在對(duì)同一類(lèi)ctDNA定量研究發(fā)現(xiàn)各項(xiàng)研究數(shù)據(jù)之間有較大差異,這可能與ctDNA提取和定量方法不同或者腫瘤患者納入和排除標(biāo)準(zhǔn)不同有關(guān)[33]。研發(fā)具有特異性和敏感性的技術(shù)仍是應(yīng)用于臨床檢測(cè)的關(guān)鍵。簡(jiǎn)便、花費(fèi)小的方法將有助于ctDNA的檢測(cè)技術(shù)盡快進(jìn)入臨床。ctDNA在胃癌轉(zhuǎn)移的早期診斷、治療、療效評(píng)估和預(yù)后分析等方面具有重要意義,有利于針對(duì)腫瘤患者實(shí)施個(gè)體化綜合治療。
[1] CORREIA M,MACHADO JC,RISTIMIKI A.Basic aspects of gastric cancer[J].Helicobacter,2009,14(S1):36-40.
[2] LGNATIADIS M,LEE M,JEFFREY SS.Circulating tumor cells and circulating tumor DNA:challenges and opportunities on the path to clinical utility[J].Clin Cancer Res,2015,21(21) :4786-4800.
[3] HAMAKAWA T,KUKITA Y,KUROKAWA Y,et al.Monitoring gastric cancer progression with circulating tumour DNA[J].Br J Cancer,2015,112(2):352-356.
[4] OPENSHAW MR,PAGE K,FERNANDEZ-GARCIA D,et al.The role of ctDNA detection and the potential of the liquid biopsy for breast cancer monitoring[J].Expert Rev Mol Diagn,2016,16(7):751-755.
[5] SANTARPIA M,KARACHALIOU N,GONZáLEZ-CAO M,et al.Feasibility of cell-free circulating tumor DNA testing for lung cancer[J].Biomark Med,2016,10(4):417-430.
[6] SAI S,ICHIKAWA D,TOMITA H,et al.Quantification of plasma cell-free DNA in patients with gastric cancer[J].Anticancer Res,2007,27(4C):2747-2751.
[7] HIBI K,GOTO T,SHIRAHATA A,et al.Detection of TFPI2 methylation in the serum of gastric cancer patients[J].Anticancer Res,2011,31(11):3835-3838.
[8] SAKAKURA C,HAMADA T,MIYAGAWA K,et al.Quantitative analysis of tumor-derived methylated RUNX3 sequences in the serum of gastric cancer patients[J].Anticancer Res,2009,29(7):2619-2625.
[9] KOLESNIKOVA EV,TAMKOVICH SN,BRYZGUNOVA OE,et al.Circulating DNA in the blood of gastric cancer patients[J].Ann N Y Acad Sci,2008,1137(1):226-231.
[10] TANI N,ICHIKAWA D,IKOMA D,et al.An early detection of recurrence using reverse transcriptase-polymerase chain reaction (RT-PCR) and methylation-specific polymerase chain reaction (MSP) from peripheral blood in patients after gastrectomy[J].Gan To Kagaku Ryoho,2006,33(12):1720-1722.
[11] 王紅兵,陳衛(wèi)昌.胃癌患者外周血與胃癌組織中ERCC1基因甲基化的關(guān)系及其意義[J].中國(guó)癌癥雜志,2013,23(11):900-903.
[12] GARCíA-OLMO D,GARCíA-OLMO DC.Functionality of circulating DNA:the hypothesis of genometastasis[J].Ann N Y Acad Sci,2001,945(1):265-275.
[13] GARCíA-OLMO DC,RUIZ-PIQUERAS R,GARCíA-OLMO D.Circulating nucleic acids in plasma and serum (CNAPS) and its relation to stem cells and cancer metastasis:state of the issue[J].Histol Histopathol,2004,19(2):575-583.
[14] GOESSL C.Diagnostic potential of circulating nucleic acids for oncology[J].Expert Rev Mol Diagn,2003,3(4):431-442.
[15] JUNG M,KLOTZEK S,LEWANDOWSKI M,et al.Changes in concentration of DNA in serum and plasma during storage of blood samples[J].Clin Chem,2003,49(6 Pt 1):1028-1029.
[16] DEVONSHIRE AS,WHALE AS,GUTTERIDGE A,et al.Towards standardisation of cell—free DNA measurement in plasma:controls for extraction efficiency,fragment size bias and quantification[J].Anal Bioanal Chem,2014,406(26):6499-6512.
[17] 傅士龍,屠紅,張國(guó)玲,等.卵巢上皮性癌患者循環(huán)DNA的定量研究[J].中國(guó)實(shí)用婦科與產(chǎn)科雜志,2015,21(11):659-662.
[18] BJ?RKMAN L,REICH CF,PISETSKY DS.The use of fluorometric assays to assess the immune response to DNA in murine systemic lupus erythematosus[J].Scand J Immunol,2003,57(6):525-533.
[19] OPENSHAW MR,PAGE K,FERNANDEZ-GARCIA D,et al.The role of ctDNA detection and the potential of the liquid biopsy for breast cancer monitoring[J].Expert Rev Mol Diagn,2016 ,16(7):751-755.
[20] TALY V,PEKIN D,BENHAIM L,et al.Multiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patients[J].Clin Chem,2013,59(12):1722-1731.
[21] OXNARD GR,PAWELETZ CP,KUANG Y,et al.Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA[J].Clin Cancer Res,2014,20(6):1698-1705.
[22] LIANOS GD,MANGANO A,KOURAKLIS G,et al.Dynamic sequencing of circulating tumor DNA:novel noninvasive cancer biomarker[J].Biomark Med,2014,8(5):629-632.
[23] DIEHL F,SCHMIDT K,CHOTI MA,et al.Circulating mutant DNA to assess tumor dynamics[J].Nat Med,2008,14(9):985-990.
[24] KIM K,SHIN DG,PARK MK,et al.Circulating cell-free DNA as a promising biomarker in patients with gastric cancer:diagnostic validity and significant reduction of cfDNA after surgical resection[J].Ann Surg Treat Res,2014,86(3):136-142.
[25] PARK JL,KIM HJ,CHOI BY,et al.Quantitative analysis of cell-free DNA in the plasma of gastric cancer patients[J].Oncol Lett,2012,3(4):921-926.
[26] BAI H,WANG ZJ,CHEN KN,et al.Influence of chemotherapy on EGFR mutation status among patients with non-small-cell lung cancer[J].J Chn Oncol,2012,30(25):3077-3083.
[27] DAWSON SJ,TSUI DW,MURTAZA M,et al.Analysis of circulating tumor DNA to monitor metastatic breast cancer[J].N Engl J Med,2013,368(13):1199-1209.
[28] FORSHEW T,MURTAZA M,PARKINSON C,et al.Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA[J].Sci Transl Med,2012,4(136):2805.
[29] DIEHL F,SCHMIDT K,CHOTI MA,et al.Circulating mutant DNA to assess tumor dynamics[J].Nat Med,2008,14(9):985-990.
[30] ALIX-PANABIèRES C,PANTEL K.Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy[J].Cancer Discov,2016,6(5):479-491.
[31] QU Z,FU J,YAN P,et al.Epigenetic repression of PDZ-LIM do-main-containing protein 2:Implications for the biology and treatment of breast cancer[J].J Biol Ehem,2010,285(16):11786-11792.
[32] CAI FF,KOHLER C,ZHANG B,et al.Epigenetic therapy for breast cancer[J].Int J Mol Sci,2011,12(7):4465-4487.
[33] DEVONSHIRE AS,WHALE AS,GUTTERIDGE A,et al.Towards standardisation of cell-free DNA measurement in plasma:controls for extraction efficiency,fragment size bias and quantification[J].Anal Bioanal Chem,2014,406(26):6499-6512.
The role of tumor circulating DNA in the development of gastric cancer and research progress
Wang Xiaonan, Cheng Min, Hu Shilian
(Anhui Provincial Hospital, Anhui Institute of Gerontology, Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Anhui Evidence-based Medicine Center, Hefei 230001, China)
HuShilian,Email:hushilian@126.com
With increasing morbidity and mortality of gastric cancer in recent years, the early detection, diagnosis and treatment are critical to the prophylaxis and therapy of gastric cancer nowadays. Tumor markers have gain growing concern to its clinical applications but still confined to the sensitivity and specificity which make it hard to fit for "precision medicine". Some research have shown that circulating tumor DNA(ctDNA) detection is a kind of "liquid biopsy " technology which is rapid、effective and noninvasive. The state of tumor, including metastasis, recurrence and prognosis can be monitored by detecting ctDNA. This review focuses on the relationship between ctDNA and gastric cancer, as well as the detection technology commonly used to explore the application of ctDNA in clinical diagnosis and treatment of gastric cancer.
Stomach neoplasms;Neoplastic cells,circulating;Genetic testing;Molecular targeted therapy
國(guó)家自然科學(xué)基金(81071808);安徽省自然科學(xué)基金面上項(xiàng)目(1408085MH167);安徽省國(guó)際科技合作項(xiàng)目(1303063017)
王曉楠,碩士在讀,Email:1006445947@qq.com
胡世蓮,主任醫(yī)師,教授,博士生導(dǎo)師,Email:hushilian@126.com
R735.2
A
10.3969/J.issn.1672-6790.2017.01.035
2016-08-17)