俞悅 周愛萍
·專家論壇·
RAS野生型轉(zhuǎn)移性結(jié)直腸癌EGFR抑制劑耐藥機(jī)制的研究進(jìn)展
俞悅 周愛萍
周愛萍,中國醫(yī)學(xué)科學(xué)院腫瘤醫(yī)院內(nèi)科副主任,主任醫(yī)師,從事實(shí)體瘤的內(nèi)科診治工作二十余年,尤其對消化道、泌尿生殖系統(tǒng)腫瘤等的化療和靶向治療有深入和系統(tǒng)的研究。兼任中國臨床腫瘤學(xué)會(CSCO)理事,中國研究型醫(yī)院學(xué)會精準(zhǔn)醫(yī)學(xué)與腫瘤MDT專業(yè)委員會副主任委員,中華醫(yī)學(xué)會腫瘤學(xué)分會結(jié)直腸癌學(xué)組委員,中國老年腫瘤學(xué)分會(CGOS)副干事長、胃腸專業(yè)委員會副主任委員;擔(dān)任《中華肝膽外科雜志》、《中華臨床醫(yī)師雜志》、《中國繼續(xù)醫(yī)學(xué)教育》、《肝癌電子雜志》、Annals of Oncology期刊編委。
抗表皮生長因子受體(epidermal growth factor receptor,EGFR)抗體的應(yīng)用是轉(zhuǎn)移性結(jié)直腸癌治療進(jìn)展中的里程碑??笶G?FR單抗和其他靶向藥物的出現(xiàn)使轉(zhuǎn)移性結(jié)直腸癌患者的中位總生存期從6個月提高至將近30個月,顯著改善轉(zhuǎn)移性結(jié)直腸癌患者的生存質(zhì)量及預(yù)后。目前KRAS和NRAS被公認(rèn)為是抗EGFR治療原發(fā)耐藥的結(jié)直腸癌患者療效預(yù)測標(biāo)志物,用于抗EGFR治療的轉(zhuǎn)移性結(jié)直結(jié)癌患者篩選。除了RAS,其他分子改變也可能影響抗EGFR的療效。即使是抗EGFR治療有效的患者也會在13~18個月間產(chǎn)生獲得性耐藥。本文將對目前已知的抗EGFR治療耐藥機(jī)制進(jìn)行綜述,并展望可能的逆轉(zhuǎn)耐藥策略,以期為轉(zhuǎn)移性結(jié)直腸癌患者精準(zhǔn)分子靶向治療提供依據(jù)和指導(dǎo)。
轉(zhuǎn)移性結(jié)直腸癌 表皮生長因子受體 耐藥 RAS
表皮生長因子受體(epidermal growth factor re?ceptor,EGFR)是HER/ErbB家族的一員,其過表達(dá)可見于包括結(jié)直腸癌在內(nèi)的多種惡性腫瘤中。EGFR通過配體偶聯(lián)、同源二聚化及異源二聚化等方式被激活,從而進(jìn)一步激活RAS/RAF/MARK/ERK和PI3K/AKT/mTOR等信號轉(zhuǎn)導(dǎo)通路,誘導(dǎo)細(xì)胞去分化、快速增殖、凋亡阻滯和新生血管形成[1-2]。西妥昔單抗和帕尼單抗是兩種抗EGFR單克隆抗體(MoAbs),兩者和化療藥物的聯(lián)合應(yīng)用使轉(zhuǎn)移性結(jié)直腸癌患者的中位總生存期從6個月提高至將近30個月,顯著改善轉(zhuǎn)移性結(jié)直腸癌患者的生存質(zhì)量及預(yù)后[3-5]。
RAS是EGFR下游信號分子之一,既往研究已證實(shí)RAS基因是抗EGFR治療原發(fā)耐藥的陽性分子預(yù)測標(biāo)志物[6-7]。即使是RAS野生型的患者,出現(xiàn)腫瘤客觀緩解的不足50%。初始對抗EGFR治療敏感的患者,同樣也會在13~18個月間產(chǎn)生獲得性耐藥[8]。原發(fā)、繼發(fā)性耐藥限制了抗EGFR治療的應(yīng)用。本文通過總結(jié)目前已知的RAS野生型轉(zhuǎn)移性結(jié)直腸癌抗EGFR治療耐藥的分子機(jī)制,以期為受益患者的精確篩選及針對性地制定克服耐藥策略提供依據(jù)。
1.1 BRAF V600E
RAF是RAS的下游因子,5%~9%結(jié)直腸癌患者存在BRAF(V600E)突變,BRAF突變只存在于RAS野生型的患者中[9-11]。非突變型BRAF基因僅在RAS激活后激活,而突變型BRAF基因不受RAS基因控制,處于連續(xù)激活狀態(tài)[12-14]。BRAF在RAS野生型轉(zhuǎn)移性結(jié)直腸癌后線治療中負(fù)性療效預(yù)測作用肯定[15-16],但在一線治療療效預(yù)測方面的作用尚不明確。
BRAF可能在抗EGFR一線治療中發(fā)揮負(fù)性療效標(biāo)志物的作用。Llovet等[17]的研究顯示,在105例接受抗EGFR單抗聯(lián)合化療作為一線治療的KRAS野生型轉(zhuǎn)移性結(jié)直腸癌患者中,BRAF基因突變型和西妥昔治療無效呈顯著相關(guān)(RR=11,P=0.015)。多因素顯示BRAF突變是治療療效唯一負(fù)性預(yù)測因素。既往COIN研究提示在BRAF突變患者一線化療中加用西妥昔單抗可能有不利影響(單純化療組對比聯(lián)合西妥昔組總生存期分別為10.0個月與7.2個月,HR=1.18,P=0.46)[18]。這些結(jié)果未能得到近年一些臨床研究的證實(shí)。在一項(xiàng)包含8項(xiàng)臨床研究共463例BRAF突變轉(zhuǎn)移性結(jié)直腸癌患者M(jìn)eta分析中[19],BRAF突變患者在抗EGFR治療組和標(biāo)準(zhǔn)治療/單純化療組間的客觀緩解率無顯著性差異(RR=1.31,P=0.25)。亞組分析顯示,一線治療中兩組的無進(jìn)展生存期(progress free survival,PFS)(P=0.34)和總生存期(overall survival,OS)(P=0.13)無明顯不同。另一項(xiàng)Meta分析納入了8項(xiàng)隨機(jī)對照臨床試驗(yàn)[20],亞組分析顯示在抗EGFR一線治療中,RAS野生型/BRAF野生型和RAS野生型/BRAF突變型轉(zhuǎn)移性結(jié)直腸癌無論是在OS(HR 0.87 vs.0.89,P=0.96)還是PFS(HR 0.75 vs.0.83,P=0.45)中均無顯著性差異。
綜上所述,鑒于BRAF突變在抗EGFR一線治療療效預(yù)測的研究中未能取得統(tǒng)一結(jié)果,因此即使肯定BRAF抗EGFR后線治療負(fù)性療效預(yù)測作用,2017年美國國立綜合癌癥網(wǎng)絡(luò)(NCCN)結(jié)直腸癌指南依然未將BRAF突變型轉(zhuǎn)移性結(jié)直腸癌患者完全排除在抗EGFR治療之外。指南同時也指出,BRAF突變型轉(zhuǎn)移性結(jié)直腸癌患者從抗EGFR治療中獲益可能性極小,推薦治療前對所有轉(zhuǎn)移性結(jié)直腸癌患者行BRAF基因檢測指導(dǎo)用藥。因此,在臨床實(shí)踐中,BRAF突變型轉(zhuǎn)移性結(jié)直腸癌患者在治療前需要更加謹(jǐn)慎地進(jìn)行方案選擇,盡量避免抗EGFR治療帶來的不利影響。
1.2 PIK3CA
PIK3CA是PI3K的p110-α催化亞單位。PI3K激活進(jìn)一步啟動PI3K/AKT/mTOR通路,促進(jìn)或調(diào)節(jié)增殖、生存、凋亡、遷徙、代謝等細(xì)胞活動[21]。在結(jié)直腸癌中,PIK3CA突變率約為20%,大于80%的PIK3CA突變見于9號外顯子(60%~65%)或20號外顯子(20%~25%)[22]。
PIK3CA在抗EGFR治療療效預(yù)測方面的作用目前尚無統(tǒng)一結(jié)論。一項(xiàng)包含22項(xiàng)研究2 395例KRAS 2號外顯子野生型轉(zhuǎn)移性結(jié)直腸患者的Meta分析顯示[23],PIK3CA突變和西妥昔治療客觀緩解率低顯著相關(guān)(OR=0.39)。然而,在Hsu等[24]的研究中,53例接受西妥昔單抗治療的KRAS野生型轉(zhuǎn)移性結(jié)直腸癌患者中PIK3CA突變組和PIK3CA野生組在治療反應(yīng)率上無顯著性差異(P=0.624)。在另一項(xiàng)包含11項(xiàng)研究864例患者的Meta分析中,研究者發(fā)現(xiàn)僅PIK3CA 20外顯子突變和西妥昔緩解率低顯著相關(guān)(OR=0.21;95%CI:0.05~0.93;P=0.04),而9號外顯子突變則對客觀緩解率無明顯影響(OR=0.54;95%CI:0.26~1.12;P=0.10)[25],提示并非所有PIK3CA突變而是僅PIK3CA20外顯子突變是抗EGFR治療療效的潛在預(yù)測因素。
PIK3CA突變率(特別是PIK3CA 20號外顯子突變率)低且部分PIK3CA突變伴有KRAS或BRAF突變[26]。因此,明確PIK3CA在抗EGFR治療中的具體作用仍較為困難。低突變率也提示PIK3CA在EGFR耐藥中的作用及影響不如BRAF廣泛,使得目前PIK3CA的受關(guān)注程度遠(yuǎn)不如BRAF。
1.3 HER-2
HER-2是人表皮受體因子家族的一員,依靠和同家族的其他配體相關(guān)受體形成異源二聚體發(fā)揮作用。異源二聚體HER-2、HER-3是細(xì)胞內(nèi)信號轉(zhuǎn)導(dǎo)強(qiáng)有力的激活劑[27]。HER-2異??杉せ頔RK1/2通路,并通過使之持續(xù)活化誘導(dǎo)西妥昔耐藥[28]。
HER-2基因擴(kuò)增可能是轉(zhuǎn)移性結(jié)直腸癌西妥昔治療耐藥的原因之一。Barry等[29]發(fā)現(xiàn),在25例帕尼單抗治療的KRAS野生型轉(zhuǎn)移性結(jié)直腸癌中,HER-2擴(kuò)增和帕尼單抗治療抵抗呈顯著相關(guān)(P=0.009)。4例HER-2基因擴(kuò)增患者中3例帕尼單抗治療后疾病進(jìn)展,1例疾病穩(wěn)定。提示HER-2擴(kuò)增可能是西妥昔原發(fā)耐藥的驅(qū)動因素。在另一項(xiàng)研究中,研究者發(fā)現(xiàn)18例西妥昔耐藥患者中1例治療前HER-2陰性的腫瘤組織在西妥昔治療后出現(xiàn)HER-2擴(kuò)增[30],提示HER-2也可能在西妥昔繼發(fā)耐藥中發(fā)揮作用。HER-2擴(kuò)增表明EGFR/HER-2的雙靶點(diǎn)阻斷或許可以逆轉(zhuǎn)RAS野生型轉(zhuǎn)移性結(jié)直腸癌西妥昔耐藥。Sartore-Bianchin等[31]設(shè)計(jì)的Ⅱ期臨床研究(HERA?CLES研究)探索了在KRAS 2號外顯子野生型、HER-2基因擴(kuò)增、既往標(biāo)準(zhǔn)治療(包括抗EGFR治療)耐藥的患者中曲妥珠聯(lián)合拉帕替尼治療的療效。結(jié)果顯示27例HER-2陽性患者中8例取得客觀緩解,達(dá)到設(shè)定的主要終點(diǎn)(緩解率6/27)。2016年美國腫瘤臨床學(xué)會(ASCO)年會報道的臨床研究進(jìn)一步證實(shí)了HER-2擴(kuò)增作為抗EGFR治療負(fù)性療效預(yù)測標(biāo)志物的作用。盡管HER-2擴(kuò)增和未擴(kuò)增的轉(zhuǎn)移性患者一線接受不含抗EGFR治療的PFS無顯著性差異,但在第二、三線抗EGFR治療中,相較于未擴(kuò)增患者,HER-2擴(kuò)增患者的PFS顯著縮短(P<0.001)。
基于上述研究,HER-2擴(kuò)增被看作可能是轉(zhuǎn)移性結(jié)直腸癌患者抗EGFR治療療效預(yù)測的負(fù)性標(biāo)志物,甚至有望成為獨(dú)立于RAS和BRAF V600E突變的另一個轉(zhuǎn)移性結(jié)直腸癌獨(dú)特的亞型。HER-2也是目前抗EGFR耐藥基因改變中唯一有上市藥物可用的靶點(diǎn)。HER-2擴(kuò)增患者可考慮接受聯(lián)合HER-2而非單獨(dú)抗EGFR的治療。
1.4 MET
MET為肝細(xì)胞生長因子(hepatocyte growth fac?tor,HGF)的酪氨酸激酶受體,其過表達(dá)可見于包括結(jié)直腸癌在內(nèi)的多種惡性腫瘤[32]。MET可通過基因擴(kuò)增、激活突變或自分泌刺激等方式被連續(xù)激活,活化的MET可進(jìn)一步激活包括RAS/RAF/MAPK/ERK,PI3K/AKT/mTOR,SRC和STAT3的多條信號通路,誘發(fā)細(xì)胞侵襲性和凋亡抑制[33]。
MET在西妥昔單抗繼發(fā)耐藥中的作用已被肯定。細(xì)胞研究方面,Song等[34]發(fā)現(xiàn),在西妥昔耐藥的KRAS、BRAF、PIK3CA野生型轉(zhuǎn)移性結(jié)直腸癌Caco-2細(xì)胞中,西妥昔誘導(dǎo)的MET磷酸化在用藥24 h后增強(qiáng)、在用藥48 h后達(dá)到頂峰。MET抑制劑(PHA-665752)和MET siRNA處理后的細(xì)胞可恢復(fù)對西妥昔單抗的敏感性。Bardelli等[35]于抗EGFR治療耐藥前后分別取3例轉(zhuǎn)移性結(jié)直腸癌患者的腫瘤組織,結(jié)果顯示3例患者耐藥后的腫瘤組織中MET基因顯著擴(kuò)增,其中2例患者治療前腫瘤組織未見擴(kuò)增,1例患者治療前組織僅見少量的MET擴(kuò)增細(xì)胞,提示抗EG?FR治療對含MET基因擴(kuò)增的腫瘤細(xì)胞來說是分選壓力,MET基因擴(kuò)增是轉(zhuǎn)移性結(jié)直腸癌患者抗EGFR治療繼發(fā)耐藥的原因之一。原發(fā)耐藥方面,Bardelli等[35]的研究中,僅有1%(2/196)患者的腫瘤組織中存在MET基因治療前擴(kuò)增,因而無足夠的數(shù)據(jù)明確MET和抗EGFR原發(fā)耐藥的關(guān)系。同樣,在Cappuzzo等[36]研究中,僅有2.6%(2/81)患者存在MET基因治療前擴(kuò)增,提示MET基因可能只在抗EGFR原發(fā)耐藥中起邊緣作用。
MET基因在抗EGFR治療原發(fā)耐藥患者中擴(kuò)增率低,其改變是否是抗EGFR繼發(fā)耐藥的原因也需要耐藥前后基因結(jié)果的對比來證實(shí),這些因素限制了對MET的進(jìn)一步探索。信號通路間復(fù)雜的交互作用導(dǎo)致MET抑制劑和抗EGFR單抗的聯(lián)合阻斷在Ⅱ期臨床研究中并未取得陽性結(jié)果[37]。因此,盡管MET基因在抗EGFR治療繼發(fā)耐藥的作用肯定,制訂相關(guān)逆轉(zhuǎn)耐藥治療措施以應(yīng)用于臨床,仍需進(jìn)一步的探索。
1.5 甲基化
近年研究提示甲基化水平可能是潛在的抗EG?FR療效預(yù)測標(biāo)志物。Ouchi等[38]對97例接受抗EG?FR治療的KRAS野生型轉(zhuǎn)移性結(jié)直腸癌患者進(jìn)行全基因組甲基化檢測,并根據(jù)檢測結(jié)果將患者分為高甲基化和低甲基化組。臨床數(shù)據(jù)顯示低甲基化組的緩解率(35.7%vs.6.3%,P=0.03)、疾病控制率(75%vs.31.3%,P=0.005)、PFS(HR=0.27;95%CI:0.13~0.57,P<0.001)、OS(HR=00.19;95%CI:0.06~0.54,P<0.001)等各項(xiàng)指標(biāo)均優(yōu)于高甲基化組。高甲基化的患者抗EGFR治療療效較差。
抗EGFR治療的原發(fā)、繼發(fā)性耐藥機(jī)制極大地限制了EGFR抑制劑的應(yīng)用。耐藥腫瘤細(xì)胞的早發(fā)現(xiàn)是克服EGFR耐藥的關(guān)鍵之一。傳統(tǒng)的組織活檢不能反映腫瘤的異質(zhì)性,部分標(biāo)本的不易獲得使得通過活檢方式獲得早期耐藥信息的可能極為有限。作為非侵入性的檢測方法,血液活檢已初步被證實(shí)是監(jiān)測腫瘤復(fù)發(fā)和藥物耐藥性的有效手段[39-40]。血液標(biāo)本中的循環(huán)DNA能有效檢測抗EGFR治療繼發(fā)性耐藥突變,這種抗EGFR治療的分子學(xué)進(jìn)展可比傳統(tǒng)的影像學(xué)進(jìn)展提前數(shù)周出現(xiàn),能為制定個體化的逆轉(zhuǎn)耐藥策略進(jìn)一步爭取時間[41]。耐藥突變早期發(fā)現(xiàn)后可對突變激活通路進(jìn)行多靶點(diǎn)阻斷。臨床前研究顯示,MEK聯(lián)合PIK3CA,EGFR聯(lián)合HER-2等不同靶點(diǎn)阻斷劑的組合可抑制西妥昔耐藥腫瘤細(xì)胞或移植瘤的增殖[42-44]。2016年歐洲臨床腫瘤協(xié)會(ESMO)年會的報道表明,在RAS野生型、BRAF V600E突變型轉(zhuǎn)移性結(jié)直腸癌患者中應(yīng)用BRAF、MEK、EGFR多靶點(diǎn)抑制劑(dabrafenib,D;trametinib,T;panitumumb,P),結(jié)果顯示三靶點(diǎn)阻斷劑無論是有效率(DTP vs.DP vs.TP:18%vs.10%vs.0)還是中位 PFS(DTP vs.DP vs.TP:未達(dá)到vs.3.4個月vs.2.8個月)均長于任意兩藥聯(lián)合。但是,細(xì)胞內(nèi)外的信號轉(zhuǎn)導(dǎo)通路是一個復(fù)雜的關(guān)系網(wǎng)絡(luò),一個或多個信號通路的阻斷可能引起其他信號通路的激活,多靶點(diǎn)阻斷可能僅作為暫時的、過渡的治療策略。此外,多靶點(diǎn)治療還受經(jīng)濟(jì)條件等社會學(xué)因素限制。因此,盡管取得了一定成果,多靶點(diǎn)治療作為逆轉(zhuǎn)耐藥的主要手段在臨床中推廣仍較為困難。
免疫治療為逆轉(zhuǎn)抗EGFR治療耐藥提供了另一種思路。Tran等[45]從轉(zhuǎn)移性結(jié)直腸癌患者腫瘤浸潤淋巴細(xì)胞中分離出以KRAS突變體G12D為靶點(diǎn)的多克隆CD8陽性T細(xì)胞,經(jīng)體外擴(kuò)大培養(yǎng)回輸體內(nèi)后,使患者獲得了臨床上的完全緩解。該研究證實(shí)了腫瘤浸潤淋巴細(xì)胞免疫療法的有效性,使以往對西妥昔治療原發(fā)性耐藥,無藥可治的KRAS突變患者有藥可醫(yī)。另外,臨床前和臨床證據(jù)表明免疫系統(tǒng)能促進(jìn)單克隆抗體在體內(nèi)的治療效果[46],這也使得研究者更加期待正在進(jìn)行中的免疫調(diào)節(jié)劑和檢查點(diǎn)抑制劑聯(lián)合西妥昔單抗一線治療KRAS野生型轉(zhuǎn)移性結(jié)直腸癌患者的臨床試驗(yàn)的最終結(jié)果。
抗EGFR治療顯著改善轉(zhuǎn)移性結(jié)直腸癌患者預(yù)后,是目前轉(zhuǎn)移性結(jié)直腸癌的一線標(biāo)準(zhǔn)治療策略。原發(fā)和繼發(fā)性耐藥極大地限制了抗EGFR治療的應(yīng)用。在精準(zhǔn)醫(yī)療和轉(zhuǎn)化醫(yī)學(xué)背景下,全面了解抗EG?FR治療耐藥的原發(fā)、繼發(fā)機(jī)制不僅能幫助臨床更準(zhǔn)確地篩選出從抗EGFR治療中獲益的目標(biāo)人群,還為不同分子特征的患者制定個體化的逆轉(zhuǎn)耐藥策略提供依據(jù),使抗EGFR治療療效最大化。多靶點(diǎn)抑制的靶向治療是目前逆轉(zhuǎn)耐藥臨時、有效的治療策略之一。未來,血液檢測有望在耐藥細(xì)胞的早期檢測中發(fā)揮主導(dǎo)作用,免疫治療有望在靶向治療之外開辟逆轉(zhuǎn)耐藥的新領(lǐng)域。
[1] Hynes NE,Lane HA.ERBB receptors and cancer:the complexity of targeted inhibitors[J].Nat Rev Cancer,2005,5(5):341-354.
[2] Yarden Y,Sliwkowski MX.Untangling the ErbB signalling network[J].Nat Rev Mol Cell Biol,2001,2(2):127-137.
[3] Bokemeyer C,Bondarenko I,Makhson A,et al.Fluorouracil,leucovorin,and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer[J].J Clin Oncol,2009,27(5):663-671.[4] Douillard JY,Siena S,Cassidy J,et al.Randomized,phase III trial of panitumumab with infusional fluorouracil,leucovorin,and oxaliplatin(FOLFOX4)versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer:the PRIME study[J].J Clin Oncol,2010,28(31):4697-4705.
[5] Van Cutsem E,Peeters M,Siena S,et al.Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer[J].J Clin Oncol,2007,25(13):1658-1664.
[6] Misale S,Di Nicolantonio F,Sartore-Bianchi A,et al.Resistance to anti-EGFR therapy in colorectal cancer:from heterogeneity to convergent evolution[J].Cancer discov,2014,4(11):1269-1280.
[7]Douillard JY,Oliner KS,Siena S,et al.Panitumumab-FOLFOX4 treatment andRASmutationsincolorectal cancer[J].NEngl J Med,2013,369(11):1023-1034.
[8] Amado RG,Wolf M,Peeters M,et al.Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer[J].J Clin Oncol,2008,26(10):1626-1634.
[9] Tol J,Dijkstra JR,Klomp M,et al.Markers for EGFR pathway activation as predictor of outcome in metastatic colorectal cancer patients treated with or without cetuximabr[J].Eur J Cancer,2010,46(11):1997-2009.
[10]Tol J,Nagtegaal ID,Punt CJ.BRAF mutation in metastatic colorectal cancer[J].N Engl J Med,2009,361(1):98-99.
[11]Davies H,Bignell GR,Cox C,et al.Mutations of the BRAF gene in human cancer[J].Nature,2002,417(6892):949-954.
[12]Ikenoue T,Hikiba Y,Kanai F,et al.Functional analysis of mutations within the kinase activation segment of B-Raf in human colorectal tumors[J].Cancer Res,2003,63(23):8132-8137.
[13]Wan PT,Garnett MJ,Roe SM,et al.Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF[J].Cell,2004,116(6):855-867.
[14]Chen D,Huang JF,Liu K,et al.BRAFV600E mutation and its association with clinicopathological features of colorectal cancer:a systematic review and meta-analysis[J].PLoS One,2014,9(3):e90607.
[15]Roock WD,Claes B,Bernasconi D,et al.Effects of KRAS,BRAF,NRAS,and PIK3CA,mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer:a retrospective consortium analysis[J].Lancet Oncol,2010,11(11):753-762.
[16]Seymour MT,Brown SR,Middleton G,et al.Panitumumab and irinotecan versus irinotecan alone for patients with KRAS wild-type,fluorouracil-resistant advanced colorectal cancer(PICCOLO):A prospectively stratified randomised trial[J].Lancet Oncol,2013,14(8):749.
[17]Llovet P,Sastre J,Ortega JS,et al.Prognostic value of BRAF,PI3K,PTEN,EGFR copy number,amphiregulin and epiregulin status in patients with KRAS codon 12 wild-type metastatic colorectal cancer receiving first-line chemotherapy with anti-EGFR therapy[J].Mol Diagn Ther,2015,19(6):397-408.
[18]Maughan TS,Adams RA,Smith CG,et al.Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer:results of the randomised phase 3 MRC COIN trial[J].Lancet,2011,377(9783):2103.
[19]Pietrantonio F,Petrelli F,Coinu A,et al.Predictive role of BRAF mu-tations in patients with advanced colorectal cancer receiving cetuximab and panitumumab:a meta-analysis[J].Eur J Cancer,2015,51(5):587-594.
[20]Rowland A,Dias MM,Wiese MD,et al.Meta-analysis of braf mutation as a predictive biomarker of benefit from anti-egfr monoclonal antibody therapy for ras wild-type metastatic colorectal cancer[J].Br J Cancer,2015,112(12):1888-1894.
[21]Samuels Y,Wang Z,Bardelli A,et al.High frequency of mutations of the PIK3CA gene in human cancers[J].Science,2004,304(5670):554.
[22]Day FL,Jorissen RN,Lipton L,et al.PIK3CA and PTEN gene and exon mutation-specific clinicopathologic and molecular associations in colorectal cancer[J].Clin Cancer Res,2013,19(12):3285-3296.
[23]Therkildsen C,Bergmann TK,Henrichsen-Schnack T,et al.The predictive value of KRAS,NRAS,BRAF,PIK3CA and PTEN for anti-EGFR treatment in metastatic colorectal cancer:A systematic review and meta-analysis.[J].Acta Oncol,2014,53(7):852-864.
[24]Hsu HC,Tan KT,Lu YJ,et al.Mutations of KRAS/NRAS/BRAF predict cetuximab resistance in metastatic colorectal cancer patients[J].Oncotarget,2016,7(16):22257-22270
[25]Huang L,Liu Z,Deng D,et al.Anti-epidermal growth factor receptor monoclonal antibody-based therapy for metastatic colorectal cancer:a meta-analysis of the effect of PIK3CA mutations in KRAS wildtype patients[[J].Arch Med Sci,2014,10(1):1-9.
[26]Ashktorab H,Rosty C,Young JP,et al.PIK3CA activating mutation in colorectal carcinoma:associations with molecular features and survival[J].PLoS One,2013,8(6):e65479.
[27]Holbro T,Beerli RR,Maurer F,et al.The ErbB2/ErbB3 heterodimer functions as an oncogenic unit:ErbB2 requires ErbB3 to drive breast tumor cell proliferation[J].Proc Natl Acad Sci U S A,2003,100(15):8933-8938.
[28]Yonesaka K,Zejnullahu K,Okamoto I,et al.Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab[J].Sci Transl Med,2011,3(99):99ra86.
[29]Barry GS,Cheang MC,Chang HL,et al.Genomic markers of panitumumab resistance including ERBB2/HER2 in a phase II study of KRAS wild-type(wt)metastatic colorectal cancer(mCRC)[J].Oncotarget,2016,7(14):18953-18964.
[30]Takegawa N,Yonesaka K,Sakai K,et al.HER-2 genomic amplification in circulating tumor DNA from patients with cetuximab-resistant colorectal cancer[J].Oncotarget,2016,7(3):3453-3460.
[31]Sartore-Bianchi A,Trusolino L,Martino C,et al.Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory,KRAS codon 12/13 wild-type,HER2-positive metastatic colorectal cancer(HERACLES):a proof-of-concept,multicentre,open-label,phase 2 trial[J].Lancet Oncol,2016,17(6):738-746.
[32]Cecchi F,Rabe DC,Bottaro DP.Targeting the HGF/Met signalling pathway in cancer[J].Eur J Cancer,2010,46(7):1260-1270.
[33]Comoglio PM,Giordano S,Trusolino L.Drug development of MET inhibitors:targeting oncogene addiction and expedience[J].Nat Rev Drug Discov,2008,7(6):504-516.
[34]Song N,Liu S,Zhang J,et al.Cetuximab-Induced MET activation acts as a novel resistance mechanism in colon cancer cells[J].Int J Mol Sci,2014,15(4):5838-5851.
[35]Bardelli A,Corso S,Bertotti A,et al.Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer[J].Cancer Discov,2013,3(6):658-673.
[36]Cappuzzo F,Varella-Garcia M,Finocchiaro G,et al.Primary resistance to cetuximab therapy in EGFR FISH-positive colorectal cancer patients[J].Br J Cancer,2008,99(1):83-89.
[37]Eng C,Bessudo A,Hart LL,et al.A randomized,placebo‐controlled,phase 1/2 study of tivantinib(ARQ 197)in combination with irinotecan and cetuximab in patients with metastatic colorectal cancer with wild‐type KRAS who have received first‐line systemic therapy[J].Int J Cancer,2016,139(1):177-186.
[38]Ouchi K,Takahashi S,Yamada Y,et al.DNA methylation status as a biomarker of anti-epidermal growth factor receptor treatment for metastatic colorectal cancer[J].Cancer Sci,2015,106(12):1722-1729.
[39]Bettegowda C,Sausen M,Leary RJ,et al.Detection of circulating tumor DNA in early-and late-stage human malignancies[J].Sci Transl Med,2014,6(224):224ra24.
[40]Murtaza M,Dawson SJ,Tsui DW,et al.Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA[J].Nature,2013,497(7447):108-112.
[41]Diaz LA Jr,Williams RT,Wu J,et al.The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers[J].Nature,2012,486(7404):537-540.
[42]Bertotti A,Migliardi G,Galimi F,et al.A molecularly annotated platform of patient-derived xenografts("xenopatients")identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer[J].Cancer Discov,2011,1(6):508-523.
[43]Wee S,Jagani Z,Xiang KX,et al.PI3K pathway activation mediates resistance to MEK inhibitors in KRAS mutant cancers[J].Cancer Res,2009,69(10):4286-4293.
[44]Troiani T,Napolitano S,Vitagliano D,et al.Primary and acquired resistance of colorectal cancer cells to anti-EGFR antibodies converge on MEK/ERK pathway activation and can be overcome by combined MEK/EGFR inhibition[J].Clin Cancer Res,2014,20(14):3775-3786.
[45]Tran E,Robbins PF,Lu YC,et al.T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer[J].N Engl J Med,2016,375(23):2255-2262.
[46]Perez EA,Thompson EA,Ballman KV,et al.Genomic analysis reveals that immune function genes are strongly linked to clinical outcome in the North Central Cancer Treatment Group n9831 Adjuvant Trastuzumab Trial[J].J Clin Oncol,2015,33(7):701-708.
(2017-02-01收稿)
(2017-06-09修回)
Advances in research on mechanisms of resistance to anti-epidermal growth factor receptor inhibitors in RAS wild-type metastatic colorectal cancer
Yue YU,Aiping ZHOU
Aiping ZHOU;E-mail:zhouap1825@126.com
Department of Medical Oncology,Cancer Hospital of Chinese Academy of Medical Sciences,Beijing,Beijing 100021,China
The development of anti-epidermal growth factor receptor monoclonal antibodies(anti-EGFR McAbs)marked a significant milestone in metastatic colorectal cancer(mCRC)treatment.The addition of anti-EGFR McAb can greatly improve quality of life of mCRC patients and mCRC prognosis and markedly increases the overall survival rate from 6 months to nearly 30 months.KRAS and NRAS mutations contribute to the primary resistance to anti-EGFR therapy and can serve as well-established predictive markers for patient selection.Apart from the RAS family,other molecular alteration in EGFR signaling pathway may also compromise the efficacy of anti-EGFR treatment.In addition,patients who responded to anti-EGFR treatment eventually develop acquired drug resistance within 13 and 18 months.In this review,the mechanisms underlying the primary and secondary resistance to anti-EGFR therapy are summarized,and a possible strategy to circumvent drug resistance is proposed.We hope this review can provide compelling evidence,deeper insights,and reasonable guidance to facilitate the precise molecular targeted therapy of mCRC.
metastatic colorectal cancer,epidermal growth factor receptor,drug resistance,RAS
10.3969/j.issn.1000-8179.2017.11.089
北京協(xié)和醫(yī)學(xué)院中國醫(yī)學(xué)科學(xué)院腫瘤醫(yī)院腫瘤內(nèi)科(北京市100021)
周愛萍 zhouap1825@126.com
俞悅 專業(yè)方向?yàn)槟[瘤內(nèi)科臨床與基礎(chǔ)研究。
E-mail:yuyueyykk123@163.com