韓淑燕,鄭文獻,何曦冉,趙 燦,蔣姍彤,龐麗娜,李曉紅,李萍萍(北京大學腫瘤醫(yī)院暨北京市腫瘤防治研究所中西醫(yī)結(jié)合科惡性腫瘤發(fā)病機制及轉(zhuǎn)化研究教育部重點實驗室,北京 100142)
?
消癌平注射液聯(lián)合吉非替尼對耐藥非小細胞肺癌H460和H1975裸鼠移植瘤的抑制作用
韓淑燕,鄭文獻,何曦冉,趙 燦,蔣姍彤,龐麗娜,李曉紅,李萍萍
(北京大學腫瘤醫(yī)院暨北京市腫瘤防治研究所中西醫(yī)結(jié)合科惡性腫瘤發(fā)病機制及轉(zhuǎn)化研究教育部重點實驗室,北京 100142)
摘要:目的 研究消癌平注射液(Xap)聯(lián)合吉非替尼(Gef)對耐藥非小細胞肺癌(NSCLC)細胞H460 和H1975裸鼠移植瘤的抑制作用及機制。方法 BALB/c裸鼠皮下接種人NSCLC的H460或H1975細胞,待腫瘤體積達到50~100 mm3,分別ip給予Xap 5 g·kg-1、ig給予Gef 50 mg·kg-1或Xap+Gef(Xap+Gef組),連續(xù)給藥21 d。每7 d測量腫瘤體積2~3次,繪制腫瘤增殖曲線,計算腫瘤增殖率,并記錄體質(zhì)量。取腫瘤組織,稱取瘤重,計算腫瘤抑制率。免疫組化法和Western蛋白印跡法檢測腫瘤組織中血管生成標志物血管內(nèi)皮生長因子(VEGF)和內(nèi)皮糖蛋白(CD105)及耐藥相關(guān)蛋白磷脂酰肌醇3-激酶(PI3K)、蛋白激酶B(Akt)、哺乳動物西羅莫司(雷帕霉素)靶蛋白(mTOR)和磷酸化胞外信號調(diào)節(jié)激酶(ERK)的表達。結(jié)果 與模型對照組相比,單用Xap或Gef對H460和H1975裸鼠移植瘤的生長無顯著影響,而兩藥聯(lián)合能顯著抑制裸鼠的瘤體積〔H460組:(1103±340)vs(3020±450)mm3,H1975組:(487±153)vs(1269± 161)mm3,P<0.05〕、降低瘤重〔H460組:(1.20±0.52)vs(2.78±0.93)g,H1975組:(0.52±0.32)vs (0.92±0.42)g,P<0.05〕,腫瘤增殖率和抑制率分別為42.1%,43.5%(H460,P<0.01)和43.0%,52.5% (H1975,P<0.01)。與單用Xap或Gef相比,聯(lián)合用藥組能顯著性地降低瘤重和腫瘤增殖率,提高腫瘤抑制率(P<0.05)。免疫組化結(jié)果表明,單用Xap或Gef對腫瘤血管新生標志物VEGF和CD105及耐藥相關(guān)蛋白p-PI3K,p-ERK,p-Akt和p-mTOR的表達無影響,而聯(lián)合用藥卻能明顯降低H460和H1975腫瘤組織中VEGF,CD105,p-ERK,p-Akt和p-mTOR的表達。Western蛋白印跡結(jié)果表明,與模型對照組相比,聯(lián)合用可顯著抑制p-PI3K及其下游蛋白p-Akt,p-ERK和p-mTOR蛋白的表達(P<0.01),并且優(yōu)于單用Xap或Gef(P<0.05)。結(jié)論 聯(lián)合Xap可顯著提高Gef在H460和H1975裸鼠移植瘤的療效,該協(xié)同作用部分是通過抑制腫瘤血管新生和PI3K及其下游信號分子等耐藥相關(guān)蛋白的表達實現(xiàn)的。
關(guān)鍵詞:消癌平注射液;吉非替尼;非小細胞肺癌;腫瘤移植
DOl:10.3867/j.issn.1000-3002.2016.01.007
近年來,針對肺腺癌驅(qū)動基因的靶向藥物已成為晚期非小細胞肺癌(non-small cell lung cancer,NSCLC)治療的發(fā)展方向。吉非替尼(gefitinib,Gef)等小分子酪氨酸激酶抑制劑(tyrosine kinase inhibitor,TKI)能顯著延長中位生存期[1],給晚期NSCLC患者的治療帶來曙光,美國國家綜合癌癥網(wǎng)絡(luò)(National Comprehensive Cancer Network,NCCN)指南推薦EGFR突變患者可一線使用EGFR-TKI[2]。雖然TKI對表皮生長因子受體(epidermal growth factor receptor,EGFR)突變的晚期NSCLC療效較好,但經(jīng)過6~12個月的緩解期后仍會發(fā)生耐藥出現(xiàn)疾病進展[3]。耐藥已成為制約TKI靶向藥療效的主要瓶頸,是治療過程亟需解決的問題。
有關(guān)Gef耐藥的機制尚未闡明,目前已明確的主要機制包括EGFR的T790M突變、c-met基因的擴增和k-ras基因突變等[4-5]。對于Gef耐藥后的主要臨床挽救措施包括厄洛替尼(erlotinib)[6]和化療[7],c-Met抑制劑和不可逆EGFR抑制劑[8]等分子抑制劑的應(yīng)用尚在研究中。盡管上述方法可在一定的程度上使Gef耐藥患者獲益,但不可避免地會發(fā)生再次耐藥[9]。如二代TKI阿法替尼對T790M突變患者有效,但耐藥性仍然是影響其療效的重要因素[10]。臨床上迄今尚缺乏解決Gef等TKI耐藥的有效藥物。因此,尋找低毒、高效的逆轉(zhuǎn)Gef耐藥的途徑仍然是臨床的迫切需求。若能從當前已在臨床應(yīng)用的抗腫瘤藥物中篩選,則更會縮短研究周期,降低研發(fā)費用。
Gef耐藥后會出現(xiàn)EGFR下游信號通路磷酸化磷脂酰肌醇3-激酶(phosphatidylinositol 3-kinase,phospho-PI3K)、磷酸化蛋白激酶B(phospho-Akt,p-AKT)、磷酸化哺乳動物西羅莫司(雷帕霉素)靶蛋白〔phospho-mammalian target of sirolimus(Rapamycin),p-mTOR〕和磷酸化胞外信號調(diào)節(jié)激酶(extracellular signal-regulated kinase,p-ERK)的持續(xù)活化[11-13]。通過抑制PI3K/Akt/mTOR通路和(或)ERK的活化,特別是當同時抑制2條通路時,可有效逆轉(zhuǎn)NSCLC對Gef的耐藥,提高耐藥細胞對Gef的響應(yīng)[14-16]。
消癌平(Xiaoaiping,Xap)是中藥通關(guān)藤的水提醇沉制劑,在臨床上可用于肺癌、消化道腫瘤和白血病等多種惡性腫瘤,該藥聯(lián)合化療對NSCLC的療效優(yōu)于單純化療。我們的前期研究表明,Xap注射液能顯著改善NSCLC對Gef的耐藥,提高腫瘤對Gef的敏感性,降低Gef的IC50,促進細胞凋亡,抑制耐藥細胞株的PI3K/Akt/mTOR通路和ERK的活化[17-18]。但Xap改善Gef耐藥的作用及其機制尚有待進一步驗證和闡明。前期在耐藥NSCLC荷瘤裸鼠上進行了Xap劑量的比較結(jié)果[18],選取具有微弱抑瘤作用的Xap 5 g·kg-1與Gef 50 mg·kg-1聯(lián)合,研究Xap改善Gef耐藥的作用。
1.1藥品和試劑
Gef,購自英國阿斯利康公司,用二甲亞砜(DMSO)配置成20 mmol·L-1的儲備液;Xap,南京圣和藥業(yè)有限公司(201107041)。Matrigel,美國BD公司;RPMI 1640培養(yǎng)基,天津百若克醫(yī)藥生物技術(shù)有限責任公司;胎牛血清(fetal bovine serum,F(xiàn)BS),美國Gibco公司;BCA蛋白定量試劑盒,美國Pierce公司。增殖細胞核抗原(proliferating cell nuclear antigen,PCNA)(1∶200)抗體,美國Eptomics公司;血管內(nèi)皮生長因子(vascular endothelial growth factor,VEGF)抗體(1∶100)和內(nèi)皮糖蛋白(endoglin,CD105)抗體(1∶100),美國Santa Cluz公司;磷酸化PI3K(phospho-PI3K,p-PI3K)(1∶1000),p-Akt抗體(Ser473)(1∶2000),p-ERK抗體(Thr202/Tyr204)(1∶2000),p-mTOR抗體(Ser2448)(1∶2000),β肌動蛋白抗體(1∶5000),甘油醛-3-磷酸脫氫酶(GAPDH)抗體(1∶5000),美國CST公司。辣根過氧化物酶標記的山羊抗兔和山羊抗小鼠IgG二抗購自北京中杉金橋公司,聚偏二氟乙烯(polyvinylidene fluoride,PVDF)膜和ECL購自美國Millipore公司。
1.2主要儀器
B5060EK-CO2型二氧化碳培養(yǎng)箱,德國Heraeus公司;1285型生物安全柜,美國Thermo Electron公司;Allegra 64R型高速低溫臺式離心機,美國Beckman Coulter公司;IMJ-213型Olympus倒置顯微鏡,MDF-U4086S型超低溫冰箱(-80℃),日本Sanyo;Western蛋白印跡電泳和轉(zhuǎn)膜系統(tǒng),美國Bio-Rad公司。
1.3細胞培養(yǎng)
人NSCLC細胞H460(k-ras突變)和H1975 (EGFR-L858R/T790M突變)購自美國ATCC細胞庫,以含10%FBS的RPMI 1640培養(yǎng)基于含5% CO2的孵箱中37℃進行培養(yǎng)。
1.4動物、模型制備及分組給藥
BALB/c-nu/nu雄性裸小鼠,體質(zhì)量18~22 g,購自中國醫(yī)學科學院基礎(chǔ)所實驗動物中心,合格證編號:SYXK(京)2006-0005,按SPF級動物實驗要求分籠飼養(yǎng)與管理。
常規(guī)培養(yǎng)H460和H1975細胞,待細胞融合度達到80%~90%時,0.25%胰酶消化細胞,PBS重懸,加入等體積的matrigel混勻,每只裸鼠皮下接種0.2 mL,即3×105個細胞。每周2次測量腫瘤體積,同時記錄動物的體質(zhì)量。腫瘤體積計算公式為:V= 1/2×a×b2,式中a為長徑,b為短徑。計算相對腫瘤體積(relative tumor volume,RTV)=Vt/V0,式中Vt是每次測量時的腫瘤體積,V0是分組給藥前的腫瘤體積;處理組RTV與模型對照組RTV比值的百分比為相對腫瘤增殖率(T/C),T/C>60%為無效,≤60%為有效。待腫瘤體積達到50~100 mm3,將裸鼠隨機分為4組,每組8只:模型對照組(不含通關(guān)藤提取物的注射液溶劑)、Xap 5 g·kg-1組(ip給予)、Gef 50 mg·kg-1組(ig給予)和Xap 5 g·kg-1+Gef 50 mg·kg-1組,給藥21 d。實驗結(jié)束,取腫瘤組織部分固定于4%多聚甲醛中,部分凍存于液氮中,待做后續(xù)檢測。
1.5免疫組化檢測腫瘤組織中VEGF,CD105,p-Akt,p-mTOR和p-ERK的表達
取多聚甲醛固定的H460和H1975裸鼠腫瘤組織,制成石蠟切片。3%H2O2消除內(nèi)源性過氧化物酶活性,微波抗原修復,室溫封閉20 min,加入一抗4℃過夜,二抗孵育后經(jīng)DAB顯色,顯微鏡下觀察結(jié)果。以胞漿或細胞核染成棕黃色或棕褐色為陽性反應(yīng),每個標本隨機觀察5個高倍鏡視野(×200),計算每個高倍鏡視野100個腫瘤細胞中的陽性細胞數(shù),取其平均值作為陽性細胞數(shù),計算陽性細胞所占百分比。
1.6Western蛋白印跡法檢測腫瘤組織的PCNA,p-AKT,p-mTOR和p-ERK蛋白表達
取凍存于液氮中的H460和H1975裸鼠腫瘤組織,預(yù)冷PBS洗滌,液氮中研磨后加入含有蛋白酶抑制劑和磷酸酶抑制劑的RIPA裂解液,冰上裂解腫瘤組織20 min,18 800×g離心20 min,收集上清液即為細胞總蛋白。BCA法測定蛋白含量,調(diào)整樣品蛋白含量為2 g·L-1。取20 μg蛋白進行SDSPAGE電泳,蛋白電轉(zhuǎn)移到PVDF膜上,以5%脫脂牛奶或5%BSA室溫封閉1 h,加入一抗4℃孵育過夜。TBST洗膜4~5次,用辣根過氧化物酶標記的二抗孵育1 h,TBST洗膜后以ECL化學發(fā)光檢測試劑盒檢測蛋白印跡。以β肌動蛋白或GAPDH進行上樣量校正,以目標蛋白與內(nèi)標蛋白積分吸光度值的比值表示蛋白的相對表達量。
1.7統(tǒng)計學分析
Tab.1 Effect of Xiaoaiping injection(Xap)combined with gefitinib(Gef)on body mass and tumor growth of H460 xenografts
2.1Xap+Gef對耐藥非小細胞肺癌H460和H1975移植瘤裸鼠腫瘤生長及體質(zhì)量的影響
表1結(jié)果顯示,H460移植瘤裸鼠給藥組的腫瘤體積及相對腫瘤增殖率均優(yōu)于單用Xap和Gef組(P<0.05)。與模型組相比,Xap組和Gef組的瘤重無統(tǒng)計學差異,聯(lián)合用藥組顯著降低(P<0.05),腫瘤抑制率為52.5%(P<0.01),瘤重及抑制率均優(yōu)于單獨應(yīng)用各組(P<0.05)。表2的結(jié)果顯示,H1975移植瘤裸鼠體質(zhì)量無顯著差異。與模型組相比,Xap和Gef單用組的腫瘤體積無顯著變化,聯(lián)合用藥組的腫瘤體積明顯縮?。≒<0.05),腫瘤增殖率顯著低于模型組(P<0.01)。
2.2Xap+Gef對耐藥非小細胞肺癌H460和H1975移植瘤裸鼠移植瘤組織VEGF,CD105,p-AKT,p-mTOR和p-ERK表達的影響
圖1結(jié)果顯示,與腫瘤模型對照組相比,單用Xap或Gef對H460移植瘤裸鼠腫瘤組織的VEGF,CD10,耐藥相關(guān)蛋白p-AKT,p-mTOR和p-ERK的表達無影響,Xap+Gef組能明顯抑制腫瘤組織中的VEGF,CD105,p-AKT,p-mTOR和p-ERK的表達(P<0.01),聯(lián)合用藥組的抑制作用顯著強于單用組(P<0.05)。
Fig.1 Effect of Xap combined with Gef on expression of vascular endothelial growth factor(VEGF),endoglin (CD105),phospho-Akt(p-AKT),phospho-mammalian target of sirolimus(rapamycin)(p-mTOR)and extracellular signal-regulated kinase(p-ERK)in tumor tissue of H460 non-small cell lung cancer(NSCLC)xenografts deter-nimined by immunohistochemistry(×200).See Tab.1 for the treatment.B was the semiquantitative result of A.±s,n=8.**P<0.01,compared with model group;#P<0.05,compared with Xap+Gef group.
圖2結(jié)果是顯示,與腫瘤模型對照組相比,單用Xap或Gef對H1975移植瘤裸鼠腫瘤組織的VEGF,CD105,耐藥相關(guān)蛋白p-AKT,p-mTOR和p-ERK的表達無影響,Xap+Gef組能明顯抑制腫瘤組織中的VEGF,CD105,p-AKT,p-mTOR和p-ERK的表達的表達(P<0.01),聯(lián)合用藥組的抑制作用顯著強于單用組(P<0.05)。表明Xap+Gef具有抑制H460和H1975移植腫瘤血管生成和耐藥相關(guān)蛋白表達的作用。
Fig.2 Effect of Xap combined with Gef on expression of VEGF,CD105,p-AKT,p-mTOR and p-ERK in tumor tis-sue of H1975 NSCLC xenografts deternimined by immunohistochemistry(×200).See Tab.1 for the treatment.B was the semiquantitative result of A.±s,n=8.**P<0.01,compared with model group;#P<0.05,compared with Xap+Gef group.
2.3Xap+Gef對耐藥非小細胞肺癌H460和H1975移植瘤裸鼠腫瘤組織PCNA,p-Pl3K,p-AKT,p-ERK和p-mTOR蛋白表達的影響
Western蛋白印跡結(jié)果顯示(圖3),與模型對照組相比,Xap 5 g·kg-1組和Gef 50 mg·kg-1組對H460裸鼠腫瘤組織中的PCNA,p-PI3K,p-Akt,p-ERK和p-mTOR(圖3A)蛋白相對表達無明顯影響。而Xap 5 g·kg-1+Gef 50 mg·kg-1組可顯著降低H460和H1975腫瘤組織中PCNA,p-PI3K,p-Akt,p-ERK和p-mTOR的表達水平(P<0.01),并且優(yōu)于單用Xap或Gef(P<0.05)(圖3B,C)。
圖4的結(jié)果表明,與模型對照組相比,Xap 5 g·kg-1組和Gef 50 mg·kg-1組對H1975移植瘤裸鼠腫瘤組織中PCNA,p-PI3K,p-Akt,p-ERK和p-mTOR蛋白相對表達量無明顯影響。而Xap 5 g·kg-1+Gef 50 mg·kg-1組可顯著降低H1975腫瘤組織中上述蛋白的表達水平(P<0.01),并且優(yōu)于單用Xap或Gef(P<0.05)(圖4B,C)。
Fig.3 Effect of Xap combined with Gef on expression of p-Pl3K,p-Akt,p-ERK and p-mTOR in tumor tissue of H460 NSCLC xenografts determined by Western blotting.See Tab.1 for the treatment.B and C were the semiquantitative re-sults of A.±s,n=8.**P<0.01,compared with model group;#P<0.05,compared with Xap+Gef group.
Fig.4 Effect of Xap combined with Gef on expression of p-Pl3K,p-Akt,p-ERK and p-mTOR in tumor tissue of H1975 NSCLC xenografts determined by Western blotting.See Tab.1 for the treatment.B and C were the semiquantitative results of A.±s,n=8.**P<0.01,compared with model group;#P<0.05,compared with Xap+Gef group.
本研究發(fā)現(xiàn),Xap+Gef處理能顯著抑制耐藥NSCLC細胞H460和H1975裸鼠移植瘤的生長,降低腫瘤體積和瘤重,表明Xap+Gef能抑制腫瘤細胞的增殖,提高Gef在耐藥NSCLC裸鼠移植瘤中的療效。
本研究中,免疫組化和Western蛋白印跡結(jié)果均顯示,Xap+Gef可顯著降低H460和H1975裸鼠移植瘤組織中PCNA的表達,表明耐藥NSCLC腫瘤細胞的增殖受到抑制。PCNA的表達除了作為評價細胞增殖狀態(tài)的一個指標外,近來的研究亦提示,PCNA作為細胞生長、死亡和修復等過程的協(xié)調(diào)者,有可能成為腫瘤治療的潛在靶點[19]。
腫瘤的增殖受多種因素的調(diào)控,其中腫瘤血管的生成發(fā)揮了重要作用。腫瘤血管生成不僅提供腫瘤生長所需的營養(yǎng),而且有利于腫瘤細胞通過血管發(fā)生轉(zhuǎn)移。VEGF是重要的血管形成分子,是目前已知作用最強、特異性最高的促血管生長因子。VEGF在大多數(shù)惡性腫瘤中均表達增加,是與腫瘤轉(zhuǎn)移和預(yù)后密切相關(guān)的重要細胞因子[20]。CD105在腫瘤組織的未成熟血管中呈高表達,而在正常血管內(nèi)皮細胞卻無表達,所以CD105是一種特異性表達于新生腫瘤血管的內(nèi)皮細胞標志物,在標記腫瘤組織微血管密度方面顯著優(yōu)于CD31和CD34等標志物[21]。相關(guān)分析顯示CD105和VEGF的表達呈正相關(guān)[22]。在本試驗中,Xap+Gef組能顯著降低VEGF和CD105的表達,說明兩藥聯(lián)用具有抑制NSCLC腫瘤血管生成的作用。
驅(qū)動基因發(fā)生突變是NSCLC對TKI靶向藥物發(fā)生原發(fā)性或獲得性耐藥的重要原因之一。H1975細胞因存在EGFR-T790M突變而耐藥,而NCIH460細胞因存在k-ras突變而對Gef原發(fā)耐藥。Gef耐藥后會出現(xiàn)PI3K/AKT和ERK等通路的持續(xù)活化,同時抑制相關(guān)通路的活化則能有效增強Gef療效。在本研究中,Xap+Gef能顯著降低耐藥相關(guān)蛋白p-PI3K,p-Akt,p-ERK和p-mTOR的表達,提示兩藥聯(lián)用能同時抑制PI3K/Akt/mTOR和MEK/ ERK通路的活化。PI3K可被細胞膜上的各種酪氨酸激酶受體激活,同時也能被Ras蛋白(Raf/MEK/ ERK通路上游的蛋白因子)所激活,PI3K被激活后通過其下游的Akt/mTOR等對細胞生長和蛋白質(zhì)合成、血管生成、炎癥反應(yīng)等均有促進作用[23-25]。由此可見,PI3K處于PI3K/Akt/mTOR和MEK/ERK兩條信號通路的上游,Xap+Gef下調(diào)耐藥相關(guān)蛋白的表達主要是通過抑制PI3K的活化實現(xiàn)的。
綜上,本研究表明Xap注射液能增強Gef在H460和H1975細胞荷瘤裸鼠的療效,提高耐藥NSCLC對Gef的敏感性,抑制腫瘤細胞的增殖,減少腫瘤血管生成。抑制EGFR下游信號通路PI3K/ Akt/mTOR和ERK的活化可能為Xap提高Gef療效的作用機制之一,是今后將繼續(xù)進行深入研究的機制。本研究在體外細胞實驗的基礎(chǔ)上,在動物體內(nèi)研究了Xap注射液改善Gef耐藥的作用及機制,為應(yīng)用Xap注射液改善Gef等抗腫瘤靶向藥的耐藥提供了實驗依據(jù),同時為鞏固或提高Gef的療效進行了新的探索。
參考文獻:
[1]Sequist LV,Martins RG,Spigel D,Grunberg SM,Spira A,J?nne PA,et al.First-line gefitinib in patientswithadvancednon-small celllung cancer harboring somatic EGFR mutations[J].J Clin Oncol,2008,26(15):2442-2449.
[2]Ettinger DS, Akerley W,Bepler G,Blum MG,Chang A,Cheney RT,et al.Non-small cell lung cancer[J].J Natl Compr Canc Netw,2010,8 (7):740-801.
[3]Hoshi S,Yamaguchi T,Kono C, Amano H,Yamada Y.Recurrenceof non-small cell lung cancer after successful treatment with gefitinibreport of three cases[J].Gan To Kagaku Ryoho,2004,31(8):1209-1213.
[4]Kosaka T,Yamaki E,Mogi A,Kuwano H.Mecha-nisms of resistance to EGFR TKIs and develop-ment of a new generation of drugs in non-small cell lung cancer[J].J Biomed Biotechnol,2011,2011:165214.doi:10.1155/2011/165214.
[5]Wang S,An T,Wang J,Zhao J,Wang Z,Zhuo M,et al.Potential clinical significance of a plasmabased KRAS mutation analysis in patients with advanced non-smallcelllungcancer[J].Clin Cancer Res,2010,16(4):1324-1330.
[6]Kaira K,Naito T,Takahashi T,Ayabe E,Shimoyama R,Kaira R,et al.Pooled analysis of the reports of erlotinib after failure of gefitinib for non-smallcelllungcancer[J].Lung Cancer,2010,68(1):99-104.
[7]Wu YL,Yang JJ,Huang YJ,Zhou Q,Huang YS,Xu CR.Docetaxel as salvage chemotherapy in patients with advanced non-small cell lung cancer after failure of cytotoxic agents and gefitinib treatment[J].Chin-German J Clin Oncol,2008,7(9):495-499.
[8]Miller VA,Hirsh V,Cadranel J,Chen YM,Park K,Kim SW,et al.Afatinib versus placebo for patients with advanced,metastatic non-small-cell lung cancer after failure of erlotinib,gefitinib,or both,and one or two lines of chemotherapy(LUX-Lung 1):a phase 2b/3 randomised trial[J].Lancet Oncol,2012,13(5):528-538.
[9]Ercan D, Zejnullahu K, Yonesaka K,Xiao Y,Capelletti M,Rogers A,et al.Amplification of EGFR T790M causes resistance to an irreversible EGFR inhibitor[J].Oncogene,2010,29(16):2346-2356.
[10]Azuma K,Kawahara A,Sonoda K,Nakashima K,Tashiro K,Watari K,et al.FGFR1 Activation is an escape mechanism in human lung cancer cells resistant to afatinib,a pan-EGFR family kinase inhibitor[J].Oncotarget,2014,5(15):5908-5919.
[11]Lopiccolo J, Blumenthal GM, Bernstein WB,Dennis PA.Targeting the PI3K/Akt/mTOR pathway:effective combinations and clinical considerations [J].Drug Resist Updat,2008,11(1/2):32-50.
[12]Miller KA,Yeager N,Baker K,Liao XH,Refetoff S,Di Cristofano A.Oncogenic Kras requires simulta-neous PI3K signaling to induce ERK activation and transform thyroid epithelial cells in vivo[J].Cancer Res,2009,69(8):3689-3694.
[13]EngelmanJA,Jaenne PA.Mechanismsof acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer[J].Clin Cancer Res,2008,14(10):2895-2899.
[14]Janmaat ML,Rodriguez JA,Gallegos-Ruiz M,Kruyt FA,Giaccone G.Enhanced cytotoxicity induced by gefitinib and specific inhibitors of the Ras or phosphatidyl inositol-3 kinase pathways in non-small cell lung cancer cells[J].Int J Cancer,2006,118(1):209-214.
[15]Milton DT, Riely GJ, Azzoli CG,Gomez JE,Heelan RT,Kris MG,et al.Phase 1 trial of everolimus and gefitinib in patients with advanced nonsmallcell lung cancer[J].Cancer,2007,110(3):599-605.
[16]La Monica S,Galetti M,Alfieri RR,Cavazzoni A,Ardizzoni A,Tiseo MA,et al.Everolimus restores gefitinib sensitivity in resistant non-small cell lung cancer cell lines[J].Biochem Pharmacol,2009,78(5):460-468.
[17]Han SY,Zhao MB,Zhuang GB,Li PP.Marsdenia tenacissima extract restored gefitinib sensitivity in resistant non-small cell lung cancer cells[J].Lung Cancer,2012,75(1):30-37.
[18]Han SY,Zhao W,Sun H,Zhou N,Zhou F,An G,et al.Marsdenia tenacissima extract enhances gefitinib efficacy in non-small cell lung cancer xenografts [J].Phytomedicine,2015,22(5):560-567.
[19]Wang SP.A silent housekeeper or a potential ther-apeutic target[J].Trends Pharmacol Sci,2014,35 (4):178-186.
[20]Hoeben A,Landuyt B,Highley MS,Wildiers H,Van Oosterom AT,De Bruijn EA.Vascular endo-thelial growth factor and angiogenesis[J].Pharmacol Rev,2004,56(4):549-580.
[21]Tanaka F,Otake Y,Yanagihara K,Kawano Y,Miyahara R,Li M,et al.Evaluation of angiogenesis in non-small cell lung cancer:comparison between anti-CD34 antibody and anti-CD105 antibody[J]. Clin Cancer Res,2001,7(11):3410-3415.
[22]Mineo TC,Ambrogi V,Baldi A,Rabitti C,Bollero P,Vincenzi B,et al.Prognostic impact of VEGF,CD31,CD34,and CD105 expression and tumour vessel invasionafter radical surgery for IB-ⅡA non-smallcelllung cancer[J].J Clin Pathol,2004,57(6):591-597.
[23]Fruman DA,Rommel C.PI3K And cancer:lessons,challenges and opportunities[J].Nat Rev Drug Discov,2014,13(2):140-156.
[24]Liu P,Cheng H,Roberts TM,Zhao JJ.Targeting the phosphoinositide 3-kinase pathway in cancer [J].Nat Rev Drug Discov,2009,8(8):627-644.
[25]Ebi H,Costa C,F(xiàn)aber AC,Nishtala M,Kotani H,Juric D,et al.PI3K Regulates MEK/ERK signaling in breast cancer via the Rac-GEF,P-Rex1[J]. Proc Natl Acad Sci USA,2013,110(52):21124-21129.
(本文編輯:喬虹)
Xiaoaiping injection combined with gefitinib inhibits resistant non-small cell lung cancer xenografts H460 and H1975
HAN Shu-yan,ZHENG Wen-xian,HE Xi-ran,ZHAO Can,JIANG Shan-tong,PANG Li-Na,LI Xiao-hong,LI Ping-ping
〔Key Laboratory of Carcinogenesis and Translational Research(Ministry of Education),Department of Integration of Chinese and Western Medicine,Peking University Cancer Hospital&Institute,Beijing 100142,China〕
Abstract:OBJECTlVE To study the antitumor activity and underlying mechanisms of Xiaoaiping injection(Xap)combined with gefitinib(Gef)in nude mice bearing resistant non-small cell lung cancer (NSCLC)cells H460 or H975.METHODS BALB/c nude mice were inoculated with human NSLCL cells H460 or H1975 and the drug treatment did not start until the tumor volume reached 50-100 mm3. The tumor bearing mice were divided into four groups:control group,Xap group(5 g·kg-1,ip),Gef group(50 mg·kg-1,ig),and Xap plus Gef group.All the administration lasted for 21 d continuous. Tumor volumes were measured two or three times per week,and the body weight of animals was recorded.At the end of the test,tumors were weighed after the sacrifice of mice.Tumor inhibition rate and relative tumor proliferation rate were calculated based on tumor weight and tumor volume.The related biomarkers and proteins in tumor tissues were detected by immunohistochemistry and Western blot assay,respectively.RESULTS Compared with the control group,no significant effect was observed on the growth of H460 and H1975 xenografts in groups of Xap or Gef alone in nude mice.However,the twodrug combination significantly suppressed tumor volume,with(1103±340)versus(3020±450)mm3for H460 and(487±153)versus(1269±161)mm3for H1975,respectively(P<0.05).The combined Xap and Gef application also significantly reduced the tumor weight,with(1.20±0.52)versus(2.78±0.93)g for H460 and(0.52±0.32)versus(0.92±0.42)g for H1975,respectively(P<0.05).The relative tumor proliferation rate and inhibition rate in the combination group was 42.1%and 43.5%for H460(P<0.01),43.0% and 52.5%for H1975(P<0.01).Compared with Xap and Gef drug alone,their combination showed significant difference in reducing tumor weight,suppressing tumor proliferation rate and increasing tumor inhibition rate(P<0.05).Immunohistochemistry results showed that each drug alone had no effect on tumor angiogenesis markers of vascular endothelial growth factor(VEGF)and CD105 expression,or on drug resistance related proteins of p-ERK,p-Akt and p-mTOR,whereas the combination of Xap and Gef obviously reduced the expression of these biomarkers in H460 and H1975 tumor tissues.The decreased drug resistance related proteins of p-PI3K and its downstream molecules p-Akt,p-ERK and p-mTOR by the two-drug combination were also confirmed by Western blot results(P<0.01,compared with control),and showed significant difference compared with each single treatment(P<0.05). CONCLUSlON The addition of Xap significantly improves the antitumor activity of Gef in H460 and H1975 xenografts,and this synergistic effect may be ascribed to the inhibition of tumor angiogenesis,the downregulation of PI3K and its downstream signaling molecules which are associated with drug resistance.
Key words:Xiaoaiping injection;geftinib;non-small cell lung cancer;neoplasm transplantation
中圖分類號:R979.1,R734.2
文獻標志碼:A
文章編號:1000-3002(2016)01-0044-09
Foundation item:The project supported by National Natural Science Foundation of China(81274148);and Specialized Research Fund for Doctoral Program of Higher Education(20110001120100) LI Ping-ping,Tel:(010)88196069,E-mail:lppma123@sina.com
收稿日期:(2015-08-03接受日期:2015-12-31)
基金項目:國家自然科學基金(81274148);高等學校博士學科點專項科研基金資助課題(20110001120100)
作者簡介:韓淑燕,女,副教授,主要從事中藥抗腫瘤的活性物質(zhì)基礎(chǔ)及作用機制研究;李萍萍,女,教授,博士生導師,主要從事中西醫(yī)結(jié)合臨床研究。
通訊作者:李萍萍,E-mail:lppma123@sina.com,Tel:(010)88196706