錢昱臻,張明睿,任剛,鄒偉
1.遼寧師范大學(xué) 生命科學(xué)學(xué)院,遼寧 大連 116081;2.遼寧省生物技術(shù)與分子藥物研發(fā)重點實驗室,遼寧 大連 116081;3.中國醫(yī)學(xué)科學(xué)院 醫(yī)藥生物技術(shù)研究所,北京 100050
嵌合抗原受體構(gòu)建與腫瘤治療的研究進(jìn)展
錢昱臻1,2,張明睿1,2,任剛3,鄒偉1,2
1.遼寧師范大學(xué) 生命科學(xué)學(xué)院,遼寧 大連 116081;2.遼寧省生物技術(shù)與分子藥物研發(fā)重點實驗室,遼寧 大連 116081;3.中國醫(yī)學(xué)科學(xué)院 醫(yī)藥生物技術(shù)研究所,北京 100050
多年來,癌癥治療一直是困擾人類的難題之一,常規(guī)的治療手段都各有局限。隨著基因轉(zhuǎn)移和細(xì)胞培養(yǎng)技術(shù)的更新?lián)Q代,細(xì)胞免疫治療逐漸走入公眾視線。嵌合抗原受體(chimeric antigen receptor,CAR)修飾的T淋巴細(xì)胞(CAR-T)治療是幾年來腫瘤過繼免疫治療的新方法,能特異性識別靶抗原,并殺傷腫瘤細(xì)胞。我們將CAR-T細(xì)胞在腫瘤治療中的應(yīng)用及構(gòu)建CAR-T細(xì)胞的策略做簡要綜述。
嵌合抗原受體;T淋巴細(xì)胞;腫瘤治療
多年來,腫瘤治療一直是困擾人類的最大難題之一,常規(guī)治療手段都各有局限。隨著基因轉(zhuǎn)移和細(xì)胞培養(yǎng)技術(shù)的更新?lián)Q代,細(xì)胞免疫治療逐漸引起人們的重視。1989年,Gross及Goverman等[1-2]提出了一個新的想法,引入細(xì)胞毒性T細(xì)胞雜交瘤細(xì)胞作為基因材料用以構(gòu)建一種抗體識別抗原的模型。1991年,Romeo和Irving等構(gòu)建了與信號分子相連的編碼CD4或CD25的嵌合受體,證實了單一的多肽鏈的確能夠復(fù)制大部分T細(xì)胞受體(T cell receptor,TCR)的定位特征[3-4]。上述基礎(chǔ)研究共同證明了重組T細(xì)胞不依賴MHC限制性,在特異性地定位靶抗原,對抗人類腫瘤上是具有可能性的。近年來,嵌合抗原受體T細(xì)胞(chimeric antigen receptor T cell,CAR-T)作為一種新的免疫細(xì)胞治療方式,在腫瘤臨床治療,尤其是在血液癌等非固態(tài)瘤的癌癥治療中,取得了突破性進(jìn)展。2016年初,《The Guardian》報道,美國弗雷德·哈欽森癌癥研究中心發(fā)現(xiàn)CAR-T可作為特定白血病的治療新方法,在不同國家進(jìn)行的早期臨床試驗效果明顯,超過90%的病人癥狀完全消失。在此,我們簡要綜述了嵌合抗原受體(chimeric antigen receptors,CARs)結(jié)構(gòu)、CAR-T細(xì)胞在腫瘤治療中的應(yīng)用及局限性,以及目前構(gòu)建CAR-T細(xì)胞的策略,旨在為臨床研究應(yīng)用提供依據(jù)。
利用基因工程方法將能與腫瘤抗原特異性結(jié)合的受體、細(xì)胞跨膜的部分和細(xì)胞內(nèi)信號轉(zhuǎn)導(dǎo)的部分結(jié)合起來,構(gòu)成新的結(jié)構(gòu),稱為CARs。用之修飾T細(xì)胞,賦予其特異性的腫瘤殺傷能力即為CAR-T細(xì)胞。簡單來說,CARs最基本的結(jié)構(gòu)由單鏈可變區(qū)(single chain fragment variable,scFv)的胞外配體結(jié)合域通過跨膜結(jié)構(gòu)域與胞內(nèi)信號域連接組成(圖1)[5]。細(xì)胞外的配體結(jié)合域一般為單鏈抗體,決定了CARs的特異性;胞內(nèi)信號域最初為T細(xì)胞活化基序,即免疫受體酪氨酸活化基序(immunoreceptor tyrosine-based activa? tion motif,ITAM),起到激活T細(xì)胞的作用,通常為CD3ζ和FcεRIγ。這種模式化的結(jié)構(gòu)方式從第一代CARs(僅有一個CD3ζ結(jié)構(gòu)域)開始一直沿用至今,只有形式上的改進(jìn),從未有結(jié)構(gòu)上的顛覆。但是,僅僅由ITAM發(fā)出的激活信號只能引起T細(xì)胞的短暫分裂和較低水平的細(xì)胞因子分泌,不能提供大量的單克隆擴增和長時間的抗腫瘤效應(yīng)[6-7]。因此,根據(jù)T細(xì)胞活化雙信號學(xué)說,在第一代CARs的基礎(chǔ)上,加入了新的單一或更多的共刺激分子(costimulatory molecule,CM),第二、三代CARs應(yīng)運而生(圖1)。與第一代CAR-T細(xì)胞相比,第二、三代CAR-T細(xì)胞毒性、增殖活性、存活時間、細(xì)胞因子釋放等均顯著增加[8-11]。而Pegram等[12]以第二代CARs為基礎(chǔ),以CD19為靶抗原,構(gòu)建了一種能分泌IL-12的新型CAR-T細(xì)胞,提出了第四代CARs的新構(gòu)想。這種新型CAR-T細(xì)胞在未用環(huán)磷酰胺預(yù)處理的情況下,具有抗CD19+腫瘤的作用。這就說明新型四代CARs可以不借助強毒副作用的放化療藥物,具有擴展CAR-T細(xì)胞的臨床應(yīng)用范圍,減少患者所受傷害的可能性。
圖1 癌癥免疫療法——CAR-T療法的細(xì)胞結(jié)構(gòu)示意圖
2.1 CAR-T細(xì)胞在腫瘤治療中的應(yīng)用
早在2006年便首次公開報道了將CAR-T細(xì)胞技術(shù)應(yīng)用于臨床試驗的相關(guān)研究[13-14],其后越來越多的文章相繼發(fā)表,提出了很多有價值的見解[15-19]。Porter等在《New England Journal of Med?icine》上報道了關(guān)于CAR-T細(xì)胞成功治療慢性淋巴細(xì)胞白血病(chronic lymphocytic leukemia,CLL)的臨床研究,3例CLL患者經(jīng)過CD19-CART細(xì)胞靶向治療后,檢測發(fā)現(xiàn)CAR-T細(xì)胞能在體內(nèi)擴增1000倍以上,可維持6個月的治療周期,而其中2例患者的病情完全得到緩解[15]。Till等,用逆轉(zhuǎn)錄病毒構(gòu)建出第三代CAR-T細(xì)胞(CD20-CD28-CD137-CD3ζ),用于治療非霍奇金淋巴瘤。臨床結(jié)果顯示,第三代CAR-T細(xì)胞在體內(nèi)存活時間超過12個月[17],較第二代CAR-T細(xì)胞6個月的存活周期相比具有更強的效用時間。
B細(xì)胞抗原CD19是典型的CARs抗原,其表達(dá)于正常B細(xì)胞與多數(shù)B細(xì)胞惡性腫瘤,而在其他組織和尚未發(fā)育完全的B細(xì)胞中不表達(dá),因此CD19可以作為一個CAR-T細(xì)胞持續(xù)作用的生物標(biāo)志物。CD19是已知最好的,也是最常用的靶向分子。賓夕法尼亞大學(xué)以抗CD19嵌合抗原受體T細(xì)胞為基礎(chǔ),開發(fā)出第一個針對特定癌癥(急性B細(xì)胞惡性腫瘤)的個性化細(xì)胞治療方式——CTL019。2014年美國食品和藥物管理局授予CTL019“突破療法”稱號。截至2015年底,已有100多個患者接受抗CD19基因重組T細(xì)胞治療,并且越來越多的關(guān)于該療法的初步臨床試驗結(jié)果被發(fā)表。目前,還有一些針對頑固惡性實體瘤的實驗在積極進(jìn)行中,目的是完善CAR-T細(xì)胞療法,使其不僅僅局限于B細(xì)胞惡性腫瘤的治療。
截止于2016年3月,CAR-T細(xì)胞在淋巴瘤、白血病臨床治療中已獲得了令人矚目的成績,全球范圍內(nèi)有記錄可查的CAR-T研發(fā)及臨床試驗已有百余項,而在中國,CAR-T細(xì)胞在卵巢癌、惡性膠質(zhì)瘤、肝癌和乳腺癌等研究中也取得了一定的突破(表1)。
2.2 CAR-T細(xì)胞腫瘤治療中的局限性
臨床應(yīng)用中CAR-T治療高潛力、快發(fā)展的背后同樣存在制約,例如不能特異性識別腫瘤抗原、過度激活大量釋放炎癥因子等現(xiàn)象。不能特異性識別腫瘤抗原即脫靶效應(yīng)(與抗原受體特異性和抗原分子擬態(tài)相關(guān)),主要原因是缺少腫瘤的特異性抗原(tumor specific antigen,TSA)[20-21],這就很可能發(fā)生腫瘤逃逸,或造成對正常細(xì)胞的殺傷。實體瘤中,若CAR-T細(xì)胞不能準(zhǔn)確歸巢到正確的腫瘤位置,由于脫靶效應(yīng),反而會和靶點表達(dá)較少的正常細(xì)胞結(jié)合,殺傷正常細(xì)胞,造成致命危害。因此,可以說脫靶效應(yīng)是CAR-T細(xì)胞治療面臨的重大挑戰(zhàn)。目前,研究者在努力尋找腫瘤新型表位的同時[22],也在積極探索能消除或沉默CAR-T細(xì)胞來控制脫靶效應(yīng)的新方法,如引入“自殺基因”等[23]。
CAR-T細(xì)胞與有效抗原結(jié)合后,能激活相關(guān)細(xì)胞因子的產(chǎn)生和釋放。其活化T細(xì)胞產(chǎn)物,釋放IFN-γ、IL-2和其他多種細(xì)胞因子,使得它們轉(zhuǎn)而激活其他免疫細(xì)胞,包括巨噬細(xì)胞等,從而形成免疫細(xì)胞間的聯(lián)合殺傷。若CAR-T細(xì)胞過度激活或在疾病根除后仍在患者體內(nèi)大量存活,就會導(dǎo)致細(xì)胞因子過度釋放,這是十分危險的[24]。目前,臨床上通過抗細(xì)胞因子療法控制其危害??辜?xì)胞因子療法能直接靶向次級介體,反應(yīng)迅速,無毒,不影響T細(xì)胞移植[25]。然而,抗細(xì)胞因子療法的前提是必須明確過度激活的細(xì)胞因子,才能針對性消除,這使得新型CAR-T細(xì)胞產(chǎn)品用于人體時,很難分辨這一新型產(chǎn)品的臨床毒性。
CARs基礎(chǔ)結(jié)構(gòu)的構(gòu)建與分子生物學(xué)的基因工程免疫受體有關(guān)。簡單來說,就是將一個來自某一抗體的輕鏈和重鏈序列的多肽結(jié)構(gòu),連接于受體上的信號傳導(dǎo)裝置,如經(jīng)典的T細(xì)胞受體——ζ鏈,通過對其三大區(qū)域(配體結(jié)合域、跨膜結(jié)構(gòu)域與胞內(nèi)信號域)基本設(shè)計上的不斷改進(jìn),從而提高其效能。
3.1 構(gòu)建CARs信號域
CARs傳輸一個信號到細(xì)胞內(nèi)的T細(xì)胞需要通過一個信號域,經(jīng)典的信號域為CD3ζ鏈,也有人使用來自Fcγ受體的信號域[14]。第二、三代CARs在初代CARs信號域CD3的基礎(chǔ)上引入新的信號域,如CD28、4-1BB或OX40,以期產(chǎn)生共同刺激,使TCR更易被抗原遞呈細(xì)胞(APC)識別,完全激活T細(xì)胞生理活性[26-27]。事實證明,引入共刺激信號分子這種方式能增加細(xì)胞因子的產(chǎn)量及促進(jìn)CAR-T細(xì)胞體外增殖,第二代CD19特異性的CARs能攜帶CD28或4-1BB信號集團,且在臨床前模型和B細(xì)胞惡性腫瘤的臨床試驗中已經(jīng)證明其具有體內(nèi)抗腫瘤活性[19,25-35]。
目前常用的共刺激分子包括CD27、CD28、CD134(OX40)、CD137(4-1BB)、CD244和ICOS等,將其合并在一起能增加ζ鏈信號,從而增強T細(xì)胞的增殖和存活[36-41]。CD28是第一個用于構(gòu)造CARs的共刺激分子,顯著增加了IL-2的產(chǎn)物和細(xì)胞毒性[9,42],加入共刺激分子CD28后,通過調(diào)節(jié)T細(xì)胞能增加CARs對細(xì)胞毒性的抗性[43]。臨床前實驗發(fā)現(xiàn)與CD28共刺激CARs相比,CD137的引入更能改善CARs的體內(nèi)存活,增強其抗腫瘤活性[36]。而ICOS信號域過表達(dá)能使人T細(xì)胞轉(zhuǎn)化為Th17表型,并且在某些情況下與CD28共刺激的Th17細(xì)胞會產(chǎn)生更高一級的抗腫瘤功效[43-45]。已清楚地發(fā)現(xiàn)引入單一共刺激分子能賦予CART細(xì)胞更高的存活或其他一些T細(xì)胞功能,而較單一共刺激分子相比,增加更多的共刺激分子的確增加了CAR-T細(xì)胞的特性[11,46]。
3.2 CARs跨膜域構(gòu)建
表1 CAR-T技術(shù)在中國的臨床試驗
配體結(jié)合域通過鉸鏈區(qū)與跨膜域相連,通常來源于CD8和IgG4分子。鉸鏈對于細(xì)胞表面的CARs表達(dá)非常重要。鉸鏈區(qū)影響scFv的韌性,從而影響其與配基的相互作用。而鉸鏈區(qū)的長度會影響T細(xì)胞和標(biāo)靶間的相互作用(取決于目標(biāo)抗原標(biāo)位的位置),例如,CD22靠近細(xì)胞膜表位較遠(yuǎn)端表位相比會引起更溶胞作用[47]。同樣,越長的連接區(qū)域越能增強CARs識別NCAM和5T4等膜近端表位的能力,還有可能增加其適應(yīng)性[48-49]。這一發(fā)現(xiàn)很可能揭示了CARs修飾的細(xì)胞和靶向細(xì)胞最佳的距離。
3.3 CARs結(jié)合域構(gòu)建
研究表明,CAR-T細(xì)胞能識別和清除相當(dāng)于自身100倍的靶向抗原[50],如果提高CAR-T細(xì)胞的特異性,會降低其激活的條件,擴大其治療窗[51]。所以說,作為與靶標(biāo)結(jié)合的至關(guān)重要的特異性區(qū)域,CARs配體結(jié)合域的構(gòu)建尤為重要。其構(gòu)建的關(guān)鍵在于如何能只特異性地與靶向細(xì)胞結(jié)合而不影響其他組織器官。目前大多數(shù)已確定的腫瘤特異性抗原是那些在早期胚胎發(fā)育期正常表達(dá),而在惡性腫瘤中被異常表達(dá)的自身抗原,例如NY-ESO1和MAGE家族抗原。
近期發(fā)表在《Cell》的一項研究中,提出提高CAR-T細(xì)胞靶向特異性和準(zhǔn)確性的一種有效方法是改造它們,使其識別組合抗原,而Kole等利用基因工程技術(shù)改造了一種需要組合激活的T細(xì)胞回路(T cell circuit)。在這一回路中,改造后的T細(xì)胞需要識別2個抗原,一種合成Notch受體能識別其中一個抗原,被激活后能夠誘導(dǎo)對應(yīng)第二個抗原的CAR表達(dá),只有在腫瘤細(xì)胞同時表達(dá)2種抗原的情況下,改造后的T細(xì)胞才會被激活,研究者將這種T細(xì)胞命名為dual-receptor AND-gate T cells。體內(nèi)實驗中,這些T細(xì)胞展現(xiàn)出精準(zhǔn)的識別與治療,能夠區(qū)分出只擁有單個抗原的非目標(biāo)腫瘤,且有效清除攜帶組合抗原的腫瘤[52]。
不同的臨床情況需要不同的scFv、鉸鏈、跨膜區(qū)、刺激和共刺激域。事實上,迄今仍沒有一個公認(rèn)最佳的方式來構(gòu)建CARs。關(guān)于CARs的最佳設(shè)計仍然需要更多的調(diào)查和對不同惡性腫瘤的臨床評估。我們也許可以運用一些簡單的克隆技術(shù)替換或改進(jìn)其不同組件,以便于評估和完善CARs模塊化設(shè)計。雖然CARs最優(yōu)設(shè)計仍未得到根本解決,但可喜的是,關(guān)于CARs設(shè)計的新思路卻層出不窮,以其為基礎(chǔ)的新型靶向免疫治療技術(shù)日新月異,如T細(xì)胞受體工程T細(xì)胞(T cell receptor-engineered T cells,TCR-T)、雙特異性T細(xì)胞銜接器(bispecific T-cell engagers,BiTE)和嵌合抗原受體NK細(xì)胞(chimeric antigen recep?tor NK cells,CAR-NK)等。這意味著CARs領(lǐng)域的發(fā)展將比經(jīng)典的新型分子藥物更為迅捷,是目前腫瘤免疫治療中炙手可熱的新貴。
癌癥免疫療法是繼細(xì)胞毒藥物化療和靶向藥物治療之后新興的癌癥治療策略。作為免疫療法的重要代表,嵌合抗原受體T細(xì)胞免疫療法在臨床白血病的治療中實現(xiàn)了治愈[34]。目前,該治療策略還存在一些問題,包括CAR-T細(xì)胞識別癌細(xì)胞時可能發(fā)生脫靶效應(yīng),以及病人在CAR-T治療后會面臨“細(xì)胞因子風(fēng)暴(細(xì)胞因子釋放綜合征)”的風(fēng)險。解決這些問題可能要依賴于基因工程手段,或配合使用其他類型的治療藥物。這些手段將有助于增加CAR-T療法的安全性、有效性,以及對更多種類癌癥治療的適用性,從而挽救更多患者的生命。
[1]Gross G,Waks T,Eshhar Z.Expression of immuno?globulin-T-cell receptor chimeric molecules as func?tional receptors with antibody-type specificity[J].Proc Natl Acad Sci USA,1989,86(24):10024-10028.
[2]Goverman J,Gomez S M,Segesman K D,et al.Chi?meric immunoglobulin-T cell receptor proteins form functional receptors:implications for T cell receptor complex formation and activation[J].Cell,1990,60(6): 929-939.
[3]Romeo C,Seed B.Cellular immunity to HIV activated by CD4 fused to T cell or Fc receptor polypeptides [J].Cell,1991,64(5):1037-1046.
[4]Irving B A,Weiss A.The cytoplasmic domain of the T cell receptor 6 chain is sufficient to couple to sig?nal transduction pathways[J].Cell,1991,64(5):891-901. [5]Cheadle E J,Gornall H,Baldan V,et al.CAR-T cells:driving the road from the laboratory to the clinic [J].Immunol Rev,2014,257(1):91-106.
[6]Jensen M C,Popplewell L,Cooper L J,et al.Anti?transgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specif?ic chimeric antigen receptor redirected T cells in hu?mans[J].Biol Blood Marrow Transplant,2010,16(9): 1245-1256.
[7]Pule M A,Savoldo B,Myers G D,et al.Virus-specif?ic T cells engineered to coexpress tumor-specific re?ceptors:persistence and antitumor activity in individu?als with neuroblastoma[J].Nat Med,2008,14(11):1264-12670.
[8]Brentjens R J,Latouche J B,Santos E,et al.Eradica?tion of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and in?terleukin-15[J].Nat Med,2003,9(3):279-286.
[9]Maher J,Brentjens R J,Gunset G,et al.Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta/CD28 receptor[J].Nat Biotech?nol,2002,20(1):70-75.
[10]Zhao Y,Wang Q J,Yang S,et al.A herceptin-based chimeric antigen receptor with modified signaling do?mains leads to enhanced survival of transduced T lym?phocytes and antitumor activity[J].Immunology,2009, 183(9):5563-5574.
[11]Carpenito C,Milone M C,Hassan R,et al.Control of large,established tumor xenografts with genetically re?targeted human T cells containing CD28 and CD137 domains[J].Proc Natl Acad Sci USA,2009,106(9): 3360-3365.
[12]Pegram H J,Lee J C,Hayman E G,et al.Tumor-tar?geted T cells modified to secrete IL-12 eradicate sys?temic tumors without need for prior conditioning[J]. Blood,2012,119(18):4133-4141.
[13]Lamers C H J,Sleijfer S,Vulto A G,et al.Treat?ment of metastatic renal cell carcinoma with autolo?gous T-lymphocytes genetically retargeted against car?bonic anhydrase IX:first clinical experience[J].J Cli?ni Oncol,2006,24(13):20-22.
[14]Kershaw M H,Westwood J A,Parker L L,et al.A phase I study on adoptive immunotherapy using genemodified T cells for ovarian cancer[J].Clin Cancer Res,2006,12(20 Pt1):6106-6115.
[15]Porter D L,Levine B L,Kalos M,et al.Chimeric an?tigen receptor-modified T cells in chronic lymphoid leukemia[J].N Engl J Med,2011,365(8):725-733.
[16]Monjezi R,Miskey C,Gogishvili T,et al.Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors[J].Leuke?mia,2016,doi:10.1038/leu.2016.180.
[17]Till B G,Jensen M C,Wang J,et al.Adoptive immu?notherapy forindolent non-Hodgkin lymphoma and man?tlecell lymphoma using genetically modifiedautologous CD20-specific T cells[J].Blood,2008,112(6):2261-2271.
[18]Long A H,Highfill S L,Cui Y,et al.Reduction of MDSCs with all-trans retinoic acid improves CAR therapy efficacy for sarcomas[J].Cancer Immunol Res, 2016,4(10):869-880.
[19]Turtle C J,Hanafi L A,Berger C,et al.Immunothera?py of non-Hodgkin's lymphoma with a defined ratio of CD8+and CD4+CD19-specific chimeric antigen receptor-modified T cells[J].Sci Transl Med,2016,8 (355):355ra116.
[20]Lamers C H,Sleijfer S,van Steenbergen S,et al. Treatment of metastatic renal cell CARscinoma with CAIX CARS-engineered T-cells:clinical evaluation and management of on-target toxicity[J].Mol Ther, 2013,21(4):904-912.
[21]Morgan R A,Yang J C,Kitano M,et al.Case report of a serious adverseevent following the administration of T-cells transduced with a chimeric antigen recep?tor recognizing ERBB2[J].Mol Ther,2010,18(4):843-851.
[22]Marcela V.Designing CAR T cells for glioblastoma[J]. Oncoimmunology,2015,4(12):e1048956.
[23]Philip B,Kokalaki E,Mekkaoui L,et al.A highly compact epitope-based marker/suicide gene for easier and safer T-cell therapy[J].Blood,2014,124(8):1277-1287.
[24]Lee D W,Gardner R,Porter D L,et al.Current con?cepts in the diagnosis and management of cytokine re?lease syndrome[J].Blood,2014,124(2):188-195.
[25]Davila M L,Riviere I,Wang X,et al.Efficacy and toxicity management of 19-28z CARS T-cell therapy in B cell acute lymphoblastic leukemia[J].Sci Transl Med,2014,6(224):268-276.
[26]Kochenderfer J N,W ilson W H,Janik J E,et al. Eradication of B-lineage cells and regression of lym?phoma in a patient treated with autologous T cells ge?netically engineered to recognize CD19[J].Blood,2010, 116(20):4099-4102.
[27]Porter D L,Levine B L,Kalos M,et al.Chimeric an?tigen receptor-modified T cells in chronic lymphoid leukemia[J].N Engl J Med,2011,365(8):725-733.
[28]Long A H,Haso W M,Shern J F,et al.4-1BB co?stimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors[J].Nat Med,2015,21(6):581-590.
[29]Li W,Guo L,Rathi P,et al.Redirecting T cells to glypican-3 with 4-1BB zeta chimeric antigen recep?tors results in Th1 polarization and potent antitumor activity[J].Hum Gene Ther,2016,doi:10.1089/hum. 2016.025.
[30]Savoldo B,Ramos C A,Liu E,et al.CD28 costimula?tion improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients [J].J Clin Invest,2011,121(5):1822-1826.
[31]Brentjens R J,Davila M L,Riviere I, et al.CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lympho?blastic leukem ia[J].Sci Transl Med,2013,5(177):638-643.
[32]Chen R,Song X T,Chen B.CD19 chimeric antigen receptor T cell therapy for the treatment of B cell lin?eage acute lymphoblastic leukemia.[J].Discovery Med, 2015,20(110):185-190.
[33]Chen F,Fan C M,Gu X Z,et al.Construction of an?ti-CD20 single-chain antibody-CD28-CD137-TCRzeta recombinant genetic modified T cells and its treat?ment effect on B cell lymphoma[J].Med Sci Monit, 2015,21:2110-2115.
[34]Maude S L,Frey N,Shaw P A,et al.Chimeric anti?gen receptor T cells for sustained remissions in leuke?mia[J].N Engl J Med,2014,371(16):1507-1517.
[35]Lee D W,Kochenderfer J N,Stetler-Stevenson M,et al.T cells expressing CD19 chimeric antigen recep?tors for acute lymphoblastic leukaemia in children and young adults:a Phase 1 dose-escalation trial[J]. Lancet,2015,385(9967):517-528.
[36]Milone M C,Fish J D,Carpenito C,et al.Chimeric receptors containing CD137 signal transduction do? mains mediate enhanced survival of T cells and in?creased antileukemic efficacy in vivo[J].Mol Ther, 2009,17(8):1453-1464.
[37]Sanchez-Paulete A R,Labiano S,Rodriguez-Ruiz M E,et al.Deciphering cd137(4-1BB)signaling in T-cell costimulation for translation into successful can?cer immunotherapy[J].Eur J Immunol,2016,46(3):513-522.
[38]Hombach A A,Heiders J,Foppe M,et al.OX40 co?stimulation by a chimeric antigen receptor abrogates CD28 and IL-2 induced IL-10 secretion by redirect?ed CD4+T cells[J].Oncoimmunology,2012,1(4):458-466.
[39]Altvater B,Landmeier S,Pscherer S,et al.2B4 (CD244)signaling via chimeric receptors costimulates tumor-antigen specific proliferation and in vitro expan?sion of human T cells[J].Cancer Immunol Immunoth?er,2009,58(12):1991-2001.
[40]Song D G,Ye Q,Poussin M,et al.CD27 costimula?tion augments the survival and antitumor activity of re?directed human T cells in vivo[J].Blood,2012,119(3): 696-706.
[41]Guedan S,Chen X,Madar A,et al.ICOS-based chi?meric antigen receptors program bipolar TH17/TH1 cells[J].Blood,2014,124(7):1070-1080.
[42]Mittal P,St Rose M C,Wang X,et al.Tumor-unrelat?ed CD4 T cell help augments CD134 plus CD137 du?al costimulation tumor therapy[J].J Immunol,2015,195 (12):5816-5826.
[43]Loskog A,Giandomenico V,Rossig C,et al.Addition of the CD28 signaling domain to chimeric T-cell re?ceptors enhances chimeric T-cell resistance to T regu?latory cells[J].Leukemia,2006,20(10):1819-1828.
[44]Paulos C M,Carpenito C,Plesa G,et al.The induc?ible costimulatory(ICOS)is critical for the develop?ment of human T(H)17 cells[J].Sci Transl Med,2010, 2(55):7444-7451.
[45]Savoldo B,Ramos C A,Liu E,et al.Brief report CD28 costimulation improves expansion and persis?tence of chimeric antigen receptor-modified T cells in lymphoma patients[J].J Clin Invest,2011,121(5):1822-1826.
[46]Zhong X-S,Matsushita M,Plotkin J et al.Chimeric antigen receptors combining 4-1BB and CD28 signal?ing domains augment PI3kinase/AKT/Bcl-XL activa?tion and CD8&plus,T cell-mediated tumor eradica?tion[J].Mol Ther,2010,18(2):413-420.
[47]James S E,Greenberg P D,Jensen M C,et al.Anti?gen sensitivity of CD22-specific chimeric TCR is mod?ulated by target epitope distance from the cell mem?brane[J].J Immunol,2008,180(10):7028-7038.
[48]Guest R D,Hawkins R E,Kirillova N,et al.The role of extracellular spacer regions in the optimal de?sign of chimeric immune receptors:evaluation of four different scFvs and antigens[J].J Immunother,2005,28 (3):203-211.
[49]Haso W,Lee D W,Shah N N,et al.Anti-CD22-chi?meric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia[J].Blood,2013,121(7): 1165-1174.
[50]Watanabe K,Terakura S,Martens A C,et al.Target antigen density governs the efficacy of anti-CD20-CD28-CD3ζ chimeric antigen receptor-modified effec?tor CD8+T-cells[J].J Immunol,2015,194(3):911-920.
[51]Song D G,Ye Q,Poussin M,et al.A fully human chimeric antigen receptor with potent activity against cancer cells but reduced risk for offtumor toxicity[J]. Oncotarget,2015,6(25):21533-21546.
[52]Roybal K T,Rupp L J,Morsut L,et al.Precision tu?mor recognition by T cells with combinatorial antigensensing circuits[J].Cell,2016,164(4):770-779.
Progress of Engineering Chimeric Antigen Receptor in Tu?mor Therapy
QIAN Yu-Zhen1,2,ZHANG Ming-Rui1,2,REN Gang3,ZOU Wei1,2*
1.College of Life Science,Liaoning Normal University,Dalian 116081;2.Liaoning Key Laboratories of Biotechnol?ogy and Molecular Drug Research and Development,Dalian 116081;3.Institute of Medicinal Biotechnology,Chi?nese Academy of Medical Sciences,Beijing 100050;China
How to treat cancer has always been one of the biggest problems that afflict humanity for years be?cause conventional treatment has its limitations.As the development of gene transfer and cell culture technology, cellular immunotherapy gradually come to public attention.Chimeric antigen receptor T-cell(CAR-T)immunothera?py is a new adoptive tumor immunotherapy approach that can target antigen specificity recognition and kill tumor cells.Therefore,this review summarizes the application of CAR-T in oncotherapy and the strategy of building CAR-T cells.
chimeric antigen receptors;T lymphocyte;oncotherapy
R392.1
A
1009-0002(2017)02-0188-08
10.3969/j.issn.1009-0002.2017.02.025
2016-09-19
錢昱臻(1990-),男,碩士研究生
鄒偉,(E-mail)weizou60@126.com
*Corresponding anthor,E-mail:weizou60@126.com