欧美综合在线观看一区二区-日韩免费av区二区电影-中文字幕免费在线一区二区-胜肽和玻色因哪个抗老效果好

加入收藏 | 設(shè)為首頁 |      中文 | English

咨詢熱線: 028-85121781

當(dāng)前位置:首頁 > 動態(tài)中心 > 企業(yè)動態(tài) 腫瘤分化誘導(dǎo)劑綠原酸抑制PD-L1表達,增強PD-1抗體的免疫,發(fā)揮抗腫瘤作用
腫瘤分化誘導(dǎo)劑綠原酸抑制PD-L1表達,增強PD-1抗體的免疫,發(fā)揮抗腫瘤作用
[ 來源:Int. J. Biol. Sci. 2024 Jan 1;20(1):61-77.   發(fā)布日期:2024-07-09 13:22:14  責(zé)任編輯:  瀏覽次 ]

1. 中國醫(yī)學(xué)科學(xué)院、北京協(xié)和醫(yī)學(xué)院,醫(yī)藥生物技術(shù)研究所,北京,100050

2. 中國醫(yī)學(xué)科學(xué)院、北京協(xié)和醫(yī)學(xué)院,藥物研究所、天然藥物生物活性物質(zhì)與功能國家重點實驗室,北京,100050

3. 中國科學(xué)院昆明植物研究所,植物化學(xué)與西部植物資源持續(xù)利用國家重點實驗室,云南昆明,650201

4. 九章生物科技發(fā)展有限公司,四川成都,610041

5. 首都醫(yī)科大學(xué)附屬北京天壇醫(yī)院腫瘤中心,神經(jīng)腫瘤科,北京,100070

摘要: 由于免疫檢查點抑制劑已顯示出良好的臨床療效,免疫檢查點阻斷已經(jīng)成為癌癥治療的重要策略。然而,大約只有12.5%的患者受益于免疫療法。在此,我們提出了一種癌癥分化誘導(dǎo)劑綠原酸(chlorogenic acid, CGA,目前正處于中國治療膠質(zhì)瘤的II期臨床試驗中),它是一種小分子免疫檢查點抑制劑,可以增強PD-1抗體的免疫,發(fā)揮抗腫瘤作用。CGA通過抑制p-STAT1-IRF1通路和增強活化T細胞的活性,可以抑制干擾素誘導(dǎo)的PD-L1在腫瘤細胞中的表達。在CGA與抗PD -1抗體聯(lián)合治療小鼠腫瘤異種移植中,可降低腫瘤細胞凋亡,抑制PD-L1IRF1的表達,增強了抗PD-1抗體對細胞的抑制作用,抑制腫瘤的生長。特別是,CGA可增強腫瘤浸潤T細胞的活性。改善腫瘤浸潤免疫細胞顆粒酶基因表達。總之,通過誘導(dǎo)分化,CGA抑制了PD-L1在癌癥細胞中的表達,有效促進腫瘤中浸潤的T細胞,增強抗腫瘤作用。因此,CGA如果與抗PD -1抗體聯(lián)合使用,可能是一種很有前景的增強抗癌作用的藥物免疫療法。

 

關(guān)鍵詞:綠原酸,腫瘤分化誘導(dǎo)劑,PD-L1,抗PD -1抗體,聯(lián)合治療

1簡介

免疫檢查點是一種避免自身免疫反應(yīng),保持免疫系統(tǒng)處于穩(wěn)態(tài)的重要機制。然而,激活免疫檢查點也是腫瘤患者帶瘤生存主要的途徑之一。檢查點通路的啟動導(dǎo)致細胞毒性T淋巴細胞的衰竭,隨后使腫瘤細胞繞過免疫監(jiān)視。PD-1/PD-L1軸是眾所周知的免疫途徑檢查點其中之一。PD-L1已在黑色素瘤,肺癌,乳腺癌、卵巢癌、胰腺癌和結(jié)腸癌等進行研究。作為一個免疫檢查點分子,PD-L1的功能是與其受體PD-1相互作用,PD-1在腫瘤浸潤性T淋巴細胞表面表達,引起細胞毒性T細胞活化的抑制,繞過對癌癥的免疫監(jiān)控。在過去的幾十年里,抗體對抗免疫檢查點分子已成為一個研究熱點,試圖發(fā)現(xiàn)癌癥藥物的免疫療法。

第一個免疫檢查點抑制劑(ICI)是伊匹單抗,是一種細胞毒 T 淋巴細胞相關(guān)抗原4 (CTLA-4),臨床有效治療轉(zhuǎn)移性黑色素瘤。隨后,PD-1抗體(派姆單抗和納武利尤單抗)及其配體PD-L1 (阿替利珠單抗,德瓦魯單抗和阿維魯單抗)已經(jīng)被批準(zhǔn),并被廣泛用于治療不同類型的癌癥。此外,新型ICIs的臨床試驗也在進行,世界各地的醫(yī)院都在對癌癥進行檢測。然而,盡管取得了巨大的成功,預(yù)期抗癌免疫反應(yīng)僅見于12.5%的部分患者。觀察患者的免疫反應(yīng),抗癌的區(qū)別歸因于各種原因,如PD-L1PD-1的表達水平,基因突變細胞,以及新抗原的發(fā)展,這是對癌癥免疫療法的巨大挑戰(zhàn)。

為避免患者使用ICIs無效,生物標(biāo)志物的研究是目前的方法之一。聯(lián)合ICI抗體與常規(guī)抗癌藥物是促進治療效果的又一嘗試。如聯(lián)合抗pd -1抗體和吉西他濱通過激活巨噬細胞和CD8+ T細胞,增強了抗體的抗癌作用。盡管PD-1單藥治療抑制劑或它們的聯(lián)合療法臨床取得顯著效果,對PD-1抑制劑的應(yīng)答率低仍然是一個大問題。因此,基于機制的理性設(shè)計對于優(yōu)化ICIs治療是非常可取的。

誘導(dǎo)腫瘤分化是我們的新課題,將癌細胞從高侵入性和轉(zhuǎn)移表型轉(zhuǎn)變?yōu)閻盒猿潭容^低或接近正常狀態(tài)。根據(jù)生物學(xué)原理,PD-L1在癌細胞中的表達與腫瘤惡性相關(guān),應(yīng)該是陽性的,就像成功創(chuàng)造了一種免疫逃逸機制。最近研究表明出現(xiàn)了PD-L1表達與癌細胞或/和干細胞分化不良有關(guān)。孫等人指出指出,間充質(zhì)干細胞(MSCs)可通過STAT3/mTORc-Myc通路 誘導(dǎo)PD-L1在胃癌細胞中的表達。同時也有人證明了這一點,證明了這一點,在非小細胞肺癌中,PD-L1介導(dǎo)的免疫逃逸與c-MycEGFR/MAPK信號有關(guān)。也有人證明了c-Myc可以誘導(dǎo)PD-L1在食管鱗狀細胞癌中的表達,而其他研究顯示PD-L1可能會增強肺腺癌中c-Myc的活性。這些發(fā)現(xiàn)提出了癌癥治療中的腫瘤分化與誘導(dǎo)的免疫檢查點。因此,我們假設(shè)一種改善的分化狀態(tài)使腫瘤細胞中PD-L1的產(chǎn)生減少,從而增強腫瘤細胞毒性t細胞的活性微環(huán)境。如果成立,癌癥分化誘導(dǎo)劑除了它們自身的抗癌作用,還可促進ICIs的抗癌活性。

綠原酸(CGA),是一種多酚化合物,可作為消炎藥使用,據(jù)報道,綠原酸具有多種藥理活性,如抗氧化、抗炎、保護神經(jīng)。最近的研究表明,CGA能抑制各種癌癥細胞的增殖。九章生物的I期臨床試驗(肌內(nèi)注射)顯示CGA耐受性很好,在復(fù)發(fā)性高級別膠質(zhì)瘤患者中,獲益性良好。基于此研究結(jié)果,中國FDA已于2017年批準(zhǔn)CGA進行復(fù)發(fā)性高級別膠質(zhì)瘤患者的II期臨床試驗(NCT2013 L01855)。對于它的作用機理,以前的研究工作已經(jīng)證明CGA通過誘導(dǎo)癌細胞分化,抑制癌和肺癌的腫瘤生長。與低分化相關(guān)的基因的表達,如c-Myc,主要由CGA,通過上調(diào)SUMO1的表達和c-Myc;,導(dǎo)致癌細胞中c-Myc的強烈抑制和向成熟表型的轉(zhuǎn)化。同時,我們也發(fā)現(xiàn)了CGA在劑量和時間依賴性的食管鱗狀細胞癌中,能夠下調(diào)腫瘤相關(guān)的干細胞標(biāo)記物BMI1SOX2的表達。在本研究中, CGA誘導(dǎo)腫瘤分化,可以下調(diào)腫瘤PD-L1表達,激活腫瘤組織中的細胞毒性T淋巴細胞,并可與目前可用的ICI抗體聯(lián)合使用。由于CGA對人體是安全的,所以此研究可能很快就會轉(zhuǎn)化為臨床使用。

2 結(jié)論

用抗體阻斷PD-1/PD-L1相互作用的免疫療法在癌癥治療應(yīng)用已經(jīng)取得了很大的成功。然而,免疫反應(yīng)在患者的情況各不相同,大多數(shù)患者未能從中受益。PD-L1在腫瘤組織中的表達被認為是一種機制,甚至是至關(guān)重要的生物標(biāo)志物,能夠預(yù)測腫瘤患者的抗癌免疫反應(yīng)。PD-L1表達下調(diào)可能會增強抗癌作用,發(fā)揮免疫治療。在本研究中,我們發(fā)現(xiàn)腫瘤分化誘導(dǎo)劑CGA可以降低癌細胞中PD-L1的表達,從而保護腫瘤浸潤的T細胞免受PD-1/PD-L1的攻擊,導(dǎo)致T細胞死亡等,增強PD -1抗體的治療效果。因此,CGA作為免疫檢查點抑制劑,聯(lián)合抗PD -1抗體可能是一種提高療效的新方法。

癌細胞分化代表了程序化細胞內(nèi)協(xié)同過程,將癌細胞從惡性轉(zhuǎn)移到良性或正常細胞,以減少增殖和轉(zhuǎn)移為特征,發(fā)揮免疫檢查點功能。這個生物過程是復(fù)雜的,并且在觸發(fā)程序信號之后,癌細胞的通路和基因表達基本都受到了監(jiān)管。由CGA重新編程,減少PD-L1表達可能是一個重要的癌細胞分化的跡象和途徑,至少涉及IFN-γ/JAK/pSTAT1/IRF1/PD-L1

IFN-γ,由細胞毒性T細胞和天然殺傷細胞分泌,是先天或適應(yīng)性免疫反應(yīng)重要分子。就像一把雙刃劍,IFN-γ不僅參與細胞毒性,同時還能誘導(dǎo)細胞中PD-L1的表達,最終導(dǎo)致腫瘤免疫逃避。在本研究中,我們發(fā)現(xiàn)CGA在黑色素瘤癌細胞,三陰性乳腺癌細胞和卵巢癌細胞中,抑制IFN-γ誘導(dǎo)的人PD-L1表達黑色素瘤癌細胞,三陰性乳腺癌細胞和卵巢癌細胞。mRNA影響蛋白表達的穩(wěn)定性,我們首先檢測PD-L1 mRNA的穩(wěn)定性,發(fā)現(xiàn)它沒有被CGA處理破壞。然后,檢測PD-L1啟動子(表達PD-L1的另一個重要因素)的活性。我們發(fā)現(xiàn)PD-L1啟動子的活性被CGA抑制。JAK-pSTAT1-IRF1通路是與IFN-γ誘導(dǎo)的,以及通路上游的IFN-γ結(jié)合IFN-γ受體(IFNGR1IFNGR2),

然后激活JAK-pSTAT1-IRF1通路。作為一種轉(zhuǎn)錄因子,IRF1與啟動子序列,從而激活PD-L1基因表達。我們首先測試了表達的IFNGR1IFNGR2,之后沒有發(fā)現(xiàn)CGA的變化。然而,過度表達轉(zhuǎn)錄因子IRF1消除了CGAPD-L1啟動子活性這種抑制作用;

表明IRF1CGA活性的主要機制。先前的研究表明STAT1是在IFN-γIRF1激活之前被磷酸化誘導(dǎo)的PD-L1刺激通路。本研究中,我們發(fā)現(xiàn)CGA下調(diào)了STAT1的磷酸化,與以前的報告一致。因此,CGA可能通過抑制STAT1磷酸化- IRF1 - PD-L1通路,抑制PD-L1在腫瘤細胞中的表達。此外,據(jù)報道,IRF1可以結(jié)合STAT1DNA并促使STAT1的磷酸化, CGA下調(diào)IRF1可以進一步減弱STAT1磷酸化和增強對PD-L1的抑制作用。同時,p-STAT1是一種直接結(jié)合PD-L1啟動子的轉(zhuǎn)錄因子; CGA的磷酸化從而抑制STAT1,抑制PD-L1的表達。事實上, CGA的增加,在腫瘤/免疫細胞共培養(yǎng)系統(tǒng),使A375MDA-MB-231SK-OV-3的易感性細胞向活化的Jurkat E6細胞轉(zhuǎn)移。這些結(jié)果表明CGA對腫瘤細胞PD-L1表達的抑制作用可以增加T細胞的活性,使阻斷腫瘤免疫檢查點功能。

簡而言之,通過聯(lián)合治療PD-L1IRF1的表達在腫瘤組織中顯著減少,以及藥物相互作用阻斷了PD-1PD-L1之間的聯(lián)系,因此T細胞介導(dǎo)的抗腫瘤免疫療法得到了推廣。在體外實驗,延長CGA處理時間,增強腫瘤細胞中PD-L1表達的抑制作用,表明在細胞分化的進展抑制作用增強了。

腫瘤對免疫療法的反應(yīng)從高到無。因此,它們被分為“Hot”、“ Cold”“Altered”,該定義與腫瘤組織浸潤的T細胞數(shù)量有關(guān)。高T細胞浸潤的癌癥在腫瘤微環(huán)境中被認為是“Hot”,低滲區(qū)為“Cold”區(qū),腫瘤浸潤邊緣有T細胞浸潤和/或顯示低CD8+免疫抑制亞型屬于“Altered”組。本研究選取兩種異種移植物模型小鼠MC38作為“Hot”腫瘤模型,4T1“Altered”模型,根據(jù)抗pd -1抗體及浸潤水平CD8+ T細胞的腫瘤免疫反應(yīng)(圖1、2)。檢測到CGA與抗pd -1抗體結(jié)合;結(jié)果表明CGA聯(lián)合抗pd -1抗體治療效果較好,效果優(yōu)于單一療法,無論是“Hot” 還是“Altered” 異種移植模型,均腫瘤生長產(chǎn)生額外抑制作用 (治療效果31.1%, MC38升高,p < 0.05;治療效果24.5%,4T1升高,p < 0.01)。

 


1   CGA在體內(nèi)增強了抗pd -1抗體的抗腫瘤作用。

a e.CGA提高了小鼠結(jié)腸癌抗PD-1抗體的抗腫瘤作用。在C57BL/6N雄性小鼠背部皮下注射2 × 105個小鼠結(jié)腸癌MC38細胞。接種3天后,隨機分組分為生理鹽水聯(lián)合IgG:NS + IgG組,生理鹽水聯(lián)合抗PD-1抗體組:NS +pd -1抗體組,CGA聯(lián)合IgG:CGA + IgG組,CGA聯(lián)合抗PD-1抗體:CGA +pd -1) (n = 7)。CGA (50 mg / kg)每天1(i.p),連續(xù)3周分別于第3、710天給予抗體200 ug。A.荷瘤小鼠的代表圖像。腫瘤用紅色圈出。B.代表切除腫瘤的圖像。C.腫瘤生長示意圖。D.各組小鼠腫瘤重量比較。E.體重的變化。F-J. CGA提高了抗PD-1抗體對小鼠乳腺癌的抗腫瘤作用。將1 × 105個小鼠乳腺癌細胞4T1注射到BALB / c雌性小鼠乳腺脂肪中。接種3天后,將小鼠隨機分為上述4組。CGA (50 mg / kg)每日1(ig),連用19例連續(xù)第3天,第7天,第10天分別給予200 ug抗體。F.荷瘤小鼠代表像。腫瘤被圈起來紅色的。G.切除腫瘤的代表圖像。H.腫瘤生長示意圖。1 .各組小鼠腫瘤重量比較。所示數(shù)據(jù)為平均值±SD。經(jīng)單因素方差分析,與NS + IgG組比較,*p< 0.05 **p< 0.01, **p< 0.001。NS表示生理鹽水;免疫球蛋白。

 

2 CGA可促進腫瘤組織中細胞毒性T淋巴細胞的浸潤。

從新鮮腫瘤組織中分離細胞毒性T淋巴細胞(CTLs)解離成單細胞懸液,流式細胞術(shù)分析。Aa. 浸潤細胞毒性T細胞(CD8+ CD3+)的代表性流式細胞儀圖譜。Ab. 浸潤的細胞毒性T細胞百分比。Ba.浸潤效應(yīng)T細胞(IFN-γ+ CD8+ CD3+)的代表性流式細胞儀圖譜。Bb. 滲透效應(yīng)T細胞百分比。C.腫瘤組織CD8(粉色)IFN-γ(紅色)染色的代表性多色免疫熒光圖像。10μm。Da.代表性流式細胞儀4T1荷瘤小鼠腫瘤組織中浸潤Treg細胞(Foxp3+ CD25+ CD4+)的譜圖。Db.浸潤Treg細胞百分比。數(shù)據(jù)以mean±SD表示(n = 5)。與NS + IgG比較,*p< 0.05, **p< 0.01 **p< 0.001。組采用單因素方差分析檢驗。

這種聯(lián)合用藥對腫瘤生長的抑制作用MC38(“Hot”)4T1(“Altered”)更顯著。對這種差異的解釋可能是相關(guān)的T細胞定位于腫瘤組織,其中浸潤T細胞的數(shù)量、細胞類型和活性會影響小鼠的抗腫瘤免疫微環(huán)境?偟膩碚f,在Hot模型中,T細胞數(shù)量和活性較高。因此,在AlteredCold模型中,需產(chǎn)生強烈的免疫反應(yīng)來對抗癌癥。在本研究中,浸潤T兩種腫瘤組織中CD3+ CD8+的表達 (2A-B)可能有助于獨特的腫瘤抑制。

細胞毒性T淋巴細胞(CTLs) 在抗腫瘤免疫是主要的免疫細胞。IFN-γ+ CD8+ T在腫瘤細胞毒性中發(fā)揮重要作用,因此這個T細胞亞群的比例是抗腫瘤免疫陽性指標(biāo)。在研究CGACTLs的影響,我們發(fā)現(xiàn)CD8+ CD3+ T細胞群(CTLs的子集),在聯(lián)合組和CGA或抗pd -1抗體單藥治療組中均增強。隨后我們分析IFN-γ+腫瘤組織在CD8+ T細胞中的浸潤比例。數(shù)據(jù)顯示,在兩種小鼠腫瘤模型中,聯(lián)合用藥組IFN-γ+ CD8+ T細胞比例增高;MC38荷瘤小鼠中IFN-γ+ CD8+ T細胞的增加更為明顯明顯優(yōu)于單藥治療(2B;2.77倍相對于NS + Anti-PD-L1上調(diào)組,p < 0.001),相同的結(jié)果見圖1A-D。這些結(jié)果表明CGA可以增加腫瘤微環(huán)境中的細胞毒性T細胞。同時,調(diào)節(jié)性T細胞(Treg),在抗腫瘤免疫治療中的一種抑制因子,與癌癥預(yù)后不良相關(guān)?pd -1單藥治療抗體增加腫瘤組織中Treg細胞的數(shù)量,然而,這種不希望的增長在CGA4T1腫瘤模型聯(lián)合治療中被逆轉(zhuǎn)了 (2D)?贵w單藥治療中Treg細胞的增加,有可能使PD-1表達的Treg細胞受到抗pd -1的保護,抗體通過阻斷PD-1之間的相互作用抑制腫瘤組織中PD-L1的表達;但CGA聯(lián)合給藥組對Treg細胞的影響需要更多的研究。實驗中發(fā)現(xiàn),CGA并沒有改變?nèi)?/span>T細胞免疫動物應(yīng)答,但可能有效調(diào)節(jié)巨噬細胞細胞因子。

顆粒酶是CTLs和殺傷細胞分泌的一組蛋白酶,在穿孔素的幫助下執(zhí)行細胞殺傷作用。顆粒酶B (Granzyme B, GZMB)是蛋白質(zhì)的主要分子之一,據(jù)報道,顆粒酶家族在PD-1封閉療法治療后,與臨床結(jié)果呈正相關(guān)。在研究中發(fā)現(xiàn),與抗PD -1單藥治療相比聯(lián)合治療中瘤顆粒酶基因上調(diào), (3)在兩種模型腫瘤蛋白組織水平上進一步得到驗證。我們認為這有益于腫瘤微環(huán)境增強T細胞激活和/T細胞計數(shù)。

 


3. CGA上調(diào)腫瘤組織中顆粒酶B (GZMB)的表達。

PD -1抗體單藥治療組(NS+pd -1, n =3)收集4T1荷瘤小鼠的CGA+PD-1抗體聯(lián)合治療組(CGA+pd -1, n = 3)。腫瘤組織總RNA采用“l(fā)imma”“DESeq”R包進行RNA序列分析。差異表達基因(DEGs)富集氧化石墨烯的功能。A. DEGs火山圖,log2FC|≥1,p≤0.05。B. GO生物過程功能項富集前20名。C. NS+Anti-PD-1CGA+Anti-PD-1組細胞溶解功能基因表達熱圖?潭缺硎緦(shù)變換后的TPMs + 1,紅到藍表示相對基因表達由高到低。實驗組中,NS+Anti-PD-1-1、NS+Anti-PD-1-2、NS+Anti-PD-1-3、CGA+Anti-PD-1-1CGA+Anti-PD-1-2、CGA+Anti-PD-1-3為測序樣品。D~ECGA+PD-1治療可上調(diào)腫瘤組織中GZMB蛋白的表達。腫瘤組織中CD8(粉色)GZMB(綠色)染色的彩色免疫熒光圖像(D)10μm。Western blot檢測GZMB蛋白表達分析(E). GZMB結(jié)果歸一化為β-肌動蛋白密度比。數(shù)據(jù)以mean±SD表示(n = 6),差異有統(tǒng)計學(xué)意義:*p< 0.05, **p< 0.01,與NS+Anti-PD-1組進行學(xué)生t檢驗。GZMB表示顆粒酶B

總之,CGA是一種已知的癌癥分化誘導(dǎo)劑(CDI),在中國,CGA治療神經(jīng)膠質(zhì)瘤的臨床試驗已進入II期臨床后期階段。CGA通過抑制IFN-γ誘導(dǎo)的干擾素-γ/ JAK / pSTAT1 / IRF1 / PD-L1通路抑制腫瘤細胞中的PD-L1。因此,CGA是一種安全的免疫增強劑,增強腫瘤中T細胞的活性微環(huán)境,與抗pd -1抗體聯(lián)合使用時可提高抗癌效果(4)。

4  CGA通過抑制STAT1磷酸化和抑制IFN-γ誘導(dǎo)的PD-L1表達,發(fā)揮腫瘤免疫治療作用

致謝

   該項目得到了CAMS醫(yī)學(xué)科學(xué)創(chuàng)新基金(No.2022-I2M-2-002, 2022-I2M-JB-012) 的支持;中國科學(xué)院植物化學(xué)與植物重點實驗室與中國西部資源國家基金(No. P2022-KF01)。

參考文獻

1. Mittendorf EA, Philips AV, Meric-Bernstam F, Qiao N, Wu Y, Harrington S, etal. PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res.2014; 2: 361-70.

2. Han J, Xu X, Liu Z, Li Z, Wu Y, Zuo D. Recent advances of molecular mechanisms of regulating PD-L1 expression in melanoma. Int Immunopharmacol. 2020; 88: 106971.

3. Wang X, Teng F, Kong L, Yu J. PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther. 2016; 9: 5023-39.

4. Bagchi S, Yuan R, Engleman EG. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu Rev Pathol. 2021; 16: 223-49.

5. Haslam A, Prasad V. Estimation of the Percentage of US Patients With Cancer Who Are Eligible for and Respond to Checkpoint Inhibitor Immunotherapy Drugs. JAMA Netw Open. 2019; 2: e192535.

6. Ott PA, Bang YJ, Piha-Paul SA, Razak ARA, Bennouna J, Soria JC, et al. T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. J Clin Oncol. 2019; 37: 318-27.

7. Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy Mechanisms response biomarkers and combinations. Sci Transl Med. 2016; 8: 328rv4.

8. Ancevski Hunter K, Socinski MA, Villaruz LC. PD-L1 Testing in Guiding Patient Selection for PD-1/PD-L1 Inhibitor Therapy in Lung Cancer. Molecular diagnosis & therapy. 2018; 22: 1-10.

9. Lantuejoul S, Sound-Tsao M, Cooper WA, Girard N, Hirsch FR, Roden AC, et al. PD-L1 Testing for Lung Cancer in 2019: Perspective From the IASLC Pathology Committee. J Thorac Oncol. 2020; 15: 499-519.

10. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 2019; 18: 197-218.

11. Ho TTB, Nasti A, Seki A, Komura T, Inui H, Kozaka T, et al. Combination of gemcitabine and anti-PD-1 antibody enhances the anticancer effect of M1 macrophages and the Th1 response in a murine model of pancreatic cancer liver metastasis. J Immunother Cancer. 2020; 8: e001367.

12. Wang H, He X, Fang D, Wang X, Guan J, Shi ZW, et al. Gemcitabine-facilitated modulation of the tumor microenvironment and PD-1/PD-L1 blockade generate a synergistic antitumor effect in a murine hepatocellular carcinoma model. Clin Res Hepatol Gastroenterol. 2022; 46: 101853.

13. Du B, Wen X, Wang Y, Lin M, Lai J. Gemcitabine and checkpoint blockade exhibit synergistic anti-tumor effects in a model of murine lung carcinoma. Int Immunopharmacol. 2020; 86: 106694.

14. Xu L, Zou C, Zhang S, Chu TSM, Zhang Y, Chen W, et al. Reshaping the systemic tumor immune environment (STIE) and tumor immune microenvironment (TIME) to enhance immunotherapy efficacy in solid tumors. J Hematol Oncol. 2022; 15: 87.

15. Qu S, Jiao Z, Lu G, Yao B, Wang T, Rong W, et al. PD-L1 lncRNA splice isoform promotes lung adenocarcinoma progression via enhancing c-Myc activity. Genome Biol. 2021; 22: 104-28.

16. Wei F, Zhang T, Deng SC, Wei JC, Yang P, Wang Q, et al. PD-L1 promotes colorectal cancer stem cell expansion by activating HMGA1-dependent signaling pathways. Cancer Lett. 2019; 450: 1-13.

17. Almozyan S, Colak D, Mansour F, Alaiya A, Al-Harazi O, Qattan A, et al. PD-L1 promotes OCT4 and Nanog expression in breast cancer stem cells by sustaining PI3K/AKT pathway activation. Int J Cancer. 2017; 141: 1402-12.

18. Mansour FA, Al-Mazrou A, Al-Mohanna F, Al-Alwan M, Ghebeh H. PD-L1 is overexpressed on breast cancer stem cells through notch3/mTOR axis. Oncoimmunology. 2020; 9: 1729299.

19. Sun L, Wang Q, Chen B, Zhao Y, Shen B, Wang H, et al. Gastric cancer mesenchymal stem cells derived IL-8 induces PD-L1 expression in gastric cancer cells via STAT3/mTOR-c-Myc signal axis. Cell Death Dis. 2018; 9: 928-39.

20. Wang J, Jia Y, Zhao S, Zhang X, Wang X, Han X, et al. BIN1 reverses PD-L1-mediated immune escape by inactivating the c-MYC and EGFR/MAPK signaling pathways in non-small cell lung cancer. Oncogene. 2017; 36: 6235-43.

21. Liang MQ, Yu FQ, Chen C. C-Myc regulates PD-L1 expression in esophageal squamous cell carcinoma. American journal of translational research. 2020; 12: 379-88.

22. Naveed M, Hejazi V, Abbas M, Kamboh AA, Khan GJ, Shumzaid M, et al. Chlorogenic acid (CGA): A pharmacological review and call for further research. Biomed Pharmacother. 2018; 97: 67-74.

23. Hou N, Liu N, Han J, Yan Y, Li J. Chlorogenic acid induces reactive oxygen species generation and inhibits the viability of human colon cancer cells. Anticancer Drugs. 2017; 28: 59-65.

24. Yan Y, Liu N, Hou N, Dong L, Li J. Chlorogenic acid inhibits hepatocellular carcinoma in vitro and in vivo. J Nutr Biochem. 2017; 46: 68-73.

25. Yamagata K, Izawa Y, Onodera D, Tagami M. Chlorogenic acid regulates apoptosis and stem cell marker-related gene expression in A549 human lung cancer cells. Mol Cell Biochem. 2018; 441: 9-19.

26. Xue N, Zhou Q, Ji M, Jin J, Lai F, Chen J, et al. Chlorogenic acid inhibits glioblastoma growth through repolarizating macrophage from M2 to M1 phenotype. Sci Rep. 2017; 7: 39011.

27. Kang Z, Li S, Kang X, Deng J, Yang H, Chen F, et al. Phase I study of chlorogenic acid injection for recurrent high-grade glioma with long-term follow-up. Cancer Biol Med. 2023; 20: 465-76.

28. Huang S, Wang LL, Xue NN, Li C, Guo HH, Ren TK, et al. Chlorogenic acid effectively treats cancers through induction of cancer cell differentiation. Theranostics. 2019; 9: 6745-63.

29. Zhan Y, Li R, Feng C, Li X, Huang S, Wang L, et al. Chlorogenic acid inhibits esophageal squamous cell carcinoma growth in vitro and in vivo by downregulating the expression of BMI1 and SOX2. Biomed Pharmacother. 2020; 121: 109602.

30. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017; 19: 1189-201.

31. Nishikawa H, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol. 2014; 27: 1-7.

32. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015; 43: e47.

33. Dennis G, Jr., Sherman Bt Fau - Hosack DA, Hosack Da Fau - Yang J, Yang J Fau - Gao W, Gao W Fau - Lane HC, Lane Hc Fau - Lempicki RA, et al. DAVID: Database for Annotation, Visualization, and Integrated Discovery.

34. Cullen SP, Brunet M, Martin SJ. Granzymes in cancer and immunity. Cell Death Differ. 2010; 17: 616-23.

35. Ewen CL, Kane KP, Bleackley RC. A quarter century of granzymes. Cell Death Differ. 2012; 19: 28-35.

36. Trapani JA, Smyth MJ. Functional significance of the perforin/granzyme cell death pathway. Nat Rev Immunol. 2002; 2: 735-47.

37. Cerezo M, Guemiri R, Druillennec S, Girault I, Malka-Mahieu H, Shen S, et al. Translational control of tumor immune escape via the eIF4F-STAT1-PD-L1 axis in melanoma. Nat Med. 2018; 24: 1877-86.

38. Wang J, Zhang R, Lin Z, Zhang S, Chen Y, Tang J, et al. CDK7 inhibitor THZ1 enhances antiPD-1 therapy efficacy via the p38α/MYC/PD-L1 signaling in non-small cell lung cancer. J Hematol Oncol. 2020; 13: 99.

39. Alspach E, Lussier DM, Schreiber RD. Interferon gamma and Its Important Roles in Promoting and Inhibiting Spontaneous and Therapeutic Cancer Immunity. Cold Spring Harb Perspect Biol. 2019; 11: a028480.

40. Andersson R, Sandelin A. Determinants of enhancer and promoter activities of regulatory elements. Nat Rev Genet. 2020; 21: 71-87.

41. Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, et al. MYC regulates the antitumor immune response through CD47 and PD-L1. SCIENCE. 2016; 352: 227-31.

42. Zenke K, Muroi M, Tanamoto KI. IRF1 supports DNA binding of STAT1 by promoting its phosphorylation. Immunol Cell Biol. 2018; 96: 1095-103.

43. Nakayama Y, Mimura K, Tamaki T, Shiraishi K, Kua LF, Koh V, et al. PhosphoSTAT1 expression as a potential biomarker for antiPD1/antiPDL1 immunotherapy for breast cancer. Int J Oncol. 2019; 54: 2030-8.

44. Xu L, Zhang Y, Tian K, Chen X, Zhang R, Mu X, et al. Apigenin suppresses PD-L1 expression in melanoma and host dendritic cells to elicit synergistic therapeutic effects. J Exp Clin Cancer Res. 2018; 37: 261-76.

45. Maleki Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer. 2018; 6: 157.

46. Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S, et al. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front Immunol. 2019; 10: 168.

47. Grasselly C, Denis M, Bourguignon A, Talhi N, Mathe D, Tourette A, et al. The Antitumor Activity of Combinations of Cytotoxic Chemotherapy and Immune Checkpoint Inhibitors Is Model-Dependent. Front Immunol. 2018; 9: 2100-13.

48. Thommen DS, Schumacher TN. T Cell Dysfunction in Cancer. Cancer Cell. 2018; 33: 547-62.

49. Hossain MA, Liu G, Dai B, Si Y, Yang Q, Wazir J, et al. Reinvigorating exhausted CD8(+) cytotoxic T lymphocytes in the tumor microenvironment and current strategies in cancer immunotherapy. Med Res Rev. 2021; 41: 156-201.

50. Liu YT, Sun ZJ. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 2021; 11: 5365-86.

51. St Paul M, Ohashi PS. The Roles of CD8(+) T Cell Subsets in Antitumor Immunity. Trends Cell Biol. 2020; 30: 695-704.

52. Kamada T, Togashi Y, Tay C, Ha D, Sasaki A, Nakamura Y, et al. PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci U S A. 2019; 116: 9999-10008.

53. Zhang Y, Yang Y, Ye J, Gao Y, Liao H, Zhou J, et al. Construction of chlorogenic acid-containing liposomes with prolonged antitumor immunity based on T cell regulation. Sci China Life Sci. 2021; 64: 1097-115.

54. Ye J, Gao Y, Ji M, Yang Y, Wang Z, Wang B, et al. Oral SMEDDS promotes lymphatic transport and mesenteric lymph nodes target of chlorogenic acid for effective T-cell antitumor immunity. J Immunother Cancer. 2021; 9:

e002753.

55. Shin HS, Satsu H, Bae MJ, Zhao Z, Ogiwara H, Totsuka M, et al. Anti-inflammatory effect of chlorogenic acid on the IL-8 production in Caco-2 cells and the dextran sulphate sodium-induced colitis symptoms in C57BL/6 mice. Food Chem. 2015; 168: 167-75.

56. Yuan Y, Gong X, Zhang L, Jiang R, Yang J, Wang B, et al. Chlorogenic acid ameliorated concanavalin A-induced hepatitis by suppression of Toll-like receptor 4 signaling in mice. Int Immunopharmacol. 2017; 44: 97-104.

57. Shan J, Fu J, Zhao Z, Kong X, Huang H, Luo L, et al. Chlorogenic acid inhibits lipopolysaccharide-induced cyclooxygenase-2 expression in RAW264.7 cells through suppressing NF-kappaB and JNK/AP-1 activation. Int Immunopharmacol. 2009; 9: 1042-8.

58. Zheng Z, Sheng Y, Lu B, Ji L. The therapeutic detoxification of chlorogenic acid against acetaminophen-induced liver injury by ameliorating hepatic inflammation. Chem Biol Interact. 2015; 238: 93-101.

59. Xu Y, Chen J, Yu X, Tao W, Jiang F, Yin Z, et al. Protective effects of chlorogenic acid on acute hepatotoxicity induced by lipopolysaccharide in mice. Inflamm Res. 2010; 59: 871-7.

30. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017; 19: 1189-201.

31. Nishikawa H, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol. 2014; 27: 1-7.

32. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015; 43: e47.

33. Dennis G, Jr., Sherman Bt Fau - Hosack DA, Hosack Da Fau - Yang J, Yang J Fau - Gao W, Gao W Fau - Lane HC, Lane Hc Fau - Lempicki RA, et al. DAVID: Database for Annotation, Visualization, and Integrated Discovery.

34. Cullen SP, Brunet M, Martin SJ. Granzymes in cancer and immunity. Cell Death Differ. 2010; 17: 616-23.

35. Ewen CL, Kane KP, Bleackley RC. A quarter century of granzymes. Cell Death Differ. 2012; 19: 28-35.

36. Trapani JA, Smyth MJ. Functional significance of the perforin/granzyme cell death pathway. Nat Rev Immunol. 2002; 2: 735-47.

37. Cerezo M, Guemiri R, Druillennec S, Girault I, Malka-Mahieu H, Shen S, et al. Translational control of tumor immune escape via the eIF4F-STAT1-PD-L1 axis in melanoma. Nat Med. 2018; 24: 1877-86.

38. Wang J, Zhang R, Lin Z, Zhang S, Chen Y, Tang J, et al. CDK7 inhibitor THZ1 enhances antiPD-1 therapy efficacy via the p38α/MYC/PD-L1 signaling in non-small cell lung cancer. J Hematol Oncol. 2020; 13: 99.

39. Alspach E, Lussier DM, Schreiber RD. Interferon gamma and Its Important Roles in Promoting and Inhibiting Spontaneous and Therapeutic Cancer Immunity. Cold Spring Harb Perspect Biol. 2019; 11: a028480.

40. Andersson R, Sandelin A. Determinants of enhancer and promoter activities of regulatory elements. Nat Rev Genet. 2020; 21: 71-87.

41. Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, et al. MYC regulates the antitumor immune response through CD47 and PD-L1. SCIENCE. 2016; 352: 227-31.

42. Zenke K, Muroi M, Tanamoto KI. IRF1 supports DNA binding of STAT1 by promoting its phosphorylation. Immunol Cell Biol. 2018; 96: 1095-103.

43. Nakayama Y, Mimura K, Tamaki T, Shiraishi K, Kua LF, Koh V, et al. PhosphoSTAT1 expression as a potential biomarker for antiPD1/antiPDL1 immunotherapy for breast cancer. Int J Oncol. 2019; 54: 2030-8.

44. Xu L, Zhang Y, Tian K, Chen X, Zhang R, Mu X, et al. Apigenin suppresses PD-L1 expression in melanoma and host dendritic cells to elicit synergistic therapeutic effects. J Exp Clin Cancer Res. 2018; 37: 261-76.

45. Maleki Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer. 2018; 6: 157.

46. Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S, et al. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front Immunol. 2019; 10: 168.

47. Grasselly C, Denis M, Bourguignon A, Talhi N, Mathe D, Tourette A, et al. The Antitumor Activity of Combinations of Cytotoxic Chemotherapy and Immune Checkpoint Inhibitors Is Model-Dependent. Front Immunol. 2018; 9: 2100-13.

48. Thommen DS, Schumacher TN. T Cell Dysfunction in Cancer. Cancer Cell. 2018; 33: 547-62.

49. Hossain MA, Liu G, Dai B, Si Y, Yang Q, Wazir J, et al. Reinvigorating exhausted CD8(+) cytotoxic T lymphocytes in the tumor microenvironment and current strategies in cancer immunotherapy. Med Res Rev. 2021; 41: 156-201.

50. Liu YT, Sun ZJ. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 2021; 11: 5365-86.

51. St Paul M, Ohashi PS. The Roles of CD8(+) T Cell Subsets in Antitumor Immunity. Trends Cell Biol. 2020; 30: 695-704.

52. Kamada T, Togashi Y, Tay C, Ha D, Sasaki A, Nakamura Y, et al. PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci U S A. 2019; 116: 9999-10008.

53. Zhang Y, Yang Y, Ye J, Gao Y, Liao H, Zhou J, et al. Construction of chlorogenic acid-containing liposomes with prolonged antitumor immunity based on T cell regulation. Sci China Life Sci. 2021; 64: 1097-115.

54. Ye J, Gao Y, Ji M, Yang Y, Wang Z, Wang B, et al. Oral SMEDDS promotes lymphatic transport and mesenteric lymph nodes target of chlorogenic acid for effective T-cell antitumor immunity. J Immunother Cancer. 2021; 9: e002753.

55. Shin HS, Satsu H, Bae MJ, Zhao Z, Ogiwara H, Totsuka M, et al. Anti-inflammatory effect of chlorogenic acid on the IL-8 production in Caco-2 cells and the dextran sulphate sodium-induced colitis symptoms in C57BL/6 mice. Food Chem. 2015; 168: 167-75.

56. Yuan Y, Gong X, Zhang L, Jiang R, Yang J, Wang B, et al. Chlorogenic acid ameliorated concanavalin A-induced hepatitis by suppression of Toll-like receptor 4 signaling in mice. Int Immunopharmacol. 2017; 44: 97-104.

57. Shan J, Fu J, Zhao Z, Kong X, Huang H, Luo L, et al. Chlorogenic acid inhibits lipopolysaccharide-induced cyclooxygenase-2 expression in RAW264.7 cells through suppressing NF-kappaB and JNK/AP-1 activation. Int Immunopharmacol. 2009; 9: 1042-8.

58. Zheng Z, Sheng Y, Lu B, Ji L. The therapeutic detoxification of chlorogenic acid against acetaminophen-induced liver injury by ameliorating hepatic inflammation. Chem Biol Interact. 2015; 238: 93-101.

59. Xu Y, Chen J, Yu X, Tao W, Jiang F, Yin Z, et al. Protective effects of chlorogenic acid on acute hepatotoxicity induced by lipopolysaccharide in mice. Inflamm Res. 2010; 59: 871-7.


上一篇:中國醫(yī)學(xué)科學(xué)院胡海宇/陳曉光揭示綠原酸抗癌靶點及相關(guān)機制
下一篇:注射用綠原酸用于復(fù)發(fā)高級別膠質(zhì)瘤的Ⅰ期臨床研究及長期隨訪
【相關(guān)資訊】
·泰國密斯卡灣健康集團與九章生物科技宣布新合作,攜手將下一代癌癥與抗衰老療法 引入泰國 2024-09-14
·藥物所陳曉光課題組和胡海宇課題組在綠原酸靶標(biāo)發(fā)現(xiàn)和抗腫瘤作用機制研究取得新進展 2024-07-11
·中國醫(yī)學(xué)科學(xué)院胡海宇/陳曉光揭示綠原酸抗癌靶點及相關(guān)機制 2024-07-09
·腫瘤分化誘導(dǎo)劑綠原酸抑制PD-L1表達,增強PD-1抗體的免疫,發(fā)揮抗腫瘤作用 2024-07-09
·注射用綠原酸用于復(fù)發(fā)高級別膠質(zhì)瘤的Ⅰ期臨床研究及長期隨訪 2023-10-28
·綠原酸通過ACAT1-TPK1-PDH途徑誘導(dǎo)神經(jīng)母細胞瘤細胞分化 2023-08-22
·綠原酸通過調(diào)節(jié)人體真皮成纖維細胞膠原的代謝和凋亡以預(yù)防UVA誘導(dǎo)下的皮膚老化 2022-08-02
·我公司參加第六屆中國創(chuàng)投大會并發(fā)布Ⅱ期臨床試驗結(jié)果 2021-09-29
时尚| 合作市| 涟水县| 宜州市| 长治市| 高要市| 海安县| 奉贤区| 双辽市| 呼图壁县| 望都县| 博乐市| 兴业县| 乐陵市| 明水县| 兴宁市| 宣化县| 宣城市| 秭归县| 屏山县| 偏关县| 惠安县| 宁明县| 日土县| 宜兰县| 大化| 抚松县| 赫章县| 垫江县| 图木舒克市| 余姚市| 陈巴尔虎旗| 南川市| 鹤壁市| 即墨市| 婺源县| 阿拉善右旗| 宿松县| 房产| 庄浪县| 芦溪县|