• 
    

    
    

      99热精品在线国产_美女午夜性视频免费_国产精品国产高清国产av_av欧美777_自拍偷自拍亚洲精品老妇_亚洲熟女精品中文字幕_www日本黄色视频网_国产精品野战在线观看

      ?

      rmhTNF-α聯(lián)合PD-1單抗治療小鼠Lewis肺癌的研究

      2020-05-25 02:34朱遼遼張存徐盈李曉菊高源韓俊張英起
      中國醫(yī)藥導報 2020年10期
      關鍵詞:藥組生理鹽水生存期

      朱遼遼 張存 徐盈 李曉菊 高源 韓俊 張英起

      [摘要] 目的 研究重組改構人腫瘤壞死因子-α(rmhTNF-α)聯(lián)合PD-1單抗(anti-PD-1)對小鼠Lewis肺癌的治療效果及作用機制。 方法 24只6~8周齡C57BL/6雌鼠,依據隨機數字表法分為4組,每組6只:生理鹽水組、rmhTNF-α組、anti-PD-1單抗組、聯(lián)合給藥組。游標卡尺測瘤體并用活體成像觀測。16 d后安樂處死小鼠,剝離腫瘤稱重。觀察小鼠生存期。免疫組化檢測腫瘤CD8+T細胞數目。 結果 與生理鹽水組比較,rmhTNF-α組、anti-PD-1組、聯(lián)合給藥組腫瘤體積、瘤重均減小,差異有高度統(tǒng)計學意義(P < 0.01);rmhTNF-α組腫瘤體積大于聯(lián)合給藥組,差異有統(tǒng)計學意義(P < 0.05);而聯(lián)合給藥組與anti-PD-1組腫瘤體積差異無統(tǒng)計學意義(P > 0.05);聯(lián)合給藥組瘤重分別與rmhTNF-α、anti-PD-1組比較,差異均無統(tǒng)計學意義(均P > 0.05)。與生理鹽水組比較,rmhTNF-α組、anti-PD-1組、聯(lián)合給藥組生存期增加,差異有統(tǒng)計學意義(P < 0.05或P < 0.01),而聯(lián)合給藥組生存期分別與rmhTNF-α組、anti-PD-1組比較,差異均無統(tǒng)計學意義(均P > 0.05)。rmhTNF-α組與生理鹽水組比較,腫瘤CD8+T細胞數目增多(P < 0.05);anti-PD-1組、聯(lián)合給藥組與生理鹽水組比較,腫瘤CD8+T細胞數目顯著增加(P < 0.01);聯(lián)合給藥組腫瘤CD8+T細胞數目少于rmhTNF-α組(P < 0.01),而與anti-PD-1組比較,腫瘤CD8+T細胞數目差異無統(tǒng)計學意義(P > 0.05)。 結論 rmhTNF-α具有抑制小鼠肺癌的作用,但不具有增強PD-1抗體治療肺癌的作用。

      [關鍵詞] 重組改構人腫瘤壞死因子-α;PD-1單抗;CD8+T細胞;Lewis肺癌

      [中圖分類號] R730.2 ? ? ? ? ?[文獻標識碼] A ? ? ? ? ?[文章編號] 1673-7210(2020)04(a)-0004-04

      Study of rmhTNF-α combined with anti-PD-1 in the treatment of Lewis lung cancer in mice

      ZHU Liaoliao ? ZHANG Cun ? XU Ying ? LI Xiaoju ? GAO Yuan ? HAN Jun ? ZHANG Yingqi

      Biopharmaceutical Teaching and Research Office, Department of Pharmacy, Air Force Medical University, Shaanxi Province, Xi′an ? 710032, China

      [Abstract] Objective To study the therapeutic effect and mechanism of recombinant mutated human tumor necrosis factor-α (rmhTNF-α) combined with PD-1 monoclonal antibody (anti-PD-1) in Lewis lung cancer in mice. Methods A total of 24 female C57BL/6 mice aged 6 to 8 weeks were divided into 4 groups according to the random number table method, with 6 mice in each group: normal saline group, rmhTNF-group, anti-PD-1 group, and combined administration group. The tumor was measured with vernier caliper and observed by in vivo imaging. After 16 days, the mice were euthanized and the tumor was removed and weighed. The survival period of mice were observed. Immunohistochemical was used to detect tumor CD8+T cells. Results Compared with the normal saline group, the tumor volume and tumor weight in the rmhTNF-α group, anti-PD-1 group and the combined administration group all decreased, with highly statistically significant differences (P < 0.01). The tumor volume of rmhTNF-α group was larger than that of the combined administration group, and the difference was statistically significant (P < 0.05). However, there was no significant difference in tumor volume between the combined administration group and the anti-PD-1 group (P > 0.05). There was no statistically significant difference in tumor weight between the combined administration group and the rmhTNF-α group and anti-PD-1 group (all P > 0.05). Compared with the normal saline group, the survival of rmhTNF-α group, anti-PD-1 group and the combined administration group increased, and the differences were statistically significant (P < 0.05 or P < 0.01), while the survival of the combined administration group was not statistically significant compared with the rmhTNF-α group and anti-PD-1 group (all P > 0.05). Compared with the normal saline group, the number of CD8+T cells increased in the rmhTNF-α group (P < 0.05). Compared with the normal saline group, the number of CD8+T cells increased significantly in the anti-PD-1 group and combined administration group (P < 0.01). The number of CD8+T cells in the combined administration group was less than that in the rmhTNF-α group (P < ?0.01), while the number of CD8+T cells was not statistically significant compared with that in the anti-PD-1 group (P > 0.05). Conclusion RmhTNF-α can inhibit lung cancer in mice, but it can not enhance the effect of anti-PD-1 to treat lung cancer.

      [Key words] Recombinant mutated human tumor necrosis factor-α; PD-1 monoclonal antibody; CD8+T cell; Lewis lung cancer

      腫瘤壞死因子-α(TNF-α)由巨噬細胞產生,介導內毒素誘導的腫瘤壞死[1],在免疫中發(fā)揮關鍵作用[2]。TNF-α可以改變血管通透性,使抗腫瘤藥物更易到達腫瘤組織周圍[3]。免疫檢查點抑制劑PD-1單抗(anti-PD-1)治療晚期肺癌有效率為20%,與細胞因子的聯(lián)合治療成為新策略[4-5]。因此,本研究利用優(yōu)化的重組改構人腫瘤壞死因子-α(recombinant mutated human tumor necrosis factor-α,rmhTNF-α)聯(lián)合anti-PD-1治療小鼠Lewis肺癌,探索rmhTNF-α是否具有提高anti-PD-1治療效果的作用及作用機制。

      1 材料與方法

      1.1 實驗材料

      rmhTNF-α(上海賽達生物藥業(yè)有限公司,批號:SC20180401);anti-PD-1(西安壯志生物科技有限公司,批號:BE0146);Luciferase慢病毒(上海吉凱基因化學技術有限公司,批號:LPK001);Lewis肺癌細胞(中國科學院上海細胞庫,目錄號:TCM7);小鼠CD8分子免疫組化試劑盒(北京中杉金橋有限公司,批號:SP9000)。

      1.2 實驗動物

      24只C57BL/6雌鼠,6~8周齡,體重(18±2)g,購自空軍軍醫(yī)大學動物中心,生產許可證號:SCXK(軍)2017-0021。飼養(yǎng)和實驗遵循實驗動物護理及使用指南[6]。

      1.3 分組與給藥

      luciferase慢病毒轉染Lewis肺癌細胞,皮下接種2×106。由隨機數字表法將小鼠分為4組,每組6只:生理鹽水組、rmhTNF-α組、anti-PD-1組、聯(lián)合給藥組。rmhTNF-α依據前期實驗的有效劑量[7]。rmhTNF-α肌肉注射10 000 U/只,anti-PD-1腹腔注射125 μg/只,3 d/次,共4次。抑瘤率(%)=(1-給藥組平均瘤重/對照組平均瘤重)×100%。腫瘤體積(mm3)=長徑×短徑2/2。

      1.4 免疫組化

      參考侯志超等[8]步驟:標本石蠟包埋切片后脫蠟水化,抗原修復,室溫冷卻,H2O2封閉,一抗孵育過夜,磷酸鹽緩沖溶液(PBS)沖洗,孵育二抗,PBS沖洗,滴加鏈霉卵白素,PBS沖洗,DAB顯色,蘇木精復染,分化,脫水,透明,中性樹脂封片。應用Image-Pro Plus 6.0軟件處理數據。

      1.5 統(tǒng)計學方法

      用SPSS 25.0軟件進行婁據分析,計量資料以均數±標準差(x±s)表示,兩組比較采用LSD-t檢驗,四組比較采用One-way ANOVA分析,生存期用log-rank分析,以P < 0.05為差異有統(tǒng)計學意義。

      2 結果

      2.1 四組小鼠腫瘤體積比較

      四組腫瘤體積比較,差異有高度統(tǒng)計學意義(F = 13.824,P < 0.01)。與生理鹽水組比較,rmhTNF-α組、anti-PD-1組、聯(lián)合給藥組腫瘤體積減小,差異有高度統(tǒng)計學意義(P < 0.01);rmhTNF-α組腫瘤體積大于聯(lián)合給藥組,差異有統(tǒng)計學意義(P < 0.05);而聯(lián)合給藥組與anti-PD-1組腫瘤體積差異無統(tǒng)計學意義(P > 0.05)。四組活體成像平均熒光值比較,差異有高度統(tǒng)計學意義(F = 12.548,P < 0.01)。rmhTNF-α組平均熒光值低于生理鹽水組(P < 0.01),anti-PD-1組、聯(lián)合給藥組平均熒光值顯著低于生理鹽水組(P < 0.01),聯(lián)合給藥組平均熒光值與rmhTNF-α組、anti-PD-1組比較,差異無統(tǒng)計學意義(P > 0.05)。見圖1。

      2.2 四組小鼠肺癌抑瘤率比較

      四組瘤重比較,差異有高度統(tǒng)計學意義(F = 9.903,P < 0.01)。與生理鹽水組比較,rmhTNF-α組、anti-PD-1組及聯(lián)合給藥組瘤重均顯著減輕(P < 0.01),且anti-PD-1組有1只小鼠腫瘤消失。聯(lián)合給藥組瘤重分別與rmhTNF-α組、anti-PD-1組比較,差異均無統(tǒng)計學意義(均P > 0.05)。見圖2。抑瘤率比較見表1。

      2.3 四組小鼠生存期比較

      與生理鹽水組比較,rmhTNF-α組生存期增加,差異有統(tǒng)計學意義(P < 0.05),anti-PD-1組、聯(lián)合給藥組生存期也增加,差異有高度統(tǒng)計學意義(P < 0.01),而聯(lián)合給藥組生存期分別與rmhTNF-α組、anti-PD-1組比較,差異均無統(tǒng)計學意義(均P > 0.05)。見圖3。

      與生理鹽水組比較,*P < 0.05,**P < 0.01

      圖3 ? 四組小鼠生存曲線(n = 6)

      2.4 四組小鼠腫瘤CD8+T細胞數目比較

      四組小鼠腫瘤CD8+T細胞數目比較,差異有高度統(tǒng)計學意義(F = 25.587,P < 0.01)。rmhTNF-α組與生理鹽水組比較,腫瘤CD8+T細胞數目增加(P < 0.05);anti-PD-1組、聯(lián)合給藥組與生理鹽水組比較,腫瘤CD8+T細胞數目顯著增加(P < 0.01);聯(lián)合給藥組腫瘤CD8+T細胞數目少于rmhTNF-α組(P < 0.01),而與anti-PD-1組比較,腫瘤CD8+T細胞數目差異無統(tǒng)計學意義(P > 0.05)。見圖4。

      3 討論

      TNF-α調節(jié)免疫系統(tǒng)[9],rmhTNF-α有高活性和低毒性[10]。臨床結果顯示,癌癥患者對rmhTNF-α聯(lián)合化療的響應率高于單獨化療(P < 0.05)[11-12]。Siurala等[13]研究結果顯示mTNF-α顯著增強過繼性T細胞的抗瘤效應。Elia等[14]發(fā)現低劑量TNF能增強免疫檢查點阻斷劑聯(lián)合過繼細胞的治療效果。近年來,免疫檢查點阻斷劑的研究成為熱點。Gettinger等[15]研究顯示納武單抗治療晚期肺癌,患者5年總生存率僅為16%。Wrangle等[16]的實驗發(fā)現納武單抗能阻止腫瘤細胞免疫逃逸。Asano等[17]報道低劑量白細胞介素-2聯(lián)合PD-1阻斷劑具有抗腫瘤免疫的協(xié)同作用。

      本研究中rmhTNF-α不具有促進anti-PD-1抑制肺癌的作用。據文獻報道[18-19],TNF可增強腫瘤細胞PD-L1表達,誘發(fā)免疫抑制,這可能抵消了TNF-α對免疫系統(tǒng)的激活作用,導致與anti-PD-1無協(xié)同抑瘤作用。Perez等[20]也發(fā)現,TNF抑制劑聯(lián)合CTLA-4和PD-1抗體可提高抗腫瘤效果。

      綜上,rmhTNF-α與anti-PD-1無協(xié)同抑瘤作用。這也提示細胞因子在體內作用的復雜性。免疫治療方法的聯(lián)合,應用在不同腫瘤、不同動物模型,可能作用都是不一致的。本研究對未來肺癌免疫治療方式的研究奠定基礎。

      [參考文獻]

      [1] ?Locksley RM,Killeen N,Lenardo MJ. The TNF and TNF receptor superfamilies:integrating mammalian biology [J]. Cell,2001,104(4):487-501.

      [2] ?Van Horssen R,Ten Hagen TL,Eggermont AM. TNF-alpha in cancer treatment:molecular insights,antitumor effects,and clinical utility [J]. Oncologist,2006,11(4):397-408.

      [3] ?Folli S,Pèlegrin A,Chalandon Y,et al. Tumor necrosis fector can enhance radio-antibody uptake in human colon carcinoma xenografts by increasing vascular permeability [J]. Int Cancer,1993,53(5): 829-836.

      [4] ?Garris CS,Arlauckas SP,Kohler RH,et al. Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12 [J]. Immunity,2018,49(6):1148-1161.e7.

      [5] ?Van den Bergh JMJ,Smits ELJM,Berneman ZN,et al. Monocyte-derived dendritic cells with silenced PD-1 ligands and transpresenting interleukin-15 stimulate strong tumor-reactive T-cell expansion [J]. Cancer Immunol Res,2017,5(8):710-715.

      [6] ?Liu Y,Zhang Y,Zheng X,et al. Galantamine improves cognition,hippocampal inflammation,and synaptic plasticity impairments induced by lipopolysaccharide in mice [J]. J Neuroinflammation,2018,15(1):112-116.

      [7] ?Yan Z,Zhao N,Wang Z,et al. A mutated human tumor necrosis factor-alpha improves the therapeutic index in vitro and in vivo [J]. Cytotherapy,2006,8(4): 415-423.

      [8] ?侯志超,伊力亞爾·夏合丁.實體腫瘤組織石蠟切片的免疫組化實驗方法總結[J].世界最新醫(yī)學信息文摘,2017, 17(41): 56.

      [9] ?Ruegg C,Yimaz A,Bieler G,et al. Evidence for the involvement of endothelial cell integrin αVβ3 in the disruption of the tumor vasculature induced by TNF and IFN-γ [J]. Nat Med,1998,4(4):408-414.

      [10] ?Li M,Qin X,Xue X,et al. Safety evaluation and pharmacokinetics of a novel human tumor necrosis factor-alpha exhibited a higher antitumor activity and a lower systemic toxicity [J]. Anticancer Drugs,2010,21(3):243-251.

      [11] ?Li M,Xu T,Zhang Z,et al. Phase Ⅱ multicenter,randomized,double-blind study of recombinant mutated human tumor necrosis factor-a in combination with che-motherapies in cancer patients [J]. Cancer Sci,2012, 103(2):288-295.

      [12] ?Ma X,Song Y,Zhang K,et al. Recombinant mutated human TNF in combination with chemotherapy for stage ⅢB/Ⅳ non-small cell lung cancer:a randomized,phase Ⅲ study [J]. Sci Rep,2015,4:9918.

      [13] ?Siurala M,Havunen R,Saha D,et al. Adenoviral delivery of tumor necrosis factor-α and interleukin-2 enables successful adoptive cell therapy of immunosuppressive melanoma [J]. Mol Ther,2016,24(8):1435-1443.

      [14] ?Elia AR,Grioni M,Basso V,et al. Targeting tumor vasculature with TNF leads effector T cells to the tumor and enhances therapeutic efficacy of immune checkpoint blockers in combination with adoptive cell therapy [J]. Clin Cancer Res,2018,24(9): 2171-2181.

      [15] ?Gettinger S,Horn L,Jackman D,et al. Five-year follow-up of nivolumab in previously treated advanced non-small-cell lung cancer:results from the CA209-003 study [J]. J Clin Oncol,2018,36(17):1675-1684.

      [16] ?Wrangle JM,Velcheti V,Patel MR,et al. ALT-803,an IL-15 superagonist,in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised,open-label,phase 1b trial [J]. Lancet Oncol,2018,19(5): 694-704.

      [17] ?Asano T,Meguri Y,Yoshioka T,et al. PD-1 modulates regulatory T-cell homeostasis during low-dose interleukin-2 therapy [J]. Blood,2017,129(15): 2186-2197.

      [18] ?Lim SO,Li CW,Xia W,et al. Deubiquitination and stabilization of PD-L1 by CSN5 [J]. Cancer Cell,2016,30(6):925-939.

      [19] ?Bertrand F,Montfort A,Marcheteau E,et al. TNF-α blockade overcomes resistance to anti-PD-1 in experimental melanoma [J]. Nat Commun,2017,8(1): 2256.

      [20] ?Perez-Ruiz E,Minute L,Otano I,et al. Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy [J]. Nature,2019,569(7756):428-432.

      (收稿日期:2019-12-04 ?本文編輯:劉永巧)

      猜你喜歡
      藥組生理鹽水生存期
      Physiological Saline
      生理鹽水
      格列美脲聯(lián)合胰島素治療2型糖尿病的臨床效果觀察
      鼻咽癌患者長期生存期的危險因素分析
      胃癌術后患者營養(yǎng)狀況及生存期對生存質量的影響
      術中淋巴結清掃個數對胃癌3年總生存期的影響
      0.9g/dl生理鹽水稀釋解決EDTA依賴性血小板假性減少的方法研究
      試論補陽還五湯中黃芪與活血藥組的配伍意義
      健脾散結法聯(lián)合化療對56例大腸癌Ⅲ、Ⅳ期患者生存期的影響
      不同時期RNA干擾水通道蛋白4對早期創(chuàng)傷性腦水腫影響的觀察
      金山区| 巴彦县| 烟台市| 广德县| 稻城县| 连南| 瓮安县| 福贡县| 屏东市| 康保县| 扎兰屯市| 江达县| 太原市| 固始县| 海南省| 永靖县| 益阳市| 金昌市| 林州市| 汉川市| 东莞市| 宁阳县| 乃东县| 苍山县| 花莲市| 从江县| 汉源县| 南城县| 偃师市| 赤城县| 凤冈县| 涡阳县| 阿鲁科尔沁旗| 滨海县| 肇庆市| 岳阳县| 邹平县| 邢台市| 台东市| 苏州市| 绥江县|