PD―1/PD―L1信号通路在恶性肿瘤免疫治疗中的作用

时间:2022-02-28 02:35:18

PD―1/PD―L1信号通路在恶性肿瘤免疫治疗中的作用

[摘要] 恶性肿瘤是目前一组严重威胁人类健康和生命的疾病,其发病率和死亡率有不断上升的趋势。免疫治疗是除手术、化疗、放疗之外一项重要的抗肿瘤治疗方式,它可通过刺激机体免疫系统提高抗肿瘤免疫效应。程序性死亡分子1(PD-1)和程序性死亡分子1配体(PD-L1)这两个免疫检查分子,通过抑制T细胞免疫与肿瘤的发生、发展密切相关,目前在部分恶性肿瘤即黑色素瘤、非小细胞肺癌(NSCLC)、肾癌、膀胱癌治疗中有重要临床意义。PD-1/PD-L1信号通路的激活可降低T细胞的免疫功能从而使肿瘤发生免疫逃逸,而阻断此通路则可以增强机体内源性的抗肿瘤免疫效应。目前临床试验显示免疫检查点阻滞剂抗PD-1、抗PD-L1抗体在肿瘤免疫治疗中有较好的疗效性及安全性。本综述旨在回顾及总结近年来PD-1/PD-L1信号通路及其阻滞剂在部分恶性肿瘤治疗中的研究进展。

[关键词] 恶性肿瘤;T细胞免疫;程序性死亡分子1;程序性死亡分子1配体;免疫治疗;免疫检查点阻滞剂

[中图分类号] R730.3 [文献标识码] A [文章编号] 1673-7210(2016)04(c)-0057-04

[Abstract] Malignancy is a group of diseases which have serious threat to human health and in recent years its morbidity and mortality has a rising trend. Immunotherapy is an important way of anti-tumor therapy, in addition to surgery, chemotherapy and radiotherapy. It can stimulate the body's immune system to improve the anti-tumor immune effects. Study had been reported that immune checkpoint molecules, including programmed death-1 (PD-1)/PD-ligand (L) 1 axis, are closely related with cancer generation and development, by inhibiting T cell immune and play a key role on clinical significance of malignancy, including Melanoma Non-small cell lung cancer (NSCLC), Renal cell carcinoma (RCC), Bladder cancer. Activation of PD-1/PD-L1 pathway contributes to tumor immune escape, and block PD-1/PD-L1 pathway can enhance endogenous antimuor immunity. Currently increasing clinical trials suggested that immune checkpoint inhibitors, including anti-PD-1 and anti-PD-L1 monoclonal antibodies turned out to be beneficial and safe in malignancy. This artical provides a review on the progress of PD-1/PD-L1 pathway and immune checkpoint inhibitors in some malignancy.

[Key words] Malignancy; T cell immune; Programmed death 1; Programmed death 1 ligand; Immuno-therapy; Checkpoint inhibitors

恶性肿瘤严重威胁人类的健康,其病因、发病机制、临床表现、治疗方法等十分复杂,随着肿瘤学、免疫学及分子生物学发展的交叉渗透,免疫治疗为恶性肿瘤治疗带来了新希望。机体免疫系统的监视功能在清除肿瘤细胞中起重要作用,但有时却促进肿瘤发生免疫逃逸、免疫耐受,这在肿瘤的发生发展过程中起不可忽略的作用。程序性死亡分子1(programmed death 1,PD-1)是CD28-B7受体家族的新成员,其在调节免疫反应中发挥着重要作用[1],配体有PD-1配体(PD-1 ligand,PD-L1)[2-3]和PD-2配体(PD-2 ligand,PD-L2)[4-5],本文主要讲述PD-L1。PD-L1调节免疫反应主要是通过下调T细胞受体的信号[6],PD-1含有290个氨基酸,是I型糖蛋白[7],T细胞抑制受体与PD-L1相互作用传递抑制性信号,在免疫紊乱中发挥作用。PD-L1蛋白在正常组织中几乎检测不到,但在T细胞和内皮细胞上经诱导可表达PD-L1,人类多种肿瘤中富含PD-L1,肿瘤相关PD-L1可增加抗原特异性肿瘤细胞凋亡。研究表明,PD-1/ PD-L1信号通路的激活在免疫抑制性肿瘤的微环境形成中起作用[8],阻断PD-1/PD-L1信号通路的激活有抑制肿瘤免疫微环境形成的作用,上调T细胞免疫功能的杀伤及监测功能使机体抗肿瘤的免疫作用提高[9-11]。PD-1/PD-L1是新发现的负向调节T细胞活化的协同激活通路,抗PD-1/PD-L1抗体用于治疗黑色素瘤、非小细胞肺癌(NSCLS)及肾癌[12-14]。

1 T细胞的抗肿瘤免疫作用

CD4+Th细胞能激活肿瘤特异性细胞毒性T淋巴细胞(cytotoxic lymphocyte,CTL),其在抗肿瘤中起到重要作用。CTL主要功能是特异性直接杀伤肿瘤细胞。CD4+Tregs细胞对CTL(CD8+T细胞)具有免疫抑制作用,而CD8+ T细胞具有肿瘤免疫监视、杀伤癌细胞和免疫记忆功能。人类CTL特征表型中CD8+T细胞可直接作为效应细胞特异地杀伤肿瘤细胞。恶性肿瘤常导致机体内环境失衡免疫功能紊乱,CD3+、CD4+T淋巴细胞含量明显低于正常人,CD3+/CD8+ T淋巴细胞比例上升,CD4+/CD8+比值下降[15-16],肿瘤细胞生长及进展加速。CD8+属抑制性淋巴T细胞,其含量越少机体免疫力越低。研究显示在一些肿瘤中CD8+T淋巴细胞不同程度地表达PD-1,且与临床肿瘤分期、转移相关。PD-1通过与肿瘤细胞表面的配体结合,抑制CD8+T细胞的肿瘤免疫应答,介导肿瘤免疫逃逸在肿瘤发生发展中起作用[17-19]。

2 PD-1/PD-L1信号通路及其免疫抑制作用

完整的免疫系统可通过免疫检查来识别和消除肿瘤细胞,但肿瘤却可适应及逃避这些防御机制[20-23]。PD-1最初在凋亡T细胞杂交瘤中发现[24],PD-1在外周组织中可调节效应T细胞对肿瘤侵袭的效应[25]。PD-L1表达在抗原提呈细胞表面,研究发现在一些肿瘤细胞中高表达[26-28]。肿瘤微环境中肿瘤细胞的浸润抑制机体的抗肿瘤免疫效应,其中TregT细胞与耐受性DCs可能与肿瘤的不良愈后相关[29]。表达于Treg的PD-1可促进Treg细胞的增殖,从而抑制免疫应答[26]。肿瘤细胞高/过表达的PD-1配体,可导致T细胞功能减弱、失能甚至死亡。研究显示,肿瘤相关PD-L1与肿瘤微环境中的TILs耗竭相关,使用抗PD-1或抗PD-L1抗体可以通过抑制Treg细胞及调节耐受性DCs增强效应性CD8+T细胞抗肿瘤效应[29]。Zhang等[17]从21例NSCLC患者中证实在肿瘤组织中CD8+T细胞PD-1的表达远高于外周血单核淋巴细胞(PBMC)中CD8+T细胞PD-1的表达,且无论PBMC中的CD8+T细胞来源于健康人还是肿瘤患者(P < 0.01)。并发现对同一患者,其肿瘤组织中CD8+T细胞PD-1的表达也远高于PBMC中CD8+T细胞的表达(P < 0.01)。针对PD-1信号通路的药物起到抗肿瘤作用尤其是PD-L1高表达的肿瘤,也与愈后不良相关[31-33]。

3 免疫检查点抑制剂抗PD-1、PD-L1抗体与免疫治疗

免疫检查点控制共抑和共刺激信号平衡的作用可调节T细胞应答持续时间和幅值。肿瘤可能采用免疫检查点导致肿瘤不平衡增长及逃避宿主监控。研究证实:PD-1在乳腺癌和黑色素瘤的高表达与肿瘤的分级、大小、淋巴结状态及转移相关[19,30]。PD-L1与黑色素瘤、NSCLS、肾癌的预后、复发相关[34]。现通过针对PD-1/PD-L1的免疫检查点抑制剂恢复肿瘤的再平衡与宿主免疫监控来治疗一些肿瘤[35]。

3.1 抗PD-1抗体

Nivolumab(BMS-936558,MDX-1106,ONO-4538)是全人源化免疫球蛋白G4、抗PD-1抗体。2014年美国FDA已批准Nivolumab用于治疗不可切除的或ipilimumab治疗后进展的或BRAF V600突变阳性用BRAF抑制剂治疗后进展的黑色素瘤患者[36]。针对BRAF V600突变的肿瘤患者,Topalian等[26]设计的Ⅰ期试验,共107名患者使用剂量在0.1~10 mg/kg的Nivolumab,每两周一次,共96周。结果显示107名患者中使用17个月后34名患者达32%CR,1年和2年生存率分别为62%和43%。2015年FDA批准Nivolumab用于以铂类为基础化疗后进展的转移性非鳞状NSCLC[1638828]。

Pembrolizumab(MK-3475)是人源化免疫球蛋白G4,最早批准用于治疗晚期黑色素瘤[37]。研究显示MK-3475治疗进展期黑色素瘤和NSCLC的客观缓解率分别为38%和21%[38-39]。

3.2 抗PD-L1抗体

MPDL3280A MEDI4736 BMS-936559是人源化IgG4抗体。PD-L1存在于肿瘤细胞质膜的细胞质中,但并不是所有的肿瘤细胞都表达PD-L1[40-41]。研究显示,预处理PD-L1直接表达在肿瘤活组织检查采用相应的抗PD-L1治疗,对于一些PD-L1阴性的肿瘤治疗效果不明显[42-44]。

各种不同临床试验阶段免疫检查点抑制剂结果之间的差异可能在于抗体亲和力。应注意的是PD-1、PD-L1阻滞剂仅在高表达PD-1及PD-L1的患者中有效。Brahmer等[45]评估BMS-936558在恶性肿瘤I期临床试验的有效性结果示25例肿瘤患者中9例表现出强烈反应[45],而53例中17例PD-L1阴性患者对阻滞剂无反应[46-47]。这表明PD-L1表达可以作为肿瘤患者进一步分层的一个潜在生物标志物并预测抗PD-L1免疫治疗的疗效。

4 小结

肿瘤免疫治疗是继手术、化疗、放疗及中医治疗后的一种新治疗策略,本综述就肿瘤免疫治疗靶点PD-1、PD-L1进行相关阐述,随着精准医疗的发展,免疫治疗是其发展的一个重要方向。抗PD-1/PD-L1抗体在一些治疗中表现出迅速、有效的药物反应为恶性肿瘤的免疫治疗奠定了基础,但PD-1/PD-L1信号通路阻滞剂对肿瘤患者的预后及其带来的副作用有待进一步研究。另实践中PD-L1阻滞剂是否优于PD-1阻滞剂并不明确。免疫治疗作为肿瘤治疗的新方向探索肿瘤和机体免疫之间的关系,达到个体及优化治疗是未来需解决的问题。

[参考文献]

[1] Francisco LM,Sage PT,Sharpe AH. The PD-1 pathway in tolerance and autoimmunity [J]. Immunolngagem Rev,2010,236:219-242.

[2] Oezcan T,Ching-Hung S,Lieping C,et al. B7-H1 (PD-L1) on T cells is required for T-cell-mediated conditioning of dendritic cell maturation [J]. Proc Natl Acad Sci USA,2009,106(8):2741-2746.

[3] Freeman GJ,Long AJ,Yoshiko I,et al. Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 Family Member Leads to Negative Regulation of Lymphocyte Activation [J]. J Exp Med,2000,192(7):1027-1034.

[4] Marguerite G,Laurent G,Nacer S,et al. PD-L1 and PD-L2 differ in their molecular mechanisms of interaction with PD-1 [J]. Int Immunol,2010,22(8):651-660.

[5] Tseng SY,Otsuji M,Gorski K,et al. B7-DC,a new dendritic cell molecule with potent costimulatory properties for T cells [J]. J Exp Med,2001,193(7):839-846.

[6] Bryan LJ,Gordon LI. Blocking tumor escape in hematologic malignancies:The anti-PD-1 strategy [J]. Blood Rev,2015,29(1):25-32.

[7] Velcheti V,Rimm DL,Schalper KA. Sarcomatoid lung carcinomas show high levels of programmed death ligand1(PD-L1) [J]. J Thorac Oncol,2013,8(6):803-805.

[8] Pardol l DM. The blockade of immune checkpoints in cancer immunotherapy [J]. Nat Rev Cancer,2012,12(4):252-264.

[9] Yael SB,Jun SL,Xingxing Z. T cell coinhibition in prostate cancer:new immune evasion pathways and emerging therapeutics [J]. Trends Mol Med,2011,17(1):47-55.

[10] Topalian SL,Drake CG,Pardoll DM,et al. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity [J]. Curr Opin Immunol,2012,24(2):207-212.

[11] Iwai Y,Ishida M,Tanaka Y,et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade [J]. Proc Natl Acad Sci USA,2002,99(19):12293-12297.

[12] Topalian SL,Hodi FS,Brahmer JR,et al. Safety,activity,and immune correlates of anti-PD-1 antibody in cancer [J]. N Engl J Med,2012, 366(26):2443-2454.

[13] Brahmer JR,Tykodi SS,Chow LQ,et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer [J]. N Engl J Med,2012,366(26):2455-2465.

[14] Hamid O,Carvajal RD. Anti-programmed death-1 and anti-programmed death-ligand 1 antibodies in cancer therapy [J]. Expert Opin Biol Ther,2013,13(6):847-861.

[15] Das S,Karim S,Datta Ray C,et al. Peripheral blood lymphocyte subpopulations in patients with cervical cancer [J]. Int J Gynaecol Obstet,2007,98(2):143-146.

[16] Wang L,Shen Y. Imbalance of circulating T-lymphocyte subpopulation in gastric cancer patiens correlated with performance status [J]. Clin Lab,2013,59(3/4):429-433.

[17] Zhang Y,Huang S,Gong D,et al. Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8+ T lymphocytes in human non-small cell lung cancer [J]. Cell Mol Immunol,2010,7(5):389-395.

[18] Mellman I,Coukos G,Dranoff G. Cancer immunotherapy comes of age [J]. Nature,2011,480(7378):480-489.

[19] Chapon M,Randriamampita C,Maubec E,et al. Progressive upregulation of PD-1 in primary and metastatic melanomas associated with blunted TCR signaling in infiltrating T lymphocytes [J]. J Invest Dermatol,2011,131(6):1300-1307.

[20] Disis ML. Immune regulation of cancer [J]. J Clin Oncol,2010,28(29):4531-4538.

[21] Anil Shanke,Marincola FM. Cooperativity of adaptive and innate immunity:implications for cancer therapy [J]. Cancer Immunol Immunother,2011,60(8):1061-1074.

[22] Dunn GP,Bruce AT,Ikeda H,et al. Cancer immunoediting:from immunosurveillance to tumor escape [J]. Nat Immunol,2002,3(11):991-998.

[23] Dolan DE,Gupta S. PD-1 pathway inhibitors:changing the landscape of cancer immunotherapy [J]. Cancer Control,2014,21(3):231-237.

[24] Ishida Y,Agata Y,Shibahara K,et al. Induced expression of PD-1,a novel member of the immunoglobulin gene superfamily,upon programmed cell death [J]. EMBO,1992,11(11):3887-3895.

[25] Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy [J]. Nat Rev Cancer,2012,12(4):252-264.

[26] Yang CY,Lin MW,Chang YL,et al. Programmed cell death-ligand 1 expression in surgically resected stageⅠ pulmonary adenocarcinoma and its correlation with driver mutations and clinical outcomes [J]. Eur J Cancer,2014,50(7):1361-1369.

[27] Zhang Y,Wang L,Li Y,et al. Protein expression of programmed death 1 ligand 1 and ligand 2 independently predict poor prognosis in surgically resected lung adenocarcinoma [J]. Onco Targets Ther,2014,7:567-573.

[28] Taube JM,Young GD,Topalian SL,et al. Differential Expression of Immune-Regulatory Genes Associated with PD-L1 Display in Melanoma:Implications for PD-1 Pathway Blockade [J]. Clin Cancer Res,2015,21(17):3969-3976.

[29] Larsen SK. Cellular immune responses towards regulatory cells [J]. Dan Med J,2016,63(1):5188.

[30] Francisco LM,Salinas VH,Brown K E,et al. PD-L1 regulates the development,maintenance,and function of induced regulatory T cells [J]. J Exp Med,2009,206(13):3015-3029.

[31] Valero-Pacheco N,Arriaga-Pizano L,Ferat-Osorio E,et al. PD-L1 expression induced by the 2009 pandemic influenza A( H1N1) virus impairs the human T cell response [J/OL]. Clin Dev Immunol,2013,2013:989673.

上一篇:七叶皂苷钠治疗2型糖尿病胃旁路术术后胃轻瘫的... 下一篇:白蛋白结合型紫杉醇治疗晚期宫颈癌的效果及安...