LAG-3分子的研究进展

田金, 张腾龙, 李玲, 王洁, 郭成业

新医学 ›› 2020, Vol. 51 ›› Issue (9) : 663-666.

PDF(753 KB)
PDF(753 KB)
新医学 ›› 2020, Vol. 51 ›› Issue (9) : 663-666. DOI: 10.3969/j.issn.0253-9802.2020.09.004
综述

LAG-3分子的研究进展

作者信息 +

Research progress on LAG-3 molecule

Author information +
文章历史 +

摘要

淋巴细胞活化基因-3(LAG-3)是Ig超家族的成员之一,主要表达于活化的自然杀伤细胞、T淋巴细胞表面,与主要组织相融性复合体-Ⅱ(MHC-Ⅱ)类分子结合,阻断T淋巴细胞抗原受体(TCR)与MHC-Ⅱ分子的结合,抑制T淋巴细胞的激活,从而发挥负性免疫调节的功能。LAG-3与多种类型肿瘤、慢性炎症等疾病相关。其发现为多种疾病的免疫治疗提供了新的靶点。该文对LAG-3的结构、表达、作用机制,及其与肺癌、胃癌等肿瘤的关系进行综述,并探讨LAG-3在临床应用中的前景。

Abstract

Lymphocyte activation gene-3 (LAG-3) is a member of immunoglobulin superfamily. LAG-3 molecule is mainly expressed on the surface of activated natural killer (NK) cells and T lymphocytes, and binds with polypeptide-major histocompatibility complex - Ⅱ (MHC-Ⅱ) molecules to block the combination of T cell antigen receptor (TCR) and MHC-Ⅱ molecules, suppress the activation of T lymphocytes and exert the function of negative immune regulation. LAG-3 is associated with multiple types of tumors, chronic inflammation and other diseases. The discovery of LAG-3 provides novel targets for the immunotherapy of various diseases. In this article, the structure, expression and mechanism of LAG-3 and its relationship with lung cancer, gastric cancer and other malignant tumors were reviewed, and the application value of LAG-3 in clinical practice was investigated.

关键词

淋巴细胞活化基因-3 / T淋巴细胞 / 肿瘤 / 免疫检查点 / 免疫治疗

Key words

Lymphocyte activation gene-3 / T cell / Tumor / Immune checkpoint / Immunotherapy

引用本文

导出引用
田金 , 张腾龙 , 李玲 , 王洁 , 郭成业. LAG-3分子的研究进展[J]. 新医学, 2020, 51(9): 663-666 https://doi.org/10.3969/j.issn.0253-9802.2020.09.004
Tian Jin , Zhang Tenglong , Li Ling , Wang Jie , Guo Chengye. Research progress on LAG-3 molecule[J]. JOURNAL OF NEW MEDICINE, 2020, 51(9): 663-666 https://doi.org/10.3969/j.issn.0253-9802.2020.09.004
人体的免疫调节机制是维持免疫系统正常运行的关键,多种细胞表面分子参与了免疫调节过程。近年,具有启动T淋巴细胞耗竭作用的共抑制分子淋巴细胞活化基因3(LAG-3,又称CD223)分子逐渐受到了关注。LAG-3是Ig超家族的成员之一,对淋巴细胞具有抑制作用。LAG-3可以增强调节性T淋巴细胞(Treg)的负性调节功能,在机体的免疫应答中发挥着重要作用。业已证实,LAG-3参与了多种肿瘤细胞的免疫逃逸[1]。为加深临床对LAG-3的了解,本文就LAG-3的结构、表达、作用机制,及其与肺癌、胃癌等肿瘤的关系进行综述,并探讨LAG-3在临床应用中的前景,现报告如下。

一、LAG-3分子的结构

LAG-3于1990年作为膜蛋白首次被克隆出来。它属于Ig超家族,是一种跨膜糖蛋白。成熟的LAG-3分子是由470个氨基酸构成,相对分子量70 kDa,表达LAG-3分子的基因定位于人12号染色体上,与表达CD4分子的基因在染色体上的定位类似,它们有共同的外显子和内含子。2个分子有部分氨基酸相同。LAG-3和CD4在肽序列和外显子或内含子结构方面的比较分析表明,这2种分子密切相关。这一点因发现2个基因均位于12号染色体短臂的远端而得到加强。LAG-3基因包括8个外显子,相应的模板DNA编码1个498个氨基酸的膜蛋白,有4个细胞外Ig超家族类似(SF)结构域,即D1 ~ D4。D1包含1个额外的环状结构和1个非典型链内二硫键,在D1与D3区域间、D2与D4区域间,有很多氨基酸序列是一致的。因此,LAG-3很可能是通过复制1个预先存在的编码2个IgSF结构域结构的基因进化而来的[2]
可溶性LAG-3(sLAG-3)在T淋巴细胞介导的免疫中起着不可忽视的作用。sLAG-3是细胞表面的LAG-3分子在去整合素-金属蛋白酶10(ADAM10)和ADAM17作用下形成并释放到细胞外的N-末端胞外段,这一过程在T淋巴细胞激活后可以显著增强。sLAG-3仅能与抗原呈递细胞少数亚群中脂筏区域中的主要组织相融性复合体-Ⅱ(MHC-Ⅱ)类分子结合。有研究显示,将sLAG-3-Ig融合蛋白(IMP321)作用于小鼠中,可增强T淋巴细胞介导的抗肿瘤免疫功能[3,4]。目前IMP321试剂作为免疫增强剂已经进入临床试验阶段,该临床试验将评估IMP321与抗病毒疫苗、化学治疗或其他抗肿瘤治疗之间的联合用药情况。

二、LAG-3的表达及其作用机制

LAG-3主要表达于活化的CD4+和CD8+ T淋巴细胞中,在自然杀伤细胞中也有表达,但幼稚及正常的T淋巴细胞仅表达低水平的LAG-3,LAG-3表达水平在被肿瘤抗原刺激时大幅度提升,提示阻断LAG-3可能会增强抗肿瘤免疫。薛妮娜等[5]在研究不同刺激剂对小鼠脾脏淋巴细胞LAG-3表达及功能影响时发现,伴刀豆球蛋白A(conA)培养24 h或72 h可剂量依赖性增加LAG-3+CD3+和LAG-3+CD3+CD4+ T淋巴细胞比例,CD3/28抗体培养72 h增加LAG-3+CD3+和LAG-3+CD3+CD4+ T淋巴细胞比例,ConA和CD3/28抗体是T淋巴细胞活化的有效刺激剂,并可促进T淋巴细胞中LAG-3的表达。魏慧等[6]报道,蕨麻多糖能促进急性低剂量镉染毒小鼠脾淋巴细胞分泌IFN-γ和sLAG-3,CD3+、CD4+ T淋巴细胞亦相应增加。J.CaM1.6细胞、CEM细胞和JurkatE6-1细胞均为来自人急性淋巴细胞白血病细胞系的不同细胞株。郝书民等[7]在探讨表观遗传因素DNA甲基化对人T淋巴细胞系LAG-3表达的影响中发现,LAG-3表达水平在J.CaM1.6和CEM细胞中极低,在JurkatE6-1细胞中相对较高。不同细胞系中,CpG岛均高度甲基化,CpG岛在J.CaM1.6和CEM细胞中高度甲基化,但在JurkatE6-1细胞中甲基化水平较低。CpG岛的甲基化与去甲基化可能是LAG-3转录的表观调控开关。
LAG-3是共抑制分子中的一员,负性调节T淋巴细胞的功能。它与MHC-Ⅱ类分子结合,阻断T淋巴细胞抗原受体(TCR)与MHC-Ⅱ分子的结合,抑制T淋巴细胞的激活,从而发挥负性免疫调节的功能,维持内环境稳定。纤维蛋白原相关蛋白1(FGL1)为肝脏分泌蛋白,是一种独立于MHC-Ⅱ的主要LAG-3功能配体。Wang等[8]的研究显示,FGL1抑制抗原特异性T淋巴细胞活化,其在小鼠体内的消融促进T淋巴细胞免疫。单克隆抗体阻断FGL1/LAG-3相互作用可刺激肿瘤免疫,并以受体-配体相互依赖的方式治疗已建立的小鼠肿瘤。FGL1是人类恶性肿瘤细胞高度分泌的产物,恶性肿瘤患者血浆中FGL1的升高与预后不良和抗程序性死亡受体-1及其配体(PD-1/PD-L1)疗法的耐药有关。Wang等的研究揭示了一种免疫逃避机制,并对恶性肿瘤免疫疗法的设计有所启示。

三、LAG-3与肿瘤增殖、进展、侵袭等之间的关系

近年研究显示在非小细胞肺癌(NSCLC)患者外周血和肿瘤组织中,CD4+ T淋巴细胞表面共抑制分子细胞毒性T淋巴细胞相关蛋白4(CTLA-4)、LAG-3和PD-1的表达增加,这可能是参与肿瘤细胞免疫逃逸,促进NSCLC疾病进展和预后不良的机制之一[9]。马成龙等[10]报道,NSCLC和良性病变肺组织中LAG-3表达差异有统计学意义,其表达与NSCLC临床分期有关,在Ⅲ期患者中LAG-3表达较高,而且LAG-3不仅在肿瘤浸润淋巴细胞中表达,还在肿瘤细胞上异位表达,并与临床分期有关。黄维等[11]在NSCLC患者肿瘤组织中免疫检查点表达与临床特征及预后的关系研究中发现,不同临床分期患者NSCLC组织中的LAG-3阳性表达率存在差异,LAG-3阳性表达患者的中位无病生存(DFS)时间长于LAG-3阴性表达患者,LAG-3阳性表达患者的3年DFS率高于LAG-3阴性表达患者,因而认为LAG-3是NSCLC患者预后的影响因素。但是LAG-3的表达水平与NSCLC 患者预后是正相关还是负相关,还有待进一步研究。
何兴桢等[12]报道,胃癌患者存在明显的LAG-3+细胞/CCR类趋化因子受体3(CCR3)+细胞的比例失调,这在一定程度上显示胃癌患者存在着Th1/Th2比例失调及免疫失衡,可能是胃癌发病的重要因素之一。江露等[13]检测了sLAG-3在胃癌患者的表达,结果显示胃癌患者血清sLAG-3水平低于健康对照组血清sLAG-3水平,且胃癌患者血清sLAG-3水平与肿瘤分期、分化程度及浸润深度均有关,sLAG-3诊断胃癌的灵敏度和准确度均高于癌胚抗原,所以sLAG-3可作为胃癌诊断的潜在血清标志物。刘玉芳[14]报道,血清LAG-3水平对早期胃癌具有诊断价值,血清LAG-3水平可评估胃癌组织中癌细胞增殖、侵袭及血管新生的程度。由此可见,LAG-3未来在消化道肿瘤的诊断中将占有重要地位。
Matsuzaki等[15]进行的研究显示,霍奇金淋巴瘤的组织和外周血中LAG-3表达高于正常组织中,肿瘤组织中特异性抗肿瘤CD8+ T淋巴细胞功能缺陷被抑制,敲除LAG-3+的T淋巴细胞,特异性CD8+ T淋巴细胞的抗肿瘤功能得以恢复,细胞因子分泌增加,抗肿瘤功能得到不同程度恢复。目前LAG-3分子在血液肿瘤中的研究较少,未来LAG-3分子或许可为血液肿瘤的治疗打开一扇大门。
在我国,食管癌的最常见类型是食管鳞癌,近年有关食管鳞癌的免疫治疗研究的前期结果较为满意[16]。孙楠等[17]的研究显示,CTLA4、LAG-3和T淋巴细胞Ig黏液素3(TIM3)在食管鳞癌中的表达水平均高于正常组织,且食管鳞癌患者的预后与PD-1和LAG-3的表达密切相关,PD-1或LAG-3表达水平较高者具有较好的预后,但PD-1和LAG-3的表达与总体生存率无关。
宫颈癌的发生发展与T淋巴细胞介导的抗肿瘤免疫功能被抑制有关。黄群欢等[18]报道,宫颈癌Ⅲ期患者LAG-3水平高于健康对照组,其余各期患者LAG-3水平与健康对照组相近。关于LAG-3与宫颈癌的关系报道较少,因此关于LAG-3与宫颈癌的相互作用关系还需进一步研究探讨。
子宫内膜癌(EC)是女性生殖系统的三大恶性肿瘤之一。近年来,我国女性EC发病率呈上升趋势。张丽萍等[19]按Stelloo等(2015年)的分型规则将EC分为POLE突变型、MSI型、NSMP型和TP53突变型进行研究,结果显示作为LAG-3的配体的FGL1主要在MSI及NSMP两种亚型中高表达。而且,FGL1高表达组的总体生存期较低表达组更长,FGL1的高表达与无复发生存期延长显著相关。作者进一步分析LAG-3/FGL1基因表达与病理类型的关系,结果显示LAG-3在子宫内膜样癌、混合型癌、浆液性癌中的表达水平相近,FGL1在子宫内膜样癌中的表达水平高于另外2种类型。该研究通过分析LAG-3/FGL1表达与肿瘤微浸润淋巴细胞的关系,推测FGL1主要表达于癌细胞,FGL1高表达的肿瘤组织中,肿瘤突变负荷显著升高,由此认为LAG-3/FGL1通路是EC潜在的免疫治疗靶点。
免疫检查点抑制剂为恶性肿瘤的治疗提供了新的策略,也为研究和探索STS提供了机会。软组织肉瘤(STS)是一种异质性的罕见间充质肿瘤。转移性或晚期STS的预后很差。Que等[20]检测了12例STS患者和11名健康人的外周血T淋巴细胞,结果显示STS患者的外周血CD8+和CD4+ T淋巴细胞LAG-3表达水平高于健康人。为了进一步确定LAG-3在STS中的表达,他们对163例接受广泛可切除手术的患者的STS组织样本进行了免疫组织化学染色,结果显示LAG-3的高表达与高病理分级和更晚期的肿瘤阶段相关。该研究同时在人STS组织样本中进行了免疫荧光检测,并主要在肿瘤浸润CD8+ T淋巴细胞中检测LAG-3的表达和定位,结果表明肿瘤浸润性T淋巴细胞特别是CD8+ T淋巴细胞中LAG-3表达水平高。研究表明,阻断LAG-3对于恢复CD8+ T淋巴细胞的抗肿瘤免疫能力是必要的,且可能是提高STS靶向治疗效果的有效策略。

四、展望

肿瘤细胞诱导的T淋巴细胞无能是肿瘤细胞的免疫逃逸机制[21]。目前多种免疫检查点抑制剂已经获批在临床应用,如伊匹单抗(靶向CTLA-4)、纳武利尤单抗和派姆单抗(靶向PD-1)。目前LAG-3靶向制剂也已进入临床试验。LAG-3分子是一类表达于活化淋巴细胞的分子,在不同类型细胞中表达水平不同,在内环境中可以负性调节T淋巴细胞的功能,参与了自身免疫病、慢性感染性疾病、肿瘤甚至神经退变性疾病的发生发展。阻断LAG-3的表达可以增强T淋巴细胞介导的抗肿瘤作用[15, 22]。此外,有研究显示同时阻断LAG-3和PD-1两条通路的抗肿瘤效应优于单独阻断其中任何一条通路,因此学者们认为LAG-3与PD-1在肿瘤免疫逃逸机制中起协同作用[8, 15, 22]。各种免疫检查点抑制剂之间如何协同作用,如何最大化免疫检查点抑制剂的临床效益,是肿瘤领域未来的研究热点。

参考文献

[1]
Goldberg MV, Drake CG. LAG-3 in cancer immunotherapy. Curr Top Microbiol Immunol, 2011,344:269-278.
LAG-3 (CD223) is a cell surface molecule expressed on activated T cells (Huard et al. Immunogenetics 39:213-217, 1994), NK cells (Triebel et al. J Exp Med 171:1393-1405, 1990), B cells (Kisielow et al. Eur J Immunol 35:2081-2088, 2005), and plasmacytoid dendritic cells (Workman et al. J Immunol 182:1885-1891, 2009) that plays an important but incompletely understood role in the function of these lymphocyte subsets. In addition, the interaction between LAG-3 and its major ligand, Class II MHC, is thought to play a role in modulating dendritic cell function (Andreae et al. J Immunol 168:3874-3880, 2002). Recent preclinical studies have documented a role for LAG-3 in CD8 T cell exhaustion (Blackburn et al. Nat Immunol 10:29-37, 2009), and blockade of the LAG-3/Class II interaction using a LAG-3 Ig fusion protein is being evaluated in a number of clinical trials in cancer patients. In this review, we will first discuss the basic structural and functional biology of LAG-3, followed by a review of preclinical and clinical data pertinent to a role for LAG-3 in cancer immunotherapy.
[2]
Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, Hercend T. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med, 1990,171(5):1393-405.
We have identified a novel human gene of the Ig superfamily, designated LAG-3. Expression of this gene is undetectable in resting PBL, while it is found (a 2-kb message) in activated T and NK cells. The LAG-3 gene includes eight exons; the corresponding cDNA encodes a 498-amino acid membrane protein with four extracellular IgSF domains. The first one belongs to the V-SET; it is particular since it includes an extra loop in the middle of the domain and an unusual intrachain disulphide bridge. The three other domains belong to the C2-SET. Strong internal homologies are found in the LAG-3 molecule between domains 1 and 3, as well as between domains 2 and 4. It is therefore likely that LAG-3 has evolved by duplication of a pre-existing gene encoding a two IgSF-domain structure. The compared analysis of LAG-3 and CD4, with respect to both their peptidic sequence as well as their exon/intron organization, indicated that the two molecules are closely related. This point is strengthened by the finding that both genes are located on the distal part of the short arm of chromosome 12.
[3]
Brignone C, Gutierrez M, Mefti F, Brain E, Jarcau R, Cvitkovic F, Bousetta N, Medioni J, Gligorov J, Grygar C, Marcu M, Triebel F. First-line chemoimmunotherapy in metastatic breast carcinoma: combination of paclitaxel and IMP321 (LAG-3Ig) enhances immune responses and antitumor activity. J Transl Med, 2010,8:71.
BACKGROUND: IMP321 is a recombinant soluble LAG-3Ig fusion protein that binds to MHC class II with high avidity and mediates APC and then antigen-experienced memory CD8+ T cell activation. We report clinical and biological results of a phase I/II in patients with metastatic breast carcinoma (MBC) receiving first-line paclitaxel weekly, 3 weeks out of 4. METHODS: MBC patients were administered one dose of IMP321 s.c. every two weeks for a total of 24 weeks (12 injections). The repeated single doses were administered the day after chemotherapy at D2 and D16 of the 28-day cycles of paclitaxel (80 mg/m2 at D1, D8 and D15, for 6 cycles). Blood samples were taken 13 days after the sixth and the twelfth IMP321 injections to determine sustained APC, NK and memory CD8 T cell responses. RESULTS: Thirty MBC patients received IMP321 in three cohorts (doses: 0.25, 1.25 and 6.25 mg). IMP321 induced both a sustained increase in the number and activation of APC (monocytes and dendritic cells) and an increase in the percentage of NK and long-lived cytotoxic effector-memory CD8 T cells. Clinical benefit was observed for 90% of patients with only 3 progressors at 6 months. Also, the objective tumor response rate of 50% compared favorably to the 25% rate reported in the historical control group. CONCLUSIONS: The absence of toxicity and the demonstration of activity strongly support the future development of this agent for clinical use in combined first-line regimens. TRIAL REGISTRATION: ClinicalTrials.gov NCT00349934.
[4]
Romano E, Michielin O, Voelter V, Laurent J, Bichat H, Stravodimou A, Romero P, Speiser DE, Triebel F, Leyvraz S, Harari A. MART-1 peptide vaccination plus IMP321 (LAG-3Ig fusion protein) in patients receiving autologous PBMCs after lymphodepletion: results of a Phase I trial. J Transl Med, 2014,12:97.
BACKGROUND: Immunotherapy offers a promising novel approach for the treatment of cancer and both adoptive T-cell transfer and immune modulation lead to regression of advanced melanoma. However, the potential synergy between these two strategies remains unclear. METHODS: We investigated in 12 patients with advanced stage IV melanoma the effect of multiple MART-1 analog peptide vaccinations with (n = 6) or without (n = 6) IMP321 (LAG-3Ig fusion protein) as an adjuvant in combination with lymphodepleting chemotherapy and adoptive transfer of autologous PBMCs at day (D) 0 (Trial registration No: NCT00324623). All patients were selected on the basis of ex vivo detectable MART-1-specific CD8 T-cell responses and immunized at D0, 8, 15, 22, 28, 52, and 74 post-reinfusion. RESULTS: After immunization, a significant expansion of MART-1-specific CD8 T cells was measured in 83% (n = 5/6) and 17% (n = 1/6) of patients from the IMP321 and control groups, respectively (P < 0.02.="" compared="" to="" the="" control="" group="" the="" mean="" fold="" increase="" of="" mart-1-specific="" cd8="" t="" cells="" in="" the="" imp321="" group="" was="" respectively="">2-, >4- and >6-fold higher at D15, D30 and D60 (P < 0.02). Long-lasting MART-1-specific CD8 T-cell responses were significantly associated with IMP321 (P < 0.02). At the peak of the response, MART-1-specific CD8 T cells contained higher proportions of effector (CCR7(-) CD45RA(+)/(-)) cells in the IMP321 group (P < 0.02) and showed no sign of exhaustion (i.e. were mostly PD1(-)CD160(-)TIM3(-)LAG3(-)2B4(+)/(-)). Moreover, IMP321 was associated with a significantly reduced expansion of regulatory T cells (P < 0.04="" consistently="" we="" observed="" a="" negative="" correlation="" between="" the="" relative="" expansion="" of="" mart-1-specific="" cd8="" t="" cells="" and="" of="" regulatory="" t="" cells.="" finally="" although="" there="" were="" no="" confirmed="" responses="" as="" per="" recist="" criteria="" a="" transient="" 30-day="" partial="" response="" was="" observed="" in="" a="" patient="" from="" the="" imp321="" group.="" conclusions:="" vaccination="" with="" imp321="" as="" an="" adjuvant="" in="" combination="" with="" lymphodepleting="" chemotherapy="" and="" adoptive="" transfer="" of="" autologous="" pbmcs="" induced="" more="" robust="" and="" durable="" cellular="" antitumor="" immune="" responses="" supporting="" further="" development="" of="" imp321="" as="" an="" adjuvant="" for="" future="" immunotherapeutic="" strategies.="">
[5]
薛妮娜, 王春阳, 陈越, 王东杰, 来芳芳, 陈晓光. 不同刺激剂对小鼠脾脏淋巴细胞LAG3表达及功能的影响. 中国生化药物杂志, 2017,37(2):1-4.
[6]
魏慧, 何多谊, 白德成, 程菊, 沈蓉, 张倩倩. 蕨麻多糖对小鼠脾淋巴细胞上清液IFN-γ和可溶性LAG-3分子水平的影响. 生物技术进展, 2015,5(4):321-324,329.
[7]
郝书民, 仇超, 张林霞, 张晓燕, 徐建青. CpG岛岸甲基化调控人T细胞系中lag3表达. 中华微生物学和免疫学杂志, 2015,35(7):491-495.
[8]
Wang J, Sanmamed MF, Datar I, Su TT, Ji L, Sun J, Chen L, Chen Y, Zhu G, Yin W, Zheng L, Zhou T, Badri T, Yao S, Zhu S, Boto A, Sznol M, Melero I, Vignali DAA, Schalper K, Chen L, Fibrinogen-like protein 1 is a major immune inhibitory ligand of LAG-3. Cell, 2019, 176(1-2):334-347.e12.
Lymphocyte-activation gene 3 (LAG-3) is an immune inhibitory receptor, with major histocompatibility complex class II (MHC-II) as a canonical ligand. However, it remains controversial whether MHC-II is solely responsible for the inhibitory function of LAG-3. Here, we demonstrate that fibrinogen-like protein 1 (FGL1), a liver-secreted protein, is a major LAG-3 functional ligand independent from MHC-II. FGL1 inhibits antigen-specific T cell activation, and ablation of FGL1 in mice promotes T cell immunity. Blockade of the FGL1-LAG-3 interaction by monoclonal antibodies stimulates tumor immunity and is therapeutic against established mouse tumors in a receptor-ligand inter-dependent manner. FGL1 is highly produced by human cancer cells, and elevated FGL1 in the plasma of cancer patients is associated with a poor prognosis and resistance to anti-PD-1/B7-H1 therapy. Our findings reveal an immune evasion mechanism and have implications for the design of cancer immunotherapy.
[9]
韦腾飞, 张军, 吴豫, 张丹丹, 卢龙坤, 沈茜. CD4+ T细胞表面共抑制分子的表达水平与非小细胞肺癌疾病进展的关系 . 中华肿瘤杂志, 2014,36(6):424-429.
[10]
马成龙, 沈冬, 孙晓, 关乃富, 孙岳军, 戚春建. LAG-3在非小细胞肺癌细胞中的异位表达及其临床意义. 中国医刊, 2019,54(9):1005-1008.
[11]
黄维, 张庆娟, 刘柯, 徐茜, 周美英. 非小细胞肺癌中免疫检查点表达与临床特征及预后的关系分析. 癌症进展, 2019,17(5):571-574.
[12]
何兴桢, 杨安, 李曦. CD4+/CD8+比例和LAG3+细胞/CCR3+细胞在胃癌患者发病机制中的意义 . 中国医药导报, 2016 , 13(8):104-107.
[13]
江露, 吴昌平, 徐斌, 蒋敬庭. 胃癌患者血清中可溶性LAG-3分子的水平及意义. 临床检验杂志, 2014,32(1):38-40.
[14]
刘玉芳. 胃癌患者的血清LAG-3、DKK-1含量测定及其与临床病理特征的相关性. 海南医学院学报, 2017,23(7):1005-1008.
[15]
Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, Eppolito C, Qian F, Lele S, Shrikant P, Old LJ, Odunsi K. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer . Proc Natl Acad Sci U S A, 2010,107(17):7875-7880.
NY-ESO-1 is a
[16]
Hellmann MD, Rizvi NA, Goldman JW, Gettinger SN, Borghaei H, Brahmer JR, Ready NE, Gerber DE, Chow LQ, Juergens RA, Shepherd FA, Laurie SA, Geese WJ, Agrawal S, Young TC, Li X, Antonia SJ. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study. Lancet Oncol, 2017,18(1):31-41.
BACKGROUND: Nivolumab has shown improved survival in the treatment of advanced non-small-cell lung cancer (NSCLC) previously treated with chemotherapy. We assessed the safety and activity of combination nivolumab plus ipilimumab as first-line therapy for NSCLC. METHODS: The open-label, phase 1, multicohort study (CheckMate 012) cohorts reported here were enrolled at eight US academic centres. Eligible patients were aged 18 years or older with histologically or cytologically confirmed recurrent stage IIIb or stage IV, chemotherapy-naive NSCLC. Patients were randomly assigned (1:1:1) by an interactive voice response system to receive nivolumab 1 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks, nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 12 weeks, or nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks until disease progression, unacceptable toxicities, or withdrawal of consent. Data from the latter two cohorts, which were considered potentially suitable for further clinical development, are presented in this report; data from the other cohort (as well as several earlier cohorts) are described in the appendix. The primary outcome was safety and tolerability, assessed in all treated patients. This ongoing study is registered with ClinicalTrials.gov, number NCT01454102. FINDINGS: Between May 15, 2014, and March 25, 2015, 78 patients were randomly assigned to receive nivolumab every 2 weeks plus ipilimumab every 12 weeks (n=38) or nivolumab every 2 weeks plus ipilimumab every 6 weeks (n=40). One patient in the ipilimumab every-6-weeks cohort was excluded before treatment; therefore 77 patients actually received treatment (38 in the ipilimumab every-12-weeks cohort; 39 in the ipilimumab every-6-weeks cohort). At data cut-off on Jan 7, 2016, 29 (76%) patients in the ipilimumab every-12-weeks cohort and 32 (82%) in the ipilimumab every-6-weeks cohort had discontinued treatment. Grade 3-4 treatment-related adverse events occurred in 14 (37%) patients in the ipilimumab every-12-weeks cohort and 13 (33%) patients in the every-6-weeks cohort; the most commonly reported grade 3 or 4 treatment-related adverse events were increased lipase (three [8%] and no patients), pneumonitis (two [5%] and one [3%] patients), adrenal insufficiency (one [3%] and two [5%] patients), and colitis (one [3%] and two [5%] patients). Treatment-related serious adverse events were reported in 12 (32%) patients in the ipilimumab every-12-weeks cohort and 11 (28%) patients in the every-6-weeks cohort. Treatment-related adverse events (any grade) prompted treatment discontinuation in four (11%) patients in the every-12-weeks cohort and five (13%) patients in the every-6-weeks cohort. No treatment-related deaths occurred. Confirmed objective responses were achieved in 18 (47% [95% CI 31-64]) patients in the ipilimumab every-12-weeks cohort and 15 (38% [95% CI 23-55]) patients in the ipilimumab every-6-weeks cohort; median duration of response was not reached in either cohort, with median follow-up times of 12.8 months (IQR 9.3-15.5) in the ipilimumab every-12-weeks cohort and 11.8 months (6.7-15.9) in the ipilimumab every-6-weeks cohort. In patients with PD-L1 of 1% or greater, confirmed objective responses were achieved in 12 (57%) of 21 patients in the ipilimumab every-12-weeks cohort and 13 (57%) of 23 patients in the ipilimumab every-6-weeks cohort. INTERPRETATION: In NSCLC, first-line nivolumab plus ipilimumab had a tolerable safety profile and showed encouraging clinical activity characterised by a high response rate and durable response. To our knowledge, the results of this study are the first suggestion of improved benefit compared with anti-PD-1 monotherapy in patients with NSCLC, supporting further assessment of this combination in a phase 3 study. FUNDING: Bristol-Myers Squibb.
[17]
孙楠, 李源, 赫捷. 常见抑制性免疫检查点基因与食管鳞癌临床相关性分析. 中华医学杂志, 2018,98(21):1703-1706.
[18]
黄群欢, 洪岭, 陶春林, 黄旭红. 不同分期宫颈癌患者外周血Treg和相关细胞因子差异分析. 检验医学, 2017,32(9):769-772.
[19]
张丽萍, 林雪萍, 宋辉, 雷萍, 朱巧英. 免疫检查点分子在子宫内膜癌中的表达及临床意义. 肿瘤预防与治疗, 2019,32(6):486-493.
[20]
Que Y, Fang Z, Guan Y, Xiao W, Xu B, Zhao J, Chen H, Zhang X, Zeng M, Liang Y, Zhang X. LAG-3 expression on tumor-infiltrating T cells in soft tissue sarcoma correlates with poor survival. Cancer Biol Med, 2019,16(2):331-340.
[21]
魏艳利, 许莲蓉. 恶性血液病的肿瘤免疫逃逸. 新医学, 2019,50(8):570-574.
[22]
Goding SR, Wilson KA, Xie Y, Harris KM, Baxi A, Akpinarli A, Fulton A, Tamada K, Strome SE, Antony PA. Restoring immune function of tumor-specific CD4+ T cells during recurrence of melanoma . J Immunol, 2013,190(9):4899-4909.

版权

版权所有,未经授权,不得转载、摘编本刊文章,不得使用本刊的版式设计。
PDF(753 KB)

146333

Accesses

0

Citation

Detail

段落导航
相关文章

/