PDF(753 KB)
PDF(753 KB)
PDF(753 KB)
LAG-3分子的研究进展
Research progress on LAG-3 molecule
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。
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.
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 {{custom_keyword}} /
Lymphocyte activation gene-3 / T cell / Tumor / Immune checkpoint / Immunotherapy {{custom_keyword}} /
| [1] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [2] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [3] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [4] |
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.="">
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [8] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [15] |
NY-ESO-1 is a
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [16] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [20] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| [22] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
| {{custom_ref.label}} |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
PDF(753 KB)
/
| 〈 |
|
〉 |