Advances in Clinical Medicine
Vol. 13  No. 11 ( 2023 ), Article ID: 75150 , 7 pages
10.12677/ACM.2023.13112471

PD-L1在不同亚型三阴性乳腺癌中的表达及与患者预后的相关性分析

王晓玮1,孔滨2*

1青岛大学医学院,山东 青岛

2青岛大学附属医院乳腺外科,山东 青岛

收稿日期:2023年10月11日;录用日期:2023年11月6日;发布日期:2023年11月13日

摘要

目的:探讨分析细胞程序性死亡–配体1 (PD-L1)在不同亚型三阴性乳腺癌组织中的表达及与患者预后的相关性。方法:选择240名接受根治性切除前未行新辅助治疗的三阴性乳腺癌(TNBC)患者进行PD-L1表达的免疫组化结果分析,其中四种亚型的TNBC患者各60例。使用Kaplan-Meier、单变量和多变量Cox比例风险模型分析来比较不同亚型PD-L1表达与患者生存时间的关系。结果:中位随访时间为55.75个月(范围:23.28~120.02个月)。与PD-L1阴性的TNBC患者相比,PD-L1阳性的TNBC患者的总生存期(OS)明显增加(P = 0.029)。在不同分型的TNBC组织中PD-L1表达率最高的是免疫调节型(IM)为26.7%,伴随其较好的预后。而不同的临床分期(TNM分期)、组织学分级、有无淋巴结转移在不同分型的TNBC中PD-L1的阳性表达率存在显著差异性(P < 0.05),其中IM在四种亚型的TNBC患者中OS明显延长。单因素Cox比例模型分析表明,PD-L1是TNBC的独立预后因素。结论:PD-L1在不同分型的TNBC中对预后的影响不同,其中在IM分型中与预后的关系最密切。

关键词

乳腺癌,三阴性乳腺癌,预后,PD-L1,分子分型

The Expression of PD-L1 in Different Molecular Subtypes of Triple-Negative Breast Cancer and Its Correlation with Prognosis

Xiaowei Wang1, Bin Kong2*

1Medical College of Qingdao University, Qingdao Shandong

2Department of Breast Surgery, Affiliated Hospital of Qingdao University, Qingdao Shandong

Received: Oct. 11th, 2023; accepted: Nov. 6th, 2023; published: Nov. 13th, 2023

ABSTRACT

Objective: To investigate the expression of PD-L1 in different subtypes of triple-negative breast cancer and its correlation with the prognosis of patients. Methods: A total of 240 patients with triple-negative breast cancer (TNBC) who had not received neoadjuvant therapy before radical resection were selected for immunohistochemical study of PD-L1 expression, including 60 patients with four subtypes of TNBC. Kaplan-Meier and univariate Cox proportional hazard model analysis were used to compare the relationship between PD-L1 expression and survival time of patients with different subtypes. Results: The median follow-up time was 55.75 months (range: 23.28~120.02 months). Overall survival (OS) was significantly increased in PD-L1-positive TNBC patients compared with PD-L1-negative TNBC patients (P = 0.029). Among the different types of TNBC tissues, the highest expression rate of PD-L1 was 26.7% in immunomodulatory type (IM), which was accompanied by a better prognosis. There were significant differences in the positive expression rate of PD-L1 among different types of TNBC with different TNM stages, histological grades, and lymph node metastasis (P < 0.05), and OS was significantly prolonged in the four subtypes of TNBC patients with IM. Univariate Cox proportional model analysis showed that PD-L1 was an independent prognostic factor for TNBC. Conclusion: PD-L1 has different effects on prognosis in different types of TNBC, and the relationship between PD-L1 and prognosis is most close in IM type.

Keywords:Breast Cancer, Triple-Negative Breast Cancer, Prognosis, PD-L1, Molecular Subtype

Copyright © 2023 by author(s) and Hans Publishers Inc.

This work is licensed under the Creative Commons Attribution International License (CC BY 4.0).

http://creativecommons.org/licenses/by/4.0/

1. 介绍

乳腺癌是女性最常见的恶性肿瘤之一,其中三阴性乳腺癌(TNBC)占所有乳腺癌病例的15%~20%且恶性程度较高、易发生早期转移、患者生存期较短,需要积极治疗 [1] 。程序性死亡配体(PD-L1)是一种凋亡相关蛋白,是肿瘤微环境中的重要分子,其上调是肿瘤免疫逃避的关键机制之一。在T淋巴细胞与其受体PD-1结合后,PD-L1可以诱导抑制信号传递/导致T细胞凋亡,从而调节外周T细胞的免疫耐受性。PD-1/PD-L1通路的破坏和肿瘤免疫规避是目前癌症研究普遍关注的免疫治疗目标,使用药物阻断PD-1/ PD-L1通路将增强T细胞功能并导致肿瘤细胞溶解,从而开辟了新的癌症治疗途径 [2] [3] 。在这项研究中,我们调查了PD-L1在不同分型的TNBC中的表达,以更好地定义其预后意义。根据雄激素受体(AR)、CD8、FOXC1和DCLK1的表达将TNBC分为四种亚型:管腔雄激素受体型(LAR; AR+)、免疫调节型(IM; AR−, CD8+)、基底样免疫抑制型(BLIS; AR−, CD8−, FOXC1+)和间充质型(MES; AR−, CD8−, FOXC1−, DCLK1+) [4] [5] ,见图1。本研究通过分析程序性死亡配体1 (PD-L1)的表达在不同亚型三阴性乳腺癌(TNBC)中的表达及对其预后的影响,以期为三阴性乳腺癌的靶向治疗提供一定的参考依据。

Figure 1. According to Zhao et al.’s classification scheme based on immunohistochemistry (LAR, luminal androgen receptor type; IM, immunomodulatory; BLIS, basal-like immunosuppression; MES, mesenchymal type; UC, cannot be classified)

图1. 根据Zhao等人的基于免疫组织化学的分类方案(LAR,管腔雄激素受体型;IM,免疫调节型;BLIS,基底样免疫抑制型;MES,间充质型;UC,无法分类)

2. 资料与方法

2.1. 一般资料

搜集240名接受根治性切除前未行新辅助治疗的TNBC患者进行PD-L1表达的免疫组化结果分析,其中四种亚型的TNBC患者各60例。所选对象均为女性,平均年龄53.2岁(范围:32~83岁),根据年龄将患者分为两类,即<50岁和≥50岁以供进一步分析。病理上,大都数肿瘤为浸润性导管癌和组织学III级(80.5%)。在诊断上,大约一半的患者处于TNMII期(46.6%),其次是TNMIII期(27.2%)。所有患者手术前均未采取放化疗及其他抗肿瘤治疗,且临床病理资料完整,排除术后没有行规范辅助治疗者 [6] 。

2.2. 方法

使用R程序进行统计分析,分类变量通过观察次数和百分比来描述,连续变量表示为平均值 ± 标准差(SD),酌情使用Fisher精确检验或Pearson卡方检验计算临床病理特征与TNBC亚型之间的相关性 [7] 。总生存期(OS)定义为从诊断日期到死亡日期或最后一次随访日期的时间间隔,OS使用Kaplan-Meier分析确定。Cox比例风险回归模型用于确定变量的预后意义,选择基于IHC的TNBC亚型和单变量分析的显著变量进行多变量分析,p值 < 0.05被认为有统计学上的显著差异 [8] 。

3. 结果

3.1. PD-L1在不同分型的三阴性乳腺癌中的表达

在不同分型的TNBC组织中PD-L1表达率不同,其中在IM中表达率最高为26.7%。而不同的TNM分期、组织学分级、阳性淋巴结数在不同分型的TNBC中PD-L1的阳性表达率存在显著差异性(P < 0.05),其中LAR型PD-L1阳性表达率为21.7%、IM型为26.7%、BLIS型为23.3%、MES型为18.3%。为了确定PD-L1与不同分型TNBC预后之间的关系,我们分析了PD-L1表达和患者的总生存率 [9] [10] 。见表1图2

Table 1. Clinicopathological features of TNBC subtypes based on IHC

表1. 基于IHC的TNBC亚型的临床病理学特征

Figure 2. Survival analysis of triple-negative breast cancer patients with different IHC molecular subtypes based on PD-L1 expression (A, IM; B, LAR; C, BLIS; D, MES)

图2. 不同IHC分子亚型三阴性乳腺癌患者根据PD-L1表达进行生存分析(A,IM;B,LAR;C,BLIS;D,MES)

3.2. 基于IHC的TNBC亚型的预后意义

表2展示了OS的单变量和多变量Cox回归分析,临床分期和基于IHC的亚型是重要因素。具体而言,与IM相比,LAR (HR: 5.04, 95%CI: 1.45~17.4)、BLIS (HR: 4.72, 95%CI: 1.46~15.23)和MES (HR: 3.85, 95%CI: 0.97~14.86)在单变量分析中与较差的OS显著相关 [11] 。然而只有BLIS在多变量模型中仍然显著(HR: 3.29, 95%CI: 1.01~10.72)。

Table 2. Univariate and multivariate Cox regression analysis of OS

表2. OS的单变量和多变量Cox回归分析

4. 讨论

我们的研究结果显示,三阴性乳腺癌中的IM型的临床病理特征明显不同于其他亚型,相比于其他亚型,其预后明显较好。我们认为这与IM型PD-L1阳性率较高、其对于化疗的敏感性更高,化疗时激活或者增强机体原本存在的抗肿瘤免疫力的效果更明显有关 [12] [13] 。根据中国队列的分子分型,CD8是排名最高的基因且具有较高的ROC曲线下面积(AUC),mRNA和蛋白质表达之间具有良好的相关性。CD8是细胞毒性T细胞的标记物,是TIL和肿瘤免疫微环境的重要组成部分 [14] 。同时我们的研究发现,以间质CD8+TIL的存在为代表的IM亚型具有良好的预后,这一发现与其他研究一致。IM亚型已被证明具有较高的免疫通路相关基因的表达,如细胞因子信号传导、免疫细胞信号传导、抗原加工和呈递、趋化因子信号传导通路、免疫信号传导通路和免疫应答过程 [15] 。使用免疫检查点抑制剂(例如PD-1抑制剂)的免疫疗法已用于治疗TNBC [16] 。先前的研究表明,PD-L1表达与TNBC中高水平的CD8+T细胞浸润相关 [17] [18] 。因此,通过IHC评估TNBC尤其是IM亚型中PD-L1的表达对预测预后和指导治疗策略具有重要意义。

在我们的研究样本中,BLIS亚型常见于年轻女性,LAR和IM亚型多见于老年女性。有研究发现组织学3级最常见于BLIS亚型,该亚型的特点是细胞周期调节因子上调、DNA修复激活和免疫反应基因下调。而这些生物机制可以解释BLIS亚型的增值表型特征导致较差的生存结果 [19] [20] 。与同类型研究对比结果表明,当前的替代分子亚型还有进一步完善的空间,以提高临床相关性。

我们的研究有一些局限性。首先,病例数较少,可能在确定亚型与临床病理特征和生存结果的关联上存在误差。其次,众所周知,基因表达转录组数据是分子亚型分型的黄金标准,但是我们没有转录组数据验证结果。因此因谨慎解释本研究结果。

综上,PD-L1在IM型TNBC中的表达水平明显升高,同时会影响到患者预后。PD-L1阳性的IM型TNBC患者术后规范治疗的生存率明显升高。

致谢

行文至此落笔中,始于初秋终于夏。“桃李不言,下自成蹊。”首先感谢我的导师——孔滨老师。孔老师专业知识渊博、为学严谨认真、待人和蔼可亲,是我们的良师益友,人生之幸,得遇良师。“树高千尺不忘根深洪土。”感谢我的家人一直在用自己的方式全心全意的爱我、尊重并支持我成长路上的每一个选择,养育之恩,无以为报。感谢走的很慢但一直向前的自己,“追风赶月莫停留,平芜尽处是春山”,愿我们前路漫漫亦灿灿。

文章引用

王晓玮,孔 滨. PD-L1在不同亚型三阴性乳腺癌中的表达及与患者预后的相关性分析
The Expression of PD-L1 in Different Molecular Subtypes of Triple-Negative Breast Cancer and Its Correlation with Prognosis[J]. 临床医学进展, 2023, 13(11): 17625-17631. https://doi.org/10.12677/ACM.2023.13112471

参考文献

  1. 1. Azim, H.A., Shohdy, K.S., Elghazawy, H., et al. (2022) Programmed Death-Ligand 1 (PD-L1) Expression Predicts Re-sponse to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer: A Systematic Review and Meta-Analysis. Bi-omarkers, 27, 764-772. https://doi.org/10.1080/1354750X.2022.2112614

  2. 2. Ji, P., Gong, Y. and Hu, X. (2019) Role of PD-1/PD-L1 in Microenvironment of Breast Cancer. Chinese Journal of Oncology, 41, 401-405.

  3. 3. Yuan, K.Y., Wu, J.P., Zhao, Y.J., et al. (2022) Consistent Expression of PD-L1 in Tumor Microenvironment with Peripheral PD-1/PD-L1 in Circulating T Lymphocytes of Operable Breast Cancer: A Diagnostic Test. Diagnostic Pathology, 17, Article No. 68. https://doi.org/10.1186/s13000-022-01249-w

  4. 4. Oner, G., Önder, S., Karatay, H., et al. (2021) Clinical Impact of PD-L1 Expression in Triple-Negative Breast Cancer Patients with Residual Tumor Burden after Neo-adjuvant Chemotherapy. World Journal of Surgical Oncology, 19, Article No. 264. https://doi.org/10.1186/s12957-021-02361-9

  5. 5. Mori, H., Kubo, M., Yamaguchi, R., et al. (2017) The Combina-tion of PD-L1 Expression and Decreased Tumor-Infil- trating Lymphocytes Is Associated with a Poor Prognosis in Tri-ple-Negative Breast Cancer. Oncotarget, 8, 15584-15592. https://doi.org/10.18632/oncotarget.14698

  6. 6. Chu, J., Yeo, M.-K., Lee, S.H., et al. (2022) Clinicopathological and Prognostic Significance of Programmed Death Ligand-1 SP142 Expression in 132 Patients with Triple-Negative Breast Cancer. In Vivo, 36, 2890-2898. https://doi.org/10.21873/invivo.13030

  7. 7. Purwanto, I., Heriyanto, D.S., Ghozali, A., et al. (2020) Overexpres-sion of Programmed Death-Ligand 1 Receptor mRNA as an Independent Negative Prognostic Factor for Triple Negative Breast Cancer. World Journal of Oncology, 11, 216-222. https://doi.org/10.14740/wjon1302

  8. 8. Zhang, M.H., Sun, H.B., Zhao, S., et al. (2017) Expression of PD-L1 and Prognosis in Breast Cancer: A Meta-Analysis. Oncotarget, 8, 31347-31354. https://doi.org/10.18632/oncotarget.15532

  9. 9. Yoshikawa, K., Ishida, M., Yanai, H., et al. (2021) Prognostic Significance of PD-L1-Positive Cancer-Associated Fibroblasts in Patients with Triple-Negative Breast Cancer. BMC Cancer, 21, Article No. 239. https://doi.org/10.1186/s12885-021-07970-x

  10. 10. Yazaki, S., Shimoi, T., Yoshida, M., et al. (2023) Integrative Prognostic Analysis of Tumor-Infiltrating Lymphocytes, CD8, CD20, Programmed Cell Death-Ligand 1, and Tertiary Lymphoid Structures in Patients with Early-Stage Triple- Negative Breast Cancer Who Did Not Receive Adjuvant Chem-otherapy. Breast Cancer Research and Treatment, 197, 287-297. https://doi.org/10.1007/s10549-022-06787-x

  11. 11. Cerbelli, B., Pernazza, A., Botticelli, A., et al. (2017) PD-L1 Ex-pression in TNBC: A Predictive Biomarker of Responseto Neoadjuvant Chemotherapy? Biomed Research International, 2017, Article ID: 1750925. https://doi.org/10.1155/2017/1750925

  12. 12. Guo, L., Li, W.B., Zhu, X.X., et al. (2016) PD-L1 Expression and CD274 Gene Alteration in Triple-Negative Breast Cancer: Implication for Prognostic Biomarker. SpringerPlus, 5, Article No. 805. https://doi.org/10.1186/s40064-016-2513-x

  13. 13. Stovgaard, E.S., Bokharaey, M., List-Jensen, K., et al. (2020) PD-L1 Diagnostics in the Neoadjuvant Setting: Implications of Intratumoral Heterogeneity of PD-L1 Expression in Triple Negative Breast Cancer for Assessment in Small Biopsies. Breast Cancer Research and Treatment, 181, 553-560. https://doi.org/10.1007/s10549-020-05655-w

  14. 14. Tomioka, N., Azuma, M., Ikarashi, M., et al. (2018) The Ther-apeutic Candidate for Immune Checkpoint Inhibitors Elucidated by the Status of Tumor-Infiltrating Lymphocytes (TILs) and Programmed Death Ligand 1 (PD-L1) Expression in Triple Negative Breast Cancer (TNBC). Breast Cancer, 25, 34-42. https://doi.org/10.1007/s12282-017-0781-0

  15. 15. Wang, H., Ding, X.-H., Liu, C.-L., et al. (2023) Genomic Alterations Impacting Tumor-Infiltrating Lymphocytes and PD-L1 Expression Patterns in Triple-Negative Breast Cancer. JNCI-Journal of the National Cancer Institute, djad154. https://doi.org/10.1093/jnci/djad154

  16. 16. Pan, B.J., Xu, C., Ping, G.Q., et al. (2017) Correlation Analysis of PD-L1 Expression and Prognosis in Triple-Negative Breast Cancers. Chinese Journal of Pathology, 46, 822-826.

  17. 17. Dieci, M.V., Tsvetkova, V., Griguolo, G., et al. (2020) Integration of Tumour Infiltrating Lymphocytes, Programmed Cell-Death Ligand-1, CD8 and FOXP3 in Prognostic Models for Triple-Negative Breast Cancer: Analysis of 244 Stage I-III Patients Treated with Standard Therapy. European Journal of Cancer, 136, 7-15. https://doi.org/10.1016/j.ejca.2020.05.014

  18. 18. Suzuki, K., Ohe, R., Kabasawa, T., et al. (2023) Histological Spatial Analysis on the Induction of PD-L1+ Macrophages by CD8+ T Cells at the Marginal Microenvironment of Tri-ple-Negative Breast Cancer. Breast Cancer, 30, 1094-1104. https://doi.org/10.1007/s12282-023-01507-9

  19. 19. Lotfinejad, P., Kazemi, T., Mokhtarzadeh, A., et al. (2020) PD-1/PD-L1 Axis Importance and Tumor Microenvironment Immune Cells. Life Sciences, 259, Article ID: 118297. https://doi.org/10.1016/j.lfs.2020.118297

  20. 20. Zhang, M.L., Song, J., Yang, H.G., et al. (2022) Efficacy and Safety of PD-1/PD-L1 Inhibitors in Triple-Negative Breast Cancer: A Systematic Review and Meta-Analysis. Acta Oncologica, 61, 1105-1115. https://doi.org/10.1080/0284186X.2022.2106795

  21. NOTES

    *通讯作者Email: qkongbin@163.com

期刊菜单