联合检测LDHA和PD-L1在晚期胃癌PD-1抑制剂疗效预测及预后评估中的价值

左学良, 陈志强, 董润雨, 王智雄, 蔡娟

  1. 1.皖南医学院第一附属医院(弋矶山医院)胃肠外科,安徽 芜湖 241001
    2.重大疾病非编码RNA转化研究安徽普通高校重点实验室,安徽 芜湖 241001
    3.南京医科大学第一附属医院肝胆中心,江苏 南京 210029
    4.皖南医学院第一附属医院(弋矶山医院)肿瘤内科,安徽 芜湖 241001
  • 收稿日期:2022-10-26 修回日期:2023-03-02 出版日期:2023-05-30 发布日期:2023-06-16
  • 通信作者: 蔡娟
  • 作者简介:左学良(ORCID: 0000-0001-5204-2585),博士,副主任医师。
  • 基金资助:
    国家自然科学基金(82103293);安徽省高校优秀青年人才支持计划(gxyq2021257);芜湖市科技计划(2022jc52)

摘要/Abstract

摘要:

背景与目的:胃癌对程序性死亡[蛋白]-1(programmed death-1,PD-1)抑制剂的应答率较低,建立有用的疗效预测方法筛选胃癌PD-1抑制剂治疗优势人群对改善患者预后有重要意义。本研究旨在探讨联合检测乳酸脱氢酶A(lactate dehydrogenase,LDHA)和程序性死亡[蛋白]配体-1(programmed death ligand-1,PD-L1)表达对接受PD-1抑制剂治疗胃癌患者的疗效预测和预后评估价值。方法:回顾性分析皖南医学院第一附属医院2020年1月—2022年3月接受PD-1抑制剂治疗的50例晚期胃癌患者的临床病理学资料,采用多因素logistic回归分析影响胃癌PD-1抑制剂疗效的独立危险因素,利用受试者工作特征(receiver operating characteristic,ROC)曲线分析联合检测LDHA和PD-L1对胃癌PD-1抑制剂疗效的预测价值,应用Kaplan-Meier法对患者进行生存分析。结果:LDHA低表达组和高表达组胃癌患者的客观缓解率(objective response rate,ORR)分别为59%和10%,疾病控制率(disease control rate,DCR)分别为83%和29%,差异有统计学意义(P<0.001)。多因素logistic回归分析结果显示,PD-L1阳性联合分数(combined positive score,CPS)<5、LDHA高表达是胃癌PD-1抑制剂疗效不佳的独立危险因素(P<0.05)。ROC曲线分析结果提示LDHA联合PD-L1具有较好的胃癌PD-1抑制剂疗效预测价值[曲线下面积(area under curve,AUC)为0.951]。Kaplan-Meier生存分析显示,LDHA低表达合并PD-L1 CPS≥5的胃癌患者在接受PD-1抑制剂治疗后具有更长的总生存期(overall survival,OS,P=0.003)和无进展生存期(progression-free survival,PFS,P<0.001)。结论:LDHA低表达、PD-L1 CPS≥5与胃癌PD-1抑制剂的疗效呈正相关,LDHA低表达合并PD-L1 CPS≥5的胃癌患者接受PD-1抑制剂治疗可显著延长OS和PFS,联合检测LDHA和PD-L1对晚期胃癌患者PD-1抑制剂的疗效预测和预后评估有一定的临床应用价值。

关键词: 胃肿瘤, 免疫检查点抑制剂, 免疫治疗, 疗效, 预后

Abstract:

Background and purpose: The response rate of gastric cancer patients to programmed death-1 (PD-1) inhibitor is relatively low. Establishing a useful efficacy prediction method to screen the superior gastric cancer patients receiving anti-PD-1 therapy could improve the prognosis of patients. This study aimed to explore the value of combined detection of lactate dehydrogenase (LDHA) and programmed death ligand-1 (PD-L1) expressions in predicting the efficacy and prognosis of gastric cancer patients treated with PD-1 inhibitor. Methods: The clinicopathological data of 50 advanced gastric cancer patients treated with PD-1 inhibitor in The First Affiliated Hospital of Wannan Medical College from January 2020 to March 2022 were retrospectively analyzed. The independent risk factors affecting the efficacy of PD-1 inhibitor were analyzed by multivariate logistic regression. The value of combined detection of LDHA and PD-L1 in predicting the efficacy of PD-1 inhibitors in gastric cancer was analyzed by receiver operating characteristic (ROC) curve analysis. Gastric cancer patient survival was analyzed by Kaplan-Meier method. Results: The objective response rate (ORR) of gastric cancer patients receiving PD-1 inhibitor therapy in LDHA low and high expression groups were 59% and 10%, respectively. The disease control rate (DCR) in LDHA low and high expression groups were 83% and 29%, respectively. The difference was statistically significant (P<0.001). Multivariate logistic regression analysis showed that PD-L1 combined positive score (CPS)<5 and LDHA high expression were independent risk factors affecting the efficacy of PD-1 inhibitor in gastric cancer (P<0.05). ROC curve analysis showed that combined detection of LDHA and PD-L1 had good predictive value for the efficacy of PD-1 inhibitor in gastric cancer [area under curve (AUC) was 0.951]. Kaplan-Meier survival analysis showed that gastric cancer patients with low LDHA expression and PD-L1 CPS≥5 had longer overall survival (OS, P=0.003) and progression-free survival (PFS, P<0.001) after receiving PD-1 inhibitor therapy. Conclusion: Low LDHA expression and PD-L1 CPS≥5 were positively correlated with the efficacy of PD-1 inhibitor in gastric cancer. Gastric cancer patients with low LDHA expression and PD-L1 CPS≥5 significantly had prolonged OS and PFS after receiving PD-1 therapy. Therefore, the combined detection of LDHA and PD-L1 expressions has good value in predicting the efficacy and evaluating prognosis of advanced gastric cancer patients treated with PD-1 inhibitor.

Key words: Stomach neoplasms, Immune checkpoint inhibitors, Immunotherapy, Efficacy, Prognosis

中图分类号: 

相关文章

[1] 杨闻箫, 国琳玮, 凌泓, 胡欣. 基于免疫微环境特征的曲妥珠单抗与免疫治疗联合应用预测模型[J]. 中国癌症杂志, 2023, 33(5): 484-498.
[2] 中国临床肿瘤学会免疫治疗专家委员会, 上海市抗癌协会肿瘤生物治疗专业委员会. 基因重组溶瘤腺病毒治疗恶性肿瘤临床应用中国专家共识(2022年版)[J]. 中国癌症杂志, 2023, 33(5): 527-548.
[3] 蒋金玲, 周尘飞, 王超, 赵丽琴, 吴珺玮, 张俊. 2022年度胃癌研究和诊疗新进展[J]. 中国癌症杂志, 2023, 33(4): 303-314.
[4] 田熙, 徐文浩, 朱殊璇, 艾合太木江·安外尔, 宿佳琦, 叶世琪, 瞿元元, 施国海, 张海梁, 叶定伟. 2022年度肾细胞癌基础研究及临床诊疗新进展[J]. 中国癌症杂志, 2023, 33(3): 191-200.
[5] 苏春霞, 周彩存. 2022年度肺癌领域重要临床研究进展[J]. 中国癌症杂志, 2023, 33(3): 218-227.
[6] 邵志博, 杨犇龙, 吴炅. 2022年中国乳腺癌重要临床试验成果及最新进展[J]. 中国癌症杂志, 2023, 33(2): 103-109.
[7] 陈玉光, 孙晓, 毕钊, 邱鹏飞, 段宝伟, 樊庆达, 王永胜. 乳腺癌内乳前哨淋巴结活检评估预后及指导个体化内乳照射:一项长期随访研究[J]. 中国癌症杂志, 2023, 33(2): 142-151.
[8] 陈瑛瑶, 储香玲, 俞昕, 苏春霞. 免疫检查点抑制剂疗效相关预测模型的研究进展[J]. 中国癌症杂志, 2023, 33(1): 61-70.
[9] 高鹤丽, 徐近, 虞先濬. 2021年胰腺神经内分泌肿瘤诊治的新进展[J]. 中国癌症杂志, 2022, 32(9): 772-778.
[10] 禹航, 刘文生, 张宁, 刘海宽, 陈洛海, 姚望, 范文哲, 李家平, 陈洁, 王于. 肝动脉栓塞术治疗囊性神经内分泌肿瘤肝转移的疗效及安全性分析[J]. 中国癌症杂志, 2022, 32(9): 794-799.
[11] 张日虹, 郭宇, 梁贇, 陈洛海, 陈洁, 王玮. 辅助化疗对胃神经内分泌癌患者预后的意义[J]. 中国癌症杂志, 2022, 32(9): 807-817.
[12] 肖玉铃, 朱秀之, 江一舟, 邵志敏. 三阴性乳腺癌精准治疗研究的新进展与未来展望[J]. 中国癌症杂志, 2022, 32(8): 669-679.
[13] 周文斌, 谢晖, 丁强, 王水. 在乳腺癌局部精准治疗中的新进展:联合免疫治疗未来可期[J]. 中国癌症杂志, 2022, 32(8): 698-704.
[14] 许宇辰, 程蕾蕾, 王妍, 林瑾仪, 陈佳慧, 陈怡帆, 周宇红, 刘天舒, 葛均波. sST2在免疫检查点抑制剂相关不良反应风险预测中的价值[J]. 中国癌症杂志, 2022, 32(8): 712-718.
[15] 曹孟轩, 胡灿, 张延强, 黄灵, 杜义安, 于鹏程, 章若岚, 徐志远, 程向东. 胃癌根治术后早期复发转移的危险因素分析[J]. 中国癌症杂志, 2022, 32(7): 588-595.