NRAS突变型晚期黑色素瘤的治疗进展

赵博伦, 朱冠男

  1. 空军军医大学西京医院皮肤科,陕西 西安 710032
  • 收稿日期:2023-05-16 修回日期:2023-10-01 出版日期:2023-10-30 发布日期:2023-10-31
  • 通信作者: 朱冠男
  • 作者简介:赵博伦(ORCID: 0000-0003-0326-459X),硕士研究生在读。
  • 基金资助:
    国家自然科学基金(82172607);国家自然科学基金(81702714)

摘要/Abstract

摘要:

黑色素瘤的发生、发展与多种癌基因的激活密切相关。15%~20%的黑色素瘤患者存在NRAS基因的激活突变,携带该突变基因的黑色素瘤具有更强的侵袭性,治疗难度大。由于RAS蛋白突变位点属于弱药物靶标,目前尚缺乏有效的靶向抑制剂,因此临床上多以免疫检查点抑制剂作为NRAS突变型晚期黑色素瘤的一线治疗方案,然而治疗反应率较低。近年来,在NRAS突变亚型中靶向治疗方案的探索主要集中在NRAS下游丝裂原活化蛋白激酶(mitogen-activated protein kinase,MAPK)通路,但结果不一:新型MEK1/2抑制剂tunlametinib,在晚期NRAS突变患者中总体客观缓解率(objective response rate,ORR)达到34.7%,较既往的binimetinib显著提高;然而泛RAF抑制剂belvarafenib和ERK抑制剂ulixertinib的Ⅰ期临床试验却未能展示出该药明显的优势。此外,以MEK抑制剂为基础的联合治疗也取得一定进展,现有证据表明,分子抑制剂类药物较免疫检查点抑制剂显示出更多的优势:选择性BRAF/CRAF抑制剂naporafenib(LXH254)与MEK抑制剂trametinib联合治疗NRAS突变型黑色素瘤的Ⅰb期临床试验中ORR达到46.7%;细胞周期蛋白依赖性激酶4/6(cyclin-dependent kinase4/6,CDK4/6)抑制剂ribociclib和binimetinib联合治疗在携带NRAS突变同时合并细胞周期蛋白基因异常的人群中可达32.5%;黏着斑激酶(focal adhesion kinase,FAK)抑制剂IN10018联合cobimetinib的研究结果也表现出较好的ORR(38.5%);但免疫检查点抑制剂程序性死亡[蛋白]配体-1(programmed death ligand-1,PD-L1)单抗durvalumab联合trametinib方案仅使27.2%的患者达到部分缓解(3/11)。与此同时,部分临床前研究结果也显示出一些转化潜质:如热激蛋白90(heat shock protein 90,HSP90)抑制剂XL888和丝氨酸/苏氨酸蛋白激酶19(serine/threonine protein kinase 19,STK19)抑制剂均在动物模型中表现出显著抑制NRAS突变型黑色素瘤细胞生长的能力。本文综述了NRAS突变型黑色素瘤的致癌机制及近年来治疗领域的研究进展,旨在展示该亚型患者的治疗现状,对多种新型治疗方法的临床研究结果进行总结和归纳,为当前临床实践和未来联合治疗方案提供依据。

关键词: NRAS基因突变, 黑色素瘤, 致癌机制, 靶向治疗

Abstract:

Oncogenic mutations are responsible for a majority of the malignancy of melanoma. Activating mutations of NRAS gene are found in 15%-20% melanoma cases, endowing the tumor cells with more aggressive phenotypes and greater difficulty to treat. The development of targeted inhibitor of mutant NRAS remains a big challenge since the mutation sites could hardly be druggable. Therefore, immune checkpoint inhibitors are currently recommended as the first-line therapy for NRAS mutant advanced melanoma albeit the response rate is still far from satisfaction. In recent years, the exploration of targeted therapy regimens has focused on the downstream pathway of NRAS, the mitogen-activated protein kinase (MAPK) pathway. A novel MEK1/2 inhibitor tunlametinib was reported to achieve an objective response rate (ORR) of 34.7% which is higher than the ORR of binimetinib in previous report. However, the phase Ⅰ trial of the pan-RAF inhibitor belvarafenib and the ERK inhibitor ulixertinib failed to show marked benefits. In the meanwhile, MEK inhibitor-based combination therapy has also achieved some progress: it was reported in the phase Ⅰb trial of the selective BRAF/CRAF inhibitor naporafenib (LXH254) combined with Trametinib in NRAS mutant melanoma that the ORR was 46.7%. The ORR of binetinib plus the cyclin-dependent kinase 4/6 (CDK4/6) inhibitor, ribociclib, was 32.5% in patients with NRAS mutation with concurrent alterations of CDKN2A, CDK4, or CCND1. The response rate of the combination of focal adhesion kinase (FAK) inhibitor, IN10018, and cobimetinib was 38.5%. On the other hand, only 27.2% of patients carrying NRAS mutation responded partially to the combined regimen of immune checkpoint inhibitor programmed death ligand-1 (PD-L1) monoclonal antibody durvalumab+trametinib. In addition, some preclinical findings have also shown translational potentials: for example, heat shock protein 90 (HSP90) inhibitor XL888 and serine/threonine protein kinase 19 (STK19) inhibitors were found to inhibit the growth of NRAS mutant melanoma in animal models. This article reviewed the oncogenic roles of NRAS mutation in melanoma and the cutting-edge clinical trials for the treatment of NRAS mutant melanoma, aiming to provide alterative treatment options for clinical practice and inspire novel combination regimen.

Key words: NRAS mutation, Melanoma, Carcinogenesis, Targeted therapy

中图分类号: 

相关文章

[1] 康殷楠, 陈顺, 解有成, 郑英, 何昱静, 李初谊, 于晓辉. 抗体药物偶联物在HER2阳性晚期胃癌中的应用进展和展望[J]. 中国癌症杂志, 2023, 33(8): 790-800.
[2] 吴晶, 周娟, 苏春霞. 肺癌脂肪酸代谢重编程的研究进展[J]. 中国癌症杂志, 2023, 33(5): 517-526.
[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] 杨梓怡, 李盼丽, 顾丙新, 刘成, 宋少莉, 许晓平. 68Ga-DOTA-PDL1P的设计合成及其在黑色素瘤小鼠模型中的应用研究[J]. 中国癌症杂志, 2023, 33(4): 354-360.
[5] 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.
[6] 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.
[7] 曹晓珊, 丛斌斌. 三阳性乳腺癌内分泌治疗联合靶向治疗的研究进展[J]. 中国癌症杂志, 2023, 33(3): 288-292.
[8] 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.
[9] 郭晴, 张剑. HER2低表达乳腺癌的靶向治疗研究进展[J]. 中国癌症杂志, 2023, 33(2): 181-190.
[10] 邹淳缘, 许晓峰, 卢仁泉, 郭林. 肺癌组织和外周血中p53、PGP9.5、SOX2、GAGE7、GBU4-5和MAGE A1蛋白水平检测及其临床价值探讨[J]. 中国癌症杂志, 2023, 33(1): 36-44.
[11] 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.
[12] 李小秋, CD30阳性淋巴瘤病理专家组. CD30在淋巴瘤中的表达及检测:现状与挑战[J]. 中国癌症杂志, 2022, 32(6): 512-518.
[13] 江健韵, 应红梅. BRAF V600突变阳性的晚期黑色素瘤治疗的临床研究进展[J]. 中国癌症杂志, 2022, 32(5): 445-450.
[14] 王子茂, 曹原, 王琪影. 皮肤梭形细胞黑色素瘤患者生存预测模型的构建及验证[J]. 中国癌症杂志, 2022, 32(3): 234-242.
[15] 陈光亮, 吴方恬, 曹军宁. 非特指EBV阳性弥漫大B细胞淋巴瘤的研究进展[J]. 中国癌症杂志, 2022, 32(3): 258-267.