王晰程①, 韦青①, 高静①, 李艳艳①, 周易②, 李洁①, 沈琳①. 转移性结直肠癌中应用二代测序技术预测EGFR单抗疗效的研究*[J]. 中国肿瘤临床, 2016, 43(4): 141-145. DOI: 10.3969/j.issn.1000-8179.2016.04.435
引用本文: 王晰程①, 韦青①, 高静①, 李艳艳①, 周易②, 李洁①, 沈琳①. 转移性结直肠癌中应用二代测序技术预测EGFR单抗疗效的研究*[J]. 中国肿瘤临床, 2016, 43(4): 141-145. DOI: 10.3969/j.issn.1000-8179.2016.04.435
Xicheng WANG1, Qing WEI1, Jing GAO1, Yanyan LI1, Yi ZHOU2, Jie LI1, Lin SHEN1. Using next-generation sequencing technology to evaluate biomarkers associated with anti-EGFR efficacy in metastatic colorectal cancer[J]. CHINESE JOURNAL OF CLINICAL ONCOLOGY, 2016, 43(4): 141-145. DOI: 10.3969/j.issn.1000-8179.2016.04.435
Citation: Xicheng WANG1, Qing WEI1, Jing GAO1, Yanyan LI1, Yi ZHOU2, Jie LI1, Lin SHEN1. Using next-generation sequencing technology to evaluate biomarkers associated with anti-EGFR efficacy in metastatic colorectal cancer[J]. CHINESE JOURNAL OF CLINICAL ONCOLOGY, 2016, 43(4): 141-145. DOI: 10.3969/j.issn.1000-8179.2016.04.435

转移性结直肠癌中应用二代测序技术预测EGFR单抗疗效的研究*

Using next-generation sequencing technology to evaluate biomarkers associated with anti-EGFR efficacy in metastatic colorectal cancer

  • 摘要: 目的:探索EGFR 单抗治疗转移性结直肠癌患者耐药性相关基因。方法:选取2012年3 月至2013年8 月在北京肿瘤医院接受EGFR 单抗治疗的31例转移性结直肠癌患者,利用二代测序技术检测肿瘤组织中与肿瘤发生、发展和EGFR 信号传导通路相关的21个基因,分析其与EGFR 单抗疗效的关系。结果:Kras 基因外显子2 为野生型的晚期结直肠癌的患者31例,接受EGFR 单抗单药三线治疗。全组患者中位PFS 为89d,OS为311 d,ORR 为16.1% 。5 例存在Kras 第3、4 外显子或者Nras 第2、3 外显子突变,均出现首次疗效评估PD(中位PFS 31d)。 其他突变包括1 例PIK3CA突变,PFS 为35d。1 例mTOR 突变,PFS 为91d。2 例患者存在SMAD 4 突变,PFS 分别是58d 及59d。1 例FBXW 7 突变,PFS 为93d。在26例全Ras基因野生型患者中MLL 3、TP53、APC 是突变频率最高的3 个基因。MLL 3 突变比例为92.3%(24/ 26),TP53为53.8%(14/ 26),APC 为42.3%(11/ 26)。 结论:进行EGFR 单抗治疗前必须进行Kras 及Nras 第2、3、4 外显子的检测,EGFR 通路下游信号分子,如PI 3KCA 、mTOR 等可能在EGFR 单抗疗效上起一定预测作用,针对EGFR 外的其他信号通路分子如SMAD 4、FBXW 7 等,需要进一步关注。

     

    Abstract: Objective:To explore genes associated with sensitivity to anti-EGFR therapy.Methods:From March2012to August 2013, 31metastatic colorectal cancer patients in Peking University Cancer Hospital & Institute were treated with anti-EGFR mono-therapy. A total of 21genes associated with oncogenesis, metastasis, and EGFR signaling pathway were profiled in these31patients by using tar-geted next-generation sequencing technology. Results:A total of 31patients with Kras exon 2 wild type received anti-EGFR therapy as third-line treatment. Among these patients, the median progression-free survival (PFS) was 89days, overall survival was311 days, and objective response rate was 16. 1%. Five cases harbored Kras exons 3/4 or Nras exons 2/3 mutations. These five Ras mutation patients showed disease progression during the first evaluation with 31-day PFS. One PIK3CA mutation case exhibited disease progression dur-ing the first evaluation (PFS 35days), and one case showed mTOR mutation with91-day PFS. The PFS of two cases with SMAD 4 muta-tion were 58and 59days, whereas that of the case containing FBXW 7 mutation was 93days. Among the 26Ras wild- type patients, MLL3, TP 53, and APC were the three genes with the highest mutation frequencies of 92. 3% (24/26), 53. 8% (14/26), and 42. 3% (11/ 26), respectively. Conclusion: Extended Ras analysis (including Kras and Nras exons 2/3/4) is recommended for patients who are candi-dates for anti-EGFR therapy. Mutations in the downstream effectors of the EGFR signaling pathway, such as PI 3KCA and mTOR, may al-so have a predictive role in anti- EGFR therapy. Mutations beyond the EGFR pathway such as FBXW7 and SMAD4 may be associated with anti-EGFR efficacy and deserve further attention.

     

/

返回文章
返回