医师报

— 医师报官方网站 —

医师报

— 医师报官方网站 —

肿瘤科
当前位置:首页/ 专科/肿瘤科/详情

进展&亮点|张兰教授:不断探索潜在生物标志物,结直肠癌患者免疫治疗前景更光明

时间:2023-10-31 11:29:25来源:医师报作者:chengshihan 阅读: 164143

结直肠癌(CRC)是消化系统常见恶性肿瘤之一,发病率和死亡率分别位居所有恶性肿瘤的第三位和第二位[1]。转移性结直肠癌(mCRC)患者的总体生存较差,临床需更有效的治疗方法[2-4]。免疫检查点抑制剂(ICI)治疗方案从晚期后线治疗逐渐前移至一线治疗,程序性死亡蛋白-1(PD-1)抑制剂对具有错配修复缺陷(dMMR)/微卫星不稳定(MSI-H)mCRC患者治疗效果良好[5-8]。《医师报》特别邀请河北医科大学第四医院消化内科副主任张兰教授分享了ICI在mCRC中的疗效及研究现状。

01

dMMR/MSI-H的mCRC患者免疫治疗效果良好

目前CRC免疫治疗的相关研究主要集中于免疫检查点抑制剂(PD-1/PD-L1、CTLA-4)。PD-1/PD-L1抑制剂可抑制PD-1信号通路,阻断肿瘤免疫抑制性信号传递,杀伤肿瘤细胞[9],而dMMR/MSI-H分子表型的mCRC患者可从PD-1抑制剂帕博利珠单抗中显著获益[5]。Ⅱ期临床研究KEYNOTE-016结果显示,经标准治疗失败的MSI-H/dMMR mCRC患者接受帕博利珠单抗治疗后,客观缓解率(ORR)可达40%,20周无进展生存(PFS)率达78%[6]。

KEYNOTE-177 是全球首个头对头比较帕博利珠单抗和标准治疗(FOLFOX或FOLFIRI化疗±靶向贝伐珠单抗或西妥昔单抗)一线治疗MSI-H/dMMR mCRC患者疗效和安全性的Ⅲ期临床研究,结果显示帕博利珠单抗组的ORR、PFS(图1)均显著优于标准治疗组,而且3-5级不良事件发生率远低于标准治疗组[7]。

帕博利珠单抗组中位OS 49.2个月,标准治疗组中位OS为36.7个月,尽管数值对比差异较大(HR,0.74;95%CI,0.53-1.03;P=0.036),但因未达到预定的统计分析边界,未取得统计学阳性结果,这可能与超过60%的标准治疗组患者一线进展后接受了免疫检查点抑制剂的交叉治疗有关。虽然研究者未能最终认定帕博利珠单抗在OS获益上优于标准治疗,但目前的结果支持帕博利珠单抗作为MSI-H/dMMR转移性结直肠癌患者的一线治疗。

为了扩大ICI治疗获益人群,继KEYNOTE-177研究之后很多研究在不断探索。Ⅱ期的GERCOR NIPICOL研究[10]评估了纳武利尤单抗+伊匹单抗治疗经标准治疗失败的MSI-H/dMMR mCRC 的疗效。受试者接受纳武利尤单抗3mg/kg+伊匹单抗1mg/kg Q3W治疗4个周期,然后纳武利尤单抗3mg/kg Q2W,直至疾病进展或最多20个周期,24个月PFS率为92.9%,这显示出纳武利尤单抗+伊匹单抗治疗对于化疗耐药MSI-H/dMMR mCRC仍显示出持久的抗肿瘤活性。

此外,为了探索更优的MSI-H/dMMR mCRC一线治疗模式,很多临床研究正在进行中,例如SWOG-S1610研究是比较atezolizumab单药与atezolizumab联合标准治疗(FOLFOX6+贝伐珠单抗)在此人群一线治疗效果的随机Ⅲ期研究。CHECKMATE 8HW同样是研究评估纳武利尤单抗(NIVO)单药或纳武利尤单抗和伊匹木单抗(NIVO+IPI)联合治疗MSI-H/dMMR mCRC的疗效或安全性。

相信MSI-H/dMMR mCRC的免疫治疗之路会更加光明。

02

ICI疗效预测的需求,筛选潜在的标志物势在必行

在Ⅳ期CRC总体人群中,dMMR/MSI-H人群占比极低,仅把dMMR/MSI-H作为预测ICI疗效的标志物远不能满足临床治疗需求,当前正开展多项研究以探索预测ICI疗效的潜在标志物。

肿瘤突变负荷(TMB)是指肿瘤组织每兆碱基中出现的体细胞突变数目,TMB越高,ICI治疗可能越有效[11]。KEYNOTE-158研究以TMB≥10 mut/Mb作为界值划分TMB-H和非TMB-H。结果显示,TMB-H组ORR显著高于非TMB-H组,可见TMB检测可筛选出MSS/TMB-H的mCRC人群[12-13]。然而目前TMB的检测成本较高,临床应用尚存局限[14]。

此外,肿瘤微环境中PD-L1上调与效应T细胞浸润增加相关,但目前在CRC中并无预测价值[15-17]。

POLE/POLD1基因突变在CRC中分别达到6.9%和7.36%[18-19],研究显示,POLE/POLD1基因突变与TMB-H相关,携带POLE/POLD1突变的患者接受ICI治疗可达更高的ORR[20-23]。目前单独应用POLE/POLD1突变预测ICI疗效尚缺乏前瞻性大规模临床研究证据。此外,CRC组织免疫组化检测结果显示高免疫评分与更高生存率显著相关[24],其能否成为预测ICI疗效的标志物值得进⼀步探索。

干扰素γ(IFN-γ)是肿瘤和宿主细胞中PD-L1表达的关键驱动因素,可预测ICI的疗效[25]。汇总研究证实,IFN-γ相关的基因表达谱(GEP)可作为ICI疗效的预测指标,且炎症性T细胞GEP联合TMB对ICI疗效预测价值更高[26]。但目前mRNA检测平台在国内的可及性较低,其针对结直肠癌预测的作用仍需大样本队列进一步验证。此外,临床研究提示特定微⽣物群(如脆弱类杆菌、双歧杆菌、厚壁杆菌等)可能与ICI疗效相关[27],肠道菌群丰富的患者表现出更优PFS[28],但相关作用机制尚不明确。

综上所述,CRC的异质性决定了应用单一生物标志物预测临床疗效仍存不足,未来联合多种生物标志物可更好筛选免疫治疗获益人群。

03

联合方案改变“冷肿瘤”状态,提高免疫治疗效果

大量临床研究证实ICI单药应用于pMMR/MSS mCRC疗效甚微,针对此类患者,需寻找克服原发免疫耐药的手段以提高ICI治疗效果。目前相关的临床研究包括化疗、放疗、靶向及CTLA-4联合PD-1/PD-L1等。

化疗联合PD-1/PD-L1方案一线治疗进展期CRC的Ⅱ期临床研究的ORR达53%(图1),8周时的疾病控制率(DCR)高达100%[29]。系列研究发现化疗联合PD-1/PD-L1治疗方案在pMMR/MSS mCRC患者中均表现出抗肿瘤活性[30-31],但其一线治疗地位仍需大规模的人群验证。

放疗联合PD-1/PD-L1研究发现[32-33],放疗杀伤肿瘤细胞后导致大量肿瘤抗原释放,通过释放组织因子促进CD8+T细胞富集,上调肿瘤细胞的PD-1表达以增强ICI疗效。放化疗序贯ICI方案的病理完全缓解(PCR)率可达30%,呈现出较优疗效[34]。

靶向联合PD-1/PD-L1的REGONIVO研究显示,中位PFS为7.9月,一年OS率达68.0%[35](图1),但在我国开展的PD-1联合TKI研究结果并不理想,因此靶向联合ICI尚待进一步证据支持。EGFR联合PD-L1抑制剂的研究结果证实二者具有协同抗肿瘤作用,其治疗MSS mCRC患者的临床研究正在进行中。此外,TGF-β细胞蛋白、炎症性环氧合酶、巨噬细胞浸润等多种方案也已投入研究,或能为肿瘤患者带来更高的生存获益。


图1靶向联合ICI治疗结直肠癌与胃癌OS情况

04

张兰教授总结

当前,ICI对具有dMMR/MSI-H的mCRC患者表现出长生存、高ORR、持久应答及低毒性等诸多优势。为满足临床需要,一方面可寻找潜在的ICI疗效预测生物标志物,扩大ICI应用范围;另一方面,对pMMR/MSS mCRC患者需考虑多种治疗手段与免疫治疗方法的联合,提高ICI疗效,未来仍需进一步探索。

专家简介


张兰 教授

河北医科大学第四医院

河北医科大学第四医院消化内科 副主任

副主任医师 硕士研究生导师

中国抗癌协会中西整合神经内分泌肿瘤专委会委员

北京癌症防治学会胃癌防治专委会委员

北京癌症防治学会食管癌防治专委会青委会委员

河北省临床肿瘤学会神经内分泌肿瘤专家委员会常委兼秘书

河北省抗癌协会老年肿瘤专委会委员兼秘书

河北省抗癌协会神经内分泌肿瘤及胃肠间质瘤专委会委员

河北省女医师协会消化专委会委员

河北省预防医学会消化疾病防控专委会委员

河北省预防医学会结直肠癌防治专委会委员

河北省急救医学会第一届胃肠外科专业委员会委员

曾荣获河北省科技进步二等奖1项,三等奖1项

参考文献:(滑动查看)

[1] Ferlay J, Ervik M, Lam F, et al. Global cancer observatory: Cancer today [EB/OL]. Lyon, France: International Agency for Research on Cancer. [2023-10-18]. https://gco.iarc.fr/today.

[2] Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol. 2016;27:1386–1422.

[3] Venook AP, Niedzwiecki D, Lenz H-J, et al. Effect of first-line chemotherapy combined with cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal cancer: A randomized clinical trial. JAMA. 2017;317:2392–2401.

[4] Vincenzi B, Cremolini C, Sartore-Bianchi A, et al. Prognostic significance of K-Ras mutation rate in metastatic colorectal cancer patients. Oncotarget. 2015;6(31):31604-12.

[5] Le DT, Uram JN, Wang H,et al . PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015; 372:2509–2520.

[6]Le DT, Uram JN, Wang H, et al. Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade[J]. Science,2017, 357( 6349) : 409-413.

[7] André T,et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer[J].N Engl J Med, 2020 ,383(23):2207-2218.

[8] Thierry Andre, First-line therapy of pembrolizumab versus standard of care (SOC) in microsatellite instability-high/mismatch repair deficient metastatic colorectal cancer: The phase III, KEYNOTE-177 study. ASCO 2020, LBA4.

[9] Diaz Jr L A, Shiu K K, Kim T W, et al. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): Final analysis of a randomised, open-label, phase 3 study[J]. The Lancet Oncology, 2022, 23(5): 659-670.

[10] Cohen, Romain; Meurisse, Aurelia; Pudlarz, Thomas,et al. One-year duration of nivolumab plus ipilimumab in patients (pts) with microsatellite instability-high/mismatch repair-deficient (MSI/dMMR) metastatic colorectal cancer (mCRC): Long-term follow-up of the GERCOR NIPICOL phase II study. J CLIN ONCOL. 2022-02-01;40(4_suppl):13-13.

[11]McGranahan N,Furness A J,Rosenthal R,et a1.Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade[J].Science,2016,351(6280):1463-1469.

[12]Marabelle A, Fakih M, Lopez J, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020; 21: 1353–1365.

[13]Fabrizio DA,George TJ Jr,Dunne R F,et a1.Beyond microsatellite testing:assessment of tumor mutational burden identifies subsets of eolorectal cancer who may respond to immune checkpoint inhibition[J].J Gastmintest Oncol,2018,9(4):610-617.

[14]McGrail DJ, Pilie PG, Rashid NU, etal. High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann Oncol 2021; 32: 661–672.

[15] O'Neil BH, Wallmark J, Lorente D, et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma.PLoS ONE,2017, 12(12):e0189848.

[16]Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency[J]. N Engl J Med, 2015, 372(26):2509-2520.

[17]Overman MJ, Lonardi S, Wong KYM, et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer[J]. J Clin Oncol, 2018, 36(8):773-779.

[18]Kothari N, Teer JK, Abbott AM, et al. Increased incidence of FBXW7 and POLE proofreading domain mutations in young adult colorectal cancers[J]. Cancer, 2016, 122(18):2828-2835.

[19]Wang F, Zhao Q, Wang YN, et al. Evaluation of POLE and POLD1 Mutations as Biomarkers for Immunotherapy Outcomes Across Multiple Cancer Types[J]. JAMA Oncol, 2019, 5(10):1504-1506.

[20]Song, Z. et al. Clinicopathological characteristics of POLE mutation in patients with non-small-cell lung cancer. Lung cancer,2018,118:57–61.

[21]Mehnert, J. M. et al. Immune activation and response to pembrolizumab in POLE-mutant endometrial cancer. Te Journal of clinical Investigation,2016,126:2334-2340.

[22]Gong J, Wang C, Lee P, Chu P. & Fakih M. Response to PD-1 Blockade in Microsatellite Stable Metastatic Colorectal Cancer Harboring a POLE Mutation. Journal of the National Comprehensive Cancer Network,2017,15:142–147.

[23]Budczies, J. et al. Integrated analysis of the immunological and genetic status in and across cancer types: impact of mutational signatures beyond tumor mutational burden. Oncoimmunology,2018,7:e1526613.

[24]Young San Ko,Jung-Soo Pyo.Clinicopathological significance and prognostic role of tumor-infiltrating lymphocytes in colorectal cancer.The International Journal of Biological Markers,2019, Vol. 34(2) 132-138.

[25]Mark Ayers,Jared Lunceford,Michael Nebozhyn,et al. IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade.The Journal of Clinical Investigation,2017,127(8):2930-2940.

[26]Cristescu R, Mogg R, Ayers M, et al. Pan-tumor genomic bioma r kers for PD-1 checkpoint blockade-based immunotherapy[J]. Science, 2018, 362(6411):eaar3593.

[27]Chaput N, Lepage P, Coutzac C,et al.Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab.Ann Oncol,2017,28(6): 1368-1379.

[28]J A Wargo, et al.Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients.Science,2018,359(6371):97-103.

[29]Shahda S,Noonan A M,Bekaii-Saab T S, O’Neil B H,Sehdev A,Shaib W L,et al. A phase II study of pembrolizumab in combination with mFOLFOX6 for patients with advanced colorectal cancer. JCO,2017,35:3541.

[30]Fumet JD,Isambert N,Hervieu A ,Ghiringhelli F,et,al.Phase Ib/II trial evaluating the safety, tolerability and immunological activity of durvalumab (MEDI4736) (anti-PD-L1) plus tremelimumab (anti-CTLA-4) combined with FOLFOX in patients with metastatic colorectal cancer.ESMO Open.2018 ;3(4) :e000375.

[31]R. Kim, et al. Safety and Efficacy of Pembrolizumab (MK-3475) Plus Binimetinib Alone or Pembrolizumab Plus Chemotherapy With or Without Binimetinib in Metastatic Colorectal Cancer (mCRC) Participants (MK-3475-651).ESMO, 2020. E-Poster 493P.

[32]Turgeon GA, Weickhardt A, Azad AA, et al. Radiotherapy and immunotherapy:a synergistic effect in cancer care[J]. Med J Aust, 2019,210(1):47-53.

[33]Stapleton S, Jaffray D, Milosevic M, et al. Radiation effects on the tumor microenvironment: Implications for nanomedicine delivery[J].Adv Drug Deliv Rev, 2017, (109):119-130.

[34]Satoshi Yuki, Hideaki Bando, et,al.Short-term results of VOLTAGE-A: Nivolumab monotherapy and subsequent radical surgery following preoperative chemoradiotherapy in patients with microsatellite stable and microsatellite instability-high locally advanced rectal cancer.Journal of Clinical Oncology,2019,38:

[35]Fukuoka S, Hara H, Takahashi N, et al. Regorafenib Plus Nivolumab in Patients With Advanced Gastric or Colorectal Cancer.An Open-Label, Dose-Escalation, and Dose-Expansion Phase Ib Trial (REGONIVO, EPOC1603)[J]. J Clin Oncol, 2020,38(18): 2053-2061.


责任编辑:李慧
本文为医师报原创文章,禁止任何机构和个人私自转载或引用,如需转载,请联系授权(微信号:DAYI2006;邮箱:yishibao2017@163.com) 如未经允许转载或使用,本报将追究法律责任!

发表评论

最新评论

相关文章推荐