中国科技论文统计源期刊 中文核心期刊  
美国《化学文摘》《国际药学文摘》
《乌利希期刊指南》
WHO《西太平洋地区医学索引》来源期刊  
日本科学技术振兴机构数据库(JST)
第七届湖北十大名刊提名奖  
HERALD OF MEDICINE, 2018, 37(5): 542-546
doi: 10.3870/j.issn.1004-0781.2018.05.008
晚期非小细胞肺癌的免疫治疗进展*
Progression of Immunotherapy in Advanced Non-small Cell Lung Cancer
王俊, 王宝成

摘要:

免疫治疗是继传统手术、放射治疗(放疗)、化学治疗(化疗)之后逐步发展起来的治疗方法。近年来,免疫治疗因其较高的有效性和安全性在晚期非小细胞肺癌(NSCLC)综合治疗中的地位日渐突出。多个程序性死亡蛋白-1(PD-1)和程序性死亡分子配体-1(PD-L1)抑制药在晚期NSCLC一线、二线治疗中获得适应证;Ⅲ期NSCLC同步放化疗后辅以PD-L1抑制药可显著延长患者无进展生存期。然而,免疫治疗目前只对部分患者有效,晚期NSCLC免疫治疗的模式还需要不断优化,包括免疫治疗联合化疗、免疫治疗联合放疗等多种免疫联合治疗,以扩大获益人群;免疫治疗的疗效及生物标志物指导的精准治疗也在快速发展、提高。

关键词: ; ; 非小细胞 ; 免疫治疗 ; 程序性死亡蛋白-1 ; 预测标志物

Abstract:

Immunotherapy acts as one of developing therapeutic strategies following traditional surgery, radiation therapy and chemotherapy. Recently, immunotherapy shows increasing importance in the comprehensive treatment of advanced non-small cell lung cancer (NSCLC) because of its significant relaibility and efficacy. Several programmed cell death protein 1/ programmed death ligand 1 (PD-1/PD-L1) inhibitors have been approved for the treatment of advanced or metastatic NCCLC in first or second-line setting. In addition, immunotherapy with PD-L1 inhibitor promotes progression-free survival in stage III NSCLC patients who received previously concurrent radiation therapy and chemotherapy. However, due to the limited number of patients who could benefit from approved immunotherapy and the aim of expanding benifit groups, the immunotherapy treatment model of advanced NSCLC still needs continuous optimization, which includes immunotherapy combined with chemotherapy or immunotherapy combined with radiation therapy . Tumor marker-based precision therapy and efficacy with immunotherapy are in rapid development and improvement.

Key words: Cancer ; lung ; non-small cell ; Immunotherapy ; Programmed cell death protein 1 ; Predictive markers

肺癌是世界范围内发病率、致死率最高的恶性肿瘤。其中,80%的肺癌是非小细胞肺癌(non-small cell lung cancer,NSCLC),确诊时大多数已处于晚期。手术、放射治疗(放疗)、化学治疗(化疗)是NSCLC传统的治疗方法[1]。近年来,免疫治疗因其较高的有效性和安全性在NSCLC综合治疗中的地位日渐突出。多个程序性死亡蛋白-1(programmed cell death protein 1,PD-1)和程序性死亡分子配体-1(programmed death ligand 1,PD-L1)抑制药在晚期NSCLC一线、二线治疗中已获得适应证;Ⅲ期NSCLC同步放化疗后辅以PD-L1抑制药可显著延长无进展生存期[2]。然而,目前获批的免疫治疗仅仅对部分患者有效,晚期NSCLC免疫治疗的模式还需要不断优化,包括免疫治疗联合化疗、免疫治疗联合放疗等多种免疫联合治疗,以扩大获益人群;免疫治疗生物标志物指导的精准治疗也在快速发展、提高。

1 解除免疫抑制是NSCLC免疫治疗成功的关键因素

自20世纪90年代开始,人们对肺癌免疫治疗的探索从未停止。既往肺癌免疫治疗集中在癌症疫苗和非特异性免疫治疗,但收效甚微。例如,START试验显示,与安慰药比较,L-BLP25肿瘤疫苗并未提高Ⅲ期NSCLC患者总生存(overall survival,OS);其中序贯放化疗组患者接受免疫治疗生存期更短[3]。为何免疫检测点抑制药能在NSCLC取得成功?主要原因是免疫治疗策略发生了转变:过去一味强调如何增强机体免疫,如今已经转向如何阻断免疫抑制,包括涉及抗原调变的被动免疫抑制,以及涉及Treg细胞、髓源性抑制细胞、肿瘤相关的巨噬细胞、免疫抑制分子的主动免疫抑制。因此,深度理解肿瘤免疫抑制的机制,有助于认清免疫靶向治疗的重点。免疫抑制导致了免疫失衡;免疫治疗的目的就是要恢复机体的免疫平衡,尤其是T细胞的功能,最终达到消灭肿瘤的目的。当然,免疫抑制的机制比较复杂,肿瘤发展的不同阶段免疫抑制的程度和范围不一样,对不同抑制途径的阻断产生的效果也有差别。

2 免疫检查点抑制药打开NSCLC免疫治疗之门

2012年,PD-1抑制药Nivolumab首次被报道二线治疗晚期NSCLC取得成功,从此NSCLC免疫治疗迎来了蓬勃发展。目前,免疫检查点抑制药在NSCLC免疫治疗中的应用包括:①多个PD-1/PD-L1抑制药批准用于治疗晚期NSCLC,包括PD-1抑制药Nivolumab和Pembrolizumab,PD-L1抑制药Atezolizumab[4,5,6,7,8,9,10,11];②大多数PD-1/PD-L1单抗批准用于二线治疗晚期NSCLC,仅Pembrolizumab用于一线治疗晚期NSCLC,且治疗之前检测PD-L1表达要大于或等于50%[8];③Pembrolizumab联合培美曲塞+卡铂用于一线治疗晚期NSCLC,不论PD-L1表达状态如何[12];④PACIFIC试验结果显示,Durvalumab用于Ⅲ期同步放化疗的NSCLC的辅助治疗,延长了无进展生存(progression-free survival,PFS)[13],提示免疫治疗已经从晚期NSCLC延伸至了Ⅲ期NSCLC;⑤Pembrolizumab也可用于那些存在高度微卫星不稳定和错配修复基因缺失的实体瘤,包括NSCLC,即使NSCLC出现高度微卫星不稳定和错配修复基因缺失的概率不高[14]。因此,NSCLC免疫治疗的适应证已经覆盖了一线、二线;可单药使用,也可以联合使用;不仅适用于晚期患者,对局部晚期的患者也能带来临床获益。最近更新的OAK试验数据显示,接受Atezolizumab治疗的晚期NSCLC,即使按RECIST标准评价为进展,继续使用Atezolizumab仍有7%患者达到部分缓解(partial response,PR),49%患者靶病灶稳定,进展后的总生存时间超过1年。

3 免疫治疗可成为NSCLC治愈性的手段

免疫检查点抑制药的出现打破了肺癌免疫治疗的困境。现阶段肺癌免疫治疗方法特指免疫检查点抑制药。目前肺癌免疫治疗的地位正在逐步上升,与化疗、靶向治疗一起成为NSCLC治疗的主要手段。近期CA209-003试验的长期随访结果显示,接受Nivolumab单药治疗的晚期NSCLC患者5年总生存率为16%;而接受化疗的晚期NSCLC患者5年生存率只有4.8%。2017年ASCO年会报道,KEYNOTE-001试验中最早接受Pembrolizumab治疗的肺癌患者的3年总生存率达20.4%,其中接受一线免疫治疗的患者甚至达到26.4%。而对CheckMate 017和CheckMate 057的联合数据分析发现,接受Nivolumab治疗的鳞癌和非鳞癌患者2年OS分别为23%和29%;接受免疫治疗获得完全缓解(complete response,CR)的患者中,分别有37%的鳞癌和34%的非鳞癌维持2年治疗停药后能维持疗效,提示免疫治疗获益的长期性[15]

4 免疫检查点抑制药丰富鳞癌治疗选择

相对于腺癌,肺鳞癌有以下典型特征:①多见于吸烟男性;②中心型为主,侵袭性强,常引起咯血、呼吸困难、感染等并发症,症状重;③属于难治类型,因疗效差和出血风险,无法使用培美曲塞、贝伐珠单抗等抗癌药物;④驱动基因突变率不足5%,多数无机会接受靶向治疗;虽然FLEX试验显示化疗加西妥昔单抗组较化疗组OS时间延长了1.2个月,但临床获益极其有限;⑤一线化疗有效率达30%~40%,但持续时间短,总体预后差,总生存时间8~10个月。CheckMate 063和017试验结果显示,鳞癌患者可直接从二线Nivolumab治疗中获益,中位OS分别为8.2和9.2个月,这已经非常接近一线化疗的OS数据;此外,KEYNOTE-010试验结果也支持鳞癌患者可获益于二线使用Pembrolizumab。在KEYNOTE-024试验中,鳞癌患者接受一线Pembrolizumab治疗,复发风险下降65%,而腺癌患者只下降45%。因此,鳞癌患者不再像以前靶向治疗时代被排除在外,免疫治疗成为鳞癌患者新的治疗标准。

5 免疫联合治疗扩大了受益人群

NSCLC免疫治疗虽然获得了突破,但总体有效率不高,仅30%患者有获益机会。如何扩大适应人群是免疫治疗需要面对的问题,因此免疫联合治疗应运而生。正在进行的免疫联合治疗策略包括:①PD-1/PD-L1抑制药联合化疗;②PD-1/PD-L1抑制药联合放疗[16];③PD-1/PD-L1抑制药联合抗血管生成治疗;④PD-1/PD-L1抑制药联合其他免疫治疗方法,如癌症疫苗、细胞因子、过继性细胞输注(ACT)等。

KEYNOTE-021试验显示,Pembrolizumab联合培美曲塞加卡铂与培美曲塞加卡铂化疗比较,客观缓解率(objective response rate,ORR)从29%提高到55%,PFS延长了4.1个月。2017年5月,美国食品药品管理局(FDA)批准了第一个免疫联合化疗的适应证,即Pembrolizumab联合培美曲塞加卡铂一线治疗晚期NSCLC,且不需要考虑PD-L1表达状态。KEYNOTE-021试验的安全性数据显示,虽然联合治疗组3~4级不良事件发生率要高于单用化疗组(39%:26%),但整体可控。Nivolumab联合化疗虽然在ORR、PFS上并未显示出优势,但Nivolumab联合紫杉醇加卡铂组57%患者OS超过了2年[12]。这些临床研究的结论改变了人们过去“化疗降低了机体免疫力”的认识。因此,基于化疗药物可以诱导肿瘤细胞发生免疫原性死亡,释放肿瘤抗原;化疗药物干扰肿瘤细胞的免疫逃逸机制,PD-1/PD-L1抑制药联合化疗是一种有前景的治疗策略。

过去在肺癌患者中人们观察到接受放疗后远处病灶会消失,但这种情况非常罕见,被认为是放疗激活了机体免疫,称为远隔效应。KEYNOTE-001试验的回顾性研究结果显示,与没有接受过放疗的患者比较,之前接受任何放疗患者再加免疫治疗,其PFS延长了2.3个月,OS延长了5.4个月[17]。而最近PACIFIC试验结果的公布更加支持PD-L1抑制药Durvalumab联合同步放化疗的有效性[13]。其他免疫联合治疗目前正在不断开展,包括免疫治疗间的组合。例如CTLA-4联合PD-1单抗与CTLA-4单抗比较,显著提高了晚期黑色素瘤患者PFS。

然而,免疫治疗的联合需要考虑用药的顺序和时间。例如,近期的动物实验结果显示,同步使用针对免疫共刺激分子OX4的激动抗体anti-OX4与PD-1抑制药不仅不产生协同效应或累加效应,反而产生了不良效果,即anti-OX4抗体的有效作用被抵消了;T细胞增殖率、治疗效果和存活率均不如anti-OX4单药治疗[18,19]。此外,免疫联合治疗也要考虑免疫治疗的种类,例如START中,L-BLP25疫苗并未提高Ⅲ期NSCLC患者OS;Ipilimumab联合化疗虽然延长了PFS及免疫相关的PFS,但获益有限,且毒性明显[20]

6 NSCLC免疫治疗逐步实现精准化

NSCLC目前在沿用病理分型的基础上已经发展到了分子分型,可以分为有驱动基因突变的NSCLC和无驱动基因突变的NSCLC。免疫治疗的疗效预测也需要根据分子分型来精确实施。有驱动基因突变的NSCLC涉及表皮生长因子基因突变、间变性淋巴瘤激酶基因重排、受体酪氨酸激酶基因重排、MET 14号染色体跳跃突变、BRAF突变等。已有明确的证据显示,有驱动基因突变的NSCLC一线使用PD-1/PD-L1抗体的疗效欠佳,ORR和PFS都有受损[21,22]。但在表皮生长因子受体敏感性突变的患者中,如果PD-L1高表达,仍有12.2%的肿瘤对PD-L1抗体起反应。因此,NCCN指南推荐无T790M突变的肺癌,如果PD-L1高表达仍可以接受单药免疫治疗。在联合治疗方面,PD-1/PD-L1抑制药联合第1代或第3代表皮生长因子受体-酪氨酸激酶抑制药有效率可达70%,但同时也产生了严重的毒副作用,3~4级的比例高达50%。因此,目前有驱动基因突变的NSCLC一线治疗不推荐使用PD-1/PD-L1抗体;联合EGFR-TKI无论是临床实践还是临床试验都需要谨慎,但PD-L1高表达的患者是否可以使用,后线治疗是否可以使用,目前仍存在争议,相关的临床试验正在进行中。

无驱动基因突变的NSCLC免疫治疗疗效预测是另一个需要探索的方向。CheckMate 026研究的回顾性分析显示,肿瘤突变负荷(tumor mutation burden,TMB)也与免疫治疗的疗效相关,高TMB的NSCLC对PD-1/PD-L1抑制药的反应率高。虽然MSI-H和d-MMR的患者是免疫治疗的适应人群,但不是所有高TMB的肿瘤都能检测到MSI-或d-MMR,其他基因改变如POLE突变也是免疫治疗起反应的标志物[23]。因此,通过全基因组或全外显子高通量分析基因突变负荷有助于筛选免疫治疗敏感人群;通过突变组分析有代表性基因的突变状态,也可以预测免疫治疗疗效,其可行性更强、临床使用更加方便,例如p53、KRAS、LKB1基因等。

来自肿瘤微环境的标志物也可以预测PD-1/PD-L1抑制药的疗效。最具有代表性的是PD-L1表达和C D 8 + T细胞浸润。虽然不同PD-1/PD-L1抑制药参与的临床试验所报道的结果有一些差别,但PD-L1高表达的人群接受免疫治疗所带来的获益明显要高于低表达的人群。因此,PD-L1表达可以作为判断PD-1/PD-L1抑制药疗效的基础标志物。

当然,除了来自肿瘤、免疫系统的标志物,源于机体的多种因子也可以预测免疫治疗疗效,例如肠道微生物群、循环淋巴细胞计数等[24,25]。目前尚无能独立预测免疫治疗的有效标志物,但将来的研究方法主要包括:①探索稳定的单标志物的临床价值,作为初步评估;②使用多种标志物预测,例如突变组分析、免疫评分,或突变负荷结合PD-L1表达;③高通量技术和生物信息学技术的发展使大数据环境下寻找更有效的生物标志物成为可能。

7 展望

近年来,NSCLC免疫治疗取得了不错的成绩,多个PD-1/PD-L1抑制药相继获批用于临床一线、二线治疗;NSCLC患者5年总生存率达到16%;精准检测的进步也使得筛选免疫治疗的优势人群成为可能。然而,也应该清醒地认识到,肺癌作为全世界发病率和死亡率极高的肿瘤类型,未来还需要在多个层面进一步探索,使得免疫治疗方法更精准、获益人群更多、疗效更显著,治疗更安全。这些措施包括:①进一步分析免疫微环境的功能作用,探索肿瘤免疫抑制、免疫耐药的机制,寻找更多免疫治疗的功能性靶点,例如其他检查点抑制剂或激动剂TIM-3、LAG-3、OX40等;针对Treg细胞、髓源性抑制细胞、肿瘤相关巨噬细胞、B细胞、肿瘤浸润的成纤维细胞等免疫抑制性细胞群;②继续开展免疫治疗的临床试验,特别是应用以生物标志物筛选甚至终点评价的临床试验、以整体观设计的免疫治疗临床试验、免疫联合治疗的临床试验;③加快转化型研究的步伐,整合目前免疫治疗的资源,在大数据上挖掘、寻找免疫精准治疗的线索;④进一步规范现有的、逐步发展的免疫治疗新方法,包括免疫治疗疗效评估、免疫治疗的安全管理、治疗持续时间、与其他治疗联合应用的间隔时间、先后顺序等;在已知的基础上探索未知细节,在未知上寻求解决方案。

The authors have declared that no competing interests exist.

参考文献

[1] SIEGEL R L,MILLER K D,JEMAL A.Cancer statistics,2017[J].CA Cancer J Clin,2017,67(1):7-30.
DOI:10.3322/caac.21387      URL    
[本文引用:1]
[2] FORMISANO L,JANSEN V M,MARCIANO R,et al.From biology to therapy:improvements of therapeutic options in lung cancer[J].Anticancer Agents Med Chem,DOI:10.2174/1871520617666170912123416.
Abstract Lung cancer is the leading cause of cancer-related mortality around the world, despite effective chemotherapeutic agents, the prognosis has remained poor for a long time. The discovery of molecular changes that drive lung cancer has led to a dramatic shift in the therapeutic landscape of this disease. In "in vitro" and "in vivo" models of NSCLC (non-small cell lung cancer), angiogenesis blockade has demonstrated an excellent anti-tumor activity, thus, a number of anti-angiogenic drugs have been approved by regulatory authorities for use in clinical practice. Much more interesting is the discovery of EGFR (epithelial growth factor receptor) mutations that predict sensitivity to the anti-EGFR tyrosine kinase inhibitors (TKIs), a class of drugs that has shown to significantly improve survival when compared with standard chemotherapy in the first-line treatment of metastatic NSCLC. Nevertheless, after an initial response, resistance often occurs and prognosis becomes dismal. Biomolecular studies on cell line models have led to the discovery of mutations (e.g., T790M) that confer resistance to anti-EGFR inhibitors. Fortunately, drugs that are able to circumvent this mechanism of resistance have been developed and have been recently approved for clinical use. The discovery of robust intra-tumor lymphocyte infiltration in NSCLC has paved the way to several strategies able to restore the immune response. Thus, agents interfering with PD-1/PD-L1 (programmed death) pathways make up a significant portion of the armamentarium of cancer therapies for NSCLC. In all the above-mentioned situations, the basis of the success in treating NSCLC has started from understanding of the mutational landscape of the tumor. Copyright Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.
DOI:10.2174/1871520617666170912123416      PMID:28901258      URL    
[本文引用:1]
[3] BUTTS C,SOCINSKI M A,MITCHELL P L,et al.Tecemo-tide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START):a randomised,double-blind,phase 3 trial[J].Lancet Oncol,2014,15(1):59-68.
Abstract BACKGROUND: Effective maintenance therapies after chemoradiotherapy for lung cancer are lacking. Our aim was to investigate whether the MUC1 antigen-specific cancer immunotherapy tecemotide improves survival in patients with stage III unresectable non-small-cell lung cancer when given as maintenance therapy after chemoradiation. METHODS: The phase 3 START trial was an international, randomised, double-blind trial that recruited patients with unresectable stage III non-small-cell lung cancer who had completed chemoradiotherapy within the 4-12 week window before randomisation and received confirmation of stable disease or objective response. Patients were stratified by stage (IIIA vs IIIB), response to chemoradiotherapy (stable disease vs objective response), delivery of chemoradiotherapy (concurrent vs sequential), and region using block randomisation, and were randomly assigned (2:1, double-blind) by a central interactive voice randomisation system to either tecemotide or placebo. Injections of tecemotide (806 g lipopeptide) or placebo were given every week for 8 weeks, and then every 6 weeks until disease progression or withdrawal. Cyclophosphamide 300 mg/m(2) (before tecemotide) or saline (before placebo) was given once before the first study drug administration. The primary endpoint was overall survival in a modified intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT00409188 . FINDINGS: From Feb 22, 2007, to Nov 15, 2011, 1513 patients were randomly assigned (1006 to tecemotide and 507 to placebo). 274 patients were excluded from the primary analysis population as a result of a clinical hold, resulting in analysis of 829 patients in the tecemotide group and 410 in the placebo group in the modified intention-to-treat population. Median overall survival was 25.6 months (95% CI 22.5-29.2) with tecemotide versus 22.3 months (19.6-25.5) with placebo (adjusted HR 0.88, 0.75-1.03; p=0.123). In the patients who received previous concurrent chemoradiotherapy, median overall survival for the 538 (65%) of 829 patients assigned to tecemotide was 30.8 months (95% CI 25.6-36.8) compared with 20.6 months (17.4-23.9) for the 268 (65%) of 410 patients assigned to placebo (adjusted HR 0.78, 0.64-0.95; p=0.016). In patients who received previous sequential chemoradiotherapy, overall survival did not differ between the 291 (35%) patients in the tecemotide group and the 142 (35%) patients in the placebo group (19.4 months [95% CI 17.6-23.1] vs 24.6 months [18.8-33.0], respectively; adjusted HR 1.12, 0.87-1.44; p=0.38). Grade 3-4 adverse events seen with a greater than 2% frequency with tecemotide were dyspnoea (49 [5%] of 1024 patients in the tecemotide group vs 21 [4%] of 477 patients in the placebo group), metastases to central nervous system (29 [3%] vs 6 [1%]), and pneumonia (23 [2%] vs 12 [3%]). Serious adverse events with a greater than 2% frequency with tecemotide were pneumonia (30 [3%] in the tecemotide group vs 14 [3%] in the placebo group), dyspnoea (29 [3%] vs 13 [3%]), and metastases to central nervous system (32 [3%] vs 9 [2%]). Serious immune-related adverse events did not differ between groups. INTERPRETATION: We found no significant difference in overall survival with the administration of tecemotide after chemoradiotherapy compared with placebo for all patients with unresectable stage III non-small-cell lung cancer. However, tecemotide might have a role for patients who initially receive concurrent chemoradiotherapy, and further study in this population is warranted. FUNDING: Merck KGaA (Darmstadt, Germany). Copyright 2014 Elsevier Ltd. All rights reserved.
DOI:10.1016/S1470-2045(13)70510-2      PMID:24331154      URL    
[本文引用:1]
[4] RIZVI N A,MAZIERES J,PLANCHARD D,et al.Activity and safety of nivolumab,an anti-PD-1 immune checkpoint inhibitor,for patients with advanced,refractory squamous non-small-cell lung cancer (CheckMate 063):a phase 2,single-arm trial[J].Lancet Oncol,2015,16(3):257-265.
Abstract BACKGROUND: Patients with squamous non-small-cell lung cancer that is refractory to multiple treatments have poor outcomes. We assessed the activity of nivolumab, a fully human IgG4 PD-1 immune checkpoint inhibitor antibody, for patients with advanced, refractory, squamous non-small-cell lung cancer. METHODS: We did this phase 2, single-arm trial at 27 sites (academic, hospital, and private cancer centres) in France, Germany, Italy, and USA. Patients who had received two or more previous treatments received intravenous nivolumab (3 mg/kg) every 2 weeks until progression or unacceptable toxic effects. The primary endpoint was the proportion of patients with a confirmed objective response as assessed by an independent radiology review committee. We included all treated patients in the analyses. This study is registered with ClinicalTrials.gov, number NCT01721759 . FINDINGS: Between Nov 16, 2012, and July 22, 2013, we enrolled and treated 117 patients. 17 (1400·5%, 95% CI 800·7-2200·2) of 117 patients had an objective response as assessed by an independent radiology review committee. Median time to response was 300·3 months (IQR 200·2-400·8), and median duration of response was not reached (95% CI 800·31-not applicable); 13 (77%) of 17 of responses were ongoing at the time of analysis. 30 (26%) of 117 patients had stable disease (median duration 600·0 months, 95% CI 400·7-1000·9). 20 (17%) of 117 patients reported grade 3-4 treatment-related adverse events, including: fatigue (five [4%] of 117 patients), pneumonitis (four [3%]), and diarrhoea (three [3%]). There were two treatment-associated deaths caused by pneumonia and ischaemic stroke that occurred in patients with multiple comorbidities in the setting of progressive disease. INTERPRETATION: Nivolumab has clinically meaningful activity and a manageable safety profile in previously treated patients with advanced, refractory, squamous non-small cell lung cancer. These data support the assessment of nivolumab in randomised, controlled, phase 3 studies of first-line and second-line treatment. FUNDING: Bristol-Myers Squibb. Copyright 0008 2015 Elsevier Ltd. All rights reserved.
DOI:10.1016/S1470-2045(15)70054-9      PMID:5726228      URL    
[本文引用:1]
[5] BRAHMER J,RECKAMP K L,BAAS P,et al.Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer[J].N Engl J Med,2015,373(2):123-135.
Abstract BACKGROUND: Nivolumab, a fully human IgG4 programmed death 1 (PD-1) immune-checkpoint-inhibitor antibody, disrupts PD-1-mediated signaling and may restore antitumor immunity. METHODS: In this randomized, open-label, international phase 3 study, we assigned patients with nonsquamous non-small-cell lung cancer (NSCLC) that had progressed during or after platinum-based doublet chemotherapy to receive nivolumab at a dose of 3 mg per kilogram of body weight every 2 weeks or docetaxel at a dose of 75 mg per square meter of body-surface area every 3 weeks. The primary end point was overall survival. RESULTS: Overall survival was longer with nivolumab than with docetaxel. The median overall survival was 12.2 months (95% confidence interval [CI], 9.7 to 15.0) among 292 patients in the nivolumab group and 9.4 months (95% CI, 8.1 to 10.7) among 290 patients in the docetaxel group (hazard ratio for death, 0.73; 96% CI, 0.59 to 0.89; P=0.002). At 1 year, the overall survival rate was 51% (95% CI, 45 to 56) with nivolumab versus 39% (95% CI, 33 to 45) with docetaxel. With additional follow-up, the overall survival rate at 18 months was 39% (95% CI, 34 to 45) with nivolumab versus 23% (95% CI, 19 to 28) with docetaxel. The response rate was 19% with nivolumab versus 12% with docetaxel (P=0.02). Although progression-free survival did not favor nivolumab over docetaxel (median, 2.3 months and 4.2 months, respectively), the rate of progression-free survival at 1 year was higher with nivolumab than with docetaxel (19% and 8%, respectively). Nivolumab was associated with even greater efficacy than docetaxel across all end points in subgroups defined according to prespecified levels of tumor-membrane expression (0909061%, 0909065%, and 09090610%) of the PD-1 ligand. Treatment-related adverse events of grade 3 or 4 were reported in 10% of the patients in the nivolumab group, as compared with 54% of those in the docetaxel group. CONCLUSIONS: Among patients with advanced nonsquamous NSCLC that had progressed during or after platinum-based chemotherapy, overall survival was longer with nivolumab than with docetaxel. (Funded by Bristol-Myers Squibb; CheckMate 057 ClinicalTrials.gov number, NCT01673867 .).
DOI:10.1056/NEJMoa1507643      PMID:26412456      URL    
[本文引用:1]
[6] BORGHAEI H,PAZ-ARES L,HORN L,et al.Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer[J].N Engl J Med,2015,373(17):1627-1639.
Abstract BACKGROUND: Nivolumab, a fully human IgG4 programmed death 1 (PD-1) immune-checkpoint-inhibitor antibody, disrupts PD-1-mediated signaling and may restore antitumor immunity. METHODS: In this randomized, open-label, international phase 3 study, we assigned patients with nonsquamous non-small-cell lung cancer (NSCLC) that had progressed during or after platinum-based doublet chemotherapy to receive nivolumab at a dose of 3 mg per kilogram of body weight every 2 weeks or docetaxel at a dose of 75 mg per square meter of body-surface area every 3 weeks. The primary end point was overall survival. RESULTS: Overall survival was longer with nivolumab than with docetaxel. The median overall survival was 12.2 months (95% confidence interval [CI], 9.7 to 15.0) among 292 patients in the nivolumab group and 9.4 months (95% CI, 8.1 to 10.7) among 290 patients in the docetaxel group (hazard ratio for death, 0.73; 96% CI, 0.59 to 0.89; P=0.002). At 1 year, the overall survival rate was 51% (95% CI, 45 to 56) with nivolumab versus 39% (95% CI, 33 to 45) with docetaxel. With additional follow-up, the overall survival rate at 18 months was 39% (95% CI, 34 to 45) with nivolumab versus 23% (95% CI, 19 to 28) with docetaxel. The response rate was 19% with nivolumab versus 12% with docetaxel (P=0.02). Although progression-free survival did not favor nivolumab over docetaxel (median, 2.3 months and 4.2 months, respectively), the rate of progression-free survival at 1 year was higher with nivolumab than with docetaxel (19% and 8%, respectively). Nivolumab was associated with even greater efficacy than docetaxel across all end points in subgroups defined according to prespecified levels of tumor-membrane expression (0909061%, 0909065%, and 09090610%) of the PD-1 ligand. Treatment-related adverse events of grade 3 or 4 were reported in 10% of the patients in the nivolumab group, as compared with 54% of those in the docetaxel group. CONCLUSIONS: Among patients with advanced nonsquamous NSCLC that had progressed during or after platinum-based chemotherapy, overall survival was longer with nivolumab than with docetaxel. (Funded by Bristol-Myers Squibb; CheckMate 057 ClinicalTrials.gov number, NCT01673867 .).
DOI:10.1056/NEJMoa1507643      PMID:26412456      URL    
[本文引用:1]
[7] GARON E B,RIZVI N A,HUI R,et al.Pembrolizumab for the treatment of non-small-cell lung cancer[J].N Engl J Med,2015,372(21):2018-2028.
We assessed the efficacy and safety of programmed cell death 1 (PD-1) inhibition with pembrolizumab in patients with advanced non-small-cell lung cancer enrolled in a phase 1 study. We also sought to define and validate an expression level of the PD-1 ligand 1 (PD-L1) that is associated with the likelihood of clinical benefit. We assigned 495 patients receiving pembrolizumab (at a dose of either 2 mg or 10 mg per kilogram of body weight every 3 weeks or 10 mg per kilogram every 2 weeks) to either a training group (182 patients) or a validation group (313 patients). We assessed PD-L1 expression in tumor samples using immunohistochemical analysis, with results reported as the percentage of neoplastic cells with staining for membranous PD-L1 (proportion score). Response was assessed every 9 weeks by central review. Common side effects that were attributed to pembrolizumab were fatigue, pruritus, and decreased appetite, with no clear difference according to dose or schedule. Among all the patients, the objective response rate was 19.4%, and the median duration of response was 12.5 months. The median duration of progression-free survival was 3.7 months, and the median duration of overall survival was 12.0 months. PD-L1 expression in at least 50% of tumor cells was selected as the cutoff from the training group. Among patients with a proportion score of at least 50% in the validation group, the response rate was 45.2%. Among all the patients with a proportion score of at least 50%, median progression-free survival was 6.3 months; median overall survival was not reached. Pembrolizumab had an acceptable side-effect profile and showed antitumor activity in patients with advanced non-small-cell lung cancer. PD-L1 expression in at least 50% of tumor cells correlated with improved efficacy of pembrolizumab. (Funded by Merck; KEYNOTE-001 ClinicalTrials.gov number, NCT01295827.).
DOI:10.1056/NEJMoa1501824      PMID:25891174      URL    
[本文引用:1]
[8] RECK M,RODRIGUEZ-ABREU D,Robinson A G,et al.Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer[J].N Engl J Med,2016,375(19):1823-1833.
DOI:10.1056/NEJMoa1606774      URL    
[本文引用:2]
[9] HERBST R S,BAAS P,KIM D W,et al.Pembrolizumab versus docetaxel for previously treated,PD-L1-positive,advanced non-small-cell lung cancer (KEYNOTE-010):a randomised controlled trial[J].Lancet,2016,387(10027):1540-1550.
DOI:10.1016/S0140-6736(15)01281-7      URL    
[本文引用:1]
[10] RITTMEYER A,BARLESI F,WATERKAMP D,et al.Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK):a phase 3,open-label,multicentre randomised controlled trial[J].Lancet,2017,389(10066):255-265.
Atezolizumab is a humanised antiprogrammed death-ligand 1 (PD-L1) monoclonal antibody that inhibits PD-L1 and programmed death-1 (PD-1) and PD-L1 and B7-1 interactions, reinvigorating anticancer immunity. We assessed its efficacy and safety versus docetaxel in previously treated patients with non-small-cell lung cancer. We did a randomised, open-label, phase 3 trial (OAK) in 194 academic or community oncology centres in 31 countries. We enrolled patients who had squamous or non-squamous non-small-cell lung cancer, were 18 years or older, had measurable disease per Response Evaluation Criteria in Solid Tumors, and had an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients had received one to two previous cytotoxic chemotherapy regimens (one or more platinum based combination therapies) for stage IIIB or IV non-small-cell lung cancer. Patients with a history of autoimmune disease and those who had received previous treatments with docetaxel, CD137 agonists, anti-CTLA4, or therapies targeting the PD-L1 and PD-1 pathway were excluded. Patients were randomly assigned (1:1) to intravenously receive either atezolizumab 1200 mg or docetaxel 75 mg/mevery 3 weeks by permuted block randomisation (block size of eight) via an interactive voice or web response system. Coprimary endpoints were overall survival in the intention-to-treat (ITT) and PD-L1-expression population TC1/2/3 or IC1/2/3 (≥1% PD-L1 on tumour cells or tumour-infiltrating immune cells). The primary efficacy analysis was done in the first 850 of 1225 enrolled patients. This study is registered with ClinicalTrials.gov, number NCT02008227. Between March 11, 2014, and April 29, 2015, 1225 patients were recruited. In the primary population, 425 patients were randomly assigned to receive atezolizumab and 425 patients were assigned to receive docetaxel. Overall survival was significantly longer with atezolizumab in the ITT and PD-L1-expression populations. In the ITT population, overall survival was improved with atezolizumab compared with docetaxel (median overall survival was 13·8 months [95% CI 11·8-15·7] vs 9·6 months [8·6-11·2]; hazard ratio [HR] 0·73 [95% CI 0·62-0·87], p=0·0003). Overall survival in the TC1/2/3 or IC1/2/3 population was improved with atezolizumab (n=241) compared with docetaxel (n=222; median overall survival was 15·7 months [95% CI 12·6-18·0] with atezolizumab vs 10·3 months [8·8-12·0] with docetaxel; HR 0·74 [95% CI 0·58-0·93]; p=0·0102). Patients in the PD-L1 low or undetectable subgroup (TC0 and IC0) also had improved survival with atezolizumab (median overall survival 12·6 months vs 8·9 months; HR 0·75 [95% CI 0·59-0·96]). Overall survival improvement was similar in patients with squamous (HR 0·73 [95% CI 0·54-0·98]; n=112 in the atezolizumab group and n=110 in the docetaxel group) or non-squamous (0·73 [0·60-0·89]; n=313 and n=315) histology. Fewer patients had treatment-related grade 3 or 4 adverse events with atezolizumab (90 [15%] of 609 patients) versus docetaxel (247 [43%] of 578 patients). One treatment-related death from a respiratory tract infection was reported in the docetaxel group. To our knowledge, OAK is the first randomised phase 3 study to report results of a PD-L1-targeted therapy, with atezolizumab treatment resulting in a clinically relevant improvement of overall survival versus docetaxel in previously treated non-small-cell lung cancer, regardless of PD-L1 expression or histology, with a favourable safety profile. F. Hoffmann-La Roche Ltd, Genentech, Inc.
DOI:10.1016/S0140-6736(16)32517-X      PMID:27979383      URL    
[本文引用:1]
[11] FEHRENBACHER L,SPIRA A,BALLINGER M,et al.Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR):a multicentre,open-label,phase 2 randomised controlled trial[J].Lancet,2016,387(10030):1837-1846.
Abstract BACKGROUND: Outcomes are poor for patients with previously treated, advanced or metastatic non-small-cell lung cancer (NSCLC). The anti-programmed death ligand 1 (PD-L1) antibody atezolizumab is clinically active against cancer, including NSCLC, especially cancers expressing PD-L1 on tumour cells, tumour-infiltrating immune cells, or both. We assessed efficacy and safety of atezolizumab versus docetaxel in previously treated NSCLC, analysed by PD-L1 expression levels on tumour cells and tumour-infiltrating immune cells and in the intention-to-treat population. METHODS: In this open-label, phase 2 randomised controlled trial, patients with NSCLC who progressed on post-platinum chemotherapy were recruited in 61 academic medical centres and community oncology practices across 13 countries in Europe and North America. Key inclusion criteria were Eastern Cooperative Oncology Group performance status 0 or 1, measurable disease by Response Evaluation Criteria In Solid Tumors version 1.1 (RECIST v1.1), and adequate haematological and end-organ function. Patients were stratified by PD-L1 tumour-infiltrating immune cell status, histology, and previous lines of therapy, and randomly assigned (1:1) by permuted block randomisation (with a block size of four) using an interactive voice or web system to receive intravenous atezolizumab 1200 mg or docetaxel 75 mg/m(2) once every 3 weeks. Baseline PD-L1 expression was scored by immunohistochemistry in tumour cells (as percentage of PD-L1-expressing tumour cells TC309090650%, TC20909065% and <50%, TC10909061% and <5%, and TC0<1%) and tumour-infiltrating immune cells (as percentage of tumour area: IC309090610%, IC20909065% and <10%, IC10909061% and <5%, and IC0<1%). The primary endpoint was overall survival in the intention-to-treat population and PD-L1 subgroups at 173 deaths. Biomarkers were assessed in an exploratory analysis. We assessed safety in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01903993 . FINDINGS: Patients were enrolled between Aug 5, 2013, and March 31, 2014. 144 patients were randomly allocated to the atezolizumab group, and 143 to the docetaxel group. 142 patients received at least one dose of atezolizumab and 135 received docetaxel. Overall survival in the intention-to-treat population was 1200·6 months (95% CI 900·7-1600·4) for atezolizumab versus 900·7 months (800·6-1200·0) for docetaxel (hazard ratio [HR] 000·73 [95% CI 000·53-000·99]; p=000·04). Increasing improvement in overall survival was associated with increasing PD-L1 expression (TC3 or IC3 HR 000·49 [000·22-100·07; p=000·068], TC2/3 or IC2/3 HR 000·54 [000·33-000·89; p=000·014], TC1/2/3 or IC1/2/3 HR 000·59 [000·40-000·85; p=000·005], TC0 and IC0 HR 100·04 [000·62-100·75; p=000·871]). In our exploratory analysis, patients with pre-existing immunity, defined by high T-effector-interferon-0206-associated gene expression, had improved overall survival with atezolizumab. 11 (8%) patients in the atezolizumab group discontinued because of adverse events versus 30 (22%) patients in the docetaxel group. 16 (11%) patients in the atezolizumab group versus 52 (39%) patients in the docetaxel group had treatment-related grade 3-4 adverse events, and one (<1%) patient in the atezolizumab group versus three (2%) patients in the docetaxel group died from a treatment-related adverse event. INTERPRETATION: Atezolizumab significantly improved survival compared with docetaxel in patients with previously treated NSCLC. Improvement correlated with PD-L1 immunohistochemistry expression on tumour cells and tumour-infiltrating immune cells, suggesting that PD-L1 expression is predictive for atezolizumab benefit. Atezolizumab was well tolerated, with a safety profile distinct from chemotherapy. FUNDING: F Hoffmann-La Roche/Genentech Inc. Copyright 0008 2016 Elsevier Ltd. All rights reserved.
DOI:10.1016/S0140-6736(16)00587-0      PMID:26970723      URL    
[本文引用:1]
[12] LANGER C J,GADGEEL S M,BORGHAEI H,et al.Carboplatin and pemetrexed with or without pembrolizumab for advanced,non-squamous non-small-cell lung cancer:a randomised,phase 2 cohort of the open-label KEYNOTE-021 study[J].Lancet Oncol,2016,17(11):1497-1508.
Merck & Co.
DOI:10.1016/S1470-2045(16)30498-3      PMID:27745820      URL    
[本文引用:2]
[13] ANTONIA S J,VILLEGAS A,DANIEL D,et al.Durva-lumab after chemoradiotherapy in stage III non-small-cell lung cancer[J].Engl J Med,2017,377(20):1919-1929.
DOI:10.1056/NEJMoa1709937      URL    
[本文引用:2]
[14] LE D T,DURHAM J N,SMITH K N,et al.Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade[J].Science,2017,357(6349):409-413.
The genomes of cancers deficient in mismatch repair (MMR) contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with MMR deficiency were sensitive to immune checkpoint blockade with anti-PD-1 antibodies. We have expanded this study to now evaluate efficacy of PD-1 blockade in patients with advanced MMR-deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients and complete responses were achieved in 21% of patients. Responses were durable with median progression-free and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in MMR-deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers tissue of origin.
DOI:10.1126/science.aan6733      PMID:28596308      URL    
[本文引用:1]
[15] HORN L,SPIGEL D R,VOKES E E,et al.Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer:two-year outcomes from two randomized,open-label,phase III trials (CheckMate 017 and CheckMate 057)[J].J Clin Oncol,2017,35(35):3924-3933.
Purpose Nivolumab, a programmed death-1 inhibitor, prolonged overall survival compared with docetaxel in two independent phase III studies in previously treated patients with advanced squamous (CheckMate 017; ClinicalTrials.gov identifier: NCT01642004) or nonsquamous (CheckMate 057; ClinicalTrials.gov identifier: NCT01673867) non-small-cell lung cancer (NSCLC). We report updated results, including a pooled analysis of the two studies. Methods Patients with stage IIIB/IV squamous (N = 272) or nonsquamous (N = 582) NSCLC and disease progression during or after prior platinum-based chemotherapy were randomly assigned 1:1 to nivolumab (3 mg/kg every 2 weeks) or docetaxel (75 mg/m2 every 3 weeks). Minimum follow-up for survival was 24.2 months. Results Two-year overall survival rates with nivolumab versus docetaxel were 23% (95% CI, 16% to 30%) versus 8% (95% CI, 4% to 13%) in squamous NSCLC and 29% (95% CI, 24% to 34%) versus 16% (95% CI, 12% to 20%) in nonsquamous NSCLC; relative reductions in the risk of death with nivolumab versus docetaxel remained similar to those reported in the primary analyses. Durable responses were observed with nivolumab; 10 (37%) of 27 confirmed responders with squamous NSCLC and 19 (34%) of 56 with nonsquamous NSCLC had ongoing responses after 2 years' minimum follow-up. No patient in either docetaxel group had an ongoing response. In the pooled analysis, the relative reduction in the risk of death with nivolumab versus docetaxel was 28% (hazard ratio, 0.72; 95% CI, 0.62 to 0.84), and rates of treatment-related adverse events were lower with nivolumab than with docetaxel (any grade, 68% v 88%; grade 3 to 4, 10% v 55%). Conclusion Nivolumab provides long-term clinical benefit and a favorable tolerability profile compared with docetaxel in previously treated patients with advanced NSCLC.
DOI:10.1200/JCO.2017.74.3062      PMID:29023213      URL    
[本文引用:1]
[16] KALBASI A,JUNE C H,HAAS N,et al.Radiation and immunotherapy:a synergistic combination[J].J Clin Invest,2013,123(7):2756-2763.
Abstract Immunotherapy can be an effective treatment for metastatic cancer, but a significant subpopulation will not respond, likely due to the lack of antigenic mutations or the immune-evasive properties of cancer. Likewise, radiation therapy (RT) is an established cancer treatment, but local failures still occur. Clinical observations suggest that RT may expand the therapeutic reach of immunotherapy. We examine the immunobiologic and clinical rationale for combining RT and immunotherapy, two modalities yet to be used in combination in routine practice. Preclinical data indicate that RT can potentiate the systemic efficacy of immunotherapy, while activation of the innate and adaptive immune system can enhance the local efficacy of RT.
DOI:10.1172/JCI69219      PMID:23863633      URL    
[本文引用:1]
[17] SHAVERDIAN N,LISBERG A E,BORNAZYAN K,et al.Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer:a secondary analysis of the KEYNOTE-001 phase 1 trial[J].Lancet Oncol,2017,18(7):895-903.
Preclinical studies have found radiotherapy enhances antitumour immune responses. We aimed to assess disease control and pulmonary toxicity in patients who previously received radiotherapy for non-small-cell lung cancer (NSCLC) before receiving pembrolizumab. We assessed patients with advanced NSCLC treated on the phase 1 KEYNOTE-001 trial at a single institution (University of California, Los Angeles, CA, USA). Patients were aged 18 years or older, had an Eastern Cooperative Oncology Group performance status of 1 or less, had adequate organ function, and no history of pneumonitis. Patients received pembrolizumab at a dose of either 2 mg/kg of bodyweight or 10 mg/kg every 3 weeks, or 10 mg/kg every 2 weeks, until disease progression, unacceptable toxicity, or other protocol-defined reasons for discontinuation. Disease response and pulmonary toxicity were prospectively assessed by Immune-related Response Criteria and Common Terminology Criteria for Adverse Events version 4.0. The primary objective of the KEYNOTE-001 trial was to assess the safety, side-effect profile, and antitumour activity of pembrolizumab. For our secondary analysis, patients were divided into subgroups to compare patients who previously received radiotherapy with patients who had not. Our primary objective was to determine whether previous radiotherapy affected progression-free survival, overall survival, and pulmonary toxicity in the intention-to-treat population. The KEYNOTE-001 trial was registered with ClinicalTrials.gov, number NCT01295827. Between May 22, 2012, and July 11, 2014, 98 patients were enrolled and received their first cycle of pembrolizumab. One patient was lost to follow-up. 42 (43%) of 97 patients had previously received any radiotherapy for the treatment of NSCLC before the first cycle of pembrolizumab. 38 (39%) of 97 patients received extracranial radiotherapy and 24 (25%) of 97 patients received thoracic radiotherapy. Median follow-up for surviving patients was 32·5 months (IQR 29·8-34·1). Progression-free survival with pembrolizumab was significantly longer in patients who previously received any radiotherapy than in patients without previous radiotherapy (hazard ratio [HR] 0·56 [95% CI 0·34-0·91], p=0·019; median progression-free survival 4·4 months [95% CI 2·1-8·6] vs 2·1 months [1·6-2·3]) and for patients who previously received extracranial radiotherapy compared with those without previous extracranial radiotherapy (HR 0·50 [0·30-0·84], p=0·0084; median progression-free survival 6·3 months [95% CI 2·1-10·4] vs 2·0 months [1·8-2·1]). Overall survival with pembrolizumab was significantly longer in patients who previously received any radiotherapy than in patients without previous radiotherapy (HR 0·58 [95% CI 0·36-0·94], p=0·026; median overall survival 10·7 months [95% CI 6·5-18·9] vs 5·3 months [2·7-7·7]) and for patients who previously received extracranial radiotherapy compared with those without previous extracranial radiotherapy (0·59 [95% CI 0·36-0·96], p=0·034; median overall survival 11·6 months [95% CI 6·5-20·5] vs 5·3 months [3·0-8·5]). 15 (63%) of 24 patients who had previously received thoracic radiotherapy had any recorded pulmonary toxicity versus 29 (40%) of 73 patients with no previous thoracic radiotherapy. Three (13%) patients with previous thoracic radiotherapy had treatment-related pulmonary toxicity compared with one (1%) of those without; frequency of grade 3 or worse treatment-related pulmonary toxicities was similar (one patient in each group). Our data suggest that previous treatment with radiotherapy in patients with advanced NSCLC results in longer progression-free survival and overall survival with pembrolizumab treatment than that seen in patients who did not have previous radiotherapy, with an acceptable safety profile. Further clinical trials investigating this combination are needed to determine the optimal treatment strategy for patients with advanced NSCLC. US National Institutes of Health.
DOI:10.1016/S1470-2045(17)30380-7      PMID:28551359      URL    
[本文引用:1]
[18] MESSENHEIMER D J,JENSEN S M,AFENTOULIS M E,et al.Timing of PD-1 blockade is critical to effective combination immunotherapy with Anti-OX40[J].Clin Cancer Res,2017,23(20):6165-6177.
Abstract Purpose: Antibodies specific for inhibitory checkpoints PD-1 and CTLA-4 have shown impressive results against solid tumors. This has fueled interest in novel immunotherapy combinations to affect patients who remain refractory to checkpoint blockade monotherapy. However, how to optimally combine checkpoint blockade with agents targeting T-cell costimulatory receptors, such as OX40, remains a critical question. Experimental Design: We utilized an anti-PD-1-refractory, orthotopically transplanted MMTV-PyMT mammary cancer model to investigate the antitumor effect of an agonist anti-OX40 antibody combined with anti-PD-1. As PD-1 naturally aids in immune contraction after T-cell activation, we treated mice with concurrent combination treatment versus sequentially administering anti-OX40 followed by anti-PD-1. Results: The concurrent addition of anti-PD-1 significantly attenuated the therapeutic effect of anti-OX40 alone. Combination-treated mice had considerable increases in type I and type II serum cytokines and significantly augmented expression of inhibitory receptors or exhaustion markers CTLA-4 and TIM-3 on T cells. Combination treatment increased intratumoral CD4 + T-cell proliferation at day 13, but at day 19, both CD4 + and CD8 + T-cell proliferation was significantly reduced compared with untreated mice. In two tumor models, sequential combination of anti-OX40 followed by anti-PD-1 (but not the reverse order) resulted in significant increases in therapeutic efficacy. Against MMTV-PyMT tumors, sequential combination was dependent on both CD4 + and CD8 + T cells and completely regressed tumors in approximately 30% of treated animals. Conclusions: These results highlight the importance of timing for optimized therapeutic effect with combination immunotherapies and suggest the testing of sequencing in combination immunotherapy clinical trials. Clin Cancer Res; 1-13. 2017 AACR. 2017 American Association for Cancer Research.
DOI:10.1158/1078-0432.CCR-16-2677      PMID:28855348      URL    
[本文引用:1]
[19] SHRIMALI R K,AHMAD S,VERMA V,et al.Concurrent PD-1 blockade negates the effects of OX40 agonist antibody in combination immunotherapy through Inducing T-cell apoptosis[J].Cancer Immunol Res,2017,5(9):755-766.
Abstract Combination therapies that depend on checkpoint inhibitor antibodies (Abs) such as for PD-1 or its ligand (PD-L1) together with immune stimulatory agonist Abs like anti-OX40 are being tested in the clinic to achieve improved antitumor effects. Here, we studied the potential therapeutic and immune effects of one such combination: Ab to PD-1 with agonist Ab to OX40/vaccine. We tested the antitumor effects of different treatment sequencing of this combination. We report that simultaneous addition of anti-PD-1 to anti-OX40 negated the antitumor effects of OX40 Ab. Antigen-specific CD8 + T-cell infiltration into the tumor was diminished, the resultant antitumor response weakened, and survival reduced. Although we observed an increase in IFN纬-producing E7-specifc CD8 + T cells in the spleens of mice treated with the combination of PD-1 blockade with anti-OX40/vaccine, these cells underwent apoptosis both in the periphery and the tumor. These results indicate that anti-PD-1 added at the initiation of therapy exhibits a detrimental effect on the positive outcome of anti-OX40 agonist Ab. These findings have important implications on the design of combination immunotherapy for cancer, demonstrating the need to test treatment combination and sequencing before moving to the clinic. Cancer Immunol Res; 5(9); 755-66. 2017 AACR . 2017 American Association for Cancer Research.
DOI:10.1158/2326-6066.CIR-17-0292      PMID:28848055      URL    
[本文引用:1]
[20] LYNCH T J,BONDARENKO I,LUFT A,et al.Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer:results from a randomized,double-blind,multicenter phase II study[J].J Clin Oncol,2012,30(17):2046-2054.
DOI:10.1200/JCO.2011.38.4032      URL    
[本文引用:1]
[21] GAINOR J F,SHAW A T,SEQUIST L V,et al.EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer:a retrospective analysis[J].Clin Cancer Res,2016,22(18):4585-4593.
PD-1 inhibitors are established agents in the management of non-small cell lung cancer (NSCLC); however, only a subset of patients derives clinical benefit. To determine the activity of PD-1/PD-L1 inhibitors within clinically relevant molecular subgroups, we retrospectively evaluated response patterns among EGFR-mutant, anaplastic lymphoma kinase (ALK)-positive, and EGFR wild-type/ALK-negative patients. We identified 58 patients treated with PD-1/PD-L1 inhibitors. Objective response rates (ORR) were assessed using RECIST v1.1. PD-L1 expression and CD8(+) tumor-infiltrating lymphocytes (TIL) were evaluated by IHC. Objective responses were observed in 1 of 28 (3.6%) EGFR-mutant or ALK-positive patients versus 7 of 30 (23.3%) EGFR wild-type and ALK-negative/unknown patients (P = 0.053). The ORR among never- or light- (≤10 pack years) smokers was 4.2% versus 20.6% among heavy smokers (P = 0.123). In an independent cohort of advanced EGFR-mutant (N = 68) and ALK-positive (N = 27) patients, PD-L1 expression was observed in 24%/16%/11% and 63%/47%/26% of pre-tyrosine kinase inhibitor (TKI) biopsies using cutoffs of ≥1%, ≥5%, and ≥50% tumor cell staining, respectively. Among EGFR-mutant patients with paired, pre- and post-TKI-resistant biopsies (N = 57), PD-L1 expression levels changed after resistance in 16 (28%) patients. Concurrent PD-L1 expression (≥5%) and high levels of CD8(+) TILs (grade ≥2) were observed in only 1 pretreatment (2.1%) and 5 resistant (11.6%) EGFR-mutant specimens and was not observed in any ALK-positive, pre- or post-TKI specimens. NSCLCs harboring EGFR mutations or ALK rearrangements are associated with low ORRs to PD-1/PD-L1 inhibitors. Low rates of concurrent PD-L1 expression and CD8(+) TILs within the tumor microenvironment may underlie these clinical observations. Clin Cancer Res; 22(18); 4585-93. 082016 AACRSee related commentary by Gettinger and Politi, p. 4539.
DOI:10.1158/1078-0432.CCR-15-3101      PMID:27225694      URL    
[本文引用:1]
[22] LEE C K,MAN J,LORD S,et al.Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer-a Meta-analysis[J].J Thorac Oncol,2017,12(2):403-407.
In EGFR- mutant advanced NSCLC, immune checkpoint inhibitors do not improve OS over that with docetaxel. Mechanisms of acquired resistance to first-line tyrosine kinase inhibitor therapy should be elucidated to guide selection of second-line treatment for these patients.
DOI:10.1016/j.jtho.2016.10.007      PMID:27765535      URL    
[本文引用:1]
[23] GONG J,WANG C,LEE P P,et al.Response to PD-1 blockade in microsatellite stable metastatic colorectal cancer harboring a POLE mutation[J].JNCCN,2017,15(2):142-147.
Recent clinical evidence has demonstrated that microsatellite instability (MSI) or defective mismatch repair (MMR) and high tumor mutational load can predict response to the programmed cell death 1 (PD-1) receptor inhibitor pembrolizumab in metastatic colorectal cancer (mCRC). Mutations in polymerase (), a DNA polymerase involved in DNA replication and repair, contribute to an ultramutated but microsatellite stable (MSS) phenotype in colorectal tumors that is uniquely distinct from MSI tumors. This report presents the first case in the literature describing a clinical response to pembrolizumab in an 81-year-old man with treatment-refractory mCRC characterized by an MSS phenotype andmutation identified on genomic profiling by next-generation sequencing. On tumor immunostaining, a large amount of CD8-positive tumor infiltrating lymphocytes (TILs) were present, with >90% of these expressing PD-1. More than 99% of PD-L1 expression was identified on nontumor cells in the tumor microenvironment that were close to the PD-1-positive CD8 TILs. mCRC tumors harboringmutations represent a hypermutated phenotype that may predict response to anti-PD-1 therapy.
DOI:10.6004/jnccn.2017.0016      PMID:28188185      URL    
[本文引用:1]
[24] ROUTY B,LE CHATELIER E,DEROSA L,et al.Gut microbiome influences efficacy of PD-1-based immu-notherapy against epithelial tumors[J].Science,2018,359(6371):91-97.
Abstract Immune checkpoint inhibitors (ICI) targeting the PD-1/PD-L1 axis induce sustained clinical responses in a sizeable minority of cancer patients. Here, we show that primary resistance to ICI can be due to abnormal gut microbiome composition. Antibiotics (ATB) inhibited the clinical benefit of ICI in patients with advanced cancer. Fecal microbiota transplantation (FMT) from cancer patients who responded to ICI (but not from non-responding patients) into germ-free or ATB-treated mice ameliorated the antitumor effects of PD-1 blockade. Metagenomics of patient stools at diagnosis revealed correlations between clinical responses to ICI and the relative abundance of Akkermansia muciniphila Oral supplementation with A. muciniphila post-FMT with non-responder feces restored the efficacy of PD-1 blockade in an IL-12-dependent manner, by increasing the recruitment of CCR9 + CXCR3 + CD4 + T lymphocytes into tumor beds. Copyright 2017, American Association for the Advancement of Science.
DOI:10.1126/science.aan3706      PMID:29097494      URL    
[本文引用:1]
[25] GOPALAKRISHNAN V,SPENCER C N,NEZI L,et al.Gut microbiome modulates response to anti-PD-1 immu-notherapy in melanoma patients[J].Science,2018,359(6371):97-103.
Abstract Pre-clinical mouse models suggest that the gut microbiome modulates tumor response to checkpoint blockade immunotherapy; however, this has not been well-characterized in human cancer patients. Here we examined the oral and gut microbiome of melanoma patients undergoing anti-PD-1 immunotherapy (n=112). Significant differences were observed in the diversity and composition of the patient gut microbiome of responders (R) versus non-responders (NR). Analysis of patient fecal microbiome samples (n=43, 30R, 13NR) showed significantly higher alpha diversity (p<0.01) and relative abundance of Ruminococcaceae bacteria (p<0.01) in responding patients. Metagenomic studies revealed functional differences in gut bacteria in R including enrichment of anabolic pathways. Immune profiling suggested enhanced systemic and anti-tumor immunity in responding patients with a favorable gut microbiome, as well as in germ-free mice receiving fecal transplants from responding patients. Together, these data have important implications for the treatment of melanoma patients with immune checkpoint inhibitors. Copyright 2017, American Association for the Advancement of Science.
DOI:10.1126/science.aan4236      PMID:29097493      URL    
[本文引用:1]
资源
PDF下载数    
RichHTML 浏览数    
摘要点击数    

分享
导出

相关文章:
关键词(key words)
非小细胞
免疫治疗
程序性死亡蛋白-1
预测标志物

Cancer
lung
non-small cell
Immunotherapy
Programmed cell death pro...
Predictive markers

作者
王俊
王宝成

WANG Jun
WANG Baocheng