寡转移性NSCLC全身联合局部治疗的临床研究进展
Progress of Clinical Research on Systemic Combined Local Treatment for Oligometastatic NSCLC
DOI: 10.12677/ACM.2023.1391938, PDF, HTML, XML, 下载: 129  浏览: 237 
作者: 刘润莎:西安医学院研究生处,陕西 西安;张燕军*:陕西省肿瘤医院内三科,陕西 西安
关键词: 非小细胞肺癌寡转移局部治疗系统治疗Non-Small Cell Lung Cancer Oligometastasis Local Therapy Systemic Therapy
摘要: 寡转移非小细胞肺癌是IV期非小细胞肺癌患者发生有限的部位和数量的转移,可能比广泛转移性具有更惰性的生物学特性,其预后比广泛性转移要好。寡转移可能受益于局部积极治疗,但目前关于寡转移性NSCLC针对转移灶治疗(metastasis directed therapy, MDT)的专家共识,缺少随机对照III期临床试验等高级别的循证医学证据。本文拟从肿瘤内科的角度,就寡转移性NSCLC不同系统治疗联合MDT的临床研究现状、影响因素作一系统回顾。
Abstract: Oligometastatic non-small cell lung cancer is stage IV non-small cell lung cancer with limited site and number of metastases that may have a more inert biology than extensive metastasis and a bet-ter prognosis than extensive metastasis. Oligometastasis may benefit from locally positive treat-ment, but the current expert consensus on metastasis directed therapy (MDT) for oligometastasis in NSCLC lacks high-level evidence-based medical evidence such as randomized controlled phase III clinical trials. From the perspective of medical oncology, this paper reviews the clinical research status and influencing factors of different systems of treatment combined with MDT for oligometa-static NSCLC.
文章引用:刘润莎, 张燕军. 寡转移性NSCLC全身联合局部治疗的临床研究进展[J]. 临床医学进展, 2023, 13(9): 13863-13873. https://doi.org/10.12677/ACM.2023.1391938

1. 背景

非小细胞肺癌(non-small cell lung cancer, NSCLC)占全部肺癌的85%~90%,诊断时早期(I期~II期)占17%,局部中晚期(III期)占22%,晚期(IV期)占57% ‎[1] 。对于III期不可手术患者,同步或序贯放化疗是标准治疗,但增加放疗剂量和延长化疗周期不改善总生存期(Overall Survival, OS) ‎[2] 。对于IV期驱动基因阴性患者,维持化疗和抗血管生成药物无进展生存期(Progression-Free Survival, PFS) (9.2月v 6.5月)和OS (24.3月v 17.2月),可见临床获益有限 ‎[3] ‎[4] ‎[5] 。回顾性研究显示 ‎[6] ‎[7] ,转移性NSCLC,同时性寡转移占16.1%,异时性寡转移占36.6%,广泛性转移占45.2%。转移性NSCLC一线全身治疗后,大多数进展事件只发生在基线时存在的病灶,而不是新病灶 ‎[8] 。因此,最大限度控制基线时存在的病灶就有可能延长PFS,进而改善OS。

1968年由Philip Rubin和Jerold Green提出恶性肿瘤“孤立性转移”的概念,并支持进行积极治疗的策略;1995年S. Hellman和R. R. Weichselbaum提出“寡转移假说(Oligometastatic Disease, OMD)”,这是一种介于局部疾病和广泛转移之间的癌症扩散的中间状态,可以受到局部治疗的影响 ‎[9] ‎[10] ‎[11] 。2020年,ESTRO和EORTC形成专家共识,定义寡转移性肺癌为最多3个器官的5个病变,分为新生性、重复性和诱导性寡转移等3种类型 ‎[12] ,每种类型又根据既往是否存在寡转移和进行治疗等情况,分别分为3种亚型,同时性寡转移、异时性寡复发和寡进展,重复性寡复发、寡进展和寡持续,诱导性寡复发、寡进展和寡持续等总共9种亚型。同年,ESTRO和ASTRO就OMD的疾病特征、转移灶的负荷、疾病进展时间和既往治疗等15个关键问题形成共识 ‎[13] 。2023年,ESTRO和ASTRO发布寡转移性NSCLC针对转移灶治疗(metastasis directed therapy, MDT)的专家共识 ‎[14] ,鉴于缺少随机对照III期临床试验等高级别的循证医学证据,对于最多有5个远处转移的NSCLC患者,强烈推荐基于患者为中心的多学科团队意见、MDT作为多模式治疗方法的组成部分,并评估对所有转移灶MDT的技术可行性和临床安全性;有条件推荐MDT同时性寡转移、异时性寡复发、诱导性寡持续和诱导性进展的NSCLC。2023年,EORTC和ESTRO同样基于低级别循证医学证据,发布免疫检查点抑制剂和靶向治疗等系统治疗联合MDT的专家共识 ‎[15] 。

目前,最新的CSCO指南仅对孤立性的脑、骨和肾上腺转移患者推荐包括手术和放疗在内的一线局部治疗。本文拟从肿瘤内科的角度,就不同系统治疗联合MDT的临床研究现状、影响因素作一系统回顾。

2. 治疗现状

2.1. 回顾性研究

2.1.1. 主要纳入人群

多数研究纳入转移灶 ≤ 3~5个的同时性或异时性寡转移性NSCLC,且全部病灶均可接受局部治疗 ‎[16] - ‎[22] ,部分研究要求局限于肺部 ‎[19] ‎[20] ‎[21] ‎[23] ,单个器官转移灶 ≤ 2~3个 ‎[17] ‎[18] ‎[24] ,最大病灶 ≤ 5.0 cm ‎[21] ‎[25] 。个别研究纳入最大病灶 ≤ 6.5 cm ‎[22] 和≤7.5 cm ‎[26] ,或纳入高龄、心肺功能差、拒绝手术或放疗,或者远处转移无法手术切除或放疗 ‎[27] ,要求针对寡转移病灶经4~6周期化疗 ‎[25] 或一代EGFR-TKI治疗2月未进展 ‎[17] ,寡转移病灶距肺门 ≥ 1 cm ‎[26] 的患者。也有研究将胸内淋巴结转移计为1个部位 ‎[18] ,肺原发灶不纳入寡转移灶数计算 ‎[17] 。一些研究排除完成根治性局部治疗前进展 ‎[16] ,临近肺门、大支气管或大肺血管(≤1.0 cm),出血性疾病对药物治疗无效 ‎[25] ,MWA后随访<6个月(不包括死亡病例),存在第二原发性肺癌和组织学上未经证实的肺内GGO ‎[19] ,术后<6月复发转移 ‎[23] ,有症状的CNS转移 ‎[21] ‎[22] ‎[24] 等的患者。

2.1.2. 主要治疗模式

局部治疗联合系统治疗旨在分别控制,多数研究采用先局部治疗后根据病情需要系统治疗 ‎[16] ‎[18] - ‎[24] ‎[26] ‎[27] ,局部治疗包括手术、各种形式的放疗和消融,要求局部治疗达到根治性目的,覆盖全部寡转移病灶。系统治疗包括化疗、靶向和免疫治疗。也有研究采用先用含铂双药化疗4~6周期未进展在RFA ‎[25] 和一代EGFR-TKI 2月未进展的患者再行手术或放疗等局部治疗 ‎[17] 的模式。

2.1.3. 主要效果结果

Table 1. Main results of curative effect after local treatment or systemic treatment

表1. 局部治疗后或行系统治疗的主要疗效结果

主要效果结果如表1表2分别摘要了先局部治疗再根据病情需要进行系统治疗和先系统治疗2~3月病情未进展时再局部治疗并系统治疗维持的主要疗效结果,全部研究的无疾病进展生存时间(progression free survival, PFS)和总生存期总生存时间(overall survival, OS),以及1年、2年和3年的PFS率及OS率均显著高于历史对照证实了局部治疗的临床获益。部分研究还报告了至局部进展时间(time to local progression, TTLP) ‎[26] ‎[27] ,均取得很好结果。也有研究设置了微波消融(microwave ablation, MWA)联合化疗组和单独化疗作为对照组 ‎[22] ,结果两组的PFS分别8.8和5.8月(HR 0.357, P = 0.007),OS分别27.7月和20.0月(HR 0.238, P = 0.0002)。当以手术或放疗作为局部治疗对比时 ‎[16] ,两组的OS分别23.4和OS 55.2月,1、3、和5年的OS率分别74.3%、35.3%、24.2%和1、3和5年OS 97.1%、58.7%和34.3%,两组间局部或全身进展以及累积死亡风险均无差异。还有研究报告了局部消融(local ablative therapy, LAT)对残留病灶的覆盖范围 ‎[17] ,分为全部消融(原发灶和全部转移灶全部覆盖)组、部分消融组(近消融原发灶或转移灶)组和未消融(原发灶和全部转移灶均不消融)组,结果3组的PFS分别20.6、15.6和13.9月(P < 0.001),OS分别40.9、34.1和30.8月(P < 0.001),全部病灶组与部分病灶或未行LAT组的PFS和OS差异显著,部分病灶组与未行LAT组的PFS和OS没有差异,提示全部病灶的局部治疗至关重要。局部治疗后,原位或局部进展率4.5%~24%,而全身或新病灶进展率高达43%~63.1%,提示局部治疗后治疗是薄弱的,主要模式是全身或新病灶进展。

Table 2. Main curative effect results of local therapy followed by systemic maintenance after systemic treatment

表2. 系统治疗后局部治疗再系统治疗维持的主要疗效结果

在预后因素方面,比较一致或符合一般临床认知的结论包括:腺癌、不吸烟、N0-1或临床I期、Del 19、1个转移灶、后续治疗、完全消融和原发灶手术或消融等与更好的PFS或OS相关 ‎[16] ‎[17] ‎[19] ‎[21] ‎[22] ‎[25] ,再次进展和远处转移是OS不好的因素 ‎[19] ,手术和放疗组间局部或全身进展以及累积死亡风险均无差异 ‎[18] ,异时性转移是OS好的因素 ‎[21] ,包括肺内孤立性转移与孤立性复发 ‎[23] 。也有相互冲突或不符合临床认知的结果,如Hiroshi Kodama ‎[20] 结果是肿瘤 ≤ 3.0 cm是OS好的因素,而Helmut Schoellnast ‎[26] 和刘宝东 ‎[23] 结果是肿瘤 ≤ 3 cm或>3 cm与OS或PFS无关;肿瘤复发时间 ≤ 36月与>36月之间的OS均无差异 ‎[23] ,存在差异的原因可能是人群选择、病理分期、组织学类型、局部治疗与系统治疗顺序以及病例数有关。

2.1.4. 主要毒性反应

有2例治疗相关性死亡,1例是Helmut Schoellnast ‎[20] 报告,1/33例(3%) RFA后30天内死亡病例,该患者手术后立即出现呼吸窘迫,有全肺切除术、放疗史和潜在的严重慢性阻塞性肺疾病;拒绝插管和机械通气,放弃救治,消融后7天死于进行性呼吸窘迫。另1例是Kyle G. Mitchell ‎[18] 报道,1/63例(1.6%)肺部病灶放疗60 Gray后,90天内死亡。其余研究的局部治疗,包括手术和各种形式的放疗及消融治疗,均未出现治疗中及治疗后90天内死亡。射频消融术(radiofrequency ablation, RFA)或微波消融(microwave ablation, MWA)主要毒性反应是术中局部发热、出汗、心率加快,无需特殊处理,术中疼痛9.1%~40%,镇痛或者降低靶温度到70℃等对症治疗好转 ‎[17] ‎[23] ‎[24] ;消融后综合征 ‎[19] ‎[21] ‎[22] ‎[23] ‎[24] ‎[25] ,包括发热(低于38.5℃)、疲劳、全身不适、恶心和呕吐,发生率15.4%~36.7%,对症治疗或自行恢复。需引流的症状性气胸或支气管胸膜瘘 ‎[17] ‎[19] ‎[20] ‎[21] ‎[22] ‎[24] ‎[25] ‎[26] ‎[27] 0.7%~29.0%,需要手术胸膜缝合1/55人次(1.8%)、胸膜硬化2/55人次(3.6%) ‎[20] 。需要引流的胸水 ‎[21] ‎[22] ‎[24] 1.4%~25.0%。自限性肺出血/咯血 ‎[19] ‎[20] ‎[21] ‎[22] ‎[27] 3.0%~29.5%,或经常规止血痊愈。肺炎 ‎[21] 7/44例(15.9%),浸润性肺曲霉病 ‎[19] 2/143例次(1.4%),均经抗感染治疗痊愈。心血管疾病 ‎[21] 3/44 (6.8%),对症治疗痊愈。1/33例消融后当日心衰,经过纠正后好转 ‎[27] 。44例51个病灶的55次消融,≥3级不良事件发生率 ‎[20] 5.5%,44例消融出现合并症55.7%,65岁及以上与65岁以下相比,更容易发生并发症(68.4%和43.9%,P = 0.028) ‎[21] 。Qinghua Xu ‎[17] 报道,145例放疗患者中,≥3级的放射性肺炎7.7%、放射性食管炎16.9%;无论是否对肺病变进行LAT,均未观察到EGFR-TKI相关的间质性肺疾病,1例放射性肺炎停用吉非替尼。其余研究未报告局部治疗加重化疗、靶向和免疫治疗的相关毒性反应。

2.2. 随机对照研究

2.2.1. 先系统治疗后联合或不联合局部治疗再系统治疗

Puneeth Iyengar ‎[28] 纳入驱动基因阴性NSCLC,≤6个颅外病灶(包括原发灶),肝脏或肺部病灶 ≤ 3个,先经一线含铂双药化疗4~6个周期未进展病例29例,1:1随机分为试验组和对照组,试验组全部病灶SAbR后观察或维持化疗,对照组观察或维持化疗。PFS分别9.7和3.5月(HR 0.304, P = 0.01),OS分别NR和12.0月(去交叉病例)/17.0月(含交叉病例) (P > 0.05)。试验组4/14进展,无原病灶处进展,对照组10/15进展,7例原病灶处进展。PFS和OS获益不受入组前接受治疗的脑转移存在与否或颅外转移灶的数量(≤2与>2个)的影响。Daniel R Gomez ‎[29] 纳入45例 ≤ 3个转移病灶(不包括原发灶)和4~6个周期的一线铂类化疗或靶向治疗 ≥ 3月未进展的NSCLC患者,1:1随机分为试验组和对照组,试验组全部病灶SAbR后靶向或观察或维持化疗,对照组靶向或观察或维持化疗。对照组17例进展病例11例交叉到试验组。OS分别41.2和17.0月(P = 0.017)。试验组与对照组接受晚期LCT的患者比例无差异(P = 0.39),两组进展后存活时间分别37.6和9.4月(P = 0.034);39例进展患者中,15例(41%;试验组6例,对照组9例)接受了晚期LCT,进展时接受晚期LCT的中位OS时间NR,未接受晚期LCT的是16.4月(P = 0.119)。LCT与OS改善相关,初始LCT与未LCT组的HR 0.30,晚期LCT与未晚期LCT组的HR 0.44 (P = 0.064),只有LCT与OS相关(HR 0.46, P = 0.048) ‎[30] 。

SINDAS是一项一代EGFR-TKI联合或不联合放疗治疗同时性寡转移性NSCLC的随机对照III期临床研究 ‎[31] ,纳入133例既往未经治疗,≤5个转移灶(包括引流区域外淋巴结,不包括引流区域内淋巴结转移),单个器官病灶 ≤ 2个的EGFR经典突变的非鳞状NSCLC,1:1随机分为试验组和对照组,分别采用一代EGFR-TKI联合全部病灶放疗或单独一代EGFR-TKI治疗。局部控制率分别55.4%和91.2% (P < 0.001),PFS12.5和20.2月(HR 0.22, P < 0.001),OS 17.6和25.5月(HR 0.44, P < 0.001)。ECOG 0分(P < 0.02)、1~2个转移灶(P < 0.001)和RT (P < 0.001)是PFS更好的预后因素,ECOG 0分(P < 0.02)、T1~2分期(P < 0.02)、1~2个转移(P < 0.001)、Del 19 (P < 0.001)和RT (P < 0.001)是OS更好的预后因素。

2.2.2. 局部治疗后或同时联合免疫治疗

Willemijn S M E Theelen ‎[32] 汇总了PEMBRO-RT研究和随机对照的1/2期临床试验MDACC的数据,前者的纳入人群和治疗模式同前,后者纳入初诊的寡转移性NSCLC患者,1:1随机分为试验组和对照组,分别采用SBRT的同时Pembrolizumab或单独Pembrolizumab治疗。总计148例寡转移性患者,两组最佳的放疗野外RR (abscopal response rate, ARR)分别41.7%和19.7% (OR 2.96, P = 0.0039),最佳放疗野外DCR (abscopal disease control rate, ACR) 65.3%和43.4% (OR 2.51, P = 0.0071),12周ACR率分别62.5%和38.2% (OR 2.71, P = 0.0033)。PFS 9.0和4.4月(HR 0.67, P = 0.045),OS 19.2和8.7月(HR 0.67, P = 0.0004)。不吸烟和50 Gy/4 f是PFS更好的预后因素,没有与OS相关的预后因素。高级别的放疗相关不良事件非常罕见,放疗联合Pembrolizumab没有引起新的安全问题。

PEMBRO-RT研究是1项随机对照的2期临床试验 ‎[33] ,纳入既往标准方案化疗进展的76寡转移性NSCLC,至少有1个可测量病灶不进行放疗,1:1随机分为试验组和对照组,分别采用立体定向放疗(Stereotactic body radiotherapy, SBRT)后Pembrolizumab或单独Pembrolizumab治疗。最佳的ORR 47.2% (17/36)和22.5% (9/40);12周的ORR分别36% (13/36)和18% (7/40) (P = 0.07),PD-L1阴性人群的ORR 22% (4/18)和4% (1/25) (P = 0.14),而PD-L阳性人群的ORR相似;12周的DCR 64% (23/36)和40% (16/40) (P = 0.04)。SBRT对之前接受过放疗的患者和从未接受过放疗的患者的缓解率的影响相似(OR 3.1 vs 2.4, P = 0.81),提示既往放疗对研究结果影响不大。PFS分别1.9和6.6月(HR 0.71, P = 0.19),SBRT的PFS获益是在PD-L1阴性人群(HR 0.49, P = 0.03),增加SBRT在PD-L1阳性人群没有PFS获益(HR 1.14, P = 0.79);OS 7.6和15.9月(HR 0.66, P = 0.16),OS只在PD-L1阴性人群获益(HR 0.48, P = 0.046),在PD-L1阳性人群没有获益(HR 1.4, P = 0.58),PD-L1是试验组OS的预测因素。Pembrolizumab相关的毒性主要是疲乏(18%),流感样症状(15%)和瘙痒(14%)。2组间3~5级pembrolizumab相关毒性没有显著差异。

系统治疗后联合局部治疗或者局部治疗联合系统治疗的随机对照研究试验效果如表3

Table 3. Results of randomized controlled trials of local therapy combined with systemic therapy

表3. 局部治疗联合系统治疗的随机对照试验治疗效果

3. 结语与展望

寡转移的概念从人群细分角度引发了一系列临床和转化研究,为转移性NSCLC患者的诊断、治疗和预后改善带来了新的挑战和机遇。针对不同的病种,NSCLC和前列腺癌的寡转移研究走在前列 ‎[36] ,已经由回顾性研究进入到前瞻性、随机、对照临床研究阶段。局部治疗联合化疗或靶向治疗旨在改善远期疗效,回顾性研究主要纳入单纯的寡转移人群,多数采取先局部治疗再系统治疗的模式;随机对照性研究更加细化人群,纳入化疗或靶向治疗2~3月内未进展病例,之后再局部治疗,并根据病情采用适当的系统治疗。目前,对于寡转移性NSCLC局部治疗联合系统治疗的研究 ‎[37] ‎[38] ,虽然不同在人群选择和治疗模式上存在差异,初步的结果显示可以改善PFS和OS,并不增加系统治疗的毒性反应,期待随机对照的3期临床试验结果。

目前,手术、各种形式的放疗和消融治疗已能对局部病灶进行满意的控制,未来研究的重点是防治出现新发病灶。随着生物标志物的不断开发,靶向治疗和免疫治疗更加有效,再引入较CT和MRI更为灵敏和特异性的PET-CT和ct-DNA和MRD等生物标志物进行人群筛选和检测疗效,或将进一步改善寡转移性NSCLC的治疗格局。

NOTES

*通讯作者。

参考文献

[1] Siegel, R.L., Miller, K.D., Fuchs, H.E., et al. (2021) Cancer Statistics, 2021. CA: A Cancer Journal for Clinicians, 71, 7-33.
https://doi.org/10.3322/caac.21654
[2] Chen, L., Hou, Y., Xia, Y., et al. (2020) Radiotherapy Dose and In-duction Chemotherapy Cycles Are Associated with Prognosis and Toxicity Risk: A Retrospective Study of 227 Patients with Unresectable Stage III Non-Small-Cell Lung Cancer. Technology in Cancer Research & Treatment, 19, 1-8.
https://doi.org/10.1177/1533033820951802
[3] Zhou, C., Wu, Y., Chen, G., et al. (2015) BEYOND: A Ran-domized, Double-Blind, Placebo-Controlled, Multicenter, Phase III Study of First-Line Carboplatin/Paclitaxel Plus Bevacizumab or Placebo in Chinese Patients with Advanced or Recurrent Nonsquamous Non-Small-Cell Lung Cancer. Journal of Clinical Oncology, 33, 2197-2204.
https://doi.org/10.1200/JCO.2014.59.4424
[4] Gridelli, C., Maione, P. and Rossi, A. (2013) The PARAMOUNT Trial: A Phase III Randomized Study of Maintenance Pemetrexed versus Placebo Immediately Following Induction First-Line Treatment with Pemetrexed plus Cisplatin for Advanced Nonsquamous Non-Small Cell Lung Cancer. Reviews on Recent Clinical Trials, 8, 23-28.
https://doi.org/10.2174/15748871112079990040
[5] Lu, J., Chu, T., Liu, H., et al. (2022) Equivalent Efficacy Assessment of QL1101 and Bevacizumab in Nonsquamous Non-Small Cell Lung Cancer Patients: A Two-Year Fol-low-Up Data Update. Chinese Journal of Cancer Research, 34, 28-39.
https://doi.org/10.21147/j.issn.1000-9604.2022.01.03
[6] Christ, S.M., Pohl, K., Muehlematter, U.J., et al. (2022) Imaging-Based Prevalence of Oligometastatic Disease: A Single-Center Cross-Sectional Study. International Journal of Radiation Oncology Biology Physics, 114, 596-602.
https://doi.org/10.1016/j.ijrobp.2022.06.100
[7] Torok, J.A., Gu, L., Tandberg, D.J., et al. (2017) Patterns of Distant Metastases after Surgical Management of Non- Small-Cell Lung Cancer. Clinical Lung Cancer, 18, e57-e70.
https://doi.org/10.1016/j.cllc.2016.06.011
[8] Rusthoven, K.E., Hammerman, S.F., Kavanagh, B.D., et al. (2009) Is There a Role for Consolidative Stereotactic Body Radiation Therapy Following First-Line Systemic Therapy for Meta-static Lung cancer? A Patterns-of-Failure Analysis. Acta Oncologica, 48, 578-583.
https://doi.org/10.1080/02841860802662722
[9] Tran, P.T., Sutera, P., Phillips, R.M., et al. (2022) From Idea to Clinical Practice: A Brief History of Oligometastatic Disease. International Journal of Radiation Oncology, Biology, Physics, 114, 576-580.
https://doi.org/10.1016/j.ijrobp.2022.04.004
[10] Milano, M.T., Biswas, T., Simone, C.N., et al. (2021) Oligome-tastases: History of a Hypothesis. Annals of Palliative Medicine, 10, 5923-5930.
https://doi.org/10.21037/apm.2020.03.31
[11] Gutiontov, S.I., Pitroda, S.P., Tran, P.T., et al. (2021) (Oli-go)metastasis as a Spectrum of Disease. Cancer Research, 81, 2577-2583.
https://doi.org/10.1158/0008-5472.CAN-20-3337
[12] Guckenberger, M., Lievens, Y., Bouma, A.B., et al. (2020) Characterisation and Classification of Oligometastatic Disease: A European Society for Radiotherapy and Oncology and European Organisation for Research and Treatment of Cancer Consensus Recommendation. The Lancet Oncology, 21, e18-e28.
https://doi.org/10.1016/S1470-2045(19)30718-1
[13] Lievens, Y., Guckenberger, M., Gomez, D., et al. (2020) Defining Oligometastatic Disease from a Radiation Oncology Perspective: An ESTRO-ASTRO Consensus Document. Radiotherapy and Oncology, 148, 157-166.
https://doi.org/10.1016/j.radonc.2020.04.003
[14] Iyengar, P., All, S., Berry, M.F., et al. (2023) Treatment of Oli-gometastatic Non-Small Cell Lung Cancer: An ASTRO/ ESTRO Clinical Practice Guideline. Practical Radiation Oncol-ogy.
https://doi.org/10.1016/j.prro.2023.04.004
[15] Kroeze, S.G.C., Pavic, M., Stellamans, K., et al. (2023) Me-tastases-Directed Stereotactic Body Radiotherapy in Combination with Targeted Therapy or Immunotherapy: Systematic Review and Consensus Recommendations by the EORTC-ESTRO OligoCare Consortium. The Lancet Oncology, 24, e121-e132.
https://doi.org/10.1016/S1470-2045(22)00752-5
[16] Kwint, M., Walraven, I., Burgers, S., et al. (2017) Outcome of Radical Local Treatment of Non-Small Cell Lung Cancer Patients with Synchronous Oligometastases. Lung Cancer (Amsterdam, Netherlands), 112, 134-139.
https://doi.org/10.1016/j.lungcan.2017.08.006
[17] Xu, Q., Zhou, F., Liu, H., et al. (2018) Consolidative Local Ablative Therapy Improves the Survival of Patients with Synchronous Oligometastatic NSCLC Harboring EGFR Acti-vating Mutation Treated with First-Line EGFR-TKIs. Journal of Thoracic Oncology, 13, 1383-1392.
https://doi.org/10.1016/j.jtho.2018.05.019
[18] Mitchell, K.G., Farooqi, A., Ludmir, E.B., et al. (2021) Pulmonary Resection Is Associated with Long-Term Survival and Should Remain a Therapeutic Option in Oligometastatic Lung Cancer. The Journal of Thoracic and Cardiovascular Surgery, 161, 1497-1504.
https://doi.org/10.1016/j.jtcvs.2020.02.134
[19] Ni, Y., Peng, J., Yang, X., et al. (2021) Multicentre Study of Mi-crowave Ablation for Pulmonary Oligorecurrence after Radical Resection of Non-Small-Cell Lung Cancer. British Jour-nal of Cancer, 125, 672-678.
https://doi.org/10.1038/s41416-021-01404-y
[20] Kodama, H., Yamakado, K., Takaki, H., et al. (2012) Lung Ra-diofrequency Ablation for the Treatment of Unresectable Recurrent Non-Small-Cell Lung Cancer after Surgical Interven-tion. CardioVascular and Interventional Radiology, 35, 563-569.
https://doi.org/10.1007/s00270-011-0220-0
[21] Wei, Z., Ye, X., Yang, X., et al. (2019) Efficacy and Safety of Microwave Ablation in the Treatment of Patients with Oligometastatic Non-Small-Cell Lung Cancer: A Retrospective Study. International Journal of Hyperthermia, 36, 826-833.
https://doi.org/10.1080/02656736.2019.1642522
[22] Ni, Y., Bi, J., Ye, X., et al. (2016) Local Microwave Abla-tion with Continued EGFR Tyrosine Kinase Inhibitor as a Treatment Strategy in Advanced Non-Small Cell Lung Cancers That Developed Extra-Central Nervous System Oligoprogressive Disease during EGFR Tyrosine Kinase Inhibitor Treatment: A Pilot Study. Medicine (Baltimore), 95, e3998.
https://doi.org/10.1097/MD.0000000000003998
[23] 刘宝东, 刘磊, 胡牧, 等. 肺癌切除术后肺内孤立性转移复发灶的射频消融治疗[J]. 中国肺癌杂志, 2014, 17(6): 460-464.
[24] Ni, Y., Liu, B., Ye, X., et al. (2019) Local Thermal Ablation with Continuous EGFR Tyrosine Kinase In-hibitors for EGFR-Mutant Non-Small Cell Lung Cancers that Developed Extra-Central Nervous System (CNS) Oli-goprogressive Disease. Cardiovascular and Interventional Radiology, 42, 693-699.
https://doi.org/10.1007/s00270-018-02153-x
[25] Li, X., Zhao, M., Wang, J., et al. (2013) Percutaneous CT-Guided Radiofrequency Ablation as Supplemental Therapy after Systemic Chemotherapy for Selected Advanced Non-Small Cell Lung Cancers. American Journal of Roentgenology (1976), 201, 1362.
https://doi.org/10.2214/AJR.12.10511
[26] Schoellnast, H., Deodhar, A., Hsu, M., et al. (2012) Recurrent Non-Small Cell Lung Cancer: Evaluation of CT-Guided Radiofrequency Ablation as Salvage Therapy. Acta Radiologica (1987), 53, 893-899.
https://doi.org/10.1258/ar.2012.110333
[27] 刘宝东, 李元博, 胡牧, 等. 射频消融在EGFR-TKIs治疗非小细胞肺癌后局部进展的初步临床应用[J]. 中国肺癌杂志, 2016, 19(12): 859-863.
[28] Iyengar, P., Wardak, Z., Gerber, D.E., et al. (2018) Consolidative Radiotherapy for Limited Metastatic Non-Small-Cell Lung Cancer: A Phase 2 Random-ized Clinical Trial. JAMA Oncology, 4, e173501.
https://doi.org/10.1001/jamaoncol.2017.3501
[29] Gomez, D.R., Blumenschein, G.R., Lee, J.J., et al. (2016) Local Consolidative Therapy versus Maintenance Therapy or Observation for Patients with Oligometastatic Non-Small-Cell Lung Cancer without Progression after First-Line Systemic Therapy: A Multicentre, Randomised, Controlled, Phase 2 Study. The Lancet Oncology, 17, 1672-1682.
https://doi.org/10.1016/S1470-2045(16)30532-0
[30] Gomez, D.R., Tang, C., Zhang, J., et al. (2019) Local Con-solidative Therapy vs. Maintenance Therapy or Observation for Patients with Oligometastatic Non-Small-Cell Lung Cancer: Long-Term Results of a Multi-Institutional, Phase II, Randomized Study. Journal of Clinical Oncology, 37, 1558-1565.
https://doi.org/10.1200/JCO.19.00201
[31] Wang, X., Bai, Y., Verma, V., et al. (2023) Randomized Trial of First-Line Tyrosine Kinase Inhibitor with or without Radiotherapy for Synchronous Oligometastatic EGFR-Mutated Non-Small Cell Lung Cancer. JNCI: Journal of the National Cancer Institute, 115, 742-748.
https://doi.org/10.1093/jnci/djac015
[32] Theelen, W.S.M.E., Chen, D., Verma, V., et al. (2021) Pembrolizumab with or without Radiotherapy for Metastatic Non-Small-Cell Lung Cancer: A Pooled Analysis of Two Randomised Trials. The Lancet Respiratory Medicine, 9, 467-475.
https://doi.org/10.1016/S2213-2600(20)30391-X
[33] Theelen, W., Peulen, H., Lalezari, F., et al. (2019) Effect of Pembrolizumab after Stereotactic Body Radiotherapy vs Pembrolizumab Alone on Tumor Response in Patients with Advanced Non-Small Cell Lung Cancer: Results of the PEMBRO-RT Phase 2 Randomized Clinical Trial. JAMA Oncology, 5, 1276-1282.
https://doi.org/10.1001/jamaoncol.2019.1478
[34] Petrelli, F., Ghidini, A., Cabiddu, M., et al. (2018) Addition of Radiotherapy to the Primary Tumour in Oligometastatic NSCLC: A Systematic Review and Meta-Analysis. Lung Cancer (Amsterdam, Netherlands), 126, 194-200.
https://doi.org/10.1016/j.lungcan.2018.11.017
[35] Bauml, J.M., Mick, R., Ciunci, C., et al. (2019) Pembrolizumab after Completion of Locally Ablative Therapy for Oligometastatic Non-Small Cell Lung Cancer: A Phase 2 Trial. JAMA Oncology, 5, 1283-1290.
https://doi.org/10.1001/jamaoncol.2019.1449
[36] Katipally, R.R., Pitroda, S.P., Juloori, A., et al. (2022) The Oli-gometastatic Spectrum in the Era of Improved Detection and Modern Systemic Therapy. Nature Reviews. Clinical On-cology, 19, 585-599.
https://doi.org/10.1038/s41571-022-00655-9
[37] Garde-Noguera, J., Martin-Martin, M., Obeso, A., et al. (2022) Current Treatment Landscape for Oligometastatic Non-Small Cell Lung Cancer. World Journal of Clinical Oncology, 13, 485-495.
https://doi.org/10.5306/wjco.v13.i6.485
[38] Jasper, K., Stiles, B., McDonald, F., et al. (2022) Practical Manage-ment of Oligometastatic Non-Small-Cell Lung Cancer. Journal of Clinical Oncology, 40, 635-641.
https://doi.org/10.1200/JCO.21.01719