综述
结直肠癌肝转移转化治疗的研究进展
中华胃肠外科杂志, 2021,24(1) : 85-93. DOI: 10.3760/cma.j.cn.441530-20200311-00135
摘要

结直肠癌患者在全病程中发生肝转移的概率达40%~50%,肝转移是影响结直肠癌患者长期预后的重要不利因素。手术切除肝转移灶是唯一可能达到近似根治效果的治疗选择。对于判断为不可切除的肝转移灶,经过综合治疗,使肿瘤缩小,进而将初始不可切除病灶转化为可切除病灶,称为转化治疗。转化治疗可分为以化疗±靶向为主的系统治疗及局部治疗。本文重点综述近年来结直肠癌肝转移转化治疗相关研究成果:(1)梳理肝转移癌手术可切除性评估标准;(2)探讨疗效评估、手术时机及肿瘤侧性对转化治疗方案选择的影响等临床问题;(3)总结转化治疗方案新进展,包括经典双药方案、三药联合的加强方案、分子靶向药物、免疫检查点抑制剂、多种局部疗法以及门静脉栓塞/两步肝切除、联合肝脏分割和门静脉结扎的分步肝切除术在转化治疗中的应用效果。本综述通过分析结直肠癌肝转移转化治疗现有问题,以期为结直肠癌肝转移的临床治疗发展提供参考。

引用本文: 张钰洋, 陈善稳, 王鹏远, 等.  结直肠癌肝转移转化治疗的研究进展 [J] . 中华胃肠外科杂志, 2021, 24(1) : 85-93. DOI: 10.3760/cma.j.cn.441530-20200311-00135.
参考文献导出:   Endnote    NoteExpress    RefWorks    NoteFirst    医学文献王
扫  描  看  全  文

正文
作者信息
基金 0  关键词  0
English Abstract
评论
阅读 0  评论  0
相关资源
引用 | 论文 | 视频

版权归中华医学会所有。

未经授权,不得转载、摘编本刊文章,不得使用本刊的版式设计。

除非特别声明,本刊刊出的所有文章不代表中华医学会和本刊编委会的观点。

结直肠癌发生同时性肝转移的概率约为25%,而全病程中最终发生肝转移的比例则高达40%~50%[1,2]。目前观点认为,对于结直肠癌肝转移(colorectal liver metastases,CRLM)患者,完整的手术切除肝转移灶是唯一可能达到近似根治效果的治疗选择。转化后切除的患者预后与初始切除者近乎一致。因此,转化治疗的概念应运而生。转化治疗是一种通过对适合患者进行术前系统或局部治疗,使肿瘤缩小,进而将初始不可切除病灶转化为可手术切除病灶的治疗方法[3]。多项研究表明,患者对于术前化疗药物的应答率与转化切除率成显著正相关[4]。因此,针对患者转移特点,个体化地选择高应答率的治疗方案可提高转化治疗成功率。经典双药化疗、三药联合加强化疗、联用靶向药物的研究成果使转化切除率不断提高。免疫检查点抑制剂(immune checkpoint inhibitor,ICI)、肝动脉灌注化疗(hepatic arterial infusion,HAI)、经动脉化疗栓塞(trans-arterial chemoembolization,TACE)、选择性内放射疗法(selective internal radiotherapy,SIRT)以及门静脉栓塞(portal vein embolization,PVE)以及联合肝脏分割和门静脉结扎的分步肝切除(associating liver partition and portal vein ligation for staged hepatectomy,ALPPS)在转化治疗中的应用也积累了初步的研究证据,有望在转化治疗领域贡献独特的力量。本文就CRLM的转化治疗进展作一综述。

一、CRLM可切除性的评估

CRLM是否可切除的评估应当由多学科团队(multidisciplinary team,MDT)完成[5]。2019年第4版美国国立综合癌症网络(National Comprehensive Cancer Network,NCCN)指南指出,适合手术切除的患者需同时满足能实现肝内肝外所有病灶的切缘阴性(R0切除)且残余肝功能足够代偿;不完整切除(R1或R2)切除并未证实能使患者获益[6]

具体而言,判断可切除性的因素包括两方面:(1)手术技术方面:要求对于肝脏血流、胆道引流以及残余肝功能的必要保留,由此导致不可切除的原因主要包括肿瘤体积过大、双叶多转移灶以及肿瘤位于不易切除的关键部位[7]。(2)肿瘤学方面:现有证据表明,只有完整切除所有病灶才能显著改善患者预后,故除肝转移灶以外,对于肝外病灶的全面评估至关重要[8]。此外,由于术前治疗效果不佳强烈预示着转化切除后的不良预后,故具有较差预后因素的患者也倾向被划为不可切除[6]

随着化疗方案、影像学分期、肿瘤分子生物学特征方面的进展,可切除肝转移灶的适应范围更加广泛,肝外淋巴结转移以及肝外远隔转移已经不再被视为切除手术的禁忌证。有证据表明,伴有以上特点的CRLM也能从切除手术中获益[9,10]。但即使术前治疗效果极佳,由于转化治疗一般并不能实现转移灶的病理完全缓解,所以因转移灶数量过多而导致无法切除的病例,目前仍不太可能转化为可切除病例,转化治疗成功的案例更多是通过显著缩小肿瘤体积而实现的[6]

二、系统治疗
(一)系统化疗

传统化疗方案包括FOLFIRI(氟尿嘧啶+伊立替康)、FOLFOX(氟尿嘧啶+奥沙利铂)和CAPEOX/XELOX(奥沙利铂+卡培他滨)。三期随机对照试验(randomized control trial,RCT)GERCOR研究发现,FOLFIRI和FOLFOX传统化疗方案能够实现9%~22%的转化率和7%~15%的肝转移灶R0切除率[11];相似的,传统化疗在1 104例初始不可切除的CRLM患者中实现了12.5%的转化切除比例[12]。但另一项包含795例患者的三期RCT回顾性研究却仅得出了3.3%的转化结果[13]

传统一线化疗方案在较大型研究中的低转化率推动了强力三药联合化疗方案FOLFOXIRI(氟尿嘧啶+奥沙利铂+伊立替康)的研究。三期RCT研究GONO表明,相比FOLFIRI方案,FOLFOXIRI方案能显著提高单纯肝转移(liver-limited disease,LLD)患者的R0转化切除率(36%比12%,P=0.017),同时延长中位生存期(23.4个月比16.7个月,P=0.03);不过,这种强化疗法带来了更多的2~3级周围神经病(19%比0)以及3~4级中性粒细胞减少(50%比28%)[14,15]。相比经典双药化疗或序贯多药应用方案,同时联用3种化疗药能够增加CRLM患者的转化切除率,改善患者预后,但由于其增加的毒副作用,三药化疗的适用人群需要经过严格选择。

(二)系统化疗联合靶向治疗

近年来,经典化疗方案与靶向药物的结合显示出了比单用化疗药更显著的疗效获益,使得转化切除率大大提高,其中抗血管内皮生长因子(vascular endothelial growth factor,VEGF)贝伐珠单抗(Bevacizumab)以及抗表皮生长因子受体(epithelial growth factor receptor,EGFR)的西妥昔单抗(Cetuximab)和帕尼单抗(Panitumumab)成为研究热点,具体用药选择需考虑KRAS/NRAS/BRAF等基因突变状况[16]。以下分别阐述抗VEGF单抗及抗EGFR单抗的研究进展。

1.抗EGFR单抗:

RAS/RAF/MAPK通路位于EGFR下游。研究已证明,该通路中KRASNRAS的突变能够预测患者对于西妥昔和帕尼单抗的耐药[17,18]。因此,考虑到抗EGFR单抗的毒副作用以及部分患者不能获益,2019年第4版NCCN指南强烈推荐对所有转移性结直肠癌(metastastic colorectal cancer,mCRC)患者进行肿瘤RAS基因检测(原发部位或转移灶),仅对KRAS和NRAS野生型的患者应用抗VEGF单抗[6]

中国的四期RCT研究BELIEF提示,对于KRAS野生型的初始不可切除LLD患者,在FOLFOX/FOLFIRI化疗方案基础上加入西妥昔单抗,在显著增加了R0切除率(25.7%比7.4%,P<0.01)之外,客观缓解率(objective response rate,ORR)、总生存期(overall survival,OS)和无进展生存期(progression-free survival,PFS)也显著改善[19,20]。此外,三期RCT研究CRYSTAL和二期RCT研究CELIM、OPUS及APEC结果也进一步支持,经典化疗方案和西妥昔的联用能够提高(KRAS野生型的初始不可切除LLD患者的R0切除率[21,22,23]。值得注意的是,在CYSTAL和OPUS研究中,非LLD的(KRAS野生型mCRC患者也实现了R0切除率的提高,这提示一部分转移范围更广泛的晚期患者,仍有希望通过转化治疗获得充分的肿瘤退缩,从而使转移灶的R0切除成为可能。

然而,三期RCT研究COIN指出,在以奥沙利铂为基础的化疗方案中加入西妥昔单抗并不能增加初始不可切除LLD患者治愈性肝切除的机会[24]。与上述试验中普遍采用的静脉输注氟尿嘧啶(5-FU)不同,COIN研究中使用的5-FU药物为非静脉输注含5-FU的化疗方案(CAPOX),这也许能够部分解释该研究得出的阴性结果。

关于帕尼单抗在mCRC转化治疗中的作用,二期RCT研究PLANET将77例KRAS野生型的LLD患者随机分成帕尼单抗+FOLFOX组或帕尼单抗+FOLFIRI组,两组分别实现了45%和59%的转化切除率,中位OS分别为37和41个月;在三期RCT研究PRIME中,FOLFOX基础上加用帕尼单抗使KRAS野生型LLD患者获得了更高的转化切除率(28%比18%,P>0.05)[25,26]

西妥昔单抗与三药强化治疗的联用方面,在两项二期单臂临床试验中,西妥昔+FOLFOXIRI治疗分别实现了37%和60%的R0切除[27,28];二期RCT研究MACBETH结果也显示,西妥昔+FOLFOXIRI方案能够为RAS/BRAF野生型患者带来28%的R0切除率,其中LDD患者实现了52%的R0切除[29]。二期头对头临床试验METHEP-2研究具有格外的意义,它将(K)RAS野生型的初始不可切除CRLM患者随机分到FOLFOX、FOLFIRI或FOLFOXIRI化疗方案组,靶向药物(贝伐珠或西妥昔单抗)的选择根据患者(KRAS状态决定。其结果不仅表明FOLFOXIRI相比FOLFOX或FOLFIRI都具有数字上更好的转化切除效果,更进一步显示出在同样化疗方案基础上,联用西妥昔单抗比贝伐珠单抗具有更高的转化切除率(55.6%比44.7%)[30]

最近,帕尼单抗与FOLFOXIRI的联用也初步得到了临床研究的结果支持:二期RCT研究VOLFI中,对96例RAS野生型初始不可切除的mCRC患者分别给予FOLFOXIRI或FOLFOXIRI+帕尼单抗治疗,与单用FOLFOXIRI相比,加用帕尼单抗的LLD患者获得了更高的转化切除率(33%比12%,P=0.03;21%比9%),以及更长的中位OS(35.7比29.8个月,P=0.12)[31]

值得注意的是,除RAS状态以外,2019年NCCN第4版指南中表示,原发肿瘤的侧性(sideness)实际上是不同的结肠癌分子亚型的集合表现;基于现有临床证据,仅对原发肿瘤位于左半结肠(结肠脾曲至直肠)的mCRC患者推荐西妥昔或帕尼单抗的一线治疗[6,32,33,34]

2.抗VEGF单抗:

抗VEGF的贝伐珠单抗与双药化疗的联用方面,三期RCT研究NO16966表明,在奥沙利铂为基础的化疗方案中加入贝伐珠单抗并不能提高R0切除率[35]。此外,来自ETNA队列的真实世界研究数据显示,在含伊立替康的化疗方案基础上加入贝伐珠单抗所获得的R0切除率与伊立替康单药治疗的数据非常接近,贝伐珠单抗并未体现出在转化治疗中的增效作用[36]

三药化疗与贝伐珠单抗联用方面,二期RCT研究OLIVIA比较了FOLFOXIRI+贝伐珠与FOLFOX+贝伐珠单抗对初始不可切除LLD患者的转化治疗效果,其中FOLFOXIRI组具有更高的R0切除率(49%比23%)[37]。另一项二期RCT研究STEAM将280例未经治疗的mCRC患者随机分为3组:同时给予FOLFOXIRI+贝伐珠单抗组、序贯给予FOLFOXIRI+贝伐珠单抗组和FOLFOX+贝伐珠单抗组;结果显示,3组的转化切除率分别为24%、17%及14%,中位PFS为11.9、11.4和9.5个月[38]。此外,2篇探讨贝伐珠+FOLFOXIRI方案下LLD患者的转化切除率的荟萃分析分别得出了62.2%和39.1%的结果[39,40]

近期另一项三期RCT研究TRIBE 2将679例不可切除的、未经治疗的mCRC患者随机分为接受FOLFOXIRI+贝伐珠单抗的实验组和FOLFOX+贝伐珠单抗的对照组,实验组在疾病进展后序贯以相同治疗方案,对照组在疾病进展后采取FOLFIRI+贝伐珠单抗的序贯治疗;研究结果显示,实验组和对照组的R0转化切除率分别为17%和12%(P=0.047),中位OS分别为27.4和22.5个月(P=0.032),提示对于不可切除的mCRC患者,在持续抑制血管生成的基础上,早期予以FOLFOXIRI三药化疗,在疾病进展后再次予以相同治疗方案,似乎能带来较FOLFOX序贯以FOLFIRI方案更好的疗效[41]。然而,此前一项三期RCT研究TRIBE并未发现贝伐珠单抗与FOLFOXIRI或FOLFIRI方案的联合应用可带来了R0切除率上的获益(15%比12%,P=0.33),可能与其未对LLD患者进行亚组分析有关[42,43]

3.西妥昔单抗与贝伐珠单抗:

比较西妥昔单抗和贝伐珠单抗的头对头试验正在逐渐引起重视。一项单中心三臂RCT研究结果显示,在KRAS野生型的初始不可切除CRLM患者中,相比双药化疗或贝伐珠+化疗方案,西妥昔+化疗方案能够获得数字上更高的转化切除率(三组切除率分别为43.3%、30.7%和51.4%;西妥昔单抗比贝伐珠单抗:HR= 0.42,P=0.07)[7];三期RCT CALGB/SWOG 80405结果也表明,接受西妥昔单抗+化疗(FOLFOX或FOLFIRI)治疗比接受贝伐珠单抗+化疗方案的患者更易实现成功的转化切除(18.2%比13.4%)[44]。然而,三期RCT研究FIRE-3未显示西妥昔单抗+FOLFIRI组相比贝伐珠单抗+FOLFIRI组有转化切除率的优势(11.6%比11.0%)[45]

除了切除率的差异以外,一项针对RAS野生型初始不可切除CRLM患者的三期头对头RCT研究提示,相比贝伐珠单抗治疗,接受西妥昔单抗治疗的患者能够获得更高的ORR,这与既往研究中发现的"更高的ORR意味着更高的转化切除率"互相印证[4]。与ORR相似的,早期肿瘤退缩(early tumor shrinkage,ETS)和肿瘤缓解深度(depth of response,DpR)也能够通过反映肿瘤对于药物的应答程度进而预测转化切除率,甚至是比ORR更有效的参数[46]

综合以上证据,笔者认为,为获得更大的转化切除机会,对于原发肿瘤位于左半结肠的RAS野生型初始不可切除CRLM患者,在基础化疗方案上叠加抗EGFR单抗治疗应当是获益更多的优先选择;对于原发肿瘤位于右半结肠,或者伴有RAS突变的CRLM患者,应首选化疗与抗VEGF单抗的联用。基础化疗方案的选择方面,FOLFOXIRI三药化疗方案及其与西妥昔单抗的联用非常有希望进一步提高RAS野生型CRLM患者的转化切除率。对于经选择的耐受性较好的患者,可优先考虑FOLFOXIRI三药治疗,否则应选用毒副作用较小的FOLFOX或FOLFIRI方案。

4.免疫检查点抑制剂:

目前已在中国上市或由美国食品药品管理局(Food and Drug Administration,FDA)批准的ICI主要包括程序性死亡受体-1(programmed death receptor-1,PD-1)单抗(帕博丽珠单抗/Pembrolizumab、纳武单抗/Nivolumab、Libtayo、特瑞普利单抗/Toripalimab、信迪利单抗/Sintilimab)、程序性死亡配体1(programmed death-ligand 1,PD-L1)单抗(阿特珠单抗/Atezolizumab、德瓦鲁单抗/Durvalumab、阿维单抗/Avelumab)以及细胞毒性T淋巴细胞相关蛋白4(cytotoxic T lymphocyte associated antigen-4,CTLA-4)单抗(伊匹木单抗/Ipilimumab)。ICI适用于微卫星高度不稳定(micrositellite instability-high,MSI-H)/错配修复缺陷(deficient mismatch repair,dMMR)的肿瘤患者[47]。既往研究表明,在CRC患者整体中,MSI-H/dMMR并不少见;相比Ⅳ期患者,MSI-H/dMMR更多地存在于Ⅱ、Ⅲ期患者中(4%比20%比12%)[48]

近年来,ICI在mCRC的治疗中得到了多种有益的尝试。非随机二期临床试验CheckMate 142结果显示,与纳武单抗单药治疗相比,纳武单抗联合伊匹木单抗免疫治疗对于接受过标准治疗的MSI-H/dMMR的复发型或mCRC患者具有较好的安全性和显著的临床疗效[49,50]。二期多中心临床试验KEYNOTE 164的结果显示,帕博丽珠单抗对于接受过一线以上的既往治疗的MSI-H/dMMR的mCRC患者具有持久的抗肿瘤效应,且安全性可控[51]

近期,三期RCT研究KEYNOTE 177的部分数据公布,其将307例初治MSI-H/dMMR的Ⅳ期结直肠癌患者随机分为两组,A组153例接受帕博丽珠单抗治疗,B组154例接受FOLFOX/FOLFIRI±贝伐珠/西妥昔单抗治疗,若疾病进展可交叉进入A组。结果显示,帕博丽珠单抗组ORR为43.8%,显著高于标准化疗组的33.1%(P=0.027 5),且免疫治疗的中位PFS为16.5个月,亦显著优于化疗组的8.2个月(P=0.000 2),两组3~5级治疗相关不良事件(treatment-related adverse event,TRAE)的发生率分别为22%和66%,化疗组中有1例治疗相关死亡[52]

2017—2018年,FDA已经批准了一定适应证下帕博丽珠单抗和纳武单抗±伊匹木单抗的临床应用:5-FU、奥沙利铂和伊立替康治疗后疾病进展的MSI-H/dMMR的12岁以上的mCRC患者。而来自诸如KEYNOTE 177研究等高质量临床数据的证据更新提示,与标准化疗相比,帕博丽珠单抗一线治疗MSI-H/dMMR的mCRC患者能够提高患者治疗反应及PFS,且安全性良好,使得免疫治疗有希望进一步挑战目前标准治疗(化疗±靶向治疗),在未来有潜力成为mCRC的一线治疗方案。

然而,值得注意的是,上述研究的对象是MSI-H/dMMR的mCRC患者,这部分患者发生肝转移的概率较pMMR (proficient mismatch repair,pMMR)患者更低[53]。但近期研究表明,结直肠癌原发灶与肝转移灶的微卫星不稳定及错配修复状态具有较高的一致性[54,55]。综上,在MSI-H/dMMR的mCRC患者中,虽然ICI的应用得到了越来越多临床试验证据支持,但其仍需针对精准定位人群开展更多临床研究以进一步探索。

三、局部治疗
(一)肝动脉灌注化疗(HAI)

肝动脉是直径>3 mm的肝脏转移瘤的主要血供,而门静脉为其余正常肝组织提供营养,这种解剖学基础为HAI药物的给药途径提供了理论支持。一系列同时应用系统治疗和HAI的研究不断为我们更新着疗效证据。一项非对照试验结果显示,HAI途径给予化疗联合贝伐珠单抗,实现了76%的ORR和47%的转化切除率[56]。二期多中心研究OPTILIV探索了HAI途径给予三联化疗药(奥沙利铂、5-FU、伊立替康)联合西妥昔单抗对于RAS野生型的初始不可切除CRLM患者的疗效,发现HAI可实现40.6%的ORR和29.7%的R0/R1转化切除率,45%成功实现转移灶切除的患者在4年后仍然存活,而未切除组全部死亡[57]。一项纳入了11个研究、1 514例患者的Meta分析探讨了HAI与CRLM转化治疗的关系,结果显示,18%患者实现了转化切除,中位OS和5年OS分别达到53个月和49%[58]。然而,需要考虑的是,伴随HAI局部药物高浓度的是更高的药物相关肝脏损伤发生概率[59];因此,HAI应用后以及肝转移灶切除术前,均需进行严格的肝功能监测。

(二)经动脉化疗栓塞(TACE)

国际介入放射学学会将TACE定义为:向肿瘤供养血管局部注射入≥1种化疗药物,使之发生栓塞。TACE包括传统TACE(conventional TACE,cTACE)和药物洗脱微珠TACE(drug-eluting bead,DEB-TACE)。相比cTACE的碘油栓塞剂,DEB-TACE利用聚乙烯醇微球溶解携带药物,可持续缓释抗癌药,更好地控制药物浓度[60]

一项纳入70例CRLM的RCT研究在静脉化疗FOLFOX基础上,分别给予DEB-伊立替康(Irinotecan-loaded DEB,DEBIRI组)或贝伐珠单抗,结果显示,DEBIRI组具有显著更高的转化切除率(35%比16%,P=0.05)[61]。近期,一项有关DEBIRI的一期剂量递增试验(微球剂量分别为50、75、100 mg/ml)公布了研究成果,其给予9个CRLM患者共22轮TACE治疗,7 d后序贯以FOLFIRI静脉化疗,实现了55.6%的ORR和18.2个月的中位OR,其中1例患者实现了转化切除(1/9,11.1%)[62]

(三)选择性内放射(SIRT)

SIRT是指将载有放射性同位素90Y的玻璃或树脂微球选择性地注射入肝动脉并到达靶组织,利用高能量β-射线杀伤局部肿瘤细胞[63]。近年来,化疗+SIRT的相关研究得出了10%~21%的转化切除结果[64,65]。三期多中心RCT研究SIRFLOX中472例CRLM患者的分析结果发现,FOLFOX与FOLFOX+SIRT方案分别带来了28.9%和38.1%的转化切除率(P<0.001),提示化疗基础上加用SIRT能够进一步增加CRLM的转化切除率[65]

然而,包含上述研究的、共纳入1 103例CRLM患者的3项大型三期多中心RCT研究(FOXFIRE,SIRFLOX和FOXFIRE-Global)却得出了不同的结论:单独应用FOLFOX组和FOLFOX+SIRT组的转化切除率分别为16%和17%(P=0.67),中位OS分别为23.3和22.6个月(P=0.61),提示一线系统化疗FOLFOX联用局部治疗SIRT不能明显改善转化切除率和CRLM患者的生存预后[66]。笔者认为,化疗+SIRT组肝脏病灶的局部控制并未带来生存优势的原因,或许与相当比例患者首先出现了肝外转移有关,这也同时降低了成功转化切除肝转移灶的可能性。

综合以上研究结果,笔者认为,HAI、TACE和SIRT在不可切除CRLM的治疗领域中得到了诸多有益的尝试,具有在化疗基础上进一步提高转化切除率、延长生存期的潜力,但以上疗法在转化治疗中的应用效力仍需更多大型RCT研究的一致结果支持,联合用药的基础上尝试不同给药方式的组合或是未来的研究热点之一。

四、其他热点问题
(一)门静脉栓塞(PVE)与联合肝脏分割和门静脉结扎的分步肝切除(ALPPS)

除了高应答的转化治疗方案选择,对于需要较大体积肝切除或存在术前肝功能不全(如由于化疗、靶向治疗)的CRLM患者,如何降低术后并发症和提高术后残余肝(future liver remnant,FLR)体积和功能是一个同样重要的议题。门静脉结扎(portal vein ligation,PVL)或栓塞通过肝切除前诱导FLR增生,可在6周内实现约40%的FLR体积增长,且安全性良好[67]。随后,Adam等[68]提出两步肝切除(two-staged hepatectomy,TSH),在第一阶段手术通过PVL或PVE等方法刺激剩余肝脏增生,为双叶CRLM的患者提供了治愈性肝切除的可能。近年来,通过阻断肝实质间门脉侧支循环的ALPPS,凭借更快的肝再生速度以及更低的肿瘤侵入FLR概率的优势而获得研究关注,有望成为一种进一步提高CRLM肝转移灶切除率的新方法。

Schnitzbauer等[69]最先在25个患者中报道了ALPPS后一周内74%的FLR体积增加,两阶段手术的中位间隔时间仅9 d,然而,64%患者出现了术后并发症。2017年的一项对比ALPSS和PVE/TSH的Meta分析结果显示,ALPSS与PVE/TSH相比,FLR体积增长无明显差别,但其术后并发症和死亡率比PVE/TSH显著更高[70]。随后,一项多中心RCT研究LIRGO研究提供了更支持ALPSS的证据:对于FLR不足够而不能单次切除的CRLM患者而言,ALPPS组比PVE/TSH组获得了显著更高的切除率(92%比57%,P<0.001),且两组的术后并发症和90 d死亡率无明显差异(43%比43%;8.3%比6.1%);该研究还提出了应用HIDA闪烁照相评估术后FLR功能变化,而非仅评估FLR体积增加的重要观点[71]。最近一项针对72例原发或继发肝脏肿瘤患者的回顾性研究结果显示,PVE/TSH、完全ALPPS或部分ALPPS治疗这3种术式导致的FRL体积增生无明显差异,但完全或部分ALPPS组的FRL功能增加显著高于PVE/TSH组(16.7%比9.3%比4.9%),且达到预期增生反应的时间均较PVE组短,提示ALPPS后肝脏功能增加的幅度与速度均优于PVE/TSH;而部分ALPPS与PVE/TSH的术后并发症及90 d死亡率相当,均显著低于完全ALPPS(17%比18%比30%;0比2%比25%)[72]

从现有研究结果看来,PVE/TSH和ALPPS均能够促进FLR增生,似乎是初始不可切除CRLM患者的一个潜在选择;但值得注意的是,相比研究证据更充分的PVE/TSH,ALPPS的应用安全性仍需更大型RCT研究进一步阐明。

(二)疗效评估及手术时机的再审视

转化治疗中客观及时的疗效评估十分重要,然而,通过影像学手段对病灶进行真实反映却是一个长久以来的难题。由于化疗药物的应用对肝实质不可避免的影响,例如伊立替康+5-FU治疗后的肝脂肪化和奥沙利铂治疗后的肝窦阻塞综合征,健康的肝组织与病灶之间的影像学对比增强会相对减弱,使得转移灶的检出率下降。另外,不同影像学手段对肝转移灶的检出率之间也存在差异:MRI、CT、FDG-PET和FDG-PET/CT的灵敏度分别为85.7%、69.9%、54.5%和51.7%[73]。故相比CT,MRI对于化疗后肝转移灶的检测似乎更加合适。不仅如此,MRI的敏感度还能通过肝脏特异性对比剂以进一步增强,如钆塞酸(gadoxetic acid)和钆布醇(gadobutrol)。

治疗中影像学上病灶的消失会导致切除范围难以确定,这是转化治疗的潜在风险与难题。另外,大部分转化治疗后在CT中消失的转移灶依然有病理学的残存病灶,仍应手术处理。因此,疗效的及时评估对于合适手术时机的选择及合理手术范围的确定具有关键意义。

基于以上认识,关于转化治疗中的疗效评价频率及手术时机,2019年第4版NCCN指南推荐,疗效评价应自转化治疗开始后2个月开始,此后每2个月评价一次;在转化治疗成功后,应尽早手术,以减少化疗相关肝损伤的进展[6]。虽然如何判定转化治疗失败目前尚无学界共识,但多数研究认为,当患者在转化治疗期间出现疾病进展或无法耐受其副作用时,便应立即停止治疗,同时认定转化治疗失败。二期RCT研究CELIM表明,从转化治疗到实现治愈性切除或以切除为目的的探查性手术的中位时间为5.1个月,平均经过8个转化治疗周期、且8个月后才完成肝转移灶切除的患者比例极少。研究者随后推荐,从转化治疗启动到初次评估的时间间隔最多为3个月,且之后每2个月(4个周期)评估一次;若6个月后如果仍然评估认定病灶为不可切除,则应重新评估治疗策略[21]

除了基于影像学检查的评估手段以外,多种血清生物标志物也在CRLM的监测中显示出越来越重要的作用。随着对肿瘤遗传学发病机制的了解加深以及基因组学相关知识技术的进展,我们认识到,近乎所有肿瘤都携带着体细胞基因突变,且这种突变在正常细胞中频率极低。因此,通过无创方式检测肿瘤DNA,以反映肿瘤负荷、识别重要突变的构想正在逐步得到验证,"液体活检"的概念随之提出[74]。最新研究显示,循环肿瘤DNA(circulating tumor DNA,ctDNA)也许是一种比影像学能够更精准地反映转化治疗效果的动态评估方式:通过识别组织特异性的循环甲基化DNA片段,并将其作为血清标志物,结合微滴式数字聚合酶链式反应(PCR)技术,或许更灵敏、特异地判断CRC有无肝转移[75]。肝脏特异的甲基化ctDNA片段mSEPT9的水平与肿瘤大小、组织学等级和类型相关,具有监测疾病动态的潜力[76]。以上证据为利用组织特异性ctDNA评估转化治疗中肝转移灶的动态变化提供了重要依据。

(三)肿瘤侧性之争

近期研究表明,不同原发部位的CRC具有不同的分子生物学特征和临床表现。在RAS野生型的CRLM患者中,左侧肿瘤的预后比右侧肿瘤显著更好[77]。右侧肿瘤更易表现为RASBRAF突变及MSI-H,而左侧肿瘤则与HER-2扩增、染色体不稳定及更有利于EGFR单抗治疗的基因表达谱相关[78]。虽然尚无不同药物对左右侧CRC转化切除影响的直接研究数据,仍有许多临床试验对CRC侧性做出了重点关注,探索其对靶向药物治疗的ORR等结局指标的影响。由于ORR与转化切除率的正向相关性,这些研究为肿瘤侧性与转化治疗选择关系的讨论提供了重要的证据基础。

两篇探究肿瘤侧性对RAS野生型mCRC患者生物制剂选择的影响的Meta分析显示,无论肿瘤位于哪一侧,与VEGF单抗相比,联合基础化疗的EGFR单抗治疗均可获得更高的ORR、ETS和DpR,故当以转化切除为目的时,在RAS野生型前提下,EGFR单抗相比VEGF单抗似乎为更优选择[78,79]。然而,一项纳入75例RAS和BRAF野生型mCRC患者的研究对比结直肠癌原发部位对于抗EGFR单抗±伊立替康的疗效影响发现,左侧肿瘤的ORR为41%,而右侧肿瘤均无应答(P=0.003),且左侧肿瘤患者的中位PFS相比右侧肿瘤患者更长(6.6个月比2.3个月,P<0.000 1)[33]。三期RCT研究CALGB/SWOG 80405和FIRE-3结果也表明,西妥昔单抗更适用于RAS野生型的左侧肿瘤,而对于RAS野生型的右侧肿瘤,相比西妥昔单抗治疗,贝伐珠单抗能够为患者带来更长的OS[80]

基于以上研究,2019年第4版NCCN指南中仅对于KRAS/NRAS/BRAF野生型且肿瘤位于左侧结肠的患者推荐西妥昔或帕尼单抗的使用[6]。另外,值得注意的是,对于达到肿瘤早期退缩的RAS野生型CRLM患者,左侧或右侧肿瘤的中位PFS和OS无明显差异。因此,ETS有潜力成为筛选右侧肿瘤患者中对EGFR单抗治疗效果较好的亚群指标[78]

五、总结

随着CRLM可切除性的定义延展,如何提高初始不可切除CRLM的转化切除率,从而改善进展期CRC患者的远期预后已经成为了CRC领域的研究焦点。转化治疗方案中,三药联合的FOLFOXIRI较传统两药方案转化率高;化疗联合靶向治疗可提高转化率,改善预后生存,对KRAS野生型的左半结肠肿瘤患者,西妥昔单抗联合化疗能提高转化率,而对于不适用西妥昔单抗的病例,联合贝伐珠单抗也是一种可选方案;ICI在mCRC的初步探索也显示了希望;多种局部治疗有潜力进一步提高转化切除率;PVE/TSH、ALPPS能够提高术后残余肝体积和功能。转化治疗的疗效评估仍以影像学为主。液体活检标志物ctDNA监测肝转移灶动态变化显现了初步的诊断价值。CRC侧性影响预后,但当以转化治疗为目的时,EGFR单抗对于无论左右侧肿瘤均更具优势和潜力。

利益冲突
利益冲突

所有作者均声明不存在利益冲突

参考文献
[1]
National Cancer Institute. SEER cancer stat facts:colon and rectum cancer,2003-2009. 2013[EB/OL].[2020-06-16]. https://seer.cancer.gov/statfacts/html/colorect.html.
[2]
[3]
HaraldsdottirS, WuC, BloomstonMet al. What is the optimal neo-adjuvant treatment for liver metastasis?[J]. Ther Adv Med Oncol20135(4): 221-234. DOI: 10.1177/1758834013485111.
[4]
OkunoM, HatanoE, NishinoHet al. Does response rate of chemotherapy with molecular target agents correlate with the conversion rate and survival in patients with unresectable colorectal liver metastases?:a systematic review[J]. Eur J Surg Oncol201743(6): 1003-1012. DOI: 10.1016/j.ejso.2016.08.019.
[5]
BlissLA, StrongEA, GamblinTC. Surgical resectability of multisite metastatic colorectal cancer:pushing the limits while appropriately selecting patients[J]. J Surg Oncol2019119(5): 623-628. DOI: 10.1002/jso.25419.
[6]
National Comprehensive Cancer Network(NCCN). NCCN Clinical Practice Guidelines in Oncology. Colon Cancer(Version 4.2019)[EB/OL].[2020-06-16]. https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf.
[7]
BassoM, DadduzioV, ArditoFet al. Conversion chemotherapy for technically unresectable colorectal liver metastases:a retrospective,STROBE-compliant,single-center study comparing chemotherapy alone and combination chemotherapy with cetuximab or bevacizumab[J]. Medicine(Baltimore)201695(20): e3722. DOI: 10.1097/MD.0000000000003722.
[8]
LeungU, GönenM, AllenPJet al. Colorectal cancer liver metastases and concurrent extrahepatic disease treated with resection[J]. Ann Surg2017265(1): 158-165. DOI: 10.1097/SLA.0000000000001624.
[9]
KhanAS, Garcia-ArozS, AnsariMAet al. Assessment and optimization of liver volume before major hepatic resection:Current guidelines and a narrative review[J]. Int J Surg201852: 74-81. DOI: 10.1016/j.ijsu.2018.01.042.
[10]
ChuaTC, SaxenaA, LiauwWet al. Hepatectomy and resection of concomitant extrahepatic disease for colorectal liver metastases--a systematic review[J]. Eur J Cancer201248(12): 1757-1765. DOI: 10.1016/j.ejca.2011.10.034.
[11]
TournigandC, AndréT, AchilleEet al. FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer:a randomized GERCOR study[J]. J Clin Oncol200422(2): 229-237. DOI: 10.1200/JCO.2004.05.113.
[12]
AdamR, DelvartV, PascalGet al. Rescue surgery for unresectable colorectal liver metastases down-staged by chemotherapy:a model to predict long-term survival[J]. Ann Surg2004240: 644-658. DOI: 10.1097/01.sla.0000141198.92114.16.
[13]
DelaunoitT, AlbertsSR, SargentDJet al. Chemotherapy permits resection of metastatic colorectal cancer:experience from Intergroup N9741[J]. Ann Oncol200516(3): 425-429. DOI: 10.1093/annonc/mdi092.
[14]
FalconeA, RicciS, BrunettiIet al. Phase III trial of infusional fluorouracil,leucovorin,oxaliplatin,and irinotecan (FOLFOXIRI)compared with infusional fluorouracil,leucovorin,and irinotecan(FOLFIRI)as first-line treatment for metastatic colorectal cancer:the Gruppo Oncologico Nord Ovest[J]. J Clin Oncol200725(13): 1670-1676. DOI: 10.1200/JCO.2006.09.0928.
[15]
MasiG, VasileE, LoupakisFet al. Randomized trial of two induction chemotherapy regimens in metastatic colorectal cancer:an updated analysis[J]. J Natl Cancer Inst2011103(1): 21-30. DOI: 10.1093/jnci/djq456.
[16]
De RoockW, De VriendtV, NormannoNet al. KRAS,BRAF,PIK3CA,and PTEN mutations:implications for targeted therapies in metastatic colorectal cancer[J]. Lancet Oncol201112(6): 594-603. DOI: 10.1016/S1470-2045(10)70209-6.
[17]
DouillardJY, OlinerKS, SienaSet al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer[J]. N Engl J Med2013369(11): 1023-1034. DOI: 10.1056/NEJMoa1305275.
[18]
SorichMJ, WieseMD, RowlandAet al. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer:a meta-analysis of randomized,controlled trials[J]. Ann Oncol201526(1): 13-21. DOI: 10.1093/annonc/mdu378.
[19]
YeLC, LiuTS, RenLet al. Randomized controlled trial of cetuximab plus chemotherapy for patients with KRAS wild-type unresectable colorectal liver-limited metastases[J]. J Clin Oncol201331(16): 1931-1938. DOI: 10.1200/JCO.2012.44.8308.
[20]
XuJ, RenL, WeiYet al. Effects of beyond KRAS mutations on the efficacy of cetuximab plus chemotherapy for patients with unresectable colorectal liver-limited metastases(BELIEF):a retrospective biomarker analysis from a Chinese population[J]. Ann Oncol201627Suppl 6: S541. DOI: 10.1093/annonc/mdw370.89.
[21]
FolprechtG, GruenbergerT, BechsteinWOet al. Tumour response and secondary resectability of colorectal liver metastases following neoadjuvant chemotherapy with cetuximab:the CELIM randomised phase 2 trial[J]. Lancet Oncol201011(1): 38-47. DOI: 10.1016/S1470-2045(09)70330-4.
[22]
KohneC, BokemeyerC, HeegerSet al. Efficacy of chemotherapy plus cetuximab according to metastatic site in KRAS wild-type metastatic colorectal cancer(mCRC):analysis of CRYSTAL and OPUS studies[J]. J Clin Oncol201129Suppl 15: S3576. DOI: 10.1200/jco.2011.29.15_suppl.3576.
[23]
ChengAL, CornelioG, ShenLet al. Efficacy,tolerability,and biomarker analyses of once-every-2-weeks cetuximab plus first-line FOLFOX or FOLFIRI in patients with KRAS or all RAS wild-type metastatic colorectal cancer:the phase 2 APEC study[J]. Clin Colorectal Cancer201716(2): e73-e88. DOI: 10.1016/j.clcc.2016.08.005.
[24]
MaughanTS, AdamsRA, SmithCGet al. Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer:results of the randomised phase 3 MRC COIN trial[J]. Lancet2011377(9783): 2103-2114. DOI: 10.1016/S0140-6736(11)60613-2.
[25]
CarratoA, AbadA, MassutiBet al. First-line panitumumab plus FOLFOX4 or FOLFIRI in colorectal cancer with multiple or unresectable liver metastases:a randomised,phase II trial (PLANET-TTD)[J]. Eur J Cancer201781: 191-202. DOI: 10.1016/j.ejca.2017.04.024.
[26]
DouillardJY, SienaS, CassidyJet al. Final results from PRIME:randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer[J]. Ann Oncol201425(7): 1346-1355. DOI: 10.1093/annonc/mdu141.
[27]
SaridakiZ, AndroulakisN, VardakisNet al. A triplet combination with irinotecan(CPT-11),oxaliplatin(LOHP),continuous infusion 5-fluorouracil and leucovorin(FOLFOXIRI)plus cetuximab as first-line treatment in KRAS wt,metastatic colorectal cancer:a pilot phase II trial[J]. Br J Cancer2012107(12): 1932-1937. DOI: 10.1038/bjc.2012.509.
[28]
GarufiC, TorselloA, TumoloSet al. Cetuximab plus chronomodulated irinotecan,5-fluorouracil,leucovorin and oxaliplatin as neoadjuvant chemotherapy in colorectal liver metastases:POCHER trial[J]. Br J Cancer2010103(10): 1542-1547. DOI: 10.1038/sj.bjc.6605940.
[29]
CremoliniC, AntoniottiC, LonardiSet al. Activity and safety of cetuximab plus modified FOLFOXIRI followed by maintenance with cetuximab or bevacizumab for RAS and BRAF wild-type metastatic colorectal cancer:a randomized phase 2 clinical trial[J]. JAMA Oncol20184(4): 529-536. DOI: 10.1001/jamaoncol.2017.5314.
[30]
YchouM, RivoireM, ThezenasSet al. FOLFIRINOX combined to targeted therapy according RAS status for colorectal cancer patients with liver metastases initially non-resectable:a phase II randomized study-prodige 13-ACCORD 21(METHEP-2),a unicancer GI trial[J]. J Clin Oncol201634Suppl: S3512.
[31]
GeisslerM, TannapfelA, Reinacher-SchickAet al. Final results of the randomized phase II VOLFI trial(AIOKRK0109):mFOLFOXIRI 1 Panitumumab versus FOLFOXIRI as firstline treatment in patients with RAS wild-type metastatic colorectal cancer(mCRC)[J]. Ann Oncol201930Suppl 4: PD030.
[32]
BruleSY, JonkerDJ, KarapetisCSet al. Location of colon cancer(right-sided versus left-sided)as a prognostic factor and a predictor of benefit from cetuximab in NCIC CO.17[J]. Eur J Cancer201551: 1405-1414.
[33]
MorettoR, CremoliniC, RossiniDet al. Location of primary tumor and benefit from anti-epidermal growth factor receptor monoclonal antibodies in patients with RAS and BRAF wild-type metastatic colorectal cancer[J]. Oncologist201621(8): 988-994. DOI: 10.1634/theoncologist.2016-0084.
[34]
LoupakisF, YangD, YauLet al. Primary tumor location as a prognostic factor in metastatic colorectal cancer[J]. J Natl Cancer Inst2015107(3): 427. DOI: 10.1093/jnci/dju427.
[35]
OkinesA, PuertoOD, CunninghamDet al. Surgery with curative-intent in patients treated with first-line chemotherapy plus bevacizumab for metastatic colorectal cancer First BEAT and the randomised phase-III NO16966 trial[J]. Br J Cancer2009101(7): 1033-1038. DOI: 10.1038/sj.bjc.6605259.
[36]
RouyerM, SmithD, LaurentCet al. Secondary metastases resection after bevacizumab plus irinotecan-based chemotherapy in first-line therapy of metastatic colorectal cancer in a real-life setting:results of the ETNA cohort[J]. Target Oncol201611(1): 83-92. DOI: 10.1007/s11523-015-0377-6.
[37]
GruenbergerT, BridgewaterJ, ChauIet al. Bevacizumab plus mFOLFOX-6 or FOLFOXIRI in patients with initially unresectable liver metastases from colorectal cancer:the OLIVIA multinational randomised phase II trial[J]. Ann Oncol201526(4): 702-708. DOI: 10.1093/annonc/mdu580.
[38]
HurwitzHI, TanBR, ReevesJAet al. Phase II randomized trial of sequential or concurrent FOLFOXIRI-bevacizumab versus FOLFOX-bevacizumab for metastatic colorectal cancer(STEAM)[J]. Oncologist201924(7): 921-932. DOI: 10.1634/theoncologist.2018-0344.
[39]
CremoliniC, CasagrandeM, LoupakisFet al. Efficacy of FOLFOXIRI plus bevacizumab in liver-limited metastatic colorectal cancer:a pooled analysis of clinical studies by Gruppo Oncologico del Nord Ovest[J]. Eur J Cancer201773: 74-84. DOI: 10.1016/j.ejca.2016.10.028.
[40]
TomaselloG, PetrelliF, GhidiniMet al. FOLFOXIRI plus bevacizumab as conversion therapy for patients with initially unresectable metastatic colorectal cancer:a systematic review and pooled analysis[J]. JAMA Oncol20173(7): e170278. DOI: 10.1001/jamaoncol.2017.0278.
[41]
CremoliniC, AntoniottiC, RossiniDet al. Upfront FOLFOXIRI plus bevacizumab and reintroduction after progression versus mFOLFOX6 plus bevacizumab followed by FOLFIRI plus bevacizumab in the treatment of patients with metastatic colorectal cancer(TRIBE2):a multicentre,open-label,phase 3,randomised,controlled trial[J]. Lancet Oncol202021(4): 497-507. DOI: 10.1016/S1470-2045(19)30862-9.
[42]
LoupakisF, CremoliniC, MasiGet al. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal cancer[J]. N Engl J Med2014371(17): 1609-1618. DOI: 10.1056/NEJMoa1403108.
[43]
CremoliniC, LoupakisF, AntoniottiCet al. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer:updated overall survival and molecular subgroup analyses of the open-label,phase 3 TRIBE study[J]. Lancet Oncol201516(13): 1306-1315. DOI: 10.1016/S1470-2045(15)00122-9.
[44]
VenookA, NiedzwieckiD, LenzHet al. CALGB/SWOG 80405:analysis of patients undergoing surgery as part of treatment strategy[J]. Ann Oncol201425Suppl: LBA10.
[45]
ModestDP, StintzingS, von WeikersthalLFet al. Impact of subsequent therapies on outcome of the FIRE-3/AIO KRK0306 trial:first-line therapy with FOLFIRI plus cetuximab or bevacizumab in patients with KRAS wild-type tumors in metastatic colorectal cancer[J]. J Clin Oncol201533(32): 3718-3726. DOI: 10.1200/JCO.2015.61.2887.
[46]
HeinemannV, StintzingS, ModestDPet al. Early tumour shrinkage(ETS)and depth of response(DpR)in the treatment of patients with metastatic colorectal cancer(mCRC)[J]. Eur J Cancer201551(14): 1927-1936. DOI: 10.1016/j.ejca.2015.06.116.
[47]
CiardielloD, VitielloPP, CardoneCet al. Immunotherapy of colorectal cancer:challenges for therapeutic efficacy[J]. Cancer Treat Rev201976: 22-32. DOI: 10.1016/j.ctrv.2019.04.003.
[48]
LeDT, UramJN, WangHet al. PD-1 blockade in tumors with mismatch-repair deficiency[J]. N Engl J Med2015372(26): 2509-2520. DOI: 10.1056/NEJMoa1500596.
[49]
OvermanMJ, McDermottR, LeachJLet al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer(CheckMate 142):an open-label,multicentre,phase 2 study[J]. Lancet Oncol201718(9): 1182-1191. DOI: 10.1016/S1470-2045(17)30422-9.
[50]
OvermanMJ, LonardiS, WongKYMet al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer[J]. J Clin Oncol201836(8): 773-779. DOI: 10.1200/JCO.2017.76.9901.
[51]
LeDT, KimTW, Van CutsemEet al. Phase II open-label study of pembrolizumab in treatment-refractory,microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer:KEYNOTE-164[J]. J Clin Oncol202038(1): 11-19. DOI: 10.1200/JCO.19.02107.
[52]
AndreTShiuKKKimTWet al. Pembrolizumab vs chemotherapy for microsatellite instability-high/mismatch repair deficient metastatic colorectal cancer:the phase 3 KEYNOTE-177 study[C/OL]. ASCO Virtual Scientific Program2020,In press.
[53]
Nordholm-CarstensenA, KrarupPM, MortonDet al. Mismatch repair status and synchronous metastases in colorectal cancer:a nationwide cohort study[J]. Int J Cancer2015137(9): 2139-2148. DOI: 10.1002/ijc.29585.
[54]
HeWZ, HuWM, WangFet al. Comparison of mismatch repair status between primary and matched metastatic sites in patients with colorectal cancer[J]. J Natl Compr Canc Netw201917(10): 1174-1183. DOI: 10.6004/jnccn.2019.7308.
[55]
EvrardC, TachonG, RandrianVet al. Microsatellite instability:diagnosis,heterogeneity,discordance,and clinical impact in colorectal cancer[J]. Cancers(Basel)201911(10)DOI: 10.3390/cancers11101567.
[56]
D'AngelicaMI, Correa-GallegoC, PatyPBet al. Phase II trial of hepatic artery infusional and systemic chemotherapy for patients with unresectable hepatic metastases from colorectal cancer:conversion to resection and long-term outcomes[J]. Ann Surg2015261(2): 353-360. DOI: 10.1097/SLA.0000000000000614.
[57]
LéviFA, BoigeV, HebbarMet al. Conversion to resection of liver metastases from colorectal cancer with hepatic artery infusion of combined chemotherapy and systemic cetuximab in multicenter trial OPTILIV[J]. Ann Oncol201627(2): 267-274. DOI: 10.1093/annonc/mdv548.
[58]
ChanDL, AlzahraniNA, MorrisDLet al. Systematic review and meta-analysis of hepatic arterial infusion chemotherapy as bridging therapy for colorectal liver metastases[J]. Surg Oncol201524(3): 162-171. DOI: 10.1016/j.suronc.2015.06.014.
[59]
AllardMA, SebaghM, BaillieGet al. Comparison of complete pathologic response and hepatic injuries between hepatic arterial infusion and systemic administration of oxaliplatin in patients with colorectal liver metastases[J]. Ann Surg Oncol201522(6): 1925-1932. DOI: 10.1245/s10434-014-4272-7.
[60]
KalliniJR, GabrA, AbouchalehNet al. New developments in interventional oncology:liver metastases from colorectal cancer[J]. Cancer J201622(6): 373-380. DOI: 10.1097/PPO.0000000000000226.
[61]
MartinRC, ScogginsCR, SchreederMet al. Randomized controlled trial of irinotecan drug-eluting beads with simultaneous FOLFOX and bevacizumab for patients with unresectable colorectal liver-limited metastasis[J]. Cancer2015121(20): 3649-3658. DOI: 10.1002/cncr.29534.
[62]
TanakaT, SatoT, NishiofukuHet al. Selective TACE with irinotecan-loaded 40 μm microspheres and FOLFIRI for colorectal liver metastases:phase I dose escalation pharmacokinetic study[J]. BMC Cancer201919(1): 758. DOI: 10.1186/s12885-019-5862-3.
[63]
WangLM, JaniAR, HillEJet al. Anatomical basis and histopathological changes resulting from selective internal radiotherapy for liver metastases[J]. J Clin Pathol201366(3): 205-211. DOI: 10.1136/jclinpath-2012-201231.
[64]
JeyarajahDR, DoyleMBM, EspatNJet al. Role of yttrium-90 selective internal radiation therapy in the treatment of liver-dominant metastatic colorectal cancer:an evidence-based expert consensus algorithm[J]. J Gastrointest Oncol202011(2): 443-460. DOI: 10.21037/jgo.2020.01.09.
[65]
GarlippB, GibbsP, Van HazelGAet al. Secondary technical resectability of colorectal cancer liver metastases after chemotherapy with or without selective internal radiotherapy in the randomized SIRFLOX trial[J]. Br J Surg2019106(13): 1837-1846. DOI: 10.1002/bjs.11283.
[66]
WasanHS, GibbsP, SharmaNKet al. First-line selective internal radiotherapy plus chemotherapy versus chemotherapy alone in patients with liver metastases from colorectal cancer (FOXFIRE,SIRFLOX,and FOXFIRE-Global):a combined analysis of three multicentre,randomised,phase 3 trials[J]. Lancet Oncol201718(9): 1159-1171. DOI: 10.1016/S1470-2045(17)30457-6.
[67]
van LiendenKP, van den EsschertJW, de GraafWet al. Portal vein embolization before liver resection:a systematic review[J]. Cardiovasc Intervent Radiol201336(1): 25-34. DOI: 10.1007/s00270-012-0440-y.
[68]
AdamR, LaurentA, AzoulayDet al. Two-stage hepatectomy:a planned strategy to treat irresectable liver tumors[J]. Ann Surg2000232(6): 777-785. DOI: 10.1097/00000658-200012000-00006.
[69]
SchnitzbauerAA, LangSA, GoessmannHet al. Right portal vein ligation combined with in situ splitting induces rapid left lateral liver lobe hypertrophy enabling 2-staged extended right hepatic resection in small-for-size settings[J]. Ann Surg2012255(3): 405-414. DOI: 10.1097/SLA.0b013e31824856f5.
[70]
MorisD, Ronnekleiv-KellyS, KostakisIDet al. Operative results and oncologic outcomes of associating liver partition and portal vein ligation for staged hepatectomy(ALPPS)versus two-stage hepatectomy(TSH)in patients with unresectable colorectal liver metastases:a systematic review and meta-analysis[J]. World J Surg201842(3): 806-815. DOI: 10.1007/s00268-017-4181-6.
[71]
SandströmP, RøsokBI, SparrelidEet al. ALPPS improves resectability compared with conventional two-stage hepatectomy in patients with advanced colorectal liver metastasis:results from a scandinavian multicenter randomized controlled trial(LIGRO Trial)[J]. Ann Surg2018267(5): 833-840. DOI: 10.1097/SLA.0000000000002511.
[72]
RassamF, OlthofPB, van LiendenKPet al. Comparison of functional and volumetric increase of the future remnant liver and postoperative outcomes after portal vein embolization and complete or partial associating liver partition and portal vein ligation for staged hepatectomy(ALPPS)[J]. Ann Transl Med20208(7): 436. DOI: 10.21037/atm.2020.03.191.
[73]
KuhlmannK, Van HilstJ, FisherSet al. Management of disappearing colorectal liver metastases[J]. Eur J Surg Oncol201642(12): 1798-1805. DOI: 10.1016/j.ejso.2016.05.005.
[74]
MurtazaM, DawsonSJ, TsuiDWet al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA[J]. Nature2013497(7447): 108-112. DOI: 10.1038/nature12065.
[75]
GaiW, JiL, LamWKJet al. Liver- and Colon-specific DNA methylation markers in plasma for investigation of colorectal cancers with or without liver metastases[J]. Clin Chem201864(8): 1239-1249. DOI: 10.1373/clinchem.2018.290304.
[76]
FuB, YanP, ZhangSet al. Cell-free circulating methylated sept9 for noninvasive diagnosis and monitoring of colorectal cancer[J]. Dis Markers20182018: 6437104. DOI: 10.1155/2018/6437104.
[77]
MissiagliaE, JacobsB, D'ArioGet al. Distal and proximal colon cancers differ in terms of molecular,pathological,and clinical features[J]. Ann Oncol201425(10): 1995-2001. DOI: 10.1093/annonc/mdu275.
[78]
ChenD, ZhangX, GaoGet al. Should anti-EGFR mAbs be discontinued for conversion surgery in untreated right-sided metastatic colorectal cancer?A systematic review and meta-analysis[J]. World J Surg Oncol201816(1): 200. DOI: 10.1186/s12957-018-1502-7.
[79]
HolchJW, RicardI, StintzingSet al. The relevance of primary tumour location in patients with metastatic colorectal cancer:a meta-analysis of first-line clinical trials[J]. Eur J Cancer201770: 87-98. DOI: 10.1016/j.ejca.2016.10.007.
[80]
ArnoldD, DouillardJY, PignonJPet al. Prognostic and predictive value of primary tumour side in patients with RAS wild-type metastatic colorectal cancer treated with chemotherapy and EGFR directed antibodies in six randomized trials[J]. Ann Oncol201728(8): 1713-1729.
 
 
展开/关闭提纲
查看图表详情
回到顶部
放大字体
缩小字体
标签
关键词