局部治疗联合系统治疗在肝细胞癌转化治疗中的价值
DOI: 10.12449/JCH240903
Value of local treatment combined with systemic therapy in conversion therapy for hepatocellular carcinoma
-
摘要: 肝细胞癌是临床中最常见的恶性肿瘤之一。由于早期缺乏典型的临床表现,我国大多数患者确诊时已达中晚期,错失手术切除机会,预后较差。因此通过相关治疗手段将不可切除肝细胞癌转化为可切除肝细胞癌的探索十分必要。近年来,随着经动脉介入治疗、放射治疗技术等局部治疗的不断进步完善、新的靶向及免疫药物的临床应用,为肝细胞癌的转化治疗模式带来了新的机遇和挑战,而局部治疗联合系统治疗产生更好联合治疗作用,提高手术转化率。本文旨在对目前局部联合系统治疗在肝细胞癌转化治疗中的价值,为不可切除肝细胞癌的临床治疗提供参考。Abstract: Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in clinical practice. Due to the lack of typical clinical manifestations in the early stage, most patients in China are in the advanced stage at the time of confirmed diagnosis and thus lose the opportunity for surgical resection, which leads to a poor prognosis. Therefore, it is necessary to explore related therapies for converting unresectable HCC into resectable HCC. In recent years, the improvement in local therapy such as transarterial interventional therapies and radiation therapy technology, together with the clinical application of new targeted therapies and immune checkpoint inhibitors, has brought new opportunities and challenges in the conversion therapy for advanced HCC, and local therapy combined with systemic therapy may have a good synergistic effect, improve the conversion rate of surgery. This article investigates the value of local therapy combined with systemic therapy in the conversion therapy for HCC, in order to provide a basis for the clinical treatment of unresectable HCC.
-
肝细胞癌是最常见的恶性肿瘤之一,占全世界所有癌症新发病例的4.7%,所有癌症相关死亡的8.3%,分别居发病率及死亡率的第六位和第三位[1]。根据我国国家癌症中心发布的数据,2022年肝细胞癌发病率在所有肿瘤中位居第四位,死亡率位居第二位[2]。外科手术(手术切除、射频消融及肝移植)是肝细胞癌的首选治疗方式,但因肝细胞癌起病隐匿,我国大多数肝细胞癌患者在初诊时已属于中晚期(CNLC-Ⅱb期、Ⅲa期和Ⅲb期),从外科学(不能实施安全的R0手术切除)及肿瘤学(切除后的疗效未能超越其他治疗方式)角度,此类患者不宜首选手术治疗,倾向于选择非手术局部治疗及系统治疗以延长生存期[3]。为改善不可切除肝细胞癌患者的预后,转化治疗作为一种新的治疗方式应运而生,即通过前期治疗使不可行手术切除的患者肿瘤降期获得手术切除机会[4]。研究[5]表明,不可切除肝细胞癌患者经转化治疗行手术切除,其5年生存率与初始接受手术的患者生存率相似。
有效降期的转化治疗方案包含以下2个因素:(1)高客观缓解率(objective response rate, ORR)。高ORR与提高手术转化率密切相关,以保证肿瘤缩小和病理坏死的程度,为后续的R0手术切除创造机会;(2)肝功能损伤小,不良反应小。由于我国肝细胞癌患者多有肝炎病史或肝硬化等基础疾病,因此转换治疗方案肝功能损伤小,不良反应相对小,从而确保转化治疗后手术的顺利进行和围手术期的安全性[6]。既往单纯局部治疗的手术转化率较低,限制了转化治疗的应用和推广。近年来,抗血管生成为主的靶向药,如酪氨酸激酶抑制剂(tyrosine kinase inhibitor,TKI)、大分子贝伐珠单抗,与免疫检查点抑制剂联合应用,将客观反应率提高了24.1%~27.3%[7-9],为转化治疗带来了新的希望,使转化率升高到15.9%~30.6%[10-11]。
介入等局部治疗联合靶向药或联合靶向和免疫治疗具有很好的理论联合基础,3种治疗方式的联合可产生更好的协同抗肿瘤作用,近年来在不可切除的肝细胞癌中应用越来越多,客观反应率不断攀升,陆续有报道达40%~60%,为手术转化成功带来更多的可能性,虽然尚无随机对照的转化治疗临床研究,但已有多个单臂研究和大样本的回顾研究报道介入联合系统药物治疗的最新数据。本文旨在探讨介入等局部治疗联合系统治疗在肝细胞癌转化治疗中的价值。
1. 经动脉化疗栓塞(transarterial chemoembolization,TACE)联合系统治疗
TACE通过肿瘤的供血动脉注射液体或颗粒性栓塞剂到达肿瘤末梢血管,使肿瘤缺血坏死,在栓塞材料中携载化疗药,在栓塞的同时在肿瘤滋养血管中释放高浓度的化疗药,实现化疗栓塞的双重目的,更大程度地导致肿瘤细胞的坏死和凋亡。TACE一直被认为是中期肝细胞癌的标准治疗,但是随着肿瘤负荷的增大,TACE治疗效果明显下降,对于中期肝细胞癌(BCLC B期)(Ⅱb期),有报道[12]肿瘤负荷(肿瘤大小+数目)>7的B2期TACE治疗的效果明显低于肿瘤负荷<7的B1期,总生存期(overall survival,OS)从42.3个月降至29.3个月,TACE进展时间从9.5个月降低至5.3个月。Wang等[13]报道,肿瘤负荷>12对比肿瘤负荷6~12,TACE治疗后生存期从32.6个月降低至16.8个月。虽然TACE对于合并Vp1/2门静脉癌栓患者有较好的治疗效果,但对于门静脉重大癌栓的肝细胞癌患者治疗效果有限,有报道[14]单纯TACE对于Vp3/4门静脉癌栓的晚期肝细胞癌,ORR仅为22.7%。因此,多数需要转化治疗的B2期患者或者肿瘤负荷比较大的Ⅱb期,以及合并Vp3/4门静脉癌栓的Ⅲa期患者难以从单纯TACE获得持久有效的转化治疗效果。TACE联合系统药物治疗提高了抗肿瘤治疗效果,扩大了转化治疗的人群和转化成功率。
1.1 TACE联合靶向治疗
TACE联合单纯靶向治疗应用于转化治疗时,多数联合客观反应率比较高的TKI类靶向药,如仑伐替尼。TACE+仑伐替尼对比单用仑伐替尼作为一线治疗晚期肝细胞癌患者疗效的Ⅲ期临床LAUNCH研究[15]中,其中70%患者有门静脉癌栓,55%有肝外转移;TACE+仑伐替尼组患者具有更高的ORR[54.1%(92/170) vs 25.0%(42/168),P<0.001](mRECIST),其中有26例(15.3%)患者因分期降低而行根治性手术切除,2例患者病理完全缓解,而单用仑伐替尼组仅有3例(1.8%)患者接受了根治性手术,且均未出现病理完全缓解。生存获益方面,TACE+仑伐替尼组的中位OS显著长于仑伐替尼组(17.8个月 vs 11.5个月,HR=0.45,P<0.001),这显示出晚期肝细胞癌在使用仑伐替尼的基础上加用TACE可提高疗效及手术转化率,并改善生存。
1.2 TACE联合靶向治疗和免疫治疗
越来越多的研究发现TACE联合TKI类靶向药的同时再联合免疫治疗,比TACE单纯联合靶向治疗能带来更多疗效,更高的客观反应率,且安全性可接受,带来更高的转化率。Chen等[16]回顾性收集了3个临床中心的数据,对比了在PD-L1表达阳性的初始不可切除的肝细胞癌患者中TACE+仑伐替尼与TACE+仑伐替尼+派姆单抗的疗效。三联治疗的ORR[47.1%(33/70) vs 27.8%(20/72)]、疾病控制率(disease control rate,DCR)[70.0%(49/70) vs 52.8%(38/72)]、手术转化率[25.7%(18/70) vs 11.1%(8/72)]显著高于二联治疗。另外,与二联治疗相比,三联治疗显著延长了中位OS(18.1个月 vs 14.1个月,HR=0.56,95%CI:0.38~0.83,P=0.004),将死亡风险降低了44%。对PD-L1表达量进行亚组分析发现,高CPS组更推荐在TACE+仑伐替尼的基础上联合免疫治疗以改善OS。该回顾性研究初步显示出在特定人群中局部联合靶免三联治疗的优越性。复旦大学中山医院的一项研究[17]显示三联治疗(TACE+仑伐替尼+特瑞普利单抗)较二联治疗(TACE+仑伐替尼)取得了更高的ORR(mRECIST)(76.7% vs 47.6%),DCR(mRECIST)(90.0% vs 57.1%),手术转化率[50.0%(15/30) vs 19.0%(4/21)]。安全性方面,在三联治疗组ALT/AST升高是最常见的3级不良反应,在药物干预下可耐受。一项多中心前瞻性单臂临床试验[18]共纳入55例患者行TACE+仑伐替尼+卡瑞丽珠单抗三联治疗,ORR达到76.4%(42/55,完全缓解率为16.4%,部分缓解率为60.0%),DCR为85.5%(47/55)(mRECIST标准)。29例(52.7%)患者转化为可切除,病理缓解(major pathologic response,MPR)率和病理完全缓解(pathological complete response,pCR)率分别为65.5%(19/29)和20.7%(6/29),中位OS和无进展生存期(progression free survival,PFS)未达到。另有一项多中心回顾性研究[19]观察了187例接受TACE+仑伐替尼+抗PD-1抗体治疗的不可切除肝细胞癌患者,其中抗PD-1抗体包括:卡瑞丽珠单抗、信迪力单抗、特瑞普利单抗、替雷利珠单抗、派安普利单抗和派姆单抗。该研究中,77例患者最终达到可切除标准,70例患者接受了手术,转化率为38.7%。29例(41.4%)患者达到了pCR,59例(84.3%)患者达到了MPR。2例(4.3%)患者发生了围手术期死亡,16例(22.9%)患者出现肝切除术后肝衰竭,是最常见的严重并发症,大多数可通过药物干预改善。Li等[20]同样关注了TACE+仑伐替尼+抗PD-1抗体(卡瑞丽珠单抗/信迪力单抗)在转化治疗中治疗效果,在94例接受治疗的患者中ORR为87.2%,DCR为93.6%(mRECIST),32例(34.0%)患者转化成功。研究者进一步将患者分为有早期肿瘤反应者(n=68,72.3%)和无早期肿瘤反应者(n=26,27.7%)。通过亚组分析发现,有早期肿瘤反应的患者的转换手术率明显高于无早期肿瘤反应的患者(44.1% vs 7.7%,P=0.001),且早期肿瘤反应可预测转化治疗的成功(OR=10.296,95%CI:2.076~51.063,P=0.004),具有早期肿瘤反应的患者可获得更长的PFS及OS(15.4个月 vs 7.8个月,P=0.005;23.1个月 vs 12.5个月,P=0.004)。有研究者[21]将TACE+仑伐替尼+抗PD-1抗体应用于预后较差的具有门静脉血栓的患者,ORR和DCR分别为69.8%(74/106)和84.0%(89/106),手术转化率为31.1%(33/106)。中位OS长于24个月,中位PFS是12.53个月。20.8%(22/106)的患者出现3/4级治疗相关不良反应,无治疗相关死亡发生。
TACE联合贝伐珠单抗和免疫检查点抑制剂也带来很好的联合治疗效果,2024年美国临床肿瘤学会胃肠肿瘤研讨会刚刚报道的全球、多中心、随机对照Ⅲ期研究EMERALD-1[22],入组中期肝细胞癌和(Vp1和Vp2型)的晚期肝细胞癌,按照1∶1∶1随机分为3组,A组为TACE+度伐利尤单抗(D+TACE);B组治疗方案为TACE+度伐利尤单抗+贝伐珠单抗(D+B+TACE);C组治疗方案为TACE+安慰剂。主要观察终点为B组对比C组的PFS(RECIST1.1)。研究达到了主要终点,D+B+TACE的中位PFS为15.0个月,而TACE的中位PFS为8.2个月(HR=0.77,95%CI:0.61~0.98,P=0.032)。次要终点方面D+TACE较TACE未能改善PFS(10.0个月 vs 8.2个月,HR=0.94,95%CI:0.75~1.19,P=0.638)。上述结果提示TACE联合贝伐珠单抗和PD-L1免疫检查点抑制剂有望成为有效的转化治疗方案。
2. 肝动脉灌注化疗(hepatic arterial infusion chemotherapy,HAIC)联合系统治疗
HAIC是在影像引导下经皮将导管留置于肝动脉行长期持续灌注化疗,提高肿瘤局部药物浓度,提高化疗效果的介入技术,应用于肝细胞癌的动脉化疗方案为奥沙利铂和5-氟尿嘧啶、顺铂、顺铂+5-氟尿嘧啶、干扰素+ 5-氟尿嘧啶等方案。目前在国内多采用奥沙利铂和5-氟尿嘧啶方案。HAIC技术:推荐采用改良式肝动脉药盒植入方式,也可以采用一次性留管的方式,术中均需完成肝内血流再分布术和肝外血流再分布术,保护胃肠道黏膜的同时,保证全肝的药物灌注。HAIC主要用于Ⅲa期肝细胞癌(尤其是伴有Vp3/Vp4型门静脉癌栓者),或者肿瘤负荷大、边界不清的Ⅱb期肝细胞癌。
2.1 HAIC联合靶向治疗
在HAIC+索拉非尼对比单用索拉非尼治疗存在门静脉侵犯的肝细胞癌患者的Ⅲ期临床研究[23]中,HAIC+索拉非尼组的中位OS为13.37个月,而单用索拉非尼组的中位OS为7.13个月。HAIC+索拉非尼组的ORR高于索拉非尼组[51例(40.8%) vs 3例(2.46%),P<0.001],手术转化率同样较高[12.8%(16/125) vs 0.8%(1/122),P<0.001]。在另一项对比HAIC+索拉非尼与单用索拉非尼在治疗肝细胞癌合并Vp3/4门静脉瘤栓的Ⅱ期随机对照临床研究[24]中,HAIC+索拉非尼组同样获得了更长的中位生存期(16.3个月 vs 6.5个月)及ORR[41%(13/32) vs 3%(1/32),P<0.001],但只有HAIC+索拉替尼组的2例(6%)患者获得了手术机会,其中1例获得pCR。
2.2 HAIC联合靶向治疗和免疫治疗
在一项研究HAIC+信迪力单抗+贝伐珠单抗类似物(IBI305)的前瞻性单臂研究[25]中,ORR为58.6%(17/29)(mRECIST),共14例(48.3%)患者完成手术切除,其中1例(5.3%)达到pCR,5例(26.3%)达到了MPR。Zhang等[26]回顾性研究了HAIC+TKI(阿帕替尼/仑伐替尼/索拉非尼)+抗PD-1抗体(卡瑞丽珠单抗/信迪力单抗)的转化治疗效果,该三联治疗组合的最佳ORR为96.0%(24/25),手术转化率为60%(15/25),其中7例(28%)患者达到了pCR。在另一项大样本量(n=145)的回顾性研究[27]中HAIC+TKI(阿帕替尼/仑伐替尼/索拉非尼)+抗PD-1抗体(卡瑞丽珠单抗/信迪力单抗/替雷利珠单抗)的ORR为57.2%(83/145),手术转化率为18.6%(27/145),其中7例达到了pCR。
2.3 TACE+HAIC联合靶向和免疫
TACE或HAIC与仑伐替尼及抗PD-1抗体联用取得了较好的ORR,有研究[28]进一步观察使用TACE-HAIC这2种局部治疗+仑伐替尼+抗PD-1抗体的转化效果,总体ORR为64.3%(18/28),DCR为89.3%(25/28),经2个周期的联合治疗后,35.7%(10/28)的患者可实现R0切除。
3. 经动脉放射栓塞(transarterial radioembolization,TARE)联合系统治疗
TARE经肝动脉灌注放射性核素标记的栓塞微球,实现肿瘤近距离内照射治疗一种经动脉介入治疗方式,通常使用钇-90作为标记核素。选择性内照射放疗(selective internal radiation therapy,SIRT)通过肝叶和肝段动脉注射,肿瘤局部可以达到更高的吸收剂量,提高治疗效果。TARE对于肿瘤负荷比较大的Ⅱb期肝细胞癌和合并门静脉癌栓的Ⅲa期肝细胞癌显示出优于TACE的疗效。在欧美国家,TARE更多的被应用于肝移植的降期转化方式。一项单中心随机对照研究[29]显示,对于不能手术切除的中期肝细胞癌,TARE较D-TACE有更长的疾病进展时间(17.1个月 vs 9.5个月,P=0.002)和更长的OS(30.2个月 vs 15.6个月,P=0.006),TARE组中38例中有10例降期转化为肝移植,而对照组TACE组34例中,仅有4例降期转化为肝移植。放射性肝段切除是近年来提出的新概念,采用选择性SIRT技术在肿瘤所在肝实现高剂量内照射,研究发现肿瘤>400 Gy的吸收剂量与病理完全坏死密切相关。放射性肝叶切除在肿瘤组织内实现高剂量照射的同时,在肿瘤所在正常肝叶组织也实时有效的照射剂量(>88 Gy),可剩余肝组织的代偿性增生,有助于转化为手术切除[30]。TARE联合靶向药的研究目前尚少,随机对照SORAMIC研究[31]显示,对于中晚期肝细胞癌,索拉非尼联合SIRT较单纯索拉非尼未显示出更长的生存获益。TARE联合PD-1免疫治疗的Ⅱ期临床研究显示出一定的联合获益。CA209-67研究[32]显示,TARE联合纳武利尤单抗的ORR为30.6%,肝内肿瘤ORR为43.5%,OS为16.9个月。Lee等[33]报道的Ⅰ/Ⅱa研究,TARE联合度伐利尤单抗,ORR为83.3%,疾病进展时间为15.2个月。
4. 放射治疗联合系统治疗
肝细胞癌对于放射治疗敏感,中等剂量的放疗即可获得较好的肿瘤缓解率。对于不适合手术切除的肝细胞癌患者,接受放疗后如能转化为手术切除,可获得更长的生存时间。近期一项前瞻性研究[34]探索了TACE+立体定向放疗+抗PD-L1抗体治疗不可切除肝细胞癌的结局,12%(4/33)的患者经过三联疗法后获得了根治性治疗。另一项回顾性研究[35]中共纳入23例接受放疗+派姆单抗+贝伐珠单抗的不可切除肝细胞癌患者,其治疗ORR和DCR分别为34.8%和91.3%。2例(8.7%)局部晚期不可切除的肝细胞癌患者在这种三联治疗后最终接受了根治性肿瘤切除术。
5. 小结与展望
单一局部治疗或者药物治疗不可切除肝细胞癌的ORR及手术转化率低,现有文献数据已表明局部治疗方式联合系统药物治疗可发挥协同作用,提高ORR及手术转化率。TACE作为中期肝细胞癌的标准治疗,对于Ⅱb期和/或合并Vp1/2瘤栓的Ⅲa期肝细胞癌,TACE联合靶向和免疫治疗可提高抗肿瘤治疗效果,提高手术转化成功率。对于肿瘤负荷大、肿瘤边界不清的Ⅱb期、Ⅲa期肝细胞癌(尤其是伴有Vp3/Vp4型门静脉癌栓者),HAIC联合系统靶免治疗转化效果或许更佳。TARE对于Ⅱb期和Ⅲa期肝细胞癌患者有可能产生比TACE更高的手术转化率,但TARE联合靶免的数据不充分,有待于研究证实。现有研究发现局部联合靶免优于局部联合靶向治疗,ORR及手术转化率的提高,未来有待于对免疫治疗获益人群进一步精准筛选,除了PD-L1表达,需发掘更多更有效的生物标志物。另外,目前现有的临床研究多为小样本回顾性研究,仍需设计严谨的大样本Ⅲ临床试验提供更高级别证据以确定最佳的局部联合系统治疗方案、联合时机、最佳获益人群等。
-
[1] SUNG H, FERLAY J, SIEGEL RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2021, 71( 3): 209- 249. DOI: 10.3322/caac.21660. [2] ZHENG RS, CHEN R, HAN BF, et al. Cancer incidence and mortality in China, 2022[J]. Chin J Oncol, 2024, 46( 3): 221- 231. DOI: 10.3760/cma.j.cn112152-20240119-00035.郑荣寿, 陈茹, 韩冰峰, 等. 2022年中国恶性肿瘤流行情况分析[J]. 中华肿瘤杂志, 2024, 46( 3): 221- 231. DOI: 10.3760/cma.j.cn112152-20240119-00035. [3] PARK JW, CHEN MS, COLOMBO M, et al. Global patterns of hepatocellular carcinoma management from diagnosis to death: The BRIDGE Study[J]. Liver Int, 2015, 35( 9): 2155- 2166. DOI: 10.1111/liv.12818. [4] GHOLAM PM, IYER R, JOHNSON MS. Multidisciplinary management of patients with unresectable hepatocellular carcinoma: A critical appraisal of current evidence[J]. Cancers, 2019, 11( 6): 873. DOI: 10.3390/cancers11060873. [5] FINN RS, IKEDA M, ZHU AX, et al. Phase Ib study of lenvatinib plus pembrolizumab in patients with unresectable hepatocellular carcinoma[J]. J Clin Oncol, 2020, 38( 26): 2960- 2970. DOI: 10.1200/JCO.20.00808. [6] HE MK, LIANG RB, ZHAO Y, et al. Lenvatinib, toripalimab, plus hepatic arterial infusion chemotherapy versus lenvatinib alone for advanced hepatocellular carcinoma[J]. Ther Adv Med Oncol, 2021, 13: 17588359211002720. DOI: 10.1177/17588359211002720. [7] KUDO M, FINN RS, QIN SK, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial[J]. Lancet, 2018, 391( 10126): 1163- 1173. DOI: 10.1016/S0140-6736(18)30207-1. [8] FINN RS, QIN SK, IKEDA M, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma[J]. N Engl J Med, 2020, 382( 20): 1894- 1905. DOI: 10.1056/NEJMoa1915745. [9] LLOVET JM, KUDO M, MERLE P, et al. Lenvatinib plus pembrolizumab versus lenvatinib plus placebo for advanced hepatocellular carcinoma(LEAP-002): A randomised, double-blind, phase 3 trial[J]. Lancet Oncol, 2023, 24( 12): 1399- 1410. DOI: 10.1016/S1470-2045(23)00469-2. [10] SUN HC, ZHOU J, WANG Z, et al. Chinese expert consensus on conversion therapy for hepatocellular carcinoma(2021 edition)[J]. Hepatobiliary Surg Nutr, 2022, 11( 2): 227- 252. DOI: 10.21037/hbsn-21-328. [11] TANG HW, CAO YB, JIAN YP, et al. Conversion therapy with an immune checkpoint inhibitor and an antiangiogenic drug for advanced hepatocellular carcinoma: A review[J]. Biosci Trends, 2022, 16( 2): 130- 141. DOI: 10.5582/bst.2022.01019. [12] ARIZUMI T, UESHIMA K, IWANISHI M, et al. The overall survival of patients with hepatocellular carcinoma correlates with the newly defined time to progression after transarterial chemoembolization[J]. Liver Cancer, 2017, 6( 3): 227- 235. DOI: 10.1159/000475777. [13] WANG QH, XIA DD, BAI W, et al. Development of a prognostic score for recommended TACE candidates with hepatocellular carcinoma: A multicentre observational study[J]. J Hepatol, 2019, 70( 5): 893- 903. DOI: 10.1016/j.jhep.2019.01.013. [14] HU JG, BAO Q, CAO G, et al. Hepatic arterial infusion chemotherapy using oxaliplatin plus 5-fluorouracil versus transarterial chemoembolization/embolization for the treatment of advanced hepatocellular carcinoma with major portal vein tumor thrombosis[J]. Cardiovasc Intervent Radiol, 2020, 43( 7): 996- 1005. DOI: 10.1007/s00270-019-02406-3. [15] PENG ZW, FAN WZ, ZHU BW, et al. Lenvatinib combined with transarterial chemoembolization as first-line treatment for advanced hepatocellular carcinoma: A phase III, randomized clinical trial(LAUNCH)[J]. J Clin Oncol, 2023, 41( 1): 117- 127. DOI: 10.1200/JCO.22.00392. [16] CHEN S, WU ZQ, SHI F, et al. Lenvatinib plus TACE with or without pembrolizumab for the treatment of initially unresectable hepatocellular carcinoma harbouring PD-L1 expression: A retrospective study[J]. J Cancer Res Clin Oncol, 2022, 148( 8): 2115- 2125. DOI: 10.1007/s00432-021-03767-4. [17] QU WF, DING ZB, QU XD, et al. Conversion therapy for initially unresectable hepatocellular carcinoma using a combination of toripalimab, lenvatinib plus TACE: Real-world study[J]. BJS Open, 2022, 6( 5): zrac114. DOI: 10.1093/bjsopen/zrac114. [18] WU XK, YANG LF, CHEN YF, et al. Transcatheter arterial chemoembolisation combined with lenvatinib plus camrelizumab as conversion therapy for unresectable hepatocellular carcinoma: A single-arm, multicentre, prospective study[J]. EClinicalMedicine, 2024, 67: 102367. DOI: 10.1016/j.eclinm.2023.102367. [19] WU JY, ZHANG ZB, ZHOU JY, et al. Outcomes of salvage surgery for initially unresectable hepatocellular carcinoma converted by transcatheter arterial chemoembolization combined with lenvatinib plus anti-PD-1 antibodies: A multicenter retrospective study[J]. Liver Cancer, 2023, 12( 3): 229- 237. DOI: 10.1159/000528356. [20] LI XZ, CHEN J, WANG XB, et al. Outcomes and prognostic factors in initially unresectable hepatocellular carcinoma treated using conversion therapy with lenvatinib and TACE plus PD-1 inhibitors[J]. Front Oncol, 2023, 13: 1110689. DOI: 10.3389/fonc.2023.1110689. [21] OU XY, WU JY, WU JY, et al. Efficacy of lenvatinib combined with anti-PD-1 antibodies plus transcatheter arterial chemoembolization for hepatocellular carcinoma with portal vein tumor Thrombus: A retrospective, multicenter study[J]. Cancer Res Treat, 2024. DOI: 10.4143/crt.2023.1165. [22] LENCIONI R, KUDO M, ERINJERI J, et al. EMERALD-1: A phase 3, randomized, placebo-controlled study of transarterial chemoembolization combined with durvalumab with or without bevacizumab in participants with unresectable hepatocellular carcinoma eligible for embolization[J]. J Clin Oncol, 2024, 42( 3_suppl): LBA432. DOI: 10.1200/jco.2024.42.3_suppl.lba432. [23] HE MK, LI QJ, ZOU RH, et al. Sorafenib plus hepatic arterial infusion of oxaliplatin, fluorouracil, and leucovorin vs sorafenib alone for hepatocellular carcinoma with portal vein invasion: A randomized clinical trial[J]. JAMA Oncol, 2019, 5( 7): 953- 960. DOI: 10.1001/jamaoncol.2019.0250. [24] ZHENG KL, ZHU X, FU SJ, et al. Sorafenib plus hepatic arterial infusion chemotherapy versus sorafenib for hepatocellular carcinoma with major portal vein tumor thrombosis: A randomized trial[J]. Radiology, 2022, 303( 2): 455- 464. DOI: 10.1148/radiol.211545. [25] LIU DM, MU H, LIU CF, et al. Sintilimab, bevacizumab biosimilar, and HAIC for unresectable hepatocellular carcinoma conversion therapy: A prospective, single-arm phase II trial[J]. Neoplasma, 2023, 70( 6): 811- 818. DOI: 10.4149/neo_2023_230806N413. [26] ZHANG JL, ZHANG XH, MU H, et al. Surgical conversion for initially unresectable locally advanced hepatocellular carcinoma using a triple combination of angiogenesis inhibitors, anti-PD-1 antibodies, and hepatic arterial infusion chemotherapy: A retrospective study[J]. Front Oncol, 2021, 11: 729764. DOI: 10.3389/fonc.2021.729764. [27] LUO LH, XIAO YQ, ZHU GQ, et al. Hepatic arterial infusion chemotherapy combined with PD-1 inhibitors and tyrosine kinase inhibitors for unresectable hepatocellular carcinoma: A tertiary medical center experience[J]. Front Oncol, 2022, 12: 1004652. DOI: 10.3389/fonc.2022.1004652. [28] ZHAO WC, LIU C, WU YT, et al. Transarterial chemoembolization(TACE)-hepatic arterial infusion chemotherapy(HAIC) combined with PD-1 inhibitors plus lenvatinib as a preoperative conversion therapy for nonmetastatic advanced hepatocellular carcinoma: A single center experience[J]. Transl Cancer Res, 2024, 13( 5): 2315- 2331. DOI: 10.21037/tcr-24-93. [29] GABR A, RIAZ A, JOHNSON GE, et al. Correlation of Y90-absorbed radiation dose to pathological necrosis in hepatocellular carcinoma: Confirmatory multicenter analysis in 45 explants[J]. Eur J Nucl Med Mol Imaging, 2021, 48( 2): 580- 583. DOI: 10.1007/s00259-020-04976-8. [30] GABR A, RIAZ A, MOULI S, et al. Modified radiation lobectomy: An evolving paradigm to convert patients to liver resection candidacy[J]. Semin Intervent Radiol, 2019, 36( 4): 343- 348. DOI: 10.1055/s-0039-1696648. [31] RICKE J, KLÜMPEN HJ, AMTHAUER H, et al. Impact of combined selective internal radiation therapy and sorafenib on survival in advanced hepatocellular carcinoma[J]. J Hepatol, 2019, 71( 6): 1164- 1174. DOI: 10.1016/j.jhep.2019.08.006. [32] KAYA NA, TAI D, LIM X, et al. Multimodal molecular landscape of response to Y90-resin microsphere radioembolization followed by nivolumab for advanced hepatocellular carcinoma[J]. J Immunother Cancer, 2023, 11( 8): e007106. DOI: 10.1136/jitc-2023-007106. [33] LEE YB, NAM JY, CHO EJ, et al. A phase I/IIa trial of yttrium-90 radioembolization in combination with durvalumab for locally advanced unresectable hepatocellular carcinoma[J]. Clin Cancer Res, 2023, 29( 18): 3650- 3658. DOI: 10.1158/1078-0432.CCR-23-0581. [34] CHIANG CL, CHIU KWH, CHAN KSK, et al. Sequential transarterial chemoembolisation and stereotactic body radiotherapy followed by immunotherapy as conversion therapy for patients with locally advanced, unresectable hepatocellular carcinoma(START-FIT): A single-arm, phase 2 trial[J]. Lancet Gastroenterol Hepatol, 2023, 8( 2): 169- 178. DOI: 10.1016/S2468-1253(22)00339-9. [35] LIANG XX, JIANG YH, YAO W, et al. Liver-directed moderately hypo-fractionated radiotherapy combined with pembrolizumab and bevacizumab for advanced hepatocellular carcinoma: A retrospective observational study of 23 cases[J]. Transl Cancer Res, 2024, 13( 3): 1508- 1518. DOI: 10.21037/tcr-23-1333. -