述评丨Liver Cancer主编:“T+A”转化疗法用于不可切除和TACE不适用中期肝细胞癌,根治率高达30%!
——  作者:    时间:2023-09-29     阅读数: 33

编者按
 
原发性肝癌是全球范围内的高发病率和高致死率恶性疾病。近年来随着免疫联合靶向治疗的迅猛发展,一种基于阿替利珠单抗+贝伐珠单抗(Atezo/Bev,T+A)免疫联合治疗的根治性转化疗法(“ABC转化疗法”)引起了学界的高度关注。Liver Cancer杂志(IF:13.8)主编、日本近畿大学医学院Masatoshi Kudo教授为此撰写了一篇社论,就不可切除和不适合经动脉化疗栓塞(TACE)治疗的中期肝细胞癌(HCC)患者采用这一新兴策略的相关进展进行了述评。《国际肝病》特此编译报道,供广大读者学习参考。
 
01
“ABC转化疗法”概述
 
阿替利珠单抗+贝伐珠单抗(Atezo/Bev)后进行根治性转化治疗(“ABC转化疗法”)在治疗不可切除和经动脉化疗栓塞(TACE)不适用的中期肝细胞癌(HCC)方面非常有效。一项多中心研究表明,根治性转化率高达30%。考虑根治性转化的适应证和时机包括:①实现肿瘤缩小;②需要改善反应,即使使用Atezo/Bev无法实现肿瘤缩小;③治疗因肿瘤负荷高以外的原因而不适合TACE治疗的结节,例如融合性多结节型、浸润性或低分化HCC;④由于不良事件而中断Atezo/Bev;⑤正电子发射计算机断层显像(PET)显示HCC阳性,即低分化HCC。
 
IMbrave 150试验显示,Atezo/Bev的总生存期(OS)相比索拉非尼具有压倒性优势[1],基于此Atezo/Bev已在全球范围内获得上市批准。IMbrave 150试验新数据分析显示,Atezo/Bev的表现优于索拉非尼,中位OS为19.2个月[风险比(HR):0.66],中位无进展生存期(PFS)为6.9个月(HR:0.65)[2]。有趣的是,Atezo/Bev在中期(OS:25.8个月;PFS:12.6个月)比晚期(OS:17.5个月;PFS:6.5个月)HCC患者获得了更有利的结果[2]。此外,根据RECIST 1.1版标准,中期HCC患者的客观缓解率(ORR:44%)优于晚期HCC患者(ORR:27%)[2]
 
Atezo/Bev的作用模式与序贯仑伐替尼(LEN)-TACE治疗患者的LEN作用模式完全不同,LEN-TACE序贯治疗方案在中期HCC患者中也显示出良好的结局[3,4]。更准确地说,Atezo/Bev主要引起肿瘤缩小,而LEN主要通过血流减少来导致肿瘤坏死[5](图1)。Atezo/Bev在TACE之前发挥抗-VEGF作用,从而增强TACE的作用,并且还可以转化为根治性治疗,例如肝切除或消融,这在肿瘤缩小后变得可行。事实上,已有一系列病例报道了根治性转化后可实现无癌且无药生存[6,7]
 
图1. 肝细胞癌对Atezo /Bev和仑伐替尼的反应模式
Atezo/Bev诱导基于肿瘤缩小的反应,而LEN诱导基于肿瘤内动脉血流量减少引起的肿瘤坏死的反应。
Atezo/Bev:阿替利珠单抗+贝伐珠单抗;LEN:仑伐替尼。
 
02
成功转化为根治性治疗的比例
 
在参与一项多中心研究的五家机构中,连续101例不可切除和不适合TACE的中期HCC患者中,30例肝功能Child-Pugh A级患者接受一线Atezo/Bev后转化为根治性治疗并实现完全缓解(CR),根治性转化率为30%。在这30例患者中,6例接受了切除术;9例在TACE(ABC-TACE三明治疗法)后接受了消融治疗,包括射频消融术(RFA);14例接受了TACE,包括ABC LEN-TACE三明治疗法;1例在单纯Atezo/Bev治疗后获得治愈。18名患者,包括15名接受切除或消融的患者,目前处于无药物治疗状态,即真正的治愈状态。
 
接受TACE的患者包括那些仍在接受Atezo/Bev治疗的患者,在这些患者中,Atezo/Bev在肿瘤抗原释放后持续激活癌症免疫周期和癌症抗原特异性免疫应答,而TACE诱导肿瘤坏死。因此,尚未获得无药物治疗生存的患者有望在未来实现无药物治疗状态。换言之,大多数达到影像学CR的患者的肿瘤标志物结果变为正常;对于肿瘤标志物结果未恢复正常,且超声造影显示存在活性肿瘤的患者,继续使用Atezo/Bev,必要时联合消融或TACE治疗,大多数患者有望实现CR或深部部分缓解(图2)。需要注意的是,局部区域治疗后必须进行序贯免疫治疗,因为癌症抗原释放和癌症抗原特异性免疫应答可以进一步增强后续Atezo/Bev治疗的有效性[8-19]
 
图2. 阿替利珠单抗+贝伐珠单抗后根治性转化对中期HCC患者的作用
即使Atezo/Bev治疗导致疾病稳定(SD)或部分缓解(PR),患者最终还是会发展为进展性疾病。在不恶化肝功能的情况下,超选择性进行根治性TACE可使30%的患者实现完全缓解(CR)和近70%的患者获得更深的PR。
 
03
转化为根治性治疗的适应证和时机选择
 
关于转化为根治性治疗的适应证和时机,主要有如下五种情况。
 
(一)肿瘤缩小后的根治性转化
 
对于Atezo/Bev治疗后实现肿瘤缩小的患者,无论是否联合TACE,均可能实现根治性治疗(例如切除或消融)。不适合TACE治疗的HCC患者[20,21],特别是融合性多结节型患者,即使在“up-to-seven”标准范围内,也可考虑转化为根治性治疗。如果Atezo/Bev之后序贯TACE达到肿瘤缩小>30%,则最初不可切除的肿瘤已转化为可切除肿瘤。在部分Atezo/Bev治疗极长时间的患者中,可能实现肝切除标本病理分析检测不到活细胞(即实现病理CR)。
 
(二)未实现肿瘤缩小时的根治性转化(ABC-TACE三明治疗法)
 
一项1b期A组的泳道图(swimmer plot)显示,在4个周期的Atezo/Bev后,≥80%的应答者实现了肿瘤缩小≥30%(据RECIST 1.1版评估,为CR或PR)[22]。因此,在<20%的患者中,肿瘤缩小从第五个周期开始变得明显。由于存在这种迟发反应者,原则上应尽可能长时间地持续使用Atezo/Bev,同时保持对晚期HCC患者不良事件的良好控制。在中期HCC患者中,只要有可能,就可以进行局部根治性治疗,例如超选择性TACE。
 
因此,对于在4~6个周期的Atezo/Bev治疗后无反应但疾病稳定或进展缓慢的患者,合理的策略包括在下一个Atezo/Bev周期前不久使用TACE,然后在TACE后肿瘤抗原释放期间尽快恢复和继续Atezo/Bev治疗,该过程被称为ABC-TACE三明治疗法,因为TACE是在Atezo/Bev治疗周期之间进行的。如果在此类患者中,肿瘤数量较少且不损害肝功能的超选择性TACE是可行的,那么根治性TACE可带来无癌和无药物治疗状态(图3)[6]
 
据报道,进行选择性(根治性)TACE以减少肿瘤大小和释放肿瘤抗原,然后再进行4~6个周期的Atezo/Bev治疗,这一过程成功地使一些病情稳定或进展缓慢的患者达到了无癌和无药状态[6]。虽然由于食欲不振导致血清白蛋白水平降低,患者有时会观察到ALBI评分的短暂恶化,但这些患者的食欲很快就改善了,且此后一直保持在初始ALBI分级(mALBI 2级或2a级)。
 
因此,ABC-TACE三明治疗法可能成为实现局部区域根治和无药物状态的重要治疗选择。即使不能通过局部区域治疗达到CR,也可以在维持肝功能的同时达到更深层次的反应(图2)。
 
图3. 中期HCC的2022治疗策略
LEN+TACE是适合TACE且肝功能为Child-Pugh A级的HCC患者的治疗选择。
LEN后序贯TACE是不适合TACE且肝功能为Child-Pugh A级的HCC患者的治疗选择。
LEN-TACE是根治性TACE的一种治疗选择。
ABC转化治疗,包括ABC(LEN)-TACE三明治治疗,是治疗TACE不适用HCC患者的一种选择。
这些策略可以带来高比例的完全缓解(CR)和深度部分缓解(PR),以及持久的无癌和无药物状态。
 
(三)ABC-LEN-TACE三明治疗法用于TACE不适用HCC(因肿瘤负荷高以外因素)
 
不适合TACE的HCC包括低分化HCC、融合性多结节型肿瘤和单纯结节伴外生长型肿瘤,以及弥漫型和块状型肿瘤[20,21]。虽然技术上可实现针对融合性多结节型肿瘤患者的TACE或局部消融,但如果结节小且较少,则TACE和局部消融被视为不合适的肿瘤学治疗选择。融合性多结节型HCC由于包膜完整性差,TACE治疗的效果不佳,仅有5.3%被包裹[21]
 
此外,TACE或局部消融术作为一线治疗与患者预后不良有关,因为多灶性复发的可能性很高[20,21],因为这些患者已经存在微小的门静脉或静脉侵犯和许多肝内微卫星灶[21,23]。因此,前期应给予系统治疗,包括免疫治疗和具有抗VEGF活性的药物,以使肿瘤血管系统正常化并增强药物递送,然后进行TACE和随后的RFA或切除[4,7]
 
小融合性多结节型HCC患者可以接受ABC-LEN-TACE三明治治疗,包括一线Atezo/Bev、在TACE前立即停用Bev、从单独给予Atezo次日起给予低剂量LEN(4~8 mg/d)4周,随后进行序贯LEN-TACE治疗[3,4,7,20,21]。采用这种方法是因为尽管TACE和局部消融在技术上可行,但在肿瘤学上没有指征。前期LEN在使选择性TACE作用最大化方面非常有效,同时可维持肝功能[3,24],建议使用TACE联合LEN治疗,而不是单纯TACE治疗。这可能导致碘油在微卫星灶和门静脉侵犯位置的密集沉积,几乎可实现CR。
 
超声造影可检测到治疗后结节内的残留活肿瘤,但在接受额外4~6个周期的Atezo/Bev治疗后,这些肿瘤会变得完全无法检测到。在TACE诱导肿瘤抗原释放后,Atezo/Bev可通过诱导CD8+T细胞的活化和浸润来增强免疫力[16,25],从而攻击残余肿瘤,从而达到CR[6,7]。ABC-LEN-TACE三明治疗法可使多数患者实现无瘤和无药物治疗状态(图2和3)。
 
(四)因不良事件中断Atezo/Bev期间的根治性转化
 
第四种情况是ABC-TACE三明治疗法,当Atezo/Bev治疗由于不良事件(如蛋白尿或AST/ALT水平升高)而必须中断时。与其等待这些不良事件的消退,不如对最大的结节(而非所有结节)进行选择性TACE治疗,从而保护肝功能并达到远隔效应[16]。该方案在中期HCC患者中显示出良好的预后(IMbrave 150试验中,Atezo/Bev组的中位OS为25.8个月)。
 
由于所有用于二线和后续治疗的分子靶向药物均具有抗VEGF活性,因此这些药物均不能用于在漫长疗程中出现严重蛋白尿的患者。作为替代方案,可以在中断或停药期间进行局部区域治疗,从而减少肿瘤大小并诱导肿瘤抗原释放。这可以增强不良事件消退后Atezo(加Bev)治疗的效果,从而达到无瘤、无药物治疗状态或深度反应(图2)。
 
(五)FDG-PET-阳性HCC的ABC转化
 
PET阳性HCC大多为低分化肿瘤[26-31]。由于其生物侵袭性,这些肿瘤在切除、消融、TACE或移植后经常复发[26,32-34]。此外,这些肿瘤含有角蛋白19(K-19)阳性的癌症干细胞[35],并经历上皮-间充质转化[36]。因此,一般而言,PET阳性HCC患者的预后较差[37-41]。ABC转化可改善这些患者的预后。例如,在PET阳性HCC患者中,切除或RFA后采用ABC-LEN-TACE三明治治疗,未观察到肿瘤复发[7]
 
许多肿瘤的PD-L1、K-19、FGFR2、FGF 19和(或)干细胞特征呈阳性,Atezo/Bev和LEN对此特别有效[42-44],部分患者由于对Atezo/Bev有强烈反应而出现肿瘤溶解综合征。Atezo/Bev治疗后的肿瘤缩小可能会增强肝切除的可行性。此外,PET阳性HCC患者可通过前期Atezo/Bev治疗后行肝切除,或ABC-LEN-TACE三明治治疗后行肝切除,来实现无药状态[45]
 
尽管Bev具有抗VEGF活性,但在TACE治疗前仍给予低剂量LEN,这是因为Bev治疗需要在TACE前3周停止[46]。这使Bev在最后一次给药后3周的抗VEGF活性降到最低。为了补偿,TACE之前的Atezo单药治疗周期内,应同时给予低剂量LEN(VEGFR和FGFR的强抑制剂),从Atezo治疗后的第二天开始,直到TACE前。LEN-TACE序贯治疗会使TACE的疗效最大化,具有非常强的抗肿瘤作用,从而导致CR(mRECIST标准)[3,4,20,21,47]
 
此外,因为LEN具有强抗FGFR2和抗FGFR4活性,低分化HCC患者单使用LEN单药治疗是有效的,ORR分别达到了47.6%[48]和92%[49]。在LEN-TACE后尽快恢复Atezo/Bev特别有效,ABC-LEN-TACE三明治疗法可导致高病理CR率[7,48]。因此,ABC-LEN-TACE三明治疗法可能是目前最有效的治疗方法,可以满足PET阳性HCC患者治疗中未满足的需求。
 
综上所述,这些发现表明,ABC-LEN-TACE三明治疗法可能是一种突破性方案,可以在PET 阳性HCC患者中实现无瘤和无药物状态,最终实现更长的OS(图4)。
 
图4. 2022年更新了不可切除HCC的治疗策略
 
结论
 
这篇社论描述了ABC转化疗法的时机、适应证和结果。根治性治疗变得可行,并且通过联合Atezo/Bev与局部区域治疗或切除可以实现无药物治疗状态,不适合TACE的中期HCC患者的影像学CR率因此可以达到30%左右。ABC转化疗法也可能治愈预后不良的HCC患者,例如PET阳性HCC患者。
 
与其他实体瘤和晚期HCC不同,中期HCC是一种可治愈的疾病。在这些患者中,使用Atezo/Bev诱导,然后进行适当的局部区域治疗,可以达到无瘤和无药物状态,并最终治愈。例如,ABC-LEN-TACE三明治疗法,包括用Atezo/Bev诱导,然后是 LEN-TACE和序贯Atezo/Bev,可能是PET阳性HCC患者的突破性治疗。先前报道的先LEN后TACE方案的有利数据[3,4]表明,这两者必须始终联合使用,LEN被视为诱导治疗(图3和4)。
 
参考文献:
 
[1] Finn?RS, Qin?S, Ikeda?M, Galle?PR, Ducreux?M, Kim?TY, et al; IMbrave150 Investigators. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med. 2020?May;382(20):1894–905.
 
[2] Cheng?AL, Hsu?C, Chan?SL, Choo?SP, Kudo?M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J Hepatol. 2020?Feb;72(2):307–19.
 
[3] Kudo?M, Ueshima?K, Chan?S, Minami?T, Chishina?H, Aoki?T, et al?Lenvatinib as an Initial Treatment in Patients with Intermediate-Stage Hepatocellular Carcinoma Beyond Up-To-Seven Criteria and Child-Pugh A Liver Function: A Proof-Of-Concept Study. Cancers (Basel). 2019?Jul;11(8):11.
 
[4] Kudo?M. A New Treatment Option for Intermediate-Stage Hepatocellular Carcinoma with High Tumor Burden: Initial Lenvatinib Therapy with Subsequent Selective TACE. Liver Cancer. 2019?Oct;8(5):299–311.
 
[5] Kudo?M, Finn?RS, Qin?S, Han?KH, Ikeda?K, Piscaglia?F, et al?Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet. 2018?Mar;391(10126):1163–73.
 
[6] Kudo?M. Hepatocellular carcinoma and NASH. J Gastroenterol. 2004;39(4):409–11.
 
[7] Kudo?M. A Novel Treatment Strategy for Patients with Intermediate-Stage HCC Who Are Not Suitable for TACE: Upfront Systemic Therapy Followed by Curative Conversion. Liver Cancer. 2021?Oct;10(6):539–44.
 
[8] den Brok?MH, Sutmuller?RP, van der Voort?R, Bennink?EJ, Figdor?CG, Ruers?TJ, et al?In situ tumor ablation creates an antigen source for the generation of antitumor immunity. Cancer Res. 2004?Jun;64(11):4024–9.
 
[9] Iida?N, Nakamoto?Y, Baba?T, Nakagawa?H, Mizukoshi?E, Naito?M, et al?Antitumor effect after radiofrequency ablation of murine hepatoma is augmented by an active variant of CC Chemokine ligand 3/macrophage inflammatory protein-1alpha. Cancer Res. 2010?Aug;70(16):6556–65.
 
[10] Mizukoshi?E, Yamashita?T, Arai?K, Sunagozaka?H, Ueda?T, Arihara?F, et al?Enhancement of tumor-associated antigen-specific T cell responses by radiofrequency ablation of hepatocellular carcinoma. Hepatology. 2013?Apr;57(4):1448–57.
 
[11] Mizukoshi?E, Nakamoto?Y, Tsuji?H, Yamashita?T, Kaneko?S. Identification of alpha-fetoprotein-derived peptides recognized by cytotoxic T lymphocytes in HLA-A24+ patients with hepatocellular carcinoma. Int J Cancer. 2006?Mar;118(5):1194–204.
 
[12] Zerbini?A, Pilli?M, Penna?A, Pelosi?G, Schianchi?C, Molinari?A, et al?Radiofrequency thermal ablation of hepatocellular carcinoma liver nodules can activate and enhance tumor-specific T-cell responses. Cancer Res. 2006?Jan;66(2):1139–46.
 
[13] Ayaru?L, Pereira?SP, Alisa?A, Pathan?AA, Williams?R, Davidson?B, et al: Unmasking of alpha-fetoprotein-specific CD4(+) T cell responses in hepatocellular carcinoma patients undergoing embolization. Journal of immunology (Baltimore, Md : 1950) 2007;178:1914-1922.
 
[14] Mizukoshi?E, Nakamoto?Y, Arai?K, Yamashita?T, Sakai?A, Sakai?Y, et al?Comparative analysis of various tumor-associated antigen-specific t-cell responses in patients with hepatocellular carcinoma. Hepatology. 2011?Apr;53(4):1206–16.
 
[15] Zerbini?A, Pilli?M, Laccabue?D, Pelosi?G, Molinari?A, Negri?E, et al?Radiofrequency thermal ablation for hepatocellular carcinoma stimulates autologous NK-cell response. Gastroenterology. 2010?May;138(5):1931–42.
 
[16] Duffy?AG, Ulahannan?SV, Makorova-Rusher?O, Rahma?O, Wedemeyer?H, Pratt?D, et al?Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017?Mar;66(3):545–51.
 
[17] Hiroishi?K, Eguchi?J, Baba?T, Shimazaki?T, Ishii?S, Hiraide?A, et al?Strong CD8(+) T-cell responses against tumor-associated antigens prolong the recurrence-free interval after tumor treatment in patients with hepatocellular carcinoma. J Gastroenterol. 2010?Apr;45(4):451–8.
 
[18] Nobuoka?D, Motomura?Y, Shirakawa?H, Yoshikawa?T, Kuronuma?T, Takahashi?M, et al?Radiofrequency ablation for hepatocellular carcinoma induces glypican-3 peptide-specific cytotoxic T lymphocytes. Int J Oncol. 2012?Jan;40(1):63–70.
 
[19] Hansler?J, Wissniowski?TT, Schuppan?D, Witte?A, Bernatik?T, Hahn?EG, et al?Activation and dramatically increased cytolytic activity of tumor specific T lymphocytes after radio-frequency ablation in patients with hepatocellular carcinoma and colorectal liver metastases. World J Gastroenterol. 2006?Jun;12(23):3716–21.
 
[20] Kudo?M, Han?KH, Ye?SL, Zhou?J, Huang?YH, Lin?SM, et al?A Changing Paradigm for the Treatment of Intermediate-Stage Hepatocellular Carcinoma: Asia-Pacific Primary Liver Cancer Expert Consensus Statements. Liver Cancer. 2020?Jun;9(3):245–60.
 
[21] Kudo?M, Kawamura?Y, Hasegawa?K, Tateishi?R, Kariyama?K, Shiina?S, et al?Management of Hepatocellular Carcinoma in Japan: JSH Consensus Statements and Recommendations 2021 Update. Liver Cancer. 2021?Jun;10(3):181–223.
 
[22] Lee?MS, Ryoo?BY, Hsu?CH, Numata?K, Stein?S, Verret?W, et al; GO30140 investigators. Atezolizumab with or without bevacizumab in unresectable hepatocellular carcinoma (GO30140): an open-label, multicentre, phase 1b study. Lancet Oncol. 2020?Jun;21(6):808–20.
 
[23] Nakashima?Y, Nakashima?O, Tanaka?M, Okuda?K, Nakashima?M, Kojiro?M: Portal vein invasion and intrahepatic micrometastasis in small hepatocellular carcinoma by gross type. Hepatology research : the official journal of the Japan Society of Hepatology 2003;26:142-147.
 
[24] Ueshima?K, Ishikawa?T, Saeki?I, Morimoto?N, Aikata?H, Tanabe?N, et al?Transcatheter arterial chemoembolization therapy in combination strategy with lenvatnib in patients with unresectable hepatocellular carcinoma (TACTICS-L) in Japan: Final analysis.?Gastrointestinal Cancers Symposium (ASCO-GI 2022), San Francisco, USA, January 20-22, 2022
 
[25] Kudo?M. Combination Immunotherapy with Anti-PD-1/PD-L1 Antibody plus Anti-VEGF Antibody May Promote Cytotoxic T Lymphocyte Infiltration in Hepatocellular Carcinoma, Including in the Noninflamed Subclass. Liver Cancer. 2022;11(3):185–91.
 
[26] Seo?S, Hatano?E, Higashi?T, Hara?T, Tada?M, Tamaki?N, et al?Fluorine-18 fluorodeoxyglucose positron emission tomography predicts tumor differentiation, P-glycoprotein expression, and outcome after resection in hepatocellular carcinoma. Clin Cancer Res. 2007?Jan;13(2 Pt 1):427–33.
 
[27] Okazumi?S, Isono?K, Enomoto?K, Kikuchi?T, Ozaki?M, Yamamoto?H, et al: Evaluation of liver tumors using fluorine-18-fluorodeoxyglucose PET: characterization of tumor and assessment of effect of treatment. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 1992;33:333-339.
 
[28] Torizuka?T, Tamaki?N, Inokuma?T, Magata?Y, Sasayama?S, Yonekura?Y, et al: In vivo assessment of glucose metabolism in hepatocellular carcinoma with FDG-PET. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 1995;36:1811-1817.
 
[29] Rigo?P, Paulus?P, Kaschten?BJ, Hustinx?R, Bury?T, Jerusalem?G, et al?Oncological applications of positron emission tomography with fluorine-18 fluorodeoxyglucose. Eur J Nucl Med. 1996?Dec;23(12):1641–74.
 
[30] Nagaoka?S, Itano?S, Ishibashi?M, Torimura?T, Baba?K, Akiyoshi?J, et al: Value of fusing PET plus CT images in hepatocellular carcinoma and combined hepatocellular and cholangiocarcinoma patients with extrahepatic metastases: preliminary findings. Liver international : official journal of the International Association for the Study of the Liver 2006;26:781-788.
 
[31] Sacks?A, Peller?PJ, Surasi?DS, Chatburn?L, Mercier?G, Subramaniam?RM. Value of PET/CT in the management of liver metastases, part 1. AJR Am J Roentgenol. 2011?Aug;197(2):W256-9.
 
[32] Kitamura?K, Hatano?E, Higashi?T, Seo?S, Nakamoto?Y, Yamanaka?K, et al?Preoperative FDG-PET predicts recurrence patterns in hepatocellular carcinoma. Ann Surg Oncol. 2012?Jan;19(1):156–62.
 
[33] Morio?K, Kawaoka?T, Aikata?H, Namba?M, Uchikawa?S, Kodama?K, et al?Preoperative PET-CT is useful for predicting recurrent extrahepatic metastasis of hepatocellular carcinoma after resection. Eur J Radiol. 2020?Mar;124:108828.
 
[34] Yaprak?O, Acar?S, Ertugrul?G, Dayangac?M. Role of pre-transplant 18F-FDG PET/CT in predicting hepatocellular carcinoma recurrence after liver transplantation. World J Gastrointest Oncol. 2018?Oct;10(10):336–43.
 
[35] Kawai?T, Yasuchika?K, Seo?S, Higashi?T, Ishii?T, Miyauchi?Y, et al?Identification of Keratin 19-Positive Cancer Stem Cells Associating Human Hepatocellular Carcinoma Using 18F-Fluorodeoxyglucose Positron Emission Tomography. Clin Cancer Res. 2017?Mar;23(6):1450–60.
 
[36] Lee?M, Jeon?JY, Neugent?ML, Kim?JW, Yun?M. 18F-Fluorodeoxyglucose uptake on positron emission tomography/computed tomography is associated with metastasis and epithelial-mesenchymal transition in hepatocellular carcinoma. Clin Exp Metastasis. 2017?Apr;34(3-4):251–60.
 
[37] Asman?Y, Evenson?AR, Even-Sapir?E, Shibolet?O. [18F]fludeoxyglucose positron emission tomography and computed tomography as a prognostic tool before liver transplantation, resection, and loco-ablative therapies for hepatocellular carcinoma. Liver Transpl. 2015?May;21(5):572–80.
 
[38] Asman?Y, Evenson?AR, Even-Sapir?E, Shibolet?O. [18F]fludeoxyglucose positron emission tomography and computed tomography as a prognostic tool before liver transplantation, resection, and loco-ablative therapies for hepatocellular carcinoma. Liver Transpl. 2015?May;21(5):572–80.
 
[39] Song?HJ, Cheng?JY, Hu?SL, Zhang?GY, Fu?Y, Zhang?YJ. Value of 18F-FDG PET/CT in detecting viable tumour and predicting prognosis of hepatocellular carcinoma after TACE. Clin Radiol. 2015?Feb;70(2):128–37.
 
[40] Na?SJ, Oh?JK, Hyun?SH, Lee?JW, Hong?IK, Song?BI, et al: (18)F-FDG PET/CT Can Predict Survival of Advanced Hepatocellular Carcinoma Patients: A Multicenter Retrospective Cohort Study. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 2017;58:730-736.
 
[41] Sposito?C, Di Sandro?S, Brunero?F, Buscemi?V, Battiston?C, Lauterio?A, et al?Development of a prognostic scoring system for resectable hepatocellular carcinoma. World J Gastroenterol. 2016?Sep;22(36):8194–202.
 
[42] Llovet?JM, Castet?F, Heikenwalder?M, Maini?MK, Mazzaferro?V, Pinato?DJ, et al?Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol. 2022?Mar;19(3):151–72.
 
[43] Montironi?C, Castet?F, Haber?PK, Pinyol?R, Torres-Martin?M, Torrens?L, et al?Inflamed and non-inflamed classes of HCC: a revised immunogenomic classification. Gut. 2022?Feb;gutjnl-2021-325918.
 
[44] Harimoto?N, Taguchi?K, Shirabe?K, Adachi?E, Sakaguchi?Y, Toh?Y, et al?The significance of fibroblast growth factor receptor 2 expression in differentiation of hepatocellular carcinoma. Oncology. 2010;78(5-6):361–8.
 
[45] Kudo?M. Atezolizumab plus Bevacizumab Followed by Curative Conversion (ABC Conversion) in Patients with Unresectable, TACE Unsuitable Intermediate-satage Hepatocellular Carcinoma. Kan Tan Sui. Hepato-Biliary and Pancreas; 2022.[(in Japanese)].
 
[46] Benson?AB, Venook?AP, Al-Hawary?MM, Arain?MA, Chen?YJ, Ciombor?KK, et al?Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2021?Mar;19(3):329–59.
 
[47] Kudo?M. Lenvatinib may drastically change the treatment landscape of hepatocellular carcinoma. Liver Cancer. 2018?Mar;7(1):1–19.
 
[48] Kudo?M, Ueshima?K, Aikata?H, Tamai?T, Saito?K, Ikeda?K. Association between tumor response by mRECIST and overall survival in patients with poorly differentiated HCC in REFLECT study. 10th Asia-Pacific Primary Liver Cancer Expert Meeting 2019, Sapporo, Aug?31, 2019.
 
[49] Kawamura?Y, Kobayashi?M, Shindoh?J, Kobayashi?Y, Kasuya?K, Sano?T, et al?Pretreatment Heterogeneous Enhancement Pattern of Hepatocellular Carcinoma May Be a Useful New Predictor of Early Response to Lenvatinib and Overall Prognosis. Liver Cancer. 2020?Jun;9(3):275–92.
 
原文链接:Masatoshi Kudo. Atezolizumab plus Bevacizumab Followed by Curative Conversion (ABC Conversion) in Patients with Unresectable, TACE-Unsuitable Intermediate-Stage Hepatocellular Carcinoma. Liver Cancer. 2022 Jul 27;11(5):399-406. doi: 10.1159/000526163.
 
https://karger.com/lic/article/11/5/399/824950/Atezolizumab-plus-Bevacizumab-Followed-by-Curative
 
 
声明:本文仅供医疗卫生专业人士了解最新医药资讯参考使用,不代表本平台观点。该信息不能以任何方式取代专业的医疗指导,也不应被视为诊疗建议,如果该信息被用于资讯以外的目的,本站及作者不承担相关责任。

发表评论

全部评论