肝细胞癌免疫治疗的现状及发展

您所在的位置:网站首页 小鼠肝细胞生长阶段 肝细胞癌免疫治疗的现状及发展

肝细胞癌免疫治疗的现状及发展

2024-06-27 08:34| 来源: 网络整理| 查看: 265

Sichuan Da Xue Xue Bao Yi Xue Ban. 2023 May 20; 54(3): 692–698. Chinese. doi: 10.12182/20230560108PMCID: PMC10475433PMID: 37248607

Language: Chinese | English

肝细胞癌免疫治疗的现状及发展Status Quo and Development of Immunotherapy for Hepatocellular Carcinoma焘 王1,2 and 文涛 王1,Δ焘 王

1 四川大学华西医院 普外科 肝脏外科 (成都 610041), Divsion of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China

2 四川大学华西医院 生物治疗科/肿瘤中心和生物治疗全国重点实验室 (成都 610041), Department of Biotherapy/Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China

Find articles by 焘 王文涛 王

1 四川大学华西医院 普外科 肝脏外科 (成都 610041), Divsion of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China

Find articles by 文涛 王Author information Article notes Copyright and License information PMC Disclaimer 1 四川大学华西医院 普外科 肝脏外科 (成都 610041), Divsion of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China 2 四川大学华西医院 生物治疗科/肿瘤中心和生物治疗全国重点实验室 (成都 610041), Department of Biotherapy/Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China 焘 王: moc.361@9121oatuohsgnaw; 文涛 王: moc.621@20rotcodtww E-mail:moc.361@20rotcodtww Received 2022 Aug 30Copyright 版权所有©《四川大学学报(医学版)》编辑部 2023Copyright ©2023 JOURNAL OF SICHUAN UNIVERSITY (MEDICAL SCIENCE EDITION). All rights reserved.Abstract

肝癌是一种严重的全球健康问题,也是常见的癌症相关死亡原因。肝细胞癌(hepatocellular carcinoma, HCC)是肝癌常见的病理类型。早期HCC临床症状不明显,50%的HCC患者确诊时已处于晚期。系统全身治疗被推荐用于晚期HCC。随着分子靶向药物(索拉非尼、仑伐替尼)的发展,晚期HCC的系统全身治疗取得了一定进展,但对HCC患者生存获益仍然不大。近年来,免疫检查点抑制剂的出现改变了HCC治疗的格局,为HCC精准治疗提供了更多的可能性并展现出较好的效果。特别是阿替利珠单抗和贝伐珠单抗的联合疗法显著改善了HCC患者的生存预后,此外,过继性细胞疗法、肿瘤疫苗、溶瘤病毒和非特异性免疫治疗也已成为免疫治疗策略。本文就HCC免疫治疗的现状及发展进行概述。

Keywords: 肝细胞癌免疫治疗, 免疫检查点抑制剂, 过继性细胞疗法, 肿瘤疫苗, 溶瘤病毒Abstract

Liver cancer is a serious global health problem and a common cause of cancer-related death. Hepatocellular carcinoma (HCC) is a common pathological type of liver cancer. The clinical symptoms of early HCC tend to be not obvious and 50% of HCC patients are already in the advanced stage by the time they are diagnosed. Systemic therapy is recommended for the treatment of advanced HCC. With the development of molecular targeted drugs (sorafenib and lenvatinib), some progress has been made in the systemic treatment of advanced HCC, but there is only modest benefit for the survival of HCC patients. In recent years, the emergence of immune checkpoint inhibitors has changed the overall outlook of HCC treatment, providing more possibilities for precise treatment of HCC and showing better treatment outcomes. In particular, the combination therapy of atezolizumab and bevacizumab significantly improved the survival outcomes in HCC patients. In addition, adoptive cell therapy, tumor vaccine, oncolytic viruses, and nonspecific immunotherapy have also emerged as strategies for immunotherapy. Herein, the status quo and development of HCC immunotherapy are reviewed.

Keywords: Immunotherapy for hepatocellular carcinoma, Immune checkpoint inhibitors, Adoptive cell therapy, Tumor vaccine, Oncolytic virus

原发性肝癌是全球第六大常见癌症和第四大癌症死亡原因[1]。从2007年到2016年,肝癌发病率以每年2%~3%的速度递增,死亡率也快速上升,近年来逐渐趋于平稳[2],但肝癌仍然是5年生存率(18%)最低的恶性肿瘤之一[3],2020年原发性肝癌新发病例已超过900000例,死亡人数约为830000例[4]。据估计,到2025年后,全球每年肝癌患病人数将超过100万[5]。肝细胞癌(hepatocellular carcinoma, HCC)是原发性肝癌中最常见的病理类型。中国是HCC高发国家,新发病例数量占据全球的47%[6]。乙型肝炎病毒(hepatitis B virus, HBV)和丙型肝炎病毒(hepatitis C virus, HCV)引起的慢性肝病是诱发HCC最常见的病因,其次为过量饮酒、与代谢综合征相关的非酒精性脂肪肝病等[7]。

HCC的治疗以肝切除术、肝移植、局部治疗〔消融、经动脉化疗栓塞(transcatheter arterial chemoembolization, TACE)、放疗等〕、系统全身治疗等为主[5]。HCC患者临床治疗方案的具体选择取决于临床分期。根据国际通用的巴塞罗那肝癌(BCLC)分期系统,HCC患者分为五个临床阶段[8]。肝切除术和肝移植是治疗早期HCC患者的主要方法,据报道早期HCC患者5年生存率可超过70%[5]。但由于早期HCC临床症状不明显,50%的HCC患者确诊时已处于晚期甚至是终末期,不符合手术治疗指征,远期预后极差,中位生存期仅为1~1.5年,系统全身治疗是唯一的选择[9]。在索拉非尼出现之前,晚期HCC的系统全身治疗仅限于细胞毒性药物,但多项大型的随机对照研究发现HCC对系统化疗的反应率低(2的效果。来自全球多中心Ⅲ期HIMALAYA研究发现,PD-L1单抗(度伐利尤单抗)联合CTLA-4单抗(替西木单抗)(STRIDE方案)治疗不可切除的HCC可显著改善患者生存状况,3年生存率高达30.7%,而索拉非尼对照组仅为20%,且3~4级治疗相关不良事件发生率明显降低[26-27]。HIMALAYA研究具有重要的意义,STRIDE方案有望成为HCC新的一线标准治疗方案。晚期肝癌免疫治疗关键临床试验结果可见表1。

表 1Results of key clinical trials of immunotherapies for advanced hepatocellular carcinoma

晚期肝癌免疫治疗关键临床试验结果

StudyTreatment optionsClinical trial stageOutcomeORR%mOSmPFSOther ORR: objective response rate; mPFS: median progression-free survival; mOS: median overall survival; OS: overall survival; mTTP: median time to progression; DCR: disease control rate; mDOR: median duration of response; mTTR: median response time.CheckMate 040[16]NivolumabⅠ/Ⅱ2015.64.0KEYNOTE-224[19]PembrolizumabⅡ1712.94.9Richard, et al[21]Pembrolizumab + lenvatinibⅠb36228.6mTTP 9.7 monthsIMbrave 150[22]Atezolizumab + bevacizumabⅢ3019.26.9DCR 74%, OS of 12 months 67%, OS of 18 months 52%ORIENT-32[24]Sintilimab + bevacizumabⅡ/Ⅲ254.6RESCUE[25]Camrelizumab + ApatinibⅡ34.35.7

OS of 9 months 86.7%, OS of 12 months 74.7%, OS of 18 months 58.1%, DCR 77.1%, mDOR 14.8 months

HIMALAYA[27]Durvalumab + tremelimumabⅢ20.116.43.8mDOR 22.34 months, mTTR 2.17 months,3-year survival rate 30.7%Open in a separate window

此外,越来越多的研究探索免疫治疗联合化疗或局部治疗对HCC的有效性及安全性。有研究发现化疗药物可以通过激活树突状细胞、增强T细胞的交叉启动以及下调MDSC和Treg细胞来改善抗肿瘤免疫反应并诱导肿瘤细胞免疫原性死亡[28], HCC的局部治疗可以通过释放炎症因子和肿瘤特异性抗原杀死肿瘤细胞来提高抗肿瘤能力[29-32]。同时,TACE联合纳武利尤单抗、TACE联合贝伐珠单抗和德瓦鲁单抗以及根治性外科手术/消融治疗联合度伐利尤单抗及贝伐珠单抗的几项临床研究正在进行中[33-34],探索不同局部治疗及免疫治疗联合的方案,将进一步推动HCC的精准治疗。ICI其他潜在的靶点还包括T细胞免疫球蛋白及黏蛋白结构域分子3(T-cell immunoglobulin and mucin domain-containing molecule 3, TIM3)和淋巴细胞活化基因-3(lymphocyte-activation gene 3, LAG-3),它们与PD-1一样在T细胞上表达,并可导致T细胞受抑制及功能障碍。有研究发现当巨噬细胞中的TIM-3被耗竭时,会抑制HCC肿瘤的生长[35]。此外,LAG-3在包括HCC在内的多种恶性肿瘤中具有异常表达[36]。因此,针对TIM-3和LAG-3靶点的免疫治疗可能为HCC患者带来获益。PD-1联合LAG-3以及PD-1联合TIM-3的双重ICI阻断的Ⅱ期实验正在进行中[37-38]。

迄今为止,基于ICI的晚期HCC免疫治疗应用虽取得了一定的效果,但仍存在许多局限性和挑战,例如仍有超过2/3的患者对免疫治疗反应性差,生存获益依然有限。现有的研究发现肿瘤基因组特征、肿瘤免疫检查点表达水平、肿瘤微环境组成、肠道菌群、肿瘤微卫星不稳定性、肿瘤突变负荷可以帮助进行ICI免疫治疗预后进行分层[39-42],但HCC发病机制复杂,单一的生物标志物并不能够准确预测HCC患者免疫治疗的预后,因此有必要进一步探索上述标志物的联合预测价值。

2. 过继性细胞疗法

在免疫治疗临床实践中,由于患者体内缺乏识别肿瘤特异性抗原的T细胞,部分患者在应用基于ICI的免疫治疗后仍无法依靠自身T细胞完全消灭肿瘤。因此,有人提出采用过继性细胞疗法,即从患者的肿瘤或外周血中提取免疫细胞在体外进行基因工程以增强细胞免疫功能,然后再输入体内,达到杀伤肿瘤细胞的作用[43]。过继性细胞疗法主要涉及到识别肿瘤细胞表达的相关抗原,并针对这些特异性肿瘤抗原激活和扩增自身幼稚的T细胞。目前过继性细胞疗法以嵌合抗原受体(chimeric antigen receptor, CAR)T细胞疗法(CAR-T)发展最为迅速。CAR-T细胞可以特异性靶向肿瘤特异性抗原并以不依赖主要组织相容性复合体(major histocompatibility complex, MHC)的方式特异性识别多种抗原,解决MHC分子下调引起的免疫逃逸[44]。CAR-T 细胞在清除肿瘤细胞的同时,也有可能错误地攻击正常组织细胞产生脱靶效应,使得CAR-T细胞疗法不能得到预期的治疗效果,因此选择合适的HCC抗原作为免疫治疗选择非常重要。磷脂酰肌醇蛋白聚糖3(glypican 3, GPC3)在HCC肿瘤细胞表面高表达,而在正常组织中不表达或低表达,因此其可作为免疫治疗的良好靶点[45]。

GAO等[46]首次构建了以GPC3为靶点的CAR-T细胞,并证明GPC3为靶点CAR-T细胞在体外和体内均可有效抑制HCC细胞的生长。另一项研究通过建立患者来源的异种移植小鼠(patient-derived tumor xenograft model, PDX)HCC模型,证明GPC3为靶点CAR-T细胞抑制了肿瘤生长,但由于肿瘤细胞上PD-L1的表达不同而具有不同的功效[47]。这表明CAR-T疗法和ICI的结合是一种可行的策略,并对PD-L1阳性HCC患者有着更佳的疗效。一项涉及13例中国难治性或复发性GPC3阳性HCC患者的Ⅰ期研究显示GPC3为靶点CAR-T是安全可行的,并具有良好的抗肿瘤潜力[48]。除GPC3外,针对其他肿瘤抗原,包括甲胎蛋白(alpha fetoprotein, AFP)、HBV表面抗原、自然杀伤细胞家族2成员D(natural killer group 2 member D, NKG2D)、细胞间质上皮转换因子(cellular-mesenchymal epithelial transition factor, c-MET)等的CAR-T细胞实验在基础研究中展现出不错的效果,但尚未在临床研究中进行测试[49-52],我们期待上述临床研究的结果。迄今为止,美国食品药品监督管理局已批准两种不同的CAR-T细胞疗法治疗血液系统恶性肿瘤[53-54]。

然而,不同于血液系统恶性肿瘤,CAR-T可以直接靶向恶性细胞,在HCC实体肿瘤中,CAR-T需要运输到肿瘤病灶才能与其靶点结合,在这个过程中往往受到肿瘤微环境的限制:首先,肿瘤周边大量和异常的新生血管、细胞外基质等造成的物理屏障使CAR-T难以有效地达到肿瘤组织[55];其次,实体肿瘤形成的免疫微环境具有低氧、低营养、低pH、高渗透的特点,不利于CAR-T细胞的存活[56],肿瘤微环境中MDSC、CAF、肿瘤相关巨噬细胞(tumor-associated macrophages, TAMs)和Treg等免疫抑制细胞也会通过多种机制使T细胞失活;此外,实体恶性肿瘤通常也会分泌趋化因子以阻碍T细胞的迁移和渗透[57]。因此我们有必要进一步探索肿瘤微环境在CAR-T细胞治疗HCC中的作用,提高CAR-T细胞治疗的有效性。

3. 肿瘤疫苗和溶瘤病毒

肿瘤疫苗是通过使用人工设计的肿瘤抗原来激发细胞毒性T细胞帮助增强抗肿瘤免疫反应的策略[58]。目前关于HCC的肿瘤疫苗主要有多肽类疫苗、DNA疫苗、RNA疫苗、树突状细胞疫苗等。溶瘤病毒(oncolytic virus, OV)是天然存在的或经过工程改造的病毒,OV主要通过直接溶解肿瘤细胞和诱导机体产生抗肿瘤免疫反应发挥治疗作用,其特征在于优先感染、复制和裂解恶性肿瘤细胞,靶向杀伤肿瘤,同时不破坏正常组织;表达肿瘤特异性免疫激活蛋白,激发全身性抗肿瘤免疫反应。

在一项Ⅰ/Ⅱ期临床试验中({"type":"clinical-trial","attrs":{"text":"NCT03480152","term_id":"NCT03480152"}}NCT03480152),在5例HCC患者身上测试了一种含有15种特异性新抗原的基于mRNA的个性化癌症疫苗,该研究最近已经完成,正在等待试验结果公布。基于AFP的肿瘤疫苗曾在早期试验中使用,并已扩展到其他肿瘤抗原例如GPC3和人端粒酶逆转录酶(human telomerase reverse transcriptase, hTERT)上[59],但上述肿瘤疫苗的治疗有效率并不高,这种不良结果的产生可能与肿瘤疫苗的抗原靶标范围不足和肝脏存在较强的免疫抑制微环境相关。但要明确其关联性,还需要进一步探索肿瘤疫苗与ICI以及其他分子靶向药物对肿瘤的治疗效果。目前,HCC的肿瘤疫苗开发已经进行了一系列的临床试验,以评估癌症疫苗在不同阶段的HCC患者中的安全性和效率,但是相关临床试验结果尚未公开。OV治疗是一种新兴的免疫疗法,许多临床前研究和临床研究已经证明其具有抗肿瘤功效。溶瘤牛痘病毒Pexa-Vec是HCC中研究最广泛的溶瘤病毒,在之前的研究中已显示出临床有效性和耐受性[60],但一项Ⅲ期临床试验(PHOCUS研究)显示尽管在治疗过程中Pexa-Vec联合索拉非尼效果好于单用索拉非尼组,但是中期分析结果表明其延长患者生存期的可能性不高,因此该试验被提前终止。在未来,我们需要更多的临床结果来验证溶瘤病毒在HCC治疗中的效果。

4. 非特异性免疫治疗

目前用于HCC治疗的非特异免疫治疗主要包括胸腺肽、干扰素(interferon, IFN)、IL-2等,其具有改善机体免疫力,促进T细胞增殖分化,增强T细胞对肿瘤抗原反应,刺激NK细胞增殖,增强NK细胞杀伤活性等作用。一项研究发现对于不可切除的晚期HCC,采用TACE联合胸腺肽治疗与单独使用TACE相比,有更高的肿瘤反应率并可延长患者生存时间[61]。一项大样本的回顾性研究分析也证实针对小HCC患者手术切除术后联合使用胸腺肽治疗可改善患者的预后[62]。IFN-α作为HCC手术切除后的辅助治疗药物已得到广泛研究,其不仅可调节宿主免疫功能,还可以抑制HBV的复制。一项研究发现IFN-α治疗可以改善HBV相关HCC患者根治性肝切除术后的长期生存期[63]。IL-2也已被用于治疗晚期HCC,有研究发现对于晚期失去手术机会的HCC患者,使用IL-2治疗后,患者的生存率得到提高[64]。但是上述细胞因子治疗毒副作用较大,总体的疗效也需要大样本量、多中心参与的随机对照临床试验去证实。

5. 小结与展望

综上,现有的免疫治疗研究证据为HCC治疗提供了更多的可能性,多种免疫治疗方案表现出潜在的临床获益,但目前仍存在一些瓶颈,例如HCC内部具有显著的异质性,不同的肿瘤细胞在形态学、分子遗传学和免疫学等方面存在显著差异,不同的分子亚型会导致HCC对免疫治疗的反应差异很大,因此,需要开发更加精准的治疗方案。此外,HCC免疫微环境中免疫抑制细胞、肿瘤细胞分泌的免疫抑制分子等会导致HCC对免疫治疗的反应减弱。为了克服免疫治疗在HCC治疗中的限制,在未来,需要继续探索免疫治疗联合手术、靶向药物、消融、放疗、介入等多种治疗模式,增强HCC对免疫治疗的反应。同时需要确定HCC分子亚型,以更好地预测治疗反应,从而整体提高HCC免疫治疗的效果。

*    *    *

利益冲突 所有作者均声明不存在利益冲突

Funding Statement

国家自然科学基金面上项目(No. 82170543)、四川省自然科学基金面上项目(No. 2023NSFSC0612)、四川省科技计划重点研发项目(No. 2023YFS0229)和四川大学专职博士后研发基金项目(No. 2023SCU12056)资助

References1. YANG J D, HAINAUT P, GORES G J, et al A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16(10):589–604. doi: 10.1038/s41575-019-0186-y. [PMC free article] [PubMed] [CrossRef] [Google Scholar]2. SIEGEL R L, MILLER K D, FUCHS H E, et al Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33. doi: 10.3322/caac.21708. [PubMed] [CrossRef] [Google Scholar]3. STORANDT M H, MAHIPAL A, TELLA S H, et al Systemic therapy in advanced hepatocellular carcinoma: patient selection and key considerations. J Hepatocell Carcinoma. 2022;9:1187–1200. doi: 10.2147/jhc.S365002. [PMC free article] [PubMed] [CrossRef] [Google Scholar]4. SUNG H, FERLAY J, SIEGEL R L, et al Global cancer statistics 2020: GLOBOCAN Estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi: 10.3322/caac.21660. [PubMed] [CrossRef] [Google Scholar]5. LLOVET J M, KELLEY R K, VILLANUEVA A, et al Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7(1):6. doi: 10.1038/s41572-020-00240-3. [PubMed] [CrossRef] [Google Scholar]6. FERLAY J, COLOMBET M, SOERJOMATARAM I, et al Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144(8):1941–1953. doi: 10.1002/ijc.31937. [PubMed] [CrossRef] [Google Scholar]7. SINGAL A G, LAMPERTICO P, NAHON P Epidemiology and surveillance for hepatocellular carcinoma: new trends. J Hepatol. 2020;72(2):250–261. doi: 10.1016/j.jhep.2019.08.025. [PMC free article] [PubMed] [CrossRef] [Google Scholar]8. REIG M, FORNER A, RIMOLA J, et al BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol. 2022;76(3):681–693. doi: 10.1016/j.jhep.2021.11.018. [PMC free article] [PubMed] [CrossRef] [Google Scholar]9. YANG C, ZHANG H, ZHANG L, et al Evolving therapeutic landscape of advanced hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2023;20(4):203–222. doi: 10.1038/s41575-022-00704-9. [PubMed] [CrossRef] [Google Scholar]10. THOMAS M B, O'BEIRNE J P, FURUSE J, et al Systemic therapy for hepatocellular carcinoma: cytotoxic chemotherapy, targeted therapy and immunotherapy. Ann Surg Oncol. 2008;15(4):1008–1014. doi: 10.1245/s10434-007-9705-0. [PubMed] [CrossRef] [Google Scholar]11. LLOVET J M, RICCI S, MAZZAFERRO V, et al Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–390. doi: 10.1056/NEJMoa0708857. [PubMed] [CrossRef] [Google Scholar]12. KUDO M, FINN R S, QIN S, et al Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet. 2018;391(10126):1163–1173. doi: 10.1016/s0140-6736(18)30207-1. [PubMed] [CrossRef] [Google Scholar]13. BRUIX J, QIN S, MERLE P, et al Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;389(10064):56–66. doi: 10.1016/s0140-6736(16)32453-9. [PubMed] [CrossRef] [Google Scholar]14. PRIETO J, MELERO I, SANGRO B Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2015;12(12):681–700. doi: 10.1038/nrgastro.2015.173. [PubMed] [CrossRef] [Google Scholar]15. HUANG S L, WANG Y M, WANG Q Y, et al Mechanisms and clinical trials of hepatocellular carcinoma immunotherapy. Front Genet. 2021;12:691391. doi: 10.3389/fgene.2021.691391. [PMC free article] [PubMed] [CrossRef] [Google Scholar]16. El-KHOUEIRY A B, SANGRO B, YAU T, et al Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet. 2017;389(10088):2492–2502. doi: 10.1016/s0140-6736(17)31046-2. [PMC free article] [PubMed] [CrossRef] [Google Scholar]17. ZHU A X, FINN R S, EDELINE J, et al Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. Lancet Oncol. 2018;19(7):940–952. doi: 10.1016/s1470-2045(18)30351-6. [PubMed] [CrossRef] [Google Scholar]18. YAU T, PARK J W, FINN R S, et al Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): a randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022;23(1):77–90. doi: 10.1016/s1470-2045(21)00604-5. [PubMed] [CrossRef] [Google Scholar]19. FINN R S, RYOO B Y, MERLE P, et al Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: a randomized, double-blind, phase Ⅲ trial. J Clin Oncol. 2020;38(3):193–202. doi: 10.1200/jco.19.01307. [PubMed] [CrossRef] [Google Scholar]20. KATO Y, TABATA K, KIMURA T, et al Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One. 2019;14(2):e0212513. doi: 10.1371/journal.pone.0212513. [PMC free article] [PubMed] [CrossRef] [Google Scholar]21. FINN R S, IKEDA M, ZHU A X, et al Phase Ⅰb study of lenvatinib plus pembrolizumab in patients with unresectable hepatocellular carcinoma. J Clin Oncol. 2020;38(26):2960–2970. doi: 10.1200/jco.20.00808. [PMC free article] [PubMed] [CrossRef] [Google Scholar]22. GALLE P R, FINN R S, QIN S, et al Patient-reported outcomes with atezolizumab plus bevacizumab versus sorafenib in patients with unresectable hepatocellular carcinoma (IMbrave150): an open-label, randomised, phase 3 trial. Lancet Oncol. 2021;22(7):991–1001. doi: 10.1016/s1470-2045(21)00151-0. [PubMed] [CrossRef] [Google Scholar]23. QIN S, REN Z, FENG Y H, et al Atezolizumab plus bevacizumab versus sorafenib in the Chinese subpopulation with unresectable hepatocellular carcinoma: phase 3 randomized, open-label IMbrave150 study. Liver Cancer. 2021;10(4):296–308. doi: 10.1159/000513486. [PMC free article] [PubMed] [CrossRef] [Google Scholar]24. REN Z, XU J, BAI Y, et al Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): a randomised, open-label, phase 2-3 study. Lancet Oncol. 2021;22(7):977–990. doi: 10.1016/s1470-2045(21)00252-7. [PubMed] [CrossRef] [Google Scholar]25. XU J, SHEN J, GU S, et al Camrelizumab in combination with apatinib in patients with advanced hepatocellular carcinoma (RESCUE): a nonrandomized, open-label, phase Ⅱ trial. Clin Cancer Res. 2021;27(4):1003–1011. doi: 10.1158/1078-0432.Ccr-20-2571. [PubMed] [CrossRef] [Google Scholar]26. Dual Immunotherapy Makes Strides against HCC. Cancer Discov. 2022;12(4):OF1. doi: 10.1158/2159-8290.Cd-nb2022-0008. [PubMed] [CrossRef] [Google Scholar]27. KUDO M Durvalumab plus tremelimumab in unresectable hepatocellular carcinoma. Hepatobiliary Surg Nutr. 2022;11(4):592–596. doi: 10.21037/hbsn-22-143. [PMC free article] [PubMed] [CrossRef] [Google Scholar]28. ZHENG Y, DOU Y, DUAN L, et al Using chemo-drugs or irradiation to break immune tolerance and facilitate immunotherapy in solid cancer. Cell Immunol. 2015;294(1):54–59. doi: 10.1016/j.cellimm.2015.02.003. [PubMed] [CrossRef] [Google Scholar]29. NOBUOKA D, MOTOMURA Y, SHIRAKAWA H, et al Radiofrequency ablation for hepatocellular carcinoma induces glypican-3 peptide-specific cytotoxic T lymphocytes. Int J Oncol. 2012;40(1):63–70. doi: 10.3892/ijo.2011.1202. [PubMed] [CrossRef] [Google Scholar]30. A trial of SHR-1210 (an Anti-PD-1 Inhibitor) in combination with FOLFOX4 in subjects with advanced HCC who have never received prior systemic treatment. (2022-02-10) [2022-12-10]. https://clinicaltrials.gov/ct2/show/study/NCT03605706.31. HARDING J J, YARMOHAMMADI H, REISS K A, et al Nivolumab (NIVO) and drug eluting bead transarterial chemoembolization (deb-TACE): preliminary results from a phase I study of patients (pts) with liver limited hepatocellular carcinoma (HCC) J Clin Oncol. 2020;38(4_suppl):525. doi: 10.1200/JCO.2020.38.4_suppl.525. [CrossRef] [Google Scholar]32. TAI W M D, LOKE K S H, GOGNA A, et al A phase Ⅱ open-label, single-center, nonrandomized trial of Y90-radioembolization in combination with nivolumab in Asian patients with advanced hepatocellular carcinoma: CA 209-678. J Clin Oncol. 2020;38(15_suppl):4590. doi: 10.1200/JCO.2020.38.15_suppl.4590. [CrossRef] [Google Scholar]33. Nivolumab in combination with TACE/TAE for patients with intermediate stage HCC (TACE-3)// ClinicalTrials. gov Identifier: NCT04268888. (2020-06-16) [2022-12-10]. https://clinicaltrials.gov/ct2/show/NCT04268888.34. Assess efficacy and safety of durvalumab alone or combined with bevacizumab in high risk of recurrence HCC patients after curative treatment (EMERALD-2). (2023-02-21) [2023-03-10]. https://clinicaltrials.gov/ct2/show/NCT03847428.35. YAN W, LIU X, MA H, et al Tim-3 fosters HCC development by enhancing TGF-β-mediated alternative activation of macrophages. Gut. 2015;64(10):1593–1604. doi: 10.1136/gutjnl-2014-307671. [PubMed] [CrossRef] [Google Scholar]36. LONG L, ZHANG X, CHEN F, et al The promising immune checkpoint LAG-3: from tumor microenvironment to cancer immunotherapy. Genes Cancer. 2018;9(5/6):176–189. doi: 10.18632/genesandcancer.180. [PMC free article] [PubMed] [CrossRef] [Google Scholar]37. A study of relatlimab in combination with nivolumab in participants with advanced liver cancer who have never been treated with immuno-oncology therapy after prior treatment with tyrosine kinase inhibitors. (2023-02-27) [2023-03-10]. https://clinicaltrials.gov/ct2/show/NCT04567615.38. TSR-022 (anti-TIM-3 antibody) and TSR-042 (anti-PD-1 antibody) in patients with liver cancer. (2022-10-25) [2022-12-10]. https://clinicaltrials.gov/ct2/show/NCT03680508.39. MAO J, WANG D, LONG J, et al Gut microbiome is associated with the clinical response to anti-PD-1 based immunotherapy in hepatobiliary cancers. J Immunother Cancer. 2021;9(12):e003334. doi: 10.1136/jitc-2021-003334. [PMC free article] [PubMed] [CrossRef] [Google Scholar]40. SANGRO B, MELERO I, WADHAWAN S, et al Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. J Hepatol. 2020;73(6):1460–1469. doi: 10.1016/j.jhep.2020.07.026. [PMC free article] [PubMed] [CrossRef] [Google Scholar]41. ESO Y, SHIMIZU T, TAKEDA H, et al Microsatellite instability and immune checkpoint inhibitors: toward precision medicine against gastrointestinal and hepatobiliary cancers. J Gastroenterol. 2020;55(1):15–26. doi: 10.1007/s00535-019-01620-7. [PMC free article] [PubMed] [CrossRef] [Google Scholar]42. MARABELLE A, FAKIH M, LOPEZ J, et al Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020;21(10):1353–1365. doi: 10.1016/s1470-2045(20)30445-9. [PubMed] [CrossRef] [Google Scholar]43. ROSENBERG S A, RESTIFO N P Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348(6230):62–68. doi: 10.1126/science.aaa4967. [PMC free article] [PubMed] [CrossRef] [Google Scholar]44. JUNE C H, O'CONNOR R S, KAWALEKAR O U, et al CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361–1365. doi: 10.1126/science.aar6711. [PubMed] [CrossRef] [Google Scholar]45. ZHOU F, SHANG W, YU X, et al Glypican-3: a promising biomarker for hepatocellular carcinoma diagnosis and treatment. Med Res Rev. 2018;38(2):741–767. doi: 10.1002/med.21455. [PubMed] [CrossRef] [Google Scholar]46. GAO H, LI K, TU H, et al Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res. 2014;20(24):6418–6428. doi: 10.1158/1078-0432.Ccr-14-1170. [PubMed] [CrossRef] [Google Scholar]47. JIANG Z, JIANG X, CHEN S, et al Anti-GPC3-CAR T cells suppress the growth of tumor cells in patient-derived xenografts of hepatocellular carcinoma. Front Immunol. 2016;7:690. doi: 10.3389/fimmu.2016.00690. [PMC free article] [PubMed] [CrossRef] [Google Scholar]48. ZHAI B, SHI D, GAO H, et al A phase Ⅰ study of anti-GPC3 chimeric antigen receptor modified T cells (GPC3 CAR-T) in Chinese patients with refractory or relapsed GPC3+ hepatocellular carcinoma (r/r GPC3+ HCC) J Clin Oncol. 2017;35(15_suppl):3049. doi: 10.1200/JCO.2017.35.15_suppl.3049. [CrossRef] [Google Scholar]49. LIU H, XU Y, XIANG J, et al Targeting alpha-fetoprotein (AFP)-MHC complex with CAR T-cell therapy for liver cancer. Clin Cancer Res. 2017;23(2):478–488. doi: 10.1158/1078-0432.Ccr-16-1203. [PubMed] [CrossRef] [Google Scholar]50. ZOU F, TAN J, LIU T, et al The CD39(+) HBV surface protein-targeted CAR-T and personalized tumor-reactive CD8(+) T cells exhibit potent anti-HCC activity. Mol Ther. 2021;29(5):1794–1807. doi: 10.1016/j.ymthe.2021.01.021. [PMC free article] [PubMed] [CrossRef] [Google Scholar]51. HUANG X, GUO J, LI T, et al c-Met-targeted chimeric antigen receptor T cells inhibit hepatocellular carcinoma cells in vitro and in vivo. J Biomed Res. 2021;36(1):10–21. doi: 10.7555/jbr.35.20200207. [PMC free article] [PubMed] [CrossRef] [Google Scholar]52. SUN B, YANG D, DAI H, et al Eradication of hepatocellular carcinoma by NKG2D-based CAR-T cells. Cancer Immunol Res. 2019;7(11):1813–1823. doi: 10.1158/2326-6066.Cir-19-0026. [PubMed] [CrossRef] [Google Scholar]53. ANDERSON L D, Jr Idecabtagene vicleucel (ide-cel) CAR T-cell therapy for relapsed and refractory multiple myeloma. Future Oncol. 2022;18(3):277–289. doi: 10.2217/fon-2021-1090. [PubMed] [CrossRef] [Google Scholar]54. RAFIQ S, HACKETT C S, BRENTJENS R J Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol. 2020;17(3):147–167. doi: 10.1038/s41571-019-0297-y. [PMC free article] [PubMed] [CrossRef] [Google Scholar]55. ZHANG G Z, LI T F, HAN S Y Mesothelin-targeted CAR-T cells for adoptive cell therapy of solid tumors. Arch Med Sci. 2021;17(5):1213–1220. doi: 10.5114/aoms.2019.84888. [PMC free article] [PubMed] [CrossRef] [Google Scholar]56. DAL BO M, De MATTIA E, BABOCI L, et al New insights into the pharmacological, immunological, and CAR-T-cell approaches in the treatment of hepatocellular carcinoma. Drug Resist Updat. 2020;51:100702. doi: 10.1016/j.drup.2020.100702. [PubMed] [CrossRef] [Google Scholar]57. KORBECKI J, KOJDER K, KAPCZUK P, et al The effect of hypoxia on the expression of CXC chemokines and CXC chemokine receptors--a review of literature. Int J Mol Sci. 2021;22(2):843. doi: 10.3390/ijms22020843. [PMC free article] [PubMed] [CrossRef] [Google Scholar]58. KANG S M, KHALIL L, El-RAYES B F, et al Rapidly evolving landscape and future horizons in hepatocellular carcinoma in the era of immuno-oncology. Front Oncol. 2022;12:821903. doi: 10.3389/fonc.2022.821903. [PMC free article] [PubMed] [CrossRef] [Google Scholar]59. JOHNSTON M P, KHAKOO S I Immunotherapy for hepatocellular carcinoma: current and future. World J Gastroenterol. 2019;25(24):2977–2989. doi: 10.3748/wjg.v25.i24.2977. [PMC free article] [PubMed] [CrossRef] [Google Scholar]60. HEO J, BREITBACH C, CHO M, et al A phase Ⅱ trial of JX-594, a targeted multimechanistic oncolytic vaccinia virus, followed by sorafenib in patients with advanced hepatocellular carcinoma (HCC) J Clin Oncol. 2012;30(15_suppl):e14566. doi: 10.1200/jco.2012.30.15_suppl.e14566. [CrossRef] [Google Scholar]61. GISH R G, GORDON S C, NELSON D, et al A randomized controlled trial of thymalfasin plus transarterial chemoembolization for unresectable hepatocellular carcinoma. Hepatol Int. 2009;3(3):480–489. doi: 10.1007/s12072-009-9132-3. [PMC free article] [PubMed] [CrossRef] [Google Scholar]62. HE C, PENG W, LI C, et al Thymalfasin, a promising adjuvant therapy in small hepatocellular carcinoma after liver resection. Medicine (Baltimore) 2017;96(16):e6606. doi: 10.1097/md.0000000000006606. [PMC free article] [PubMed] [CrossRef] [Google Scholar]63. SUN H C, TANG Z Y, WANG L, et al Postoperative interferon alpha treatment postponed recurrence and improved overall survival in patients after curative resection of HBV-related hepatocellular carcinoma: a randomized clinical trial. J Cancer Res Clin Oncol. 2006;132(7):458–465. doi: 10.1007/s00432-006-0091-y. [PubMed] [CrossRef] [Google Scholar]64. BERTELLI R, NERI F, TSIVIAN M, et al Endolymphatic immunotherapy in inoperable hepatocellular carcinoma. Transplant Proc. 2008;40(6):1913–1915. doi: 10.1016/j.transproceed.2008.05.049. [PubMed] [CrossRef] [Google Scholar]


【本文地址】


今日新闻


推荐新闻


CopyRight 2018-2019 办公设备维修网 版权所有 豫ICP备15022753号-3