切换至 "中华医学电子期刊资源库"

中华移植杂志(电子版) ›› 2021, Vol. 15 ›› Issue (06) : 334 -340. doi: 10.3877/cma.j.issn.1674-3903.2021.06.003

论著

使用程序性死亡受体-1单克隆抗体治疗肝癌肝移植术后复发的临床策略
鲁旭1, 张英才1, 李华1, 赵辉1, 李海波1, 张剑文1, 邱春辉1, 易述红1, 汪根树1, 张剑1, 杨扬1, 陈规划1, 汪国营1,()   
  1. 1. 510630 广州,中山大学附属第三医院肝脏外科暨肝脏移植中心
  • 收稿日期:2021-04-06 出版日期:2021-12-25
  • 通信作者: 汪国营
  • 基金资助:
    十三五科技重大专项(2017ZX10203205-006-001); 陈孝平科技发展基金(CXPJJH12000001-2020218)

Clinical strategy of programmed death-1 monoclonal antibody in patients with recurrent liver cancer after liver transplantation

Xu Lu1, Yingcai Zhang1, Hua Li1, Hui Zhao1, Haibo Li1, Jianwen Zhang1, Chunhui Qiu1, Shuhong Yi1, Genshu Wang1, Jian Zhang1, Yang Yang1, Guihua Chen1, Guoying Wang1,()   

  1. 1. Department of Hepatic Surgery, Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
  • Received:2021-04-06 Published:2021-12-25
  • Corresponding author: Guoying Wang
引用本文:

鲁旭, 张英才, 李华, 赵辉, 李海波, 张剑文, 邱春辉, 易述红, 汪根树, 张剑, 杨扬, 陈规划, 汪国营. 使用程序性死亡受体-1单克隆抗体治疗肝癌肝移植术后复发的临床策略[J]. 中华移植杂志(电子版), 2021, 15(06): 334-340.

Xu Lu, Yingcai Zhang, Hua Li, Hui Zhao, Haibo Li, Jianwen Zhang, Chunhui Qiu, Shuhong Yi, Genshu Wang, Jian Zhang, Yang Yang, Guihua Chen, Guoying Wang. Clinical strategy of programmed death-1 monoclonal antibody in patients with recurrent liver cancer after liver transplantation[J]. Chinese Journal of Transplantation(Electronic Edition), 2021, 15(06): 334-340.

目的

探讨肝癌肝移植术后复发患者安全应用程序性死亡受体-1(PD-1)单克隆抗体(以下简称单抗)的治疗策略。

方法

本研究纳入4例一线靶向药物治疗后肿瘤仍进展的肝癌肝移植术后复发患者,进行移植肝穿刺免疫组织化学检测程序性死亡配体-1(PD-L1)表达均为阴性。患者接受PD-1抑制剂卡瑞利珠单抗治疗,每3周1次,200 mg/次,并选择合适的靶向药物联合治疗,同时将免疫抑制方案调整为哺乳动物雷帕霉素靶蛋白(mTOR)抑制剂联合霉酚酸类药物。随访期间综合分析患者肝功能、甲胎蛋白(AFP)以及胸腹部影像学检查等变化情况,评估PD-1单抗的有效性与安全性。

结果

截至2021年2月,4例患者中存活2例,分别随访17.3和15.3个月,期间均接受PD-1单抗治疗6次,有乏力、腹泻和高血压等不良反应,经对症治疗后均好转,仍处于持续随访中。病例1因肺部转移瘤压迫气管和支气管引起严重肺部感染,于PD-1单抗治疗后6.3个月死亡(期间共接受PD-1单抗治疗6次);病例4因肺部转移瘤压迫支气管引起肺炎、肺不张和双侧胸水,于PD-1单抗治疗后10.9个月死亡(期间共接受PD-1单抗治疗3次)。4例患者使用PD-1单抗治疗后,AST水平均处于相对稳定状态,偶有波动也可通过护肝治疗较快恢复正常;血清白蛋白波动于30~45 g/L;总胆红素处于正常水平,均未发生黄疸;3例患者AFP均有不同程度下降。随访期间患者均未发生明显排斥反应。使用PD-1单抗治疗后,改良的实体肿瘤疗效评价标准评估3例患者为疾病稳定,中位无进展生存期为141 d;另1例患者评估为部分缓解,中位无进展生存期为180 d。

结论

移植肝组织PD-L1表达阴性的肝癌肝移植术后复发患者,在mTOR抑制剂联合霉酚酸类药物维持有效免疫抑制状态下使用PD-1单抗是相对安全的,但其有效性与长期临床获益仍需进一步观察。

Objective

To investigate the safe application of programmed death-1 (PD-1) monoclonal antibody in patients with recurrent liver cancer after liver transplantation.

Methods

Four patients with recurrent hepatocellular carcinoma after liver transplantation who still progressed after first-line targeted drug therapy were included in this study, and programmed death ligand-1 (PD-L1) expression was negative by immunohistochemistry of liver allograft puncture. Patients were treated with the PD-1 inhibitor Camrelizumab, 200 mg every 3 weeks, and the appropriate targeted drug combination was selected, while the immunosuppressive regimen was adjusted to mammalian target of rapamycin (mTOR) inhibitors in combination with mycophenolate mofetil. Blood routine, liver and kidney function, alpha-fetoprotein (AFP) and thoracic and abdominal imaging examinations were measured during follow-up, and changes in liver function and tumor parameters during follow-up were comprehensively analyzed to evaluate the efficacy and safety of PD-1 monoclonal antibody.

Results

By February 2021, 2 of the 4 patients survived and were followed up for 17.3 and 15.3 months, respectively, during which they all received PD-1 monoclonal antibody for 6 times, with adverse reactions such as fatigue, diarrhea and hypertension, which were improved after symptomatic treatment and are still under continuous follow-up. In Case 1, the patient died 6.3 months after PD-1 monoclonal antibody treatment due to severe lung infection caused by compression of trachea and bronchus by pulmonary metastases (a total of 6 times of PD-1 monoclonal antibody treatment were performed during this period); in Case 4, the patient died 10.9 months after PD-1 monoclonal antibody treatment due to pneumonia, atelectasis and pleural effusion caused by compression of bronchus by pulmonary metastases (a total of 3 times of PD-1 monoclonal antibody treatment were performed during this period). Four patients were treated with PD-1 monoclonal antibody. However, AST levels were in a relatively stable state, and occasional fluctuations can also be quickly restored to normal by liver protection therapy; serum albumin fluctuated between 30 and 45 g/L; total bilirubin was in a normal level, and no jaundice occurred; AFP decreased to varying degrees in 3 patients. None of the patients developed significant rejection during the follow-up period. After treatment with PD-1 monoclonal antibody, 3 patients had stable disease assessed by the modified Response Evaluation Criteria in Solid Tumors, with a progression-free survival of 141 days; the other patient had a partial response assessed, with a progression-free survival of 180 days.

Conclusions

PD-1 monoclonal antibody is relatively safe in patients with PD-L1-negative recurrent hepatocellular carcinoma after liver transplantation under the effective immunosuppressive state of mTOR inhibitor combined with mycophenolate mofetil, but its effectiveness and long-term clinical benefit still need further observation.

表1 肝癌肝移植术后复发接受PD-1单抗治疗的患者资料
图1 肝癌肝移植术后复发患者接受PD-1单抗治疗期间肝功能和APF水平变化注:PD-1.程序性死亡受体-1; ALB.血清白蛋白;TBIL.总胆红素;AFP.甲胎蛋白
图2 肝癌肝移植术后复发病例1接受PD-1单抗治疗前后肿瘤影像学变化注:PD-1.程序性死亡受体-1; a、b、c分别为入组前肝、肺、纵隔CT影像;d、e、f分别为入组126 d复查肝、肺、纵隔CT影像,可见肝、肺结节均较前增大
图3 肝癌肝移植术后复发病例2接受PD-1单抗治疗前后肿瘤影像学变化注:PD-1.程序性死亡受体-1; a、b、c分别为入组前腹部、纵隔、肺CT影像;d、e、f分别为入组181 d复查腹部、纵隔、肺CT影像,可见双肺新发结节
图4 肝癌肝移植术后复发病例3接受PD-1单抗治疗前后肿瘤影像学变化注:PD-1.程序性死亡受体-1; a、b、c分别为入组前肝、肺CT影像;d、e、f分别为入组180 d复查肝、肺CT影像,可见肝内新发子灶
图5 肝癌肝移植术后复发病例4接受PD-1单抗治疗前后肿瘤影像学变化注:PD-1.程序性死亡受体-1; a、b、c分别为入组前双下肢、肺、纵隔CT影像;d、e、f分别为入组117 d复查双下肢、肺、纵隔影像,可见纵隔淋巴结转移瘤较前增大
1
中国医师协会器官移植医师分会,中华医学会器官移植学分会. 中国肝癌肝移植临床实践指南(2018版)[J/CD]. 中华移植杂志:电子版201812 (4): 145-150.
2
王波,钱叶本,罗雪莲,等. 肝癌复发的诊断与治疗进展[J]. 肝胆外科杂志2016, 24(4): 307-312.
3
Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer[J]. N Engl J Med, 2012, 366(26): 2443-2454.
4
Donisi C, Puzzoni M, Ziranu P, et al. Immune checkpoint inhibitors in the treatment of HCC[J]. Front Oncol, 2020, 10: 601240.
5
Aitcheson G, Pillai A, Dahman B, et al. Recent advances in systemic therapies for advanced hepatocellular carcinoma[J]. Curr Hepatol Rep, 2021, 20(1): 23-33.
6
Llovet JM, Lencioni R. mRECIST for HCC: performance and novel refinements[J]. J Hepatol, 2020, 72(2): 288-306.
7
刘秀峰,秦叔逵. 肝细胞癌免疫治疗的现状与未来[J]. 医药导报2019, 38(8): 1008-1013.
8
Hall KH, Liu Y, Jiang C, et al. New and worsening long-term immune-related adverse events with PD-1/PD-L1 pathway agents in patients with cancer[J]. Pharmacotherapy, 2020, 40(2): 133-141.
9
Qin S, Ren Z, Meng Z, et al. Camrelizumab in patients with previously treated advanced hepatocellular carcinoma: a multicentre, open-label, parallel-group, randomised, phase 2 trial[J]. Lancet Oncol, 2020, 21(4): 571-580.
10
Biondani P, De Martin E, Samuel D. Safety of an anti-PD-1 immune checkpoint inhibitor in a liver transplant recipient[J]. Ann Oncol, 2018, 29(1): 286-287.
11
De Toni EN, Gerbes AL. Tapering of immunosuppression and sustained treatment with nivolumab in a liver transplant recipient[J]. Gastroenterology, 2017, 152(6): 1631-1633.
12
Qiu J, Tang W, Du C. Immune checkpoint inhibitors in patients with recurrent hepatocellular carcinoma after liver transplantation: a case report and literature review[J]. Curr Cancer Drug Targets, 2020, 20(9): 720-727.
13
Owoyemi I, Vaughan LE, Costello CM, et al. Clinical outcomes of solid organ transplant recipients with metastatic cancers who are treated with immune checkpoint inhibitors: a single-center analysis[J]. Cancer, 2020, 126(21): 4780-4787.
14
Fisher J, Zeitouni N, Fan W, et al. Immune checkpoint inhibitor therapy in solid organ transplant recipients: a patient-centered systematic review[J]. J Am Acad Dermatol, 2020, 82(6): 1490-1500.
15
Khalaileh A, Khoury T, Harkrosh S, et al. Multiplication product of Model for End-stage Liver Disease and Donor Risk Index as predictive models of survival after liver transplantation[J]. Eur J Gastroenterol Hepatol, 2019, 31(9): 1116-1120.
16
De Bruyn P, Van Gestel D, Ost P, et al. Immune checkpoint blockade for organ transplant patients with advanced cancer: how far can we go?[J]. Curr Opin Oncol, 2019, 31(2): 54-64.
17
DeLeon TT, Salomao MA, Aqel BA, et al. Pilot evaluation of PD-1 inhibition in metastatic cancer patients with a history of liver transplantation: the Mayo Clinic experience[J]. J Gastrointest Oncol, 2018, 9(6): 1054-1062.
18
汪国营,唐晖,张英才,等. 程序性死亡受体(PD)-1单克隆抗体治疗肝癌肝移植术后复发诱发急性免疫性肝炎:附1例报告[J]. 器官移植2016, 7(1): 44-47.
19
汪国营. 肝癌肝移植新进展——2019年ILTS年会速递[J]. 器官移植2020, 11(1): 47-53.
20
Rodríguez-Perálvarez M, Guerrero M, Barrera L, et al. Impact of early initiated everolimus on the recurrence of hepatocellular carcinoma after liver transplantation[J]. Transplantation, 2018, 102(12): 2056-2064.
21
Grigg SE, Sarri GL, Gow PJ, et al. Systematic review with meta-analysis: sirolimus- or everolimus-based immunosuppression following liver transplantation for hepatocellular carcinoma[J]. Aliment Pharmacol Ther, 2019, 49(10): 1260-1273.
22
郑树森. 肝癌患者肝移植后应用mTOR抑制剂预防肿瘤复发的研究进展[J]. 中华器官移植杂志2011, 32(8): 453-456.
23
Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy[J]. Nat Rev Cancer, 2012, 12(4): 252-264.
24
Boussiotis VA. Molecular and biochemical aspects of the PD-1 checkpoint pathway[J]. N Engl J Med, 2016, 375(18): 1767-1778.
25
Ozkaynak E, Wang L, Goodearl A, et al. Programmed death-1 targeting can promote allograft survival[J]. J Immunol, 2002, 169(11): 6546-6553.
26
Cao Y, Zhou H, Tao J, et al. Keratinocytes induce local tolerance to skin graft by activating interleukin-10-secreting T cells in the context of costimulation molecule B7-H1[J]. Tranplantation, 2003, 75(8): 1390-1396.
27
Lee BT, Horwich BH, Chopra S, et al. Checkpoint inhibitor-induced rejection of a liver allograft: A combination of acute T cell-mediated and antibody-mediated rejection[J]. Liver Transpl, 2019, 25(12): 1845-1848.
28
易善永,赵玲,易强,等. 肿瘤突变负荷在非小细胞肺癌免疫靶向治疗疗效预测中的应用[J]. 中国临床研究2019, 32(4): 566-569.
[1] 李淼, 朱连华, 韩鹏, 姜波, 费翔. 高帧频超声造影评价肝细胞癌血管形态与风险因素的研究[J]. 中华医学超声杂志(电子版), 2023, 20(09): 911-915.
[2] 丁建民, 秦正义, 张翔, 周燕, 周洪雨, 王彦冬, 经翔. 超声造影与普美显磁共振成像对具有高危因素的≤3 cm肝结节进行LI-RADS分类诊断的前瞻性研究[J]. 中华医学超声杂志(电子版), 2023, 20(09): 930-938.
[3] 董杰, 杨松, 杨浩, 陈翔, 张万里. 乙酰辅酶A羧化酶2基因高甲基化与肝细胞癌临床病理因素和生存期的关系[J]. 中华普通外科学文献(电子版), 2023, 17(06): 433-437.
[4] 黄威, 刘启, 陈流华, 滕茶香, 区喆建, 刘韩笑, 陈健聪, 张昆松. 新定义的可预测肝癌预后的焦亡相关lncRNA模型[J]. 中华普通外科学文献(电子版), 2023, 17(05): 357-365.
[5] 李坤河, 寇萌佳, 邝立挺. 肝移植术后二次气管插管的危险因素及预测模型的建立[J]. 中华普通外科学文献(电子版), 2023, 17(05): 366-371.
[6] 中华医学会器官移植学分会, 中国医师协会器官移植医师分会, 上海医药行业协会. 中国肝、肾移植受者霉酚酸类药物应用专家共识(2023版)[J]. 中华移植杂志(电子版), 2023, 17(05): 257-272.
[7] 陆闻青, 陈昕怡, 任雪飞. 遗传代谢病儿童肝移植受者术后生活质量调查研究[J]. 中华移植杂志(电子版), 2023, 17(05): 287-292.
[8] 范铁艳, 李君, 陈虹. 肝移植术后新发戊型病毒性肝炎的诊治经验[J]. 中华移植杂志(电子版), 2023, 17(05): 293-296.
[9] 陈朔, 陈峰, 程飞, 项捷. 糖原累积病Ⅰ型并发胰腺炎肝移植术后胰腺梗死一例[J]. 中华移植杂志(电子版), 2023, 17(05): 300-302.
[10] 严庆, 刘颖, 邓斐文, 陈焕伟. 微血管侵犯对肝癌肝移植患者生存预后的影响[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 624-629.
[11] 廖梅, 张红君, 金洁玚, 吕艳, 任杰. 床旁超声造影对肝移植术后早期肝动脉血栓的诊断价值[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 630-634.
[12] 李秉林, 吕少诚, 潘飞, 姜涛, 樊华, 寇建涛, 贺强, 郎韧. 供肝灌注液病原菌与肝移植术后早期感染的相关性分析[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 656-660.
[13] 吕垒, 冯啸, 何凯明, 曾凯宁, 杨卿, 吕海金, 易慧敏, 易述红, 杨扬, 傅斌生. 改良金氏评分在儿童肝豆状核变性急性肝衰竭肝移植手术时机评估中价值并文献复习[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 661-668.
[14] 胡宝茹, 尚乃舰, 高迪. 中晚期肝细胞癌的DCE-MRI及DWI表现与免疫治疗预后的相关性分析[J]. 中华消化病与影像杂志(电子版), 2023, 13(06): 399-403.
[15] 吴凤芸, 滕鑫, 刘连娟. 高帧频超声造影与增强磁共振对不同直径原发性高分化肝细胞癌的诊断价值[J]. 中华消化病与影像杂志(电子版), 2023, 13(06): 404-408.
阅读次数
全文


摘要