Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2022, Vol. 16 Issue (3) : 467-482    https://doi.org/10.1007/s11684-021-0869-y
RESEARCH ARTICLE
Rapamycin enhances the anti-tumor activity of cabozantinib in cMet inhibitor-resistant hepatocellular carcinoma
Chao Gao1, Shenghao Wang1, Weiqing Shao1, Yu Zhang1, Lu Lu1, Huliang Jia1, Kejin Zhu2, Jinhong Chen1, Qiongzhu Dong1,3, Ming Lu1, Wenwei Zhu1(), Lunxiu Qin1,3()
1. Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
2. Kanion Research Institute, Lianyungang 222002, China
3. Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China
 Download: PDF(9161 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Cabozantinib, mainly targeting cMet and vascular endothelial growth factor receptor 2, is the second-line treatment for patients with advanced hepatocellular carcinoma (HCC). However, the lower response rate and resistance limit its enduring clinical benefit. In this study, we found that cMet-low HCC cells showed primary resistance to cMet inhibitors, and the combination of cabozantinib and mammalian target of rapamycin (mTOR) inhibitor, rapamycin, exhibited a synergistic inhibitory effect on the in vitro cell proliferation and in vivo tumor growth of these cells. Mechanically, the combination of rapamycin with cabozantinib resulted in the remarkable inhibition of AKT, extracellular signal-regulated protein kinases, mTOR, and common downstream signal molecules of receptor tyrosine kinases; decreased cyclin D1 expression; and induced cell cycle arrest. Meanwhile, rapamycin enhanced the inhibitory effects of cabozantinib on the migration and tubule formation of human umbilical vascular endothelial cells and human growth factor-induced invasion of cMet inhibitor-resistant HCC cells under hypoxia condition. These effects were further validated in xenograft models. In conclusion, our findings uncover a potential combination therapy of cabozantinib and rapamycin to combat cabozantinib-resistant HCC.

Keywords hepatocellular carcinoma      cabozantinib      primary resistance      rapamycin     
Corresponding Author(s): Wenwei Zhu,Lunxiu Qin   
Just Accepted Date: 30 August 2021   Online First Date: 15 October 2021    Issue Date: 18 July 2022
 Cite this article:   
Chao Gao,Shenghao Wang,Weiqing Shao, et al. Rapamycin enhances the anti-tumor activity of cabozantinib in cMet inhibitor-resistant hepatocellular carcinoma[J]. Front. Med., 2022, 16(3): 467-482.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-021-0869-y
https://academic.hep.com.cn/fmd/EN/Y2022/V16/I3/467
Fig.1  Low expression of cMet is correlated with poor response of HCC cells to cabozantinib. (A) The expression of cMet, pMet (Y1234/1235), cyclin D1, and cleaved-PARP levels in Huh7 cells treated with cabozantinib (CAB, 2 or 4 μmol/L) or NZ001 (NZ, 2 or 4 μmol/L) was determined by Western blot analysis. (B) Cell survival curves of a panel of HCC cell lines treated with cabozantinib (CAB) or NZ001 (NZ) for 3 days were detected by CCK8 assay, and the IC50 values of cabozantinib and NZ001 are shown. (C) Drug response data (IC50) of HCC cell lines to cabozantinib in the Genomics of Drug Sensitivity in Cancer database are shown. (D) Representative images of IHC staining of cMet in 10 HCC samples. For patient 6, the areas labeled with black boxes in the left panel are shown in the right panels with higher magnification. Bar of left panel, 500 μm. Bar of right panels, 100 μm. For patients 5 and 8, bar, 200 μm. Data are shown as the mean±SD from three independent biological replicates.
Fig.2  Rapamycin can be used in combination with cabozantinib to inhibit the proliferation of cMet inhibitor-resistant HCC cells. (A) Percentage of living Huh7 and PLC cells after treatment with cabozantinib (CAB) alone or in combination with a panel of inhibitors for 3 days. The relative cell number of the control group was set as 100%; the three most effective inhibitors are marked in red. The significance was determined by one-way ANOVA (Bonferroni post test). (B–E) The phosphorylation levels of AKT (S473) (B), ERK (T202/Y204) (C), mTOR (S2448 and S2481) (D), and p65 (S536) (E) in Huh7 cells treated with the indicated inhibitors (MK2206 (AKTi), SCH772984 (ERKi), rapamycin (RAPA) (mTORi), and SC75741 (p65i)) or in combination with cabozantinib (CAB) were determined by Western blot analysis. Representative images from three independent experiments are shown.
Fig.3  Rapamycin sensitizes cMet-low HCC cells to cabozantinib. (A, B) The proliferation of Huh7 (A) and PLC (B) cells treated with cMet inhibitor (cabozantinib (CAB) or NZ001 (NZ)) and rapamycin (RAPA) alone or in combination at the indicated concentration for 3 days was measured using CCK8 assay. The significance was determined by two-way ANOVA (Bonferroni post test). (C, D) The CI of rapamycin (RAPA) and cMet inhibitor (cabozantinib (CAB) or NZ001 (NZ)) in inhibiting the proliferation of Huh7 (C) and PLC (D) cells was calculated using the Chou–Talalay method. (E, F) Colony formation assay of Huh7 and PLC cells treated with cMet inhibitor (cabozantinib (CAB, 2 μmol/L) or NZ001 (NZ, 2 μmol/L)) and rapamycin (RAPA, 10 nmol/L) alone or in combination for 14 days. Representative images (E) and bar charts of the colony numbers (F) are shown. For (F), the significance was determined by one-way ANOVA (Bonferroni post test). Data are shown as the mean±SD from three independent biological replicates. *P<0.05; **P<0.01; ***P<0.001; ns: not significant.
Fig.4  The combination treatment of cabozantinib and rapamycin shows enhanced anti-angiogenesis effect. (A, B) In the migration assay, 2 μmol/L of cabozantinib (CAB), 2 μmol/L of NZ001 (NZ), and 10 nmol/L of rapamycin (RAPA) alone or in combination were used to treat HUVECs. The representative images of scratches after 0 and 24 h (A) and the percentage of wound healing 24 h after scratching are shown (B). Bar, 200 μm. Data are shown as the mean±SD from three independent biological replicates. (C, D) Tubule formation assay of HUVECs treated with 2 μmol/L of cabozantinib (CAB), 2 μmol/L of NZ001 (NZ), and 10 nmol/L of rapamycin (RAPA) alone or in combination for 4 h. Representative images are shown (C), and the angiogenic index was calculated (D). Bar, 200 μm. Data are shown as the mean±SD from three independent biological replicates. (E, F) CD31 expression of subcutaneous implantation tumors from C57BL/6 mice treated with indicated inhibitors was detected by IHC staining. The representative images are shown (E), and CD31+ blood vessels per mm2 were calculated accordingly (n = 6) (F). Bar, 100 μm. Data are shown as the mean±SD from six independent biological replicates. For (B), (D), and (F), the statistical significance was determined by one-way ANOVA (Bonferroni post test). *P<0.05, **P<0.01, ***P<0.001.
Fig.5  Combination treatment results in stronger inhibition of HGF-induced cell invasion of Huh7 cells under hypoxia condition. (A, C) Invasion assay of Huh7 cells treated with 1 or 10 ng/mL of HGF under normoxia or hypoxia condition for 48 h. The representative images are shown (A), and the chemotactic index was calculated as the ratio of the cell number of treatment groups to that of the control group (C). Bar, 100 μm. (B, D) The inhibitory effects of cabozantinib (CAB, 0.5 μmol/L and 1 μmol/L) alone or combined with rapamycin (RAPA, 10 nmol/L) on the invasion ability of Huh7 cells treated with 1 ng/mL of HGF under hypoxia condition were measured (B), and the chemotactic index was calculated (D). Bar, 100 μm. Data are shown as the mean±SD from three independent biological replicates. For (C) and (D), the statistical significance was determined by one-way ANOVA (Bonferroni post test). (E, F) The effects of rapamycin (RAPA) and cabozantinib (CAB) alone or in combination on tumor metastasis in lung metastasis models. The representative HE staining images of lung tissues (E) and the average numbers of lung metastatic lesions (F) from three mice per group are shown. Arrows indicate lung metastatic lesions, and the areas labeled with a black box in the upper panel are shown in the lower panel with higher magnification. Scale bar, 100 μm. For (F), data are shown as the mean±SD (n = 3), and the significance was determined by one-way ANOVA (Bonferroni post test). *P<0.05, **P<0.01, ***P<0.001.
Fig.6  Rapamycin augments the inhibitory effect of cabozantinib on the tumor growth of HCC xenograft in vivo. (A–C) Effects of rapamycin (RAPA) and cabozantinib (CAB) alone or in combination on tumor growth in subcutaneous xenograft models. Photos (A), tumor volume (B), and endpoint tumor weight (C) are shown. For (B) and (C), data are shown as mean±SD (n = 6). The significance was determined by two-way ANOVA (Bonferroni post test) (B) and one-way ANOVA (Bonferroni post test) (C). (D) The body weight of mice from different groups at day 1 (subcutaneous injection) and day 28 (mice sacrificed) was measured. Data are shown as mean±SD (n = 6). The significance was determined by one-way ANOVA (Bonferroni post test). (E–I) The expression of cyclin D1, PCNA, pmTOR (S2448), and pAKT (S473) in tumors from different experimental groups treated with the indicated inhibitors was detected by IHC staining. The representative images are shown (E). The IHC score of cyclin D1 (F), proliferation index based on the IHC score of PCNA (G), IHC score of pmTOR (S2448) (H), and IHC score of pAKT (S473) (I) are shown. Bar, 100 μm. HE, hematoxylin and eosin. For (F–I), data are shown as the mean±SD (n = 3), and the significance was determined by one-way ANOVA (Bonferroni post test). *P<0.05, **P<0.01, ***P<0.001; ns, not significant.
Fig.7  Combination treatment of cabozantinib and rapamycin leads to the suppression of RTK signaling downstream effectors and cell cycle arrest. (A, C) The inhibitory effect of cMet inhibitor (cabozantinib (CAB) or NZ001 (NZ)) and rapamycin (RAPA) alone or in combination on the phosphorylation of cMet (Y1234/1235) and downstream effectors AKT (S473), ERK (T202/Y204), mTOR (S2448), and P70S6K (T421/S424) (A) and on the expression of cyclin D1 (C) in Huh7 cells was analyzed by Western blot. (B, D) Huh7 cells treated with cMet inhibitor (cabozantinib (CAB) or NZ001 (NZ), 2 μmol/L) and rapamycin (RAPA, 10 nmol/L) alone or in combination were subjected to PI staining and assessed by flow cytometry (B). The percentage of Huh7 cells in each cell cycle stage was calculated (D). For (D), the statistical significance was determined by one-way ANOVA (Bonferroni post test). Data are shown as the mean±SD from three independent biological replicates. *P<0.05; ns, not significant.
1 F Bray, J Ferlay, I Soerjomataram, RL Siegel, LA Torre, A Jemal. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6): 394–424
https://doi.org/10.3322/caac.21492 pmid: 30207593
2 A Villanueva. Hepatocellular carcinoma. N Engl J Med 2019; 380(15): 1450–1462
https://doi.org/10.1056/NEJMra1713263 pmid: 30970190
3 JD Yang, P Hainaut, GJ Gores, A Amadou, A Plymoth, LR Roberts. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol 2019; 16(10): 589–604
https://doi.org/10.1038/s41575-019-0186-y pmid: 31439937
4 CR Maroun, T Rowlands. The Met receptor tyrosine kinase: a key player in oncogenesis and drug resistance. Pharmacol Ther 2014; 142(3): 316–338
https://doi.org/10.1016/j.pharmthera.2013.12.014 pmid: 24384534
5 FM Yakes, J Chen, J Tan, K Yamaguchi, Y Shi, P Yu, F Qian, F Chu, F Bentzien, B Cancilla, J Orf, A You, AD Laird, S Engst, L Lee, J Lesch, YC Chou, AH Joly. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol Cancer Ther 2011; 10(12): 2298–2308
https://doi.org/10.1158/1535-7163.MCT-11-0264 pmid: 21926191
6 GK Abou-Alfa, T Meyer, AL Cheng, AB El-Khoueiry, L Rimassa, BY Ryoo, I Cicin, P Merle, Y Chen, JW Park, JF Blanc, L Bolondi, HJ Klümpen, SL Chan, V Zagonel, T Pressiani, MH Ryu, AP Venook, C Hessel, AE Borgman-Hagey, G Schwab, RK Kelley. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med 2018; 379(1): 54–63
https://doi.org/10.1056/NEJMoa1717002 pmid: 29972759
7 JM Llovet, R Montal, D Sia, RS Finn. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol 2018; 15(10): 599–616
https://doi.org/10.1038/s41571-018-0073-4 pmid: 30061739
8 S Faivre, G Kroemer, E Raymond. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov 2006; 5(8): 671–688
https://doi.org/10.1038/nrd2062 pmid: 16883305
9 F Chiarini, C Evangelisti, JA McCubrey, AM Martelli. Current treatment strategies for inhibiting mTOR in cancer. Trends Pharmacol Sci 2015; 36(2): 124–135
https://doi.org/10.1016/j.tips.2014.11.004 pmid: 25497227
10 A Carracedo, L Ma, J Teruya-Feldstein, F Rojo, L Salmena, A Alimonti, A Egia, AT Sasaki, G Thomas, SC Kozma, A Papa, C Nardella, LC Cantley, J Baselga, PP Pandolfi. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest 2008; 118(9): 3065–3074
https://doi.org/10.1172/JCI34739 pmid: 18725988
11 RA Saxton, DM Sabatini. mTOR signaling in growth, metabolism, and disease. Cell 2017; 168(6): 960–976
https://doi.org/10.1016/j.cell.2017.02.004 pmid: 28283069
12 Y Zhang, X Gao, Y Zhu, D Kadel, H Sun, J Chen, Q Luo, H Sun, L Yang, J Yang, Y Sheng, Y Zheng, K Zhu, Q Dong, L Qin. The dual blockade of MET and VEGFR2 signaling demonstrates pronounced inhibition on tumor growth and metastasis of hepatocellular carcinoma. J Exp Clin Cancer Res 2018; 37(1): 93
https://doi.org/10.1186/s13046-018-0750-2 pmid: 29712569
13 TC Chou. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 2010; 70(2): 440–446
https://doi.org/10.1158/0008-5472.CAN-09-1947 pmid: 20068163
14 E Gherardi, W Birchmeier, C Birchmeier, G Vande Woude. Targeting MET in cancer: rationale and progress. Nat Rev Cancer 2012; 12(2): 89–103
https://doi.org/10.1038/nrc3205 pmid: 22270953
15 A D’Errico, M Fiorentino, A Ponzetto, Y Daikuhara, H Tsubouchi, C Brechot, JY Scoazec, WF Grigioni. Liver hepatocyte growth factor does not always correlate with hepatocellular proliferation in human liver lesions: its specific receptor c-met does. Hepatology 1996; 24(1): 60–64
https://doi.org/10.1053/jhep.1996.v24.pm0008707284 pmid: 8707284
16 P Kaposi-Novak, JS Lee, L Gòmez-Quiroz, C Coulouarn, VM Factor, SS Thorgeirsson. Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype. J Clin Invest 2006; 116(6): 1582–1595
https://doi.org/10.1172/JCI27236 pmid: 16710476
17 L Rimassa, E Assenat, M Peck-Radosavljevic, V Zagonel, M Pracht, ER Caremoli, P Mathurin, WP Harris, L Bolondi, M Reig, N Damjanov, B Daniele, C Porta, V Mazzaferro, G Abbadessa, BE Schwartz, M Lamar, TR Goldberg, A Santoro, J Bruix. Second-line tivantinib (ARQ 197) vs placebo in patients (Pts) with MET-high hepatocellular carcinoma (HCC): Results of the METIV-HCC phase III trial. J Clin Oncol 2017; 35(15 suppl): 4000
https://doi.org/10.1200/JCO.2017.35.15_suppl.4000
18 D Hanahan, RA Weinberg. Hallmarks of cancer: the next generation. Cell 2011; 144(5): 646–674
https://doi.org/10.1016/j.cell.2011.02.013 pmid: 21376230
19 I Dagogo-Jack, AT Shaw. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol 2018; 15(2): 81–94
https://doi.org/10.1038/nrclinonc.2017.166 pmid: 29115304
20 X Li, MT Lewis, J Huang, C Gutierrez, CK Osborne, MF Wu, SG Hilsenbeck, A Pavlick, X Zhang, GC Chamness, H Wong, J Rosen, JC Chang. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 2008; 100(9): 672–679
https://doi.org/10.1093/jnci/djn123 pmid: 18445819
21 J Bruix, LG da Fonseca, M Reig. Insights into the success and failure of systemic therapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2019; 16(10): 617–630
https://doi.org/10.1038/s41575-019-0179-x pmid: 31371809
22 Z Ezzoukhry, C Louandre, E Trécherel, C Godin, B Chauffert, S Dupont, M Diouf, JC Barbare, JC Mazière, A Galmiche. EGFR activation is a potential determinant of primary resistance of hepatocellular carcinoma cells to sorafenib. Int J Cancer 2012; 131(12): 2961–2969
https://doi.org/10.1002/ijc.27604 pmid: 22514082
23 DY Chiang, A Villanueva, Y Hoshida, J Peix, P Newell, B Minguez, AC LeBlanc, DJ Donovan, SN Thung, M Solé, V Tovar, C Alsinet, AH Ramos, J Barretina, S Roayaie, M Schwartz, S Waxman, J Bruix, V Mazzaferro, AH Ligon, V Najfeld, SL Friedman, WR Sellers, M Meyerson, JM Llovet. Focal gains of VEGFA and molecular classification of hepatocellular carcinoma. Cancer Res 2008; 68(16): 6779–6788
https://doi.org/10.1158/0008-5472.CAN-08-0742 pmid: 18701503
24 Y Hoshida, SM Nijman, M Kobayashi, JA Chan, JP Brunet, DY Chiang, A Villanueva, P Newell, K Ikeda, M Hashimoto, G Watanabe, S Gabriel, SL Friedman, H Kumada, JM Llovet, TR Golub. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res 2009; 69(18): 7385–7392
https://doi.org/10.1158/0008-5472.CAN-09-1089 pmid: 19723656
25 S Boyault, DS Rickman, A de Reyniès, C Balabaud, S Rebouissou, E Jeannot, A Hérault, J Saric, J Belghiti, D Franco, P Bioulac-Sage, P Laurent-Puig, J Zucman-Rossi. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 2007; 45(1): 42–52
https://doi.org/10.1002/hep.21467 pmid: 17187432
26 M Laplante, DM Sabatini. mTOR signaling in growth control and disease. Cell 2012; 149(2): 274–293
https://doi.org/10.1016/j.cell.2012.03.017 pmid: 22500797
27 L Asnaghi, P Bruno, M Priulla, A Nicolin. mTOR: a protein kinase switching between life and death. Pharmacol Res 2004; 50(6): 545–549
https://doi.org/10.1016/j.phrs.2004.03.007 pmid: 15501691
28 A Villanueva, DY Chiang, P Newell, J Peix, S Thung, C Alsinet, V Tovar, S Roayaie, B Minguez, M Sole, C Battiston, S Van Laarhoven, MI Fiel, A Di Feo, Y Hoshida, S Yea, S Toffanin, A Ramos, JA Martignetti, V Mazzaferro, J Bruix, S Waxman, M Schwartz, M Meyerson, SL Friedman, JM Llovet. Pivotal role of mTOR signaling in hepatocellular carcinoma. Gastroenterology 2008; 135(6): 1972–1983.e1–11
https://doi.org/10.1053/j.gastro.2008.08.008 pmid: 18929564
29 Y Imura, H Yasui, H Outani, T Wakamatsu, K Hamada, T Nakai, S Yamada, A Myoui, N Araki, T Ueda, K Itoh, H Yoshikawa, N Naka. Combined targeting of mTOR and c-MET signaling pathways for effective management of epithelioid sarcoma. Mol Cancer 2014; 13(1): 185
https://doi.org/10.1186/1476-4598-13-185 pmid: 25098767
30 C Wigerup, S Påhlman, D Bexell. Therapeutic targeting of hypoxia and hypoxia-inducible factors in cancer. Pharmacol Ther 2016; 164: 152–169
https://doi.org/10.1016/j.pharmthera.2016.04.009 pmid: 27139518
31 CW Pugh, PJ Ratcliffe. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 2003; 9(6): 677–684
https://doi.org/10.1038/nm0603-677 pmid: 12778166
32 GL Semenza. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci 2012; 33(4): 207–214
https://doi.org/10.1016/j.tips.2012.01.005 pmid: 22398146
33 H You, W Ding, H Dang, Y Jiang, CB Rountree. c-Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinoma. Hepatology 2011; 54(3): 879–889
https://doi.org/10.1002/hep.24450 pmid: 21618573
34 V Finisguerra, G Di Conza, M Di Matteo, J Serneels, S Costa, AA Thompson, E Wauters, S Walmsley, H Prenen, Z Granot, A Casazza, M Mazzone. MET is required for the recruitment of anti-tumoural neutrophils. Nature 2015; 522(7556): 349–353
https://doi.org/10.1038/nature14407 pmid: 25985180
35 C Zhu, Y Wei, X Wei. AXL receptor tyrosine kinase as a promising anti-cancer approach: functions, molecular mechanisms and clinical applications. Mol Cancer 2019; 18(1): 153
https://doi.org/10.1186/s12943-019-1090-3 pmid: 31684958
36 AY Li, MG McCusker, A Russo, KA Scilla, A Gittens, K Arensmeyer, R Mehra, V Adamo, C Rolfo. RET fusions in solid tumors. Cancer Treat Rev 2019; 81: 101911
https://doi.org/10.1016/j.ctrv.2019.101911 pmid: 31715421
37 F Janku, TA Yap, F Meric-Bernstam. Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol 2018; 15(5): 273–291
https://doi.org/10.1038/nrclinonc.2018.28 pmid: 29508857
[1] FMD-21023-OF-QLX_suppl_1 Download
[1] Jinrong Liu, Rongfang Shen, Lin Feng, Shujun Cheng, Jun Chen, Ting Xiao, Shunying Zhao. Proteomics study of Mycoplasma pneumoniae pneumonia reveals the Fc fragment of the IgG-binding protein as a serum biomarker and implicates potential therapeutic targets[J]. Front. Med., 2022, 16(3): 378-388.
[2] Wei Zhang, Bixiang Zhang, Xiao-ping Chen. Adjuvant treatment strategy after curative resection for hepatocellular carcinoma[J]. Front. Med., 2021, 15(2): 155-169.
[3] Jianpeng Liu, Xinhua Chen, Shusen Zheng. Immune response triggered by the ablation of hepatocellular carcinoma with nanosecond pulsed electric field[J]. Front. Med., 2021, 15(2): 170-177.
[4] Amy Lee, Fa-Chyi Lee. Medical oncology management of advanced hepatocellular carcinoma 2019: a reality check[J]. Front. Med., 2020, 14(3): 273-283.
[5] Renyu Zhang, Zhao Zhang, Zekun Liu, Ding Wei, Xiaodong Wu, Huijie Bian, Zhinan Chen. Adoptive cell transfer therapy for hepatocellular carcinoma[J]. Front. Med., 2019, 13(1): 3-11.
[6] Shasha Zhu, Huimin Zhang, Li Bai. NKT cells in liver diseases[J]. Front. Med., 2018, 12(3): 249-261.
[7] Guangbiao Zhou, Xinchun Zhao. Carcinogens that induce the A:T>T:A nucleotide substitutions in the genome[J]. Front. Med., 2018, 12(2): 236-238.
[8] Min Yu, Zonghai Li. Natural killer cells in hepatocellular carcinoma: current status and perspectives for future immunotherapeutic approaches[J]. Front. Med., 2017, 11(4): 509-521.
[9] Zhen He,Cheng Hu,Weiping Jia. miRNAs in non-alcoholic fatty liver disease[J]. Front. Med., 2016, 10(4): 389-396.
[10] Xinsen Xu,Yanyan Zhou,Runchen Miao,Wei Chen,Kai Qu,Qing Pang,Chang Liu. Transcriptional modules related to hepatocellular carcinoma survival: coexpression network analysis[J]. Front. Med., 2016, 10(2): 183-190.
[11] Zhi Xu,Chunxiang Cao,Haiyan Xia,Shujing Shi,Lingzhi Hong,Xiaowei Wei,Dongying Gu,Jianmin Bian,Zijun Liu,Wenbin Huang,Yixin Zhang,Song He,Nikki Pui-Yue Lee,Jinfei Chen. Protein phosphatase magnesium-dependent 1δ is a novel tumor marker and target in hepatocellular carcinoma[J]. Front. Med., 2016, 10(1): 52-60.
[12] Felice Ho-Ching Tsang,Sandy Leung-Kuen Au,Lai Wei,Dorothy Ngo-Yin Fan,Joyce Man-Fong Lee,Carmen Chak-Lui Wong,Irene Oi-Lin Ng,Chun-Ming Wong. MicroRNA-142-3p and microRNA-142-5p are downregulated in hepatocellular carcinoma and exhibit synergistic effects on cell motility[J]. Front. Med., 2015, 9(3): 331-343.
[13] Farhad Sahebjam,John M. Vierling. Autoimmune hepatitis[J]. Front. Med., 2015, 9(2): 187-219.
[14] Guanghua Rong,Wenlin Bai,Zheng Dong,Chunping Wang,Yinying Lu,Zhen Zeng,Jianhui Qu,Min Lou,Hong Wang,Xudong Gao,Xiujuan Chang,Linjing An,Yan Chen,Yongping Yang. Cryotherapy for cirrhosis-based hepatocellular carcinoma: a single center experience from 1595 treated cases[J]. Front. Med., 2015, 9(1): 63-71.
[15] Kai Qu,Ting Lin,Zhixin Wang,Sinan Liu,Hulin Chang,Xinsen Xu,Fandi Meng,Lei Zhou,Jichao Wei,Minghui Tai,Yafeng Dong,Chang Liu. Reactive oxygen species generation is essential for cisplatin-induced accelerated senescence in hepatocellular carcinoma[J]. Front. Med., 2014, 8(2): 227-235.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed