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.    2024, Vol. 18 Issue (4) : 708-720    https://doi.org/10.1007/s11684-024-1071-9
Moderate expression of CD39 in GPC3-CAR-T cells shows high efficacy against hepatocellular carcinoma
Fan Zou1,5, Jialiang Wei3, Jialang Zhuang6,7, Yafang Liu2, Jizhou Tan4(), Xianzhang Huang2(), Ting Liu2()
1. Guangdong Cardiovsacular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Science, Guangzhou 510080, China
2. The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Department of Laboratory Medicine/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
3. Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
4. Department of Stomatology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
5. Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Science), Southern Medical University, Guangzhou 510080, China
6. School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China
7. Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518055, China
 Download: PDF(13117 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

CD39 serves as a crucial biomarker for neoantigen-specific CD8+ T cells and is associated with antitumor activity and exhaustion. However, the relationship between CD39 expression levels and the function of chimeric antigen receptor T (CAR-T) cells remains controversial. This study aimed to investigate the role of CD39 in the functional performance of CAR-T cells against hepatocellular carcinoma (HCC) and explore the therapeutic potential of CD39 modulators, such as mitochondrial division inhibitor-1 (mdivi-1), or knockdown CD39 through short hairpin RNA. Our findings demonstrated that glypican-3-CAR-T cells with moderate CD39 expression exhibited a strong antitumor activity, while high and low levels of CD39 led to an impaired cellular function. Methods modulating the proportion of CD39 intermediate (CD39int)-phenotype CAR-T cells such as mdivi-1 and CD39 knockdown enhanced and impaired T cell function, respectively. The combination of mdivi-1 and CD39 knockdown in CAR-T cells yielded the highest proportion of infiltrated CD39int CAR-T cells and demonstrated a robust antitumor activity in vivo. In conclusion, this study revealed the crucial role of CD39 in CAR-T cell function, demonstrated the potential therapeutic efficacy of combining mdivi-1 with CD39 knockdown in HCC, and provided a novel treatment strategy for HCC patients in the field of cellular immunotherapy.

Keywords CD39      CAR-T cells      mdivi-1      hepatocellular carcinoma      antitumor activity     
Corresponding Author(s): Jizhou Tan,Xianzhang Huang,Ting Liu   
Just Accepted Date: 10 May 2024   Online First Date: 05 June 2024    Issue Date: 30 August 2024
 Cite this article:   
Fan Zou,Jialiang Wei,Jialang Zhuang, et al. Moderate expression of CD39 in GPC3-CAR-T cells shows high efficacy against hepatocellular carcinoma[J]. Front. Med., 2024, 18(4): 708-720.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-024-1071-9
https://academic.hep.com.cn/fmd/EN/Y2024/V18/I4/708
Fig.1  CD39int CD8+ CAR-T cells presented an antitumor activity in vivo. (A) In vivo experimental design. GPC3+ Huh7 cell lines at 1 × 106 cells were inoculated s.c. into NSG mice. Fourteen days later, GPC3-CAR-T cells were infused via tail injection, n = 6. On day 14, tumors were resected, followed by digestion and FACS analysis. (B,C) Flow cytometry for analyzing the CD39hi/int/- subsets gated from tumor-infiltrating GPC3-CAR-T cells. The frequency of IFN-γ+ from each subset is revealed. Statistical data is also shown. All data are the mean of three independent experiments performed in duplicate. Mean ± SEM, unpaired t-test, *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.2  CD39 knockdown compromised cytotoxicity and proliferation but reduced apoptosis in CAR-T cells in vitro. (A) Schematic of the lentiviral vectors carrying a GPC3-specific CAR moiety and a cluster of sh-CD39. (B) In vitro experimental design. GPC3-CAR-T and shCD39-GPC3-CAR-T cells were separately cocultured and stimulated by targeted antigen rhGPC3 protein. T cells were harvested and tested by flow cytometry to analyze the CAR-T function. (C) Bar plots revealing the proportion of CD39-/int/hi expression in GPC3-CAR-T and shCD39-GPC3-CAR-T cells. (D) Bar plots demonstrating the expression of exhaustion markers, including PD-1, Tim-3, and Lag-3, in GPC3-CAR-T and shCD39-GPC3-CAR-T cells. (E) Flow analysis and bar plot presenting the proportion of IFN-γ+ GPC3-CAR-T and shCD39-GPC3-CAR-T cells. (F) Flow analysis and bar plot showing the proportion of Ki67+ GPC3-CAR-T and shCD39-GPC3-CAR-T cells. (G) Flow analysis and bar plot indicating the level of apoptosis in GPC3-CAR-T and shCD39-GPC3-CAR-T cells in the presence of TNF-a and FasL (100 ng/mL). All data are the mean of three independent experiments performed in duplicate. Mean ± SEM, unpaired t-test, *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.3  Mdivi-1 promoted the cytokine secretion, proliferation, and cytotoxicity but induced exhaustion and apoptosis in CAR-T cells in vitro. (A) Three candidate chemical compounds that potentially affect CD39 expression. (B) IC50 of three compounds in GPC3-CAR-T cells. (C) Bar plots showing the proportion of CD39int CAR-T cells with different dosages of compound stimulation. (D) In vitro experimental design. CD8+ CAR-T cells were sorted into CD39hi, CD39int, and CD39- subpopulations using a CD39 flow cytometry-based sorting method. Then, the sorted CAR-T cells were stimulated in the presence or absence of 50 µM mdivi-1 and incubated for 24 h. Afterward, the cells were labeled with the same flow cytometry antibodies and analyzed. The flow analysis revealed that mdivi-1 reverted the CD39- CAR-T cells into the CD39int CAR-T cell population. (E) In vitro experimental design. GPC3-CAR-T cells in the presence or absence of 50 µM mdivi-1 were stimulated by targeted antigen rhGPC3 protein. T cells were harvested and tested by flow cytometry to analyze the function of GPC3-CAR-T cells. (F) Bar plots revealing the proportion of CD39-/int/hi expression in GPC3-CAR-T cells in the presence or absence of 50 µM mdivi-1. (G) Bar plots demonstrating the expression of exhaustion markers, including PD-1, Tim-3, and Lag-3, in GPC3-CAR-T cells in the presence or absence of 50 µM mdivi-1. (H) Expression of INF-γ and Ki-67 of GPC3-CAR-T cells in the presence or absence of 50 µM mdivi-1. (I) Level of apoptosis of GPC3-CAR-T cells in the presence or absence of 50 µM mdivi-1 treated with TNF-a and FasL (100 ng/mL). Bar plots showing the statistical data of apoptotic assay. All data are the mean of three independent experiments performed in duplicate. Mean ± SEM, unpaired t-test, *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.4  shCD39-CAR-T cells combined with mdivi-1 exhibited a synergistic effect and enhanced the antitumor activity in the HCC organoid model. (A) GPC3-CAR-T and shCD39-GPC3-CAR-T cells cocultured with HCC tumor organoids for 24 h in the presence or absence of 50 µM mdivi-1. CAR-T cells were labeled with CellTrace Far Red (red), and apoptotic cells were labeled with caspase3/7 probe (green). A real-time Biotech imaging system was used to take 1 picture per hour. Scale bar = 300 mm. (B) Summary of quantitative statistics. Apoptotic cells (green) were calculated using the Spot function of Imaris software on the basis of the size and intensity threshold. The initial number of infiltrating GPC3-CAR-T cells at 0 h was defined as the base point. (C) Flow analysis revealing the proportion of CD39 and IFN-γ expression in CAR-T cells. (D) Bar plots showing the statistical data of the percentage of CD39int/hi CD8+ GPC3-CAR-T cells. (E) Bar plots presenting the proportion of IFN-γ+ GPC3-CAR-T and shCD39-GPC3-CAR-T cells in the presence or absence of 50 µM mdivi-1. (F) Bar plots demonstrating the proportion of Ki67+ GPC3-CAR-T and shCD39-GPC3-CAR-T cells in the presence or absence of 50 µM mdivi-1. *P < 0.05, **P < 0.01, ***P < 0.001 (one-way ANOVA). All data are means ± SEMs.
Fig.5  shCD39-CAR-T cells combined with mdivi-1 exhibited a potent antitumor activity by increasing the proportion of infiltrated CD39int CAR-T cells. GPC3+ Huh7 cell lines were inoculated s.c. into NSG mice at a concentration of 1 × 106 cells. After 14 days, CAR-T cells were infused via tail injection, and mdivi-1 was administered via intraperitoneal injection at a dose of 20 mg/kg. (A) Representative photo of the tumor (n = 6). (B) Line chart depicting the trend in tumor volume (n = 6). (C) Dot plots showing the tumor weight on day 40 (n = 6). (D) Tumor-infiltrating CAR-T cells analyzed by flow cytometry. The percentage of CD39int/hi CAR-T cells in tumor of different groups is shown (n = 6). (E) Flow analysis revealing the proportion of CD39 and IFN-γ expression in CAR-T cells. (F) Tumor-infiltrating CAR-T cells analyzed by flow cytometry. The percentages of IFN-γ+ of CD39int/hi CAR-T cells of different groups are presented (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001 (one-way ANOVA). All data are means ± SEMs.
Fig.6  shCD39-GPC3-CAR-T combined with mdivi-1 exerted strong infiltration and cytotoxicity. (A) Immunofluorescence staining from tumors of the Huh7 cell injection model treated with GPC3-CAR-T cells. Scale bar = 20 μm. (B) Dot plots representing infiltrated CAR-T cells and GZMB+ CAR-T cells. n = 6. *P < 0.05, **P < 0.01, ***P < 0.001 (one-way ANOVA). All data are means ± SEMs.
1 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
2 JM Llovet, F Castet, M Heikenwalder, MK Maini, V Mazzaferro, DJ Pinato, E Pikarsky, AX Zhu, RS Finn. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol 2022; 19(3): 151–172
https://doi.org/10.1038/s41571-021-00573-2
3 F Zhou, W Shang, X Yu, J Tian. Glypican-3: a promising biomarker for hepatocellular carcinoma diagnosis and treatment. Med Res Rev 2018; 38(2): 741–767
https://doi.org/10.1002/med.21455
4 M Taniguchi, S Mizuno, T Yoshikawa, N Fujinami, M Sugimoto, S Kobayashi, S Takahashi, M Konishi, N Gotohda, T Nakatsura. Peptide vaccine as an adjuvant therapy for glypican-3-positive hepatocellular carcinoma induces peptide-specific CTLs and improves long prognosis. Cancer Sci 2020; 111(8): 2747–2759
https://doi.org/10.1111/cas.14497
5 Y Sawada, T Yoshikawa, D Nobuoka, H Shirakawa, T Kuronuma, Y Motomura, S Mizuno, H Ishii, K Nakachi, M Konishi, T Nakagohri, S Takahashi, N Gotohda, T Takayama, K Yamao, K Uesaka, J Furuse, T Kinoshita, T Nakatsura. Phase I trial of a glypican-3-derived peptide vaccine for advanced hepatocellular carcinoma: immunologic evidence and potential for improving overall survival. Clin Cancer Res 2012; 18(13): 3686–3696
https://doi.org/10.1158/1078-0432.CCR-11-3044
6 N Pang, J Shi, L Qin, A Chen, Y Tang, H Yang, Y Huang, Q Wu, X Li, B He, T Li, B Liang, J Zhang, B Cao, M Liu, Y Feng, X Ye, X Chen, L Wang, Y Tian, H Li, J Li, H Hu, J He, Y Hu, C Zhi, Z Tang, Y Gong, F Xu, L Xu, W Fan, M Zhao, D Chen, H Lian, L Yang, P Li, Z Zhang. IL-7 and CCL19-secreting CAR-T cell therapy for tumors with positive glypican-3 or mesothelin. J Hematol Oncol 2021; 14(1): 118
https://doi.org/10.1186/s13045-021-01128-9
7 E Mizukoshi, S Kaneko. Immune cell therapy for hepatocellular carcinoma. J Hematol Oncol 2019; 12(1): 52
https://doi.org/10.1186/s13045-019-0742-5
8 B Allard, MS Longhi, SC Robson, J Stagg. The ectonucleotidases CD39 and CD73: novel checkpoint inhibitor targets. Immunol Rev 2017; 276(1): 121–144
https://doi.org/10.1111/imr.12528
9 S Krishna, FJ Lowery, AR Copeland, E Bahadiroglu, R Mukherjee, L Jia, JT Anibal, A Sachs, SO Adebola, D Gurusamy, Z Yu, V Hill, JJ Gartner, YF Li, M Parkhurst, B Paria, P Kvistborg, MC Kelly, SL Goff, G Altan-Bonnet, PF Robbins, SA Rosenberg. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 2020; 370(6522): 1328–1334
https://doi.org/10.1126/science.abb9847
10 T Liu, J Tan, M Wu, W Fan, J Wei, B Zhu, J Guo, S Wang, P Zhou, H Zhang, L Shi, J Li. High-affinity neoantigens correlate with better prognosis and trigger potent antihepatocellular carcinoma (HCC) activity by activating CD39+CD8+ T cells. Gut 2021; 70(10): 1965–1977
https://doi.org/10.1136/gutjnl-2020-322196
11 PK Gupta, J Godec, D Wolski, E Adland, K Yates, KE Pauken, C Cosgrove, C Ledderose, WG Junger, SC Robson, EJ Wherry, G Alter, PJ Goulder, P Klenerman, AH Sharpe, GM Lauer, WN Haining. CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog 2015; 11(10): e1005177
https://doi.org/10.1371/journal.ppat.1005177
12 A Chow, FZ Uddin, M Liu, A Dobrin, BY Nabet, L Mangarin, Y Lavin, H Rizvi, SE Tischfield, A Quintanal-Villalonga, JM Chan, N Shah, V Allaj, P Manoj, M Mattar, M Meneses, R Landau, M Ward, A Kulick, C Kwong, M Wierzbicki, J Yavner, J Egger, SS Chavan, A Farillas, A Holland, H Sridhar, M Ciampricotti, D Hirschhorn, X Guan, AL Richards, G Heller, J Mansilla-Soto, M Sadelain, CA Klebanoff, MD Hellmann, T Sen, E de Stanchina, JD Wolchok, T Merghoub, CM Rudin. The ectonucleotidase CD39 identifies tumor-reactive CD8+ T cells predictive of immune checkpoint blockade efficacy in human lung cancer. Immunity 2023; 56(1): 93–106.e6
https://doi.org/10.1016/j.immuni.2022.12.001
13 F Zou, J Tan, T Liu, B Liu, Y Tang, H Zhang, J Li. 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
https://doi.org/10.1016/j.ymthe.2021.01.021
14 M Cui, H Ding, F Chen, Y Zhao, Q Yang, Q Dong. Mdivi-1 protects against ischemic brain injury via elevating extracellular adenosine in a cAMP/CREB-CD39-dependent manner. Mol Neurobiol 2016; 53(1): 240–253
https://doi.org/10.1007/s12035-014-9002-4
15 R Bao, X Shui, J Hou, J Li, X Deng, X Zhu, T Yang. Adenosine and the adenosine A2A receptor agonist, CGS21680, upregulate CD39 and CD73 expression through E2F-1 and CREB in regulatory T cells isolated from septic mice. Int J Mol Med 2016; 38(3): 969–975
https://doi.org/10.3892/ijmm.2016.2679
16 H Liao, MC Hyman, AE Baek, K Fukase, DJ Pinsky. cAMP/CREB-mediated transcriptional regulation of ectonucleoside triphosphate diphosphohydrolase 1 (CD39) expression. J Biol Chem 2010; 285(19): 14791–14805
https://doi.org/10.1074/jbc.M110.116905
17 F Zou, L Lu, J Liu, B Xia, W Zhang, Q Hu, W Liu, Y Zhang, Y Lin, S Jing, M Huang, B Huang, B Liu, H Zhang. Engineered triple inhibitory receptor resistance improves anti-tumor CAR-T cell performance via CD56. Nat Commun 2019; 10(1): 4109
https://doi.org/10.1038/s41467-019-11893-4
18 L Broutier, G Mastrogiovanni, MM Verstegen, HE Francies, LM Gavarró, CR Bradshaw, GE Allen, R Arnes-Benito, O Sidorova, MP Gaspersz, N Georgakopoulos, BK Koo, S Dietmann, SE Davies, RK Praseedom, R Lieshout, JNM IJzermans, SJ Wigmore, K Saeb-Parsy, MJ Garnett, der Laan LJ van, M Huch. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med 2017; 23(12): 1424–1435
https://doi.org/10.1038/nm.4438
19 S Nuciforo, I Fofana, MS Matter, T Blumer, D Calabrese, T Boldanova, S Piscuoglio, S Wieland, F Ringnalda, G Schwank, LM Terracciano, CKY Ng, MH Heim. Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep 2018; 24(5): 1363–1376
https://doi.org/10.1016/j.celrep.2018.07.001
20 Y Qiao, J Chen, X Wang, S Yan, J Tan, B Xia, Y Chen, K Lin, F Zou, B Liu, X He, Y Zhang, X Zhang, H Zhang, X Wu, L Lu. Enhancement of CAR-T cell activity against cholangiocarcinoma by simultaneous knockdown of six inhibitory membrane proteins. Cancer Commun (Lond) 2023; 43(7): 788–807
https://doi.org/10.1002/cac2.12452
21 Y Simoni, E Becht, M Fehlings, CY Loh, SL Koo, KWW Teng, JPS Yeong, R Nahar, T Zhang, H Kared, K Duan, N Ang, M Poidinger, YY Lee, A Larbi, AJ Khng, E Tan, C Fu, R Mathew, M Teo, WT Lim, CK Toh, BH Ong, T Koh, AM Hillmer, A Takano, TKH Lim, EH Tan, W Zhai, DSW Tan, IB Tan, EW Newell. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018; 557(7706): 575–579
https://doi.org/10.1038/s41586-018-0130-2
22 M Qiao, F Zhou, X Liu, T Jiang, H Wang, Y Jia, X Li, C Zhao, L Cheng, X Chen, S Ren, H Liu, C Zhou. Interleukin-10 induces expression of CD39 on CD8+ T cells to potentiate anti-PD1 efficacy in EGFR-mutated non-small cell lung cancer. J Immunother Cancer 2022; 10(12): e005436
https://doi.org/10.1136/jitc-2022-005436
23 G Oliveira, K Stromhaug, S Klaeger, T Kula, DT Frederick, PM Le, J Forman, T Huang, S Li, W Zhang, Q Xu, N Cieri, KR Clauser, SA Shukla, D Neuberg, S Justesen, G MacBeath, SA Carr, EF Fritsch, N Hacohen, M Sade-Feldman, KJ Livak, GM Boland, PA Ott, DB Keskin, CJ Wu. Phenotype, specificity and avidity of antitumour CD8+ T cells in melanoma. Nature 2021; 596(7870): 119–125
https://doi.org/10.1038/s41586-021-03704-y
24 N Xie, C Wang, Y Lian, C Wu, H Zhang, Q Zhang. Inhibition of mitochondrial fission attenuates Aβ-induced microglia apoptosis. Neuroscience 2014; 256: 36–42
https://doi.org/10.1016/j.neuroscience.2013.10.011
25 LF Fan, PY He, YC Peng, QH Du, YJ Ma, JX Jin, HZ Xu, JR Li, ZJ Wang, SL Cao, T Li, F Yan, C Gu, L Wang, G Chen. Mdivi-1 ameliorates early brain injury after subarachnoid hemorrhage via the suppression of inflammation-related blood-brain barrier disruption and endoplasmic reticulum stress-based apoptosis. Free Radic Biol Med 2017; 112: 336–349
https://doi.org/10.1016/j.freeradbiomed.2017.08.003
[1] FMD-24013-OF-LT_suppl_1 Download
[1] Zhiqin Zhang, Xinli Shi, Jingmin Ji, Yinglin Guo, Qing Peng, Liyuan Hao, Yu Xue, Yiwei Liu, Caige Li, Junlan Lu, Kun Yu. Dihydroartemisinin increased the abundance of Akkermansia muciniphila by YAP1 depression that sensitizes hepatocellular carcinoma to anti-PD-1 immunotherapy[J]. Front. Med., 2023, 17(4): 729-746.
[2] Chengdong Wu, Dekai Liu, Lufei Zhang, Jingjie Wang, Yuan Ding, Zhongquan Sun, Weilin Wang. 5′-tiRNA-Gln inhibits hepatocellular carcinoma progression by repressing translation through the interaction with eukaryotic initiation factor 4A-I[J]. Front. Med., 2023, 17(3): 476-492.
[3] Chao Gao, Shenghao Wang, Weiqing Shao, Yu Zhang, Lu Lu, Huliang Jia, Kejin Zhu, Jinhong Chen, Qiongzhu Dong, Ming Lu, Wenwei Zhu, Lunxiu Qin. Rapamycin enhances the anti-tumor activity of cabozantinib in cMet inhibitor-resistant hepatocellular carcinoma[J]. Front. Med., 2022, 16(3): 467-482.
[4] Wei Zhang, Bixiang Zhang, Xiao-ping Chen. Adjuvant treatment strategy after curative resection for hepatocellular carcinoma[J]. Front. Med., 2021, 15(2): 155-169.
[5] 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.
[6] Amy Lee, Fa-Chyi Lee. Medical oncology management of advanced hepatocellular carcinoma 2019: a reality check[J]. Front. Med., 2020, 14(3): 273-283.
[7] 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.
[8] Shasha Zhu, Huimin Zhang, Li Bai. NKT cells in liver diseases[J]. Front. Med., 2018, 12(3): 249-261.
[9] 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.
[10] 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.
[11] Zhen He,Cheng Hu,Weiping Jia. miRNAs in non-alcoholic fatty liver disease[J]. Front. Med., 2016, 10(4): 389-396.
[12] 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.
[13] 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.
[14] 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.
[15] Farhad Sahebjam,John M. Vierling. Autoimmune hepatitis[J]. Front. Med., 2015, 9(2): 187-219.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed