Anti-GPC3-CAR T Cells Suppress the Growth of Tumor Cells in Patient-Derived Xenografts of Hepatocellular Carcinoma

Zhiwu Jiang, Xiaofeng Jiang, Suimin Chen, Yunxin Lai, Xinru Wei, Baiheng Li, Simiao Lin, Suna Wang, Qiting Wu, Qiubin Liang, Qifa Liu, Muyun Peng, Fenglei Yu, Jianyu Weng, Xin Du, Duanqing Pei, Pentao Liu, Yao Yao, Ping Xue, Peng Li, Zhiwu Jiang, Xiaofeng Jiang, Suimin Chen, Yunxin Lai, Xinru Wei, Baiheng Li, Simiao Lin, Suna Wang, Qiting Wu, Qiubin Liang, Qifa Liu, Muyun Peng, Fenglei Yu, Jianyu Weng, Xin Du, Duanqing Pei, Pentao Liu, Yao Yao, Ping Xue, Peng Li

Abstract

Background: The lack of a general clinic-relevant model for human cancer is a major impediment to the acceleration of novel therapeutic approaches for clinical use. We propose to establish and characterize primary human hepatocellular carcinoma (HCC) xenografts that can be used to evaluate the cytotoxicity of adoptive chimeric antigen receptor (CAR) T cells and accelerate the clinical translation of CAR T cells used in HCC.

Methods: Primary HCCs were used to establish the xenografts. The morphology, immunological markers, and gene expression characteristics of xenografts were detected and compared to those of the corresponding primary tumors. CAR T cells were adoptively transplanted into patient-derived xenograft (PDX) models of HCC. The cytotoxicity of CAR T cells in vivo was evaluated.

Results: PDX1, PDX2, and PDX3 were established using primary tumors from three individual HCC patients. All three PDXs maintained original tumor characteristics in their morphology, immunological markers, and gene expression. Tumors in PDX1 grew relatively slower than that in PDX2 and PDX3. Glypican 3 (GPC3)-CAR T cells efficiently suppressed tumor growth in PDX3 and impressively eradicated tumor cells from PDX1 and PDX2, in which GPC3 proteins were highly expressed.

Conclusion: GPC3-CAR T cells were capable of effectively eliminating tumors in PDX model of HCC. Therefore, GPC3-CAR T cell therapy is a promising candidate for HCC treatment.

Keywords: CAR; PDX; T cells; cell therapy; hepatocellular carcinoma.

Figures

Figure 1
Figure 1
Characterization of patient-derived xenograft (PDX) model of hepatocellular carcinoma. (A) Patient-derived xenografts (PDXs) were established, and tumor growth was measured at a given time for each PDX xenograft. Tumor volume was calculated as (a × b2)/2, the length (a), and width (b) of tumor. (B,C) H&E and IHC staining tissue of primary tumor and PDXs. The xenograft demonstrates morphology, glypican 3, and AFP staining consistent with the original human tumor. Brown color indicates positive staining. IgG was used as a negative control. Scale bar is 50 μm. (D) mRNA expression level of tumor-related genes [MET (MET proto-oncogene, receptor tyrosine kinase), CTNNB1 (catenin beta 1), AXIN1 (axin 1), TP53 (tumor protein p53), RB1 (RB transcriptional corepressor 1), PTEN (phosphatase and tensin homolog), BCL2 (BCL2, apoptosis regulator), AFP (alpha fetoprotein), KRT19 (keratin 19), CDKN2A (cyclin-dependent kinase inhibitor 2A), CDKN1B (cyclin-dependent kinase inhibitor 1B), and CCND1 (cyclin D1)] in primary tumor and PDXs. Results represent mean ± SD of three individual experiments.
Figure 2
Figure 2
Construction of chimeric anti-glypican 3 (GPC3) vectors and generation of GPC3-chimeric antigen receptor (CAR) T cells. (A) Schematic representation of a lentiviral vector encoding the signal peptide, anti-GPC3 scFv, CD28 transmembrane domain, 4-1BB costimulatory endodomain, and CD3ζ signaling domain along with eGFP using 2A. (B) A representative of GPC3-CARs expression on human T cells transduced with lentivirus was analyzed using flow cytometry, which detected eGFP at days 7 and 14. (C) Transduction efficiency. Results represent mean ± SD of six individual experiments. No difference was detected between the percentages of GPC3-CAR T and Control CAR T. (D) The expansion of transduced T cells in vitro from day 0 to 21. Results represent mean ± SD of three individual experiments.
Figure 3
Figure 3
Phenotypic analysis and cytokines produced in glypican 3 (GPC3)-chimeric antigen receptor (CAR) T cells. (A) Flow cytometry comparison of the common surface phenotype of GPC3-CAR T cells (red line) at day 14 of culture with freshly isolated T cells (blue line). Black line represents the isotype control. Histogram overlays show 16 markers related to lymphocyte activation, differentiation, migration, adhesion, and exhaustion. (B) Interferon-γ and (C) interleukin-2 were secreted by the indicated modified CAR T cells co-cultured with hepatocellular carcinoma cell lines and A549 for 24 h. Results represent triplicates. ***P < 0.001 with T-test.
Figure 4
Figure 4
Cytotoxicity of glypican 3 (GPC3)-chimeric antigen receptor (CAR) T cells against targeted cells in vitro. (A) Cytotoxicity of GPC3-CAR T cells was assayed by coincubation with Huh-7-GL (GPC3+), HepG2-GL (GPC3+), and A549-GL (GPC3−) cells at E:T ratios ranging from 1:3 to 5:1. (B) Serial killing of GPC3-CAR cells against Huh-7-GL and HepG2-GL cells at E: T ratios of 1:1 in indicated durations. Results represent mean ± SEM.
Figure 5
Figure 5
Cytotoxicity of glypican 3 (GPC3)-chimeric antigen receptor (CAR) T cells against hepatocellular carcinoma cell lines in xenografts. (A,B) Growth curve of Huh-7 and HepG2 xenografts (n = 5) treated with the Control- or GPC3-CAR T cells at indicated time point (arrow). At the end of the experiment, the tumors treated with GPC3-CAR T cells were significantly smaller than those in the Control group. (C,D) Huh-7 and HepG2 tumor weights from the mice treated with CAR T cells at the end of the experiment, respectively. (E,F) GPC3-CAR T cells in tumors were significantly higher than Control-CAR T groups. Results represent mean ± SD. *P < 0.05, **P < 0.01, and ***P < 0.001 with T-test.
Figure 6
Figure 6
Glypican 3 (GPC3)-chimeric antigen receptor (CAR) T cells efficiently abolish growth of patient-derived xenografts (PDXs) of hepatocellular carcinoma. (A–C) Growth curve of PDX1, PDX2, and PDX3 (n = 5) treated with the Control- or GPC3-CAR T cells at indicated time point (arrow). At the end of the experiment, the tumors treated with GPC3-CAR T cells were significantly smaller than those in the Control group. (D–F) PDX1, PDX2, and PDX3 tumor weights from the mice treated with CAR T cells at the end of the experiment. (G–I) GPC3-CAR T cells in tumors were significantly higher than Control-CAR T groups. Results represent mean ± SD. ***P < 0.001 with T-test.

References

    1. Sharma S, Khosla R, David P, Rastogi A, Vyas A, Singh D, et al. CD4+CD25+CD127low regulatory T cells play predominant anti-tumor suppressive role in hepatitis B virus associated hepatocellular carcinoma. Front Immunol (2015) 6:49.10.3389/fimmu.2015.00049
    1. Page AJ, Cosgrove DC, Philosophe B, Pawlik TM. Hepatocellular carcinoma: diagnosis, management, and prognosis. Surg Oncol Clin N Am (2014) 23(2):289–311.10.1016/j.soc.2013.10.006
    1. Jayachandran A, Dhungel B, Steel JC. Epithelial-to-mesenchymal plasticity of cancer stem cells: therapeutic targets in hepatocellular carcinoma. J Hematol Oncol (2016) 9(1):74.10.1186/s13045-016-0307-9
    1. Clavien PA, Petrowsky H, DeOliveira ML, Graf R. Strategies for safer liver surgery and partial liver transplantation. N Engl J Med (2007) 356(15):1545–59.10.1056/NEJMra065156
    1. Waghray A, Murali AR, Menon KN. Hepatocellular carcinoma: from diagnosis to treatment. World J Hepatol (2015) 7(8):1020–9.10.4254/wjh.v7.i8.1020
    1. Toyoda H, Kumada T, Kiriyama S, Sone Y, Tanikawa M, Hisanaga Y, et al. Changes in the characteristics and survival rate of hepatocellular carcinoma from 1976 to 2000: analysis of 1365 patients in a single institution in Japan. Cancer (2004) 100(11):2415–21.10.1002/cncr.20289
    1. Lee SS, Shin HS, Kim HJ, Lee SJ, Lee HS, Hyun KH, et al. Analysis of prognostic factors and 5-year survival rate in patients with hepatocellular carcinoma: a single-center experience. Korean J Hepatol (2012) 18(1):48–55.10.3350/kjhep.2012.18.1.48
    1. Tentler JJ, Tan AC, Weekes CD, Jimeno A, Leong S, Pitts TM, et al. Patient-derived tumour xenografts as models for oncology drug development. Nat Rev Clin Oncol (2012) 9(6):338–50.10.1038/nrclinonc.2012.61
    1. Jin K, Teng L, Shen Y, He K, Xu Z, Li G. Patient-derived human tumour tissue xenografts in immunodeficient mice: a systematic review. Clin Transl Oncol (2010) 12(7):473–80.10.1007/s12094-010-0540-6
    1. Bissig-Choisat B, Kettlun-Leyton C, Legras XD, Zorman B, Barzi M, Chen LL, et al. Novel patient-derived xenograft and cell line models for therapeutic testing of pediatric liver cancer. J Hepatol (2016) 65(2):325–33.10.1016/j.jhep.2016.04.009
    1. Huynh H, Soo KC, Chow PK, Panasci L, Tran E. Xenografts of human hepatocellular carcinoma: a useful model for testing drugs. Clin Cancer Res (2006) 12(14 Pt 1):4306–14.10.1158/1078-0432.CCR-05-2568
    1. Dargel C, Bassani-Sternberg M, Hasreiter J, Zani F, Bockmann JH, Thiele F, et al. T cells engineered to express a T-cell receptor specific for glypican-3 to recognize and kill hepatoma cells in vitro and in mice. Gastroenterology (2015) 149(4):1042–52.10.1053/j.gastro.2015.05.055
    1. Gao H, Li K, Tu H, Pan X, Jiang H, Shi B, et al. Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res (2014) 20(24):6418–28.10.1158/1078-0432.CCR-14-1170
    1. Wenpeng L, Linjie G, Purva R, Ekaterina M, Xiuhua G, Meng-Feng W, et al. Redirecting T cells to glypican-3 with 4-1BB zeta chimeric antigen receptors results in Th1 polarization and potent antitumor activity. Hum Gene Ther (2016).10.1089/hum.2016.025
    1. Ye W, Jiang Z, Li GX, Xiao Y, Lin S, Lai Y, et al. Quantitative evaluation of the immunodeficiency of a mouse strain by tumor engraftments. J Hematol Oncol (2015) 8:59.10.1186/s13045-015-0156-y
    1. Jiang Z, Deng M, Wei X, Ye W, Xiao Y, Lin S, et al. Heterogeneity of CD34 and CD38 expression in acute B lymphoblastic leukemia cells is reversible and not hierarchically organized. J Hematol Oncol (2016) 9(1):94.10.1186/s13045-016-0310-1
    1. Xiao Y, Wei X, Jiang Z, Wang X, Ye W, Liu X, et al. Loss of Angiopoietin-like 7 diminishes the regeneration capacity of hematopoietic stem and progenitor cells. J Hematol Oncol (2015) 8:7.10.1186/s13045-014-0102-4
    1. Nakano KYT, Nezu JI, Tsumoda H, Igawa T, Konishi H, Inventor. Anti-Glypican 3 Antibody. United States patent US 20070190599 (2007).
    1. Venepalli NK, Goff L. Targeting the HGF-cMET axis in hepatocellular carcinoma. Int J Hepatol (2013) 2013:341636.10.1155/2013/341636
    1. Nault JC, De Reynies A, Villanueva A, Calderaro J, Rebouissou S, Couchy G, et al. A hepatocellular carcinoma 5-gene score associated with survival of patients after liver resection. Gastroenterology (2013) 145(1):176–87.10.1053/j.gastro.2013.03.051
    1. Jeannin P, Herbault N, Delneste Y, Magistrelli G, Lecoanet-Henchoz S, Caron G, et al. Human effector memory T cells express CD86: a functional role in naive T cell priming. J Immunol (1999) 162(4):2044–8.
    1. Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, Ingaramo M, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med (2015) 21(6):581–90.10.1038/nm.3838
    1. Hoffmann P, Hofmeister R, Brischwein K, Brandl C, Crommer S, Bargou R, et al. Serial killing of tumor cells by cytotoxic T cells redirected with a CD19-/CD3-bispecific single-chain antibody construct. Int J Cancer (2005) 115(1):98–104.10.1002/ijc.20908
    1. Xin H, Wang K, Hu G, Xie F, Ouyang K, Tang X, et al. Establishment and characterization of 7 novel hepatocellular carcinoma cell lines from patient-derived tumor xenografts. PLoS One (2014) 9(1):e85308.10.1371/journal.pone.0085308
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol (2008) 26:677–704.10.1146/annurev.immunol.26.021607.090331
    1. Polakis P. Wnt signaling and cancer. Genes Dev (2000) 14(15):1837–51.10.1101/gad.14.15.1837
    1. Zhang YJ, Chen SY, Chen CJ, Santella RM. Polymorphisms in cyclin D1 gene and hepatocellular carcinoma. Mol Carcinog (2002) 33(2):125–9.10.1002/mc.10028.abs
    1. Domaica CI, Fuertes MB, Rossi LE, Girart MV, Avila DE, Rabinovich GA, et al. Tumour-experienced T cells promote NK cell activity through trogocytosis of NKG2D and NKp46 ligands. EMBO Rep (2009) 10(8):908–15.10.1038/embor.2009.92
    1. Han EQ, Li XL, Wang CR, Li TF, Han SY. Chimeric antigen receptor-engineered T cells for cancer immunotherapy: progress and challenges. J Hematol Oncol (2013) 6:47.10.1186/1756-8722-6-47
    1. Newick K, O’Brien S, Moon E, Albelda SM. CAR T cell therapy for solid tumors. Annu Rev Med (2017) 68(1).10.1146/annurev-med-062315-120245
    1. John LB, Devaud C, Duong CP, Yong CS, Beavis PA, Haynes NM, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res (2013) 19(20):5636–46.10.1158/1078-0432.CCR-13-0458
    1. Moon EK, Wang LC, Dolfi DV, Wilson CB, Ranganathan R, Sun J, et al. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clin Cancer Res (2014) 20(16):4262–73.10.1158/1078-0432.CCR-13-2627

Source: PubMed

3
订阅