Selection of a Clinical Lead TCR Targeting Alpha-Fetoprotein-Positive Liver Cancer Based on a Balance of Risk and Benefit

Xiaobing Luo, Huijuan Cui, Lun Cai, Wei Zhu, Wei-Chih Yang, Michael Patrick, Shigui Zhu, Jiaqi Huang, Xin Yao, Yihong Yao, Yukai He, Yun Ji, Xiaobing Luo, Huijuan Cui, Lun Cai, Wei Zhu, Wei-Chih Yang, Michael Patrick, Shigui Zhu, Jiaqi Huang, Xin Yao, Yihong Yao, Yukai He, Yun Ji

Abstract

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer with a poor prognosis and limited therapeutic options. Alpha-fetoprotein (AFP), an established clinical biomarker of HCC, has been employed as an attractive target for T cell-based immunotherapy against this disease given its high expression in the tumor and restricted expression in normal tissues. We have identified a number of T cell receptors (TCRs) recognizing the HLA-A*02:01 restricted AFP158-166 peptide FMNKFIYEI, providing a TCR candidate pool for identifying TCRs with optimal clinical benefit. To select the ideal AFP TCR for clinical use, we evaluated the efficacy and safety profile of 7 TCRs by testing their potency toward AFP-expressing HCC cells and their specificity based upon reactivity to normal and transformed cells covering a wide variety of primary cell types and HLA serotypes. Furthermore, we assessed their cross-reactivity to potential protein candidates in the human genome by an extensive alanine scan (X-scan). We first selected three TCR candidates based on the in vitro anti-tumor activity. Next we eliminated two potential cross-reactive TCRs based on their reactivity against normal and transformed cells covering a variety of primary cell types and HLA serotypes, respectively. We then excluded the potential cross-reactivity of the selected TCR with a protein candidate identified by X-scan. At present we have selected an AFP TCR with the optimal affinity, function, and safety profile, bearing properties that are expected to allow AFP TCR redirected T cells to specifically differentiate between AFP levels on tumor and normal tissues. An early phase clinical trial using T cells transduced with this TCR to treat HCC patients (NCT03971747) has been initiated.

Keywords: T cell receptor (TCR); X-scan; alloreactivity; alpha-fetoprotein (AFP); cross-reactivity; hepatocellular carcinoma (HCC); immunotherapy.

Copyright © 2020 Luo, Cui, Cai, Zhu, Yang, Patrick, Zhu, Huang, Yao, Yao, He and Ji.

Figures

Figure 1
Figure 1
AFP TCR1, 2, and 3 demonstrate comparable potent reactivity toward the target cells in vitro. (A) Alignment of amino acid sequence of CDR3 regions of the α (left) and β (right) chains of the 7 AFP TCRs. (B) FACS analysis of the surface expression of TCR β chain and AFP tetramer staining of T cells 8 days after transducing of indicated AFP TCRs. The percentage of TCR β (top) or AFP tetramer (bottom) positive cells as indicated. (C) Cytokine production and degranulation of AFP TCR T cells upon encountering HepG2 target cells as revealed by intracellular staining of IL-2, IFN-γ (top), TNF-α and CD107a (bottom). (D) IFN-γ concentration in the supernatant of overnight, 1:1 ratio co-culture of AFP TCR T cells with HepG2 target cells or control Huh7 cells measured by ELISA. Data represents the mean + s.d. of quadruplicate co-culture samples. (E) Cytolytic capacity of AFP TCR T cells toward HepG2 target cells over a 4-day co-culture. Data represents the mean of triplicate samples derived from RTCA instrument, as compared to target only wells. (E:T): effect to target ratio. Colors are represented as in (D). (F) Time to eradicate 50% of target cells (KT50) of different AFP TCR T cells at the indicated effect to target (E:T) ratio. Colors are represented as in (D). T cells transduced with TCR 8 and TCR 11 did not eradicate 50% of the target by the end of the analysis at E:T = 0.25:1, thus were not plotted. Data shown in b-f is representative of at least 3 independent experiments on T cells isolated from 3 healthy donors. UT, untransduced T cells.
Figure 2
Figure 2
AFP TCR 2 shows lower than background activity toward primary neurons and no alloreactivity compared to TCR 1 and 3. (A) IFN-γ concentration in the supernatant of overnight, 1:1 ratio co-culture of the indicated AFP TCR T cells with iCell derived neurons measured by ELISA. Data represents the mean + s.d. of the triplicate co-cultures and is representative of T cells prepared from 2 healthy donors. (B) IFN-γ concentration in the supernatant of overnight, 1:1 ratio co-culture of the indicated AFP TCR T cells with a panel of Epstein-Barr virus transformed B cell lines expressing various HLA alleles measured by ELISA. Data represents the mean + s.d. of quadruplicate co-cultures and is representative of T cells prepared from 2 healthy donors. HepG2 and Huh7 cells were included as positive and negative controls, respectively, in both (A,B). *P < 0.05 (unpaired two-tailed Student's t-test).
Figure 3
Figure 3
AFP TCR 2 displays no reactivity toward a variety of HLA-A*02:01+ primary cells. (A) IFN-γ concentration in the supernatant of overnight, 1:1 ratio co-culture of UT or T cells expressing AFP TCR 2 with primary hepatocytes measured by ELISA. T cells were prepared from 2 healthy donors and human primary hepatocytes were obtained from 2 adult HLA-A*02:01+ donors. Data represents the mean + s.d. of quadruplicate co-cultures. (B) 4-1BB upregulation by T cells after co-culture as described in a. Cells were further analyzed by FACS for surface expression of 4-1BB on total CD3+ untransduced T cells or AFP TCR 2 transduced T cells. HepG2 and Huh7 cells were included as positive and negative controls, respectively, in both (A,B). (C) Summary of reactivity from co-culture experiment of T cells expressing AFP TCR 2 with primary hepatocytes, primary lung and kidney epithelial cells, and iCell derived primary cells including neurons, astrocytes, cardiomyocyte, and endothelial cells.
Figure 4
Figure 4
AFP TCR 2 shows no alloreactivity. (A) IFN-γ concentration in the supernatant of overnight, 1:1 ratio co-culture of UT or AFP TCR 2 transduced T cells with a panel of 14 Epstein-Barr virus transformed B cell lines expressing various HLA alleles measured by ELISA. Data represents the mean + s.d. of quadruplicate co-cultures and is representative of 3 healthy donors. A total of 38 transformed B cell lines were tested. (B) Frequencies of class I HLA serotypes in Chinese Han population. Serotypes were sorted by frequency for each subclass from high to low; those covered by the 38 transformed B cell lines in our test are shown in blue while those not covered are shown in orange. For HLA-B, only the top 20 serotypes are displayed. (C) Summary of the number of class I HLA serotypes covered in this study and corresponding cumulative percentage in Chinese Han population.
Figure 5
Figure 5
AFP TCR 2 is unlikely to cross-react with other proteins in the human genome. (A) Graphic presentation of X-scan results showing the replaceability of each amino acid of AFP158−166 epitope. Peptides with replacement of each amino acid residue with every other 19 amino acids were used to activate the TCR 2 transduced T cells. The amount of IFN-γ produced by AFP TCR 2 transduced T cells after stimulation with mutated epitope (mut) was compared to that from wild type AFP158−166 peptide (wt) and the ratios are presented. Dashed line indicates the cut-off for tolerance. Data represents the mean + s.d. of duplicate co-cultures and is representative of 3 healthy donors. (B) ENPP1 and AFP expression in the indicated cell lines tested by Western blot. GAPDH was used as loading control. (C) IFN-γ concentrations in the supernatant of overnight, 1:1 ratio co-culture of UT or AFP TCR 2 transduced T cells with the indicated cell lines measured by ELISA. Data represents the mean + s.d. of quadruplicate co-cultures and is representative of 3 healthy donors.

References

    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2018) 68:394–424. 10.3322/caac.21492
    1. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. . Cancer statistics in China, 2015. CA Cancer J Clin. (2016) 66:115–32. 10.3322/caac.21338
    1. Wege H, Li J, Ittrich H. Treatment lines in hepatocellular carcinoma. Visc Med. (2019) 35:266–72. 10.1159/000501749
    1. Yao W, He JC, Yang Y, Wang JM, Qian YW, Yang T, et al. . The prognostic value of tumor-infiltrating lymphocytes in hepatocellular carcinoma: a systematic review and meta-analysis. Sci Rep. (2017) 7:7525. 10.1038/s41598-017-08128-1
    1. Ding W, Xu X, Qian Y, Xue W, Wang Y, Du J, et al. . Prognostic value of tumor-infiltrating lymphocytes in hepatocellular carcinoma: a meta-analysis. Medicine. (2018) 97:e13301. 10.1097/MD.0000000000013301
    1. Shen Y, Xia M, Zhang J, Xu L, Yang J, Chen A, et al. . IRF-1 and p65 mediate upregulation of constitutive HLA-A antigen expression by hepatocellular carcinoma cells. Mol Immunol. (2009) 46:2045–53. 10.1016/j.molimm.2009.03.001
    1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. (2015) 348:62–8. 10.1126/science.aaa4967
    1. June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. (2018) 359:1361–5. 10.1126/science.aar6711
    1. Majzner RG, Mackall CL. Clinical lessons learned from the first leg of the CAR T cell journey. Nat Med. (2019) 25:1341–55. 10.1038/s41591-019-0564-6
    1. Ji Y, Hocker JD, Gattinoni L. Enhancing adoptive T cell immunotherapy with microRNA therapeutics. Semin Immunol. (2016) 28:45–53. 10.1016/j.smim.2015.11.006
    1. D'Ippolito E, Schober K, Nauerth M, Busch DH. T cell engineering for adoptive T cell therapy: safety and receptor avidity. Cancer Immunol Immunother. (2019) 68:1701–12. 10.1007/s00262-019-02395-9
    1. Wolf B, Zimmermann S, Arber C, Irving M, Trueb L, Coukos G. Safety and tolerability of adoptive cell therapy in cancer. Drug Saf. (2019) 42:315–34. 10.1007/s40264-018-0779-3
    1. Galle PR, Foerster F, Kudo M, Chan SL, Llovet JM, Qin S, et al. . Biology and significance of alpha-fetoprotein in hepatocellular carcinoma. Liver Int. (2019) 39:2214. 10.1111/liv.14223
    1. Sun L, Guo H, Jiang R, Lu L, Liu T, He X. Engineered cytotoxic T lymphocytes with AFP-specific TCR gene for adoptive immunotherapy in hepatocellular carcinoma. Tumour Biol. (2016) 37:799–806. 10.1007/s13277-015-3845-9
    1. Docta RY, Ferronha T, Sanderson JP, Weissensteiner T, Pope GR, Bennett AD, et al. . Tuning T-cell receptor affinity to optimize clinical risk-benefit when targeting alpha-fetoprotein-positive liver cancer. Hepatology. (2019) 69:2061–75. 10.1002/hep.30477
    1. Liu H, Xu Y, Xiang J, Long L, Green S, Yang Z, et al. . Targeting alpha-fetoprotein. (AFP)-MHC complex with CAR T-cell therapy for liver cancer. Clin Cancer Res. (2017) 23:478–88. 10.1158/1078-0432.CCR-16-1203
    1. Lipika Goyal MF, Meyer T, Lynn G, Jordi B, El-Khoueiry A, Hausner P, et al. Abstract 3183: initial safety of AFP SPEAR T-cells in patients with advanced hepatocellular carcinoma. Cancer Res. (2019) 79:3183 10.1158/1538-7445.AM2019-3183
    1. Eureka Therapeutics Achieves Regression Of Metastatic Liver Cancer Using Et140202 T-Cell Therapy 2019. Available online at: (accessed April 1, 2020).
    1. Zhu W, Peng Y, Wang L, Hong Y, Jiang X, Li Q, et al. . Identification of alpha-fetoprotein-specific T-cell receptors for hepatocellular carcinoma immunotherapy. Hepatology. (2018) 68:574–89. 10.1002/hep.29844
    1. Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, et al. . Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. (2009) 114:535–46. 10.1182/blood-2009-03-211714
    1. Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DA, Feldman SA, et al. . T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. (2011) 19:620–6. 10.1038/mt.2010.272
    1. Cameron BJ, Gerry AB, Dukes J, Harper JV, Kannan V, Bianchi FC, et al. . Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med. (2013) 5:197ra103. 10.1126/scitranslmed.3006034
    1. Morgan RA, Chinnasamy N, Abate-Daga D, Gros A, Robbins PF, Zheng Z, et al. . Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. (2013) 36:133–51. 10.1097/CJI.0b013e3182829903
    1. Cai L, Caraballo Galva LD, Peng Y, Luo X, Zhu W, Yao Y, et al. Preclinical Studies of the Off-Target Reactivity of AFP158-Specific TCR Engineered T Cells. Front Immunol. (2020) 11:607 10.3389/fimmu.2020.00607
    1. Gatz SA, Pohla H, Schendel DJ. A PCR-SSP method to specifically select HLA-A*0201 individuals for immunotherapeutic studies. Tissue Antigens. (2000) 55:532–47. 10.1034/j.1399-0039.2000.550604.x
    1. Kawai HF, Kaneko S, Honda M, Shirota Y, Kobayashi K. alpha-fetoprotein-producing hepatoma cell lines share common expression profiles of genes in various categories demonstrated by cDNA microarray analysis. Hepatology. (2001) 33:676–91. 10.1053/jhep.2001.22500
    1. Zhou F, Cao H, Zuo X, Zhang T, Zhang X, Liu X, et al. . Deep sequencing of the MHC region in the Chinese population contributes to studies of complex disease. Nat Genet. (2016) 48:740–6. 10.1038/ng.3576
    1. Gonzalez-Galarza FF, Takeshita LY, Santos EJ, Kempson F, Maia MH, da Silva AL, et al. . Allele frequency net 2015 update: new features for HLA epitopes, KIR and disease and HLA adverse drug reaction associations. Nucleic Acids Res. (2015) 43(Database issue):D784–8. 10.1093/nar/gku1166
    1. Bacci S, De Cosmo S, Prudente S, Trischitta V. ENPP1 gene, insulin resistance and related clinical outcomes. Curr Opin Clin Nutr Metab Care. (2007) 10:403–9. 10.1097/MCO.0b013e3281e386c9
    1. Price DA, Asher TE, Wilson NA, Nason MC, Brenchley JM, Metzler IS, et al. . Public clonotype usage identifies protective Gag-specific CD8+ T cell responses in SIV infection. J Exp Med. (2009) 206:923–36. 10.1084/jem.20081127
    1. Valkenburg SA, Josephs TM, Clemens EB, Grant EJ, Nguyen TH, Wang GC, et al. . Molecular basis for universal HLA-A*0201-restricted CD8+ T-cell immunity against influenza viruses. Proc Natl Acad Sci USA. (2016) 113:4440–5. 10.1073/pnas.1603106113
    1. Irving M, Zoete V, Hebeisen M, Schmid D, Baumgartner P, Guillaume P, et al. . Interplay between T cell receptor binding kinetics and the level of cognate peptide presented by major histocompatibility complexes governs CD8+ T cell responsiveness. J Biol Chem. (2012) 287:23068–78. 10.1074/jbc.M112.357673
    1. Heemskerk MH, de Paus RA, Lurvink EG, Koning F, Mulder A, Willemze R, et al. . Dual HLA class I and class II restricted recognition of alloreactive T lymphocytes mediated by a single T cell receptor complex. Proc Natl Acad Sci USA. (2001) 98:6806–11. 10.1073/pnas.111162298
    1. Obenaus M, Leitao C, Leisegang M, Chen X, Gavvovidis I, van der Bruggen P, et al. . Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice. Nat Biotechnol. (2015) 33:402–7. 10.1038/nbt.3147

Source: PubMed

3
Abonneren