Lymphocytes transiently expressing virus-specific T cell receptors reduce hepatitis B virus infection

Janine Kah, Sarene Koh, Tassilo Volz, Erica Ceccarello, Lena Allweiss, Marc Lütgehetmann, Antonio Bertoletti, Maura Dandri, Janine Kah, Sarene Koh, Tassilo Volz, Erica Ceccarello, Lena Allweiss, Marc Lütgehetmann, Antonio Bertoletti, Maura Dandri

Abstract

Adoptive transfer of T cells engineered to express a hepatitis B virus-specific (HBV-specific) T cell receptor (TCR) may supplement HBV-specific immune responses in chronic HBV patients and facilitate HBV control. However, the risk of triggering unrestrained proliferation of permanently engineered T cells raises safety concerns that have hampered testing of this approach in patients. The aim of the present study was to generate T cells that transiently express HBV-specific TCRs using mRNA electroporation and to assess their antiviral and pathogenetic activity in vitro and in HBV-infected human liver chimeric mice. We assessed virological and gene-expression changes using quantitative reverse-transcriptase PCR (qRT-PCR), immunofluorescence, and Luminex technology. HBV-specific T cells lysed HBV-producing hepatoma cells in vitro. In vivo, 3 injections of HBV-specific T cells caused progressive viremia reduction within 12 days of treatment in animals reconstituted with haplotype-matched hepatocytes, whereas viremia remained stable in mice receiving irrelevant T cells redirected toward hepatitis C virus-specific TCRs. Notably, increases in alanine aminotransferase levels, apoptotic markers, and human inflammatory cytokines returned to pretreatment levels within 9 days after the last injection. T cell transfer did not trigger inflammation in uninfected mice. These data support the feasibility of using mRNA electroporation to engineer HBV TCR-redirected T cells in patients with chronic HBV infection.

Conflict of interest statement

Conflict of interest: S. Koh is the Director of Research at Lion TCR Pte. Ltd., a biotech company developing T cell receptors for treatment of virus-related cancers and chronic viral diseases. A. Bertoletti is a cofounder of Lion TCR Pte. Ltd.

Figures

Figure 1. Lytic and antiviral function of…
Figure 1. Lytic and antiviral function of mRNA HBV–specific TCR–electroporated T cells in vitro.
(A) Activated T cells were electroporated with HBV s183–TCR or c18-TCR mRNA, and TCR expression was determined 24 hours after electroporation. Mock electroporated T cells served as negative control. Shown are representative plots. The percentages of HLA-A2/pentamer+ cells out of CD8+ or CD8− T cells are indicated. (B) TCR expression on electroporated cells was measured longitudinally from 24 hours to 96 hours. Electroporated T cells were cocultured with their respective peptide-pulsed T2 cells for 18 hours, and the frequencies of IFN-γ–producing CD8+ T cells out of total lymphocytes were quantified. (C) The ability of mRNA TCR–electroporated T cells to lyse HepG2.2.15 HBV–producing cells at 1:3, 1:5, and 1:100 E:T ratios within 24 hours after T cell addition was compared with that of retroviral transduced (TCR RV) T cells. (D) Sensitivity of T cell activation, displayed as percentage of maximum IFN-γ response using mRNA TCR–electroporated T cells compared with retroviral-transduced T cells (upper panel). MRNA HBV s183–TCR–electroporated T cells were cocultured with HBV s183–191 genotype B (FLLTKILTI) or genotype A/C/D (FLLTRILTI) peptide-loaded T2 cells. The percentages of CD8+ or CD8− T cells producing IFN-γ are indicated (lower panel). (E) Mock, mRNA HBV s183–TCR, or c18-TCR–electroporated T cells were cocultured with either HepG2 or HepG2.2.15 cells for 24 hours. The percentages of CD8+ or CD8− T cells producing IFN-γ are indicated. (F) mRNA HBV s183–TCR or c18-TCR–electroporated T cells were cocultured with mock or HBV-infected HepG2-NTCP for 24 hours, and IFNG gene expression was determined using NanoString analysis. (G) Mock or mRNA HBV s183–TCR–electroporated T cells were cocultured with HepG2.2.15 cells at a 1:3 E:T ratio for 24 hours, and intracellular HBV DNA was quantified by real-time quantitative PCR (qPCR). AST levels were determined in coculture media. Shown are means of percentage reduction in intracellular HBV DNA ± SD (black bars) and means of AST ± SD (gray bars) from 3 independent experiments (right panel).
Figure 2. mRNA HBV–specific TCR–electroporated T cells…
Figure 2. mRNA HBV–specific TCR–electroporated T cells show antiviral efficacy in vivo.
(A) Schematic representation of the experiment performed to assess the effect of 1 single injection of electroporated effector T cells in high viremic mice reconstituted with haplotype-matched hepatocytes. (B) Viremia changes relative to baseline levels determined after 4 and 6 days in individual mice upon 1 injection of mRNA HBV s183–TCR T cells (n = 5) and in untreated controls (n = 2). (C) ALT levels determined in HBV-infected and uninfected mice receiving a single injection of effector T cells. (D) Schematic representation of the experiment performed to assess the antiviral effect of multiple injections of electroporated effector T cells in high viremic mice reconstituted either with haplotype-matched (n = 3) or -mismatched (n = 3) human hepatocytes and in comparison with mice that were left untreated (n = 3). (E) Median viremia changes determined within each group depicted in D and relative to baseline levels determined in individual mice upon 3 injections of mRNA HBV s183–TCR T cells. Blood was taken 4, 8, and 12 days after the first T cell injection. (F) Median changes in levels of circulating HBeAg were determined by ELISA in all animal groups. BL, baseline.
Figure 3. mRNA HBV–specific TCR–electroporated T cells…
Figure 3. mRNA HBV–specific TCR–electroporated T cells are specifically recruited and activated in livers of haplotype-matched HBV-infected mice.
(A) Liver tissues of humanized HBV-infected mice and uninfected mice that underwent 3 injections of HBV s183–TCR T cells and were sacrificed 4 days after the third T cell transfer were used for immunofluorescence. Human hepatocytes were identified using human-specific CK18 Abs (red). Transferred human immune cells were visualized using human-specific CD45 Abs (green). (B) Liver tissues of HBV-infected mice adoptively transferred with either HLA-A2– or HLA-A1–presenting human hepatocytes were costained with HBV core–specific Ab (green) and human CD45-specific Ab (red). (C) Transcript levels of human T cell response–related genes (GZMB and IFNG) and human ISGs (MxA, STAT1, TAP1, and HLA-A) were measured by quantitative reverse-transcriptase PCR (qRT-PCR) and normalized against human housekeeping transcripts. Statistical analysis was performed with GraphPad Prism 6 software.
Figure 4. Antiviral and inflammatory events after…
Figure 4. Antiviral and inflammatory events after in vivo multiple injections of different mRNA HBV–specific TCR–electroporated T cells.
(A) Median viremia changes relative to baseline levels were determined as indicated after 4, 8, and 12 days upon multiple injections of mRNA HBV s183–TCR T cells (n = 3), mRNA c18-TCR T cells (n = 3), and mRNA mock TCR T cells (n = 3) as well as in untreated controls (n = 6). (B) Individual reduction (shown as percentages) of viremia relative to baseline levels determined on days 4, 8, and 12 upon transfer of mRNA s183–191 T cells and mRNA c18 T cells. Transcriptional changes of human T cell response–related genes (C, GZMB; E, IFNG) were measured by qRT-PCR and normalized against human housekeeping transcripts. (D) ALT levels were determined in uninfected (n = 3) and HBV-infected mice receiving multiple injections of effector T cells presenting s183 (n = 3), c18 (n = 3), or HCV NS3 as mock control (n = 3) in comparison with HBV-infected control mice (n = 6). (F) Median changes in human IFN-γ serum protein levels were determined by multiplex measurement in HBV-infected (s183 median = 1280 ng/ml or c18 median = 160 ng/ml) as well as uninfected (s183 median = 3.6 ng/ml) mice that received 3 injections of HBV-specific effector T cells relative to HBV-infected control mice (n = 4; median = 0.53 ng/ml).
Figure 5. Adoptive transfer of mRNA HBV–specific…
Figure 5. Adoptive transfer of mRNA HBV–specific TCR–electroporated T cells leads to temporary limited liver inflammation and cell damage.
(A) Schematic representation of the experiment performed to assess the effect of multiple injections of electroporated effector T cells after treatment cessation both on inflammatory and virological parameters. (B) ALT levels were determined in 1 HBV-infected animal shortly before T cell injection (baseline) after receiving 3 injections of HBV-specific c18-TCR T cells (day 12) and 9 days after the last T cell injection (day 21). (C) Longitudinal changes in viremia relative to baseline were determined at 4, 8, 12, and 21 days after the first T cell transfer as depicted in A. (D) Serum protein levels of CCL2 were determined in both 1 HBV-infected control mouse (gray) and 1 mouse receiving multiple T cell injections (c18-TCR), and that was monitored for 21 days. (E) Longitudinal changes in levels of circulating HBeAg were determined by ELISA in the same mice described in D.

References

    1. Feldman SA, Assadipour Y, Kriley I, Goff SL, Rosenberg SA. Adoptive cell therapy–tumor-infiltrating lymphocytes, T-cell receptors, and chimeric antigen receptors. Semin Oncol. 2015;42(4):626–639. doi: 10.1053/j.seminoncol.2015.05.005.
    1. Schub A, Schuster IG, Hammerschmidt W, Moosmann A. CMV-specific TCR-transgenic T cells for immunotherapy. J Immunol. 2009;183(10):6819–6830. doi: 10.4049/jimmunol.0902233.
    1. Banu N, et al. Building and optimizing a virus-specific T cell receptor library for targeted immunotherapy in viral infections. Sci Rep. 2014;4:4166.
    1. Oh HL, et al. Engineering T cells specific for a dominant severe acute respiratory syndrome coronavirus CD8 T cell epitope. J Virol. 2011;85(20):10464–10471. doi: 10.1128/JVI.05039-11.
    1. Zhang Y, et al. Transduction of human T cells with a novel T-cell receptor confers anti-HCV reactivity. PLoS Pathog. 2010;6(7):e1001018. doi: 10.1371/journal.ppat.1001018.
    1. Gehring AJ, et al. Engineering virus-specific T cells that target HBV infected hepatocytes and hepatocellular carcinoma cell lines. J Hepatol. 2011;55(1):103–110. doi: 10.1016/j.jhep.2010.10.025.
    1. Mueller K, et al. Protective capacity of virus-specific T cell receptor-transduced CD8 T cells in vivo. J Virol. 2012;86(19):10866–10869. doi: 10.1128/JVI.01472-12.
    1. Stemberger C, et al. Lowest numbers of primary CD8(+) T cells can reconstitute protective immunity upon adoptive immunotherapy. Blood. 2014;124(4):628–637. doi: 10.1182/blood-2013-12-547349.
    1. Leen AM, et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood. 2013;121(26):5113–5123. doi: 10.1182/blood-2013-02-486324.
    1. Thimme R, et al. CD8(+) T cells mediate viral clearance and disease pathogenesis during acute hepatitis B virus infection. J Virol. 2003;77(1):68–76. doi: 10.1128/JVI.77.1.68-76.2003.
    1. Bertoletti A, Ferrari C. Adaptive immunity in HBV infection. J Hepatol. 2016;64(1 Suppl):S71–S83.
    1. Boni C, et al. Characterization of hepatitis B virus (HBV)-specific T-cell dysfunction in chronic HBV infection. J Virol. 2007;81(8):4215–4225. doi: 10.1128/JVI.02844-06.
    1. Webster GJ, et al. Longitudinal analysis of CD8+ T cells specific for structural and nonstructural hepatitis B virus proteins in patients with chronic hepatitis B: implications for immunotherapy. J Virol. 2004;78(11):5707–5719. doi: 10.1128/JVI.78.11.5707-5719.2004.
    1. Rehermann B, Ferrari C, Pasquinelli C, Chisari FV. The hepatitis B virus persists for decades after patients’ recovery from acute viral hepatitis despite active maintenance of a cytotoxic T-lymphocyte response. Nat Med. 1996;2(10):1104–1108. doi: 10.1038/nm1096-1104.
    1. Revill P, Testoni B, Locarnini S, Zoulim F. Global strategies are required to cure and eliminate HBV infection. Nat Rev Gastroenterol Hepatol. 2016;13(4):239–248. doi: 10.1038/nrgastro.2016.7.
    1. Ilan Y, et al. Adoptive transfer of immunity to hepatitis B virus after T cell-depleted allogeneic bone marrow transplantation. Hepatology. 1993;18(2):246–252.
    1. Lau GK, et al. Clearance of hepatitis B surface antigen after bone marrow transplantation: role of adoptive immunity transfer. Hepatology. 1997;25(6):1497–1501. doi: 10.1002/hep.510250631.
    1. Loggi E, et al. Anti-HBs re-seroconversion after liver transplantation in a patient with past HBV infection receiving a HBsAg positive graft. J Hepatol. 2009;50(3):625–630. doi: 10.1016/j.jhep.2008.08.026.
    1. Bohne F, et al. T cells redirected against hepatitis B virus surface proteins eliminate infected hepatocytes. Gastroenterology. 2008;134(1):239–247. doi: 10.1053/j.gastro.2007.11.002.
    1. Krebs K, et al. T cells expressing a chimeric antigen receptor that binds hepatitis B virus envelope proteins control virus replication in mice. Gastroenterology. 2013;145(2):456–465. doi: 10.1053/j.gastro.2013.04.047.
    1. Qasim W, et al. Immunotherapy of HCC metastases with autologous T cell receptor redirected T cells, targeting HBsAg in a liver transplant patient. J Hepatol. 2015;62(2):486–491. doi: 10.1016/j.jhep.2014.10.001.
    1. Guidotti LG, et al. Immunosurveillance of the liver by intravascular effector CD8(+) T cells. Cell. 2015;161(3):486–500. doi: 10.1016/j.cell.2015.03.005.
    1. Sitia G, Isogawa M, Iannacone M, Campbell IL, Chisari FV, Guidotti LG. MMPs are required for recruitment of antigen-nonspecific mononuclear cells into the liver by CTLs. J Clin Invest. 2004;113(8):1158–1167. doi: 10.1172/JCI21087.
    1. Koh S, et al. A practical approach to immunotherapy of hepatocellular carcinoma using T cells redirected against hepatitis B virus. Mol Ther Nucleic Acids. 2013;2:e114. doi: 10.1038/mtna.2013.43.
    1. Dandri M, Petersen J. Chimeric mouse model of hepatitis B virus infection. J Hepatol. 2012;56(2):493–495. doi: 10.1016/j.jhep.2011.05.037.
    1. Koh S, Bertoletti A. Circumventing failed antiviral immunity in chronic hepatitis B virus infection: triggering virus-specific or innate-like T cell response? Med Microbiol Immunol. 2015;204(1):87–94. doi: 10.1007/s00430-014-0377-7.
    1. Tan AT, et al. Host ethnicity and virus genotype shape the hepatitis B virus-specific T-cell repertoire. J Virol. 2008;82(22):10986–10997. doi: 10.1128/JVI.01124-08.
    1. Chulanov VP, Shipulin GA, Schaefer S, Gerlich WH. Kinetics of HBV DNA and HBsAg in acute hepatitis B patients with and without coinfection by other hepatitis viruses. J Med Virol. 2003;69(3):313–323. doi: 10.1002/jmv.10291.
    1. Allweiss L, et al. Immune cell responses are not required to induce substantial hepatitis B virus antigen decline during pegylated interferon-alpha administration. J Hepatol. 2014;60(3):500–507. doi: 10.1016/j.jhep.2013.10.021.
    1. Bissig KD, et al. Human liver chimeric mice provide a model for hepatitis B and C virus infection and treatment. J Clin Invest. 2010;120(3):924–930. doi: 10.1172/JCI40094.
    1. Nakagawa S, et al. Targeted induction of interferon-λ in humanized chimeric mouse liver abrogates hepatotropic virus infection. PLoS One. 2013;8(3):e59611. doi: 10.1371/journal.pone.0059611.
    1. Werle-Lapostolle B, et al. Persistence of cccDNA during the natural history of chronic hepatitis B and decline during adefovir dipivoxil therapy. Gastroenterology. 2004;126(7):1750–1758. doi: 10.1053/j.gastro.2004.03.018.
    1. Dandri M, Locarnini S. New insight in the pathobiology of hepatitis B virus infection. Gut. 2012;61(Suppl 1):i6–i17. doi: 10.1136/gutjnl-2012-302056.
    1. Volz T, et al. The entry inhibitor Myrcludex-B efficiently blocks intrahepatic virus spreading in humanized mice previously infected with hepatitis B virus. J Hepatol. 2013;58(5):861–867. doi: 10.1016/j.jhep.2012.12.008.
    1. Petersen J, et al. Prevention of hepatitis B virus infection in vivo by entry inhibitors derived from the large envelope protein. Nat Biotechnol. 2008;26(3):335–341. doi: 10.1038/nbt1389.
    1. Belloni L, et al. IFN-α inhibits HBV transcription and replication in cell culture and in humanized mice by targeting the epigenetic regulation of the nuclear cccDNA minichromosome. J Clin Invest. 2012;122(2):529–537. doi: 10.1172/JCI58847.
    1. Allweiss L, et al. Proliferation of primary human hepatocytes and prevention of hepatitis B virus reinfection efficiently deplete nuclear cccDNA in vivo. Gut. [published online ahead of print on April 20, 2017]. .
    1. Lucifora J, et al. Specific and nonhepatotoxic degradation of nuclear hepatitis B virus cccDNA. Science. 2014;343(6176):1221–1228. doi: 10.1126/science.1243462.
    1. Giersch K, et al. Hepatitis Delta co-infection in humanized mice leads to pronounced induction of innate immune responses in comparison to HBV mono-infection. J Hepatol. 2015;63(2):346–353. doi: 10.1016/j.jhep.2015.03.011.

Source: PubMed

3
Subscribe