A practical approach to immunotherapy of hepatocellular carcinoma using T cells redirected against hepatitis B virus

Sarene Koh, Noriko Shimasaki, Rossarin Suwanarusk, Zi Zong Ho, Adeline Chia, Nasirah Banu, Shanshan Wu Howland, Alice Soh Meoy Ong, Adam J Gehring, Hans Stauss, Laurent Renia, Matti Sällberg, Dario Campana, Antonio Bertoletti, Sarene Koh, Noriko Shimasaki, Rossarin Suwanarusk, Zi Zong Ho, Adeline Chia, Nasirah Banu, Shanshan Wu Howland, Alice Soh Meoy Ong, Adam J Gehring, Hans Stauss, Laurent Renia, Matti Sällberg, Dario Campana, Antonio Bertoletti

Abstract

Hepatocellular carcinoma (HCC) cells often have hepatitis B virus (HBV)-DNA integration and can be targeted by HBV-specific T cells. The use of viral vectors to introduce exogenous HBV-specific T-cell receptors (TCR) on T cells to redirect their specificity is complex and expensive to implement in clinical trials. Moreover, it raises safety concerns related to insertional mutagenesis and potential toxicity of long-lived HBV-specific T cells in patients with persistent infection. To develop a more practical and safer approach to cell therapy of HCC, we used electroporation of mRNA encoding anti-HBV TCR. Approximately 80% of CD8(+) T cells expressed functional HBV TCR 24 hours postelectroporation, an expression efficiency much higher than that obtained by retroviral transduction (~18%). Antigen-specific cytokine production of electroporated T cells was efficient within 72-hour period, after which the redirected T cells lost their HBV-specific function. Despite this transient functionality, the TCR-electroporated T cells efficiently prevented tumor seeding and suppressed the growth of established tumors in a xenograft model of HCC. Finally, we established a method for large-scale TCR mRNA electroporation that yielded large numbers of highly functional clinical-grade anti-HBV T cells. This method represents a practical approach to cell therapy of HCC and its inherently self-limiting toxicity suggests potential for application in other HBV-related pathologies.Molecular Therapy-Nucleic Acids (2013) 2, e114; doi:10.1038/mtna.2013.43; published online 13 August 2013.

Figures

Figure 1
Figure 1
High level of TCR expression and polyfunctionality of mRNA electroporated T cells. (a) Dot plots from a representative HLA-A2-HBs183-191 pentamer staining in HBV s183-TCR mRNA electroporated T cells at 6, 24, and 72 hours postelectroporation and retrovirally transduced T cells at 72 hours. T cells that were mock-electroporated or electroporated with an irrelevant CMV pp65-TCR mRNA and mock-transduced served as negative controls. The percentages of pentamer+ cells out of CD8+ or CD8− cells are indicated. (b) Expression of TCR on electroporated CD8+ T cells and frequency of IFN-γ-producing CD8+ T cells after overnight coculture with s183 peptide-loaded T2 cells were determined at several time points as indicated. Results expressed as mean + SD (n = 5). (c) Dot plots from a representative activated T cells electroporated or retrovirally transduced with s183-TCR, after overnight coculture with s183 peptide-loaded T2 cells and stained for CD8 and IFN-γ (top row), TNF-α (middle row) and IL-2 (bottom row). Mock- and CMV pp65-TCR mRNA electroporated T cells cocultured with s183 peptide-loaded T2 cells served as negative controls. The percentages of cytokine-producing cells out of total T cells are indicated. (d) TNF-α and IL-2 production by IFN-γ+ T cells demonstrate polyfunctionality of electroporated T cells. (e) Cytokine co-expression subsets expressed as a percentage of total cytokine-producing electroporated or retrovirally transduced T cells. Mean for each group is shown. Single producers (blue slice), IFN-γ+, TNF-α+ or IL-2+; double producers (red slice), IFN-γ+ TNF-α+, IFN-γ+ IL-2+ or IL-2+ TNF-α+; triple producers (green slice), IFN-γ+ IL-2+ TNF-α+.
Figure 2
Figure 2
Similar signaling capacity and cytotocixity of mRNA electroporated and retrovirally transduced T cells. (a) Sensitivity of T-cell activation using T cells produced by mRNA electroporation compared with retroviral transduction. Results are displayed as percentage of maximum IFN-γ response obtained from intracellular cytokine staining. (b) Dose dependent lysis of HepG2-env targets (solid symbols and line) or HepG2-core targets (open symbols and dotted line) by electroporated T cells (triangle) compared with retrovirally transduced T cells (circle). Results are displayed as mean of triplicate measurements + SD. (c) Expression of perforin (left panel) and granzyme (right panel) on a representative activated T cells electroporated (red histograms) or retrovirally transduced (blue histograms) with s183-TCR, after 5 hours coculture with peptide-loaded T2 cells. Coculture with unpulsed T2 cells served as negative control (gray histograms). MFI of perforin and granzyme are indicated. (d) mRNA electroporated T cells have TCM-like phenotype. Phenotype of total lymphocytes (left panel), CD8+ pentamer+ T cells (middle panel) and CD4+ Vb3+ T cells (right panel) in s183-TCR mRNA electroporated T cells (red shaded area) compared with retrovirally transduced T cells (blue shaded area). The percentages of CD45RA+/- and CD62L+/- cells within total lymphocytes or the gated CD8+ pentamer+ or CD4+ Vb3+ populations were determined by FACS. Cells were classified into different subsets: naive (CD45RA+CD62L+), TCM (CD45RA−CD62L+), TEM (CD45RA−CD62L−) and terminally differentiated EM (CD45RA+CD62L−). Results expressed as mean + SD (n = 3).
Figure 3
Figure 3
Efficient tumor clearance requires both CD8 cytotoxic and CD4 helper TCR-retrovirally transduced T cells. (a) One million HepG2-env tumor cells were inoculated by intrasplenic injection in NSG mice (n = 22). Ten days after tumor inoculation, mice were treated with 3 × 106 CD8 + 3 × 105 CD4 (n = 4), 3 × 106 CD8 (n = 4), 1.5 × 106 CD8 (n = 4), 1.5 × 106 CD4 (n = 3), or 3 × 105 CD4 (n = 4) s183-TCR transduced T cells injected i.v. Mice treated with 3 × 106 mock transduced T cells served as controls (n = 3). In all experiments, tumor size was monitored by bioluminescence imaging and results are displayed as average radiance (p/s/cm2/sr) of each mouse (colored lines) and the mean of each group (in black bold line). Control group (grey shaded area) is plotted as average radiance (p/s/cm2/sr) of the mean + SD. (b) Tumors were significantly eliminated in mice treated with total (3 × 106 CD8 + 3 × 105 CD4, n = 10) (P < 0.05) compared with 3 × 106 CD8 alone (n = 10) or 3 × 105 CD4 alone (n = 6) s183-TCR transduced T cells. Results from two independent experiments. (c) TissueFAXS staining of spleen tissue from mice treated with total s183-TCR transduced T cells at day 1 after adoptive cell transfer. Figure shows DAPI staining (blue), HepG2-env tumor cells expressing GFP (green) and CD8 (red, bottom panel; isotype control, top panel). Multiple CD8 T cells were detected in the tumor (white arrows) and in multiple fields. Two representative fields shown at 40× magnification.
Figure 4
Figure 4
Multiple infusions of activated mRNA electroporated T cells control tumor growth and maintained stable disease. One million HepG2-env tumor cells were inoculated by intrasplenic injection in NSG mice (n = 11). Nine days after tumor inoculation, mice were treated with three doses of 3 × 106 activated s183-TCR mRNA electroporated T cells per dose, (n = 4, red line) injected i.v once every 3 days. Mice treated with 3 × 106 mock-electroporated T cells served as controls (n = 3, grey shaded area). Tumor size was monitored by bioluminescence imaging and plotted as average radiance (p/s/cm2/sr) of the mean + SD.
Figure 5
Figure 5
Prevention of HCC tumor cells seeding by mRNA electroporated T cells. One million HepG2-env tumor cells were inoculated by intrasplenic injection in NSG mice (n = 14). Four hours later, mice (n = 4 or 3 per group) were treated with graded doses (0.75, 1.5, 3 × 106 pentamer+ CD8) of s183-TCR mRNA electroporated T cells injected i.v. Mice treated with 3 × 106 mock-electroporated T cells served as controls (grey shaded area). Tumor size was monitored by bioluminescence imaging and plotted as average radiance (p/s/cm2/sr) of the mean + SD.
Figure 6
Figure 6
High level of TCR expression and multifunctionality of mRNA electroporated T cells produced in large-scale, clinical-grade conditions. A schematic illustrating cell numbers, efficiency, yield, and functionality of laboratory-grade (top row) vs. clinical-grade (bottom row) electroporation of T cells. Dot plot of CD8 and HLA-A2-HBs183-191 pentamer staining in s183-TCR mRNA electroporated T cells at 24 hours postelectroporation. Bar charts show the frequency of IFN-γ, TNF-α, and IL-2 producing cells out of CD8 or CD4 electroporated T cells.

References

    1. Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000. Int J Cancer. 2001;94:153–156.
    1. El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012;142:1264–1273.e1.
    1. Sung WK, Zheng H, Li S, Chen R, Liu X, Li Y, et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet. 2012;44:765–769.
    1. Brechot C, Pourcel C, Louise A, Rain B, Tiollais P. Presence of integrated hepatitis B virus DNA sequences in cellular DNA of human hepatocellular carcinoma. Nature. 1980;286:533–535.
    1. Edman JC, Gray P, Valenzuela P, Rall LB, Rutter WJ. Integration of hepatitis B virus sequences and their expression in a human hepatoma cell. Nature. 1980;286:535–538.
    1. Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet. 2003;362:1907–1917.
    1. Straathof KC, Bollard CM, Popat U, Huls MH, Lopez T, Morriss MC, et al. Treatment of nasopharyngeal carcinoma with Epstein-Barr virus–specific T lymphocytes. Blood. 2005;105:1898–1904.
    1. Rooney CM, Smith CA, Ng CY, Loftin SK, Sixbey JW, Gan Y, et al. Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients. Blood. 1998;92:1549–1555.
    1. Gehring AJ, Ho ZZ, Tan AT, Aung MO, Lee KH, Tan KC, et al. Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology. 2009;137:682–690.
    1. June CH, Blazar BR, Riley JL. Engineering lymphocyte subsets: tools, trials and tribulations. Nat Rev Immunol. 2009;9:704–716.
    1. Park TS, Rosenberg SA, Morgan RA. Treating cancer with genetically engineered T cells. Trends Biotechnol. 2011;29:550–557.
    1. Gehring AJ, Xue SA, Ho ZZ, Teoh D, Ruedl C, Chia A, et al. Engineering virus-specific T cells that target HBV infected hepatocytes and hepatocellular carcinoma cell lines. J Hepatol. 2011;55:103–110.
    1. Ando K, Moriyama T, Guidotti LG, Wirth S, Schreiber RD, Schlicht HJ, et al. Mechanisms of class I restricted immunopathology. A transgenic mouse model of fulminant hepatitis. J Exp Med. 1993;178:1541–1554.
    1. Maini MK, Boni C, Lee CK, Larrubia JR, Reignat S, Ogg GS, et al. The role of virus-specific CD8(+) cells in liver damage and viral control during persistent hepatitis B virus infection. J Exp Med. 2000;191:1269–1280.
    1. Bertoletti A, Maini MK. Protection or damage: a dual role for the virus-specific cytotoxic T lymphocyte response in hepatitis B and C infection. Curr Opin Microbiol. 2000;3:387–392.
    1. Marshall E. Gene therapy a suspect in leukemia-like disease. Science. 2002;298:2002–2003.
    1. Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008;118:3132–3142.
    1. Recchia A, Bonini C, Magnani Z, Urbinati F, Sartori D, Muraro S, et al. Retroviral vector integration deregulates gene expression but has no consequence on the biology and function of transplanted T cells. Proc Natl Acad Sci USA. 2006;103:1457–1462.
    1. Hunziker L, Klenerman P, Zinkernagel RM, Ehl S. Exhaustion of cytotoxic T cells during adoptive immunotherapy of virus carrier mice can be prevented by B cells or CD4+ T cells. Eur J Immunol. 2002;32:374–382.
    1. Antony PA, Piccirillo CA, Akpinarli A, Finkelstein SE, Speiss PJ, Surman DR, et al. CD8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells. J Immunol. 2005;174:2591–2601.
    1. Ho WY, Yee C, Greenberg PD. Adoptive therapy with CD8(+) T cells: it may get by with a little help from its friends. J Clin Invest. 2002;110:1415–1417.
    1. Robbins PF, Dudley ME, Wunderlich J, El-Gamil M, Li YF, Zhou J, et al. Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy. J Immunol. 2004;173:7125–7130.
    1. Zhou J, Dudley ME, Rosenberg SA, Robbins PF. Persistence of multiple tumor-specific T-cell clones is associated with complete tumor regression in a melanoma patient receiving adoptive cell transfer therapy. J Immunother. 2005;28:53–62.
    1. Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118:294–305.
    1. Gattinoni L, Klebanoff CA, Palmer DC, Wrzesinski C, Kerstann K, Yu Z, et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin Invest. 2005;115:1616–1626.
    1. Klebanoff CA, Gattinoni L, Torabi-Parizi P, Kerstann K, Cardones AR, Finkelstein SE, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci USA. 2005;102:9571–9576.
    1. van Loenen MM, de Boer R, Amir AL, Hagedoorn RS, Volbeda GL, Willemze R, et al. Mixed T cell receptor dimers harbor potentially harmful neoreactivity. Proc Natl Acad Sci USA. 2010;107:10972–10977.
    1. Zhao Y, Moon E, Carpenito C, Paulos CM, Liu X, Brennan AL, et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res. 2010;70:9053–9061.
    1. Llovet JM, Schwartz M, Mazzaferro V. Resection and liver transplantation for hepatocellular carcinoma. Semin Liver Dis. 2005;25:181–200.
    1. Faria LC, Gigou M, Roque-Afonso AM, Sebagh M, Roche B, Fallot G, et al. Hepatocellular carcinoma is associated with an increased risk of hepatitis B virus recurrence after liver transplantation. Gastroenterology. 2008;134:1890–9; quiz 2155.
    1. Rabinovich PM, Komarovskaya ME, Ye ZJ, Imai C, Campana D, Bahceci E, et al. Synthetic messenger RNA as a tool for gene therapy. Hum Gene Ther. 2006;17:1027–1035.
    1. Shimasaki N, Fujisaki H, Cho D, Masselli M, Lockey T, Eldridge P, et al. A clinically adaptable method to enhance the cytotoxicity of natural killer cells against B-cell malignancies. Cytotherapy. 2012;14:830–840.
    1. Li L, Liu LN, Feller S, Allen C, Shivakumar R, Fratantoni J, et al. Expression of chimeric antigen receptors in natural killer cells with a regulatory-compliant non-viral method. Cancer Gene Ther. 2010;17:147–154.
    1. Belnoue E, Guettier C, Kayibanda M, Le Rond S, Crain-Denoyelle AM, Marchiol C, et al. Regression of established liver tumor induced by monoepitopic peptide-based immunotherapy. J Immunol. 2004;173:4882–4888.
    1. Rénia L, Rodrigues MM, Nussenzweig V. Intrasplenic immunization with infected hepatocytes: a mouse model for studying protective immunity against malaria pre-erythrocytic stage. Immunology. 1994;82:164–168.

Source: PubMed

3
Prenumerera