Mini review

Genetically-engineered T cells to treat viral hepatitis-associated liver cancer: is it possible?

  • Received: 07 June 2017 Accepted: 14 July 2017 Published: 19 July 2017
  • Adoptive T-cell immunotherapy is gaining credibility and popularity as a potential tool to cure cancer. Genetic engineering utilization of this therapeutic mode currently comes in the forms of chimeric antigen receptor- and T-cell receptor-engineered T cells. This short review focuses on opportunities to use engineered T cells to treat viral hepatitis-associated hepatocellular carcinoma.

    Citation: Johan Garssen, Juandy Jo. Genetically-engineered T cells to treat viral hepatitis-associated liver cancer: is it possible?[J]. AIMS Allergy and Immunology, 2017, 1(1): 43-49. doi: 10.3934/Allergy.2017.1.43

    Related Papers:

    [1] Kochar Khasro Saleh, Semih Dalkiliç, Lütfiye Kadioğlu Dalkiliç, Bahra Radhaa Hamarashid, Sevda Kirbağ . Targeting cancer cells: from historic methods to modern chimeric antigen receptor (CAR) T-Cell strategies. AIMS Allergy and Immunology, 2020, 4(2): 32-49. doi: 10.3934/Allergy.2020004
    [2] Issam Tout, Marie Marotel, Isabelle Chemin, Uzma Hasan . HBV and the importance of TLR9 on B cell responses. AIMS Allergy and Immunology, 2017, 1(3): 124-137. doi: 10.3934/Allergy.2017.3.124
    [3] Andrey Mamontov, Alexander Polevshchikov, Yulia Desheva . Mast cells in severe respiratory virus infections: insights for treatment and vaccine administration. AIMS Allergy and Immunology, 2023, 7(1): 1-23. doi: 10.3934/Allergy.2023001
    [4] Ling Wang, Shunbin Ning . “Toll-free” pathways for production of type I interferons. AIMS Allergy and Immunology, 2017, 1(3): 143-163. doi: 10.3934/Allergy.2017.3.143
    [5] Daniil Shevyrev, Valeriy Tereshchenko, Olesya Manova, Vladimir kozlov . Homeostatic proliferation as a physiological process and a risk factor for autoimmune pathology. AIMS Allergy and Immunology, 2021, 5(1): 18-32. doi: 10.3934/Allergy.2021002
    [6] Stefano Regis, Fabio Caliendo, Alessandra Dondero, Francesca Bellora, Beatrice Casu, Cristina Bottino, Roberta Castriconi . Main NK cell receptors and their ligands: regulation by microRNAs. AIMS Allergy and Immunology, 2018, 2(2): 98-112. doi: 10.3934/Allergy.2018.2.98
    [7] Ken S. Rosenthal, Daniel H. Zimmerman . J-LEAPS vaccines elicit antigen specific Th1 responses by promoting maturation of type 1 dendritic cells (DC1). AIMS Allergy and Immunology, 2017, 1(2): 89-100. doi: 10.3934/Allergy.2017.2.89
    [8] James Peterson . Affinity and avidity models in autoimmune disease. AIMS Allergy and Immunology, 2018, 2(1): 45-81. doi: 10.3934/Allergy.2018.1.45
    [9] Michael D. Caponegro, Jeremy Tetsuo Miyauchi, Stella E. Tsirka . Contributions of immune cell populations in the maintenance, progression, and therapeutic modalities of glioma. AIMS Allergy and Immunology, 2018, 2(1): 24-44. doi: 10.3934/Allergy.2018.1.24
    [10] Caterina Marangio, Rosa Molfetta, Erisa Putro, Alessia Carnevale, Rossella Paolini . Exploring the dynamic of NKG2D/NKG2DL axis: A central regulator of NK cell functions. AIMS Allergy and Immunology, 2025, 9(2): 70-88. doi: 10.3934/Allergy.2025005
  • Adoptive T-cell immunotherapy is gaining credibility and popularity as a potential tool to cure cancer. Genetic engineering utilization of this therapeutic mode currently comes in the forms of chimeric antigen receptor- and T-cell receptor-engineered T cells. This short review focuses on opportunities to use engineered T cells to treat viral hepatitis-associated hepatocellular carcinoma.


    1. The Adoptive Cellular Therapy

    We are witnessing a rapid advancement in immunotherapy, in particular adoptive T-cell immunotherapy, against specific cancers over the past decade. Currently, adoptive T-cell immunotherapy comprises of three different classes through the use of tumor-infiltrating lymphocytes (TILs), chimeric antigen receptor (CAR)-and T-cell receptor (TCR)-engineered T cells [1]. While TILs are obtained through the isolation from tumor mass, the latter two methods obtain T cells through genetic engineering. Both CAR-and TCR-redirected systems result in the form of antigen-specific T cells, which would permit the immune system to confer an adequate anti-tumor immune response that maybe not present naturally [1].

    Several published articles on the distinction between TCR-and CAR-engineered T-cell systems are available (please read review [2] in particular). TCR is an αβ heterodimer receptor, naturally expressed on the T-cell surface, which binds to a specific peptide-major histocompatibility complex/MHC unit. TCR associates with CD3 molecules (γ, δ, ε and ζ chains) to provide intracellular signaling domains, which is a prerequisite for a T cell to confer an immune response. In addition, the presence of either co-receptor CD4 or CD8 supports the responsiveness of a T cell to be activated by TCR binding to as few as one peptide-MHC unit. This sensitive, yet specific system allows T cells to physiologically target intracellular antigens in a form of peptide-MHC complexes [2]. The CAR refers to a synthetic construct typically comprising a single-chain antibody variable fragment, an extracellular domain/hinge, a transmembrane domain, one or more intracellular signaling domains (e.g. CD28 or 4-1BB) and cytoplasmic immunoreceptor tyrosine activation motifs derived from CD3-ζ chain [3]. Less commonly, the CD3-ζ chain is substituted with the γ chain of FcεRI or the CD3-ε chain [4]. The CAR system can target cell surface antigens independent of MHC. Transformed cells, such as cancerous cells, usually express a particular cell surface antigen at high density, hence these kinds of target cells can be recognized and eliminated by the CAR-engineered T cells [2]. Due to the pronounced differences between the CAR and TCR systems (as shown in Figure 1), it is indeed difficult to directly compare these two systems. Nonetheless, it is fair to state that CAR system has more potential to be utilized in a wide population, since it does not face any MHC restriction. On the other hand, TCR is a more specific system than CAR due to its capability to recognize and respond to as few as only one particular peptide-MHC complex [2].

    Figure 1. Distinction between T-cell receptor and chimeric antigen receptor. (A & B) T-cell receptor/TCR is an αβ heterodimer that binds to a peptide presented by major histocompatibility complex/MHC. TCR is expressed as a functional complex along with CD3 molecules (γ, δ, ε and ζ chains). CD4 or CD8 molecule, as a co-receptor, supports the effective binding of a TCR complex to a particular peptide-MHC unit, i.e. CD4 recognizes MHC class Ⅱ, while CD8 recognizes MHC class Ⅰ. Furthermore, both CD4 and CD8 interact and bind lymphocyte-specific protein kinase/Lck. (C) Chimeric antigen receptor/CAR is a hybrid molecule contains a single-chain variable fragment (scFv) recognition domain, a hinge region, a transmembrane domain, a co-stimulatory molecule (typically CD28 or 4-1BB) and cytoplasmic immunoreceptor tyrosine-based activation motifs (not shown) derived from CD3-ζ chain. The scFV domain is derived from the heavy-and light-chain variable regions of a specific monoclonal antibody (VH and VL, respectively), hence it is capable to bind particular cell surface antigens (e.g. CD19). Both the TCR and CAR signalling are initiated by Lck-mediated phosphorylation of immuno-tyrosine activation motifs within the cytoplasmic domains of CD3 molecules.

    As mentioned above, both CAR and TCR systems are currently being explored as potential treatment targets in different cancers. While the clinical efficacy of CAR-engineered T cells against solid tumors is still limited, partly due to the local immunosuppressive tumor environment, it has been demonstrated that CAR treatment against CD19 antigen (expressed by B cells) resulted in complete remission in several patients suffering from B-cell malignancies [5,6,7,8]. Despite its efficacy, CAR therapy, however, is associated with several side effects. In general, it could cause "the cytokine release syndrome" because of the systemic release of pro-inflammatory cytokines, e.g. TNF-α and IL-6, at high levels [9]. In particular, CD19-specific CAR therapy has been reported to result in B-cell aplasia that prompts for exogenous administration of immunoglobulin [2]. On the other hand, trials using TCR-engineered T cells demonstrated some success in treating both solid and hematological tumors [2]. However, current TCR-engineered T cells have been developed to target tumor-associated antigens (e.g. MAGE-A3 or NY-ESO-1), which are actually self antigens perse [10]. This implies that TCR-generated T cells could cross-react to similar peptide-MHC complexes in heathy tissue, although are presented at very low levels, resulting in severe, sometime lethal adverse events [11,12]. Taken together, despite these two systems are potentially efficacious in treating certain cancers, their safety profiles are still of concern and they need to be addressed and properly rectified before they can be routinely used in the clinical setting.

    In addition, due to the complex process to produce a specific type of CAR-or TCR-engineered T cells per patient, there is a cautious remark questioning the ability of patients or public health system to pay substantial costs incurred by these modes of treatment. It is forecasted that the price ranges of CAR-or TCR-engineered T cells are between $120,000 and $300,000 per patient [13,14]. This estimated price seriously challenges the likelihood to implement these modes of treatment in the clinical setting [13]. Therefore, any creative solution to reduce the incurred costs, e.g. converting the engineered T cells from a specific patient-customized item to a mass-produced item [13], will be crucial in order to support and sustain the economic viability of these novel therapeutic modes.


    2. The Adoptive T Cells Against Viral Hepatitis

    Functional T-cell responses are required to control HBV and HCV replication, as well as to eliminate viral infection [15,16]. Therefore, both CAR and TCR systems have been studied the context of HBV and HCV infection as well. The published viral epitopes, targeted by CAR and TCR-engineered T cells, are summarized in Table 1. With respect to HCV infection, both systems have been extensively studied in vitro, i.e. by targeting HLA-A*02:01-restricted epitopes within HCV NS3 or NS5A protein, as well as HCV E2 glycoprotein in the TCR and CAR systems, respectively [17,18,19,20]. Both systems demonstrated their efficacy in controlling HCV replication in vitro with a low level of cytotoxicity. Nonetheless, subsequent in vivo and clinical studies are required to confirm whether these efficacies observed in vitro can be replicated without a profound risk of morbidity or mortality. Another hindrance is that potent antiviral drugs to cure HCV-infected patients are already available on the market [21], hence questioning the necessity to develop adoptive T-cell immunotherapy against chronic HCV infection.

    Table 1. Published HBV and HCV epitopes targeted by CAR-or TCR-engineered T cells.
    System HBV HCV
    CAR S domain of HBs antigen [22] e137 of HCV/E2 glycoprotein [18]
    TCR HLA-A*02:01-restricted HBs183-191 [14,24] HLA-A*02:01-restricted NS31073-1081 [17,19,20]
    HLA-A*02:01-restricted HBs370-379 [23,24] HLA-A*02:01-restricted NS5A1992-2000 [17,20]
    HLA-A*02:01-restricted HBc18-27 [23,24]
    HLA-C*08:01-restricted HBs171-80 [24]
     | Show Table
    DownLoad: CSV

    Lack of potent anti-HBV drugs in contrast, has accelerated studies on adoptive T-cell immunotherapy. A research group led by Ulrike Protzer uses CAR that targets HBV envelope protein and has tested this system in a HBV-transgenic mouse model. This group demonstrated that CAR-engineered T cells were able to control HBV replication with a transient liver damage in vivo. In addition, their study showed that the presence of HBs antigens in murine sera did not interfere with the functionality of HBV-specific CAR-engineered T cells [22]. However, it is noteworthy that the levels of HBs antigen in murine sera (~1,000–1,200 IU/mL) correspond only to the levels observed in the low-replicate phase of chronic Hepatitis B [22]. This implies that it is elusive yet on whether CAR-engineered T cells can be functional in patients with higher levels of HBs antigen. As an alternative, the HBV-specific TCR-engineered T-cell research, led primarily by Antonio Bertoletti's group, is focusing on targeting certain MHC-restricted epitopes within viral antigens, e.g. HBV surface or core antigen. This group has demonstrated that TCR-engineered T cells are also able to control HBV replication in both cell lines and xenograft mice [14,23].

    Taken together, both CAR-and TCR-engineered T cells have merits to be further developed as a potential treatment tool against chronic HBV infection. However, both methods are associated with a risk of significant liver inflammation and consequently, liver damage. Therefore, extensive studies are required to provide sufficient evidence in order to support the efficacy and safety of these treatment modes for patients with chronic HBV infection.


    3. The Adoptive T Cells Against Viral Hepatitis-associated Liver Cancer

    It has been acknowledged that chronic viral hepatitis contributes to the majority of primary hepatocellular carcinoma/HCC cases [25]. In line with the primary usage of adoptive T-cell therapy against cancers, this mode of treatment has a potential utility for the treatment of HBV-or HCV-associated HCC.

    It is important to point out that a high frequency of HBV DNA integration is observed in the genome of HBV-associated HCC cells, resulting in the expression of HBV antigens by tumor cells [26]. This allows the usage of TCR-engineered T cells to treat HBV-associated HCC. Unlike self antigens, HBV antigens are not found in healthy tissues. Hence, these viral antigens theoretically can serve as better target antigens in certain HCC cases. However, since non-cancerous but HBV-infected hepatocytes also express HBV antigens, HBV-specific TCR-engineered T cells could attack those infected hepatocytes. This potentially could cause severe liver damage.

    To address this concern, Bertoletti's group decided to treat a liver-transplanted patient who developed extrahepatic HCC metastasis with HBV-DNA integration, as the first use of HBV-specific TCR-engineered T cells in a clinical setting [27]. The patient importantly exhibited HBV surface antigen restricted by HLA-A*02:01 (i.e., HBs183-191 epitope), only in tissues with the extrahepatic HCC metastasis. Hence, the metastatic tissue could be attacked by HBV-specific TCR-engineered T cells with a significant reduced risk of damaging healthy liver tissue. Indeed, this group demonstrated the clinical potential and safety of using HBV-specific TCR-engineered T cells by choosing a suitable patient [27]. This finding still needs to be validated in clinical studies using large number of patients. Nonetheless, this milestone study suggests that the adoptive T-cell therapy can be used against a selected group of HBV-associated HCC cases, such as to prevent or treat HCC recurrence in liver-transplanted patients with HBV-positive HCC [28]. In addition, this research group recently modified its TCR engineering technology from the viral vector-based to mRNA electroporation-based method [14]. This TCR mRNA electroporation method is, arguably, a promising technology [14] due to its successful rate of engineering T cells (approximately half of the engineered T cells were endowed with antigen-specific functionality), its better safety profile (because of the transient functionality of engineered T cells) and its much reduced costs (approximately $30,000 per patient because of the less complexity and effort to engineer T cells). This improved technology, if proven, would sustain the economic viability of developing and implementing HBV-specific TCR-engineered T cells for clinical use.

    In contrast, HCV as an RNA virus does not integrate with the host genome. Therefore, despite a study demonstrated the utilization of HCV-specific TCR-engineered T cells against HCV-associated HCC in cell lines and xenograft mice [29], it will be difficult to select a suitable group of HCV-associated HCC patients in order to be treated with this treatment mode.


    4. Concluding Remark

    We are entering a new exciting era where adoptive T-cell immunotherapy is extensively studied against viral hepatitis-associated liver cancer. Based on the evidence presented in this review, we are optimistic and feel that the adoptive T-cell immunotherapy, at least in a form of TCR-engineered T cells, could serve as a novel alternative yet effective treatment for a selected group of HBV-associated HCC patients.


    Acknowledgment

    No fund or grant was received for this article.


    Conflict of Interest

    Both authors are also employees of Nutricia Research and therefore declare potential conflicts of interest.


    [1] June CH, Riddell SR, Schumacher TN (2015) Adoptive cellular therapy: a race to the finish line.Sci Transl Med 7: 280–287.
    [2] Harris DT, Kranz DM (2016) Adoptive T cell therapies: a comparison of t cell receptors and chimeric antigen receptors. Trends Pharmacol Sci 37: 220–230. doi: 10.1016/j.tips.2015.11.004
    [3] Figueroa JA, Reidy A, Mirandola L, et al. (2015) Chimeric antigen receptor engineering: a right step in the evolution of adoptive cellular immunotherapy. Int Rev Immunol 34: 154–187. doi: 10.3109/08830185.2015.1018419
    [4] Ramos CA, Dotti G (2011) Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer therapy. Expert Opin Biol Th 11: 855–873. doi: 10.1517/14712598.2011.573476
    [5] Porter DL, Levine BL, Kalos M, et al. (2011) Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. New Engl J Med 365: 725–733. doi: 10.1056/NEJMoa1103849
    [6] Brentjens RJ, Davila ML, Riviere I, et al. (2013) CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 5: 177ra38.
    [7] Grupp SA, Kalos M, Barrett D, et al. (2013) Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. New Engl J Med 368: 1509–1518. doi: 10.1056/NEJMoa1215134
    [8] Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. (2015) T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385: 517–528.
    [9] Ramos CA, Heslop HE, Brenner MK (2016) CAR-T cell therapy for lymphoma. Annu Rev Med 67: 165–183. doi: 10.1146/annurev-med-051914-021702
    [10] Vigneron N (2015) Human tumor antigens and cancer immunotherapy. Biomed Res Int 2015: 948501.
    [11] Linette GP, Stadtmauer EA, Maus MV, et al. (2013) Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood 122: 863–871. doi: 10.1182/blood-2013-03-490565
    [12] Morgan RA, Chinnasamy N, Abatedaga DD, et al. (2013) Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother 36: 133–151. doi: 10.1097/CJI.0b013e3182829903
    [13] Walker A, Johnson R (2016) Commercialization of cellular immunotherapies for cancer. Biochem Soc Trans 44: 329–332. doi: 10.1042/BST20150240
    [14] Koh S, Shimasaki N, Suwanarusk R, et al. (2013) A practical approach to immunotherapy of hepatocellular carcinoma using T cells redirected against hepatitis B virus. Mol Ther Nucl Acids 2: e114. doi: 10.1038/mtna.2013.43
    [15] Bertoletti A, Ferrari C (2016) Adaptive immunity in HBV infection. J Hepatol 64: S71–83. doi: 10.1016/j.jhep.2016.01.026
    [16] Jo J, Lohmann V, Bartenschlager R, et al. (2011) Experimental models to study the immunobiology of hepatitis C virus. J Gen Virol 92: 477–493. doi: 10.1099/vir.0.027987-0
    [17] Pasetto A, Frelin L, Aleman S, et al. (2012) TCR-redirected human T cells inhibit hepatitis C virus replication: hepatotoxic potential is linked to antigen specificity and functional avidity. J Immunol 189: 4510–4519. doi: 10.4049/jimmunol.1201613
    [18] Sautto GA, Wisskirchen K, Clementi N, et al. (2016) Chimeric antigen receptor (CAR)-engineered T cells redirected against hepatitis C virus (HCV) E2 glycoprotein. Gut 65: 512–523. doi: 10.1136/gutjnl-2014-308316
    [19] Zhang Y, Liu YY, Moxley KM, et al. (2010) Transduction of human T cells with a novel T-cell receptor confers anti-HCV reactivity. PLoS Pathog 6: e1001018. doi: 10.1371/journal.ppat.1001018
    [20] Balasiddaiah A, Davanian H, Aleman S, et al. (2017) Hepatitis C virus-specific T cell receptor mRNA-engineered human t cells: impact of antigen specificity on functional properties. J Virol 91: e00010–17.
    [21] Pawlotsky JM, Aghemo A, Back D (2017) EASL recommendations on treatment of hepatitis C 2016. J Hepatol 66: 153–194. doi: 10.1016/j.jhep.2016.09.001
    [22] Krebs K (2013) T cells expressing a chimeric antigen receptor that binds hepatitis B virus envelope proteins control virus replication in mice. Gastroenterology 145: 456–465. doi: 10.1053/j.gastro.2013.04.047
    [23] Gehring AJ, Xue SA, Ho ZZ, et al. (2011) Engineering virus-specific T cells that target HBV infected hepatocytes and hepatocellular carcinoma cell lines. J Hepatol 55: 103–110. doi: 10.1016/j.jhep.2010.10.025
    [24] Banu N, Chia A, Zi ZH, et al. (2014) Building and optimizing a virus-specific T cell receptor library for targeted immunotherapy in viral infections. Sci Rep 4: 4166–4175.
    [25] Perz JF, Armstrong GL, Farrington LA, et al. (2006) The contributions of hepatitis B virus and hepatitis C virus infections to cirrhosis and primary liver cancer worldwide. J Hepatol 45: 529–538. doi: 10.1016/j.jhep.2006.05.013
    [26] Brechot C, Pourcel C, Louise A, et al. (1980) Presence of integrated hepatitis B virus DNA sequences in cellular DNA of human hepatocellular carcinoma. Nature 286: 533–535. doi: 10.1038/286533a0
    [27] Qasim W, Brunetto M, Gehring A, et al. (2015) Immunotherapy of HCC metastases with autologous T cell receptor redirected T cells, targeting HBsAg in a liver transplant patient. J Hepatol 62: 486–491. doi: 10.1016/j.jhep.2014.10.001
    [28] Koh S, Tan A, Li L, et al. (2016) Targeted therapy of hepatitis B virus-related hepatocellular carcinoma: present and future. Diseases 4: 10–16. doi: 10.3390/diseases4010010
    [29] Spear TT, Callender GG, Roszkowski JJ, et al. (2016) TCR gene-modified T cells can efficiently treat established hepatitis C-associated hepatocellular carcinoma tumors. Cancer Immunol Immun 65: 293–304. doi: 10.1007/s00262-016-1800-2
  • Reader Comments
  • © 2017 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(5392) PDF downloads(1115) Cited by(0)

Article outline

Figures and Tables

Figures(1)  /  Tables(1)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog