Page 29 - Read Online
P. 29

Li et al. Hepatoma Res 2020;6:15  I  http://dx.doi.org/10.20517/2394-5079.2019.34                                                      Page 7 of 13


               stimulatory molecule ICOSL-41BB promotes CAR-T cell proliferation and tumor rejection [130] . Besides
               GPC3, MUC-1, EpCAM, AFP, and CEA might be potential targets of CAR T cells for HCC treatment,
               which have been registered for clinical trials on the applicability of CAR-T therapy as a treatment strategy
               for HCC [131] . Moreover, these classical tumor-associated antigens and ligands for receptors expressed on T
               cells also act as the targets for HCC recognition. For instance, NKG2D-based CAR-T cells could potently
                                +
               eliminate NKG2DL  HCC cells [132] . A CD147-targeted inducible CAR-T cell system has been developed
               for HCC treatment [133] . Although clinical trials of CAR-T therapy against HCC have not been completed,
               CAR-T therapy might provide effective therapeutic modalities for HCC treatment. Nonetheless, to date,
               the therapeutic efficacy of CAR-T cells remains limited owing to the lack of cancer-specific targets, weak
               expansion, poor infiltration, and induced exhaustion of CAR-T cells. Hence, smarter optimization strategies
               and more clinical trials are required for the confirmation and improvement of clinical outcomes of CAR-T
               cells in HCC treatment.

               T cell receptor-genetically engineered T cell therapy
               The success of T cell receptor-genetically engineered T (TCR-T) cells in melanoma treatment has encouraged
               the use of TCR-T cells in HCC treatment. Autologous T cells forced to express an HBV-specific TCR
                               +
               recognized HBsAg  HCC cells and decreased HBsAg levels in a patient who underwent liver transplant [134] .
               T cells genetically engineered with HCV NS3:1406-1415-reactive TCR recognized the naturally processed
                                                 +
               antigen and led to suppression of HCV  HCC in vivo [135] . T cells genetically engineered with AFP-specific
                                                      +
               TCR specifically recognized and killed AFP  HepG2 cells, both in vitro and in vivo [136] . Although there
               remain many challenges such as off-target cross-reactivity and low TCR affinity that need to be overcome
               before successful translation into clinical practice [137,138] , increasing findings suggest that TCR-T therapy
               might be an attractive alternative immunotherapeutic modality for HCC treatment.

               DC-vaccines adjuvant immunotherapy
               Briefly, DCs are professional antigen-presenting cells with the capacity to prime antigen-specific T-cell
               immunity. DC vaccines are recognized as promising agents for activating T cells to eliminate cancer cells;
               their role and functions have been evaluated in some malignancies in clinical trials, including HCC. A phase
               II study using intravenous vaccination with DCs pulsed with HepG2 lysate was found to be safe and showed
               evidence of anticancer efficacy in some patients with advanced HCC [139] . Another phase I/II study reported
               that vaccination with DCs pulsed with AFP peptides induced strong T-cell immunity against AFP but no
               clinical responses in HCC patients [140] . Other phase I/IIa studies also reported that subcutaneous vaccination
               with DC pulsed with multiple antigens such as AFP, glypican-3 (GPC-3), and melanoma-associated antigen
               1 (MAGE-1) enhanced anticancer immunity and prolonged time-to-recurrence and recurrence-free survival
               in HCC patients [141-143] . Interestingly, Lu et al. [144]  showed that exosomes derived from AFP-expressing DCs
               elicited potent anticancer immune responses and cancer regression in HCC mice, thus providing a novel
               option for vaccine-based immunotherapy of HCC. Pang et al. [145]  reported that DCs fused with cancer stem
               cells could efficiently stimulate T lymphocytes to generate specific CD8 T cells against cancer stem cells.
               Collectively, several studies indicate that DC vaccine-based adjuvant therapy enhances anticancer immunity
               and improves the survival of patients with HCC. Nonetheless, further improvements such as specific
               immunogenic neoantigens for HCC, safe and feasible DC source, potent adjuvant, and access to vaccination
               are required for future success of DC-based HCC immunotherapy.

               Combination therapy of immune checkpoint blockade and adoptive cell transfer
               The existence of cancer immunosuppressive microenvironment limits the effector function of adoptive
               immune cells. Therefore, it is reasonable to improve the cancer immunosuppressive microenvironment to
               enhance the curative efficacy of adoptive immune cells on HCC. Kodumudi et al. [146]  reported that adoptive
               transfer of tumor infiltrating lymphocytes from tumors with anti-PD-L1 antibody treatment led to a
               significant delay in tumor growth, suggesting that pretreatment with immune checkpoint blockade could be
   24   25   26   27   28   29   30   31   32   33   34