Page 25 - Read Online
P. 25

Page 6 of 15                                            Feng et al. Hepatoma Res 2021;7:3  I  http://dx.doi.org/10.20517/2394-5079.2020.107

                 NCT04039607 PD-1         Recruiting, Phase III  Nivolumab  1,084 OS, 4 years
                             CTLA-4                       Ipilimumab
                             Raf-1                        Sorafenib
                             VEGFR/FGFR                   lenvatinib
                 NCT04472767 PD-1         Not yet recruiting,   Nivolumab  35  Percentage of Progression-free
                             CTLA-4       Phase II        Ipilimumab         Survival, 6 Months;
                             Multikinase                  Cabozantinib       Complete Response Rate, 1 year
                             Embolotherapy                Transarterial
                                                          Chemoembolization
                 NCT04050462 PD-1         Not yet recruiting,   Nivolumab   74  ORR, 6 years
                             Multikinase  Phase II        Cabiralizumab
                             IL-8                         BMS-986253
                 NCT03071094 Oncolytic therapy  Active, not recruiting,  Pexastimogene   30  DLTs, 4Weeks;
                             PD-1         Phase I/II      Devacirepvec;      ORR, 6 months
                                                          Nivolumab
                 NCT03897543 PD-1         Recruiting Phase 1   Nivolumab   48  AEs, 1 year
                             INK T cells Agonist Phase 2  ABX196
               Most data were obtained from findings from www.clinicaltrials.gov using the search terms “hepatocellular carcinoma” and “Immune
               Checkpoint Inhibitors”. AEs: rate of adverse events; ORR: objective response rate; MTD: maximum tolerated dose; DLTs: dose-limiting
               toxicities; OS: overall survival; TTTP: time to TACE progression

               macrophage colony-stimulating factor (GM-CSF) into the oncolytic virus sequence, GM-CSF recruits
                                                                                                     [53]
               myeloid cells in the periphery to enhance the immune response in the tumor microenvironment . So
               far, preclinical studies for HCC oncolytic virus therapy have been very encouraging. We have compiled
               preclinical studies on HCC oncolytic therapy for the past ten years, as shown in Table 2.

               Although many preclinical research attempts have been made in oncolytic therapy in recent years, there
               are still very few programs that have entered the clinical stage. At present, the only HCC oncolytic virus
               entering clinical research is JX-549, with VV as an engineered vector. VV has the stability and efficiency
               of intravenous administration, is widely used in the safety of live vaccines, has the advantages of immune-
               inducing activity and better editability, and has become a carrier of various engineered tumor-melting
               viruses [73-75] . The thymidine kinase gene (TK) gene of JX-594 (also known as PexaVec; Jennerex Inc.) was
               deleted to make it more specific for cancer cell infection. In addition, hGM-CSF and β-galactosidase were
               inserted to enhance its immunostimulatory activity and replication capabilities [73,76,77] . JX-594 showed
               complete tumor response and systemic efficacy in a phase I clinical study . In the phase II trial, low-
                                                                                [78]
                                                                                  [79]
               dose JX-594 has significant anti-cancer effect and immune activation ability , but this requires earlier
               interventional therapy . Currently, a large-scale 600-person multicenter Phase 3 trial is still in progress
                                  [80]
               (NCT02562755). More clinical studies of HCC oncovirus are shown in Table 3.

               HCC VACCINE
               Tumor vaccine is a treatment program to increase the specificity of tumor antigens, mainly antigen peptide
               vaccines and DCs vaccines, which are used to stimulate specific immune responses. The clinical trials of
               therapeutic vaccines for HCC are summarized in Table 4. At present, there are relatively few registered
               clinical trials for DCs vaccines in HCC, partly because of the unsatisfactory results of previous clinical trials
                             [86]
               of such vaccines . On the other hand, the tumor heterogeneity of HCC also limits the development of a
               single antigen peptide or DCs vaccine. With the development of large-scale DNA sequencing technology,
               patient-specific multi-target peptide or DCs vaccine is still a promising strategy for the treatment of HCC.
               DC, as professional antigen-presenting cells (APC), recognize, process and present TAA. Allogeneic DC
               vaccines can provide T cells with antigens and co-stimulatory molecules needed for immune response.
               In short, DCs are mobilized from peripheral blood and their expansion is stimulated with GM-CSF to
               produce DCs for reinfusion. Prior to this, DC needs to be exposed to TAA to trigger the specificity of the
               vaccine . DCs can be transduced with DNA or RNA encoding known TAA, or directly co-cultured with
                     [87]
                                [88]
               patient tumor lysate . Phase I clinical studies have shown that the allogeneic DCs vaccine can produce a
                                                          [89]
               specific immune response in 73% of HCC patients .
   20   21   22   23   24   25   26   27   28   29   30