Page 210 - Read Online
P. 210

Page 10 of 16              Landi et al. Neuroimmunol Neuroinflammation 2018;5:29  I  http://dx.doi.org/10.20517/2347-8659.2018.35


               Table 3. Notable past and ongoing active immunization trials for pediatric brain tumors
                Trial/therapy                                Description                       Reference
                Monocyte-derived DCs loaded   Phase I trial evaluating DCs pulsed with tumor RNA in children with brain tumors  [93]
                with tumor RNA
                Monocyte-derived DC loaded   Phase I trial evaluating DCs pulsed with whole tumor lysate in children and adults   [94]
                with whole tumor lysate  with relapsed malignant glioma
                Monocyte-derived DC loaded   Phase I trial with DC pulsed with tumor lysate for children with newly diagnosed or   [95]
                with whole tumor lysate  recurrent high-grade gliomas
                Monocyte-derived DC loaded   Phase I trial with postoperative DC loaded with tumor lysate for children and adults   [96]
                with whole tumor lysate  with recurrent GBM
                Peptide vaccine targeting   Phase I trial evaluating peptide vaccine targeting H3.K27M neoantigen for HLA-A2+   NCT02960230
                H3.K27M                children with H3K27M mutated glioma                     [101]
                Peptide vaccine targeting CMV   Phase I trial evaluating peptide vaccine targeting CMV pp65 and glycoprotein B for   NCT03299309
                epitopes pp65 and glycoprotein B   children with recurrent malignant glioma and medulloblastoma  [102]
                Peptide vaccine targeting glioma   Phase I peptide trial using glioma-associated antigens for HLA-A2+ children with   NCT01130077
                antigens               malignant brainstem and non-brainstem gliomas, including low-grade glioma  [103]

               are genetically modified to express the CAR molecule, which is designed to bind a tumor-restricted antigen
               and cause tumor cell death.

               The CD19 CAR, which is effective against B-lineage lymphoid malignancies [111,112] , is FDA approved and
               induces remission in most patients with relapsed CD19-positive leukemia. CAR T cells targeting HER2 [113] ,
               IL13rα2 [114] , EGFRvIII [115] , and EphA2 [116]  have been used to treat adults with GBM. A trial involving CMV-
               specific cytotoxic T lymphocytes expressing a HER2 CAR treated seven children with GBM. There were
               no serious adverse events or instances of cytokine release syndrome, and at least one child had a partial
               response [113] . Transient responses following adoptive CAR T cell therapy are not infrequent, but almost all
               patients ultimately suffer disease progression.

               There are multiple reasons the success of the CD19 CAR for B-lymphoblastic leukemia has not been dupli-
               cated by CAR T cells for brain tumors. The CD19 CAR targets an antigen that is ubiquitous and expressed
               solely on tumor cells or non-essential B cells without a strongly immunosuppressive tumor bed. Additional-
               ly, the CD19 single chain variable fragment (scFv) that guides the CAR T cell imparts an optimal activation
               profile and supports continued T cell killing [117] . This characteristic of the scFv is a key and unique distinc-
               tion in this T cell product. ScFvs for other CAR T cells cause tonic signaling, which can cause T cell exhaus-
               tion and limits anti-tumor activity in patients following adoptive transfer [117] .

               Antigen escape, tumor heterogeneity, and a harshly immunosuppressive immune microenvironment also
               contribute to treatment failure by CAR T cells. In a recently completed phase I trial for adults with recurrent
               GBM, EGFRvIII CAR T cells reliably reached the tumor bed following peripheral administration. However,
               ex vivo analyses from resected tumor showed dramatic adaptive resistance, with markedly increased PD-
               L1 expression and an influx of regulatory T cells, as well as decreased expression of the targeted EGFRvIII
               antigen [115] .


               CONCLUSIONS
               Immunotherapy holds tremendous promise for improving outcomes for children with brain tumors. While
               checkpoint inhibitors and CAR T cells are well suited for hypermutated, immunologically hot tumors and
               B-cell malignancies, respectively, these modalities are less of a fit for pediatric brain tumors. Rather, immu-
               notherapy approaches that induce inflammation and an innate immune response may be a better starting
               point, on which checkpoint agents and other T cell-directed agents can build.

               While we are optimistic about immunotherapy in pediatric neuro-oncology, it is important to recognize that
               conventional chemotherapy and radiation will likely retain a role in treatment, particularly as both of these
   205   206   207   208   209   210   211   212   213   214   215