Page 380 - Read Online
P. 380

Page 10 of 18              Yelton et al. Neuroimmunol Neuroinflammation 2018;5:46  I  http://dx.doi.org/10.20517/2347-8659.2018.58


               p21Waf1/Cip1 and the pro-apoptotic protein Bad and the decreased expression of the anti-apoptotic proteins
                              [73]
               Bcl-xL and Bcl-2) . Finally, of the two miscellaneous HDAC inhibitors, DATS has been shown to cause
               upregulation of the cell cycle inhibitor p21Waf1/Cip1 and the tumor suppressor p53 in order to cause cell
               cycle arrest in glioblastoma cells and is unique in that it is derived from garlic and demonstrates less toxicity
                                                      [74]
               to normal cells than other HDAC inhibitors . Tubacin, the other miscellaneous HDAC inhibitor, is a
                                [75]
               specific for HDAC6  and it has been proposed to be useful because HDAC6 is known to be increased in
               certain high-grade gliomas. All the above HDAC inhibitors have shown considerable promise for growth
               inhibition in glioblastoma, but only a few of these agents have made it into the clinical trials as of now.


               Only a few selected HDAC inhibitors that have shown promise in the pre-clinical realm translate so
               seamlessly over to show efficacy in the clinical realm. Vorinostat, romidepsin, and valproic acid are
               particularly notable to have seen translational promise in the preclinical realm as well as in the clinical
               realm. Vorinostat as a monotherapy progressed through multiple Phase I and Phase II trials, with the
               results of one Phase II trial indicating that it was well tolerated in recurrent glioblastoma patients,
               and its efficacy was seen to extend life by a few months in a subpopulation of those with recurrent
                          [76]
               glioblastoma . Romidepsin also went through both Phase I and Phase II trials but had disappointing
               outcomes in progression free survival with the conclusion that although the drug demonstrated success
               in the preclinical arena, when used in clinics an inadequate amount of the drug reached the actual tumor
                         [77]
               in the CNS . However, this agent showed success when used in combination therapies. Valproic acid
               similarly showed success in clinical trials, but only when used in combination therapies and not when
                                   [78]
               used as a monotherapy . In fact, many other HDAC inhibitors listed in Table 2 in various trials for use in
               glioblastoma are in combination therapies and may yet show results when combined with the standard-of-
               care agents. However, HDAC inhibitors when used as monotherapy have yet to yield the progression free
               survival results that the preclinical mechanistic evidence would suggest, with an exception to vorinostat (and
               even then, only modestly so). To understand the full picture of these promising new agents, one must look at
               both the preclinical and the clinical data housed in trials. Unfortunately, there seems to be a wealth of new
               mechanisms to be revealed and understood as to the biological pathways these agents are inhibiting. A more
               profound understanding of glioblastoma pathogenesis and the associated aberrant pathways inhibited by
               these agents is essential to translate the benefits from the preclinical bench to the clinical arena.


               HDAC INHIBITORS FOR ENHANCING ANTITUMOR IMMUNITY IN GLIOBLASTOMA
               While there have been a variety of preclinical studies regarding the effects of HDAC inhibitors specifically in
               glioblastoma, one of the most interesting effects is alteration of the tumor itself to increase tumor susceptibility
               to antitumor immune attack. Many cells of the immune system act as surveillance cells, effectively patrolling
                                                                     [79]
               the body to eliminate neoplastic cells as soon as they are found . However, many tumors are notorious for
               down regulating these markers on their surfaces, effectively “hiding” from the immune system to evade
               elimination and continue their unfettered growth. While there are many important cell types (microglia, T
               cells, etc.) involved in the surveillance of the body’s somatic tissues for signs of pathological changes, one of
               the cell types most important to the preclinical mechanism of HDAC inhibitors and antitumor immunity
               are NK cells. These cells act to “check” or surveille surface proteins displayed on the exterior of many cells,
               checking these cells for signs of stress, infection, or neoplastic change. If an NK cell finds a cell within the
               body that has undergone one of these pathological changes, the NK cell releases cytotoxic granules containing
               toxic compounds known as perforins and granzymes, which act synergistically to induce apoptosis in the
               target cell. These NK cells have been described in the literature to be the agents that lyse GBM cells as they are
               recognized during their surveillance, with HDAC inhibitors playing a role in upregulating the surface markers
               that help to mark these malignant cells as a target for elimination [19,22] .

               The NK cells possess a constitutively expressed receptor on their surface known as natural killer group 2D
                                                                          [80]
               (NKG2D) that is essential for recognition of abnormal human tissues . This receptor recognizes a ligand
   375   376   377   378   379   380   381   382   383   384   385