Page 64 - Read Online
P. 64

Page 60                          Hunt et al. Extracell Vesicles Circ Nucleic Acids 2020;1:57-62  I  http://dx.doi.org/10.20517/evcna.2020.04

               including breast cancer and hepatocellular carcinoma, among others [20-25] . Additionally, several ongoing clinical
               trials are testing anti-adrenergic medications as an adjuvant or stand-alone therapy in prostate and pancreatic
                                                                                                        [18]
               cancers (NCT03152786, NCT02944201, NCT03838029, NCT04245644). Within this study, Amit et al.
               found that Carvedilol, which non-selectively blocks α1, β1, and β2 adrenergic receptors, dramatically
               decreased tumor growth and proliferation rates when compared to tumors within vehicle-treated mice
                                                      null
               harboring patient-derived xenografted p53  OCSCC tumors. Supporting the use of anti-adrenergic
               therapies in humans, this group found that the level of TH staining in human OCSCC samples was an
               independent predictor of increased tumor recurrence and decreased patient survival. Thus, this work
               argues for an increased focus on anti-adrenergic approaches for treating OCSCC as well as other cancers.

                                                        [1]
                                           [18]
               Similar to the findings by Amit et al , Magnon et al.  found an increased density of TH-positive adrenergic neurons
               within the tumor microenvironments of patients with high-risk prostate adenocarcinomas . This group showed
                                                                                        [1]
               that the number of adrenergic neurons within the tumor microenvironment was an independent predictor of
               tumor recurrence. They also found a similar relationship between tumor aggression and the amount of
               parasympathetic vesicular acetylcholine transporter (VAChT)-positive staining within the tumor body.
               However, they were not able to determine whether this increase in autonomic innervation was due to
               neurogenesis, neuritogenesis, neuron-type switching, or some other mechanism. Additionally, they were
               not able to determine if the tumor was driving this increase in autonomic neurite number, and by what
               means these aggressive tumors were communicating with local neurons. In contrast, the findings presented
                           [18]
               by Amit et al.  shed light on possible mechanisms of communication between prostate tumors and local
               innervating autonomic neurons. These experiments add to the already rich literature describing the role
               that EVs play in regulating the tumor microenvironment and cancer metastasis. Future work in prostate
               adenocarcinoma should examine the EVs released by these tumors, and the miRNA contents that may be
               manipulating local neurons. A recent report systematically demonstrated the importance of EV contents
                                                               [26]
               in the development and growth of a variety of cancers . Though small RNA messengers and regulators
               were found to be important in mediating tumor growth in this study as well, this group provided ample
               evidence supporting damage-associated molecular patterns (DAMPs) and other cancer-associated proteins
               as essential mediators of neuron-tumor trophic interactions. These findings point to the possibility that
               protein messengers are important in establishing and maintaining the trophic relationships between
               prostate adenocarcinomas and local autonomic fibers, as well as p53  OCSCC tumors and local sensory/
                                                                          null
               adrenergic neurons.
                                                                                            [18]
               Though the mechanisms governing neuron-tumor relationships described by Amit et al.  were worked
               out between OCSCC tumors and sensory/adrenergic neurons of the oral cavity, many of these principles
               might be generalizable to other tumor types and neuronal subtypes. Already, increased neuritogenesis and
               sensory-autonomic neurotype switching has been reported in pancreatic nerves in the context of pancreatic
                     [27]
               cancer . However, the findings presented here also differ from those published in previous work. In
                                                   [1]
               contrast to the findings of Magnon et al. , Amit et al.  found no increase in parasympathetic VAChT-
                                                               [18]
                                    null
               positive neurites in p53  OCSCC tumors. Thus, it is possible that for each tumor type and each region
               of local autonomic fibers, a balance of sympathetic and parasympathetic neurons guides tumor growth
               and survival. Future work will need to examine these balancing forces within each tumor type and the
               corresponding microenvironment.

               This work describes novel mechanisms of EV-mediated regulation of tumor activity, which highlights new
               hypotheses regarding the biology underlying these mechanisms. Differential delivery of miR-34a vs. miR-21
               and miR-324 to local neurons could be the result of differential loading of miRNAs into EVs. Alternatively,
               the differing contents of these cancer-derived EVs could be dictated strictly by expression changes within
                            WT
                  null
                                                                                             WT
               p53  and p53  cancer cells. Specific analysis of transcriptional changes in p53  and p53  cancer cells,
                                                                                    null
               combined with detailed profiling of EV contents will allow for a deeper understanding of the mechanisms
                                                                                                        null
               governing this described specificity of EV contents. In tandem with this, specific subpopulations of p53
   59   60   61   62   63   64   65   66   67   68   69