Page 178 - Read Online
P. 178

Ansari et al. J Cancer Metastasis Treat 2019;5:20  I  http://dx.doi.org/10.20517/2394-4722.2018.68                           Page 9 of 14

               SRC-mediated activation of PIK3/AKT/mTOR signaling, we treated BBM1 cells with 100 nmol/L of AC93253
               for 48 h, followed by Western blot analysis of the total protein extract. Our analysis showed that inhibition
               of SRC leads to inhibition of PIK3CA and mTOR phosphorylation [Figure 5D]. As our analysis indicated
               that SRC is an upstream regulator of ERBB2 and the PIK3-AKT/mTOR pathway in BBM1 cells and that
               inhibition of both SCR and ERBB2 induces BBM1 cell death, we analyzed the combined effect of SRC and
               ERBB2 inhibitors on BBM1 cell viability. We treat the cells with 50 nmol/L of AC93253 and 50 nmol/L of
               Lapatinib, separately and in combination, for 48 h. Our analysis showed robust inhibition of BBM1 cells
               in the presence of either inhibitor alone, however, the effect was greatest in presence of both [Figure 5E].
               Similar to combinatorial SRC/ERBB2 treatment, concurrent inhibition of ERBB2 and PIK3CA showed a
               significant additive inhibitory effect on BBM1 cell survival [Figure 5E].

               We also evaluated the combinatorial treatment of ERBB2 and SRC inhibitors with a GABA agonist.
               BBM1 cells were treated with either 100 nmol/L of Isoguvacine alone or in combination with Lapatinib
               or AC93253 for 48 h. Cell viability and apoptosis analyses showed that the combination of Isoguvacine
               with both Lapatinib and AC93253 induced higher inhibition of cell viability compared to independent
               treatments [Figure 6A and B]. Our analysis showed that the expression of GABA receptors and subunits was
               significantly higher in nBrain compared to PT, MT, and nBreast [Figure 6C, Supplementary Figure 5].


               DISCUSSION
               Brain metastases are the most common and complicated central nervous system disease in adults. The
               incidence of brain metastases is increasing due to both improved diagnosis and increased cancer patient
               survival through advanced systemic treatments. Outcomes of patients remain disappointing and treatment
               options are limited, usually involving multimodality approaches of radiation and palliative chemotherapy.
               Brain metastases represent an unmet clinical challenge in caring for solid tumor, especially in breast cancer,
               where the incidence of brain metastases are frequent and that result in impaired quality of life.


               BBMs are common in patients with the HER2-positive and TNBC breast cancer subtypes, and the natural
               course of BBM is strongly influenced by the biology of the primary tumor subtype. Although the biology of
               BBM according to tumor subtypes is still poorly understood, recent breakthroughs have been achieved in
               the identification of specific mediators of BBM and in the development of preclinical models for therapeutic
               studies. However, the use of established cell lines and comparative analysis of unrelated tissue samples
               often identify factors that eventually fail to represent as a key regulator in preclinical and clinical stage. To
               overcome this issue we obtained both primary and metastasis tissue from same individual patients (PT1/
               MT2 and PT2/MT2). Due to the inherent difficulty in the procurement of such tissue pairs we have used
               limited numbers of tissue samples. Our RNA-seq analysis revealed that ERBB2-mediated activation of
               PIK3CA and its downstream AKT/mTOR pathway plays a critical role in the survival and proliferation of
               breast to brain metastatic cells. Inhibition of ERBB2 or its downstream targets suppresses BBM cell viability
               to different degrees. The critical function of ERBB2 in HER2+ primary and metastatic breast cancer has been
               well established, and the ERBB2 inhibitor Lapatinib and trastuzumab have been used as chemotherapeutic
               agents targeting HER2+ breast cancer [25-28] . In addition, our earlier studies revealed the contribution of neural
               factors, such as the astrocyte-secreted brain-derived neurotrophic factor, in the activation of HER2-TrkB
                                                                          [25]
               signaling that leads to AKT1 activation and BBM1 cell proliferation . Several studies have also reported
               mutations and/or copy number changes in PIK3CA in hormone receptor-positive breast cancers [29-31] . Indeed,
               targeting PIK3-AKT/mTOR signaling to inhibit breast cancer has been evaluated extensively, and there is
               phase I to III preclinical trial data demonstrating that inhibition of this pathway leads to regression of solid
               tumors and breast cancer [32-37] .

               Recent gene expression analyses of BBM samples identifed cyclooxygenase (COX)-2, EGFR ligands, and a
                                                                           [38]
               sialyltransferase, as mediators of cancer cell passage through the BBB . In contrast to COX-2 and EGFR,
   173   174   175   176   177   178   179   180   181   182   183