Page 364 - Read Online
P. 364

Rizzieri et al. J Cancer Metastasis Treat 2019;5:26  I  http://dx.doi.org/10.20517/2394-4722.2019.05                         Page 7 of 16

               contributor of carbon substrates to the TCA cycle whereas glucose is an important contributor of carbon
                                                                    [37]
               substrates for the formation of lactate in clonal plasma cells . This was also supported by the finding
               that clonal bone marrow plasma cells in MM patients have higher glutamine uptake compared with the
               remainder of bone marrow mononuclear cells. Additionally, 2-hydroxyglutarate (2-HG) - an oncometabolite
               of glutaminolysis - is significantly increased in the bone marrow supernatant of MM patients compared to
               that of MGUS patients. Similarly, levels of 2-HG in the bone marrow supernatant or in the peripheral blood
               plasma of patients with smoldering multiple myeloma was associated with higher risk of progression to
                                                                                            [37]
               MM, and correlates with the percentage of bone marrow plasma cells expressing c-MYC . These results
               demonstrate the utility of measuring 2-HG levels in BM supernatant or peripheral blood plasma of SMM
               patients as a potential biomarker for disease progression and for identification of patients who may benefit
               from early therapeutic intervention.

                                                                  [38]
               To characterize the lipid profile of MM cells, Hossen et al.  sorted single MM and normal plasma cells
               using flow cytometry and performed matrix-assisted laser desorption/ionization-imaging mass spectrometry.
               They found that PC (16:0/20:4) was significantly decreased in MM cells compared to normal plasma cells,
               which is likely due to the decrease of its integrated fatty acids or to increased metabolism of C .
                                                                                             16:0
                                                                                                        [39]
               The development of drug resistance is also associated with metabolic alterations in MM. Zub et al.
               performed global proteomic and transcriptomic profiling on melphalan sensitive and resistant RPMI8226
               MM cell lines. Glycolysis was found to be the most significantly altered pathway. Gluconeogenesis, glutaryl-
               CoA degradation, isoleucine and tryptophan degradation, TCA cycle, ketolysis and ketogenesis were also
               significantly altered when MM cells develop resistance to melphalan. Specifically, the glycolytic and pentose
               phosphate pathway enzymes were up-regulated in the melphalan resistant cells, whereas the TCA cycle and
               ETC proteins were down-regulated. Additionally, the melphalan-resistant cells displayed increased tolerance
               to overall oxidative stress, but were sensitive to mitochondrial electron transport inhibitors. It was further
               demonstrated that lactate accumulation, Interleukin-8 and vascular endothelial growth factor (VEGF)
               signaling, as well as aldo-keto reductase played a role in the development of drug resistance to melphalan in
                           [39]
               MM cell lines . Similarly, in MM cell lines resistant to proteasome inhibitors (bortezomib and carfilzomib),
                                                                                               [40]
               complex proteomic changes, particularly in redox and energy metabolism were observed . Proteins
               involved in metabolic regulation, redox homeostasis, and protein folding and destruction were upregulated
               in proteasome inhibitor-resistant MM cell lines, which exhibited metabolic adaptations that favored the
                                                           [40]
               generation of reducing equivalents such as NADPH . The glucose metabolism was rewired in bortezomib-
               resistant MM cells, leading to higher activity of both the pentose phosphate pathway and serine synthesis
                                                                               [41]
               pathway and an increased anti-oxidant capacity in bortezomib-resistant cells . Interestingly, the expression
               of 3-phosphoglycerate dehydrogenase (PHGDH), which catalyzes the rate-limiting step of serine synthesis,
               was up-regulated in bortezomib-resistant MM cells. Consistent with increased serine synthesis observed
               in bortezomib-resistant MM cells, serine starvation enhances the cytotoxicity of bortezomib. These data
               suggest that interfering with serine metabolism could be a novel strategy to improve bortezomib therapy and
                                                                      [41]
               PHGDH could be a potential biomarker for bortezomib resistance .
                                                                                              [42]
               To identify therapeutic targets for the prevention and treatment of drug resistance, Maiso et al.  performed
               metabolic profiling of multiple myeloma cells in normoxic and hypoxic conditions. It was found that in
               hypoxic conditions, intermediates of the TCA cycle and the ETC were reduced whereas intermediates of
               glycolysis were elevated. Specifically, glucose 6-phosphate, fructose 6-phosphate, fructose 1,6-bisphosphate,
               pyruvate, and lactate were significantly increased in hypoxic cells. Gene expression profiling further
               demonstrated that the main pathways altered by hypoxic culture were those involved in glucose metabolism
               and the TCA cycle including key metabolic genes such as hexokinase 2 (HK2) and lactate dehydrogenase
               A (LDHA). Interestingly, bortezomib treatment inhibited HK2 activity but not lactate activity, suggesting
               LDHA may play a role in modulating drug resistance of MM cells in hypoxic conditions.
   359   360   361   362   363   364   365   366   367   368   369