Page 54 - Read Online
P. 54

Sale et al. Cancer Drug Resist 2019;2:365-80  I  http://dx.doi.org/10.20517/cdr.2019.14                                                    Page 371

               activation arising from BRAF V600E  mutation, sufficient ERK1/2-responsive tumour suppressive mechanisms
               remain intact to drive proliferative arrest or cell death following the hyperactivation of ERK1/2 that occurs
               upon MEKi withdrawal.



               BRAF  V600E -MUTANT CRC CELLS EVOLVE TO REINSTATE THE SAME OPTIMAL LEVEL OF ERK1/2
               ACTIVITY REGARDLESS OF WHETHER ERK1/2 ARE INHIBITED OR HYPERACTIVATED
               Cell cycle phase profile, EdU incorporation and C6244-R fitness vs. COLO205 were all optimal and/or
               maximal when C6244-R cells were maintained in 1 µmol/L selumetinib, the concentration in which they
               were selected and at which ERK1/2 phosphorylation matched that in parental cells [9,11] . However, these
               observations were not unique to selumetinib. C6244-R cells were cross-resistant to the clinically approved
               MEKis cobimetinib and trametinib, and to the ERK1/2 inhibitor SCH772984, and in each case proliferated
               optimally at inhibitor concentrations that imposed ERK1/2 or RSK phosphorylation at close to parental
               levels . These effects were recapitulated in vivo: C6244-R tumours grew better in mice dosed with 10 mg/kg
                    [11]
               selumetinib compared to those dosed with vehicle only or 25 mg/kg selumetinib . Thus, although achieving
                                                                                  [11]
               a steady-state concentration of selumetinib in mice akin to that in vitro is not possible, C6244-R cells were
               addicted to a tight window or “sweet-spot” of ERK1/2 pathway output optimal for proliferation both in vitro
               and in vivo.


               This evolutionary pressure to restore ERK1/2 activity to an optimal “sweet-spot” was strikingly exemplified
               in an experiment in which separate COLO205 cell lines with resistance to a range of distinct selumetinib
               concentrations were established . The higher the concentration of selumetinib, the longer the cells took to
                                          [11]
               evolve resistance and proliferate normally. Remarkably, however, all resistant cells proliferated optimally in
               the presence of the selumetinib concentration to which they had adapted, and at this concentration exhibited
               equivalent p-ERK1/2 levels as parental COLO205 cells . This was enabled by a progressive increase in
                                                               [11]
               BRAF expression: cells adapted to higher concentrations of selumetinib through higher BRAF expression
               that restored parental ERK1/2 activity and a normal cell cycle profile in the respective drug concentration .
                                                                                                        [11]
               However, in the absence of selumetinib ERK1/2 were hyperactivated in proportion to the degree of BRAF
               expression. Consequently COLO205 cells with resistance to higher concentrations of selumetinib exhibited
               greater ERK1/2 activation in the absence of selumetinib and underwent G1 cell cycle arrest .
                                                                                            [11]
               Thus regardless of whether ERK1/2 were inhibited in parental COLO205 cells, or ERK1/2 were hyperactivated
               following MEKi withdrawal from C6244-R cells, cells evolved accordingly to increase or decrease BRAF copy
               number and BRAF expression to a level that restored ERK1/2 activity and pathway output back to parental
               levels. Mass spectrometry was used to define this optimal “sweet-spot” of ERK1/2 activation; quantifying
               ERK1/2 activation loop dual pT-E-pY phosphorylation revealed that COLO205 cells, and C6244-R cells
               maintained in selumetinib, proliferated with just 2%-3% of the total ERK1/2 pool active, and cellular p-ERK1
               and p-ERK2 concentrations of ~2 nmol/L and 3 nmol/L, respectively . MEKi withdrawal increased the
                                                                           [11]
               stoichiometry of phosphorylated ERK1/2 to ~20%-30%, and cellular p-ERK1 and p-ERK2 concentrations to
               ~10 and 20 nmol/L, respectively . HT29 cells, and HT6244-R cells in selumetinib, also exhibited a ~2%-5%
                                          [11]
               stoichiometry of ERK1/2 phosphorylation and cellular p-ERK1 and p-ERK2 concentrations of < 2 nmol/L and
               < 5 nmol/L, respectively . This suggests, even in tumour cells with BRAF V600E  mutation, there is substantial
                                   [11]
               spare capacity within the ERK1/2 pathway under basal conditions.


               MEK1/2 INHIBITOR WITHDRAWAL FROM KRAS-MUTANT CRC CELLS WITH ACQUIRED MEK1/2
               INHIBITOR RESISTANCE PROMOTES EMT AND CHEMORESISTANCE
               HCT116 CRC cells harbour a KRAS G13D  mutation and acquired resistance to selumetinib through KRAS G38A
               gene amplification and striking upregulation of KRAS protein  [Figure 4]. As with the BRAF V600E -amplified
                                                                    [9]
   49   50   51   52   53   54   55   56   57   58   59