Page 31 - Read Online
P. 31

Page 982                                                  Lee et al. Cancer Drug Resist 2020;3:980-91  I  http://dx.doi.org/10.20517/cdr.2020.73

               Table 1. Mutational status and TNBC subgroup of the cells line used in this work
                Cell line                                 Mutations                               Subgroup
                HCC1806    TCF12-A482V                                                             Basal A
                MDA-MB-157  FAT4-L4468P; MSH6-R644S                                                Basal B
                MDA-MB-231  BRAF-G4646V; CD79A-C106Y; KRAS-G13D; NF2-E231*; PBRM1-I228V; PDGFRA-Y172F; TP53-R280K  Basal B
                MDA-MB-468  CACNA1D-E953D; TP53-R273H, PTEN V85_splice                             Basal A
                HCC1937    TP53-R308; PTEN deletion; BRCA-5396insC                                 Basal A
                MX-1       BRCA1-3363delGAAA                                                       Basal B
               TNBC: triple negative breast cancer


               METHODS
               Cell culture
               The TNBC cell lines HCC1806, HCC1937, MDA-MB-157 (MDA-157), MDA-MB-231 (MDA-231), and
               MDA-MB-468 (MDA-468) were purchased from the American Type Culture Collection (Manassas, VA;
               CRL-2335, CRL-2336, HTB-24, HTB-26, and HTB-132). MX-1 was purchased from the NCI repository. All
               cell lines were purchased within the previous 24 months and passaged < 15 times for all experiments [Table 1].
               Cells were tested biweekly for mycoplasma contamination (MycoAlert, Lonza, Basel, Switzerland). MX-1,
               MDA-157, MDA-231, and MDA-468 were grown in Dulbecco Modified Eagle Medium (DMEM High
               Glucose with GlutaMAX, Life Technologies, Carlsbad, CA) and supplemented with 1% sodium pyruvate
               (Life Technologies) and 10% fetal bovine serum (FBS) (Premium Select, Atlantic Biologicals, Miami, FL).
               HCC1937 and HCC1806 were grown in RPMI supplemented with 10% FBS. Cells were maintained in a
               humidified 37 °C incubator with 5% carbon dioxide.

               Cytotoxicity
               Cytotoxicity of monolayer and 3D cultures were determined with a cell viability assay, CellTiter-Glo and
               CellTiter-Glo 3D (Promega, Madison, WI). For monolayer experiments, HCC1806, HCC1937, MDA-157,
               MDA-468, and MX-1 were plated at 5000 cells per well in clear-bottomed white 96-well plates. MDA-231
               cells were plated at 2500 cells per well in clear-bottomed white 96-well plates. The cells were allowed to
               attach for two days, and then they were treated with increasing concentrations of prexasertib (LY2606368,
               Selleck Chemicals, Houston, TX), erlotinib (Selleck Chemicals), or both agents. The cells were exposed
               continuously for four days, and the viability was assessed with CellTiter-Glo. For EGF stimulation, MDA-231
               or MDA-468 were treated with EGF (human recombinant proteins, Life Technologies, Carlsbad, CA) at
               50 or 500 nmol/L at the same time as the indicated concentrations of prexasertib were added. Cells were
               exposed continuously for four days and viability assessed with CellTiter-Glo.


               For spheroid growth, clear-bottomed white 96-well plates were coated with 50 µL of Corning Matrigel
               Growth Factor Reduced (GFR) Basement Membrane Matrix (Fisher Scientific, Waltham, MA). MDA-231
               and MDA-468 cells were then seeded at 5000 cells per well on top of the gel layer in DMEM with 2.5%
               Matrigel. The cells were grown for four days to allow spheroids to form. The cells were then treated with
               increasing concentrations of prexasertib, erlotinib, or both agents. Cells were exposed continuously for four
               days, and the viability was assessed with CellTiter-Glo 3D.

               The assay reagent (CellTiter-Glo or CellTiter-Glo 3D) was added to plates and incubated according to the
               manufacturer’s instructions. Luminescence was read on a multimodal plate reader (Infinite M1000, Tecan,
               Männedorf, Switzerland). All viability assays were performed with technical triplicates over three biological
               replicates. The results were normalized to values for cells exposed to vehicle control and graphed to
               generate half-maximal inhibitory concentration (IC ) using software (Prism, GraphPad, San Diego, CA).
                                                           50
   26   27   28   29   30   31   32   33   34   35   36