Development of a test that measures real-time HER2 signaling function in live breast cancer cell lines and primary cells

Yao Huang, David J Burns, Benjamin E Rich, Ian A MacNeil, Abhijit Dandapat, Sajjad M Soltani, Samantha Myhre, Brian F Sullivan, Carol A Lange, Leo T Furcht, Lance G Laing, Yao Huang, David J Burns, Benjamin E Rich, Ian A MacNeil, Abhijit Dandapat, Sajjad M Soltani, Samantha Myhre, Brian F Sullivan, Carol A Lange, Leo T Furcht, Lance G Laing

Abstract

Background: Approximately 18-20% of all human breast cancers have overexpressed human epidermal growth factor receptor 2 (HER2). Standard clinical practice is to treat only overexpressed HER2 (HER2+) cancers with targeted anti-HER2 therapies. However, recent analyses of clinical trial data have found evidence that HER2-targeted therapies may benefit a sub-group of breast cancer patients with non-overexpressed HER2. This suggests that measurement of other biological factors associated with HER2 cancer, such as HER2 signaling pathway activity, should be considered as an alternative means of identifying patients eligible for HER2 therapies.

Methods: A new biosensor-based test (CELxTM HSF) that measures HER2 signaling activity in live cells is demonstrated using a set of 19 human HER2+ and HER2- breast cancer reference cell lines and primary cell samples derived from two fresh patient tumor specimens. Pathway signaling is elucidated by use of highly specific agonists and antagonists. The test method relies upon well-established phenotypic, adhesion-related, impedance changes detected by the biosensor.

Results: The analytical sensitivity and analyte specificity of this method was demonstrated using ligands with high affinity and specificity for HER1 and HER3. The HER2-driven signaling quantified ranged 50-fold between the lowest and highest cell lines. The HER2+ cell lines were almost equally divided into high and low signaling test result groups, suggesting that little correlation exists between HER2 protein expression and HER2 signaling level. Unexpectedly, the highest HER2-driven signaling level recorded was with a HER2- cell line.

Conclusions: Measurement of HER2 signaling activity in the tumor cells of breast cancer patients is a feasible approach to explore as a biomarker to identify HER2-driven cancers not currently diagnosable with genomic techniques. The wide range of HER2-driven signaling levels measured suggests it may be possible to make a distinction between normal and abnormal levels of activity. Analytical validation studies and clinical trials treating HER2- patients with abnormal HER2-driven signaling would be required to evaluate the analytical and clinical validity of using this functional biomarker as a diagnostic test to select patients for treatment with HER2 targeted therapy. In clinical practice, this method would require patient specimens be delivered to and tested in a central lab.

Keywords: Breast cancer; Breast tumor; CELx HSF Test; Cancer diagnostic; HER2-negative; HER2-positive; Oncology; Primary epithelial cells; Signaling pathway; Targeted therapeutics.

Figures

Fig. 1
Fig. 1
Representative CI versus time-course curves for basic cell attachment. Human breast cancer BT474 cells (a) or R56 patient-derived primary breast tumor cells (b) were seeded in a sensor plate and allowed to adhere, spread, and proliferate. Impedance was recorded as Cell Index (CI) versus time for 100 h after seeding. Cell attachment, stabilization, proliferation, and confluent phases are shown as indicated. c Representative images captured by an inverted phase contrast microscope (magnification: X40) showing cell morphology of BT474 and breast cancer R56 primary cells. Scale bar, 100 μm
Fig. 2
Fig. 2
Dose–response curves of EGF and NRG1b stimulation of HER2 signaling in SKBr3 cells. SKBr3 cells were seeded in the sensor plates and stimulated with serial titrations of a EGF (0 pM to 1200 pM) or b NRG1b (0 pM to 1350 pM). Instrument data for CELx curves are displayed using Delta CI values to demonstrate the relative signals to the time point (arrow) when the stimulus (EGF or NRG1b) was added. Log plots of dose-response curves with error bars of EGF and NRG1b stimulation are shown in the insets for a and b, respectively
Fig. 3
Fig. 3
Dose–response curves showing the effects of HER2 inhibitors on EGF- and NRG1b-directed HER2 signaling. a Neither pertuzumab nor lapatinib has significant effect on baseline cell signal determined before agonist addition. SKBr3 cells were seeded in sensor plates and treated with pertuzumab (10 μg/mL), lapatinib (200nM), or vehicle (control) 18 h prior to stimulation with NRG1 or EGF. CELx curves are displayed using Delta CI values to easily compare the relative change in signals from the time point of drug addition. The time points for drug addition and growth factor (GF) addition are indicated by black arrows. b and c SKBr3 cells were seeded in sensor plates and treated with serial titrations of lapatinib (0 nM to 3200 nM) or pertuzumab (0 μg/mL to 40 μg/mL) two hours prior to stimulation with EGF or NRG1b. Dose–response curves of drug inhibition on NRG1b and EGF-driven cell index signals are displayed
Fig. 4
Fig. 4
The PI3K/AKT pathway significantly contributes to the ligand-driven HER2 signaling activities detected by CELx HSF tests. a and b SKBr3 cells were seeded in sensor plates and then treated with a serial titration of the PI3K/AKT pathway inhibitor GSK1059615 (0 nM to 810 nM) two hours prior to maximal stimulation with NRG1b (800 pM) (a) or EGF (600 pM) (b). CELx curves are displayed using Delta CI values to demonstrate the relative signals to the time point (arrow) when the stimulus (EGF or NRG1b) was added. Dose–response curves of GSK1059615 inhibition on NRG1b and EGF-driven HER2 signals are shown in the insets
Fig. 5
Fig. 5
Comparison of EGF–HER2, NRG1b–HER2, and IGF-1–IGF-1R signaling systems in CELx assays. Human breast cancer T47D cells pre-seeded in sensor plates were treated with (a) pertuzumab (10 μg/mL), (b) lapatinib (200 nM), (c) linsitinib (200 nM), or (d) GSK1059615 (300 nM) two hours prior to stimulation with NRG1b (800 pM), EGF (600 pM), or IGF-1 (8 nM). CELx curves are displayed using Delta CI values to demonstrate the relative signals to the time point (arrow) when the stimulus (NRG1b, EGF, or IGF-1) was added. Blue curves, unstimulated cells (baseline); Green curves, cells stimulated with ligand (NRG1b, EGF, or IGF-1); Red curves, cells stimulated with ligand in the presence of drug (pertuzumab, lapatinib, linsitinib, or GSK1059615)
Fig. 6
Fig. 6
CELx HSF Test signals in HER2+ and HER2- breast cancer cell lines. a HER2+ cell lines (n = 9) and HER2- cell lines (n = 10) were evaluated with the CELx HSF test as described in the Methods. The sum of NRG1b- and EGF-driven HER2 signals that can be inhibited by the HER2-specific mAb pertuzumab was approximated as response units for all cell lines and plotted. b Comparison of NRG1b-driven CELx signals in AU565, BT483, SKBr3 (HER2+ reference cell line), and MDA-MB231 (HER2- reference cell line) and sensitivities to HER2-targeted drugs (pertuzumab, lapatinib, and afatinib). c HER2 expression levels in HER2+ (n = 9) and HER2- cell lines (n = 10) were determined by fluorescence flow cytometry (mean fluorescence channel units, MFC) and plotted against the corresponding HER2 signal determined by CELx HSF test (response units) for each cell line. No correlation between the two parameters was observed (P = 0.204, R2 = 0.0929). Empty circles, HER2- cell lines; Filled circles, HER2+ cell lines. The locations of BT483, AU565, SKBr3 (HER2+ reference cell line) and MDA-MB231 (HER2- reference cell line) are indicated
Fig. 7
Fig. 7
Subtypes of CELx HSF curves. Representative CELx time-course curves representing HER2+/HSF+ (HER2+ cells having high HER2 signaling activities) (a), HER2+/HSF- (HER2+ cells having low HER2 signaling activities) (b), HER2-/HSF+ (HER2- cells having high HER2 signaling activities) (c), and HER2-/HSF- (HER2- cells having low HER2 signaling activities) (d) are shown. For display purposes, NRG1b and EGF-driven HER2 CELx signals are shown in separated panels. CELx curves are displayed using Delta CI values to demonstrate the relative signals to the time point (arrow) when the stimulus (EGF or NRG1b) was added. Red curves, unstimulated cells (control); Green curves, cells stimulated with ligand (NRG1b or EGF); Blue curves, cells stimulated with ligand in the presence of drug (pertuzumab)
Fig. 8
Fig. 8
Validation of CELx HSF test in patient tissue specimen-derived primary cells ex vivo. Primary epithelial cells derived from two HER2- (R39 and R49) patients with breast cancer and one healthy control subject (R62) were subjected to CELx HSF tests. Responses of NRG1b-driven HER2 CELx signals with and without pertuzumab for these primary cells are plotted along with those for the HER2+ reference cell line (SKBr3) and the HER2- reference cell line (MDA-MB231) as bar charts. Black bars, cells stimulated with NRG1b; Grey bars, cells stimulated with NRG1b in the presence of pertuzumab. HER2- Patient R39 has approximately 80% of the NRG1 CELx signal of HER2+ cell line SKBr3
Fig. 9
Fig. 9
The PI3K/AKT pathway significantly contributes to the ligand-driven HER2 signaling activities detected by CELx HSF tests in patient-derived breast tumor primary cells. Patient R54 breast tumor-derived primary cells (15,000 cells per well) pre-seeded in sensor plates were treated with a serial titration of GSK1059615 (0 nM to 2700 nM) two hours prior to stimulation with NRG1b (800 pM) (a) or EGF (600 pM) (b). The dose-dependent inhibitory effect of GSK1059615 on ligand-driven HER2 signals is shown

References

    1. Gonzalez-Angulo AM, Litton JK, Broglio KR, Meric-Bernstam F, Rakkhit R, Cardoso F, Peintinger F, Hanrahan EO, Sahin A, Guray M, et al. High risk of recurrence for patients with breast cancer who have human epidermal growth factor receptor 2-positive, node-negative tumors 1 cm or smaller. J Clin Oncol. 2009;27(34):5700–5706. doi: 10.1200/JCO.2009.23.2025.
    1. Santa-Maria CA, Nye L, Mutonga MB, Jain S, Gradishar WJ. Management of Metastatic HER2-Positive Breast Cancer: Where Are We and Where Do We Go From Here? Oncology (Williston Park). 2016;30(2):148–55.
    1. Eroglu Z, Tagawa T, Somlo G. Human epidermal growth factor receptor family-targeted therapies in the treatment of HER2-overexpressing breast cancer. Oncologist. 2014;19(2):135–150. doi: 10.1634/theoncologist.2013-0283.
    1. Johnston S, Pippen J, Jr, Pivot X, Lichinitser M, Sadeghi S, Dieras V, Gomez HL, Romieu G, Manikhas A, Kennedy MJ, et al. Lapatinib combined with letrozole versus letrozole and placebo as first-line therapy for postmenopausal hormone receptor-positive metastatic breast cancer. J Clin Oncol. 2009;27(33):5538–5546. doi: 10.1200/JCO.2009.23.3734.
    1. Coombes RC, Tat T, Miller ML, Reise JA, Mansi JL, Hadjiminas DJ, Shousha S, Elsheikh SE, Lam EW, Horimoto Y, et al. An open-label study of lapatinib in women with HER-2-negative early breast cancer: the lapatinib pre-surgical study (LPS study) Ann Oncol. 2013;24(4):924–930. doi: 10.1093/annonc/mds594.
    1. Paik S, Kim C, Wolmark N. HER2 status and benefit from adjuvant trastuzumab in breast cancer. N Engl J Med. 2008;358(13):1409–1411. doi: 10.1056/NEJMc0801440.
    1. Perez EA, Reinholz MM, Hillman DW, Tenner KS, Schroeder MJ, Davidson NE, Martino S, Sledge GW, Harris LN, Gralow JR, et al. HER2 and chromosome 17 effect on patient outcome in the N9831 adjuvant trastuzumab trial. J Clin Oncol. 2010;28(28):4307–4315. doi: 10.1200/JCO.2009.26.2154.
    1. Leary A, Evans A, Johnston SR, A’Hern R, Bliss JM, Sahoo R, Detre S, Haynes BP, Hills M, Harper-Wynne C, et al. Antiproliferative Effect of Lapatinib in HER2-Positive and HER2-Negative/HER3-High Breast Cancer: Results of the Presurgical Randomized MAPLE Trial (CRUK E/06/039) Clin Cancer Res. 2015;21(13):2932–2940. doi: 10.1158/1078-0432.CCR-14-1428.
    1. Pogue-Geile KL, Kim C, Jeong JH, Tanaka N, Bandos H, Gavin PG, Fumagalli D, Goldstein LC, Sneige N, Burandt E, et al. Predicting degree of benefit from adjuvant trastuzumab in NSABP trial B-31. J Natl Cancer Inst. 2013;105(23):1782–1788. doi: 10.1093/jnci/djt321.
    1. Huang Y, Chang Y. Epidermal Growth factor receptor (EGFR) phosphorylation, signaling and trafficking in prostate cancer. In: Spiess PE: InTech, editor. Prostate Cancer - From Bench to Bedside. 2011.
    1. Olayioye MA, Graus-Porta D, Beerli RR, Rohrer J, Gay B, Hynes NE. ErbB-1 and ErbB-2 acquire distinct signaling properties dependent upon their dimerization partner. Mol Cell Biol. 1998;18(9):5042–5051. doi: 10.1128/MCB.18.9.5042.
    1. Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 2009;9(7):463–475. doi: 10.1038/nrc2656.
    1. Campbell MR, Moasser MM. HER Targeting in HER2-Negative Breast Cancers: Looking for the HER3 Positive. Clin Cancer Res. 2015;21(13):2886–2888. doi: 10.1158/1078-0432.CCR-14-3012.
    1. Holbro T, Beerli RR, Maurer F, Koziczak M, Barbas CF, 3rd, Hynes NE. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc Natl Acad Sci U S A. 2003;100(15):8933–8938. doi: 10.1073/pnas.1537685100.
    1. Lapin V, Shirdel EA, Wei X, Mason JM, Jurisica I, Mak TW. Kinome-wide screening of HER2+ breast cancer cells for molecules that mediate cell proliferation or sensitize cells to trastuzumab therapy. Oncogenesis. 2014;3:e133. doi: 10.1038/oncsis.2014.45.
    1. Hendriks BS, Orr G, Wells A, Wiley HS, Lauffenburger DA. Parsing ERK activation reveals quantitatively equivalent contributions from epidermal growth factor receptor and HER2 in human mammary epithelial cells. J Biol Chem. 2005;280(7):6157–6169. doi: 10.1074/jbc.M410491200.
    1. Schulze WX, Deng L, Mann M. Phosphotyrosine interactome of the ErbB-receptor kinase family. Mol Syst Biol. 2005;1:2005 0008. doi: 10.1038/msb4100012.
    1. Scott CW, Peters MF. Label-free whole-cell assays: expanding the scope of GPCR screening. Drug Discov Today. 2010;15(17–18):704–716. doi: 10.1016/j.drudis.2010.06.008.
    1. Armando Gagliardi P, Puliafito A, di Blasio L, Chianale F, Somale D, Seano G, Bussolino F, Primo L. Real-time monitoring of cell protrusion dynamics by impedance responses. Sci Rep. 2015;5:10206. doi: 10.1038/srep10206.
    1. Peters MF, Scott CW. Evaluating cellular impedance assays for detection of GPCR pleiotropic signaling and functional selectivity. J Biomol Screen. 2009;14(3):246–255. doi: 10.1177/1087057108330115.
    1. Halai R, Croker DE, Suen JY, Fairlie DP, Cooper MA. A Comparative Study of Impedance versus Optical Label-Free Systems Relative to Labelled Assays in a Predominantly Gi Coupled GPCR (C5aR) Signalling. Biosensors (Basel) 2012;2(3):273–290. doi: 10.3390/bios2030273.
    1. Stampfer MR, Yaswen P, Taylor-Papadimitriou J. Culture of human mammary epithelial cells. In: Freshney RI, Freshney MG, editors. Culture of Epithelial Cells. 2 2002.
    1. Proia DA, Kuperwasser C. Reconstruction of human mammary tissues in a mouse model. Nat Protoc. 2006;1(1):206–214. doi: 10.1038/nprot.2006.31.
    1. Campbell LH, Brockbank KG. Serum-free solutions for cryopreservation of cells. In Vitro Cell Dev Biol Anim. 2007;43(8–9):269–275. doi: 10.1007/s11626-007-9039-z.
    1. Nelson MH, Dolder CR. Lapatinib: a novel dual tyrosine kinase inhibitor with activity in solid tumors. Ann Pharmacother. 2006;40(2):261–269. doi: 10.1345/aph.1G387.
    1. Agus DB, Akita RW, Fox WD, Lewis GD, Higgins B, Pisacane PI, Lofgren JA, Tindell C, Evans DP, Maiese K, et al. Targeting ligand-activated ErbB2 signaling inhibits breast and prostate tumor growth. Cancer Cell. 2002;2(2):127–137. doi: 10.1016/S1535-6108(02)00097-1.
    1. Jackson JG, St Clair P, Sliwkowski MX, Brattain MG. Blockade of epidermal growth factor- or heregulin-dependent ErbB2 activation with the anti-ErbB2 monoclonal antibody 2C4 has divergent downstream signaling and growth effects. Cancer Res. 2004;64(7):2601–2609. doi: 10.1158/0008-5472.CAN-03-3106.
    1. Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486(7404):532–536.
    1. Junttila TT, Akita RW, Parsons K, Fields C, Lewis Phillips GD, Friedman LS, Sampath D, Sliwkowski MX. Ligand-independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively inhibited by the PI3K inhibitor GDC-0941. Cancer Cell. 2009;15(5):429–440. doi: 10.1016/j.ccr.2009.03.020.
    1. Salama AK, Kim KB. Trametinib (GSK1120212) in the treatment of melanoma. Expert Opin Pharmacother. 2013;14(5):619–627. doi: 10.1517/14656566.2013.770475.
    1. Kuma Y, Sabio G, Bain J, Shpiro N, Marquez R, Cuenda A. BIRB796 inhibits all p38 MAPK isoforms in vitro and in vivo. J Biol Chem. 2005;280(20):19472–19479. doi: 10.1074/jbc.M414221200.
    1. Bennett BL, Sasaki DT, Murray BW, O’Leary EC, Sakata ST, Xu W, Leisten JC, Motiwala A, Pierce S, Satoh Y, et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc Natl Acad Sci U S A. 2001;98(24):13681–13686. doi: 10.1073/pnas.251194298.
    1. Mulvihill MJ, Cooke A, Rosenfeld-Franklin M, Buck E, Foreman K, Landfair D, O’Connor M, Pirritt C, Sun Y, Yao Y, et al. Discovery of OSI-906: a selective and orally efficacious dual inhibitor of the IGF-1 receptor and insulin receptor. Future Med Chem. 2009;1(6):1153–1171. doi: 10.4155/fmc.09.89.
    1. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, Wilson CJ, Lehar J, Kryukov GV, Sonkin D, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483(7391):603–607. doi: 10.1038/nature11003.
    1. Czyz ZT, Hoffmann M, Schlimok G, Polzer B, Klein CA. Reliable single cell array CGH for clinical samples. PLoS One. 2014;9(1):e85907. doi: 10.1371/journal.pone.0085907.
    1. Heiser LM, Sadanandam A, Kuo WL, Benz SC, Goldstein TC, Ng S, Gibb WJ, Wang NJ, Ziyad S, Tong F, et al. Subtype and pathway specific responses to anticancer compounds in breast cancer. Proc Natl Acad Sci U S A. 2012;109(8):2724–2729. doi: 10.1073/pnas.1018854108.
    1. Minkovsky N, Berezov A. BIBW-2992, a dual receptor tyrosine kinase inhibitor for the treatment of solid tumors. Curr Opin Investig Drugs. 2008;9(12):1336–1346.
    1. Perez EA, Cortes J, Gonzalez-Angulo AM, Bartlett JM. HER2 testing: current status and future directions. Cancer Treat Rev. 2014;40(2):276–284. doi: 10.1016/j.ctrv.2013.09.001.
    1. Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark L, Bayani N, Coppe JP, Tong F, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006;10(6):515–527. doi: 10.1016/j.ccr.2006.10.008.
    1. Kao J, Salari K, Bocanegra M, Choi YL, Girard L, Gandhi J, Kwei KA, Hernandez-Boussard T, Wang P, Gazdar AF, et al. Molecular profiling of breast cancer cell lines defines relevant tumor models and provides a resource for cancer gene discovery. PLoS One. 2009;4(7):e6146. doi: 10.1371/journal.pone.0006146.
    1. Holliday DL, Speirs V. Choosing the right cell line for breast cancer research. Breast Cancer Res. 2011;13(4):215. doi: 10.1186/bcr2889.
    1. Lim E, Vaillant F, Wu D, Forrest NC, Pal B, Hart AH, Asselin-Labat ML, Gyorki DE, Ward T, Partanen A, et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat Med. 2009;15(8):907–913. doi: 10.1038/nm.2000.
    1. Hinow P, Wang SE, Arteaga CL, Webb GF. relocating job wise? A mathematical model separates quantitatively the cytostatic and cytotoxic effects of a HER2 tyrosine kinase inhibitor. Theor Biol Med Model. 2007;4:14. doi: 10.1186/1742-4682-4-14.
    1. Burris HA, 3rd, Hurwitz HI, Dees EC, Dowlati A, Blackwell KL, O’Neil B, Marcom PK, Ellis MJ, Overmoyer B, Jones SF, et al. Phase I safety, pharmacokinetics, and clinical activity study of lapatinib (GW572016), a reversible dual inhibitor of epidermal growth factor receptor tyrosine kinases, in heavily pretreated patients with metastatic carcinomas. J Clin Oncol. 2005;23(23):5305–5313. doi: 10.1200/JCO.2005.16.584.
    1. Ringerike T, Stang E, Johannessen LE, Sandnes D, Levy FO, Madshus IH. High-affinity binding of epidermal growth factor (EGF) to EGF receptor is disrupted by overexpression of mutant dynamin (K44A) J Biol Chem. 1998;273(27):16639–16642. doi: 10.1074/jbc.273.27.16639.
    1. Sliwkowski MX, Schaefer G, Akita RW, Lofgren JA, Fitzpatrick VD, Nuijens A, Fendly BM, Cerione RA, Vandlen RL, Carraway KL., 3rd Coexpression of erbB2 and erbB3 proteins reconstitutes a high affinity receptor for heregulin. J Biol Chem. 1994;269(20):14661–14665.
    1. Steinkamp MP, Low-Nam ST, Yang S, Lidke KA, Lidke DS, Wilson BS. erbB3 is an active tyrosine kinase capable of homo- and heterointeractions. Mol Cell Biol. 2014;34(6):965–977. doi: 10.1128/MCB.01605-13.
    1. Shi F, Telesco SE, Liu Y, Radhakrishnan R, Lemmon MA. ErbB3/HER3 intracellular domain is competent to bind ATP and catalyze autophosphorylation. Proc Natl Acad Sci U S A. 2010;107(17):7692–7697. doi: 10.1073/pnas.1002753107.
    1. Huang X, Gao L, Wang S, McManaman JL, Thor AD, Yang X, Esteva FJ, Liu B. Heterotrimerization of the growth factor receptors erbB2, erbB3, and insulin-like growth factor-i receptor in breast cancer cells resistant to herceptin. Cancer Res. 2010;70(3):1204–1214. doi: 10.1158/0008-5472.CAN-09-3321.
    1. Xia W, Petricoin EF, 3rd, Zhao S, Liu L, Osada T, Cheng Q, Wulfkuhle JD, Gwin WR, Yang X, Gallagher RI, et al. An heregulin-EGFR-HER3 autocrine signaling axis can mediate acquired lapatinib resistance in HER2+ breast cancer models. Breast Cancer Res. 2013;15(5):R85. doi: 10.1186/bcr3480.
    1. Sergina NV, Rausch M, Wang D, Blair J, Hann B, Shokat KM, Moasser MM. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature. 2007;445(7126):437–441. doi: 10.1038/nature05474.
    1. Hollestelle A, Elstrodt F, Nagel JH, Kallemeijn WW, Schutte M. Phosphatidylinositol-3-OH kinase or RAS pathway mutations in human breast cancer cell lines. Mol Cancer Res. 2007;5(2):195–201. doi: 10.1158/1541-7786.MCR-06-0263.
    1. Rodriguez-Viciana P, Warne PH, Dhand R, Vanhaesebroeck B, Gout I, Fry MJ, Waterfield MD, Downward J. Phosphatidylinositol-3-OH kinase as a direct target of Ras. Nature. 1994;370(6490):527–532. doi: 10.1038/370527a0.
    1. Fallahi-Sichani M, Honarnejad S, Heiser LM, Gray JW, Sorger PK. Metrics other than potency reveal systematic variation in responses to cancer drugs. Nat Chem Biol. 2013;9(11):708–714. doi: 10.1038/nchembio.1337.
    1. Knight SD, Adams ND, Burgess JL, Chaudhari AM, Darcy MG, Donatelli CA, Luengo JI, Newlander KA, Parrish CA, Ridgers LH, et al. Discovery of GSK2126458, a Highly Potent Inhibitor of PI3K and the Mammalian Target of Rapamycin. ACS Med Chem Lett. 2010;1(1):39–43. doi: 10.1021/ml900028r.
    1. Brady SW, Zhang J, Seok D, Wang H, Yu D. Enhanced PI3K p110alpha signaling confers acquired lapatinib resistance that can be effectively reversed by a p110alpha-selective PI3K inhibitor. Mol Cancer Ther. 2014;13(1):60–70. doi: 10.1158/1535-7163.MCT-13-0518.
    1. Mukohara T. PI3K mutations in breast cancer: prognostic and therapeutic implications. Breast Cancer (Dove Med Press) 2015;7:111–123.
    1. Duncan JS, Whittle MC, Nakamura K, Abell AN, Midland AA, Zawistowski JS, Johnson NL, Granger DA, Jordan NV, Darr DB, et al. Dynamic reprogramming of the kinome in response to targeted MEK inhibition in triple-negative breast cancer. Cell. 2012;149(2):307–321. doi: 10.1016/j.cell.2012.02.053.
    1. Stuhlmiller TJ, Miller SM, Zawistowski JS, Nakamura K, Beltran AS, Duncan JS, Angus SP, Collins KA, Granger DA, Reuther RA, et al. Inhibition of Lapatinib-Induced Kinome Reprogramming in ERBB2-Positive Breast Cancer by Targeting BET Family Bromodomains. Cell Rep. 2015;11(3):390–404. doi: 10.1016/j.celrep.2015.03.037.
    1. Mylona A, Theillet FX, Foster C, Cheng TM, Miralles F, Bates PA, Selenko P, Treisman R. Opposing effects of Elk-1 multisite phosphorylation shape its response to ERK activation. Science. 2016;354(6309):233–237. doi: 10.1126/science.aad1872.
    1. Santarpia L, Bottai G, Kelly CM, Gyorffy B, Szekely B, Pusztai L. Deciphering and Targeting Oncogenic Mutations and Pathways in Breast Cancer. Oncologist. 2016;21(9):1063–1078. doi: 10.1634/theoncologist.2015-0369.
    1. Lee CY, Lin Y, Bratman SV, Feng W, Kuo AH, Scheeren FA, Engreitz JM, Varma S, West RB, Diehn M. Neuregulin autocrine signaling promotes self-renewal of breast tumor-initiating cells by triggering HER2/HER3 activation. Cancer Res. 2014;74(1):341–352. doi: 10.1158/0008-5472.CAN-13-1055.
    1. Ithimakin S, Day KC, Malik F, Zen Q, Dawsey SJ, Bersano-Begey TF, Quraishi AA, Ignatoski KW, Daignault S, Davis A, et al. HER2 drives luminal breast cancer stem cells in the absence of HER2 amplification: implications for efficacy of adjuvant trastuzumab. Cancer Res. 2013;73(5):1635–1646. doi: 10.1158/0008-5472.CAN-12-3349.
    1. Mayer IA, Arteaga CL. Does lapatinib work against HER2-negative breast cancers? Clin Cancer Res. 2010;16(5):1355–1357. doi: 10.1158/1078-0432.CCR-09-3223.

Source: PubMed

3
Prenumerera