A preclinical evaluation of the MEK inhibitor refametinib in HER2-positive breast cancer cell lines including those with acquired resistance to trastuzumab or lapatinib

John O'Shea, Mattia Cremona, Clare Morgan, Malgorzata Milewska, Frankie Holmes, Virginia Espina, Lance Liotta, Joyce O'Shaughnessy, Sinead Toomey, Stephen F Madden, Aoife Carr, Naomi Elster, Bryan T Hennessy, Alex J Eustace, John O'Shea, Mattia Cremona, Clare Morgan, Malgorzata Milewska, Frankie Holmes, Virginia Espina, Lance Liotta, Joyce O'Shaughnessy, Sinead Toomey, Stephen F Madden, Aoife Carr, Naomi Elster, Bryan T Hennessy, Alex J Eustace

Abstract

Purpose: The MEK/MAPK pathway is commonly activated in HER2-positive breast cancer, but little investigation of targeting this pathway has been undertaken. Here we present the results of an in vitro preclinical evaluation of refametinib, an allosteric MEK1/2 inhibitor, in HER2-positive breast cancer cell lines including models of acquired resistance to trastuzumab or lapatinib.

Methods: A panel of HER2-positive breast cancer cells were profiled for mutational status and also for anti-proliferative response to refametinib alone and in combination with the PI3K inhibitor (PI3Ki) copanlisib and the HER2-targeted therapies trastuzumab and lapatinib. Reverse phase protein array (RPPA) was used to determine the effect of refametinib alone and in combination with PI3Ki and HER2-inhibitors on expression and phosphorylation of proteins in the PI3K/AKT and MEK/MAPK pathways. We validated our proteomic in vitro findings by utilising RPPA analysis of patients who received either trastuzumab, lapatinib or the combination of both drugs in the NCT00524303/LPT109096 clinical trial.

Results: Refametinib has anti-proliferative effects when used alone in 2/3 parental HER2-positive breast cancer cell lines (HCC1954, BT474), along with 3 models of these 2 cell lines with acquired trastuzumab or lapatinib resistance (6 cell lines tested). Refametinib treatment led to complete inhibition of MAPK signalling. In HCC1954, the most refametinib-sensitive cell line (IC50= 397 nM), lapatinib treatment inhibits phosphorylation of MEK and MAPK but activates AKT phosphorylation, in contrast to the other 2 parental cell lines tested (BT474-P, SKBR3-P), suggesting that HER2 may directly activate MEK/MAPK and not PI3K/AKT in HCC1954 cells but not in the other 2 cell lines, perhaps explaining the refametinib-sensitivity of this cell line. Using RPPA data from patients who received either trastuzumab, lapatinib or the combination of both drugs together with chemotherapy in the NCT00524303 clinical trial, we found that 18% (n=38) of tumours had decreased MAPK and increased AKT phosphorylation 14 days after treatment with HER2-targeted therapies. The combination of MEK inhibition (MEKi) with refametinib and copanlisib led to synergistic inhibition of growth in 4/6 cell lines tested (CI @ED75 = 0.39-0.75), whilst the combinations of lapatinib and refametinib led to synergistic inhibition of growth in 3/6 cell lines (CI @ED75 = 0.39-0.80).

Conclusion: Refametinib alone or in combination with copanlisib or lapatinib could represent an improved treatment strategy for some patients with HER2-positive breast cancer, and should be considered for clinical trial evaluation. The direct down-regulation of MEK/MAPK but not AKT signalling by HER2 inhibition (e.g. by lapatinib or trastuzumab), which we demonstrate occurs in 18% of HER2-positive breast cancers may serve as a potential biomarker of responsiveness to the MEK inhibitor refametinib.

Keywords: HER2-positive breast cancer; MEK inhibitor; acquired resistance to HER2-targeted therapies; reverse phase protein array.

Conflict of interest statement

CONFLICTS OF INTEREST The authors state that they have no conflicts of interest in relation to this article or the funding bodies.

Figures

Figure 1. Differential expression of MEK1 as…
Figure 1. Differential expression of MEK1 as determined by RPPA in a panel of breast cancer cell lines (n=28) dependant on their sensitivity to the MEK inhibitors tremitinib and PD-0325901
Standard deviations are calculated from analysis of MEK expression in the sensitive (n=15) versus the resistant (n=12) cell lines analysed on the same RPPA slide. ‘*’ indicates a significant p value of <0.05 as determined by the students t-test.
Figure 2
Figure 2
(A) RPPA analysis displaying the fold-change in protein expression or phosphorylation relative to untreated controls in cell lines (HCC1954-P, -L) treated with 300nM refametinib (MEKi). (B) RPPA analysis displaying the basal levels of EGFR and ERBB3 protein expression and phosphorylation in the HCC1954-P and HCC1954-L cells. Standard deviations are calculated from triplicate independent protein samples analysed on the same RPPA slide. ‘*’ indicates proteins which have a change of signal intensity greater than 1.2 fold and a p-value of <0.05 as determined by the students t-test.
Figure 3
Figure 3
(A) RPPA analysis displaying the fold-change in protein expression or phosphorylation relative to control treated cells in cell lines (HCC1954-P, BT474-P and SKBR3-P) treated with 150nM lapatinib for 30 minutes (Lap30min). (B) Representative figure demonstrating hypothesised inhibition of MAPK/ERK signalling in HCC1954 and SKBR3 cells as a result of lapatinib treatment. Standard deviations are calculated from triplicate independent protein samples analysed on the same RPPA slide. ‘*’ indicates proteins which have a change of signal intensity of greater than 1.2 fold and a p value of <0.05 as determined by the students t-test.
Figure 4
Figure 4
RPPA analysis of the NCT00524303 clinical trial looking at changes in AKT and MAPK signalling pathways in tumours in response to treatment with either (A) trastuzumab, (B) lapatinib or (C) a combination of both. Patient samples where AKT (S473) is increased by >20% and MAPK(T202/Y204) is decreased by >20% are indicated in red.
Figure 5. Efficacy of refametinib (MEKi) (-□-),…
Figure 5. Efficacy of refametinib (MEKi) (-□-), copanlisib (PI3Ki) (-◊-) and a combination of refametinib and copanlisib (--∆--) in a panel of HER2-positive breast cancer cell lines, including parental cells (-P) and those with acquired resistance to either trastuzumab (-T or -Res) or lapatinib (-L)
Error bars are representative of standard deviations across triplicate independent experiments. The ratio of refametinib:copanlisib in this assay was fixed at either 20:1 or 200:1.
Figure 6. RPPA analysis displaying the fold-change…
Figure 6. RPPA analysis displaying the fold-change in protein expression or phosphorylation relative to control treated cells in cell lines treated with either 300nM refametinib (MEKi) or 15nM copanlisib (PI3Ki) alone or in combination (MEKi - 300nM: PI3Ki - 15nM) in HCC1954-P (parental) and -L (lapatinib resistant) cells
Standard deviations are calculated from at least triplicate biologically independent protein samples analysed on the same RPPA slide. ‘*’ indicates proteins which have a change of signal intensity of greater than 1.2 fold and a p-value of

Figure 7. RPPA analysis displaying the fold-change…

Figure 7. RPPA analysis displaying the fold-change in protein expression or phosphorylation relative to control…

Figure 7. RPPA analysis displaying the fold-change in protein expression or phosphorylation relative to control treated cells in cell lines treated with either 300nM refametinib (MEKi) or Lapatinib (HCC1954-P - 150nM: HCC1954-L - 500nM) alone or in combination in HCC1954-P and -L cells
Standard deviations are calculated from at least triplicate biologically independent protein samples analysed on the same RPPA slide. ‘*’ indicates proteins which have a change of signal intensity of greater than 1.2 fold and a p-value of

Figure 8. Efficacy of refametinib (MEKi) (-□-),…

Figure 8. Efficacy of refametinib (MEKi) (-□-), lapatinib (-◊-) and a combination of refametinib and…

Figure 8. Efficacy of refametinib (MEKi) (-□-), lapatinib (-◊-) and a combination of refametinib and lapatinib (--∆--) in a panel of HER2-positive breast cancer cell lines, including parental cell lines(-P) and matched cells with acquired resistance to either trastuzumab (-T or -Res) or lapatinib (-L)
Error bars are representative of standard deviations across triplicate independant experiments. The ratio of refametinib:lapatinib in this assay was fixed at 2:1.
All figures (8)
Similar articles
Cited by
References
    1. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer. 2013;49:1374–403. https://doi.org/10.1016/j.ejca.2012.12.027. - DOI - PubMed
    1. Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51. https://doi.org/10.1056/NEJMra043186. - DOI - PubMed
    1. Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG, Udove J, Ullrich A, Press MF. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707–12. - PubMed
    1. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–92. https://doi.org/10.1056/NEJM200103153441101. - DOI - PubMed
    1. Esteva FJ, Valero V, Booser D, Guerra LT, Murray JL, Pusztai L, Cristofanilli M, Arun B, Esmaeli B, Fritsche HA, Sneige N, Smith TL, Hortobagyi GN. Phase II study of weekly docetaxel and trastuzumab for patients with HER-2-overexpressing metastatic breast cancer. J Clin Oncol. 2002;20:1800–8. - PubMed
Show all 43 references
Associated data
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 7. RPPA analysis displaying the fold-change…
Figure 7. RPPA analysis displaying the fold-change in protein expression or phosphorylation relative to control treated cells in cell lines treated with either 300nM refametinib (MEKi) or Lapatinib (HCC1954-P - 150nM: HCC1954-L - 500nM) alone or in combination in HCC1954-P and -L cells
Standard deviations are calculated from at least triplicate biologically independent protein samples analysed on the same RPPA slide. ‘*’ indicates proteins which have a change of signal intensity of greater than 1.2 fold and a p-value of

Figure 8. Efficacy of refametinib (MEKi) (-□-),…

Figure 8. Efficacy of refametinib (MEKi) (-□-), lapatinib (-◊-) and a combination of refametinib and…

Figure 8. Efficacy of refametinib (MEKi) (-□-), lapatinib (-◊-) and a combination of refametinib and lapatinib (--∆--) in a panel of HER2-positive breast cancer cell lines, including parental cell lines(-P) and matched cells with acquired resistance to either trastuzumab (-T or -Res) or lapatinib (-L)
Error bars are representative of standard deviations across triplicate independant experiments. The ratio of refametinib:lapatinib in this assay was fixed at 2:1.
All figures (8)
Figure 8. Efficacy of refametinib (MEKi) (-□-),…
Figure 8. Efficacy of refametinib (MEKi) (-□-), lapatinib (-◊-) and a combination of refametinib and lapatinib (--∆--) in a panel of HER2-positive breast cancer cell lines, including parental cell lines(-P) and matched cells with acquired resistance to either trastuzumab (-T or -Res) or lapatinib (-L)
Error bars are representative of standard deviations across triplicate independant experiments. The ratio of refametinib:lapatinib in this assay was fixed at 2:1.

References

    1. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer. 2013;49:1374–403. .
    1. Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51. .
    1. Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG, Udove J, Ullrich A, Press MF. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707–12.
    1. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–92. .
    1. Esteva FJ, Valero V, Booser D, Guerra LT, Murray JL, Pusztai L, Cristofanilli M, Arun B, Esmaeli B, Fritsche HA, Sneige N, Smith TL, Hortobagyi GN. Phase II study of weekly docetaxel and trastuzumab for patients with HER-2-overexpressing metastatic breast cancer. J Clin Oncol. 2002;20:1800–8.
    1. O’Donovan N, Byrne AT, O’Connor AE, McGee S, Gallagher WM, Crown J. Synergistic interaction between trastuzumab and EGFR/HER-2 tyrosine kinase inhibitors in HER-2 positive breast cancer cells. Invest New Drugs. 2011;29:752–9. .
    1. Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B, Skarlos D, Campone M, Davidson N, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733–43. .
    1. Blackwell KL, Burstein HJ, Storniolo AM, Rugo HS, Sledge G, Aktan G, Ellis C, Florance A, Vukelja S, Bischoff J, Baselga J, O’Shaughnessy J. Overall survival benefit with lapatinib in combination with trastuzumab for patients with human epidermal growth factor receptor 2-positive metastatic breast cancer: final results from the EGF104900 Study. J Clin Oncol. 2012;30:2585–92. .
    1. Garrett JT, Arteaga CL. Resistance to HER2-directed antibodies and tyrosine kinase inhibitors: mechanisms and clinical implications. Cancer Biol Ther. 2011;11:793–800.
    1. Elster N, Collins DM, Toomey S, Crown J, Eustace AJ, Hennessy BT. HER2-family signalling mechanisms, clinical implications and targeting in breast cancer. Breast Cancer Res Treat. 2015;149:5–15. .
    1. Weigelt B, Lo AT, Park CC, Gray JW, Bissell MJ. HER2 signaling pathway activation and response of breast cancer cells to HER2-targeting agents is dependent strongly on the 3D microenvironment. Breast Cancer Res Treat. 2010;122:35–43. .
    1. Gayle SS, Castellino RC, Buss MC, Nahta R. MEK inhibition increases lapatinib sensitivity via modulation of FOXM1. Curr Med Chem. 2013;20:2486–99.
    1. Cheng H, Liu P, Ohlson C, Xu E, Symonds L, Isabella A, Muller WJ, Lin NU, Krop IE, Roberts TM, Winer EP, Arteaga CL, Zhao JJ. PIK3CA(H1047R)- and Her2-initiated mammary tumors escape PI3K dependency by compensatory activation of MEK-ERK signaling. Oncogene. 2016;35:2961–70. .
    1. McDermott MS, Browne BC, Conlon NT, O’Brien NA, Slamon DJ, Henry M, Meleady P, Clynes M, Dowling P, Crown J, O’Donovan N. PP2A inhibition overcomes acquired resistance to HER2 targeted therapy. Mol Cancer. 2014;13:157. .
    1. McDermott M, Eustace AJ, Busschots S, Breen L, Crown J, Clynes M, O’Donovan N, Stordal B. In vitro development of chemotherapy and targeted therapy drug-resistant cancer cell lines: a practical guide with case studies. Front Oncol. 2014;4:40. .
    1. Eustace AJ, Crown J, Clynes M, O’Donovan N. Preclinical evaluation of dasatinib, a potent Src kinase inhibitor, in melanoma cell lines. J Transl Med. 2008;6:53. .
    1. Holmes FA, Espina V, Liotta LA, Nagarwala YM, Danso M, McIntyre KJ, Osborne CR, Anderson T, Krekow L, Blum JL, Pippen J, Florance A, Mahoney J, et al. Pathologic complete response after preoperative anti-HER2 therapy correlates with alterations in PTEN, FOXO,
    1. Hennessy BT, Lu Y, Gonzalez-Angulo AM, Carey MS, Myhre S, Ju Z, Davies MA, Liu W, Coombes K, Meric-Bernstam F, Bedrosian I, McGahren M, Agarwal R, et al. A technical assessment of the utility of reverse phase protein arrays for the s tudy of the functional proteome in non-microdissected human breast cancers. Clin Proteomics. 2010;6:129–51. .
    1. Stemke-Hale K, Gonzalez-Angulo AM, Lluch A, Neve RM, Kuo WL, Davies M, Carey M, Hu Z, Guan Y, Sahin A, Symmans WF, Pusztai L, Nolden LK, et al. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 2008;68:6084–91. .
    1. Paweletz CP, Charboneau L, Bichsel VE, Simone NL, Chen T, Gillespie JW, Emmert-Buck MR, Roth MJ, Petricoin IE, Liotta LA. Reverse phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front. Oncogene. 2001;20:1981–9. .
    1. Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984;22:27–55.
    1. Elster N, Cremona M, Morgan C, Toomey S, Carr A, O’Grady A, Hennessy BT, Eustace AJ. A preclinical evaluation of the PI3K alpha/delta dominant inhibitor BAY 80-6946 in HER2-positive breast cancer models with acquired resistance to the HER2-targeted therapies trastuzumab and lapatinib. Breast Cancer Res Treat. 2015;149:373–83. .
    1. Liu N, Haegebarth A, Pühler F, Scholz A, Hoffmann J, Dubowy R, Krissel H, Wilhelm SM, Mumberg D, Ziegelbauer K. BAY. 80-6946 and allosteric MEK inhibitor BAY 86-9766 (RDEA119), a promising approach for the treatment of colorectal cancers. EJC Supplements. 2010;8:50–50.
    1. Liu N, Puehler F, Haegebarth A, Scholz A, Hoffmann J, Mumberg D, Ziegelbauer K. Combination of PI3K inhibitor BAY 80-6946 with allosteric MEK inhibitor BAY 86-9766 (RDEA119) and with erlotinib for the treatment of non-small-cell lung cancer. EJC Supplements. 2010;8:50–50.
    1. Iverson C, Larson G, Lai C, Yeh LT, Dadson C, Weingarten P, Appleby T, Vo T, Maderna A, Vernier JM, Hamatake R, Miner JN, Quart B. RDEA119/BAY 869766: a potent, selective, allosteric inhibitor of MEK1/2 for the treatment of cancer. Cancer Res. 2009;69:6839–47.
    1. Daemen A, Griffith OL, Heiser LM, Wang NJ, Enache OM, Sanborn Z, Pepin F, Durinck S, Korkola JE, Griffith M, Hur JS, Huh N, Chung J, et al. Modeling precision treatment of breast cancer. Genome Biol. 2013;14:R110. .
    1. Ventura AP, Radhakrishnan S, Green A, Rajaram SK, Allen AN, O’Briant K, Schummer M, Karlan B, Urban N, Tewari M, Drescher C, Knudsen BS. Activation of the MEK-S6 pathway in high-grade ovarian cancers. Appl Immunohistochem Mol Morphol. 2010;18:499–508. .
    1. She QB, Chandarlapaty S, Ye Q, Lobo J, Haskell KM, Leander KR, DeFeo-Jones D, Huber HE, Rosen N. Breast tumor cells with PI3K mutation or HER2 amplification are selectively addicted to Akt signaling. PLoS One. 2008;3:e3065. .
    1. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, Cameron D, Dowsett M, Barrios CH, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659–72. .
    1. Andre F, O’Regan R, Ozguroglu M, Toi M, Xu B, Jerusalem G, Masuda N, Wilks S, Arena F, Isaacs C, Yap YS, Papai Z, Lang I, et al. Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2014;15:580–91. .
    1. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, Wilson CJ, Lehar J, Kryukov GV, Sonkin D, Reddy A, Liu M, Murray L, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7. .
    1. Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, Greninger P, Thompson IR, Luo X, Soares J, Liu Q, Iorio F, Surdez D, et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature. 2012;483:570–5. .
    1. Yang W, Soares J, Greninger P, Edelman EJ, Lightfoot H, Forbes S, Bindal N, Beare D, Smith JA, Thompson IR, Ramaswamy S, Futreal PA, Haber DA, et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41:D955–61. .
    1. Schmieder R, Puehler F, Neuhaus R, Kissel M, Adjei AA, Miner JN, Mumberg D, Ziegelbauer K, Scholz A. Allosteric MEK1/2 inhibitor refametinib (BAY 86-9766) in combination with sorafenib exhibits antitumor activity in preclinical murine and rat models of hepatocellular carcinoma. Neoplasia. 2013;15:1161–71.
    1. Weekes CD, Von Hoff DD, Adjei AA, Leffingwell DP, Eckhardt SG, Gore L, Lewis KD, Weiss GJ, Ramanathan RK, Dy GK, Ma WW, Sheedy B, Iverson C, et al. Multicenter phase I trial of the mitogen-activated protein kinase 1/2 inhibitor BAY 86-9766 in patients with advanced cancer. Clin Cancer Res. 2013;19:1232–43. .
    1. Adjei AA, Richards DA, El-Khoueiry A, Braiteh F, Becerra CH, Stephenson JJ, Jr, Hezel AF, Sherman M, Garbo L, Leffingwell DP, Iverson C, Miner JN, Shen Z, et al. A phase I study of the safety, pharmacokinetics, and pharmacodynamics of combination therapy with refametinib plus sorafenib in patients with advanced cancer. Clin Cancer Res. 2016;22:2368–76. .
    1. Ramanathan RK, Von Hoff DD, Eskens F, Blumenschein GR, Richards DA, Renshaw FG, Rajagopalan P, Kelly A, Pena CE, Mross KB. A phase 1b trial of PI3K inhibitor copanlisib (BAY 80-6946) combined with the allosteric-MEK inhibitor refametinib (BAY 86-9766) in patients with advanced cancer. Cancer Res. 2014;32:5s, (suppl; abstr 2588).
    1. Van Laethem J, Jassem J, Heinemann V, Weekes C, Bridgewater J, Cascinu S, Melichar B, Peeters M, Ross P, Saramak P, Giurescu M, Garosi V, Roth K, et al. Phase II study of refametinib (BAY 86-9766), an allosteric dual MEK 1/2 inhibitor, and gemcitabine in patients with unresectable, locally advanced, or metastatic pancreatic cancer. J Clin Oncol. 2014;32:5s, (suppl; abstr 4025).
    1. Henjes F, Bender C, von der Heyde S, Braun L, Mannsperger HA, Schmidt C, Wiemann S, Hasmann M, Aulmann S, Beissbarth T, Korf U. Strong EGFR signaling in cell line models of ERBB2-amplified breast cancer attenuates response towards ERBB2-targeting drugs. Oncogenesis. 2012;1:e16. .
    1. Carlino MS, Todd JR, Gowrishankar K, Mijatov B, Pupo GM, Fung C, Snoyman S, Hersey P, Long GV, Kefford RF, Rizos H. Differential activity of MEK and ERK inhibitors in BRAF inhibitor resistant melanoma. Mol Oncol. 2014;8:544–54. .
    1. Fattore L, Marra E, Pisanu ME, Noto A, de Vitis C, Belleudi F, Aurisicchio L, Mancini R, Torrisi MR, Ascierto PA, Ciliberto G. Activation of an early feedback survival loop involving phospho-ErbB3 is a general response of melanoma cells to RAF/MEK inhibition and is abrogated by anti-ErbB3 antibodies. J Transl Med. 2013;11:180. .
    1. Petricoin EF, 3rd, Espina V, Araujo RP, Midura B, Yeung C, Wan X, Eichler GS, Johann DJ, Jr, Qualman S, Tsokos M, Krishnan K, Helman LJ, Liotta LA. Phosphoprotein pathway mapping: Akt/mammalian target of rapamycin activation is negatively associated with childhood rhabdomyosarcoma survival. Cancer Res. 2007;67:3431–40. .
    1. von der Heyde S, Bender C, Henjes F, Sonntag J, Korf U, Beissbarth T. Boolean ErbB network reconstructions and perturbation simulations reveal individual drug response in different breast cancer cell lines. BMC Syst Biol. 2014;8:75. .

Source: PubMed

3
Subscribe