Skip to main content
Top
Published in: Cancer Cell International 1/2019

Open Access 01-12-2019 | Ovarian Cancer | Primary research

Markers of MEK inhibitor resistance in low-grade serous ovarian cancer: EGFR is a potential therapeutic target

Authors: Marta Llaurado Fernandez, Amy Dawson, Joshua Hoenisch, Hannah Kim, Sylvia Bamford, Clara Salamanca, Gabriel DiMattia, Trevor Shepherd, Mattia Cremona, Bryan Hennessy, Shawn Anderson, Stanislav Volik, Colin C. Collins, David G. Huntsman, Mark S. Carey

Published in: Cancer Cell International | Issue 1/2019

Login to get access

Abstract

Background

Although low-grade serous ovarian cancer (LGSC) is rare, case-fatality rates are high as most patients present with advanced disease and current cytotoxic therapies are not overly effective. Recognizing that these cancers may be driven by MAPK pathway activation, MEK inhibitors (MEKi) are being tested in clinical trials. LGSC respond to MEKi only in a subgroup of patients, so predictive biomarkers and better therapies will be needed.

Methods

We evaluated a number of patient-derived LGSC cell lines, previously classified according to their MEKi sensitivity. Two cell lines were genomically compared against their matching tumors samples. MEKi-sensitive and MEKi-resistant lines were compared using whole exome sequencing and reverse phase protein array. Two treatment combinations targeting MEKi resistance markers were also evaluated using cell proliferation, cell viability, cell signaling, and drug synergism assays.

Results

Low-grade serous ovarian cancer cell lines recapitulated the genomic aberrations from their matching tumor samples. We identified three potential predictive biomarkers that distinguish MEKi sensitive and resistant lines: KRAS mutation status, and EGFR and PKC-alpha protein expression. The biomarkers were validated in three newly developed LGSC cell lines. Sub-lethal combination of MEK and EGFR inhibition showed drug synergy and caused complete cell death in two of four MEKi-resistant cell lines tested.

Conclusions

KRAS mutations and the protein expression of EGFR and PKC-alpha should be evaluated as predictive biomarkers in patients with LGSC treated with MEKi. Combination therapy using a MEKi with EGFR inhibition may represent a promising new therapy for patients with MEKi-resistant LGSC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Society AC. Cancer facts and figures 2017. Society AC. Cancer facts and figures 2017.
2.
go back to reference Society CC. Canadian cancer statistics 2017. Society CC. Canadian cancer statistics 2017.
3.
go back to reference Matsuo K, Machida H, Grubbs BH, Sood AK, Gershenson DM. Trends of low-grade serous ovarian carcinoma in the United States. J Gynecol Oncol. 2018;29:e15.CrossRef Matsuo K, Machida H, Grubbs BH, Sood AK, Gershenson DM. Trends of low-grade serous ovarian carcinoma in the United States. J Gynecol Oncol. 2018;29:e15.CrossRef
4.
go back to reference Gourley C, Farley J, Provencher DM, Pignata S, Mileshkin L, Harter P, Maenpaa J, Kim JW, Pujaide-Lauraine E, Glasspool RM, Ray-Coquard I, Gershenson D. Gynecologic Cancer InterGroup (GCIG) consensus review for ovarian and primary peritoneal low-grade serous carcinomas. Int J Gynecol Cancer. 2014;24:S9–13.CrossRef Gourley C, Farley J, Provencher DM, Pignata S, Mileshkin L, Harter P, Maenpaa J, Kim JW, Pujaide-Lauraine E, Glasspool RM, Ray-Coquard I, Gershenson D. Gynecologic Cancer InterGroup (GCIG) consensus review for ovarian and primary peritoneal low-grade serous carcinomas. Int J Gynecol Cancer. 2014;24:S9–13.CrossRef
5.
go back to reference Grabowski JP, Harter P, Heitz F, Pujade-Lauraine E, Reuss A, Kristensen G, Ray-Coquard I, Heitz J, Traut A, Pfisterer J, du Bois A. Operability and chemotherapy responsiveness in advanced low-grade serous ovarian cancer. An analysis of the AGO Study Group metadatabase. Gynecol Oncol. 2016;140:457–62.CrossRef Grabowski JP, Harter P, Heitz F, Pujade-Lauraine E, Reuss A, Kristensen G, Ray-Coquard I, Heitz J, Traut A, Pfisterer J, du Bois A. Operability and chemotherapy responsiveness in advanced low-grade serous ovarian cancer. An analysis of the AGO Study Group metadatabase. Gynecol Oncol. 2016;140:457–62.CrossRef
6.
go back to reference Gershenson DM. Low-grade serous carcinoma of the ovary or peritoneum. Ann Oncol. 2016;27(Suppl 1):i45–9.CrossRef Gershenson DM. Low-grade serous carcinoma of the ovary or peritoneum. Ann Oncol. 2016;27(Suppl 1):i45–9.CrossRef
7.
go back to reference Kobel M, Kalloger SE, Boyd N, McKinney S, Mehl E, Palmer C, Leung S, Bowen NJ, Ionescu DN, Rajput A, Prentice LM, Miller D, Santos J, Swenerton K, Gilks CB, Huntsman D. Ovarian carcinoma subtypes are different diseases: implications for biomarker studies. PLoS Med. 2008;5:e232.CrossRef Kobel M, Kalloger SE, Boyd N, McKinney S, Mehl E, Palmer C, Leung S, Bowen NJ, Ionescu DN, Rajput A, Prentice LM, Miller D, Santos J, Swenerton K, Gilks CB, Huntsman D. Ovarian carcinoma subtypes are different diseases: implications for biomarker studies. PLoS Med. 2008;5:e232.CrossRef
8.
go back to reference Kurman RJ, Shih Ie M. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer—shifting the paradigm. Hum Pathol. 2011;42:918–31.CrossRef Kurman RJ, Shih Ie M. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer—shifting the paradigm. Hum Pathol. 2011;42:918–31.CrossRef
9.
go back to reference Singer G, Stohr R, Cope L, Dehari R, Hartmann A, Cao DF, Wang TL, Kurman RJ, Shih Ie M. Patterns of p53 mutations separate ovarian serous borderline tumors and low- and high-grade carcinomas and provide support for a new model of ovarian carcinogenesis: a mutational analysis with immunohistochemical correlation. Am J Surg Pathol. 2005;29:218–24.CrossRef Singer G, Stohr R, Cope L, Dehari R, Hartmann A, Cao DF, Wang TL, Kurman RJ, Shih Ie M. Patterns of p53 mutations separate ovarian serous borderline tumors and low- and high-grade carcinomas and provide support for a new model of ovarian carcinogenesis: a mutational analysis with immunohistochemical correlation. Am J Surg Pathol. 2005;29:218–24.CrossRef
10.
go back to reference Cancer Genome Atlas Research N. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–15.CrossRef Cancer Genome Atlas Research N. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–15.CrossRef
11.
go back to reference Wong KK, Lu KH, Malpica A, Bodurka DC, Shvartsman HS, Schmandt RE, Thornton AD, Deavers MT, Silva EG, Gershenson DM. Significantly greater expression of ER, PR, and ECAD in advanced-stage low-grade ovarian serous carcinoma as revealed by immunohistochemical analysis. Int J Gynecol Pathol. 2007;26:404–9.CrossRef Wong KK, Lu KH, Malpica A, Bodurka DC, Shvartsman HS, Schmandt RE, Thornton AD, Deavers MT, Silva EG, Gershenson DM. Significantly greater expression of ER, PR, and ECAD in advanced-stage low-grade ovarian serous carcinoma as revealed by immunohistochemical analysis. Int J Gynecol Pathol. 2007;26:404–9.CrossRef
12.
go back to reference Ayhan A, Kurman RJ, Yemelyanova A, Vang R, Logani S, Seidman JD, Shih Ie M. Defining the cut point between low-grade and high-grade ovarian serous carcinomas: a clinicopathologic and molecular genetic analysis. Am J Surg Pathol. 2009;33:1220–4.CrossRef Ayhan A, Kurman RJ, Yemelyanova A, Vang R, Logani S, Seidman JD, Shih Ie M. Defining the cut point between low-grade and high-grade ovarian serous carcinomas: a clinicopathologic and molecular genetic analysis. Am J Surg Pathol. 2009;33:1220–4.CrossRef
13.
go back to reference Singer G, Oldt R 3rd, Cohen Y, Wang BG, Sidransky D, Kurman RJ, Shih Ie M. Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst. 2003;95:484–6.CrossRef Singer G, Oldt R 3rd, Cohen Y, Wang BG, Sidransky D, Kurman RJ, Shih Ie M. Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst. 2003;95:484–6.CrossRef
14.
go back to reference Tsang YT, Deavers MT, Sun CC, Kwan SY, Kuo E, Malpica A, Mok SC, Gershenson DM, Wong KK. KRAS (but not BRAF) mutations in ovarian serous borderline tumour are associated with recurrent low-grade serous carcinoma. J Pathol. 2013;231:449–56.CrossRef Tsang YT, Deavers MT, Sun CC, Kwan SY, Kuo E, Malpica A, Mok SC, Gershenson DM, Wong KK. KRAS (but not BRAF) mutations in ovarian serous borderline tumour are associated with recurrent low-grade serous carcinoma. J Pathol. 2013;231:449–56.CrossRef
15.
go back to reference Jones S, Wang TL, Kurman RJ, Nakayama K, Velculescu VE, Vogelstein B, Kinzler KW, Papadopoulos N, Shih Ie M. Low-grade serous carcinomas of the ovary contain very few point mutations. J Pathol. 2012;226:413–20.CrossRef Jones S, Wang TL, Kurman RJ, Nakayama K, Velculescu VE, Vogelstein B, Kinzler KW, Papadopoulos N, Shih Ie M. Low-grade serous carcinomas of the ovary contain very few point mutations. J Pathol. 2012;226:413–20.CrossRef
16.
go back to reference Malpica A, Wong KK. The molecular pathology of ovarian serous borderline tumors. Ann Oncol. 2016;27(Suppl 1):i16–9.CrossRef Malpica A, Wong KK. The molecular pathology of ovarian serous borderline tumors. Ann Oncol. 2016;27(Suppl 1):i16–9.CrossRef
17.
go back to reference Grisham RN, Iyer G. Low-grade serous ovarian cancer: current treatment paradigms and future directions. Curr Treat Options Oncol. 2018;19:54.CrossRef Grisham RN, Iyer G. Low-grade serous ovarian cancer: current treatment paradigms and future directions. Curr Treat Options Oncol. 2018;19:54.CrossRef
18.
go back to reference Emmanuel C, Chiew YE, George J, Etemadmoghadam D, Anglesio MS, Sharma R, Russell P, Kennedy C, Fereday S, Hung J, Galletta L, Hogg R, Wain GV, Brand A, Balleine R, MacConaill L, Palescandolo E, Hunter SM, Campbell I, Dobrovic A, Wong SQ, Do H, Clarke CL, Harnett PR, Bowtell DD, deFazio A, Australian Ovarian Cancer S. Genomic classification of serous ovarian cancer with adjacent borderline differentiates RAS pathway and TP53-mutant tumors and identifies NRAS as an oncogenic driver. Clin Cancer Res. 2014;20:6618–30.CrossRef Emmanuel C, Chiew YE, George J, Etemadmoghadam D, Anglesio MS, Sharma R, Russell P, Kennedy C, Fereday S, Hung J, Galletta L, Hogg R, Wain GV, Brand A, Balleine R, MacConaill L, Palescandolo E, Hunter SM, Campbell I, Dobrovic A, Wong SQ, Do H, Clarke CL, Harnett PR, Bowtell DD, deFazio A, Australian Ovarian Cancer S. Genomic classification of serous ovarian cancer with adjacent borderline differentiates RAS pathway and TP53-mutant tumors and identifies NRAS as an oncogenic driver. Clin Cancer Res. 2014;20:6618–30.CrossRef
19.
go back to reference Hunter SM, Anglesio MS, Ryland GL, Sharma R, Chiew YE, Rowley SM, Doyle MA, Li J, Gilks CB, Moss P, Allan PE, Stephens AN, Huntsman DG, deFazio A, Bowtell DD, Australian Ovarian Cancer Study G, Gorringe KL, Campbell IG. Molecular profiling of low grade serous ovarian tumours identifies novel candidate driver genes. Oncotarget. 2015;6(35):37663–77.CrossRef Hunter SM, Anglesio MS, Ryland GL, Sharma R, Chiew YE, Rowley SM, Doyle MA, Li J, Gilks CB, Moss P, Allan PE, Stephens AN, Huntsman DG, deFazio A, Bowtell DD, Australian Ovarian Cancer Study G, Gorringe KL, Campbell IG. Molecular profiling of low grade serous ovarian tumours identifies novel candidate driver genes. Oncotarget. 2015;6(35):37663–77.CrossRef
20.
go back to reference Xing D, Suryo Rahmanto Y, Zeppernick F, Hannibal CG, Kjaer SK, Vang R, Shih IM, Wang TL. Mutation of NRAS is a rare genetic event in ovarian low-grade serous carcinoma. Hum Pathol. 2017;68:87–91.CrossRef Xing D, Suryo Rahmanto Y, Zeppernick F, Hannibal CG, Kjaer SK, Vang R, Shih IM, Wang TL. Mutation of NRAS is a rare genetic event in ovarian low-grade serous carcinoma. Hum Pathol. 2017;68:87–91.CrossRef
21.
go back to reference Hsu CY, Bristow R, Cha MS, Wang BG, Ho CL, Kurman RJ, Wang TL, Shih Ie M. Characterization of active mitogen-activated protein kinase in ovarian serous carcinomas. Clin Cancer Res. 2004;10:6432–6.CrossRef Hsu CY, Bristow R, Cha MS, Wang BG, Ho CL, Kurman RJ, Wang TL, Shih Ie M. Characterization of active mitogen-activated protein kinase in ovarian serous carcinomas. Clin Cancer Res. 2004;10:6432–6.CrossRef
22.
go back to reference Farley J, Brady WE, Vathipadiekal V, Lankes HA, Coleman R, Morgan MA, Mannel R, Yamada SD, Mutch D, Rodgers WH, Birrer M, Gershenson DM. Selumetinib in women with recurrent low-grade serous carcinoma of the ovary or peritoneum: an open-label, single-arm, phase 2 study. Lancet Oncol. 2013;14:134–40.CrossRef Farley J, Brady WE, Vathipadiekal V, Lankes HA, Coleman R, Morgan MA, Mannel R, Yamada SD, Mutch D, Rodgers WH, Birrer M, Gershenson DM. Selumetinib in women with recurrent low-grade serous carcinoma of the ovary or peritoneum: an open-label, single-arm, phase 2 study. Lancet Oncol. 2013;14:134–40.CrossRef
23.
go back to reference Han C, Bellone S, Zammataro L, Schwartz PE, Santin AD. Binimetinib (MEK162) in recurrent low-grade serous ovarian cancer resistant to chemotherapy and hormonal treatment. Gynecol Oncol Rep. 2018;25:41–4.CrossRef Han C, Bellone S, Zammataro L, Schwartz PE, Santin AD. Binimetinib (MEK162) in recurrent low-grade serous ovarian cancer resistant to chemotherapy and hormonal treatment. Gynecol Oncol Rep. 2018;25:41–4.CrossRef
24.
go back to reference Fernandez ML, DiMattia GE, Dawson A, Bamford S, Anderson S, Hennessy BT, Anglesio MS, Shepherd TG, Salamanca C, Hoenisch J, Tinker A, Huntsman DG, Carey MS. Differences in MEK inhibitor efficacy in molecularly characterized low-grade serous ovarian cancer cell lines. Am J Cancer Res. 2016;6:2235–51.PubMedPubMedCentral Fernandez ML, DiMattia GE, Dawson A, Bamford S, Anderson S, Hennessy BT, Anglesio MS, Shepherd TG, Salamanca C, Hoenisch J, Tinker A, Huntsman DG, Carey MS. Differences in MEK inhibitor efficacy in molecularly characterized low-grade serous ovarian cancer cell lines. Am J Cancer Res. 2016;6:2235–51.PubMedPubMedCentral
25.
go back to reference Chahal M, Pleasance E, Grewal J, Zhao E, Ng T, Chapman E, Jones MR, Shen Y, Mungall KL, Bonakdar M, Taylor GA, Ma Y, Mungall AJ, Moore RA, Lim H, Renouf D, Yip S, Jones SJM, Marra MA, Laskin J. Personalized oncogenomic analysis of metastatic adenoid cystic carcinoma: using whole-genome sequencing to inform clinical decision-making. Cold Spring Harb Mol Case Stud. 2018;4:a002626.CrossRef Chahal M, Pleasance E, Grewal J, Zhao E, Ng T, Chapman E, Jones MR, Shen Y, Mungall KL, Bonakdar M, Taylor GA, Ma Y, Mungall AJ, Moore RA, Lim H, Renouf D, Yip S, Jones SJM, Marra MA, Laskin J. Personalized oncogenomic analysis of metastatic adenoid cystic carcinoma: using whole-genome sequencing to inform clinical decision-making. Cold Spring Harb Mol Case Stud. 2018;4:a002626.CrossRef
26.
go back to reference Tomkinson H, McBride E, Martin P, Lisbon E, Dymond AW, Cantarini M, So K, Holt D. Comparison of the pharmacokinetics of the phase II and phase III capsule formulations of selumetinib and the effects of food on exposure: results from two randomized crossover trials in healthy male subjects. Clin Ther. 2017;39(2260–2275):e2261. Tomkinson H, McBride E, Martin P, Lisbon E, Dymond AW, Cantarini M, So K, Holt D. Comparison of the pharmacokinetics of the phase II and phase III capsule formulations of selumetinib and the effects of food on exposure: results from two randomized crossover trials in healthy male subjects. Clin Ther. 2017;39(2260–2275):e2261.
27.
go back to reference Infante JR, Papadopoulos KP, Bendell JC, Patnaik A, Burris HA 3rd, Rasco D, Jones SF, Smith L, Cox DS, Durante M, Bellew KM, Park JJ, Le NT, Tolcher AW. A phase 1b study of trametinib, an oral Mitogen-activated protein kinase kinase (MEK) inhibitor, in combination with gemcitabine in advanced solid tumours. Eur J Cancer. 2013;49:2077–85.CrossRef Infante JR, Papadopoulos KP, Bendell JC, Patnaik A, Burris HA 3rd, Rasco D, Jones SF, Smith L, Cox DS, Durante M, Bellew KM, Park JJ, Le NT, Tolcher AW. A phase 1b study of trametinib, an oral Mitogen-activated protein kinase kinase (MEK) inhibitor, in combination with gemcitabine in advanced solid tumours. Eur J Cancer. 2013;49:2077–85.CrossRef
28.
go back to reference Tibes R, Qiu Y, Lu Y, Hennessy B, Andreeff M, Mills GB, Kornblau SM. Reverse phase protein array: validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells. Mol Cancer Ther. 2006;5:2512–21.CrossRef Tibes R, Qiu Y, Lu Y, Hennessy B, Andreeff M, Mills GB, Kornblau SM. Reverse phase protein array: validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells. Mol Cancer Ther. 2006;5:2512–21.CrossRef
29.
go back to reference Aslan O, Cremona M, Morgan C, Cheung LW, Mills GB, Hennessy BT. Preclinical evaluation and reverse phase protein array-based profiling of PI3K and MEK inhibitors in endometrial carcinoma in vitro. BMC Cancer. 2018;18:168.CrossRef Aslan O, Cremona M, Morgan C, Cheung LW, Mills GB, Hennessy BT. Preclinical evaluation and reverse phase protein array-based profiling of PI3K and MEK inhibitors in endometrial carcinoma in vitro. BMC Cancer. 2018;18:168.CrossRef
30.
go back to reference Wiegand KC, Hennessy BT, Leung S, Wang Y, Ju Z, McGahren M, Kalloger SE, Finlayson S, Stemke-Hale K, Lu Y, Zhang F, Anglesio MS, Gilks B, Mills GB, Huntsman DG, Carey MS. A functional proteogenomic analysis of endometrioid and clear cell carcinomas using reverse phase protein array and mutation analysis: protein expression is histotype-specific and loss of ARID1A/BAF250a is associated with AKT phosphorylation. BMC Cancer. 2014;14:120.CrossRef Wiegand KC, Hennessy BT, Leung S, Wang Y, Ju Z, McGahren M, Kalloger SE, Finlayson S, Stemke-Hale K, Lu Y, Zhang F, Anglesio MS, Gilks B, Mills GB, Huntsman DG, Carey MS. A functional proteogenomic analysis of endometrioid and clear cell carcinomas using reverse phase protein array and mutation analysis: protein expression is histotype-specific and loss of ARID1A/BAF250a is associated with AKT phosphorylation. BMC Cancer. 2014;14:120.CrossRef
31.
go back to reference Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA. 2001;98:5116–21.CrossRef Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA. 2001;98:5116–21.CrossRef
32.
go back to reference 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.CrossRef 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.CrossRef
33.
go back to reference Corcoran RB, Andre T, Atreya CE, Schellens JHM, Yoshino T, Bendell JC, Hollebecque A, McRee AJ, Siena S, Middleton G, Muro K, Gordon MS, Tabernero J, Yaeger R, O’Dwyer PJ, Humblet Y, De Vos F, Jung AS, Brase JC, Jaeger S, Bettinger S, Mookerjee B, Rangwala F, Van Cutsem E. Combined BRAF, EGFR, and MEK inhibition in patients with BRAF(V600E)-mutant colorectal cancer. Cancer Discov. 2018;8:428–43.CrossRef Corcoran RB, Andre T, Atreya CE, Schellens JHM, Yoshino T, Bendell JC, Hollebecque A, McRee AJ, Siena S, Middleton G, Muro K, Gordon MS, Tabernero J, Yaeger R, O’Dwyer PJ, Humblet Y, De Vos F, Jung AS, Brase JC, Jaeger S, Bettinger S, Mookerjee B, Rangwala F, Van Cutsem E. Combined BRAF, EGFR, and MEK inhibition in patients with BRAF(V600E)-mutant colorectal cancer. Cancer Discov. 2018;8:428–43.CrossRef
34.
go back to reference Kim JY, Welsh EA, Fang B, Bai Y, Kinose F, Eschrich SA, Koomen JM, Haura EB. Phosphoproteomics reveals MAPK inhibitors enhance MET- and EGFR-driven AKT signaling in KRAS-mutant lung cancer. Mol Cancer Res. 2016;14:1019–29.CrossRef Kim JY, Welsh EA, Fang B, Bai Y, Kinose F, Eschrich SA, Koomen JM, Haura EB. Phosphoproteomics reveals MAPK inhibitors enhance MET- and EGFR-driven AKT signaling in KRAS-mutant lung cancer. Mol Cancer Res. 2016;14:1019–29.CrossRef
35.
go back to reference Li J, Sordella R, Powers S. Effectors and potential targets selectively upregulated in human KRAS-mutant lung adenocarcinomas. Sci Rep. 2016;6:27891.CrossRef Li J, Sordella R, Powers S. Effectors and potential targets selectively upregulated in human KRAS-mutant lung adenocarcinomas. Sci Rep. 2016;6:27891.CrossRef
36.
go back to reference Kim CW, Asai D, Kang JH, Kishimura A, Mori T, Katayama Y. Reversal of efflux of an anticancer drug in human drug-resistant breast cancer cells by inhibition of protein kinase Calpha (PKCalpha) activity. Tumour Biol. 2016;37:1901–8.CrossRef Kim CW, Asai D, Kang JH, Kishimura A, Mori T, Katayama Y. Reversal of efflux of an anticancer drug in human drug-resistant breast cancer cells by inhibition of protein kinase Calpha (PKCalpha) activity. Tumour Biol. 2016;37:1901–8.CrossRef
37.
go back to reference Muscella A, Vetrugno C, Antonaci G, Cossa LG, Marsigliante S. PKC-delta/PKC-alpha activity balance regulates the lethal effects of cisplatin. Biochem Pharmacol. 2015;98:29–40.CrossRef Muscella A, Vetrugno C, Antonaci G, Cossa LG, Marsigliante S. PKC-delta/PKC-alpha activity balance regulates the lethal effects of cisplatin. Biochem Pharmacol. 2015;98:29–40.CrossRef
38.
go back to reference Momeny M, Yousefi H, Eyvani H, Moghaddaskho F, Salehi A, Esmaeili F, Alishahi Z, Barghi F, Vaezijoze S, Shamsaiegahkani S, Zarrinrad G, Sankanian G, Sabourinejad Z, Hamzehlou S, Bashash D, Aboutorabi ES, Ghaffari P, Dehpour AR, Tavangar SM, Tavakkoly-Bazzaz J, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Blockade of nuclear factor-kappaB (NF-kappaB) pathway inhibits growth and induces apoptosis in chemoresistant ovarian carcinoma cells. Int J Biochem Cell Biol. 2018;99:1–9.CrossRef Momeny M, Yousefi H, Eyvani H, Moghaddaskho F, Salehi A, Esmaeili F, Alishahi Z, Barghi F, Vaezijoze S, Shamsaiegahkani S, Zarrinrad G, Sankanian G, Sabourinejad Z, Hamzehlou S, Bashash D, Aboutorabi ES, Ghaffari P, Dehpour AR, Tavangar SM, Tavakkoly-Bazzaz J, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Blockade of nuclear factor-kappaB (NF-kappaB) pathway inhibits growth and induces apoptosis in chemoresistant ovarian carcinoma cells. Int J Biochem Cell Biol. 2018;99:1–9.CrossRef
39.
go back to reference Kankia IH, Khalil HS, Langdon SP, Moult PR, Bown JL, Deeni YY. NRF2 regulates HER1 signaling pathway to modulate the sensitivity of ovarian cancer cells to lapatinib and erlotinib. Oxid Med Cell Longev. 2017;2017:1864578.CrossRef Kankia IH, Khalil HS, Langdon SP, Moult PR, Bown JL, Deeni YY. NRF2 regulates HER1 signaling pathway to modulate the sensitivity of ovarian cancer cells to lapatinib and erlotinib. Oxid Med Cell Longev. 2017;2017:1864578.CrossRef
40.
go back to reference Yeo WL, Riely GJ, Yeap BY, Lau MW, Warner JL, Bodio K, Huberman MS, Kris MG, Tenen DG, Pao W, Kobayashi S, Costa DB. Erlotinib at a dose of 25 mg daily for non-small cell lung cancers with EGFR mutations. J Thorac Oncol. 2010;5:1048–53.CrossRef Yeo WL, Riely GJ, Yeap BY, Lau MW, Warner JL, Bodio K, Huberman MS, Kris MG, Tenen DG, Pao W, Kobayashi S, Costa DB. Erlotinib at a dose of 25 mg daily for non-small cell lung cancers with EGFR mutations. J Thorac Oncol. 2010;5:1048–53.CrossRef
42.
go back to reference Safaee Ardekani G, Jafarnejad SM, Tan L, Saeedi A, Li G. The prognostic value of BRAF mutation in colorectal cancer and melanoma: a systematic review and meta-analysis. PLoS ONE. 2012;7:e47054.CrossRef Safaee Ardekani G, Jafarnejad SM, Tan L, Saeedi A, Li G. The prognostic value of BRAF mutation in colorectal cancer and melanoma: a systematic review and meta-analysis. PLoS ONE. 2012;7:e47054.CrossRef
43.
go back to reference Cheng Y, Tian H. Current development status of MEK inhibitors. Molecules. 2017;22(10):1551.CrossRef Cheng Y, Tian H. Current development status of MEK inhibitors. Molecules. 2017;22(10):1551.CrossRef
45.
go back to reference Gershenson DM, Sun CC, Bodurka D, Coleman RL, Lu KH, Sood AK, Deavers M, Malpica AL, Kavanagh JJ. Recurrent low-grade serous ovarian carcinoma is relatively chemoresistant. Gynecol Oncol. 2009;114:48–52.CrossRef Gershenson DM, Sun CC, Bodurka D, Coleman RL, Lu KH, Sood AK, Deavers M, Malpica AL, Kavanagh JJ. Recurrent low-grade serous ovarian carcinoma is relatively chemoresistant. Gynecol Oncol. 2009;114:48–52.CrossRef
46.
go back to reference Grisham RN, Sylvester BE, Won H, McDermott G, DeLair D, Ramirez R, Yao Z, Shen R, Dao F, Bogomolniy F, Makker V, Sala E, Soumerai TE, Hyman DM, Socci ND, Viale A, Gershenson DM, Farley J, Levine DA, Rosen N, Berger MF, Spriggs DR, Aghajanian CA, Solit DB, Iyer G. Extreme outlier analysis identifies occult mitogen-activated protein kinase pathway mutations in patients with low-grade serous ovarian cancer. J Clin Oncol. 2015;33(34):4099–105.CrossRef Grisham RN, Sylvester BE, Won H, McDermott G, DeLair D, Ramirez R, Yao Z, Shen R, Dao F, Bogomolniy F, Makker V, Sala E, Soumerai TE, Hyman DM, Socci ND, Viale A, Gershenson DM, Farley J, Levine DA, Rosen N, Berger MF, Spriggs DR, Aghajanian CA, Solit DB, Iyer G. Extreme outlier analysis identifies occult mitogen-activated protein kinase pathway mutations in patients with low-grade serous ovarian cancer. J Clin Oncol. 2015;33(34):4099–105.CrossRef
47.
go back to reference Takekuma M, Wong KK, Coleman RL. A long-term surviving patient with recurrent low-grade serous ovarian carcinoma treated with the MEK1/2 inhibitor, selumetinib. Gynecol Oncol Res Pract. 2016;3:5.CrossRef Takekuma M, Wong KK, Coleman RL. A long-term surviving patient with recurrent low-grade serous ovarian carcinoma treated with the MEK1/2 inhibitor, selumetinib. Gynecol Oncol Res Pract. 2016;3:5.CrossRef
48.
go back to reference Altman AD, Nelson GS, Ghatage P, McIntyre JB, Capper D, Chu P, Nation JG, Karnezis AN, Han G, Kalloger SE, Kobel M. The diagnostic utility of TP53 and CDKN2A to distinguish ovarian high-grade serous carcinoma from low-grade serous ovarian tumors. Mod Pathol. 2013;26:1255–63.CrossRef Altman AD, Nelson GS, Ghatage P, McIntyre JB, Capper D, Chu P, Nation JG, Karnezis AN, Han G, Kalloger SE, Kobel M. The diagnostic utility of TP53 and CDKN2A to distinguish ovarian high-grade serous carcinoma from low-grade serous ovarian tumors. Mod Pathol. 2013;26:1255–63.CrossRef
49.
go back to reference Kuo KT, Guan B, Feng Y, Mao TL, Chen X, Jinawath N, Wang Y, Kurman RJ, Shih Ie M, Wang TL. Analysis of DNA copy number alterations in ovarian serous tumors identifies new molecular genetic changes in low-grade and high-grade carcinomas. Cancer Res. 2009;69:4036–42.CrossRef Kuo KT, Guan B, Feng Y, Mao TL, Chen X, Jinawath N, Wang Y, Kurman RJ, Shih Ie M, Wang TL. Analysis of DNA copy number alterations in ovarian serous tumors identifies new molecular genetic changes in low-grade and high-grade carcinomas. Cancer Res. 2009;69:4036–42.CrossRef
50.
go back to reference McIntyre JB, Rambau PF, Chan A, Yap S, Morris D, Nelson GS, Kobel M. Molecular alterations in indolent, aggressive and recurrent ovarian low-grade serous carcinoma. Histopathology. 2017;70:347–58.CrossRef McIntyre JB, Rambau PF, Chan A, Yap S, Morris D, Nelson GS, Kobel M. Molecular alterations in indolent, aggressive and recurrent ovarian low-grade serous carcinoma. Histopathology. 2017;70:347–58.CrossRef
51.
go back to reference Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A, Bernards R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100–3.CrossRef Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A, Bernards R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100–3.CrossRef
52.
go back to reference Whittaker SR, Cowley GS, Wagner S, Luo F, Root DE, Garraway LA. Combined Pan-RAF and mek inhibition overcomes multiple resistance mechanisms to selective RAF inhibitors. Mol Cancer Ther. 2015;14:2700–11.CrossRef Whittaker SR, Cowley GS, Wagner S, Luo F, Root DE, Garraway LA. Combined Pan-RAF and mek inhibition overcomes multiple resistance mechanisms to selective RAF inhibitors. Mol Cancer Ther. 2015;14:2700–11.CrossRef
53.
go back to reference Carter CA, Rajan A, Keen C, Szabo E, Khozin S, Thomas A, Brzezniak C, Guha U, Doyle LA, Steinberg SM, Xi L, Raffeld M, Tomita Y, Lee MJ, Lee S, Trepel JB, Reckamp KL, Koehler S, Gitlitz B, Salgia R, Gandara D, Vokes E, Giaccone G. Selumetinib with and without erlotinib in KRAS mutant and KRAS wild-type advanced nonsmall-cell lung cancer. Ann Oncol. 2016;27:693–9.CrossRef Carter CA, Rajan A, Keen C, Szabo E, Khozin S, Thomas A, Brzezniak C, Guha U, Doyle LA, Steinberg SM, Xi L, Raffeld M, Tomita Y, Lee MJ, Lee S, Trepel JB, Reckamp KL, Koehler S, Gitlitz B, Salgia R, Gandara D, Vokes E, Giaccone G. Selumetinib with and without erlotinib in KRAS mutant and KRAS wild-type advanced nonsmall-cell lung cancer. Ann Oncol. 2016;27:693–9.CrossRef
54.
go back to reference Corcoran RB, Ebi H, Turke AB, Coffee EM, Nishino M, Cogdill AP, Brown RD, Della Pelle P, Dias-Santagata D, Hung KE, Flaherty KT, Piris A, Wargo JA, Settleman J, Mino-Kenudson M, Engelman JA. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2012;2:227–35.CrossRef Corcoran RB, Ebi H, Turke AB, Coffee EM, Nishino M, Cogdill AP, Brown RD, Della Pelle P, Dias-Santagata D, Hung KE, Flaherty KT, Piris A, Wargo JA, Settleman J, Mino-Kenudson M, Engelman JA. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2012;2:227–35.CrossRef
55.
go back to reference Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O’Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT, McArthur GA, Group B-S. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–16.CrossRef Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O’Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT, McArthur GA, Group B-S. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–16.CrossRef
56.
go back to reference Kopetz S, Desai J, Chan E, Hecht JR, O’Dwyer PJ, Maru D, Morris V, Janku F, Dasari A, Chung W, Issa JP, Gibbs P, James B, Powis G, Nolop KB, Bhattacharya S, Saltz L. Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J Clin Oncol. 2015;33:4032–8.CrossRef Kopetz S, Desai J, Chan E, Hecht JR, O’Dwyer PJ, Maru D, Morris V, Janku F, Dasari A, Chung W, Issa JP, Gibbs P, James B, Powis G, Nolop KB, Bhattacharya S, Saltz L. Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J Clin Oncol. 2015;33:4032–8.CrossRef
57.
go back to reference Grisham RN, Iyer G, Garg K, Delair D, Hyman DM, Zhou Q, Iasonos A, Berger MF, Dao F, Spriggs DR, Levine DA, Aghajanian C, Solit DB. BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer. 2013;119:548–54.CrossRef Grisham RN, Iyer G, Garg K, Delair D, Hyman DM, Zhou Q, Iasonos A, Berger MF, Dao F, Spriggs DR, Levine DA, Aghajanian C, Solit DB. BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer. 2013;119:548–54.CrossRef
58.
go back to reference Wong KK, Tsang YT, Deavers MT, Mok SC, Zu Z, Sun C, Malpica A, Wolf JK, Lu KH, Gershenson DM. BRAF mutation is rare in advanced-stage low-grade ovarian serous carcinomas. Am J Pathol. 2010;177:1611–7.CrossRef Wong KK, Tsang YT, Deavers MT, Mok SC, Zu Z, Sun C, Malpica A, Wolf JK, Lu KH, Gershenson DM. BRAF mutation is rare in advanced-stage low-grade ovarian serous carcinomas. Am J Pathol. 2010;177:1611–7.CrossRef
Metadata
Title
Markers of MEK inhibitor resistance in low-grade serous ovarian cancer: EGFR is a potential therapeutic target
Authors
Marta Llaurado Fernandez
Amy Dawson
Joshua Hoenisch
Hannah Kim
Sylvia Bamford
Clara Salamanca
Gabriel DiMattia
Trevor Shepherd
Mattia Cremona
Bryan Hennessy
Shawn Anderson
Stanislav Volik
Colin C. Collins
David G. Huntsman
Mark S. Carey
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2019
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-019-0725-1

Other articles of this Issue 1/2019

Cancer Cell International 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine