Display options
Share it on

Mol Cancer Res. 2021 Mar;19(3):361-374. doi: 10.1158/1541-7786.MCR-20-0687. Epub 2020 Nov 02.

The MEK/ERK Network as a Therapeutic Target in Human Cancer.

Molecular cancer research : MCR

Renee Barbosa, Lucila A Acevedo, Ronen Marmorstein

Affiliations

  1. School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.
  2. Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
  3. Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
  4. Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. [email protected].

PMID: 33139506 PMCID: PMC7925338 DOI: 10.1158/1541-7786.MCR-20-0687

Abstract

The RAS-RAF-MEK-ERK pathway is the most well-studied of the MAPK cascades and is critical for cell proliferation, differentiation, and survival. Abnormalities in regulation resulting from mutations in components of this pathway, particularly in upstream proteins, RAS and RAF, are responsible for a significant fraction of human cancers and nearly all cutaneous melanomas. Activation of receptor tyrosine kinases by growth factors and various extracellular signals leads to the sequential activation of RAS, RAF, MEK, and finally ERK, which activates numerous transcription factors and facilitates oncogenesis in the case of aberrant pathway activation. While extensive studies have worked to elucidate the activation mechanisms and structural components of upstream MAPK components, comparatively less attention has been directed toward the kinases, MEK and ERK, due to the infrequency of oncogenic-activating mutations in these kinases. However, acquired drug resistance has become a major issue in the treatment of RAS- and RAF-mutated cancers. Targeting the terminal kinases in the MAPK cascade has shown promise for overcoming many of these resistance mechanisms and improving treatment options for patients with MAPK-aberrant cancers. Here, we will describe the role of MEK and ERK in MAPK signaling and summarize the current understanding of their interaction and activation mechanisms. We will also discuss existing approaches for targeting MEK and ERK, and the benefits of alternative strategies. Areas requiring further exploration will be highlighted to guide future research endeavors and aid in the development of alternative therapeutic strategies to combat surmounting drug resistance in treating MAPK-mediated cancers. VISUAL OVERVIEW: http://mcr.aacrjournals.org/content/molcanres/19/3/361/F1.large.jpg.

©2020 American Association for Cancer Research.

References

  1. Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 2007;26:3291–310. - PubMed
  2. Roskoski R. Allosteric MEK1/2 inhibitors including cobimetanib and trametinib in the treatment of cutaneous melanomas. Pharmacol Res. 2017;117:20–31. - PubMed
  3. Wortzel I, Seger R. The ERK cascade: distinct functions within various subcellular organelles. Genes Cancer. 2011;2:195–209. - PubMed
  4. Marais R, Light Y, Paterson HF, Marshall CJ. Ras recruits Raf-1 to the plasma membrane far activation by tyrosine phosphorylation. EMBO J. 1995;14:3136–45. - PubMed
  5. Macdonald SG, Crews CM, Wu L, Driller J, Clark R, Erikson RL, et al. Reconstitution of the Raf-1-MEK-ERK signal transduction pathway in vitro. Mol Cell Biol. 1993;13:6615–20. - PubMed
  6. Haling JR, Sudhamsu J, Yen I, Sideris S, Sandoval W, Phung W, et al. Structure of the BRAF-MEK complex reveals a kinase activity independent role for BRAF in MAPK signaling. Cancer Cell. 2014;26:402–13. - PubMed
  7. Seger R, Ahn NG, Posada J, Munar ES, Jensen AM, Cooper JA, et al. Purification and characterization of mitogen-activated protein kinase activator(s) from epidermal growth factor-stimulated A431 cells. J Biol Chem. 1992;267:14373–81. - PubMed
  8. Roskoski R. ERK1/2 MAP kinases: structure, function, and regulation. Pharmacol Res. 2012;66:105–43. - PubMed
  9. Nan X, Tamgüney TM, Collisson EA, Lin Li-J, Pitt C, Galeas J, et al. Ras-GTP dimers activate the mitogen-activated protein kinase (MAPK) pathway. Proc Natl Acad Sci U S A. 2015;112:7996–8001. - PubMed
  10. Yao Z, Torres NM, Tao A, Gao Y, Luo L, Li Qi, et al. BRAF mutants evade ERK-dependent feedback by different mechanisms that determine their sensitivity to pharmacologic inhibition. Cancer Cell. 2015;28:370–83. - PubMed
  11. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298:1912–34. - PubMed
  12. Burack WR, Sturgill TW. The activating dual phosphorylation of MAPK by MEK is nonprocessive. Biochemistry. 1997;36:5929–33. - PubMed
  13. Gonzalez FA, Raden DL, Davis RJ. Identification of substrate recognition determinants for human ERK1 and ERK2 protein kinases. J Biol Chem. 1991;266:22159–63. - PubMed
  14. Mebratu Y, Tesfaigzi Y. How ERK1/2 activation controls cell proliferation and cell death is subcellular localization the answer?. Cell Cycle. 2009;8:1168–75. - PubMed
  15. Roskoski R. MEK1/2 dual-specificity protein kinases: Structure and regulation. Biochem Biophys Res Commun. 2012;417:5–10. - PubMed
  16. Knighton D, Zheng J, Ten Eyck L, Ashford V, Xuong N, Taylor S, et al. Crystal structure of the catalytic subunit of cyclic adenosine monophosphate-dependent protein kinase. Science. 1991;253:407–14. - PubMed
  17. Zheng CF, Guan KL. Activation of MEK family kinases requires phosphorylation of two conserved Ser/Thr residues. EMBO J. 1994;13:1123–31. - PubMed
  18. Alessi DR, Saito Y, Campbell DG, Cohen P, Sithanandam G, Rapp U, et al. Identification of the sites in MAP kinase kinase-1 phosphorylated by p74raf-1. EMBO J. 1994;13:1610–19. - PubMed
  19. Kornev AP, Haste NM, Taylor SS, Ten Eyck LF. Surface comparison of active and inactive protein kinases identifies a conserved activation mechanism. Proc Natl Acad Sci U S A. 2006;103:17783–8. - PubMed
  20. Kornev AP, Taylor SS, Ten Eyck LF. A helix scaffold for the assembly of active protein kinases. Proc Natl Acad Sci U S A. 2008;105:14377–82. - PubMed
  21. Fischmann TO, Smith CK, Mayhood TW, Myers JE, Reichert P, Mannarino A, et al. Crystal structures of MEK1 binary and ternary complexes with nucleotides and inhibitors. Biochemistry. 2009;48:2661–74. - PubMed
  22. Yuan J, Ng WH, Tian Z, Yap J, Baccarini M, Chen Z, et al. Activating mutations in MEK1 enhance homodimerization and promote tumorigenesis. Sci Signal. 2018;11:eaar6795. - PubMed
  23. Emery CM, Vijayendran KG, Zipser MC, Sawyer AM, Niu L, Kim JJ, et al. MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc Natl Acad Sci U S A. 2009;106:20411–6. - PubMed
  24. Fukuda M, Gotoh Y, Nishida E. Interaction of MAP kinase with MAP kinase kinase: its possible role in the control of nucleocytoplasmic transport of MAP kinase. EMBO J. 1997;16:1901–8. - PubMed
  25. Dang A, Frost JA, Cobb MH. The MEK1 proline-rich insert is required for efficient activation of the mitogen-activated protein kinases ERK1 and ERK2 in mammalian cells. J Biol Chem. 1998;273:19909–13. - PubMed
  26. Catalanotti F, Reyes G, Jesenberger V, Galabova-Kovacs G, de Matos Simoes R, Carugo O, et al. A Mek1-Mek2 heterodimer determines the strength and duration of the Erk signal. Nat Struct Mol Biol. 2009;16:294–303. - PubMed
  27. Wilsbacher JL, Goldsmith EJ, Cobb MH. Phosphorylation of MAP kinases by MAP/ERK involves multiple regions of MAP kinases. J Biol Chem. 1999;274:16988–94. - PubMed
  28. Robinson MJ, Cheng M, Khokhlatchev A, Ebert D, Ahn N, Guan KL, et al. Contributions of the mitogen-activated protein (MAP) kinase backbone and phosphorylation loop to MEK specificity. J Biol Chem. 1996;271:29734–9. - PubMed
  29. Eblen ST, Catling AD, Assanah MC, Weber MJ. Biochemical and biological functions of the N-terminal, noncatalytic domain of extracellular signal-regulated kinase 2. Mol Cell Biol. 2001;21:249–59. - PubMed
  30. Burack WR, Shaw AS. Live cell imaging of ERK and MEK: Simple binding equilibrium explains the regulated nucleocytoplasmic distribution of ERK. J Biol Chem. 2005;280:3832–7. - PubMed
  31. Lenormand P, Brondello J, Brunet A, Pouysségur J. Activation and neosynthesis of nuclear anchoring proteins. Cell. 1998;142:625–33. - PubMed
  32. Khokhlatchev AV, Canagarajah B, Wilsbacher J, Robinson M, Atkinson M, Goldsmith E, et al. Phosphorylation of the MAP kinase ERK2 promotes its homodimerization and nuclear translocation. Cell. 1998;93:605–15. - PubMed
  33. Matsubayashi Y, Fukuda M, Nishida E. Evidence for existence of a nuclear pore complex-mediated, cytosol-independent pathway of nuclear translocation of ERK MAP kinase in permeabilized cells. J Biol Chem. 2001;276:41755–60. - PubMed
  34. Adachi M, Fukuda M, Nishida E. Two co-existing mechanisms for nuclear import of MAP kinase: passive diffusion of a monomer and active transport of a dimer. EMBO J. 1999;18:5347–58. - PubMed
  35. Kosako H, Yamaguchi N, Aranami C, Ushiyama M, Kose S, Imamoto N, et al. Phosphoproteomics reveals new ERK MAP kinase targets and links ERK to nucleoporin-mediated nuclear transport. Nat Struct Mol Biol. 2009;16:1026–35. - PubMed
  36. Zehorai E, Yao Z, Plotnikov A, Seger R. The subcellular localization of MEK and ERK-a novel nuclear translocation signal (NTS) paves a way to the nucleus. Mol Cell Endocrinol. 2010;314:213–20. - PubMed
  37. Lange A, Mills RE, Lange CJ, Stewart M, Devine SE, Corbett AH. Classical nuclear localization signals: definition, function, and interaction with importin α. J Biol Chem. 2007;282:5101–5. - PubMed
  38. Zehorai E, Seger R. Beta-like importins mediate the nuclear translocation of MAPKs. Cell Physiol Biochem. 2019;52:802–21. - PubMed
  39. Pouysségur J, Volmat V, Lenormand P. Fidelity and spatio-temporal control in MAP kinase (ERKs) signalling. Biochem Pharmacol. 2002;64:755–63. - PubMed
  40. Adachi M, Fukuda M, Nishida E. Nuclear export of MAP kinase (ERK) involves a MAP kinase (MEK)-dependent active transport mechanism. J Cell Biol. 2000;148:849–56. - PubMed
  41. Costa M, Marchi M, Cardarelli F, Roy A, Beltram F, Maffei L, et al. Dynamic regulation of ERK2 nuclear translocation and mobility in living cells. J Cell Sci. 2006;119:4952–63. - PubMed
  42. Furuno T, Hirashima N, Onizawa S, Sagiya N, Nakanishi M. Nuclear shuttling of mitogen-activated protein (MAP) kinase (extracellular signal-regulated kinase (ERK) 2) was dynamically controlled by MAP/ERK kinase after antigen stimulation in RBL-2H3 cells. J Immunol. 2001;166:4416–21. - PubMed
  43. Formstecher E, Ramos JW, Fauquet M, Calderwood DA, Hsieh JC, Canton B, et al. PEA-15 mediates cytoplasmic sequestration of ERK MAP kinase. Dev Cell. 2001;1:239–50. - PubMed
  44. Tohgo A, Pierce KL, Choy EW, Lefkowitz RJ, Luttrell LM. β-arrestin scaffolding of the ERK cascade enhances cytosolic ERK activity but inhibits ERK-mediated transcription following angiotensin AT1a receptor stimulation. J Biol Chem. 2002;277:9429–36. - PubMed
  45. Yoon S, Seger R. The extracellular signal-regulated kinase: multiple substrates regulate diverse cellular functions. Growth Factors. 2006;24:21–44. - PubMed
  46. Eblen ST. Extracellular-regulated kinases: signaling from Ras to ERK substrates to control biological outcomes. Adv Cancer Res. 2018;138:99–142. - PubMed
  47. Maik-Rachline G, Hacohen-Lev-Ran A, Seger R. Nuclear ERK: mechanism of translocation, substrates, and role in cancer. Int J Mol Sci. 2019;20:1–18. - PubMed
  48. Ünal EB, Uhlitz F, Blüthgen N. A compendium of ERK targets. FEBS Lett. 2017;591:2607–15. - PubMed
  49. Murphy LO, Smith S, Chen RH, Fingar DC, Blenis J. Molecular, interpretation of ERK signal duration by immediate early gene products. Nat Cell Biol. 2002;4:556–64. - PubMed
  50. Cruzalegui FH, Cano E, Treisman R. ERK activation induces phosphorylation of Elk-1 at multiple S/T-P motifs to high stoichiometry. Oncogene. 1999;18:7948–57. - PubMed
  51. Gille H, Kortenjann M, Thomae O, Moomaw C, Slaughter C, Cobb MH, et al. ERK phosphorylation potentiates Elk-1-mediated ternary complex formation and transactivation. EMBO J. 1995;14:951–62. - PubMed
  52. Weber JD, Raben DM, Phillips PJ, Baldassare JJ. Sustained activation of extracellular-signal-regulated kinase 1 (ERK1) is required for the continued expression of cyclin D1 in G1 phase. Biochem J. 1997;326:61–8. - PubMed
  53. Balmanno K, Cook SJ. Sustained MAP kinase activation is required for the expression of cyclin D1, p21(Cip1) and a subset of AP-1 proteins in CCL39 cells. Oncogene. 1999;18:3085–97. - PubMed
  54. Maekawa M, Nishida E, Tanoue T. Identification of the anti-proliferative protein Tob as a MAPK substrate. J Biol Chem. 2002;277:37783–7. - PubMed
  55. Yang JY, Zong CS, Xia W, Yamaguchi H, Ding Q, Xie X, et al. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation. Nat Cell Biol. 2008;10:138–48. - PubMed
  56. Lu Z, Xu S. ERK1/2 MAP kinases in cell survival and apoptosis. IUBMB Life. 2006;58:621–31. - PubMed
  57. Biswas SC, Greene LA. Nerve growth factor (NGF) down-regulates the Bcl-2 homology 3 (BH3) domain-only protein Bim and suppresses its proapoptotic activity by phosphorylation. J Biol Chem. 2002;277:49511–6. - PubMed
  58. Domina AM, Vrana JA, Gregory MA, Hann SR, Craig RW. MCL1 is phosphorylated in the PEST region and stabilized upon ERK activation in viable cells, and at additional sites with cytotoxic okadaic acid or Taxol. Oncogene. 2004;23:5301–15. - PubMed
  59. Liu ZX, Yu CF, Nickel C, Thomas S, Cantley LG. Hepatocyte growth factor induces ERK-dependent paxillin phosphorylation and regulates paxillin-focal adhesion kinase association. J Biol Chem. 2002;277:10452–8. - PubMed
  60. Ishibe S, Joly D, Zhu X, Cantley LG. Phosphorylation-dependent paxillin-ERK association mediates hepatocyte growth factor-stimulated epithelial morphogenesis. Mol Cell. 2003;12:1275–85. - PubMed
  61. Ishibe S, Joly D, Liu ZX, Cantley LG. Paxillin serves as an ERK-regulated scaffold for coordinating FAK and Rac activation in epithelial morphogenesis. Mol Cell. 2004;16:257–67. - PubMed
  62. Sturgill TW, Ray LB, Erikson E, Maller JL. Insulin-stimulated MAP-2 kinase phosphorylates and activates ribosomal protein S6 kinase II. Nature. 1988;334:715–8. - PubMed
  63. Deak M, Clifton AD, Lucocq JM, Alessi DR. Mitogen- and stress-activated protein kinase-1 (MSK1) is directly activated by MAPK and SAPK2/p38, and may mediate activation of CREB. EMBO J. 1998;17:4426–41. - PubMed
  64. Hauge C, Frödin M. RSK and MSK in MAP kinase signalling. J Cell Sci. 2006;119:3021–3. - PubMed
  65. Chen N, Fang W, Lin Z, Peng P, Wang J, Zhan J, et al. KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol Immunother. 2017;66:1175–87. - PubMed
  66. Zdanov S, Mandapathil M, Abu Eid R, Adamson-Fadeyi S, Wilson W, Qian J, et al. Mutant KRAS conversion of conventional T cells into regulatory T cells. Cancer Immunol Res. 2016;4:354–65. - PubMed
  67. Salaroglio IC, Campia I, Kopecka J, Gazzano E, Orecchia S, Ghigo D, et al. Zoledronic acid overcomes chemoresistance and immunosuppression of malignant mesothelioma. Oncotarget. 2014;6:1128–42. - PubMed
  68. Kopecka J, Porto S, Lusa S, Gazzano E, Salzano G, Pinzòn-Daza ML, et al. Zoledronic acid-encapsulating self-assembling nanoparticles and doxorubicin: a combinatorial approach to overcome simultaneously chemoresistance and immunoresistance in breast tumors. Oncotarget. 2016;7:20753–72. - PubMed
  69. Salaroglio IC, Mungo E, Gazzano E, Kopecka J, Riganti C. ERK is a pivotal player of chemo-immune-resistance in cancer. Int J Mol Sci. 2019;20:2505. - PubMed
  70. Yuan J, Ng WH, Lam PYP, Wang Yu, Xia H, Yap J, et al. The dimer-dependent catalytic activity of RAF family kinases is revealed through characterizing their oncogenic mutants. Oncogene. 2018;37:5719–34. - PubMed
  71. Hu J, Stites EC, Yu H, Germino EA, Meharena HS, Stork PJS, et al. Allosteric activation of functionally asymmetric RAF kinase dimers. Cell. 2013;154:1036–46. - PubMed
  72. Haystead AJ, Dent P, Wu J, Haystead GMM, Sturgill TW. Ordered phosphorylation. Assessment. 1992;306:17–22. - PubMed
  73. Zhou B, Zhang ZY. The activity of the extracellular signal-regulated kinase 2 is regulated by differential phosphorylation in the activation loop. J Biol Chem. 2002;277:13889–99. - PubMed
  74. Cha H, Shapiro P. Tyrosine-phosphorylated extracellular signal-regulated kinase associates with the Golgi complex during G2–M phase of the cell cycle: evidence for regulation of Golgi structure. J Cell Biol. 2001;153:1355–67. - PubMed
  75. Freeman AK, Ritt DA, Morrison DK. Effects of raf dimerization and its inhibition on normal and disease-associated raf signaling. Mol Cell. 2013;49:751–8. - PubMed
  76. Aoki K, Yamada M, Kunida K, Yasuda S, Matsuda M. Processive phosphorylation of ERK MAP kinase in mammalian cells. Proc Natl Acad Sci U S A. 2011;108:12675–80. - PubMed
  77. Zhou LEI, Tan X, Kamohara H, Wang W, Wang B, Liu J, et al. MEK1 and MEK2 isoforms regulate distinct functions in pancreatic cancer cells. Oncol Rep. 2010;24:251–5. - PubMed
  78. Canagarajah BJ, Khokhlatchev A, Cobb MH, Goldsmith EJ. Activation mechanism of the MAP kinase ERK2 by dual phosphorylation. Cell. 1997;90:859–69. - PubMed
  79. Wilsbacher JL, Juang YC, Khokhlatchev AV, Gallagher E, Binns D, Goldsmith EJ, et al. Characterization of mitogen-activated protein kinase (MAPK) dimers. Biochemistry. 2006;45:13175–82. - PubMed
  80. Ohren JF, Chen H, Pavlovsky A, Whitehead C, Zhang E, Kuffa P, et al. Structures of human MAP kinase 1 (MEK1) and MEK2 describe novel noncompetitive kinase inhibition. Nat Struct Mol Biol. 2004;11:1192–7. - PubMed
  81. Philipova R, Whitaker M. Active ERK1 is dimerized in vivo: bisphosphodimers generate peak kinase activity and monophosphodimers maintain basal ERK1 activity. J Cell Sci. 2005;118:5767–76. - PubMed
  82. Lake D, Corrêa SAL, Müller J. Negative feedback regulation of the ERK1/2 MAPK pathway. Cell Mol Life Sci. 2016;73:4397–413. - PubMed
  83. Dougherty MK, Müller J, Ritt DA, Zhou M, Zhou XZ, Copeland TD, et al. Regulation of Raf-1 by direct feedback phosphorylation. Mol Cell. 2005;17:215–24. - PubMed
  84. Ritt DA, Monson DM, Specht SI, Morrison DK. Impact of feedback phosphorylation and raf heterodimerization on normal and mutant B-Raf signaling. Mol Cell Biol. 2010;30:806–19. - PubMed
  85. Langlois WJ, Sasaoka T, Saltiel AR, Olefsky JM. Negative feedback regulation and desensitization of insulin- and epidermal growth factor- stimulated p21 ras activation. J Biol Chem. 1995;270:25320–3. - PubMed
  86. Douville E, Downward J. EGF induced SOS phosphorylation in PC12 cells involves P90 RSK-2. Oncogene. 1997;15:373–83. - PubMed
  87. Waters SB, Holt KH, Ross SE, Syu LJ, Guan KL, Saltiel AR, et al. Desensitization of Ras activation by a feedback disassociation of the SOS-Grb2 complex. J Biol Chem. 1995;270:20883–6. - PubMed
  88. Kawasaki Y, Sakimura A, Park CM, Tomaru R, Tanaka T, Ozawa T, et al. Feedback control of ErbB2 via ERK-mediated phosphorylation of a conserved threonine in the juxta membrane domain. Sci Rep. 2016;6:1–9. - PubMed
  89. McKay MM, Ritt DA, Morrison DK. Signaling dynamics of the KSR1 scaffold complex. Proc Natl Acad Sci U S A. 2009;106:11022–7. - PubMed
  90. Lavoie H, Sahmi M, Maisonneuve P, Marullo SA, Thevakumaran N, Jin T, et al. MEK drives BRAF activation through allosteric control of KSR proteins. Nature. 2018;554:549–53. - PubMed
  91. Kolch W. Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat Rev Mol Cell Biol. 2005;6:827–37. - PubMed
  92. Yeung K, Janosch P, McFerran B, Rose DW, Mischak H, Sedivy JM, et al. Mechanism of suppression of the Raf/MEK/extracellular signal-regulated kinase pathway by the raf kinase inhibitor protein. Mol Cell Biol. 2000;20:3079–85. - PubMed
  93. Shin SY, Rath O, Choo SM, Fee F, McFerran B, Kolch W, et al. Positive- and negative-feedback regulations coordinate the dynamic behavior of the Ras-Raf-MEK-ERK signal transduction pathway. J Cell Sci. 2009;122:425–35. - PubMed
  94. Corbit KC, Trakul N, Eves EM, Diaz B, Marshall M, Rosner MR, et al. Activation of raf-1 signaling by protein kinase C through a mechanism involving Raf kinase inhibitory protein. J Biol Chem. 2003;278:13061–8. - PubMed
  95. Caunt CJ, Keyse SM. Dual-specificity MAP kinase phosphatases (MKPs): shaping the outcome of MAP kinase signalling. FEBS J. 2013;280:489–504. - PubMed
  96. Ozaki K, Kadomoto R, Asato K, Tanimura S, Itoh N, Kohno M. Erk pathway positively regulates the expression of sprouty genes. Biochem Biophys Res Commun. 2001;285:1084–8. - PubMed
  97. Kucharska A, Rushworth LK, Staples C, Morrice NA, Keyse SM. Regulation of the inducible nuclear dual-specificity phosphatase DUSP5 by ERK MAPK. Cell Signal. 2009;21:1794–805. - PubMed
  98. Caunt CJ, Rivers CA, Conway-Campbell BL, Norman MR, McArdle CA. Epidermal growth factor receptor and protein kinase C signaling to ERK2: Spatiotemporal regulation of ERK2 by dual specificity phosphatases. J Biol Chem. 2008;283:6241–52. - PubMed
  99. Karlsson M, Mathers J, Dickinson RJ, Mandl M, Keyse SM. Both nuclear-cytoplasmic shuttling of the dual specificity phosphatase MKP-3 and its ability to anchor MAP kinase in the cytoplasm are mediated by a conserved nuclear export signal. J Biol Chem. 2004;279:41882–91. - PubMed
  100. Ekerot M, Stavridis MP, Delavaine L, Mitchell MP, Staples C, Owens DM, et al. Negative-feedback regulation of FGF signalling by DUSP6/MKP-3 is driven by ERK1/2 and mediated by Ets factor binding to a conserved site within the DUSP6/MKP-3 gene promoter. Biochem J. 2008;412:287–98. - PubMed
  101. Brondello JM, Pouysségur J, McKenzie FR. Reduced MAP kinase phosphatase-1 degradation after p42/p44(MAPK)- dependent phosphorylation. Science. 1999;286:2514–7. - PubMed
  102. Cagnol S, Rivard N. Oncogenic KRAS and BRAF activation of the MEK/ERK signaling pathway promotes expression of dual-specificity phosphatase 4 (DUSP4/MKP2) resulting in nuclear ERK1/2 inhibition. Oncogene. 2013;32:564–76. - PubMed
  103. Liu S, Sun JP, Zhou B, Zhang ZY. Structural basis of docking interactions between ERK2 and MAP kinase phosphatase 3. Proc Natl Acad Sci U S A. 2006;103:5326–31. - PubMed
  104. Yusoff P, Lao DH, Ong SH, Wong ESM, Lim J, Lo TL, et al. Sprouty2 inhibits the Ras/MAP kinase pathway by inhibiting the activation of Raf. J Biol Chem. 2002;277:3195–201. - PubMed
  105. Gross I, Bassit B, Benezra M, Licht JD. Mammalian sprouty proteins inhibit cell growth and differentiation by preventing ras activation. J Biol Chem. 2001;276:46460–8. - PubMed
  106. Sasaki A, Taketomi T, Kato R, Saeki K, Nonami A, Sasaki M, et al. Mammalian Sprouty4 suppresses Ras-independent ERK activation by binding to Raf1. Nat Cell Biol. 2003;5:427–32. - PubMed
  107. Hanafusa H, Torii S, Yasunaga T, Nishida E. Sprouty1 and Sprouty2 provide a control mechanism for the Ras/MAPK signalling pathway. Nat Cell Biol. 2002;4:850–8. - PubMed
  108. Brady SC, Coleman ML, Munro J, Feller SM, Morrice NA, Olson MF. Sprouty2 association with B-Raf is regulated by phosphorylation and kinase conformation. Cancer Res. 2009;69:6773–81. - PubMed
  109. Avraham R, Yarden Y. Feedback regulation of EGFR signalling: decision making by early and delayed loops. Nat Rev Mol Cell Biol. 2011;12:104–17. - PubMed
  110. Liu F, Yang X, Geng M, Huang M. Targeting ERK, an Achilles’ heel of the MAPK pathway, in cancer therapy. Acta Pharm Sin B. 2018;8:552–62. - PubMed
  111. Ramos JW. The regulation of extracellular signal-regulated kinase (ERK) in mammalian cells. Int J Biochem Cell Biol. 2008;40:2707–19. - PubMed
  112. Roskoski R. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol Res. 2016;103:26–48. - PubMed
  113. Wu PK, Park JI. MEK1/2 inhibitors: molecular activity and resistance mechanisms. Semin Oncol. 2015;42:849–62. - PubMed
  114. Markham A, Keam SJ. Selumetinib: first approval. Drugs. 2020;80:931–7. - PubMed
  115. Robarge KD, Lee W, Eigenbrot C, Ultsch M, Wiesmann C, Heald R, et al. Structure based design of novel 6,5 heterobicyclic mitogen-activated protein kinase kinase (MEK) inhibitors leading to the discovery of imidazo[1,5-a] pyrazine G-479. Bioorganic Med Chem Lett. 2014;24:4714–23. - PubMed
  116. Ryan MB, Der CJ, Wang-Gillam A, Cox AD. Targeting RAS-mutant cancers: is ERK the key?. Trends Cancer. 2015;1:183–98. - PubMed
  117. Deng Y, Shipps GW, Cooper A, English JM, Annis DA, Carr D, et al. Discovery of novel, dual mechanism ERK inhibitors by affinity selection screening of an inactive kinase. J Med Chem. 2014;57:8817–26. - PubMed
  118. Merchant M, Moffat J, Schaefer G, Chan J, Wang Xi, Orr C, et al. Combined MEK and ERK inhibition overcomes therapy-mediated pathway reactivation in RAS mutant tumors. PLoS One. 2017;12:1–22. - PubMed
  119. Germann UA, Furey BF, Markland W, Hoover RR, Aronov AM, Roix JJ, et al. Targeting the MAPK signaling pathway in cancer: promising preclinical activity with the novel selective ERK1/2 inhibitor BVD-523 (ulixertinib). Mol Cancer Ther. 2017;16:2351–63. - PubMed
  120. Tomasovic A, Brand T, Schanbacher C, Kramer S, Hümmert MW, Godoy P, et al. Interference with ERK-dimerization at the nucleocytosolic interface targets pathological ERK1/2 signaling without cardiotoxic side-effects. Nat Commun. 2020;11:1733. - PubMed
  121. Lorenz K, Schmitt JP, Schmitteckert EM, Lohse MJ. A new type of ERK1/2 autophosphorylation causes cardiac hypertrophy. Nat Med. 2009;15:75–83. - PubMed
  122. Chandarlapaty S. Negative feedback and adaptive resistance to the targeted therapy of cancer. Cancer Discov. 2012;2:311–9. - PubMed
  123. Su F, Bradley WD, Wang Q, Yang H, Xu L, Higgins B, et al. Resistance to selective BRAF inhibition can be mediated by modest upstream pathway activation. Cancer Res. 2012;72:969–78. - PubMed
  124. Plotnikov A, Flores K, Maik-Rachline G, Zehorai E, Kapri-Pardes E, Berti DA, et al. The nuclear translocation of ERK1/2 as an anticancer target. Nat Commun. 2015;6:6685. - PubMed
  125. VanBrocklin MW, Verhaegen M, Soengas MS, Holmen SL. Mitogen-activated protein kinase inhibition induces translocation of bmf to promote apoptosis in melanoma. Cancer Res. 2009;69:1985–94. - PubMed
  126. Jiang CC, Lai F, Tay KH, Croft A, Rizos H, Becker TM, et al. Apoptosis of human melanoma cells induced by inhibition of B-RAF V600E involves preferential splicing of bimS. Cell Death Dis. 2010;1:1–14. - PubMed
  127. Beck D, Niessner H, Smalley KSM, Flaherty K, Paraiso KHT, Busch C, et al. Vemurafenib potently induces endoplasmic reticulum stress-mediated apoptosis in BRAFV600E melanoma cells. Sci Signal. 2013;6:ra7. - PubMed
  128. Eroglu Z, Ribas A. Combination therapy with BRAF and MEK inhibitors for melanoma: latest evidence and place in therapy. Ther Adv Med Oncol. 2016;8:48–56. - PubMed

MeSH terms

Publication Types

Grant support