Skip to main content

Advertisement

Log in

The Potential of Targeting P53 and HSP90 Overcoming Acquired MAPKi-Resistant Melanoma

  • Skin Cancer (T Ito, Section Editor)
  • Published:
Current Treatment Options in Oncology Aims and scope Submit manuscript

Opinion Statement

Melanoma is the deadliest form of skin cancer worldwide. The rising melanoma incidence and mortality, along with its high propensity for metastasis highlights the urgency to identify more effective therapeutic targets. Approximately, one half of advanced melanoma bears a mutation in the BRAF gene that makes BRAF as an important therapeutic target. Significant clinical benefit is associated with BRAF and MEK inhibitors (MAPKi) on targeting patients with BRAF V600 mutations. However, the frequent and rapid development of acquired resistance still is the major challenge facing the melanoma. Several mechanisms by which melanoma passes the inhibitory effects of MAPKi have been characterized and clinically translated, but additional alternations of genetic and epigenetic regulators outside of MAPK and/or AKT networks occurs in a quarter of patients with acquired MAPKi resistance. These studies implicate that targeting signaling networks external MAPK or AKT pathways is critical. In this review, we will focus on two approaches that are under evaluating for targeting melanoma: (1) against genome instability by p53 network restoration and (2) disrupt cancer proteome by chaperone inhibition.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30.

    Article  PubMed  Google Scholar 

  2. Chang JW. Cutaneous melanoma: Taiwan experience and literature review. Chang Gung Med J. 2010;33(6):602–12.

    PubMed  Google Scholar 

  3. Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004;116(6):855–67.

    Article  CAS  PubMed  Google Scholar 

  4. Salama AK, Flaherty KT. BRAF in melanoma: current strategies and future directions. Clin Cancer Res. 2013;19(16):4326–34.

    Article  CAS  PubMed  Google Scholar 

  5. Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012;367(18):1694–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Long GV, Hauschild A, Santinami M, Atkinson V, Mandala M, Chiarion-Sileni V, et al. Adjuvant dabrafenib plus trametinib in stage III BRAF-mutated melanoma. N Engl J Med. 2017;377(19):1813–23.

    Article  CAS  PubMed  Google Scholar 

  7. Amaria RN, Prieto PA, Tetzlaff MT, Reuben A, Andrews MC, Ross MI, et al. Neoadjuvant plus adjuvant dabrafenib and trametinib versus standard of care in patients with high-risk, surgically resectable melanoma: a single-centre, open-label, randomised, phase 2 trial. Lancet Oncol. 2018;19(2):181–93.

    Article  CAS  PubMed  Google Scholar 

  8. Hauschild A, Dummer R, Schadendorf D, Santinami M, Atkinson V, Mandala M, et al. Longer follow-up confirms relapse-free survival benefit with adjuvant dabrafenib plus trametinib in patients with resected BRAF V600-mutant stage III melanoma. J Clin Oncol. 2018;JCO1801219.

  9. •• Solit DB, Rosen N. Resistance to BRAF inhibition in melanomas. N Engl J Med. 2011;364(8):772–4 This article provides a comprehensive review of mechanisms of MAPKi resistance in melanoma and summarizes landmark trials for metastatic melanoma treatment.

    Article  CAS  PubMed  Google Scholar 

  10. Shi H, Hugo W, Kong X, Hong A, Koya RC, Moriceau G, et al. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov. 2014;4(1):80–93.

    Article  CAS  PubMed  Google Scholar 

  11. Kakadia S, Yarlagadda N, Awad R, Kundranda M, Niu J, Naraev B, et al. Mechanisms of resistance to BRAF and MEK inhibitors and clinical update of US Food and Drug Administration-approved targeted therapy in advanced melanoma. Onco Targets Ther. 2018;11:7095–107.

    Article  PubMed  PubMed Central  Google Scholar 

  12. • Shen CH, Kim SH, Trousil S, Frederick DT, Piris A, Yuan P, et al. Loss of cohesin complex components STAG2 or STAG3 confers resistance to BRAF inhibition in melanoma. Nat Med. 2016;22(9):1056–61 A novel genome-wide chromatin interaction leads to MAPKi resistance in melanoma.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. • Strub T, Ghiraldini FG, Carcamo S, Li M, Wroblewska A, Singh R, et al. SIRT6 haploinsufficiency induces BRAF(V600E) melanoma cell resistance to MAPK inhibitors via IGF signalling. Nat Commun. 2018;9(1):3440 A novel epigenetic regulators involved in MAPKi resistance development in melanoma.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Lane DP. Cancer. p53, guardian of the genome. Nature. 1992;358(6381):15–6.

    Article  CAS  PubMed  Google Scholar 

  15. Zilfou JT, Lowe SW. Tumor suppressive functions of p53. Cold Spring Harb Perspect Biol. 2009;1(5):a001883.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Wade M, Wang YV, Wahl GM. The p53 orchestra: Mdm2 and Mdmx set the tone. Trends Cell Biol. 2010;20(5):299–309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jackson MW, Berberich SJ. MdmX protects p53 from Mdm2-mediated degradation. Mol Cell Biol. 2000;20(3):1001–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Montes de Oca Luna R, Wagner DS, Lozano G. Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature. 1995;378(6553):203–6.

    Article  CAS  PubMed  Google Scholar 

  19. Jones SN, Roe AE, Donehower LA, Bradley A. Rescue of embryonic lethality in Mdm2-deficient mice by absence of p53. Nature. 1995;378(6553):206–8.

    Article  CAS  PubMed  Google Scholar 

  20. Parant J, Chavez-Reyes A, Little NA, Yan W, Reinke V, Jochemsen AG, et al. Rescue of embryonic lethality in Mdm4-null mice by loss of Trp53 suggests a nonoverlapping pathway with MDM2 to regulate p53. Nat Genet. 2001;29(1):92–5.

    Article  CAS  PubMed  Google Scholar 

  21. Gannon HS, Donehower LA, Lyle S, Jones SN. Mdm2-p53 signaling regulates epidermal stem cell senescence and premature aging phenotypes in mouse skin. Dev Biol. 2011;353(1):1–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gembarska A, Luciani F, Fedele C, Russell EA, Dewaele M, Villar S, et al. MDM4 is a key therapeutic target in cutaneous melanoma. Nat Med. 2012;18(8):1239–47.

    Article  CAS  PubMed  Google Scholar 

  23. Marine JC, Francoz S, Maetens M, Wahl G, Toledo F, Lozano G. Keeping p53 in check: essential and synergistic functions of Mdm2 and Mdm4. Cell Death Differ. 2006;13(6):927–34.

    Article  CAS  PubMed  Google Scholar 

  24. Matin RN, Chikh A, Chong SL, Mesher D, Graf M. Sanza P, et al. p63 is an alternative p53 repressor in melanoma that confers chemoresistance and a poor prognosis. J Exp Med. 2013;210(3):581–603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lu M, Breyssens H, Salter V, Zhong S, Hu Y, Baer C, et al. Restoring p53 function in human melanoma cells by inhibiting MDM2 and cyclin B1/CDK1-phosphorylated nuclear iASPP. Cancer Cell. 2013;23(5):618–33.

    Article  CAS  PubMed  Google Scholar 

  26. •• Nardella C, Clohessy JG, Alimonti A, Pandolfi PP. Pro-senescence therapy for cancer treatment. Nat Rev Cancer. 2011;11(7):503–11 Provide concepts that are relevant to a pro-senescence approach to therapy in tumours.

    Article  CAS  PubMed  Google Scholar 

  27. Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 2004;303(5659):844–8.

    Article  CAS  PubMed  Google Scholar 

  28. Ray-Coquard I, Blay JY, Italiano A, Le Cesne A, Penel N, Zhi J, et al. Effect of the MDM2 antagonist RG7112 on the P53 pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: an exploratory proof-of-mechanism study. Lancet Oncol. 2012;13(11):1133–40.

    Article  CAS  PubMed  Google Scholar 

  29. Patnaik A, Tolcher A, Beeram M, Nemunaitis J, Weiss GJ, Bhalla K, et al. Clinical pharmacology characterization of RG7112, an MDM2 antagonist, in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2015;76(3):587–95.

    Article  CAS  PubMed  Google Scholar 

  30. Andreeff M, Kelly KR, Yee K, Assouline S, Strair R, Popplewell L, et al. Results of the phase I trial of RG7112, a small-molecule MDM2 antagonist in leukemia. Clin Cancer Res. 2016;22(4):868–76.

    Article  CAS  PubMed  Google Scholar 

  31. Reis B, Jukofsky L, Chen G, Martinelli G, Zhong H, So WV, et al. Acute myeloid leukemia patients’ clinical response to idasanutlin (RG7388) is associated with pre-treatment MDM2 protein expression in leukemic blasts. Haematologica. 2016;101(5):e185–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Canon J, Osgood T, Olson SH, Saiki AY, Robertson R, Yu D, et al. The MDM2 inhibitor AMG 232 demonstrates robust antitumor efficacy and potentiates the activity of p53-inducing cytotoxic agents. Mol Cancer Ther. 2015;14(3):649–58.

    Article  CAS  PubMed  Google Scholar 

  33. Her NG, Oh JW, Oh YJ, Han S, Cho HJ, Lee Y, et al. Potent effect of the MDM2 inhibitor AMG232 on suppression of glioblastoma stem cells. Cell Death Dis. 2018;9(8):792.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Cheok CF, Verma CS, Baselga J, Lane DP. Translating p53 into the clinic. Nat Rev Clin Oncol. 2011;8(1):25–37.

    Article  CAS  PubMed  Google Scholar 

  35. Wang YC, Wu YT, Huang HY, Lin HI, Lo LW, Tzeng SF, et al. Sustained intraspinal delivery of neurotrophic factor encapsulated in biodegradable nanoparticles following contusive spinal cord injury. Biomaterials. 2008;29(34):4546–53.

    Article  CAS  PubMed  Google Scholar 

  36. Joseph TL, Madhumalar A, Brown CJ, Lane DP, Verma CS. Differential binding of p53 and nutlin to MDM2 and MDMX: computational studies. Cell Cycle. 2010;9(6):1167–81.

    Article  CAS  PubMed  Google Scholar 

  37. Wachter F, Morgan AM, Godes M, Mourtada R, Bird GH, Walensky LD. Mechanistic validation of a clinical lead stapled peptide that reactivates p53 by dual HDM2 and HDMX targeting. Oncogene. 2017;36(15):2184–90.

    Article  CAS  PubMed  Google Scholar 

  38. Chang YS, Graves B, Guerlavais V, Tovar C, Packman K, To KH, et al. Stapled alpha-helical peptide drug development: a potent dual inhibitor of MDM2 and MDMX for p53-dependent cancer therapy. Proc Natl Acad Sci U S A. 2013;110(36):E3445–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Carvajal LA, Neriah DB, Senecal A, Benard L, Thiruthuvanathan V, Yatsenko T, et al. Dual inhibition of MDMX and MDM2 as a therapeutic strategy in leukemia. Sci Transl Med. 2018;10(436).

  40. Amit S, Hatzubai A, Birman Y, Andersen JS, Ben-Shushan E, Mann M, et al. Axin-mediated CKI phosphorylation of beta-catenin at Ser 45: a molecular switch for the Wnt pathway. Genes Dev. 2002;16(9):1066–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Pribluda A, Elyada E, Wiener Z, Hamza H, Goldstein RE, Biton M, et al. A senescence-inflammatory switch from cancer-inhibitory to cancer-promoting mechanism. Cancer Cell. 2013;24(2):242–56.

    Article  CAS  PubMed  Google Scholar 

  42. Huart AS, MacLaine NJ, Meek DW, Hupp TR. CK1alpha plays a central role in mediating MDM2 control of p53 and E2F-1 protein stability. J Biol Chem. 2009;284(47):32384–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chen L, Li C, Pan Y, Chen J. Regulation of p53-MDMX interaction by casein kinase 1 alpha. Mol Cell Biol. 2005;25(15):6509–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Elyada E, Pribluda A, Goldstein RE, Morgenstern Y, Brachya G, Cojocaru G, et al. CKIalpha ablation highlights a critical role for p53 in invasiveness control. Nature. 2011;470(7334):409–13.

    Article  CAS  PubMed  Google Scholar 

  45. Minzel W, Venkatachalam A, Fink A, Hung E, Brachya G, Burstain I, et al. Small molecules co-targeting CKIalpha and the transcriptional kinases CDK7/9 control AML in preclinical models. Cell. 2018;175(1):171–85.e25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kronke J, Fink EC, Hollenbach PW, MacBeth KJ, Hurst SN, Udeshi ND, et al. Lenalidomide induces ubiquitination and degradation of CK1alpha in del(5q) MDS. Nature. 2015;523(7559):183–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Li G, Xiao Z, Liu J, Li C, Li F, Chen Z. Cancer: a proteomic disease. Sci China Life Sci. 2011;54(5):403–8.

    Article  CAS  PubMed  Google Scholar 

  48. •• Moulick K, Ahn JH, Zong H, Rodina A, Cerchietti L, Gomes DaGama EM, et al. Affinity-based proteomics reveal cancer-specific networks coordinated by Hsp90. Nat Chem Biol. 2011;7(11):818–26 Targeting chaperone HSP90 is a considerable anticancer strategy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Whitesell L, Lindquist SL. HSP90 and the chaperoning of cancer. Nat Rev Cancer. 2005;5(10):761–72.

    Article  CAS  PubMed  Google Scholar 

  50. Verba KA, Wang RY, Arakawa A, Liu Y, Shirouzu M, Yokoyama S, et al. Atomic structure of Hsp90-Cdc37-Cdk4 reveals that Hsp90 traps and stabilizes an unfolded kinase. Science. 2016;352(6293):1542–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Karagoz GE, Duarte AM, Akoury E, Ippel H, Biernat J, Moran Luengo T, et al. Hsp90-Tau complex reveals molecular basis for specificity in chaperone action. Cell. 2014;156(5):963–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Kirschke E, Goswami D, Southworth D, Griffin PR, Agard DA. Glucocorticoid receptor function regulated by coordinated action of the Hsp90 and Hsp70 chaperone cycles. Cell. 2014;157(7):1685–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Mayer MP. Hsp70 chaperone dynamics and molecular mechanism. Trends Biochem Sci. 2013;38(10):507–14.

    Article  CAS  PubMed  Google Scholar 

  54. Panaretou B, Prodromou C, Roe SM, O'Brien R, Ladbury JE, Piper PW, et al. ATP binding and hydrolysis are essential to the function of the Hsp90 molecular chaperone in vivo. EMBO J. 1998;17(16):4829–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ali MM, Roe SM, Vaughan CK, Meyer P, Panaretou B, Piper PW, et al. Crystal structure of an Hsp90-nucleotide-p23/Sba1 closed chaperone complex. Nature. 2006;440(7087):1013–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Panaretou B, Siligardi G, Meyer P, Maloney A, Sullivan JK, Singh S, et al. Activation of the ATPase activity of hsp90 by the stress-regulated cochaperone aha1. Mol Cell. 2002;10(6):1307–18.

    Article  CAS  PubMed  Google Scholar 

  57. Trepel J, Mollapour M, Giaccone G, Neckers L. Targeting the dynamic HSP90 complex in cancer. Nat Rev Cancer. 2010;10(8):537–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Neckers L, Trepel JB. Stressing the development of small molecules targeting HSP90. Clin Cancer Res. 2014;20(2):275–7.

    Article  CAS  PubMed  Google Scholar 

  59. Xu W, Neckers L. Targeting the molecular chaperone heat shock protein 90 provides a multifaceted effect on diverse cell signaling pathways of cancer cells. Clin Cancer Res. 2007;13(6):1625–9.

    Article  CAS  PubMed  Google Scholar 

  60. Whitesell L, Mimnaugh EG, De Costa B, Myers CE, Neckers LM. Inhibition of heat shock protein HSP90-pp60v-src heteroprotein complex formation by benzoquinone ansamycins: essential role for stress proteins in oncogenic transformation. Proc Natl Acad Sci U S A. 1994;91(18):8324–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Roe SM, Prodromou C, O'Brien R, Ladbury JE, Piper PW, Pearl LH. Structural basis for inhibition of the Hsp90 molecular chaperone by the antitumor antibiotics radicicol and geldanamycin. J Med Chem. 1999;42(2):260–6.

    Article  CAS  PubMed  Google Scholar 

  62. Grenert JP, Sullivan WP, Fadden P, Haystead TA, Clark J, Mimnaugh E, et al. The amino-terminal domain of heat shock protein 90 (hsp90) that binds geldanamycin is an ATP/ADP switch domain that regulates hsp90 conformation. J Biol Chem. 1997;272(38):23843–50.

    Article  CAS  PubMed  Google Scholar 

  63. Powers MV, Workman P. Inhibitors of the heat shock response: biology and pharmacology. FEBS Lett. 2007;581(19):3758–69.

    Article  CAS  PubMed  Google Scholar 

  64. Jhaveri K, Taldone T, Modi S, Chiosis G. Advances in the clinical development of heat shock protein 90 (Hsp90) inhibitors in cancers. Biochim Biophys Acta. 2012;1823(3):742–55.

    Article  CAS  PubMed  Google Scholar 

  65. Schulte TW, Neckers LM. The benzoquinone ansamycin 17-allylamino-17-demethoxygeldanamycin binds to HSP90 and shares important biologic activities with geldanamycin. Cancer Chemother Pharmacol. 1998;42(4):273–9.

    Article  CAS  PubMed  Google Scholar 

  66. Chatterjee S, Burns TF. Targeting heat shock proteins in cancer: a promising therapeutic approach. Int J Mol Sci. 2017;18(9).

  67. Pacey S, Gore M, Chao D, Banerji U, Larkin J, Sarker S, et al. A Phase II trial of 17-allylamino, 17-demethoxygeldanamycin (17-AAG, tanespimycin) in patients with metastatic melanoma. Investig New Drugs. 2012;30(1):341–9.

    Article  CAS  Google Scholar 

  68. Vaishampayan UN, Burger AM, Sausville EA, Heilbrun LK, Li J, Horiba MN, et al. Safety, efficacy, pharmacokinetics, and pharmacodynamics of the combination of sorafenib and tanespimycin. Clin Cancer Res. 2010;16(14):3795–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Garcia-Carbonero R, Carnero A, Paz-Ares L. Inhibition of HSP90 molecular chaperones: moving into the clinic. Lancet Oncol. 2013;14(9):e358–69.

    Article  CAS  PubMed  Google Scholar 

  70. • Sidera K, Patsavoudi E. HSP90 inhibitors: current development and potential in cancer therapy. Recent Pat Anticancer Drug Discov. 2014;9(1):1–20 The first clinical trail for evaluation of the combinaiton of BRAFi and HSP90i in melanoma.

    Article  CAS  PubMed  Google Scholar 

  71. Eroglu Z, Chen YA, Gibney GT, Weber JS, Kudchadkar RR, Khushalani NI, et al. Combined BRAF and HSP90 inhibition in patients with unresectable BRAF (V600E)-mutant melanoma. Clin Cancer Res. 2018;24(22):5516–24.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Yuno A, Lee MJ, Lee S, Tomita Y, Rekhtman D, Moore B, et al. Clinical evaluation and biomarker profiling of Hsp90 inhibitors. Methods Mol Biol. 2018;1709:423–41.

    Article  CAS  PubMed  Google Scholar 

  73. Aguila M, Bevilacqua D, McCulley C, Schwarz N, Athanasiou D, Kanuga N, et al. Hsp90 inhibition protects against inherited retinal degeneration. Hum Mol Genet. 2014;23(8):2164–75.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Che-Hung Shen PhD.

Ethics declarations

Conflict of Interest

Chi-Che Hsieh and Che-Hung Shen declare they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Skin Cancer

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hsieh, CC., Shen, CH. The Potential of Targeting P53 and HSP90 Overcoming Acquired MAPKi-Resistant Melanoma. Curr. Treat. Options in Oncol. 20, 22 (2019). https://doi.org/10.1007/s11864-019-0622-9

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11864-019-0622-9

Keywords

Navigation