Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Copper ionophore elesclomol selectively targets GNAQ/11-mutant uveal melanoma

Abstract

Unlike cutaneous melanoma, uveal melanoma (UM) is characterized by mutations in GNAQ and GNA11 and remains a fatal disease because there is essentially no effective targeted therapy or immunotherapy available. We report the discovery of the copper ionophore elesclomol as a GNAQ/11-specific UM inhibitor. Elesclomol was identified in a differential cytotoxicity screen of an in-house tool compound library, and its in vivo pharmacological efficacy was further confirmed in zebrafish and mouse UM models. Mechanistically, elesclomol transports copper to mitochondria and produces a large amount of reactive oxygen species (ROS) as Cu(II) is reduced to Cu(I) in GNAQ/11-mutant UM cells, which selectively activates LATS1 kinase in the Hippo signaling pathway and consequently promotes YAP phosphorylation and inhibits its nuclear accumulation. The inactivation of YAP downregulates the expression of SNAI2, which in turn suppresses the migration of UM cells. These findings were cross validated by our clinical observation that YAP activation was found specifically in UM samples with a GNAQ/11 mutation. Furthermore, addition of binimetinib, a MEK inhibitor, to elesclomol increased its synthetic lethality to GNAQ/11-mutant UM cells, thereby overriding drug resistance. This effect was confirmed in an orthotopic xenograft model and in a patient-derived xenograft model of UM. These studies reveal a novel mechanistic basis for repurposing elesclomol by showing that copper homeostasis is a GNAQ/11-specific vulnerability in UM. Elesclomol may provide a new therapeutic path for selectively targeting malignant GNAQ/11-mutant UM.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Discovery of the GNAQ/11-specific compound elesclomol.
Fig. 2: Elesclomol selectively elevates intracellular ROS in GNAQ/11-mutant UM cells.
Fig. 3: Specific cellular events in elesclomol-treated GNAQ/11-mutant UM cells.
Fig. 4: Elesclomol impairs UM tumor growth in zebrafish and mouse models.
Fig. 5: Elesclomol selectively induces YAP phosphorylation in GNAQ/11-mutant UM cells.
Fig. 6: Elesclomol promotes YAP phosphorylation via ROS-induced Hippo signaling pathway activation.
Fig. 7: Antiproliferative effect of elesclomol on MEKi-resistant UM cells.
Fig. 8: Elesclomol regulates SNAI2 expression and cell migration through YAP.

Similar content being viewed by others

Data availability

All sequencing data are available through the Gene Expression Omnibus (GEO) via accession code PRJNA733836.

References

  1. Singh AD, Topham A. Incidence of uveal melanoma in the United States: 1973-97. Ophthalmology. 2003;110:956–61.

    Article  PubMed  Google Scholar 

  2. Shields CL, Shields JA. Ocular melanoma: relatively rare but requiring respect. Clin Dermatol. 2009;27:122–33.

    Article  PubMed  Google Scholar 

  3. Khoja L, Atenafu EG, Suciu S, Leyvraz S, Sato T, Marshall E, et al. Meta-analysis in metastatic uveal melanoma to determine progression free and overall survival benchmarks: an international rare cancers initiative (IRCI) ocular melanoma study. Ann Oncol. 2019;30:1370–80.

    Article  CAS  PubMed  Google Scholar 

  4. Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, O’Brien JM, et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457:599–602.

    Article  PubMed  CAS  Google Scholar 

  5. Van Raamsdonk CD, Griewank KG, Crosby MB, Garrido MC, Vemula S, Wiesner T, et al. Mutations in GNA11 in uveal melanoma. N Engl J Med. 2010;363:2191–9.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Smit KN, Jager MJ, de Klein A, Kili E. Uveal melanoma: towards a molecular understanding. Prog Retin Eye Res. 2020;75:100800.

    Article  PubMed  Google Scholar 

  7. Kleuss C, Raw AS, Lee E, Sprang SR, Gilman AG. Mechanism of GTP hydrolysis by G-protein alpha subunits. Proc Natl Acad Sci USA. 1994;91:9828–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Populo H, Soares P, Rocha AS, Silva P, Lopes JM. Evaluation of the mTOR pathway in ocular (uvea and conjunctiva) melanoma. Melanoma Res. 2010;20:107–17.

    Article  CAS  PubMed  Google Scholar 

  9. Feng X, Degese MS, Iglesias-Bartolome R, Vaque JP, Molinolo AA, Rodrigues M, et al. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry. Cancer Cell. 2014;25:831–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yu FX, Luo J, Mo JS, Liu G, Kim YC, Meng Z, et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell. 2014;25:822–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Campbell AP, Smrcka AV. Targeting G protein-coupled receptor signalling by blocking G proteins. Nat Rev Drug Discov. 2018;17:789–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Schrage R, Schmitz AL, Gaffal E, Annala S, Kehraus S, Wenzel D, et al. The experimental power of FR900359 to study Gq-regulated biological processes. Nat Commun. 2015;6:10156.

    Article  CAS  PubMed  Google Scholar 

  13. Onken MD, Makepeace CM, Kaltenbronn KM, Kanai SM, Todd TD, Wang S, et al. Targeting nucleotide exchange to inhibit constitutively active G protein alpha subunits in cancer cells. Sci Signal. 2018;11:eaao6852.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Steeb T, Wessely A, Ruzicka T, Heppt MV, Berking C. How to MEK the best of uveal melanoma: a systematic review on the efficacy and safety of MEK inhibitors in metastatic or unresectable uveal melanoma. Eur J Cancer. 2018;103:41–51.

    Article  CAS  PubMed  Google Scholar 

  15. Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med. 2014;371:1877–88.

    Article  PubMed  CAS  Google Scholar 

  16. Ambrosini G, Musi E, Ho AL, de Stanchina E, Schwartz GK. Inhibition of mutant GNAQ signaling in uveal melanoma induces AMPK-dependent autophagic cell death. Mol Cancer Ther. 2013;12:768–76.

    Article  CAS  PubMed  Google Scholar 

  17. Shoushtari AN, Kudchadkar RR, Panageas K, Murthy RK, Jung M, Shah R, et al. A randomized phase 2 study of trametinib with or without GSK2141795 in patients with advanced uveal melanoma. J Clin Oncol. 2016;34:9511.

    Article  Google Scholar 

  18. Nagai M, Vo NH, Shin Ogawa L, Chimmanamada D, Inoue T, Chu J, et al. The oncology drug elesclomol selectively transports copper to the mitochondria to induce oxidative stress in cancer cells. Free Radic Biol Med. 2012;52:2142–50.

    Article  CAS  PubMed  Google Scholar 

  19. Tsvetkov P, Detappe A, Cai K, Keys HR, Brune Z, Ying W, et al. Mitochondrial metabolism promotes adaptation to proteotoxic stress. Nat Chem Biol. 2019;15:681–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hedley D, Shamas-Din A, Chow S, Sanfelice D, Schuh AC, Brandwein JM, et al. A phase I study of elesclomol sodium in patients with acute myeloid leukemia. Leuk Lymphoma. 2016;57:2437–40.

    Article  PubMed  Google Scholar 

  21. Monk BJ, Kauderer JT, Moxley KM, Bonebrake AJ, Dewdney SB, Secord AA, et al. A phase II evaluation of elesclomol sodium and weekly paclitaxel in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube or primary peritoneal cancer: An NRG oncology/gynecologic oncology group study. Gynecol Oncol. 2018;151:422–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Berkenblit A, Eder JP Jr., Ryan DP, Seiden MV, Tatsuta N, Sherman ML, et al. Phase I clinical trial of STA-4783 in combination with paclitaxel in patients with refractory solid tumors. Clin Cancer Res. 2007;13:584–90.

    Article  CAS  PubMed  Google Scholar 

  23. O’Day SJ, Eggermont AM, Chiarion-Sileni V, Kefford R, Grob JJ, Mortier L, et al. Final results of phase III SYMMETRY study: randomized, double-blind trial of elesclomol plus paclitaxel versus paclitaxel alone as treatment for chemotherapy-naive patients with advanced melanoma. J Clin Oncol. 2013;31:1211–8.

    Article  PubMed  CAS  Google Scholar 

  24. O’Day S, Gonzalez R, Lawson D, Weber R, Hutchins L, Anderson C, et al. Phase II, randomized, controlled, double-blinded trial of weekly elesclomol plus paclitaxel versus paclitaxel alone for stage IV metastatic melanoma. J Clin Oncol. 2009;27:5452–8.

    Article  PubMed  CAS  Google Scholar 

  25. Parry D, Guzi T, Shanahan F, Davis N, Prabhavalkar D, Wiswell D, et al. Dinaciclib (SCH 727965), a novel and potent cyclin-dependent kinase inhibitor. Mol Cancer Ther. 2010;9:2344–53.

    Article  CAS  PubMed  Google Scholar 

  26. Carlson BA, Dubay MM, Sausville EA, Brizuela L, Worland PJ. Flavopiridol induces G1 arrest with inhibition of cyclin-dependent kinase (CDK) 2 and CDK4 in human breast carcinoma cells. Cancer Res. 1996;56:2973–8.

    CAS  PubMed  Google Scholar 

  27. Gunst JD, Kjaer K, Olesen R, Rasmussen TA, Ostergaard L, Denton PW, et al. Fimepinostat, a novel dual inhibitor of HDAC and PI3K, effectively reverses HIV-1 latency ex vivo without T cell activation. J Virus Erad. 2019;5:133–7.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Harrington BS, Ozaki MK, Caminear MW, Hernandez LF, Jordan E, Kalinowski NJ, et al. drugs targeting tumor-initiating cells prolong survival in a post-surgery, post-chemotherapy ovarian cancer relapse model. Cancers. 2020;12:1645.

    Article  PubMed Central  CAS  Google Scholar 

  29. Harris IS, DeNicola GM. The Complex Interplay between Antioxidants and ROS in Cancer. Trends Cell Biol. 2020;30:440–51.

    Article  CAS  PubMed  Google Scholar 

  30. Shimada K, Reznik E, Stokes ME, Krishnamoorthy L, Bos PH, Song Y, et al. Copper-binding small molecule induces oxidative stress and cell-cycle arrest in glioblastoma-patient-derived cells. Cell Chem Biol. 2018;25:585–94.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tardito S, Bassanetti I, Bignardi C, Elviri L, Tegoni M, Mucchino C, et al. Copper binding agents acting as copper ionophores lead to caspase inhibition and paraptotic cell death in human cancer cells. J Am Chem Soc. 2011;133:6235–42.

    Article  CAS  PubMed  Google Scholar 

  32. Hasinoff BB, Wu X, Yadav AA, Patel D, Zhang H, Wang DS, et al. Cellular mechanisms of the cytotoxicity of the anticancer drug elesclomol and its complex with Cu(II). Biochem Pharmacol. 2015;93:266–76.

    Article  CAS  PubMed  Google Scholar 

  33. Kirshner JR, He S, Balasubramanyam V, Kepros J, Yang CY, Zhang M, et al. Elesclomol induces cancer cell apoptosis through oxidative stress. Mol Cancer Ther. 2008;7:2319–27.

    Article  CAS  PubMed  Google Scholar 

  34. van der Ent W, Burrello C, Teunisse AF, Ksander BR, van der Velden PA, Jager MJ, et al. Modeling of human uveal melanoma in zebrafish xenograft embryos. Investig Ophthalmol Vis Sci. 2014;55:6612–22.

    Article  CAS  Google Scholar 

  35. Kluza J, Corazao-Rozas P, Touil Y, Jendoubi M, Maire C, Guerreschi P, et al. Inactivation of the HIF-1alpha/PDK3 signaling axis drives melanoma toward mitochondrial oxidative metabolism and potentiates the therapeutic activity of pro-oxidants. Cancer Res. 2012;72:5035–47.

    Article  CAS  PubMed  Google Scholar 

  36. Li Y, Shi J, Yang J, Ge S, Zhang J, Jia R, et al. Uveal melanoma: progress in molecular biology and therapeutics. Ther Adv Med Oncol. 2020;12:1758835920965852.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Brady DC, Crowe MS, Turski ML, Hobbs GA, Yao X, Chaikuad A, et al. Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature. 2014;509:492–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70:440–6.

    Article  CAS  PubMed  Google Scholar 

  39. Nieto MA, Sargent MG, Wilkinson DG, Cooke J. Control of cell behavior during vertebrate development by Slug, a zinc finger gene. Science. 1994;264:835–9.

    Article  CAS  PubMed  Google Scholar 

  40. Wu WS, Heinrichs S, Xu D, Garrison SP, Zambetti GP, Adams JM, et al. Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell. 2005;123:641–53.

    Article  CAS  PubMed  Google Scholar 

  41. Tripathi MK, Misra S, Khedkar SV, Hamilton N, Irvin-Wilson C, Sharan C, et al. Regulation of BRCA2 gene expression by the SLUG repressor protein in human breast cells. J Biol Chem. 2005;280:17163–71.

    Article  CAS  PubMed  Google Scholar 

  42. Bolos V, Peinado H, Perez-Moreno MA, Fraga MF, Esteller M, Cano A. The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J Cell Sci. 2003;116:499–511.

    Article  CAS  PubMed  Google Scholar 

  43. Barrallo-Gimeno A, Nieto MA. The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development. 2005;132:3151–61.

    Article  CAS  PubMed  Google Scholar 

  44. Hajra KM, Chen DY, Fearon ER. The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res. 2002;62:1613–8.

    CAS  PubMed  Google Scholar 

  45. Fan H, Wang X, Li W, Shen M, Wei Y, Zheng H, et al. ASB13 inhibits breast cancer metastasis through promoting SNAI2 degradation and relieving its transcriptional repression of YAP. Genes Dev. 2020;34:1359–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Li W, Shen M, Jiang YZ, Zhang R, Zheng H, Wei Y, et al. Deubiquitinase USP20 promotes breast cancer metastasis by stabilizing SNAI2. Genes Dev. 2020;34:1310–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Asnaghi L, Gezgin G, Tripathy A, Handa JT, Merbs SL, van der Velden PA, et al. EMT-associated factors promote invasive properties of uveal melanoma cells. Mol Vis. 2015;21:919–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang Z, Li L, Wu C, Yin G, Zhu P, Zhou Y, et al. Inhibition of Slug effectively targets leukemia stem cells via the Slc13a3/ROS signaling pathway. Leukemia. 2020;34:380–90.

    Article  CAS  PubMed  Google Scholar 

  49. Wang W, Hind T, Lam BWS, Herr DR. Sphingosine 1-phosphate signaling induces SNAI2 expression to promote cell invasion in breast cancer cells. FASEB J. 2019;33:7180–91.

    Article  CAS  PubMed  Google Scholar 

  50. Ma Q. Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol. 2013;53:401–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci. 2014;39:199–218.

    Article  CAS  PubMed  Google Scholar 

  52. Albayrak G, Korkmaz FD, Tozcu D, Dogan, Turacli I. The outcomes of an impaired powerhouse in KRAS mutant lung adenocarcinoma cells by Elesclomol. J Cell Biochem. 2019;120:10564–71.

    Article  CAS  PubMed  Google Scholar 

  53. Qu Y, Wang J, Sim MS, Liu B, Giuliano A, Barsoum J, et al. Elesclomol, counteracted by Akt survival signaling, enhances the apoptotic effect of chemotherapy drugs in breast cancer cells. Breast Cancer Res Treat. 2010;121:311–21.

    Article  CAS  PubMed  Google Scholar 

  54. Brouwer NJ, Konstantinou EK, Gragoudas ES, Marinkovic M, Luyten GPM, Kim IK, et al. Targeting the YAP/TAZ pathway in uveal and conjunctival melanoma with verteporfin. Investig Ophthalmol Vis Sci. 2021;62:3.

    Article  CAS  Google Scholar 

  55. White SM, Avantaggiati ML, Nemazanyy I, Di Poto C, Yang Y, Pende M, et al. YAP/TAZ inhibition induces metabolic and signaling rewiring resulting in targetable vulnerabilities in NF2-deficient tumor cells. Dev Cell. 2019;49:425–43.e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Aubert L, Nandagopal N, Steinhart Z, Lavoie G, Nourreddine S, Berman J, et al. Copper bioavailability is a KRAS-specific vulnerability in colorectal cancer. Nat Commun. 2020;11:3701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Shanbhag V, Jasmer-McDonald K, Zhu S, Martin AL, Gudekar N, Khan A, et al. ATP7A delivers copper to the lysyl oxidase family of enzymes and promotes tumorigenesis and metastasis. Proc Natl Acad Sci USA. 2019;116:6836–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. De Waard-Siebinga I, Blom DJ, Griffioen M, Schrier PI, Hoogendoorn E, Beverstock G, et al. Establishment and characterization of an uveal-melanoma cell line. Int J Cancer. 1995;62:155–61.

    Article  PubMed  Google Scholar 

  59. Chen PW, Murray TG, Uno T, Salgaller ML, Reddy R, Ksander BR. Expression of MAGE genes in ocular melanoma during progression from primary to metastatic disease. Clin Exp Metastasis. 1997;15:509–18.

    Article  CAS  PubMed  Google Scholar 

  60. Luyten GP, Naus NC, Mooy CM, Hagemeijer A, Kan-Mitchell J, Van Drunen E, et al. Establishment and characterization of primary and metastatic uveal melanoma cell lines. Int J Cancer. 1996;66:380–7.

    Article  CAS  PubMed  Google Scholar 

  61. Ksander BR, Rubsamen PE, Olsen KR, Cousins SW, Streilein JW. Studies of tumor-infiltrating lymphocytes from a human choroidal melanoma. Investig Ophthalmol Vis Sci. 1991;32:3198–208.

    CAS  Google Scholar 

  62. Amirouchene-Angelozzi N, Nemati F, Gentien D, Nicolas A, Dumont A, Carita G, et al. Establishment of novel cell lines recapitulating the genetic landscape of uveal melanoma and preclinical validation of mTOR as a therapeutic target. Mol Oncol. 2014;8:1508–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Seftor EA, Meltzer PS, Kirschmann DA, Pe’er J, Maniotis AJ, Trent JM, et al. Molecular determinants of human uveal melanoma invasion and metastasis. Clin Exp Metastasis. 2002;19:233–46.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research was supported by the National Natural Science Foundation of China (grant 82073889), the Science and Technology Commission of Shanghai (20DZ2270800), the Shanghai Municipal Science and Technology Major Project (19JC1410200), the National Facility for Translational Medicine (Shanghai) (TMSZ-2020-206), the Innovative Research Team of High-level Local Universities in Shanghai (SHSMU- ZDCX20210900), and startup funding of State Key Laboratory of Medical Genomics to JZ. We thank Drs. David E. Fisher and Xu Wu (Massachusetts General Hospital), Yantao Chen, and Yunqi Li (Shanghai Jiao Tong University) for review of this work and discussions on various topics relating to this project.

Author information

Authors and Affiliations

Authors

Contributions

JZ, YL, and JY designed and performed the experiments; JY, SX, and SG were responsible for the animal model; YL, JY, QZ, XX, and RJ analyzed the data; WS designed the ES analog ELE-003; YL, JZ, MJJ, and XF drafted the paper. All authors read and approved the final paper.

Corresponding authors

Correspondence to Renbing Jia, Jianming Zhang or Xianqun Fan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

For the patient-derived cell line and the tissue used for the PDX model, written informed consent was obtained from all involved patients. The process was approved by the ethics committee of Shanghai Ninth People’s Hospital (reference number: 20202356). For animal experiments, all procedures followed were in accordance with the Declaration of Helsinki. All animal research was conducted in accordance with the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research, all procedures were reviewed and approved by Shanghai Ninth People’s Hospital Central Lab IACUC (Permit Number: SYXK (Shanghai) 2016-0016), and all experiments conformed to the relevant regulatory standards. The experimental design followed the Guide for the Care and Use of Laboratory Animals of AAALAC international.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Y., Yang, J., Zhang, Q. et al. Copper ionophore elesclomol selectively targets GNAQ/11-mutant uveal melanoma. Oncogene 41, 3539–3553 (2022). https://doi.org/10.1038/s41388-022-02364-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02364-0

This article is cited by

Search

Quick links