Chonnam Med J.  2018 Sep;54(3):135-142. 10.4068/cmj.2018.54.3.135.

Melanoma Cell Death Mechanisms

Affiliations
  • 1Department of Dermatology, University of Colorado Denver School of Medicine, Aurora, CO, USA. peter.song@ucdenver.edu
  • 2Department of Internal Medicine, University of Colorado Denver School of Medicine, Aurora, CO, USA.
  • 3Marian University College of Osteopathic Medicine, Indianapolis, IN, USA.

Abstract

Over recent years, several new molecular and immunogenic therapeutic approaches to melanoma treatment have been approved and implemented in clinical practice. Mechanisms of resistance to these new therapies have become a major problem. Mutation-specific pharmacotherapy can result in simultaneous emergence of resistant clones at many separate body sites despite an initially positive therapeutic response. Additionally, treatments aimed at inducing apoptosis are subject to resistance due to escape through other known mechanisms of regulated cell death (RCD). In this review, we discuss the complexity in pharmacological manipulation of melanoma with c-Kit, BRAF, MEK, and/or mTOR mutant cell lines. This study also addresses melanoma evasion of cell death through modalities of RCD such as apoptosis, autophagy, and necroptosis. This study also examines new combination therapies which have been approved to target both cell cycle dysregulation and cell death pathways. Lastly, we recognize the importance of immunomodulation though manipulation of the body's natural killing mechanisms with CTLA4, PD1, and CSF1 inhibition. As we begin to recognize tumor cell activation of alternate pathways, evasion of programmed cell death, and manipulation of the tumor microenvironment, it is increasingly important to grasp the complexity of personalized therapy in melanoma treatment.

Keyword

Melanoma; Autophagy; Apoptosis; CTLA4 protein, human; TOR Serine-Threonine Kinases

MeSH Terms

Apoptosis
Autophagy
Cell Cycle
Cell Death*
Cell Line
Clone Cells
Drug Therapy
Hand Strength
Homicide
Humans
Immunomodulation
Melanoma*
TOR Serine-Threonine Kinases
Tumor Microenvironment
United Nations
TOR Serine-Threonine Kinases

Figure

  • FIG. 1 Regulated Cell Death (RCD) in Melanoma: Apoptosis, Autophagy, and Necroptosis. Classification of basic Caspase-dependent and Caspase-independent cell death processes and key players.21222529

  • FIG. 2 Mechanisms of Autophagy Initiation in Melanoma: (A) Macroautophagy. Initiation of autophagosome formation requires the ULK1-Atg13-FIP200 complex and Beclin1-class III PI3K complexes along with two conjugation systems, Atg12-Atg5-Atg16 and Atg8-PE. LC3 proteolysis and lipidation is involved in the elongation phase. Cytoplasmic matter is engulfed by the phagophore surrounded by a double-membrane vesicle referred to as the autophagosome. Next, the autophagosome fuses with a lysosome to form an autolysosome. The contents within the autolysosome are degraded by the lysosome. Low ATP levels and hypoxia induce autophagy. Conversely, mTOR is activated in a nutrient rich environment which ultimately inhibits autophagy through suppression of ULK1 activity. Insulin and other growth factors can also stimulate phosphorylation and activation of mTOR. (B) Microautophagy. Cellular contents are directly sequestered into the lysosome by self-invagination of lysosomal membranes. (C) Chaperone-mediated autophagy. Hsc70 or Hsc90 chaperone facilitates binding of cytosolic proteins to LAMP2A receptor resulting in lysosomal translocation and internalization.252627


Reference

1. Mattia G, Puglisi R, Ascione B, Malorni W, Carè A, Matarrese P. Cell death-based treatments of melanoma:conventional treatments and new therapeutic strategies. Cell Death Dis. 2018; 9:112.
Article
2. Whiteman DC, Green AC, Olsen CM. The growing burden of invasive melanoma: projections of incidence rates and numbers of new cases in six susceptible populations through 2031. J Invest Dermatol. 2016; 136:1161–1171.
Article
3. Carvajal RD. Targeting KIT for treatment of advanced melanoma. Melanoma Lett Winter. 2011; 29.
4. Amann VC, Ramelyte E, Thurneysen S, Pitocco R, Bentele-Jaberg N, Goldinger SM, et al. Developments in targeted therapy in melanoma. Eur J Surg Oncol. 2017; 43:581–593.
Article
5. Smalley KS, Sondak VK. Melanoma--an unlikely poster child for personalized cancer therapy. N Engl J Med. 2010; 363:876–878.
Article
6. Cheng L, Lopez-Beltran A, Massari F, MacLennan GT, Montironi R. Molecular testing for BRAF mutations to inform melanoma treatment decisions: a move toward precision medicine. Mod Pathol. 2018; 31:24–38.
Article
7. Hayward NK, Wilmott JS, Waddell N, Johansson PA, Field MA, Nones K, et al. Whole-genome landscapes of major melanoma subtypes. Nature. 2017; 545:175–180.
Article
8. Moon KR, Choi YD, Kim JM, Jin S, Shin MH, Shim HJ, et al. Genetic alterations in primary acral melanoma and acral melanocytic nevus in Korea: common mutated genes show distinct cytomorphological features. J Invest Dermatol. 2018; 138:933–945.
Article
9. Hodi FS, Corless CL, Giobbie-Hurder A, Fletcher JA, Zhu M, Marino-Enriquez A, et al. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J Clin Oncol. 2013; 31:3182–3190.
Article
10. Guo J, Si L, Kong Y, Flaherty KT, Xu X, Zhu Y, et al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol. 2011; 29:2904–2909.
Article
11. Gounder MM, Maki RG. Molecular basis for primary and secondary tyrosine kinase inhibitor resistance in gastrointestinal stromal tumor. Cancer Chemother Pharmacol. 2011; 67:Suppl 1. S25–S43.
Article
12. Boni A, Cogdill AP, Dang P, Udayakumar D, Njauw CN, Sloss CM, et al. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res. 2010; 70:5213–5219.
Article
13. Manzano JL, Layos L, Bugés C, de Los Llanos Gil M, Vila L, Martínez-Balibrea E, et al. Resistant mechanisms to BRAF inhibitors in melanoma. Ann Transl Med. 2016; 4:237.
Article
14. Ryu S, Youn C, Moon AR, Howland A, Armstrong CA, Song PI. Therapeutic inhibitors against mutated BRAF and MEK for the treatment of metastatic melanoma. Chonnam Med J. 2017; 53:173–177.
Article
15. Welsh SJ, Rizos H, Scolyer RA, Long GV. Resistance to combination BRAF and MEK inhibition in metastatic melanoma: where to next? Eur J Cancer. 2016; 62:76–85.
Article
16. Grimaldi AM, Simeone E, Festino L, Vanella V, Strudel M, Ascierto PA. MEK inhibitors in the treatment of metastatic melanoma and solid tumors. Am J Clin Dermatol. 2017; 18:745–754.
Article
17. Lugowska I, Koseła-Paterczyk H, Kozak K, Rutkowski P. Trametinib: a MEK inhibitor for management of metastatic melanoma. Onco Targets Ther. 2015; 8:2251–2259.
18. Cobimetinib for metastatic melanoma. Aust Prescr. 2017; 40:30–31.
19. Kim DW, Patel SP. Profile of selumetinib and its potential in the treatment of melanoma. Onco Targets Ther. 2014; 7:1631–1639.
20. Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet. 2015; 386:444–451.
Article
21. Karbowniczek M, Spittle CS, Morrison T, Wu H, Henske EP. MTOR is activated in the majority of malignant melanomas. J Invest Dermatol. 2008; 128:980–987.
Article
22. Kong Y, Si L, Li Y, Wu X, Xu X, Dai J, et al. Analysis of mTOR gene aberrations in melanoma patients and evaluation of their sensitivity to PI3K-AKT-mTOR pathway inhibitors. Clin Cancer Res. 2016; 22:1018–1027.
Article
23. Vera Aguilera J, Rao RD, Allred JB, Suman VJ, Windschitl HE, Kaur JS, et al. Phase II study of Everolimus in metastatic malignant melanoma (NCCTG-N0377, alliance). Oncologist. 2018; 23:887–e94.
Article
24. Hirayama Y, Gi M, Yamano S, Tachibana H, Okuno T, Tamada S, et al. Anti-PD-L1 treatment enhances antitumor effect of everolimus in a mouse model of renal cell carcinoma. Cancer Sci. 2016; 107:1736–1744.
Article
25. Wang Y, Wang XY, Subjeck JR, Shrikant PA, Kim HL. Temsirolimus, an mTOR inhibitor, enhances anti-tumour effects of heat shock protein cancer vaccines. Br J Cancer. 2011; 104:643–652.
Article
26. Xie X, White EP, Mehnert JM. Coordinate autophagy and mTOR pathway inhibition enhances cell death in melanoma. PLoS One. 2013; 8:e55096.
Article
27. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on Cell death 2018. Cell Death Differ. 2018; 25:486–541.
28. Adewale FO, Basiru AO, Ayorinde OO, Israel OI, Oluwafemi OA. Regulation of apoptotic and necroptotic cell death in skin cancer. J Cancer Biol Res. 2017; 5:1108–1117.
29. Wong RS. Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res. 2011; 30:87.
Article
30. Avery-Kiejda KA, Bowden NA, Croft AJ, Scurr LL, Kairupan CF, Ashton KA, et al. P53 in human melanoma fails to regulate target genes associated with apoptosis and the cell cycle and may contribute to proliferation. BMC Cancer. 2011; 11:203.
Article
31. Vucic D, Stennicke HR, Pisabarro MT, Salvesen GS, Dixit VM. ML-IAP, a novel inhibitor of apoptosis that is preferentially expressed in human melanomas. Curr Biol. 2000; 10:1359–1366.
Article
32. Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol. 2010; 221:3–12.
Article
33. Duffy A, Le J, Sausville E, Emadi A. Autophagy modulation: a target for cancer treatment development. Cancer Chemother Pharmacol. 2015; 75:439–447.
Article
34. Cheng Y, Ren X, Hait WN, Yang JM. Therapeutic targeting of autophagy in disease: biology and pharmacology. Pharmacol Rev. 2013; 65:1162–1197.
Article
35. Egger ME, Huang JS, Yin W, McMasters KM, McNally LR. Inhibition of autophagy with chloroquine is effective in melanoma. J Surg Res. 2013; 184:274–281.
Article
36. Kaczmarek A, Vandenabeele P, Krysko DV. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity. 2013; 38:209–223.
Article
37. Geserick P, Wang J, Schilling R, Horn S, Harris PA, Bertin J, et al. Absence of RIPK3 predicts necroptosis resistance in malignant melanoma. Cell Death Dis. 2015; 6:e1884.
Article
38. Caldarola G, Carbone A, Arena V, Pennacchia I, De Waure C, Vianale G, et al. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL): a possible pathogenic role in chronic plaque psoriasis. G Ital Dermatol Venereol. 2016; 151:17–24.
39. Karlsson AK, Saleh SN. Checkpoint inhibitors for malignant melanoma: a systematic review and meta-analysis. Clin Cosmet Investig Dermatol. 2017; 10:325–339.
Article
40. Neubert NJ, Schmittnaegel M, Bordry N, Nassiri S, Wald N, Martignier C, et al. T cell-induced CSF1 promotes melanoma resistance to PD1 blockade. Sci Transl Med. 2018; 10:eaan3311.
Article
Full Text Links
  • CMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr