Notch receptors as a therapeutic target in melanoma: a narrative bibliographic review

Conteúdo do artigo principal

Renato Santos de Oliveira Filho
Guilherme Routh Peixoto
Lorena Dal Collina Sangiuliano
Daniel Arcuschin de Oliveira

Resumo

O melanoma é o câncer de pele com maior mortalidade e mais e mais casos surgem a cada ano. A superexpressão de elementos da via de sinalização Notch já foi encontrada em linhagens de melanoma primário e metastático e está diretamente correlacionada ao desenvolvimento, crescimento, angiogênese, metástase e resistência ao tratamento do melanoma. Assim, a manipulação de Notch apresenta alto potencial terapêutico. A presente revisão teve como objetivo realizar uma revisão sobre os possíveis tratamentos do melanoma pela inibição da via de sinalização Notch. Pesquisamos a literatura existente sobre os inibidores da via de sinalização Notch em melanoma cutâneo humano, publicada entre 2000 e 2020, usando as bases de dados MEDLINE (via PubMed), LILACS (via Virtual Health Library) e Cochrane Library. Os artigos selecionados foram analisados, resumidos, tabelados e utilizados para a produção da presente revisão narrativa. Nos 24 artigos selecionados e em artigos referenciados neles, apresentaram-se como terapias os inibidores de γ-secretase (GSIs), principalmente, mas também gliotoxina, honokiol, fosfolipase A2, andrografolide e anticorpos monoclonais, que, entretanto, não foram estudados diretamente no melanoma. Outra terapia que interfere indiretamente na via de sinalização Notch e foi encontrada nesses artigos foram os inibidores de G9a. Com a revisão foi possível concluir que GSIs provavelmente não são a melhor opção para o tratamento do melanoma, excedendo cenários específicos ou pelo seu uso concomitante com outros inibidores de vias. Já o uso dos demais compostos tem maior potencial, porém mais estudos são necessários para comprovar sua eficácia e viabilidade para o tratamento do melanoma cutâneo humano.  


Palavras-chave: Melanoma;  inibidores; Receptores Notch

Detalhes do artigo

Como Citar
1.
Santos de Oliveira Filho R, Routh Peixoto G, Dal Collina Sangiuliano L, Arcuschin de Oliveira D. Notch receptors as a therapeutic target in melanoma: a narrative bibliographic review. Braz. J. Nat. Sci [Internet]. 3º de abril de 2021 [citado 22º de outubro de 2024];4(1):614 - 628. Disponível em: https://bjns.com.br/index.php/BJNS/article/view/138
Seção
Artigo em fluxo contínuo

Referências

Liu Y, Sheikh MS. Melanoma: Molecular Pathogenesis and Therapeutic Management. Mol Cell Pharmacol. 2014; 6(3):228.

IARC (a): Global Cancer Observatory. Cancer Today, 2018. Available form: https://gco.iarc.fr/today. Accessed in 2020 (Sep 27)

IARC (b): Global Cancer Observatory. Cancer Tomorrow, 2018. Available from https://gco.iarc.fr/tomorrow (2018). Accessed 2020 (Sep 27).

Liu J, Fukunaga-Kalabis M, Li L, Herlyn M. Developmental pathways activated in melanocytes and melanoma. Arch Biochem Biophys. 2014; 563: 13-21.

Oliveira Filho RS, Soares AL, Paschoal FM, Rezze GG, Oliveira E, Macarenco R, et al. Literature review of Notch melanoma receptors. Surgical and Experimental Pathology. 2019; 2(1): 1-6.

Sangiuliano LD, de Oliveira Filho RS, de Oliveira DA, Gomes HC, Ferreira LM. Identification and quantification of notch receptors in human cutaneous melanoma using molecular biology techniques: literature review. Surgical and Experimental Pathology. 2020; 3(1): 1-9.

Howard JD, Moriarty WF, Park J, Riedy K, Panova IP, Chung CH, et al. Notch signaling mediates melanoma–endothelial cell communication and melanoma cell migration. Pigment Cell Melanoma Res. 2013; 25(5):697-707.

Huynh C, Poliseno L, Segura MF, Medicherla R, Haimovic A, Menendez S, et al. The novel gamma secretase inhibitor RO4929097 reduces the tumor initiating potential of melanoma. PloS one. 2011; 6(9):e25264.

Massi D, Tarantini F, Franchi A, Paglierani M, Di Serio C, Pellerito S, et al. Evidence for differential expression of Notch receptors and their ligands in melanocytic nevi and cutaneous malignant melanoma. Mod Pathol. 2006; 19:246–254

Golan T, Messer AR, Amitai-Lange A, Melamed Z, Ohana R, Bell RE, et al. Interactions of melanoma cells with distal keratinocytes trigger metastasis via Notch signaling inhibition of MITF. Mol Cell. 2015; 59(4): 664-676.

Yamamoto S. Making sense out of missense mutations: Mechanistic dissection of Notch receptors through structure‐function studies in Drosophila. Develop Growth Differ. 2019; 62(1): 15-34.

Luger K, Rechsteiner TJ, Flaus AJ, Waye MM, Richmond TJ. Characterization of nucleosome core particles containing histone proteins made in bacteria. J Mol Biol. 1997; 272(3):301–11.

Shiio Y, Eisenman RN. Histone sumoylation is associated with transcriptional repression. Proc Natl Acad Sci U S A. 2003; 100(23):13225–30.

Biran A, Meshorer E. Concise review: chromatin and genome organization in reprogramming. Stem Cells. 2012; 30(9):1793–9.

Jenuwein T, Allis CD. Translating the histone code. Science. 2001; 293(5532):1074–80.

Martin C, Zhang Y. The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol. 2005; 6(11):838–49.

Black JC, Van Rechem C, Whetstine JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell. 2012; 48(4):491–507.

Sun XJ, Wei J, Wu XY, Hu M, Wang L, Wang HH, et al. Identification and characterization of a novel human histone H3 lysine 36-specific methyltransferase. J Biol Chem. 2005; 280(42):35261–71.

Chen MW, Hua KT, Kao HJ, Chi CC, Wei LH, Johansson G, et al. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res. 2010; 70(20):7830–40.

Wozniak RJ, Klimecki WT, Lau SS, Feinstein Y, Futscher BW. 5-Aza-2’-deoxycytidine-mediated reductions in G9A histone methyltransferase and histone H3 K9 di-methylation levels are linked to tumor suppressor gene reactivation. Oncogene. 2007; 26(1):77–90.

Dang NN, Jiao J, Meng X, An Y, Han C, Huang S. Abnormal overexpression of G9a in melanoma cells promotes cancer progression via upregulation of the Notch1 signaling pathway. Aging (Albany NY). 2020 Feb 3;12(3):2393-2407.

Balint K, Xiao M, Pinnix C, Soma A, Veres I, Juhansz I, et al. Activation of Notch1 signaling is required for beta-catenin-mediated 6 human primary melanoma progression. J Clin Invest. 2005; 115(11): 3166–3176.

Bedogni B, Warneke JA, Nickoloff BJ, Giaccia AJ, Powell MB. Notch1 is an effector of Akt and hypoxia in melanoma development. J Clin Invest. 2008; 118(11):3660-70.

Bhat KM, Maddodi N, Shashikant C, Setaluri V. Transcriptional regulation of human MAP2 gene in melanoma: role of neuronal bHLH factors and Notch1 signaling. Nucleic Acids Res. 2006;34(13):3819-32.

Liu S, Tetzlaff MT, Wang T, Chen X, Yang R, Kumar SM, Vultur A, Li P, Martin JS, Herlyn M, Amaravadi R, Li B, Xu X. Hypoxia-activated prodrug enhances therapeutic effect of sunitinib in melanoma. Oncotarget. 2017;8(70):115140-115152.

Lin X, Sun B, Zhu D, Zhao X, Sun R, Zhang Y, Zhang D, Dong X, Gu Q, Li Y, Liu F. Notch4+ cancer stem-like cells promote the metastatic and invasive ability of melanoma. Cancer Sci. 2016;107(8):1079-91.

Keyghobadi F, Mehdipour M, Nekoukar V, Firouzi J, Kheimeh A, Nobakht Lahrood F, Azimian Zavareh V, Azimi M, Mohammadi M, Sodeifi N, Ebrahimi M. Long-Term Inhibition of Notch in A-375 Melanoma Cells Enhances Tumor Growth Through the Enhancement of AXIN1, CSNK2A3, and CEBPA2 as Intermediate Genes in Wnt and Notch Pathways. Front Oncol. 2020;10:531.

Nair JS, Sheikh T, Ho AL, Schwartz GK. PTEN regulates sensitivity of melanoma cells to RO4929097, the γ-secretase inhibitor. Anticancer research. 2013; 33(4): 1307-16.

Gajewski TF. Molecular profiling of melanoma and the evolution of patient-specific therapy. Semin Oncol. 2011;38(2):236-42.

Lee SM, Moon J, Redman BG, Chidiac T, Flaherty LE, Zha Y, et al. Phase 2 study of RO4929097, a gamma-secretase inhibitor, in metastatic melanoma: SWOG 0933. Cancer. 2015;121(3):432-440.

Krepler C, Xiao M, Samanta M, Vultur A, Chen HY, Brafford P, et al. Targeting Notch enhances the efficacy of ERK inhibitors in BRAF-V600E melanoma. Oncotarget. 2016; 7(4): 712211.

Skarmoutsou E, Bevelacqua V, D' Amico F, Russo A, Spandidos DA, Scalisi A, et al. FOXP3 expression is modulated by TGF‑β1/NOTCH1 pathway in human melanoma. Int J Mol Med. 2018; 42(1): 392–404.

Zhang, K., Wong, P., Zhang, L., Jacobs, B., Borden, E. C., Aster, J. C., & Bedogni, B. (2012). A Notch1–neuregulin1 autocrine signaling loop contributes to melanoma growth. Oncogene. 2012;31: 4609–4618

Zhang K, Wong P, Salvaggio C, Salhi A, Osman I, Bedogni, B. Synchronized targeting of Notch and ERBB signaling suppresses melanoma tumor growth through inhibition of Notch1 and ERBB3. J Invest Dermatol. 2016; 136(2):464-472.

González-Cao M, Rodón J, Karachaliou N, Sánchez J, Santarpia M, Viteri S, Pilotto S, Teixidó C, Riso A, Rosell R. Other targeted drugs in melanoma. Ann Transl Med. 2015;3(18):266.

Dantonio PM, Klein MO, Freire MRVB, Araujo CN, Chiacetti AC, Correa RG. Exploring major signaling cascades in melanomagenesis: a rationale route for targetted skin cancer therapy. Biosci Rep. 2018;38(5):BSR20180511.

Shahbazian MD, Zhang K, Grunstein M. Histone H2B ubiquitylation controls processive methylation but not monomethylation by Dot1 and Set1. Mol Cell. 2005; 19(2):271–7.

Liu F, Barsyte-Lovejoy D, Li F, Xiong Y, Korboukh V, Huang XP, et al. Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP. J Med Chem. 2013; 56(21):8931–42.

Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007; 129(4):823–37.

Collins R, Cheng X. A case study in cross-talk: the histone lysine methyltransferases G9a and GLP. Nucleic Acids Res. 2010; 38(11):3503–11.

Ke XX, Zhang D, Zhu S, Xia Q, Xiang Z, Cui H. Inhibition of H3K9 methyltransferase G9a repressed cell proliferation and induced autophagy in neuroblastoma cells. PLoS One. 2014; 9(9):e106962.

Casciello F, Windloch K, Gannon F and Lee JS. Functional role of G9a histone methyltransferase in cancer. Front. Immunol. 2015; 6:487.

Kaniskan HU, Martini ML, and Jin J. Inhibitors of Protein Methyltransferases an Demethylases. Chem . Rev. 2018;118:989-1068.

Hubmann R, Sieghart W, Schnabl S, Araghi M, Hilgarth M, Reiter M, et al. Gliotoxin targets nuclear Notch2 in human solid tumor derived cell lines in vitro and inhibits melanoma growth in xenograft mouse model. Front Pharmacol. 2017; 8: 319.

Khunsap S, Khow O, Buranapraditkun S, Suntrarachun S, Puthong S, Boonchang, S. Anticancer properties of phospholipase A2 from Daboia siamensis venom on human skin melanoma cells. J Venom Anim Toxins incl Trop Dis. 2016; 22(1): 1-8

Strickland LR, Pal HC, Elmets CA, Afaq F. Targeting drivers of melanoma with synthetic small molecules and phytochemicals. Cancer Lett. 2015;359(1):20-35.

Kaushik G, Ramalingam S, Subramaniam D, Rangarajan P, Protti P, Rammamoorthy P, Anant S, Mammen JM. Honokiol induces cytotoxic and cytostatic effects in malignant melanoma cancer cells. Am J Surg. 2012; 204(6):868-73.

Kaushik G, Venugopal A, Ramamoorthy P, Standing D, Subramaniam D, Umar S, et al. Honokiol inhibits melanoma stem cells by targeting notch signaling. Mol Carcin. 2015; 54(12):1710-21.

Kumar D, Gorain M, Kundu G, Kundu GC. Therapeutic implications of cellular and molecular biology of cancer stem cells in melanoma. Mol Cancer. 2017;16(1):7.

Artigos mais lidos pelo mesmo(s) autor(es)