Comparison between antineoplastic treatments with selective inhibitors (BRAF/MEK) and the new potentials combinatorial therapies for metastatic melanoma

Authors

DOI:

https://doi.org/10.33448/rsd-v11i14.36275

Keywords:

Melanoma; Immunotherapy; Drug therapy; Quality of life; BRAF Kinase; MEK.

Abstract

Melanoma is responsible for most skin cancer related mortalities, and its best prognosis is related to early diagnosis and treatment. In this sense much has been studied about the treatment of the disease, relating mainly to selective inhibitors of signaling pathways. Therefore, the present review addresses the use of monotherapies of BRAF (vemurafenib and dabrafenib) and MEK (trametinib) inhibitors, the possible combinations between them and other therapeutic options that may be promising to participate on the neoadjuvant and adjuvant treatment with the selective inhibitors, since the main limitations in treatment with these inhibitors is the frequent development of drug resistance mechanisms. In order to obtain the necessary content, a bibliographic review was carried out in the following databases of the Virtual Health Library, PubMed, Scielo and Google Academic, using the following keywords: melanoma, BRAF and MEK inhibitors, dabrafenib monotherapy and vemurafenib, dabrafenib and trametinib combinations, protein disulfide isomerase action, immunotherapy in the treatment of metastatic melanoma. After the information survey it was possible to conclude that dabrafenib + trametinib combination therapy is more advantageous in view of the adverse effects, quality of life and progression free survival of the patient when compared directly to selective chemotherapy monotherapies, and this may be considered for future studies that include, for example, combination with immunotherapy or quercetin, a potential natural inhibitor for PDI.

References

Araujo, T. L. S., Zeidler, J. D., Oliveira, P. V. S., Dias, M. H., Armelin, H. A., & Laurindo, F. R. M. (2017). Protein disulfide isomerase externalization in endothelial cells follows classical and unconventional routes. Free Radical Biology and Medicine, 103, 199–208. https://doi.org/10.1016/j.freeradbiomed.2016.12.021

Aydin, E., Johansson, J., Nazir, F. H., Hellstrand, K., & Martner, A. (2017). Role of NOX2-Derived Reactive Oxygen Species in NK Cell–Mediated Control of Murine Melanoma Metastasis. Cancer Immunology Research, 5(9), 804–811. https://doi.org/10.1158/2326-6066.CIR-16-0382

Brandes, N., Schmitt, S., & Jakob, U. (2009). Thiol-Based Redox Switches in Eukaryotic Proteins. Antioxidants & Redox Signaling, 11(5), 997–1014. https://doi.org/10.1089/ars.2008.2285

Câmara de Regulação do Mercado de Medicamentos. (2022). Preços Máximos de Medicamentos por Princípio ativo. Retrieved August 01, 2022 from https://www.gov.br/anvisa/pt-br/assuntos/medicamentos/cmed/precos

Cellone Trevelin, S., & Rossetti Lopes, L. (2015). Protein disulfide isomerase and Nox: new partners in redox signaling. Current Pharmaceutical Design, 21(41), 5951–5963.

Cesi, G., Walbrecq, G., Zimmer, A., Kreis, S., & Haan, C. (2017). ROS production induced by BRAF inhibitor treatment rewires metabolic processes affecting cell growth of melanoma cells. Molecular Cancer, 16(1). https://doi.org/10.1186/s12943-017-0667-y

Comissão Nacional de Incorporação de Tecnologias no SUS. (2020). Terapia-alvo (Vemurafenibe, Dabrafenibe, Cobimetinibe, Trametinibe) e Imunoterapia (Ipilimumabe, Nivolumabe, Pembrolizumabe) para o Tratamento de Primeira Linha do Melanoma Avançado não-cirúrgico e Metastático. https://www.gov.br/conitec/pt-br/midias/relatorios/2020/relatorio_541_terapiaalvo_melanoma_final_2020.pdf

Czarnecka, A. M., Ostaszewski, K., Borkowska, A., Szumera-Ciećkiewicz, A., Kozak, K., Świtaj, T., Rogala, P., Kalinowska, I., Koseła-Paterczyk, H., Zaborowski, K., Teterycz, P., Tysarowski, A., Makuła, D., & Rutkowski, P. (2021). Efficacy of Neoadjuvant Targeted Therapy for Borderline Resectable III B-D or IV Stage BRAF V600 Mutation-Positive Melanoma. Cancers, 14(1), 110. https://doi.org/10.3390/cancers14010110

Damsky, W. E., Rosenbaum, L. E., & Bosenberg, M. (2010). Decoding Melanoma Metastasis. Cancers, 3(1), 126–163. https://doi.org/10.3390/cancers3010126

Davies, H., Bignell, G. R., Cox, C., Stephens, P., Edkins, S., Clegg, S., Teague, J., Woffendin, H., Garnett, M. J., Bottomley, W., Davis, N., Dicks, E., Ewing, R., Floyd, Y., Gray, K., Hall, S., Hawes, R., Hughes, J., Kosmidou, V., … Futreal, P. A. (2002). Mutations of the BRAF gene in human cancer. In Nature (Vol. 417, Issue 6892, pp. 949–954). https://doi.org/10.1111/ced.12015

Duggan, M. C., Stiff, A. R., Bainazar, M., Regan, K., Olaverria Salavaggione, G. N., Maharry, S., Blachly, J. S., Krischak, M., Walker, C. J., Latchana, N., Tridandapani, S., de La Chapelle, A., Eisfeld, A. K., & Carson, W. E. (2017). Identification of NRAS isoform 2 overexpression as a mechanism facilitating BRAF inhibitor resistance in malignant melanoma. Proceedings of the National Academy of Sciences of the United States of America, 114(36), 9629–9634. https://doi.org/10.1073/pnas.1704371114

Fidler, I. J. (2003). The pathogenesis of cancer metastasis: the “seed and soil” hypothesis revisited. Nature Reviews Cancer, 3(6), 453–458. https://doi.org/10.1038/nrc1098

Galvão, M. C. B., & Ricarte, I. L. M. (2019). Revisão sistemática da literatura: conceituação, produção e publicação. Logeion: Filosofia Da Informação, 6(1), 57–73. https://doi.org/10.21728/logeion.2019v6n1.p57-73

Goplen, D., Wang, J., Enger, P. Ø., Tysnes, B. B., Terzis, A. J. A., Laerum, O. D., & Bjerkvig, R. (2006). Protein Disulfide Isomerase Expression Is Related to the Invasive Properties of Malignant Glioma. Cancer Research, 66(20), 9895–9902. https://doi.org/10.1158/0008-5472.CAN-05-4589

Halmos, B., Burke, T., Kalyvas, C., Insinga, R., Vandormael, K., Frederickson, A., & Piperdi, B. (2022). Indirect comparison of pembrolizumab monotherapy versus nivolumab + ipilimumab in first-line metastatic lung cancer. Immunotherapy, 14(5), 295–307. https://doi.org/10.2217/imt-2021-0273

Hauschild, A., Grob, J. J., Demidov, L. v., Jouary, T., Gutzmer, R., Millward, M., Rutkowski, P., Blank, C. U., Miller, W. H., Kaempgen, E., Martín-Algarra, S., Karaszewska, B., Mauch, C., Chiarion-Sileni, V., Martin, A. M., Swann, S., Haney, P., Mirakhur, B., Guckert, M. E., … Chapman, P. B. (2012). Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. The Lancet, 380(9839), 358–365. https://doi.org/10.1016/S0140-6736(12)60868-X

Hughes, T., Klairmont, M., Sharfman, W. H., & Kaufman, H. L. (2021). Interleukin-2, Ipilimumab, and Anti-PD-1: clinical management and the evolving role of immunotherapy for the treatment of patients with metastatic melanoma. Cancer Biology & Therapy, 22(10–12), 513–526. https://doi.org/10.1080/15384047.2015.1095401

Instituto Nacional de Câncer José Alencar Gomes da Silva. (2019). Estimativa 2020 - Incidência de Câncer no Brasil. http://www.inca.gov.br

Kramkimel, N., Thomas-Schoemann, A., Sakji, L., Golmard, J. L., Noe, G., Regnier-Rosencher, E., Chapuis, N., Maubec, E., Vidal, M., Avril, M. F., Goldwasser, F., Mortier, L., Dupin, N., & Blanchet, B. (2016). Vemurafenib pharmacokinetics and its correlation with efficacy and safety in outpatients with advanced BRAF-mutated melanoma. Targeted Oncology, 11(1), 59–69. https://doi.org/10.1007/s11523-015-0375-8

Lee, E., & Lee, D. H. (2017). Emerging roles of protein disulfide isomerase in cancer. In BMB Reports (Vol. 50, Issue 8, pp. 401–410). The Biochemical Society of the Republic of Korea. https://doi.org/10.5483/BMBRep.2017.50.8.107

Lidsky, M., Antoun, G., Speicher, P., Adams, B., Turley, R., Augustine, C., Tyler, D., & Ali-Osman, F. (2014). Mitogen-activated Protein Kinase (MAPK) hyperactivation and enhanced NRAS expression drive acquired vemurafenib resistance in V600E BRAF melanoma cells. Journal of Biological Chemistry, 289(40), 27714–27726. https://doi.org/10.1074/jbc.M113.532432

Lin, L., Gopal, S., Sharda, A., Passam, F., Bowley, S. R., Stopa, J., Xue, G., Yuan, C., Furie, B. C., Flaumenhaft, R., Huang, M., & Furie, B. (2015). Quercetin-3-rutinoside Inhibits Protein Disulfide Isomerase by Binding to Its b′x Domain. Journal of Biological Chemistry, 290(39), 23543–23552. https://doi.org/10.1074/jbc.M115.666180

Liu, Y., & Sheikh, M. S. (2015). Melanoma: Molecular Pathogenesis and Therapeutic Management. Molecular and Cellular Pharmacology, 6(3), 228. http://www.ncbi.nlm.nih.gov/pubmed/25745537

Liu-Smith, F., Dellinger, R., & Meyskens, F. L. (2014). Updates of reactive oxygen species in melanoma etiology and progression. Archives of Biochemistry and Biophysics, 563, 51–55. https://doi.org/10.1016/j.abb.2014.04.007

Lodish, H., Berk, A., Kaiser, C. A., Krieger, M., Bretscher, A., Ploegh, H., Amon, A., & Scott, M. P. (2014). Biologia Celular e molecular (S. de Fraga, Ed.; 7th ed.). Artmed.

Long, G. v, Atkinson, V., Cebon, J. S., Jameson, M. B., Fitzharris, B. M., McNeil, C. M., Hill, A. G., Ribas, A., Atkins, M. B., Thompson, J. A., Hwu, W.-J., Hodi, F. S., Menzies, A. M., Guminski, A. D., Kefford, R., Kong, B. Y., Tamjid, B., Srivastava, A., Lomax, A. J., … Carlino, M. S. (2017). Standard-dose pembrolizumab in combination with reduced-dose ipilimumab for patients with advanced melanoma (KEYNOTE-029): an open-label, phase 1b trial. The Lancet Oncology, 18(9), 1202–1210. https://doi.org/10.1016/S1470-2045(17)30428-X

Long, G. v., Stroyakovskiy, D., Gogas, H., Levchenko, E., de Braud, F., Larkin, J., Garbe, C., Jouary, T., Hauschild, A., Grob, J. J., Chiarion-Sileni, V., Lebbe, C., Mandalà, M., Millward, M., Arance, A., Bondarenko, I., Haanen, J. B. A. G., Hansson, J., Utikal, J., … Flaherty, K. (2015). Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. The Lancet, 386(9992), 444–451. https://doi.org/10.1016/S0140-6736(15)60898-4

Lovat, P. E., Corazzari, M., Armstrong, J. L., Martin, S., Pagliarini, V., Hill, D., Brown, A. M., Piacentini, M., Birch-Machin, M. A., & Redfern, C. P. F. (2008). Increasing Melanoma Cell Death Using Inhibitors of Protein Disulfide Isomerases to Abrogate Survival Responses to Endoplasmic Reticulum Stress. Cancer Research, 68(13), 5363–5369. https://doi.org/10.1158/0008-5472.CAN-08-0035

Luebker, S. A., & Koepsell, S. A. (2019). Diverse Mechanisms of BRAF Inhibitor Resistance in Melanoma Identified in Clinical and Preclinical Studies. Frontiers in Oncology, 9. https://doi.org/10.3389/fonc.2019.00268

Lugowska, I., Koseła-Paterczyk, H., Kozak, K., & Rutkowski, P. (2015). Trametinib: A MEK inhibitor for management of metastatic melanoma. In OncoTargets and Therapy (Vol. 8, pp. 2251–2259). Dove Medical Press Ltd. https://doi.org/10.2147/OTT.S72951

Massi, A., Bortolini, O., Ragno, D., Bernardi, T., Sacchetti, G., Tacchini, M., & de Risi, C. (2017). Research Progress in the Modification of Quercetin Leading to Anticancer Agents. Molecules, 22(8), 1270. https://doi.org/10.3390/molecules22081270

Menzies, A. M., Long, G. v., & Murali, R. (2012). Dabrafenib and its potential for the treatment of metastatic melanoma. In Drug Design, Development and Therapy (Vol. 6, pp. 391–405). https://doi.org/10.2147/DDDT.S38998

Meyle, K. D., & Guldberg, P. (2009). Genetic risk factors for melanoma. In Human Genetics (Vol. 126, Issue 4, pp. 499–510). https://doi.org/10.1007/s00439-009-0715-9

Miller, A. J., & Mihm, M. C. (2006). Melanoma. New England Journal of Medicine, 355(1), 51–65. https://doi.org/10.1056/NEJMra052166

National Center for Biotechnology Information. (2022a). PubChem Compound Summary for CID 11707110, Trametinib. National Center for Biotechnology Information. Retrieved August 20, 2022 from https://pubchem.ncbi.nlm.nih.gov/compound/trametinib

National Center for Biotechnology Information. (2022b). PubChem Compound Summary for CID 42611257, Vemurafenib. National Center for Biotechnology Information. Retrieved July 25, 2022 from https://pubchem.ncbi.nlm.nih.gov/compound/Vemurafenib

National Center for Biotechnology Information. (2022c). PubChem Compound Summary for CID 44462760, Dabrafenib. National Center for Biotechnology Information. Retrieved August 20, 2022 from https://pubchem.ncbi.nlm.nih.gov/compound/dabrafenib

Nazarian, R., Shi, H., Wang, Q., Kong, X., Koya, R. C., Lee, H., Chen, Z., Lee, M. K., Attar, N., Sazegar, H., Chodon, T., Nelson, S. F., McArthur, G., Sosman, J. A., Ribas, A., & Lo, R. S. (2010). Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature, 468(7326), 973–977. https://doi.org/10.1038/nature09626

Pescatore-Alves, L. (2012). Papel da dissulfeto isomerase proteica (PDI) na migração de células musculares lisas vasculares: possível envolvimento de Nox1 NADPH oxidase e RhoGTPases [Tese de Doutorado, Universidade de São Paulo]. https://doi.org/10.11606/T.5.2012.tde-25042012-165242

Queirolo, P., Boutros, A., Tanda, E., Spagnolo, F., & Quaglino, P. (2019). Immune-checkpoint inhibitors for the treatment of metastatic melanoma: a model of cancer immunotherapy. Seminars in Cancer Biology, 59, 290–297. https://doi.org/10.1016/j.semcancer.2019.08.001

Ribas, A., Lawrence, D., Atkinson, V., Agarwal, S., Miller, W. H., Carlino, M. S., Fisher, R., Long, G. v., Hodi, F. S., Tsoi, J., Grasso, C. S., Mookerjee, B., Zhao, Q., Ghori, R., Moreno, B. H., Ibrahim, N., & Hamid, O. (2019). Combined BRAF and MEK inhibition with PD-1 blockade immunotherapy in BRAF-mutant melanoma. Nature Medicine, 25(6), 936–940. https://doi.org/10.1038/s41591-019-0476-5

Sale, M. J., & Cook, S. J. (2013). That which does not kill me makes me stronger; Combining ERK1/2 pathway inhibitors and BH3 mimetics to kill tumour cells and prevent acquired resistance. In British Journal of Pharmacology (Vol. 169, Issue 8, pp. 1708–1722). https://doi.org/10.1111/bph.12220

Shi, H., Kong, X., Ribas, A., & Lo, R. S. (2011). Combinatorial Treatments That Overcome PDGFRβ-Driven Resistance of Melanoma Cells to V600EB-RAF Inhibition. Cancer Research, 71(15), 5067–5074. https://doi.org/10.1158/0008-5472.CAN-11-0140

Sirokmány, G., Donkó, Á., & Geiszt, M. (2016). Nox/Duox Family of NADPH Oxidases: Lessons from Knockout Mouse Models. Trends in Pharmacological Sciences, 37(4), 318–327. https://doi.org/10.1016/j.tips.2016.01.006

Soares Moretti, A. I., & Martins Laurindo, F. R. (2017). Protein disulfide isomerases: Redox connections in and out of the endoplasmic reticulum. Archives of Biochemistry and Biophysics, 617, 106–119. https://doi.org/10.1016/j.abb.2016.11.007

Sobierajska, K., Skurzynski, S., Stasiak, M., Kryczka, J., Cierniewski, C. S., & Swiatkowska, M. (2014). Protein Disulfide Isomerase Directly Interacts with β-Actin Cys374 and Regulates Cytoskeleton Reorganization. Journal of Biological Chemistry, 289(9), 5758–5773. https://doi.org/10.1074/jbc.M113.479477

Sun, C., Wang, L., Huang, S., Heynen, G. J. J. E., Prahallad, A., Robert, C., Haanen, J., Blank, C., Wesseling, J., Willems, S. M., Zecchin, D., Hobor, S., Bajpe, P. K., Lieftink, C., Mateus, C., Vagner, S., Grernrum, W., Hofland, I., Schlicker, A., … Bernards, R. (2014). Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature, 508(1), 118–122. https://doi.org/10.1038/nature13121

Thota, R., Johnson, D. B., & Sosman, J. A. (2015). Trametinib in the treatment of melanoma. Expert Opinion on Biological Therapy, 15(5), 735–747. https://doi.org/10.1517/14712598.2015.1026323

Wan, P. T. C., Garnett, M. J., Roe, S. M., Lee, S., Niculescu-Duvaz, D., Good, V. M., Project, C. G., Jones, C. M., Marshall, C. J., Springer, C. J., Barford, D., & Marais, R. (2004). Mechanism of Activation of the RAF-ERK Signaling Pathway by Oncogenic Mutations of B-RAF. Cell, 116(6), 855–867. https://doi.org/10.1016/S0092-8674(04)00215-6

Winterbourn, C. C. (2008). Reconciling the chemistry and biology of reactive oxygen species. Nature Chemical Biology, 4(5), 278–286. https://doi.org/10.1038/nchembio.85

Xu, S., Sankar, S., & Neamati, N. (2014). Protein disulfide isomerase: A promising target for cancer therapy. In Drug Discovery Today (Vol. 19, Issue 3, pp. 222–240). Elsevier Ltd. https://doi.org/10.1016/j.drudis.2013.10.017

Published

28/10/2022

How to Cite

DAL’AVA, B. T. .; SOUZA, N. F. de; CHAGAS, T. dos S. .; PEREIRA , G. J. V. . Comparison between antineoplastic treatments with selective inhibitors (BRAF/MEK) and the new potentials combinatorial therapies for metastatic melanoma. Research, Society and Development, [S. l.], v. 11, n. 14, p. e326111436275, 2022. DOI: 10.33448/rsd-v11i14.36275. Disponível em: https://www.rsdjournal.org/index.php/rsd/article/view/36275. Acesso em: 26 apr. 2024.

Issue

Section

Review Article