Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1619 2022-10-24 16:17:12 |
2 format correct Meta information modification 1619 2022-10-25 02:54:28 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sabag, N.;  Yakobson, A.;  Retchkiman, M.;  Silberstein, E. Biomarkers and Therapeutic Targets for Melanoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/30974 (accessed on 29 March 2024).
Sabag N,  Yakobson A,  Retchkiman M,  Silberstein E. Biomarkers and Therapeutic Targets for Melanoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/30974. Accessed March 29, 2024.
Sabag, Noa, Alexander Yakobson, Meir Retchkiman, Eldad Silberstein. "Biomarkers and Therapeutic Targets for Melanoma" Encyclopedia, https://encyclopedia.pub/entry/30974 (accessed March 29, 2024).
Sabag, N.,  Yakobson, A.,  Retchkiman, M., & Silberstein, E. (2022, October 24). Biomarkers and Therapeutic Targets for Melanoma. In Encyclopedia. https://encyclopedia.pub/entry/30974
Sabag, Noa, et al. "Biomarkers and Therapeutic Targets for Melanoma." Encyclopedia. Web. 24 October, 2022.
Biomarkers and Therapeutic Targets for Melanoma
Edit

Malignant melanoma is one of the most common cancers in the world. In the disease’s early stages, treatment involves surgery, in advanced stages however, treatment options were once scarce. There has been a paradigm shift in advanced melanoma treatment with the introduction of immunotherapy and targeted therapies. Understanding the molecular pathways and their pathologic counterparts helped identifying specific biomarkers that lead to the development of specific targeted therapies.

malignant melanoma immunotherapy adjuvant therapy targeted therapy

1. Introduction

Malignant melanoma is one of the most common cancers in the world, ranking fifth for the highest number of estimated new cases in 2022 according to Cancer Statistics Cases [1]. In the disease’s early stages, treatment involves surgery; in advanced stages, however, treatment options were once scarce. There has been a paradigm shift in advanced melanoma treatment with the introduction of immunotherapy and targeted therapies. This began in 2011 when the U.S. Food and Drug Administration (FDA) approved the first drug, Ipilimumab, that proved to decrease mortality rates [2]. Since then, the FDA has approved eight more drugs for the treatment of melanoma that have resulted in improved overall survival rates [3].
Many patients, however, develop either primary or even secondary resistance to current systemic treatment options. Resistance is due to multiple complex mechanisms driven by a set of rewiring processes that involve cancer metabolism, epigenetics, gene expression and interactions in the tumour microenvironment.
There are several molecular pathways that, when unregulated, may become pathological and influence the onset, proliferation and invasion of melanoma.
The five main pathways are:
  • The Mitogen-activated Protein Kinase (MAPK) pathway, where serine/threonine-protein kinase BRAF (BRAF) is involved [4];
  • The phosphatidylinositol-3-kinases (PI3K)-AKT pathway [5];
  • The gene suppressor Cyclin-Dependent Kinase Inhibitor 2A (CDKN2A) and tumour suppressors pathway [6];
  • The microphthalmia-associated transcription factor (MITF) pathway [7];
  • The nuclear factor-kappa B (NFκB) pathway [8].
Understanding these molecular pathways and their pathologic counterparts helps identify specific biomarkers that lead to the development of specific targeted therapies.

2. Novel Therapeutic Strategies in the Field of Melanoma

2.1. Gender-Adapted Therapy

Female patents have a survival advantage in the case of melanoma, regardless of other tumour characteristics such as Breslow thickness or ulceration [9]. This leads to the assumption that the genomic differences between males and females may be potentially exploited as a therapeutic target. Differences in melanoma mutation burden associated with gender have already been found [10]. Further gender differences could be related to innate factors such as hormone levels, hormone receptor expression, immune system function and cell apoptosis susceptibility [11]. The connection between immune response and mutation burden explains the superiority of females over males regarding survival outcomes. The more that is revealed about the exact gender-related differences in melanoma, the more treatment may be accurately adapted.

2.2. Molecular Targeting Strategies

Six hundred and thirty-five biomarkers are associated with malignant melanoma [12]. There are numerous clinical trials aimed at discovering treatments for specific melanoma mutations. Two novel treatments showing promising results with respect to efficacy are:
Targeted therapy for c-KIT-mutated melanoma: The c-KIT gene mutation has been observed in gastrointestinal stromal tumours. Imatinib blocks the receptor tyrosine kinase activity of c-KIT and is a first-line therapy for gastrointestinal stromal tumours (GIST) [13]. Inhibitors of c-KIT, including Nilotinib, Dasatinib and Imatinib, have all been assessed in clinical trials and have delivered disappointing results with minimal activity in c-KIT-mutant melanoma patients. Further studies are required, as the long-term efficacy of c-KIT inhibitors in melanoma patients is uncertain, as well as its efficacy compared to other immunotherapies [14][15][16]. Immunotherapy in combination with c-KIT inhibitors provides hope for future therapeutic responses in mutant melanoma.
MEK inhibitors for NF1-mutation melanoma: Although NF1-mutant melanoma patients do not usually express NRAS or BRAF mutations, inhibition of the BRAF, MEK and mTOR pathways may be therapeutic since the NF1 mutation increases RAS/MAPK pathway signalling [17]. The greatest evidence of this achieved so far is related to treatment with MEK inhibitors for NF1-mutant melanoma [18][19]. Although therapeutic response to MEK inhibitors is impressive, a long period of treatment is necessary to achieve this response [20]. Immunotherapy and targeted therapy are both found to be effective for the treatment of NF1-mutant melanoma; however, further studies are required to determine the best treatment protocol with minimal side effects.
The field of targeted therapy is continuously evolving, with existing agents being investigated for new combinations and indications. Immunotherapy is also making continuous progress, and new standards of treatment are determined frequently.
Immunotherapy as a neoadjuvant therapy: Currently there are no standardized neo-adjuvant treatment regimens; however, there are several trials showing good pathological response rates, ranging from 50–80%, including OpACIN-neo studies and the PRADO trial, which are both phase II studies. These trials have confirmed the safety and high pathologic response rate (approximately 60%) of neoadjuvant Ipilimumab and Nivolumab. Patients with resectable BRAF-mutant stage III melanoma who received concurrent treatment with Dabrafenib+Trametinib+Pembrolizumab triple combination therapy in a new phase II trial had a high pathological response rate (80%) and a pathological complete response of 50% [21].
Adjuvant therapy: Anti-PD-1 monotherapy is an adjuvant treatment option for stage III melanoma and stage IV resectable patients. Standardized treatment options include adjuvant nivolumab treatment, which has shown significantly longer relapse-free survival (RFS) than adjuvant Ipilimumab in patients with resected stage IIIB, IIIC and IV resectable melanoma. In the KEYNOTE-054 study [22] assessing patients with stage IIIA, B or C melanoma, adjuvant Pembrolizumab had an improved RFS compared to the placebo. The COMBI-AD [23] trial of adjuvant BRAF/MEK-inhibitor therapy and dabrafenib and trametinib also exhibited improved RFS and overall survival (OS) compared to the placebo. In a new trial, KEYNOTE-716 [24], patients with stage IIB or IIC melanoma received pembrolizumab and, compared to the placebo, treated patients showed reduced risk of recurrence and improved distant metastatic-free survival. Although not yet a standardized treatment, this regimen has recently received FDA approval.
Immunotherapy is discussed as a neoadjuvant and adjuvant therapy in the current standards of treatment in melanoma, while surgery is still an essential part of the treatment when possible. The new immunotherapy drugs and the development of targeted therapy, with a face towards personalised therapy, bring up the question of the place of surgical treatment in the rapidly changing medical field. Surgical treatment is the gold standard for resectable cases and takes an important part in staging determination, but despite its values, it may lead to complications and morbidity. Therefore, a question about the potential of advanced medical treatments to replace surgical treatment has to be asked, at least for cases of mutilating resection surgeries.
Triple therapy for BRAF-mutant patients: the standard of care for BRAF-mutant melanoma patients is either BRAF/MEK inhibitor therapy or immunotherapy. There are three studies that have assessed anti-PD1+BRAF inhibitors + MEK inhibitors. Firstly, IMspire 150 [25] was the only study with significant results; it showed that Atezolizumab + Cobimetinib improved PFS compared to targeted therapy + placebo. This regimen is not commonly used because usually one needs to begin with BRAF-targeted therapy (Vemurafenib + Cobimetinib) before administering Atezolizumab, which may be complicated. In addition, Atezolizumab is often considered to be inferior compared to anti-PD-1 therapy, even though there is no directly comparative data to support this. The second trial, KEYNOTE-022 [26], showed no significant difference in PFS between triple therapy and targeted therapy. The third trial, COMBI-I [27], also showed no significant support for triple therapy using Sparta-DabTram therapy. There is currently an ongoing phase 3 STARBOARD trial [28] assessing the triple combination of Encorafenib + Binimetinib + Pembrolizumab versus Pembrolizumab alone. This is highly relevant because, as opposed to previous studies, STARBOARD is using front-line anti-PD-1 immunotherapy for comparison, as opposed to BRAF/MEK inhibitors, which currently are rarely used as first-line therapy for melanoma treatment. In general, triple therapy is only considered the best treatment approach in patients with high disease burden that is rapidly progressing.
New immunotherapy combinations: A new study, RELATIVITY-047, suggested using combined immunotherapy for unresectable or metastatic melanoma with anti-PD-1 and anti-lymphocyte-activation gene 3 (LAG-3) as active agents [29]. While anti-PD-1 is a known and proven agent, anti-LAG-3 is a relatively new immune checkpoint inhibitor, and its role as an inhibitory target seems promising [30]. RELATIVITY-047 [29] compared Relatlimab (a new checkpoint inhibitor) + nivolumab to nivolumab alone and found a significantly improved PFS in favour of the combination therapy. This trial showed that inhibition of these two immune checkpoints provided a significant benefit compared to anti-PD-1 alone with regards to progression-free survival. Currently there are ongoing studies researching the efficacy of this combination therapy as an adjuvant therapy [31]. There are currently pre-clinical studies supporting the use of T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain (TIGIT), an inhibitory receptor known as Vibostolimab, in metastatic melanoma [32]. A phase III clinical trial is expected to begin relatively soon to compare TIGIT in combination with anti-PD-1 therapy vs. anti-PD-1 therapy alone as an adjuvant therapy for stage IIB, C, III and IV resectable cutaneous melanoma. In addition, there are current, ongoing trials assessing the addition of the BCL-2 inhibitor navitoclax to the combination of dabrafenib + trametinib [33]. There is also a phase 2 BAMM2 trial examining the addition of hydroxychloroquine to dabrafenib + trametinib in patients with stage IIIC and IV BRAF-positive melanoma [34]. Lastly, the use of ipilimumab/nivolumab + the interleukin 6 (IL-6) receptor blocker tocilizumab in patients with untreated, unresectable advanced or metastatic melanoma has been reported. Results are promising, and the study is ongoing [35]. In a very recent study, the phase I AMBER study presented at the 2022 ASCO Annual Meeting, dual T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) with anti-PD1 showed promising anti-tumour activity. This study utilized Cobolimab, a selective anti-TIM-3 monoclonal antibody [36].
An additional study assessed a TOLL-like receptor (TLR)-9 agonist: vidutolimod. This drug has shown promise in patients as a monotherapy as well as in combination with anti-PD-1 therapy, with a response rate of approximately 20% in both regimens; however, it is more durable when combined with anti-PD-1 therapy [37].

References

  1. The cancer statistics center of the American Cancer Society. Available online: https://cancerstatisticscenter.cancer.org (accessed on 12 January 2022).
  2. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. NEJM 2010, 363, 711–723.
  3. Sharif, K.; Coplan, L.; Lichtbroun, B.; Amital, H. Unmasking a new potential role of intravenous immunoglobulins: Could it stand the cancer challenge? IMAJ 2018, 20, 780–781.
  4. Raman, M.; Chen, W.; Cobb, M.H. Differential regulation and properties of MAPKs. Oncogene 2007, 26, 3100–3112.
  5. Kong, Y.; Si, L.; Li, Y.; Wu, X.; Xu, X.; Dai, J.; Tang, H.; Ma, M.; Chi, Z.; Sheng, X.; 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.
  6. Fargnoli, M.C.; Gandini, S.; Peris, K.; Maisonneuve, P.; Raimondi, S. MC1R variants increase melanoma risk in families with CDKN2A mutations: A meta-analysis. EJC 2010, 46, 1413–1420.
  7. Garraway, L.A.; Widlund, H.R.; Rubin, M.A.; Getz, G.; Berger, A.J.; Ramaswamy, S.; Beroukhim, R.; Milner, D.A.; Granter, S.R.; Du, J.; et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 2005, 436, 117–122.
  8. Gajos-Michniewicz, A.; Czyz, M. WNT Signaling in Melanoma. IJMS 2020, 21, 4852.
  9. Joosse, A.; Collette, S.; Suciu, S.; Nijsten, T.; Lejeune, F.; Kleeberg, U.R.; Coebergh, J.W.W.; Eggermont, A.M.M.; de Vries, E. Superior Outcome of Women with Stage I/II Cutaneous Melanoma: Pooled Analysis of Four European Organisation for Research and Treatment of Cancer Phase III Trials. JCO 2012, 30, 2240–2247.
  10. Gupta, S.; Artomov, M.; Goggins, W.; Daly, M.; Tsao, H. Gender Disparity and Mutation Burden in Metastatic Melanoma. JNCI J. 2015, 107, djv221.
  11. Joosse, A.; Collette, S.; Suciu, S.; Nijsten, T.; Patel, P.M.; Keilholz, U.; Eggermont, A.M.M.; Coebergh, J.W.W.; de Vries, E. Sex Is an Independent Prognostic Indicator for Survival and Relapse/Progression-Free Survival in Metastasized Stage III to IV Melanoma: A Pooled Analysis of Five European Organisation for Research and Treatment of Cancer Randomized Controlled Trials. JCO 2013, 31, 2337–2346.
  12. Melanoma Biomarkers by “My Cancer Genom” from Genetically Informed Cancer Medicine. Available online: https://www.mycancergenome.org/content/biomarkers/#search=Melanoma (accessed on 12 January 2022).
  13. Kelly, C.M.; Gutierrez Sainz, L.; Chi, P. The management of metastatic GIST: Current standard and investigational therapeutics. J Hematol. Oncol. 2021, 14, 2.
  14. Guo, J.; Si, L.; Kong, Y.; Flaherty, K.T.; Xu, X.; Zhu, Y.; Corless, C.L.; Li, L.; Li, H.; Sheng, X. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic mela- noma harboring c-Kit mutation or amplification. J. Clin. Oncol. 2011, 29, 2904.
  15. Steeb, T.; Wessely, A.; Petzold, A.; Kohl, C.; Erdmann, M.; Berking, C.; Heppt, M.V. c-Kit inhibitors for unresectable or metastatic mucosal, acral or chronically sun-damaged melanoma: A systematic review and one-arm meta-analysis. EJC 2021, 157, 348–357.
  16. Py, C.; Christinat, Y.; Kreutzfeldt, M.; McKee, T.A.; Dietrich, P.-Y.; Tsantoulis, P. Response of NF1-Mutated Melanoma to an MEK Inhibitor. JCO Precis. Oncol. 2018, 1–11, 1380–1387.
  17. Kiuru, M.; Busam, K.J. The NF1 gene in tumor syndromes and melanoma. Lab. Invest. 2017, 97, 146–157.
  18. Guo, J.; Carvajal, R.D.; Dummer, R.; Hauschild, A.; Daud, A.; Bastian, B.C.; Markovic, S.N.; Queirolo, P.; Arance, A.; Berking, C.; et al. Efficacy and safety of nilotinib in patients with KIT-mutated metastatic or inoperable melanoma: Final results from the global, single-arm, phase II TEAM trial. Ann. Oncol. 2017, 28, 1380–1387.
  19. Nissan, M.H.; Pratilas, C.; Jones, A.M.; Ramirez, R.; Won, H.; Liu, C.; Tiwari, S.; Kong, L.; Hanrahan, A.J.; Yao, Z.; et al. Loss of NF1 in cutaneous melanoma is associated with RAS activation and MEK dependence. Cancer. Res. 2014, 74, 2340–2350.
  20. Solares, I.; Viñal, D.; Morales-Conejo, M.; Rodriguez-Salas, N.; Feliu, J. Novel molecular targeted therapies for patients with neurofibromatosis type 1 with inoperable plexiform neurofibromas: A comprehensive review. ESMO Open 2021, 6, 100223.
  21. Rozeman, E.A.; Menzies, A.M.; van Akkooi, A.C.; Adhikari, C.; Bierman, C.; van de Wiel, B.A.; Scolyer, R.A.; Krijgsman, O.; Sikorska, K.; Eriksson, H.; et al. Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): A multicentre, phase 2, randomised, controlled trial. Lancet Oncol. 2019, 20, 948–960.
  22. Eggermont, A.M.M.; Blank, C.U.; Mandala, M.; Long, G.V.; Atkinson, V.; Dalle, S.; Haydon, A.; Lichinitser, M.; Khattak, A.; Carlino, M.S.; et al. Adjuvant pembrolizumab versus placebo in respected stage 3 melanoma. NEJM 2018, 378, 1789–1801.
  23. Long, G.V.; Hauschild, A.; Santinami, M.; Atkinson, V.; Mandalà, M.; Chiarion-Sileni, V.; Larkin, J.; Nyakas, M.; Dutriaux, C.; Haydon, A.; et al. Adjuvant Dabrafenib plus Trametinib in Stage IIIBRAF-Mutated Melanoma. NEJM 2017, 377, 1813–1823.
  24. Luke, J.J.; Rutkowski, P.; Queirolo, P.; Del Vecchio, M.; Mackiewicz, J.; Chiarion-Sileni, V.; de la Cruz Merino, L.; Khattak, M.A.; Schadendorf, D.; Long, G.V.; et al. Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): A randomised, double-blind, phase 3 trial. Lancet 2022, 399, 1718–1729.
  25. Gutzmer, R.; Stroyakovskiy, D.; Gogas, H.; Robert, C.; Lewis, K.; Protsenko, S.; Pereira, R.P.; Eigentler, T.; Rutkowski, P.; Demidov, L.; et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): Primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020, 395, 1835–1844.
  26. Ferrucci, P.F.; Di Giacomo, A.M.; Del Vecchio, M.; Atkinson, V.; Schmidt, H.; Schachter, J.; Queirolo, P.; Long, G.V.; Stephens, R.; Svane, I.M.; et al. KEYNOTE-022 part 3: A randomized, double-blind, phase 2 study of pembrolizumab, dabrafenib, and trametinib in BRAF-mutant melanoma. J. Immunother. Cancer 2020, 8, e001806.
  27. Dummer, R.; Long, G.V.; Robert, C.; Tawbi, H.A.; Flaherty, K.T.; Ascierto, P.A.; Nathan, P.D.; Rutkowski, P.; Leonov, O.; Dutriaux, C.; et al. Randomized Phase III Trial Evaluating Spartalizumab Plus Dabrafenib and Trametinib for BRAF V600–Mutant Unresectable or Metastatic Melanoma. JCO 2022, 40, 1428–1438.
  28. Schadendorf, D.; Dummer, R.; Robert, C.; Ribas, A.; Sullivan, R.J.; Panella, T.; McKean, M.; Santos, E.S.; Brill, K.; Polli, A.; et al. STARBOARD: Encorafenib + binimetinib + pembrolizumab for first-line metastatic/unresectable BRAF V600-mutant melanoma. Future Oncol. 2022, 18, 2041–2051.
  29. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Gutiérrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. NEJM 2022, 386, 24–34.
  30. Long, L.; Zhang, X.; Chen, F.; Pan, Q.; Phiphatwatchara, P.; Zeng, Y.; Chen, H. The promising immune checkpoint LAG-3: From tumor microenvironment to cancer immunotherapy. Genes Cancer 2018, 9, 176–189.
  31. Available online: https://clinicaltrials.gov/ct2/show/NCT05002569 (accessed on 21 August 2021).
  32. Ge, Z.; Peppelenbosch, M.P.; Sprengers, D.; Kwekkeboom, J. TIGIT, the Next Step Towards Successful Combination Immune Checkpoint Therapy in Cancer. Front. Immunol. 2021, 12, 699895.
  33. Available online: https://clinicaltrials.gov/ct2/show/NCT01989585 (accessed on 19 August 2022).
  34. Available online: https://www.clinicaltrials.gov/ct2/show/NCT04527549 (accessed on 14 January 2022).
  35. Sullivan, R.J. The role of triple therapy in BRAF-positive melanoma. Clin. Adv. Hematol. Oncol. HO 2022, 20, 359–361.
  36. Ribas, A.; Eroglu, Z.; Trigo Perez, J.M.M.; Di Pace, B.; Wang, T.; Ghosh, S.; Dhar, A.; Borgovan, T.; Waszak, A.; Davar, D. AMBER parts 1c and 1e: A phase 1 study of cobolimab plus dostarlimab in patients (pts) with advanced/metastatic melanoma. JCO 2022, 40, 9513.
  37. Geoffrey, T.; Gibney, M.D. Discusses Current Treatment Options for Patients with Melanoma, Clinical Trials That Have Explored Different Agents and Combinations, and Developing Treatment Options in Uveal Melanoma. Available online: https://www.onclive.com/view/gibney-breaks-down-the-evolving-treatment-landscape-of-melanoma (accessed on 15 June 2022).
More
Information
Subjects: Surgery
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 286
Revisions: 2 times (View History)
Update Date: 25 Oct 2022
1000/1000