Targeted therapeutic strategies for melanoma : Chinese Medical Journal

Secondary Logo

Journal Logo

Review Article

Targeted therapeutic strategies for melanoma

Zhang, Shiwei; Xie, Ruxin; Zhong, Ai; Chen, Junjie

Editor(s): Gao, Ting; Hao, Xiuyuan

Author Information
Chinese Medical Journal ():10.1097/CM9.0000000000002692, May 05, 2023. | DOI: 10.1097/CM9.0000000000002692
  • Open
  • PAP

Abstract

Introduction

In the past ten years, the incidence of melanoma continued to rise, accounting for 1.7% of diagnosed cancers worldwide. It is the fifth most common cancer in the United States and the annual incidence of melanoma in China has risen to about 16.83 per 10,000, while the annual death toll is about 4369. The relatively poor prognosis of melanoma also brings a great disease burden to society.[1–5] Over the past 15 years, with the improvement in our understanding of the pathophysiology of melanoma, it has been recognized that melanoma development is closely related to genetic mutations, and the emergence of targeted drugs has also changed the comprehensive treatment of melanoma. Vemurafenib was the first BRAF inhibitor that was approved by the Food and Drug Administration (FDA, in 2011) for BRAF-mutant melanoma, and since that time, the prognosis of patients with advanced melanoma has undergone continuous and significant improvement.[6] This article reviews and provides evidence on the advances in targeted therapy of melanoma.

Melanoma Subtypes and Common Gene Mutations

Based on the 2018 World Health Organization Classification of cutaneous, mucosal, and uveal melanoma, which encompasses mutational signatures, anatomical site, and epidemiology, melanoma is divided into etiologies that are related and unrelated to sun exposure. The histological degree of cumulative solar damage (CSD) of the surrounding skin, based on the level of associated solar elastosis, further categorizes melanomas on sun-exposed skin into low and high CSD. In the low-CSD group, melanomas include superficial spreading melanoma, while those in the high-CSD group include lentigo malignant and sclerosing melanomas. The "non-CSD" classification includes acral melanoma, melanoma in congenital nevus, melanoma in blue nevus, Spitz melanoma, mucosal melanoma, and uveal melanoma. Meanwhile, nodular and nevoid melanomas can occur by both routes.[7,8]

The major melanoma subtypes are associated with different mutational profiles, with frequently mutated genes, including BRAF, CDKN2A, NRAS, and TP53 in cutaneous melanoma; BRAF, NRAS, NF1, and KIT in acral melanoma (although the rate of mutation is less frequent compared with cutaneous melanoma); and SF3B1 in mucosal melanoma.[9] In all melanomas, the mutation rate of BRAF V600 is about 40–50%, NRAS is about 15–20%, and the mutation rate of KIT is only 1–2% (but the KIT mutation rate is relatively high in acral melanoma and mucosal melanoma, reaching nearly 15–20%).[10,11] Based on this, these mutations are often used as therapeutic targets.

BRAF-Mutant Melanoma

BRAF mutation is the most common mutation in melanoma and accounts for nearly 50% of all mutations.[12] The role of the MAPK (mitogen activated kinase-like protein) pathway in the proliferation of melanoma involves the activation of RAF that leads to phosphorylation of MEK (MAPK kinase) and the phosphorylation of ERK, which stimulates cell proliferation and activates mitochondrial proteins, thus promoting growth and inhibiting apoptosis.[13] Based on this, BRAF is an effective target for the treatment of melanoma with BRAF mutation, and many targeted agents are being developed. Since the approval of the BRAF inhibitor, vemurafenib, by the FDA for the treatment of advanced melanoma with BRAF V600E/K mutation, in 2011, the comprehensive treatment of melanoma has been revolutionized. A total of five BRAF and MEK inhibitors were approved by the FDA in the following years [Figure 1]. The results of several trials showed that the combination of BRAF and MEK inhibitors was more effective than BRAF inhibitor monotherapy, due to its capacity to delay the emergence of BRAF inhibitor resistance and reduce skin toxicity. Currently, regimens of BRAF inhibitors combined with MEK inhibitors are approved by the FDA and recommended as first-line therapy for advanced melanoma. These regimens include dabrafenib plus trametinib, vemurafenib plus cobimetinib, and encorafenib plus binimetinib.[14]

F1
Figure 1:
The timeline of significant clinical trials of targeted therapy and the mechanism of targeted drugs. BRIM3, BREAK3, METRIC, coBRIM, COMBI-v, COMBI-d, COLUMBUS, and IMspire150 are phase III trials. BRIM3, BREAK3, and METRIC demonstrated that vemurafenib and dabrafenib were more effective than traditional chemotherapy drug dacarbazine and had lower adverse reaction rate. coBRIM, COMBI-v, COMBI-d, and COLUMBUS demonstrated that the combination of BRAF and MEK inhibitors improved OS in melanoma patients compared with BRAF inhibitor monotherapy; besides, the combination of encorafenib and binimetinib in melanoma increases OS over 30 months for the first time. The results of IMspire150 suggested that the combination of spartalizumab with vemurafenib and cobimetinib could improve PFS and objective response and induce long-term responses in patients. TRK receptors family is composed of three receptor tyrosine kinases (RTKs): TRKA, TRKB and TRKC, also called neurotrophic tyrosine receptor kinases (NTRK1-NTRK3) that function as high affinity signaling receptors for various neurotrophins, including NGF, BDGF and NT3. BDGF: Brain-derived neurotrophic factor; ERK: extracellular regulated protein kinases; MEK: mitogen activated kinase-like protein kinase; NFI: Neurofibromin 1. NGF: Nerve growth factor NT3: Neurotrophin-3; OS: Overall survival; PFS: Progression-free survival; PLCγ: phospholipase C gamma; PI3K: phosphatidylinositol 3-kinase, putative; TRK: tropomyosin receptor kinase.

BRAF inhibitors and MEK inhibitors for advanced melanoma

A clinical trial (NCT01072175) demonstrated the efficacy and safety of the combination of dabrafenib and trametinib in patients with BRAF V600 mutations.[15,16] Based on a longer-time follow-up, the overall survival (OS) (4 years, 30%; 5 years, 28%) and progression-free survival (PFS) (4 years and 5 years, both 13%) in the case of receiving dabrafenib plus trametinib appeared to be stabilized.[17] Although NCT01072175 demonstrated the safety and efficacy of the combination of dabrafenib and trametinib, it did not compare the effects of monotherapy and combination therapy, and thus, a series of randomized controlled trials (COMBI-v, COMBI-d, coBRIM, and COLUMBUS) were carried out. The combination of dabrafenib and trametinib improved OS in patients with BRAF mutations without increasing overall toxicity (COMBI-v and COMBI-d) compared with vemurafenib or dabrafenib monotherapy.[18–21] In COMBI-v and COMBI-d, the 5-year outcomes were associated with a PFS of 21% at 4 years (95% confidence interval [CI], 17–24%) and 19% at 5 years (95% CI, 15–22%). Overall survival was 37% (95% CI, 33–42%) at 4 years and 34% (95% CI, 30–38%) at 5 years.[22] Compared with vemurafenib monotherapy, vemurafenib plus cobimetinib, encorafenib plus binimetinib, or encorafenib monotherapy could improve patients' PFS and OS. Compared with encorafenib monotherapy, encorafenib plus binimetinib could improve patient tolerability. These trials (coBRIM, COLUMBUS) supported the use of vemurafenib plus cobimetinib and encorafenib plus binimetinib as first-line treatment options for patients with advanced BRAF-mutant melanoma.[23–25] In coBRIM, the median PFS with cobimetinib and vemurafenib was 12.3 (95% CI, 9.5–13.4) months.[26] According to the updated data of COLUMBUS, the combination group reduced the risk of death by 39% compared with that of vemurafenib alone (hazard ratio [HR], 0.61). Besides, the PFS was doubled to 14.9 months (HR, 0.51).[27]

A real-world study showed that brain metastases often represented a poor prognosis.[28] A study that combined dabrafenib and trametinib suggested its efficacy and safety with a relatively short median duration of response (DoR) for patients with brain metastases. However, whether combined therapy can improve the prognosis of patients with brain metastases requires more clinical evidence.[29]

BRAF inhibitors and MEK inhibitors for adjuvant therapy

Surgery is an indispensable part of the comprehensive treatment of melanoma, but for patients with stage IIC–III, there is still a risk of recurrence and death even if R0 resection is achieved. To explore whether the use of post-surgery targeted drugs improves outcomes in patients with stage IIC and III melanoma, COMBI-AD and BRIM8 had been conducted and data from these trials showed that the use of BRAF and MEK inhibitors could reduce the risk of recurrence. From the results of COMBI-AD, patients with stage III melanoma with a BRAF V600Eor V600K mutation, who received 12 months of adjuvant dabrafenib plus trametinib after surgery, lived longer without recurrence or distant metastasis, while there was no significant long-term toxic effect compared with patients on placebo. This clinical trial suggested that dabrafenib plus trametinib adjuvant therapy might be beneficial in preventing or delaying recurrence in patients with a completely resected stage III melanoma. At 5 years, the percentage of living patients without relapse was 52% (95% CI, 48–58%) in the dabrafenib plus trametinib group and 36% (95% CI, 32–41%) in the placebo group who accepted two matched placebo (HR for recurrence or death, 0.51; 95% CI, 0.42–0.61). The percentage of living patients without distant metastases was 65% (95% CI, 61–71%) in the dabrafenib plus trametinib group and 54% (95% CI, 49–60%) in the placebo group (distant HR for metastasis or death, 0.55; 95% CI, 0.44–0.70).[30–32] In BRIM-8, the median disease-free survival was 23.1 (95% CI, 18.6–26.5) months in the vemurafenib group, compared with 15.4 (95% CI, 11.1–35.9) months in the placebo group (HR, 0.80, log-rank P = 0.26).[33]

BRAF inhibitors and MEK inhibitors for neoadjuvant therapy

Although upfront surgery and postoperative adjuvant therapy improve recurrence-free survival in stage III melanoma patients with BRAF mutation, there are still locally advanced stage III melanoma patients with BRAF mutation who are inoperable, and therefore, clinical trials of preoperative treatment with dabrafenib plus trametinib were conducted. These trials suggested that the neoadjuvant (dabrafenib plus trametinib) was beneficial for high-risk populations and could help patients with locally advanced melanoma who previously could not undergo radical tumor resection to achieve radical tumor resection after neoadjuvant therapy. Besides, these trials demonstrated the efficacy and safety of neoadjuvant therapy (dabrafenib plus trametinib). In a phase II trial of neoadjuvant plus adjuvant (NCT02231775), survival without disease progression was significantly higher in patients treated with the neoadjuvant plus adjuvants than that in patients treated with standard care (10 of 14 patients [71%] vs. none of 7 in the standard of care group; median event-free survival was 19.7 [95% CI, 16.2–not estimable] months and 2.9 [95% CI, 1.7–not estimable] months, respectively; HR, 0.016, 95% CI, 0.00012–0.14, P <0.0001). Neoadjuvant plus adjuvant was well-tolerated and without grade 4 adverse events or treatment-related deaths. The most common adverse reactions in the neoadjuvant plus adjuvant therapy group were grade 1–2 toxicities, including chills (12 of 14, 92%), headaches (12 of 14, 92%), and pyrexia (10 of 14, 77%). The most common grade 3 adverse event was diarrhea (2 of 14, 15%).[34]

In another phase II trial of neoadjuvant therapy (NeoCombi), 30 of 35 patients achieved a RECIST response, 16 patients (16 of 35, 46%; 95% CI, 29–63%) had complete responses, and 14 patients (14 of 35, 40%; 95% CI, 24–58%) had partial responses at resection. Five patients (5 of 35, 14%; 95% CI, 5–30%) had stable disease, and none had progressive disease. After resection and evaluation of the results of 18F-fluorodeoxyglucose (FDG) PET scan, all 35 patients achieved response, including 18 patients (18 of 35, 51%; 95% CI, 34–69%) with complete response and 17 patients (17 of 35, 49%; 95% CI, 31–66%) with incomplete response. Compared to traditional chemotherapy, the incidence of treatment-related adverse events (TRAEs) was relatively low, and these adverse reactions were acceptable to patients. Ten patients (10 of 35, 29%) experienced grade 3–4 adverse events, and none experienced treatment-related deaths.[35]

In a phase II study of neoadjuvant therapy, 21 patients, with unresectable BRAF mutated stage III melanoma or stage IV melanoma with ≤3 metastases, were enrolled for treatment. Surgery was performed in 18 patients (18 of 21, 86%), 17 with R0 resection and 1 with R1 resection. Patients who underwent surgery had a median recurrence-free survival of 9.9 (95% CI, 7.52–not reached) months. Through neoadjuvant therapy, 1 patient (1 of 21, 4.8%) achieved complete response, 16 (16 of 21, 76.2%) achieved partial response, and 1 (1 of 21, 4.8%) achieved stable disease, while 1 (1 of 21, 4.8%) had disease progression. Half of the patients who underwent surgery experienced recurrence (9 of 18) and most patients experienced distant recurrence (6 of 9), while 3 patients experienced local recurrence for the first time. Three patients with local recurrence underwent surgery, but all developed distant metastases.[36] Although neoadjuvant therapy can shrink and downgrade the tumor, the prognosis of patients with stage IIIC–IV melanoma is still poor, and more effective treatment options are still worth exploring.

Combination of BRAF plus MEK inhibitors with immune checkpoint inhibitors to achieve long-term response

Although treatment with BRAF plus MEK inhibitors can improve outcomes in patients with BRAF mutations, the major disadvantage of targeted therapy is that the tolerance to treatment increases with the shortening of the DoR with the patients usually relapsing within 9–12 months of treatment commencement. Patients are less responsive to immune checkpoint inhibitors than targeted therapy, but once immune memory is developed, there is a durable response to immunotherapy.[37] Based on this, several clinical trials involving the combination of targeted therapy with immunotherapy had been carried out, and the results confirmed that the combination of targeted therapy with immunotherapy could produce long-term responses in a subset of patients with BRAF mutations.

The results of IMspire150 showed that the addition of atezolizumab to patients with BRAF V600-mutant advanced melanoma, receiving vemurafenib and cobimetinib, improved PFS and was safely tolerated by the patients. PFS was significantly longer compared with that of the control group (15.1 months vs. 10.6 months; HR, 0.78; 95% CI, 0.63–0.97; P = 0.025).[38] In KEYNOTE-022 (part 3), the results suggested that pembrolizumab plus dabrafenib and trametinib improved PFS, DoR, and OS in patients with BRAF V600E/K-mutant advanced melanoma; however, the treatment-related adverse effects were higher. The median PFS was 16.9 (95% CI, 11.3–27.9) months in the pembrolizumab plus dabrafenib and trametinib group and 10.7 (95% CI, 7.2–16.8) months in the dabrafenib plus trametinib group (HR, 0.53; 95% CI, 0.34–0.83). Grade 3–5 TRAEs occurred in 35 patients (35 of 60, 58%, including one death) who received pembrolizumab plus dabrafenib and trametinib and 15 patients (15 of 60, 25%) who received dabrafenib plus trametinib.[39] The increase in the incidence of adverse reactions did not support the combination of pembrolizumab with dabrafenib and trametinib as a first-line treatment option. In COMBI-i, the results showed that the median PFS was 16.2 (95% CI, 12.7–23.9) months in the spartalizumab plus dabrafenib and trametinib group (experimental group), and 12.0 (95% CI, 10.2–15.4) months in the dabrafenib plus trametinib (control group) (HR, 0.82; P = 0.042). The objective response rates were 69% (183 of 267 patients) and 64% (170 of 265 patients), respectively. Grade 3 TRAEs occurred in 55% (146 of 267) in the experimental group and 33% (88 of 264) in the control group. It is worth noting that although the combination of spartalizumab plus dabrafenib and trametinib improved response rates, there was a corresponding increase in the incidence of adverse effects.[40] More medication regimens that can improve long-term response rates and lower TRAEs are worthy of further exploration.

KIT-Mutant Melanoma

Imatinib is a KIT inhibitor that shows effect on tumors with KIT mutation. It has a high response rate in the treatment of gastrointestinal stromal tumors and greatly improves the prognosis of patients.[41,42] Although KIT mutations account for only 1–2% of all mutations in melanoma, the KIT mutation rate is approximately 15–20% in acral and mucosal melanomas, accounting for 70% of all melanomas in Asian populations. Therefore, KIT is a reasonable therapeutic target that was tried for melanoma with KIT mutation[11,43]. Several clinical trials were conducted to explore the benefit of imatinib and confirmed that imatinib was effective for a subset of patients with KIT mutations (exon-11 or exon-13), but not for patients with KIT amplification only. In a phase II trial, 10 patients responded to imatinib, while 9 of them had exon-11 or exon-13 mutation. However, for patients who had developed resistance to imatinib, increasing the dose of imatinib to 800 mg/day in patients with disease progression did not restore disease control.[44] Meanwhile, a clinical trial showed that the emergence of imatinib resistance might be related to NRAS mutation and increase in KIT copies.[45]

Although imatinib clinical trials demonstrated the benefit of imatinib in patients with specific KIT-mutant advanced melanoma, the response rate was relatively low, and therefore, several clinical trials evaluated the efficacy of nilotinib in the treatment of advanced KIT-mutant melanoma. In a single-center phase II trial, seven patients who responded to nilotinib, and two patients who partially responded, had the KIT mutation. This result demonstrated the antitumoral effect of nilotinib in melanoma.[46] Another multicenter clinical trial further illustrated the effectiveness of nilotinib (UN10-06) by showing an overall response rate to nilotinib of 16.7% and a disease control rate of 57.1% (1 patient had complete response, 6 had a partial response, and 17 had a stable disease). Of the 7 responders, 6 patients had KIT mutations (exon-11: 5 patients; exon-17: 1 patient) and 1 patient just had a KIT amplification.[47] In a global phase II study of nilotinib, the results showed that nilotinib had a greater activity in patients with exon-11 mutation with an objective response rate of 26.2% and a median duration of overall response of 7.1 months.[48] Those trials confirmed the benefit of nilotinib in patients with KIT mutations (exon-11, exon-13, and exon-17), especially those with exon-11 mutation. The toxicity of nilotinib was also acceptable. In another phase II study, the results showed that nilotinib could achieve disease control in patients with KIT mutations and disease progression after imatinib treatment, but nilotinib had a limited efficacy in patients with brain metastases. Two partial responses were observed in patients without brain metastases (18.2%; 90% CI, 3–47%), whereas no response to nilotinib was observed in the treated patients with brain metastases. The median time to progression and OS for all treated patients were, respectively, 3.3 (90% CI, 2.1–3.9) months and 9.1 (90% CI, 4.3–14.2) months.[49]

Overall, nilotinib has a better clinical efficacy than imatinib, but only patients with specific KIT mutations can benefit from it. In a phase II study, the researchers observed decreases in the phosphorylation of the signal transducer and activator of transcription, STAT3, and its effectors (BCL-2 [BCL2 apoptosis regulator], MCL-1 [MCL1 apoptosis regulator, BCL2 family member]) in tumors during follow-up, that were significantly associated with clinical response. In the KIT-mutant melanoma cell line M230, nilotinib reduced STAT3 signaling, and STAT inhibitors were as effective as KIT inhibitors in reducing cell proliferation, demonstrating a significant association between STAT3 inhibition and response to nilotinib. These findings suggest that SATA3 may be a beneficial therapeutic target for patients with the KIT mutation and may increase the response of KIT inhibitors.[50]

At the 2021 European Society for Medical Oncology (ESMO) annual meeting, the results of a phase I study of ripretinib in KIT-mutant melanoma were presented. The data showed that in the treatment of KIT-mutant melanoma, the objective response rate (ORR) was similar to the previously reported data for other KIT inhibitors, and that the median PFS and DoR were superior. More clinical trials should be conducted to explore its efficacy and safety.[51]

NRAS-Mutant Melanoma

NRAS-mutant melanomas account for 15–20% of cutaneous melanomas.[52] So far, the first-line treatment for NRAS-mutant melanoma that is incurable by stage III/IV surgery has been immunotherapy with programed cell death protein (anti-PD-1) checkpoint inhibitors, such as nivolumab or pembrolizumab. The efficacy of immunotherapy in NRAS-mutant melanoma is controversial,[53] and a phase III trial (NEMO) suggested that binimetinib, a MEK inhibitor, might be a treatment option for patients with NRAS mutation. The median PFS in the binimetinib group was longer than that in the dacarbazine group. Median PFS was 2.8 months in the binimetinib group and 1.5 months in the dacarbazine group (HR, 0.62; 95% CI, 0.47–0.80; one-sided P <0.001).[54] Pimasertib has also demonstrated its safety and efficacy in clinical trials. In a phase II trial, the results suggested that PFS was significantly improved compared to DTIC (Dacarbazine) (median, 13 weeks and 7 weeks; HR, 0.59; 95% CI, 0.42–0.83; P = 0.002).[55] Besides, another trial of pimasertib demonstrated that pERK phosphorylated extracellular regulated protein kinases inhibition was associated with the clinical activity of pimasertib.[56] Statistically, MEK inhibitors prolong the survival period of NRAS mutant patients, but the prolongation time is relatively short, and thus, more effective targeted drugs are needed for NRAS-mutant melanoma.

NTRK-Mutant Melanoma

Fusions involving neurotrophic tyrosine receptor kinase (NTRK), a known driver of tumorigenesis, also occurs in melanoma. NTRK fusions are relatively common in spitzoid melanoma with a prevalence of 21–29%, whereas <1% are observed in cutaneous or mucosal melanoma and 2.5% in acral melanoma.[57] Therefore, NTRK is a reasonable therapeutic target for patients with advanced melanoma who have failed first-line therapy or who do not have BRAF and KIT mutations. From the results of two phase I studies of entrectinib (ALKA-372-001 and STARTRK-1), a potent oral inhibitor of the tyrosine kinases TRKA/B/C (tropomyosin receptor kinase A/B/C), ROS1, and ALK (ALK receptor tyrosine kinase), only one patient with acral melanoma responded to entrectinib. Through the evaluation of computed tomography, the patient achieved partial response without evidence of disease progression at 11 months.[58,59]

Conclusions

The emergency of targeted agents had revolutionized the treatment of advanced melanoma, especially for BRAF V600-mutant melanoma and the current major targeted agents and their indications were summarized in Table 1. The combination of encorafenib and binimetinib improved median survival to more than 30 months. Although the advance of targeted therapy has led to a decline in mortality of patients with advanced melanoma, survival rates for those diagnosed with stage IIIC or IV disease remain poor. Targeted therapy is extremely effective, but the outcome of long-term use of molecule-targeted inhibitors is the emergence of drug resistance. In melanoma, genetic mutations and cellular plasticity are mechanisms of resistance to targeted therapy.[60] To improve the prognosis of patients with advanced melanoma (the final goal is complete response), investigating the mechanism of drug resistance and identifying potential targets and treatment options (such as combination with immune checkpoint inhibitors) should be further explored.

Table 1 - Targeted therapy approved by FDA for melanoma.
Agent Mechanism Recommended indications
Vemurafenib BRAF inhibitor As monotherapy and in combination with cobimetinib for BRAF V600E/K-mutant disease
Dabrafenib BRAF inhibitor As monotherapy and in combination with trametinib for BRAF V600E/K-mutant disease
Encorafenib BRAF inhibitor As monotherapy and in combination with binimetinib for BRAF V600E/K-mutant disease
Trametinib MEK inhibitor As monotherapy and in combination with dabrafenib for BRAF V600E/K-mutant disease
Cobimetinin MEK inhibitor In combination with vemurafenib for BRAF V600E/K-mutant disease
Binimetinib MEK inhibitor In combination with encorafenib for BRAF V600E/K-mutant disease
Imatinib KIT inhibitor As monotherapy for c-KIT-mutant disease
Nilotinib KIT inhibitor As monotherapy for c-KIT-mutant disease
FDA: Food and drug administration.

Acknowledgments

We thank all the investigators who participated in preparing the present review.

Funding

This study was funded by 1·3·5 Project for Disciplines of Excellence, West China Hospital, Sichuan University (Nos. ZYPY20001 and ZYPY20002).

Conflicts of interest

None.

References

1. Leiter U, Keim U, Garbe C. Epidemiology of skin cancer: Update 2019. Adv Exp Med Biol 2020;1268: 123–139. doi: 10.1007/978-3-030-46227-7_6.
2. Rastrelli M, Tropea S, Rossi CR, Alaibac M. Melanoma: Epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo 2014;28: 1005–1011.
3. Saginala K, Barsouk A, Aluru JS, Rawla P, Barsouk A. Epidemiology of melanoma. Med Sci (Basel) 2021;9: 63. doi: 10.3390/medsci9040063.
4. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71: 209–249. doi: 10.3322/caac.21660.
5. Xia C, Dong X, Li H, Cao M, Sun D, He S, Yang F, Yan X, Zhang S, Li N, Chen W. Cancer statistics in China and United States, 2022: profiles, trends, and determinants. Chin Med J 2022;135: 584–590. doi: 10.1097/CM9.0000000000002108.
6. Jenkins RW, Fisher DE. Treatment of advanced melanoma in 2020 and beyond. J Invest Dermatol 2021;141: 23–31. doi: 10.1016/j.jid.2020.03.943.
7. Elder DE, Bastian BC, Cree IA, Massi D, Scolyer RA. The 2018 World Health Organization classification of cutaneous, mucosal, and uveal melanoma: Detailed analysis of 9 distinct subtypes defined by their evolutionary pathway. Arch Pathol Lab Med 2020;144: 500–522. doi: 10.5858/arpa.2019-0561-RA.
8. Bobos M. Histopathologic classification and prognostic factors of melanoma: A 2021 update. Ital J Dermatol Venerol 2021;156: 300–321. doi: 10.23736/S2784-8671.21.06958-3.
9. 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. doi: 10.1038/nature22071.
10. Davis EJ, Johnson DB, Sosman JA, Chandra S. Melanoma: What do all the mutations mean? Cancer 2018;124: 3490–3499. doi: 10.1002/cncr.31345.
11. Nassar KW, Tan AC. The mutational landscape of mucosal melanoma. Semin Cancer Biol 2020;61: 139–148. doi: 10.1016/j.semcancer.2019.09.013.
12. Leonardi GC, Falzone L, Salemi R, Zanghì A, Spandidos DA, Mccubrey JA, et al. Cutaneous melanoma: From pathogenesis to therapy (Review). Int J Oncol 2018;52: 1071–1080. doi: 10.3892/ijo.2018.4287.
13. Sun J, Carr MJ, Khushalani NI. Principles of targeted therapy for melanoma. Surg Clin North Am 2020;100: 175–188. doi: 10.1016/j.suc.2019.09.013.
14. Teixido C, Castillo P, Martinez-Vila C, Arance A, Alos L. Molecular markers and targets in melanoma. Cells 2021;10: 2320. doi: 10.3390/cells10092320.
15. Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med 2012;367: 1694–1703. doi: 10.1056/NEJMoa1210093.
16. Long GV, Weber JS, Infante JR, Kim KB, Daud A, Gonzalez R, et al. Overall survival and durable responses in patients with BRAF V600-mutant metastatic melanoma receiving dabrafenib combined with trametinib. J Clin Oncol 2016;34: 871–878. doi: 10.1200/JCO.2015.62.9345.
17. Long GV, Eroglu Z, Infante J, Patel S, Daud A, Johnson DB, et al. Long-term outcomes in patients with BRAF V600-mutant metastatic melanoma who received dabrafenib combined with trametinib. J Clin Oncol 2018;36: 667–673. doi: 10.1200/jco.2017.74.1025.
18. 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: 1877–1888. doi: 10.1056/NEJMoa1406037.
19. Grob JJ, Amonkar MM, Karaszewska B, Schachter J, Dummer R, Mackiewicz A, et al. Comparison of dabrafenib and trametinib combination therapy with vemurafenib monotherapy on health-related quality of life in patients with unresectable or metastatic cutaneous BRAF Val600-mutation-positive melanoma (COMBI-v): Results of a phase 3, open-label, randomised trial. Lancet Oncol 2015;16: 1389–1398. doi: 10.1016/s1470-2045(15)00087-x.
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. doi: 10.1016/s0140-6736(15)60898-4.
21. Robert C, Karaszewska B, Schachter J, Rutkowski P, Mackiewicz A, Stroiakovski D, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med 2015;372: 30–39. doi: 10.1056/NEJMoa1412690.
22. Robert C, Grob JJ, Stroyakovskiy D, Karaszewska B, Hauschild A, Levchenko E, et al. Five-year outcomes with dabrafenib plus trametinib in metastatic melanoma. N Engl J Med 2019;381: 626–636. doi: 10.1056/NEJMoa1904059.
23. Larkin J, Dréno B, Atkinson V, Liszkay G, Maio M, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med 2014;371: 1867–1876. doi: 10.1056/NEJMoa1408868.
24. Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, et al. Overall survival in patients with BRAF-mutant melanoma receiving encorafenib plus binimetinib versus vemurafenib or encorafenib (COLUMBUS): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2018;19: 1315–1327. doi: 10.1016/s1470-2045(18)30497-2.
25. Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF -mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2018;19: 603–615. doi: 10.1016/s1470-2045(18)30142-6.
26. Ascierto PA, McArthur GA, Dréno B, Atkinson V, Liszkay G, Di Giacomo AM, et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): Updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol 2016;17: 1248–1260. doi: 10.1016/s1470-2045(16)30122-x.
27. Ascierto PA, Dummer R, Gogas HJ, Flaherty KT, Arance A, Mandala M, et al. Update on tolerability and overall survival in COLUMBUS: Landmark analysis of a randomised phase 3 trial of encorafenib plus binimetinib vs vemurafenib or encorafenib in patients with BRAF V600-mutant melanoma. Eur J Cancer 2020;126: 33–44. doi: 10.1016/j.ejca.2019.11.016.
28. Saiag P, Robert C, Grob JJ, Mortier L, Dereure O, Lebbe C, et al. Efficacy, safety and factors associated with disease progression in patients with unresectable (stage III) or distant metastatic (stage IV) BRAF V600-mutant melanoma: An open label, non-randomized, phase IIIb study of trametinib in combination with dabrafenib. Eur J Cancer 2021;154: 57–65. doi: 10.1016/j.ejca.2021.05.031.
29. Davies MA, Saiag P, Robert C, Grob JJ, Flaherty KT, Arance A, et al. Dabrafenib plus trametinib in patients with BRAFV600-mutant melanoma brain metastases (COMBI-MB): A multicentre, multicohort, open-label, phase 2 trial. Lancet Oncol 2017;18: 863–873. doi: 10.1016/S1470-2045(17)30429-1.
30. Long GV, Hauschild A, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, et al. Adjuvant dabrafenib plus trametinib in stage III BRAF-mutated melanoma. N Engl J Med 2017;377: 1813–1823. doi: 10.1056/NEJMoa1708539.
31. Schadendorf D, Hauschild A, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, et al. Patient-reported outcomes in patients with resected, high-risk melanoma with BRAFV600E or BRAFV600K mutations treated with adjuvant dabrafenib plus trametinib (COMBI-AD): A randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2019;20: 701–710. doi: 10.1016/S1470-2045(18)30940-9.
32. Dummer R, Hauschild A, Santinami M, Atkinson V, Mandalà M, Kirkwood JM, et al. Five-year analysis of adjuvant dabrafenib plus trametinib in stage III melanoma. N Engl J Med 2020;383: 1139–1148. doi: 10.1056/NEJMoa2005493.
33. Maio M, Lewis K, Demidov L, Mandalà M, Bondarenko I, Ascierto PA, et al. Adjuvant vemurafenib in resected, BRAF V600 mutation-positive melanoma (BRIM8): A randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol 2018;19: 510–520. doi: 10.1016/s1470-2045(18)30106-2.
34. Amaria RN, Prieto PA, Tetzlaff MT, Reuben A, Andrews MC, Ross MI, et al. Neoadjuvant plus adjuvant dabrafenib and trametinib versus standard of care in patients with high-risk, surgically resectable melanoma: A single-centre, open-label, randomised, phase 2 trial. Lancet Oncol 2018;19: 181–193. doi: 10.1016/s1470-2045(18)30015-9.
35. Long GV, Saw RPM, Lo S, Nieweg OE, Shannon KF, Gonzalez M, et al. Neoadjuvant dabrafenib combined with trametinib for resectable, stage IIIB–C, BRAFV600 mutation-positive melanoma (NeoCombi): A single-arm, open-label, single-centre, phase 2 trial. Lancet Oncol 2019;20: 961–971. doi: 10.1016/s1470-2045(19)30331-6.
36. Blankenstein SA, Rohaan MW, Klop WMC, van der Hiel B, Lahaye MJ, et al.; van de Wiel BA Neoadjuvant cytoreductive treatment with BRAF/MEK inhibition of prior unresectable regionally advanced melanoma to allow complete surgical resection, REDUCTOR: A prospective, single-arm, open-label phase II trial. Ann Surg 2021;274: 383–389. doi: 10.1097/SLA.0000000000004893.
37. Ferrucci PF, Lens M, Cocorocchio E. Combined BRAF-targeted therapy with immunotherapy in BRAF-mutated advanced melanoma patients. Curr Oncol Rep 2021;23: 138. doi: 10.1007/s11912-021-01134-7.
38. Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, 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. doi: 10.1016/s0140-6736(20)30934-x.
39. Ferrucci PF, Di Giacomo AM, Del Vecchio M, Atkinson V, Schmidt H, Schachter J, 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. doi: 10.1136/jitc-2020-001806.
40. Dummer R, Long GV, Robert C, Tawbi HA, Flaherty KT, Ascierto PA, et al. Randomized phase III trial evaluating spartalizumab plus dabrafenib and trametinib for BRAF V600-mutant unresectable or metastatic melanoma. J Clin Oncol 2022;40: 1428–1438. doi: 10.1200/JCO.21.01601.
41. Arshad J, Ahmed J, Subhawong T, Trent JC. Progress in determining response to treatment in gastrointestinal stromal tumor. Expert Rev Anticancer Ther 2020;20: 279–288. doi: 10.1080/14737140.2020.1745068.
42. Cavnar MJ, Seier K, Curtin C, Balachandran VP, Coit DG, Yoon SS, et al. Outcome of 1000 patients with gastrointestinal stromal tumor (GIST) treated by surgery in the pre- and post-imatinib eras. Ann Surg 2021;273: 128–138. doi: 10.1097/SLA.0000000000003277.
43. Basurto-Lozada P, Molina-Aguilar C, Castaneda-Garcia C, Vázquez-Cruz ME, Garcia-Salinas OI, Álvarez-Cano A, et al. Acral lentiginous melanoma: Basic facts, biological characteristics and research perspectives of an understudied disease. Pigment Cell Melanoma Res 2021;34: 59–71. doi: 10.1111/pcmr.12885.
44. 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. doi: 10.1200/JCO.2010.33.9275.
45. 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. doi: 10.1200/JCO.2012.47.7836.
46. Cho JH, Kim KM, Kwon M, Kim JH, Lee J. Nilotinib in patients with metastatic melanoma harboring KIT gene aberration. Invest New Drugs 2012;30: 2008–2014. doi: 10.1007/s10637-011-9763-9.
47. Lee SJ, Kim TM, Kim YJ, Jang KT, Lee HJ, Lee SN, et al. Phase II trial of nilotinib in patients with metastatic malignant melanoma harboring KIT gene aberration: A multicenter trial of Korean cancer study group (UN10-06). Oncologist 2015;20: 1312–1319. doi: 10.1634/theoncologist.2015-0161.
48. Guo J, Carvajal RD, Dummer R, Hauschild A, Daud A, Bastian BC, 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. doi: 10.1093/annonc/mdx079.
49. Carvajal RD, Lawrence DP, Weber JS, Gajewski TF, Gonzalez R, Lutzky J, et al. Phase II study of nilotinib in melanoma harboring KIT alterations following progression to prior KIT inhibition. Clin Cancer Res 2015;21: 2289–2296. doi: 10.1158/1078-0432.CCR-14-1630.
50. Delyon J, Chevret S, Jouary T, Dalac S, Dalle S, Guillot B, et al. STAT3 mediates nilotinib response in KIT-altered melanoma: A phase II multicenter trial of the French skin cancer network. J Invest Dermatol 2018;138: 58–67. doi: 10.1016/j.jid.2017.07.839.
51. Janku F, Bauer S, Shoumariyeh K, Jones RL, Spreafico A, Jennings J, et al. Phase I study of ripretinib, a broad-spectrum KIT and PDGFRA inhibitor, in patients with KIT-mutated or KIT-amplified melanoma. Ann Oncol 2021;32: S896–S897. doi: 10.1016/j.annonc.2021.08.1467.
52. Delyon J, Lebbe C, Dumaz N. Targeted therapies in melanoma beyond BRAF: Targeting NRAS-mutated and KIT-mutated melanoma. Curr Opin Oncol 2020;32: 79–84. doi: 10.1097/CCO.0000000000000606.
53. Randic T, Kozar I, Margue C, Utikal J, Kreis S. NRAS mutant melanoma: Towards better therapies. Cancer Treat Rev 2021;99: 102238. doi: 10.1016/j.ctrv.2021.102238.
54. Dummer R, Schadendorf D, Ascierto PA, Arance A, Dutriaux C, Di Giacomo AM, et al. Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2017;18: 435–445. doi: 10.1016/s1470-2045(17)30180-8.
55. Lebbé C, Dutriaux C, Lesimple T, Kruit W, Kerger J, Thomas L, et al. Pimasertib versus dacarbazine in patients with unresectable NRAS-mutated cutaneous melanoma: Phase II, randomized, controlled trial with crossover. Cancers (Basel) 2020;12: 1727. doi: 10.3390/cancers12071727.
56. Lebbé C, Italiano A, Houédé N, Awada A, Aftimos P, Lesimple T, et al. Selective oral MEK1/2 inhibitor pimasertib in metastatic melanoma: Antitumor activity in a phase I, dose-escalation trial. Target Oncol 2021;16: 47–57. doi: 10.1007/s11523-020-00767-1.
57. Forschner A, Forchhammer S, Bonzheim I. NTRK gene fusions in melanoma: Detection, prevalence and potential therapeutic implications. J Dtsch Dermatol Ges 2020;18: 1387–1392. doi: 10.1111/ddg.14160.
58. Couts KL, McCoach CE, Murphy D, Christiansen J, Turner J, Lewis KD, et al. Acral lentiginous melanoma harboring a ROS1 gene fusion with clinical response to entrectinib. JCO Precis Oncol 2017;1: 1–7. doi: 10.1200/po.16.00013.
59. Drilon A, Siena S, Ou SI, Patel M, Ahn MJ, Lee J, et al. Safety and antitumor activity of the multitargeted Pan-TRK, ROS1, and ALK inhibitor entrectinib: Combined results from two phase I trials (ALKA-372-001 and STARTRK-1). Cancer Discov 2017;7: 400–409. doi: 10.1158/2159-8290.CD-16-1237.
60. Boumahdi S, de Sauvage FJ. The great escape: Tumour cell plasticity in resistance to targeted therapy. Nat Rev Drug Discov 2020;19: 39–56. doi: 10.1038/s41573-019-0044-1.
Keywords:

Melanoma; Molecular targeted therapy; BRAF inhibitors, KIT inhibitors

Copyright © 2023 The Chinese Medical Association, produced by Wolters Kluwer, Inc. under the CC-BY-NC-ND license.