Management of Advanced Invasive Melanoma: New Strategies

Reprinted with permission. Adv Ther. 2023;40:3381–3394*

by Alessia Villani, Massimiliano Scalvenzi, Giuseppe Micali, Francesco Lacarrubba,
Luigi Fornaro,  Fabrizio Martora, 
Luca Potestio

Authors Villani, Scalvenzi, Fornaro, and Martora are with Dermatology Unit, Department of Clinical Medicine  and Surgery, University of Naples Federico II, in Naples, Italy. Authors Lacarrubba and Micali are with Dermatology Clinic, University of Catania, in Catania, Italy.

FUNDING: Open access funding provided by Universita` degli Studi di Napoli Federico II within the CRUI-CARE Agreement.

DISCLOSURES: The authors report no conflicts of interest relevant to the content of this article. 


ABSTRACT: The incidence of cutaneous melanoma (CM) is increasing. CM is defined as melanoma in situ when limited within the epidermis and invasive when atypical melanocytes progressively invade the dermis. Treatment of CM is challenging. On one hand, melanoma in situ does not require further treatment except for a limited secondary excision with reduced margins to minimize the risk of local recurrences; on the other, invasive melanoma requires a personalized approach based on tumor staging. Consequently, an association of surgical and medical treatments is often necessary for invasive forms of the disease. In this scenario, new knowledge on melanoma pathogenesis has led to the development of safe and effective treatments, and several drugs are currently under investigation. However, extensive knowledge is required to offer patients a tailored-tail approach. The aim of our article was to review current literature to provide an overview of treatment options for invasive melanoma, highlighting strategical approaches that can be used in patients with these forms of disease.

Key Summary Points

Why carry out this study? Treatment of cutaneous melanoma is challenging. On one hand, melanoma in situ does not require further treatments except for a limited secondary excision with reduced margins to minimize the risk of local recurrences; on the other, invasive melanoma requires a personalized approach based on tumor staging. New knowledge on melanoma pathogenesis has led to the development of safe and effective treatments, and several drugs are currently under investigation; however, extensive knowledge is required to offer patients a tailored approach.

What was learned from the study? The management of metastatic melanoma has been revolutionized by the introduction of immune checkpoint inhibitors and targeted therapies. Moreover, the association of targeted therapies and immunotherapies and their sequential use has been shown to be a valid weapon in melanoma management. Finally, other therapeutic options (e.g., T-cell agonists, intravenous oncolytic virus, vaccines, cytokines) are currently under investigation. The future of melanoma management will be characterized by the association of surgical and medical interventions, as well as several biomarkers that will allow identifying at-risk patients and defining the right treatment at the right moment.

Keywords: Melanoma, management, invasive melanoma, immune checkpoint inhibitors, targeted therapy, vaccines, small molecules

*Copyright: © 2023 The Author(s). This article is licensed under a Creative Commons Attribution-Non- Commercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.** The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http:// creativecommons.org/licenses/by-nc/4.0/.

**Minor grammatical and style edits were made to this article.


Cutaneous melanoma (CM) is a malignant tumor arising from melanocytes located in skin tissue1 and accounts for greater than 90 percent of the deaths caused by cutaneous tumors.2,3 The worldwide prevalence of CM is increasing, with 232,100 new diagnoses and about 55,500 cancer deaths per year.4–6 Several risk factors have been associated with CM, including exposure to ultraviolet (UV) radiation, sunburns, indoor tanning, high socioeconomic status, presence of melanocytic or dysplastic naevi, a family or personal history of CM, and certain phenotypic features (e.g., fair hair, eye, and skin colors and the tendency to freckle).7,8  Of note, melanoma may also derive from melanocyte residents on the meninges, eyes, and various mucosal surfaces.9

Clinically, four common subtypes of CM can be distinguished: superficial spreading melanoma (about 41% of cases), nodular melanoma (about 16% of cases), lentigo maligna melanoma (2.7–14% of cases), and acral lentiginous melanoma (1–5% of cases); uncommon subtypes include desmoplastic melanoma, spitzoid melanoma, and amelanotic or hypomelanotic melanoma.10–12

 CM is defined as melanoma in situ when limited within the epidermis and invasive melanoma when atypical melanocytes progressively invade the dermis.13 Regarding CM staging, tumor thickness (Breslow score, which measures the depth of the melanoma from the granular layer of the epidermis down through to the deepest point of the tumor),14 lymph node involvement, and the presence of metastasis are the key factors, categorizing melanoma severity into four stages: Stages I and II (localized disease), Stage III (local lymph nodes involvement), and Stage IV (distant metastasis).13 Moreover, vertical tumor thickness (Breslow’s depth), presence of ulceration, mitotic rate, and level of invasion (Clark’s level, used to determine the number of the skin into whiich the melanoma has grown) are the main prognostic factors for CM.13,15,16  The majority of CMs (about 90%) are diagnosed as primary tumors without metastasis.13,16,17 

Current guidelines proposed by the American Academy of Dermatology (AAD) and a collaboration of multidisciplinary experts from the European Association of Dermato-Oncology, the European Organization of Research and Treatment of Cancer, and the European Dermatology Forum recommend that local excision with different safety margins, depending on Breslow’s thickness, is the mainstay of treatment for CM, followed by sentinel lymph node biopsy (SLNB) if tumor thickness is greater than 0.8mm (if histologic examination has revealed additional risk factors).13,15,16 Although excision with a safety margin and eventual SLNB are the standard of care in patients with melanoma, this approach may not bring resolution in patients with invasive melanoma. 

The aim of our study was to review current literature and provide a complete overview regarding treatment options for invasive melanoma to highlight strategic approaches that can be used in patients with these forms of disease. Of note, we focused our attention on tumor stage to describe and provide the correct treatment option tailored for each patient at the right time.

Methods

A search of the current literature was performed using the Embase, PubMed, Cochrane Skin and www.clinicaltrials.gov databases (until 15 March 2023) using the following research terms: “cutaneous melanoma,” “invasive melanoma,” “surgical treatment,” “sentinel lymph node biopsy,” “immunotherapy,” “checkpoint immunotherapy,” “targeted therapies,” “ipilimumab,” “pembrolizumab,” “nivolumab,” “BRAF inhibitors,” “vemurafenib,” “dabrafenib,” “MEK inhibitors,” “binimetinib,” “trametinib,” “sequential treatment,” “vaccines,” “anti-vascular endothelial growth factor,” “cytokines,” “inhibitory molecules,” and “T-cell agonists.” Investigated articles included reviews, meta-analyses, clinical trials, real-world studies, and case series and reports. The most relevant articles were examined. Bibliographies were reviewed to include articles that could have been missed. Articles regarding treatments for noninvasive CM were not considered. Finally, the research was refined by reviewing the abstracts and texts of collected texts. Of note, only English language articles were considered, while other language manuscripts were excluded. The current review article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.

Results

Herein, we report the current therapeutic scenario for CM and provide an overview on the currently used systemic therapies, as well as drugs under development.

Current therapeutic scenario. Surgical treatment. Surgery is the gold standard treatment of CM.18 When melanoma is clinically and dermoscopically suspected, an excisional biopsy should be performed to allow a histologic diagnosis.18 Of note, the use of reflectance confocal microscopy (RCM) for noninvasive imaging of the skin has shown promising results in cancer diagnosis, limiting use of the surgical approach only to at risk cases.19,20 However, RCM is currently not available in tertiary referral centers, nor is the knowledge of the pros and cons of this tool; its clinical applicability is currently under investigation.21

Generally, incisional biopsies may be considered on large CM lesions or lesions located on sensitive areas, such as lentigo maligna, acral, and genital lesions.13,15,16 In cases of histologic confirmation of CM, a subsequent excision should be performed, thus reducing the risk of local recurrences.18 However, peripheral surgical margins depend on tumor thickness.

One-centimeter margins are recommended for CM with a Breslow thickness greater than 2mm (except for melanoma in situ, which is not considered invasive melanoma when margins are reduced to 0.5cm), while 2cm margins are required for CM (Breslow) 2mm  or greater.13,15,16 Of note, in patients presenting with lentigo maligna melanoma or genital and acral melanoma, a microscopically controlled surgery (Mohs micrographic surgery) may be a valuable option to spare tissue, ensuring complete tumor resection.13,15,16,22

SLNB. SLNB is a standard staging procedure recommended for patients with CM that is thicker than 1mm or 0.8mm if histologic examination has revealed additional risk factors (e.g., ulceration, 1 mitosis/mm2 or greater, and/or lymphovascular invasion, particularly in the younger patients).13,15,16 In patients with negative sentinel lymph nodes, no further actions are needed.13,15,16 Contrarily, in cases of positive lymph node involvement, complete lymph node dissection, previously routinely practiced, has been replaced by adjuvant therapies after similar outcomes have been reported in clinical trials involving patients with microscopic nodal metastases undergoing or not undergoing complete lymph node dissection.13,15,16 

Metastasis. CM metastasis should be divided into satellite and/or in-transit metastasis, defined as lesions occurring within 2cm of the primary tumor and any cutaneous or subcutaneous metastases that are greater than 2cm from the primary lesion but are not beyond the regional nodal basin, respectively, and distant metastases.23 Surgery or other destructive therapies (e.g., radiotherapy, cryotherapy, electrochemotherapy, laser therapy) can be used for the management of satellite and/or in-transit metastasis while distant metastasis can be managed with complete resection or other destructive procedures only in cases of oligometastatic disease or as a palliative procedure.13,15,16 For metastatic disease, several systemic therapies are available.13,15,16

Neoadjuvant treatment. Neoadjuvant therapy is defined as a first step treatment to reduce tumor size before the gold standard treatment.24,25 Neoadjuvant therapy should be considered in patients with resectable metastasis, particularly Stage III melanoma.24,25 While trials investigating the use of adjuvant treatment in CM management are scant, preliminary data suggest that immunotherapy is superior to targeted therapy, resulting in high response rates, but with lower durability.13,15,16 Moreover, it has been reported that combination of ipilimumab [anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4)] plus nivolumab [anti-programmed cell death protein-1 (PD-1)] is more effective than monotherapy with an anti-PD-1 agent and that the initial pathologic response is predictive of recurrence-free survival (RFS).24,25

Adjuvant treatment. Adjuvant therapy is a treatment given in addition to the primary or initial therapy to maximize its effectiveness.13,15,16 Adjuvant therapy plays a key role in melanoma management in that its use has been shown to impact overall survival (OS) and RFS.26 Historically, interferon-alpha (α) was used as adjuvant therapy in melanoma patients with tumors greater than 1.5mm, despite a significant toxicity profile.26 Currently, new effective and safe drugs, such as immune checkpoint inhibitors [PD-1 and its ligand (PDL-1) and anti-CTLA-4] and targeted therapies (BRAF/MEK inhibitors) have replaced its use.26 Currently, the adjuvant therapy is offered to patients with lymph node involvement using BRAF/MEK inhibitor if BRAFV600 E/K mutation is detected. Of note, anti-PD-1 can be used regardless of the mutation status.26 Moreover, for patients who develop progressive disease more than six months after adjuvant therapy, re-challenge can be considered, evaluating BRAF status also in case of acquired resistance setting.26 For BRAF-mutated melanoma, switching from immunotherapy to targeted therapy or vice versa can be considered, whereas combination therapy with anti-PD-1 plus ipilimumab or ipilimumab alone can be an option in patients without BRAF mutation.26

Management of systemic disease. Recent knowledge on CM pathogenesis has led to the development of safer and more effective treatments, leading to an improvement in the RFS, OS, progression-free survival (PFS), disease free survival (DFS), durable response rate (DRR), and overall response rate (ORR).17 The introduction of these drugs has allowed avoiding the use of chemotherapy, which is now only considered as a last-line treatment in patients with resistance to immunotherapies and targeted therapies (if applicable).17 However, many patients with Stage IV disease do not benefit from these therapies and have died from the disease.17 Immune checkpoint inhibitors are used as a first-line treatment option, regardless BRAF status, as anti-PD-1 monotherapy or a combination of anti-PD-1 plus anti- CTLA-4 [13, 15, 16]. Targeted therapies are used in patients with resistance to immunotherapy, with BRAF-V600 E/K mutation, or as first-line option in selected cases .13,15,16 Currently approved drugs for the management of melanoma, along with mechanisms of action and other therapeutic indications, are reported in Table 1. 


Overview on the currently used systemic therapies. Immune checkpoint inhibitors. Immune checkpoint inhibitors target small proteins produced by immune cells and cancer cells called “checkpoints.”27,28 Among these, PD-1 and CTLA-4 are checkpoints produced by cancer cells to escape from the immune system through the downregulation of T-cell activation, leading to immune tolerance.27,28 On one hand, the binding of PD-1, expressed on the surface of monocytes, T and B and natural killer cells, to its ligand PDL-1 promotes the apoptosis of T lymphocytes and activates the regulatory T cells, preventing the inflammation cascade; on the other hand, CTLA-4 is constitutively expressed in regulatory T lymphocytes and prevents the activation of T cells. Thus, targeting these cytokines is a valuable option to reduce melanoma immune escape mechanisms.27,28

Nivolumab. Nivolumab is an immune checkpoint inhibitor approved for the management of metastatic or unresectable melanoma and as adjuvant therapy. It acts through the blockage of PD-L1 binding to PD-1.29 It can be used as monotherapy or combination therapy for melanoma management and is also approved for the treatment of non-small cell lung cancer (NSCLC), malignant pleural mesothelioma (MPM), renal cell carcinoma (RCC), classical Hodgkin lymphoma (cHL), squamous cell cancer of the head and neck (SCCHN), urothelial carcinoma, mismatch repair deficient (dMMR) or microsatellite instability-high (MSI-H) colorectal cancer (CRC), esophageal squamous cell carcinoma (OSCC), adjuvant treatment of esophageal or gastroesophageal junction cancer (OC or GEJC), and gastric, gastro-esophageal junction (GEJ) or esophageal adenocarcinoma.30

Regarding melanoma management, nivolumab is scheduled as an intravenous infusion at a dosage of 240mg every two weeks (Q2W) or 480mg every four weeks (Q4W) as monotherapy or in combination with ipilimumab (nivolumab 1mg/kg + ipilimumab 3mg/kg every 3 weeks [Q3W] up to 4 doses followed by nivolumab as single agent at standard dosage of 240mg Q2W or 480mg Q4W).30 Nivolumab was reported to be superior to chemotherapy in previously treated (ipilimumab and BRAF inhibitor, if BRAF V600 mutation was positive) metastatic melanoma (CHECKMATE-037 study) and in untreated patients with metastatic melanoma (CHECKMATE-066 study).31,32 Moreover, nivolumab as single treatment or combined with ipilimumab was shown to be statistically significantly more effective than ipilimumab (CHECKMATE-067 study) in untreated patients with unresectable or metastatic melanoma.33 Finally, nivolumab was reported to be significantly superior to ipilimumab also as adjuvant treatment (CHECKMATE-238 study).34

Pembrolizumab. Pembrolizumab is an anti-PD-1 drug currently approved for the treatment of metastatic or unresectable melanoma and as adjuvant treatment of patients with melanoma. Pembrolizumab is administered at a scheduled dosage of 200mg every three weeks (Q3W) or 400mg every six weeks (Q6W).35 Pembrolizumab is also approved for NSCLC, cHL, SCCHN, RCC, urothelial carcinoma, dMMR or MSI-H cancers, esophageal carcinoma, triple-negative breast cancer (TNBC), endometrial carcinoma and cervical cancer.35

Pembrolizumab has been reported to be significantly superior to ipilimumab for the treatment of untreated metastatic or unresectable melanoma (KEYNOTE-006 study) and to chemotherapy in patients with previously treated metastatic or unresectable disease (KEYNOTE-002 study).36,37 Finally, a statistically significant improvement in RFS was reported in patients receiving pembrolizumab compared to placebo as adjuvant treatment of resected melanoma (KEYNOTE-054 study).38

Ipilimumab. Ipilimumab is an anti-CTLA-4 antibody targeting CTLA-4 activity, approved at the dosage of 3mg/kg Q3W for up to four doses.39 As adjuvant treatment, it has been reported to be significantly superior to placebo in terms of RFS (CA184-029 study).39 Studies reporting its effectiveness as combination therapy with nivolumab in metastatic disease have been previously reported. Currently, ipilimumab is also approved for RCC, NSCLC, MPM, dMMR or MSI-H CRC, and OSCC.39

Targeted therapies. BRAF is a serine/threonine protein kinase that activates the mitogen-activated protein (MAP) kinase/ERK-signaling pathway.39 This signaling cascade leads to the evasion of apoptosis, senescence, and immune response; unchecked replicative potential; angiogenesis; tissue invasion; and metastasis.39 Globally, about 50 percent of melanomas have BRAF mutations. In particular, over 90 percent of these are located on codon 600, with 90 percent represented by a single nucleotide mutation resulting in substitution of glutamic acid for valine (BRAFV600E: nucleotide 1799 T>A; codon GTG>GAG).39 Combining BRAF and MEK inhibition is an innovative strategy for melanoma management.39

Dabrafenib. Dabrafenib is a kinase inhibitor approved for the treatment of metastatic or unresectable melanoma with a BRAF V600E or V600K mutation and as an adjuvant treatment at the dosage of 150mg twice daily, used either as monotherapy or in combination with trametinib.40 It was also approved for the management of NSCLC.40 Dabrafenib was shown to be significantly superior to chemotherapy in melanoma management (BREAK-3 study).41 Dabrafenib has also been investigated in metastatic melanoma with brain metastases (COMBI-d study and COMBI-v study).42 Finally, the superiority of dabrafenib plus placetinib with respect to placebo as adjuvant treatment has been reported (COMBI-AD study).43

Trametinib. Trametinib is a kinase inhibitor targeting MEK1 and MEK2, approved for the management of metastatic or BRAF V600E- or BRAF V600K-mutated, unresectable melanoma or as adjuvant treatment at an oral dosage of 2mg a day, used as either monotherapy or in combination with dabrafenib.44 It is also approved for NSCLC.44 Trametinib was shown to be significantly superior to chemotherapy in metastatic melanoma (METRIC study) as well as in combination with dabrafenib in brain metastases or adjuvant therapy (COMBI-d and COMBI-AD studies, respectively).42,43,45

Vemurafenib. Vemurafenib is a targeted therapy approved in monotherapy, four tablets of 240mg every 12 hours, for the management of patients with metastatic or unresectable melanoma with BRAF V600E mutation.46 A statistically significant superiority to chemotherapy has been reported (BRIM3 study), also in previously treated patients and in patients with metastatic BRAF V600E mutation-positive melanoma with brain metastasis.46

Encorafenib and binimetinib. Encorafenib is a kinase inhibitor approved for the treatment of unresectable or metastatic melanoma with BRAF V600E or BRAF V600K mutation in combination with binimetinib at the dosage of 450mg once a day. It is also approved in combination with cetuximab for the treatment of CRC in adult patients.47

Binimetinib is a MEK inhibitor approved for the management of metastatic or BRAF V600E-mutated, unresectable melanoma at the dosage of 45mg twice a day in combination with encorafenib.48

Encorafenib + binimetinib was shown to be statistically significantly superior in terms of PFS compared to vemurafenib (CMEK162B2301 study).47,48

Emerging treatments. Several treatment strategies or clinical options are currently under investigation for the management of melanoma. First, the combination or sequential use of immune checkpoint inhibitors plus targeted therapies is considered a valuable option due to their high effectiveness. Many trials on this subject are ongoing.24 However, several drugs are under investigation to combat acquired resistance to both targeted therapy and immunotherapy. Among these, an oncolytic viral immunotherapy (talimogene laherparepvec [TVEC]) has been recently approved for the local treatment of unresectable, metastatic, Stage IIIB/C and IV melanoma.49 However, its place in the combined therapy of T-VEC and anti-PD-1 is unclear.49 Other drugs, such as engineered cytokine (e.g., darleukin and bempegaldesleukin), intravenous oncolytic virus (e.g., ICOVIR-5), antivascular endothelial growth factor (e.g., bevacizumab), drugs targeting inhibitory molecules (e.g., colony-stimulating factor 1 receptor inhibitors and indoleamine 2,3-dioxygenase 1 inhibitors), t-cell agonists, Toll-like receptor agonists, agonistic anti-OX40 antibodies, glucocorticoid-induced tumor necrosis factor receptor family-related protein, and adoptive T-cell therapy, are currently under investigation, opening a new era in the therapy of advanced melanoma.50–60 Finally, another potential treatment option in CM under inbvestigation is the combination of immunotherapy and local therapies such as radiotherapy.61 In particular, radiotherapy may be used when starting immunotherapy peri-induction radiotherapy (PIR) or after systemic treatment failure as postescape radiotherapy (PER) to improve treatment outcomes.61 Indeed, radiation can increase tumor antigen visibility and promote priming of T cells but can also exert immunosuppressive action on the tumor microenvironment.61 To sum up, the combination of immunotherapy and radiotherapy provides an opportunity to increase the immunostimulatory potential of radiation.61

Discussion

The increasing knowledge on the pathogenesis of skin cancers has led to the development of more effective and safer treatments.62–67 In particular, new strategies have been adopted for the management of melanoma.68–70 While­ in-situ melanoma does not require further treatments except for a limited secondary excision with reduced margins to minimize the risk of local recurrences, invasive melanomas (defined by the presence of atypical melanocytes invading the dermis) require a personalized approach based on tumor staging (tumor thickness + lymph node involvement + presence of metastasis).13,15,16 Even if a secondary excision with larger margins (1cm if thickness <2mm, 2cm if thickness ≥2mm) is required for invasive melanoma, SLNB is recommended only for patients with CM greater than 1mm in thickness or 0.8mm or greater in the presence of peculiar histologic features.13,15,16. Although the therapeutic approach seems standardized up to this point, several strategies may be used to personalize the management of patients with a thick melanoma without lymph node involvement or with an unresectable disease. In this scenario, some patients may benefit from neoadjuvant therapies. However, neoadjuvant treatments are not yet a standard of care because there is a lack of data demonstrating the superiority of neoadjuvant approaches over conventional surgery plus postoperative adjuvant treatment, though Phase III trials are ongoing. Similarly, although adjuvant treatment is well established for patients with Stages III or IV melanoma, a subgroup of patients with Stage II disease may benefit from adjuvant therapies because of the high risk of tumor recurrence. Regarding the management of metastatic melanoma, it has been revolutionized by the introduction of immune checkpoint inhibitors and targeted therapies. Moreover, the association of targeted therapies and immunotherapies or their sequential use has been shown to be effective in melanoma management. Finally, other therapeutic options (e.g., T-cell agonists, intravenous oncolytic virus, vaccines, cytokines) are currently under investigation as therapeutic alternatives in patients unresponsive to immune checkpoint inhibitors or targeted therapies. In our opinion, the future of melanoma management will be characterized by the association of surgical and medical interventions, as well as several biomarkers that will allow identifying at-risk patients and defining the right treatment at the right moment. Additionally studies are required to increase our knowledge on melanoma pathogenesis and treatment to enable us to offer therapeutic options that increase survival with good safety profiles.

Conclusion

The management of invasive melanoma is challenging. Despite current guidelines that seem to offer a complete overview, there are still many uncovered areas. In this scenario, several studies are needed to establish the role of innovative drugs and surgical procedures in melanoma management at each stage of the disease to increase treatment efficacy and reduce safety concerns.

Acknowledgements

Author contributions. Alessia Villani: data curation, formal analysis, investigation, visualization, writing-original draft preparation, writing-review, and editing; Massimiliano Scalvenzi: data curation, formal analysis, investigation, visualization, writing-original draft preparation, writing-review, and editing; Giuseppe Micali and Francesco Lacarrubba: data curation, formal analysis, investigation, visualization, writing-original draft preparation, writing-review, and editing; Luigi Fornaro: data curation, formal analysis, investigation, visualization, and writing-original draft preparation; Fabrizio Martora: data curation, formal analysis, investigation, visualization, and writing-original draft preparation; Luca Potestio: data curation, formal analysis, investigation, visualization, writing- original draft preparation, writing-review, and editing; All authors read and approved the final version of the manuscript.

Compliance and ethics guidelines. This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.

Data availability. Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

  1. Leonardi GC, Falzone L, Salemi R, et al. Cutaneous melanoma: from pathogenesis to therapy (review). Int J Oncol. 2018;52(4):1071–1080. 
  2. Villani A, Scalvenzi M, Fabbrocini G, et al. Looking into a better future: novel therapies for metastatic melanoma. Dermatol Ther (Heidelb). 2021;11(3):751–767.
  3. Abbas O, Miller DD, Bhawan J. Cutaneous malignant melanoma: update on diagnostic and prognostic biomarkers. Am J Dermatopathol. 2014;36(5):363–379. 
  4. Tı´ma´r J, Lada´nyi A. Molecular pathology of skin melanoma: epidemiology, differential diagnostics, prognosis and therapy prediction. Int J Mol Sci. 2022. 
  5. Villani A, Scalvenzi M, Fabbrocini G, et al. Effects of COVID-19 pandemic on malignant melanoma diagnosis. J Eur Acad Dermatol Venereol. 2023;37(1):e22–e23. 
  6. Villani A, Fabbrocini G, Scalvenzi M. The reduction in the detection of melanoma during the coronavirus disease 2019 (COVID-19) pandemic in a melanoma center of South Italy. J Dermatol Treat. 2022;33(3):1778. 
  7. Rastrelli M, Tropea S, Rossi CR, Alaibac M. Melanoma: epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo. 2014;28(6):1005–11.
  8. Dzwierzynski WW. Melanoma risk factors and prevention. Clin Plast Surg. 2021;48(4):543–550.
  9. Ahmed B, Qadir MI, Ghafoor S. Malignant melanoma: skin cancer-diagnosis, prevention, and treatment. Crit Rev Eukaryot Gene Expr. 2020;30(4):291–7. 
  10. Minini R, Rohrmann S, Braun R, et al. Incidence trends and clinical-pathological characteristics of invasive cutaneous melanoma from 1980 to 2010 in the Canton of Zurich, Switzerland. Melanoma Res. 2017;27(2):145–51. 
  11. Clark WHJ, From L, Bernardino EA, Mihm MC. The histogenesis and biologic behavior of primary human malignant melanomas of the skin. Cancer Res. 1969;29(3):705–27.
  12. Villani A, Fabbrocini G, Costa C, et al. Second primary melanoma: incidence rate and risk factors. J Eur Acad Dermatol Venereol. 2020;34(10):e623–e624. 
  13. Garbe C, Amaral T, Peris K, et al. European consensus-based interdisciplinary guideline for melanoma. Part 1: diagnostics: update 2022. Eur J Cancer. 2022;170:236–255. 
  14. Saldanha G, Yarrow J, Pancholi J, et al. Breslow density is a novel prognostic feature that adds valueto melanoma staging. Am J Surg Pathol. 2018;42(6): 715–725. 
  15. Swetter SM, Tsao H, Bichakjian CK, et al. Guidelines of care for the management of primary cutaneous melanoma. J Am Acad Dermatol. 2019;80(1):208–250. 
  16. Garbe C, Amaral T, Peris K, et al. European consensus-based interdisciplinary guideline for melanoma. Part 2: treatment—update 2022. Eur J Cancer. 2022;170:256–284. 
  17. Villani A, Potestio L, Fabbrocini G, et al. The treatment of advanced melanoma: therapeutic update. Int J Mol Sci. 2022. 
  18. Schadendorf D, van Akkooi ACJ, Berking C, et al. Melanoma. Lancet (London, England). 2018;392(10151):971–984.
  19. Villani A, Scalvenzi M, Peduto T, et al. Dermoscopy and reflectance confocal microscopy of Kaposi’s sarcoma: an overview. J Eur Acad Dermatol Venereol. 2022;36(4):e272–e274. 
  20. Potestio L, Villani A, Ocampo-Garza SS, et al. Dermatophytid evaluated by in vivo reflectance confocal microscopy: a new approach? Int J Dermatol. 2022;61(10):e394–e396. 
  21. Agozzino M, Moscarella E, Babino G,et al. The use of in vivo reflectance confocal microscopy for the diagnosis of melanoma. Expert Rev Anticancer Ther. 2019;19(5):413–421. 
  22. Golda N, Hruza G. Mohs micrographic surgery.Dermatol Clin. 2023;41(1):39–47.
  23. Poetsch M, Dittberner T, Woenckhaus C. Can different genetic changes characterize histogenetic subtypes and biologic behavior in sporadic malignant melanoma of the skin? Cell Mol Life Sci. 2003;60(9):1923–1932. 
  24. Curti BD, Faries MB. Recent advances in the treatment of melanoma. N Engl J Med. 2021;384(23):2229–2240. 
  25. Lee AY, Brady MS. Neoadjuvant immunotherapy for melanoma. J Surg Oncol. 2021;123(3):782–788.
  26. de Meza MM, Ismail RK, Rauwerdink D, et al. Adjuvant treatment for melanoma in clinical practice—trial versus reality. Eur J Cancer. 2021;158:234–45. 
  27. DeRogatis JM, Viramontes KM, Neubert EN, et al. Targeting the PSGL-1 immune checkpoint promotes immunity to PD-1 resistant melanoma. Cancer Immunol Res. 2022. 
  28. Lee JB, Kim HR, Ha SJ. Immune checkpoint inhibitors in 10 years: contribution of basic research and clinical application in cancer immunotherapy. Immune Netw. 2022;22(1): e2. 
  29. Koppolu V, RekhaVasigala VK. Checkpoint immunotherapy by nivolumab for treatment of metastatic melanoma. J Cancer Res Ther. 2018;14(6):1167–1175. 
  30. European Medicines Agency. ODIVO (Nivolumab): summary of product characteristics. 2014. https://www.ema.europa.eu/en/documents/product-information/opdivo-epar-product-information_en.pdf. Accessed on Mar 16, 2023.
  31. Larkin J, Minor D, D’Angelo S, et al. Overall survival in patients with advanced melanoma who received nivolumab versus investigator’s choice chemotherapy in CheckMate 037: a randomized, controlled, open-label phase III trial. J Clin Oncol Off J Am Soc Clin Oncol. 2018;36(4):383–90.
  32. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320–330.
  33. Hodi FS, Chiarion-Sileni V, Gonzalez R, et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018;19(11):1480–1492. 
  34. Ascierto PA, Del Vecchio M, Mandala´ M, et al. Adjuvant nivolumab versus ipilimumab in resected stage IIIB-C and stage IV melanoma (CheckMate 238): 4-year results from a multicentre, doubleblind, randomised, controlled, phase 3 trial. Lancet Oncol. 2020;21(11):1465–1477. 
  35. European Medicines Agency. KEYTRUDA (Pemrbolizumab): summary of product characteristics. 2015. https://www.ema.europa. eu/en/documents/product-information/keytrudaepar-product-information_en.pdf. Accessed on Mar 16, 2023.
  36. Ribas A, Puzanov I, Dummer R, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol. 2015;16(8):908–918. 
  37. Schachter J, Ribas A, Long GV, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet (London, England). 2017;390(10105):1853–1862.
  38. Eggermont AMM, Blank CU, Mandala` M, et al. Adjuvant pembrolizumab versus placebo in resected stage III melanoma (EORTC 1325-MG/KEYNOTE- 054): distant metastasis-free survival results from a double-blind, randomised, controlled, phase 3 trial. Lancet Oncol. 2021;22(5):643–54. 
  39. European Medicines Agency. YERVOY (Ipilimumab): summary of product characteristics. 2011. https://www.ema.europa.eu/en/documents/product-information/yervoy-eparproduct-information_en.pdf. Accessed on Mar 16, 2023.
  40. European Medicines Agency. TAFINLAR (Dabrafenib): Summary of Product Characteristics. 2013. https://www.ema.europa.eu/en/documents/product-information/tafinlar-eparproduct-information_en.pdf. Accessed on Mar 16, 2023.
  41. Latimer NR, Abrams KR, Amonkar MM, et al. Adjusting for the confounding effects of treatment switching—the BREAK-3 trial: dabrafenib versus dacarbazine. Oncologist. 2015;20(7):798–805. 
  42. Robert C, Grob JJ, Stroyakovskiy D, et al. Five-year outcomes with dabrafenib plus trametinib in metastatic melanoma. N Engl J Med. 2019;381(7):626–636. 
  43. Dummer R, Brase JC, Garrett J, et al. Adjuvant dabrafenib plus trametinib versus placebo in patients with resected, BRAF(V600)-mutant, stage III melanoma (COMBI-AD): exploratory biomarker analyses from a randomised, phase 3 trial. Lancet Oncol. 2020;21(3):358–372. 
  44. European Medicines Agency. MEKINIST (Trametinib): summary of product characteristics. 2014. https://www.ema.europa.eu/en/documents/product-information/mekinist-eparproduct-information_en.pdf. Accessed on Mar 16, 2023.
  45. Latimer NR, Bell H, Abrams KR, et al. Adjusting for treatment switching in the METRIC study shows further improved overall survival with trametinib compared with chemotherapy. Cancer Med. 2016;5(5):806–815. 
  46. European Medicines Agency. ZELBORAF (Vemurafenib): summary of product characteristics. 2012. https://www.ema.europa.eu/en/documents/product-information/zelboraf-eparproduct- information_en.pdf. Accessed on Mar 16, 2023.
  47. European Medicines Agency. BRAFTOVI (Encorafenib): summary of product characteristics. 2018. Encorafenib. 2018. https://www.ema.europa.eu/en/documents/productinformation/braftovi-epar-product-information_en.pdf. Accessed on Mar 16, 2022.
  48. European Medicines Agency. MEKTOVI (Binimetinib): summary of product characteristics. 2018. https://www.ema.europa.eu/en/documents/product-information/mektovi-eparproduct-information_en.pdf. Accessed on Mar 16, 2023.
  49. European Medicines Agency. IMLYGIC (Talimogene laherparepvec): summary of product characteristics. 2020. https://www.ema. europa.eu/en/documents/product-information/imlygic-epar-product-information_en.pdf. Accessed on Mar 16, 2023.
  50. Goff SL, Dudley ME, Citrin DE, et al. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol Off J Am Soc Clin Oncol. 2016;34(20):2389–2397. 
  51. Weide B, Eigentler T, Catania C, et al. A phase II study of the L19IL2 immunocytokine in combination with dacarbazine in advanced metastatic melanoma patients. Cancer Immunol Immunother. 2019;68(9):1547–1559. 
  52. Megna M, Camela E, Villani A, et al. Teledermatology: a useful tool also after COVID-19 era? J Cosmet Dermatol. 2022;21(6):2309–2310. 
  53.  Khushalani NI, Diab A, Ascierto PA, et al. Bempegaldesleukin plus nivolumab in untreated, unresectable or metastatic melanoma: Phase III PIVOT IO 001 study design. Future Oncol. 2020;16(28):2165–2175. 
  54. Corrie PG, Marshall A, Dunn JA, et al. Adjuvant bevacizumab in patients with melanoma at high risk of recurrence (AVAST-M): preplanned interim results from amulticentre, open-label, randomised controlled phase 3 study. Lancet Oncol. 2014;15(6):620–630.
  55. Neubert NJ, Schmittnaegel M, Bordry N, et al. T cell induced CSF1 promotes melanoma resistance to PD1 blockade. Sci Transl Med. 2018. 
  56. Jung KH, LoRusso P, Burris H, et al. Phase I study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) administered with PD-L1 inhibitor (Atezolizumab) in advanced solid tumors. Clin Cancer Res. 2019;25(11):3220–3228.
  57. Deng J, Zhao S, Zhang X, et al. OX40 (CD134) and OX40 ligand, important immune checkpoints in cancer. Onco Targets Ther. 2019;12:7347–7353.
  58.  Zappasodi R, Sirard C, Li Y, et al. Rational design of anti-GITR-based combination immunotherapy. Nat Med. 2019;25(5):759–766. 
  59. Scalvenzi M, Costa C, Cappello M, Villani A. Reply to Woltsche N. et al. Managing adverse effects by dose reduction during routine treatment of locally advanced basal cell carcinoma with the hedgehog inhibitor vismodegib: a single-centre experience. J Eur Acad Dermatol Venereol. 2019;33(4):e145–147.
  60. Villani A, Annunziata MC, Abategiovanni L, Fabbrocini G. Teledermatology for acne patients: how to reduce face-to-face visits during COVID-19 pandemic. J Cosmet Dermatol. 2020;19(8):1828. 
  61.  Tagliaferri L, Lancellotta V, Fionda B, et al. Immunotherapy and radiotherapy in melanoma: a multidisciplinary comprehensive review. Hum Vaccin Immunother. 2022;18(3):1903827. 
  62. Villani A, Cinelli E, Fabbrocini G, et al. Hedgehog inhibitors in the treatment of advanced basal cell carcinoma: risks and benefits. Expert Opin Drug Saf. 2020;19(12):1585–1594.
  63. Villani A, Fabbrocini G, Costa C, et al. Sonidegib with and without adjunctive treatment for locally advanced basal cell carcinomas. Oncologist. 2022;27(6): e533. 
  64. Villani A, Potestio L, Fabbrocini G, et al. Long-term efficacy of telemedicine for patients with locally advanced basal cell carcinoma during COVID-19 pandemic. Dermatol Ther. 2022. 
  65. Villani A, Potestio L, Fabbrocini G, Scalvenzi M. New emerging treatment options for advanced basal cell carcinoma and squamous cell carcinoma. Adv Ther. 2022;39(3):1164–1178. 
  66. Villani A, Ocampo-Garza SS, Potestio L, et al. Cemiplimab for the treatment of advanced cutaneous squamous cell carcinoma. Expert Opin Drug Saf. 2022;21(1):21–29. 
  67.  Scalvenzi M, Villani A, Mazzella C, et al. Vismodegib treatment in a HIV positive patient on antiretroviral therapy. Ind J Dermatol Venereol Leprol. 2018;84(6):758–760. 
  68. Jenkins RW, Fisher DE. Treatment of advanced melanoma in 2020 and beyond. J Invest Dermatol. 2021;141(1):23–31. 
  69. Eddy K, Chen S. Overcoming immune evasion in melanoma. Int J Mol Sci. 2020. 
  70. Alicea GM, Rebecca VW. Emerging strategies to treat rare and intractable subtypes of melanoma. Pigment Cell Melanoma Res. 2021;34(1):44–58.  
Facebook
Twitter
LinkedIn