Abstract
Neurocutaneous melanocytosis (NCM) is a rare, congenital condition primarily affecting children, characterized by large or giant congenital melanocytic nevi (L/GCMN) on the skin and pigmented lesions in the brain. The presence of pigmented lesions in the brain often leads to neurological symptoms like headaches, seizures, and hydrocephalus, typically manifesting before age two. Melanocytic lesions in the brain can range from benign melanocytosis to malignant melanoma. NCM is nearly always fatal if symptomatic, with a high risk of malignant transformation. Patients with larger skin nevi with neurological involvement tend to have a greater lifetime risk of malignancy. There is no specific treatment, and current therapies focus on palliative care, including surgery, radiation, and chemotherapy. Malignant transformation into melanoma requires aggressive treatments like surgery, radiation, and chemotherapy. Despite these approaches, outcomes remain poor, with no definitive cure for NCM. This study aims to review and critically evaluate the current therapeutic strategies for NCM while also exploring prospective avenues for developing specific and effective treatments. It aims to highlight recent advancements in the molecular understanding of NCM and examine how these insights may inform the development of novel therapeutic approaches. Additionally, the study will address the significant unmet medical needs associated with this rare and often fatal condition, emphasizing the importance of continued research and innovation in its management.
Neurocutaneous melanocytosis (NCM) is described as a neoplasm that is congenital in origin, categorized as a rare disease predominantly affecting the pediatric population, and characterized by large or giant congenital melanocytic nevi on the skin (L/GCMN) and pigmented lesions in the brain (1-3). This condition is also dubbed the Congenital Melanocytic Nevi (CMN) syndrome (4). Incidence of congenital skin nevi varies from 1/50,000-200,000 births, among them, a subset of patients also presents with CNS involvement. The risk of having a central nervous system (CNS) involvement appears to be proportional to the size of the cutaneous nevi. Brain lesions are commonly detected in the leptomeninges, frequently infiltrating into the brain parenchyma (5-7). The newborn may either remain asymptomatic or experience neurological symptoms that usually appear before two years of age. Frequently experienced symptoms are headache, nausea, vomiting, seizures, hydrocephalus, intracranial hypertension, and deficit in sensory and motor capabilities (8-10). Symptomatic NCM is almost 100% fatal, and in most symptomatic NCM cases reported in the literature, death has occurred within three years of the onset of symptoms. There is also an increased risk of malignant transformation from benign CNS or cutaneous lesions noted in these patients. Primary leptomeningeal melanoma arising within NCM patients was found to constitute about 9% of all published case reports of NCM (11). Although small and medium cutaneous nevi are thought to pose a lesser risk for malignancy, large or giant nevi (L/GCMN), on the contrary, remain at a greater lifetime risk of developing cutaneous melanoma (12). A worldwide registry of patients noted that approximately 6.68% of cases of L/GCMN had associated NCM, 65% of whom were symptomatic (13).
This study aims to review and discuss the current therapeutic strategies for neurocutaneous melanocytosis (NCM), as well as to explore future directions for developing specific and effective treatments. The study seeks to highlight recent research on the molecular aspects of NCM and how this knowledge can inform potential therapeutic advancements, addressing the significant unmet medical needs associated with this rare and often fatal condition. Although first described by Bohemian pathologist Rokitansky in 1861 (14), the molecular etiology of the disease remains unclear to date, and there is still no specific therapy for NCM. Current treatment options are mostly directed to palliative care, which includes surgery to relieve intracranial pressure, radiation, and treatment with chemotherapeutic drugs.
Methods
Inclusion and exclusion criteria. At the time of preparation of this manuscript, a search at the National Library of Medicine with keywords “Neurocutaneous melanocytosis” yielded only 116 results showing publications spanning a period from 1976 to the current year. For this review, only studies involving individuals with neurocutaneous melanocytosis, including children and adults, are included. Articles such as case reports, original experimental research, and reviews, as well as articles on diagnostic methods, treatment modalities, or management strategies for NCM, are included. Systematic reviews and meta-analyses relevant to NCM were included. Only peer-reviewed articles published in English were included. Non-peer-reviewed articles, opinion pieces, editorials, and non-original research were excluded. Studies not focused on neurocutaneous melanocytosis or involving unrelated conditions and articles that do not report on NCM-specific interventions or outcomes were excluded. This approach narrowed down to 92 publications to date on Pubmed, and in this review, we present the current understanding of the field based on a comprehensive review of these studies.
Clinical presentation and diagnostic criteria. Although rare, the clinical presentation of NCM is diverse. Reports of clinical characteristics of NCM in literature cover a wide spectrum of diffuse melanocytic proliferations in the meninges, ranging from melanocytosis, melanomatosis, melanocytoma, and melanoma (15-18). Diagnostically, there is consensus around the classical criteria provided by Kadonaga and Freiden in 1991, which is considered the gold standard for clinical diagnosis of NCM. According to these criteria, NCM diagnosis will be based on 1) evidence of congenital cutaneous nevi together with melanocytosis in CNS, 2) absence of cutaneous melanoma except in patients whose brain lesions are histologically benign, and 3) evidence of melanoma in meninges absent except where cutaneous nevi are histologically benign (19). Currently, NCM diagnosis is based on not only clinical presentation but also characteristics, such as brain MRI, histological examination of nevi, and cerebrospinal fluid (CSF) analysis are also taken into account. Some case reports have demonstrated that NCM patient’s CSF analysis may detect macrophages containing melanin (15). The presence of CNS lesions can be detected in T1 or T2 weighted MRI with gadolinium contrast that identifies diffuse thickening of leptomeninges and the presence of melanin. Commonly, neurological lesions are detected as diffuse pigmented deposits in either the leptomeninges or brain parenchyma, but in some cases of extensive spread, spinal cord infiltration is also possible (20, 21). Predominantly, three categories of cutaneous lesions associated with NCM have been observed – 1) benign, 2) proliferative nodules within benign nevi, and 3) malignancies arising within the benign CMN. These lesions are usually evident at birth or appear shortly thereafter, while brain lesions are detected later after the appearance of neurological symptoms in most cases (6, 9, 15, 22, 23). Due to the consistent association of CNS involvement with large/giant nevi in case reports, the current evaluation of the patients includes a recommendation for a gadolinium-enhanced MRI and a longitudinal neurological assessment, especially for L/GCMN patients younger than six months. Due to the rapid increase in the severity of the neurological symptoms observed in these patients, early diagnosis is important to apply palliative surgical interventions (24). Common clinical symptoms of NCM include headaches, nausea, vomiting, and seizures accompanied by hydrocephalus. Other conditions often included cranial nerve palsy and developmental delay (19, 25). Dandy-Walker malformation, a structural defect, was reported in 8-10% of the symptomatic patients. Occasionally, other forms of non-melanocytic tumors were also noted in association with NCM, such as rhabdomyosarcoma (26), meningioma (27), and lipoma (28). For NCM patients exhibiting hydrocephalus, attempts to reduce intracranial hypertension by placing a ventriculoperitoneal shunt (VPS) is effective management (also known as CSF shunting). Insertion of CSF shunt was reported in about 80% of the symptomatic cases (25, 26). However, this process is not without its risks, considering 70% of NCM patients died within three years of shunt insertion on account of peritoneal seeding, malignant transformation, and increased leptomeningeal spread. Peritoneal seeding frequently occurred with “metastatic” dissemination of the leptomeningeal tumor to the peritoneal surface via the inserted shunt (29-31). Lesions that underwent a malignant transformation into melanoma are commonly recommended for melanoma treatment with the existing paradigm that includes gross total resection by surgery, radiotherapy, chemotherapy, immunotherapy, etc. Often, surgical decisions are challenging and complicated by the actual anatomical location of the tumor and areas of the brain that are affected. Tumor recurrence post-resection can be monitored with periodic postoperative MRI surveillance. A recently published meta-analysis determined a 9% prevalence of leptomeningeal melanoma among all reported NCM cases (11). Patients with aggressive diseases have been treated with multiple modalities, such as surgery, radiation, chemotherapy, and immunotherapy without significant impact on progression-free survival (32, 33). Recently, Rades et al. reviewed the role of radiotherapy for meningeal melanocytomas, rare central nervous system tumors clinically similar to NCM, from published studies throughout a twenty-year period. This study investigated the treatment options by comparing different surgical and postoperative radiation therapy (RT) approaches. Analyzing data from 184 patients, the study found that gross total resection (GTR) led to better local control (LC) and overall survival (OS) than subtotal resection (STR). Specifically, GTR combined with RT yielded the best outcomes, while STR alone had the worst. RT following STR improved LC but did not affect OS, and for GTR cases, RT did not significantly enhance LC or OS. In summary, GTR is recommended for optimal outcomes, with STR+RT improving LC compared to STR alone. This finding could be of significant therapeutic importance to NCM patients drawing clinical experience from meningeal melanocytoma. To date, NCM is without a specific therapy, and clinical management of symptoms is the recommended course of action.
Molecular Mechanisms and Experimental Models
Experimental research involving drug discovery approaches has been largely impeded in NCM due to the scarcity of human samples and the lack of a faithful experimental model that recapitulates the disease. Historically, lack of interest from funding agencies also accounts for a dearth of support for basic, experimental research in NCM. Notwithstanding, the last decade saw an increase in NCM research initiatives, including the formation of a clinical registry of patients prospectively collecting information about the regular medical care of L/GCMN and NCM patients (ClinicalTrials.gov ID NCT04548817; Sponsor Memorial Sloan Kettering Cancer Center). Identification of a genetic driver of the disease facilitated the development of targeted therapies. Genetic mosaicism and clonal expansion of oncogenically mutant nevocytes have been proposed as the underlying cause of the formation of pigmented lesions, but the cell of origin in NCM remains elusive. It is still open for debate if the oncogenic mutation is acquired in developing melanocytes or even earlier in the developmental timeline at the undifferentiated neural crest cell stage (34). Considering that fully differentiated skin melanocytes in adults respond to oncogenic mutation (BRAF V600E or otherwise) by an oncogene-induced senescent (OIS) benign phenotype comprising of mostly pigmented growth-arrested cells, commonly identified as a small nevus or “mole”, congenital nevi are significantly larger, comprises of cell division capable cells displaying plasticity and, in some cases, proliferative nodules suggest a distinctly different etiology. Additionally, brain lesions observed in NCM patients in conjunction with their skin lesions carrying the same oncogenic mutation point to an embryonic origin, perhaps before the migration of embryonic melanocytes from the neural crest rather than a skin melanocytic origin. A plausible hypothesis is that oncogenic mutations acquired during the embryonic neural crest stage induce the proliferation of melanocytic progenitors. These progenitors, upon differentiating into pigment-producing melanocytes (or nevocytes), will likely form pigmented lesions. The embryonic environment, abundant in growth factors, may facilitate this proliferation by delaying the onset of oncogene-induced senescence, leading to the formation of larger, benign lesions at birth (Figure 1). However, currently, there is no experimental model available to test this hypothesis. Expanding further on this line of thought, undifferentiated nevocytes may persist in specific niches that support their undifferentiated state, thereby acting as a reservoir for mature nevocytes. This hypothesis may account for the slow growth of congenital nevi and NCM compared to acquired nevi (that is, growth-arrested) and aligns with experimental observations of nevocyte behavior both in vitro and in vivo.
Neural crest cells (NCCs) are transient embryonic cells unique to vertebrates with stem cell-like properties having the potential to give rise to neurons and mesenchymal progenitors, including components of the peripheral nervous system and craniofacial osteocytes other than melanocytes (35). The observation that very rarely, ensuant non-melanocytic lesions of neural crest derivatives are also detected in NCM, such as rhabdomyosarcoma, meningioma, and lipoma, lends credence to this possibility (26-28). Experimentally, forced expression of NRAS Q61K under the control of tyrosinase promoter (expression of a mutant oncogene is targeted to melanocytic lineage progenitors) in mice failed to produce brain lesions, although hyperpigmentation of the skin in this model histologically was reminiscent of human congenital nevi (36). Interestingly, the hepatocyte growth factor (HGF), when overexpressed in mice during development, produced both skin hyperpigmentation and pigmented lesions in the brain neonatally (37). HGF plays a crucial role in neural crest delamination and differentiation into several cell types, including melanocytes (38). HGF binding to its receptor MET activates a variety of intracellular signaling. HGF is known to enhance RAS signaling through the MAPK pathway culminating in cell proliferation, activate PI3K-Akt-mTOR signaling to support cell survival and induce motility through p21 activating kinase (PAK) and STAT3 (39). Increased bioavailability of HGF would likely amplify the MAP kinase pathway downstream of RAS, resulting in higher rates of cell proliferation (Figure 2). The HGF-Tg overexpression model, although produced benign leptomeningeal lesions resembling human NCM, this model did not address the role of mutant NRAS Q61K or BRAF V600E as the driver of the disease. It can be surmised that HGF-MET hyperactivity in the same cells affected by NRAS oncogenic activation would likely overamplify the RAS-MAP kinase pathway, leading to a more aggressively proliferating phenotype in addition to supporting cell survival and resisting senescence. Investigations into the possible role of HGF in suppression or delaying OIS are ongoing. Likely, HGF contributes indirectly to senescence prevention by reducing cellular stress and maintaining growth and survival. Takayama et al. reported higher levels of MET protein, a cellular receptor for HGF, in NCM lesions in a case report (40). Unfortunately, the presence of oncogenic mutations in NRAS and/or BRAF was not investigated in that case. Later case reports of NCM patients that detected NRAS Q61K/R or BRAF oncogenic mutations did not investigate the abundance of MET protein in lesional tissues either. Therefore, the molecular events underlying the origin of NCM are still unclear. A pertinent question to ask is if increased HGF activity/bioavailability and oncogenic activation of NRAS or BRAF act synergistically and whether such cooperation is necessary to produce NCM brain lesions. Considering the important role of HGF during fetal development and the possibility of maternal metabolic changes during pregnancy leading to fluctuations of HGF in the fetal microenvironment, this becomes a clinically relevant question (41, 42). Ackerman et al. reported the development of a transgenic mouse where forced expression of NRAS Q61K in developing melanocytes produced only skin hyperpigmentation due to melanocytosis, however, these animals failed to develop leptomeningeal melanocytosis, leaving this question open (36). It remains to be explored if HGF overactivity aids in the development of leptomeningeal lesions in Tyr-NRAS Q61K transgenic mice. On the other hand, Richard Marais’s group reported the development of primary leptomeningeal melanoma when oncogenic NRAS G12D is expressed in transgenic animals under the control of the same tyrosinase promoter (43). This also points to the diversity of outcomes regarding the oncogenicity of different NRAS hotspots in the context of developing melanocytes. Notwithstanding, the majority of NCM patients reported in the literature were positive for either NRAS Q61K or BRAF V600E and not NRAS G12D, the mutations being mutually exclusive (4, 44, 45). Thus, a transgenic mouse model harboring either NRAS Q61K/R or BRAF V600E with pigmented benign leptomeningeal lesions will likely be a faithful model of the disease provided histopathology is comparable with the human disease.
Following in these lines, Pawlikowski et al. reported a transgenic mouse model where Tyr-NRAS Q61K expression was coupled with aberrant Wnt signaling. In this model, brain lesions were produced in transgenic animals in addition to benign melanocytic proliferation in the skin (46, 47). This model remains the only animal model closely resembling NRAS-mutant NCM to date. However, the genetic defect resulting in aberrant Wnt signaling in this model has not been detected in human patients. Defects in any component of the Wnt signaling pathway have not been reported in human patients so far. Some of the polymorphisms in Wnt pathway genes detected by the authors in their cohort of patients can be expected to produce Wnt signaling aberrations based on computer modeling predictions, but actual functional data is still elusive in humans. This remains a serious deficiency of this animal model. Interestingly, this also highlights the role of molecular factors of the embryonic microenvironment in addition to oncogenic NRAS contributing to the development of NCM.
The development of faithful animal models of the disease must rely on the genetic data of human patients, and to that end, more high throughput sequencing and transcriptomic profiling of patients’ lesions are necessary. The information available currently suggests possible cooperation between a genetic mutation in NRAS or BRAF and additional factors present in the embryonic environment during development playing a role. It is not known for certainty what these factors are, but HGF seems to deserve a strong consideration. Therefore, a transgenic mouse model combining these aspects may produce a true recapitulation of NCM. Nonetheless, it is also imperative that the genetic changes created in the animal model remain relevant to human patients. That is yet to be achieved.
Drug Discovery and Targeted Therapies
To develop effective therapies targeting aberrant cells of NCM, it is important to identify the cells of origin and special features of NCM transcriptome to understand the vulnerabilities and drug susceptibilities of these cells. Recently, experimental studies were reported in models derived from patients’ cells in vitro and xenografts. Based on these reports, some new strategies are now being discussed, although their translatability will require rigorous trials.
The majority of NCM cases have NRAS Q61K as the driver mutation, although BRAF mutant NCM cases have also been reported (48). Oncogenic NRAS and BRAF both act through the MAP kinase signaling pathway, which is linked to the cellular proliferation machinery. NRAS being a GTPase, direct targeting of such a molecule has remained challenging for scientists and, in many instances, was deemed undruggable (49). NRAS, when mutated at the 61st residue to code for Lysine (K) instead of Glutamic acid (Q), becomes constitutively active, bound to GTP, and sequentially activates downstream effectors. RAF kinase (activated by NRAS) phosphorylates MEK (MAPK/ERK kinase), which subsequently activates ERK (extracellular signal-regulated kinase). Expression of cell proliferation, differentiation, and survival genes are influenced by ERK post-activation and upon its translocation to the nucleus (50, 51). Therefore, targeted inhibition of MAP kinase signaling intermediates was considered an attractive strategy to disrupt NRAS oncogenic signaling (52-54). There are ongoing clinical trials that use small molecule inhibitors of the MAP kinase pathway in several NRAS-driven cancers. There are only a few reports in the literature where such potential drugs have been experimentally tested in NCM. Trametinib, or MEK162, which inhibits MEK, a major kinase mediating signaling through the MAP kinase pathway, has been well studied in NRAS-driven cancers and has been approved by the FDA for conducting clinical trials of NRAS mutant melanoma. In preclinical studies, MEK162 was effective in regressing NCM-like CNS lesions in a transgenic mouse (47). However, oncogenic NRAS is also likely to activate redundant signaling parallelly through Akt and continue to function even when MAP kinase signaling is blocked. In line with this idea, MEK inhibition was found to be not as effective alone in a three-dimensional cellular model wherein activated Akt was shown to be functional in NRAS mutant NCM cells (55, 56). Additionally, trametinib may not be useful in NCM cases having wild-type NRAS and BRAF. Therefore, alternative strategies are needed to target cellular mechanisms that are common in most NCM cases. In a report by Ruan et al., 2014, cells isolated from an NRAS and BRAF wild-type NCM patient xenografted in immunocompromised mice showed sensitivity to an IGF receptor blocker, which was proposed as a druggable target in NCM (57). This molecule successfully reduced cell viability also in NRAS mutant NCM cells, which led to the idea that IGF is a common contributor to lesional growth in NCM irrespective of the mutation status (58). Since the IGF receptor predominantly signals through the PI3K – Akt signaling pathway, which is also known to be used by oncogenic NRAS as a redundant signaling alternative, this idea seems tenable. However, PI3K-Akt is also connected through a positive feedback loop to the mTOR signaling pathway that regulates cell survival. Therefore, treatment with Omipalisib, a dual inhibitor of PI3K-mTOR, abrogated cell survival and caused autophagic cell death in the anchorage-independent nevosphere model of NCM patients’ cells (56). These studies point to the importance of targeting multiple signaling intermediates in NCM cells as an effective therapeutic paradigm. Following in these lines, several experimental therapeutic regimens have been tested in the clinic. In most cases, biomarker analysis indicated target validation and initial response indicated tumor regression, but ultimately, the patient did not survive due to advanced disease and/or secondary complications. It would be premature to conclude the efficacy of these experimental treatments without appropriately designed clinical trials and statistically significant datasets. The rarity of the disease poses significant challenges to designing of clinical trials of NCM to test investigational drugs.
Experimental Treatments With Targeted Drugs and Clinical Outcomes
Conventional chemotherapy with Dacarbazine, Vincristine, and Cyclophosphamide has been tried unsuccessfully with NCM patients in the clinic before a driver mutation was identified in NCM and targeted therapy was proposed. Makin et al. reported 5 cases of NCM, including one with primary leptomeningeal melanoma spanning a period of 13 years (1984-1997), who were treated at two hospitals in Northern England. Several combinations of the above-mentioned drugs were used with or without Etoposide. The patients, all children, died from disease complications within 2-8 months of diagnosis without any notable improvement in the clinical presentation of the disease (59). These drugs are classified as broad-spectrum chemotherapeutics with cytotoxic effects on a variety of tissue cells, owing to their inhibitory effect on cellular processes such as replication of DNA and microtubule assembly, etc., during cell division. On the contrary, rational drug discovery approaches showed effective responses when oncogenic NRAS signaling was targeted based on mutation analysis of tumor biopsy. These included MEK162 (known as Trametinib), either alone or in combination with other drugs. Case reports are scarce but provide important insights into the roles of such treatments in regression and disease-free survival of the patients (Table I).
MEK162 has been reported to produce favorable results in clinical trials with cancers that involved dysregulated MAP kinase or ERK (60, 61), suggesting it could be tested as a therapeutic candidate for NRAS mutant NCM. A 13-year-old patient with NCM positive for somatic NRAS Q61K mutation was treated with MEK162 on a compassionate use basis after repeated surgery failed to prevent disease progression. The patient died five days after the therapy was started. Leptomeningeal tumor samples were studied for the effect of the drug, which showed a marked decrease in MIB1 and phosphorylated ERK (target of MEK162) proteins, suggesting a positive biomarker response (62). Important to note here is the histological finding that the lesion was comprised of cells ovoid and monotonous and exhibited low mitotic activity and no necrosis, which differs from typical melanoma histological features, although the lesion was extensive and covered significant areas of the brain and spinal cord.
Following this report, there was another report of four patients with NRAS-mutant brain melanoma that arose in conjunction with CMN and associated CNS abnormalities who received Trametinib as treatment. All these patients were diagnosed with diffuse leptomeningeal melanoma that harbored NRAS Q61K mutation and were symptomatic at the time of diagnosis. After they were started on Trametinib treatment, mild improvement was noted in all patients, and the symptom-free period varied from one month to 9 months; however, discontinuation of therapy resulted in rapid deterioration and death. The authors noted that the underlying cause of disease progression could be attributed to the presence of additional mutations in the melanoma tumor (63).
In 2018, Sharouf et al. described a case diagnosed with giant congenital melanocytic nevus, hydrocephalus, and leptomeningeal melanocytosis at five years of age. The patient was treated with Trametinib for seven months in addition to a ventriculoperitoneal shunt placement. The patient experienced symptomatic relief during this period, followed by detection of status epilepticus at 30 months of age. NRAS mutant melanoma was detected at 21 months of age from leptomeningeal and skin biopsies. At this stage, the patient’s condition started deteriorating rapidly and finally succumbed to death at 32 months of age (64). The authors did not present data on biomarker evaluation of drug response in this study. Although Trametinib was not as effective in controlling melanocytic tumor progression in the CNS context, interestingly, trametinib treatment of skin lesions of L/GCMN was found to be extremely effective in a patient whose lesional cells carried an AKAP9-BRAF fusion and, hence, should have activated the MAP kinase pathway (65).
Trametinib, when combined with other drugs, has shown improved treatment outcomes. In 2020, Vanood et al. reported NCM diagnosis in a 13-year-old patient with multiple congenital melanocytic nevi and clinical symptoms of seizure. The patient was found positive for NRAS Q61K mutation, and a diagnosis of NCM was made from MRI analysis. The patient was initially treated with Levetiracetam to manage seizures, the dosage of which was later increased and coupled with Trametinib. The authors report that the patient had no seizure episode reported at the 10-month follow-up, and at the one-year follow-up, no further clinical progression of neurological symptoms was noted (66). The authors describe the veracity of the disease as not so severe, offering hope that drug administration at the early onset of symptoms could offer a favorable prognosis.
Another interesting approach was followed by Hanft et al., 2022 to treat a patient with diffuse leptomeningeal melanosis and CNS melanoma. In this case, the patient was treated with azacitidine (DNA methyltransferase inhibitor) in combination with trametinib, and a durable positive clinical response was noted. The authors also showed that azacitidine acted synergistically with Trametinib in trametinib-resistant melanoma cells in vitro (67). More work needs to be done to understand the underlying molecular mechanism.
In a comprehensive analysis reported by Abele et al. on the treatment outcome of CNS melanoma patients, documented in the “German Registry for Rare Pediatric Tumors (STEP registry)”, aged 0-18 years, whose melanoma arose from NCM, nearly 100% fatality was noted. 85% of patients carried NRAS oncogenic mutation, and n=9 patients were treated with Trametinib. The authors noted short-term clinical improvement in some patients from treatment with Trametinib or Temozolomide, adding that the regimens failed to alter the overall survival rates. They infer that the overall survival rate of patients with CNS melanoma arising in NCM, even after targeted chemotherapy, was found to be worse in the pediatric group when compared to adults with primary CNS melanoma, whose one-year overall survival was around 50% (68).
These studies indicate that Trametinib, despite its targeted inhibition of the MAP kinase pathway, may not be sufficient as a monotherapy to produce an effective, durable clinical response in NCM in the context of CNS lesions. NCM, with all its similarities with other melanocytic malignancies, is also different in its response to chemotherapy targeting oncogenic NRAS signaling alone. In conclusion, more research needs to be done to understand the cellular and molecular characteristics of NRAS/BRAF mutant NCM, identify drug susceptibilities of mutant NCM cells, delineate the origin and progression of the disease and the role of microenvironment in it towards developing a specific therapeutic regimen for NCM.
Future Directions
Future research investigations into the therapeutic landscape of NCM would benefit from a multidisciplinary approach drawing from advances in genetics, molecular biology, pharmacology, and improved patient care strategies. Several key areas of investigation could be:
Targeted therapies. Investigations aimed at understanding the molecular etiology of NCM need to be undertaken to thoroughly understand the drug susceptibilities of NCM cells. High throughput sequencing studies at the level of whole genome and/or transcriptome would reveal new insights into specific mutations and associations between other genetic and/or non-genetic factors contributing to altered cell signaling, which in turn could contribute to the development and progression of the disease. With significant technological advances in the field of transcriptomics, gene expression patterns can now be analyzed at single-cell resolution, allowing for a deeper analysis of genes, gene networks, and cellular pathways underlying a disease phenotype. This approach could lead to the identification of druggable targets within NCM cells and/or the tumor microenvironment that more precisely addresses the underlying pathology.
Novel drug combination therapies. New drug development approaches may be followed that target multiple cellular pathways in addition to the major driver mutation(s) to provide for a more robust and durable response. Some of the probable targets could be epigenetic modifiers [such as azacytidine (67) or Vorinostat (69)], drugs targeting growth factor receptors (such as IGF1 receptor blocker or MET inhibitors), drugs targeting cell survival mechanisms (such as mToR inhibitors) in addition to targeting the oncogenic signaling through the MAP kinase pathway. Combination therapies may also be tested for more effective management options for patients depending on the severity of disease progression and clinical symptoms.
Gene therapy. This approach involves gene editing technology to correct or compensate for a defective gene that is linked to the development of disease (in this case, the mutant oncogene that drives NCM cell proliferation). Although the technology is still in its early stages of development, it represents a promising new strategy to be tested in NCM since the underlying pathogenic driver of NCM is genetic mutations in NRAS or BRAF.
Immunotherapy. Although published reports using PD1 blockers and interferons did not improve overall survival in NCM, and immunotherapy using monoclonal antibodies could be challenging considering their inability to effectively cross the blood-brain barrier (70), more studies need to be undertaken to investigate the overall role of the immune system in NCM. Inflammation and pruritus have been observed in patients with L/GCMN and cutaneous lesions of NCM patients. It remains to be explored if targeting inflammatory pathways produces any beneficial outcome in the context of CNS lesions.
Developing a relevant animal model of NCM. Efficient animal models are needed to study in vivo drug efficacies observed in vitro for translating those findings into effective treatments in the clinic. Previous studies by our group and others have suggested that NRAS Q61K mutant cells isolated from L/GCMN and NCM lesions may be responsive to dual PI3K-mTOR inhibitors owing to the redundant signaling of mutant NRAS through the MAP kinase and Akt pathways in vitro (56, 71, 72). A faithful animal model of NCM will facilitate experimental approaches testing gene therapy, and immunotherapy approaches in addition to targeted and combinatorial chemotherapies in vivo. Combination therapies aimed at alleviating neurological symptoms may also be tested in animal models that can recapitulate them along with targeted inhibition of the mutagenic signaling.
Advanced imaging techniques. With more and more advancements in the field of imaging technology, the ability to monitor and evaluate the progression of NCM will be improved. This will not only empower diagnostic capabilities but also will enable more specific and timely interventions.
Patient registries and data collection. Considering the rarity of the disease, data on the patient’s disease pathology, clinical outcomes concerning treatment responses, and overall clinical and molecular parameters are extremely valuable to devise future directions of research. Increased interaction and partnerships between clinicians, researchers, and patient advocacy groups are vital to developing collaborations facilitating such collection of data.
Clinical trials. Participation of patients in ongoing and future clinical trials is key to the development of novel therapies and the improvement of current therapies. Being a rare disease, conducting clinical trials for NCM could be a challenging task in terms of patient enrolment and participation. Currently, NCM patients participate in trials designed for other closely related diseases, such as primary CNS melanoma and/or other cancers of CNS.
Conclusion
The last decade saw great advances in experimental approaches to assess therapeutic capabilities toward the treatment of NCM. Considering the rarity of the disease and the slow advancement of research in the field, so far, only few therapeutic strategies have been tested in the clinic. These included targeted chemotherapeutic Trametinib as monotherapy or in combination with other drugs. More work needs to be done to understand the molecular underpinnings of the disease and to examine the combinatorial approach towards designing a specific therapy for NCM that improves the progression-free survival of patients.
Footnotes
Conflicts of Interest
The Author declares that there are no conflicts of interest related to the publication of this work. The research and findings presented in this manuscript were conducted unbiasedly, and no financial or personal relationships have influenced the content of this article.
- Received September 24, 2024.
- Revision received October 25, 2024.
- Accepted November 7, 2024.
- Copyright © 2024 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.
This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).