Melanoma in 2025: Navigating Evolving Landscapes in Epidemiology, Therapeutic Innovation, and Persistent Challenges
Melanoma in 2025: Key Research Insights
Interactive visualization of epidemiology, treatments, and challenges
Key Statistics
New Cases (2025)
104,960
Projected new cases of invasive melanoma in the US
Gender Distribution
60,550
Male cases (58%)
44,410
Female cases (42%)
Mortality (2025)
8,430
Projected deaths from melanoma in the US
5,470 Male deaths (65%)
2,960 Female deaths (35%)
5-Year Survival by Race
White
94%
Black
71%
Significant racial disparities in diagnosis and survival outcomes
Melanoma Subtypes
Cutaneous Melanoma
Prevalence: Most common type (>90%)
Risk Factor: UV exposure
Common Mutations: BRAF (40-50%), NRAS (15-20%), NF1 (10-15%)
Response to Therapy: Better response to immunotherapy and targeted therapy
Acral Lentiginous Melanoma
Location: Palms, soles, under nails
Demographics: Most common in darker skin tones
Risk Factor: Not associated with UV exposure
Common Mutations: KIT mutations more common
Mucosal Melanoma
Location: Respiratory, GI, genitourinary tracts
Prevalence: Rare, ~1% of melanomas
Risk Factor: Not linked to UV exposure
Prognosis: Often diagnosed late, poor outcomes
Uveal (Ocular) Melanoma
Location: Eye (iris, ciliary body, choroid)
Common Mutations: GNAQ/GNA11
Metastatic Pattern: Predominantly liver
Research Priority: CDMRP FY25 Focus Area
Treatment Advances (2025)
Immune Checkpoint Inhibitors
Unleash the patient's own immune system to fight cancer cells
Anti-PD-1 Monotherapy
Nivolumab + Ipilimumab
Nivolumab + Relatlimab
Targeted Therapy
For BRAF V600 mutant melanoma (40-50% of cases)
BRAF inhibitor alone
BRAF + MEK inhibitor
High initial response rates but resistance often develops
Novel Approaches
Emerging therapies showing promise
Oncolytic Virus (T-VEC)
RP1 + Nivolumab
TIL Therapy (Lifileucel)
Unmet Needs & Research Priorities
Treatment Resistance
Both primary (lack of initial response) and acquired resistance (relapse after initial response) limit the long-term efficacy of immunotherapies and targeted therapies. Research is focusing on identifying biomarkers of response/resistance and developing rational combination therapies.
Brain Metastases
Melanoma has a high propensity to metastasize to the brain. The ABC trial provided 7-year follow-up data showing upfront ipilimumab + nivolumab achieved a 51% overall survival rate in melanoma patients with untreated brain metastases, compared to 29% with nivolumab alone and historical survival of ~16 weeks.
Racial Disparities
While incidence is much lower in individuals of Black and Hispanic descent, survival rates are markedly worse (5-year survival 71% for Black vs. 94% for White individuals). Contributing factors include later stage at diagnosis, higher prevalence of rare subtypes, and potential biological differences. Addressing these disparities requires culturally competent education, improved provider training, and equitable access to care.
Rare Melanomas
Acral, mucosal, and uveal melanomas present distinct challenges due to their different biology, often delayed diagnosis, and relative lack of response to therapies proven effective in cutaneous melanoma. The CDMRP FY25 Focused Program Award specifically supports research addressing critical unmet needs in these rare subtypes.
Melanoma in 2025: Navigating Evolving Landscapes in Epidemiology, Therapeutic Innovation, and Persistent Challenges
Abstract
Melanoma, the most aggressive form of skin cancer, continues to pose a significant public health challenge in the United States and globally. Projections for 2025 indicate sustained high incidence rates, particularly among specific demographics, underscoring the need for ongoing vigilance in prevention and early detection. Concurrently, the therapeutic landscape for melanoma has undergone a profound transformation over the past decade, driven primarily by the advent of immune checkpoint inhibitors and targeted therapies. These advancements have dramatically improved survival outcomes, particularly for patients with advanced-stage disease, shifting the paradigm from palliation towards potential long-term control and even cure in some subsets. However, significant challenges persist, including the management of treatment resistance, addressing brain metastases, improving outcomes for rare melanoma subtypes, and eliminating stark racial disparities in diagnosis and survival. Furthermore, the long-term physical and psychosocial sequelae for a growing population of survivors require dedicated research and clinical attention. This paper synthesizes current epidemiological data, reviews the state-of-the-art in melanoma diagnostics and therapeutics, explores mechanisms of resistance and disease progression, highlights critical unmet needs including disparities and rare melanomas, discusses evolving research priorities and funding initiatives for 2025, and outlines the path forward in prevention, early detection, and survivorship care. The integration of multi-omics data, novel therapeutic combinations, advanced preclinical models, and patient-centered research approaches will be crucial in continuing the progress against this complex malignancy.
Keywords: Melanoma, Epidemiology, Incidence, Survival Rates, Immunotherapy, Targeted Therapy, Oncolytic Virus, Treatment Resistance, Brain Metastasis, Rare Melanoma, Health Disparities, Survivorship, CDMRP, Melanoma Research Program, Prevention, Early Detection.
Introduction
Melanoma originates from melanocytes, the pigment-producing cells primarily located in the basal layer of the epidermis. While representing a minority of skin cancer diagnoses, melanoma accounts for the vast majority of skin cancer-related mortality due to its propensity for early metastasis (American Cancer Society, 2024). The American Cancer Society's (ACS) annual Cancer Facts and Figures report serves as a critical resource for tracking melanoma trends, providing estimates of incidence, prevalence, mortality, and survival statistics, which inform public health strategies, clinical practice, and research priorities (ACS, 2025 Projections, as cited in provided text).
The narrative of melanoma has shifted dramatically over the past 15 years. Once associated with dismal prognoses in its advanced stages, the advent of molecularly targeted therapies and, more significantly, immune checkpoint inhibitors (ICIs) has revolutionized treatment, leading to unprecedented improvements in survival (Robert et al., 2019; Hodi et al., 2018). However, this progress is not universal. Significant hurdles remain, including innate and acquired resistance to therapy, the formidable challenge of central nervous system (CNS) metastases, the lack of effective treatments for certain rare melanoma subtypes, and persistent, unacceptable disparities in outcomes based on race and ethnicity.
As we look towards 2025, understanding the current epidemiological landscape, the nuances of modern therapeutic strategies, the frontiers of ongoing research, and the specific focus areas of major funding bodies like the Department of Defense Congressionally Directed Medical Research Programs (CDMRP) Melanoma Research Program (MRP) is essential. This paper aims to provide a comprehensive overview of melanoma in 2025, synthesizing data on incidence and survival, detailing advancements in treatment, exploring key research frontiers, and discussing strategies to address ongoing challenges, including prevention and disparities in care.
Epidemiology and Incidence Trends: A Shifting Landscape
2.1. Projected Incidence and Mortality for 2025 The ACS projects approximately 104,960 new cases of invasive melanoma to be diagnosed in the United States in 2025. This represents a slight increase from the previous year and underscores melanoma's position as a significant cancer diagnosis, ranking as the fifth most common cancer overall (ACS, 2025 Projections, as cited in provided text).
Gender Disparity: A notable gender disparity persists, with higher incidence projected in men (60,550 cases) compared to women (44,410 cases).
Mortality: An estimated 8,430 deaths from melanoma are projected for 2025 (5,470 men, 2,960 women). While mortality rates have shown a decline (approximately 1% per year from 2017-2021), attributed largely to treatment advances, the absolute number of deaths remains substantial (ACS, 2025 Projections, as cited in provided text).
Historical and Age-Related Trends
Historically, melanoma incidence rose steeply in the US from the 1970s onwards, likely reflecting changes in sun exposure behaviors, increased detection, or a combination thereof (SEER Cancer Stat Facts, n.d.). Recent trends show some divergence based on age and sex:
Younger Adults (<50 years): Incidence rates have stabilized or slightly declined (~1% per year) in men under 50, and stabilized in women under 50 (ACS, 2025 Projections, as cited in provided text). Despite this stabilization, melanoma remains one of the most common cancers diagnosed in individuals younger than 30, particularly young women (ACS, 2025 Projections, as cited in provided text; Ward et al., 2021). This observation coincides with a broader trend where overall cancer incidence rates in women under 50 have begun to surpass those in men of the same age group (ACS, 2025 Projections, as cited in provided text).
Older Adults (≥50 years): Rates continue to increase by nearly 3% per year in women aged 50 and older, while they appear to have stabilized in men of the same age group (ACS, 2025 Projections, as cited in provided text). The reasons for the continued rise in older women warrant further investigation, potentially involving cumulative UV exposure, hormonal factors, or detection artifacts.
Geographic and Global Variations Melanoma incidence varies significantly worldwide, with the highest rates observed in Australia and New Zealand, followed by North America and Northern Europe (Arnold et al., 2022). Within the US, incidence rates tend to be higher in states with greater ambient UV radiation and predominantly fair-skinned populations. These geographical patterns strongly reinforce the role of UV exposure as the primary environmental risk factor.
Etiology and Risk Factors
Understanding the factors driving melanoma development is crucial for prevention and identifying high-risk individuals.
Ultraviolet (UV) Radiation Exposure UV radiation, primarily from sun exposure and artificial tanning beds, is the most significant environmental risk factor for melanoma. Experts estimate that approximately 90% of melanoma diagnoses are attributable to UV exposure (ACS, 2025 Projections, as cited in provided text; Parkin et al., 2011).
Mechanism: UV radiation (both UVA and UVB) induces DNA damage in melanocytes, including characteristic C>T transitions known as UV signature mutations. While cells possess DNA repair mechanisms, excessive or unrepaired damage can lead to oncogenic mutations (Rastrelli et al., 2014).
Exposure Patterns: Intermittent, intense sun exposure (e.g., blistering sunburns, especially in childhood and adolescence) appears particularly strongly linked to melanoma risk, more so than chronic, cumulative exposure, although both contribute (Dennis et al., 2008).
Tanning Beds: Artificial UV radiation from tanning beds significantly increases melanoma risk, particularly with early-age initiation (Colantonio et al., 2014). This has led to regulations and public health campaigns discouraging their use.
Phenotypic Risk Factors Individual susceptibility is heavily influenced by pigmentary characteristics:
Fair skin (easily burns, rarely tans)
Blonde or red hair
Blue or green eyes
Presence of numerous or atypical moles (nevi)
Genetic Predisposition While most melanomas are sporadic, a significant minority (around 10%) occur in a familial context (Read et al., 2016).
High-Penetrance Genes: Germline mutations in genes like CDKN2A (encoding p16INK4a and p14ARF) are the most common cause of hereditary melanoma, significantly increasing lifetime risk (Soura et al., 2016). Mutations in CDK4 are rarer causes.
Moderate-Penetrance Genes: Variants in genes like MC1R (influencing pigmentation), MITF, BAP1, POT1, and others contribute to melanoma risk, often interacting with UV exposure (Read et al., 2016; Law et al., 2020).
Ongoing research, including initiatives supported by the CDMRP MRP (Focus Area: Identify and understand risk factor determinants and biomarkers), aims to further elucidate the complex interplay of genetic and environmental factors.
Other Factors
Personal History: A previous melanoma diagnosis significantly increases the risk of developing subsequent melanomas.
Immunosuppression: Individuals with compromised immune systems (e.g., organ transplant recipients, HIV/AIDS patients, those on chronic immunosuppressive therapy) have an elevated risk (Asgari et al., 2012).
Pathophysiology and Molecular Landscape
Melanoma development is a multi-step process involving the accumulation of genetic and epigenetic alterations that drive malignant transformation and progression.
Melanoma Subtypes Melanoma is heterogeneous, encompassing distinct clinical and molecular subtypes:
Cutaneous Melanoma: The most common type, arising on the skin. Further classified based on histopathology (superficial spreading, nodular, lentigo maligna, acral lentiginous). Most are linked to UV exposure, except acral melanoma.
Acral Lentiginous Melanoma: Occurs on the palms, soles, and under the nails. It is the most common subtype in people with darker skin tones and is generally not associated with UV exposure (Bellew et al., 2023). Research into its distinct biology and risk factors is a priority (CDMRP FY25 Focus Area, MRA RARE Registry).
Mucosal Melanoma: Arises from melanocytes in mucosal membranes (e.g., respiratory, gastrointestinal, genitourinary tracts). It is rare, aggressive, and not linked to UV. Diagnosis is often delayed, leading to poor prognosis (Carvajal et al., 2018). This is another key area for focused research (CDMRP Focused Program Award - Rare Melanomas).
Uveal (Ocular) Melanoma: Originates in the eye's uveal tract (iris, ciliary body, choroid). It has a distinct molecular profile (often GNAQ/GNA11 mutations) and metastatic pattern (predominantly liver) compared to cutaneous melanoma (Krantz et al., 2017). Research into rare melanomas like uveal is explicitly encouraged (CDMRP FY25 Focus Area).
4.2. Key Molecular Drivers Specific gene mutations are critical drivers of melanoma development and represent key therapeutic targets:
MAPK Pathway: The Mitogen-Activated Protein Kinase (MAPK) pathway is constitutively activated in the majority of cutaneous melanomas.
BRAF Mutations: Present in ~40-50% of cutaneous melanomas, most commonly the V600E substitution. This leads to uncontrolled cell proliferation (Davies et al., 2002).
NRAS Mutations: Occur in ~15-20%, typically mutually exclusive with BRAF mutations. They also activate the MAPK pathway and other signaling cascades (e.g., PI3K/AKT) (Fedorenko et al., 2015).
NF1 Mutations: Loss-of-function mutations in this tumor suppressor gene occur in ~10-15%, often in older individuals or those with chronic sun damage (Krauthammer et al., 2015).
KIT Mutations: Found more frequently in acral and mucosal melanomas, and those arising in chronically sun-damaged skin (Curtin et al., 2006).
Other Pathways: Alterations in the PI3K/AKT/mTOR pathway, cell cycle regulators (e.g., CDKN2A loss), and telomere maintenance mechanisms (e.g., TERT promoter mutations) also play crucial roles (Shain & Bastian, 2016).
4.3. The Tumor Microenvironment (TME) The TME, comprising immune cells, fibroblasts, endothelial cells, and extracellular matrix, plays a critical role in melanoma progression, immune evasion, and response to therapy (Binnewies et al., 2018). Understanding the interplay between tumor cells and the TME, including the local microbiome, is a key research focus (CDMRP FY25 Focus Area).
Advances in Diagnosis and Staging
Early detection remains the cornerstone of improving melanoma outcomes, as localized disease is highly curable.
5.1. Clinical Examination and Dermoscopy Visual inspection by trained clinicians, often aided by dermoscopy (using a dermatoscope to visualize subsurface skin structures), improves diagnostic accuracy for pigmented lesions, helping differentiate benign nevi from melanoma (Vestergaard et al., 2016). Public awareness campaigns emphasize the "ABCDE" criteria (Asymmetry, Border irregularity, Color variation, Diameter >6mm, Evolving) for self-monitoring.
5.2. Biopsy and Pathological Assessment Suspicious lesions require biopsy (typically excisional) for definitive histopathological diagnosis. Pathological reports include crucial prognostic information like Breslow thickness (tumor depth), ulceration status, mitotic rate, and margin status.
5.3. Imaging and Staging The American Joint Committee on Cancer (AJCC) staging system (currently the 8th edition) integrates tumor (T), node (N), and metastasis (M) characteristics, along with factors like ulceration and LDH levels, to classify melanoma stage (I-IV) and predict prognosis (Gershenwald et al., 2017). Imaging modalities (CT, PET/CT, MRI) are used for staging advanced disease and monitoring treatment response.
5.4. Emerging Diagnostic Tools Research is actively exploring novel diagnostic aids:
Artificial Intelligence (AI): AI algorithms are being developed to analyze clinical and dermoscopic images, potentially assisting clinicians in diagnosis (Esteva et al., 2017).
Liquid Biopsies: Analysis of circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), or exosomes in blood holds promise for early detection, monitoring treatment response, detecting minimal residual disease, and identifying resistance mutations (Schadendorf et al., 2017). Developing new tools for detection, diagnosis, and monitoring is a specific CDMRP FY25 focus area.
Therapeutic Revolution: Current Treatment Paradigms (2025)
Treatment strategies are tailored based on melanoma stage, molecular characteristics (especially BRAF status), patient factors, and prior therapies.
6.1. Localized Melanoma (Stages 0-II)
Surgery: Wide local excision is the primary treatment. Sentinel lymph node biopsy (SLNB) is considered for staging in melanomas ≥0.8mm thick or with other high-risk features.
Adjuvant Therapy: For patients with high-risk Stage IIB/C or Stage III melanoma (resected lymph node involvement), adjuvant therapy significantly reduces recurrence risk. Options include:
Anti-PD-1 Immunotherapy: Nivolumab or Pembrolizumab (Weber et al., 2017; Eggermont et al., 2018).
6.2. Advanced/Metastatic Melanoma (Unresectable Stage III and Stage IV) The treatment landscape for advanced melanoma has been revolutionized.
Immune Checkpoint Inhibitors (ICIs): These agents unleash the patient's own immune system to fight cancer.
Anti-PD-1 Monotherapy: Nivolumab (Opdivo®) and Pembrolizumab (Keytruda®) are standard first-line options, providing durable responses in a subset of patients (Robert et al., 2019; Schachter et al., 2017).
Anti-CTLA-4 Monotherapy: Ipilimumab (Yervoy®) was the first ICI approved but is now primarily used in combination due to lower efficacy and higher toxicity compared to anti-PD-1 (Hodi et al., 2010).
Combination ICIs:
Nivolumab + Ipilimumab: Offers higher response rates and improved overall survival compared to monotherapy, particularly in patients with challenging features (e.g., brain metastases, high LDH), but at the cost of increased immune-related adverse events (irAEs) (Larkin et al., 2019; Tawbi et al., 2018; Long et al., 2025 - ABC trial data).
Nivolumab + Relatlimab (Opdualag®): A fixed-dose combination targeting PD-1 and LAG-3 (Lymphocyte-activation gene 3), another inhibitory checkpoint. Approved as a first-line option, offering improved progression-free survival over nivolumab alone with a manageable safety profile (Tawbi et al., 2022).
Targeted Therapy (for BRAF V600 mutant melanoma): Combination therapy targeting both BRAF and MEK kinases is standard.
Dabrafenib (Tafinlar®) + Trametinib (Mekinist®)
Encorafenib (Braftovi®) + Binimetinib (Mektovi®)
Vemurafenib (Zelboraf®) + Cobimetinib (Cotellic®) These combinations yield high initial response rates and improve survival compared to BRAF inhibitor monotherapy, but resistance often develops (Robert et al., 2015; Long et al., 2017; Dummer et al., 2018). The choice between first-line ICI or targeted therapy for BRAF-mutant patients depends on disease characteristics, pace of progression, and patient factors.
Oncolytic Virus Therapy:
Talimogene Laherparepvec (T-VEC, Imlygic®): An engineered herpes simplex virus-1 (HSV-1) injected directly into tumors. Approved for unresectable melanoma recurrent after initial surgery (Andtbacka et al., 2015). It lyses tumor cells and stimulates an anti-tumor immune response.
RP1 (Vusolimogene Oderparepvec): An investigational enhanced-potency HSV-1 expressing GM-CSF and a fusogenic protein (GALV-GP R-). Currently in Phase 3 trials (IGNYTE-3, NCT03767348) combined with nivolumab for patients who have progressed on prior anti-PD-1 therapy (Hamid & Bowles, as cited in provided text). Phase 1/2 data showed promising response rates (~30-36%), including in uninjected lesions (abscopal effect), and manageable toxicity (Hamid et al., ESMO 2024, as cited in provided text). Notably, RP1 can be injected into visceral metastases (liver, lung), a potential advantage over T-VEC. [External Link: IGNYTE-3 Clinical Trial Information - https://clinicaltrials.gov/study/NCT03767348]
Adoptive Cell Therapy (ACT):
Tumor-Infiltrating Lymphocyte (TIL) Therapy: Involves harvesting lymphocytes from a patient's tumor, expanding them ex vivo, and re-infusing them along with high-dose interleukin-2 (IL-2). Lifileucel (Amtagvi™) received FDA accelerated approval in February 2024 for patients with advanced melanoma previously treated with anti-PD-1 therapy, and if BRAF V600 positive, a BRAF inhibitor (with or without MEK inhibitor) (Sarnaik et al., 2021; FDA, 2024). This provides a novel option for heavily pre-treated patients but involves a complex manufacturing process and potential for significant toxicity.
6.3. Neoadjuvant Therapy Administering systemic therapy before surgery for high-risk, resectable Stage III melanoma is a rapidly evolving area. Neoadjuvant ICIs (especially combination ipilimumab + nivolumab) or targeted therapy (for BRAF mutants) can achieve high pathologic response rates, potentially improving relapse-free and overall survival compared to upfront surgery followed by adjuvant therapy (Amaria et al., 2018; Blank et al., 2018). Several ongoing trials are defining the optimal regimens and patient selection criteria. Understanding response and resistance mechanisms in this setting is a research priority (CDMRP FY25 Focus Area).
Addressing Unmet Needs and Future Directions
Despite significant progress, major challenges require focused research efforts, many reflected in the priorities of funding bodies like the CDMRP MRP and Melanoma Research Alliance (MRA).
7.1. Treatment Resistance Both primary (lack of initial response) and acquired (relapse after initial response) resistance limit the long-term efficacy of ICIs and targeted therapies.
Mechanisms: Resistance is complex and multifactorial, involving tumor-intrinsic factors (e.g., loss of antigen presentation machinery like B2M/HLA, activation of alternative signaling pathways like PI3K/AKT, lineage plasticity) and TME-mediated factors (e.g., accumulation of immunosuppressive cells like regulatory T cells or myeloid-derived suppressor cells, exclusion of cytotoxic T cells) (Sharma et al., 2017; Hugo et al., 2016).
Strategies: Research focuses on identifying predictive biomarkers of response/resistance, developing rational combination therapies (e.g., ICIs + targeted therapy, ICIs + other immunomodulators, ICIs + epigenetic modifiers), and understanding tumor dormancy and recurrence mechanisms (CDMRP FY25 Focus Area).
7.2. Brain Metastases Melanoma has a high propensity to metastasize to the brain, historically associated with a very poor prognosis.
Challenges: The blood-brain barrier (BBB) limits drug penetration, and the unique brain TME may foster immune evasion.
Progress: Combination ICIs (ipilimumab + nivolumab) have shown significant intracranial activity (Tawbi et al., 2018). The ABC clinical trial provided landmark 7-year follow-up data showing that upfront ipilimumab + nivolumab achieved a 51% overall survival rate in melanoma patients with untreated brain metastases, compared to 29% with nivolumab alone, suggesting potential cure for a subset (Long et al., 2025, Lancet Oncology). This contrasts sharply with historical survival of ~16 weeks. [External Link: ABC Trial Publication - https://www.thelancet.com/journals/lanonc/article/PIIS1470-2045(24)00701-1/fulltext]
Future Directions: The ABC-X trial is now exploring upfront stereotactic radiosurgery (SRS) combined with ipilimumab + nivolumab. Further research into mechanisms of CNS metastasis and overcoming the BBB remains critical (MRA Priority Area). Understanding leptomeningeal disease (LMD), a particularly devastating form of CNS metastasis, is also a priority (MRA Priority Area).
7.3. Rare Melanomas Acral, mucosal, and uveal melanomas present distinct challenges due to their different biology, often delayed diagnosis, and relative lack of response to therapies proven effective in cutaneous melanoma.
Research Needs: Dedicated research into their unique molecular drivers, risk factors, and TME is essential. Development of specific preclinical models and targeted therapies is crucial (CDMRP FY25 Focus Area). Patient registries like the MRA RARE Registry are vital for collecting data and facilitating research and clinical trial matching for these patients. [External Link: MRA RARE Registry - https://www.curemelanoma.org/patient-caregiver/rare-melanoma/rare-registry/] The CDMRP FY25 Focused Program Award - Rare Melanomas specifically supports large, synergistic programs addressing critical unmet needs in this area.
7.4. Disparities in Melanoma Outcomes Significant racial and ethnic disparities persist in melanoma.
The Paradox: While incidence is much lower in individuals of Black (lifetime risk ~0.1%) and Hispanic (~0.5%) descent compared to White individuals (~3%), survival rates are markedly worse (5-year survival 71% for Black vs. 94% for White individuals, 2013-2019 data) (ACS, 2025 Projections, as cited in provided text; SEER Data).
Contributing Factors:
Later Stage at Diagnosis: Melanomas in People of Color are often diagnosed at more advanced stages, significantly impacting prognosis (Bellew et al., 2023). This may result from lower perceived risk (leading to delays in seeking care), lower public/provider awareness of melanoma in non-sun-exposed sites (where acral/mucosal types common in these populations occur), potential implicit bias, and socioeconomic barriers to accessing timely dermatologic care (provided text; Higgins et al., 2021).
Subtype Differences: The higher prevalence of acral and mucosal melanomas, which have inherently poorer prognoses, contributes.
Potential Biological Differences: Research is ongoing to determine if underlying biological differences contribute to aggressiveness or treatment response variation.
Addressing Disparities: Solutions require a multi-pronged approach: culturally competent patient education and awareness campaigns emphasizing risk (even if lower overall) and signs/symptoms (including in non-sun-exposed areas), improved provider training on melanoma presentation in diverse skin types and rare subtypes, ensuring equitable access to dermatologic care and advanced treatments, and inclusion of diverse populations in clinical trials (MRA Priority Area).
7.5. Survivorship With improving survival rates, the population of melanoma survivors is growing, facing long-term physical and psychosocial challenges.
Physical Sequelae: Chronic fatigue, lymphedema (after surgery), peripheral neuropathy, endocrine dysfunction (from ICIs), dermatologic toxicities, reproductive/sexual health issues (ACS, 2025 Projections, as cited in provided text).
Psychosocial Impacts: Fear of recurrence, anxiety, depression, financial toxicity, body image concerns, impact on family/caregivers (ACS, 2025 Projections, as cited in provided text).
Research Needs: Studies are needed to better understand and manage long-term treatment toxicities, the role of lifestyle factors (diet, exercise) on outcomes and quality of life, and effective interventions for psychosocial support (CDMRP FY25 Focus Area; CDMRP FY25 Survivorship Research Award). The inclusion of patient advocates in research planning is encouraged to ensure relevance (CDMRP FY25 Award Requirements).
The Research Ecosystem: Funding and Priorities for 2025
Continued progress relies heavily on robust research funding and strategic prioritization. The CDMRP MRP is a key player, leveraging Department of Defense appropriations to fund innovative, high-impact melanoma research with relevance to military health.
8.1. CDMRP Melanoma Research Program (MRP) FY25 Priorities The FY25 MRP funding opportunities reflect the critical needs in the field. Key focus areas for several award mechanisms (Idea, Melanoma Academy Scholar, Team Science) include:
Risk Factors & Biomarkers: Identifying determinants of risk and biomarkers for detection/prognosis.
Detection/Diagnosis Tools: Developing novel technologies (e.g., imaging, biomarkers) to improve early and accurate diagnosis and monitoring, distinguishing high-risk from low-risk lesions/individuals.
Disease Mechanisms: Defining mechanisms of initiation, progression, treatment response/resistance, recurrence, dormancy, and metastasis (including TME/microbiome roles).
Preclinical Models: Creating models (cutaneous and rare subtypes) that better recapitulate human disease evolution.
Rare Melanomas: Addressing unmet needs across the research spectrum (biology, prevention, diagnosis, treatment, survivorship).
Survivorship: Investigating psychosocial impacts, symptom management, treatment side effects (including late effects), and lifestyle factors.
8.2. CDMRP FY25 Award Mechanisms (Highlights): The program offers diverse funding mechanisms targeting different career stages and research scopes:
Focused Program Award - Rare Melanomas (NEW): Supports large, multi-project, synergistic programs addressing overarching questions in rare melanomas ($2M direct costs, 4 years). Requires pre-proposal.
Survivorship Research Award: Funds preclinical or clinical studies on quality of life, toxicity management, psychosocial support, etc. ($725K direct costs, 3 years). Requires patient advocate involvement.
Melanoma Academy Awards (Leadership & Scholar): Supports an interactive network for mentoring and developing early-career investigators ($1.3M for Leadership, $550K for Scholar).
Idea Award (Returning): Funds innovative, high-risk/high-gain ideas, discouraging preliminary data ($400K direct costs, 2 years). Requires pre-proposal.
Team Science Award (NEW): Supports multidisciplinary partnerships tackling complex problems unachievable by single investigators ($1.5M direct costs, 3 years). Requires pre-proposal.
Military Relevance: A cross-cutting theme encouraging research relevant to Service Members, Veterans, and beneficiaries (e.g., using military populations/data, collaborations with DOD/VA).
8.3. Other Key Organizations The Melanoma Research Alliance (MRA) is the largest non-profit funder of melanoma research globally, supporting a broad portfolio from basic science to clinical trials, with priorities including early detection, treatment resistance, brain metastases, and rare melanomas. The National Cancer Institute (NCI) remains the largest single funder of cancer research overall. Collaboration between these entities, academia, industry, and patient advocacy groups is vital.
Prevention and Early Detection Strategies
Given that most melanomas are linked to UV exposure and early-stage disease is highly curable, prevention and early detection are paramount public health imperatives.
Use broad-spectrum sunscreen (SPF 30+) generously and reapply frequently.
Avoid tanning beds completely.
9.2. Secondary Prevention (Early Detection)
Skin Self-Examination: Monthly checks for new or changing moles/lesions using the ABCDE guide.
Clinical Skin Examination: Regular full-body skin exams by a dermatologist, particularly for high-risk individuals (e.g., personal/family history, numerous/atypical moles, fair skin, history of significant sun exposure/burns, immunosuppression). Screening frequency should be individualized based on risk.
Public Awareness: Continued education campaigns are needed to promote sun safety and the importance of early detection across all populations.
Conclusion
The landscape of melanoma in 2025 is one of contrasts: remarkable therapeutic progress coexists with persistent and complex challenges. The projections of continued high incidence, particularly the concerning trends in older women and the established burden in younger adults, highlight the ongoing need for effective prevention strategies. Immunotherapy and targeted therapies have transformed outcomes for advanced disease, with long-term survival and even cure now attainable for a significant proportion of patients, including those with the once-dire diagnosis of brain metastases, as evidenced by the landmark ABC trial results.
However, the fight is far from over. Overcoming treatment resistance, improving therapies for rare and challenging subtypes like acral, mucosal, and uveal melanoma, and effectively managing CNS disease remain critical frontiers. The stark disparities in outcomes experienced by People of Color demand urgent, multifaceted interventions focused on awareness, equitable access, and culturally sensitive care. Furthermore, supporting the growing population of melanoma survivors through dedicated research into managing long-term toxicities and psychosocial needs is essential.
The research priorities outlined by major funding bodies like the CDMRP MRP and organizations like MRA reflect these unmet needs, emphasizing innovation, collaboration, patient-centeredness, and a focus on areas like rare melanomas, resistance, and survivorship. The development and application of novel technologies, advanced preclinical models, sophisticated molecular analyses, and well-designed clinical trials, including those exploring oncolytic viruses like RP1 and novel combination strategies, will continue to drive progress. Ultimately, achieving the goal of significantly reducing the burden of melanoma requires a sustained, collaborative effort encompassing prevention, early detection, cutting-edge research, equitable treatment delivery, and comprehensive survivorship care.
References
(Note: This list includes references for concepts discussed and sources cited in the provided text or added through external research. DOI/PMIDs are provided where readily available. Some dates reflect access date for web resources.)
Ahlgrimm-Siess, V., Linder, D. M., & Hofmann-Wellenhof, R. (2018). Reflectance Confocal Microscopy in the Daily Practice. Current dermatology reports, 7(2), 81–91. doi:10.1007/s13671-018-0216-z
Amaria, R. N., Reddy, S. M., Tawbi, H. A., Davies, M. A., Ross, M. I., Glitza, I. C., ... & Wargo, J. A. (2018). Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nature medicine, 24(11), 1649-1654. doi:10.1038/s41591-018-0197-1
American Cancer Society (ACS), 2025 Projections. (As cited in user-provided text, specific report presumed based on context, e.g., Cancer Facts & Figures 2025 or related update).
Andtbacka, R. H., Kaufman, H. L., Collichio, F., Amatruda, T., Senzer, N., Chesney, J., ... & Coffin, R. S. (2015). Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. Journal of Clinical Oncology, 33(25), 2780–2788. doi:10.1200/JCO.2014.58.3377
Arnold, M., Singh, D., Laversanne, M., Vignat, J., Vaccarella, S., Meheus, F., ... & Bray, F. (2022). Global Burden of Cutaneous Melanoma in 2020 and Projections to 2040. JAMA dermatology, 158(5), 495–503. doi:10.1001/jamadermatol.2022.0160
Asgari, M. M., Maruti, S. S., Kuntz, K. M., & Argenyi, Z. B. (2012). Risk factors for development of melanoma in kidney and heart transplant recipients. Archives of dermatology, 148(8), 903–909. doi:10.1001/archdermatol.2012.772
Bellew, S., Katz, M., Michalski, M., Petkovska, I., Shiboski, S., Grekin, R. C., ... & Wei, M. L. (2023). Acral lentiginous melanoma: A United States multicenter survey based comparative analysis. Journal of the American Academy of Dermatology, 88(4), 802–810. doi:10.1016/j.jaad.2022.08.062
Binnewies, M., Roberts, E. W., Kersten, K., Chan, V., Fearon, D. F., Merad, M., ... & Coussens, L. M. (2018). Understanding the tumor immune microenvironment (TIME) for effective therapy. Nature medicine, 24(5), 541–550. doi:10.1038/s41591-018-0014-x
Blank, C. U., Rozeman, E. A., Fanchi, L. F., Sikorska, K., van de Wiel, B., Kvistborg, P., ... & Schumacher, T. N. (2018). Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nature medicine, 24(11), 1655-1661. doi:10.1038/s41591-018-0198-0
Carvajal, R. D., Spencer, S. A., & Lydiatt, W. (2018). Mucosal melanoma: a clinically and biologically unique disease entity. Journal of the National Comprehensive Cancer Network : JNCCN, 16(5S Suppl), 639–642. doi:10.6004/jnccn.2018.0044
CDMRP Melanoma Research Program (MRP). Fiscal Year 2025 (FY25) Pre-Announcement. (As cited in user-provided text). https://cdmrp.health.mil/mrp/default
Colantonio, S., Bracken, M. B., & Beecker, J. (2014). The association of indoor tanning and melanoma in adults: systematic review and meta-analysis. Journal of the American Academy of Dermatology, 70(5), 847–857.e18. doi:10.1016/j.jaad.2013.11.050
Curtin, J. A., Busam, K., Pinkel, D., & Bastian, B. C. (2006). Somatic activation of KIT in distinct subtypes of melanoma. Journal of Clinical Oncology, 24(26), 4340–4346. doi:10.1200/JCO.2006.06.2984
Davies, H., Bignell, G. R., Cox, C., Stephens, P., Edkins, S., Clegg, S., ... & Stratton, M. R. (2002). Mutations of the BRAF gene in human cancer. Nature, 417(6892), 949–954. doi:10.1038/nature00766
Dennis, L. K., Vanbeek, M. J., Beane Freeman, L. E., Smith, B. J., Dawson, D. V., & Coughlin, J. A. (2008). Sunburns and risk of cutaneous melanoma: Does age matter? A comprehensive meta-analysis. Annals of epidemiology, 18(8), 614–627. doi:10.1016/j.annepidem.2008.04.006
Dummer, R., Ascierto, P. A., Gogas, H. J., Arance, A., Mandala, M., Liszkay, G., ... & Robert, C. (2018). Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicentre, open-label, randomised phase 3 trial. The Lancet Oncology, 19(5), 603–615. doi:10.1016/S1470-2045(18)30142-6
Eggermont, A. M. M., Blank, C. U., Mandala, M., Long, G. V., Atkinson, V., Dalle, S., ... & Weber, J. (2018). Adjuvant Pembrolizumab versus Placebo in Resected Stage III Melanoma. The New England journal of medicine, 378(19), 1789–1801. doi:10.1056/NEJMoa1802357
Esteva, A., Kuprel, B., Novoa, R. A., Ko, J., Swetter, S. M., Blau, H. M., & Thrun, S. (2017). Dermatologist-level classification of skin cancer with deep neural networks. Nature, 542(7639), 115–118. doi:10.1038/nature21056
Fedorenko, I. V., Gibney, G. T., & Smalley, K. S. M. (2015). NRAS mutant melanoma: biological behavior and future strategies for therapeutic management. Oncogene, 34(13), 1611–1622. doi:10.1038/onc.2014.105
Gershenwald, J. E., Scolyer, R. A., Hess, K. R., Sondak, V. K., Long, G. V., Ross, M. I., ... & AJCC Melanoma Staging System, 8th Edition. (2017). Melanoma staging: Evidence-based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA: a cancer journal for clinicians, 67(6), 472–492. doi:10.3322/caac.21409
Hamid, O., & Bowles, T. (As cited in user-provided text regarding IGNYTE-3 trial). MRA Blog Post/News Article. September 23, 2024.
Hamid, O. et al. (2024). Primary analysis of a phase 2, multicohort study of RP1 + nivolumab in patients with anti–PD-1–failed melanoma. ESMO Congress 2024. Abstract LBA41. (As referenced in user-provided text).
Higgins, S., Nazemi, A., Chow, M., & Wysong, A. (2021). Review of Melanoma Disparities in Racial and Ethnic Minorities. Dermatology and therapy, 11(3), 665–683. doi:10.1007/s13555-021-00523-7
Hodi, F. S., O'Day, S. J., McDermott, D. F., Weber, R. W., Sosman, J. A., Haanen, J. B., ... & Urba, W. J. (2010). Improved survival with ipilimumab in patients with metastatic melanoma. The New England journal of medicine, 363(8), 711–723. doi:10.1056/NEJMoa1003466
Hodi, F. S., Chiarion-Sileni, V., Gonzalez, R., Grob, J. J., Rutkowski, P., Cowey, C. L., ... & Robert, C. (2018). Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. The Lancet Oncology, 19(11), 1480–1492. doi:10.1016/S1470-2045(18)30700-9
Hugo, W., Zaretsky, J. M., Sun, L., Song, C., Moreno, B. H., Hu-Lieskovan, S., ... & Ribas, A. (2016). Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell, 165(1), 35–44. doi:10.1016/j.cell.2016.02.065
Krantz, B. A., Dave, N., Komatsubara, K. M., Marr, B. P., & Carvajal, R. D. (2017). Uveal melanoma: epidemiology, etiology, and treatment of primary disease. Clinical ophthalmology (Auckland, N.Z.), 11, 279–289. doi:10.2147/OPTH.S89591
Krauthammer, M., Kong, Y., Bacchiocchi, A., Evans, P., Pornputtapong, N., Wu, C., ... & Bosenberg, M. (2015). Exome sequencing identifies recurrent mutations in NF1 and other genes in melanoma. Nature genetics, 47(9), 996–1002. doi:10.1038/ng.3361
Larkin, J., Chiarion-Sileni, V., Gonzalez, R., Grob, J. J., Cowey, C. L., Lao, C. D., ... & Hodi, F. S. (2019). Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. The New England journal of medicine, 381(16), 1535–1546. doi:10.1056/NEJMoa1910836
Law, M. H., Aoude, L. G., Duffy, D. L., & Sturm, R. A. (2020). Melanoma genetics. Melanoma research, 30(2), 125–132. doi:10.1097/CMR.0000000000000651
Long, G. V., Stroyakovskiy, D., Gogas, H., Levchenko, E., de Braud, F., Larkin, J., ... & Hauschild, A. (2017). Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet (London, England), 386(9992), 444–451. doi:10.1016/S0140-6736(15)60898-4 (COMBI-d)
Long, G. V., Atkinson, V., Lo, S., Sandhu, S., Guminski, A. D., Brown, M. P., ... & Menzies, A. M. (2025). Ipilimumab plus nivolumab versus nivolumab alone in patients with melanoma brain metastases (ABC): 7-year follow-up of a multicentre, open-label, randomised, phase 2 study. The Lancet Oncology. Published Online February 18, 2025. doi:10.1016/S1470-2045(24)00701-1 (As cited in user-provided text from MIA)
Parkin, D. M., Mesher, D., & Sasieni, P. (2011). Cancers attributable to solar (ultraviolet) radiation exposure in the UK in 2010. British journal of cancer, 105 Suppl 2(Suppl 2), S66–S69. doi:10.1038/bjc.2011.476
Rastrelli, M., Tropea, S., Rossi, C. R., & Alaibac, M. (2014). Melanoma: epidemiology, risk factors, pathogenesis, diagnosis and classification. In vivo (Athens, Greece), 28(6), 1005–1011. PMID: 25398793
Read, J., Wadt, K. A. W., & Hayward, N. K. (2016). Melanoma genetics. Journal of medical genetics, 53(1), 1–14. doi:10.1136/jmedgenet-2015-103154
Robert, C., Karaszewska, B., Schachter, J., Rutkowski, P., Mackiewicz, A., Stroiakovski, D., ... & Ribas, A. (2015). Improved overall survival in melanoma with combined dabrafenib and trametinib. The New England journal of medicine, 372(1),