Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2024 Jul 31;39(1):39–51. doi: 10.1111/jdv.20269

An overview of benefits and risks of chronic melanocortin‐1 receptor activation

M Böhm 1,, C Robert 2,3, S Malhotra 4,5,6, K Clément 7,8, S Farooqi 9
PMCID: PMC11664455  PMID: 39082868

Abstract

The melanocortin‐1 receptor (MC1R) is a G protein‐coupled receptor that plays a pivotal role in human skin pigmentation, melanin synthesis, redox homeostasis and inflammation. Loss‐of‐function MC1R variants suppress G protein‐coupled receptor coupling or cell surface expression leading to a decrease in adenyl cyclase activation and intracellular levels of cyclic adenosine monophosphate. Chronic activation of MC1R can occur in certain medical conditions such as Addison's disease and physiologic states such as pregnancy melasma. MC1R activation is more commonly caused by environmental exposure to ultraviolet (UV) radiation. Approved pharmacologic melanocortin agonists that activate MC1R signalling in a targeted manner or as a bystander effect have recently become available for erythropoietic protoporphyria, sexual desire disorders, monogenic obesity and syndromic obesity. Further, small peptide analogues of α–melanocortin‐stimulating hormone, human MC1R selective agonists, are photoprotective, decreasing the adverse impact of UV radiation (a primary risk factor for skin cancer) and are being investigated as potential chemoprevention strategies. MC1R activation through induction of UV‐protective skin pigmentation increased DNA repair, and control of aberrant cell growth may reduce the risk of melanoma but importantly does not prevent melanoma particularly in individuals with risk factors and regular skin examination remains critical in high‐risk individuals.


Thinner line widths indicate typical physiologic signalling, thicker lines indicate increased signalling and red x's indicate suppressed signalling. Larger eumelanin and pheomelanin ovals indicate higher protein synthesis, while smaller ovals indicate lower protein synthesis. Eumelanin and pheomelanin protein synthesis levels as compared to levels at MC1R basal activity are indicated as lower or higher by ‘<’ and ‘>’, respectively.

graphic file with name JDV-39-39-g003.jpg


Key points.

Why Was the Study Undertaken?

Although the structure of the melanocortin‐1 receptor (MC1R) and its variants in melanoma risk have been previously described, the impact of constitutive and pharmacologic activation of MC1R have not been extensively covered.

What Does this Study Add?

Long‐term activation of MC1R, either physiologically or pharmacologically, has not been associated with increased incidence of melanoma, whereas loss‐of‐function variants in MC1R have been associated with an increased risk of melanoma.

What Are the Implications of this Study for Disease Understanding and/or Clinical Care?

Although chronic MC1R activation has demonstrated benefits, it does not serve as a complete preventative measure for melanoma. Patients who receive melanocortin receptor pharmacotherapies should continue to limit ultraviolet radiation exposure and have regular dermatologic surveillance as part of standard, dermatologic health practice.

INTRODUCTION AND OVERVIEW OF MELANOCORTIN‐1 RECEPTOR STRUCTURE AND FUNCTION

Melanocortin receptor family

The melanocortin system is composed of multiple signalling hormones, ligands and associated melanocortin receptors (MCRs; melanocortin‐1 receptor [MC1R] through MC5R) that are involved in many biologic pathways and bodily functions. 1 , 2 , 3 , 4 MC1R plays a pivotal role in human skin pigmentation, melanin synthesis, DNA repair and inflammation (Figure 1). 1 , 2 , 3 , 5 , 6 , 7 , 8 , 9 MC2R, expressed in the adrenal cortex, is involved in steroidogenesis. 2 , 3 , 4 Adrenocorticotropic hormone (ACTH) binds to MC2R located on the surface of adrenal zona fasciculata cells, thereby producing cortisol. 10 MC3R and MC4R are expressed in the central nervous system and are pivotal in the regulation of hunger, satiety, and overall energy homeostasis. 3 , 4 , 11 , 12 MC3R has been linked to timing of sexual maturation and linear growth. 13 Some studies have reported the association between MC4R and sexual function. 2 , 3 MC5R has been linked to exocrine function, especially of the sebaceous glands. 2 , 3 , 4

FIGURE 1.

FIGURE 1

Melanocortin signalling for melanin production and anti‐inflammatory effects in melanocytes. 5 , 6 , 7 , 8 , 9 α‐MSH, α–melanocortin‐stimulating hormone; AC, adenylyl cyclase; cAMP, cyclic adenosine monophosphate; ACTH, adrenocorticotropic hormone; CREB, cAMP response element–binding protein; DCT, dopachrome tautomerase; IκBα, NF‐κB inhibitor α; MC1R, melanocortin‐1 receptor; MITF, microphthalmia‐associated transcription factor; NF‐κB, nuclear factor κB; PC1, proprotein convertase 1; PC2, proprotein convertase 2; PKA, protein kinase A; POMC, proopiomelanocortin; ROS, reactive oxygen species; TNFα, tumour necrosis factor α; TNFRI, tumour necrosis factor receptor type I; TYR, tyrosinase; UV, ultraviolet.

MC1R and pigmentation

MC1R is a 7‐transmembrane G protein‐coupled receptor expressed on the cell membrane of melanocytes that regulates melanocyte proliferation, melanin pigment synthesis, melanocyte differentiation and ultraviolet (UV) radiation sensitivity. 4 , 14 , 15 , 16 , 17 , 18 UV radiation is a natural inducer of MC1R expression and physiologic activation. 4 The high‐affinity MCR agonists, α–melanocyte‐stimulating hormone (α‐MSH) and ACTH are endogenous physiologic ligands that can activate MC1R. UV irradiation of skin induces p53 within the epidermis and directly upregulates expression of the precursor protein proopiomelanocortin (POMC). POMC is subsequently cleaved by prohormone convertase 1 to produce ACTH; prohormone convertase 2 then cleaves ACTH to generate α‐MSH. 3 , 4 , 14 , 19 , 20 For biologic activation, α‐MSH is further processed by C‐terminal carboxypeptidase, α‐amidating monooxygenase and N‐acetyltransferase. 19 POMC‐derived peptides including ACTH, α‐MSH and β‐endorphin are generated in the skin, especially in keratinocytes and melanocytes. 14 α‐MSH is also produced in the hypothalamus of adults but not in the pituitary gland. 3 , 21 The endogenous ligand α‐MSH activates MC4R, a key receptor in the melanocortin‐MC4R pathway, which is the primary pathway in the brain that mediates energy expenditure, hunger, and consequently, body weight. 22

α‐MSH also stimulates MC1R, and MC1R signalling can lead to the synthesis of melanin pigments: a dark brown/black UV radiation‐blocking pigment (eumelanin) and a yellow/red pigment (pheomelanin). 14 , 23 The balance of eumelanin and pheomelanin determines skin and hair colour. 24 When the skin is exposed to UV radiation and MC1R is activated by engagement with its newly synthesized ligand α‐MSH, the balance of melanin synthesis switches within melanocytes towards eumelanin. 5 , 8 Increased eumelanin production leads to darkening of the skin and pre‐existing nevi (i.e. benign skin growths formed by a cluster of melanocytes). 2 , 8 , 25

Anti‐inflammatory and immunomodulatory effects of MC1R activation

Eumelanin is photoprotective because it resists photodegradation, shields skin from UV radiation and scavenges reactive oxygen radicals. 23 Pheomelanin, a potential independent risk factor for melanoma, can amplify reactive oxygen species production and oxidative DNA damage. 26 , 27 , 28 α‐MSH has indirect antioxidative effects in melanocytes by regulating catalase and antioxidant response transcription factors such as nuclear factor κB (NF‐κB) and NF‐E2–related factor 2 (Nrf2). 19 , 29 , 30 α‐MSH inhibits activation of NF‐κB, a master regulator of inflammation by increasing intracellular levels of cyclic adenosine monophosphate (cAMP). Elevated levels of cAMP prevent degradation of NF‐κB inhibitor α, the inhibitory subunit of NF‐κB, thereby preventing p65 subunit translocation. 19 UV radiation‐induced down‐regulation of Nrf2 and the Nrf2‐dependent genes HMOX1, GCLC and GSTP1 is prevented or even overcorrected by α‐MSH. Of note, oxidized α‐MSH is unable to rescue suppression of Nrf2‐ and Nrf2‐dependent gene expression; therefore, this antioxidative mechanism may occur through the MC1R pathway. 29 UV radiation can lead to genotoxic stress from the generation of cyclobutene pyrimidine dimers, 6–4 photoproducts and 7,8‐dihydro‐8‐oxoguanine, resulting in DNA damage, including DNA breaks. 30 , 31 , 32 α‐MSH–mediated MC1R activation increases catalase and ferritin, improving cellular antioxidant status and mitigating oxidative DNA damage. 30 Given that pheomelanin incorporates cysteine into its structure, it may generate reactive oxygen species through depletion of glutathione stores. 27 α‐MSH has been shown to protect against UV radiation‐induced genotoxic stress and reduce cell death mediated by caspase‐3, H2O2, tumour necrosis factor α (TNF‐α) and interleukin‐1β (IL‐1β). 19

Along with regulating pigmentation, MC1R signalling is a key mediator of DNA repair and reactive oxygen species regulation following UV‐induced damage. 5 , 9 , 33 , 34 Studies have demonstrated that MC1R signalling promotes UV‐induced DNA repair via nucleotide excision repair pathways independent of melanin pigmentation. 9 , 35 , 36 , 37 In an in vitro study, α‐MSH demonstrated antiapoptotic effects following UV radiation in human melanocytes and keratinocytes that were independent of melanin content via nucleotide excision repair of cyclobutene pyrimidine dimer DNA lesions. 35 Conversely, loss‐of‐function MC1R variants can lead to compromised DNA repair response and increased apoptosis because expression of protective genes is reduced. 33 , 37

In addition to having a role in pigmentation and DNA repair, in vitro and in vivo studies have demonstrated that MC1R activation exhibits anti‐inflammatory and immunomodulatory properties (Figure 1). 2 , 38 , 39 , 40 Leukocytes and other cell types involved in inflammation express MC1R, which can mediate the immunomodulatory properties of melanocortins. 2 , 39 , 40 Melanocortins, including α‐MSH, can stimulate the production of immunosuppressive factors and inhibit the production of proinflammatory factors. 2 , 19 α‐MSH was initially demonstrated to suppress the well‐known proinflammatory cytokines interferon‐γ and TNF‐α and was later shown to suppress others such as IL‐1β, IL‐1, IL‐6, growth‐related oncogene α and lymphotactin. 19 The MC1R‐mediated anti‐inflammatory role of α‐MSH has also been shown to suppress allergen‐induced basophilic activation and cytokines including IL‐4, IL‐6 and IL‐13. 41 Further, α‐MSH induces expression of the immunosuppressive cytokine IL‐10 and exhibits further immunomodulatory effects through suppressing the expression of intercellular adhesion molecules and cell surface molecules including intercellular adhesion molecule 1, CD40, CD86 and ectopic major histocompatibility complex class I. α‐MSH can also suppress noncytokine proinflammatory mediators such as prostaglandins and nitric oxide. 19 In mice, α‐MSH has been shown to upregulate CD8+ T cells, reduce allergic contact sensitivity and reduce progressive tumour growth. 42

LOSS‐OF‐FUNCTION MELANOCORTIN‐1 RECEPTOR POLYMORPHISMS

Population‐based studies have demonstrated that MC1R is highly polymorphic, with ~200 identified allelic variants to date, with an ~60% prevalence for any MC1R variant among individuals of European ancestry. 4 , 43 , 44 , 45 Loss‐of‐function MC1R variants are associated with decreased eumelanin production, impaired tanning ability and UV radiation‐induced DNA damage in skin cells. 14 Loss of MC1R signalling can occur because of MC1R variants associated with key positions that reduce or block receptor function (i.e. activation of cAMP) via loss of G protein‐coupled receptor coupling or reduced cell surface expression. 46 Activation of loss‐of‐function MC1R variants has also been shown to activate p38 mitogen‐activated protein kinase (MAPK), extracellular signal‐regulated kinase 1 and 2 (ERK‐1/2) and Akt pathways. 47 , 48 , 49 , 50 , 51

An early study of MC1R variants in humans by Valverde et al reported an association of specific variants with red hair colour, fair skin and impaired tanning response to UV radiation, with MC1R variants identified in 82% of patients with red hair. 52 Individuals with both biallelic and heterozygous MC1R variants demonstrate impaired tanning following UV radiation. 53 , 54 In a study comparing patients with and without cutaneous malignant melanoma, the frequency of specific MC1R variants was significantly higher in patients with melanoma versus the control population, with a 2.2‐fold increase in risk for those with one active variant and a 4.1‐fold increase for those with two variants. 55 Although many variants affect the signalling ability of MC1R, the red hair colour variants D84E, R142H, R151C, R160W and D294H—which are associated with complete or partial loss of function of the MC1R—are especially associated with increased melanoma risk. 33 , 53 , 56 , 57 Mechanisms of loss of function of MC1R red hair colour variants include reduced receptor functional activity and reduced cell surface expression of the receptor. 53

MELANOMA: AETIOLOGY, MECHANISMS OF DEVELOPMENT AND THE ROLE OF MELANOCORTIN‐1 RECEPTOR SIGNALLING

Melanomas are malignant tumours that arise from uncontrolled melanocyte proliferation (Figure 2, 5 , 7 , 8 , 50 , 58 , 59 , 60 , 61 , 62 , 63 , 64 , 65 , 66 ). Malignant melanoma accounts for most skin cancer‐related deaths. Sun exposure and UV radiation damage are the primary causes of melanoma, with most cases of melanoma (range, ~60%–85%) believed to be caused by excess UV radiation. 9 , 67 , 68 , 69 UV radiation and the resulting DNA damage cause deleterious variants that drive the oncogenic process. 9 Both UVA and UVB radiation can lead to direct DNA damage, with UVB generating cyclobutene pyrimidine dimer formation and cell apoptosis and UVA damage promoting reactive oxygen species that induce direct DNA damage. 9 , 68

FIGURE 2.

FIGURE 2

MC1R signalling pathway during basal physiologic (left), hyperactivation (middle) and loss of function with UV radiation (right) conditions. 5 , 7 , 8 , 50 , 58 , 59 , 60 , 61 , 62 , 63 , 64 , 65 , 66 Thinner line widths indicate typical physiologic signalling, thicker lines indicate increased signalling and red x's indicate suppressed signalling. Larger eumelanin and pheomelanin ovals indicate higher protein synthesis, while smaller ovals indicate lower protein synthesis. Eumelanin and pheomelanin protein synthesis levels as compared to levels at MC1R basal activity are indicated as lower or higher by ‘<’ and ‘>’, respectively. α‐MSH, α–melanocortin‐stimulating hormone; AC, adenylyl cyclase; cAMP, cyclic adenosine monophosphate; BRAF, B‐Raf proto‐oncogene, serine/threonine kinase; CREB, cAMP response element–binding protein; ERK‐1/2, extracellular signal–regulated kinase 1 and 2; MC1R, melanocortin‐1 receptor; MEK, mitogen‐activated protein kinase kinase; MITF, microphthalmia‐associated transcription factor; pAkt, phospho‐Akt; PI3K, phosphatidylinositol 3‐kinase; PKA, protein kinase A; PIP2, phosphatidylinositol 4,5‐bisphosphate; PIP3, phosphatidylinositol 3,4,5‐trisphosphate; RAS, rat sarcoma; ROS, reactive oxygen species; TYR, tyrosinase; UV, ultraviolet.

Melanoma development can have multifactorial causes, with driving contributions from the environment (i.e. UV radiation) and genetic predisposition. 68 Seven of the nine most‐studied MC1R variants are positively associated with melanoma, while five and two of these variants are also positively associated with red hair and fair skin, respectively (Table 1, 70 ). All of the most studied MC1R variants positively associated with red hair and/or fair skin were also positively associated with melanoma. 70 In most cases, melanoma proliferation is under the constitutive activation of the MAPK signalling pathway. 71 Mutations in the proto‐oncogenes BRAF (50%–70%) and, to a lesser extent, NRAS (15%–30%) and KIT (2%–5%), are primarily responsible for MAPK activation in melanoma. 71 , 72 NF1 variants, which are typically loss‐of‐function variants, can also lead to dysregulation of the MAPK signalling pathway and account for an estimated 12%–18% of melanomas. 73

TABLE 1.

Association of phenotype characteristics and melanoma in the most‐studied MC1R variants. 70

MC1R variant Variant association with
Fair skin Red hair Melanoma
V60L
V92M
I155T +
R163Q +
D84E + +
R142H + +
R151C + +
R160W + + +
D294H + + +

Characteristics positively associated with a variant are denoted with ‘+’ and characteristics not positively associated with a variant are denoted with ‘–’.

The neoplastic transformation process requires additional genetic changes, namely telomerase reverse transcriptase overexpression or suppression of the retinoblastoma and/or p53 pathways. 71 A variant in the promoter of TERT is involved in ~70% of sporadic melanomas, and this variant is induced by UV radiation. 71 Variants in CDKN2A, an important tumour suppressor, have also been identified in 10%–40% of familial cases of melanoma. 74 MC1R variants have also been associated with other pathways, including the MAPK, ERK and Akt pathways. 49 , 50 , 65 , 75 , 76 Although there is evidence that hyperactivation of cAMP, ERK and Akt signalling can drive oncogenic transformation, these pathways have also been demonstrated to promote DNA damage response and mitigate oxidative stress and DNA damage. 47 , 49 , 50 , 75 , 76 MC1R signalling may be protective by contributing to eumelanin production and preventing or reducing DNA genotoxicity and enhancing DNA repair mechanisms. 5 , 30 , 71

While MC1R polymorphisms can increase phosphatidylinositol 3‐kinase signalling and senescence in melanocytes, hyperactivation of this pathway in melanocytes expressing BRAF V600E can lead to oncogenesis. 77 However, UVB irradiation in transgenic mice with compromised MC1R signalling and conditional melanocyte‐specific Braf V600E expression led to the development of melanoma. Notably, inhibition of the depalmitoylation enzyme acyl‐protein thioesterase 2 led to delayed onset and lower rates of melanoma in mice expressing melanocyte‐specific zinc finger DHHC‐type palmitoyltransferase 13 and an MC1R variant. 77 , 78

There are also population‐based risk factors associated with melanoma. Globally, age‐standardized rates of melanoma incidence vary among regions, ranging from the highest in Australia and New Zealand (35.8 per 100,000 population) to the lowest in West Africa (0.33 per 100,000 population). 79 Variations in melanoma incidence could be attributed to skin phenotype and excessive sun exposure potentially related to cultural and/or economic differences. 80 , 81 Overall, men have a higher incident age‐standardized rate of melanoma than women. 79 , 82 Notably, women have higher incidence in younger age groups and men have a higher incidence in older age groups. 81 However, an increased trend in incidence was observed in individuals aged ≥50 years, regardless of sex. 79 The role of MC1R in other cancers is under investigation. Interestingly, MC1R expression and signalling has been associated with colon and breast cancer and may serve as an independent prognosis marker. 76 , 83 , 84 , 85

EXAMPLES OF CONDITIONS AND SITUATIONS OF CONSTITUTIVE MELANOCORTIN‐1 RECEPTOR ACTIVATION

Ultraviolet radiation

As discussed previously, UV radiation, such as sun exposure or tanning, induces α‐MSH and ACTH production and thus stimulates MC1R‐mediated eumelanin synthesis. 2 , 4 Whereas UV‐associated radiation is a primary cause of DNA damage and melanoma, UV radiation‐induced MC1R activation can be protective against melanoma by enhancing DNA repair pathways (i.e. nucleotide excision repair), protecting against reactive oxygen species and promoting melanogenesis. 4 , 9 , 30 , 35 , 36 , 37 , 86

Addison's disease

Addison's disease, or chronic primary adrenal insufficiency, is a rare endocrine disorder resulting in cortisol deficiency. Decreased cortisol levels cause decreased feedback to the hypothalamic–pituitary axis leading to increased secretion of ACTH, which contributes to skin and mucous membrane hyperpigmentation via MC1R activation. 61 Although an overall increase in mortality due to malignancies is observed in Addison's disease, no elevated risk of any skin cancer, including melanoma, has been reported in epidemiologic studies. 87 One study followed 3299 patients with primary adrenocortical insufficiency for 40 years, determining that the standardized incidence ratio (SIR) for melanoma skin cancer was numerically less than expected (0.7; 95% confidence interval, 0.2–1.6), consistent with the eumelanin‐mediated protective effects of MC1R agonism, although a higher incidence of nonmelanoma skin cancer was reported (SIR, 2.1; 95% confidence interval, 1.3–3.1), along with higher incidences of oral cancer and male genital system cancer. 23 , 88 To our knowledge, no studies of primary adrenal insufficiency (and the associated chronic activation of MC1R) suggesting an association with increased incidence of melanoma have been published.

Pregnancy Melasma

Hyperpigmentation occurs in ~85%–90% of pregnant women, appears to be associated, at least in part, with increased expression and/or signalling of MC1R, and typically resolves post‐partum. 64 Notably, increased levels of α‐MSH in plasma have been observed during the third trimester of normal pregnancy. 89 During pregnancy, melanocytes may be sensitive to elevated levels of α‐ and β‐MSH, as well as oestrogen, progesterone and β‐endorphins, which likely stimulate melanin production. 64 While skin folds and intertriginous areas may darken, the most commonly affected areas include the areolas, nipples, genitalia, axillae, periumbilical area and inner thighs. 64 Further, the linea alba may darken, and acanthosis nigricans can develop. 64 In a cohort of 2025 patients with melanoma, 156 (7.7%) had pregnancy‐associated melanoma; no significant difference in survival outcomes was observed between people developing pregnancy‐ and non–pregnancy‐associated melanoma. 90

PHARMACOLGIC ACTIVATION OF MELANOCORTIN‐1 RECEPTOR

Approved Melanocortin pharmacotherapies

In addition to conditions and environmental factors that promote physiologic MC1R activation, there are also MC1R agonists and various other pharmacologic treatments that activate MC1R signalling that are commercially available and in development in the United States and/or European Union for indications including erythropoietic protoporphyria (EPP), sexual desire disorders and monogenic and syndromic obesity (Table 2, 60 , 91 , 92 , 93 , 94 , 95 , 96 , 97 , 98 , 99 , 100 , 101 , 102 ). Various investigational MC1R‐targeting compounds are also in clinical development.

TABLE 2.

Approved melanocortin pharmacotherapies and melanocortin pharmacotherapies in development.

Pharmacotherapy Indications or treatment population Mechanism of action Structure MCR interactions Common adverse events (≥10%)
Approved melanocortin pharmacotherapies
Afamelanotide 91 , 92 , 93 Approved to increase pain‐free light exposure in adults with a history of phototoxic reactions from erythropoietic protoporphyria MC1R agonist

Linear 13‐amino acid peptide with the following structure:

Ac‐Ser‐Tyr‐Ser‐Nle‐Glu‐His‐(D)Phe‐Arg‐Trp‐Gly‐Lys‐Pro‐Val‐NH2 • × CH3COOH

MC1R > MC3R > MC4R > MC5R > MC2R Nausea and headache
Bremelanotide 60 Approved for acquired, generalized hypoactive sexual desire disorder in premenopausal women Nonselective MCR agonist

Cyclic 7‐amino acid peptide with the following structure:

Ac‐Nle‐cyclo‐(Asp‐His‐D‐Phe‐Arg‐Trp‐Lys‐OH) • × CH3COOH

MC1R > MC4R > MC3R > MC5R > MC2R Nausea, flushing, injection site reactions a and headache
Setmelanotide 94 , 95 , b Approved for treatment of obesity and control of hunger in adult and paediatric patients with specifics forms of monogenic or syndromic obesity due to variants in POMC, PCSK1 or LEPR variants and BBS MC4R agonist

Cyclic 8–amino acid peptide with the following structure:

Ac‐Arg‐Cys‐(D)Ala‐His‐(D)Phe‐Arg‐Trp‐Cys‐NH2 • × CH3COOH

MC4R > MC3R ≈ MC1R Skin hyperpigmentation, injection site reactions, c nausea, vomiting, headache and spontaneous penile erection d
Melanocortin pharmacotherapies in development
Repository corticotropin injection 96 , 97 , 98 Adults with rheumatoid arthritis e Nonselective MCR agonist Mixture of adrenocorticotropic hormone analogues and other pituitary peptides MC4R > MC3R > MC1R > MC2R > MC5R f Infection, g cushingoid, irritability, diarrhoea, acne and convulsion h
PL‐8177 99 , 100 Adults with active ulcerative colitis Selective MC1R agonist Cyclic 7‐amino acid peptide MC1R NA
RM‐718 102 , b Adult and paediatric patients with hypothalamic obesity Highly selective MC4R agonist 7‐((3H‐indol‐3‐yl) methyl)‐19‐(2‐acetamido‐5‐guanidinopentanamido)‐13‐benzyl‐10‐(3‐guanidinopropyl)‐16‐(1‐hydroxyethyl)‐3,3‐dimethyl‐6,9,12,15,18‐pentaoxo‐1,2‐dithia‐5,8,11,14,17‐pentaazacyclohenicosane‐4‐carboxamide hydrochloride MC4R > MC5R > MC3R > MC1R > MC2R NA

α‐MSH, α–melanocyte‐stimulating hormone; BBS, Bardet–Biedl syndrome; MCR, melanocortin receptor; M1CR, MC1 receptor; NA, not applicable or not available.

a

Includes injection site pain, unspecified injection site reactions, erythema, hematoma, pruritus, haemorrhage, bruising, paresthesia and hypoesthesia.

b

Unpublished data, Rhythm Pharmaceuticals, Inc.; 2023.

c

Injection site reactions include injection site‐associated events of erythema, pruritus, oedema, pain, induration, bruising, reaction, swelling, haemorrhage, hypersensitivity, hematoma, nodule, discoloration, erosion, inflammation, irritation, warmth, atrophy, discomfort, dryness, mass, hypertrophy, rash, scar, abscess and urticaria.

d

Male patients.

e

Acthar Gel is indicated as monotherapy for the treatment of infantile spasms in infants and children age <2 years, exacerbations of multiple sclerosis in adults and may be used for the following disorders and diseases: rheumatic, collagen, dermatologic, allergic states, ophthalmic, respiratory and oedematous state.

f

Partial agonist.

g

Including candidiasis, otitis media, pneumonia and upper respiratory tract infections.

h

In the treatment of infantile spasms, other types of seizures/convulsions may occur because some patients with infantile spasms progress to other forms of seizures (e.g. Lennox–Gastaut syndrome). Additionally, the spasms sometimes mask other seizures, and once the spasms resolve after treatment, the other seizures may become visible.

Afamelanotide

Afamelanotide, the first and most studied synthetic α‐MSH analogue, is a nonselective melanocortin tridecapeptide that can bind and activate MC1R. 63 , 103 , 104 The structure of afamelanotide, [Nle, 4 D‐Phe 7 ]‐α‐MSH (also referred to as NDP‐α‐MSH), is identical to physiologic α‐MSH, with the exception of two amino acids at positions 4 and 7. 63 , 104 Afamelanotide is a subcutaneous implant allowing for controlled release that is approved by the United States Food and Drug Administration (FDA) to increase pain‐free light exposure in adults with a history of phototoxic reactions from EPP and by the European Medicines Agency (EMA) to prevent phototoxicity in adults with EPP. 91 , 92 Following visible light exposure, patients with EPP experience severe, phototoxic‐related pain and consequently may dramatically limit their exposure to light. Therefore, this population may represent a low–UV‐exposed population in which to assess the impact of chronic MC1R agonism without the major risk factor of developing UV‐mediated melanoma. 63

To date, 201 patients have been treated in a clinical setting. 105 , 106 In an open‐label, Phase 2 trial, 5 patients with solar urticaria received a single subcutaneous dose of afamelanotide, which resulted in increased melanin density and photosensitivity improvement, such as increased minimum urticarial dose and decreased weal area. An increase in pre‐existing nevi pigmentation was reported. 107 Afamelanotide may also have an anti‐inflammatory effect in patients with mild‐to‐moderate acne vulgaris, as demonstrated by an open‐label, Phase 2 trial (n = 3) in which afamelanotide resulted in a decreased number of total and inflammatory acne lesions. 108 In Phase 3 double‐blind randomized placebo‐controlled trials of 167 patients with EPP, melanocytic nevi were reported in 2 of 86 patients (4%) receiving afamelanotide and 1 of 81 patients (2%) receiving placebo. Mild hyperpigmentation occurred at the implant site in one‐third of the patients who received afamelanotide. No melanomas were reported in the afamelanotide group. 106 In a placebo‐controlled study of 65 White individuals, melanin density was significantly increased in all those receiving 3 months of treatment with afamelanotide, which was accompanied by decreased thymine dimer formation (indicative of reduced DNA damage). 109 In a longitudinal observational study of 115 patients with EPP treated with afamelanotide for up to 8 years, 2 patients reported a new melanocytic nevus, which appeared 2.5 years after the first dose of afamelanotide in 1 patient and 5 years after the first dose in the other patient. 105 More than 1000 patients have been exposed to afamelanotide with no melanoma events reported, with many who receive continuous treatment for 5 years and some for >10 years. 91 , 110

Bremelanotide

Bremelanotide is a nonselective MCR agonist approved by the FDA for the treatment of acquired generalized hypoactive sexual desire disorder in premenopausal women. 60 To date, 684 patients have been treated in a clinical setting. 111 While bremelanotide is an agonist for all 5 MCRs, bremelanotide primarily acts through MC1R and MC4R. 60 In Phase 3 placebo‐controlled trials of bremelanotide, 1% of individuals who received bremelanotide (n = 627) reported hyperpigmentation. 60 During the open‐label periods of these trials, an additional 7 individuals receiving bremelanotide (1%; n = 684) experienced hyperpigmentation, 6 of whom had received placebo during double‐blind treatment. 111 Two patients experienced melanocytic nevi; cases were mild and resolved in both patients, and only 1 case was considered possibly related to bremelanotide. No malignant melanomas or other cutaneous malignancies were reported in any of these studies. 111

Setmelanotide

Setmelanotide is approved by the FDA, EMA and Medicines and Healthcare products Regulatory Agency for the management of obesity and approved by the EMA and Healthcare products Regulatory Agency for the control of hunger in patients with certain rare genetic forms of monogenic or syndromic obesity. 94 , 95 , 112 Setmelanotide is an MCR agonist that acts primarily on MC4R but also activates MC1R with >20‐fold less activity than that for MC4R. 62 To date, 926 patients have been treated in a clinical setting (unpublished data, Rhythm Pharmaceuticals, Inc.; 2023). While setmelanotide partially acts on MC1R, only 56% of patients reported skin hyperpigmentation (i.e. tanning) during the first month of setmelanotide treatment at clinical doses (unpublished data, Rhythm Pharmaceuticals, Inc.; 2023). 113 , 114 , 115 , 116 Although comparisons between different studies are limited, the proportion of patients who experienced hyperpigmentation was greater than that among those treated with afamelanotide; the mechanism of this discrepancy is currently unknown (unpublished data, Rhythm Pharmaceuticals, Inc.; 2023). 106 The observed hyperpigmentation is generally uniform and plateaus before returning to baseline within ~1 month of treatment discontinuation (data on file, Rhythm Pharmaceuticals, Inc.) 115 , 117 , 118 Some patients also experience darkening of pre‐existing nevi or development of new nevi (clinical trial setting, n = 1; real‐world setting, n = 66 [data on file, Rhythm Pharmaceuticals, Inc.; 2023]) and changes in hair colour. 115 , 117 , 118 , 119 Outcomes following long‐term exposure of setmelanotide were assessed in 2 patients with POMC deficiency who participated in a Phase 2, open‐label pilot study, followed by an open‐label extension study, with each receiving setmelanotide daily for a range of 6.8–7.2 years. Although these patients experienced changes in hair colour and darkening of nevi, no additional persistent treatment‐related adverse events associated with skin were observed. 119 , 120

All patients receiving setmelanotide treatment, whether in the clinical or real‐world setting, are recommended to have a baseline dermatologic examination and regular follow‐up visits because of skin changes due to the increased pigmentation. 95 Across the setmelanotide clinical program, 1 patient with Bardet–Biedl syndrome, fair skin (Fitzpatrick skin type II), freckling, nevi and a history of excessive sun exposure was diagnosed with stage pT1a melanoma after 3 years of continuous treatment (unpublished data, Rhythm Pharmaceuticals, Inc.) The lesion was excised for cure. A second patient on commercial therapy with Bardet–Biedl syndrome, fair skin (Fitzpatrick skin type II), and a skin examination revealing extensive chronic UV‐related damage who lived on an island close to the equator experienced increased pigmentation of the skin and existing melanocytic nevi and was diagnosed with stage pT1a superficial spreading melanoma after 6 months of setmelanotide treatment. In both cases, the individuals were at high risk of melanoma. These patients receive regular dermatologic examinations as recommended, which resulted in earlier melanoma diagnosis and timely excision (data on file, Rhythm Pharmaceuticals, Inc.).

Nonlicensed and investigational Melanocortin pharmacotherapies

Melanocortin pharmacotherapies in development

Several melanocortin pharmacotherapies are currently in development, including repository corticotropin injection, PL‐8177, RM‐718, PL‐8331 and PL‐5000 (data on file, Rhythm Pharmaceuticals, Inc.). 96 , 97 , 99 , 100 , 101 , 102 Details of these pharmacotherapies are included in Table 2.

Melanotan II

Melanotan II is a synthetic cyclic α‐MSH analogue that is a nonselective MCR agonist and is not approved for use in any therapeutic indication. 121 , 122 Melanotan II can be purchased online and is typically self‐administered by individuals seeking the cosmetic effects of MC1R activation with the intent of increasing skin pigmentation. 121 Melanotan II induces eumelanin synthesis through MC1R activation and exaggerates UV‐mediated tanning. 123 Because melanotan II can cross the blood–brain barrier and is nonselective, it also activates the other MCRs, resulting in adverse events including fatigue, loss of appetite and penile erection. 121 , 124 However, loss of appetite resulting in weight loss and increased libido by actions of melanotan II within the central nervous system are often welcome side effects for the purchasers of this ‘Barbie drug’. 121 , 125 , 126 As an unregulated ‘treatment’, melanotan II has the additional potential risks associated with unknown impurities and varied concentrations. 121 , 125

There have been at least five reports of melanomas during or after melanotan II use. All individuals had other risk factors such as fair skin, excess sun exposure/tanning bed use and/or family history of melanoma. 125 , 127 , 128 , 129 , 130 A 20‐year‐old woman with Fitzpatrick skin type II and a history of tanning bed use was diagnosed with melanoma on her left gluteal region 3 months after a 3‐week course of self‐administered melanotan II treatment. 129 In one report of a 23‐year‐old White woman with red hair and a familial history of melanoma, three melanoma lesions were identified on the back, abdomen and right arm following 3 years of self‐administered melanotan II treatment. Genetic testing revealed heterozygous gene variants in MC1R and CDKN2A, a high‐risk melanoma‐associated gene. Multiple melanoma risk factors such as familial history, sun‐seeking behaviour and other genetic factors, including MC1R variants, may increase the risk associated with melanotan II. 125 A 23‐year‐old woman with Fitzpatrick skin type II and a history of tanning bed use developed melanoma on a pre‐existing nevus on her left knee 4 months after seven weekly self‐administered melanotan II treatments. 130 A 42‐year‐old woman with Fitzpatrick skin type III and a history of sun exposure and tanning bed use developed melanoma on a nevus on the abdomen 3 months after a 1‐week course of self‐administered melanotan II treatment. 128 A 66‐year‐old male with Fitzpatrick skin type II developed melanoma in‐situ on his right mandibular angle after 4 weeks of self‐administered melanotan II treatment. 127 Additional skin conditions, such as dysplastic compound nevus, have also been reported after melanotan II use. 131 Individuals should avoid the use of nonlicensed pharmaceutical agents, including melanotan II, and consult with a healthcare professional about the risks and benefits of such an unregulated purchase. Safety of this nonlicensed drug has not been proven and, because of the potential risks associated with melanotan II, thorough skin examinations may lead to an earlier diagnosis of melanoma or other skin diseases. 128 , 130

Small peptide analogues of α‐MSH

Peptides, or any polymer of amino acids, are classified by the number of amino acids they contain (e.g. tripeptides are composed of three amino acid residues and tetrapeptides are composed of four amino acid residues). The development of small peptide analogues of α‐MSH that are human MC1R selective agonists for topical application can allow for photoprotection and chemoprevention of skin cancers. 14 Tetrapeptide α‐MSH analogues have been shown to be more potent than endogenous α‐MSH in activating MC1R and have improved selectivity over NDP‐α‐MSH for MC1R. 103 , 132 Compared with α‐MSH, tripeptide α‐MSH analogues are similarly effective in activating cAMP, inhibiting H2O2 formation and enhancing DNA repair. 133 KdPT is an α‐MSH–related tripeptide derivative that is structurally similar to the three amino acid residues of the α‐MSH C‐terminal and amino acids 193–195 of IL‐1β. 134 KdPT has potent anti‐inflammatory effects capable of suppressing IL‐β–induced NF‐κB activation and IL‐6 and IL‐8 expression. 19 KdPT is also able to attenuate the TNF‐α– and interferon‐γ–induced breakdown of transepithelial electrical resistance in models of intestinal inflammation. KdPT treatment significantly reduced the severity of dextran sodium sulfate‐induced colitis in IL‐1R–deficient mice and severity of piroxicam‐induced colitis in IL‐10–deficient mice compared with control phosphate‐buffered saline treatment. The same beneficial effect of KdPT treatment on dextran sodium sulfate‐induced colitis was not seen in MC1R‐deficient mice, which suggests a reduced capacity of KdPT to mitigate colitis in the absence of MC1R and a potential role of MC1R in the mechanism of action of KdPT. 134 However, KdPT does not appear to bind to the MC1R and is not pigment inducing (i.e. melanotropic). 135 A Phase 2 trial in patients with ulcerative colitis confirmed the anti‐inflammatory action of KdPT without any evidence for pigment induction as an adverse effect. 136 In accordance with these in vivo observations, KdPT did not induce pigmentation in B16 melanoma cells in vitro, indicating that this tripeptide is not melanotropic and not a bona fide melanocortin. 134 , 135

Dersimelagon

Dersimelagon, an oral selective MC1R agonist, significantly inhibited skin fibrosis and lung inflammation when administered prophylactically and suppressed skin fibrosis development when administered as a therapeutic treatment in a bleomycin‐induced systemic sclerosis mouse model. 137 , 138 Further, dersimelagon suppressed the activation of inflammatory cells and inflammation‐related signals. 137

Dersimelagon has been evaluated in a randomized, placebo‐controlled, Phase 2 trial in patients with EPP (n = 93) or X‐linked protoporphyria (n = 9). Following 16 weeks of dersimelagon treatment, the least‐squares mean difference from placebo in the change from baseline in mean daily time to the first prodromal symptom associated with sun exposure increased by ~1 hour (53.8 min for those who received 100 mg and 62.5 min for those who received 300 mg) compared with placebo. Hyperpigmentation was reported in 14 patients (21%) who received dersimelagon and no patients who received placebo. Melanocytic nevus was reported in 11 patients (16%) who received dersimelagon compared with 3 (9%) who received placebo. Melanoma was not reported as an adverse event in this study. 139

SUMMARY AND CONCLUSIONS

Activation of MC1R in melanocytes results in increased eumelanin synthesis that shields against UV radiation, enhances DNA repair activity and increases antioxidant capacity. Increased skin pigmentation may be associated with a darkening of pre‐existing nevi. Long‐term activation of MC1R, either physiologically or pharmacologically, has not been shown to be associated with increased incidence of melanoma whereas loss‐of‐function variants in MC1R have been associated with an increased risk of melanoma. MC1R activation through induction of UV radiation‐protective skin pigmentation, increased DNA repair and control of aberrant cell growth may reduce the risk of melanoma but does not prevent the development of melanoma.

UV light exposure is the primary risk factor for the development of melanoma, and fair‐skinned individuals (Fitzpatrick ≤3) are at increased risk of melanoma independent of pharmacologic activation of MC1R; these individuals should have regular close dermatologic surveillance. Because the pigment‐enhancing effect of MC1R agonism may unmask a pre‐existing melanoma, careful surveillance in this setting may allow for an earlier diagnosis with a greater chance of curative resection. Although dark‐skinned individuals may be at lower risk of melanoma, and chronic stimulation of MC1R with proportionally more eumelanin secretion may be further protective, it does not prevent melanoma. Patients who receive MCR pharmacotherapies should continue to limit UV radiation exposure and have regular dermatologic surveillance, which can lead to early identification of pre‐existing melanoma.

Although chronic MC1R activation has demonstrated benefits, such as UV‐protective skin pigmentation, enhanced DNA repair, regulation of aberrant cell growth and reduction of melanoma risk, it does not serve as a complete preventive measure, especially in individuals with melanoma risk factors. Regular skin examination remains crucial for those at high risk of developing melanoma, including those with fair skin, excess sun exposure/tanning bed use and/or family history of melanoma.

FUNDING INFORMATION

Writing and editorial assistance for the preparation of this article was provided under the direction of the authors by MedThink SciCom and funded by Rhythm Pharmaceuticals, Inc.

CONFLICT OF INTEREST STATEMENT

MB, CR and SF have no conflicts of interests to disclose. At the time of first submission, SM was a full‐time employee of Rhythm Pharmaceuticals, Inc. and held company‐awarded stocks or stock options. KC is primary investigator for setmelanotide trials in the obesity field.

ETHICS STATEMENT

Not relevant because this is a review article and did not involve any human or animal research.

ACKNOWLEDGEMENTS

Writing and editorial assistance for the preparation of this article was provided under the direction of the authors by Kelby Killoy, PhD, and David Boffa, ELS, MedThink SciCom. Open Access funding enabled and organized by Projekt DEAL.

Böhm M, Robert C, Malhotra S, Clément K, Farooqi S. An overview of benefits and risks of chronic melanocortin‐1 receptor activation. J Eur Acad Dermatol Venereol. 2025;39:39–51. 10.1111/jdv.20269

DATA AVAILABILITY STATEMENT

Data sharing not applicable to this article as no data sets were generated or analysed during the current study.

REFERENCES

  • 1. Bashmakova EE, Krasitskaya VV, Bondar AA, Eremina EN, Slepov EV, Zukov RA, et al. Bioluminescent SNP genotyping technique: development and application for detection of melanocortin 1 receptor gene polymorphisms. Talanta. 2018;189:111–115. [DOI] [PubMed] [Google Scholar]
  • 2. Gantz I, Fong TM. The melanocortin system. Am J Physiol Endocrinol Metab. 2003;284(3):E468–E474. [DOI] [PubMed] [Google Scholar]
  • 3. Ericson MD, Lensing CJ, Fleming KA, Schlasner KN, Doering SR, Haskell‐Luevano C. Bench‐top to clinical therapies: a review of melanocortin ligands from 1954 to 2016. Biochim Biophys Acta Mol Basis Dis. 2017;1863(10 Pt A):2414–2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. García‐Borrón JC, Abdel‐Malek Z, Jiménez‐Cervantes C. MC1R, the cAMP pathway, and the response to solar UV: extending the horizon beyond pigmentation. Pigment Cell Melanoma Res. 2014;27(5):699–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Swope VB, Abdel‐Malek ZA. MC1R: front and center in the bright side of dark eumelanin and DNA repair. Int J Mol Sci. 2018;19(9):2667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Catania A. The melanocortin system in leukocyte biology. J Leukoc Biol. 2007;81(2):383–392. [DOI] [PubMed] [Google Scholar]
  • 7. Takeuchi R, Kambe M, Miyata M, Jeyadevan U, Tajima O, Furukawa K, et al. TNFα‐signal and cAMP‐mediated signals oppositely regulate melanoma‐ associated ganglioside GD3 synthase gene in human melanocytes. Sci Rep. 2019;9(1):14740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Wolf Horrell EM, Boulanger MC, D'Orazio JA. Melanocortin 1 receptor: structure, function, and regulation. Front Genet. 2016;7:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Yin K, Sturm RA, Smith AG. MC1R and NR4A receptors in cellular stress and DNA repair: implications for UVR protection. Exp Dermatol. 2014;23(7):449–452. [DOI] [PubMed] [Google Scholar]
  • 10. Angelousi A, Margioris AN, Tsatsanis C. ACTH action on the adrenals. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext [Internet]. South Dartmouth, MA: MDText.com, Inc.; 2000. Updated June 13, 2020. https://www.ncbi.nlm.nih.gov/books/NBK279118/. [Google Scholar]
  • 11. Huvenne H, Dubern B, Clément K, Poitou C. Rare genetic forms of obesity: clinical approach and current treatments in 2016. Obes Facts. 2016;9(3):158–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Yazdi FT, Clee SM, Meyre D. Obesity genetics in mouse and human: back and forth, and back again. PeerJ. 2015;3:e856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Lam BYH, Williamson A, Finer S, Day FR, Tadross JA, Gonçalves Soares A, et al. MC3R links nutritional state to childhood growth and the timing of puberty. Nature. 2021;599(7885):436–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Abdel‐Malek ZA, Swope VB, Starner RJ, Koikov L, Cassidy P, Leachman S. Melanocortins and the melanocortin 1 receptor, moving translationally towards melanoma prevention. Arch Biochem Biophys. 2014;563:4–12. [DOI] [PubMed] [Google Scholar]
  • 15. Holcomb NC, Bautista RM, Jarrett SG, Carter KM, Gober MK, D'Orazio JA. cAMP‐mediated regulation of melanocyte genomic instability: a melanoma‐preventive strategy. Adv Protein Chem Struct Biol. 2019;115:247–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Aberdam E, Bertolotto C, Sviderskaya EV, de Thillot V, Hemesath TJ, Fisher DE, et al. Involvement of microphthalmia in the inhibition of melanocyte lineage differentiation and of melanogenesis by agouti signal protein. J Biol Chem. 1998;273(31):19560–19565. [DOI] [PubMed] [Google Scholar]
  • 17. Suzuki I, Cone RD, Im S, Nordlund J, Abdel‐Malek ZA. Binding of melanotropic hormones to the melanocortin receptor MC1R on human melanocytes stimulates proliferation and melanogenesis. Endocrinology. 1996;137(5):1627–1633. [DOI] [PubMed] [Google Scholar]
  • 18. Swope VB, Jameson JA, McFarland KL, Supp DM, Miller WE, McGraw DW, et al. Defining MC1R regulation in human melanocytes by its agonist α‐melanocortin and antagonists agouti signaling protein and β‐defensin 3. J Invest Dermatol. 2012;132(9):2255–2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Brzoska T, Luger TA, Maaser C, Abels C, Böhm M. Alpha‐melanocyte‐stimulating hormone and related tripeptides: biochemistry, antiinflammatory and protective effects in vitro and in vivo, and future perspectives for the treatment of immune‐mediated inflammatory diseases. Endocr Rev. 2008;29(5):581–602. [DOI] [PubMed] [Google Scholar]
  • 20. Cui R, Widlund HR, Feige E, Lin JY, Wilensky DL, Igras VE, et al. Central role of p53 in the suntan response and pathologic hyperpigmentation. Cell. 2007;128(5):853–864. [DOI] [PubMed] [Google Scholar]
  • 21. Harno E, White A. Chapter 8—adrenocorticotropic hormone. In: Jameson JL, De Groot LJ, de Kretser DM, Giudice LC, Grossman AB, Melmed S, et al., editors. Endocrinology: adult and pediatric. 7th ed. Philadelphia: W.B. Saunders; 2016. p. 129–146.e5. [Google Scholar]
  • 22. Baldini G, Phelan KD. The melanocortin pathway and control of appetite‐progress and therapeutic implications. J Endocrinol. 2019;241(1):R1–R33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Swope VB, Abdel‐Malek ZA. Significance of the melanocortin 1 and endothelin B receptors in melanocyte homeostasis and prevention of sun‐induced genotoxicity. Front Genet. 2016;7:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Haddadeen C, Lai C, Cho SY, Healy E. Variants of the melanocortin‐1 receptor: do they matter clinically? Exp Dermatol. 2015;24(1):5–9. [DOI] [PubMed] [Google Scholar]
  • 25. Roh MR, Eliades P, Gupta S, Tsao H. Genetics of melanocytic nevi. Pigment Cell Melanoma Res. 2015;28(6):661–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Jarrett SG, D'Orazio JA. Hormonal regulation of the repair of UV photoproducts in melanocytes by the melanocortin signaling axis. Photochem Photobiol. 2017;93(1):245–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Morgan AM, Lo J, Fisher DE. How does pheomelanin synthesis contribute to melanomagenesis? Two distinct mechanisms could explain the carcinogenicity of pheomelanin synthesis. Bioessays. 2013;35(8):672–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Mitra D, Luo X, Morgan A, Wang J, Hoang MP, Lo J, et al. An ultraviolet‐radiation‐independent pathway to melanoma carcinogenesis in the red hair/fair skin background. Nature. 2012;491(7424):449–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Kokot A, Metze D, Mouchet N, Galibert MD, Schiller M, Luger TA, et al. Alpha‐melanocyte‐stimulating hormone counteracts the suppressive effect of UVB on Nrf2 and Nrf‐dependent gene expression in human skin. Endocrinology. 2009;150(7):3197–3206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Song X, Mosby N, Yang J, Xu A, Abdel‐Malek Z, Kadekaro AL. Alpha‐MSH activates immediate defense responses to UV‐induced oxidative stress in human melanocytes. Pigment Cell Melanoma Res. 2009;22(6):809–818. [DOI] [PubMed] [Google Scholar]
  • 31. Cadet J, Douki T, Ravanat JL. Oxidatively generated damage to cellular DNA by UVB and UVA radiation. Photochem Photobiol. 2015;91(1):140–155. [DOI] [PubMed] [Google Scholar]
  • 32. Hung KF, Sidorova JM, Nghiem P, Kawasumi M. The 6‐4 photoproduct is the trigger of UV‐induced replication blockage and ATR activation. Proc Natl Acad Sci U S A. 2020;117(23):12806–12816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Kadekaro AL, Leachman S, Kavanagh RJ, Swope V, Cassidy P, Supp D, et al. Melanocortin 1 receptor genotype: an important determinant of the damage response of melanocytes to ultraviolet radiation. FASEB J. 2010;24(10):3850–3860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Robinson S, Dixon S, August S, Diffey B, Wakamatsu K, Ito S, et al. Protection against UVR involves MC1R‐mediated non‐pigmentary and pigmentary mechanisms in vivo. J Invest Dermatol. 2010;130(7):1904–1913. [DOI] [PubMed] [Google Scholar]
  • 35. Böhm M, Wolff I, Scholzen TE, Robinson SJ, Healy E, Luger TA, et al. Alpha melanocyte‐stimulating hormone protects from ultraviolet radiation‐induced apoptosis and DNA damage. J Biol Chem. 2005;280(7):5795–5802. [DOI] [PubMed] [Google Scholar]
  • 36. Hauser JE, Kadekaro AL, Kavanagh RJ, Wakamatsu K, Terzieva S, Schwemberger S, et al. Melanin content and MC1R function independently affect UVR‐induced DNA damage in cultured human melanocytes. Pigment Cell Res. 2006;19(4):303–314. [DOI] [PubMed] [Google Scholar]
  • 37. Kadekaro AL, Kavanagh R, Kanto H, Terzieva S, Hauser J, Kobayashi N, et al. Alpha‐melanocortin and endothelin‐1 activate antiapoptotic pathways and reduce DNA damage in human melanocytes. Cancer Res. 2005;65(10):4292–4299. [DOI] [PubMed] [Google Scholar]
  • 38. Yu S, Doycheva DM, Gamdzyk M, Yang Y, Lenahan C, Li G, et al. Activation of MC1R with BMS‐470539 attenuates neuroinflammation via cAMP/PKA/Nurr1 pathway after neonatal hypoxic‐ischemic brain injury in rats. J Neuroinflammation. 2021;18(1):26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Maaser C, Kannengiesser K, Specht C, Lügering A, Brzoska T, Luger TA, et al. Crucial role of the melanocortin receptor MC1R in experimental colitis. Gut. 2006;55(10):1415–1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Wu X, Fu S, Liu Y, Luo H, Li F, Wang Y, et al. NDP‐MSH binding melanocortin‐1 receptor ameliorates neuroinflammation and BBB disruption through CREB/Nr4a1/NF‐κB pathway after intracerebral hemorrhage in mice. J Neuroinflammation. 2019;16(1):192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Böhm M, Apel M, Sugawara K, Brehler R, Jurk K, Luger TA, et al. Modulation of basophil activity: a novel function of the neuropeptide α‐melanocyte‐stimulating hormone. J Allergy Clin Immunol. 2012;129(4):1085–1093. [DOI] [PubMed] [Google Scholar]
  • 42. Loser K, Brzoska T, Oji V, Auriemma M, Voskort M, Kupas V, et al. The neuropeptide alpha‐melanocyte‐stimulating hormone is critically involved in the development of cytotoxic CD8+ T cells in mice and humans. PLoS One. 2010;5(2):e8958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Kanetsky PA, Hay JL. Marshaling the translational potential of MC1R for precision risk assessment of melanoma. Cancer Prev Res (Phila). 2018;11(3):121–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Pasquali E, García‐Borrón JC, Fargnoli MC, Gandini S, Maisonneuve P, Bagnardi V, et al. MC1R variants increased the risk of sporadic cutaneous melanoma in darker‐pigmented Caucasians: a pooled‐analysis from the M‐SKIP project. Int J Cancer. 2015;136(3):618–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Tagliabue E, Gandini S, García‐Borrón JC, Maisonneuve P, Newton‐Bishop J, Polsky D, et al. Association of melanocortin‐1 receptor variants with pigmentary traits in humans: a pooled analysis from the M‐Skip project. J Invest Dermatol. 2016;136(9):1914–1917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Beaumont KA, Liu YY, Sturm RA. The melanocortin‐1 receptor gene polymorphism and association with human skin cancer. Prog Mol Biol Transl Sci. 2009;88:85–153. [DOI] [PubMed] [Google Scholar]
  • 47. Castejón‐Griñán M, Herraiz C, Olivares C, Jiménez‐Cervantes C, García‐Borrón JC. cAMP‐independent non‐pigmentary actions of variant melanocortin 1 receptor: AKT‐mediated activation of protective responses to oxidative DNA damage. Oncogene. 2018;37(27):3631–3646. [DOI] [PubMed] [Google Scholar]
  • 48. Cerdido S, Sánchez‐Beltrán J, Lambertos A, Abrisqueta M, Padilla L, Herraiz C, et al. A side‐by‐side comparison of wildtype and variant melanocortin 1 receptor signaling with emphasis on protection against oxidative damage to DNA. Int J Mol Sci. 2023;24(18):14381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Herraiz C, Journé F, Abdel‐Malek Z, Ghanem G, Jiménez‐Cervantes C, García‐Borrón JC. Signaling from the human melanocortin 1 receptor to ERK1 and ERK2 mitogen‐activated protein kinases involves transactivation of cKIT. Mol Endocrinol. 2011;25(1):138–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Mosca S, Cardinali G, Flori E, Briganti S, Bottillo I, Mileo AM, et al. The PI3K pathway induced by αMSH exerts a negative feedback on melanogenesis and contributes to the release of pigment. Pigment Cell Melanoma Res. 2021;34(1):72–88. [DOI] [PubMed] [Google Scholar]
  • 51. Smalley K, Eisen T. The involvement of p38 mitogen‐activated protein kinase in the alpha‐melanocyte stimulating hormone (alpha‐MSH)‐induced melanogenic and anti‐proliferative effects in B16 murine melanoma cells. FEBS Lett. 2000;476(3):198–202. [DOI] [PubMed] [Google Scholar]
  • 52. Valverde P, Healy E, Jackson I, Rees JL, Thody AJ. Variants of the melanocyte‐stimulating hormone receptor gene are associated with red hair and fair skin in humans. Nat Genet. 1995;11(3):328–330. [DOI] [PubMed] [Google Scholar]
  • 53. Beaumont KA, Shekar SN, Newton RA, James MR, Stow JL, Duffy DL, et al. Receptor function, dominant negative activity and phenotype correlations for MC1R variant alleles. Hum Mol Genet. 2007;16(18):2249–2260. [DOI] [PubMed] [Google Scholar]
  • 54. Healy E, Flannagan N, Ray A, Todd C, Jackson IJ, Matthews JN, et al. Melanocortin‐1‐receptor gene and sun sensitivity in individuals without red hair. Lancet. 2000;355(9209):1072–1073. [DOI] [PubMed] [Google Scholar]
  • 55. Palmer JS, Duffy DL, Box NF, Aitken JF, O'Gorman LE, Green AC, et al. Melanocortin‐1 receptor polymorphisms and risk of melanoma: is the association explained solely by pigmentation phenotype? Am J Hum Genet. 2000;66(1):176–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Ringholm A, Klovins J, Rudzish R, Phillips S, Rees JL, Schiöth HB. Pharmacological characterization of loss of function mutations of the human melanocortin 1 receptor that are associated with red hair. J Invest Dermatol. 2004;123(5):917–923. [DOI] [PubMed] [Google Scholar]
  • 57. Scott MC, Wakamatsu K, Ito S, Kadekaro AL, Kobayashi N, Groden J, et al. Human melanocortin 1 receptor variants, receptor function and melanocyte response to UV radiation. J Cell Sci. 2002;115(Pt 11):2349–2355. [DOI] [PubMed] [Google Scholar]
  • 58. García‐Borrón JC, Sánchez‐Laorden BL, Jiménez‐Cervantes C. Melanocortin‐1 receptor structure and functional regulation. Pigment Cell Res. 2005;18(6):393–410. [DOI] [PubMed] [Google Scholar]
  • 59. Hennessy A, Oh C, Diffey B, Wakamatsu K, Ito S, Rees J. Eumelanin and pheomelanin concentrations in human epidermis before and after UVB irradiation. Pigment Cell Res. 2005;18(3):220–223. [DOI] [PubMed] [Google Scholar]
  • 60. VYLEESI (bremelanotide injection) [package insert]. Waltham, MA: AMAG Pharmaceuticals, Inc 2019.
  • 61. Bornstein SR, Allolio B, Arlt W, Barthel A, Don‐Wauchope A, Hammer GD, et al. Diagnosis and treatment of primary adrenal insufficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2016;101(2):364–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Collet TH, Dubern B, Mokrosinski J, Connors H, Keogh JM, Mendes de Oliveira E, et al. Evaluation of a melanocortin‐4 receptor (MC4R) agonist (setmelanotide) in MC4R deficiency. Mol Metab. 2017;6(10):1321–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Minder EI, Barman‐Aksoezen J, Schneider‐Yin X. Pharmacokinetics and pharmacodynamics of afamelanotide and its clinical use in treating dermatologic disorders. Clin Pharmacokinet. 2017;56(8):815–823. [DOI] [PubMed] [Google Scholar]
  • 64. Motosko CC, Bieber AK, Pomeranz MK, Stein JA, Martires KJ. Physiologic changes of pregnancy: a review of the literature. Int J Womens Dermatol. 2017;3(4):219–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Buscà R, Abbe P, Mantoux F, Aberdam E, Peyssonnaux C, Eychène A, et al. Ras mediates the cAMP‐dependent activation of extracellular signal‐regulated kinases (ERKs) in melanocytes. EMBO J. 2000;19(12):2900–2910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Herraiz C, Martínez‐Vicente I, Maresca V. The α‐melanocyte‐stimulating hormone/melanocortin‐1 receptor interaction: a driver of pleiotropic effects beyond pigmentation. Pigment Cell Melanoma Res. 2021;34(4):748–761. [DOI] [PubMed] [Google Scholar]
  • 67. Parkin DM, Mesher D, Sasieni P. 13. Cancers attributable to solar (ultraviolet) radiation exposure in the UK in 2010. Br J Cancer. 2011;105(suppl 2):S66–S69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Dzwierzynski WW. Melanoma risk factors and prevention. Clin Plast Surg. 2021;48(4):543–550. [DOI] [PubMed] [Google Scholar]
  • 69. Matthews NH, Li WQ, Qureshi AA, Weinstock MA, Cho E. Epidemiology of melanoma. In: Ward WH, Farma JM, editors. Cutaneous melanoma: etiology and therapy [Internet]. Brisbane, Australia: Codon Publications; 2017. [PubMed] [Google Scholar]
  • 70. Raimondi S, Sera F, Gandini S, Iodice S, Caini S, Maisonneuve P, et al. MC1R variants, melanoma and red hair color phenotype: a meta‐analysis. Int J Cancer. 2008;122(12):2753–2760. [DOI] [PubMed] [Google Scholar]
  • 71. Olbryt M. Molecular background of skin melanoma development and progression: therapeutic implications. Postepy Dermatol Alergol. 2019;36(2):129–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Berger MF, Garraway LA. Applications of genomics in melanoma oncogene discovery. Hematol Oncol Clin North Am. 2009;23(3):397–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Kiuru M, Busam KJ. The NF1 gene in tumor syndromes and melanoma. Lab Invest. 2017;97(2):146–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Potjer TP, Bollen S, Grimbergen A, van Doorn R, Gruis NA, van Asperen CJ, et al. Multigene panel sequencing of established and candidate melanoma susceptibility genes in a large cohort of Dutch non‐CDKN2A/CDK4 melanoma families. Int J Cancer. 2019;144(10):2453–2464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Cao J, Wan L, Hacker E, Dai X, Lenna S, Jimenez‐Cervantes C, et al. MC1R is a potent regulator of PTEN after UV exposure in melanocytes. Mol Cell. 2013;51(4):409–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Chelakkot VS, Thomas K, Romigh T, Fong A, Li L, Ronen S, et al. MC1R signaling through the cAMP‐CREB/ATF‐1 and ERK‐NFκB pathways accelerates G1/S transition promoting breast cancer progression. NPJ Precis Oncol. 2023;7(1):85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Guida S, Guida G, Goding CR. MC1R functions, expression, and implications for targeted therapy. J Invest Dermatol. 2022;142(2):293–302.e1. [DOI] [PubMed] [Google Scholar]
  • 78. Chen S, Han C, Miao X, Li X, Yin C, Zou J, et al. Targeting MC1R depalmitoylation to prevent melanomagenesis in redheads. Nat Commun. 2019;10(1):877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Huang J, Chan SC, Ko S, Lok V, Zhang L, Lin X, et al. Global incidence, mortality, risk factors and trends of melanoma: a systematic analysis of registries. Am J Clin Dermatol. 2023;24(6):965–975. [DOI] [PubMed] [Google Scholar]
  • 80. Ali Z, Yousaf N, Larkin J. Melanoma epidemiology, biology and prognosis. EJC Suppl. 2013;11(2):81–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Conforti C, Zalaudek I. Epidemiology and risk factors of melanoma: a review. Dermatol Pract Concept. 2021;11(suppl 1):e2021161S. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Sinikumpu SP, Jokelainen J, Keinänen‐Kiukaanniemi S, Huilaja L. Skin cancers and their risk factors in older persons: a population‐based study. BMC Geriatr. 2022;22(1):269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Liu C, Liu D, Wang F, Xie J, Liu Y, Wang H, et al. An intratumor heterogeneity‐related signature for predicting prognosis, immune landscape, and chemotherapy response in colon adenocarcinoma. Front Med (Lausanne). 2022;9:925661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Wang XQ, Xu SW, Wang W, Piao SZ, Mao XL, Zhou XB, et al. Identification and validation of a novel DNA damage and DNA repair related genes based signature for colon cancer prognosis. Front Genet. 2021;12:635863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Zhang N, Li Y, Sundquist J, Sundquist K, Ji J. Identifying actionable druggable targets for breast cancer: Mendelian randomization and population‐based analyses. EBioMedicine. 2023;98:104859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Maresca V, Flori E, Bellei B, Aspite N, Kovacs D, Picardo M. MC1R stimulation by alpha‐MSH induces catalase and promotes its re‐distribution to the cell periphery and dendrites. Pigment Cell Melanoma Res. 2010;23(2):263–275. [DOI] [PubMed] [Google Scholar]
  • 87. Bergthorsdottir R, Leonsson‐Zachrisson M, Odén A, Johannsson G. Premature mortality in patients with Addison's disease: a population‐based study. J Clin Endocrinol Metab. 2006;91(12):4849–4853. [DOI] [PubMed] [Google Scholar]
  • 88. Bensing S, Brandt L, Tabaroj F, Sjoberg O, Nilsson B, Ekbom A, et al. Increased death risk and altered cancer incidence pattern in patients with isolated or combined autoimmune primary adrenocortical insufficiency. Clin Endocrinol (Oxf). 2008;69(5):697–704. [DOI] [PubMed] [Google Scholar]
  • 89. Clark D, Thody AJ, Shuster S, Bowers H. Immunoreactive alpha‐MSH in human plasma in pregnancy. Nature. 1978;273(5658):163–164. [DOI] [PubMed] [Google Scholar]
  • 90. Jones MS, Lee J, Stern SL, Faries MB. Is pregnancy‐associated melanoma associated with adverse outcomes? J Am Coll Surg. 2017;225(1):149–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. European Medicines Agency . https://www.ema.europa.eu/en/documents/product‐information/scenesse‐epar‐product‐information_en.pdf. Accessed February 21, 2021.
  • 92. SCENESSE (afamelanotide) [package insert]. West Menlo Park, CA: Clinuvel, Inc. 2019.
  • 93. Minder EI. Afamelanotide, an agonistic analog of α‐melanocyte‐stimulating hormone, in dermal phototoxicity of erythropoietic protoporphyria. Expert Opin Investig Drugs. 2010;19(12):1591–1602. [DOI] [PubMed] [Google Scholar]
  • 94. European Medicines Agency . https://www.ema.europa.eu/en/medicines/human/EPAR/imcivree#product‐information‐section. Accessed February 21, 2021.
  • 95. IMCIVREE (setmelanotide) [package insert]. Boston, MA: Rhythm Pharmaceuticals, Inc. 2022.
  • 96. ACTHAR GEL (repository corticotropin injection) [package insert]. Bridgewater, NJ: Mallinckrodt ARD LLC. 2022.
  • 97. ClinicalTrials.gov . https://clinicaltrials.gov/study/NCT03511625. Accessed April 2, 2024.
  • 98. Huang YJ, Galen K, Zweifel B, Brooks LR, Wright AD. Distinct binding and signaling activity of Acthar gel compared to other melanocortin receptor agonists. J Recept Signal Transduct Res. 2021;41(5):425–433. [DOI] [PubMed] [Google Scholar]
  • 99. ClinicalTrials.gov . https://clinicaltrials.gov/study/NCT05466890. Accessed April 3, 2024.
  • 100. Garrido‐Mesa J, Thomas BL, Dodd J, Spana C, Perretti M, Montero‐Melendez T. Pro‐resolving and anti‐arthritic properties of the MC(1) selective agonist PL8177. Front Immunol. 2022;13:1078678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Ng TF, Dawit K, Taylor AW. Melanocortin receptor agonists suppress experimental autoimmune uveitis. Exp Eye Res. 2022;218:108986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. ClinicalTrials.gov . https://clinicaltrials.gov/study/NCT06239116. Accessed April 2, 2024.
  • 103. Koikov L, Starner RJ, Swope VB, Upadhyay P, Hashimoto Y, Freeman KT, et al. Development of hMC1R selective small agonists for sunless tanning and prevention of genotoxicity of UV in melanocytes. J Invest Dermatol. 2021;141(7):1819–1829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Sawyer TK, Sanfilippo PJ, Hruby VJ, Engel MH, Heward CB, Burnett JB, et al. 4‐Norleucine, 7‐D‐phenylalanine‐alpha‐melanocyte‐stimulating hormone: a highly potent alpha‐melanotropin with ultralong biological activity. Proc Natl Acad Sci U S A. 1980;77(10):5754–5758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Biolcati G, Marchesini E, Sorge F, Barbieri L, Schneider‐Yin X, Minder EI. Long‐term observational study of afamelanotide in 115 patients with erythropoietic protoporphyria. Br J Dermatol. 2015;172(6):1601–1612. [DOI] [PubMed] [Google Scholar]
  • 106. Langendonk JG, Balwani M, Anderson KE, Bonkovsky HL, Anstey AV, Bissell DM, et al. Afamelanotide for Erythropoietic protoporphyria. N Engl J Med. 2015;373(1):48–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Haylett AK, Nie Z, Brownrigg M, Taylor R, Rhodes LE. Systemic photoprotection in solar urticaria with α‐melanocyte‐stimulating hormone analogue [Nle4‐D‐Phe7]‐α‐MSH. Br J Dermatol. 2011;164(2):407–414. [DOI] [PubMed] [Google Scholar]
  • 108. Böhm M, Ehrchen J, Luger TA. Beneficial effects of the melanocortin analogue Nle4‐D‐Phe7‐α‐MSH in acne vulgaris. J Eur Acad Dermatol Venereol. 2014;28(1):108–111. [DOI] [PubMed] [Google Scholar]
  • 109. Barnetson RS, Ooi TK, Zhuang L, Halliday GM, Reid CM, Walker PC, et al. [Nle4‐D‐Phe7]‐alpha‐melanocyte‐stimulating hormone significantly increased pigmentation and decreased UV damage in fair‐skinned Caucasian volunteers. J Invest Dermatol. 2006;126(8):1869–1878. [DOI] [PubMed] [Google Scholar]
  • 110. Clinuvel . https://www.clinuvel.com/wp‐content/uploads/2020/11/CLINUVEL‐STRATEGIC‐UPDATE.pdf. Accessed June 7, 2023.
  • 111. Center for Drug Evaluation and Research . Application number: 210557Orig1s000 other reviews (Bremelanotide). Silver Spring, MD: Department of Health and Human Services, Food and Drug Administration; 2019. [Google Scholar]
  • 112. Medicines and Healthare Products Regulatory Agency . https://www.medicines.org.uk/emc/product/14068/smpc. Accessed April 20, 2023.
  • 113. Haqq AM, Chung WK, Dollfus H, Haws RM, Martos‐Moreno G, Poitou C, et al. Efficacy and safety of setmelanotide, a melanocortin‐4 receptor agonist, in patients with Bardet‐Biedl syndrome and Alström syndrome: a multicentre, randomised, double‐blind, placebo‐controlled, phase 3 trial with an open‐label period. Lancet Diabetes Endocrinol. 2022;10(12):859–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Haws R, Brady S, Davis E, Fletty K, Yuan G, Gordon G, et al. Effect of setmelanotide, a melanocortin‐4 receptor agonist, on obesity in Bardet‐Biedl syndrome. Diabetes Obes Metab. 2020;22(11):2133–2140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Clément K, van den Akker E, Argente J, Bahm A, Chung WK, Connors H, et al. Efficacy and safety of setmelanotide, an MC4R agonist, in individuals with severe obesity due to LEPR or POMC deficiency: single‐arm, open‐label, multicentre, phase 3 trials. Lancet Diabetes Endocrinol. 2020;8(12):960–970. [DOI] [PubMed] [Google Scholar]
  • 116. Clément K, van dan Akker E, Gordon G, Yuan G, Kühnen P. Timing of onset of adverse events with setmelanotide, an MC4R agonist, in patients with severe obesity due to LEPR or POMC deficiency. Poster Presented at: The Endocrine Society Annual Meeting; March 20–23, 2021; virtual.
  • 117. Clément K, Biebermann H, Farooqi IS, Van der Ploeg L, Wolters B, Poitou C, et al. MC4R agonism promotes durable weight loss in patients with leptin receptor deficiency. Nat Med. 2018;24(5):551–555. [DOI] [PubMed] [Google Scholar]
  • 118. Kanti V, Puder L, Jahnke I, Krabusch PM, Kottner J, Vogt A, et al. A melanocortin‐4 receptor agonist induces skin and hair pigmentation in patients with monogenic mutations in the leptin‐melanocortin pathway. Skin Pharmacol Physiol. 2021;34(6):307–316. [DOI] [PubMed] [Google Scholar]
  • 119. Kühnen P, Clément K, Wiegand S, Blankenstein O, Gottesdiener K, Martini LL, et al. Proopiomelanocortin deficiency treated with a melanocortin‐4 receptor agonist. N Engl J Med. 2016;375(3):240–246. [DOI] [PubMed] [Google Scholar]
  • 120. Kühnen P, Clément K. Long‐term MC4R agonist treatment in POMC‐deficient patients. N Engl J Med. 2022;387(9):852–854. [DOI] [PubMed] [Google Scholar]
  • 121. Callaghan DJ 3rd. A glimpse into the underground market of melanotan. Dermatol Online J. 2018;24(5):13030. [PubMed] [Google Scholar]
  • 122. Dorr RT, Lines R, Levine N, Brooks C, Xiang L, Hruby VJ, et al. Evaluation of melanotan‐II, a superpotent cyclic melanotropic peptide in a pilot phase‐I clinical study. Life Sci. 1996;58(20):1777–1784. [DOI] [PubMed] [Google Scholar]
  • 123. Cardones AR, Grichnik JM. Alpha‐melanocyte‐stimulating hormone‐induced eruptive nevi. Arch Dermatol. 2009;145(4):441–444. [DOI] [PubMed] [Google Scholar]
  • 124. Wessells H, Levine N, Hadley ME, Dorr R, Hruby V. Melanocortin receptor agonists, penile erection, and sexual motivation: human studies with melanotan II. Int J Impot Res. 2000;12(suppl):4S74–4S79. [DOI] [PubMed] [Google Scholar]
  • 125. Böhm M, Jagirdar K, Sturm RA, König S, Bauer J, Metze D, et al. Lack of protection from development of multiple melanomas by an injected melanocortin analogue in a combined high‐risk MC1R/CDKN2A genotype patient. J Eur Acad Dermatol Venereol. 2016;30(10):e65–e67. [DOI] [PubMed] [Google Scholar]
  • 126. Peters B, Hadimeri H, Wahlberg R, Afghahi H. Melanotan II: a possible cause of renal infarction: review of the literature and case report. CEN Case Rep. 2020;9(2):159–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Mallalieu JE. Melanoma in‐situ associated with melanotan II use. J Case Rep Clin Med. 2020;3(1):147. [Google Scholar]
  • 128. Paurobally D, Jason F, Dezfoulian B, Nikkels AF. Melanotan‐associated melanoma. Br J Dermatol. 2011;164(6):1403–1405. [DOI] [PubMed] [Google Scholar]
  • 129. Hjuler KF, Lorentzen HF. Melanoma associated with the use of melanotan‐II. Dermatology. 2014;228(1):34–36. [DOI] [PubMed] [Google Scholar]
  • 130. Ong S, Bowling J. Melanotan‐associated melanoma in situ. Australas J Dermatol. 2012;53(4):301–302. [DOI] [PubMed] [Google Scholar]
  • 131. Sivyer GW. Changes of melanocytic lesions inducedby Melanotan injections and sun bed use in a teenage patient with FAMMM syndrome. Dermatol Pract Concept. 2012;2(3):203a10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Abdel‐Malek ZA, Kadekaro AL, Kavanagh RJ, Todorovic A, Koikov LN, McNulty JC, et al. Melanoma prevention strategy based on using tetrapeptide alpha‐MSH analogs that protect human melanocytes from UV‐induced DNA damage and cytotoxicity. FASEB J. 2006;20(9):1561–1563. [DOI] [PubMed] [Google Scholar]
  • 133. Abdel‐Malek ZA, Ruwe A, Kavanagh‐Starner R, Kadekaro AL, Swope V, Haskell‐Luevano C, et al. Alpha‐MSH tripeptide analogs activate the melanocortin 1 receptor and reduce UV‐induced DNA damage in human melanocytes. Pigment Cell Melanoma Res. 2009;22(5):635–644. [DOI] [PubMed] [Google Scholar]
  • 134. Bettenworth D, Buyse M, Böhm M, Mennigen R, Czorniak I, Kannengiesser K, et al. The tripeptide KdPT protects from intestinal inflammation and maintains intestinal barrier function. Am J Pathol. 2011;179(3):1230–1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Mastrofrancesco A, Kokot A, Eberle A, Gibbons NC, Schallreuter KU, Strozyk E, et al. KdPT, a tripeptide derivative of alpha‐melanocyte‐stimulating hormone, suppresses IL‐1 beta‐mediated cytokine expression and signaling in human sebocytes. J Immunol. 2010;185(3):1903–1911. [DOI] [PubMed] [Google Scholar]
  • 136. Kucharzik T, Lemmnitz G, Abels C, Maaser C. Tripeptide K(D)PT is well tolerated in mild‐to‐moderate ulcerative colitis: results from a randomized multicenter study. Inflamm Bowel Dis. 2017;23(2):261–271. [DOI] [PubMed] [Google Scholar]
  • 137. Kondo M, Suzuki T, Kawano Y, Kojima S, Miyashiro M, Matsumoto A, et al. Dersimelagon, a novel oral melanocortin 1 receptor agonist, demonstrates disease‐modifying effects in preclinical models of systemic sclerosis. Arthritis Res Ther. 2022;24(1):210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Suzuki T, Kawano Y, Matsumoto A, Kondo M, Funayama K, Tanemura S, et al. Melanogenic effect of dersimelagon (MT‐7117), a novel oral melanocortin 1 receptor agonist. Skin Health Dis. 2022;2(1):e78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Balwani M, Bonkovsky HL, Levy C, Anderson KE, Bissell DM, Parker C, et al. Dersimelagon in erythropoietic protoporphyrias. N Engl J Med. 2023;388(15):1376–1385. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing not applicable to this article as no data sets were generated or analysed during the current study.


Articles from Journal of the European Academy of Dermatology and Venereology are provided here courtesy of Wiley

RESOURCES