Melanoma is a malignant tumor of melanocytes, which are the cells that make the pigment melanin and are derived from the neural crest. Although most melanomas arise in the skin, they may also arise from mucosal surfaces or at other sites to which neural crest cells migrate, including the uveal tract. Uveal melanomas differ significantly from cutaneous melanoma in incidence, prognostic factors, molecular characteristics, and treatment. For more information, see Intraocular (Uveal) Melanoma Treatment.
Estimated new cases and deaths from melanoma in the United States in 2024:[1]
Skin cancer is the most common malignancy diagnosed in the United States, and invasive melanoma represents about 1% of skin cancers but results in the most deaths.[1,2] Since the early 2000s, the incidence of melanoma in people younger than 50 years declined by about 1% per year in men and stabilized in women. In people aged 50 years and older, incidence stabilized in men and increased by about 3% per year in women.[1] Older men are at highest risk; however, melanoma is the most common cancer in young adults aged 25 to 29 years and the second most common cancer in those aged 15 to 29 years.[3] Ocular melanoma is the most common cancer of the eye, with approximately 2,000 cases diagnosed annually.
Risk factors for melanoma include both intrinsic (genetic and phenotype) and extrinsic (environmental or exposure) factors:
For more information about risk factors, see Skin Cancer Prevention and Genetics of Skin Cancer.
For more information, see Skin Cancer Screening.
Melanoma occurs predominantly in adults, and more than 50% of the cases arise in apparently normal areas of the skin. Although melanoma can occur anywhere, including on mucosal surfaces and the uvea, in women it occurs more commonly on the extremities, and in men it occurs most commonly on the trunk or head and neck.[4]
Early signs in a nevus that would suggest a malignant change include the following:
A biopsy, preferably by local excision, should be performed for any suspicious lesions. Suspicious lesions should never be shaved off or cauterized. An experienced pathologist should examine the specimens to allow for microstaging.
Studies show that distinguishing between benign pigmented lesions and early melanomas can be difficult, and even experienced dermatopathologists can have differing opinions. To reduce the possibility of misdiagnosis for an individual patient, a second review by an independent qualified pathologist should be considered.[5,6] Agreement between pathologists in the histological diagnosis of melanomas and benign pigmented lesions has been found to be considerably variable.[5,6]
Evidence (discordance in histological evaluation):
Prognosis is affected by the characteristics of primary and metastatic tumors. The most important prognostic factors have been incorporated into the revised 2009 American Joint Committee on Cancer staging and include the following:[4,7-9]
Patients who are younger, who are female, and who have melanomas on their extremities generally have better prognoses.[4,7-9]
Microscopic satellites, recorded as present or absent, in stage I melanoma may be a poor prognostic histological factor, but this is controversial.[10] The presence of tumor infiltrating lymphocytes, which may be categorized as brisk, nonbrisk, or absent, is under study as a potential prognostic factor.[11]
The risk of relapse decreases substantially over time, although late relapses are not uncommon.[12,13]
The descriptive terms for clinicopathological cellular subtypes of malignant melanoma should be considered of historic interest only; they do not have independent prognostic or therapeutic significance. The cellular subtypes are the following:
The Cancer Genome Atlas (TCGA) Network performed an integrative multiplatform characterization of 333 cutaneous melanomas from 331 patients.[1] Using six types of molecular analysis at the DNA, RNA, and protein levels, the researchers identified four major genomic subtypes:
Genomic subtypes may suggest drug targets and clinical trial design, as well as guide clinical decision-making for targeted therapies. For more information, see Table 1.
To date, targeted therapies have demonstrated efficacy and received U.S. Food and Drug Administration approval only for the BRAF-mutant subtype of melanoma. Combination therapies with a BRAF plus a MEK inhibitor have shown improvement in outcomes over a single-agent inhibitor alone; yet, virtually all patients acquire resistance to therapy and relapse. Therefore, clinical trials remain an important option for patients with BRAF-mutant subtype, as well as other genomic subtypes of melanoma. See the individual treatment sections for more information.
A variety of immunotherapies have been approved for the treatment of melanoma regardless of genetic subtype. The benefit of immunotherapy has not been associated with a specific mutation or molecular subtype. The TCGA analysis identified immune markers (in a subset within each molecular subtype) that were associated with improved survival and that may have implications for immunotherapy. Identification of predictive biomarkers remains an active area of research. See the individual treatment sections for more information.
Genomic Subtype | % Samples With Mutation | Increased Lymphocytic Infiltration (%) | Clinical Management Implications for Targeted Therapyc,d | ||
---|---|---|---|---|---|
FDA = U.S. Food and Drug Administration; WT = wild-type. | |||||
aPrimary melanoma with matched normal samples; N = 67 (20%). Metastatic melanoma with matched normal samples; N = 266 (80%). Matched is defined as sample from the same patient. | |||||
bTriple WT was defined as a heterogeneous subgroup lacking BRAF, NRAS, HRAS, and KRAS, and NF1 mutations. | |||||
cThe indications for immunotherapy are not known to be determined or limited by genomic subtype. | |||||
dRisks and benefits of single versus combination therapies are detailed in the Treatment Option Overview for Melanoma section of this summary. | |||||
eResearch includes but is not limited to these examples. Clinical trials are posted on clinicaltrials.gov. | |||||
fIndicated when mutation is diagnosed by an FDA-approved assay. | |||||
FDA Approved | Researche (single agent or in combination) | ||||
BRAF mutant | 52 | ~ 30 | BRAF inhibitorsf | CDK inhibitors, PI3K/Akt/mTOR inhibitors, ERK inhibitors, IDH1 inhibitors, EZH2 inhibitors, Aurora kinase inhibitors, ARID2 chromatin remodelers | |
– Vemurafenib | |||||
–Dabrafenib | |||||
MEK inhibitors | |||||
–Trametinib | |||||
–Cobimetinib | |||||
Combination of BRAF + MEK inhibitors | |||||
–Vemurafenib + cobimetinib | |||||
–Dabrafenib + trametinib | |||||
RAS mutant (NRAS, HRAS, and KRAS ) | 28 | ~ 25 | MEK inhibitors, CDK inhibitors, PI3K/Akt/mTOR inhibitors, ERK inhibitors, IDH1 inhibitors, EZH2 inhibitors, Aurora kinase inhibitors, ARID2 chromatin remodelers | ||
NF1 mutant | 14 | ~ 25 | PI3K/Akt/mTOR inhibitors, ERK inhibitors, IDH1 inhibitors, EZH2 inhibitors, ARID2 chromatin remodelers | ||
Triple WTb | 14.5 | ~ 40 | KIT-mutated/amplified CDK inhibitors (i.e., imatinib and dasatinib), MDM2/p53 interaction inhibitors, PI3K/Akt/mTOR inhibitors, IDH1 inhibitors, EZH2 inhibitors |
Uveal melanomas differ significantly from cutaneous melanomas. ln one series, 83% of 186 uveal melanomas were found to have a constitutively active somatic mutation in GNAQ or GNA11.[2,3] For more information, see Intraocular (Uveal) Melanoma Treatment.
Clinical staging is based on whether the tumor has spread to regional lymph nodes or distant sites. For melanoma that is clinically confined to the primary site, the chance of lymph node or systemic metastases increases as the thickness and depth of local invasion increases, which worsens the prognosis. Melanoma can spread by local extension (through lymphatics) and/or by hematogenous routes to distant sites. Any organ may be involved by metastases, but the lungs and liver are common sites.
The microstage of malignant melanoma is determined on histological examination by the vertical thickness of the lesion in millimeters (Breslow classification) and/or the anatomical level of local invasion (Clark classification). The Breslow thickness is more reproducible and more accurately predicts subsequent behavior of malignant melanoma in lesions thicker than 1.5 mm and should always be reported.
Accurate microstaging of the primary tumor requires careful histological evaluation of the entire specimen by an experienced pathologist.
Level of Invasion | Description |
---|---|
Level I | Lesions involving only the epidermis (in situ melanoma); not an invasive lesion. |
Level II | Invasion of the papillary dermis; does not reach the papillary-reticular dermal interface. |
Level III | Invasion fills and expands the papillary dermis but does not penetrate the reticular dermis. |
Level IV | Invasion into the reticular dermis but not into the subcutaneous tissue. |
Level V | Invasion through the reticular dermis into the subcutaneous tissue. |
The American Joint Committee on Cancer (AJCC) has designated staging by TNM (tumor, node, metastasis) classification to define melanoma.[1]
Cancers staged using this staging system include cutaneous melanoma. Cancers not staged using this system include melanoma of the conjunctiva; melanoma of the uvea; mucosal melanoma arising in the head and neck; mucosal melanoma of the urethra, vagina, rectum, and anus; Merkel cell carcinoma; and squamous cell carcinoma.[1]
AJCC Prognostic Stage Groups-Clinical (cTNM)
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) |
---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; c = clinical; LDH = lactate dehydrogenase; No. = number. | ||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | ||||
bThickness and ulceration status not applicable. | ||||
0 | Tis, N0, M0 | Tis = Melanoma in situ.b | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) |
---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; c = clinical; LDH = lactate dehydrogenase; No. = number. | ||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | ||||
IA | T1a, N0, M0 | T1a = <0.8 mm/without ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
IB | T1b, N0, M0 | T1b = <0.8 mm with ulceration; 0.8–1.0 mm with or without ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
T2a, N0, M0 | T2a = >1.0–2.0 mm/without ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) |
---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; c = clinical; LDH = lactate dehydrogenase; No. = number. | ||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | ||||
IIA | T2b, N0, M0 | T2b = >1.0–2.0 mm/with ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
T3a, N0, M0 | T3a = >2.0–4.0 mm/without ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. | |
IIB | T3b, N0, M0 | T3b = >2.0–4.0 mm/with ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
T4a, N0, M0 | T4a = >4.0 mm/without ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. | |
IIC | T4b, N0, M0 | T4b = >4.0 mm/with ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH) |
---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; c = clinical; LDH = lactate dehydrogenase; No. = number. | ||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | ||||
bFor example, diagnosis by curettage. | ||||
cFor example, unknown primary or completely regressed melanoma. | ||||
dThickness and ulceration status not applicable. | ||||
eDetected by sentinel lymph node biopsy. | ||||
III | Any T, Tis, ≥N1, M0 | TX = Primary tumor cannot be assessed.b,d | N1a = One clinically occult nodee /in-transit, satellite, and/or microsatellite metastases not present. | M0 = No evidence of distant metastasis. |
T0 = No evidence of primary tumor.c,d | ||||
Tis = Melanoma in situ.d | N1b = One clinically detected node/in-transit, satellite, and/or microsatellite metastases not present. | |||
T1a = <0.8 mm/without ulceration. | N1c = No regional lymph node disease/in-transit, satellite, and/or microsatellite metastases present. | |||
T1b = <0.8 mm with ulceration; 0.8–1.0 mm with or without ulceration. | N2a = Two or three clinically occult nodese/in-transit, satellite, and/or microsatellite metastases not present. | |||
T2a = >1.0–2.0 mm/without ulceration. | N2b = Two or three nodes at least one of which was clinically detected/in-transit, satellite, and or microsatellite metastases not present. | |||
T2b = >1.0–2.0 mm/with ulceration. | N2c = One clinically occult or clinically detected node/in-transit, satellite, and/or microsatellite metastases present. | |||
T3a = >2.0–4.0 mm/without ulceration. | N3a = Four or more clinically occult nodese/in-transit, satellite, and/or microsatellite metastases not present. | |||
T3b = >2.0–4.0 mm/with ulceration. | N3b = Four or more nodes, at least one of which was clinically detected, or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases not present. | |||
T4a = >4.0 mm/without ulceration. | N3c = Two or more clinically occult or clinically detected nodes and/or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases present. | |||
T4b = >4.0 mm/with ulceration. |
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) |
---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; c = clinical; CNS = central nervous system; LDH = lactate dehydrogenase; No. = number. | ||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | ||||
bFor example, sentinel lymph node biopsy not performed, regional nodes previously removed for another reason. (Exception: pathological N category is not required for T1 melanomas, use cN). | ||||
IV | Any T, Any N, M1 | Any T = See Table 6 for description. | NX = Regional nodes not assessed;b N0 = No regional metastases; ≥N1 = See Table 6 for description. | M1 = Evidence of distant metastasis. |
–M1a = Distant metastasis to skin, soft tissue including muscle, and/or nonregional lymph nodes [M1a(0) = LDH not elevated; M1a(1) = LDH elevated]. | ||||
–M1b = Distant metastasis to lung with or without M1a sites of disease [M1b(0) = LDH not elevated; M1b(1) = LDH elevated]. | ||||
–M1c = Distant metastasis to non-CNS visceral sites with or without M1a or M1b sites of disease [M1c(0) = LDH not elevated; OR M1c(1) = LDH elevated]. | ||||
–M1d = Distant metastasis to CNS with or without M1a, M1b, or M1c sites of disease [M1d(0) = LDH not elevated; M1d(1) = LDH elevated]. |
AJCC Prognostic Stage Groups-Pathological (pTNM)
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) | Illustration |
---|---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; cN = clinical N; LDH = lactate dehydrogenase; No. = number; p = pathological. | |||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | |||||
bPathological stage 0 (melanoma in situ) and T1 do not require pathological evaluation of lymph nodes to complete pathological staging; use cN information to assign their pathological stage. | |||||
cThickness and ulceration status not applicable. | |||||
0 | Tis, N0, M0 | Tis = Melanoma in situ.b,c | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) | Illustration |
---|---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; cN = clinical N; LDH = lactate dehydrogenase; No. = number; p = pathological. | |||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | |||||
bPathological stage 0 (melanoma in situ) and T1 do not require pathological evaluation of lymph nodes to complete pathological staging; use cN information to assign their pathological stage. | |||||
IA | T1a, N0, M0 | T1a = <0.8 mm/without ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. | |
T1b, N0, M0 | T1b = <0.8 mm with ulceration; 0.8–1.0 mm with or without ulceration. | ||||
IB | T2a, N0, M0 | T2a = >1.0–2.0 mm/without ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) | Illustration |
---|---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; LDH = lactate dehydrogenase; No. = number; p = pathological. | |||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | |||||
IIA | T2b, N0, M0 | T2b = >1.0–2.0 mm/with ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. | |
T3a, N0, M0 | T3a = >2.0–4.0 mm/without ulceration. | ||||
IIB | T3b, N0, M0 | T3b = >2.0–4.0 mm/with ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. | |
T4a, N0, M0 | T4a = >4.0 mm/without ulceration. | ||||
IIC | T4b, N0, M0 | T4b = >4.0 mm/with ulceration. | N0 = No regional metastases detected. | M0 = No evidence of distant metastasis. |
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) |
---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; LDH = lactate dehydrogenase; No. = number; p = pathological. | ||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | ||||
bDetected by sentinel lymph node biopsy. | ||||
cFor example, unknown primary or completely regressed melanoma. | ||||
dThickness and ulceration status not applicable. | ||||
IIIA | T1a/b–T2a, N1a or N2a, M0 | T1a = <0.8 mm/without ulceration/T1b = <0.8 mm with ulceration; 0.8–1.0 mm with or without ulceration. | N1a = One clinically occult nodeb/in-transit, satellite, and/or microsatellite metastases not present; OR N2a = Two or three clinically occult nodesb/in-transit, satellite, and/or microsatellite metastases not present. | M0 = No evidence of distant metastasis. |
T2a = >1.0–2.0 mm/without ulceration. | ||||
IIIB | T0, N1b, N1c, M0 | T0 = No evidence of primary tumor.c,d | N1b = One clinically detected node/in-transit, satellite, and/or microsatellite metastases not present. | M0 = No evidence of distant metastasis. |
N1c = No regional lymph node disease/in-transit, satellite, and/or microsatellite metastases present. | ||||
T1a/b–T2a, N1b/c or N2b, M0 | T1a = <0.8 mm/without ulceration/T1b <0.8 mm with ulceration; 0.8–1.0 mm with or without ulceration. | N1b = One clinically detected node/in-transit, satellite, and/or microsatellite metastases not present;/N1c = No regional lymph node disease/in-transit, satellite, and/or microsatellite metastases present; OR | M0 = No evidence of distant metastasis. | |
T2a = >1.0–2.0 mm/without ulceration. | ||||
N2b = Two or three nodes at least one of which was clinically detected/in-transit, satellite, and or microsatellite metastases not present. | ||||
T2b/T3a, N1a–N2b, M0 | T2b = >1.0–2.0 mm/with ulceration/T3a = >2.0–4.0 mm/without ulceration. | N1a = One clinically occult nodeb/in-transit, satellite, and/or microsatellite metastases not present. | M0 = No evidence of distant metastasis. | |
N1b = One clinically detected node/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N1c = No regional lymph node disease/in-transit, satellite, and/or microsatellite metastases present. | ||||
N2a = Two or three clinically occult nodesb/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N2b = Two or three nodes, at least one of which was clinically detected/in-transit, satellite, and/or microsatellite metastases not present. | ||||
IIIC | T0, N2b, N2c, N3b, or N3c, M0 | T0 = No evidence of primary tumor.c,d | N2b = Two or three nodes, at least one of which was clinically detected/in-transit, satellite, and/or microsatellite metastases not present. | M0 = No evidence of distant metastasis. |
N2c = One clinically occult or clinically detected node/in-transit, satellite, and/or microsatellite metastases present. | ||||
N3b = Four or more nodes, at least one of which was clinically detected, or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases not present; OR | ||||
N3c = Two or more clinically occult or clinically detected nodes and/or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases present. | ||||
T1a–T3a, N2c or N3a/b/c, M0 | T1a = <0.8 mm/without ulceration/T1b = <0.8 mm with ulceration; 0.8–1.0 mm with or without ulceration. | N2c = One clinically occult or clinically detected node/in-transit, satellite, and/or microsatellite metastases present; OR | M0 = No evidence of distant metastasis. | |
T2a = >1.0–2.0 mm/without ulceration. | ||||
T2b = >1.0–2.0 mm/with ulceration. | ||||
N3a = Four or more clinically occult nodesb/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N3b = Four or more nodes, at least one of which was clinically detected, or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases not present. | ||||
T3a = >2.0–4.0 mm/without ulceration. | N3c = Two or more clinically occult or clinically detected nodes and/or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases present. | |||
T3b/T4a, Any N ≥N1, M0 | T3b = >2.0–4.0 mm/with ulceration/T4a = >4.0 mm/without ulceration. | N1a = One clinically occult nodeb/in-transit, satellite, and/or microsatellite metastases not present. | M0 = No evidence of distant metastasis. | |
N1b = One clinically detected node/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N1c = No regional lymph node disease/in-transit, satellite, and/or microsatellite metastases present. | ||||
N2a = Two or three clinically occult nodesb/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N2b = Two or three nodes, at least one of which was clinically detected/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N2c = One clinically occult or clinically detected node/in-transit, satellite, and/or microsatellite metastases present. | ||||
N3a = Four or more clinically occult nodesb/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N3b = Four or more nodes, at least one of which was clinically detected, or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N3c = Two or more clinically occult or clinically detected nodes and/or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases present. | ||||
T4b, N1a–N2c, M0 | T4b = >4.0 mm/with ulceration. | N1a = One clinically occult nodeb/in-transit, satellite, and/or microsatellite metastases not present. | M0 = No evidence of distant metastasis. | |
N1b = One clinically detected node/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N1c = No regional lymph node disease/in-transit, satellite, and/or microsatellite metastases present. | ||||
N2a = Two or three clinically occult nodesb/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N2b = Two or three nodes, at least one of which was clinically detected/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N2c = One clinically occult or clinically detected node/in-transit, satellite, and/or microsatellite metastases present. | ||||
IIID | T4b, N3a/b/c, M0 | T4b = >4.0 mm/with ulceration. | N3a = Four or more clinically occult nodesb/in-transit, satellite, and/or microsatellite metastases not present. | M0 = No evidence of distant metastasis. |
N3b = Four or more nodes, at least one of which was clinically detected, or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases not present. | ||||
N3c = Two or more clinically occult or clinically detected nodes and/or presence of any number of matted nodes/in-transit, satellite, and/or microsatellite metastases present. |
Stage | TNM | T Category (Thickness/Ulceration Status) | N Category (No. of Tumor-Involved Regional Lymph Nodes/Presence of In-Transit, Satellite, and/or Microsatellite Metastases) | M Category (Anatomic Site/LDH Level) | Illustration |
---|---|---|---|---|---|
T = primary tumor; N = regional lymph node; M = distant metastasis; cN = clinical N; CNS = central nervous system; LDH = lactate dehydrogenase; No. = number; p = pathological. | |||||
aAdapted from AJCC: Melanoma of the Skin. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 563–85. | |||||
bFor example, sentinel lymph node biopsy not performed, regional nodes previously removed for another reason. (Exception: pathological N category is not required for T1 melanomas, use cN). | |||||
cPathological stage 0 (melanoma in situ) and T1 do not require pathological evaluation of lymph nodes to complete pathological staging; use cN information to assign their pathological stage. | |||||
dThickness and ulceration status not applicable. | |||||
IV | Any T, Tis, Any N, M1 | Any T = See Table 6 for description. | NX = Regional nodes not assessed;d N0 = No regional metastases; ≥N1 = See Table 6 for description. | M1 = Evidence of distant metastasis. | |
Tis = Melanoma in situ.b,c | –M1a = Distant metastasis to skin, soft tissue including muscle, and/or nonregional lymph nodes [M1a(0) = LDH not elevated; M1a(1) = LDH elevated]. | ||||
–M1b = Distant metastasis to lung with or without M1a sites of disease [M1b(0) = LDH not elevated; M1b(1) = LDH elevated]. | |||||
–M1c - Distant metastasis to non-CNS visceral sites with or without M1a or M1b sites of disease [M1c(0) = LDH not elevated; M1c(1) = LDH elevated]. | |||||
–M1d = Distant metastasis to CNS with or without M1a, M1b, or M1c sites of disease [M1d(0) = LDH not elevated; M1d(1) = LDH elevated]. |
Stage (TNM Staging Criteria) | Standard Treatment Optionsa |
---|---|
aClinical trials are an important option for patients with all stages of melanoma because advances in understanding the aberrant molecular and biological pathways have led to rapid drug development. Standard treatment options are available in many clinical trials. Information about ongoing clinical trials is available from the NCI website. | |
Stage 0 melanoma | Excision |
Stage I melanoma | Excision +/− lymph node management |
Stage II melanoma | Excision +/− lymph node management |
Resectable Stage III melanoma | Excision +/− lymph node management |
Unresectable Stage III, Stage IV, and Recurrent melanoma | Intralesional therapy |
Immunotherapy | |
Signal transduction inhibitors | |
Chemotherapy | |
Palliative local therapy |
Surgical excision remains the primary modality for treating melanoma. Cutaneous melanomas that have not spread beyond the initial site are highly curable. The treatment for localized melanoma is surgical excision with margins proportional to the microstage of the primary lesion.
Lymphatic mapping and SLNB can be considered to assess the presence of occult metastasis in the regional lymph nodes of patients with primary tumors larger than 1 to 4 mm, potentially identifying individuals who may be spared the morbidity of regional lymph node dissections and individuals who may benefit from adjuvant therapy.[1-6]
To ensure accurate identification of the sentinel lymph node, lymphatic mapping and removal of the SLN should precede wide excision of the primary melanoma.
Multiple studies have demonstrated the diagnostic accuracy of SLNB, with false-negative rates of 0% to 2%.[1,6-11] If metastatic melanoma is detected, a complete regional lymphadenectomy can be performed in a second procedure.
Patients can be considered for CLND if the sentinel node(s) is microscopically or macroscopically positive for regional control or considered for entry into the Multicenter Selective Lymphadenectomy Trial II (NCT00297895) to determine whether CLND affects survival. SLNB should be performed before wide excision of the primary melanoma to ensure accurate lymphatic mapping.
Adjuvant therapy options are expanding for patients at high risk of recurrence after complete resection and include checkpoint inhibitors and combination signal transduction inhibitor therapy. Ipilimumab was the first checkpoint inhibitor to be approved by the U.S. Food and Drug Administration (FDA) as adjuvant therapy, and it has demonstrated improved overall survival (OS) at 10 mg/kg (ipi10) compared with placebo (EORTC 18071 [NCT00636168]).[12] However, ipi10 has significant toxicity at this dose. The North American Intergroup Trial E1609 (NCT01274338), designed with three treatment groups, compared ipi10 with ipilimumab at a lower dose of 3 mg/kg (ipi3) (approved for metastatic melanoma) and with high-dose interferon (HDI). Ipi3 showed a significant improvement in OS whereas ipi10 did not.[13] These data remove support for HDI as adjuvant treatment of melanoma. As newer checkpoint inhibitors emerge, the role of ipi3 remains to be further defined.
Large randomized trials with the newer checkpoint inhibitors (nivolumab and pembrolizumab) and with combination signal transduction inhibitors (dabrafenib plus trametinib) showed a clinically significant impact on relapse-free survival (RFS). CheckMate 238 (NCT02388906) compared nivolumab with ipi10 and found that nivolumab was superior in RFS and had a more tolerable safety profile.[14] Pembrolizumab was superior to placebo in RFS, with data on OS still maturing in the MK-3475-054/KEYNOTE-054 trial (NCT02362594).[15] Dabrafenib plus trametinib was superior to placebo in RFS, with data on OS still maturing in the COMBI-AD trial (NCT01682083).[16] Single-agent BRAF-inhibitor therapy with vemurafenib did not show improved RFS compared with placebo in the BRIM8 trial (NCT01667419).[17]
The benefit of immunotherapy with ipilimumab, nivolumab, and pembrolizumab has been seen regardless of programmed death-ligand 1 (PD-L1) expression or BRAF mutations. Combination signal transduction inhibitor therapy is an additional option for patients with BRAF mutations.
Participation in clinical trials designed to identify treatments that will further extend RFS and OS with less toxicity and shorter treatment schedules is an important option for all patients.
A completed, multicenter, phase III randomized trial (SWOG-8593) of patients with high-risk primary stage I limb melanoma did not show a disease-free survival or OS benefit from isolated limb perfusion with melphalan, when compared with surgery alone.[5]
Although melanoma that has spread to distant sites is rarely curable, treatment options are rapidly expanding. Two approaches—checkpoint inhibition and targeting the mitogen-activated protein kinase pathway—have demonstrated improvement in OS in randomized trials. Given the rapid development of new agents and combinations, patients and their physicians are encouraged to consider treatment in a clinical trial for initial treatment and at the time of progression.
The FDA has approved three checkpoint inhibitors: pembrolizumab, nivolumab, and ipilimumab. Each has demonstrated the ability to impact OS against different comparators in unresectable or advanced disease. For more information, see the sections on Pembrolizumab, Nivolumab, and Ipilimumab. Multiple phase III trials are in progress to determine optimal sequencing of immunotherapies, immunotherapy with targeted therapy, and whether combinations of immunotherapies or immunotherapy plus targeted therapy are superior for increasing OS.
The FDA approved IL-2 in 1998 because of durable complete response (CR) rates in a minority of patients (6%–7%) with previously treated metastatic melanoma in eight phase I and II studies. Phase III trials comparing high-dose IL-2 with other treatments and providing an assessment of relative impact on OS have not been conducted.
The combination of anti–programmed death-1 (PD-1) and anti–cytotoxic T-lymphocyte antigen-4 (CTLA-4) immunotherapies (nivolumab and ipilimumab) has demonstrated prolongation of progression-free survival (PFS) and OS compared with ipilimumab monotherapy, but the combination is associated with significant toxicity.
Studies to date indicate that both BRAF and MEK inhibitors can significantly impact the natural history of melanoma, although they do not appear to be curative as single agents. Two combination regimens of BRAF and MEK inhibitors have demonstrated improved PFS and OS compared with BRAF inhibitor monotherapy.
Vemurafenib, approved by the FDA in 2011, has shown an improvement in PFS and OS in patients with unresectable or advanced disease. Vemurafenib is an orally available, small-molecule, selective BRAF V600E kinase inhibitor, and its indication is limited to patients with a demonstrated BRAF V600E mutation by an FDA-approved test.[11]
Dabrafenib is an orally available, small-molecule, selective BRAF inhibitor that was approved by the FDA in 2013. It showed improvement in PFS when compared with dacarbazine in an international multicenter trial (BREAK-3 [NCT01227889]).
Trametinib is an orally available, small-molecule, selective inhibitor of MEK1 and MEK2. The FDA approved trametinib in 2013 for patients with unresectable or metastatic melanoma with BRAF V600E or K mutations. Trametinib demonstrated improved PFS over dacarbazine.
Cobimetinib is an orally available, small-molecule, selective MEK inhibitor. The FDA approved cobimetinib in 2015 for use in combination with the BRAF inhibitor vemurafenib. For more information, see the Combination signal transduction inhibitor therapy section.
Early data suggest that mucosal or acral melanomas with activating mutations or amplifications in c-KIT may be sensitive to a variety of c-KIT inhibitors.[18-20] Phase II and phase III trials are available for patients with unresectable stage III or stage IV melanoma harboring the c-KIT mutation.
The FDA approved two combination regimens, dabrafenib plus trametinib and vemurafenib plus cobimetinib, in patients with unresectable or metastatic melanomas that carry the BRAF V600E or V600K mutation as confirmed by an FDA-approved test. The approvals were based on improved PFS and OS when compared with a single-agent BRAF inhibitor (either dabrafenib or vemurafenib).
The triplet regimen of cobimetinib (MEK inhibitor), vemurafenib (BRAF kinase inhibitor), and atezolizumab (PD-L1 inhibitor) showed improved PFS over the combination of cobimetinib and vemurafenib.[21]
Dacarbazine was approved in 1970 on the basis of overall response rates. Phase III trials indicated an overall response rate of 10% to 20%, with rare CRs observed. An impact on OS has not been demonstrated in randomized trials.[22-25] When used as a control arm for recent registration trials of ipilimumab and vemurafenib in previously untreated patients with metastatic melanoma, dacarbazine was shown to be inferior for OS.
Temozolomide, an oral alkylating agent, appeared to be similar to intravenous dacarbazine in a randomized phase III trial with a primary end point of OS. However, because the trial was designed to demonstrate the superiority of temozolomide, which was not achieved, the trial was left with a sample size that was inadequate to provide statistical proof of noninferiority.[23]
Melanoma metastatic to distant, lymph node–bearing areas may be palliated by regional lymphadenectomy. Isolated metastases to the lung, gastrointestinal tract, bone, or sometimes the brain may be palliated by resection, with occasional long-term survival.[14,26,27]
Standard treatment options for stage 0 melanoma include the following:
Patients with stage 0 disease may be treated by excision with minimal, but microscopically free, margins.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
Standard treatment options for stage I melanoma include the following:
Evidence suggests that lesions no thicker than 2 mm may be treated conservatively with radial excision margins of 1 cm.
Depending on the location of the melanoma, most patients can now have the excision performed on an outpatient basis.
Evidence (excision):
Elective regional lymph node dissection is of no proven benefit for patients with stage I melanoma.[8]
Lymphatic mapping and sentinel lymph node biopsy (SLNB) for patients who have tumors of intermediate thickness and/or ulcerated tumors may identify individuals with occult nodal disease. These patients may benefit from regional lymphadenectomy and adjuvant therapy.[6,9-11]
Evidence (immediate lymphadenectomy vs. observation with delayed lymphadenectomy):
Treatment options under clinical evaluation for patients with stage I melanoma include the following:
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
Standard treatment options for stage II melanoma include the following:
For melanomas with a thickness between 2 and 4 mm, surgical margins need to be 2 to 3 cm or smaller.
Few data are available to guide treatment in patients with melanomas thicker than 4 mm; however, most guidelines recommend margins of 3 cm whenever anatomically possible.
Depending on the location of the melanoma, most patients can have the excision performed on an outpatient basis.
Evidence (excision):
Lymphatic mapping and SLNB have been used to assess the presence of occult metastasis in the regional lymph nodes of patients with stage II disease, potentially identifying individuals who may be spared the morbidity of regional lymph node dissections (LNDs) and individuals who may benefit from adjuvant therapy.[3-7]
To ensure accurate identification of the sentinel lymph node, lymphatic mapping and removal of the SLN should precede wide excision of the primary melanoma.
With the use of a vital blue dye and a radiopharmaceutical agent injected at the site of the primary tumor, the first lymph node in the lymphatic basin that drains the lesion can be identified, removed, and examined microscopically. Multiple studies have demonstrated the diagnostic accuracy of SLNB, with false-negative rates of 0% to 2%.[3,8-12] If metastatic melanoma is detected, a complete regional lymphadenectomy can be performed in a second procedure.
No published data on the clinical significance of micrometastatic melanoma in regional lymph nodes are available from prospective trials. Some evidence suggests that for patients with tumors of intermediate thickness and occult metastasis, survival is better among patients who undergo immediate regional lymphadenectomy than it is among those who delay lymphadenectomy until the clinical appearance of nodal metastasis.[13] This finding should be viewed with caution because it arose from a post hoc subset analysis of data from a randomized trial.
Evidence (regional lymphadenectomy):
Postsurgical systemic adjuvant treatment has not been adequately studied in patients with stage II disease; therefore, clinical trials are an important therapeutic option for patients at high risk of relapse.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
Standard treatment options for resectable stage III melanoma include the following:
The primary tumor may be treated with wide local excision with 1 to 3-cm margins, depending on tumor thickness and location.[1-7] Skin grafting may be necessary to close the resulting defect.
Lymphatic mapping and SLNB can be considered to assess the presence of occult metastases in the regional lymph nodes of patients with primary tumors larger than 1 to 4 mm, potentially identifying individuals who may be spared the morbidity of regional lymph node dissections and individuals who may benefit from adjuvant therapy.[3,8-12]
To ensure accurate identification of the sentinel lymph node (SLN), lymphatic mapping and removal of the SLN should precede wide excision of the primary melanoma.
Multiple studies have demonstrated the diagnostic accuracy of SLNB, with false-negative rates of 0% to 2%.[8,12-17] If metastatic melanoma is detected, a complete regional lymphadenectomy can be performed in a second procedure.
Patients can be considered for CLND if the sentinel node(s) is microscopically or macroscopically positive for regional control or considered for entry into the Multicenter Selective Lymphadenectomy Trial II (NCT00297895) to determine whether CLND affects survival. SLNB should be performed prior to wide excision of the primary melanoma to ensure accurate lymphatic mapping.
Adjuvant therapeutic options are expanding for patients at high risk of recurrence after complete resection and include checkpoint inhibitors and combination signal transduction inhibitor therapy. Ipilimumab was the first checkpoint inhibitor to be approved by the U.S. Food and Drug Administration (FDA) as adjuvant therapy, and it has demonstrated improved overall survival (OS) at 10 mg/kg (ipi10) compared with placebo (EORTC 18071 [NCT00636168]).[18] However, ipi10 has significant toxicity at this dose. The North American Intergroup Trial E1609 (NCT01274338), designed with three treatment groups, compared ipi10 with ipilimumab at a lower dose of 3 mg/kg (ipi3) (approved for metastatic disease) and with high-dose interferon (HDI). Ipi3 showed a significant improvement in OS whereas ipi10 did not.[19] These data remove support for HDI as adjuvant treatment for melanoma. As newer checkpoint inhibitors emerge, the role of ipi3 remains to be further defined.
Large randomized trials with the newer checkpoint inhibitors (nivolumab and pembrolizumab) and with combination signal transduction inhibitors (dabrafenib plus trametinib) showed a clinically significant impact on relapse-free survival (RFS). CheckMate 238 (NCT02388906) compared nivolumab with ipi10 and found that nivolumab was superior in RFS and had a more tolerable safety profile.[20] Pembrolizumab was superior to placebo in RFS, with data on OS still maturing in the MK-3475-054/KEYNOTE-054 trial (NCT02362594).[21] Dabrafenib plus trametinib was superior to placebo in RFS, with data on OS still maturing in the COMBI-AD trial (NCT01682083).[22] Single-agent BRAF-inhibitor therapy with vemurafenib did not show improved RFS compared with placebo in the BRIM8 trial (NCT01667419).[23]
The benefit of immunotherapy with ipilimumab, nivolumab, and pembrolizumab has been seen regardless of programmed death-ligand 1 (PD-L1) expression or BRAF mutations. Combination signal transduction inhibitor therapy with dabrafenib plus trametinib is an additional option for patients with BRAF mutations.
Participation in clinical trials to identify treatments that will further extend RFS and OS with less toxicity and shorter treatment schedules is an important option for all patients.
Evidence (nivolumab):
A total of 906 patients were randomly assigned: 453 patients to nivolumab and 453 patients to ipilimumab. Baseline characteristics were balanced. Approximately 81% of patients had stage III disease, 32% had ulcerated primary melanoma, 48% had macroscopic lymph node involvement, 62% had less than a 5% PD-L1 expression, and 42% harbored BRAF mutations.
Evidence (pembrolizumab):
A total of 1,019 patients were randomly assigned: 514 to pembrolizumab and 505 to placebo. Baseline characteristics were balanced. Approximately 40% had ulcerated primary melanoma, 66% had macroscopic lymph node involvement, 84% had positive PD-L1 expression (melanoma score >2 by 22C3 antibody assay), and 44% harbored BRAF mutations.
Evidence (ipilimumab):
The trial was designed with two coprimary end points, RFS and OS, with a hierarchic analysis to evaluate ipi3 versus HDI followed by ipi10 versus HDI. The time to event was longer than anticipated and the design was amended for a final analysis at a data cutoff date giving a median follow-up time of 57.4 months (range, 0.03 months−86.6 months).
Toxicity with ipi3 was lower than with ipi10; however, both had treatment-related discontinuations and death.
The study concluded that evidence no longer supports a role for HDI as adjuvant therapy for patients with high-risk melanoma. Further, ipi3 provides OS data superior to ipi10 compared with HDI. The role of ipilimumab as adjuvant monotherapy is unclear because CheckMate 238 demonstrated that nivolumab was superior to ipi10 in improving RFS, with OS data still maturing.
An updated analysis was performed at a median follow-up of 5.3 years.[18]
Data from this trial (EORTC 18071), which tested high-dose ipilimumab at 10 mg/kg compared with placebo, served as the basis for the approval of ipilimumab in the adjuvant setting. However, the subsequent intergroup trial, E1609 (described above), demonstrated better outcomes with low-dose (3 mg/kg) ipilimumab, which is also the dose approved for metastatic disease.
Evidence (dabrafenib plus trametinib):
Treatment options under clinical evaluation for patients with resectable stage III melanoma include the following:
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
Treatment options for unresectable stage III, stage IV, and recurrent melanoma include the following:
Two approaches—checkpoint inhibition and targeting the mitogen-activated protein kinase (MAPK) pathway—demonstrated improvement in progression-free survival (PFS) and overall survival (OS) in randomized trials. Anti–PD-1 monotherapy (pembrolizumab or nivolumab) demonstrated improved efficacy outcomes with better safety profiles when compared with treatment using single-agent anti–CTLA-4 (ipilimumab) or investigator choice of chemotherapy. The combination of anti–PD-1 and anti–CTLA-4 immunotherapies (nivolumab and ipilimumab) also prolongs PFS and OS compared with ipilimumab, but the combination is associated with significant toxicity. The efficacy seen with immunotherapy is independent of BRAF mutation status.
Combinations of BRAF and MEK inhibitors have consistently shown superior efficacy compared with BRAF monotherapy. Improved PFS was seen when a PD-L1 inhibitor (atezolizumab) was added to the combination of a BRAF plus MEK inhibitor (vemurafenib plus cobimetinib); however, data on OS is immature. Further questions remain regarding triplet therapy, including how it compares with monotherapy checkpoint inhibition and if the concurrent administration is superior to sequential therapy (NCT02224781).
Because of the rapid development of new agents, combinations, and remaining questions, patients and their physicians are encouraged to consider a clinical trial for initial treatment and at the time of progression. Clinical trials are addressing the following issues:
T-VEC is a genetically modified, herpes simplex virus type 1 (HSV1) oncolytic therapy approved for local intralesional injection into unresectable cutaneous, subcutaneous, and nodal lesions in patients with melanoma that recurs after initial surgery. T-VEC is designed to replicate within tumors, causing lysis, and to produce granulocyte-macrophage colony-stimulating factor (GM-CSF). Release of antigens together with virally derived GM-CSF may promote an antitumor immune response; however, the exact mechanism of action is unknown.
The approval of T-VEC by the U.S. Food and Drug Administration (FDA) is based on data that demonstrated shrinkage of lesions. However, improvement of OS or an effect on visceral metastases or improvement in quality of life has not been shown.
Evidence (T-VEC):
Precautions: T-VEC is a live, attenuated HSV and may cause life-threatening, disseminated herpetic infection. It is contraindicated in immunocompromised or pregnant patients. Health care providers and close contacts should avoid direct contact with injected lesions. Biohazard precautions for preparation, administration, and handling are provided in the label.
Detailed prescribing information by treatment cycle and lesion size are provided in the FDA label.
The PD-1 pathway is a key immunoinhibitory mediator of T-cell exhaustion. Blockade of this pathway can lead to T-cell activation, expansion, and enhanced effector functions. PD-1 has two ligands, PD-L1 and PD-L2. Two anti–PD-1 antibodies, pembrolizumab and nivolumab, were approved by the FDA on the basis of improved OS in randomized trials.
Evidence (pembrolizumab):
Pembrolizumab was discontinued because of AEs in 7% of the patients treated with 2 mg/kg, with 3% considered drug-related AEs by the investigators. The most common AEs in the 2 mg/kg versus 10 mg/kg arms were the following:
Other common AEs included cough, nausea, decreased appetite, constipation, arthralgia, and diarrhea. The most frequent and serious AEs that occurred in more than 2% of a total of 411 patients treated with pembrolizumab included renal failure, dyspnea, pneumonia, and cellulitis. Additional clinically significant irAEs included pneumonitis, colitis, hypophysitis, hyperthyroidism, hypothyroidism, nephritis, and hepatitis.
The FDA label provides recommendations for suspected irAEs, including withholding the drug and administering corticosteroids.
Approximately 66% of patients had received no previous systemic therapy for advanced melanoma. BRAF V600 mutations were present in 36% of patients and of these, approximately 50% had received previous BRAF inhibitor treatments. The study did not enroll patients with BRAF V600 mutations with high LDH levels and symptomatic or rapidly progressive disease who had not received anti-BRAF therapy, which could provide rapid clinical benefit. Approximately 80% of patients had PD-L1–positive tissue samples.
Evidence (nivolumab):
The FDA label provides recommendations for suspected irAEs, including withholding the drug and administering corticosteroids.
Ipilimumab is a human monoclonal antibody that binds to CTLA-4, thereby blocking its ability to downregulate T-cell activation, proliferation, and effector function.
Approved by the FDA in 2011, ipilimumab has demonstrated clinical benefit by prolonging OS in randomized trials. Two prospective, randomized, international trials, one each in previously untreated and treated patients, supported the use of ipilimumab.[7,8]
Evidence (ipilimumab):
Clinicians and patients should be aware that immune-mediated adverse reactions may be severe or fatal. Early identification and treatment are necessary, including potential administration of systemic glucocorticoids or other immunosuppressants according to the immune-mediated adverse reaction management guide provided by the manufacturer.[9]
IL-2 was approved by the FDA in 1998 because of durable CRs in eight phase I and II studies. Phase III trials comparing high-dose IL-2 to other re-treatments, providing an assessment of relative impact on OS, have not been conducted.
Evidence (high-dose IL-2):
Strategies to improve this therapy are an active area of investigation.
T-cells coexpress several receptors that inhibit T-cell function. Preclinical data and early clinical data suggest that co-blockade of the two inhibitory receptors, CTLA-4 and PD-1, may be more effective than blockade of either alone.[12] This has led to a phase III trial (NCT01844505) comparing each single agent with the combination.
Evidence (ipilimumab plus nivolumab):
PFS and OS were coprimary end points. The study was powered to compare the combination of nivolumab plus ipilimumab with ipilimumab monotherapy, and nivolumab monotherapy with ipilimumab monotherapy; the study was not powered to compare combination ipilimumab plus nivolumab with nivolumab.
Patients were stratified according to tumor PD-L1 status assessed in a central laboratory by immunohistochemical testing (positive vs. negative or indeterminate), BRAF mutation status (V600 mutation−positive vs. wild-type), and American Joint Committee on Cancer stage.[13][Level of evidence A1]
Treatment consisted of nivolumab (1 mg/kg) plus ipilimumab (3 mg/kg) every 3 weeks for up to 4 doses, followed by nivolumab (3 mg/kg) every 2 weeks until progression or unacceptable toxicity.
The primary end point was rate of intracranial clinical benefit assessed by the investigator per RECIST criteria and defined as the percentage of patients with CR, PR, or stable disease for at least 6 months. A total of 28 sites in the United States enrolled 101 patients, of whom 94 had a minimum follow-up of 6 months; the data on that population are reported below.
Studies to date indicate that both BRAF and MEK (mitogen-activated ERK-[extracellular signal-regulated kinase] activating kinase) inhibitors, as single agents and in combination, can significantly impact the natural history of melanoma, although they do not appear to provide a cure.
Treatment with BRAF inhibitors is discouraged in wild-type BRAF melanoma because data from preclinical models have demonstrated that BRAF inhibitors can enhance rather than downregulate the mitogen-activated protein kinase (MAPK) pathway in tumor cells with wild-type BRAF and upstream RAS mutations.[16-19]
Vemurafenib is an orally available, small-molecule, selective BRAF kinase inhibitor that was approved by the FDA in 2011 for patients with unresectable or metastatic melanoma who test positive for the BRAF V600E mutation.
Evidence (vemurafenib):
Dabrafenib is an orally available, small-molecule, selective BRAF inhibitor that was approved by the FDA in 2013. It is used for treatment of patients with unresectable or metastatic melanoma who test positive for the BRAF V600E mutation as detected by an FDA-approved test. Dabrafenib and other BRAF inhibitors are not recommended for treatment of BRAF wild-type melanomas, as in vitro experiments suggest there may be a paradoxical stimulation of MAPK signaling resulting in tumor promotion.
Evidence (dabrafenib):
Trametinib is an orally available, small-molecule, selective inhibitor of MEK1 and MEK2. BRAF activates MEK1 and MEK2 proteins, which in turn, activate MAPK. Preclinical data suggest that MEK inhibitors can restrain growth and induce cell death of some BRAF-mutated human melanoma tumors.
In 2013, trametinib was approved by the FDA for patients with unresectable or metastatic melanoma with BRAF V600E or V600K mutations, as determined by an FDA-approved test.
Evidence (trametinib):
Cobimetinib is a small-molecule, selective MEK inhibitor that the FDA approved in 2015 for use in combination with the BRAF inhibitor vemurafenib. For more information, see the Combination therapy with signal transduction inhibitors section.
Early data suggest that mucosal or acral melanomas with activating mutations or amplifications in c-KIT may be sensitive to a variety of c-KIT inhibitors.[24-26] Phase II and phase III trials are available for patients with unresectable stage III or stage IV melanoma harboring the c-KIT mutation.
Results from phase III trials comparing three different combinations of BRAF-MEK inhibitors with BRAF inhibitor monotherapy have consistently shown efficacy superior to BRAF monotherapy.
Secondary resistance to BRAF inhibitor monotherapy in patients with BRAF V600 mutations may be associated with reactivation of the MAPK pathway. Therefore, combinations of signal transduction inhibitors that block different sites in the same pathway or sites in multiple pathways are an active area of research.
Evidence (dabrafenib plus trametinib):
Evidence (vemurafenib plus cobimetinib):
Encorafenib is a small-molecule BRAF inhibitor that is approved in combination with binimetinib, a small-molecule MEK inhibitor, for the treatment of unresectable or metastatic melanoma with a BRAF V600E or V600K mutation, as detected by an FDA-approved test. The combination has demonstrated improved PFS and OS compared with vemurafenib; however, neither is approved as single-agent therapy.
Evidence (encorafenib plus binimetinib):
Evidence (cobimetinib and vemurafenib plus atezolizumab):
After all patients in both arms received a 28-day cycle of cobimetinib and vemurafenib, patients received atezolizumab (840 mg IV every 2 weeks) or placebo in addition to the combination BRAF-MEK inhibitor therapy. The primary efficacy end point was investigator-assessed PFS per RECIST 1.1 criteria.
The impact of triplet therapy on OS, or when compared with checkpoint inhibitor monotherapy, or to sequential therapy with combination BRAF-MEK inhibitor therapy, preceded or followed by checkpoint inhibition (ongoing trial NCT02224781) is unknown.
Dacarbazine was approved in 1970 on the basis of objective response rates. Phase III trials indicate an objective response rate of 10% to 20%, with rare CRs observed. An impact on OS has not been demonstrated in randomized trials.[7,20,35-37] When used as a control arm for recent registration trials of ipilimumab and vemurafenib in previously untreated patients with metastatic melanoma, dacarbazine was inferior for OS.
Temozolomide, an oral alkylating agent that hydrolyzes to the same active moiety as dacarbazine, appeared to be similar to dacarbazine (IV administration) in a randomized, phase III trial with a primary end point of OS. However, the trial was designed for superiority, and the sample size was inadequate to prove equivalency.[36]
The objective response rate to dacarbazine and the nitrosoureas, carmustine and lomustine, is approximately 10% to 20%.[35,38-40] Responses are usually short-lived, ranging from 3 to 6 months, although long-term remissions can occur in a limited number of patients who attain a CR.[38,40]
A randomized trial compared IV dacarbazine with temozolomide, an oral agent; OS was 6.4 months for dacarbazine versus 7.7 months for temozolomide (HR, 1.18; 95% CI, 0.92–1.52). While these data suggested similarity between dacarbazine and temozolomide, no benefit in survival has been demonstrated for either dacarbazine or temozolomide; therefore, demonstration of similarity did not result in FDA approval of temozolomide.[36][Level of evidence A1]
An extended schedule and escalated dose of temozolomide was compared with dacarbazine in a multicenter trial by the European Organisation for Research and Treatment of Cancer (EORTC) (EORTC-18032 [NCT00101218]) randomly assigning 859 patients. No improvement was seen in OS or PFS for the temozolomide group, and this dose and schedule resulted in more toxicity than standard-dose, single-agent dacarbazine.[41][Level of evidence A1]
Two randomized phase III trials in previously untreated patients with metastatic melanoma (resulting in FDA approval for vemurafenib [20] and ipilimumab [7]) included dacarbazine as the standard therapy arm. Both vemurafenib (in BRAF V600 mutant melanoma) and ipilimumab showed superior OS compared with dacarbazine in the two separate trials.
Other agents with modest, single-agent activity include vinca alkaloids, platinum compounds, and taxanes.[38,39]
Attempts to develop combination regimens that incorporate chemotherapy (e.g., multiagent chemotherapy,[42,43] combinations of chemotherapy and tamoxifen,[44-46] and combinations of chemotherapy and immunotherapy [10,11,42,47-50]) have not demonstrated an improvement in OS.
A published data meta-analysis of 18 randomized trials (15 of which had survival information) that compared chemotherapy with biochemotherapy (i.e., the same chemotherapy plus interferon alone or with IL-2) reported no impact on OS.[51][Level of evidence A1]
Melanoma metastatic to distant, lymph node–bearing areas may be palliated by regional lymphadenectomy. Isolated metastases to the lung, gastrointestinal tract, bone, or sometimes the brain may be palliated by resection, with occasional long-term survival.[48-50]
Although melanoma is a relatively radiation-resistant tumor, palliative radiation therapy may alleviate symptoms. Retrospective studies have shown that symptom relief and some shrinkage of the tumor with radiation therapy may occur in patients with the following:[52,53]
The most effective dose-fractionation schedule for palliation of melanoma metastatic to the bone or spinal cord is unclear, but high-dose-per-fraction schedules are sometimes used to overcome tumor resistance. For more information, see Cancer Pain.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
General Information About Melanoma
Updated statistics with estimated new cases and deaths for 2024 (cited American Cancer Society as reference 1).
Revised text to state that since the early 2000s, the incidence of melanoma in people aged 50 years and older has stabilized in men and increased by about 3% per year in women.
This summary is written and maintained by the PDQ Adult Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of melanoma. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
This summary is reviewed regularly and updated as necessary by the PDQ Adult Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Melanoma Treatment are:
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Adult Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Melanoma Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: https://www.cancer.gov/types/skin/hp/melanoma-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389469]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.