You are on page 1of 55

The Management of Head and Neck Melanoma

The worldwide incidence of melanoma continues to increase faster than any other cancer.1 It is currently the fth leading cause of cancer among men and the sixth leading cause among women.2 Fortunately, the management of melanoma is also 1 of the fastest evolving elds in cancer, with promising research taking place at both the molecular and clinical level. Despite these research efforts, many controversies still surround the management of melanoma. What is the optimal evaluation for a newly diagnosed melanoma patient? What are the ideal surgical margins? Does early management of draining nodal basins impact overall survival? Does sentinel lymph node biopsy (SLNB) impart a survival benet? What is the role of adjuvant interferon- 2b? The head and neck (HN) region adds further complexity with its rich and often watershed lymphatic system, intricate anatomy, and vital structures such as the carotid artery and facial nerve. The goal of this issue is to review the evaluation and management of patients with HN melanoma. In doing so, the new 2002 American Joint Committee on Cancer (AJCC) staging system will be highlighted, along with the current National Cancer Comprehensive Network (NCCN) practice guidelines. The majority of the monograph chapter will focus specically on the management of cutaneous head and neck melanoma, which is far more common than mucosal melanoma. Mucosal melanoma and melanoma of unknown primary origin will be discussed briey as independent sections.

Demographics
Despite a recent decline in the overall trend in cancer incidence and mortality, the incidence of cutaneous melanoma continues to rise faster than any other cancer.3 The National Cancer Institutes Surveillance, Epidemiology and End Results (SEER) Data estimates that 59,580 new cases of invasive cutaneous melanoma will be diagnosed in 2005. An additional 46,170 in situ cases are estimated that same year. For women
Curr Probl Surg 2006;43:781-835. 0011-3840/2006/$30.00 0
doi:10.1067/j.cpsurg.2006.08.003

Curr Probl Surg, November 2006

781

under the age of 40, the incidence of melanoma is second only to breast cancer.1 Over the past 50 years, the annual percentage change in invasive melanoma incidence has steadily increased 4.3% each year.3 The lifetime risk for developing invasive melanoma has climbed at epidemic proportions.4 This risk increased from 1 in 1500 for individuals born in 1935, to 1 in 250 in 1980. This trend is expected to continue at a startling rate, with 1 in 39 individuals born in 2010 projected to develop melanoma.5 Melanoma is the most lethal form of skin cancer. In 2005, it will be responsible for an estimated 7770 deaths within the United States.3 This estimate averages to approximately 1 American dying from melanoma every hour. Over the past 50 years, the annual percentage change in mortality rate has increased at a steady rate of 1.8% per year. Melanoma accounts for the second highest increase in mortality rate, especially for men over the age of 65. However, it should not be viewed as a cancer limited to the elderly. Melanoma is known to afict young adults, with 1 in 4 patients diagnosed before the age of 40.5,6 Consequently, melanoma is responsible for 1 of the leading cancer causes of lost potential life years. Approximately 25% of all cutaneous melanomas arise in the head and neck (HN) region, with more than 9000 cases diagnosed annually. A slight male predominance has been consistently reported throughout the literature.7-9 The median age of diagnosis for patients with HN melanoma is 55 years, slightly younger than the median age of 57 years reported for all melanoma sites.6 However, juvenile cases account for 1.66% of HN melanoma cases, with patients being diagnosed as young as 4 years of age.10,11

Tumor Biology
Melanocytes are dendritic cells of neural crest origin, located at the epidermal-dermal junction. Within these cells, cytoplasmic organelles termed melanosomes synthesize melanin, which is distributed to surrounding keratinocytes to form a supranuclear cap that provides protection from damaging ultraviolet radiation (UVR).12 The photo-protective property of melanin results from the absorption of both UVR photons and its oxygen radical by-products.13-15 The protective role of melanin is evident through tanning in which UVR exposure increases melanogenesis. The increased amount of melanin granules is transferred to surrounding keratinoctyes, thereby darkening the skin color. This protective layer of melanin serves as an endogenous sunscreen, persisting for approximately 3 weeks following exposure.15 Whereas cutaneous squamous cell carcinoma (SCC) and basal cell
782 Curr Probl Surg, November 2006

carcinoma (BCC) are associated with cumulative sun exposure, melanoma development correlates with intermittent and intense exposures common in sunburns.16-18 This difference is thought to be due to intrinsic differences in tumor cell origin. SCC and BCC both arise from keratinocytes that undergo apoptosis in response to severe UVR damage. With repeated low-dose solar exposure, keratinocytes can accumulate a significant amount of unrepaired DNA damage to result in SCC or BCC of the skin. Melanocytes, however, have not been found to undergo apoptosis.19 Gilcrest and colleagues speculate that the absence of apoptosis in DNA-damaged melanocytes represents a protective measure since these cells have a limited ability to proliferate. By escaping apoptosis, the photo-protective role of melanocytes is preserved. However, this protective measure comes at the expense of increased melanoma risk. The appearance of freckles after intense sun exposure supports this theory, because freckles represent clones of mutated melanocytes and carry an increased risk for melanoma.20

Head and Neck Distribution


All individuals, regardless of ethnicity, share approximately the same number of melanocytes. It is a difference in the number, distribution, and density of melanin granules within keratinocytes that account for racial variation in skin color.21 Within the human body, melanocyte density varies. The average number of melanocytes per square millimeter squared (mm2) in an adult is 1194 for the face, 1060 for the scalp, and 926 for the neck. This concentration is considerably higher compared with other anatomic sites such as the buttock and abdomen, which contain only 565 and 578 melanocytes/mm2, respectively. The concentration of melanocytes within sun-exposed regions of the body emphasizes the UVR protective role of melanin.22 The majority of HN cutaneous melanomas arise on the cheek, scalp, and neck. Among 857 HN melanoma patients, Fisher and colleagues found that the face and neck region accounted for more than 60% of all primary tumors. An additional 26% arouse from the scalp, with the ear and nose accounting for only 9% and 4% of primary tumors, respectively.6 These results were conrmed by OBrien and colleagues who reported the face as the most common site (47%), followed by the neck (29%), scalp (14%), and ear (10%).8 The cheek and forehead region contain a 2- to 3-fold greater density of melanocytes compared with other anatomic sites. This difference, coupled with increased sun exposure, is thought to account for the distribution of melanoma within the HN region.
Curr Probl Surg, November 2006 783

TABLE 1. Cutaneous melanoma risk factors Environmental Inability to tan Fair complexion Blue/green eyes Blonde/red hair Freckling History of blistering/peeling sunburns Teenage outdoor summer jobs Genetic/past medical history CDKN2A (p16) mutation History of prior melanoma Family history of melanoma Actinic keratoses Non-melanoma skin cancer Xeroderma pigmentosa (XP) Atypical (dysplastic) nevus Giant congenital melanocytic nevus Immunosuppression

Etiology and Risk Factors


Several environmental and genetic risk factors have been implicated in the development of cutaneous melanoma.23 Numerous risk factors are summarized in Table 1.

Sun Exposure
Sun exposure is the leading cause of melanoma.23,24 As noted above, patients with a signicant history of sun exposure are at particularly high risk, especially if they experienced intense exposure resulting in blistering sunburns.25 Risk factors for sunburns such as red or blond hair, green or blue eyes, or fair skin consistent with Fitzpatrick skin type I-III share a strong correlation with the development of melanoma.26 Patients previously diagnosed with melanoma are also at increased risk, with 5% to 10% of individuals developing a second primary cancer.27 This risk is life-long and can occur anywhere on the skin. Therefore, long-term, annual follow-up with a thorough total body examination is critical. In addition, adults with more than 100 clinically normal appearing nevi, children with more than 50 clinically normal appearing nevi, and any patient with atypical or dysplastic nevi are at risk.28

Genetics
A genetic etiology has also been implicated in the pathogenesis of melanoma.29 CDKN2A, also known as p16, remains the most common chromosomal mutation associated with melanoma.30 Overall, the mutation accounts for only a small percentage of melanoma cases observed. For example, Aitken and colleagues identied the mutation in only 0.2% of the melanoma cases diagnosed in Australia.31 Up to15% of melanoma patients report a positive family history.32 In the 1970s, Clark and colleagues noted the hereditary nature of
784 Curr Probl Surg, November 2006

cutaneous melanoma when they described several family members aficted with acquired, large, irregular, and dysplastic nevi, often in sun-protected regions of the body such as the scalp and trunk.33 They coined the term B-K mole syndrome after 2 families in their study. During this time period, Lynch and colleagues independently reported a familial association of melanoma among individuals with atypical nevi. They proposed the term familial atypical multiple mole-melanoma syndrome or FAMMM syndrome for family members.34 Today, the term atypical mole syndrome is applied to familial cases of melanoma. The syndrome is inherited in an autosomally dominant fashion.35 Patients suffering from atypical mole syndrome carry a 10-year melanoma risk of 10.7%, which is signicantly greater than the 0.62% risk reported in control patients.36,37 A 56% cumulative risk is estimated in these patients from age 20 to 59 years, with 100% of atypical mole syndrome patients developing melanoma by age 76.38 Another hereditary disorder associated with melanoma is xeroderma pigmentosa (XP). This rare disease is inherited in an autosomally recessive fashion.39 The broblasts in XP patients have an impaired ability to repair DNA damaged by ultraviolet light.40 As a result, multiple primary cutaneous malignancies including melanoma, BCC, and SCC develop. Individuals are usually diagnosed with their rst cancer before the age of 10. Despite ultraviolet light precautions, careful surveillance, and aggressive treatment, the development of skin cancers is relentless. XP patients ultimately succumb to their disease at an early age.

Immunosuppression
Numerous studies throughout the literature provide supporting evidence for a role of immunosuppression in the development of melanoma. Children suffering from immunodeciency disorders carry a 3-fold increased risk for melanoma.41 Renal transplant patients requiring immunosuppressive therapy may have a higher incidence of melanoma compared with the general population.42 Although the SEER program for San Francisco has not reported an increased trend in melanoma among men at risk for human immunodeciency virus (HIV),43 several case series detail aggressive melanomas in this setting.44,45 In addition, higher rates of premalignant, melanocytic nevi are associated with renal transplantation,46 chemotherapy,47 and childhood leukemia.48

Congenital Nevi
Congenital melanocytic nevi (CMN) are pigmented lesions present at birth or within the rst 6 months of infancy.49 Up to 6% of children are
Curr Probl Surg, November 2006 785

born with CMN. These nevi are classied according to size, with small CMN measuring less than 1.5 cm in diameter and accounting for the majority of lesions. Medium CMN range between 1.5 and 19.9 cm in diameter. Large CMN, also termed giant congenital nevi, measure 20 cm or greater. The large size of these lesions carries signicant cosmetic, as well as psychosocial, implications.50 The CMN classication system is important because nevi size correlates with melanoma risk. The risk of melanoma development from small and medium sized CMN is similar to any other nevus. Cancer development in this setting is usually after childhood. Malignant transformation tends to arise at the dermo-epidermal junction, allowing for obvious changes to the nevus and early detection of cancer. For this reason, routine prophylactic removal of small and medium CMN is rarely indicated in the absence of signs or symptoms concerning for malignant progression. However, large/giant CMN carry an increased risk for melanoma, with development in an estimated 5% to 20% of individuals.51,52 Seventy percent of these patients are diagnosed before age 10.37 Melanoma can originate deep to the epidermis, going unnoticed within the large pigmented lesion, thereby delaying diagnosis until advanced disease has developed. For this reason, prophylactic excision of large CMN is advocated if the nevus is in an anatomic location amenable to surgical excision.

Melanoma Classication
Several histologic subtypes of melanoma are encountered within the HN region. Despite the classication system, it is important to realize that melanoma subtype does not generally inuence prognosis once tumor thickness and other prognostic variables such as ulceration are taken into account.53

Common HN Melanoma Subtypes


The most common type of cutaneous melanoma is supercial spreading melanoma (SSM), accounting for approximately 70% of all cases. SSM commonly arises in the setting of a preexisting nevus, with a diagnosis rendered during the fourth or fth decade. The characteristic feature of SSM is color variation, which is often haphazard in nature. Black, dark brown, tan, and blue-gray areas of pigmentation are commonly observed. Areas of pink and white represent hypopigmentation secondary to tumor regression. Although SSM lesions are well circumscribed, the borders tend to be scalloped and asymmetric. Nodular melanoma (NM) is the second most common variant, account786 Curr Probl Surg, November 2006

ing for 15% to 30% of cases. The majority of mucosal melanomas are classied as the nodular subtype (see below). This lesion typically appears as a blue-black or blue-red, raised nodule. For this reason, NM must be differentiated from a hemangioma, blue nevus, pyogenic granuloma, and pigmented BCC. Lentigo maligna (LM) represents intraepidermal or melanoma in situ. Histologically, it is seen in the background of chronic solar damage. LM is considered the precursor to invasive lentigo malignant melanoma (LMM). The exact percentage of LMs that progress to invasive LMM remains unknown54; however, it is speculated that if patients live long enough, all LMs will eventually progress to invasive melanoma. LM/ LMM is commonly found within the HN region. This subtype has been associated with older individuals, but the frequency in younger patients is increasing.28 The LM/LMM pattern warrants special comment because this subtype is often characterized by subclinical and extensive peripheral involvement of atypical junctional melanocytic hyperplasia (AJMH). Therefore, management with adequate wide margins can be challenging from both a functional and cosmetic standpoint. Additionally, amelanotic and invasive desmoplastic melanoma (see discussion here) often arise within LM/LMM.

Desmoplastic-Neurotropic Melanoma
In 1971, Conley and colleagues introduced the term desmoplastic melanoma (DM) to describe a melanoma subtype comprised of spindle cells, abundant collagen, and features resembling bromas.55 Reed and colleagues further noted a propensity for perineural spread among a subset of DMs.56 This prompted further subclassication of a desmoplastic-neurotropic melanoma (DNM) variant. DM/DNM subtypes are rare, accounting for approximately 1% of all cutaneous melanomas.57 However, more than three fourths of the lesions are diagnosed within the HN region. DM/DNM often present as hard, brous, subcutaneous lesions within a background of LM/LMM. It is important to note that the clinical presentation and biologic behavior of these tumors are distinct from traditional cutaneous melanomas. Although amelanotic cases account for only 4% to 5% of cutaneous melanomas, up to 73% of DM/DNMs are amelanotic.57,58 In addition, the neoplasms often lack the typical ABCD criteria for melanoma (described herein). They have a challenging histologic pattern that requires combined histopathologic and immunophenotypic evaluation by an experienced pathologist.59 DM/DNM is recognized as locally aggressive and highly inltrative
Curr Probl Surg, November 2006 787

melanoma. Tumors are often associated with cranial nerve and skull base involvement. It is estimated that 50% of all cases will recur locally.57 Explanations for this high rate include the association with neurotropism, failure to recognize and adequately clear peripheral AJMH margins, and delay in diagnosis due to the atypical appearance. It is interesting to note that although DM/DNM demonstrate greater tumor thickness at the time of diagnosis, the rate of regional lymph node metastasis is estimated between 8% and 15%, much lower than that of other melanoma subtypes.56

Unknown Primary
Three percent to 8% of melanoma patients present initially with an unknown primary site.60-62 Two thirds of these individuals present with regional metastasis in the absence of an identiable primary lesion or history of melanoma. The remaining one third of cases involve distant metastasis to sites such as the subcutaneous tissues, lung, and brain.61-64 Several theories have been proposed to explain melanoma of unknown primary origin. The identication of melanocytes and nevus cells within lymph node capsules and visceral organ epithelium have led some scientists to propose that unknown primary cases result from melanoma arising de novo at regional and distant sites.61,65 An alternative explanation is that the original primary melanoma site undergoes complete, spontaneous regression secondary to antitumor immune response. Therefore, it is no longer identiable.65,66 All patients presenting with melanoma of unknown origin warrant a thorough examination of both the total body skin and the mucosal surfaces. A careful history must also be obtained, specically inquiring about previous skin biopsies, lesions that were frozen, skin tags removed, and moles that spontaneously disappeared. All pathology slides from previously excised lesions should be re-reviewed. The metastatic evaluation is identical to that of known primary cases described below. After adjusting for tumor stage, melanoma of unknown primary origin shares the same disease-free interval and overall survival rates as their counterparts with known primary sites.61-64

Mucosal Melanoma
Mucosal melanoma is recognized as a rare, distinct, and separate subtype from its cutaneous counterpart. Only 1074 (1.3%) of the 84,836 melanoma cases registered in the National Cancer Database from 1985 to 1994 were mucosal in origin.60 Fifty percent of all mucosal melanoma
788 Curr Probl Surg, November 2006

cases arise within the HN region, making it the most common location.67 However, less than 2% of all HN melanomas are of the mucosal variant. The demographic aspects of mucosal melanoma differ in several respects from cutaneous melanoma. Mucosal melanoma is usually diagnosed during the sixth to seventh decade, approximately 10 to 15 years later than cutaneous melanomas.67-71 Whereas cutaneous melanoma is recognized as a disease among non-Hispanic Caucasians, a substantial number of mucosal melanomas are also diagnosed among African Americans, Hispanics, and the Japanese.61,72 Similar to cutaneous melanoma, a slight male predominance has been reported for mucosal tumors.69,71,73 The nasal cavity (NC) is the most common site for HN mucosal melanoma. Specically, the origin is most often along the anterior nasal septum, followed by the inferior and middle turbinates.70,73-75 The second most common site is the oral cavity (OC), where a predilection for the hard palate and maxillary alveolar gingivae has been reported. Laryngeal primary tumors account for less than 4% of all cases.74 Within the larynx, the supraglottis was the most common subsite involved. The presenting signs and symptoms for mucosal melanoma are quite different from cutaneous melanoma and correlate directly with the anatomic origin. Patients harboring NC primaries present most often with nasal obstruction and epistaxis.73-75 Proptosis, diplopia, facial pain, and facial asymmetry are less common and raise concern for advanced disease. Patients with OC mucosal melanoma are often asymptomatic, with the cancer going undiagnosed until a neck mass develops from regional metastasis.68,73 The most common local sign for OC tumors is a mass lesion.75 The sparse number of mucosal melanoma cases limits research to anecdotal reports. The paucity of cases simply precludes the ability to conduct meaningful clinical trials. It is recognized that mucosal melanoma does not share the same important prognostic markers as cutaneous melanoma.67 For example, the development of regional metastasis does not impact survival for patients with mucosal melanoma, yet it is the most important prognostic marker for cutaneous variants.70 For this reason, the melanoma staging system set forth by the AJCC and described herein specically applies to cutaneous, not mucosal, melanoma cases. The majority of mucosal melanoma patients present with localized disease. Only 18.7% will demonstrate stage III, regional spread, at the time of diagnosis. However, the high percentage of patients with localized disease is deceiving because local recurrence is the major reason for treatment failure.74 Fifty percent of patients recur locally, usually within
Curr Probl Surg, November 2006 789

12 months of diagnosis. Despite the fact that most patients present with stage I/II disease, the overall 5-year survival rate is a dismal 10% to 20%.69-71,74 Mucosal melanoma of the paranasal sinuses portends the worst prognosis, followed in decreasing order by OC, pharyngeal, and NC tumors.70 The rich vasculature and lymphatics of HN mucosa possibly contributes to the aggressive behavior of mucosal melanoma.74

Diagnostic Evaluation
History
The majority of melanoma lesions are rst detected by the patient or his/her partner.76,77 Less than one fourth of lesions are diagnosed during routine ofce physical examination; however, when lesions are detected by the physician, they tend to be thinner.77 Change in color, size, or shape of a lesion represent the earliest signs for melanoma. The earliest symptom is persistent pruritus. Bleeding, ulceration, and pain represent later signs and symptoms concerning for advanced disease. Patients should be questioned about a previous personal and family history of melanoma. Information including previous skin biopsies, sun exposure, history of blistering sunburns, tanning booth use, and occupation should be obtained. Johnson and colleagues investigated characteristics of 1515 melanoma patients and found that 81% recalled a history of at least 1 sunburn.77

Physical Examination
All patients who present with a suspicious lesion warrant a full body evaluation of the skin and nodal basins by a physician well versed in cutaneous cancers. This thorough examination is important because up to 8% of newly diagnosed patients can have multiple primary cutaneous melanomas.78-81 The differential diagnosis for cutaneous melanoma is quite broad, including: seborrheic keratosis, hemangioma, blue nevus, spitz nevus, pyogenic granuloma, pigmented basal cell carcinoma, and even cutaneous squamous cell carcinoma. To help educate both patients and physicians in early detection of melanoma, the American Cancer Society has published the ABCD checklist.82 Under these guidelines, concerning signs for melanoma include: Asymmetry in lesion appearance, Border irregularity such as scalloped, poorly circumscribed, or ill-dened margins, Color variation within a lesion, or Diameter greater than 6 mm. The ABCD checklist is quite helpful in identifying melanoma; however, it will not detect every case.81,83 It is important to realize that a subset of cancers
790 Curr Probl Surg, November 2006

such as nodular, amelanotic, and desmoplastic melanomas lack the common features of the ABCDs. For this reason, the Europeans implemented a 7-point checklist that focuses on the importance of change within an existing lesion.84 In 1 series, 88% melanoma patients (615 of 696) recalled change in their pigmented lesion before melanoma diagnosis.85 Due to the signicance of change, a proposal has been set forth to add E evolving changes to the traditional ABCD warning signs.86 Clinicians are hopeful that the new, more comprehensive ABCDE criteria will lead to even further detection of melanoma at an earlier stage.85 A nal, useful screening tool is the ugly duckling sign87 in which any pigmented lesion that appears signicantly and individually different from other surrounding lesions should be viewed with a high index of suspicion. This suspicion should remain high, even if the ugly duckling lesion lacks the traditional ABCD warning signs.

Biopsy
Any pigmented lesion that fullls any of the ABCD criteria, has undergone change, or appears different from surrounding nevi on the body, should undergo histologic evaluation. We consider the biopsy of potential melanoma as a distinct, 2-staged process. The rst step involves biopsy not only for tissue diagnosis, but also to evaluate important prognostic factors such as tumor depth, ulceration, mitotic rate, angiolymphatic invasion, and perineural spread. Ideally, a complete excisional biopsy, with narrow 1- to 2-mm clinical margins of surrounding skin, is performed. Some lesions are simply not amenable to excisional biopsy due to large size or anatomic location. In such cases, punch biopsy or incisional biopsy through the thickest portion of the neoplasm is recommended. Shave biopsy and ne needle aspiration are discouraged because the thickness of the tumor, which dictates further diagnostic evaluation and treatment, cannot be obtained. Unfortunately punch and incisional biopsies are subject to sampling error. For this reason, if a diagnosis of melanoma is not rendered following the initial procedure, repeat biopsy is recommended. Biopsy results obtained during the rst stage then serve as the guide for the second stage which entails wide local excision (WLE) using a 0.5 to 2 cm margin of normal surrounding tissue, with or without SLNB. The role of SLNB ultimately depends on the nal microstaging of the primary lesion and is detailed below. Obtaining wider margins at the time of the initial biopsy may seem both cost and time efcient. However, this practice is highly discouraged because clinical accuracy is uncertain, and removal of signicant amounts of skin surrounding the lesion may
Curr Probl Surg, November 2006 791

TABLE 2. University of Michigan Guidelines for Initial HN Melanoma evaluation Stage 0 (in situ) History & physical examination No routine tests Stage I (mm thickness 1) History & physical examination Consider SLNB for select pts; (see Table 11) Stage I-II (N0; any thickness) History & physical examination SLNB if indicated (see Table 11) CXR Optional LDH levels optional Stage III (N ; in transit) History & physical examination FNA neck mass CXR LDH Levels Other imaging studies if clinically indicated Stage IV (distant mets) History & physical examination CXR LDH levels Consider head, chest, abdomen, pelvis CT/MRI if symptomatic Other imaging studies per clinical trial SLNB, Sentinel lymph node biopsy; LDH, lactate dehydrogenase; CXR, chest radiograph; FNA, ne needle aspiration; CT, computed tomography; MRI, magnetic resonance imaging.

preclude accurate regional staging using lymphoscintigraphy and the SLNB technique.88 Instead, biopsy with narrow margins in a 2-staged fashion is advocated.

Metastatic Evaluation
The majority of patients presenting with melanoma have localized disease, are asymptomatic, and lack clinical ndings suggestive of regional or distant spread. In an attempt to standardize the staging evaluation for cutaneous melanoma, the National Comprehensive Cancer Network (NCCN) published guidelines that are available at: http:// www.nccn.org.89 The staging evaluation performed at our institution incorporates these guidelines and is outlined in Table 2.90 The foundation for melanoma evaluation in the setting of localized stage I disease remains a thorough history and physical examination. The most common site for distant metastasis is the lungs.88 Many physicians advocate a screening chest x-ray (CXR) because the study is viewed as an inexpensive, noninvasive means for metastatic evaluation. However, the incidence of occult pulmonary metastasis in
792 Curr Probl Surg, November 2006

TABLE 3. Review of systems for melanoma evaluation Skin/lymphatics Lesion change (size, shape, color) Pruritus Mass/nodule/enlarged lymph node Nonhealing skin lesions Easy bruising/bleeding New pigmented skin lesions Constitutional Weight loss/decreased appetite Malaise Weakness/fatigue Fever Respiratory Cough Hemoptysis Pneumonia Pleurisy Chest pain Dyspnea Hepatic Abdominal pain Back/scapula pain on inspiration Jaundice Adapted from Johnson TM, Chang A, Redman B, Rees R, Bradford CR, Riba M, et al. Management of melanoma with a multidisciplinary melanoma clinic model, J Am Acad Dermatol 2000; 42:820 6. Neurological/psychiatric Headache Memory disturbance Depression Focal neurologic decits Visual disturbances Balance problems Blackouts/seizures Numbness Local weakness Paralysis Mood changes Gastrointestinal Cramping/Abdominal pain Bleeding Nausea/anorexia Vomiting Constipation Musculoskeletal Bone pain

an asymptomatic patient with stage I or II disease is exceedingly low.91,92 We evaluated 210 patients with a screening CXR and found the true positive rate, dened as the percentage of CXRs interpreted as positive or possibly melanoma related to be 0%.93 Yet, the high false positive rate of 7% necessitated additional and costly evaluations. Similarly, evidence supporting the use of other screening modalities such as computed tomography (CT),94,95 liver-spleen scans, magnetic resonance imaging (MRI), and bone scans for patients with limited stage I and II disease is lacking.96,97 For stage II and III patients who lack clinical evidence of regional disease (N-zero neck), CXR and screening lactate dehydrogenase (LDH) levels are deemed optional under the NCCN guidelines. It has been our experience that screening LDH carries a 15% false positive rate, does not correlate with SLN status, and has not been helpful in detecting occult
Curr Probl Surg, November 2006 793

TABLE 4. Melanoma TNM classication T classication T1 T2 T3 T4 N classication N1 N2 Thickness 1.0 mm 1.01-2.0 mm 2.01-4.0 mm 4.0 mm No. of metastatic nodes 1 node 2-3 nodes a: b: a: b: a: b: a: b: Ulceration status without ulceration and level II/III with ulceration or level IV/V without ulceration with ulceration without ulceration with ulceration without ulceration with ulceration Nodal metastatic mass a: micrometastasis* b: macrometastasis a: micrometastasis* b: macrometastasis c: in transit met(s)/satellite(s) without metastatic nodes

N3

4 or more metastatic nodes, or matted nodes, or in transit met(s)/satellite(s) with metastatic node(s) Site Distant skin, subcutaneous, or nodal mets Lung metastases All other visceral metastases Any distant metastasis Serum lactate dehydrogenase Normal Normal Normal Elevated

M classication M1a M1b M1c

*Micrometastases are diagnosed after sentinel or elective lymphadenectomy. Macrometastases are dened as clinically detectable nodal metastases conrmed by therapeutic lymphadenectomy or when nodal metastasis exhibits gross extracapsular extension. From Balch CM, Buzaid AC, Soong SJ, Atkins MB, Cascinelli N, Coit DG, et al. Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 2001;19:3635-48.

disease in asymptomatic patients.93 For this reason these studies are ordered at our institution only when the history or physical examination reveals jaundice, abdominal pain, or other specic ndings concerns for distant metastasis.90 A comprehensive list of concerning symptoms warranting a focused investigation for distant metastasis is summarized in Table 3.98 Patients with stage III disease who present with clinically or radiographically suspicious lymph nodes, satellite lesions, or in-transit lesions (dened by melanoma located more than 2 cm from the primary lesion), carry a signicant risk of distant metastasis. Fine
794 Curr Probl Surg, November 2006

TABLE 5. Proposed stage groupings for cutaneous melanoma Clinical staging* T O IA IB IIA IIB IIC III Tis T1a T1b T2a T2b T3a T3b T4a T4b Any T N N0 N0 N0 N0 N0 N0 N0 N0 N0 N1 N2 N3 M M0 M0 M0 M0 M0 M0 M0 M0 M0 M0 T Tis T1a T1b T2a T2b T3a T3b T4a T4b Pathologic staging N N0 N0 N0 N0 N0 N0 N0 N0 N0 M M0 M0 M0 M0 M0 M0 M0 M0 M0

IIIA IIIB

IIIC

IV

Any T

Any N

Any M1

T1-4a T1-4a T1-4b T1-4b T1-4a T1-4a T1-4a/b T1-4b T1-4b Any T Any T

N1a N2a N1a N2a N1b N2b N2c N1b N2b N3 Any N

M0 M0 M0 M0 M0 M0 M0 M0 M0 M0 Any M1

*Clinical staging includes microstaging of the primary melanoma and clinical/radiologic evaluation for metastases. By convention, it should be used after complete excision of the primary melanoma with clinical assessment for regional and distant metastases. Pathologic staging includes microstaging of the primary melanoma and pathologic information about the regional lymph nodes after partial or complete lymphadenectomy. Pathologic stage 0 or stage 1A patients are the exception; they do not require pathologic evaluation of their lymph nodes. There are no stage III subgroups for clinical staging. From Balch CM, Buzaid AC, Soong SJ, Atkins MB, Cascinelli N, Coit DG, et al. Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 2001;19:3635-48.

needle aspiration has been shown to be an accurate and cost-effective means of conrming metastatic melanoma within a lymph node.99 CXR and LDH should be ordered for all patients diagnosed with regional disease. The patients history and physical examination dictate whether additional imaging studies are required to evaluate specic distant sites (Table 3). Patients with known stage IV disseminated melanoma require a complete evaluation for systemic metastasis. Clinical trial protocols often dictate the evaluation in this setting. Unfortunately, a survival benet has
Curr Probl Surg, November 2006 795

TABLE 6. 2002 AJCC cutaneous melanoma staging criteria based on the most important prognostic markers of survival* Stages I and II localized disease 1) Tumor thickness (even integer tumor thickness) 2) Tumor ulceration 3) Histologic (Clarks) level of invasion for thin T1 lesions only Stage III regional disease 1) Number of metastatic lymph nodes 2) Tumor burden (microscopic vs. macroscopic nodal disease) 3) Primary tumor uleration Stage IV distant disease 1) Site of distant metastasis 2) Elevated LDH LDH, Lactate dehydrogenase. *Adapted from Schmalbach CE, Johnson TM, Bradford CR. The management of head and neck melanoma. In: Cummings CW, Flint PW, Harker LA, Haughey BH, Richardson MA, Robbins KT, et al, editors. Cummings Otolaryngology Head & Neck Surgery. Philadelphia: Elsevier Mosby; 2005.

not been found for patients who are asymptomatic when diagnosed with distant, stage IV disease compared with their counterparts who are diagnosed with symptomatic, stage IV disease.90 However, a thorough evaluation for systemic metastasis may lead to improvement in patient quality of life.

Prognostic Factors and Tumor Staging


To gain increased understanding of the natural history and behavior of cutaneous melanoma, the AJCC Melanoma Task Force conducted a multi-intuitional study, comprised of 13 major cancer centers and 17,600 patients.100 This investigation marks the largest analysis of its kind. In 2002, results from the landmark study were used to revise the AJCC cutaneous melanoma staging system.100,101 The goal in modifying the classication system was 2-fold. First, the Staging Committee wanted to develop categories based on the most important, independent prognostic markers for melanoma. This goal was established in an attempt to better identify cohorts of patients sharing similar survival rates, thereby leading to increased homogeneity for future, meaningful clinical trials. The second goal was to provide physicians with a practical classication system that truly mirrored clinical practice. For this reason, the Committee used physician input related to both clinical management and research protocols.
796 Curr Probl Surg, November 2006

TABLE 7. Revisions to the AJCC staging system 1) Tumor thickness (Breslow depth measured in mm) and ulceration are now utilized in T classication. Clarks histologic level of invasion is now only applicable in the staging of thin ( 1 mm) T1 lesions. 2) The number of metastatic lymph nodes, as opposed to the gross size of the metastatic node, denes the N category. 3) The technique of sentinel lymph node mapping and biopsy (SLNB) is now incorporated into the staging system. Delineation of occult (microscopic/ subclinical) nodal metastasis vs. clinically/radiographically apparent (macroscopic) nodal metastasis is outlined within the N category. 4) Both anatomic site of distant metastasis and elevated LDH level are utilized in the M classication. 5) Ulceration of the primary lesions upstages the patient. 6) Primary lesions with surrounding satellite metastasis or in-transit metastasis are dened as stage III regional disease regardless of the nodal status.

Summary of Revisions
The 2002 AJCC staging system for cutaneous melanoma remains founded on the traditional Tumor-Node-Metastasis (TNM) classication system (Tables 4 and 5). Stages I and II represent localized disease, stage III is regional disease in which the melanoma has spread to draining nodal basins, and stage IV is reserved for distant metastatic disease, most commonly to the lungs and liver. Table 6 summarizes the most important predictors for melanoma survival, all of which are now used in classifying tumor stage. The comprehensive, multiinstitutional study conducted by the AJCC led to 6 major revisions in the staging system (Table 7).

T Classication/Localized Disease
Tumor thickness and ulceration emerged as the 2 most important predictors for outcome when multivariate analysis of 13,581 patients with localized disease was performed.100 Overall, tumor thickness was the most powerful prognostic indicator for this subgroup. Tumor thickness was previously incorporated in the 1997 version of the staging system. However, the 0.75-mm cut point between T1 and T2 lesions was empirically based on the Breslow classication.102 The revised staging system now uses practical, even-integer cut points of 1.0, 2.0, and 4.0 mm to delineate T stage, since these cut points represent the best statistical t in correlating tumor thickness and survival. In the 1997 staging system, the histologic level of invasion, as represented by the Clark scale was incorporated into the T classication.
Curr Probl Surg, November 2006 797

798 TABLE 8. Survival rates for melanoma TNM and staging categories Pathologic Thickness TNM stage (mm) IA IB IIA IIB IIC IIIA IIIB T1a T1b T2a T2b T3a T3b T4a T4b N1a N2a N1a N2a N1b N2b N1b N2b N3 M1a M1b M1c 1 1 1.01-2.0 1.01-2.0 2.01-4.0 2.01-4.0 4.0 4.0 Any Any Any Any Any Any Any Any Any Any Any Any Ulceration No Yes or level IV, V No Yes No Yes No Yes No No Yes Yes No No Yes Yes Any Any Any Any No. nodes 0 0 0 0 0 0 0 0 1 2-3 1 2-3 1 2-3 1 2-3 4 Any Any Any Nodal size Distant metastasis No. of patients 4,510 1,380 3,285 958 1,717 1,523 563 978 252 130 217 111 122 93 98 109 396 179 186 793 17,600 Survival 1-Year 99.7 99.8 99.5 98.2 98.7 95.1 94.8 89.9 95.9 93.0 93.3 92.0 88.5 76.8 77.9 74.3 71.0 59.3 57.0 40.6 0.1 0.1 0.1 0.5 0.3 0.6 1.0 1.0 1.3 2.4 1.8 2.7 2.9 4.4 4.3 4.3 2.4 3.7 3.7 1.8 2-Year 99.0 98.7 97.3 92.9 94.3 84.8 88.6 70.7 88.0 82.7 75.0 81.0 78.5 65.6 54.2 44.1 49.8 36.7 23.1 23.6 0.2 0.3 0.3 0.9 0.6 1.0 1.5 1.6 2.3 3.8 3.2 4.1 3.7 5.0 5.2 4.9 2.7 3.6 3.2 1.5 SE 5-Year 95.3 90.9 89.0 77.4 78.7 63.0 67.4 45.1 69.5 63.3 52.8 49.6 59.0 46.3 29.0 24.0 26.7 18.8 6.7 9.5 0.4 1.0 0.7 1.7 1.2 1.5 2.4 1.9 3.7 5.6 4.1 5.7 4.8 5.5 5.1 4.4 2.5 3.0 2.0 1.1 10-Year 87.9 83.1 79.2 64.4 63.8 50.8 53.9 32.3 63.0 56.9 37.8 35.9 47.7 39.2 24.4 15.0 18.4 15.7 2.5 6.0 1.0 1.5 1.1 2.2 1.7 1.7 3.3 2.1 4.4 6.8 4.8 7.2 5.8 5.8 5.3 3.9 2.5 2.9 1.5 0.9 IIIC IV Micro Micro Micro Micro Macro Macro Macro Macro Micro/macro Any Skin, SQ Any Lung Any Other Visceral Curr Probl Surg, November 2006 Total From Balch CM, Buzaid AC, Soong SJ, Atkins MB, Cascinelli N, Coit DG, et al. Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 2001;19:3635-48.

FIG 1. Survival curves of 1528 melanoma patients with lymph node metastases subgrouped by actual number of metastatic lymph nodes. (From Balch CM, Soong SJ, Gershenwald JE, Thompson JF, Reintgen DS, Cascinelli N, et al. Prognostic factors analysis of 17,600 melanoma patients: validation of the American Joint Committee on Cancer melanoma staging system. J Clin Oncol 2001;19:362234.)

However, recent analysis by Balch and colleagues found the histologic level of invasion to be prognostic only for thin T1 lesions.100 For tumors up to 1 mm without ulceration, invasion to Clark level II or III is considered T1a, whereas invasion into the reticular dermis (level IV or V) is considered T1b. With the exception of T1 lesions, the Clark level of invasion is no longer used in staging melanoma. Tumor ulceration was the second most important prognostic indicator for patients with localized disease. It is important to note that ulceration is not a visible crater on gross examination, but rather a histologic diagnosis in which intact epidermis overlying the melanoma is absent. Patients with ulcerated primary lesions were found to have signicantly lower survival rates compared with their nonulcerated counterparts (Table 8). Closer analysis revealed that the survival of patients with an ulcerated tumor mirrored that of nonulcerated patients in the next highest T category, as opposed to their original T category. For this reason, ulceration warrants upstaging of tumors. The prognostic signicance of
Curr Probl Surg, November 2006 799

FIG 2. Survival curves of 1429 patients with lymph node metastases subgrouped by presenting
clinical stage. Survival rates calculated signicantly different for the 2 groups (P Thompson JF, Reintgen DS, Cascinelli N, patients: validation of the American Joint Oncol 2001;19:3622-34.) from the time of primary melanoma diagnoses were 0.0001). (From Balch CM, Soong SJ, Gershenwald JE, et al. Prognostic factors analysis of 17,600 melanoma Committee on Cancer melanoma staging system. J Clin

ulceration,103 as well as the correlation between ulceration and mitotic rate, has been reported.104,105 Although mitotic rate was not evaluated during the revision of the AJCC staging system, it may emerge as an important prognostic marker in the future.

N Classication/Regional Disease
Similar multivariate analysis was performed for more than 1000 melanoma patients diagnosed with lymph node metastasis. Three statistically signicant prognostic factors were identied: number of metastatic lymph nodes, tumor burden as represented by microscopic vs. macroscopic disease, and primary tumor ulceration. The gross diameter of metastatic nodes was previously used to dene the N category in the 1997 staging system. However, this most recent study failed to nd a meaningful predictive value for metastatic nodal size, and it is no longer incorporated into melanoma staging.100,106,107
800 Curr Probl Surg, November 2006

TABLE 9. Five-year survival rates for stage III (nodal metastases) patients stratied by number of
metastatic nodes, ulceration, and tumor burden
Microscopic 1 Melanoma ulceration % Absent Present 69 52 Node SE No. 2-3 Nodes % SE No. >3 Nodes % 1 Nodes SE No. Macroscopic 2-3 Nodes % SE No. >3 Nodes % SE No.

SE No. % 9.3 57 59 8.8 46 29

3.7 252 63 4.1 217 50

5.6 130 27 5.7 111 37

4.7 122 46 5.0 98 25

5.5 93 27 4.4 109 13

4.6 109 3.5 104

From Balch CM, Soong SJ, Gershenwald JE, Thompson JF, Reintgen DS, Cascinelli N, et al. Prognostic factors analysis of 17,600 melanoma patients: validation of the American Joint Committee on Cancer melanoma staging system. J Clin Oncol 2001;19:3622-34.

The most important predictive marker for patients with nodal metastasis was the number of positive lymph nodes. Survival curves are depicted in Fig 1. The greatest difference in 5-year survival rates was used to subclassify the N category. Patients with 1 metastatic node are now categorized as N1, patients with 2 or 3 metastatic nodes as N2, and patients with 4 or more nodes as N3. Tumor burden was identied as the second most important prognostic indicator for patients with regional metastasis. Patients identied with microscopic nodal disease (also referred to as occult or subclinical nodal disease), either through SLNB or elective lymphadenectomy, were found to have a signicantly better survival compared with individuals diagnosed clinically or radiographically with gross, macroscopic disease (Fig 2). This difference was so compelling that microscopic versus macroscopic nodal disease is now a subclassication within the N category. In addition, the AJCC Melanoma Committee strongly recommends staging with SLNB for patients with T2N0M0, T3N0M0, and T4N0M0 disease before entry into clinical trials.101 It is the Committees hope that identication of occult nodal disease will lead to accurate staging and increased homogeneity among investigational cohorts. Ulceration was identied as the only primary tumor characteristic indicative of outcome for patients with stage III disease. Depth of invasion, as represented by tumor thickness, was no longer prognostic once adjustment was made for regional metastasis (P 0.16). Intralymphatic metastasis is new a criterion now used in classifying stage III regional disease. The presence of satellite metastasis surrounding a primary lesion, and in-transit metastasis identied between the primary melanoma and draining nodal basin, emerged as poor prognostic indicators.100,107,108 Both ndings portend a prognosis similar to nodal metastasis. Under the new staging system, satellite metastasis and in-transit metastasis are classied as N2C disease, even in the absence of nodal
Curr Probl Surg, November 2006 801

TABLE 10. Recommended surgical margins for primary cutaneous melanoma Tumor thickness (mm) In situ 1.0 1.012.0 2.0 Surgical margin (cm) 0.5 1.0 1.02.0 2.0

disease (N-zero). If synchronous nodal metastasis is found along with satellite or in-transient metastasis, the prognosis is exceedingly poor. These patients are automatically upstaged to N3 disease regardless of the number of positive lymph nodes. Overall 5-year and 10-year survival for patients with metastatic nodal disease was 49% and 37%, respectively.100 However, reported survival rates for stage III patients were quite broad, ranging from a dismal 13% in the setting of an ulcerated primary lesion with macroscopic disease identied in 4 lymph nodes to a promising 69% survival in the setting of a nonulcerated primary tumor with microscopic disease conned to 1 lymph node (Table 9). This nding conrms previous studies that demonstrated that stage III disease represents a heterogeneous group of patients.106,109 It further highlights the critical impact on survival afforded by the early diagnosis of regional metastasis, as well as the necessity for accurate pathologic staging of all patients enrolled in clinical trials.

M Classication/Distant Metastasis
A total of 1158 patients in the study were diagnosed with disseminated stage IV disease at the time of presentation. The most signicant prognostic factor correlated to anatomic site of distant metastasis. Patients with skin, subcutaneous tissue, or distant lymph node involvement (M1a) had a slightly higher survival rated compared with patients with lung metastasis (M1b). Overall, patients with metastasis involving other visceral organs (M1c) had the worst prognosis. Previous studies have also identied elevated LDH as a poor prognostic marker.110-113 For this reason, any patient with elevated LDH and distant metastasis, regardless of site, is classied as M1c. Overall survival for patients with distant metastasis is extremely grave, measured in months as opposed to years. The median survival time following diagnosis of disseminated disease is only 6 to 8 months, with a dismal 5-year survival rate of 6%.110,114 For this reason, the AJCC staging system does not subclassify stage IV melanoma.
802 Curr Probl Surg, November 2006

Anatomic Site
The anatomic site of the primary lesion is not used within the formal staging system. However, prognostic differences have been reported in the literature.6,115 A review of 6300 cutaneous melanomas treated at the Duke Medical Center identied the worst 10-year survival rate among HN primary tumors (54%) compared with trunk (61%), lower extremity (71%), and upper extremity (76%) sites.115 A 50% tumor recurrence rate has been reported for HN cutaneous melanomas.116,117 One explanation for this aggressive behavior and higher local recurrence is the development of thicker melanoma lesions within the HN region compared with other sites. In addition, the surgeons may be reluctant to clear tumor margins adequately and to remove peripheral AJMH in the HN region due to the visible anatomic location and related psychosocial implications.

Surgical Management of the Primary Tumor


Wide Local Excision and Surgical Margins
The standard of care for primary melanoma treatment is complete surgical excision. However, the extent of surgical margins remains an unanswered question despite numerous retrospective studies, meta-analyses, and clinical trials. Historically, an extensive 5-cm margin of surrounding normal tissue was practiced. However, this recommendation was based on a 1907 autopsy report of a patient with advanced melanoma.118 The use of a 5-cm surgical margins was routine practice until the 1970s when Breslow and Macht challenged the concept by successfully treating a cohort of 35 patients with thin melanomas using narrower margins.119 Several prospective, randomized trials investigating surgical margins for cutaneous melanoma have since followed. The World Health Organization (WHO) conducted an international trial in which 612 patients with thin melanomas (up to 2 mm) were randomized to surgical excision with 1-cm vs. greater than 3-cm margins.120 At a mean follow-up of 8 years, the disease-free survival and overall survival rates were reported to be equivalent between the 2 groups.121 The WHO concluded that wide excision did not inuence survival for patients with thin melanomas; for patients with melanomas less than 1 mm in thickness, the authors advocated narrow 1-cm margins to the muscular fascia plane. Within the WHO trial, a subset of 245 patients had tumors measuring 1.1 to 2.0 mm in thickness. Although a difference in disease-free survival and overall survival was not observed with respect to margins, a local recurrence rate of 3.3% was reported among patients undergoing narCurr Probl Surg, November 2006 803

row excision. This nding prompted the Intergroup Melanoma Surgical Trial, which prospectively randomized 740 patients with intermediate thickness (1-4 mm) melanomas to WLE with 2-cm vs. 4-cm margins.122 Local recurrence rates and 10-year survival rates were reported to be equivalent between the 2 groups. This nding led to the recommendation of a 2-cm surgical margin for patients with intermediate melanomas measuring 1.1 to 4.0 mm in thickness. The most recent prospective clinical trial conducted by the United Kingdom Melanoma Study Group randomized 900 patients with localized cutaneous melanomas of at least 2 mm in thickness to 1-cm vs. 3-cm margins.123 A statistically signicant difference was not identied between the 2 groups when local, regional, and distant recurrences were individually compared. Overall mortality rates were found to be identical between the 2 arms. However, when all recurrences (local, in-transit, and nodal) were pooled together, the 1-cm margin group experienced a statistically higher recurrence rate. This is the rst clinical trial comparing tumor margins to report a statistically signicant difference in tumor recurrence. From a practical standpoint, however, it is the 1-cm vs. 2-cm margin that is debated more often in the clinical setting.124 No prospective randomized trial has investigated the optimal surgical margin for thick ( 4 mm) melanomas. A retrospective study of 278 thick melanomas found that surgical margins greater than 2 cm did not lead to a difference in local recurrence rate, disease-free survival, or overall survival when compared with margins less than 2 cm.125 Within this study, 16% of the tumors involved HN subsites. The primary goal of melanoma excision is to eliminate local recurrence secondary to persistent disease. The rate of local recurrence from narrow-margin excisions is admittedly low; however, the consequences are potentially fatal. It has been estimated that 100% achievement of ideal margins would lead to a reduction in melanoma-related mortality and an increase of life expectancy of melanoma patients by 0.4 years.126 Although this difference appears small at rst glance, it equates to an estimated 11 additional years of life expectancy for those individuals who would have recurred locally following a 1-cm margin, but instead achieved a disease-free state following a wider surgical margin. Current guidelines for surgical margins are based on primary tumor thickness (Table 10). It is important to realize that these recommendations serve merely as a guideline. Each melanoma case must be individualized. The depth of excision includes full thickness skin and underlying subcutaneous tissue. Resection of fascia, perichondrium, and periosteum
804 Curr Probl Surg, November 2006

is required only in the setting of direct tumor invasion or if the surgical plane was violated during a previous biopsy.127 LMM warrants special consideration because it has a propensity for wide subclinical spread, which often results in positive margins.128 In an attempt to address this challenge, we developed the square procedure.53,129 This staged procedure entails complete excision of the peripheral margins using a double-bladed instrument, followed by permanent histologic evaluation of 100% of the peripheral margins surrounding the entire tumor.

Closure and Reconstruction


The majority of surgical sites can be closed primarily with the use of wide undermining. Larger defects may require reconstruction with a split thickness skin graft, full thickness skin graft, local advancement ap, or regional ap. The method of reconstruction depends on the anatomic location including skin color and texture, depth of the defect, and patient, as well as surgeon, preference. Initially surgeons were reluctant to graft excision sites for fear that surveillance for future melanoma recurrence within the surgical bed would be hindered. However, the method of closure has not been shown to impact survival.130 Once clear margins have been conrmed, surgeons are encouraged to close surgical defects using the technique that they think will yield the best cosmetic result.

Auricular Melanoma
Originally, auricular melanoma was thought to carry a worse prognosis compared with other sites within the HN region.117,131,132 This increased risk was attributed to rich lymphatics, complex anatomic subdivisions of the ear, and a paucity of subcutaneous tissue between the thin auricular skin and underlying perichondrium.133 For these reasons, full thickness excision or total auriculectomy was often advocated. Research conducted over the past decade has led to a shift in the treatment paradigm for auricular melanoma. After accounting for tumor thickness, recent studies have demonstrated that melanoma in this region carries the same prognosis as other HN sites.88,134 Outcome differences were not observed between auricular subsites.133 In addition, retrospective reviews failed to demonstrate a difference in local recurrence based on the extent of surgical excision, even when perichondrium was preserved.134 Today, the same prognostic indicators and surgical principles of obtaining wide, clear margins for treatment of cutaneous melanoma can be applied safely to the auricle.
Curr Probl Surg, November 2006 805

Surgical Management of Regional Lymph Nodes


Therapeutic Lymph Node Dissection
The most common sites for metastasis of HN cutaneous melanoma are the cervical and parotid lymph node basins.7,135,136 Therapeutic lymph node dissection (TLND) is accepted universally as the treatment of choice for regional disease. The neck dissection must include all draining nodal basins as well as the intervening lymphatics between the primary tumor and the site of regional disease. The location of the primary tumor dictates the specic type of TLND, as well as the need for a supercial parotidectomy. In the absence of gross tumor involvement, or disruption from open biopsy or previous surgical dissection, concerted efforts should be made to preserve the spinal accessory nerve, internal jugular vein, and sternocleidomastoid muscle.137 Melanomas of the anterolateral scalp, temple, lateral forehead, lateral cheek, and ear, arising anterior to an imaginary coronal plane through the external auditory canals (EACs), drain via the parotid nodal basin to the jugular lymph node chain.137 For this reason, melanomas anterior to this coronal plane require a supercial parotidectomy and modied radical neck dissection (MRND). If the melanoma arises in a more inferior location, such as the chin or neck, a supercial parotidectomy is not warranted. Melanomas located on the scalp and occiput, posterior to the imaginary coronal plane through the EACs, can drain to postauricular, suboccipital, and posterior triangle lymph nodes. These nodal basins are not addressed during routine MRND. In this situation, a posterolateral neck dissection, which extends to the midline of the posterior neck, is required.138

Elective Lymph Node Dissection


Historically, 1 of the most controversial debates in melanoma surrounded treatment of regional nodal basins in the absence of clinical metastasis (prophylactic treatment of the N-zero neck). Melanomas measuring less than 1.0 mm in thickness have an excellent prognosis, with a 5-year survival rate approaching 95% to 99%. For this reason, elective treatment of the neck is considered unnecessary for the majority of thin stage I melanomas since the risk of occult nodal metastasis is less than 5%. Conversely, melanomas measuring greater than 4.0 mm in thickness have an extremely poor prognosis. The high 70% rate of systemic metastasis is thought to negate any benet that may be gained by electively treating regional nodal basins.88 The real controversy sur806 Curr Probl Surg, November 2006

TABLE 11. University of Michigan considerations for melanoma sentinel lymph node mapping and
biopsy

Clinically localized melanoma 1 mm No signicant comorbidities No previous wide excision (usually) 0.750.99 mm, with Ulceration Extensive regression to 1 mm Young age High mitotic rate Any site, any age, any number of basins After discussion of risks and benets

rounded elective treatment of the neck for patients with intermediate thickness (1.0-3.9 mm) melanomas. Opponents of elective lymph node dissection (ELND) contended that melanoma metastasis is unpredictable. In Fishers retrospective review of 1444 HN melanoma patients, up to 16% developed distant metastasis in the absence of regional disease.7 The potential for hematogenous melanoma spread to bypass regional nodal basins theoretically limits the utility of an ELND. Opponents further argue that all 4 prospective, randomized trials failed to demonstrate an overall survival benet for patients undergoing ELND in the absence of regional metastasis.139-142 In 1967 the WHO Melanoma Group conducted the rst prospective, randomized trial (No. 1) between 1967 and 1974.141 A total of 535 patients with stage I and II melanoma of the extremity were enrolled. No difference in survival benet was found between patients who underwent WLE and observation, with TLND reserved for the development of gross nodal metastasis, compared with patients who underwent WLE and ELND. Similarly, surgeons at the Mayo Clinic randomized 171 patients with stage I disease to: 1) WLE and observation, 2) WLE and delayed (30-60 days) ELND, or 3) WLE with concomitant ELND.139 ELND was not found to provide a survival benet compared with observation. Although both prospective trials represent pioneering research in a challenging area, both study designs have been criticized.135 At the time of the studies, the prognostic signicance of tumor thickness and ulceration was unknown. Later analysis of the WHO Melanoma Group Trial No. 1 found signicant discrepancy in the distribution of tumor thickness between the 2 treatment arms. Furthermore, 52% of lesions in the ELND group were ulcerated compared with only 19% in the observation group.135 Subsequent re-analysis of these data with respect to tumor thickness and ulceration identied a subset of patients with a 22%
Curr Probl Surg, November 2006 807

FIG 3. Preoperative lymphoscintigraphy is performed approximately 3 hours before SLNB. A total of


2 Ci of technetium Tc 99m sulfur colloid (CIS-US, Inc., Bedford, MA) is injected intradermally into the 4 quadrants surrounding the primary melanoma lesion located midline on the posterior occiput. (A) The right posterior occiput primary lesion with an overlying shield. (B) Lateral views taken 45 minutes after injection. An SLN is observed in the right posterior triangle. Reprinted from Schmalbach C, Johnson T, Bradford C. In: Cummings Otolaryngology, The Management of Head and Neck Melanoma. 4th Ed. Elsevier Inc. pp 563-564, 2005 with permission.

improved 10-year survival in the setting of ELND. The accuracy of clinical staging within the trial was also questioned because several institutions reported a 30% rate of occult nodal metastasis. This rate is quite high compared with reports in the literature that range from 10% to 20%. Last, Balch argues that the failure to detect a survival benet with ELND was not surprising, given that both trials included patients with an overall low risk for regional metastasis at the time of diagnosis.135
808 Curr Probl Surg, November 2006

FIG 4. After administration of anesthesia, intraoperative lymphatic mapping is performed using approximately 1 mL of isosulfan blue dye (Lymphazurin 1%, Hirsh Industries, Inc., Richmond, VA) injected into the intradermal layer surrounding the primary melanoma. Reprinted from Schmalbach C, Johnson T, Bradford C. In: Cummings Otolaryngology, The Management of Head and Neck Melanoma. 4th Ed. Elsevier Inc. pp 563-564, 2005 with permission.

Approximately 85% of patients enrolled in the WHO Melanoma Group Trial No.1 were women with extremity melanomas, a group that is recognized to have a low rate of metastasis compared with other sites. In addition, the Mayo Clinic excluded patients with HN and midline trunk melanomas. To address these concerns, the Intergroup Melanoma Surgical Trial (IMST) was initiated.142 The IMST was a prospective, multi-institutional study of 740 patients with intermediate tumor thickness (1-4 mm) melanomas of the trunk, extremity, and HN region. Patients were once again randomized to WLE and observation vs. WLE and ELND. Cox regression analysis identied ulceration, site, tumor thickness, and age as independent markers for survival. Overall 5-year survival rates were not found to be different between the 2 treatment groups. However, a signicant survival benet was found in patients 60 years of age and younger who underwent ELND, especially if their tumor was nonulcerated or measured 1 to 2 mm in thickness. Although this subgroup analysis
Curr Probl Surg, November 2006 809

FIG 5. Wide local excision of the primary melanoma is performed before SLNB. Otherwise, the close proximity of the primary lesion to the draining nodal basins within the head and neck region will cause signicant radioactive shine-through and difculty in localizing the SLN. Each SLN is then identied using a combination of the gamma probe, which detects radioactive activity from the Tc 99m sulfur colloid, and visual cues from the isosulfan blue dye. Reprinted from Schmalbach C, Johnson T, Bradford C. In: Cummings Otolaryngology, The Management of Head and Neck Melanoma. 4th Ed. Elsevier Inc. pp 563-564, 2005 with permission.

is subject to the shortcomings of retrospective review, and it is criticized because patients were not randomized on age, patients were randomized prospectively on tumor thickness and ulceration. The fourth prospective trial was initiated in 1982 by the WHO Melanoma Group (No. 14). In an attempt to study patients truly at high risk for occult nodal metastasis, 240 patients with trunk melanomas measuring greater than 1.5 mm in thickness were enrolled.140 A difference in survival was not observed between patients randomized to observation vs. ELND. In a multivariate analysis including sex, age, tumor thickness, and treatment, only sex and tumor thickness were found to have a signicant impact on survival. However, this study did identify a statistically signicant 5-year survival difference for patients with micrometastasis identied during ELND (47%) compared with patients in the observation arm who underwent TLND only after the development of
810 Curr Probl Surg, November 2006

gross nodal disease (27%). For this reason, the WHO Melanoma Group advocated early detection of nodal metastasis using procedures such as SLNB. Statistical power remains 1 of the greatest challenges in investigating the survival benets of ELND and early detection of nodal disease.143 Only 20% of melanoma patients presenting with localized disease actually harbor occult nodal metastasis. It is only this 20% who would potentially benet from early removal of nodal basins. Adjuvant melanoma therapy imparts a survival benet in 25% to 50% of cases. If a similar survival benet is applied to the ELND group, 25% to 50% of the 20% of patients with occult disease should benet. In other words, only 5% to 10% of patients undergoing ELND are expected to experience a survival advantage. Detecting this small difference requires extremely large clinical trial enrollment, numbering in the thousands. McMasters and colleagues point out that the IMST, WHO, and Mayo Clinic trials lacked adequate statistical power to detect this small survival benet. For this reason, the group concluded that the 4% survival benet observed in the elective lymph node dissection (ELND) group of the IMST study is clinically signicant, despite the fact that statistical signicance was not reached. In summary, numerous prospective, randomized trials have failed to demonstrate an overall survival benet for patients undergoing ELND.139-146 Therefore, routine ELND is no longer advocated for melanoma. Instead, the procedure has been replaced by SLNB.

Sentinel Lymph Node Biopsy


Sentinel lymph node biopsy represents a minimally invasive, costeffective, and efcient means of screening patients for regional metastasis. Nodal status is currently recognized as the single most important prognostic factor for melanoma patients.101 Ten percent to 20% of individuals harbor occult, microscopic nodal disease. In an attempt to identify this small group of patients who warranted TLND, while sparing the remaining 80% of patients without regional disease the morbidity associated with a neck dissection, Morton and colleagues introduced SLNB for the evaluation of patients with trunk and extremity cutaneous melanoma.146 These investigators demonstrated that the status of the SLN accurately represented the status of the entire nodal basin from which it was obtained. SLNB is the best staging modality for regional disease, with the highest sensitivity and specicity of any modality currently available. Among major melanoma cancer centers across the country, it is now accepted as the standard of care.147-149
Curr Probl Surg, November 2006 811

Success of SLNB hinges on appropriate patient selection. Patients presenting with palpable regional disease or distant metastasis are not candidates for SLNB because additional prognostic information will not be gained. In addition, patients who have undergone previous neck dissection or resection of the primary site with wide margins are not deemed candidates due to lack of accuracy. Guidelines for SLNB practiced at our institution are summarized in Table 11. The SLN technique has evolved to include preoperative lymphoscintigraphy by nuclear medicine.150 Approximately 2 to 4 hours before surgery, patients undergo intradermal injection of a radioactive colloid into the 4 quadrants surrounding the primary melanoma tumor. Lymphoscintigraphy is then performed to guide the surgeon in determining the number, location, and laterality of nodal basins at risk for metastatic disease (Fig 3). It is particularly helpful in the setting of midline HN melanomas that have the potential for bilateral lymphatic drainage. Once under anesthesia, intraoperative lymphatic mapping with isosulfan blue dye (Lymphazurin 1%, Hirsch Industries, Inc., Richmond, VA) is performed.146 Approximately 1 mL of dye is injected into the intradermal layer surrounding the primary melanoma lesion (Fig 4). Unlike melanoma of the trunk and extremity, the primary tumor and draining lymphatics are in close proximity within the HN region. Therefore, WLE of the primary tumor is performed rst to reduce radioactive shine-through, which will render the intraoperative gamma probe useless in identifying SLNs (Fig 5). Following WLE of the primary melanoma, nodal basins at risk for metastasis are evaluated for increased radioactivity using a handheld gamma probe. A 1- to 3-cm incision was made overlying the areas of increased radioactivity. A preauricular incision is recommended for SLNB in the parotid region. Facial nerve monitoring is also recommended in this setting. SLNs are then identied using a combination of the gamma probe and visual cues from the blue dye (Fig 5). Each SLN is individually dissected from surrounding tissue. Within the parotid bed, gentle dissection in the anticipated direction of the facial nerve is imperative. The staging procedure is considered complete when all nodal basins demonstrate minimal background radioactivity ( 10%) relative to the primary lesion and sentinel nodes. Histopathologic protocols for SLN evaluation vary from institution to institution. At the University of Michigan, all SLNs are sent for histologic evaluation using permanent sections. We do not use frozen sections because this practice is less reliable, carrying a false negative rate between 5% to 10%.151 Our evaluation includes serial sectioning (5- m
812 Curr Probl Surg, November 2006

thick sections) and staining with hematoxylin and eosin (H&E). All SLNs negative on H&E staining are then subjected to melanoma-specic immunohistochemical staining (IHCS) for S-100 and Melan-A (MART1). This panel was chosen after pathologic evaluation of 99 positive SLNs from 72 patients treated at our institution.152 The sensitivities for S-100, Melan-A, and HMB-45 were found to be 97%, 96%, and 75%, respectively. In addition, we found that HMB-45 stained a smaller percentage of cells (25% to 75%), with weaker intensity compared with S-100 and Melan-A. For this reason, we no longer routinely stain for HMB-45. Patients with a positive SLN return to the operating room within 2 weeks of diagnosis for denitive TLND. Patients with a negative biopsy are followed clinically. An alternative to this 2-staged technique is immediate TLND based on frozen section evaluation of the SLNs. However, it is important to realize that the reliability of frozen sections for melanoma analysis has been questioned,151,153 and permanent sections remain the gold standard. The pathologist plays an extremely critical role in the success of the SLNB. Occult lymphatic metastasis from cutaneous melanoma can be difcult to detect, with tumor cells occupying less that 2% of the entire lymph node volume.151 Therefore, rigorous pathological analysis including serial sectioning, special immunohistochemical study when indicated, and interpretation by an experienced pathologist is necessary. Wagner and colleagues reported the mean tumor volume in positive SLNs to be only 4.7 mm3.154 Joseph and colleagues reported identication of only 73% of metastatic SLNs using standard H&E staining alone.155 In our study, 20 of the 97 positive SLNs (21%) were negative on initial H&E staining.152 This high false negative rate highlights the importance of IHCS for accurate diagnosis of occult nodal disease. From a practical standpoint, the histologic analysis of SLNs is more thorough, cost-effective, and complete compared with traditional evaluation of the entire lymphadenectomy specimen because the technique provides the pathologist with a limited number of nodes to evaluate thoroughly.156 In an effort to further increase SLNB sensitivity, Morton and colleagues are investigating the utility of carbon dye as a mapping adjunct. Unlike isosulfan blue dye, the carbon remains as a permanent marker to aid the pathologist in identifying the specic intranodal site of lymphatic drainage, which is the most likely area for occult metastatic disease.157 We have found SLNB particularly helpful in the diagnosis of occult DM/DNM. Here, a focused histopathologic evaluation is particularly important because the microscopic features of metastatic DM/DMN are quite variable, often lack resemblance of the primary tumor, are limited to
Curr Probl Surg, November 2006 813

a paucity of tumor cells, and demonstrated inconsistencies on HMB-45 and Melan-A staining.158 SLNB is a team effort involving experienced surgeons, nuclear medicine staff, and pathologists. The experience and technical skill of the surgeon is vital and may account for some of the variability observed in HN cutaneous melanoma SLN studies.159 Morton and colleagues previously suggested a 30 case learning curve.150 However, long-term follow-up of their international Multicenter Selective Lymphadenectomy Trial (MSLT-I) found the 30 case learning curve to be too shallow. Analysis of the rst 25 cases performed at the 10 highest volume centers in the trial revealed a nodal basin recurrence rate of 10.3%.151 This false negative rate dropped to 5.2% after 25 additional cases. The authors now conclude that a 55 case learning curve is required to achieve at least 95% accuracy with SLNB. An experienced nuclear medicine staff is necessary because inappropriate administration of the radioactive tracer can lead to shine through, which renders the handheld gamma probe useless in the operating room. Communication with the nuclear medicine team is critical not only in interpreting the lymphoscintigram, but also in ensuring that the appropriate lesion is mapped since patients with melanoma often present with multiple pigmented lesions and signicant solar changes. Recent multivariate analysis involving patients with stage I and II melanoma by Greshenwald and colleagues found the pathologic status (positive or negative for metastasis) of the SLN to be the most important prognostic factor for both recurrence and overall survival.160 For stage III melanoma, a survival benet was found in patients with occult microscopic disease compared with their counterparts who had palpable, macroscopic disease (Table 9).101 This survival benet was so compelling that the AJCC has now incorporated SLNB into the revised staging system for cutaneous melanoma. Although SLNB has a dened role in the evaluation of cutaneous melanoma of the trunk and extremities, several questions have been posed with respect to its application in the HN region.150,161,162 The complexity of the HN lymphatic system has caused concern surrounding the reliability of the SLN to represent the status of the entire nodal basin accurately. The interlacing network of cervical lymphatic vessels is often deemed watershed in nature. The complexity of this lymphatic system was demonstrated by OBrien and colleagues who reported 34% discordance between the clinical prediction of lymphatic drainage and lymphoscintigraphy ndings in 97 cases of HN cutaneous melanoma.163 The popularity of SLNB in the HN region has also been limited by concerns surrounding damage to vital structures such as the facial nerve,164
814 Curr Probl Surg, November 2006

technical difculties,161,164 and the necessity for nuclear medicine staff as well as pathologists who specialize in SLNB technique. Our experience in 80 patients with HN cutaneous melanoma demonstrated that the complexity of HN anatomy does not preclude the use of SLNB for staging of cutaneous melanoma.159 SLNB accurately predicted the status of the nodal basin in this region. Fourteen (17.5%) of 80 patients were identied with a positive SLNB. Only 3 (4.5%) of 66 patients developed regional recurrence following a negative SLNB. The 17.5% positivity rate of SLNs and the 4.5% false negative rate both mirror the results of SLNB achieved in other anatomic sites such as the trunk and extremities.156,165 Similar success in the application of SLNB for HN cutaneous melanoma has been reported by others,166,167 and the technique has successfully been applied in pediatric HN cases.11 Approximately 25% to 30% of HN cutaneous melanomas drain to lymph nodes within the parotid bed.159,163 Potential injury to the facial nerve from SLNB has led some surgeons to advocate supercial parotidectomy over the mapping procedure.164 In our retrospective analysis, 28 (93.3%) of 30 patients draining to the parotid nodal basin successfully underwent staging using SLNB.159 One patient required a supercial parotidectomy due to the location of the SLN deep to the facial nerve. A second patient experienced signicant bleeding from surrounding parotid tissue, which could have placed the facial nerve at increased risk. A total of 39 nodes from 28 parotid basins were removed without facial nerve injury. Continuous facial nerve monitoring for SLNB within the parotid nodal basin can be helpful when performing the biopsy with the parotid bed. Concern has also been expressed that SLNB causes inammation and brosis that could place the facial nerve at increased risk when reoperation is required to treat the parotid basin denitively in the setting of a positive SLN.164 In our experience, all patients with a positive parotid SLN underwent a supercial parotidectomy as a subsequent procedure, without facial nerve injury. Our ndings are consistent with other reports demonstrating that SLNB can be performed reliably and safely within the parotid nodal basin.132,168 Other authors have suggested that SLNB increases the risk of in-transit metastasis (ITM), which is dened as intralymphatic tumor dissemination within cutaneous or subcutaneous tissue located between the primary lesion and draining nodal basin.169-174 It is theorized that ITM develops when melanoma cells detach from the primary lesion and become lodged in the dermal plexus of lymphatics before reaching the lymph nodes. The development of ITM presents a therapeutic challenge and carries a poor
Curr Probl Surg, November 2006 815

prognosis as indicated by the new changes to the AJCC staging system cited above.101 Original reports citing increased ITM following SLNB must be viewed with caution because the studies often entailed pooled data from small cohorts and failed to control for important prognostic factors such as tumor thickness and ulceration.169,175 A more recent prospective review comparing 4412 patients undergoing WLE alone, WLE with SLNB, and ELND identied a correlation between ITM and increasing Breslow depth, Clark level, and T stage.175 A statistically signicant difference between ITM and tumor recurrence was not found among the treatment groups, once adjustment was made for T stage, age, sex, tumor thickness, and site. Additional studies have concluded that it is the tumor biology, as opposed to the surgical procedure (SLNB; ELND), which dictates melanoma metastatic behavior.176,177 Finally, a correlation between ITM and SLNB was not reported with MSLT-I, thus negating this concern.151 The impact that SLNB imparts on overall survival remains to be determined. The answer will hopefully be provided through the multiinstitutional Sunbelt Melanoma Trial, which is a prospective, randomized clinical trial that uses SLN staging to determine the need for adjuvant therapy.178 While we await these results, McMasters and colleagues outlined 4 compelling reasons to use SLNB for accurate regional staging of cutaneous melanoma.148 First, the SLNB technique provides important prognostic information to the physician, patient, and family members in guiding subsequent treatment options. Second, SLNB helps identify patients harboring nodal metastasis, who then may benet from early TLND. Third, SLNB identies patients who are candidates for adjuvant treatment such as interferon- 2b. Fourth, SLNB provides the most accurate means of regional staging. In doing so, the technique enables the identication of a homogeneous population of patients for enrollment into clinical trials. Regional metastasis is recognized as the most important prognostic factor in melanoma. Without accurate pathologic staging, stratication is impossible, and the results of clinical trials will remain inconsistent and difcult to interpret. The fth and nal analysis of MSLT-I will provide additional insight into the potential therapeutic benet of SLNB.151 Recent publication of the MSLT-I interim analysis is exciting because it is the rst randomized, prospective trial to demonstrate that SLNB accurately identies occult nodal metastasis, which will lead to advanced, palpable nodal disease if left in situ. The authors argue that there is no reason not to perform SLNB because a zero mortality rate was reported, and the complication rate of SLNB (10%) was signicantly lower than for TLND (37%). Morton
816 Curr Probl Surg, November 2006

concluded that SLNB should be considered standard of care for regional staging of primary cutaneous melanomathe key term being staging. At the present time SLNB is a diagnostic tool for regional staging, not a therapeutic modality. Although it is not 100% accurate, SLNB is the most reliable means for regional staging. It is more sensitive and specic than CT, MRI, PET, ELND, and clinical examination.179 McMasters astutely points out that we do not impart a survival benet for any other cancer staging test, and therefore we should not ask the same of SLNB.

Future SLNB Investigations


SLNB research endeavors hold exciting and great promise in the future. In an effort to further investigate the therapeutic potential of SLNB, recent research efforts have focused on identifying markers of both the primary lesion and the SLN that are predictive of tumor containing non-SLNs.180-182 Absence of these markers would then allow identication of the subset of SLNB positive patients who may not warrant further treatment with a formal TLND. Unfortunately, current studies have failed to identify a consistent and 100% accurate marker. For this reason, the future MSLT-II trial is designed to investigate the indications for TLND following a positive SLNB. Specically, it will determine whether immediate TLND provides a survival benet over postoperative, diligent, ultrasonographic monitoring of the draining nodal basins.151 Molecular staging of melanoma is also gaining increased interest. Reverse transcriptase polymerase chain reaction (RT-PCR) analysis of SLNs for melanoma-associated genes such as MART-1, tyrosinase, microphthalmia-associated transcription factor (MIFT), and tyrosinaserelated protein 2 (TRP-2) has proven helpful in identifying a subset of patients harboring occult nodal disease at a submicroscopic level that cannot be detected with traditional IHCS.183-185 In 1 study, 30% (49 of 162) of patients had negative SLNs on IHCS but at least 1 melanoma marker identied with RT-PCR.186 This subset of patients experienced an increased rate of tumor recurrence. Currently, RT-PCR lacks specicity. The high false positive rate may be from the inability to differentiate melanoma cells from occult benign nevus cells.187 Through future research efforts, molecular staging may prove helpful in the identication of a subset of high-risk melanoma patients who develop nodal or distant metastases despite presentation with a thin primary tumor. This information has the potential to change the manner in which these individuals are counseled both with respect to adjuvant therapy and follow-up surveillance.188
Curr Probl Surg, November 2006 817

Surgical Management of Distant Metastasis


Patients with stage IV melanoma involving distant sites have an exceedingly grave prognosis. Surgical treatment has a limited role. Operation has been used as a means of palliative treatment in patients suffering from brain, lung, gastrointestinal, subcutaneous soft tissue, and distant lymph node metastasis.189 The success of surgery in the palliative setting is heavily dependent on appropriate patient selection. Surgery should be considered only if clearly identiable and specic symptoms are associated with a metastatic lesion. In selecting patients, it is also important to consider surgical morbidity, expected quality of life, expected survival, and most importantly, the patients wishes.190 The patient and family must understand that the goal of surgery is palliative in nature. Several prognostic markers have been identied in patients with disseminated stage IV melanoma.190,191 These markers are reected in the AJCC staging system described above and should serve as a guide when considering surgical resection of distant tumors. Patients with metastatic disease limited to 1 or 2 isolated sites experience a better prognosis compared with patients with multiple metastatic lesions. In addition, a short disease-free interval between initial diagnosis and the development of distant metastasis correlates with a poor overall prognosis, even when complete resection of the metastatic lesion is achieved.190 Last, the anatomic site of the metastatic lesion is of importance. Patients with metastatic spread to nonvisceral sites such as distant subcutaneous tissues or lymph nodes have a better prognosis compared with individuals with visceral metastasis. Within the group of patients suffering from visceral metastasis, individuals with pulmonary lesions experience improved survival compared with other visceral sites.

Radiation Therapy
Melanoma has traditionally been classied as a radioresistant tumor.192,193 Although adjuvant radiation has not been shown to impact survival,194 researchers at the M.D. Anderson Cancer Center completed a phase II clinical trial supporting the efcacy of large dose, hypofractionated radiation as an adjuvant treatment to surgery for HN cutaneous melanoma patients at high risk for local-regional recurrence.195-197 Local-regional control was achieved in 88% of patients, an improvement over the historical control rates of 50% to 70%.8,198 Late radiation complications were rare (3 of 174 patients) and included moderate neck brosis, mild ipsilateral hearing loss, and transient exposure of external auditory canal cartilage. Although an overall survival benet has not been
818 Curr Probl Surg, November 2006

demonstrated, local-regional control is of great importance because recurrence can signicantly impact the quality of life by causing pain, wound breakdown, and socially debilitating cosmetic disgurement, especially in the HN region.199,200 Ultimately, the survival benet achieved with adjuvant radiation therapy will be determined through the prospective, randomized phase III clinical trial (E3697) currently under investigation by the Radiation Therapy Oncology Group.199 Until this randomized phase III clinical trial is completed, most authors advocate the use of adjuvant radiation for patients demonstrating adverse prognostic markers such as neurotropism, extracapsular spread (ECS), multiple node involvement ( 4), or tumor recurrence.201-203 These patients are often eligible to receive adjuvant interferon- 2b (see discussion herein). Interferon is thought to act as a radiosensitizer; therefore, it is common practice to delay radiation until the 4-week induction phase of interferon therapy is complete.204 On rare occasion, primary radiation can be used to treat extensive LM/LMM in an elderly patient who is not deemed a surgical candidate or if the lesion is so extensive that surgical resection would leave the patient functionally and socially crippled.205 Radiation therapy can also be administered as palliative treatment. Patients suffering from painful, systemic stage IV disease, such as brain metastasis, bone metastasis, spinal cord compression, and isolated, symptomatic visceral metastasis have gained benet from such treatment.201

Chemotherapy
Melanoma is a relatively chemoresistant tumor.206-212 A small subset of patients is thought to benet from chemotherapy; however, a regimen that denitively impacts survival has not emerged. The challenge lies in the limited number of stage IV patients, the inherently short survival period for this group, and the vast number of treatment options being studied in phase I and II trials. The main role for chemotherapy remains as palliative treatment in the setting of disseminated stage IV disease. Dacarbazine (DTIC) was the rst chemotherapeutic agent to show signicant activity against melanoma. Today it remains the only agent approved for treatment of advance stage IV melanoma. Unfortunately, response rates following DTIC administration are modest at best, ranging from 10% to 20%.26,76,102,109,130 This prognosis has not changed over the past 2 decades, despite dedicated research efforts using a host of chemotherapeutic agent regimens. Overall, fewer than 5% of individuals experience a complete response with DTIC.
Curr Probl Surg, November 2006 819

Immunotherapy
Classically, immunotherapy is divided into 2 categories. Specic immunotherapeutic agents upregulate the antibody and cytotoxic T-cell immune response specically to the patients tumor or to known melanoma antigens. The majority of melanoma vaccines fall into this category. In contrast, nonspecic immunotherapeutic agents stimulate the hosts immune system without targeting melanoma tumor antigens. Examples include interferon, interleukin, and microbacterial products such as Bacille Calmette-Guin (BCG) and Cryptosporidium parvum. These agents are often administered as an adjuvant to specic immunotherapeutic agents in an attempt to augment the immune system.204,208

Interferon
Despite myriad clinical trials involving adjuvant regimens, high dose interferon- 2b (IFN- 2b) remains the only U.S. Food and Drug Administration (FDA) approved adjuvant treatment for stage III melanoma. It functions as a biologic response modier. Mechanisms of action include direct antiproliferative effects, immune stimulation through enhancement of natural killer cells (NKCs), increased histocompatibility antigen expression on melanoma cells, macrophage phagocytosis, and enhanced T-cell mediated cytotoxicity.128,212 Although all 3 types of interferon (IFN- , IFN- , IFN- ) demonstrate antitumor activity,212 IFN- 2b is the only treatment currently approved for adjuvant treatment of melanoma patients at high risk of recurrence following surgery. Three large clinical trials involving adjuvant IFN- 2b have been conducted by the Eastern Cooperative Oncology Group (ECOG).213-215 In brief, ECOG trial E1684 was the rst study to demonstrate the efcacy of IFN- 2b.213 The regimen consisted of high dose interferon (20 million units[MU]/m2/d) administered intravenously, 5 days per week for 4 weeks. Maintenance treatment followed, consisting of 48 weeks of subcutaneous IFN- 2b (10 MU/m2/d), administered 3 days per week. The prolonged disease-free survival rate and overall survival rate in the IFN- 2b arm of E1684 ultimately led to FDA approval of adjuvant high dose IFN- 2b. Although the follow-up trial E1690 failed to conrm the efcacy of high dose IFN- 2b,214 the results require careful interpretation.148 Unlike E1684, patients enrolled in E1690 did not require pathologic staging with ELND or SLNB, nor were they stratied on ulceration. In addition, a disproportionate number of individuals from the observation arm crossed over into the IFN- 2b arm to receive salvage therapy for recurrent
820 Curr Probl Surg, November 2006

disease. Any therapeutic benet provided to this subgroup by IFN- 2b went unrecognized since the crossover patients remained in the observation arm for the purposes of statistical analysis. The most recent and largest of the 3 studies, ECOG 1694,215 once again conrmed the efcacy of high dose IFN- 2b. In fact, the relapse-free and overall survival benet observed in the high dose IFN- 2b control arm compared with the GMK ganglioside vaccine treatment arm was so compelling that the data safety monitoring committee terminated the trial early. With close follow-up, dose modication, and pharmacologic intervention, the majority of melanoma patients are able to tolerate the 1-year course of IFN- 2b.148,204,214,215 Almost every individual experiences u-like symptoms (fevers, chills, malaise) during the initial treatment course. Severe and intolerable chronic fatigue is experienced in 20% to 30% of patients. An additional 2% to 10% of patients experience neurologic and psychiatric side effects including depression, anxiety, suicidal ideation, and difculty with cognition. Myelosuppression, thyroid dysfunction, and elevated liver enzymes require close monitoring. Contraindications include a history of myocardial infarction or dysrhythmia, liver disease, CNS disorder, and severe psychiatric illness.204 Approximately 50% of patients require a dose reduction or delay as a result of these side effects. Despite these risk factors, the majority of melanoma patients are willing to accept the side effects given the potential benet of IFN- 2b.216 While clinical trials continue to investigate alternative dosages and schedules,217,218 only high dose IFN- 2b is FDA approved and used as routine adjuvant therapy within the United States. Given the toxicities associated with IFN- 2b, the adjuvant therapy is reserved for patients at high risk for tumor recurrence. Candidates include patients with regional lymph node metastasis or a primary tumor thickness greater than 4 mm. All patients should be informed of the option to receive postoperative IFN- 2b. To make an educated decision, an objective discussion including the side effects associated with IFN- 2b, as well as the opportunity to enroll in other clinical trials, must be provided.

Interleukin-2 and Other Cytokines


Interleukin-2 (IL-2) is another form of immunotherapy used in the primary treatment of patients with disseminated stage IV disease. Unlike interferon, IL-2 lacks direct antitumor activity and in vitro activity.128 However, in vivo, IL-2 stimulates the host immune system by activating effector cells such as NKCs, monocytes, cytotoxic T cells, and helper T
Curr Probl Surg, November 2006 821

cells. It also induces cytokines such as IFN- and tumor necrosis factor (TNF)- .128,219 Rosenberg and the National Cancer Institute Surgery Branch successfully used high dose IL-2 to treat 134 melanoma patients.220 An overall response was observed in 17% of patients, with 10% experiencing a partial response, and an additional 7% experiencing complete regression. The therapeutic benet was substantial, lasting between 2 and 8 years. IL-2 side effects are signicant and potentially lethal. Acute toxicities include myocardial infarction, arrhythmia, respiratory distress, hypotension, capillary leak syndrome, nephrotoxicity, hepatic toxicity, and sepsis.219 Other toxicities include anemia, thrombocytopenia, nausea, emesis, diarrhea, myalgia/arthralgia, skin erythema, and pruritus. Only patients who demonstrate excellent cardiopulmonary health and performance status should be considered for clinical trials involving IL-2. Subsequent efforts to enhance the response to IL-2 by altering the schedule, the dose, and by combining other therapeutic agents such as lymphokine-activated killer (LAK) cells have not proven benecial.128,208,219 In addition to IL-2, other cytokines have been studied alone and in combination with various chemotherapeutic agents. The list includes IL-1, IL-4, IL-6, and TNF- .219 To date, substantial therapeutic benet has not been demonstrated.

Follow-up and Surveillance


The primary goals in melanoma follow-up are: 1) early detection of local-regional tumor recurrence, 2) early identication of second primary tumors (including melanoma as well as other skin cancers), 3) continuing patient education, and 4) psychological support.193 Each follow-up visit should include an inquiry into new or changing skin lesions. A review of systems (Table 3) concerning distant metastasis should be asked.98 A thorough examination of the skin and mucosa is required, with particular attention paid to the original melanoma site and associated draining nodal basins. Photodocumentation and dermoscopy have proven helpful in monitoring change in patients who have a substantial number of nevi.221 This vigilant monitoring is particularly important among elderly patients where a change in nevus appearance is more likely to be a melanoma compared with younger patients.221 Each follow-up visit should be viewed as an opportunity to re-educate patients on the ABCDE melanoma warning signs and the importance of monthly skin self-examination. Studies have demonstrated that melanoma patients who practice monthly self-examination are diagnosed with thinner lesions at the time of recurrence.221 Sun education including the use of sunscreen, avoidance of
822 Curr Probl Surg, November 2006

peak sun hours, shade seeking, sun protective clothing, and the potential risks of tanning booths should all be emphasized.

Conclusion
Over the past few decades, melanoma has escalated into a problem of epidemic proportions. Fortunately, signicant research advances have been achieved in recent years.204,208,212,222,223 Intense efforts in the areas of melanoma vaccination, gene therapy, HLA immunoprinting, and gene proling at the genomic, as well as the proteomic level, will likely play an important role in future research endeavors. Given the association with sun exposure, melanoma is considered a preventable disease. Decreased incidence and mortality hinges on patient as well as physician education, prevention, early diagnosis, and improved treatment for advanced disease. Ultimately, the key to impacting melanoma survival rates lies in wellorganized, multi-institutional studies that enroll patients who are staged accurately and therefore share a similar prognosis.

REFERENCES
1. Rigel DS. The effect of sunscreen on melanoma risk. Dermatol Clin 2002;20:601-6. 2. Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, et al. Cancer statistics, 2005. CA Cancer J Clin 2005;55:10-30. 3. Surveillance, Epidemiology and End Results. 2005. http://seer.cancer.gov. Accessed December 13, 2005. 4. Beddingeld FC. The melanoma epidemic: Res Ipsa Loquitur. The Oncologist 2003;8:459-65. 5. Rigel DS, Carucci JA. Malignant melanoma: prevention, early detection, and treatment in the 21st century. CA Cancer J Clin 2000;50:215-36. 6. Fisher SR, OBrien CJ Head and neck melanoma. In: Balch CM, Houghton AN, Sober AJ, Soong SJ, editors. Cutaneous Melanoma. St. Louis, MO: Quality Medical Publishing; 2003. 7. Fisher SR. Elective, therapeutic, and delayed lymph node dissection for malignant melanoma of the head and neck: analysis of 1444 patients from 1970 to 1998. Laryngoscope 2002;112:99-110. 8. OBrien CJ, Coates AS, Petersen-Schaefer K, Shannon K, Thompson JF, Milton GW, et al. Experience with 998 cutaneous melanomas of the head and neck over 30 years. Am J Surg 1991;162:310-4. 9. Peralta EA, Yarington T, Glenn MG. Malignant melanoma of the head and neck: effect of treatment on survival. Laryngoscope 1998;108:220-3. 10. Fisher SR, Reintgen DS, Seigler HF. Juvenile malignant melanoma of the head and neck. Laryngoscope 1988;98:184-9. 11. Pacella SJ, Lowe L, Bradford C, Marcus BC, Johnson T, Rees R. The utility of sentinel lymph node biopsy in head and neck melanoma in the pediatric population. Plast Reconstr Surg 2003;112:1257-65. 12. Kobayashi N, Muramatsu T, Yamashina Y, Shirai T, Ohnishi T, Mori T. Melanin
Curr Probl Surg, November 2006 823

13. 14. 15. 16.

17.

18.

19.

20. 21.

22. 23.

24.

25.

26. 27. 28.

29. 30.
824

reduces ultraviolet-induced DNA damage and killing rate in cultured human melanoma cells. J Invest Dermatol 1993;101:685-9. Pathak MA, Stratton K. Free radicals in human skin before and after exposure to light. Arch Biochem Biophys 1968;123:468-76. Riley PA. Melanin. Int J Biochem Cell Biol 1997;29:1235-9. Gilchrest BA, Eller MS, Gellar AC, Yaar M. The pathogenesis of melanoma induced by ultraviolet radiation. N Engl J Med 1999;340:1341-8. Holman CD, Armstrong BK, Heenan PJ. Etiology of common acquired melanocytic nevi: constitutional variables, sun exposure, and diet. J Natl Cancer Inst 1986;77:329-35. Nelemans PJ, Groenendal H, Kiemeney LA, Rampen FH, Ruiter DJ, Verbeek AL. Effect of intermittent exposure to sunlight on melanoma risk among indoor workers and sun-sensitive individuals. Environ Health Perspect 1993;101:252-5. Bentham G, Aase A. Incidence of malignant melanoma of the skin in Norway, 1955-1989: associations with solar ultraviolet radiation, income and holidays abroad. Int J Epidemiol 1996;25:1132-8. Bayerl C, Taake S, Moll I, Jung EG. Characterization of sunburn cells after exposure to ultraviolet light. Photodermatol Photoimmunol Photomed 1995; 11:149-54. Gilcrest BA, Treloar V, Grass AM, Yaar M, Szabo G, Flynn E. J Invest Dermatol 1986;87:102-7. Fitzpatrick TB, Ortonne JP: Normal skin color and general considerations of pigmentary disorders. In: Freedberg IM, Eisen AZ, Wolff K, Austen KF, Goldsmith LA, Katz AI, editors. Fitzpatricks Dermatology in General Medicine. New York: McGraw Hill Medical Publishing; 2003. Yaar M, Gilcrest BA. Ageing and photoageing of keratinocytes and melanocytes. Clin Exp Dermatol 2001;26:583-91. Tsao H, Sober AJ Atypical melanocytic nevi. In: Freedberg IM, Eisen AZ, Wolff K, Austen KF, Goldsmith LA, Katz AI, editors. Fitzpatricks Dermatology in General Medicine. New York: McGraw Hill Medical Publishing; 2003. Berwick M, Weinstock MA. Epidemiology: current trends. In: Balch CM, Houghton AN, Sober AJ, Soong SJ, editors. Cutaneous Melanoma. St. Louis, MO: Quality Medical Publishing; 2003. Kennedy C, Bajdik CD, Willemze R, et al. The inuence of painful sunburns and lifetime sun exposure on the risk of actinic keratoses, seborrheic warts, melanocytic nevi, atypical nevi, and skin cancer. J Invest Dermatol 2003;1087-93. Rigel DS. Identication of those at highest risk for development of malignant melanoma. Adv Dermatol 1995;10:151-70. Johnson TM, Hamilton T, Lowe L. Multiple primary melanomas. J Am Acad Dermatol 1998;39:422-7. Schmalbach CE, Johnson TM, Bradford CR. The management of head and neck melanoma. In: Cummings CW, Flint PW, Harker LA, Haughey BH, Richardson MA, Robbins KT, et al, editors. Cummings Otolaryngology Head & Neck Surgery. Philadelphia: Elsevier Mosby; 2005. Kraehn GM, Schartl M, Peter RU. Human malignant melanoma: a genetic disease? Cancer 1995;75:1228-37. Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K, Tavtigian SV, et al.
Curr Probl Surg, November 2006

31.

32. 33.

34. 35.

36.

37.

38.

39.

40. 41. 42. 43.

44. 45.

46.

47. 48.

A cell cycle regulator potentially involved in genesis of many tumor types. Science 1994;264:436-40. Aitken J, Welch J, Duffy D, Milligan A, Green A, Martin N, et al. CDKN2A variants in a population-based sample of Queensland families with melanoma. J Natl Cancer Inst 1999;91:446-52. Piepkorn K. Melanoma genetics: and update with focus on the CDKN2A(p16)/ARF tumor suppressors. J Am Acad Dermatol 2000;42:705-22. Clark WH Jr., Reimer RR, Greene MH, Ainsworth AM, Mastrangelo MJ. Origin of familial malignant melanoma from heritable melanocytic lesions: the B-K mole syndrome. Arch Dermatol 1978;114:732-8. Lynch HT, Frichot BC, Lynch JF. Familial atypical multiple mole-melanoma syndrome. J Med Genet 1978;15:352-6. Greene MH, Goldin LR, Clark WH Jr, Lovien E, Kraemer KH, Tucker MA, et al. Familial cutaneous malignant melanoma: autosomal dominant trait possibly lined to the Rh locus. Proc Natl Acad Sci USA 1983;80:6071-5. Marghoob AA, Kopf AW, Rigel DS, Bart RS, Friedman RS, Yadav S, et al. Risk of cutaneous malignant melanoma in patients with classic atypical mole syndrome. A case-control study. Arch Dermatol 1994;130:993-8. Marghoob AA, Schoenbach SP, Kopf AW, Orlow SJ, Nossa R, Bart RS. Large congenital melanocytic nevi and the risk of developing malignant melanoma: a prospective study and review of the world literature. Arch Dermatol 1997;132:170-5. Greene MH, Clark WH Jr, Tucker MA, Mraiemer KH, Elder DE, Fraser MC. High risk of malignant melanoma in melanoma-prone families with dysplastic nevi. Ann Int Med 1985;102:458-65. Kraemer KH, Levy DD, Parris CN, Gozukara EM, Moriwaki S, Adelberg S, et al. Xeroderma pigmentosum and related disorders: examining the linkage between defective DNA repair and cancer. J Invest Dermatol 1994;103:96S-101S. Cleaver JE. Defective repair replication of DNA in xeroderma pigmentosum. Nature 1968;218:652-6. Ceballos PI, Ruiz-Maldonado R, Milun MC Jr. Melanoma in children. N Engl J Med 1995;332:656-62. Greene MH, Young TI, Clark WH Jr. Malignant melanoma in renal-transplant recipients. Lancet 1981;1:1196-9. Reynolds P, Suanders LD, Layefsky ME, Lemp GF. The spectrum of acquired immunodeciency syndrome (AIDS)-associated malignancies in San Francisco, 1980-1987. Am J Epidemiol 1993;137:19-30. Aboulaa DM. Malignant melanoma in an HIV-infected man: a case report and literature review. Cancer Invest 1998;16:217-24. McGregor JM, Newell M, Ross J, Krkam N, McGibbon DH, Barley C. Cutaneous malignant melanoma and human immunodeciency virus (HIV) infection: a report of three cases. Br J Dermatol 1992;126:516-9. Smith CH, McGregor JM, Barker JN, Morris RW, Rigdon SP, MacDonald DM. Excess melanocytic nevi in children with renal allografts. J Am Acad Dermatol 1993;28:51-5. Baird EA, McHenry PM, Mackie RM. Effect of maintenance chemotherapy in childhood on numbers of melanocytic naevi. BMJ 1992;305:799-801. Naldi L, Adamoli L, Fraschini D, Corbelta A, Imberti L, Reseghett A, et al. Number
825

Curr Probl Surg, November 2006

49.

50. 51. 52.

53.

54. 55. 56. 57. 58.

59.

60.

61. 62. 63.

64. 65. 66.

67.
826

and distribution of melanocytic nevi in individuals with a history of childhood leukemia. Cancer 1996;77:1402-8. Rhodes AR, Albert LS, Weinstock MA. Congenital nevomelanocytic nevi: proportionate area expansion during infancy and early childhood. J Am Acad Dermatol 1996;34:51-62. Marghoob AA. Congenital melanocytic nevi: evaluation and management. Dermatol Clin 2002;20:607-16. Consensus Development Panel. Precursors to malignant melanoma. JAMA 1984;251:1864-94. Bittencourt FV, Marghoob AA, Kopf AW, Koenig KL, Bart RS. Large congenital melanocytic nevi and the risk of developing malignant melanoma and neurocutaneous melanocytosis. Pediatrics 2000;106:736-41. Anderson KW, Baker SR, Lowe L, Su L, Johnson TM. Treatment of head and neck melanoma, lentigo maligna subtype: a practical surgical technique. Arch Facial Plast Surg 2001;3:202-6. Cohen LM. Lentigo maligna and lentigo maligna melanoma. J Dermatol 1995;33:913. Conley J, Lattes R, Orr W. Desmoplastic malignant melanoma (a rare variant of spindle cell melanoma). Cancer 1971;28:914-36. Reed JG, Leonard DD. Neurotropic melanoma: a variant of desmoplastic melanoma. Am J Surg Pathol 1979;3:310-1. Carlson JA, Dickersin GR, Sober AJ, Barnhill RL. Desmoplastic neurotropic melanoma, a clinicopathologic analysis of 28 cases. Cancer 1995;75:478-94. Quinn MJ, Crotty KA, Thompson JG, Coates AS, OBrien CJ, McCarthy WH. Desmoplastic and desmoplastic neurotropic melanoma, experience with 280 patients. Cancer 1998;83:1128-35. Fullen DR, Lowe L, Wang TS, Schwartz JL, Cimmino VM, Sondak VK, Johnson TM. Desmoplastic and neurotropic melanoma: analysis of 33 patients with lymphatic mapping and sentinel lymph node biopsy. Cancer 2003;100:598-604. Chang AE, Karnell LH, Menck HR. The national cancer database report on cutaneous and noncutaneous melanoma: a summary of 84,836 cases from the past decade. Cancer 1998;83:1664-78. Chang P, Knapper WH. Metastatic melanoma of unknown primary. Cancer 1982;49:1106-11. Norman J, Cruse CW, Wells KE, Saba HI, Reintgen DS. Metastatic melanoma with an unknown primary. Ann Plast Surg 1992;28:81-4. Nasri S, Namazie A, Dulguerov P, Mickel R. Malignant melanoma of cervical and parotid lymph nodes with an unknown primary site. Laryngoscope 1994;104: 1194-8. Santini H, Byers RM, Wolf PF. Melanoma metastatic to cervical and parotid nodes from an unknown primary site. Am J Surg 1985;150:510-2. Jonk A, Kroon BBR, Rmke P, Maooi WJ, Hart AA, van Dongen JA. Lymph node metastasis from melanoma with an unknown primary site. Br J Surg 1990;77:665-8. Bulkey GB, Cohen MH, Banks PM, Char DH, Ketcham AS. Long-term spontaneous regression of malignant melanoma with visceral metastasis, report of a case with immunologic prole. Cancer 1975;36:485-94. Ross MI, Stern SJ Mucosal melanomas. In: Balch CM, Houghton AN, Sober AJ,
Curr Probl Surg, November 2006

68. 69. 70. 71. 72. 73. 74. 75.

76.

77.

78. 79. 80. 81. 82.

83.

84. 85. 86.

87.

Soong SJ, editors. Cutaneous Melanoma. St. Louis, MO: Quality Medical Publishing; 2003. Berthelsen A, Andersen AP, Jensen TS, Hansen HS. Melanomas of the mucosa in the oral cavity and the upper respiratory passages. Cancer 1984;54:907-12. Hoyt DJ, Jordan T, Fisher SR. Mucosal melanoma of the head and neck. Arch Otolaryngol Head Neck Surg 1989;115:1096-9. Manolidis S, Donald PJ. Malignant mucosal melanoma of the head and neck: review of the literature and report of 14 patients. Cancer 1997;80:1373-86. Patel SG, Prasad ML, Escrig M, Singh B, Shaha AR, Kraus DH, et al. Primary mucosal malignant melanoma of the head and neck. Head Neck 2002;24:247-57. Kato T, Takematsu H, Tomita Y, Takahash M, Abe R. Malignant melanoma of mucous membranes. Arch Dermatol 1987;123:216-20. Stern SJ, Guillamondegui OM. Mucosal melanoma of the head and neck. Head Neck 1991;13:22-7. Batsakis JG, Regezi JA, Solomon AR, Rice DH. The pathology of head and neck tumors: mucosal melanoma, part 13. Head Neck Surg 1982;4:404-18. Lee SP, Shimizu KT, Tran LM, Juillard G, Calcaterra TC. Mucosal melanoma of the head and neck: the impact of local control on survival. Laryngoscope 1994;104:121-6. Koh HK, Miller DR, Geller AC, Clapp RW, Mercer MB, Lew RA. Who discovers melanoma? Patterns from a population-based survey. J Am Acad Dermatol 1992;26:914-9. Schwartz JL, Wang TS, Hamilton TA, Clapp RW, Mercer MB, Lew RA. Thin primary cutaneous melanomas: associated detection patterns, lesion characteristics, and patient characteristics. Cancer 2002;95:1562-8. Beardmore GL, Cavis NC. Multiple primary cutaneous melanomas. Arch Dermatol 1975;111:603-9. Kang S, Barnhill RL, Mihm MC Jr, Sober AJ. Multiple primary cutaneous melanomas. Cancer 1992;70:1911-6. Johnson TM, Hamilton T, Lowe L. Multiple primary melanomas. J Am Acad Dermatol 1998;39:422-7. Moseley HS, Giuliano AE, Storm FK, Clark WH, Robinson DS, Morton DL. Multiple primary melanoma. Cancer 1979;43:939-44. American Academy of Dermatology Melanoma/skin cancer: you can recognize the signs. AAD patient handout. Evanston, IL: American Academy of Dermatology; 1986. McGovern TW, Litaker MS. Clinical predictors of malignant pigmented lesions, a comparison of the Glasgow seven-point checklist and the American Cancer Societys ABCDs of pigmented lesions. J Dermatol Surg Oncol 1992;18:22-6. MacKie RM. Clinical recognition of early invasive melanoma. BMJ 1990;301:1005-6. Rigel DS, Friedman RJ, Kopf AW, Polsky D. ABCDEan evolving concept in the early detection of melanoma. Arch Dermatol 2005;141:1032-4. Abbasi NR, Shaw HM, Rigel DS, Friedman JL, McCarthy WH, Osman I, et al. Early diagnosis of cutaneous melanoma: revisiting the ABCD criteria. JAMA 2004;292:2771-6. Grob JJ, Bonerandi JJ. The ugly ducklingsign: identication of the common
827

Curr Probl Surg, November 2006

88.

89.

90.

91. 92. 93.

94.

95.

96. 97.

98.

99.

100.

101.

102. 103.

104.
828

characteristics of nevi in an individual as a basis for melanoma screening. Arch Dermatol 1998;134:103-4. Stadelmann WK, McMasters K, Digenis AG, Reintgen DS. Cutaneous melanoma of the head and neck: advances in evaluation and treatment. Plast Reconstr Surg 2000;05:2105-26. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology, v. 2.2005, Melanoma. Rockledge PA: NCCN, Inc.; 2005. Visit www.nccn.org for recent updates. Accessed December 13, 2005. Johnson TM, Bradford CR, Gruber SB, Sondak VK, Schwartz JL. Staging workup, sentinel node biopsy, and follow-up tests for melanoma. Arch Dermatol 2004;140:107-13. Ardizzoni A, Grimaldi A, Repetto L, Bruzzone M, Sertoli MR, Rosso R. Stage I-II melanoma: the value of metastatic work-up. Oncology 1987;44:87-9. Khansur T, Sanders J, Das SK. Evaluation of staging workup in malignant melanoma. Arch Surg 1989;124:847-9. Wang TS, Johnson TM, Cascade PN, Redman BG, Sondak VK, Schwartz JL. Evaluation of staging chest radiographs and serum lactate dehydrogenase for localized melanoma. J Am Acad Dermatol 2004;51:399-405. Buzaid AC, Sandler AB, Mani S, Curtis AM, Poow J, Bolognia JL, et al. Role of computed tomography in the staging of primary melanoma. J Clin Oncol 1993;11:638-43. Curtis AM, Ravin CE, Deering TF, Putman CE, McCloud TC, Greenspan RH. The efcacy of full-lung tomography in the detection of early metastatic disease from melanoma. Radiology 1982;144:27-9. Au FC, Maier WP, Malmud LS, Goldman LI, Clark WH Jr. Preoperative nuclear scans in patients with melanoma. Cancer 1984;53:2095-7. Roth JA, Eilber FR, Bennett LR, Morton DL. Radionuclide photoscanning: usefulness in preoperative evaluation of melanoma patients. Arch Surg 1975;110:1211-2. Johnson TM, Chang A, Redman B, Rees R, Bradford CR, Riba M, et al. Management of melanoma with a multidisciplinary melanoma clinic model. J Am Acad Dermatol 2000;42:820-6. Basler GC, Fader DJ, Yahanda A, Sondak VK, Johnson TM. The utility of ne needle aspiration in the diagnosis of melanoma metastatic to lymph nodes. J Am Acad Dermatol 1997;36:403-8. Balch CM, Soong SJ, Gershenwald JE, Thompson JF, Reintgen DS, Cascinelli N, et al. Prognostic factors analysis of 17,600 melanoma patients: validation of the American Joint Committee on Cancer melanoma staging system. J Clin Oncol 2001;19:3622-34. Balch CM, Buzaid AC, Soong SJ, Atkins MB, Cascinelli N, Coit DG, et al. Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 2001;19:3635-48. Breslow A. Thickness, cross-sectional areas and depth of invasion in the prognosis of cutaneous melanoma. Ann Surg 1970;172:902-8. Balch CM, Wilkerson JA, Murad TM, Soong SJ, Ingalls AL, Maddox WA. The prognostic signicance of ulceration of cutaneous melanoma. Cancer 1980;45:3012-7. Azzola MF, Shaw HM, Thompson JF. Tumor mitotic rate is a more powerful
Curr Probl Surg, November 2006

105.

106.

107.

108.

109.

110.

111.

112.

113.

114. 115.

116. 117. 118. 119. 120.

121.

prognostic indicator than ulceration in patients with primary cutaneous melanoma, an analysis of 3661 patients from a single center. Cancer 2003;97:1488-98. Ostmeier H, Fuchs B, Otto F, Mawick R, Lippold A, Krieg V, et al. Can immunohistochemical markers and mitotic rate improve prognostic precision in patients with primary melanoma? Cancer 1999;85:2391-99. Buzaid AC, Tinoco LA, Jendiroba D, Tu ZN, Lee JJ, Legha SS, et al. Prognostic value of size of lymph node metastases in patients with cutaneous melanoma. J Clin Oncol 1995;13:2361-8. Buzaid AC, Ross MI, Balch CM, Soong S, McCarthy WH, Tinoco L, et al. Critical analysis of the current American Joint Committee on Cancer staging system for cutaneous melanoma and proposal of a new staging system. J Clin Oncol 1997;15:1039-51. Harrist T, Rigel D, Day C Jr, Sober AJ, Lew RA, Rhodes AR, et al. Microscopic satellites are more highly associated with regional lymph node metastases than with primary melanoma thickness. Cancer 1984;53:2183-7. Morton DL, Davtyan DG, Wanek LA, Foshag ILA, Cochran AJ. Multivariate analysis of the relationship between survival and the microstage of primary melanoma by Clark level and Breslow thickness. Cancer 1993;71:3737-43. Brand CU, Ellwanger U, Stroebel W, Meier F, Schlagenhauff B, Rassner G, et al. Prolonged survival of 2 years or longer for patients with disseminated melanoma, an analysis of related prognostic factors. Cancer 1997;79:2345-53. Deichmann M, Benner A, Bock M, Jackel A, Uhl K, Waldmann V, et al. S100-Beta, melanoma-inhibiting activity, and lactate dehydrogenase discriminate progressive from nonprogressive American Joint Committee on Cancer stage IV melanoma. J Clin Oncol 1999;17:1891-6. Eton O, Legha SS, Moon TE, Buzaid AC, Papadopoulos NE, Plager C, et al. Prognostic factors for survival of patients treated systemically for disseminated melanoma. J Clin Oncol 1998;16:1103-11. Sirott MN, Bajorin DF, Wong GY, Tao Y, Chapman PB, Templeton MA, et al. Prognostic factors in patients with metastatic malignant melanoma, a multivariate analysis. Cancer 1993;72:3091-8. Barth A, Wanek LA, Morton DL. Prognostic factors in 1,521 melanoma patients with distant metastases. J Am Coll Surg 1995;181:193-201. Garbe C, Bttner P, Bertz J, Burg G, dHoedt B, Drepper H, et al. Primary cutaneous melanoma: prognostic classication of anatomic location. Cancer 1995;75:2492-8. Fisher SR. Cutaneous malignant melanoma of the head and neck. Laryngoscope 1989;99:822-36. Fisher SR, Seigler HF, George SL. Therapeutic and prognostic considerations of head and neck melanoma. Ann Plast Surg 1992;28:78-80. Handley WS. The pathology of melanotic growths in relation to their operative treatment. Lancet 1907;1:996-1003. Breslow A, Macht SD. Optimal size of resection for thin cutaneous melanoma. Surg Gynecol Obstet 1997;145:691-2. Veronesi U, Adamus J, Bandiera DC, Brennhovd O, Caceres E, Cascinelli N, et al. Delayed regional lymph node dissection in stage I melanoma of the skin of the lower extremities. Cancer 1982;49:2420-30. Veronesi U, Cascinelli N, Adamus J, Balch C, Bandiera D, Barchuk A, et al. Thin
829

Curr Probl Surg, November 2006

122.

123. 124. 125.

126. 127.

128. 129.

130. 131.

132. 133. 134. 135. 136.

137. 138. 139.

140.
830

stage I primary cutaneous malignant melanoma: comparison of excision with margins of 1 or 3 cm. N Engl J Med 1998;318:1159-62. Balch CM, Soong SJ, Smith T, Ross MI, Urist MM, Karakousis CP, et al. Long-term results of a prospective surgical trial comparing 2 cm vs. 4 cm excision margins for 740 patients with 1-4 mm melanomas. Ann Surg Oncol 2001;8:101-8. Thomas JM, Newton-Bishop J, AHern R, Coombes G, Timmons M, Evans J, et al. Excision margins in high-risk melanoma. N Engl J Med 2004;350:757-66. Johnson TM, Sondak VK. Melanoma margins: the importance and need for more evidence-based trials. Arch Dermatol 2004;140:1148-50. Heaton KM, Sussman JJ, Gershenwald JE, Lee JR, Reintgen DS, Manseld PF, et al. Surgical margins and prognostic factors in patients with thick ( 4mm) primary melanomas. Ann Surg Oncol 1998;5:322-8. Barzilai DA, Singer ME. The potential impact on melanoma mortality of reducing rates of suboptimal excision margins. J Invest Dermatol 2003;120:1067-72. Kenady DE, Brown BE, McBride CM. Excision of underlying fascia with a primary malignant melanoma: effect on recurrence and survival rates. Surgery 1982;92:615-8. Johnson TM, Smith JW II, Nelson BR, Chang A. Current therapy for cutaneous melanoma. J Am Acad Dermatol 1995;32:689-707. Johnson TM, Headington JT, Baker SR, Lowe L. Usefulness of the staged excision for lentigo maligna and lentigo maligna melanoma: the square procedure. J Am Acad Dermatol 1997;37:758-64. Lent WM, Ariyan S. Flap reconstruction following wide local excision for primary malignant melanoma of the head and neck region. Ann Plast Surg 1994;33:23-7. Uren RF, Howman-Giles RB, Shaw HM, Thompson JF, McCarthy WH. Lymphoscintigraphy in high risk melanoma of the trunk: predicting draining node groups, dening lymphatic channels and locating the sentinel node. J Nucl Med 1993;34:1435-40. Wanebo HJ, Cooper PH, Young DV, Harpole DH, Kaiser DL. Prognostic factors in head and neck melanoma. Cancer 1988;62:831-7. Byers RM, Smith JL, Russell N, Rosenberg V. Malignant melanoma of the external ear, review of 102 cases. Am J Surg 1980;140:518-21. Cole DJ, Mackay GJ, Walker BF, Wooden WA, Murray DR, Coleman JJ 3rd. Melanoma of the external ear. J Surg Oncol 1992;50:110-4. Balch CM. The role of elective lymph node dissection in melanoma: rationale, results, and controversies. J Clin Oncol 1988;6:163-72. Myers JN. Value of neck dissection in the treatment of patients with intermediatethickness cutaneous malignant melanoma of the head and neck. Arch Otolaryngol Head Neck Surg 1999;25:110-7. Byers RM. Treatment of the neck in melanoma. Otolaryngol Clin North Am 1998;31:833-9. Goepfert H, Jesse RH, Ballantyne AJ. Posterolateral neck dissection. Arch Otolaryngol 1980;106:618-20. Sim FH, Taylor WF, Ivins JC, Pritchard DJ, Soule EH. A prospective randomized study of the efcacy of routine elective lymphadenopathy in management of malignant melanoma. Cancer 1978;41:948-56. Cascinelli N, Morabito A, Santinami M, MacKie RM, Belli F. Immediate or
Curr Probl Surg, November 2006

141.

142.

143. 144.

145. 146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

delayed dissection of regional nodes in patients with melanoma of the trunk: a randomized trial. WHO Melanoma Programme. Lancet 1998;351:793-6. Veronesi U, Adamus J, Bandiera DC, Brennhovd IO, caceres E, Cascinelli N, et al. Inefcacy of immediate node dissection in stage I melanoma of the limbs. N Engl J Med 1977;297:627-30. Balch CM, Soong SJ, Bartolucci AA, Urist MM, Karakousis CP, Smith TJ, et al. Efcacy of an elective regional lymph node dissection of 1 to 4 mm thick melanomas for patients 60 years of age and younger. Ann Surg 1996;224:255-66. McMasters KM, Sondak VK, Lotze MT, Ross MI. Recent advances in melanoma staging and therapy. Ann Surg Oncol 1999;6:467-75. Balch CM, Soong SJ, Ross MI, Urist MM, Karakousis CP, Temple WJ, et al. Long-term results of a multi-institutional randomized trial comparing prognostic factors and surgical results for intermediate thickness melanomas (1.0 to 4.0 mm): Intergroup Melanoma Surgical Trial. Ann Surg Oncol 2000;7:87-97. Urist MN, Balch CM, Soong S, Milton GW, Shaw HM, McGovern VJ, et al. Head and neck melanoma in 534 clinical stage I patients. Ann Surg 1984;200:769-75. Morton DL, Wen DR, Wong JH, Economou JS, Cagle LA, Storm FK, et al. Technical details of intraoperative lymphatic mapping for early stage melanoma. Ann Surg 1992;127:392-9. Rousseau DL, Ross MI, Johnson MM, Prieto VG, Lee JE, Manseld PF, et al. Revised American joint committee on cancer staging criteria accurately predict sentinel lymph node positivity in clinically node-negative melanoma patients. Ann Surg Oncol 2003;10:569-74. McMasters KM, Reintgen DS, Ross MI, Gershenwald JE, Edwards MJ, Sober A, et al. Sentinel lymph node biopsy for melanoma: controversy despite widespread agreement. J Clin Oncol 2001;19:2851-5. McMasters KM, Noyes RD, Reintgen DS, Goydos JS, Beitsch PD, Davidson BS, et al. Lessons learned from the sunbelt melanoma trial. J Surg Oncol 2004;86:212-23. Morton DL, Thompson JF, Essner R, Elashoff R, Stern SL, Nieweg OE, et al. Validation of the accuracy of intraoperative lymphatic mapping and sentinel lymphadenectomy for early-stage melanoma. Ann Surg 1999;230:453-63. Morton DL, Cochran AJ, Thompson JF, Elashoff R, Essner R, Glass ED, Mozzillo N, et al. Sentinel node biopsy for early-stage melanoma accuracy and morbidity in MSLT-I, an international multicenter trial. Ann Surg 2005;242:302-13. Karimipour DJ, Lowe L, Su L, Hamilton T, Sondak V, Johnson TM, et al. Standard immunostains for melanoma in sentinel lymph node specimens: which ones are most useful? J Am Acad Dermatol 2004;50:759-64. Cohen LM, McCall MW, Hodge SJ. Successful treatment of lentigo maligna and lentigo maligna melanoma with Mohs micrographic surgery aided by rush permanents sections. Cancer 1994;73:2964-70. Wagner JD, Davidson D, Coleman JJ III, Hutchins G, Schauwecker D, Park HM, et al. Lymph node tumor volumes in patients undergoing sentinel lymph node biopsy for cutaneous melanoma. Ann Surg Oncol 1999;6:398-404. Joseph E, Brobeil A, Glass F, Messina J, DeConti R, Cruse CW, et al. Results of complete lymph node dissection in 83 melanoma patients with positive sentinel nodes. Ann Surg Oncol 1998;5:119-25. Cascinelli N, Belli F, Santinami M, Fait V, Testori Q, Ruka W, et al. Sentinel
831

Curr Probl Surg, November 2006

157.

158.

159.

160.

161. 162. 163.

164.

165.

166.

167.

168.

169.

170.

171. 172. 173.


832

lymph node biopsy in cutaneous melanoma: the WHO Melanoma Program experience. Ann Surg Oncol 2000;7:469-774. Haigh PI, Lucci A, Turner RR, Bostick PH, Krasne DL, Stern SL, et al. Carbon dye histologically conrms the identity of sentinel lymph nodes in cutaneous melanoma. Cancer 2001;92:535-41. Su LD, Fullen DR, Lowe L, Wang TS, Schwartz JL, Cimmino VM, et al. Desmoplastic and neurotropic melanoma analysis of 33 patients with lymphatic mapping and sentinel lymph node biopsy. Cancer 2004;100:598-604. Schmalbach CE, Nussenbaum B, Rees RS, Schwartz J, Johnson TM, Bradford CR. Reliability of sentinel lymph node mapping with biopsy for head and neck cutaneous melanoma. Arch Otolaryngol Head Neck Surg 2003;129:61-5. Gershenwald JE, Thompson W, Manseld PF, Lee JE, Colome MI, Tseng CH, et al. Multi-institutional melanoma lymphatic mapping experience: the prognostic value of sentinel lymph node status in 612 stage I or II melanoma patients. J Clin Oncol 1999;17:976-83. Chao C, Wong SL, Edwards MJ, Ross MI, Reintgen DS, Noyes D, et al. Sentinel lymph node biopsy for head and neck melanoma. Ann Surg Oncol 2003;10:21-6. McMasters KM, Noyes D, Reintgen D, Goydos JS, Beitsch PD, Davidson BS. Lessons learned from the sunbelt melanoma trial. J Surg Oncol 2004;86:212-23. OBrien CJ, Uren RF, Thompson JF, Howman-Giles RB, Petersen-Schaefer K, Shaw HM, et al. Prediction of potential metastatic sites in cutaneous head and neck melanoma using lymphoscintigraphy. Am J Surg 1995;170:461-6. Eicher SA, Clayman GL, Myers JN, Gillenwater AM. A prospective study of intraoperative lymphatic mapping for head and neck cutaneous melanoma. Arch Otolaryngol Head Neck Surg 2002;128:241-6. Gershenwald JE, Colome MI, Lee JE, Manseld PF, Tseng C, Lee JJ, et al. Patterns of recurrence following a negative sentinel lymph node biopsy in 243 patients with stage I or II melanoma. J Clin Oncol 1998;16:2253-4. Alex JC, Krag DN, Harlow SP, Meijer Sl, Loggie BW, Kuhn J, et al. Localization of regional lymph nodes in melanomas of the head and neck. Arch Otolaryngol Head Neck Surg 1998;124:135-40. Patel SG, Coit DG, Shaha AR, Brady MS, Moyle JO, Singh B, et al. Sentinel lymph node biopsy for cutaneous head and neck melanomas. Arch Otolaryngol Head Neck Surg 2002;128:285-91. Ollila DW, Foshag LJ, Essner R, Stern SL, Morton DL. Parotid region lymphatic mapping and sentinel lymphadenectomy for cutaneous melanoma. Ann Surg Oncol 1999;6:150-4. Pawlik TM, Ross MI, Thompson JF, Eggermont AMM, Gershenwald JE. The risk of in-transit melanoma metastasis depends on tumor biology and not the surgical approach to regional lymph nodes. J Clin Ocol 2005;23:4588-90. Thomas JM, Clark MA. Selective lymphadenectomy in sentinel node-positive patients may increase the risk of local/in-transit recurrence in malignant melanoma. Eur J Surg Oncol 2004;30:686-91. Thomas JM, Clark MA. Sentinel lymph node biopsy: not yet standard of care for melanoma. BMJ 2004;329:170. Thomas JM, Clark MA. Are there guidance issues relating to sentinel node biopsy for melanoma in the UK? Br J Plast Surg 2004;57:689-90. Estourgie SH, Nieweg OE, Valdes Olmos RA, Hoefnagel CA, Kroon BB. Review
Curr Probl Surg, November 2006

174. 175.

176.

177.

178. 179. 180.

181.

182.

183.

184.

185.

186.

187. 188.

189. 190. 191.

and evaluation of sentinel node procedures in 250 melanoma patients with a median follow-up of 6 years. Ann Surg Oncol 2003;10:681-8. Estourgie SH, Nieweg OE, Kroon BB. High incidence of in-transit metastases after sentinel node biopsy in patients with melanoma. Br J Surg 2004;91:1370-1. Kang JC, Wanek LA, Essner R, Faries MB, Foshag LJ, Morton DL. Sentinel lymphadenectomy does not increase the incidence of in-transit metastases in primary melanoma. J Clin Oncol 2005;23:4764-70. Pawlik T, Ross M, Johnson M, Schacherer CW, McClain DM, Manseld PF, et al. Predictors and natural history of in-transit melanoma after sentinel lymphadenectomy. Ann Surg Oncol 2005;12:587-96. Van Poll D, Thompson JF, Colman MH, McKinnon JG, Saw RP, Stretch JR, et al. A sentinel node biopsy does not increase the incidence of in-transit metastasis in patients with primary cutaneous melanoma. Ann Surg Oncol 2005;12:597-608. McMasters KM. The sunbelt melanoma trial. Ann Surg Oncol 2001;8:41S-43S. McMasters KM. What good is sentinel lymph node biopsy for melanoma if it does not improve survival? Ann Surg Oncol 2004;11:810-2. Lee JH, Essner R, Torisu-Itakure H, Wanek L, Wang H, Morton DL. Factors predictive of tumor-positive nonsentinel lymph nodes after tumor-positive sentinel lymph node dissection for melanoma. J Clin Oncol 2004;22:3677-84. Cochran AJ, Wen DR, Huang RR, Wang HJ, Elashoff R, Morton DL. Prediction of metastatic melanoma in nonsentinel nodes and clinical outcome based on the primary melanoma and the sentinel node. Mod Pathol 2004;17:747-55. Sabel MS, Strecher VJ, Schwartz JL, Wang TS, Marimipour DJ, Orringer JS, et al. Predictors of nonsentinel lymph node positivity in patients with a positive sentinel node for melanoma. J Am Coll Surg 2005;201:37-47. Ribuffo D, Gradilone A, Vonella M, Chiummariello S, Cigna E, Haliassos N, et al. Prognostic signicance of reverse transcriptase-polymerase chain reaction-negative sentinel nodes in malignant melanoma. Ann Surg Oncol 2003;10:396-402. Shivers S, Wang X, Li W, Joseph E, Messina J, Glass LF, et al. Molecular staging of malignant melanoma: correlation with clinical outcome. JAMA 1998;280: 1410-5. Bostick PJ, Morton DL, Turner RR, Huyuh KT, Wang HJ, Elashoff R, et al. Prognostic signicance of occult metastases detected by sentinel lymphadenectomy and reverse transcriptase-polymerase chain reaction in early stage melanoma patients. J Clin Oncol 1999;17:3238-44. Morton DL, Hoon DSB, Cochran AJ, Turner RR, Essner R, Takeuchi J, et al. Lymphatic mapping and sentinel lymphadenectomy for early-stage melanoma. Ann Surg 2003;238:538-50. McMasters KM. Molecular staging of melanoma: sensitivity, specicity, and the search for clinical signicance. Ann Surg Oncol 2003;10:336-7. Kalady MF, White RR, Johnson JL, Tyler DS, Seigler HF. Thin melanomas: predictive lethal characteristics form a 30-year clinical experience. Ann Surg 2003;238:528-37. Whited JD, Grichnik JM. Does this patient have a mole or a melanoma? JAMA 1998;279:696-701. Allen PJ, Coit DG. The surgical management of metastatic melanoma. Ann Surg Oncol 2002;9:762-70. Morton DL, Essner R Surgical excision of distant metastases. In: Balch CM,
833

Curr Probl Surg, November 2006

192. 193.

194.

195.

196.

197.

198. 199. 200. 201.

202.

203.

204. 205.

206.

207. 208. 209.


834

Houghton AN, Sober AJ, Soong SJ, editors. Cutaneous Melanoma. St. Louis, MO: Quality Medical Publishing; 2003. Lentsch EJ, Myers JN. Melanoma of the head and neck: current concepts in diagnosis and management. Laryngoscope 2001;111:1209-22. Trask PC, Paterson AG, Hayasaka S, Dunn RL, Riba M, Johnson T. Psychosocial characteristics of individuals with non-stage IV melanoma. J Clin Oncol 2001;19:2844-50. Creagan ET, Cupps RE, Ivins JC, Pritchard DJ, Sim FH, Soule EH, et al. Adjuvant radiation therapy for regional nodal metastases from malignant melanoma, a randomized prospective study. Cancer 1978;42:2206-10. Ang KK, Byers RM, Peters LJ, Maor MH, Wendt CD, Morrison WH, et al. Regional radiotherapy as adjuvant treatment for head and neck malignant melanoma. Arch Otolaryngol Head Neck Surg 1990;116:169-72. Ang KK, Peters LJ, Weber RS, Morrison WH, Frankenthaler RA, Garden AS, et al. Postoperative radiotherapy for cutaneous melanoma of the head and neck region. Int J Radiat Oncol 1994;30:795-8. Ballo MT, Bonnen MD, Garden AS, Myers JN, Gershenwald JE, Zagars GK, et al. Adjuvant irradiation for cervical lymph node metastases from melanoma. Cancer 2003;97:1789-96. Byers RM. The role of modied neck dissection in the treatment of cutaneous melanoma of the head and neck. Arch Surg 1986;121:1338-41. Ridge JA. Adjuvant radiation after lymph node dissection for melanoma. Ann Surg Oncol 2000;7:550-1. Shen P, Wanek LA, Morton DL. Is adjuvant radiotherapy necessary after positive lymph node dissection in head and neck melanoma. Ann Surg Oncol 2000;7:554-9. Ainslie J, Peters LJ, McKay MS Radiotherapy for primary and regional melanoma. In: Balch CM, Houghton AN, Sober AJ, Soong SJ, editors. Cutaneous Melanoma. St. Louis, MO: Quality Medical Publishing; 2003. OBrien CJ, Petersen-Schaefer K, Stevens GN, Bass PC, Tew P, Gebski VJ, et al. Adjuvant radiotherapy following neck dissection and parotidectomy for metastatic malignant melanoma. Head Neck 1997;19:589-94. Stevens G, Thompson JF, Firth I, OBrien CH, McCarthy WH, Quinn MJ. Locally advanced melanoma, results of postoperative hypofractionated radiation therapy. Cancer 2000;88:88-94. Terando A, Sabel MS, Sondak VK. Melanoma: adjuvant therapy and other treatments. Curr Treat Options Oncol 2003;4:187-99. Harwood AR. Conventional fractionated radiotherapy for 51 patients with lentigo maligna and lentigo maligna melanoma. Int J Radiat Oncol Biol Phys 1983;9:1019-21. Atkins MB, Buzaid AC, Houghton AN. Systemic chemotherapy and biochemotherapy. In: Balch CM, Houghton AN, Sober AJ, Soong SJ, editors. Cutaneous Melanoma. St. Louis, MO: Quality Medical Publishing; 2003. Soengas MS, Lowe SW. Apoptosis and melanoma chemoresistance. Oncogene 2003;22:3138-51. Pawlik TM, Sondak VK. Malignant melanoma: current state of primary and adjuvant treatment. Clin Rev Oncol Heme 2003;45:245-64. Li Y, McClay EF. Systemic chemotherapy for the treatment of metastatic melanoma. Semin Oncol 2003;29:413-26.
Curr Probl Surg, November 2006

210. Hill GJ, Moss SE, Golomb FM, Grage TB, Fletcher WS, Minton JP, et al. DTIC and combination therapy for melanoma: III. DTIC (NSC 45388) Surgical Adjuvant study COG Protocol 7040.Cancer 1981;47:2556-62. 211. Biasco G, Pantaleo MA, Casadei S. Treatment of brain metastases of malignant melanoma with temazolomide. N Engl J Med 2001;345:621-2. 212. Shaw PM, Sivanandham M, Bernik SF, Ditaranto K, Wallack MK. Adjuvant immunotherapy for patients with melanoma: are patients with melanoma of the head and neck candidates for this therapy? Head Neck 1997;19:595-603. 213. Kirkwood JM, Strawderman MH, Ernstoff MC, Smith TJ, Borden EC, Blum RH. Interferon alpha-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol 1996;14:7-17. 214. Kirkwood JM, Ibrahim JG, Sondak VK, Richards J, Flaherty LE, Ernstoff MS, et al. High- and low-dose interferon alfa- 2b in high-risk melanoma: rst analysis of Intergroup Trial E1690/S9111/C9190. J Clin Oncol 2000;18:2444-59. 215. Kirkwood JM, Ibrahim JG, Sosman JA, Sondak VK, Agarwala SS, Ernstoff MS, et al. High-dose interferon alfa-2b signicantly prolongs relapse-free and overall survival compared with the GM2-KLH/QS-21 vaccine in patients with resected stage IIB-III melanoma: results of Intergroup Trial E1694/S9512/C509801. J Clin Oncol 2001;19:2370-80. 216. Kilbridge KL, Cole BF, Kirkwood JM, Haluska FG, Atkins MA, Ruckdeschel JC, et al. Quality-of-life-adjusted survival analysis of high-dose adjuvant interferon alpha-2b for high-risk melanoma patients using intergroup clinical trial data. J Clin Oncol 2002;20:1311-8. 217. Cascinelli N, Belli F, MacKie RM, Santinami M, Bufalino R, Morabito A. Effect of long-term adjuvant therapy with interferon alpha-2a in patients with regional node metastases from cutaneous melanoma: a randomized trial. Lancet 2001;358:866-9. 218. Sabel MS, Sondak VK. Pros and cons of adjuvant interferon in the treatment of melanoma. Oncologist 2003;8:451-8. 219. Robbins PG, Rosenberg SA Melanoma antigens and their use as vaccines. In: Balch CM, Houghton AN, Sober AJ, Soong SJ, editors. Cutaneous Melanoma. St. Louis, MO: Quality Medical Publishing; 2003. 220. Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin-2. JAMA 1994;271:907-13. 221. Banky JP, Kelly JW, English DR, Yeaman JM, Dowling JP. Incidence of new and changed nevi and melanomas detected using baseline images and dermoscopy in patients at high risk for melanoma. Arch Dermatol 2005;141:998-1006. 222. Bittner M, Meltzer P, Chen Y, Jiang Y, Seftor E, Hendrix M, et al. Molecular classication of cutaneous malignant melanoma by gene expression proling. Nature 2000;406:536-40. 223. Myers JN. Adjuvant immunotherapy for patients with melanoma. Head Neck 1998;20:270.

Curr Probl Surg, November 2006

835

You might also like