当前位置: 首页 > 期刊 > 《新英格兰医药杂志》 > 2004年第15期 > 正文
编号:11306683
Management of Cutaneous Melanoma
http://www.100md.com 《新英格兰医药杂志》
     PubMed Citation

    Cutaneous melanoma remains a management challenge. The National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) database documents increases of 619 percent in annual diagnoses of cutaneous melanoma and of 165 percent in annual mortality from 1950 to 2000.1 In 2004, an estimated 55,000 Americans will receive a diagnosis of cutaneous melanoma, and 7900 will die from the disease.2 Of all cancers in the United States, cutaneous melanoma ranks fifth in incidence among men and seventh among women2 and is the second leading cause of lost productive years3,4; it is also the most common cancer among women 20 to 29 years of age.5

    Melanoma Screening and Identification of High-Risk Persons

    Efforts to reduce the incidence of cutaneous melanoma have focused on identifying and screening persons at high risk and on promoting sun protection. People with light complexions, an inability to tan, blond or red hair, or blue eyes have a higher risk of melanoma than the general population.6 Recently, inherited mutations in the melanocortin-1 receptor have been linked to people with red hair, to photosensitivity, and to an increased risk of cutaneous melanoma.7

    Having many pigmented lesions, including freckles and either common or clinically atypical moles (Figure 1), is also associated with an increased risk of cutaneous melanoma.7 Although some moles are precursors to cutaneous melanoma,8 more often they are markers of an increased risk.7 Intermittent sun exposure and severe sunburns, especially during childhood, and use of tanning beds have been associated with an increased risk of cutaneous melanoma.9,10

    Figure 1. Clinical Images of Pigmented Lesions.

    Multiple clinically atypical nevi are distributed over the back of a patient (Panel A). In Panel B, a cluster of clinically atypical moles have central papular components ("fried-egg nevi") and peripheral diffusion of pigment. A large (2.5-cm) nevus has a fuzzy border but relatively symmetric features (Panel C). In Panel D, superficial spreading melanoma is characterized by a dark brown plaque with highly irregular, scalloped borders and extensive color variegation. In Panel E, acral–lentiginous melanoma appears as a large ulcerative nodule on the plantar surface. In Panel F, lentigo maligna melanoma is manifested as an irregular, kidney-shaped, thin brown plaque on the face. In Panel G, nodular melanoma appears as a relatively symmetric, sharply circumscribed nodule with a blue–gray dermal invasive component. Photographs courtesy of Dr. Richard Allen Johnson.

    Prior cutaneous melanoma is associated with an increased risk of a second primary cutaneous melanoma.11,12 Recent analysis from the SEER database showed that the rate of subsequent cutaneous melanomas among persons with a history of melanoma was more than 10 times the rate of a first cutaneous melanoma among the general SEER population. Furthermore, the period of greatest risk is the first two years after diagnosis.12

    Patients with a strong family history of melanoma and multiple clinically atypical moles are at the greatest risk for cutaneous melanoma.13,14,15,16,17 Inherited mutations in the CDKN2A and CDK4 genes, which have been documented in some families with hereditary melanoma,7 confer a 60 to 90 percent lifetime risk of melanoma.18 Although commercially available, genetic testing for hereditary melanoma is currently considered a research tool and is not routinely recommended.

    Because skin is readily accessible to direct visual inspection, screening of high-risk persons is strongly recommended. Educational efforts to reduce the incidence of melanoma and the associated mortality promote recognition of early lesions (including an "ABCD" evaluation for asymmetry, border irregularity, color variegation, and a diameter that is greater than 6 mm or increasing in size) and avoidance of sun exposure. The benefit of mass screening has been studied in Australia,19 the Netherlands,20 and the United States.21 The American Academy of Dermatology has long sponsored annual free-access skin checks. In a study conducted from 1992 to 1994, about 2 percent of 282,000 patients screened had suspicious lesions, approximately 8 percent of which proved to be melanoma. Notably, screening through the American Academy of Dermatology program detected a higher percentage of early lesions than were recorded in the 1990 SEER database.21 Similarly, melanomas that are detected by physicians are diagnosed earlier than lesions that are recognized by patients.22 Although such efforts have promoted earlier identification of lesions, a direct reduction in mortality has yet to be documented. Nevertheless, for persons at highest risk (i.e., those with a strong family history of melanoma and multiple clinically atypical moles), frequent self-examination and professional evaluation at least once a year are necessary.

    Identification of High-Risk Lesions

    A full skin examination, including mucous membranes, is essential for all people at risk; prior photographs of atypical lesions can be helpful. Clinical recognition of cutaneous melanomas and discrimination from surrounding clinically atypical moles can be difficult (Figure 1). Melanomas frequently display irregularities in shape, color, and border; however, these features are neither invariant nor specific. Changing or symptomatic lesions or moles that stand out from other surrounding moles deserve prompt histologic evaluation by excision with narrow margins (1 to 3 mm). Subsequent pathological assessment requires multiple sections through the lesion to establish the thickness of the tumor (a measurement, in millimeters, of the distance from the epidermal granular layer to the base of the tumor at its thickest point) and the pathological stage of the disease (Figure 2 and Table 1). Superficial shave biopsies are suboptimal, since deep margins cannot be fully ascertained.

    Figure 2. Relationship between the Stage of Melanoma and Survival.

    Kaplan–Meier survival curves are adapted from the American Joint Committee on Cancer.23

    Table 1. Staging Criteria for Melanoma.

    Melanomas in the nail matrix often produce longitudinal pigmentation and require sampling of the matrix beneath the proximal fold. Nail-bed lesions frequently distort the nail plate, simulating a fungal infection, and require evaluation of both the plate and the underlying tissue bed.

    Staging and Treatment of Primary Melanoma

    A management schema is shown in Figure 3. After histologic confirmation of cutaneous melanoma, pathological staging of the primary tumor guides the prognosis and decisions regarding further surgery.23 Increases in the maximal thickness of a tumor and microscopical ulceration are both inversely correlated with survival and can be used to provide reasonable survival estimates. However, nodal status — determined by sentinel-lymph-node biopsy — has emerged as the most powerful predictor of recurrence and survival.24

    Figure 3. Algorithm for the Management of Cutaneous Melanoma.

    SLNB denotes sentinel-lymph-node biopsy, FNA fine-needle aspiration, CLND completion lymph-node dissection, CT computed tomography, MRI magnetic resonance imaging, PET positron-emission tomography, CNS central nervous system, and XRT external radiation therapy. A minus sign indicates no evidence of disease, and a plus sign evidence of metastasis. Patients who are candidates for sentinel-lymph-node biopsy should be referred to a surgeon experienced in performing the procedure so that the benefits and complications can be fully discussed.

    Wide local excision is the treatment for the primary melanoma. Several randomized, controlled trials have determined what the proper resection margin should be according to the thickness of the lesion (Table 2).25,26,27,28,29,30 In these trials, patients were enrolled on the basis of the thickness of the tumor and randomly assigned to excision with wide margins (3 to 5 cm) or narrow margins (1 or 2 cm). Because most of the trials showed similar outcomes in the two groups, narrower margins have been accepted for tumors of most thicknesses.

    Table 2. Rates of Recurrence and Overall Survival in Randomized, Prospective Studies of Excision Margins in Patients with Primary Melanoma.

    For melanoma in situ, margins of 0.5 to 1 cm around the visible lesion or biopsy scar are recommended. For thin melanomas (1.0 mm or less), a 1-cm margin is accepted.25,26 Although no trials have specifically targeted melanomas that are 1.0 to 2.0 mm thick, most centers recommend a margin of 2 cm if anatomically possible (otherwise, a margin of 1 cm is recommended). This recommendation is derived from two trials showing that margins of 2 cm and 5 cm were equivalent for treating melanomas of 2.0 mm or less27,28 and one trial that reported a higher, but nonsignificant, rate of local recurrence at 12 years among patients with lesions that are 1.0 to 2.0 mm thick who are treated with a 1-cm excision margin as compared with those treated with a 3-cm margin (4.2 percent vs. 1.5 percent).25

    For tumors between 2.0 and 4.0 mm in thickness, the U.S. Intergroup Melanoma Surgical Trial established the adequacy of a 2-cm margin.29 The recent United Kingdom Melanoma Study Group Trial30 randomly assigned 900 patients with melanomas that had a thickness of 2.0 mm or more to two groups, with excision margins of 1 cm and 3 cm. The study showed no significant difference in the rate of local recurrence or in overall survival between the two groups, although the group with 1-cm margins had more combined locoregional recurrences. Although the findings of the study argue against a 1-cm margin for lesions that are 2.0 to 4.0 mm in thickness, the data do not support a preference for margins of 2 cm versus 3 cm.

    Since thick melanomas (larger than 4.0 mm) are associated with a high risk of nodal and distant metastases, more extensive resection is unlikely to mitigate the outcome substantially. One retrospective analysis showed no significant benefit with respect to either local recurrence or overall survival rates among patients with thick tumors who underwent excision with margins greater than 2 cm31; thus, a 2-cm margin is probably adequate. On the basis of its recent findings, the United Kingdom Melanoma Study Group does recommend a 3-cm margin for thick tumors,30 even though its superiority over a 2-cm margin has not been established. Melanomas in unusual sites (e.g., in the nail bed or the nail matrix, on the fingers, and on the soles of the feet) are uncommon and require special surgical attention.32

    Since patients who are rendered disease-free by surgery are still at risk for regional and distant relapse and for additional primary melanomas, surveillance aimed at early detection of new lesions is needed. Although an optimal follow-up interval has not been determined, at minimum, an annual routine physical examination, including a full skin assessment and palpation of lymph nodes, is important. More frequent visits are appropriate for patients at high risk for multiple primary lesions (i.e., patients with multiple clinically atypical moles or a family history of melanoma) or for relapse (i.e., stage II or III disease). Routine laboratory tests, including serum lactate dehydrogenase, albumin, and plasma hemoglobin measurements and chest radiography,33,34 have not been shown to be beneficial in screening for visceral disease in asymptomatic patients; therefore, these tests are usually reserved for patients with disease of stage II or higher.

    Nodal Sampling and Staging

    Interest in elective lymph-node dissection grew out of the hypothesis that melanoma cells spread to regional nodal basins before metastasizing widely. Thus, early removal of nodal deposits may prevent subsequent dissemination. Four prospective trials were designed to test this hypothesis,35,36,37,38 and all four failed to show an overall survival benefit. Debate over the merits of elective lymph-node dissection has largely been subsumed by the emergence of sentinel-lymph-node biopsy as a staging, and possibly therapeutic, procedure.

    In the early 1990s, Morton et al.39 developed sentinel-lymph-node biopsy to sample selectively the first draining lymph node (or nodes) from a tumor site. In their initial report, the investigators (with the use of vital blue dye) identified the sentinel lymph node in 82 percent of lymphadenectomies and identified metastases in 21 percent of sentinel lymph nodes; in the absence of involved sentinel lymph nodes, subsequent completion lymphadenectomy revealed melanoma in only 1 percent of the nonsentinel nodes. Current protocols use technetium-99m–labeled radiocolloids in addition to vital dye40,41 for maximal accuracy. Successful sentinel-lymph-node biopsy requires a multidisciplinary team that includes an experienced surgeon, a nuclear medicine radiologist, and a pathologist. In skilled hands, the sentinel lymph node can be identified more than 95 percent of the time with less than 5 percent false negative results.41

    Sentinel nodes should be fixed and processed with both hematoxylin and eosin and immunohistochemical stains (e.g., S-100, HMB-45, and MART-1), since hematoxylin and eosin alone will miss up to 12 percent of positive nodes.42 Since fewer nodes are submitted for examination, more detailed serial analysis, with detection of small micrometastases, is possible. Although benign collections of melanocytes may occasionally be present, the detection of intranodal deposits of melanocytic cells usually indicates metastasis.43 More recent investigations suggest that the amount and pattern of metastatic deposits in the sentinel lymph node (e.g., tyrosinase messenger RNA molecules and single cells, cell clusters, micrometastases, and macrometastases),44,45 as well as the function of the immune cells within the nodal basin,46 may have prognostic significance.

    The likelihood of detecting metastatic deposits in the sentinel lymph node is approximately 1 percent if the thickness of the tumor is less than 0.8 mm, 8 percent if it is 0.8 to less than 1.5 mm, 23 percent if it is 1.5 to less than 4.0 mm, and 36 percent if it is 4.0 mm or greater.47,48 Since many large retrospective studies24,44,48,49,50,51,52 have shown a strong negative correlation between the presence of metastatic melanoma in sentinel nodes and survival, sentinel-lymph-node biopsy is currently the most powerful staging and prognostic tool. As such, proponents argue that the performance of this minimally invasive procedure provides prognostic information and identifies candidates for systemic adjuvant treatment, such as with interferon alfa-2b. However, critics argue that sentinel-lymph-node biopsy has not been shown to improve survival and that interferon alfa-2b has not been proven effective for microscopical nodal disease. The Multicenter Selective Lymphadenectomy Trial and the Sunbelt Melanoma Trial were designed to address these concerns.53

    The Multicenter Selective Lymphadenectomy Trial assigned 2001 patients whose tumors were 1.0 mm or greater or were classified as Clark level IV or V to undergo wide resection, either alone or with sentinel-lymph-node biopsy. The more complex design of the Sunbelt Melanoma Trial assessed the value of a reverse-transcriptase–polymerase-chain-reaction (RT-PCR) assay for molecular identification of regional disease and the roles of completion lymph-node dissection and adjuvant interferon alfa-2b therapy on the basis of the extent of regional nodal involvement. Until complete data from these trials become available, judicious use of sentinel-lymph-node biopsy must balance the pretest likelihood of detecting melanoma within a node against the complication rate associated with the procedure and its cost. Early results for 2120 patients in the Sunbelt Melanoma Trial estimate complication rates of 4 percent for sentinel-lymph-node biopsy alone and 23 percent for sentinel-lymph-node biopsy along with completion lymph-node dissection.54

    Since patients with melanomas that are 1.0 mm or less in thickness rarely have nodal disease,48 sentinel-lymph-node biopsy is not commonly performed but could be considered if pathological examination showed negative prognostic features such as ulceration or Clark level IV to V invasion.23 For patients with melanomas that are more than 1 mm thick, sentinel-lymph-node staging can be considered in order to estimate the prognosis and to determine eligibility for clinical trials and the need for adjuvant therapy. Further studies are needed to determine the additional benefit of completion lymph-node dissection in patients who have positive results on sentinel-lymph-node biopsy, the effect of prior wide resection on the accuracy of sentinel-lymph-node biopsy, and the role of sentinel-lymph-node biopsy in patients who are not candidates for completion lymph-node dissection or adjuvant therapy.

    Adjuvant Therapy

    One of the advantages of increasingly accurate staging is that it can identify patients whose risk of recurrence is sufficiently high to justify adjuvant systemic treatment.23 Attempts to reduce recurrent melanoma with adjuvant therapy date back several decades and have been studied in more than 100 randomized, controlled trials.55 Recent efforts focus on approaches involving interferon alfa-2b, vaccines, or both.

    Although a variety of approaches have been beneficial in comparisons with the use of historical controls, only interferon alfa-2b has been shown to have a reproducible benefit. Three U.S. cooperative group studies showed improvement in relapse-free survival, and two of the three also showed significantly improved overall survival among patients receiving high-dose interferon alfa-2b (Table 3).56,57,58 Since therapy with high-dose interferon alfa-2b improved relapse-free survival by 20 to 30 percent and overall survival by as much as 30 percent, the Food and Drug Administration (FDA) approved this treatment for patients with primary lesions thicker than 4 mm (i.e., stage IIB or IIC) or melanoma involving regional lymph nodes that have been rendered disease-free by surgery (i.e., stage III). Although analyses in individual studies suggested that the benefit of high-dose interferon alfa-2b may be restricted to certain patient populations on the basis of the number of nodes that are involved, no consistent pattern has been observed. Thus, it is reasonable to propose that the benefit of interferon alfa-2b is proportional to the risk of recurrence and that therapy can be considered for all patients in whom the potential benefit outweighs the expected toxic effects, regardless of the number of diseased nodes. In general, this would include patients with stage IIB, IIC, or III disease, no serious coexisting illnesses, and a life expectancy of more than 10 years.59

    Table 3. Relapse-free and Overall Survival in Trials of Interferon Alfa for Stage III Melanoma.

    High-dose interferon alfa-2b has many side effects, including acute constitutional symptoms, chronic fatigue, headache, nausea, weight loss, myelosuppression, and depression.60 Although these side effects can often be managed with appropriate supportive care,61,62 most patients will require an adjustment of the dose during therapy. Thus, high-dose interferon alfa-2b should be administered by health care professionals who are familiar with its side effects.

    Despite the toxicity, modest efficacy, and considerable expense of interferon alfa-2b, retrospective analyses show that the agent is associated with improved quality-of-life-adjusted survival63 and is relatively cost-effective.64 Additional evidence suggests that patients at high risk for a recurrence of melanoma prefer interferon alfa-2b therapy and its attendant toxic effects to even a slight increase in the risk of relapse.65 Critics, however, have expressed concern about toxicity and the lack of a consistent survival advantage.66 For example, the significant survival benefit with interferon alfa-2b, as compared with observation alone, in the initial report of the E1684 trial was no longer apparent when the data were reanalyzed at a median follow-up of 12.6 years.67 Furthermore, other investigators have suggested without substantiation that the marked survival advantage observed in the more recent E1694 trial may be due, in part, to a deleterious effect of the experimental ganglioside vaccine rather than to a therapeutic effect of interferon alfa-2b.66 Finally, meta-analyses of many randomized, controlled trials involving treatment with interferon at various doses failed to show a survival advantage in the aggregate.68

    Since high-dose interferon alfa-2b has considerable toxic effects and prevents recurrence and death in only a minority of the patients at risk, it is not currently accepted worldwide and is inconsistently used in the United States. In addition, the intensity and duration of the regimen make it difficult to combine with other approaches. Efforts to improve the efficacy and toxicity profiles of interferon have included the use of lower-dose regimens and combination with various vaccines.

    Low-dose interferon has been extensively tested in Europe. Several trials have failed to show that low-dose interferon, as compared with observation alone, results in an increase in recurrence-free survival or overall survival in patients with high-risk stage II or III disease,57,69,70 although two trials did report an increase in recurrence-free survival among patients with intermediate-risk melanoma.71,72 This benefit led to approval, in several European countries, of low-dose interferon alfa-2b for patients with lesions of intermediate thickness. Nevertheless, no studies have yet shown that regimens of low-dose interferon alfa-2b provide an overall survival advantage; therefore, such regimens cannot be routinely recommended.

    Other trials have evaluated interferon regimens that omit either the high-dose intravenous induction phase or the subcutaneous maintenance phase. A recent European trial suggested a significant delay in distant recurrence, but not an improvement in overall survival, with interferon alfa-2b (5 million units) administered subcutaneously three times a week for two years.66 This delayed rate of relapse prompted European investigators to study longer durations of interferon therapy with the use of a pegylated interferon. In contrast, the early improvement in recurrence-free survival but not overall survival that is consistently seen with high-dose interferon prompted U.S. investigators to compare the four-week intravenous induction phase alone with observation alone in patients with stage IB to IIIA melanoma (in the E1697 study) and in those with regional nodal disease detected only by RT-PCR for melanoma-associated mRNA (in the Sunbelt Melanoma Trial).73

    Other investigators have attempted to improve the efficacy of interferon therapy in patients with stage III disease by combining intermediate-dose interferon with Melacine (a vaccine comprising melanoma-cell lysates mixed in an adjuvant called Detox74) or by incorporating intermediate-dose interferon into a regimen that includes several chemotherapy drugs and interleukin-2 (biochemotherapy). A trial of high-dose interferon as compared with Melacine plus intermediate-dose interferon has completed enrollment, and a preliminary analysis showed no significant difference in efficacy between the two study groups.75 Studies comparing a nine-week course of intensive biochemotherapy with a year of therapy with high-dose interferon are under way as a joint effort among the various U.S. oncology cooperative groups.

    The appropriate adjuvant treatment for patients with intermediate- or high-risk melanoma remains controversial. Since interferon alfa-2b is the only FDA-approved therapy, it is reasonable to inform patients for whom this therapy would be appropriate about its potential risks and benefits and known adverse effects. Patients who are not able to tolerate interferon or who prefer treatments that are less toxic or more aggressive should be encouraged to consider participating in controlled clinical trials. Such studies ideally enroll patients who have undergone adequate staging and whose disease has been stratified accordingly.

    Various tumor-specific vaccines have been investigated as potential adjuvant therapy for patients with high-risk melanoma. These vaccines include a GM2-ganglioside–based vaccine76; a shed melanoma-antigen vaccine77; a polyvalent whole-cell vaccine, Canvaxin78; a dinitrophenyl-conjugated autologous tumor vaccine, M-Vax79; and the aforementioned Melacine vaccine.74 When administered as adjuvant therapy, many of these vaccines have been reported to produce responses in some patients with advanced disease and to prolong survival significantly in comparison with historical controls.

    Given the evolution in the methods for staging melanoma (including high-resolution computed tomography, positron-emission tomography, and sentinel-lymph-node biopsy), contemporary controls are essential to evaluate adjuvant treatments. To date, few large-scale, randomized, controlled trials of vaccine efficacy have been reported. Livingston et al. initially reported a small phase 3 trial comparing a GM2 ganglioside vaccine administered along with bacille Calmette–Guérin (BCG) with BCG alone in patients with stage III disease.76 Immunoglobulin M antibodies developed in 80 percent of patients who received the GM2 vaccine, whereas 10 percent of patients in both groups had antibody present at enrollment. When patients with preexisting antibodies were excluded from the analysis, a significant improvement in recurrence-free survival was observed in patients receiving the GM2 vaccine. Unfortunately, a modified, more immunogenic version of this vaccine fared poorly when compared with high-dose interferon.58

    When treatment with Melacine was compared with observation alone in patients with stage II disease, the vaccine did not increase either recurrence-free or overall survival in the treated population as a whole. However, a prospectively defined subgroup of patients with expression of either the HLA-A2 haplotype or the HLA-C3 haplotype, or both, had significantly longer disease-free and overall survival, as compared with patients with other HLA types who received the vaccine and with patients who were followed by observation alone.80 However, prospective confirmation of this intriguing result is lacking.

    Investigators have identified HLA-restricted melanoma antigens recognized by CD8 T cells in patients who have a response to T-cell–derived immunotherapy.81 These peptide antigens have been cloned and are being tested in patients with stage IV melanoma, either alone or in combination with dendritic cells,82 anti–cytokine-T-lymphocyte–associated protein 4 (CTLA-4) antibody,83 or cytokines, such as interleukin-2,84 interferon, and granulocyte–macrophage colony-stimulating factor (GM-CSF).85 Many of these promising strategies are also being investigated in the adjuvant setting. Although vaccines are less toxic than some other treatments, their efficacy relative to that of interferon and their ability to synergize with interferon remain to be established.

    Treatment of Distant Disease

    For patients with stage IV melanoma, no randomized, controlled trials have shown a significant survival advantage with the use of any specific drug or combination of drugs, including dacarbazine and a high-dose bolus of interleukin-2, both of which have been approved by the FDA for the treatment of stage IV disease (Table 4).86 Although surgical resection can result in five-year survival rates of up to 25 percent,87 these results have usually been achieved in highly selected patients with isolated metastases or stage M1a disease. The usual outcome for patients with distant metastases remains bleak, with median survival of 6 to 10 months and less than 5 percent of patients surviving for more than 5 years.23,88

    Table 4. Key Randomized Trials for Stage IV Melanoma.

    High-dose bolus interleukin-2 (600,000 or 720,000 U per kilogram of body weight, administered intravenously three times daily on days 1 to 5 and 15 to 19) received FDA approval in 1998 because the regimen led to durable responses in a meaningful proportion of patients with stage IV disease.89 Updated information regarding these patients, which was presented to the FDA, confirms the durability of the response in this patient population. Although a tumor response occurred in only 16 percent of patients, the median duration of the response has yet to be reached among those with a complete response, and disease progression has not been noted in any patient with a response lasting for more than 30 months. Unfortunately, high-dose bolus interleukin-2 therapy is associated with many severe toxic effects (including hypotension, the capillary-leak syndrome, myocarditis, transient renal insufficiency, and catheter-related sepsis90). These effects have restricted the use of this therapy to highly selected patients being treated by experienced clinicians in specialized programs that are equipped to manage the potential complications.

    Efforts to improve on the therapeutic index associated with the administration of interleukin-2 have included altered dose levels and schedules, measures to block the toxic effects of high-dose bolus interleukin-2, and combinations with other cytokines (e.g., interferon alfa and interleukin-12), immune protectors (e.g., histamine), and various vaccines.84,91,92,93,94 Despite a promising preclinical rationale and encouraging findings in a few small clinical trials, these approaches have yet to yield activity that is sufficient to justify either FDA approval or routine clinical administration.

    Among cytotoxic agents, dacarbazine remains the benchmark, since it produces responses in 15 to 20 percent of patients and the median duration of the response is four months.54,95 The major side effects of dacarbazine are limited to nausea and vomiting. Response rates and the duration of the response do not appear to be affected by administration schedules. The availability of potent antiemetic agents has permitted outpatient administration of dacarbazine at a dose of 1 g per square meter of body-surface area in a convenient schedule of one day every three to four weeks. Temozolomide, an analogue of dacarbazine, is characterized by significant central nervous system penetration and can be absorbed orally.96 Clinical trials have suggested that temozolomide has activity that is equal to that of dacarbazine97 and may be associated with a lower frequency of central nervous system relapse.98,99 However, FDA approval has not been forthcoming. Investigations continue into temozolomide used alone in various dosing schedules and in combination with other therapies. For patients with brain metastases, the combination of temozolomide with thalidomide,100 especially in conjunction with radiation therapy, has generated particular interest; however, data supporting the superiority of these approaches to standard therapy have yet to be produced.

    A variety of regimens combining dacarbazine with other cytotoxic agents, tamoxifen, or interferon alfa have shown promising response rates in single-institution phase 2 trials and potential survival advantages in small phase 3 trials.86 Unfortunately, despite extensive investigation, no randomized, controlled trials have shown these approaches to be superior to dacarbazine alone (Table 4).

    Recent attention has focused on combinations of dacarbazine and cisplatin with interleukin-2 and interferon alfa (i.e., biochemotherapy). Phase 2 studies of such regimens have typically shown tumor responses in 40 to 50 percent of patients,90 and meta-analyses suggest their superiority to regimens involving cytotoxic chemotherapy and immunotherapy alone.101,102 In addition, a single-institution phase 3 trial showed that as compared with chemotherapy, biochemotherapy resulted in a doubling of the response rate and the time to progression, with a prolongation of median survival that approached significance (from 9.2 to 11.9 months).103 Unfortunately, several other phase 3 trials, including two recent studies of the Eastern Cooperative Oncology Group104 and the European Organization for the Research and Treatment of Cancer,105 showed no survival benefit with biochemotherapy as compared with chemotherapy or immunotherapy alone.104,105,106,107 Consequently, routine use of these more toxic combination approaches can no longer be justified.

    Future Directions

    Ongoing therapeutic investigations focus on vaccine-based immunotherapy and targeted-chemotherapy approaches. Although many melanomas carry distinct tumor-associated antigens that can be recognized by the immune system, recent studies have suggested that most patients become tolerant to these antigens early in the course of their disease.108 Efforts to overcome immune tolerance include the use of dendritic-cell or heat-shock-protein–based vaccines,109 concurrent use of immune adjuvants (interleukin-12, cytidine phosphate guanosines, GM-CSF, and anti-CTLA-4 antibody) and gene therapy (intratumoral injection of Allovectin-7 and GM-CSF),110,111 and adoptive transfer of tumor-specific cytolytic T cells after the use of lymphocyte-depleting chemotherapy to eliminate regulatory T cells.112 Although these approaches have begun to show encouraging immunologic activity — including vitiligo,113 enhanced reactivity of tumor-specific T cells in the peripheral blood,85 and occasional clinical responses112 — much work is required before they can be used in the majority of patients with stage IV disease.

    Recent advances in our understanding of the biologic features of melanoma indicate that many tumors evolve strong defenses against chemotherapy-induced apoptosis, including methylation-mediated silencing of the APAF-1 gene,114 sustained expression of Bcl-2, and activating mutations of the BRAF gene.115 Investigations combining chemotherapy with demethylating agents (e.g., 5-aza-2'-deoxycytidine), antisense Bcl-2 oligonucleotides, or RAF inhibitors (e.g., BAY 43-9006) have been initiated, with some encouraging early results.116,117 It is hoped that this better understanding of the biologic features of melanoma and the mechanisms underlying tumor-induced immune suppression will lead to more precisely targeted treatment approaches and rational treatment selection. Given the promise of these new approaches and the limited value of existing standard therapies, advanced disease is best managed by encouraging participation in clinical trials.

    Dr. Atkins reports having received consulting fees from Bayer and Celgene, consulting and lecture fees from Chiron, and grant support from Celgene and Antigenics.

    Source Information

    From the Department of Dermatology (H.T., A.J.S.), Massachusetts General Hospital Melanoma Center (H.T., A.J.S.), and the Wellman Center for Photomedicine (H.T.), Massachusetts General Hospital and Harvard Medical School; and the Division of Hematology/Oncology, Department of Medicine, Biologic Therapy and Cutaneous Oncology Programs, Beth Israel Deaconess Medical Center and Harvard Medical School (M.B.A.) — all in Boston.

    Address reprint requests to Dr. Tsao at the Department of Dermatology, Massachusetts General Hospital, Bartlett 622, 48 Blossom St., Boston, MA 02114.

    References

    National Cancer Institute. 51-Year trends in U.S. cancer death rates. In: SEER cancer statistics review, 1975-2000. (Accessed August 9, 2004, at http://seer.cancer.gov/csr/1975_2000/results_merged/topic_inc_mor_trends.pdf.)

    Jemal A, Tiwari RC, Murray T, et al. Cancer statistics, 2004. CA Cancer J Clin 2004;54:8-29.

    Albert LS, Rhodes AR, Sober AJ. Dysplastic melanocytic nevi and cutaneous melanoma: markers of increased melanoma risk for affected persons and blood relatives. J Am Acad Dermatol 1990;22:69-75.

    Berwick M, Weinstock MA. Epidemiology: current trends. In: Balch C, Houghton A, Sober A, Soong S, eds. Cutaneous melanoma. 4th ed. St. Louis: Quality Medical Publishing, 2003:15-23.

    National Cancer Institute. SEER cancer statistics review, 1975-2001. (Accessed August 9, 2004, at http://seer.cancer.gov/cgi-bin/csr/1975_2001/search.pl#results.)

    Bliss JM, Ford D, Swerdlow AJ, et al. Risk of cutaneous melanoma associated with pigmentation characteristics and freckling: systematic overview of 10 case-control studies. Int J Cancer 1995;62:367-376.

    Tsao H, Sober A. Acquired precursor lesions and markers of increased risk for cutaneous melanoma. In: Balch C, Houghton A, Sober A, Soong S, eds. Cutaneous melanoma. 4th ed. St. Louis: Quality Medical Publishing, 2003:121-34.

    Tsao H, Bevona C, Goggins W, Quinn T. The transformation rate of moles (melanocytic nevi) into cutaneous melanoma: a population-based estimate. Arch Dermatol 2003;139:282-288.

    Elwood JM, Jopson J. Melanoma and sun exposure: an overview of published studies. Int J Cancer 1997;73:198-203.

    Tsao H, Sober AJ. Ultraviolet radiation and malignant melanoma. Clin Dermatol 1998;16:67-73.

    Kang S, Barnhill RL, Mihm MC Jr, Sober AJ. Multiple primary cutaneous melanomas. Cancer 1992;70:1911-1916.

    Goggins WB, Tsao H. A population-based analysis of risk factors for a second primary cutaneous melanoma among melanoma survivors. Cancer 2003;97:639-643.

    Clark WH Jr, Reimer RR, Greene M, Ainsworth AM, Mastrangelo MJ. Origin of familial malignant melanomas from heritable melanocytic lesions: the `B-K mole syndrome.' Arch Dermatol 1978;114:732-738.

    Lynch HT, Frichot BC III, Lynch JF. Familial atypical multiple mole-melanoma syndrome. J Med Genet 1978;15:352-356.

    NIH Consensus Conference: diagnosis and treatment of early melanoma. JAMA 1992;268:1314-1319.

    Kraemer KH, Tucker M, Tarone R, Elder DE, Clark WH Jr. Risk of cutaneous melanoma in dysplastic nevus syndrome types A and B. N Engl J Med 1986;315:1615-1616.

    Greene MH, Clark WH Jr, Tucker MA, Kraemer KH, Elder DE, Fraser MC. High risk of malignant melanoma in melanoma-prone families with dysplastic nevi. Ann Intern Med 1985;102:458-465.

    Bishop DT, Demenais F, Goldstein AM, et al. Geographical variation in the penetrance of CDKN2A mutations for melanoma. J Natl Cancer Inst 2002;94:894-903.

    Katris P, Crock JG, Gray BN. Research note: the Lions Cancer Institute and the Western Australian Society of Plastic Surgeons skin cancer screening programme. Aust N Z J Surg 1996;66:101-104.

    Rampen FH, van Huystee BE, Kiemeney LA. Melanoma/skin cancer screening clinics: experiences in the Netherlands. J Am Acad Dermatol 1991;25:776-777.

    Koh HK, Norton LA, Geller AC, et al. Evaluation of the American Academy of Dermatology's national skin cancer early detection and screening program. J Am Acad Dermatol 1996;34:971-978.

    Epstein DS, Lange JR, Gruber SB, Mofid M, Koch SE. Is physician detection associated with thinner melanomas? JAMA 1999;281:640-643.

    Balch CM, Buzaid AC, Soong SJ, et al. Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 2001;19:3635-3648.

    Gershenwald JE, Thompson W, Mansfield PF, 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;17:976-983.

    Cascinelli N. Margin of resection in the management of primary melanoma. Semin Surg Oncol 1998;14:272-275.

    Veronesi U, Cascinelli N, Adamus J, et al. Thin stage I primary cutaneous malignant melanoma: comparison of excision with margins of 1 or 3 cm. N Engl J Med 1988;318:1159-1162.

    Khayat D, Rixe O, Martin G, et al. Surgical margins in cutaneous melanoma (2 cm versus 5 cm for lesions measuring less than 2.1-mm thick). Cancer 2003;97:1941-1946.

    Cohn-Cedermark G, Rutqvist LE, Andersson R, et al. Long term results of a randomized study by the Swedish Melanoma Study Group on 2-cm versus 5-cm resection margins for patients with cutaneous melanoma with a tumor thickness of 0.8-2.0 mm. Cancer 2000;89:1495-1501.

    Balch CM, Soong SJ, Smith T, 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;8:101-108.

    Thomas JM, Newton-Bishop J, A'Hern R, et al. Excision margins in high-risk malignant melanoma. N Engl J Med 2004;350:757-766.

    Heaton KM, Sussman JJ, Gershenwald JE, et al. Surgical margins and prognostic factors in patients with thick (>4 mm) primary melanoma. Ann Surg Oncol 1998;5:322-328.

    Ross MI, Balch C, Cascinelli N, Edwards MJ. Excision of primary melanoma. In: Balch C, Houghton A, Sober A, Soong S, eds. Cutaneous melanoma. 4th ed. St. Louis: Quality Medical Publishing, 2003:209-30.

    Terhune MH, Swanson N, Johnson TM. Use of chest radiography in the initial evaluation of patients with localized melanoma. Arch Dermatol 1998;134:569-572.

    Tsao H, Feldman M, Fullerton JE, Sober AJ, Rosenthal D, Goggins W. Early detection of asymptomatic pulmonary melanoma metastases by routine chest radiographs is not associated with improved survival. Arch Dermatol 2004;140:67-70.

    Veronesi U, Adamus J, Bandiera DC, et al. Inefficacy of immediate node dissection in stage I melanoma of the limbs. N Engl J Med 1977;297:627-630.

    Cascinelli N, Morabito A, Santinami M, MacKie RM, Belli F. Immediate or delayed dissection of regional nodes in patients with melanoma of the trunk: a randomised trial: WHO Melanoma Programme. Lancet 1998;351:793-796.

    Sim FH, Taylor WF, Pritchard DJ, Soule EH. Lymphadenectomy in the management of stage I malignant melanoma: a prospective randomized study. Mayo Clin Proc 1986;61:697-705.

    Balch CM, Soong S, Ross MI, 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.

    Morton DL, Wen D-R, Foshag LJ, Essner R, Cochran A. Intraoperative lymphatic mapping and selective cervical lymphadenectomy for early-stage melanomas of the head and neck. J Clin Oncol 1993;11:1751-1756.

    Thompson JF, Uren RF, Coventry BJ, Chatterton BE. Lymphoscintigraphy. In: Balch C, Houghton A, Sober A, Soong S, eds. Cutaneous melanoma. 4th ed. St. Louis: Quality Medical Publishing, 2003:329-52.

    Reintgen D, Thompson JF, Gershenwald JE. Intraoperative mapping and sentinel node technology. In: Balch C, Houghton A, Sober A, Soong S, eds. Cutaneous melanoma. 4th ed. St. Louis: Quality Medical Publishing, 2003:353-77.

    Yu LL, Flotte TJ, Tanabe KK, et al. Detection of microscopic melanoma metastases in sentinel lymph nodes. Cancer 1999;86:617-627.

    Carson KF, Wen DR, Li PX, et al. Nodal nevi and cutaneous melanomas. Am J Surg Pathol 1996;20:834-840.

    Shivers SC, Wang X, Li W, et al. Molecular staging of malignant melanoma: correlation with clinical outcome. JAMA 1998;280:1410-1415.

    Carlson GW, Murray DR, Lyles RH, Staley CA, Hestley A, Cohen C. The amount of metastatic melanoma in a sentinel lymph node: does it have prognostic significance? Ann Surg Oncol 2003;10:575-581.

    Cochran AJ, Morton DL, Stern S, Lana AM, Essner R, Wen DR. Sentinel lymph nodes show profound downregulation of antigen-presenting cells of the paracortex: implications for tumor biology and treatment. Mod Pathol 2001;14:604-608.

    Lens MB, Dawes M, Goodacre T, Bishop JA. Excision margins in the treatment of primary cutaneous melanoma: a systematic review of randomized controlled trials comparing narrow vs wide excision. Arch Surg 2002;137:1101-1105.

    Bleicher RJ, Essner R, Foshag LJ, Wanek LA, Morton DL. Role of sentinel lymphadenectomy in thin invasive cutaneous melanomas. J Clin Oncol 2003;21:1326-1331.

    Jansen L, Nieweg OE, Peterse JL, Hoefnagel CA, Olmos RA, Kroon BB. Reliability of sentinel lymph node biopsy for staging melanoma. Br J Surg 2000;87:484-489.

    Essner R, Chung MH, Bleicher R, Hsueh E, Wanek L, Morton DL. Prognostic implications of thick (> or = 4-mm) melanoma in the era of intraoperative lymphatic mapping and sentinel lymphadenectomy. Ann Surg Oncol 2002;9:754-761.

    Carlson GW, Murray DR, Hestley A, Staley CA, Lyles RH, Cohen C. Sentinel lymph node mapping for thick (> or = 4-mm) melanoma: should we be doing it? Ann Surg Oncol 2003;10:408-415.

    Vuylsteke RJ, van Leeuwen PA, Statius Muller MG, Gietema HA, Kragt DR, Meijer S. Clinical outcome of stage I/II melanoma patients after selective sentinel lymph node dissection: long-term follow-up results. J Clin Oncol 2003;21:1057-1065.

    Perrott RE, Glass LF, Reintgen DS, Fenske NA. Reassessing the role of lymphatic mapping and sentinel lymphadenectomy in the management of cutaneous malignant melanoma. J Am Acad Dermatol 2003;49:567-592.

    Wrightson WR, Wong SL, Edwards MJ, et al. Complications associated with sentinel lymph node biopsy for melanoma. Ann Surg Oncol 2003;10:676-680.

    Santinami M, Maurichi A, Patuzzo R, Pennacchioli E, Cascinelli N. Impact of clinical trials on the treatment of melanoma. Surg Oncol Clin N Am 2001;10:935-947.

    Kirkwood JM, Strawderman MH, Ernstoff MC, Smith TJ, Borden EC, Blum RH. Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group trial EST 1684. J Clin Oncol 1996;14:7-17.

    Kirkwood JM, Ibrahim JG, Sondak VK, et al. High- and low-dose interferon alfa-2b in high-risk melanoma: first analysis of intergroup trial E1690/S9111/C9190. J Clin Oncol 2000;18:2444-2458.

    Kirkwood JM, Ibrahim JG, Sosman JA, et al. High-dose interferon alfa-2b significantly 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;19:2370-2380.

    Dubois RW, Swetter SM, Atkins M, et al. Developing indications for the use of sentinel lymph node biopsy and adjuvant high-dose interferon alfa-2b in melanoma. Arch Dermatol 2001;137:1217-1224.

    Agarwala SS, Kirkwood JM. Interferons in melanoma. Curr Opin Oncol 1996;8:167-174.

    Kirkwood JM, Bender C, Agarwala S, et al. Mechanisms and management of toxicities associated with high-dose interferon alfa-2b therapy. J Clin Oncol 2002;20:3703-3718.

    Musselman DL, Lawson DH, Gumnick JF, et al. Paroxetine for the prevention of depression induced by high-dose interferon alfa. N Engl J Med 2001;344:961-966.

    Cole BF, Gelber RD, Kirkwood JM, Goldhirsch A, Barylak E, Borden E. Quality-of-life-adjusted survival analysis of interferon alfa-2b adjuvant treatment of high-risk resected cutaneous melanoma: an Eastern Cooperative Oncology Group study. J Clin Oncol 1996;14:2666-2673.

    Hillner BE, Kirkwood JM, Atkins MB, Johnson ER, Smith TJ. Economic analysis of adjuvant interferon alfa-2b in high-risk melanoma based on projections from Eastern Cooperative Oncology Group 1684. J Clin Oncol 1997;15:2351-2358.

    Kilbridge KL, Weeks JC, Sober AJ, et al. Patient preferences for adjuvant interferon alfa-2b treatment. J Clin Oncol 2001;19:812-823.

    Eggermont AM. The role interferon-alpha in malignant melanoma remains to be defined. Eur J Cancer 2001;37:2147-2153.

    Kirkwood JM, Manola J, Ibrahim J, Sondak V, Ernstoff MS, Rao U. A pooled analysis of Eastern Cooperative Oncology Group and intergroup trials of adjuvant high-dose interferon for melanoma. Clin Cancer Res 2004;10:1670-1677.

    Lens MB, Dawes M. Interferon alfa therapy for malignant melanoma: a systematic review of randomized controlled trials. J Clin Oncol 2002;20:1818-1825.

    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 randomised trial. Lancet 2001;358:866-869.

    Hancock BW, Wheatley K, Harris S, et al. Adjuvant interferon in high-risk melanoma: the AIM HIGH Study -- United Kingdom Coordinating Committee on Cancer Research randomized study of adjuvant low-dose extended-duration interferon alfa-2a in high-risk resected malignant melanoma. J Clin Oncol 2004;22:53-61.

    Grob JJ, Dreno B, de la Salmoniere P, et al. Randomised trial of interferon alpha-2a as adjuvant therapy in resected primary melanoma thicker than 1.5 mm without clinically detectable node metastases. Lancet 1998;351:1905-1910.

    Pehamberger H, Soyer HP, Steiner A, et al. Adjuvant interferon alfa-2a treatment in resected primary stage II cutaneous melanoma. J Clin Oncol 1998;16:1425-1429.

    McMasters KM. The Sunbelt Melanoma Trial. Ann Surg Oncol 2001;8:Suppl:41S-43S.

    Mitchell MS. Perspective on allogeneic melanoma lysates in active specific immunotherapy. Semin Oncol 1998;25:623-635.

    Mitchell MS, Abrams J, Kashani-Sabet M, Thompson J. Interim analysis of a phase III stratified randomized trial of Melacine + low-dose intron-A versus high-dose intron-A for resected stage III melanoma. Prog Proc Am Soc Clin Oncol 2003;22:709. abstract.

    Livingston PO, Wong GY, Adluri S, et al. Improved survival in stage III melanoma patients with GM2 antibodies: a randomized trial of adjuvant vaccination with GM2 ganglioside. J Clin Oncol 1994;12:1036-1044.

    Bystryn JC, Zeleniuch-Jacquotte A, Oratz R, Shapiro RL, Harris MN, Roses DF. Double-blind trial of a polyvalent, shed-antigen, melanoma vaccine. Clin Cancer Res 2001;7:1882-1887.

    Hsueh EC, Gupta RK, Qi K, Morton DL. Correlation of specific immune responses with survival in melanoma patients with distant metastases receiving polyvalent melanoma cell vaccine. J Clin Oncol 1998;16:2913-2920.

    Berd D, Maguire HC Jr, Schuchter LM, et al. Autologous hapten-modified melanoma vaccine as postsurgical adjuvant treatment after resection of nodal metastases. J Clin Oncol 1997;15:2359-2370.

    Sosman JA, Unger JM, Liu PY, et al. Adjuvant immunotherapy of resected, intermediate-thickness, node-negative melanoma with an allogeneic tumor vaccine: impact of HLA class I antigen expression on outcome. J Clin Oncol 2002;20:2067-2075.

    Kawakami Y, Eliyahu S, Jennings C, et al. Recognition of multiple epitopes in the human melanoma antigen gp100 by tumor-infiltrating T lymphocytes associated with in vivo tumor regression. J Immunol 1995;154:3961-3968.

    Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med 1998;4:328-332.

    Weber JS, Snively J, Sian S, et al. Phase I trial of antibody to CTLA-4 (MDX-010) with melanoma peptides/Ifa for resected stages III/IV melanoma. Prog Proc Am Soc Clin Oncol 2003;22:710. abstract.

    Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med 1998;4:321-327.

    Kirkwood JM, Lee S, Land S, et al. E1696: phase II trial of multi-epitope peptide vaccination for melanoma with MGT (MART-1 (27-35), gp100 (209-217, 210M) and tyrosinase (368-376, 370D)) +/– IFN alfa-2b and GM-CSF — immunological and clinical results. Prog Proc Am Soc Clin Oncol 2003;22:709. abstract.

    Atkins MB, Buzaid AC, Houghton AN Jr. Chemotherapy and biochemotherapy. In: Balch C, Houghton A, Sober A, Soong S, eds. Cutaneous melanoma. 4th ed. St. Louis: Quality Medical Publishing, 2003:589-604.

    Morton DL, Essner R, Balch C. Surgical excision of distant metastases. In: Balch C, Houghton A, Sober A, Soong S, eds. Cutaneous melanoma. 4th ed. St. Louis: Quality Medical Publishing, 2003:547-72.

    Manola J, Atkins M, Ibrahim J, Kirkwood J. Prognostic factors in metastatic melanoma: a pooled analysis of Eastern Cooperative Oncology Group trials. J Clin Oncol 2000;18:3782-3793.

    Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999;17:2105-2116.

    Margolin K, Atkins M, Sparano J, et al. Prospective randomized trial of lisofylline for the prevention of toxicities of high-dose interleukin 2 therapy in advanced renal cancer and malignant melanoma. Clin Cancer Res 1997;3:565-572.

    Atkins MB, Shet A, Sosman JA. Interleukin-2: clinical applications: melanoma. In: DeVita VT, Hellman S, Rosenberg SA, eds. Principles and practice of the biologic therapy of cancer. Philadelphia: Lippincott Williams & Wilkins, 2000:50-73.

    Agarwala SS, Glaspy J, O'Day SJ, et al. Results from a randomized phase III study comparing combined treatment with histamine dihydrochloride plus interleukin-2 versus interleukin-2 alone in patients with metastatic melanoma. J Clin Oncol 2002;20:125-133.

    Gollob JA, Veenstra KG, Parker RA, et al. Phase I trial of concurrent twice-weekly recombinant human interleukin-12 plus low-dose IL-2 in patients with melanoma or renal cell carcinoma. J Clin Oncol 2003;21:2564-2573.

    Gollob J, Flaherty L, Smith J, et al. A Cytokine Working Group (CWG) phase II trial of a modified gp100 melanoma peptide (gp100 (209M)) and high dose interleukin-2 (HD IL-2) administered q3 weeks in patients with stage IV melanoma: limited antitumor activity. Prog Proc Am Soc Clin Oncol 2001;20:357a. abstract.

    Anderson CM, Buzaid AC, Legha SS. Systemic treatments for advanced cutaneous melanoma. Oncology (Huntingt) 1995;9:1149-1158.

    Crosby T, Fish R, Coles B, Mason MD. Systemic treatments for metastatic cutaneous melanoma. Cochrane Database Syst Rev 2000;2:CD001215-CD001215.

    Middleton MR, Grob JJ, Aaronson N, et al. Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol 2000;18:158-166.

    Summers Y, Middleton MR, Calvert H, et al. Effect of temozolomide (TMZ) on central nervous system (CNS) relapse in patients with advanced melanoma. Prog Proc Am Soc Clin Oncol 1999;18:531a. abstract.

    Atkins MB, Gollob JA, Sosman JA, et al. A phase II pilot trial of concurrent biochemotherapy with cisplatin, vinblastine, temozolomide, interleukin 2, and IFN-alpha 2B in patients with metastatic melanoma. Clin Cancer Res 2002;8:3075-3081.

    Hwu WJ, Krown SE, Menell JH, et al. Phase II study of temozolomide plus thalidomide for the treatment of metastatic melanoma. J Clin Oncol 2003;21:3351-3356.

    Keilholz U, Conradt C, Legha SS, et al. Results of interleukin-2-based treatment in advanced melanoma: a case record-based analysis of 631 patients. J Clin Oncol 1998;16:2921-2929.

    Allen IE, Kupelnick B, Kumashiro M, Luo D, Ross SD, Wolin MJ. Efficacy of interleukin-2 in the treatment of metastatic melanoma. Cancer Ther 1998;1:168-73.

    Eton O, Legha SS, Bedikian AY, et al. Sequential biochemotherapy versus chemotherapy for metastatic melanoma: results from a phase III randomized trial. J Clin Oncol 2002;20:2045-2052.

    Atkins MB, Lee S, Flaherty LE, Sosman JA, Sondak VK, Kirkwood JM. A prospective randomized phase III trial of concurrent biochemotherapy (BCT) with cisplatin, vinblastine, dacarbazine (CVD), IL-2 and interferon alpha-2b (IFN) versus CVD alone in patients with metastatic melanoma (E3695): an ECOG-coordinated intergroup trial. Prog Proc Am Soc Clin Oncol 2003;22:708. abstract.

    Keilholz U, Punt CJ, Gore M, et al. Dacarbazine, cisplatin and IFN-2b with or without IL-2 in advanced melanoma: final analysis of EORTC randomized phase III trial 18951. Prog Proc Am Soc Clin Oncol 2003;22:708. abstract.

    Keilholz U, Goey SH, Punt CJ, et al. Interferon alfa-2a and interleukin-2 with or without cisplatin in metastatic melanoma: a randomized trial of the European Organization for Research and Treatment of Cancer Melanoma Cooperative Group. J Clin Oncol 1997;15:2579-2588.

    Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Prospective randomized trial of the treatment of patients with metastatic melanoma using chemotherapy with cisplatin, dacarbazine, and tamoxifen alone or in combination with interleukin-2 and interferon alfa-2b. J Clin Oncol 1999;17:968-975.

    Keilholz U, Weber J, Finke JH, et al. Immunologic monitoring of cancer vaccine therapy: results of a workshop sponsored by the Society for Biological Therapy. J Immunother 2002;25:97-138.

    Parmiani G, Rivoltini L, Belli F, et al. Vaccination of metastatic melanoma patients with the autologous heat-shock protein peptide complex-96 (HSPPC-96: Oncophage) which contains melanoma peptides, results in a specific T-cell response and clinical response. Prog Proc Am Soc Clin Oncol 2002;21:13a. abstract.

    Bergen M, Chen R, Gonzalez R. Efficacy and safety of HLA-B7/beta-2 microglobulin plasmid DNA/lipid complex (Allovectin-7) in patients with metastatic melanoma. Expert Opin Biol Ther 2003;3:377-384.

    Soiffer R, Hodi FS, Haluska F, et al. Vaccination with irradiated, autologous melanoma cells engineered to secrete granulocyte-macrophage colony-stimulating factor by adenoviral-mediated gene transfer augments antitumor immunity in patients with metastatic melanoma. J Clin Oncol 2003;21:3343-3350.

    Dudley ME, Wunderlich JR, Robbins PF, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 2002;298:850-854.

    Tsao H, Millman P, Linette GP, et al. Hypopigmentation associated with an adenovirus-mediated gp100/MART-1-transduced dendritic cell vaccine for metastatic melanoma. Arch Dermatol 2002;138:799-802.

    Soengas MS, Capodieci P, Polsky D, et al. Inactivation of the apoptosis effector Apaf-1 in malignant melanoma. Nature 2001;409:207-211.

    Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature 2002;417:949-954.

    Millward MJ, Bedikian AY, Conry RM, et al. Randomized multinational phase 3 trial of dacarbazine (DTIC) with or without Bcl-2 antisense (oblimersen sodium) in patients (pts) with advanced malignant melanoma (MM): analysis of long-term survival. abstract. (Available at http://www.asco.org/ac/1,1003,_12-002636-00_18-0026-00_19-003620,00.asp.)

    Flaherty KT, Brose M, Schuchter L, et al. Phase I/II trial of BAY 43-9006, carboplatin (C) and paclitaxel (P) demonstrates preliminary antitumor activity in the expansion cohort of patients with metatastic melanoma. abstract. (Available at http://www.asco.org/ac/1,1003,_12-002636-00_18-0026-00_19-004164,00.asp.)

    Related Letters:

    Management of Cutaneous Melanoma

    de Giorgi V., Carli P., Giannotti B., Sampsel J. W., Barbera-Guillem E., Miranda E. P., Tsao H., Atkins M. B., Sober A. J.(Hensin Tsao, M.D., Ph.D.,)