Newer
Older
indexation / test / dataset / in / resources / corpus / Clean_0C1C12E1156F7025DBB91BEDD4B8D1E7BB81C36A.txt
@kieffer kieffer on 27 Feb 2017 43 KB v0
annoncannoncAnnals of Oncology1569-80410923-7534Oxford University Press10.1093/annonc/mdl050reviewsMerkel cell cancer of the skinPectasidesD*PectasidesMEconomopoulosTSecond Department of Internal Medicine, Oncology Section, University of Athens, ‘Attikon’ University Hospital, Haidari, 1 Rimini, Athens, Greece*Correspondence to: Dr D. Pectasides, Second Department of Internal Medicine, Propaedeutic, Oncology Section, Attikon University Hospital, Rimini 1, Haidari, Athens, Greece. Tel: +3210-5831691; Fax: +3210-5831690; E-mail: pectasid@otenet.gr102006243200617101489149525920051220061022006© 2006 European Society for Medical Oncology2006Merkel cell carcinoma (MCC) is a rare malignant cutaneous tumor of the elderly with rapidly growing skin nodules found predominantly on sun-exposed areas of the body. The vast majority of patients present with localized disease, while up to 30% have regional lymph node metastases. Despite local excision and the incidence of local recurrence, regional lymph node metastases and distant metastases is high and usually occurs within 2 years of primary diagnosis. The optimal treatment for patients with MCC remains unclear. The best outcome is achieved with multidisciplinary management including surgical excision of primary tumor with adequate margins and post-operative radiotherapy (RT) to control local and regional disease. Patients with regional nodal metastases should be treated with lymph node dissection plus RT. Adjuvant chemotherapy (CT) should be considered as part of the initial management. In case of metastatic disease CT based on regimens used for small-cell lung cancer is the standard treatment of care.chemotherapyMerkel cell carcinomaprognosisradiotherapysurgeryintroductionMerkel cell carcinoma (MCC) is a rare malignant cutaneous tumor of the elderly that is characterized by an aggressive course with regional nodal involvement, distant metastases and a high rate of recurrence. MCC was first described by Toker in 1972 [1] as trabecular carcinoma of the skin and, since then, has also been considered as cutaneous APUDoma, neuroendocrine tumor of the skin, Merkel cell tumor, primary small cell carcinoma of the skin, primary undifferentiated carcinoma of the skin, anaplastic carcinoma of the skin and murky cell carcinoma [2]. MCC and neuroendocrine carcinoma of the skin are the most widely used terms and may best reflect postulated origin and immunocytologic characteristics of this neoplasm.epidemiologyEvery year in the United States, there are approximately 470 new recorded cases. The estimated annual incidence according to US Surveillance data, Epidemiology, and the End-Results Program is 0.23 per 100 000 people for whites [3]. From 1987 to 2001, the Finnish Cancer Registry recorded 141 cases of MCC in a population of 5 000 000, which gives an annual prevalence of 0.2 cases per 100 000 [4]. The incidence in blacks (0.01 cases per 100 000) as well as for Polynesians seems to be lower. Both sexes are affected, but there seems to be a male predominance (2.3:1) [3, 5–7]. In some series the incidence in women was reported higher than that in men [8, 9]. MCC occurs in elderly patients, with an average age of diagnosis at 69 years (range 7–104 years). Only less than 5% of cases occur before the age of 50 years [7, 9–13].clinical manifestation and diagnosisMost patients present with rapidly growing, painless, firm, non-tender, dome-shaped red, occasionally ulcerated skin nodules, which have a red or bluish color, measuring up to several centimeters, on predominantly sun-exposed areas of the body [11]. The overlying skin is smooth and shiny, sometimes exhibiting ulcerative, acneiform or telangiectatic features [14]. However, the clinical feature of Merkel cell tumor is typically not acneiform. MCC can spread through the dermal lymphatic system, resulting in the development of multiple satellite lesions. MCC involves predominantly the head and neck (>50% of cases) [9, 15]; it involves the extremities in 40% of cases and the trunk in less than 10% of cases [2, 16–20]. On the face, the eyelids are involved more frequently [21]. MCC involving other sites not exposed to sunlight has been reported in a small percentage of cases [22–24]. At presentation most patients with MCC present with localized disease (70%–80%), 9%–26% have regional lymph node metastases and 1%–4% distant metastases [11, 18]. Despite local excision, the incidence of local recurrence (27%–60%), regional lymph node metastases (45%–91%) and distant metastases (18%–52%) is high and usually occurs within 2 years of primary diagnosis [11, 25, 26]. The majority of patients die from distant metastases involving liver, bone, lung, brain or distant lymph nodes [11, 26, 27]. Because the early course of MCC is asymptomatic, diagnosis may be delayed until the detection of regional lymphadenopathy and distant metastases. Symptoms are related to local spread or to involvement of regional lymph nodes. Superior vena cava syndrome due to obstruction by tumor has been reported [28]. Spontaneous regression of the primary lesion has been reported [29] and 20%–30% of patients present without obvious primary lesion. Patients who present with no obvious primary tumor seem to have a better prognosis [30, 31]. The differential diagnosis of MCC includes basal cell carcinoma, lymphoma, melanoma and squamous cell carcinoma, with MCC usually being overlooked [14]. The correct diagnosis of MCC is not made until tissue sampling and pathologic evaluation are performed.staging and prognosisThere is no universal accepted staging system for MCC, but tentative classification relies on clinical manifestation and includes: stage I, localized cutaneous involvement (IA: ≤2 cm, IB: >2 cm); stage II, regional nodal involvement; stage III, systemic metastases [7, 32]. Even though the diagnosis of MCC is exclusively pathologic, imaging is useful for staging, surgical guidance, therapeutic management and follow-up. This staging system allows therapeutic approaches according to the stage: stage I and II should be treated with curative intent and stage III as a palliative treatment.The most important prognostic factor for survival and development of distant metastases is the presence of lymph node involvement [6, 33]. In the series of Poulsen et al. [30], most important predictors for local control and survival were the tumor site and the presence of regional lymph node metastases. Morrison et al. [34] reported that the median survival of patients with lymph node metastases was 13 months and for those without lymph node involvement it was 40 months. Other reported poor prognostic factors include location on the lower extremities [31] because of difficulties in local control and the high incidence of local failure due to the poor blood supply in the legs, which limits big surgical procedures, and the subsequent poor tolerability of radiotherapy (RT) by the elderly. Furthermore, they include tumor size ≥2cm [4, 35], male sex [6, 9, 11, 18], age ≥60 years [7], no treatment with radiotherapy [36] and positive surgical margins [37].etiologyRisk factors include exposure to sunlight and ultraviolet (UV) light. MCC seems to have a relationship with sun exposure, both by its anatomic predilection and geographic distribution. Miller and Rabkin reported a correlation of the UV B index and the incidence of MCC [3]. An association has been reported between MCC and other UV light-related skin cancers (squamous cell carcinoma, basal cell carcinoma) previous irradiation and exposure to radiant infrared heat [10, 38–43]. The development of MCC in non-sun-exposed regions of the body suggests that factors other than sunlight have been implicated in the pathogenesis of MCC. Lunder and Stern [44] reported a 100-fold increased incidence of MCC in patients treated with methoxsalen and UVA for psoriasis. Exposure to arsenic has also been implicated [45]. Immunocompromise and immunosuppression may represent additional risk factors for MCC, whose prevalence is unusually high in organ recipients compared with the general population [46, 47]. The risk of MCC in renal recipients has been estimated at 0.13/1000 person-years [48]. A high prevalence of second neoplasm occurring before, concurrent with or after the diagnosis of MCC (25%–28%) has recently been reported. These neoplasms include squamous cell carcinoma, hematological malignancies, breast cancer and ovarian cancer [43]. MCC has also been reported in patients with HIV infection and in those with chronic lymphocytic leukemia, suggesting that these diseases have an association with MCC. Engels et al. [49] reported that the relative risk of developing MCC in patients with HIV infection is approximately 13.4.pathologyMCC involves the dermis and frequently extends into the subcutaneous fat. Sometimes, MCC involves the overlying epidermis [50]. The tumor consists of small blue, ovoid cells with hyper-chromatic nuclei, a very fine chromatine and minimal cytoplasm. Mitoses are frequent and the apoptotic index high. The triad of vesicular nuclei with small nucleoli, abundant mitosis and apoptosis is suggestive of MCC.The most widely accepted origin of MCC is the Merkel cell [51], whose function is not clear. This clear, oval cell is located within or close to the basal layer of the epidermis and functions as a cutaneous mechanoreceptor [52]. The ultrastructural and immunohistochemical similarities between MCC and Merkel cells support the Merkel cell origin of MCC [53–55]. However, there is evidence that runs counter to this postulate: MCC and Merkel cells have different location (dermis and epidermis, respectively) and MCC does not express vasoactive intestinal peptides and metenkephalin specific to Merkel cells [56]. An intermediate consideration suggests that the origin of MCC is associated with an immature, totipotential stem cell that acquires neuroendocrine characteristics during malignant transformation [56, 57]. The same concept of precursor stem cells may explain the coexistence of MCC and squamous cell carcinoma of the skin [38].Although MCC can usually be diagnosed histologically rather easily, it sometimes may be difficult using light microscopic findings due to its similarities with other small cell tumors. These include metastatic oat cell carcinoma, metastatic carcinoid tumor, neuroblastoma, and some types of melanoma, lymphoma and squamous cell carcinoma [11, 57]. MCC can be definitively diagnosed with hematoxylin–eosin and immunohistochemical staining, electron microscopy or both [15, 25]. Immunohistochemical staining can distinguish MCC from these tumors. MCC express both neuroendocrine (neuron-specific enolase, synaptophysin, chromogranin) and cytokeratin markers (cytokeratin 20, as a paranuclear dot, CAM 5.2) and is negative for S100 and common leukocyte antigen. Epithelial membrane antigen and BEP-EP4 may also be expressed in MCC. Cytokeratin 7, which identifies bronchial small-cell carcinoma, is negative in MCC. In addition, a homeodomain-containing nuclear transcription factor expresses in bronchial small-cell carcinoma and is negative in MCC [58, 59]. On the contrary, neurofilament protein is commonly expressed in MCC and often not in bronchial small-cell carcinoma [60, 61]. CD 117 (KIT receptor) is also present in 95% of MCC, but its expression does not correlate with more aggressive tumor [62].Three histological subtypes of MCC have been reported: intermediate, small cell and trabecular, but these variants have no clinical relevance [63]. Intermediate variant is the most common seen subtype; the small-cell variant is histologically similar to other small-cell carcinomas and should be distinguished from the bronchial small-cell carcinomas.molecular biology of MCCNumerous chromosomal abnormalities have been reported in MCC, but the exact pathways of neuroendocrine differentiation are yet unclear. The most interesting chromosomal abnormality is the deletion of the short arm of chromosome 1 (1p36) [64, 65], which is also common in melanoma and neuroblastoma [66, 67]. These findings lead to arguments about the neurocrest origin of MCC [68]. Another abnormality is the loss of heterozygosity in chromosome 3p21, the same region that is also affected in small-cell lung cancer [69].Several other abnormalities have been reported, including trisomy 1, trisomy 6, trisomy 11, trisomy 18 and deletion of chromosome 7 [70–73]. Loss of heterozygosity has also been reported in chromosomes 10q and 13 [74, 75]. The tumor suppressor gene p53 is also occasionally present in MCC, and the expression of mutant p53 may be associated with poor outcome [76]. P73 has been localized in 1p36.33 and, as in melanoma, is expressed infrequently [77, 78].Recently, Leonard et al. [79] reported that MNF (Merkel nuclear factor) contains the POU-IV family member Brn-3c and that Brn-3c is expressed in normal Merkel cells. In addition, Brn-3c protein reactivity is restricted to a subset of MCC biopsies and is not seen in biopsies revealing adherent, variant cell lines lacking neuroendocrine markers. In mice POU-IV Brn-3 and the transcription factor ATOH1 are very important for the function of normal Merkel cells and for neuroendocrine differentiation [31]. The precise role of these abnormalities in the prognosis of MCC has to be determined.imagingAfter diagnosis, patients should be assessed by imaging techniques to define the exact extent of disease. Because MCC behaves like malignant melanoma, lymphoscintigraphy may be used to localize sentinel nodes, which may harbor micrometastases. Lymphoscintigraphy in combination with intra-operative lymphatic mapping allows accurate and selective sampling of sentinel nodes [80]. Distant metastases are present in some cases at presentation. The pattern of metastases is not specific for MCC and seems to mimic multifocal invasion by other small-cell carcinomas. Ultrasonography of soft tissue may show hypoechoic nodules arising from the dermis. CT scan may demonstrate regional lymph node involvement or systematic metastases in the lungs, liver, bones and other sites [81]. Invasion of CNS is rare but should not be overlooked. In such cases, it is best to perform MRI.Distant metastases and locoregional recurrence may be evaluated with somatostatin receptor scintigraphy (SRS) based on the neuroendocrine characteristics of MCC [80]. SRS is helpful in pre- and post-therapeutic evaluation of MCC. Compared with CT scan and MRI, SRS is less affected by inflammation, edema and tissues at the surgical and irradiated site [80, 82]. However, SRS may be limited in evaluating metastases in liver, kidneys and spleen. The presence of somatostatin receptors has also been used therapeutically in MCC [83]. Guitera-Rovel et al. [84] reported a sensitivity of 78% and a specificity of 96% in 20 MCC patients assessed by SRS.Positron emission tomography with the glucose analog 2-[fluorine-18] fluoro-2-deoxy-D-glucose (FDG) also plays an important role in assessing and monitoring MCC [85].treatmentTreatment options will be discussed according to the disease stages. Because of the rarity of MCC, there are no prospective trials investigating surgery, RT or chemotherapy (CT).stage IFor stage I disease the main treatment strategies include surgery and radiation therapy. In these cases the reported 5-year survival reached 64%. Controversies exist regarding the best excision margins in MCC, as there are no prospective, controlled studies dealing with this aspect. Margins 2–3 cm wide and 2 cm deep have been generally accepted [2, 11, 18, 32, 86–88]. In the older series [14, 32], the recommended margins were ≥3 cm. In the studies by O'Connor et al. [14] and by Yiengpruksawan et al. [32], margins ≥3 cm were associated with reduction in local recurrence. Updating results were reported based on 102 patients with MCC treated at Memorial Sloan Kettering Cancer Center [35]. After a median follow-up of 35 months, the overall 5-year disease-specific survival rate was 74%. Recurrence of disease occurred in 55 patients (55%), and the most common site of first recurrence was within the draining lymph nodes (n = 35). Elective lymph node dissection was the only independent predictor of improved relapse-free survival. However, in the study by Ott et al. [9] and in the study by Gillenwater et al. [89], 66 patients with MCC of the head and neck region, no difference in outcome (locoregional control and survival) was noted when the resection margins were >2 cm or <2 cm or the patients had an adequate RT or <1 cm, or 1–2 cm, or >2 cm, respectively. In contrast, in the latest study, a comparison of the patients who did (n = 26) and did not (n = 34) receive post-operative radiation therapy revealed a significant difference in local [3 (12%) versus 15 (44%), respectively; P <0.01] and regional [7 (27%) versus 29 (85%), respectively; P <0.01] recurrence rates. There was, however, no significant difference in the disease-specific survival between these groups (P = 0.30). Distant disease developed in 36% of all patients regardless of therapy [89].Mohs micrographic surgery has been considered as the best method of wide clearance. However, no controlled clinical trials comparing this method with the traditional wide excision have shown its benefit. O'Connor et al. [90] evaluated the use of Mohs micrographic surgery for this aggressive neoplasm. Standard surgical excision for local disease was associated with high rates of local persistence [13 of 41 (31.7%)] and regional metastasis [20 of 41 (48.8%)]. Only one of 12 (8.3%) Mohs-treated patients, with histologically confirmed clearance, had local persistence of disease. Regional metastasis developed in four of 12 cases (33.3%). Regional metastasis developed in none of the four patients treated with RT after Mohs surgery and in four of eight patients treated with Mohs surgery without post-operative RT. Boyer et al. [91] retrospectively conducted a study; the study group consisted of 45 patients with stage I MCC who were histologically and clinically free of disease after Mohs excision. Twenty patients subsequently received elective post-operative RT to the primary site and 25 patients had no adjuvant radiation therapy. One marginal recurrence (4%) and three in-transit metastases were observed in the Mohs surgery alone group, whereas none were observed in the Mohs surgery and RT group. The proportion of patients with these events was not significantly different between treatment groups. Overall survival, relapse-free survival and disease-free survival were not significantly different between treatment groups [91]. They concluded that adjuvant RT appears unessential to secure local control of primary MCC lesions completely excised with Mohs micrographic surgery.MCC is a radiosensitive malignancy and adjuvant RT has been advocated for local and regional disease control [6]. Because of the high rate of local or regional relapse, RT has been used post-operatively, in an adjuvant basis in patients with MCC. However, most of the studies argued for the benefits of post-surgery RT [6, 33, 92–97]. Allen et al. [35] found that local recurrence occurred in approximately 12% of patients and was not more common in those who did not receive adjuvant RT after resection of the primary lesion. Adjuvant RT should be considered in patients with primary disease in whom wide surgical margins are unattainable or in those found to have pathologically close or involved margins after excision. In contrast, Wilder et al. [97] reported in a review that the administration of RT post-operatively to both the surgical bed and the draining lymph nodes improves locoregional control and may result in long-term disease-free survival when administered after the initial surgical resection. Similarly, Shaw et al. [18] in a review of 30 reports of MCC reported that after excision alone of the primary lesion, local recurrence occurred in 39% of patients and regional failure occurred in 46%. In contrast, patients treated by excision plus prophylactic treatment (adjuvant node dissection and/or adjuvant RT), experienced local recurrence in 26% and regional failure in 22%. Locoregional recurrence carried an ominous significance with 67% of patients subsequently dying of the disease. For patients who either presented with regional disease or later developed regional disease, the best outcome (44% survival with mean follow-up of 40 months) was obtained following treatment by therapeutic node dissection with or without RT. On the contrary, treatment of regional disease with RT alone was associated with only a 20% survival rate. In addition, Ott et al. [9] treated 31 patients with MCC of the head and neck region. Therapy included local excision with or without RT in 19 patients, local resection and lymphadenectomy with or without RT in eight patients, and RT alone in four patients with head and neck tumors. They concluded that locoregional recurrence correlates with inadequate margins and lack of RT, but remission is possible with multimodality therapy. Similarly, Meeuwissen et al. [6] demonstrated the importance of surgery plus RT over surgery alone in preventing local recurrence of this highly malignant skin cancer. Medina-Franco et al. [98] reviewed 1024 cases with MCC and reported that adjuvant RT was associated with a reduced risk of local recurrence (P < 0.00001). RT has also been used alone and has been reported to achieve local control [92, 94, 99, 100]. The administered dose of RT ranges from 30 to 70 Gy, but the majority of patients received 45–50 Gy [31, 70].Patients with MCC have at presentation 10%–30% regional lymph node involvement [11] and micrometastatic disease in prophylactic lymph node dissection has been found in 100% of patients [31]. Because the incidence of regional lymph node metastases is extremely high, ranging from 45% to 91% at diagnosis [11, 25, 26], elective nodal surgery with RT should be considered in patients with stage I disease. Controversy regarding the management of clinically negative lymph nodes has been eliminated with the use of sentinel lymph node mapping (SLN) for patients with MCC [15, 101, 102]. Approximately 25% of patients with MCC have metastatic disease in the SLN [15, 35]. Prophylactic lymph node dissection is not recommended as standard management [63, 87]. However, some investigators have recommended the prophylactic use of lymph node dissection for tumors at high risk for recurrence [87]. If metastatic disease is proved in SLN, management includes complete lymphadenectomy plus post-operative RT. Early removal of microscopic disease in regional lymph nodes detected by SLN biopsy may afford the patient a greater opportunity for cure than expectant management of the regional nodal basin, although this remains unproven [103]. Unfortunately, over half of the patients who undergo regional lymphadenectomy for clinically positive nodes will die of distant metastases [18, 32]. Lymphoscintigraphy and sentinel node biopsy have been proposed to overcome the problem of unpredictable lymphatic drainage and to minimize the need for node dissection [104, 105]. A meta-analysis of case series of 60 patients with Merkel cell carcinoma managed with sentinel lymph node biopsy was performed by Mehrany et al. [101]. Forty of 60 patients (67%) had a biopsy-negative sentinel lymph node; 97% of this group had no recurrence at 7.3 months median follow-up. Twenty patients (33%) had a biopsy-positive sentinel lymph node; 33% of this group experienced local, regional or systemic recurrence at 12 months median follow-up. Risk of recurrence or metastasis was 19-fold greater in biopsy-positive patients (odds ratio, 18.9; P = 0.005). None of 15 biopsy-positive patients who underwent therapeutic lymph node dissection experienced a regional recurrence. They concluded that sentinel lymph node positivity is strongly predictive of a high short-term risk of recurrence or metastasis in patients with MCC. Therapeutic lymph node dissection appears effective in preventing short-term regional nodal recurrence. The role of adjuvant CT in stage I MCC remains experimental. However, aggressive adjuvant treatment should be considered for patients with positive sentinel lymph nodes [101].stage IIIf stage II disease has been documented, management should include simultaneous excision of the primary site, regional lymph node dissection and RT to the primary tumor and regional nodal site. Lymph node involvement is frequently bulky and the decision for operation should be based on the operability of the nodal mass, the patient's performance status and the site of lymph node involvement [31]. RT at a dose of 50–60 Gy should be used for the regional nodal mass.CT can be given in high-risk patients [106–109]. In a small series of 35 patients [86] no benefit from adjuvant CT was obtained. In contrast, Boyle et al. [7] reported a 40% (eight of 20) RR to CT, particularly CBDCA plus VP-16 given to patients with regional lymph node disease. They proposed that for such poor-prognosis tumors, further investigation of adjuvant RT and CT is warranted, as responsiveness of recurrent disease has been confirmed. In patients with recurrent or locally advanced disease and adequate performance status, Tai et al. [110] feel that chemoradiation (radiotherapy plus CAV-cyclophosphamide, doxorubicin or epirubicin, and vincristine or cisplatin and etoposide) may be considered as a treatment option.stage IIIDistant metastases are frequent at initial presentation [11, 32]. These lesions may be found incidentally at routine radiographic work-up or imagine evaluation for unrelated causes. The pattern of metastasis is not specific for MCC and mimics multifocal invasion by other small-cell carcinomas. However, organs most frequently affected are the liver, bone, lung, brain and skin. The prognosis of patients with stage III MCC is poor, with an average interval of 8 months between diagnosis and death [10]. Treatment consists of CT, palliative RT and possibly surgery. CT for MCC includes several regimens consisting of cyclophosphamide (CTX), doxorubicin (ADR), vincristine (VCR), 5-fluoruracil (5-FU), cisplatin (CDDP), carboplatin (CBDCA) and etoposide (VP-16) [10, 86, 106, 110, 111]. CT has only short-lived success in patients with MCC [10, 14].RT can be given for palliation of bone and brain metastases or for palliation of cutaneous deposits that are bleeding or fungating. CT is usually reserved for those patients with generalized disease or advanced local or regional disease. Several investigators have reported favorable responses using a variety of regimens [110, 111]. Complete response rates of 20%–35% (median duration 6 months) and partial response rates of 23%–50% have been reported in patients with stage III disease [86, 110–112]. Voog et al. [111] reviewed 37 reports on the use of CT for 107 patients with MCC. The reported response rate was 60% for patients with locally advanced disease and 57% for those with metastatic disease. Cisplatin, ADR and 5-FU were the agents most commonly associated with a significant response. Cyclophosphamide was utilized in 56% of cases, ADR in 49% of cases and platinum derivatives in 25% of cases. Tai et al. [110] confirmed these results, reporting RR in 68% of patients with locally advanced disease and 59% in those with metastatic disease. Cyclophosphamide, ADR or epirubicin (EPI) and VCR appeared to be the most active agents. The reported RR was 75.7% (CR 35.1%). The combination of CDDP or CBDCA plus VP-16 had a RR of 60% (CR 36%). These responses did not differ significantly (P = 0.19). In our study [112] the RR was 83% (CR 33%, PR 50%) for patients both with locally advanced and metastatic disease.conclusionsMCC is a rare and aggressive neuroendocrine tumor of the skin. The optimal treatment for patients with MCC remains unclear. The best outcome is achieved with multidisciplinary management. Optimal management of patients with MCC consists of optimal surgery of primary tumor with adequate margins and post-operative RT to control local and regional disease. SLN mapping is essential in patients with clinically negative lymph nodes because it provides important prognostic information and allows selection of patients for additional nodal therapy. Patients with localized disease should be treated with conservative surgery followed by post-operative RT. The dose of post-operative RT ranges from 45 Gy to 60 Gy in 25–30 fractions. Patients with resectable nodal disease should be treated with regional node dissection followed by post-operative RT. Patients with fixed, unresectable nodal metastases should be treated with pre-operative CT plus RT followed by a nodal dissection. Because MCC has a propensity to develop hematogenous metastases and the responses to CT are high, adjuvant CT should be considered as part of the initial treatment. CT based on regimens for small-cell lung cancer resulted in tumor regression in up to 70% of cases with metastatic disease.1.TokerCTrabecular carcinoma of the skinArch Dermatol19721051071102.HaagMLGlassLFFenskeNAMerkel cell carcinoma. Diagnosis and treatmentDermatol Surg1995216696833.MillerRWRabkinCSMerkel cell carcinoma and melanoma: etiological similarities and differencesCancer Epidemiol Biomarkers Prev199981531584.KoljonenVBohlingTGranhrothGTukiainenEMerkel cell carcinoma: a clinicopathological study of 34 patientsEur J Surg Oncol2003296076105.FeunLGSavarajNLeghaSSChemotherapy for metastatic Merkel cell carcinoma. Review of the M.D. Anderson Hospital's experienceCancer1988626836856.MeeuwissenJABourneRGKearsleyJHThe importance of postoperative radiation therapy in the treatment of Merkel cell carcinomaInt J Radiat Oncol Biol Phys1995313253317.BoyleFPendleburySBellDFurther insights into the natural history and management of primary cutaneous neuroendocrine (Merkel cell) carcinomaInt J Radiat Oncol Biol Phys1995313153238.Meyer-PannwittUKummerfeldtKBoubarisPCaselitzJMerkel cell tumor or neuroendocrine skin carcinomaLangenbecks Arch Chir19973823493589.OttMJTanabeKKGaddMAMultimodality management of Merkel cell carcinomaArch Surg199913438839310.SmithDFMessinaJLPerrottRClinical approach to neuroendocrine carcinoma of the skin (Merkel cell carcinoma)Cancer Control20007728311.HitchcockCLBlandKILaneyRGNeuroendocrine (Merkel cell) carcinoma of the skin. Its natural history, diagnosis, and treatmentAnn Surg19882072012073rd12.SchmidCBehamAFeichtingerJRecurrent and subsequently metastasizing Merkel cell carcinoma in a 7-year-old girlHistopathology19922043743913.SonakRATredeKGerharzCDMerkel cell tumor of the hand in a 104-year-old patient. Case report with review of the literatureHandchir Mikrochir Plast Chir199628434514.O'ConnorWJBrodlandDGMerkel cell carcinomaDermatol Surg19962226226715.MessinaJLReintgenDSCruseCWSelective lymphadenectomy in patients with Merkel cell (cutaneous neuroendocrine) carcinomaAnn Surg Oncol1997438939516.RaafJHUrmacherCKnapperWKTrabecular (Merkel cell) carcinoma of the skin. Treatment of primary, recurrent, and metastatic diseaseCancer19865717818217.GollardRWeberRKostyMPMerkel cell carcinoma: review of 22 cases with surgical, pathologic, and therapeutic considerationsCancer2000881842185118.ShawJHRumballEMerkel cell tumour: clinical behaviour and treatmentBr J Surg19917813814219.SavagePConstenlaDFisherCThe natural history and management of Merkel cell carcinoma of the skin: a review of 22 patients treated at the Royal Marsden HospitalClin Oncol (R Coll Radiol)1997916416720.SkeltonHGSmithKJHitchcockCLMerkel cell carcinoma: analysis of clinical, histologic, and immunohistologic features of 132 cases with relation to survivalJ Am Acad Dermatol19973773473921.SoltauJBSmithMECusterPLMerkel cell carcinoma of the eyelidAm J Ophthalmol199612133133222.TennvallJBiorklundAJohanssonLAkermanMMerkel cell carcinoma: management of primary, recurrent and metastatic disease. A clinicopathological study of 17 patientsEur J Surg Oncol1989151923.ChenKTMerkel's cell (neuroendocrine) carcinoma of the vulvaCancer1994732186219124.TomicSWarnerTFMessingEWildingGPenile Merkel cell carcinomaUrology1995451062106525.SibleyRKDahlDPrimary neuroendocrine (Merkel cell?) carcinoma of the skin. II. An immunocytochemical study of 21 casesAm J Surg Pathol1985910911626.MelandNBJacksonITMerkel cell tumor: diagnosis, prognosis, and managementPlast Reconstr Surg19867763263827.EckerHAJrAbtABGrahamWPHercegSTrabecular or Merkel-cell carcinoma of the skinPlast Reconstr Surg1982704854903rd28.RouthAHickmanBTJohnsonWWSuperior vena cava obstruction from Merkel cell carcinomaArch Dermatol198712371471629.SaisGAdmellaCSolerTSpontaneous regression in primary cutaneous neuroendocrine (Merkel cell) carcinoma: a rare immune phenomenon?J Eur Acad Dermatol Venereol200216828330.PoulsenMRischinDWalpoleETrans-Tasman Radiation Oncology Group. High-risk Merkel cell carcinoma of the skin treated with synchronous carboplatin/etoposide and radiation: a Trans-Tasman Radiation Oncology Group Study–TROG 96:07J Clin Oncol2003214371437631.PoulsenMMerkel-cell carcinoma of the skinLancet Oncol2004559359932.YiengpruksawanACoitDGThalerHTMerkel cell carcinoma: Prognosis and managementArch Surg19911261514151933.FenigEBrennerBKatzAThe role of radiation therapy and chemotherapy in the treatment of Merkel cell carcinomaCancer19978088188534.MorrisonWHPetersLJSilvaEGThe essential role of radiation therapy in securing locoregional control of Merkel cell carcinomaInt J Radiat Oncol Biol Phys19901958359135.AllenPJZhangZFCoitDGSurgical management of Merkel cell carcinomaAnn Surg19992299710536.EichHTEichDStaarSRole of postoperative radiotherapy in the management of Merkel cell carcinomaAm J Clin Oncol200225505637.CoitDGMerkel cell carcinomaAnn Surg Oncol20019Suppl99S102S38.IacoccaMVAbernethyJLStefanatoCMMixed Merkel cell carcinoma and squamous cell carcinoma of the skinJ Am Acad Dermatol19983988288739.HewittJBSherifAKerrKMStanklerLMerkel cell and squamous cell carcinomas arising in erythema ab igneBr J Dermatol199312859159240.CerroniLKerlHPrimary cutaneous neuroendocrine (Merkel cell) carcinoma in association with squamous- and basal-cell carcinomaAm J Dermatopathol19971961061341.JonesCSTyringSKLeePCFineJDDevelopment of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igneArch Dermatol198812411011342.GomezLGDiMaioSSilvaEGMackayBAssociation between neuroendocrine (Merkel cell) carcinoma and squamous carcinoma of the skinAm J Surg Pathol1983717117743.BrennerBSulkesARakowskyESecond neoplasms in patients with Merkel cell carcinomaCancer2001911358136244.LunderEJSternRSMerkel-cell carcinomas in patients treated with methoxsalen and ultraviolet A radiationN Engl J Med19983391247124845.TsurutaDHamadaTMochidaKMerkel cell carcinoma, Bowen's disease and chronic occupational arsenic poisoningBr J Dermatol199813929129446.PennIFirstMRMerkel's cell carcinoma in organ recipients: report of 41 casesTransplantation1999681717172147.BuellJFTrofeJHanawayMJImmunosuppression and Merkel cell cancerTransplant Proc2002341780178148.BordeaCWojnarowskaFMillardPRSkin cancers in renal-transplant recipients occur more frequently than previously recognized in a temperate climateTransplantation20047757457949.EngelsEAFrischMGoedertJJMerkel cell carcinoma and HIV infectionLancet200235949749850.SilvaEGMackayBGoepfertHEndocrine carcinoma of the skin (Merkel cell carcinoma)Pathol Annu19841913051.TangCKTokerCTrabecular carcinoma of the skin: an ultrastructural studyCancer1978422311232152.WinkelmannRKThe Merkel cell system and a comparison between it and the neurosecretory or APUD cell systemJ Invest Dermatol197769414653.WarnerTFUnoHHafezGRMerkel cells and Merkel cell tumors. Ultrastructure, immunocytochemistry and review of the literatureCancer19835223824554.GuJPolakJMTapiaFJNeuron-specific enolase in the Merkel cells of mammalian skin. The use of specific antibody as a simple and reliable histologic markerAm J Pathol1981104636855.WilsonBSLloydRVDetection of chromogranin in neuroendocrine cells with a monoclonal antibodyAm J Pathol198411545846856.HoeflerHKerlHRauchHJDenkHNew immunocytochemical observations with diagnostic significance in cutaneous neuroendocrine carcinomaAm J Dermatopathol1984652553057.FrigerioBCapellaCEusebiVMerkel cell carcinoma of the skin: the structure and origin of normal Merkel cellsHistopathology1983722924958.AgoffSNLampsLWPhilipATThyroid transcription factor-1 is expressed in extrapulmonary small cell carcinomas but not in other extrapulmonary neuroendocrine tumorsMod Pathol20001323824259.CheukWKwanMYSusterSChanJKImmunostaining for thyroid transcription factor 1 and cytokeratin 20 aids the distinction of small cell carcinoma from Merkel cell carcinoma, but not pulmonary from extrapulmonary small cell carcinomasArch Pathol Lab Med200112522823160.SchmidtUMullerUMetzKALederLDCytokeratin and neurofilament protein staining in Merkel cell carcinoma of the small cell type and small cell carcinoma of the lungAm J Dermatopathol19982034635161.ShahIANettoDSchlageterMONeurofilament immunoreactivity in Merkel-cell tumors: a differentiating feature from small-cell carcinomaMod Pathol199363962.SuLDFullenDRLoweLCD117 (KIT receptor) expression in Merkel cell carcinomaAm J Dermatopathol20022428929363.PilottiSRilkeFBartoliCGrisottiAClinicopathologic correlations of cutaneous neuroendocrine Merkel cell carcinomaJ Clin Oncol198861863187364.Van GeleMVan RoyNRonanSGMolecular analysis of 1p36 breakpoints in two Merkel cell carcinomasGenes Chromosomes Cancer199823677165.LeonardJHCookALNancarrowDDeletion mapping on the short arm of chromosome 1 in Merkel cell carcinomaCancer Detect Prev20002462062766.MarisJMMatthayKKMolecular biology of neuroblastomaJ Clin Oncol1999172264227967.SmedleyDSidharSBirdsallSCharacterization of chromosome 1 abnormalities in malignant melanomasGenes Chromosomes Cancer20002812112568.HarnettPRKearsleyJHHaywardNKLoss of allelic heterozygosity on distal chromosome 1p in Merkel cell carcinomaA marker of neural crest origins? Cancer Genet Cytogenet19915410911369.LeonardJHWilliamsGWaltersMKDeletion mapping of the short arm of chromosome 3 in Merkel cell carcinomaGenes Chromosomes Cancer19961510210770.GoesslingWMcKeePHMayerRJMerkel cell carcinomaJ Clin Oncol20022058859871.LeonardJHLeonardPKearsleyJHChromosomes 1, 11, and 13 are frequently involved in karyotypic abnormalities in metastatic Merkel cell carcinomaCancer Genet Cytogenet199367657072.HarleMArensNMollIComparative genomic hybridization (CGH) discloses chromosomal and subchromosomal copy number changes in Merkel cell carcinomasJ Cutan Pathol19962339139773.LarsimontDVerhestAChromosome 6 trisomy as sole anomaly in a primary Merkel cell carcinomaVirchows Arch199642830530974.Van GeleMLeonardJHVan RoyNFrequent allelic loss at 10q23 but low incidence of PTEN mutations in Merkel cell carcinomaInt J Cancer20019240941375.LeonardJHHayardNLoss of heterozygosity of chromosome 13 in Merkel cell carcinomaGenes Chromosomes Cancer199720939776.CarsonHJLueckNEHortenBCComparison of mutant and wild-type p53 proteins in Merkel cell carcinomaClin Diagn Lab Immunol2000732677.Van GeleMKaghadMLeonardJHMutation analysis of P73 and TP53 in Merkel cell carcinomaBr J Cancer20008282382678.TsaoHZhangXMajewskiPHaluskaFGMutational and expression analysis of the p73 gene in melanoma cell linesCancer Res19995917217479.LeonardJHCookALVan GeleMProneural and proneuroendocrine transcription factor expression in cutaneous mechanoreceptor (Merkel) cells and Merkel cell carcinomaInt J Cancer200210110311080.NguyenBDMcCulloughAEImaging of Merkel cell carcinomaRadiographics20022236737681.GollubMJGruenDRDershawDDMerkel cell carcinoma: CT findings in 12 patientsAJR Am J Roentgenol199616761762082.KwekkeboomDJHoffAMLambertsSWSomatostatin analogue scintigraphy. A simple and sensitive method for the in vivo visualization of Merkel cell tumors and their metastasesArch Dermatol199212881882183.di Bartolomeo M, Bajetta E, Buzzoni R et al. Clinical efficacy of octreotide in the treatment of metastatic neuroendocrine tumors. A study by the Italian Trials in Medical Oncology GroupCancer19967740240884.Guitera-RovelPLumbrosoJGautier-GougisMSIndium-111 octreotide scintigraphy of Merkel cell carcinomas and their metastasesAnn Oncol20011280781185.LampreaveJLBenardFAlaviAPET evaluation of therapeutic limb perfusion in Merkel's cell carcinomaJ Nucl Med1998392087209086.KokoskaERKokoskaMSCollinsBTEarly aggressive treatment for Merkel cell carcinoma improves outcomeAm J Surg199717468869387.RatnerDNelsonBRBrownMDJohnsonTMMerkel cell carcinomaJ Am Acad Dermatol19932914315688.Al-GhazalSKAroraDSSimpsonRHSaxbyPMerkel cell carcinoma of the skinBr J Plast Surg19964949149689.GillenwaterAMHesselACMorrisonWHMerkel cell carcinoma of the head and neck: effect of surgical excision and radiation on recurrence and survivalArch Otolaryngol Head Neck Surg200112714915490.O'ConnorWJRoenigkRKBrodlandDGMerkel cell carcinoma. Comparison of Mohs micrographic surgery and wide excision in eighty-six patientsDermatol Surg19972392993391.BoyerJDZitelliJABrodlandDGD'AngeloGLocal control of primary Merkel cell carcinoma: review of 45 cases treated with Mohs micrographic surgery with and without adjuvant radiationJ Am Acad Dermatol20024788589292.AshbyMAJonesDHTaskerADBlackshawAJPrimary cutaneous neuroendocrine (Merkel cell or trabecular carcinoma) tumour of the skin: a radioresponsive tumourClin Radiol198940858793.CotlarAMGatesJOJrGibbs FAMerkel cell carcinoma: combined surgery and radiation therapyAm Surg19865215916494.PacellaJAshbyMAinslieJMintyCThe role of radiotherapy in the management of primary cutaneous neuroendocrine tumors (Merkel cell or trabecular carcinoma): experience at the Peter MacCallum Cancer Institute (Melbourne, Australia)Int J Radiat Oncol Biol Phys1988141077108495.BischofMvan KampenMHuberPWannenmacherMMerkel cell carcinoma: the role of radiation therapy in general managementStrahlenther Onkol199917561161596.MarksMEKimRYSalterMMRadiotherapy as an adjunct in the management of Merkel cell carcinomaCancer199065606497.WilderRBHarariPMGrahamARMerkel cell carcinoma. Improved locoregional control with postoperative radiation therapyCancer1991681004100898.Medina-FrancoHUristMMFiveashJMultimodality treatment of Merkel cell carcinoma: case series and literature review of 1024 casesAnn Surg Oncol2001820420899.MorrisonWHPetersLJSilvaEGThe essential role of radiation therapy in securing locoregional control of Merkel cell carcinomaInt J Radiat Oncol Biol Phys199019583591100.MortierLMirabelXFournierCRadiotherapy alone for primary Merkel cell carcinomaArch Dermatol200313915871590101.MehranyKOtleyCCWeenigRHA meta-analysis of the prognostic significance of sentinel lymph node status in Merkel cell carcinomaDermatol Surg200228113117102.PfeiferTWeinbergHBradyMSLymphatic mapping for Merkel cell carcinomaJ Am Acad Dermatol199737650651103.BradyMSCurrent management of patients with merkel cell carcinomaDermatol Surg200430321325104.HillADMannGBBorgenPICodyHSSentinel lymphatic mapping in breast cancerJ Am Coll Surg19991885455493rd105.RodriguesLKLeongSPKashani-SabetMWongJHEarly experience with sentinel lymph node mapping for Merkel cell carcinomaJ Am Acad Dermatol200145303308106.PectasidesDMoutzouridesGDimitriadisMVarthalitisJAthanassiouAChemotherapy for Merkel cell carcinoma with carboplatin and etoposideAm J Clin Oncol199518418420107.FenigELurieHKleinBSulkesAThe treatment of advanced Merkel cell carcinoma: A multimodality chemotherapy and radiation therapy treatment approachJ Dermatol Surg Oncol199319860864108.FenigELurieHSulkesAThe use of cyclophosphamide, methotrexate, and 5-fluorouracil in the treatment of Merkel cell carcinomaAm J Clin Oncol1993165457109.DavisMPMillerEMRauRCThe use of VP16 and cisplatin in the treatment of Merkel cell carcinomaJ Dermatol Surg Oncol199016276278110.TaiPTYuEWinquistEChemotherapy in neuroendocrine/Merkel cell carcinoma of the skin: case series and review of 204 casesJ Clin Oncol20001824932499111.VoogEBironPMartinJPBlayJYChemotherapy for patients with locally advanced or metastatic Merkel cell carcinomaCancer19998525892595112.PectasidesDPectasidesMKoumarianouACisplatin-based chemotherapy for Merkel cell carcinomaCancer Investin press