Your activity: 26966 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: [email protected]

Staging, treatment, and surveillance of Merkel cell carcinoma

Staging, treatment, and surveillance of Merkel cell carcinoma
Authors:
Patricia Tai, MB, BS, FRCPC
Song Youn Park, MD
Paul T Nghiem, MD, PhD
Section Editors:
Robert S Stern, MD
June K Robinson, MD
Michael B Atkins, MD
Deputy Editor:
Sonali Shah, MD
Literature review current through: Feb 2022. | This topic last updated: Nov 19, 2020.

INTRODUCTION — Merkel cell carcinoma (MCC) is a rare, aggressive, cutaneous malignancy that predominantly affects older adults with light skin types and has a high propensity to metastasize.

The staging, treatment, and surveillance of MCC are discussed here. The clinical features and initial diagnosis of MCC are reviewed separately. (See "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma".)

STAGING SYSTEM — The eighth edition American Joint Committee on Cancer (AJCC) staging system provides important information for both management and prognosis of patients with MCC [1]. Prior to adoption of the AJCC staging system, the standard in the field, multiple other staging systems were used [2].

MCC staging is assigned based upon tumor, node, and metastasis assessment. These stages can be summarized as follows (table 1 and table 2):

Stage I – Primary tumors ≤2 cm in maximum dimension (T1), without evidence of regional lymph node involvement.

Stage II – Primary tumors >2 cm (T2 or T3) or a primary tumor with invasion into bone, muscle, fascia, or cartilage (T4), without evidence of lymph node involvement. Stage II is further divided into two subgroups based upon the size and depth of invasion of the primary tumor.

Stage III – Any primary tumor with in-transit metastasis or regional lymph node disease. Pathologic stage III is divided into subgroups based upon the extent of in-transit or regional lymph node involvement.

Stage IV – Metastasis beyond the regional lymph nodes, regardless of the status of the primary tumor and regional nodes.

PROGNOSTIC FACTORS — The extent of disease at presentation, which is represented by the American Joint Committee on Cancer (AJCC) staging system, is the single most important determinant of prognosis. Along with staging, several clinical and pathologic features independently affect prognosis. (See 'Surveillance after initial therapy' below and "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma", section on 'Pathogenesis'.)

Stage — Stage-specific recurrence rates, disease-specific survival, and overall survival (OS) vary between study cohorts. However, stage is consistently reported to be the most important prognostic factor [3-5].

An analysis of 9387 patients with MCC from the National Cancer Database (NCDB) provides more detailed information about the extent of disease and OS (figure 1A-C) [3]. Of note, disease-specific survival by stage was not evaluated in this study, so clinicians should be aware that, in the older population affected by MCC, a significant proportion of deaths captured by OS are not due to MCC.

Local disease – At initial presentation, 65 percent of patients had local disease without evidence of regional lymph node involvement or metastatic disease. The five-year OS for these patients was 55.6 percent, and there was a progressive decrease in OS for those with a T1 (<2 cm), T2-3 (>2 to 5, >5 cm), and T4 (invasion of fascia or deeper tissues) primary lesion, with five-year OS rates of 55.8, 41.1, and 31.8 percent, respectively.

Regional disease – At initial presentation, 26 percent of patients had regional lymph node involvement without disseminated metastatic disease. This group included those with occult disease (detected by sentinel lymph node (SLN) biopsy or regional lymph node dissection), those with clinically detected regional lymph node involvement, and those with in-transit metastases. The five-year OS for the entire group was 35.4 percent, 39.7 percent for patients with occult disease, 26.8 percent for those with clinically detected nodal disease, and 41.4 percent for those with in-transit disease.

Patients with lymph node metastasis and an unknown primary tumor constituted 3.6 percent of the total cohort. The five-year OS for unknown primary patients was 42.2 percent.

Distant metastases – At initial presentation, approximately 8 percent of patients had distant metastatic disease. The five-year OS for these patients was 13.5 percent.

Positive Merkel cell polyomavirus status — The presence of the positive Merkel cell polyomavirus (MCPyV) antigen within the tumor is associated with a favorable prognosis, including improved disease-specific survival and reduced risk of recurrence [6-8]. Observational data supporting the prognostic utility of the MCPyV status are discussed separately [6,7]. (See "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma", section on 'Prognostic factors'.)

Other clinical and histologic factors — Clinical prognostic factors that are significantly favorable for OS, after adjustment for stage, include female sex, younger age at diagnosis, immune competence, and absence of comorbid conditions [9,10]. Inclusion of these factors could provide more precise prognostic information. A web-based individualized risk containing these risk factors is available.

INITIAL EVALUATION AND MANAGEMENT — Once a tumor biopsy has confirmed the diagnosis of MCC, the following evaluation and initial management are suggested. The diagnostic clinical and histopathologic features of a primary MCC tumor are discussed separately. (See "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma", section on 'Diagnosis'.)

Physical examination — The initial evaluation of a patient with MCC should include a complete examination of the skin and regional lymph nodes, with appropriate evaluation of any detected abnormalities. The clinical features of MCC are discussed separately. (See "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma", section on 'Clinical features'.)

Imaging — For initial imaging, we offer either positron emission tomography (PET) with fluorodeoxyglucose combined with either computed tomography (CT) or magnetic resonance imaging (MRI), or a CT of the chest, abdomen, and pelvis. MRI of the brain may be obtained if the patient is symptomatic to assess for the presence of central nervous system metastases. These imaging modalities are critical due to the metastatic potential of MCC and may influence subsequent management [11-14].

Imaging studies are strongly indicated if metastatic disease is suspected by physical examination. In addition, we offer baseline imaging to most patients to identify those with a higher stage of disease who would benefit from a different treatment approach. In one observational series, baseline imaging detected occult metastatic disease in 13 percent of patients who presented without suspicious physical examination findings (eg, palpable adenopathy) [15].

Merkel cell polyomavirus status — Baseline determination of Merkel cell polyomavirus (MCPyV) oncoprotein antibody titer can further assist with prognosis and subsequent detection of recurrent disease [8,16]. (See "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma", section on 'Prognostic factors' and "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma", section on 'Merkel cell polyomavirus serology' and 'Positive Merkel cell polyomavirus status' above.)

Approach to initial management — Based upon the initial evaluation, the approach to initial management of patients with MCC is dependent on the pathologic characteristics of the primary tumor and the presence of clinical evidence of lymph node or distant metastases.

Clinically negative nodes – For patients with a biopsy-confirmed MCC and no clinical evidence of regional lymph node involvement, a sentinel lymph node (SLN) biopsy is generally indicated prior to, or in conjunction with, definitive local therapy. (See 'Clinically negative nodes' below.)

However, the role of SLN biopsy is less clear for lesions arising in the head and neck region, due to the variable lymphatic drainage of primary tumors in this region. (See 'Head and neck lesions' below.)

Negative SLN biopsy – For patients with a negative SLN biopsy, an excision of the primary tumor is indicated. For patients in whom surgery is not technically feasible or for those who are not surgical candidates, radiation therapy (RT) to the primary tumor and nodal region is an alternative [17,18]. (See 'Primary tumor' below.)

Positive SLN biopsy – For patients with a positive SLN biopsy, careful evaluation for occult metastatic disease is indicated. If no distant metastatic disease is detected, treatment of the primary tumor by excision and adjuvant RT and definitive treatment of regional lymph node basin by nodal dissection or definitive RT is indicated. However, performing both RT and nodal dissection of the lymph node basin is significantly morbid and does not improve control for SLN-positive disease [19]. (See 'Regional lymph nodes' below.)

No SLN biopsy – For patients not undergoing SLN mapping and biopsy, adjuvant RT to the regional lymph nodes may be indicated. Adjuvant RT to the regional lymph nodes is a reasonable approach to avoid relapse as nodal evaluation is not performed. Without any elective regional lymph node treatment, at least 17 percent of patients with a primary tumor ≤1 cm will eventually develop nodal disease [20,21]. (See 'Adjuvant radiation therapy' below.)

Clinically positive nodes – For patients with suspected clinical lymph node involvement, biopsy confirmation (fine needle aspirate, core needle biopsy, or excisional biopsy) is indicated.

If the fine needle aspirate or core needle biopsy of the suspicious node is negative, an excisional nodal biopsy may be indicated [22].

If the biopsy confirms the presence of nodal disease, patients should be carefully evaluated for the presence of metastatic disease. If there are no distant metastases detected, we recommend excision of the primary tumor and definitive therapy of the draining lymphatics. Treatment options include: (a) regional lymph node dissection, or (b) nodal excision followed by regional RT covering the intervening lymphatics if the RT field size is not too large or morbid. For very frail patients who cannot tolerate nodal excision even if carried out only with local anesthetic, definitive regional RT may be used instead, although local control will only be approximately 75 to 85 percent if clinically evident nodal disease is not surgically excised before RT [19,23-25]. (See 'Regional lymph nodes' below.)

For patients who have completed a wide local excision of the primary tumor and a regional lymph node dissection, potential risk factors indicating the need for adjuvant RT to the primary site and/or involved node bed include the presence of a primary tumor ≥1 cm, positive or limited surgical resection margins, lymphovascular invasion, a head and neck primary, and an immunocompromised host. In addition, extracapsular extension of tumor outside the lymph node(s) or the involvement of multiple nodes are both associated with an increased risk of recurrence and are indications for adjuvant regional RT. Participation in a clinical trial of adjuvant immunotherapy, if available, may be offered to high-risk patients following the completion of surgery with or without RT of the regional lymph nodes. (See 'Adjuvant immunotherapy' below.)

Metastatic disease – For patients with suspected disseminated disease after their initial evaluation, biopsy confirmation of the metastasis is indicated prior to systemic therapy. The treatment approach to metastatic disease is discussed below. (See 'Metastatic disease' below.)

LOCOREGIONAL DISEASE

Primary tumor — Surgery, rather than radiation therapy (RT), is preferred as the initial therapy of the primary tumor. However, given that adjuvant RT to the primary site is often indicated after excision, it is important to balance the extent of surgery and accompanied morbidity (eg, wound healing, cosmesis, and function).

Surgery — Wide excision of the primary MCC tumor is the standard initial management whenever possible [26,27]. A margin of at least 1 to 2 cm of normal-appearing skin is recommended. If the margins are close to or involved with tumors, postoperative RT is indicated to increase the probability of local disease control [16,26]. (See 'Adjuvant radiation therapy' below.)

Achieving wide negative margins with the initial excision is important for long-term disease control, especially if no adjuvant RT to the primary site is planned [17,28]. However, data are limited regarding the appropriate surgical margin size if postoperative RT will be delivered. Furthermore, critical information, such as sentinel lymph node (SLN) positivity, pathologic surgical margins, and lymphovascular invasion, is missing at the time of the surgery. As examples:

In a retrospective observational study of 179 patients with MCC treated with curative intent, the addition of adjuvant RT to narrow excision (<10 mm margin) was associated with a reduction of local recurrence rates (25 versus 5 percent) and was also effective in the presence of microscopic positive margins. Irrespective of whether RT was used or not, the local recurrence rate was 7 percent when the margin size was >10 mm [28].

In a retrospective analysis of 188 patients with MCC, 48 were patients treated with surgery alone (all of whom had microscopically negative margins). Of those with surgical margins ≤1 cm, 20 percent (7 of 35 patients) developed a local recurrence, whereas none (0 of 13 patients) developed a local recurrence among those with margins >1.0 cm. In contrast, among 140 patients treated with adjuvant RT, local control was excellent for both large and small surgical margins, even among patients who had microscopically positive margins (1 percent local recurrence for both margin groups, n = 70 for both groups) [29].

Based upon the available data, our practice aims to select a surgical margin size that offers complete excision of disease yet is conservative enough to avoid significant delay in the initiation of adjuvant RT when it is indicated. Therefore, it is recommended that when adjuvant RT is planned, extensive tissue movement and grafting are avoided [22].

Mohs micrographic surgery can be offered where cosmetic outcomes are important, such as the face. If an SLN biopsy is to be carried out, it should be performed prior to Mohs surgery as the tumor excision site may interfere with a subsequent node biopsy [22]. With the Mohs approach, 100 percent of all major borders, including the deep margins, are evaluated histologically. However, adjuvant RT still has a role in preventing locoregional recurrences when Mohs surgery is used since MCC tends to recur beyond pathologically negative margins more often than other skin cancers [30-32].

Definitive radiation therapy — For patients in whom surgical resection is not technically feasible or who are medically unfit for surgery, definitive RT serves as an alternative treatment option [16,17,23,33]. Higher doses of radiation (60 to 66 Gy) are recommended if RT is used alone rather than in the adjuvant setting [16]. RT achieves an in-field disease control rate of only 75 to 85 percent, and systemic relapses are not rare [23,24].

The role of chemoradiotherapy versus RT alone is controversial, and there are no randomized trials comparing chemoradiotherapy with RT alone as the primary therapy for nonsurgical candidates. (See 'Adjuvant chemotherapy or chemoradiotherapy' below.)

Regional lymph nodes — Pathologic staging of lymph node status is important for both prognosis and treatment planning [34]. Optimal management of the regional lymphatics depends on whether there is clinical or pathologic evidence of MCC involvement as well as the size and pathologic characteristics of the primary tumor. The management approach of the regional lymph nodes is based upon retrospective series since there are no randomized trials that provide definitive guidance.

Clinically negative nodes — Most patients with a biopsy-proven MCC should have SLN mapping and biopsy, including immunohistochemistry [16,26].

However, some clinicians do not perform SLN mapping and biopsy when the primary tumor size is ≥2 cm, since RT is indicated due to the high risk of lymph node metastasis.

In addition, the role of SLN biopsy is less clear for patients with a lesion arising in the head and neck region since the lymphatic drainage from primary tumors in this region is highly variable to multiple nodal basins. If an SLN biopsy is not carried out, lymph nodes should usually be treated with prophylactic RT in higher-risk cases [16]. (See 'Head and neck lesions' below.)

The SLN mapping and biopsy should be performed prior to or in the same setting as the wide local excision because surgery may disrupt the lymphatic drainage from the primary tumor. This technique, which is widely used in patients with melanoma, is discussed separately. (See "Evaluation and treatment of regional lymph nodes in melanoma", section on 'Sentinel lymph node biopsy'.)

The results of the SLN biopsy provide important prognostic information and help guide further treatment. The impact on prognosis is illustrated by an analysis of 9387 patients with MCC from the National Cancer Database (NCDB) [3]. Patients with a negative SLN biopsy had improved five-year overall survival (OS) compared with those with a positive biopsy (55.6 versus 39.7 percent).

Approximately one-third of patients with clinically negative lymph nodes will have microscopic tumor involvement on pathologic examination [4]. However, a negative SLN biopsy does not preclude a subsequent regional recurrence. In an analysis of 721 patients, the false-negative rate for an SLN biopsy (as defined by a subsequent regional recurrence) was 17 percent [35]. Thus, adjuvant treatment to regional lymph nodes may rarely be indicated even after a negative SLN biopsy, depending upon the characteristics of the primary tumor or in high-risk situations such as an immunosuppressed patient. (See 'Adjuvant therapy' below.)

For patients with a negative SLN biopsy, adjuvant regional lymph node RT may be indicated if there is a high risk of recurrence due to anatomic, technical, or histologic factors (eg, previous wide local excision or immunohistochemistry evaluation of SLN biopsy was not performed). If none of these factors are present and the patient is compliant with follow-up and imaging, observation is appropriate after SLN biopsy.

If the SLN contains a tumor, regional lymph node dissection or definitive RT to the lymph node basin is indicated. However, performing both RT and nodal dissection is significantly morbid and does not improve control for SLN-positive disease [19]. For patients managed with a regional lymph node dissection, postoperative RT may be indicated, particularly in those with more extensive lymph node disease or extracapsular extension.

Clinically positive nodes — Patients with suspected lymph node involvement based upon clinical or radiographic findings require histologic confirmation of disease [16]. This may be obtained by a fine needle aspirate, core needle biopsy, or open biopsy.

If pathologic involvement of the regional nodes is confirmed, regional nodal therapy with surgery is indicated with or without postoperative RT. Excision of the clinically abnormal node followed by RT is supported by preliminary evidence [17]. Node dissection is recommended in the National Comprehensive Cancer Network guidelines. RT as the primary modality is an option in selected frail patients who cannot tolerate even nodal excision under local anesthetic [19,25].

The role of adjuvant RT following complete lymph node dissection has not been definitively established by randomized trials. However, multiple observational studies provide evidence that RT can decrease the rate of locoregional recurrence and improve OS [28,36-38]. Involvement of multiple lymph nodes or extracapsular extension of tumor outside the lymph node(s) is associated with an increased risk of recurrence and is an indication for adjuvant regional RT following lymph node dissection. The risks of node dissection plus RT include lymphoedema, fibrosis of the joints, and subsequent functional impairment. Therefore, despite retrospective studies showing decreased recurrence rates with adjuvant RT, clinical judgment is required to decide which patients can be followed without adjuvant RT after nodal dissection. In general, patients with clinically positive nodes have a five-year disease-specific survival of approximately 50 percent [39].

The role of adjuvant immunotherapy is the subject of ongoing clinical trials. (See 'Adjuvant immunotherapy' below.)

For patients with a clinically positive lymph node that cannot be confirmed by appropriate pathologic examination (eg, confirmatory nodal biopsy is not feasible), we generally suggest RT to the primary tumor and draining lymph node basin. (See 'Adjuvant therapy' below.)

Special considerations

Head and neck lesions — Management of MCC arising in the head and neck region can present a unique problem because achieving an adequate wide resection in these patients is often particularly difficult and the lymphatic drainage from primary tumors in this region is highly variable [40-42]. MCCs arising in the head and neck appear to be associated with a worse prognosis than those arising in other sites [43].

Although there are no randomized trials in patients with MCC arising in the head and neck region, RT to the resection bed, in-transit lymphatics, and regional lymph nodes is generally recommended [16].

The benefit of adjuvant RT was reported in multiple studies. In an analysis of 46 low-risk head and neck MCC patients, the group that did not receive postoperative RT had a significantly higher risk of local recurrence (26 versus 0 percent) [44]. In another study of 4815 patients with MCC of the head and neck region, OS was significantly improved with postoperative RT compared with surgery alone (hazard ratio [HR] 0.80, 95% CI 0.70-0.92) [37].

Analyses of other large series and a review of the literature have also concluded that the inclusion of RT to the tumor bed and regional lymphatics as part of initial treatment improves local and regional tumor control [40-42,45].

The role of adjuvant chemoradiotherapy is uncertain and/or controversial. (See 'Adjuvant chemotherapy or chemoradiotherapy' below.)

Unknown primary — Among patients presenting with clinically detectable lymph nodes at the time of MCC diagnosis (stage IIIB), one-third to one-half of patients do not have an identifiable primary tumor [46]. In these cases, the primary tumor most likely regressed due to immune surveillance as these patients show markedly stronger immune responses across multiple parameters [46]. The concept that the MCC may have arisen within the node itself is unlikely as MCC tumor cells in the node have extensive ultraviolet-induced DNA mutations, strongly suggesting a cutaneous origin [46].

Observational series suggest that patients with an unknown primary, regional nodal disease and no evidence of distant metastases do significantly better (with approximately one-half the risk of disease progression) compared with those with a primary cutaneous site in addition to clinically positive lymph nodes [46-50].

As an example, an observational series of 321 patients with MCC included a subset of 38 patients (12 percent) who presented with an unknown primary, nodal disease and no evidence of distant metastases [50]. Among this cohort of patients, median recurrence-free survival was 35 months. Ten patients (26 percent) died, five of disease and five of other causes. Median OS was 104 months. This study did not have comparative data for patients with a known primary. However, in the 5371-patient NCDB, as shown in the figure (figure 1B), the median survival of node-positive patients with a known primary (pathologic IIIB) is around two years and is around 3.5 years for those with an unknown primary, almost double that of pathologic IIIB.

Management should be individualized based upon the specific pattern of disease presentation and should follow that for patients with an identifiable primary tumor and clinically positive lymph nodes or distant metastases. (See 'Clinically positive nodes' above and 'Metastatic disease' below.)

Adjuvant therapy — RT has an important role in treating occult lymph node disease or minimum residual tumors at the primary site in patients whose MCC is primarily managed with surgery. The role of systemic treatment (chemotherapy, immunotherapy) as part of a combined modality approach is not yet well established.

Adjuvant radiation therapy — Adjuvant RT is indicated in patients with any of the following risk factors: a primary tumor ≥1 cm in maximum dimension, a head and neck primary, positive or limited surgical resection margins, lymphovascular invasion, or an immunocompromised host. RT to the nodal basin is an option for patients with profound immunosuppression [51] or for those with a suspected false-negative SLN biopsy.

MCC is a radiosensitive malignancy. RT has been used as an adjuvant treatment after surgery to prevent recurrence in the primary tumor bed and regional lymphatics. Although there are no randomized trials supporting the role of adjuvant RT, multiple studies provide evidence that RT can decrease the rate of locoregional recurrence and improve OS [28,36-38].

The most extensive evidence supporting the role of adjuvant RT comes from an analysis of 6908 patients with MCC in the NCDB [38]. In the 4843 patients without evidence of lymph node involvement (stage I or II), adjuvant RT was associated with significantly improved OS (HR 0.71, 95% CI 0.64-0.80). In contrast, there was no improvement in OS observed in the 2065 patients with lymph node-positive (stage III) MCC (HR 0.98, 95% CI 0.86-1.12).

When RT is used as an adjuvant or definitive treatment, it is important to treat all regional lymphatics to avoid geographic miss since a recurrence in untreated regions may occur. Some providers use a 5 cm margin to cover the surgical bed for definitive or adjuvant RT [52,53]. As an example, the Trans Tasman Radiation Oncology Group (TROG) 96:07 study used a 3 to 5 cm margin, and the draining lymph nodes were treated in the same field as the primary if the nodal region was within 20 cm of the primary to reduce the risk of in-transit recurrence [54,55].

The RT dose required to treat either the primary tumor or regional lymph nodes is based upon the tumor burden [16]. Following a resection with negative margins, a total RT dose of 50 to 56 Gy is generally adequate for those at significant risk for residual subclinical disease. A dose of 56 to 60 Gy is recommended for those with microscopically positive resection margins, and a dose of 60 to 66 Gy is recommended for definitive treatment of those with grossly positive margins, an unresectable primary lesion, or unresectable regional lymph nodes.

Adjuvant chemotherapy or chemoradiotherapy — The role of chemotherapy, either alone or in combination with RT, as an adjuvant following surgery for locoregional therapy is uncertain and/or controversial.

Most studies are retrospective and were carried out prior to the introduction of immunotherapy. In addition, there are no randomized trials evaluating the efficacy of adjuvant chemotherapy or chemoradiotherapy in patients with MCC. Furthermore, there is concern about the immunosuppressive effects of chemotherapy. The immune system plays an important role in defense against MCC, based upon the increased incidence in immunosuppressed patients, association with Merkel cell polyomavirus (MCPyV), and reports of spontaneous regression. Data are as follows:

The most extensive data regarding chemoradiotherapy derive from a retrospective study of 4815 patients with MCC of the head and neck region, based on the NCDB between 1998 and 2011 [37]. In this analysis, 1995 patients were managed with surgery alone, 2330 managed with postoperative RT, 393 managed with postoperative chemoradiotherapy, and 97 managed with postoperative chemotherapy alone. With a median survival of 3.4 years, postoperative chemoradiotherapy and RT both provided a survival benefit over surgery alone (HR 0.62, 95% CI 0.47-0.81, and HR 0.80, 95% CI 0.70-0.92, respectively). However, postoperative chemotherapy was associated with decreased OS (HR 1.74, 95% CI 1.10-2.75) when compared with surgery alone.

Another large study from the NCDB included 6908 patients with MCC at all sites, including 2065 patients with stage III disease [38]. Neither adjuvant chemotherapy nor chemoradiotherapy was associated with statistically improved or worsened OS compared with patients treated without chemotherapy (stage I HR 0.79, 95% CI 0.60-1.05; stage II HR 1.14, 95% CI 0.89-1.45; stage III HR 0.97, 95% CI 0.85-1.12).

In the prospective TROG 96:07 study, 53 patients were classified as being at high risk of recurrence based upon having a recurrence after initial therapy, positive regional lymph nodes, a primary tumor ≥1 cm, or gross residual disease after surgery [54,55]. Treatment consisted of RT (50 Gy in 25 fractions over five weeks) and synchronous carboplatin (area under the curve 4.5) plus etoposide (80 mg/m2 intravenously on days 1 to 3) during weeks 1, 4, 7, and 10. With a median follow-up of 48 months, the three-year OS, locoregional control, and distant control rates were 76, 75, and 76 percent, respectively [55]. However, a comparison with historical controls suggested that chemotherapy did not affect OS compared with surgery plus RT without chemotherapy [54].

Patients exposed to prior chemotherapy may have a lower response rate to immunotherapy. In the phase 2 JAVELIN Merkel 200 trial, the objective response rate to avelumab in patients previously treated with chemotherapy was lower than those who had received avelumab as first-line therapy (28 versus 62 percent) [56,57]. (See 'Avelumab' below.)

When carboplatin is being given during RT, a weekly regimen (area under the curve 2) may be a safer way to deliver synchronous chemotherapy [58]. This approach reduced the incidence of febrile neutropenia and grade 3 skin toxicity compared with the every-three-week regimen of carboplatin and etoposide used in the TROG 96:07 study [54].

In patients with metastatic disease, systemic chemotherapy has demonstrated high response rates but short duration of responses. The use of systemic chemotherapy in metastatic disease is discussed below. (See 'Metastatic disease' below.)

Adjuvant immunotherapy — In patients with resected MCC, there are limited data on the use of immunotherapy in the adjuvant setting. Adjuvant immunotherapy with checkpoint inhibitors is being evaluated in randomized trials. Enrollment in such a trial is encouraged when feasible.

Checkpoint inhibitor immunotherapy is now the preferred therapy for patients with metastatic MCC, and its use is discussed below. (See 'Metastatic disease' below.)

IS THERE A ROLE FOR NEOADJUVANT IMMUNOTHERAPY? — The role of neoadjuvant immunotherapy in the management of patients with MCC is not established and remains under investigation. Neoadjuvant immunotherapy may potentially decrease the extent of necessary surgery or the need for postoperative radiation therapy (RT) in patients with a good pathologic response. While preliminary data suggest pathologic complete response (pCR) in approximately one-half of patients [59], additional studies are required to confirm these results.

Nivolumab – Nivolumab was evaluated in the neoadjuvant setting in a phase I/II study (Checkmate 358). In this study of 39 patients with resectable MCC, surgical resection specimens were obtained after two doses of nivolumab (240 mg on days 1 and 15) [59]. Among the 36 patients who underwent surgery, 17 patients (47 percent) achieved a pCR. Among 33 radiographically evaluable patients, 18 (55 percent) had tumor reductions greater than 30 percent. At a median follow-up of 20.3 months, recurrence-free survival significantly correlated with pCR and radiographic response at the time of surgery. No patient with a pCR had tumor relapse during observation.

SURVEILLANCE AFTER INITIAL THERAPY

Frequency of follow-up — Patients with MCC may benefit from frequent follow-up because of the high rate of recurrences. Follow-up should be individualized according to risk factors and potential therapeutic options.

The National Comprehensive Cancer Network guidelines recommend that patients should be followed every three to six months for three years and then every 6 to 12 months thereafter. Physical examination, with special emphasis on total-body skin examination and palpation of lymph nodes, is justified by the frequency of skin and nodal recurrences and the lack of expense or toxicity associated with such examinations.

Helpful data for deciding ongoing follow-up frequency after initial management are available in the form of stage-specific recurrence-free survival rates [60] and a web-based risk calculator tool.

Imaging studies — The use of imaging studies (eg, magnetic resonance imaging [MRI], computed tomography [CT]/positron emission tomography [PET]) should be based upon clinical indications [9,61]. Routine imaging should be considered for high-risk patients [16].

Merkel cell polyomavirus serology — We suggest that the Merkel cell polyomavirus (MCPyV) oncoprotein antibody test be performed every three months while the patient remains at risk for MCC recurrence (typically up to approximately five years). If the titer increases more than 30 percent from the previous value, an imaging study is warranted to evaluate possible recurrence. (See "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma", section on 'Merkel cell polyomavirus serology'.)

Changes in the titer of antibodies that recognize MCPyV oncoproteins reflect alterations in a patient's MCC tumor burden [62-66]. As such, this test can be a useful component of ongoing surveillance for seropositive patients. A rising titer can be an early indicator of recurrence. Additionally, for patients who do not have recurrent disease, titers are expected to decrease significantly within three months of successful treatment of MCC [8]. A combination of the MCPyV oncoprotein antibody test and imaging studies can help guide surveillance.

In an analysis of 260 patients with MCC who had a positive MCPyV oncoprotein antibody test, the positive predictive value of recurrence was 99 percent (eg, fraction of patients with rising titers that had or soon developed clinically evident MCC recurrence) and the negative predictive value was 99 percent (eg, fraction of patients with stable or declining titers who did not have detectable recurrent disease) [67].

Secondary malignancies — MCC is associated with an increased incidence of other skin cancers, particularly hematologic malignancies that are associated with immunosuppression. Patients with treated MCC who are under surveillance should also be evaluated for the potential development of such secondary malignancies (eg, on physical examination). (See "Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma", section on 'Epidemiology'.)

RECURRENT DISEASE — Following initial therapy, MCC has a 20 to 75 percent chance of recurrence, depending on stage at diagnosis [68]. In one literature review, the reported local recurrence rate was 29 percent; nodal or distant disease recurrence rates were 50 and 25 percent, respectively [9,61]. Patients with an initial nodal recurrence had a significantly higher chance of developing subsequent distant metastases than those without a nodal recurrence.

Local recurrences – Local recurrences usually occur within one year of initial therapy, with a median time to recurrence of four months (range one to nine months) [9,39,61,69]. Patients with locoregional recurrence should be carefully reevaluated to rule out disseminated disease. Although isolated locoregional recurrences are associated with a relatively poor prognosis, some patients with disease limited to locoregional sites can be salvaged with multimodal therapy. The impact of a local recurrence on survival is controversial [70,71].

Nodal recurrences – The median time to develop a clinically detectable nodal recurrence after resection of the primary lesion is seven to eight months. Among patients with nodal involvement, either at presentation or at recurrence, 11 to 66 percent die of their disease within five years [72-74]. As with local recurrences, multimodal approaches have been associated with improved outcome following a nodal recurrence. In a literature review of 661 patients, median overall survival (OS) was 27 months (range 1 to 216 months) among those who received salvage treatments for recurrent disease [9,61]. Combined modality therapy (surgery, radiation therapy [RT], and/or systemic therapy) is associated with the best salvage potential [72,75,76].

METASTATIC DISEASE — For patients with metastatic disease, biopsy confirmation of the metastasis is often indicated. Clinical factors that should be taken into account when deciding treatment course include sites of disease involvement, age, comorbidities, and patient preferences. A clinical trial of systemic therapy is preferred whenever possible.

Immunotherapy — Checkpoint inhibitor immunotherapy is the preferred initial treatment approach for patients with advanced MCC without absolute contraindications to such therapy [22,77]. (See "Toxicities associated with checkpoint inhibitor immunotherapy".)

In patients with advanced or metastatic MCC, we suggest initial treatment with a programmed cell death ligand 1 (PD-L1)-blocking agent (avelumab) or programmed cell death protein 1 (PD-1) inhibitor-based immunotherapy (pembrolizumab, nivolumab), rather than chemotherapy, as immunotherapy has a greater capacity to induce clinically meaningful, durable responses in such patients. The US Food and Drug Administration has approved avelumab and pembrolizumab to treat patients with advanced or metastatic MCC, irrespective of prior therapy.

Avelumab — Avelumab is a monoclonal antibody that binds to PD-L1.

In the international, multicenter, phase II JAVELIN Merkel 200 trial part A, 88 patients were treated with avelumab (10 mg/kg every two weeks) [56,78,79]. All patients had received prior cytotoxic chemotherapy, and 40 percent had received two or more lines of chemotherapy. With a minimum follow-up of two years (median 29.2 months), there were 29 objective responses (33 percent), including 10 complete responses (11 percent). The two-year progression-free survival rate was 26 percent, and the two-year overall survival (OS) rate was 36 percent. In this trial, avelumab was associated with infusion-related adverse reactions in 17 percent of patients; all were grade 1 or 2. Premedication with acetaminophen and an antihistamine is recommended prior to the first four infusions and subsequently as needed [80]. There were no grade 4 or 5 treatment-related adverse events in the JAVELIN Merkel 200 trial. Only 4 of 88 patients (5 percent) had grade 3 adverse events. (See "Toxicities associated with checkpoint inhibitor immunotherapy".)

The role of avelumab in MCC is also supported by preliminary results from part B of the JAVELIN Merkel 200 study in 29 previously untreated patients [57]. In a preplanned analysis with a median follow-up of five months, the objective response rate was 62 percent. In responding patients, 83 percent had a durable response for at least six months. No treatment-related deaths or grade 4 adverse events occurred.

Pembrolizumab — Pembrolizumab is a monoclonal antibody that binds to PD-1. In a phase II study, 50 patients who were systemic therapy-naïve were treated with pembrolizumab (2 mg/kg every three weeks) for up to two years [81,82]. Overall, 43 patients had stage IV disease and seven had stage III unresectable disease. At a median follow-up of 14.9 months, objective responses were observed in 28 of 50 evaluable patients (56 percent), which included 12 complete responses and 16 partial responses. Responses were durable, with 85 percent of responders remaining in remission at one year and 79 percent in remission at two years. OS rates at one, two, and three years were 72, 69, and 64 percent, respectively. There was no statistically significant difference in response rate or duration of response in patients whether or not tumors were positive for the Merkel cell polyomavirus (MCPyV) or based upon PD-L1 expression.

Nivolumab — Nivolumab is a monoclonal antibody that binds to PD-1. In a phase I/II study, 25 patients who were treatment-naïve or who had received one to two prior systemic chemotherapies were treated with nivolumab (240 mg every two weeks) [83]. In the 22 response-evaluable patients, the overall response rate was 68 percent, with ongoing responses in 13 of 15 patients (87 percent). At three months, progression-free survival and OS rates were 82 and 92 percent, respectively. Responses occurred in 71 percent of treatment-naïve patients and in 63 percent of patients with one to two prior systemic therapies, in both virus-positive and virus-negative tumors.

Other agents (ipilimumab) — Due to limited evidence on efficacy and safety, we do not offer the cytotoxic T-lymphocyte associated antigen 4 (CTLA-4) inhibitor ipilimumab monotherapy as a first-line treatment for metastatic MCC. Data are as follows:

The efficacy of ipilimumab monotherapy for MCC was initially studied in the adjuvant setting. In a randomized phase II study, compared with observation, ipilimumab did not improve disease-free survival and worsened toxicity. Given the lack of efficacy of ipilimumab with the pronounced toxicity, enrollment for the trial was discontinued [84].

For patients with metastatic MCC, ipilimumab monotherapy has not been studied in a trial, but one small case series reports efficacy of ipilimumab monotherapy in metastatic MCC [85].

For those with advanced disease that has progressed through anti-PD-1 therapy, one retrospective case series reported the efficacy of ipilimumab alone or in combination with an anti-PD-1 agent. Among the 13 patients evaluated, objective responses were seen in four patients (31 percent) [86].

Chemotherapy — Chemotherapy retains a role in the treatment of patients who do not respond to or relapse after immunotherapy or in those patients in whom immunotherapy is contraindicated. (See "Toxicities associated with checkpoint inhibitor immunotherapy", section on 'Preexisting autoimmune disease'.)

There are no randomized trials or prospective studies of chemotherapy in patients with distant metastases. Carboplatin plus etoposide is generally the initial treatment option, and weekly carboplatin (area under the curve 2.0) has been used to reduce toxicity [58]. Cyclophosphamide plus doxorubicin and vincristine is an alternative. Alternative regimens and second-line therapies are patterned after those used in small cell carcinoma of the lung. (See "Extensive-stage small cell lung cancer: Initial management", section on 'Rationale for addition of immune checkpoint inhibitors to chemotherapy' and "Treatment of refractory and relapsed small cell lung cancer".)

The most extensive results of chemotherapy come from a retrospectively analyzed series of 103 patients with distant metastases [61]. Patients were treated with a wide range of chemotherapy regimens (59 percent); the median duration for complete responses and partial responses was six and three months, respectively.

The impact of systemic chemotherapy on survival in patients with metastatic MCC is unclear; however, relapse-free survival averages only three months from the time of initiating therapy [87]. Furthermore, despite a relatively high response rate, cisplatin-based chemotherapy is toxic, especially in this relatively older adult population, and multiple treatment-related deaths have been reported [61].

Palliative treatment — For patients with metastatic disease that is symptomatic or nonresponsive to systemic therapy, options include local treatment of the symptomatic primary tumor and/or regional nodes with surgery or radiation therapy (RT). This approach is useful in a palliative setting as well. If impending organ damage is suspected (eg, spinal cord compression or superior vena cava obstruction), RT should be initiated promptly.

RT is a particularly effective modality as MCC is a radiosensitive tumor. The dose is typically delivered in multiple (between 5 and 20) fractions. One palliative option that has been reported to be well tolerated for symptomatic tumor masses is single-fraction radiotherapy. In a retrospective analysis of 93 tumors treated with single-fraction radiotherapy, objective response rate was 94 percent with complete responses in 45 percent [88]. Other palliative RT options such as brachytherapy have been reported as well [89].

EXPERIMENTAL APPROACHES — Newer approaches to advanced disease are required, given that checkpoint inhibitor immunotherapy only provides a durable benefit in approximately one-half of patients and chemotherapy offers short durations of response. Patients should be enrolled in clinical studies whenever possible.

Pazopanib – The tyrosine kinase inhibitor pazopanib has activity against metastatic MCC in at least one report [90]. In a phase II trial (UKMCC-01), pazopanib demonstrated clinical benefit in 9 of 16 patients with metastatic MCC (56 percent): 3 of 16 with partial responses and 6 of 16 with stable disease [91,92].

Somatostatin receptor-based therapy – Like other neuroendocrine tumors, MCC possesses receptors for somatostatin. These receptors can be demonstrated in vivo by somatostatin receptor-based diagnostic imaging (indium-111 pentetreotide single-photon emission computed tomography [SPECT; OctreoScan] or gallium Ga-68 DOTATATE positron emission tomography [PET] scanning) [93-97]. Peptide receptor radionuclide therapy with lutetium-177 isotope has been successfully used in other neuroendocrine tumors, and MCC also responds to the lutetium treatment [97,98].

Somatostatin analogs (octreotide and lanreotide) have been studied in MCC [93-96,99]. As an example, in one case report, a patient with metastatic MCC had a complete response to treatment with octreotide after receptors were demonstrated using indium-111 pentetreotide [96]. In another study of patients whose metastatic MCC was treated with somatostatin analogs, among seven patients who had a response-evaluable target lesion, three patients (43 percent) experienced progression-free survival of more than 120 days (median 237 days). This is a favorable outcome compared with historical data of chemotherapy, with median progression-free survival of approximately 90 days [87]. (See "Metastatic well-differentiated gastroenteropancreatic neuroendocrine tumors: Presentation, prognosis, imaging, and biochemical monitoring", section on 'Somatostatin receptor-based imaging techniques' and "Metastatic well-differentiated gastrointestinal neuroendocrine (carcinoid) tumors: Systemic therapy options to control tumor growth", section on 'Radiolabeled somatostatin analogs'.)

SUMMARY AND RECOMMENDATIONS

Prognostic factors for Merkel cell carcinoma (MCC) – MCC is an aggressive cutaneous malignancy that has a high propensity for regional spread via the lymphatics and for distant metastasis. Prognostic factors include stage, Merkel cell polyomavirus (MCPyV) status, and immune competence, among other clinical factors. A multidisciplinary consultation at a center with significant MCC expertise, including dermatology, medical oncology, radiation oncology, and surgery, is in the best interest of the patient. (See 'Prognostic factors' above.)

Initial evaluation and management – For a patient with a biopsy-confirmed cutaneous MCC, the initial evaluation should focus on determining whether there is clinical evidence of lymph node involvement or distant metastasis. Baseline determination of Merkel virus oncoprotein antibody titer can assist with prognosis and subsequent detection of recurrent disease. Imaging studies are important because of the metastatic potential of this cancer. (See 'Initial evaluation and management' above.)

Indications for sentinel lymph node (SLN) biopsy – For patients without evidence of regional lymph node or distant metastases, SLN mapping and biopsy are generally indicated to rule out occult disease. (See 'Approach to initial management' above and 'Clinically negative nodes' above.)

Positive SLN biopsy – For patients with a positive SLN biopsy, careful evaluation for occult metastatic disease is indicated (including appropriate imaging if not already done). If no distant metastatic disease is detected, treatment of the regional lymph node basin is indicated with either nodal dissection or definitive radiation therapy (RT). Adjuvant RT is offered after node dissection for multiple nodal involvement or extranodal extension.

Surgery – For patients without regional lymphatic or distant metastases, we suggest excision of the primary lesion with a margin of 1 to 2 cm as a component of initial treatment (Grade 2C). (See 'Primary tumor' above and 'Surgery' above.)

Adjuvant radiation – For patients who undergo surgical resection and are at high risk for local or regional recurrence, we suggest adjuvant RT, rather than observation (Grade 2C). High-risk factors include a primary tumor ≥1 cm in maximum dimension, positive or limited surgical resection margins, lymphovascular invasion, or an immunocompromised host. Adjuvant nodal RT is indicated if there is a high risk of anatomic, technical, or histologic failure (previous wide local excision, immunohistochemistry of the SLN not performed). (See 'Adjuvant therapy' above.)

For patients not at high risk for locoregional recurrence, observation may be an alternative to adjuvant RT following surgery.

Definitive RT – For patients who are not surgical candidates or for those in whom wide excision of the primary tumor is not feasible, definitive RT is an alternative to surgery. (See 'Primary tumor' above and 'Definitive radiation therapy' above.)

Clinical lymph node involvement – For patients suspected to have regional lymph node involvement based upon clinical evaluation, biopsy confirmation (fine needle aspirate, core biopsy, or excisional biopsy) is necessary. (See 'Approach to initial management' above and 'Regional lymph nodes' above and 'Clinically positive nodes' above.)

If regional lymph node disease is pathologically confirmed and there is no evidence of distant metastasis, we suggest definitive surgery (Grade 2C). Adjuvant RT may also be indicated. (See 'Clinically positive nodes' above and 'Adjuvant radiation therapy' above.)

If regional lymph node disease cannot be confirmed by appropriate pathologic examination (eg, confirmatory nodal biopsy is not feasible), we suggest adjuvant RT to the suspicious node (Grade 2C).

Patients at high risk for recurrence following definitive therapy of the primary tumor and regional lymph nodes should be considered for enrollment in formal clinical trials. (See 'Adjuvant immunotherapy' above.)

Surveillance – Following initial definitive therapy, patients with MCC should have frequent follow-up due to the high incidence of recurrent disease and evolving options for effective therapy. Decisions regarding follow-up frequency after initial management can be informed by accurate risk data for a given patient, via a web-based risk calculator tool. (See 'Surveillance after initial therapy' above.)

Recurrent locoregional disease – Patients with an isolated locoregional recurrence should be carefully reevaluated to definitively exclude the presence of disseminated disease. If no distant metastases are identified, patients should be treated with an individualized multimodality salvage treatment. (See 'Recurrent disease' above.)

Initial management of metastatic disease (immunotherapy) – For patients with metastatic disease, we suggest initial treatment with a programmed cell death ligand 1 (PD-L1)-blocking agent (avelumab) or programmed cell death protein 1 (PD-1) inhibitor-based immunotherapy (pembrolizumab, nivolumab) rather than chemotherapy (Grade 2C). Whenever possible, patients with advanced MCC should be referred for participation in PD-1/PD-L1 inhibitor-based immunotherapy trials. (See 'Metastatic disease' above and 'Immunotherapy' above.)

Subsequent management of metastatic disease – For patients with metastatic disease who progress on or are not candidates for checkpoint inhibitor immunotherapy, systemic chemotherapy may provide palliative benefit, although its impact on overall survival (OS) is uncertain. Most contemporary experience has used a regimen of a platinum (cisplatin, carboplatin) plus etoposide, as is used in small cell carcinoma of the lung. (See 'Chemotherapy' above and "Extensive-stage small cell lung cancer: Initial management", section on 'Initial treatment'.)

REFERENCES

  1. AJCC Cancer Staging Manual, 8th ed, Springer, New York 2016. p.240.
  2. Moshiri AS, Nghiem P. Milestones in the staging, classification, and biology of Merkel cell carcinoma. J Natl Compr Canc Netw 2014; 12:1255.
  3. Harms KL, Healy MA, Nghiem P, et al. Analysis of Prognostic Factors from 9387 Merkel Cell Carcinoma Cases Forms the Basis for the New 8th Edition AJCC Staging System. Ann Surg Oncol 2016; 23:3564.
  4. Lemos BD, Storer BE, Iyer JG, et al. Pathologic nodal evaluation improves prognostic accuracy in Merkel cell carcinoma: analysis of 5823 cases as the basis of the first consensus staging system. J Am Acad Dermatol 2010; 63:751.
  5. Farley CR, Perez MC, Soelling SJ, et al. Merkel Cell Carcinoma Outcomes: Does AJCC8 Underestimate Survival? Ann Surg Oncol 2020; 27:1978.
  6. Sihto H, Kukko H, Koljonen V, et al. Merkel cell polyomavirus infection, large T antigen, retinoblastoma protein and outcome in Merkel cell carcinoma. Clin Cancer Res 2011; 17:4806.
  7. Moshiri AS, Doumani R, Yelistratova L, et al. Polyomavirus-Negative Merkel Cell Carcinoma: A More Aggressive Subtype Based on Analysis of 282 Cases Using Multimodal Tumor Virus Detection. J Invest Dermatol 2017; 137:819.
  8. Paulson KG, Lewis CW, Redman MW, et al. Viral oncoprotein antibodies as a marker for recurrence of Merkel cell carcinoma: A prospective validation study. Cancer 2017; 123:1464.
  9. Tai PT, Yu E, Tonita J, Gilchrist J. Merkel cell carcinoma of the skin. J Cutan Med Surg 2000; 4:186.
  10. McEvoy A, Lachance K, Hippe D. Addressing the missing components for appropriate surveillance of Merkel cell carcinoma patients: Detailed recurrence data and an integrated risk analysis approach. J Invest Dermatol 2019; 139(5):S99.
  11. George A, Girault S, Testard A, et al. The impact of (18)F-FDG-PET/CT on Merkel cell carcinoma management: a retrospective study of 66 scans from a single institution. Nucl Med Commun 2014; 35:282.
  12. Hawryluk EB, O'Regan KN, Sheehy N, et al. Positron emission tomography/computed tomography imaging in Merkel cell carcinoma: a study of 270 scans in 97 patients at the Dana-Farber/Brigham and Women's Cancer Center. J Am Acad Dermatol 2013; 68:592.
  13. Siva S, Byrne K, Seel M, et al. 18F-FDG PET provides high-impact and powerful prognostic stratification in the staging of Merkel cell carcinoma: a 15-year institutional experience. J Nucl Med 2013; 54:1223.
  14. Poulsen M, Macfarlane D, Veness M, et al. Prospective analysis of the utility of 18-FDG PET in Merkel cell carcinoma of the skin: A Trans Tasman Radiation Oncology Group Study, TROG 09:03. J Med Imaging Radiat Oncol 2018; 62:412.
  15. Singh N, Alexander NA, Lachance K, et al. Clinical benefit of baseline imaging in Merkel cell carcinoma: Analysis of 584 patients. J Am Acad Dermatol 2021; 84:330.
  16. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Merkel cell carcinoma. Available at: https://www.nccn.org/professionals/physician_gls/pdf/mcc.pdf (Accessed on October 19, 2020).
  17. Harrington C, Kwan W. Outcomes of Merkel cell carcinoma treated with radiotherapy without radical surgical excision. Ann Surg Oncol 2014; 21:3401.
  18. Veness M, Howle J. Radiotherapy alone in patients with Merkel cell carcinoma: the Westmead Hospital experience of 41 patients. Australas J Dermatol 2015; 56:19.
  19. Fang LC, Lemos B, Douglas J, et al. Radiation monotherapy as regional treatment for lymph node-positive Merkel cell carcinoma. Cancer 2010; 116:1783.
  20. Tai P, Yu E, Assouline A, et al. Management of Merkel cell carcinoma with emphasis on small primary tumors--a case series and review of the current literature. J Drugs Dermatol 2010; 9:105.
  21. Jouary T, Leyral C, Dreno B, et al. Adjuvant prophylactic regional radiotherapy versus observation in stage I Merkel cell carcinoma: a multicentric prospective randomized study. Ann Oncol 2012; 23:1074.
  22. National Comprehensive Cancer Network (NCCN) clinical practice guidelines in oncology. http://www.nccn.org/professionals/physician_gls/f_guidelines.asp (Accessed on October 27, 2020).
  23. Veness M, Foote M, Gebski V, Poulsen M. The role of radiotherapy alone in patients with merkel cell carcinoma: reporting the Australian experience of 43 patients. Int J Radiat Oncol Biol Phys 2010; 78:703.
  24. Sundaresan P, Hruby G, Hamilton A, et al. Definitive radiotherapy or chemoradiotherapy in the treatment of Merkel cell carcinoma. Clin Oncol (R Coll Radiol) 2012; 24:e131.
  25. Becker JC, Stang A, DeCaprio JA, et al. Merkel cell carcinoma. Nat Rev Dis Primers 2017; 3:17077.
  26. Lebbe C, Becker JC, Grob JJ, et al. Diagnosis and treatment of Merkel Cell Carcinoma. European consensus-based interdisciplinary guideline. Eur J Cancer 2015; 51:2396.
  27. NCCN Guidelines https://www.nccn.org/professionals/physician_gls/default.aspx (Accessed on March 29, 2020).
  28. Harrington C, Kwan W. Radiotherapy and Conservative Surgery in the Locoregional Management of Merkel Cell Carcinoma: The British Columbia Cancer Agency Experience. Ann Surg Oncol 2016; 23:573.
  29. Tarabadkar ES, Fu T, Lachance K, et al. Narrow excision margins are appropriate for Merkel cell carcinoma when combined with adjuvant radiation: Analysis of 188 cases of localized disease and proposed management algorithm. J Am Acad Dermatol 2021; 84:340.
  30. O'Connor WJ, Roenigk RK, Brodland DG. Merkel cell carcinoma. Comparison of Mohs micrographic surgery and wide excision in eighty-six patients. Dermatol Surg 1997; 23:929.
  31. Boyer JD, Zitelli JA, Brodland DG, D'Angelo G. Local control of primary Merkel cell carcinoma: review of 45 cases treated with Mohs micrographic surgery with and without adjuvant radiation. J Am Acad Dermatol 2002; 47:885.
  32. Snow SN, Larson PO, Hardy S, et al. Merkel cell carcinoma of the skin and mucosa: report of 12 cutaneous cases with 2 cases arising from the nasal mucosa. Dermatol Surg 2001; 27:165.
  33. Pape E, Rezvoy N, Penel N, et al. Radiotherapy alone for Merkel cell carcinoma: a comparative and retrospective study of 25 patients. J Am Acad Dermatol 2011; 65:983.
  34. Cheraghlou S, Agogo GO, Girardi M. Evaluation of Lymph Node Ratio Association With Long-term Patient Survival After Surgery for Node-Positive Merkel Cell Carcinoma. JAMA Dermatol 2019; 155:803.
  35. Gunaratne DA, Howle JR, Veness MJ. Sentinel lymph node biopsy in Merkel cell carcinoma: a 15-year institutional experience and statistical analysis of 721 reported cases. Br J Dermatol 2016; 174:273.
  36. Strom T, Carr M, Zager JS, et al. Radiation Therapy is Associated with Improved Outcomes in Merkel Cell Carcinoma. Ann Surg Oncol 2016; 23:3572.
  37. Chen MM, Roman SA, Sosa JA, Judson BL. The role of adjuvant therapy in the management of head and neck merkel cell carcinoma: an analysis of 4815 patients. JAMA Otolaryngol Head Neck Surg 2015; 141:137.
  38. Bhatia S, Storer BE, Iyer JG, et al. Adjuvant Radiation Therapy and Chemotherapy in Merkel Cell Carcinoma: Survival Analyses of 6908 Cases From the National Cancer Data Base. J Natl Cancer Inst 2016; 108.
  39. Allen PJ, Bowne WB, Jaques DP, et al. Merkel cell carcinoma: prognosis and treatment of patients from a single institution. J Clin Oncol 2005; 23:2300.
  40. Clark JR, Veness MJ, Gilbert R, et al. Merkel cell carcinoma of the head and neck: is adjuvant radiotherapy necessary? Head Neck 2007; 29:249.
  41. Lawenda BD, Arnold MG, Tokarz VA, et al. Analysis of radiation therapy for the control of Merkel cell carcinoma of the head and neck based on 36 cases and a literature review. Ear Nose Throat J 2008; 87:634.
  42. Gillenwater AM, Hessel AC, Morrison WH, et al. Merkel cell carcinoma of the head and neck: effect of surgical excision and radiation on recurrence and survival. Arch Otolaryngol Head Neck Surg 2001; 127:149.
  43. Morand GB, Madana J, Da Silva SD, et al. Merkel cell carcinoma of the head and neck: poorer prognosis than non-head and neck sites. J Laryngol Otol 2016; 130:393.
  44. Takagishi SR, Marx TE, Lewis C, et al. Postoperative radiation therapy is associated with a reduced risk of local recurrence among low risk Merkel cell carcinomas of the head and neck. Adv Radiat Oncol 2016; 1:244.
  45. Bishop AJ, Garden AS, Gunn GB, et al. Merkel cell carcinoma of the head and neck: Favorable outcomes with radiotherapy. Head Neck 2016; 38 Suppl 1:E452.
  46. Vandeven N, Lewis CW, Makarov V, et al. Merkel Cell Carcinoma Patients Presenting Without a Primary Lesion Have Elevated Markers of Immunity, Higher Tumor Mutation Burden, and Improved Survival. Clin Cancer Res 2018; 24:963.
  47. Tarantola TI, Vallow LA, Halyard MY, et al. Unknown primary Merkel cell carcinoma: 23 new cases and a review. J Am Acad Dermatol 2013; 68:433.
  48. Chen KT, Papavasiliou P, Edwards K, et al. A better prognosis for Merkel cell carcinoma of unknown primary origin. Am J Surg 2013; 206:752.
  49. Foote M, Veness M, Zarate D, Poulsen M. Merkel cell carcinoma: the prognostic implications of an occult primary in stage IIIB (nodal) disease. J Am Acad Dermatol 2012; 67:395.
  50. Deneve JL, Messina JL, Marzban SS, et al. Merkel cell carcinoma of unknown primary origin. Ann Surg Oncol 2012; 19:2360.
  51. Song Y, Zheng C, Shannon AB, et al. Sentinel lymph node positivity and overall survival in immunosuppressed patients with Merkel cell carcinoma: a national cohort study. Br J Dermatol 2020; 183:569.
  52. Hruby G, Scolyer RA, Thompson JF. The important role of radiation treatment in the management of Merkel cell carcinoma. Br J Dermatol 2013; 169:975.
  53. Foote M, Harvey J, Porceddu S, et al. Effect of radiotherapy dose and volume on relapse in Merkel cell cancer of the skin. Int J Radiat Oncol Biol Phys 2010; 77:677.
  54. Poulsen MG, Rischin D, Porter I, et al. Does chemotherapy improve survival in high-risk stage I and II Merkel cell carcinoma of the skin? Int J Radiat Oncol Biol Phys 2006; 64:114.
  55. Poulsen M, Rischin D, Walpole E, et al. High-risk Merkel cell carcinoma of the skin treated with synchronous carboplatin/etoposide and radiation: a Trans-Tasman Radiation Oncology Group Study--TROG 96:07. J Clin Oncol 2003; 21:4371.
  56. Kaufman HL, Russell J, Hamid O, et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol 2016; 17:1374.
  57. D'Angelo SP, Russell J, Lebbé C, et al. Efficacy and Safety of First-line Avelumab Treatment in Patients With Stage IV Metastatic Merkel Cell Carcinoma: A Preplanned Interim Analysis of a Clinical Trial. JAMA Oncol 2018; 4:e180077.
  58. Poulsen M, Walpole E, Harvey J, et al. Weekly carboplatin reduces toxicity during synchronous chemoradiotherapy for Merkel cell carcinoma of skin. Int J Radiat Oncol Biol Phys 2008; 72:1070.
  59. Topalian SL, Bhatia S, Amin A, et al. Neoadjuvant Nivolumab for Patients With Resectable Merkel Cell Carcinoma in the CheckMate 358 Trial. J Clin Oncol 2020; 38:2476.
  60. Merkel cell disease recurrence. https://www.merkelcell.org/prognosis/disease-recurrence/ (Accessed on April 08, 2019).
  61. Tai PT, Yu E, Winquist E, et al. Chemotherapy in neuroendocrine/Merkel cell carcinoma of the skin: case series and review of 204 cases. J Clin Oncol 2000; 18:2493.
  62. Paulson KG, Carter JJ, Johnson LG, et al. Antibodies to merkel cell polyomavirus T antigen oncoproteins reflect tumor burden in merkel cell carcinoma patients. Cancer Res 2010; 70:8388.
  63. Pastrana DV, Wieland U, Silling S, et al. Positive correlation between Merkel cell polyomavirus viral load and capsid-specific antibody titer. Med Microbiol Immunol 2012; 201:17.
  64. Lyngaa R, Pedersen NW, Schrama D, et al. T-cell responses to oncogenic merkel cell polyomavirus proteins distinguish patients with merkel cell carcinoma from healthy donors. Clin Cancer Res 2014; 20:1768.
  65. Fleury MJ, Nicol JT, Samimi M, et al. Identification of the neutralizing epitopes of Merkel cell polyomavirus major capsid protein within the BC and EF surface loops. PLoS One 2015; 10:e0121751.
  66. Samimi M, Molet L, Fleury M, et al. Prognostic value of antibodies to Merkel cell polyomavirus T antigens and VP1 protein in patients with Merkel cell carcinoma. Br J Dermatol 2016; 174:813.
  67. Lachance K, Akaike T, Cahill K, et al. 590 Detecting Merkel cell carcinoma recurrence using a blood test: Outcomes from 774 patients. J Invest Dermatol 2019; 139:S101.
  68. Merkel cell disease recurrence. https://www.merkelcell.org/prognosis/disease-recurrence/ (Accessed on March 29, 2020).
  69. Goessling W, McKee PH, Mayer RJ. Merkel cell carcinoma. J Clin Oncol 2002; 20:588.
  70. Yiengpruksawan A, Coit DG, Thaler HT, et al. Merkel cell carcinoma. Prognosis and management. Arch Surg 1991; 126:1514.
  71. Pitale M, Sessions RB, Husain S. An analysis of prognostic factors in cutaneous neuroendocrine carcinoma. Laryngoscope 1992; 102:244.
  72. Eng TY, Naguib M, Fuller CD, et al. Treatment of recurrent Merkel cell carcinoma: an analysis of 46 cases. Am J Clin Oncol 2004; 27:576.
  73. Smith DE, Bielamowicz S, Kagan AR, et al. Cutaneous neuroendocrine (Merkel cell) carcinoma. A report of 35 cases. Am J Clin Oncol 1995; 18:199.
  74. Pilotti S, Rilke F, Bartoli C, Grisotti A. Clinicopathologic correlations of cutaneous neuroendocrine Merkel cell carcinoma. J Clin Oncol 1988; 6:1863.
  75. Fenig E, Brenner B, Katz A, et al. The role of radiation therapy and chemotherapy in the treatment of Merkel cell carcinoma. Cancer 1997; 80:881.
  76. Fenig E, Lurie H, Sulkes A. The use of cyclophosphamide, methotrexate, and 5-fluorouracil in the treatment of Merkel cell carcinoma. Am J Clin Oncol 1993; 16:54.
  77. Localized radiation therapy or recombinant interferon beta and avelumab with or without cellular adoptive immunotherapy in treating patients with metastatic Merkel cell carcinoma. (NCT 02584829). https://clinicaltrials.gov/ct2/show/NCT02584829?term=02584829&rank=1 (Accessed on April 08, 2019).
  78. Kaufman HL, Russell JS, Hamid O, et al. Updated efficacy of avelumab in patients with previously treated metastatic Merkel cell carcinoma after ≥1 year of follow-up: JAVELIN Merkel 200, a phase 2 clinical trial. J Immunother Cancer 2018; 6:7.
  79. Nghiem P, Bhatia S, Brohl AS, et al. Two-year efficacy and safety update from JAVELIN Merkel 200 part A: A registrational study of avelumab in metastatic Merkel cell carcinoma progressed on chemotherapy. J Clin Oncol 2018; 36S: ASCO #9507.
  80. Avelumab injection. United States Prescribing Information. US National Library of Medicine. Available online at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761049s000lbl.pdf (Accessed on April 08, 2019).
  81. Nghiem P, Bhatia S, Lipson EJ, et al. Durable Tumor Regression and Overall Survival in Patients With Advanced Merkel Cell Carcinoma Receiving Pembrolizumab as First-Line Therapy. J Clin Oncol 2019; 37:693.
  82. Nghiem PT, Bhatia S, Lipson EJ, et al. PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N Engl J Med 2016; 374:2542.
  83. Topalian SL, Bhatia S, Hollebecque A, et al. Non-comparative, open-label, multiple cohort, phase 1/2 study to evaluate nivolumab (NIVO) in patients with virus-associated tumors (CheckMate 358): Efficacy and safety in Merkel cell carcinoma (MCC). Cancer Res 2017; 77S: AACR #CT074.
  84. Becker JC, Hassel JC, Menzer C, et al. Adjuvant ipilimumab compared with observation in completely resected Merkel cell carcinoma (ADMEC): A randomized, multicenter DeCOG/ADO study. J Clin Oncol 2018; 36S:ASCO #9527.
  85. Winkler JK, Dimitrakopoulou-Strauss A, Sachpekidis C, et al. Ipilimumab has efficacy in metastatic Merkel cell carcinoma: a case series of five patients. J Eur Acad Dermatol Venereol 2017; 31:e389.
  86. LoPiccolo J, Schollenberger MD, Dakhil S, et al. Rescue therapy for patients with anti-PD-1-refractory Merkel cell carcinoma: a multicenter, retrospective case series. J Immunother Cancer 2019; 7:170.
  87. Iyer JG, Blom A, Doumani R, et al. Response rates and durability of chemotherapy among 62 patients with metastatic Merkel cell carcinoma. Cancer Med 2016; 5:2294.
  88. Iyer JG, Parvathaneni U, Gooley T, et al. Single-fraction radiation therapy in patients with metastatic Merkel cell carcinoma. Cancer Med 2015; 4:1161.
  89. Garibyan L, Cotter SE, Hansen JL, et al. Palliative treatment for in-transit cutaneous metastases of Merkel cell carcinoma using surface-mold computer-optimized high-dose-rate brachytherapy. Cancer J 2013; 19:283.
  90. Davids MS, Charlton A, Ng SS, et al. Response to a novel multitargeted tyrosine kinase inhibitor pazopanib in metastatic Merkel cell carcinoma. J Clin Oncol 2009; 27:e97.
  91. A trial of pazopanib for Merkel cell skin cancer (UKMCC-01). www.cancerresearchuk.org/about-cancer/find-a-clinical-trial/a-trial-of-pazopanib-for-merkel-cell-carcinoma-ukmcc-01 (Accessed on April 08, 2019).
  92. Nathan PD, Gaunt P, Wheatley K, et al. UKMCC-01: A phase II study of pazopanib (PAZ) in metastatic Merkel cell carcinoma. J Clin Oncol 2016; 34S: ASCO #9542.
  93. Meier G, Waldherr C, Herrmann R, et al. Successful targeted radiotherapy with 90Y-DOTATOC in a patient with Merkel cell carcinoma. A Case Report. Oncology 2004; 66:160.
  94. Kau R, Arnold W. Somatostatin receptor scintigraphy and therapy of neuroendocrine (APUD) tumors of the head and neck. Acta Otolaryngol 1996; 116:345.
  95. 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 Group. Cancer 1996; 77:402.
  96. Cirillo F, Filippini L, Lima GF, et al. [Merkel cell tumor. Report of case and treatment with octreotide]. Minerva Chir 1997; 52:1359.
  97. Salavati A, Prasad V, Schneider CP, et al. Peptide receptor radionuclide therapy of Merkel cell carcinoma using (177)lutetium-labeled somatostatin analogs in combination with radiosensitizing chemotherapy: a potential novel treatment based on molecular pathology. Ann Nucl Med 2012; 26:365.
  98. Basu S, Ranade R. Favorable Response of Metastatic Merkel Cell Carcinoma to Targeted 177Lu-DOTATATE Therapy: Will PRRT Evolve to Become an Important Approach in Receptor-Positive Cases? J Nucl Med Technol 2016; 44:85.
  99. Fakiha M, Letertre P, Vuillez JP, Lebeau J. Remission of Merkel cell tumor after somatostatin analog treatment. J Cancer Res Ther 2010; 6:382.
Topic 7609 Version 58.0

References

1 : AJCC Cancer Staging Manual, 8th ed, Springer, New York 2016. p.240.

2 : Milestones in the staging, classification, and biology of Merkel cell carcinoma.

3 : Analysis of Prognostic Factors from 9387 Merkel Cell Carcinoma Cases Forms the Basis for the New 8th Edition AJCC Staging System.

4 : Pathologic nodal evaluation improves prognostic accuracy in Merkel cell carcinoma: analysis of 5823 cases as the basis of the first consensus staging system.

5 : Merkel Cell Carcinoma Outcomes: Does AJCC8 Underestimate Survival?

6 : Merkel cell polyomavirus infection, large T antigen, retinoblastoma protein and outcome in Merkel cell carcinoma.

7 : Polyomavirus-Negative Merkel Cell Carcinoma: A More Aggressive Subtype Based on Analysis of 282 Cases Using Multimodal Tumor Virus Detection.

8 : Viral oncoprotein antibodies as a marker for recurrence of Merkel cell carcinoma: A prospective validation study.

9 : Merkel cell carcinoma of the skin.

10 : Addressing the missing components for appropriate surveillance of Merkel cell carcinoma patients: Detailed recurrence data and an integrated risk analysis approach

11 : The impact of (18)F-FDG-PET/CT on Merkel cell carcinoma management: a retrospective study of 66 scans from a single institution.

12 : Positron emission tomography/computed tomography imaging in Merkel cell carcinoma: a study of 270 scans in 97 patients at the Dana-Farber/Brigham and Women's Cancer Center.

13 : 18F-FDG PET provides high-impact and powerful prognostic stratification in the staging of Merkel cell carcinoma: a 15-year institutional experience.

14 : Prospective analysis of the utility of 18-FDG PET in Merkel cell carcinoma of the skin: A Trans Tasman Radiation Oncology Group Study, TROG 09:03.

15 : Clinical benefit of baseline imaging in Merkel cell carcinoma: Analysis of 584 patients.

16 : Clinical benefit of baseline imaging in Merkel cell carcinoma: Analysis of 584 patients.

17 : Outcomes of Merkel cell carcinoma treated with radiotherapy without radical surgical excision.

18 : Radiotherapy alone in patients with Merkel cell carcinoma: the Westmead Hospital experience of 41 patients.

19 : Radiation monotherapy as regional treatment for lymph node-positive Merkel cell carcinoma.

20 : Management of Merkel cell carcinoma with emphasis on small primary tumors--a case series and review of the current literature.

21 : Adjuvant prophylactic regional radiotherapy versus observation in stage I Merkel cell carcinoma: a multicentric prospective randomized study.

22 : Adjuvant prophylactic regional radiotherapy versus observation in stage I Merkel cell carcinoma: a multicentric prospective randomized study.

23 : The role of radiotherapy alone in patients with merkel cell carcinoma: reporting the Australian experience of 43 patients.

24 : Definitive radiotherapy or chemoradiotherapy in the treatment of Merkel cell carcinoma.

25 : Merkel cell carcinoma.

26 : Diagnosis and treatment of Merkel Cell Carcinoma. European consensus-based interdisciplinary guideline.

27 : Diagnosis and treatment of Merkel Cell Carcinoma. European consensus-based interdisciplinary guideline.

28 : Radiotherapy and Conservative Surgery in the Locoregional Management of Merkel Cell Carcinoma: The British Columbia Cancer Agency Experience.

29 : Narrow excision margins are appropriate for Merkel cell carcinoma when combined with adjuvant radiation: Analysis of 188 cases of localized disease and proposed management algorithm.

30 : Merkel cell carcinoma. Comparison of Mohs micrographic surgery and wide excision in eighty-six patients.

31 : Local control of primary Merkel cell carcinoma: review of 45 cases treated with Mohs micrographic surgery with and without adjuvant radiation.

32 : Merkel cell carcinoma of the skin and mucosa: report of 12 cutaneous cases with 2 cases arising from the nasal mucosa.

33 : Radiotherapy alone for Merkel cell carcinoma: a comparative and retrospective study of 25 patients.

34 : Evaluation of Lymph Node Ratio Association With Long-term Patient Survival After Surgery for Node-Positive Merkel Cell Carcinoma.

35 : Sentinel lymph node biopsy in Merkel cell carcinoma: a 15-year institutional experience and statistical analysis of 721 reported cases.

36 : Radiation Therapy is Associated with Improved Outcomes in Merkel Cell Carcinoma.

37 : The role of adjuvant therapy in the management of head and neck merkel cell carcinoma: an analysis of 4815 patients.

38 : Adjuvant Radiation Therapy and Chemotherapy in Merkel Cell Carcinoma: Survival Analyses of 6908 Cases From the National Cancer Data Base.

39 : Merkel cell carcinoma: prognosis and treatment of patients from a single institution.

40 : Merkel cell carcinoma of the head and neck: is adjuvant radiotherapy necessary?

41 : Analysis of radiation therapy for the control of Merkel cell carcinoma of the head and neck based on 36 cases and a literature review.

42 : Merkel cell carcinoma of the head and neck: effect of surgical excision and radiation on recurrence and survival.

43 : Merkel cell carcinoma of the head and neck: poorer prognosis than non-head and neck sites.

44 : Postoperative radiation therapy is associated with a reduced risk of local recurrence among low risk Merkel cell carcinomas of the head and neck.

45 : Merkel cell carcinoma of the head and neck: Favorable outcomes with radiotherapy.

46 : Merkel Cell Carcinoma Patients Presenting Without a Primary Lesion Have Elevated Markers of Immunity, Higher Tumor Mutation Burden, and Improved Survival.

47 : Unknown primary Merkel cell carcinoma: 23 new cases and a review.

48 : A better prognosis for Merkel cell carcinoma of unknown primary origin.

49 : Merkel cell carcinoma: the prognostic implications of an occult primary in stage IIIB (nodal) disease.

50 : Merkel cell carcinoma of unknown primary origin.

51 : Sentinel lymph node positivity and overall survival in immunosuppressed patients with Merkel cell carcinoma: a national cohort study.

52 : The important role of radiation treatment in the management of Merkel cell carcinoma.

53 : Effect of radiotherapy dose and volume on relapse in Merkel cell cancer of the skin.

54 : Does chemotherapy improve survival in high-risk stage I and II Merkel cell carcinoma of the skin?

55 : High-risk Merkel cell carcinoma of the skin treated with synchronous carboplatin/etoposide and radiation: a Trans-Tasman Radiation Oncology Group Study--TROG 96:07.

56 : Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial.

57 : Efficacy and Safety of First-line Avelumab Treatment in Patients With Stage IV Metastatic Merkel Cell Carcinoma: A Preplanned Interim Analysis of a Clinical Trial.

58 : Weekly carboplatin reduces toxicity during synchronous chemoradiotherapy for Merkel cell carcinoma of skin.

59 : Neoadjuvant Nivolumab for Patients With Resectable Merkel Cell Carcinoma in the CheckMate 358 Trial.

60 : Neoadjuvant Nivolumab for Patients With Resectable Merkel Cell Carcinoma in the CheckMate 358 Trial.

61 : Chemotherapy in neuroendocrine/Merkel cell carcinoma of the skin: case series and review of 204 cases.

62 : Antibodies to merkel cell polyomavirus T antigen oncoproteins reflect tumor burden in merkel cell carcinoma patients.

63 : Positive correlation between Merkel cell polyomavirus viral load and capsid-specific antibody titer.

64 : T-cell responses to oncogenic merkel cell polyomavirus proteins distinguish patients with merkel cell carcinoma from healthy donors.

65 : Identification of the neutralizing epitopes of Merkel cell polyomavirus major capsid protein within the BC and EF surface loops.

66 : Prognostic value of antibodies to Merkel cell polyomavirus T antigens and VP1 protein in patients with Merkel cell carcinoma.

67 : 590 Detecting Merkel cell carcinoma recurrence using a blood test: Outcomes from 774 patients

68 : 590 Detecting Merkel cell carcinoma recurrence using a blood test: Outcomes from 774 patients

69 : Merkel cell carcinoma.

70 : Merkel cell carcinoma. Prognosis and management.

71 : An analysis of prognostic factors in cutaneous neuroendocrine carcinoma.

72 : Treatment of recurrent Merkel cell carcinoma: an analysis of 46 cases.

73 : Cutaneous neuroendocrine (Merkel cell) carcinoma. A report of 35 cases.

74 : Clinicopathologic correlations of cutaneous neuroendocrine Merkel cell carcinoma.

75 : The role of radiation therapy and chemotherapy in the treatment of Merkel cell carcinoma.

76 : The use of cyclophosphamide, methotrexate, and 5-fluorouracil in the treatment of Merkel cell carcinoma.

77 : The use of cyclophosphamide, methotrexate, and 5-fluorouracil in the treatment of Merkel cell carcinoma.

78 : Updated efficacy of avelumab in patients with previously treated metastatic Merkel cell carcinoma after≥1 year of follow-up: JAVELIN Merkel 200, a phase 2 clinical trial.

79 : Two-year efficacy and safety update from JAVELIN Merkel 200 part A: A registrational study of avelumab in metastatic Merkel cell carcinoma progressed on chemotherapy

80 : Two-year efficacy and safety update from JAVELIN Merkel 200 part A: A registrational study of avelumab in metastatic Merkel cell carcinoma progressed on chemotherapy

81 : Durable Tumor Regression and Overall Survival in Patients With Advanced Merkel Cell Carcinoma Receiving Pembrolizumab as First-Line Therapy.

82 : PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma.

83 : Non-comparative, open-label, multiple cohort, phase 1/2 study to evaluate nivolumab (NIVO) in patients with virus-associated tumors (CheckMate 358): Efficacy and safety in Merkel cell carcinoma (MCC)

84 : Adjuvant ipilimumab compared with observation in completely resected Merkel cell carcinoma (ADMEC): A randomized, multicenter DeCOG/ADO study.

85 : Ipilimumab has efficacy in metastatic Merkel cell carcinoma: a case series of five patients.

86 : Rescue therapy for patients with anti-PD-1-refractory Merkel cell carcinoma: a multicenter, retrospective case series.

87 : Response rates and durability of chemotherapy among 62 patients with metastatic Merkel cell carcinoma.

88 : Single-fraction radiation therapy in patients with metastatic Merkel cell carcinoma.

89 : Palliative treatment for in-transit cutaneous metastases of Merkel cell carcinoma using surface-mold computer-optimized high-dose-rate brachytherapy.

90 : Response to a novel multitargeted tyrosine kinase inhibitor pazopanib in metastatic Merkel cell carcinoma.

91 : Response to a novel multitargeted tyrosine kinase inhibitor pazopanib in metastatic Merkel cell carcinoma.

92 : UKMCC-01: A phase II study of pazopanib (PAZ) in metastatic Merkel cell carcinoma.

93 : Successful targeted radiotherapy with 90Y-DOTATOC in a patient with Merkel cell carcinoma. A Case Report.

94 : Somatostatin receptor scintigraphy and therapy of neuroendocrine (APUD) tumors of the head and neck.

95 : Clinical efficacy of octreotide in the treatment of metastatic neuroendocrine tumors. A study by the Italian Trials in Medical Oncology Group.

96 : [Merkel cell tumor. Report of case and treatment with octreotide].

97 : Peptide receptor radionuclide therapy of Merkel cell carcinoma using (177)lutetium-labeled somatostatin analogs in combination with radiosensitizing chemotherapy: a potential novel treatment based on molecular pathology.

98 : Favorable Response of Metastatic Merkel Cell Carcinoma to Targeted 177Lu-DOTATATE Therapy: Will PRRT Evolve to Become an Important Approach in Receptor-Positive Cases?

99 : Remission of Merkel cell tumor after somatostatin analog treatment.