Your activity: 43602 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: [email protected]

Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma

Pathogenesis, clinical features, and diagnosis of Merkel cell (neuroendocrine) carcinoma
Authors:
Patricia Tai, MB, BS, FRCPC
Paul T Nghiem, MD, PhD
Song Youn Park, MD
Section Editors:
Robert S Stern, MD
June K Robinson, MD
Deputy Editors:
Rosamaria Corona, MD, DSc
Sonali Shah, MD
Literature review current through: Feb 2022. | This topic last updated: Apr 02, 2021.

INTRODUCTION — Merkel cell carcinoma (MCC) of the skin is a rare, aggressive, cutaneous malignancy that predominantly affects older adults with light skin types and has a high propensity for recurrence and metastases [1].

The pathogenesis, clinical features, and diagnosis of MCC are discussed here. The staging and treatment of patients with MCC are discussed separately. (See "Staging, treatment, and surveillance of Merkel cell carcinoma".)

EPIDEMIOLOGY

Incidence — Multiple studies report increasing incidence of Merkel cell carcinoma (MCC) in several countries over the past decades [2-9]. Data from the Surveillance, Epidemiology, and End Results (SEER) Program database indicate that in the United States, the estimated annual incidence rate rose from 0.5 cases per 100,000 persons in 2000 (95% CI 0.4-0.5) to 0.7 cases per 100,000 persons in 2013 (95% CI 0.7-0.8). MCC incidence increases exponentially with advancing age, from 0.1 to 1 to 9.8 (per 100,000 person-years) among age groups 40 to 44, 60 to 64, and >85 years, respectively. Due to aging of the "baby boomer" generation, the United States' MCC incidence is predicted to climb to 2835 cases in 2020 and 3284 cases in 2025 [2].

Risk factors — Recognized risk factors for MCC include:

Light skin color – MCC predominantly affects persons with light skin [10,11]. A study analyzing SEER data from 1973 to 2006 found that 95 percent of cases diagnosed during this interval arose in the White population [10].

Increasing age – MCC is typically seen in older individuals, with a mean age at diagnosis of 76 years for women and 74 years for men [10,11]. MCC incidence increases in advanced age groups, with an almost 10-fold increase every 20 years of increasing age [11].

Male sex – The incidence of MCC is higher in men than in women, in general. In an analysis of United States cancer registries including the National Program of Cancer Registries, the reported incidence rate was 4.2 and 1.9 per 100,000 persons for men and women, respectively, and the male to female ratio was 2.2 (95% CI 2.1-2.4) in the United States White population [9].

Immunosuppression – The incidence of MCC is more frequent in immunosuppressed patients, such as organ transplant recipients [12-17], HIV-infected individuals [18,19], and those with hematologic malignancies [11,12,20]. MCC occurs at a younger age and higher incidence in immunosuppressed subjects [11-14,18-20]. Data from the United States Scientific Registry of Transplant Recipients and 15 population-based cancer registries indicate that transplant recipients have a 24-fold higher risk of MCC than immunocompetent patients [12]. The highest incidence occurs 10 or more years after transplant. Maintenance treatment with mammalian (mechanistic) target of rapamycin (mTOR) inhibitors and azathioprine or cyclosporine confers a higher risk of MCC [12]. (See 'Immunosuppression' below.)

Other malignancies – The risk of MCC is significantly increased in patients with other malignancies, especially hematologic malignancies [20,21]. In a SEER database study of over two million patients with cancer, the risk of MCC was significantly increased in those with multiple myeloma, chronic lymphocytic leukemia, and malignant melanoma (standardized incidence ratio [SIR] 3.7, 6.9, and 3.1, respectively) [20]. A retrospective study of 4164 patients with chronic lymphocytic leukemia and 172 patients with MCC supported the association between these two disorders reported in the SEER database study [22]. Among patients with a diagnosis of chronic lymphocytic leukemia, the incidence of a subsequent diagnosis of MCC was greatly elevated (SIR 15.7, 95% CI 3.2-46). Patients with a preceding diagnosis of MCC also exhibited an increased risk for developing chronic lymphocytic leukemia (SIR 17.9, 95% CI 2.2-64.6).

HISTOGENESIS — Based upon early histologic and ultrastructural studies, Merkel cell carcinoma (MCC) has been traditionally believed to arise from Merkel cells, which are located in the basal layer of the epidermis and hair follicles and are associated with sensory neurites in the dermal papillae, the skin mechanoreceptors [23]. However, this hypothesis is controversial. An alternative hypothesis is that these tumors originate from an immature totipotential stem cell that acquires neuroendocrine features during malignant transformation [24]. Based on cancer genomic studies and an understanding of the two different etiologies of MCC (Merkel cell polyomavirus [MCPyV]-positive or -negative MCCs), it is plausible that MCC tumors might not have a single cell of origin. MCPyV-positive MCCs might arise from dermal fibroblasts and MCPyV-negative MCCs from epidermal keratinocytes, but this is a highly controversial area [25,26]. (See 'Merkel cell polyomavirus' below.)

PATHOGENESIS — Several factors have been associated with the development of Merkel cell carcinoma (MCC). These include infection with the Merkel cell polyomavirus (MCPyV), ultraviolet (UV) radiation exposure, and immunosuppression. (See 'Risk factors' above.)

Merkel cell polyomavirus — Merkel cell polyomavirus (MCPyV) is a nonenveloped, double-stranded DNA virus that has been causally linked to the development of MCC (picture 1) [27-30]. Previous studies have consistently reported that MCPyV can be detected in approximately 80 and 60 percent of all MCCs by real-time polymerase chain reaction (PCR) and immunohistochemistry, respectively [31]. A 2015 meta-analysis of 23 studies found an overall MCPyV prevalence of 79 percent (95% CI 72-84 percent) in Merkel cell tumors versus 12 percent (95% CI 8-9 percent) in control skin samples [32].

MCPyV is a ubiquitous virus, with five specific geographically related genotypic variants [33]. MCPyV is thought to be part of the human skin microbiome and appears to be chronically shed from the skin in the form of assembled virions [34]. The seroprevalence of antibodies specific to the capsid viral protein 1 (VP1) appears to increase with age, from approximately 40 percent in children to up to over 80 percent in older individuals [30,35-37].

The observation that the integration of the virus into the tumor genome precedes the clonal expansion of tumor cells suggests that the virus is present at tumor initiation and that one or more viral proteins are oncogenic drivers [27,38]. MCPyV encodes two main oncoproteins, the large tumor (LT) antigen and small tumor (sT) antigen, which are both persistently expressed in MCC and have been implicated in oncogenesis through multiple mechanisms. Sequential events involved in the pathogenesis of MCPyV-positive tumors include clonal integration into the host genome, expression of sT, acquisition of mutations in the 3' end of LT, and evasion of a destructive immune response [1,39,40].

Mutations in LT result in a truncated molecule that lacks the DNA binding domain and the helicase domain, which renders the virus replication incompetent while preserving its retinoblastoma (RB1) gene binding property and the expression of the oncogenic viral proteins [41]. LT antigens with intact RB1 binding domain sequester and inactivate the tumor suppressor, resulting in sustained tumor growth.

MCPyV has been detected in normal-appearing skin and in other tumors, including cutaneous squamous cell carcinoma, chronic lymphocytic leukemia, and folliculotropic mycosis fungoides [42-48]. In one series of patients with MCC, skin samples taken from sites distant from the tumor were positive for the virus in 10 out of 14 cases (71 percent), a frequency higher than that was observed in six skin samples from normal individuals (17 percent) [42]. Importantly, outside of MCC, evidence of clonal integration of the virus and expression of viral proteins in tumor cells is almost invariably lacking in other cancers, suggesting that the occasional detection of the virus was likely coincidental rather than causal in non-MCC tumors [49].

Ultraviolet radiation — Ultraviolet (UV) radiation exposure, especially to the sun, is thought to play an important role in the etiology of many cases of MCC. The following observations are consistent with an etiologic role for UV radiation exposure:

MCC has a predilection for sun-exposed areas. In a study of the clinical manifestations in 195 cases of MCC, the tumor arose in a UV-exposed region in 81 percent of assessable cases [11]. Furthermore, 98 percent of cases occurred in fair-skinned individuals.

Regional incidence rates correlate with increasing sun exposure, as measured by the ultraviolet B (UVB) solar index [7,8,50,51].

Presentation with other skin cancers, for which sun exposure is a major risk factor, is common [21,52]. (See "Epidemiology, pathogenesis, and clinical features of basal cell carcinoma" and "Cutaneous squamous cell carcinoma: Epidemiology, risk factors, and molecular pathogenesis".)

MCC has been described in patients treated with psoralen plus ultraviolet A (PUVA) photochemotherapy [53].

Multiple reports describe UVB signature mutations in MCC tumors [54-57].

UV radiation exposure may be especially relevant in the pathogenesis of the virus-negative subtype of MCC. This hypothesis is supported by the observation that in Australia the incidence of MCC is highest, but the prevalence of MCPyV-positive tumors is much lower than in other geographic areas. An Australian study of 95 MCC reported a 23 percent prevalence of MCPyV positivity; the prevalence was even lower in tumors occurring in skin areas with evidence of sun damage (ie, elastosis, squamous dysplasia) [58].

Immunosuppression — The incidence of MCC is increased in immunosuppressed patients, including organ transplant recipients [12-17], HIV-infected individuals [18,19], and those with hematologic malignancies [11,12,20]. (See "Epidemiology and risk factors for skin cancer in solid organ transplant recipients", section on 'Merkel cell carcinoma' and "HIV infection and malignancy: Management considerations", section on 'Merkel cell carcinoma'.)

The mechanism by which immunosuppression interacts with MCPyV and UV radiation exposure in the pathogenesis of MCC is unknown. Immunosuppression may facilitate the replication of MCPyV and increase the chance of virus integration in the MCC progenitor cell. In addition, reduced immune surveillance may contribute to the survival and proliferation of atypical cells. Finally, immunosuppressive agents, such as azathioprine or calcineurin inhibitors, have been shown to act synergistically with UV radiation in inducing mutagenesis and promoting skin carcinogenesis in an immunosuppression-independent manner [59-63].

Virus-negative Merkel cell carcinoma — Some MCCs have low or negligible levels of MCPyV T antigen expression. The mechanisms of oncogenesis underlying MCPyV-negative MCC are incompletely understood, but they are thought to involve somatic mutations in tumor suppressor genes such as TP53 and retinoblastoma (RB1), as well as epigenetic alterations, such as DNA methylation and microRNAs, resulting in aberrant expression and activity of oncogenes [41,64-67]. Mutations in TP53 (encoding p53) occur infrequently in MCCs and have been found almost exclusively in tumors with low or complete lack of MCPyV LT antigen expression [56,68]. The expression of retinoblastoma protein (RB), a key cell cycle regulator, is low or absent in viral-negative tumors, whereas nearly all MCPyV-positive tumors are also RB-positive [68].

The frequency of TP53 mutations, either UV signature mutations or non-UV signature mutations, appears to be higher in MCPyV-negative MCCs than in MCPyV-positive tumors [69]. An analysis of MCPyV-positive and MCPyV-negative MCCs using next-generation sequencing found an overall high mutational burden in MCPyV-negative tumors compared with MCPyV-positive tumors [56]. In MCPyV-negative tumors, the majority of tandem substitutions were CC > TT substitutions, characteristic of UV mutational signature, whereas only one CC > TT tandem substitution was detected across all MCPyV-positive tumors. Activating oncogenic mutations in HRAS, PIK3CA, KNSTRN, PREX2, and RAC1 were found in six of eight MCPyV-negative tumors but only in two of eight MCPyV-positive tumors. Highly recurrent mutations in tumor suppressor genes, including TP53, RBI, NOTCH1, and PRUNE2, were also found in MCPyV-negative tumors. These findings suggest that genetic aberrations independent of MCPyV infection are involved in the pathogenesis of viral-negative MCCs.

CLINICAL FEATURES — Merkel cell carcinoma (MCC) typically presents in older patients with light skin tones as a rapidly growing, firm, nontender, shiny, flesh-colored or bluish-red, intracutaneous nodule (picture 2A-C). Ulceration and crusting are infrequent. MCCs range in size from less than 1 cm to over 2 cm and are most often located in sun-exposed areas.

In an analysis of 9387 MCC cases from the National Cancer Database between 1998 and 2012 (median age 76 years), the most frequent anatomic locations for the primary tumor were the following [70]:

Head and neck: 43 percent

Upper limbs and shoulder: 24 percent

Lower limbs and hip: 15 percent

Trunk: 11 percent

Other areas: 9 percent

In this series, patients presented with local disease only in 65 percent of cases, while 26 percent had regional lymph node involvement at presentation, and 8 percent had distant metastases; 3.6 percent had lymph node involvement with an unknown primary tumor and without distant metastases. In these patients, the primary tumor may have spontaneously regressed [71-74].

DIAGNOSIS — Merkel cell carcinoma (MCC) is often clinically misdiagnosed as a benign lesion (eg, cyst, lipoma, pyogenic granuloma) [11]. A high index of suspicion is required if the diagnosis is to be made without delay. An analysis of data from the Surveillance, Epidemiology, and End Results (SEER) registry from 1973 to 2014 on over 3400 patients with MCC found that younger age (<65 years), male sex, and location of the tumor on the trunk or extremities were all associated with late-stage disease at diagnosis [75].

Clinical — Lesion features that suggest a biopsy may be appropriate to evaluate for a nonmelanoma skin cancer, such as MCC, include a rapidly growing, nontender lesion on sun-exposed skin in an older person with lightly pigmented skin, particularly if chronic immune suppression is present. These features can be easily remembered by the acronym AEIOU, derived from a series of 195 cases seen over a period of 27 years [11]:

Asymptomatic: 88 percent

Expanding rapidly (significant growth in ≤3 months): 63 percent

Immune suppression (HIV infection, solid organ transplant recipient, chronic lymphocytic leukemia): 8 percent

Older than 50 years age: 90 percent

Ultraviolet (UV)-exposed area in a fair-skinned individual: 81 percent

The presence of at least three of these features increases the suspicion of MCC. Lesion biopsy and histologic examination are necessary to establish the diagnosis. (See 'Histopathology' below.)

Dermoscopy — Data on the dermoscopic findings of MCC are limited, and features specific to a diagnosis of MCC have not been identified. Some of the dermoscopic findings reported in small, retrospective series include milky red areas; linear, irregular vessels; and polymorphous vessels (picture 3) [76-79]. However, dermoscopy is unlikely to be broadly useful for the clinical diagnosis of MCC due to a lack of specific features, the rarity of MCC, and the need for a highly expert dermoscopist to potentially recognize features suspicious of MCC. (See "Overview of dermoscopy".)

Histopathology — Both routine studies with hematoxylin and eosin as well as immunohistochemical stains are usually required to distinguish MCC from other poorly differentiated tumors. MCC typically presents as a dermal mass that frequently extends into the subcutis. The epidermis is infrequently involved, and the overlying skin is rarely ulcerated.

The tumor is composed of strands or nests of monotonously uniform, round, blue cells, containing large basophilic nuclei with powdery dispersed chromatin and inconspicuous nucleoli, and minimal cytoplasm [80]. Other features may include single-cell necrosis, frequent mitoses, lymphovascular invasion, perineural invasion, and epidermal involvement via pagetoid spread.

There are three main histologic patterns, which have not been demonstrated to have prognostic or therapeutic implications but may be helpful in differentiating MCC from other entities (picture 4) [80]:

Intermediate type – This variant, which is the most common, shows large, solid nodules made of diffuse sheets of basophilic cells with the characteristic round to oval nucleus, powdery chromatin, and inconspicuous nucleoli.

Small cell type – The small cell variant has small, round cells with scant cytoplasm, oval hyperchromatic nuclei, and prominent nucleoli. The tumor cells form a solid sheet or clusters, often with crush artifact and nuclear molding.

Trabecular type – This variant, which is the least common, has round to polygonal cells with abundant cytoplasm; round, centrally located vesicular nuclei; and inconspicuous nucleoli arranged in an organoid, trabecular, or ribbon-like arrangement.

Ultrastructurally, MCC tumor cells, like normal Merkel cells, contain paranuclear electron-dense neurosecretory granules (dense core granules), 10 mm filaments, and desmosomes [81].

Combined Merkel cell carcinoma — MCC may rarely show squamous, eccrine, glandular, and melanocytic differentiation [82,83]. The occurrence of focal areas of squamous or sarcomatous differentiation in MCC has been documented in several patients [84,85]. The histogenesis and pathogenesis of these tumors are incompletely understood. They are Merkel cell polyomavirus (MCPyV)-negative and have high frequency of retinoblastoma (RB1) and p53 mutations [85].

Immunohistochemistry — On immunohistochemistry, the Merkel cells show features of both epithelial and neuroendocrine cells [81]. They express epithelial markers, such as AE1/AE3, CAM 5.2, pan-cytokeratin, epithelial membrane antigen, and Ber-EP4, and may stain for various neuroendocrine markers, including chromogranin, synaptophysin, calcitonin, vasoactive intestinal peptide, and somatostatin receptor [86].

Immunoreactivity for low-molecular-weight cytokeratins (eg, CK20, CK5/6) distinguishes MCC from other undifferentiated tumors (table 1) [87]. MCC consistently stains positively for low-molecular-weight CK20, which is a fairly specific and sensitive marker for MCC, with a characteristic paranuclear dot-like positivity (picture 5) [80].

MCPyV large tumor (LT) antigen expression can be detected by using the commercially available mouse monoclonal antibody CM2B4 (picture 1). The LT antigen expression is highly correlated with the viral load (amount of MCPyV DNA) measured by real-time polymerase chain reaction [31].

Cytogenetic and molecular analysis — Multiple chromosome abnormalities, including gains, losses, and rearrangements, have been detected in MCC. However, the relationship of these genetic changes to pathogenesis, natural history, and therapeutic outcomes is unclear.

The chromosomes most frequently affected are 1, 5, 6, 8, and 13 [41,88-90]. The pattern of chromosome gains and losses is similar to those seen in small cell cancer of the lung (SCLC) [89] (see "Pathobiology and staging of small cell carcinoma of the lung"). Structural abnormalities involving chromosome 1p have been noted in up to 40 percent of examined cases [91].

Somatic mutations in tumor suppressor genes, including RB1 and TP53, have been documented in subsets of MCCs. Mutations in TP53 have been found in 0 to 28 percent of MCCs, the majority of which represent single nucleotide polymorphisms or silent mutations of unknown clinical significance [41]. RB1-inactivating mutations occur in nearly all MCPyV-negative MCCs.

Other molecular alterations found with varying frequency in MCC include mutations in genes involved in the tyrosine kinase signaling (PIK3CA, KIT, and PDGFR) and microRNA expression [92,93]. Distinct expression profiles of microRNA (noncoding RNA sequences that silence translation of complementary messenger RNA transcripts) have been found in MCPyV-positive and -negative tumors, suggesting distinct molecular mechanisms in tumor development and progression based upon MCPyV status [94].

DIFFERENTIAL DIAGNOSIS — Clinically, Merkel cell carcinoma (MCC) may closely mimic many benign and malignant lesions occurring on sun-exposed skin, such as basal cell carcinoma, squamous cell carcinoma, keratoacanthoma, amelanotic melanoma, pyogenic granuloma, lipoma, and adnexal tumors.

Histology can clarify the diagnosis in most cases. However, on conventional light microscopy, MCC is difficult to distinguish from other poorly differentiated cutaneous and noncutaneous "small, blue cell tumors" [95,96]. Immunohistochemistry is required for a definitive diagnosis (table 1) [81,97,98] (see 'Immunohistochemistry' above):

Small cell carcinoma of the lung – In contrast to MCC, cutaneous metastases of small cell carcinoma of the lung do not stain with CK20 but are positive for CK7, neuron-specific enolase, and thyroid transcription factor-1 (table 1) [87,99]. (See "Pathobiology and staging of small cell carcinoma of the lung", section on 'Pathology'.)

Small cell melanoma – Melanoma with small cell phenotype is a rare variant of melanoma that resembles MCC [100,101]. Positive staining for HMB-45, Melan-A, and S-100 protein confirms the diagnosis of melanoma. (See "Pathologic characteristics of melanoma", section on 'Rare variants'.)

PROGNOSTIC FACTORS — The extent of disease at presentation, and in particular the involvement of regional lymph nodes, is the most important predictor of survival for Merkel cell carcinoma (MCC). However, several clinicopathologic features may have an influence on prognosis independent of stage [102]. (See "Staging, treatment, and surveillance of Merkel cell carcinoma", section on 'Prognostic factors'.)

Histologic features and viral status — Several histologic features and the tumor Merkel cell polyomavirus (MCPyV) status may have a prognostic significance:

Lymphovascular invasion – In a retrospective series of 156 patients with MCC, when clinical stage was incorporated into a multivariate analysis, only lymphovascular invasion and infiltrative, rather than nodular, growth pattern were associated with a poorer prognosis (odds ratios [ORs] for death 3.8 and 6.9, respectively) [103]. In another retrospective study of 500 MCC patients, age, pathologic stage at diagnosis, and lymphovascular invasion were independent determinants of prognosis. Patients with lymphovascular invasion of primary tumor had a twofold increased risk of death compared with those without lymphovascular invasion [104].

Tumor-infiltrating lymphocytes – Tumor-infiltrating lymphocytes (TILs), also known as intratumoral CD8+ lymphocyte infiltration, may be associated with favorable survival outcome [105,106]. In a multivariate analysis of 2182 patients with nonmetastatic MCC, the presence of TILs was associated with improved overall survival, including TILs with either brisk (hazard ratio [HR] 0.50, 95% CI 0.34-0.74) or nonbrisk (HR 0.75, 95% CI 0.60-0.93) features [106].

MCPyV positivity – The presence of the MCPyV large tumor (LT) antigen and the expression of the retinoblastoma protein (RB) appear to be associated with a favorable prognosis. MCPyV DNA-positive tumors are preferentially located on the limbs and tend to metastasize less frequently compared with MCPyV DNA-negative MCCs [107]. In a study of 91 patients with MCC, of whom 61 were MCPyV DNA-positive, LT antigen expression was independently associated with better MCC-specific survival in a multivariate model that also included postsurgical stage (HR 0.22, 95% CI 0.09-0.52) [68]. In a study of 282 patients, 19 percent were MCPyV-negative, and this virus-negative subset had a significantly increased risk of death from MCC (HR 1.85, 95% CI 1.19-2.89) [108].

p63 expression – The expression of p63, a member of the p53 family, has been proposed as a prognostic marker for patients with low-stage MCC. In a study of 70 patients with MCC, p63 expression was independently associated with an increased risk of death (HR 7.26, 95% CI 2.069-25.506) [109]. Another study of 128 patients confirmed that p63 expression predicts a poorer survival (HR 2.05, 95% CI 1.1-3.8) but did not serve as an independent predictor if stage was considered [110].

Merkel cell polyomavirus serology — Multiple studies found that changes in the titer of antibodies that recognize MCPyV oncoproteins (small tumor antigen) reflect recent decreases or increases in a patient's MCC tumor burden [111-114]. This test can serve as an additional prognostic marker as well as an effective method of surveillance. Based on the clinical utility of this test, the National Comprehensive Cancer Network guidelines for MCC suggest that clinicians consider determining MCPyV oncoprotein antibody titer as part of the initial work-up of MCC and of ongoing surveillance for seropositive patients [115]:

Prognostic marker – The determination of oncoprotein antibody titer at baseline (ideally within three months of the initial treatment) can assist in the clinical management of MCC patients. In a cohort of 219 patients newly diagnosed with MCC and followed for more than five years, 114 (52 percent) were MCPyV-oncoprotein-seropositive at the time of diagnosis [116]. In this study, seropositivity at diagnosis was associated with a reduced risk of recurrence (HR 0.58, 95% CI 0.36-0.97), after adjusting for age, sex, stage, and immunosuppression. Thus, patients who are MCPyV-oncoprotein-seronegative at diagnosis may be at increased risk of recurrence and may benefit from more intensive surveillance using radiologic imaging [116].

Surveillance – For seropositive patients, MCPyV-oncoprotein antibody determination may be a useful component of ongoing surveillance, as a rising titer can be an early indicator of recurrence [115,116]. 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 (fraction of patients with rising titers that currently had or soon developed clinically evident MCC recurrence), and the negative predictive value was 99 percent (fraction of patients with stable or declining titers who did not have recurrent disease detectable at that time) [117]. In some cases, the antibody test outperformed imaging by identifying recurrences before they were detectable by imaging.

In clinical practice, a combination of the MCPyV oncoprotein antibody test and imaging studies can help guide surveillance so that is appropriate for MCC patients with varying levels of recurrence risk.

Immune status — The MCC-specific mortality rate is higher in patients with T cell immunosuppression (eg, HIV infection, solid organ transplant, chronic lymphocytic leukemia). In a cohort of 218 patients with MCC, those with immunosuppression had an increased risk of overall and MCC-specific mortality (HR 1.9, 95% CI 0.9-4 and HR 4.9, 95% CI 1.7-14.4, respectively) [72]. In another study of 87 patients with MCC, patients with chronic immunosuppression had an increased risk of death when compared with immune-competent individuals (HR 2.01, 95% CI 1.1-3.7) [118].

SUMMARY AND RECOMMENDATIONS

Merkel cell carcinoma (MCC) is a rare, cutaneous malignancy with a propensity for local recurrence and distant metastases. It predominantly affects older individuals with fair skin types. (See 'Introduction' above and 'Epidemiology' above.)

Multiple factors appear to contribute to the etiology of MCC, including the Merkel cell polyomavirus (MCPyV), ultraviolet (UV) radiation exposure, and immunosuppression. (See 'Pathogenesis' above.)

Patients with MCC typically present with a rapidly growing, painless, firm, nontender, shiny, flesh-colored or bluish-red, intracutaneous nodule commonly located in the head and neck region (picture 2A-C). (See 'Clinical features' above.)

The diagnosis of MCC requires a high index of suspicion, as it is often clinically misdiagnosed as a benign lesion (eg, cyst, lipoma, pyogenic granuloma). Clinical warning signs that should prompt a biopsy are summarized in the acronym AEIOU (asymptomatic lesion, expanding rapidly, immunosuppression, age older than 50 years, lesion on UV-exposed skin). The definitive diagnosis is made by histologic examination of a biopsy specimen and generally requires immunohistochemistry as well as routine staining to distinguish MCC from other poorly differentiated tumors (table 1). (See 'Diagnosis' above and 'Differential diagnosis' above.)

Patient's immune status, histologic features of tumor (eg, lymphovascular invasion, intratumoral lymphocyte infiltration), and MCPyV serology provide prognostic information. MCPyV serology can be used as a follow-up test for surveillance in MCPyV serology-positive patients. (See 'Prognostic factors' above.)

The staging and management of patients with MCC are discussed separately. (See "Staging, treatment, and surveillance of Merkel cell carcinoma".)

REFERENCES

  1. Tothill R, Estall V, Rischin D. Merkel cell carcinoma: emerging biology, current approaches, and future directions. Am Soc Clin Oncol Educ Book 2015; :e519.
  2. Paulson KG, Park SY, Vandeven NA, et al. Merkel cell carcinoma: Current US incidence and projected increases based on changing demographics. J Am Acad Dermatol 2018; 78:457.
  3. Zaar O, Gillstedt M, Lindelöf B, et al. Merkel cell carcinoma incidence is increasing in Sweden. J Eur Acad Dermatol Venereol 2016; 30:1708.
  4. Reichgelt BA, Visser O. Epidemiology and survival of Merkel cell carcinoma in the Netherlands. A population-based study of 808 cases in 1993-2007. Eur J Cancer 2011; 47:579.
  5. Fondain M, Dereure O, Uhry Z, et al. Merkel cell carcinoma in France: a registries-based, comprehensive epidemiological survey. J Eur Acad Dermatol Venereol 2018; 32:1292.
  6. Youlden DR, Soyer HP, Youl PH, et al. Incidence and survival for Merkel cell carcinoma in Queensland, Australia, 1993-2010. JAMA Dermatol 2014; 150:864.
  7. Robertson JP, Liang ES, Martin RC. Epidemiology of Merkel cell carcinoma in New Zealand: a population-based study. Br J Dermatol 2015; 173:835.
  8. Girschik J, Thorn K, Beer TW, et al. Merkel cell carcinoma in Western Australia: a population-based study of incidence and survival. Br J Dermatol 2011; 165:1051.
  9. Stang A, Becker JC, Nghiem P, Ferlay J. The association between geographic location and incidence of Merkel cell carcinoma in comparison to melanoma: An international assessment. Eur J Cancer 2018; 94:47.
  10. Albores-Saavedra J, Batich K, Chable-Montero F, et al. Merkel cell carcinoma demographics, morphology, and survival based on 3870 cases: a population based study. J Cutan Pathol 2010; 37:20.
  11. Heath M, Jaimes N, Lemos B, et al. Clinical characteristics of Merkel cell carcinoma at diagnosis in 195 patients: the AEIOU features. J Am Acad Dermatol 2008; 58:375.
  12. Clarke CA, Robbins HA, Tatalovich Z, et al. Risk of merkel cell carcinoma after solid organ transplantation. J Natl Cancer Inst 2015; 107.
  13. Penn I, First MR. Merkel's cell carcinoma in organ recipients: report of 41 cases. Transplantation 1999; 68:1717.
  14. Tadmor T, Aviv A, Polliack A. Merkel cell carcinoma, chronic lymphocytic leukemia and other lymphoproliferative disorders: an old bond with possible new viral ties. Ann Oncol 2011; 22:250.
  15. Bajetta E, Platania M, Catena L, et al. Merkel cell carcinoma after liver transplantation: a case report. Tumori 2007; 93:323.
  16. Bensaleh H, Perney P, Dereure O, et al. Merkel cell carcinoma in a liver transplant patient. Am J Clin Dermatol 2007; 8:239.
  17. Koljonen V, Kukko H, Tukiainen E, et al. Incidence of Merkel cell carcinoma in renal transplant recipients. Nephrol Dial Transplant 2009; 24:3231.
  18. Engels EA, Frisch M, Goedert JJ, et al. Merkel cell carcinoma and HIV infection. Lancet 2002; 359:497.
  19. Manganoni MA, Farisoglio C, Tucci G, et al. Merkel cell carcinoma and HIV infection: a case report and review of the literature. AIDS Patient Care STDS 2007; 21:447.
  20. Howard RA, Dores GM, Curtis RE, et al. Merkel cell carcinoma and multiple primary cancers. Cancer Epidemiol Biomarkers Prev 2006; 15:1545.
  21. Kaae J, Hansen AV, Biggar RJ, et al. Merkel cell carcinoma: incidence, mortality, and risk of other cancers. J Natl Cancer Inst 2010; 102:793.
  22. Koljonen V, Kukko H, Pukkala E, et al. Chronic lymphocytic leukaemia patients have a high risk of Merkel-cell polyomavirus DNA-positive Merkel-cell carcinoma. Br J Cancer 2009; 101:1444.
  23. Ratner D, Nelson BR, Brown MD, Johnson TM. Merkel cell carcinoma. J Am Acad Dermatol 1993; 29:143.
  24. Tilling T, Moll I. Which are the cells of origin in merkel cell carcinoma? J Skin Cancer 2012; 2012:680410.
  25. Sunshine JC, Jahchan NS, Sage J, Choi J. Are there multiple cells of origin of Merkel cell carcinoma? Oncogene 2018; 37:1409.
  26. Nirenberg A, Steinman H, Dixon J, Dixon A. Merkel cell carcinoma update: the case for two tumours. J Eur Acad Dermatol Venereol 2020; 34:1425.
  27. Feng H, Shuda M, Chang Y, Moore PS. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 2008; 319:1096.
  28. Kassem A, Schöpflin A, Diaz C, et al. Frequent detection of Merkel cell polyomavirus in human Merkel cell carcinomas and identification of a unique deletion in the VP1 gene. Cancer Res 2008; 68:5009.
  29. Sharp CP, Norja P, Anthony I, et al. Reactivation and mutation of newly discovered WU, KI, and Merkel cell carcinoma polyomaviruses in immunosuppressed individuals. J Infect Dis 2009; 199:398.
  30. Carter JJ, Paulson KG, Wipf GC, et al. Association of Merkel cell polyomavirus-specific antibodies with Merkel cell carcinoma. J Natl Cancer Inst 2009; 101:1510.
  31. Leroux-Kozal V, Lévêque N, Brodard V, et al. Merkel cell carcinoma: histopathologic and prognostic features according to the immunohistochemical expression of Merkel cell polyomavirus large T antigen correlated with viral load. Hum Pathol 2015; 46:443.
  32. Santos-Juanes J, Fernández-Vega I, Fuentes N, et al. Merkel cell carcinoma and Merkel cell polyomavirus: a systematic review and meta-analysis. Br J Dermatol 2015; 173:42.
  33. Martel-Jantin C, Filippone C, Tortevoye P, et al. Molecular epidemiology of merkel cell polyomavirus: evidence for geographically related variant genotypes. J Clin Microbiol 2014; 52:1687.
  34. Schowalter RM, Pastrana DV, Pumphrey KA, et al. Merkel cell polyomavirus and two previously unknown polyomaviruses are chronically shed from human skin. Cell Host Microbe 2010; 7:509.
  35. Viscidi RP, Rollison DE, Sondak VK, et al. Age-specific seroprevalence of Merkel cell polyomavirus, BK virus, and JC virus. Clin Vaccine Immunol 2011; 18:1737.
  36. Martel-Jantin C, Pedergnana V, Nicol JT, et al. Merkel cell polyomavirus infection occurs during early childhood and is transmitted between siblings. J Clin Virol 2013; 58:288.
  37. Chen T, Hedman L, Mattila PS, et al. Serological evidence of Merkel cell polyomavirus primary infections in childhood. J Clin Virol 2011; 50:125.
  38. Sastre-Garau X, Peter M, Avril MF, et al. Merkel cell carcinoma of the skin: pathological and molecular evidence for a causative role of MCV in oncogenesis. J Pathol 2009; 218:48.
  39. Church CD, Nghiem P. How does the Merkel polyomavirus lead to a lethal cancer? Many answers, many questions, and a new mouse model. J Invest Dermatol 2015; 135:1221.
  40. DeCaprio JA, Garcea RL. A cornucopia of human polyomaviruses. Nat Rev Microbiol 2013; 11:264.
  41. Erstad DJ, Jr JC. Mutational analysis of merkel cell carcinoma. Cancers (Basel) 2014; 6:2116.
  42. Foulongne V, Dereure O, Kluger N, et al. Merkel cell polyomavirus DNA detection in lesional and nonlesional skin from patients with Merkel cell carcinoma or other skin diseases. Br J Dermatol 2010; 162:59.
  43. Kassem A, Technau K, Kurz AK, et al. Merkel cell polyomavirus sequences are frequently detected in nonmelanoma skin cancer of immunosuppressed patients. Int J Cancer 2009; 125:356.
  44. Rollison DE, Giuliano AR, Messina JL, et al. Case-control study of Merkel cell polyomavirus infection and cutaneous squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev 2012; 21:74.
  45. Murakami M, Imajoh M, Ikawa T, et al. Presence of Merkel cell polyomavirus in Japanese cutaneous squamous cell carcinoma. J Clin Virol 2011; 50:37.
  46. Dworkin AM, Tseng SY, Allain DC, et al. Merkel cell polyomavirus in cutaneous squamous cell carcinoma of immunocompetent individuals. J Invest Dermatol 2009; 129:2868.
  47. Trizuljak J, Srovnal J, Plevová K, et al. Analysis of prognostic significance of merkel cell polyomavirus in chronic lymphocytic leukemia. Clin Lymphoma Myeloma Leuk 2015; 15:439.
  48. Du-Thanh A, Dereure O, Guillot B, Foulongne V. Merkel cell polyomavirus: its putative involvement in a particular subset of cutaneous lymphoma with possibly unfavorable outcome. J Clin Virol 2014; 61:161.
  49. Reisinger DM, Shiffer JD, Cognetta AB Jr, et al. Lack of evidence for basal or squamous cell carcinoma infection with Merkel cell polyomavirus in immunocompetent patients with Merkel cell carcinoma. J Am Acad Dermatol 2010; 63:400.
  50. Miller RW, Rabkin CS. Merkel cell carcinoma and melanoma: etiological similarities and differences. Cancer Epidemiol Biomarkers Prev 1999; 8:153.
  51. Agelli M, Clegg LX. Epidemiology of primary Merkel cell carcinoma in the United States. J Am Acad Dermatol 2003; 49:832.
  52. Zhan FQ, Packianathan VS, Zeitouni NC. Merkel cell carcinoma: a review of current advances. J Natl Compr Canc Netw 2009; 7:333.
  53. Lunder EJ, Stern RS. Merkel-cell carcinomas in patients treated with methoxsalen and ultraviolet A radiation. N Engl J Med 1998; 339:1247.
  54. Van Gele M, Kaghad M, Leonard JH, et al. Mutation analysis of P73 and TP53 in Merkel cell carcinoma. Br J Cancer 2000; 82:823.
  55. Popp S, Waltering S, Herbst C, et al. UV-B-type mutations and chromosomal imbalances indicate common pathways for the development of Merkel and skin squamous cell carcinomas. Int J Cancer 2002; 99:352.
  56. Harms PW, Vats P, Verhaegen ME, et al. The Distinctive Mutational Spectra of Polyomavirus-Negative Merkel Cell Carcinoma. Cancer Res 2015; 75:3720.
  57. Goh G, Walradt T, Markarov V, et al. Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget 2016; 7:3403.
  58. Dabner M, McClure RJ, Harvey NT, et al. Merkel cell polyomavirus and p63 status in Merkel cell carcinoma by immunohistochemistry: Merkel cell polyomavirus positivity is inversely correlated with sun damage, but neither is correlated with outcome. Pathology 2014; 46:205.
  59. Perrett CM, Walker SL, O'Donovan P, et al. Azathioprine treatment photosensitizes human skin to ultraviolet A radiation. Br J Dermatol 2008; 159:198.
  60. O'Donovan P, Perrett CM, Zhang X, et al. Azathioprine and UVA light generate mutagenic oxidative DNA damage. Science 2005; 309:1871.
  61. Han W, Soltani K, Ming M, He YY. Deregulation of XPC and CypA by cyclosporin A: an immunosuppression-independent mechanism of skin carcinogenesis. Cancer Prev Res (Phila) 2012; 5:1155.
  62. Han W, Ming M, He TC, He YY. Immunosuppressive cyclosporin A activates AKT in keratinocytes through PTEN suppression: implications in skin carcinogenesis. J Biol Chem 2010; 285:11369.
  63. Ming M, Zhao B, Qiang L, He YY. Effect of immunosuppressants tacrolimus and mycophenolate mofetil on the keratinocyte UVB response. Photochem Photobiol 2015; 91:242.
  64. Cimino PJ, Robirds DH, Tripp SR, et al. Retinoblastoma gene mutations detected by whole exome sequencing of Merkel cell carcinoma. Mod Pathol 2014; 27:1073.
  65. Harms PW, Patel RM, Verhaegen ME, et al. Distinct gene expression profiles of viral- and nonviral-associated merkel cell carcinoma revealed by transcriptome analysis. J Invest Dermatol 2013; 133:936.
  66. Greenberg ES, Chong KK, Huynh KT, et al. Epigenetic biomarkers in skin cancer. Cancer Lett 2014; 342:170.
  67. Veija T, Sahi H, Koljonen V, et al. miRNA-34a underexpressed in Merkel cell polyomavirus-negative Merkel cell carcinoma. Virchows Arch 2015; 466:289.
  68. 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.
  69. Higaki-Mori H, Kuwamoto S, Iwasaki T, et al. Association of Merkel cell polyomavirus infection with clinicopathological differences in Merkel cell carcinoma. Hum Pathol 2012; 43:2282.
  70. 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.
  71. Haymerle G, Fochtmann A, Kunstfeld R, et al. Management of Merkel cell carcinoma of unknown primary origin: the Vienna Medical School experience. Eur Arch Otorhinolaryngol 2015; 272:425.
  72. Asgari MM, Sokil MM, Warton EM, et al. Effect of host, tumor, diagnostic, and treatment variables on outcomes in a large cohort with Merkel cell carcinoma. JAMA Dermatol 2014; 150:716.
  73. Deneve JL, Messina JL, Marzban SS, et al. Merkel cell carcinoma of unknown primary origin. Ann Surg Oncol 2012; 19:2360.
  74. 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.
  75. Liu MA, Nguyen J, Driver JA. Influence of age and marital status on stage at diagnosis and survival of patients with Merkel cell carcinoma: A Surveillance, Epidemiology, and End Results registry-based cohort study. J Am Acad Dermatol 2018; 79:1146.
  76. Jalilian C, Chamberlain AJ, Haskett M, et al. Clinical and dermoscopic characteristics of Merkel cell carcinoma. Br J Dermatol 2013; 169:294.
  77. Harting MS, Ludgate MW, Fullen DR, et al. Dermatoscopic vascular patterns in cutaneous Merkel cell carcinoma. J Am Acad Dermatol 2012; 66:923.
  78. Dalle S, Parmentier L, Moscarella E, et al. Dermoscopy of Merkel cell carcinoma. Dermatology 2012; 224:140.
  79. Scalvenzi M, Palmisano F, Ilardi G, et al. Clinical, dermoscopic and histological features of a Merkel cell carcinoma of the hand. J Dermatol Case Rep 2013; 7:15.
  80. Wong HH, Wang J. Merkel cell carcinoma. Arch Pathol Lab Med 2010; 134:1711.
  81. Warner TF, Uno H, Hafez GR, et al. Merkel cells and Merkel cell tumors. Ultrastructure, immunocytochemistry and review of the literature. Cancer 1983; 52:238.
  82. Martin B, Poblet E, Rios JJ, et al. Merkel cell carcinoma with divergent differentiation: histopathological and immunohistochemical study of 15 cases with PCR analysis for Merkel cell polyomavirus. Histopathology 2013; 62:711.
  83. Saeb-Lima M, Montante-Montes de Oca D, Albores-Saavedra J. Merkel cell carcinoma with eccrine differentiation: a clinicopathologic study of 7 cases. Ann Diagn Pathol 2008; 12:410.
  84. Narisawa Y, Koba S, Inoue T, Nagase K. Histogenesis of pure and combined Merkel cell carcinomas: An immunohistochemical study of 14 cases. J Dermatol 2015; 42:445.
  85. Pulitzer MP, Brannon AR, Berger MF, et al. Cutaneous squamous and neuroendocrine carcinoma: genetically and immunohistochemically different from Merkel cell carcinoma. Mod Pathol 2015; 28:1023.
  86. Gardair C, Samimi M, Touzé A, et al. Somatostatin Receptors 2A and 5 Are Expressed in Merkel Cell Carcinoma with No Association with Disease Severity. Neuroendocrinology 2015; 101:223.
  87. Chan JK, Suster S, Wenig BM, et al. Cytokeratin 20 immunoreactivity distinguishes Merkel cell (primary cutaneous neuroendocrine) carcinomas and salivary gland small cell carcinomas from small cell carcinomas of various sites. Am J Surg Pathol 1997; 21:226.
  88. Leonard JH, Leonard P, Kearsley JH. Chromosomes 1, 11, and 13 are frequently involved in karyotypic abnormalities in metastatic Merkel cell carcinoma. Cancer Genet Cytogenet 1993; 67:65.
  89. Van Gele M, Speleman F, Vandesompele J, et al. Characteristic pattern of chromosomal gains and losses in Merkel cell carcinoma detected by comparative genomic hybridization. Cancer Res 1998; 58:1503.
  90. Sozzi G, Bertoglio MG, Pilotti S, et al. Cytogenetic studies in primary and metastatic neuroendocrine Merkel cell carcinoma. Cancer Genet Cytogenet 1988; 30:151.
  91. Van Gele M, Van Roy N, Ronan SG, et al. Molecular analysis of 1p36 breakpoints in two Merkel cell carcinomas. Genes Chromosomes Cancer 1998; 23:67.
  92. Swick BL, Srikantha R, Messingham KN. Specific analysis of KIT and PDGFR-alpha expression and mutational status in Merkel cell carcinoma. J Cutan Pathol 2013; 40:623.
  93. Iwasaki T, Matsushita M, Nonaka D, et al. Comparison of Akt/mTOR/4E-BP1 pathway signal activation and mutations of PIK3CA in Merkel cell polyomavirus-positive and Merkel cell polyomavirus-negative carcinomas. Hum Pathol 2015; 46:210.
  94. Xie H, Lee L, Caramuta S, et al. MicroRNA expression patterns related to merkel cell polyomavirus infection in human merkel cell carcinoma. J Invest Dermatol 2014; 134:507.
  95. Daoud MA, Mete O, Al Habeeb A, Ghazarian D. Neuroendocrine carcinoma of the skin--an updated review. Semin Diagn Pathol 2013; 30:234.
  96. Pulitzer MP, Amin BD, Busam KJ. Merkel cell carcinoma: review. Adv Anat Pathol 2009; 16:135.
  97. Sibley RK, Dehner LP, Rosai J. Primary neuroendocrine (Merkel cell?) carcinoma of the skin. I. A clinicopathologic and ultrastructural study of 43 cases. Am J Surg Pathol 1985; 9:95.
  98. Meyer-Pannwitt U, Kummerfeldt K, Boubaris P, Caselitz J. [Merkel cell tumor or neuroendocrine skin carcinoma]. Langenbecks Arch Chir 1997; 382:349.
  99. Leech SN, Kolar AJ, Barrett PD, et al. Merkel cell carcinoma can be distinguished from metastatic small cell carcinoma using antibodies to cytokeratin 20 and thyroid transcription factor 1. J Clin Pathol 2001; 54:727.
  100. Song HS, Kim YC. Small cell melanoma. Ann Dermatol 2014; 26:419.
  101. House NS, Fedok F, Maloney ME, Helm KF. Malignant melanoma with clinical and histologic features of Merkel cell carcinoma. J Am Acad Dermatol 1994; 31:839.
  102. Moshiri AS, Nghiem P. Milestones in the staging, classification, and biology of Merkel cell carcinoma. J Natl Compr Canc Netw 2014; 12:1255.
  103. Andea AA, Coit DG, Amin B, Busam KJ. Merkel cell carcinoma: histologic features and prognosis. Cancer 2008; 113:2549.
  104. Fields RC, Busam KJ, Chou JF, et al. Five hundred patients with Merkel cell carcinoma evaluated at a single institution. Ann Surg 2011; 254:465.
  105. Paulson KG, Iyer JG, Tegeder AR, et al. Transcriptome-wide studies of merkel cell carcinoma and validation of intratumoral CD8+ lymphocyte invasion as an independent predictor of survival. J Clin Oncol 2011; 29:1539.
  106. Butala AA, Jain V, Reddy VK, et al. Impact of Tumor-Infiltrating Lymphocytes on Overall Survival in Merkel Cell Carcinoma. Oncologist 2021; 26:63.
  107. Sihto H, Kukko H, Koljonen V, et al. Clinical factors associated with Merkel cell polyomavirus infection in Merkel cell carcinoma. J Natl Cancer Inst 2009; 101:938.
  108. 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.
  109. Asioli S, Righi A, de Biase D, et al. Expression of p63 is the sole independent marker of aggressiveness in localised (stage I-II) Merkel cell carcinomas. Mod Pathol 2011; 24:1451.
  110. Stetsenko GY, Malekirad J, Paulson KG, et al. p63 expression in Merkel cell carcinoma predicts poorer survival yet may have limited clinical utility. Am J Clin Pathol 2013; 140:838.
  111. 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.
  112. 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.
  113. 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.
  114. 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.
  115. National Comprehensive Cancer Network (NCCN). NCCN Clinical practice guidelines in oncology. www.nccn.org/professionals/physician_gls/pdf/neuroendocrine.pdf (Accessed on June 28, 2018).
  116. 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.
  117. 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.
  118. Liang E, Brower JV, Rice SR, et al. Merkel Cell Carcinoma Analysis of Outcomes: A 30-Year Experience. PLoS One 2015; 10:e0129476.
Topic 7623 Version 30.0

References

1 : Merkel cell carcinoma: emerging biology, current approaches, and future directions.

2 : Merkel cell carcinoma: Current US incidence and projected increases based on changing demographics.

3 : Merkel cell carcinoma incidence is increasing in Sweden.

4 : Epidemiology and survival of Merkel cell carcinoma in the Netherlands. A population-based study of 808 cases in 1993-2007.

5 : Merkel cell carcinoma in France: a registries-based, comprehensive epidemiological survey.

6 : Incidence and survival for Merkel cell carcinoma in Queensland, Australia, 1993-2010.

7 : Epidemiology of Merkel cell carcinoma in New Zealand: a population-based study.

8 : Merkel cell carcinoma in Western Australia: a population-based study of incidence and survival.

9 : The association between geographic location and incidence of Merkel cell carcinoma in comparison to melanoma: An international assessment.

10 : Merkel cell carcinoma demographics, morphology, and survival based on 3870 cases: a population based study.

11 : Clinical characteristics of Merkel cell carcinoma at diagnosis in 195 patients: the AEIOU features.

12 : Risk of merkel cell carcinoma after solid organ transplantation.

13 : Merkel's cell carcinoma in organ recipients: report of 41 cases.

14 : Merkel cell carcinoma, chronic lymphocytic leukemia and other lymphoproliferative disorders: an old bond with possible new viral ties.

15 : Merkel cell carcinoma after liver transplantation: a case report.

16 : Merkel cell carcinoma in a liver transplant patient.

17 : Incidence of Merkel cell carcinoma in renal transplant recipients.

18 : Merkel cell carcinoma and HIV infection.

19 : Merkel cell carcinoma and HIV infection: a case report and review of the literature.

20 : Merkel cell carcinoma and multiple primary cancers.

21 : Merkel cell carcinoma: incidence, mortality, and risk of other cancers.

22 : Chronic lymphocytic leukaemia patients have a high risk of Merkel-cell polyomavirus DNA-positive Merkel-cell carcinoma.

23 : Merkel cell carcinoma.

24 : Which are the cells of origin in merkel cell carcinoma?

25 : Are there multiple cells of origin of Merkel cell carcinoma?

26 : Merkel cell carcinoma update: the case for two tumours.

27 : Clonal integration of a polyomavirus in human Merkel cell carcinoma.

28 : Frequent detection of Merkel cell polyomavirus in human Merkel cell carcinomas and identification of a unique deletion in the VP1 gene.

29 : Reactivation and mutation of newly discovered WU, KI, and Merkel cell carcinoma polyomaviruses in immunosuppressed individuals.

30 : Association of Merkel cell polyomavirus-specific antibodies with Merkel cell carcinoma.

31 : Merkel cell carcinoma: histopathologic and prognostic features according to the immunohistochemical expression of Merkel cell polyomavirus large T antigen correlated with viral load.

32 : Merkel cell carcinoma and Merkel cell polyomavirus: a systematic review and meta-analysis.

33 : Molecular epidemiology of merkel cell polyomavirus: evidence for geographically related variant genotypes.

34 : Merkel cell polyomavirus and two previously unknown polyomaviruses are chronically shed from human skin.

35 : Age-specific seroprevalence of Merkel cell polyomavirus, BK virus, and JC virus.

36 : Merkel cell polyomavirus infection occurs during early childhood and is transmitted between siblings.

37 : Serological evidence of Merkel cell polyomavirus primary infections in childhood.

38 : Merkel cell carcinoma of the skin: pathological and molecular evidence for a causative role of MCV in oncogenesis.

39 : How does the Merkel polyomavirus lead to a lethal cancer? Many answers, many questions, and a new mouse model.

40 : A cornucopia of human polyomaviruses.

41 : Mutational analysis of merkel cell carcinoma.

42 : Merkel cell polyomavirus DNA detection in lesional and nonlesional skin from patients with Merkel cell carcinoma or other skin diseases.

43 : Merkel cell polyomavirus sequences are frequently detected in nonmelanoma skin cancer of immunosuppressed patients.

44 : Case-control study of Merkel cell polyomavirus infection and cutaneous squamous cell carcinoma.

45 : Presence of Merkel cell polyomavirus in Japanese cutaneous squamous cell carcinoma.

46 : Merkel cell polyomavirus in cutaneous squamous cell carcinoma of immunocompetent individuals.

47 : Analysis of prognostic significance of merkel cell polyomavirus in chronic lymphocytic leukemia.

48 : Merkel cell polyomavirus: its putative involvement in a particular subset of cutaneous lymphoma with possibly unfavorable outcome.

49 : Lack of evidence for basal or squamous cell carcinoma infection with Merkel cell polyomavirus in immunocompetent patients with Merkel cell carcinoma.

50 : Merkel cell carcinoma and melanoma: etiological similarities and differences.

51 : Epidemiology of primary Merkel cell carcinoma in the United States.

52 : Merkel cell carcinoma: a review of current advances.

53 : Merkel-cell carcinomas in patients treated with methoxsalen and ultraviolet A radiation.

54 : Mutation analysis of P73 and TP53 in Merkel cell carcinoma.

55 : UV-B-type mutations and chromosomal imbalances indicate common pathways for the development of Merkel and skin squamous cell carcinomas.

56 : The Distinctive Mutational Spectra of Polyomavirus-Negative Merkel Cell Carcinoma.

57 : Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy.

58 : Merkel cell polyomavirus and p63 status in Merkel cell carcinoma by immunohistochemistry: Merkel cell polyomavirus positivity is inversely correlated with sun damage, but neither is correlated with outcome.

59 : Azathioprine treatment photosensitizes human skin to ultraviolet A radiation.

60 : Azathioprine and UVA light generate mutagenic oxidative DNA damage.

61 : Deregulation of XPC and CypA by cyclosporin A: an immunosuppression-independent mechanism of skin carcinogenesis.

62 : Immunosuppressive cyclosporin A activates AKT in keratinocytes through PTEN suppression: implications in skin carcinogenesis.

63 : Effect of immunosuppressants tacrolimus and mycophenolate mofetil on the keratinocyte UVB response.

64 : Retinoblastoma gene mutations detected by whole exome sequencing of Merkel cell carcinoma.

65 : Distinct gene expression profiles of viral- and nonviral-associated merkel cell carcinoma revealed by transcriptome analysis.

66 : Epigenetic biomarkers in skin cancer.

67 : miRNA-34a underexpressed in Merkel cell polyomavirus-negative Merkel cell carcinoma.

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

69 : Association of Merkel cell polyomavirus infection with clinicopathological differences in Merkel cell carcinoma.

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

71 : Management of Merkel cell carcinoma of unknown primary origin: the Vienna Medical School experience.

72 : Effect of host, tumor, diagnostic, and treatment variables on outcomes in a large cohort with Merkel cell carcinoma.

73 : Merkel cell carcinoma of unknown primary origin.

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

75 : Influence of age and marital status on stage at diagnosis and survival of patients with Merkel cell carcinoma: A Surveillance, Epidemiology, and End Results registry-based cohort study.

76 : Clinical and dermoscopic characteristics of Merkel cell carcinoma.

77 : Dermatoscopic vascular patterns in cutaneous Merkel cell carcinoma.

78 : Dermoscopy of Merkel cell carcinoma.

79 : Clinical, dermoscopic and histological features of a Merkel cell carcinoma of the hand.

80 : Merkel cell carcinoma.

81 : Merkel cells and Merkel cell tumors. Ultrastructure, immunocytochemistry and review of the literature.

82 : Merkel cell carcinoma with divergent differentiation: histopathological and immunohistochemical study of 15 cases with PCR analysis for Merkel cell polyomavirus.

83 : Merkel cell carcinoma with eccrine differentiation: a clinicopathologic study of 7 cases.

84 : Histogenesis of pure and combined Merkel cell carcinomas: An immunohistochemical study of 14 cases.

85 : Cutaneous squamous and neuroendocrine carcinoma: genetically and immunohistochemically different from Merkel cell carcinoma.

86 : Somatostatin Receptors 2A and 5 Are Expressed in Merkel Cell Carcinoma with No Association with Disease Severity.

87 : Cytokeratin 20 immunoreactivity distinguishes Merkel cell (primary cutaneous neuroendocrine) carcinomas and salivary gland small cell carcinomas from small cell carcinomas of various sites.

88 : Chromosomes 1, 11, and 13 are frequently involved in karyotypic abnormalities in metastatic Merkel cell carcinoma.

89 : Characteristic pattern of chromosomal gains and losses in Merkel cell carcinoma detected by comparative genomic hybridization.

90 : Cytogenetic studies in primary and metastatic neuroendocrine Merkel cell carcinoma.

91 : Molecular analysis of 1p36 breakpoints in two Merkel cell carcinomas.

92 : Specific analysis of KIT and PDGFR-alpha expression and mutational status in Merkel cell carcinoma.

93 : Comparison of Akt/mTOR/4E-BP1 pathway signal activation and mutations of PIK3CA in Merkel cell polyomavirus-positive and Merkel cell polyomavirus-negative carcinomas.

94 : MicroRNA expression patterns related to merkel cell polyomavirus infection in human merkel cell carcinoma.

95 : Neuroendocrine carcinoma of the skin--an updated review.

96 : Merkel cell carcinoma: review.

97 : Primary neuroendocrine (Merkel cell?) carcinoma of the skin. I. A clinicopathologic and ultrastructural study of 43 cases.

98 : [Merkel cell tumor or neuroendocrine skin carcinoma].

99 : Merkel cell carcinoma can be distinguished from metastatic small cell carcinoma using antibodies to cytokeratin 20 and thyroid transcription factor 1.

100 : Small cell melanoma.

101 : Malignant melanoma with clinical and histologic features of Merkel cell carcinoma.

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

103 : Merkel cell carcinoma: histologic features and prognosis.

104 : Five hundred patients with Merkel cell carcinoma evaluated at a single institution.

105 : Transcriptome-wide studies of merkel cell carcinoma and validation of intratumoral CD8+ lymphocyte invasion as an independent predictor of survival.

106 : Impact of Tumor-Infiltrating Lymphocytes on Overall Survival in Merkel Cell Carcinoma.

107 : Clinical factors associated with Merkel cell polyomavirus infection in Merkel cell carcinoma.

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

109 : Expression of p63 is the sole independent marker of aggressiveness in localised (stage I-II) Merkel cell carcinomas.

110 : p63 expression in Merkel cell carcinoma predicts poorer survival yet may have limited clinical utility.

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

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

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

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

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

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

117 : 590 Detecting Merkel cell carcinoma recurrence using a blood test: Outcomes from 774 patients

118 : Merkel Cell Carcinoma Analysis of Outcomes: A 30-Year Experience.