Next Article in Journal
The Influence of the Ketogenic Diet on the Immune Tolerant Microenvironment in Glioblastoma
Next Article in Special Issue
Management and Prognosis of Cardiac Metastatic Merkel Cell Carcinoma: A Case–Control Study and Literature Review
Previous Article in Journal
COVID-19 Vaccination Campaign in Cancer Patients and Healthcare Workers-Results from a French Prospective Multicenter Cohort (PAPESCO-19)
Previous Article in Special Issue
Defining the Profile of Patients with Primary Merkel Cell Carcinoma in Central Italy: A Retrospective Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Merkel Cell Polyomavirus (MCPyV) and Cancers: Emergency Bell or False Alarm?

by
Maria Georgia Dimitraki
and
George Sourvinos
*
Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Crete, Greece
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(22), 5548; https://doi.org/10.3390/cancers14225548
Submission received: 25 September 2022 / Revised: 6 November 2022 / Accepted: 8 November 2022 / Published: 11 November 2022
(This article belongs to the Special Issue Merkel Cell Carcinoma: An Update and Review)

Abstract

:

Simple Summary

Merkel cell polyomavirus is a widespread pathogen, strongly associated with the highly aggressive Merkel cell carcinoma. In this review paper we aim to examine a possible association between Merkel cell polyomavirus and the emergence of different types of cancer. Evidently, Merkel cell polyomavirus is the major oncogenic factor for Merkel cell carcinomas, but no establishing results can be derived about the virus’ prevalence and role in the pathogenesis of other malignant diseases. Most certainly, it is imperative that the virus’ biology, infection cycle, as well as its interactions with the host, are further investigated. Identifying a link between a virus and tumorigenesis is crucial and could lead to new virus-targeted therapeutic approaches with one final target: cancer elimination.

Abstract

Merkel cell polyomavirus (MCPyV), the sole member of Polyomavirus associated with oncogenesis in humans, is the major causative factor of Merkel cell carcinoma (MCC), a rare, neuroendocrine neoplasia of the skin. Many aspects of MCPyV biology and oncogenic mechanisms remain poorly understood. However, it has been established that oncogenic transformation is the outcome of the integration of the viral genome into the host DNA. The high prevalence of MCPyV in the population, along with the detection of the virus in various human tissue samples and the strong association of MCPyV with the emergence of MCC, have prompted researchers to further investigate the role of MCPyV in malignancies other than MCC. MCPyV DNA has been detected in several different non-MCC tumour tissues but with significantly lower prevalence, viral load and protein expression. Moreover, the two hallmarks of MCPyV MCC have rarely been investigated and the studies have produced generally inconsistent results. Therefore, the outcomes of the studies are inadequate and unable to clearly demonstrate a direct correlation between cellular transformation and MCPyV. This review aims to present a comprehensive recapitulation of the available literature regarding the association of MCPyV with oncogenesis (MCC and non-MCC tumours).

1. Introduction

Cancer is a group of diseases with a major global health impact, estimated to become the leading cause of premature death in this century, according to GLOBOCAN 2020 [1]. The emergence of cancer is primarily linked to both genetic predisposition and environmental factors [2]. Nevertheless, recent studies have identified some infectious agents as risk factors of cancer development in humans [3]. According to data from 2018, infection-attributable cancers represented the third leading cause of cancer development (2.2 million cases) [4], following smoking and diet [5]. Additionally, infection-related cancer cases have mostly been reported in the developing parts of the world [4]. Most of those oncogenic agents are characterized as Group I human carcinogens by the International Agency for Research on Cancer (IARC) and they include viruses, bacteria, and parasites [6,7,8]. Recent advances in the fields of molecular biology have shed light on the correlation between fluke infections and cancer formation and, therefore, have provided valuable insights into the molecular basis of oncogenesis [9]. Generally, infectious agents seem to be able to cause malignant transformation via direct and indirect mechanisms: persistent infection that eventually leads to inflammation, DNA alternations and cell damage, irregular oncogene expression, and immunologic recognition disruption [7,8,9]. Amongst the infectious agents, studies targeting the precise oncogenic mechanisms of the viruses are the most abundant.
Human oncogenic viruses have been the in the centre of scientific attention for over 50 years. They represent a variety of viruses with a range in morphology, genetic material and, overall, viral life cycle and reproduction [4,10]. According to The World Health Organization, approximately 10% of infection-attributed cancers have viral aetiology [11]. The carcinogenic mechanisms of oncoviruses also vary widely [12] but they generally involve continued expression of specific viral oncogenes that regulate proliferative and antiapoptotic activities, dysregulation of cellular genomic instability through integration of the viral DNA into the host genome, viral promotion of DNA damage and immune evasion strategies [10,12]. However, it is significant to note that oncogenesis is an uncommon consequence of the viral infection, and it is developed mainly after a long-lasting chronic infection [10]. Moreover, the detection of a virus in a cancerous tissue does not necessarily establish causality [10]. Therefore, the direct or indirect involvement [13] of suspected viruses in the tumorigenesis process has been proposed to be made through the following list of characteristics: (1) presence and persistence of viral genome in biopsies obtained from tumour tissues; (2) presence of growth-promoting viral genes in model systems; (3) emergence of the malignant tissue exclusively due to continuous viral oncogene expression or dysregulation of host genes; and (4) epidemiological evidence of correlation between viral infection and cancer development [14]. So far, seven viruses have been confirmed to have an established causative link to oncogenesis. These are human papillomaviruses (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), Epstein–Barr virus (EBV), Kaposi’s sarcoma associated herpes virus (KSHV) (also called human herpes virus 8), human T-cell leukaemia virus (HTLV-1), and Merkel cell polyomavirus (MCPyV) [5,11]. This review focuses on Merkel cell polyomavirus and examines its association with various types of cancer.

2. Merkel Cell Polyomavirus and Merkel Cell Carcinoma

2.1. Merkel Cell Polyomavirus (MCPyV)

Merkel cell polyomavirus (MCPyV) is the only member of the Polyomaviridae family with scientifically proven ability to cause oncogenesis in humans [15]. MCPyV exhibits the characteristic genomic and structural organization of the family (Figure 1). It is a small, non-enveloped virus, approximately 45 nm in diameter, with icosahedral capsids [16]. The genome is represented by a short (approximately 5.4 kb) double-stranded circular DNA [17], packed together with host-derived histones [18]. It is comprised of three main regions, classified based on the sequential priority in which they are expressed during infection: the early-coding region, the late-coding region, as well as a noncoding, regulatory region (NCRR) [19]. NCRR is located in-between the other two and does not encode any functional protein or RNA. It possesses an essential role in the viral life cycle regulation, since it contains both the viral origin of replication (Ori) and, also, various transcription promoters [20,21]. Immediately upon viral infection, the expression of the early region is initiated, encoding multiple, alternatively spliced RNA transcripts, leading to distinctive gene products that are generally involved in the replication of the viral genome [17]. Those are the large tumor antigen (LT), the small tumor antigen (sT), as well as a 57 kT antigens, along with the alternate LT ORF (ALTO) product [20,21]. The LT, sT and 57 kT antigens all share an identical sequence of 78 amino acids at their N-terminal [22]. Lastly, the viral genome also contains a gene, located on the late strand of the T antigen genes, with an opposite transcription orientation to the T antigens. The transcriptional product of this gene is a non-coding miRNA [20,21]. The most important viral antigens proven to play a vital part in the lytic infection are the MCPyV LT and sT antigens [23]. More specifically, the LT antigen is a multifunctional, pleiotropic protein of 700 amino acids that impacts several cellular proteins involved in both the host cell and the viral life cycles [18,22]. It is mainly nuclear located, though phosphorylation modifications may lead to different localization within the cell [18]. Structurally, it is composed of a highly conserved N-terminal region (CR1) and a C-terminal region with an origin-binding domain (OBD) and a helicase domain, both critical for viral DNA replication. In-between the two terminal regions there can be found sequences responsible for the protein–protein interactions of LT with host proteins that end up alternating the normal cell cycle (RB,p53) [22]. Regarding the sT protein, it shares the LT N-terminal region, but has a unique C-region made of a protein phosphatase 2A (PP2A) binding site [22,24]. Following the expression of the early-coding locus and the replication of the viral DNA, the late region is transcriptionally activated to produce the structural components of the viral capsid, major capsid protein viral protein 1 (VP1) and the minor capsid protein 2 and 3 (VP2 and VP3) [20]. The presence of VP1 and VP2 is crucial for the viral entry into the cell, whereas VP3 cannot be detected in MCPyV-infected cells or in MCPyV virions, indicating a possible conditional expression pattern [18].
In general, MCPyV is a widespread and mostly asymptomatic resident of the skin in the human population [25,26,27]. Serological surveys for antibodies against viral antigens demonstrate that 20–40% of children 0–5 years old test positive [28], meaning that the first contact with this infectious agent occurs early in life [29], whereas positivity reaches 80% in individuals of age of 50 years and above [28,30,31]. Therefore, it is highly suggested that MCPyV is a member of the skin microbiome (virome) of the human body [32]. Moreover, the precise host cell tropism of MCPyV remains unclear. Interestingly, MCPyV seems to require both salicylic acid heparan sulphate upon its entrance [29,32,33], molecules ubiquitous in several cell types, though the replication of MCPyV has so far shown to be limited to keratinocytes [29] and fibroblasts from the dermis, and the lung [34]. Furthermore, among fibroblasts isolated from common model animals, only chimpanzee and human ones can support a vital level of MCPyV replication [35]. This lack of an eligible cell culture system for MCPyV infection [21], as well as the inability to establish an in vivo infection system [35], has limited our ability to understand the fundamentals of the viral behavior. Consequently, there is much to be elucidated with respect to MCPyV host potential reservoir cells and the complete MCPyV infectious cycle during infection. Despite the limitation regarding the target-cell preferences, MCPyV is not solely detectable within the skin, but also in respiratory samples, urine, and blood [20,36], as well as in samples extracted from numerous non-malignant tissues (e.g., spleen, bone marrow, stomach, heart), although with a comparatively low viral load [37]. Lastly, the route of MCPyV transmission has yet to be established, but suggested routes include primarily direct contact with the skin or saliva, and recently proposed ones, i.e., airborne and fecal–oral [18,29].
Nevertheless, MCPyV is not harmless in the minority of individuals and, on the contrary, it is considered the first, and, to date, the only polyomavirus directly implicated in the emergence of an aggressive, lethal human cancer, Merkel cell carcinoma (MCC).

2.2. Merkel Cell Carcinoma (MCC)

Merkel cell carcinoma (MCC) is a rare, aggressive, neuroendocrine neoplasia of the human skin [38], originally described in 1972 by Cyril Taker [39]. With a nearly 50% mortality rate [40] and a high case-fatality [21], it is considered one of the most lethal skin malignancies, surpassing melanoma [41]. The low survival rates are a direct consequence of the rapid metastatic ability (distant, regional metastases or lymph node metastases) of MCC [29,40,42,43], combined with its intrinsic ability to resist immunological eradication [29,43], as well as a relatively poorly response to chemotherapeutic agents and constant recurrence [44,45,46]. Furthermore, MCC is characterized by several established risk factors, including advanced age (50 years and older), population features (fair skin), exposure to intense solar radiation (skin displayed mainly to UV rays) and immune deficiencies [42,43,47]. In the last few years, even though MCC is still considered uncommon, both the incidence as well as the mortality rate of MCC have sharply tripled [21] and are anticipated to rise further [48]. Once emerged, MCC appears as a fast growing red or purple nodule of trabecular, nodular, or diffuse shape [47]. Despite the given name, attributed to the phenotypical similarities of the tumour with the Merkel cells of the skin, the true cell origin of MCC is rigorously debated [43,49]. The initial favoured theory, that MCC arises from unrestrained growth of differentiated Merkel cells, is debated due to the post-mitotic nature of the Merkel cells, which makes them terminally differentiated, unable to undergo further cell division, and consequently, making their oncogenic potential limited [20,50]. In addition, Merkel cell have epidermal origin, in contrast to the dermis or subcutis layer origin of the MCCs [20]. Therefore, plenty of other suggestions have been made, such as that MCCs could potentially occur from Merkel cell precursor cells present at the hair follicles of the skin, or other epithelial, fibroblastic, or even B-cells, primarily in accordance with morphological similarities shared by MCC and these potential pedigrees [20,51,52].
The two major causative factors of MCC are Merkel cell polyomavirus (MCPyV) and ultraviolet radiation [41,53]. Hence, Merkel cell carcinomas can be subdivided into two major groups: the virus-associated MCCs (VP-MCCs) and the virus-negative MCCs (VN-MCCs or UV-MCCs). Despite evidently different aetiologies, both groups share mutations in vital cell-cycle related genes (meaning they regulate similar molecular paths in different means) [29,54,55,56], as well as similar disease phenotype [57] and same emergence location (on sun-exposed regions of the body, such as the head, neck, and limbs [58,59]). On the contrary, VP-MCCs seem to have an insignificant mutational burden and a lower number of somatic mutations compared to VN-MCCs [60,61,62,63]. Initially, the MCCs caused by UV-light overexposure have been attributed to approximately 20% of the total cases and are characterized by high accumulation of UV-derived DNA mutations, specifically on genes that encode tumour-specific UV-neoantigens, such as the retinoblastoma (pRb) pathway, RB1, TP53, and PIK3CA, along with mutations in host DDR and chromatin modulation pathways [43,53,60,61]. Such aberrations are crucial for MCC emergence, since the dysregulation results in both interruption of the cell cycle and induction of SOX2 expression, leading to neuroendocrine transformation [64]. MCPyV-negative MCCs also have increased levels of activation-induced cytidine deaminase (AID) which could potentially enhance carcinogenesis [17].
Whilst UV radiation is considered a strong risk factor and leads to MCC-related mutagenesis in VN tumours, the major causative factor of MCC is MCPyV, evident in the vast majority of MCC cases [40]. More specifically, MCPyV was first detected and isolated in 2008 by Feng and colleagues at the Pittsburgh Cancer Institute within the cells of the primary MCC tumour and a metastatic lymph node, using digital transcriptome subtraction assays. The viral genome, which was detected in 80% of the MCC samples, also exhibited a clonal integration pattern within several different chromosomal sites [15,29,40]. This identification of MCPyV was a significant leap in the comprehension of the pathogenesis of MCC in virus-associated MCC, but the exact role of MCPyV in MCC pathogenesis requires further investigation [21]. In the last few years, significant progress in decoding the mechanism of MCC-VP tumorigenesis has been observed. Thus, oncogenic transformation by MCPyV is hypothesized to be the outcome of two consecutive events [29]: (1) the incorporation of the viral DNA into the host genome, (2) the subsequent expression of the viral oncogenic proteins. Initially, the clonal incorporation of MCPyV DNA into the host genome transpires at generally random genomic sites, most commonly on chromosome 5. This event has been evident in up to 80% of all studied MCCs [65,66]. The integration takes place as either a single copy or as a concatemer of multiple copies, always in a manner that results in loss of replicative abilities of the virus before MCC development, but, simultaneously, in a way that preserves the expression of the so-called viral tumour (T) antigens [20,67]. The MCPyV-encoded antigens, sT and LT, are highly immunogenic and are required for the MCC development as well as the oncogenic phenotype maintenance [23,50]. They contribute to oncogenesis by targeting various host cell proteins involved in cell cycle control and proliferation [20]. The significance of the T antigens in MCC tumorigenesis is highlighted by the fact that their expression is required for optimal MCC cell growth and proliferation [9,68]. Depletion of these tumour antigens from VP-MCC cell lines, on the other hand, leads to cancer cell death [20]. Starting with the LT antigen, sequencing assays have indicated several truncating mutations within the C-terminus of LT, most probably caused by and during the viral genome integration process. These mutated LT antigens seem to lack the DNA binding domain–helicase activity, thus interrupting the viral DNA replication in MCC tumours [17]. However, the truncated LT proteins retain several other structural domains and motifs within the N-terminus region, resulting in interaction with the tumour-repressor retinoblastoma protein (pRb) [68], thus inactivating it and promoting cell cycle deregulation and cellular proliferation (Figure 2). MCPyV LT is also associated with Vam6/Vps39-like protein, which is potentially significant for the viral cell cycle [54]. The oncogenic significance of the LT antigen is further indicated by the observation that the silence of this antigen in VP-positive MCC-derived cells intervenes with the cell growth and promotes cell death [17,69]. All in all, the accumulation of mutations in the LT antigen plays a vital role in the carcinogenesis process, since these mutations not only downregulate the viral replication and viral load, promoting immune evasion, but also promote unrestrained cell proliferation. Regarding the sT antigen, it is more frequently detected in human MCC tumours than the LT antigen and is considered a more crucial ‘player’ for oncogenesis [18]. Experiments on transgenic mouse models have indicated a transformative ability of sTs in various organ systems, including in the epidermis [54]. Furthermore, the sT antigen is able to transform rat-1 fibroblasts in cell cultures [54]. ST has been found to interact with a variety of host cellular proteins and, therefore, exhibits a range of activities. Initially, sT’s PP2A region interacts and inhibits PP2A and PP4 phosphate complexes. This interaction promotes inhibition of the nuclear factor-κB (NF-κB), as well as alternations in the cellular cytoskeleton, resulting in cell motility. Nevertheless, the PP2A binding domain is not considered crucial for carcinogenesis, since studies have shown that its presence is not required for cellular transformation by MCPyV ST31 [54]. An sT antigen region that is vital for the virus-induced oncogenesis is the LT-stabilizing domain (LSD), which has been suggested to interact with F-box/WD repeat-containing protein 7 (FBXW7) and cell division cycle protein 20 homologue (CDC20), resulting in inhibition of E3 ubiquitin ligases, and subsequent enhancement of the oncoprotein stability and cap-dependent mRNA translation, respectively. Additionally, research on sT has demonstrated its ability to promote changes in gene expression, by regulating both EP400 histone acetyltransferase and chromatin remodelling complex66 [54]. Lastly, studies suggest that sT might also interfere with the cell metabolism [54]. Therefore, sT antigen appears to possess a substantial role in MCPyV-related cancer emergence. Finally, immunocompromised patients display a higher (16-fold) relative risk for MCC development, opposed to the healthy population [18]. Therefore, immuno-suppression is classified as another crucial contributing factor in MCPyV-mediated carcinogenesis [43,70,71]. It is also imperative to mention that in MCPyV-positive MCCs, UV light may simply promote tumour growth through immunosuppressive effects on the tumour microenvironment [9].
According to molecular epidemiological studies, the prevalence of MCPyV in MCC varies widely depending on the region of interest [5,72], ranging between approximately 25% in Australia and 100% in a French study [72]. Subsequent studies demonstrated values of 76.0% in the United States population and 66.6% in Switzerland [5]. Furthermore, MCPyV DNA has been shown to be present in 55–79% of MCCs in Japanese patients [72]. The detection methods used for MCC diagnosis initiate with clinical examination and are followed by tissue biopsy through immunochemistry, a method that demonstrates characteristic histopathologic neuroendocrine features [43]. Immunohistochemistry aims to differentiate MCCs from other, morphologically similar, neuroendocrine tumours through characteristic staining of a variety of MCC epithelial and neuroendocrine markers, such as cytokeratin-20 (CK20), neurofilaments, CAM 5.2, TTF-1 and AE1/3 [73,74]. With the purpose of distinguishing the VP-MCCs from VN-MCCs, a series of identification techniques are performed, including serological anti-oncoprotein antibody detection and molecular techniques of viral DNA amplification and detection (PCR, qPCR) [75]. For the molecular assays, the most common primers used target the LT and sT gene and the VP1 and NCCR regions of the genome [75]. Lastly, for confirmation purposes, a combination of molecular and immunohistochemical analyses can be performed (since viral proteins can be detected in tumour tissues) [75]. On average, the viral genome copy number of MCPyV has been estimated to be 60 times lower in healthy tissues across the body compared to MCC samples [76]. The available treatment options for both UV- and virus-induced MCCs, are, so far, very limited. Surgical extraction followed by rounds of chemotherapy is the recommended route [43]. Due to the immunogenic properties of MCCs, a novel therapy based on immune checkpoint inhibitors has recently shown encouraging survival outcomes [62,77,78], though this approach is considered insufficient for the systemically immunosuppressed patients [79,80]. Lastly, therapeutic vaccines are currently being developed [9,81].

2.3. MCPyV and Non–Merkel Cell Carcinomas

As aforementioned, MCPyV is chronically shed from the skin [32] of the vast majority of the healthy population globally and it has been detected, not solely on the skin, but also in a variety of other tissues, thus raising the possibility that the virus establishes an infectious reservoir in body tissues other than the skin, such as the lymph nodes or within the blood. Accordingly, the widespread prevalence of the virus across the human body in combination with the strong association of MCPyV in the development of MCC have led to the following question in the scientific community: what if MCPyV is present and has an oncogenic linkage to other, non-MCC cancers? This question has prompted researchers to further investigate a possible existence of MCPyV in cancers emerging throughout the body, searching for a possible linkage between malignancies and the MCC-inducing virus. Therefore, cancerous tumours, derived from different body locations, have been systematically tested for the presence of viral DNA, antigen transcripts, and proteins. In most cases, the MCPyV DNA was extracted from the tumour biopsies using PCR-based methods, whereas the expression of the LT antigen was monitored by immunohistochemistry. Interestingly, LT could only be detected in a handful of cases, even though the viral DNA was present [82]. Lastly, it is important to mention that the research projects only investigated the presence of the viral genome and the LT antigens and scarcely the integration of the genome or the truncated LT antigen [82], even though these characteristics are the hallmarks for the MCPyV-positive MCCs.
The outcomes of these investigations are listed below, divided into groups according to the system of the body where each malignancy was isolated (Table 1).

2.3.1. Circulatory System

The presence of MCPyV in tumours has been tested in different types of leukaemia, such as acute myeloid leukaemia (AML), cutaneous T-cell leukaemia (CTCL), cutaneous B-cell lymphomas (CBCL), and chronic lymphocytic leukaemia/small lymphocytic lymphoma (CLL/SLL). Initially, acute myeloid leukaemia (AML) is a type of blood cancer of the myeloid line of blood cells (granulocytes or monocytes) in the bone marrow, which results in interference with normal blood cell production. AML progresses rapidly and is typically fatal in the span of a few weeks if left untreated. Samples of AML cells have been tested using qPCR as well as NGS. From the 29 samples, only one was found positive for MCPyV DNA [83,84]. Furthermore, the results from studies on cutaneous T-cell lymphoma (CTCL), a special non-Hodgkin’s lymphoma caused by a mutation of T cells which migrate to and reside in the skin, do not demonstrate a clear outcome regarding MCPyV. In particular, from the 352 studied samples of CTCL, some contained no detectable levels of MCPyV cells [85,86], while others reported some level of MCPyV DNA [87,88] (viral load 0.0012–12.467 viral copies per cell) [82]. The methods used were PCR/Southern blot and qPCR. Regarding cutaneous B-cell lymphomas (CBCL), slowly growing B-cell lymphomas of the skin, studies detected the virus in CBCLs using PCR/qPCR, with a relatively low prevalence (5.6%) [82], a low viral load per cell, and without viral protein expression (IHC for LT antigen was negative for all samples) [88,89,90]. Another type of B-cell leukaemia/lymphoma is chronic lymphocytic leukaemia/small cell lymphoma (CLL/SLL). It is the most common B-cell-related leukaemia affecting >15,000 patients/year [91,92], and it has an established epidemiological link with Merkel cell carcinoma (a large population-based study revealed that CLL patients have a higher risk of developing VP-MCCs) [92,93]. Therefore, there has been some interest in investigating a possible leukemogenic role of Merkel cell polyomavirus (MCPyV). PCR studies have demonstrated the presence of MCPyV in peripheral blood at low copies [94,95,96,97]. The viral load was only determined in one case, and it was extremely low, with 0.000017–0.002 viral copies per cell [82,96]. Moreover, amongst the positive samples, six contained a truncated LT antigen of which two also harboured full-length LT mRNA [94]. Lastly, no MCPyV genome was detected in chronic myelomonocytic leukaemia cells [84], mantle cell lymphoma cells [98], follicular lymphomas [99], primary effusion lymphomas [100] and small-cell carcinomas of the lymph nodes [101].

2.3.2. Digestive System

MCPyV sequences have been detected in various tumour samples from the digestive tract. Amongst the different cancers, oesophagus cancer (156 specimens), liver cancer (27 specimens) and salivary gland cancer (185 specimens) seem to have the highest prevalence of the virus, reaching up to 62%, 45.1% and 26.2%, respectively [76,82,102,103]. The detection was performed with the use of qPCR assays. In contrast, qPCR analyses of gallbladder [37], pancreas [37], intestine [106,107], appendix [106] and gastrointestinal cancers [108] indicated no trace of MCPyV genome. Another examined digestive-tract-associated cancer is colon cancer. Colon cancer is the third on the list of cancer-associated fatalities in Western countries, estimated to have caused 9% among all cancer deaths in 2020. Some evidence points to pathogens, such as viruses, as risk factors for its oncogenesis. [159,160,161]. Nevertheless, studies showed a very low prevalence (3.5%) with low and even no positivity toward viral transcripts [82,109,110,111,112,113]. The same pattern was observed in stomach cancer (1.7% prevalence) [82,109,113]. Regarding the upper regions of the digestive system, the oral cavity, some viral genome copies have been detected there. Lastly, some MCPyV-positive tumours were identified in oral cavity cancers; LT protein was not detected in any of them, and the copy number of the virus/cell was low (0.00024–0.026) [82,102,104,105,114,115].

2.3.3. Excretory System

The excretory system is the regulator of the chemical composition of body fluids. It is responsible for the discarding of metabolic wastes of the body, as well as the preserving of the adequate water, salts and nutrients levels within the cells and tissues. Studies have investigated the presence of MCPyV in cancers extracted from the key components of this system, the kidneys [76,109] and the bladder [76,109,113]. In both cases, the prevalence of the virus was low (4% derived from 149 samples of bladder cancer and 3.7% from 81 samples of renal cancer), with a viral load of 0.001 and 0.004 copies/cell [76,82]. Furthermore, IHC assays could not detect any sign of the LT antigen in any of the samples [109,113].

2.3.4. Integumentary System

The integumentary system is the body’s outer layer, meaning it consists of the skin with its glands and nerves, nails, and hair. Since the Merkel cell carcinomas originate from and emerge on the skin, other skin cells in proximity to the tumorigenesis location could potentially be susceptible to the presence of the virus and, consequently, become malignant. Therefore, MCPyV has been searched for and eventually detected in the integumentary system. Initially, MCPyV DNA was detected at a varying level in non-melanoma cancers, acquired from immunocompromised patients (prevalence 31.3%) [82,112]. In this rate are included many non-melanoma skin cancers, such as squamous cell carcinomas (SCC) [112,114,116,117,118,119] and basal cell carcinomas (BCC) [112,120,121,122]. The presence of the virus was also detected in a few cases of keratoacanthoma (viral load 0.0001–0.10 viral genome copies/cell, no LT detected) [82,117,120,123], Kaposi’s sarcoma (0.00001–0.00685 viral genome copies/cell) [82,100,124], porocarcinoma (0.00022–0.212 viral genome copies/cell) [82,114,125] and atypical fibroxanthoma (0.0001–0.031 viral genome copies/cell) [82,120]. On the contrary, melanomas do not seem to typically harbour MCPyV [106,126,127]. There is only one documented case in which acral lentiginous melanomas and in nodular melanomas tested positive [121].

2.3.5. Lymphatic System

In the available data derived from studies on some lymphatic system cancers (hypertrophy adenoid [132]), the prevalence of MCPyV was found to be low. Nevertheless, tonsillar squamous cell carcinoma (Tonsillar SCC) and thymoma qPCR analysis demonstrated a 32% and 15.2% prevalence of the viral DNA, respectively [82,128,129,130,131]. Additionally, three MCPyV DNA-positive thymoma samples also expressed the LT antigen [131]. It is crucial to mention, though, that Toracchio et al. [137] identified transcripts of the other oncogenic antigen of MCPyV, sT antigen, within benign, non-malignant lymph nodes.

2.3.6. Nervous System

Studies focusing on tumours of the nervous system are scarce. Only a limited number of cases in which the MCPyV genome was detected are available and they include brain tumours [101,109,133,134,135,136], schwannomas [102,115,137], a rare type of nerve sheath tumour, mostly benign, meningiomas [102] and glioblastomas [102,138]. Furthermore, Matthay et al. analysed the viral presence in neuroblastomas, the most common malignant extracranial solid tumour in childhood [139]. The results indicated no correlation between the viral DNA in the neuroblastoma samples [113,139,140,141].

2.3.7. Respiratory System

As mentioned already, MCPyV has been detected on respiratory samples and lung fibroblasts are an eligible cell system for the in vitro viral reproduction [34]. Therefore, a lot of hypotheses have arisen regarding the possibility of a linkage between MCPyV and carcinogenesis events in the respiratory system. Additionally, healthy lung tissues have been examined regarding the presence of the virus, and genome transcripts have been detected [37,76,100,142]. Therefore, much research has focused on the detection of genome transcripts in respiratory malignancies. Initially, MCPyV was detected via qPCR and nPCR in lung cancer only in 10 positive cases out of the 388 tested samples, and IHC did not show any detectable level of LT antigen expression in any of the positive samples [37,76,142]. Similarly, the majority of the studies on small-cell lung carcinomas point out a general lack of viral presence, with only a few tumours being MCPyV DNA-positive (5.2% prevalence with 0.000005–0.026 viral copies per cell) [82,107,113,143,144,145,146]. In contrast, scientific evidence has led to a strong association between non-small-cell lung carcinoma pathogenesis and the presence of MCPyV (16.3% prevalence) [82,130,147,148,149]. Even though most of the small-cell lung carcinoma samples showed no LT expression [150], two of the ones that did are of exceptional significance. In particular, those two samples contained a truncated LT antigen and one of those seemed to express both the full-length and truncated LT protein [130]. Further investigations on this specific, peculiar sample, led to the finding of two viral DNAs in the same malignancy sample, both in episomal and integrated form. A similar observation was made in a few Merkel cell carcinomas, where episomal and integrated viral genomes co-existed in the cytoplasm and nucleus simultaneously [130]. Additionally, studies have observed an association between the presence of Merkel cell polyomavirus with a deregulated expression of two genes (BRAF and Bcl-2) in non-small cell lung cancer [151], as well as virus-specific microRNA signature [152].

2.3.8. Reproductive System

The data regarding the involvement of MCPyV in tumorigenesis in the human reproductive system are inadequate. However, there are studies focused on the detection of the virus in malignant samples acquired from reproductive organs that demonstrate some trace of viral presence both in females and males. The samples tested originated from prostate [76,153] and testicular cancer in males [76] (the two most common male-reproductive system cancers) and cervical [155,156], breast [157], ovarian and vulvar cancer [156,157] in women. Initially, MCPyV was detected via qPCR (LT, VP1 sequences) in one case of testicular cancer, a very common, highly treatable, and usually curable cancer in men [76]. Interestingly, the viral load was relatively high, at 0.934 copies/cell [76,82]. The LT protein expression, though, was not assessed [76]. Furthermore, MCPyV-positive prostate cancers, the second-most common type of cancer and the fifth-leading cause of cancer-related death in men [154], have been analysed with a variety of methods for both the viral genome and the LT antigen (qRT-PCR, NGS, IHC). The viral load was found to be lower than in the testicular cancer case, at 0.002 copies/cell, whereas no LT antigen was detected in the tumour [76]. Regarding the female reproduction system, breast cancer is the most prevalent cancer in the female population worldwide [158], with an estimated 1.38 million new cases and over 450,000 deaths annually. The aetiology of the minority of breast cancers is genetic-related, though the rest of the sporadic breast cancers remain a mystery. Studies have proposed a possible aetiological relationship between viral infection breast cancer oncogenesis, which remains to be established. Hence, approximately 474 samples of breast cancer tissue have been used for MCPyV genome detection, via PCR along with whole transcriptome sequencing [82]. Out of all samples, viral genomic transcripts were identified in only a few cases of breast cancer cells [113,157]. Furthermore, other research studies have attempted to determine the prevalence of MCPyV in cervical cancer (the third most prevalent malignancy and the fourth most fatal cancer in women [3,162]) as well as to assess a possible connection between MCPyV and HPV-related cervical cancer. The results demonstrated that the cervical cancer samples contained a relatively low viral copy number per tumour cell, between 0.00003055 and 0.0015 [82,155,156]. Despite the low viral genome quantity, LT antigens (transcripts and proteins) were detected superficially in samples isolated from HIV-positive women [156]. Finally, MCPyV viral transcripts were not detected in either ovarian or vulvar cancer samples [113,140].

2.3.9. Soft Tissue

Lastly, soft tissue is all the tissue in the body that is not ossified or calcified, and, therefore, remains unhardened. The role of soft tissue is the connection, surrounding and support of other tissues, internal organs, and bones. This tissue category comprises muscles (including the heart), fat, nerves, as well as lymph and blood vessels, ligaments, tendons, and tissues that surround the bones and joints [163,164]. Soft tissue cancers can arise at any age, with an increased prevalence amongst children and young adults, along with adults over the age of 55. Thus far, the soft tissue-related tumours examined (all by Sastre-Garau et al. [140]) are a limited number of rhabdomyosarcoma tumours (25 samples, a rare type of cancer that forms in skeletal muscle tissue), desmoplastic tumours (24 samples, these grow in the abdomen and pelvic area of the body) and fibromatosis tumours (4 samples, a connective tissue malignancy developing in musculoaponeurotic tissues). The detection of the viral sequences was performed by PCR (primers for the conserved LT sequences). The results demonstrated that none of the soft tissue malignant specimens harboured any MCPyV DNA.

3. Conclusions

In summary, a causality connection between infectious agents and carcinogenesis is well established. Merkel cell polyomavirus (MCPyV), a small DNA virous, is, thus far, the only polyomavirus associated with oncogenesis in humans. Normally, it is a widespread member of the skin microbiome. However, in some rare cases, especially in immunocompromised individuals, it evidently triggers the pathogenesis of Merkel cell carcinomas (MCC) on the skin. MCC is a rare, extremely aggressive neuroendocrine neoplasia of the skin, of uncertain cell origin, that emerges mainly on UV-exposed areas. It is characterized by low survival rates, rapid metastatic ability, and ever-increasing incidence. Additionally, the available therapeutic approaches are considered inadequate. The data regarding the biology of the virus are insufficient and the precise oncogenic mechanism remains to be elucidated. However, the MCC onset is confirmed to be dependent on two events: viral DNA integration into the host genome and viral oncoprotein expression, such as a truncated version of the LT antigen. Both events, along with other UV-related mutations, lead to interference with the normal cellular life circle and cell proliferation and, consequently, lead to cell transformation.
MCPyV is not detected solely in skin samples, but also in other tissues of the human body. This finding, along with the high seroprevalence of MCPyV in the human population, the in vitro oncogenic potentials of LT and sT, as well as its strong tumorigenic properties on MCC, have triggered attempts to detect MCPyV (DNA and proteins) in non-MCC tumour samples. In total, many of the samples tested displayed the presence of the virus in various non-MCC cancers. Nevertheless, the results do not clearly demonstrate a direct connection between cellular transformation and the presence of the virus, due to some pitfalls of the studies [82]. Firstly, for the virus-positive cases, the numbers of the same tumour types tested are insufficient for a conclusion to be made, and many of the studies were case reports. For statistical significance, large cohort studies are needed. Furthermore, the tested samples were extracted from patients of different health backgrounds, for instance both immunocompetent and immunocompromised patients, and the abstracted results were considered, in many cases, as one category, even though the condition of the immune system of an individual plays a crucial role in the onset and progress of malignant tumours. Additionally, even with the presence of the viral DNA, the prevalence, viral load, and protein expression were significantly lower compared to MCCs and the viral load (genome copies/cell) was scarcely determined. Moreover, the two hallmarks of MCPyV MCC, namely the state of the viral genome and whether a truncated large T antigen is expressed, have not systematically been investigated in non-MCC tumours. Expression of truncated LT and/or integration of the viral genome was only reported in a few cases. Lastly, in several reported cases, the prevalence in healthy tissue was comparable to malignant tissue.
Regarding the virus-negative tumours, the lack of the viral genome within a tumour cannot eliminate the viral–oncogenesis possibility. Initially, the absence of the virus from the tested samples could be attributed to dysfunction of molecular techniques used for the detection. In particular, PCR-based methods were the choice of the vast majority of the research. The results can be affected by the primers that were used, the quality of the samples, as well as possible mishandling and contamination (considering MCPyV is chronically shed from the skin [32]). Other more sensitive techniques such as NGS could provide a highly sensitive approach to detect the viral genome in non-MCC tumours. Furthermore, the total absence of the viral genome does not necessarily exclude a possible scenario where the viral presence is not needed for the tumour progression, as it has been described before. In this scenario, MCPyV would be necessary for tumour initiation, but at later stages the virus is dispensable. In some MCC cases, this “hit-and-run mechanism” for MCPyV, has been suggested based on the observation that a knockdown LT antigen in the MCPyV-positive MKL-1 cell induced growth repression, whereas no impaired growth was reported in LoKe cells that were LT-deprived [165]. Lastly, the viral proteins, without the presence of its genetic material, could have enhancive effects on other co-infecting oncovirus, since the co-presence of MCPyV and human papillomavirus and Epstein–Barr virus in tumours has been reported [129,153,166,167].
In conclusion, MCPyV is the major oncogenic factor for MCCs, but no establishing results can be derived about the virus’ prevalence in non-MCC skin abnormalities and its potential role in the pathogenesis of other malignant diseases. Most certainly, it is imperative that the biochemical, molecular, and immunological characteristics of the virus are further investigated. The more understanding we have on the virus’ biology and infection, the easier it will be to investigate its correlation with the tumorigenesis of other malignant cells, and, also, many more possible preventative and therapeutic targets will be found, taking us one step closer to the elimination of Merkel cell carcinomas.

Author Contributions

Conceptualization, G.S.; methodology, M.G.D.; software, M.G.D.; validation, M.G.D. and G.S.; formal analysis, M.G.D.; investigation, M.G.D.; resources, M.G.D.; writing—original draft preparation, M.G.D.; writing—review and editing, G.S.; visualization, M.G.D.; supervision, G.S.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Khan, N.; Afaq, F.; Mukhtar, H. Lifestyle as risk factor for cancer: Evidence from human studies. Cancer Lett. 2010, 293, 133–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hatta, M.N.A.; Mohamad Hanif, E.A.; Chin, S.F.; Neoh, H.M. Pathogens and Carcinogenesis: A Review. Biology 2021, 10, 533. [Google Scholar] [CrossRef] [PubMed]
  4. de Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. Lancet Glob. Health 2020, 8, e180–e190. [Google Scholar] [CrossRef] [Green Version]
  5. Chua, T.H.; Punjabi, L.S.; Khor, L.Y. Tissue Pathogens and Cancers: A Review of Commonly Seen Manifestations in Histo- and Cytopathology. Pathogens 2021, 10, 1410. [Google Scholar] [CrossRef] [PubMed]
  6. De Flora, S.; La Maestra, S. Epidemiology of cancers of infectious origin and prevention strategies. J. Prev. Med. Hyg. 2015, 56, E15–E20. [Google Scholar]
  7. Masrour-Roudsari, J.; Ebrahimpour, S. Causal role of infectious agents in cancer: An overview. Casp. J. Intern. Med. 2017, 8, 153–158. [Google Scholar]
  8. Zella, D.; Gallo, R.C. Viruses and Bacteria Associated with Cancer: An Overview. Viruses 2021, 13, 1039. [Google Scholar] [CrossRef] [PubMed]
  9. Cai, Q.; Yuan, Z.; Lan, K. (Eds.) Infectious agents associated cancers: Epidemiology and molecular biology. In Advances in Experimental Medicine and Biology; Springer: Singapore, 2017; Volume 1018. [Google Scholar]
  10. Schiller, J.T.; Lowy, D.R. An Introduction to Virus Infections and Human Cancer. Recent Results Cancer Res. 2021, 217, 1–11. [Google Scholar] [PubMed]
  11. Plummer, M.; de Martel, C.; Vignat, J.; Ferlay, J.; Bray, F.; Franceschi, S. Global burden of cancers attributable to infections in 2012: A synthetic analysis. Lancet Glob. Health. 2016, 4, e609–e616. [Google Scholar] [CrossRef] [Green Version]
  12. Schiller, J.T.; Lowy, D.R. Virus infection and human cancer: An overview. Recent Results Cancer Res. 2014, 193, 1–10. [Google Scholar] [PubMed]
  13. Morales-Sánchez, A.; Fuentes-Pananá, E.M. Human Viruses and Cancer. Viruses 2014, 6, 4047–4079. [Google Scholar] [CrossRef] [PubMed]
  14. Krump, N.A.; You, J. Molecular mechanisms of viral oncogenesis in humans. Nat. Rev. Microbiol. 2018, 16, 684–698. [Google Scholar] [CrossRef] [PubMed]
  15. Feng, H.; Shuda, M.; Chang, Y.; Moore, P.S. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 2008, 319, 1096–1100. [Google Scholar] [CrossRef] [Green Version]
  16. Becker, M.; Dominguez, M.; Greune, L.; Soria-Martinez, L.; Pfleiderer, M.M.; Schowalter, R.; Buck, C.B.; Blaum, B.S.; Schmidt, M.A.; Schelhaas, M. Infectious Entry of Merkel Cell Polyomavirus. J. Virol. 2019, 93, e02004-18. [Google Scholar] [CrossRef] [Green Version]
  17. Krump, N.A.; You, J. From Merkel Cell Polyomavirus Infection to Merkel Cell Carcinoma Oncogenesis. Front. Microbiol. 2021, 12, 739695. [Google Scholar] [CrossRef]
  18. Prado, J.C.M.; Monezi, T.A.; Amorim, A.T.; Lino, V.; Paladino, A.; Boccardo, E. Human polyomaviruses and cancer: An overview. Clinics 2018, 73, e558s. [Google Scholar] [CrossRef]
  19. Spurgeon, M.E.; Lambert, P.F. Merkel cell polyomavirus: A newly discovered human virus with oncogenic potential. Virology 2013, 435, 118–130. [Google Scholar] [CrossRef] [Green Version]
  20. Yang, J.F.; You, J. Merkel cell polyomavirus and associated Merkel cell carcinoma. Tumour Virus Res. 2022, 13, 200232. [Google Scholar] [CrossRef]
  21. Liu, W.; Krump, N.A.; Buck, C.B.; You, J. Merkel Cell Polyomavirus Infection and Detection. J. Vis. Exp. 2019, 144, e58950. [Google Scholar] [CrossRef]
  22. Wendzicki, J.A.; Moore, P.S.; Chang, Y. Large T and small T antigens of Merkel cell polyomavirus. Curr. Opin. Virol. 2015, 11, 38–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Baez, C.F.; Brandão Varella, R.; Villani, S.; Delbue, S. Human Polyomaviruses: The Battle of Large and Small Tumor Antigens. Virology 2017, 8, 1178122X17744785. [Google Scholar] [CrossRef] [PubMed]
  24. Kwun, H.J.; Shuda, M.; Camacho, C.J.; Gamper, A.M.; Thant, M.; Chang, Y.; Moore, P.S. Restricted protein phosphatase 2A targeting by Merkel cell polyomavirus small T antigen. J. Virol. 2015, 89, 4191–4200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kamminga, S.; van der Meijden, E.; Feltkamp, M.C.W.; Zaaijer, H.L. Seroprevalence of fourteen human polyomaviruses determined in blood donors. PLoS ONE 2018, 13, e0206273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Pastrana, D.V.; Tolstov, Y.L.; Becker, J.C.; Moore, P.S.; Chang, Y.; Buck, C.B. Quantitation of human seroresponsiveness to Merkel cell polyomavirus. PLoS Pathog. 2009, 5, e1000578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Kean, J.M.; Rao, S.; Wang, M.; Garcea, R.L. Seroepidemiology of human polyomaviruses. PLoS Pathog. 2009, 5, e1000363. [Google Scholar] [CrossRef] [Green Version]
  28. Chen, T.; Hedman, L.; Mattila, P.S.; Jartti, T.; Ruuskanen, O.; Söderlund-Venermo, M.; Hedman, K. Serological evidence of Merkel cell polyomavirus primary infections in childhood. J. Clin. Virol. 2011, 50, 125–129. [Google Scholar] [CrossRef]
  29. Stachyra, K.; Dudzisz-Śledź, M.; Bylina, E.; Szumera-Ciećkiewicz, A.; Spałek, M.J.; Bartnik, E.; Rutkowski, P.; Czarnecka, A.M. Merkel Cell Carcinoma from Molecular Pathology to Novel Therapies. Int. J. Mol. Sci. 2021, 22, 6305. [Google Scholar] [CrossRef]
  30. Tolstov, Y.L.; Pastrana, D.V.; Feng, H.; Becker, J.C.; Jenkins, F.J.; Moschos, S.; Chang, Y.; Buck, C.B.; Moore, P.S. Human Merkel cell polyomavirus infection II. MCV is a common human infection that can be detected by conformational capsid epitope immunoassays. Int. J. Cancer. 2009, 125, 1250–1256. [Google Scholar] [CrossRef] [Green Version]
  31. Viscidi, R.P.; Rollison, D.E.; Sondak, V.K.; Silver, B.; Messina, J.L.; Giuliano, A.R.; Fulp, W.; Ajidahun, A.; Rivanera, D. Age-specific seroprevalence of Merkel cell polyomavirus, BK virus, and JC virus. Clin. Vaccine Immunol. 2011, 18, 1737–1743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Schowalter, R.M.; Pastrana, D.V.; Pumphrey, K.A.; Moyer, A.L.; Buck, C.B. Merkel cell polyomavirus and two previously unknown polyomaviruses are chronically shed from human skin. Cell Host Microbe. 2010, 7, 509–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Schowalter, R.M.; Pastrana, D.V.; Buck, C.B. Glycosaminoglycans and sialylated glycans sequentially facilitate Merkel cell polyomavirus infectious entry. PLoS Pathog. 2011, 7, e1002161. [Google Scholar] [CrossRef]
  34. Liu, W.; Yang, R.; Payne, A.S.; Schowalter, R.M.; Spurgeon, M.E.; Lambert, P.F.; Xu, X.; Buck, C.B.; You, J. Identifying the Target Cells and Mechanisms of Merkel Cell Polyomavirus Infection. Cell Host Microbe. 2016, 19, 775–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Liu, W.; Krump, N.A.; MacDonald, M.; You, J. Merkel Cell Polyomavirus Infection of Animal Dermal Fibroblasts. J Virol. 2018, 92, e01610-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Mertz, K.D.; Junt, T.; Schmid, M.; Pfaltz, M.; Kempf, W. Inflammatory monocytes are a reservoir for Merkel cell polyomavirus. J. Investig. Dermatol. 2010, 130, 1146–1151. [Google Scholar] [CrossRef] [Green Version]
  37. Matsushita, M.; Kuwamoto, S.; Iwasaki, T.; Higaki-Mori, H.; Yashima, S.; Kato, M.; Murakami, I.; Horie, Y.; Kitamura, Y.; Hayashi, K. Detection of Merkel cell polyomavirus in the human tissues from 41 Japanese autopsy cases using polymerase chain reaction. Intervirology 2013, 56, 1–5. [Google Scholar] [CrossRef]
  38. Elder, D.E.; Bastian, B.C.; Cree, I.A.; Massi, D.; Scolyer, R.A. The 2018 World Health Organization Classification of Cutaneous, Mucosal, and Uveal Melanoma: Detailed Analysis of 9 Distinct Subtypes Defined by Their Evolutionary Pathway. Arch. Pathol. Lab Med. 2020, 144, 500–522. [Google Scholar] [CrossRef] [Green Version]
  39. Toker, C. Trabecular carcinoma of the skin. Arch. Dermatol. 1972, 105, 107–110. [Google Scholar] [CrossRef]
  40. Becker, J.C.; Stang, A.; DeCaprio, J.A.; Cerroni, L.; Lebbé, C.; Veness, M.; Nghiem, P. Merkel cell carcinoma. Nat. Rev. Dis. Primers 2017, 26, 17077. [Google Scholar] [CrossRef]
  41. Pietropaolo, V.; Prezioso, C.; Moens, U. Merkel Cell Polyomavirus and Merkel Cell Carcinoma. Cancers 2020, 12, 1774. [Google Scholar] [CrossRef]
  42. Walsh, N.M.; Cerroni, L. Merkel cell carcinoma: A review. J. Cutan. Pathol. 2021, 48, 411–421. [Google Scholar] [CrossRef] [PubMed]
  43. Dellambra, E.; Carbone, M.L.; Ricci, F.; Ricci, F.; Di Pietro, F.R.; Moretta, G.; Verkoskaia, S.; Feudi, E.; Failla, C.M.; Abeni, D.; et al. Merkel Cell Carcinoma. Biomedicines 2021, 9, 718. [Google Scholar] [CrossRef] [PubMed]
  44. Allen, P.J.; Bowne, W.B.; Jaques, D.P.; Brennan, M.F.; Busam, K.; Coit, D.G. Merkel cell carcinoma: Prognosis and treatment of patients from a single institution. J. Clin. Oncol. 2005, 23, 2300–2309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Nghiem, P.; Kaufman, H.L.; Bharmal, M.; Mahnke, L.; Phatak, H.; Becker, J.C. Systematic literature review of efficacy, safety and tolerability outcomes of chemotherapy regimens in patients with metastatic Merkel cell carcinoma. Future Oncol. 2017, 13, 1263–1279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Iyer, J.G.; Blom, A.; Doumani, R.; Lewis, C.; Tarabadkar, E.S.; Anderson, A.; Ma, C.; Bestick, A.; Parvathaneni, U.; Bhatia, S.; et al. Response rates and durability of chemotherapy among 62 patients with metastatic Merkel cell carcinoma. Cancer Med. 2016, 5, 2294–2301. [Google Scholar] [CrossRef]
  47. Patel, P.; Hussain, K. Merkel cell carcinoma. Clin. Exp. Dermatol. 2021, 46, 814–819. [Google Scholar] [CrossRef]
  48. Paulson, K.G.; Park, S.Y.; Vandeven, N.A.; Lachance, K.; Thomas, H.; Chapuis, A.G.; Harms, K.L.; Thompson, J.A.; Bhatia, S.; Stang, A.; et al. Merkel cell carcinoma: Current US incidence and projected increases based on changing demographics. J. Am. Acad. Dermatol. 2018, 78, 457–463. [Google Scholar] [CrossRef]
  49. Liu, W.; MacDonald, M.; You, J. Merkel cell polyomavirus infection and Merkel cell carcinoma. Curr. Opin. Virol. 2016, 20, 20–27. [Google Scholar] [CrossRef] [Green Version]
  50. Becker, J.C.; Stang, A.; Hausen, A.Z.; Fischer, N.; DeCaprio, J.A.; Tothill, R.W.; Lyngaa, R.; Hansen, U.K.; Ritter, C.; Nghiem, P.; et al. Epidemiology, biology and therapy of Merkel cell carcinoma: Conclusions from the EU project IMMOMEC. Cancer Immunol. Immunother. 2018, 67, 341–351. [Google Scholar] [CrossRef]
  51. Akaike, T.; Nghiem, P. Scientific and clinical developments in Merkel cell carcinoma: A polyomavirus-driven, often-lethal skin cancer. J. Dermatol. Sci. 2022, 105, 2–10. [Google Scholar] [CrossRef]
  52. Sauer, C.M.; Haugg, A.M.; Chteinberg, E.; Rennspiess, D.; Winnepenninckx, V.; Speel, E.J.; Becker, J.C.; Kurz, A.K.; Zur Hausen, A. Reviewing the current evidence supporting early B-cells as the cellular origin of Merkel cell carcinoma. Crit. Rev. Oncol. Hematol. 2017, 116, 99–105. [Google Scholar] [CrossRef] [PubMed]
  53. Wong, S.Q.; Waldeck, K.; Vergara, I.A.; Schröder, J.; Madore, J.; Wilmott, J.S.; Colebatch, A.J.; De Paoli-Iseppi, R.; Li, J.; Lupat, R.; et al. UV-Associated Mutations Underlie the Etiology of MCV-Negative Merkel Cell Carcinomas. Cancer Res. 2015, 75, 5228–5234. [Google Scholar] [CrossRef] [PubMed]
  54. Harms, P.W.; Harms, K.L.; Moore, P.S.; DeCaprio, J.A.; Nghiem, P.; Wong, M.K.K.; Brownell, I. International Workshop on Merkel Cell Carcinoma Research (IWMCC) Working Group. The biology and treatment of Merkel cell carcinoma: Current understanding and research priorities. Nat. Rev. Clin. Oncol. 2018, 15, 763–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. DeCaprio, J.A. Merkel cell polyomavirus and Merkel cell carcinoma. Phil. Trans. R. Soc. 2017, 372, 20160276. [Google Scholar] [CrossRef]
  56. Gravemeyer, J.; Spassova, I.; Verhaegen, M.E.; Dlugosz, A.A.; Hoffmann, D.; Lange, A.; Becker, J.C. DNA-methylation patterns imply a common cellular origin of virus- and UV-associated Merkel cell carcinoma. Oncogene 2022, 41, 37–45. [Google Scholar] [CrossRef]
  57. Ahmed, M.M.; Cushman, C.H.; DeCaprio, J.A. Merkel Cell Polyomavirus: Oncogenesis in a Stable Genome. Viruses 2022, 14, 58. [Google Scholar] [CrossRef]
  58. Schadendorf, D.; Lebbé, C.; Zur Hausen, A.; Avril, M.F.; Hariharan, S.; Bharmal, M.; Becker, J.C. Merkel cell carcinoma: Epidemiology, prognosis, therapy and unmet medical needs. Eur. J. Cancer 2017, 71, 53–69. [Google Scholar] [CrossRef] [Green Version]
  59. Becker, J.C.; Kauczok, C.S.; Ugurel, S.; Eib, S.; Bröcker, E.B.; Houben, R. Merkel cell carcinoma: Molecular pathogenesis, clinical features and therapy. J. Dtsch. Dermatol. Ges. 2008, 6, 709–719. [Google Scholar] [CrossRef]
  60. Horny, K.; Gerhardt, P.; Hebel-Cherouny, A.; Wülbeck, C.; Utikal, J.; Becker, J.C. Mutational Landscape of Virus- and UV-Associated Merkel Cell Carcinoma Cell Lines Is Comparable to Tumor Tissue. Cancers 2021, 13, 649. [Google Scholar] [CrossRef]
  61. DeCaprio, J.A. Molecular Pathogenesis of Merkel Cell Carcinoma. Annu. Rev. Pathol. 2021, 16, 69–91. [Google Scholar] [CrossRef]
  62. Knepper, T.C.; Montesion, M.; Russell, J.S.; Sokol, E.S.; Frampton, G.M.; Miller, V.A.; Albacker, L.A.; McLeod, H.L.; Eroglu, Z.; Khushalani, N.I.; et al. The Genomic Landscape of Merkel Cell Carcinoma and Clinicogenomic Biomarkers of Response to Immune Checkpoint Inhibitor Therapy. Clin. Cancer Res. 2019, 25, 5961–5971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Goh, G.; Walradt, T.; Markarov, V.; Blom, A.; Riaz, N.; Doumani, R.; Stafstrom, K.; Moshiri, A.; Yelistratova, L.; Levinsohn, J.; et al. Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget 2016, 7, 3403–3415. [Google Scholar] [CrossRef] [PubMed]
  64. Harold, A.; Amako, Y.; Hachisuka, J.; Bai, Y.; Li, M.Y.; Kubat, L.; Gravemeyer, J.; Franks, J.; Gibbs, J.R.; Park, H.J. Conversion of Sox2-dependent Merkel cell carcinoma to a differentiated neuron-like phenotype by T antigen inhibition. Proc. Natl. Acad. Sci. USA 2019, 116, 20104–20114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Martel-Jantin, C.; Filippone, C.; Cassar, O.; Peter, M.; Tomasic, G.; Vielh, P.; Brière, J.; Petrella, T.; Aubriot-Lorton, M.H.; Mortier, L.; et al. Genetic variability and integration of Merkel cell polyomavirus in Merkel cell carcinoma. Virology 2012, 426, 134–142. [Google Scholar] [CrossRef]
  66. Starrett, G.J.; Thakuria, M.; Chen, T.; Marcelus, C.; Cheng, J.; Nomburg, J.; Thorner, A.R.; Slevin, M.K.; Powers, W.; Burns, R.T.; et al. Clinical and molecular characterization of virus-positive and virus-negative Merkel cell carcinoma. Genome Med. 2020, 12, 30. [Google Scholar] [CrossRef] [Green Version]
  67. Czech-Sioli, M.; Günther, T.; Therre, M.; Spohn, M.; Indenbirken, D.; Theiss, J.; Riethdorf, S.; Qi, M.; Alawi, M.; Wülbeck, C.; et al. High-resolution analysis of Merkel Cell Polyomavirus in Merkel Cell Carcinoma reveals distinct integration patterns and suggests NHEJ and MMBIR as underlying mechanisms. PLoS Pathog. 2020, 16, e1008562. [Google Scholar] [CrossRef]
  68. Spurgeon, M.E.; Cheng, J.; Ward-Shaw, E.; Dick, F.A.; DeCaprio, J.A.; Lambert, P.F. Merkel cell polyomavirus large T antigen binding to pRb promotes skin hyperplasia and tumor development. PLoS Pathog. 2022, 18, e1010551. [Google Scholar] [CrossRef]
  69. Houben, R.; Shuda, M.; Weinkam, R.; Schrama, D.; Feng, H.; Chang, Y.; Moore, P.S.; Becker, J.C. Merkel cell polyomavirus-infected Merkel cell carcinoma cells require expression of viral T antigens. J. Virol. 2010, 84, 7064–7072. [Google Scholar] [CrossRef] [Green Version]
  70. Ma, J.E.; Brewer, J.D. Merkel cell carcinoma in immunosuppressed patients. Cancers 2014, 6, 1328–1350. [Google Scholar] [CrossRef]
  71. Koljonen, V.; Kukko, H.; Pukkala, E.; Sankila, R.; Böhling, T.; Tukiainen, E.; Sihto, H.; Joensuu, H. Chronic lymphocytic leukaemia patients have a high risk of Merkel-cell polyomavirus DNA-positive Merkel-cell carcinoma. Br. J. Cancer 2009, 101, 1444–1447. [Google Scholar] [CrossRef]
  72. Wijaya, W.A.; Liu, Y.; Qing, Y.; Li, Z. Prevalence of Merkel Cell Polyomavirus in Normal and Lesional Skin: A Systematic Review and Meta-Analysis. Front. Oncol. 2022, 12, 868781. [Google Scholar] [CrossRef] [PubMed]
  73. Pasternak, S.; Carter, M.D.; Ly, T.Y.; Doucette, S.; Walsh, N.M. Immunohistochemical profiles of different subsets of Merkel cell carcinoma. Hum. Pathol. 2018, 82, 232–238. [Google Scholar] [CrossRef] [PubMed]
  74. Jaeger, T.; Ring, J.; Andres, C. Histological, immunohistological, and clinical features of merkel cell carcinoma in correlation to merkel cell polyomavirus status. J. Skin Cancer 2012, 2012, 983421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ursu, R.G.; Damian, C.; Porumb-Andrese, E.; Ghetu, N.; Cobzaru, R.G.; Lunca, C.; Ripa, C.; Costin, D.; Jelihovschi, I.; Petrariu, F.D.; et al. Merkel Cell Polyoma Virus and Cutaneous Human Papillomavirus Types in Skin Cancers: Optimal Detection Assays, Pathogenic Mechanisms, and Therapeutic Vaccination. Pathogens 2022, 11, 479. [Google Scholar] [CrossRef] [PubMed]
  76. Loyo, M.; Guerrero-Preston, R.; Brait, M.; Hoque, M.O.; Chuang, A.; Kim, M.S.; Sharma, R.; Liégeois, N.J.; Koch, W.M.; Califano, J.A. Quantitative detection of Merkel cell virus in human tissues and possible mode of transmission. Int. J. Cancer 2010, 126, 2991–2996. [Google Scholar] [CrossRef] [Green Version]
  77. Nghiem, P.; Bhatia, S.; Lipson, E.J.; Sharfman, W.H.; Kudchadkar, R.R.; Brohl, A.S.; Friedlander, P.A.; Daud, A.; Kluger, H.M.; Reddy, S.A.; et al. Three-year survival, correlates and salvage therapies in patients receiving first-line pembrolizumab for advanced Merkel cell carcinoma. J. Immunother. Cancer 2021, 9, e002478. [Google Scholar] [CrossRef]
  78. Rabinowits, G.; Lezcano, C.; Catalano, P.J.; McHugh, P.; Becker, H.; Reilly, M.M.; Huang, J.; Tyagi, A.; Thakuria, M.; Bresler, S.C.; et al. Cabozantinib in Patients with Advanced Merkel Cell Carcinoma. Oncologist 2018, 23, 814–821. [Google Scholar] [CrossRef] [Green Version]
  79. Leroy, V.; Gerard, E.; Dutriaux, C.; Prey, S.; Gey, A.; Mertens, C.; Beylot-Barry, M.; Pham-Ledard, A. Adverse events need for hospitalization and systemic immunosuppression in very elderly patients (over 80 years) treated with ipilimumab for metastatic melanoma. Cancer Immunol. Immunother. 2019, 68, 545–551. [Google Scholar] [CrossRef]
  80. Johnson, D.B.; Sullivan, R.J.; Menzies, A.M. Immune checkpoint inhibitors in challenging populations. Cancer 2017, 123, 1904–1911. [Google Scholar] [CrossRef] [Green Version]
  81. Tabachnick-Cherny, S.; Pulliam, T.; Church, C.; Koelle, D.M.; Nghiem, P. Polyomavirus-driven Merkel cell carcinoma: Prospects for therapeutic vaccine development. Mol. Carcinog. 2020, 59, 807–821. [Google Scholar] [CrossRef]
  82. Csoboz, B.; Rasheed, K.; Sveinbjørnsson, B.; Moens, U. Merkel cell polyomavirus and non-Merkel cell carcinomas: Guilty or circumstantial evidence? APMIS 2020, 128, 104–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Song, Y.; Gyarmati, P. Identification of Merkel Cell Polyomavirus from a Patient with Acute Myeloid Leukemia. Genome Announc. 2017, 5, e01241-16. [Google Scholar] [CrossRef] [PubMed]
  84. Hashida, Y.; Imajoh, M.; Taniguchi, A.; Kamioka, M.; Daibata, M. Absence of Merkel cell polyomavirus in monocytic leukemias. Acta. Haematol. 2013, 130, 135–137. [Google Scholar] [CrossRef] [PubMed]
  85. Dereure, O.; Cheval, J.; Du Thanh, A.; Pariente, K.; Sauvage, V.; Claude Manuguerra, J.; Caro, V.; Foulongne, V.; Eloit, M. No evidence for viral sequences in mycosis fungoides and Sézary syndrome skin lesions: A high-throughput sequencing approach. J. Investig. Dermatol. 2013, 133, 853–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Mirvish, E.D.; Pomerantz, R.G.; Geskin, L.J. Infectious agents in cutaneous T-cell lymphoma. J. Am. Acad. Dermatol. 2011, 64, 423–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Andres, C.; Belloni, B.; Puchta, U.; Sander, C.A.; Flaig, M.J. Prevalence of MCPyV in Merkel cell carcinoma and non-MCC tumors. J. Cutan. Pathol. 2010, 37, 28–34. [Google Scholar] [CrossRef] [PubMed]
  88. Kreuter, A.; Silling, S.; Dewan, M.; Stücker, M.; Wieland, U. Evaluation of 4 recently discovered human polyomaviruses in primary cutaneous B-cell and T-cell lymphoma. Arch. Dermatol. 2011, 147, 1449–1451. [Google Scholar] [CrossRef] [Green Version]
  89. Shuda, M.; Arora, R.; Kwun, H.J.; Feng, H.; Sarid, R.; Fernández-Figueras, M.T.; Tolstov, Y.; Gjoerup, O.; Mansukhani, M.M.; Swerdlow, S.H.; et al. Human Merkel cell polyomavirus infection I. MCV T antigen expression in Merkel cell carcinoma, lymphoid tissues and lymphoid tumors. Int. J. Cancer 2009, 125, 1243–1249. [Google Scholar] [CrossRef]
  90. Andres, C.; Puchta, U.; Sander, C.A.; Ruzicka, T.; Flaig, M.J. Prevalence of Merkel cell polyomavirus DNA in cutaneous lymphomas, pseudolymphomas, and inflammatory skin diseases. Am. J. Dermatopathol. 2010, 32, 593–598. [Google Scholar] [CrossRef]
  91. Jeon, Y.W.; Cho, S.G. Chronic lymphocytic leukemia: A clinical review including Korean cohorts. Korean J. Intern. Med. 2016, 31, 433–443. [Google Scholar] [CrossRef] [PubMed]
  92. Cimino, P.J., Jr.; Bahler, D.W.; Duncavage, E.J. Detection of Merkel cell polyomavirus in chronic lymphocytic leukemia T-cells. Exp. Mol. Pathol. 2013, 94, 40–44. [Google Scholar] [CrossRef] [PubMed]
  93. Howard, R.A.; Dores, G.M.; Curtis, R.E.; Anderson, W.F.; Travis, L.B. Merkel cell carcinoma and multiple primary cancers. Cancer Epidemiol. Biomark. Prev. 2006, 15, 1545–1549. [Google Scholar] [CrossRef] [PubMed]
  94. Pantulu, N.D.; Pallasch, C.P.; Kurz, A.K.; Kassem, A.; Frenzel, L.; Sodenkamp, S.; Kvasnicka, H.M.; Wendtner, C.M.; Zur Hausen, A. Detection of a novel truncating Merkel cell polyomavirus large T antigen deletion in chronic lymphocytic leukemia cells. Blood 2010, 116, 5280–5284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Prezioso, C.; Di Lella, F.M.; Rodio, D.M.; Bitossi, C.; Trancassini, M.; Mele, A.; de Vito, C.; Antonelli, G.; Pietropaolo, V. Merkel Cell Polyomavirus DNA Detection in Respiratory Samples: Study of a Cohort of Patients Affected by Cystic Fibrosis. Viruses 2019, 11, 571. [Google Scholar] [CrossRef] [Green Version]
  96. Imajoh, M.; Hashida, Y.; Taniguchi, A.; Kamioka, M.; Daibata, M. Novel human polyomaviruses, Merkel cell polyomavirus and human polyomavirus 9, in Japanese chronic lymphocytic leukemia cases. J. Hematol. Oncol. 2012, 5, 25. [Google Scholar] [CrossRef] [Green Version]
  97. Trizuljak, J.; Srovnal, J.; Plevová, K.; Brychtová, Y.; Semerád, L.; Bakešová, D.; Létalová, E.; Benedíková, A.; Mayer, J.; Hajdúch, M.; et al. Analysis of prognostic significance of merkel cell polyomavirus in chronic lymphocytic leukemia. Clin. Lymphoma Myeloma Leuk. 2015, 15, 439–442. [Google Scholar] [CrossRef]
  98. Haugg, A.M.; Speel, E.J.; Pantulu, N.D.; Pallasch, C.; Kurz, A.K.; Kvasnicka, H.M.; Cathomas, G.; Wendtner, C.M.; zur Hausen, A. Fluorescence in situ hybridization confirms the presence of Merkel cell polyomavirus in chronic lymphocytic leukemia cells. Blood 2011, 117, 5776–5777. [Google Scholar] [CrossRef] [Green Version]
  99. Teman, C.J.; Tripp, S.R.; Perkins, S.L.; Duncavage, E.J. Merkel cell polyomavirus (MCPyV) in chronic lymphocytic leukemia/small lymphocytic lymphoma. Leuk. Res. 2011, 35, 689–692. [Google Scholar] [CrossRef]
  100. Katano, H.; Ito, H.; Suzuki, Y.; Nakamura, T.; Sato, Y.; Tsuji, T.; Matsuo, K.; Nakagawa, H.; Sata, T. Detection of Merkel cell polyomavirus in Merkel cell carcinoma and Kaposi’s sarcoma. J. Med. Virol. 2009, 81, 1951–1958. [Google Scholar] [CrossRef]
  101. Paik, J.Y.; Hall, G.; Clarkson, A.; Lee, L.; Toon, C.; Colebatch, A.; Chou, A.; Gill, A.J. Immunohistochemistry for Merkel cell polyomavirus is highly specific but not sensitive for the diagnosis of Merkel cell carcinoma in the Australian population. Hum. Pathol. 2011, 42, 1385–1390. [Google Scholar] [CrossRef]
  102. Yahyapour, Y.; Sadeghi, F.; Alizadeh, A.; Rajabnia, R.; Siadati, S. Detection of Merkel Cell Polyomavirus and Human Papillomavirus in Esophageal Squamous Cell Carcinomas and Non-Cancerous Esophageal Samples in Northern Iran. Pathol. Oncol. Res. 2016, 22, 667–672. [Google Scholar] [CrossRef]
  103. Chen, A.A.; Gheit, T.; Stellin, M.; Lupato, V.; Spinato, G.; Fuson, R.; Menegaldo, A.; Mckay-Chopin, S.; Dal Cin, E.; Tirelli, G.; et al. Oncogenic DNA viruses found in salivary gland tumors. Oral Oncol. 2017, 75, 106–110. [Google Scholar] [CrossRef] [PubMed]
  104. Mohebbi, E.; Noormohamadi, Z.; Sadeghi-Rad, H.; Sadeghi, F.; Yahyapour, Y.; Vaziri, F.; Rahimi, A.; Rahimi Jamnani, F.; Mehrabi, S.; Siadat, S.D.; et al. Low viral load of Merkel cell polyomavirus in Iranian patients with head and neck squamous cell carcinoma: Is it clinically important? J. Med. Virol. 2018, 90, 344–350. [Google Scholar] [CrossRef] [PubMed]
  105. Yahyapour, Y.; Rahmani, R.; Alipour, M.; Alizadeh, A.; Khademian, A.; Sadeghi, F. Prevalence and association of human papillomavirus, Epstein-Barr virus and Merkel Cell polyomavirus with neoplastic esophageal lesions in northern Iran. Caspian J. Intern. Med. 2018, 9, 353–360. [Google Scholar] [PubMed]
  106. Ly, T.Y.; Walsh, N.M.; Pasternak, S. The spectrum of Merkel cell polyomavirus expression in Merkel cell carcinoma, in a variety of cutaneous neoplasms, and in neuroendocrine carcinomas from different anatomical sites. Hum. Pathol. 2012, 43, 557–566. [Google Scholar] [CrossRef] [PubMed]
  107. Touzé, A.; Gaitan, J.; Maruani, A.; Le Bidre, E.; Doussinaud, A.; Clavel, C.; Durlach, A.; Aubin, F.; Guyétant, S.; Lorette, G.; et al. Merkel cell polyomavirus strains in patients with merkel cell carcinoma. Emerg. Infect. Dis. 2009, 15, 960–962. [Google Scholar] [CrossRef] [PubMed]
  108. Rodig, S.J.; Cheng, J.; Wardzala, J.; DoRosario, A.; Scanlon, J.J.; Laga, A.C.; Martinez-Fernandez, A.; Barletta, J.A.; Bellizzi, A.M.; Sadasivam, S.; et al. Improved detection suggests all Merkel cell carcinomas harbor Merkel polyomavirus. J. Clin. Investig. 2012, 122, 4645–4653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Toptan, T.; Yousem, S.A.; Ho, J.; Matsushima, Y.; Stabile, L.P.; Fernández-Figueras, M.T.; Bhargava, R.; Ryo, A.; Moore, P.S.; Chang, Y. Survey for human polyomaviruses in cancer. JCI Insight 2016, 1, e85562. [Google Scholar] [CrossRef] [Green Version]
  110. Fiorina, L.; Ricotti, M.; Vanoli, A.; Luinetti, O.; Dallera, E.; Riboni, R.; Paolucci, S.; Brugnatelli, S.; Paulli, M.; Pedrazzoli, P.; et al. Systematic analysis of human oncogenic viruses in colon cancer revealed EBV latency in lymphoid infiltrates. Infect. Agent Cancer 2014, 9, 18. [Google Scholar] [CrossRef] [Green Version]
  111. Campello, C.; Comar, M.; D’Agaro, P.; Minicozzi, A.; Rodella, L.; Poli, A. A molecular case-control study of the Merkel cell polyomavirus in colon cancer. J. Med. Virol 2011, 83, 721–724. [Google Scholar] [CrossRef] [Green Version]
  112. Kassem, A.; Technau, K.; Kurz, A.K.; Pantulu, D.; Löning, M.; Kayser, G.; Stickeler, E.; Weyers, W.; Diaz, C.; Werner, M. Merkel cell polyomavirus sequences are frequently detected in nonmelanoma skin cancer of immunosuppressed patients. Int. J. Cancer. 2009, 125, 356–361. [Google Scholar] [CrossRef] [PubMed]
  113. Jung, H.S.; Choi, Y.; Choi, J.S.; Roh, J.H.; Pyon, J.K.; Woo, K.J.; Lee, E.H.; Jang, K.T.; Han, J.; Park, C.S. Detection of Merkel cell polyomavirus in Merkel cell carcinomas and small cell carcinomas by PCR and immunohistochemistry. Histol. Histopathol. 2011, 26, 1231–1241. [Google Scholar] [PubMed]
  114. Ramqvist, T.; Ursu, R.G.; Haeggblom, L.; Mirzaie, L.; Gahm, C.; Hammarstedt-Nordenvall, L.; Dalianis, T.; Näsman, A. Human Polyomaviruses Are Not Frequently Present in Cancer of the Salivary Glands. Anticancer Res. 2018, 38, 2871–2874. [Google Scholar] [PubMed]
  115. Tanio, S.; Matsushita, M.; Kuwamoto, S.; Horie, Y.; Kodani, I.; Murakami, I.; Ryoke, K.; Hayashi, K. Low prevalence of Merkel cell polyomavirus with low viral loads in oral and maxillofacial tumours or tumour-like lesions from immunocompetent patients: Absence of Merkel cell polyomavirus-associated neoplasms. Mol. Clin. Oncol. 2015, 3, 1301–1306. [Google Scholar] [CrossRef] [Green Version]
  116. Purdie, K.J.; Proby, C.M.; Rizvi, H.; Griffin, H.; Doorbar, J.; Sommerlad, M.; Feltkamp, M.C.; der Meijden, E.V.; Inman, G.J.; South, A.P. The Role of Human Papillomaviruses and Polyomaviruses in BRAF-Inhibitor Induced Cutaneous Squamous Cell Carcinoma and Benign Squamoproliferative Lesions. Front. Microbiol. 2018, 9, 1806. [Google Scholar] [CrossRef] [Green Version]
  117. Schrama, D.; Groesser, L.; Ugurel, S.; Hafner, C.; Pastrana, D.V.; Buck, C.B.; Cerroni, L.; Theiler, A.; Becker, J.C. Presence of human polyomavirus 6 in mutation-specific BRAF inhibitor-induced epithelial proliferations. JAMA Dermatol. 2014, 150, 1180–1186. [Google Scholar] [CrossRef] [Green Version]
  118. Dworkin, A.M.; Tseng, S.Y.; Allain, D.C.; Iwenofu, O.H.; Peters, S.B.; Toland, A.E. Merkel cell polyomavirus in cutaneous squamous cell carcinoma of immunocompetent individuals. J. Investig. Dermatol. 2009, 129, 2868–2874. [Google Scholar] [CrossRef] [Green Version]
  119. Rollison, D.E.; Giuliano, A.R.; Messina, J.L.; Fenske, N.A.; Cherpelis, B.S.; Sondak, V.K.; Roetzheim, R.G.; Iannacone, M.R.; Michael, K.M.; Gheit, T.; et al. Case-control study of Merkel cell polyomavirus infection and cutaneous squamous cell carcinoma. Cancer Epidemiol. Biomark. Prev. 2012, 21, 74–81. [Google Scholar] [CrossRef] [Green Version]
  120. Scola, N.; Wieland, U.; Silling, S.; Altmeyer, P.; Stücker, M.; Kreuter, A. Prevalence of human polyomaviruses in common and rare types of non-Merkel cell carcinoma skin cancer. Br. J. Dermatol. 2012, 167, 1315–1320. [Google Scholar] [CrossRef]
  121. Imajoh, M.; Hashida, Y.; Nakajima, H.; Sano, S.; Daibata, M. Prevalence and viral DNA loads of three novel human polyomaviruses in skin cancers from Japanese patients. J. Dermatol. 2013, 40, 657–660. [Google Scholar] [CrossRef]
  122. Bellott, T.R.; Baez, C.F.; Almeida, S.G.; Venceslau, M.T.; Zalis, M.G.; Guimarães, M.A.; Rochael, M.C.; Luz, F.B.; Varella, R.B.; Almeida, J.R. Molecular prevalence of Merkel cell polyomavirus in nonmelanoma skin cancer in a Brazilian population. Clin. Exp. Dermatol. 2017, 42, 390–394. [Google Scholar] [CrossRef] [PubMed]
  123. Haeggblom, L.; Franzén, J.; Näsman, A. Human polyomavirus DNA detection in keratoacanthoma and Spitz naevus: No evidence for a causal role. J. Clin. Pathol. 2017, 70, 451–453. [Google Scholar] [CrossRef] [PubMed]
  124. Du-Thanh, A.; Guillot, B.; Dereure, O.; Foulongne, V. Detection of Merkel cell and other human polyomavirus DNA in lesional and nonlesional skin from patients with Kaposi sarcoma. Br. J. Derm. 2015, 173, 1063–1065. [Google Scholar] [CrossRef]
  125. Urso, C.; Pierucci, F.; Sollai, M.; Arvia, R.; Massi, D.; Zakrzewska, K. Detection of Merkel cell polyomavirus and human papillomavirus DNA in porocarcinoma. J. Clin. Virol. 2016, 78, 71–73. [Google Scholar] [CrossRef]
  126. Koburger, I.; Meckbach, D.; Metzler, G.; Fauser, U.; Garbe, C.; Bauer, J. Absence of merkel cell polyoma virus in cutaneous melanoma. Exp. Dermatol. 2011, 20, 78–79. [Google Scholar] [CrossRef] [PubMed]
  127. Murakami, M.; Imajoh, M.; Ikawa, T.; Nakajima, H.; Kamioka, M.; Nemoto, Y.; Ujihara, T.; Uchiyama, J.; Matsuzaki, S.; Sano, S.; et al. Presence of Merkel cell polyomavirus in Japanese cutaneous squamous cell carcinoma. J. Clin. Virol. 2011, 50, 37–41. [Google Scholar] [CrossRef] [PubMed]
  128. Saláková, M.; Košlabová, E.; Vojtěchová, Z.; Tachezy, R.; Šroller, V. Detection of human polyomaviruses MCPyV, HPyV6, and HPyV7 in malignant and non-malignant tonsillar tissues. J. Med. Virol. 2016, 88, 695–702. [Google Scholar] [CrossRef] [PubMed]
  129. Herberhold, S.; Hellmich, M.; Panning, M.; Bartok, E.; Silling, S.; Akgül, B.; Wieland, U. Human polyomavirus and human papillomavirus prevalence and viral load in non-malignant tonsillar tissue and tonsillar carcinoma. Med. Microbiol. Immunol. 2017, 206, 93–103. [Google Scholar] [CrossRef]
  130. Hashida, Y.; Imajoh, M.; Nemoto, Y.; Kamioka, M.; Taniguchi, A.; Taguchi, T.; Kume, M.; Orihashi, K.; Daibata, M. Detection of Merkel cell polyomavirus with a tumour-specific signature in non-small cell lung cancer. Br. J. Cancer 2013, 108, 629–637. [Google Scholar] [CrossRef]
  131. Chteinberg, E.; Klufah, F.; Rennspiess, D.; Mannheims, M.F.; Abdul-Hamid, M.A.; Losen, M.; Keijzers, M.; De Baets, M.H.; Kurz, A.K.; Zur Hausen, A. Low prevalence of Merkel cell polyomavirus in human epithelial thymic tumors. Thorac Cancer. 2019, 10, 445–451. [Google Scholar] [CrossRef] [Green Version]
  132. Cason, C.; Monasta, L.; Zanotta, N.; Campisciano, G.; Maestri, I.; Tommasino, M.; Pawlita, M.; Villani, S.; Comar, M.; Delbue, S. Antibody response to polyomavirus primary infection: High seroprevalence of Merkel cell polyomavirus and lymphoid tissue involvement. J. Neurovirol. 2018, 24, 314–322. [Google Scholar] [CrossRef] [PubMed]
  133. Mancuso, G.; Antona, J.; Sirini, C.; Salvo, M.; Giacometti, L.; Olivero, C.; Trisolini, E.; Indellicato, R.; Boldorini, R. Frequent detection of Merkel cell polyomavirus DNA in tissues from 10 consecutive autopsies. J. Gen. Virol. 2017, 98, 1372–1376. [Google Scholar] [CrossRef] [PubMed]
  134. Schmitt, M.; Höfler, D.; Koleganova, N.; Pawlita, M. Human polyomaviruses and other human viruses in neuroendocrine tumors. Cancer Epidemiol. Biomark. Prev. 2011, 20, 1558–1561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Dang, X.; Bialasiewicz, S.; Nissen, M.D.; Sloots, T.P.; Koralnik, I.J.; Tan, C.S. Infrequent detection of KI, WU and MC polyomaviruses in immunosuppressed individuals with or without progressive multifocal leukoencephalopathy. PLoS ONE 2011, 6, e16736. [Google Scholar] [CrossRef] [PubMed]
  136. Lam, W.Y.; Leung, B.W.; Chu, I.M.; Chan, A.C.; Ng, H.K.; Chan, P.K. Survey for the presence of BK, JC, KI, WU and Merkel cell polyomaviruses in human brain tissues. J. Clin. Virol. 2010, 48, 11–14. [Google Scholar] [CrossRef]
  137. Toracchio, S.; Foyle, A.; Sroller, V.; Reed, J.A.; Wu, J.; Kozinetz, C.A.; Butel, J.S. Lymphotropism of Merkel cell polyomavirus infection, Nova Scotia, Canada. Emerg. Infect. Dis. 2010, 16, 1702–1709. [Google Scholar] [CrossRef]
  138. Hashida, Y.; Taniguchi, A.; Yawata, T.; Hosokawa, S.; Murakami, M.; Hiroi, M.; Ueba, T.; Daibata, M. Prevalence of human cytomegalovirus, polyomaviruses, and oncogenic viruses in glioblastoma among Japanese subjects. Infect. Agent Cancer 2015, 10, 3. [Google Scholar] [CrossRef] [Green Version]
  139. Matthay, K.; Maris, J.; Schleiermacher, G.; Nakagawara, A.; Mackall, C.L.; Diller, L.; Weiss, W. Neuroblastoma. Nat. Rev. Dis. Primers 2016, 2, 1–21. [Google Scholar] [CrossRef]
  140. Sastre-Garau, X.; Peter, M.; Avril, M.F.; Laude, H.; Couturier, J.; Rozenberg, F.; Almeida, A.; Boitier, F.; Carlotti, A.; Couturaud, B.; et al. Merkel cell carcinoma of the skin: Pathological and molecular evidence for a causative role of MCV in oncogenesis. J. Pathol. 2009, 218, 48–56. [Google Scholar] [CrossRef]
  141. Giraud, G.; Ramqvist, T.; Pastrana, D.V.; Pavot, V.; Lindau, C.; Kogner, P.; Orrego, A.; Buck, C.B.; Allander, T.; Holm, S.; et al. DNA from KI, WU and Merkel cell polyomaviruses is not detected in childhood central nervous system tumours or neuroblastomas. PLoS ONE 2009, 4, e8239. [Google Scholar] [CrossRef] [Green Version]
  142. Shikova, E.; Emin, D.; Alexandrova, D.; Shindov, M.; Kumanova, A.; Lekov, A.; Moens, U. Detection of Merkel Cell Polyomavirus in Respiratory Tract Specimens. Intervirology 2017, 60, 28–32. [Google Scholar] [CrossRef]
  143. Karimi, S.; Yousefi, F.; Seifi, S.; Khosravi, A.; Nadji, S.A. No evidence for a role of Merkel cell polyomavirus in small cell lung cancer among Iranian subjects. Pathol. Res. Pract. 2014, 210, 836–839. [Google Scholar] [CrossRef] [PubMed]
  144. Wetzels, C.T.; Hoefnagel, J.G.; Bakkers, J.M.; Dijkman, H.B.; Blokx, W.A.; Melchers, W.J. Ultrastructural proof of polyomavirus in Merkel cell carcinoma tumour cells and its absence in small cell carcinoma of the lung. PLoS ONE 2009, 4, e4958. [Google Scholar] [CrossRef] [PubMed]
  145. Helmbold, P.; Lahtz, C.; Herpel, E.; Schnabel, P.A.; Dammann, R.H. Frequent hypermethylation of RASSF1A tumour suppressor gene promoter and presence of Merkel cell polyomavirus in small cell lung cancer. Eur. J. Cancer 2009, 45, 2207–2211. [Google Scholar] [CrossRef] [PubMed]
  146. Andres, C.; Ihrler, S.; Puchta, U.; Flaig, M.J. Merkel cell polyomavirus is prevalent in a subset of small cell lung cancer: A study of 31 patients. Thorax 2009, 64, 1007–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Behdarvand, A.; Zamani, M.S.; Sadeghi, F.; Yahyapour, Y.; Vaziri, F.; Jamnani, F.R.; Nowruzi, B.; Fateh, A.; Siadat, S.D. Evaluation of Merkel cell polyomavirus in non-small cell lung cancer and adjacent normal cells. Microb. Pathog. 2017, 108, 21–26. [Google Scholar] [CrossRef]
  148. Kim, G.J.; Lee, J.H.; Lee, D.H. Clinical and prognostic significance of Merkel cell polyomavirus in nonsmall cell lung cancer. Medicine 2017, 96, e5413. [Google Scholar] [CrossRef]
  149. Joh, J.; Jenson, A.B.; Moore, G.D.; Rezazedeh, A.; Slone, S.P.; Ghim, S.J.; Kloecker, G.H. Human papillomavirus (HPV) and Merkel cell polyomavirus (MCPyV) in non small cell lung cancer. Exp. Mol. Pathol. 2010, 89, 222–226. [Google Scholar] [CrossRef]
  150. Trejo Bittar, H.E.; Pantanowitz, L. Merkel Cell Polyomavirus is Not Detected in Lung Adenocarcinomas by Immunohistochemistry. Appl. Immunohistochem Mol. Morphol. 2016, 24, 427–430. [Google Scholar] [CrossRef]
  151. Lasithiotaki, I.; Antoniou, K.M.; Derdas, S.P.; Sarchianaki, E.; Symvoulakis, E.K.; Psaraki, A.; Spandidos, D.A.; Stathopoulos, E.N.; Siafakas, N.M.; Sourvinos, G. The presence of Merkel cell polyomavirus is associated with deregulated expression of BRAF and Bcl-2 genes in non-small cell lung cancer. Int. J. Cancer 2013, 133, 604–611. [Google Scholar] [CrossRef]
  152. Lasithiotaki, I.; Tsitoura, E.; Koutsopoulos, A.; Lagoudaki, E.; Koutoulaki, C.; Pitsidianakis, G.; Spandidos, D.A.; Siafakas, N.M.; Sourvinos, G.; Antoniou, K.M. Aberrant expression of miR-21, miR-376c and miR-145 and their target host genes in Merkel cell polyomavirus-positive non-small cell lung cancer. Oncotarget 2016, 8, 112371–112383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Smelov, V.; Bzhalava, D.; Arroyo Mühr, L.S.; Eklund, C.; Komyakov, B.; Gorelov, A.; Dillner, J.; Hultin, E. Detection of DNA viruses in prostate cancer. Sci/ Rep. 2016, 6, 25235. [Google Scholar] [CrossRef] [PubMed]
  154. Rawla, P. Epidemiology of Prostate Cancer. World J. Oncol. 2019, 10, 63–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Salehi-Vaziri, M.; Sadeghi, F.; Alamsi-Hashiani, A.; Haeri, H.; Monavari, S.H.; Keyvani, H. Merkel cell polyomavirus and human papillomavirus infections in cervical disease in Iranian women. Arch. Virol. 2015, 160, 1181–1187. [Google Scholar] [CrossRef] [PubMed]
  156. Imajoh, M.; Hashida, Y.; Nemoto, Y.; Oguri, H.; Maeda, N.; Furihata, M.; Fukaya, T.; Daibata, M. Detection of Merkel cell polyomavirus in cervical squamous cell carcinomas and adenocarcinomas from Japanese patients. Virol. J. 2012, 9, 154. [Google Scholar] [CrossRef] [Green Version]
  157. Peng, J.; Wang, T.; Zhu, H.; Guo, J.; Li, K.; Yao, Q.; Lv, Y.; Zhang, J.; He, C.; Chen, J.; et al. Multiplex PCR/mass spectrometry screening of biological carcinogenic agents in human mammary tumors. J. Clin. Virol. 2014, 61, 255–259. [Google Scholar] [CrossRef]
  158. Lei, S.; Zheng, R.; Zhang, S.; Wang, S.; Chen, R.; Sun, K.; Zeng, H.; Zhou, J.; Wei, W. Global patterns of breast cancer incidence and mortality: A population-based cancer registry data analysis from 2000 to 2020. Cancer Commun. 2021, 41, 1183–1194. [Google Scholar] [CrossRef]
  159. Hossain, M.S.; Karuniawati, H.; Jairoun, A.A.; Urbi, Z.; Ooi, J.; John, A.; Lim, Y.C.; Kibria, K.M.K.; Mohiuddin, A.K.M.; Ming, L.C.; et al. Colorectal Cancer: A Review of Carcinogenesis, Global Epidemiology, Current Challenges, Risk Factors, Preventive and Treatment Strategies. Cancers 2022, 14, 1732. [Google Scholar] [CrossRef]
  160. Li, S.; Liu, J.; Zheng, X.; Ren, L.; Yang, Y.; Li, W.; Fu, W.; Wang, J.; Du, G. Tumorigenic bacteria in colorectal cancer: Mechanisms and treatments. Cancer Biol. Med. 2021, 19, 147–162. [Google Scholar] [CrossRef]
  161. Dolci, M.; Signorini, L.; Toumi, W.; Basile, G.; D’Alessandro, S.; Ferrante, P.; Delbue, S. Human polyomaviruses genomes in clinical specimens of colon cancer patients. J. Med. Virol. 2021, 93, 6333–6339. [Google Scholar] [CrossRef]
  162. Jemal, A.; Bray, F.; Center, M.M.; Ferlay, J.; Ward, E.; Forman, D. Global cancer statistics. CA Cancer J. Clin. 2011, 61, 69–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Merriam-Webster Medical. Available online: https://www.merriam-webster.com/medical/soft%20tissue (accessed on 23 July 2022).
  164. NCI Dictionary of Cancer Terms-NCI. Available online: https://www.cancer.gov/publications/dictionaries/cancer-terms/def/soft-tissue?redirect=true (accessed on 23 July 2022).
  165. Houben, R.; Grimm, J.; Willmes, C.; Weinkam, R.; Becker, J.C.; Schrama, D. Merkel cell carcinoma and Merkel cell polyomavirus: Evidence for hit-and-run oncogenesis. J. Investig. Dermatol. 2012, 132, 254–256. [Google Scholar] [CrossRef] [PubMed]
  166. Moens, U.; Van Ghelue, M.; Ehlers, B. Are human polyomaviruses co-factors for cancers induced by other oncoviruses? Rev. Med. Virol. 2014, 24, 343–360. [Google Scholar] [CrossRef] [PubMed]
  167. Vazquez-Guillen, J.M.; Palacios-Saucedo, G.C.; Rivera-Morales, L.G.; Alonzo-Morado, M.V.; Burciaga-Bernal, S.B.; Montufar-Martinez, M.; Ortiz-Lopez, R.; Gonzalez-Villasana, V.; Martinez-Torres, A.C.; Serna-Hernandez, J.C.; et al. Infection and coinfection by human papillomavirus, Epstein-Barr virus and Merkel cell polyomavirus in patients with squamous cell carcinoma of the larynx: A retrospective study. PeerJ 2018, 6, e5834. [Google Scholar] [CrossRef]
Figure 1. The genomic map of MCPyV circular, double stranded DNA. The early genes encode oncogenic proteins (LT, sT, 57 kT) and the late genes structural proteins (VP1. VP2, VP3). The NCCR (non-coding control region) includes Ori and regulates the viral gene expression. A gene, located on the late strand of the T antigen genes, with an opposite transcription orientation (reversed complementarity) to the T antigens produces a non-coding miRNA. The total length of the viral genome is approximately 5.4 bp. Created with BioRender.com (visited on 1 July 2022).
Figure 1. The genomic map of MCPyV circular, double stranded DNA. The early genes encode oncogenic proteins (LT, sT, 57 kT) and the late genes structural proteins (VP1. VP2, VP3). The NCCR (non-coding control region) includes Ori and regulates the viral gene expression. A gene, located on the late strand of the T antigen genes, with an opposite transcription orientation (reversed complementarity) to the T antigens produces a non-coding miRNA. The total length of the viral genome is approximately 5.4 bp. Created with BioRender.com (visited on 1 July 2022).
Cancers 14 05548 g001
Figure 2. The structural domains of the early gene region antigens, LT and sT. The LT and ST are products of alternative splicing of the region. In MCC, a truncated version of LT is expressed, characterized by mutations that eliminate domains of the C-terminus of LT, naming the nuclear localization signal (NLS), origin binding domain (OBD), and helicase domains. The crucial for the oncogenesis LXCXE RB-binding motif is preserved. ST contains several unique motifs with multiple sites that bind and interact with a variety of host cellular proteins. The DnaJ domains, conserved in both antigens, are related to viral replication. NLS, Nuclear localization signal; MUR, MCPyV unique region; OBD, Origin binding domain, PP2A protein phosphatase 2A; EP400, E1A-binding protein p400; MYCL, L-myc-1 proto-oncogene protein; LSD, NF-κΒ, Nuclear factor kappa B; LT-stabilizing domain; FBXW7m F-box/WD repeat-containing protein 7; CDC20, cell division cycle protein 20 homolog; PP4, Protein phosphatase 4.
Figure 2. The structural domains of the early gene region antigens, LT and sT. The LT and ST are products of alternative splicing of the region. In MCC, a truncated version of LT is expressed, characterized by mutations that eliminate domains of the C-terminus of LT, naming the nuclear localization signal (NLS), origin binding domain (OBD), and helicase domains. The crucial for the oncogenesis LXCXE RB-binding motif is preserved. ST contains several unique motifs with multiple sites that bind and interact with a variety of host cellular proteins. The DnaJ domains, conserved in both antigens, are related to viral replication. NLS, Nuclear localization signal; MUR, MCPyV unique region; OBD, Origin binding domain, PP2A protein phosphatase 2A; EP400, E1A-binding protein p400; MYCL, L-myc-1 proto-oncogene protein; LSD, NF-κΒ, Nuclear factor kappa B; LT-stabilizing domain; FBXW7m F-box/WD repeat-containing protein 7; CDC20, cell division cycle protein 20 homolog; PP4, Protein phosphatase 4.
Cancers 14 05548 g002
Table 1. Detection results and method of the MCPyV DNA in tumours derived from different tissues. For the positive cases of the same type of tumours tested, the prevalence of the viral DNA varies greatly.
Table 1. Detection results and method of the MCPyV DNA in tumours derived from different tissues. For the positive cases of the same type of tumours tested, the prevalence of the viral DNA varies greatly.
Histological Tumour TypeSystemMCPyV DNAMethodComplementary FindingsReferences
AMLCirculatory System+qPCR, NGSViral DNA found in only 1 sample[82,83,84]
CTCLCirculatory System+PCR/Southern blot, qPCR-[82,85,86,87,88]
CBCLCirculatory System+PCR/Southern blot, qPCRAll samples tested by IHC were negative for LT[82,88,89,90]
CLL/SLLCirculatory System+PCR, qPCR, FISH, IHC
  • CLL patients have a higher risk of developing VP-MCCs.
  • six samples contained a truncated LT antigen
  • two of the above also harboured full-length LT mRNA
[82,91,92,93,94,95,96,97]
Chronic myelomonocytic leukemiaCirculatory SystemqPCR-[84]
FISH-[98]
Mantle cell lymphomaCirculatory System
Follicular lymphomaCirculatory SystemPCR, qPCR-[99]
Primary effusion lymphomaCirculatory SystemqPCR-[100]
Small-cell carcinoma of the lymph nodesCirculatory SystemIHC-[101]
Esophagus cancerDigestive System+qPCR-[76,82,102]
Liver cancerDigestive System+qPCR, IHC-[82]
Salivary gland cancerDigestive System+qPCR, IHCIn one positive parotid sample truncated LT was detected[82,102,103,104,105]
Gallbladder cancerDigestive SystemqPCR-[37]
Pancreas cancerDigestive SystemqPCR-[37]
Intestine cancerDigestive SystemnPCR-[106,107]
Appendix cancerDigestive SystemIHC-[106]
Gastrointestinal cancerDigestive SystemIHC-[108]
Colon cancerDigestive System+PCR, nPCR, qPCR, FISH, IHC-[82,109,110,111,112,113]
Stomach cancerDigestive System+PCR, IHC-[82,109,113]
Oral cavity cancerDigestive System+PCR, qPCR, IHC-[82,102,104,105,114,115]
Bladder cancerExcretory System+qPCR, IHC-[76,82,109,113]
Renal cancerExcretory System+qPCR, IHC-[76,82,109,113]
SCCIntegumentary System+PCR, nPCR, qPCR, ddPCR, NGS, IHC
  • All samples tested by IHC were negative for LT
  • Truncated LT was found in some samples after sequencing
[112,114,116,117,118,119]
BCCIntegumentary System+PCR, nPCR, IHC, nPCR,All samples tested by IHC were negative for LT[112,120,121,122]
Keratoacanthoma cancerIntegumentary System+PCR, nPCR, qPCRAll samples tested by IHC were negative for LT[82,117,120,123]
Kaposi’s SarcomaIntegumentary System+PCR, nPCR, qPCR-[82,100,124]
PorocarcinomaIntegumentary System+PCR, nPCR, qPCR, ddPCR-[82,114,125]
Atypical fibroxanthomaIntegumentary System+PCR, qPCR, IHC, PCR/Southern blotAll samples tested by IHC were negative for LT[82,120]
MelanomaIntegumentary SystemPCR, nPCRonly 1 documented case, in which acral lentiginous melanomas and nodular melanomas were tested positive[106,121,126,127]
Tonsillar SCCLymphatic System+qPCR-[82,128,129,130,131]
Hypertrophy adenoidLymphatic System+qPCR-[132]
Brain tumoursNervous System+qPCR, nPCR, IHC-[101,109,133,134,135,136]
SchwannomasNervous System+qPCROne examined sample contained LT transcripts[102,115,137]
MeningiomasNervous System+qPCR-[102]
GlioblastomasNervous System+qPCR-[102,138]
NeuroblastomasNervous System-qPCR-[113,139,140,141]
Lung cancerRespiratory System+PCR, qPCR-[37,76,107,117,142,143]
SCLCRespiratory System+PCR, qPCR, nPCR, IHC-[82,107,113,143,144,145,146]
NSCLCRespiratory System+PCR, qPCR, RT-PCR
  • Integrated viral DNA in one adenosarcoma + expression of truncated LT.
  • Integrated + episomal viral DNA in one SCC sample + expression of full-length and truncated LT
  • association between the presence of Merkel cell polyomavirus with a disrupted expression of two genes (BRAF and Bcl-2) as well as virus-specific microRNA signature
[82,130,147,148,149,150,151,152]
Prostate cancerReproductive System+qPCR, qRT-PCR, NGC, IHC-[76,153]
Testicular cancerReproductive System+qPCR, nPCR-[76,154]
Cervical cancerReproductive System+PCR, qPCR, RT-PCR, IHC140 of the examined patients were also HIV positive[82,155,156]
Breast cancerReproductive System+PCR, qPCR, qRT-PCR-[82,157,158]
Rhabdomyosarcoma tumoursSoft TissuePCR-[140]
Desmoplastic tumoursSoft TissuePCR-[140]
Fibromatosis tumoursSoft TissuePCR-[140]
Acute Myeloid Leukemia (AML); Cutaneous T-cell leukemia (CTCL); Cutaneous B-cell lymphomas (CBCL); Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma (CLL/SLL); Squamous Cell Carcinomas (SCC); Basal Cell Carcinomas (BCC); Tonsillar Squamous cell carcinoma (Tonsillar SCC); small-cell lung carcinomas (SCLC), non-small-cell lung carcinomas (NSCLC).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dimitraki, M.G.; Sourvinos, G. Merkel Cell Polyomavirus (MCPyV) and Cancers: Emergency Bell or False Alarm? Cancers 2022, 14, 5548. https://doi.org/10.3390/cancers14225548

AMA Style

Dimitraki MG, Sourvinos G. Merkel Cell Polyomavirus (MCPyV) and Cancers: Emergency Bell or False Alarm? Cancers. 2022; 14(22):5548. https://doi.org/10.3390/cancers14225548

Chicago/Turabian Style

Dimitraki, Maria Georgia, and George Sourvinos. 2022. "Merkel Cell Polyomavirus (MCPyV) and Cancers: Emergency Bell or False Alarm?" Cancers 14, no. 22: 5548. https://doi.org/10.3390/cancers14225548

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop