G Model ACTHIS-50982; No. of Pages 12

ARTICLE IN PRESS Acta Histochemica xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Acta Histochemica journal homepage: www.elsevier.de/acthis

Review

Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors Lucia Salvatorelli a,∗ , Rosalba Parenti b , Giorgia Leone c , Giuseppe Musumeci d , Enrico Vasquez a , Gaetano Magro a a Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, Azienda Ospedaliero-Universitaria “Policlinico-Vittorio Emanuele”, Anatomic Pathology Section, University of Catania, Catania, Italy b Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania, Italy c Anatomic Pathology, Hospital of Sondrio, Sondrio, Italy d Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy

a r t i c l e

i n f o

Article history: Received 18 December 2014 Received in revised form 15 March 2015 Accepted 20 March 2015 Available online xxx Keywords: WT1 Tumors Children Adolescents Review

a b s t r a c t Despite Wilms tumor 1 (WT1) protein was originally considered as a specific immunomarker of Wilms tumor, with the increasing use of immunohistochemistry, there is evidence that other tumors may share WT1 protein expression. This review focuses on the immunohistochemical profile of WT1 protein in the most common malignant tumors of children and adolescents. The variable expression and distribution patterns (nuclear vs cytoplasmic) in the different tumors, dependent on the antibodies used (anti-C or N-terminus WT1 protein), will be emphasized by providing explicative illustrations. Potential diagnostic pitfalls from unexpected WT1 protein expression in some tumors will be discussed in order to avoid diagnostic errors, especially when dealing with small biopsies. © 2015 Elsevier GmbH. All rights reserved.

Introduction The Wilms tumor 1 (WT1) gene on chromosome 11p13, encodes a zinc-finger transcription factor with developmental, tumor suppressor and oncogenic properties (Call et al., 1990; Gessler et al., 1990; Hohenstein and Hastie, 2006; Lee and Haber, 2001; Parenti et al., 2014b). These different, occasionally opposite, functions may be explained if we consider that various isoforms of WT1 do exist, as a result of alternative splicing (Menke et al., 1998; Lee and Haber, 2001; Hohenstein and Hastie, 2006; Roberts, 2006; Hartkamp and Roberts, 2008; Huff, 2011). Several studies have shown that WT1 plays an important role during human development, being expressed in several tissues, both at nuclear and cytoplasmic level. In this regard, nuclear immunohistochemical expression is frequently found in metanephric and mesonephric glomeruli, primary sex cords, gonadic stroma, mesothelial and submesothelial cells, while cytoplasmic expression is usually seen in developing skeletal muscles, myocardium, radial glia of spinal cord and cerebral cortex, sympathetic neuroblasts, adrenal cortical cells and endothelial cells of blood vessels (Pritchard-Jones et al., 1990; Sharma et al., 1992; Armstrong et al., 1993; Mundlos et al., 1993;

∗ Corresponding author. E-mail address: [email protected] (L. Salvatorelli).

Ramani and Cowell, 1996; Charles et al., 1997; Parenti et al., 2013, 2014a, 2015). With regard to neoplastic tissues, WT1 protein has been found in several tumors with variable immunostaining patterns, i.e., exclusively nuclear, cytoplasmic or both, according to the antibodies used (anti-C or N-terminus WT1 protein) (Carpentieri et al., 2002; Nakatsuka et al., 2006; Schittenhelm et al., 2010; Bisceglia et al., 2011a,b; Salvatorelli et al., 2011; Singh et al., 2012; Magro et al., 2014a,b, 2015c,d). The different sub-cellular distribution of WT1 protein is consistent with its nucleo-cytoplasmic shuttling properties (Niksic et al., 2004), suggesting its involvement as regulator of transcription or post-transcriptional processes, depending on the tumor tissue and environmental contex (Hohenstein and Hastie, 2006). Interestingly, in some tumors, such as Wilms tumor, ovarian, mesothelial neoplasms, Sertoli cell tumor, and rhabdomyosarcoma, the variable nuclear and cytoplasmic WT1 staining may be explained by assuming that the expression of this transcription factor in some neoplastic tissues mirrors its normal developmental regulation (Parenti et al., 2013, 2014a; Magro et al., 2015c). Recently, we have also shown that WT1 shows an oncofetal expression pattern, being abundantly detected in developing and neoplastic skeletal muscle tissues, while its expression is downregulated in adult normal skeletal muscle tissues (Salvatorelli et al., 2011; Magro et al., 2015c). However, over the last years there is increasing evidence showing that WT1 protein is expressed “de

http://dx.doi.org/10.1016/j.acthis.2015.03.010 0065-1281/© 2015 Elsevier GmbH. All rights reserved.

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12 2

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

novo” in some tumors (colorectal, breast cancer, and brain tumors) which arise from tissues that normally do not, suggesting a potential oncogenic role in these neoplasms (Loeb et al., 2001; Oji et al., 2002, 2004a,b; Koesters et al., 2004; Hohenstein and Hastie, 2006; Kang et al., 2010; Oue et al., 2011). The diagnosis of benign and malignant pediatric tumors is often challenging, mainly due to the fact that some of these tumors share overlapping morphological and/or immunohistochemical features, with the possibility of misdiagnosing a benign lesion with a malignant tumor or vice versa (Magro et al., 2005, 2008, 2011; Alaggio et al., 2009). In addition a subset of malignant tumors may present unusual clinical, morphological and immunohistochemical features which make difficult their recognition, especially when dealing with small biopsies. Therefore there is an increasing need of highly sensitive and specific immunomarkers which can be exploitable in daily practice in order to achieve a correct diagnosis (Magro et al., 2015a,b). Among these immunomarkers, WT1 protein, traditionally considered as specific for Wilms tumor, is expressed in a wide variety of other neoplasms of children and adolescents, even if with a variable cellular distribution (nuclear vs cytoplasmic expression). The aim of this review is to focus on the immunohistochemical expression of WT1 protein in the most common tumors of children and adolescents. The potential diagnostic role of WT1 protein will be emphasized, providing illustrations and discussion about its usefulness in the differential diagnosis. Potential diagnostic pitfalls, which can arise from unexpected WT1 protein immunoreactivity, will be also discussed. WT1 antibodies: Nuclear versus cytoplasmic immunoreactivity For a long time, it was believed that immunohistochemical expression of WT1 protein was exclusively limited to the nucleus and that the cytoplasmic localization was an artefact, likely due

to cross-reactivity of the antibodies used. Nuclear staining has been obtained especially with antibodies directed against the Cterminal portion of WT1 protein (clone WT 1C19) (Ramani and Cowell, 1996; Charles et al., 1997). However with the advent of new available antibodies against the N-terminal portion of WT1 protein (clone WT 6F-H2), more recent studies have shown that cytoplasmic WT1 staining can be obtained in the nucleus or cytoplasm, or concurrently in both nucleus and cytoplasm (Carpentieri et al., 2002; Bisceglia et al., 2011a; Parenti et al., 2013, 2014a; Magro et al., 2014a,b, 2015c,d). This variable immunoreactivity may be explained assuming that WT1 acts as regulator of either transcriptional or translational processes, shuttling between the nucleus and the cytoplasm (Niksic et al., 2004; Hohenstein and Hastie, 2006). WT1 protein expression in tumors of children and adolescents Wilms tumor (nephroblastoma) It is a malignant pediatric embryonal neoplasm that occurs as a result of a disturbance of cellular differentiation of the metanephric blastema (Davies et al., 2004; Chau and Hastie, 2012; Miller-Hodges and Hohenstein, 2012; Al-Hussain et al., 2014). Accordingly, this neoplasm replicates, at least partially, the morphology of the developing metanephros (Fig. 1A and B). Wilms tumor is the most common genitourinary tumor of childhood, with most cases diagnosed in children between 2 and 4 years of age. Both kidneys are equally affected, and there is the possibility of synchronous or metachronous bilateral involvement in 5–10% of cases. Classically, Wilms tumors presents as an abdominal mass, with abdominal pain, hematuria and hypertension. Histologically, the majority of Wilms tumors usually exhibits triphasic histological components, consisting of blastematous, epithelial, and stromal components (Al-Hussain et al., 2014) (Fig. 1C). The blastematous component

Fig. 1. (A and B) Metanephros of human fetus of 11 weeks of gestational age. (A) Histological illustration showing developing metanephros. The sub-capsular nephrogenic zone, containing blastema and developing epithelial structures, is seen (hematoxylin and eosin, original magnification ×80); (B) Nuclear WT1 expression is found in the blastema and podocytes of developing glomeruli (immunoperoxidase method with WT1 C-terminus antibodies, original magnification ×80). (C and D) Wilms tumor. (C) Histological examination showing the triphasic components: blastema (B), epithelial structures and stromal component (S) (hematoxylin and eosin, original magnification ×100); (D) WT1 is expressed in the nuclei of the blastematous component, in the cytoplasm of stromal cells, while it is lacking in the epithelial structures (immunoperoxidase method with WT1 N-terminus antibodies, original magnification ×100).

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

3

Fig. 2. Intra-abdominal desmoplastic small round cell tumor of a 11 year-old female. (A) Tumor nests surrounded by fibro-sclerotic stroma are typical features (hematoxylin and eosin, original magnification×80); neoplastic cells showing cytoplasmic desmin (B) and cytokeratin (C) expression (B and C: immunoperoxidase method, original magnification×150); (D) Nuclear WT1 staining is obtained with WT1 C-terminus antibodies (immunoperoxidase method, original magnification ×200).

consists of densely packed small round blue cells with scanty cytoplasm, dark nuclei and frequent mitotic figures. They are usually arranged in several growth patterns, including diffuse, nodular and cord-like patterns. Notably, some cases of Wilms tumors are composed almost exclusively of the above mentioned blastematous component and, thus, may result diagnostically challenging, especially when dealing with small biopsies (differential diagnosis with other small round blue cell tumors). The epithelial component is often composed of tubules, papillary and glomerular-like structures which are closely reminiscent of normal nephrogenesis. Primitive rosette-like structures, representing early tubular forms and cysts lined by primitive columnar or cuboidal cells can be usually seen. The stromal component exhibits a variety of patterns of differentiation. Most tumors are composed of spindle-shaped cells set in a myxoid stroma, vaguely resembling embryonal mesenchyme. Heterologous stromal elements, such as skeletal muscle, cartilage, bone, fat, or more rarely neuroglia and mature ganglion cells can be seen. Skeletal muscle in various stages of differentiation, including rhabdomyoblasts, represents the most common divergent line of differentiation. Identification of anaplastic cells (anaplasia), i.e. large neoplastic cells with nuclear pleomorphism and multipolar mitotic figures, is important because it may have prognostic implications (favorable vs unfavorable histology), according to its tumor extension (focal vs diffuse). Anaplasia is usually seen in about 5% of Wilms tumors and seems to be associated with a poor prognosis and increased risk of treatment failure, especially when it is diffusely distributed. The immunohistochemical profile depends on the different tumor components examined. Blastematous component shows diffuse expression of vimentin and WT1, while a variable staining is found with CD56, CD57, cytokeratins, EMA, desmin and PAX2 (Ramani and Cowell, 1996; Charles et al., 1997; Nakatsuka et al., 2006; Vasei et al., 2008; Davis et al., 2011; Arnold et al., 2014; Sehic et al., 2014). The epithelial elements stain for cytokeratin,

EMA, CD56 and variably for PAX2 and WT1 (Ramani and Cowell, 1996; Charles et al., 1997; Vasei et al., 2008; Davis et al., 2011). The cells of the stromal component are usually vimentin positive, while the heterologous skeletal muscle component is reactive for desmin, myogenin and MyoD1. Among the above mentioned markers, WT1 is certainly the most sensitive and specific marker for the diagnosis of Wilms tumor, being dected in more than 90% of cases (Ramani and Cowell, 1996; Charles et al., 1997; Nakatsuka et al., 2006). WT1 protein is expressed mainly at nuclear level, using antibodies directed both to C-terminal or N-terminal portions of the WT1 protein (Ramani and Cowell, 1996; Charles et al., 1997; Nakatsuka et al., 2006), with an immunoreactivity which can be detected in blastematous, and less frequently, in epithelial and stromal cells. This immunoreactivity mirrors the developmental expression of WT1 protein during human nephrogenesis (Fig. 1B). A concurrent or exclusively (more often in blastematous and stromal cells) cytoplasmic WT1 expression can be found, especially by using WT1 N-terminus antibodies (Nakatsuka et al., 2006) (Fig. 1D). Similarly for desmoplastic small round cell tumor, only nuclear staining should be considered as specific for diagnosis of Wilms tumor. Desmoplastic small round cell tumor This highly aggressive tumor, originally described as multiple intra-abdominal tumor masses, is now recognized to arise from many other sites, including pleura, kidney, ovary, scrotum, meninges, bone, scalp, paranasal sinuses, pancreas, and parotid gland (Antonescu and Ladanyi, 2013). Because of its predominant location in mesothelium-lined cavities and its immunohistochemical profile, a primitive origin from mesothelial/sub-mesothelial cells was originally postulated, but later denied. This tumor usually occurs in children and adolescents, with male predilection. Abdominal pain and multiple abdominal masses, without a primary organ involved, are the usual presenting findings (Antonescu and

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12 4

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

Fig. 3. Renal rhabdoid tumor of 1 year-old female (A) histological examination showing the typical rhabdoid cells with abundant eosinophilic cytoplam (hematoxylin and eosin, original magnification ×80); neoplastic cells are positive for cytokeratin (B), while they typically lack nuclear INI-1 expression (C); intratumoral lymphocytes are stained and served as internal control (immunoperoxidase method, original magnification ×150); (D) Unusual nuclear WT1 staining is obtained with WT1 N-terminus antibodies; endothelial cells of intra-tumoral vessels show cytoplasmic staining, and served as internal control (immunoperoxidase method, original magnification ×200).

Ladanyi, 2013). The most common sites of origin are the mesentery, omentum, surface of the liver, and pelvic peritoneum (Antonescu and Ladanyi, 2013). Histologically, it is composed of variably-sized nests, trabeculae, or lobules of malignant small cells, usually separated by a prominent fibro-sclerotic stroma (Fig. 2A). Neoplastic cells are round in shape, with scant cytoplasm, indistinct cell borders and hyperchromatic round to oval, or slightly angulated, nuclei that have finely granular chromatin and small nucleoli. Central necrosis and calcification may be seen within the nests (Fig. 2A). Mitoses are usually observed. In some cases, unusual morphological features, such as rhabdoid or signet ring appearance, as well as glandular or pseudorosettes formation have been described (Antonescu and Ladanyi, 2013). Immunohistochemically, desmoplastic small round cell tumor is characterized by a polyphenotypic profile, with co-expression of vimentin, desmin (Fig. 2B), epithelial markers (cytokeratin; epithelial membrane antigen) (Fig. 2C), and WT1 (Fig. 2D) (Antonescu and Ladanyi, 2013). Although other markers, such as CD99, smooth muscle actin, neuron-specific enolase, CD57 and synaptophysin are variably found, the co-expression of epithelial markers, desmin and WT1 have a major diagnostic value. With regard to WT1, it should be emphasized that most cases (>90%) of desmoplastic small round cell tumor show strong nuclear staining with antibodies directed against the C-terminal portion of WT1 protein (clone WT1 C19) (Barnoud et al., 2000; Hill et al., 2000). This unexpected WT1 positivity was originally considered as an evidence of the possible origin of the tumor from mesothelial cells, which are WT1-positive since fetal development (Parenti et al., 2013, 2015). However, the lack of staining for other mesothelial markers, such as cytokeratins 5/6 and calretinin, argues against this hypothesis. It is widely accepted that the aberrant nuclear expression of WT1 protein in this tumor is due to a recurrent chromosomal translocation t(11;22)(p13;q12),

which can be found in about 90% of cases (Antonescu and Ladanyi, 2013). Two genes, EWSR1 gene and WT-1 gene, are fusion partners, resulting in EWSR1-WT1 fusion transcript (Antonescu and Ladanyi, 2013). Some authors state that only nuclear staining obtained with the WT1 C-terminus antibodies (clone WT C19) is of diagnostic utility in diagnosis of desmoplastic small round cell tumor, because they are predictive of the EWS-WT1 translocation with high sensitivity and specificity (Hill et al., 2000; Carpentieri et al., 2002). Infact nuclear staining is found with the former antibodies in >95% of cases, in contrast to a minority of cases which results to be positive also with WT1 N-terminus antibodies (Carpentieri et al., 2002; Nakatsuka et al., 2006; Murphy et al., 2008). It has been suggested that the unusual nuclear WT1 immunoreactivity with N-terminus antibodies is likely due to novel fusion transcripts (Murphy et al., 2008). Interestingly, apart from nuclear staining, a weak to moderate cytoplasmic WT1 positivity has been reported in some cases of desmoplastic small round cell tumor, especially by using N-terminus antibodies (Nakatsuka et al., 2006; Wang et al., 2007; Bisceglia et al., 2011a). Based on these findings, we suggest the use of WT1 C-terminus (clone WT C19) antibodies for the diagnosis of desmoplastic small round cell tumor, and that only a strong nuclear staining should be considered as a positive result. Malignant rhabdoid tumor Malignant rhabdoid tumor is a highly aggressive neoplasm that usually occurs in the kidney of children, but less frequently may arise from other sites, including central nervous system, somatic soft tissues, abdomen, pelvis, retroperitoneum, liver, heart, and gastrointestinal tract (Alaggio et al., 2012). Interestingly, renal and nervous system tumors occur frequently in children younger than

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

5

Fig. 4. (A and B) Embryonal rhabdomyosarcoma, botryoid type, of the external auditory canal of a 5 year-old boy. (A) Histological examination showing a proliferation of small round to spindle cells set in a myxoid stroma, beneath the squamous epithelium (hematoxylin and eosin, original magnification ×60). (B) Neoplastic cells showing strong and diffuse cytoplasmic WT1 staining (immunoperoxidase method with WT1 N-terminus antibodies, original magnification ×60). (C and D) Spindle cell/sclerosing rhabdomyosarcoma of the oral cavity. (C) Histological examination showing a proliferation of spindle cells set in fibrous stroma (hematoxylin and eosin, original magnification ×60). (D) Neoplastic cells showing strong and diffuse cytoplasmic WT1 staining (immunoperoxidase method with WT1 N-terminus antibodies, original magnification ×60).

10 years, while soft tissue tumors are usually diagnosed in fetuses, newborns and young children (Alaggio et al., 2012). A variable number of cases (15–85%), including congenital cases, are metastastic at presentation, and pursue a fatal course (Alaggio et al., 2012). Histologically, it is composed of round/epithelioid to polygonal cells, variably arranged in solid or trabecular growth patterns. Characteristically, neoplastic cells have abundant, deeply eosinophilic cytoplasm with a paranuclear eosinophilic, PAS-positive inclusion and large, round, vescicular nuclei with finely dispersed chromatin containing a prominent eosinophilic nucleolus (Fig. 3A). Mitoses and necrosis are commonly seen. A minor tumor component is represented by smaller, round, undifferentiated cells with scant cytoplasmic rim. In some cases, this unusual cellular component may be prominent, posing serious differential diagnostic problems with small round cell tumors, especially Ewing’s sarcoma/peripheral primitive neuroectodermal tumors (Sigauke et al., 2006; Alaggio et al., 2009; Machado et al., 2010). Immunohistochemically, malignant rhabdoid tumor, like desmoplastic small round cell tumor, exhibits a poliphenotypic profile, with variable co-expression of different markers, including vimentin, cytokeratin (Fig. 3B), epithelial membrane antigen, and CD99 (Alaggio et al., 2012). However the most useful diagnostic marker is the complete absence of nuclear immunoreactivity for INI1 protein (Hoot et al., 2004; Sigauke et al., 2006; Judkins, 2007; Machado et al., 2010) (Fig. 3C). Additional markers, such as muscle specific actin, alpha-smooth muscle actin, S100 protein, synapthophisin, CD56 and also WT1 can be occasionally expressed (Ramani and Cowell, 1996; Charles et al., 1997; Alaggio et al., 2012). As far as WT1 protein expression is concerned, immunostaining, exclusively nuclear or nucleo-cytoplasmic, can be detected using antibodies directed against the C-terminal portion of WT1 protein (clone WT C19) also in some cases of malignant rhabdoid tumor (Ramani and Cowell, 1996; Charles et al., 1997). We have experience of similar results also by using antibodies against the N-terminal portion (clone WT 6F-H2) (Fig. 3D).

Rhabdomyosarcoma Rhabdomyosarcoma (RMS) is a malignant tumor composed of cells which show variable morphological, immunohistochemical and ultrastructural evidence of skeletal muscle differentiation. Based on morphological, immunohistochemical and molecular features, at least four major subtypes can be recognized: (i) embryonal; (ii) alveolar; (iii) spindle cell/sclerosing; (iv) pleomorphic. While pleomorphic rhabdomyosarcoma is typically a tumor of adults, the other subtypes occur predominantly in children and adolescents. Embryonal rhabdomyosarcoma, the most common subtype, occurring in children aged 50% of neoplastic cells) of spindle-shaped cells arranged in whorls or in a fascicular/

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12 6

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

Fig. 5. (A) Poorly differentiated neuroblastoma: neoplastic cells lacking WT1 staining; intra-tumoral endothelial cells of blood vessels show cytoplasmic staining, and served as internal control (immunoperoxidase method with WT1 N-terminus antibodies, original magnification ×80); (B) Intermixed ganglioneuroblastoma: immature ganglion cells and Schwann cells of the stromal component, show cytoplasmic WT1 staining (immunoperoxidase method with WT1 N-terminus antibodies, original magnification ×100).

storiform growth pattern closely reminiscent of fibrosarcoma or leiomyosarcoma (Parham et al., 2012) (Fig. 4C). A variable amount of extracellular sclerosis is seen and, when extensive, the term “sclerosing rhabdomyosarcoma” can be used. Alveolar rhabdomyosarcoma is a distinct subtype of rhabdomyosarcoma usually associated with aggressive behavior. It affects adolescents and young adults, with a peak incidence at 10–25 years of age. The most common sites of occurrence are the deep soft tissues of the extremities and axial musculature. Histologically, it is characterized by cellular nests separated by fibro-vascular septa. Neoplastic cells, with scant cytoplasm and large hyperchromatic nuclei, are mainly discohesive in the center of the nests, while they are attached to the fibro-vascular septa at the periphery of the nests. This histological growth pattern is designated with the term “alveolar” because it is vaguely reminiscent of lung alveoli. Some cases, which lack the alveolar growth pattern, are classified as “solid variant”, being predominantly composed of sheets of closely packed neoplastic cells with no intervening fibrous septa. Interestingly, some cases of rhabdmyosarcoma may have overlapping or mixed morphology, i.e. areas of embryonal and alveolar subtypes may coexist in the same tumor. Immunohistochemically, virtually all cases of rhabdomyosarcoma, whatever the subtype, are positive, albeit with a variable extension, to desmin, myogenin and MyoD1, currently considered the most reliable markers of this tumor (Parham et al., 2012). Although it is true that immunohistochemistry staining pattern is not reliable in subtyping rhabdomyosarcoma, there is evidence that alveolar rhabdomyosarcoma usually exhibits a more diffuse staining for both desmin and myogenin compared with embryonal rhabdomyosarcoma. Among these markers, myogenin and MyoD1 are considered highly specific markers of skeletal muscle differentiation, as desmin can be expressed by several myofibroblastic/leiomyomatous lesions (Parham et al., 2012). Interestingly, both embryonal and alveolar rhabdomyosarcoma may also variably express CD99, cytokeratin, S100 protein, alpha-smooth muscle actin, and neuroendocrine markers, such as chromogranin A and synaptophysin (Parham et al., 2012). In addition some cases of rhabdomyosarcoma have been reported to be WT1-positive. In this regard it should be emphasized that nuclear WT1 staining

can be focally demonstrated only in a few cases, by using WT1 Cterminus antibodies (Barnoud et al., 2000). Conversely, with the advent of new available WT1 N-terminus antibodies (clone WT 6F-H2), several studies have reported a diffuse and strong cytoplasmic expression in embryonal (Fig. 4B), sclerosing/spindle cell (Fig. 4D) and alveolar subtypes of rhabdomyosarcoma (Carpentieri et al., 2002; Sebire et al., 2005; Bisceglia et al., 2011a,b; Magro et al., 2015c). This unexpected finding is explained by the detection of WT1 in the cytoplasm of human developing myoblasts and myotubes during the early phases of skeletal myogenesis (Salvatorelli et al., 2011; Parenti et al., 2013, 2015; Magro et al., 2015c,d; Musumeci et al., 2015), suggesting its potential role in the molecular mechanisms that regulate skeletal muscle differentiation. Although we admit that the diagnosis of rhabdomyosarcoma is based on the demonstration of myogenic markers (desmin, myogenin, MyoD1), the strong and diffuse cytoplasmic WT1 staining can be useful in confirming the diagnosis when dealing with a small round cell tumor morphologically consistent with a rhabdomyosarcoma (i.e., identification, even if only focally, of cells with the features of rhabdomyoblasts) but which lacks myogenin and MyoD1 (Parham et al., 2012). In our experience, a strong and diffuse cytoplasmic WT1 expression (WT1 N-terminus-clone WT 6F-H2) can be exploitable as an adjunct helpful immunomarker of rhabdomyosarcoma versus other small round cell tumors (Magro et al., 2015c,d). Neuroblastic tumors Neuroblastic tumors occur in children and adolescents, especially from the adrenal gland, retroperitoneum or, more rarely, in the posterior mediastinum. They represent a heterogeneous group of lesions characterized by a wide morphological spectrum which reflects a different degree of differentiation from immature neuroblastic cells to mature ganglionic cells (Rosai, 2011). According to the degree of maturation, the following histological categories are defined: (i) neuroblastoma (Schwannian stroma-poor tumors), including undifferentiated, poorly differentiated and differentiating neuroblastomas; (ii) ganglioneuroblastoma (Schwannian stroma-rich tumors), including intermixed and nodular

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

7

Fig. 6. Infantile fibrosarcoma of the leg of a 4 month-old male infant. (A) Histological examination showing spindle cells arranged in intersecting fascicles (hematoxylin and eosin, original magnification ×80); (B) neoplastic cells showing strong and diffuse cytoplasmic WT1 staining (immunoperoxidase method with WT1 N-terminus antibodies, original magnification ×100).

ganglioneuroblastomas; (iii) ganglioneuroma (Schwannian stroma predominant tumors), including maturing and mature ganglioneuromas (Rosai, 2011). Neuroblastoma consists of small, round cells, with scant cytoplasm and round, hyperchromatic nuclei. The tumor cells sometimes are arranged around a fibrillar area (neuropil), resulting in the so-called Homer Wright rosettes. If ≤5% or more than 5% of the tumor cells shows ganglionic differentiation, the neuroblastoma is classified, respectively, as poorly differentiated or differentiating neuroblastoma; in contrast when neoplastic cell do not show any differentiation toward ganglionic cells and the neuropil is almost or completely absent, the neuroblastoma is classified as undifferentiated neuroblastoma (Rosai, 2011). Ganglioneuroblastoma exhibits features of differentiation that are intermediate between neuroblastoma and ganglioneuroma. Histologically, two distinct entities can be recognized, intermixed and nodular. In both variants there is a ganglioneuromatous component, but in the former collection of immature ganglion cells are interspersed among the Schwannian stroma, while in the latter, in addition to the ganglioneuromatous component, there is an area of neuroblastoma (Rosai, 2011). Ganglioneuroma rapresents the fully differentiated member of neuroblastic tumors, being composed of mature or maturing ganglion cells surrounded by fascicles of Schwann cells (Rosai, 2011). Several morphological, immunohistochemical and in vitro studies support the concept that childhood peripheral neuroblastic tumors recapitulate the developmental stages of normal peripheral sympathetic nervous system (Magro et al., 1995, 2000; Magro and Grasso, 1997, 2001; De Preter et al., 2007; Hoehner et al., 1998). Neuroblastoma is traditionally considered as a WT1-negative tumor, with only a few studies reporting focal and weak nuclear WT1 staining in neuroblastoma (Barnoud et al., 2000; Sebire et al., 2005; Carpentieri et al., 2002). A more recent study showed WT1 expression preferentially in ganglioneuroblastoma

and ganglioneuroma compared with neuroblastoma (Wang et al., 2007). In our experience, by using antibodies against the Nterminal portion of WT1 protein (clone WT 6F-H2), the more undifferentiated neuroblastic component in both neuroblastoma and ganglioneuroblastoma is usually WT1-negative (Fig. 5A), while a variable cytoplasmic WT1 staining is found in ganglion cell component of both ganglioneuroblastoma and ganglioneuroma (Fig. 5B). Congenital/infantile fibrosarcoma It is a malignant tumor currently classified in the category of the intermediate (rarely metastasizing) neoplasms, which typically occurs in the first 2 years of life, with a significant number of cases diagnosed at birth or antenatally (Coffin and Alaggio, 2012). The most commonly involved sites are soft tissues of the trunk and distal extremities, with only rare cases reported in the retroperitoneum. Prognosis is favorable, with distant metastases documented in only a few cases (Coffin and Alaggio, 2012). Histologically, it is a highly cellular neoplasm composed predominantly of spindle cells variably arranged in fascicles, focally exhibiting a herringbone pattern (Fig. 6A). Areas with more immature round to polygonal cells can be seen. Generally, neoplastic cells show only a mild to focally moderate degree of nuclear atypia. Mitoses are usually numerous, even if atypical mitoses are absent. A hemangiopericytoma-like vascular pattern and hypocellular fibrotic areas can be seen (Coffin and Alaggio, 2012). Although congenital/infantile fibrosarcoma may variably express, more frequently with focal extension, alpha-smooth muscle actin and/or desmin, highly specific markers are not available for this tumor. Accordingly, the diagnosis is frequently of exclusion, being mainly based on negative results for specific lineage markers such as desmin, myogenin, CD34, S-100, HMB-45, and

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12 8

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

Fig. 7. (A and B) Blastemal-dominant Wilms tumor. (A) A close resemblance with desmoplastic small round cell tumor is evident (hematoxylin and eosin, original magnification ×60). (B) Blastematous cells showing strong and diffuse nuclear WT1 staining (immunoperoxidase method with WT1 C-terminus antibodies, original magnification ×100). (C and D) Desmoplastic small round cell tumor. (C) Typical histological picture of desmoplastic small round cell tumor (hematoxylin and eosin, original magnification ×60). (D) Neoplastic cells lacking WT1 expression (immunoperoxidase method with WT1 N-terminus antibodies, original magnification ×100).

cytokeratins (Coffin and Alaggio, 2012). Given the non-specificity of immunohistochemistry, the diagnosis of congenital/infantile fibrosarcoma is usually confirmed, especially for morphologically and/or immunohistochemically ambiguous cases, by the identification of the recurrent translocation t(12;15) (p13;q25) with an ETV6-NTRK3 gene fusion (Bourgeois et al., 2000; Sheng et al., 2001). Recently, we first showed that congenital/infantile fibrosarcoma is a WT1-positive tumor by using antibodies against the N-terminal portion of WT1 protein (clone WT 6F-H2) (Fig. 6B) (Magro et al., 2014a). In this regard a strong and diffuse cytoplasmic WT1 staining is obtained not only in surgically resected samples but also in small biopsies, suggesting that WT1 can be successfully used in confirming the diagnosis of congenital/infantile fibrosarcoma (Magro et al., 2014a). In this regard, WT1 is useful in daily practice, especially in distinguishing congenital/infantile fibrosarcoma from desmoid-type fibromatosis, a lesion which, albeit may show overlapping morphological and immunohistochemical features with the former, is typically WT1-negative (Magro et al., 2014a). WT1 immunoreactivity: Potential diagnostic pitfalls Although WT1 is a useful marker in the diagnosis of pediatric tumors, an erroneous interpretation of its staining in some clinicopathologic settings may lead pathologists to a misdiagnosis. It is commonly believed that a pediatric small round blue cell tumor with nuclear immunostaining for WT1 protein is a Wilms tumor. However, even if WT1 protein is a highly sensitive immunomarker of Wilms tumor, it is not specific, being expressed in other morphologic mimics. Wilms’ tumor versus desmoplastic small round cell tumor A pediatric renal tumor which expresses WT1 protein in the nuclei of neoplastic cells is considered a Wilms’ tumor. This is

true if an epithelial component can be demonstrated, at least focally, in association with a blastematous and/or stromal component. However differential diagnostic problems may arise when dealing with a blastemal-dominant Wilms tumor, in which the epithelial component is lacking or only focally detectable (Fig. 7A). In this regard, differential diagnostic problems mainly revolves around desmoplastic small round cell tumor which, albeit commonly observed in the mesentery, omentum, surface of the liver, and pelvic peritoneum, may primarily arise in kidney or retroperitoneum of children and adolescents (Wang et al., 2007; da Silva et al., 2009) (Fig. 7C). Moreover there is the possibility that Wilms’ tumor be so large in size to be diagnosed, by radiological imaging, as abdominal or retroperitoneal mass which involves secondarily the kidney. Accordingly, distinguishing a blastemal-dominant Wilms tumor from a desmoplastic small round cell tumor may be challenging (Wang et al., 2007; da Silva et al., 2009; Arnold et al., 2014), especially if pathologist is dealing with a small, round- to ovoid-shaped cell component as the sole cytotype in small biopsies obtained from renal masses. The possibility of misdiagnosing a desmoplastic small round cell tumor with a Wilms tumor is mainly due to the fact both tumors share nuclear WT1 expression by using WT1 (C-terminus; clone WT C19) antibodies. In addition, the confusion may be also enhanced by the evidence that desmin, a marker expressed in >90% of cases of desmoplastic small round cell tumors, is occasionally found in the blastemal component of the Wilms’ tumor, especially with a dot-like, perinuclear staining pattern (Arnold et al., 2014). A correct diagnostic interpretation relies on the differential expression of WT1 protein in the two tumors by using different WT1-antibodes. In this regard, pathologists should keep in mind that the majority (>95%) of desmoplastic small round cell tumors results to be WT1-positive (nuclear staining) only if antibodies against the C-terminal portion (clone WT C19) are used (Fig. 2D) (Hill et al., 2000; Carpentieri et al., 2002; Murphy et al., 2008). In contrast nuclear staining for WT1 is still maintained in

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

9

Fig. 8. Renal rhabdoid tumor of a 1 year-old female. (A–C) Core biopsy. (A) Histological examination showing a proliferation of small, round cells without any feature consistent with rhabdoid tumor. (B) At higher magnification cellular details of neoplastic cells can be better appreciated. (C) Neoplastic cells showing a strong and diffuse nuclear staining for WT1 (A: hematoxylin and eosin, original magnification ×80; B: hematoxylin and eosin, original magnification ×150; C: immunoperoxidase method with WT1 N-terminus antibodies; original magnification ×100). (D) Histological examination of the surgically resected sample, showing that tumor was composed predominantly of the typical rhabdoid cells with abundant eosinophilic cytoplasm, which were lacking in the core biopsy (hematoxylin and eosin, original magnification ×200).

Wilms’ tumor (Fig. 7B), whereas it is lacking in the majority (>97%) of cases of desmoplastic small round cell tumor by using WT1 Nterminus antibodies (clone WT 6F-H2) (Fig. 7D). Based on these considerations, we strongly suggest that when pathologists are dealing with a small round blue cell tumor of the kidney or abdominal/retroperitoneal cavity, WT1 C- and N-terminus antibodies should be included in the immunohistochemical panel, including desmin, cytokeratins and INI1 protein. The last antibodies (antiINI1 protein) are useful in ruling out malignant rhabdoid tumor. If tumor exhibits diffuse and strong nuclear immunoreactivity with WT1 N-terminus antibodies, it is likely a Wilms’ tumor, even if desmin and cytokeratin immunostaining is variably represented and INI1 expression is retained. A similar diagnostic conclusion should be considered if immunostaining is also obtained with WT1 C-terminus antibodies, along with absence of both desmin and cytokeratins and concurrent expression of INI1 protein. Lastly, if tumor exhibits strong nuclear immunoreactivity only with WT1 C-terminus antibodies, in addition to a diffuse staining for desmin, cytokeratins and INI1, the possibility of a desmoplastic small round cell tumor should be seriously considered, especially if the morphological features are suggestive. Molecular tests, especially the detection of EWS-WT1 fusion transcript by reverse transcription polymerase chain reaction (RT-PC), are mandatory in confirming the diagnosis of desmoplastic small round cell tumor in those cases with ambiguous morphology and/or immunohistochemical profile. Wilms’ tumor versus malignant rhabdoid tumor Kidney is one of the most common site of malignant rhabdoid tumor in children (Alaggio et al., 2012). Although this tumor is suspected if the typical cells with rhabdoid morphology are present,

it should be emphasized that a small round cell component may variably be represented throughout the tumor (Fowler et al., 2006; Alaggio et al., 2009). It consists of cells with a lymphocyte-like appearance, showing scant cytoplasm and slightly hyperchromatic, round-shaped nuclei with rare and inconspicuous nucleoli (Alaggio et al., 2009). The identification, at least focally, of larger neoplastic cells with nuclei containing finely dispersed chromatin, and one or two small evident nucleoli, may help in the correct interpretation of tumor histotype (Alaggio et al., 2009). However serious differential diagnostic problems between a blastemal-dominant Wilms tumor and a malignant rhabdoid tumor do exist when dealing with small biopsies from renal tumor masses which contain exclusively/predominantly a small, round cell component (Fig. 8A and B). The potential diagnostic pitfall is mainly due to the fact nuclear WT1 staining can be detected using WT1 Cterminus antibodies in some cases of malignant rhabdoid tumor (Ramani and Cowell, 1996; Charles et al., 1997); in our experience, we have seen a case of renal malignant rhabdoid tumor (Fig. 8D) with a small cell round component at core biopsy, which expressed nuclear staining by using WT1 (N-terminus-clone WT 6F-H2) antibodies (Fig. 8C). As both Wilms’ tumor and rhabdoid tumor may variably stain positively with vimentin and cytokeratins, the most reliable marker helpful in their distinction is INI1 protein. If a renal WT1-positive tumor lacks nuclear immunoreactivity for INI1 protein, the diagnosis of malignant rhabdoid tumor is likely (Hoot et al., 2004; Sigauke et al., 2006; Judkins, 2007; Machado et al., 2010). Based on these considerations, pathologists should be aware that not all WT1-positive renal tumors are Wilms tumors, and when approaching a small biopsy from a renal tumor mass, apart from WT1 (both C- and N-terminus) antibodies, INI protein should be routinely included in the immunohistochemical panel.

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12 10

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

Rhabdomyosarcoma versus other small round cell tumors All subtypes of rhabdomyosarcoma show a diffuse and strong cytoplasmic WT1 staining by using WT1 (N-terminus) antibodies (Fig. 4B and D) (Carpentieri et al., 2002; Sebire et al., 2005; Bisceglia et al., 2011a,b; Magro et al., 2015c,d). A similar staining has also been reported in up 43% of cases with antibodies directed against C-terminus of WT1 protein (Nakatsuka et al., 2006). Although this finding is helpful in confirming diagnosis of rhabdomyosarcoma, especially in tumors with ambiguous immunoprofile, WT1 should not be used alone in daily practice. This is due to the evidence that rhabdomyosarcoma mimics, such as EWS/pPNET and desmoplatic small round cell tumor, may express, even if less frequently and usually with focal extension, WT1 at the cytoplasmic level. With regard to desmoplastic small round cell tumor, a cytoplasmic WT1 staining can be obtained by using WT1 (N-terminus-clone 6F-H2) antibodies (Nakatsuka et al., 2006; Bisceglia et al., 2011a), while a similar cytoplasmic immunoreactivity has been reported in up 43% or 63% of cases of EWS/pPNET by using antibodies against both Cterminus or N-terminus of WT1 protein, respectively (Nakatsuka et al., 2006). This strongly suggests that a small round cell tumor which expresses a diffuse and strong cytoplasmic WT1 staining is likely to be a rhabdomyosarcoma, but this finding should be interpreted in the context of other antibodies, such CD99, FLI-1, desmin, myogenin, MyoD1, cytokeratins, and INI1 protein. Congential/infantile fibrosarcoma versus other spindle cell tumors There are no specific immunomarkers for congenital/infantile fibrosarcoma, even if this tumor may show focally alpha-smooth muscle actin and/or desmin, suggesting that, at least, a minor component of neoplastic cells is myofibroblastic in nature. Recently, we have shown that congenital/infantile fibrosarcoma exhibits a strong and diffuse cytoplasmic WT1 immunoreactivity by using WT1 N-terminus antibodies (Fig. 7B) (Magro et al., 2014a). Although sensitive, this staining is not specific because other spindle cell lesions, such spindle cell rhabdomyosarcoma, benign and malignant peripheral nerve sheath tumors, leyomyosarcoma, myofibroma/myofibromatosis, are reported to be variably WT1positive (Nakatsuka et al., 2006; Schittenhelm et al., 2010; Singh et al., 2012; Magro et al., 2014a, 2015c). This possibility should be kept in mind by pathologists, especially when dealing with small biopsies containing a spindle cell proliferation. However we suggest that WT1 staining is helpful in confirming the diagnosis of congential/infantile fibrosarcoma in the appropriate clinicopathologic context (tumor localized in the extremities of an infant, highly cellular and composed of interlacing fascicles of uniform spindle cells with only low nuclear atypia, and focal expression of alpha-smooth muscle actin and/or desmin) (Magro et al., 2014a). The morphologically and immunohistochemically ambiguous cases can be correctly diagnosed by means of molecular analyses showing the recurrent translocation t(12;15) (p13;q25) with an ETV6-NTRK3 gene fusion, typically found in most cases of infantile fibrosarcoma (Coffin and Alaggio, 2012). Conclusions WT1 protein is a useful marker for diagnosis of malignant tumors in children and adolescents. However, it should be emphasized that not only Wilms tumor, but also other neoplasms, including desmoplastic small round cell tumor, malignant rhabdoid tumor, can express WT1 protein at nuclear level. Pathologists should be aware of the possibility that nuclear and/or cytoplasmic WT1 immunostaining mainly depends on the tumor examined

and WT1 antibodies used (anti-C or N-terminus portion of the protein). This review suggests that WT1 antibodies (both anti-C or N-terminus portions of the protein) should be included in the immunohistochemical panel when approaching a small round blue cell or spindle cell tumor of children and adolescents. Acknowledgement The authors would like to thank Dr. Graziana Spoto for her excellent technical support. References Alaggio R, Boldrini R, di Venosa B, Rosolen A, Bisogno G, Magro G. Pediatric extrarenal rhabdoid tumors with unusual morphology: a diagnostic pitfall for small biopsies. Pathol Res Pract 2009;205:451–7. Alaggio R, Coffin CM, Vargas SO. Soft tissue tumors of uncertain origin. Pediatr Dev Pathol 2012;15:267–305. Al-Hussain T, Ali A, Akhtar M. Wilms tumor: an update. Adv Anat Pathol 2014;21:166–73. Antonescu CR, Ladanyi M. Desmoplastic small round cell tumour. In: Fletcher CDM, Bridge JA, Hogendoorn PCW, Mertens F, editors. WHO classification of tumours of soft tissue and bone. Lyon: IARC; 2013. p. 225–7. Armstrong JF, Pritchard-Jones K, Bickmore WA, Hastie ND, Bard JB. The expression of the Wilms’ tumour gene, WT1, in the developing mammalian embryo. Mech Dev 1993;40:85–97. Arnold MA, Schoenfield L, Limketkai BN, Arnold CA. Diagnostic pitfalls of differentiating desmoplastic small round cell tumor (DSRCT) from Wilms tumor (WT): overlapping morphologic and immunohistochemical features. Am J Surg Pathol 2014;38:1220–6. Barnoud R, Sabourin JC, Pasquier D, Ranchère D, Bailly C, TerrierLacombe MJ, et al. Immunohistochemical expression of WT1 by desmoplastic small round cell tumor: a comparative study with other small round cell tumors. Am J Surg Pathol 2000;24:830. Bisceglia M, Vairo M, Galliani C, Lastilla G, Parafioriti A, De Maglio, et al. Immunohistochemical investigation of WT1 expression in 117 embryonal tumors. Pathologica 2011a;103:182–3. Bisceglia M, Magro G, Carosi I, Cannazza V, Ben Dor D. Primary embryonal rhabdomyosarcoma of the prostate in adults: report of a case and review of the literature. Int J Surg Pathol 2011b;19:831–7. Bourgeois JM, Knezevich SR, Mathers JA, Sorensen PH. Molecular detection of the ETV6-NTRK3 gene fusion differentiates congenital fibrosarcoma from other childhood spindle cell tumors. Am J Surg Pathol 2000;24:937–46. Call KM, Glaser T, Ito CY, Buckler AJ, Pelletier J, Rose EA, et al. Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms’ tumor locus. Cell 1990;60:509–20. Charles AK, Mall S, Watson J, Berry PJ. Expression of the Wilms’ tumour gene WT1 in the developing human and in paediatric renal tumours: an immunohistochemical study. Mol Pathol 1997;50:138–44. Carpentieri DF, Nichols K, Chou PM, Matthews M, Pawel B, Huff D. The expression of WT1 in the differentiation of rhabdomyosarcoma from other pediatric small round blue cell tumors. Mod Pathol 2002;15:1080–6. Chau YY, Hastie ND. The role of Wt1 in regulating mesenchyme in cancer, development, and tissue homeostasis. Trends Genet 2012;28:515–24. Coffin CM, Alaggio R. Fibroblastic and myofibroblastic tumors in children and adolescents. Pediatr Dev Pathol 2012;15:127–80.

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

Davis JL, Matsumura L, Weeks DA, Troxell ML. PAX2 expression in Wilms tumors and other childhood neoplasms. Am J Surg Pathol 2011;35:1186–94. da Silva RC, Medeiros Filho P, Chioato L, Silva TR, Ribeiro SM, Bacchi CE. Desmoplastic small round cell tumor of the kidney mimicking Wilms tumor: a case report and review of the literature. Appl Immunohistochem Mol Morphol 2009;17:557–62. Davies JA, Ladomery M, Hohenstein P, Michael L, Shafe A, Spraggon L, et al. Development of an siRNA-based method for repressing specific genes in renal organ culture and its use to show that the Wt1 tumour suppressor is required for nephron differentiation. Hum Mol Genet 2004;13:235–46. De Preter K, Vandesompele J, Heimann P, Yigit N, Beckman S, Schramm A, et al. Human fetal neuroblast and neuroblastoma transcriptome analysis confirms neuroblast origin and highlights neuroblastoma candidate genes. Genome Biol 2007;8:401. Fowler DJ, Malone M, Chisholm J, Roebuck D, Sebire NJ. Primary thoracic myxoid variant of extrarenal rhabdoid tumor in childhood. Fetal Pediatr Pathol 2006;25:159–68. Gessler M, Poustka A, Cavenee W, Neve RL, Orkin SH, Bruns GA. Homozygous deletion in Wilms tumours of a zinc-finger gene identified by chromosome jumping. Nature 1990;343:774–8. Hartkamp J, Roberts SG. The role of the Wilms’ tumour-suppressor protein WT1 in apoptosis. Biochem Soc Trans 2008;36:629–31. Hill DA, Pfeifer JD, Marley EF, Dehner LP, Humphrey PA, Zhu X, et al. WT1 staining reliably differentiates desmoplastic small round cell tumor from Ewing sarcoma/primitive neuroectodermal tumor. An immunohistochemical and molecular diagnostic study. Am J Clin Pathol 2000;114:345–53. Hoehner JC, Hedborg F, Eriksson L, Sandstedt B, Grimelius L, Olsen L, et al. Developmental gene expression of sympathetic nervous system tumors reflects their histogenesis. Lab Invest 1998;78:29–45. Hohenstein P, Hastie ND. The many facets of the Wilms’ tumour gene, WT1. Hum Mol Genet 2006;15:R196–201 (Spec No). Hoot AC, Russo P, Judkins AR, Perlman EJ, Biegel JA. Immunohistochemical analysis of hSNF5/INI1 distinguishes renal and extra-renal malignant rhabdoid tumors from other pediatric soft tissue tumors. Am J Surg Pathol 2004;28:1485–91. Huff V. Wilms’ tumours: about tumour suppressor genes, an oncogene and a chameleon gene. Nat Rev Cancer 2011;11:111–21. Judkins AR. Immunohistochemistry of INI1 expression: a new tool for old challenges in CNS and soft tissue pathology. Adv Anat Pathol 2007;14:335–9. Kang GH, Kim KM, Noh JH, Sohn TS, Kim S, Park CK, et al. WT1 expression in gastrointestinal stromal tumours. Pathology 2010;42:54–7. Koesters R, Linnebacher M, Coy JF, Germann A, Schwitalle Y, Findeisen P, et al. WT1 is a tumor associated antigen in colon cancer that can be recognized by in vitro stimulated cytotoxic T cells. Int J Cancer 2004;109:385–92. Lee SB, Haber DA. Wilms tumor and the WT1 gene. Exp Cell Res 2001;264:74–99. Loeb DM, Evron E, Patel CB, Sharma PM, Niranjan B, Buluwela L, et al. Wilms’ tumor suppressor gene (WT1) is expressed in primary breast tumors despite tumor-specific promoter methylation. Cancer Res 2001;61:921–5. Machado I, Noguera R, Santonja N, Donat J, Fernandez-Delgado R, Acevedo A, et al. Immunohistochemical study as a tool in differential diagnosis of pediatric malignant rhabdoid tumor. Appl Immunohistochem Mol Morphol 2010;18:150–8. Magro G, Grasso S, Emmanuele C. Immunohistochemical distribution of S-100 protein and type IV collagen in human embryonic and fetal sympathetic neuroblasts. Histochem J 1995;27:694–701.

11

Magro G, Grasso S. Immunohistochemical identification and comparison of glial cell lineage in fetal, neonatal, adult and neoplastic human adrenal medulla. Histochem J 1997;29:293–9. Magro G, Ruggieri M, Fraggetta F, Grasso S, Viale G, Cathepsin D. is a marker of ganglion cell differentiation in the developing and neoplastic human peripheral sympathetic nervous tissues. Virchows Arch 2000;437:406–12. Magro G, Grasso S. The glial cell in the ontogenesis of the human peripheral sympathetic nervous system and in neuroblastoma. Pathologica 2001;93:505–16. Magro G, Di Benedetto A, Sanges G, Scalisi F, Alaggio R. Rhabdomyomatous mesenchymal hamartoma of oral cavity: an unusual location for such a rare lesion. Virchows Arch 2005;446:346–7. Magro G, Greco P, Alaggio R, Gangemi P, Ninfo V. Polypoid angiomyofibroblastoma-like tumor of the oral cavity: a hitherto unreported soft tissue tumor mimicking embryonal rhabdomyosarcoma. Pathol Res Pract 2008;204:837–43. Magro G, Michal M, Alaggio R, D’Amore E. Intradermal proliferative fasciitis in childhood: a potential diagnostic pitfall. J Cutan Pathol 2011;38:59–62. Magro G, Salvatorelli L, Vecchio GM, Musumeci G, Rita A, Parenti R. Cytoplasmic expression of Wilms tumor transcription factor-1 (WT1): a useful immunomarker for young-type fibromatoses and infantile fibrosarcoma. Acta Histochem 2014a;116:1134–40. Magro G, Longo F, Salvatorelli L, Vecchio GM, Parenti R. Wilms’ tumor protein (WT1) in mammary myofibroblastoma: an immunohistochemical study. Acta Histochem 2014b;116:905–10. Magro G, Salvatorelli L, Di Cataldo A, Musumeci G, Spoto G, Parenti R. Cyclin D1 in human neuroblastic tumors recapitulates its developmental expression. An Immunohistochemical study. Acta Histochem 2015a [Epub ahead of print – Special Issue]. Magro G, Brancato F, Alaggio R, Musumeci G, Parenti R, Salvatorelli L. Cyclin D1 is a useful marker for pediatric soft tissue Ewing’s sarcoma/peripheral primitive neuroectodermal tumor: a comparative immunohistochemical study with rhabdomyosarcoma. Acta Histochem 2015b [Epub ahead of print – Special Issue]. Magro G, Salvatorelli L, Puzzo L, Musumeci G, Bisceglia M, Parenti R. Oncofetal expression of Wilms’ tumor 1 (WT1) protein in human fetal, adult and neoplastic skeletal muscle tissues. Acta Histochem 2015c [Epub ahead of print – Special Issue]. Magro G, Longo F, Angelico G, Spadola S, Amore FF, Salvatorelli L. Immunohistochemistry as potential diagnostic pitfall in the most common solid tumors of children and adolescents. Acta Histochem 2015d [Epub ahead of print – Special Issue]. Menke AL, van der Eb AJ, Jochemsen AG. The Wilms’ tumor 1 gene: oncogene or tumor suppressor gene? Int Rev Cytol 1998;181:151–212. Miller-Hodges E, Hohenstein P. WT1 in disease: shifting the epithelial–mesenchymal balance. J Pathol 2012;226:229–40. Mundlos S, Pelletier J, Darveau A, Bachmann M, Winterpacht A, Zabel B. Nuclear localization of the protein encoded by the Wilms’ tumor gene WT1 in embryonic and adult tissues. Development 1993;119:1329–41. Murphy AJ, Bishop K, Pereira C, Chilton-MacNeill S, Ho M, Zielenska M, et al. A new molecular variant of desmoplastic small round cell tumor: significance of WT1 immunostaining in this entity. Hum Pathol 2008;39:1763–70. Musumeci G, Castrogiovanni P, Coleman R, Szychlinska MA, Salvatorelli L, Parenti R, et al. Somitogenesis: from somite to skeletal muscle. Acta Histochem 2015 [Epub ahead of print – Special Issue]. Nakatsuka S, Oji Y, Horiuchi T, Kanda T, Kitagawa M, Takeuchi T, et al. Immunohistochemical detection of WT1 protein in a variety of cancer cells. Mod Pathol 2006;19:804–14.

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

G Model ACTHIS-50982; No. of Pages 12 12

ARTICLE IN PRESS L. Salvatorelli et al. / Acta Histochemica xxx (2015) xxx–xxx

Niksic M, Slight J, Sanford JR, Caceres JF, Hastie ND. The Wilms’ tumour protein (WT1) shuttles between nucleus and cytoplasm and is present in functional polysomes. Hum Mol Genet 2004;13:463–71. Oji Y, Miyoshi S, Maeda H, Hayashi S, Tamaki H, Nakatsuka S, et al. Overexpression of the Wilms’ tumor gene WT1 in de novo lung cancers. Int J Cancer 2002;100:297–303. Oji Y, Nakamori S, Fujikawa M, Nakatsuka S, Yokota A, Tatsumi N, et al. Overexpression of the Wilms’ tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci 2004a;95:583–7. Oji Y, Suzuki T, Nakano Y, Maruno M, Nakatsuka S, Jomgeow T, et al. Overexpression of the Wilms’ tumor gene WT1 in primary astrocytic tumors. Cancer Sci 2004b;95:822–7. Oue T, Uehara S, Yamanaka H, Takama Y, Oji Y, Fukuzawa M. Expression of Wilms tumor 1 gene in a variety of pediatric tumors. J Pediatr Surg 2011;46:2233–8. Parenti R, Perris R, Vecchio GM, Salvatorelli L, Torrisi A, Gravina L. Immunohistochemical expression of Wilms’ tumor protein (WT1) in developing human epithelial and mesenchymal tissues. Acta Histochem 2013;115:70–5. Parenti R, Cardile V, Graziano AC, Parenti C, Venuti A, Bertuccio MP, et al. Wilms’ tumor gene 1 (WT1) silencing inhibits proliferation of malignant peripheral nerve sheath tumor sNF96.2 cell line. PLoS ONE 2014a;9(12):e114333. Parenti R, Puzzo L, Vecchio GM, Gravina L, Salvatorelli L, Musumeci G, et al. Immunolocalization of Wilms’ tumor protein (WT1) in developing human peripheral sympathetic and gastroenteric nervous system. Acta Histochem 2014b;116:48–54. Parenti R, Salvatorelli L, Musumeci G, Parenti C, Giorlandino A, Motta F, et al. Wilms’ Tumor 1 (WT1) protein expression in human developing tissues. Acta Histochem 2015 [Epub ahead of print – Special Issue]. Parham DM, Alaggio R, Coffin CM. Myogenic tumors in children and adolescents. Pediatr Dev Pathol 2012;15:211–38. Parham DM, Barr FG. Classification of rhabdomyosarcoma and its molecular basis. Adv Anat Pathol 2013;20:387–97. Pritchard-Jones K, Fleming S, Davidson EC, Bickmore W, Porteus DJ, Gosden C, et al. The candidate Wilms’ tumor gene is involved in genitourinary development. Nature (Lond) 1990;346:194–7. Salvatorelli L, Bisceglia M, Vecchio G, Parenti R, Galliani C, Alaggio R, et al. A comparative immunohistochemical study of oncofetal cytoplasmic WT1 expression in human fetal, adult and neoplastic skeletal muscle. Pathologica 2011;103:186.

Ramani P, Cowell JK. The expression pattern of Wilms’ tumour gene (WT1) product in normal tissues and paediatric renal tumours. J Pathol 1996;179:162–8. Roberts SG. The modulation of WTI transcription function by cofactors. Biochem Soc Symp 2006;73:191–201. Rosai J. Rosai and Ackerman’s surgical pathology. 10th ed. New York, NY: Elsevier; 2011. Schittenhelm J, Thiericke J, Nagel C, Meyermann R, Beschorner R. WT1 expression in normal and neoplastic cranial and peripheral nerves is independent of grade of malignancy. Cancer Biomark 2010;7:73–7. Sebire NJ, Gibson S, Rampling D, Williams S, Malone M, Ramsay AD. Immunohistochemical findings in embryonal small round cell tumors with molecular diagnostic confirmation. Appl Immunohistochem Mol Morphol 2005;13:1–5. Sehic D, Ciornei CD, Gisselsson D. Evaluation of CITED1, SIX1, and CD56 protein expression for identification of blastemal elements in Wilms tumor. Am J Clin Pathol 2014;141:828–33. Sharma PM, Yang X, Bowman M, Roberts V, Sukumar S. Molecular cloning of rat Wilms’ tumor complementary DNA and a study of messenger RNA expression in the urogenital system and the brain. Cancer Res 1992;52:6407–12. Sheng WQ, Hisaoka M, Okamoto S, Tanaka A, Meis-Kindblom JM, Kindblom LG, et al. Congenital–infantile fibrosarcoma: a clinicopathologic study of 10 cases and molecular detection of the ETV6-NTRK3 fusion transcripts using paraffin embedded tissues. Am J Clin Pathol 2001;115:348–55. Sigauke E, Rakheja D, Maddox DL, Hladik CL, White CL, Timmons CF, et al. Absence of expression of SMARCB1/INI1 in malignant rhabdoid tumors of the central nervous system, kidneys and soft tissue: an immunohistochemical study with implications for diagnosis. Mod Pathol 2006;19:717–25. Singh A, Mishra AK, Ylaya K, Hewitt SM, Sharma KC, Saxena S. Wilms tumor-1, claudin-1 and ezrin are useful immunohistochemical markers that help todistinguish schwannoma from fibroblastic meningioma. Pathol Oncol Res 2012;18:383–9. Vasei M, Moch H, Mousavi A, Kajbafzadeh AM, Sauter G. Immunohistochemical profiling of Wilms tumor: a tissue microarray study. Appl Immunohistochem Mol Morphol 2008;16: 128–34. Wang LL, Perlman EJ, Vujanic GM, Zuppan C, Brundler MA, Cheung CR, et al. Desmoplastic small round cell tumor of the kidney in childhood. Am J Surg Pathol 2007;31:576–84.

Please cite this article in press as: Salvatorelli L, et al. Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors. Acta Histochemica (2015), http://dx.doi.org/10.1016/j.acthis.2015.03.010

Wilms tumor 1 (WT1) protein: Diagnostic utility in pediatric tumors.

Despite Wilms tumor 1 (WT1) protein was originally considered as a specific immunomarker of Wilms tumor, with the increasing use of immunohistochemist...
10MB Sizes 3 Downloads 14 Views