Introduction

Urothelial carcinoma (UC) accounts for about 90% of bladder cancers in industrialized countries with squamous cell carcinoma (1–7%) and adenocarcinomas (0.5–2%) forming a minor component of bladder cancers [1, 2]. UC is remarkable for showing marked diversity in its morphological appearance, which may in part be a reflection of its molecular heterogeneity, resulting in the recognition of various histologic variants. Variant histology is not uncommon and can be seen in as many as 33% of the cystectomy specimens [3]. Despite the high prevalence of variant histology its recognition remains challenging because of under-recognition, misclassification, lack of ancillary testing to confirm variant diagnosis, sampling limitations and the high interobserver variability in part due to evolving diagnostic criteria [4••, 5••, 6,7,8,9,10]. In some patients multiple variants may occur within a tumor, with many pathologists reporting the percentage of each variant within the lesion [11]. These variants are important to recognize because they may have important prognostic or therapeutic implications and misdiagnosis may result in inappropriate treatment [10]. The WHO 2016 classification of tumors of the urinary system and male genital organs addresses many of these issues by updating the diagnostic criteria and the molecular characteristics of the various subtypes (Table 1).

Table 1 Histologic variants of invasive urothelial carcinoma as per the WHO 2016 classification of tumors of the urinary

This review describes the diagnostic criteria of the histologic variants of UC with special emphasis on the morphologic features, molecular profiles and prognostic implications of the identification of the various subtypes. A brief description of the evolving UC subtypes not currently included in the WHO classification is included. Non-urothelial bladder cancer subtypes are not addressed in this review.

Urothelial Carcinoma with Divergent Differentiation

The most common UC variant is UC with divergent differentiation, which includes squamous, glandular, trophoblastic and small cell differentiation (Fig. 1a–d). These tumors are characterized by varying components of urothelial carcinoma (invasive and or in situ) with the aforementioned morphological subtypes [5••]. The general consensus among urologic pathologists is to report these cases with an estimated percentage of the divergent component.

Fig. 1
figure 1

Urothelial carcinoma (UC) with divergent differentiation. Squamous differentiation with squamous pearls (a); glandular spaces lined by cuboidal appearing cells in UC with glandular differentiation (b); syncytiotrophoblastic giant cells scattered along with UC component (c); small cell carcinoma component with urothelial carcinoma in situ (d)

Urothelial Carcinoma with Squamous Differentiation

Squamous differentiation is defined by the presence of intercellular bridges or keratinization (Fig. 1a) and is seen in up to 40% of urothelial carcinomas of the bladder [7, 8]. They are frequently associated with high-grade and high-stage tumors [12•] and may show less favorable response to intravesical BCG and intravesical chemotherapy [13•, 14]; however, the response to neoadjuvant chemotherapy may be comparable with that of conventional urothelial carcinoma [15•, 16,17,18,19]. The presence of squamous differentiation, however, has been associated with higher rates of recurrences and poor prognosis [13•, 18, 20].

One of the challenges in the identification of this variant, especially in transurethral resection (TUR) and biopsy specimens, is its distinction from pure squamous cell carcinoma of the bladder and from secondary involvement of the bladder by squamous cell carcinoma of other pelvic organs. Both squamous cell carcinomas and UC with squamous differentiation share many basal markers including cytokeratin 5/6 and p63. In the absence of specific immunohistochemical markers to make this distinction, and as suggested by previous studies, I rely on the clinical history and the absence of a conventional urothelial carcinoma component to make this distinction [7, 10]. Unlike squamous carcinoma of the cervix, human papilloma virus is generally not considered to be causative in the development of squamous cell carcinoma of the bladder or UC with squamous differentiation [21].

Urothelial Carcinoma with Glandular Differentiation

Glandular differentiation is not as frequent as squamous differentiation but has been documented in 6–18% of invasive bladder cancers [7, 22,23,24]. Glandular differentiation is characterized by true gland formation (Fig. 1b) resembling colonic adenocarcinoma, signet ring carcinoma or mucinous/colloid carcinoma (tumor cells floating in a pool of mucin). They may be associated with urothelial carcinoma in situ or rarely with in situ urothelial carcinoma with glandular differentiation [25]. Pseudo glandular spaces with intracellular mucin may be seen in conventional UC and should not be confused with either UC with glandular differentiation or bladder adenocarcinoma [26].

Robust immunohistochemical markers to distinguish UC with glandular differentiation, from primary bladder adenocarcinomas and secondary involvement /metastatic colonic adenocarcinoma are lacking, making this distinction challenging [27, 28•]. Often in limited samples a definite diagnosis may not be possible and pathologists may give a general descriptive diagnosis and favor one of the differentials over the other. TERT mutations are seen in up to 70% of urothelial carcinoma with glandular differentiation and not in bladder adenocarcinomas [23]; however, this is seldom used in routine clinical practice.

UC with glandular differentiation tends to present at a higher stage but it is not a negative prognostic indicator in stage-matched patients [29].

Urothelial Carcinoma with Trophoblastic Differentiation

UC showing syncytiotrophoblastic giant cells are rare (Fig. 1c), although the use of immunohistochemical markers for the beta subunit of human chorionic gonadotropin (βhCG) can highlight trophoblastic differentiation in up to 35% of UC [30].

βhCG expression within urothelial carcinoma cells without morphological evidence of trophoblastic differentiation correlates with the grade and stage of tumor; however, it is not included in this subgroup. These variants need to be distinguished from pure choriocarcinoma of the bladder, which is extremely rare and requires the demonstration of isochromosome 12p, which is a hallmark of germ cell tumors [31]. Elevated serum βhCG has been reported in 20–76% of metastatic urothelial carcinomas, and response to chemotherapy in these patients can be correlated with the levels of this marker [32,33,34,35].

Other Forms of Divergent Differentiation

UC can show neuroendocrine differentiation in the form of small cell carcinoma (Fig. 1d), and any small cell carcinoma component when present should be reported. TERT promoter mutations have been reported in 55–95% of these tumors, similar to the concomitant UC component suggesting a common clonality [36••, 37]. TERT promoter mutation status may also help identify the site of origin of these tumors, as they are not present in small cell carcinomas of other sites including the prostate [36••, 38]. Coalterations in P53 and RB1, with resultant loss in function, are commonly seen in small cell carcinoma [37, 39••]. The small cell component is usually negative for both luminal and basal markers with few cases showing focal expression of CK5/6, a marker for the basal molecular subtype. This suggests evolution from a basal phenotype and better response to cisplatin-based chemotherapy [39••]. These tumors are treated similar to their counterpart in the lungs. Rarely urothelial carcinomas may show germ cell differentiation, including yolk sac tumor [5••].

Nested, Including Large Nested Urothelial Carcinoma

As per the 2016 WHO classification, large nested variant of UC and UC with small tubules and microcysts that were earlier considered as distinct entities are now included within the category of nested variant [5••, 40, 41••]. This variant is characterized by its deceptively benign cytology and its superficial resemblance to benign mimickers of UC including von Brunn nests, nephrogenic adenoma, and paraganglia [40, 41••, 42,43,44,45].

Nested variant is characterized by a disorderly proliferation of discrete to confluent nests of urothelial cells usually with minimal cytologic atypia (Fig. 2a). The absence of cytologic atypia can make the diagnosis of this variant extremely challenging in small biopsy specimens and transurethral resections. In many instances, the diagnosis may be delayed until the tumor declares itself with unequivocal evidence of invasion of the muscularis propria. Unlike the usual nested variant, which is composed of small- to intermediate-sized nests, the large nested variant is characterized by irregularly infiltrating large aggregates of bland tumor cells which may sometimes be difficult to differentiate from conventional UC with an inverted growth pattern [46].

Fig. 2
figure 2

Cytologically bland nests of invasive carcinoma characteristic of nested variant (a); micropapillary UC with small nests within lacunar spaces (b); tumor cells obscured by brisk inflammatory infiltrate in lymphoepithelioma-like UC (c); singly discohesive bland plasma cell-like cells of plasmacytoid UC (d)

Nested urothelial carcinomas may be admixed with a component of conventional UC or may occur in a pure form. They usually present as a high-stage disease, which may in part reflect the delay in diagnosis, associated with this tumor [47]. Pure large nested variant may have a better prognosis than cases with a mixed histology [41].

The immunohistochemical profile of this variant is similar to conventional UC. In particularly challenging cases the presence of TERT promoter mutation within the tumor cells can help distinguish it from its benign mimics [48]. Preliminary studies have shown that these tumors express immunohistochemical markers characterized by the luminal molecular subtype of urothelial carcinomas [49, 50••].

Microcystic Urothelial Carcinoma

Microcystic urothelial carcinoma, like nested urothelial carcinoma, is an example of urothelial carcinoma characterized by bland cytologic features. It is comprised of round-oval cysts up to 2 mm in diameter lined by urothelial, low columnar or flattened epithelium [51]. The cyst lining may be focally denuded and intraluminal secretions and calcifications may be present within some of the cysts. The cysts are infiltrating; however, a stromal response may be lacking. Focal high-grade conventional urothelial carcinoma may be present in up to 40% of the cases. Their immunohistochemical profile is similar to that of conventional UC [52].

These tumors should be differentiated from their benign mimics including cystitis cystica and cystitis glandularis [53]. TERT promoter mutation studies may help in difficult cases.

Micropapillary Urothelial Carcinoma

This variant has been reported more commonly in men with a peak incidence in the 6th decade of life. It is associated with a poorer prognosis and usually presents at a higher stage with lymph node metastasis [54, 55]. The reported prevalence of this variant varies from 0.7 to 8% [56,57,58] and this may in part reflect the high interobserver variability and the cutoffs used in the diagnosis of this variant [58, 59]. The diagnosis of this tumor should be restricted to infiltrating tumors with slender filiform processes without fibrovascular cores and/or multiple small tumor nests within a single lacunar space (Fig. 2b). The tumor nests often show peripherally oriented nuclei with “reverse polarization” of the basal and luminal aspects of the cell, highlighted on electron microscopy and MUC1 staining [60]. Cytoplasmic vacuolization with indentation of the nuclei (ring forms) is also a characteristic of this tumor [59]. Lymphovascular invasion has also been commonly reported with this variant. Micropapillary carcinoma is frequently admixed with conventional UC and carcinoma in situ is present in more than 50% of the cases [61]. It is unclear if the prognosis of this variant depends on the proportion of the micropapillary component [61, 62]; however, most urologic pathologists report the percentage of this variant in the tumor. Although a surface micropapillary component may be identified in a non-invasive UC or rarely as a variant of urothelial carcinoma in situ, these should not be interpreted as micropapillary urothelial carcinoma. Micropapillary UC in addition to the usual urothelial markers also show immunoreactivity for MUC1 and CA-125 [28•]. They also usually express luminal markers including FOXA1 [37, 49] and have been reported to have a high prevalence of TERT promoter mutations [63••, 64]. HER2 amplifications have been documented in 15–42% of the cases [63, 65, 66].

Limited response to BCG therapy and adverse outcome despite chemotherapy has been reported in some studies, prompting early cystectomy in patients with T1 disease [67]. However, other studies have shown the utility of a bladder-sparing approach in select non-muscle-invasive patients [68, 69]. A recent prospective trial has also reported efficacy with aggressive neoadjuvant chemotherapy [70]. Neoadjuvant chemotherapy although decreased the frequency of non-organ confined disease it did not translate into a statistically significant overall survival benefit [71••]. HER2-amplified tumors have been associated with a worse cancer-specific survival, opening up the potential role of ERBB2-targeted therapy [65, 63].

Lymphoepithelioma-Like Urothelial Carcinoma

Lymphoepithelioma-like urothelial carcinoma is similar in morphology to the lymphoepitheliomas arising elsewhere in the body including the nasopharynx. However, unlike tumors of the nasopharynx this variant of UC is not associated with Epstein-Barr virus infection [72, 73]. It is more commonly seen in older men (mean age, 69 years) and usually presents as a stage 2 or 3 tumor [5••, 74].

It may occur either in the pure form or admixed with conventional urothelial carcinoma or other variants. The tumor is composed of sheets of undifferentiated cells with large pleomorphic nuclei and prominent nucleoli with indistinct cell borders forming a syncytium. The background comprises of an inflammatory infiltrate comprising predominantly of lymphocytes, which in some instances may be so dense so as to obscure the tumor cells (Fig. 2c). The tumor cells mark for urothelial markers including p63 and GATA-3.

Although it was suggested that pure/predominant forms of this carcinoma are associated with a relatively favorable outcome with good response to chemotherapy, a recent study of 30 cases has reported similar outcomes to conventional UC [75].

Plasmacytoid Urothelial Carcinoma

This is a rare but aggressive variant of UC characterized by cells resembling plasma cells that present at a high stage with extravesical disease and intraperitoneal spread in 27–33% of the cases [76, 77, 78•]. They also have higher incidence of lymph node metastasis and are more likely to have positive margins after radical cystectomy [78•]. They generally have a poor outcome with higher rates of recurrence and death but in a recent study of 98 patients, plasmacytoid urothelial carcinoma was not associated with worse overall mortality compared with conventional UC [79••].

This variant is characterized by singly infiltrating, discohesive cells in an edematous or myxoid stroma. The tumor cells have eccentric enlarged hyperchromatic nuclei with eosinophilic to clear cytoplasm, without significant cytologic atypia (Fig. 2d). Signet ring cells with or without intracytoplasmic mucin may be identified, but unlike a signet ring adenocarcinoma extracellular mucin is absent [80, 81, 5••]. A high-grade urothelial carcinoma component is seen in about 50% of the cases. Perrino et al. [82•] recently classified plasmacytoid UC into three morphological subtypes (classic, desmoplastic, and pleomorphic) and reported that the desmoplastic variant had the worst clinical behavior. However, many of their cases of the desmoplastic and pleomorphic variants appear to have a morphologic overlap with UC with rhabdoid morphology. Further studies are required to validate the significance of these findings.

Up to 84% of these tumors show truncating mutations in the CDH1 gene, which encodes for E-cadherin, whereas these mutations have not been reported in conventional UC ( [83••, 84]. These mutations lead to the loss of E-cadherin (a cell adhesion molecule), which may be responsible for the marked discohesion of tumor cells seen in this variant [85]. Recently Fox et al. showed the lack of immunohistochemical staining for RB (retinoblastoma) protein in 62% of their cases, suggesting abnormal function of the RB gene [78•]. Plasmacytoid urothelial carcinoma must be differentiated from tumors arising from the breast and gastrointestinal tract among others because of overlapping morphological features. This is possible by the judicious use of immunohistochemical markers [85, 86•].

Giant Cell Urothelial Carcinoma

Giant cell urothelial carcinoma is a rare aggressive variant characterized by highly atypical giant cells usually admixed with conventional UC component [87•]. Rarely the tumor may be composed entirely of large pleomorphic tumor giant cells, and in such instances immunohistochemical stains may be necessary to identify its urothelial origin. Atypical mitotic figures and areas of necrosis are often present. They are usually highly invasive with involvement of the muscularis propria. The prognosis is uniformly poor [88].

Lipid-Rich Urothelial Carcinoma

This is another rare variant of urothelial carcinoma, with less than 40 cases reported in literature [5••]. The tumor cells resemble lipoblasts and have one or more vacuoles in the cytoplasm that indent the nucleus. The lipid-rich component usually comprises 10–50% of the tumor and is usually admixed with a component of conventional or other UC variants. These tumors usually present at a higher stage, with 60% of the patients dying of the disease within 58 months [89].

Clear Cell (Glycogen-Rich) Urothelial Carcinoma

The clear cytoplasm seen in this variant is because of the presence of intracytoplasmic glycogen, which stains with periodic-acid-Schiff (PAS) stain and disappears after diastase digestion [90].They resemble the cells seen in clear cell renal cell carcinoma, but can be differentiated from it because of its immunoreactivity with urothelial markers including GATA-3 and p63. This variant is also usually associated with in situ, papillary or conventional urothelial carcinoma [5••]. They are extremely rare with fewer than 25 cases reported in literature and therefore it is difficult to comment upon the effect of this variant morphology on prognosis.

Sarcomatoid Urothelial Carcinoma

Sarcomatoid urothelial carcinoma usually presents as advanced disease with poor clinical outcomes [91]. Radiation exposure and chemotherapy with cyclophosphamide have been documented as known risk factors for this variant [5••]. On histology the sarcomatoid component is composed of high-grade spindle or pleomorphic cells indistinguishable from those of a sarcoma. Heterologous components including osteosarcoma, chondrosarcoma, and rhabdomyosarcoma may be identified in some of the tumors and may portend a worse prognosis [92]. Conventional urothelial, squamous, glandular, or small cell carcinoma may be identified admixed with the sarcomatoid component. The epithelial component expresses vimentin in up to 100% of the cases while the sarcomatoid component shows at least focal immunoreactivity for high molecular weight cytokeratins, p63 and GATA-3 [93]. Markers of epithelial-mesenchymal transition have been documented in this tumor both immunohistochemically and at a molecular level with nearly half of the tumors showing a heavily infiltrated immune phenotype [93, 94••]. These findings have important implications for prognostication and development of therapies for this aggressive variant of bladder cancer. Neoadjuvant chemotherapy may be beneficial in muscle-invasive cancers to downstage the tumor at the time of cystectomy, but the overall prognosis remains poor [71••].

Poorly Differentiated Tumors (Including Those with Osteoclast-Like Giant Cells)

This entity has been introduced in the WHO 2016 classification and represents a wide range of tumors with mixed morphologies such as small cell carcinoma, giant cell carcinoma, and osteoclast-rich undifferentiated tumors. The latter tumors are characterized by osteoclast-like giant cells (CD68 positive) with undifferentiated UC cells (cytokeratin positive). Most of these cases are associated with in situ or invasive carcinoma, and the mononuclear cells mark for urothelial markers including GATA-3 [95••]. These are rare tumors and have been reported to be associated with a poor prognosis [95••].

Evolving Entities of Urothelial Carcinoma

There are certain morphological variants of urothelial carcinoma that have not yet been included in the 2016 WHO classification of bladder cancers, because of their rarity and consequent paucity of literature regarding their biology and behavior. A brief description of these tumors is included below.

Pseudoangiosarcomatous Variant of Urothelial Carcinoma

A total of 15 cases of this rare variant have been reported in literature and they have all been associated with poor prognosis [96, 97]. These tumors histologically resemble angiosarcomas with discohesive tumor cells forming pseudolumina (Fig. 3a). They are commonly accompanied with other UC types and show immunoreactivity for urothelial markers [96].

Fig. 3
figure 3

Vascular-like spaces lined by tumor cells in pseudoangiosarcomatous UC (a); rhabdoid UC with singly dispersed high-grade malignant cells in a myxoid stroma (b)

Urothelial Carcinoma with Myxoid Stroma

This variant shows extensive mucinous-myxoid stroma with cords of cells suspended within the stroma, morphologically resembling patterns seen in chordoma, yolk sac tumor, and myxoid chondrosarcoma. These tumors present with high-stage disease, have an urothelial immunophenotype, and usually are associated with some component of conventional UC [98].

Urothelial Carcinoma with Rhabdoid Features

UC with rhabdoid features are tumors comprised of undifferentiated tumor cells having eccentric nuclei, prominent nucleoli, and abundant inclusion like eosinophilic cytoplasm (Fig. 3b). These tumors are extremely rare and portend a poor prognosis [99]. Up to 70% of these tumors show loss of at least one SW1/SNF (chromatin remodeling complex) subunit. SMARCA2 is most frequently lost followed by ARID1A, SMARCB1/INI1, SMARCA4, and SMARCC1 [100•].

Molecular Classification of UC with Reference to UC variants

The molecular landscape of UC is very large and complex and recently there have been many attempts to classify invasive UC using molecular classifiers based on RNA expression profiles. Many of these molecular classifiers have overlapping profiles, and the most comprehensive of these classifications were proposed by the Lund University group and the TCGA [101, 102••]. The TCGA classification identifies 5 subtypes of muscle-invasive UC, including luminal, luminal-papillary, luminal infiltrated, basal-squamous, and neuronal. These subtypes are characterized by specific RNA expression signatures and have different prognostic and therapeutic implications. Although some of the UC variants are characterized by certain characteristic molecular alterations as mentioned previously in this review, there is limited information regarding the classification of UC variants using the abovementioned molecular classifiers. Using RNA expression or immunohistochemical profiles, micropapillary, nested, and plasmacytoid urothelial carcinomas were found to have luminal features, whereas urothelial carcinoma with squamous differentiation was found to have basal characteristics [37, 49, 50••]. However, the available data in these variants is limited and further comprehensive studies are needed to test the validity of these molecular classifiers in UC variants.

Conclusions

UC with variant histology is a heterogeneous group of tumors that are being increasingly identified because of their distinct morphological features and their reported association with variable clinical prognosis. Distinct molecular alterations have been described for some of these morphologic subtypes, which have implications for the use of targeted therapies. The use of molecular classifiers, currently used for invasive conventional UC, has yet to be fully explored in UC variants.