The impact of histological variants on bladder cancer outcomes
Review Article: Medical Tests and Health Care: Pathology

The impact of histological variants on bladder cancer outcomes

Tania Processali#, Alberto Diminutto#, Maria Angela Cerruto, Alessandro Antonelli

Urology Clinic, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy

Contributions: (I) Conception and design: All authors; (II) Administrative support: MA Cerruto, A Antonelli; (III) Provision of study materials or patients: T Processali, A Diminutto; (IV) Collection and assembly of data: T Processali, A Diminutto; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Professor Alessandro Antonelli. Urology Clinic, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Italy Borgo Trento Hospital, Piazzale Stefani, 1, 37126, Verona, Italy. Email: alessandro_antonelli@me.com.

Abstract: Pure urothelial carcinoma is the most common histology of bladder cancer, consisting of about 75% of all cases. The remaining cases are represented by histological variants (HVs). Despite an apparent and recent rise in the incidence of variant histology (VH) in pathological reports, the real epidemiology, presentation and prognostic value on oncological outcomes are still not well established. Pathological identification of HVs indeed is challenging and at risk of under-recognition and misclassification, both at transurethral resection (TUR), due to the lack of a standardized technique, and at definitive histology after cystectomy because of the need for dedicated genitourinary pathologists. Genomic sequencing has led to identify molecular subtypes of bladder cancer eventually related to HVs and potentially implicated in the management of bladder cancer. The aim of this review is to present an overview of the literature regarding the impact of VH and molecular subtypes on bladder cancer outcomes. A narrative review of the literature was conducted applying several terms and combinations of them regarding HVs and molecular subtypes. The main findings are that HVs are indicative of a more aggressive disease, often advanced at presentation. When matched for stage and grade, the oncological outcomes are similar with pure urothelial carcinoma, except for pure squamous cell and signet ring cell carcinoma variants that invariably portend a poor prognosis. Radical cystectomy (RC) remains the treatment of choice and should be considered even in non-muscle-invasive cases if some forms of VH are present. Neoadjuvant chemotherapy is recommended in chemo-sensitive variants. Molecular composition could be related to the expression of specific VH and new subtypes could be identified with further investigations. Future developments in TUR procedures, genomic techniques and dedicated genitourinary pathologists will lead to a better definition of each HV, along with related prognosis and response to treatments.

Keywords: Bladder cancer; urothelial; variant histology (VH); molecular subtypes; oncological outcomes


Received: 10 December 2019; Accepted: 20 January 2020; Published: 25 March 2020.

doi: 10.21037/amj.2020.02.02


Background

Pure urothelial carcinoma is the most common histology of bladder cancer, consisting of about 75% of all cases (1). The remaining cases are represented by variant histology (VH), divided into urothelial and non-urothelial subtypes.

Histological variants (HVs) can arise in variable proportion in bladder cancer, as part of urothelial carcinoma or in pure forms.

The 2016 World Health Organization (WHO) classification of tumours in the urinary tract stressed out the need to mention the presence and percentage of VH in the pathological report (2), as commonly associated, like in upper urinary tract neoplasm, with advanced disease and correlated with clinical implications affecting the prognosis of patients and guiding the treatment algorithm (3,4).

Pathological identification of VH is challenging, with high risk of under-recognition and misclassification due to several reasons (5), both at transurethral resection (TUR) and radical cystectomy (RC) levels.

The techniques for TUR of bladder tumours are quite heterogeneous and still poorly standardized, with great variability in quality and quantity of specimens retrieved: a resection in multiple specimens is often a “puzzling challenge” for histological report due to cautery damage and lack of spatial orientation (6). An “en-bloc” resection could be preferable to recognize HVs, even if present in small percentage (7).

Few studies in literature evaluated the accuracy of initial TUR in the identification of VH compared to RC, with conflict results. While Abufaraj et al. demonstrated similar accuracy in VH reporting between TUR and RC, up to 84% (8), other three studies showed poor concordance between the two specimens, depending on the type of HVs (9-11).

Moreover, several studies highlighted the importance of the analysis of TUR specimens by expert and dedicated genitourinary pathologists. In particular, Shah et al. demonstrated that HVs had not been reported in 44% of cases evaluated by non-dedicated pathologists (12).

Despite an apparent and recent rise in the incidence of VH in pathological reports, probably due to the increased awareness by pathologists, the real epidemiology, presentation and prognostic value on oncological outcomes are still not well established. Literature is still based principally on case reports or retrospective studies, even for the exclusion of patients with HVs from clinical trials.

Latest researches postulated that the phenotype of different HVs could be the expression of underlying diversity in molecular composition of the tumour, through a process of progressive dedifferentiation as observed in other organs (13).

In recent years, the increasing spread of research in genomic next-generation sequencing has led to identify specific molecular subtypes of bladder cancer. Most of classification systems differentiate urothelial tumours in major subtypes luminal and basal, respectively characterized by the expression of urothelial or basal cell/squamous differentiation markers, and in additional subtypes (14-17). Moreover, evidences from molecular analysis of some histologically defined variants, like squamous differentiation or neuroendocrine tumour, led to identify specific mutations present also in neoplasms lacking classic histologic hallmarks (18).

Molecular classification has potential implications in the management of bladder cancer, both for therapeutic and prognostic purposes: recent studies demonstrated different survival and response to chemotherapies and target therapies for the different subtypes of urothelial carcinoma (19).

Therefore, the optimal management of bladder cancer with VH is already not established, with a general recommendation by international guidelines for early RC also in non-muscle-invasive cases (20,21).

The aim of this review is to present an up-to-date overview of the literature regarding the impact of VH and molecular subtypes on bladder cancer outcomes in relation to the actual standard of care treatment options for muscle-invasive (RC, perioperative neoadjuvant or adjuvant chemotherapy and trimodality therapy) and non-muscle-invasive (intravesical immunotherapy and early RC) urothelial carcinoma.


Methods

A non-systematic review of the literature was performed in October 2019, using the PubMed/MEDLINE database applying the following terms and combinations: “bladder cancer”, “bladder tumour”, “variant histology”, “histological variants”, “urothelial carcinoma”, “non-urothelial carcinoma”, “nested”, “micropapillary”, “signet ring cell carcinoma”, “microcystic”, “sarcomatoid”, “squamous differentiation”, “glandular differentiation”, “clear cell”, “plasmacytoid”, “lymphoepithelioma-like carcinoma”, “squamous cell carcinoma”, “small cell carcinoma”, “large cell carcinoma”, “neuroendocrine”, “adenocarcinoma”, “mixed variants”, “radical cystectomy”, “radiotherapy”, “neoadjuvant chemotherapy”, “adjuvant chemotherapy”, “molecular subtypes”, “basal”, “luminal”. Meta-analysis, original articles, reviews, case reports and case series were selected. Only English-language papers were included.

Due to the rarity of VH, no time restriction was applied and studies on non-muscle-invasive bladder cancer were not excluded.

Urachal adenocarcinoma, endometrioid carcinoma, melanocytic tumours, mesenchymal tumours, haematopoietic and lymphoid tumours were excluded.


Results

The characteristics and oncological outcomes for each VH, related to each treatment option, are summarized in Table 1.

Table 1

Variant histology outcomes for treatment option, compared with pure urothelial carcinoma, and molecular subtypes

Variant Outcomes after RC (adjusted for stage): inferior/similar/superior/variable Outcomes after perioperative CT: beneficial/no effect/no data/not preferred Outcomes after trimodality therapy: inferior/similar/superior/no data/not preferred Outcomes after intravesical therapy with BCG (NMIBC): inferior/similar/superior/variable/no data Molecular subtype: luminal/basal/unknown
Nested Similar No data No data Inferior Luminal/basal
Microcystic Similar No data No data No data Luminal/basal
Micropapillary Similar/inferior Beneficial/no effect No data Inferior Luminal
Lymphoepithelioma-like Superior Beneficial No data No data Basal
Plasmacytoid/signet ring cell Similar/inferior Beneficial/no effect Inferior (Signet ring cell) No data Luminal
Sarcomatoid Inferior/similar No benefit No data No data Basal
Clear cell Variable No data/not preferred No data No data Luminal/basal
Mixed variants Similar Beneficial (MVAC) Similar Similar (glandular) Luminal (glandular)/basal (squamous)
Neuroendocrine tumours Inferior/similar Beneficial (Etoposide) No data No data Basal
Adenocarcinoma Similar No benefit No data No data Unknown
Pure squamous cell carcinoma Inferior No benefit No data No data Unknown

RC, radical cystectomy; CT, chemotherapy; BCG, bacillus Calmette-Guerin; NMIBC, non-muscle-invasive bladder cancer; MVAC, methotrexate vinblastine adriamycin cisplatin.


VH of bladder cancer

Urothelial variants

Nested

Nested is a rare HV, representing the 0.3% of invasive bladder cancers. It usually presents with high rates of muscle invasion (70%), locally advanced disease and metastases (22).

The morphological appearance of nested variant resembles benign von Brunn nests or nephrogenic adenoma on the surface but demonstrates focal high-grade cytological atypia within the base, with a disordered and infiltrative proliferation (23).

In two cohort studies, patients with nested variant showed similar oncological outcomes, in terms of 5-year recurrence-free survival (RFS) and cancer-specific survival (CSS), compared to patients with pure urothelial carcinoma when treated with RC and matched for stage (5,23).

No data exists on the impact of perioperative chemotherapy (neoadjuvant or adjuvant) or trimodality therapy in patients harbouring nested variant.

The use of intravesical immunotherapy in non-muscle-invasive nested variant is reported in only one study in literature: in seven patients treated with bacillus of Calmette-Guerin (BCG) the RFS, progression-free survival (PFS), CSS and overall survival (OS) were significantly inferior compared to patients with pure urothelial carcinoma (24).

Microcystic

Microcystic variant has an incidence around 1% of bladder tumours (25) and it is histologically composed of varying sizes cysts with intraluminal secretions, usually lined by transitional cells or low columnar cells with mucinous differentiation. It is deeply infiltrative, with early invasion of muscularis propria and usually associated with high-grade urothelial carcinoma (26).

Poor and limited data exists in literature on microcystic variant, mostly case reports and small case series, the biggest counting 20 patients. This VH is associated with poor prognosis but, when matched for stage, no difference in oncological outcomes after RC is observed compared with pure urothelial carcinoma (27).

No data exists on the use of perioperative chemotherapy or trimodality therapy, neither on the use of intravesical therapy in patients with microcystic variant.

Micropapillary

Micropapillary is an aggressive urothelial carcinoma variant that accounts for 2% to 5% of all bladder cancers. It histologically presents with multiple small nests and papillae in lacunar spaces and it is usually diagnosed at advanced stage, with high rates of carcinoma in situ and regional lymph node metastases (28-30).

For its behaviour of rapid progression to muscle invasion and metastases, early cystectomy in non-muscle-invasive micropapillary carcinoma is advocated as the optimal management over intravesical immunotherapy in many centres (31-33).

Nevertheless, bladder preserving strategies were explored in other studies for non-muscle-invasive stage, with a non-inferior OS in selected patients harbouring small percentage of micropapillary component (32,34).

Data from National Cancer Data Base (NCDB) and single-centre retrospective studies showed inferior OS in patients with micropapillary variant compared to patients with pure urothelial carcinoma (31,35), nevertheless when matched for disease stage, OS is similar (11,28-30).

Despite a single report of Monn et al. showing inferior OS for micropapillary variant (36), a recent meta-analysis of seven studies demonstrated that patients with this variant have similar RFS, CSS and OS to patients with pure urothelial carcinoma after RC (37).

The role of neoadjuvant chemotherapy remains unclear due to conflicting results reported in literature.

In a comparative study on 100 consecutive patients treated with RC with or without neoadjuvant chemotherapy, despite a 61% of pathological downstaging, no benefit on 5-year OS was observed in the neoadjuvant chemotherapy group, which experienced a higher non-organ-confined disease rate (33).

Same results were showed by a similar study on 82 patients (38) and by a recent meta-analysis (37): despite a higher rate of downstaging to pT0, no significant differences were noted in RFS, CSS and OS (38).

The MD Anderson Cancer Center experience on 103 patients with micropapillary variant was reviewed accounting for possible selection bias, showing a beneficial effect of neoadjuvant chemotherapy in those with muscle-invasive disease and absence of tumour-associated hydronephrosis (39).

Only one study explored the role of adjuvant chemotherapy in micropapillary variant, showing a higher recurrence rate compared to pure urothelial carcinoma (40), but did not evaluate if adjuvant chemotherapy could give an advantage with respect to no adjuvant treatment.

No data regarding the use of trimodality therapy is available.

Lymphoepithelioma-like

Named for its histological similarities to nasopharyngeal lymphoepithelioma, this variant represents less than 1% of all urothelial carcinomas (41).

Its peculiar characteristics are represented by poorly differentiated tumour cells with nuclear atypia and indistinct cell borders, with a massive infiltration of polyclonal T and B cells (42).

In a systematic review of 140 cases, Yang et al. demonstrated a higher disease-free survival (DFS) for patients harbouring this variant when treated with RC compared to partial cystectomy or TUR alone (43).

In its pure form, this variant has a high response rate to platinum-based chemotherapy and a low metastatic potential. In mixed forms, the prognosis mostly depends by the other associated variants (42).

The optimal management of this VH should be therefore RC with neoadjuvant chemotherapy.

No data is available regarding intravesical or trimodality therapy.

Plasmacytoid and signet ring cell

Plasmacytoid and signet ring cell carcinoma are classified in the same group by the 2016 WHO update (2).

Data from recent series estimates that plasmacytoid variant represents 1% to 4.9% of urothelial carcinoma and a specific differential diagnosis with other plasmacytoid bladder neoplasms, such as melanoma and lymphoma, is mandatory (44).

Plasmacytoid variant is histologically characterized by discohesive cells with eccentrical nuclei and eosinophilic cytoplasm (45) and has a characteristic pattern of invasion, with distant microscopic satellite lesions associated with a lack of desmoplastic reaction between normal and neoplastic tissues. These characteristics determine a difficult surgical resection and a consequent high risk of positive surgical margins at RC, with high rates of recurrence (46).

This variant is frequently associated to peritoneal carcinomatosis probably related to loss-of-function mutations of E-cadherin, conferring enhanced cell migration activity to the tumour (47-49).

Despite multiple studies in literature showed that plasmacytoid variant is associated with locally advanced disease and lymph node metastases (44,45,49-51), when matched for stage OS is similar to pure urothelial carcinoma (46,50).

Nevertheless, in a single centre comparative study including patients with plasmacytoid variant underwent RC, Monn et al. found that this variant was independently associated with an increased mortality risk. Considering its aggressive nature, early RC should be suggested even in non-muscle-invasive cases (36).

Promising initial data on the chemo-sensitivity of plasmacytoid variant (47) was not confirmed by recent experiences that failed to demonstrate a benefit from neoadjuvant chemotherapy in OS, even if pT0 is achieved (51,52).

Considering all these findings, no study regarding the role of trimodality therapy on this variant is available.

Based on data from the 685 patients with signet ring cell carcinoma of the Surveillance Epidemiology and End Results (SEER) database and from the NCDB, signet-ring variant is associated with inferior OS after RC compared to pure urothelial carcinoma (53,54), with a 5-year CSS of 24.8% (55).

Nevertheless, RC guarantees better outcomes when compared to bladder-preserving strategies including trimodality therapy (56).

No data on neoadjuvant or adjuvant chemotherapy is available.

Sarcomatoid

Sarcomatoid variant represents 0.3% of all urothelial neoplasms. The peculiar characteristic of this VH is the presence of high-grade spindle tumour cells and both epithelial and mesenchymal differentiation. Previous pelvic irradiation and intravesical cyclophosphamide exposure are two established risk factors (57).

Differential diagnosis is challenging as this variant can mimic the morphology of a variety of other neoplasms.

Sarcomatoid variant presents usually at advanced stages and is associated with poor prognosis and inferior OS compared to pure urothelial carcinoma when treated with RC (57-60).

Only Moschini et al. (11) and Monn et al. (36) did not found sarcomatoid variant as an independent predictor of worse OS.

Based on data from NCDB, neoadjuvant chemotherapy seems to offer no significant benefit on OS in patients with sarcomatoid variant (61-63).

In their study, Sui et al. analysed the role of trimodality therapy, demonstrating a worse OS when compared to RC with or without neoadjuvant chemotherapy (61).

No study regarding effectiveness of intravesical therapy is available in literature. The optimal management for this VH remains therefore RC even in non-muscle-invasive cases.

Clear cell

Clear cell is an extremely rare VH composed of cells with glycogen-rich cytoplasm. Literature is sparse with only 16 case reports, so poor data exists on prognosis and oncological outcomes. This variant seems to have a male sex predisposition (male to female ratio of 16:1) and to be associated with an aggressive behaviour, as fast progression to muscle invasion and metastases.

Differential diagnosis includes clear cell adenocarcinoma of the bladder, clear cell carcinoma of the female genital tract or metastatic clear cell carcinoma from other organs (64).

Perioperative chemotherapy was rarely used with upfront RC as preferred treatment with wide variable oncological outcomes (65-67).

No data regarding trimodality therapy is available.

Urothelial carcinoma with squamous and/or glandular divergent differentiation (mixed variant)

Urothelial carcinoma with divergent differentiation, also known as mixed variant, is the most common VH of bladder cancer. The term “mixed variant” refers to those tumours that harbour urothelial carcinoma and squamous and/or glandular differentiation in variable proportion.

Squamous is the most common divergent differentiation, counting up to 40% of invasive urothelial carcinoma, while glandular differentiation occurs in up to 18% of invasive tumours.

Mixed variants are morphologically characterized by the presence of intercellular bridges or keratinization in squamous differentiation and intratumoural tubular or enteric gland-like spaces in glandular differentiation.

These entities must be distinguished from pure squamous cell carcinoma and pure adenocarcinoma of the bladder, which are different HVs without urothelial component, with different behaviour and prognosis.

Mixed variant usually presents with locally advanced disease and lymph node metastases (68-73).

While earlier reports showed inferior OS (71,72), Mitra et al. and Kim et al. demonstrated a similar 5-year OS and CSS after RC for patients with mixed variant compared to those with pure urothelial carcinoma in two single-centre studies (68,69).

Data regarding the benefits of neoadjuvant chemotherapy remains conflicting.

Despite Zargar-Shoshtari et al. reported a significant pathological downstaging after neoadjuvant chemotherapy (74), Minato et al. showed poorer response to this treatment (75).

The strongest evidences for neoadjuvant chemotherapy come from a secondary analysis of the Southwest Oncology Group (SWOG)-directed intergroup randomized trial S8710 of neoadjuvant platinum-based chemotherapy (with MVAC: methotrexate, vinblastine, doxorubicin and cisplatin): 32 of 59 patients with VH that received neoadjuvant chemotherapy showed a large survival benefit in multivariate analysis (70).

Available data is scarce regarding the role of trimodality therapy in patients with mixed HVs. In a retrospective single centre study, patients treated with trimodality therapy showed similar 5- and 10-year CSS and OS when compared with patients with pure urothelial carcinoma (76).

The only data available regarding the use of intravesical therapy in mixed variants derives from a small case series of 24 patients with glandular divergent differentiation: six patients were successfully treated with BCG (77).

The optimal management of mixed variant remains RC with or without neoadjuvant chemotherapy but, in case of glandular divergent differentiation, intravesical therapy with BCG can be considered (78).

Non-urothelial variants

Neuroendocrine tumours (small/large cell)

Neuroendocrine tumours of the bladder, especially small cell VH, represent 0.5–0.7% of bladder cancer.

Bladder neuroendocrine small cell tumour is histologically similar to pulmonary small cell carcinoma, characterized by small poorly differentiated cells with high atypia, and is frequently present together with urothelial carcinoma, squamous cell carcinoma and adenocarcinoma of the bladder.

Small cell carcinoma is an extremely aggressive tumour with early metastases to multiple sites including bone and brain, rarely affected in urothelial carcinoma (79-81).

In a phase II trial on neoadjuvant chemotherapy in patients with small cell carcinoma of the bladder, a strong association between advanced disease stage (≥T3b, N+ or M+) and brain metastases was observed, with an increased incidence up to 50% (82).

For this reason, it is mandatory an imaging evaluation of central nervous system at the preoperative diagnosis, even in asymptomatic patients.

Due to its aggressive behaviour, differential diagnosis is crucial and includes poorly differentiated high-grade urothelial carcinoma and bladder metastases of small cell carcinoma from other organs (especially from prostate).

Data on small cell carcinoma comes mostly from comparative studies based on SEER database and NCDB, and shows inferior OS when patients are treated with only RC compared to pure urothelial carcinoma (11,53,54,83).

Nevertheless, Kaushik et al. demonstrated no significant 5-year CSS differences compared to pure urothelial carcinoma when adjusted for stage and lymph node status (84).

Like its pulmonary counterpart, data from literature demonstrates the chemo-sensitivity of small cell carcinoma of the bladder when platinum- and etoposide-based chemotherapy is used, and three studies on neoadjuvant chemotherapy demonstrated a significant survival benefit (62,80,85).

Early neoadjuvant chemotherapy and extent radical surgery should be considered therefore the optimal treatment for this VH, even in non-muscle-invasive disease (78).

Adenocarcinoma

While secondary adenocarcinoma interesting the bladder by direct or distant spread from other organs (prostate, gastrointestinal tract, endometrium, cervix, lung) is the most frequent form, primary adenocarcinoma is a rare variant of bladder cancer, representing 2% of all bladder neoplasms.

It has to be distinguished from urachal adenocarcinoma that develops from the urachal remnant, which usually shows a less aggressive behaviour and a favourable prognosis and could be treated with bladder preserving techniques (86).

Bladder exstrophy and non-functioning neurogenic bladder are well established risk factors for adenocarcinoma of the bladder.

Primary adenocarcinoma exists in different morphologies including intestinal, papillary, signet ring cell, clear cell or mixed. It usually presents at advanced stages with local invasion or distant metastases.

Data from SEER database shows that 75% of adenocarcinomas were locally advanced at the time of surgery (87), however when stage- and grade-adjusted, cancer specific mortality after RC is similar to urothelial carcinoma (88).

Recent multivariable analysis of data from patients treated with RC from NCDB and SEER database confirmed comparable outcomes (53,54).

Owing the radio-sensitivity of adenocarcinoma histology, the role of radiotherapy was explored, but at present is still not well established. Although a previous report on the beneficial effect on 5-year DFS, CSS and lower local failure of adjuvant radiotherapy (89), more recent data from NCDB failed to confirm these results, showing worse outcomes for patients receiving radiotherapy alone or after RC (90).

Data on the role of perioperative chemotherapy in patients with adenocarcinoma is still scarce, with apparent no beneficial effects on OS (62,63).

RC with or without adjuvant radiotherapy remains therefore the standard of care for patients harbouring primary adenocarcinoma of the bladder.

Pure squamous cell carcinoma

Pure squamous cell carcinoma is the most common non-urothelial variant, accounting 5% of all bladder cancers in western countries and reaching 30% in Schistosoma haematobium endemic regions like Egypt and Sudan (91).

Risk factors for squamous cell carcinoma, other than schistosomiasis, are represented by causes of chronic inflammation of the bladder, including permanent/intermittent catheterization, urinary tract infections and bladder calculi.

It is microscopically characterized by abundant keratin production as pearl formations and intracytoplasmic granules.

Approximatively 70% of pure squamous cell carcinomas are muscle-invasive at initial diagnosis, with high rate of local recurrence (92).

Based on data from NCDB, OS in squamous cell carcinoma remains inferior compared to urothelial carcinoma, even when adjusted for grade and stage (83,93).

SEER database analysis showed that patients treated with RC have a significant superior CSS and OS compared to patients treated with only radiotherapy or to untreated patients (94), but pure squamous cell variant remains an independent predictor of inferior CSS and OS compared to pure urothelial carcinoma.

Moreover, bladder preserving techniques including trimodality therapy showed worse OS in patients with pure squamous cell carcinoma compared to those with pure urothelial histology (95).

From analyses based on NCDB patients, perioperative both neoadjuvant and adjuvant chemotherapy seem to confer no beneficial effects on OS in patients with pure squamous cell carcinoma. In patients underwent only neoadjuvant chemotherapy no downstaging effect was observed, with poorer OS (62,63,93,96).

The standard of care for squamous cell carcinoma remains therefore RC even for pT1 disease (97).


Molecular subtypes and VH

Four classification methods, University of North Carolina (UNC), MD Anderson Cancer Center (MDA), The Cancer Genome Atlas (TCGA) and Lund University, have identified at least five different molecular profiles for muscle-invasive bladder cancer: luminal, luminal-papillary/uroA, luminal-infiltrated/p53-like, basal-squamous, basal-neuronal (98,99).

Another molecular subtype has been recently identified as a basal mesenchymal-like/claudin-low type, different from either basal-squamous or neuronal (100).

Basal subtypes, both squamous and neuronal, seem to have a more aggressive behavior than luminal subtypes, with a tendency to early invasion and metastases. However, Seiler et al. demonstrated that basal-squamous is the most chemo-sensitive subtype, with a significative improvement in prognosis after neoadjuvant cisplatin-based chemotherapy (19). Neuronal tumours are only initially chemo-sensitive, especially to etoposide-platinum combined chemotherapy, but with short-term responses (101). Therefore, new therapeutic approaches are under study, in particular immune checkpoint inhibitors due to the high tumour mutational burdens of neuronal subtypes (98).

Claudin-law subtype has the worst outcome, with no beneficial effect from neoadjuvant chemotherapy (19).

Luminal-papillary subtype is associated with the best survival outcomes, regardless the low likelihood of response to platinum-based chemotherapy (19). This subtype is also described as an “immune desert” phenotype for its low expression of immune markers and nonimmune-infiltrated nature. For this reason, luminal-papillary tumours do not respond to immunotherapy (102). Nevertheless, the high rates of FGFR3 mutations may be a target for therapies based on FGFR inhibitors.

Instead, luminal-infiltrated is the most chemo-resistant but immunosensitive subtype (19,102).

Luminal subtype is clinically aggressive, but it seems to be sensitive to chemotherapy and immunotherapy (18).

Recent studies suggested that some HVs tend to have specific molecular subtypes (13,103), as reported in Table 1. Micropapillary, plasmacytoid/signet ring cell variants and glandular divergent differentiation express typically luminal and luminal-infiltrated biomarkers (104,105). Lymphoepithelioma-like, sarcomatoid and neuroendocrine variants and squamous divergent differentiation express instead basal molecular mutations.


Conclusions

Accurate pathological identification and report of VH is crucial for a precise risk stratification of patients with bladder cancer, in order to choose the optimal management and improve the oncological outcomes.

The presence of HVs is indicative of a more aggressive disease, often advanced at presentation. Nevertheless, when matched for stage and grade, the oncological outcomes are similar with pure urothelial carcinoma, except for pure squamous cell and signet ring cell carcinoma variants which have the worst prognosis.

RC remains the treatment of choice and should be considered even in non-muscle-invasive cases if some forms of VH are present, in particular micropapillary, pure squamous, plasmacytoid and sarcomatoid. Further studies are necessary to fully explore the role of early RC in non-muscle-invasive HVs. Neoadjuvant chemotherapy is recommended in chemo-sensitive variants, in particular for neuroendocrine tumour and lymphoepithelioma-like carcinoma. Adjuvant radiotherapy could be considered in pure adenocarcinoma.

Basal molecular subtype relates to a more aggressive disease, with poor survival outcomes but high response to neoadjuvant chemotherapy. Luminal subtype has instead a better prognosis but low response to chemotherapy. Recent evidences suggest that molecular composition could be related to the expression of specific HVs and new subtypes could be identified with further investigations.

Improvement in TUR procedures, dedicated genitourinary pathologists and emerging genomic techniques for molecular subtyping will lead to a better definition of each HV, along with related prognosis and response to treatments.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editor (Marco Moschini) for the series “Bladder Cancer” published in AME Medical Journal. The article has undergone external peer review.

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://amj.amegroups.com/article/view/10.21037/amj.2020.02.02/coif). The series “Bladder Cancer” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Rogers CG, Palapattu GS, Shariat SF, et al. Clinical outcomes following radical cystectomy for primary nontransitional cell carcinoma of the bladder compared to transitional cell carcinoma of the bladder. J Urol 2006;175:2048-53. [Crossref] [PubMed]
  2. Humphrey PA, Moch H, Cubilla AL, et al. The 2016 WHO classification of tumours of the urinary system and male genital organs-part B: prostate and bladder tumours. Eur Urol 2016;70:106-19. [Crossref] [PubMed]
  3. Chalasani V, Chin JL, Izawa JI. Histologic variants of urothelial bladder cancer and nonurothelial histology in bladder cancer. Can Urol Assoc J 2009;3:S193-8. [Crossref] [PubMed]
  4. Zamboni S, Foerster B, Abufaraj M, et al. Incidence and survival outcomes in patients with upper urinary tract urothelial carcinoma diagnosed with variant histology and treated with nephroureterectomy. BJU Int 2019;124:738-45. [Crossref] [PubMed]
  5. Linder BJ, Boorjian SA, Cheville JC, et al. The impact of histological reclassification during pathology re-review--evidence of a Will Rogers effect in bladder cancer? J Urol 2013;190:1692-6. [Crossref] [PubMed]
  6. Ukai R, Hashimoto K, Iwasa T, et al. Transurethral resection in one piece (TURBO) is an accurate tool for pathological staging of bladder tumor. Int J Urol 2010;17:708-14. [Crossref] [PubMed]
  7. Hansel DE, Amin MB, Comperat E, et al. A contemporary update on pathology standards for bladder cancer: transurethral resection and radical cystectomy specimens. Eur Urol 2013;63:321-32. [Crossref] [PubMed]
  8. Abufaraj M, Shariat SF, Foerster B, et al. Accuracy and prognostic value of variant histology and lymphovascular invasion at trans-urethral resection of bladder. World J Urol 2018;36:231-40. [Crossref] [PubMed]
  9. Abd El-Latif A, Watts KE, Elson P, et al. The sensitivity of initial transurethral resection or biopsy of bladder tumor(s) for detecting bladder cancer variants on radical cystectomy. J Urol 2013;189:1263-7. [Crossref] [PubMed]
  10. Cai T, Tiscione D, Verze P, et al. Concordance and clinical significance of uncommon variants of bladder urothelial carcinoma in trans-urethral resection and radical cystectomy specimens. Urology 2014;84:1141-6. [Crossref] [PubMed]
  11. Moschini M, Shariat SF, Freschi M, et al. Is transurethral resection alone enough for the diagnosis of histological variants? A single-center study. Urol Oncol 2017;35:528.e1-528.e5. [Crossref] [PubMed]
  12. Shah RB, Montgomery JS, Montie JE, et al. Variant (divergent) histologic differentiation in urothelial carcinoma is under-recognized in community practice: impact of mandatory central pathology review at a large referral hospital. Urol Oncol 2013;31:1650-5. [Crossref] [PubMed]
  13. Lopez-Beltran A, Henriques V, Montironi R, et al. Variants and new entities of bladder cancer. Histopathology 2019;74:77-96. [Crossref] [PubMed]
  14. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014;507:315-22. [Crossref] [PubMed]
  15. Choi W, Porten S, Kim S, et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 2014;25:152-65. [Crossref] [PubMed]
  16. Damrauer JS, Hoadley KA, Chism DD, et al. Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology. Proc Natl Acad Sci U S A 2014;111:3110-5. [Crossref] [PubMed]
  17. Sjödahl G, Lauss M, Lövgren K, et al. A molecular taxonomy for urothelial carcinoma. Clin Cancer Res 2012;18:3377-86. [Crossref] [PubMed]
  18. Robertson AG, Kim J, Al-Ahmadie H, et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder cancer. Cell 2017;171:540-556.e25. [Crossref] [PubMed]
  19. Seiler R, Ashab HAD, Erho N, et al. Impact of Molecular Subtypes in Muscle-invasive Bladder Cancer on Predicting Response and Survival after Neoadjuvant Chemotherapy. Eur Urol 2017;72:544-54. [Crossref] [PubMed]
  20. Babjuk M, Burger M, Compérat EM, et al. European Association of Urology Guidelines on Non-muscle-invasive Bladder Cancer (TaT1 and Carcinoma In Situ) - 2019 Update. Eur Urol 2019;76:639-57. [Crossref] [PubMed]
  21. Chang SS, Boorjian SA, Chou R, et al. Diagnosis and treatment of non-muscle invasive bladder cancer: AUA/SUO guideline. J Urol 2016;196:1021-9. [Crossref] [PubMed]
  22. Mally AD, Tin AL, Lee JK, et al. Clinical outcomes of patients with T1 nested variant of urothelial carcinoma compared to pure urothelial carcinoma of the bladder. Clin Genitourin Cancer 2017; [Epub ahead of print]. [PubMed]
  23. Beltran AL, Cheng L, Montironi R, et al. Clinicopathological characteristics and outcome of nested carcinoma of the urinary bladder. Virchows Arch 2014;465:199-205. [Crossref] [PubMed]
  24. Gofrit ON, Yutkin V, Shapiro A, et al. The Response of Variant Histology Bladder Cancer to Intravesical Immunotherapy Compared to Conventional Cancer. Front Oncol 2016;6:43. [Crossref] [PubMed]
  25. Paz A, Rath-Wolfson L, Lask D, et al. The clinical and histological features of transitional cell carcinoma of the bladder with microcysts: analysis of 12 cases. Br J Urol 1997;79:722-5. [Crossref] [PubMed]
  26. Young RH, Zukerberg LR. Microcystic transitional cell carcinomas of the urinary bladder. A report of four cases. Am J Clin Pathol 1991;96:635-9. [Crossref] [PubMed]
  27. Lopez Beltran A, Montironi R, Cheng L. Microcystic urothelial carcinoma: morphology, immunohistochemistry and clinical behaviour. Histopathology 2014;64:872-9. [Crossref] [PubMed]
  28. Wang JK, Boorjian SA, Cheville JC, et al. Outcomes following radical cystectomy for micropapillary bladder cancer versus pure urothelial carcinoma: a matched cohort analysis. World J Urol 2012;30:801-6. [Crossref] [PubMed]
  29. Fairey AS, Daneshmand S, Wang L, et al. Impact of micropapillary urothelial carcinoma variant histology on survival after radical cystectomy. Urol Oncol 2014;32:110-6. [Crossref] [PubMed]
  30. Mitra AP, Fairey AS, Skinner EC, et al. Implications of micropapillary urothelial carcinoma variant on prognosis following radical cystectomy: a multi-institutional investigation. Urol Oncol 2019;37:48-56. [Crossref] [PubMed]
  31. Ghoneim IA, Miocinovic R, Stephenson AJ, et al. Neoadjuvant systemic therapy or early cystectomy? Single-center analysis of outcomes after therapy for patients with clinically localized micropapillary urothelial carcinoma of the bladder. Urology 2011;77:867-70. [Crossref] [PubMed]
  32. Willis DL, Fernandez MI, Dickstein RJ, et al. Clinical outcomes of cT1 micropapillary bladder cancer. J Urol 2015;193:1129. [Crossref] [PubMed]
  33. Kamat AM, Dinney CPN, Gee JR, et al. Micropapillary bladder cancer: a review of the University of Texas M. D. Anderson Cancer Center experience with 100 consecutive patients. Cancer 2007;110:62-7. [Crossref] [PubMed]
  34. Spaliviero M, Dalbagni G, Bochner BH, et al. Clinical outcome of patients with T1 micropapillary urothelial carcinoma of the bladder. J Urol 2014;192:702-7. [Crossref] [PubMed]
  35. Sui W, Matulay JT, James MB, et al. Micropapillary Bladder Cancer: Insights from the National Cancer Database. Bladder Cancer 2016;2:415-23. [Crossref] [PubMed]
  36. Monn MF, Kaimakliotis HZ, Pedrosa JA, et al. Contemporary bladder cancer: variant histology may be a significant driver of disease. Urol Oncol 2015;33:18.e15-18.e20. [Crossref] [PubMed]
  37. Abufaraj M, Foerster B, Schernhammer E, et al. Micropapillary Urothelial Carcinoma of the Bladder: A Systematic Review and Meta-analysis of Disease Characteristics and Treatment Outcomes. Eur Urol 2019;75:649-58. [Crossref] [PubMed]
  38. Meeks JJ, Taylor JM, Matsushita K, et al. Pathological response to neoadjuvant chemotherapy for muscle-invasive micropapillary bladder cancer. BJU Int 2013;111:E325-30. [Crossref] [PubMed]
  39. Fernández MI, Williams SB, Willis DI, et al. Clinical risk stratification in patients with surgically resectable micropapillary bladder cancer. BJU Int 2017;119:684-91. [Crossref] [PubMed]
  40. Masson-Lecomte A, Xylinas E, Bouquot M, et al. Oncological outcomes of advanced muscle-invasive bladder cancer with a micropapillary variant after radical cystectomy and adjuvant platinum-based chemotherapy. World J Urol 2015;33:1087-93. [Crossref] [PubMed]
  41. Williamson SR, Zhang S, Lopez-Beltran A, et al. Lymphoepithelioma-like carcinoma of the urinary bladder: clinicopathologic, immunohistochemical, and molecular features. Am J Surg Pathol 2011;35:474-83. [Crossref] [PubMed]
  42. Lopez-Beltrán A, Luque RJ, Vicioso L, et al. Lymphoepithelioma-like carcinoma of the urinary bladder: a clinicopathologic study of 13 cases. Virchows Arch 2001;438:552-7. [Crossref] [PubMed]
  43. Yang AW, Pooli A, Lele SM, et al. Lymphoepithelioma-like, a variant of urothelial carcinoma of the urinary bladder: a case report and systematic review for optimal treatment modality for disease-free survival. BMC Urol 2017;17:34. [Crossref] [PubMed]
  44. Sood S, Paner GP. Plasmacytoid Urothelial Carcinoma: An Unusual Variant That Warrants Aggressive Management and Critical Distinction on Transurethral Resections. Arch Pathol Lab Med 2019;143:1562-67. [Crossref] [PubMed]
  45. Lopez-Beltran A, Requena MJ, Montironi R, et al. Plasmacytoid urothelial carcinoma of the bladder. Hum Pathol 2009;40:1023-8. [Crossref] [PubMed]
  46. Cockerill PA, Cheville JC, Boorjian SA, et al. Outcomes Following Radical Cystectomy for Plasmacytoid Urothelial Carcinoma: Defining the need for improved local cancer control. Urology 2017;102:143-7. [Crossref] [PubMed]
  47. Dayyani F, Czerniak BA, Sircar K, et al. Plasmacytoid urothelial carcinoma, a chemosensitive cancer with poor prognosis, and peritoneal carcinomatosis. J Urol 2013;189:1656-61. [Crossref] [PubMed]
  48. Al-Ahmadie HA, Iyer G, Lee BH, et al. Frequent somatic CDH1 loss-of-function mutations in plasmacytoid variant bladder cancer. Nat Genet 2016;48:356-8. [Crossref] [PubMed]
  49. Simon CT, Skala SL, Killen PD, et al. Plasmacytoid urothelial carcinoma: a rapid autopsy case report with unique clinicopathologic and genomic profile. Diagn Pathol 2019;14:113. [Crossref] [PubMed]
  50. Li Q, Assel M, Benfante NE, et al. The Impact of Plasmacytoid Variant Histology on the Survival of Patients with Urothelial Carcinoma of Bladder after Radical Cystectomy. Eur Urol Focus 2019;5:104-8. [Crossref] [PubMed]
  51. Ericson KJ, Thomas L, Lee BH. Plasmacytoid variant urothelial carcinoma: clinicopathologic outcomes and experience with neoadjuvant chemotherapy. J Clin Oncol 2019;37:483. [Crossref]
  52. Diamantopoulos LN, Khaki AR, Vakar-Lopez F, et al. Patient characteristics, treatment patterns, outcomes and prognostic factors in plasmacytoid urothelial carcinoma. J Clin Oncol 2019;37:e16007. [Crossref]
  53. Patel SG, Weiner A, Keegan K, et al. Oncologic outcomes in patients with nonurothelial bladder cancer. Indian J Urol 2018;34:39-44. [Crossref] [PubMed]
  54. Jue JS, Koru-Sengul T, Moore KJ, et al. Sociodemographic and survival disparities for histologic variants of bladder cancer. Can J Urol 2018;25:9179-85. [PubMed]
  55. Wu SG, Chen XT, Zhang WW, et al. Survival in signet ring cell carcinoma varies based on primary tumor location: a Surveillance, Epidemiology, and End Results database analysis. Expert Rev Gastroenterol Hepatol 2018;12:209-14. [Crossref] [PubMed]
  56. Wang J, Wang FW. Clinical characteristics and outcomes of patients with primary signet-ring cell carcinoma of the urinary bladder. Urol Int 2011;86:453-60. [Crossref] [PubMed]
  57. Lopez-Beltran A, Pacelli A, Rothenberg HJ, et al. Carcinosarcoma and sarcomatoid carcinoma of the bladder: clinicopathological study of 41 cases. J Urol 1998;159:1497-503. [Crossref] [PubMed]
  58. Sanfrancesco J, McKenney JK, Leivo MZ, et al. Sarcomatoid Urothelial Carcinoma of the Bladder: Analysis of 28 Cases with Emphasis on Clinicopathologic Features and Markers of Epithelial-to-Mesenchymal Transition. Arch Pathol Lab Med 2016;140:543-51. [Crossref] [PubMed]
  59. Cheng L, Zhang S, Alexander R, et al. Sarcomatoid carcinoma of the urinary bladder: the final common pathway of urothelial carcinoma dedifferentiation. Am J Surg Pathol 2011;35:e34-46. [Crossref] [PubMed]
  60. Wright JL, Black PC, Brown GA, et al. Differences in survival among patients with sarcomatoid carcinoma, carcinosarcoma and urothelial carcinoma of the bladder. J Urol 2007;178:2302-6. [Crossref] [PubMed]
  61. Sui W, Matulay JT, Onyeji IC, et al. Contemporary treatment patterns and outcomes of sarcomatoid bladder cancer. World J Urol 2017;35:1055-61. [Crossref] [PubMed]
  62. Vetterlein MW, Wankowicz SAM, Seisen T. Neoadjuvant chemotherapy prior to radical cystectomy for muscle-invasive bladder cancer with variant histology. Cancer 2017;123:4346-55. [Crossref] [PubMed]
  63. Berg S, D’Andrea D, Vetterlein MW. Impact of adjuvant chemotherapy in patients with adverse features and variant histology at radical cystectomy for muscle-invasive carcinoma of the bladder: Does histologic subtype matter? Cancer 2019;125:1449-58. [Crossref] [PubMed]
  64. Lopez-Beltran A. Further commentary: tumours of the urinary system. In: Eble JN, Sauter G, Epstein JI, et al. editors. Pathology and genetics of tumours of the urinary system and male genital organs. Lyon, France: IARC Press, 2004:90-134.
  65. Mai KT, Bateman J, Djordjevic B, et al. Clear Cell Urothelial Carcinoma: A Study of 10 Cases and Meta-analysis of the Entity. Evidence of Mesonephric Differentiation. Int J Surg Pathol 2017;25:18-25. [Crossref] [PubMed]
  66. Kumar L, Narwal A, Kumar M, et al. Primary clear-cell urothelial carcinoma of urinary bladder: a not-so-clear entity with review of literature. BMJ Case Rep 2019; [Crossref] [PubMed]
  67. Mihai I, Taban S, Cumpanas A, et al. Clear cell urothelial carcinoma of the urinary bladder - a rare pathological entity. A case report and a systematic review of the literature. Bosn J Basic Med Sci 2019;19:400-3. [PubMed]
  68. Mitra AP, Bartsch CC, Bartsch G Jr, et al. Does presence of squamous and glandular differentiation in urothelial carcinoma of the bladder at cystectomy portend poor prognosis? An intensive case-control analysis. Urol Oncol 2014;32:117-27. [Crossref] [PubMed]
  69. Kim SP, Frank I, Cheville JC, et al. The impact of squamous and glandular differentiation on survival after radical cystectomy for urothelial carcinoma. J Urol 2012;188:405-9. [Crossref] [PubMed]
  70. Scosyrev E, Ely BW, Messing EM, et al. Do mixed histological features affect survival benefit from neoadjuvant platinum-based combination chemotherapy in patients with locally advanced bladder cancer? A secondary analysis of Southwest Oncology Group-Directed Intergroup Study (S8710). BJU Int 2011;108:693-9. [Crossref] [PubMed]
  71. Erdemir F, Tunc M, Ozcan F, et al. The effect of squamous and/or glandular differentiation on recurrence, progression and survival in urothelial carcinoma of bladder. Int Urol Nephrol 2007;39:803-7. [Crossref] [PubMed]
  72. Antunes AA, Nesrallah LJ, Dall’Oglio MF, et al. The role of squamous differentiation in patients with transitional cell carcinoma of the bladder treated with radical cystectomy. Int Braz J Urol 2007;33:339-45. [Crossref] [PubMed]
  73. Liu Y, Bui MM, Xu B. Urothelial Carcinoma With Squamous Differentiation Is Associated With High Tumor Stage and Pelvic Lymph-Node Metastasis. Cancer Control 2017;24:78-82. [Crossref] [PubMed]
  74. Zargar-Shoshtari K, Sverrisson EF, Sharma P, et al. Clinical Outcomes After Neoadjuvant Chemotherapy and Radical Cystectomy in the Presence of Urothelial Carcinoma of the Bladder With Squamous or Glandular Differentiation. Clin Genitourin Cancer 2016;14:82-8. [Crossref] [PubMed]
  75. Minato A, Fujimoto N, Kubo T. Squamous Differentiation Predicts Poor Response to Cisplatin-Based Chemotherapy and Unfavorable Prognosis in Urothelial Carcinoma of the Urinary Bladder. Clin Genitourin Cancer 2017;15:e1063-7. [Crossref] [PubMed]
  76. Krasnow RE, Drumm M, Roberts HJ, et al. Clinical Outcomes of Patients with Histologic Variants of Urothelial Cancer Treated with Trimodality Bladder-sparing Therapy. Eur Urol 2017;72:54-60. [Crossref] [PubMed]
  77. Miller JS, Epstein JI. Noninvasive urothelial carcinoma of the bladder with glandular differentiation: report of 24 cases. Am J Surg Pathol 2009;33:1241-8. [Crossref] [PubMed]
  78. Baumeister P, Zamboni S, Mattei A, et al. Histological variants in non-muscle invasive bladder cancer. Transl Androl Urol 2019;8:34-8. [Crossref] [PubMed]
  79. Mukesh M, Cook N, Hollingdale AE, et al. Small cell carcinoma of the urinary bladder: a 15-year retrospective review of treatment and survival in the Anglian Cancer Network. BJU Int 2009;103:747-52. [Crossref] [PubMed]
  80. Wang G, Xiao L, Zhang M, et al. Small cell carcinoma of the urinary bladder: a clinicopathological and immunohistochemical analysis of 81 cases. Hum Pathol 2018;79:57-65. [Crossref] [PubMed]
  81. Priemer DS, Wang M, Zhang S, et al. Small-cell Carcinomas of the Urinary Bladder and Prostate: TERT Promoter Mutations Status Differentiates Sites of Malignancy and Provides Evidence of Common Clonality Between Small-cell Carcinoma of the Urinary Bladder and Urothelial Carcinoma. Eur Urol Focus 2018;4:880-8. [Crossref] [PubMed]
  82. Siefker-Radtke AO, Kamat AM, Grossman HB. Phase II clinical trial of neoadjuvant alternating doublet chemotherapy with ifosfamide/doxorubicin and etoposide/cisplatin in small-cell urothelial cancer. J Clin Oncol 2009;27:2592-7. [Crossref] [PubMed]
  83. Royce TJ, Lin CC, Gray PJ, et al. Clinical characteristics and outcomes of nonurothelial cell carcinoma of the bladder: Results from the National Cancer data Base. Urol Oncol 2018;36:78.e1-78.e12. [Crossref] [PubMed]
  84. Kaushik D, Frank I, Boorjian SA, et al. Long-term results of radical cystectomy and role of adjuvant chemotherapy for small cell carcinoma of the bladder: small cell carcinoma of the bladder. Int J Urol 2015;22:549-54. [Crossref] [PubMed]
  85. Lynch SP, Shen Y, Kamat A, et al. Neoadjuvant chemotherapy in small cell urothelial cancer improves pathologic downstaging and long-term outcomes: results from a retrospective study at the MD Anderson Cancer Center. Eur Urol 2013;64:307-13. [Crossref] [PubMed]
  86. Natale C, Leinwand G, Chiang J, et al. Reviewing the Demographic, Prognostic and Treatment Factor of Primary Adenocarcinoma of the Bladder: A SEER Population-based Study. Clin Genitourin Cancer 2019;17:380-8. [Crossref] [PubMed]
  87. Zaffuto E, Gazdovich S, Leyh-Bannurah S, et al. Contemporary rates of pathological features and mortality for adenocarcinoma of the urinary bladder in the USA. Int J Urol 2017;24:117-23. [Crossref] [PubMed]
  88. Lughezzani G, Sun M, Jeldres C, et al. Adenocarcinoma versus urothelial carcinoma of the urinary bladder: comparison between pathologic stage at radical cystectomy and cancer-specific mortality. Urology 2010;75:376-81. [Crossref] [PubMed]
  89. Zaghloul MS, Nouh A, Nazmy M, et al. Long-term results of primary adenocarcinoma of the urinary bladder: a report on 192 patients. Urol Oncol 2006;24:13-20. [Crossref] [PubMed]
  90. Davaro F, Schaefer J, May A, et al. Invasive non-urachal adenocarcinoma of the bladder: analysis of the National Cancer Database. World J Urol 2019;37:497-505. [Crossref] [PubMed]
  91. Felix AS, Soliman AS, Khaled H, et al. The changing patterns of bladder cancer in Egypt over the past 26 years. Cancer Causes Control 2008;19:421-9. [Crossref] [PubMed]
  92. Kassouf W, Speiss PE, Sieker-Radtke A, et al. Outcome and patterns of recurrence of nonbilharzial pure squamous cell carcinoma of the bladder: a contemporary review of the University of Texas M D Anderson Cancer Center experience. Cancer 2007;110:764-9. [Crossref] [PubMed]
  93. Matulay JT, Woldu SL, Lim A, et al. The impact of squamous histology on survival in patients with muscle-invasive bladder cancer. Urol Oncol 2019;37:353.e17-24. [Crossref] [PubMed]
  94. Abdel-Rahman O. Squamous cell carcinoma of the bladder: a SEER database analysis. Clin Genitourin Cancer 2017;15:e463-8. [Crossref] [PubMed]
  95. Fischer-Valuck BW, Michalski JM, Contreras JA, et al. A propensity analysis comparing definitive chemo-radiotherapy for muscle-invasive squamous cell carcinoma of the bladder vs. urothelial carcinoma of the bladder using the National Cancer Database. Clin Transl Radiat Oncol 2018;15:38-41. [Crossref] [PubMed]
  96. Dotson A, May A, Davaro F, et al. Squamous cell carcinoma of the bladder: poor response to neoadjuvant chemotherapy. Int J Clin Oncol 2019;24:706-11. [Crossref] [PubMed]
  97. Scosyrev E, Yao J, Messing E. Urothelial carcinoma versus squamous cell carcinoma of bladder: is survival different with stage adjustment? Urology 2009;73:822-7. [Crossref] [PubMed]
  98. McConkey DJ, Choi W. Molecular Subtypes of Bladder Cancer. Curr Oncol Rep 2018;20:77. [Crossref] [PubMed]
  99. Lerner SP, McConkey DJ, Hoadley KA, et al. Bladder Cancer Molecular Taxonomy: Summary from a Consensus Meeting. Bladder Cancer 2016;2:37-47. [Crossref] [PubMed]
  100. Kardos J, Chai S, Mose LE, et al. Claudin-low bladder tumors are immune infiltrated and actively immune suppressed. JCI Insight 2016;1:e85902. [Crossref] [PubMed]
  101. Geynisman DM, Handorf E, Wong YN, et al. Advanced small cell carcinoma of the bladder: clinical characteristics, treatment patterns and outcome in 960 patients and comparison with urothelial carcinoma. Cancer Med 2016;5:192-9. [Crossref] [PubMed]
  102. Rosenberg JE, Hoffman-Censits J, Powels T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909-20. [Crossref] [PubMed]
  103. Warrick JI, Sjödahl G, Kaag M, et al. Intratumoral Heterogeneity of Bladder Cancer by Molecular Subtypes and Histologic Variants. Eur Urol 2019;75:18-22. [Crossref] [PubMed]
  104. Dadhania V, Zhang M, Zhang L, et al. Meta-Analysis of the Luminal and Basal Subtypes of Bladder Cancer and the Identification of Signature Immunohistochemical Markers for Clinical Use. EBioMedicine 2016;12:105-17. [Crossref] [PubMed]
  105. Warrick JI, Kaag M, Raman JD, et al. FOXA1 and CK14 as markers of luminal and basal subtypes in histologic variants of bladder cancer and their associated conventional urothelial carcinoma. Virchows Arch 2017;471:337-45. [Crossref] [PubMed]
doi: 10.21037/amj.2020.02.02
Cite this article as: Processali T, Diminutto A, Cerruto MA, Antonelli A. The impact of histological variants on bladder cancer outcomes. AME Med J 2020;5:4.

Download Citation