Next Article in Journal
Selectively Modified Lactose and N-Acetyllactosamine Analogs at Three Key Positions to Afford Effective Galectin-3 Ligands
Next Article in Special Issue
Crosstalk between Mesenchymal Stem Cells and Cancer Stem Cells Reveals a Novel Stemness-Related Signature to Predict Prognosis and Immunotherapy Responses for Bladder Cancer Patients
Previous Article in Journal
Dependence on MUC1-C in Progression of Neuroendocrine Prostate Cancer
Previous Article in Special Issue
Ex Vivo Fluorescence Confocal Microscopy (FCM) Ensures Representative Tissue in Prostate Cancer Biobanking: A Feasibility Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

HER2 Expression in Bladder Cancer: A Focused View on Its Diagnostic, Prognostic, and Predictive Role

1
Pathology Unit, Policlinico Riuniti, University of Foggia, 71122 Foggia, Italy
2
Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
3
Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
4
Pathology Unit, Azienda Ospedaliera Santa Maria di Terni, University of Perugia, 05100 Terni, Italy
5
Urology Unit, Azienda Ospedaliero-Universitaria Ospedali Riuniti Di Ancona, Università Politecnica Delle Marche, 60126 Ancona, Italy
6
Department of Urology and Renal Transplantation, Policlinico Riuniti, University of Foggia, 71122 Foggia, Italy
7
Department of Urology, Bonomo Teaching Hospital, 76123 Andria, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(4), 3720; https://doi.org/10.3390/ijms24043720
Submission received: 21 January 2023 / Revised: 4 February 2023 / Accepted: 7 February 2023 / Published: 13 February 2023
(This article belongs to the Special Issue Urogenital Tumors: From Molecular Basis to Therapy)

Abstract

:
Bladder cancer (BC) is a heterogeneous disease from a molecular, morphological, and clinical standpoint. HER2 is a known oncogene involved in bladder carcinogenesis. Assessing HER2 overexpression as a result of its molecular changes in a routine pathology practice using immunohistochemistry might be a useful adjunct in several scenarios, namely (1) to correctly identify flat urothelial lesions and inverted urothelial lesions in the diagnostic setting; (2) to provide prognostic hints in both non-muscle invasive (NMI) and muscle invasive (MI) tumors, thus supplementing risk stratification tools, especially when evaluating higher-risk tumors such as those with variant morphology; (3) to improve antibody panels as a surrogate marker of BC molecular subtyping. Furthermore, the potential of HER2 as a therapeutic target has been only partly explored so far, in light of the ongoing development of novel target therapies.

1. Introduction

Bladder cancer (BC) is the seventh most prevalent malignancy and the thirteenth cause of cancer death worldwide, accounting for 1,720,625 cases overall and 573,278 newly diagnosed cases each year [1]. Approximately a quarter of BC patients present with advanced (muscle-invasive or metastatic) disease (MIBC), whereas the remaining 75% are diagnosed with non-muscle invasive BC (NMIBC), with both having different clinical behavior and therapeutic strategies [2,3]. TURBT/radiotherapy/chemotherapy is not comparable to radical cystectomy, though responses to treatment may be variable and sometimes barely predictable. Novel target treatment options have been introduced in the advanced setting of the disease, including anti-Fibroblast Growth Factor Receptor (FGFR) and immune checkpoint inhibitors (ICIs), as well as antibody–drug conjugates (ADCs) (see below) [4].
On the other hand, most NMIBC patients undergo favorable outcomes upon early diagnosis; nevertheless, these tumors carry both high recurrence rates and a significant risk of progression to muscle-invasive disease, especially in high-risk cases, with inherent increased lifetime costs per patient [5]. Such high-risk tumors may be cured with transurethral resection along with intravesical instillations of chemotherapeutic or immunotherapeutic agents (namely, mitomycin C and Bacillus Calmette–Guérin (BCG)), or early cystectomy. Yet, the latter may carry postoperative life-changing disadvantages, whereas BCG intravesical therapy may be ineffective and yield a high rate of adverse effects, with a varying potential to disease relapse and progression [2,3].
The underlying biology of such clinical heterogeneity is still a matter of study. As far as we know, BC is a heterogeneous disease from a molecular standpoint as well, resulting from a complex multi-step carcinogenesis that includes several changes involving genes and molecular pathways with specific functions in tumor development and progression. In an attempt to catch this biological variability, several efforts have been made over the last decade to develop of a molecular classification encompassing a discrete category of BC harboring different clinical and prognostic features, matching their DNA and RNA profiles [6,7,8,9]. In routine practice, current risk assessment models in NMIBC, as well as predictive/prognostic systems in MIBC, are based on clinical and histopathological features [10]. During the last few years, attempts have been made to improve the management of BC patients by introducing more effective risk stratification tools, including molecular markers; as a result of this, a more comprehensive analysis of genomic, epigenetic, and transcriptomic features have been accomplished, providing novel insights into bladder carcinogenesis [9,11]. The ultimate goal is to find biomarkers with a prognostic/predictive role and which, hopefully, may act as potential therapeutic targets as well; they should be based upon a clear rationale, detectable both in vitro and in vivo in distinct specimens (urine, blood, tissue) using selected technologies, and be easily assessed and quantified [12]. Finally, they should demonstrate clinical soundness and utility upon assay standardization, threshold establishment, and external validation [13].
Human Epidermal growth factor Receptor 2 (HER2/ERBB2) is a member of a family of epithelial growth factor receptors, along with HER1/EGFR, HER3, and HER4 [14], which are transmembrane receptor tyrosine kinases involved in cell proliferation, survival, and mobility via the downstream activation of different intracellular signaling pathways such as the mitogen-activated protein kinase (MAPK) and the phosphoinositide 3-kinase (PI3K/Akt) pathways [14]. The HER2 proto-oncogene, located at the long arm of chromosome 17 (17q12), codes for the HER2 protein, and its mutation and amplification mostly result in HER2 overexpression [14]. Higher HER2 levels have well-known prognostic and predictive roles in breast and gastroesophageal cancers, where immunohistochemistry (IHC) is routinely used to assess HER2 status at the protein level [15]. In this setting, patients are stratified according to the presence/absence, intensity, and completeness of the membrane staining in the tumor cells, whereas in situ hybridization methods (namely, chromogenic in situ hybridization (CISH) and fluorescence in situ hybridization (FISH)), are regarded as second-level techniques to be performed in case of equivocal results [16,17]. HER2-positivity is currently defined as intense protein overexpression in >10% of the tumor cells by IHC, or HER2 gene amplification by FISH in breast and gastroesophageal cancer [18]; these classification systems have been applied by most but not all studies, resulting in poor validation.
Using different techniques may yield discordant results, with IHC positivity rates being higher than ISH ones across different studies; further issues include assay standardization and the definition of distinct cut-offs based on the sensitivity level of each method [19]. Though widely observed, especially in NMIBCs, gender differences have not been described in this setting [20]. However, a high concordance between HER2 protein overexpression and gene amplification in BC has been described in studies based on large data sets [18,21,22]. Conversely, other authors have failed to report an optimal correlation between HER2 gene amplification and protein overexpression, possibly due to epigenetic factors and technique-related factors [16,17]. Obviously, literature findings should be weighted in relation to the geographical, clinical, and pathological characteristics, as well as the number of study cohorts.
Over the years, HER2 expression has been evaluated in BC as well, among other potential biomarkers, with a view to implementing its use in clinical practice [23,24]. As a potential therapeutic target, HER2 has been assessed in clinical trials using anti-HER2 monoclonal antibodies and tyrosine-kinase inhibitors, either in monotherapy or combined with conventional chemotherapy, as second-line treatments in patients with advanced or metastatic BC; however, such phase II trials did not yield acceptable results, with low overall response rates [16]. More recently, ADCs targeting HER2 (among other molecules), such as trastuzumab emtansine, trastuzumab duocarmazine, and disitamab vedotin, showed promising results in multi-tumor basket clinical trials; a further such agent with an enhanced pharmacokinetic profile, namely trastuzumab deruxtecan, is currently under evaluation [16].
Herein, we aim to highlight the updated data on the diagnostic, prognostic, and predictive role of HER2 in BC, and provide a critical discussion on current and emerging issues in the field.

2. HER2 Expression in Flat and Inverted Urothelial Lesions

Flat urothelial lesions with atypia encompass a spectrum of pathological entities ranging from non-neoplastic to frankly malignant, including reactive atypia, urothelial dysplasia, and carcinoma in situ (CIS) [25]. The differential diagnosis among such lesions in the appropriate clinical framework mostly relies on their appearance in light microscopy [26]; however, even in the context of proper clinical information, the assessment of morphological parameters alone may be not sufficient to distinguish among these lesions, especially between reactive atypia and CIS [25]. Therefore, in the last few decades, selected immunohistochemical markers have been progressively studied and introduced in the routine pathology practice, including HER2.
In normal urothelium, membranous HER2 expression ranges from lacking to present in the superficial cell layer only and, occasionally, by intermediate cells, with stronger staining on the basal and lateral side [27,28,29,30,31]. This is in keeping with the theory that the orderly maturation of urothelial cells is supported by the coordinated up-regulation and down-regulation of the class I tyrosine kinase receptors, namely HER2, HER3, HER4, and EGFR, respectively [27]. Normal urothelium showing such HER2 expression patterns has been shown to be diploid in FISH analysis [29], accordingly. A few samples of normal or reactive urothelium displayed full-thickness HER2 immunoreactivity in one study, and the staining was described as weak and focal [30].
Wagner et al. [32] described two different HER2 expression patterns in normal, dysplastic, and neoplastic flat urothelium. The diffuse pattern (namely, a focal or diffuse HER2 expression in deeper cell layers) was observed with increasing frequency in mild-to-moderate dysplasia (8/22, 36%) and CIS (12/22, 55%), thus suggesting that HER2 may play a role in malignant transformation. HER2 gene amplification was detected in 33% (2/6) of biopsy samples showing diffuse overexpression of the protein. Conversely, a superficial pattern (namely, HER2 expression in superficial urothelial cells only) was reported more often in patients with mild dysplasia (26/58, 45%) compared to moderate dysplasia (10/58, 17%), normal urothelium in absence of previous BC neoplasms (5/58, 9%), and CIS (3/58, 5%) (Figure 1).
In keeping with these results, a strong, full-thickness HER2 expression has been described in the majority of CIS cases by several authors [25,28,29,30,31,33]. Two investigators independently evaluated HER2 expression by separately assessing the lower/basal and upper/luminal halves of the urothelium in the study by Gunia et al. [33], yielding significantly different scores in both cases between CIS and non-CIS entities (namely, dysplasia and reactive atypia). Accordingly, Barth et al. analyzed a large panel of luminal and basal markers in a series of 156 CIS cases [31], most of them being characterized by the expression of luminal markers, including HER2; such findings were confirmed in a more recent study from the same group [22]. Interestingly, HER2 expression showed higher specificity in identifying CIS across studies, as compared to other markers (namely, CK20, CK5/6, P53, CD138) [30,33].
An additional advantage in using HER2 as a diagnostic tool is the fact that its positive staining in CIS cases involves the deeper urothelial layers; therefore, HER2 may be applied even to tissue samples whose urothelial lining is not intact, which is a quite common occurrence in TUR specimens [26].
A further point is the higher presence of HER2 immunostaining in morphologically normal urothelium, either from patients with positive as compared to negative BC history (64% versus 33%) in one study [32], or adjacent to urothelial neoplasms [31], thus suggesting that early molecular changes may precede the development of morphologically detectable features of malignancy. In keeping with this, a normal urothelium adjacent to CIS showed weak positivity compared to the moderate to strong expression of HER2 in the series by Barth et al. [31].
HER2 enrichment at the protein level is mostly attributable to polysomy 17 rather than gene amplification in both FISH and SISH analysis [29,31,33,34]. The results from a recent next-generation sequencing (NGS) study on CIS cases reported a rate of missense mutations in the extracellular domain of HER2 as high as 16%, encompassing the pathogenic activating S310F mutation, which is a common HER2 alteration in BC [34].
Beyond its diagnostic role in CIS, HER2 alone or in combination with other agents may be the leading actor of intravesical targeted therapies, including ADCs [33,34], in order to supply an alternative bladder-sparing approach in the subset of BCG-refractory CIS patients otherwise amenable to cystectomy [31,34].
A subset of urothelial lesions, ranging from benign to frankly malignant, displays a partial to diffuse inverted/endophytic pattern of growth; in this scenario, distinguishing lesions with different outcomes may be challenging due to their morphological similarities, hence the need to find diagnostic biomarkers. Since moderate to strong HER2 overexpression has been described in inverted UCs as compared to their benign counterparts both in the bladder and upper urinary tract [35], this might be a useful adjunct in the diagnosis of malignant urothelial lesions with an inverted/endophytic pattern.

3. Prognostic Role of HER2 in NMIBC

BC ranks third among all cancers in terms of HER2 overexpression, carrying as much as 6–17% of gene mutations and/or amplification in tissue samples [36].
HER2 protein overexpression has been reported as a marker of poor prognosis in BC. Despite disagreeing results, the finding that the nodal metastases consistently showed HER2 overexpression as compared to the respective primary strongly supports this hypothesis [37,38], although it might be related to tumor heterogeneity. Accordingly, a higher HER2 status was described by several authors as significantly associated with a higher stage and grade and a poor disease-specific survival, mainly in the muscle-invasive and metastatic setting [39,40,41,42,43,44,45]. Furthermore, the higher rates of HER2 positivity in advanced cases may suggest its use as a marker of circulating tumor cells assessed through liquid biopsy, thus overcoming the need to take further tissue biopsies in such patients [46].
The prognostic role of HER2 in NMIBC is more debatable. In a next-generation sequencing analysis of a cohort of 105 NMIBCs of varying stage (pTis, pTa, pT1) and grade (high and low), higher-stage and grade tumors were consistently enriched with HER2 mutations [47]. Increased HER2 expression has been reported in patients with relapsed NMIBC after adjuvant intravesical therapy [48]. HER2 overexpression has been significantly associated with shorter progression-free survival (PFS) [49,50] and especially recurrence-free survival (RFS) [48,51,52], or both [53], whereas earlier studies failed to confirm these findings [54,55,56]. Conversely, HER2-positive tumors from a cohort of 60 NMIBCs had favorable outcomes in terms of lower odds ratios of grade progression at any subsequent biopsy diagnosis [57].
In two further studies, our group reported HER2 overexpression as an independent predictor of RFS and PFS, either alone [58] or in combination with microsatellite instability markers (MLH1 and MSH2) [59], the latter being potential therapeutic targets as well. In our studies, the proposed markers even outperformed BCG treatment in predicting PFS.
Risk stratification tools combining clinical and pathological parameters are currently used to inform patients’ therapeutic and follow-up strategies [3,60,61], despite their less-than-optimal performance, especially in the high-risk group. In this scenario, the available data on the putative prognostic role of HER2 is notably intriguing, since patients with high-risk NMIBCs may benefit from a spectrum of treatment options carrying different side effects and/or risk of failure.
However, in order to best assess the role of HER2 as a risk stratification marker, alone or in combination, some issues need to be fixed. The findings in the literature are biased by a series of inherent limitations that affect the possibility to draw consistent results—mostly (1) the retrospective fashion of available data, (2) intra- and inter-tumor heterogeneity, and (3) discrepancies among assessment methods used in different studies, namely antibody clones, evaluation criteria, and cut-offs adopted for the definition of HER2 positivity [37,45,62,63]. Hence, large multi-center studies are needed in order to overcome such limitations.

4. HER2 in Divergent Differentiation and Histological Subtypes of BC

Some types of BCs may show peculiar morphological and biological features, which accounts for them being classified as divergent forms (such as squamous, glandular, trophoblastic) or even distinct subtypes, including micropapillary, plasmacytoid, and sarcomatoid tumors [64]. These variants are believed to carry a worse clinical outcome than conventional urothelial carcinoma (UC), both in the NMIBC and MIBC setting [65], although there is no universal agreement on this point; nevertheless, it is highly recommended to carefully assess these features in tissue samples, even when present to a small extent (see below). The use of reliable prognostic and predictive markers might be of pivotal importance in stratifying such BC patients [66].
Variable rates of HER2 expression have been described in these patients. Behzatoglu et al. reported the presence of HER2 overexpression in 56% of micropapillary carcinomas (MPCs) and only 36% of conventional BCs; on the other hand, HER2 positivity rates declined to 20% in the group of BCs with squamous differentiation (SD-BC), and no expression was seen in the cases of sarcomatoid carcinoma (SCs) and BC with glandular differentiation (GD-BC) [67], in keeping with the findings by Wang et al. on a cohort of upper urinary tract UCs [68]. Even lower positivity rates (3–11%) of HER2 amplification/overexpression in SD-BCs were reported in two studies [69,70].
HER2 alterations have been extensively studied in MPCs [71,72,73], with rates of overexpression and amplification in up to 75% and 42% of cases, respectively [74,75], supporting the classification of MPCs as luminal tumors [7,71]. Two studies comparing stage-matched MPC to conventional BC reported higher amplification rates in the first group (12% and 15% vs. 6% and 9%, respectively) [70,73].
When evaluating the presence of MP architecture in BC, it may happen that such morphological features can be detected only in part of an otherwise conventional UC. There is still no agreement about which proportion of MPC within a tumor yields clinical significance; therefore, any MP component, possibly with its percentage, should be reported [76]. According to Bertz et al., CISH disclosed HER2 gene amplification in 30% (3/10) of the BCs harboring a ≥30% MP component [77]. On the other hand, 77% of the HER2-amplified tumors in the cohort studied by Tschui et al. presented with an MP morphology, ranging from <10% to 100%, and the HER2-positive group had significantly higher rates of morphological heterogeneity than the control group [78]. Interestingly, Isharwal et al. reported frequent intratumoral heterogeneity of HER2 amplification within combined tumors (i.e., containing both MPC and conventional UC), in that the MP component showed higher rates of amplification than the conventional one [79]. Furthermore, the presence of higher HER2 rates in the conventional component in these tumors compared to both pure conventional UC and those not combined with MP tumor [66,79] suggests that HER2 activation may play a role in the carcinogenesis of this variant. Interestingly, a gene-expression meta-cohort study of 2411 tumors hinted at a subclassification of MPCs into HER2-like and mesenchymal-like [80], in keeping with the findings by Han et al. [81].
Since HER2 positivity has been described in conventional BC as well, it cannot be used to support the diagnosis of MPC [82,83]. On the other hand, there is no consensus on how to treat MPC, especially the NMI cases [66]; therefore, the therapeutic implications of HER2 overexpression in this variant are yet to be explored [84].
According to some authors, HER2 assessment in MPC may have a prognostic potential, in that some authors have reported an association with worse cancer-specific survival (CSS) after radical cystectomy [73,75]. In keeping with this, HER2 was overexpressed in as many as 70% of patients with angiolymphatic invasion in a cohort of 27 patients analyzed by Goodman et al., both with an early and advanced disease [74].
Moreover, the association between HER2 expression and amplification is not linear [70,84]; Moktefi et al. reported HER2 protein overexpression in as many as 60% of MPC cases, with only 12% showing HER2 amplification with FISH analysis [70]. Such discrepancies may be due to the low frequency of pure MPC, or to other mechanisms supporting the overexpression at the protein level in these tumors, such as mutations in known hotspots, which have been frequently described in MPC [72]. Nevertheless, a rate as high as 40% of activating HER2 mutations has been reported in MPC in the absence of protein overexpression [72]. A further D769N mutation was detected by Tschui et al. in a HER2-amplified tumor, occurring at the same amino acid position than two other mutations (D769H and D769Y) associated with breast cancer, both resulting in the constitutive activation of the enzyme [78].
Plasmacytoid carcinomas (PCs) are rare and biologically aggressive urothelial malignancies, mostly carrying low levels of HER2 expression and amplification [70,85]. Interestingly, a recent study by Kossaï et al. showed that HER2 positivity rates, though overall low in their cohort of PCs (8/32, 25%), were indeed higher as compared to conventional high-grade UCs (0/30) [86].
Small cell neuroendocrine carcinoma (SCNEC) is an uncommon high-grade biologically aggressive malignancy that may affect several organs, including the bladder, carrying poorer outcomes than conventional urothelial BC [64]. A comprehensive whole-genome analysis of these tumors demonstrated a novel in-frame Prt1 oncogene (PVT1)-ERBB2 fusion, resulting in the aberrant expression of the HER2 gene [87,88]. An earlier study reported a 50% positivity of HER2 protein in a cohort of 10 bladder SCNECs [88].

5. HER2 and BC Molecular Subtypes

The biological and clinical heterogeneity of BC has led several researchers to develop classification schemes able to mirror this variability and translate it into distinct subtypes according to their mRNA expression profiles in the last decade. The next steps were to combine these findings into a consensus classification and to implement such molecular subtyping as a risk-stratification tool in routine practice, using IHC in order to assess subtype-specific biomarkers at the protein level [7,8].
According to an earlier classification system proposed by the University of Lund, Sweden, MIBCs can be divided in four molecular subtypes by gene expression profiling, namely UroA, UroB, GU, and SCCL tumors [88]. Using IHC, HER2 expression rates were distinctly different among the groups, ranging from strong (GU) to moderate–low (UroA and UroB) to almost absent (SCCL) [89]. Conversely, HER2 overexpression is associated with Clusters I and II and luminal-like tumors, according to the 2014 Cancer Genome Atlas Network (TGCA) and MD Anderson Cancer Center (MDACC) classifications, respectively [90,91].
These and other groups reported apparently distinct molecular frameworks including varying numbers of subtypes (from 2 to 5), yet overlapping in the top-level distinction between luminal and basal clusters [11]. Accordingly, the Consensus Molecular Classification of MIBC by the Bladder Cancer Molecular Taxonomy Group, based on the analysis of 1750 transcriptomic profiles from sixteen published datasets and two additional cohorts, identifies six classes, namely luminal papillary (LumP), Luminal Non-Specified (LumNS), Luminal Unstable (LumU), Stroma-rich, Basal/Squamous (Ba/Sq), and Neuroendocrine-like (NE-like), with different biological and clinical/prognostic features [6]. Herein, HER2 amplifications were enriched in the LumU subtype, which features the highest cell cycle activity among luminal tumors (p < 0.001), as well as the uppermost somatic mutation load overall (p = 0.009) and a poor prognosis [6]. Nevertheless, patients with LumU BCs may show a good response to radiotherapy and ICI (atezolizumab) [6].
According to Kiss et al. [92], the rate of HER2 alterations at both the gene and protein level is higher in the luminal rather than the basal subtype of MIBC. In keeping with this, HER2 has been used as a surrogate marker of luminal phenotype, usually along with GATA3 and CK20, by a few authors [93,94,95]. Accordingly, Yorozu et al. assessed HER2 status (protein overexpression and/or gene amplification) in a series of 148 UCs of the upper urinary tract, reporting that HER2 positivity was significantly associated with the luminal subtype (p = 0.0030) and a shorter overall survival at univariate analysis (p = 0.0265) [96].
The proposed molecular classifications mostly focus on MIBC. According to the UROMOL study, a comprehensive multi-institutional transcriptional analysis project aiming to classify NMIBC through molecular methods, Class 2 tumors frequently harbored HER2 mutations and were associated to the CIS pathway of progression, carrying higher progression rates to MIBC [97].
All in all, the available data support the hypothesis that HER2 may be an optimal candidate marker to be included in a small and effective antibody panel suitable for BC molecular subtyping in clinical practice.

6. With a Little Help from AI: A Step beyond on the Way of Standardization

A major issue preventing the implementation of routine biomarker assessment in BC is the poor reproducibility of results due to the subjective evaluation done by light microscopy. In recent years, artificial intelligence (AI)-based techniques in pathology have been steadily implemented, leading to the development of automated image analysis tools with the ability to evaluate whole slide images (WSIs) in order to determine several types of parameters [98,99]. Such digital image analysis (DIA) algorithms quantify the expression of IHC biomarkers in manually outlined region of interests (ROIs) and scan through the whole slide, thus yielding highly reproducible and accurate results [98].
In routine practice, the semiquantitative assessment of HER2 IHC-stained slides is performed by pathologists manually, as mentioned before, resulting in interobserver variability, in spite of the presence of international widespread guidelines. A further drawback is the relatively high levels of equivocal cases, especially when such evaluation is performed by non-experienced pathologists [18]. DIA may be of pivotal importance in this setting, to the extent that it has been acknowledged as a diagnostic tool for HER2 status evaluation to be implemented into pathology practice according to focused guidelines by the American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) [100] through several commercially available FDA-cleared and CE-certified algorithms for HER2 IHC quantification [101]. These systems run through a first step of segmentation in order to arrange cells and/or nuclei into discrete staining classes, and they later quantify the percentage of cells in each class. The output may be expressed as membrane connectivity, which is a continual measure of the size distribution of stained membrane fragments ranging from 0 to 1, and is later converted into the guideline-recommended categories of 0, 1+, 2+, 3+ (Figure 2). Since HER2 is a marker of tumor cells only, the step of ROI definition may be skipped; furthermore, sensitivity may be manually set by the user according to perceived staining intensity [98].
High rates of agreement between manual and DIA HER2 scoring and between IHC and FISH results have been reported in a study on breast carcinoma [102]. In a recent study, HER2 DIA connectivity showed the strongest association among other prognostic parameters with pathologic complete response in a cohort of HER2+ invasive breast carcinomas treated with anti-HER2 agents in the neoadjuvant setting [103].
The application of such methods in clinical practice needs (1) prior intra-laboratory validation through a comparison with surrogate methods (such as, detection of HER2 at gene level) or consensus images, along with supervision by expert pathologists, and (2) to follow evidence-based guidelines and implementation of regular maintenance and accreditation programs [101,103].
Since FISH is a more complex and time-consuming method than IHC in assessing HER2 status, DIA systems have also been established in order to overcome these issues by automatically detecting, classifying, and counting cells of interest within the slides on the basis of pre-set parameters, including color, intensity, size, pattern, and shape, yielding overall concordance rates approaching 100% [101].

7. Conclusions

HER2 is a versatile molecule to be exploited as a diagnostic, prognostic, and predictive tissue biomarker in the assessment of urothelial lesions. Available findings so far suggest that is can be easily implemented in clinical practice, especially with the aid of novel AI-based methods of assessment. In order to define its real potential for patients’ risk stratification and as a therapeutic target, further well-designed and more focused studies are warranted.

Author Contributions

Conceptualization, F.S. and L.C.; methodology, F.S., M.Z., M.C.P., A.B. and U.G.F.; writing—original draft preparation, F.S., M.Z. and A.P.; writing—review and editing, all authors; supervision, L.C. and G.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. IARC-WHO. Global Cancer Observatory. Available online: https://gco.iarc.fr (accessed on 9 January 2023).
  2. Witjes, J.A.; Bruins, H.M.; Carrión, A.; Cathomas, R.; Compérat, E.M.; Efstathiou, J.A.; Kietkau, R.; Gakis, G.; Van der Heijden, A.G.; Lorch, A.; et al. (Eds.) EAU Guidelines on Muscle-Invasive and Metastatic Bladder Cancer; EAU: Arnhem, The Netherlands, 2022. [Google Scholar]
  3. Babjuk, M.; Burger, M.; Compérat, E.; Gontero, P.; Liedberg, F.; Masson-Lecomte, A.; Mostafid, A.H.; Palou, J.; Van Rhijn, B.W.G.; Roupret, M.; et al. EAU Guidelines on Non-Muscle-Invasive Bladder Cancer (TaT1 and CIS); EAU: Arnhem, The Netherlands, 2022. [Google Scholar]
  4. D’Angelo, A.; Chapman, R.; Sirico, M.; Sobhani, N.; Catalano, M.; Mini, E.; Roviello, G. An update on antibody-drug conjugates in urothelial carcinoma: State of the art strategies and what comes next. Cancer Chemother. Pharmacol. 2022, 90, 191–205. [Google Scholar] [CrossRef] [PubMed]
  5. Smith, N.D.; Prasad, S.M.; Patel, A.R.; Weiner, A.B.; Pariser, J.J.; Razmaria, A.; Maene, C.; Schuble, T.; Pierce, B.; Steinberg, G.D. Bladder Cancer Mortality in the United States: A Geographic and Temporal Analysis of Socioeconomic and Environmental Factors. J. Urol. 2016, 195, 290–296. [Google Scholar] [CrossRef] [PubMed]
  6. Kamoun, A.; De Reyniès, A.; Allory, Y.; Sjödahl, G.; Robertson, A.G.; Seiler, R.; Hoadley, K.A.; Groeneveld, C.S.; Al-Ahmadie, H.; Choi, W.; et al. A Consensus Molecular Classification of Muscle-invasive Bladder Cancer. Eur. Urol. 2020, 77, 420–433. [Google Scholar] [CrossRef]
  7. Sanguedolce, F.; Zanelli, M.; Palicelli, A.; Ascani, S.; Zizzo, M.; Cocco, G.; Björnebo, L.; Lantz, A.; Landriscina, M.; Conteduca, V.; et al. Are We Ready to Implement Molecular Subtyping of Bladder Cancer in Clinical Practice? Part 2: Subtypes and Divergent Differentiation. Int. J. Mol. Sci. 2022, 23, 7844. [Google Scholar] [CrossRef]
  8. Sanguedolce, F.; Zanelli, M.; Palicelli, A.; Ascani, S.; Zizzo, M.; Cocco, G.; Björnebo, L.; Lantz, A.; Landriscina, M.; Conteduca, V.; et al. Are We Ready to Implement Molecular Subtyping of Bladder Cancer in Clinical Practice? Part 1: General Issues and Marker Expression. Int. J. Mol. Sci. 2022, 23, 7819. [Google Scholar] [CrossRef]
  9. Cooley, L.F.; McLaughlin, K.A.; Meeks, J.J. Genomic and Therapeutic Landscape of Non-muscle-invasive Bladder Cancer. Urol. Clin. N. Am. 2020, 47, 35–46. [Google Scholar] [CrossRef]
  10. EAU Guidelines. Edn. EAU: Arnhem, The Netherlands, 2023. Available online: https://uroweb.org/eau-guidelines (accessed on 9 January 2023).
  11. Fong, M.H.Y.; Feng, M.; McConkey, D.J.; Choi, W. Update on bladder cancer molecular subtypes. Transl. Androl. Urol. 2020, 9, 2881–2889. [Google Scholar] [CrossRef]
  12. Pritzker, K.P. Predictive and prognostic cancer biomarkers revisited. Expert Rev. Mol. Diagn. 2015, 15, 971–974. [Google Scholar] [CrossRef]
  13. Taylor, C.R. Introduction to Predictive Biomarkers: Definitions and Characteristics. In Predictive Biomarkers in Oncology; Badve, S., Kumar, G.L., Eds.; Applications in Precision Medicine; Springer: Berlin/Heidelberg, Germany, 2019. [Google Scholar]
  14. Moasser, M.M. The oncogene HER2: Its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene 2007, 26, 6469–6487. [Google Scholar] [CrossRef] [PubMed]
  15. Meric-Bernstam, F.; Johnson, A.M.; Dumbrava, E.E.I.; Raghav, K.; Balaji, K.; Bhatt, M.; Murthy, R.K.; Rodon, J.; Piha-Paul, S.A. Advances in HER2-Targeted Therapy: Novel Agents and Opportunities Beyond Breast and Gastric Cancer. Clin. Cancer Res. 2019, 25, 2033–2041. [Google Scholar] [CrossRef] [Green Version]
  16. Albarrán, V.; Rosero, D.I.; Chamorro, J.; Pozas, J.; San Román, M.; Barrill, A.M.; Alía, V.; Sotoca, P.; Guerrero, P.; Calvo, J.C.; et al. Her-2 Targeted Therapy in Advanced Urothelial Cancer: From Monoclonal Antibodies to Antibody-Drug Conjugates. Int. J. Mol. Sci. 2022, 23, 12659. [Google Scholar] [CrossRef] [PubMed]
  17. Sanguedolce, F.; Bufo, P. HER2 assessment by silver in situ hybridization: Where are we now? Expert Rev. Mol. Diagn. 2015, 15, 385–398. [Google Scholar] [CrossRef]
  18. Wolff, A.C.; Hammond, M.E.H.; Allison, K.H.; Harvey, B.E.; Mangu, P.B.; Bartlett, J.M.S.; Bilous, M.; Ellis, I.O.; Fitzgibbons, P.; Hanna, W.; et al. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J. Clin. Oncol. 2018, 36, 2105–2122. [Google Scholar] [CrossRef]
  19. Mooso, B.A.; Vinall, R.L.; Mudryj, M.; Yap, S.A.; deVere White, R.W.; Ghosh, P.M. The role of EGFR family inhibitors in muscle invasive bladder cancer: A review of clinical data and molecular evidence. J. Urol. 2015, 193, 19–29. [Google Scholar] [CrossRef]
  20. Bilski, K.; Zapała, Ł.; Skrzypczyk, M.A.; Oszczudłowski, M.; Dobruch, J. Review on gender differences in non-muscle invasive bladder cancer. Transl. Androl. Urol. 2019, 8, 12–20. [Google Scholar] [CrossRef]
  21. Yan, M.; Schwaederle, M.; Arguello, D.; Millis, S.Z.; Gatalica, Z.; Kurzrock, R. HER2 expression status in diverse cancers: Review of results from 37,992 patients. Cancer Metastasis Rev. 2015, 34, 157–164. [Google Scholar] [CrossRef]
  22. Lae, M.; Couturier, J.; Oudard, S.; Radvanyi, F.; Beuzeboc, P.; Vieillefond, A. Assessing HER2 gene amplification as a potential target for therapy in invasive urothelial bladder cancer with a standardized methodology: Results in 1005 patients. Ann. Oncol. 2010, 21, 815–819. [Google Scholar] [CrossRef] [PubMed]
  23. Sanguedolce, F.; Bufo, P.; Carrieri, G.; Cormio, L. Predictive markers in bladder cancer: Do we have molecular markers ready for clinical use? Crit. Rev. Clin. Lab. Sci. 2014, 51, 291–304. [Google Scholar] [CrossRef] [PubMed]
  24. Sanguedolce, F.; Cormio, A.; Bufo, P.; Carrieri, G.; Cormio, L. Molecular markers in bladder cancer: Novel research frontiers. Crit. Rev. Clin. Lab. Sci. 2015, 52, 242–255. [Google Scholar] [CrossRef]
  25. McKenney, J.K. Precursor lesions of the urinary bladder. Histopathology 2019, 74, 68–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sanguedolce, F.; Brunelli, M.; D’amuri, A.; Calò, B.; Mancini, V.; Carrieri, G.; Cormio, L. Evolving concepts and use of immunohistochemical biomarkers in flat non-neoplastic urothelial lesions: WHO 2016 classification update with diagnostic algorithm. Biomarkers 2018, 23, 305–314. [Google Scholar] [CrossRef] [PubMed]
  27. Chow, N.H.; Liu, H.S.; Yang, H.B.; Chan, S.H.; Su, I.J. Expression patterns of erbB receptor family in normal urothelium and transitional cell carcinoma. An immunohistochemical study. Virchows Arch. 1997, 430, 461–466. [Google Scholar] [CrossRef] [PubMed]
  28. Petraki, C.D.; Sfikas, C.P. Non-papillary urothelial lesions of the urinary bladder: Morphological classification and immunohistochemical markers. In Vivo 2008, 22, 493–501. [Google Scholar]
  29. Schwarz, S.; Rechenmacher, M.; Filbeck, T.; Knuechel, R.; Blaszyk, H.; Hartmann, A.; Brockhoff, G. Value of multicolour fluorescence in situ hybridisation (UroVysion) in the differential diagnosis of flat urothelial lesions. J. Clin. Pathol. 2008, 61, 272–277. [Google Scholar] [CrossRef] [PubMed]
  30. Jung, S.; Wu, C.; Eslami, Z.; Tanguay, S.; Aprikian, A.; Kassouf, W.; Brimo, F. The role of immunohistochemistry in the diagnosis of flat urothelial lesions: A study using CK20, CK5/6, P53, Cd138, and Her2/Neu. Ann. Diagn. Pathol. 2014, 18, 27–32. [Google Scholar] [CrossRef] [PubMed]
  31. Barth, I.; Schneider, U.; Grimm, T.; Karl, A.; Horst, D.; Gaisa, N.T.; Knüchel, R.; Garczyk, S. Progression of urothelial carcinoma in situ of the urinary bladder: A switch from luminal to basal phenotype and related therapeutic implications. Virchows Arch. 2018, 472, 749–758. [Google Scholar] [CrossRef] [PubMed]
  32. Wagner, U.; Sauter, G.; Moch, H.; Novotna, H.; Epper, R.; Mihatsch, M.J.; Waldman, F.M. Patterns of p53, erbB-2, and EGF-r expression in premalignant lesions of the urinary bladder. Hum. Pathol. 1995, 26, 970–978. [Google Scholar] [CrossRef]
  33. Gunia, S.; Koch, S.; Hakenberg, O.W.; May, M.; Kakies, C.; Erbersdobler, A. Different HER2 protein expression profiles aid in the histologic differential diagnosis between urothelial carcinoma in situ (CIS) and non-CIS conditions (dysplasia and reactive atypia) of the urinary bladder mucosa. Am. J. Clin. Pathol. 2011, 136, 881–888. [Google Scholar] [CrossRef]
  34. Garczyk, S.; Ortiz-Brüchle, N.; Schneider, U.; Lurje, I.; Guricova, K.; Gaisa, N.T.; Lorsy, E.; Lindemann-Docter, K.; Heidenreich, A.; Knüchel, R. Next-Generation Sequencing Reveals Potential Predictive Biomarkers and Targets of Therapy for Urothelial Carcinoma in Situ of the Urinary Bladder. Am. J. Pathol. 2020, 190, 323–332. [Google Scholar] [CrossRef]
  35. Sanguedolce, F.; Calò, B.; Chirico, M.; Falagario, U.; Busetto, G.M.; Zanelli, M.; Bisagni, A.; Zizzo, M.; Ascani, S.; Carrieri, G.; et al. Distinctive morphological and molecular features of urothelial carcinoma with an inverted growth pattern. J. Pathol. Transl. Med. 2021, 55, 239–246. [Google Scholar] [CrossRef]
  36. Iyer, G.; Al-Ahmadie, H.; Schultz, N.; Hanrahan, A.J.; Ostrovnaya, I.; Balar, A.; Kim, P.H.; Lin, O.; Weinhold, N.; Sander, C.; et al. Prevalence and Co-Occurrence of Actionable Genomic Alterations in High-Grade Bladder Cancer. J. Clin. Oncol. 2013, 31, 3133–3140. [Google Scholar] [CrossRef]
  37. Jimenez, R.E.; Hussain, M.; Bianco, F.J.; Vaishampayan, U.; Tabazcka, P.; Sakr, W.A.; Pontes, J.E.; Wood, D.P., Jr.; Grignon, D.J. Her-2/neu overexpression in muscle-invasive urothelial carcinoma of the bladder: Prognostic significance and comparative analysis in primary and metastatic tumors. Clin. Cancer Res. 2001, 7, 2440–2447. [Google Scholar] [PubMed]
  38. Fleischmann, A.; Rotzer, D.; Seiler, R.; Studer, U.E.; Thalmann, G.N. Her2 amplification is significantly more frequent in lymph node metastases from urothelial bladder cancer than in the primary tumours. Eur. Urol. 2011, 60, 350–357. [Google Scholar] [CrossRef] [PubMed]
  39. Grivas, P.D.; Day, M.; Hussain, M. Urothelial carcinomas: A focus on human epidermal receptors signaling. Am. J. Transl. Res. 2011, 3, 362–373. [Google Scholar] [PubMed]
  40. Latif, Z.; Watters, A.D.; Dunn, I.; Grigor, K.; Underwood, M.A.; Bartlett, J.M. HER2/neu gene amplification and protein overexpression in G3 pT2 transitional cell carcinoma of the bladder: A role for anti-HER2 therapy? Eur. J. Cancer 2004, 40, 56–63. [Google Scholar] [CrossRef]
  41. Nedjadi, T.; Al-Maghrabi, J.; Assidi, M.; Dallol, A.; Al-Kattabi, H.; Chaudhary, A.; Al-Sayyad, A.; Al-Ammari, A.; Abuzenadah, A.; Buhmeida, A.; et al. Prognostic value of HER2 status in bladder transitional cell carcinoma revealed by both IHC and BDISH techniques. BMC Cancer 2016, 16, 653. [Google Scholar] [CrossRef]
  42. Kolla, S.B.; Seth, A.; Singh, M.K.; Gupta, N.P.; Hemal, A.K.; Dogra, P.N.; Kumar, R. Prognostic significance of Her2/neu overexpression in patients with muscle invasive urinary bladder cancer treated with radical cystectomy. Int. Urol. Nephrol. 2008, 40, 321–327. [Google Scholar] [CrossRef] [PubMed]
  43. Krüger, S.; Weitsch, G.; Büttner, H.; Matthiensen, A.; Böhmer, T.; Marquardt, T.; Sayk, F.; Feller, A.C.; Böhle, A. Overexpression of c-erbB-2 oncoprotein in muscle-invasive bladder carcinoma: Relationship with gene amplification, clinicopathological parameters and prognostic outcome. Int. J. Oncol. 2002, 21, 981–987. [Google Scholar] [CrossRef] [PubMed]
  44. Kriegmair, M.C.; Wirtz, R.M.; Worst, T.S.; Breyer, J.; Ritter, M.; Keck, B.; Boehmer, C.; Otto, W.; Eckstein, M.; Weis, C.A.; et al. Prognostic Value of Molecular Breast Cancer Subtypes based on Her2, ESR1, PGR and Ki67 mRNA-Expression in Muscle Invasive Bladder Cancer. Transl. Oncol. 2018, 11, 467–476. [Google Scholar] [CrossRef] [PubMed]
  45. Zhao, J.; Xu, W.; Zhang, Z.; Song, R.; Zeng, S.; Sun, Y.; Xu, C. Prognostic role of HER2 expression in bladder cancer: A systematic review and meta-analysis. Int. Urol. Nephrol. 2015, 47, 87–94. [Google Scholar] [CrossRef]
  46. Nini, A.; Hoffmann, M.J.; Lampignano, R.; Große Siemer, R.; van Dalum, G.; Szarvas, T.; Cotarelo, C.L.; Schulz, W.A.; Niederacher, D.; Neubauer, H.; et al. Evaluation of HER2 expression in urothelial carcinoma cells as a biomarker for circulating tumor cells. Cytom. B Clin. Cytom. 2020, 98, 355–367. [Google Scholar] [CrossRef] [Green Version]
  47. Pietzak, E.J.; Bagrodia, A.; Cha, E.K.; Drill, E.N.; Iyer, G.; Isharwal, S.; Ostrovnaya, I.; Baez, P.; Li, Q.; Berger, M.F.; et al. Next-generation Sequencing of Nonmuscle Invasive Bladder Cancer Reveals Potential Biomarkers and Rational Therapeutic Targets. Eur. Urol. 2017, 72, 952–959. [Google Scholar] [CrossRef] [PubMed]
  48. Moustakas, G.; Kampantais, S.; Nikolaidou, A.; Vakalopoulos, I.; Tzioufa, V.; Dimitriadis, G. HER-2 Overexpression is a Negative Predictive Factor for Recurrence in Patients With non-Muscle-Invasive Bladder Cancer on Intravesical Therapy. J. Int. Med. Res. 2020, 48, 300060519895847. [Google Scholar] [CrossRef]
  49. Ding, W.; Tong, S.; Gou, Y.; Sun, C.; Wang, H.; Chen, Z.; Tan, J.; Xu, K.; Xia, G.; Ding, Q. Human epidermal growth factor receptor 2: A significant indicator for predicting progression in non-muscle-invasive bladder cancer especially in high-risk groups. World J. Urol. 2015, 33, 1951–1957. [Google Scholar] [CrossRef] [PubMed]
  50. Breyer, J.; Wirtz, R.M.; Otto, W.; Laible, M.; Schlombs, K.; Erben, P.; Kriegmair, M.C.; Stoehr, R.; Eidt, S.; Denzinger, S.; et al. Predictive value of molecular subtyping in NMIBC by RT-qPCR of ERBB2, ESR1, PGR and MKI67 from formalin fixed TUR biopsies. Oncotarget 2017, 8, 67684–67695. [Google Scholar] [CrossRef] [PubMed]
  51. Lim, S.D.; Cho, Y.M.; Choi, G.S.; Park, H.K.; Paick, S.H.; Kim, W.Y.; Kim, S.N.; Yoon, G. Clinical Significance of Substaging and HER2 Expression in Papillary Nonmuscle Invasive Urothelial Cancers of the Urinary Bladder. J. Korean Med. Sci. 2015, 30, 1068–1077. [Google Scholar] [CrossRef]
  52. Sikic, D.; Eckstein, M.; Weyerer, V.; Kubon, J.; Breyer, J.; Roghmann, F.; Kunath, F.; Keck, B.; Erben, P.; Hartmann, A.; et al. High expression of ERBB2 is an independent risk factor for reduced recurrence-free survival in patients with stage T1 non-muscle-invasive bladder cancer. Urol. Oncol. 2022, 40, 63.e9–63.e18. [Google Scholar] [CrossRef] [PubMed]
  53. Chen, P.C.H.; Yu, H.J.; Chang, Y.H.; Pan, C.C. Her2 amplification distinguishes a subset of non-muscle-invasive bladder cancers with a high risk of progression. J. Clin. Pathol. 2013, 66, 113–119. [Google Scholar] [CrossRef]
  54. Olsson, H.; Fyhr, I.M.; Hultman, P.; Jahnson, S. HER2 status in primary stage T1 urothelial cell carcinoma of the urinary bladder. Scand. J. Urol. Nephrol. 2012, 46, 102–107. [Google Scholar] [CrossRef]
  55. Sato, K.; Moriyama, M.; Mori, S.; Saito, M.; Watanuki, T.; Terada, K.; Okuhara, E.; Akiyama, T.; Toyoshima, K.; Yamamoto, T.; et al. An immunohistologic evaluation of C-erbB-2 gene product in patients with urinary bladder carcinoma. Cancer 1992, 70, 2493–2498. [Google Scholar] [CrossRef]
  56. Bongiovanni, L.; Arena, V.; Vecchio, F.M.; Racioppi, M.; Bassi, P.; Pierconti, F. HER-2 immunohistochemical expression as prognostic marker in high-grade T1 bladder cancer (T1G3). Arch. Ital. Urol. Androl. 2013, 85, 73–77. [Google Scholar] [CrossRef] [Green Version]
  57. Rodriguez Pena, M.D.C.; Chaux, A.; Eich, M.L.; Tregnago, A.C.; Taheri, D.; Borhan, W.; Sharma, R.; Rezaei, M.K.; Netto, G.J. Immunohistochemical assessment of basal and luminal markers in non-muscle invasive urothelial carcinoma of bladder. Virchows Arch. 2019, 475, 349–356. [Google Scholar] [CrossRef] [PubMed]
  58. Cormio, L.; Sanguedolce, F.; Cormio, A.; Massenio, P.; Pedicillo, M.C.; Cagiano, S.; Calò, G.; Pagliarulo, V.; Carrieri, G.; Bufo, P. Human epidermal growth factor receptor 2 expression is more important than bacillus calmette guerin treatment in predicting the outcome of T1G3 bladder cancer. Oncotarget 2017, 8, 25433–25441. [Google Scholar] [CrossRef] [PubMed]
  59. Sanguedolce, F.; Cormio, A.; Massenio, P.; Pedicillo, M.C.; Cagiano, S.; Fortunato, F.; Calò, B.; Di Fino, G.; Carrieri, G.; Bufo, P.; et al. Altered expression of HER-2 and the mismatch repair genes MLH1 and MSH2 predicts the outcome of T1 high-grade bladder cancer. J. Cancer Res. Clin. Oncol. 2018, 144, 637–644. [Google Scholar] [CrossRef] [PubMed]
  60. Cambier, S.; Sylvester, R.J.; Collette, L.; Gontero, P.; Brausi, M.A.; Van Andel, G.; Kirkels, W.J.; Silva, F.C.; Oosterlinck, W.; Prescott, S.; et al. EORTC Nomograms and Risk Groups for Predicting Recurrence, Progression, and Disease-specific and Overall Survival in Non-Muscle-invasive Stage Ta-T1 Urothelial Bladder Cancer Patients Treated with 1–3 Years of Maintenance Bacillus Calmette-Guérin. Eur. Urol. 2016, 69, 60–69. [Google Scholar] [CrossRef]
  61. Chang, S.S.; Boorjian, S.A.; Chou, R.; Clark, P.E.; Daneshmand, S.; Konety, B.R.; Pruthi, R.; Quale, D.Z.; Ritch, C.R.; Seigne, J.D.; et al. Diagnosis and Treatment of Non-Muscle Invasive Bladder Cancer: AUA/SUO Guideline. J. Urol. 2016, 196, 1021–1029. [Google Scholar] [CrossRef]
  62. De Carlo, C.; Valeri, M.; Corbitt, D.N.; Cieri, M.; Colombo, P. Non-muscle invasive bladder cancer biomarkers beyond morphology. Front. Oncol. 2022, 12, 947446. [Google Scholar] [CrossRef]
  63. Gandour-Edwards, R.; Lara, P.N., Jr.; Folkins, A.K.; LaSalle, J.M.; Beckett, L.; Li, Y.; Meyers, F.J.; DeVere-White, R. Does HER2/neu expression provide prognostic information in patients with advanced urothelial carcinoma? Cancer 2002, 95, 1009–1015. [Google Scholar] [CrossRef]
  64. WHO Classification of Tumours Editorial Board. Urinary and Male Genital Tumours, 5th ed.; WHO Classification of Tumours Series; International Agency for Research on Cancer: Lyon, France; WHO: Geneva, Switzerland, 2022; Volume 8, Available online: https://tumourclassification.iarc.who.int/chapters/36 (accessed on 9 January 2023).
  65. Veskimae, E.; Espinos, E.L.; Bruins, H.M.; Yuan, Y.; Sylvester, R.; Kamat, A.M.; Shariat, S.F.; Witjes, J.A.; Comperat, E.M. What Is the Prognostic and Clinical Importance of Urothelial and Nonurothelial Histological Variants of Bladder Cancer in Predicting Oncological Outcomes in Patients with Muscle-invasive and Metastatic Bladder Cancer? A European Association of Urology Muscle Invasive and Metastatic Bladder Cancer Guidelines Panel Systematic Review. Eur. Urol. Oncol. 2019, 2, 625–642. [Google Scholar]
  66. Sanguedolce, F.; Calò, B.; Mancini, V.; Zanelli, M.; Palicelli, A.; Zizzo, M.; Ascani, S.; Carrieri, G.; Cormio, L. Non-Muscle Invasive Bladder Cancer with Variant Histology: Biological Features and Clinical Implications. Oncology 2021, 99, 345–358. [Google Scholar] [CrossRef]
  67. Behzatoğlu, K.; Yörükoğlu, K.; Demir, H.; Bal, N. Human Epidermal Growth Factor Receptor 2 Overexpression in Micropapillary and Other Variants of Urothelial Carcinoma. Eur. Urol. Focus 2018, 4, 399–404. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, X.; MacLennan, G.T.; Zhang, S.; Montironi, R.; Lopez-Beltran, A.; Tan, P.H.; Foster, S.; Baldridge, L.A.; Cheng, L. Sarcomatoid carcinoma of the upper urinary tract: Clinical outcome and molecular characterization. Hum. Pathol. 2009, 40, 211–217. [Google Scholar] [CrossRef] [PubMed]
  69. Wucherpfennig, S.; Rose, M.; Maurer, A.; Cassataro, M.A.; Seillier, L.; Morsch, R.; Hammad, E.; Baldia, P.H.; Ecke, T.H.; Vögeli, T.A.; et al. Evaluation of Therapeutic Targets in Histological Subtypes of Bladder Cancer. Int. J. Mol. Sci. 2021, 22, 11547. [Google Scholar] [CrossRef]
  70. Moktefi, A.; Pouessel, D.; Liu, J.; Sirab, N.; Maille, P.; Soyeux, P.; Bergman, C.C.; Auriault, M.L.; Vordos, D.; Taille, A.; et al. Reappraisal of HER2 status in the spectrum of advanced urothelial carcinoma: A need of guidelines for treatment eligibility. Mod. Pathol. 2018, 31, 1270–1281. [Google Scholar] [CrossRef] [PubMed]
  71. Guo, C.C.; Dadhania, V.; Zhang, L.; Majewski, T.; Bondaruk, J.; Sykulski, M.; Wronowska, W.; Gambin, A.; Wang, Y.; Zhang, S.; et al. Gene Expression Profile of the Clinically Aggressive Micropapillary Variant of Bladder Cancer. Eur. Urol. 2016, 70, 611–620. [Google Scholar] [CrossRef]
  72. Ross, J.S.; Wang, K.; Gay, L.M.; Al-Rohil, R.N.; Nazeer, T.; Sheehan, C.E.; Jennings, T.A.; Otto, G.A.; Donahue, A.; He, J.; et al. A high frequency of activating extracellular domain ERBB2 (HER2) mutation in micropapillary urothelial carcinoma. Clin. Cancer Res. 2014, 20, 68–75. [Google Scholar] [CrossRef] [PubMed]
  73. Schneider, S.A.; Sukov, W.R.; Frank, I.; Boorjian, S.A.; Costello, B.A.; Tarrell, R.F.; Thapa, P.; Houston Thompson, R.; Tollefson, M.K.; Jeffrey Karnes, R.; et al. Outcome of patients with micropapillary urothelial carcinoma following radical cystectomy: ERBB2 (HER2) amplification identifies patients with poor outcome. Mod. Pathol. 2014, 27, 758–764. [Google Scholar] [CrossRef] [PubMed]
  74. Goodman, A.L.; Osunkoya, A.O. Human epidermal growth factor receptor 2 expression in micropapillary urothelial carcinoma of the bladder: An analysis of 27 cases. Hum. Pathol. 2016, 57, 160–164. [Google Scholar] [CrossRef] [PubMed]
  75. Zinnall, U.; Weyerer, V.; Compérat, E.; Camparo, P.; Gaisa, N.T.; Knuechel-Clarke, R.; Perren, A.; Lugli, A.; Toma, M.; Baretton, G.; et al. Micropapillary urothelial carcinoma: Evaluation of HER2 status and immunohistochemical characterization of the molecular subtype. Hum. Pathol. 2018, 80, 55–64. [Google Scholar] [CrossRef]
  76. Sanguedolce, F.; Cormio, A.; Calò, B.; Landriscina, M.; Carvalho-Dias, E.; Cormio, L. Micropapillary bladder cancer, a variant histology of the elderly. J. Gerontol. Geriatr. 2018, 66, 222–227. [Google Scholar]
  77. Bertz, S.; Wach, S.; Taubert, H.; Merten, R.; Krause, F.S.; Schick, S.; Ott, O.J.; Weigert, E.; Dworak, O.; Rödel, C.; et al. Micropapillary morphology is an indicator of poor prognosis in patients with urothelial carcinoma treated with transurethral resection and radiochemotherapy. Virchows Arch. 2016, 469, 339–344. [Google Scholar] [CrossRef]
  78. Tschui, J.; Vassella, E.; Bandi, N.; Baumgartner, U.; Genitsch, V.; Rotzer, D.; Seiler, R.; Thalmann, G.N.; Fleischmann, A. Morphological and molecular characteristics of HER2 amplified urothelial bladder cancer. Virchows Arch. 2015, 466, 703–710. [Google Scholar] [CrossRef] [PubMed]
  79. Isharwal, S.; Huang, H.; Nanjangud, G.; Audenet, F.; Chen, Y.B.; Gopalan, A.; Fine, S.W.; Tickoo, S.K.; Lee, B.H.; Iyer, G.; et al. Intratumoral heterogeneity of ERBB2 amplification and HER2 expression in micropapillary urothelial carcinoma. Hum. Pathol. 2018, 77, 63–69. [Google Scholar] [CrossRef]
  80. Tan, T.Z.; Rouanne, M.; Tan, K.T.; Huang, R.Y.; Thiery, J.P. Molecular Subtypes of Urothelial Bladder Cancer: Results from a Meta-Cohort Analysis of 2411 Tumors. Eur. Urol. 2019, 75, 423–432. [Google Scholar] [CrossRef] [PubMed]
  81. Han, L.; Gallan, A.J.; Steinberg, G.D.; Sweis, R.F.; Paner, G.P. Morphological correlation of urinary bladder cancer molecular subtypes in radical cystectomies. Hum. Pathol. 2020, 106, 54–61. [Google Scholar] [CrossRef] [PubMed]
  82. Warrick, J.I.; Knowles, M.A.; Yves, A.; van der Kwast, T.; Grignon, D.J.; Kristiansen, G.; Egevad, L.; Hartmann, A.; Cheng, L. Report From the International Society of Urological Pathology (ISUP) Consultation Conference On Molecular Pathology Of Urogenital Cancers. II. Molecular Pathology of Bladder Cancer: Progress and Challenges. Am. J. Surg. Pathol. 2020, 44, e30–e46. [Google Scholar] [CrossRef]
  83. Sanguedolce, F.; Russo, D.; Mancini, V.; Selvaggio, O.; Calo, B.; Carrieri, G.; Cormio, L. Prognostic and therapeutic role of HER2 expression in micropapillary carcinoma of the bladder. Mol. Clin. Oncol. 2019, 10, 205–213. [Google Scholar] [CrossRef]
  84. Takahara, T.; Murase, Y.; Tsuzuki, T. Urothelial carcinoma: Variant histology, molecular subtyping, and immunophenotyping significant for treatment outcomes. Pathology 2021, 53, 56–66. [Google Scholar] [CrossRef]
  85. Kim, B.; Kim, G.; Song, B.; Lee, C.; Park, J.H.; Moon, K.C. HER2 Protein Overexpression and Gene Amplification in Plasmacytoid Urothelial Carcinoma of the Urinary Bladder. Dis. Markers 2016, 2016, 8463731. [Google Scholar] [CrossRef]
  86. Kossaï, M.; Radulescu, C.; Adam, J.; Dziegielewski, A.; Signolle, N.; Sibony, M.; Lebret, T.; Allory, Y.; Rouanne, M. Plasmacytoid urothelial carcinoma (UC) are luminal tumors with similar CD8+ Tcell density and PD-L1 protein expression on immune cells as compared to conventional UC. Urol. Oncol. 2022, 40, 12.e1–12.e11. [Google Scholar] [CrossRef]
  87. Shen, P.; Jing, Y.; Zhang, R.; Cai, M.C.; Ma, P.; Chen, H.; Zhuang, G. Comprehensive genomic profiling of neuroendocrine bladder cancer pinpoints molecular origin and potential therapeutics. Oncogene 2018, 37, 3039–3044. [Google Scholar] [CrossRef]
  88. Soriano, P.; Navarro, S.; Gil, M.; Llombart-Bosch, A. Small-cell carcinoma of the urinary bladder: A clinico-pathological study of ten cases. Virchows Arch. 2004, 445, 292–297. [Google Scholar] [CrossRef] [PubMed]
  89. Sjödahl, G.; Lövgren, K.; Lauss, M.; Patschan, O.; Gudjonsson, S.; Chebil, G.; Aine, M.; Eriksson, P.; Månsson, W.; Lindgren, D.; et al. Toward a molecular pathologic classification of urothelial carcinoma. Am. J. Pathol. 2013, 183, 681–691. [Google Scholar] [CrossRef] [Green Version]
  90. Robertson, A.G.; Kim, J.; Al-Ahmadie, H.; Bellmunt, J.; Guo, G.; Cherniack, A.D.; Hinoue, T.; Laird, P.W.; Hoadley, K.A.; Akbani, R.; et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell 2017, 171, 540–556.e25. [Google Scholar] [CrossRef]
  91. Choi, W.; Porten, S.; Kim, S.; Willis, D.; Plimack, E.R.; Hoffman-Censits, J.; Roth, B.; Cheng, T.; Tran, M.; Lee, I.L.; et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 2014, 25, 152–165. [Google Scholar] [CrossRef] [PubMed]
  92. Kiss, B.; Wyatt, A.W.; Douglas, J.; Skuginna, V.; Mo, F.; Anderson, S.; Rotzer, D.; Fleischmann, A.; Genitsch, V.; Hayashi, T.; et al. Her2 alterations in muscle-invasive bladder cancer: Patient selection beyond protein expression for targeted therapy. Sci. Rep. 2017, 7, 42713. [Google Scholar] [CrossRef]
  93. Garczyk, S.; Bischoff, F.; Schneider, U.; Golz, R.; von Rundstedt, F.C.; Knüchel, R.; Degener, S. Intratumoral heterogeneity of surrogate molecular subtypes in urothelial carcinoma in situ of the urinary bladder: Implications for prognostic stratification of high-risk non-muscle-invasive bladder cancer. Virchows Arch. 2021, 479, 325–335. [Google Scholar] [CrossRef]
  94. Haghayeghi, K.; Lu, S.; Matoso, A.; Schiff, S.F.; Mueller-Leonhard, C.; Amin, A. Association of current molecular subtypes in urothelial carcinoma with patterns of muscularis propria invasion. Virchows Arch. 2021, 479, 515–521. [Google Scholar] [CrossRef] [PubMed]
  95. Schnitzler, T.; Ortiz-Brüchle, N.; Schneider, U.; Lurje, I.; Guricova, K.; Buchner, A.; Schulz, G.B.; Heidenreich, A.; Gaisa, N.T.; Knüchel, R.; et al. Pure high-grade papillary urothelial bladder cancer: A luminal-like subgroup with potential for targeted therapy. Cell. Oncol. 2020, 43, 807–819. [Google Scholar] [CrossRef]
  96. Yorozu, T.; Sato, S.; Kimura, T.; Iwatani, K.; Onuma, H.; Yanagisawa, T.; Miki, J.; Egawa, S.; Ikegami, M.; Takahashi, H. HER2 Status in Molecular Subtypes of Urothelial Carcinoma of the Renal Pelvis and Ureter. Clin. Genitourin. Cancer 2020, 18, e443–e449. [Google Scholar] [CrossRef]
  97. Hedegaard, J.; Lamy, P.; Nordentoft, I.; Algaba, F.; Høyer, S.; Ulhøi, B.P.; Vang, S.; Reinert, T.; Hermann, G.G.; Mogensen, K.; et al. Comprehensive Transcriptional Analysis of Early-Stage Urothelial Carcinoma. Cancer Cell 2016, 30, 27–42. [Google Scholar] [CrossRef] [PubMed]
  98. Li, Z.; Bui, M.M.; Pantanowitz, L. Clinical tissue biomarker digital image analysis: A review of current applications. Hum. Pathol. Rep. 2022, 28, 300633. [Google Scholar] [CrossRef]
  99. Abels, E.; Pantanowitz, L.; Aeffner, F.; Zarella, M.D.; van der Laak, J.; Bui, M.M.; Vemuri, V.N.; Parwani, A.V.; Gibbs, J.; Agosto-Arroyo, E.; et al. Computational pathology definitions, best practices, and recommendations for regulatory guidance: A white paper from the Digital Pathology Association. J. Pathol. 2019, 249, 286–294. [Google Scholar] [CrossRef] [Green Version]
  100. Bui, M.M.; Riben, M.W.; Allison, K.H.; Chlipala, E.; Colasacco, C.; Kahn, A.G.; Lacchetti, C.; Madabhushi, A.; Pantanowitz, L.; Salama, M.E.; et al. Quantitative Image Analysis of Human Epidermal Growth Factor Receptor 2 Immunohistochemistry for Breast Cancer: Guideline From the College of American Pathologists. Arch. Pathol. Lab. Med. 2019, 143, 1180–1195. [Google Scholar] [CrossRef]
  101. Lara, H.; Li, Z.; Abels, E.; Aeffner, F.; Bui, M.M.; ElGabry, E.A.; Kozlowski, C.; Montalto, M.C.; Parwani, A.V.; Zarella, M.D.; et al. Quantitative Image Analysis for Tissue Biomarker Use: A White Paper From the Digital Pathology Association. Appl. Immunohistochem. Mol. Morphol. 2021, 29, 479–493. [Google Scholar] [CrossRef] [PubMed]
  102. Hartage, R.; Li, A.C.; Hammond, S.; Parwani, A.V. A Validation Study of Human Epidermal Growth Factor Receptor 2 Immunohistochemistry Digital Imaging Analysis and its Correlation with Human Epidermal Growth Factor Receptor 2 Fluorescence In situ Hybridization Results in Breast Carcinoma. J. Pathol. Inform. 2020, 11, 2. [Google Scholar] [CrossRef]
  103. Li, A.C.; Zhao, J.; Zhao, C.; Ma, Z.; Hartage, R.; Zhang, Y.; Li, X.; Parwani, A.V. Quantitative digital imaging analysis of HER2 immunohistochemistry predicts the response to anti-HER2 neoadjuvant chemotherapy in HER2-positive breast carcinoma. Breast Cancer Res. Treat. 2020, 180, 321–329. [Google Scholar] [CrossRef]
Figure 1. Differential HER2 expression in flat urothelial lesions. CIS: carcinoma in situ.
Figure 1. Differential HER2 expression in flat urothelial lesions. CIS: carcinoma in situ.
Ijms 24 03720 g001
Figure 2. Digital image analysis of HER2-stained BC tissue samples with low (left) and high (right) protein expression (original magnification 200×).
Figure 2. Digital image analysis of HER2-stained BC tissue samples with low (left) and high (right) protein expression (original magnification 200×).
Ijms 24 03720 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sanguedolce, F.; Zanelli, M.; Palicelli, A.; Bisagni, A.; Zizzo, M.; Ascani, S.; Pedicillo, M.C.; Cormio, A.; Falagario, U.G.; Carrieri, G.; et al. HER2 Expression in Bladder Cancer: A Focused View on Its Diagnostic, Prognostic, and Predictive Role. Int. J. Mol. Sci. 2023, 24, 3720. https://doi.org/10.3390/ijms24043720

AMA Style

Sanguedolce F, Zanelli M, Palicelli A, Bisagni A, Zizzo M, Ascani S, Pedicillo MC, Cormio A, Falagario UG, Carrieri G, et al. HER2 Expression in Bladder Cancer: A Focused View on Its Diagnostic, Prognostic, and Predictive Role. International Journal of Molecular Sciences. 2023; 24(4):3720. https://doi.org/10.3390/ijms24043720

Chicago/Turabian Style

Sanguedolce, Francesca, Magda Zanelli, Andrea Palicelli, Alessandra Bisagni, Maurizio Zizzo, Stefano Ascani, Maria Carmela Pedicillo, Angelo Cormio, Ugo Giovanni Falagario, Giuseppe Carrieri, and et al. 2023. "HER2 Expression in Bladder Cancer: A Focused View on Its Diagnostic, Prognostic, and Predictive Role" International Journal of Molecular Sciences 24, no. 4: 3720. https://doi.org/10.3390/ijms24043720

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

Article Metrics

Back to TopTop