You are on page 1of 13

EUROPEAN UROLOGY 70 (2016) 93–105

available at www.sciencedirect.com
journal homepage: www.europeanurology.com

Platinum Priority – Guidelines


Editorial by Rodolfo Montironi, Liang Cheng, Marina Scarpelli and Antonio Lopez-Beltran on pp. 120–123 of
this issue

The 2016 WHO Classification of Tumours of the Urinary System


and Male Genital Organs—Part A: Renal, Penile, and Testicular
Tumours

Holger Moch a,*, Antonio L. Cubilla b, Peter A. Humphrey c,


Victor E. Reuter d, Thomas M. Ulbright e
a
Department of Pathology, University Hospital Zurich, Zurich, Switzerland; b Instituto de Patologı´a e Investigación, Facultad de Ciencias Médicas, Universidad
c d
Nacional de Asunción, San Lorenzo, Paraguay; Department of Pathology, Yale University School of Medicine, New Haven, CT, USA; Department of
e
Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pathology and Laboratory Medicine, Indiana University Health
Partners, Indiana University School of Medicine, Indianapolis, IN, USA

Article info Abstract

Article history: The fourth edition of the World Health Organization (WHO) classification of urogenital
Accepted February 4, 2016 tumours (WHO ‘‘blue book’’), published in 2016, contains significant revisions. These
revisions were performed after consideration by a large international group of pathol-
Associate Editor: ogists with special expertise in this area. A subgroup of these persons met at the WHO
James Catto Consensus Conference in Zurich, Switzerland, in 2015 to finalize the revisions. This
review summarizes the most significant differences between the newly published
classification and the prior version for renal, penile, and testicular tumours. Newly
Keywords: recognized epithelial renal tumours are hereditary leiomyomatosis and renal cell
WHO classification carcinoma (RCC) syndrome–associated RCC, succinate dehydrogenase–deficient RCC,
Male genital organs tubulocystic RCC, acquired cystic disease–associated RCC, and clear cell papillary RCC.
The WHO/International Society of Urological Pathology renal tumour grading system
Urogenital tract was recommended, and the definition of renal papillary adenoma was modified. The
new WHO classification of penile squamous cell carcinomas is based on the presence of
human papillomavirus and defines histologic subtypes accordingly. Germ cell neoplasia
in situ (GCNIS) of the testis is the WHO-recommended term for precursor lesions of
invasive germ cell tumours, and testicular germ cell tumours are now separated into two
fundamentally different groups: those derived from GCNIS and those unrelated to
GCNIS. Spermatocytic seminoma has been designated as a spermatocytic tumour and
placed within the group of non–GCNIS-related tumours in the 2016 WHO classification.
Patient summary: The 2016 World Health Organization (WHO) classification contains
new renal tumour entities. The classification of penile squamous cell carcinomas is
based on the presence of human papillomavirus. Germ cell neoplasia in situ of the testis is
the WHO-recommended term for precursor lesions of invasive germ cell tumours.
# 2016 European Association of Urology. Published by Elsevier B.V. All rights reserved.

* Corresponding author. Department of Pathology, University Hospital Zurich, Schmelzbergstrasse


12, CH-8091 Zurich, Switzerland. Tel. +41 442552500; Fax: +41 442553194.
E-mail address: holger.moch@usz.ch (H. Moch).

http://dx.doi.org/10.1016/j.eururo.2016.02.029
0302-2838/# 2016 European Association of Urology. Published by Elsevier B.V. All rights reserved.
94 EUROPEAN UROLOGY 70 (2016) 93–105

1. The new renal tumour classification RCC), anatomic location of tumours (eg, collecting duct and
renal medullary carcinomas), and correlation with a specific
The Vancouver consensus conference of the International renal disease background (eg, acquired cystic disease–
Society of Urological Pathology (ISUP) provided the associated RCCs) as well as molecular alterations patho-
foundation for much of the 2016 World Health Organization gnomonic for RCC subtypes (eg, MiT family translocation
(WHO) renal tumour classification [1,2] (Fig. 1). The carcinomas and succinate dehydrogenase [SDH]–deficient
revision of the 2004 WHO renal tumour classification renal carcinomas) or familial predisposition syndromes (eg,
was performed after consideration of new knowledge about hereditary leiomyomatosis and RCC [HLRCC] syndrome–
pathology, epidemiology, and genetics [3–5]. associated RCC). In contrast to the 2004 WHO classification,
familial forms of RCC, which also occur in sporadic form (eg,
2. Important changes from existing renal tumour clear cell RCC [ccRCC] in patients with von Hippel-Lindau
types [VHL] syndrome or chromophobe RCC in patients with Birt-
Hogg-Dubé syndrome) are now discussed with the corre-
First, the new 2016 WHO classification refers to subtypes sponding sporadic tumour types in joint chapters.
that have been named on the basis of predominant Second, multiple publications report no recurrence
cytoplasmic features (eg, clear cell and chromophobe renal or metastasis in patients with multilocular cystic RCC
cell carcinomas [RCCs]), architectural features (eg, papillary [6]. Consequently, multilocular cystic renal neoplasm of low
[(Fig._1)TD$IG]

Fig. 1 – World Health Organization (WHO) classification of tumours of the kidney. Reproduced with permission from the WHO International Agency for
Research on Cancer [1].
WHO = World Health Organization.
EUROPEAN UROLOGY 70 (2016) 93–105 95

malignant potential is now the WHO-recommended term for paragangliomas (extrarenal pheochromocytoma). The
this lesion. Such tumours are defined as tumours composed term carcinoid of the kidney is obsolete.
entirely of numerous cysts with low-grade tumour cells
(WHO/ISUP grade 1 or 2). The cysts are lined by a single 3. New renal tumour entities
layer of tumour cells with abundant clear cytoplasm. The
septa contain, at the maximum, groups of clear cells but Over the past decade, several new tumour entities have
without expansile growth. emerged; therefore, the WHO working group was entrusted
Third, the entity of papillary RCC has traditionally been with the responsibility to decide whether enough molecular
subdivided into type 1 and type 2 papillary RCCs [7,8]. It has clinical follow-up data and pathologic data justify the
been accepted that a subset of tumours have mixed recognition of any new distinct tumour entity within the
histology. Recent molecular studies suggest that type classification system. The newly recognized epithelial renal
2 papillary RCCs may constitute not a single well-defined tumours in the 2016 WHO classification are HLRCC-
entity but rather individual subgroups with different associated RCC, SDH-deficient RCC, tubulocystic RCC,
molecular backgrounds [9]. Papillary RCCs with eosinophil- acquired cystic RCC, and clear cell papillary RCC. Paediatric
ic (oncocytic) cytoplasm and oncocytoma-like low-grade cystic nephroma represents a new entity in the group of
nuclei have been called oncocytic papillary RCCs. Because nephroblastic and cystic tumours occurring mainly in
tumours with this morphology have not yet been fully children (as described under ‘‘Important changes’’).
characterized, they are not considered a distinct WHO
entity. The Vancouver consensus conference has recom- 3.1. Hereditary leiomyomatosis and renal cell carcinoma (RCC)
mended diagnosing such tumours as type 2 papillary RCC syndrome–associated RCC
for the time being [2].
Fourth, papillary adenomas were defined until 2015 as HLRCC-associated RCCs are rare tumours occurring in the
tumours measuring 0.5 cm. The WHO 2016 classification setting of nonrenal leiomyomatosis and demonstrate
defines papillary adenomas as unencapsulated tumours with germline fumarate hydratase mutations [12]. The tumours
papillary or tubular architecture, low WHO/ISUP grade, and a have papillary architecture with abundant eosinophilic
diameter 1.5 cm. The decision to increase the cut-off size cytoplasm, large nuclei, and very prominent nucleoli with
was based on available data showing that unencapsulated perinucleolar clearing. The prognosis of these tumours is
grade 1–2 tumours have no capacity to metastasize [10]. It is poor [13].
emphasized, however, that a diagnosis of papillary adenoma
based on needle biopsy should be made with extreme caution 3.2. Succinate dehydrogenase–deficient renal cell carcinoma
because the presence of any capsule or grade heterogeneity
may not be visualized. Current protocols, which define SDH-deficient RCC is composed of vacuolated eosinophilic
papillary adenomas as 0.5 cm in size, state that the or clear cells [14,15]. Immunohistochemistry is a useful tool
presence of papillary adenoma in a donor kidney is not a for their diagnosis because there is a loss of expression of
contraindication for renal transplantation. The new cut-off of SDHB, a marker of dysfunction of mitochondrial complex II
1.5 cm could have a significant impact on some clinical (Fig. 2). It presents mainly in young adults, and most
situations (eg, small renal papillary tumours detected in patients have germline mutations in an SDH gene [15]. Most
transplanted kidneys). tumours are solid, with a brown, sometimes red, cut surface.
Fifth, mixed epithelial and stromal tumours (MEST) The most distinctive feature is the presence of cytoplasmic
encompass a spectrum of tumours including predominantly vacuoles. There are sometimes flocculent inclusions. Most
cystic tumours (adult cystic nephromas) and tumours that SDH-deficient RCC has good prognosis. In cases with
are more solid. Adult cystic nephroma was previously sarcomatoid differentiation and necrosis, the prognosis is
classified, along with paediatric cystic nephroma, as a less favourable.
separate entity from MEST. On the basis of similar age and
sex distributions and a similar histochemical profile, adult 3.3. Tubulocystic renal cell carcinoma
cystic nephroma is now classified within this spectrum of
MEST, and the WHO renal tumour subcommittee recom- Tubulocystic RCC is a dominantly cystic renal epithelial
mended using the term mixed epithelial and stromal tumour neoplasm. Macroscopically, it is composed of multiple small
family for both entities. In contrast to adult cystic nephroma, to medium-sized cysts and has a spongy cut surface. The
paediatric cystic nephroma is a distinct entity with specific nuclei are enlarged with WHO/ISUP grade 3 nucleoli. The
DICER1 mutations [11]. cytoplasm has abundant eosinophilic and oncocytoma-
Sixth, most carcinoids of the kidney have a poor like aspects. Only 4 of 70 reported cases showed metastasis
prognosis, with frequent occurrence of metastasis after to bone, liver, and lymph nodes [16–19].
nephrectomy. The renal tumour subcommittee recom-
mended renal carcinoids should be newly designated as 3.4. Acquired cystic disease–associated renal cell carcinoma
well-differentiated neuroendocrine tumours of the kidney
and simultaneously placed within the group of endocrine Acquired cystic disease–associated RCC occurs in the
tumours encompassing small cell neuroendocrine carci- kidneys of end-stage renal disease and acquired cystic
nomas, large cell neuroendocrine carcinomas, and kidney disease [20]. Histologically, these tumours show a
96 EUROPEAN UROLOGY 70 (2016) 93–105
[(Fig._2)TD$IG] [(Fig._3)TD$IG]

Fig. 2 – (A) Succinate dehydrogenase–deficient renal cell carcinoma


composed of vacuolated cells. (B) Immunohistochemically, there is a
loss of SDHB expression (with positivity in normal tubular cells).

broad spectrum with a cribriform, microcystic, or sieve-


like architecture. They have an eosinophilic and/or clear
cytoplasm and prominent nucleoli. Calcium oxalate crystal
deposition is common. CK7 is typically not expressed. Most
tumours have indolent behaviour.

3.5. Clear cell papillary renal cell carcinoma

Clear cell papillary RCC is a renal epithelial neoplasm


composed of low-grade clear epithelial cells arranged in
Fig. 3 – (A) Clear cell papillary renal cell carcinoma is a renal epithelial
tubules and papillae with a predominantly linear nuclear neoplasm composed of low-grade clear epithelial cells arranged in
alignment away from the basement membrane [20]. They tubules and papillae with a predominantly linear nuclear alignment. (B)
The tumour cells have a characteristic diffuse CK7 positivity.
account for up to 5% of all resected renal tumours and arise
sporadically in end-stage renal disease and VHL syndrome
[21,22]. Some of these tumours were previously referred to
as renal angioadenomatous tumours. The tumour cells have and others were kept as emerging. The WHO classification
characteristic diffuse CK7 positivity and carbonic anhydrase noted that although these entities appear to be distinct, they
IX positivity in a cuplike distribution. CD10 is negative or are rare tumours that are not yet fully characterized by
only focally positive (Fig. 3). According to current knowl- morphology, immunohistochemistry, and molecular stud-
edge, these tumours have indolent behaviour. ies. Consequently, further reports are needed to refine their
diagnostic criteria and established clinical outcomes.
4. Emerging or provisional renal tumour entities SDH-deficient RCC was regarded as an emerging entity in
the Vancouver classification but now is considered to be an
The 2013 ISUP Vancouver classification identified a established entity. RCC in neuroblastoma survivors was
category of emerging or provisional new entities. Some of included in the 2004 WHO classification [23]; however, it is
these emerging entities have been accepted by the WHO, now recognized that some of these tumours represent MiT
EUROPEAN UROLOGY 70 (2016) 93–105 97
[(Fig._4)TD$IG]
seven potential ccRCC tumour suppressor genes: VHL,
PBRM1, BAP1, SETD2, RASSF1A, TU3A, and DLEC1. The
elucidation of the effects of different combinations of
mutations on the initiation and progression of ccRCC will be
an important future area of research [4].
Second, the identification of novel therapeutic agents
that are effective against RCC cells that harbour specific
genetic alterations is an important ongoing research topic
[35]. This area includes emerging immunotherapies target-
ing immune checkpoints and tumour-associated antigens in
patients with RCC [36]. In contrast to other solid tumours
(eg, melanoma or lung cancer), there are presently no
predictive molecular biomarkers suitable for routine use
Fig. 4 – World Health Organization/International Society of Urological [37]. The use of such potential biomarkers must account for
Pathology grading system for clear cell renal cell carcinoma and the considerable genetic intratumoural heterogeneity with
papillary renal cell carcinoma [34].
the parallel evolution of multiple tumour clones [38,39].
Third, there has been a remarkable expansion of
knowledge about the genetics of renal cancer in recent
family translocation RCCs [24], and others are difficult to years. This has led to greater understanding of the
classify based on the published pathologic details. Conse- molecular pathogenesis of renal cancer [40]. Such knowl-
quently, it was decided to remove this entity from the edge will be incorporated into a future WHO classification
2016 WHO classification, although it may be distinct, and to and will clarify the position of some emerging renal tumour
continue to consider it as an emerging entity. Very few entities (eg, TCEB1-mutated RCC and angiomyoadenoma-
thyroid-like follicular RCCs have been described [25,26]. Most tous RCC) or the subgroups of papillary RCC [9,30,32].
of these tumours have indolent behaviour. Fewer than 10 Fourth, the novel WHO/ISUP grading system has been
RCCs associated with ALK gene rearrangements have been validated for ccRCC and papillary RCC but not for other
reported in the literature [27–29]; some are medullary- tumour types [34]. Several grading schemes have been
based tumours. Recently, reports of RCC associated with proposed for chromophobe RCC to predict its behavioural
prominent angioleiomyomatous stroma have been pub- outcome [41–44]. It is important to have an internationally
lished. It is unclear if the renal angiomyoadenomatous accepted chromophobe RCC grading system in the near
tumour represents a variant of ccRCC or clear cell papillary future.
RCC [30]. Some tumours were sporadic, and others were
associated with tuberous sclerosis [31]. A recent report 7. The new classification of penile tumours
identified TCEB1 gene mutations in tumours with this
morphology [32]. The vast majority of malignant tumours of the penis are
squamous cell carcinomas (SCCs) originating in the inner
5. Grading of renal tumours mucosal lining of the glans, coronal sulcus, or foreskin. Most
previous classification schemes were exclusively morphol-
Many grading systems have been proposed for renal cell ogy based. The 2016 WHO classification presents a new
neoplasia. The Fuhrman system was the most frequently classification based on clinicopathologic distinctiveness
used grading system in RCC but should not be applied for and relation to human papillomavirus (HPV) infection
chromophobe RCC [33]. Furthermore, the Fuhrman system (Fig. 5).
has not been validated for most of the new subtypes of renal
carcinoma. For these reasons, the four-tiered WHO/ISUP 7.1. Non–human papillomavirus–related subtypes
grading system is recommended by the WHO [34]. For
grade 1–3 tumours, the system defines tumour grade based Non–HPV-related subtypes of SCC are mainly SCC of the usual
on nucleolar prominence. Grade 4 is defined by the presence type (Fig. 6A). Pseudohyperplastic carcinomas [45] and
of pronounced nuclear pleomorphism, tumour giant cells, pseudoglandular carcinomas [46] are also non-HPV related.
and/or rhabdoid and/or sarcomatoid differentiation (Fig. 4). Pseudohyperplastic carcinomas [45] occur in patients in the
This grading system has been validated for ccRCC and seventh and eighth decades of life and are associated
papillary RCC. It has not yet been validated for other tumour with lichen sclerosus. Pseudoglandular carcinomas [46] are
types because of the small numbers of reported cases. aggressive tumours simulating adenocarcinomas. Verrucous
carcinoma is a nonmetastasizing low-grade neoplasm with
6. Important future issues in renal tumour carcinoma cuniculatum as a variant [47]. Carcinoma cunicu-
pathology latum is a rare low-grade tumour with a labyrinthine growth
pattern with no metastatic potential. Other non–HPV-related
First, the VHL tumour suppressor protein pVHL functions as subtypes of SCC are papillary [48], adenosquamous [49], and
a tumour suppressor via HIF-dependent regulation in most sarcomatoid SCC, the latter with the worst prognosis among
ccRCC. The chromosome 3p locus, however, contains up to all penile carcinomas.
98 EUROPEAN UROLOGY 70 (2016) 93–105
[(Fig._5)TD$IG]

Fig. 5 – World Health Organization classification of tumours of the penis. Reproduced with permission from the World Health Organization
International Agency for Research on Cancer [1].
[(Fig._6)TD$IG]

Fig. 6 – (A) Well differentiated squamous cell carcinoma (SCC; usual type), grade I. Multiple well-delineated invasive tumour nests. (B) Basaloid SCC.
Interanastomosing sheets and nests of basaloid tumour. (C) Basaloid SCC, p16 staining. Dense staining of all tumour cells by p16.

7.2. Human papillomavirus–related carcinomas cell carcinomas [54]. Very unusual HPV-related tumours are
lymphoepithelioma-like [55] and medullary SCC [56].
HPV-related carcinomas are basaloid [50] (Fig. 6B) and warty Penile intraepithelial neoplasia (PeIN) is a precursor lesion
(condylomatous) SCC [51]. Basaloid SCC has a high rate of of invasive SCC showing a penile squamous epithelium
nodal metastasis, whereas warty (condylomatous) carcino- characterized by dysplastic changes with an intact basement
mas [51] are rarely associated with regional nodal membrane. Non–HPV-related PeIN is the so-called differen-
metastasis. Other HPV-related SCCs include warty-basaloid tiated PeIN, whereas basaloid and warty (or mixed basaloid-
[52] and the rare variants of papillary-basaloid [53] and clear warty) PeINs are usually associated with HPV (Fig. 7).
EUROPEAN UROLOGY 70 (2016) 93–105 99
[(Fig._7)TD$IG]

Fig. 7 – Penile intraepithelial neoplasia (PeIN). (A) Differentiated PeIN. There is parakeratosis and elongation of rete ridges. Cells are maturing,
keratinizing; atypical cells predominate at basal layers. Note the changes of lichen sclerosus in lamina propria. (B) Basaloid PeIN. Atypical small
basaloid cells involving full epithelial thickness. (C) Basaloid PeIN, p16 staining. Note the dense ‘‘on block’’ staining with p16.

The three-tiered WHO/ISUP grading system is the lesion is a well-established precursor to an invasive germ
recommended grading system for penile SCC. Well-differ- cell tumour and simultaneously does away with both the
entiated carcinomas (grade 1) have cytologic aspects of misleading epithelial-based nomenclature and the use of
normal squamous tissue. The tumour cells grow in an the word unclassified. It was recognized that IGCNU, and
irregular nesting pattern with little intervening stroma. specifically the unclassified designation, was originally
Poorly differentiated carcinomas (grade 3) have an irregular adopted to distinguish it from differentiated forms of
growth of small tumour cell nests, poor keratinization, intratubular neoplasia [58], most commonly intratubular
polymorphic tumour cells with hyperchromatic nuclei, seminoma and intratubular embryonal carcinoma. There
frequent mitoses, and marked stromal reaction. The cases was concern that GCNIS did not make this distinction, but
that do not qualify as grade 1 or 3 are grade 2. the testis subcommittee made the important point that
GCNIS refers to malignant germ cells that develop in the
8. The new classification of testicular tumours spermatogonial niche (Fig. 9), which is the usual in situ
location for germ cells in the early differentiated testis
Some important changes have been made in the WHO [61]. All other forms of intratubular neoplasia are no longer
classification of testicular tumours in 2016 compared with confined to this location but typically fully occupy the
that adopted in 2004 [3]. These revisions were discussed tubular diameter. GCNIS is now the WHO-recommended
over the course of several months in 2014 through e-mail term for this precursor lesion, and other forms of
communications and were finalized in Zurich, Switzerland, intratubular neoplasia should be referred to by their
in March 2015. The most significant differences between differentiated phenotype with the prefix intratubular.
the newly published classification and the prior version A new emphasis in WHO 2016 is the distinction of GCNIS
concern the germ cell tumours (Fig. 8), but other categories from maturation-delayed germ cells, which are a relatively
are also affected. common feature in the gonads of patients with disorders of
It has been recognized for several decades that the sex development [61–63]. It is believed that maturation-
majority of germ cell tumours arise from progression of an delayed germ cells likely give rise to GCNIS, but they should
intratubular malignant germ cell that has the morphologic be distinguished from the latter because progression is not
and immunohistochemical features of a seminoma cell. invariable. Unfortunately, both lesions express the usual
Such cells were usually designated as either carcinoma in markers used for the identification of GCNIS (eg, OCT3/4,
situ (CIS) [57] or intratubular germ cell neoplasia, unclassified placental alkaline phosphatase, AP-2g). The differentiation
(IGCNU) [58], with those terms receiving preferential use in of the two relies on the more diffuse distribution and central
Europe and North America, respectively. Some others, tubular location of maturation-delayed germ cells (Fig. 10)
however, have been used, including testicular intraepithelial as well as the lack of expression of KIT ligand (stem cell
neoplasia [59] and gonocytoma in situ [60]. The use of factor) in the associated seminiferous tubules, which
different terms for the identical lesion has been a source of contrast with the findings in GCNIS [61,63,64].
some confusion, and neither the two predominant terms The prior version of the WHO classification was purely
nor any of the previously used alternatives were considered morphologically based and divided the germ cell tumours
entirely satisfactory by the testis subcommittee. The main into those of single or more than one histologic type. In so
objection to CIS was that the malignant germ cells to which doing, quite disparate tumours came to be placed under
it referred were not epithelial (a similar objection applied to similar diagnostic terms. The new approach recognizes that
testicular intraepithelial neoplasia), whereas IGCNU, be- there are significantly different pathogeneses for testicular
cause it contained the word unclassified, was felt to falsely germ cell tumours, despite only subtle or even—in the case
convey an element of doubt concerning the nature and of yolk sac tumour of paediatric and adult types—no
clinical behaviour of the lesion. Germ cell neoplasia in situ perceptible morphologic differences. The germ cell tumours
(GCNIS) was proposed as an alternative term, retaining the are now broadly separated into two fundamentally different
in situ nomenclature that correctly conveys the fact that the groups: those derived from GCNIS and those unrelated to
100 EUROPEAN UROLOGY 70 (2016) 93–105
[(Fig._8)TD$IG]

Fig. 8 – World Health Organization classification of germ cell tumours of the testis. Reproduced with permission from the World Health Organization
International Agency for Research on Cancer [1].

GCNIS (Fig. 8). It is apparent, however, that the latter group In making this separation among the germ cell tumours,
is heterogeneous. In contrast, the former group shows a it was necessary to remove entities from the GCNIS-derived
number of basic similarities despite varied morphologies group and to introduce changes in nomenclature. The testis
and, to some extent, behaviours. The GCNIS-derived subcommittee recommended that the entity known as
tumours have comparable epidemiologic associations and spermatocytic seminoma be newly designated as
usually occur in a background of perturbed testicular spermatocytic tumour and simultaneously placed within
development that has recognizable morphologic features: the non-GCNIS related tumours (Fig. 8) because its lack of
impaired spermatogenesis, tubular shrinkage, peritubular association with GCNIS and different molecular features are
sclerosis, immature Sertoli cells, interstitial widening, well established, and it shows no relationship with
hyalinized tubules, and microlithiasis [65,66]. They share seminoma or any other neoplasm in the GCNIS-derived
the finding of amplification of genetic material from the group [67–72]. Because teratomas and yolk sac tumours
short arm of chromosome 12, often in the form of may develop from GCNIS or apart from it, it was
isochromosome 12p, and represent progression from recommended that the GCNIS-derived entities be designat-
GCNIS, often through at least a transient stage of seminoma ed as postpubertal type and the non–GCNIS-related ones be
that may be intratubular. designated as prepubertal type (Fig. 8) because the former
EUROPEAN UROLOGY 70 (2016) 93–105 101
[(Fig._9)TD$IG]

Fig. 9 – (A) Germ cell neoplasia in situ showing confinement of the lesional cells to the base of a seminiferous tubule, the ‘‘spermatogonial niche.’’ In
(B) intratubular seminoma and (C) intratubular embryonal carcinoma, the neoplastic cells have expanded beyond the tubular base and, as shown,
typically occupy most of the tubule.

usually develops in adults and the latter in children. It is with other forms of germ cell tumour. In addition to lacking
recognized, however, that the prepubertal-type tumours GCNIS [77,78], prepubertal-type teratoma and yolk sac
may, rarely, occur in postpubertal patients [73,74], and the tumour also lack 12p amplification and do not occur in
postpubertal-type tumours may occur in paediatric patients malformed testes [79]. The prepubertal-type teratomas show
who have disorders of sex development [75,76]. The evidence no genetic abnormalities, whereas the prepubertal-type yolk
for making these changes is abundant. Spermatocytic tumour sac tumours show characteristic gains and losses of portions
lacks not only association with GCNIS but also 12p of several chromosomes that differ from those frequently
amplification, shows a unique amplification of chromosome occurring in the GCNIS-derived tumours [79]. Although there
[(Fig._10)TD$IG]9 corresponding to the DMRT1 gene, and is never associated are no apparent differences in tumour morphology between
the prepubertal and postpubertal forms of yolk sac tumour,
the prepubertal teratomas—in contrast to the postpubertal
ones—lack any cytologic atypia and are more frequently
organoid with prominent components of smooth muscle and
ciliated and squamous epithelium [73]. The benign behav-
iour of the prepubertal-type teratomas [73,80] contrasts with
that of the postpubertal type [81,82], and the prepubertal-
type yolk sac tumours behave less aggressively than the
postpubertal-type tumours, with a significantly lower
frequency of relapse on surveillance of clinical stage I
patients and of lymphatic-based metastases [83–85].
An additional change that naturally follows the recogni-
tion of the prepubertal-type teratomas is placement of
dermoid cyst, epidermoid cyst, and carcinoid tumour (well-
differentiated neuroendocrine tumour) as specialized forms
of prepubertal-type teratomas (Fig. 8). This is supported by
the absence of GCNIS and 12p amplification in association
with these entities, consistently so for the first two and in the
majority of cases for carcinoid tumour [73,78,86,87],
although some contradictory information in the literature
suggests the possibility of a dual pathogenesis for carcinoid
tumour [88,89]. Additional work is required to resolve this
question.
In the prior WHO classification, the trophoblastic
tumours were divided into choriocarcinoma and nonchor-
iocarcinomatous trophoblastic tumours, the latter repre-
sented by placental-site trophoblastic tumour (PSTT).
So-called monophasic choriocarcinoma was also discussed
in the nonchoriocarcinomatous trophoblastic tumours cate-
gory. The current classification, however, does not separately
recognize monophasic choriocarcinoma but considers it
a morphologic variant of choriocarcinoma. In addition, the
Fig. 10 – (A) An immature testis from a child shows delayed maturation nonchoriocarcinomatous trophoblastic tumour group has
of germ cells, with gonocyte-like cells localized to the central portion of
the tubules (arrows). (B) The nuclei of the gonocyte-like cells are been expanded, recognizing not only PSTT but also epitheli-
highlighted by an immunostain directed against OCT3/4. oid trophoblastic tumour (ETT) and cystic trophoblastic
102 EUROPEAN UROLOGY 70 (2016) 93–105
[(Fig._1)TD$IG]

Fig. 11 – (A) A focus of placental site trophoblastic tumour showing loose clusters of variably sized mononucleated and occasionally multinucleated
cells in a hyalinized, nonhaemorrhagic background. (B) Epithelioid trophoblastic tumour shows cohesive nests of atypical squamoid cells with
admixed apoptotic cells and deposits of fibrinoid material. (C) Cystic trophoblastic tumour in an untreated testis showing the characteristic vacuolated
tumour cells lining a cystic space.

tumour (CTT) (Figs. 8 and 11) [90,91]. Although these entities testis subcommittee to be a variant of mesothelioma that
have most frequently been reported in metastatic sites after tends to behave indolently, but the members noted that
chemotherapy, their de novo development in the testis is progression has been identified in other sites by morpho-
now established. The PSTT consists of intermediate tropho- logically identical tumours [108,109]. In addition, cystic
blast cells that are positive for human placental lactogen mesothelioma, which had also been placed in the benign
(HPL) and negative for p63. They are usually loosely cohesive mesothelioma category, was regarded as either a non-
and tend to invade the walls of blood vessels, provoking a neoplastic condition (mesothelial cysts) [110] or a variant of
fibrinoid reaction. ETT has a more cohesive arrangement of conventional mesothelioma.
squamoid cells, typically displaying apoptotic and fibrinoid
material within cell nests, usually lacks vascular invasion, Author contributions: Holger Moch had full access to all the data in the
and is HPL negative and p63 positive. CTT consists of study and takes responsibility for the integrity of the data and the
frequently vacuolated trophoblast cells that line gaping accuracy of the data analysis.
spaces that may contain eosinophilic material. Based on the
Study concept and design: Moch, Cubilla, Humphrey, Reuter, Ulbright.
available evidence, these lesions are less aggressive than Acquisition of data: Moch, Cubilla, Humphrey, Reuter, Ulbright.
choriocarcinoma, but the data are limited. Analysis and interpretation of data: Moch, Cubilla, Humphrey, Reuter,
In the sex cord–stromal tumours, the sclerosing Sertoli Ulbright.
cell tumour [92,93] is no longer separately classified. These Drafting of the manuscript: Moch, Cubilla, Humphrey, Reuter, Ulbright.
tumours are now considered to be morphologic variants of Critical revision of the manuscript for important intellectual content: Moch,
Sertoli cell tumour, not otherwise specified (NOS), based on the Cubilla, Humphrey, Reuter, Ulbright.
occurrence of CTNNB1 gene mutations and nuclear b-catenin Statistical analysis: None.
Obtaining funding: None.
staining in a similar proportion of both [94–96]. Nonetheless,
Administrative, technical, or material support: None.
it is recommended to continue to use this term for those
Supervision: Moch, Cubilla, Humphrey, Reuter, Ulbright.
Sertoli cell tumours, NOS, that have a hypocellular fibrous
Other (specify): None.
stroma in excess of 50% of the tumour based on their overall
better prognosis than more cellular tumours. Intratubular Financial disclosures: Holger Moch certifies that all conflicts of interest,
large cell hyalinizing Sertoli cell tumour [97] has been added including specific financial interests and relationships and affiliations
to the classification as a distinct entity associated with the relevant to the subject matter or materials discussed in the manuscript
(eg, employment/affiliation, grants or funding, consultancies, honoraria,
Peutz-Jeghers syndrome and as having a characteristic
stock ownership or options, expert testimony, royalties, or patents filed,
mutation of the STK11 gene. Myoid gonadal stromal tumour
received, or pending), are the following: None.
[98–100] is considered an emerging entity characterized by
fusiform cells arranged in short fascicles that coexpress S-100 Funding/Support and role of the sponsor: None[1_TD$IF].
protein and smooth muscle actin.
Acknowledgment: The authors thank Dr. Hiroko Ohgaki and Dr. Paul
Gonadoblastoma [101–103] is now recognized as the
Kleihues for their engagement in the WHO ‘‘Blue Books’’.
only entity in the mixed germ cell–sex cord–stromal
category, with the unclassified form of germ cell–sex
cord–stromal tumour considered not sufficiently estab- References
lished by the available evidence [104,105]. There is
[1] Moch H, Humphrey PA, Ulbright TM, Reuter V. WHO Classifica-
additional emphasis on the recognition of ‘‘undifferentiated
tion of Tumours of the Urinary System and Male Genital Organs.
gonadal tissue’’ as a frequent finding that accompanies Lyon, France: International Agency for Research on Cancer;
gonadoblastoma and is its likely precursor [106,107]. 2016.
In contrast to the prior classification, there is no [2] Srigley JR, Delahunt B, Eble JN, et al. The International Society of
recognized benign mesothelioma category. The well-differ- Urological Pathology (ISUP) Vancouver classification of renal neo-
entiated papillary mesothelioma was considered by the plasia. Am J Surg Pathol 2013;37:1469–89.
EUROPEAN UROLOGY 70 (2016) 93–105 103

[3] Eble JN, Sauter G, Epstein JI, Sesterhenn IA. World Health Organi- [22] Zhou H, Zheng S, Truong LD, Ro JY, Ayala AG, Shen SS. Clear cell
zation Classification of Tumours. Pathology and Genetics of papillary renal cell carcinoma is the fourth most common histo-
Tumours of the Urinary System and Male Genital Organs. Lyon, logic type of renal cell carcinoma in 290 consecutive nephrecto-
France: IARC Press; 2004. mies for renal cell carcinoma. Hum Pathol 2014;45:59–64.
[4] Frew IJ, Moch H. A clearer view of the molecular complexity [23] Medeiros LJ, Palmedo G, Krigman HR, Kovacs G, Beckwith JB.
of clear cell renal cell carcinoma. Annu Rev Pathol 2015;10: Oncocytoid renal cell carcinoma after neuroblastoma: a report
263–89. of four cases of a distinct clinicopathologic entity. Am J Surg Pathol
[5] Moch H. Kidney cancer. In: Stewart BW, Wild CP, editors. World 1999;23:772–80.
Cancer Report 2014. Lyon, France: World Health Organization [24] Argani P, Lae M, Ballard ET, et al. Translocation carcinomas of the
International Agency for Research on Cancer (IARC); 2014. p. 2–9. kidney after chemotherapy in childhood. J Clin Oncol 2006;24:
[6] Eble JN, Bonsib SM. Extensively cystic renal neoplasms: cystic 1529–34.
nephroma, cystic partially differentiated nephroblastoma, multi- [25] Amin MB, Gupta R, Ondrej H, et al. Primary thyroid-like follicular
locular cystic renal cell carcinoma, and cystic hamartoma of renal carcinoma of the kidney: report of 6 cases of a histologically
pelvis. Semin Diagn Pathol 1998;15:2–20. distinctive adult renal epithelial neoplasm. Am J Surg Pathol
[7] Delahunt B, Eble J. Papillary renal cell carcinoma: a clinicopatho- 2009;33:393–400.
logic and immunohistochemical study of 105 tumors. Mod Pathol [26] Berens S, Vogt P, Alkadhi H, Berger N, Moch H. Thyroid-like
1997;10:537–44. follicular carcinoma of the kidney: a separate tumor entity? [in
[8] Jiang F, Richter J, Schraml P, et al. Chromosomal imbalances in German] Pathologe 2014;35:83–7.
papillary renal cell carcinoma: genetic differences between histo- [27] Sukov WR, Hodge JC, Lohse CM, et al. ALK alterations in adult renal
logic subtypes. Am J Pathol 1998;153:1467–73. cell carcinoma: frequency, clinicopathologic features and out-
[9] Cancer Genome Atlas Research Network. Linehan WM, Spellman come in a large series of consecutively treated patients. Mod
PT, Ricketts CJ, et al. Comprehensive molecular characterization of Pathol 2012;25:1516–25.
papillary renal-cell carcinoma. N Engl J Med 2016;374:135–45. [28] Sugawara E, Togashi Y, Kuroda N, et al. Identification of anaplastic
[10] Lohse CM, Gupta S, Cheville JC. Outcome prediction for patients lymphoma kinase fusions in renal cancer: large-scale immuno-
with renal cell carcinoma. Semin Diagn Pathol 2015;32:172–83. histochemical screening by the intercalated antibody-enhanced
[11] Doros LA, Rossi CT, Yang J, et al. DICER1 mutations in childhood polymer method. Cancer 2012;118:4427–36.
cystic nephroma and its relationship to DICER1-renal sarcoma. [29] Debelenko LV, Raimondi SC, Daw N, et al. Renal cell carcinoma
Mod Pathol 2014;27:1267–80. with novel VCL-ALK fusion: new representative of ALK-associated
[12] Tomlinson IP, Alam NA, Rowan AJ, et al. Germline mutations in FH tumor spectrum. Mod Pathol 2011;24:430–42.
predispose to dominantly inherited uterine fibroids, skin leiomyo- [30] Deml KF, Schildhaus HU, Comperat E, et al. Clear cell papillary renal
mata and papillary renal cell cancer. Nat Genet 2002;30:406–10. cell carcinoma and renal angiomyoadenomatous tumor: two var-
[13] Merino MJ, Torres-Cabala C, Pinto P, Linehan WM. The morpho- iants of a morphologic, immunohistochemical, and genetic distinct
logic spectrum of kidney tumors in hereditary leiomyomatosis and entity of renal cell carcinoma. Am J Surg Pathol 2015;39:889–901.
renal cell carcinoma (HLRCC) syndrome. Am J Surg Pathol 2007; [31] Yang P, Cornejo KM, Sadow PM, et al. Renal cell carcinoma in
31:1578–85. tuberous sclerosis complex. Am J Surg Pathol 2014;38:895–909.
[14] Gill AJ, Hes O, Papathomas T, et al. Succinate dehydrogenase [32] Hakimi AA, Tickoo SK, Jacobsen A, et al. TCEB1-mutated renal cell
(SDH)-deficient renal carcinoma: a morphologically distinct carcinoma: a distinct genomic and morphological subtype. Mod
entity: a clinicopathologic series of 36 tumors from 27 patients. Pathol 2015;28:845–53.
Am J Surg Pathol 2014;38:1588–602. [33] Delahunt B, Sika-Paotonu D, Bethwaite PB, et al. Fuhrman grading
[15] Ricketts C, Woodward ER, Killick P, et al. Germline SDHB mutations is not appropriate for chromophobe renal cell carcinoma. Am J
and familial renal cell carcinoma. J Natl Cancer Inst 2008;100: Surg Pathol 2007;31:957–60.
1260–2. [34] Delahunt B, Cheville JC, Martignoni G, et al. The International
[16] Zhou M, Yang XJ, Lopez JI, et al. Renal tubulocystic carcinoma is Society of Urological Pathology (ISUP) grading system for renal
closely related to papillary renal cell carcinoma: implications for cell carcinoma and other prognostic parameters. Am J Surg Pathol
pathologic classification. Am J Surg Pathol 2009;33:1840–9. 2013;37:1490–504.
[17] Amin MB, MacLennan GT, Gupta R, et al. Tubulocystic carcinoma of [35] Moch H, Montironi R, Lopez-Beltran A, Cheng L, Mischo A. Onco-
the kidney: clinicopathologic analysis of 31 cases of a distinctive targets in different renal cancer subtypes. Curr Drug Targets
rare subtype of renal cell carcinoma. Am J Surg Pathol 2009;33: 2015;16:125–35.
384–92. [36] Kucharczyka J, Matrana MR, Santoni M, et al. Emerging immuno-
[18] Yang XJ, Zhou M, Hes O, et al. Tubulocystic carcinoma of the targets in metastatic renal cell carcinoma. Curr Drug Targets
kidney: clinicopathologic and molecular characterization. Am J 2016;17:771–6.
Surg Pathol 2008;32:177–87. [37] Tan PH, Cheng L, Rioux-Leclercq N, et al. Renal tumors: diagnostic
[19] Azoulay S, Vieillefond A, Paraf F, et al. Tubulocystic carcinoma of and prognostic biomarkers. Am J Surg Pathol 2013;37:1518–31.
the kidney: a new entity among renal tumors. Virchows Arch [38] Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity
2007;451:905–9. and branched evolution revealed by multiregion sequencing. N
[20] Tickoo SK, Deperalta-Venturina MN, Harik LR, et al. Spectrum of Engl J Med 2012;366:883–92.
epithelial neoplasms in end-stage renal disease: an experience [39] Gerstung M, Beisel C, Rechsteiner M, et al. Reliable detection of
from 66 tumor-bearing kidneys with emphasis on histologic subclonal single-nucleotide variants in tumour cell populations.
patterns distinct from those in sporadic adult renal neoplasia. Nat Commun 2012;3:811.
Am J Surg Pathol 2006;30:141–53. [40] Comprehensive molecular characterization of clear cell renal cell
[21] Rohan SM, Xiao Y, Liang Y, et al. Clear-cell papillary renal cell carcinoma. Nature 2013;499:43–9.
carcinoma: molecular and immunohistochemical analysis with [41] Finley DS, Shuch B, Said JW, et al. The chromophobe tumor grading
emphasis on the von Hippel-Lindau gene and hypoxia-inducible system is the preferred grading scheme for chromophobe renal
factor pathway-related proteins. Mod Pathol 2011;24:1207–20. cell carcinoma. J Urol 2011;186:2168–74.
104 EUROPEAN UROLOGY 70 (2016) 93–105

[42] Paner GP, Amin MB, Alvarado-Cabrero I, et al. A novel tumor [60] Giwercman A, Skakkebaek NE. Carcinoma-in-situ (gonocytoma-
grading scheme for chromophobe renal cell carcinoma: prognostic in-situ) of the testis. In: Burger H, de Kretser D, editors. The Testis.
utility and comparison with Fuhrman nuclear grade. Am J Surg ed 2. New York, NY: Raven Press; 1989. p. 475–91.
Pathol 2010;34:1233–40. [61] Cools M, van Aerde K, Kersemaekers AM, et al. Morphological and
[43] Cheville JC, Lohse CM, Sukov WR, Thompson RH, Leibovich BC. immunohistochemical differences between gonadal maturation
Chromophobe renal cell carcinoma: the impact of tumor grade on delay and early germ cell neoplasia in patients with underviriliza-
outcome. Am J Surg Pathol 2012;36:851–6. tion syndromes. J Clin Endocrinol Metab 2005;90:5295–303.
[44] Amin MB, Paner GP, Alvarado-Cabrero I, et al. Chromophobe renal [62] Kaprova-Pleskacova J, Stoop H, Bruggenwirth H, et al. Complete
cell carcinoma: histomorphologic characteristics and evaluation androgen insensitivity syndrome: factors influencing gonadal
of conventional pathologic prognostic parameters in 145 cases. histology including germ cell pathology. Mod Pathol 2014;27:
Am J Surg Pathol 2008;32:1822–34. 721–30.
[45] Cubilla AL, Velazquez EF, Young RH. Pseudohyperplastic squa- [63] Rajpert-De Meyts E, Jorgensen N, Brondum-Nielsen K, Muller J,
mous cell carcinoma of the penis associated with lichen sclerosus. Skakkebaek NE. Developmental arrest of germ cells in the patho-
An extremely well-differentiated, nonverruciform neoplasm that genesis of germ cell neoplasia. APMIS 1998;106:198–204.
preferentially affects the foreskin and is frequently misdiagnosed: [64] Stoop H, Honecker F, van de Geijn GJ, et al. Stem cell factor as a
a report of 10 cases of a distinctive clinicopathologic entity. Am J novel diagnostic marker for early malignant germ cells. J Pathol
Surg Pathol 2004;28:895–900. 2008;216:43–54.
[46] Cunha IW, Guimaraes GC, Soares F, et al. Pseudoglandular (ade- [65] Hoei-Hansen CE, Holm M, Rajpert-De Meyts E, Skakkebaek NE.
noid, acantholytic) penile squamous cell carcinoma: a clinicopath- Histological evidence of testicular dysgenesis in contralateral
ologic and outcome study of 7 patients. Am J Surg Pathol 2009; biopsies from 218 patients with testicular germ cell cancer. J
33:551–5. Pathol 2003;200:370–4.
[47] Barreto JE, Velazquez EF, Ayala E, Torres J, Cubilla AL. Carcinoma [66] Hoei-Hansen CE, Rajpert-De Meyts E, Daugaard G, Skakkebaek NE.
cuniculatum: a distinctive variant of penile squamous cell carci- Carcinoma in situ testis, the progenitor of testicular germ cell
noma: report of 7 cases. Am J Surg Pathol 2007;31:71–5. tumours: a clinical review. Ann Oncol 2005;16:863–8.
[48] Chaux A, Soares F, Rodriguez I, et al. Papillary squamous cell [67] Talerman A. Spermatocytic seminoma: clinicopathological study
carcinoma, not otherwise specified (NOS) of the penis: clinico- of 22 cases. Cancer 1980;45:2169–76.
pathologic features, differential diagnosis, and outcome of [68] Muller J, Skakkebaek NE, Parkinson MC. The spermatocytic semi-
35 cases. Am J Surg Pathol 2010;34:223–30. noma: views on pathogenesis. Int J Androl 1987;10:147–56.
[49] Cubilla AL, Ayala MT, Barreto JE, Bellasai JG, Noel JC. Surface [69] Verkerk AJ, Ariel I, Dekker MC, et al. Unique expression patterns of
adenosquamous carcinoma of the penis. A report of three cases. H19 in human testicular cancers of different etiology. Oncogene
Am J Surg Pathol 1996;20:156–60. 1997;14:95–107.
[50] Cubilla AL, Reuter VE, Gregoire L, et al. Basaloid squamous cell [70] Kraggerud SM, Berner A, Bryne M, Pettersen EO, Fossa SD. Sperma-
carcinoma: a distinctive human papilloma virus-related penile tocytic seminoma as compared to classical seminoma: an immu-
neoplasm: a report of 20 cases. Am J Surg Pathol 1998;22:755–61. nohistochemical and DNA flow cytometric study. APMIS 1999;
[51] Cubilla AL, Velazques EF, Reuter VE, Oliva E, Mihm Jr MC, Young 107:297–302.
RH. Warty (condylomatous) squamous cell carcinoma of the [71] Looijenga LH, Hersmus R, Gillis AJ, et al. Genomic and expression
penis: a report of 11 cases and proposed classification of ‘verruci- profiling of human spermatocytic seminomas: primary spermato-
form’ penile tumors. Am J Surg Pathol 2000;24:505–12. cyte as tumorigenic precursor and DMRT1 as candidate chromo-
[52] Chaux A, Tamboli P, Ayala A, et al. Warty-basaloid carcinoma: some 9 gene. Cancer Res 2006;66:290–302.
clinicopathological features of a distinctive penile neoplasm. Re- [72] Goriely A, Hansen RM, Taylor IB, et al. Activating mutations in
port of 45 cases. Mod Pathol 2010;23:896–904. FGFR3 and HRAS reveal a shared genetic origin for congenital
[53] Cubilla AL, Lloveras B, Alemany L, et al. Basaloid squamous cell disorders and testicular tumors. Nat Genet 2009;41:1247–52.
carcinoma of the penis with papillary features: a clinicopathologic [73] Zhang C, Berney DM, Hirsch MS, Cheng L, Ulbright TM. Evidence
study of 12 cases. Am J Surg Pathol 2012;36:869–75. supporting the existence of benign teratomas of the postpubertal
[54] Sanchez DF, Rodriguez IM, Piris A, et al. Clear cell carcinoma of the testis: a clinical, histopathologic, and molecular genetic analysis of
penis. A human papillomavirus (HPV) related variant of squamous 25 cases. Am J Surg Pathol 2013;37:827–35.
cell carcinoma. A report of 3 cases. Am J Surg Pathol. In press. [74] Oosterhuis JW, Stoop JA, Rijlaarsdam MA, et al. Pediatric germ cell
[55] Mentrikoski MJ, Frierson Jr HF, Stelow EB, Cathro HP. Lymphoe- tumors presenting beyond childhood? Andrology 2015;3:70–7.
pithelioma-like carcinoma of the penis: association with human [75] Scully RE. Neoplasia associated with anomalous sexual develop-
papilloma virus infection. Histopathology 2014;64:312–5. ment and abnormal sex chromosomesIn: Jass J, editor. The
[56] Cañete S, Clavero O, Sanchez DF, et al. Poorly differentiated solid Intersex Child Pediatric Adolescent Endocrinology, vol 8. Basel,
(medullary) carcinoma: a distinctive HPV-related penile neo- Switzerland: Karger; 1981. p. 203–17.
plasm. A report of 12 cases. Mod Pathol. In press. [76] Muller J, Skakkebaek NE, Ritzen M, Ploen L, Petersen KE. Carcino-
[57] Skakkebaek NE. Carcinoma in situ of the testis: frequency and ma in situ of the testis in children with 45,X/46,XY gonadal
relationship to invasive germ cell tumours in infertile men. His- dysgenesis. J Pediatr 1985;106:431–6.
topathology 1978;2:157–70. [77] Manivel JC, Simonton S, Wold LE, Dehner LP. Absence of intra-
[58] Scully RE. Intratubular germ cell neoplasia (carcinoma in situ). tubular germ cell neoplasia in testicular yolk sac tumors in chil-
What is it and what should be done about itIn: Fraley EE, editor. dren. A histochemical and immunohistochemical study. Arch
World Urology Update Series (lesson 17), vol. 1. Princeton, NJ: Pathol Lab Med 1988;112:641–5.
Continuing Professional Education Center; 1982. p. 1–8. [78] Manivel JC, Reinberg Y, Niehans GA, Fraley EE. Intratubular germ
[59] Dieckmann KP, Loy V. Prevalence of bilateral testicular germ cell cell neoplasia in testicular teratomas and epidermoid cysts. Cor-
tumors and early detection by testicular intraepithelial neoplasia. relation with prognosis and possible biologic significance. Cancer
Eur Urol 1993;23(Suppl 2):22–3. 1989;64:715–20.
EUROPEAN UROLOGY 70 (2016) 93–105 105

[79] Mostert M, Rosenberg C, Stoop H, et al. Comparative genomic and [95] Colecchia M. Observations on the paper ‘‘sclerosing Sertoli cell
in situ hybridization of germ cell tumors of the infantile testis. Lab tumor of the testis: a clinicopathologic study of 20 cases’’ by Kao
Invest 2000;80:1055–64. et al. Am J Surg Pathol 2014;38:1160.
[80] Visfeldt J, Jorgensen N, Muller J, Moller H, Skakkebaek NE. Testic- [96] Perrone F, Bertolotti A, Montemurro G, Paolini B, Pierotti MA,
ular germ cell tumours of childhood in Denmark, 1943–1989: Colecchia M. Frequent mutation and nuclear localization of be-
incidence and evaluation of histology using immunohistochemi- ta-catenin in sertoli cell tumors of the testis. Am J Surg Pathol
cal techniques. J Pathol 1994;174:39–47. 2014;38:66–71.
[81] Leibovitch I, Foster RS, Ulbright TM, Donohue JP. Adult primary [97] Ulbright TM, Amin MB, Young RH. Intratubular large cell hyalinizing
pure teratoma of the testis. The Indiana experience. Cancer sertoli cell neoplasia of the testis: a report of 8 cases of a distinctive
1995;75:2244–50. lesion of the Peutz-Jeghers syndrome. Am J Surg Pathol 2007;31:
[82] Simmonds PD, Lee AH, Theaker JM, Tung K, Smart CJ, Mead GM. 827–35.
Primary pure teratoma of the testis. J Urol 1996;155:939–42. [98] Kao CS, Ulbright TM. Myoid gonadal stromal tumor: a clinicopath-
[83] Cornejo KM, Frazier L, Lee RS, Kozakewich HP, Young RH. Yolk sac ologic study of three cases of a distinctive testicular tumor. Am J
tumor of the testis in infants and children: a clinicopathologic Clin Pathol 2014;142:675–82.
analysis of 33 cases. Am J Surg Pathol 2015;39:1121–31. [99] Weidner N. Myoid gonadal stromal tumor with epithelial differen-
[84] Grady RW, Ross JH, Kay R. Patterns of metastatic spread in tiation (? testicular myoepithelioma) Ultrastruct Pathol 1991;15:
prepubertal yolk sac tumor of the testis. J Urol 1995;153: 409–16.
1259–61. [100] Du S, Powell J, Hii A, Weidner N. Myoid gonadal stromal tumor: a
[85] Foster RS, Hermans B, Bihrle R, Donohue JP. Clinical stage I pure distinct testicular tumor with peritubular myoid cell differentia-
yolk sac tumor of the testis in adults has different clinical behavior tion. Hum Pathol 2012;43:144–9.
than juvenile yolk sac tumor. J Urol 2000;164:1943–4. [101] Scully RE. Gonadoblastoma. A review of 74 cases. Cancer 1970;
[86] Dieckmann KP, Loy V. Epidermoid cyst of the testis: a review of 25:1340–56.
clinical and histogenetic considerations. Br J Urol 1994;73:436–41. [102] Ulbright TM. Gonadoblastoma and hepatoid and endometrioid-like
[87] Wang WP, Guo C, Berney DM, et al. Primary carcinoid tumors of yolk sac tumor: an update. Int J Gynecol Pathol 2014;33:365–73.
the testis: a clinicopathologic study of 29 cases. Am J Surg Pathol [103] Ulbright TM, Young RH. Gonadoblastoma and selected other
2010;34:519–24. aspects of gonadal pathology in young patients with disorders
[88] Abbosh PH, Zhang S, Maclennan GT, et al. Germ cell origin of of sex development. Semin Diagn Pathol 2014;31:427–40.
testicular carcinoid tumors. Clin Cancer Res 2008;14:1393–6. [104] Michal M, Vanecek T, Sima R, et al. Mixed germ cell sex cord-
[89] Merino J, Zuluaga A, Gutierrez-Tejero F, Del Mar Serrano M, Ciani S, stromal tumors of the testis and ovary. Morphological, immuno-
Nogales FF. Pure testicular carcinoid associated with intratubular histochemical, and molecular genetic study of seven cases. Virch-
germ cell neoplasia. J Clin Pathol 2005;58:1331–3. ows Arch 2006;448:612–22.
[90] Idrees MT, Kao CS, Epstein JI, Ulbright TM. Nonchoriocarcinoma- [105] Ulbright TM, Srigley JR, Reuter VE, Wojno K, Roth LM, Young RH.
tous trophoblastic tumors of the testis: the widening spectrum of Sex cord-stromal tumors of the testis with entrapped germ cells: a
trophoblastic neoplasia. Am J Surg Pathol 2015;39:1468–78. lesion mimicking unclassified mixed germ cell sex cord-stromal
[91] Ulbright TM, Henley JD, Cummings OW, Foster RS, Cheng L. Cystic tumors. Am J Surg Pathol 2000;24:535–42.
trophoblastic tumor: a nonaggressive lesion in postchemotherapy [106] Beaulieu Bergeron M, Lemieux N, Brochu P. Undifferentiated
resections of patients with testicular germ cell tumors. Am J Surg gonadal tissue, Y chromosome instability, and tumors in XY
Pathol 2004;28:1212–6. gonadal dysgenesis. Pediatr Dev Pathol 2011;14:445–59.
[92] Kao CS, Kum JB, Idrees MT, Ulbright TM. Sclerosing Sertoli cell [107] Cools M, Stoop H, Kersemaekers AM, et al. Gonadoblastoma
tumor of the testis: a clinicopathologic study of 20 cases. Am J Surg arising in undifferentiated gonadal tissue within dysgenetic
Pathol 2014;38:510–7. gonads. J Clin Endocrinol Metab 2006;91:2404–13.
[93] Zukerberg LR, Young RH, Scully RE. Sclerosing Sertoli cell tumor [108] Galateau-Salle F, Vignaud JM, Burke L, et al. Well-differentiated
of the testis. A report of 10 cases. Am J Surg Pathol 1991;15: papillary mesothelioma of the pleura: a series of 24 cases. Am J
829–34. Surg Pathol 2004;28:534–40.
[94] Zhang C, Ulbright TM. Nuclear localization of beta-catenin in [109] Churg A, Allen T, Borczuk AC, et al. Well-differentiated papillary
Sertoli cell tumors and other sex cord-stromal tumors of the testis: mesothelioma with invasive foci. Am J Surg Pathol 2014;38:990–8.
an immunohistochemical study of 87 cases. Am J Surg Pathol [110] Ross MJ, Welch WR, Scully RE. Multilocular peritoneal inclusion
2015;39:1390–4. cysts (so-called cystic mesotheliomas). Cancer 1989;64:1336–46.

You might also like