You are on page 1of 15

Seminars in Diagnostic Pathology 34 (2017) 438–452

Contents lists available at ScienceDirect

Seminars in Diagnostic Pathology


journal homepage: www.elsevier.com/locate/serndb

Fibroepithelial lesions; The WHO spectrum


Gregor Krings n, Gregory R. Bean, Yunn-Yi Chen
Department of Pathology, University of California San Francisco (UCSF), San Francisco, CA, USA

ar t ic l e i nf o a b s t r a c t

Fibroepithelial lesions of the breast comprise a morphologically and biologically heterogeneous group of
Keywords: biphasic tumors with epithelial and stromal components that demonstrate widely variable clinical be-
Phyllodes havior. Fibroadenomas are common benign tumors with a number of histologic variants, most of which
Fibroepithelial pose no diagnostic challenge. Cellular and juvenile fibroadenomas can have overlapping features with
Fibroadenoma phyllodes tumors and should be recognized. Phyllodes tumors constitute a spectrum of lesions with
Sarcoma varying clinical behavior and are graded as benign, borderline or malignant based on a set of histologic
Breast features according to recommendations by the World Health Organization (WHO). Recent developments
have significantly expanded our understanding of the pathogenesis of fibroepithelial lesions, highlighting
fibroadenomas as true neoplasms and underscoring a commonality with phyllodes tumors in the form of
recurrent MED12 exon 2 mutations. In addition, sequencing studies have elucidated pathways associated
with phyllodes tumor progression. Accurate diagnosis and grading of phyllodes tumors are important for
patient management and prognosis, as grade broadly correlates with increasing local recurrence risk, and
essentially only malignant tumors metastasize. However, classification of fibroepithelial lesions in many
cases remains challenging on both core biopsy and excision specimens. A commonly encountered pro-
blem at the benign end of the spectrum is the distinction of benign phyllodes tumor from cellular fi-
broadenoma, which is largely due to the subjective nature of histologic features used in diagnosis and
histologic overlap between lesions. Grading is further complicated by the requirement to integrate
multiple subjective and ill-defined parameters. On the opposite end of the histologic spectrum, malig-
nant phyllodes tumors must be distinguished from more common metaplastic carcinomas and from
primary or metastatic sarcomas, which can be especially difficult in core biopsies. Im-
munohistochemistry can be useful in the differential diagnosis but should be interpreted with attention
to caveats. This review provides an overview and update on the spectrum of fibroepithelial lesions, with
special emphasis on common problems and practical issues in diagnosis.
& 2017 Elsevier Inc. All rights reserved.

Introduction fibroadenomas and a commonality with phyllodes tumors in the


form of recurrent MED12 mutations, and also highlight the hetero-
Fibroepithelial lesions of the breast constitute a heterogeneous geneity and molecular progression of phyllodes tumors.
group of biphasic tumors with stromal and epithelial components Whereas the diagnosis of usual type fibroadenomas is typically
that demonstrate wide ranging biologic behavior and differences in straightforward, accurate diagnosis of phyllodes tumors can be
clinical management. These include common benign fibroadenomas challenging in many cases. Commonly encountered problems in-
and fibroadenoma variants, as well as the spectrum of rare phyllodes clude phyllodes tumor grading and distinction of cellular fi-
tumors, ranging from benign tumors with variable recurrence po- broadenomas from the lower end of the histologic spectrum of
tential to frankly malignant tumors with risk of metastasis and phyllodes tumors. The diagnostic difficulty arises in large part due
death.1 Periductal stromal tumors likely represent phyllodes tumor to overlapping histologic features and the subjective nature of the
variants with periductal growth and are also included in this review. assessed histologic parameters. In addition, phyllodes tumor
Although the pathogenesis of fibroepithelial lesions remains poorly grading is further complicated by the need to evaluate and in-
understood overall, recent genomic studies have begun to uncover tegrate multiple parameters whose relative weighted importance
the molecular underpinnings of fibroadenomas and phyllodes can be observer dependent in the absence of clearly defined cri-
tumors.2–7 These studies provide support for a clonal origin of teria. Nonetheless, accurate diagnosis is important to help predict

n
Correspondence to: Department of Pathology, University of California San Francisco, 1825 4th Street, Room M-2355, San Francisco, CA 94143, USA.
E-mail address: gregor.krings@ucsf.edu (G. Krings).

http://dx.doi.org/10.1053/j.semdp.2017.05.006
0740-2570/& 2017 Elsevier Inc. All rights reserved.
G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452 439

recurrence potential and guide clinical management. On the ma- features.28,30 On the other hand, a diagnosis of phyllodes tumor
lignant end of the spectrum, distinction of malignant phyllodes should be carefully considered in mitotically active cellular fi-
tumors from metaplastic carcinoma or sarcoma can be challenging broepithelial lesions presenting in women significantly past ado-
on core biopsy and excision, in which generous sampling and lescence. Pediatric fibroadenomas may also demonstrate ill-de-
immunohistochemistry can be useful to reach the correct diag- fined borders and often have small leaf-like fronds, although the
nosis. Here, we provide a review on fibroepithelial lesions, in- latter should not be diffuse or well-developed for this diagnosis.
cluding recent advances in molecular pathogenesis, and with an While mitotic activity 4 2/10 HPF correlated with recurrence in
emphasis on problems and practical issues in diagnosis. one recent study on pediatric fibroepithelial lesions, stromal cel-
lularity, border irregularity and frond-like growth in the absence
of other features of phyllodes tumor did not.30
Fibroadenomas
Practical problems and challenges in diagnosis
Fibroadenomas are the most common fibroepithelial lesions
and most common benign tumors of the breast. The incidence is The most challenging differential diagnosis is between cellular
highest in women less than 30 years old, but these tumors are not fibroadenoma and phyllodes tumor, which is due to overlapping
uncommon in older age groups.1,8,9 Fibroadenomas are often small and subjective histologic features and ill-defined criteria, as well
(o 3 cm) but may be large and even rapidly growing, especially as tumor heterogeneity and sampling issues in core biopsies; this
juvenile fibroadenomas, which can raise clinical concern for is discussed in more detail below. In contrast, the diagnosis of a
phyllodes tumor.1,10–12 An increased prevalence in younger wo- usual type fibroadenoma is often facile if typical features are
men, tendency to regress or hyalinize with age, increased risk of present. Indeed, a diagnosis of fibroadenoma can be confidently
development with estrogen replacement therapy, and an associa- made on core biopsy if the lesion lacks stromal fronds, stromal
tion with gynecomastia or hormonal imbalance in men all suggest hypercellularity, atypia or mitotic activity. In this context, the al-
a role for hormonal influence in fibroadenoma development and ternate diagnosis of phyllodes tumor on excision is o1% despite
growth.10,13–17 Cyclosporine use in organ transplant patients has the heterogeneity of many phyllodes tumors.34,35 Some fi-
been associated with multiple and/or bilateral fibroadenomas.18–20 broadenomas may show focal leaf-like architecture; if focal and
Genetic risk factors remain unknown, but multiple and/or bilateral lacking hypercellular stroma, this should not be misinterpreted as
fibroadenomas have been associated with a family history, and evidence for phyllodes tumor. Benign lipomatous metaplasia may
Carney complex patients may develop multiple or bilateral myxoid raise concern for infiltrative growth, especially on biopsy; low
fibroadenomas.21,22 stromal cellularity and lesional circumscription is often the clue to
Fibroadenomas are well-circumscribed epithelial and stromal the correct diagnosis in this context (see Fig. 6F). Fibroadenomas
proliferations derived from the terminal duct lobular unit (Fig. 1A). with lipomatous metaplasia may also mimic hamartomas, but this
Growth may be intracanalicular or pericanalicular, and both pat- distinction is not clinically relevant. In limited and hypocellular
terns often coexist (Fig. 1B-C). In usual type fibroadenomas, stro- biopsy material, myxoid fibroadenomas may resemble mucinous
mal cellularity is similar to that of normal perilobular stroma, and carcinomas (Fig. 3A–B), which are also radiologically similar.36
there is no stromal atypia and no or only very widely scattered Mucin of mucinous carcinomas is often more fibrillary and blue-
mitotic activity. The stroma may be fibrous, myxoid (Fig. 1D), tinged than the homogenous white-tan stroma of myxoid fi-
hyalinized (Fig. 1E), or mixed. Benign heterologous stroma is rare broadenomas. Level sections or immunohistochemistry for
but most often lipomatous, with myoid, cartilaginous or osseous myoepithelial cells may be helpful in challenging cases. Un-
metaplasia being exceedingly rare. Fibroadenomas with sclerosing commonly, the stroma of higher grade phyllodes tumors can also
adenosis, papillary apocrine metaplasia, cysts or epithelial calcifi- be paradoxically hypocellular and myxoid-appearing. Rarely, fi-
cations have been called complex fibroadenomas (Fig. 1F) and may broadenomas with cyst-like spaces from stromal retraction can
be associated with slightly higher risk of subsequent carcinoma mimic intraductal papillomas, and vice versa (Fig. 3C–D). On core
development, but this term is not recommended in routine biopsy, sclerosing adenosis, cysts, apocrine change or epithelial
practice.23–25 Tubular adenomas are characterized by adenosis-like proliferation may mask an underlying fibroepithelial lesion
proliferations of closely packed ductules with small lumina that (Fig. 3E), and sclerosing lesions with fibroglandular distortion and
may have scant secretions (Fig. 1G); identification of MED12 mu- altered stroma may conversely mimic complex fibroadenoma.
tations in these lesions supports their classification as fi- Tubular adenomas may resemble ductal adenomas or intraductal
broadenoma variants with epithelial overgrowth 26, although papillomas with adenosis; the ductules of tubular adenomas are
others have found no such association.27 Some fibroadenoma round to oval, in contrast to the irregular jagged ductules em-
variants demonstrate a more lobular rather than ductal archi- bedded in hyalinized stroma of sclerosed papillomas/ductal ade-
tecture, and this is more common in juvenile fibroadenomas.28 nomas. Fibroadenomas can rarely infarct sporadically or during
Cellular fibroadenomas are defined by mild to moderately in- pregnancy, lactation or following biopsy procedures, which should
creased stromal cellularity, although this feature is subjective and not necessarily be misinterpreted as a worrisome feature
lacking a diagnostic threshold (Fig. 2A–C). Juvenile fibroadenomas (Fig. 3F).37–40
are cellular fibroadenomas additionally characterized by perica-
nalicular growth and epithelial “gynecomastoid”/micropapillary Pathogenesis and molecular pathology
usual ductal hyperplasia (Fig. 2D–F). Juvenile fibroadenomas are
more common in adolescents and young women, but prevalence is The pathogenesis of fibroadenomas overall remains poorly
bimodal, with a second peak in women 4 40 years old.11,28–30 understood, but epithelial-stromal interactions are thought to be
Stromal mitoses may be increased in cellular fibroadenomas, with central to their development and to the development and growth
some suggesting an upper threshold of 2 mitotic figures/10 high of fibroepithelial lesions in general. Sawyer et al. postulated a role
power fields (HPF).28,30–33 Although still averaging o2 mitotic for stromal insulin-like growth factor (IGF) 1 and IGF-2 signaling
figures per 10 HPF in most cases, a subset of usual type and ju- through β-catenin.41 Along with epidemiologic and clin-
venile fibroadenomas in adolescents may be more mitotically ac- icopathologic evidence, a role for hormonal influence is supported
tive (up to 6 and 7 mitoses/10 HPF, respectively), which is not by stromal ERβ expression and recurrent somatic mutations in
diagnostic of phyllodes tumor in the absence of other histologic MED12, a component of the multiprotein transcriptional regulator
440 G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452

Fig. 1. Fibroadenoma and Fibroadenoma Variants. (A) Fibroadenomas are well-circumscribed biphasic tumors with epithelial and stromal components. (B) Intracanalicular
growth pattern, with compression of ducts into elongated curvilinear structures (C) Pericanalicular growth pattern, with circumferential stromal proliferation around non-
compressed ducts with open lumina. (D) Myxoid fibroadenoma (E) Hyalinized fibroadenoma, with dense sclerotic stroma and associated calcifications (F) Complex fi-
broadenoma, in this case showing epithelial cysts and foci of sclerosing adenosis. (G) Tubular adenomas are characterized by adenosis-like proliferations of closely-packed
ductules, often with luminal secretions (inset). (H) Lactational change may be seen during pregnancy or lactation.
G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452 441

Fig. 2. Cellular and Juvenile Fibroadenomas. (A-C) Cellular fibroadenoma (A, B) Well-circumscribed cellular fibroepithelial lesion with uniform distribution of epithelium and
stroma (C) High power magnification shows mild to moderately cellular stroma without significant nuclear atypia. (D–F) Juvenile fibroadenoma (D) Peripheral circum-
scription, pericanalicular growth pattern, uniformly distributed epithelium, and mildly proliferative epithelium are evident on low power evaluation. High power view
reveals (E) mild stromal cellularity without atypia and (F) characteristic gynecomastoid-type/micropapillary usual ductal hyperplasia of epithelial component.

Mediator complex that shows a similar spectrum of mutations in receptor α (RARA) gene have also been identified in fibroadenomas
uterine leiomyomas.2,10,13–17,26,42–47 Mutations in exon 2 of MED12 and phyllodes tumors and were shown to impact RARA-mediated
have been identified across the spectrum of breast fibroepithelial transcription.3 The identification of recurrent stromal MED12 and
lesions, including up to 65% of fibroadenomas, with the vast ma- RARA mutations is indicative of monoclonality and supports the
jority of these occurring at codon 44.2,26,45,46 MED12 mutations classification of at least some fibroadenomas as neoplastic rather
were shown to be confined to the microdissected stromal than hyperplastic lesions. This is in contrast to some earlier studies
component,2 and mutation status correlates with stromal but not that used comparative genomic hybridization and polymerase
epithelial MED12 protein expression.47 MED12 mutations do not chain reaction (PCR)-based approaches to suggest a polyclonal
clearly correlate with stromal or epithelial ERα/ERβ expression, stromal origin.49–52 The apparent discrepancy can likely be ex-
indicating that the proposed functional impact on ER signaling plained at least in part by low lesional stromal cellularity that can
requires additional study.47 MED12 mutations may be more fre- mask clonality in the latter assays. Indeed, fibroadenomas with
quently associated with intracanalicular growth.45,48 Mutations more cellular stroma or “phyllodal features” were shown to be
clustering in the ligand-binding domain of the retinoic acid clonal using these techniques.51–53 Interestingly, a recent study
442 G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452

Fig. 3. Histologic Mimics of Fibroadenoma. (A) Myxoid fibroadenoma and (B) mucinous carcinoma may sometimes mimic each other, especially in scant core biopsy spe-
cimens. (C) Gaping ducts in fibroadenoma with stromal retraction can sometimes mimic intraductal papilloma if the intracanalicular growth pattern is not recognized (inset),
especially in core biopsies. (D) Conversely, fragmentation of a biopsied intraductal papilloma may sometimes mimic features of a fibroepithelial lesion. (E) Sclerosing
adenosis, variably dilated cysts and stromal fibrosis of complex fibroadenomas can mimic a sclerosing lesion on core biopsy. The presence of elongated curvilinear ducts
(bottom left) can be a clue to the correct diagnosis. (F) Necrosis (shown here in a core biopsy of fibroadenoma) can be seen in fibroepithelial lesions due to infarction during
pregnancy or lactation, following a prior procedure, or spontaneously, and is not necessarily worrisome in the absence of other features of phyllodes tumor.

found that myxoid fibroadenomas did not harbor MED12 muta- and 10–20%, respectively.1,55,56 Phyllodes tumors often present as a
tions. Although limited in number of cases, this report potentially rapidly growing mass or accelerated growth of a previously stable
suggests an alternate pathogenesis for these variants.54 lesion, and growth rate may be demonstrably faster than fi-
broadenomas by ultrasound.57 However, phyllodes tumors cannot
be reliably distinguished from fibroadenomas by imaging.58–61
Phyllodes tumors Average tumor size is 4–5 cm, but size ranges widely from sub-
centimeter to 430 cm.55,62–68 Large tumors may cause skin dis-
Phyllodes tumors are rare, comprising 2.5% of fibroepithelial coloration, thinning, or ulceration.1,64,67 Although axillary lympha-
lesions and 0.3–1% of all primary breast tumors.1,31 These tumors denopathy is common, nodal metastases are very rare.69 Age at
have inherent recurrence and/or metastatic potential, which varies presentation (40–50 years) is on average 10–20 years older than
based on histologic grade.1 The majority of tumors (60–75%) are fibroadenomas, but these tumors can present at any
benign, with borderline and malignant tumors constituting 15–20% age.1,28,30,63,64,70,71 Phyllodes tumors are more common and may
G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452 443

present at earlier age in Asian and Latina women.1,72 Rare reports in epithelium (Fig. 4C).1 Mitotic activity may be increased in this
men are often associated with gynecomastia, suggesting a role for zone. The stroma is usually fibrous, myxoid, or hyalinized and not
hormonal imbalance.73–75 Genetic risk factors are largely unknown, infrequently shows heterogeneity in terms of cellularity, matrix
but phyllodes tumors have been described in Li-Fraumeni syn- type and epithelial density. Pseudoangiomatous stromal hyper-
drome patients and in a mother and daughter pair.76–78 plasia (PASH) may be seen. Mature stromal metaplasia is rare but if
Phyllodes tumors are hypercellular fibroepithelial tumors and present is often lipomatous.79
characteristically demonstrate an exaggerated intracanalicular Phyllodes tumors are graded according to recommendations by
growth pattern with leaf-like stromal proliferations protruding the World Health Organization (WHO) as benign, borderline, or
into dilated clefts or gaping cyst-like spaces (Fig. 4A–B). Some malignant based on the presence and degree of stromal cellularity,
cases demonstrate periductal or subepithelial stromal condensa- atypia, mitotic activity, border infiltration versus circumscription
tion, with increased stromal cell density subjacent to the and stromal overgrowth (Table 1).1 Stromal overgrowth is defined

Fig. 4. Histologic Features of Phyllodes Tumors. (A–B) Benign phyllodes tumor showing (A) well-circumscribed borders without infiltrative growth and with characteristic leaf-
like architecture (B) High power magnification reveals mild cellularity of the stromal fronds with no to mild stromal cell cytologic atypia. Mitotic activity was scant
(averaging 1 mitosis/10 HPF; not shown). (C) Stromal condensation, with increased stromal cellularity subjacent to the epithelium; mitotic activity may be identified in this
zone. Stromal condensation can be subjective to evaluate but supports a diagnosis of phyllodes tumor. (D–E and F) Borderline phyllodes tumors (D) This tumor demonstrates
well-developed leaf-like growth (inset) and circumscribed borders (not shown), in most areas showing mild to moderate stromal cellularity, mild stromal cell atypia and few
mitoses. A more cellular area with (E) moderate cytologic atypia and increased mitoses (up to 9/10 HPF) was identified, most consistent with borderline phyllodes tumor.
(F) Infiltrative growth and stromal overgrowth, even if focal, are generally features of at least borderline tumors.
444 G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452

as proliferative stroma resulting in lack of epithelium in at least one periductal stromal sarcoma was defined as having moderate
low-power (4x objective, 10x ocular) microscopic field. Grading stromal atypia, at least 3 mitotic figures/10 HPF, and infiltrative
requires evaluation of the entire lesion and should be based on an growth, whereas periductal stromal hyperplasia was non-in-
excision specimen.80,81 Benign phyllodes tumors are mildly cellular filtrative with less atypia and mitoses. However, this classification
without stromal overgrowth, have only mild atypia with o5 mi- does not clearly correlate with recurrence, and the term periductal
totic figures/10 HPF, and have well-circumscribed, pushing borders stromal tumor is preferred over sarcoma.86 Recurrences often re-
without infiltrative growth (Fig. 4A–B). On the other end of the semble phyllodes tumors.
spectrum, malignant tumors show marked stromal cellularity,
marked atypia, numerous mitoses (Z10 mitotic figures/10 HPF) Practical problems and challenges in diagnosis
and at least focal stromal overgrowth with infiltrative borders
(Fig. 5A–C). Malignant heterologous stroma defines malignancy Phyllodes tumor grading
regardless of the presence of the other features. Liposarcomatous Phyllodes tumor grade correlates with risk of recurrence and
differentiation is most common, but chondrosarcoma, osteosarco- metastasis and is important for patient management, but grading
ma, rhabdomyosarcoma and others may rarely be seen, sometimes can be challenging in practice. This is highlighted in a study by
in combination (Fig. 5D–F).1,63 Borderline tumors show some but Lawton et al., in which individual diagnoses ranged from fi-
not all features of malignancy, often with an intermediate mitotic broadenoma to borderline phyllodes tumor in 9 (43%) of 21 chal-
count, and accordingly may be varied in appearance (Fig. 4D–F). lenging cellular fibroepithelial lesions reviewed by ten breast pa-
Numerous studies have evaluated immunohistochemical markers thologists. The grade of one phyllodes tumor in this study ranged
as adjuncts for grading and tumor behavior; although many (in- from benign to malignant.87 Of the criteria determining grade,
cluding p53, CD117, EGFR, VEGF, CD10 and Ki67) correlate with stromal cellularity and atypia in particular are each inherently
grade, none has been shown to independently predict recurrence or subjective and may be heterogeneous throughout the tumor. In-
metastasis.82 terpretative differences are expected to be greatest in lesions
Although histologic grading estimates the risk of phyllodes bordering between mild and moderate or between moderate and
tumor recurrence and/or metastasis, grading is not necessarily marked categories. A reasonable approach is to define mild hy-
predictive of tumor aggression in individual cases.1,63 Nonetheless, percellularity as increased in comparison to normal perilobular
grade has been shown to correlate with behavior, with malignant stroma but with well-spaced nuclei showing no overlap, whereas
phyllodes tumors showing the highest risk for local recurrence (up the nuclei of markedly hypercellular stroma show confluent areas
to 23–30%) and also demonstrating metastatic potential (up to 22% of dense nuclear overlap; moderate cellularity is intermediate
of cases).1,63 Borderline tumors may have very low (0–4%) meta- between the two extremes.32,88 Mild atypia is characterized by
static potential, whereas benign tumors are generally not thought small, uniform nuclei with smooth borders, not much different
to metastasize. Rare benign tumors preceding metastasis have from normal perilobular stroma, and marked atypia is defined by
been reported, but inadequate sampling of higher grade areas of marked nuclear pleomorphism with coarse chromatin, irregular
heterogeneous tumors cannot be excluded.83 Although local re- membranes and nucleoli; moderate atypia falls between the
currences are usually of the same grade as the primary tumor, two.32,88
tumors may occasionally recur with higher or lower grade than Intratumoral heterogeneity may further complicate the inter-
initial presentation, which may be due to progression and/or in- pretation, and good practice may be to evaluate atypia, cellularity
tratumoral heterogeneity.63,67,71,84 Metastases are essentially al- and mitotic activity in the most affected area of the tumor, which
ways composed of the stromal component and are most com- should be adequately sampled to include at least 1 section per cm
monly of spindle cell morphology.85 Heterologous sarcomatous of gross tumor size, as well as grossly heterogenous areas. In ad-
elements in the primary tumor may or may not be present in the dition to subjectivity of individual criteria, grading requires in-
metastasis, and the metastasis rarely demonstrates heterologous tegration of the multiple parameters, which can be further con-
stroma not present in the primary tumor. founding. A particular problem in this regard is the determination
Periductal stromal tumors are considered phyllodes tumor of malignancy, which is a critical distinction given the high risk for
variants characterized by periductal stromal proliferation asso- metastatic disease and poor outcome. Indeed, in one study, all
ciated with non-compressed or dilated ducts. Adjacent pro- phyllodes tumor metastases and deaths were preceded by a ma-
liferative nodules can coalesce to form a larger mass. Initially, lignant primary diagnosis.68 Despite these prognostic implications,
the histologic threshold required for a malignant diagnosis re-
Table 1 mains poorly defined. From a practical standpoint, we require the
Histologic features of phyllodes tumorsa. presence of all five main malignant features (marked stromal
cellularity, Z 10 stromal mitoses/10 HPF, marked stromal atypia,
Benign Borderline Malignant
stromal overgrowth and infiltrative tumor borders) or the pre-
Stromal Mild, non-uni- Moderate, non- Marked, sence of malignant heterologous stromal differentiation for a di-
hypercellularity form or diffuse uniform or diffuse usually agnosis of malignant phyllodes tumor.1,88 Occasional tumors have
diffuse only focal marked cellularity, atypia and elevated mitotic rate, with
Stromal mitotic 0–4/10 HPF 5–9/10 HPF Z 10/10 HPF
activity
the rest of the tumor resembling lower grade phyllodes tumor
Stromal cell atypia Mild or none Mild or moderate Marked (Fig. 4D–E). We recommend classification of such tumors as bor-
Stromal overgrowth Absent May be focal Usually pre- derline, with an acknowledgment of possible metastatic potential.
sent, may be
diffuse
Differential diagnosis of benign cellular fibroepithelial lesions
Tumor borders Circumscribed, Circumscribed or Infiltrative
pushing focally infiltrative Distinction of cellular fibroadenoma from benign phyllodes tu-
Malignant hetero- Absent Absent Presentb mor is a common diagnostic challenge (Table 2). This was further
logous highlighted in the interobserver study by Lawton et al., in which
differentiation agreement among all pathologists was seen in only 2 (9.5%) of the
Adapted from.1
a 21 challenging fibroepithelial lesions.87 The diagnosis of phyllodes
b
Malignant heterologous differentiation defines malignancy even in the tumor is usually not problematic in the presence of a cellular fi-
absence of the other features. broepithelial tumor with classic well-developed leaf-like
G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452 445

Fig. 5. Malignant Phyllodes Tumor. (A) Markedly hypercellular stroma is evident at low power of this malignant phyllodes tumor, which retains areas of well-developed leaf-
like growth. (B) In other cases, the epithelial component is scant due to marked stromal overgrowth, and extensive tumor sampling may be required. Note also the markedly
infiltrative tumor border. (C) High power magnification reveals markedly cellular stroma and marked cytologic atypia with pleomorphic nuclei and abundant mitotic activity
( Z 10 mitoses/10 HPF). All histologic criteria should generally be present for a diagnosis of malignant phyllodes tumor, although (D–F) malignant heterologous stroma itself
is sufficient for a malignant diagnosis. (D) Liposarcomatous differentiation is the most common type of malignant heterologous differentiation. (E) Mixed osteosarcomatous
and chondrosarcomatous differentiation in malignant phyllodes tumor. Note background fibroepithelial architecture in D and E insets. (F) Leiomyosarcomatous differ-
entiation in malignant phyllodes tumor, supported by desmin expression by the malignant smooth muscle tumor cells (inset).

architecture. The difficulty in other cases stems from the application exclude the diagnosis when a cellular fibroepithelial lesion shows
of subjective criteria (stromal cellularity, atypia, well-developed other features of phyllodes tumor, such as clear cut stromal atypia,
leaf-like growth, stromal condensation) with overlapping features overgrowth, and/or infiltrative borders. On the other hand, leaf-like
between groups. Whereas fibroadenomas classically lack stromal areas may be seen in some fibroadenomas (Fig. 6C), but these are
cell atypia, this feature can be subjective, especially at the mild end characteristically focal and less cellular without stromal condensa-
of the spectrum as would be typical of benign phyllodes tumor. tion. The fronds of such fibroadenomas appear to fit together like a
Cellularity and mitotic activity of benign phyllodes tumor (0–4 jigsaw puzzle, as opposed to phyllodes tumor, in which the bulbous
mitoses/10 HPF) overlap with cellular and juvenile fibroadenomas stroma protrudes into gaping cyst-like spaces. Focal leaf-like growth
in adults and children.28,30,32 Leaf-like growth is the best dis- may be especially common in pediatric fibroepithelial lesions and
criminator between the two groups but is not always prominent or should be interpreted with caution in the absence of other phyllodal
well-developed in phyllodes tumors, and lack thereof should not features.30
446 G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452

Table 2 heterogeneity, subepithelial condensation, and stromal pleo-


Morphologic features of benign phyllodes tumor versus cellular fibroadenoma. morphism were considered most useful, although the subjective
nature of these features again questions their overall practical
Benign phyllodes Cellular
tumor Fibroadenoma utility.33 Tan et al. described a 5-gene RT-PCR signature that may
help predict phyllodes tumor on excision, but included fi-
Leaf-like growth pattern Present, well- Absent or focal broadenomas of the usual type, and validation with cellular fi-
developed
broadenomas is needed.92 In summary, no feature(s) on core
Periductal stromal condensation May be present Absent
Stromal heterogeneity May be present Usually absent biopsy can reliably predict phyllodes tumor over cellular fi-
Stromal cell atypia Mild Nonea broadenoma in lesions that are not obviously at the higher end of
Stromal hypercellularity Mild Mild the phyllodes tumor spectrum. A diagnosis of fibroepithelial lesion
Stromal mitoses 0–4/10 HPF Fewb
with cellular stroma with recommendation for excision for more
Pseudoangiomatous stromal May be present Rarely present
hyperplasia complete characterization is most prudent in such cases.
Squamous metaplasia May be present Virtually absent
Differential diagnosis of malignant phyllodes tumor
a
Multinucleated stromal giant cells may be seen. The differential diagnosis of malignant phyllodes tumors pri-
b
Typically r 2/10 HPF, but may be more numerous in children, adolescents
and young women.
marily includes metaplastic carcinoma and primary or metastatic
sarcoma, which can be problematic on core biopsy and in excision
specimens (Table 3). Metaplastic carcinoma is more common than
Given the overlap between cellular fibroadenoma and benign malignant phyllodes tumor or breast sarcoma and should always be
phyllodes tumor, stromal cellularity alone is usually not a useful excluded.1 Accurate diagnosis on core biopsy can have important
discriminator on the low end of the histologic spectrum; however, clinical implications, as neoadjuvant chemotherapy and sentinel
cellular fibroadenomas lack significant nuclear overlap or sheet- lymph node biopsy at time of surgery may be considered for me-
like stromal growth, which favor phyllodes tumor. In our experi- taplastic carcinoma but not phyllodes tumor.93 A recent study
ence, loose fascicular stromal growth can also favor phyllodes suggested that malignant phyllodes tumors and breast sarcomas
tumor. Prominent stromal heterogeneity may also support phyl- have similar clinicopathologic features, outcomes and treatment,
lodes tumor (Fig. 6G-H), as cellular fibroadenomas are typically and the relevance of this distinction is currently less clear.94
more uniform in epithelial-stromal distribution and character of The presence of characteristic leaf-like growth facilitates the
the stroma (Fig. 2). Anecdotally, we find squamous metaplasia of
diagnosis of malignant phyllodes tumor in straightforward cases
the epithelial component to be more common in phyllodes tumors
(Fig. 5A). Breast sarcomas usually displace rather than entrap
(Fig. 6E), which may result from friction of the bulbous stromal
mammary epithelium. An associated malignant epithelial com-
fronds with the duct walls.
ponent or in situ carcinoma favors metaplastic carcinoma, but
Benign multinucleated giant cells may be encountered in fi-
heterologous elements are not specific to any of the entities and
broepithelial lesions, including both fibroadenomas and phyllodes
are not useful. Malignant phyllodes tumors with extensive stromal
tumors, and may be a diagnostic pitfall. These cells are typically
overgrowth are more problematic and can mimic spindle cell or
present as discrete subpopulations with degenerative-type atypia,
other metaplastic carcinomas and sarcomas. Extensive sampling of
which contrasts with the more dispersed distribution of neoplastic
an excision specimen may be required to identify the associated
stromal cells with irregular nuclear membranes, coarse chromatin,
epithelial component of phyllodes tumor, which can be very focal.
and possible mitotic activity in phyllodes tumors.68,89
The distinction may be especially difficult in core biopsies
Some fibroepithelial lesions may simply not be histologically
(Fig. 7A), and immunohistochemistry can be useful. Metaplastic
distinguishable as cellular fibroadenomas or benign phyllodes tu-
carcinomas express p63 and cytokeratins, especially high-mole-
mors. In such cases, a diagnosis of benign fibroepithelial neoplasm
cular-weight keratins (such as 34βE12, CK5/6, CK14 and AE1/3),
can be rendered, with an explanation of the features present, the
but staining can vary between markers and may be focal.1,95–98
difficulty in categorization, distance to margins, and some risk but
low likelihood of local recurrence.1,31 Importantly, phyllodes tumors can be focally (1–5%) cytokeratin
The distinction between cellular fibroadenoma and phyllodes positive (Fig. 7B), which increases with grade and was only seen in
tumor is particularly problematic in core biopsies (Fig. 6A). Nu- malignant tumors in one study.95,96,99,100 Similarly, malignant
merous studies have attempted to identify histopathologic factors phyllodes tumors may focally express p63 in up 57% of cases
in core biopsies that could predict phyllodes tumor on (Fig. 7C).95 Accordingly, focal keratin or p63 positivity does not
excision.32–34,80,90,91 Not surprisingly, features reported to be use- exclude phyllodes tumor, especially on core biopsy, and a diag-
ful are variable between studies, which can be explained by the nosis of malignant neoplasm with spindle cell features may be
inherently heterogeneous character of fibroepithelial lesions, the prudent. On the other hand, diffuse strong positive keratin or p63
subjectivity of histologic criteria and thresholds, and tissue sam- staining favors metaplastic carcinoma. The TP63 splice variant p40
pling. Stromal cellularity and/or proliferative indices (mitotic ac- may be more specific (86% vs. 43%) but less sensitive (46% vs. 62%)
tivity, Ki67) have been described as the most promising than p63 for metaplastic carcinoma over malignant phyllodes tu-
features.32–34,80,90 However, due to subjectivity of stromal cellu- mors, but this is based on a small tissue microarray study and
larity evaluation and overlap of proliferative indices between requires further validation.95 CD34 is negative in metaplastic car-
groups, practical utility is likely limited to lesions at the more cinomas, and expression decreases with increasing phyllodes tu-
extreme ends of the spectrum.32,90 PASH stroma in a fi- mor grade, being negative in up to 43% of malignant tumors.101,102
broepithelial lesion may be a more objective feature to raise Therefore, CD34 expression excludes metaplastic carcinoma
concern for phyllodes tumor on core biopsy and can provide (Fig. 7E), but negative staining is not helpful. We recommend a
support for excision and evaluation of the entire lesion (Fig. 6D).90 large panel of high- and low-molecular weight cytokeratins, p63
Consistent with the inter- and intra-lesional heterogeneity of fi- and CD34 to help establish the diagnosis. In one study, 66% of
broepithelial lesions, Yasir et al. concluded that a combination of metaplastic carcinomas but no phyllodes tumors (including 14
3 or more of 7 adverse features may be more predictive than in- malignant tumors) expressed Sox10, and GATA3 may be more
dividual features, with phyllodes tumors averaging more features frequently positive in metaplastic carcinomas, but these markers
(3.9) than fibroadenomas (1.4). A combination of stromal require further study.103,104
G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452 447

Fig. 6. Histologic Features of Cellular Fibroadenomas versus Benign Phyllodes Tumors. (A) Core biopsy of fibroepithelial lesion with cellular stroma. Distinction between cellular
fibroadenoma and phyllodes tumor is often not possible on core biopsy, and final classification can be deferred to an excision specimen. (B) Tissue fragmentation in core
biopsy specimens is not specific to phyllodes tumor and can also be seen in fibroadenomas with intracanalicular growth, as in this case (inset shows fibroadenoma in the
excision specimen of the biopsied lesion). (C) Focal leaf-like growth in a fibroadenoma. When present, this is generally focal, poorly-developed and less cellular, in contrast to
phyllodes tumors. (D) Pseudoangiomatous stromal hyperplasia in benign phyllodes tumor (see inset). On core biopsy, this feature in combination with others can raise
consideration for phyllodes tumor and advocate for excision. (E) Squamous metaplasia in benign phyllodes tumor (inset with high power magnification). (F) Lipomatous
metaplasia in a cellular fibroadenoma. Note the lack of leaf-like growth and overall lesional circumscription, with fat confined within the rounded lesional borders, in
contrast to infiltrative growth that can be seen in phyllodes tumors. (G, H) Heterogeneity in phyllodes tumors with fibroadenoma-like areas. (G) Benign phyllodes tumor with
area of distinct leaf-like growth and hypercellular stroma (top) adjacent to area with hypocellular hyalinized stroma that is otherwise indistiguishable form fibroadenoma
(bottom). (H) Phyllodes tumor with central area of stromal overgrowth and focal leaf-like architecture (bottom right), as well as other peripheral areas indistinguishable from
hyalinized fibroadenoma.
448 G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452

Table 3
Useful histopathologic features in the differential diagnosis of malignant spindle cell lesions of the breast.

Malignant phyllodes tumor Metaplastic spindle cell carcinoma Sarcoma (primary or metastatic)

Leaf-like growth pattern Often presenta Absent Absent


Heterologous differentiation May be present May be present Tumor lineage-dependent
Conventional invasive carcinoma Absent May be presentb Absent
Ductal carcinoma in situ Absentc May be present Absent
Cytokeratin(s)d Negative or focal Positive Negative or focal
p63 Negative or focal Positive Negative or focal
CD34 Positive or negativee Negative Positive or negative

a
May be focal or absent with extensive stromal overgrowth, especially on core biopsy.
b
Most often ductal, may be squamous.
c
May rarely be involved by ductal carcinoma in situ.
d
Panel of high- and low- molecular weight cytokeratins is recommended.
e
Decreases with increasing phyllodes tumor grade; often negative in malignant tumors.

Fig. 7. Immunohistochemistry in the Differential Diagnosis of High-Grade Spindle Cell Lesions of the Breast. (A) Core biopsy of a high-grade spindle cell neoplasm - The
differential diagnosis includes malignant phyllodes tumor, metaplastic (spindle cell) carcinoma and primary/metastatic sarcoma, which often cannot be distinguished on
core biopsy. Malignant phyllodes tumors may focally express (B) cytokeratin(s) and/or (C) p63, and focal staining for these markers does not exclude the diagnosis. On the
other hand, diffuse keratin and/or p63 expression can favor metaplastic carcinoma. (D) Excision specimen of tumor shown in B and C reveals residual leaf-like growth
characteristic of malignant phyllodes tumor with prominent stromal overgrowth. Extensive tumor sampling may be required to identify the epithelial component. CD34
expression decreases with increasing phyllodes tumor grade, and CD34 is immunopositive in only a subset of malignant phyllodes tumors. (E) Strong diffuse CD34 im-
munostaining excludes metaplastic carcinoma, but negative staining is not helpful. (F) Nuclear β-catenin expression can be seen in metaplastic carcinomas, malignant
phyllodes tumors (shown here), and other lesions (such as fibromatosis) and is not helpful in the differential diagnosis.
G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452 449

Pathogenesis and molecular pathology More recent targeted sequencing revealed shared mutations in a
The stromal component of phyllodes tumors is neoplastic.49,52,105 fibroepithelial tumor with fibroadenoma-like and phyllodes tu-
Similar to fibroadenomas, epithelial-stromal interactions are con- mor-like areas, as well as in fibroadenoma preceding phyllodes
sidered to play a key role in tumor development. Supportive evi- tumor recurrence.3,46 Nonetheless, the data overall are scarce, and
dence for this view includes morphologic observations of stromal inherent intratumoral heterogeneity as an explanation for these
condensation and mitotic activity in the subepithelial zone, de- findings cannot be excluded.
creasing epithelial ER and PR expression with increasing tumor
grade, and correlation of epithelial Wnt5a expression with stromal β-
catenin activation.106–108 Some studies have also demonstrated
monoclonality of the epithelial component.52,109 It has been postu- Issues in clinical management and outcomes
lated that the stroma becomes independent of epithelial interactions
with progression towards malignancy.107 Fibroadenomas of usual type generally do not recur, and those
A number of studies have attempted to characterize the mo- diagnosed on core biopsy without concerning clinical behavior can be
lecular features of phyllodes tumors in order to understand their enucleated or managed non-surgically. Treatment depends on patient
pathogenesis and identify factors that may predict outcome. Col- preference and risk factors. Nonsurgical options include observation
lectively, these have shown increased karyotypic complexity, more with follow-up, percutaneous vacuum-assisted biopsy, and
chromosomal gains and losses, and increased loss of hetero- cryoablation.122–124 Incomplete removal with these approaches is as-
zygosity (LOH) with increasing tumor grade.84,109–115 The presence
sociated with larger size (42 cm).122,123,125 Juvenile fibroadenomas
of spatial and temporal intratumoral heterogeneity with respect to
are often excised, given the rapid growth of some lesions and the
copy number changes, including in morphologically similar and
inability to distinguish them from phyllodes tumor on core biopsy.
distinct areas, may in part explain the imperfect correlation be-
Excision aims to preserve as much breast tissue as possible in ado-
tween histologic grading and tumor behavior, with implications
for tumor recurrence and progression.6,84 lescent girls to allow for proper breast development.11
More recent DNA sequencing studies have identified recurrent Phyllodes tumors are treated by surgical excision, and most do
MED12 exon 2 mutations in phyllodes tumors. The mutational not recur. Risk of recurrence is associated with increasing tumor
spectrum is similar to that of fibroadenomas, with enrichment of grade and is reported to be 10–17%, 14–25% and 23–30% for benign,
mutations at codon 44, supporting an underlying commonality in borderline and malignant tumors, respectively.1,63 In addition, mar-
pathogenesis.3,4,26,45–48,105,116,117 All grades of phyllodes tumor gin status is a strong predictor of local recurrence.56,63,65,71,126,127 In a
harbor MED12 mutations, with some but not all studies suggesting large study, Tan et al. found that stromal cell atypia, stromal over-
a lower frequency in malignant tumors.45,46,48,105,117,118 Given a growth and surgical margin status were most predictive of recur-
postulated association of MED12 with estrogen signaling, this rence-free survival (RFS), with mitotic activity nearing statistical
suggests possible loss of hormonal dependence in malignant tu- significance on multivariate analysis (A.M.O.S. criteria). A nomogram
mors but requires further study. Interestingly, Ng et al. reported using these features was developed to help predict RFS more accu-
longer disease-free survival in patients with MED12-mutated rately than grade or a sum of histologic features, which has since
compared to MED12 wild-type tumors on univariate analysis, been validated in other cohorts.63,128,129 The nomogram can be
which was not identified with grade alone.116 Additional aberra-
adapted to routine practice to provide clinicians with personalized
tions in cancer-related genes are associated with tumor progres-
local recurrence risk data.
sion in borderline and malignant tumors. These include the tumor
In general, excision to negative margins and including a rim of
suppressors TP53, RB1, NF1 and PTEN, oncogenes such as PIK3CA,
normal tissue has been recommended for phyllodes tumors.
BRAF and EGFR, and chromatin remodeling genes SETD2 and
However, adequate distance to negative margins has not been
KMT2D/MLL2, among others.3–6 In one study, 80% of malignant
phyllodes tumors showed aberrations in phosphatidyl inositol-3 randomly or prospectively evaluated. A 1 cm rim of uninvolved
kinase or Ras signaling pathways.6 Telomerase reverse tran- tissue has been recommended based on retrospective analyses,
scriptase (TERT) hotspot promoter mutations or amplifications are but others have shown no clear relationship between size of
also associated with phyllodes tumor progression, being most margin width and recurrence or RFS, and optimal margin width
frequent in malignant tumors.4–6,119 Discrepant reports of TERT remains uncertain.56,67,71,93,126,127,130–132 Recent studies have ad-
promoter mutations in fibroadenomas and benign phyllodes tu- vocated less aggressive treatment for benign phyllodes
mors may be due to differing diagnostic thresholds or sample tumors.56,67,131,133,134 Based on overall low recurrence rates and
sizes.5,6,119 Tumors with malignant heterologous stroma show lack of correlation with margin status, the findings can be used to
evidence of divergent evolution between heterologous and non- support a more conservative approach without requirement for
heterologous areas; interestingly, those with liposarcomatous negative margins for benign phyllodes tumors. This is especially
differentiation lack CDK4/MDM2 amplification characteristic of relevant in cases diagnosed as cellular fibroepithelial lesion on core
well-differentiated soft tissue liposarcoma, suggesting an alternate biopsy and subsequently found to be benign phyllodes tumor on
pathogenesis despite histologic similarity.6,120 Together, these excision, as the extent of initial excision (enucleation for fi-
studies highlight pathways important for tumor progression and
broadenoma vs. wider excision to negative margins for phyllodes
may identify potentially actionable targets, especially in malignant
tumor) may be less important than previously thought. However,
tumors without effective treatment strategies beyond surgery.
close follow-up is required with timely re-excision to negative
Given their morphologic similarities, it has long been suggested
that some phyllodes tumors may arise from fibroadenomas. Per- margins for any subsequent recurrence. Borderline and malignant
haps the subset of heterogeneous phyllodes tumors with fi- phyllodes tumors should be excised to negative margins.
broadenoma-like areas may arise from fibroadenomas (Fig. 6G-H), Routine use of adjuvant radiation therapy currently has no role
but evidence is limited. Noguchi et al. demonstrated inactivation of in phyllodes tumor treatment, and its use is often determined on a
the same AR allele in three fibroadenomas that subsequently re- case-by case basis 1,135,136 Radiation may decrease local recur-
curred as phyllodes tumors,53 and another study described a fi- rence, but survival benefit has not been demonstrated.130,137 There
broepithelial lesion with shared LOH in areas morphologically is no proven benefit for chemotherapy.135,136,138 The optimal local
resembling fibroadenoma and malignant phyllodes tumor.121 treatment for phyllodes tumor continues to evolve.
450 G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452

Acknowledgments clinicopathologic study of 21 patients. Arch Pathol Lab Med 1987;111:427–432.


30. Tay TK, Chang KT, Thike AA, et al. Paediatric fibroepithelial lesions revisited:
pathological insights. J Clin Pathol 2015;68:633–641.
We thank Sarah Bowman for technical expertise in preparing 31. Jacobs TW. Update on Fibroepithelial Lesions of the Breast. Surg Pathol Clin
the figures. 2009;2:301–317.
32. Jacobs TW, Chen YY, Guinee Jr DG, et al. Fibroepithelial lesions with cellular
stroma on breast core needle biopsy: are there predictors of outcome on sur-
gical excision? Am J Clin Pathol 2005;124:342–354.
References 33. Yasir S, Gamez R, Jenkins S, et al. Significant histologic features differentiating
cellular fibroadenoma from phyllodes tumor on core needle biopsy specimens.
Am J Clin Pathol 2014;142:362–369.
1. Tan PH, Tse G, Lee A, Simpson JF, Hanby AM. Fibroepithelial Tumours. In: Lakhani 34. Lee AH, Hodi Z, Ellis IO, et al. Histological features useful in the distinction of
SR, Ellis IO, Schnitt SJ, Tan PT, Vijver MJ, eds. WHO Classification of Tumours of the phyllodes tumour and fibroadenoma on needle core biopsy of the breast. His-
Breast. 4th ed., Lyon, France: International Agency for Research on Cancer (IARC); topathology 2007;51:336–344.
2012:141–147. 35. Jacobs TW, Chen YY, Guinee DG, et al. Phyllodes tumor subsequent to a di-
2. Lim WK, Ong CK, Tan J, et al. Exome sequencing identifies highly recurrent agnosis of fibroadenoma on breast core needle biopsy: frequency and char-
MED12 somatic mutations in breast fibroadenoma. Nat Genet 2014;46:877–880. acteristics. Mod Pathol 2014;27(Supplement 2):56A.
3. Tan J, Ong CK, Lim WK, et al. Genomic landscapes of breast fibroepithelial tu- 36. Yamaguchi R, Tanaka M, Mizushima Y, et al. Myxomatous fibroadenoma of the
mors. Nat Genet 2015;47:1341–1345. breast: correlation with clinicopathologic and radiologic features. Hum Pathol
4. Cani AK, Hovelson DH, McDaniel AS, et al. Next-Gen Sequencing Exposes Fre- 2011;42:419–423.
quent MED12 Mutations and Actionable Therapeutic Targets in Phyllodes Tu- 37. Skenderi F, Krakonja F, Vranic S. Infarcted fibroadenoma of the breast: report of
mors. Mol Cancer Res 2015. two new cases with review of the literature. Diagn Pathol 2013;8:38.
5. Piscuoglio S, Ng CK, Murray M, et al. Massively parallel sequencing of phyllodes 38. Ichihara S, Matsuyama T, Kubo K, et al. Infarction of breast fibroadenoma in a
tumours of the breast reveals actionable mutations, and TERT promoter hotspot postmenopausal woman. Pathol Int 1994;44:398–400.
mutations and TERT gene amplification as likely drivers of progression. J Pathol 39. Pinto RG, Couto F, Mandreker S. Infarction after fine needle aspiration. A report
2016;238:508–518. of four cases. Acta Cytol 1996;40:739–741.
6. Liu SY, Joseph NM, Ravindranathan A, et al. Genomic profiling of malignant 40. Oh YJ, Choi SH, Chung SY, et al. Spontaneously infarcted fibroadenoma mi-
phyllodes tumors reveals aberrations in FGFR1 and PI-3 kinase/RAS signaling micking breast cancer. J Ultrasound Med 2009;28:1421–1423.
pathways and provides insights into intratumoral heterogeneity. Mod Pathol 41. Sawyer EJ, Hanby AM, Poulsom R, et al. Beta-catenin abnormalities and asso-
2016;29:1012–1027. ciated insulin-like growth factor overexpression are important in phyllodes
7. Nozad S, Sheehan CE, Gay LM, et al. Comprehensive genomic profiling of ma- tumours and fibroadenomas of the breast. J Pathol 2003;200:627–632.
lignant phyllodes tumors of the breast. Breast Cancer Res Treat 2017;162:597– 42. Sapino A, Bosco M, Cassoni P, et al. Estrogen receptor-beta is expressed in
602. stromal cells of fibroadenoma and phyllodes tumors of the breast. Mod Pathol
8. Brogi E. Fibroepithelial Neoplasms. In: Hoda S, Brogi E, Koerner FC, Rosen PR, eds. 2006;19:599–606.
Rosen's Breast Pathology. 4th ed., Philadelphia, PA: Wolters Kluwer; 2014:1379. 43. Je EM, Kim MR, Min KO, et al. Mutational analysis of MED12 exon 2 in uterine
9. Weaver DL, Rosenberg RD, Barlow WE, et al. Pathologic findings from the Breast leiomyoma and other common tumors. Int J Cancer 2012;131:E1044–E1047.
Cancer Surveillance Consortium: population-based outcomes in women under- 44. Makinen N, Mehine M, Tolvanen J, et al. MED12, the mediator complex subunit
going biopsy after screening mammography. Cancer 2006;106:732–742. 12 gene, is mutated at high frequency in uterine leiomyomas. Science
10. Foster ME, Garrahan N, Williams S. Fibroadenoma of the breast: a clinical and 2011;334:252–255.
pathological study. J R Coll Surg Edinb 1988;33:16–19. 45. Pfarr N, Kriegsmann M, Sinn P, et al. Distribution of MED12 mutations in fi-
11. Pike AM, Oberman HA. Juvenile (cellular) adenofibromas. A clinicopathologic broadenomas and phyllodes tumors of the breast–implications for tumor biology
study. Am J Surg Pathol 1985;9:730–736. and pathological diagnosis. Genes Chromosomes Cancer 2015;54:444–452.
12. Song BS, Kim EK, Seol H, et al. Giant juvenile fibroadenoma of the breast: a case 46. Piscuoglio S, Murray M, Fusco N, et al. MED12 somatic mutations in fi-
report and brief literature review. Ann Pediatr Endocrinol Metab 2014;19:45–48. broadenomas and phyllodes tumours of the breast. Histopathology
13. Trapido EJ, Brinton LA, Schairer C, et al. Estrogen replacement therapy and 2015;67:719–729.
benign breast disease. J Natl Cancer Inst 1984;73:1101–1105. 47. Tan WJ, Chan JY, Thike AA, et al. MED12 protein expression in breast fi-
14. Dixon JM. Cystic disease and fibroadenoma of the breast: natural history and broepithelial lesions: correlation with mutation status and oestrogen receptor
relation to breast cancer risk. Br Med Bull 1991;47:258–271. expression. J Clin Pathol 2016;69:858–865.
15. Gupta P, Foshee S, Garcia-Morales F, et al. Fibroadenoma in male breast: case 48. Mishima C, Kagara N, Tanei T, et al. Mutational analysis of MED12 in fi-
report and literature review. Breast Dis 2011;33:45–48. broadenomas and phyllodes tumors of the breast by means of targeted next-
16. Kanhai RC, Hage JJ, Bloemena E, et al. Mammary fibroadenoma in a male-to- generation sequencing. Breast Cancer Res Treat 2015;152:305–312.
female transsexual. Histopathology 1999;35:183–185. 49. Noguchi S, Motomura K, Inaji H, et al. Clonal analysis of fibroadenoma and
17. Hilton DA, Jameson JS, Furness PN. A cellular fibroadenoma resembling a be- phyllodes tumor of the breast. Cancer Res 1993;53:4071–4074.
nign phyllodes tumour in a young male with gynaecomastia. Histopathology 50. Kobayashi S, Iwase H, Kuzushima T, et al. Consecutively occurring multiple
1991;18:476–477. fibroadenomas of the breast distinguished from phyllodes tumors by clonality
18. Iaria G, Pisani F, De Luca L, et al. Prospective study of switch from cyclosporine analysis of stromal tissue. Breast Cancer 1999;6:201–206.
to tacrolimus for fibroadenomas of the breast in kidney transplantation. 51. Kasami M, Vnencak-Jones CL, Manning S, et al. Monoclonality in fi-
Transplant Proc 2010;42:1169–1170. broadenomas with complex histology and phyllodal features. Breast Cancer Res
19. Seo YL, Choi CS, Yoon DY, et al. Benign breast diseases associated with cy- Treat 1998;50:185–191.
closporine therapy in renal transplant recipients. Transplant Proc 52. Kuijper A, Buerger H, Simon R, et al. Analysis of the progression of fi-
2005;37:4315–4319. broepithelial tumours of the breast by PCR-based clonality assay. J Pathol
20. Alkhunaizi AM, Ismail A, Yousif BM. Breast fibroadenomas in renal transplant 2002;197:575–581.
recipients. Transplant Proc 2004;36:1839–1840. 53. Noguchi S, Yokouchi H, Aihara T, et al. Progression of fibroadenoma to phyl-
21. Williamson ME, Lyons K, Hughes LE. Multiple fibroadenomas of the breast: a lodes tumor demonstrated by clonal analysis. Cancer 1995;76:1779–1785.
problem of uncertain incidence and management. Ann R Coll Surg Engl 54. Lozada JRPF, Burke KA, Maguire A, et al. Massively parallel sequencing analysis
1993;75:161–163. of myxoid fibroadenomas reveals a genomic landscape distinct from that of
22. Carney JA, Toorkey BC. Myxoid fibroadenoma and allied conditions (myx- conventional fibroadenomas. Mod Pathol 2017;30(supplement 2):57A.
omatosis) of the breast. A heritable disorder with special associations including 55. Karim RZ, Gerega SK, Yang YH, et al. Phyllodes tumours of the breast: a clin-
cardiac and cutaneous myxomas. Am J Surg Pathol 1991;15:713–721. icopathological analysis of 65 cases from a single institution. Breast
23. Kuijper A, Mommers EC, van der Wall E, et al. Histopathology of fibroadenoma 2009;18:165–170.
of the breast. Am J Clin Pathol 2001;115:736–742. 56. Kim S, Kim JY, Kim do H, et al. Analysis of phyllodes tumor recurrence ac-
24. Sklair-Levy M, Sella T, Alweiss T, et al. Incidence and management of complex cording to the histologic grade. Breast Cancer Res Treat 2013;141:353–363.
fibroadenomas. AJR Am J Roentgenol 2008;190:214–218. 57. Gordon PB, Gagnon FA, Lanzkowsky L. Solid breast masses diagnosed as fi-
25. Dupont WD, Page DL, Parl FF, et al. Long-term risk of breast cancer in women broadenoma at fine-needle aspiration biopsy: acceptable rates of growth at
with fibroadenoma. N Engl J Med 1994;331:10–15. long-term follow-up. Radiology 2003;229:233–238.
26. Lien HC, Huang CS, Yang YW, et al. Mutational analysis of MED12 exon 2 in a 58. Liberman L, Bonaccio E, Hamele-Bena D, et al. Benign and malignant phyllodes
spectrum of fibroepithelial tumours of the breast: implications for pathogen- tumors: mammographic and sonographic findings. Radiology 1996;198:121–124.
esis and histogenesis. Histopathology 2015. 59. Buchberger W, Strasser K, Heim K, et al. Phylloides tumor: findings on
27. Volckmar AL, Leichsenring J, Flechtenmacher C, et al. Tubular, lactating, and mammography, sonography, and aspiration cytology in 10 cases. AJR Am J
ductal adenomas are devoid of MED12 Exon2 mutations, and ductal adenomas Roentgenol 1991;157:715–719.
show recurrent mutations in GNAS and the PI3K-AKT pathway. Genes Chro- 60. Chao TC, Lo YF, Chen SC, et al. Sonographic features of phyllodes tumors of the
mosomes Cancer 2017;56:11–17. breast. Ultrasound Obstet Gynecol 2002;20:64–71.
28. Ross DS, Giri DD, Akram MM, et al. Fibroepithelial Lesions in the Breast of 61. Wurdinger S, Herzog AB, Fischer DR, et al. Differentiation of phyllodes breast
Adolescent Females: a Clinicopathological Study of 54 Cases. Breast J tumors from fibroadenomas on MRI. AJR Am J Roentgenol 2005;185:1317–1321.
2017;23:182–192. 62. Keelan PA, Myers JL, Wold LE, et al. Phyllodes tumor: clinicopathologic review
29. Fekete P, Petrek J, Majmudar B, et al. Fibroadenomas with stromal cellularity. A of 60 patients and flow cytometric analysis in 30 patients. Hum Pathol
G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452 451

1992;23:1048–1054. 97. Adem C, Reynolds C, Adlakha H, et al. Wide spectrum screening keratin as a
63. Tan PH, Thike AA, Tan WJ, et al. Predicting clinical behaviour of breast phyl- marker of metaplastic spindle cell carcinoma of the breast: an im-
lodes tumours: a nomogram based on histological criteria and surgical mar- munohistochemical study of 24 patients. Histopathology 2002;40:556–562.
gins. J Clin Pathol 2012;65:69–76. 98. Carter MR, Hornick JL, Lester S, et al. Spindle cell (sarcomatoid) carcinoma of
64. Norris HJ, Taylor HB. Relationship of histologic features to behavior of cysto- the breast: a clinicopathologic and immunohistochemical analysis of 29 cases.
sarcoma phyllodes. Analysis of ninety-four cases Cancer 1967;20:2090–2099. Am J Surg Pathol 2006;30:300–309.
65. Cohn-Cedermark G, Rutqvist LE, Rosendahl I, et al. Prognostic factors in cy- 99. Aranda FI, Laforga JB, Lopez JI. Phyllodes tumor of the breast. An im-
stosarcoma phyllodes. A clinicopathologic study of 77 patients. Cancer munohistochemical study of 28 cases with special attention to the role of
1991;68:2017–2022. myofibroblasts. Pathol Res Pract 1994;190:474–481.
66. Moffat CJ, Pinder SE, Dixon AR, et al. Phyllodes tumours of the breast: a clin- 100. Chia Y, Thike AA, Cheok PY, et al. Stromal keratin expression in phyllodes
icopathological review of thirty-two cases. Histopathology 1995;27:205–218. tumours of the breast: a comparison with other spindle cell breast lesions. J
67. Tan EY, Tan PH, Yong WS, et al. Recurrent phyllodes tumours of the breast: Clin Pathol 2012;65:339–347.
pathological features and clinical implications. ANZ J Surg 2006;76:476–480. 101. Lee AH. Recent developments in the histological diagnosis of spindle cell
68. Tan PH, Jayabaskar T, Chuah KL, et al. Phyllodes tumors of the breast: the role carcinoma, fibromatosis and phyllodes tumour of the breast. Histopathology
of pathologic parameters. Am J Clin Pathol 2005;123:529–540. 2008;52:45–57.
69. Gullett NP, Rizzo M, Johnstone PA. National surgical patterns of care for pri- 102. Moore T, Lee AH. Expression of CD34 and bcl-2 in phyllodes tumours, fi-
mary surgery and axillary staging of phyllodes tumors. Breast J 2009;15:41–44. broadenomas and spindle cell lesions of the breast. Histopathology
70. Macdonald OK, Lee CM, Tward JD, et al. Malignant phyllodes tumor of the 2001;38:62–67.
female breast: association of primary therapy with cause-specific survival from 103. Cimino-Mathews A, Subhawong AP, Elwood H, et al. Neural crest transcrip-
the Surveillance, Epidemiology, and End Results (SEER) program. Cancer tion factor Sox10 is preferentially expressed in triple-negative and metaplastic
2006;107:2127–2133. breast carcinomas. Hum Pathol 2013;44:959–965.
71. Barrio AV, Clark BD, Goldberg JI, et al. Clinicopathologic features and long-term 104. Cimino-Mathews A, Subhawong AP, Illei PB, et al. GATA3 expression in breast
outcomes of 293 phyllodes tumors of the breast. Ann Surg Oncol 2007;14:2961–2970. carcinoma: utility in triple-negative, sarcomatoid, and metastatic carcinomas.
72. Chua CL, Thomas A, Ng BK. Cystosarcoma phyllodes–Asian variations. Aust N Z J Hum Pathol 2013;44:1341–1349.
Surg 1988;58:301–305. 105. Yoshida M, Sekine S, Ogawa R, et al. Frequent MED12 mutations in phyllodes
73. Keelan PA, Myers JL, Wold LE, et al. Phyllodes Tumor - Clinicopathological tumours of the breast. Br J Cancer 2015;112:1703–1708.
Review of 60 Patients and Flow Cytometric Analysis in 30 Patients. Human 106. Tse GM, Lee CS, Kung FY, et al. Hormonal receptors expression in epithelial
Pathol 1992;23:1048–1054. cells of mammary phyllodes tumors correlates with pathologic grade of the
74. Nielsen VT, Andreasen C. Phyllodes tumour of the male breast. Histopathology tumor: a multicenter study of 143 cases. Am J Clin Pathol 2002;118:522–526.
1987;11:761–762. 107. Sawyer EJ, Hanby AM, Rowan AJ, et al. The Wnt pathway, epithelial-stromal
75. Konstantakos AK, Graham DJ. Cystosarcoma phyllodes tumors in men. Am Surg interactions, and malignant progression in phyllodes tumours. J Pathol
2003;69:808–811. 2002;196:437–444.
76. Giacomazzi J, Koehler-Santos P, Palmero EI, et al. A TP53 founder mutation, p. 108. Tsang JY, Mendoza P, Lam CC, et al. Involvement of alpha- and beta-catenins
R337H, is associated with phyllodes breast tumors in Brazil. Virchows Arch and E-cadherin in the development of mammary phyllodes tumours. Histo-
2013;463:17–22. pathology 2012;61:667–674.
77. Foucar CE, Hardy A, Siziopikou KP, et al. A mother and daughter with phyllodes 109. Dietrich CU, Pandis N, Rizou H, et al. Cytogenetic findings in phyllodes tumors
tumors of the breast. Clin Breast Cancer 2012;12:373–377. of the breast: karyotypic complexity differentiates between malignant and
78. Birch JM, Alston RD, McNally RJ, et al. Relative frequency and morphology of benign tumors. Hum Pathol 1997;28:1379–1382.
cancers in carriers of germline TP53 mutations. Oncogene 2001;20:4621–4628. 110. Lae M, La Rosa P, Mandel J, et al. Whole-genome profiling helps to classify
79. Powell CM, Rosen PP. Adipose differentiation in cystosarcoma phyllodes. A phyllodes tumours of the breast. J Clin Pathol 2016;69:1081–1087.
study of 14 cases. Am J Surg Pathol 1994;18:720–727. 111. Ang MK, Ooi AS, Thike AA, et al. Molecular classification of breast phyllodes
80. Tsang AK, Chan SK, Lam CC, et al. Phyllodes tumours of the breast - differ- tumors: validation of the histologic grading scheme and insights into malig-
entiating features in core needle biopsy. Histopathology 2011;59:600–608. nant progression. Breast Cancer Res Treat 2011;129:319–329.
81. Choi J, Koo JS. Comparative study of histological features between core needle 112. Lae M, Vincent-Salomon A, Savignoni A, et al. Phyllodes tumors of the breast
biopsy and surgical excision in phyllodes tumor. Pathol Int 2012;62:120–126. segregate in two groups according to genetic criteria. Mod Pathol
82. Krings G, Chen YY. Fibroepithelial Lesions. In: Shin S, ed. A Comprehensive Guide 2007;20:435–444.
to Core Needle Biopsies of the Breast. 1st ed., Switzerland: Spinger International; 113. Lu YJ, Birdsall S, Osin P, et al. Phyllodes tumors of the breast analyzed by
2016:241–319. comparative genomic hybridization and association of increased 1q copy
83. Chaney AW, Pollack A, McNeese MD, et al. Primary treatment of cystosarcoma number with stromal overgrowth and recurrence. Genes Chromosomes Cancer
phyllodes of the breast. Cancer 2000;89:1502–1511. 1997;20:275–281.
84. Jones AM, Mitter R, Springall R, et al. A comprehensive genetic profile of 114. Lv S, Niu Y, Wei L, et al. Chromosomal aberrations and genetic relations in
phyllodes tumours of the breast detects important mutations, intra-tumoral benign, borderline and malignant phyllodes tumors of the breast: a comparative
genetic heterogeneity and new genetic changes on recurrence. J Pathol genomic hybridization study. Breast Cancer Res Treat 2008;112:411–418.
2008;214:533–544. 115. Tan WJ, Lai JC, Thike AA, et al. Novel genetic aberrations in breast phyllodes
85. Kracht J, Sapino A, Bussolati G. Malignant phyllodes tumor of breast with lung tumours: comparison between prognostically distinct groups. Breast Cancer
metastases mimicking the primary. Am J Surg Pathol 1998;22:1284–1290. Res Treat 2014;145:635–645.
86. Burga AM, Tavassoli FA. Periductal stromal tumor: a rare lesion with low-grade 116. Ng CC, Tan J, Ong CK, et al. MED12 is frequently mutated in breast phyllodes
sarcomatous behavior. Am J Surg Pathol 2003;27:343–348. tumours: a study of 112 cases. J Clin Pathol 2015;68:685–691.
87. Lawton TJ, Acs G, Argani P, et al. Interobserver variability by pathologists in the 117. Yoon N, Bae GE, Kang SY, et al. Frequency of MED12 mutations in phyllodes
distinction between cellular fibroadenomas and phyllodes tumors. Int J Surg tumors: inverse correlation with histologic grade. Genes Chromosomes Cancer
Pathol 2014;22:695–698. 2016;55:495–504.
88. Tan BY, Acs G, Apple SK, et al. Phyllodes tumours of the breast: a consensus 118. Lien HC, Huang CS, Yang YW, et al. MED12 exon 2 mutation as a highly
review. Histopathology 2016;68:5–21. sensitive and specific marker in distinguishing phyllodes tumours from other
89. Tse GM, Law BK, Chan KF, et al. Multinucleated stromal giant cells in mammary spindle neoplasms of the breast. APMIS 2016;124:356–364.
phyllodes tumours. Pathology 2001;33:153–156. 119. Yoshida M, Ogawa R, Yoshida H, et al. TERT promoter mutations are frequent
90. Jara-Lazaro AR, Akhilesh M, Thike AA, et al. Predictors of phyllodes tumours on and show association with MED12 mutations in phyllodes tumors of the
core biopsy specimens of fibroepithelial neoplasms. Histopathology breast. Br J Cancer 2015;113:1244–1248.
2010;57:220–232. 120. Lyle PL, Bridge JA, Simpson JF, et al. Liposarcomatous differentiation in ma-
91. Resetkova E, Khazai L, Albarracin CT, et al. Clinical and radiologic data and core lignant phyllodes tumours is unassociated with MDM2 or CDK4 amplification.
needle biopsy findings should dictate management of cellular fibroepithelial Histopathology 2016;68:1040–1045.
tumors of the breast. Breast J 2010;16:573–580. 121. Hodges KB, Abdul-Karim FW, Wang M, et al. Evidence for transformation of
92. Tan WJ, Cima I, Choudhury Y, et al. A five-gene reverse transcription-PCR assay fibroadenoma of the breast to malignant phyllodes tumor. Appl Im-
for pre-operative classification of breast fibroepithelial lesions. Breast Cancer munohistochem Mol Morphol 2009;17:345–350.
Res 2016;18:31. 122. Sperber F, Blank A, Metser U, et al. Diagnosis and treatment of breast fi-
93. National Comprehensive Cancer Network (NCCN) Guidelines Version 1. 2015 broadenomas by ultrasound-guided vacuum-assisted biopsy. Arch Surg
Phyllodes Tumor. Fort Washington, P.A.2015. Available from: 〈http://www.nccn. 2003;138:796–800.
org/professionals/physician_gls/f_guidelines.asp〉. 123. Kaufman CS, Littrup PJ, Freeman-Gibb LA, et al. Office-based cryoablation of
94. Lim SZ, Selvarajan S, Thike AA, et al. Breast sarcomas and malignant phyllodes breast fibroadenomas with long-term follow-up. Breast J 2005;11:344–350.
tumours: comparison of clinicopathological features, treatment strategies, 124. Nurko J, Mabry CD, Whitworth P, et al. Interim results from the Fi-
prognostic factors and outcomes. Breast Cancer Res Treat 2016;159:229–244. broAdenoma Cryoablation Treatment Registry. Am J Surg 2005;190:647–651
95. Cimino-Mathews A, Sharma R, Illei PB, et al. A subset of malignant phyllodes [discussion 651-2].
tumors express p63 and p40: a diagnostic pitfall in breast core needle biopsies. 125. Grady I, Gorsuch H, Wilburn-Bailey S. Long-term outcome of benign fi-
Am J Surg Pathol 2014;38:1689–1696. broadenomas treated by ultrasound-guided percutaneous excision. Breast J
96. Dunne B, Lee AH, Pinder SE, et al. An immunohistochemical study of meta- 2008;14:275–278.
plastic spindle cell carcinoma, phyllodes tumor and fibromatosis of the breast. 126. Chen WH, Cheng SP, Tzen CY, et al. Surgical treatment of phyllodes tumors of
Hum Pathol 2003;34:1009–1015. the breast: retrospective review of 172 cases. J Surg Oncol 2005;91:185–194.
452 G. Krings et al. / Seminars in Diagnostic Pathology 34 (2017) 438–452

127. Mangi AA, Smith BL, Gadd MA, et al. Surgical management of phyllodes tu- 133. Moutte A, Chopin N, Faure C, et al. Surgical management of benign and
mors. Arch Surg 1999;134:487–492 [discussion 492-3]. borderline phyllodes tumors of the breast. Breast J 2016;22:547–552.
128. Chng TW, Lee JY, Lee CS, et al. Validation of the Singapore nomogram for 134. Cowan ML, Argani P, Cimino-Mathews A. Benign and low-grade fibroepithelial
outcome prediction in breast phyllodes tumours: an Australian cohort. J Clin neoplasms of the breast have low recurrence rate after positive surgical
Pathol 2016;69:1124–1126. margins. Mod Pathol 2016;29:259–265.
129. Nishimura R, Tan PH, Thike AA, et al. Utility of the Singapore nomogram for 135. Khosravi-Shahi P. Management of non metastatic phyllodes tumors of the
predicting recurrence-free survival in Japanese women with breast phyllodes breast: review of the literature. Surg Oncol 2011;20:e143–e148.
tumours. J Clin Pathol 2014;67:748–750. 136. Guillot E, Couturaud B, Reyal F, et al. Management of phyllodes breast tumors.
130. Belkacemi Y, Bousquet G, Marsiglia H, et al. Phyllodes tumor of the breast. Int Breast J 2011;17:129–137.
J Radiat Oncol Biol Phys 2008;70:492–500. 137. Barth Jr RJ, Wells WA, Mitchell SE, et al. A prospective, multi-institutional
131. Teo JY, Cheong CS, Wong CY. Low local recurrence rates in young Asian pa- study of adjuvant radiotherapy after resection of malignant phyllodes tumors.
tients with phyllodes tumours: less is more. ANZ J Surg 2012;82:325–328. Ann Surg Oncol 2009;16:2288–2294.
132. Yom CK, Han W, Kim SW, et al. Reappraisal of conventional risk stratification 138. Telli ML, Horst KC, Guardino AE, et al. Phyllodes tumors of the breast: natural
for local recurrence based on clinical outcomes in 285 resected phyllodes history, diagnosis, and treatment. J Natl Compr Canc Netw 2007;5:324–
tumors of the breast. Ann Surg Oncol 2015;22:2912–2918. 330.

You might also like