10.1007@978 3 319 62539 3
10.1007@978 3 319 62539 3
10.1007@978 3 319 62539 3
Anna Sapino
Janina Kulka Editors
Breast
Pathology
Encyclopedia of Pathology
Series Editor
J. H. J. M. van Krieken
The scope of this 15–20-volume set encompasses the entire field of pathology
ranging from general pathological terms to specific diseases to diagnostic
methods. Published as print edition and online version (eReference) in the
Springer Reference Program each topical volume sticks out by clearly and
homogenously structured entries. A team of international experts guarantee
that the essays and definitions are scientifically sound. The A-Z format of each
topical volume allows readers to quickly and easily find the information they
need. The major advantage of the encyclopedia is the way it makes relevant
information available not only to pathologists, but also to all clinicians and
researchers of the neighboring disciplines working together with pathologists
who occasionally might wish to look up terms online.
Breast Pathology
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
To our Masters and Teachers,
Dr. Gianni Bussolati
Dr. Pietro Maria Gullino
Dr. Judit Mohácsy
Dr. John Douglas Davies
ThiS is a FM Blank Page
Series Preface
When Denis Diderot started the first encyclopedia in the eighteenth century, it
was a groundbreaking and timely event. It was the time of the Enlightenment,
and knowledge was seen as something which was to be spread to many and to
build upon by creating new knowledge. His ambition was to bring all available
knowledge together in one series of books so that every person who could read
has access to all there is to know. Nowadays, in a time of easily accessible
knowledge, the question is whether there is still need of an encyclopedia. It is
obvious that the amount of knowledge is such that it is not possible to bring it
all together in one encyclopedia. One may argue that the Internet is the
encyclopedia of today, but that misses an important point of Diderot, a point
that is probably even more valid today. He created a team that valued infor-
mation and selected what was worth to be presented in the encyclopedia. He
recognized that science is not a democratic process where the majority decides
what is true and valuable, but rather a growing body of knowledge in which
radical ideas from individuals may bring about huge changes, even though
most would reject these new ideas in the beginning. Indeed, the Internet lacks
such authority and it is not easy to select valuable information from nonsense,
especially when one is not an expert in a certain field.
It is therefore that an encyclopedia is only as good as the team that creates
it. It goes without saying the team that is responsible for the Encyclopedia of
Pathology consists of recognized experts in the field. Pathology is a growing
medical discipline in which the amount of information is probably already
more than that the whole encyclopedia of Diderot contained. For experts in
subspecialties within pathology, it is already almost impossible to keep an
overview on new developments and to select relevant from less relevant new
information. There are plenty of textbooks for every disease group, and
scientific literature is available for most pathologists through PubMed or
GoogleScholar. What is lacking is a systematic overview of what we know
in an alphabetical order, easily accessible to all. The encyclopedia of pathology
fills that gap. It is written by experts with the general pathologist in mind and
also specialist from other disciplines. It will consist of a series of volumes on
subspecialties, and when it is completed there will be an online version
combining these. Yearly updates from the online version is foreseen and
readers are welcome to provide suggestions for improvement. These will be
judged by the editorial team in order to keep the encyclopedia authoritative yet
using the expertise of many.
vii
viii Series Preface
Finally, it is my hope that the encyclopedia will grow into a reliable body of
knowledge in pathology, enabling communication through a common lan-
guage, and that it will grow and adapt to new developments.
ix
Editors Biography
Dr. Anna Sapino obtained her M.D. degree at the University of Turin (Italy)
in 1982 and completed her residency in anatomic pathology at the University
of Milan in 1986. She started her career as a consultant pathologist at
the University Hospital in Turin in 1984 under the supervision of Prof.
Gianni Bussolati. In 1987 she accrued experience in experimental studies on
pre-neoplastic breast lesions through a sabbatical period spent in the USA at
the Michigan Cancer Foundation and at the Columbia University. Upon her
return to Italy, she set up her own cell biology lab working on effects of
hormones on breast cancer cells and mouse mammary gland organ cultures.
Her research activity has always been paralleled and inspired by breast cancer
diagnostic pathology and her career orientation reflects this approach. In 1998
Dr. Sapino was appointed Associate Professor and in 2005 full Professor of
Anatomic Pathology and Histopathology at the School of Medicine of the
University of Turin. From 2010 to 2015 she was recruited as Director of
Surgical Pathology at the University Hospital (Città della Salute e della
Scienza) in Turin and from 2013 to 2015 as Director of the Department of
Laboratory Medicine, then she moved to Candiolo Cancer Centre FPO-IRCCS
(Italy) as Director of the Pathology Unit. This unit is recognized as training
center for breast pathology by the European Society of Pathology (ESP). In
2017 she became Scientific Director of Candiolo Cancer Institute FPO-
IRCCS, a private nonprofit institution endorsed by the Italian Ministry of
Health for oncology research. In 2018 Dr. Sapino had also been appointed
Director of the Department of Medical Sciences of the School of Medicine at
the University of Turin (Italy). She has been member of the European Working
Group for Screening of Breast Pathology and coauthor of the European
Guideline for Breast Cancer Screening. From 2014 to 2018 she had been
chairing the European Working Group of Breast Pathology of the ESP. She
is member of the teaching staff for breast pathology of the European School of
xi
xii Editors Biography
xv
Contributors
xvii
xviii Contributors
Noëlle Weingertner and Jean-Pierre Bellocq Puerperal abscess is the natural course of acute
Department of Pathology, Strasbourg University puerperal mastitis. It occurs at the end of preg-
Hospital, Strasbourg, France nancy but especially during lactation. Clinically,
it presents with the same symptoms as mastitis
but with a palpable mass (Mahoney and Ingram
Synonyms 2014). This mass is painful and inflammatory, with
a purulent flow at incision and possible general
Pyogenic mastitis signs (fever and malaise). Spontaneous evolution
may be fistulization. There is usually a context of
cracked nipple or skin abrasion, leading to retro-
Definition grade infection. Staphylococcus aureus is the most
common organism responsible (50% of cases), but
A breast abscess is defined as a collection of Staphylococcus epidermidis and streptococci are
infected fluid or pus within the breast. occasionally isolated. Breast abscesses associated
Breast abscesses are mainly classified as with methicillin-resistant Staphylococcus aureus
“puerperal” (or “lactational”) – related to (MRSA) has been reported and is likely to be an
childbirth – and “non-puerperal.” In a non- increasing problem. Typhoid is a well-recognized
puerperal context, the subareolar abscess is cause of breast abscess in countries where this
the most frequently observed. Other rarer disease is common (Kataria et al. 2013).
situations include peripheral non-puerperal
abscesses, neonatal infections, skin-associated Clinical Features
infections, infections associated with breast • Incidence
surgery, with medical or non-medical proce- Puerperal abscesses affect 1–9% of breast-
dures, and unusual infections. Abscesses repre- feeding mothers (Mahoney and Ingram 2014).
sent 3–4% of all benign breast masses (Kasales Infection is more frequent following a first
et al. 2014). child and occurs most commonly within the
In this chapter, we will detail the most frequent first 6 weeks of breastfeeding. Risk factors
presentations of breast abscesses, namely, puer- include the first child at maternal age over
peral abscesses and non-puerperal periareolar or 30, gestation of more than 41 weeks, and mas-
subareolar abscesses. titis (Kataria et al. 2013). Breast abscesses may
Subareolar abscesses present with acute, keratin plugs (Fig. 4). A foreign-body-like reaction
subacute, or chronic inflammatory infiltrate with to keratin may also be seen (Kasales et al. 2014).
fibrosis. Some subareolar abscesses may show
ducts with squamous metaplasia that contain Differential Diagnosis
Subareolar abscesses have to be differentiated
from breast cancer, especially inflammatory
carcinoma.
Subareolar abscess are distinct from duct
ectasia (▶ Duct Ectasia and Periductal Mastitis).
Duct ectasia affects an older age group and is not
associated with smoking. The subareolar ducts are
dilated and the inflammation is less extensive and
less active.
Granulomatous lobular mastitis (▶ Granulo-
matous Mastitis) (GLM) may be associated with
abscess formation. This condition should be differ-
entiated from non-puerperal subareolar abscesses,
in which granulomatous inflammation may
develop in the late stages. In contrast to subareolar
abscess, GLM is generally peripheral and lobulo-
centric, is not associated with epithelial duct squa-
mous metaplasia and with smoking, but is associ-
ated with pregnancy (Pereira et al. 2012).
Others
Abscess, Fig 4 Non-puerperal subareolar abscess – epidermis of the nipple (arrow) and malpighian metaplasia
microscopic features (A: H&E x4 and B: H&E x20). of the distal part of a galactophorous duct (star)
Dense, subacute, inflammatory infiltrate beyond the
Acinic Cell Carcinoma 5
Neonatal breast infection and abscess Habif, D. V., Perzin, K. H., Lipton, R., & Lattes, R. (1970).
are uncommon but can occur in the first few Subareolar abscess associated with squamous metapla-
sia of the lactiferous ducts. American Journal of
weeks of life when the breast bud is enlarged. Surgery, 119, 523–526. A
Staphylococcus aureus is the usual organism, but Kasales, C. J., Han, B., Stanley Smith, J., Jr.,
occasionally it may be due to Escherichia coli Chetlen, A. L., Kaneda, H. J., & Shereef, S. (2014).
(Weidner 2012). Nonpuerperal mastitis and subareolar abscess of
the breast. American Journal of Roentgenology, 202,
Primary infection of the skin of the breast, W133–W139.
which can present as cellulitis or an abscess, most Kataria, K., Srivastava, A., & Dhar, A. (2013). Manage-
commonly affects the skin of the lower half of the ment of lactational mastitis and breast abscesses:
breast. These infections are often recurrent in review of current knowledge and practice. Indian
Journal of Surgery, 75(6), 430–435.
women who are overweight, have large breasts Mahoney, M. C., & Ingram, A. D. (2014). Breast
or have poor personal hygiene, and in people emergencies: types, imaging features, and manage-
with skin conditions such as eczema. Epidermal ment. American Journal of Roentgenology, 202(4),
cysts are common in the skin of the breast and W390–W399.
Pereira, F. A., Mudgil, A. V., Macias, E. S., &
may become infected. Hidradenitis suppurativa Karsif, K. (2012). Idiopathic granulomatous lobular
may affect the breast. Pilonidal abscesses affect- mastitis. International Journal of Dermatology, 51(2),
ing the nipple have been reported in hairdressers 142–151.
or sheep shearers (Weidner 2012). Staphylococ- Weidner, N. (2012). Infections of the breast. In D. J. Dabbs
(Ed.), Breast pathology (pp. 34–43). Philadelphia:
cus aureus is the usual causative organisms in Elsevier Saunders.
skin-associated infections (Weidner 2012).
Breast infection and abscesses may occur
following needle biopsy, or surgery for breast
cancer, breast implants or mammoplasty. Acinic Cell Carcinoma
Nipple rings can cause recurrent infection,
particularly in smokers. Maria P. Foschini and Luca Morandi
Factitial diseases are created by the patient. Such Department of Biomedical and Neuromotor
patients may undergo many investigations before Sciences (DIBINEM), Unit of Anatomic
the nature of the disease is recognized. The diagnosis Pathology at Bellaria Hospital, University of
is difficult to establish but should be considered Bologna, Bologna, Italy
when the clinical situation appears suspicious.
Immunophenotype
and most importantly, none of these latter tumors Damiani, S., Pasquinelli, G., Lamovec, J., Peterse, J. L., &
show serous acinar differentiation, demonstrated Eusebi, V. (2000). Acinic cell carcinoma of the breast:
An immunohistochemical and ultrastructural study.
with anti-lysozyme, anti-salivary type amylase, Virchows Archiv, 437, 74–81.
and anti-alpha-1- antichimotrypsin. Eusebi, V., Ichihara, S., Vincent-Salomon, A., Sniege, A.,
Secretory carcinoma (▶ Invasive Secretory Car- & Sapino, A. (2012). Exceptionally rare types and
cinoma) of the breast can simulate ACC. Differen- variants. In S. R. Lakhani, I. O. Ellis, S. J. Schnitt,
P. H. Tan, & M. van de Vijver (Eds.), WHO classifica-
tial diagnosis is based on the cytological atypias tion of tumours of the breast (4th ed., p. 75). Lyon:
that in secretory carcinoma are quite bland, while IARC Press.
are more marked in ACC. In addition secretory Foschini, M. P., Morandi, L., Asioli, S., Giove, G.,
breast carcinoma is characterized by a typical Corradini, A. G., & Eusebi, V. (2017). The morpholog-
ical spectrum of salivary gland type tumours of the
ETV6 rearrangement that is absent in ACC. breast. Pathology, 49, 215–227.
Differential diagnosis between ACC with Guerini-Rocco, E., Hodi, Z., Piscuoglio, S., Ng, C. K.,
prominent microglandular pattern and micro- Rakha, E. A., Schultheis, A. M., Marchiò, C., da
glandular adenosis (MGA) (▶ Microglandular Cruz, P. A., De Filippo, M. R., Martelotto, L. G., De
Mattos-Arruda, L., Edelweiss, M., Jungbluth, A. A.,
Adenosis) is a more controversial issue, and Fusco, N., Norton, L., Weigelt, B., Ellis, I. O., & Reis-
recent papers describe similar molecular fea- Filho, J. S. (2015). The repertoire of somatic genetic
tures. According to the original morphological alterations of acinic cell carcinomas of the breast: An
description (Clement and Azzopardi 1983), exploratory, hypothesis-generating study. Journal of
Pathology, 237, 166–178.
MGA is characterized by small glandular struc- Piscuoglio, S., Hodi, Z., Katabi, N., Guerini-Rocco, E.,
tures lined by clear cells with empty cytoplasm, Macedo, G. S., Ng, C. K., Edelweiss, M., De Mattos-
surrounded by basal membrane. On the contrary, Arruda, L., Wen, H. Y., Rakha, E. A., Ellis, I. O., Rubin,
ACC with microglandular pattern is composed B. P., Weigelt, B., & Reis-Filho, J. S. (2015). Are acinic
cell carcinomas of the breast and salivary glands dis-
of cells with eosinophilic cytoplasm, with tinct diseases? Histopathology, 67, 529–537.
intracytoplasmic granules (demonstrated both Ripamonti, C. B., Colombo, M., Mondini, P., Siranoush,
with PAS after diastase digestion and electron M., Peissel, B., Bernard, L., Radice, P., & Carcangiu,
microscopy) and absence of basal membrane. M. L. (2013). First description of an acinic cell carci-
noma of the breast in a BRCA1 mutation carrier: A case
report. BMC Cancer, 13, 46.
Roncaroli, F., Lamovec, J., Zidar, A., & Eusebi, V. (1996).
References and Further Reading Acinic cell-like carcinoma of the breast. Virchows
Archiv, 429, 69–74.
Clement, P. B., & Azzopardi, J. G. (1983). Microglandular Shimao, K., Haga, S., Shimizu, T., Imamura, H.,
adenosis of the breast – A lesion simulating tubular Watanabe, O., Kinoshita, J., Nagumo, H., Utada, Y.,
carcinoma. Histopathology, 7, 169–180. Okabe, T., Kajiwara, T., Oshibe, N., & Aiba,
Conlon, N., Sadri, N., Corben, A. D., & Tan, L. K. (2016). M. (1998). Acinic cell adenocarcinoma arising in the
Acinic cell carcinoma of breast: Morphologic and breast of a young male: A clinicopathological, immu-
immunohistochemical review of a rare breast cancer nohistochemical and ultrastructural study. Breast Can-
subtype. Human Pathology, 51, 16–24. cer, 5, 77–81.
10 Adenoid Cystic Carcinoma
Adenoid Cystic
Carcinoma, Fig. 1 AdCC
classical variant, showing
a central part with A
cribriform architecture and
a peripheral part (arrows)
with tubular architecture.
The AdCC with tubular
architecture shows
infiltrative pattern of
growth, invading the fatty
tissue (H&E)
Adenoid Cystic
Carcinoma, Fig. 2 AdCC
classical variant with
tubular architecture is
composed of elongated
glands, surrounded by two
or more cell layers. Focally
structures typical of AdCC
with two types of mucins
are visible (H&E)
features and nuclear atypia. Atypical mitotic fig- cells are positive with high molecular weight CK,
ures are frequent; necrosis can be present as well as CK 14 and CK 5\6, p63 (Fig. 7 (a) p63
as perineural invasion (Figs. 5 and 6 H&E). When immunostaining in nuclei of myoepithelial cells in
perineural invasion is present a SB-AdCC variant cribriform and (b) and tubular components).
should always be suspected. Myoepithelial cells are evidenced by myoepithelial
AdCC with high-grade transformation: In rare markers, as smooth muscle actin (SMA), calponin,
cases, in addition to the classical AdCC features, caldesmon. Basal cell markers, as collagen IV and
areas of high-grade tumor of different origin laminin, stain the basal membrane present in the
(melanoma, spindle cell carcinoma, ▶ Invasive pseudoluminal spaces (Fig. 8 immunostaining with
Carcinoma NST, and ▶ Malignant Adenomyoe- Collagen IV). CD117 (cKit) is strongly present in
pithelioma (M-AME)) are present. The presence of all the epithelial components. Proliferative markers,
these high-grade tumor areas has prognostic impact, as Ki67, stain a variable percentage of neoplastic
as metastases and disease progression can occur. cells. High proliferative index is usually encoun-
tered in the SB-AdCC variant or in the areas of
high-grade transformation.
Immunophenotype
Adenoid Cystic
Carcinoma, Fig. 3 AdCC
classical variant with
cribriform architecture
(a and b) shows neoplastic
nests composed of cells
with mild atypia,
surrounding two types of
spaces. Glandular spaces
contain an epithelial type of
mucin that is weakly
basophilic on H&E stained
sections. Pseudoglandular
spaces contain eosinophilic
basal membrane (H&E)
progesterone receptors (PR) are usually negative Epidermal growth factor receptor 1 (EGFR)
and HER2 protein is not overexpressed (Foschini protein can be overexpressed, even if this over-
et al. 2017). A weak expression for androgen recep- expression is not accompanied by gene amplifica-
tor (AR) has been described (Vranic et al. 2010). tion (Vranic et al. 2010).
Nevertheless, papers, based on large cohorts of MYB-NFIB fusion gene has been detected in
AdCC, report focal ER positivity in a minority of the majority of AdCC (Martelotto et al. 2015); in
cases. In our experience ER positivity can be seen addition it is maintained in SB-AdCC and in
in a minority of epithelial cells in AdCC classical AdCC with high-grade transformation (Fusco
variant, while ER and PR are completely absent et al. 2016; Righi et al. 2011).
in SB-AdCC and in AdCC with high-grade The mutational profile of AdCC has been
transformation. described using whole exome sequencing
A novel isoform of ER, namely ER-alpha36, (Martelotto et al. 2015), which showed a sub-
has been detected in AdCC of the breast. This stantial heterogeneity with lack of recurrent
novel isoform differs from the classical isoform mutations. The mutation rate was found to be
(ER-alpha66) as it does not show the transcrip- very low (0.27 nonsilent mutations/Mb), similar
tional activation domains; in addition, on to that reported for pediatric malignancies and
immunohistochemically stained sections, it is salivary gland AdCCs (0.31 nonsilent mutations/
mainly localized in the cytoplasm and on the cell Mb), and lower than that reported for triple neg-
membrane. It mediates nonclassic (nongenomic) ative breast cancers (TNBCs) (1.38 nonsilent
estrogen signaling, therefore opening the possibil- mutations/Mb). Unlike common TNBCs and
ity of novel therapies (Vranic et al. 2011). basal-like breast cancers, no somatic mutations
Adenoid Cystic Carcinoma 13
Adenoid Cystic Carcinoma, Fig. 4 SB-AdCC variant shows a multinodular type of growth, with neoplastic nodules
varying in size from a few millimeters (a) to larger neoplastic masses (b) (H&E)
Adenoid Cystic
Carcinoma, Fig. 5 SB-
AdCC at higher power
shows cellular atypia,
mitoses, and necrosis
(H&E)
targeting TP53, PIK3CA, RB1, BRCA1, or have been detected. Many of the mutations iden-
BRCA2 were identified in breast AdCCs. The tified were likely clonal with a cancer cell frac-
most frequently mutated genes are: MYB, tion >80%. Evaluating gene copy number
BRAF, FBXW7, SMARCA5, SF3B1, FGFR2, alterations, low level of genetic instability and
TLN2, PRKD1, RASA1, PTPN11, and MTOR. no amplifications or homozygous deletions were
On rare occasions mutations in RAS pathway identified.
14 Adenoid Cystic Carcinoma
When AdCC shows high-grade transformation, firm the morphological hypothesis, that high-grade
progression to high-grade TNBC was found to transformation is related to selected neoplastic
involve clonal shifts with enrichment of mutations clones that acquire additional somatic mutations.
affecting EP300, NOTCH1, ERBB2, FGFR1,
KMT2C, STAG2, KDM6A, CDK12. These data con-
Differential Diagnosis
Adenoid Cystic
Carcinoma, Fig. 7 p63
immunostaining shows the
presence of basal and
myoepithelial cells both in
the cribriform (a) and in the
tubular (b) components
Adenoid Cystic Carcinoma 15
References and Further Reading EGFR gene amplification. Human Pathology, 41,
1617–1623.
Brill, L. B., Kanner, W. A., Fehr, A., Andrén, Y., Vranic, S., Gatalica, Z., Deng, H., Frkovic-Grazio, S.,
Moskaluk, C. A., Löning, T., Stenman, G., & Frierson, Lee, L. M., Gurjeva, O., & Wang, Z. Y. (2011).
H. F., Jr. (2011). Analysis of MYB expression and ER-a36, a novel isoform of ER-a66, is commonly
MYB-NFIB gene fusions in adenoid cystic carcinoma over-expressed in apocrine and adenoid cystic carci-
and other salivary neoplasms. Modern Pathology, 24, nomas of the breast. Journal of Clinical Pathology,
1169–1176. 64, 54–57.
D’Alfonso, T. M., Mosquera, J. M., MacDonald, T. Y., Wetterskog, D., Wilkerson, P. M., Rodrigues, D. N.,
Padilla, J., Liu, Y. F., Rubin, M. A., & Shin, S. J. (2014). Lambros, M. B., Fritchie, K., Andersson, M. K.,
MYB-NFIB gene fusion in adenoid cystic carcinoma of Natrajan, R., Gauthier, A., Di Palma, S., Shousha, S.,
the breast with special focus paid to the solid variant with Gatalica, Z., Töpfer, C., Vukovic, V., A’Hern, R.,
basaloid features. Human Pathology, 45, 2270–2280. Weigelt, B., Vincent-Salomon, A., Stenman, G., Rubin,
Foschini, M. P., & Krausz, T. (2010). Salivary gland-type B. P., & Reis-Filho, J. S. (2013). Mutation profiling of
tumors of the breast: A spectrum of benign and malig- adenoid cystic carcinomas from multiple anatomical
nant tumors including “triple negative carcinomas” of sites identifies mutations in the RAS pathway, but no
low malignant potential. Seminar in Diagnostic Pathol- KIT mutations. Histopathology, 62, 543–550.
ogy, 27, 77–90.
Foschini, M. P., Rizzo, A., De Leo, A., Laurino, L., Sironi,
M., & Rucco, V. (2016). Solid variant of adenoid cystic
carcinoma of the breast: A case series with proposal of a
new grading system. International Journal of Surgical
Adenomyoepithelioma
Pathology, 24(2), 97–102.
Foschini, M. P., Morandi, L., Asioli, S., Giove, G., Eugenio Maiorano1, Enrica Macorano2 and
Corradini, A. G., & Eusebi, V. (2017). The morpholog- Mauro Giuseppe Mastropasqua2
ical spectrum of salivary gland type tumours of the 1
Department of Emergency and Organ
breast. Pathology, 49, 215–227.
Fusco, N., Geyer, F. C., De Filippo, M. R., Martelotto, Transplantation, Unit of Pathology, University of
L. G., Ng, C. K., Piscuoglio, S., Guerini-Rocco, E., Bari, Bari, Italy
Schultheis, A. M., Fuhrmann, L., Wang, L., Jungbluth, 2
Department of Emergency and Organ
A. A., Burke, K. A., Lim, R. S., Vincent-Salomon, A.,
Bamba, M., Moritani, S., Badve, S. S., Ichihara, S.,
Transplantation, Universitá degli Studi di Bari
Ellis, I. O., Reis-Filho, J. S., & Weigelt, B. (2016). Aldo Moro – Policlinico, Bari, Italy
Genetic events in the progression of adenoid cystic
carcinoma of the breast to high-grade triple-negative
breast cancer. Modern Pathology, 29, 1292–1305.
Martelotto, L. G., De Filippo, M. R., Ng, C. K., Natrajan, R.,
Definition
Fuhrmann, L., Cyrta, J., Piscuoglio, S., Wen, H. C., Lim,
R. S., Shen, R., Schultheis, A. M., Wen, Y. H., Edelweiss, Adenomyoepithelioma (AME) is a benign tumor of
M., Mariani, O., Stenman, G., Chan, T. A., Colombo, the breast, composed by a biphasic proliferation of
P. E., Norton, L., Vincent-Salomon, A., Reis-Filho, J. S.,
phenotypically distinct myoepithelial cells and
& Weigelt, B. (2015). Genomic landscape of adenoid
cystic carcinoma of the breast. Journal of Pathology, luminal epithelial cells.
237, 179–189. Following the first description by Hamperl
Righi, A., Lenzi, M., Morandi, L., Flamminio, F., De (1970), Tavassoli (1991) identified three histo-
Biase, D., Farnedi, A., & Foschini, M. P. (2011).
Adenoid cystic carcinoma of the breast
logic variants, in which myoepithelial and epithe-
associated with invasive duct carcinoma: A case lial cells are present in variable percentages,
report. International Journal of Surgical Pathology, namely, spindle, tubular, and lobulated AME
19, 230–234. (Tavassoli 1991).
Shin, S. J., & Rosen, P. P. (2002). Solid variant of mam-
AME with malignant features (▶ Malignant
mary adenoid cystic carcinoma with basaloid features:
A study of nine cases. American Journal of Surgical Adenomyoepithelioma (M-AME)) have seldom
Pathology, 26, 413–420. been reported (Hayes 2011) and named AME
Vranic, S., Frkovic-Grazio, S., Lamovec, J., Serdarevic, with carcinoma, when epithelial cells or myoepi-
F., Gurjeva, O., Palazzo, J., Bilalovic, N., Lee, L. M.,
thelial cells show malignant cytomorphological
& Gatalica, Z. (2010). Adenoid cystic carcinomas
of the breast have low Topo IIa expression but fre- features and malignant AME when these are pre-
quently overexpress EGFR protein without sent in both components (Brogi 2014).
Adenomyoepithelioma 17
Clinical Features nodule. Its average size is 2.5 cm, ranging from
0.5 to 8.0 cm, without distinctive discoloration,
• Incidence the cut surface appearing as tan, gray, white, yel- A
AME is an extremely rare tumor, a few small low, or pink (Rosen 1987). Occasionally, a small
case series and single case reports having been multicystic component or multinodulation may
reported so far. (Decorsiere et al. 1988; McLaren be detected (Laforga et al. 1998). Its clinical pre-
et al. 2005; Loose et al. 1992; Rosen 1987; sentations include nipple discharge, pain, or
Tavassoli 1991). AME with carcinoma and tenderness at palpation and, mammographically,
malignant AME are even more infrequent than it appears as a rounded or lobulated, well-
their benign counterpart (Ahmed et al., Ahmed circumscribed dense mass, sometimes showing
and Heller 2000). microcalcifications, while ultrasonography high-
• Age lights a solid or cystic structure (Park et al. 2013;
It mostly occurs in postmenopausal women, Lee et al. 2010). Quite interestingly, strong
with sporadic cases occurring both in younger uptake of 18 F-Fluorodeoxyglucose by Positron
(26 and 27 year old; Loose et al. 1992; Rosen Emission Tomography has recently been reported
1987) and older patients (84 year old; Berna (Oba et al. 2017); this should be taken into
et al. 1997). account as a potential pitfall of clinico-
• Sex instrumental investigations.
Women are far more commonly affected, with
sporadic cases described in males (Berna et al.
1997; Tamura et al. 1993).
Microscopy
• Site
The cytomorphological features of AME have
AME usually is a solitary and unilateral mass,
been described in occasional cases or small case
which more commonly occurs in the peripheral
series (Chang et al. 2002; Hock and Chan 1994;
parts of the breast, and only occasionally may
Kurashina 2002; Mercado et al. 2007; Ng 2002;
be centrally located (Adejolu et al. 2011).
Niemann et al. 1995; Saad et al. 2012; Valente and
• Treatment
Stuckey 1994; Zhang et al. 2004): clusters of
The standard treatment is surgical excision
epithelial and myoepithelial cells represent the
with clear margins (Rosen 1987). Currently,
major finding, the latter also displaying occasional
adjuvant treatments are considered only for
intracytoplasmic vacuolization. Mild to moderate
the management of those exceptional cases of
nuclear atypia was also noted, in the absence of
AME with carcinoma.
necrosis or mitotic activity. Nevertheless, the
• Outcome
diagnosis of AME was not achieved, and based
AME is considered a benign neoplasm; never-
on fine needle aspiration cytology, most cases
theless, local recurrence has been reported in
were interpreted as benign neoplasms but suspi-
sporadic cases, usually more than 2 years after
cion of malignancy was raised in some instances
surgical excision (Loose et al. 1992) and pos-
(Brogi 2014). Proper identification of the
sibly ascribed to incomplete tumor eradication.
myoepithelial cell component, admixed with duc-
AME with carcinoma may present either as
tal cells, seem the most reliable tool to rule out
synchronous or metachronous lesions, follow-
malignancy.
ing recurrence of previously excised AMEs
In histologic preparations, AME usually is
(Tavassoli 1991).
sharply separated from adjacent tissues and may
show discrete fibrous encapsulation. Different
Macroscopy morphological patterns may distinctly or simulta-
neously be present, according to the variable per-
AME generally is a circumscribed, uniformly centage of myoepithelial and epithelial cells in
solid, sometimes lobulated, firm and small individual lesions (Hock and Chan 1994). The
18 Adenomyoepithelioma
spindle cell growth pattern consists in a predom- infrequently occur, possibly as a consequence of
inant proliferation of myoepithelial cells, some- infarction, while cartilaginous metaplasia is
times showing clear and vacuolated (“soap exceptionally uncommon.
bubble”) cytoplasms, and sparse or hardly visible The detection of infiltrative growth towards the
epithelial-lined spaces (Fig. 1), resembling surrounding tissues, necrosis, prominent cyto-
myoepithelioma (Weidner et al., Weidner and logic atypia, and exuberant mitotic activity in
Levine 1988).
In the tubular pattern, glandular spaces are
far more evident (Fig. 2), oval or rounded in
shape, reminiscent of tubular adenoma. They
are composed by epithelial cells with darkly
stained cytoplasms and slightly hyperchromatic
nuclei. Myoepithelial cells may surround epithe-
lial cells in a basal position or form small
clusters. They appear as small cuboidal cells
with barely visible cytoplasm or as larger cells
with evident cytoplasmic clearing; occasionally,
the exuberant proliferation of myoepithelial cells
may lead to compression of the lumen in tubular
structures.
In the lobulated variant (Fig. 3), the growth Adenomyoepithelioma,Fig.2 Adenomyoepithelioma–
pattern is mostly solid and the tumor nests include tubular variant: duct-like structures are well evident, oval
an admixture of clear, eosinophilic, and or rounded in shape, and composed by epithelial cells, with
plasmacytoid myoepithelial cells along with com- darkly stained cytoplasms and slightly hyperchromatic
nuclei and myoepithelial cells in a basal position. Occa-
pressed epithelial-lined spaces. A fibro-sclerotic sionally, the latter may be assembled in small clusters and
core is at times evident. show either scant cytoplasm or prominent cytoplasmic
Papillary structures may seldom be present and clearing. The exuberant proliferation of myoepithelial
apocrine metaplasia may be detected in such areas cells may lead to compression of the lumen in tubular
structures (H&E 100)
(Eusebi et al. 1987); squamous metaplasia may
Immunophenotype
Dewar, R., Fadare, O., & Gilmore, H. (2011). Best prac- Laforga, J. B., Aranda, F. I., & Sevilla, F. (1998).
tices in diagnostic immunohistochemistry: Adenomyoepithelioma of the breast: Report of two
Myoepithelial markers in breast pathology. Archives cases with prominent cystic changes and intranuclear
of Pathology & Laboratory Medicine, 135, 422–429. inclusions. Diagnostic Cytopathology, 19, 55–58. A
Eusebi, V., Casadei, G. P., & Bussolati, G. (1987). Lee, J. H., Kim, S. H., & Kang, B. J. (2010). Ultrasono-
Adenomyoepithelioma of the breast. A spectrum of graphic features of benign adenomyoepithelioma of the
biologic behavior. The American Journal of Surgical breast. Korean Journal of Radiology, 11, 522–527.
Pathology, 16, 868–876. Loose, J. H., Patchefsky, A. S., & Hollander, I. J. (1992).
Foschini, M. P., Pizzicannella, G., & Peterse, J. L. (1995). Adenomyoepithelioma of the breast. A spectrum of
Adenomyoepithelioma of the breast associated with biologic behavior. The American Journal of Surgical
low-grade adenosquamous and sarcomatoid carcino- Pathology, 16, 868–876.
mas. Virchows Archiv, 427, 243–250. McLaren, B. K., Smith, J., & Schuyler, P. A. (2005).
Gatalica, Z., Velagaleti, G., & Kuivaniemi, H. (2005). Adenomyoepithelioma: Clinical, histologic, and
Gene expression profile of an adenomyoepithelioma immunohistologic evaluation of a series of related
of the breast with a reciprocal translocation involving lesions. The American Journal of Surgical Pathology,
chromosomes 8 and 16. Cancer Genetics and Cytoge- 29, 1294–1299.
netics, 156, 14–22. Mercado, C. L., Toth, H. K., & Axelrod, D. (2007). Fine-
Guarino, M., Raele, D., & Squillaci, S. (1993). Ductal needle aspiration biopsy of benign adenomyoe-
adenoma of the breast. An immunohistochemical pithelioma of the breast: Radiologic and pathologic
study of five cases. Pathology, Research and Practice, correlation in four cases. Diagnostic Cytopathology,
189, 515–520. 35, 690–694.
Gusterson, B. A., Sloane, J. P., & Middwood, C. (1987). Michal, M., Baumruk, L., & Burger, J. (1994). Adenomyoe-
Ductal adenoma of the breast – A lesion exhibiting a pithelioma of the breast with undifferentiated carcinoma
myoepithelial/epithelial phenotype. Histopathology, component. Histopathology, 24, 274–276.
11, 103–110. Narita, T., & Matsuda, K. (1995). Pleomorphic adenoma of
Hamperl, H. (1970). Normal state, regressive changes, hyper- the breast: Case report and review of the literature.
plasia, tumors. Current Topics in Pathology, 53, 194–198. Pathology International, 45, 441–447.
Han, J. S., & Peng, Y. (2010). Multicentric adenomyoe- Nevado, M., Lopez, J. I., & Dominguez, M. P. (1991).
pithelioma of the breast with atypia and associated Pleomorphic adenoma of the breast. Case report.
ductal carcinoma in situ. Breast J, 16, 547–549. APMIS, 99, 866–868.
Hayes, M. M. (2011). Adenomyoepithelioma of the breast: Ng, W. K. (2002). Adenomyoepithelioma of the breast.
A review stressing its propensity for malignant transfor- A review of three cases with reapprasail of the fine needle
mation. Journal of Clinical Pathology, 64, 477–484. aspiration biopsy findings. Acta Cytologica, 46, 317–324.
Hegy, L., Thway, K., & Newton, R. (2009). Malignant Niemann, T. H., Benda, J. A., & Cohen, M. B. (1995).
myoepithelioma arising in adenomyoepithelioma of Adenomyoepithelioma of the breast: Fine-needle aspi-
the breast and coincident multiple gastrointestinal stro- ration biopsy and histologic findings. Diagnostic Cyto-
mal tumours in a patient with neurofibromatosis type pathology, 12, 245–250.
1. Journal of Clinical Pathology, 62, 653–655. Oba, T., Maeno, K., Ono, M., Iesato, A., Ito, T., Kanai, T.,
Hock, Y. L., & Chan, S. Y. (1994). Adenomyoepithelioma Mochizuki, Y., Ito, K.I., Yoshizawa, A. & Takayama, F.
of the breast. A case report correlating cytologic and (2017). A case of adenomyoepithelioma of the breast
histologic features. Acta Cytologica, 38, 953–956. showing strong uptake of 18 F-Fluorodeoxyglucose on a
Honda, Y., & Iyama, K. (2009). Malignant adenomyoe- positron emission tomography. Breast J, 23, 220–224.
pithelioma of the breast combined with invasive lobular Oka, K., Sando, N., & Moriya, T. (2007). Malignant
carcinoma. Pathology International, 59, 179–184. adenomyoepithelioma of the breast with matrix pro-
Iyengar, P., Cody, H. S., & Brogi, E. (2005). Pleomorphic duction may be compatible with one variant form of
adenoma of the breast: Case report and review of the matrix-producting carcinoma: A case report. Pathol-
literature. Diagnostic Cytopathology, 33, 416–420. ogy, Research and Practice, 203, 599–604.
Jones, C., Tooze, R., & Lakhani, S. R. (2003). Malignant Park, Y. M., Park, J. S., & Jung, H. S. (2013). Imaging
adenomyoepithelioma of the breast metastasizing to the features of benign adenomyoepithelioma of the breast.
liver. Virchows Archiv, 442, 504–506. Journal of Clinical Ultrasound, 41, 218–223.
Kihara, M., Yokomise, H., & Irie, A. (2001). Malignant Qureshi, A., Kayani, N., & Gulzar, R. (2009). Malignant
adenomyoepithelioma of the breast with lung metasta- adenomyoepithelioma of the breast: A case report with
ses: Report of a case. Surgery Today, 31, 899–903. review of literature. BML Case Reports, 2009.
Kurashina, M. (2002). Fine-needle aspiration cytology of Rasbridge, S. A., & Millis, R. R. (1998). Adenomyoe-
benign and malignant adenomyoepithelioma: Report of pithelioma of the breast with malignant features.
two cases. Diagnostic Cytopathology, 26, 29–34. Virchows Archiv, 432, 123–130.
Kuroda, N., Fujishima, N., & Ohara, M. (2008). Coexistent Reid-Nicholson, M., Bleiwess, I., & Pace, B. (2003). Pleo-
adenomyoepithelioma and invasive ductal carcinoma morphic adenoma of the breast. A case report and
of the breast: Presentation as separate tumor. Medical distinction from mucinous carcinoma. Archives of
Molecular Morphology, 41, 238–242. Pathology & Laboratory Medicine, 127, 474–477.
22 Adenosis, Other Types
Adenosis, Other Types, Fig. 1 Adenomyoepithelial Adenosis, Other Types, Fig. 3 Adenosis tumor
adenosis (H&Ex50) (H&Ex25)
Adenosis, Other Types, Fig. 2 Adenomyoepithelial Adenosis, Other Types, Fig. 4 Adenosis tumor (p63 IHC
adenosis (H&Ex400) reaction x63)
Adenosis, Other Types 25
adenomyoepithelial adenosis but differs from Oberman, H. A. (1984). Benign breast lesions confused
them by the interdigitating tubular configuration; with carcinoma. In R. W. McDivtt, H. A. Oberman,
L. Ozzello, N. Kaufman (Eds.), The breast. Baltimore:
it also differs from microglandular adenosis Williams and Wilkins. United States-Canadian
due to the presence of myoepithelial cells. The Academy of Pathology Monographs in Pathology,
most important differential diagnosis is with 25, 1–33.
tubular carcinoma (▶ Tubular Carcinoma). The Rageth, C. J., O’Flynn, E. A. M., Comstock, C., Kurtz, C.,
Kubik, R., Madjar, H., Lepori, D., Kampmann, G.,
absence of angular shaped tubules with occa- Mundinger, A., Baege, A., Decker, T., Hosch, S.,
sional transverse bars, a lack of prominent lumi- Tausch, C., Delaloye, J. F., Morris, E., & Varga, Z.
nal snouts, a generally acellular collagenous (2016). First International Consensus Conference on
stroma rather than a desmoplastic one and the lesions of uncertain malignant potential in the breast
(B3 lesions). Breast Cancer Research and Treatment,
presence of the myoepithelial cells highlighted 159, 203–213.
by immunostaining, support the diagnosis of Reston, V. A., & American College of Radiology. (1998).
tubular adenosis (Lee et al. 1996). In core biopsy Illustrated breast imaging report and system
this lesion is classified in the B3 category. (BI-RADS), 3rd edn. American College of Radiology
5B 6.
Sloane, J. P. (1985). Non-neoplastic epithelial changes. In
J. P. Sloane (Ed.), Biopsy pathology of the breast,
References and Further Reading Biopsy pathology series (pp. 69–92). London:
Chapman and Hall Medical.
Bonser, G. M., Dossett, J. A., & Jull, J. W. (1961). Human Tavassoli, F. A. (1991). Myoepithelial lesions of the breast.
and experimental breast cancer. London: Pitman Myoepitheliosis, adenomyoepithelioma and
Medical Publications Co. Ltd. myoepithelial carcinoma. American Journal of
Da Silva, L., Buck, L., Simpson, P. T., Reid, L., Surgical Pathology, 15, 554–568.
McCallum, N., Madigan, B. J., & Lakhani, Tsuda, H., Mukai, K., Fukutomi, T., & Hirohashi, S.
S. R. (2009). Molecular and morphological analysis of (1994). Malignant progression of adenomyoepithelial
adenoid cystic carcinoma of the breast with synchronous adenosis of the breast. Pathology International,
tubular adenosis. Virchows Archive, 454, 107–114. 44, 475–479.
DiPiro, P., Gulizia, J. A., Lester, S. C., & Meyer, J. E. (2000).
American Journal of Radiology, 175, 31–34.
Erel, S., Tuncbilek, I., Kismet, K., Kilicoglu, B., Ozer, E.,
& Mehmet, A. A. (2008). Adenomyoepithelial
adenosis of the breast: Clinical, radiological and path- Angiosarcoma of the Breast
ological findings for differential diagnosis. Breast
Cancer, 3, 427–430.
European guidelines for quality assurance in breast Alberto Pisacane
cancer screening and diagnosis. (2006). European Unit of Pathology, Candiolo Cancer Institute
Commission, 4th edn 6a (pp. 221–256). FPO-IRCCS, Candiolo, TO, Italy
Hamperl, H. (1970) The myoepithelia (myoepithelial cells)
normal state; regressive changes; hyperplasia; tumors.
In H-W. Altmann, et al., (Eds.), Current topics in
pathology (Vol. 53, pp. 161–213). Berlin/Heidelberg: Definition
Springer.
Heller, E. L., & Fleming, J. C. (1950). Fibrosing A tumor composed of malignant cells with endo-
adenomatosis of the breast. American Journal of
Clinical Pathology, 20, 141–146. thelial differentiation.
Kiaer, H., Nielsen, B., Paulsen, S., Sørensen, I. M.,
Dyrebog, U., & Blichert-Tft, M. (1984). Adenomyoe-
pithelial adenosis and low-grade malignant Clinical Features
adenomyoepithelioma of the breast. Virchows Archive
(A), 405, 55–67.
Lee, K. C., Chan, J. K., & Gwi, E. (1996). Tubular adenosis • Incidence
of the breast: A distinctive lesion mimicking invasive Angiosarcoma (AS) of the breast is a rare dis-
carcinoma. American Journal of Surgical Pathology, ease accounting for about 1% of all soft tissue
20, 46–54.
breast tumors.
Nielsen, B. B. (1987). Adenosis tumor of the breast – a
clinic – pathological investigation of 27cases. It may present as primary AS, without pre-
Histopathology, 11, 1259–1275. vious history of mammary carcinoma or any
Angiosarcoma of the Breast 27
associated factor, or as secondary AS, most Low-grade tumors confer better outcomes
commonly associated with previous breast in terms of disease-free survival interval.
irradiation. Conversely, no statistically significant corre- A
Primary AS represents <0.05% of all pri- lation between tumor size and likelihood of
mary breast cancers. local recurrence has been identified in pri-
Secondary AS presents a median of mary AS, as far as no correlation seems to
10.5 years after radiotherapy for breast cancer. exist between tumor grade and the rate of
In those patients undergoing breast conserving local recurrence, distant metastases and
surgery with adjuvant radiotherapy, the esti- death owing to disease.
mated incidence of radiation-induced AS is
0.05–0.3%.
Macroscopy
AS rarely occurs after mastectomy with
axillary dissection without irradiation and dur-
In secondary AS single or multiple erythematous-
ing pregnancy.
violaceous nodules and/or papules may be present
• Age
on the breast skin. On gross examination AS
Primary AS more frequently affects young and
appear as poorly circumscribed, brown and hem-
middle-aged women (median age 30–55 ys).
orrhagic masses, friable, spongy or firm in breast
Secondary AS presents in older women
parenchyma.
(median age 67–71 ys) after radiotherapy for
breast cancer.
• Sex
Microscopy
Rare cases of AS have been described in male
breast.
Microscopic spectrum ranges from tumors dem-
• Site
onstrating well-formed vessels with mild cyto-
Primary AS arises within the breast paren-
logic atypia, to morphologically atypical and
chyma and usually presents with fullness or
undifferentiated pleomorphic sarcoma
swelling, which can rapidly grow forming pal-
(Nascimento et al. 2008).
pable mass or masses, without skin changes.
Tumors are histologically graded using
Secondary AS arises in the cutaneous tissue
Rosen’s three-tier system into low (I), intermedi-
and might secondarily invade breast
ate (II), or high (III) grade.
parenchyma.
Low-grade AS (Fig. 1 H&E) are composed of
• Treatment
complex anastomosing vascular channels. The
Because of rarity of AS, few data are available
endothelium is flat with minimal nuclear atypia
to support treatment decisions. Surgical resec-
tion is key, with mastectomy preferred over
breast-conserving therapy. Lymph node dis-
section is not routinely recommended.
The role of adjuvant radiotherapy and che-
motherapy is poorly defined, with many agents
showing activity and varying clinical benefit. It
is generally agreed that multimodality therapy
is the best for this aggressive tumor.
• Outcome
The 5-year survival rate of secondary AS
patients is in the range of 27%–48%. The
5-year disease-free survival rate is 35%.
Tumor size, grade, and number of foci are Angiosarcoma of the Breast, Fig. 1 Low-grade AS:
important prognostic factors. neoplastic vessels dissect mammary parenchyma (H&E)
28 Angiosarcoma of the Breast
and no cellular stratification. Tumor dissects the channels, often with prominent areas of hemor-
stroma, causing distortion with little destruction rhage (so called “blood lakes”). Tumor cells are
of the preexisting lobules and ducts. The mitotic mostly spindled or epithelioid and show diffuse
rate is low (median 2/10 HPF). Necrosis is gener- and prominent nuclear atypia. Mitotic figures are
ally absent. easily observed. Necrosis is frequent, and the
Intermediate-grade AS (Fig. 2 H&E) are more tumor growth is infiltrating with dissection of
cellular and are characterized by intermingled adjacent breast parenchyma. High-grade AS may
areas of low and moderate nuclear atypia. The contain low- or intermediate-grade elements,
endothelial cells lining neoplastic vessels are gen- especially at the periphery of the lesion.
erally plump, multilayered, frequently arranged in
papillary structures. Solid areas are not present.
Immunophenotype
Mitotic rate is low (median 3/10 HPF). Necrosis
may be present.
Fli-1, FactorVIII, CD31, CD34, and ERG used in
High-grade AS (Fig. 3) show a generally solid
combination have the greatest sensitivity (94%)
growth pattern with very poorly defined vascular
and specificity (up to 100%).
Molecular Features
Differential Diagnosis
The most challenging differential diagnoses of
breast AS are represented by low-grade pseudo-
angiomatous and angiomatous proliferations.
Above all, hemangiomas, pseudoangiomatous
stromal hyperplasia (PASH), and atypical vascu-
Angiosarcoma of the Breast, Fig. 2 Intermediate-grade lar lesions (AVLs) must be kept in mind (Bowman
AS: cellular area of moderate nuclear atypia; an entrapped et al. 2012; Rosen et al. 1988).
duct is in the center (H&E)
Hemangiomas and other benign vascular
tumors are generally well-circumscribed lesions
composed of well-formed vascular channels and
lack a dissecting growth pattern within breast
parenchyma (▶ Hemangioma of the Breast).
PASH is a benign proliferation of stromal
myofibroblasts, which express CD34 and focally
smooth muscle actin or desmin, but not other
more specific endothelial markers (CD31, Factor
VIII, or ERG) (▶ Pseudoangiomatous Stromal
Hyperplasia).
AVLs, typically develop 2–5 years after
radiotherapy and is histologically characterized
by a focal proliferation of dilated, sometimes
anastomosing, vascular channels lined by a sin-
Angiosarcoma of the Breast, Fig. 3 High-grade AS:
cellular area of solid growth with very poorly defined gle layer of endothelial cells in the papillary and
vascular channels (H&E) reticular dermis. Although AVLs lack cytologic
Apocrine Carcinoma 29
• Incidence
Apocrine Carcinoma Invasive apocrine carcinoma is a rare special
type of breast cancer constituting 0.3–4% of all
Semir Vranic1,2 and Zoran Gatalica3 breast cancers (depending on the criteria used
1
Department of Pathology, Clinical Centre and for its definition). When strictly defined as
School of Medicine, University of Sarajevo, above, it constitutes ~1% of all breast cancers.
Sarajevo, Bosnia and Herzegovina • Age
2
College of Medicine, Qatar University, Typically affects postmenopausal women.
Doha, Qatar A recent comprehensive analysis based on the
3
Caris Life Sciences, Phoenix, AZ, USA SEER [The Surveillance, Epidemiology, and
End Results] data confirmed the prevalence of
apocrine carcinomas among the elderly women
Synonyms (Zhang et al. 2017).
• Sex
Carcinoma with apocrine differentiation; Invasive Invasive apocrine carcinoma predominantly
apocrine carcinoma affects women although rare cases of apocrine
30 Apocrine Carcinoma
carcinoma (both in situ and invasive) among due to encysted (encapsulated) lesions with pap-
males have also been described (Vranic illary architecture and apocrine morphology
et al. 2013). (Vranic et al. 2013).
• Site
The tumor location of invasive apocrine carci-
noma does not differ from that of invasive Microscopy
carcinoma of no special type (NST) (▶ Inva-
sive Carcinoma NST). Apocrine cells have abundant eosinophilic/granu-
• Treatment lar cytoplasm (called “type A” cells), with cen-
Current therapeutic approach to the patients trally/eccentrically located nuclei and prominent
with invasive apocrine carcinoma is similar to nucleoli (positive for Periodic Acid Schiff
that of breast carcinoma NST. A subset of staining, PAS). The tumor cells usually show dis-
invasive apocrine carcinomas (30–50%) are tinct cell borders (Fig. 1a). Another, less common
HER-2/neu positive (amplified) and are ame- type of apocrine cells (called “type B” cells)
nable for anti-HER2 treatment modalities. exhibits foamy and vacuolated cytoplasm resem-
Consistent AR expression and activation of bling histiocytes (O’Malley and Lakhani 2012;
AR signaling pathways may represent a new Vranic et al. 2013). The tumor cells show marked
avenue for the anti-AR treatment modalities of nuclear pleomorphism and atypia while the num-
patients with advanced and/or metastatic apo- ber of mitotic figures (easily appreciated by light
crine carcinomas. microscopy) may vary. Invasive apocrine carcino-
• Outcome mas are typically grade 2 or 3 carcinomas (graded
Clinical studies reporting outcome (overall by the Elston-Ellis modification of the Bloom and
survival and disease-free survival) of the Richardson grading system). Apocrine ductal car-
patients with invasive apocrine carcinoma cinoma in situ (DCIS) is commonly seen in asso-
are contradictory due to the lack of consistent ciation with its invasive counterpart. Apocrine
criteria for its definition (O’Malley and DCIS is usually high-grade DCIS with
Lakhani 2012; Vranic et al. 2013). A recent comedonecrosis and calcifications. Similar to
SEER analysis, based on 840 patients with its ductal counterpart, a subset of apocrine
invasive apocrine carcinoma, revealed that carcinomas may develop through a multistep pro-
apocrine carcinomas tend to behave more gression process (“linear progression model”):
aggressively in comparison with NST carci- Apocrine hyperplasia (metaplasia)!apocrine
nomas. However, the worse outcome was atypia!apocrine DCIS!invasive apocrine
significantly diminished after matching for carcinoma) (Fig. 2) (Costa et al. 2013; Gromov
demographic and clinicopathologic character- et al. 2015). This model has also been confirmed
istics (Zhang et al. 2017). by a molecular study that revealed common
mutations in both preinvasive (e.g., apocrine
DCIS) and invasive apocrine carcinomas (Costa
Macroscopy et al. 2013).
Apocrine Carcinoma, Fig. 1 Hematoxylin and Eosin positively for androgen receptor (AR). Her-2/neu protein
(H&E) staining of a case of invasive carcinoma of the was overexpressed exhibiting a complete, intense mem-
breast with a characteristic apocrine morphology; the brane positivity in the majority of neoplastic apocrine cells
tumor cells are negative for ER-a66 (ER), but stain (HER2).
Apocrine Carcinoma, Fig. 2 A concept of apocrine carcinoma progression and development: Apocrine hyperplasia
(metaplasia)!apocrine atypia!apocrine DCIS!invasive apocrine carcinoma.
ER-alpha36 variant that locates to the cytoplasm [acyl-CoA synthetase medium-chain family mem-
and cytoplasmic membrane (Vranic et al. 2011). ber 1]), and negative biomarkers (ER, PR, Bcl-2,
Apocrine cells are also positive for gross cystic GATA-3) (Gromov et al. 2015). Recently, additional
disease fluid protein-15 (GCDFP-15) and novel biomarkers of apocrine differentiation have
negative for Bcl-2 (Gatalica 1997). Celis et al. been identified (HMGCS2 and FABP7) (Gromov
defined a specific “apocrine protein signature” et al. 2015).
that includes positive biomarkers AR, CD24, HER-2 protein is overexpressed (with under-
15-PDGH [prostaglandin dehydrogenase], ACMS1 lying HER-2/neu gene amplification) in 30–50%
32 Apocrine Carcinoma
of invasive apocrine carcinomas (Fig. 1d) (Vranic factor receptor 1 (IGF-1R) expression appears to
et al. 2013). be downregulated in apocrine carcinomas.
Invasive apocrine carcinomas typically stain for
luminal cytokeratins including CK7, CK8/18, and
CK9/19. CK20 expression has also been described Differential Diagnosis
in one study (Shao et al. 2012). Basal markers
(basal cytokeratins [CK5/6, CK14, CK17], p63, Apocrine differentiation may be seen in invasive
P-cadherin, S-100, CD10, vimentin) are usually carcinomas NST (▶ Invasive Carcinoma NST) as
absent or positive in only small subset of invasive well as some special types such as pleomorphic
apocrine carcinomas, while EGFR protein is pre- lobular carcinoma (▶ Pleomorphic Lobular Carci-
sent in ~50–60% of cases (with uncommon EGFR noma) with which apocrine carcinoma shares some
gene alterations) (Vranic et al. 2013). morphologic and molecular characteristics; anti-
body to E-cadherin can aid diagnosis in such
cases as apocrine carcinomas predominantly
Molecular Features (>80%) retain E-cadherin expression in contrast
to pleomorphic lobular carcinomas. Primary breast
“Molecular apocrine breast cancers” exhibit a oncocytic carcinoma (▶ Invasive Oncocytic Carci-
poor prognostic gene signature with a high-risk noma) and a subset of lipid-rich and secretory car-
recurrence score and overall poor outcome. If cinomas (▶ Invasive Secretory Carcinoma) may
strictly defined by morphology and ER-/AR+ ste- also pose diagnostic dilemmas as well as metaplas-
roid receptor profile, invasive apocrine carcino- tic carcinomas (▶ Invasive Metaplastic Carci-
mas are either triple-negative (50–70%) or HER2- noma), which latter also may have morphologic
positive breast carcinomas (30–50%). characteristics similar to apocrine carcinomas
Several comprehensive molecular studies (e.g., eosinophilic cytoplasm in squamous cells).
revealed common mutations of TP53 and Apocrine carcinomas may be occasionally con-
PIK3CA/PTEN/AKT genes. Interestingly, patients fused with a granular cell tumor (▶ Granular Cell
with germline PTEN mutations (Cowden syn- Tumor) (S-100 and pancytokeratins may help),
drome, OMIM #158350) are prone to develop while apocrine carcinomas composed of “type B”
breast carcinomas with apocrine differentiation. cells may resemble a benign histiocytic
Mutations within the mitogen-activated protein (inflammatory) lesion (CD68, CD163, and
kinase [MAPK] pathway (KRAS, NRAS, BRAF) pancytokeratins are helpful).
may also be occasionally seen in a subset of In addition, metastatic neoplasms to the breast
invasive apocrine carcinomas. Other targetable exhibiting eosinophilic cytoplasm may mimic apo-
mutations are much less common (Vranic et al. crine carcinoma. These include metastatic malig-
2013, 2017). nant melanoma and oncocytic carcinomas. In such
HER-2/neu gene is amplified in 30–50% of cases, a comprehensive immunohistochemical
invasive apocrine carcinomas, while HER-2/neu workup along with the clinical history may help
gene mutations are rare. EGFR protein expression render the correct diagnosis.
is common while underlying EGFR gene alter-
ations are uncommon: EGFR gene amplification
is present <10% of the cases and EGFR gene
References
mutations have not been described so far. Simi- Costa, L. J., Justino, A., Gomes, M., Alvarenga, C. A.,
larly, cMET overexpression is present in ~25% of Gerhard, R., Vranic, S., Gatalica, Z., Machado, J. C., &
the cases without cMET gene alterations (Vranic Schmitt, F. (2013). Comprehensive genetic characteri-
et al. 2017). Growth hormone-releasing hormone zation of apocrine lesions of the breast. Cancer
Research, 73, 2013.
(GHRH) and its receptors (GHRH-R) are also
Gatalica, Z. (1997). Immunohistochemical analysis of apo-
overexpressed in vast majority of invasive apo- crine breast lesions. Consistent over-expression of
crine carcinomas. In contrast, insulin-like growth androgen receptor accompanied by the loss of estrogen
Atypical Ductal Hyperplasia 33
and progesterone receptors in apocrine metaplasia and micropapillae, arcades, Roman bridges, bars
apocrine carcinoma in situ. Pathology Research and crossing the glandular space, and finally full-
Practice, 193, 753–758.
Gromov, P., Espinoza, J. A., & Gromova, I. (2015). blown lesions with solid or cribriform growth A
Molecular and diagnostic features of apocrine breast (Fig. 1). As discussed below, the definition of
lesions. Expert Review of Molecular Diagnostics, 15, ADH has undergone remarkable changes. It is
1011–1022. the great achievement of Azzopardi who shifted
O’Malley, F., & Lakhani, S. R. (2012). Carcinoma with
apocrine differentiation. In S. R. Lakhani, I. O. Ellis, our view from usual ductal hyperplasia to ductal
S. J. Schnitt, P. H. Tan, & M. J. van de Vijver (Eds.), in situ microcancer as the decisive step in early
World Health Organization classification of tumours of breast cancer development.
the breast (4th ed., pp. 53–54). Lyon: International In rare cases, atypical ductal proliferation
Agency of Research and Cancer (IARC).
Shao, M. M., Chan, S., Yu, A., Lam, C., Tsang, J., Lui, P., (ADH/DCIS) may evolve from primarily benign
Law, B., Tan, P. H., & Tse, G. (2012). Keratin expres- proliferative breast lesions, or they may show
sion in breast cancers. Virchows Archiv, 461, 313–322. apocrine features (see below).
Vranic, S., Gatalica, Z., Frkovic-Grazio, S., Deng, H.,
Lee, L. M. J., Gurjeva, O., & Wang, Z. Y. (2011).
ER-a36 a novel isoform of ER-a66 is commonly over-
expressed in apocrine and adenoid cystic carcinoma of Clinical Features
the breast. Journal of Clinical Pathology, 64, 54–57.
Vranic, S., Schmitt, F., Sapino, A., Costa, J. L., Castro, M., • Incidence
Reddy, S., & Gatalica, Z. (2013). Apocrine carcinoma
of the breast: A comprehensive review. Histology and The frequency of ADH diagnosis has multi-
Histopathology, 28, 1393–1409. plied with the advent of mammographic
Vranic, S., Feldman, R., & Gatalica, Z. (2017). Apocrine screening with figures of 12–17% of cases
carcinoma of the breast: A brief update on the molec- performed due to the presence of micro-
ular features and targetable biomarkers. Bosnian
Journal of Basic Medical Sciences, 17, 9–11. calcifications. In biopsies from mass lesions
Zhang, N., Zhang, H., Chen, T., & Yang, Q. (2017). Dose identified in screening programs, ADH is
invasive apocrine adenocarcinoma has worse prognosis only found in 2–4% of specimens.
than invasive ductal carcinoma of breast: Evidence • Age
from SEER database. Oncotarget, 8, 24579–24592.
Late 40s, screening programs.
• Sex
Atypical Ductal Hyperplasia Female
• Site
Werner Boecker Either breast, any quadrant, probably prefer-
Gerhard Domagk-Institute of Pathology, ence of upper outer quadrant.
University of Münster, Münster, • Clinic, Treatment, and Core Biopsy
North-Rhine Westphalia, Germany ADH, generally, is asymptomatic. Clustered
microcalcifications (Fig. 2) are thought to be
the most common findings on a screening
Synonyms mammogram that usually require minimal
invasive biopsy (MIB) techniques such as
Atypical intraductal hyperplasia; Ductal vacuum-assisted core biopsy. However, due
intraepithelial neoplasia 1B (DIN 1B) to the limited amount of tissue in core biopsies,
ADH cannot be reliably diagnosed with
these techniques because the atypical epithelial
Definition foci in these biopsies may form part of an
established in situ neoplastic lesion, with or
Atypical ductal hyperplasia (ADH) is defined as a without associated invasion. For this reason,
local proliferation of evenly spaced monotonous the European Working Group for Breast Screen-
cells with an atypical architecture, characterized ing Pathology (EWGBSP) recommended to
by smooth geometrical growth patterns as abandon the term ADH in core biopsies in
34 Atypical Ductal Hyperplasia
favor of “atypical epithelial proliferation, ductal categorized as B3, and an indication for a
type” (AEPDT) (Wells et al. 2006). In the diagnostic biopsy to exclude a DCIS should
five category reporting system of core needle be made. Nevertheless, the number of foci of
biopsies, which was initially proposed by the atypical ductal proliferation and the number
UK National Coordinating Group for Breast of cores involved may help in making a clin-
Screening Pathology and later adopted and ical decision about a surgical diagnostic exci-
recommended by the EWGBSP, “atypical epi- sion. In the study of Sneige et al. (2003) on
thelial proliferation, ductal type,” is listed as B3 61 cases of vacuum-assisted biopsy atypical
which includes lesions with uncertain malignant ductal proliferations, the lesions were con-
potential. This explains why in the literature fined to an average of 1.5 large ducts or lob-
“upstaging” of excisions after stereotactic breast ular units and were associated with
core biopsies of patients with nonpalpable sus- microcalcifications in all cases. Surgical spec-
picious microcalcifications and/or lesions of imens showed ADH in 15 cases, no residual
AEPDT to DCIS or invasive carcinoma are lesion in 24 cases, and ductal carcinoma in
found in 12–52%; likewise, using vacuum- situ only in 3 cases. It was found that micro-
assisted biopsy devices, which reveal more con- calcifications that contain atypical ductal pro-
tiguous tissue than core biopsy needles, the liferations in less than three lobules or ducts
reported rate of “upstaging” of AEPDT to and/or that are removed completely do not
breast cancers ranges from 0% to 28%. reveal higher-risk lesions on excision; thus,
Recently Weigel et al. (2011) analyzed the it was concluded removal in such cases to be
rate and the histological spectrum for malig- unnecessary. Similar results were published
nancy of minimally invasive biopsies with by Wagoner et al. (2009). In practice, we
“uncertain malignant potential (B3)” in digital therefore modify the NHSBSP and EWGBSP
mammography screening. One hundred forty- guidelines for cases in which sufficient mate-
eight of 979 MIBs (15.1%) were categorized as rial is available and in which the mammogram
B3. Among these B3 lesions, calcifications shows only minor changes that have been
(61.5%) were the most important screening removed by the procedure and finally where
abnormalities. In agreement with several ana- the atypical epithelial proliferation is con-
log studies, the authors found AEPDT to be the fined to single terminal duct-lobular units
most frequent entity (35%), followed by radial (TDLUs) and spares the ducts. Follow-up of
scars (28%) and papillary lesions (20%). these patients with annual control mammog-
Furthermore, with 40.4% AEPDT was the sub- raphy seems a reasonable management
type with the highest rate of finally detected option. The same holds true for ADH in diag-
malignancy (DCIS n = 16, invasive cancer nostic biopsies. In contrast, if ducts or several
n = 3) in surgical excision biopsies (19/47). TDLU areas are involved by atypical prolif-
The lesion-specific positive predictive value erations in core biopsies or in cases in which
(PPV) was highest for AEPDT compared to the mammogram shows more extended
other B3-lesions with a PPV of 0.40, similar changes, which usually indicates grade
to the respective PPVs of 0.32 to 0.59 reported 1 DCIS, we prefer further diagnostic and, if
from analog screening settings (see also Menes necessary, therapeutic procedures.
et al. 2017). • Outcome
The NHSBSP guidelines (2001) suggest Dupont and Page (1989) proposed the term
that if a low- or intermediate-grade atypical ADH for lesions in which “some but not all
epithelial proliferation is found in a vacuum- the features of DCIS” are present and demon-
assisted biopsy that is lacking in extent or strated that the subsequent general risk of inva-
degree of ductal/lobular involvement to be sive breast carcinoma for these lesions is about
classified as DCIS, the lesion should be four to five times that of the general population.
Atypical Ductal Hyperplasia 35
Atypical Ductal Hyperplasia, Fig. 1 (H&E) Atypical in a. (b) ADH with monotonous atypical cells with atypical
ductal hyperplasia. (a) Scanning view showing unfolded architecture characterized by cribriform growth. (c)
TDLU with atypical ductal hyperplasia (center) with two Columnar cell change with psammomatous type micro-
foci of columnar cell change (arrows) and lobular hyper- calcification. (d) Atypical lobular hyperplasia
plasia (asterisks); (b–d) higher magnification of the lesions
36 Atypical Ductal Hyperplasia
Atypical Ductal Hyperplasia, Fig. 3 Cellular algorithm low-grade intraepithelial neoplasias (LG-IEN), including
of ductal-type lesions of the breast. Usual ductal hyperpla- flat epithelial atypia, atypical ductal hyperplasia, low-grade
sia (UDH) contains phenotypically the whole set of lumi- DCIS, and lobular neoplasia. (From Boecker et al. 2017)
nal cells contrasting with the robust CK8/18+ phenotype of
Atypical Ductal Hyperplasia, Table 1 Key features of benign apocrine cells, and moderately stained
atypical ductal hyperplasia regular nuclei with usually small nucleoli and a
ADH Monomorphic epithelial proliferation with higher nucleus-to-cytoplasm ratio. In contrast,
regular placement of cells and atypical high-grade apocrine lesions are defined by pleo-
architecture with smooth geometrical growth morphic, hyperchromatic nuclei, coarse chroma-
patterns such as micropapillae, arcades, Roman
bridges, bars crossing the glandular space, and tin, and prominent nucleoli.
in full-blown lesions with cribriform growth
The cells contain eosinophilic to pale cytoplasm
and round to oval monotonous nuclei Immunophenotype
Confined to individual TDLU(s), lacking the
ductal segmental spread of low-grade DCIS
Several groups demonstrated that the epithelial
Often associated with lamellar
microcalcifications cells in the putative low-grade ductal pathway
precursors, including classical ADH, were posi-
tive for glandular keratins CK7, CK19, and CK8/
criteria of atypical apocrine hyperplasia are met CK18 but negative for basal keratins CK5
whenever a lesion shows features of low-grade (Fig. 5) and CK14 and myoepithelial markers.
apocrine DCIS and is of small size. There is Strong E-cadherin membranous staining has
good reason to believe that the same fundamen- been used to define ductal-type carcinomas,
tal extension criteria used to define classical both in situ and invasive. Overexpression of
atypical ductal proliferations (distinguishing HER2 is rare with only weak basal expression
such lesions from low-grade DCIS) also in ADH, in contrast to high expression and
apply to lesions with low-grade atypical apo- amplification rates in high-grade DCIS (▶ Duc-
crine proliferations. Among the cytological hall- tal Carcinoma In Situ). The cellular constituents
marks of low-grade apocrine lesions are a of ADH demonstrate increased expression rates
smaller cell size, cytoplasmic eosinophilia or with Ki-67 (usually <5%), ERa, the anti-
pallor with less coarse granularity than in classic apoptotic proto-oncogene bcl-2, and the cell
38 Atypical Ductal Hyperplasia
Atypical Ductal Hyperplasia, Fig. 4 (H&E) Growth crossing the lumen in the background of FEA. The nuclei
patterns of ADH. High-power views. (a) Ductule are round and hyperchromatic. (d) Ductule showing pro-
highlighting the proliferation of the same cell type both liferation of cells forming irregular bars and an abortive
in the periphery and within a single micropapilla. The cribriform pattern and micropapillae with psammomatous
tumor cells in the micropapilla, however, tend to be smaller microcalcifications. (e) In this ductule, many rigid bars
with more darkly staining nuclei with slight variation in cross the lumen forming a cribriform growth pattern.
size and spacing. (b) Ductule with multiple micropapillae Same case as c. (f–g) Proliferation of a single cell type
in a background of FEA. Same case as a. (c) Ductule with forming many arcades resembling the pillars supporting
proliferation of a single cell type forming a rigid bar Roman bridges
cycle regulator cyclin D1, compared with adja- Molecular Features and Tumorigenesis
cent normal breast lobular units. The
myoepithelial cells stain strongly for p63, CK5/ The traditional tumorigenesis model of linear
14, and myoepithelial markers such as smooth- progression implies a cascade from benign duc-
muscle actin, calponin etc. tal proliferations to invasive breast carcinoma.
Atypical Ductal Hyperplasia 39
Atypical Ductal Hyperplasia, Table 2 Comparison of histological features of UDH, ADH, and low-grade DCISa
Histological Low-nuclear-grade
features UDH ADH DCIS
Size Variable size but rarely extensive Small More extensive,
ductal-lobular
involvement
Cellular Mixed epithelial cells with Monotonous cell population with Identical to ADH
composition variations in size and shape regular spacing
Myoepithelial cells usually around Myoepithelial cells around the ductal
the ductal periphery periphery
Architecture Fenestrating growth pattern with Atypical with well-delineated Identical to ADH
irregular lumina: Streaming pattern geometric structures
common with long axes of nuclei (micropapillae, bars, Roman bridges,
arranged parallel to the direction of arcades, and cribriform pattern) or
cellular bridges, which often have a solid growth
“tapering” appearance
Lumina Irregular lumina, often ill-defined Usually well-formed rounded Identical to ADH
peripheral slit-like spaces are spaces
common and useful distinguishing
features
IH Mosaicism of CK5- (CK14-) CK8-/CK18-positive cells, lacking Identical to ADH
positive cells. Myoepithelial cells CK5 and CK14. Residual normal
around the periphery CK5- and/or CK14-positive cells
usually attenuated or in a luminal
position
Focal ER positivity Uniformly and strongly ER+ cells
Nuclear Uneven Even, occasionally uneven Identical to ADH
spacing
Nucleoli Indistinct Single, small Identical to ADH
Mitoses Infrequent with no abnormal forms Infrequent, abnormal forms rare Identical to ADH
Necrosis Rare Rare If present, confined
to small particulate
debris in luminal
spaces
Major diagnostic features are shown in bold type
a
Modified version of the table in “European Guidelines for Quality Assurance in Mammography Screening – Fourth
Edition,” Office for Official Publications of the European Communities
Atypical Ductal Hyperplasia, Fig. 6 Atypical ductal CK5/CK14-positive myoepithelial cells in the periphery in
hyperplasia versus usual ductal hyperplasia. H&E stain of (b). Usual ductal hyperplasia showing the fenestrating
ADH showing monomorphous cell proliferation with crib- growth pattern in H&E routine section in (c) and strong
riform growth pattern in (a) and CK5/CK14 immunostain immunostaining for CK5/CK14 in proliferating cells with
with CK5/CK14-negative neoplastic cells contrasting with the typical CK5/CK14þ mosaicism (d)
in clinical practice, this could lead to a decrease challenges. In the authors’ experience, the typi-
in the number of surgeries carried out for cal CK5/CK14 staining pattern usually helps to
intraductal proliferative lesions. One important solve the problem as intermediate malignant
problem remains: as intermediate-grade neoplas- ductal-type proliferations usually fail to stain
tic intraepithelial proliferations, ductal type, may for keratins CK5 and CK14. Finally, micro-
exhibit more variability in cytological and archi- papillary ductal growth patterns of epithelial
tectural characteristic than classical prolifera- proliferations may occasionally also pose prob-
tions of the low-grade ductal pathway, the lems in the differential diagnosis in which, again,
differentiation of these lesions from conven- CK5/CK14 immunohistochemistry may help to
tional ductal hyperplasia is one of the greatest solve the problem.
42 Atypical Vascular Lesions
Occasionally cells with more advanced cellular Moinfar, F., Man, Y. G., Bratthauer, G. L., Ratschek, M., &
atypia may grow in just one or two layers. Tavassoli, F. A. (2000). Genetic abnormalities in mam-
mary ductal intraepithelial neoplasia-flat type (“cling-
Currently, most experts in the field agree that flat ing ductal carcinoma in situ”): A simulator of normal
epithelial lesions with high-nuclear-grade mor- mammary epithelium. Cancer, 88, 2072–2081.
phology should be termed high-grade DCIS O’Malley, F. P., & Bane, A. L. (2004). The spectrum of
irrespective of their size. apocrine lesions of the breast. Advances in Anatomic
Pathology, 11, 1–9.
The most important distinguishing feature of Page, D. L., & Rogers, L. W. (1992). Combined histologic
ADH to lobular neoplasia (▶ Lobular In Situ Neo- and cytologic criteria for the diagnosis of mammary
plasia) is its cohesive growth; the latter, by its atypical ductal hyperplasia. Human Pathology, 23,
discohesive growth and typical change of cell 1095–1097.
Raju, U., Zarbo, R. J., Kubus, J., & Schultz, D. S. (1993).
shape from polygonal to round tumor cells, is The histologic Spectrum of apocrine breast prolifera-
usually easily recognizable on H&E sections. tions: A comparative study of morphology and DNA
Molecularly the presence or loss of the adhesion content by image analysis. Human Pathology, 24,
molecule E-cadherin may be helpful in 1973–1981.
Sneige, N., Lim, S. C., Whitman, G., Krishnamurthy, S.,
distinguishing between growth patterns of ductal Sahin, A. A., Smith, T. L., & Stelling, C. B. (2003).
and lobular type. Atypical ductal hyperplasia diagnosis by directional
vacuum-assisted stereotactic biopsy of breast micro-
calcifications. Considerations for surgical excision.
American Journal of Clinical Pathology, 119, 218–243.
References and Further Reading Wagoner, M. J., Laronga, C., & Acs, G. (2009). Extent and
histologic pattern of atypical ductal hyperplasia present
Aulmann, S., Elsawaf, Z., Penzel, R., Schirmacher, P., & on core needle biopsy specimens of the breast can
Sinn, H. P. (2009). Invasive tubular carcinoma of the predict ductal carcinoma in situ in subsequent excision.
breast frequently is clonally related to flat epithelial American Journal of Clinical Pathology, 131, 112–121.
atypia and low-grade ductal carcinoma in situ. Weigel, S., Decker, T., Korsching, E., Biesheuvel, C.,
The American Journal of Surgical Pathology, 33, Wöstmann, A., Böcker, W., Hungermann, D.,
1646–1653. Roterberg, K., Tio, J., & Heindel, W. (2011). Minimal
Azzopardi, J. (1979). Problems in breast pathology. invasive biopsy results of “uncertain malignant poten-
London: W.B. Saunders. tial” in digital mammography screening: High preva-
Boecker, W., Stenman, G., Schroeder, T., Schumacher, U., lence but also high predictive value for malignancy.
Loening, T., Stahnke, L., Löhnert, C., Siering, R. M., Fortschritte auf dem Gebiet der Röntgenstrahlen und
Kuper, A., Samoilova, V., Tiemann, M., Korsching, E., der bildgebenden Verfahren, 183, 743–748.
& Buchwalow, I. (2017). Multicolor immunofluores- Wells, C. A., Amendoeira, I., Apostolikas, N., et al. (2006).
cence reveals that p63- and/or K5-positive progenitor Quality assurance guidelines for pathology.
cells contribute to normal breast epithelium and usual In N. M. Perry, M. Broeders, & C. de Wolf (Eds.),
ductal hyperplasia but not to low grade intraepithelial European guidelines for quality assurance in breast
neoplasia of the breast: New concepts on the cellular cancer screening and diagnosis (p. 219). Luxembourg:
hierarchy using in-situ multicolour experiments. Office for Official Publication of the European
Virchows Archiv, 470, 493–504. Communities.
Dupont, W. D., & Page, D. L. (1989). Relative risk of
breast cancer varies with time since diagnosis of atyp-
ical hyperplasia. Human Pathology, 20, 723–725.
Jain, R. K., Mehta, R., Dimitrov, R., Larsson, L. G.,
Musto, P. M., Hodges, K. B., Ulbright, T. M.,
Atypical Vascular Lesions
Hattab, E. M., Agaram, N., Idrees, M. T., & Badve, S.
(2011). Atypical ductal hyperplasia: Interobserver and Lauren E. Rosen and Thomas Krausz
intraobserver variability. Modern Pathology, 24(7), Department of Pathology, University of Chicago,
917–923.
Chicago, IL, USA
Lakhani, S. R., Ellis, I. O., Schnitt, S. J., Tan, P. H., &
van de Vijver, M. J. (2012). WHO-classification of
tumours of the breast. Lyon: IARC.
Menes, T. S., Kerlikowske, K., Lange, J., Jaffer, S., Synonyms
Rosenberg, R., & Miglioretti, D. L. (2017). Subsequent
breast cancer risk following diagnosis of atypical
ductal hyperplasia on needle biopsy. JAMA Oncology, Cutaneous atypical vascular lesions (AVLs) of
3, 36–41. the breast have been described using a variety
Atypical Vascular Lesions 43
• Incidence
Macroscopy
AVLs occur in the field of radiation after radi-
ation therapy for breast cancer, with a latency
AVLs typically present as discrete flesh-toned to
period of 1 month to 20 years, occurring most
reddish or bluish papules arising in the field of
frequently between 3 and 6 years post-
radiation, and less frequently as vesicles, cystic
treatment (Ginter et al. 2017).
lesions, or erythematous plaques (Brenn and
• Age
Fletcher 2005; Koerner 2014). The lesions are
AVLs are seen in a population ranging in age
generally less than 5 mm in size (range
from 29 to 95 years, most occurring in the fifth
1–20 mm), and can be single or multiple, with
and sixth decades (Ginter et al. 2017).
up to half of patients developing multiple lesions
• Sex
either at the time of initial diagnosis or subse-
All reported cases of AVLs of the breast have
quently (Baker and Schnitt 2017; Ginter et al.
occurred in females, likely reflecting the higher
2017; Flucke et al. 2013).
incidence of breast cancer in this population
(Weaver and Billings 2009).
• Site
AVLs occur most commonly in mammary or Microscopy
axillary skin, and can rarely involve the breast
parenchyma. AVLs also occur in sites other On low power, AVLs are frequently circum-
than the breast following radiation therapy for scribed lesions located within the superficial der-
various malignancies, such as gynecologic mis. AVLs can be wedge-shaped with their base
tumors and multiple myeloma (Brenn and oriented toward the dermal-epidermal junction
Fletcher 2005). (Baker and Schnitt 2017). The lesions are
44 Atypical Vascular Lesions
generally flat, but may appear as exophytic pap- and rare mitotic figures; however, endothelial
ules that project from the skin surface. The over- multilayering and necrosis are absent. The adja-
lying epidermis is usually normal or mildly cent stroma may contain sparse to dense chronic
acanthotic (Koerner 2014). When AVLs involve inflammatory infiltrate composed of lymphocytic
the breast parenchyma, they are usually encased aggregates with occasional germinal centers, or
within fibrous tissue without involvement of the there may be adjacent hemorrhage or hemosiderin
mammary glandular tissue (Brenn and Fletcher (Ginter et al. 2017; Patton et al. 2008).
2005). AVLs are composed of ectatic, angulated,
and variably anastomosing thin-walled vascular
channels lined by a single layer of monomorphous Immunophenotype
endothelial cells (Baker and Schnitt 2017; Weaver
and Billings 2009) (Fig. 1a: H&E). While in Lymphatic type AVLs are positive for CD31,
most cases the lesions are limited to the superficial D2-40, ERG, and positive or negative for CD34
dermis, in some cases the vessels extend into (Fig. 1b: CD34 immunostaining; 1C: D2-40
the deep dermis where they are often smaller, immunostaining; 1D: CD31 immunostaining). Vas-
compressed, and infiltrative with focal dissection cular type AVLs are positive for CD31, CD34, and
of collagen and infiltration of adnexal structures ERG, but negative for D2-40 (Baker and Schnitt
(Brenn and Fletcher 2005). More rarely, the 2017). The pericytes surrounding the vessels in
lesions can extend into the subcutis. Two distinct vascular type AVLs are positive for smooth muscle
morphologic subtypes of AVL have been actin (Brenn and Fletcher 2005).
described, lymphatic type and vascular type.
Both patterns are often seen within the same
lesion (Patton et al. 2008; Ginter et al. 2017). Molecular Features
The lymphatic type is the most common sub-
type of AVL, and is comprised of variably-sized Amplification of MYC has been implicated in
anastomosing vascular channels lined by a mono- the development of PIAS (Fernandez et al.
layer of attenuated to hobnail endothelial cells 2012; Fraga-Guedes et al. 2015; Mentzel et al.
reminiscent of lymphangioma (Gengler et al. 2012). The MYC oncogene is a transcription fac-
2007; Koerner 2014). Endothelial-lined stromal tor that has a role in cell proliferation, cellular
projections can be seen within the vascular differentiation, apoptosis, and angiogenesis.
lumens, which are characteristically devoid of Dysregulated MYC expression due to gene ampli-
erythrocytes (Flucke et al. 2013; Patton et al. fication promotes cell proliferation through inap-
2008). The vessels are typically tightly clustered propriate progression to S-phase from G1 phase
within the superficial dermis; however, in a minor- of the cell cycle. Amplification of MYC occurs
ity of cases, they extend into the deep dermis in other sarcomas, such as high-grade chon-
where they have a more infiltrative appearance. drosarcomas, proximal type epithelioid sarcomas,
The lining endothelium is commonly hyper- and myxoid liposarcomas (Fraga-Guedes et al.
chromatic, but lacks significant nuclear atypia, 2015). Many studies have found high-level ampli-
prominent nucleoli, and mitoses (Patton et al. fications of MYC by fluorescence in situ hybridi-
2008; Requena et al. 2002). zation (FISH) in the majority of post-radiation and
Vascular type AVLs are composed of round to lymphedema associated angiosarcomas, but not
elongated capillary sized vessels that grow irreg- in primary cutaneous angiosarcomas or AVLs
ularly within the superficial or deep dermis, rem- (Fernandez et al. 2012; Fraga-Guedes et al.
iniscent of a capillary hemangioma (Patton et al. 2015; Mentzel et al. 2012). Interestingly in one
2008). In contrast to lymphatic type AVLs, these study, two patients had AVLs lacking MYC ampli-
lesions show intraluminal red cells and the vessels fication adjacent to MYC amplified PIAS (Fraga-
are surrounded by a layer of pericytes. Vascular Guedes et al. 2015). Amplification of MYC by
type AVLs can show mild random nuclear atypia FISH has a strong correlation with MYC
Atypical Vascular Lesions 45
Atypical Vascular Lesions, Fig. 1 (a) AVL composed of cytologic atypia [H&E, 40X], (b) AVL with variable pos-
irregular thin-walled dilated vascular spaces dissecting itivity for CD34 [20x], (c and d) AVL with strong diffuse
through collagen. The nuclei are widely spaced and lack positivity for D2-40 (c) and CD31 (d) [20x]
expression by immunohistochemistry (IHC). (Weaver and Billings 2009). The majority of PIAS
FLT4 is a gene that encodes for VEGFR3 and are high grade, but low and intermediate grade
plays a role in lymphatic differentiation. FLT4 tumors have been described in which there is
gene amplification is detected by FISH in 25% morphologic overlap with AVLs. AVLs may be
of PIAS, and is absent in AVLs, but the gene is multiple but are usually small pink papules (<
co-amplified with MYC limiting its utility in this 1 cm) while PIAS are often larger (>2 cm) and
setting (Cornejo et al. 2015; Guo et al. 2011). One present as violaceous or erythematous plaques
study demonstrated TP53 mutations in the major- with ulceration (Weaver and Billings 2009;
ity of AVLs studied, and hypothesized that TP53 Brenn and Fletcher 2005). AVLs are relatively
mutations may occur early in neoplastic develop- circumscribed and limited to the superficial der-
ment of AVL (Santi et al. 2011). mis, however can invade into the deep reticular
dermis and rarely into the subcutis; while PIAS
often invade through the dermis and into the sub-
Differential Diagnosis cutis (Weaver and Billings 2009; Brenn and
Fletcher 2005). The nuclei of AVLs are hyper-
The main differential diagnosis for AVL is chromatic and can show mild random atypia
angiosarcoma (▶ Angiosarcoma of the Breast), (Fig. 2a: H&E), while the nuclei in PIAS are
particularly those that arise following radiation, uniformly enlarged with prominent nucleoli
termed PIAS. Both AVL and PIAS arise in the (Weaver and Billings 2009; Brenn and Fletcher
skin of the breast or chest wall following radia- 2005). Features that favor a diagnosis of AVL
tion, occur in middle aged to elderly women, and include circumscription, vessels lined by a single
are characterized by anastomosing vascular chan- layer of endothelial cells, bloodless spaces, deli-
nels. PIAS have a latency period of 5–6 years, cate projections of endothelial lined stroma into
which overlaps with the 3–6 year interval of AVLs the vascular spaces, and associated chronic
46 Atypical Vascular Lesions
Atypical Vascular Lesions, Fig. 2 (a) AVL with irregu- (dissecting collagen), irregular vascular channels lined by
larly shaped, dilated vascular channels and random cyto- focally multilayered neoplastic endothelium with marked
logic atypia with variably enlarged and hyperchromatic diffuse nuclear atypia and extravasated red blood cells
nuclei [H&E, 40x]. (b and c) AVL with low Ki-67 prolif- [H&E, 40x]. (e and f) Post-radiation angiosarcoma
eration index (b) and negativity for c-myc (c) [40x]. (d) with high Ki-67 proliferation index (e) and c-myc positiv-
Post-radiation angiosarcoma is composed of invasive ity (f) [40x]
inflammation. Proliferation activity evaluated by (Fig. 2e and f: Ki67 immunostaining and cMYC
Ki67 is low and cMYC is negative (Udager et al. immunostaining, respectively). AVLs can very
2016; Requena et al. 2002) (Fig. 2b and c: Ki67 rarely show more worrisome features such as
immunostaining and cMYC immunostaining, scattered variably enlarged nuclei with prominent
respectively); (Table 1). Findings that favor nucleoli, an occasional mitosis, focal, limited
PIAS include infiltration of the subcutis, promi- multilayering (usually not more than a double
nent dissection of dermal collagen, hemorrhage layer of endothelium), poor circumscription, and
and extravasated red blood cells, marked cyto- extension into subcutis (Weaver and Billings
logic atypia, mitotic figures, necrosis, and multi- 2009). In addition, AVLs and PIAS can coexist,
layering of the endothelial cells (Fig. 2d: H&E). and PIAS may exhibit AVL-like areas, which can
Proliferation activity evaluated by Ki67 is be indistinguishable from a true AVL in a limited
high and cMYC is overexpressed in nuclei biopsy. For this reason, it is recommended that a
(Udager et al. 2016; Requena et al. 2002) diagnosis of AVL should not be made on a small
Atypical Vascular Lesions 47
pathology (pp. 1143–1148). Philadelphia: Lippincott Requena, L., Kutzner, H., Mentzel, T., et al. (2002). Benign
Williams & Wilkins. vascular proliferations in irradiated skin. American
Mentzel, T., Schildhaus, H. U., Palmedo, G., et al. (2012). Journal of Surgical Pathology, 26, 328–337.
Postradiation cutaneous angiosarcoma after treatment Santi, R., Cetica, V., Franchi, A., et al. (2011). Tumour A
of breast carcinoma is characterized by MYC amplifi- suppressor gene TP53 mutations in atypical vascular
cation in contrast to atypical vascular lesions after lesions of breast following radiotherapy. Histopathol-
radiotherapy and control cases: Clinicopathological, ogy, 58, 455–466.
immunohistochemical and molecular analysis of Udager, A. M., Ishikawa, M. K., Lucas, D. R., et al. (2016).
66 cases. Modern Pathology, 25, 75–85. MYC immunohistochemistry in angiosarcoma and
Patton, K. T., Deyrup, A. T., & Weiss, S. W. (2008). atypical vascular lesions: Practical considerations
Atypical vascular lesions after surgery and radiation based on a single institutional experience. Pathology,
of the breast. A clinicopathologic study of 32 cases 47, 697–704.
analyzing histologic heterogeneity and association Weaver, J., & Billings, S. D. (2009). Postradiation cutane-
with angiosarcoma. American Journal of Surgical ous vascular tumors of the breast: A review. Seminars
Pathology, 32, 943–950. in Diagnostic Pathology, 26, 141–149.
B
Definition
Clinical Features
Apocrine metaplasia: This is a change of native
breast ductal epithelial cells into cells showing • Incidence
eosinophilic granular cytoplasm, a large open Microscopic apocrine change is frequent in the
nucleus with a single nucleolus, and decapitation female breast after the age of 30, is uncommonly
secretion. Apocrine metaplasia can be subtyped observed in women younger than 19, and
into simple, papillary, and complex depending on increases with age, persisting postmenopausally.
architectural complexity. Apocrine metaplasia can Atypical apocrine lesions are uncommon, and
coexist with columnar cell change (▶ Columnar the precise incidence is unknown.
Cell Lesions), sometimes even involving the same • Age
duct spaces. 19–90
Apocrine adenosis: This is apocrine change • Sex
in sclerosing adenosis (▶ Sclerosing Adenosis). Female
Some authors use a different definition, as • Site
a haphazard proliferation of bland glands Breast parenchyma, no specific localizations
with apocrine differentiation and use this • Treatment
term as synonymous with adenomyoepithelial Benign apocrine changes and apocrine cell meta-
adenosis. plasia: Apocrine metaplasia is quite common
© Springer Nature Switzerland AG 2020
A. Sapino, J. Kulka (eds.), Breast Pathology, Encyclopedia of Pathology,
https://doi.org/10.1007/978-3-319-62539-3
52 Benign and Atypical Apocrine Lesions
within the breast, is not premalignant and hence but when found in the ducts and lobules within the
does not require surgical intervention. breast, these appear to be a consequence of meta-
Atypical apocrine change within sclerosing plasia, possibly related to columnar cell change
adenosis (atypical apocrine adenosis): Insufficient (▶ Columnar Cell Lesions) and hormonal stress.
cases have been studied to determine if surgical Apocrine cells have eosinophilic granular cyto-
excision is warranted or not (in view of the clin- plasm due to secretory vacuoles, a large open
ical concern of concurrent apocrine DCIS or nucleus with a single nucleolus, and decapitation
invasive apocrine carcinoma (▶ Apocrine Carci- secretion. Apocrine cells carry androgen receptor,
noma). Atypical apocrine change in a papilloma: as do some examples of columnar cell change and
Complete surgical excision recommended. therefore this may be a response to androgenic
Atypical apocrine hyperplasia: Complete stimulation. Although some authors believe that
surgical excision recommended. apocrine cells are a normal constituent of the
• Outcome breast, they can sometimes be seen as partial
Benign apocrine lesions pose no long-term risk metaplasia within a duct, the rest of the lining of
to the patient. which is columnar cell change (Fig. 1 (H&E)).
Regarding atypical apocrine lesions, Apocrine cells often form the lining of type
Fuehrer et al. (2012) showed that the rate of 1 cysts. Type 1 cysts have a high concentration of
breast cancer diagnosis in follow-up of patients potassium relative to sodium and chloride
diagnosed with atypical apocrine change
within sclerosing adenosis (8.1%) was not
appreciably different from that of the cohort
overall (7.8%). Calhoun and Booth (2014)
showed that out of seven patients diagnosed
with atypical apocrine change within scleros-
ing adenosis, none of these diagnoses were
upgraded to ductal carcinoma in situ (▶ Ductal
Carcinoma In Situ) or invasive carcinoma fol-
lowing surgical excision; hence it remains
unclear whether a diagnosis of atypical apo-
crine change within sclerosing adenosis in iso-
lation requires surgical excision or not.
Macroscopy
Microscopy
Benign and Atypical Apocrine Lesions,
Fig. 1 Columnar cell change and apocrine metaplasia
Apocrine cells are a normal constituent of the occurring within same duct spaces, with associated micro-
sweat glands around the nipple and in the axilla, calcifications (H&E)
Benign and Atypical Apocrine Lesions 53
Differential Diagnosis
A clinicopathologic study of 37 patients with 8.7-year Sometimes, the cause of fat necrosis is unknown
follow-up. Cancer, 77, 2529–2537. (Tan et al. 2006; Kaplan et al. 2005).
Selim, A. G. A., Ryan, A., El-Ayat, G., & Wells, C. A.
(2002). Loss of heterozygosity and allelic imbalance in Fat necrosis of the breast may be a challenging
apocrine metaplasia of the breast: Microdissection diagnosis on mammography, ultrasound, CT,
microsatellite analysis. The Journal of Pathology, PET-CT, and MRI. The extent of associated fibro-
196, 287–291. sis, liquefied fat, and calcifications determine the
imaging findings of fat necrosis. The diagnosis of
fat necrosis using fine needle aspiration cytology
(FNAC) is limited by inadequate samples. Core
Breast Fat Necrosis biopsy is more sensitive.
Clinical presentation of fat necrosis is variable
Inta Liepniece-Karele and Sergejs Isajevs (Chala et al. 2004) ranging from an incidental
Riga East University Hospital, benign finding (indolent single or multiple round
Centre of Pathology/Faculty of Medicine, oil cysts or nodules on imaging) to a lump highly
University of Latvia, Riga, Latvia suggestive of cancer (palpable and radiologically
visible irregular masses with overlying skin
retraction). It can be associated with pain, skin or
Synonyms nipple retraction, erythema and lymphadenopa-
thy. The lesion characteristically is situated near
Adipose tissue necrosis the skin or areola, since these are the sites within
the breast that are most vulnerable to trauma. The
average period for patients to present with a breast
Definition lump from time of trauma is 68.5 weeks (Tan et al.
2006; Kaplan et al. 2005).
Breast fat necrosis is a benign nonsuppurative
inflammatory process of adipose tissue usually • Incidence
complicating breast trauma, breast surgery, or The incidence of the disease is estimated to be
radiation treatment. It was initially described in up to 0.6% in the breast, representing 2.75% of
1920 by Lee, BE & Munzer, JT. all benign lesions. Fat necrosis is found in
0.8% in surgical specimens of breast tumors
and in 1% of breast reduction surgery cases
Clinical Features (Tan et al. 2006; Kaplan et al. 2005).
• Age
The etiological factors include trauma or micro- Variable, the average age of clinical presenta-
trauma, radiotherapy, biopsy, anticoagulation tion is 50 years.
(warfarin), surgery, duct ectasia (▶ Duct Ectasia • Sex
and Periductal Mastitis), and breast infections Mainly described in female breast.
(Tan et al. 2006; Kaplan et al. 2005). The breast • Site
can be injured in a motor vehicle accident, which Variable.
occurs in 16% of cases of seat belt-restrained • Treatment
victims. Fat necrosis was observed secondary In patients with a new palpable finding, the
to the direct injection of silicone or paraffin into history of a traumatic event can be helpful in
the breast. Transplantation of a large amount of making the diagnosis of fat necrosis. Manage-
autologous fat can result in fat necrosis, because ment includes short-term follow-up with imag-
fat cells are not nourished adequately until neo- ing and physical examination, including
vascularization occurs. Other rare causes mammographic follow-up. Fat necrosis often
include polyarteriitis nodosa, Weber-Christian resolves on its own. If the lump does not dis-
disease, and granulomatous angiopanniculitis. appear over time, or increase in size, operation
Breast Fat Necrosis 61
Macroscopy
Microscopy
Immunophenotype
In rare cases, immunohistochemical stains for Breast Fat Necrosis, Fig. 3 Magnification 100, H&E
cytokeratin may be necessary to exclude an staining
62 Breast Fat Necrosis
Most cases present as a periprosthetic effusion presentation was found to correlate with an infe-
(seroma or “in situ” lymphoma) as illustrated in rior overall survival (Ferrufino-Schmidt et al.
Fig. 1. Figure 1a shows a lymphoid proliferation 2018). Rare cases present with disseminated
embedded in fibrinous material at the surface of disease.
the periprosthetic capsule; panel 1b shows an
area of mild infiltration into the capsule
(arrow). Only a minority of cases present as a Microscopy
tumor mass infiltrating into the adjacent breast
parenchyma (Fig. 3a), with or without an In cases with seroma-associated presentation,
associated effusion (Clemens et al. 2016). tumor cells may be identified in cytological
A significant proportion of the patients samples from pericapsular effusions or on
(around 30%) may present with axillary lymph- capsulectomy specimens. The tumor cells are
adenopathy, which is not proven involved large and pleomorphic, with large nuclei and
by lymphoma in all instances. Lympho- abundant cytoplasm (Fig. 2). Cells with
matous involvement of axillary lymph nodes horseshoe-shaped nuclei and a paranuclear
is usually characterized by a low-tumor cytoplasmic inclusion (“hallmark cells”)
burden and can feature a sinusoidal, perifollicular, typically encountered in all forms of ALCL are
diffuse, or Hodgkin-like pattern, often associated also found in BI-ALCL (Fig. 2, arrows). The
to fibrosis. Lymph node involvement at initial neoplastic cells may resemble Hodgkin-like
Breast Implant-Associated Malignant Lymphoma 65
Breast Implant-Associated Malignant Lymphoma, a very high proportion of the nuclei indicating a high
Fig. 4 Immunohistochemical findings in BI-ALCL: the proliferation fraction (E) and perforin, another marker of
tumor cells are strongly positive for CD30 with a mem- activated cytotoxic phenotype, produces fine granular pos-
brane and paranuclear dot-like pattern (A), negative for itivity in a subset of the tumor cells (F). Images A-D are
ALK (B), positive for EMA (C), and positive for granzyme from the tumoral BI-ALCL and images E-F from the
B indicating an activated cytotoxic profile (D); Ki67 stains seroma-associated case
Three IL2-dependent TLBR (T-cell breast origin of the neoplasm may be questioned. Stag-
lymphoma) cell lines established from seroma- ing and clinical history are therefore critical to
associated BI-ALCL had clonally abnormal establish the correct diagnosis.
complex karyotypes with a modal number – Other types of lymphomas have been reported to
of 47 chromosomes in one cell line (TLBR-1) occur in association with breast implants: mycosis B
and a hypertriploid pattern in TLBR-2 and fungoides/Sézary syndrome, extranodal NK/T-
TLBR-3. Functional studies on these three cell cell lymphoma, nasal type, and different types
lines showed evidence of STAT3 activation, of B-cell lymphomas (diffuse large B-cell lym-
while pharmacological inhibition of STAT3 inhi- phoma, follicular lymphoma, lymphoplasmacytic
bition induced in vitro cell death. lymphoma, primary effusion lymphoma, mar-
Seven cases of BI-ALCL were successfully ginal zone B-cell lymphoma). These lymphomas
analyzed by whole exome or targeted next- differ from BI-ALCL both by their morphology
generation sequencing using a large panel of and immunophenotype.
465 cancer-associated genes, and of these only – Hodgkin lymphoma is an entity with
four showed somatic variants (reviewed in significant overlapping morphological and
Letourneau et al. 2018). The STAT3 S614R variant immunophenotypical features with BI-ALCL.
was detected by whole exome sequencing or The Hodgkin/Reed-Sternberg cells of Hodgkin
targeted sequencing in two cases (including the lymphoma are typically scarce in a prominent
primary tumor from which TLBR-1 cell line was inflammatory background, coexpress CD30
derived), as the sole abnormality in the latter and in and CD15, and have a B-cell genotype with
combination with pathogenic mutations in TP53 expression of a markedly attenuated B-cell
and SOCS1 in the other case. One somatic variant immunophenotype. They are usually negative
was detected in the other two cases, affecting JAK1 for T-cell antigens. Although Hodgkin lym-
(G1097 V) or DNMT3A (W176X). In one case of phoma very rarely occurs in extranodal locali-
BI-ALCL which presented as a solid tumor mass zations, an exceptional case of Hodgkin
and recurred as an in situ capsular lesion, dual gain- lymphoma arising adjacent to a breast implant
of-function mutations in JAK1 and STAT3 were has been reported in a woman who had a his-
identified in both specimens, suggesting pathoge- tory of follicular lymphoma, and the Hodgkin
netic mechanisms overlapping with those of sys- lymphoma was shown to represent transforma-
temic ALK-negative ALCLs (Letourneau et al. tion from the preexisting follicular lymphoma.
2018) However, rearrangements of the IRF4/ – Chronic inflammatory lesions associated to a
DUSP22 locus at 6p25 and of TP63 (frequently breast implant may contain occasional CD30+
observed in systemic or primary cutaneous ALCL) activated lymphoid cells and must be distin-
have not been found in any of the cases tested so far guished from BI-ALCL.
(Oishi et al. 2018).
Ferrufino-Schmidt, M. C., Medeiros, L. J., Liu, H., Clemens, Letourneau, A., Maerevoet, M., Milowich, D., Dewind, R.,
M. W., Hunt, K. K., Laurent, C., Lofts, J., Amin, M. B., Bisig, B., Missiaglia, E., & de Leval, L. (2018, 2018).
Ming Chai, S., Morine, A., Di Napoli, A., Dogan, A., Dual JAK1 and STAT3 mutations in a breast implant-
Parkash, V., Bhagat, G., Tritz, D., Quesada, A. E., Pina- associated anaplastic large cell lymphoma. Virchow
Oviedo, S., Hu, Q., Garcia-Gomez, F. J., Jose Borrero, J., Archiv. https://doi.org/10.1007/s00428-018-2352-y.
Horna, P., Thakral, B., Narbaitz, M., Hughes, R. C., 3rd, Oishi, N., Brody, G. S., Ketterling, R. P., Viswanatha, D. S.,
Yang, L. J., Fromm, J. R., Wu, D., Zhang, D., Sohani, He, R., Dasari, S., Mai, M., Benson, H. K., Sattler, C. A.,
A. R., Hunt, J., Vadlamani, I. U., Morgan, E. A., Ferry, Boddicker, R. L., McPhail, E. D., Bennani, N. N.,
J. A., Szigeti, R., C Tardio, J., Granados, R., Dertinger, S., Harless, C. A., Singh, K., Clemens, M. W., Medeiros,
Offner, F. A., Pircher, A., Hosry, J., Young, K. H., & L. J., Miranda, R. N., & Feldman, A. L. (2018). Genetic
Miranda, R. N. (2018). Clinicopathologic features and subtyping of breast implant-associated anaplastic large
prognostic impact of lymph node involvement in patients cell lymphoma. Blood, 132(5), 544–547.
with breast implant-associated anaplastic large cell lym- Rupani, A., et al. (2015). Lymphomas associated with
phoma. The American Journal of Surgical Pathology, breast implants: A review of the literature. Aesthetic
42(3), 203–305. Surgery Journal, 35(5), 533–544.
C
Collagenous Spherulosis, Fig. 3 Immunostainings with p63 (a) and CK 14 (b) highlight myoepithelial cells, which
form incomplete lining of the spherules
a surgical excision specimen is not an indica- and Collins 2009). An association with tubular
tion for further surgery. carcinoma (▶ Tubular Carcinoma), which
If FEA is found to coexist with atypical together with columnar cell lesions and lobular
ductal hyperplasia, in situ or invasive carci- neoplasia forms the eponymously named
noma, then naturally the treatment recommen- Rosen’s triad, was first described in 1999
dation is dictated by the higher grade lesion. (Rosen 1999). Whether or not columnar cell
Other treatment recommendations for lesions constitute a direct precursor lesion or C
FEA found on core needle biopsy include radi- are an indicator of a more general increased
ology and pathology correlation with multi- risk of subsequent breast cancer has not been
disciplinary team discussion. Some may established. The time course of possible pro-
recommend using vacuum-assisted biopsy to gression to in situ or invasive carcinoma and
excise any residual tissue showing micro- actual upgrade rates are also contentious areas.
calcification or other areas of radiological con- There is probably sufficient evidence to sup-
cern, instead of advocating surgical excision. port columnar cell lesions being at least one
If no further microcalcification is identified early feature of the low-grade breast neoplasia
radiologically, if the extent of involvement of pathway, but the data are difficult to compare
the terminal duct lobular units by FEA is small and analyze because of the range of methodol-
and the patient does not have a personal history ogies the various studies have used. In some
of breast cancer, ongoing radiological surveil- follow-up studies it is not clear if the subse-
lance instead of surgical excision has been quent carcinoma has been in the same area as
suggested as the preferred management the initial columnar cell lesion. When colum-
(Berry et al. 2016; Acott and Mancino 2016). nar cell lesions are removed at core biopsy it is
However, long-term follow-up studies of generally not known how much, if any, lesional
women with FEA diagnosed on core needle tissue remains. It is difficult to then interpret
biopsy for whom the management has been the significance of subsequent cancers as it
prolonged surveillance are not yet available, would not be known from precisely what tissue
and some use this lack of definitive informa- they may have arisen.
tion to support the use of surgical excision as An association between columnar cell
the primary treatment recommendation. lesions and carcinoma has also been investi-
• Outcome gated by assessing whether columnar cell
The clinical outcomes of the various colum- lesions are present in breast tissue adjacent to
nar cell lesions have not been precisely in situ and invasive carcinomas. Another way
defined, and while some generalizations to explore this association is to retrospectively
can be made, there is still a lack of clarity study biopsies taken previously from women
with respect to the long-term significance of with invasive carcinomas to see if columnar
these lesions. A lower level of risk has been cell lesions, including FEA, might have been
attributed to columnar cell change and present earlier. Follow-up studies have investi-
columnar cell hyperplasia compared with gated whether cancers arising subsequent to
that for FEA. columnar cell lesions have occurred in the
The biological importance of columnar cell same area or somewhere else in the ipsilateral
lesions is underscored by their association with breast or indeed in the contralateral breast, the
other breast lesions such as atypical ductal latter, of course, not being used to support any
hyperplasia (▶ Atypical Ductal Hyperplasia) direct precursor role (Boulos et al. 2008).
and lobular neoplasia, including atypical lobu- Another confounding factor is that many of
lar hyperplasia and lobular carcinoma in situ the studies have not been able to provide infor-
(▶ Lobular In Situ Neoplasia), which have mation as to the size or possible multifocality
been noted to coexist with them, sometimes of the columnar cell lesions although
in the same terminal duct lobular unit (Schnitt pathology-radiology correlation as to the
74 Columnar Cell Lesions
extent of residual calcification has been very had atypical ductal hyperplasia (Verschuur-
helpful in this regard. Maes et al. 2012b).
Overall, the long-term relative risk of carci-
noma for those women with nonatypical
Macroscopy
columnar cell lesions diagnosed in isolation
appears to be very low, similar to that seen for
Columnar cell lesions, including FEA are diag-
nonatypical proliferative breast disease in gen-
nosed microscopically, and except for occasional
eral, around the order of a 1.5-fold increase
floridly cystic examples, are not able to be identi-
(Verschuur-Maes et al. 2012b).
fied macroscopically.
A closely related issue with columnar cell
lesion is that of the potential upgrade to more
significant lesions, particularly in situ and
Microscopy
invasive carcinoma, which occurs when there
is an immediate surgical excision taken on
A distinctive feature of columnar cell lesions on
the basis of a columnar cell lesion identified
low power examination is the cystic dilatation of
on core biopsy. A very wide range of upgrade
enlarged terminal duct lobular units, and for
rates (between 0% and 67%) has been
columnar cell change and columnar cell hyperpla-
described in the literature, and this consider-
sia, these gland spaces often have an irregular
able variation partly reflects the very different
contour (Fig. 1). The lesions may also be conspic-
ways in which the upgrade rate has been cal-
uous on low power examination because of a
culated. The numerator is generally those who
basophilic appearance, partly due to the regular
have had a malignancy in the subsequent exci-
arrangement of the cells, which are generally uni-
sion specimen, while the denominator can
form, columnar to cuboidal in shape, and mostly
include all women who have had a columnar
orientated perpendicular to the basement mem-
cell lesion on core biopsy, or only those who
brane. Quite frequently, small apical cytoplasmic
have gone on to surgical excision. This of
blebs or snouts are seen, projecting into the lumen
course would give a higher upgrade rate. In
of some glandular spaces, but these are not usually
this latter group too, other clinical and radio-
prominent. Thin, pale proteinaceous secretions
logical features may have also contributed to
which may be pink or blue are commonly seen
the decision to recommend surgical excision,
in the gland lumina (Fig. 2). Luminal micro-
thus confounding the analysis of columnar cell
calcification, often the reason for these lesions
lesions. Some studies have reviewed columnar
cell lesions when there is also atypical ductal or
lobular hyperplasia present, while others have
specifically excluded cases with concomitant
atypical lesions. Many more studies have
addressed upgrade rates from FEA than from
pure columnar cell change or columnar cell
hyperplasia. The problem of correctly diagnos-
ing these lesions histologically only adds to
the difficulty of obtaining meaningful outcome
data. Verschuur-Maes in a comprehensive
meta-analysis of 24 papers concerning the
risk of carcinoma in women with columnar
cell lesions derived pooled underestimation
risks of 1.5% for columnar cell lesions without
atypia, 9% for columnar cell lesions with Columnar Cell Lesions, Fig. 1 Columnar cell lesion.
atypia compared with 20% for those who also Cystic dilatation of terminal duct lobular unit. (H & E)
Columnar Cell Lesions 75
Columnar Cell Lesions, Fig. 2 Columnar cell Columnar Cell Lesions, Fig. 4 Columnar cell hyperpla-
lesion. dilated gland lumina containing watery secretions. sia. irregularly dilated gland spaces with some overlapping
(H & E) of cells. (H & E)
Columnar Cell Lesions, Fig. 3 Columnar cell lesion and Columnar Cell Lesions, Fig. 5 Columnar cell hyperpla-
columnar cell hyperplasia. Luminal microcalcification. sia. Luminal tufting of cells and small micropapillae. (H & E)
(H & E)
being detected radiologically, may be seen and micropapillae, and luminal secretions can occur
these range from being fine to granular or coarse (Fig. 5).
and even sometimes psammomatous (Fig. 3). The Enlarged or dilated acini of terminal duct lob-
nuclei of columnar cell change or hyperplasia are ular units, often appearing rounded or distended
generally oval or elongated, with smooth nuclear (Fig. 6) rather than the more convoluted contours
membranes and fine chromatin. Nucleoli usually of columnar cell change, characterize lesions of
are not seen and mitoses are rarely identified. FEA and these are lined by 3–5 layers of cells.
Occasionally the underlying myoepithelial cells With a loss of polarity, the cells have a more
become cuboidal and even relatively prominent. haphazard overlapping arrangement. “Flat” refers
For columnar cell change, 1–2 layers of cells are to the absence of complex architectural features
seen above the basement membrane, with greater such as Roman bridges or well-developed rigid
numbers seen for columnar cell hyperplasia micropapillae. Apical cytoplasmic projections,
(Fig. 4), where stratification of cells can occur characteristic of columnar cell change, may on
within dilated acini, often giving an impression occasion be more pronounced. The lining epithe-
of hyperchromasia. Luminal tufting of cells, blunt lial cells are usually rounded, with a low-grade
76 Columnar Cell Lesions
Immunophenotype
in situ and invasive carcinoma, as part of the low- Twenty-five percent of invasive breast carcino-
grade breast neoplasia pathway. A variety of mas have been demonstrated to contain an acti-
methods of analysis have been used, and while vating mutation in the catalytic subunit of
the findings so far are suggestive of a causal rela- phosphatidylinositol-3-kinase (PIK3CA), and the
tionship, rather than just co-occurrence, the evi- presence of these mutations have been investi-
dence currently available is relatively scanty. gated in columnar cell lesions, with one study
A loss of heterozygosity study of clinging ductal showing them to be present in 13/24 (54%) cases C
carcinoma in situ, also called ductal intraepithelial (Troxell et al. 2012) and in 31/62 (50%) of colum-
neoplasia (DIN) flat type, one of the earlier names nar cell/usual ductal hyperplasia lesions in another
for what is now referred to as FEA, demonstrated (Ang et al. 2014). However, the same point muta-
changes at chromosomes 11q21-23.2, 16q23.1- tions were not consistently identified in the
24.2, and 3p14.2 in 50%, 45%, and 41% of infor- corresponding carcinoma, sometimes with differ-
mative cases, respectively, with corresponding ent mutations identified, or wild-type PIK3CA in
changes identified in the in situ and invasive carci- the carcinoma with mutations in the columnar cell
noma also present in these cases (Moinfar 2009). lesions, or vice versa, wild-type in the columnar
These findings were later supported by the work of cell lesions and point mutations identified in the
Aulman in a study of FEA, tubular carcinoma, and carcinomas making the association less certain.
low-grade ductal carcinoma in situ where loss of Promoter methylation of the tumor suppressor
heterozygosity was noted for 16q in up to 80% of genes ID4, CCND2, and CDH13 has been dem-
cases, 8p21 (26% of cases) 3p14 (20% of cases), onstrated in columnar cell lesions with increasing
1p35 (20% of cases), and 11q14 (24% of cases), levels shown to occur in parallel with progression
with many comparable losses seen in the adjacent to invasive carcinoma (Verschuur-Maes et al.
low-grade ductal carcinoma in situ and tubular 2012a) supporting a precursor role for these
carcinoma (Aulmann et al. 2009). lesions, although atypia was not associated with
Using comparative genomic hybridization increased levels. Other changes identified as
Simpson was able to demonstrate chromosomal linked to the early phase of the low-grade pathway
losses (6q, 11q, 12q, 16q, 17p, 19q, and 22) and include, in columnar cell hyperplasia, down-
gains (3p, 3q, 7, 8q, 12p, 15q, 16p, 19, and 20) in regulation of the let-7c microRNA in the epithelial
columnar cell lesions, with increasing chromo- compartment and upregulation of miR-132 in the
somal changes seemingly occurring in parallel stroma surrounding columnar cell hyperplasia
with increasing cytological and architectural (Bjorner et al. 2014) possibly reducing some con-
atypia, particularly the recurrent loss of 16q trols on anti-proliferative activity.
(Simpson et al. 2005).
Gains and losses of 17 breast cancer-related
genes were documented for columnar cell lesions Differential Diagnosis
with accompanying ductal carcinoma in situ and
invasive carcinoma. Copy number gains were The diagnosis of columnar cell change, columnar
identified for C11orf30, MYC, CPD, MTDH, cell hyperplasia, and FEA can be difficult to be
CCND1, CCNE1, ESR1, and TOP2A, while losses certain about, particularly on a core needle biopsy
were seen for CDH1 and TOP2A in a small num- where there may be only limited material for
ber of cases, the copy number changes increasing evaluation; however, distinguishing FEA is par-
from columnar cell lesions through to invasive ticularly important as it is associated with a dif-
carcinomas. The relative infrequency of the ferent management outcome for the patient.
changes in the columnar cell lesions compared Similar to columnar cell lesions, apocrine
with those for the in situ and invasive carcinomas metaplasia (▶ Benign and Atypical Apocrine
suggested that they may represent relatively late Lesions) can also be seen in cystically dilated
events in the presumed low-grade pathway acini and the lining cells may also show apical
(Verschuur-Maes et al. 2014). snouts. The cytoplasm of apocrine cells is more
78 Columnar Cell Lesions
Troxell, M. L., Brunner, A. L., Neff, T., Warrick, A., variety of epithelial structures with a complex
Beadling, C., Montgomery, K., et al. (2012). pattern of growth, which can include intraductal
Phosphatidylinositol-3-kinase pathway mutations are
common in breast columnar cell lesions. Modern papillomas (▶ Intraductal Papilloma), sclerosing
Pathology, 25, 930–937. adenosis (▶ Sclerosing Adenosis), and usual-type
Verschuur-Maes, A. H., de Bruin, P. C., & van Diest, P. J. ductal hyperplasia (▶ Usual Ductal Hyperplasia
(2012a). Epigenetic progression of columnar cell (UDH)) (Racz et al. 2017; Ellis and Simpson
lesions of the breast to invasive breast cancer. Breast
Cancer Research and Treatment, 136, 705–715. 2012; Eusebi and Millis 2010; Kennedy C
Verschuur-Maes, A. H., van Deurzen, C. H., et al. 2003).
Monninkhof, E. M., & van Diest, P. J. (2012b). Colum-
nar cell lesions on breast needle biopsies: Is surgical
excision necessary? A systematic review. Annals of
Surgery, 255, 259–265. Clinical Features
Verschuur-Maes, A. H., Moelans, C. B., de Bruin, P. C., &
van Diest, P. J. (2014). Analysis of gene copy number Occasionally, CSLs may be palpable firm irregu-
alterations by multiplex ligation-dependent probe lar masses (Hicks and Lester 2017) or a radiolog-
amplification in columnar cell lesions of the breast.
Cellular Oncology (Dordrecht), 37, 147–154. ical finding of a mass with spiculated margins,
Yu, C. C., Ueng, S. H., Cheung, Y. C., Shen, S. C., but more commonly CSLs cause architectural dis-
Kuo, W. L., Tsai, H. P., et al. (2015). Predictors of tortion on imaging, frequently containing micro-
underestimation of malignancy after image-guided calcifications. These radiological patterns may be
core needle biopsy diagnosis of flat epithelial atypia
or atypical ductal hyperplasia. The Breast Journal, 21, indistinguishable from invasive carcinoma. On
224–232. ultrasound, CSL is seen as hypoechoic area or
mass and at Magnetic Resonance Imaging (MRI)
as an irregular enhancing lesion, usually with
lower enhancement than invasive carcinoma
Complex Sclerosing Lesion (Hicks and Lester 2017).
needle biopsy (CNB) varies widely, between epitheliosis/usual type hyperplasia, apocrine
0% and 23% (Nakhlis et al. 2018). However, metaplasia, and sclerosing adenosis (Racz et al.
the majority of cancers associated with CSL 2017; Ellis and Simpson 2012). All these lesions
are small, well-differentiated invasive carci- may be associated with cytological atypia.
noma and low grade DCIS. On the other At low magnification, two different regions
hand, some authors suggest that CSLs are asso- can be identified, although not pathognomonic
ciated with considerable risk of finding breast of CSL:
carcinoma in adjacent tissue, prompting rec-
ommendation for excision (Kennedy et al. • Central nidus: It is formed by hyalinized col-
2003). In conclusion, if epithelial atypia is lagen stroma with elastosis entrapping and
present on CNB, excision is recommended distorting tubules (two cell layers are retained)
due to the risk of having malignancy associated (Fig. 1, H&E).
to the CSL. It is possible to proceed with • Corona: Sclero-hyalinized bands of connec-
observation when radiological and pathologi- tive tissue radiating from central nidus and
cal features agree on CSL diagnosed on containing distorted tubular structures, fre-
vacuum-assisted CNBs in the absence of quently pointed and of different size. These
other associated high risk lesions (Racz et al. bands dissect normal parenchyma and/or cystic
2017). CSLs diagnosed on excisional surgery lobules and/or lobular structures with various
specimens do not require additional treatment degrees of proliferation and atypia (Fig. 2,
(Patterson et al. 2004). H&E).
• Outcome
As above reported CSL itself is considered
a proliferative benign lesion which exhibits CSL frequently involves papillomas leading
various secondary alterations in the epithelium to the diagnosis of “complex papillary
and carries an approximately twofold sclerosing lesions” (CPSL). The sclero-
increased risk of breast cancer (Racz et al. hyalinazed tissue is predominant in such cases
2017). For this reason, CSLs are diagnosed as and areas of infiltrating epitheliosis (▶ Usual
“lesion of undetermined risk” on core biopsies. Ductal Hyperplasia (UDH)) may be frequently
seen. In some cases of CSL, multiple foci of
“adenosquamous proliferation” (ASP), often
Macroscopy
regarded as representing benign squamous meta-
plasia, are situated within the nidus and
At difference with radial scar, CSLs are generally
permeated between ducts beyond the corona
of sufficient size to produce an irregular firm
mass, of yellow-white color, indurated, some-
times with central retraction. The central core is
usually firmer than the area of radiating arms;
most lesions are <2 cm. The gross appearance
may be indistinguishable from that of an invasive
carcinoma (Ellis and Simpson 2012); neverthe-
less, CSLs are usually firm but not as hard as
invasive carcinomas.
Microscopy
Immunophenotype
• Incidence Macroscopy
DMP accounts for approximately 1% of
benign lesions of the breast (Foschini et al. Diagnosis of DMP is a challenge for both
1990). It is usually associated with type clinicians and radiographers. On mammography,
I DM but it can also be seen, though rarely, the lesion is not well-delineated, with heter-
in type II DM patients (Weidner and Dabbs ogeneous density and no calcifications. It may
2012). be a focally asymmetric mass or a well-
• Age circumscribed nodule resembling a
The average age of female patients with DM, fibroadenoma. Some cases can be interpreted as
suffering from DMP varies between 34 and a carcinoma. All these characteristics make the
47 years. In most of the DM-associated cases, findings rather unspecific (Rosen 2009; Weidner
the interval between DM onset and its clinical and Dabbs 2012).
presentation of DMP is approximately 20 years Ultrasound examination may be a helpful diag-
(Rosen 2009). Long-term insulin-dependent nostic method in DMP cases. It is also useful in
DM and premenopause are considered to be core-needle biopsy diagnosis and follow-up of
among the main predisposing factors for the patients (Rosen 2009).
development of DMP in females (Weidner and The hyperdensity seen in DMP can also cause
Dabbs 2012). difficulties in distinguishing DPM from breast
• Sex carcinoma. In such cases, MRI is a reliable tech-
The changes manifest mostly in female nique for diagnosing malignant neoplastic lesions
patients. Single cases have been reported in (Rosen 2009).
male patients with insulin-dependent DM At macroscopy, DMP usually presents as pal-
(Lee et al. 1996; Rosen 2009). pable, mobile lump, firm on palpation, with gray-
Diabetic Mastopathy 85
white surface. Lumps are irregular in shape, with stroma (Fig. 4a, b). These same cells were
no distinct borders between the lesion and the interpreted as epithelioid fibroblasts and first
surrounding healthy tissue. Occasionally, DMP reported by Tomaszewski et al. (1992) and are
may present as a tumor-like lesion (Foschini seen in about 75% of the cases.
et al. 1990; Weidner and Dabbs 2012). Though rare, there are cases of DMP showing
necrosis and granuloma formation (Weidner and
Dabbs 2012). The histological findings in DMP
Microscopy in male patients share many similarities with the
findings seen in women. Morphologically, DMP D
Histologically, DMP is characterized by presence in the male breast is characterized by lympho-
of spindle to epithelioid myofibroblasts within a cytic periductitis, perivasculitis, dense fibrotic
collagenized frequently dense stroma. In addition, stroma, and epithelioid myofibroblasts (Lee
the main features of the lesion include circumfer- et al. 1996).
ential intralobular, periductal, and perivascular
B type lymphocyte infiltration. The intralobular
lymphocytic reaction is frequently very intense
and shows neat borders, as seen at low power, a
feature often very indicative of the disease. Duct-
ules are consistently atrophic and show thick basal
lamina. (Foschini et al. 1990; Tomaszewski et al.
1992) (Figs. 1, 2, and 3). The stroma varies from
loose to dense. It lacks inflammatory cells. Never-
theless, in the stroma, cells with round to elon-
gated nuclei, evident nucleoli, and abundant
amphophilic cytoplasm are present. These cells
are sparse or can be clumped in nests within the
Diabetic Mastopathy, Fig. 4 (H&E) (a) Sclerohyaline (b) Hyaline fibrous tissue with sparse fibroblasts showing
acellular fibrous tissue centered by a clump of epithelioid abundant cytoplasm. The nuclei are irregular and may
fibroblasts showing ovoid nuclei and well visible nucleoli. simulate neoplastic cells
• Site
Major ducts. May be bilateral.
• Treatment
PDM/DE is a benign lesion with no increased
risk of malignancy. Excision biopsy should be
curative, although not always recommended.
Recurrence is not common (Dabbs 2016).
• Outcome
Although it may be asymptomatic, spontane-
ous and intermittent nipple discharge (clear,
yellow, green, or brown), pain, mass, and Duct Ectasia and Periductal Mastitis, Fig. 1 Gross.
edematous skin changes are described as the
first symptoms and are seen in younger
patients. In a later stage, duct dilatation, nipple 2017). The ductal epithelium is often thin and
retraction, and periductal fibrosis are seen more may be disrupted by inflammatory cells and mac-
preferentially in older patients (Dixon et al. rophages (Fig. 2) (O’Malley and Pinder 2011).
1983, 1996). Periductal inflammatory infiltrate, predominantly
Ulceration of the skin, abscess, and fistulas lymphocytes, and fibrosis are also
may occur and are often described at a later characteristic. Granulomatous inflammation with
stage and can mimic a carcinoma (Moinfar giant cells can occur (O’Malley and Pinder 2011).
2007). Although the inflammatory process is centered in
Mammography abnormalities include large major ducts, it can extend to the peripheral lob-
rod-like branching calcifications, usually ules. Lymphocytic infiltrate in lobules without
>1 mm, with lucent centers. Calcifications granulomas can be seen (Rosen et al. 2014). Stasis
are usually widespread, bilateral, and radially material can be discharged in stroma by disruption
oriented to the nipple-areolar complex of the ducts and cause inflammation and abscess
(Kamal et al. 2009). Lobulated smooth nod- formation (Rosen et al. 2014).
ules or spiculated masses suggesting carci- Duct content is often varied: granular amor-
noma have been described as well (Rosen phous, proteinaceous material admixed with
et al. 2014). foam-cells and desquamated duct epithelial cells.
Histiocytic reaction around cholesterol crystals
can be identified (Rosen et al. 2014).
Macroscopy
The thick layer of hyaline and elastic tissue in a
later stage can encase ducts and in some cases the
The most characteristic alteration in the mammary
lumen becomes totally obliterated by granulation
specimen is the presence of dilated subareolar
tissue – the so called “mastitis obliterans.” Within
ducts; depending on duct contents, cream-
the sclerotic duct, the persisting epithelium may
colored, soft or brown, different kinds of secretion
proliferate forming secondary glands, a pattern
can be observed (Fig. 1 gross appearance:
that resembles a recanalized thrombus in a blood
enlarged ducts are converging toward nipple).
vessel (Rosen et al. 2014).
Microscopy Immunophenotype
Duct Ectasia and Periductal Mastitis, Fig. 2 (H&E) Histiocytes and lymphocytes disrupt the duct epithelium
(Left (400x)); The lumen of the dilated duct is filled with granular, amorphous, eosinophilic material (right (100x))
• Site
Ductal Adenoma Mostly unilateral, in the form of single or mul-
tiple nodules. Bilaterality has been reported in
Handan Kaya patients with Carney Syndrome (Carney and
Pathology Department, Marmara University, Stratakis 1996; Carney and Toorkey 1991)
Istanbul, Turkey • Symptoms and Treatment
Ductal adenoma may present as a palpable
mass or sometimes with nipple discharge.
Synonyms On mammography, it is a well-delineated
nodule, with smooth margins with or
Sclerosing Papilloma is used as a synonym in without coarse calcifications; ultrasonography
WHO 2012 (Foschini et al. 2012). may exhibit hypoechoic structure, sometimes
located within a cyst and MR imaging shows
smooth and clear margins (Matsubayashi et al.
Definition 2016). Excision is curative.
• Outcome
Solid benign tumor of the breast ducts, involving Benign lesion with no reported tendency to
small or medium size ducts described by recurrence.
Azzopardi and Salm (Azzopardi and Salm
1984). These lesions may rarely arise from large-
Macroscopy
sized ducts (Lammie and Millis 1989).
The typical gross appearance is well-
circumscribed, round nodule(s). Macroscopically,
Clinical Features
the lesions’ size varies from 0.5 cm to 4.0 cm. The
cut surface is greyish-white and may be gritty.
• Incidence
It is an uncommon disease.
• Age Microscopy
It is most frequently seen in women over
50 years. Microscopically these lesions are typically
• Sex located in the lumen of a duct demonstrating
Female single or multiple adenomatous nodules
Ductal Adenoma,
Fig. 1 Dilated duct
showing in intraductal
adenomatous growth with
areas of poorly cellular
connective tissue (H&E)
Ductal Carcinoma In Situ 91
composed of ductal epithelial and myoepithelial and complex sclerosing lesions (▶ Complex Scle-
cells surrounding a central scar (Fig. 1). Charac- rosing Lesion). Although sometimes it is not pos-
teristically, they are circumscribed by a thick layer sible to make a differential diagnosis,
of hyalinized fibrosis which often contains demonstrating only small foci of papillary growth
elastic fibers derived from the original ducts. pattern by examining multiple section levels
Hyalinized fibrosis with entrapped tubules, favors ductal adenoma diagnosis.
epithelial atypia, and pseudoinvasion pattern
may mimic malignancy. Demonstration of base-
ment membrane, dual cell population with epithe-
References and Further Reading D
lial and myoepithelial cells forming the tubules Azzopardi, J. G., & Salm, R. (1984). Ductal adenoma of
can be helpful in differential diagnosis. Myxoid the breast: A lesion which can mimic carcinoma.
change, calcifications, and infarction can be Journal of Pathology, 144, 15–23. https://doi.org/
10.1002/path.1711440103.
encountered. More than half of the reported
Carney, J. A., & Stratakis, C. A. (1996). Ductal adenoma
cases showed apocrine differentiation; squamous of the breast and the Carney complex. American Jour-
metaplasia has also been described. nal of Surgical Pathology, 20, 1154–1155.
Carney, J. A., & Toorkey, B. C. (1991). Ductal adenoma
of the breast with tubular features. A probable compo-
nent of the complex of myxomas, spotty pigmentation,
Immunophenotype endocrine overactivity, and schwannomas. American
Journal of Surgical Pathology, 15, 722–731.
Epithelial, myoepithelial, and basal membrane Foschini, M., Simpson, J., & O’Malley, F. (2012). Ductal
markers are expressed as in normal structures. adenoma. In S. Lakhani, I. Ellis, S. Schnitt, P. Tan, &
M. Van de Vijyer (Eds.), WHO classification of tumors
of the breast (pp. 117–118). Lyon: IARC.
Lammie, G. A., & Millis, R. R. (1989). Ductal adenoma
Molecular Features of the breast – A review of fifteen cases. Human Pathol-
ogy, 20, 903–908.
Matsubayashi, R. N., Momosaki, S., & Muranaka, T.
Molecular basis of these tumors is unknown. Next (2016). Ductal adenoma of breast: imaging characteris-
generation sequencing of 50 cancer-related genes tics and radiologic-pathologic correlation of unique find-
and Sanger sequencing analysis of nine ductal ings which reflect “pseudoinvasion”. Breast Cancer, 23,
597–606. https://doi.org/10.1007/s12282-015-0608-9.
adenomas showed 56% (5/9) harboring mutated
Troxell, M. L., Levine, J., Beadling, C., Warrick, A., Dunlap,
genes and from those 30% demonstrated mutant J., Presnell, A., Patterson, J., Shukla, A., Olson, N. R.,
AKT1 and 22% (3/9) an additional GNAS muta- Heinrich, M. C., & Corless, C. L. (2010). High preva-
tion, 11% (1/9) had mutant PIK3CA. AKT lence of PIK3CA/AKT pathway mutations in papillary
neoplasms of the breast. Modern Pathology, 23, 27.
1 E17 K mutations demonstrated in breast papil-
lomas without atypia supports the relation
between sclerosing intraductal papillomas and
ductal adenomas (Troxell et al. 2010). Ductal Carcinoma In Situ
Definition • Site
The anatomic location of DCIS is similar to
Ductal carcinoma in situ (DCIS) is a neoplastic invasive carcinoma location. The majority
proliferation confined to the mammary ductal- of tumors are found in the upper outer quad-
lobular system, which is surrounded by a rant (43.9%), followed by the upper inner
myoepithelial layer and a basal membrane. DCIS quadrant (9.0%), the central quadrant
covers a heterogeneous group of diseases charac- (8.5%), the lower outer quadrant (8.1%), and
terized by subtle to marked cytological atypia, finally the lower inner quadrant (6.9%)
without any invasion of the stromal tissue (Ernster et al. 2000).
(Lakhani et al. 2012). In a study by Verkooijen et al. (2002), 35%
of DCIS were present in two different quad-
rants. Multi-quadrant presentation is often
Clinical Features observed, especially in micropapillary DCIS
(Bellamy et al. 1993).
• Incidence • Treatment
DCIS incidence gradually and constantly Surgery: over the past 20 years, the surgical
increased over the past decades (from 5.8 approach of DCIS changed from radical mastec-
per 100,000 women in 1975 to 34.4 per tomy with sentinel node (▶ Sentinel Node) to
100,000 women in 2014), according to data conservative surgery, without any intervention
of the National Cancer Institute’s Surveil- on axilla, as recommended by ASCO (Lyman
lance, Epidemiology, and End Results Pro- et al. 2014) and NCCN guidelines (2015)).
gram (SEER 2014). DCIS consequently Some studies suggest that low-grade lesions can
constitutes around 20% of all breast cancer be treated only with surgical excision excluding
diagnoses. radiotherapy (EORTC Breast Cancer Coopera-
The widespread use of mammography, as tive Group et al. 2006), while others reported
well as implementation of national breast that being often multicentric, low-grade DCIS
screening programs (Ernster and Barclay may recur and it may remain radiologically
1997), contributed to the diagnosis of breast silent, even if extensive (Tot and Gere 2008).
cancer in early stage. According to different Mastectomy with immediate breast recon-
studies, approximately 1 in every 1300 screen- struction is used for large/diffuse DCIS, in case
ing mammography leads to a diagnosis of of contralateral prophylactic mastectomy
DCIS (Ernster et al. 2002). (Rutter et al. 2015), especially in younger
• Age women, as well as in patients with BRCA
DCIS rarely occurs in women younger than mutations or with positive family history for
40 years, and its incidence rises between ovarian cancer (Elsayegh et al. 2014). The
40 and 70 years, with a plateau after the age main feature that guides surgical approach is
of 70. the lesion size, measured with mammography
• Sex and magnetic resonance (MR). In case of con-
DCIS is common in women. However, servative approach, the margins should be free
according to SEER database, DCIS represents of disease with a minimum recommended dis-
9.4% of all male breast carcinomas (Anderson tance of 2 mm (Pilewskie and Morrow 2018).
and Devesa 2005) with an average age of Very recently three multi-institutional, random-
62 years. Although all DCIS subtypes could ized phase III trials are addressed to avoid sur-
be present in both genders, in general, papillary gery in case of low-grade DCIS, in favor of an
in situ (▶ Intraductal Papillary Carcinoma) active surveillance strategy (Elshof et al. 2015;
lesions are more common in men (Staerkle COMET trial- https://clinicaltrials.gov/ct2/show/
et al. 2006). NCT02926911; Francis et al. 2015).
Ductal Carcinoma In Situ 93
Radiotherapy: conservative surgery is gen- radiation were 73% and 78%, respectively
erally associated with postsurgical radiother- (Kong et al. 2014). The presence of comedo-
apy, regardless of DCIS histology. necrosis is closely related to the risk of ipsilat-
Randomized trials have proven that RT reduces eral recurrences following lumpectomy, and a
local recurrence rates by 50% (McCormick meta-analysis demonstrated that this risk
et al. 2015; EORTC Breast Cancer Cooperative ranges from 1.3 to 5.0 (Wapnir et al. 2011;
Group et al. 2006); however, many studies are Wai et al. 2011).
now addressed to avoid radiation in low-risk Observational studies reported that about
DCIS. Several trials have been performed 14–50% of DCIS may progress to invasive D
using the multigene assay, Oncotype DX ® carcinomas (King et al. 2017; Erbas et al.
(Genentech, Houston, TX), able to quantify 2006; Pluchinotta 2018). Many evidences
the 10-year risk of local recurrences, both for suggested that the majority of invasive carci-
in situ and invasive breast cancer. The results noma may derive from DCIS disease. Actually,
are reported as a numerical score called “DCIS invasive cancers are generally accompanied by
Score,” which classifies patients into low-, DCIS, which share similar molecular features.
intermediate-, and high-risk group. It has However, a high number of DCIS was found in
been proposed to avoid radiotherapy in low- autopsy studies, to emphasize that not all DCIS
risk categories (Solin et al. 2013). will progress to invasive cancer (Pluchinotta
Hormonal treatment: 75–80% of DCIS 2018).
express estrogen receptor (ER). Hormonal treat-
ment (HT) is debated; nevertheless, HT for
patients with ER-positive DCIS has been dem-
onstrated to reduce recurrent events of about Macroscopic Features
37% in patients with conservative surgery
(Wapnir et al. 2011). The NSABP B-35 trial The majority of DCIS are detected by mammog-
demonstrated similar rates of disease-free sur- raphy due to microcalcifications; thus, the macro-
vival comparing tamoxifen and aromatase scopic appearance may be completely negative.
inhibitors in postmenopausal patients with Sometimes these lesions may form a nodular
DCIS (Margolese et al. 2016). thickening or spot gray-yellow lesions with strand
Anti-HER2 therapy: Although trastuzumab of dense material that may exude from the cut
treatment (Kuerer et al. 2011) or lapatinib surface, especially in case of comedo-necrosis.
(Decensi et al. 2011) in HER2-positive DCIS To better define extension of the lesion, margin
is proposed, so far, the clinical effect of anti- status, and the correlation with radiological fea-
HER2 blockade is unclear. A prospective, ran- tures (calcifications on mammography and on
domized phase III multi-institutional clinical intraoperative radiogram), the use of large histo-
trial – National Surgical Adjuvant Breast Pro- logical sections in routine clinical practice is
ject (NSABP) – treating HER2-positive DCIS encouraged. However, not every case of DCIS
patients with trastuzumab associated with calcifies, or calcifications may be partially pre-
radiotherapy (Siziopikou et al. 2013) is cur- sent, and, when measured by mammography, the
rently in progress. real size of the lesion may be underestimated. In
• Outcome 10% of cases, DCIS grows in form of nodular
The age at diagnosis has been identified as the mass or architectural distortion (Giuseppetti
strongest predictor of invasive and in situ local et al. 2018). At ultrasound, DCIS calcifications
recurrences. For women under 45 years of age manifest as echogenic foci within the breast tis-
at diagnosis, the 10-year local recurrence-free sue, unlike non-calcified DCIS, which may appear
survival and invasive local recurrence-free sur- as hypoechoic mass with microlobulated margins.
vival after breast-conserving surgery and MR is the most sensitive examination to evaluate
94 Ductal Carcinoma In Situ
the correct size of DCIS, mainly in high-grade The lumens may contain amorphous material with
lesions (Greenwood et al. 2013). The most apoptotic cells, calcifications (generally with
common morphologic feature of DCIS at MR is powdered/granular pattern), and necrosis. Micro-
“nonmass” enhancement (60–81% of cases) papillary DCIS is composed of numerous epithe-
(Greenwood et al. 2013). Although many lial projections lacking fibrovascular cores
studies have demonstrated overestimations of the (Fig. 2c. H&E); these cell clusters may detach in
disease extent by MR (Rominger et al. 2016), this the lumen and flow to the nipple (Castellano et al.
method principally guides the surgical treatment 2010). Papillary DCIS (▶ Intraductal Papillary
approach. Carcinoma) is characterized by fibrovascular pap-
illary fronds. In some cases, the papillary over-
growth fills the lumen completely, simulating
Microscopy solid DCIS, but fibrovascular cores are still iden-
tifiable. WHO classification system categorized
Nuclear Grade papillary lesions as a separate group, composed
Classification of DCIS should be based primarily of intraductal (▶ Intraductal Papillary Carci-
on nuclear grade, as it represents the most noma), encapsulated (▶ Encapsulated Papillary
significant predictor of local recurrence at both Carcinoma), and solid papillary carcinoma
univariate and multivariate analyses (Wapnir (▶ Solid Papillary Carcinoma) (Lakhani et al.
et al. 2011). 2012). While the intraductal papillary carcinoma
Depending on the degree of nuclear atypia, is characterized by the presence of myoepithelial
DCIS is generally classified in low-, intermedi- layer, the other two entities may lack it at the
ate-, and high-grade DCIS. In particular, low periphery of the lesion. For this reason, there is
grade is characterized by small, monomorphic, no universal agreement on how to stage papillary
well-polarized cells, with uniform size and carcinomas; however, it is suggested that both
regular chromatin pattern and rare mitotic encapsulated and solid type should be managed
figures (Fig. 1a. H&E). Microcalcifications as DCIS form. Comedo DCIS is characterized by
are often of psammomatous/granular/powdery a strongly atypical cell proliferation distending
type. DCIS of intermediate nuclear grade and enlarging ducts with central necrotic debris
consists of cells similar to those of low grade and amorphous calcifications (Fig. 2d. H&E).
but with variability in size, shape and placement, These generally appear as crushed-stone-like/
occasional nucleoli, mitotic figures, and casting-type calcifications, distributed with a lin-
coarse chromatin (Fig. 1b. H&E). Finally, high- ear or segmental pattern at mammography exam-
nuclear-grade DCIS is composed of highly ination. The periductal stroma may be dense and
atypical cells, large in size, with pleomorphic thick and frequently shows a mild inflammatory
and poorly polarized irregular nuclei, prominent reaction. Comedo DCIS usually belongs to high
nucleoli, numerous mitotic figures, and presence or intermediate histopathology nuclear grade.
of necrosis (Fig. 1c. H&E) (Tavassoli and
Devilee 2003). The “DIN” Classification
In 2003 Tavassoli et al. proposed a novel classi-
Histological Growth Patterns fication system (Tavassoli and Devilee 2003),
The histological variants are solid, cribriform, suggesting to replace the term DCIS in favor of
micropapillary, papillary, comedo (Fig. 2. H&E), “ductal intraepithelial neoplasia” (DIN), reserv-
apocrine, and mixed (Bellamy et al. 1993). Solid ing the term “carcinoma” only for invasive
DCIS is characterized by an orderly proliferation tumors.
of crowded neoplastic cells that expand the ducts The subgroup of lesions classified as “DIN 1”
(Fig. 2a. H&E). Cribriform DCIS is formed by encompasses a series of low-grade intraductal
well-defined roundish lumens regularly spaced, proliferations, such as flat epithelial atypia
lined by uniform polarized cells (Fig. 2b. H&E). (▶ Columnar Cell Lesions) (DIN1a), atypical
Ductal Carcinoma In Situ 95
Ductal Carcinoma In Situ, Fig. 1 (a) Low-grade DCIS presents occasional mitotic figures and coarse chromatin.
is characterized by small, monomorphic, well-polarized (c) High-grade DCIS is composed of highly atypical cells,
cells, with uniform size. (b) Intermediate-grade DCIS con- large in size, with pleomorphic and poorly polarized irreg-
sists of cells variable in size, shape, and placement and ular nuclei and presence of necrosis (H&E, 20x)
ductal hyperplasia (▶ Atypical Ductal Hyperpla- and pleomorphic cells and shows genetic alter-
sia) (ADH) (DIN 1b), and low-grade DCIS ations of high-grade neoplasia (Reis-Filho
(DIN1c). Molecular studies demonstrated that et al. 2005).
these lesions share similar genetic alterations, typ- DIN terminology did not reach widespread
ical of low-grade neoplasia (Reis-Filho consensus. Thus, the WHO in 2012 (Lakhani
et al. 2005). et al. 2012) abandoned the term DIN in favor of
DIN 2 represents intermediate-nuclear-grade DCIS classifications based on the nuclear grade.
DCIS, with intermediate level of differentiation Some authors still support the use of “DIN”
between low- and high-grade lesions. This nomenclature, in order to reduce negative psycho-
latter called DIN 3 is characterized by atypical logical impact on patients and confusion in
96 Ductal Carcinoma In Situ
Ductal Carcinoma In Situ, Fig. 2 Different histologic composed of numerous epithelial projections into the
subtypes of DCIS. (a) Solid DCIS is characterized by a lumen. Clusters of neoplastic cells without fibrovascular
proliferation of crowded neoplastic cells, which fills the cores are floating in the lumen. (d) Comedo DCIS is
lumen. (b) Cribriform DCIS is represented by neoplastic characterized by enlarged ducts containing necrotic debris.
lumens regularly spaced. The lumens contain amorphous The periductal stroma shows mild inflammatory reaction
material with calcifications. (c) Micropapillary DCIS is (H&E, 10x)
medical management of in situ lesions CD10, cytokeratin 5/6) or their nucleus (p63 and
(Galimberti et al. 2013). p40) (Fig. 3. p63 immunostaining). In routine
practice, the use of both nuclear and cytoplasmic
antibodies is recommended. ERs and progester-
Immunophenotype one receptors (PR) are included as part of the
workup of DCIS by the National Comprehensive
Myoepithelial cells around DCIS may be Cancer Network guidelines. ER is found positive
highlighted by antibodies which stain either in 75–80% of DCIS lesions. Its expression is
their cytoplasm (smooth muscle actin, calponin, generally related to low-intermediate-nuclear-
Ductal Carcinoma In Situ 97
characterized by complex karyotypes (Reis- composed of collagen type IV, laminin 1, and
Filho et al. 2005; Lopez-Garcia et al. 2010), some proteoglycans. The overexpression of
including gain in 1q+, 5p+, 8q+, and 17q+; loss some markers, such as matrix metalloproteinase
in 8p , 11q , 13 , and 14q ; and focal ampli- 2 (MMP2), involved in extracellular matrix
fications on 6q22, 8q22, 11q13, 17q12, remodeling, may provoke basal membrane degra-
17q22–24, and 20q13 (Reis-Filho and Lakhani dation (Abba et al. 2004), allowing cancer stromal
2003; Shackney and Silverman 2003). As a result invasion.
of such molecular profile, it is hypothesized that Recently, a number of studies have focused
DCIS may progress along two distinct pathways on analysis of microRNA (miRNA), a class of
of low or high grade. The low-grade arm is char- small RNA molecules that control mRNA expres-
acterized by pathways related to ER activation sion and regulate stem cell division, cell
and well-differentiated invasive carcinoma (such growth, apoptosis, and carcinogenesis. In com-
as tubular carcinoma (▶ Tubular Carcinoma)). parison to normal breast tissue, in DCIS,
The majority of high-grade DCIS lesions show some miRNAs are under- or overexpressed
identical genomic profile of invasive cancers (Hannafon et al. 2011). miRNA deregulations
(Abba et al. 2015). It has been shown that TP53 seem to occur during the transition from
(Kim et al. 2015), PTEN (Yates et al. 2015) normal to DCIS epithelium, such as the loss of
mutations, as well as amplifications of chromo- miR-125b and miR-132 and the gain of miR-182
some 20, 11, and 17 (Aubele et al. 2000) may and miR-183 (Hannafon et al. 2011). A specific
occur in the transition from normal epithelium to miRNA signature was described as differently
in situ carcinoma cells (Casasent et al. 2017). expressed in invasive and DCIS lesions (Volinia
Concomitant factors such as stromal environ- et al. 2012).
ment, cell-mediated immune mechanisms, vas- Finally, DNA methylation seems to play a piv-
cular spaces, and myoepithelial cells contribute otal role in early breast carcinogenesis with an
to progression of DCIS to invasive breast cancer increase of methylated genes from normal breast
(Mardekian et al. 2016; Adriance et al. 2005). In to DCIS (Mardekian et al. 2016).
particular, tumor suppressor functions of
myoepithelial layer are well established, with
the maintenance of integrity of basal membrane Differential Diagnosis
and epithelial cell polarity (Polyak and Hu 2005).
A study conducted by Allinen et al. (2004), com- The differential diagnosis of DCIS may include
paring the microenvironment of normal and can- (i) epitheliosis/usual ductal hyperplasia (UDH),
cerous breast tissue, discovered that the DCIS (ii) atypical ductal hyperplasia (ADH), (iii) lobu-
myoepithelial layer displayed the most abundant lar carcinoma in situ (LCIS), and (iv) minimally
gene expression changes of all the microenviron- invasive carcinoma:
mental cell types. Actually, myoepithelium sur-
rounding DCIS shows some morphological 1. The solid growth pattern of DCIS can be
differences compared to normal breast tissue, misinterpreted as florid epitheliosis/UDH
losing the power to polarize luminal epithelial lesions (▶ Usual Ductal Hyperplasia (UDH)).
cells and showing a discontinuity in the expres- Cell proliferation of epitheliosis is heteroge-
sion of myoepithelial markers (Rakha et al. neous; cells show different shapes, sizes, and
2017). Moreover, it has been shown that genes streaming arrangements and irregular periph-
that are specific for normal myoepithelial cells, eral fenestrations. Cells forming the bridges in
such as CTK14, CTK17, and EGFR, are absent low grade DCIS are more clonal and arranged
or downregulated in DCIS myoepithelial cells haphazardly or transversely to the long axis
(Polyak and Hu 2005). and do not manifest streaming pattern. More-
Besides myoepithelium, basal membrane rep- over, an immunohistochemical mosaicism of
resents a constitutive barrier to invasion, CK5/14+ and CK5/14- cells is characteristic
Ductal Carcinoma In Situ 99
relevance of histologic categorization. Human Pathol- mammography: A dilemma. Journal of the National
ogy, 24, 16–23. Cancer Institute Monographs, 22, 151–156.
Böcker, W. (2006). Preneoplasia of the breast. A new Ernster, V. L., Barclay, J., Kerlikowske, K., et al. (2000).
conceptual approach to proliferative breast disease. Mortality among women with ductal carcinoma in
Munich: Elsevier GmbH. situ of the breast in the population-based surveillance,
Bombonati, A., & Sgroi, D. C. (2011). The molecular epidemiology and end results program. Archives of
pathology of breast cancer progression. Journal of Internal Medicine, 160, 953–958.
Pathology, 223, 307–317. Ernster, V. L., Ballard-Barbash, R., Barlow, W. E., et al.
Bratthauer, G. L., Moinfar, F., Stamatakos, M. D., et al. (2002). Detection of ductal carcinoma in situ in women
(2002). Combined E-cadherin and high molecular undergoing screening mammography. Journal of the
weight cytokeratin immunoprofile differentiates lobu- National Cancer Institute, 94, 1546–1554.
lar, ductal, and hybrid mammary intraepithelial neo- Francis, A., Fallowfield, L., & Rea, D. (2015). The LORIS
plasias. Human Pathology, 33, 620–627. trial: Addressing overtreatment of ductal carcinoma
Casasent, A. K., Edgerton, M., & Navin, N. E. (2017). in situ. Clinical Oncology, 27, 6–8.
Genome evolution in ductal carcinoma in situ: Invasion Galimberti, V., Monti, S., & Mastropasqua, M. G. (2013).
of the clones. Journal of Pathology, 241, 208–218. DCIS and LCIS are confusing and outdated terms.
Castellano, I., Marchiò, C., Tomatis, M., et al. (2010). They should be abandoned in favor of ductal
Micropapillary ductal carcinoma in situ of the breast: intraepithelial neoplasia (DIN) and lobular
An inter-institutional study. Modern Pathology, 23, intraepithelial neoplasia (LIN). Breast, 22, 431–435.
260–269. Giuseppetti, G. M., Boria, F., & Baldassare, S. (2018).
Comparison of Operative to Monitoring and Endocrine DCIS imaging. In C. Mariotti (Ed.), Ductal carcinoma
Therapy (COMET). Trial For Low Risk DCIS in situ (pp. 39–57). Cham: Springer.
(COMET). https://clinicaltrials.gov/ct2/show/NCT029 Greenwood, H. I., Heller, S. L., Kim, S., et al. (2013).
26911 Ductal carcinoma in situ of the breasts: Review of
Curigliano, G., Disalvatore, D., Esposito, A., et al. (2015). MR imaging features. Radiographics, 33, 1569–1588.
Risk of subsequent in situ and invasive breast cancer in Hannafon, B. N., Sebastiani, P., de las Morenas, A., et al.
human epidermal growth factor receptor 2-positive (2011). Expression of microRNA and their gene targets
ductal carcinoma in situ. Annals of Oncology, 26, are dysregulated in preinvasive breast cancer. Breast
682–687. Cancer Research, 13, R24.
Decensi, A., Puntoni, M., Pruneri, G., et al. (2011). Holmes, P., Lloyd, J., Chervoneva, I., et al. (2011).
Lapatinib activity in premalignant lesions and HER-2- Prognostic markers and long-term outcomes in ductal
positive cancer of the breast in a randomized, placebo- carcinoma in situ of the breast treated with excision
controlled presurgical trial. Cancer Prevention alone. Cancer, 117, 3650–3657.
Research, 4, 1181–1189. Kim, S. Y., Jung, S. H., Kim, M. S., et al. (2015). Genomic
Elsayegh, N., Kuerer, H. M., Lin, H., et al. (2014). differences between pure ductal carcinoma in situ and
Predictors that influence contralateral prophylactic synchronous ductal carcinoma in situ with invasive
mastectomy election among women with ductal carci- breast cancer. Oncotarget, 6, 7597–7607.
noma in situ who were evaluated for BRCA genetic King, M. T., Winters, Z. E., Olivotto, I. A., et al. (2017).
testing. Annals of Oncology, 21, 3466–3472. Patient-reported outcomes in ductal carcinoma in
Elshof, L. E., Tryfonidis, K., Slaets, L., et al. (2015). situ: A systematic review. European Journal of Cancer,
Feasibility of a prospective, randomised, open-label, 71, 95–108.
international multicentre, phase III, non-inferiority Kong, I., Narod, S. A., Taylor, C., et al. (2014). Age at
trial to assess the safety of active surveillance for low diagnosis predicts local recurrence in women treated
risk ductal carcinoma in situ – the LORD study. with breast-conserving surgery and postoperative
European Journal of Cancer, 51, 1497–1510. radiation therapy for ductal carcinoma in situ:
EORTC Breast Cancer Cooperative Group, EORTC A population-based outcomes analysis. Current Oncol-
Radiotherapy Group, Bijker, N., et al. (2006). Breast- ogy, 21, e96–e104.
conserving treatment with or without radiotherapy in Kuerer, H. M., Buzdar, A. U., Mittendorf, E. A., et al.
ductal carcinoma-in-situ: Ten-year results of European (2011). Biologic and immunologic effects of preopera-
Organisation for Research and Treatment of Cancer tive trastuzumab for ductal carcinoma in situ of the
randomized phase III trial 10853 – a study by the breast. Cancer, 117, 39–47.
EORTC Breast Cancer Cooperative Group and Lakhani, S. R., Ellis, I. O., Schnitt, S. J., et al. (2012). WHO
EORTC Radiotherapy Group. Journal of Clinical classification of tumors of the breast. Lyon: Interna-
Oncology, 24, 3381–3387. tional Agency for Research on Cancer.
Erbas, B., Provenzano, E., Armes, J., et al. (2006). The Lopez-Garcia, M. A., Geyer, F. C., Lacroix-Triki, M., et al.
natural history of ductal carcinoma in situ of the (2010). Breast cancer precursors revisited: Molecular
breast: A review. Breast Cancer Research and Treat- features and progression pathways. Histopathology, 57,
ment, 97, 135–144. 171–192.
Ernster, V. L., & Barclay, J. (1997). Increases in ductal Lyman, G. H., Temin, S., Edge, S. B., et al. (2014). Sentinel
carcinoma in situ (DCIS) of the breast in relation to lymph node biopsy for patients with early-stage breast
Ductal Carcinoma In Situ 101
cancer: American Society of Clinical Oncology clinical of tumour size measurements in breast cancer?
practice guideline update. Journal of Clinical Oncol- European Radiology, 26, 1457–1465.
ogy, 32, 1365–1383. Rutter, C. E., Park, H. S., Killelea, B. K., et al. (2015).
Mardekian, S. K., Bombonati, A., & Palazzo, J. P. (2016). Growing use of mastectomy for ductal carcinoma-in
Ductal carcinoma in situ of the breast: The importance situ of the breast among young women in the United
of morphologic and molecular interactions. Human States. Annals of Surgical Oncology, 22, 2378–2386.
Pathology, 49, 114–123. Shackney, S. E., & Silverman, J. F. (2003). Molecular
Margolese, R. G., Cecchini, R. S., Julian, T. B., et al. evolutionary patterns in breast cancer. Advances in
(2016). Anastrozole versus tamoxifen in postmeno- Anatomic Pathology, 10, 278–290.
pausal women with ductal carcinoma in situ undergo- Siziopikou, K. P., Anderson, S. J., Cobleigh, M. A., et al.
ing lumpectomy plus radiotherapy (NSABP B-35): (2013). Preliminary results of centralized HER2 testing D
A randomised, double-blind, phase 3 clinical trial. in ductal carcinoma in situ (DCIS): NSABP B-43.
Lancet, 387, 849–856. Breast Cancer Research and Treatment, 142, 415–421.
McCormick, B., Winter, K., Hudis, C., et al. (2015). RTOG Solin, L. J., Gray, R., Baehner, F. L., et al. (2013).
9804: A prospective randomized trial for good-risk A multigene expression assay to predict local recur-
ductal carcinoma in situ comparing radiotherapy rence risk for ductal carcinoma in situ of the breast.
with observation. Journal of Clinical Oncology, 33, Journal of the National Cancer Institute, 105,
709–715. 701–710.
Mustonen, M., Raunio, H., Pääkkö, P., et al. (1997). The Staerkle, R. F., Lenzlinger, P. M., Suter, S. L., et al. (2006).
extent of apoptosis is inversely associated with bcl-2 Synchronous bilateral ductal carcinoma in situ of the
expression in premalignant and malignant breast male breast associated with gynecomastia in a 30-year-
lesions. Histopathology, 31, 347–354. old patient following repeated injections of stanozolol.
NCCN. (2015). Guidelines: https://www.nccn.org/profes Breast Cancer Research and Treatment, 97, 173–176.
sionals/physician_gls/default.aspx Surveillance, Epidemiology, and End Results (SEER) Pro-
Page, D. L., & Rogers, L. W. (1992). Combined histologic gram. (2014). National Cancer Institute. https://seer.
and cytologic criteria for the diagnosis of mammary cancer.gov/csr/1975_2014/
atypical ductal hyperplasia. Human Pathology, 23, Tavassoli, F. A., & Devilee, P. (2003). World Health
1095–1097. Organization classification of tumors of the breast.
Patchefsky, A. S., Schwartz, G. F., Finkelstein, S. D., et al. Lyon: International Agency for Research on Cancer.
(1989). Heterogeneity of intraductal carcinoma of the Tot, T., & Gere, M. (2008). Radiological-pathological
breast. Cancer, 63, 731–741. correlation in diagnosing breast carcinoma: The role
Pilewskie, M., & Morrow, M. (2018). Margins in of pathology in the multimodality era. Pathology &
breast cancer: How much is enough? Cancer, 124, Oncology Research, 14, 173–178.
1335–1341. Verkooijen, H. M., Fioretta, G., De Wolf, C., et al. (2002).
Pluchinotta, A. (2018). History of the DCIS of the breast Management of women with ductal carcinoma in situ
and the evolution of knowledge based on ductal tree of the breast: A population-based study. Annals of
anatomy. In C. Mariotti (Ed.), Ductal Carcinoma in situ Oncology, 13, 1236–1245.
(pp. 1–23). Cham: Springer. Vincent-Salomon, A., Lucchesi, C., Gruel, N., et al.
Polyak, K., & Hu, M. (2005). Do myoepithelial cells hold (2008). Integrated genomic and transcriptomic analysis
the key for breast tumor progression? Journal of Mam- of ductal carcinoma in situ of the breast. Clinical
mary Gland Biology and Neoplasia, 10, 231–247. Cancer Research, 14, 1956–1965.
Rakha, E. A., Miligy, I. M., Gorringe, K. L., et al. Volinia, S., Galasso, M., Sana, M. E., et al. (2012). Breast
(2017). Invasion in breast lesions: The role of the cancer signatures for invasiveness and prognosis
epithelial-stroma barrier. Histopathology, 72, 1075. defined by deep sequencing of microRNA. Proceed-
https://doi.org/10.1111/his.13446. ings of the National Academy of Sciences of the United
Ravaioli, S., Tumedei, M. M., Foca, F., et al. (2017). Andro- States of America, 109, 3024–3029.
gen and oestrogen receptors as potential prognostic Wai, E. S., Lesperance, M. L., Alexander, C. S., et al.
markers for patients with ductal carcinoma in situ treated (2011). Predictors of local recurrence in a population-
with surgery and radiotherapy. International Journal of based cohort of women with ductal carcinoma in situ
Experimental Pathology, 98, 289–295. treated with breast conserving surgery alone. Annals of
Reis-Filho, J. S., & Lakhani, S. R. (2003). The diagnosis Surgical Oncology, 18, 119–124.
and management of pre-invasive breast disease: Wapnir, I. L., Dignam, J. J., Fisher, B., et al. (2011). Long-
Genetic alterations in pre-invasive lesions. Breast term outcomes of invasive ipsilateral breast tumor
Cancer Research, 5, 313–319. recurrences after lumpectomy in NSABP B-17 and
Reis-Filho, J. S., Simpson, P. T., Gale, T., et al. (2005). The B-24 randomized clinical trials for DCIS. Journal of
molecular genetics of breast cancer: The contribution of the National Cancer Institute, 103, 478–488.
comparative genomic hybridization. Pathology, Yates, L. R., Gerstung, M., Knappskog, S., et al. (2015).
Research and Practice, 201, 713–725. Subclonal diversification of primary breast cancer
Rominger, M., Berg, D., Frauenfelder, T., et al. (2016). revealed by multiregion sequencing. Nature Medicine,
Which factors influence MRI-pathology concordance 21, 751–759.
E
• Treatment
Because of its low incidence, no clearly
defined guidelines have been established for
EPC treatment. Breast conserving surgery is
the standard treatment for patients with the
pure form. In addition to surgical excision,
several studies report on the use of adjuvant
radiation, chemotherapy, or endocrine therapy
in the management of EPC; however, their role
remains undefined (Solorzano et al. 2002;
Fayanju et al. 2007).
Due to the low incidence of axillary lymph
node metastases, sentinel node (▶ Sentinel Encapsulated Papillary Carcinoma, Fig. 1 Low power
Node) evaluation is suggested only in EPC magnification of encapsulated papillary carcinoma shows a
associated to invasive component (Solorzano well-defined lesion surrounded by a fibrous capsule. The
proliferating epithelium has a papillary and cribriform pat-
et al. 2002). tern (H&E 4)
• Outcome
EPC presents a favorable prognosis, even
when associated with invasion (Rakha et al.
2011), showing low local recurrence, rare dis-
tant metastases, and an excellent survival (91%
disease-free survival rate at 10-year follow-up)
(Lefkowitz et al. 1994).
Macroscopy
Differential Diagnosis
Fayanju, O. M., Ritter, J., Gillanders, W. E., Eberlein, T. J., the breast: A SEER database analysis of implications
Dietz, J. R., Aft, R., & Margenthaler, J. A. (2007). for therapy. The Breast, 27, 87–92.
Therapeutic management of intracystic papillary carci- Rakha, E. A., Gandhi, N., Climent, F., van Deurzen, C. H.,
noma of the breast: The roles of radiation and endocrine Haider, S. A., Dunk, L., Lee, A. H., Macmillan, D., &
therapy. The American Journal of Surgery, 194, Ellis, I. O. (2011). Encapsulated papillary carcinoma of
497–500. the breast: An invasive tumor with excellent prognosis.
Hill, C. B., & Yeh, I. T. (2005). Myoepithelial cell staining The American Journal of Surgical Pathology, 35,
patterns of papillary breast lesions: From intraductal 1093–1103.
papillomas to invasive papillary carcinomas. American Solorzano, C. C., Middleton, L. P., Hunt, K. K., Mirza, N.,
Journal of Clinical Pathology, 123, 36–44. Meric, F., Kuerer, H. M., Ross, M. I., Ames, F. C., Feig,
Kővári, B., Ormándi, K., Simonka, Z., Vörös, A., & B. W., Pollock, R. E., Singletary, S. E., & Babiera,
Cserni, G. (2018). Apocrine encapsulated papillary G. (2002). Treatment and outcome of patients with
carcinoma of the breast: The first reported case with intracystic papillary carcinoma of the breast. The Amer-
an infiltrative component. Journal of Breast Cancer, ican Journal of Surgery, 184, 364–368.
21, 227–230. Turahvili, G., Brogi, E., Morrow, M., Hudis, C., Dickler,
Lakhani, S.R., Ellis, I.O., Schnitt, S.J., Tan, P.H., & van de M., Norton, L., & Wen, H. Y. (2017). The 21-gene
Vijver, M.J.. (2012). WHO classification of tumors of recurrence score in special histologic subtypes of breast
the breast (pp. 106–107). World Health Organization. cancer with favorable prognosis. Breast Cancer
ISBN-10: 9283224337. Lyon, IARC press. Research and Treatement, 165(1), 65–76.
Lefkowitz, M., Lefkowitz, W., & Wargotz, E. S. (1994). Valdes, E. K., Tartter, P. I., Genelus-Dominique, E.,
Intraductal (intracystic) papillary carcinoma of the Guilbaud, D. A., Rosenbaum-Smith, S., & Estabrook,
breast and its variants: A clinicopathological study of A. (2006). Significance of papillary lesions at percuta-
77 cases. Human Pathology, 25, 802–809. neous breast biopsy. Annals of Surgical Oncology, 13,
Li, X., Xu, Y., Ye, H., Qin, S., Hou, F., & Liu, W. (2018). 480–482.
Encapsulated papillary carcinoma of the breast: Zhang, J., Zhang, T., Wu, N., Zhao, X., Wang, Q., Jiang, Y.,
A clinicopathological study of 49 cases. Current Prob- Gao, M., & Gu, L. (2018). Intracystic papillary carci-
lems in Cancer, 42, 291–301. noma of the breast: Experience of a major Chinese
Mogal, H., Brown, D. R., Isom, S., Griffith, K., & Howard- cancer center. Pathology-Research and Practice, 214,
McNatt, M. (2016). Intracystic papillary carcinoma of 579–585.
F
Fibroadenoma • Imaging
Mammography can detect FA if a calcific com-
Elena Maldi1 and Anna Sapino1,2 ponent is present, otherwise it cannot distin-
1
Unit of Pathology, Candiolo Cancer Institute guish FAs and cysts. FA is a round or ovoid
FPO-IRCCS, Candiolo, Italy hypoechoic lesion at ultrasound evaluation.
2
Department of Medical Sciences, University of • Treatment & Outcome
Turin, Turin, Italy FAs are painless lesions with slow growth thus
they do not require any treatment, but imaging
follow up. About 10% to 40% of FAs resolve
Synonyms spontaneously or if completely excised they do
not recur (Song et al. 2014). Cases of juvenile FA
Benign fibroepithelial lesion (see below) can grow rapidly and occupy most of
the breast resulting in esthetic problems due to
breast asymmetry or hypertrophy. Complete
Definition lumpectomy is recommended because they may
recur in 33% at 5 years (Song et al. 2014).
Fibroadenoma (FA) is a common benign neo- Complex FAs (see below) are associated
plasm of the breast. It is classically defined as a with increased risk (1.89 times) of breast can-
biphasic lesion, meaning a proliferation of both cer compared to simple FAs if associated with
the epithelial and stromal component. proliferative diseases and when there is a rele-
vant family history of breast cancer (Dupont
et al. 1994). In general, FA-carcinoma associ-
Clinical Features ation varies between 0.1% and 0.3% in the
screened population (Dupont et al. 1994).
• Age & Sex
FA has been described at any age, but it
occurs mainly in young women of Macroscopy
reproductive age. Juvenile FA occurs at ages
between 10 and 18 years (Wechselberger FA is a firm, fibro-elastic, round–oval nodule with
et al. 2002). smooth or rarely nodular margins. The cut surface
• Site shows a relatively homogenous whitish tissue,
It may be multiple and bilateral, with no site with a delicate lobulated pattern, and in many
preference. cases, slits are also present.
© Springer Nature Switzerland AG 2020
A. Sapino, J. Kulka (eds.), Breast Pathology, Encyclopedia of Pathology,
https://doi.org/10.1007/978-3-319-62539-3
108 Fibroadenoma
typically in adolescence. They show high development (Loke et al. 2018). A recent study
stromal cellularity and pericanalicular has shown that MED12 mutations are common
growth pattern with usual hyperplasia (Song particularly among intracanalicular FAs
et al. 2014; Calcaterra et al. 2009). Apical (Volckmar et al. 2017).
snouts and gynecomastia-like micropapillary
projections and rare mitoses are features
of the luminal epithelial layer (Fig. 4 H&E). Differential Diagnosis
Genetics and genomics of breast fibroadenomas. stated in a consensus meeting in New York in
Journal of Clinical Pathology, 71, 381–387. 1985 (Hutter et al. 1986). In addition, for some
Page, D. L., & Anderson, T. J. (1987). Diagnostic
histopathology of the breast (p. 362). Edinburgh: authors this entity may be simply an “aberration of
Churchill Livingstone. normal development and involution of breast tis-
Song, B. S., Kim, E. K., Seol, H., et al. (2014). Giant sue,” in which the diagnostic definition of FCs is
juvenile fibroadenoma of the breast: A case report and more quantitative than qualitative (Tot et al.
brief literature review. Annals of Pediatric
Endocrinology & Metabolism, 19, 45–48. 2014). However, it is necessary to underline that
Volckmar, A. L., Leichsenring, J., Flechtenmacher, C., et al. the term FCs is used occasionally in a redundant
(2017). Tubular, lactating, and ductal adenomas are way, including also other benign breast entities
devoid of MED12 Exon2 mutations, and ductal adenomas such as fibroadenoma (▶ Fibroadenoma), ductal
show recurrent mutations in GNAS and the PI3K–AKT
pathway. Genes, Chromosomes & Cancer, 56, 11–17. hyperplasia (▶ Usual Ductal Hyperplasia (UDH)),
Wechselberger, G., Schoeller, T., & Piza-Katzer, H. (2002). papilloma (▶ Intraductal Papilloma), and scleros-
Juvenile fibroadenoma of the breast. Surgery, ing adenosis (▶ Sclerosing Adenosis).
132, 106–107.
Yasir, S., Gamez, R., Jenkins, S., Visscher, D. W., &
Nassar, A. (2014). Significant histological features dif-
ferentiating cellular fibroadenoma from phyllodes Clinical Features
tumor on core needle biopsies. American Journal of
Clinical Pathology, 142, 362–369. • Incidence
FCs represent the most common breast modifica-
tion. Their incidence is so high that they are
considered as a paraphysiological condition by
Fibrocystic Breast Changes some authors (Tot et al. 2014). From 58% to 61%
of breast tissue examined at autopsy show micro-
Isabella Castellano1 and Jasna Metovic2 scopic evidence of FCs (Bartow et al. 1982).
1
Department of Medical Sciences, University of • Age
Turin, Turin, Italy FCs are rarely encountered in women younger
2
Department of Oncology, University of Turin, than 21 years. Their incidence rises during the
Turin, Italy second decade of life, with a peak in the fourth
and fifth decade during premenopause period.
Family cancer history is frequently reported
Synonyms positive (Kabat et al. 2010; Shaaban
et al. 2002).
Fibrocystic condition; Fibrocystic disease; Fibro- • Sex
cystic mastopathy; Mammary dysplasia FCs are typical of female breast, because they
classically arise in the terminal ductal lobular
unit (TDLU); thus they are only occasionally
Definition seen in male breast tissue.
• Site
The term “fibrocystic changes” (FCs) covers a FCs tend to be bilateral and symmetrical. They
wide variety of clinical and pathological condi- are mainly located at the upper outer quadrants,
tions from painful breast to solid lump, multiple where glandular tissue is more abundant.
cysts, or diffuse nodularity. These abnormalities Occasionally, they may produce a clinically
are noncancerous disorders and do not represent a palpable nodule.
specific disease but rather a sign of response of the • Treatment
breast tissue to hormone levels in different phases FCs mainly occur due to relative hormone
of women life. Since FCs are the most frequent imbalance, in particular, hyperestrogenism.
breast abnormalities, the previous definition This imbalance may cause gradual dilatation
referred as “fibrocystic breast disease” was aban- of the ducts with an overgrowth of stromal
doned in favor of “fibrocystic breast changes,” as tissue and blood vessel dilatation with fluid
Fibrocystic Breast Changes 111
retention, resulting in swelling, edema, and of 15 years, the relative risk to develop a
sometimes pain in the breast. Symptoms cancer was of 1.27 (Hartmann et al. 2005).
related to FCs are generally associated with On the other hand, this risk may increase to
menstrual cycles, and they may subside with 1.62 in women with a strong family history of
menopause. Although the relationship breast cancer (Hartmann et al. 2005). More
between hormones and development of FCs recently, a study on a screening population, in
is clear, hormonal treatment with progestins which predominated postmenopausal
and tamoxifen is prescribed only for patients women, demonstrated a higher (OR = 2.23)
with severe and prolonged pain (Faiz and relative risk to develop cancer from non-
Fentiman 2000). The synthetic steroid most proliferative benign breast disease (Castells
commonly prescribed is Danazol, which has et al. 2015). The current view is that FCs per
high affinity for androgen receptor (AR), se have no impact on cancer development. F
moderate affinity for progesterone receptor However, the risk of malignant differentiation
(PR), and poor affinity for estrogen receptor may vary depending on age, family history,
(ER) (Mousavi et al. 2010). Its efficacy may and/or the association with other lesions, such
be explained by the presence of AR in the as ductal atypical hyperplasia (▶ Atypical
apocrine epithelium that normally lines cysts Ductal Hyperplasia), papillomas
of FCs. A wide variety of non- (▶ Intraductal Papilloma), radial scars
pharmacological interventions has been pro- (▶ Radial Scar), etc.
posed, such as quitting smoking and dietary
changes. In particular, lowering the intake of
Macroscopy
tea, coffee, chocolate, and fat may cause
reduction of methylxanthines that are impli-
FCs may be palpable, generally in the form of a
cated in the development of cystic formation
nodule or plaque. Radiologically, FCs can mimic
(Smith et al. 2004). In more painful cases,
other pathological conditions forming nodular
therapeutic efficacy of vitamin E on
thickening, microcalcifications, or cystic lesions.
mastalgia, with minimal or absent side
Ultrasound examination is the most accurate
effects, has been described (Pruthi et al.
method to detect cystic lesions, which appear
2010). Surgical removal of the lesion is not
commonly as well-defined anechoic nodules.
recommended.
Low-level echoes may be seen in cysts
• Outcome
containing inflammatory reactive tissue, blood
Page et al. (1978) classified FCs into
clots, and dense protein. FCs at magnetic reso-
(i) non-proliferative FCs, (ii) proliferative
nance (MR) appear as non-mass enhancement,
FCs without atypia, and (iii) proliferative
mainly due to apocrine cysts, and share the same
FCs with atypia. In this chapter only the first
features with ductal carcinoma in situ (▶ Ductal
group will be described, because the second
Carcinoma In Situ) leading sometimes to unnec-
includes papilloma, radial scar, or sclerosing
essary biopsy (Milosevic et al. 2017).
adenosis, while the third encompasses atypi-
Dense white stromal tissue with a number of
cal ductal and lobular hyperplasia. Most of
blue-greenish cysts of different size characterizes
the studies report that women with FCs are
the cut surface of breast surgical specimens (Fig. 1).
not at greater risk for subsequent carcinoma
than women in the general population (Page
et al. 1978; Dupont and Page 1985) in the
absence of positive family history, while in Microscopy
other studies, the relative risk is 1.6 (Wang
et al. 2004) with an increase to 1.95 in women FCs are characterized by histological tetrad
50 years of age and older. In one of the largest consisting of fibrosis, cysts, adenosis, and epithe-
cohorts of 9087 women affected by non- lial hyperplasia: (i) Areas of fibrosis with
proliferative FCs, with a median follow-up hyalinized stroma are prominent and cause
112 Fibrocystic Breast Changes
Fibrocystic Breast
Changes, Fig. 1 FCs in a
breast nodule: white stromal
tissue with cystic structures
is visible on cut surface
Fibrocystic Breast
Changes, Fig. 2 (H&E)
(a) Vacuum-assisted biopsy
sampling of FCs.
(b) Cluster of variably
enlarged cystic acini
containing amorphous
material and bluish
calcifications
permanent atrophy of the glandular breast tissue. entire lobule and contain amorphous secretion
(ii) Cysts are the most frequent condition, proba- rich in proteins and microcalcifications (Fig. 2).
bly due to obstruction of the duct by the exuberant Microcalcifications are present in form of calcium
fibrosis that causes dilatation of terminal ducts phosphate, appearing as basophilic and non-
and acini. Cysts may vary in size from very large birefringent round deposits forming psammoma
to microscopic. Often multiple cysts occupy an bodies easily recognizable at routine staining or
Fibrocystic Breast Changes 113
Fibrocystic Breast
Changes, Fig. 3 Single
cyst containing many
psammoma bodies (H&E)
Fibrocystic Breast
Changes,
Fig. 4 Micropapillae of
apocrine cells protrude
within the cyst (H&E)
Fibrocystic Breast Changes, Fig. 5 (IHC) (a) Nuclei of apocrine cells in a cyst are positive for AR and negative for
(b) ER, (c) PR, (d) IHC of GCDFP-15
GCDFP-15 is usually not expressed in normal aggressive potential from those at risk to become
non-apocrine breast epithelium (Fig. 5). malignant (Gromov et al. 2015).
Molecular profile of FCs was rarely analyzed. Concomitant presence of fibrosis, cysts with
High levels of ER gene transcripts (Boyd et al. apocrine metaplasia, (sclerosing) adenosis, and
1996) and ER aberrant promoter methylation have ductal hyperplasia of usual type is patho-
been described (Parrella et al. 2004). The molec- gnomonic of FCs. However, each singular histo-
ular origin of apocrine epithelium remains contro- logic pattern is subjected per se to differential
versial. Washington et al. (2000) showed loss of diagnosis with other lesions. Validated criteria
heterozygozity (LOH) in the apocrine compo- used to differentiate benign from atypical or
nents of FCs, as well as in associated carcinoma, neoplastic non-apocrine lesions are not readily
indicating possible common clonal precursors. In applicable to apocrine lesions. For example,
line with these findings, IHC screening of a vari- sclerosing adenosis with apocrine epithelium
ety of apocrine lesions from benign to malignant may simulate an apocrine-invasive carcinoma
showed similar protein signature characterized by (▶ Apocrine Carcinoma) and may require the
positive or negative expression of the following use of myoepithelial markers (such as p63,
proteins: 15-PGDH, HMGCR, S110A7, S100A9, Calponin, SMA, CK5/6). Nevertheless, as men-
and p53 (Celis et al. 2006). Thus, so far, available tioned above, pathologists should be aware that
protein markers are not sufficiently specific to benign apocrine papillary lesions, which may be
differentiate benign apocrine lesions without placed nearby to foci of sclerosing adenosis, may
Fibrocystic Breast Changes 115
lack myoepithelial layer (Cserni 2012) and should expression signature specific for benign apocrine meta-
not be confused with invasive lesions. Apocrine plasia. FEBS Letters, 580, 2935–2944.
Cserni, G. (2012). Benign apocrine papillary lesions of the
epithelium should be differentiated from atypical breast lacking or virtually lacking myoepithelial cells-
apocrine adenosis, atypical apocrine hyperplasia, potential pitfalls in diagnosing malignancy. Acta
and/or apocrine ductal carcinoma in situ. The Pathologica, Microbiologica, et Immunologica
presence of nuclear atypia (threefold nuclear Scandinavica, 120, 249–252.
Dupont, W. D., & Page, D. L. (1985). Risk factors for
enlargement with presence of large nucleolus or breast cancer in women with proliferative breast dis-
with multiple small nucleoli) seems to be the ease. New England Journal of Medicine, 312, 146–151.
major diagnostic criteria. In addition to nuclear Faiz, O., & Fentiman, I. S. (2000). Management of breast
atypia, the extent of the apocrine proliferation, pain. International Journal of Clinical Practice, 54,
228–232.
architectural features, and presence of necrosis Gromov, P., Espinoza, J. A., & Gromova, I. (2015). Molec-
have been emphasized to differentiate non-atypical ular and diagnostic features of apocrine breast lesions. F
from atypical apocrine proliferations or in situ car- Expert Review of Molecular Diagnostics, 15,
cinomas (Tavassoli and Norris 1994; O’Malley and 1011–1022.
Hartmann, L. C., Sellers, T. A., Frost, M. H., Lingle, W. L.,
Bane 2008). Finally, foci of epithelial hyperplasia Degnim, A. C., Ghosh, K., Vierkant, R. A.,
of usual type (▶ Usual Ductal Hyperplasia (UDH)) Maloney, S. D., Pankratz, V. S., Hillman, D. W., Suman,
in FCs should be differentiated from atypical V. J., Johnson, J., Blake, C., Tlsty, T., Vachon, C. M.,
hyperplasia and low-grade ductal in situ carcinoma. Melton, L. J., 3rd, & Visscher, D. W. (2005). Benign
breast disease and the risk of breast cancer. The New
Briefly, epithelial hyperplasia of usual type is a England Journal of Medicine, 353, 229–237.
non-monoclonal proliferation of cells filling Hutter, R. V. P., Albores-Saavedra, J., & Anderson, E. et al.
lumens of ducts or acini with irregular fenestrations (1986). Is ‘fibrocystic disease’ of the breast precancer-
at the periphery and expresses both low and high ous? Consensus meeting, New York, October 1985.
Archives of Pathology & Laboratory Medicine, 110,
molecular weight cytokeratins (CK), such as 121.
CK8/18 and CK5/6 and 14. In addition at differ- Kabat, G. C., Jones, J. G., Olson, N., Negassa, A.,
ence with atypical hyperplasia (▶ Atypical Ductal Duggan, C., & Ginsberg, M. (2010). A multi-center
Hyperplasia) and low-grade ductal carcinoma in prospective cohort study of benign breast disease and
risk of subsequent breast cancer. Cancer Causes and
situ (▶ Ductal Carcinoma In Situ), ER expression Control, 21, 821–828.
is heterogeneous. Milosevic, Z. C., Nadrljanski, M. M., Milovanovic, Z. M.,
Gusic, N. Z., Vucicevic, S. S., & Radulovic, O. S.
(2017). Breast dynamic contrast enhanced MRI: Fibro-
cystic changes presenting as a non-mass enhancement
Reference and Further Reading mimicking malignancy. Radiology and Oncology, 51,
130–136.
Bartow, S. A., Black, W. C., Waeckerlin, R. W., & Mousavi, S. R., Mousavi, S. M., Samsami, M., &
Mettler, F. A. (1982). Fibrocystic disease: Mahdikhah, Z. (2010). Comparison of tamoxifen with
A continuing enigma. Pathology Annual, 2, 93–111. danazol in the management of fibrocystic disease.
Boyd, M., Hildebrandt, R. H., & Bartow, S. A. (1996). International Journal of Medicine and Medical Sci-
Expression of the estrogen receptor gene in developing ences, 2, 329–331.
and adult human breast. Breast Cancer Research and O’Malley, F. P., & Bane, A. (2008). An update on apocrine
Treatment, 37, 243–251. lesions of the breast. Histopathology, 52, 3–10.
Castells, X., Domingo, L., Corominas, J. M., Page, D. L., Vander Zwaag, R., Rogers, L. W.,
Torá-Rocamora, I., Quintana, M. J., Baré, M., Williams, L. T., Walker, W. E., & Hartmann, W. H.
Vidal, C., Natal, C., Sánchez, M., Saladié, F., (1978). Relation between component parts of fibrocys-
Ferrer, J., Vernet, M., Servitja, S., Rodríguez-Arana, A., tic disease complex and breast cancer. Journal of the
Roman, M., Espinàs, J. A., & Sala, M. (2015). Breast National Cancer Institute, 61(4), 1055–1063.
cancer risk after diagnosis by screening mammography Parrella, P., Poeta, M. L., Gallo, A. P., Prencipe, M.,
of nonproliferative or proliferative benign breast dis- Scintu, M., Apicella, A., Rossiello, R., Liguoro, G.,
ease: A study from a population-based screening pro- Seripa, D., Gravina, C., Rabitti, C., Rinaldi, M.,
gram. Breast Cancer Research and Treatment, 149, Nicol, T., Tommasi, S., Paradiso, A., Schittulli, F.,
237–244. Altomare, V., & Fazio, V. M. (2004). Nonrandom dis-
Celis, J. E., Gromova, I., Gromov, P., Moreira, J. M., tribution of aberrant promoter methylation of cancer-
Cabezón, T., Friis, E., & Rank, F. (2006). Molecular related genes in sporadic breast tumors. Clinical Can-
pathology of breast apocrine carcinomas: A protein cer Research, 10, 5349–5354.
116 Fibrocystic Breast Changes
Pruthi, S., Wahner-Roedler, D. L., Torkelson, C. J., Tavassoli, F. A., & Norris, H. J. (1994). Intraductal apo-
Cha, S. S., Thicke, L. S., Hazelton, J. H., & Bauer, crine carcinoma: A clinicopathologic study of 37 cases.
B. A. (2010). Vitamin E and evening primrose oil for Modern Pathology, 7, 813–818.
management of cyclical mastalgia: A randomized pilot Tot, T., Tabàr, L., & Dean, P. B. (2014). Practical breast
study. Alternative Medicine Review, 15, 59–67. pathology. Normal breast tissue or fibrocystic change?
Radi, M. J. (1989). Calcium oxalate crystals in breast biop- (pp. 2–23). Stuttgart: Thieme Publishing Group.
sies. An overlooked form of microcalcification associ- Tramm, T., Kim, J. Y., & Tavassoli, F. A. (2011).
ated with benign breast disease. Archives of Pathology & Diminished number or complete loss of myoepithelial
Laboratory Medicine, 113(12), 1367–1369. cells associated with metaplastic and neoplastic apo-
Shaaban, A. M., Sloane, J. P., West, C. R., Moore, F. R., crine lesions of the breast. American Journal of Surgi-
Jarvis, C., & Williams, E. M. I. (2002). Histopathologic cal Pathology, 35, 202–211.
types of benign breast lesions and the risk of breast Wang, J., Costantino, J. P., Tan-Chiu, E.,
cancer: case-control study. American Journal of Surgi- Wickerham, D. L., Paik, S., & Wolmark, N. (2004).
cal Pathology, 26, 421–430. Lower-category benign breast disease and the risk of
Simard, J., Hatton, A. C., Labrie, C., Dauvois, S., invasive breast cancer. Journal of the National Cancer
Zhao, H. F., Haagensen, D. E., & Labrie, F. (1989). Institute, 96, 616–620.
Inhibitory effect of estrogens on GCDFP-15 mRNA Washington, C., Dalbègue, F., Abreo, F.,
levels and secretion in ZR-75-1 human breast cancer Taubenberger, J. K., & Lichy, J. H. (2000). Loss of
cells. Molecular Endocrinology, 3, 694–702. heterozygosity in fibrocystic change of the breast:
Smith, R. L., Pruthi, S., & Fitzpatrick, L. A. (2004). Eval- Genetic relationship between benign proliferative
uation and management of breast pain. Mayo Clinic lesions and associated carcinomas. American Journal
Proceedings, 79, 353–372. of Pathology, 157, 323–329.
G
Glycogen-Rich Clear Cell carcinoma cases when strict criteria are applied
Carcinoma (Eusebi et al. 2012). In everyday practice, the
frequency seems to be much lower.
Noëlle Weingertner and Jean-Pierre Bellocq • Age
Department of Pathology, Strasbourg University Patients are 37–78 years old (median age
Hospital, Strasbourg, France of 57).
• Treatment
Patients with GRCCC are treated according to
Definition routine guidelines for breast cancer in general.
• Outcome
Glycogen-rich clear cell carcinoma (GRCCC) Opinions differ concerning the prognosis of
occurs in many organs. GRCCC of the breast is GRCCC. Most reports suggest more aggres-
an exceptionally rare subtype of infiltrative carci- sive behavior than carcinoma NST, but Hayes
noma in which more than 90% of the neoplastic et al. contend that prognosis is not different
cells have abundant clear cytoplasm containing once tumors are matched by size, grade, and
glycogen (Eusebi et al. 2012). Extraction of the lymph node status (Hayes et al. 1995).
water-soluble glycogen during tissue processing
gives the cytoplasm an optically clear, vacuolated
appearance on tissue sections. It was first
Macroscopy
described by Hull in 1981 (Hull et al. 1981), and
The tumor size ranges from 1 to 15 cm.
since then, fewer than 200 cases have been
reported. According to Hull et al., the features of
GRCCC are very similar to the morphology of the
developing mammary gland in 13-week-old fetus. Microscopy
The clinical features of GRCCC are similar to
those of invasive carcinoma NST (▶ Invasive Histologically, GRCCC shows clusters of clear
Carcinoma NST). cells (Figs. 1 and 2 H&E) and must have more
than 90% of cells containing intracytoplasmic
Clinical Features glycogen, stained by periodic acid stain (PAS)
but not stained by diastase-periodic acid stain
• Incidence (Fig. 3 PAS staining). The tumor cells tend to
According to the 2012 WHO classification, the have sharply defined borders and polygonal con-
incidence is estimated to be 1–3% of breast tours. The cytoplasm is clear or finely granular.
© Springer Nature Switzerland AG 2020
A. Sapino, J. Kulka (eds.), Breast Pathology, Encyclopedia of Pathology,
https://doi.org/10.1007/978-3-319-62539-3
118 Glycogen-Rich Clear Cell Carcinoma
Molecular Features
M. J. van de Vijver (Eds.), WHO classification of elderly ladies aged more than 70 years (Torous
tumours of the breast (pp. 74–75). Lyon: IARC. et al. 2017).
Hayes, M. M. M., Seidman, J. D., & Ashton, M. A. (1995).
Glycogen rich clear cell carcinoma of the breast. • Sex
A clinicopathologic study of 21 cases. American Breast GCT affects more frequently female
Journal of Surgical Pathology, 19, 904–911. patients, but rare cases have been reported in
Hull, M. T., Priest, J. B., Broadie, T. A., Ransburg, R. C., & males (Damiani et al. 1992). African-American
Mc Carthy, L. J. (1981). Glycogen-rich clear cell car-
cinoma of the breast: A light and electron microscopic women are more frequently affected than Cau-
study. Cancer, 48, 2003–2009. casian women (Torous et al. 2017).
Kim, Z. E., Koo, J. S., & Jung, W. H. (2012). • Site
Immunophenotypes of glycogen rich clear cell carci- GCT is usually located in the upper-internal
noma. Yonsei Medical Journal, 53(6), 1142–1146.
Markopoulos, C., Mantas, D., Philipidis, T., Kouskos, E., quadrant of the breast, even if all the breast
Antonopoulou, Z., Hatzinikolaou, M. L., & Gogas, H. quadrants can be affected, comprising the axil-
(2008). Glycogen-rich clear cell carcinoma of the lary tail (Damiani et al. 1992, Torous et al.
breast. World Journal of Surgical Oncology, 6, 44. 2017). GCT of the breast can be located both G
in the deep parenchyma and in the subcutane-
ous tissue. Cases deeply located and infiltrating
the pectoralis muscle are on record (Damiani
Granular Cell Tumor et al. 1992). Cases arising in the retro-areolar
region can cause nipple retraction. Subcutane-
Maria P. Foschini and Luca Morandi ous lesions can cause skin retraction.
Department of Biomedical and Neuromotor • Treatment
Sciences (DIBINEM), Unit of Anatomic GCT is more frequently benign; therefore it is
Pathology at Bellaria Hospital, University of usually treated with tumor resection, with clear
Bologna, Bologna, Italy margins. Sentinel lymph node dissection is not
needed. Correct treatment largely depends on
correct preoperative diagnosis. As GCT fre-
Synonyms quently mimics malignancy both on clinical
and histological grounds (Torous et al. 2017),
Abrikossoff’s tumor, Granular cell myoblastoma sometimes an erroneous aggressive treatment
is performed.
Aggressive treatment is needed in those rare
Definition cases showing malignant features.
• Outcome
Granular cell tumor (GCT) is a rare tumor, of Most of breast GCT follow a benign clinical
neurogenic derivation, composed of cells with course. Recurrences can appear when initial
pale and granular cytoplasm. surgical excision has not achieved free
margins.
Clinical Features
Macroscopy
• Incidence
GCT usually arises in the tongue, but all the On macroscopy GCT presents as a firm nodule,
organs can be affected. About 6% of GCT gray-whitish in color. Margins are more fre-
arises in the breast (Damiani et al. 1992; quently invasive, therefore simulating a breast
Sanguinetti et al. 2016; Rexeena et al. 2015). carcinoma. Rare cases with well-defined margins,
• Age simulating fibroadenoma have been described
Breast GCT most frequently presents in middle (Damiani et al. 1992). Size varies, but it is usually
aged women, but age can vary from puberty to smaller than 3 cm.
120 Granular Cell Tumor
Microscopy Immunophenotype
On histology breast GCT is similar to GCT of other GCT cells are typically strongly and diffusely immu-
body sites. It is characterized by a proliferation of noreactive with S-100 protein. In addition, CD68,
cells with wide, pale eosinophilic granular cyto- carcino-embryonic antigen (CEA), vimentin, and
plasm (Figs. 1 and 2). Nucleus is centrally located, calretinin have been described as positive
small, with delicate chromatin, and inconspicuous (Sanguinetti et al. 2016; Rexeena et al. 2015). Epithe-
nucleolus (Fig. 3a, b). Cytoplasmic granules are lial markers as low and high molecular weight
positive with PAS after diastase reaction. No in cytokeratins, epithelial membrane antigen, and apo-
situ carcinoma or lymph-vascular invasion is seen. crine marker GCDFP-15 are negative. Estrogen, Pro-
GCT has invasive margins, therefore infiltrat- gesterone and Androgen receptors are negative as
ing the surrounding breast and fat tissue. well as HER2 does not show features of amplification.
Mitoses, cellular atypia, and necrosis are usually Proliferative index, as evaluated with Ki67, is low.
absent, as most of the GCT are benign. These latter On electron microscopy the GCT cells show
features can be encountered in those rare cases of numerous membrane-bound vesicles, mostly
malignant GCT (Sanguinetti et al. 2016; Rexeena filled with flocculent and solid electron-dense
et al. 2015). material (Damiani et al. 1992).
• Incidence
IGM is rare, encountered in less than 0.5% of
Granulomatous Mastitis breast samples (Fazzio et al. 2016).
• Age
Noëlle Weingertner and Jean-Pierre Bellocq IGM mainly affects childbearing age women,
Department of Pathology, Strasbourg University most often in postpartum or breastfeeding
Hospital, Strasbourg, France mothers (Fazzio et al. 2016). The age at
Granulomatous Mastitis 123
Granulomatous Mastitis,
Fig. 1 Idiopathic
granulomatous mastitis –
mammographic features.
Mammography shows
nodular opacities (stars).
Courtesy of Dr Sébastien
Molière, Department of
Radiology, Strasbourg
University Hospital
diagnosis is generally 20–50 years (age range, (Lai et al. 2005). Treatment of IGM
11–83 years) (Seo et al. 2012). is not standardized, but corticosteroids and
• Sex methotrexate are generally used, as primary
Mainly females. Males may rarely be affected treatment or after surgery to prevent recur-
(Seo et al. 2012). rences. Steroids can be used before surgery to
• Site shrink the mass (Cheng et al. 2015). Topical
IGM equally affects the right or left breast, corticosteroids may be effective (Tahmasebi
bilaterality being exceptionally rare. Oran et al. 2016) and prevent systemic adverse
et al. found the most frequent site of the lesion effect. Antibiotics are sometimes used in com-
was the upper outer quadrant, followed by the bination with these treatments (D’Alfonso
inferior outer quadrant, the areola, the inferior et al. 2015). Surgery is controversial due to
inner quadrant, and the upper inner quadrant poor wound healing, fistula formation, and dis-
(Oran et al. 2013). ease recurrence (Tahmasebi et al. 2016), with
• Treatment reported recurrence rates after surgery as high
Spontaneous resolution occurs in 50% as 50% (Pereira et al. 2012). It is usually
of patients after a mean duration of 14.5 months reserved for refractory or aggressive cases. It
124 Granulomatous Mastitis
is recommended that implants not be used if Therefore, tissue core biopsy with histologic
breast reconstruction is undertaken (Pereira evaluation is the most reliable diagnostic method.
et al. 2012), because the implant may act as a Histologic evaluation demonstrates non-
foreign body and trigger further granulomatous necrotizing granuloma, with an infiltrate of multi-
inflammation. Clinical follow-up is also nucleated giant cells, epithelioid histiocytes,
recommended. plasma cells, and lymphocytes. A neutrophilic
• Outcome infiltrate may also occur with formation of micro-
Variable recurrence rates (5.5–50%) are abscesses (Pereira et al. 2012). A histological
reported in the literature (Seo et al. 2012) pattern of neutrophil aggregates surrounding
with complications including sinuses requiring microcystic spaces associated with the granulo-
draining and abscess formation. The patients matous inflammation has been called “cystic neu-
often have to undergo multiple surgical pro- trophilic granulomatous mastitis”. This subset of
cedures. Upon resolution, common sequelae granulomatous mastitis is reported to be linked to
include scarring, retraction of skin and nipple, Corynebacterium species infection in some cases
and shrinkage of breast tissue (Pereira et al. (Gautham et al. 2019) (Figs. 2 and 3). Eosinophils
2012). Therefore, IGM is disfiguring and pain- can also be seen. The inflammation is centered on
ful but does not pose a threat to general health. lobules (Fig. 4), but extensive inflammation may
There is no evidence that this condition obliterate this feature.
increases the incidence of breast carcinoma The diagnosis of IGM should be made after
(Pereira et al. 2012). exclusion of all other possibilities and specific
etiological features (Differential Diagnosis).
Necrosis (apart from microabscesses occasion-
Macroscopy ally), foreign body material, infective agents, or
vasculitis should not be present. Proper evaluation
The most common clinical/macroscopic presenta- of the specimen requires special stains to search
tion is one or multiple firm, unilateral, and palpa- for mycobacteria and a variety of other organisms
ble breast mass(es), often associated with and also requires that samples for microbial exam
inflammation of the overlying skin, with “peau and cultures are obtained in specific conditions.
d’orange” appearance being possible. The mass
can vary in size from 1 to 10 cm (Pereira et al.
2012) and is gray-tan and irregular. The lesion is Immunophenotype
painful in approximately 25% of cases (Korkut
et al. 2015). Skin ulceration, nipple discharge There are no particular immunohistochemical
or nipple retraction, abscesses, sinus formation, features in IGM. A pankeratin immunostaining
fistulas, and axillary lymphadenopathy may be may be helpful to exclude underlying breast
seen, this latter in approximately 15% of patients carcinoma.
(Seo et al. 2012; Korkut et al. 2015). Bilateral
involvement has been described. Rare patients
are asymptomatic, with an abnormality detected Molecular Features
on mammography (Fazzio et al. 2016).
Mammography may show nodular opacities To date, no particular molecular feature has been
(Fig. 1). described for IGM.
IGM can mimic breast malignancy cytologically One of the most important clinical differential
(Al-Khaffaf et al. 2008; Oran et al. 2013). diagnoses of IGM is breast carcinoma. Indeed, in
Granulomatous Mastitis 125
Granulomatous Mastitis,
Fig. 2 Cystic neutrophilic
granulomatous mastitis –
microscopic features
(microbiospy, H&E 10):
inflammatory infiltrate with
epithelioid cells,
macrophages and
neutrophil leucocytes
forming non-necrotizing
granulomas (stars). This
type of granulomatous
mastitis has association
with Corynebacteria
Granulomatous Mastitis, Fig. 3 Cystic neutrophilic Granulomatous Mastitis, Fig. 4 Idiopathic granuloma-
granulomatous mastitis – microscopic features tous mastitis – microscopic features (microbiospy,
(microbiospy, H&E 20). The inflammatory infiltrate is H&E 20): Multinucleated giant-cells (arrow)
in contact with a lobule and shows a cyst-like space
surrounded by neutrophil leucocytes
Non-tuberculous Infectious Granulomatous
Mastitis
more than 50% of cases, the initial diagnosis was Infection is the first possibility to consider when
suspected to be breast cancer (Seo et al. 2012). faced with an inflammatory granulomatous
Furthermore, a granulomatous inflammation may infiltrate of the breast. Infectious granulomatous
occasionally accompany an infiltrating carci- mastitis can be due to mycobacteriosis,
noma, and this situation must be ruled out before bartonellosis (cat scratch disease), brucellosis,
a diagnosis of granulomatous mastitis is made, corynebacteriosis, and salmonella typhi, as well
particularly in postmenopausal women. as fungal agents such as histoplasma and parasites
Once malignancy is excluded, pathologists such as filariasis. These granulomatous inflamma-
have to think about other causes of mammary tions are often necrotizing. Therefore, special
granulomatous inflammation before a diagnosis stains are essential in considering granulomatous
of IGM can be made. mastitis, including PAS, Grocott, Ziehl-Neelsen,
126 Granulomatous Mastitis
and Gram. When possible, some tissue should be well-circumscribed, non-necrotizing granulomas
sent to bacteriology, mycology, and parasitology with epithelioid histiocytes and multinucleated
laboratories. giant cells, which may contain asteroid or
Schaumann bodies.
Tuberculosis of the Breast Mammary localization of vasculitis is rare,
Tuberculosis of the breast is rare in Western coun- with approximately 40 cases described in the
tries, accounting for less than 1% of tuberculous literature. Suggestive clinical context is often
cases, and is most frequent in endemic zones. present. Nevertheless, patients often present a
Its incidence can increase in immunosuppressed breast mass, and biopsies of these lesions are
patients. Patients are mostly female, 20 to 50 years therefore taken to exclude malignancy. With
old. Breast tuberculosis is most frequently sec- microbiopsies, when the vasculitis is not known,
ondary to a generalized tuberculous infection the diagnosis is very difficult to ascertain and can
(Korkut et al. 2015). It may infrequently be often only be suggested. Therefore, pathologists
primary, due to main galactophorous duct lesion, have to analyze the vessels carefully in cases of
linked to a trauma during pregnancy or lactation. granulomatous mastitis.
Patients present in general with a breast mass, or Granulomatosis with polyangiitis mainly
an abscess, sometimes associated with satellite affects the respiratory tract and the kidneys, but
axillary lymphadenopathy. Histological aspects rarely the breast. Patients are mostly females, aged
are similar to its appearance in other organs, 40 to 50. In general, breast lesions are accompa-
with an inflammatory infiltrate of epithelioid his- nied by respiratory tract and kidney involvement,
tiocytes, with Langhans-type multinucleated giant associated with biological inflammatory syn-
cells accompanied by lymphocytes and occasion- drome and ANCA antibodies. Breast lesions pre-
ally by neutrophils. Caseous necrosis is character- sent as one or multiple breast lump(s), uni- or
istic and is the main pathologic aspect bilaterally, measuring 2–6 centimeters (Pereira
differentiating IGM from tuberculous mastitis et al. 2012). The skin may be retracted, ulcerated,
(Pereira et al. 2012). As in other organs, Ziehl- or with “peau d’orange” effect, and axillary
Neelsen staining rarely identifies acid-fast bacilli. lymphadenopathy may be present.
Histologically, non-well-circumscribed epithe-
Granulomatous Mastitis Associated with lioid and giant-cell granulomas are associated
Systemic Diseases with large geographic zones of “blue necrosis”
Granulomatous mastitis can be part of systemic (due to the large amounts of basophilic nuclear
diseases such as sarcoidosis, granulomatosis with debris) and vasculitis of small- and medium-sized
polyangiitis (Wegener’s granulomatosis), giant- vessels. Eosinophils may be encountered.
cell arteritis (Horton disease), rheumatoid arthri- Giant-cell arteritis (Horton’s disease) has been
tis, necrobiotic xanthogranuloma, etc. Extranodal described in all branches of the subclavicular
Rosai-Dorfman disease of the breast has also been artery. The breast is vascularized by the internal
reported (Chen et al. 2016) and may appear as a mammary artery and lateral thoracic artery, which
granulomatous process. are branches of the subclavicular artery.
The histological features of systemic disease- Patients are 50 to 80 years old. It presents as
associated granulomatous mastitis are quite clas- one or several, frequently painful breast lumps
sic, similar to the histology seen in other organs. (Pereira et al. 2012). Bilaterality is encountered
Sarcoidosis-related mastitis is rare (less than in approximately 50% of cases. General symp-
1% of sarcoidosis) and, in the large majority of toms are often present, with arthralgias, anorexia,
cases, occurs in the context of generalized disease asthenia, fever, myalgia, biological inflammatory
with pulmonary, cutaneous, lymph node, articular, syndrome, and headache. Histologically, an
and ocular manifestations (Pereira et al. 2012). inflammatory infiltrate centered on small- and
Patients present with one or several breast medium-sized arterial vessels is composed of
masses. Histology is characterized by small, macrophages and giant cells with lymphocytes
Granulomatous Mastitis 127
and neutrophils. The inflammatory infiltrate is often refracting under polarized light) or second-
segmental and focal, with fragmentation of ary to macrobiopsy or to breast implant (Oran
the internal elastic lamina. et al. 2013).
Necrobiotic xanthogranuloma displays degen- Breast implants are in general composed of
eration of collagen fibers and foam cells besides silicone. A fibrous capsule develops surrounding
the granulomas (Cheng et al. 2015). Benign the implant. The pocket of the implant can fissure
monoclonal gammopathy or myeloma is associ- with a clinical aspect of skin fixation and retrac-
ated with 80% of cases. tion. Axillary lymphadenopathy may be encoun-
tered. Periprosthetic capsules may be addressed
Granulomatous Mastitis Associated with to the pathologists. The inflammatory infiltrate
Trauma, Surgery, or Foreign Body Material is composed mainly of macrophages, with a
Cytosteatonecrosis (▶ Breast Fat Necrosis) is in “pseudo-synovial” palisaded disposition on the
general secondary to a trauma or a surgery internal side of the capsule. Multinucleated for-
(Pereira et al. 2012), but may be spontaneous, eign body giant cell may be seen sometimes asso- G
especially in older women. Clinical presentation ciated with optically clear holes corresponding to
may be worrisome, particularly if the trauma is not silicone, which is non-refracting. Lymphocytes
known or forgotten. The forms of trauma are may be associated. Pathologists have to examine
many: traumas of everyday life, massage, pressure these specimens carefully, to exclude a carcinoma
related to mammography, cyst aspiration, core and a breast implant-associated lymphoma
biopsy, mammoplasty, implant removal, radiation (▶ Breast Implant-Associated Malignant Lym-
therapy, etc. The lesion may persist several phoma), the latter being recently described
months or years after the trauma. Women with (Laurent et al. 2016).
large (adipous) breasts are particularly at risk. Rarely, nodular deposition of amyloid can
Fat necrosis presents as a breast mass, sometimes cause granulomatous reaction in the breast,
with skin retraction. Cystic zones may be seen on which will be diagnosed with special stains
ultrasonography, and there may be calcifications (Cheng et al. 2015).
on mammography (D’Alfonso et al. 2015).
The macroscopic aspect is characteristic, with Other Conditions
adipose tissue appearing pale yellow. Fibrosis, Other conditions may be associated with a granu-
pseudocysts, and calcifications are seen in lomatous inflammation of the breast tissue, such
old lesions. Histologically, an inflammatory infil- as subareolar abscesses, duct ectasia, or
trate is centered on adipose tissue, composed of hidradenitis suppurativa.
foamy macrophages, multinucleated giant cells, Subareolar abscess (SA) is also known as
lymphocytes, and plasma cells. Hemorrhage may Zuska’s disease, mastitis obliterans, or lactiferous
be seen in the acute phase. Immunohistochemistry fistula (Pereira et al. 2012). It results from obstruc-
can be helpful to rule out a carcinoma (particularly tion, distension, inflammation, and rupture of the
a lipid-rich carcinoma or a carcinoma with lactiferous ducts. SA occurs when the normal
“histiocytoid” morphology), which would be pos- epithelium of the distal ducts undergoes squa-
itive in fat necrosis for histiocytic markers such mous metaplasia and keratin obstructs the duct
as CD163 and CD68 and negative for pankeratin lumen. The duct may then rupture, and release of
(D’Alfonso et al. 2015). Lipid pseudocysts, calci- keratin into the breast parenchyma leads to an
fications, and ossifications can be seen in old inflammatory response. Granulomatous inflam-
lesions. When cytosteatonecrosis is secondary to mation is a common histologic feature in the
radiotherapy, cytological atypia in fibroblasts, chronic stage of SA. Nipple discharge is common,
endothelial cells, or mammary epithelial cells usually non-bloody, whereas it is not a common
may be seen. feature in IGM. Smoking is a major risk factor for
Foreign body granulomas may be encountered SA, in contrast to IGM, which is often associated
and associated with surgical thread (which is with pregnancy and lactation. The SA abscess
128 Granulomatous Mastitis
involves the main lactiferous ducts and occurs just Cheng, L., Reddy, V., Solmos, G., Watkins, L.,
below the nipple and areola, whereas IGM involves Cimbaluk, D., Bitterman, P., Ghai, R., & Gattuso,
P. (2015). Mastitis, a radiographic, clinical, and Histo-
the breast lobules at the periphery of the breast. pathologic review. The Breast Journal, 21, 403–409.
Fistulas from SA involve the nipple and areola, D’Alfonso, T. M., Ginter, P. S., & Shin, S. J. (2015).
whereas fistulas from IGM can occur in any part A review of inflammatory processes of the breast
of the breast. Histologically, keratinizing squamous with a focus on diagnosis in Core biopsy samples.
Journal of Pathology and Translational Medicine,
epithelium can be observed in SA, and the inflam- 49, 279–287.
mation is localized around the ducts. These features Fazzio, R. T., Shah, S. S., Sandhu, N. P., &
are not observed in IGM. Glazebrook, K. N. (2016). Idiopathic granulomatous
In duct ectasia, subareolar ducts are dilated, mastitis: Imaging update and review. Insights Imaging,
7, 531–539.
full of milk-like or butter-like secretion, Gautham, I., Radford, D. M., Kovacs, C. S., Calhoun, B.
surrounded by fibrosis of variable intensity, and C., Procop, G. W., Shepardson, L. B., Dawson, A. E.,
accompanied by a chronic inflammatory infiltrate Downs-Kelly, E. P., Zhang, G. X., Al-Hilli, Z., Fan-
also of variable intensity that may sometimes ning, A. A., Wilson, D.A., Sturgis, C. D. (2019) Cystic
neutrophilic granulomatous mastitis: The Cleveland
become granulomatous (periductal mastitis, Clinic experience with diagnosis and management.
comedomastitis). The Breast Journal, 25, 80–85.
Hidradenitis suppurativa can involve the Kessler, E., & Wolloch, Y. (1972). Granulomatous masti-
breast, but the breast localization is usually asso- tis: A report of seven cases. American Journal of Clin-
ical Pathology, 58, 642–646.
ciated with axillary and inguinal involvement Korkut, E., Akcay, M. N., Karadeniz, E., Subasi, I. D., &
(Pereira et al. 2012). Granulomatous inflamma- Gursan, N. (2015). Granulomatous mastitis: A ten-year
tion is present in the chronic stages. experience at a university hospital. The Eurasian Jour-
In conclusion, when dealing with a granulo- nal of Medicine, 47, 165–173.
Lai, E. C., Chan, W. C., Ma, T. K., Tang, A. P., Poon, C. S.,
matous inflammation in breast specimens, pathol- & Leong, H. T. (2005). The role of conservative treat-
ogists have to: ment in granulomatous mastitis. The Breast Journal.
11, 454–456.
• Rule out an underlying breast carcinoma Laurent, C., Delas, A., Gaulard, P., Haioun, C.,
Moreau, A., Xerri, L., Traverse-Glehen, A., Rousset,
• Search for foreign body material T., Quintin-Roue, I., Petrella, T., Emile, J. F., Amara,
• Think about a possible infection, and therefore N., Rochaix, P., Chenard-Neu, M. P., Tasei, A. M.,
search for necrosis and use special stains or, Menet, E., Chomarat, H., Costes, V., Andrac-Meyer,
more especially, microbiologic analyses on L., Michiels, J. F., Chassagne-Clement, C.,
De Leval, L., Brousset, P., Delsol, G., & Lamant,
fresh tissue if possible L. (2016). Breast implant-associated anaplastic large
• Analyze vessels carefully, searching for cell lymphoma: Two distinct clinicopathological vari-
vasculitis ants with different outcomes. Annals of Oncology, 27,
• Of course be aware of the clinical context 306–314.
Oran, E. S., Gürdal, S. Ö., Yankol, Y., Öznur, M., Calay, Z.,
Tunaci, M., & Soybir, G. R. (2013). Management of
idiopathic granulomatous mastitis diagnosed by core
biopsy: A retrospective multicenter study. The Breast
References and Further Reading Journal, 19, 411–418.
Pereira, F. A., Mudgil, A. V., Macias, E. S., &
Al-Khaffaf, B., Knox, F., & Bundred, N. J. (2008). Karsif, K. (2012). Idiopathic granulomatous lobular
Idiopathic granulomatous mastitis: A 25-years experi- mastitis. International Journal of Dermatology, 51,
ence. Journal of the American College of Surgeons, 142–151.
206, 269–273. Seo, H. R., Na, K. Y., Yim, H. E., Kim, T. H., Kang, D. K.,
Bani-Hani, K. E., Yaghan, R. J., Matalka, I. I., & Oh, K. K., Kang, S. Y., An, Y. S., Chun, M., Kim, W.,
Shatnawi, N. J. (2004). Idiopathic granulomatous mas- Park, R. W., Jung, Y. S., & Kim, K. S. (2012).
titis: Time to avoid unnecessary mastectomies. The Differential diagnosis in idiopathic granulomatous
Breast Journal, 10, 318–322. mastitis and tuberculous mastitis. Journal of Breast
Chen, Y. P., Jiang, X. N., Lu, J. P., Zhang, H., Li, X. Q., & Cancer, 15, 111–118.
Cheng, G. (2016). Clinicopathologic analysis of extra- Tahmasebi, S., Karami, M. Y., & Maalhagh, M. (2016).
nodal Rosai-Dorfman disease of breast: a report of 12 Granulomatous mastitis: Time to introduce new
cases. Zhonghua Bing Li Xue Za Zhi. 45, 556–60. weapons. World Journal of Surgery, 40, 2827–2828.
Gynecomastia 129
Gynecomastia, Fig. 1 (H&E) Low-power hematoxylin intermediate type shows increasing concentric fibrosis
and eosin stains illustrate the three types of gynecomastia. around the individual ducts which start to merge (b). The
The florid type shows edematous increased periductal stro- fibrous type shows confluent fibrous collagen-rich stroma
mal cellularity concentrically forming around the rudimen- and less prominent epithelial proliferation (c)
tary ducts that show ductal epithelial hyperplasia (a). The
Gynecomastia 131
Gynecomastia, Fig. 2 Medium-power hematoxylin and Gynecomastia, Fig. 3 High-power hematoxylin and
eosin stain illustrates pseudoangiomatous stromal hyper- eosin stain illustrates gynecomastoid hyperplasia. The
plasia (PASH). The inter-anastomosing clefts are angulated duct is lined by several layers that tuft into the lumina;
and slit-like, lined by myofibroblasts and surrounded by note the pyramid shape of the tufts (H&E)
wavy, acellular collagenous stroma (H&E)
Atypical ductal hyperplasia (ADH) G
suggests, lined by myofibroblasts and not by (▶ Atypical Ductal Hyperplasia) and ductal carci-
endothelium, thereby positive for stromal markers noma in situ (DCIS) (▶ Ductal Carcinoma In
like vimentin only and not for (lympho)vascular Situ), two known clonal precursor lesions for
markers. The spaces are angulated and slit-like breast cancer in females, have rarely been
and surrounded by wavy, acellular collagenous reported in male breasts with gynecomastia
stroma. The myofibroblasts are spindle to ovoid (Hamady et al. 2005; Doebar et al. 2017). DCIS
but are not atypical (Fig. 2). Multinucleated giant is the most commonly observed precursor lesion
cells can be seen. in male breast cancer, and its presence seems to be
The rudimentary ducts in gynecomastia are associated with favorable outcome (Doebar et al.
lined by at least three cell layers: an outer layer 2017). It usually concerns the papillary variant or
of myoepithelial cells and two layers of epithelial low-grade DCIS (▶ Intraductal Papillary Carci-
cells. The myoepithelial cells may have clear noma), although all the subtypes described in
cytoplasm. The intermediate luminal layer con- women can occur in men. DCIS is mainly associ-
sists of cuboidal to columnar vertically orientated ated with invasive breast cancer.
cells, with regular oval nuclei and occasional The presence of lobular carcinoma in situ
small nucleoli. The cytoplasm is eosinophilic (▶ Lobular In Situ Neoplasia) has also been
with indistinct cell borders, and these cells often described but is very rare, in line with the rarity
show luminal tufts. The inner luminal layer com- of invasive lobular cancer (▶ Invasive Lobular
prised slightly smaller and more horizontally ori- Carcinoma) in men (Doebar et al. 2005).
entated cells, most often arranged in a single layer,
with regular to slightly irregular small nuclei and
sporadic small nucleoli (Kornegoor et al. 2012b). Gynecomastia and Invasive Cancer
More layers may be present, resembling the
sloppy ductal hyperplasia of usual type of the The role of gynecomastia in the development of
female breast, with flat peripheral layers or a mildly male breast cancer (▶ Male Breast Cancer)
cribriform architecture with irregular lumina with remains uncertain. Gynecomastia is often seen
regard to spacing, shape, and size, or a central sea alongside invasive breast cancer, but gynecomas-
of epithelium with slit-like peripheral residual tia is also frequently encountered in healthy men
lumina. Some tufting into the lumina may be pre- (Braunstein 1993; Fentiman et al. 2006). How-
sent, and micropapillae showing a pyramid shape ever, other studies found a significant correlation
instead of the rod shape of micropapillae of clonal between gynecomastia and male breast cancer
female breast epithelial proliferations usually (Brinton et al. 2010).
denoted gynecomastoid hyperplasia (Fig. 3). It has been suggested that there is a difference
Microcalcifications and secretions are very rare. in carcinogenesis between male and female breast
132 Gynecomastia
cancer. One of the largest series of male breast SMA+ and CD34- staining are not specific to malig-
cancers demonstrates that luminal type A is by far nancy because such findings are also encountered in
the most common breast cancer subtype in males. reactive fibrosis (Kalekou et al. 2005).
Luminal type B breast cancer is less common and In most cases moderate to strong Bcl-2
represents a subgroup of ER-positive tumors with staining is seen in stromal cells, especially in
highly malignant phenotype. HER2-driven, basal- florid and intermediate types. p21 shows spo-
like, and unclassifiable triple-negative breast can- radic positive cells in the surrounding stroma,
cers in men seem to be very rare. This distribution and p53 shows wild-type expression. Prominent
of breast cancers subtypes in men is different stromal staining is often seen for EGFR, espe-
compared with females (Kornegoor 2012a). cially in the presence of PASH (Kornegoor et al.
2012a). PASH areas do not stain with (lympho)
vascular makers like D2–40 (podoplanin),
Immunophenotype LYVE-1, and CD31.
Epithelium
The normal ductal male breast epithelial cells are Molecular Features
very often ER and Bcl-2 positive (>69%), and PR
and AR expression is also common (>39%). As Until now no chromosomal abnormalities have been
described above, gynecomastia appears to show a detected in cases of gynecomastia with no other
consistent three-layered pattern. The immunohis- concomitant breast disease. For male DCIS and
tochemical pattern is distinctive as the outer invasive cancer, however, a clonal relationship has
myoepithelial layer shows expression of CK5, been described, based on similar copy number pro-
CK14, and p63. The two epithelial cell layers file for 21 out of 22 interrogated breast cancer-related
differ from each other. The intermediate luminal genes, underlining that DCIS is a true precursor for
layer, consisting of vertically oriented cuboidal to male invasive breast cancer (▶ Male Breast Cancer)
columnar cells, is hormone receptor positive and (Vermeulen et al. 2017). Doebar et al. showed iden-
expresses Bcl-2 and CyclinD1. The inner luminal tical genomic aberrations, including PIK3CA,
layer is composed of smaller cells expressing CK5 GATA3, TP53, and MAP2K4 mutations, based on
and often CK14 but are usually negative for hor- next-generation sequencing (Doebar et al. 2017). For
mone receptors and Bcl-2. All epithelial cells male breast cancer it has been suggested that the
show strong staining with CK7 and E-cadherin. X chromosome gain plays a role in the neoplastic
HER2 staining is completely negative in the epi- transformation of male breast epithelial cells and that
thelium, and BRST2 and EGFR are also almost the gain of chromosome 5, loss of the
always negative. Only wild-type p53 staining is Y chromosome, loss of chromosome 17, and del
seen in ductal epithelium (Kornegoor (18)(q21) are described as nonrandom abnormalities
et al. 2012b). (Di Oto et al. 2015).
Male DCIS is consistently ER positive and
CK5/CK14 negative. Ki67-positive cells are
rarely seen in the intermediate and inner luminal Differential Diagnoses
layers (Kornegoor et al. 2012a).
The main most important differential diagnosis is to
Stroma discriminate gynecomastia from male breast cancer
The immunophenotype of periductal connective tis- (▶ Male Breast Cancer), especially if it is a unilat-
sue stroma in gynecomastia appears to parallel the eral, fixed palpable mass with nipple discharge
phenotype of normal breast stroma. In male breast (Johnson and Murad 2009). Mammography and
carcinoma the stromal cell immunophenotype is high-resolution ultrasound can be helpful to distin-
similar to that of its female counterpart showing guish between these breast lesions (Munoz
myofibroblastic differentiation. However, alpha- Carrasco et al. 2010). Also pseudogynecomastia
Gynecomastia 133
and other benign lesions, such as lipoma, hema- Johnson, R. E., & Murad, M. H. (2009). Gynecomastia:
toma, fat necrosis (▶ Breast Fat Necrosis), dermoid Pathophysiology, evaluation, and management. Mayo
Clinic Proceedings, 84, 1010–1015.
cyst, sebaceous cyst, or lymphoplasmacytic inflam- Kalekou, H., Kostopoulos, I., Milias, S., & Papadimitriou,
mation, should be considered. C. S. (2005). Comparative study of CD34, alpha-SMA
and h-caldesmon expression in the stroma of
gynaecomastia and male breast carcinoma. Histopa-
thology, 47, 74–81.
References Kanhai, R. C., Hage, J. J., van Diest, P. J., Bloemena, E., &
Mulder, J. W. (2000). Short-term and long-term histo-
Blau, M., Hazani, R., & Hekmat, D. (2016). Anatomy of logic effects of castration and estrogen treatment on
the gynecomastia tissue and its clinical significance. breast tissue of 14 male-to-female transsexuals in com-
Plastic and Reconstructive Surgery – Global Open, 4, parison with two chemically castrated men. The Amer-
e854. ican Journal of Surgical Pathology, 24, 74–80.
Braunstein, G. D. (1993). Gynecomastia. New England Kornegoor, R., Verschuur-Maes, A. H., Buerger, H., & van
Journal of Medicine, 328, 490–495. Diest, P. J. (2012a). The 3-layered ductal epithelium in
Brinton, L. A., Carreon, J. D., Gierach, G. L., McGlynn,
K. A., & Gridley, G. (2010). Etiologic factors for male
gynecomastia. The American Journal of Surgical G
Pathology, 36, 762–768.
breast cancer in the U.S. veterans affairs medical care Kornegoor, R., Verschuur-Maes, A. H., Buerger, H.,
system database. Breast Cancer Research and Treat- Hogenes, M. C., de Bruin, P. C., Oudejans, J. J., van
ment, 119, 185–192. der Groep, P., Hinrichs, B., & van Diest, P. J. (2012b).
Di Oto, E., Monti, V., Cucchi, M. C., Masetti, R., Molecular subtyping of male breast cancer by immu-
Varga, Z., & Foschini, M. P. (2015). X chromosome nohistochemistry. Modern Pathology, 25, 398–404.
gain in male breast cancer. Human Pathology, 46, Milanezi, M. F., Saggioro, F. P., Zanati, S. G., Bazan, R., &
1908–1912. Schmitt, F. C. (1998). Pseudoangiomatous hyperplasia
Doebar, S. C., Doebar, S. C., Slaets, L., Cardoso, F., of mammary stroma associated with gynaecomastia.
Giordano, S. H., Bartlett, J. M., Tryfonidis, K., Journal of Clinical Pathology, 51, 204–206.
Dijkstra, N. H., Schröder, C. P., van Asperen, C. J., Munoz Carrasco, R., Alvarez Benito, M., Muñoz Gomariz,
Linderholm, B., Benstead, K., Dinjens, W. N., van E., Raya Povedano, J. L., & Martínez Paredes,
Marion, R., van Diest, P. J., Hamady, Z. Z., Carder, M. (2010). Mammography and ultrasound in the eval-
P. J., & Brennan, T. G. (2005). Atypical ductal hyper- uation of male breast disease. European Radiology, 20,
plasia in male breast tissue with gynaecomastia. Histo- 2797–2805.
pathology, 47, 111–112. Narula, H. S., & Carlson, H. E. (2014). Gynaecomastia –
Doebar, S. C., Slaets, L., Cardoso, F., Giordano, S. H., Pathophysiology, diagnosis and treatment. Nature
Bartlett, J. M., Tryfonidis, K., Dijkstra, N. H., Reviews Endocrinology, 10, 684–698.
Schröder, C. P., van Asperen, C. J., Linderholm, B., Sansone, A., Romanelli, F., Sansone, M., Lenzi, A., & Di
Benstead, K., Dinjens, W. N., van Marion, R., van Luigi, L. (2017). Gynecomastia and hormones. Endo-
Diest, P. J., Martens, J. W., & van Deurzen, C. H. crine, 55, 37–44.
(2017). Male breast cancer precursor lesions: Analysis Vega, R. M., Pechman, D., Ergonul, B., Gomez, C., &
of the EORTC 10085/TBCRC/BIG/NABCG Interna- Moller, M. G. (2015). Bilateral pseudoangiomatous
tional Male Breast Cancer Program. Modern Pathol- stromal hyperplasia tumors in axillary male gyneco-
ogy, 30, 509–518. mastia: Report of a case. Surgery Today, 45, 105–109.
Fentiman, I. S., Fourquet, A., & Hortobagyi, G. N. (2006). Vermeulen, M. A., Doebar, S. C., van Deurzen, C. H. M.,
Male breast cancer. Lancet, 367(9510), 595–604. Martens, J. W. M., van Diest, P. J., & Moelans, C. B.
Hamady, Z. Z., Carder, P. J., & Brennan, T. G. (2005). (2017). Copy number profiling of oncogenes in ductal
Atypical ductal hyperplasia in male breast tissue with carcinoma in situ of the male breast. Endocrine Related
gynaecomastia. Histopathology, 47, 111–112. Cancer, 25, 173–184.
H
Immunophenotype
Hamartoma, Fig. 6 Symptomatic hamartoma consisting
of a predominantly dense hyalinized stroma with atrophic One of the characteristic immunophenotypic fea-
sparsely distributed ducts and lobular units (H&E) tures of hamartomas is that both the stroma and
epithelial elements are positive for estrogen recep-
tors and progesterone receptors, i.e., ER and PR
positive (Herbert et al. 2002). This differentiates
hamartomas from normal breast tissue which is
usually ER and PR positive in the epithelium, but
ER and PR negative in the stroma. This suggests
simultaneous response to hormonal stimulation of
the constituents of the hamartoma (Fig. 8). As
expected the epithelial elements are positive for
cytokeratins and the stroma for vimentin similar to
normal breast tissue. Hamartomas do not express
HER2 or p53 and the Ki67 proliferation activity is
Hamartoma, Fig. 7 Pseudoangiomatous hyperplasia in a
needle core biopsy from a hamartoma in Fig. 6 which could usually low (Herbert et al. 2002). Markers such as
have led to erroneous diagnosis of primary PASH (H&E) desmin highlight smooth muscle fibers in the
140 Hamartoma
Hamartoma, Fig. 8 The hamartoma from the patient in Fig. 6 expresses estrogen receptors (a) and progesterone
receptors (b) in the stroma and epithelial components. The PR appears stronger than the ER
hamartoma or support the diagnosis of a myoid gastrointestinal tract, bone, central nervous sys-
hamartoma. tem, eyes and genitourinary tract (Fiala (2016)
Medscape). Cowden syndrome is associated with
malignancy in particular breast cancer and thyroid
Genetic Aberrations cancer. Gomez et al. (2012) reported a carcinoma
of the breast arising in a calcified hamartoma. The
As the breast hamartoma is not considered neo- case was unusual in that although Cowden syn-
plastic there are very few genetic studies on this drome is a multiple hamartoma syndrome, breast
lesion. Aberrations involving 12q12-15 in a breast hamartomas are not even in the minor criteria for
hamartoma were reported independently by dif- diagnosis of this condition, but breast cancer is a
ferent authors (Dietrich et al. 1994 and Rohen major diagnostic criterion. Furthermore, the patient
et al. 1995). Using FISH studies, the authors had bilateral breast microcalcification which on
reported aberrations involving 12q12-15 which mammography involved over two-thirds of the
was mapped to the multiple aberration region breasts. Massive calcification is not one of the
(MAR). The MAR is a major cluster region of features of hamartomas. This link between PTEN
chromosome 12 breakpoints of benign solid mutations and breast hamartoma confirmed an ear-
tumors such as uterine leiomyoma, lipoma and lier study by the same authors (Sabate et al. 2006)
pleomorphic adenoma of the salivary gland. The which reported hamartomas in two patients with
authors identified the aberrations in the stroma Cowden syndrome.
and not the epithelial cells and concluded that
hamartomas arose due to mutated mesenchymal
stromal cells, and therefore adenolipomas or Differential Diagnosis
hamartomas of the breast were not due to
entrapped epithelial cells and mesenchymal cells The most common lesion which mimics a
with proliferating adipocytes but mutated mesen- hamartoma clinically is a fibroadenoma
chymal cells capable of differentiating into stro- (▶ Fibroadenoma). Fibroadenomas tend to affect
mal cells as well as adipocytes. younger women in the 20–30 age groups. Although
Breast hamartomas have also been linked to clinically and radiologically fibroadenomas resem-
Cowden disease. Cowden disease also termed ble hamartomas, pathologically the two lesions are
Cowden syndrome or multiple hamartoma syn- very different, more significantly, fibroadenomas
drome is an autosomal dominant condition with lack fat infiltration. Furthermore the epithelial com-
variable expression associated with the mutation ponent of a fibroadenoma can be very heteroge-
of the PTEN suppressor gene on 10q23.3 neous to include: sclerosing adenosis (▶ Sclerosing
(Liaw et al. 1997). Cowden disease causes Adenosis), fibrocystic change (▶ Fibrocystic Breast
hamartomatous neoplasms of the skin, mucosa, Changes) with apocrine metaplasia, columnar cell
Hamartoma 141
change (▶ Columnar Cell Lesions), epithelial detected. The well-circumscribed lesions could
hyperplasia of usual type (▶ Usual Ductal Hyper- be difficult to differentiate from a hamartoma
plasia (UDH)) without atypia. The fibroadenoma radiologically. However, the lack of sonographic
is lobulated and the epithelial proliferation exhibits compressibility in the fibrous tumor should dis-
the so called intracanalicular or pericanalicular tinguish this lesion from a hamartoma. Fibrous
growth patterns. In older patients, fibroadenomas tumors can be clinically palpable but radiologi-
become hyalinized with reduced cellularity but cally occult, which is not the case with a
with preservation of the lobular architecture. The hamartoma. In the absence of appropriate radio-
stroma in fibroadenoma is also prone to myxoid logical studies, a fibrous tumor can be difficult to
degeneration. If these features are present, the diag- differentiate from a hamartoma in a needle core
nosis of hamartoma would be questionable. biopsy (Fig. 9). In the presence of fat, a fibrous
Fibroadenomatoid hyperplasia is thought to be tumor can mimic a hamartoma with a predomi-
a harbinger of a fibroadenoma and can be difficult nantly fibrous component. The cause of a fibrous
to differentiate from a hamartoma on a needle core tumor is unknown but transforming growth factor
biopsy. This lesion consists of small lobules of beta (TGF-beta) is implicated in promoting spin-
hyalinized stroma admixed with benign ducts and dle cell proliferation (Gold 1999). H
lobular units. Inclusion of fat in the needle core Diabetic mastopathy can mimic a hamartoma
biopsy may give rise to erroneous diagnosis of radiologically. Histologically, the diagnosis
hamartoma. However, the lobulated architecture should be clear because of the presence of lobular
of a fibroadenomatoid hyperplasia plus the radio- mastitis in the background of fibrotic stroma with
logical findings should exclude a hamartoma. or without islands of mature adipocytes
Tubular adenoma (▶ Tubular Adenoma of the (▶ Diabetic Mastopathy).
Breast) which belongs to the same family as Pseudoangiomatous stromal hyperplasia
fibroadenoma can mimic a hamartoma clinically (PASH) (▶ Pseudoangiomatous Stromal Hyper-
and radiologically, but the histological appearance plasia) can be present as a primary lesion of the
of crowded tubules usually with luminal secre- breast but can arise in the stroma of other breast
tions with no intervening stroma has no resem- lesions such as gynecomastia or hamartoma. The
blance to a hamartoma. Coincidentally, a patient main pathological appearance of PASH is the
with a tubular adenoma of the breast and Cowden presence of slit-like structures which resemble
disease was reported in the same study which capillaries in the stroma. If a needle core biopsy
reported two patients with PTEN mutation in samples the pseudoangiomatous component in a
hamartomas (Sabate et al. 2006). hamartoma, as illustrated in Fig. 7, it is almost
Phyllodes tumors (▶ Phyllodes Tumor) arise in impossible to exclude primary PASH; more so
the same age groups as hamartomas. A giant
hamartoma (Cazorla and Arentz 2015) would be
clinically indistinguishable from a phyllodes
tumor. However, histologically the increase in
cellularity of the stroma with associated clefting
would distinguish a phyllodes tumor from a
hamartoma (▶ Phyllodes Tumor).
Fibromatosis of the breast may clinically and
radiologically mimic a hamartoma, but the pre-
dominance of a cellular infiltrative fibrous prolif-
eration with no epithelial elements is
characteristic of this lesion and clearly differenti- Hamartoma, Fig. 9 Needle core biopsy from a palpable
but mammorgraphically and sonographically occult
ates it from a hamartoma.
fibrous tumor. The stroma is hyalinized with a scar-like
Fibrous mastopathy or fibrous tumor can be appearance. The presence of fat and benign ducts and
present symptomatically or mammographically lobular units resembles a hamartoma (H&E)
142 Hamartoma
Hemangiomas
of the breast:
localized: diffuse:
angiomatosis
nonparenchymal: parenchymal:
cutaneous
hemangiomas
Hemangioma of the Breast, Fig. 1 A 10 mm large septa between them (c H&E, 100 magnification). Mam-
cavernous hemangioma of the breast in a 52-year-old mography (a) and ultrasonography picture (b) by courtesy
female patient. The tumor is composed of dilated, blood- of Dr. Eva Molnar, Department of Radiology, Sahlgrenska
filled spaces of different sizes and shape with thin fibrous University Hospital, Gothenburg, Sweden
Eusebi 2009). Any vascular lesion with instability or ulceration and should prevent
atypical morphology on a needle core or correct deformity.
biopsy should be completely excised, • Outcome
regardless of the degree of Ki-67, to exclude Favorable, but hemangiomas can grow over time.
an underlying angiosarcoma. Treatment of Malignant transformation is rare; they are not
hemangiomas should avoid hemodynamic likely precursors of angiosarcoma.
146 Hemangioma of the Breast
Hemangioma of the Breast, Fig. 2 (a, b) Perilobular (H&E staining, 40 and 100 magnification). (c) Neg-
hemangioma as an incidental finding in a 57-year- ative E-cadherin immunostaining in LCIS (200 magnifi-
old female patient. The coexistence of perilobular cation). (d) Positive 34bE12 immunostaining in LCIS
hemangioma and lobular carcinoma in situ (LCIS) (100 magnification)
Hemangioma of the Breast 147
Hemangioma of the Breast, Fig. 3 A 3 mm large courtesy of Dr. Bronislava Vályová, Department of
cavernous hemangioma in a 64-year-old female Radiology, Sahlgrenska University Hospital, Gothenburg,
patient, in the mastectomy scar (b H&E staining, Sweden
100 magnification). Ultrasonography picture (a) by
148 Hemangioma of the Breast
occasional mitoses, and microthrombi. They can Erythrocyte-type glucose transporter protein
be categorized into four groups according to their 1 (GLUT1) immunoreactivity is found in
growth pattern: (a) cavernous, (b) compact capil- all phases of hemangiomas and can be helpful
lary, (c) capillary budding type, and (d) combined for differential diagnosis. Pericytes are a-SMA-
atypical hemangiomas with a combination of cav- positive, whereas most angiosarcomas lack this
ernous (a) and compact capillary subtype (b). marker.
There are no signs of destructive invasion, solid
areas, hemorrhage, or necrosis. Atypical heman-
giomas have no greater risk to develop into Differential Diagnosis
angiosarcoma.
Angiomatosis is an extensive, acquired, or Hemangiomas should be differentiated from
congenital vascular lesion in young women, angiosarcoma, atypical vascular lesions (AVLs),
which may involve large areas (9–22 cm) in the pseudoangiomatous stromal hyperplasia
breast growing as non-anastomosing cystic (PASH), Mondor’s disease, angiolipoma,
spaces in between but not infiltrating the breast papillary endothelial hyperplasia (Masson’s
glandular structures. The vascular spaces are tumor), Kaposi-form hemangioendothelioma,
lined by flat endothelial cells without atypia and hemangiopericytoma, epithelioid hemangioen-
mitotic activity. The presence of pericytes can be dothelioma, benign lymphangioendothelioma,
detected by a-smooth muscle actin (a-SMA) and lymphangioma.
positivity. Because of lacking the circumscrip- Cutaneous hemangioma should be differenti-
tion that characterizes mostly the other benign ated from low-grade, postradiation cutaneous
vascular lesions, distinction from a low-grade angiosarcoma in the breast (▶ Angiosarcoma of
angiosarcoma can be difficult. However, the Breast). Angiosarcomas are located usually in
angiomatosis surrounds the ducts and the lobules the breast parenchyma and are usually larger than
without invading them. Hormonal replacement 3 cm (Howitt and Nascimento 2012). They appear
therapy with the use of exogenous estrogen may as hemorrhagic masses which involves both the
favor the appearance of angiomatosis. mammary parenchyma and the overlying skin.
Angiosarcomas usually lack a-SMA. MYC gene
amplification has been detected in secondary,
Immunophenotype postradiation angiosarcomas but not in AVLs.
Amplification of FLT4 gene (which encodes
Endothelial markers are overexpressed for CD31, VEGFR-3 and belongs to the tyrosine kinase
CD34, factor VIII, and Fli-1. However, Fli-1 is a receptor family) was found in 25% of secondary
marker for both benign and malignant vascular angiosarcomas but not in AVLs.
tumors. The Ki-67 proliferation marker is rarely Atypical vascular lesions (AVLs) (▶ Atypical
>5% in breast hemangiomas, whereas in Vascular Lesions) are limited to the upper dermis
angiosarcomas, it is usually >20%. Ki-67 may in patients with prior exposure to radiation ther-
help to highlight proliferative activity in border- apy. Two distinct morphological patterns exist: a
line vascular lesions. However, increased Ki-67 lymphatic type and a vascular type. Occasionally,
immunoreactivity can be seen in a hemangioma at both types may be seen simultaneously within
the site of a prior core biopsy and in thrombi; the same lesion. The vascular type of the lesion
therefore, fields with these reactive lesions should shows round to linear, capillary-sized vessels
be avoided during assessment of Ki-67 index. The which grow irregularly within the superficial or
absence of cell cycle regulatory protein Skp2 can deep dermis. Moderate nuclear atypia may be
be used in combination with Ki-67 for hemangi- encountered, but no mitotic figures. The lesions
oma diagnoses. Androgen receptor expression frequently show endothelial hobnailing and anas-
may be seen in hemangiomas. tomosing vascular channels.
Hemangioma of the Breast 149
Hemangioma of the Breast, Fig. 4 Mondor’s disease. lesion. (a, b) H&E staining (200 magnification). (c–e)
A 48-year-old female patient with abscess in the right Masson trichrome staining (100 , 200 and
breast. There was thrombophlebitis at the periphery of the 100 magnification)
References and Further Reading Howitt, B., & Nascimento, A. F. (2012). Vascular lesions
of the breast. Surgical Pathology, 5, 645–659.
Brodie, C., & Provenzano, E. (2008). Vascular prolifera- Rosen, P. P., Jozefczyk, M. A., & Boram, L. H. (1985).
tions of the breast. Histopathology, 52, 30–44. Vascular tumors of the breast. IV. The venous heman-
Calonje, E., & Fletcher, C. D. M. (1991). Sinusoidal hem- gioma. The American Journal of Surgical Pathology, 9,
angioma. A distinctive benign vascular neoplasm within 659–665.
the group of cavernous hemangiomas. The American Tavassoli, F. A., & Eusebi, V. (2009). Tumors of the mammary
Journal of Surgical Pathology, 15, 1130–1135. gland (AFIP atlas of tumor pathology, series 4, fascicles
10, pp. 283–290). Silver Spring, Maryland, USA.
HER2 in Breast Cancer 151
of the target (HER2) by either frontline in situ ISH (SISH). ISH tests, either in dark or in bright
hybridization (ISH) or immunohistochemistry field, can be performed with a single-color probe
(IHC) followed by ISH in borderline cases. (HER2 gene probe) or with a dual-color probe
When applying a two-step approach, IHC is (probes for the HER2 gene and the centromere
applied first and scored according to a three-tier of the chromosome 17 (CEP17)).
scoring system according to the ASCO/CAP 2018 Alternative probes mapping to the chromo-
guidelines (score 0/1+, score 2+, score 3+; see some 17 have been used and proposed by several
Fig. 1a–c, respectively) (Wolff et al. 2018). When- groups; however, it is important to note that chro-
ever an equivocal overexpression (score 2+) is mosome 17 is highly recurrently altered in breast
encountered, the sample must be subjected to cancers (Arriola et al. 2008; Isola et al. 2004;
ISH to verify the presence of HER2 gene Marchio et al. 2009; Rondon-Lagos et al. 2014);
amplification. therefore this approach should be strongly
discouraged.
Cleared Methods Alternative techniques to assess the HER2 sta-
Three methods of ISH have been introduced in tus, such as mRNA assays (e.g., RT-PCR), did not
breast pathology and are FDA approved for HER2 reach sufficient evidence to warrant inclusion, at
gene assessment in breast cancer: fluorescence least when dealing with unselected patients (Wolff
ISH (FISH), chromogenic ISH (CISH), and silver et al. 2018).
HER2 in Breast Cancer, Fig. 1 HER2 immunohisto- membrane staining in >10% of tumor cells (score 2+). (c)
chemistry. (a) weak, incomplete membrane staining in strong, complete membrane staining in the vast majority of
>10% of tumor cells (score 1+). (b) moderate, complete tumor cells (score 3+)
HER2 in Breast Cancer 153
HER2 in Breast Cancer, Table 1 Summary of changes of HER2 evaluation by ISH over the ASCO/CAP guideline
updates (2007, 2013, 2018). FDA-recommended thresholds are also reported H
NOT
Guideline Method AMP/positive Equivocal AMP/negative
FDA/ HER2/ 2 // <2
EMA CEP17
ratio
HER2 >4 4
copy
number
ASCO/ HER2/ >2.2 1.8–2.2 <1.8
CAP CEP17
2007 ratio
HER2 >6 4–6 <4
copy
number
ASCO/ ISH (a) HER2/CEP17 ratio 2, regardless of HER2/CEP17 <2 with HER2/CEP17
CAP algorithm HER2 copy number HER2 copy number 4 and <2 with HER2
2013 (b) HER2/CEP17 <2 but HER2 copy <6 copy number
dual- number 6 <4
color
ASCO/ HER2 6 4 and <6 <4
CAP copy
2013 number
single-
color
ASCO/ ISH (a) HER2/CEP17 ratio 2, regardless of HER2/CEP17 <2 with HER2/CEP17
CAP algorithm mean HER2 copy number (but additional HER2 copy number 4 and <2 with HER2
2018 work-up needed in case HER2 copy <6 (additional work-up copy number
dual- number mean is <4) needed) <4
color (b) HER2/CEP17 <2 but HER2 copy
number 6 (but additional work-up
needed)
ASCO/ HER2 6 4 and <6 (additional <4
CAP copy work-up needed)
2018 number
single-
color
AMP Amplified, ASCO American Association of Clinical Oncology, CAP College of American Pathologists, EMA
European Medicines Agency, FDA Food and Drug Administration, NOT AMP Not Amplified
154 HER2 in Breast Cancer
low power objective and observed within a homo- Additional work-up for scenarios in which the
geneous and contiguous invasive cell population). tumor cell population shows HER2/CEP17 ratio
2.0 and average HER2 copy number <4.0 sig-
By ISH:
nals/cell.
This is a challenging scenario in which, for
– Dual-probe assay: HER2/CEP17 ratio 2.0
instance, tumors harboring monosomy of chro-
and average HER2 copy number 4.0 sig-
mosome 17 easily lead to ratio values >2 despite
nals/cell (Fig. 2a).
a not high absolute HER2 copy number (mean of
– Dual-probe assay: HER2/CEP17 ratio 2.0
2 or 3). Importantly, Press and colleagues have
and average HER2 copy number <4.0 sig-
recently shown that these patients seem not to
nals/cell: additional work-up needed.
benefit from trastuzumab treatment (Press
– Dual-probe assay: HER2/CEP17 ratio <2.0
et al. 2016).
and average HER2 copy number 6 signals/
The updated guidelines suggest that, if not
cell: additional work-up needed.
already assessed by the institution or laboratory
– Single-probe assay: average HER2 copy num-
performing the ISH test, IHC testing for HER2
ber 6.0 signals/cell: concurrent score 3+ in
should be performed (using sections from the
IHC should be documented, otherwise dual
same tissue sample used for ISH) and the slides
colour ISH should be performed.
HER2 in Breast Cancer, Fig. 2 Dual-probe HER2 fluo- CEP17 copy number of 3 (green signals) leading to a
rescence in situ hybridization (FISH). (a) nuclei of tumor HER2/CEP17 ratio of 1.4. (c) tumor cells harbor a mean
cells homogeneously show multiple red signals (HER2 HER2 copy number of 2 (red signals) and a CEP17 copy
signals) clustering together. (b) tumor cells display a number of 1.9 with a HER2/CEP17 ratio of 1.05
mean HER2 copy number of 4.1 (red signals) and a
HER2 in Breast Cancer 155
from both ISH and IHC should be reviewed The ISH algorithm in this case has allowed
together to guide the selection of areas to score HER2 gene amplification not to be
by ISH: underestimated in a subset of tumors harbor-
ing high HER2 copy numbers together with
(a) IHC result is 3+: diagnosis is HER2-positive. CEP17 gain or amplification, a scenario that
(b) If the IHC result is 0 or 1+: diagnosis is would lead to a HER2/CEP17 ratio <2 even in
HER2-negative with a comment*. the presence of a high HER2 copy number.
(c) If IHC result is 2+, recount ISH by having an The phenomenon of CEP17 gain/amplifica-
additional observer, blinded to previous ISH tion has been demonstrated by the study of
results, and count at least 20 cells that include microarray-based comparative genomic
the area of invasive cancer with IHC 2+ hybridization (array CGH) (Marchio et al.
staining: 2009; Moelans et al. 2010; Yeh et al. 2009)
– If reviewing the count by the additional and further confirmed by other indirect
observer changes the result into another methods (Ercolani et al. 2017; Sapino et al.
ISH category, the result should be adjudi- 2014). Although these carcinomas account for
cated per internal procedures to define the a minority of cases, they are also the most H
final category. challenging in diagnostic practice.
– If the count remains an average of <4.0 Now, in the 2018 updated recommenda-
HER2 signals per cell and the HER2/ tions, the recommended additional work-up
CEP17 ratio is >2.0, diagnosis is HER2- is as follows: if not already assessed by the
negative with a comment*. institution or laboratory performing the ISH
test, IHC testing for HER2 should be
*The Expert Panel recommends the fol- performed (using sections from the same tis-
lowing comment: “Evidence is limited on the sue sample used for ISH), and the slides from
efficacy of HER2-targeted therapy in the both ISH and IHC should be reviewed
small subset of cases with a HER2/CEP17 together to guide the selection of areas to
ratio of 2.0 and an average HER2 copy score by ISH:
number of <4.0 per cell. In the first generation (a) IHC result is 3+: diagnosis is HER2-positive.
of adjuvant trastuzumab trials, patients in this (b) If the IHC result is 0 or 1+, diagnosis is
subgroup who were randomly assigned to the HER2-negative with a comment**.
trastuzumab arm did not seem to derive an (c) If the IHC result is 2+, recount ISH by having
improvement in disease-free or overall sur- an additional observer, blinded to previous
vival, but there were too few such cases to ISH results, and count at least 20 cells that
draw definitive conclusions. IHC expression include the area of invasion with IHC 2+
for HER2 should be used to complement ISH staining:
and define HER2 status. If the IHC result is – If reviewing the count by the additional
not 3+ positive, it is recommended that the observer changes the result into another
specimen be considered HER2 negative ISH category, the result should be adjudi-
because of the low HER2 copy number by cated per internal procedures to define the
ISH and the lack of protein overexpression.” final category.
(Type, evidence based; evidence quality, – If the HER2/CEP17 ratio remains <2.0
intermediate; strength of recommendation, with >6.0 HER2 signals per cell, diagnosis
strong) is HER2-positive.
Additional work-up for scenarios in which **The Expert Panel recommends the fol-
the tumor cell population shows HER2/ lowing comment: “There are insufficient data
CEP17 ratio <2.0 and average HER2 copy on the efficacy of HER2-targeted therapy in
number 6.0 signals/cell. cases with a HER2 ratio of <2.0 in the
156 HER2 in Breast Cancer
threshold for positive, there is a high likeli- and usually display a relatively high mean of
hood that repeat testing will result in different Ki67 indices (Ballard et al. 2017; Ragazzi et al.
results by chance alone. Therefore, when IHC 2017; Sapino et al. 2014). If compared to HER2-
results are not 3+ positive, it is recommended negative disease, these tumors display a signifi-
that the sample be considered HER2 negative cantly higher prevalence of grade 3 carcinomas,
without additional testing on the same harbor significantly higher proliferation indices,
specimen.” and show a significantly higher proportion of
(Type, evidence based; evidence quality, lymph node metastases at presentation (Ballard
intermediate; strength of recommendation, et al. 2017; Ragazzi et al. 2017; Sapino et al.
strong) 2014). If compared to HER2-positive disease,
HER2 double-equivocal carcinomas show a sig-
The experts also suggest that clinical correlation nificantly higher rate of ER positivity and a sig-
with other factors in a particular case (such as grade nificantly lower prevalence of histological grade
and special histologic subtypes) or repeat testing of 3 carcinomas (Ballard et al. 2017; Ragazzi et al.
other tissue samples from the patient may also be 2017; Sapino et al. 2014). Data on the efficacy of
appropriate in this setting. In particularly challeng- response to anti-HER2 therapy are not available at H
ing cases or if the results are in question, expert present, and we only know that these carcinomas
consultation may be appropriate and may include do not do worse than HER2-negative breast car-
alternative probes or other genetic methods. How- cinomas treated with anthracycline- and taxane-
ever, alternative probes should not be used as stan- based chemotherapy (Press et al. 2016). Recently,
dard practice due to limited data on outcomes for an extensive genomic analysis has revealed that
this subset of patients. Regarding the latter, the double-equivocal carcinomas are preferentially
experts acknowledge that this group of carcinomas luminal B also at the transcriptomic level. In addi-
has been frequently tested by ISH using multiple tion, in these cases, HER2 mRNA levels are more
chromosome 17 probes at once, many not analyti- widely overlapping with HER2-negative rather
cally or clinically validated. Such indiscriminate than with HER2-positive breast carcinomas.
testing often results in four or more ISH ratios However, they display a high risk of recurrence
being described in a single test report and a final by Prosigna ® assay, even when they present good
designation of HER2 gene amplified if just a single prognostic features (unifocal, <2 cm, node nega-
ratio is >2.0. After careful consideration of this tive/micrometastatic carcinomas) (Marchio et al.
practice and available data, the Expert Panel 2018). Interestingly, a subset of double-equivocal
strongly recommends against this as a routine test- cases is classified as HER2-enriched by gene
ing strategy. expression analysis in spite of their double-
According to this work-up, the equivocal result equivocal status, opening the possibility to
is in a way “sorted out” as negative; nevertheless, explore the beneficial effect of anti-HER2 agents
the experts acknowledge that data on this sub- in this subgroup of patients (Marchio et al. 2018).
group of patients are scant. Cases as such would In addition, at the mutational level, double-
be defined as HER2 double-equivocal breast car- equivocal carcinomas show PIK3CA and TP53
cinomas, i.e., breast carcinomas with an equivocal mutation rates more similar to ER-positive/
expression of the HER2 protein (score 2+ by IHC) HER2-positive rather than to ER-positive/HER2-
and mean HER2 copy numbers in the equivocal negative carcinomas, supporting the theory that
range (i.e., HER2/CEP17 ratio <2.0 and mean these cases represent an aggressive subgroup of
HER2 copy numbers 4 and <6 by ISH). These ER-positive carcinomas (Marchio et al. 2018).
breast carcinomas fall into the luminal B subtype The responses to chemotherapy and
when using the IHC surrogate proposed by the trastuzumab in this category of patients are still
St. Gallen Consensus (▶ Invasive Carcinoma unclear. In a cohort of patients treated in the neo-
NST), as they are typically estrogen receptor pos- adjuvant setting with trastuzumab-containing che-
itive (▶ Hormone Receptors in Breast Cancer) motherapy, double-equivocal carcinoma patients
158 HER2 in Breast Cancer
HER2 in Breast Cancer, Table 2 Prevalence of HER2 derived from the TCGA study by Curtis et al. (2012) and
mutations in the distinct PAM50-defined molecular sub- from Ciriello et al. (2015))
types (TCGA data from www.cbioportal.org). (Data
Frequency of HER2 mutations
N %
Luminal (A/B) 5/321 1.6
Luminal A 3/235 1.3
ILC Luminal A 4/106 4
Luminal B 2/133 1.5
HER2-enriched 2/58 3
Basal-like 0/81 0
ILC invasive lobular carcinoma, N number of cases
had low rates of pathologic complete response et al. 2013; Lee et al. 2006). These cases account
(pCR), but a great proportion reached near-pCR: for about 1.3–4% of all breast cancers, depending
when these two categories were grouped, the dif- on the molecular subtype and the histologic type.
ference in terms of response rate was not statisti- They represent an important subgroup of HER2-
cally significant between double-equivocal and activated cancers that may be missed by standard
HER2-positive carcinomas (Marchio et al. HER2 positivity assessment (IHC or ISH).
2018). However, larger studies are warranted to Indeed, these mutations are found in breast carci-
ascertain the real impact of chemotherapy and nomas regardless of the presence of HER2 ampli-
anti-HER2 therapy in this specific subset of breast fication, occurring in both ER-positive/HER2-
carcinomas. negative diseases and HER2-enriched cancers
(Table 2) (Petrelli et al. 2017). For this reason,
HER2 Negative Result they represent tumors considered to be HER2-
By IHC: negative (by the routinely used methods and algo-
– Score 1+: incomplete membrane staining rithms at present) but with active HER2 signaling
that is faint/barely perceptible and in that can be targeted by anti-HER2 agents.
>10% of tumor cells. In breast cancer, HER2 mutations are clustered
– Score 0: (i) no staining is observed or mainly in two different regions: the extracellular
(ii) membrane staining that is incomplete domain, at exon 8, and the kinase domain, at exons
and is faint/barely perceptible and in 19 and 20. The majority of HER2 mutations
10% of tumor cells. described so far in the context of breast cancer
By ISH: affect codons belonging to the kinase domain.
– Single-probe assay: average HER2 copy The most frequent HER2 mutations are L755S,
number <4.0 signals/cell: double-check V777L, and D769H or D769Y: all of them are
whether concurrent IHC 0, 1+ and/or con- activating mutations, likely driving tumorigene-
current dual-probe ISH with a result of sis. In preclinical studies, L755S is related to
HER2 copy number <4.0 signals/cell. resistance to the reversible HER2 inhibitor
– Dual-probe assay: HER2/CEP17 ratio <2.0 lapatinib, but it is sensitive to the irreversible
and average HER2 copy number <4.0 sig- HER2 inhibitor, neratinib (Bose et al. 2013;
nals/cell (Fig. 2c). Kancha et al. 2011), while the other two mutations
(V777L, and D769H or D769Y) are associated
with sensitivity to trastuzumab, lapatinib, and
HER2 Mutations neratinib (Bose et al. 2013; Petrelli et al. 2017).
Different studies have suggested that invasive
Studies based on massively parallel sequencing lobular carcinomas (▶ Invasive Lobular Carci-
have unveiled the presence of a subgroup of breast noma) are enriched in HER2 mutations (Ciriello
carcinomas harboring HER2 mutations (Bose et al. 2015; Lien et al. 2015; Ross et al. 2013).
HER2 in Breast Cancer 159
A high HER2 mutation rate has also been reported cancer: A multi-institutional study. Modern Pathology,
in metastatic breast cancer. Interestingly, in this 30, 227–235.
Bose, R., Kavuri, S. M., Searleman, A. C., Shen, W., Shen,
subset of tumors, mutations are more frequent in D., Koboldt, D. C., Monsey, J., Goel, N., Aronson,
patient after the administration of trastuzumab A. B., Li, S., Ma, C. X., Ding, L., Mardis, E. R., &
(Fang et al. 2014). In addition, HER2 mutations Ellis, M. J. (2013). Activating HER2 mutations in
can represent a marker of resistance to anti-HER2 HER2 gene amplification negative breast cancer. Can-
cer Discovery, 3, 224–237.
therapy in this subset of patients (Park et al. 2015). Ciriello, G., Gatza, M. L., Beck, A. H., Wilkerson, M. D.,
Using a basket-trial approach, Hyman et al. Rhie, S. K., Pastore, A., Zhang, H., McLellan, M., Yau,
(2018) have tested the effects of neratinib on C., Kandoth, C., Bowlby, R., Shen, H., Hayat, S.,
many patients with HER2 mutations. Sequenc- Fieldhouse, R., Lester, S. C., Tse, G. M., Factor,
R. E., Collins, L. C., Allison, K. H., Chen, Y. Y., Jensen,
ing analysis demonstrated that most patients K., Johnson, N. B., Oesterreich, S., Mills, G. B.,
whose HER2 mutations were present in all the Cherniack, A. D., Robertson, G., Benz, C., Sander,
tumor cells responded to neratinib, whereas C., Laird, P. W., Hoadley, K. A., King, T. A., Network,
those with HER2 mutations in only a subset of T. R., & Perou, C. M. (2015). Comprehensive molecu-
lar portraits of invasive lobular breast cancer. Cell, 163,
the tumor cells did not respond. Neratinib
exhibited the greatest degree of activity in
506–519.
Cocco, E., Javier Carmona, F., Razavi, P., Won, H. H., Cai,
H
patients with breast cancer (n = 25 total, objec- Y., Rossi, V., Chan, C., Cownie, J., Soong, J., Toska, E.,
tive response rate at week 8 of 32%) with mis- Shifman, S. G., Sarotto, I., Savas, P., Wick, M. J.,
Papadopoulos, K. P., Moriarty, A., Cutler, R. E., Jr.,
sense mutations involving the extracellular and Avogadri-Connors, F., Lalani, A. S., Bryce, R. P.,
kinase domains, as well as insertions in the Chandarlapaty, S., Hyman, D. M., Solit, D. B., Boni,
kinase domain. These tumors were all classified V., Loi, S., Baselga, J., Berger, M. F., Montemurro, F.,
as HER2-negative (lack of amplification) at the & Scaltriti, M. (2018). Neratinib is effective in breast
tumors bearing both amplification and mutation of
time of enrolment per established guidelines ERBB2 (HER2). Science Signaling, 11, pii: eaat9773.
(Wolff et al. 2013). Curtis, C., Shah, S. P., Chin, S. F., Turashvili, G., Rueda,
Recently, preclinical data have shown that the O. M., Dunning, M. J., Speed, D., Lynch, A. G.,
co-occurrence of a HER2 mutation in HER2 Samarajiwa, S., Yuan, Y., Graf, S., Ha, G., Haffari,
G., Bashashati, A., Russell, R., McKinney, S., META-
amplified tumors reduces the efficacy of therapies BRIC Group, Langerod, A., Green, A., Provenzano, E.,
commonly used to treat HER2-positive breast Wishart, G., Pinder, S., Watson, P., Markowetz, F.,
cancer, particularly in metastatic and previously Murphy, L., Ellis, I., Purushotham, A., Borresen-Dale,
HER2 inhibitor-treated patients, as well as poten- A. L., Brenton, J. D., Tavare, S., Caldas, C., &
Aparicio, S. (2012). The genomic and transcriptomic
tially in patients scheduled for first-line treatment architecture of 2,000 breast tumours reveals novel sub-
(Cocco et al. 2018). These data may suggest the groups. Nature, 486, 346–352.
need of clinical studies testing the efficacy of Ercolani, C., Marchio, C., Di Benedetto, A., Fabi, A.,
neratinib in breast cancer patients whose tumors Perracchio, L., Vici, P., Sperati, F., Buglioni, S.,
Arena, V., Pescarmona, E., Sapino, A., Terrenato, I.,
carry both amplification and mutation of the & Mottolese, M. (2017). Breast carcinomas with low
HER2 gene (Cocco et al. 2018). amplified/equivocal HER2 by Ish: Potential supporting
role of multiplex ligation-dependent probe amplifica-
tion. Journal of Experimental & Clinical Cancer
Research, 36, 143.
References and Further Reading Fang, Y., Jiang, Y., Wang, X., Yang, X., Gao, Y., & Wang,
J. (2014). Somatic mutations of the HER2 in metastatic
Arriola, E., Marchio, C., Tan, D. S., Drury, S. C., Lambros, breast cancer. Tumour Biology, 35, 11851–11854.
M. B., Natrajan, R., Rodriguez-Pinilla, S. M., Mackay, Fehrenbacher, L., Cecchini, R., Geyer, C., Rastogi, P.,
A., Tamber, N., Fenwick, K., Jones, C., Dowsett, M., Costantino, J., Atkins, J., Polikoff, J., Boileau, J.-F.,
Ashworth, A., & Reis-Filho, J. S. (2008). Genomic Provencher, L., Stokoe, C., Moore, T., Robidoux, A.,
analysis of the HER2/TOP2A amplicon in breast can- Borges, V., Albain, K., Swain, S., Paik, S., Mamounas,
cer and breast cancer cell lines. Laboratory Investiga- E., & Wolmark, N. (2018). Abstract GS1-02: NSABP
tion, 88, 491–503. B-47 (NRG oncology): Phase III randomized trial com-
Ballard, M., Jalikis, F., Krings, G., Schmidt, R. A., Chen, paring adjuvant chemotherapy with adriamycin (A) and
Y. Y., Rendi, M. H., Dintzis, S. M., Jensen, K. C., West, cyclophosphamide (C) ! weekly paclitaxel (WP), or
R. B., Sibley, R. K., Troxell, M. L., & Allison, K. H. docetaxel (T) and C with or without a year of
(2017). ‘Non-classical’ HER2 FISH results in breast trastuzumab (H) in women with node-positive or
160 HER2 in Breast Cancer
high-risk node-negative invasive breast cancer (IBC) Orlassino, R., Cassoni, P., Pich, A., Montemurro, F.,
expressing HER2 staining intensity of IHC 1+ or 2+ Mottolese, M., Vincent-Salomon, A., Penault-Llorca,
with negative FISH (HER2-Low IBC). Cancer F., Medico, E., Ng, C. K. Y., Viale, G., & Sapino, A.
Research, 78(4 Supplement), GS1-02-GS01-02. (2018). The dilemma of HER2 double-equivocal breast
Hyman, D. M., Piha-Paul, S. A., Won, H., Rodon, J., Saura, carcinomas: Genomic profiling and implications for
C., Shapiro, G. I., Juric, D., Quinn, D. I., Moreno, V., treatment. American Journal of Surgical Pathology,
Doger, B., Mayer, I. A., Boni, V., Calvo, E., Loi, S., 42, 1190–1200.
Lockhart, A. C., Erinjeri, J. P., Scaltriti, M., Ulaner, Moelans, C. B., de Weger, R. A., & van Diest, P. J. (2010).
G. A., Patel, J., Tang, J., Beer, H., Selcuklu, S. D., Absence of chromosome 17 polysomy in breast cancer:
Hanrahan, A. J., Bouvier, N., Melcer, M., Murali, R., Analysis by CEP17 chromogenic in situ hybridization
Schram, A. M., Smyth, L. M., Jhaveri, K., Li, B. T., and multiplex ligation-dependent probe amplification.
Drilon, A., Harding, J. J., Iyer, G., Taylor, B. S., Berger, Breast Cancer Research and Treatment, 120, 1–7.
M. F., Cutler, R. E., Jr., Xu, F., Butturini, A., Eli, L. D., Park, Y. H., Shin, H. T., Jung, H. H., Choi, Y. L., Ahn, T.,
Mann, G., Farrell, C., Lalani, A. S., Bryce, R. P., Park, K., Lee, A., Do, I. G., Kim, J. Y., Ahn, J. S., Park,
Arteaga, C. L., Meric-Bernstam, F., Baselga, J., & W. Y., & Im, Y. H. (2015). Role of HER2 mutations in
Solit, D. B. (2018). HER kinase inhibition in patients refractory metastatic breast cancers: Targeted sequenc-
with HER2- and HER3-mutant cancers. Nature, 554, ing results in patients with refractory breast cancer.
189–194. Oncotarget, 6, 32027–32038.
Isola, J., Tanner, M., Forsyth, A., Cooke, T. G., Watters, Petrelli, F., Tomasello, G., Barni, S., Lonati, V.,
A. D., & Bartlett, J. M. (2004). Interlaboratory com- Passalacqua, R., & Ghidini, M. (2017). Clinical and
parison of HER-2 oncogene amplification as detected pathological characterization of HER2 mutations in
by chromogenic and fluorescence in situ hybridization. human breast cancer: A systematic review of the liter-
Clinical Cancer Research, 10, 4793–4798. ature. Breast Cancer Research and Treatment, 166,
Kancha, R. K., von Bubnoff, N., Bartosch, N., Peschel, C., 339–349.
Engh, R. A., & Duyster, J. (2011). Differential sensi- Press, M. F., Sauter, G., Buyse, M., Fourmanoir, H.,
tivity of ERBB2 kinase domain mutations towards Quinaux, E., Tsao-Wei, D. D., Eiermann, W., Robert,
lapatinib. PLoS One, 6, e26760. N., Pienkowski, T., Crown, J., Martin, M., Valero, V.,
Lee, J. W., Soung, Y. H., Seo, S. H., Kim, S. Y., Park, C. H., Mackey, J. R., Bee, V., Ma, Y., Villalobos, I., Campeau,
Wang, Y. P., Park, K., Nam, S. W., Park, W. S., A., Mirlacher, M., Lindsay, M. A., & Slamon, D. J.
Kim, S. H., Lee, J. Y., Yoo, N. J., & Lee, S. H. (2016). HER2 gene amplification testing by fluorescent
(2006). Somatic mutations of ERBB2 kinase domain in situ hybridization (FISH): Comparison of the ASCO-
in gastric, colorectal, and breast carcinomas. Clinical College of American pathologists guidelines with FISH
Cancer Research, 12, 57–61. scores used for enrollment in Breast Cancer Interna-
Lien, H. C., Chen, Y. L., Juang, Y. L., & Jeng, Y. M. (2015). tional Research Group Clinical Trials. Journal of Clin-
Frequent alterations of HER2 through mutation, ampli- ical Oncology, 34, 3518–3528.
fication, or overexpression in pleomorphic lobular car- Ragazzi, M., Bisagni, A., Gasparini, E., Kuhn, E.,
cinoma of the breast. Breast Cancer Research and Bassano, C., Tamagnini, I., Foroni, M., Bortesi, M.,
Treatment, 150, 447–455. Falco, G., Ferrari, G., Braglia, L., Savoldi, L., Bologna,
Ma, C. X., Bose, R., Gao, F., Freedman, R. A., Telli, M. L., A., Di Cicilia, R., Bisagni, G., & Gardini, G. (2017).
Kimmick, G., Winer, E., Naughton, M., Goetz, M. P., Impact of 2013 ASCO/CAP guidelines on HER2 deter-
Russell, C., Tripathy, D., Cobleigh, M., Forero, A., mination of invasive breast cancer: A single institution
Pluard, T. J., Anders, C., Niravath, P. A., Thomas, S., experience using frontline dual-color FISH. Breast, 34,
Anderson, J., Bumb, C., Banks, K. C., Lanman, R. B., 65–72.
Bryce, R., Lalani, A. S., Pfeifer, J., Hayes, D. F., Rondon-Lagos, M., Verdun Di Cantogno, L., Rangel, N.,
Pegram, M., Blackwell, K., Bedard, P. L., Al-Kateb, Mele, T., Ramirez-Clavijo, S. R., Scagliotti, G., March-
H., & Ellis, M. J. C. (2017). Neratinib efficacy and io, C., & Sapino, A. (2014). Unraveling the chromo-
circulating tumor DNA detection of HER2 mutations some 17 patterns of FISH in interphase nuclei: An
in HER2 nonamplified metastatic breast cancer. Clini- in-depth analysis of the HER2 amplicon and chromo-
cal Cancer Research, 23, 5687–5695. some 17 centromere by karyotyping, FISH and
Marchio, C., Lambros, M. B., Gugliotta, P., Di Cantogno, M-FISH in breast cancer cells. BMC Cancer, 14, 922.
L. V., Botta, C., Pasini, B., Tan, D. S., Mackay, A., Ross, J. S., Wang, K., Sheehan, C. E., Boguniewicz, A. B.,
Fenwick, K., Tamber, N., Bussolati, G., Ashworth, A., Otto, G., Downing, S. R., Sun, J., He, J., Curran, J. A.,
Reis-Filho, J. S., & Sapino, A. (2009). Does chromo- Ali, S., Yelensky, R., Lipson, D., Palmer, G., Miller,
some 17 centromere copy number predict polysomy in V. A., & Stephens, P. J. (2013). Relapsed classic
breast cancer? A fluorescence in situ hybridization and E-cadherin (CDH1)-mutated invasive lobular breast
microarray-based CGH analysis. Journal of Pathology, cancer shows a high frequency of HER2 (ERBB2)
219, 16–24. gene mutations. Clinical Cancer Research, 19,
Marchio, C., Dell’Orto, P., Annaratone, L., Geyer, F. C., 2668–2676.
Venesio, T., Berrino, E., Verdun di Cantogno, L., Sapino, A., Maletta, F., Verdun di Cantogno, L., Macri, L.,
Garofoli, A., Rangel, N., Casorzo, L., dell’Aglio, C., Botta, C., Gugliotta, P., Scalzo, M. S., Annaratone, L.,
Gugliotta, P., Trisolini, E., Beano, A., Pietribiasi, F., Balmativola, D., Pietribiasi, F., Bernardi, P., Arisio, R.,
Hormone Receptors in Breast Cancer 161
Growth factor
E2
ER ER
Cytoplasm
ER
ER P
Fos
Foss JJun
un
n
H
Nucleus AP1/SP1
P P
ER ER ER
ER P
Fos
Foss Jun
n
ERE
Non-classical pathway
mRNA
Protein
Estrogen action
Hormone Receptors in Breast Cancer, traditional ligand- dependent transcription pathway and
Fig. 1 Mechanism of action of estrogen receptor. The the indirect activation through growth factor binding
receptor can be activated via two mechanisms; the
The most widely used scoring system is the possibility is to evaluate the percentage of any
Allred (Quick) score. The score is a sum of the nuclear staining, independently from the intensity.
intensity (0, 1, 2, 3) and percentage (scores 0, 1, 2,
3, 4, 5) scores giving a final score ranging from
zero (negative) to 8/8 (strongly positive) (Harvey Relevance for Breast Pathology
et al. 1999). Another commonly used system is
the H-score which is obtained by adding the sum ER is routinely assessed on all newly diagnosed
of multiplying the percentage by intensity scores primary breast cancers, and this is mandated
(including different staining intensities within in the pathology and management guidelines
the tumor, thus accounting for heterogeneity). (Guidelines Working Group of the UK National
The final score ranges from 0 to 300. Another Coordinating Committee for Breast Pathology
164 Hormone Receptors in Breast Cancer
2016; National Institute for Health and Care The prognostic significance of ERb protein
Excellence 2009). Depending on the guidelines, and mRNA expression in breast carcinoma
PR assessment is optional or mandatory has been conflicting. A recent meta-analysis
(American Society of Clinical Oncology/College of 21 studies of 6769 patients for ERb1, 2295
of American Pathologists guideline 2010). Both patients for ERb2, and 2271 patients for ERb5
receptors are increasingly tested in recurrent concluded that ERb1 protein expression corre-
and metastatic breast cancers to aid management lated with favorable survival (DFS, OS)
decisions. AR and ERb are not analyzed in the and ERb2 with improved DFS only, whereas
routine setting. ERb5 was not associated with DFS (Liu
Traditionally, testing for ER/PR was et al. 2016).
performed on surgical resection specimens. With AR has been shown to be an independent
increasing demands for early availability of prognosticator (when tumor size, grade, and
results to help select patients for neoadjuvant ther- nodal status were included in the model) of favor-
apy and preoperative trials, testing is currently able outcome not only in ER-positive breast car-
performed on core biopsies in the majority of cinoma but also in ER-negative tumors
cases. Studies have shown excellent concordance (Aleskandarany et al. 2016).
of receptor results between core and excision A prognostic index (ERPI) was created using
samples (Hodi et al. 2007). AR status (positive or negative) together with
Repeat testing on surgical specimens is advised tumor size and lymph node status that clearly
in the presence of multiple tumors, tumor hetero- separated patients with luminal-A and luminal-B
geneity, if tumor cells on core biopsy were scanty, breast cancer into good and poor prognosis groups
or if the staining on core sections was technically (Castellano et al. 2013).
suboptimal/failed.
Hammond ME, Hayes DF, Wolff AC, Mangu PB, Temin S. receptors. Nature Reviews Molecular Cell Biology,
American Society of Clinical Oncology/College of 17, 783–797.
American Pathologists Guideline Recommendations Liu, J., Guo, H., Mao, K., Zhang, K., Deng, H., & Liu, Q.
for Immunohistochemical testing of estrogen and pro- (2016). Impact of estrogen receptor-beta expression
gesterone receptors in breast cancer. J Oncol Pract. on breast cancer prognosis: A meta-analysis. Breast
2010 Jul;6(4):195–7. https://doi.org/10.1200/JOP. Cancer Research and Treatment, 156, 149–162.
777003 Mote, P. A., Gompel, A., Howe, C., Hilton, H. N.,
Harvey, J. M., Clark, G. M., Osborne, C. K., & Sestak, I., Cuzick, J., Dowsett, M., Hugol, D.,
Allred, D. C. (1999). Estrogen receptor status by immu- Forgez, P., Byth, K., Graham, J. D., & Clarke, C. L.
nohistochemistry is superior to the ligand-binding (2015). Progesterone receptor A predominance is a
assay for predicting response to adjuvant endocrine discriminator of benefit from endocrine therapy in the
therapy in breast cancer. Journal of Clinical Oncology, ATAC trial. Breast Cancer Research and Treatment,
17, 1474–1481. 151, 309–318.
Hodi, Z., Chakrabarti, J., Lee, A. H., Ronan, J. E., National Institute for Health and Care Excellence.
Elston, C. W., Cheung, K. L., Robertson, J. F., & (2009). Early and locally advanced breast cancer:
Ellis, I. O. (2007). The reliability of assessment Diagnosis and treatment, NICE guidelines [CG80].
of oestrogen receptor expression on needle core Accessed 29 Dec 2017, from http://www.nice.org.uk/
biopsy specimens of invasive carcinomas of the breast. guidance/cg80.
Journal of Clinical Pathology, 60, 299–302.
Howell, A., Cuzick, J., Baum, M., Buzdar, A.,
Nemere, I., Pietras, R. J., & Blackmore, P. F. (2003).
Membrane receptors for steroid hormones: Signal
H
Dowsett, M., Forbes, J. F., Hoctin-Boes, G., transduction and physiological significance. Journal
Houghton, J., Locker, G. Y., Tobias, J. S., & of Cellular Biochemistry, 88, 438–445.
A. T. Group. (2005). Results of the ATAC (Arimidex, Riemsma, R., Forbes, C. A., Kessels, A., Lykopoulos, K.,
tamoxifen, alone or in combination) trial after comple- Amonkar, M. M., Rea, D. W., & Kleijnen, J. (2010).
tion of 5 years’ adjuvant treatment for breast cancer. Systematic review of aromatase inhibitors in the first-
Lancet, 365, 60–62. line treatment for hormone sensitive advanced or met-
Humphries, M. P., Sundara Rajan, S., Honarpisheh, H., astatic breast cancer. Breast Cancer Research and
Cserni, G., Dent, J., Fulford, L., Jordan, L. B., Treatment, 123, 9–24.
Jones, J. L., Kanthan, R., Litwiniuk, M., Shaaban, A. M., Ball, G. R., Brannan, R. A.,
Di Benedetto, A., Mottolese, M., Provenzano, E., Cserni, G., Di Benedetto, A., Dent, J., Fulford, L.,
Shousha, S., Stephens, M., Kulka, J., Ellis, I. O., Honarpisheh, H., Jordan, L., Jones, J. L., Kanthan, R.,
Titloye, A. N., Hanby, A. M., Shaaban, A. M., & Maraqa, L., Litwiniuk, M., Mottolese, M., Pollock, S.,
Speirs, V. (2017). Characterisation of male breast Provenzano, E., Quinlan, P. R., Reall, G., Shousha, S.,
cancer: A descriptive biomarker study from a large Stephens, M., Verghese, E. T., Walker, R. A.,
patient series. Scientific Reports, 7, 45293. Hanby, A. M., & Speirs, V. (2012). A comparative bio-
Lee, A. V., Cui, X., & Oesterreich, S. (2001). Cross-talk marker study of 514 matched cases of male and female
among estrogen receptor, epidermal growth factor, and breast cancer reveals gender-specific biological differences.
insulin-like growth factor signaling in breast cancer. Breast Cancer Research and Treatment, 133, 949–958.
Clinical Cancer Research, 7(12 Suppl), 4429s–4435s; van de Ven, S., Smit, V. T., Dekker, T. J., Nortier, J. W., &
discussion 4411s–4412s. Kroep, J. R. (2011). Discordances in ER, PR and HER2
Levin, E. R., & Hammes, S. R. (2016). Nuclear receptors receptors after neoadjuvant chemotherapy in breast
outside the nucleus: Extranuclear signalling by steroid cancer. Cancer Treatment Reviews, 37, 422–430.
I
References and Further Reading tendency to recur in up to 25% of the cases and
with a slight risk of distant metastasis. This is
Flint, A., & Oberman, H. A. (1984). Infarction and squa- especially true if IMT typically arises in the lung
mous metaplasia of intraductal papilloma: A benign
and abdomen of children/adolescents (Coffin
breast lesion that may simulate carcinoma. Human
Pathology, 15, 764–767. and Fletcher 2013). As future studies are needed
Han, B., Zhang, H., Jiang, P., Zheng, C., Bi, L., Lu, L. U., to better define the boundaries between a reac-
& Fan, Z. (2015). Breast infarction during pregnancy tive and a neoplastic process, the combined
and lactation: A case report. Experimental and Thera-
diagnosis of “inflammatory pseudotumor/
peutic Medicine, 10, 1888–1892.
Hasson, J., & Pope, C. H. (1961). Mammary infarcts asso- inflammatory myofibroblastic tumor,” specify-
ciated with pregnancy presenting as breast tumors. ing the presence or not of ALK-1 expression
Surgery, 49, 313–316. (see below), seems actually to be more appropri-
Isenberg, J. S., Tu, Q., & Rainey, W. (1996). Mammary
ate for these lesions.
gangrene associated with warfarin ingestion. Annals of
Plastic Surgery, 37, 553–555.
Clinical Features
Inflammatory Myofibroblastic Tumor of the Breast, Fig. 1 (a) Proliferation of bland-looking spindle cells with
fascicular and swirling growth patterns. (b) Higher magnification showing the cytological details
expression of ALK-1 protein has been demon- and/or ALK gene rearrangement does not seem
strated in three cases (Zhou et al. 2013; Bosse to predict clinical course. In this regard, only a
et al. 2014), proving that, at least, a subset of few cases, regardless of ALK-1 expression and/or
these lesions could represent true IMT. Occasion- ALK rearrangements, develop local recurrence or
ally immunostaining for cytokeratins has been more rarely distant metastases (Zhao et al. 2013;
described. No immunoreactivity is reported for Bosse et al. 2014; Choi et al. 2015; Markopoulos
CD34, CD21, CD35, S100, and estrogen/proges- et al. 2015; Talu et al. 2016).
terone receptors (Table 1).
The role of ALK gene is still unclear in the path- The diagnosis of IMT/IP arising in the breast is
ogenesis of IMT. The distinction between IMT challenging, especially on a core needle biopsy.
and IP still remains arbitrary, especially in the The pathologists facing with a bland-looking
absence of a proven immunohistochemical spindle cell lesion of the breast should keep in
expression of ALK-1 or rearrangements of ALK mind a wide variety of benign and malignant
gene by means of FISH. Some investigators prefer lesions, including reactive spindle cell nodule,
the diagnosis of IP when dealing with ALK- nodular fasciitis, IgG4-related sclerosing mastitis,
negative lesions arising at unusual sites, including myofibroblastoma (▶ Mammary Myofibro-
the breast, of adult patients (Vecchio et al. 2011). blastoma), solitary fibrous tumor, desmoid-type
Among the cases of IMT/IP of the breast tested by fibromatosis, low-grade (fibromatosis-like) spin-
FISH, only four cases have been found to be dle cell carcinoma (▶ Invasive Metaplastic Carci-
ALK-rearranged. Notably ALK1 expression noma), low-grade myofibroblastic sarcoma, and
172 Inflammatory Myofibroblastic Tumor of the Breast
Inflammatory Myofibroblastic Tumor of the Breast, some areas the spindle cells may exhibit moderate nuclear
Fig. 2 Proliferating spindle cells are closely admixed with atypia (c) (H&E). Immunostaining with alpha-smooth
lymphocytes (a) (H&E) and plasma cells (b) (H&E). In muscle actin is a common finding (d)
eosinophilic spindle cells arranged in short, hap- Finally, dermatofibrosarcoma protuberans may
hazardly intersecting fascicles with interspersed share with IMT/IP the storiform/swirling growth
keloid-like collagen fibers. Unlike IMT/IP, pattern which can be focally encountered in the
myofibroblastoma contains mast cells but not latter. Unlike IMT/IP, dermatofibrosarcoma pro-
lymphocytes, plasma cells, or eosinophils in the tuberans exhibits a diffuse storiform pattern and is
stroma. In addition myofibroblastoma usually diffusely positive for CD34, while myogenic
co-expresses diffusely CD34, desmin, and markers (desmin, a-smooth muscle actin) are
a-smooth muscle actin, while ALK1 immunore- absent.
activity is absent.
In contrast to IMT/IP, solitary fibrous tumor
characteristically shows short fibroblast-like cells References and Further Reading
haphazardly arranged and medium-sized blood
vessels with hyalinized walls and branching con- Bosse, K., Ott, C., Biegner, T., Fend, F., Siegmann-Luz,
K., Wallwiener, D., & Hahn, M. (2014). 23-year-old
figuration. In addition solitary fibrous tumor is
female with an inflammatory Myofibroblastic tumour
diffusely positive for CD34 and STAT6, which of the breast: A case report and a review of the
are not expressed in IMT/IP. literature. Geburtshilfe und Frauenheilkunde, 74,
wDesmoid-type fibromatosis usually exhibits 167–170.
Cheuk, W., Chan, A. C., Lam, W. L., Chow, S. M.,
extensive infiltrative margins with fingerlike pro-
Crowley, P., Lloydd, R., Campbell, I., Thorburn, M.,
jections into the adjacent fibro-fatty tissue and & Chan, J. K. (2009). IgG4-related sclerosing mastitis:
breast parenchyma. The diagnosis of desmoid- Description of a new member of the IgG4-related scle-
type fibromatosis is based on the presence of rosing diseases. The American Journal of Surgical
Pathology, 33, 1058–1064.
long, sweeping fascicles set in a variable fibrous
Choi, E. J., Jin, G. Y., Chung, M. J., Moon, W. S., &
stroma. Spindle cells are aligned parallel and their Youn, H. J. (2015). Primary inflammatory
nuclei are spaced, without overlapping. Inflamma- myofibroblastic tumors of the breast with metastasis:
tory cells admixed with the proliferating spindle Radiographic and histopathologic predictive factors.
Journal of Breast Cancer, 18, 200–205.
cells are lacking in desmoid-type fibromatosis.
Coffin, C. M., & Fletcher, J. A. (2013). Inflammatory
Although desmoid-type fibromatosis may myofibroblastic tumour. In C. D. M. Fletcher, J. A.
share with IMT/IP the immunohistochemical Bridge, P. C. W. Hogendoorn, & F. Mertens (Eds.),
expression of a-smooth muscle actin, the former WHO classification of tumours of soft tissue and bone
(4th ed., pp. 83–84). Lyon: WHO Press.
is typically positive for b-catenin in about 80% of
Haj, M., Weiss, M., Loberant, N., & Cohen, I. (2003).
cases. Inflammatory pseudotumor of the breast: Case report
Low-grade (fibromatosis-like) spindle cell car- and literature review. The Breast Journal, 9, 423–425.
cinoma (▶ Invasive Metaplastic Carcinoma) is Markopoulos, C., Charalampoudis, P., Karagiannis, E.,
Antonopoulou, Z., & Mantas, D. (2015). Inflammatory
included in the differential diagnosis because neo-
myofibroblastic tumor of the breast. Case Reports in
plastic cells adopt a spindle cell morphology. Surgery, 2015, 705127.
Detection of epithelioid-polygonal cells arranged Talu, C. K., Çakır, Y., Hacıhasanoğlu, E., Leblebici, C.,
in small cohesive clusters, in addition to spindle Aksoy, Ş., & Nazlı, M. A. (2016). Inflammatory
myofibroblastic tumor of the breast coexisting with
cell component, and immunoreactivity for
pseudoangiomatous stromal hyperplasia. The Journal
cytokeratins and myoepithelial markers, espe- of Breast Health, 12, 171–173.
cially p63, supports the diagnosis of carcinoma. Vecchio, G. M., Amico, P., Grasso, G., Vasquez, E., La
Low-grade myofibroblastic sarcoma shares with Greca, G., & Magro, G. (2011). Post-traumatic inflam-
matory pseudotumor of the breast with atypical mor-
IMT/IP the expression of a-smooth muscle actin.
phological features: A potential diagnostic pitfall.
However, its presentation (nodule with well- Report of a case and a critical review of the literature.
circumscribed margins); presence, at least focally, Pathology, Research and Practice, 207, 322–326.
of moderate nuclear atypia; and high mitotic count Zhao, H. D., Wu, T., Wang, J. Q., Zhang, W. D., He, X. L.,
Bao, G. Q., Li, Y., Gong, L., & Wang, Q. (2013).
(from 7 to 35 mitoses per 10 HPF), along with
Primary inflammatory myofibroblastic tumor of the
absence of inflammatory cells, favor the diagnosis breast with rapid recurrence and metastasis: A case
of low-grade myofibroblastic sarcoma. report. Oncology Letters, 5, 97–100.
Intraductal Papillary Carcinoma 175
Zhou, Y., Zhu, J., Zhang, Y., Jiang, J., & Jia, M. (2013). • Incidence
An inflammatory myofibroblastic tumour of the breast The incidence of DCIS has dramatically
with ALK overexpression. BMJ Case Reports,
2013. https://doi.org/10.1136/bcr-07-2011-4474, pii: increased since the introduction of population-
bcr0720114474. based breast cancer screening. Nowadays,
approximately 20–25% of all breast neoplastic
lesions are DCIS. A pure or predominant papil-
lary growth pattern is however rare.
Intraductal Papillary • Age
Carcinoma The reported age range of the patients with
malignant papillary lesions in general is
Emma Josephine Groen and Jelle Wesseling 27–89 years with a mean age of 60 (Tse 2005).
Department of Pathology, The Netherlands • Sex
Cancer Institute-Antoni van Leeuwenhoek Predominantly in women.
Hospital, Amsterdam, The Netherlands • Site
The vast majority is asymptomatic and
is detected by screening mammography. Symp-
Synonyms toms such as nipple discharge, Paget’s disease of
I
the nipple, or a palpable mass sometimes occur.
Noninvasive papillary carcinoma; Papillary duc- There is no preferential site for intraductal pap-
tal carcinoma in situ illary carcinoma.
• Treatment
Treatment is similar to DCIS in general. Cur-
Definition rently almost all lesions are being treated to
prevent the development of breast cancer.
In literature, an unambiguous definition and uni- Breast conserving treatment followed by radio-
form criteria of intraductal papillary carcinoma therapy or in case of extensive lesions a mas-
is lacking. Often, it is described as a luminal tectomy with or without immediate
epithelial neoplastic proliferation of the breast reconstruction are generally recommended.
confined to the mammary ducts and lobules There is no consensus on the value of adjuvant
(also known as ductal carcinoma in situ, DCIS hormonal treatment.
(▶ Ductal Carcinoma In Situ)), characterized by • Outcome
a papillary growth pattern. It is considered a The natural course of intraductal papillary car-
potential precursor lesion for breast cancer. cinoma or papillary DCIS is largely unknown,
Whether these lesions represent de novo malig- as almost all lesions are treated. Fifteen years
nant papillary epithelial proliferations or malig- after DCIS diagnosis, including all subtypes,
nant epithelial populations transforming and cumulative incidence of ipsilateral breast can-
replacing benign papillomas (▶ Intraductal Pap- cer was 1.9% after mastectomy, 8.8% after
illoma) remains unclear. breast conservative surgery (BCS) plus radio-
therapy, and 15.4% after BCS alone (Elshof
et al. 2016).
Clinical Features Despite the increased detection and treat-
ment of these potential precursor lesions for
As intraductal papillary carcinoma is a morpho- breast cancer, the incidence of breast cancer
logical subtype of DCIS, the following features has not declined, suggesting overdiagnosis
are derived from literature based on DCIS in gen- exists resulting in overtreatment. Unfortu-
eral, combined with data specifically focusing on nately, we cannot, at present, distinguish inno-
intraductal papillary carcinoma (also known as cent from aggressive DCIS lesions regarding
papillary DCIS). their capacity to progress to invasive breast
176 Intraductal Papillary Carcinoma
cancer. Therefore, biomarker studies in order and Neubecker 1962; Yamaguchi et al. 2014).
to enable risk stratification and prospective The neoplastic epithelium often consists of one
trials investigating the safety of a wait-and- or more layers of uniform cuboidal to columnar
see strategy for low-risk ductal carcinoma in epithelium with hyperchromatic nuclei of low
situ have emerged. to intermediate grade (Fig. 1b, Fig. 2-plate A2).
Myoepithelial cells are absent within the fibrovas-
cular cores but are seen at the periphery of the
Macroscopy
involved spaces, in line with an in situ process. It
should be noted that a pure papillary architecture
As DCIS in general, intraductal papillary
is rarely seen; often it is seen intermixed with
carcinoma is a segmental disease that can be asso-
other growth patterns, such as micropapillary,
ciated with an impeccable breast parenchyma.
cribriform, or solid DCIS.
Occasionally, dilated mammary ducts filled with
necrotic debris and calcifications with or without
Pitfalls
fibrosis can be seen.
In some cases, intraductal papillary carcinoma
constitutes of a dimorphic epithelial cell popula-
tion, in which an often basally located second
Microscopy population of cells with abundant pale cytoplasm
(“globoid cells”) is seen. These cells may be
Intraductal papillary carcinoma is characterized incorrectly interpreted as myoepithelial cells
by a segmental disease distribution. Involved (Collins and Schnitt 2007). Immunohistochemical
spaces show intraluminal arborizing fibrovascular staining for epithelial and myoepithelial cells can
cores covered by neoplastic epithelium (Fig. 1a, resolve this issue and confirm the epithelial nature
Fig. 2-plate A1). In 1962, delicate and relatively of these cells.
inconspicuous fibrovascular cores were described Dislodgement and displacement of fragments
as characteristic for malignant lesions such as from papillary lesions into the surrounding
intraductal papillary carcinoma (Kraus and breast stroma or even within lymphatic vascular
Neubecker 1962). More recently, however, others spaces, especially after a biopsy procedure, is
have refuted this statement, as they found broad a well-known occurrence and should be
fibrovascular cores even more often in malignant interpreted with caution (Collins and Schnitt
papillary lesions and concluded this morphologic 2007; Kraus and Neubecker 1962; Ni and Tse
feature was not a helpful feature to distinguish 2015). In case of displacement into the surround-
between benign and malignant lesions (Kraus ing breast stroma, epithelial components with
Intraductal Papillary Carcinoma, Fig. 1 Intraductal papillary carcinoma, haemotoxylin-eosin stain (H&E),
(a) magnification 25; (b) magnification 200
Intraductal Papillary Carcinoma 177
Intraductal Papillary Carcinoma, Fig. 2 (a) Intraductal involved space; B4 – ER 400, heterogeneous ER expres-
papillary carcinoma: A1 – H&E 50; A2 – H&E 400; sion (mosaic pattern). (c) Encapsulated papillary carci-
A3 – p63 400, showing presence of myoepithelial cells at noma: C1 – H&E 50; C2 – H&E 400; C3 – p63
the periphery of the involved space but not within the 400, showing no myoepithelial cells; C4 – ER 400,
fibrovascular structure; A4 – ER 400, strong diffuse strong diffuse homogeneous ER expression. (d) Solid
homogeneous ER expression. (b) Intraductal benign pap- papillary carcinoma: D1 – H&E 50; D2 – H&E 400;
illoma: B1 – H&E 50; B2 – H&E 400; B3 – p63 400, D3 – p63 400, showing sporadic myoepithelial cells at
showing presence of myoepithelial cells both within the the periphery of the nodules; D4 – ER 400, strong diffuse
fibrovascular structures and at the periphery of the homogeneous ER expression
some degree of degenerative changes are present fat necrosis, and some degree of inflammation.
within the confines of the biopsy tract with Invasion should only be considered when epithe-
concomitant reactive changes such as reactive lial structures are found well away from the
fibrous stroma, signs of associated hemorrhage, biopsy site and show morphological features
178 Intraductal Papillary Carcinoma
consistent with invasive cancer. Likewise, the Whereas LOH at loci 16p13 and 16q21 could
presence of epithelial fragments in lymphatic be detected in malignant as well as paired benign
vascular spaces or even lymph nodes should papillary lesions as obtained from adjacent tissue,
be doubted when no convincing invasive cancer LOH at locus 16q23 and at the TP53 locus was
is found elsewhere within the breast. limited to malignant lesions, suggesting a role
Immunohistochemistry to verify the presence for the latter two in malignant transformation
or absence of myoepithelial cells in the distinction (Di Cristofano et al. 2005; Lininger et al. 1998).
between true invasion and displacement is only LOH at loci 16q12.2 and 16q21 was found
useful when myoepithelial cells can be detected, exclusively in malignant intraductal papillary
because often only the luminal epithelial cells lesions in core needle biopsies. Therefore, these
are displaced. Especially in intraductal papillary loci of LOH might be potential biomarkers in
carcinoma where myoepithelial cells are only the differential diagnosis with benign lesions
found at the periphery, immunohistochemistry is (Yoshida et al. 2012).
of limited value (Collins and Schnitt 2007; Ni and
Tse 2015).
Differential Diagnosis
a hyperplastic process (Fig. 2-plate B1, B2, B3). expression of neuroendocrine markers such as
A heterogeneous ER expression pattern is often synaptophysin and chromogranin A. Mucin pro-
seen (Fig. 2-plate B4), but homogeneous ER duction or even a mucinous invasive component
expression does not rule out benignancy, espe- can be seen in association with these lesions.
cially when epithelium with columnar cell type Myoepithelial cells can be present or absent
change is encountered (Ni and Tse 2015). Other at the periphery of these lesions while ER is
useful findings are the presence of apocrine meta- homogeneously expressed in epithelial cells
plasia within a papillary lesion and ▶ usual ductal (Fig. 2-plate D1, D2, D3, D4).
hyperplasia and ▶ sclerosing adenosis in the adja- The true nature of both these entities is uncer-
cent breast tissue, which are suggestive of tain. Historically, these lesions were considered
a benign papilloma (Collins and Schnitt 2007; variants of DCIS but given the often lack of
Kraus and Neubecker 1962). myoepithelial cells, these lesions may perhaps be
When an undoubtedly neoplastic epithelial better classified as low grade invasive carcinomas
population with papillary growth has been identi- with pushing growth or intermediates during the
fied one should keep in mind the following. progression from in situ to invasive breast cancer.
A recognizable architecture of a benign papilloma In the absence of a component of frank invasive
in some part of the lesion precludes the diagnosis growth or high-grade morphology, the outcome is
I
of intraductal papillary carcinoma and should excellent.
rather be considered an atypical papilloma or As described above, the presence of
a papilloma with DCIS depending on the myoepithelial cells both at the periphery
classification used. (DD encapsulated/solid papillary carcinoma)
and in the fibrovascular structures and/or cell
Encapsulated Papillary Carcinoma and Solid proliferation (DD benign papilloma) is a key
Papillary Carcinoma distinguishing feature. One should be careful
Encapsulated Papillary Carcinoma (▶ Encapsu- interpreting immunohistochemical stains for
lated Papillary Carcinoma) often have myoepithelial cells as both stromal fibroblasts
a symptomatic presentation with a subareolar and pericytes can show cross-reactivity, and all
mass and/or nipple discharge mostly occurring stains have their inherent sensitivity and specific-
in elderly women. Microscopically, one or several ity. A combination of more than one marker
nodules of often but not exclusively low to inter- is therefore advised. P63 is superior to other
mediate grade papillary neoplasms can be seen myoepithelial markers in the classification of pap-
originating in a cystically dilated duct surrounded illary lesions, as it is a nuclear stain with only
by a thick fibrous capsule (Collins and Schnitt minimal cross-reactivity and high sensitivity.
2007; Ni and Tse 2015). In contrast to intraductal For evaluating the nature of solid epithelial
papillary carcinoma, these lesions do not show proliferations often seen in papillary lesions,
a rim of myoepithelial cells at the periphery and cytokeratin 5/6 is recommended, as among the
ER is homogeneously and strongly expressed high molecular weight cytokeratins (HMWCKs),
(Fig. 2-plate C1, C2, C3, C4). it has the highest sensitivity and specificity
Solid papillary carcinoma (▶ Solid Papillary (Ni and Tse 2015; Tse et al. 2009).
Carcinoma) also tends to present in elderly Classifying papillary lesions on fine-needle
women as a circumscribed mass. As in aspiration is challenging and should generally
intraductal papillary carcinoma a papillary be discouraged, given the necessity to visualize
ground structure can be appreciated in solid pap- not only the presence of myoepithelial cells but
illary carcinoma. In the latter, however, fibrovas- also their exact location within the lesion.
cular cores are often completely surrounded by a
solid epithelial cell proliferation. These epithelial Immunohistochemical Biomarkers
cells often show ovoid to spindle bland cells and Several studies have evaluated the effectivity
can show neuroendocrine features, both morpho- of new immunohistochemical biomarkers to
logically and immunohistochemically, with the distinguish benign from malignant papillary
180 Intraductal Papilloma
lesions. The absence of staining for stem cell Loh, S. F., Cooper, C., Selinger, C. I., Barnes, E. H.,
markers CD44 and CD133 may be useful in iden- Chan, C., Carmalt, H., et al. (2015). Cell cycle marker
expression in benign and malignant intraductal papil-
tifying malignant papillary lesions. Positive CD44 lary lesions of the breast. Journal of Clinical Pathol-
expression has been proposed as a marker to dif- ogy, 68, 187–191. https://doi.org/10.1136/jclinpath-
ferentiate between benign and malignant papillary 2014-202331.
lesions with a reported sensitivity of 45% and Ni, Y.-B., & Tse, G. M. (2015). Pathological criteria and
practical issues in papillary lesions of the breast –
a specificity of 92% (Tse 2005) and CD133 A review. Histopathology, 68, 22–32. https://doi.org/
expression was significantly lower in papillary 10.1111/his.12866.
carcinomas than in benign or atypical papillomas Troxell, M. L., Levine, J., Beadling, C., Warrick, A.,
(p < 0.001) (Lin et al. 2014). Dunlap, J., Presnell, A., et al. (2009). High prevalence
of PIK3CA/AKT pathway mutations in papillary neo-
Cell cycle markers cyclin B1 and cyclin D1 plasms of the breast. Modern Pathology, 23, 27–37.
have been shown to be independently associated https://doi.org/10.1038/modpathol.2009.142.
with malignancy in papillary lesions. Positive Tse, G. M. K. (2005). CD44s is useful in the differentiation
staining for cyclin B1 and cyclin D1 was found of benign and malignant papillary lesions of the breast.
Journal of Clinical Pathology, 58, 1185–1188. https://
to be helpful for identifying malignant papillary doi.org/10.1136/jcp.2005.026906.
lesions with a sensitivity of 80% and a specificity Tse, G. M., Tan, P. H., & Moriya, T. (2009). The role of
of 72.7% for cyclin B1 and a sensitivity of 86.4% immunohistochemistry in the differential diagnosis of
but only a specificity of 32.6% for cyclin D1 (Loh papillary lesions of the breast. Journal of Clinical
Pathology, 62, 407–413. https://doi.org/10.1136/
et al. 2015). The expression was however fre- jcp.2008.063016.
quently heterogeneous and only focal, limiting Tsuda, H., Takarabe, T., Inazawa, J., &
its usefulness in clinical practice. Hirohashi, S. (1997). Detection of numerical alterations
of chromosomes 3, 7, 17 and X in low-grade intracystic
papillary tumors of the breast by multi-color fluores-
References and Further Reading cence in situ hybridization. Breast Cancer, 4, 247–252.
Yamaguchi, R., Tanaka, M., Tse, G. M., Yamaguchi, M.,
Collins, L. C., & Schnitt, S. J. (2007). Papillary lesions of Terasaki, H., Nomura, Y., et al. (2014). Broad
the breast: Selected diagnostic and management issues. fibrovascular cores may not be an exclusively benign
Histopathology, 52, 20–29. https://doi.org/10.1111/ feature in papillary lesions of the breast: A cautionary
j.1365-2559.2007.02898.x. note. Journal of Clinical Pathology, 67, 258–262.
Di Cristofano, C., Mrad, K., Zavaglia, K., Bertacca, G., https://doi.org/10.1136/jclinpath-2013-201749.
Aretini, P., Cipollini, G., et al. (2005). Papillary lesions Yoshida, M., Tsuda, H., Yamamoto, S., Kinoshita, T., Akashi-
of the breast: A molecular progression? Breast Cancer Tanaka, S., Hojo, T., & Fukutomi, T. (2012). Loss of
Research and Treatment, 90, 71–76. https://doi.org/ heterozygosity on chromosome 16q suggests malignancy
10.1007/s10549-004-3003-3. in core needle biopsy specimens of intraductal papillary
Elshof, L. E., Schaapveld, M., Schmidt, M. K., breast lesions. Virchows Archiv, 460, 497–504. https://doi.
Rutgers, E. J., Leeuwen, F. E., & Wesseling, org/10.1007/s00428-012-1200-8.
J. (2016). Subsequent risk of ipsilateral and contralat-
eral invasive breast cancer after treatment for ductal
carcinoma in situ: Incidence and the effect of
radiotherapy in a population-based cohort of 10,090
women. Breast Cancer Research and Treatment, 159, Intraductal Papilloma
553–563. https://doi.org/10.1007/s10549-016-3973-y.
Kraus, F. T., & Neubecker, R. D. (1962). The differential Ales Ryska and Folakemi A. Torgersen
diagnosis of papillary tumors of the breast. Cancer, 15,
444–455. The Fingerland Department of Pathology, Charles
Lin, C.-H., Liu, C.-H., Wen, C.-H., Ko, P.-L., & Chai, C.-Y. University Medical Faculty and University
(2014). Differential CD133 expression distinguishes Hospital, Hradec Kralove, Czech Republic
malignant from benign papillary lesions of the breast.
Virchows Archiv, 466, 177–184. https://doi.org/
10.1007/s00428-014-1695-2.
Lininger, R. A., Park, W. S., Man, Y. G., Pham, T., Synonyms
MacGrogan, G., Zhuang, Z., & Tavassoli, F. A.
(1998). LOH at 16p13 is a novel chromosomal alter- Ductal adenoma; Large duct papilloma; Major
ation detected in benign and malignant microdissected
papillary neoplasms of the breast. Human Pathology, duct papilloma; Microscopic papilloma; Small
29, 1113–1118. duct papilloma
Intraductal Papilloma 181
Macroscopy
Microscopy
Intraductal Papilloma, Fig. 2 (H&E) Intraductal papil-
Cytologically, smears from papillomas are loma with complex structure and signs of bleeding
moderately cellular. The fronds of intraductal
papillomas show sharp delineation and cells
in tissue fragments are more cohesive. The papil- hyperchromasia (O’Malley et al. 2012). Regressive
lae feature fibrovascular stalks lined by changes may frequently occur in papillomas, mak-
columnar cells. Frequently, epithelial cells ing them difficult to assess when only a limited
arranged in a honeycomb pattern are seen. amount of tissue is available for histological exam-
Alternatively, columnar cells can be arranged ination (▶ Infarct of Breast Tissue). Such changes
in small papillae and/or can be seen isolated. include bleeding and infarction. These are typically
The presence of apocrine metaplasia and seen in large central lesions and may occur as a
bipolar naked nuclei is very frequent (Gomez- result of torsion of fibrovascular cores or prior
Aracil et al. 2002). sampling. When no residual vital tissue is present,
Histologically intraductal papillomas are the lesion should be simply called an infarcted
characterized by the presence of a complex papillary lesion (Wei 2016).
arborizing structure comprised of fibrovascular Stromal fibrosis or sclerosis is another feature
cores lined by myoepithelial (basally located) commonly associated with intraductal papillomas.
and epithelial (luminal) cell layers (Fig. 1 The sclerosis may distort the tissue to such an
(H&E) (Fig. 2 (H&E). The epithelial cell nuclei extent that a pseudo-infiltrative pattern is observed
vary in shape and size but are lacking (Fig. 3 (H&E). In these sclerosing intraductal
Intraductal Papilloma 183
Intraductal Papilloma, Fig. 3 (H&E) Pseudo-infiltra- Intraductal Papilloma, Fig. 4 (H&E) Peripheral papil-
tive pattern in intraductal papilloma with stromal sclerosis loma with apocrine metaplasia and microcalcifications in
adjacent TDLU
Molecular Features
(Nagi et al. 2005). This phenomenon – although Shiino, S., Tsuda, H., Yoshida, M., Jimbo, K., Asaga, S.,
relatively rare – is well known and represents an Hojo, T., & Kinoshita, T. (2015). Intraductal papillo-
mas on core biopsy can be upgraded to malignancy on
important diagnostic pitfall. Displaced benign subsequent excisional biopsy regardless of the presence
epithelial structures are usually represented by of atypical features. Pathology International, 65,
small isolated clusters of cells present in the 293–300.
stroma or lymphatic channels. Unlike truly inva- Tse, G. M., Tan, P. H., & Moriya, T. (2009). The role of
immunohistochemistry in the differential diagnosis of
sive nests of neoplastic cells, displaced epithelial papillary lesions of the breast. Journal of Clinical
clusters are always located within the biopsy tract Pathology, 62, 407–413.
and lack desmoplastic stromal reaction (Ueng >Ueng, S. H., Mezzetti, T., & Tavassoli, F. A. (2009).
et al. 2009). Though the biologic significance of Papillary neoplasms of the breast: A review. The
Archives of Pathology and Laboratory Medicine, 133,
epithelial displacement is unknown, preliminary 893–907.
studies suggest that displaced cells most probably Volckmar, A. L., Leichsenring, J., Flechtenmacher, C.,
do not survive the preceding trauma or eventually Pfarr, N., Siebolts, U., Kirchner, M., Budczies, J.,
die as time progresses (Nagi et al. 2005). Bockmayr, M., Ridinger, K., Lorenz, K., Herpel, E.,
Noske, A., Weichert, W., Klauschen, F., Schirmacher,
P., Penzel, R., Endris, V., & Stenzinger, A. (2017).
Tubular, lactating, and ductal adenomas are devoid
of MED12 Exon2 mutations, and ductal
References and Further Reading adenomas show recurrent mutations in GNAS and I
the PI3K-AKT pathway. Genes, Chromosomes &
Bianchi, S., Bendinelli, B., Saladino, V., Vezzosi, V., Cancer, 56, 11–17.
Brancato, B., Nori, J., & Palli, D. (2015). Non- Wei, S. (2016). Papillary lesions of the breast: An update.
malignant breast papillary lesions – b3 diagnosed on The Archives of Pathology and Laboratory Medicine,
ultrasound – guided 14-gauge needle core biopsy: 140, 628–643.
Analysis of 114 cases from a single institution and
review of the literature. Pathology and Oncology
Research, 21, 535–546.
Cserni, G. (2012). Benign apocrine papillary lesions of the
breast lacking or virtually lacking myoepithelial cells- Invasive Carcinoma NST
potential pitfalls in diagnosing malignancy. Acta
Pathologica, Microbiologica, et Immunologica
Scandinavica, 120, 249–252. Ian Ellis
Cyr, A. E., Novack, D., Trinkaus, K., Margenthaler, J. A., Department of Histopathology, City Hospital
Gillanders, W. E., Eberlein, T. J., Eberlein, T. J., Ritter, Campus, Nottingham University Hospitals,
J., & Aft, R. L. (2011). Are we overtreating papillomas Nottingham City Hospital, Nottingham, UK
diagnosed on core needle biopsy? Annals of Surgical
Oncology, 18, 946–951.
Gomez-Aracil, V., Mayayo, E., Azua, J., & Arraiza, A.
(2002). Papillary neoplasms of the breast: Clues in Synonyms
fine needle aspiration cytology. Cytopathology, 13,
22–30.
MacGrogan, G., & Tavassoli, F. A. (2003). Central atypical Carcinoma simplex; Ductal carcinoma NST; Infil-
papillomas of the breast: A clinicopathological study of trating ductal carcinoma; Invasive carcinoma of
119 cases. Virchows Archiv, 443, 609–617. no specific type (NST); Invasive ductal carcinoma
Nagi, C., Bleiweiss, I., & Jaffer, S. (2005). Epithelial not otherwise specified (ductal NOS); Scirrhous
displacement in breast lesions: A papillary phenome-
non. The Archives of Pathology and Laboratory Med- carcinoma; Spheroidal cell carcinoma
icine, 129, 1465–1469.
Ni, Y. B., & Tse, G. M. (2016). Pathological criteria and
practical issues in papillary lesions of the breast – a
review. Histopathology, 68(1), 22–32. https://doi.org/ Definition
10.1111/his.12866.
O’Malley, F., Visscher, D., MacGrogan, G., & Tan, P. H. Invasive breast carcinoma of no special type
(2012). Intraductal papilloma. In S. R. Lakhani, I. O.
is also commonly known as invasive ductal
Ellis, S. J. Schnitt, P. H. Tan, & M. J. van de Vijver
(Eds.), WHO classification of tumours of the breast carcinoma not otherwise specified, comprises
(pp. 100–102). Lyon: IARC Press. the largest group of invasive breast cancers. It
186 Invasive Carcinoma NST
is a morphologically heterogeneous group which the tumor including the histological grade
does not exhibit the features or purity of tumors (or growth fraction), size, lymph node stage,
of recognized special types of breast cancer, vascular invasion, hormone receptor, and
such as classical invasive lobular (▶ Invasive HER2 receptor status of the cancer. Treatments
Lobular Carcinoma) or tubular carcinoma are more aggressive when the prognosis is
(▶ Tubular Carcinoma). worse or there is a higher risk of recurrence of
the cancer following treatment. Surgery with or
without radiation therapy is the usual initial
Clinical Features treatment in early stage disease, which may
be followed by adjuvant systemic therapy
• Incidence such as endocrine therapy, chemotherapy, and
Invasive carcinoma NST forms a large propor- targeted anti HER2 therapy where appropriate.
tion of mammary carcinomas and its epidemio- Approximately, 70–80% of NST breast can-
logical characteristics are similar to those of cers are estrogen receptor (ER)-positive and
breast cancer as a whole. The WHO currently 12–20% of cases are HER2 positive.
identifies that breast cancer is the most frequent Neoadjuvant chemotherapy (NACT) has
cancer among women, impacting 2.1 million been shown to be effective in downstaging
women each year, and also causes the greatest breast cancer and is now commonly used as
number of cancer-related deaths among women. primary therapy (Early Breast Cancer Trialists’
In 2018, it is estimated that 627,000 women die Collaborative Group 2018). NACT for early
from breast cancer – that is approximately 15% breast cancer can make breast-conserving sur-
of all cancer deaths among women. While gery more feasible. Patients with high-grade,
breast cancer rates are higher among women in hormone receptor-negative tumors are most
more developed regions, rates are increasing in likely to achieve a complete clinical response
nearly every region globally. of the primary tumor after NACT. Pathological
• Age complete response (pCR) of the axilla can
It is rare below the age of 40 years, but be achieved in 41–75% of patients with
the proportion of tumors classified as such in HER2-positive or triple-negative cancer
young women with breast cancer is in general receiving NACT.
similar to that in older women. • Outcome
• Sex Outcomes for breast cancer vary depending on
Breast cancer of NST is a disease predomi- the extent of disease, prognostic characteristics
nantly of females with male breast cancer and response to treatment. Survival rates have
being very rare. Less than 1% of all breast improved significantly in recent years due to
cancer cases develop in men, and only one in multiple variables including early detection,
a thousand men will ever be diagnosed with screening and an increasing range of effective
breast cancer. treatment options. Survival rates in the devel-
• Site oped world are now high, with between 80%
Breast cancer of NST occurs in the parenchyma and 90% of those in Europe and the United
of the breast or very rarely in extra mammary States alive for at least 5 years. In developing
ectopic breast tissue. Approximately 50% of countries survival rates are poorer.
breast cancers arise in the upper outer quadrant Tools and methods have been developed to
of the breast, 15% in the inner upper quadrant, assist in predicting patient outcome and to sup-
and <10% in the inner lower and outer lower port clinical decision making in breast cancer
quadrants respectively. management. Examples of such methods
• Treatment include the Nottingham Prognostic Index (NPI)
The management of breast cancer of NST (Blamey et al. 2007), St Gallen consensus
depends on the prognostic characteristics of criteria, the National Comprehensive Cancer
Invasive Carcinoma NST 187
Network (NCCN) guidelines, and Predict in this chapter illustrate examples of NST histo-
(https://www.predict.nhs.uk). logical type, please note the ranges of non
The NPI is based on a combination of specific appearances). In many of cases, glandular
histopathological examination of tumor size, differentiation may be apparent as tubular or
lymph node stage, and tumor grading assem- acinar structures with central lumina formed by
bled in a prognostic index formula. Prognosis the tumor cell population. Occasionally,
worsens as the NPI numerical value increases areas with single-file infiltration or targetoid
and by using cut-off points patients may be features mimicking invasive lobular carcinoma
stratified into good, moderate and poor prog- (▶ Invasive Lobular Carcinoma) occur but
nostic groups. The NPI has been confirmed lacking its distinct cytomorphological character-
after long-term follow-up, validated indepen- istics. NST carcinoma cells have a variable
dently in large multi-center studies, and revised appearance. Their cytoplasm may be abundant
in order to stratify patients into 5 prognostic and eosinophilic. Nuclei may be regular and uni-
groups (Blamey et al. 2010). form or highly pleomorphic with prominent, often
multiple, nucleoli. Mitotic activity ranges from
virtually absent to frequent. In up to 80% of
Macroscopy
cases, foci of associated ductal carcinoma in situ
I
(DCIS) (▶ Ductal Carcinoma In Situ) will be pre-
Due to their morphological, biological, and
sent. Any associated DCIS is usually of same
behavioral diversity, these tumors have no specific
nuclear grade as the invasive carcinoma. Occa-
macroscopic features. They can range in size from
sionally lobular neoplasia, either ALH or LCIS
<10 mm to >100 mm. They can have a rounded,
may be present (▶ Lobular In Situ Neoplasia).
irregular, stellate, diffuse, or nodular configura-
The stromal component is extremely variable.
tion. The invasive border is variable in appearance
There may be a highly cellular fibroblastic prolif-
but usually moderately or ill-defined and lacks
eration, a scanty element of connective tissue or
sharp circumscription. Classically, NST carcino-
marked hyalinization. Foci of elastosis may also
mas form a mass lesion and are firm or even hard
be present in a periductal or perivenous distribu-
on palpation, and may have a “gritty” feel when
tion. Focal necrosis may be present and this is
cut with a blade. The cut surface appearance is
occasionally extensive with secondary formation
varied but is usually pale and grey to cream white.
of cysts. In a minority of cases, a distinct
lymphoplasmacytoid infiltrate can be identified.
Vascular tumor emboli can be observed within or
Microscopy adjacent to the tumor.
Invasive Carcinoma NST, Fig. 1 (H&E) (a, b, c) Exam- morphological appearances seen in the NST tumor type.
ples of three invasive carcinomas of NST type showing no Please note that all the figures in this chapter are of NST
specific characteristics and a range of morphological tumors, with the exception of Fig. 2 which shows a tumour
appearances. Note the lack of special characteristic of mixed type with both NST and Tubular features
exhibited by each tumor and also the range of
will fall into one of the mixed groups: carcinoma, carcinoma with osteoclast-like stro-
mixed NST and special type (Fig. 2) or mal giant cells, carcinoma with choriocarci-
mixed NST and lobular carcinoma. Apart nomatous features, and carcinoma with
from these considerations, there are very melanotic features. There is also a current debate
few lesions that should be confused with NST whether to include medullary like cancers within a
carcinomas. group of NST cancers with associated
lymphocyte-rich stroma rather than as a distinct
separate morphological type.
Rare Morphological Variants of NST
Carcinoma Pleomorphic carcinoma. The 2012 WHO
The 2012 WHO classification of breast tumors classification defines pleomorphic carcinoma
(Lakhani et al. 2012) recognizes a number of as a rare variant of high-grade NST carcinoma,
morphological forms of breast cancer that are which is characterized by a proliferation of
not currently recognized as distinct special types pleomorphic and bizarre, sometimes multi-
of invasive breast cancer but as variants of nucleated, tumor giant cells comprising
NST breast cancer. These include pleomorphic >50% of the tumor cells in a background
Invasive Carcinoma NST 189
Invasive Carcinoma NST, Fig. 2 (H&E) (a, b) An Note the presence of a normal lobular unit (b) for compar-
example of a tumor of mixed type with focal (>50% and ison of tumor cells’ nuclear size. The carcinoma has similar
<90%) tubular carcinoma characteristics with background sized nuclei and score 1 for nuclear pleomorphism. There
(<50%) non specialised, NST, features. Please note that is marked tubule/gland formation, score 1 and no visible
this tumour shows grade 1 histological characteristics. mitoses, score 1, giving a total score of 3 (grade 1)
I
Additional Classification Systems Ellis 1991). The method has now been shown to
have good reproducibility in other centres and it
Histological Grade has been adopted for use in the pathological data
One of the most fundamental aspects of oncological set of the United Kingdom NHS BSP and in the
pathology, which has undoubtedly stood the test of USA and Europe (Rakha et al. 2010). Examples
time, has been the recognition that the detailed of grade 1, grade 2, and grade 3 carcinomas
morphological structure of tumors can be correlated are shown in Figs. 2a, b, 3, 4, and 5.
with their degree of malignancy. Greenhough in
1928, in Boston, USA, undertook the first formal Tumor Infiltrating Lymphocytes
study of the grading of breast cancer. Scarff and his It has been recognized that some types of breast
colleagues at the Middlesex Hospital in London cancer such as medullary like, NST, triple negative
re-examined Greenhough’s method and decided and HER2 positive, are associated with tumor-
that only three factors, tubule formation, nuclear infiltrating lymphocytes (TILs) (Fig. 6). Formal
pleomorphism, and hyperchromatism were of evaluation of TILs is gaining momentum as evi-
importance. Scarff’s method has formed the basis dence strengthens for the clinical relevance of phe-
of all subsequent grading systems. The perceived nomenon which is regarded as an immunological
poor reproducibility and consistency has been biomarker. The extent of lymphocytic infiltration in
largely resolved by use of semi objective scoring tumor tissue can be assessed as by evaluation of
systems and adherence to written criteria. These hematoxylin and eosin (H&E)-stained tumor sec-
studies have highlighted the need for grading to be tions and has been shown to provide prognostic
carried out by trained histopathologists who work to and potentially predictive value, particularly in the
an agreed protocol. triple-negative and HER 2 positive settings.
The Nottingham method, outlined in A standardized methodology for evaluating
Tables 1 and 2, is the most widely used method. TILs has been developed for visual assessment
Three characteristics of the tumor are evaluated on H&E sections (Salgado et al. 2015) by the
according to semi quantitative criteria, tubule International TILs Working Group. Their key rec-
formation as an expression of glandular differen- ommendations are:
tiation, nuclear pleomorphism and mitotic counts.
It has been validated through long term follow 1. TILs should be reported for the stromal com-
up of a large series of patients confirming partment (=% stromal TILs).
conclusively the highly significant relationship 2. TILs should be evaluated within the borders
between histological grade and prognosis; sur- of the invasive tumor.
vival worsens with increasing grade (Elston and 3. Exclude TILs outside of the tumor border and
around DCIS and normal lobules.
Invasive Carcinoma NST, Table 1 Semiquantitative
4. Exclude TILs in tumor zones with crush arti-
method for assessing histological grade in breast
carcinoma facts, necrosis, regressive hyalinization as
well as in the previous core biopsy site.
Feature Score
5. All mononuclear cells (including lymphocytes
Tubule formation
Majority of tumor (>75%) 1
and plasma cells) should be scored, but poly-
Moderate degree (10–75%) 2 morphonuclear leukocytes are excluded.
Little or none (<10%) 3 6. One section (4–5 mm, magnification
Nuclear pleomorphism 200–400) per patient is currently consid-
Small, regular uniform cells 1 ered to be sufficient.
Moderate increase in size and variability 2 7. Full sections are preferred over biopsies
Marked variation 3 whenever possible.
Mitotic counts 8. A full assessment of average TILs in the
Dependent on microscope field area 1–3 tumor area by the pathologist should be
(see Table 2) used. Do not focus on hotspots.
Invasive Carcinoma NST 191
Invasive Carcinoma NST, Table 2 Assignment of points for mitotic counts according to the field area
Number of mitoses corresponding to
Field diameter in mm Score 1 Score 2 Score 3
0.40 Up to 4 5–8 9 or more
0.41 Up to 4 5–9 10 or more
0.42 Up to 4 5–9 10 or more
0.43 Up to 4 5–10 11 or more
0.44 Up to 5 6–10 11 or more
0.45 Up to 5 6–11 12 or more
0.46 Up to 5 6–11 12 or more
0.47 Up to 5 6–12 13 or more
0.48 Up to 6 7–12 13 or more
0.49 Up to 6 7–13 14 or more
0.50 Up to 6 7–13 14 or more
0.51 Up to 6 7–14 15 or more
0.52 Up to 7 8–14 15 or more
0.53 Up to 7 8–15 16 or more
0.54 Up to 7 8–16 17 or more I
0.55 Up to 8 9–16 17 or more
0.56 Up to 8 9–17 18 or more
0.57 Up to 8 9–17 18 or more
0.58 Up to 9 10–18 19 or more
0.59 Up to 9 10–19 20 or more
0.60 Up to 9 10–19 20 or more
0.61 Up to 9 10–20 21 or more
0.62 Up to 10 11–21 22 or more
0.63 Up to 10 11–21 22 or more
0.64 Up to 11 12–22 23 or more
0.65 Up to 11 12–23 24 or more
0.66 Up to 11 12–24 25 or more
0.67 Up to 12 13–25 26 or more
0.68 Up to 12 13–25 26 or more
0.69 Up to 12 13–26 27 or more
0.70 Up to 13 14–27 28 or more
9. TILs should be assessed as a continuous low molecular weight cytokeratin, including CK’s
parameter. 7, 8 and 18. All of the major gene expression
10. No formal recommendation for a clinically molecular classes (luminal, basal/triple negative
relevant TIL threshold(s) can be given at and HER2 positive) are represented in the NST
this stage. group with the luminal ER positive tumors
predominating, approximately 70–80% of NST
breast cancers are ER-positive (▶ Hormone
Immunophenotype Receptors in Breast Cancer). The basal/triple neg-
ative for ER, progesterone receptor (PR)
Being a heterogeneous form of breast and HER2 group occur at a lower frequency
cancer tumors of NST type have not distinct (10–20%), as does the HER2 positive/ER positive
immunephenotypic characteristics. The majority and negative subgroups, with 12–20% of cases
of NST tumors are positive for GCDFP-15 and being HER2 positive (▶ HER2 in Breast Cancer).
192 Invasive Carcinoma NST
propose the collective term, “Carcinoma with BRCA1 gene mutations are also at increased
Medullary Features,” abbreviated to CMF for the risk of developing breast cancer. BRCA2
remainder of this chapter (Lakhani et al. 2012). mutations confer a higher risk of breast cancer
in men than in women.
• Site and Presentation
Clinical Features Patients may present with a discrete palpable
breast mass or may be diagnosed through
• Incidence mammographic screening, population based
Classical MC, as defined by Ridolfi et al. in or high-risk surveillance programs. CMF is
1977, is generally considered to be a rare breast typically well circumscribed and may have a
tumor accounting for <1% of all breast cancers soft consistency due to the lack of a stromal
(Ridolfi et al. 1977). Rates of up to 7% have desmoplastic reaction. In view of these fea-
been reported and this is likely attributable to tures and the relatively young age at presen-
interobserver and inter-institutional variation tation, these tumors may mimic benign
in the application of the histological criteria lesions both clinically and on imaging studies.
used to make the diagnosis. The overlying skin may be red. Bilateral syn-
In reported series, MC is less common chronous MCs have been reported, more com-
than atypical MC but the incidence is monly in patients with a positive family
largely unknown due to variation in defi- history.
nition and lack of and/or inconsistent coding CMF may be associated with a rapid growth
documentation. rate and may also come to medical attention as
CMF has a broader, more inclusive, defini- an interval screening breast cancer.
tion and includes a subset of tumors with med- Axillary lymph node involvement may occur
ullary characteristics that would previously but is relatively less common in CMF compared
have been categorized as invasive carcinoma with invasive carcinoma NST (▶ Invasive Car-
NST. It is difficult to obtain precise figures on cinoma NST). Concomitant enlarged axillary
overall incidence, but it is estimated that CMF lymph nodes are more likely to be reactive than
accounts for between 10% and 20% of invasive involved by metastatic CMF and presentation as
breast tumors (Hicks and Lester 2017). axillary lymph node involvement is rare (Hicks
• Age and Lester 2017). Presentation as distant metas-
The age at diagnosis of women with CMF ranges tases is also rare.
from 21 to 95 years, with an average age at • Treatment Surgery
presentation of between 45 and 50 years. The The primary goal of treatment is complete sur-
latter is approximately 10 years younger than the gical excision with clear margins. Conservation
average age profile of women diagnosed with surgery is clearly desirable and is feasible in
other types of invasive breast carcinoma. One most cases due to the absence of ductal carci-
in four diagnoses of CMF occurs in women noma in situ (▶ Ductal Carcinoma In Situ).
under the age of 35 years. This is likely to be Mastectomy may be necessary to treat large
due to the association of CMF with inherited and some centrally located tumors.
Breast Cancer susceptibility gene 1 (BRCA1) A diagnosis of CMF, in some countries, will
germline mutation (Eisinger et al. 1998). lead to genetic testing in women who have not
• Sex been previously tested. The results of this
CMF is predominantly reported in females in may lead to consideration of additional breast
line with all other forms of breast carcinoma. and other prophylactic surgery. This requires
The literature is scarce on histological tumor careful evaluation by the patient in consulta-
type in men. Most breast tumors in men are tion with her/his clinical team.
luminal A or triple negative. MC has been • Chemotherapy
reported in male patients. Male patients with
Invasive Carcinoma with Medullary Features 197
CMF is typically triple negative although cases was associated with a significantly higher sur-
of hormone receptor positive MC have been vival rate at 10 years, estimated at 84% for
reported. In view of the more favorable prog- patients with classical MC compared with
nosis associated with CMF, it has been 63% for patients with non-medullary invasive
suggested that patients with lymph node nega- breast carcinoma (Ridolfi et al. 1977). This
tive disease may possibly avoid chemotherapy. also pertained to patients with
In general, this tumor type tends to occur in positive axillary lymph nodes when the
younger women and the constellation of mor- two groups were compared. In addition to
phological features is usually associated with 57 patients with classical MC, the study popu-
triple negative status. At the present time it is lation included 79 patients with AMC and
recommended that adjuvant or neoadjuvant 56 with non-medullary carcinoma (invasive
chemotherapy treatment recommendations for carcinoma NST, using modern terminology).
patients with CMF should take account of the Patients with AMC also had an improved prog-
clinical, pathological, and biological character- nosis with a 10-year survival rate of 74% over-
istics as for all invasive breast cancers. PARP all. Interestingly, AMC with a sparse lymphoid
inhibitors and platinum agents are reported to infiltrate was associated with a relatively poor
be more effective in the treatment of BRCA1- prognosis. Apart from this observation, the
I
related tumors. authors were unable to draw any firm conclu-
• Immunotherapy sions regarding the prognostic effect of the
Several clinical trials are in progress to investi- other morphological features of AMC.
gate the efficacy of immunotherapy agents in In a more recent study, Mateo et al.
the treatment of triple negative breast carci- reviewed the outcome of patients diagnosed
noma (TNBC) (Dua and Tan 2017). This relates with MC (n = 3,688) and AMC (n = 288)
to the discovery that approximately 25% of registered with the National US Cancer Data-
TNBCs express the T-cell inhibitory molecule base (NCDB) during the period 2004 to 2013
PD-L1 (programmed death ligand-1), particu- (Mateo et al. 2016). Cases are reported to
larly in tumors with a high lymphocyte count. the NCDB using codes from the International
PDL1 is expressed on cancer cells, tumor infil- classification of diseases for Oncology
trating lymphocytes (TILs), and immune cells (ICD-O-3). During the study period 918,870
(Tung et al. 2016). The binding of PD-L1 to the cases of non-medullary breast carcinoma
PD-1 (programmed death 1) checkpoint recep- were registered. Over 20% of tumors diag-
tor on T cells is one mechanism for tumor nosed as MC or AMC were recorded as hor-
evasion of the immune response. As detailed mone receptor positive. The study did not
in subsequent sections, CMF is characterized include a review of histopathological slides
by a prominent inflammatory cell infiltrate with and the number of AMC tumors was rela-
a high TIL count. CMF is also associated with a tively small. Notwithstanding these limita-
high level of genomic instability leading to tions the authors found no significant
increased production of neoantigens and greater difference between MC and AMC in terms
immunogenicity (Weigelt et al. 2010). These of clinicopathological characteristics and
features suggest that CMF is likely to be a can- clinical outcome.
didate tumor type for targeted immunotherapy. Subsequent studies from several institutions
Adjuvant Radiotherapy: Recommenda- have also reported a more favorable outcome in
tions for post-surgical radiotherapy should be patients with MC compared with grade matched
made in accordance with those for other pri- invasive carcinoma NST. In a compilation of
mary invasive breast carcinomas. data from 13 International Breast Cancer Study
• Outcome Group (IBCSG) trials, Huober et al. found that
In the original report from Ridolfi et al., clas- patients with MC diagnosed on morphology
sical MC, diagnosed using their strict criteria, alone had an improved prognosis in terms of
198 Invasive Carcinoma with Medullary Features
disease free and overall survival at 14 years Foci of hemorrhage and necrosis may be evi-
(Huober et al. 2012). Restricting the diagnosis dent but are not extensive.
to patients with hormone receptor negative dis- Ridolfi et al. observed an average tumor size of
ease was associated with an even more favor- 2.9 cm (Ridolfi et al. 1977). Some studies report
able outlook. an average tumor size of up to 4 cm but in
In the last decade, the role of TILs in medi- most studies the recorded median size is in the
ating response to chemotherapy and improving range of 2– 3 cm.
clinical outcomes in all subtypes of breast can- The imaging characteristics of CMF, including
cer has been recognized. TNBCs are associated those observed on mammography and ultrasound
with a greater degree of lymphoid infiltration examination, may resemble a fibroadenoma.
than luminal tumors and survival benefit Magnetic resonance imaging (MRI) does not
increases with each 10% increase in TILs differentiate between CMF and other breast
(Stanton and Disis 2016). The survival advan- tumor types.
tage associated with TILs appears to be more Calcification is rare in CMF due to the lack of
powerful in TNBCs than in HER2 positive stromal desmoplasia and to the absence or low
tumors that may also feature lymphoid infiltra- volume of accompanying DCIS.
tion. Considering that a prominent lymphoid
infiltrate is one of the key morphological
criteria for a diagnosis of CMF, it appears likely Microscopy
that the improved prognosis in CMF is related
to the host immune response, particularly in The original criteria for a diagnosis of classical
triple negative tumors. Gene expression profil- MC included complete tumor circumscription
ing studies (GEP) have also demonstrated that with a pushing border and no extension of tumor
the expression levels of immune response genes cells into adjacent stroma, a syncytial growth
are independent predictors of outcome in highly pattern in at least 75% of the tumor, a moderate
proliferative breast cancers. A high TIL count to marked mononuclear stromal infiltrate, high
also has implications for targeted immunother- nuclear grade, no glandular features and no
apy as discussed above. intraduct component (DCIS). The last criterion
In the context of prognosis and treatment, was shown to be less important with a DCIS
the decision by the WHO to broaden the defi- component having no impact on prognosis.
nition and to create the designation of CMF A tumor showing the majority of these features
(to include MC, AMC, and invasive carcinoma can be diagnosed as CMF. This approach should
NST with specific histological appearances) facilitate recognition and identification of CMF.
takes account of the compelling data regarding However, due to the inherent flexibility in the
the improved outcome associated with specific application of the diagnostic criteria (“some but
MC morphology and that reported in relation not all of the features of MC”), interobserver
to a high TIL count in TNBC. variability and reproducibility of diagnosis con-
tinue to be a challenge.
Despite these limitations, tumors categorized
Macroscopy as CMF share the following morphological
features:
CMF, by definition, is a well-circumscribed tumor Outline: CMF is a well-delineated tumor
with an expansile or pushing border and may with a predominant pushing or expansile margin
resemble a fibroadenoma on gross inspection. The (Figs. 1 and 2).
cut surface is fleshy and the consistency is soft, Syncytial growth pattern: This refers to the
attributed to the absence of a stromal desmoplastic arrangement of tumor cells in solid, broad anasto-
reaction. The color varies from tan to grey-white. mosing sheets with minimal intervening stroma.
In classical MC, this pattern is present in at least
Invasive Carcinoma with Medullary Features 199
Invasive Carcinoma with Medullary Features, Fig. 1 Invasive Carcinoma with Medullary Features, Fig. 3
(H&E 4) Low-power view of CMF showing the classical (H&E 20) Tumor cells of CMF show a syncytial growth
well-circumscribed outline with an expansile edge pattern, high nuclear grade and mitotic activity
Invasive Carcinoma with Medullary Features, Fig. 2 Invasive Carcinoma with Medullary Features, Fig. 4
(H&E 10) CMF at higher magnification showing the (H&E 10) CMF showing a prominent lymphoplas-
classical well-circumscribed outline with an expansile edge macytic infiltrate
75% of the tumor. Although the latter is not a Lymphoplasmacytic infiltrate: This is
requirement for diagnosis, CMF shows a predom- usually a prominent feature of CMF (Fig. 4). The
inant solid growth pattern with scant intervening infiltrate tends to be more marked at the periphery
stroma. The relative paucity of stroma and lack of of the tumor and comprises a mixed population of
a desmoplastic reaction confer a soft consistency CD3+ T lymphocytes, cytotoxic CD8+ T lympho-
on these tumors which, together with the typical cytes, and plasma cells.
pushing border, may give a false impression of a The presence of a prominent lympho-
benign lesion on gross examination. plasmacytic infiltrate is associated with an
High nuclear grade: CMF is composed of improved prognosis related to the host lymphocyte
cells with high nuclear grade and prominent response.
nucleoli that may be multiple. Tumor giant cells Gland formation: The complete absence of
are commonly seen. Mitotic figures, including gland formation is a strict requirement for a
atypical forms, are frequent (Fig. 3).
200 Invasive Carcinoma with Medullary Features
lymphoepithelioma-like carcinoma of the breast, that approximately 20% of patients with CMF
expression of EBV protein is rare. test positive for BRCA1 mutation. CMF is
uncommon in patients with BRCA2 mutations.
Somatic BRCA1 mutations and BRCA1 pro-
Molecular Features moter hypermethylation have also been reported
in CMF. Promoter hypermethylation may lead to
Molecular Classification inactivation of the BRCA1 gene and contribute to
Gene expression profiling (GEP) studies, using breast carcinogenesis.
unsupervised cluster analysis, has led to the classi- The most common somatic mutation in CMF is
fication of breast cancer based on molecular char- TP53, which occurs at a higher rate than in non
acteristics (Perou et al. 2000). The vast majority of CMF tumors. TP53 mutations are common in all
CMF tumors segregate as basal-type, which are forms of breast cancer and overall are associated
typically characterized by lack of expression of with a worse prognosis. However, the prognostic
ER, PR, and HER2 and by positive expression of impact appears to vary with molecular subtype
genes that are expressed in the basal-like cells of the and may be limited to ER positive tumors.
normal breast and those associated with high pro- In keeping with the high rate of TP53 mutations
liferative activity. Further transcriptional analysis in CMF, high-density array comparative genomic
I
of the basal-like TNBC group has led to the recog- hybridization (CGH) studies have demonstrated a
nition of heterogeneity within this group of tumors high level of genomic instability in these tumors.
and the identification of basal-like TNBC molecu- Gains of 1q and 8q are common to all basal-like
lar subtypes (Lehmann and Pietenpol 2013). carcinomas and are observed in CMF. CMF
CMF tumors display a unique gene expression tumors also show a specific pattern of genetic
profile (Bertucci et al. 2006). CMF tumors are alteration including recurrent gains at 3p, 9p,10p,
characterized by upregulation of genes involved and 16q and losses at 4p and amplicons of 1q, 8p,
in the immune response including those that con- and 10p (Vincent-Salomon et al. 2007).
trol interleukin, interferon, and cytokine produc-
tion. Genes involved in activation of the apoptosis
pathway are also over-expressed compared with Differential Diagnosis
non-CMF basal-like tumors. In contrast, genes
that are involved in remodeling of the cytoskeleton When confronted with a well-circumscribed inva-
and those that control cell invasiveness are under- sive breast tumor with no associated DCIS and
expressed in CMF (Weigelt et al. 2010). This pro- triple negative for ER, PR, and HER2, it is always
file correlates with the immunomodulatory subtype wise to maintain a broad differential diagnosis.
of TNBC tumors, associated with an improved Recognition of CMF is important due to its
response to chemotherapy, a more favorable prog- association with the BRCA1 germline mutation.
nosis and potential for treatment with A diagnosis of CMF, particularly in a younger
immunotherapy. patient, will lead to consideration of genetic test-
ing for the patient and her/his family. CMF also
Genetic Alterations has a more favorable prognosis than a grade
BRCA1, located at chromosome 17q12–21, is a 3 invasive carcinoma NST. These characteristics
tumor suppressor gene. Germline mutations of emphasize the importance and value of careful
BRCA1 account for 30–40% of hereditary breast assessment of breast tumor morphology, even in
carcinomas. Most breast tumors that develop in this era of sophisticated genetic methodologies.
patients with BRCA1 germline mutations are of The main alternative diagnosis is invasive car-
CMF type. A diagnosis of CMF may, therefore, cinoma NST (▶ Invasive Carcinoma NST). To
signify a BRCA1 mutation and consideration of date there has been considerable interobserver var-
genetic testing. However, not all patients with iation in the diagnosis of MC and AMC. Applica-
CMF have a BRCA1 mutation. It is estimated tion of the more relaxed histological criteria
202 Invasive Carcinoma with Medullary Features
proposed by the WHO and outlined above should Dua, I., & Tan, A. R. (2017). Immunotherapy for triple-
improve observer agreement in recognizing CMF negative breast cancer: A focus on immune checkpoint
inhibitors. American Journal of Haematology and
and distinguishing it from NST. In view of the Oncology, 13, 20–27.
clinical relevance, it is more important to entertain Eisinger, F., Noguès, C., Birnbaum, D., Jacquemier, J., &
the diagnosis than to assign these tumors to the Sobol, H. (1998). BRCA1 and medullary breast cancer.
NST category. The preferential expression of basal Journal of the American Medical Association, 280,
1227–1228.
type immunohistochemical markers may be help- Ellis, I. O., Al-Sam, S., Anderson, N., Carder, P., Deb, R.,
ful in making a diagnosis of CMF. Girling, A., et al. (2016). Pathology reporting of breast
The possibility of a metastasis from an alternative disease in surgical excision specimens incorporating
primary site or an intramammary lymph node the dataset for histological reporting of breast cancer.
(2016). Rcpath.org. Available from: http://www.rcpath.
replaced by metastatic tumor should be borne in org. Accessed Sept 2017.
mind. This is less likely when dealing with a single Flucke, U., Flucke, M., Hoy, L., Breuer, E., Goebbels, R.,
tumor. Tumors that metastasize to the breast include Rhiem, K., et al. (2010). Distinguishing medullary car-
melanoma, which may be pigmented, ovarian carci- cinoma of the breast from high-grade hormone receptor-
negative invasive ductal carcinoma: An immunohisto-
noma, lung carcinoma, sarcomatoid carcinoma, and chemical approach. Histopathology, 56, 852–859.
sarcoma and primary sarcoma. Thorough evaluation Hicks, D. G., & Lester, S. C. (2017). Carcinoma with
of tumor morphology and consideration of the clin- medullary features. Available from: https://app.
ical history and radiological findings are key to expertpath.com. Accessed Sept 2014.
Huober, J., Gelber, S., Coates, A. S., Viale, G.,
reaching a correct diagnosis. In the case of an Ohlschlegel, C., et al. (2012). Prognosis of medullary
intramammary lymph node replaced by tumor, a breast cancer: Analyses of 13 international breast can-
search for lymph node capsule and any residual cer study group (IBCSG) trials. Journal of Clinical
normal lymph node architecture should assist the Oncology, 11, 2843–2851.
Jacquemier, J., Padovani, L., Rabayrol, L., Lakhani, S.,
diagnosis. Comparison with previous histology Penault-Llorca, F., Denoux, Y., et al. (2005). Typical
and the use of immunohistochemistry will also assist medullary breast carcinomas have a basal/myo-
accurate diagnosis of an intramammary metastasis. epithelial phenotype. The Journal of Pathology, 207,
Lymphoma of the breast is a rare condition that 260–268.
Lakhani, S., Ellis, I. O., Schnitt, S. J., Tan, P. H., & van de
should also be considered in the differential diag- Vijver, M. J. (2012). WHO classification of tumours of
nosis. Breast lymphoma (mostly diffuse large the breast (pp. 46–47). Lyon: International Agency for
B-cell lymphoma) may occur as a primary process Research on Cancer.
or represent breast involvement by known sys- Lehmann, B., & Pietenpol, J. (2013). Identification and use
of biomarkers in treatment strategies for triple-negative
temic lymphoma. It usually forms a solid mass breast cancer subtypes. The Journal of Pathology, 232,
and may mimic carcinoma both radiologically 142–150.
and histologically. On close inspection, it lacks Mateo, A., Pezzi, T., Sundermeyer, M., Kelley, C.,
the syncytial growth pattern observed in CMF. Klimberg, V., & Pezzi, C. (2016). Chemotherapy
significantly improves survival for patients with
Consideration of the diagnosis and the use of T1c-T2N0M0 medullary breast cancer: 3739 cases
immunohistochemistry will clarify the diagnosis. from the National Cancer Data Base. Annals of Surgi-
cal Oncology, 24, 1050–1056.
Matkovic, B., Juretic, A., Separovic, V., Novosel, I.,
Separovic, R., Gamulin, M., et al. (2008). Immunohis-
References and Further Reading tochemical analysis of ER, PR, HER-2, CK 5/6,
p 63 and EGFR antigen expression in medullary breast
Badve, S., Dabbs, D., Schnitt, S., Baehner, F., Decker, T., cancer. Tumori, 94, 838–844.
Eusebi, V., et al. (2010). Basal-like and triple-negative Perou, C. M., Sorlie, T., Eisen, M. B., van de Rijn, M.,
breast cancers: A critical review with an emphasis Jeffrey, S. S., Rees, C. A., et al. (2000). Molecular
on the implications for pathologists and oncologists. portraits of human breast tumors. Nature, 406,
Modern Pathology, 24, 157–167. 474–452.
Bertucci, F., Finetti, P., Cervera, N., Charafe-Jauffret, E., Rakha, E., & Green, A. (2017). Molecular classification of
Mamessier, E., Adélaïde, J., et al. (2006). Gene expres- breast cancer: What the pathologist needs to know.
sion profiling shows medullary breast cancer is a sub- Pathology, 49, 111–119.
group of basal breast cancers. Cancer Research, 66, Ridolfi, R., Rosen, P., Port, A., Kinne, D., & Miké, V.
4636–4644. (1977). Medullary carcinoma of the breast.
Invasive Carcinoma with Neuroendocrine Differentiation 203
A clinicopathologic study with 10 year follow-up. carcinomas/small cell carcinomas, and invasive
Cancer, 40, 1365–1385. carcinomas with NE differentiation.
Silwal-Pandit, L., Vollan, H., Chin, S., Rueda, O.,
McKinney, S., Osako, T., et al. (2014). TP53 mutation
Spectrum in breast cancer is subtype specific and has
distinct prognostic relevance. Clinical Cancer Clinical Features
Research, 20, 3569–3580.
Stanton, S., & Disis, M. (2016). Clinical significance
of tumor-infiltrating lymphocytes in breast cancer. • Incidence
Journal for Immunotherapy of Cancer, 4, 59. Invasive breast carcinomas with NE features
Tung, N., Garber, J., Hacker, M., Torous, V., Freeman, G., are rare and represent less than 5% of breast
Poles, E., et al. (2016). Prevalence and predictors of cancers (Wang et al. 2014).
androgen receptor and programmed death-ligand 1 in
BRCA1-associated and sporadic triple-negative breast • Age
cancer. Breast Cancer, 2, 16002. Most patients are in the sixth or seventh
Vincent-Salomon, A., Gruel, N., Lucchesi, C., decades of life.
MacGrogan, G., Dendale, R., Sigal-Zafrani, B., et al. • Sex
(2007). Identification of typical medullary breast carci-
noma as a genomic sub-group of basal-like carcinomas, There is a female predilection. Single case
a heterogeneous new molecular entity. Breast Cancer reports of invasive breast carcinomas with
Research, 9, R24. NE features in male patients were occasionally
Weigelt, B., Geyer, F., & Reis-Filho, J. (2010). Histological I
described.
types of breast cancer: How special are they?
Molecular Oncology, 4, 192–208. • Site
Usually, invasive breast carcinomas with NE
differentiation present unilaterally, neverthe-
less a single case report with bilateral primary
breast NE carcinoma in a young woman has
Invasive Carcinoma with been described (Zhang and Chen 2011). The
Neuroendocrine central and subareolar region of the breast or
Differentiation the upper outer quadrant are the most frequent
locations where this type of carcinoma arises.
Ewa Chmielik Rarely, multicentric forms were reported.
Tumor Pathology Department, Maria • Treatment
Sklodowska-Curie Memorial Cancer Center and Surgery is the standard initial treatment for early
Institute of Oncology, Gliwice, Poland invasive carcinomas with NE features. Patients
with locally advanced cases or with inoperable
carcinomas are treated with neoadjuvant ther-
Synonyms apy. Patients with luminal A or B carcinomas
are candidates for adjuvant endocrine therapy. In
Carcinoid tumor of the breast; Endocrine hormone receptor positive cases with high
carcinoma Ki-67 proliferation index, adjuvant chemother-
apy in addition to endocrine therapy may be
beneficial. Anti-HER2 therapy is an option for
Definition patients with HER2-positive carcinomas with
NE features, which is a quite rare event. Peptide
Invasive carcinoma with neuroendocrine receptor radionuclide therapy (PPRT) has been
(NE) differentiation presents microscopic features reported in patients with carcinomas expressing
similar to NE neoplasms of other organs. somatostatin receptors (Inno et al. 2016).
Bussolati and Badve (2012) classifies Breast NE carcinomas can metastasize to
invasive carcinomas with NE differentiation multiple organs even years after treatment of
into three subtypes: well-differentiated NE the primary tumor, and therefore long-term
tumors, poorly differentiated neuroendocrine follow-up is advisable (Inno et al. 2016).
204 Invasive Carcinoma with Neuroendocrine Differentiation
• Outcome
Well-differentiated NE tumors have a favorable
prognosis and have significantly less frequent
lymph node metastases compared with those with
partial NE differentiation (Sapino et al. 2001a).
Mucinous differentiation, estrogen receptor
(ER) expression, and progesterone receptor
(PR) expression are important parameters for
favorable prognosis in invasive carcinomas
with NE features (Sapino et al. 2001a).
However, as a group, patients with breast car-
cinomas showing NE differentiation have worse
overall survival (OS) and disease-free survival
(DFS) than those without (Kwon et al. 2014).
In particular, poorly differentiated/small cell
carcinomas are aggressive with poor prognosis
(Sapino et al. 2001a). The most frequent meta-
static sites of breast NE carcinomas are bone,
lungs, bone marrow, liver, pleura, skin, adrenal
gland, and pancreas (Lavigne et al. 2017) and
less frequently in other organs.
Invasive Carcinoma with Neuroendocrine Differenti-
ation, Fig. 1 Mammography shows a stellate lesion
Macroscopy which was histologically a well-differentiated NE breast
tumor with microcalcifications, BI-RADS 5, ACR 2
Invasive breast carcinomas with NE differentia-
tion may be grossly well circumscribed or with
infiltrative border (Fig. 1 radiogram of invasive
breast carcinoma with NE differentiation).
Microscopy
Invasive Carcinoma with Neuroendocrine Differenti- Invasive Carcinoma with Neuroendocrine Differenti-
ation, Fig. 3 Carcinoid-like pattern with rosettes (H&E, ation, Fig. 4 Strongly and diffusely positive
40) immunostaining for synaptophysin (10)
I
is the preinvasive counterpart of mucinous carci-
noma with NE differentiation.
The mucinous variant of NE carcinoma shows
lakes of mucin populated by large clusters of cells
unlike non-NE mucinous carcinoma (▶ Invasive
Mucinous Carcinoma).
In alveolar growth pattern, large clear cells are
separated by scanty dense stroma. The number of
mitoses can be higher than in the other
histotypes.
Within poorly differentiated NE carcinomas
of the breast, the small cell variant is character-
ized by high nuclear/cytoplasmic ratio, dark Invasive Carcinoma with Neuroendocrine Differenti-
chromatin, inconspicuous nucleoli, and scant ation, Fig. 5 Carcinoma in situ component of NE breast
eosinophilic cytoplasm. The mitotic activity is carcinoma positive for Chromogranin A (40)
brisk (approximately 10 mitoses/10HPF). Zones
of necrosis or hemorrhage are often encountered.
The presence of an intraductal component of differentiation detected only by immunohistochem-
small cell carcinoma supports the breast origin. istry occurs in up to 30% of invasive carcinomas
NST (▶ Invasive Carcinoma NST) or other special
types. CD56 immunostaining and TTF-1 (20% of
Immunophenotype cases) have been documented in primary breast
small cell carcinoma.
All invasive carcinomas with NE differentiation ER are expressed in all of well-differentiated NE
express NE markers. Synaptophysin (Fig. 4) and tumors (Lavigne et al. 2017) or nearly in all (95%)
chromogranin A (Fig. 5) are considered as the cases (Bogina et al. 2016). The vast majority of
most sensitive and specific NE markers. cases (89%) express PR (Lavigne et al. 2017).
Seventy percent of NE carcinomas express Only few cases show immunophenotype
chromogranin A, 96% express synaptophysin, and ER+/HER2+, ER /HER2+, or lack ER and
66% express both markers (Lavigne et al. 2017). NE HER2 expression (Bogina et al. 2016).
206 Invasive Carcinoma with Neuroendocrine Differentiation
Definition • Treatment
These tumors are generally recognized to have
The definition of primary mammary invasive signet an aggressive behavior with higher frequency
ring cell carcinoma (SRCC) varies in the limited of vascular invasion and lymph node involve-
number of published series to date. Some restrict ment. Treatment will be governed but the char-
the definition to lobular carcinoma (Merino and acteristics of the individual patient and based
Livolsi 1981), with a population of signet ring on tumor size, grade, lymph node stage, hor-
cells present and exclude signet ring cell variant mone receptor (▶ Hormone Receptors in
of invasive ductal/NST carcinoma or vica versa. Breast Cancer), and HER2 (▶ HER2 in Breast
Others recognize signet ring cell tumor cell mor- Cancer) status.
phology in invasive ductal, lobular, and mucinous • Outcome
carcinoma, and one recent study has shown no Most but not all of the published series recog-
difference between these variants (Ohashi et al. nize SRC to be more aggressive than mucinous
2016). The current 2012 WHO classification of carcinoma (▶ Invasive Mucinous Carcinoma),
breast tumors (Lakhani et al. 2012) includes invasive ductal carcinoma of no special type
SRCC as a subcategory of invasive mucinous car- (▶ Invasive Carcinoma NST), and classic
cinoma (▶ Invasive Mucinous Carcinoma) and invasive lobular carcinoma (▶ Invasive Lobu-
uses the following definition, “Carcinomas with lar Carcinoma) of the breast, with frequent
signet-ring-cell differentiation are characterized by metastasis to regional lymph nodes and distant
abundant intracellular mucin that pushes the metastasis to the lung, liver, and bone but also
nucleus to one side, creating the characteristic with unusual sites of metastasis including the
signet-ring-cell morphology.” The published series stomach, endometrium, cervix, serosa, gastro-
differ on the number of signet cells that must be intestinal tract, urinary tract, and spleen.
present to label a carcinoma as SRCC, but generally
require >10–20% signet ring cells as a minimum
Macroscopy
requirement.
There are no specific macroscopic features.
Clinical Features
• Incidence Microscopy
Primary SRCC carcinoma of the breast is a
very rare tumor subtype with few series SRCC has been defined as a lesion with diffuse
published to date. It is exceptionally rare as a infiltration of the stroma by mucin containing signet
pure form but presence of SRCC cells have ring cells where mucin fills the cytoplasm and dis-
been reported to occur in between 1% and 4% places nucleus. Prominent signet-ring-cell differen-
of primary breast carcinomas; however, there tiation is most common in invasive lobular
are no detailed epidemiological or comparative carcinomas but may also be seen in invasive carci-
studies available. noma NST and other special type cancers, particu-
• Age larly mucinous carcinoma. Pure SRCC is
No large series exist but the average age at exceptionally rare, and it has been argued that car-
presentation appears to be older than that for cinomas with signet-ring-cell differentiation do not
breast cancer as a whole. represent a distinct entity. The current 2012 WHO
• Sex classification of breast tumors (Bussolati and
Marked female predominance. Sapino 2012) describes two cytological types of
• Site carcinomas with signet-ring-cell differentiation.
Breast but exclusion of metastatic SRCC from One type characterized by large intracytoplasmic
distant sites particularly GI tract needs to be vacuoles, with a “target” appearance owing to the
excluded. presence of large intracytoplasmic lumina
Invasive Carcinoma with Signet Ring Cell Differentiation 209
I
containing a periodic acid Schiff (PAS)/Alcian blue Differential Diagnosis
and HMFG2-positive central globule. This cytolog-
ical pattern is typically observed in lobular neopla- The differential diagnosis includes mucinous
sia, in classic ▶ invasive lobular carcinoma, and it carcinomas and clear cell carcinomas and metas-
has also been associated with the pleomorphic var- tasis from other sites. It is important to differen-
iant of lobular carcinoma (▶ Pleomorphic Lobular tiate from these carcinomas as the treatment
Carcinoma). The other cytological type is similar to options and the outcome varies considerably.
the cells of diffuse gastric carcinoma (Fig. 1) and is Metastases will lack an accompanying in situ
characterized by acidic muco-substances that dif- carcinoma (DCIS or LCIS) component.
fusely fill the cytoplasm and dislodge the nucleus to A variety of immunohistochemical markers
one pole of the cell. This type of signet-ring cell has have been applied to distinguish SRCC from
also been described as a variant of DCIS. metastasis from different organs. SRCCs of the
breast are generally immunohistochemically
positive for GCDFP-15, whereas SRCCs of the
Immunophenotype gastrointestinal tract are negative. ER is very
often positive in primary SRCC of the breast,
Primary breast SRCC are usually GCDFP-15 pos- but commonly negative in gastric and colonic
itive and approximately 80% are positive for SRCC. Adenocarcinomas of the breast, stomach,
estrogen receptor. HER2 status frequency is not and colon also show different CK7 and CK20
well described but HER2 positive cases occur but expression patterns (Tot 2000). While primary
at a lower frequency than breast cancer as a whole. SRCC of the breast is typically positive for
These tumors typically have a luminal type CK7 but negative for CK20, the gastrointestinal
cytokeratin profile being low molecular weight SRCCs are commonly positive for CK20 but
cytokeratin CK8/18 and CK7 positive. Lobular usually negative for CK7. In combination with
variants are usually E-Cadherin negative. ER staining, CK7 and CK20 expression patterns
can be used to distinguish gastrointestinal SRCC
from SRCC of the breast but it should be borne in
Molecular Features mind that approximately 20% of SRCCs of the
breast are negative for ER (Tot 2000). Mucin
There are no known specific genetic or molecular secretion type may also be helpful (Chu and
features. Weiss 2004). Adenocarcinomas of the breast
210 Invasive Cribriform Carcinoma
Invasive Cribriform Carcinoma, Fig. 1 (H&E) a) A arranged in cribriform pattern and desmoplastic stroma.
case of invasive cribriform carcinoma showing irregular Higher-power view is shown in Fig. 1b
infiltrating islands of low-nuclear-grade malignant cells
References Definition
Ellis, I. O., Galea, M., Broughton, N., Locker, A., Invasive lobular carcinoma (ILC) is characterized
Blamey, R. W., & Elston, C. W. (1992). Pathological
by noncohesive cells, usually E-cadherin negative,
prognostic factors in breast cancer. II. Histological
type. Relationship with survival in a large study with that are dispersed or arranged in single-file linear
long-term follow-up. Histopathology, 20, 479–489. pattern, but can show several additional architec-
Li, C. I., Uribe, D. J., & Daling, J. R. (2005). Clinical tural patterns (Tavassoli and Eusebi 2009; Lakhani
characteristics of different histologic types of breast
cancer. British Journal of Cancer, 93, 1046–1052.
et al. 2012). ILC is frequently associated with
Louwman, M. W., Vriezen, M., van Beek, M. W., lobular carcinoma in situ (LCIS).
Nolthenius-Puylaert, M. C., van der Sangen, M. J.,
Roumen, R. M., Kiemeney, L. A., & Coebergh, J. W.
(2007). Uncommon breast tumors in perspective: Inci-
dence, treatment and survival in the Netherlands. Inter-
Clinical Features
national Journal of Cancer, 121, 127–135.
Page, D. L., Dixon, J. M., Anderson, T. J., Lee, D., & • Incidence
Stewart, H. J. (1983). Invasive cribriform carcinoma ILC is the second most common type of breast
of the breast. Histopathology, 7, 525–536.
cancer and constitutes 5–15% of all invasive
Venable, J. G., Schwartz, A. M., & Silverberg, S. G. (1990).
Infiltrating cribriform carcinoma of the breast: breast tumors (Lakhani et al. 2012), although
A distinctive clinicopathologic entity. Human Pathology, incidence varies according to the criteria
21, 333–338. applied for diagnosis. The incidence of ILC is
Wells, C. A., & Ferguson, D. J. (1988). Ultrastructural and
immunocytochemical study of a case of invasive crib-
increasing, especially among postmenopausal
riform breast carcinoma. Journal of Clinical Pathology, women, probably as consequence of hormonal
41, 17–20. replacement therapy and alcohol abuse (Rakha
and Ellis 2010).
• Age
ILC usually affects peri and postmenopausal
Invasive Lobular Carcinoma women aged 50 or more but it can be seen in a
wide age range of patients, from young, in their
Maria P. Foschini and Luca Morandi third decade of life, to elderly women, aged more
Department of Biomedical and Neuromotor than 80 years (Tavassoli and Eusebi 2009).
Sciences (DIBINEM), Unit of Anatomic • Sex
Pathology at Bellaria Hospital, University of ILC affects more frequently female patients
Bologna, Bologna, Italy and it is rare in men.
Invasive Lobular Carcinoma 213
Invasive Lobular Carcinoma, Fig. 1 A: ILC-C is char- stained with low molecular weight cytokeratin antibody)
acterized by a diffuse growth of neoplastic cells infiltrat- are arranged around a ductule in a targetoid fashion
ing the mammary tissue (H&E). B: Neoplastic cell, (here
Invasive Lobular Carcinoma, Fig. 2 ILC-C sometimes a solid architecture can be seen (A) (H&E). Negative
immunohistochemistry for E-cadherin helps in the differential diagnosis with invasive carcinoma of no special type (B)
immunohistochemistry (IHC)) due to invasion of prognosis. Mitotic count is low and necrosis is
the prelymphatic channels of Hartveit (Tavassoli rarely seen in this variant.
and Eusebi 2009). No inflammatory reaction of ILC with solid architecture (ILC-S) can present
the stroma is seen. Neoplastic cells have monoto- as a solid mass, of varying size, usually >2
nous morphology, roundish central nucleus, with cm. ILC-S is composed of neoplastic cells similar
delicate chromatin, and scarce cytoplasm. to those of the ILC-C, but often showing higher
Intracytoplasmic vacuoles are not infrequent and nuclear grade (Fig. 2a, H&E). Atypical mitotic
can be highlighted by Alcian blue pH2.5/PAS figures and necrotic areas are frequent findings in
after diastase staining. On electron microscopy, ILC-S. Negative immunohistochemistry for
the intracytoplasmic lumina are lined by E-cadherin helps in the differential diagnosis with
microvilli (Tavassoli and Eusebi 2009). invasive carcinoma of no special type (▶ Invasive
ILC-C neoplastic cells can invade diffusely the Carcinoma NST) (Fig. 2b, (IHC) E-cadherin high-
mammary gland, in a “spider–web” fashion (Tot lights the epithelial cells of a residual duct. ILC
2016). This type of growth indicates poor nests are negative).
Invasive Lobular Carcinoma 215
Cytologic Variants
All these features may manifest within each his-
tologic variants. Invasive Lobular Carcinoma, Fig. 5 ILC-SR shows
Histiocytoid variant (ILC-H) (Tan et al. 2011) numerous cells with intracytoplasmic vacuole (H&E)
is composed of neoplastic cells that resemble his-
tiocytes having a clear and finely foamy cyto-
plasm (Fig. 4, H&E). On electron microscopy, two types of signet ring cells can be encountered
the neoplastic histiocytoid cells show large clear (Tavassoli and Eusebi 2009). The first and more
vesicles, typical of apocrine cells. The apocrine common type is characterized by intracytoplasmic
nature of these neoplastic cells can be demon- mucin-filled vacuoles, with microvilli seen on elec-
strated with positive immunohistochemical reac- tron microscopy. The second type shows dilated
tion for the apocrine marker Gross Cystic Disease cisternae of the endoplasmic reticulum filled with
Fluid Protein 15 (GCDFP-15). PAS or Alcian blue positive mucin similar to the SR
Apocrine variant (ILC-A): the neoplastic cells cells of the stomach.
show eosinophilic and granular cytoplasm, central Myoepithelial cell variant (ILC-M): rare
nucleus with evident nucleolus. Diffuse positivity cases showing evidence of myoepithelial cell dif-
for GCDFP-15 is usually encountered. ILC-A may ferentiation are on record (Del Vecchio et al. 2005;
be associated with LCIS with apocrine features. Tavassoli and Eusebi 2009). The cases described
Signet ring cell variant (ILC-SR): cells with a by Del Vecchio et al. (2005) were composed of
signet ring appearance are the predominant cells having features intermediate between
neoplastic population (Fig. 5, H&E). In ILC-SR, epithelial-secretory and myoepithelial (therefore
216 Invasive Lobular Carcinoma
mucin production—a novel pattern of lobular carcino- breast carcinoma. Evidence of their frequent expression
mas of the breast. Clinico-pathological description of in lobular carcinoma. Virchows Arch, 447(4), 695–700.
eight cases. Virchows Arch, 471(1), 3–12. Sakr, R. A., Schizas, M., Carniello, J. V., Ng, C. K.,
Dabbs, D. J., Schnitt, S. J., Geyer, F. C., Weigelt, B., Baehner, Piscuoglio, S., Giri, D., Andrade, V. P., De Brot, M.,
F. L., Decker, T., Eusebi, V., Fox, S. B., Ichihara, S., Lim, R. S., Towers, R., Weigelt, B., Reis-Filho, J. S., &
Lakhani, S. R., Palacios, J., Rakha, E., Richardson, King, T. A. (2016). Targeted capture massively parallel
A. L., Schmitt, F. C., Tan, P. H., Tse, G. M., Vincent- sequencing analysis of LCIS and invasive lobular can-
Salomon, A., Ellis, I. O., Badve, S., & Reis-Filho, J. S. cer: Repertoire of somatic genetic alterations and clonal
(2013). Lobular neoplasia of the breast revisited with relationships. Mol Oncol, 10(2), 360–370.
emphasis on the role of E-cadherin immunohistochemis- Tan, P. H., Harada, O., Thike, A. A., & Tse, G. M. (2011).
try. Am J Surg Pathol, 37(7), e1–11. Histiocytoid breast carcinoma: an enigmatic lobular
Del Vecchio, M., Foschini, M. P., Peterse, J. L., & entity. J Clin Pathol, 64(8), 654–659.
Eusebi, V. (2005). Lobular carcinoma of the breast Tavassoli, F. A., & Eusebi, V. (2009). Invasive lobular
with hybrid myoepithelial and secretory carcinoma. In Tumors of the mammary gland, AFIP
("myosecretory") cell differentiation. Am J Surg Atlas of tumor pathology, series 4 (pp. 156–168).
Pathol, 29(11), 1530–1536. Washington, DC: American Registry of Pathology.
Desmedt, C., Zoppoli, G., Gundem, G., Pruneri, G., Tot, T. (2016). Diffuse invasive breast carcinoma of no
Larsimont, D., Fornili, M., Fumagalli, D., Brown, D., special type. Virchows Arch, 468(2), 199–206.
Rothé, F., Vincent, D., Kheddoumi, N., Rouas, G., Majjaj, van Deurzen, C. H., Cserni, G., Bianchi, S., Vezzosi, V., Arisio,
S., Brohée, S., Van Loo, P., Maisonneuve, P., Salgado, R., R., Wesseling, J., Asslaber, M., Foschini, M. P., Sapino, A.,
Van Brussel, T., Lambrechts, D., Bose, R., Metzger, O., Castellano, I., Callagy, G., Faverly, D., Martin-Martinez,
Galant, C., Bertucci, F., Piccart-Gebhart, M., Viale, G., M. D., Quinn, C., Amendoeira, I., Kulka, J., Reiner- I
Biganzoli, E., Campbell, P. J., & Sotiriou, C. (2016). Concin, A., Cordoba, A., Seldenrijk, C. A., van Diest, P.
Genomic characterization of primary invasive lobular J. (2010). Nodal-stage classification in invasive lobular
breast cancer. J Clin Oncol, 34(16), 1872–1881. breast carcinoma: influence of different interpretations of
Eusebi, V., Foschini, M. P., Bussolati, G., Rosen, P. P. the pTNM classification. J Clin Oncol, 28(6), 999–1004.
(1995). Myoblastomatoid (histiocytoid) carcinoma of
the breast. A type of apocrine carcinoma. Am J Surg
Pathol, 19(5), 553–62.
Foschini, M. P., Righi, A., Cucchi, M. C., Ragazzini, T.,
Merelli, S., Santeramo, B., & Eusebi, V. (2006). The Invasive Metaplastic
impact of large sections and 3D technique on the study Carcinoma
of lobular in situ and invasive carcinoma of the breast.
Virchows Arch, 448(3), 256–261.
Iorfida, M., Maiorano, E., Orvieto, E., Maisonneuve, P., Caterina Marchiò1,2,3, Suzanne Chartier3,
Bottiglieri, L., Rotmensz, N., Montagna, E., Guillaume Bataillon3 and
Dellapasqua, S., Veronesi, P., Galimberti, V., Luini, A., Anne Vincent-Salomon3
Goldhirsch, A., Colleoni, M., & Viale, G. (2012). Inva- 1
Unit of Pathology, Candiolo Cancer Institute
sive lobular breast cancer: subtypes and outcome. Breast
Cancer Res Treat, 133(2), 713–723. FPO-IRCCS, Candiolo, Italy
2
Lakhani, S. R., Rakha, E. A., & Simpson, P. T. (2012). Department of Medical Sciences, University of
Invasive lobular carcinoma. In S. R. Lakhani, I. O. Turin, Turin, Italy
Ellis, S. J. Schnitt, P. H. Tan, & M. van de Vijver 3
Institut Curie, Paris, France
(Eds.), WHO classification of tumors of the breast
(4th ed., pp. 40–42). Lyon: IARC Press.
Morandi, L., Marucci, G., Foschini, M. P., Cattani, M. G.,
Pession, A., Riva, C., & Eusebi, V. (2006). Genetic Synonyms
similarities and differences between lobular in situ neo-
plasia (LN) and invasive lobular carcinoma of the
breast. Virchows Arch, 449(1), 14–23. Carcinoma with pseudosarcomatous metaplasia;
Orvieto, E., Maiorano, E., Bottiglieri, L., Maisonneuve, P., Carcinosarcoma; Matrix-producing carcinoma;
Rotmensz, N., Galimberti, V., Luini, A., Brenelli, F., Sarcomatoid carcinoma; Spindle cell metaplastic
Gatti, G., & Viale, G. (2008). Clinicopathological char-
tumor
acteristics of invasive lobular carcinoma of the breast:
results of an analysis of 530 cases from a single insti-
tution. Cancer, 113(7), 1511–1520.
Rakha, E. A., & Ellis, I. O. (2010). Lobular breast carcinoma Definition
and its variants. Semin Diagn Pathol, 27(1), 49–61.
Riva, C., Dainese, E., Caprara, G., Rocca, P. C.,
Massarelli, G., Tot, T., Capella, C., & Eusebi, V. (2005). Metaplastic carcinomas represent a morphologi-
Immunohistochemical study of androgen receptors in cally heterogeneous group of invasive breast
220 Invasive Metaplastic Carcinoma
particularly the case for squamous cell carcino- organs such as the skin and, when dealing with
mas; in addition, when heterologous elements male patients or whenever a patient has a history
are abundant and predominant, the macroscopic of lung cancer or is heavy smoker, the lung (see
appearance may be more distinctive. For below for section “Differential Diagnosis”).
instance, on cut surface, squamous or chondroid Squamous cell carcinomas of the breast
areas may appear as pearly white to gray glisten- typically present as a cystic lesion where the cav-
ing areas. ity is lined by a squamous epithelium with vari-
Finally, at the time of diagnosis, they tend to be able nuclear atypia, and there is evidence of
larger than IC-NSTs (▶ Invasive Carcinoma neoplastic infiltration in the adjacent stroma that
NST), with a reported mean size of 3.9 cm usually displays a marked inflammatory infiltrate
(Collins and Schnitt 2013). (Lakhani et al. 2012).
The squamous differentiation of the neoplastic
cells can show well to poorly differentiated fea-
Microscopy tures (Fig. 1a and b, H&E), and a spindle cell
morphology can also be appreciated, typically at
Histologically these tumors are most often of high the infiltrative border of the tumor (Lakhani
grade, with conspicuous nuclear pleomorphism et al. 2012).
I
and mitotic activity; however they constitute a An acantholytic variant of squamous cell car-
heterogeneous group of lesions encompassing cinoma has also been described, featuring irregu-
several entities (Fig. 1). The 2012 WHO classifi- lar spaces lined by squamous cells. These spaces
cation has adopted a descriptive classification can be misinterpreted as vascular spaces and lead
including: to a wrong diagnosis of angiosarcoma (Lakhani
et al. 2012; Collins and Schnitt 2013).
– Squamous cell carcinoma
– Metaplastic carcinoma with mesenchymal Metaplastic Carcinoma with Mesenchymal
differentiation Differentiation
– Spindle cell carcinoma These lesions are defined by the presence of a
– Fibromatosis-like metaplastic carcinoma varying degree of heterologous differentiation of
– Low-grade adenosquamous carcinoma mesenchymal origin (Collins and Schnitt 2013)
and can also be called “matrix-producing carcino-
When diagnosing a metaplastic breast cancer, mas.” The most frequent heterologous differen-
one should clearly describe the distinct morpho- tiation encountered in metaplastic carcinomas
logical components present as this may have clin- of the breast is either osseous or cartilaginous
ical implications. (Fig. 1e and f, H&E) (Collins and Schnitt 2013).
As a general rule, the heterologous compo-
Squamous Cell Carcinoma nents may appear either benign or malignant, in
Metaplastic squamous cell carcinomas can be the latter case resembling/mimicking a sarcoma-
pure or admixed with an IC-NST (Lakhani et al. tous component. Based on this, whenever the
2012; Collins and Schnitt 2013). Pure squamous mesenchymal component is predominant a differ-
cell carcinomas of the breast are rare. It is impor- ential diagnosis with (i) a malignant phyllodes
tant to keep in mind that some degree of squamous tumor (▶ Phyllodes Tumor) with heterologous
differentiation can be observed in invasive carci- differentiation or (ii) a sarcoma (primary or meta-
nomas of no special type (▶ Invasive Carcinoma static) has to be taken into account (Collins and
NST) and is more commonly seen in carcinomas Schnitt 2013; Rakha et al. 2016).
with medullary features (▶ Invasive Carcinoma In these cases, an extensive sampling of the
with Medullary Features) (Lakhani et al. 2012; lesion is often required to look for unequivocal
Collins and Schnitt 2013). It is good practice to epithelial elements or foci of ductal carcinoma in
rule out the possibility of a metastasis from other situ (▶ Ductal Carcinoma In Situ) (Collins and
222 Invasive Metaplastic Carcinoma
Invasive Metaplastic Carcinoma, Fig. 1 H&E. Repre- carcinoma. (d) Fibromatosis-like metaplastic carcinoma. (e,
sentative microphotographs of the heterogeneity of metaplas- f) Two examples of a metaplastic carcinoma with mesenchy-
tic carcinomas. (a) squamous cell carcinoma. (b) A higher mal differentiation, one featuring chondroid differentiation
magnification of a metaplastic carcinoma with epidermoid (e) and the other showing bone tissue formation (f)
features showing high mitotic count. (c) Spindle cell
Schnitt 2013; Rakha et al. 2016). As discussed in is employed to demonstrate even a focal expres-
the differential diagnosis paragraph, this is the sion. It has to be stressed that not all metaplastic
typical scenario in which a panel of cytokeratins carcinomas express cytokeratins and that focal
Invasive Metaplastic Carcinoma 223
cytokeratin expression can also be observed in carcinoma with spindle-like features. The authors
phyllodes tumors (▶ Phyllodes Tumor), thus originally described this lesion by using the term
making the differential diagnosis particularly “tumor” to avoid the word “carcinoma” because
challenging and, in some cases, not possible neither the phenotype nor the behavior seemed to
(Collins and Schnitt 2013). be that of a carcinoma (Gobbi et al. 1999); how-
ever this entity was labeled as “carcinoma” in the
Spindle Cell Carcinoma 2012 WHO classification.
Metaplastic carcinomas can present as a prolifer- The microscopic growth pattern is typically
ation of atypical spindle cells (Lakhani et al. infiltrative, with finger-like projections extending
2012), thus posing important differential diagno- into adjacent mammary structures and fat tissue;
sis issues with other spindle cell lesions of the however lesions with ill-defined border or nodular
breast. The neoplastic spindle cells show a wide appearance have also been described (Gobbi et al.
morphological spectrum and may have a fasci- 1999). It is characterized by a dominant prolifer-
cular, storiform, or haphazard growth pattern ation of spindle cells displaying pale eosinophilic
(Fig. 1c, H&E) with infiltrative borders (Lakhani cytoplasm and slender nuclei with mild cytologic
et al. 2012). The nuclear atypia varies from mod- atypia (Fig. 1d H&E, and Fig. 2a and b, H&E)
erate to high grade. An inflammatory infiltrate of (Gobbi et al. 1999). The stroma shows varying
I
lymphocytes and dendritic cells is often found degree of collagenization. Focal plump fusiform
(Lakhani et al. 2012). and polygonal tumor cells, with more rounded
The presence of a carcinomatous component nuclei, arranged in “epithelioid” clumps can be
in the mixed tumors makes the diagnosis rela- observed. Spindle cells are often arranged in
tively straightforward. On the other side, the wavy interlacing fascicles (Lakhani et al. 2012).
diagnosis of high-grade spindle cell carcinoma In addition, foci of glandular or squamous ele-
with no morphological evidence of epithelial dif- ments associated with the spindle cells can be
ferentiation is challenging (Rakha et al. 2016). encountered posing issues in differential diagno-
This represents another scenario in which exten- sis with ▶ low grade adenosquamous carcinoma
sive sampling and thorough immunohistochemi- (see below); in fibromatosis-like carcinomas, foci
cal investigation with a panel of markers of glandular or squamous elements should repre-
including more than one cytokeratin are useful sent less than 5% of the overall tumor cell com-
(Collins and Schnitt 2013; Rakha et al. 2016). As ponent. Ductal carcinoma in situ (▶ Ductal
discussed above for metaplastic carcinomas with carcinoma in situ) can be associated (Gobbi
mesenchymal differentiation, foci of ductal car- et al. 1999).
cinoma in situ (▶ Ductal Carcinoma In Situ) or Cytokeratin expression is typically
small cohesive epithelial foci should be sought. found; however it is important to note that it
A pure spindle cell malignancy with no evidence can be focal and occasionally restricted to the
of these features or even focal cytokeratin plump spindle and more epithelioid cells.
expression should prompt to consider, in the dif- Expression of p63 is invariably observed in
ferential diagnosis, malignant phyllodes tumor these lesions.
(▶ Phyllodes Tumor), sarcomas, and metastatic
sarcomatoid tumors (Collins and Schnitt 2013; Low-Grade Adenosquamous Carcinoma
Rakha et al. 2016). This is a rare histologic type of metaplastic
A low-grade form of spindle cell carcinoma carcinoma showing a distinctive combination of
has also been described and is labeled as glandular and squamous differentiation. It is
“Fibromatosis-like Metaplastic Carcinoma” and characterized by well-developed gland/tubule
is discussed here below. formation intimately admixed with solid
nests of squamous cells in a spindle cell back-
Fibromatosis-like Metaplastic Carcinoma ground. Despite the presence of metaplastic
This tumor type, described by Gobbi et al. (1999), elements, these tumors display a low-grade
represents a low-grade variant of metaplastic histological pattern. In agreement with their
224 Invasive Metaplastic Carcinoma
Invasive Metaplastic Carcinoma, Fig. 2 A core biopsy atypia (A,B). The neoplastic cells show mild
sample showing a spindle cell proliferation in the mam- pancytokeratin expression (C), focal CK5/6 expression
mary gland featuring fusiform cells displaying pale eosin- (D), and more pervasive and intense CK14 expression
ophilic cytoplasm and slender nuclei with bland cytologic (E). The lesion shows diffuse p63 expression (F)
low-grade morphological features, the majority lymph node metastasis (Collins and Schnitt
of low-grade adenosquamous carcinomas exhibit 2013). A proportion of cases, however, can
an excellent prognosis, with a low incidence of behave in a locally aggressive manner.
Invasive Metaplastic Carcinoma 225
Invasive Metaplastic Carcinoma, Table 1 Expression carcinomas. This table has been modified and adapted
of CKs and epithelial differentiation markers in the main from Table 2 reported in Rakha et al. (2017). Number of
subgroups of metaplastic carcinomas. Data are extracted positive cases/total number of cases (percentages). LGASC
from a series of published cohorts of metaplastic low-grade adenosquamous carcinoma
Metaplastic carcinoma Fibromatosis-
Squamous with mesenchymal Spindle cell like
carcinoma differentiation carcinoma carcinoma LGASC
Cytokeratins AE1/ 11/13 (85) 36/36 (100) 99/117 (85) 30/30 (100) 13/14 (93)
AE3
CK8/18 17/18 (94) 7/13 (94) 14/54 (26) 1/2 (50) 18/21 (86)
CK7 // 4/4 (100) 0/2 (0) 0/25 (0) 21/22 (95)
CK19 // 5/7 (71) 0/2 (0) // 2/2 (100)
MNF116 // 7/10 (70) 38/40 (95) // 2/2 (100)
34bE12 // 10/15 (67) 7/9 (78) 28/28 (100) 23/23
(100)
CK5/6 59/63 (94) 41/56 (73) 25/34 (74) 6/6 (100) 25/27 (93)
CK14 17/19 (89) 38/52 (73) 35/43 (81) // 2/3 (67)
CK17 // 5/9 (56) 6/7 (86) // //
Myoepithelial p63 46/53 (87) 58/84 (69) 73/100 (73) 7/7 (100) 32/88 (84)
markers SMA 3/6 (50) 14/30 (47) 77/92 (84) 22/30 (73) 5/23 (22)
SMM // 2/16 (12) 1/10 (10) 0/24 (0) 0/19 (0)
CD10 // 9/18 (50) 14/15 (93) // 0/17 (0)
Calponin // 24/30 (80) 1/7 (14) // 0/7 (0)
S100 // 59/63 (94) 32/86 (37) 1/5 (20) //
Epithelial EGFR 95/114 47/76 (62) 35/49 (71) // //
differentiation (83)
EMA 0/3 (0) 14/15 (93) 32/101 (32) // //
E-CAD 18/20 (90) 0/19 (0) 0/10 (0) 0/3 (0) //
mutations in genes of the PI3K/AKT/mTOR path- differentiation and is associated with conven-
way and of the Wnt pathway, respectively tional mammary invasive or in situ carcinoma,
(Ng et al. 2017). These data are of clinical interest the diagnosis of a metaplastic carcinoma is usu-
as they provide a molecular basis for the recent ally straightforward. If these features are absent,
preclinical and clinical observations that Wnt and evidence for epithelial differentiation of the neo-
PI3K/AKT/mTOR pathway inhibition may be plastic cells should be provided by using immu-
beneficial for a subset of patients with metaplastic nohistochemistry (Fig. 2) (Rakha et al. 2017).
carcinoma (Ng et al. 2017). A wide spectrum of benign, locally aggressive,
and highly aggressive lesions has to be taken
in account for the differential diagnosis (Rakha
Differential Diagnosis et al. 2017). For instance, in order to render
a diagnosis of primary squamous cell carcino-
Due to the rarity and the histological diversity of mas of the breast, a squamous cell carcinoma
metaplastic carcinoma of the breast, its diagnosis of other sites, and in particular of the skin and
in routine diagnostic practice can be sometimes of the lung, should be ruled out (Lakhani
challenging (Rakha et al. 2017). et al. 2012).
As a general rule, if an invasive lesion of the Clinical history and radiological findings
breast shows mesenchymal or squamous would be crucial in this scenario. However, if
Invasive Metaplastic Carcinoma 227
Lehmann, B. D., Jovanović, B., Chen, X., Estrada, M. V., Weigelt, B., Kreike, B., & Reis-Filho, J. S. (2009). Meta-
Johnson, K. N., Shyr, Y., Moses, H. L., Sanders, M. E., plastic breast carcinomas are basal-like breast cancers:
& Pietenpol, J. A. (2016). Refinement of triple-negative A genomic profiling analysis. Breast Cancer Research
breast cancer molecular subtypes: Implications for neo- and Treatment, 117(2), 273–280.
adjuvant chemotherapy selection. PLoS One, 11(6), Weigelt, B., Ng, C. K., Shen, R., Popova, T., Schizas, M.,
e0157368. Natrajan, R., Mariani, O., Stern, M. H., Norton, L.,
Lien, H. C., Lin, C. W., Mao, T. L., Kuo, S. H., Hsiao, C. H., Vincent-Salomon, A., & Reis-Filho, J. S. (2015). Meta-
& Huang, C. S. (2004). p53 overexpression and mutation plastic breast carcinomas display genomic and trans-
in metaplastic carcinoma of the breast: Genetic evidence criptomic heterogeneity. Modern Pathology, 28(3),
for a monoclonal origin of both the carcinomatous and 340–351.
the heterogeneous sarcomatous components. The Jour- Wendroth, S. M., Mentrikoski, M. J., & Wick, M. R.
nal of Pathology, 204(2), 131–139. (2015). GATA3 expression in morphologic subtypes
Marchio, C., Geyer, F. C., & Reis-Filho, J. S. (2016). Pathol- of breast carcinoma: A comparison with gross cystic
ogy and molecular pathology of breast cancer. In M. Loda disease fluid protein 15 and mammaglobin. Annals of
et al. (Eds.), Pathology and epidemiology of cancer Diagnostic Pathology, 19(1), 6–9.
(pp. 173–232). Cham: Springer International Publishing.
Ng, C. K. Y., Piscuoglio, S., Geyer, F. C., Burke, K. A.,
Pareja, F., Eberle, C. A., Lim, R. S., Natrajan, R.,
Riaz, N., Mariani, O., Norton, L., Vincent-Salomon, A.,
Wen, Y. H., Weigelt, B., & Reis-Filho, J. S. (2017). The
landscape of somatic genetic alterations in metaplastic Invasive Micropapillary
breast carcinomas. Clinical Cancer Research, 23(14), Carcinoma
3859–3870.
Pezzi, C. M., Patel-Parekh, L., Cole, K., Franko, J.,
Klimberg, V. S., & Bland, K. (2007). Characteristics Lilla Madaras and Janina Kulka
and treatment of metaplastic breast cancer: Analysis of 2nd Department of Pathology, Semmelweis
892 cases from the National Cancer Data Base. Annals University, Budapest, Hungary
of Surgical Oncology, 14(1), 166–173.
Tanabe, Y., Tsuda, H., Yoshida, M., Yunokawa, M.,
Yonemori, K., Shimizu, C., Yamamoto, S., Kinoshita, T.,
Fujiwara, Y., & Tamura, K. (2017). Pathological fea- Synonyms
tures of triple-negative breast cancers that showed pro-
gressive disease during neoadjuvant chemotherapy. Micropapillary carcinoma
Cancer Science, 108(7), 1520–1529.
Provenzano, E., Byrne, D. J., Russell, P. A., Wright, G. M.,
Generali, D., & Fox, S. B. (2016). Differential expres-
sion of immunohistochemical markers in primary lung Definition
and breast cancers enriched for triple-negative tumours.
Histopathology, 68(3), 367–377.
Rakha, E. A., Aleskandarany, M. A., Lee, A. H., & Invasive micropapillary carcinoma (IMPC) is a
Ellis, I. O. (2016). An approach to the diagnosis of special type of invasive breast carcinoma (IBC)
spindle cell lesions of the breast. Histopathology, characterized by cancer cells with eosinophilic or
68(1), 33–44. granular cytoplasm, forming morule-like clus-
Rakha, E. A., Coimbra, N. D., Hodi, Z., Juneinah, E.,
Ellis, I. O., & Lee, A. H. (2017). Immunoprofile of ters, surrounded by clear stromal spaces. Tumor
metaplastic carcinomas of the breast. Histopathology, cells have characteristic reverse polarity, also
70(6), 975–985. known as an “inside-out” growth pattern: the
Reis-Filho, J. S., Milanezi, F., Steele, D., Savage, K., apical pole of tumor cells faces the empty stromal
Simpson, P. T., Nesland, J. M., Pereira, E. M.,
Lakhani, S. R., & Schmitt, F. C. (2006). Metaplastic spaces.
breast carcinomas are basal-like tumours. Histopathol-
ogy, 49(1), 10–21.
Reyes, C., Gomez-Fernández, C., & Nadji, M. (2012).
Metaplastic and medullary mammary carcinomas
Clinical Features
do not express mammaglobin. American Journal of
Clinical Pathology, 137(5), 747–752. • Incidence
Sneige, N., Yaziji, H., Mandavilli, S. R., Perez, E. R., Pure IMPC is a rare subtype of invasive breast
Ordonez, N. G., Gown, A. M., & Ayala, A. (2001).
Low-grade (fibromatosis-like) spindle cell carcinoma
cancer, accounting in its pure form for 0.9–2%
of the breast. American Journal of Surgical Pathology, of IBCs. The reported incidence of IBC with a
25(8), 1009–1016. micropapillary component is up to 8%.
Invasive Micropapillary Carcinoma 229
• Age Microscopy
The reported median age of IMPC is from
48 to 62 years, consistent with that of IMPC is characterized by cell clusters arranged
patients with estrogen receptor (ER) in pseudopapillary structures, devoid of fibrovas-
positive IBC. cular cores and surrounded by clear stromal spaces,
• Sex delineated by delicate fibers of a fibro-collagenous
IMPC usually affects women, but single cases stroma (Fig. 1a. H&E: shows the low power pattern
of male IMPC had been reported. In studies on of IMPC). The empty spaces surrounding the
male breast cancer, the reported incidence of tumor cell morules resemble lymphatic channels
IBC with micropapillary features varies but lack endothelial lining (Fig. 1b. H&E: lack of
between 0% and 11%. endothelial lining of stromal spaces).
• Site The amount of a micropapillary component
No specific site within the breast parenchyma required for the diagnosis of IMPC in a tumor is
was identified for these tumors. not yet established. Some authors have used the
• Treatment diagnosis of IMPC in tumors with at least a 50%
Currently stage I and II tumors are treated as micropapillary component, while others require
IBC of no special type (IBC NST) (▶ Invasive that the entire tumor should demonstrate the char-
I
Carcinoma NST); however there are some sug- acteristic micropapillary growth pattern in order
gestions that modification of current to be diagnosed as pure IMPC. For practical
locoregional treatment would be beneficial in reasons at least 75% of the tumor should demon-
cases of IMPC: wider surgical margins and strate the characteristic micropapillary growth in
more aggressive axillary and supraclavicular order to be characterized as pure IMPC. There are
management would enhance local disease con- some suggestions that IBC with any well-defined
trol (Yu et al. 2015). micropapillary carcinoma component should pro-
• Outcome mpt the diagnosis of IMPC and the percentage of
Since the incidence of IMPC is low, studies the micropapillary component clearly stated in the
on its outcome are sparse. It seems that pathology report (Yi-Ling et al. 2016) (Fig. 2.
despite the reported unfavorable tumor char- H&E: mixed IBC NST and IMPC).
acteristics of IMPC (higher nuclear grade, The tumor cells of IMPC usually have eosino-
frequent lymphovascular invasion (LVI), philic cytoplasm, and they demonstrate a charac-
lymph node metastases (LNM), and extra- teristic reverse polarity (“inside-out” pattern): the
capsular spread) (Chen et al. 2014; Yu et al. apical surface of the cells facing the empty stromal
2015) the overall prognosis of IMPC patients spaces and not the central pseudolumen, if pre-
in multivariate analysis – when stratified for sent. In some cases apocrine features can be iden-
number of positive lymph nodes and other tified. True tubule formation is lacking; nuclear
prognostic factors – is comparable to that of pleomorphism is only rarely pronounced. Number
IBC NST patients (Yu et al. 2015). of mitoses is usually low to moderate. Most
According to Chen’s study of 624 patients IMPCs are grade 2 or 3 lesions. Necrosis, tumor
with IMPC, the prognosis is poor if the tumor infiltrating lymphocytes (TILs) are not common.
is ER negative. Peritumoral LVI is detected in up to 70% of
IMPC cases, while the LVI rate of IBC NST cases
is 20% (Gruel 2014). It seems that neither the
Macroscopy tumor size alone nor the extent of the micro-
papillary component but the presence of the
IMPCs do not have any particular features on micropapillary pattern correlates with the agg-
grossing. The reported mean tumor size is between ressive locoregional presentation of the tumor
1.5 cm and 3.9 cm, not significantly different from (Yi-Ling et al. 2016).
that of IBC NST (▶ Invasive Carcinoma NST) The majority of patients – up to 84% – present
(Yi-Ling et al. 2016). with axillary LNM at initial diagnosis. This rate is
230 Invasive Micropapillary Carcinoma
Invasive Micropapillary Carcinoma, Fig. 1 A: Low power pattern of IMPC (H&E); B: Lack of endothelial lining of
stromal spaces around the morule-like clusters formed by tumor cells (H&E)
significantly higher than that of IBC NST patients American Pathologists (CAP) 2013 guidelines for
with LNM, and the number of lymph nodes HER2 testing suggest that micropapillary carci-
involved by metastatic disease is also higher in noma with HER2 immunohistochemistry staining
IMPC than in IBC NST cases. that is intense but incomplete (basolateral or
Associated intraductal carcinoma (DCIS) U-shaped) and therefore would be considered
(▶ Ductal Carcinoma In Situ) is usually of micro- score 1+ may actually be amplified by fluorescent
papillary architecture with high-grade nuclei. in situ hybridization (FISH). Thus the guideline
Necrosis and microcalcifications may also be recommends that these IMPC cases should be
present. reported as equivocal (score 2+), and alternative
testing should be carried out. This recommendation
of the 2013 ASCO/CAP guideline was based solely
Immunophenotype on the results of a single study, which was
reinforced later by others.
IMPC is frequently positive for ER (66–94%) and Epithelial membrane antigen (EMA) or mucin
progesterone receptor (PR) (50–84%). The prog- 1 (MUC1 [CD227]) is a high-molecular weight
nosis of ER-negative IMPCs is usually poor. In (>400 kDa), type I membrane-tethered glycopro-
reported series to date, basal cytokeratins are not tein that may serve to demonstrate the reverse
expressed in IMPCs. Pure IMPCs are likely to polarity of IMPC cells by staining the apical sur-
have high Ki67 index, and cyclin D1 is also highly face of tumor cells facing the empty stromal
expressed (Marchiò et al. 2008). spaces (Fig. 3a: EMA immunohistochemistry
The reported incidence of HER2-positive IMPC showing positive reaction at the periphery of
is inconsistent, which varies between 8.3% and the morule-like micropapillary tumor cell clusters
95% (Yi-Ling et al. 2016). The American Society highlighting their inverse polarity in a pure IMPC
of Clinical Oncology (ASCO) and the College of case and Fig. 3b, in the IMPC component of a
Invasive Micropapillary Carcinoma 231
Invasive Micropapillary Carcinoma, Fig. 3 EMA of cancer cells and (B) of a mixed IBC NST and IMPC,
immunohistochemical reaction (A) of a pure IMPC with where the characteristic pattern of EMA positivity high-
EMA positivity at the periphery of the morule-like micro- lights the IMPC component of the tumor
papillary tumor cell clusters showing the inverse polarity
Invasive Micropapillary Carcinoma, Fig. 4 A: IMPC with mucinous differentiation (H&E); B: Invasive mucinous
carcinoma with IMPC growth pattern (IMMPC) (H&E)
Invasive Micropapillary Carcinoma 233
(IMMPC) (Fig. 4b H&E) are relatively newly References and Further Reading
described entities.
Serous ovarian carcinoma metastatic to the breast Chen, A. C., Paulino, A. C., Schwartz, M. R.,
Rodriguez, A. A., Bass, B. L., Chang, J. C., &
may mimic the histological features of IMPC and
Teh, B. S. (2014). Population-based comparison of
usually also displays lymphatic tumor emboli. How- prognostic factors in invasive micropapillary and inva-
ever, the presence of psammoma bodies and the sive ductal carcinoma of the breast. British Journal of
immunoreactivity for WT1 together with the clinical Cancer, 111, 619–622.
Fisher, E. R., Palekar, A. S., Redmond, C., Barton, B., &
history may assist to make the right diagnosis. The
Fisher, B. (1980). Pathologic findings from the
caveat here is that IMPCs may express WT1, albeit National Surgical Adjuvant Breast Project (protocol
infrequently (Lee et al. 2007). no. 4). VI. Invasive papillary cancer. American Journal
IMPC should be distinguished from IBC NST of Clinical Pathology, 73, 313–322.
Gruel, N., Benhamo, V., Bhalshankar, J., Popova, T.,
(▶ Invasive Carcinoma NST) with widespread
Freneaux, P., Arnould, L., Mariani, O., Stern, M. H.,
lymphovascular invasion. Although the clear Raynal, V., Sastre-Garau, X., Rouzier, R., Delattre, O.,
spaces around the morule-like clusters of IMPC & Vincent-Salomon, A. (2014). Polarity gene alter-
cells resemble lymphatic channels, they are ations in pure invasive micropapillary carcinomas of
the breast. Breast Cancer Research, 16, R46. https://
devoid of endothelial cell lining. The characteris-
doi.org/10.1186/bcr3653
tic reverse polarity of IMPC cells is usually not Lee, A. H., Paish, E. C., Marchio, C., Sapino, A., I
observed in IBC NST. Schmitt, F. C., Ellis, I. O., & Reis-Filho, J. S. (2007).
It is important that widespread retraction arti- The expression of Wilms’ tumour-1 and Ca125 in
invasive micropapillary carcinoma of the breast.
fact be not overdiagnosed as IMPC.
Histopathology, 51(6), 824–828.
Li, S., Yang, C., Zhai, L., Zhang, W., Yu, J., Gu, F.,
Fact sheet
Lang, R., Fan, Y., Gong, M., Zhang, X., & Fu, L.
Definition (2012). Deep sequencing reveals small RNA charac-
Rare type of invasive breast cancer displaying tumor terization of invasive micropapillary carcinomas of the
cell clusters in empty spaces. The tumor cells have breast. Breast Cancer Research and Treatment, 136(1),
inverse polarity. 77–87.
Microscopy Marchiò, C., Iravani, M., Natrajan, R., Lambros, M. B.,
The inverse polarity of tumor cells can be Savage, K., Tamber, N., Fenwick, K., Mackay, A.,
demonstrated by EMA immunohistochemistry. Senetta, R., Di Palma, S., Schmitt, F. C., Bussolati, G.,
The cells may form morule-like clusters or Ellis, I. O., Ashworth, A., Sapino, A., & Reis-Filho, J. S.
pseudotubules. (2008). Genomic and immunophenotypical characteri-
Lymphovascular invasion is a common feature. zation of pure micropapillary carcinomas of the breast.
Immunohistochemistry The Journal of Pathology, 215, 398–410.
IMPCs are usually ER/PR positive. Some cases belong Marchiò, C., Iravani, M., Natrajan, R., Lambros, M. B.,
to the HER2-positive subtype. Few IMPCs belong to the Geyer, F. C., Savage, K., Parry, S., Tamber, N.,
triple-negative non-basal group of breast carcinomas. Fenwick, K., Mackay, A., Schmitt, F. C., Bussolati, G.,
Ellis, I., Ashworth, A., Sapino, A., & Reis-Filho, J. S.
Molecular features
(2009). Mixed micropapillary-ductal carcinomas of the
IMPCs are characterized by genetic heterogeneity.
breast: A genomic and immunohistochemical analysis
Gains and losses of large chromosomal segments are
of morphologically distinct components. The Journal
common. Genes responsible for cell polarity were found
of Pathology, 218(3), 301–315.
to be downregulated.
Natrajan, R., Wilkerson, P. M., Marchiò, C., Piscuoglio, S.,
Treatment Ng, C. K., Wai, P., Lambros, M. B., Samartzis, E. P.,
Wide excision with sentinel lymph node (▶ Sentinel Dedes, K. J., Frankum, J., Bajrami, I., Kopec, A.,
Node) biopsy. IMPCs are more likely to develop axillary Mackay, A., A’Hern, R., Fenwick, K., Kozarewa, I.,
LN metastases than IBC NSTs. Hakas, J., Mitsopoulos, C., Hardisson, D., Lord, C. J.,
Outcome Kumar-Sinha, C., Ashworth, A., Weigelt, B., Sapino, A.,
According to recent large studies with follow-up data, the Chinnaiyan, A. M., Maher, C. A., & Reis-Filho, J. S.
outcome is not worse than that of stage-matched ER-positive (2014). Characterization of the genomic features and
IBC NSTs. ER-negative IMPCs have poor prognosis. expressed fusion genes in micropapillary carcinomas of
Differential diagnosis the breast. The Journal of Pathology, 232(5), 553–565.
Widespread lymphovascular invasion in IBC NST Peterse, J. L. (1993). Breast carcinomas with an
Mucinous carcinoma with micropapillary features unexpected inside-out growth pattern: rotation of
Metastatic serous papillary ovarian carcinoma polarization associated with angioinvasion. (Abstract).
Retraction artifact in poorly fixed specimens Pathology Research and Practice, 189, 780.
234 Invasive Mucinous Carcinoma
Siriaunkgul, S., & Tavassoli, F. A. (1993). Invasive micro- diagnosis of MC, the mucinous component should
papillary carcinoma of the breast. Modern Pathology, represent over 90% of the tumor (Harris
6, 660–662.
Wang, X. X., Liu, B. B., Wu, X., Su, D., Zhu, Z., & Fu, L. et al. 2011).
(2015). Loss of leucine zipper putative tumor supp-
ressor 1 (LZTS1) expression contributes to lymph
node metastasis of breast invasive micropapillary car- Clinical Features
cinoma. Pathology Oncology Research: POR, 21(4),
1021–1026.
Weigelt, B., Horlings, H. M., Kreike, B., Hayes, M. M., • Incidence
Hauptmann, M., Wessels, L. F. A., de Jong, D., Van de Pure mucinous carcinoma (PMC) of the breast
Vijver, M. J., Van’t Veer, L. J., & Peterse, J. L. (2008). represents one of the rarest subgroups (Park
Refinement of breast cancer classification by molecular
characterization of histological special types. The Jour- et al. 2010) and accounts for approximately
nal of Pathology, 216, 141–150. 2% of all breast carcinomas (Bussolati and
Yi-Ling, Y., Bing-Bing, L., Xinmin, Z., & Li, F. (2016). Sapino 2012; Harris et al. 2011).
Invasive micropapillary carcinoma of the breast: An • Age
update. Archives of Pathology and Laboratory Medi-
cine, 140, 799–805. Compared to conventional invasive breast
Yu, J. I., Choi, D. H., Huh, S. J., Cho, E. Y., Kim, K., carcinoma, PMC tends to affect an older pop-
Chie, E. K., Ha, S. W., Park, I. A., Ahn, S. J., Lee, J. S., ulation, typically in the postmenopausal age
Shin, K. H., Kwon, Y., Kim, Y. B., Suh, C. O., group (Park et al. 2010).
Koo, J. S., Kim, J. H., Jeong, B. G., Kim, I. A.,
Lee, J. H., & Park, W. (2015). Differences in prognostic • Sex
factors and failure patterns between invasive micro- While MC of the breast primarily affects
papillary carcinoma and carcinoma with micropapillary women, isolated case reports of male breast
component versus invasive ductal carcinoma of the MC exist. Furthermore, some of these
breast: Retrospective Multicenter Case-Control Study
(KROG 13-06). Clinical Breast Cancer, 15, 353-361. cases of male breast MC have presented
e351-352. with metastasis to axillary lymph nodes.
Metastasis to the lung has also been
documented.
• Symptoms and Imaging Findings
Invasive Mucinous Carcinoma The presentation is similar to other breast carci-
nomas. Patients typically present with a palpable
James S. DeGaetano and Ian Said Huntingford mass. Not surprisingly, the radiological findings
Pathology Department, Mater Dei Hospital, can mimic those of a benign neoplasm, typically
Msida, Malta presenting as a lobulated and well-circumscribed
lesion (Park et al. 2010). The majority of tumors
show high signal intensity on T2 weighted and
Synonyms short tau inversion recovery (STIR) magnetic
resonance imaging (MRI). This is due to the
Colloid carcinoma; Gelatinous carcinoma; high water content owing to the copious amounts
Mucinous adenocarcinoma; Mucoid carcinoma of extracellular mucin seen in such lesions.
• Treatment
By definition, these are rare cancers hence most
Definition of the data on management is based on smaller
case series as well as isolated case reports. Man-
Mucinous carcinoma (MC) of the breast forms part agement is usually no different to the more
of the special type of breast cancers and common invasive breast carcinoma of no spe-
is characterized histologically by aggregates or cial type (IBC NST) (▶ Invasive Carcinoma
clusters of typically uniform cells floating within NST). Breast conserving therapy is typically
copious amounts of extracellular mucin (Bussolati undertaken, interestingly with a similar rate of
and Sapino 2012). In order to qualify for a local recurrence compared to IBC NST. Since
Invasive Mucinous Carcinoma 235
PMC is regarded to have a good prognosis, the size does not have such an important impact on
need to undergo axillary dissection was histori- overall prognosis, since there typically is over-
cally a rather contentious issue; although, this is estimation of tumor size owing to the presence
less so given that most patients undergo sentinel of abundant extra-cellular mucin. Other studies
node (▶ Sentinel Node) biopsy in the first (Di Saverio et al. 2008) still utilize tumor size as
instance, ultimately dictating whether axillary an independent prognostic factor, although it
dissection is required (Dieci et al. 2014). The has less relevance compared to nodal status
National Comprehensive Cancer Network and stage. When compared to IBC NST
(NCCN) guidelines state that adjuvant endo- (▶ Invasive Carcinoma NST), MC displays bet-
crine therapy should be considered if the case ter disease-free survival; however, overall sur-
is hormone receptor (HR) (▶ Hormone Recep- vival appears to be similar.
tors in Breast Cancer) positive, node negative A study (Tseng et al. 2013) retrospectively
(or nodal metastasis size is equal to or less than reviewed 93 cases of PMC and amongst other
2 mm), and the tumor size is less than 3 cm. If aspects looked at the effects that the various
the tumor size is less than 1 cm, then adjuvant clinicopathological features had on disease-
endocrine therapy is mainly indicated for risk free and overall survival. These factors
reduction. If the tumor size is equal to or greater included age, tumor size, grade (I vs. II), HR
I
than 3 cm, then endocrine therapy is definitely status, HER2 status, primary treatment status
recommended. If the tumor is HR positive yet (mastectomy vs. lumpectomy with and without
there is ipsilateral axillary nodal involvement radiotherapy), as well as systemic therapy
(one or more ipsilateral metastases greater than (endocrine therapy alone vs. with chemother-
2 mm), then adjuvant chemotherapy may be apy). A univariate analysis of these character-
indicated in addition to adjuvant endocrine ther- istics interestingly revealed that none of these
apy. In the context of HR negative cases, the HR factors had an impact on survival.
status should be re-evaluated, and if it is It is important to distinguish between PMC
reconfirmed that the case is HR negative, then and mixed mucinous carcinoma (MMC), since
the treatment follows that of IBC NST (NCCN the prognosis between the two is different. Stud-
2017). ies have estimated the 10 year survival rate of
• Outcome PMC to be 90.4% while that of MMC to be 66%.
PMC has an excellent prognosis with a low rate The importance of this distinction is further
of local recurrence and a 5-year disease-free highlighted since MMC demonstrates a higher
survival rate ranging from 81% to 94%. The rate of recurrence. The study also confirmed that
disease-specific survival rate is quoted at in particular MC, Type A, with a high mucin
95.3%, while the overall 5-year survival rate is content has a favorable prognosis. When dealing
80%. Lymph node involvement plays an impor- with Type B MC, treatment should be based on
tant role in determining survival rates with the whether there is lymph node involvement or not.
overall 5-year survival rate increasing to 86% if As shall be eluded to later, there is no significant
there is no nodal involvement (Harris et al. difference in survival between the two main his-
2011). Case series have estimated the axillary tological types of PMC: Type A and Type B.
lymph node involvement rate at about 12%
(Di Saverio et al. 2008). The greater the nodal
involvement, the higher the likelihood of death Macroscopy
from malignancy and the greater the risk of
tumor recurrence (Harris et al. 2011). Tumor The typical appearance is that of a gelatinous
size has an impact on the incidence of lymph lesion reflecting the high mucinous component
node involvement, with tumors less than 10 mm (Bussolati and Sapino 2012) (Fig. 1). The tumor
displaying a low risk of lymph node metastasis, size typically ranges from 3 to 120 mm and has an
namely, 0–4%. Some reports show that tumor average dimension of 20 mm (Harris et al. 2011).
236 Invasive Mucinous Carcinoma
Histopathology
MC of the breast is characterized by copious pools
of extraceullar mucin-containing free floating
tumor cells typically arranged as nests, trabeculae,
sheets, or acini. The nuclear grade is usually low
to intermediate (Harris et al. 2011); however, rare
cases with pronounced atypia and mitoses have
been reported (Bussolati and Sapino 2012).
PMC has been traditionally subtyped into Type
A and Type B. Type A is described as the hypo-
cellular variant and displays a variety of architec-
tural pattern ranging from cribriform, tubular,
cord like, papillary, as well as micropapillary
(Fig. 3). Type B is the hypercellular variant and
grows in solid nests (Fig. 3). Type B mucinous
carcinomas often show neuroendocrine differen-
tiation (Capella et al. 1980). The morphological
similarities are also confirmed at molecular level
(Weigelt et al. 2009).
MMC is a well-described entity, and the most
common admixture is with IBC NST (▶ Invasive
Carcinoma NST) and lobular carcinoma
(▶ Invasive Lobular Carcinoma) (Fig. 3).
A particular histological variant, namely, mucin-
ous micropapillary carcinoma (▶ Invasive Micro-
papillary Carcinoma), is associated with higher
Invasive Mucinous Carcinoma, Fig. 1 Classic macro-
scopic appearance of mucinous carcinoma. One can
rates of lymphovascular invasion, lymph node
appreciate the mucinous cut surface involvement, and greater expression of human epi-
dermal growth factor receptor 2 (HER2) positivity.
This entity is characterized by the coexistence of
Microscopy mucinous and micropapillary morphology. Charac-
teristic histological features of this entity include
Role of Fine Needle Aspiration (FNA) Biopsy in the presence of a micropapillary pattern, a mucin-
Diagnosis of Mucinous Carcinoma ous appearance, psammomatous calcifications,
Fine needle aspiration can play an important role hobnailing, and an intermediate/high grade nuclear
in the diagnosis of both PMC and MMC (Cyrta cytology. The study (Barbashina et al. 2013)
et al. 2013). The most common features encoun- described 15 cases and found that 60% of cases
tered on cytology include copious mucin and showed lymphovascular invasion, with 33%
bland nuclear cytology (regular nuclear con- of patients displaying synchronous axillary lymph
tours, small nuclear size) (Fig. 2). If the lesion node involvement.
is mixed, one can expect sparser mucin content
and more pronounced nuclear cytological fea-
tures such as larger nuclei, nuclear contour irreg- Immunophenotype
ularity, and prominent nucleoli. The presence of
necrosis is also suggestive of a mixed mucinous The receptor status profile of MC has also
component. been studied quite intensely. It is typically
Invasive Mucinous Carcinoma 237
Invasive Mucinous Carcinoma, Fig. 2 FNA of mucinous carcinoma (H&E, clockwise, x4, x10, x20, x40). Groups of
fairly monomorphic tumor cells float in a sea of mucinous material
Invasive Mucinous Carcinoma, Fig. 3 Types and pat- C), mucinous carcinoma solid papillary architecture (H&E,
terns of mucinous carcinoma. Mucinous carcinoma B), mucinous carcinoma with invasive breast carcinoma
Type A (H&E, A), mucinous carcinoma Type B (H&E, (H&E, D)
Invasive Mucinous Carcinoma, Fig. 4 Diffuse, strong ER positivity (A); diffuse, strong PR positivity (B); HER2
negative expression (C); unusual HER2 expression in Type B mucinous carcinoma (D)
mucinous carcinoma since it is believed that differences in MUC protein expression profiles
MUC2 has tumor suppressor activity and forms between the two types of MC. Type A breast
a rather viscid tumor environment perhaps mucinous carcinoma shows luminal and apical
inhibiting spread of cancer cells. There are even expression of MUC proteins, while the expression
Invasive Mucinous Carcinoma 239
Invasive Mucinous Carcinoma, Fig. 5 Mucinous (B). Mucinous carcinoma Type B neuroendocrine marker
carcinoma Type A Ki67 proliferation index (A). expression (Synaptophysin (C) and Chromogranin (D))
Mucinous carcinoma Type B Ki67 proliferation index
profile in Type B is membrano-cytoplasmic. Inter- breast MC, which is a similar finding in various
estingly MUC2 expression is inversely associated other types of cancers including prostate, colon,
with the rate of lymph node involvement and bladder, amongst others (Feng et al. 2013).
lymphovascular invasion in non-mucinous inva- miR-143 downregulation is also associated with
sive carcinomas. MUC6 expression is also asso- mucinous phenotype within colon cancer, hence it
ciated with MC (Rakha et al. 2005). is plausible to consider miR-143 downregulation
The pathogenesis of MC is also thought to as an important common factor in the mucinous
result from altered expression of MUC2 within cancer pathway. Interestingly, unlike in colonic
normal breast tissue. The study also concluded mucinous cancer, microsatellite instability is not
that out of all mucins it is MUC1 and MUC3 a prevalent occurrence (Hugen et al. 2014).
which have the most profound impact on out- Studies have shown that specialized breast
come. MUC1 expression appears to be associated cancers typically adopt a particular category of
with a better prognosis mostly because it is molecular profiling with mucinous cancer
associated with more well differentiated neo- expressing a luminal phenotype (Weigelt et al.
plasms. Tumors expressing MUC3 (membranous 2010). As has already been stated, there is consid-
staining) tend to have a higher rate of lymph node erable overlap between Type B mucinous carci-
involvement and local recurrence as well as being noma and neuroendocrine carcinoma (▶ Invasive
associated with poorer histological parameters Carcinoma with Neuroendocrine Differentiation),
(such as worse grade) (Rakha et al. 2005). and this is mirrored at the genetic level, with both
types of tumors exhibiting identical genomic
Molecular Profile profiles (Weigelt et al. 2009). They are also
MicroRNA has been the subject of intense genomically distinct from conventional IBC
research and is enabling us to better understand NST (▶ Invasive Carcinoma NST) (Lacroix-
the molecular pathways involved in cancer. There Triki et al. 2010). The pure variant typically
is downregulation of miR-143 and miR-224-5p in shows genetic stability and characteristically
240 Invasive Mucinous Carcinoma
lacks 1q gain and 16q loss which is typically seen Hruban, R. H., & Klimstra, D. S. (2003). Pathogenesis
in low grade IBC NST (Lacroix-Triki et al. 2010; of colloid (pure mucinous) carcinoma of exocrine
organs: Coupling of gel-forming mucin (MUC2)
Dieci et al. 2014; Hugen et al. 2014). They also production with altered cell polarity and abnormal
demonstrate a low incidence of PIK3CA muta- cell-stroma interaction may be the key factor in the
tions (Dieci et al. 2014). Studies which carried out morphogenesis and indolent behavior of colloid carci-
hierarchical molecular clustering revealed that noma carcinoma in the breast and pancreas. American
Journal of Surgical Pathology, 27, 571–578.
even mixed mucinous carcinomas are Barbashina, V., Corben, A. D., Akram, M., Vallejo, C., & Tan,
genomically very similar to pure mucinous carci- L. K. (2013). Mucinous micropapillary carcinoma of
noma rather than IBC NSTs (Lacroix-Triki the breast: An aggressive counterpart to conventional
et al. 2010). pure mucinous tumors. Human Pathology, 44,
1577–1585.
Bussolati, G., & Sapino, A. (2012). Mucinous carcinoma
and carcinomas with signet-ring-cell differentiation.
Differential Diagnosis In S. R. Lakhani, I. O. Ellis, S. J. Schnitt, P. H. Tan, &
M. J. van de Vijver (Eds.), WHO classification of
tumours of the breast (pp. 60–61). Lyon: IARC Press.
The differential diagnosis of mucinous carcinoma Capella, C., Eusebi, V., Mann, B., & Azzopardi, J. G.
typically includes the following: Mucocele-like (1980). Endocrine differentiation in mucoid carcinoma
lesion (▶ Mucocele-like Lesion), Mucinous of the breast. Histopathology, 6, 174–188.
DCIS, CIS with mucocele-like lesion and signet Cyrta, J., Andreiuolo, F., Azoulay, S., Balleyguier, C.,
Bourgier, C., Mazouni, C., Mathieu, M. C.,
ring cell carcinoma (▶ Invasive Carcinoma with Delaloge, S., & Vielh, P. (2013). Pure and mixed
Signet Ring Cell Differentiation). A newly mucinous carcinoma of the breast: Fine needle aspira-
described entity of invasive lobular carcinoma tion cytology findings and review of the literature.
(ILC) (▶ Invasive Lobular Carcinoma) with Cytopathology, 24, 377–384.
De Andrade, N. R., Derchain, S. F., Pavanello, M.,
extracellular mucin production certainly enters Paiva, G. R., Sarian, L. O., & Vassallo, J. (2017).
into the differential diagnosis. This entity can be Expression of unusual immunohistochemical markers
easily confused with mucinous carcinoma in mucinous breast carcinoma. Acta Histochemica, 119,
exhibiting a separate lobular component. Obvi- 327–336.
Di Saverio, S. D., Guitierrez, J., & Avisar, E. (2008).
ously if one overlooks the lobular component, A retrospective review with long term follow up of
then an incorrect diagnosis of PMC can easily be 11,400 cases of pure mucinous breast carcinoma.
made. There do exist a number of useful histolog- Breast Cancer Reserch and Treatment, 111, 541–547.
ical clues when making a diagnosis of this entity. Dieci, M. V., Orvieto, E., Dominici, M., Conte, P., &
Guarnieri, V. (2014). Rare breast cancer subtypes:
These include the presence of classical ILC Histological, molecular,and clinical peculiarities.
(▶ Invasive Lobular Carcinoma) around and not The Oncologist, 19, 805–881.
necessarily within the mucinous lakes. The iden- Feng, Z., Shuai, L., Meng, H.-M., Li-Qiang, Q., &
tification of lobular neoplasia (▶ Lobular In Situ Lin, G. (2013). MicroRNA and histopathological
characterization of pure mucinous breast carcinoma.
Neoplasia) is another helpful feature. Also impor- Cancer Biology and Medicine, 10, 22–27.
tant is the lack of E-cadherin staining, and cyto- Harris, G. C., O’Malley, F. P., & Pinder, S. E. (2011).
plasmic expression of p120 by tumor cells in the Invasive carcinoma: Special Types. In F. P. O’Malley,
mucinous areas. One may also include solid pap- S. E. Pinder, & A. M. Mulligan (Eds.), Breast
pathology: A volume in the foundations in diagnostic
illary carcinoma (SPC) (▶ Solid Papillary Carci- pathology series (2nd ed., pp. 235–237). London:
noma) in the differential diagnosis since there may Elsevier Saunders.
be co-existing mucin, and it is not unusual for Hugen, N., Simons, M., Halilovic, A., Van der Post, R. S.,
SPC to demonstrate neuroendocrine differentia- Bogers, A. J., Marijnissen-Van Zanten, M. A. J., de Wilt,
J. H. W., & Nagtegaal, I. D. (2014). The molecular
tion similar to Type B MC. background of mucinous carcinoma beyond MUC2.
The Journal of Pathology: Clinical Research, 1, 3–17.
Lacroix-Triki, M., Suarez, P. H., MacKay, A.,
Lambros, M. B., Natrajan, R., Savage, K.,
References Geyer, F. C., Weigelt, B., Ashworth, A., & Reis-Filho,
J. S. (2010). Mucinous carcinoma of the breast
Adsay, N. V., Merati, K., Nassar, H., Shia, J., Sarkar, F., is genomically distinct from invasive ductal carcinomas
Pierson, C. R., Cheng, J. D., Visscher, D. W., of no special type. Journal of Pathology, 222, 282–298.
Invasive Oncocytic Carcinoma 241
National Comprehensive Cancer Network, Clinical Oncocyte is a Greek term, first applied to sal-
Practice Guidelines in Oncology. NCCN guidelines, ivary glands by Hamperl (1931) in 1931, which
Breast Cancer v2.2.2017. Available at http://www.
nccn.org/professionals/physician_gls/pdf/breast.pdf. means “swollen cell.” Under light microscope,
Accessed 15 Aug 2017. oncocytes are characterized by abundant granular
Park, S., Koo, J., Kim, J. H., Yang, W. I., Park, B. W., & eosinophilic cytoplasm due to a high content of
Lee, K. S. (2010). Clinicopathological characteristics mitochondria, demonstrable at ultrastructural
of mucinous carcinoma of the breast in Korea:
Comparison with invasive ductal carcinoma-not other- level (Roth et al. 1962). By definition, mitochon-
wise specified. Journal of Korean Medical Science, 25, dria must occupy at least 60% of the total cyto-
361–368. plasmic volume (Ghadially 1985) and confer an
Rakha, E. A., Boyce, R. W. G., Abd El-Rehim, D., intense cytoplasmic granular positivity for anti-
Kurien, T., Green, A. R., Paish, E. C.,
Robertson, J. F. R., & Ellis, I. O. (2005). Expression of mitochondrion antibody (Damiani et al. 1998;
mucins (MUC1, MUC2, MUC3, MUC4, MUC5AC and Ragazzi et al. 2011). Namely, a carcinoma can
MUC6) and their prognostic significance in human be classified as oncocytic if it shows a strong
breast cancer. Modern Pathology, 18, 1295–1304. cytoplasmic positivity for anti-mitochondrion
Tseng, H. S., Lin, C., Chan, S. E., Chien, S. Y., Kuo, S. J.,
Chen, S. T., Chang, T. W., & Chen, D. R. (2013). antibody in at least 70% of tumor cells.
Pure mucinous carcinoma of the breast. World Journal
of Surgical Oncology, 11, 139.
Weigelt, B., Geyer, F. C., Horlings, H. M., Kreike, B., I
Clinical Features
Halfwerk, H., & Reis-Filho, J. S. (2009). Mucinous
and neuroendocrine breast carcinomas are transcrip-
tionally distinct from invasive ductal carcinomas of • Incidence
no special type. Modern Pathology, 22, 1401–1414. This tumor is considered very rare, but is
Weigelt, B., Geyer, F. C., & Reis-Filho, J. S. (2010). suspected to be overlooked, and probably mis-
Histological types of breast cancer: How special are
they? Molecular Oncology, 4, 192–208. diagnosed as apocrine carcinoma. The largest
series on record retrospectively evaluating
76 consecutive breast carcinomas using immu-
nohistochemistry for anti-mitochondrion
antibody reported an incidence of 19.6%
Invasive Oncocytic Carcinoma (Ragazzi et al. 2011).
• Age
Dario de Biase1 and Moira Ragazzi2 Patient age ranges from 26 to 94 years with an
1
Department of Pharmacy and Biotechnology, average age at presentation of 67 years (Costa
University of Bologna, Bologna, Italy and Silverberg 1989; Damiani et al. 1998;
2
Department of Oncology and Advanced Ragazzi et al. 2011; Marla et al. 2013; Itagaki
Technologies, Operative Unit of Pathology, et al. 2017).
Azienda USL-IRCCS, Reggio Emilia, Italy • Sex
Thirty-seven cases of invasive oncocytic carci-
noma have been reported in English-language
Synonyms literature. Among them, four cases occurred
in men.
Malignant oncocytoma • Site
All breast quadrants can be affected and a
multifocal and bilateral case has also been
Definition reported (Itagaki et al. 2017).
• Treatment
Invasive oncocytic carcinoma (OC) is a “special Clinical features of oncocytic carcinomas are
type” of breast carcinoma that is composed of at similar to those of invasive carcinomas, no
least 70% of cells with oncocytic features special type (NST) (▶ Invasive Carcinoma
(so-called oncocytes) on both morphologic and NST) (Ragazzi et al. 2011). Thus, the thera-
immunophenotypic grounds (Lakhani et al. 2012). peutic strategies are the same. Resistance to
242 Invasive Oncocytic Carcinoma
radiotherapy has been hypothesized just as it pushing (Fig. 1). The main difference between OC
has been reported in oncocytic tumors at other and carcinoma NST resides in cytologic features.
sites, including the rectum, thyroid, and menin- OCs are typically composed of large polygonal
ges, but it has been never investigated in the cells with abundant granular eosinophilic cyto-
breast. plasm, neat cytoplasmic borders, centrally located
• Outcome nuclei, and prominent nucleoli (Fig. 2).
Presently, knowledge available on the prog- A moderate to marked nuclear pleomorphism is
nostic impact of oncocytic differentiation in often observed. Coherently, OCs are mostly grade
breast carcinomas is scant. According to the 3 (53%) according to the Nottingham scoring
largest series reported (Ragazzi et al. 2011), system, but they can be also grade 2 (36%),
prognosis depends on grading and staging while a minority of cases is grade 1 (11%).
and is similar to the matched breast carcinomas A discrete lymphoid infiltrate can be also seen in
NST (▶ Invasive Carcinoma NST). the majority of cases. Invasive carcinoma is asso-
ciated with an in situ component in a prevalence
of cases.
Macroscopy
In a retrospective series, tumors with oncocytic
features were originally diagnosed as invasive car-
Tumor size ranges from 0.8 to 9 cm (mean 3 cm).
cinoma NST in the majority of patients (78%) but
On cut section, OCs are mainly well-defined, solid,
special-types were also present including pure
and firm, from whitish to tan brown nodules.
mucinous carcinomas (▶ Invasive Mucinous Car-
cinoma), apocrine (▶ Apocrine Carcinoma), and
neuroendocrine carcinomas (▶ Invasive Carci-
Microscopy
noma with Neuroendocrine Differentiation)
(Ragazzi et al. 2011). One tall cell variant of pap-
OC shows the spectrum of different architectural
illary breast carcinoma (▶ Tall Cell Variant of Pap-
patterns seen in invasive carcinoma NST (▶ Inva-
illary Breast Carcinoma) was also reported in that
sive Carcinoma NST), even if a solid growth
series (Fig. 3). In these mixed cases, aspects of both
pattern with nests, sheets, and cords is
differentiations usually coexist.
characteristic. Tumor margins are more frequently
Invasive Oncocytic
Carcinoma,
Fig. 1 Invasive oncocytic
carcinoma (OC), low
magnification: Nodule has
typically pushing margins
and solid architecture
(H&E)
Invasive Oncocytic Carcinoma 243
Invasive Oncocytic Carcinoma, Fig. 2 Invasive Antimitochondrion antibody shows intense and diffuse
oncocytic carcinoma (OC), higher magnification: (a) Neo- positivity; (c) A discrete lymphocytic infiltrate is often
plastic cells have abundant eosinophilic and finely granular present at the edge of the tumour (H&E); (d) Tumour
cytoplasm, with neat borders, centrally located round cells forming cords in desmoplastic stroma can be seen
nuclei, and prominent nucleoli (H&E); (b) (H&E)
Invasive Oncocytic Carcinoma, Fig. 3 Tall cell variant composed of columnar cells with abundant granular cyto-
of papillary breast carcinoma: (a) Follicular structures plasm and grooved nuclei (H&E); (c) Tumour cells show
filled with colloid-like material with scalloped borders. intense and diffuse positivity for antimitochondrion anti-
Small papillae are evident as well as some calcifications body (Courtesy of Professor Vincenzo Eusebi)
(H&E). (b) High magnification of a solid structure
the 5p13.33 region, the Telomerase Reverse Tran- notably increase of SOD2 immunostaining in a
scriptase (TERT) gene has been mapped and it has series of OC breast carcinomas compared with
been established that TERT protein can localize to non-OC. These high SOD2 levels seem to be cor-
the mitochondrion and modulate its activity related with the high activity of the SIRT/FOXO/
(Sahin et al. 2011). Based on these data, Geyer SOD2 axis. Moreover, the authors observed that
et al. concluded that OCs of the breast are a group patients with high SOD2 expression tumors had a
of tumors with a distinctive pattern of chromo- significantly worse outcome and that a SOD2 pos-
somal aberrations, previously associated with itive expression correlated with lymph nodal
OCs of other anatomical sites (e.g., oncocytic involvement (Kenny et al. 2017).
tumors of the kidney and thyroid) (Geyer
et al. 2012).
A recent in vitro study, performed on six breast Differential Diagnosis
cancer cell lines, investigated the role of SIRT/
FOXO/SOD2 axis of the mitochondrial unfolded Carcinoma with apocrine differentiation (▶ Apo-
protein response in promoting breast tumor inva- crine Carcinoma): The differential diagnosis
siveness. In the same paper, Kenny et al. found a between OC and carcinomas with apocrine
Invasive Oncocytic Carcinoma 245
features is usually difficult on morphology and Eusebi, V., Damiani, S., Ellis, I. O., Azzopardi, J. G., &
mainly relies on immunohistochemistry. Apo- Rosai, J. (2003). Breast tumor resembling the tall cell
variant of papillary thyroid carcinoma: Report of
crine carcinomas are positive for GCDFP-15 and 5 cases. American Journal of Surgical Pathology, 27,
AR, while hormone receptors are typically nega- 1114–1118.
tive and antibodies for mitochondria are seen in Eusebi, V., Tallini, G., & Rosai, J. (2004). Nuclear alter-
cells below 50% of the total neoplastic prolifera- ations and RET/PTC activation. American Journal of
Surgical Pathology, 28, 974–975.
tion. Occasional cases, however, are oncocytic Foschini, M. P., Asioli, S., Foreid, S., Cserni, G., Ellis,
and apocrine at the same time. I. O., Eusebi, V., & Rosai, J. (2017). Solid papillary
Acinic cell carcinoma (ACC) (▶ Acinic Cell breast carcinomas resembling the tall cell variant of
Carcinoma): Granular cytoplasm in ACC is due to papillary thyroid neoplasms: A unique invasive tumor
with indolent behavior. American Journal of Surgical
zymogen granules that are evidenced by PAS after Pathology, 41, 887–895.
diastase digestion. Anti-mitochondrion antibodies Geyer, F. C., de Biase, D., Lambros, M. B., Ragazzi, M.,
are negative. Tumor cells in ACC show serous Lopez-Garcia, M. A., Natrajan, R., Mackay, A.,
differentiation and are positive with salivary type Kurelac, I., Gasparre, G., Ashworth, A., Eusebi, V.,
Reis-Filho, J. S., & Tallini, G. (2012). Genomic profil-
amylase, lysozyme, and alpha-1-antichimotrypsin ing of mitochondrion-rich breast carcinoma: Chromo-
immunostains. In addition, ACC cells are typi- somal changes may be relevant for mitochondria
cally immunoreactive for S-100 and they are triple accumulation and tumour biology. Breast Cancer
Research and Treatment, 132, 15–28. I
negative with no expression of both estrogen and
Ghadially, F. N. (1985). Diagnostic electron microscopy of
progesterone receptors and no Her2 amplification. tumours (2nd ed.). London: Butterworth & Company.
Neuroendocrine carcinoma (▶ Invasive Carci- Hamperl, H. (1931). Beiträge zur normalen und
noma with Neuroendocrine Differentiation): pathologischen Histologie menschlieher
Well-differentiated neuroendocrine carcinoma of Speicheldriisen. Zeitschrift für Mikroskopisch-
Anatomische Forschung, 27, 1–55.
the breast could be very similar to low-grade Itagaki, H., Yamamoto, T., Hiroi, A., Kawanishi, K., Nogu-
OC. Expression of chromogranin and/or syn- chi, E., Ohchi, T., Kamio, T., Kameoka, S., Oda, H., &
aptophysin in the majority of tumor cells is char- Nagashima, Y. (2017). Synchronous and bilateral
acteristic of neuroendocrine differentiation. oncocytic carcinoma of the breast: A case report and
review of the literature. Oncology Letters, 13,
Granular cell tumors (▶ Granular Cell Tumor): 1714–1718.
They are composed of cells with eosinophilic Kenny, T. C., Hart, P., Ragazzi, M., Sersinghe, M., Chipuk,
granular cytoplasm that can mimic oncocytes, J., Sagar, M. A. K., Eliceiri, K. W., LaFramboise, T.,
but they derive from Schwann cells of peripheral Grandhi, S., Santos, J., Riar, A. K., Papa, L., D'Aurello,
M., Manfredi, G., Bonini, M. G., & Germain,
nerves. Accordingly, neoplastic cells in granular D. (2017). Selected mitochondrial DNA landscapes
cell tumors are strongly and diffusely positive for activate the SIRT3 axis of the UPRmt to promote
S100 and CD68 immunostains while they are metastasis. Oncogene, 36, 4393–4404.
negative for keratins. Lakhani, S. R., Ellis, I. O., Schnitt, S. J., Tan, P. H., & van
de Vijver, M. J. (2012). WHO classification of tumours
of the breast (4th ed.). Lyon: IARC.
Marla, N. J., Pai, M. R., Swethadri, G. K., & Fernandes,
References and Further Reading H. (2013). Male breast cancer-review of literature on a
rare microscopic variant (oncocytic carcinoma). Indian
Bhargava, R., Florea, A. V., Pelmus, M., Jones, M. W., Journal of Surgery, 75(Suppl 1), 240–242.
Bonaventura, M., Wald, A., & Nikiforova, M. (2017). Masood, S., Davis, C., & Kubik, M. J. (2012). Changing
Breast tumor resembling tall cell variant of papillary the term “breast tumor resembling the tall cell variant of
thyroid carcinoma: A solid papillary neoplasm with papillary thyroid carcinoma” to “tall cell variant of
characteristic immunohistochemical profile and few papillary breast carcinoma”. Advances in Anatomic
recurrent mutations. American Journal of Clinical Pathology, 19, 108–110.
Pathology, 147, 399–410. Ragazzi, M., de Biase, D., Betts, C. M., Farnedi, A.,
Costa, M. J., & Silverberg, S. G. (1989). Oncocytic carci- Ramadan, S. S., Tallini, G., Reis-Filho, J. S., & Eusebi,
noma of the male breast. Archives of Pathology and V. (2011). Oncocytic carcinoma of the breast: Fre-
Laboratory Medicine, 113, 1396–1399. quency, morphology and follow-up. Human Pathology,
Damiani, S., Eusebi, V., Losi, L., D’Adda, T., & Rosai, 42, 166–175.
J. (1998). Oncocytic carcinoma (malignant Roth, S. I., Olen, E., & Hansen, L. S. (1962). The eosino-
oncocytoma) of the breast. American Journal of Surgi- philic cells of the parathyroid (oxyphil cells), salivary
cal Pathology, 22, 221–230. (oncocytes), and thyroid (Huerthle cells) glands. Light
246 Invasive Secretory Carcinoma
and electron microscopic observations. Laboratory very rarely in the elderly. The age range of
Investigation, 11, 933–941. the reported cases varies from 3 to 91 years.
Sahin, E., Colla, S., Liesa, M., Moslehi, J., Muller, F. L.,
Guo, M., Cooper, M., Kotton, D., Fabian, A. J., • Sex
Walkey, C., Maser, R. S., Tonon, G., Foerster, F., Mainly observed in adult female and children of
Xiong, R., Wang, Y. A., Shukla, S. A., Jaskelioff, M., both sexes, very rare in men over the age of 30.
Martin, E. S., Heffernan, T. P., Protopopov, A., A case in a male-to-female transgender patient
Ivanova, E., Mahoney, J. E., Kost-Alimova, M.,
Perry, S. R., Bronson, R., Liao, R., Mulligan, R., has been described (Grabellus et al. 2005).
Shirihai, O. S., Chin, L., & DePinho, R. A. (2011). • Site
Telomere dysfunction induces metabolic and mito- ISC may appear in any part of the breast; how-
chondrial compromise. Nature, 470, 359–365. ever, a subareolar location is prevalent, particu-
larly in men and children, and frequently
associated with nipple discharge. The usual clin-
ical presentation is a slow growing nodule,
Invasive Secretory Carcinoma mobile, often present for a long time and easily
mistaken with a fibroadenoma (▶ Fibroadenoma)
Angelo Sidoni or a papilloma (▶ Intraductal Papilloma) on
Department of Experimental Medicine, Section of imaging studies. Rare cases are reported in the
Pathologic Anatomy and Histology, Medical axilla, conceivably originating from ectopic
School, University of Perugia, Perugia, Italy breast tissue or from skin appendage glands.
• Treatment
Treatment varies depending on the age and sex
Synonyms of patients. In children, considering the favor-
able prognosis and the need to safeguard the
Juvenile secretory carcinoma, Juvenile breast car- development of the breast, a conservative local
cinoma, Secretory carcinoma. excision is the most advisable treatment. In post-
menarchal girls and women wide local excision,
quadrantectomy, or mastectomy are reported
Definition based on tumor dimensions in order to obtain
negative surgical margins. In males, due to small
Invasive secretory carcinoma (ISC) is one of the dimension of breast, mastectomy is the rule.
rarest histological subtypes of breast cancer, of Sentinel lymph node (▶ Sentinel Node) map-
low-grade and translocation-associated. ISC ping is suggested. No definitive conclusions are
shows distinctive histological features consisting available on the benefits of systemic adjuvant
in intracellular and extracellular production of chemotherapy and radiotherapy.
secretory material. • Outcome
The clinical course of ISC is mainly character-
ized by favorable prognosis and a prolonged
Clinical Features survival, particularly in children and young
adults under 30 years of age, even in presence
• Incidence of lymph node metastasis (rarely are involved
Less than 0.15% of all breast carcinomas more than three lymph nodes). In adult women
(about 200 cases published in English ISC are more aggressive, especially if the
literature). tumor is of high-grade (see below) and locally
• Age advanced, although distant metastases are
Originally described in children and termed exceptional and still correlated with long sur-
“juvenile secretory carcinoma” (McDivitt and vival. Late local recurrence (even after
Stewart 1966), it appears that two thirds of the 20 years) may occur and a prolonged follow-
published cases actually occur in adults and up is necessary.
Invasive Secretory Carcinoma 247
Invasive Secretory Carcinoma, Fig. 1 Coexistence of Invasive Secretory Carcinoma, Fig. 3 Despite the well
solid, microcystic, follicular, tubular, and papillary patterns circumscribed appearance, invasion is present at the border
(H&E) (H&E)
248 Invasive Secretory Carcinoma
Lobular In Situ Neoplasia, Fig. 1 (H&E) Proliferation nuclei are uniform with evenly distributed chromatin and
of monomorphic, evenly spaced cells that are loosely indistinct nucleoli (A). Intracytoplasmic lumina are often
cohesive and slightly larger than normal acinar cells. The present (B – arrows)
Lobular In Situ Neoplasia, Fig. 2 LCIS distribution can spread along larger ducts or even along the main lactiferous
involve lobules causing expansion (A, H&E) and E- ducts, causing cloverleaf or necklace patterns (C, H&E). E-
cadherin immunostaining persists in myoepithelial cells cadherin negative LCIS cells spread along ducts between
around E-cadherin–negative LCIS cells (B). LCIS can E-cadherin positive epithelial and myopeithelial cells (D)
In Situ Carcinoma with Mixed Ductal and characteristically diffusely localized to the cyto-
Lobular Features plasm in cases of lobular neoplasia, whereas in
It is very rare. This is considered collision in situ ductal lesions it remains localized to the mem-
carcinoma containing both LCIS-C and ductal car- brane. Immunoreactivity for ER and progesterone
cinoma in situ (DCIS) within the same structure. receptor (PR) is found in virtually all LCIS-C, in
about 66% of LCIS-P and in less than 25% of
apocrine LCIS-P. PR expression is reduced in
Immunophenotype LCIS associated with ILC (▶ Invasive Lobular
Carcinoma). LCIS-C is typically negative for
LCIS-C and variants of LCIS can exhibit strong HER2 protein overexpression/gene amplification,
reactivity for the high-molecular-weight CK lacks p53 mutations, and has a low Ki67 labeling
34bE12 that is absent or weakly expressed in index. Conversely, variants of LCIS and LCIS-P
DCIS. E-cadherin expression can be reliably may show HER2 protein overexpression/gene
used as a marker to differentiate between ductal amplification (especially when associated with
and lobular neoplasia (>95% of cases of LCIS invasive carcinoma), p53 positivity and a moder-
lack E-cadherin). However, morphologically ate to high Ki67 labeling index. GCDFP-15 is
unequivocal cases of LCIS may demonstrate positive in >90% of LCIS-C, 75% of LCIS-P,
focal E-cadherin positivity. p120-catenin is and in virtually all cases of apocrine LCIS-P.
Lobular In Situ Neoplasia 255
Lobular In Situ Neoplasia, Fig. 3 Apocrine type of LCIS (A, H&E); E-Cadherin is negative in neoplastic cells (B);
GCDFP-15 (C) and PAS (D) are diffusely positive in apocrine LCIS
Lobular In Situ Neoplasia, Fig. 4 Pleomorphic LCIS, The black arrows on pictures A-C point at the characteris-
central comedo necrosis (A, H&E); diffuse p120 cytoplas- tics multinucleation which is a distinctive feature of
mic positivity (B); PAS-AB staining (C); E-Cadherin (D). LCIS-P
diagnosis of lobular neoplasia. On the other hand, central necrosis and calcifications. The
the presence of distinct cell membranes supports a dyscohesive appearance, lack of E-cadherin
diagnosis of DCIS (▶ Ductal Carcinoma In Situ). expression and diffuse p120 staining of the cells
E-cadherin, p120 IHC can be particularly helpful. are helpful in making the LCIS-P diagnosis.
King, T. A., Pilewskie, M., Muhsen, S., Patil, S., carcinomas. Such a variability of incidence is
Mautner, S. K., Park, A., Oskar, S., Guerini-Rocco, E., due to a plethora of terminologies used by
Boafo, C., Gooch, J. C., De Brot, M., Reis-Filho, J. S.,
Morrogh, M., Andrade, V. P., Sakr, R. A., & different authors to designate these tumors.
Morrow, M. (2015). Lobular carcinoma in situ: Consequently, it is not surprising that there
A 29-year longitudinal experience evaluating clinico- are series of metaplastic carcinomas that do
pathologic features and breast cancer risk. Journal of not even report a single case of LGASC,
Clinical Oncology, 33, 3945–3952.
Mastracci, T. L., Tjan, S., Bane, A. L., O’malley, F. P., & while other authors estimate their incidence
Andrulis, I. L. (2005). E-cadherin alterations in atypical around 0.58% of the total of invasive breast
lobular hyperplasia and lobular carcinoma in situ of the cancers. The real prevalence of this histotype is
breast. Modern Pathology, 18, 741–751. probably underestimated due to the diagnostic
Pieri, A., Harvey, J., & Bundred, N. (2014). Pleomorphic
lobular carcinoma in situ of the breast: Can the difficulties in recognizing it correctly. In any
evidence guide practice? World Journal of Clinical case this entity has to be considered exception-
Oncology, 5, 546–553. ally uncommon and no more than 130 cases
Simpson, P. T., Reis-Filho, J. S., Gale, T., & Lakhani, S. R. have been published in English literature
(2005). Molecular evolution of breast cancer. Journal
of Pathology, 205, 248–254. until now.
• Age
In the original series of Rosen and Ernsberger
(1987) the age of patients affected by LGASC
Low-Grade Adenosquamous ranged from 42 to 76 years with a mean age of
Carcinoma 59. In later studies the extent of age range was
19–88 years, with prevalence in the peri-
Angelo Sidoni meopausal or postmenopausal age.
Department of Experimental • Sex L
Medicine, Section of Pathologic Anatomy Female. No cases of LGASC in males have
and Histology, Medical School, been reported so far.
University of Perugia, Perugia, Italy • Site
LGASC may appear in any part of the breast
parenchyma not infrequently in peria-
Synonyms reolar location. The most frequent clin-
ical presentation is a palpable mass followed
Adenosquamous carcinoma; Infiltrative syringomatous by mammographic abnormalities or nipple dis-
adenoma; Syringomatous squamous tumor charge. Due to the frequent association with
other lesions imaging studies can be inconclu-
Definition sive or confounding.
• Treatment
Low-grade adenosquamous carcinoma (LGASC) Because of the paucity of reported cases, there
of the breast is exceedingly uncommon, and it is is no consensus on the optimal treatment of
characterized by well-developed gland formation LGASC. On the other hand, when compared
mixed with solid nests of squamous cells in a with the majority of metaplastic carcinomas,
spindle cell background. which are also triple negative but highly
aggressive and chemoresistant, LGASC por-
tends an indolent course with a low metastatic
Clinical Features potential and thus a complete local excision or
mastectomy to achieve clear margins appear
• Incidence appropriate.
LGASC belongs to the family of “metaplastic Lymph node metastases are exceedingly
breast cancers,” (▶ Invasive Metaplastic Car- rare and routine sentinel node biopsy and axil-
cinoma) representing 0.2 to 5% of all breast lary dissection may not be necessary. There is
258 Low-Grade Adenosquamous Carcinoma
no unequivocal evidence of the usefulness of H&E). Epithelial islands are irregularly distrib-
both adjuvant radiotherapy and chemotherapy. uted into an abundant collagenous and lamellar
• Outcome stroma rich in plump spindle cells, which subtly
Unlike the aggressive tumor behavior and poor merge with epithelial cells (Fig. 3 H&E). Some-
clinical outcomes seen in other forms of meta- times tubular structures appear to radiate from a
plastic mammary carcinomas, the majority of fibrosclerotic center similarly to a radial scar
LGASC have been described to have an excel- (▶ Radial Scar). Within glands, varying degrees
lent prognosis with greater predilection of squamous differentiation are observed
towards local recurrence, even multiple, (syringoid appearance), from focal not
which can occur within 2–3 years especially keratinizing lining of glandular lumens to sizable
in patients with excisional biopsy alone. Usu- squamous pearls (Fig. 4 H&E). An attenuated
ally all patients with recurrences remain dis- outer myoepithelial layer around glands and
ease free after definitive re-excision. The only cords can be seen (see below). Cytological atypia
particularly aggressive examples reported in
the literature are: one lethal case due to a recur-
rent tumor infiltrating the chest wall; one
patient with pulmonary metastasis and one
with a single lymph node metastasis.
Macroscopy
Microscopy
Low-Grade Adenosquamous Carcinoma, Fig. 3 Low- Low-Grade Adenosquamous Carcinoma, Fig. 5 Low-
grade adenosquamous carcinoma. Fibrous stroma grade adenosquamous carcinoma. Peripheral staining for
containing numerous spindle cell elements disposed in a the myoepithelial marker p63 in the glandular elements and
lamellar pattern blending with epithelial component in the compressed epithelial cords (H&E)
(H&E)
scenarios, LGASC may be an incidental finding
on histological examination of excisional biopsy
or mastectomy specimens.
Immunophenotype L
needle aspirates, core biopsy specimens, or frozen Badve, S., & Reis-Filho, J. S. (2010). Genomic and
sections. immunohistochemical analysis of adenosquamous car-
cinoma of the breast. Modern Pathology, 23, 951–960.
Kawaguchi, K., & Shin, S. J. (2012). Immuno-
histochemical staining characteristics of low-grade
References and Further Reading adenosquamous carcinoma of the breast. American
Journal of Surgical Pathology, 36, 1009–1020.
Boecker, W., Stenman, G., Loening, T., Andersson, M. K., Rosen, P. P., & Ernsberger, D. (1987). Low-grade
Sinn, H. P., Barth, P., Oberhellmann, F., adenosquamous carcinoma: A variant of metaplastic
Bos, I., Berg, T., Marusic, Z., Samoilova, V., & mammary carcinoma. American Journal of Surgical
Buchwalow, I. (2014). Differentiation and histogenesis Pathology, 11, 351–358.
of syringomatous tumour of the nipple and low-grade Senger, J. L., Meiers, P., & Kanthan, R. (2015). Bilateral
adenosquamous carcinoma: Evidence for a common synchronous low-grade adenosquamous carcinoma of
origin. Histopathology, 65, 9–23. the breast: A case report with review of current litera-
Geyer, F. C., Lambros, M. B. K., Natrajan, R., Mehta, R., ture. International Journal of Surgery Case Reports,
Mackay, A., Savage, K., Parry, S., Ashworth, A., 14, 53–57.
L
M
the same way as women. Usually this is mas- Survival rates for MBC are generally assumed
tectomy followed by chemotherapy and radio- to be lower than female breast cancer, probably
therapy. Some men opt for breast-conserving as a result of presentation at a more advanced
surgery, if appropriate, because of reduced stage than in women. However, in general, stud-
morbidity and a better cosmetic outcome. As ies in which male and female breast cancer are
most MBCs express estrogen receptor (ER), matched for key prognostic factors (size, grade,
they are suitable for treatment with endocrine and lymph node status) have refuted this, with
therapy, predominantly adjuvant tamoxifen. outcome almost identical between genders.
Data from clinical trials such as ATAC, in
which the efficacies of aromatase inhibitors
Macroscopy
(AIs) were compared to tamoxifen, have revo-
lutionized the treatment of ER positive breast
Most specimens are mastectomies (Fig. 1a) with
cancer in postmenopausal women, such that
either sentinel node biopsy or axillary node clear-
AIs are now considered standard therapy.
ance according to the presurgical nodal assess-
Despite the fact that aromatase is expressed
ment results. The macroscopic tumors are often
in MBC, initial case series have shown nega-
large, with frequent skin and nipple involvement
tive or equivocal results meaning the efficacy
(Fig. 1b). Similar to the female type, the tumor
of aromatase inhibition in men has been
may appear circumscribed and infiltrative or show
questioned. Because of the increase in testos-
cystic changes.
terone seen after aromatase inhibition, this may
overcome the effect of AI blockade, by
saturating the enzyme pathway with substrate,
resulting in only a modest suppression of estro- Microscopy
gen. Hence LHRH agonists are required to
reduce excessive substrate and maximize the The main histological types and their frequency
effect of aromatase inhibition. As a result, AIs in MBC are shown in Table 1. Of these, the
are generally contraindicated in MBC, unless commonest type of MBC is invasive carcinoma
used with medical or surgical orchiectomy. of no special type (NST) (▶ Invasive Carcinoma
• Outcome NST) of grade 2 differentiation (Fig. 2). The
About 80 men will die of breast cancer in the tumors comprise nests and trabeculae of malig-
UK each year. In the USA, this is around 460. nant cells within fibrous stroma. Invasive lobular
Male Breast Cancer, Fig. 1 (a) A mastectomy specimen surgical margin is painted. (b) Slicing of the posterior
with orientation sutures. It includes fibrofatty breast tissue surface shows a well-defined, grayish-white hemorrhagic
with an overlying nipple bearing ellipse of the skin. The tumor. The tumor is clear of surgical margins
Male Breast Cancer 265
Male Breast Cancer, Table 1 Main histological sub- reported from the EORTC10085, TBCRC, BIG,
types observed in MBC and their relative frequencies and NABCG International Male Breast Cancer
Histology % Frequency Program and other smaller studies from single
NST 85 centers. This is illustrated in Table 2. As with
Lobular 1–2 female breast cancer, ERa and PR are used to
Papillary 3–5 guide treatment and prognosis in MBC (▶ Hor-
Mucinous 1 mone Receptors in Breast Cancer). Other bio-
Medullary 0.5 markers that have been examined in MBC
Cribriform 0.5–1
using immunohistochemistry include AR,
Tubular 0.5
ERb1, ERb2, ERb5, Bcl-2, p53, E-cadherin,
Mixed 5
Ki67, survivin (both in the nucleus and cyto-
Data was obtained from several published articles, includ-
plasm), prolactin, and FOXA1. While these
ing the Veterans Affairs (VA) Central Cancer Registry and
Shaaban et al. (2012) biomarkers are not unique to MBC, some of
them, e.g., AR, is frequently expressed at
relativity high levels in men. Although AR is
currently not used for routine diagnostics
or management in MBC, its relative abundance
suggests that this could be used as a therapeutic
target for antiandrogen therapies. Examples
of ERa, PR, and AR expression in MBC are
shown in Fig. 3.
Although ERa expression is higher in
MBC than in female breast cancer (typically in
approximately 80% of MBCs vs. 60–70% of
female breast cancers), it has been proposed that
M
not all ERa-positive MBCs behave in the same
way as their female counterparts. This is founded
Male Breast Cancer, Fig. 2 (H&E) image of the most through observations of different grouping of hor-
frequent type of MBC, invasive carcinoma NST
mone receptor patterns when applying hierarchical
clustering. In female breast cancers, PR and ERa
carcinoma is extremely rare and so is lobular clustered together, while in MBC, ERa clustered
carcinoma in situ. Compared with female breast together with ERb and AR, with PR clustering
cancer, incidence of papillary carcinoma (both independently (Shaaban et al. 2012). Further clus-
intraductal and invasive) and mucinous ter analysis in MBC showed significant correlation
carcinoma is higher in the male breast (Shaaban of ERa, AR, and FOXA1 (Humphries et al. 2017).
et al. 2012). Ductal carcinoma in situ (DCIS; A number of cell cycle proteins have also been
▶ Ductal Carcinoma In Situ) is reported in examined in MBC. Enhanced proliferation seems
5–15% of cases. Reporting of MBC and cancer to be associated with poorer outcome in MBC,
datasets is the same as for female breast cancer. exemplified analysis of proliferation by mitotic
count, and the expression of cyclins -A, -B, and
D1 biomarkers.
Immunophenotype
Male Breast Cancer, Table 2 Overview of the clinico- group reported differential driver genes in MBC
pathological features of MBC in 16 published studies versus female breast cancer. Somatic genetic
examining a total of 1986 cases from 1996 to 2017
alterations typically seen in ERa-positive/HER2-
Feature Number (%) negative female breast cancers (the most common
Histology phenotype seen in MBC), e.g., PIK3CA and TP53
NST 1615 (84) mutations and 16q loss, were much less frequent in
Lobular 19 (1)
subtype-matched MBC, suggesting that MBC is
Other 239 (12)
driven by different somatic changes (Piscuoglio
N/A 60 (3)
et al. 2016). In a number of smaller studies,
Grade
gender-specific differentially expressed genes,
1 239 (13)
2 872 (48)
including those involved in translation, cell migra-
3 597 (33) tion/motility, immune response, membrane trans-
N/A 115 (6) port, apoptosis, and energy metabolism, have been
Node reported, adding weight to the hypothesis that male
+ 734 (42) and female breast cancers are biologically distinct.
742 (42) Promoter hypermethylation has been reported
N/A 275 (16) in MBC with methylation of a number of
ERa genes that are known to act as tumor suppressors.
+ 1584 (86) Accumulation of methylated genes and an overall
193 (10) high methylation pattern were correlated with a
N/A 74 (4) more aggressive phenotype and poorer survival.
PR Interestingly, RASSF1A, a well-characterized
+ 1321 (72) tumor suppressor gene, was significantly more
436 (24) frequently methylated in MBC than female breast
N/A 84 (5)
cancer, providing further evidence of likely bio-
HER2
logical differences between genders.
+ 160 (9)
Expression of microRNAs (miRs), small non-
1319 (77)
coding RNAs that alter gene expression at the
N/A 241 (14)
posttranscriptional level, has been examined in
Data adapted from Humphries et al. 2017, where details on
specific studies can be found MBC. Differential expression of several miRNAs
N/A not available has been reported between gynecomastia and
MBC and also between MBC and female breast
cancer. Despite these observational reports, the
starting to suggest there may be underlying role of miRs in MBC, and if this may contrast
gender-specific biological differences in their from female breast cancer, has not been studied
genomic landscapes, both genetically and fully.
epigenetically. Germline mutations of BRCA2 and, to a
As reported above, in general, the main histo- lesser extent, BRCA1 are associated with
logical subtype observed in MBC is invasive car- increased risk of men developing breast cancer
cinoma NST with the most common phenotype (approximately 5–10% and 1–5%, respectively).
ER- and/or PR-positive; HER2-negative, which A genome-wide association study of MBC
falls into the luminal A-like subgroup. However, identified a SNP in RAD51B which was associ-
one of the first gene profiling studies to compare ated with MBC susceptibility, but there is
male and female breast cancer showed two unique evidence that common variants associated
subgroups of MBC, termed luminal M1 and lumi- with female breast cancer may also impact
nal M2 (Johansson et al. 2012). These differed from MBC risk (Orr et al. 2012). More recent genotyping
the conventional molecular classifications observed of 1802 male carriers of BRCA1/2 mutations
in female breast cancer. Consequently, the same showed that weighted polygenic risk scores based
Male Breast Cancer 267
Male Breast Cancer, Fig. 3 Immunohistochemical examples of hormone receptors which are commonly expressed in
MBC. ERa (a), PR (b), and AR (c) are frequently expressed
M
on 88 female breast cancer susceptibility variants to the architectural features of hyperplasia, admix-
were similarly associated with breast cancer risk in ture of luminal and basaloid cells, and the absence
men (Lecarpentier et al. 2017). A large ongoing of significant atypia support a benign diagnosis.
study into the causes of MBC has identified several Basal cytokeratins such as CK5 and CK14 show a
genetic variations associated with risk of develop- three-layered ductal epithelium in gynecomastia
ing the disease. These genetic variations appear to (Kornegoor et al. 2012) and negative staining in
have a different effect on risk between genders, DCIS. Metastases to the breast should be identi-
lending further support to the idea that the biology fied by clinical correlation, unusual histological
of breast cancer is diverse in men and women. features, and a suitable panel of immunohisto-
chemical biomarkers.
Differential Diagnosis
References and Further Reading
The most important differentials are gynecomas-
tia, a benign enlargement of the male breast Humphries, M. P., Sundara Rajan, S., Honarpisheh, H.,
Cserni, G., Dent, J., Fulford, L., Jordan, L. B.,
(▶ Gynecomastia) and metastases to the breast,
Jones, J. L., Kanthan, R., Litwiniuk, M., et al. (2017).
such as metastatic prostatic carcinoma, mela- Characterisation of male breast cancer: A descriptive
noma, etc. Gynecomastia may present with florid biomarker study from a large patient series. Scientific
hyperplasia of the mammary ducts which can Reports, 7, 45293.
Johansson, I., Nilsson, C., Berglund, P., Lauss, M.,
mimic in situ carcinoma particularly on small
Ringner, M., Olsson, H., Luts, L., Sim, E.,
biopsy or in cytological preparations. Attention Thorstensson, S., Fjallskog, M. L., et al. (2012).
268 Malignant Adenomyoepithelioma (M-AME)
Cancer Agency, the mean age is 70.6 years, 26 months, and two are alive and well after
and the range is 42–96 years with only two local recurrences. Eleven patients are alive
2 patients aged <50 years. and one is dead with no evidence of disease
• Sex ranging from 6 to 130 months with a mean
The reported cases in the literature have been follow-up of 50 months.
females, and no males are recorded in the • Clinical Presentation
30 cases in our database. However, since Most cases present as a palpable mass. Some
there are rare case reports of benign AME in patients with M-AME report a long-standing
males, M-AME is most likely not confined to stable mass followed by a period of accelerated
females. growth. A few cases present on screening
• Site mammography as mass lesions.
No specific site in the breast
• Treatment
Macroscopy
Surgical excision of the primary tumor mass is
the mainstay of treatment. Since axillary lymph
M-AME with ductal carcinoma in situ
node involvement is rare, axillary node dissec-
(DCIS) typically presents as a multilobu-
tion is contraindicated in the absence of
lated circumscribed mass similar to that of
involved nodes on clinical or imaging studies,
benign AME.
but sentinel node biopsy is appropriate.
The mass is often reported to be firm, rubbery,
Regional radiation may be helpful and is used
and yellow-white in color. A few cases show
empirically by most oncologists. The value of
cystic change and necrosis. M-AME with an inva-
standard chemotherapy is unknown with few
sive carcinoma presents as an irregular infiltrative
positive responses reported, but most cases are
mass. The size ranges from 1.4 to 14 cm in
treated as for triple negative breast cancer of
usual or metaplastic type. In the case of NST
greatest dimension. Recognition of a pre-existing M
or associated low-grade or “benign” AME is key
carcinomas arising from AME, the manage-
to the diagnosis of carcinoma ex-AME. This
ment should be appropriate to the breast bio-
requires thorough sampling of the gross specimen
marker status of the malignant component.
to include any focal mass lesions within the con-
A recent case report describes the response of
fines of the main tumor. The precursor lesion is
M-AME to eribulin (Lee et al. 2015).
typically nodular but may be papillary in nature.
• Outcome
Many cases of EMEC are also associated with a
There is little data available on the outcome of
low-grade benign AME.
M-AME with invasive carcinoma due to the
rarity of the tumor, its occurrence in the older
age group where life span is already limited,
and poor follow-up in the small series of Microscopy
cases reported to date. There is some
evidence that M-AME with components of Malignant AME of biphasic epithelial-
squamous or spindle cell metaplastic carci- myoepithelial carcinoma (EMEC) type may
noma do better than those with components arise de novo without a precursor low-grade AME
of carcinosarcoma or metaplastic matrix- (Petrozza et al. 2013), but some cases appear to
producing carcinoma. In our records, 5 of represent high-grade dual-lineage transformation
23 patients with M-AME and follow-up data of a low-grade precursor lesion. The tumors are
died of disease at 3, 24, 31, 36, and 54 months. typically large and have a mixture of partially
Three patients died of unknown cause at circumscribed nodules and diffuse areas of growth
36, 44, and 132 months. One patient is alive with infiltrative borders. Glandular and papillary
with pulmonary and nodal metastases after structures exhibiting a disorganized architecture
270 Malignant Adenomyoepithelioma (M-AME)
Malignant Adenomyoepithelioma (M-AME), Fig. 1 (H&E) (a) EMEC showing lobulated architecture, (b, c)
disorganized ductal and myoepithelial elements, and (d) hyaline matrix material
are present, but the degree of cytological atypia necrosis. Commonly, the malignant spindle cell
varies from case to case and in different areas of component of the tumor outgrows the epithelial
the same tumor (Fig. 1). Often, in some foci, the component in many areas of the tumor. Metastatic
tumor retains a partially organized relationship EMEC may retain its dual phenotype in both nodal
between the luminal cells lining the tubules and and pulmonary metastases (Fig. 4).
papillary structures and the layers of underlying It is likely that the entity of “borderline” or
malignant myoepithelial cells (MECs) (Fig. 2). “atypical” AME represents the precursor lesion
Recognition of these poorly organized dual-lineage to this group of tumors. Borderline AME has the
foci is key to differentiating this neoplasm from the morphological appearance of benign AME but
more common breast metaplastic carcinoma of car- with superimposed cytological atypia and an
cinosarcoma or matrix-producing type (Yoon and increased proliferative rate. Its non-infiltrative
Chitale 2013). It is these areas that should be care- growth pattern and generally benign clinical
fully scrutinized using immunohistochemistry in behavior suggest that these “borderline” lesions
order to confirm the dual lineage of the cells for are better grouped with the benign AME category
diagnostic purposes. The MEC component of for practical purposes. However, one has to be
EMEC is typically plump spindle cells that encircle aware that even so-called “benign” AME has the
the epithelial structures and extend as infiltrative potential to recur locally and even rarely metasta-
sheets and strands into the surrounding native size (Nadelman et al. 2006).
breast stroma (Fig. 3). Again, the morphology of The key to the diagnosis of carcinoma ex-AME
this component of the tumor is exceedingly vari- is the recognition of the underlying precursor low-
able between different cases and even within the grade or benign AME within the background of the
same tumor. Areas with a bland spindle cell mor- carcinoma (Fig. 5). The precursor lesion can be
phology often associated with production of hya- detected grossly or on low-power examination of
line collagenous matrix are seen. Elsewhere the the tumor where it appears as a multilobulated
cells may exhibit marked cytological atypia, complex mass, sometimes with a papillary compo-
numerous mitoses, matrix production, and nent. High-power examination will reveal the
Malignant Adenomyoepithelioma (M-AME) 271
Malignant Adenomyoepithelioma (M-AME), Fig. 3 (IHC) (a) CK18 and (b) estrogen receptors (ER) label the
epithelial component; (c) CD10 and (d) smooth muscle heavy chain myosin (SMM) label the MEC component
typical combination of luminal cells arranged in exceeding one cell thickness, and nests of cells
ducts and chords surrounded by numerous atypical that have larger nuclei than normal MECs often
MECs. The MEC component forms layers have abundant eosinophilic or clear cytoplasm
272 Malignant Adenomyoepithelioma (M-AME)
Malignant Adenomyoepithelioma (M-AME), Fig. 4 (H&E) (a) Metastases of EMEC to lymph node and (c) to lung
retaining dual lineage. (IHC) (b, d) p63 highlights MECs
Malignant Adenomyoepithelioma (M-AME), Fig. 5 (H&E) (a) Carcinoma ex-AME: low-grade AME component;
(IHC) (b) CK14, (c) p63, and (d) SMM
Malignant Adenomyoepithelioma (M-AME) 273
Malignant Adenomyoepithelioma (M-AME), Fig. 6 Carcinoma ex-AME. (H&E) (a) Malignant component – disor-
ganized ducts and (b); atypical spindle cells immunostained by (c) p63 and (d) CK14
and show mitotic activity. The cell shape of the The morphology of the invasive carcinoma
M
MECs varies from polygonal through to spindled ex-AME depends on the type of carcinoma that
and often lies within an eosinophilic hyaline col- evolves (see classification above). The most com-
lagenous or chondroid matrix. The luminal cells mon variant is metaplastic carcinoma with spindle
may show squamous or sebaceous metaplasia. cells and foci of squamous differentiation, which
The spectrum of pathological changes seen in varies from low-grade through to tumors that have
benign AME has been published in detail else- the morphology of high-grade carcinosarcoma
where (Ali and Hayes 2012). Many cases of (Fig. 6). Sometimes the metaplastic carcinoma
AME, both benign and malignant, are initially mis- produces malignant chondroid tissue or osteoid
classified as invasive NST carcinoma or invasive and woven bone and may contain osteoclast-like
metaplastic carcinoma especially on core biopsy. giant cells (Fig. 7). Rare cases resemble carci-
Malignant transformation of the luminal cells may noma ex pleomorphic adenoma of salivary
result in the classical morphology of lobular carci- gland, which may be considered an entity distinct
noma in situ or low-grade cribriform DCIS with from malignant AME.
rounded uniform nuclei evenly spaced and polar-
ized around secondary lumens (Warrier et al. 2013).
Transformation to high-grade DCIS can also occur Ultrastructural Features
within an AME, a diagnosis that is based on the
cytological atypia, mitotic activity, and necrosis Electron microscopy may be helpful in confirming
and encompassing the morphological range of myoepithelial differentiation within M-AME. The
DCIS encountered unassociated with AME. myoepithelial cells exhibit the presence of both
AMEs harboring such in situ neoplastic prolifera- tonofilaments and myofibrils in their cytoplasm.
tions are probably best regarded as “borderline” or Pinocytotic vesicles are also seen. A thick basal
“atypical” AME rather than frankly malignant. lamina surrounds the cells, and hemidesmosomes
274 Malignant Adenomyoepithelioma (M-AME)
Malignant Adenomyoepithelioma (M-AME), Fig. 7 (H&E) (a) AME (b) transforming to EMEC and (c) metaplastic
chondroid and (d) osseous metaplasia in the MEC component
attach the MECs to adjacent cells and to the base- encounter positive staining of the luminal cell
ment membrane (Trojani et al. 1992). component for high molecular weight keratins,
and occasionally CD10 and SMM, especially
when the cells show squamous metaplasia but
Immunophenotype even when they resemble typical luminal epithe-
lial cells on morphology. Similarly, the MECs
The diagnosis of M-AME is often suspected can show staining for low molecular weight ker-
based on the morphology of the H&E stained atins, but this is usually much weaker than that
sections but is greatly supported by the use of seen in the luminal cell component of the lesion.
immunohistochemistry (Ali and Hayes 2012). Often the MECs lose their expected
The aim is to delineate a dual population of immunoprofile in an unpredictable pattern.
luminal cells and MECs within the lesion. In Thus, some MECs lose the myogenic markers
theory, this is simpler than in practice because and other cytoplasmic markers but retain the
the staining pattern of both the MECs and the nuclear markers, but the reverse pattern also
luminal cells in M-AME may be aberrant. occurs. The expression of the cytoplasmic
Classically, the luminal cells stain for low molec- markers is unpredictable and variable. Therefore
ular weight keratins such as CK18, CK7, and a wide range of immunostains may be required in
CAM5.2, and the MEC component stains with order to investigate a particular case thoroughly
nuclear markers p63 and p40 and cytoplasmic as sometimes only one of the many markers is
markers CD10 and S-100 protein, myogenic expressed. The immunostain for Ki-67 enables
markers (SMM, calponin, and actin), and high assessment of the degree of proliferative activity
molecular weight keratins CK 34-beta-E12, of M-AME (which typically has an index
CK5/6, and CK14. The expression of these of >15%) and indirectly reflects the malignant
markers varies from case to case and in different potential of the neoplasm (Hungermann et al.
areas of the same tumor. It is frequent to 2005). With regard to breast biomarkers, stains
Malignant Adenomyoepithelioma (M-AME) 275
for ER and progesterone receptors are patchy borderline) precursor AME intimately associ-
positive or completely negative in the luminal ated with the high-grade malignant epithelial
cell compartment in most M-AMEs especially or myoepithelial component of the neoplasm.
those with a metaplastic component, but some This depends on the history suggesting a pre-
cases show strong diffuse staining. These stains existing stable mass followed by a phase of
are negative in the myoepithelial or spindle cell accelerated growth, a careful gross examina-
compartment of the neoplasm. Staining for tion to search for such a precursor lesion,
androgen receptor does not appear to have been and/or a thorough sampling of the tumor.
studied in these neoplasms. HER2 is typically 3. Distinction from malignant myoepithelioma
not overexpressed or amplified. (▶ Myoepithelial Carcinoma of the Breast)
depends on the demonstration of both luminal
and MEC components in M-AME, whereas
Molecular Features luminal components are not seen in pure
malignant myoepithelioma.
A few cases of AME have been subject to genetic
and molecular analysis, and no clear pattern
has emerged. A p53 mutation has been identified References and Further Reading
in several cases of M-AME (Angèle et al. 2004;
Han et al. 2006) and a balanced t(8;16)(p23;q21) Ali, R. H., & Hayes, M. M. (2012). Combined epithelial-
translocation in a benign AME (Gatalica et al. myoepithelial lesions of the breast. Surgical Pathology
2005). A case report of a M-AME associated Clinics, 5(3), 661–699.
Angèle, S., Jones, C., Reis Filho, J. S., Fulford, L. G.,
with GIST tumors, both exhibiting c-kit mutation Treilleux, I., Lakhani, S. R., & Hall, J. (2004).
in a patient with NF-1, has also been reported Expression of ATM, p53, and the MRE11-Rad50-
(Hegyi et al. 2009). Comparative genomic hybrid- NBS1 complex in myoepithelial cells from benign
ization (CGH) analysis of benign AME showed and malignant proliferations of the breast. Journal of M
Clinical Pathology, 57(11), 1179–1184.
alterations comprising 13 gains and 5 losses Gatalica, Z., Velagaleti, G., Kuivaniemi, H., Tromp, G.,
of chromosomal regions in 5 of 7 cases. In Palazzo, J., Graves, K. M., Guigneaux, M., Wood, T.,
M-AME 14 of 18 cases showed DNA copy num- Sinha, M., & Luxon, B. (2005). Gene expression pro-
ber changes. Changes on chromosome 8 and 17p file of an adenomyoepithelioma of the breast with a
reciprocal translocation involving chromosomes 8 and
were most frequent (Hungermann et al. 2005). 16. Cancer Genetics and Cytogenetics, 156(1), 14–22.
Han, B., Mori, I., Nakamura, M., Wang, X., Ozaki, T.,
Nakamura, Y., & Kakudo, K. (2006). Myoepithelial car-
Differential Diagnosis cinoma arising in an adenomyoepithelioma of the breast:
Case report with immunohistochemical and mutational
analysis. Pathology International, 56(4), 211–216.
1. The differential diagnosis of M-AME Hegyi, L., Thway, K., Newton, R., Osin, P., Nerurkar, A.,
of EMEC type is from “benign” or “border- Hayes, A. J., & Fisher, C. (2009). Malignant
line” AME (▶ Adenomyoepithelioma). The myoepithelioma arising in adenomyoepithelioma of
the breast and coincident multiple gastrointestinal stro-
“malignant” designation is based on the pres- mal tumors in a patient with neurofibromatosis
ence of marked cytological atypia, presence of type 1. Journal of Clinical Pathology, 62(7), 653–655.
numerous mitoses and corresponding high Hungermann, D., Buerger, H., Oehlschlegel, C., Herbst, H.,
Ki-67 index (>15%), presence of necrosis, & Boecker, W. (2005). Adenomyoepithelial tumors and
myoepithelial carcinomas of the breast – A spectrum of
and an infiltrative growth pattern. monophasic and biphasic tumors dominated by imma-
2. Distinction between M-AME of carcinoma ture myoepithelial cells. BMC Cancer, 5, 92.
ex-AME type and metaplastic carcinoma Lee, S., Oh, S. Y., Kim, S.-H., et al. (2015). Malignant
(▶ Invasive Metaplastic Carcinoma) of the adenomyoepithelioma of the breast and responsive-
ness to eribulin. Journal of Breast Cancer, 18(4),
breast (including poorly differentiated 400–403.
myoepithelial-rich carcinoma) depends on the Nadelman, C. M., Leslie, K. O., & Fishbein, M. C. (2006).
demonstration of a low-grade (benign or “Benign,” metastasizing adenomyoepithelioma of the
276 Mammary Myofibroblastoma
breast: A report of 2 cases. Archives of Pathology & immunohistochemical, and ultrastructural level
Laboratory Medicine, 130(9), 1349–1353. (Wargotz et al. 1987b; Magro et al. 2001, 2002a,
Petrozza, V., Pasciuti, G., Pacchiarotti, A., et al. (2013).
Breast adenomyoepithelioma: A case report with 2016; Magro 2008). Other tumors, such as the
malignant proliferation of epithelial and benign fibroblastic spindle cell tumor, the spindle
myoepithelial elements. World Journal of Surgical cell lipoma of the breast, and tumors with hybrid
Oncology, 11, 285. features, have a fibroblastic profile, with cells
Trojani, M., Guiu, M., Trouette, H., De Mascarel, I., &
Cocquet, M. (1992). Malignant adenomyo- stained with vimentin and CD34 by means of immu-
epithelioma of the breast. An immunohistochemical, nohistochemistry (Magro et al. 2001, 2002a).
cytophotometric, and ultrastructural study of a case Although MFB was originally described as a spindle
with lung metastases. American Journal of Clinical cell tumor, over time several morphological variants
Pathology, 98(6), 598–602.
Warrier, S., Hwang, S., Ghaly, M., & Matthews, A. (2013). have been recognized: fibrous/collagenized,
Adenomyoepithelioma with ductal carcinoma in situ: myxoid, epithelioid/deciduoid cell, lipomatous, and
A case report and review of the literature. Case Reports palisaded/Schwannian-like variant (Magro et al.
in Surgery, 2013, 521417. 2000a; Reis-Filho et al. 2001; Simsir et al. 2001;
Yoon, J. Y., & Chitale, D. (2013). Adenomyoepithelioma of
the breast: A brief diagnostic review. Archives Magro 2008; Laforga and Escandón 2017).
of Pathology & Laboratory Medicine, 137(5), 725–729.
Clinical Features
Mammary Myofibroblastoma, Fig. 1 Classic-type arranged in short fascicles interrupted by keloid-like colla-
myofibroblastoma. Tumor showing well-circumscribed gen fibers (c) (H&E), are stained with desmin (d), CD34
borders (a) (H&E), bland-looking spindle cells and (e), alpha-smooth muscle actin (f) and estrogen
keloid-like collagen fibers (b) (H&E). Neoplastic cells, receptors (g)
278 Mammary Myofibroblastoma
Mammary Myofibroblastoma, Fig. 2 (H&E) Fibrous/ and histiocytes in their walls (a). Spindle cells, arranged in
collagenized myofibroblastoma. Low magnification show- distorted fascicles, are seen at higher magnification (b)
ing a fibrous lesion containing blood vessels with fibrin
Mammary Myofibroblastoma, Fig. 3 Myxoid myo- fibers (a) (H&E). Neoplastic cells show focal nuclear atypia
fibroblastoma. Tumor with extensive myxoid stroma (b) (H&E) and are stained with desmin (c)
containing spindle- to stellate-shaped cells and thick collagen
Mammary Myofibroblastoma 281
Mammary Myofibroblastoma, Fig. 4 (H&E) Epitheli- magnification showing mono- or bi-nucleated cells with
oid myofibroblastoma. Tumor is composed predominantly of mild to moderate nuclear atypia. Mitoses are absent (b)
epithelioid cells set in a fibrous stroma (a). Higher
M
“Palisaded/Schwannian-like MFB” is composed of some cases, can be absent. Accordingly, the major-
bland-looking spindle cells with a diffuse nuclear ity of mammary MFBs are desmin-positive
palisading and formation of numerous Verocay- myofibroblastic tumors which usually express estro-
like bodies (Magro et al. 2013b; Laforga and gen, progesterone, and androgen receptors (Magro
Escandón 2017) (Fig. 7). Keloid-like collagen et al. 2000b, 2012a; Magro 2008, 2016). Calponin,
fibers, a typical feature of classic-type MFB, are bcl-2 protein, CD99, and CD10 are also variably
seen. This tumor is closely reminiscent of expressed (Magro et al. 2007b; Magro 2016).
schwannoma and immunohistochemistry is helpful H-caldesmon expression can be focally observed
for a correct diagnosis (absence of S100 protein (Magro et al. 2003). Epithelial markers
along with the expression of myogenic markers). (cytokeratins, EMA) and S100 protein are negative.
Classic-type MFB and all its morphological vari- Cytogenetic studies have showed that MFB is a
ants show a myofibroblastic profile. The neoplastic tumor with the loss of material from chromosome
cells are characteristically stained with desmin in 13 and less frequently from chromosome
addition to vimentin and CD34 (Magro 2008, 16 (Pauwels et al. 2000). FISH analyses, using
2016). Alpha-smooth muscle actin is also locus specific probes, reveal that about 70–80% of
expressed, but with a more intralesional and mammary MFB exhibits the loss of the 13q14
interlesional heterogeneous distribution and, in region, which can be shown by the losses of
282 Mammary Myofibroblastoma
Mammary Myofibroblastoma, Fig. 5 Deciduoid mono- or multinucleated cells with abundant cytoplasm
myofibroblastoma. Tumor is composed of closely packed and large nuclei with evident nucleoli (b) (H&E). Neoplas-
large-sized cells closely reminiscent of decidua (a) (H&E). tic cells are diffusely stained with desmin (c)
Higher magnification showing alarming features: large
RB/13q14 and/or FOX1(FKHR)/13q14 loci in mitoses 10 high power fields), atypical mitoses,
tumor cells (Magro et al. 2012b; Trépant et al. necrosis, and extensive infiltrative margins are not
2014). Interestingly similar results have been features of MFB. The diagnosis is challenging on
reported in extra-mammary MFB (Maggiani core biopsy, but MFB can be suspected if tumor has
et al. 2006; Magro et al. 2012b). As spindle cell pushing borders (imaging information) and the
lipoma and cellular angiofibroma share with MFB spindle cells are positive for desmin, CD34,
the same chromosomal alterations (Maggiani alpha-smooth muscle actin, estrogen/progesterone
et al. 2007; Flucke et al. 2011), it has been receptors and negative for epithelial markers,
suggested a genetic link among these three tumors h-caldesmon, beta-catenin, S100 protein, and
(Flucke et al. 2011; Magro et al. 2012b). STAT6. The differential diagnosis includes all the
bland-looking tumor or tumor-like spindle cell
lesions arising in the breast parenchyma, such as
Differential Diagnosis the fascicular variant of pseudoangiomatous
stromal hyperplasia (▶ Pseudoangiomatous Stro-
The diagnosis of classic-type MFB is relatively mal Hyperplasia), reactive spindle cell nodule,
straightforward in surgical specimens if strict nodular fasciitis, inflammatory pseudotumor/
morphological criteria are applied. MFB of the inflammatory myofibroblastic tumor (▶ Inflamma-
breast should be suspected when dealing with a tory Myofibroblastic Tumor of the Breast),
pure spindle cell lesion with pushing borders, leiomyoma, solitary fibrous tumor, desmoid-type
composed of bland-looking spindle cells, fibromatosis, low-grade (fibromatosis-like) spindle
arranged in short, haphazardly intersecting fasci- cell carcinoma (▶ Invasive Metaplastic Carci-
cles with interspersed keloid-like collagen fibers noma), and low-grade myofibroblastic sarcoma
(Magro 2008, 2016). High mitotic count (>3 (Table 3). Among these lesions, the distinction
Mammary Myofibroblastoma 283
Mammary Myofibroblastoma, Fig. 6 Lipomatous growth pattern (b) (H&E). Fibrous area exhibiting the
myofibroblastoma. Low-magnification showing a fibro- characteristic features of classic-type myofibroblastoma M
fatty tumor with well-circumscribed borders (a) (H&E). (c) (H&E). The spindle cells are diffusely stained
In some areas, there is a close admixture of fibrous areas with CD34 (d)
with adipose tissue, resulting in a pseudo-infiltrative
Mammary Myofibroblastoma, Fig. 7 (H&E) picture is highly suggestive of schwannoma (a). High mag-
Palisaded/Schwannian-like myofibroblastoma. Low- nification showing the cellular details (b). Neoplastic cells
magnification showing a tumor with well-circumscribed are stained with desmin (inset)
borders and numerous Verocay-like bodies. The overall
stroma and a tissue culture/granulation-like h-caldesmon, while CD34 is lacking (Jones et al.
appearance (Kang et al. 2015; Paliogiannis et al. 1994; Vecchio et al. 2013). As solitary fibrous
2016; Hayashi et al. 2017). Numerous mitoses tumor and MFB share several morphological and
and entrapped ducts/lobules can be seen, espe- immunohistochemical features, some authors
cially at the periphery of the lesion. The cells of have used interchangeably their names in the
nodular fasciitis are diffusely stained with alpha- past (Damiani et al. 1994; Magro et al. 2002a).
smooth muscle actin but are usually negative However, unlike MFB, SFT shows nuclear
for desmin and CD34 (Kang et al. 2015; expression of STAT6 (Magro et al. 2016) and is
Paliogiannis et al. 2016; Hayashi et al. 2017). usually desmin-negative. Desmoid type-
Unlike MFB, inflammatory pseudotumor/inflam- fibromatosis differs from MFB in that it is an
matory myofibroblastic tumor contains spindle infiltrative lesion composed of spindle cells, usu-
cells admixed with chronic inflammatory infiltrate ally aligned parallel to one another, separated by
of plasma cells, lymphocytes, and less frequently fibrous stroma and arranged in long fascicles
eosinophils and neutrophils. The spindle cells (Wargotz et al. 1987a; Devouassoux-Shisheboran
show a variable expression of alpha-smooth mus- et al. 2000; Magro et al. 2002b). In addition, the
cle actin, while desmin and CD34 are usually cells are usually stained with beta-catenin (nuclear
negative or only focally positive (Vecchio et al. staining) and alpha-smooth muscle actin, while
2011; Zhou et al. 2013; Bosse et al. 2014). ALK-1 they are negative for desmin and CD34 (Magro
protein can be expressed in about 40–50% of 2016). Unlike MFB, low-grade (fibromatosis-
cases (Zhou et al. 2013; Bosse et al. 2014). like) spindle cell carcinoma exhibits, at least
Apart from the more eosinophilic cytoplasm of focally, infiltrative margins with projections into
the cells and the more organized fascicular growth adjacent mammary parenchyma (Wargotz et al.
pattern, leiomyoma can be distinguished from 1989; Gobbi et al. 1999; Sneige et al. 2001; Carter
MFB by demonstrating diffuse coexpression of et al. 2006; Dwyer and Clark 2015).
desmin, alpha-smooth muscle actin, and Immunostaining of neoplastic cells, at least
Mammary Myofibroblastoma 285
focally, with epithelial (cytokeratins) and progesterone receptors, while it is negative for
myoepithelial (p63) markers is mandatory for beta-catenin (Magro et al. 2000a). Epithelioid-
making the diagnosis of spindle cell carcinoma. cell MFB may be misinterpreted as an invasive
Apart from the spindle cells, cytokeratins usually lobular carcinoma (▶ Invasive Lobular Carci-
highlight the neoplastic cells with epithelioid to noma) as the neoplastic cells may adopt a single
polygonal morphology, often arranged in small cell and/or single-cell files growth patterns
cohesive clusters (Dwyer and Clark 2015). This (Magro 2009, 2012; Magro et al. 2013a). How-
finding is an important diagnostic clue. Finally, ever, lobular carcinoma has infiltrative margins,
low-grade myofibroblastic sarcoma (low-grade expresses epithelial markers (cytokeratins and
myofibrosarcoma) is a tumor with higher mitotic EMA), and is negative for myogenic markers
activity (7 to 35 mitoses per10 high power fields) (desmin, alpha-smooth muscle actin). Patholo-
and stained with alpha-smooth muscle actin, gists should also keep in mind that epithelioid
while the expression of desmin and CD34 is cells may occasionally exhibit a deciduoid-like
lacking (Taccagni et al. 1997; Gocht et al. 1999; appearance, making more difficult the recognition
Lucin et al. 2003; Morgan et al. 2005). of a tumor as MFB (Magro et al. 2008). Deciduoid
Among the morphological variants of MFB, cell MFB should be distinguished from malignant
lipomatous and epithelioid/deciduoid cell MFB tumors such as invasive pleomorphic lobular carci-
are unusual enough to deserve a separate differ- noma, apocrine carcinoma, metastatic pleomorphic
ential diagnosis. Due to their alarming morphol- rhabdomyosarcoma, melanoma, or malignant
ogy, they can represent potential diagnostic rhabdoid tumor (Magro et al. 2008). The well-
pitfalls and confused with more aggressive circumscribed margins, the absence or low mitotic
lesions, especially in core biopsies (Magro 2009; count (up 2 mitoses 10 HPF), the presence of
Wahbah et al. 2011; Bakuła-Zalewska et al. 2012; keloid-like collagen fibers, and the expression of
Alizadeh et al. 2015; Arafah et al. 2015). The myogenic markers are all features supporting the
former is an uncommon variant, which shows a diagnosis of MFB (Magro et al. 2008).
close admixture of spindle cells with adipocytes, Interestingly some cases of mammary MFB
resulting in a fibromatosis-like growth pattern. may exhibit hybrid features with a lipomatous com-
However, lipomatous MFB has well- ponent intermingling with epithelioid cells
circumscribed margins and the lipomatous com- (Wahbah et al. 2011; Alizadeh et al. 2015). These
ponent is an integral part of the tumor and not are diagnostically challenging cases which are
entrapped tissue due to infiltration of neoplastic often misdiagnosed as invasive lobular carcinomas
cells into adjacent breast parenchyma (Magro (▶ Invasive Lobular Carcinoma) in core biopsy due
et al. 2000a). Unlike desmoid-type fibromatosis, to the apparent pseudo-infiltrative pattern of epithe-
it is stained with desmin, CD34, and estrogen/ lioid cells, singly dispersed or arranged in files,
Mammary Myofibroblastoma 287
admixed with the intratumoral fatty tissue. The Flucke, U., van Krieken, J. H., & Mentzel, T. (2011).
diagnosis of invasive lobular carcinoma will prompt Cellular angiofibroma: Analysis of 25 cases emphasiz-
ing its relationship to spindle cell lipoma and
pathologists to perform immunohistochemical ana- mammary-type myofibroblastoma. Modern Pathology,
lyses for defining the tumor profile and the demon- 24, 82–89.
stration of estrogen/progesterone receptors, Fukunaga, M., & Ushigome, S. (1997). Myofibroblastoma
typically observed in MFB, will further support of the breast with diverse differentiation. Archives of
Pathology & Laboratory Medicine, 121, 599–603.
the mistaken diagnosis of carcinoma (Magro 2009; Fukunaga, M., Endo, Y., & Ushigome, S. (1996). Atypical
Wahbah et al. 2011). Awareness of the lipomatous leiomyomatous features in myofibroblastoma of the
and epithelioid/deciduoid cell MFB is crucial for breast. Histopathology, 29, 592–593.
pathologists, who should always correlate morphol- Garijo, M. F., Val-Bernal, J. F., Vega, A., & Val, D. (2008).
Postoperative spindle cell nodule of the breast: Pseudo-
ogy with clinico-radiologic information. Immuno- sarcomatous myofibroblastic proliferation following
histochemistry, including myogenic and epithelial endosurgery. Pathology International, 58, 787–791.
markers, is mandatory in ambiguous cases. Gobbi, H., Simpson, J. F., Borowsky, A., Jensen, R. A., &
Page, D. L. (1999). Metaplastic breast tumors with a
dominant fibromatosis-like phenotype have a high risk
of local recurrence. Cancer, 85, 2170–2182.
References and Further Reading Gobbi, H., Tse, G., Page, D. L., Olson, S. J., Jensen, R. A.,
& Simpson, J. F. (2000). Reactive spindle cell nodules
Alam, F. M., Samarasinghe, D. S., & Pillai, R. G. (2002). of the breast after core biopsy or fine-needle aspiration.
Myofibroblastoma of the breast in an adolescent. Saudi American Journal of Clinical Pathology, 113,
Medical Journal, 23, 232–233. 288–294.
Alizadeh, L., Alkhasawneh, A., Reith, J. D., & Gocht, A., Bosmuller, H. C., Bassler, R., Tavassoli, F. A.,
Al-Quran, S. Z. (2015). Epithelioid myofibro- Moinfar, F., Katenkamp, D., Schirrmacher, K., Lüders,
blastoma of the breast: A report of two cases with P., & Saeger, W. (1999). Breast tumors with
discussion of diagnostic pitfalls. The Breast Journal, myofibroblastic differentiation: Clinicopathological
21, 669–673. observations in myofibroblastoma and
Arafah, M. A., Ginter, P. S., D’Alfonso, T. M., & Hoda,
S. A. (2015). Epithelioid mammary myofibroblastoma
myofibrosarcoma. Pathology, Research and Practice,
195, 1–10.
M
mimicking invasive lobular carcinoma. International Gurzu, S., & Jung, I. (2012). Male breast cellular myofibro-
Journal of Surgical Pathology, 23, 284–288. blastoma with a rich reticulinic network: Case report.
Bakuła-Zalewska, E., Piasek, P., Wawryszuk, J., & American Journal of Men’s Health, 6, 344–348.
Domanski, H. A. (2012). Myofibroblastoma: Hamele-Bena, D., Cranor, M., Sciotto, C., Erlandson, R.,
A potential pitfall in core needle biopsy of breast & Rosen, P. P. (1996). Uncommon presentation of
lesions. Polish Journal of Pathology, 63, 131–133. mammary myofibroblastoma. Modern Pathology, 9,
Baxendine-Jones, J., Theaker, J. M., & Baldwin, L. J. 786–790.
(2001). Predominant fatty variant of myofibrolastoma Hayashi, S., Yasuda, S., Takahashi, N., Okazaki, S.,
of breast. Journal of Clinical Pathology, 54, 568–569. Ishibashi, K., Kitada, M., & Miyokawa, N. (2017).
Bosse, K., Ott, C., Biegner, T., Fend, F., Siegmann-Luz, K., Nodular fasciitis of the breast clinically resembling
Wallwiener, D., & Hahn, M. (2014). 23-year-old breast cancer in an elderly woman: A case report.
female with an inflammatory myofibroblastic tumour Journal of Medical Case Reports, 11, 57.
of the breast: A case report and a review of the litera- Jones, M. W., Norris, H. J., & Wargotz, E. S. (1994).
ture. Geburtshilfe und Frauenheilkunde, 74, 167–170. Smooth muscle and nerve sheath tumors of the breast.
Carter, M. R., Hornick, J. L., Lester, S., & Fletcher, C. D. A clinicopathologic study of 45 cases. International
(2006). Spindle cell (sarcomatoid) carcinoma of the Journal of Surgical Pathology, 2, 85–92.
breast: A clinicopathologic and immunohistochemical Kang, A., Kumar, J. B., Thomas, A., & Bourke, A. G. (2015).
analysis of 29 cases. The American Journal of Surgical A spontaneously resolving breast lesion: Imaging and
Pathology, 30, 300–309. cytological findings of nodular fasciitis of the breast
Damiani, S., Miettinen, M., Peterse, J. L., & Eusebi, with FISH showing gene rearrangement. BMJ Case
V. (1994). Solitary fibrous tumour (myofibroblastoma) Reports. pii: bcr2015213076. https://doi.org/10.1136/
of the breast. Virchows Archiv, 425, 89–92. bcr-2015-213076.
Devouassoux-Shisheboran, M., Schammel, M. D. P., Man, Kobayashi, N., Oda, K., Yokoi, S., Kanda, H., Hayakawa,
Y. G., & Tavassoli, F. A. (2000). Fibromatosis of the S., Tang, X., & Osamura, Y. (1996). Myofibroblastoma
breast. Archives of Pathology & Laboratory Medicine, of the breast: Report of a case. Surgery Today, 26,
124, 276–280. 727–729.
Dwyer, J. B., & Clark, B. Z. (2015). Low-grade fibromatosis- Laforga, J. B., & Escandón, J. (2017). Schwannoma-like
like spindle cell carcinoma of the breast. Archives of (Palisaded) myofibroblastoma: A challenging diagno-
Pathology & Laboratory Medicine, 139, 552–557. sis on core biopsy. The Breast Journal, 23, 354–356.
288 Mammary Myofibroblastoma
Lázaro-Santander, R., García-Prats, M. D., Nieto, S., Magro, G., Gurrera, A., Scavo, N., Lanzafame, S., &
Andrés-Gozalvo, C., Cortés-Vizcaino, V., Vargas- Bisceglia, M. (2002b). Fibromatosis of the breast:
Holguin, S., & Vera-Roman, J. M. (1999). Myofibro- A clinical, radiological and pathological study of
blastoma of the breast with diverse histological fea- 6 cases. Pathologica, 94, 238–246.
tures. Virchows Archiv, 434, 547–550. Magro, G., Gurrera, A., & Bisceglia, M. (2003).
Lee, J. W., Jung, G. S., Kim, J. B., Choi, K. Y., Chung, H-caldesmon expression in myofibroblastoma of the
H. Y., Cho, B. C., Park, J. Y., Kim, H. J., Park, H. Y., & breast: Evidence supporting the distinction from
Yang, J. D. (2016). Pseudoangiomatous stromal hyper- leiomyoma. Histopathology, 42, 233–238.
plasia presenting as rapidly growing bilateral breast Magro, G., Amico, P., & Gurrera, A. (2007a). Myxoid
enlargement refractory to surgical excision. Archives myofibroblastoma of the breast with atypical cells:
of Plastic Surgery, 43, 218–221. A potential diagnostic pitfall. Virchows Archiv, 450,
Lucin, K., Mustać, E., & Jonjić, N. (2003). Breast sarcoma 483–485.
showing myofibroblastic differentiation. Virchows Magro, G., Caltabiano, R., Di Cataldo, A., & Puzzo,
Archiv, 443, 222–224. L. (2007b). CD10 is expressed by mammary myofibro-
Maggiani, F., Debiec-Rychter, M., Verbeeck, G., & Sciot, blastoma and spindle cell lipoma of soft tissue: An
R. (2006). Extramammary myofibroblastoma is genet- additional evidence of their histogenetic linking.
ically related to spindle cell lipoma. Virchows Archiv, Virchows Archiv, 450, 727–728.
449, 244–247. Magro, G., Gangemi, P., & Greco, P. (2008). Deciduoid-
Maggiani, F., Debiec-Rychter, M., Vanbockrijck, M., & like myofibroblastoma of the breast: A potential pitfall
Sciot, R. (2007). Cellular angiofibroma: Another mes- of malignancy. Histopathology, 52, 652–654.
enchymal tumour with 13q14 involvement, suggesting Magro, G., Fletcher, C. D. M., & Eusebi, V. (2012a). WHO
a link with spindle cell lipoma and (extra)-mammary classification of tumours of the breast. In S. Lakhani,
myofibroblastoma. Histopathology, 51, 410–412. I. O. Ellis, S. J. Schnitt, P. H. Tan, & M.J. van de Vijver
Magro, G. (2008). Mammary myofibroblastoma: A tumor M.J. (Eds.), Myofibroblastoma (pp. 130–131). Lyon:
with a wide morphologic spectrum. Archives of Pathol- IARC press.
ogy & Laboratory Medicine, 132, 1813–1820. Magro, G., Righi, A., Casorzo, L., Torrisi, A., Salvatorelli,
Magro, G. (2009). Epithelioid-cell myofibroblastoma of the L., Kacerovská, D., & Kazakov, D. (2012b). Mammary
breast. Expanding the morphologic spectrum. The Amer- and vaginal myofibroblastomas are genetically related
ican Journal of Surgical Pathology, 33, 1085–1092. lesions: Fluorescence in situ hybridization analysis
Magro, G. (2012). Epithelioid cell myofibroblastoma of the shows deletion of 13q14 region. Human Pathology,
breast. A potential diagnostic pitfall. The Breast Jour- 43, 1887–1893.
nal, 18, 278–279. Magro, G., Vecchio, G. M., Michal, M., & Eusebi,
Magro, G. (2016). Mammary myofibroblastoma: An update E. (2013a). Atypical epithelioid cell myofibroblastoma
with emphasis on the most diagnostically challenging of the breast with multinodular growth pattern:
variants. Histology and Histopathology, 31, 1–23. A potential pitfall of malignancy. Pathology, Research
Magro, G., & Bisceglia, M. (2005). Myofibroblastoma-like and Practice, 209, 463–466.
changes in fibro(stroma)-epithelial lesions of the breast: Magro, G., Foschini, M. P., & Eusebi, V. (2013b).
Report of two cases. Virchows Archiv, 446, 95–96. Palisaded myofibroblastoma of the breast: A tumor
Magro, G., Fraggetta, F., Torrisi, A., Emmanuele, C., & closely mimicking schwannoma. Report of two cases.
Lanzafame, S. (1999). Myofibroblastoma of the breast Human Pathology, 44, 1941–1946.
with haemagiopericytoma- like pattern and pleomor- Magro, G., Salvatorelli, L., Spadola, S., & Angelico, G. (2014).
phic lipoma-like areas. Pathology, Research and Prac- Mammary myofibroblastoma with extensive
tice, 195, 257–262. myxoedematous stromal changes: A potential diagnostic
Magro, G., Michal, M., Vasquez, E., & Bisceglia, pitfall. Pathology, Research and Practice, 210, 1106–1111.
M. (2000a). Lipomatous myofibroblastoma: A potential Magro, G., Angelico, G., Leone, G., & Palazzo, J. (2016).
diagnostic pitfall in the spectrum of the spindle cell Solitary fibrous tumor of the breast: Report of a case
lesions of the breast. Virchows Archiv, 437, 540–544. with emphasis on diagnostic role of STAT6
Magro, G., Bisceglia, M., & Michal, M. (2000b). Expres- immunostaining. Pathology, Research and Practice,
sion of steroid hormone receptors, their regulated pro- 212, 463–477.
teins, and bcl-2 protein in myofibroblastoma of the Mnif, H., Charfi, S., Abid, N., & Sallemi-Boudawara,
breast. Histopathology, 36, 515–521. T. (2013). Mammary myofibroblastoma with
Magro, G., Michal, M., & Bisceglia, M. (2001). Benign leiomyomatous differentiation: Case report and litera-
spindle cell tumors of the mammary stroma: Diagnostic ture review. Pathologica, 105, 142–145.
criteria, classification, and histogenesis. Pathology, Morgan, P. B., Chundru, S., Hatch, S. S., Hawkins, H. K.,
Research and Practice, 197, 453–466. Adegboyega, P. A., & Eltorky, M. A. (2005). Uncom-
Magro, G., Bisceglia, M., Michal, M., & Eusebi, mon malignancies: Case 1. Low-grade myofibroblastic
V. (2002a). Spindle cell lipoma-like tumor, solitary sarcoma of the breast. Journal of Clinical Oncology,
fibrous tumor and myofibroblastoma of the breast: 23, 6249–6251.
A clinicopathological analysis of 13 cases in favor of Ozerdem, U., Wells, J., & Hoda, S. A. (2015). Hyaline
a unifying histologic concept. Virchows Archiv, 440, globules in mammary myofibroblastoma: A case report.
249–260. International Journal of Surgical Pathology, 23, 89–91.
Microglandular Adenosis 289
Paliogiannis, P., Cossu, A., Palmieri, G., Scognamillo, F., Virk, R. K., & Khan, A. (2010). Pseudoangiomatous stro-
Pala, C., Nonnis, R., Sotgiu, G., Fois, A., Palomba, G., mal hyperplasia an overview. Archives of Pathology &
& Attene, F. (2016). Breast nodular fasciitis: Laboratory Medicine, 134, 1070–1074.
A comprehensive review. Breast Care (Basel), 11, Wahbah, M. M., Michael, Z., Gilcrease, M. D., & Yun,
270–274. W. (2011). Lipomatous variant of myofibroblastoma
Pauwels, P., Sciot, R., Croiset, F., Rutten, H., Van den with epithelioid features: A rare and diagnostically
Berghe, H., & Dal Cin, P. (2000). Myofibroblastoma challenging breast lesion. Annals of Diagnostic Pathol-
of the breast: Genetic link with spindle cell lipoma. The ogy, 15, 454–458.
Journal of Pathology, 191, 282–285. Wargotz, E. S., Norris, H. J., Austin, R. M., & Enzinger,
Reis-Filho, J. S., Faoro, L. N., Gasparetto, E. L., Totsugui, F. M. (1987a). Fibromatosis of the breast. A clinical and
J. T., & Schmitt, F. C. (2001). Mammary epithelioid pathological study of 28 cases. The American Journal
myofibroblastoma arising in bilateral gynecomastia: of Surgical Pathology, 11, 38–44.
Case report with immunohistochemical profile. Inter- Wargotz, E. S., Weiss, S. W., & Norris, H. J. (1987b).
national Journal of Surgical Pathology, 9, 331–334. Myofibroblastoma of the breast. Sixteen cases of a
Sciallis, A. P., Chen, B., & Folpe, A. L. (2012). Cellular distinctive benign mesenchymal tumor. The
spindled histiocytic pseudotumor complicating mam- American Journal of Surgical Pathology, 11,
mary fat necrosis. The American Journal of Surgical 493–502.
Pathology, 36, 1571–1578. Wargotz, E. S., Deos, P. H., & Norris, H. J. (1989). Meta-
Simsir, A., Cangiarella, J., Boppana, S., & Waisman, plastic carcinomas of the breast. II. Spindle cell carci-
J. (2001). Aspiration cytology of the collagenized var- noma. Human Pathology, 20, 732–740.
iant of mammary myofibroblastoma: A case report with Yoo, C. C., Pui, J. C., & Torosian, M. H. (1998). Myofibro-
review of the literature. Diagnostic Cytopathology, 24, blastoma associated with bilateral gynecomastia:
399–402. A case report and literature review. Oncology Reports,
Sneige, N., Yaziji, H., Mandavilli, S. R., Perez, E. R., 5, 731–733.
Ordonez, N. G., Gown, A. M., & Ayala, A. (2001). Zhou, Y., Zhu, J., Zhang, Y., Jiang, J., & Jia, M (2013). An
Low-grade (fibromatosis-like) spindle cell carcinoma inflammatory myofibroblastic tumour of the breast
of the breast. The American Journal of Surgical Pathol- with ALK overexpression. BMJ Case Reports. pii:
ogy, 25, 1009–1016. bcr0720114474. https://doi.org/10.1136/bcr-07-2011-
Soyer, T., Ayva, S., Senyucel, M. F., Senyucel, C., Aslan, 4474.
M. K., & Cakmak, M. (2012). Myofibroblastoma of
breast in a male infant. Fetal and Pediatric Pathology,
M
31, 164–168.
Taccagni, G., Rovere, E., Masullo, M., Christensen, L., &
Eyden, B. (1997). Myofibrosarcoma of the breast:
Review of the literature on myofibroblastic tumors and Microglandular Adenosis
criteria for defining myofibroblastic differentiation. The
American Journal of Surgical Pathology, 21, 489–496. Eliano Cascardi1,2, Caterina Marchiò2,3,4 and
Teng, X. D., & You, Q. H. (2005). Infiltrating myofibro-
blastoma of the breast in female: A case report. Eugenio Maiorano1
1
Zhonghua Bing Li Xue Za Zhi, 34, 186. Department of Emergency and Organ
Thomas, T. M., Myint, A., Mak, C. K., & Chan, J. K. Transplantation, Unit of Pathology, University
(1997). Mammary myofibroblastoma with of Bari, Bari, Italy
leiomyomatous differentiation. American Journal of 2
Clinical Pathology, 107, 52–55. Unit of Pathology, Candiolo Cancer Institute
Trépant, A. L., Sibille, C., Frunza, A. M., Simon, P., & FPO-IRCCS, Candiolo, Italy
3
Noël, J. C. (2014). Myofibroblastoma of the breast with Department of Medical Sciences,
smooth muscle differentiation showing deletion of University of Turin, Turin, Italy
13q14 region: Report of a case. Pathology, Research 4
and Practice, 210, 389–391. Institut Curie, Paris, France
Vecchio, G. M., Amico, P., Grasso, G., Vasquez, E., La
Greca, G., & Magro, G. (2011). Post-traumatic inflam-
matory pseudotumor of the breast with atypical mor- Definition
phological features: A potential diagnostic pitfall.
Report of a case and a critical review of the literature.
Pathology, Research and Practice, 207, 322–326. Microglandular adenosis (MGA) of the breast is
Vecchio, G. M., Cavaliere, A., Cartaginese, F., a glandular proliferation of uncertain malignant
Lucaccioni, A., Lombardi, T., Parenti, R., potential, which mimics invasive carcinoma due
Salvatorelli, L., & Magro, G. (2013). Intraparen-
chymal leiomyoma of the breast: Report of a case to haphazard infiltrative growth in breast stroma
with emphasis on needle core biopsy-based diagnosis. and the lack of a myoepithelial cell layer. The
Pathologica, 105, 122–127. term atypical microglandular adenosis (AMGA)
290 Microglandular Adenosis
M
Microglandular Adenosis, Fig. 1 Representative pho- for S100 protein. (c) Estrogen and progesterone receptors
tomicrographs of microglandular adenosis diagnosed are not expressed. (d) Absence of a myoepithelial cell layer
on core biopsy. (a) The microglandular proliferation around the glandular structures, as shown by immunohis-
shows an infiltrative growth pattern and is haphazardly tochemistry with anti-p63 antibody (a normal lobule serves
distributed in the fibro-fatty mammary tissue (H&E). (b) as internal positive control)
The cells lining the tubules show strong nuclear staining
Immunophenotype
Molecular Features
MGA and AMGA are characterized by a triple
negative phenotype, lacking expression for Genetic analyses by array-based comparative
estrogen receptor, progesterone receptor, and genomic hybridization and next-generation
292 Microglandular Adenosis
sequencing have shown that MGA and AMGA AMGA, the glandular distribution of tubular car-
associated with triple negative carcinomas share cinoma is usually stellate, the glands are angular
clonal genetic alterations with invasive carcino- and the stroma is frequently desmoplastic. Most
mas, such as TP53 mutations (Guerini-Rocco important, tubular carcinomas typically are estro-
et al. 2016; Geyer et al. 2009, 2012; Shin et al. gen receptor positive.
2009). Of note, pure MGA/AMGA (i.e., with MGA and AMGA also share striking similarities
no association with an infiltrative carcinoma) fea- with acinic cell carcinomas (▶ Acinic Cell Carci-
ture simpler genetic profiles and lack TP53 muta- noma), showing a microglandular growth pattern, a
tions (Guerini-Rocco et al. 2016; Geyer et al. triple negative phenotype, and S100 protein expres-
2012). Taken together, these data suggest that sion. Contrary to MGA/AMGA, acinic cell carcino-
a subset of MGA/AMGA can be considered as mas may display a solid pattern with comedo-like
a clonal neoplastic lesion and a non-obligate pre- necrosis and intracytoplasmic zymogen-type gran-
cursor of high-grade, triple negative breast cancer, ules, relevant mitotic activity and express high
underpinned by TP53 mutations. levels of EMA and acinar differentiation markers
It is interesting to note that MGA/AMGA (lysozyme and amylase) (Lakhani et al. 2012).
and acinic cell carcinomas (ACC) (▶ Acinic Cell
Carcinoma), a special histologic subtype of
triple negative breast cancer, share histologic and References and Further Reading
immunophenotypic similarities. When analyzed at
the genetic level, no significant differences Clement, P. B., & Azzopardi, J. G. (1983). Microglandular
in mutation burden and repertoire were detected adenosis of the breast – A lesion simulating tubular
carcinoma. Histopathology, 7, 169–180.
between MGA/AMGA/ACC analyzed (either indi- Foschini, M. P., & Eusebi, V. (2018). Microglandular
vidually or as a whole group) and conventional adenosis of the breast: A deceptive and still mysterious
triple negative breast carcinomas (Geyer et al. benign lesion. Human Pathology, 82, 1–9.
2017). These data support the existence of a “low- Geyer, F. C., Kushner, Y. B., Lambros, M. B., Natrajan, R.,
Mackay, A., Tamber, N., . . . & Reis-Filho, J. S. (2009).
grade breast neoplasia family,” which encompasses Microglandular adenosis or microglandular adenoma?
triple negative lesions such as MGA, AMGA, and A molecular genetic analysis of a case associated with
ACC that, despite their low-grade morphology and atypia and invasive carcinoma. Histopathology, 55,
good outcome, recapitulate the complex genomic 732–743.
Geyer, F. C., Lacroix-Triki, M., Colombo, P. E., Patani, N.,
landscape of common forms of triple negative Gauthier, A., Natrajan, R., . . . & Marchio, C. (2012).
breast cancers (Geyer et al. 2017). Molecular evidence in support of the neoplastic and
precursor nature of microglandular adenosis.
Histopathology, 60, E115–E130.
Geyer, F. C., Berman, S. H., Marchiò, C., Burke, K. A.,
Differential Diagnosis Guerini-Rocco, E., Piscuoglio, S., . . . & Schnitt, S. J.
(2017). Genetic analysis of microglandular adenosis and
Numerous breast lesions may display a micro- acinic cell carcinomas of the breast provides evidence for
glandular pattern, such as sclerosing adenosis the existence of a low-grade triple-negative breast neo-
plasia family. Modern Pathology, 30, 69.
(▶ Sclerosing Adenosis), adenomyoepithelial Guerini-Rocco, E., Piscuoglio, S., Ng, C. K., Geyer, F. C.,
adenosis (▶ Adenosis, Other Types), or tubular De Filippo, M. R., Eberle, C. A., . . . & Yatabe, Y. (2016).
carcinoma (▶ Tubular Carcinoma), and can be Microglandular adenosis associated with triple-negative
misinterpreted as MGA (Foschini and Eusebi breast cancer is a neoplastic lesion of triple-negative
phenotype harbouring TP53 somatic mutations. The
2018). Contrary to MGA, sclerosing adenosis Journal of Pathology, 238, 677–688.
shows a dense and hyalinized stroma that creates James, B. A., Cranor, M. L., & Rosen, P. P. (1993).
distorted glandular structures. Of note, sclerosing Carcinoma of the breast arising in microglandular
adenosis and adenomyoepithelial adenosis retain adenosis. American Journal of Clinical Pathology,
100, 507–513.
a myoepithelial cell layer (Foschini and Eusebi Khalifeh, I. M., Albarracin, C., Diaz, L. K.,
2018). Adenomyoepithelial adenosis also is Symmans, F. W., Edgerton, M. E., Hwang, R. F., &
strongly immunoreactive for GCDFP-15. Unlike Sneige, N. (2008). Clinical, histopathologic, and
Microinvasive Carcinoma of the Breast 293
Microinvasive Carcinoma of the Breast, Fig. 2 Microinvasive Carcinoma of the Breast, Fig. 3
(H&E): Microinvasive lobular carcinoma associated (H&E): Microinvasive lobular carcinoma not associated
with LCIS with ductal or lobular carcinoma in situ
microinvasion (DCIS-MI) type 1 and type 2, accepted agreement particularly about the maxi-
respectively. mum diameter compatible with a diagnosis of
microinvasion.
This lack of a uniform definition for MIC has In the fourth edition of the European Guide-
clearly contributed to the confusion regarding this lines for Quality Assurance in Breast Cancer
entity. Screening and Diagnosis published in 2006,
The fifth edition of the AJCC Cancer Staging formed on the major basis of UK guidelines,
Manual published in 1997 recognizes a specific MIC was defined as a “tumor in which the dom-
T substage for MIC, defined as “the extension of inant lesion is in-situ carcinoma (usually exten-
cancer cells beyond the basement membrane into sive high nuclear grade DCIS, rarely other types
the adjacent tissues with no focus more than of DCIS or LCIS) but in which there are one or
0.1 cm in greatest dimension” and formally more, clearly separate, foci of infiltration of non-
reported it as pT1mic. The AJCC Cancer Staging specialized interlobular or interductal fibrous or
Manual further stated that “when there are multi- adipose tissue, none measuring more than 1 mm
ple foci of microinvasion, the size of only the (about 2 high power fields) in maximum diameter.
largest focus is used to classify the microinvasion When there are multiple foci of MIC only the
and that the size of the individual foci should not size of the largest focus is used to classify the
be added together; the presence of multiple foci of microinvasion; the presence of multiple foci of
microinvasion should however be noted and/or microinvasion should however be noted and/or
quantified, as it is with multiple larger invasive quantified.”
carcinomas”. Not all authors accept the definition of MIC
Following the establishment of a National that requires “clearly separate foci of infiltration
Breast Screening Programme in the United of nonspecialized interlobular stroma”: tumor
Kingdom, a Working Group of the Royal Col- cells are, in fact, not known to leap or jump
lege of Pathologists produced in 1990 a docu- or dissociate from the in situ component in order
M
ment on Pathology Reporting in Breast Cancer to invade the stroma, in three dimension micro-
Screening where MIC was defined as “a tumor in invasive tumor foci would be seen to grow
which the dominant lesion is noninvasive but in as solid cords extending from in situ area; further-
which there are one or more foci of infiltration, more the unequivocal influence of the plane
none of which measures more than 1 mm (about of section on the detection of continuity between
2 high power fields) in maximum diameter. the in situ and invasive component render
Small invasive carcinomas without an in situ the requirement “clearly separate foci” invalid;
component are classified as invasive.” In the moreover the “infiltration of nonspecialized
second edition of the Pathology Reporting in interlobular stroma” has been considered a
Breast Cancer Screening published in 1995, it requirement untenable since vascular invasion
is proposed that “only when unequivocal inva- can occur even within the lobular stroma due to
sion is seen outside the specialized lobular the presence of vascular channels both within
stroma, namely into the non-specialized the specialized lobular stroma and immediately
interlobular stroma, should MIC be diagnosed. surrounding the basement membrane that invests
If there is sufficient doubt about the presence of ductules.
invasion, the case should be classified as in situ Considering the difficulty to ascertain exten-
carcinoma.” sion of malignant cells beyond the specialized
In the 2003 edition of WHO Classification of lobular stroma, the current WHO Classification
Tumours of the Breast and Female Genital of Tumours of the Breast (Pinder et al. 2012)
Organs, in spite of the pT1mic category officially reports that MIC is diagnosed when convincing
being recognized by the fifth edition of the AJCC histological features are present, despite malig-
Cancer Staging Manual, MIC was still considered nant cells not being clearly beyond the specialized
an evolving concept as there was no generally lobular stroma.
296 Microinvasive Carcinoma of the Breast
can be particularly harmful because it may lead to needling procedure. In cases with a history
overtreatment. of a prior needling procedure for preoperative
The differential diagnosis of MIC includes diagnosis (FNA, NCB or VANCB), the
pure in situ disease and, conversely, invasive diagnosis of MIC should be made with cau-
breast carcinoma measuring >1 mm; the size of tion because artifactual disruption of the
the focus should be carefully measured with an epithelial-stromal junction of glandular struc-
ocular micrometer to exclude frankly invasive tures involved by in situ carcinoma is not
breast carcinoma. infrequently encountered in subsequent exci-
A variety of patterns in DCIS (▶ Ductal sion biopsy or therapeutic specimen. Granu-
Carcinoma In Situ) and, more rarely, in LCIS lation tissue, old or recent hemorrhage (iron-
(▶ Lobular In Situ Neoplasia) may be mis- laden macrophages, cholesterol crystals,
interpreted as stromal invasion (Schnitt and reactive fibrosis, hemosiderin deposition), tis-
Collins 2013). Lesions and artifacts commonly sue tears, and a degenerative appearance of
mistaken for MIC include: the dislodged tumor cells can help in
distinguishing pseudo-invasion from true
1. DCIS involving lobules (“lobular invasion.
cancerization”) 8. DCIS or LCIS involving benign complex scle-
2. Chronic inflammatory reaction present in rosing lesions such as radial scar (▶ Radial
association with, and obscuring, involved Scar), sclerosing papilloma (▶ Intraductal
ducts and acini Papilloma), ductal adenoma (▶ Ductal Ade-
3. Branching of involved ducts noma), and sclerosing adenosis (▶ Sclerosing
4. Distorsion or entrapment of involved ducts or Adenosis) (Fig. 4, Ductal carcinoma in situ
acini by fibrosis (sometime due to prior nee- involving sclerosing adenosis. (a) H&E.
dling procedure) (b) Calponin immunostain).
5. Crush artifacts
M
6. Cautery effects Additional H&E-stained sections may help to
7. Artifactual displacement of DCIS or LCIS define the nature of the process in problematic
cells into the surrounding stroma or adipose cases. In many cases, IHC for myoepithelial
tissue due to tissue manipulation or a prior cells markers is of greater value for distinguishing
Microinvasive Carcinoma of the Breast, Fig. 4 Ductal the suspicion of microinvasive carcinoma.
carcinoma in situ involving sclerosing adenosis. (b) (Immunohistochemistry): Calponin immunostain dem-
(a) (H&E): Solid nests and glands composed onstrates a myoepithelial cell layer around nests and glands
of neoplastic cells in a desmoplastic stroma raising supporting the diagnosis of ductal carcinoma in situ
300 Mucocele-like Lesion
true MIC from its mimics (see “Microscopy” Low-grade luminal pathway and high grade HER2
section). pathway based on high tumor-infiltrating lymphocytes.
Journal of Clinical Pathology, 69, 890–898.
When there is doubt about the diagnosis of Pinder, S. E., Ellis, I. O., Schnitt, S. J., et al. (2012).
MIC or the suspicious area is no longer seen on Microinvasive carcinoma. In S. R. Lakhani, I. O.
any further sections or IHC, it is recommended Ellis, S. J. Schnitt, et al. (Eds.), WHO classification of
that the case should be diagnosed as in situ lesion tumours of the breast (pp. 96–97). Lyon: International
Agency for Research on Cancer.
with no definite evidence of MIC. Schnitt, S. J., & Collins, L. C. (Eds.). (2013). Biopsy
As reported above, MIC can be not only over- interpretation of the breast (2nd ed.pp. 267–281).
diagnosed but also underdiagnosed. MIC cannot Philadelphia: Lippincott Williams & Wilkins.
be reliably excluded unless all tissue is serially
sectioned, completely embedded, and submitted
for histological examination. This method is
now recommended in clinical guidelines Mucocele-like Lesion
as well in breast screening programs.
However it is well-known that even with a Janina Kulka
large number of paraffin blocks, only a part of 2nd Department of Pathology, Semmelweis
the tissue is examined microscopically, and University, Budapest, Hungary
pathologists can never be absolutely certain that
MIC is really absent.
Serial sections supported by IHC usually Synonyms
provide the best evidence of MIC. Care should
be taken to obtain IHC early in the evaluation Mucocele-like tumor
of suspected MIC, before the sample has been
sectioned excessively, first to confirm MIC and
second to exclude the possibility of larger Definition
invasive foci.
Rosen, who first described these lesions in 1986,
gave the following definition: “mucin containing
cysts lined by flat or low cubical epithelium with
References and Further Reading
or without extravasated mucin” (Rosen 1986).
Amin, M. B., Edge, S. B., Greene, F. L., et al. (Eds.).
(2017). AJCC cancer staging manual (8th ed.
pp. 589–628). New York: Springer. Clinical Features
Bianchi, S., & Vezzosi V. (2008). Microinvasive Carci-
noma of the Breast. Pathology and Oncology Research,
14, 105–111. • Incidence
Cserni, G., Wells, C. A., Kaia, H., et al. (2016). Since the first description by Rosen in 1986,
Consistency in recognizing microinvasion in breast single case reports or relatively small series
carcinomas is improved by immunohistochemistry for have been published. With the introduction of
myoepithelial markers. Virchows Archive, 468,
473–481. screening programs, the incidence increased
Hilson, J. B., Schnitt, S. J., & Collins, L. C. (2009). due to the associated microcalcifications. The
Phenotypic alterations in ductal carcinoma in situ- incidence of MCLLs is very low, ranging from
associated myoepithelial cells: Biologic and diagnostic 0.25% (Jaffer et al. 2011) to <1% (102 MCLL
implications. The American Journal Surgical Pathol-
ogy, 33, 227–232. cases in a population of 13,412 women with
Hoda, S. A. (2014). Ductal carcinoma in situ. biopsy proven benign breast disease) (Meares
In S. A. Hoda, E. Brogi, F. C. Koerner, & P. P. Rosen et al. 2016).
(Eds.), Rosen’s breast pathology (4th ed., pp. 331–411). • Age
Philadelphia: Lippincott Williams & Wilkins.
Morita, M., Yamaguchi, R., Tanaka, M., et al. (2016). Two Since MCLLs are very often associated with
progressive pathways of microinvasive carcinoma: microcalcifications and are discovered mainly
Mucocele-like Lesion 301
on screening mammograms, the mean age atypical ductal hyperplasia (▶ Atypical Ductal
reported is 53–55 years. Hyperplasia) or DCIS (▶ Ductal Carcinoma In
• Sex Situ), even if VACB has been the diagnostic
No MCLLs have been described in male method, further surgical excision seems the
patients to date. right treatment.
• Site • Outcome
No specific site in the breast has been According to the few large series (Jaffer et al.
described. 2011; Meares et al. 2016; Dash et al. 2017), the
• Imaging upgrade rate of MCLLs devoid of atypia diag-
The majority of these lesions are nonpalpable, nosed on VACB specimens is very low. Very
detected on screening mammograms due to close radio-pathological correlation is necessary
associated indetermined, suspicious clustered, and surgical excision can be avoided only if the
coarse or pleomorphic microcalcifications lesion had been removed entirely. In the Not-
(Davies et al. 1995) (Fig. 1). Some MCLLs tingham series (Rakha et al. 2013), 2/54 MCLL
present as a well-circumscribed mass. On ultra- without atypia on core biopsy were upgraded to
sound, cysts, cyst with calcification or a hypo- DCIS in the surgical excision specimen (4%
echoic lesion may be present. upgrade rate). However, altogether in published
• Treatment series until 2013 among all MCLLs with atypi-
The appropriate treatment of MCLL is cal hyperplasia diagnosed on either 14G needle
debated. By definition, these lesions belong to biopsies or VACB the upgrade rate is 21%. In an
the B3 category of core biopsy diagnoses, and Australian series of 117 MCLLs, the upgrade
earlier publications concluded that further sur- rate to DCIS – all low to intermediate grade –
gical excision is necessary. However, in recent was 5% (Dash et al. 2017).
years, studies documented successful removal The question of MCLL as a risk factor was
of MCLLs by vacuum-assisted core biopsy recently analyzed in one of the largest MCLL
M
(VACB), a more conservative approach that series to date (Meares et al. 2016). Thirteen out
appears to be the appropriate treatment in of 102 patients developed in situ or invasive
MCLLs without associated atypical epithelial breast cancer during the follow-up period, the
proliferation. In MCLLs associated with median time to cancer was 11.8 years. It is
Mucocele-like Lesion,
Fig. 1 Specimen
mammogram of a MCLL
detected in screening due to
the presence of coarse,
clustered
microcalcifications
302 Mucocele-like Lesion
noteworthy that almost half of the cancers numerous authors as part of the spectrum of
developed in the contralateral breast. With mucinous breast lesions, representing the first
14.8 years median follow-up, it was found step along the line leading to invasive mucinous
that in women younger than 45, MCLL caused carcinoma through MCLL with atypia and mucin-
only a nonsignificant increase in cancer risk ous DCIS with mucocele-like features. MCLL
compared to the general population. In may have completely bland, flattened, cuboidal,
women above that age, the cancer risk incre- or low columnar epithelium, but some cases are
ment is low – equals that of proliferative breast associated with papillary projections, epithelial
disease. atypia, or frank DCIS (mucinous type, with crib-
riform, solid, papillary, or micropapillary archi-
tecture) and association with mucinous carcinoma
Macroscopy
(▶ Invasive Mucinous Carcinoma) or no special
type invasive breast carcinoma (▶ Invasive Car-
Since MCLLs are usually less than 1 cm, non-
cinoma NST) was also reported. MCLLs associ-
palpable lesions, description of their macroscopic
ated with DCIS (▶ Ductal Carcinoma In Situ)
appearance is missing from most case reports and
may be extensive (Fig. 3, H&E: Low grade
is not emphasized either in larger case series. In
DCIS associated with MCLL). The mucin filling
the original description by Rosen, the gross
the ducts and extravasated in the stroma is PAS,
appearances were described as follows: “irregular,
mucicarmin, and also Alcian blue positive at
gelatinous, shiny area” or “ill-defined mucinous
pH 2.5, suggesting the presence of neutral to
mass,” or “multicystic colloid nodule.”
acidic mucin in these lesions. An important clue
to the diagnosis is the lack of epithelial cell clus-
ters in the extravasated mucin pools. However, the
Microscopy epithelial cells can detach from the cyst walls and
superficially may be reminiscent to the floating
MCLLs of the breast are localized lesions, affect- epithelial cell clusters so characteristic in mucin-
ing the terminal duct-lobular unit. The main fea- ous carcinomas. It is of utmost importance that in
ture is the dilatation and partial rupture of the such cases the epithelial cell clusters be thor-
mucin filled- and cystically dilated ductules and oughly analyzed: those in MCLLs are linear in
as a consequence, mucin leakage into the adjacent most of the cases, as they represent a cohesive
stroma. (Fig. 2). MCLL has been described by fragment of the lining epithelium of a mucin filled
Mucocele-like Lesion,
Fig. 2 Subgross
appearance of a MCLL:
cystic, distended ductules in
a TDLU. Arrows point to
the extravasated mucin
Mucocele-like Lesion 303
Mucocele-like Lesion,
Fig. 3 H&E: Low-grade
DCIS associated
with MCLL
and/or ruptured cyst. In some cases, a complete showed mainly luminal/apical or luminal/apical
arborizing papillary structure may detach from the and cytoplasmic expression. MUC2 was only
cyst wall and be present in the extravasated expressed in MCLLs associated with atypia or
mucin. Lack of cellular atypia and presence of DCIS and was positive in almost every mucinous
M
myoepithelial cells are the key for the correct carcinoma studied.
diagnosis (Tan et al. 2008). A common feature
in MCLLs leading to their identification on
screening mammograms are (indeterminate – or Molecular Features
suspicious, coarse, and polymorphic) micro-
calcifications. Calcifications are common in the To date, no molecular studies have been
mucin pools in MCLL (Fig. 4, H&E). In some performed on MCLL series.
cases, a heavy stromal lymphoid cell infiltrate is
also present around the areas of extravasated
mucin (Fig. 5). Differential Diagnosis
Mucocele-like Lesion,
Fig. 4 H&E: MCLL with
microcalcification
Mucocele-like Lesion,
Fig. 5 H&E: Foci of
lymphoid cell infiltrate in
the vicinity of acellular
mucin pools
mucinous carcinomas’ cell clusters are more MCLLs. Cytology diagnosis of MCLL requires
abundant and are formed by uniformly atypical great caution, since the presence of mucinous
cells. When a MCLL is associated with atypia or material on the smears may lead to erroneous
DCIS, the correct diagnosis requires even greater conclusion. It is also important to keep in mind
caution. that mucinous carcinomas occur mostly in older
The diagnostic dilemma is even more diffi- women than do MCLLs. The difficulty is even
cult in core biopsies, when only fragments of the greater when MCLL is associated with atypia or
lesion can be evaluated. A helpful feature is the frank DCIS. In such cases, fragments of the
presence of totally acellular mucin in cases of atypical epithelium may detach and float in the
MCLL and lack of atypia of the epithelial cells, extravasated mucin. The linear appearance of
if present. Multiple levels may help to confirm these epithelial fragments may help not to over-
the lack of epithelium in the mucin pools. In diagnose the case as mucinous carcinoma
mucinous carcinoma the areas of extracellular (Fig. 3). Nevertheless, in some cases, it is
mucin often contain small vessels embedded in impossible to unequivocally differentiate a
thin fibrous bands, which is not a feature in MCLL from a mucinous carcinoma on core
Mucoepidermoid Carcinoma of the Breast 305
Mucocele-like Lesion, Table 1 Differential diagnosis of MCLL and mucinous carcinoma (Based on Torous et al. 2017)
MCLL Mucinous carcinoma
Clinical presentation Most often incidentally detected on Usually presents as a mass (palpable or
(screening) mammography due to radiologically detected)
microcalcifications
Microscopy
Extracellular mucin Acellular; may contain detached linear Tumor cell clusters floating in the mucin;
fragments of epithelium evidence of neovascularization
Epithelium Most often low cuboidal to columnar Low to intermediate grade nuclear atypia of
without atypia, proliferative changes and the tumor cells that form small clusters in
papillarization may be present; usual type- the mucin pools; myoepithelial cells are not
or atypical ductal hyperplasia and DCIS seen
may be associated; myoepithelial cells are
present
Immunohistochemistry Myoepithelial markers stain the No myoepithelial cells can be identified
myoepithelial cells, but staining may be
absent in associated ADH or DCIS
biopsy and these doubts have to be stated in the & Visscher, D. W. (2016). Mucocele-like lesions of the
report (Table 1). breast: A cliniical outcome and histologic analysis of
102 cases. Human Pathology, 49, 33–38.
Cystic hypersecretory breast lesions may Rakha, E. A., Shaaban, A. M., Asma Haider, S., Jankins, J.,
superficially resemble to MCLL at low power. Menon, S., Johnson, C., Yamaguchi, R., Murphy, A.,
However, in cystic hypersecretory breast lesions Liston, J., Cornford, E., Hamilton, L., James, J., Ellis,
(hyperplasias and DCIS), the secretion that fills I. O., & Lee, A. H. S. (2013). Histopathology, 62,
894–898.
the cystic spaces is deeply eosinophilic and Rosen, P. P. (1986). Mucocele-like tumors of the
resembles thyroid colloid. breast. American Journal of Surgical Pathology, 10, M
An extremely rare breast tumor, mucinous 464–469.
cystadenocarcinoma may also possess extrava- Tan, P. H., Tse, G. M. K., & By, B. H. (2008). Mucinous
breast lesions: Diagnostic challenges. Journal of Clin-
sated mucin pools, but myoepithelial cells are ical Pathology, 6, 11–19.
not present, the epithelial cells are tall columnar Torous, V. F., Schnitt, S. J., & Collins, L. C. (2017). Benign
cells and contain intracellular mucin, both fea- breast lesions that mimic malignancy. Pathology, 49,
tures lacking in MCLL. 181–196.
Low-grade MEC are characterized by mixture seen. In situ component having the same cell
of basaloid, intermediate, epidermoid, and population can be present, associated with inva-
mucous-secreting cells arranged mainly in cystic sive MEC.
and solid nests (Figs. 1 and 2 H&E). Basaloid High-grade MEC share the same cell features
cells are usually located at the periphery of the of low-grade MEC but show a higher degree of
neoplastic nodules, while the center of the nod- cellular atypia and higher mitotic count; necrosis
ule is mainly composed of intermediate and epi- can be encountered.
dermoid cells (Fig. 3 H&E). True keratinization One case only of MEC of intermediate malig-
is not a feature of MEC. Epidermoid and inter- nancy grade was reported by Di Tommaso et al.
mediate cells have wide and granular eosino- (2004). The same case showed solid architectural
philic, less frequently clear cytoplasm that pattern with prevalence of intermediate and epi-
can be enriched in mitochondria. Nuclei are dermoid cells and focal presence of mucous-
centrally located. Mucous cells usually line the secreting cells.
cystic areas (Fig. 3 H&E). Alcian Blue pH 2.5
and PAS after diastase digestion can evidence
the mucous-secreting cells. On rare occasions, Immunophenotype
mucous-secreting cells have a signet ring
appearance (Fig. 4a and b H&E). In low-grade Immunohistochemistry can be helpful in the
MEC, atypia is mild, atypical mitotic diagnosis of breast MEC, and each single cell
figures are extremely rare, and no necrosis is type has its peculiar immunohistochemical
pattern.
Basaloid, intermediate, and epidermoid cells
are mainly positive with high-molecular-weight
cytokeratins (CK) as CK 14 and with p63. These
same cells can be positive with anti-mitochondrial
M
antibody but to a lesser intensity than observed in
salivary gland counterpart.
On the contrary mucous-secreting cells are
positive with low-molecular-weight CK as CK7,
epithelial membrane antigen (EMA), and MUC 1,
MUC 5 AC, and MUC 6.
Mucoepidermoid Carcinoma of the Breast,
Estrogen, progesterone receptors, and HER2
Fig. 3 Low-grade MEC with cystic features; mucous-
secreting cells line the cystic component. Epidermoid are consistently negative in all the reported
cells, devoid of true keratinization, are present (H&E) cases.
Mucoepidermoid Carcinoma of the Breast, Fig. 4 (a and b) Low-grade (a) and intermediate-grade (b) MEC: single
mucous-secreting cells are indicated by arrows (H&E)
308 Myoepithelial Carcinoma of the Breast
Molecular Features Tjalma, W. A., Verslegers, I. O., De Loecker, P. A., & Van
Marck, E. A. (2002). Low and high grade
mucoepidermoid carcinomas of the breast. European
Salivary gland MEC is characterized by the Journal of Gynaecological Oncology, 23, 423–425.
MECT-MAML2 translocation. Similarly, Camelo-
Piragua et al. (2009) found the 11q21 deletion at
the site of MAML2 gene.
Myoepithelial Carcinoma of
the Breast
Differential Diagnosis
Horst Bürger
Low-grade MEC diagnosis can be difficult when Institute of Pathology Paderborn/Höxter,
clear cells or mitochondrion-rich cells are numer- Paderborn, Germany
ous, raising the suspicion of clear cell carcinoma
or of oncocytic carcinoma (▶ Invasive Oncocytic
Carcinoma). The differential diagnosis is mainly Synonyms
based on the detection of mucous-secreting cells
intermingled with the neoplastic population. Malignant myoepithelioma; Metaplastic carci-
Diagnosis is more difficult in high-grade MEC. noma; Spindle cell carcinoma
Differential diagnosis should be carried out
mainly with carcinomas showing adenosquamous Definition
features (▶ Low-Grade Adenosquamous Carci-
noma). These latter tumors usually show true A malignant lesion composed of myoepithelial
keratinization features that are usually lacking in cells with increased mitotic activity and expres-
both low- and high-grade MEC. sion of myoepithelial markers, such as high
molecular weight cytokeratins, p63, smooth mus-
cle actin, and others.
References
Basbug, M., Akbulut, S., Arikanoglu, Z., Sogutcu, N., Clinical Features
Firat, U., & Kucukoner, M. (2011). Mucoepidermoid
carcinoma in a breast affected by burn scars: Compre-
hensive literature review and case report. Breast Care • Incidence
(Basel), 6, 293–297. Myoepithelial carcinomas of the breast repre-
Camelo-Piragua, S. I., Habib, C., Kanumuri, P., Lago, sent an extremely rare and poorly understood
C. E., Mason, H. S., & Otis, C. N. (2009).
entity of invasive breast cancer. The exact inci-
Mucoepidermoid carcinoma of the breast shares cyto-
genetic abnormality with mucoepidermoid carcinoma dence of these tumors is unknown, partially due
of the salivary gland: A case report with molecular to the disputed definition. According to the
analysis and review of the literature. Human Pathology, actual WHO classification, myoepithelial carci-
40, 887–892.
nomas are a subgroup of metaplastic carcinoma
Di Tommaso, L., Foschini, M. P., Ragazzini, T., Magrini,
E., Fornelli, A., Ellis, I. O., & Eusebi, V. (2004). (Reis-Filho et al. 2012). Under these assump-
Mucoepidermoid carcinoma of the breast. Virchows tions, the incidence is probably significantly
Archives, 444, 13–19. lower than <1%.
Foschini, M. P., & Krausz, T. (2010). Salivary gland-type
• Age
tumors of the breast: A spectrum of benign and malig-
nant tumors including “triple negative carcinomas” of Myoepithelial carcinomas have been seen in all
low malignant potential. Seminar in Diagnostic Pathol- age groups from 25 to 81 years.
ogy, 27, 77–90. • Sex
Foschini, M. P., Morandi, L., Asioli, S., Giove, G.,
Rare cases of myoepithelial carcinoma of the
Corradini, A. G., & Eusebi, V. (2017). The morpholog-
ical spectrum of salivary gland type tumours of the breast have been described in women. The
breast. Pathology, 49, 215–227. incidence in male patients is not known.
Myoepithelial Carcinoma of the Breast 309
• Site
A predominant site within the breast has not
been described.
• Treatment
Recommendations concerning the treatment of
these lesions are differing. This is mainly due
to the unsharp definition of this tumor entity.
Since some regard atypical adenomyoe-
pithelioma as one end of a morphological spec-
trum and myoepithelial carcinoma as the other
end, the treatment of these former tumors sig-
nificantly differs from myoepithelial carci-
noma fulfilling the criteria of ▶ metaplastic
carcinoma. Myoepithelial Carcinoma of the Breast,
Fig. 1 Myoepithelial carcinoma of the breast (H&E)
Whereas atypical adenomyoepithelioma (200)
does not fulfil the criteria to justify a treatment
as invasive carcinoma, myoepithelial carcino-
mas are treated according to the respective supports the diagnosis of myoepithelial carci-
guidelines for metaplastic, hormone receptor- noma (Tan and Ellis 2013). Extensive sampling
negative cancers. of the tumor is required since in most cases an
• Outcome adenomyoepithelial component may give hints to
Reliable data concerning the outcome are rare. the myoepithelial differentiation of this tumor.
Limited follow-up data report an increased In-situ carcinoma can also be observed in a large
local recurrence rate and a rather low incidence subset of tumors, indicating the epithelial origin of
of regional lymph node metastasis. the malignant lesion.
M
Macroscopy
Immunophenotype
Myoepithelial carcinomas might present as round
The immunophenotype reflects the expression
to oval, well-demarcated nodules as well as
patterns observed in normal myoepithelial cells.
tumors with irregular borders and overt infiltrating
This includes the expression of keratins, mainly
growth pattern. Hemorrhage, cystic degenera-
high molecular weight cytokeratins (Ck5, 14, 17),
tions, and calcifications can be seen. In most
smooth muscle actin, vimentin, caldesmon,
cases, the tumors appear as white-grey tumors
calponin, S-100, CD10, GFAP, D2–40 (examples
with largely varying size (up to >20 cm diameter).
given in Fig. 2a–c) and the lack of expression of
estrogen receptor (ER), progesterone receptor (PR),
and negativity for HER2 (Schmitt et al. 2012).
Microscopy
Myoepithelial Carcinoma of the Breast, Fig. 2 (a) (c) Myoepithelial carcinoma of the breast with strong and
Myoepithelial carcinoma of the breast with focal positivity homogeneous positivity for D2–40 as a marker of
for p63 (200). (b) Myoepithelial carcinoma of the breast myoepithelial differentiation (200)
with focal expression of cytokeratin (200).
In general, these tumors reveal a high number exact classification of these tumors (Weidner and
of genetic alterations, indicating a high genetic Dabbs 2012; Schnitt and Collins 2009; Fine and
instability. Major hallmarks, however, only seen Kurdek 2016).
in rather small proportions are p53-alterations, Therefore, myoepithelial carcinomas should be
EGFR amplification, lack of HER2 expression, separated from all kind of spindle cell lesions of
and negativity for ER and PR. the breast, mainly spindle cell carcinomas and all
However, one series described a lower rate of forms of ▶ metaplastic carcinoma.
genetic alterations per case compared to invasive Since the morphological and immunohisto-
breast cancer no-special type. This discrepancy chemical differential diagnosis between these
seems to be a reflection of the controversial defi- tumors might be arbitrary, some authors regard
nitions of myoepithelial carcinoma. the distinction between these tumors rather as a
problem of semantics.
At the current state, it is not clear if this sub-
Differential Diagnosis grouping is supported by other hallmarks, e.g.,
molecular findings. Other authors argue that due
The differential diagnosis of these tumors is, to a very close morphological relationship
despite a clear immunophenotype, complicated between myoepithelial carcinomas and
and reflects the current discussion concerning the adenomyoepithelial tumors, these tumors might
Myoepithelial Carcinoma of the Breast 311
be seen as the extreme end of a morphological Reis-Filho, J. S., Lakhani, S. R., Gobbi, H., & Sneige,
continuum of tumors with a predominant N. (2012). Metaplastic carcinoma. In S. R. Lakhani,
I. O. Ellis, S. J. Schnitt, P. H. Tan, & M. J. van de Vijver
myoepithelial differentiation. (Eds.), WHO classification of tumours of the breast
Malignant ▶ phyllodes tumor can be a differ- (pp. 119–124). IACR: Lyon.
ential diagnosis. However, the lack of epithelial Schmitt, F., Tan, P. H., Dabbs, D., & Jones, L. (2012).
glands, expression of high molecular weight Myoepithelial and epithelial–myoepithelial lesions.
In S. R. Lakhani, I. O. Ellis, S. J. Schnitt, P. H.
cytokeratins, p63, and other markers of Tan, & M. J. van de Vijver (Eds.), WHO classifi-
myoepithelial differentiation allows a clear dis- cation of tumours of the breast (pp. 119–124).
tinction from myoepithelial carcinomas in the IACR: Lyon.
vast majority of the cases. Schnitt, S. J., & Collins, L. C. (2009). Spindle cell lesions.
In S. J. Schnitt (Ed.), Biopsy interpretation of the breast
Fibromatosis, a spindle cell proliferation (pp. 323–343). Philadelphia: Lippincott Williams &
rarely seen in the breast may also be considered. Wilkins.
Lack of p63 and high molecular weight Tan, P. H., & Ellis, I. O. (2013). Myoepithelial and
cytokeratin positivity in these lesions helps to epithelial–myoepithelial, mesenchymal and
fibroepithelial breast lesions: Updates from the WHO
make distinction from myoepthelial carcinoma. Classification of Tumours of the Breast 2012. Journal
of Clinical Pathology, 66, 465–470.
Tavassoli, F. A., & Eusebi, V. (2009). Myoepithelial
lesions. In F. A. Tavassoli & V. Eusebi (Eds.), Tumors
References and Further Reading of the mammary gland (pp. 249–262). Washington,
DC: Armed Forces Institute of Pathology.
Fine, M. A., & Kurdek, L. A. (2016). An approach to the Weidner, N., & Dabbs, J. D. (2012). Myoepithelial lesions
diagnosis of spindle cell lesions of the breast. Histopa- of the breast. In D. J. Dabbs (Ed.), Breast pathology
thology, 68, 33–44. (pp. 307–323). Philadelphia: Saunders.
M
P
Paget Disease of the Nipple, Fig. 1 (a) Paget disease of highlighted by CK 7 positivity (CK7 IHC reaction). (d)
the nipple with underlying DCIS in large ducts (H&E). Paget cells are GATA3 positive (GATA3 IHC reaction). (e)
(b) Paget cells in the epidermis forming nests (H&E). In this case, Paget cells are strongly HER2 positive (HER2
(c) Massive intraepidermal spread of Paget cells is IHC reaction)
Paget Disease of the Nipple, Fig. 2 Melanin pigment is Paget Disease of the Nipple, Fig. 3 Acantholysis in a
present in the cytoplasm of a Paget cell (H&E) case of PD (H&E)
Paget Disease of the Nipple, Table 1 Immunohistochemical characteristics of the main entities in the differential
diagnosis of Paget disease
Paget In situ squamous cell carcinoma/Bowen Malignant
disease Toker cells disease melanoma
Cytokeratin 7 + +
Cam 5.2 + +
EMA + +
CEA +
GATA3 + No reports
available
Cytokeratin +
5/6
p63 +
S100 + or + or +
Melan-A +
HMB45 +
SOX10 +
ER/PR Usually May be + or
HER2 Often +
Mucin May be + Not present Not present Not present
Major source: Jacobs, T.W. Clear Cells of Toker in the Nipple Epidermis, Case presentation, USCAP 2010 Annual
Meeting
KMT2C (MLL3, 39%) and ARID2 (22%), with glandular structures are absent. As opposed to
additional recurrent somatic mutations, such as PD, high molecular weight cytokeratins are
CDCC168 (34%), FSIP2 (29%), CASP8AP2 expressed by the atypical cells. Paget cells may
(29%), and BIRC6 (24%). Interestingly, in paired take up melanin pigment released by epidermal
mammary PD and underlying breast carcinoma cells or melanocytes (referred to as pigmented PD
samples, in 3 cases out of 20, distinct gene muta- in the literature), mimicking malignant mela-
tions were detected, suggesting independent noma: both PD and melanoma in situ may show
oncogenic events in the pathogenesis of these an intraepidermal population of epithelioid cells
lesions (Zhang et al. 2019). Mai et al. (2018) individually or arranged in small nests and the
described dysregulation of FOXA1 and the presence of atypical epithelioid cells in the supra-
nuclear receptor signaling pathway in both mam- basal layer. Both lesions may show melanin pig-
mary and extramammary PD. ment not only within tumor cells’ cytoplasm but
also in dermal melanophages and within dendritic
melanocytes between tumor cells (Solinas et al.
Differential Diagnosis 2018). HER2 staining if positive is particularly
useful (especially in the differential diagnosis
Differential diagnosis of PD includes those with melanoma, pagetoid Spitz nevus, in situ
lesions characterized by so-called intraepidermal squamous cell carcinoma/pagetoid Bowen disease
“pagetoid spread” of atypical cells: pagetoid and Toker cells (▶ Toker Cells of the Nipple),
Bowen’s disease, malignant melanoma, pagetoid which are all negative). The distinction between
Spitz nevus, Merkel cell carcinoma, mycosis LCIS involving the nipple skin in the form of PD
fungoides, Langerhans histiocytosis, and ductal versus Toker cells may be difficult since both have
eccrine carcinoma (Sandoval-Leon et al. 2013). bland appearing cells. However, Toker cells are
Paget cells are not to be mistaken with Toker cells E-cadherin positive as opposed to LCIS cells. The
(▶ Toker Cells of the Nipple). In Bowen’s disease, neoplastic T cells in mycosis fungoides have large
intracellular mucin, signet ring cells, and convoluted (cerebriform) nuclei, pale cytoplasm,
Paget Disease of the Nipple 317
and show CD3 positivity. Similar to occasional Mai, R., Zhou, S., Zhou, S., Zhong, W., Hong, L., Wang,
cases of PD, a clear halo is often seen around Y., Lu, S., Pan, J., Huang, Y., Su, M., Crawford, R.,
Zhou, Y., & Zhang, G. (2018). Transcriptome analyses
single neoplastic cells. Furthermore, Pautrier reveal FOXA1 dysregulation in mammary and extra-
microabscesses may resemble glandular struc- mammary Paget’s disease. Human Pathology, 77,
tures in PD. In various forms of histiocytoses, 152–158.
the nuclei of the atypical cells are large, bean Meissner, K., Riviere, A., Haupt, G., & Loning, T. (1990).
Study of neu-protein expression in mammary Paget’s
shaped, and the cytoplasm stains pale. If in disease with and without underlying breast carcinoma
doubt, CD1a, HLA-DR, and S100 immunohisto- and in extramammary Paget’s disease. American Jour-
chemical positivity of the atypical histiocytes is nal of Pathology, 137, 1305–1309.
helpful (Lloyd and Flanagan 2000). Ductal Ozerdem, U., McNiff, J. M., & Tavassoli, F. A. (2016).
Cytokeratin 7-negative mammary Paget’s disease:
eccrine carcinoma arising in the nipple-areola A diagnostic pitfall. Pathology Research and Practice,
region may mimic PD both clinically and histo- 212, 279–281. https://doi.org/10.1016/j.prp.2016.01.004.
logically in a biopsy specimen (Park et al. 2001). Epub 22 Jan 2016.
As opposed to PD, eccrine adnexal tumors are p63 Park, B. W., Kim, S. I., Lee, K. S., & Yang, W. I. (2001).
Ductal eccrine carcinoma presenting as a Paget’s
positive. Merkel cell carcinomas may also have a disease-like lesion of the breast. The Breast Journal,
pagetoid intraepidermal component. The tumor 7, 358–362.
cells, however, are positive for neuroendocrine Rayne, S. C., & Santa Cruz, D. J. (1992). Anaplastic
immunohistochemical markers unlike Paget cells Paget’s disease. American Journal of Surgical Pathol-
ogy, 16, 1085–1091.
(Stanoszek et al. 2017). Satellite invasive breast Sahoo, S., Green, I., & Rosen, P. P. (2002). Bilateral Paget
carcinoma focus involving the areolar skin with disease of the nipple associated with lobular carcinoma
exulceration (pT4b) is a different entity and in situ. Archives of Pathology and Laboratory Medi-
should not be misinterpreted as PD. cine, 126, 90–92.
Sanders, M. A. G. Paget disease. PathologyOutlines.com
website. http://www.pathologyoutlines.com/topic/breast
malignantpaget.html. Accessed 13 Jan 2019.
References and Further Reading Sandoval-Leon, A. C., Drews-Elger, K., Gomez-
Fernandez, C. R., Yepes, M. M., & Lippman, M. E.
Adams, S. J., & Kanthan, R. (2016). Paget’s disease of the (2013). Paget’s disease of the nipple. Breast Cancer
male breast in the 21st century: A systematic review. Research and Treatment, 141, 1–12.
The Breast, 29, 14–23. Shousha, S. (2007). Glandular Paget’s disease of the nip-
Anderson, J. M., Ariga, R., Govil, H., Bloom, K. J., ple. Histopathology, 50, 812–814.
Francescatti, D., Reddy, V. B., et al. (2003). Assess- Shousha, S., Eusebi, V., & Lester, S. (2012). Paget disease P
ment of Her-2/Neu status by immunohistochemistry of the nipple. In S. R. Lakhani, I. O. Ellis, S. J. Schnitt,
and fluorescence in situ hybridization in mammary P. H. Tan, & M. J. van de Vijver (Eds.), WHO classifi-
Paget disease and underlying carcinoma. Applied cation of tumours of the breast (4th ed., pp. 152–153).
Immunohistochemistry and Molecular Morphology, Lyon: IARC.
11, 120–124. Solinas, A., Mahar, A., Cooper, W. A., Thompson, J. F.,
Barnes, P. J., Dumont, R. J., & Higgins, H. G. (2007). Spillane, A. J., & Scolyer, R. A. (2018). Pigmented
Acinar pattern of mammary Paget’s disease: A case Paget’s disease of the nipple mistaken for melanoma
report. The Breast Journal, 13, 520–526. in situ: A diagnostic pitfall for the unwary. Pathology,
Bianco, M. K., & Vasef, M. A. (2006). HER-2 gene ampli- 50, 364–367.
fication in Paget disease of the nipple and extra- Stanoszek, L. M., Wang, G. Y., & Harms, P. W. (2017).
mammary site: A chromogenic in situ hybridization Archives of Pathology and Laboratory Medicine, 141,
study. Diagnostic Molecular Pathology, 15, 131–135. 1490–1502.
Jacobs, T. W. Clear cells of Toker in the Nipple Epidermis. Wachter, D. L., Wachter, P. W., Fasching, P. A.,
USCAP 2010 Annual Meeting, Specialty Conference, Beckmann, M. W., Hack, C. C., Riener, M. O.,
Breast Pathology, Case 2. http://uscapknowledgehub. Hartmann, A., & Strehl, J. D. (2019). Characterization
org/newindex.htm?99th/specbreah2.htm of molecular subtypes of Paget disease of the breast
Lee, H. W., Kim, T. E., Cho, S. Y., Kim, S. W., Kil, W. H., using immunohistochemistry and in situ hybridiza-
Lee, J. E., Nam, S. J., & Cho, E. Y. (2014). Invasive tion. Archives of Pathology and Laboratory Medicine,
Paget disease of the breast: 20 years of experience at a 143, 206–211.
single institution. Human Pathology, 45, 2480–2487. Wolber, R. A., Dupuis, B. A., & Wick, M. R. (1991).
Lloyd, J., & Flanagan, A. M. (2000). Mammary and extra- Expression of c-erbB-2 oncoprotein in mammary and
mammary Paget’s disease. Journal of Clinical Pathol- extramammary Paget’s disease. American Journal of
ogy, 53, 742–749. Clinical Pathology, 96, 243–247.
318 Phyllodes Tumor
Wong, S. M., Freedman, R. A., Stamell, E., Sagara, Y., its malignant potential became known when a
Brock, J. E., Desantis, S. D., & Golshan, M. (2015). metastatic case was reported by Lee and Pack.
Modern trends in the surgical management of Paget’s
disease. Annals of Surgical Oncology, 22, 3308–3316. Phyllodes tumors are classified as benign, border-
Zhang, G., Zhou, S., Zhong, W., Hong, L., Wang, Y., Lu, line, or malignant (Ellis et al. 2013).
S., Pan, J., Huang, Y., Su, M., Crawford, R., Zhou, Y., Usage of the original term of cystosarcoma
& Mai, R. (2019). Whole-exome sequencing reveals phyllodes is discouraged given that the majority
frequent mutations in chromatin remodeling genes in
mammary and Extramammary Paget’s diseases. Jour- of these neoplasms are benign. The preferred term
nal of Investigative Dermatology, 139, 789–795. as recommended by the World Health Organiza-
tion in 1981 is phyllodes tumor, with the benign,
borderline, or malignant prefix used as appropri-
ate (Tan et al. 2016).
Phyllodes Tumor
Clinical Features
Mark O’Loughlin1 and Grace Callagy1,2
1
Discipline of Pathology, NUI Galway, Galway,
• Incidence
Ireland
2 Phyllodes tumors are uncommon, representing
NUI Galway Clinical Science Institute, Galway,
approximately 1% of all breast neoplasms at
Ireland
most (0.2–1%) and only 2.5% of fibroepithelial
breast lesions. The exact incidence of tumors
that fall into the benign, borderline, and malig-
Synonyms
nant categories is uncertain. This is because the
rates reported for each category vary consider-
Benign phyllodes tumor; Benign phylloides
ably between studies due to the use of different
tumor; Borderline phyllodes tumor; Borderline
classification systems and to the well-
phylloides tumor; Cystosarcoma phyllodes;
recognized difficulty in distinguishing between
Cystosarcoma phylloides; Malignant phyllodes
tumors in the different categories. It is esti-
tumor; Malignant phylloides tumor; Phyllodes
mated that benign phyllodes tumors are likely
tumor; Phylloides tumor
to account for at least 70%, malignant tumors
for 10–20%, and borderline tumors for the
remainder of all these cases (Ellis et al. 2013;
Definition Tan et al. 2012, 2016).
Malignant phyllodes tumors are more com-
Phyllodes tumors are true fibroepithelial neo- mon among Hispanic populations than other
plasms characterized by a biphasic growth pattern ethnic groups, particularly those born in Cen-
which comprises an epithelial and a stromal com- tral and South America. Phyllodes tumors are
ponent and resembles an intracanalicular more common in nulliparous women (Tan
fibroadenoma. The epithelial component is dou- et al. 2012).
ble layered and arranged in clefts and is • Age
surrounded by the hypercellular stromal compo- These tumors are more common in perimeno-
nent. Together these give the appearance of leaf- pausal and postmenopausal women. The aver-
like structures. This appearance is the origin of the age age at presentation is usually 40–50 years,
name cystosarcoma phyllodes when they were with the median age being 45 years. Phyllodes
first described in 1838 by Müller (phyllos being tumors tend to develop a decade later than the
the Greek word for leaf, cysto- pertaining to the usual age for fibroadenomas and are relatively
cystic spaces identified, and sarcoma pertaining to rare below the age of 25. However, there have
the sarcomatous stroma). The lesion was initially been reports with ages ranging from younger
described as benign, and it was not until 1931 that than 10 years to older than 70. In Asian
Phyllodes Tumor 319
populations, age at diagnosis is lower than for retrospective reviews is contradictory with
other populations, approximately 25–30 years some recording no relationship between mar-
(Tan et al. 2012). gin width and recurrence rates, whereas others
• Sex found that recurrence increased with margins
It predominantly affects women though there <1 cm. Interpretation is compounded by the
have been a small number of cases described in fact that the rate of recurrence for
men, which is slightly more often found in fibroadenomas, the main differential diagnosis
those with preexisting gynecomastia (Tan for a benign phyllodes tumor, is in the order of
et al. 2012). 15%. Moreover, not all benign phyllodes
• Site tumors with a positive margin recur. In the
These neoplasms affect breast tissue and rarely absence of definitive evidence, many would
involve the skin, though they can stretch the consider the presence of tumor cells touching
skin if grown to a large size, giving a bluish ink or within 1 mm of the margin as a positive
discoloration and dilated veins. This margin. A benign phyllodes tumor with a pos-
stretching, nipple retraction, and potential itive margin can be managed conservatively;
pain can all occur with both benign and malig- however, most would advocate clear margins
nant phyllodes tumors. Ulceration onto the for recurrent, borderline, and malignant phyl-
skin surface is very rare and is far more likely lodes tumors (Tan et al. 2016).
to be observed with malignant phyllodes than Mastectomy may be considered in some
with benign or borderline tumors, although cases depending on the size of the tumor and
very large benign tumors can ulcerate the over- the malignancy status. Axillary node sampling
lying skin. Phyllodes tumors are typically uni- is not recommended given the low rate of nodal
lateral, with multifocal or bilateral lesions metastasis.
being regarded as rare (Ellis et al. 2013). The role of adjuvant radiotherapy in the treat-
• Clinical Presentation ment for malignant tumors is unclear. There are
The presentation is classically a clinically pal- some reports of reduced rates of local recurrence
pable mass in the breast parenchyma with an but no effect on overall survival. There is no
average size of 4–5 cm. This lump is usually, evidence to support the use of cytotoxic or
but not always, painless, well defined, and targeted chemotherapy (Tan et al. 2016).
mobile. It is not possible to distinguish a phyl- • Outcome P
lodes tumor from a fibroadenoma or other Phyllodes tumors of all grades have the potential
benign breast lesion at clinical presentation, for recurrence, although recurrence is rare when
although a phyllodes tumor is often rapidly the tumor is completely excised. Recurrence is
growing. Smaller tumors can present on rou- more common for malignant tumors (23–30%)
tine mammography as well-defined lesions or than for borderline (14–25%) or benign
as hypoechoic masses with hyperechoic stria- (10–17%) tumors. In the vast majority of cases,
tions on ultrasound imaging (Ellis et al. 2013). recurrence is local and usually occurs within
• Treatment 2–3 years of initial diagnosis. Recurrent disease
The currently accepted gold standard of treat- is typically of the same grade as the initial tumor,
ment for phyllodes tumors is surgical resection though progression to a higher-, or rarely a
with a margin of normal tissue. Unlike the case lower-, grade tumor can occur (Tan et al. 2012).
for invasive carcinoma, there is no consensus Metastatic rates vary from 1% to 22%, and
on what constitutes an adequate margin thick- metastasis is associated with a poor prognosis
ness, especially for benign tumors. Most data with death from disease most commonly ensu-
on margin status comes from retrospective ing within 5–8 years after diagnosis. Virtually
studies. Some advocate a margin of 1 cm all metastases occur following a diagnosis of
even for benign tumors, but robust evidence malignant phyllodes tumor. There are reports
for this is lacking. The data on recurrence from of metastases occurring following a diagnosis
320 Phyllodes Tumor
of borderline and, less commonly, benign phyl- more likely to be malignant. Phyllodes tumors in
lodes tumor, but these are considered excep- general have a faster growth rate than
tionally rare occurrences. Metastatic sites fibroadenomas (Ellis et al. 2013; Tan et al. 2012).
include the lungs, pleura, bone, and very rarely Phyllodes tumors are round to oval in shape,
the central nervous system. The metastases are with a well-circumscribed border and a firm tex-
frequently composed of the sarcomatous stro- ture. They vary between lobulated contours to hav-
mal component of the tumor without epithelial ing a multinodular surface. Benign phyllodes
elements. Spread to lymph nodes is rare, and tumors can be grossly indistinguishable from
hence nodal sampling is not routinely fibroadenomas. The cut surface shows a tan or
recommended in the management of phyllodes pink to gray-white whorled pattern that may have
tumors. In extremely rare cases, metabolic dis- a fleshy or mucoid consistency, though they are
turbances caused by paraneoplastic syndrome more frequently firm. They typically have cleft
can occur whereby phyllodes tumors secrete spaces, and larger tumors can have areas of cystic
insulin-like growth factor II, causing severe degeneration, hemorrhage, or necrosis. These fea-
(or occasionally fatal) hypoglycemia (Ellis tures are more common in malignant compared to
et al. 2013; Tan et al. 2012). benign phyllodes tumors. The borders of benign
• Factors Influencing Recurrence and tumors are usually well defined, whereas those of a
Behavior malignant phyllodes tumor may be grossly infiltra-
Margin status is one of the most important tive. Periductal stromal hyperplasia can occur adja-
factors determining the risk for recurrence. In cent to the phyllodes tumor and can make
many instances, recurrence is due to inade- assessment of the border difficult. In some
quate resection, and a further wide local exci- instances, a “pseudocapsule” composed of com-
sion can be curative. Conversely, positive pressed normal breast parenchyma can envelop
margins do not result in inevitable recurrence. the tumor (Ellis et al. 2013).
A review of factors associated with an
increased risk of adverse outcome identified
margin status as being the most important Microscopy
with atypia, mitoses, stromal overgrowth also
being important, but the evidence for this is Phyllodes tumors are derived from the specialized
weak. As yet, there are no validated prognostic lobular stroma of the breast, and the biphasic
features to identify those patients who will growth pattern is the characteristic feature of this
develop metastatic disease (Ellis et al. 2013). tumor. Phyllodes tumors are composed of a dou-
ble layer of glandular epithelium with
myoepithelial and luminal epithelial cells that
Macroscopy line cleft-like spaces. The clefts create the appear-
ance of large leaflike structures in which the
Tumor size is extremely variable with case reports stroma is cellular. The stroma is the dominant
documenting cases from 1 cm in diameter up to component of the tumor; the epithelium appears
greater than 40 cm. The average size at diagnosis benign. Tumors are classified as benign, border-
is usually 4 cm or 5 cm with the size range most line, or malignant based on the assessment of a
commonly being between 2 and 10 cm. Mammo- number of interrelated histological features, as
graphic screening has seen an increase in detec- outlined in the WHO Classification of Tumors of
tion of smaller tumors that are between 2 and the Breast (Tan et al. 2012).
3 cm. It is not possible to distinguish between
the different grades of phyllodes tumor by size Benign Phyllodes Tumor
because there is significant overlap in the sizes of These lesions characteristically have pushing,
benign, borderline, and malignant tumors. How- lobulated borders that are well circumscribed. In
ever, some reports suggest that larger tumors are benign phyllodes tumors, the biphasic growth
Phyllodes Tumor 321
Phyllodes Tumor, Fig. 1 (H&E) (a) Architecture of a without pleomorphism, mitotic activity or stromal over-
benign phyllodes tumour. The characteristic leaf-like pro- growth. In this case, the overall cellularity is not increased
cesses are seen throughout a benign phyllodes tumor. (b) illustrating the fact that all of the classic features of a
High power image of benign phyllodes tumour (the same tumour grade may not be present together
case as in Figure 2) shows a minimal increase in cellularity,
322 Phyllodes Tumor
Phyllodes Tumor, Fig. 2 (H&E) (a) Stromal overgrowth Heterologous elements present within a malignant phyl-
and increased cellularity in a malignant phyllodes tumor. lodes tumor. Liposarcomatous differentiation is seen at low
Note the absence of epithelial elements in this medium power (c). Pleopmorphic lipoblasts are seen at high power
power view. (b) Malignant phyllodes tumor shows hyper- (d)
cellular stroma infiltrating normal breast tissue. (c and d)
Phyllodes Tumor, Fig. 3 (H&E) (a and b) Leaf-like overgrowth and no infiltrative margin. A high power view
architecture within a borderline phyllodes tumor (a). The of the same case shows an accentuation of stromal hyper-
mitotic activity was low in this case. There was no stromal cellularity in the subepithelial zone (b)
Phyllodes Tumor, Table 1 Histological features of fibroadenoma, benign, borderline, and malignant phyllodes tumor
Fibroadenoma Benign PT Borderline PT Malignant PT
Stromal Hypocellular but Mild, can be diffuse or Moderate Marked and
hypercellularity can be cellular heterogeneous diffuse
Cellular Absent Mild Moderate Marked
pleomorphism
Mitoses Absent or low (<2 Few (<5 per 10 hpf) Frequent (5–9 per Numerous (10
per 10 hpf) 10 hpf) per 10 hpf)
Borders Well defined Well circumscribed, Well defined, can be Infiltrative
pushing focally infiltrative
Stromal overgrowth None None None or present focally Usually present
Malignant None Absent Absent Can be present
heterologous
differentiation
P
Adapted from WHO Classification of Tumors of the Breast (Tan et al. 2012)
the different grades of tumor and combine the cellularity and architectural abnormality (Tan
assessment of the same features: tumor border, et al. 2012).
stromal cellularity, stromal overgrowth, nuclear As a general guide, it is recommended that
pleomorphism, mitotic activity, and the presence tumors are graded as malignant when all the fea-
of heterologous elements. The WHO three-tier tures of malignancy are present and as borderline
system is the most widely used (Table 1) (Tan when only some are present. The presence of any
et al. 2012). malignant heterologous elements implies a diag-
Assessment and quantification of the histolog- nosis of malignancy (Fig. 2c, d H&E) (Tan
ical features are subjective, and the thresholds for et al. 2016).
each parameter that assign a tumor in one grade
over another are unclear. For example, a benign
phyllodes tumor may show an increase in cellu- Immunophenotype
larity with a focally infiltrative border but with
minimal mitotic activity and few leaflike struc- The diagnosis of phyllodes tumor is made on
tures. It is generally recommended that grading H&E staining; immunohistochemistry is not
should be based on the most florid area of required to make a diagnosis. Stromal cells
324 Phyllodes Tumor
express CD34 and b-catenin, and their expression upregulation of beta-catenin and cyclin D1
is inversely related to tumor grade. Expression of (among others), and the stroma promotes epithe-
proliferation markers, e.g., Ki-67 as well as c-kit lial proliferation via pathways such as IGF and
(CD117), p53, p16, pRb, VEGF, CD10, and IGFR1. The epithelial-stromal feedback is dimin-
EGFR, can be observed in the stromal cells with ished in malignant tumors where there is also
increasing levels of expression from benign to frequent loss of beta-catenin expression (Tan
borderline to malignant phyllodes tumors. et al. 2012).
Immunohistochemistry is required to distin- Detailed genomic analysis has revealed chro-
guish phyllodes tumors from other entities. mosomal changes in phyllodes tumors with the
CD34, p63, p40, and cytokeratins, e.g., aberrations being more frequent and more com-
MNF116, CK 5/6, 34be12, and AE1/3, are most plex in malignant and borderline phyllodes
useful in the distinction between phyllodes tumor tumors compared to benign tumors. Clonal
and metaplastic spindle cell carcinoma. Tradition- changes are detectable in both the epithelial and
ally, CD34 was reportedly positive in the stromal stromal component of phyllodes tumors. Specific
cells in all grades of phyllodes tumor and negative genomic aberrations include gains in chromo-
in carcinomas, whereas p63, p40, and some 1q, losses in chromosome 13, deletions of
cytokeratins were positive in carcinomas and neg- chromosomal 9p, and MYC amplification (Tan
ative in phyllodes tumors. Focal positivity for et al. 2012).
cytokeratin can be observed in malignant phyl-
lodes tumor but not in lower-grade phyllodes
tumors. While benign and borderline phyllodes Differential Diagnosis
tumors and fibroadenomas are negative for p63,
its isoform p40 and cytokeratin, malignant phyl- Benign Phyllodes Tumor
lodes tumors can express p63, p40, and The main differential diagnosis for a benign phyl-
cytokeratin in 57%, 29%, and 21% of cases, lodes tumor is a fibroadenoma (▶ Fibroadenoma).
respectively, and 43% of malignant phyllodes It can often be very challenging because many of
tumors can be negative for CD34. the features relied upon are subject to
interobserver disagreement, and it is important
not to rely on and overinterpret the significance
Molecular Features of any single criterion to distinguish between
these two entities (Tan et al. 2016).
Phyllodes tumors are considered to arise de novo. The distinction is particularly heightened for a
There are very obvious morphological similarities cellular fibroadenoma, a juvenile fibroadenoma,
between fibroadenomas and phyllodes tumors, and fibroadenomas occurring in children and ado-
but direct evidence supporting a precursor role lescents. Intracanalicular growth pattern and leaf-
for fibroadenomas has been lacking. Recent like structures can be seen in both entities. In a
work, however, identified mutations in the medi- phyllodes tumor, the latter structures are generally
ator complex subunit (MED) 12 gene, which is in present throughout the tumor and are hyper-
both fibroadenomas and phyllodes tumors, cellular, whereas they are present only focally
supporting a common molecular link between and are often hyaline and hypocellular in a
both lesions (Tan et al. 2016). fibroadenoma. If stromal fronds are not present
The biphasic appearance of a phyllodes tumor in a benign phyllodes tumor, then the long clefts
is characterized by benign-appearing epithelium may also be a clue to the diagnosis of phyllodes
and a dominant neoplastic stroma, particularly in a tumor (Fig. 4a H&E) (Tan et al. 2016).
periductal location. This feature is in keeping with It is recommended that fibroadenomas are
the hypothesis of an epithelial-stromal cross talk, assessed for features of phyllodes tumors on first
in which the epithelium induces stromal prolifer- viewing whenever there is an increase in stromal
ation via the Wnt signalling pathway and cellularity. A more uniform increase in cellularity
Phyllodes Tumor 325
Phyllodes Tumor, Fig. 4 (H&E) (a) Benign phyllodes lesion. This was classified as a fibroepithelial lesion with a
tumor in which the features overlap with those of a cellular comment that a benign phyllodes tumor could not be
fibroadenoma. In this low power view, leaf-like processes excluded. At low power, an obvious leaf-like architecture
are not seen; however, there are some cleft-like spaces. The is seen with only a minimal increase in stromal cellularity
stromal cellularity and mitotic activity were increased and and no mitotic activity. Fragmentation of the biopsy, as a
some stromal fronds were seen elsewhere in the lesion. result of the needle passing through the fronds, is a clue to
This illustrates the heterogeneity that can be seen within a the diagnosis of a benign phyllodes tumor. This lesion was
benign phyllodes tumor and the diagnostic difficulty in excised because it was large and a diagnosis of a benign
distinguishing benign phyllodes tumors from cellular phyllodes tumor was made based on the features in the
fibroadenomas. (b) Needle core biopsy of a fibroepithelial excised tumor
favors a fibroadenoma. Mitotic activity may not through the frond (Fig. 4b H&E). Mitotic activ-
be particularly helpful in distinguishing between ity is often unhelpful on a core biopsy (Jara-
these entities as it is low in both. Mitotic counts Lazaro et al. 2010).
above 2 per 10 hpf favor a phyllodes tumor how- In many cases, stating that the lesion is a
ever (Tan et al. 2012). fibroepithelial lesion and that a phyllodes
Caution is advised in diagnosing a benign tumor cannot be excluded is the best approach
phyllodes tumor in children and adolescents with the most appropriate management
where increased stromal cellularity and high decided upon through multidisciplinary P
mitotic activity (up to 7 mitoses per 10 hpf) can discussion. Clinical and radiological features
be observed. In this situation, other features of may aid in the decision with excision performed
benign phyllodes tumor, e.g., leaflike architec- if the lesion is growing to >3 cm in size (Jara-
ture, should also be seen to arrive at a diagnosis Lazaro et al. 2010).
of a phyllodes tumor (Tan et al. 2016). Pure stromal tumor is a rare entity in which a
The distinction between fibroadenoma and spindle cell proliferation surrounds open ducts.
benign phyllodes tumor can be especially chal- This entity lacks the architectural leaflike archi-
lenging on a needle core biopsy. Increased stro- tecture of phyllodes tumor, but some reports sug-
mal cellularity has been found to be the most gest that pure stromal tumor and phyllodes tumor
helpful distinguishing feature on core biopsy. may be part of the same disease spectrum (Tan
The threshold cellularity that favors a phyllodes et al. 2012).
tumor is unclear, and some have proposed that it
be present in over half of the core biopsy. The Malignant Phyllodes Tumors
presence of stroma without epithelial elements in Malignant phyllodes tumors need to be distin-
a 10 field, if seen, also favors a phyllodes guished from metaplastic carcinomas (▶ Inva-
tumor. Fragmentation of the biopsy is another sive Metaplastic Carcinoma) because the
clue to the diagnosis of a phyllodes tumor, management of both entities is different. Both
which is most likely due to the core being taken the malignant spindle cells and the heterologous
326 Pleomorphic Lobular Carcinoma
elements within a malignant phyllodes tumor breast core needle biopsies. American Journal of Sur-
can lead to the tumor being confused with the gical Pathology, 38, 1689–1696.
Ellis, I., Lee, A., Pinder, S., & Rakha, E. (2013). Tumors of
different morphological subtypes of a metaplas- the breast. In C. Fletcher (Ed.), Diagnostic histopathol-
tic carcinoma (Tan et al. 2016). ogy of tumors (4th ed., pp. 1064–1065). Philadelpia:
The key feature in this distinction is the dem- Elsevier.
onstration of epithelial differentiation in the Jara-Lazaro, A. R., Akhilesh, M., Thike, A. A., Lui, P. C.-
W., Tse, G. M.-K., & Tan, P. H. (2010). Predictors of
spindle cells to support a diagnosis of carcinoma phyllodes tumours on core biopsy specimens of
and the demonstration of benign admixed epi- fibroepithelial neoplasms. Histopathology, 57, 220–232.
thelium (i.e., biphasic growth) to support a diag- Tan, P. H., Tse, G., & Kee, A. (2012). Fibroepithelial
nosis of malignant phyllodes tumor. In phyllodes tumours. In S. Lakhani, I. Ellis, S. Schnitt, P. H. Tan,
& M. van de Vijver (Eds.), WHO classification of
tumors, epithelium is both morphologically tumours of the breast (4th ed., pp. 143–147). Geneva:
benign and distinct from the stromal elements, World Health Organisation.
and thorough sampling may be required to iden- Tan, B. Y., Acs, G., Apple, S. K., Badve, S., Bleiweiss, I. J.,
tify it. Demonstration of convincing epithelial Brogi, E., . . . Tan, P. H. (2016). Phyllodes tumours of
the breast: A consensus review. Histopathology, 68,
differentiation within the stromal compartment 5–21.
supports a diagnosis of carcinoma. Again, this
may require thorough sampling and the demon-
stration of cytokeratin positivity, bearing the
caveats outlined above in mind (see Pleomorphic Lobular
immunophenotype). The presence of ductal car- Carcinoma
cinoma in situ adjacent to malignant spindle cells
also favors a metaplastic carcinoma (Ellis et al. Sofia Asioli1 and Chiara Baldovini2
1
2013; Tan et al. 2016). In difficult cases, GATA3 Department of Biomedical and Neuromotor
and SOX10 can also be helpful. GATA3 is rarely Sciences (DIBINEM)-Surgical Pathology
expressed in malignant phyllodes tumors and is Section, Alma Mater Studiorum – University of
expressed in up to 50% of metaplastic carcino- Bologna, Bologna, Italy
2
mas; SOX10 is negative in malignant phyllodes Pathology Unit, Giannina Gaslini Institute,
tumors and is expressed in 40% of metaplastic Genoa, Italy
carcinomas. b-catenin is expressed in both
malignant phyllodes tumors and metaplastic car-
cinoma and is therefore unhelpful in Definition
distinguishing these entities (Cimino-Mathews
et al. 2014). Pleomorphic lobular carcinoma (PLC) is currently
Distinction between malignant phyllodes defined by the World Health Organization
tumor and the rare entity of pure stromal sarcoma (Lakhani et al. 2012) as a rare variant of invasive
is made by the finding of the epithelial elements lobular carcinoma (ILC), exhibiting marked
of a phyllodes tumor. These can be very focal if nuclear pleomorphism and cellular atypia. It may
the stromal overgrowth is particularly pro- be associated with pleomorphic lobular carcinoma
nounced and thorough sampling is required. in situ (PLCIS), which refers to a lobular carci-
However, the management of both is similar noma in situ (LCIS) with high-grade cytological
(Tan et al. 2016). features. PLC was first documented by Martinez
and Azzopardi (1979) then subsequently reported
by Dixon (Dixon et al. 1982) and formally
described by Page and Anderson (1987). Further
References and Further Reading
characterization was provided in 1992 by Eusebi
Cimino-Mathews, A., Sharma, R., Illei, P. B., Vang, R., & (Eusebi et al. 1992) and Weidner (Weidner and
Argani, P. (2014). A subset of malignant phyllodes Semple 1992), who emphasized its aggressive
tumors express p63 and p40: A diagnostic pitfall in behavior and poor clinical outcome.
Pleomorphic Lobular Carcinoma 327
resonance imaging (MRI) has become increas- (WHO 2012). Sometimes, neoplastic cells may
ingly important in the detection of multifocal, also show singular intracytoplasmic vacuoles giv-
multicentric or contralateral disease, especially ing them the appearance of signet-ring cells. Polyg-
for patients diagnosed with lobular carcinoma onal cells with eosinophilic cytoplasm reminiscent
including PLC. of rhabdomyoblasts are also occasionally found
(Fig. 3 H&E). In contrast with ILC-C, PLC often
displays evidence of vascular invasion and high
Microscopy mitotic index. Owing to the marked degree of
nuclear pleomorphism, PLCs are more likely to
The original morphologic criteria are still applied to be scored as G2, G3 within the Nottingham grading
identify PLC. Although some investigators found system, depending on mitotic activity.
that nonclassical growth patterns were more fre-
quent in tumors with marked nuclear pleomor-
phism, in most studies PLC did not display
significant differences from ILC in terms
of architecture. Similar to ILC-C, PLC displays
poorly circumscribed lesions, characterized by dis-
sociated neoplastic cells, which may delineate
“targetoid” patterns around ducts and acini (Fig. 1
H&E). Alveolar, solid, and mixed patterns of
growth may be seen as well. In contrast to ILC-C,
PLC nuclei are larger – approximately four times
the size of a lymphocyte (Weidner et al. 1992) –
indented and often eccentrically placed. Prominent
and multiple nucleoli are generally present (Fig. 2
H&E). The cytoplasm is abundant, pale to eosino-
Pleomorphic Lobular Carcinoma, Fig. 2 The neoplas-
philic, but varying degrees of granular to vacuo- tic nuclei of PLC are larger, indented and often eccentri-
lated cells have been described. Indeed, according cally placed with prominent and multiple nucleoli (H&E)
with the last WHO classification, PLC may dem-
onstrate apocrine or histiocytoid differentiation
pattern typical of ductal carcinoma along with Derksen, P. W., Braumuller, T. M., et al. (2011). Mammary-
E-cadherin positivity. specific inactivation of E-cadherin and p53 impairs
functional gland development and leads to pleomorphic
invasive lobular carcinoma in mice. Disease Models &
Pleomorphic lobular carcinoma
Mechanisms, 4, 347–358.
Definition Dixon, J. M., Anderson, T. J., et al. (1982). Infiltrat-
– A rare variant of invasive lobular carcinoma (ILC), ing lobular carcinoma of the breast. Histopathology,
exhibiting marked nuclear pleomorphism 6, 149–161.
Age distribution Eusebi, V., Magalhaes, F., et al. (1992). Pleomorphic lob-
– Postmenopausal women aged between 60 and 80 ular carcinoma of the breast: An aggressive tumor
Clinical features showing apocrine differentiation. Human Pathology,
– Often arises bilaterally, showing multifocal 23, 655–662.
distribution Jung, S. P., Lee, S. K., et al. (2012). Invasive pleomorphic
Microscopic findings lobularcarcinoma of the breast: Clinicopathologic char-
– Dissociated neoplastic cells acteristics and prognosis compared with invasive duc-
– Nonspecific architectural pattern (classic, alveolar, tal carcinoma. Journal of Breast Cancer, 15, 313–319.
solid, mixed) Lakhani, S., Ellis, I., et al. (2012). WHO classification of
– Nuclei: Large, indented, lobulated, and eccentric Tumours of the breast (4th ed.). Lyon: IARC Press.
– Cytoplasm: Abundant, eosinophilic, granular, Liu, Y. L., Choi, C., et al. (2017). Invasive lobular breast
vacuolated (single or multiple vacuoles) carcinoma: Pleomorphic versus classical subtype, asso-
– Frequent vascular invasion and high mitotic activity ciations and prognosis. Clinical Breast Cancer,
– PLCIS 8209(16), 30558–30554.
Immunohistochemical findings Martinez, V., & Azzopardi, J. G. (1979). Invasive lobular
– E-cadherin: Negative carcinoma of the breast: Incidence and variants. Histo-
– Estrogen, progesterone, androgen receptors: pathology, 3, 467–488.
Frequently positive Narendra, S., Jenkins, S. M., et al. (2015). Clinical
– HER2: May be positive (2+/3+: 40–80%, 3+: 13%) outcome in pleomorphic lobular carcinoma: A case-
– P53: Frequently positive control study with comparison to classic invasive
– GCDFP-15: Frequently positive lobular carcinoma. Annals of Diagnostic Pathology,
19, 64–69.
Molecular findings
Page, D. L., & Anderson, T. J. (1987). Diagnostic his-
– Lobular histotype typical alterations: 1q+, 11q ,
topathology of the breast. Edinburgh: Churchill
16p+, and 16q
Livingstone.
– Ductal histotype typical alterations: Gains of HER2/
Simpson, P. T., Reis-Filho, J. S., et al. (2008). Molecular
neu, c-myc, p53 positivity, amplification of 8q24, 12q14,
profiling pleomorphic lobular carcinomas of the breast:
and 20q13
Evidence for a common molecular genetic pathway
– LOH BRCA2 40%
with classic lobular carcinomas. Journal of Pathology,
Differential diagnosis 215, 231–244.
– High-grade invasive carcinoma no special type Weidner, N., & Semple, J. P. (1992). Pleomorphic variant
(NST) of invasive lobular carcinoma of the breast. Human
– Apocrine carcinoma Pathology, 23, 1167–1171.
Prognosis and treatment
– Unfavorable prognostic factors
– Recent evidence of DFS and OS similar to ILC-C
– Mastectomy +/ adjuvant therapy (endocrine,
trastuzumab) Polymorphous Carcinoma
• Incidence
Microscopy
Five cases of this special type of primary breast
carcinoma have been reported in literature with
Polymorphus carcinoma shows the same morpho-
morphological description.
logical features as that of minor salivary glands
• Age
counterpart.
Median age at presentation is 55 years, ranging
In particular, this neoplasm is non-encapsu-
from 37 to 74 years.
lated and is constituted of neoplastic cells show-
• Sex
ing a polymorphous structure that consists of
All the patients reported in literature are
central areas showing solid nests, surrounded at
female.
the periphery by alveolar and cribriform struc-
• Site
tures as well as trabeculae and “Indian file”
Breast without the prevalence of a specific
arranged cells. Combinations and transitions
mammary quadrant.
among all these patterns are frequent. The neo-
• Treatment
plastic cells are uniformly small, have round to
Wide excision with axillary lymph node dis-
ovoid nuclei with dispersed vesicular chromatin,
section is the choice of treatment of the cases
and have inconspicuous to small nucleoli.
reported in the literature, followed by radio-
Mitotic activity ranges from 10 to 12 per
therapy in two patients. Although no lymph
10 high-power fields; necrotic areas are not P
node metastases are found, polymorphous car-
seen. Altogether the histological grade,
cinoma seems to be an aggressive neoplasm
according to the Elston and Ellis grading system
and recommended treatment consists of wide
of breast cancer (Elston and Ellis 1991), is grade
excision with sentinel node biopsy and even-
2. Neither vascular nor perineural invasion has
tually axillary lymph node dissection.
been reported (Fig. 1).
• Outcome
Follow-up has been reported in three cases
(Asioli et al. 2006). In two out of these three
patients, the follow-up was very short. The Immunophenotype
other case had a very aggressive clinical course
with the patient developing liver metastases Estrogen and progesterone receptors, as well as
and dying of widespread disease 3 years after HER2, were consistently negative, so this entity is
the diagnosis. Therefore, it was suggested to considered as triple-negative breast carcinoma by
name the tumor in the breast as “polymorphous immunohistochemistry (Asioli et al. 2006). The
adenocarcinoma,” avoiding the term of “low neoplastic cells are faintly positive for cytokeratin
grade.” The same conclusion has led to rename 7, E-cadherin, and EGFR, and strongly stain for
the salivary gland counterpart as polymor- Bcl-2. On the contrary, EMA, cytokeratin
phous carcinoma, as at long-term follow-up 14, smooth muscle actin, and c-Kit are negative
the incidence of local recurrences and distant (Foschini et al. 2017; Reyes et al. 2013).
332 Polymorphous Carcinoma
Polymorphous Carcinoma, Fig. 1 Polymorphous car- magnification x100), trabeculae (c; H&E, Original magni-
cinoma shows different growth patterns including central fication x200) and “Indian file” infiltration pattern (d;
areas with solid nests (a; H&E, Original magnification H&E, Original magnification x40)
x40), alveolar and cribriform structures (b; H&E, Original
Clinical Features P
References
• Incidence
Asioli, S., Marucci, G., Ficarra, G., Stephens, M.,
Foschini, M. P., Ellis, I. O., & Eusebi, V. (2006).
PASH of the breast was first described by
Polymorphous adenocarcinoma of the breast. Report Vuitch et al. in 1986 (Vuitch et al. 1986). Its
of three cases. Virchows Archiv, 448, 29–34. incidence was later investigated in a series of
Elston, C. W., & Ellis, I. O. (1991). Pathological prognostic 200 consecutive breast biopsies and surgical
factors in breast cancer. 1. The value of histological
grade in breast cancer: Experience from a large study
specimens and was found to be an incidental
with long-term follow-up. Histopathology, 19, finding in breast biopsies in 23% of the cases
403–410. (Ibrahim et al. 1989). Incidental PASH can be
Foschini, M. P., Morandi, L., Asioli, S., Giove, G., found in fibroadenomas, gynecomastia, nor-
Corradini, A. G., & Eusebi, V. (2017). The morpholog-
ical spectrum of salivary gland type tumours of the
mal breast tissue, hamartomas, and sclerosing
breast. Pathology, 49, 215–227. lobular hyperplasia. It has also been described
Reyes, C., Jorda, M., & Gomez-Fernández, C. (2013). in surgical specimens of gigantomastia. Two
Salivary gland-like tumors of the breast express cases were described in immunosuppressed
basal-type immunohistochemical markers. Applied
Immunohistochemistry and Molecular Morphology,
patients (Seidman et al. 1993 and de Saint
21, 283–286. Aubain Somerhausen et al. 1997). In a case
334 Pseudoangiomatous Stromal Hyperplasia
series of 24 mass forming PASH, it was found appears as an oval, circumscribed, hypoechoic
that 40% of the female patients had family mass. On magnetic resonance imaging, PASH
history of breast cancer (Bowman et al. 2012). usually has progressive (Type 1) enhancement,
• Age and high-signal slit-like spaces may be seen on
PASH occurs in pre- or perimenopausal T2-weighted and short tau inversion recovery
patients and is often associated with fibrocystic (STIR) images (Raj et al. 2017). A case of
changes (Ibrahim et al. 1989). The mean age of PASH (confirmed in a subsequent core biopsy)
patients diagnosed with tumor forming was incidentally revealed due to focal
(nodular) PASH is 37 years (Powell et al. increased radiotracer uptake in the left breast
1995). However, PASH was also described in of a prostate cancer patient undergoing
postmenopausal women: among them more Ga-labeled prostate-specific membrane anti-
than half of those with tumor forming PASH gen ligand PET-CT (Malik et al. 2017).
were receiving hormone replacement therapy • Treatment
(Virk and Khan 2010). Rare cases of PASH The current recommendations are to excise
causing voluminous and rapidly growing lesions more than 2 cm and conservative man-
breast mass with severe breast asymmetry agement and watchful waiting with smaller
occurring in adolescents have been described lesions. Successful treatment of PASH with
(Pellini et al. 2018). Tamoxifen in a patient with PASH presenting
• Sex with breast enlargement, pain, and breast
PASH is mainly found in female breast tissue, masses was described (Pruthi et al. 2001).
but it is also common in male gynecomastia: • Outcome
according to one study, 23.8% of gynecomastia Sporadic cases of recurrent PASH have been
cases harbored PASH (Milanezi et al. 1998). described following incomplete excision. On
Occurrence of PASH in the breast of a trans- the other hand, spontaneous regression of
gender male patient has been described PASH is also documented in the literature.
(Bowman et al. 2012).
• Site
Macroscopy
No specific site in the breast has been
described. PASH was reported in axillary
Only PASH forming a nodular lesion in the breast
accessory breast tissue (Lee et al. 2005).
can be seen macroscopically: the typical appear-
• Symptoms
ance is a well-circumscribed firm or rubbery
PASH is found incidentally in most of the cases
tumor with a white-gray/tan cut surface (Powell
without any clinical symptoms. Fewer cases or
et al. 1995). The largest lesion described in the
case series are described in the literature with
literature measured 20 cm (Sasaki et al. 2008).
PASH occurring as palpable mass. Raj et al.
reported 44% of PASH presenting as palpable
breast lesion (Raj et al. 2017). A rare manifes-
tation is diffuse massive process with asymme- Microscopy
try of the breast (Shahi et al. 2015). PASH was
also described as a cause of gigantomastia Microscopically, a spectrum of pathological stro-
(Roy et al. 2015). In a published series of mal changes can be seen ranging from anastomos-
seven cases, the lesions were identified on ing slit-like spaces lined by flat, bland spindle
mammograms as noncalcified masses measur- cells (Figs. 1 and 2) to more proliferative lesions
ing 1.1–11 cm in largest diameter (Polger et al. composed of bundles of plump spindle cells that
1996). Mammographic and ultrasound appear- may obscure the underlying pseudoangiomatous
ance of nodular PASH is very similar to those architecture in the most florid lesions (Powell
of fibroadenomas: on ultrasound, PASH often et al. 1995).
Pseudoangiomatous Stromal Hyperplasia 335
Differential Diagnosis Malik, D., Basher, R. K., Mittal, B. R., Jain, T. K., Bal, A.,
& Singh, S. K. (2017). 68Ga-PSMA expression in
Pseudoangiomatous stromal hyperplasia of the breast.
The most important differential diagnosis is well Clinical Nuclear Medicine, 42, 58–60.
differentiated angiosarcoma (▶ Angiosarcoma of Milanezi, M. F., Saggioro, F. P., Zanati, S. G., Bazan, R., &
the Breast). Schmitt, F. C. (1998). Pseudoangiomatous hyperplasia
of mammary stroma associated with gynaecomastia.
Journal of Clinical Pathology, 51, 204–206.
Nassar, H., Elieff, M. L., Kronz, J. D., & Argani, P. (2010).
References and Further Reading Pseudoangiomatous stromal hyperplasia (PASH) of the
breast with focal of morphologic malignancy: A case of
Bowman, E., Oprea, G., Okoli, J., Gundry, K., Rizzo, M., PASH with malignant transformation? International
Gabram-Mendola, S., Manne, U., Smith, Journal of Surgical Pathology, 18, 564–569.
G., Pambuccian, S., & Bumpers, H. L. (2012). Pseudo- Noda, Y., Nishimae, A., Sawai, Y., Inaji, H., & Yamasaki, M.
angiomatous stromal hyperplasia (PASH) of the breast: (2019). Atypical pseudoangiomatous stromal hyperpla-
A series of 24 patients. The Breast Journal, 18, sia showing rapid growth of the breast: Report of a case.
242–247. Pathology International. https://doi.org/10.1111/
Comunoğlu, N., Comunoğlu, C., Ilvan, S., Calay, Z., & pin.12786.
Müslümanoğlu, M. (2007). Mammary Pseudo- Pellini, D. F., Lorenzi, M., Gaudino, R., Accordini, B.,
angiomatous stromal hyperplasia composed of pre- Mirandola, S., Invento, A., & Pollini, P. D. (2018).
dominantly Giant cells: An unusual variant. The Pseudoangiomatous stromal hyperplasia (PASH) in
Breast Journal, 13, 568–570. adolescence: A systematic review. World Journal of
Damiani, S., & Eusebi, V. (2001). Gynecomastia in type-1 Surgery and Surgical Research, 1, 1058.
neurofibromatosis with features of pseudoangiomatous Polger, M. R., Denison, C. M., Lester, S., & Meyer, J. E.
stromal hyperplasia with giant cells. Report of two (1996). Pseudoangiomatous stromal hyperplasia:
cases. Virchows Archiv, 438, 513–516. Mammographic and sonographic appearances. Ameri-
de Saint Aubain Somerhausen, N., Larsimont, D., Cluydts, N., can Journal of Roentgenology, 166, 349–352.
Heymans, O., & Verhest, A. (1997). Pseudo- Powell, C. M., Cranor, M. L., & Rosen, P. P. (1995).
angiomatous hyperplasia of mammary stroma in an Pseudoangiomatous stromal hyperplasia (PASH):
HIV patient. General & Diagnostic Pathology, 143, A mammary stromal tumor with myofibroblastic dif-
251–254. ferentiation. American Journal of Surgical Pathology,
Ferreira, M., Albarracin, C. T., & Resetkova, E. (2008). 19, 270–277.
Pseudoangiomatous stromal hyperplasia tumor: Pruthi, S., Reynolds, C., Johnson, R. E., & Gisvold, J. J.
A clinical, radiologic and pathologic study of (2001). Tamoxifen in the management of Pseudo-
26 cases. Modern Pathology, 21, 201–207. angiomatous stromal hyperplasia. The Breast Journal,
Ibrahim, R. E., Sciotto, C. G., & Weidner, N. (1989). 7, 434–439.
Pseudoangiomatous hyperplasia of mammary stroma. Raj, S. D., Sahani, V. G., Adrada, B. E., Scoggins, M. E.,
Some observations regarding its clinicopathologic Albarracin, C. T., Woodtichartpreecha, P., Posleman
spectrum. Cancer, 63, 1154–1160. Monetto, F. E., & Whitman, G. J. (2017). Pseudo-
Krawczyk, N., Fehm, T., Ruckhäberle, E., Mohrmann, S., angiomatous stromal hyperplasia of the breast: Multi-
Riemer, J. Braunstein, S. Hoffmann, J. (2016) Bilateral modality review with pathologic correlation. Current
diffuse Pseudoangiomatous stromal hyperplasia Problems in Diagnostic Radiology, 46, 130–135.
(PASH) causing Gigantomastia in a 33-year-old preg- Recavarren R. A., Chivukula M., Carter G., Dabbs D. J.
nant woman: Case report. Breast Care, 11, 356–358. (2009). Columnar cell lesions and pseudoangiomatous
Krings, G., McIntire, P., & Shin, S. J. (2017). hyperplasia like stroma: is there an epithelial-stromal
Myofibroblastic, fibroblastic and myoid lesions of the interaction? International Journal of Clinical and
breast. Seminars in Diagnostic Pathology, 34, Experimental Pathology, 3, 87–97.
427–437. Rosen, P. P. (2001). Rosen’s breast pathology. Philadel-
Lee, J. S., Oh, H. S., & Min, K. W. (2005). Mammary phia: Lippincott Williams & Wilkins.
pseudoangiomatous stromal hyperplasia presenting as Roy, M., Lee, J., Aldekhayel, S., & Dionisopoulos, T.
an axillary mass. The Breast, 14, 61–64. (2015). Pseudoangiomatous stromal hyperplasia:
Leon, M. E., Leon, M. A., Ahuja, J., & Garcia, F. U. A rare cause of idiopathic Gigantomastia. Plastic and
(2002). Nodular myofibroblastic stromal hyper- Reconstructive Surgery. Global Open, 3, e501.
plasia of the mammary gland as an accurate name Sasaki, Y., Kamata, S., Saito, K., Nishikawa, Y., & Ogawa, J.
for pseudoangiomatous stromal hyperplasia of (2008). Pseudoangiomatous stromal hyperplasia (PASH)
the mammary gland. The Breast Journal, 8, of the mammary gland: Report of a case. Surgery Today,
290–293. 38, 340–343.
Pseudoangiomatous Stromal Hyperplasia 337
Seidman, J. D., Borkowski, A., Aisner, S. C., & Sun, A rare tumor of the breast. Journal of Cancer Research
C. C. (1993). Rapid growth of pseudoangiomatous and Therapy, 11, 1032.
hyperplasia of mammary stroma in axillary Virk, R. K., & Khan, A. (2010). Pseudoangiomatous stro-
gynecomastia in an immunosuppressed patient. mal hyperplasia. An overview. Archives of Pathology
Archives of Pathology and Laboratory Medicine, and Laboratory Medicine, 134, 1070–1074.
117, 736–738. Vuitch, M. F., Rosen, P. P., & Erlandson, R. A. (1986).
Shahi, K. S., Bhandari, G., Gupta, R. K., & Sawai, M. Pseudoangiomatous hyperplasia of mammary stroma.
(2015). Pseudoangiomatous stromal hyperplasia: Human Pathology, 17, 185–191.
P
R
on average 15.5 per breast. Linell who of the cases diagnosed as radial scar. In almost
performed a study of 555 mastectomy speci- 10% of the cases, the lesion is better visualized
mens found radial scars in 16%, but in the by ultrasound than mammography. On MRI,
group of breast cancer patients radial scars radial scars possess variable morphology and
were present in half of the specimens (Linell their kinetic features are also not uniform.
1985). These findings led to suggest that radial Since they can demonstrate false positive
scars may be precancerous lesions. enhancement, ill-defined borders, and irregular
• Age shape, not infrequently MRI findings mimic
In large studies, the age range of radial scars invasive carcinoma.
varies between 25 and 89 years. The median of • Treatment
this range is around the perimenopausal age Given that core biopsy diagnosis of radial scars
group. is coded as B3 according to present guidelines,
• Sex these lesions need further multidisciplinary
Radial scars are mainly found in female work-up. Until a few years ago, core biopsy
breasts, but in occasional rare cases it may diagnosis of a radial scar was an absolute indi-
occur in male breasts as well. cation for surgical excision, and in large series
• Site the upgrade rate for malignancy varied between
No specific site had been described as concerns 0% and 24%. More recently, with the wide-
laterality or quadrant of the breast. spread use of vacuum-assisted core biopsy
• Imaging (VACB), the indications for surgical excision
Many papers had been published describing became more restricted. In the latest update of
the imaging features of radial scars given that the UK Breast Screening Quality Assurance
the structure of these lesions may mimic small Guideline (2016 update), the management of
stellate carcinoma on mammography. Tabar radial scars differs if the lesion was incidental
and Dean (1983) have first described system- (not seen on mammography) or was identified
atically the mammographic features that enable as a mammographic abnormality. For those
the radiology diagnosis, as follows: radial scars detected only histologically on
(i) depending on the view, the appearance of core biopsy performed for other radiologically
the detected lesion is variable, (ii) in relation to identified benign lesion, no further intervention
the long and thin spicules, the central opacity is is needed. For those radial scars detected as
small, (iii) parallel to the spicules a very thin, mammographic abnormality, at least 12 VACB
radiolucent line can be seen, (iv) the center of cores is recommended as a second intervention.
the lesion is radiolucent, and (v) there is dis- Radial scars completely removed by VACB
cordance between the mammographic finding require surgical excision only if epithelial atypia
of a stellate lesion and the lack of a palpable is associated to the lesion.
lesion by physical examination. Digital mam- • Outcome
mography is more sensitive than analogue In one of the longest follow-up study of
mammography, and breast tomosynthesis 32 patients with radial scar, only one patient
apparently discovers even more stellate lesions developed ipsilateral breast cancer 16 years
than analogue or digital mammography, thus after the diagnosis of the radial scar
seems to increase slightly the recall rate in (Andersen and Gram 1984). The mean
screening programs. In the Malmo Breast follow-up period in this study was 19.5 years
Tomosynthesis Screening Trial, around 10% (range 15–24 years). Many studies in the eight-
of the false positive recalls were due to radial ies suggested an elevated cancer risk associ-
scars. Ultrasound detects up to two-third of ated to radial scars; however, large recent
radial scars, most commonly as a hypoechoic studies are less convincing especially
area or mass. Parenchymal distortion without a concerning radial scars devoid of atypical epi-
hypoechoic mass is present in about one-fifth thelial proliferation and assessed on VACB.
Radial Scar 341
Macroscopy
Microscopy
Radial Scar, Fig. 2 H&E: (a) Distorted small ducts lacking any organoid arrangement in the central part of a small radial
scar. (b) Radial scar associated with flat epithelial atypia
342 Radial Scar
periphery. Within the central scar, very often one In some lesions, lymphoid inflammatory infiltrate
finds entrapped and distorted or ramifying small is also found. Presence of small microcalcification
ducts lacking any organoid arrangement particles is not uncommon. Radial scars often
(Fig. 2a). The nuclei of the epithelial cells lying occur in the background of fibrocystic breast
the entrapped ducts are devoid of atypia and no parenchyma. The presence of atypical epithelial
mitotic figures are present. Occasionally, the proliferations (Fig. 2b), in situ carcinomas, or
central elastosis and the scar itself is so much invasive carcinomas in radial scars has been
reminiscent to the contour of a totally obliterated described in larger lesions of older women
larger duct, for in the past periductal mastitis was (Sloane and Mayers 1993). Association of inva-
supposed as an etiologic factor for these lesions. sive carcinoma has not been described in lesions
However, there are opponents to this theory who that are smaller than 5 mm.
believe that radial scars are per se proliferative
lesions of the breast.
The radiating projections contain epithelial Immunophenotype
structures: ducts with various degree of dilatation
(Fig. 3), often with usual type ductal hyperplasia. High molecular weight cytokeratins (cytokeratin
Sclerosing adenosis, small papillomas, adenosis, 5/6, 14) and myoepithelial markers (p63, p40,
and columnar cell change are also common fea- calponin, CD10, etc.) can help to identify the
tures. Apocrine changes may also be associated. benign nature of the epithelial proliferation asso-
ciated with a radial scar or may help to identify an
associated in situ or invasive cancer (Fig. 4).
Molecular Features
Radial Scar, Fig. 4 (a) p63 immunohistochemistry highlights the myoepithelial cells and (b) CK14 immunohistochem-
ical reaction shows the heterogeneous epithelial cell population in entrapped small ducts in the center of a radial scar
Radial Scar 343
opposed to only 50% of tubular carcinomas that The differential diagnosis may be more diffi-
showed diploidy. However, using flow cytometry, cult in 14G needle core biopsies. However, in
both radial scars and tubular carcinomas proved close cooperation with the radiologist, the diag-
diploid. In the early 2000s, stromal mRNA nosis of a radial scar can be made with confi-
expression pattern of radial scars and invasive dence even in such specimens. Tissue fragments
cancers was compared using small number of showing the central elastotic scar with entrapped
cases and normal controls. Findings suggested small ducts with normal two layered epithelial
that expression of mRNAs encoding proteins lining (or usual type hyperplasia) are features
involved in the vascular stroma shows similarities that help to identify RSs even in fragmented
between radial scars and invasive breast cancers specimens.
(Jacobs et al. 2002). Chromosomal alterations
have also been described in radial scars, namely,
allelic imbalance of chromosome 16q and 8p.
References and Further Reading
This finding suggested that some areas of radial
scars are clonal and may be neoplastic (Iqbal et al. Andersen, J. A., & Gram, J. B. (1984). Radial scar in the
2002). More recently a few studies using next female breast. A long-term follow-up study of 32 cases.
generation sequencing (NGS) detected PIK3CA Cancer, 53, 2557–2560.
D’Amore, E., Montes, E., Le, M. G., Lacombe, M. J.,
mutations in radial scars with higher frequency
Bertin, F., Castaigne, D., & Contesso, G. (1985).
compared to the 25–30% PIK3CA mutation fre- Aschoff’s center of proliferation. Experience of the
quency detected in series of invasive breast carci- Gustave Roussy Institute. Annals of Pathology, 5,
nomas (Wilsher et al. 2017). 173–182.
Hamperl, H. (1975). Strahlige Narben und obliterierende
Mastopathie. Virchows Archiv (Pathological Anatomy),
369, 55.
Differential Diagnosis Iqbal, M., Shoker, B. S., Foster, C. S., Jarvis, C.,
Sibson, D. R., & Davies, M. P. (2002). Molecular and
genetic abnormalities in radial scar. Human Pathology,
Radial scars may be misdiagnosed by the
33, 715–722.
unexperienced pathologist as invasive breast car- Jacobs, T. W., Byrne, C., Colditz, G., Connolly, J. L.,
cinoma NST (▶ Invasive Carcinoma NST) or & Schnitt, S. J. (1999). Radial scars in benign
tubular carcinoma (▶ Tubular Carcinoma). It is breast-biopsy specimens and the risk of breast can-
cer. New England Journal of Medicine, 340(6),
noteworthy to mention that the irregular, haphaz-
430.
ardly arranged, compressed tubular structures in Jacobs, T. W., Schnitt, S. J., Tan, X., & Brown, L. F. (2002).
radial scars are exclusively seen in the central Radial scars of the breast and breast carcinomas have
elastotic nidus. In invasive cancers, the neoplastic similar alterations in expression of factors involved in
vascular stroma formation. Human Pathology, 33, R
tubules often invade the fat tissue which is not
29–38.
a feature in radial scars. Furthermore, in the rela- Lång, K., Nergården, M., Andersson, I., Rosso, A., &
tively wide radiating arms, there are ducts and/or Zackrisson, S. (2016). False positives in breast cancer
lobules showing various degree of the screening with one-view breast tomosynthesis: An
analysis of findings leading to recall, work-up and
abovementioned proliferative epithelial changes.
biopsy rates in the Malmö Breast Tomosynthesis
As opposed to this, in small grade 1 carcinomas or Screening Trial. European Radiology, 26(389),
tubular carcinomas, the neoplastic tubules build 9–3907.
up the whole lesion and most of their stellate Linell, F. (1985). Radial scars of the breast and their sig-
nificance for diagnosis and prognosis. Verhandlungen
peripheral projections are devoid of preexisting
der Deutschen Gesellschaft für Pathologie, 69,
ducts or lobules. In equivocal cases, immunohis- 108–118.
tochemical markers for myoepithelial cells can Mengmeng, L. V., Xingya, Z., Shanliang, Z., et al. (2014).
assist to exclude malignancy. In case of uncer- Radial scars and subsequent breast Cancer risk: A meta
analysis. PLoS One, 9, e102503.
tainty, high molecular weight cytokeratins are
NHS Breast Screening Programme Clinical guidance for
useful to characterize the associated intraductal breast cancer screening assessment. (2016). NHSBSP
proliferations. publication number 49, 4th ed.
344 Radial Scar
Osborn, G., Wilton, F., Stevens, G., Vaughan-Williams, E., sclerosing lesions: Importance of lesion size and patient
& Gower-Thomas, K. (2011). A review of needle core age. Histopathology, 23, 225–231.
biopsy diagnosed radial scars in the Welsh Breast Tabar, L., & Dean, P. B. (1983). Teaching atlas of mammog-
Screening Programme. Annals of Royal College of raphy (pp. 95–96). Stuttgart: Georg Thieme Verlag.
Surgeons England, 93, 123–126. Wellings, S. R., & Alpers, C. E. (1984). Subgross patho-
Ruiz-Saurí, A., Almenar-Medina, S., Callaghan, R. C., logic features and incidence of radial scars in the breast.
Calderon, J., & Llombart-Bosch, A. (1995). Radial Human Pathology, 15, 475–479.
scar versus tubular carcinoma of the breast. Wilsher, M. J., Owens, T. W., & Allcock, R. J. (2017).
A comparative study with quantitative techniques Next generation sequencing of the nidus of early
(morphometry, image- and flow cytometry). Pathology (adenosquamous proliferation rich) radial sclerosing
Research and Practice, 191, 547–554. lesions of the breast reveals evidence for a neoplastic
Sloane, J. P., & Mayers, M. M. (1993). Carcinoma and precursor lesion. Journal of Pathology: Clinical
atypical hyperplasia in radial scars and complex Research, 3, 115–122.
S
Sclerosing Adenosis, Fig. 1 SA associated with micro- appearance (b), and localized within enlarged and distorted
calcifications. Specimen mammography shows clusters of lobules (c, d). The single black arrows on pictures d
cotton ball-like, powdery calcifications (a). Thick-section represent the two expanded foci of SA comparing to an
images demonstrate that the radio dense phosphate calcifi- intermingled normal lobule depicted with double black
cations can have “onion ring” like psammomatous- arrows
calcifications may be amorphous and pleo- and the distinction from infiltrating carci-
morphic and in such cases indistinguishable noma is impossible.
from non-comedo DCIS. SA is less com- – Magnetic resonance imaging (MRI)
monly detected as a mass lesion, which is Typical SA lesions are indistinguishable
generally circumscribed or lobulated and from breast parenchyma, but the enhance-
may be associated with microcalcifications. ment may increase moderately or rapidly
In rare cases, SA presents as an architectural over time making it difficult to distinguish
distortion. nodular SA from malignant lesions.
– Ultrasound • Treatment
Nodular SA often appears as a circum- Some evidence suggests that SA may regress
scribed, hypoechoic solid lesion. In cases after the menopause. When SA is associated
with asymmetrical density on mammogra- with non-proliferative lesions as a part of fibro-
phy, the typical ultrasound feature is focal cystic change, further excision is not
acoustic shadowing without a mass config- recommended. If SA is diagnosed in a core nee-
uration. Mass-like lesions with irregular dle biopsy (CNB), surgical excision of
contour in association with marked poste- the lesional area is recommended to rule out
rior acoustic shadowing are also observed, carcinoma, particularly in cases associated with
Sclerosing Adenosis 347
Immunophenotype
Microscopy
Due to its origin, SA maintains the immuno-
At low power three main forms of SA could phenotype of normal lobules (i.e., CK7, CK 8/18,
be observed: tiny foci in an otherwise normal breast p63, p40, smooth muscle actin, calponin, and high-
tissue, especially in perimenopausal women, larger molecular weight cytokeratins (CK5/6, CK14)).
but still microscopic lesions as part of the spectrum Using of more than one myoepithelial marker or
of fibrocystic change, and lesions that may form a “cocktail” of markers can be helpful (Pavlakis et al.
348 Sclerosing Adenosis
Sclerosing Adenosis, Fig. 2 Histology of SA. Enlarged partly obscured lumens on higher magnification (c, H&E
lobule with dilated glands (a, H&E 20) and retained 100) and the outer layer of myoepithelial cells being
myoepithelial cell layer which is highlighted with the p63 positive with CK14 IHC (d)
immunostain (b). Array of microtubular structures with
Sclerosing Adenosis, Fig. 3 SA harboring LCIS. The cytoplasmic membrane positivity and mixed with focally
part of SA shows irregularly distributed acini expanded by remaining normal glandular cells with continuous and
atypical cells of lobular type (a, H&E 80). E-cadherin intensive membrane staining (b). The myoepithelial cells
IHC shows that the cells of LCIS are completely lacking are intact as indicated by p63 IHC (c)
Microglandular Adenosis (MGA) A nested case-control study from the Mayo Benign
MGA (▶ Microglandular Adenosis) is composed Breast Disease Cohort. Breast Cancer Research and
Treatment, 151, 89–97.
of small round tubules scattered around normal Pavlakis, K., Zoubouli, C., Liakakos, T., Messini, I.,
breast structures. Luminal spaces are opened with Keramopoullos, A., Athanassiadou, S., Kafousi, M.,
round contours and eosinophilic secretions with- & Stathopoulos, E. N. (2006). Myoepithelial cell cock-
out obvious stromal response. MGA can be dis- tail (P63+SMA) for the evaluation of sclerosing breast
lesions. Breast, 15, 705–712.
tinguished from SA by lacking myoepithelial Urban, J. A., & Adair, F. E. (1949). Sclerosing adenosis. S
cells, ER negativity, and S100 strong positivity Cancer, 2, 625–634.
with IHC. Visscher, D. W., Nassar, A., Degnim, A. C., Frost, M. H.,
Vierkant, R. A., Frank, R. D., Tarabishy, Y.,
Radisky, D. C., & Hartmann, L. C. (2014). Sclerosing
adenosis and risk of breast cancer. Breast Cancer
References and Further Reading Research and Treatment, 144, 205–212.
Winham, S. J., Mehner, C., Heinzen, E. P.,
Jensen, R. A., Page, D. L., Dupont, W. D., & Rogers, Broderick, B. T., Stallings-Mann, M., Nassar, A.,
L. W. (1989). Invasive breast cancer risk in Vierkant, R. A., Hoskin, T. L., Frank, R. D.,
women with sclerosing adenosis. Cancer, 64, Wang, C., Denison, L. A., Vachon, C. M.,
1977–1983. Frost, M. H., Hartmann, L. C., Aubrey Thompson,
Nassar, A., Hoskin, T. L., Stallings-Mann, M. L., E., Sherman, M. E., Visscher, D. W., Degnim, A. C.,
Degnim, A. C., Radisky, D. C., Frost, M. H., & Radisky, D. C. (2017). NanoString-based breast
Vierkant, R. A., Hartmann, L. C., & Visscher, D. W. cancer risk prediction for women with sclerosing
(2015). Ki-67 expression in sclerosing adenosis and adenosis. Breast Cancer Research and Treatment,
adjacent normal breast terminal ductal lobular units: 166, 641–650.
350 Sebaceous Carcinoma of the Breast
Macroscopy
Molecular Features
Differential Diagnosis
the reported cases is G2 or G3; until now there is
no case reported with well differentiated The main differential diagnosis encompasses a
(G1) morphology (Acosta et al. 2018; Maia and primary skin adnexal tumor with sebaceous mor-
Amendoeira 2018; Sakai et al. 2018; Martin et al. phology. The presence of in situ components
2017; Yamamoto et al. 2017; Švajdler et al. 2015; (DCIS, LN) in the tumor and the lack of connec-
Wachter et al. 2014; Carlucci et al. 2012; Müller tion to the skin of the breast are useful features,
et al. 2011; Murakami et al. 2009; Hisaoka et al. which favor a primary sebaceous breast carci-
2006; Varga et al. 2000; Propeck et al. 2000). noma. Additionally, the use of immunohisto-
chemistry (especially a positive AR reaction and
the negativity for S100 and GCDFP-15 can sup-
Immunophenotype port breast origin) (Table 1).
Further differential diagnosis is the different
Most studies confirm that sebaceous cells are usu- types of breast cancers; especially the spectrum
ally strongly positive for keratins, EMA, and of primary clear cell carcinomas needs to be
adipophilin. Recent case reports could addition- excluded. Apocrine breast carcinoma (▶ Apo-
ally show GATA 3 and AR positivity in the seba- crine Carcinoma), lipid-rich breast carcinoma,
ceous cells. Three cases analyzed mismatch repair and glycogen-rich breast carcinoma (▶ Glyco-
proteins showing preserved epitopes without any gen-Rich Clear Cell Carcinoma) are in the differ-
selective loss. Interestingly, GCDFP-15 and S100 ential diagnosis. Morphology (large lipid
are consequently negative in the sebaceous cells. droplets, water-clear cytoplasm, or eosinophilic
One case was shown to exhibit neuroendocrine cytoplasm) and positive special stains (granular
differentiation by synaptophysin positivity PAS stain and large fatty droplets with Sudan
(Hisaoka et al. 2006). Black and Oil Red O) favor another type of clear
Predictive markers were tested in a large subset cell breast carcinoma. Expansive growth in a lob-
of reported cases; most cases are hormone recep- ular pattern and the presence of a dual neoplastic
tor positive and HER2 negative, and three cases cell population of larger sebaceous cells and
were triple negative and two cases HER2 positive smaller cells of different type are suggestive of a
(Acosta et al. 2018; Maia and Amendoeira 2018; sebaceous carcinoma (Acosta et al. 2018; Maia
Sakai et al. 2018; Martin et al. 2017; Yamamoto and Amendoeira 2018; Murakami et al. 2009;
et al. 2017; Švajdler et al. 2015; Wachter et al. Hisaoka et al. 2006; Varga et al. 2000).
Sebaceous Carcinoma of the Breast, Table 1 Summary of reported cases
Tumor
Number Age size Nodal Histological ER/
Publication of cases (years) (in mm) status grade PR HER2 IHC positive IHC negative other Follow-up
Acosta 1 51 20 mm pNO 2 +/+ Neg AR BRCA2+ NA
(2018) MMRP
Maia 2(1) 65 70 mm pN1(1/2) NA Neg/ Neg AR Died 9 months after
(2018) neg MMRP initial diagnosis
Sebaceous Carcinoma of the Breast
GATA 3
Maia 2 71 37 mm pNO NA +/ Neg GATA3 AR 100 months uneventful
(2018) neg MMRP
Sakai 2018 1 74 23 mm NA G2 +/+ Neg AR Mammoglobin NA
GATA3
Adipophylin
Martin 1 59 NA NA G2 +/+ Neg EMA Eyelid metastasis
2017 CK7 2 years after initial
CAM2 diagnosis
Yamamoto 1 80 35 mm NA NA Neg/ Neg EMA AR NA
2017 neg BerEp4
Adipophylin
Svajdler 4(1) 65 16 mm pN1(1/1) G3 +/+ Neg EMA S100 27 months uneventful
2015
Svajdler (2) 61 17 mm pN1(2/5) G3 Neg/ Neg EMA Died after 28 months
2015 neg
Svajdler 3 66 30 mm pN1(1/10) G2 +/+ Neg GCDFP-15 70 months uneventful
2015 EMA
S100
Svajdler 4 25 NA NA G3 +/+ Neg EMA GCDFP-15 75 months uneventful
2015 S100
Wachter 1 53 12 mm pNO G3 +/+ Neg GATA3 NA
2014 MMRP
(continued)
353
S
354
regional lymph nodes as emboli via lymphatics pN1(sn), or pN2(sn) TNM categories denote path-
reach the sentinel node(s) (first echelon nodes) ological nodal stage categories established with
first and nonsentinel nodes (second or further sentinel node biopsy.
echelon nodes) only after. This explains why the Nodal staging of clinically node-negative
sentinel nodes are the most likely site of nodal (cN0) early breast cancer patients with sentinel
metastasis, and also suggests that if they are not node biopsy followed a stepwise evolution
involved by metastatic disease, the rest of the reflected in the surgical consequences of finding
lymph nodes can also be considered free of metas- metastases in sentinel nodes. The concept of sen-
tases. Therefore the sentinel nodes “guard” the tinel nodes was validated in different clinical tri-
regional nodal basin, what explains their name. als, where lymphatic mapping was followed by
The kinetics of tumor cell migration from complete axillary lymph node dissection. These
the primary site to the sentinel nodes also apply studies proved that the sentinel nodes are really
to tracer molecules which travel via lymphatics, more likely to harbor metastasis than nonsentinel
and first accumulate in the sentinel lymph nodes. nodes. As sentinel nodes were often more thor-
Vital dyes (such as patent blue, isosulfan blue, or oughly analyzed than the remaining of the lymph
isocyanine green), 99-meta-technecium labeled nodes, a few validation studies have also sub-
sulfur or albumin colloids, or super paramagnetic jected nonsentinel nodes to the same scrutiny as
iron oxide nanoparticles all use the same path, the sentinel nodes, and these studies also rein-
i.e. the lymphatic vessels to reach and selectively forced the theory that sentinel nodes are more
label the sentinel nodes. Depending on the tracers often involved by metastases (Turner et al. 1997;
used, the sentinel nodes can be selectively Weaver et al. 2000). Following these initial vali-
removed after being detected by their color, dation studies and early learning periods (all com-
(probe-detected) radioactivity or magnetism. pleted with axillary nodal dissection), the scenario
Identification of the sentinel nodes with such of performing axillary lymph node dissection (as
tracers is called lymphatic mapping, and their a surgical treatment of the axilla) only in patients
removal sentinel (lymph) node biopsy or sentinel with positive (i.e., metastatic) sentinel lymph
lymphadenectomy. nodes became the rule. Patients with negative
Lymphatic mapping studies have revealed sentinel nodes are spared from axillary clearance
that most sentinel nodes are located in the lower and its potential morbidity (lymphedema, motor
axilla, but some can be located at higher levels of dysfunction, sensory loss), although it must be
the axilla or at extra-axillary sites like the sub- admitted that sentinel node biopsy can also have
clavicular or supraclavicular regions, the breast complications, but less frequently and to a lesser
itself (intramammary sentinel nodes), or along degree. This new era of axillary nodal staging has
the internal mammary artery (internal mammary also initiated a novel approach to teaching sentinel
or parasternal sentinel nodes). The theoretical def- node biopsy: supervision by surgeons already
inition of the sentinel node given at the beginning trained in the procedure has become general
of the present description can be complemented and no complete axillary dissection was mandated
with a second practical one: the sentinel nodes are any more to prove proficiency in removing the
the ones that are identified as such during lym- proper lymph nodes. As the decision to clear the
phatic mapping. Their number is highly variable, axilla or not was dependent on the status of the
but is generally 1–3. If the number of sentinel sentinel nodes, intraoperative assessment has
nodes is consistently higher than this, one must gained emphasis similarly to preoperative staging
doubt about the adequacy of lymphatic mapping; by axillary ultrasound. Patients with negative axil-
the American Joint Committee on Cancer has lary palpation and axillary ultrasound findings
adopted a rule of allowing the (sn) classifier in (cN0 patients) are offered lymphatic mapping
the Tumor Node Metastasis (TNM) classification and sentinel node biopsy, whereas those with sus-
only for breast cancers where the number of picious findings have ultrasound-guided sampling
sentinel nodes does not exceed 6. The pN0(sn), (mostly fine needle aspiration, but sometimes core
Sentinel Node 357
needle biopsy) of the suspicious nodes or the most comparable with that of sentinel
suspicious one. The microscopic evidence of lymphadenectomy of cN0 patient not requiring
nodal involvement excludes patients from lym- neoadjuvant treatment (Geng et al. 2016). It is
phatic mapping, as the procedure is advocated therefore common to perform sentinel node
for cN0 patients. Later, a few studies explored biopsy after the completion of primary systemic
the idea that once nodal positivity was proven treatment, but the procedure has also been
by means of sentinel node biopsy, there was no performed before. For patients with initially
more need for axillary lymph node dissection in node-positive disease, the false-negative rates
the majority of patients. The American College have been found to be somewhat higher, but
of Surgeons Oncology Group (ACOSOG) trial with at least two sentinel nodes identified, and
Z-0011 concluded that patients with limited nodal adequate patient selection, post-neoadjuvant ther-
involvement (up to two metastatic lymph nodes apy sentinel node biopsy has also been found
independently of the size of the metastasis) under- reliable enough (El Hage Chehade et al. 2016;
going breast conserving surgery complemented van Nijnatten et al. 2015).
with whole breast irradiation needed no axillary Most surgeons remove only axillary sentinel
nodal dissection (Giuliano et al. 2017). The 6-year nodes and do not search for extra-axillary ones.
results of the European Organization for Research The latter, to be known about, require lympho-
and Treatment of Cancer (EORTC) After Map- scintigraphy, which visualizes them. Even if they
ping of the Axilla: Radiotherapy Or Surgery? are evidenced by lymphoscintigraphy, internal
(AMAROS) trial (Donker et al. 2014) and the mammary sentinel nodes are rarely removed by
8-year results of the Hungarian National Institute surgeons, despite the fact that reports indicate that
of Oncology Optimal Treatment of the Axilla: metastatic involvement of a parasternal sentinel
Surgery Or Radiotherapy (OTOASOR) trial node may impact on the planning of radiotherapy
(Sávolt et al. 2017) both concluded that axillary and the indication of systemic therapy. As a rule,
irradiation was as effective as axillary lymph node internal mammary sentinel nodes are smaller than
dissection in preventing regional recurrences in axillary ones, often less than 5 mm in size, and
patients with metastatic sentinel nodes. These commonly they are identified by gamma-probes
results have also led to a change in policy: at only on the basis of their radioactivity, and do not
present many patients with positive sentinel label with dyes.
nodes do not have an axillary lymph node dissec- Pathological assessment of sentinel nodes
tion (Lyman et al. 2016). This change had obvi- can be divided into two broad categories: the
ously resulted in a decrease in intraoperative intraoperative and the postoperative setting. As
examinations of the sentinel nodes, as the result concerns the intraoperative assessment, imprint
had no impact on subsequent surgery. (or scrape) cytology, frozen section histology,
Another setting of applying sentinel node and molecular analysis of sentinel nodes have all S
biopsy as a staging procedure with lower morbidity been in use to assess metastatic involvement of the
is its use after primary systemic (neoadjuvant) sentinel nodes. Imprint cytology is based on the
treatment of locally advanced breast cancer. Neo- exfoliation of tumor cells from the lymph node
adjuvant therapy is used for both clinically node- freshly cut surface to the glass slides. This spon-
negative and node-positive patients. In the second taneous detachment can be enhanced by scraping
set, it is widely accepted that sentinel node biopsy the cut surface and smearing the cells on the
should be offered only to patients who become slides. Different stains have been described and
clinically node-negative after systemic treatment used (Figs. 1 and 2).
(ycN0). The false-negative rate (the proportion The advantages of intraoperative cytology
of cases without sentinel node involvement but specimens include fast preparation, relatively
with metastasis in nonsentinel nodes) of sentinel low costs, lack of tissue loss during the procedure,
lymphadenectomy after neoadjuvant treatment of and a microscopic verification of malignant cell
initially cN0 patients has been found to be morphology. Its disadvantages include the
358 Sentinel Node
different methods. It is accepted that different detecting these metastases (Giuliano et al. 1995;
parts of the sentinel nodes have different risks of Cserni et al. 2003). As a consequence, the average
being involved by the metastatic process, i.e., the size of nodal tumor deposits became also smaller.
areas at the junction of the tumor draining afferent Micrometastases (earlier defined by an upper
lymphatic vessels are involved at a much higher inclusive size limit of 2 mm) were identified at
frequency than areas away from this point. This is much higher rates than before the era of lymphatic
why, the best comparisons were made when alter- mapping. This led to a phenomenon called stage
nating slices of the lymph nodes were subjected to migration: the same patients who were classified
molecular and microscopic examinations. The as having node-negative disease after a conven-
comparison of larger series suggests that molecu- tional nodal examination following axillary
lar methods are substantially more sensitive in lymph node dissection could be identified as
detecting micrometastatic involvement of the sen- being node-positive by detecting tiny foci of met-
tinel nodes than either frozen sections or imprint astatic disease in the better investigated sentinel
cytology, but when it comes to macrometastases, nodes that the conventional evaluation had no
frozen sections are nearly as sensitive as molecu- chance to visualize. To limit stage migration, stag-
lar methods (Cserni 2012). To allow a nearly ing authorities have introduced a lower non-
complete assessment, many laboratories using a inclusive limit of 0.2 mm (and later an additional
molecular intraoperative assay use most if not all cell count limit of 200 for discohesive lobular
nodal tissue to adequately sample the sentinel carcinoma cells) to the definition of micro-
nodes, and this leaves no tissue for a morpholog- metastasis and have considered anything smaller
ical evaluation. A common recommendation pro- than that as isolated tumors cells or clusters (ITC)
poses to use at least one slice of tissue (generally to be staged as part of the node-negative set of
the central one) for histology and use only the patients (Figs. 3 and 4).
remaining parts of the sentinel node for the molec- At the beginning, the definitions of isolated
ular assay. Recommendations are in agreement tumor cells were not straight forward and uniform,
that intraoperative assessment of any type should and this has limited the proper prognostic evalua-
be restricted to cases where it has immediate tion of this subcategory, and its discrimination
influence on the intervention to follow (Wells from micrometastasis. At present, it seems that
et al. 2012). neither isolated tumor cells or clusters nor micro-
The traditional evaluation of sentinel nodes, metastases have major impact on breast cancer
which has been used as a gold standard for com-
parison with intraoperative examinations, is his-
topathological assessment. As the most likely
sites of nodal involvement, sentinel nodes have
been given more scrutiny: gross slicing, serial S
sectioning at equidistant steps (including step sec-
tioning till the extinction of the tissue blocks), and
immunohistochemistry have been used, often in
combination. This approach led to detection of
metastatic nodal involvement at a higher rate
than the previous conventional approach of
assessing one hematoxylin and eosin stained
slide of each lymph node or of each slice of larger
lymph nodes divided into pieces. Increased detec-
tion was partly due to the fact that sentinel nodes
Sentinel Node, Fig. 3 Isolated tumor cells of lobular
really harbor metastases more often than other
carcinoma detected by cytokeratin immunohistochemistry
lymph nodes of the same nodal basin, but also (Cytokeratin AE1/AE3 immunohistochemistry, original
due to the use of more sensitive techniques of magnification 400)
360 Sentinel Node
patients and therefore should not initiate any epithelial can lead to erroneous interpretation in
therapeutic intervention on their own. This clari- any intraoperative test.
fication of the prognostic impact of low-volume Endosalpingiosis of axillary sentinel nodes has
tumor deposits in sentinel nodes discovered by also been reported. Epithelial cells or structures
enhanced pathological evaluation has resulted like glands may be lodged artifactually during
in recommendations proposing not to search for tissue processing into holes without tissue
micrometastases and smaller volume involvement, (cracks) on the sections (Fig. 7).
but to devise assessment protocols that identify Similarly, epithelial displacement by needling
virtually all macrometastases (Cserni 2012). procedures (e.g., core needle biopsy, wire locali-
It must be kept in mind that sentinel nodes zation of the primary tumor, or intratumoral injec-
(as any other lymph nodes) may harbor a number tion of the tracer) or massage used to help tracer
of other changes or diseases than metastasis from migration during lymphatic mapping can lead to
breast cancer. This has often been an argument the misdiagnosis of displaced epithelium as a real
against the use of all nodal tissue for molecular metastasis. Papillary lesions have been reported to
analysis. Capsular nevi are a relatively common be especially prone for misplacement: most
finding, and generally do not pose a major differ-
ential diagnostic problem. Often they do not show
melanin content (Fig. 5).
On occasions they may involve the trabeculae
and even the nodal parenchyma when the differ-
ential diagnostic concern may be greater. Being
completely different in nature, they can easily
be clarified by their positivity for melanocytic
markers of nevi and their negativity for epithelial
markers. Involvement by other neoplastic (com-
monly chronic lymphoid leukemia, less commonly
other lymphomas or metastatic melanoma, etc.)
or infectious diseases (e.g., cat scratch disease
or toxoplasmosis) can also be encountered. Epi-
thelial inclusions (often of mammary type) Sentinel Node, Fig. 5 Capsular nevus without
(Fig. 6) are not very common, but by being melanin content. (H&E, original magnification 400)
van Nijnatten, T. J., Schipper, R. J., Lobbes, M. B., solid nodules of neoplastic cells arranged around
Nelemans, P. J., Beets-Tan, R. G., & Smidt, M. L. multiple fibrovascular spaces. Despite the lack of
(2015). The diagnostic performance of sentinel lymph
node biopsy in pathologically confirmed node positive the external myoepithelial layer, these lesions are
breast cancer patients after neoadjuvant systemic ther- currently considered as noninvasive (Lakhani et al.
apy: A systematic review and meta-analysis. European 2012). However, cases of true invasive SPC exist.
Journal of Surgical Oncology, 41, 1278–1287.
Weaver, D. L., Krag, D. N., Ashikaga, T., Harlow, S. P., &
O’Connell, M. (2000). Pathologic analysis of sentinel
and nonsentinel lymph nodes in breast carcinoma: a Clinical Features
multicenter study. Cancer, 88, 1099–1107.
Wells, C. A., Amendoeira, I., Bellocq, J. P., Bianchi, S., • Incidence
Boecker, W., Borisch, B., Bruun Rasmussen, B., Callagy,
G. M., Chmielik, E., Cordoba, A., Cserni, G., Decker, T., SPC is a rare entity, accounting for 1–2% of
DeGaetano, J., Drijkoningen, M., Ellis, I. O., Faverly, breast carcinomas (BCs) (Otsuki et al. 2007).
D. R., Foschini, M. P., Frković-Grazio, S., Grabau, D., • Age
Heikkilä, P., Iacovou, E., Jacquemier, J., Kaya, H., Kulka, It usually affects women in the seventh decade,
J., Lacerda, M., Liepniece-Karele, I., Martinez-Penuela,-
J., Quinn, C. M., Rank, F., Regitnig, P., Reiner-Concin, with a mean age of 73.2 years (Maluf and
A., Sapino, A., Tot, T., Van Diest, P. J., Varga, Z., Koerner 1995).
Wesseling, J., Zolota, V., & Zozaya-Alvarez, E. (2012). • Sex
S2: Pathology update. Quality assurance guidelines for It is more frequently observed in female
pathology. In N. Perry, M. Broeders, C. de Wolf,
S. Törnberg, R. Holland, & L. von Karsa (Eds.), patients, but it has been sporadically described
European guidelines for quality assurance in breast can- in men (Nassar et al. 2006).
cer screening and diagnosis. Fourth edition, supplements • Side
(pp. 73–120). Luxembourg: European Commission, SPC is typically unifocal and localized in the
Office for Official Publications of the European Union.
central region of the breast (Guo et al. 2016),
although a single case of bilateral synchronous
SPC has been reported in the literature
(Yoshimura et al. 2013).
Solid Papillary Carcinoma • Clinical presentation
The clinical presentation does not differ from
Elena Vissio2, Caterina Marchiò1,2,3 and Anna the other BC histological types, being usually
Sapino1,2 diagnosed after the detection of a mammo-
1
Unit of Pathology, Candiolo Cancer Institute graphic density or a palpable mass. In
FPO-IRCCS, Candiolo, Italy 20–25% of cases, a bloody nipple discharge
2
Department of Medical Sciences, University of may occur (Lakhani et al. 2012).
Turin, Turin, Italy • Treatment
3
Institut Curie, Paris, France Mastectomy or conservative surgery represents
the usual treatment for these tumors. However,
since the risk of recurrence is not related to the
Synonyms surgical technique, mastectomy, as well as
lymph node excision, seems to represent an
Solid Papillary Neuroendocrine Breast Carcinoma overtreatment for small tumors with no signs
of invasion (Guo et al. 2016). There is no
agreement on the necessity of sentinel lymph
Definition node biopsy for these patients. Regarding adju-
vant treatment, the benefit of radiation and
Solid papillary carcinoma (SPC) is described in the hormone therapy is still debated.
2012 WHO classification within the group of the • Outcome
intraductal papillary lesions and is defined as a var- SPCs present an excellent prognosis. Lymph
iant of papillary carcinoma characterized by multiple node and distant metastasis are rare. Local
Solid Papillary Carcinoma 363
Solid Papillary Carcinoma, Fig. 1 (a) Solid (b) calponin immunohistochemical reaction shows focal
papillary carcinoma is characterized by a dense neoplastic lack of myoepithelial peripheral cell layer
proliferation featuring well-defined borders (H&E);
recurrences have been reported, even if appear well defined, the myoepithelial peripheral
uncommon, and death for SPC should be con- layer is frequently absent at immunohistochemi-
sidered exceptional (Guo et al. 2016). cal analyses (Fig. 1a, b). To date, it is still debated
Recently, the 21-gene recurrence score if SPC should be considered as a low-grade inva-
(RS) multigene assay was performed in sive carcinoma or a variant of ductal carcinoma in
5 cases of SPC, and the RS resulted low in situ (▶ Ductal Carcinoma In Situ). Nonetheless,
3 cases and intermediate in the remaining because of their generally favorable clinical
2 cases, confirming the good prognosis of this course, the current WHO classification recom-
histological type. The only woman who pre- mends to consider SPCs as in situ lesions
sented a local recurrence showed a RS of (Lakhani et al. 2012). By contrast, irregular mar-
25 (Turashvili et al. 2017). gins and multiple small nests arranged in a jigsaw
When SPCs are associated to an invasive car- pattern represent typical features that suggest
cinoma, the outcome is related to the features invasive growth (Lakhani et al. 2012).
of the infiltrative component (Lakhani et al. Inside the nodules, neoplastic cells are densely
2012; Tan et al. 2016). organized. They may form typical peripheral
palisading around delicate fibrovascular struc-
tures (Fig. 2a). Cells usually present small dimen-
Macroscopy sions, hyperchromatic nuclei, granular S
cytoplasmic pattern, and occasionally spindle
cell features (Lakhani et al. 2012). They are gen-
At macroscopic examination, SPC usually
erally low- or intermediate-grade lesions, with
appears as a well-circumscribed lobulated mass,
low mitotic activity (Guo et al. 2016). SPC may
characterized by a whitish-gray or yellowish-
produce extracellular and/or intracellular mucin
brown color (Lakhani et al. 2012). The size may
(Wei 2016), and neuroendocrine differentiation
be variable, with a reported average diameter of
is reported in about half of cases (Fig. 2b)
2.5 cm (Guo et al. 2016).
(Otsuki et al. 2007).
SPCs may present foci of invasive carcinoma,
Microscopy frequently characterized by neuroendocrine or
mucinous features (Nassar et al. 2006; Otsuki
At low magnification, SPC is characterized by et al. 2007), even if other histotypes have
multiple cellular nodules. Although its borders been described. Invasive mucinous carcinomas
364 Solid Papillary Carcinoma
Solid Papillary Carcinoma, Fig. 2 (a) Densely (b) SPCs may show neuroendocrine differentiation
organized neoplastic cells form a peripheral palisading (immunohistochemical reaction with anti-synaptophysin
layer around delicate fibrovascular structures (H&E). antibody)
associated with SPCs are usually classified as weight CKs, such as CK8 and CK18, are fre-
type B, characterized by high cellularity, lower quently positive (Lakhani et al. 2012).
mucin production, and neuroendocrine differenti- Synaptophysin (Fig. 2b) and/or chromogranin,
ation (Nassar et al. 2006) (▶ Invasive Mucinous considered typical neuroendocrine markers, is
Carcinoma). expressed in more than 50% of cases (Guo et al.
Mixed cases, where SPC and encapsulated 2016; Lakhani et al. 2012; Otsuki et al. 2007).
papillary carcinoma (EPC) (▶ Encapsulated Pap-
illary Carcinoma) coexist, have been reported,
hypothesizing a common carcinogenetic process Molecular Features
for the two tumors (Cui and Wei 2015; Duprez
et al. 2012). Copy number aberration patterns are similar
between papillary carcinomas (PCs) and ER+
invasive carcinomas of no special type of
Immunophenotype similar grade (Duprez et al. 2012). However,
gene expression analysis showed a down-
Similarly to other papillary tumors, SPC is com- regulation of genes related to cell migration, pro-
monly classified as Luminal A type, showing an liferation, assembly, and organization in PCs,
intensely positive expression of estrogen receptor whereas genes involved in cellular homeostasis
(ER) and progesterone receptor (PR), lack of and angiogenesis were upregulated (Piscuoglio
HER2 protein overexpression, and a low prolifer- et al. 2014).
ation activity (Guo et al. 2016; Otsuki et al. 2007), Among the main types of PCs (SPC, EPC, and
and a similar immunohistochemical pattern is fre- invasive papillary carcinoma), no differences in
quently reported also for the possibly associated copy number aberrations were noted (Piscuoglio
invasive component (Nassar et al. 2006). et al. 2014). On the other hand, SPC, when com-
Nonetheless, the PAM50 gene signature pared with EPC, presents a higher expression of
applied on four cases of SPCs assigned them to neuroendocrine-related genes (e.g., RET, ASCL1,
the Luminal B subtype (Piscuoglio et al. 2014). and DOK7), whereas EPC shows down-
Myoepithelial markers, like p63 and high regulation of genes involved in cell migration,
molecular weight cytokeratins (CK), may show possibly explaining the histological differences
positive or negative result by immunohistochem- between these two entities (Piscuoglio
ical staining (Ni and Tse 2016). Low molecular et al. 2014).
Syringomatous Tumor of the Nipple 365
Differential Diagnosis Otsuki, Y., Yamada, M., Shimizu, S., Suwa, K.,
Yoshida, M., Tanioka, F., Ogawa, H., et al. (2007).
Solid-papillary carcinoma of the breast: Clinicopatho-
Intraductal papilloma (▶ Intraductal Papilloma) logical study of 20 cases. Pathology International, 57,
and florid usual ductal hyperplasia (▶ Usual Duc- 421–429.
tal Hyperplasia (UDH)) may present similar fea- Piscuoglio, S., Ng, C. K. Y., Martelotto, L. G., Eberle,
tures with SPC, but they usually present no C. A., Cowell, C. F., Natrajan, R., Bidard, F. C., et al.
(2014). Integrative genomic and transcriptomic charac-
mitoses and do not show neuroendocrine or terization of papillary carcinomas of the breast. Molec-
mucinous differentiation. Furthermore, in benign ular Oncology, 8, 1588–1602.
lesions, CK5/6 is usually intensely positive; there- Rabban, J. T., Koerner, F. C., & Lerwill, M. F. (2006). Solid
fore, its evaluation may be helpful in challenging papillary ductal carcinoma in situ versus usual ductal
hyperplasia in the breast: A potentially difficult distinc-
cases (Lakhani et al. 2012; Rabban et al. 2006). tion resolved by cytokeratin 5/6. Human Pathology,
In case ER and/or PR expression are unusually 37, 787–793.
weak or absent, metastatic neuroendocrine tumors Tan, B. Y., Thike, A. A., Ellis, I. O., & Tan, P. H. (2016).
to the breast should be taken into account, and the Clinicopathologic characteristics of solid papillary car-
cinoma of the breast. American Journal of Surgical
clinical history of the patient should be accurately Pathology, 40, 1334–1342.
reviewed in order to identify the primary malig- Turashvili, G., Brogi, E., Morrow, M., Hudis, C.,
nancy (Burt et al. 2016). Dickler, M., Norton, L., & Wen, H. Y. (2017). The
21-gene recurrence score in special histologic subtypes
of breast cancer with favorable prognosis. Breast
Cancer Research and Treatment, 165, 65–76.
References and Further Reading Wei, S. (2016). Papillary lesions of the breast: An
update. Archives of Pathology & Laboratory Medicine,
Burt, M., Madan, R., & Fan, F. (2016). Metastatic 140, 628–643.
gastrinoma in the breast mimicking primary solid pap- Yoshimura, N., Murakami, S., Kaneko, M., Sakatani, A.,
illary carcinoma. Human Pathology, 56, 143–146. Hirabayashi, N., & Takiyama, W. (2013). Synchronous
Cui, X., & Wei, S. (2015). Composite encapsulated bilateral solid papillary carcinomas of the breast.
papillary carcinoma and solid papillary carcinoma. Case Reports in Surgery, 2013, 812129.
Pathology International, 65, 133–137.
Duprez, R., Wilkerson, P. M., Lacroix-Triki, M., Lambros,
M. B., MacKay, A., A’Hern, R., Gauthier, A., et al.
(2012). Immunophenotypic and genomic characteriza-
tion of papillary carcinomas of the breast. The Journal Syringomatous Tumor of the
of Pathology, 226, 427–441.
Guo, S., Wang, Y., Rohr, J., Fan, C., Li, Q., Li, X., &
Nipple
Wang, Z. (2016). Solid papillary carcinoma of the
breast: A special entity needs to be distinguished from Zsuzsanna Varga and Linda Moskovszky
conventional invasive carcinoma avoiding over- Institute of Pathology and Molecular Pathology,
treatment. The Breast, 26, 67–72.
University Hospital Zurich, Zurich, Switzerland
Lakhani, S. R., Ellis, I. O., Schnitt, S. J., Tan, P. H.,
van de Vijver, M. J. (2012). WHO classification S
of tumours of the breast. World Health Organization
classification of tumours. Lyon, IARC press. ISBN-10: Synonyms
9283224337.
Maluf, H. M., & Koerner, F. C. (1995). Solid papillary
carcinoma of the breast. A form of intraductal carci- Infiltrating syringomatous adenoma of the nipple;
noma with endocrine differentiation frequently associ- Syringomatous adenoma of the nipple
ated with mucinous carcinoma. The American Journal
of Surgical Pathology, 19, 1237–1244.
Nassar, H., Qureshi, H., Adsay, N. V., Volkanadsay, N., &
Visscher, D. (2006). Clinicopathologic analysis of solid Definition
papillary carcinoma of the breast and associated inva-
sive carcinomas. The American Journal of Surgical Syringomatous tumor of the nipple/areola region
Pathology, 30, 501–507.
(SAN) is a benign rare condition, which is typi-
Ni, Y. B., & Tse, G. M. (2016). Pathological criteria and
practical issues in papillary lesions of the breast – cally observed in the dermal/subcutaneous region
A review. Histopathology, 68, 22–32. of the nipple area and which exhibits sweat gland
366 Syringomatous Tumor of the Nipple
differentiation (Ishikawa et al. 2015; Montgomery (Ishikawa et al. 2015; Montgomery et al. 2014;
et al. 2014; Boecker et al. 2014). These tumors Oo and Xiao 2009; Page et al. 2009).
have no metastastic potential but can recur if • Treatment
removed incompletely (Ishikawa et al. 2015; Therapy is surgical excision with clear mar-
Montgomery et al. 2014; Boecker et al. 2014). gins. If the tumor is close to the nipple and a
complete excision is therefore not possible, in
such cases, a thorough postoperative follow-up
Clinical Features with imaging is recommended (Ishikawa et al.
2015; Oo and Xiao 2009).
• Incidence • Outcome
This type of nipple tumor is very rare, account- Prognosis is favorable in all cases; until now, no
ing less than 1% of all tumors arising in the metastastic lesions and no death due to tumor
breast. Until now, 39 cases were reported in the progression were reported. However, local
English literature, mostly in form of case tumor recurrence in a small subset of patients
reports. Thirty seven patients were female, in the literature was reported mainly due to
and two were males (Ishikawa et al. 2015; incomplete resection (Ishikawa et al. 2015;
Montgomery et al. 2014; Oo and Xiao 2009; Montgomery et al. 2014; Boecker et al. 2014;
Page et al. 2009; Jones et al. 1989; Carter and Oo and Xiao 2009; Page et al. 2009; Jones et al.
Dyess 2004; Rosen 1983). The original 1989; Carter and Dyess 2004).
description of nipple syringomatous adenoma
was published by PP Rosen in 1983. Since this
Macroscopy
description, this entity has been occasionally
addressed in reviews on diagnostic challenges
Syringomatous tumor usually presents as a solid
and immunohistochemical phenotypes as well
mass or ill-defined tumor in the dermis of the
as reported in the above mentioned case report
nipple. Rarely the lesion is detected due to
series in different journals (Ishikawa et al.
mammographic calcifications in the surrounding
2015; Montgomery et al. 2014; Boecker et al.
breast. Size of the lesion is variable, generally
2014; Oo and Xiao 2009; Page et al. 2009;
between 5 mm and 50 mm in diameter, with an
Jones et al. 1989; Carter and Dyess 2004).
average size of 17.7 mm in largest diameter
• Age
(Ishikawa et al. 2015; Montgomery et al. 2014;
Patients are mostly peri-/postmenopausal at the
Oo and Xiao 2009).
time of the diagnosis; however, one case was
described in a young prepubertal 11-year-old
girl occurring in a supernumerary nipple, and
one elderly patient of 87 years was reported Microscopy
with this entity (Oo and Xiao 2009; Page
et al. 2009). Syringomatous tumor of the nipple has an appar-
• Sex ent infiltrative growth in most cases arising in
Thirty-seven patients were female, and two a recognizable lobular growth pattern with nests
patients were males (Ishikawa et al. 2015; and branching cords of inconspicuous epithelial
Montgomery et al. 2014; Oo and Xiao 2009). cells situated in the dermis and in the subcutis of
• Site the nipple or areola (Ishikawa et al. 2015) (Fig. 1).
Both sides are equally affected and most The tumor cells typically show an infiltrative
lesions are unifocal. Two papers reported growth between the stroma and the smooth mus-
a syringomatous tumor arising in a supernu- cle cells of the dermis and subcutis (Fig. 2)
merary breast, each in one patient; two further (Ishikawa et al. 2015; Montgomery et al. 2014).
papers, each of them described one female Typically, there is no relationship between the
patient with bilateral syringomatous adenoma tumor and the overlying skin or the nipple
Syringomatous Tumor of the Nipple 367
Syringomatous Tumor of the Nipple, Fig. 1 Low Syringomatous Tumor of the Nipple, Fig. 3 High
power appearance of a syringomatous tumor containing power appearance of a syringomatous tumor showing infil-
infiltrative bland solid epithelial formations in the dermis trative bland small epithelial formations surrounded by
of the skin in the vicinity of thin epithelial cysts (H&E, low smooth muscles and vessels of the dermal-subcutaneous
magnification) junction without any significant stromal desmoplasia
(H&E, high magnification)
subset with squamoid differentiation, also for p63. negative in both components. Ki67 index is usually
The outer layer is consequently positive for very low (<5%) (Ishikawa et al. 2015; Montgomery
myoepithelial markers as p63 (Fig. 4), CK5/6 et al. 2014; Boecker et al. 2014).
(Fig. 5), or smooth muscle myosin heavy chain
and also for high molecular weight cytokeratins as
34betaE12. Hormone receptors and HER2 are Molecular Features
Differential Diagnosis
Syringomatous Tumor of the Nipple, note also the vicinity of the cystic protruding epidermal
Fig. 5 Cytokeratin 5/6 immunohistochemistry decorates structures (cytokeratin 5/6 immunohistochemistry, clone
both the inner epithelial and the outer myoepithelial layer, D5/16B4, high magnification)
Syringomatous Tumor of the Nipple 369
Macroscopy
Molecular Feature
Tall Cell Variant of Papillary Breast Carcinoma, RET/PTC and BRAF genes alterations commonly
Fig. 3 The neoplastic cells are columnar to cuboidal in
shape, with granular eosinophilic cytoplasm. The nucleus seen in papillary thyroid carcinomas have been
occupies about 1/3 of the cell height and it is centrally explored, with negative results (Cameselle-
located, elongated and has clear chromatin. Nuclear Teijeiro et al. 2006; Eusebi et al. 2003; Hameed
grooves and pseudoinclusions are present (H&E) et al. 2009). Mutations in isocitrate dehydroge-
nase 2 (IDH2) have been reported. In particular,
morphological and immunohistochemical features Chiang et al. (2016) detected hotspot mutations at
with the primary neoplasm. R172 IDH2 gene in 10 out of 13 cases of tall cell
variant of papillary breast carcinomas. Eight of
the 10 mutated cases displayed concomitant path-
Immunophenotype ogenic mutations at PIK3CA or PIK3R1. In addi-
tion, the same authors, through functional studies,
Estrogen and progesterone receptors are demonstrated that IDH2 and PIK3CA hotspot
completely missing or rarely (7.6% of cases) mutations were crucial in the neoplastic transfor-
expressed in less than 1% of the total neoplastic mation of tall cell variant of papillary breast car-
proliferation and HER2 is negative, so that tall cinoma and probably responsible of its peculiar
cell variant of papillary breast carcinoma is phenotype.
classified as triple-negative. Androgen receptor
is negative as well excluding an apocrine nature.
Ki67 proliferative index is low. Differential Diagnosis
Cytokeratin 7 expression is strong and diffuse
within the neoplastic cells, while occasional Lymph node metastases of papillary thyroid car-
focal positivity for GCDFP-15, mammaglobin, cinoma have to be considered in the rare occa-
and GATA 3, which are “luminal” type markers, sion of metastatic tall cell papillary carcinoma of T
may be observed. On the other hand, immunocy- the breast. However, the site of metastases, the
tochemical expression of “nonluminal/basal-like” absence of history of a primary thyroid carci-
markers such as cytokeratin 14 and cytokeratin noma, the immunohistochemical expression of
5/6 has been reported in 8 cases (Eusebi et al. GCDFP-15, GATA3, and mammaglobin, and the
2003; Tosi et al. 2007; Foschini et al. 2017). negativity of markers of thyroid origin as TTF1
Myoepithelial markers such as p63, calponin, and thyroglobulin and the absence of common
and smooth muscle actin are negative or mark molecular alterations seen in papillary thyroid
rare myoepithelial cells that surround occasional carcinomas help distinguish between these two
neoplastic areas. entities. A possible differential diagnosis may
Collagen IVand laminin highlight the fibrovas- be required with the solid papillary variant of
cular cores present within the solid papillary nests breast carcinoma (▶ Solid Papillary Carcinoma).
374 Toker Cells of the Nipple
The latter, at difference with the tall cell variant K. Y., Jungbluth, A. A., Balss, J., Pusch, S., Baker, G.
of papillary breast carcinoma, is positive for M., Cole, K. S., von Deimling, A., Batten, J. M.,
Marotti, J. D., Soh, H-C., McCalip, B. L., Serrano, J.,
hormone receptor expression and frequently Lim, R. S., Siziopikou, K. P., Lu, S., Liu, X., Hammour,
exhibits neuroendocrine marker expression. T., Brogi, E., Snuderl, M., Iafrate, A. J., Reis-Filho, J.
S., Schnitt, S. J. (2016). IDH2 mutations define a
Tall cell variant of papillary breast carcinoma: fact unique subtype of breast cancer with altered nuclear
sheet polarity. Cancer Research, 76, 1–12.
Definition Elston, C. W., & Ellis, I. O. (1991). Pathological prognostic
– Very rare type of primary breast neoplasm not related factors in breast cancer. 1. The value of histological
to a thyroid gland neoplasm, showing very frequently a grade in breast cancer: Experience from a large
triple-negative profile study with long-term follow-up. Histopathology, 19,
Age distribution 403–410.
– Median age: 61 years (range 45–85 years) Eusebi, V., Damiani, S., Ellis, I. O., Azzopardi, J. G.,
Rosai, J. (2003). Breast tumor resembling the tall cell
Clinical features
variant of papillary thyroid carcinoma: Report of
– Palpable and well-defined nodules with regular
5 cases. The American Journal of Surgical Pathology,
margins
27, 1114–1118.
Microscopic findings Foschini, M. P., Asioli, S., Foreid, S., Cserni, G., Ellis, I.
– Three architectural patterns: papillary, solid, and O., Eusebi, V., Rosai, J. (2017). Solid papillary breast
follicular carcinomas resembling the tall cell variant of papillary
– Neoplastic cells: columnar to cuboidal in shape, with thyroid neoplasms: A unique invasive tumor with indo-
granular eosinophilic cytoplasm lent behavior. The American Journal of Surgical
– Presence of nuclear grooves, pseudoinclusions, and Pathology, 41, 887–895.
psammoma bodies Hameed, O., Perry, A., Banerjee, R., Zhu, X., Pfeifer, J. D.
– Mitosis is rare, and vascular invasion is uncommon (2009). Papillary carcinoma of the breast lacks evi-
Immunohistochemical findings dence of RET rearrangements despite morphological
– Estrogen, progesterone, androgen receptors, and similarities to papillary thyroid carcinoma. Modern
HER2: negative Pathology, 22, 1236–1242.
– “Luminal” type markers (cytokeratin 7, GCDFP-15, Tosi, A. L., Ragazzi, M., Asioli, S., Del Vecchio, M.,
mammaglobin, and GATA 3): variably positive Cavalieri, M., Eusebi, L. H. U., Foschini, M. P.
– Myoepithelial markers (p63, calponin, and smooth (2007). Breast tumor resembling the tall cell variant
muscle actin): negative of papillary thyroid carcinoma: Report of 4 cases with
– Thyroglobulin and TTF1: negative evidence of malignant potential. International Journal
Molecular findings of Surgical Pathology, 15, 14–19.
– Absence of alterations of RET/PTC and BRAF genes
– Mutations of IDH2 gene in 77% of cases tested
Differential diagnosis
– Solid papillary carcinoma of the breast
– Metastasis from papillary thyroid carcinomas Toker Cells of the Nipple
Prognosis and treatment
– Wide excision without aggressive treatment Luca Di Tommaso
strategies Pathology Unit, Humanitas Clinical and Research
– Indolent clinical behavior
Center, Department of Biomedical Sciences,
Humanitas University, Milan, Rozzano, Italy
Toker Cells of the Nipple, Fig. 1 (H&E) Toker cells higher magnification single elements and small glands are
(TC) of the nipple. (a) Occasional clear cells are seen in the characterized by bland cytological features
lower layers of the epidermis of the nipple (arrows); (b) at
Toker Cells of the Nipple, Fig. 2 (H&E) Hyperplastic TC. (a) Numerous TC are easily seen at low magnification; (b)
while most of TC are still devoid of atypia, scattered prominent nucleoli and irregular nuclear profile can be observed
Toker Cells of the Nipple, Fig. 3 Atypical TC. (a) The TC show one or two prominent eosinophilic nucleoli
basal layer of this nipple is characterized by the presence of (H&E) (c) and demonstrate a faint, continuous
several TC, isolated and in small clusters (H&E); (b) most immunostaining for HER2
Toker Cells of the Nipple, Fig. 4 Phenotypical features of TC. These hyperplastic TC show strong immunoreactivity to
estrogen receptors (a) and faint immunoreactivity to progesterone receptors (b)
confirmed by Lundquist in his autopsy series explanation on TC origin came from an embry-
(Lundquist et al. 1999). Subsequent studies ological study conducted on the anogenital
investigated the presence of TC in patients region of female and male fetus (van der
undergoing mastectomy for breast cancer: epi- Putte 2011). This study demonstrated that
demiological features of TC were confounded the primordial follicular cells involved in the
by those of the underlining neoplastic disease. formation of apocrine and mammary-like
Indeed, patients showing TC are significantly glands are displaced into the epidermis where
younger if undergoing prophylactic mastec- they proliferate and differentiate as
tomy for BRCA mutation rather than mastec- TC. Accordingly, it was speculated that TC of
tomy for breast cancer (Nofech-Mozes and the anogenital region may likely represent the
Hanna 2009). precursors of primary anogenital Paget disease. T
• Site and Origin Similarly, TC of the nipple may represent the
TC have been reported not only in the nipple precursors of primary Paget disease of the
but also in the areola, in the accessory nipple, nipple.
in the axilla, and in the vulva. Accordingly, it
has been suggested that TC represent (1) ele-
ments that migrated from mammary excretory Microscopy
ducts into the epidermis of the nipple, or
(2) abortive mammary differentiation within TC are cells larger than keratinocytes, round to
the basal layer of the epidermis, or (3) embry- polygonal in shape, with faintly eosinophilic or
onic nests of cells related to eccrine and apo- clear cytoplasm, oval nuclei, and small nucleoli.
crine structures. A possible comprehensive Electron microscopy showed that the clear
378 Toker Cells of the Nipple
Toker Cells of the Nipple, Table 1 Proposed criteria for the classification of normal, hyperplastic, and atypical TC
Normal TC Hyperplastic TC Atypical TC
Isolated TC (n ) <10 10–20 Any; usually 10–20
Clusters of TC (n ) <3 3–10 Any; usually 3–10
TC after recuts Disappear Conserved/increased Conserved/increased
Cytological atypia Absent Absent Mild to moderate
Toker Cells of the Nipple, Table 2 Phenotypical features of Toker cells (TC) and Paget disease (PD) cells
ER (%) PgR (%) HER2/neu (%) p53 (%)
Normal TC 100 70 0 na
Hyperplastic TC 100 90 10 0
Atypical TC 100 100 80 0
PD (without coexisting carcinoma) 50–60 20 70–100 80
PD (with coexisting carcinoma) 10–40 0–20 80–100 10–60
Abbreviation: ER estrogen receptors, PgR progesterone receptors, na not available, DCIS ductal carcinoma in situ
cytoplasm is due to paucity of organelles and their role in the differential diagnosis with ▶ Paget
filaments (Marucci et al. 2002). TC are seen Disease (PD). Hyperplastic and atypical TC are
within the basal and/or spinous layers; they pre- always p53 negative; a faint immunoreactivity for
sent as single elements or as small glands. Hyper- HER2 is frequently seen in atypical TC and, more
plastic TC retain cytological and architectural rarely, in hyperplastic conditions.
features of normal TC. The distinctive diagnostic Table 2 illustrates the immunohistochemical
feature is the increased number of single cells profile of TC (normal, hyperplastic, and atypical)
and/or glands within the lower layers of the epi- as compared to PD (without or with a coexisting
dermis, which makes these cases easily recogniz- carcinoma in the underlining breast parenchyma: for
able at H&E as compared to normal TC even at a detailed explanation, see Differential Diagnosis).
scanning magnification. Atypical TC are charac-
terized by nuclei with irregular contour and eosin-
ophilic nucleoli but with preserved nucleus/ Differential Diagnosis
cytoplasmic ratio. There are no signs of architec-
tural disorder, being most of the cell still localized As mentioned, the main differential diagnosis
in the lower layers of the epidermis. of TC is PD of the nipple. In most of the cases,
PD represents an epidermotropic migration of
cancer cells from an underlying breast carci-
Immunophenotype noma. However, the existence of PD with
extensive involvement of the epidermidis
TC are consistently immunoreactive with CK7. lacking any carcinoma in the parenchyma or
This marker, as well as EMA, can be used to that of PD in the areola in patient with con-
reveal TC or to disclose additional TC at the genital absence of the nipple does not support
periphery of those seen on H&E. Interestingly, the epidermotropic hypothesis. The few reports
CK7 also highlights the dendritic profile about PD associated with TC hyperplasia in the
of rare TC. TC are p63 negative/ER positive/ areola and that of PD indistinguishable from
PgR positive: a phenotype further reinforcing TC hyperplasia in supernumerary nipple fur-
the difference with keratinocytes and supporting ther reinforce the hypothesis that rare
a mammary or mammary-like origin. Additional cases of PD may represent a malignant
markers tested in TC, mostly in the hyperplastic transformation of TC, as suggested by Toker
and atypical groups, are p53 and HER2, due to himself (Toker 1970). This hypothesis was
Tubular Adenoma of the Breast 379
(Foschini et al. 2012) and vanishingly rare in in pregnant and puerperal women (Salemis
elderly people (Nishimori et al. 2000; Rovera et al. 2012).
et al. 2006). Pregnancy-associated tubular ade- The imaging features of breast tubular ade-
nomas can be observed and may show rapid noma are nondiagnostic and consistent to
enlargement in tumor size (Nagata et al. 1998; those of a benign breast lesion. At mammog-
O’Hara and Page 1985; Salemis et al. 2012). raphy, it appears as a well-circumscribed
• Sex fibroadenoma-like nodule (Fig. 1a). Calcifica-
Despite tubular adenoma has been described tions have been rarely observed, particularly
only in females, the possible occurrence in in elderly women (Soo et al. 2000; D’Orsi
male individuals should not be excluded. et al. 2013). Ultrasonography reveals a well-
• Site defined hypoechoic mass with a relatively
Akin to most benign breast lesions, tubular homogeneous internal texture, showing low-
adenomas arise within the terminal duct- to-mild posterior enhancement (Irshad et al.
lobular unit (Foschini et al. 2012; Sciarra 2008) (Fig. 1b).
et al. 2017). They preferentially involve the • Treatment
upper and outer quadrants of the breasts Given its benign clinicoradiological features
(Sengupta et al. 2014). and low tendency to increase in size, clinical
• Clinical Presentation observation usually suffices for the proper
Breast tubular adenoma is usually asymptom- management of these patients. However, in
atic and detected incidentally during screening high-risk individuals (e.g., elderly women and
examinations (Rovera et al. 2006). When pre- young women with strong family history for
sent, the symptoms are nonspecific, being breast cancer), or in cases of fast-growing
related to the mass effect. They include a pal- lesions with significant mass effect, a diagnos-
pable smooth, mobile, well-defined nodular tic core biopsy or surgical excision should be
mass and breast tenderness in the area of the performed (Salemis et al. 2012; Sengupta et al.
lump (Salemis et al. 2012; Sengupta et al. 2014; Rovera et al. 2006; Calderaro et al.
2014). These benign alterations cause no nip- 2010).
ple discharge or pain. Although the majority of • Outcome
tubular adenomas grow slowly, they can also Tubular adenoma of the breast has an indo-
present as a fast-growing nodule, particularly lent clinical course (Foschini et al. 2012).
breast tubular adenoma and cylindroma, the latter Schmid, K. W. (1992). An immunohistochemical
is consistently ER-negative and shows recurrent study of the breast using antibodies to basal and luminal
keratins, alpha-smooth muscle actin, vimentin, colla-
splice-site somatic mutations in the tumor sup- gen IV and laminin. Part II: Epitheliosis and ductal
pressor cylindromatosis gene (CYLD). carcinoma in situ. Virchows Archive. A, Pathological
The presence of luminal eosinophilic secre- Anatomy and Histology, 421, 323–330.
tions in the duct-like structures of tubular adeno- Case, T. C. (1977). Adenocarcinoma of breast arising in
adenoma. New York State Journal of Medicine, 77,
mas has allowed some authors to speculate on its 2122–2123.
possibly shared phylogenetic origin with lactating Calderaro, J., Bayou, E. H., Castaigne, D., Mathieu, M. C.,
adenoma (O’Hara and Page 1985). However, Andreiuolo, F., Suciu, V., Delaloge, S., Vielh, P. (2010).
based on their pathophysiology and morphology, Tubular adenoma of the breast with associated mucin-
ous features: A cytological diagnostic trap. Cytopathol-
these two benign lesions should be considered ogy, 21, 191–193.
discrete conditions. Indeed, lactating adenomas D’Orsi, C. J., Sickles, E. A., Mendelson, E. B., et al.
are hormone-related and tend to reflect lactational (2013). ACR BI-RADS ® atlas, breast imaging
alterations in hyperplastic and enlarged lobules reporting and data system. Reston: American College
of Radiology.
with variably complex architectural patterns. On Domoto, H., Tsuda, H., Miyakawa, K., Shinoda, A.,
the contrary, tubular adenomas can occur Nanasawa, T. (2002). Invasive ductal carcinoma asso-
irrespectively of the obstetric history of the ciated with tubular adenoma of the breast. Pathology
patients and may show superimposed lactational International, 52, 244–248.
Düşünceli, F., Manukyan, M. N., Midi, A., Deveci, U.,
changes. Furthermore, the tubular structures of Yener, N. (2012). Giant tubular adenoma of the breast:
breast tubular adenoma are characteristically A rare entity. Breast Journal, 18, 79–80.
small and organized in a uniform architecture. Foschini, M. P., Simpson, J. F., & O’Malley, F. (2012).
Differently from tubular adenomas, ductal ade- Benign epithelial proliferations. In S. R. Lakhani, I. O.
Ellis, S. J. Schnitt, P. H. Tan, & M. J. van de Vijver
nomas (▶ Ductal Adenoma) of the breast classi- (Eds.), World health organization classification of
cally display glands that vary in size, sclerosing tumours of the breast (p. 115). Lyon: IARC Press.
stroma, and a fibrous capsule. Despite they tend to Fusco, N., Colombo, P. E., Martelotto, L. G., De Filippo,
be localized lesions, they are not completely M. R., Piscuoglio, S., Ng, C. K., Lim, R. S., Jacot, W.,
Vincent-Salomon, A., Reis-Filho, J. S., Weigelt, B.
circumscribed nodules like tubular adenomas (2016). Resolving quandaries: Basaloid adenoid cys-
(Saimura et al. 2015). tic carcinoma or breast cylindroma? The role of mas-
The absence of compression and distortion of sively parallel sequencing. Histopathology, 68,
the ducts within the cellular stroma is a key mor- 262–271.
Guerini-Rocco, E., Piscuoglio, S., Ng, C. K., Geyer, F. C.,
phological feature that may help in the differenti- De Filippo, M. R., Eberle, C. A., Akram, M., Fusco, N.,
ation of tubular adenoma with sclerosing adenosis Ichihara, S., Sakr, R. A., Yatabe, Y., Vincent-Salomon,
(▶ Sclerosing Adenosis) and microglandular A., Rakha, E. A., Ellis, I. O., Wen, Y. H., Weigelt, B.,
adenosis (▶ Microglandular Adenosis), a rare Schnitt, S. J., & Reis-Filho, J. S. (2016). Micro-
glandular adenosis associated with triple-negative
proliferative lesion of the breast composed of breast cancer is a neoplastic lesion of triple-negative
small glands in a hypocellular stroma (Guerini- phenotype harbouring TP53 somatic mutations. The
Rocco et al. 2016; Foschini et al. 2012). The latter, Journal of Pathology, 238, 677–688. T
however, is a non-lobulocentric proliferation of Hertel, B. F., Zaloudek, C., & Kempson, R. L. (1976).
Breast adenomas. Cancer, 37, 2891–2905.
ER-negative glands lacking the myoepithelial Hill, R. P., & Miller, F. N. J. (1954). Adenomas of the
cell layer, while tubular adenoma ducts are com- breast with case report of carcinomatous transformation
posed of both epithelial and myoepithelial cells in an adenoma. Cancer, 7, 318–324.
and express ER. Irshad, A., Ackerman, S. J., Pope, T. L., Moses, C. K.,
Rumboldt, T., & Panzegrau, B. (2008). Rare breast
lesions: Correlation of imaging and histologic features
with WHO classification. Radiographics, 28,
1399–1414.
References and Further Reading Komaki, K., Morimoto, T., Mori, Sasa, M., Oshimo, K.,
Monden, Y., Hirose, T., & Hizawa, K. (1992). A rare
Böcker, W., Bier, B., Freytag, G., Brömmelkamp, B., case of fibroadenoma in a tubular adenoma of the
Jarasch, E. D., Edel, G., Dockhorn-Dworniczak, B., breast. Surgery Today, 22, 163–165.
384 Tubular Carcinoma
Maiorano, E., & Albrizio, M. (1995). Tubular adenoma of North American Journal of Medical Sciences, 6,
the breast: An immunohistochemical study of ten cases. 219–223.
Pathology Research and Practice, 191, 1222–1230. Soo, M. S., Dash, N., Bentley, R., Lee, L. H., & Nathan,
Moross, T., Lang, A. P., & Mahoney, L. (1983). Tubular G. (2000). Tubular adenomas of the breast: Imaging
adenoma of breast. Archives of Pathology & Labora- findings with histologic correlation. AJR The American
tory Medicine, 107, 84–86. Journal of Roentgenology, 174, 757–761.
Nagata, Y., Horimi, T., Ichikawa, J., Nishioka, Y., Tavassoli, F. A. (1999). Benign lesions. Pathology of the
Okabayashi, T., Inagaki, M., Okazaki Y., Noguchi, breast (pp. 115–204). New York: McGraw-Hill Profes-
H., nagano, K., Teraishi, H., Kotaka, M., Sasaoka, K. sional Publishing.
(1998). A case of rapidly enlarging tubular adenoma of Yeh, I. T., & Mies, C. (2008). Application of immunohis-
the breast occurred in a pregnant woman. Nihon Rinsho tochemistry to breast lesions. Archives of Pathology &
Geka Gakkai Zasshi (Journal of Japan Surgical Asso- Laboratory Medicine, 132, 349–358.
ciation), 59, 2764–2768.
Nishimori, H., Sasaki, M., Hirata, Zembutsu, H.,
Yasoshima, T., Fukui, R., Kobayashi, K. (2000). Tubu-
lar adenoma of the breast in a 73-year-old woman.
Breast Cancer, 7, 169–172. Tubular Carcinoma
O’Hara, M. F., & Page, D. L. (1985). Adenomas of the
breast and ectopic breast under lactational influences.
Emad Rakha
Human Pathology, 16, 707–712.
Persaud, V., Talerman, A., Jordan, R. (1968). Pure ade- Department of Histopathology, Division of
noma of the breast. Arch Pathol, 86, 481–3. Cancer and Stem Cells, University of
Piscuoglio, S., Murray, M., Fusco, N., Marchiò, C.,1, Loo, Nottingham, Nottingham City Hospital,
F. L., Martelotto, L. G., Schultheis, A. M., Akram, M.,
Nottingham, UK
Weigelt, B., Brogi, E., Reis-Filho, J. S. (2015). MED12
somatic mutations in fibroadenomas and phyllodes
tumours of the breast. Histopathology, 67, 719–729.
Rovera, F., Ferrari, A., Carcano, G., Dionigi, G., Synonyms
Cinquepalmi, L., Boni, L., Diurni, M., & Dionigi,
R. (2006). Tubular adenoma of the breast in an
84-year-old woman: Report of a case simulating breast Pure tubular carcinoma; Tubular carcinoma
cancer. Breast Journal, 12, 257–259.
Rosen, P. P. (2014). Fibroepithelial neoplasms. In S. A.
Hoda, P. P. Rosen, E. Brogi, & K. F.C. (Eds.), Rosen’s
breast pathology (pp. 187–229). Philadelphia: Definition
Lippincott Williams & Wilkins.
Shet, T. M., & Rege, J. D. (1998). Aspiration cytology of
A special type of breast carcinoma with a partic-
tubular adenomas of the breast. An analysis of eight
cases. Acta Cytologica, 42, 657–662. ularly favorable prognosis composed of well-
Saimura, M., Anan, K., Mitsuyama, S., Ono, M., & differentiated tubular structures with open lumina
Toyoshima, S. (2015). Ductal carcinoma in situ arising lined by a single layer of low-nuclear-grade neo-
in tubular adenoma of the breast. Breast Cancer, 22,
plastic epithelial cells.
428–431.
Sciarra, A., Lopez, G., Corti, C., Runza, L., Ercoli, G.,
Bonometti, A., Despini, L., Blundo, C., Gambini, D., &
Fusco, N. (2017). Columnar cell lesion and apocrine Clinical Features
hyperplasia of the breast: Is there a common origin?
The role of prolactin-induced protein. Applied Immu-
nohistochemistry & Molecular Morphology. https:// • Incidence
doi.org/10.1097/pai.0000000000000604. Published Pure tubular carcinoma accounts for approxi-
Ahead-of-Print. PubMed PMID: 00129039- mately 2% of invasive breast cancer in most
900000000-98888.
series. Higher frequencies are found in cohorts
Salemis, N. S., Gemenetzis, G., Karagkiouzis, G., Seretis,
C., Sapounas, K., Tsantilas, V., Sambaziotis, D., & enriched with small T1 breast cancers
Lagoudianakis, E. (2012). Tubular adenoma of the and in mammographic screening programs
breast: A rare presentation and review of the literature. (Anderson et al. 2004; Rakha et al. 2010).
Journal of Clinical Medicine Research, 4, 64–67.
• Age
Sengupta, S., Pal, S., Biswas, B. K., Phukan, J. P., Sinha,
A., & Sinha, R. (2014). Preoperative diagnosis of tubu- Approximately 60–70% of tubular carcinomas
lar adenoma of breast – 10 years of experience. The present as non-palpable, mammographically
Tubular Carcinoma 385
detected lesions and are more common in stromal desmoplasia. Foci of hemorrhage are usu-
women in their sixth or seventh decade who ally attributable to a prior diagnostic needling
are undergoing breast cancer screening. procedure. Necrosis is not a feature of tubular
• Sex carcinoma. Calcification is common in tubular
Female. Rare in male as for other forms carcinoma and may be the presenting sign on
of breast cancer. mammography.
• Site
Anywhere in the breast but most frequent in the
upper outer quadrant like other types of breast Microscopy
cancer. Multifocality has been reported in up to
20% of tubular carcinomas. The characteristic feature of tubular carcinoma
• Treatment is the predominance of tubules composed of
Local control and hormone therapy are the a single layer of epithelial cells with open lumen
main treatment options. Even when patients (Fig. 1). The tubules are arranged haphazardly;
have axillary metastases from tubular carci- are generally oval, rounded, and angulated in
noma, the prognosis is very good, and the shape; and lack peripheral myoepithelial cells.
axillary node dissection could be considered The cells are small to moderate in size, regular
unnecessary, and the use of systemic adjuvant with little nuclear pleomorphism, inconspicuous
chemotherapy is typically not needed in pure nucleoli, and scanty mitotic figures. Apical snouts
tubular carcinomas (Diab et al. 1999; are seen in up to a third of cases but are not
Papadatos et al. 2001; Fedko et al. 2010). pathognomonic. Multilayering of nuclei, marked
• Outcome nuclear pleomorphism, and high mitotic activity
Tubular carcinoma has an exceptionally good are contraindications for a diagnosis of tubular
long-term prognosis (Diab et al. 1999; carcinoma. The characteristic tubules of tubular
Papadatos et al. 2001; Anderson et al. 2004; carcinoma comprise more than 90% of the tumor.
Fedko et al. 2010; Rakha et al. 2010), and in Adherence to the 90% rule is essential to correctly
some series, the outcome is similar to age- identify tubular carcinoma which carries an excel-
matched population without breast cancer lent prognosis. A tumor that shows features of
(Diab et al. 1999; Sullivan et al. 2005; Fedko tubular carcinoma in 50–90% of the tumor and
et al. 2010; Rakha et al. 2010). The risk of another breast tumor type in the remainder of
local recurrence is so low that some centers the tumor, usually ▶ Invasive Carcinoma NST is
consider adjuvant radiotherapy unnecessary. regarded as a tubular mixed carcinoma.
Ten-year disease-free and overall survival The stroma is typically cellular and
rates after mastectomy or breast-conserving desmoplastic and commonly accompanying
surgery were reported to be 93.1–99.1% and the tubular structures. A consistent feature of
99–100%, respectively. Axillary node metasta- tubular carcinoma and one that is particularly
ses occur infrequently (average 10%; range useful on needle core biopsy is the extension of T
0–22%) and rarely involve more than one or tubules into surrounding fat.
two axillary lymph nodes. Tubular carcinoma frequently coexists with
other low-nuclear-grade breast lesions including
▶ Columnar Cell Lesions, flat epithelial atypia,
Macroscopy ▶ Atypical Ductal Hyperplasia, low-grade
▶ Ductal Carcinoma In Situ, and ▶ Lobular In
Tubular carcinoma presents as an ill-defined Situ Neoplasia. These lesions share morphologi-
gray to white, firm or hard mass with irregular cal, immunophenotypic, and molecular character-
outlines measuring between 0.1 cm and 2 cm in istics and constitute the so-called low-nuclear-
diameter; the majority are 1.5 cm or less. The grade neoplasia family. An association with
consistency is generally hard due to prominent ▶ Radial Scar has also been proposed.
386 Tubular Carcinoma
Tubular Carcinoma, Fig. 1 (H&E) A case of tubular (shown in Fig. 1c) and desmoplastic stroma. Tubular car-
carcinoma showing haphazardly arranged tubules lacking cinoma is associated with columnar cell change, flat epi-
myoepithelial cells and with characteristically open lumina thelial atypia, and low-grade DCIS (Figs. 1a and b )
is characterized by the lobular architecture and the carcinoma. Cancer Epidemiology, Biomarkers &
compression and distortion of glandular struc- Prevention, 13, 1128–1135.
Diab, S. G., Clark, G. M., Osborne, C. K., Libby, A.,
tures. Myoepithelial cells are always present. Allred, D. C., & Elledge, R. M. (1999). Tumor
Complex sclerosing lesions/radial scars have cen- characteristics and clinical outcome of tubular and
tral fibrosis and elastosis containing a few small, mucinous breast carcinomas. Journal of Clinical
often distorted, tubular structures around which Oncology, 17, 1442–1448.
Fedko, M. G., Scow, J. S., Shah, S. S., Reynolds, C.,
myoepithelial cells are present. The peripheral Degnim, A. C., Jakub, J. W., & Boughey, J. C.
glandular structures in a radial scar show varying (2010). Pure tubular carcinoma and axillary nodal
degrees of dilatation and ductal epithelial hyper- metastases. Annals of Surgical Oncology, 17(Suppl 3),
plasia. Microglandular adenosis that typically 338–342.
Papadatos, G., Rangan, A. M., Psarianos, T., Ung, O.,
shows haphazard, diffuse, infiltrative pattern Taylor, R., & Boyages, J. (2001). Probability of axil-
with tubules lacking peripheral myoepithelial lary node involvement in patients with tubular carci-
cells shows more rounded and regular tubules noma of the breast. British Journal of Surgery, 88,
that often contain colloid-like secretory material. 860–864.
Rakha, E. A., Lee, A. H., Evans, A. J., Menon, S.,
Microglandular adenosis also lacks desmoplastic Assad, N. Y., Hodi, Z., Macmillan, D.,
stroma characteristic of tubular carcinoma. Blamey, R. W., & Ellis, I. O. (2010). Tubular carcinoma
of the breast: Further evidence to support its excellent
prognosis. Journal of Clinical Oncology, 28, 99–104.
Sullivan, T., Raad, R. A., Goldberg, S., Assaad, S. I.,
References and Further Reading Gadd, M., Smith, B. L., Powell, S. N., Taghian, A. G.
(2005). Tubular carcinoma of the
Anderson, W. F., Chu, K. C., Chang, S., Sherman, M. E. breast: A retrospective analysis and review of the
(2004). Comparison of age-specific incidence rate literature. Breast Cancer Research and Treatment, 93,
patterns for different histopathologic types of breast 199–205.
T
U
Impalpable lesion. No specific clinical features are UDH is discovered at microscopy, the gross fea-
described being a microscopic finding. ture is not specific.
© Springer Nature Switzerland AG 2020
A. Sapino, J. Kulka (eds.), Breast Pathology, Encyclopedia of Pathology,
https://doi.org/10.1007/978-3-319-62539-3
390 Usual Ductal Hyperplasia (UDH)
Molecular Features
Differential Diagnosis
Usual Ductal
Hyperplasia (UDH),
Fig. 2 UDH showing
typical peripheral slit-like
spaces lined by luminal
cells with occasional apical
snouts. (High
magnification, H&E)
Usual Ductal
Hyperplasia (UDH),
Fig. 3 Florid UDH and
infiltrating epitheliosis
within keloid like stroma.
(H&E)
Usual Ductal
Hyperplasia (UDH),
Fig. 4 Mosaic like CK14
immunostaining of cells
in UDH
U
392 Usual Ductal Hyperplasia (UDH)
Usual Ductal Hyperplasia (UDH), Fig. 5 (a) ER immunostaining is almost negative in a field of infiltrating UDH.
(b) p63 highlights myoepithelial cells