《Modern Breast Cancer Imaging》 - 240323 - 092230
《Modern Breast Cancer Imaging》 - 240323 - 092230
《Modern Breast Cancer Imaging》 - 240323 - 092230
Cancer Imaging
Su Jin Kim Hsieh
Elizabeth Anne Morris
Editors
123
Modern Breast Cancer Imaging
Su Jin Kim Hsieh • Elizabeth Anne Morris
Editors
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature
Switzerland AG 2022
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether
the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and
transmission or information storage and retrieval, electronic adaptation, computer software, or by similar
or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the
editors give a warranty, expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
“To Mariana, Lucas and Pedro”
—Dr Su Jin Kim Hsieh
“For my two beautiful children, who are a
wonder” —Dr Elizabeth Anne Morris
Foreword
Breast cancer has haunted the female universe for millennia. And this agony has
been portrayed in many ways, both in history and in art. Sometimes in an accidental
way as in some well-known paintings and sculptures, while at other times directly,
as in hieroglyphic scripts that described it as an incurable disorder. Today, in the
third millennium of the Christian era, our knowledge about this disease has com-
pletely changed. We already have the sequencing of the main genes related to breast
cancer and specific targeted therapies, which totally altered its evolution. We know
that early detection is still the best way to increase the chances of cure and the pos-
sibility of less aggressive surgical and adjuvant treatments. Currently there are
imaging techniques capable of detecting increasingly smaller and biologically sig-
nificant tumors. And this was only achieved thanks to the tireless work of doctors
and scientists from different specialties, who joined together for a single objective:
to reduce mortality from breast cancer and provide a better quality of life for women
affected by it.
In the book Modern Breast Cancer Imaging, the editors, Su Jin Kim Hsieh and
Elizabeth Morris, both with extensive expertise in breast radiology and international
projection, brought a multidisciplinary approach to breast cancer, in the areas of
pathology, oncology, radiotherapy, and surgery, in addition to an extensive update
on diagnostic imaging applied to clinic. Divided into three large parts, the book
initially brings the main advances in science from a clinical and pathological point
of view. It covers from the molecular basis of breast cancer, risk factors, and genetic
testing to advances in surgical and pathological treatment. In Part II, the imaging
methods used in breast radiology, their indications and limitations, as well as their
future perspectives are discussed, mainly how to transpose the information obtained
to improve clinical outcomes. Part III presents the main clinical problems and how
imaging methods can help: from screening to diagnosis, including treatment and
follow-up of women diagnosed with breast cancer.
At last, despite breast cancer being one of the oldest and most studied diseases in
science, we do have a lot to evolve. Mainly because fear and prejudice against this
disease still exist, in the twenty-first century. And also, because we still witness
many lives lost, despite being a potentially curable disease. We currently live in the
vii
viii Foreword
The field of breast imaging has been evolving very fast lately, especially concerning
breast cancer. Not only screening protocols have suffered some adjustments over
the past years, but also new imaging methods, software and even new types of pro-
cedures have emerged. New concepts regarding molecular and genetic basis and
therapeutic regimens have changed greatly.
The understanding of these new concepts become essential to give the patient the
best possible care, since the confection of a relevant and useful breast imaging
report from the radiologist, and also for the other professionals to understand what
type of imaging exams to order in each scenario and how to interpret the results.
Keeping up to date demanded a great deal of effort, considering the vast amount
of information in the literature, not only in the area of imaging, but also in other
fields such as pathology, surgery and oncology.
The idea of this book was to compile up-to-date and established information
about breast cancer in all these areas, in a multimodality approach. It is intended to
be useful for daily practice not only for breast radiologists, but also for everyone
who is in this field of medical practice.
This book has been written by very experienced and renowned professionals and
would not be possible without their valuable contributions!
Special thanks to Dr Morris, who believed in this project from the beginning!
ix
Contents
xi
xii Contents
Index�������������������������������������������������������������������������������������������������������������������� 463
Contributors
1.1 Introduction
Breast cancer is the second most common cancer among women, after non-
melanoma skin cancer, with a one in eight probability of developing invasive carci-
noma in a US woman during her lifetime. In 2021, an estimated 281,550 new cases
of invasive breast carcinoma are expected to be diagnosed in American women,
along with 49,290 new cases of in situ breast cancer [1].
About 5–10% of breast cancer can be linked to a hereditary mutation of a known
gene, such as BRCA1 and BRCA2. On average, women with a BRCA1 mutation
have up to a 72% lifetime risk of developing breast cancer, while the risk is 69%
with BRCA2 mutation [2].
After the year 2000, with the sequencing of multiple types of tumors, the under-
standing of the molecular biology of breast cancer brought extremely important
knowledge, which changed the clinical practice of its treatment. It has been recog-
nized as a heterogeneous neoplasia by several biomarkers, now used in diagnosis,
prognosis, and treatment according to each subtype.
The heterogeneity of breast carcinomas is due to the involvement of different
genes, related to the proliferative activity, differentiation, and suppression of tumors,
several of them studied alone or in small groups, regarding the implication in carci-
nogenesis and/or as prognostic factors. However, the enormous heterogeneity and
the number of factors involved make interpretation difficult. With the development
of genome analysis technologies with the possibility of concomitant analysis of
thousands of genes, it was possible to identify intrinsic gene profiles in breast
R. C. M. Fernandes (*)
Hospital Beneficência Portuguesa de São Paulo/Laboratório Bacchi, São Paulo, SP, Brazil
Pathology Department Director, Hospital Pérola Byington, São Paulo, SP, Brazil
e-mail: [email protected]
tumors called basal; HER2-enriched; luminal A, B, and C; and normal breast type
according to the classification of Perou et al. [3–7]. These presentation profiles
showed consistency even in different study groups and using different array tech-
niques [4, 5, 8].
Over the past few years, there has been considerable effort to characterize and
classify breast cancer at the molecular level in order to effectively tailor treatment.
However, due to time and cost constraints, in the great majority of health-care sys-
tems, a surrogate molecular breast cancer classification is still largely based on
immunohistochemical (IHC) assessment of biomarkers: ER (estrogen receptor), PR
(progesterone receptor), HER2, and Ki-67. Nevertheless, the examination of global
patterns of gene expression (especially of genes involved in the regulation of cell
growth and other important aspects of cell behavior, such as invasion) has resulted
in the identification of intrinsic molecular subtypes of biological and clinical rele-
vance and of gene signatures predictive of outcome or response to therapy [9].
It is extremely important to remember that to avoid false-negative result for any
molecular or IHC study, the biopsy or surgical specimen must be properly fixed at
10% buffered formalin to ensure pH range 7.0 and 7.4 within 1 hour after resection
averting prolonged cold ischemic time, which may result in antigenic degradation.
Under- or over-fixation is not recommended. Fixation for at least 6 hours and a
maximum of 72 hours are recommendations from the College of American
Pathologists (CAP) [10].
In this scenario, immunotherapy with checkpoint inhibitors has made great prog-
ress. Early in 2019, atezolizumab, an anti-PD-L1 monoclonal antibody, was
approved in combination with nanoparticle albumin-bound (nab) paclitaxel, a form
of paclitaxel that does not require steroid premedication, for the treatment of patients
with locally advanced or metastatic TNC whose tumors are positive for PD-L1
expression (≥1%) on immune infiltrate, evaluated by the Ventana PD-L1 (SP142)
antibody [18].
The tumor mutational burden (TMB) is substantially lower in breast cancer than
in other solid malignancies such as lung cancer or melanoma, with low ability to
develop a competent immune response related only to this variant [19].
Along with BRCA1/2-mutation breast cancer, about 50% of sporadic TNC have
the BRCAness phenotype, which gives them, clinical and pathological characteris-
tics to be highlighted: large tumors (>2.0 cm), circumscribed with pushing borders,
sheets of pleomorphic tumor cells, syncytial-like growth pattern, brisk lymphocytic
stromal reaction, with geographic pattern necrosis or fibrosis, usually occurring in
young patients and high KI-67 levels.
They are frequently associated with TP53 gene mutation and EGFR expression,
deficient in homologous recombination and with sensitivity to (adenosine
diphosphate-ribose) polymerase inhibitors and platinum salts [20, 21].
Fig. 1.1
Immunohistochemical
reaction illustrating
positive case for estrogen
receptor. Nuclear staining
in >1% of neoplastic cells
8 R. C. M. Fernandes
types with the best prognosis (pure mucinous and tubular carcinomas) presented
the lowest levels of gene copy number changes and corresponded to the luminal
subtype A.
In the 2013 St. Gallen consensus, the approximate cut-point in immunohisto-
chemistry to separate the luminal subtypes A and B was KI67 ≤20% and/or PR
<20% [26].
Currently, we also have genomic tests for HER-negative LCs, which can be used
to define treatment in cases of discordant clinical and genomic risks, formerly used
only to guide prognosis, such as the distance recurrence index determined by 21
genes [27] and the Amsterdam test of 70 genes [28]. These will be discussed in a
specific chapter.
As for the metastatic scenario, Chen et al. [29] provided a large single center
cohort to assess the frequency of receptor conversion in metastatic breast cancer.
The overall discordant rates were 18.3%, 40.3%, and 13.7% for ER, PR, and HER2,
respectively. The discordance was significantly higher for PR when compared with
ER and HER2. The conversion occurred significantly as a switch from positive to
negative receptor status when compared to the conversion from negative to positive
for all three receptors. Semiquantitative analyses revealed a significantly decreased
expression of both ER (25%) and PR (57%) in the metastases. There was a higher
rate of PR discordance in bone metastases when compared to other common organs
of relapse.
A positive ER status, in primary or metastatic breast cancer, was associated with
a prolonged metastasis-free survival when compared with ER-negative primary
tumors without conversion. Furthermore, a positive ER status in metastatic breast
cancer regardless of the primary tumor was associated with a superior overall sur-
vival when compared with an ER-negative tumor without conversion. Thus, recep-
tor conversion is a frequent event in the course of breast cancer progression and can
also be seen between different metastatic sites. Moreover, some conversions are of
prognostic significance. The findings may reflect tumor heterogeneity, sampling, or
treatment effect but may also indicate alteration in tumor biology. Repeat biomarker
testing is justified when making appropriate treatment plans in the pursuit of preci-
sion medicine.
Despite continuous expression of the ER, for unknown reasons, many ER+
breast cancers in the metastatic setting become refractory to the inhibition of estro-
gen action. Thirty-one biopsy samples collected after progression on hormonal
therapy from patients with metastatic ER+ breast cancer were subjected to targeted
DNA sequencing in a study [30]. ESR1 mutations (a total of 9/36 cases) were
detected at much higher rates than in those reported by TCGA. The mutations in
ESR1 clustered in the ligand-binding domain. Furthermore, analysis of the avail-
able paired primary samples showed that these mutations were an acquired event.
The mutations clearly occurred in a population of patients with hormone refractory
disease, and the biochemical and structural data demonstrated that these mutations
promoted the agonist conformation of ER in the absence of ligand. Furthermore, the
mutant ER isoforms were only partially inhibited by direct receptor antagonists
such as tamoxifen or fulvestrant and needed higher doses than those used clinically
1 Molecular Basis of Breast Cancer 9
factor [10]. HER2 status is primarily evaluated to determine patient eligibility for
anti-HER2 therapy. It may identify patients who have a greater benefit from
anthracycline-based adjuvant therapy.
HER2 status can be determined in formalin-fixed paraffin-embedded tissue by
assessing protein expression on the membrane of tumor cells using IHC or by
assessing the number of HER2 gene copies using in situ hybridization (ISH). When
both IHC and ISH are performed on the same tumor, the results should be corre-
lated. The most likely reason for a discrepancy is that one of the assays is incorrect,
but in a small number of cases, there may be protein overexpression without ampli-
fication, amplification without protein overexpression, or marked intratumoral het-
erogeneity. We must always keep in mind the intratumoral heterogeneity; when
there is a negative result in a small biopsy sample, repeated testing on a subsequent
specimen with a larger area of carcinoma sampled should be considered, particu-
larly if the tumor has characteristics associated with HER2 positivity (i.e., tumor
grade 2 or 3, weak or negative PR expression, increased proliferation index) [10].
When multiple invasive foci are present, the largest invasive focus should be tested.
Testing smaller invasive carcinomas is also recommended if they are of different
histologic type or higher grade.
The major challenge for the characterization of the HER2 group was the ade-
quate laboratory standardization. The American Society of Clinical Oncology
(ASCO) and CAP standardized criteria for positivity and laboratory control for
adequate testing [10]. The immunohistochemical criteria are illustrated in Table 1.1
(Figs. 1.2 and 1.3), remembering that all HER2 2+ tumors (equivocal) must be sub-
jected to ISH to confirm the real status of HER2. It is recommended that hormone
receptor and HER2 testing be performed in all primary invasive breast carcinomas
and in recurrent or metastatic tumors.
Fluorescence in situ hybridization (FISH), chromogenic in situ hybridization
(CISH), and silver-enhanced in situ hybridization (SISH) studies for HER2 deter-
mine the presence or absence of gene amplification. Some assays use a single probe
to determine the number of HER2 gene copies present, but most assays include a
chromosome enumeration probe (CEP17) to determine the ratio of HER2 signals to
copies of chromosome 17. Although 10% to 50% of breast carcinomas have more
Fig. 1.2
Immunohistochemical
reaction illustrating a
positive case (score 3+) for
HER2. Strong and
complete membrane
staining in >10% of
neoplastic cells
Fig. 1.3
Immunohistochemical
reaction illustrating an
equivocal case (score 2+)
for HER2. Weak to
moderate complete
membrane staining in
>10% of neoplastic cells
than two CEP17 copies, only 1% to 2% of carcinomas show true polysomy (i.e.,
duplication of the entire chromosome) (10).
Dual probe defines five groups by ISH: Group 1 = HER2/CEP17 ratio ≥2.0, ≥4.0
HER2 signals/cell; Group 2 = HER2/CEP17 ratio ≥2.0, <4.0 HER2 signals/cell;
Group 3 = HER2/CEP17 ratio <2, ≥6.0 signals/cell; Group 4 = HER2/CEP17 ratio
<2, ≥4.0 and <6.0 HER2 signals/cell; and Group 5 = HER2/CEP17 ratio <2.0, <4.0
HER2 signal/cell. Reporting results recommended by CAP/ASCO are shown in
Table 1.2.
Group 1 is positive, group 5 is negative, and groups 2, 3, and 4 require a new
count, together with immunohistochemistry and preferably by a second observer, to
determine whether the result is positive or negative, without further misunderstand-
ing after ISH. Regardless of the result, the group must be returned to the final report
after the ISH.
12 R. C. M. Fernandes
Table 1.2 Reporting results of HER2 testing by in situ hybridization (dual-probe assay)
Result Criteria (dual-probe assay)
Negative Group 5
Negative – results based on concurrent review of IHC Group 2 and concurrent IHC 0-1+ or 2+
Group 3 and concurrent IHC 0-1+
Group 4 and concurrent IHC 0-1+ or 2+
Positive – results based on concurrent review of IHC Group 2 and concurrent IHC 3+
Group 3 and concurrent IHC 2+ or 3+
Group 4 and concurrent IHC 3+
Positive – results based on concurrent review of IHC Group 1
Adapted from 2018 CAP/ASCO Update recommendations
References
1. American Cancer Society. How common is breast cancer? Jan. 2021. Available at: https://
www.cancer.org/cancer/breast-cancer/about/how-common-is-breast-cancer.html.
2. National Cancer Institute. BRCA mutations: cancer risk and genetic testing. Jan. 2018. Available
at: https://www.cancer.gov/about-cancer/causes-prevention/genetics/brca-fact-sheet.
3. Perou CM, Sorlie T, Eisen MB, van de RM JSS, Rees CA, et al. Molecular portraits of human
breast tumours. Nature. 2000;406(6797):747–52.
4. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns
of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad
Sci U S A. 2001;98(19):10869–74.
5. West M, Blanchette C, Dressman H, Huang E, Ishida S, Spang R, et al. Predicting the clini-
cal status of human breast cancer by using gene expression profiles. Proc Natl Acad Sci U S
A. 2001;98(20):11462–7.
6. Sotiriou C, Neo SY, McShane LM, Korn EL, Long PM, Jazaeri A, et al. Breast cancer clas-
sification and prognosis based on gene expression profiles from a population-based study. Proc
Natl Acad Sci U S A. 2003;100(18):10393–8.
7. Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, et al. Repeated observation
of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S
A. 2003;100(14):8418–23.
8. Van't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, et al. Gene expression profil-
ing predicts clinical outcome of breast cancer. Nature. 2002;415(6871):530–6.
9. WHO Classification of Tumours Editorial Board. Breast tumours, WHO classification of
tumours series, vol. 2. 5th ed. Lyon: International Agency for Research on Cancer; 2019.
10. Cancer Protocols Templates of College of American Pathologists. Available at: https://www.
cap.org/protocols-and-guidelines/cancer-reporting-tools/cancer-protocol-templates.
11. Birnbaum D, Bertucci F, Ginestier C, Tagett R, Jacquemier J, Charafe-Jauffret E. Basal and
luminal breast cancers: basic or luminous? (review). Int J Oncol. 2004;25(2):249–58.
12. Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, et al. Immunohistochemical and
clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer
Res. 2004;10(16):5367–74.
13. Livasy CA, Karaca G, Nanda R, Tretiakova MS, Olopade OI, Moore DT, et al. Phenotypic eval-
uation of the basal-like subtype of invasive breast carcinoma. Mod Pathol. 2006;19(2):264–71.
14. Stingl J, Caldas C. Molecular heterogeneity of breast carcinomas and the cancer stem cell
hypothesis. Nat Rev Cancer. 2007;7(10):791–9.
15. Thike AA, Cheok PY, Jara-Lazro AR, Tan B, Tan P, et al. Triple-negative breast cancer: clini-
copathological characteristics and relationship with basal-like breast cancer. Mod Pathol.
2010;23(1):123–33.
16. Lehmann BD, Jovanović B, Chen X, Estrada MV, Johnson KN, Shyr Y, et al. Refinement of
triple-negative breast cancer molecular subtypes: implications for neoadjuvant chemotherapy
selection. PLoS One. 2016;11(6):e0157368.
14 R. C. M. Fernandes
17. Wimberly H, Brown JR, Schlper K, Haack H, Silver MR, et al. PD-L1 expression correlates
with tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy in breast can-
cer. Cancer Immunol Res. 2015;3(4):326–32.
18. Schmid P, Adams S, Rugo HS, Schneeweiss A, et al. Atezolizumab and Nab-Pacltaxel in
advanced triple-negative breast cancer. N Engl J Med. 2018;379:2108–21.
19. Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, et al. Mutational landscape and sig-
nificance across 12 major cancer types. Nature. 2013;502(7471):333–9.
20. Tian T, Shan L, Yang W, Zhou X, Shui R. Evaluation of the BRCAness phenotype and its
correlation with clinicopathological features in triple-negative breast cancers. Hum Pathol.
2019;84:231–8.
21. Gross E, van Tinteren H, Li Z, Raab S, Meul C, Avril S, et al. Identification of BRCA1-like
triple-negative breast cancers by quantitative multiplex-ligation-dependent probe amplifica-
tion (MLPA) analysis of BRCA1-associated chromosomal regions: a validation study. BMC
Cancer. 2016;16(1):811.
22. Fernandes RCM, Bevilacqua JLB, Soares IC, Siqueira SAC, Pires L, et al. Coordinated expres-
sion of ER, PR and HER2 define different prognostic subtypes among poorly differentiated
breast carcinomas. Histopathology. 2009;55(3):346–52.
23. Loi S, Haibe-Kains B, Desmedt C, Lallemand F, Tutt AM, Gillet C, et al. Definition of clini-
cally distinct molecular subtypes in estrogen receptor-positive breast carcinomas through
genomic grade. J Clin Oncol. 2007;25(10):1239–46.
24. Allison KH, Hammond MEH, Dowsett M, McKernin SE, Carey AL, et al. Estrogen and
progesterone receptor testing in breast cancer: ASCO/CAP guideline update. J Clin Oncol.
2020;38(12):1246–66.
25. Dieci MV, Orvieto E, Dominici M, Conte P, Guarneri V. Rare breast cancer subtypes: histo-
logical, molecular, and clinical peculiarities. Oncologist. 2014;19(8):805–13.
26. Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thürlimann B, et al.
Personalizing the treatment of women with early breast cancer: highlights of the St Gallen
International Expert Consensus on the primary therapy of early breast cancer 2013. Ann
Oncol. 2013;24:2206–23.
27. Sparano JA, Gray RJ, Makower DF, Pritchard KI, Albain KS, Hayes DF, et al. Adjuvant
chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med.
2018;379(2):111–21.
28. Cardoso F, van’t Veer LJ, Bogaerts J, Slaets L, Viale G, Delaloge S, et al. MINDACT
Investigators. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N
Engl J Med. 2016;375(8):717–29.
29. Chen R, Qarmali M, Siegal GP, Wei S. Receptor conversion in metastatic breast cancer: analy-
sis of 390 cases from a single institution. Modern Pathol. 2020;33:2499–506.
30. Toy W, Shen Y, Won H, Green B, Sakr RA. ESR1 ligand-binding domain mutations in
hormone-resistant breast cancer. Nat Genet. 2013;45:1439–45.
31. Aftimos P Jr, AAAJ, Sotiriou C. Tumours of the breast. In: Coleman WB, Tsongalis GJ, editors.
Molecular pathology – the molecular basis of human disease. Academic Press; 2017. p. 583.
32. Andre F, Ciruelos E, Rubovszky G, Campone M, Loibl S, et al. Alpelisib for PIK3CA-mutated,
hormone receptor–positive advanced breast cancer. N Engl J Med. 2019;380(20):1929–40.
33. Choi YJ, Anders L. Signaling through cyclin D-dependent kinases. Oncogene.
2014;33:1890–903.
34. Konecny G, Pauletti G, Pegram M, Untch M, Dandekar S, Aguilar Z, et al. Quantitative asso-
ciation between HER-2/neu and steroid hormone receptors in hormone receptor-positive pri-
mary breast cancer. J Natl Cancer Inst. 2003;95(2):142–53.
35. Ellis MJ. Neoadjuvant endocrine therapy for breast cancer: medical perspectives. Clin Cancer
Res. 2001;7(12 Suppl):4388s–4391s.
36. Villman K, Sjostrom J, Heikkila R, Hultborn R, Malmstrom P, Bengtsson NO, et al. TOP2A
and HER2 gene amplification as predictors of response to anthracycline treatment in breast
cancer. Acta Oncol. 2006;45(5):590–6.
1 Molecular Basis of Breast Cancer 15
2.1 Introduction
Breast cancer is the second most common malignancy globally after lung cancer
and the most frequent cancer among women.
About 2 million new cases of cancer occur each year. In 2020, there were
2,261,419 new cases and 684,996 deaths due to breast cancer worldwide [1, 2]. The
incidence of breast cancer has been increasing in recent decades. In Brazil, the esti-
mate for the year 2020 is 66,280 new cases, with 16,927 deaths [3]. Due to its sig-
nificant incidence and prevalence, it is important to understand the risk factors for
its development and what can be done to reduce the risk of acquiring it.
Studies in the literature show that about 10% of breast cancer cases are related to
hereditary genetic mutations, while the other 90% occur randomly. Risk factors can
be related to age, sex, and reproductive cycle. There are also behavioral factors.
Other factors, such as precursor lesions and genetic mutations, must be evaluated.
Breast cancer mainly affects women after 50 years old. Its incidence ranges from
14.5 to 55.6 (ASR-age standardized rate) per 100,000 women in Africa to 84.9
(ASR) per 100,000 in Brazil, 62.9 (ASR) per 100,000 in the United States, 93.6
(ASR) per 100,000 in the United Kingdom, and 99.1 (ASR) per 100,000 in France.
The male sex corresponds to only 1% of cases, with one man per 100,000 standard
population. When it affects men, they are generally older and may present some
hormonal alteration, or it may be related to the BRCA2 gene mutation [4].
Regarding age, the incidence of breast cancer is higher among women after
menopause. In the United States, about 77% of cases occur in women after 50 years
of age, and the average age at diagnosis is 62 years old [5, 6]. In the United Kingdom,
between 2015 and 2017, about 85% of cases were diagnosed in women over
50 years old [7]. Therefore, the population at the most significant risk for breast
cancer are women over 50 years old, usually after menopause. They must undergo
screening with annual or biannual mammograms according to each country’s pro-
gram [1, 8].
Early menarche, nulliparity, and late pregnancy are risk factors for the development
of breast cancer. In the literature, studies show a relationship between these factors
and the appearance of positive hormone receptor breast cancer, which is not related
to negative hormone receptor breast cancer. Probably the longer exposure time of
the breast tissue to the hormonal action, such as estrogen and progesterone, the
greater the likelihood of the appearance of breast neoplasms [9]. Thakur and col-
laborators [10] carried out a case-control study, which included 377 women with
breast cancer and 346 controls. In this study, early menarche (<12 years) increased
the risk for breast cancer by up to two times (odds ratio (OR) = 2.83, 95% confi-
dence interval (CI) = 1.02–7.86); the same was found for late menopause (>50 years)
(OR = 2.43, 95% CI = 1.2–4.9).
Huo and collaborators [11] also sought to assess menarche’s influence on the
onset of breast cancer. They carried out a case-control study, evaluating 819 patients
with breast cancer and 569 controls, and found that for every 2 years of delay in
menarche, there was a reduction in 7% risk of developing breast cancer.
Pregnancy and lactation are considered protective factors against developing breast
cancer, and the age of the pregnancy and the duration of lactation are responsible for
providing this protection. Ma and collaborators [12] evaluated that pregnancy
decreases the risk of developing breast cancer by 11%. Still, it presents itself as a
protective factor when it occurs before the age of 30 and can be a risk factor when
it occurs at a late age. Women who became pregnant after the age of 30 were 27%
more likely to have hormonal positive breast cancer than the patients who became
pregnant before the age of 30. This difference is probably due to the breast’s matura-
tion stages, as it completes its embryological development with pregnancy. The
2 Risk Factors for Breast Cancer 19
sooner this occurs, the less the chance of cellular changes that can be stimulated by
the hormonal action of pregnancy, leading to the onset of neoplasms [13]. Thakur
et al. [10], in a case-control study, found a risk of up to six times greater than having
breast cancer when pregnancy occurs at an older age (>30 years) (OR = 6.34, 95%
CI = 2.04–27).
A meta-analysis carried out by the collaborative group on hormonal factors of
the breast [14] evaluated the impact of pregnancy on breast cancer; forty-seven stud-
ies were carried out, were conducted in 30 countries, and compared 12,214 nulli-
gravid women with 16,900 women who had children but did not breastfeed. The
result found that the earlier the pregnancy occurred, the lower the relative risk of
developing breast cancer. The risk of developing breast cancer decreases by 7%
with each delivery, demonstrating that pregnancy is a protective factor against the
onset of breast cancer.
The practice of breastfeeding is a protective factor for breast cancer develop-
ment, but there is still no consensus on the ideal breastfeeding time for its preven-
tion [12, 15]. Huo and collaborators [11] found that every 12 months, lactation
reduces the risk of developing breast cancer by 7%. A meta-analysis carried out by
the collaborative group on hormonal factors of the breast [14] also identified breast-
feeding as a protective factor. Every 12 months of breastfeeding, there is a decrease
in the relative risk estimated at 4.5%.
Studies in the literature that have assessed the influence of hormonal contraceptives
on breast cancer have shown variable findings ranging from no increased risk to a
relative risk of 9.5, thus being a controversial subject [4, 16].
Marshbanks and collaborators [16] conducted a case-control study, evaluating
2282 women with breast cancer and 2424 women without breast cancer, and found
no association between the use of hormonal contraceptives and an increased risk of
developing breast cancer.
Mørch and collaborators [17], seeking a more recent analysis on the use of hor-
monal contraceptives and their relationship with breast cancer, conducted a cohort
study evaluating approximately 1.8 million women between 15 and 49 years old,
who were followed up by about 10.9 years. In this period, there were 11,517 cases
of breast cancer. The relative risk found for women who used contraceptives com-
pared to women who never used them was 1.20 (95% CI, 1.14–1.26). The risk
increased from 1.09 (95% CI, 0.96–1.23) in women with less than 1 year of use to
1.38 (95% CI, 1.26–1.51) in women over 10 years of using hormonal contracep-
tives. The absolute risk for using hormonal contraceptives was 13 (95% CI, 10–16)
per 100,000 women-years or approximately one extra breast cancer case for every
7690 women using hormonal contraceptives for 1 year. Therefore, the use of hor-
monal contraceptives should be discussed with the patient, individualizing personal
risk factors and deciding with her the use thereof.
20 S. Masili-Oku et al.
2.6 Obesity
Obesity has been described as a risk factor for the development of breast cancer.
Adipose tissue is an androgen producer and allows more significant hormonal action
in the breasts through their peripheral conversion [9].
Obesity is a risk factor for breast cancer in postmenopausal women. Kabat and
collaborators [22] evaluated obesity and metabolic syndrome as a risk factor for
breast cancer. An arm of the WHI study included 20,944 women with obesity and
metabolic syndrome. Blood glucose, triglycerides, HDL-cholesterol, blood pres-
sure, waist circumference, and body mass index (BMI) measurements were evalu-
ated. Women were classified according to their BMI (18.5–<25.0, normal weight;
25.0–<30.0, overweight; and ≥30.0 kg/m2, obese) and the presence of metabolic
syndrome (≥3 of the following: waist circumference ≥88 cm, triglycerides
≥150 mg/dL, HDL-C <50 mg/dL, glucose ≥100 mg/dL, and systolic/diastolic
blood pressure ≥130/85 mmHg or treatment for hypertension), yielding six groups:
metabolically healthy/normal weight (MHNW), metabolically unhealthy/normal
weight (MUNW), metabolically healthy/overweight (MHOW), metabolically
2 Risk Factors for Breast Cancer 21
Alcohol consumption and smoking are associated with the development of breast
cancer. The greater the consumption before pregnancy, the greater the risk [4]. Park
and collaborators [23] evaluated 85,089 women and alcohol consumption in a
cohort with women from different ethnicities in Hawaii and California, with an
average follow-up of 12.4 years. During this time, 3885 cases of breast cancer were
identified. The hazard ratios (HRs) found demonstrated that alcohol consumption
increases the risk of developing breast cancer compared to not consuming it. The
HRs were 1.23 (95% CI, 1.06–1.42), 1.21 (95% CI, 1.00–1.45), 1.12 (95% CI,
0.95–1.31), and 1.53 (95% CI, 1.32–1.77) for 5–9.9, 10–14.9, 15–29.9, and ≥30 g/
day of alcohol consumption, respectively.
A cohort between 1899 and 1975 with 302,865 Norwegian women evaluated the
effect of smoking on cancer and concluded that women who smoked had a 15%
increased risk of having breast cancer (HR = 1.15; 95% CI, 1.10–1.21) [24].
Chest radiotherapy performed to treat Hodgkin’s disease increases the risk of devel-
oping breast cancer. Studies have shown that this risk is greater the younger the
woman is when undergoing chest radiotherapy. The relative risk for developing
breast cancer for women who received chest radiotherapy between the ages of 15
and 19 is 30.7; between 20 and 24 years, the RR is 14.0; between 25 and 29 years
old, the RR is 5.5, and between 30 and 35 years old, the RR is 5.3. Thoracic radio-
therapy after age 35 does not seem to increase this risk [25].
The risk of developing breast cancer is also related to the time that elapses after
the end of chest radiotherapy. The RR is 2.9 after 5 years of the end of irradiation;
the peak is after 15 to 19 years when this risk reaches an RR of 11.1, and after 20 to
25 years, there is a drop to an RR of 8.0 [25].
22 S. Masili-Oku et al.
The proliferative intraductal lesions usually have their origin in the terminal duct
lobular unit. They are composed of a heterogeneous group of lesions concerning
cytology and architecture. Their presence confers an increased risk of developing an
invasive lesion. The usual ductal hyperplasia, atypical hyperplasia, ductal carci-
noma in situ, columnar cell lesion, columnar cell lesion with atypia, lobular carci-
noma in situ, and flat epithelial atypia are part of this group. Since the increase in
risk for breast cancer is 1.5 times in the presence of usual ductal hyperplasia, this
rises to four to five times in ductal hyperplasia with atypia/columnar cell lesion with
atypia and eight to ten times in ductal carcinoma in situ/lobular carcinoma in
situ [28].
Atypical hyperplasia corresponds to a monotonous proliferation of epithelial
cells inside the mammary duct. Without breaching through the myoepithelial layer,
it does not compromise two consecutive mammary ducts and measures less than
2 mm. In terms of cytological characteristics, it is similar to low-grade ductal carci-
noma in situ. It is classified when atypical cells affect two consecutive whole ducts
and are present in an extension of 2 mm or more.
Columnar cell lesion with atypia is characterized by the proliferation of monoto-
nous, noncohesive cells that fill the mammary duct and compromise less than 50%
of the acini. In contrast, lobular carcinoma in situ is formed by these same cellular
changes that occupy more than 50% of acini [28, 29].
The greater access to breast screening programs and the more significant number
of mammograms performed have increased the diagnosis of atypia in percutaneous
biopsies performed for detected masses and microcalcifications found in imaging
exams. The presence of atypia in a breast biopsy, besides representing a greater risk
for this patient to develop breast cancer, implies the need for surgical excision for
the total removal of the lesion, since the percutaneous biopsy may underestimate the
presence of an invasive lesion in 15% to 30% [29].
As for the flat epithelial atypia (FEA), few data establish its risk for breast cancer
development as it is a rare finding. A study conducted by the Mayo Clinic [30]
evaluating 11,591 women with benign breast biopsies found 282 biopsies with
2 Risk Factors for Breast Cancer 23
FEA. Of these, 130 were associated with atypical hyperplasia (AH) (46%), and the
others were associated with proliferative disease without atypia (PDWA). These
patients were followed for an average of 16.8 years. The standardized incidence
ratios (SIRs) were the following: SIR for breast cancer in women with AH + FEA
was 4.74 (95% confidence interval [95% CI], 3.17–6.81) versus 4.23 (95% CI,
3.44–5.13) for AH without FEA (P = 0.59). SIR for PDWA + FEA was 2.04 (95%
CI, 1.23–3.19) versus 1.90 (95% CI, 1.72–2.09) for patients with PDWA without
FEA (P = 0.76), so the risk of developing breast cancer was mainly attributed to the
presence of other hyperplasias with atypia rather than the presence of flat epithelial
atypia itself.
In the literature, there is a conundrum about the evolution of a precursor lesion to
an invasive breast cancer. It is known that breast cancer’s natural course does not
always follow this order: usual hyperplasia, atypical hyperplasia, ductal carcinoma
in situ, and then to an invasive ductal carcinoma. Some studies demonstrate a more
complex path, which does not follow these steps for the development of an invasive
breast cancer. To et al. [31] followed 146 women for 25 years, 111 with ductal car-
cinoma in situ (DCIS) and 35 with lobular carcinoma in situ (LCIS). Among these,
26 (19 from the DCIS group and seven from LCIS) developed invasive breast cancer
(17.8%), and 12 died of invasive cancer (8.2%). The average time for the diagnosis
of invasive cancer was 6.3 years. The probability of progressing to invasive breast
cancer was 10.0% in 5 years, 13.7% in 10 years, 17.6% in 15 years, and 19.7% in
20 years. Although 20% were related to the appearance of invasive cancer, in the
other 80%, this relationship was not clear, allowing the discussion that not all in situ
cancers evolve to invasive ones.
Familial breast cancer is responsible for 5% to 10% of all breast cancers. Most of
these cases may be associated with mutations in the BRCA1 or BRCA2 genes [32].
The lifetime risk of developing breast cancer for a woman is approximately
12.9%. That is, one in eight women will be diagnosed with breast cancer at some
point in their lives, leading to the estimated prevalence of 3,577,264 women living
with breast cancer in the United States in 2017 [33].
Several factors suggest a genetic contribution to breast cancer, such as [34]:
1. Increased incidence among individuals with a family history of these cancers
2. Several family members affected by these and other types of cancer
3. A cancer pattern compatible with autosomal dominant inheritance
Didactically, we can classify cancer cases [35]:
(a) Sporadic: when there are no cases of the same type of cancer in two genera-
tions, that is, brothers, children, parents, aunts, and uncles, and both pairs of
unaffected grandparents.
24 S. Masili-Oku et al.
(b) Familial: when there are two or more second-degree relatives with breast can-
cer, regardless of age, bilaterality, or other associated cancers. It corresponds to
about 25% of the total diagnoses. Its etiology is multifactorial or random.
(c) Hereditary: its genealogy leads us to think of an autosomal dominant distribu-
tion characterized by early age onset, a more significant number of cases with
bilaterality, and multiple primary cancers. It corresponds to about 10% of the
total cases.
Breast and ovarian cancers are present in several autosomal dominant cancer syn-
dromes, although they are more strongly associated with highly penetrating patho-
genic variants in BRCA1 and BRCA2. Other genes, such as PALB2, TP53
(associated with Li-Fraumeni syndrome), PTEN (associated with Cowden syn-
drome), CDH1 (associated with diffuse gastric and lobular cancer syndrome), and
STK11 (associated with Peutz-Jeghers syndrome), confer risk for one or both can-
cers with relatively high penetrance [34].
(a) TP53
Mutations involving p53 are inherited in an autosomal dominant manner, which
results in a familial predisposition to various cancers, with an estimated risk of
breast cancer of around 49% at 60 years old and, according to several studies, a
large percentage of diagnoses in young people (about 30 years old). It is associated
with sarcomas, tumors of the central nervous system, leukemia, and adrenocortical
carcinoma.
(b) STK11
Characterized by mutations in the serine-threonine kinase STK11, Peutz-Jeghers
syndrome manifests itself by the appearance of hamartomas (perioral, hands, and
genitals) and polyps in the gastrointestinal tract, in addition to malignant tumors,
including uterus, ovary, and breast (relative risk, 15 times higher than the general
population) [36].
(c) PTEN
Mutations in the tumor suppressor gene PTEN constitute an autosomal dominant
inheritance known as Cowden syndrome, which is characterized by the develop-
ment of multiple hamartomas on the skin and mucous membranes and an increased
risk of developing thyroid, kidney, endometrial, and breast cancer (estimated risk of
85%) [37].
2 Risk Factors for Breast Cancer 25
(d) BRCA
BRCA is a tumor suppressor gene responsible for maintaining the integrity of the
genome since it makes possible the recombination of homologous bases of DNA,
repairs the breakdown of the double helix, and controls DNA damage in the S
phase [38].
A recent study [39] estimated that about 72% of women who inherit a harmful
BRCA1 mutation and about 69% of women who inherit a harmful BRCA2 muta-
tion would develop breast cancer at the age of 80 years old.
It is estimated that in 20 years after the first diagnosis of breast cancer, about
40% of women who inherit a BRCA1 mutation and about 26% of women who
inherit a BRCA2 mutation will manifest cancer in their contralateral breast.
About 1.3% of women in the general population will develop ovarian cancer
at some point in their lives. On the other hand, it is estimated that approximately
44% of women who inherit a deleterious BRCA1 mutation and about 17% of
women who inherit a BRCA2 mutation will develop ovarian cancer at 80 years
old [40].
The BRCA mutation increases the risk for several other types of cancer, includ-
ing fallopian tube cancer and peritoneal cancer. Men with BRCA2 mutations are at
increased risk of breast and prostate cancer. Men and women with BRCA mutations
are at increased risk of pancreatic cancer [40].
Ashkenazi Jewish ancestry and Norwegian, Dutch, and Icelandic people have a
higher prevalence of deleterious BRCA1 and BRCA2 mutations than people in the
general population.
(a) CHEK2
CHEK2 gene is a member of the Fanconi anemia (FA)-BRCA pathway and is
involved in both the checkpoint function and repair mediated by BRCA1 and p53.
CHEK * 1100delC mutations are associated with a three- to fivefold increase in
breast cancer. The cumulative risk of breast cancer at the age of 70 years old among
CHEK * 1100delC heterozygotes reaches 37% [41].
(b) PALB2
PALB2 is another gene on the FA-BRCA pathway. It encodes a protein that
binds closely to BRCA2, stabilizing it and allowing it to perform its repairing
functions. This interaction is essential for tumor suppression by BRCA2.
Germline mutations in PALB2 increase the chance of early-onset breast cancer,
with a five- to ninefold elevated risk. Unlike BRCA mutations, patients with a
PALB2 mutation do not experience a statistically significant increase in ovar-
ian cancer.
26 S. Masili-Oku et al.
(c) ATM
Mutations in the ATM gene result in ataxia-telangiectasia, an autosomal reces-
sive neurodegenerative disease that causes cerebellar dysfunction and a weakened
immune response.
ATM protein is an essential kinase of the cell cycle within the FA-BRCA path-
way and, among its functions, phosphoryl BRCA1. Studies have shown that patients
with the ATM mutation have a breast cancer risk of approximately 2.37 [41].
Risk assessment models have been developed to clarify the lifetime risk of develop-
ing breast cancer and the likelihood of having a pathogenic variant in BRCA1 or
BRCA2 (3). When this risk is greater than or equal to 10%, conducting genetic test-
ing should be suggested.
Among these risk calculators, we can mentio Claus and Gail’s models, which
estimate cancer-based risk only on personal and family characteristics.
To assess the probability of BRCA mutation, BRCAPRO, Penn II, Tyrer-Cuzick
model, and BOADICEA can be utilized.
The BRCA mutation analysis can range from direct, including only a specific muta-
tion in BRCA1 or BRCA2 (single site analysis), to more complete tests that include
sequencing of the entire gene [41].
Given the number of genes associated with an increased risk of breast cancer,
several genes’ massive sequencing may be available. Such an option may be of
interest in patients whose family history is not very informative, that is, a restricted
or unknown family structure (for instance, in case of adoption).
The strategy is to offer the broadest genetic test to the family member affected by
the neoplasia and the guided analysis for the mutation then defined to the other fam-
ily members.
Possible results for genetic tests are:
(a) True positive: the identified mutation is associated with an increased risk
of cancer.
(b) True negative: no mutation is identified in an individual from a family known to
have a mutation.
(c) Meaningless: no mutation is identified in an individual whose family also has
no mutations.
(d) Uncertain meaning: a mutation has been identified; however, its clinical signifi-
cance is unknown at the moment.
2 Risk Factors for Breast Cancer 27
References
22. Kabat GC, Kim MY, Lee JS, Ho GY, Going SB, Beebe-Dimmer J, et al. Metabolic obesity
phenotypes and risk of breast cancer in postmenopausal women. Cancer Epidemiol Biomark
Prev. 2017;26(12):1730–5.
23. Park SY, Kolonel LN, Lim U, White KK, Henderson BE, Wilkens LR. Alcohol consumption
and breast cancer risk among women from five ethnic groups with light to moderate intakes:
the Multiethnic Cohort Study. Int J Cancer. 2014;134(6):1504–10.
24. Bjerkaas E, Parajuli R, Weiderpass E, Engeland A, Maskarinec G, Selmer R, et al. Smoking
duration before first childbirth: an emerging risk factor for breast cancer? Results from 302,865
Norwegian women. Cancer Causes Control. 2013;24(7):1347–56.
25. Bloom JR, Stewart SL, Hancock SL. Breast cancer screening in women surviving Hodgkin
disease. Am J Clin Oncol. 2006;29(3):258–66.
26. Wolden SL, Hancock SL, Carlson RW, Goffinet DR, Jeffrey SS, Hoppe RT. Management of
breast cancer after Hodgkin's disease. J Clin Oncol. 2000;18(4):765–72.
27. Cutuli B, Borel C, Dhermain F, Magrini SM, Wasserman TH, Bogart JA, et al. Breast can-
cer occurred after treatment for Hodgkin's disease: analysis of 133 cases. Radiother Oncol.
2001;59(3):247–55.
28. Board WCoTE. Breast Tumours. Lyon: International Agency for Research on Cancer; 2019.
29. Hartmann LC, Degnim AC, Santen RJ, Dupont WD, Ghosh K. Atypical hyperplasia of the
breast--risk assessment and management options. N Engl J Med. 2015;372(1):78–89.
30. Said SM, Visscher DW, Nassar A, Frank RD, Vierkant RA, Frost MH, et al. Flat epithelial
atypia and risk of breast cancer: a Mayo cohort study. Cancer. 2015;121(10):1548–55.
31. To T, Wall C, Baines CJ, Miller AB. Is carcinoma in situ a precursor lesion of invasive breast
cancer? Int J Cancer. 2014;135(7):1646–52.
32. Ford D, Easton DF, Stratton M, et al. Genetic heterogeneity and penetrance analysis of the
BRCA1 and BRCA2 genes in breast cancer families. Am J Hum Genet. 1998;62:676–89.
33. Female breast cancer — cancer stat facts. Available at: https://seer.cancer.gov/statfacts/html/
breast.html. Accessed on 30/09/2020.
34. Genetics of Breast and Gynecologic Cancers (PDQ®)–Health professional version https://
www.cancer.gov/types/breast/hp/breast-ovarian-genetics-pdq.
35. Lynch HT, Watson P, Lynch JF. Epidemiology and risk factors. Clin Obstet Gynecol.
1989;32(4):750–60.
36. Giardiello FM, Brensinger JD, Tersmette AC, Goodman SN, et al. Very high risk of cancer in
familial Peutz–Jeghers syndrome. Gastroenterology. 2000;119(6):1447–53.
37. Tan MH, Mester JL, Ngeow J, Rybicki LA, et al. Lifetime cancer risks in individuals with
Germline PTEN mutations. Clin Cancer Res. 2012;18(2):400–7.
38. De GrèveJ, Sermijn E, Brakeleer SD, et al. Hereditary breast cancer from bench to bedside.
Curr Opin Oncol. 2008;20:605–13.
39. Kuchenbaecker KB, Hopper JL, Barnes DR, et al. Risks of breast, ovarian, and contralateral
breast cancer for BRCA1 and BRCA2 mutation carriers. JAMA. 2017;317(23):2402–16.
40. BRCA mutations: cancer risk and genetic testing fact sheet. Available at: https://www.cancer.
gov/about-cancer/causesprevention/genetics/brca-fact-sheet. Accessed on 30/09/2020.
41. Scalia-Wilbur J, Colins BL, Penson RT, Dizon DS. Breast cancer risk assessment: moving
beyond BRCA 1 and 2. Semin Radiat Oncol. 2016;26(1):3–8.
42. NCCN Guidelines. Available at: https://www.nccn.org/professionals/physician_gls/pdf/genet-
ics_bop.pdf. Accessed on 30/09/2020.
Chapter 3
Genomic Tests
3.1 Introduction
3.2.1 Oncotype DX
The Oncotype DX assay evaluates 21 genes (16 tumor associated and five controls)
and provides a recurrence score (RS) that ranges from 0 to 100. Depending on the
RS, patients are classified as low risk, intermediate risk, or high risk. Oncotype DX
was validated as a prognostication tool for estrogen receptor (ER)-positive early
breast cancer in various trials [1–4].
Among patients with node-negative ER-positive breast cancer enrolled in the
NSABP B-14 trial, recurrence scores (RS) of <18, 18–30, and ≥31 were associated
with 10-year distant recurrence rates of 6.8%, 14.3%, and 30.5%, respectively [1]. A
study with the population of another trial, the NSABP B-20, suggested that patients
in the low-risk category (RS < 18) derived no benefit from the addition of adjuvant
chemotherapy, while those with high risk (RS ≥ 31) had a significant reduction in
distant recurrence with chemotherapy. Finally, for patients in the intermediate-risk
category (RS 18–30), benefit from chemotherapy was uncertain [2].
The cutoff values for risk categories were refined in subsequent studies, and the
ability of Oncotype DX to predict chemotherapy benefit was evaluated prospec-
tively. The TAILORx trial enrolled patients with node-negative HR-positive breast
cancer, tailoring adjuvant systemic therapy according to RS. Patients with an RS
lower than 11 (low risk) were treated with endocrine therapy alone. Those with RS
greater than 25 (high risk) received chemotherapy plus endocrine therapy. Finally,
patients in the intermediate-risk category (RS 11–25) were randomized to receive
endocrine therapy alone or chemotherapy plus endocrine therapy. Results showed
similar outcomes from endocrine therapy and chemoendocrine therapy for patients
in the intermediate group [5]. The study confirmed that Oncotype DX can be used
to predict chemotherapy benefit.
Further analysis of the TAILORx trial suggested a different test behavior for
women younger than 50 years. For those in the intermediate-risk group, patients
with an RS of 16–25 seemed to benefit from chemoendocrine therapy. The absolute
distant recurrence rate decreased 1.6% for RS 16–20 and 6.4% for RS 21–25 at
9 years with the addition of chemotherapy. A question that remains is if this differ-
ence observed in younger women is due to the effect of chemotherapy itself or the
induction of menopause caused by it.
The RxPONDER trial evaluated the role of Oncotype DX for predicting che-
motherapy benefit for patients with one to three positive lymph nodes. Patients
with an RS ≤25 were assigned to receive either endocrine therapy or endocrine
therapy plus standard chemotherapy. No benefit was seen for chemotherapy in
postmenopausal women, but premenopausal women again had a different out-
come. Five-year invasive disease-free survival rates were 91.9% for endocrine
therapy plus chemotherapy vs 91.6% for endocrine therapy alone in the post-
menopausal patient subset and 94.2% vs 89.0% in the premenopausal patient
3 Genomic Tests 33
subset [6]. The question remains as to whether this is a direct benefit of chemo-
therapy or an indirect effect of ovarian suppression.
3.2.2 MammaPrint
The MammaPrint assay classifies HR-positive early breast cancer patients into two
categories, low risk and high risk, based on the evaluation of 70 genes. Retrospective
and prospective studies have validated MammaPrint as a prognostic tool for breast
cancer recurrence [7–9].
In the MINDACT trial, genomic risk based on MammaPrint was used together
with clinical risk to define the use of adjuvant chemotherapy for patients with
HR-positive breast cancer with negative lymph nodes or one to three positive lymph
nodes. The clinical risk was defined using Adjuvant! Online, a web-based tool that
provides recurrence risk according to clinical and pathological features [10].
Patients with low clinical and genomic risk received endocrine therapy alone, while
patients with high clinical and genomic risk received chemoendocrine therapy. The
cohort of patients with discordant results (low clinical risk and high genomic risk,
or high clinical risk and low genomic risk) were randomized to receive adjuvant
chemotherapy or not. The trial aimed to evaluate if patients with high clinical risk
and low genomic risk could be spared chemotherapy. Results showed that 5-year
distant metastasis-free survival (DMFS) rate was 94.7% (95% confidence interval
92.5–96.2%), which was considered satisfactory when compared with the noninfe-
riority boundary of 92% established by the study. Additionally, in this group, the
5-year distant metastasis rate of survival (DMRS) was similar in patients with che-
motherapy and without chemotherapy. The study suggested that MammaPrint can
be used for patients with high clinical risk to select patients for whom chemotherapy
can be avoided, although the trial was not powered for this comparison (yes versus
no chemotherapy) [11].
Similar to what was observed with both randomized Oncotype DX trials, sub-
group analysis of MINDACT suggested that for women younger than 50 years with
high clinical risk and low genomic risk, a relevant absolute difference of 5% in
8-year DMFS was observed with the addition of chemotherapy [12]. Thus, for
women younger than 50 years, additional studies are necessary to clarify the role of
genomic assays for predicting chemotherapy benefit.
EndoPredict assay analyzes 12 genes (eight cancer-related genes and four reference
genes), providing a risk score ranging from 0 to 15. The test also classifies patients
into low- and high-risk groups. Moreover, an EndoPredict clinical score is
34 L. Testa and R. C. Bonadio
Although genomic tests available have similar objectives, they differ in terms of
genes evaluated and population in which the tests were validated. Importantly, dis-
cordances have been demonstrated among the tests. For instance, Barlett et al.
showed that in a cohort of patients with early breast cancer, the proportion consid-
ered as low/intermediate risk was 82.1%, 65.6%, and 61.4% with Oncotype DX,
Prosigna, and MammaPrint, respectively [24]. In another study, the concordance
between Oncotype DX and MammaPrint was only 0.64 [25].
These results are similar to what was observed in a review that summarized head-
to-head comparisons of Oncotype DX with other genomic tests. In this review, a
high-risk score was observed: 11.5% with Oncotype DX, 16.2% with BCI, 30.7%
with ROR (Prosigna), 63% with EndoPredict, and 45.8% with MammaPrint. Overall
discordance between Oncotype DX and other tests ranged from 42% to 66% [26].
Thus, genomic tests for recurrence risk are not interchangeable, and careful selec-
tion of the appropriate test for a patient is necessary. A summary of these tests’
characteristics is shown in Table 3.1.
3 Genomic Tests 35
Table 3.1 Summary of the characteristics of genomic tests for breast cancer recurrence risk
Type of
Number Risk Population Current validation
of genes categories evaluated validation studies
Oncotype DX 21 Low ER+/HER2- Prognostic Prospective-
Intermediate pT1-2 Predictive of retrospective
High pN0-1 chemotherapy [1–4]
benefit Prospective
randomized [5,
6]
MammaPrint 70 Low pT1-2 Prognostic Prospective-
High pN0-1 Predictive of retrospective
chemotherapy [7–9]
benefit Prospective
randomized [11]
EndoPredict 12 Low ER+/HER2- Prognostic Prospective-
High pT1-2 retrospective
pN0-1 [13–15]
Postmenopausal
Prosigna 58 Low ER+ Prognostic Prospective-
(PAM50) Intermediate pT1-2 retrospective
High pN0-1 [16–18]
Postmenopausal
Breast Cancer 11 Low ER+/HER2- Prognostic Prospective-
Index High pT1-3 retrospective
pN0 [20–23]
3.4 Conclusion
Genomic tests are adding valuable information on recurrence risk for ER/PR-positive
early breast cancer. Their use to estimate prognosis and predict the benefit of adju-
vant therapy has been increasing in clinical practice [27]. Nevertheless, some chal-
lenges still need to be faced.
Despite the number of tests available, they were validated in different scenarios
and are not totally concordant with one other. Carefulness is necessary for the
appropriate test selection for a patient. More prospective validation is still required
for some groups, such as premenopausal women and patients with positive lymph
nodes. Moreover, tests’ costs remain a barrier for wider access to these tests.
References
1. Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, et al. A multigene assay to predict recur-
rence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004;351(27):2817–26.
2. Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, et al. Gene expression and benefit of chemo-
therapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol.
2006;24(23):3726–34.
36 L. Testa and R. C. Bonadio
3. Dowsett M, Cuzick J, Wale C, Forbes J, Mallon EA, Salter J, et al. Prediction of risk of distant
recurrence using the 21-gene recurrence score in node-negative and node-positive postmeno-
pausal patients with breast cancer treated with anastrozole or tamoxifen: a TransATAC study.
J Clin Oncol. 2010;28(11):1829–34.
4. Albain KS, Barlow WE, Shak S, Hortobagyi GN, Livingston RB, Yeh IT, et al. Prognostic and
predictive value of the 21-gene recurrence score assay in postmenopausal women with node-
positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis
of a randomised trial. Lancet Oncol. 2010;11(1):55–65.
5. Francis PA, Pagani O, Fleming GF, Walley BA, Colleoni M, Láng I, et al. Tailoring adjuvant
endocrine therapy for premenopausal breast cancer. N Engl J Med. 2018;379(2):122–37.
6. Kalinsky K, Barlow W, Meric-Bernstam F, et al.. SWOG S1007: adjuvant trial randomized
ER+ patients who had a recurrence score < 25 and 1–3 positive nodes to endocrine therapy
(ET) versus ET + chemotherapy. Presented at: 2020 virtual San Antonio breast cancer sympo-
sium; December 8–11, 2020. Abstract GS3-00.
7. van de Vijver MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW, et al. A gene-expression
signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347(25):1999–2009.
8. Buyse M, Loi S, van't Veer L, Viale G, Delorenzi M, Glas AM, et al. Validation and clinical
utility of a 70-gene prognostic signature for women with node-negative breast cancer. J Natl
Cancer Inst. 2006;98(17):1183–92.
9. Bueno-de-Mesquita JM, van Harten WH, Retel VP, van't Veer LJ, van Dam FS, Karsenberg
K, et al. Use of 70-gene signature to predict prognosis of patients with node-negative
breast cancer: a prospective community-based feasibility study (RASTER). Lancet Oncol.
2007;8(12):1079–87.
10. Olivotto IA, Bajdik CD, Ravdin PM, Speers CH, Coldman AJ, Norris BD, et al. Population-
based validation of the prognostic model ADJUVANT! for early breast cancer. J Clin Oncol.
2005;23(12):2716–25.
11. Cardoso F, van't Veer LJ, Bogaerts J, Slaets L, Viale G, Delaloge S, et al. 70-gene signature as
an aid to treatment decisions in early-stage breast cancer. N Engl J Med. 2016;375(8):717–29.
12. Cardoso F, van't Veer L, Poncet C, et al. MINDACT: long-term results of the large prospec-
tive trial testing the 70-gene signature MammaPrint as guidance for adjuvant chemotherapy in
breast cancer patients. J Clin Oncol. 2020;38(suppl; abstr):506.
13. Filipits M, Rudas M, Jakesz R, Dubsky P, Fitzal F, Singer CF, et al. A new molecular predictor
of distant recurrence in ER-positive, HER2-negative breast cancer adds independent informa-
tion to conventional clinical risk factors. Clin Cancer Res. 2011;17(18):6012–20.
14. Dubsky P, Filipits M, Jakesz R, Rudas M, Singer CF, Greil R, et al. EndoPredict improves
the prognostic classification derived from common clinical guidelines in ER-positive, HER2-
negative early breast cancer. Ann Oncol. 2013;24(3):640–7.
15. Martin M, Brase JC, Calvo L, Krappmann K, Ruiz-Borrego M, Fisch K, et al. Clinical valida-
tion of the EndoPredict test in node-positive, chemotherapy-treated ER+/HER2- breast cancer
patients: results from the GEICAM 9906 trial. Breast Cancer Res. 2014;16(2):R38.
16. Dowsett M, Sestak I, Lopez-Knowles E, Sidhu K, Dunbier AK, Cowens JW, et al. Comparison
of PAM50 risk of recurrence score with oncotype DX and IHC4 for predicting risk of distant
recurrence after endocrine therapy. J Clin Oncol. 2013;31(22):2783–90.
17. Gnant M, Filipits M, Greil R, Stoeger H, Rudas M, Bago-Horvath Z, et al. Predicting distant
recurrence in receptor-positive breast cancer patients with limited clinicopathological risk:
using the PAM50 risk of recurrence score in 1478 postmenopausal patients of the ABCSG-8
trial treated with adjuvant endocrine therapy alone. Ann Oncol. 2014;25(2):339–45.
18. Liu S, Chapman JA, Burnell MJ, Levine MN, Pritchard KI, Whelan TJ, et al. Prognostic and
predictive investigation of PAM50 intrinsic subtypes in the NCIC CTG MA.21 phase III che-
motherapy trial. Breast Cancer Res Treat. 2015;149(2):439–48.
19. Sestak I, Cuzick J, Dowsett M, Lopez-Knowles E, Filipits M, Dubsky P, et al. Prediction of
late distant recurrence after 5 years of endocrine treatment: a combined analysis of patients
from the Austrian breast and colorectal cancer study group 8 and arimidex, tamoxifen alone
3 Genomic Tests 37
or in combination randomized trials using the PAM50 risk of recurrence score. J Clin Oncol.
2015;33(8):916–22.
20. Sgroi DC, Sestak I, Cuzick J, Zhang Y, Schnabel CA, Schroeder B, et al. Prediction of late
distant recurrence in patients with oestrogen-receptor-positive breast cancer: a prospective
comparison of the breast-cancer index (BCI) assay, 21-gene recurrence score, and IHC4 in the
TransATAC study population. Lancet Oncol. 2013;14(11):1067–76.
21. Sgroi DC, Carney E, Zarrella E, Steffel L, Binns SN, Finkelstein DM, et al. Prediction of
late disease recurrence and extended adjuvant letrozole benefit by the HOXB13/IL17BR bio-
marker. J Natl Cancer Inst. 2013;105(14):1036–42.
22. Zhang Y, Schnabel CA, Schroeder BE, Jerevall PL, Jankowitz RC, Fornander T, et al. Breast
cancer index identifies early-stage estrogen receptor-positive breast cancer patients at risk for
early- and late-distant recurrence. Clin Cancer Res. 2013;19(15):4196–205.
23. Sanft T, Aktas B, Schroeder B, Bossuyt V, DiGiovanna M, Abu-Khalaf M, et al. Prospective
assessment of the decision-making impact of the breast cancer index in recommending
extended adjuvant endocrine therapy for patients with early-stage ER-positive breast cancer.
Breast Cancer Res Treat. 2015;154(3):533–41.
24. Bartlett JM, Bayani J, Marshall A, Dunn JA, Campbell A, Cunningham C, et al. Comparing
breast cancer multiparameter tests in the OPTIMA prelim trial: no test is more equal than the
others. J Natl Cancer Inst. 2016;108(9):djw050.
25. Denduluri N, Rugo H, Davis S, et al. Concordance between the 21-gene recurrence score
(RS) and the 70-gene profile (MP) in breast cancer (BC) patients (pts). J Clin Oncol.
2011;27_suppl:13.
26. Varga Z, Sinn P, Seidman AD. Summary of head-to-head comparisons of patient risk clas-
sifications by the 21-gene Recurrence Score® (RS) assay and other genomic assays for early
breast cancer. Int J Cancer. 2019;145(4):882–93.
27. Duffy MJ, Harbeck N, Nap M, Molina R, Nicolini A, Senkus E, et al. Clinical use of biomark-
ers in breast cancer: updated guidelines from the European Group on Tumor Markers (EGTM).
Eur J Cancer. 2017;75:284–98.
Chapter 4
Updates in Surgical Approaches for Breast
and Axilla
4.1 Introduction
Axillary surgery provides valuable staging information to guide the adjuvant treat-
ment of breast cancer and is key to the local control of axillary disease, being a
well-established procedure for the management of primary breast cancer.
However, the surgical management of the axilla has changed substantially in the
last three decades, from the axillary lymph node dissection (ALND), which has a
high morbidity and decreases the women’s quality of life, to a sentinel lymph node
biopsy for most patients.
This radical change in the surgical approach started in 1970; the Mayo Clinic
published strong evidence for the axillary nodal metastases predictive values. All
patients had undergone ALND, and the data showed 5-year survival rates of 86% in
node-negative patients compared to 50% in node-positive patients. After that, the
value of axillary clearance for all breast cancer surgery became questionable since
it did not influence the survival rates and increased the chances of lymphedema [1].
In 1994, Giuliano et al. [2], after the successful use of sentinel lymph node
biopsy (SLNB) for melanoma staging [3], published the first description of SLNB
for breast cancer treatment, with the potential of axillary staging with less morbid-
ity. Since then, its use has been widely accepted as the single axillary surgical pro-
cedure among all clinically node-negative (cN0) breast cancer patients and selected
patients with low-volume axillary metastases. Those results significantly impacted
breast cancer surgery management since it avoids as much as 70% of all complete
axillary node dissections.
We will discuss in this chapter the surgical management of the axilla according
to the axillary stage: N (0), micrometastases (N1mic) or isolated tumor cells, clini-
cally N0 with macrometastases in the pathological exam, and clinically positive
axilla patients. This approach aims to facilitate the understanding of axillary treat-
ment in different scenarios.
The effectiveness of SLNB in clinically N0 breast cancer patients has been proven
throughout the years with the publication of a comprehensive study assortment.
These trials all have demonstrated low rates of axillary recurrences (0.2–0.5%) [4–
7] when SLNB was compared to ALND (0–0.8%) in patients with a negative SLNB
who had no further surgery.
The National Surgical Adjuvant Breast and Bowel Project (NSABP) B-32 trial is
the most extensive study, with 5611 participants, to compare survival and regional
control between SLNB alone and axillary lymph node dissection (ALND) in clini-
cally node-negative patients. Patients were assigned to SLNB plus ALND or SLNB
alone with ALND only if the sentinel node(s) was positive [5]. The latest report, in
2010, showed no difference in overall and disease-free survival between the two
groups with a median follow-up of 8 years. Axillary recurrence as a first disease
relapse event occurred in less than 1% of both the SLNB followed by ALND group
(0.4%) and the SLNB-only group (0.6%), p = 0.22 [8].
In 2017, a meta-analysis from the Cochrane database, including seven clinical
trials, compared ALND versus SLNB. There was no difference in overall survival in
the comparison between SLNB and ALND (HR 1.05, 95% CI 0.89 to 1.25; 6352
participants; three studies; moderate-quality evidence) [9].
The pathology review of nodal specimens of SLNB negative patients in the NSABP
B-32 trial presented the question about the role of ALND in the axillary treatment
in micrometastases (N1m1) or isolated tumor cells N (+1) (ITC) patients. The abso-
lute reduction in overall survival in those patients with ITC or micrometastases was
only 1.2% in patients submitted to ALND in that trial [5].
The ACOSOG Z0010 trial confirmed that women with a low volume of nodal
metastases who underwent an SLNB plus ALND did not have significant benefit
in overall or disease-free survival over patients undergoing SLNB alone in this
4 Updates in Surgical Approaches for Breast and Axilla 41
multicentric prognostic study, which had 10.5% rates of occult sentinel node
metastases [10].
The International Breast Cancer Study Group (IBCSG) 23-01 trial answered the
question regarding the role of axillary surgery in breast cancer patients with N1mic
definitively. In this trial, clinically node-negative patients with an SLNB containing
micrometastases were randomized to ALND or no further axillary surgery. Thirteen
percent of patients who had an ALND presented additional involved axillary nodes.
However, 5-year disease-free survival did not significantly differ in the SLNB-alone
(87.8%) vs. the ALND (84.4%) group. In patients who did not undergo ALND, the
rate of disease recurrence was less than 1% [11].
In the past, the standard of care for patients who had positive sentinel lymph nodes
with macrometastases (tumor deposits of more than 2.0 mm) was ALND to achieve
local control. However, with the increasing use and efficacy of adjuvant chemo-
therapy, hormonal therapy, and radiation, the scientific community started question-
ing the real importance of ALND in providing regional control.
The American College of Surgeons Oncology Group Z0011 trial recruited
patients with clinically T1 to T2, node-negative breast cancer selected to undergo
breast-conserving surgery and SLNB. All patients who had a positive SLNB by
routine H&E staining were randomized to ALND completion or no ALND and no
further axillary specific radiotherapy. Any patient with three or more positive senti-
nel nodes was excluded. Accrual to the study and event rates were lower than
expected, resulting in the trial’s early closure. All patients received whole-breast
irradiation, and almost all received adjuvant systemic therapy (58% chemotherapy,
46% hormonal therapy). Forty-one percent of the study patients had small volume
metastases (micrometastases or ITCs). In the cohort who did undergo an ALND,
additional positive axillary nodes were found in 27% of cases. At a median follow-
up of 6 years, there were no differences between the SLNB followed by ALND and
SLNB groups in the rates of overall locoregional recurrence (4.1% vs. 2.8%), breast
recurrence (3.6% vs. 1.9%), and axillary recurrence (0.5% vs. 0.9). The 10-year
follow-up publication confirmed the low rate of axillary recurrence of <2% in both
groups, denying any concerns that a spate of late axillary recurrences may be seen
with the SLNB-only group [12]. The trial’s major weakness focused on the design
of the radiotherapy fields; the tangential field whole-breast irradiation with high
tangents used in both groups resulted in a portion of the axilla receiving radiation in
patients on both arms of the study. However, trying to evaluate the radiotherapy as
a confounder, further statistical analysis did not show a significant difference
between treatment arms in the use of protocol-prohibited nodal fields [13].
42 B. S. Mota et al.
response. Caudle et al., in 2017, conducted a prospective study, including 208 par-
ticipants, using iodine-125 seed localization. The marked node was not recovered as
an SLN in 23% (31 of 134) of patients, including six with negative SLNs but metas-
tasis in the clipped node. A total of 120 patients still had residual disease; from
those, the clipped node showed metastases in 115 patients, resulting in a false-
negative rate of 4.2% [29].
The routine use of SLNB using iodine-125 seed localization has not been estab-
lished yet since there is an increase in treatment costs and questions regarding the
technique due to the need of specialized breast radiologists, notably experienced in
targeting techniques [29]. Otherwise, the trials published regarding this theme sup-
port the management of SLND with blue dye and radiocolloid with acceptable
false-negative rates.
Although there are increasing efforts to avoid the ALND in patients identified as
node-positive before systemic treatment that converted to negative after NAC, there
are still almost 40% of patients who have residual axillary disease and need ALND
following standard adjuvant treatment. The ALND is still the standard treatment in
this setting; there is no scientific data to support radiotherapy as a unique treatment
for this group of patients. The updated analysis of NSABP B-18 and B-27 neoadju-
vant trials showed high rates of locoregional recurrence (LRR) observed in patients
with pathologically positive nodes at surgery. The 10-year cumulative incidence of
LRR was 12.3% for mastectomy patients and 10.3% for lumpectomy plus breast
radiotherapy patients. The independent predictors of LRR in lumpectomy patients
were age, clinical nodal status (before NAC), and pathologic nodal status/breast
tumor response. In mastectomy patients, predictors of LRR were clinical tumor size
(before NAC), clinical nodal status (before NAC), and pathologic nodal status/
breast tumor response. The risk of LRR was increased by 2.71 times if residual axil-
lary disease was present [30].
The Alliance A11202 study is a randomized clinical trial that evaluates the role
of axillary radiotherapy (axilla, supraclavicular nodes, and internal mammary
nodes) compared to ALND in patients who remain node-positive after NAC irre-
spective of the breast surgery type. The main outcome of this trial is recurrence-free
survival rate in a non-inferiority design.
We will still observe extraordinary advances and practice changes in axilla man-
agement for the next decades due to preoperative image improvement and profile
signatures. There are two exciting trials in the recruitment phase: the SOUND trial
[31] and the SWOG RxPONDER trial [32]. In the first one, the research question is
based on patients with a low burden of axillary disease, with a hypothesis that the
results of SLNB are unlikely to alter adjuvant treatment decisions. Regarding this,
the patients will be randomized to perform SLNB versus ultrasound only. The sec-
ond protocol, SWOG RxPONDER trial, will assess gene expression profiling’s abil-
ity in the setting of lymph node positivity to identify patients with excellent predicted
long-term survival with adjuvant hormonal treatment alone in which chemotherapy
would not confer any additional benefit.
4 Updates in Surgical Approaches for Breast and Axilla 45
References
1. Fisher B, Montague E, Redmond C, Barton B, Borland D, Fisher ER, et al. Comparison of rad-
ical mastectomy with alternative treatments for primary breast cancer. A first report of results
from a prospective randomized clinical trial. Cancer. 1977;39(6 Suppl):2827–39.
2. Giuliano AE, Kirgan DM, Guenther JM, Morton DL. Lymphatic mapping and sentinel lymph-
adenectomy for breast cancer. Ann Surg. 1994;220(3):391–8. discussion 398–401.
3. Morton DL, Wen DR, Wong JH, Economou JS, Cagle LA, Storm FK, et al. Technical details of
intraoperative lymphatic mapping for early stage melanoma. Arch Surg. 1992;127(4):392–9.
4. Mansel RE, Fallowfield L, Kissin M, Goyal A, Newcombe RG, Dixon JM, et al. Randomized
multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast
cancer: the ALMANAC trial. J Natl Cancer Inst. 2006;98(9):599–609.
5. Krag DN, Julian TB, Harlow SP, Weaver DL, Ashikaga T, Bryant J, et al. NSABP-32: phase
III, randomized trial comparing axillary resection with sentinal lymph node dissection: a
description of the trial. Ann Surg Oncol. 2004;11(3 Suppl):208S–10S.
6. Zavagno G, De Salvo GL, Scalco G, Bozza F, Barutta L, Del Bianco P, et al. A randomized
clinical trial on sentinel lymph node biopsy versus axillary lymph node dissection in breast
cancer: results of the Sentinella/GIVOM trial. Ann Surg. 2008;247(2):207–13.
7. Veronesi U, Paganelli G, Viale G, Luini A, Zurrida S, Galimberti V, et al. A randomized com-
parison of sentinel-node biopsy with routine axillary dissection in breast cancer. N Engl J Med.
2003;349(6):546–53.
8. Krag DN, Anderson SJ, Julian TB, Brown AM, Harlow SP, Costantino JP, et al. Sentinel-
lymph-node resection compared with conventional axillary-lymph-node dissection in clini-
cally node-negative patients with breast cancer: overall survival findings from the NSABP
B-32 randomised phase 3 trial. Lancet Oncol. 2010;11(10):927–33.
9. Bromham N, Schmidt-Hansen M, Astin M, Hasler E, Reed MW. Axillary treatment for oper-
able primary breast cancer. Cochrane Database Syst Rev. 2017;1:CD004561.
10. Giuliano AE, Ballman K, McCall L, Beitsch P, Whitworth PW, Blumencranz P, et al.
Locoregional recurrence after sentinel lymph node dissection with or without axillary dissec-
tion in patients with sentinel lymph node metastases: long-term follow-up from the American
College of Surgeons Oncology Group (Alliance) ACOSOG Z0011 randomized trial. Ann Surg.
2016;264(3):413–20.
11. Galimberti V, Cole BF, Viale G, Veronesi P, Vicini E, Intra M, et al. Abstract GS5-02: axillary
dissection vs. no axillary dissection in patients with cT1-T2cN0M0 breast cancer and only
micrometastases in the sentinel node(s): ten-year results of the IBCSG 23-01 trial. Cancer Res.
2018;78(4 Supplement):GS5-02-GS5-02.
12. Giuliano AE, Ballman KV, McCall L, Beitsch PD, Brennan MB, Kelemen PR, et al. Effect of
axillary dissection vs no axillary dissection on 10-year overall survival among women with
invasive breast cancer and sentinel node metastasis: the ACOSOG Z0011 (Alliance) random-
ized clinical trial. JAMA. 2017;318(10):918–26.
13. Haffty BG, McCall LM, Ballman KV, McLaughlin S, Jagsi R, Ollila DW, et al. Patterns of
local-regional management following neoadjuvant chemotherapy in breast cancer: results
from ACOSOG Z1071 (Alliance). Int J Radiat Oncol Biol Phys. 2016;94(3):493–502.
14. Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, et al. Breast cancer,
version 3.2020, NCCN clinical practice guidelines in oncology. J Natl Compr Cancer Netw.
2020;18(4):452–78.
15. Straver ME, Meijnen P, van Tienhoven G, van de Velde CJH, Mansel RE, Bogaerts J, et al.
Sentinel node identification rate and nodal involvement in the EORTC 10981-22023 AMAROS
trial. Ann Surg Oncol. 2010;17(7):1854–61.
16. Rutgers EJ, Donker M, Poncet C, Straver ME, Meijnen P, van de Velde CJ, et al. Abstract
GS4-01: radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer
patients: 10 year follow up results of the EORTC AMAROS trial (EORTC 10981/22023).
Cancer Res. 2019;79(4 Supplement):GS4-01-GS4-01.
46 B. S. Mota et al.
17. Singletary SE, Connolly JL. Breast cancer staging: working with the sixth edition of the AJCC
Cancer staging manual. CA Cancer J Clin. 2006 Jan;56(1):37–47; quiz 50–1.
18. Mamtani A, Barrio AV, King TA, Van Zee KJ, Plitas G, Pilewskie M, et al. How often does
neoadjuvant chemotherapy avoid axillary dissection in patients with histologically confirmed
nodal metastases? Results of a prospective study. Ann Surg Oncol. 2016;23(11):3467–74.
19. Schwentner L, Helms G, Nekljudova V, Ataseven B, Bauerfeind I, Ditsch N, et al. Using ultra-
sound and palpation for predicting axillary lymph node status following neoadjuvant chemo-
therapy - results from the multi-center SENTINA trial. Breast. 2017;31:202–7.
20. Geng C, Chen X, Pan X, Li J. The feasibility and accuracy of sentinel lymph node biopsy in
initially clinically node-negative breast cancer after neoadjuvant chemotherapy: a systematic
review and meta-analysis. PLoS One. 2016;11(9):e0162605.
21. Lee S, Kim EY, Kang SH, Kim SW, Kim S-K, Kang KW, et al. Sentinel node identification
rate, but not accuracy, is significantly decreased after pre-operative chemotherapy in axillary
node-positive breast cancer patients. Breast Cancer Res Treat. 2007;102(3):283–8.
22. Shen J, Gilcrease MZ, Babiera GV, Ross MI, Meric-Bernstam F, Feig BW, et al. Feasibility
and accuracy of sentinel lymph node biopsy after preoperative chemotherapy in breast cancer
patients with documented axillary metastases. Cancer. 2007;109(7):1255–63.
23. Alvarado R, Yi M, Le-Petross H, Gilcrease M, Mittendorf EA, Bedrosian I, et al. The role for
sentinel lymph node dissection after neoadjuvant chemotherapy in patients who present with
node-positive breast cancer. Ann Surg Oncol. 2012;19(10):3177–84.
24. Newman EA, Sabel MS, Nees AV, Schott A, Diehl KM, Cimmino VM, et al. Sentinel lymph
node biopsy performed after neoadjuvant chemotherapy is accurate in patients with docu-
mented node-positive breast cancer at presentation. Ann Surg Oncol. 2007;14(10):2946–52.
25. Boughey JC, Suman VJ, Mittendorf EA, Ahrendt GM, Wilke LG, Taback B, et al. Factors
affecting sentinel lymph node identification rate after neoadjuvant chemotherapy for breast
cancer patients enrolled in ACOSOG Z1071 (Alliance). Ann Surg. 2015;261(3):547–52.
26. Boughey JC, Suman VJ, Mittendorf EA, Ahrendt GM, Wilke LG, Taback B, et al. Sentinel
lymph node surgery after neoadjuvant chemotherapy in patients with node-positive breast can-
cer: the ACOSOG Z1071 (Alliance) clinical trial. JAMA. 2013;310(14):1455–61.
27. Kuehn T, Bauerfeind I, Fehm T, Fleige B, Hausschild M, Helms G, et al. Sentinel-lymph-node
biopsy in patients with breast cancer before and after neoadjuvant chemotherapy (SENTINA):
a prospective, multicentre cohort study. Lancet Oncol. 2013;14(7):609–18.
28. Boileau J-F, Poirier B, Basik M, Holloway CMB, Gaboury L, Sideris L, et al. Sentinel node
biopsy after neoadjuvant chemotherapy in biopsy-proven node-positive breast cancer: the SN
FNAC study. J Clin Oncol. 2015;33(3):258–64.
29. Caudle AS, Yang WT, Krishnamurthy S, Mittendorf EA, Black DM, Gilcrease MZ, et al.
Improved axillary evaluation following neoadjuvant therapy for patients with node-positive
breast cancer using selective evaluation of clipped nodes: implementation of targeted axillary
dissection. J Clin Oncol. 2016;34(10):1072–8.
30. Mamounas EP, Anderson SJ, Dignam JJ, Bear HD, Julian TB, Geyer CE Jr, et al. Predictors
of locoregional recurrence after neoadjuvant chemotherapy: results from combined analy-
sis of National Surgical Adjuvant Breast and Bowel Project B-18 and B-27. J Clin Oncol.
2012;30(32):3960–6.
31. Gentilini O, Veronesi U. Abandoning sentinel lymph node biopsy in early breast cancer? A
new trial in progress at the European Institute of Oncology of Milan (SOUND: sentinel node
vs observation after axillary UltraSouND). Breast. 2012;21(5):678–81.
32. Ramsey SD, Barlow WE, Gonzalez-Angulo AM, Tunis S, Baker L, Crowley J, et al. Integrating
comparative effectiveness design elements and endpoints into a phase III, randomized clini-
cal trial (SWOG S1007) evaluating oncotypeDX-guided management for women with breast
cancer involving lymph nodes. Contemp Clin Trials. 2013;34(1):1–9.
Chapter 5
Pathological Aspects for Diagnosis
Fixation of biopsies and surgical specimens in neutral buffered 10% formalin must
be immediate, and an adequate volume of fixative (at least ten times the volume of
the specimen – for biopsy fragments, about 10 ml) should be used for histological,
immunohistochemical, in situ hybridization, and molecular studies (e.g., Oncotype
DX). Large specimens should be sectioned in slices with a maximum thickness of
10 mm for adequate fixation.
Bar code identification and individual analysis in each step of histological pro-
cessing are recommended to avoid errors.
The pathologist must have access to lesion imaging characteristics and clinical data.
If the purpose of the biopsy is to study calcifications, the identification and segrega-
tion of biopsy fragments with calcifications may facilitate analysis and radiological-
pathological correlation. Some rules are recommended to avoid mistakes:
M. A. Giannotti (*)
Hospital Sírio Libanês and Departamento de Patologia, Hospital das Clinicas HCFMUSP,
Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
e-mail: [email protected]
F. N. Aguiar
Departamento de Patologia, Instituto do Cancer do Estado de Sao Paulo ICESP,
São Paulo, SP, Brazil
e-mail: [email protected]
Fibroadenomas and phyllodes tumors are lesions that exhibit proliferation of epithe-
lial and stromal cells. The architectural aspects of stromal growth can be divided
into two patterns. In the peri-canalicular pattern, the stroma grows around rounded
tubules, while in the intracanalicular pattern, the stroma compresses the ducts and
forms arciform slit-like structures with epithelial revestment. In phyllodes tumors,
this intracanalicular pattern is exaggerated and associated to stromal hypercellular-
ity, with formation of leaf-like structures.
Fibroadenomas are benign lesions, may be multiple, and may increase with hor-
monal stimuli. If asymptomatic, excision of fibroadenomas is not required.
Phyllodes tumors are classified according to some histological findings: its cel-
lularity, atypia, the presence of stromal overgrowth (i.e., areas greater than a lower
magnification field with only stromal component), growth pattern (permeative x well
defined), the presence of malignant heterologous elements, and, mainly, by their
mitotic count. Although benign phyllodes tumors and fibroadenomas may have
molecular differences [1], they share molecular alterations (e.g., MED12-mutant
pathway) and recurrence rates, even when margins are positive [2]. In core needle
5 Pathological Aspects for Diagnosis 49
With the improvement of biopsy devices, i.e., vacuum-assisted biopsies (VAB), the
ratio of false-negative results in percutaneous biopsies has been reduced, and surgi-
cal approach to avoid underestimation is no longer recommended for some high-
risk lesions [3], i.e., lesions with risk for subsequent development of breast cancer
lower than 2.5-fold. When appropriate sampling is achieved on a percutaneous
biopsy, patients diagnosed with papilloma, radial scar, mucocele-like lesion, flat
epithelial atypia, lobular atypical hyperplasia, and classic in situ lobular carcinoma
may be spared of surgical intervention. It is very important, however, before adopt-
ing a conservative approach, to verify if pathological and radiological aspects are
compatible; for example, the diagnosis of in situ lobular carcinoma in a nodular
lesion on mammogram should be considered inappropriate. The sampled proportion
of the lesion should also be regarded. Large lesions, i.e., larger than 10 mm, will
5 Pathological Aspects for Diagnosis 51
Fig. 5.3 Ductal carcinoma in situ (DCIS) of intermediate nuclear grade (grade 2)
Fig. 5.4 Ductal carcinoma in situ (DCIS) of high nuclear grade (grade 3)
variably prominent nucleoli [4] (Fig. 5.3). Necrosis and microcalcifications in simi-
lar patterns seen in low- or high-grade DCIS may be present. By sharing some his-
tological characteristics with low- and/or high-grade DCIS, it is no surprise that
intermediate-grade DCIS has the least agreement between pathologists [5]. Studies
show that the agreement is better when only two categories, low or high grade, are
considered [6, 7]. Molecular studies support this as the intermediate-grade DCIS
separates into these two and not into an individual category [8].
In summary, DCIS of intermediate grade can show any histological characteris-
tics and immunohistochemical profile, overlapping with low- or high-grade DCIS.
High-grade DCIS presents highly atypical cells with pleomorphic nuclei, irregu-
lar contours, clumped chromatin, and prominent nucleoli [4]. Comedonecrosis is
frequently seen, although not obligatory. Mitotic figures are common. High-grade
DCIS may present as a mass or calcifications on mammography and as an enhance-
ment on magnetic resonance imaging (MRI) [9] (Fig. 5.4).
The pleomorphic LCIS variant has inactivation of e-cadherin which can be con-
firmed by immunohistochemistry, lacking membrane expression of e-cadherin and
B-catenin and showing cytoplasmic expression of p120 [10]. Histologically, they
are more similar to high-grade DCIS, showing higher nuclear grade and
5 Pathological Aspects for Diagnosis 53
comedonecrosis [11, 12] (Fig. 5.5). Compared with classic LCIS, PLCIS shows less
expression of hormone receptors (estrogen and progesterone) and more chance of
overexpression of HER2 [10, 11, 13].
PLCIS shares some genetic similarities with classic LCIS, including 16q loss
and 1q gain, but might show a higher number of genetic alterations like gain of 16p,
loss of 8p, and amplification of cyclin D1 gene [11, 14]. Biological behavior of
PLCIS is more similar to high-grade DCIS [15].
Table 5.3 Nottingham criteria for invasive carcinomas of no special type (NST)
Feature Score
Tubule and gland formation
Majority of tumor (>75%) 1
Moderate degree (10–75%) 2
Little or none (<10%) 3
Nuclear pleomorphism
Small, regular, uniform cells 1
Moderate increase in size and variability 2
Marked variation 3
Mitotic count (dependent on microscopic field
area)
Low 1
Moderate 2
High 3
Total score (add the scores) Final grading
3–5 Grade 1
6 or 7 Grade 2
8 or 9 Grade 3
56 M. A. Giannotti and F. N. Aguiar
predictive factors [49]. Although the gold standard for identification of these classes
is the molecular tests, in practice, immunohistochemical profile shows a good cor-
relation with those [50, 51]. Based on that, the main molecular classes of invasive
breast carcinoma can be correlated as this:
• Luminal A or B: carcinomas that express hormone receptors (ER and/or PR),
without overexpression of HER2; the division between A and B has many
options, but most incorporate Ki67 and the level of progesterone receptor expres-
sion – we follow this group criteria [52] (Table 5.4).
• Hybrid luminal/HER2: carcinomas that express hormone receptor (ER and/or
PR) associated with overexpression of HER2 (Fig. 5.8).
• HER2: carcinomas that overexpress HER2 without expression of hormone
receptors.
• Triple negative (TN): carcinomas that do not express hormone receptors
nor HER2.
Invasive carcinomas of no special type are present in all of those groups, even
though we can see some patterns of characteristics in some of them. Luminal A
carcinomas are mostly Nottingham grade 1 with some grade 2. Hybrid, HER2, and
Table 5.4 Immunohistochemical profile and its correlation with molecular classes of invasive
breast cancer
Luminal A All of:
ER positive
HER2 negative
And at least one of:
Ki67 <14%
Ki67 14–19% and PR ≥20%
Luminal B (HER2 negative) All of:
ER positive
HER2 negative
And at least one of:
Ki67 14–19% and PR negative or <20%
Ki67 ≥20%
TN are mostly grade 2 or 3 [53]. Special histological types show a more specific
association with those classes: classic invasive lobular carcinoma is almost always
luminal, predominantly A, and Nottingham grade 1 or 2. Pleomorphic lobular car-
cinoma can be luminal B (majority), but also hybrid, HER2, or even TN, being most
Nottingham grade 2 or 3. Metaplastic and medullary carcinomas are TN and
Nottingham grade 2 or 3. Some special types of carcinomas, such as adenoid cystic
and secretory, can be TN and have a Nottingham low grade, but they are rare
carcinomas.
With the improvement in the knowledge on breast carcinomas, its histological
evaluation must have complementary immunohistochemistry to better understand
its biology, predict the behavior, and offer adequate treatment.
General rules for evaluation of breast specimens are well established and available
in various publications like the College of American Pathologists [54] and other
papers [55–57]. Here, we would like to reinforce some fundamental aspects in an
attempt to better evaluate and obtain the best possible report for the other specialties
that receive the anatomopathological summary.
The pathologist must have access to all data regarding the clinical, imaginologi-
cal, and histological aspects about the specimen that he is evaluating. Most hospitals
have integrated systems available to the pathologist, but in other scenarios, that may
not be available. In this case, the pathologist must be proactive in looking for those
data before evaluating and processing the specimen, as the optimal strategies in
choosing which areas to evaluate happen in that first time – subsequent new cuts can
be problematic as the tissue has already been cut before and its recomposition is
difficult and also add to the delay in delivering the final pathological report.
Another extremely important factor in the evaluation of these specimens is the
adequate fixation which does not mean just placing the specimens in formalde-
hyde, but it implies making sure that it can reach all areas of the specimen.
Without an adequate fixation, morphological and biomarker evaluation will suf-
fer [58, 59]. In larger specimens, it is necessary to make sections prior to fixation,
so the maximum thickness should be of 10 mm or less. If the pathology labora-
tory is not integrated with the operating room or if the pathologist is not present
at the surgery to make an evaluation and make the sections, the surgeon can do it
himself.
With all specimen-related information and an adequate fixation, the pathologist
can make optimal sections to provide the best anatomopathological report.
Low-grade DCIS specimens normally do not present any macroscopic lesions,
so small specimens are entirely included for analysis and larger specimens can have
only selected areas included, which should be chosen based on the lesion radiologic
characteristics.
58 M. A. Giannotti and F. N. Aguiar
Fig. 5.9 Breast and lymph node with histological features of post-neoadjuvant chemotherapy
regression
5 Pathological Aspects for Diagnosis 59
5.13.1 Immunohistochemistry
Immunohistochemistry is the most used technique, and usual antibodies are well
described in the carcinoma section. It is also used to help pathologists decide
between typical and atypical hyperplasias, to define invasion (Fig. 5.12), and to
subclassify carcinomas. Besides HER2 therapy, immune-checkpoint therapy is also
based on immunohistochemical assays. The table below shows some of the most
used antibodies (Table 5.6).
In situ hybridization allows the search of genetic alterations in slides from paraffin-
embedded biopsies. In breast pathology, the main utility is to verify HER2 equivo-
cal staining cases. The number of genes per cell and the ratio of genes and
Table 5.6 Most common antibodies used for breast tumor evaluation
Antibody Utility
P63, calponin, SMA, p40 Identify myoepithelial cells
CK5/6 ADH versus UDH
MUC1 Peripheral in micropapillary carcinoma
Cytokeratins Diagnosis of spindle cell metaplastic carcinoma
E-cadherin, B-catenin, Loss of expression in ILC
GP120
GATA3, mammaglobin, Breast primary tumors
Sox10
Pax-8, Melan A, TTF1, Cdx2 Identification of metastasis to the breast
PD-L1 Identify cases that benefit from immuno-therapy (e.g.,
atezolizumab)
*ADH = Atypical Ductal Hyperplasia, UDH = Usual Ductal Hyperplasia, ILC = Invasive Lobular
Carcinoma
centromeres are calculated. Amplified cases are those with HER2/centromere ratio
higher than 2.0 and more than 4.0 signals per cell [66].
References
1. Ng CCY, Md Nasir ND, Loke BN, Tay TKY, Thike AA, Rajasegaran V, et al. Genetic differ-
ences between benign phyllodes tumors and fibroadenomas revealed through targeted next
generation sequencing. Mod Pathol. 2021;34:1320.
2. Lu Y, Chen Y, Zhu L, Cartwright P, Song E, Jacobs L, et al. Local recurrence of Benign,
Borderline, and malignant Phyllodes tumors of the breast: a systematic review and meta-anal-
ysis. Ann Surg Oncol. 2019;26(5):1263.
3. Raza S. Management of high-risk breast lesions: counterpoint—time for personalized surveil-
lance. Am J Roentgenol. 2020;216:1434.
4. Lakhani SR, Eliis IO, Schnitt SJ, Tan PH, van de Vijver MJ. WHO classification of tumors of
the breast, vol. 4. Lyon: IARC Press; 2012. p. 240.
5 Pathological Aspects for Diagnosis 63
5. van Dooijeweert C, van Diest PJ, Willems SM, Kuijpers CCHJ, Overbeek LIH, Deckers
IAG. Significant inter- and intra-laboratory variation in grading of ductal carcinoma in situ of
the breast: a nationwide study of 4901 patients in the Netherlands. Breast Cancer Res Treat
[Internet]. 2019 [cited 2020 Feb 1];174(2):479–88. Available at: http://www.ncbi.nlm.nih.gov/
pubmed/30539380
6. Alghamdi SA, Krishnamurthy K, Garces Narvaez SA, Algashaamy KJ, Aoun J, Reis IM,
et al. Low-grade ductal carcinoma in situ. Am J Clin Pathol [Internet]. 2020;153(3):360–7.
Available at: https://academic.oup.com/ajcp/article/153/3/360/5643824
7. van Bockstal M, Baldewijns M, Colpaert C, Dano H, Floris G, Galant C, et al. Dichotomous
histopathological assessment of ductal carcinoma in situ of the breast results in substantial
interobserver concordance. Histopathology [Internet]. 2018;73(6):923–32. Available at: http://
doi.wiley.com/10.1111/his.13741
8. Hannemann J, Velds A, Halfwerk JBG, Kreike B, Peterse JL, van de Vijver MJ. Classification
of ductal carcinoma in situ by gene expression profiling. Breast Cancer Res. 2006;8(5):1–20.
9. Shehata M, Grimm L, Ballantyne N, Lourenco A, Demello LR, Kilgore MR, et al. Ductal
carcinoma in situ: current concepts in biology, imaging, and treatment. J Breast Imag.
2019;1(3):166–76.
10. Dabbs DJ, Schnitt SJ, Geyer FC, Weigelt B, Baehner FL, Decker T, et al. Lobular neo-
plasia of the breast revisited with emphasis on the role of E-Cadherin immunohistochem-
istry. Am J Surg Pathol [Internet]. 2013;37(7):e1–11. Available at: http://journals.lww.
com/00000478-201307000-00001
11. Wen HY, Brogi E. Lobular Carcinoma In Situ. Surg Pathol Clin [Internet]. 2018;11(1):123–45.
Available at: https://linkinghub.elsevier.com/retrieve/pii/S1875918117301435
12. Ginter PS, D’Alfonso TM. Current concepts in diagnosis, molecular features, and manage-
ment of lobular carcinoma in situ of the breast with a discussion of morphologic variants. Arch
Pathol Lab Med [Internet]. 2017;141(12):1668–78. Available at: http://meridian.allenpress.
com/aplm/article/141/12/1668/65743/Current-Concepts-in-Diagnosis-Molecular-Features
13. Shamir ER, Chen Y-Y, Chu T, Pekmezci M, Rabban JT, Krings G. Pleomorphic and florid lob-
ular carcinoma in situ variants of the breast. Am J Surg Pathol [Internet]. 2019;43(3):399–408.
Available at: http://journals.lww.com/00000478-201903000-00013
14. Chen Y-Y, Hwang E-SS, Roy R, DeVries S, Anderson J, Wa C, et al. Genetic and phenotypic char-
acteristics of pleomorphic lobular carcinoma in situ of the breast. Am J Surg Pathol [Internet].
2009;33(11):1683–94. Available at: http://journals.lww.com/00000478-200911000-00015
15. Foschini MP, Miglio R, Fiore R, Baldovini C, Castellano I, Callagy G, et al. Pre-
operative management of Pleomorphic and florid lobular carcinoma in situ of the breast:
report of a large multi-institutional series and review of the literature. Eur J Surg Oncol
[Internet]. 2019;45(12):2279–86. Available at: https://linkinghub.elsevier.com/retrieve/pii/
S0748798319305487
16. Allison KH, Hammond MEH, Dowsett M, McKernin SE, Carey LA, Fitzgibbons PL,
et al. Estrogen and progesterone receptor testing in breast cancer: American Society of
Clinical Oncology/College of American Pathologists Guideline Update. Arch Pathol Lab
Med [Internet]. 2020;144(5):545–63. Available at: http://meridian.allenpress.com/aplm/
article/144/5/545/427509/Estrogen-and-Progesterone-Receptor-Testing-in
17. Tamimi RM, Baer HJ, Marotti J, Galan M, Galaburda L, Fu Y, et al. Comparison of molec-
ular phenotypes of ductal carcinoma in situ and invasive breast cancer. Breast Cancer Res
[Internet]. 2008;10(4):R67. Available at: http://breast-cancerresearch.biomedcentral.com/
articles/10.1186/bcr2128
18. Mardekian SK, Bombonati A, Palazzo JP. Ductal carcinoma in situ of the breast: the impor-
tance of morphologic and molecular interactions. Human Pathol. 2016;49:114–23.
19. Miligy IM, Toss MS, Gorringe KL, Lee AHS, Ellis IO, Green AR, et al. The clinical and bio-
logical significance of HER2 overexpression in breast ductal carcinoma in situ: a large study
from a single institution. Br J Cancer [Internet]. 2019;120(11):1075–82. Available at: http://
www.nature.com/articles/s41416-019-0436-3
64 M. A. Giannotti and F. N. Aguiar
20. Sue GR, Lannin DR, Killelea B, Chagpar AB. Predictors of microinvasion and its prognos-
tic role in ductal carcinoma in situ. Am J Surg [Internet]. 2013;206(4):478–81. Available at:
https://linkinghub.elsevier.com/retrieve/pii/S0002961013002882
21. Badve S, A’hern RP, Ward AM, Millis RR, Pinder SE, Ellis IO, et al. Prediction of local recur-
rence of ductal carcinoma in situ of the breast using five histological classifications: a com-
parative study with long follow-up. Human Pathol [Internet]. 1998;29(9):915–23. Available at:
https://linkinghub.elsevier.com/retrieve/pii/S0046817798901964
22. Silverstein MJ, Lagios MD. Choosing treatment for patients with ductal carcinoma in situ: fine
tuning the University of Southern California/Van Nuys prognostic index. J Natl Cancer Inst -
Monographs. 2010;41:193–6.
23. Silverstein MJ. The University of Southern California/Van Nuys prognostic index for duc-
tal carcinoma in situ of the breast. Am J Surg [Internet]. 2003;186(4):337–43. Available at:
https://linkinghub.elsevier.com/retrieve/pii/S0002961003002654
24. Hilson JB, Schnitt SJ, Collins LC. Phenotypic alterations in ductal carcinoma in situ-associ-
ated myoepithelial cells. Am J Surg Pathol [Internet]. 2009;33(2):227–32. Available at: http://
journals.lww.com/00000478-200902000-00008
25. Aguiar FN, Cirqueira CS, Bacchi CE, Carvalho FM. Morphologic, molecular and microen-
vironment factors associated with stromal invasion in breast ductal carcinoma in situ: role of
myoepithelial cells. Breast Dis. 2015;35(4):249–52.
26. Livasy CA, Perou CM, Karaca G, Cowan DW, Maia D, Jackson S, et al. Identification of a basal-
like subtype of breast ductal carcinoma in situ. Human Pathol [Internet]. 2007;38(2):197–204.
Available at: https://linkinghub.elsevier.com/retrieve/pii/S004681770600534X
27. Meijnen P, Peterse JL, Antonini N, Rutgers EJT, van de Vijver MJ. Immunohistochemical cate-
gorisation of ductal carcinoma in situ of the breast. Br J Cancer [Internet]. 2008;98(1):137–42.
Available at: http://www.nature.com/articles/6604112
28. Doebar SC, van den Broek EC, Koppert LB, Jager A, Baaijens MHA, Obdeijn IMAM, et al.
Extent of ductal carcinoma in situ according to breast cancer subtypes: a population-based
cohort study. Breast Cancer Res Treat. 2016;158(1):179–87.
29. Chou S-HS, Gombos EC, Chikarmane SA, Giess CS, Jayender J. Computer-aided hetero-
geneity analysis in breast MR imaging assessment of ductal carcinoma in situ: correlating
histologic grade and receptor status. J Magn Resonan Imag [Internet]. 2017;46(6):1748–59.
Available at: http://doi.wiley.com/10.1002/jmri.25712
30. Esserman LJ, Kumar AS, Herrera AF, Leung J, Au A, Chen Y-Y, et al. Magnetic reso-
nance imaging captures the biology of ductal carcinoma in situ. J Clin Oncol [Internet].
2006;24(28):4603–10. Available at: http://ascopubs.org/doi/10.1200/JCO.2005.04.5518
31. Porter D, Lahti-Domenici J, Keshaviah A, Bae YK, Argani P, Marks J, et al. Molecular markers
in ductal carcinoma in situ of the breast. Mol Cancer Res. 2003;1:362.
32. Hernandez L, Wilkerson PM, Lambros MB, Campion-Flora A, Rodrigues DN, Gauthier A,
et al. Genomic and mutational profiling of ductal carcinomas in situ and matched adjacent
invasive breast cancers reveals intra-tumour genetic heterogeneity and clonal selection. J
Pathol [Internet]. 2012;227(1):42–52. Available at: http://doi.wiley.com/10.1002/path.3990
33. Aguiar FN, Mendes HN, Bacchi CE, Carvalho FM. Comparison of nuclear grade and immuno-
histochemical features in situ and invasive components of ductal carcinoma of breast. Revista
Brasileira de Ginecologia e Obstetrícia [Internet]. 2013;35(3):97–102. Available at: http://
www.scielo.br/scielo.php?script=sci_arttext&pid=S0100-72032013000300002&lng=en&nr
m=iso&tlng=en
34. Pareja F, Brown DN, Lee JY, da Cruz PA, Selenica P, Bi R, et al. Whole-exome sequencing
analysis of the progression from non–low-grade ductal carcinoma in situ to invasive ductal
carcinoma. Clinical Cancer Res [Internet]. 2020;26(14):3682–93. Available at: http://clincan-
cerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-19-2563
35. Aguiar F, Mendes H, Cirqueira C, Bacchi C, Carvalho F. Basal cytokeratin as a potential marker
of low risk of invasion in ductal carcinoma in situ. Clinics [Internet]. 2013;68(5):638–43.
Available at: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3654300/?report=classic
5 Pathological Aspects for Diagnosis 65
36. Vargas AC, Reed AEM, Waddell N, Lane A, Reid LE, Smart CE, et al. Gene expression
profiling of tumour epithelial and stromal compartments during breast cancer progression.
Breast Cancer Res Treat [Internet]. 2012;135(1):153–65. Available at: http://link.springer.
com/10.1007/s10549-012-2123-4
37. Sharma M, Beck AH, Webster JA, Espinosa I, Montgomery K, Varma S, et al. Analysis of
stromal signatures in the tumor microenvironment of ductal carcinoma in situ. Breast Cancer
Res Treat [Internet]. 2010;123(2):397–404. Available at: http://link.springer.com/10.1007/
s10549-009-0654-0
38. Muggerud AA, Hallett M, Johnsen H, Kleivi K, Zhou W, Tahmasebpoor S, et al.
Molecular diversity in ductal carcinoma in situ (DCIS) and early invasive breast cancer.
Mol Oncol [Internet]. 2010;4(4):357–68. Available at: http://doi.wiley.com/10.1016/j.
molonc.2010.06.007
39. Lee S, Stewart S, Nagtegaal I, Luo J, Wu Y, Colditz G, et al. Differentially expressed genes
regulating the progression of ductal carcinoma in situ to invasive breast cancer. Cancer
Res [Internet]. 2012;72(17):4574–86. Available at: http://cancerres.aacrjournals.org/cgi/
doi/10.1158/0008-5472.CAN-12-0636
40. Hendry S, Pang J-MB, Byrne DJ, Lakhani SR, Cummings MC, Campbell IG, et al. Relationship
of the breast ductal carcinoma in situ immune microenvironment with clinicopathological and
genetic features. Clin Cancer Res [Internet]. 2017;23(17):5210–7. Available at: http://clincan-
cerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-17-0743
41. Pruneri G, Lazzeroni M, Bagnardi V, Tiburzio GB, Rotmensz N, DeCensi A, et al. The preva-
lence and clinical relevance of tumor-infiltrating lymphocytes (TILs) in ductal carcinoma in
situ of the breast. Ann Oncol. 2017;28(2):321–8.
42. Man Y, Tai L, Barner R, Vang R, Saenger JS, Shekitka KM, et al. Cell clusters overlying
focally disrupted mammary myoepithelial cell layers and adjacent cells within the same duct
display different immunohistochemical and genetic features: implications for tumor progres-
sion and invasion. Breast Cancer Res [Internet]. 2003;5(6):R231. Available at: http://breast-
cancer-research.biomedcentral.com/articles/10.1186/bcr653
43. Gudjonsson T, Adriance MC, Sternlicht MD, Petersen OW, Bissell MJ. Myoepithelial
cells: their origin and function in breast morphogenesis and neoplasia. J Mammary Gland
Biol Neoplas [Internet]. 2005;10(3):261–72. Available at: http://link.springer.com/10.1007/
s10911-005-9586-4
44. Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, et al. Molecular char-
acterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004;6(1):17–32.
45. Hilson JB, Schnitt SJ, Collins LC. Phenotypic alterations in ductal carcinoma in situ-associ-
ated myoepithelial cells. Am J Surg Pathol. 2009;33(2):227–32.
46. Lokuhetty D, White VA, Watanabe R 1966-, Cree IA, WHO Classification of Tumours
Editorial Board, International Agency for Research on Cancer. Breast tumours.
47. Elston CW, Ellis IO. pathological prognostic factors in breast cancer. I. The value of histological
grade in breast cancer: experience from a large study with long-term follow-up. Histopathology
[Internet]. 1991;19(5):403–10. Available at: http://doi.wiley.com/10.1111/j.1365-2559.1991.
tb00229.x
48. Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular por-
traits of human breast tumours. Nature [Internet]. 2000;406(6797):747–52. Available at: http://
www.nature.com/articles/35021093
49. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns
of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad
Sci [Internet]. 2001;98(19):10869–74. Available at: http://www.pnas.org/cgi/doi/10.1073/
pnas.191367098
50. Cheang MCU, Chia SK, Voduc D, Gao D, Leung S, Snider J, et al. Ki67 index, HER2 sta-
tus, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst [Internet].
2009;101(10):736–50. Available at: https://academic.oup.com/jnci/article-lookup/
doi/10.1093/jnci/djp082
66 M. A. Giannotti and F. N. Aguiar
51. Nielsen TO. Immunohistochemical and clinical characterization of the basal-like subtype of
invasive breast carcinoma. Clinical Cancer Res [Internet]. 2004;10(16):5367–74. Available at:
http://clincancerres.aacrjournals.org/cgi/doi/10.1158/1078-0432.CCR-04-0220
52. Maisonneuve P, Disalvatore D, Rotmensz N, Curigliano G, Colleoni M, Dellapasqua S, et al.
Proposed new clinicopathological surrogate definitions of luminal A and luminal B (HER2-
negative) intrinsic breast cancer subtypes. Breast Cancer Res [Internet]. 2014;16(3):R65.
Available at: http://breast-cancer-research.biomedcentral.com/articles/10.1186/bcr3679
53. Lopez-Garcia MA, Geyer FC, Lacroix-Triki M, Marchió C, Reis-Filho JS. Breast can-
cer precursors revisited: molecular features and progression pathways. Histopathology.
2010;57(2):171–92.
54. Protocol for the Examination of Resection Specimens From Patients With Invasive Carcinoma
of the Breast [Internet]. 2020. Available at: www.cap.org/cancerprotocols.
55. Bossuyt V, Provenzano E, Symmans WF, Boughey JC, Coles C, Curigliano G, et al.
Recommendations for standardized pathological characterization of residual disease for
neoadjuvant clinical trials of breast cancer by the BIG-NABCG collaboration. Ann Oncol.
2015;26(7):1280–91.
56. Pinder SE, Rakha EA, Purdie CA, Bartlett JMS, Francis A, Stein RC, et al. Macroscopic
handling and reporting of breast cancer specimens pre- and post-neoadjuvant chemotherapy
treatment: review of pathological issues and suggested approaches. Histopathology. Blackwell
Publishing Ltd. 2015;67:279–93.
57. Marchiò C, Maletta F, Annaratone L, Sapino A. The perfect pathology report after neoadjuvant
therapy. J Natl Cancer Inst - Monographs. 2015;2015(51):47–50.
58. Khoury T. Delay to formalin fixation (cold ischemia time) effect on breast cancer molecules.
Am J Clin Pathol. Oxford University Press. 2018;149:275–92.
59. Carson FL. Formaldehyde as a fixative for light and electron microscopy. Micros Today.
2000;8(5):30–1.
60. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological com-
plete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analy-
sis. Lancet [Internet]. 2014;384(9938):164–72. Available at: https://linkinghub.elsevier.com/
retrieve/pii/S0140673613624228
61. Battisti NML, True V, Chaabouni N, Chopra N, Lee K, Shepherd S, et al. Pathological
complete response to neoadjuvant systemic therapy in 789 early and locally advanced
breast cancer patients: the Royal Marsden experience. Breast Cancer Res Treat [Internet].
2020;179(1):101–11. Available at: http://link.springer.com/10.1007/s10549-019-05444-0
62. Gianni L, Pienkowski T, Im Y-H, Roman L, Tseng L-M, Liu M-C, et al. Efficacy and safety of
neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or
early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase
2 trial. Lancet Oncol [Internet]. 2012;13(1):25–32. Available at: https://linkinghub.elsevier.
com/retrieve/pii/S1470204511703369
63. Santonja A, Sánchez-Muñoz A, Lluch A, Chica-Parrado MR, Albanell J, Chacón JI, et al.
Triple negative breast cancer subtypes and pathologic complete response rate to neoadjuvant
chemotherapy. Oncotarget [Internet]. 2018;9(41):26406–16. Available at: https://www.onco-
target.com/lookup/doi/10.18632/oncotarget.25413
64. Cottu P, D’Hondt V, Dureau S, Lerebours F, Desmoulins I, Heudel P-E, et al. Letrozole and
palbociclib versus chemotherapy as neoadjuvant therapy of high-risk luminal breast cancer.
Ann Oncol [Internet]. 2018;29(12):2334–40. Available at: https://linkinghub.elsevier.com/
retrieve/pii/S0923753419342358
65. Haque W, Verma V, Hatch S, Suzanne Klimberg V, Brian Butler E, Teh BS. Response rates
and pathologic complete response by breast cancer molecular subtype following neoadjuvant
chemotherapy. Breast Cancer Res Treat [Internet]. 2018;170(3):559–67. Available at: http://
link.springer.com/10.1007/s10549-018-4801-3
5 Pathological Aspects for Diagnosis 67
66. Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, et al. Human
epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical
Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J
Clin Oncol. 2018;36(20):2105.
67. Colomer R, Mondejar R, Romero-Laorden N, Alfranca A, Sanchez-Madrid F, Quintela-
Fandino M. When should we order a next generation sequencing test in a patient with cancer?
EClinicalMedicine. 2020;25:100487.
Part II
Update in Imaging Method
Chapter 6
Radiation Based Imaging: Digital
Mammography, Tomosynthesis
6.1 Introduction
Mammography is the most used imaging method for breast cancer screening in
average-risk women. In fact, it is still the mainstay of breast cancer screening
because it is broadly available, with established quality assurance, and has demon-
strated within multiple historic randomized controlled trials to reduce breast cancer
mortality in this population [1–4]. One of the main advantages of mammography
over other modalities is the ability to identify calcifications, which are the only ini-
tial manifestation in about 30–50% of non-palpable breast cancers, especially in
ductal carcinoma in situ (DCIS). However, mammography sensitivity for breast
cancer detection varies with breast density and is lower for women with heteroge-
neously dense or extremely dense fibroglandular tissue [5].
Radiation-based breast imaging has undergone an impressive evolution in the
past decades [6, 7]. The transition from conventional screen-film mammography to
full-field digital mammography (DM) allowed higher accuracy for women younger
than 50 years and women with heterogeneously dense or extremely dense breasts,
in addition to improvement in mammography workflow, lower radiation dose, and
faster localization procedures [8, 9]. More recently, digital breast tomosynthesis
(DBT), which is an evolution of DM, has improved both screening and diagnostic
A. G. V. Bitencourt (*)
A.C.Camargo Cancer Center, São Paulo, SP, Brazil
Diagnósticos da América SA, São Paulo, SP, Brazil
e-mail: [email protected]
C. Rossi Saccarelli
Diagnósticos da América SA, São Paulo, SP, Brazil
Hospital Sírio-Libânes, São Paulo, SP, Brazil
e-mail: [email protected]
6.2 Technique
a b
Fig. 6.1 Example of standard mammography views: (a) craniocaudal (CC); (b) mediolateral
oblique (MLO)
6 Radiation Based Imaging: Digital Mammography, Tomosynthesis 73
• True lateral view: it is used to determine the exact location of a lesion, to guide
stereotactic percutaneous procedures, and to evaluate calcifications of probable
intracystic nature; it can be performed in mediolateral (ML) or lateromedial
(LM) views; in both, the device will be positioned at 90° (it is usually positioned
at 45° on MLO view) (Fig. 6.2).
• Magnification view: it is mandatory for the evaluation of the morphology and
distribution of calcifications; the breast is positioned on a device which brings
the breast closer to the X-ray source and allows the acquisition of magnified
images (1.5–2x larger) of the region of interest (Fig. 6.3).
• Spot compression view: it is used on DM to perform a focal compression over
asymmetries and masses, which is used to separate overlapping structures and
improve lesion characterization (Fig. 6.4); this additional view is becoming less
frequently used after the advent of DBT.
a b
Fig. 6.5 Example of regular and rolled CC view on left mammography. Regular CC view (a)
showed an asymmetry in the outer quadrants (arrow), which was not characterized on rolled CC
view (b), corresponding to overlapped breast tissue
a b
Fig. 6.6 Example of regular and exaggerated CC view on left mammography. Regular CC view
(a) partially showed an asymmetry in the posterior third of the outer quadrants (arrow), which was
better characterized on exaggerated CC view (b)
a b
Fig. 6.7 Example of cleavage view on mammography. Regular CC view (a) showed an asymme-
try in the inner quadrant of the right breast (arrow). Cleavage view (b) confirmed the finding and
also showed a contralateral symmetric finding, compatible with sternalis muscle (anatomic variant)
6.3 Indications
a b
Fig. 6.10 Example of standard mammography views (a) and mammography views using Eklund
technique (b) in a patient with breast implants
between age 40 and 49, the indication for screening is controversial, because in this
population the incidence of cancer is lower and there is a higher frequency of dense
breasts, reducing the effectiveness of screening. However, several studies have
proven the benefit of mammographic screening also in this age group, especially
with DM and DBT. For women under the age of 40 years, without a high risk of
developing breast cancer, mammographic screening is not recommended due to the
low frequency of the tumor, lower sensitivity of the mammogram, and greater radio-
sensitivity of the parenchyma. For women over age of 70, the decision to suspend
screening should be individualized, considering global health, comorbidities, and
life expectancy.
Several studies have demonstrated the benefit of DBT over DM for breast cancer
screening, especially in women with dense breasts, with an increased rate of breast
cancer detection, an increase in the positive predictive value of patients summoned
for reassessment, and a reduction in the recall rates [19–24]. DBT also allows better
80 A. G. V. Bitencourt and C. Rossi Saccarelli
a b c d
Fig. 6.11 Example of architectural distortion in the upper outer quadrant of the left breast (circle),
better seen on tomosynthesis images ((b): MLO view; (d): CC view) than digital mammography
images ((a): MLO view; (c): CC view), which was confirmed as an invasive lobular carcinoma
6.4 Interpretation
Both DM and DBT reports should follow the American College of Radiology –
Breast Imaging Reporting and Data System (ACR-BI-RADS®) lexicon and must
include indication of the exam, breast composition, and comparison with previous
exams [26]. The report must also include an assessment category and manage-
ment recommendation based on the relevant findings described on the exam and
their probability of malignancy.
6.4.2 Masses
They are three-dimensional findings and must be described according to the shape
(oval, round or irregular), margins (circumscribed, obscured, indistinct, microlobu-
lated and spiculated), and density in relation to the normal fibroglandular tissue
(fat-containing, low, equal, or high density) (Figs. 6.13, 6.14, and 6.15).
a b c d
Fig. 6.12 Examples of different composition subtypes on mammography: (a) almost entirely fatty
breast; (b) breast with scattered areas of fibroglandular density; (c) heterogeneously dense breast;
(d) extremely dense breast
Shape
Fibroglandular
Tissue
Fig. 6.13 Illustration of mass shape and margins according to the BI-RADS fifth edition
82 A. G. V. Bitencourt and C. Rossi Saccarelli
a b c
d e f
Fig. 6.14 Examples of masses with different shapes and margins on mammography: (a) round
shape and circumscribed margins; (b) oval shape and circumscribed margins; (c) oval shape and
obscured margins; (d) irregular shape and microlobulated margins; (e) irregular shape and indis-
tinct margins;(f) irregular shape and spiculated margins
a b c d
Fig. 6.15 Examples of masses with different densities on mammography: (a) fat-containing oval
masses; (b) low-density oval mass; (c) equal-density (isodense) oval mass; (d) high-density irreg-
ular mass
6.4.3 Calcifications
Their morphology and distribution in the breast parenchyma must be taken into
account. According to their distribution (Fig. 6.16), they should be classified as:
• Diffuse, when they are present in the entire breast.
• Grouped, when at least five calcifications are observed in a space of less than
1 cm2, with the upper limit of 2 cm.
• Regional, when they occupy an area greater than 2 cm.
• Linear, suggesting ductal involvement.
• Segmental, inferring involvement of a ductal tree.
According to the morphology, calcifications should be classified as typically
benign (skin, vascular, coarse or popcorn-like, large rod-like, rim, dystrophic, milk
of calcium, and suture) or suspicious (amorphous, coarse heterogeneous, fine pleo-
morphic, fine linear, or fine linear branching) (Figs. 6.17 and 6.18). Round or
6 Radiation Based Imaging: Digital Mammography, Tomosynthesis 83
a b c d e
Fig. 6.16 Illustration of calcification distribution according to the BI-RADS fifth edition: (a)
grouped; (b) regional; (c) diffuse; (d) linear; (e) segmental
a b
c d
Fig. 6.17 Examples of typically benign breast calcifications on mammography: (a) skin lucent-
centered calcifications; (b) vascular calcifications in blood vessels; (c) coarse or popcorn-like cal-
cifications, typical of involuting fibroadenomas; (d) large rod-like intraductal calcifications; (e)
rim “eggshell” calcification; (f) dystrophic calcifications; (g) suture calcifications; (h) milk of cal-
cium sedimented calcifications in macro- or microcysts, usually more clearly defined on the true
lateral view
84 A. G. V. Bitencourt and C. Rossi Saccarelli
e f
g h
a b c d
Fig. 6.18 Examples of calcifications with suspicious morphology on mammography: (a) coarse
heterogeneous calcifications; (b) amorphous calcifications; (c) fine pleomorphic calcifications; (d)
fine linear and fine linear branching calcifications
punctate calcifications are classified based on their distribution into benign (scat-
tered or diffuse distribution), probably benign (grouped), or suspicious (linear or
segmental distribution) (Fig. 6.19).
6 Radiation Based Imaging: Digital Mammography, Tomosynthesis 85
a b
c d
Fig. 6.19 Examples of round and punctate (<0.5 mm) calcifications with different distributions on
mammography: (a) scattered isolated round calcifications (typically benign); (b) diffuse small
punctate calcifications (typically benign); (c) grouped round calcifications (probably benign); (d)
segmental round calcifications (suspicious)
This finding may be related to previous surgeries and the presence or absence of
other associated findings such as nodules or microcalcifications (Fig. 6.20). If there
is no personal history of surgery, trauma, or radiation therapy, any architectural
distortion should be considered suspicious for malignancy. These findings are par-
ticularly best characterized on DBT (Fig. 6.11) [27].
6.4.5 Asymmetries
a b
Fig. 6.20 Examples of architectural distortions on mammography: (a) postoperative changes; (b)
invasive lobular carcinoma
• Asymmetry: Lesion seen in only one breast and in only one incidence of the
mammographic exam. About 80% of the asymmetries are images formed by the
overlapping of the breast tissue and complementary views such as magnification
and compression or even the repetition of the exam can be sufficient to define
whether the lesion is an image overlay or not (Fig. 6.21).
• Global asymmetry: They are seen in only one breast but in both views (caudal
skull and oblique mediolateral) and occupy an area larger than one quadrant,
being benign in most cases, except when there is an associated clinical symptom
(Fig. 6.22).
• Focal asymmetry: Lesion seen in only one breast but in both views, occupying an
area smaller than one quadrant. If there is no correspondent on ultrasound, they
are considered ACR-BI-RADS® 3 because they have a malignancy index below
1%, when they are not associated with other suspicious findings (Fig. 6.23).
• Developing asymmetry: It has the same concept as a focal asymmetry, but it is
more dense or larger when compared to the previous exam (Fig. 6.24). Therefore,
for its characterization, it is mandatory to have a previous exam for comparison,
and it has a malignancy index of 13 to 27% and should always be submitted to a
histological study (ACR-BI-RADS 4) [28].
They are regular nodules with a fatty central hilum, often appear close to the ves-
sels, and can present in any location of the breast, being more common in the upper
outer quadrant (Fig. 6.25) [29].
6 Radiation Based Imaging: Digital Mammography, Tomosynthesis 87
Fig. 6.23 Example of focal asymmetry in the upper outer quadrant of the right breast (arrows)
a b
Fig. 6.24 Example of developing asymmetry in the central portion of the right breast (arrow in a),
not seen on prior exam (b)
6 Radiation Based Imaging: Digital Mammography, Tomosynthesis 89
Skin breast lesions are usually associated to benign conditions; however, they may
mimic breast lesions at mammography (Fig. 6.26). Placement of a metallic marker
on skin lesions identified prior to mammography is essential to reduce additional
a b
Fig. 6.26 Example of cutaneous cyst mimicking a mass in the left breast. (a) CC view with no
marker. (b) CC view with metallic marker in the skin lesion
90 A. G. V. Bitencourt and C. Rossi Saccarelli
workup. On the other hand, breast cancer may present as a superficial lesion, and
thus, any new, increasing, or suspicious superficial finding, not related to known
skin lesions, should undergo additional imaging for further characterization [30].
It is a rare finding (Fig. 6.27), but it can be associated with DCIS. Recent studies
have suggested that the positive predictive value of this findings is low and ultra-
sound should be performed to better assess ductal content [31].
They are the result of the effect that a lesion can cause in the surrounding tissues, includ-
ing skin thickening and retraction, nipple retraction, trabecular thickening (edema), axil-
lary adenopathy, architectural distortion, and calcifications (Figs. 6.28 and 6.29).
a b
Fig. 6.27 Example of single dilated duct in the right breast. (a) MLO view of both breasts. (b)
Spot compression view of the right breast
6 Radiation Based Imaging: Digital Mammography, Tomosynthesis 91
Fig. 6.28 Example of a malignant tumor with associated findings in the left breast, characterized
by skin and trabecular thickening (edema) and axillary adenopathy
may be different in the true lateral view. Thus, it is important to know how to prop-
erly locate the lesions, both for correlation with other imaging methods and to indi-
cate an additional incidence in the correct topography, when necessary. In general,
lesions located in the inner quadrants tend to have an upper location in the true lat-
eral view, when compared to the MLO view, while lesions located in the outer
quadrants tend to have a lower location in the true lateral view.
Based on the positive predictive values of each finding, final assessment should be
categorized in:
• BI-RADS 1 (negative; normal exam).
• BI-RADS 2 (benign findings): it includes typically benign calcifications, round
or punctate calcifications with diffuse distribution, fat-containing masses or
masses with typically benign calcifications (e.g., popcorn-like, rim or milk of
calcium), architectural distortion related to prior surgery, asymmetry or global
asymmetry with no other findings, and intramammary lymph node.
• BI-RADS 3 (probably benign; <2% likelihood of malignancy): it includes round
or punctate calcifications with grouped or regional distribution, oval masses
with circumscribed or obscured margins, and focal asymmetry with no other
associated findings.
• BI-RADS 4 (suspicious; 2–95% likelihood of malignancy): calcifications
with suspicious morphology, linear or segmental distribution, masses with
irregular shape or non-circumscribed margins, architectural distortion not
related to prior surgery, developing asymmetry, and other asymmetries with
other suspicious findings. This category can be further subdivided into 4A,
92 A. G. V. Bitencourt and C. Rossi Saccarelli
a b
Fig. 6.29 Examples of nipple retraction on mammography: (a) postoperative changes; (b) inva-
sive ductal carcinoma
4B, and 4C, with 2–10%, 10–50%, and 50–95% likelihood of malignancy,
respectively.
• BI-RADS 5 (highly suggestive of malignancy; >95% likelihood of malignancy):
more than one suspicious (BI-RADS 4) feature associated to additional findings.
• BI-RADS 6 (known biopsy-proven malignancy).
• BI-RADS 0 (incomplete; needs additional imaging evaluation or prior exams for
comparison).
6 Radiation Based Imaging: Digital Mammography, Tomosynthesis 93
6.4.12 Recommendation
6.5 Conclusions
Radiation-based breast imaging is still the most used method for screening and
diagnostic workup in clinical practice. The advent of DM and DBT has improved
mammography quality and results. Mammography findings should be reported
according to the BI-RADS lexicon, which is widely available and universally
accepted.
References
1. Myers ER, Moorman P, Gierisch JM, et al. Benefits and harms of breast cancer screening.
JAMA. 2015;314:1615. https://doi.org/10.1001/jama.2015.13183.
2. Mainiero MB, Moy L, Baron P, et al. ACR appropriateness criteria ® breast cancer screen-
inG. J Am Coll Radiol. 2017;14:S383–90. https://doi.org/10.1016/j.jacr.2017.08.044.
94 A. G. V. Bitencourt and C. Rossi Saccarelli
3. Helvie MA, Bevers TB. Screening mammography for average-risk women: the controversy
and NCCN’s position. J Natl Compr Cancer Netw. 2018;16:1398–404. https://doi.org/10.6004/
jnccn.2018.7081.
4. Saccarelli CR, Bitencourt AGV, Morris EA. Is It the era for personalized screening? Radiol
Clin N Am. 2021;59:129–38. https://doi.org/10.1016/j.rcl.2020.09.003.
5. Vourtsis A, Berg WA. Breast density implications and supplemental screening. Eur Radiol.
2019;29:1762–77. https://doi.org/10.1007/s00330-018-5668-8.
6. Joe BN, Sickles EA. The evolution of breast imaging: past to present. Radiology.
2014;273:S23–44. https://doi.org/10.1148/radiol.14141233.
7. Garcia EM, Crowley J, Hagan C, Atkinson LL. Evolution of imaging in breast cancer. Clin
Obstet Gynecol. 2016;59:322–35. https://doi.org/10.1097/GRF.0000000000000193.
8. Lewin JM, D’Orsi CJ, Hendrick RE. Digital mammography. Radiol Clin N Am.
2004;42:871–84. https://doi.org/10.1016/j.rcl.2004.06.004.
9. Bick U, Diekmann F. Digital mammography: what do we and what don’t we know? Eur
Radiol. 2007;17:1931–42. https://doi.org/10.1007/s00330-007-0586-1.
10. Kopans DB. Digital breast tomosynthesis from concept to clinical care. Am J Roentgenol.
2014;202:299–308. https://doi.org/10.2214/AJR.13.11520.
11. Nguyen T, Levy G, Poncelet E, et al. Overview of digital breast tomosynthesis: clinical cases,
benefits and disadvantages. Diagn Interv Imaging. 2015;96:843–59. https://doi.org/10.1016/j.
diii.2015.03.003.
12. Abdullah P, Alabousi M, Ramadan S, et al. Synthetic 2D mammography versus standard 2D
digital mammography: a diagnostic test accuracy systematic review and meta-analysis. Am J
Roentgenol AJR. 2020;20:24204. https://doi.org/10.2214/AJR.20.24204.
13. Oeffinger KC, Fontham ETH, Etzioni R, et al. Breast cancer screening for women at average
risk. JAMA. 2015;314:1599. https://doi.org/10.1001/jama.2015.12783.
14. Siu AL. Screening for breast cancer: U.S. Preventive Services Task Force recommendation
statement. Ann Intern Med. 2016;164:279. https://doi.org/10.7326/M15-2886.
15. Monticciolo DL, Newell MS, Hendrick RE, et al. Breast cancer screening for average-risk
women: recommendations from the ACR commission on breast imaging. J Am Coll Radiol.
2017;14:1137–43. https://doi.org/10.1016/j.jacr.2017.06.001.
16. Sardanelli F, Aase HS, Álvarez M, et al. Position paper on screening for breast cancer by
the European Society of Breast Imaging (EUSOBI) and 30 national breast radiology bod-
ies from Austria, Belgium, Bosnia and Herzegovina, Bulgaria, Croatia, Czech Republic,
Denmark, Estonia, Finland, France, G. Eur Radiol. 2017;27:2737–43. https://doi.org/10.1007/
s00330-016-4612-z.
17. Qaseem A, Lin JS, Mustafa RA, et al. Screening for breast cancer in average-risk women: a
guidance statement from the American College of Physicians. Ann Intern Med. 2019;170:547.
https://doi.org/10.7326/M18-2147.
18. Cardoso F, Kyriakides S, Ohno S, et al. Early breast cancer: ESMO Clinical Practice
Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 201930(8):1194–220. https://
doi.org/10.1093/annonc/mdz173.
19. Sharpe RE, Venkataraman S, Phillips J, et al. Increased cancer detection rate and variations
in the recall rate resulting from implementation of 3D digital breast tomosynthesis into a
population-based screening program. Radiology. 2016;278:698–706. https://doi.org/10.1148/
radiol.2015142036.
20. Rose SL, Shisler JL. Tomosynthesis impact on breast cancer screening in patients younger than
50 years old. Am J Roentgenol. 2018;210:1401–4. https://doi.org/10.2214/AJR.17.18839.
21. Alsheik NH, Dabbous F, Pohlman SK, et al. Comparison of resource utilization and clini-
cal outcomes following screening with digital breast tomosynthesis versus digital mammog-
raphy: findings from a learning health system. Acad Radiol. 2019;26:597–605. https://doi.
org/10.1016/j.acra.2018.05.026.
6 Radiation Based Imaging: Digital Mammography, Tomosynthesis 95
22. Conant EF, Barlow WE, Herschorn SD, et al. Association of digital breast tomosynthesis vs
digital mammography with cancer detection and recall rates by age and breast density. JAMA
Oncol. 2019;5:635.
23. Dang PA, Wang A, Senapati GM, et al. Comparing tumor characteristics and rates of breast
cancers detected by screening digital breast tomosynthesis and full-field digital mammogra-
phy. Am J Roentgenol. 2020;214:701–6. https://doi.org/10.2214/AJR.18.21060.
24. Bahl M, Mercaldo S, Dang PA, et al. Breast cancer screening with digital breast tomosyn-
thesis: are initial benefits sustained? Radiology. 2020;295:529–539 https://doi.org/10.1148/
radiol.2020191030.
25. Jackson VP. Diagnostic mammography. Radiol Clin N Am. 2004;42:853–70. https://doi.
org/10.1016/j.rcl.2004.06.002.
26. Sickles EA, D’Orsi CJ, Mendelson EB, Morris E, et al. ACR BI-RADS® mammography. In:
ACR BI-RADS® Atlas, breast imaging reporting and data system. 5th ed. Reston; 2013.
27. Durand MA, Wang S, Hooley RJ, et al. Tomosynthesis-detected architectural distortion: man-
agement algorithm with radiologic-pathologic correlation. Radiographics. 2016;36:311–21.
https://doi.org/10.1148/rg.2016150093.
28. Chesebro AL, Winkler NS, Birdwell RL, Giess CS. Developing asymmetries at mammography:
a multimodality approach to assessment and management. Radiographics. 2016;36:322–34.
https://doi.org/10.1148/rg.2016150123.
29. Bitencourt AG, Ferreira EV, Bastos DC, et al. Intramammary lymph nodes: normal and abnor-
mal multimodality imaging features. Br J Radiol. 2019;92:20190517. https://doi.org/10.1259/
bjr.20190517.
30. Giess CS, Raza S, Birdwell RL. Distinguishing breast skin lesions from superficial breast
parenchymal lesions: diagnostic criteria, imaging characteristics, and pitfalls. Radiographics.
2011;31:1959–72. https://doi.org/10.1148/rg.317115116.
31. Choudhery S, Simmons C, Woodard GA, et al. Outcomes of solitary dilated breast ducts
in symptomatic and asymptomatic patients. Br J Radiol. 2020;93:20191039. https://doi.
org/10.1259/bjr.20191039. https://doi.org/10.1001/jamaoncol.2018.7078
32. Rao AA, Feneis J, Lalonde C, Ojeda-Fournier H. A pictorial review of changes in the BI-RADS
fifth edition. Radiographics. 2016;36:623–39. https://doi.org/10.1148/rg.2016150178.
Chapter 7
Sonographic Based Imaging:
Ultrasound, Color Doppler, Elastography,
and Automated Breast Imaging
Abbreviations
7.1 Introduction
Ultrasound (US) is an interactive, dynamic, and real-time method that has become
an indispensable resource for breast assessment, both together and complementary
to mammography and magnetic resonance imaging (MRI). In the past, only mam-
mography has been useful for population-based screening. However, high-resolution
and quality-controlled ultrasound can further improve early cancer detection.
US has been used to classify benign, solid lesions with a negative predictive
value of 99.5% [1].
US advantages include:
(a) No exposure to radiation and its related risks
J. H. Catani (*)
Instituto de Radiologia INRAD, Hospital das Clinicas HCFMUSP, Faculdade de Medicina,
Universidade de Sao Paulo, São Paulo, SP, Brazil
e-mail: [email protected]
For positioning, the patient should lie down in the supine position with the chest
undressed and with arms relaxed and flexed behind the head to flatten the breast. It
might be necessary to roll the patient slightly to access the breast evenly.
The scan with the transducer should be done in at least two orthogonal planes,
including the peripheral regions of the breasts and lymphatic drainage pathways. As the
lactiferous ducts are arranged radially in the nipple, and the lesions tend to grow along
the ducts, the radial scan is favored by being anatomically guided (Figs. 7.1 and 7.2).
Medial structures should generally be scanned in the supine position, and lateral
structures including the armpit should generally be scanned with the patient in the
contralateral oblique position. This allows for the elimination of possible artifacts
secondary to inadequate compression of the breast tissue.
excellent spatial and soft tissue resolution, allowing for substantially improved dif-
ferentiation of subtle shades of gray, margin resolution, and conspicuousness of the
lesion at the bottom of normal breast tissue [2, 4–8].
The initial gain settings must be adjusted so that the fat at all levels is displayed
as medium level gray (calibrated by the superficial fat to the breast area).
The echogenicity of the structures is determined by comparison with the echo-
genicity of the fat. In comparison with breast fat, most solid masses are hypoechoic
and simple cysts anechoic, while skin, Cooper’s ligaments, and fibrous tissue are
echogenic.
100 J. H. Catani
Gentle pressure should be applied to the transducer during the examination. The
smaller the thickness of the tissue, the higher the image resolution. The increase in
pressure can have a beneficial effect on the acquired image, which can reduce arti-
factual shadows, as well as making it difficult to evaluate compressible structures
(ducts and vessels).
7.2.2 Anatomy
The region of interest in the breast comprises the portion from the skin surface (a)
and subcutaneous tissue (b) to the pleural surface/posterior chest wall (Fig. 7.3).
The areolopapillary complex is formed by the areola, papilla, and lactifer-
ous ducts.
The mammary zone (c) is formed by glandular and adipose tissue and Cooper’s
ligaments, where most of the breast ducts and lobules are located, and therefore the
main area of breast diseases.
The retromammary zone (d) is formed by retromammary fat, pectoral muscle,
ribs, and pleural surface.
The axillary region is the pyramidal space inferior to the glenohumeral joint, at
the junction between the arm and thorax, and contains many neurovascular struc-
tures, including the axillary artery, axillary vein, brachial plexus, and lymph nodes.
The anatomical repair used to classify lymph node levels in the axilla is the pec-
toralis minor muscle (highlighted in red in Fig. 7.4), being:
• Level I: lateral to the pectoralis minor muscle
• Level II: between the medial and lateral borders of the pectoralis minor muscle
• Level III: medial to the pectoralis minor muscle
d
7 Sonographic Based Imaging: Ultrasound, Color Doppler, Elastography, and… 101
7.2.3 Harmonic
The harmonic image can also be applied to better characterize a cyst or a subtle
solid mass. The generation of harmonic images allows the higher harmonic waves
to be selected and used to create the grayscale images with fewer artifacts [9].
Low-frequency surface reverberation echoes are thus reduced, allowing better
characterization of simple cysts (particularly if small) by eliminating artificial inter-
nal echoes often seen in fluids (Fig. 7.5).
Fig. 7.5 Cyst with harmonic resource (left) and without harmonic resource (right), showing atten-
uation of internal echoes and confirming that it is a simple cyst
102 J. H. Catani
Harmonic imaging also improves lateral resolution and can improve the contrast
between adipose tissue and subtle lesions, allowing for better definition of lesion
margins and posterior shading.
The evaluation through color mapping on the breast is based on the argument that
malignant or inflammatory lesions can cause angiogenesis and these vessels are
identified on the periphery or inside the lesions.
Currently, both power and color Doppler are complementary tools to the gray-
scale image, although power Doppler mode is favored for being more sensitive
when viewing small vessels, and factors such as flow direction and spectral evalua-
tion are not relevant.
Fig. 7.10 New, solid, hypoechoic, oval, and circumscribed mass, with peripheral and central vas-
cularization, and branched vessels on color Doppler. Patient underwent a percutaneous biopsy and
was diagnosed with adrenal carcinoma metastasis
7 Sonographic Based Imaging: Ultrasound, Color Doppler, Elastography, and… 105
7.4 Elastography
a b
c d
Fig. 7.11 (a) Score 1: complete deformability of lesion. (b) Score 2: most of the lesion is deform-
able although there are areas which are not deformable. (c) Score 3: presence of stiff area in center
with peripheral deformability of lesion. (d) Score 4: no deformability throughout the entire lesion
only. (e) Score 5: no deformation throughout the entire lesion or in adjacent tissue
7 Sonographic Based Imaging: Ultrasound, Color Doppler, Elastography, and… 107
Fig. 7.12 Solid, hypoechoic, irregular, and microlobulated lesion on the grayscale B-mode image
(right). Color mapping SE image (left) showing the same lesion shaded in blue color (hard lesion
in the color scale used)
With the use of light transducer pressure, automatic transient pulses can be gen-
erated by the US probe, inducing shear waves transversely oriented in the tissue.
The system captures the speed of these shear waves, which move faster in hard tis-
sue compared to soft tissue.
To measure the elasticity quantitatively in SWE for breast lesions, the most com-
mon practice is to place a 2- to 3-mm circular ROI over the stiffest part of the lesion,
including the immediately adjacent stiff tissue or halo [14].
Shear wave elastography provides quantitative information because tissue elas-
ticity can be measured in meters per second or kilopascals, a unit of pressure. A
value of over 80 kPa or velocity results of over 2 m/s are considered suspicious
(Figs. 7.13 and 7.14).
Fig. 7.13 Solid, hypoechoic, irregular, and microlobulated lesion on the grayscale B-mode image
(right), poorly deformable in color-mode SWE (left)
Fig. 7.14 Velocities measured in the lesions are very high (>3 m/s), which suggests a malig-
nant lesion
7 Sonographic Based Imaging: Ultrasound, Color Doppler, Elastography, and… 109
Fig. 7.15 When performing the correlation with mammography and MRI, take into account loca-
tion (superficial, medium, and deep thirds), dimensions, shape, and margin
Fig. 7.16 Oily cysts in the posterior third of the right breast seen on mammography and their cor-
respondence in the ultrasound study, next to the pectoral muscle
7 Sonographic Based Imaging: Ultrasound, Color Doppler, Elastography, and… 111
According to ACR BI-RADS®, the documentation must contain the breast that is
being studied, the laterality, and the place of the “body mark,” thus reducing any
errors in laterality and quadrant (Fig. 7.18).
When documenting findings, the adjustment of focus and gain and frequency
must be made and must contain images with and without measures to allow the
evaluation of margins in still images. The location should be noted in laterality,
quadrant, and/or clock face notation on the breast [9].
The size of the lesion should be measured in three dimensions, first reporting the
longest diameter and the rest in the orthogonal plane (Figs. 7.19 and 7.20). The
measurement can be made in millimeters or centimeters, rounding to just one deci-
mal place [7].
In the presence of multiple cysts, documentation of the largest cyst in each breast
and the measurement of its largest dimension can be made. In the presence of a
normal lymph node, the same documentation can be performed.
Measure the longest axis (1) and perpendicular to the first (2). The third measure
(3) should be taken from a plane orthogonal to the first image.
Fig. 7.17 Non-mass enhancement associated with architectural distortion in the posterior third of
the outer quadrants of the left breast in the mammary zone (surrounded by fibroglandular tissue)
and its ultrasound correspondence (“second-look” ultrasound)
112 J. H. Catani
Fig. 7.18 US
documentation with
laterality, quadrant, and
body mark
Fig. 7.19 Mass documentation in two plans without and with measures
The distance between the papilla and the lesion, and from this to the skin, is use-
ful information and easy to identify by any operator, facilitating eventual localiza-
tion during surgery and for evolutive control (Fig. 7.21).
A color Doppler/power Doppler image is recommended to assess the vascular-
ization of the documented lesion (Fig. 7.22).
The documentation on breast screening without changes should contain the four
quadrants and the retroareolar region.
7 Sonographic Based Imaging: Ultrasound, Color Doppler, Elastography, and… 113
a b
Fig. 7.22 Isoechoic mass that is difficult to characterize in B mode (a), but well delimited with the
use of power Doppler (b)
1. Breast composition
Can be homogeneous background – fat or fibroglandular or heterogeneous back-
ground echotexture (Figs. 7.25, 7.26, and 7.27).
2. The characteristics of a mass that must be evaluated and included in the report
include:
(a) Shape
Shape can be oval, round, or irregular (Figs. 7.28, 7.29, and 7.30).
(b) Margin
Can be circumscribed or non-circumscribed (indistinct, angular, microlobu-
lated, or spiculated) (Figs. 7.31, 7.32, 7.33, 7.34, and 7.35).
(c) Orientation (in relation to the skin surface)
Parallel or non-parallel (Figs. 7.36 and 7.37).
(d) Echogenicity
Can be anechoic, hyperechoic, hypoechoic, isoechoic, complex cystic solid,
or solid, heterogeneous (Figs. 7.38, 7.39, 7.40, 7.41, 7.42, and 7.43).
(e) Posterior features
116 J. H. Catani
a b
Fig. 7.49 A 75-year-old patient undergoing breast cancer screening. Linear calcifications on the
right papilla on mammography (a). US shows hyperechoic foci within the papilla (b). In color
Doppler (c), these foci have a “twinkle” artifact
5. Special cases
(a) Simple cyst (Fig. 7.55).
(b) Clustered microcysts (Fig. 7.56).
(c) Complicated cyst (Fig. 7.57).
(d) Mass in or on skin (Fig. 7.58).
(e) Foreign body, including implants (Fig. 7.59).
(f) Lymph nodes: intramammary, axillary (Fig. 7.60).
(g) Vascular abnormalities (Fig. 7.61).
(h) Postsurgical fluid collection (Fig. 7.62).
(i) Fat necrosis (Fig. 7.63).
126 J. H. Catani
The preparation of the ultrasound report must contain the indication of the exam, the
breast composition, a brief description of the lesion according to ACR BI-RADS,
comparison with previous exams, final impression, and recommendation.
The report should conclude a summary of relevant US findings with a final
assessment using BI-RADS® US categories 1–6 and the phrases associated
with them.
If report of a US examination is integrated with a concurrently mammographic
examination, the combined final assessment should reflect the highest likelihood of
malignancy assessed by the two exams [7].
7 Sonographic Based Imaging: Ultrasound, Color Doppler, Elastography, and… 127
Fig. 7.58 Skin lesion. Ultrasound demonstrates skin thickening with increased vascularity
Fig. 7.61 An 8-month-old with circumscribed, hypoechoic, and heterogeneous mass in the upper
outer quadrant of the left breast. Color Doppler shows high flow in the entire lesion with a vascular
pedicle, consistent with arteriovenous malformation
Fig. 7.63 Irregular mass in a patient with a history of surgical manipulation, requiring further
evaluation with mammography. The correlation with mammography showed that it was
steatonecrosis
References
1. Stavros AT, Thickman D, Rapp CL, Dennis MA, Parker SH, Sisney GA. Solid breast nod-
ules: use of sonography to distinguish between benign and malignant lesions. Radiology.
1995;196(1):123–34.
2. Watanabe T, Kaoku S, Yamaguchi T, Izumori A, Konno S, Okuno T, Tsunoda H, Ban K,
Hirokaga K, Sawada T, Ito T, Nakatani S, Yasuda H, Tsuruoka M, Ueno E, Tohno E, Umemoto
T, Shirakawa T. Multicenter prospective study of color Doppler ultrasound for breast masses:
utility of our color Doppler method. Ultrasound Med Biol. 2019;45(6):1367–79. https://doi.
org/10.1016/j.ultrasmedbio.2019.01.021. PMID: 30905536.
3. Cho N, Jang M, Lyou CY, Park JS, Choi HY, Moon WK. Distinguishing benign from malig-
nant masses at breast US: combined US elastography and color doppler US–influence on radi-
ologist accuracy. Radiology. 2012;262(1):80–90. https://doi.org/10.1148/radiol.11110886.
PMID: 22084209.
4. Hooley RJ, Scoutt LM, Philpotts LE. Breast ultrasonography: state of the art. Radiology.
2013;268(3):642–59. https://doi.org/10.1148/radiol.13121606. PMID: 23970509.
5. https://www.acr.org/-/media/ACR/Files/Practice-Parameters/US-Breast.pdf. Assessed 15
Dec 2020.
6. Monticciolo DL, Newell MS, Moy L, Niell B, Monsees B, Sickles EA. Breast cancer screen-
ing in women at higher-than-average risk: recommendations from the ACR. J Am Coll
Radiol. 2018;15(3 Pt A):408–14. https://doi.org/10.1016/j.jacr.2017.11.034. PubMed PMID:
29371086.
7. Mendelson EB, Bõhm-Vélez M, Berg WA, et al. ACR BI-RADS® ultrasound. In: ACR
BI-RADS® atlas, breast imaging reporting and data system. Reston: American College of
Radiology; 2013.
8. Leung JW, Sickles EA. Multiple bilateral masses detected on screening mammography:
assessment of need for recall imaging. AJR Am J Roentgenol. 2000;175(1):23–9. https://doi.
org/10.2214/ajr.175.1.1750023. PMID: 10882241.
9. Barr RG. Breast elastography: how to perform and integrate into a “best-practice” patient
treatment algorithm. J Ultrasound Med. 2020;39(1):7–17. https://doi.org/10.1002/jum.15137.
PMID: 31617225.
10. Vourtsis A. Three-dimensional automated breast ultrasound: technical aspects and first results.
Diagn Interv Imaging. 2019;100(10):579–92. https://doi.org/10.1016/j.diii.2019.03.012.
PMID: 30962169.
11. Hao SY, Jiang QC, Zhong WJ, Zhao XB, Yao JY, Li LJ, Luo BM, Ou B, Zhi H. Ultrasound
elastography combined with BI-RADS-US classification system: is it helpful for the diag-
nostic performance of conventional ultrasonography? Clin Breast Cancer. 2016;16(3):e33–41.
https://doi.org/10.1016/j.clbc.2015.10.003. PMID: 26639065.
12. Itoh A, Ueno E, Tohno E, Kamma H, Takahashi H, Shiina T, Yamakawa M, Matsumura
T. Breast disease: clinical application of US elastography for diagnosis. Radiology.
2006;239(2):341–50. https://doi.org/10.1148/radiol.2391041676. PMID: 16484352.
13. Vourtsis A, Kachulis A. The performance of 3D ABUS versus HHUS in the visualisation and
BI-RADS characterisation of breast lesions in a large cohort of 1,886 women. Eur Radiol.
2018;28(2):592–601. https://doi.org/10.1007/s00330-017-5011-9. PMID: 28828640.
14. Youk JH, Gweon HM, Son EJ. Shear-wave elastography in breast ultrasonography: the state
of the art. Ultrasonography. 2017;36(4):300–9. https://doi.org/10.14366/usg.17024. PMID:
28513127; PMCID: PMC5621798.
15. Kim SH. Image quality and artifacts in automated breast ultrasonography. Ultrasonography.
2019;38(1):83–91. https://doi.org/10.14366/usg.18016. PMID: 30139244; PMCID:
PMC6323315.
16. Kaplan SS. Automated whole breast ultrasound. Radiol Clin N Am. 2014;52(3):539–46.
https://doi.org/10.1016/j.rcl.2014.01.002. PMID: 24792655.
Chapter 8
Magnetic Resonance Imaging: Regular
Protocols and Fast Protocols
8.1 Introduction
Breast cancer diagnosis on magnetic resonance imaging (MRI) relies on the estab-
lishment of standard features for imaging acquisition. Patient positioning, temporal
and spatial resolution, and pulse sequences should be standardized to provide high-
quality images for adequate interpretation. The protocol should consist of sequences
that can be obtained from different equipment within the same institution. The
sequences obtained must provide images that are familiar to the radiology staff and
that satisfy certain imaging interpretation objectives, such as differentiating cystic
from solid lesions [1].
The imaging acquisition must not be extensively long. Long protocols can cause
discomfort to the patient and may result in interruptions during the examination or
patient motion that decreases the image quality. Long protocols reduce the number
of patients that can be scanned within a certain amount of time, which may result in
the elevation of costs. On the other hand, time should not be the most important fac-
tor in a protocol. If essential sequences are left out in a protocol, misdiagnosis may
occur [2].
A reliable protocol must provide images that maintain a high sensitivity for the
diagnosis of breast cancer and that are capable of differentiating between benign
and malignant lesions [3]. Lesion morphology on gadolinium contrast-enhanced
sequences is essential for this accomplishment. For malignant lesions, there are
several morphological features that must be demonstrated, including shape, margin,
internal enhancement pattern, and the presence of tumoral necrosis. Features that
are typical of benign lesions such as the presence of fat should be clearly evident. A
Table 8.1 Most common signal intensity of different structures in the breast on MRI sequences
Complicated Benign Malignant Lymph nodes
Sequence Fluid cysts Fat tumors tumors (cortex)
T2 with fat sat High Low to high Low Low to high Low High
T1 without fat Low Low to high High Low Low Low
sat
T1 with fat sat Low Low to high Low Low Low Low
T1 post Low Low to high Low Low to high High High
contrast
Subtraction Low Low Low Low to high High High
T2-weighted images with fat suppression are the most important sequence for the
detection of fluid [7]. Some facilities may opt to utilize a short tau inversion recov-
ery (STIR) sequence instead with the same objective. Simple cysts present typical
high signal intensity on T2 and have absence of internal enhancement on T1-weighted
contrast-enhanced images. Complicated cysts may present high protein or hemor-
rhagic fluid content that may reduce T2 and increase T1 signal intensity. Subtraction
images may be helpful in demonstrating the absence of internal enhancement within
a complicated cyst.
T2-weighted images with fat suppression may aid in the identification of a solid
portion on the wall of a cyst or a mass inside a fluid-filled duct. Cystic areas inside
a mass are also visible on T2. The presence of tumoral necrosis is associated with
poor prognosis and may also be identified as cystic areas with high signal intensity
inside a tumor. Likewise, the presence of perilesional edema on T2-weighted images
is also associated with malignancy and poor prognosis. Breast cancer on T2-weighted
images usually presents low signal due to its high cellularity. Enhancing circum-
scribed masses that are T2 bright are most frequently benign [8]. Some tumors, on
the other hand, may have a different biological profile and present high signal inten-
sity on T2, like mucinous carcinomas [9].
Sequences without fat suppression, either T1 or T2, may be used to evaluate fat-
containing lesions. Fat necrosis and mammary hamartoma, also known as fibroad-
enolipoma, can be easily detected as they present internal high signal similar to
adjacent fatty tissue [10]. Due to its high resolution, radiologists may use this
sequence to better visualize the margins of a mass, especially if it is surrounded by
fat. Similarly, fat surrounding axillary lymph nodes may facilitate their identifica-
tion and the diagnosis of nodal disease. Lastly, the distinction between a breast mass
and an intramammary lymph node is not always easy. The identification of a fatty
hilum in a sequence without fat suppression together with other morphological fea-
tures may indicate that it corresponds to an intramammary lymph node. Some breast
imaging services opt to include in their protocol only T2-weighted images with fat
134 J. V. Horvat and S. B. Thakur
suppression. As there is need for a sequence without fat suppression for the reasons
mentioned, a T1-weighted sequence without fat suppression may be used instead.
Post-processed images are extremely valuable in the analysis of breast MRI studies.
Subtraction images correspond to the deletion of signal of the unenhanced
T1-weighted sequence from a given contrast-enhanced phase. The result is the sub-
traction of all T1 signal except the signal from contrast enhancement. Subtraction
images are capable of demonstrating only structures that truly present enhancement
to gadolinium. There are, on the other hand, certain situations where the subtracted
signal may not be optimal. Patient motion between pre- and post-contrast sequences
may cause uneven subtraction. This may result in areas of false enhancement dem-
onstrated in the subtraction images. When patient motion is suspected, visual com-
parison between pre- and post-contrast T1-weighted sequences is necessary to
evaluate if it has occurred. Nowadays, there are several softwares that are capable of
automatically correcting patient motion.
Variations in signal intensity between dynamic contrast-enhanced phases can be
post-processed and graphically evaluated. Once a mass is visualized, it is possible
to position a region of interest over it and create an enhancement kinetic curve.
Three distinct enhancement curve patterns can be identified: persistent, plateau, and
washout. Most malignant masses present a rapid increase in signal intensity fol-
lowed by a contrast washout with a decay in signal intensity [12]. Most benign
lesions, on the other hand, present a slow and persistent increase in signal intensity
in the first 5 min after contrast injection [13].
3D MIP images have a significant contribution to the detection of breast cancer.
They are obtained from subtracted images that are post-processed to create a three-
dimensional volumetric display of both breasts. 3D MIP can better demonstrate
abnormal enhancement with the comparison between both sides. Not only it is used
to highlight malignancies, but it can also be useful in the comprehension of their
position and distribution within the breast and the proximity to the papilla, skin, and
chest wall.
limited spatial resolution and are prone to artifacts that may jeopardize imaging
interpretation [17]. Recent improvements in technique such as the use of readout-
segmented echo planar imaging or multi-shot multiplexed sensitivity- encoding
(MUSE) reduce these artifacts and improve lesion characterization [18, 19].
Post-processing images from the DWI sequence may be achieved with the cre-
ation of ADC maps that can be used to calculate the numerical amount of restriction
in a mass. High-cellularity lesions present high diffusion restriction that is repre-
sented as low signal on ADC maps [20]. By drawing a region of interest on a lesion,
ADC values can be calculated. Mean ADC values that are lower than 1.3 × 10−3 mm2/s
are associated with malignancy.
Breast implants have become widely present in our society. MRI is the best modal-
ity for the evaluation of implant ruptures and other implant-related abnormalities.
The vast number of different types of implants is specially challenging for radiolo-
gists. Implants may be filled with saline or silicone, which have different signal
intensities on MRI. They may have one, two or, more lumens. They may present
inner structures such as seals, valves, cannulas, markers, and chips that may cause
confusion during imaging interpretation. Nevertheless, a standardized protocol to
evaluate breast implants is necessary despite these variations [21].
Protocols to evaluate breast implants may be used alone or in addition to the
standard protocol. Patients who are having an examination for the sole reason of
evaluating implant rupture may opt not to receive intravenous contrast media. For
these patients, a combination of unenhanced sequences of the standard protocol and
sequences for implant evaluation should be used.
The sequences for implant evaluation consist of T2-weighted images with fat
suppression and silicone or water suppression. These two sequences should be done
in the same axis, as comparison between the two is of paramount importance. The
suppression of silicone and water makes it possible for radiologists to evaluate
abnormalities on the contours or within the implants that could be an indication of
intracapsular rupture. The signal shift of silicone on the sequences with and without
silicone suppression can demonstrate its presence within the breast parenchyma or
inside internal mammary or axillary lymph nodes. Additionally, peri-implant fluid
may be better depicted on the sequences with silicone suppression [22].
Because of the high sensitivity of MRI for the detection of breast cancer, several
studies have been conducted to evaluate its use in screening. There are a number of
obstacles in MRI screening, including equipment lower availability, need of
8 Magnetic Resonance Imaging: Regular Protocols and Fast Protocols 137
intravenous contrast media, higher costs, and longer examination and reading times.
To overcome some of these factors, abbreviated protocols with shorter examination
times and with a reduced number of sequences have been proposed [23]. The main
objective of these protocols is to maintain high sensitivity for breast cancer diagno-
sis [24]. Some protocols are proposed only to be used for patients with a previous
full imaging protocol study, while others are used on the patient’s first breast MRI
examination [25].
The majority of abbreviated protocols consists of dynamic contrast-enhanced
images with fat suppression. One pre-contrast and one post-contrast sequences are
acquired followed by post-processed images, including subtraction and 3D
MIP. Some authors have included other sequences such as more than one contrast-
enhanced phase or T2-weighted images that may improve lesion characterization
but increase examination and reading times. Since breast cancers usually present
rapid contrast enhancement, images obtained 90–120 s after the injection of gado-
linium depict the majority of malignancies on MRI.
On MRI post-contrast sequences, breast malignant tumors enhance early, fast and
avidly to gadolinium-based contrast agent. To evaluate the first stages of contrast
inflow of breast lesions, ultrafast sequences were developed and have been investi-
gated in several studies that were recently published. These sequences have high tem-
poral resolution with an imaging acquisition of the whole breast that takes less than
7 s. Multiple repeated sequences can be obtained after contrast media injection or after
it reaches the aorta. Kinetic curves of the first 1–2 min of contrast enhancement can
demonstrate how early and intense a lesion enhances, and measurements of the
enhancement curve slope can be obtained [26, 27]. These measurements have been
used to improve specificity in the differentiation between benign and malignant lesions
[28]. Some authors have proposed that ultrafast breast MRI can substitute the delayed
standard dynamic contrast-enhanced sequences and reduce examination time [17].
8.10 Conclusion
References
1. Newstead GM. MR imaging in the management of patients with breast cancer. Semin
Ultrasound CT MR. 2006;27(4):320–32.
2. Clauser P, Mann R, Athanasiou A, Prosch H, Pinker K, Dietzel M, et al. A survey by the
European Society of Breast Imaging on the utilisation of breast MRI in clinical practice. Eur
Radiol. 2018;28(5):1909–18.
3. Huang W, Fisher PR, Dulaimy K, Tudorica LA, O’Hea B, Button TM. Detection of breast
malignancy: diagnostic MR protocol for improved specificity. Radiology. 2004;232(2):585–91.
4. Thompson JL, Wright GP. The role of breast MRI in newly diagnosed breast cancer: an
evidence-based review. Am J Surg. 2020;221(3):525–8.
5. Yitta S, Joe BN, Wisner DJ, Price ER, Hylton NM. Recognizing artifacts and optimizing breast
MRI at 1.5 and 3 T. AJR Am J Roentgenol. 2013;200(6):W673–82.
6. Radiology ACo. Breast MRI clinical image review category D: spatial and temporal resolution
2019 [MRI and breast MRI accreditation]. Available at: https://accreditationsupport.acr.org/
support/solutions/articles/11000070275-breast-mri-clinical-image-review-category-d-spatial-
and-temporal-resolution
7. Santamaría G, Velasco M, Bargalló X, Caparrós X, Farrús B, Luis FP. Radiologic and
pathologic findings in breast tumors with high signal intensity on T2-weighted MR images.
Radiographics. 2010;30(2):533–48.
8. Lee JY, Jang M, Kim SM, Yun B, Jang JY, Ahn HS. Preoperative magnetic resonance imaging
characteristics of oval circumscribed fast enhancing lesions in patients with newly diagnosed
breast cancer. Medicine (Baltimore). 2018;97(19):e0704.
9. Pintican R, Duma M, Chiorean A, Fetica B, Badan M, Bura V, et al. Mucinous versus
medullary breast carcinoma: mammography, ultrasound, and MRI findings. Clin Radiol.
2020;75(7):483–96.
10. Taboada JL, Stephens TW, Krishnamurthy S, Brandt KR, Whitman GJ. The many faces of fat
necrosis in the breast. AJR Am J Roentgenol. 2009;192(3):815–25.
11. Carbonaro LA, Pediconi F, Verardi N, Trimboli RM, Calabrese M, Sardanelli F. Breast MRI
using a high-relaxivity contrast agent: an overview. AJR Am J Roentgenol. 2011;196(4):942–55.
12. Partridge SC, Stone KM, Strigel RM, DeMartini WB, Peacock S, Lehman CD. Breast DCE-
MRI: influence of postcontrast timing on automated lesion kinetics assessments and discrimi-
nation of benign and malignant lesions. Acad Radiol. 2014;21(9):1195–203.
13. Schnall MD, Blume J, Bluemke DA, DeAngelis GA, DeBruhl N, Harms S, et al. Diagnostic
architectural and dynamic features at breast MR imaging: multicenter study. Radiology.
2006;238(1):42–53.
14. Iacconi C, Thakur SB, Dershaw DD, Brooks J, Fry CW, Morris EA. Impact of fibroglandular
tissue and background parenchymal enhancement on diffusion weighted imaging of breast
lesions. Eur J Radiol. 2014;83(12):2137–43.
15. Durando M, Gennaro L, Cho GY, Giri DD, Gnanasigamani MM, Patil S, et al. Quantitative
apparent diffusion coefficient measurement obtained by 3.0Tesla MRI as a potential noninva-
sive marker of tumor aggressiveness in breast cancer. Eur J Radiol. 2016;85(9):1651–8.
16. Horvat JV, Durando M, Milans S, Patil S, Massler J, Gibbons G, et al. Apparent diffusion coef-
ficient mapping using diffusion-weighted MRI: impact of background parenchymal enhance-
ment, amount of fibroglandular tissue and menopausal status on breast cancer diagnosis. Eur
Radiol. 2018;28(6):2516–24.
17. Mann RM, Cho N, Moy L. Breast MRI: state of the art. Radiology. 2019;292(3):520–36.
18. Naranjo I, Gullo R, Morris E, Larowin T, Fung M, Guidon A, et al. High-spatial-resolution
multishot multiplexed sensitivity-encoding diffusion-weighted imaging for improved qual-
ity of breast images and differentiation of breast lesions: a feasibility study. Radiol Imaging
Cancer. 2020;2(3):e190076.
8 Magnetic Resonance Imaging: Regular Protocols and Fast Protocols 139
19. Bogner W, Pinker K, Zaric O, Baltzer P, Minarikova L, Porter D, et al. Bilateral diffusion-
weighted MR imaging of breast tumors with submillimeter resolution using readout-segmented
echo-planar imaging at 7 T. Radiology. 2015;274(1):74–84.
20. Horvat JV, Bernard-Davila B, Helbich TH, Zhang M, Morris EA, Thakur SB, et al. Diffusion-
weighted imaging (DWI) with apparent diffusion coefficient (ADC) mapping as a quantitative
imaging biomarker for prediction of immunohistochemical receptor status, proliferation rate,
and molecular subtypes of breast cancer. J Magn Reson Imaging. 2019;50(3):836–46.
21. Maijers MC, Niessen FB, Veldhuizen JF, Ritt MJ, Manoliu RA. MRI screening for silicone
breast implant rupture: accuracy, inter- and intraobserver variability using explantation results
as reference standard. Eur Radiol. 2014;24(6):1167–75.
22. Brenner RJ. Evaluation of breast silicone implants. Magn Reson Imaging Clin N Am.
2013;21(3):547–60.
23. Greenwood HI. Abbreviated protocol breast MRI: the past, present, and future. Clin Imaging.
2019;53:169–73.
24. Gao Y, Heller SL. Abbreviated and ultrafast breast MRI in clinical practice. Radiographics.
2020;40(6):1507–27.
25. Oldrini G, Derraz I, Salleron J, Marchal F, Henrot P. Impact of an abbreviated protocol for
breast MRI in diagnostic accuracy. Diagn Interv Radiol. 2018;24(1):12–6.
26. Mori N, Abe H, Mugikura S, Takasawa C, Sato S, Miyashita M, et al. Ultrafast dynamic
contrast-enhanced breast MRI: kinetic curve assessment using empirical mathematical model
validated with histological microvessel density. Acad Radiol. 2019;26(7):e141–e9.
27. Shin SU, Cho N, Kim SY, Lee SH, Chang JM, Moon WK. Time-to-enhancement at ultra-
fast breast DCE-MRI: potential imaging biomarker of tumour aggressiveness. Eur Radiol.
2020;30(7):4058–68.
28. Onishi N, Sadinski M, Gibbs P, Gallagher KM, Hughes MC, Ko ES, et al. Differentiation
between subcentimeter carcinomas and benign lesions using kinetic parameters derived from
ultrafast dynamic contrast-enhanced breast MRI. Eur Radiol. 2020;30(2):756–66.
Chapter 9
Nuclear Medicine Based Methods: PET
FDG and Other Tracers
9.1 Introduction
M. T. Sapienza (*)
Departamento de Radiologia e Oncologia, Hospital das Clinicas HCFMUSP, Faculdade de
Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
e-mail: [email protected]
P. F. Zampieri
Departamento de Medicina Nuclear, Instituto do Cancer do Estado de Sao Paulo ICESP,
São Paulo, SP, Brazil
e-mail: [email protected]
511keV
511keV
H2C OH
Hexokinase Glycolysis
H O H
GLUT
OH H 18
F-FDG 18
F-FDG-6P
OH OH
18
F
H
Fig. 9.2 FDG is transported by GLUTs and metabolized in FDG-6P by hexokinase but does not
follow the subsequent glycolysis steps, being accumulated in the cell
group has been replaced by fluorine-18, obtained in a cyclotron. The uptake of 18F-
FDG by tumor cells occurs through non-insulin-dependent glucose transport sys-
tems (GLUTs 1 and 4) and, like glucose, undergoes phosphorylation due to
hexokinase. Phosphorylated FDG does not progress in the metabolic pathway
beyond this step and remains trapped in the tumor cell (Fig. 9.2). Most breast can-
cers present increased metabolic activity, although there are variations according to
tumor characteristics and histology, to be discussed below.
9 Nuclear Medicine Based Methods: PET FDG and Other Tracers 143
MBq
Tissue activity concentration
mL × body weight g
SUV = ( )
Injected dose ( MBq )
Other tissues also show physiological uptake of FDG. Among the organs with
intense physiological uptake of 18F-FDG is the brain, impairing the ability to detect
metastases at this site. Activity in the kidneys, ureters, and bladder resulting from
the excretion of the radiopharmaceutical generally does not interfere with the inter-
pretation of the study. It should also be remembered that inflammatory changes with
infiltration by macrophages or granulation tissue (e.g., postoperative) have high
metabolic activity and may lead to a false-positive study. Other causes of false-
positive studies in the breast include benign conditions such as breast changes in
pregnancy and lactation (Fig. 9.3), gynecomastia, mastitis, fat necrosis (Fig. 9.4),
fibroadenoma, intraductal papilloma, and atypical ductal hyperplasia [1].
a b
Fig. 9.3 FDG PET scan of a 39-year-old breastfeeding patient demonstrating diffuse intense
activity in both breasts on MIP (a) and axial PET/CT (b), without tomographic lesions (c)
144 M. T. Sapienza and P. F. Zampieri
a b
Fig. 9.4 Restaging FDG PET/CT scan in a patient suspected of having recurrent breast carci-
noma. Axial fusion (a) and dedicated CT (b) images show areas of fat necrosis with increased
FDG uptake
a b c
d e
Fig. 9.5 Restaging with PET/CT in a patient with triple negative breast cancer presenting with
reduction of strength in the left hemibody. MIP (a), sagittal PET/CT fusion (b), and axial PET/CT
fusion (c–e) images show multiple metastasis with intense FDG uptake on the central nervous
system, lungs, liver, and bones
FDG PET/CT is not indicated for screening in early-stage cancers [8]. Although it
offers the opportunity to provide an overview of disease in a single procedure, the
recommendation against the use of PET scanning is supported by many factors [3,
8]: it has low sensitivity for the primary breast tumor, and neither FDG PET nor CT
is sensitive enough to detect breast cancers smaller than 1 cm [4]; the SUV in small
lesions is susceptible to the partial volume averaging effect, which may lead to a
lower value. This is further hampered by potential breathing motion artifact, as the
PET acquisition is performed at tidal volume breathing [8]; 18F-FDG imaging has
lower sensitivity than the sentinel node technique in assessing axillary lymph node
involvement, and the risk of distant metastases in early-stage cases is also low [3].
In addition to that, there is a high rate of false-positive scans in early-stage cancers,
leading to unwarranted patient anxiety and delay of care in those patients [3].
However, in high-risk patients like those with inflammatory (T4d) or locally
advanced breast cancer (LABC), the role of 18F-FDG imaging in detecting local and
distant metastasis has been highlighted [3]. FDG PET is helpful in local staging and
particularly helpful in evaluation of internal mammary nodes and distant nodal
metastasis [7]. It also helps to evaluate the equivocal findings on standard imaging
and, in some cases, can detect unknown sites of distant metastasis even though the
standard imaging is negative for lesions [7].
Any FDG-avid breast focus found during staging or surveillance of an extra-
mammary malignancy should be thoroughly investigated in patients with reason-
able life expectancy [8] (Fig. 9.6). These lesions have a 30–40% chance of being
a b
c d
Fig. 9.6 FDG PET/CT used to stage lung cancer. Lung CT demonstrates partially cavitated pul-
monary nodule (arrowhead in a), and PET/CT shows a solid nodule on the left breast (arrows in b,
c, and d) with FDG uptake, which was later diagnosed as ductal invasive carcinoma by biopsy
9 Nuclear Medicine Based Methods: PET FDG and Other Tracers 147
The most common sites of distant metastasis in breast cancer are bones, lungs, liver,
and brain, and the conventional imaging studies for detecting distant metastasis
include contrast-enhanced CT, bone scintigraphy, and MRI [10]. FDG PET/CT has
148 M. T. Sapienza and P. F. Zampieri
a b
Fig. 9.7 Restaging with PET/CT in a patient with breast cancer presenting rising levels of CA
15-3. MIP (a), axial PET/CT fusion (b), and axial CT (c) images show intense FDG uptake in
internal mammary lymph node (arrows in a, b, and c)
a b c
Fig. 9.8 Metastatic disease in a patient with biopsy-proven breast cancer. Arrows in sagittal PET
(a) and sagittal CT (b) demonstrate FDG uptake in lytic bone lesions, with no uptake in 18F-NaF
(sodium fluoride) (c)
that are normally performed months after initiating therapy. Rather, FDG metabolic
flare may be an indicator of future response to therapy [4].
PET is generally considered to be superior to CT and bone scintigraphy in detect-
ing lytic or mixed bone metastases and bone marrow metastases (Fig. 9.8), but a
multimodality approach is recommended for the investigation of bone metastases
due to the low sensitivity of PET in detecting sclerotic bone metastases in some
cases. In addition, sclerotic lesions without FDG accumulation can be detected on
CT images of a PET/CT study [13].
FDG PET is not as sensitive as MRI in the evaluation of brain metastases.
Cerebral cortex is highly FDG avid, and metastases often appear as focal areas of
hypometabolism, which may also be seen in non-neoplastic entities. Some lesions
do manifest as focal areas of hypermetabolism, although this can be difficult to
detect in the setting of normal physiologic gray matter metabolism [14]. Furthermore,
inflammatory tissue can also exhibit high FDG tracer uptake, diminishing diagnos-
tic specificity [15].
150 M. T. Sapienza and P. F. Zampieri
Question Why, despite the high global sensitivity for breast cancer metastases,
PET FDG is not indicated for the detection of brain lesions?
9.2.4 Recurrence
a b
Fig. 9.9 PET/CT in a patient with breast cancer presenting rising levels of tumor markers. MIP
(a), axial PET/CT fusion (b), and axial CT (c) images show FDG uptake on mediastinal soft tissue
mass (arrows in a, b, and c)
9 Nuclear Medicine Based Methods: PET FDG and Other Tracers 151
a b
Fig. 9.10 PET/CT in a patient suspected of having recurrent breast carcinoma due to back pain.
Sagittal PET/CT fusion (a) and sagittal CT (b) images show mixed bone lesion on the vertebral
body of L4, consistent with metastatic disease
a b
c d
Fig. 9.11 Response assessment in a patient with metastatic breast cancer after chemotherapy.
Pretreatment FDG PET/CT images (a and b) show intense bone lesion FDG uptake in the sternum.
(b) Follow-up FDG PET/CT images after chemotherapy (c and d) show significant decrease in the
intensity and extent of FDG uptake in the sternum
9 Nuclear Medicine Based Methods: PET FDG and Other Tracers 153
Question Is FDG PET a good method to assess whether a persistent residual mass
after therapy corresponds to a viable tumor or fibrosis?
FDG PET has been found to have also a role in predicting the prognosis of breast
cancer [23], indicating those patients more likely to progress, also with a correlation
between SUV measurement and histologic grade, proliferation index, and triple-
negative status [3]. High uptake reflects aggressive tumors and has poor prognosis
[7]. Although SUVmax is one of the most widely used parameters in clinical set-
tings, it does not show the uptake of the entire tumor mass and may not reflect the
intratumor heterogeneity sufficiently [23]. Therefore, there has been an increasing
interest in volumetric parameters such as metabolic tumor volume (MTV) and total
lesion glycolysis (TLG), which are becoming easier to measure due to commer-
cially available softwares [23]. The metabolic parameters SUVmax, MTV, and TLG
of the lesion before treatment are related to the recurrence rate. The higher the meta-
bolic parameters of the primary lesion, the greater the possibility of recurrence and
distant metastasis [24].
After the rapid incorporation of PET/CT into clinical practice for staging and prog-
nostic evaluation of breast cancer patients, hybrid PET/MRI equipment was devel-
oped. MRI provides not only high resolution but also high contrast images and the
possibility of better tissue characterization with the combination of different pulse
sequences. MRI sensitivity in the detection of breast lesions is well established,
especially for patients with limitations in conventional mammographic/ultrasound
evaluation, such as young women, dense breasts, and multifocal/multicentric
lesions. Incorporation of metabolic information by FDG PET can increase the spec-
ificity of the method, but care must be taken with false-negative results in cases of
small tumors (<1–2 cm) and histologic variations such as tubular carcinoma, well-
differentiated, or in situ ductal carcinomas.
PET/MR for breast imaging includes specific position and pulse sequences.
PET/MR mammography is acquired in the prone position with the breast hanging
and allows better identification of lesions in the breast and regional lymph nodes
[25]. MR imaging sequences usually include a T2 fat-suppressed sequence, a T1
non-fat-suppressed sequence, and post-contrast T1 sequences. Even though there is
no increase in radiation dose, the additional time spent on a PET/MRI exam should
154 M. T. Sapienza and P. F. Zampieri
While 18F-FDG remains the most widely used radiopharmaceutical in PET studies,
other tracers available or under development have a great prospect of clinical appli-
cation in breast cancer imaging. New radiotracers allow a noninvasive method not
only for staging but also for assessment of receptor status, metabolic activity, and
proliferation [35]. The clinical introduction of these radiopharmaceuticals depends
on factors such as local production and availability, clinical validation, and national
regulatory agencies approval.
18
F-NaF (sodium fluoride) presents high affinity for areas of bone remodeling,
determined by the fluoride ion exchange in hydroxyapatite crystals. Its higher and
faster uptake, together with the improved resolution of PET in relation to scintigra-
phy, allows a higher sensitivity than a bone scan, especially for osteoblastic metas-
tasis (Fig. 9.12). However, there is still a need for further cost-benefit analysis
before the recommendation to replace the methods [36].
18
FMISO and 18FAZA are hypoxia markers that have prognostic value. Hypoxia,
in addition to radioresistance, is associated with greater tumor aggressiveness and
worse response to treatment, and a boost in hypoxemic tumors may allow optimiza-
tion of radiotherapy results [37].
18
F-FLT (fluorothymidine) is a labeled nucleotide that traces DNA synthesis and
correlates with Ki-67 expression, used as an imaging proliferation marker. FLT PET
may be used in the assessment of early response to chemotherapy and endocrine
therapy [38].
18
F-FES (fluoroestradiol) and other receptor tracers
18
F-FES is currently used to evaluate the estrogen receptor status in breast cancer
patients, with the advantage of a simultaneous evaluation of multiple sites and of
sites not accessible to a biopsy. Visual and semiquantitative measures (SUV) in a
FES PET/CT can identify patients that will most likely benefit from endocrine
156 M. T. Sapienza and P. F. Zampieri
a b c d
Fig. 9.12 18F-NaF PET shows multiple areas of increased radioactivity (a, b, and d), predomi-
nantly in the axial skeleton, consistent with osseous metastases. CT image (c) shows both lytic and
sclerotic bone metastases, demonstrating fluoride uptake only on sclerotic lesions
therapy [39, 40]. Clinical trials are currently under way to assess the value of FES
PET/CT as predictive marker of response to endocrine therapy.
18
F-Fluoro Furanyl Norprogesterone (18F-FFNP) is a progesterone receptor
tracer, also under evaluation to determine its value as a predictor of response to
hormone therapy [41].
89
Zr-trastuzumab is one of many different tracers developed to study human epi-
dermal growth factor type 2 (HER2) receptor status. HER2 PET is a possible method
to predict response to trastuzumab-based therapy [41, 42].
Question FES PET can be used to evaluate hormone receptor status. Which lesions
should be presumed to respond to hormonal therapy, those with high or with low
radiopharmaceutical uptake?
9.5 Conclusion
18
F-FDG PET/CT is indicated for distant metastases detection in patients with
advanced breast cancer, particularly when other methods are inconclusive. It is also
indicated to detect suspected recurrence. New tracers and equipment may increase
the future role of PET imaging in breast cancer patients.
9 Nuclear Medicine Based Methods: PET FDG and Other Tracers 157
References
1. Dong A, Wang Y, Lu J, Zuo C. Spectrum of the breast lesions with increased 18F-FDG uptake
on PET/CT. Clin Nucl Med. 2016;41:543–57. https://doi.org/10.1097/rlu.0000000000001203.
2. Rashmi Kumar JB. NCCN Guidelines Version 6.2020 Breast Cancer. Version 6.2020 —
September 8, 2020. NCCN Guidelines Version 6.2020 Breast Cancer. Version 6.2020 —
September 8, 2020. [cited 2020 Sep 27]. Available at: https://www.nccn.org/professionals/
physician_gls/pdf/breast.pdf
3. Groheux D, Cochet A, Humbert O, Alberini J-L, Hindie E, Mankoff D. 18F-FDG PET/CT for
staging and restaging of breast cancer. J Nucl Med. 2016;57:17S–26S. https://doi.org/10.2967/
jnumed.115.157859.
4. Ulaner GA. PET/CT for patients with breast cancer: where is the clinical impact? Am J
Roentgenol. 2019;213:254–65. https://doi.org/10.2214/ajr.19.21177.
5. Hortobagyi GN, Connolly JL, D’Orsi CJ, Edge SB, Mittendorf EA, Rugo HS, et al. AJCC cancer
staging manual. Breast. 2017;2017:589–636. https://doi.org/10.1007/978-3-319-40618-3_48.
6. Provenzano E, Ulaner GA, Chin S-F. Molecular classification of breast cancer. PET Clin.
2018;13(3):325–38.
7. Lebron L, Greenspan D, Pandit-Taskar N. PET imaging of breast cancer. PET Clin.
2015;10:159–95. https://doi.org/10.1016/j.cpet.2014.12.004.
8. Benveniste AP, Marom EM, Benveniste MF, Mawlawi O, Fox PS, Yang W. Incidental primary
breast cancer detected on PET–CT. Breast Cancer Res Treat. 2015;151:261–8. https://doi.
org/10.1007/s10549-015-3402-7.
9. Yararbas U, Avci NC, Yeniay L, Argon AM. The value of 18F-FDG PET/CT imaging in breast
cancer staging. Bosn J Basic Med Sci. 2018;18(1):72–9.
10. Paydary K, Seraj SM, Zadeh MZ, Emamzadehfard S, Shamchi SP, Gholami S, et al. The
evolving role of FDG-PET/CT in the diagnosis, staging, and treatment of breast cancer. Mol
Imaging Biol. 2019;21:1–10. https://doi.org/10.1007/s11307-018-1181-3.
11. Park S, Yoon J-K, Lee SJ, Kang SY, Yim H, An Y-S. Prognostic utility of FDG PET/CT and bone
scintigraphy in breast cancer patients with bone-only metastasis. Medicine. 2017;96:e8985.
https://doi.org/10.1097/md.0000000000008985.
12. Peterson LM, O’Sullivan J, Wu Q, et al. Prospective study of serial 18F-FDG PET and
18F-fluoride PET to predict time to skeletal-related events, time to progression, and survival in
patients with bone-dominant metastatic breast cancer. J Nucl Med. 2018;59:1823–30. https://
doi.org/10.2967/jnumed.118.211102.
13. Aliyev A, Aksoy SY, Özhan M, Ekmekçi̇oğlu Ö, Vatankulu B, Kocael PÇ, et al. The role of
FDG PET/CT in detection of distant metastasis in theinitial staging of breast cancer. Turk J
Med Sci. 2016;46:349–60. https://doi.org/10.3906/sag-1409-1.
14. Fink KR, Fink JR. Imaging of brain metastases. Surg Neurol Int. 2013;4(Suppl 4):S209–19.
15. Galldiks N, Langen K-J, Albert NL, Chamberlain M, Soffietti R, Kim MM, et al. PET imag-
ing in patients with brain metastasis—report of the RANO/PET group. Neuro-Oncology.
2019;21:585–95. https://doi.org/10.1093/neuonc/noz003.
16. Göktaş İ, Cayvarlı H. The role of F-FDG PET/CT in evaluating elevated levels of tumor mark-
ers in breast cancer. Mol Imaging Radionucl Ther. 2018;27(1):3–9.
17. Piva R, Ticconi F, Ceriani V, Scalorbi F, Fiz F, Capitanio S, et al. Comparative diagnostic accu-
racy of 18F-FDG PET/CT for breast cancer recurrence. Breast Cancer. 2017;9:461–71. https://
doi.org/10.2147/bctt.s111098.
18. Liu Q, Wang C, Li P, Liu J, Huang G, Song S. The role of18F-FDG PET/CT and MRI in
assessing pathological complete response to neoadjuvant chemotherapy in patients with breast
cancer: a systematic review and Meta-analysis. Biomed Res Int. 2016;2016:1–10. https://doi.
org/10.1155/2016/3746232.
19. Tian F, Shen G, Deng Y, Diao W, Jia Z. The accuracy of 18F-FDG PET/CT in predicting
the pathological response to neoadjuvant chemotherapy in patients with breast cancer: a
158 M. T. Sapienza and P. F. Zampieri
V. C. Zanetta (*)
Department of Radiology, Instituto de Radiologia INRAD, Hospital das Clinicas HCFMUSP,
Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
e-mail: [email protected]
(MRI)-guided biopsies possible, increasing the range of lesions that are amenable to
percutaneous biopsy, while the development of more efficient sampling needles,
including larger core needles and vacuum-assisted devices, has improved diagnostic
accuracy.
Currently, the goal of the breast team is to obtain a definitive, nonoperative diag-
nosis of all potential breast abnormalities in a timely and cost-effective way [5]. The
standard approach to potential breast abnormalities is the “Triple Assessment”
derived from clinical evaluation, radiological information, and pathological assess-
ment. When there is agreement between these three assessments, the level of diag-
nostic accuracy exceeds 99%. For cases without clinical information, such as
nonpalpable breast lesions, similar levels of accuracy have been achieved by com-
bining only imaging tests and pathological evaluation [5].
Preferences for guidance methods and needle types for specific breast lesions vary
across institutions. A major factor in choosing the adequate imaging modality to
guide a biopsy procedure is lesion visibility. Breast lesions may be better depicted
at one modality over another, to the point that some highly suspicious lesions in one
modality may be completely invisible in another method.
For lesions that are well seen in all different modalities, the order of choice
among imaging methods will usually be ultrasound, mammography, and MRI. It is
easy to understand this order when comparing modalities. Ultrasound-guided biop-
sies have several advantages, including real-time sampling, lower cost, and the
absence of radiation or gadolinium injection, and are more comfortable for patients
when compared to MRI and mammographic guided procedures. These last two
methods require breast compression and immobile position in order to target the
lesion and complete the sampling, occasionally lasting more than 30 minutes.
Whenever a suspicious lesion is first seen at mammography or MRI, a targeted
ultrasound can be performed to look for a possible correlate, and if found, ultra-
sound is used to guide the procedure. Likewise, stereotactic and tomosynthesis-
guided biopsies must be preferred instead of MRI-guided procedures, given the
technical difficulties associated with MRI biopsy, higher costs, and limitations
related to contrast injection.
Usually masses, cysts, and every other lesion detected on ultrasound will be sub-
mitted to ultrasound-guided biopsy. Calcifications, breast asymmetries, and archi-
tectural distortions without a sonographic correlate will undergo
mammography-guided biopsy, and non-mass enhancements, foci, and masses with-
out correlation in other modalities will undergo MRI-guided biopsy. It is thus criti-
cal to perform an adequate imaging workup before any procedure to ensure accurate
targeting, safety of the procedure, and cost-effectiveness.
10 Image-Guided Percutaneous Biopsies 163
There are currently three different options of biopsy needles that are used for
sampling breast lesions: fine needle aspiration (FNA), conventional core needle
biopsy (CNB), and vacuum-assisted core needle biopsy (VAB). Although all imag-
ing modalities can guide procedures using any kind of needles, VAB is preferably
guided by mammography and MRI, while ultrasound is used for all types of biop-
sies and needles. This matter will be further discussed.
Prior to any percutaneous breast biopsy, there are several common steps that should
be undertaken regardless of the procedure. It is critical to ensure that complete
imaging workup has been performed, to obtain confirmation that the lesion is real
and requires tissue sampling, as well as to select the proper imaging guiding method.
For instance, the radiologist should review all available images and assure that the
lesion is not actually a benign finding with incomplete workup, such as skin calcifi-
cations without proper tangential mammographic views demonstrating them or fat
necrosis presenting as a suspicious complex mass on ultrasound without proper
mammographic correlation, that would reveal benign lucent-center calcification. If
there is any concern about the biopsy need due to incomplete workup or doubts
about what imaging guidance method is appropriate, additional studies may be per-
formed, such as additional mammographic views or a second-look ultrasound look-
ing for a sonographic correlate.
The patient should be oriented to keep normal activities and medications until the
day of the procedure; no fasting is required. Discontinuing blood thinning medica-
tions is a controversial topic. Blood thinning medications may increase the risk of
bleeding and hematoma formation due to biopsy; however, there are potential life-
threatening risks in stopping them, such as myocardial infarction and stroke. Current
literature supports that it is safe to perform percutaneous breast biopsy under medi-
cations known to impact bleeding, such as aspirin and anticoagulants [6]. Decisions
regarding discontinuing blood thinning medications should be made on a case-by-
case basis and shared with referring physician. If the radiologist opts for cessation,
it is necessary to ensure adequate timing for normalization of coagulation and, if
needed, check laboratory tests such as international normalized ratio (IRN) before
performing the procedure.
On the day of the procedure, the patient should be instructed to avoid applying
deodorant, lotion, or powder in the chest and axilla, since these may cause
164 V. C. Zanetta
The radiologist may choose to deploy a post-biopsy marker (clip) at the biopsy site
to facilitate the subsequent location. These markers are designed to be seen at dif-
ferent imaging modalities, allowing accurate localization of the biopsy site. Post-
biopsy markers are useful for several reasons:
1. Correlation of imaging findings between MRI, US, and mammography. This is
especially useful to check for accurate targeting when the lesion was first iden-
tified in one modality and biopsied using another one, for instance, a suspi-
cious MRI finding, submitted to a second-look ultrasound-guided biopsy. A
post-biopsy non-enhanced MRI T1-weighted sequence can be used to check
for the marker location and whether it matches the original diagnostic MRI
location.
2. Tissue markers allow for reliable re-identification of a biopsy site. Tissue mark-
ers are considered vital when the target is no longer visible after biopsy, for
instance, after complete removal during percutaneous biopsy and the histologic
results warrant surgical excision. In addition, re-identification of the biopsy
assists in evaluation for sampling error and imaging-pathology concordance.
3. Malignant lesions submitted to neoadjuvant chemotherapy may reduce to the
point of complete radiological response, becoming invisible in control examina-
tions. The post-biopsy marker is usually a reliable landmark to guide additional
surgical excision. For large lesions, more than one marker may be used for lesion
extent bracketing.
4. Dealing with breasts containing multiple sampled lesions is facilitated when
biopsy markers are used, clearly demonstrating which lesions have already been
sampled. Multiple biopsy sites may require different management, and biopsy
markers streamline this assessment. Post-biopsy markers are available in several
different shapes, and specific clips can be used to mark different targets
(Fig. 10.1).
Clips are frequently made of titanium or stainless steel. Other materials include
metal alloys and non-metallic alternatives, such as carbon-coated ceramic. Metallic
clips produce a susceptibility artifact on MRI, being easily identified, whereas non-
metallic clips may be less evident. On the other hand, metallic clips cause an artifact
on tomosynthesis that may obscure subtle findings.
Clips are available either alone, as bare clips, or associated with other materials,
such as bio-absorbable materials or non-absorbable polymers. Examples of associ-
ated materials include beta glucan, polyethylene glycol (PEG)-based hydrogel,
bovine collagen, polyvinyl alcohol (PVA) polymer, polyglycolic acid (PGA) micro-
fiber pad, PGA microfiber–PVA polymer combinations, starch pellets, polylactic
acid–PGA pellets, and suture-like netting [7]. These associated materials are
designed by vendors to improve or confer the clips certain features, such as improved
hemostasis, ultrasound visualization, or reduced clip migration.
166 V. C. Zanetta
a b
c d
Fig. 10.1 Malignant mass marked for neoadjuvant chemotherapy, with two different clips. (a, b)
sonography views of the markers before starting chemotherapy. One of the markers was deployed
at the center of the mass (a) and the other at the posterior and lateral edge (b), to mark lesion
extent. (c) Post-neoadjuvant chemotherapy mammography; the two markers are easily identified
and no residual lesion is seen. (d) MRI T1W axial image; notice the different susceptibility arti-
facts produced by each different clip
The clip must be deployed at the biopsy site and must remain close to it to mark
location accurately. Complications with clip placement are related to inaccurate
deployment away from the lesion, mechanical failure to deploy the clip (rare), and
clip migration, which may occur immediately after the procedure or later. Clip
migration is the most frequent cause of clip displacement, and there are several
reasons that can account for it:
1. The accordion effect: in procedures that require breast compression, such as
mammography or MRI-guided biopsy, the clip might not adhere to the biopsy
site and can be moved away from the biopsy site during compression release.
The displacement of the clip in those cases occurs in the direction of the needle
track (usually the Z axis, which will be further explained in the following
sections).
10 Image-Guided Percutaneous Biopsies 167
2. Clip migration in fatty tissue: predominantly fatty tissue breasts are more likely
to present clip migration within or outside the biopsy cavity, which is unrelated
to breast compression.
3. Bleeding – post-procedure continuous bleeding may displace a clip that is not
firmly attached to the biopsy cavity.
4. Hematoma – hematoma formation may prevent adequate clip attachment (float-
ing clip inside the biopsy cavity) or even cause significant mass effect displacing
the clip.
5. Changes in the biopsy site – Any factor that can modify the biopsy site may be
responsible for clip migration, such as air reabsorption in the biopsy cavity or
breast surgery.
Obtaining proper imaging documentation after clip deployment is essential. For
ultrasound-guided procedures, it is critical to seek real-time visualization of the
echogenic clip being deployed, while for MRI or mammography-guided proce-
dures, post-procedure imaging provides immediate clip identification.
Regardless of the method being used to guide the procedure, it may be useful to
obtain post procedure two orthogonal (craniocaudal (CC) and mediolateral (ML))
mammographic views to document clip location. Although uncommon, delayed
migration may occur, and immediate post-clip documentation is critical to assess
the extent of migration and accurate target location.
Ultrasound can guide fine-needle aspiration (FNA), CNB, and VAB. Currently,
US-guided CNB is the most popular modality of breast tissue sampling and consid-
ered very safe, diagnostically accurate, and cost-effective compared to other modal-
ities. In everyday practice, US-guided biopsy is the first option for sonographically
visible lesions. In the following sections, we will discuss the three main types of
US-guided procedures: FNA, CNB, and VAB.
All US-guided procedures can be performed either by a single operator or two
operators. A single operator uses the free-hand technique, in which one hand holds
the transducer in place while the “free hand” performs needle manipulation. Two
operators may be helpful for challenging cases such as lesions too close to a blood
vessel or in a very deep location, where one operator holds the transducer in place
while the other is responsible for needle handling and sampling.
during the procedure, even when no local anesthesia is used. Prior to the develop-
ment of core biopsy in the 1990s, FNA was the standard test used for breast lesion
assessment. However, FNA has several limitations compared to core biopsy and has
been progressively replaced by the latter. FNA retrieves small amount of tissue,
samples are frequently non-diagnostic or inadequate, and cytologic analysis is lim-
ited in differentiating invasive and in situ carcinoma, as well as tumoral subtype.
Currently, FNA is still employed in specific scenarios, especially for the evalua-
tion of metastasis in axillary lymph nodes, workup of breast cysts, and evaluation of
peri-implant effusions suspected of breast implant–associated anaplastic large cell
lymphoma (BIA-ALCL). Rarely, FNA might be preferred for very superficial or
deep lesions.
Axillary management is an evolving and complex subject, and it is beyond the scope
of this chapter to conduct a thorough discussion of axillary management and the
role of image-guided biopsy of lymph nodes (see Chap. 4).
Staging of axillary lymph nodes is an important prognostic factor, and sentinel
lymph node biopsy (SLNB) is the standard procedure to define axillary status.
SLNB could be skipped in cases of positive image-guided biopsy of axillary nodes,
leading straight to complete axillary dissection.
This paradigm has changed since the ACOSOG Z0011 study. The ACOSOG
Z0011 trial showed that for clinical T1–T2 N0 invasive breast cancer patients, with
up to two positive sentinel nodes in SNLB, there is no need for complete dissection
of axillary nodes for patients undergoing conservative surgery and whole breast
radiotherapy. In this setting, the role of preoperative axillary imaging has been ques-
tioned. Nevertheless, axillary image-guided biopsy with FNA or CNB still plays an
important role in assessing axillary status in the clinically node-positive axilla and
also in determining patients with extensive axillary disease burden, who may still
benefit from additional axillary management. In addition, assessment of nodal sta-
tus with ultrasound-guided biopsy should be considered for patients eligible for
neoadjuvant chemotherapy, as SLNB can be plagued by a high false-negative rate
afterward. Although with inferior diagnostic accuracy than CNB, FNA is still a
reasonable option to assess lymph nodes, being less traumatic and more affordable.
10.8.2 Cysts
FNA is indicated for symptomatic large simple cysts, without a true solid compo-
nent or other associated suspicious features (Fig. 10.2).
10 Image-Guided Percutaneous Biopsies 169
a b
c d
Usually, simple cysts are easily aspirated, leaving no residual lesions in the imag-
ing evaluation. For most of the cases, non-bloody and non-purulent aspirates are
deemed benign and can be discarded. However, purulent aspirates should be sent for
microbiologic analysis and culture, while bloody aspirates should be sent for cyto-
logic evaluation. In the last case, it is also important to place a post-biopsy marker,
especially if no residual lesion is visible.
Occasionally, cysts may be complicated and filled with echogenic material from
pus, clot, or debris. If ultrasound and Doppler evaluation cannot discern whether the
echogenic material is actually a solid component, FNA may be indicated to differ-
entiate them. True complicated cysts will likely disappear, while true solid compo-
nents will persist. It should be noted that if a true solid component is present, this
should be submitted for histologic analysis preferably with VAB. Nevertheless,
whenever US evaluation confirms a solid component within a cystic mass by flow
detection on Doppler, FNA should not preclude histologic analysis by VAB or
CNB [8].
170 V. C. Zanetta
10.9 Technique
The patient is positioned in a supine or supine oblique position, with the ipsilateral
arm raised behind the head. Usually, the oblique position is preferred for lateral
lesions, as well as axillary targets, especially in large breasts. In these situations, the
10 Image-Guided Percutaneous Biopsies 171
Skin entry
Skin entry
Fig. 10.3 Breast illustration for adequate needle entry point. (a) For superficial lesions, a closer
entry point from the lesion is desired, while for deep lesions (b), a farther entry point is required to
allow the needle to be parallel to the chest wall while sampling
oblique position may facilitate lesion visualization and improve needle path, allow-
ing it to pass parallel to the chest wall.
The skin is cleansed, the transducer is properly covered with a sterile cover, and
sterile gel is used to allow ultrasound imaging.
Planning the needle trajectory and skin entry site is one of the most crucial steps
in the procedure. In general, the needle is better visualized while perpendicular to
the transducer beam, and the needle tip should not be pointing to the chest wall to
avoid unwarranted trauma (Fig. 10.3). Given the breast natural curvature, superficial
lesions are better approached by a closer entry skin point in relation to the lesion,
while deep lesions require a farther entry point. These considerations are valid for
all US-guided procedures, including all types of biopsy needles, such as CNB and
VAB, although different adjustments are required according to the needle length.
Once proper skin entry point has been selected, proceed applying local anesthe-
sia, such as lidocaine, to decrease discomfort or pain during the procedure. The
sampling needle attached to a 10–20-ml disposable syringe should be gently intro-
duced and accurately placed within the lesion. The tip of the needle should be seen
inside the target. For solid lesions, sampling is performed while applying suction to
the syringe and using small and rapid back and forth stroke movements for cell
harvesting. Coupling the syringe with a pistol-grip mechanic syringe holder
(Fig. 10.4) is not required but highly recommended as it eases maintaining a nega-
tive pressure, allowing the operator to be more accurate during the back and forth
movements.
172 V. C. Zanetta
a b
Fig. 10.5 After removing the needle and drawing air into the syringe, the operator replaces the
needle onto the syringe and squirts one drop in the center of a slide (a). A second slide is used to
gently spread the sample and make the smear (b)
Whenever suitable material is collected, the negative pressure is relieved and the
needle withdrawn. Handling and preparation of the sampled cytologic material is as
important as targeting.
If an on-site cytotechnologist or pathologist is not available, the operator must be
familiar with pathology service standards and be able to prepare the direct smears
on slides (Fig. 10.5) or any other desired preparation, such as cell blocks. If a slide
is being submitted to fixation, it is imperative to be as expedite as possible, since
delayed fixation may impair cellular preservation.
It is recommended to perform multiple needle passes, aiming to sample different
portions of the target lesion to improve representativeness and diagnostic accuracy.
A cytotechnologist or pathologist present on-site during the procedure can assist
and ensure adequate sample collection.
FNA is minimally invasive, usually very well tolerated by patients, and considered
a safe breast procedure. Patients are released after the procedure and do not require
further care. Rarely, the procedure may be complicated with pain, bleeding, or
10 Image-Guided Percutaneous Biopsies 173
Core needle biopsies can be divided into two main groups: conventional core needle
biopsy (CNB) and vacuum-assisted biopsy (VAB), which, although both use hollow
needles to obtain the samples, are substantially different systems.
Conventional US-guided core needle biopsy (CNB) uses a hollow needle to
retrieve tissue samples and overpowered FNA with its higher diagnostic accuracy,
capability of differentiating in situ and invasive carcinomas, tumor grade, subtype,
and receptor status. The procedure is currently the most popular modality of breast
tissue sampling.
CNB uses a spring-loaded mechanism to obtain the samples with 18–12G nee-
dles. They are available as automatic or semiautomatic devices (Figs. 10.6 and 10.7).
The mechanism for cutting and collecting each sample is similar for both: the
needle contains an outer cutting cannula and an inner stylet, with a notch for speci-
men collection. With the inner stylet retracted, the needle tip is first placed adjacent
to the target border. The inner stylet is advanced inside the target. Once the inner
trocar is in place, the outer cannula is rapidly advanced, cutting and storing each
sample in the inner trocar notch. The whole needle is then withdrawn from the
breast, and the sample is collected after being exposed in the trocar notch (Figs. 10.8,
10.9, 10.10, and 10.11).
The main difference between automatic and semiautomatic devices is to the
advancement of the inner trocar. In semiautomatic devices, the inner trocar is manu-
ally advanced inside the lesion by the operator. Once the operator triggers (needle
firing), the outer cutting cannula is spring deployed (one-stage mechanism). In auto-
matic needles, the needle tip is positioned adjacent to the lesion border, and once the
operator triggers, the inner trocar is spring deployed inside the lesion, followed by
automatic spring deployment of the outer cutting cannula (two-stage mechanism).
Since the inner trocar of the most commonly used 14-gauge needle has a throw of
2.2 cm, before firing the needle, the operator has to ensure the presence of adequate
tissue beyond the lesion to avoid unwarranted trauma to the surroundings.
Because the procedure has to be repeated for each sample and requires multiple
passes, a coaxial device may be used to facilitate repeated access to the biopsy site.
The semiautomatic method allows for a steady and operator-controlled advance-
ment of the inner trocar, with real-time tracking of the needle tip. Thus, it is popular
in challenging cases such as sampling of subtle atypical axillary lymph nodes,
where it is possible to avoid the vascular hilum and better target the suspicious cor-
tex, lesions near vascular structures or next to breast implants, avoiding unwar-
ranted rupture of the implant shell. Nevertheless, the automatic procedure is slightly
174 V. C. Zanetta
Fig. 10.6 Examples of automatic core biopsy devices: (a) Max-Core™ Disposable Core Biopsy
(Bard Medical Division, Covington, GA, USA). (b) Magnum Reusable Core Biopsy Instrument
(Bard Medical Division, Covington, GA, USA). (c) BioPince™ Full Core Biopsy (Argon Medical
Devices, Inc., Athens, TX, USA). (d) Pro-Mag™ Ultra (Argon Medical Devices, Inc., Athens,
TX, USA)
a b
Fig. 10.9 (a) Semiautomatic needle device with the inner stylet retracted. (b) After pushing the
plunger (arrowhead), the inner stylet is advanced, exposing the sampling notch. (c) In detail, the
sampling notch (arrow) and outer cutting cannula (void arrow)
176 V. C. Zanetta
a b
c d
e f
Fig. 10.10 Core biopsy of a lymph node with a semiautomatic device. (a) Ultrasound shows the
abnormal cortical thickening (void arrow) and vascular hilum (arrow) of the target lymph node. (b)
The needle is introduced fully retracted until reaching the border of the cortex. (c) Gradual and
controlled advancement of the inner stylet, piercing the cortical and avoiding the vascular hilum.
(d) The inner stylet fully extended while the sampling notch placed in the region of interest. (e) 90°
rotation of the transducer confirming in both planes the accurate position of the sampling notch. (f)
Post-fire with advancement of the outer cutting cannula over the sampling notch, colleting
the sample
a b
Fig. 10.11 Core biopsy of irregular mass with an automatic device (a) Pre-fire – the needle is
introduced until the tip is positioned near the border of the mass. (b) Post-fire – both the inner stylet
and outer cutting cannula have been spring deployed
10 Image-Guided Percutaneous Biopsies 177
more comfortable for the operator and less likely to displace the lesion as the inner
trocar advances. Especially for hard lesions surrounded by breast soft tissue, the
vigorous mechanical inner trocar throw in automatic devices is more likely to pierce
the lesion instead of displacing it forward.
Vacuum-assisted breast biopsy (VAB) uses a vacuum-powered biopsy probe to
obtain samples from larger core needles, available in a variety of sizes, needle
gauges range from 14- to 7-gauge. The needle sampling chamber is positioned into
or underneath the lesion, and after activation, the system continuously opens the
needle sampling chamber while using suction to pull the tissue into it; a rotating
cutting device advances through this tissue, closing the sampling chamber and aspi-
rating the sampled material through the probe into a collection chamber. This pro-
cess is repeated several times. The operator holds the probe in place and may rotate
it to acquire samples from different locations, requiring only one needle pass for the
whole procedure (Figs. 10.12 and 10.13).
a b
c d
Fig. 10.12 (a) VAB needle with a sampling chamber positioned underneath the lesion. (b) Vacuum
pulls the lesion inside the sampling chamber. (c) Cutting cannula advances and shaves part of the
lesion. (d) The specimen is aspirated (e) followed by retraction of the cutting cannula, uncovering
the sampling chamber for the next sample
178 V. C. Zanetta
a b
c d
Fig. 10.13 VAB of an irregular mass. (a) A mass is situated near a breast implant. (b) The VAB
needle is introduced until the sampling chamber is located underneath the target, being careful not
to pierce the breast implant. Once in position, sampling is performed with (c) retraction of the cut-
ting cannula, lesion aspiration, and (d) advancement of the cutting cannula
Adequate sampling requires not only accurate targeting of the lesion but also repre-
sentative and satisfactory sampling; in other words, adequate sampling requires
samples of sufficient quality from the most suspicious parts of the lesion.
Adequate sampling is ultimately related to operator skills and needle selection.
While the operator is responsible for adequately targeting the lesion, needle selec-
tion impacts the amount of tissue retrieved per biopsy and the quality of each sample.
The amount of tissue retrieved per biopsy and its quality are dependent on needle
gauge as well as the method of choice: VAB vs CNB. Larger core needles yield
larger sample sizes, and for the same needle gauge, vacuum-assisted biopsy obtains
larger amounts of tissue when compared to CNB. For instance, the average speci-
men weight for the 14-gauge CNB sample is 17 mg, while per VAB, it is 37 mg. An
11-gauge VAB probe is capable of obtaining on average 94 mg per sampling, which
is over five times the amount of tissue retrieved by the 14-gauge CNB [12]. In addi-
tion, another key factor is the quality of the sampled material. VAB devices can
provide superior quality samples using suction, avoiding fragmented or floating
specimens that are worse for pathologic assessment. For instance, bleeding in the
biopsy cavity during the procedure may occur to varying degrees and can be severe
10 Image-Guided Percutaneous Biopsies 179
during a CNB, leading to the removal of a blood clot instead of an actual tissue. An
interesting feature available in most VAB devices is manual aspiration of the biopsy
cavity. Applying suction to the biopsy site can draw blood from the biopsy cavity,
improving actual tissue sampling instead of blood clots.
A skilled operator should sample different portions of the lesion and consider the
minimum amount of required tissue for a definitive diagnosis, which is dependent
on lesion type and size. For instance, when sampling a complex solid cystic mass,
the main objective should be to target the solid portion, and when sampling a very
heterogeneous lesion, the goal is to obtain fragments from different parts of the
lesion. Due to the risk of carcinoma in situ, non-mass lesions require greater sam-
pling for a definitive diagnosis. This is also true for irregular areas of distortion,
comprising a differential diagnosis of sclerosing adenosis, radial scar, and invasive
carcinoma [13].
There is no consensus on the number of fragments that must be retrieved during
a standard CNB. The American College of Radiology suggests that at least three to
six fragments should be obtained from each lesion. However, instead of looking for
a fixed number of fragments, the operator should seek for adequate quality of the
samples obtained according to the lesion type.
The two main advantages of VAB are the convenient single-pass procedure and
capability of acquiring high-quality samples. A study by the American Society of
Breast Surgeons Mastery of Breast Surgery Registry showed that larger core nee-
dles and tethered vacuum-assisted biopsy are associated with lower re-biopsy rates.
One potential drawback from VAB devices is its higher cost per procedure when
compared to standard CNB, although this might be balanced when comparing the
cost per diagnosis, since VAB is more accurate [13].
Although the benefits of performing VAB in stereotactic biopsy for calcified
lesions have been extensively shown in the literature, the evidence supporting VAB
instead of CNB for US-guided biopsies, mainly for solid masses, is less clear. For
biopsy of solid masses, both methods have shown excellent diagnostic performance,
and CNB is a reasonable option in most scenarios. The operator should take into
consideration its familiarity with each method, the type of the lesion, and cost of
each method. In addition, while CNB is restricted to diagnostic purpose, VAB has
been playing an increasing role as a therapeutic procedure, which will be further
discussed below.
180 V. C. Zanetta
VAB is also suitable for therapeutic removal of benign breast lesions, such as fibro-
adenomas. In such cases, the operator seeks the complete removal of a symptomatic
benign entity, thus avoiding excisional surgery. Moreover, VAB has an increasing
role in the management of benign lesions with heterogeneity in histological find-
ings, such as papillary lesions.
Papillary lesions are a broad category ranging from definitely benign papilloma
to lesions containing varying degrees of atypia, in situ and invasive carcinoma. One
of the problems dealing with papillary lesions is that atypical cells can be found
only in part of the lesion, as small foci, or even in adjacent areas outside the lesions
themselves. Thus, although most papillary lesions are benign, CNB tends to mislead
the pathologist and is associated with underestimation. For papillary lesions diag-
nosed through CNB, the risk of malignant upgrade on surgery has been reported as
high as 15.7% [14].
Although surgical excision is still appropriated to atypical papilloma and malig-
nant papillary lesions, most papillary lesions are in fact benign papilloma, which
frequently present as a small intraductal solid mass and bloody nipple discharge.
Surgical excision for papilloma without atypia diagnosed by VAB may not be nec-
essary, since pathologic underestimation of heterogeneous lesions is reduced in
biopsies with larger gauge needles, such as VAB. Moreover, VAB can be a therapeu-
tic option when complete removal of the lesion is achieved, resolving the nipple
discharge in symptomatic cases. An ongoing research has shown that if complete
removal of papilloma has been achieved by VAB and no atypia is found, the upgrade
rate to malignancy is close to 0% [15]. This approach to use VAB instead of surgery
for papilloma without atypia may also be suitable for papilloma first diagnosed by
standard CNB, where VAB could be used as a final diagnostic and therapeutic tool
(Fig. 10.14).
This trend to de-escalate treatment with complete removal of the lesion by VAB
has been expanding to other histological diagnosis with uncertain malignant poten-
tial and may be appropriate for selected cases of flat epithelial atypia, classic lobular
neoplasia, radial scar, and benign phyllodes tumor [15, 16].
10 Image-Guided Percutaneous Biopsies 181
a b
c d
Fig. 10.14 Diagnostic and therapeutic VAB for an intraductal papilloma. (a) Intraductal solid
mass in a patient presenting with bloody nipple discharge. (b) Color Doppler detected flow within
the lesion, confirming the solid nature of the mass. (c) Complete removal of the intraductal mass
with US-guided VAB. (d) Post-biopsy marker deployed immediately after the procedure. (e)
Biopsy site 4 years after the procedure, with no residual lesion
The initial planning of both VAB and CNB is very similar to FNA. The patient is
positioned in a supine or supine oblique position, with the ipsilateral arm behind the
head. The oblique position is preferred for lateral lesions, as well as axillary targets,
especially in large breasts, improving lesion visualization and needle trajectory,
allowing it to pass parallel to the chest wall.
Similar to FNA, the skin is cleansed and asepticized, the transducer is properly
covered with a sterile cover, and sterile gel is used to allow ultrasound imaging of
the targeted lesion. Once the initial preparations are complete, the operator has to
plan the skin entry point of the needle and its trajectory, taking into account the
lesion depth and location. The optimal needle visualization is obtained with a per-
pendicular angle between the needle and the transducer beam. Superficial lesions
are best approached by a skin entry point closest to the transducer, while deep
lesions require a farther entry point (Fig. 10.3).
182 V. C. Zanetta
a b
Fig. 10.16 (a) Scalpel incision in the skin and (b) core needle introduction
The entry skin point is anesthetized with lidocaine or other local anesthetic
(Fig. 10.15). The needle trajectory should also be anesthetized with lidocaine alone
or in combination with epinephrine, and the anesthetic liquid can be used to lift
lesions close to the chest wall or breast implants. While delivering anesthesia, the
operator uses this opportunity to confirm that the planned entry site in the skin and
needle trajectory is optimal for performing the biopsy.
A small incision is made in the skin with a scalpel blade, and a needle is inserted
under real-time guidance into the desired location for specimen collection
(Fig. 10.16). VAB needles are ideally placed underneath the lesion, allowing the
suction of the vacuum to assist in the lesion sampling, while CNB needles should
pierce the lesion in each sampling (multiple passes). Once sampling is adequate, a
marker should be deployed inside the lesion or in its original location in case of
complete lesion removal with VAB.
Direct compression and ice should be applied to the breast for hemostasis for
about 10 minutes. The most frequent biopsy complication is related to persistent
bleeding and hematoma formation. Since VAB yields more tissue per biopsy, there
10 Image-Guided Percutaneous Biopsies 183
may be more tissue damage and bleeding, although the suction during the procedure
is an effective measure to minimize hematomas in most cases. Usually 10 minutes
of direct compression is sufficient for adequate hemostasis for both VAB and CNB;
however, additional compression for 20 minutes or longer may be required in refrac-
tory cases. Continuous bleeding that requires surgical intervention is extremely
rare. Albeit rare, other possible complications include infection, pneumothorax, and
pseudoaneurysm formation. Implant rupture and milk fistula in lactating patients
may occur, although they do not represent a threat but rather an inconvenient
outcome.
Once hemostasis is achieved, a sterile dressing is applied to the skin wound, and
the patient may be submitted to mammographic CC and ML views to document
post-biopsy clip location. A compression bandage can be used for a few hours to
further enhance hemostasis, and the patient is discharged after receiving general
post-procedure orientations.
Lesion
Compression
paddle
+15˚ –15˚
10 Image-Guided Percutaneous Biopsies 185
different locations in each image. Using trigonometry and the apparent shift of the
target lesion, the computer software is able to calculate the depth of the target lesion
and provide coordinates in all axes.
Both stereotactic biopsy and DBT biopsy systems are offered by vendors as either
dedicated prone tables or as add-on units to standard mammogram devices
(Fig. 10.18).
In dedicated prone tables, the system is designed solely for breast biopsies. The
patient is in prone position on the table, with the target breast inserted through a
table opening. Under the table, the breast is compressed by a fenestrated compres-
sion paddle for needle introduction and tissue sampling.
On the other hand, upright add-on units are attached to a diagnostic mammogra-
phy unit; the patient is positioned either in a lateral decubitus on a table or in an
upright position seated on a chair, while the breast is compressed by a fenestrated
compression paddle for sampling. The patient is able to see the whole procedure,
including the needle manipulation and tissue sampling, which can increase vasova-
gal reactions.
The major advantage of the prone table system is that it is more comfortable for
the patients; however, these systems are more expensive and cannot be used for
routine mammographic imaging. The prone table system has a weight limit, which
can preclude its use for overweight patients who exceed the limit. Also, targeting of
far posterior lesions may be difficult as the breast cannot be pulled down enough
Image detector
Fig. 10.18 (a) A dedicated prone table; the patient lies prone on the table, with the target breast
inserted through a hole in the table. Under the table, the breast is compressed with a fenestrated
paddle, through which the biopsy needle is inserted. (b, c) Conventional mammography unit with
an add-on device for biopsy; the patient can be positioned either in a lateral decubitus or upright
(seated)
186 V. C. Zanetta
The first step is to select the most appropriate approach for breast compression
and needle introduction. The operator should review the CC and ML views and be
attentive to two factors: conspicuity of the lesion in both views and in which view
the lesion is more superficial to the skin. Ideally, the radiologist chooses the inci-
dence in which the lesion is most superficial on the skin, as long as it is well
visualized. In some situations, the operator has to choose one or the other
(Fig. 10.19).
For instance, a lesion located at the junction of the inner quadrants has the medio-
lateral view as the shortest skin to lesion approach; however, if this lesion is best
seen at the craniocaudal view, being very subtle at the lateromedial view, the opera-
tor may choose the CC approach to avoid sampling error.
After selecting the biopsy approach, the patient is positioned on the available
biopsy system (prone or upright), and the technologist acquires a scout view visual-
izing the lesion through a fenestrated compression paddle. This preliminary image
is acquired with the x-ray tube perpendicular to the breast (angle of 0°) to position
the lesion at the center of the field of view (FOV). This step may be hard, long, and
strenuous, especially for subtle findings in large breasts, given the small FOV of the
a b
Fig. 10.19 Approach selection for a pleomorphic cluster of calcifications. (a) ML view and (b)
CC view. The cluster of calcifications (circle) is closer to the inferior skin than the medial (arrows)
and is well seen in both views; thus, the best approach is caudocranial
10 Image-Guided Percutaneous Biopsies 187
a b c
fenestrated paddle. If needed, the radiologist may help in positioning, using what-
ever helpful landmarks are available, such as blood vessels or coarse calcifications
near the lesion to guide the correct positioning. The available screen in the proce-
dure room does not have the same resolution as the dedicated breast workstations
used for reporting, and therefore, the findings usually become less evident.
After finding the lesion, it is also important to check if there is any major blood
vessel overlapping the lesion, a situation that will require repositioning, for instance,
with maneuvers such as rolling.
Once the lesion is properly found and centered, the two views of the “stereopair”
are obtained. Each stereopair view is 15° off the scout view, one angled to the left
and the other angled to the right (+15° and −15°) (Fig. 10.20).
The radiologist should mark the target on each of the stereopair views so the
software can calculate X-Y and Z coordinates. Using the Z coordinate (depth), the
thickness of the breast, and the needle measurements, the radiologist has to check
the “stroke margin” to ensure that the tip of the needle will not hit the posterior
aspect of the breast (Fig. 10.21).
After targeting, the coordinates are transferred to the computer connected to the
needle probe holder. The system automatically drives the needle to the desired loca-
tion in the horizontal and vertical planes (X and Y coordinates), while advancing the
needle in depth (Z-axis) is performed manually by the operator. The radiologist
should disinfect the skin through the fenestrated paddle and administer local subcu-
taneous and deep anesthesia with lidocaine alone or in combination with epineph-
rine. For accurate local administration of anesthesia, it is helpful to manually
advance the tip of the needle close to the skin to ensure the entry point is being
numbed and seek for the needle trajectory for the deep anesthesia. A small scalpel
incision (2–3 mm) is made in the skin, and the needle is manually advanced to the
target. Once the needle is in place, the operator may request pre-fire images, two
stereopair angulated views (+15 and −15 degrees) to ensure that the lesion has not
moved due to anesthesia injection or inadvertent patient movement. The radiologist
may obviate the pre-fire images if there is confidence that no movement has
occurred. The needle is then fired deeper into the breast, and two post-fire stereopair
188 V. C. Zanetta
Target
Detector
or breast Compression
platform plate
Compression thickness
a b c
Fig. 10.22 (a, b) Post-fire stereopair images; part of the calcifications are obscured by the needle,
with the sampling chamber correctly placed. (c) The specimen radiograph shows multiple frag-
ments with pleomorphic calcifications
images are usually obtained to ensure the sampling chamber is within the target
lesion (Fig. 10.22).
Generally, samples are obtained in a clockwise fashion if the needle is right in
the center of the target; however, obtaining post-fire images can tailor sampling; for
example, if more calcifications are seen under the sampling chamber, more samples
should be obtained with the sampling chamber directed downward.
Usually, from 6 to 12 tissue fragments should be obtained. However, the number
of fragments is variable and depends on the radiologist’s discretion, needle gauge,
and lesion size. For calcified lesions, radiographs of the sample should be obtained
10 Image-Guided Percutaneous Biopsies 189
a b c
Fig. 10.23 (a) Biopsy probe slightly pulled back showing adequately deployed clip. (b, c) Post-
biopsy clip documentation in CC and ML views
Table 10.1 Positive predictive value for architectural distortions seen only on tomosynthesis
Author AD only seen on DBT Malignant outcome PPV (%)
Alshafeiy TI et al. 59 6 10.2
Partyka L et al. 9 4 44
Patel BK et al. 34 9 26
AD architectural distortion, PPV positive predictive value
Digital breast tomosynthesis (DBT) has been increasingly used since the first device
received its Food and Drug Administration (FDA) approval in 2011. The technology
uses an x-ray tube that moves in an arc over the compressed breast and captures
several images of each breast from different angles, which are reconstructed by
software into a set of “three-dimensional” images. Also, acquired data can be col-
lapsed into a single synthesized 2D image, which may obviate the need for conven-
tional 2D images, reducing radiation dose in half. The three-dimensional set of
images is converted into 1-mm sections of the breast, reducing tissue overlapping,
improving analysis of the margins, facilitating the identification of subtle findings
such as architectural distortions, and providing the location of lesions visible only
in one mammographic view [22]. Studies have shown that DBT reduces false-
positive and recall rates while improving cancer detection [8, 23, 24]. Its adoption
by medical facilities has been increasing and replacing older mammographic 2D
units (conventional full field digital mammography and computed radiography
units). To illustrate, on December 1, 2017, 3866 certified facilities had DBT units in
the United States of America (USA), and this number jumped to 6.111 on June 1,
2020. Currently, over 70% of all US certified facilities [25] have already adopted at
least one DBT unit.
With increasing number of examinations being done with DBT technology, we
have been facing a growth of findings visible only on DBT. There is still an ongoing
debate of the positive predictive value of these findings; however, initial data sug-
gests that biopsy is required. For architectural distortions (AD) seen only on DBT,
10–44% had a malignant biopsy outcome [26–28] (Table 10.1).
DBT biopsy is critical for tissue sampling of findings only seen on DBT, such as
AD, asymmetries, and some masses. Prior to DBT biopsy development, findings
seen only on DBT had to be surgically excised after preoperative localization; thus,
DBT biopsy reduces the costs and morbidity related to surgical excision. Moreover,
even for noncalcified lesions seen at 2D mammography, DBT biopsy usually depicts
these findings better than digital mammography (DM), which translates into better
confidence in targeting.
10 Image-Guided Percutaneous Biopsies 191
Table 10.2 Comparison of technical success rate, mean procedure time, and radiation exposure
between digital breast tomosynthesis and stereotactic biopsy
Mean procedure time Median number of
Author Technical success rate (min) images acquired
PSVAB DBTVAB PSVAB DBTVAB
Schrading 154/165 51/51 (100%) 27 13 8 5
et al. (93%)
Bahl et al. 410/431 695/700 27 12 12 3
(95.1%) (99.3%)
Ariaratnam N/A 38/38 (100%) N/A 15 N/A N/A
et al.
PSVAB prone stereotactic vacuum-assisted biopsy,. DBTVAB digital breast tomosynthesis vacuum-
assisted biopsy
In addition, DBT biopsy obviates the need for the stereopair images, with direct
depth calculation (Z-axis), decreasing procedure time and radiation exposure. A
large retrospective study conducted by Bahl et al. comparing 706 DBT-guided biop-
sies in upright positions with 439 stereotactic biopsies in prone tables suggested that
DBT biopsy prevailed over stereotactic as a faster procedure (12 vs 27 min), with
lower radiation dose (3 vs 12 exposures) and greater technical success (99.3% vs
95.1%) [29]. Those findings are similar to those of smaller earlier studies comparing
DBT and stereotactic guided biopsies [30, 31] (Table 10.2).
Overall, aside from lesion identification and targeting, DBT-guided biopsy is very
similar to the standard stereotactic biopsy. The choice of approach is identical,
requiring the needle to go through the shortest distance to the skin or use the inci-
dence that provides the best conspicuity of the lesion.
The patient is positioned according to the available system (upright or prone),
and tomosynthesis images or a 2D scout image is obtained by the technologist cen-
tralizing the lesion within the FOV. Once the lesion is accurately identified, a tomo-
synthesis scout image is obtained. The radiologist scrolls through the slices until he/
she finds the slice where the lesion is more conspicuous (Fig. 10.24).
In the computer screen, the target is marked and the software readily calculates
the Cartesian coordinates X, Z, and Y. It is important to emphasize when going
through the images, the radiologist should be aware if there are no vessels along the
needle path, and if applicable, repositioning should be done. The breast can be
rolled to remove the vessel from the path or change the approach view, such as
orthogonal view.
After the proper coordinates are obtained, the radiologist has to ensure that there
is sufficient tissue beyond the lesion for the needle to be fired within the breast
without hitting the posterior aspect of the breast and also that the skin is not included
in the sampling chamber of the needle. This has become very easy and intuitive with
192 V. C. Zanetta
newer software that displays a graphical representation of the needle inside the
breast, as shown in Fig. 10.25.
Once targeting has been completed, the coordinates are sent to the computer
attached to the needle probe. Asepsis of the skin, anesthesia, scalpel incision, and
needle introduction are conducted similarly to stereotactic biopsy. Once in target,
pre-fire images may be obtained, which can be either 2D stereopair angulated views
(+15 and −15 degrees) or a pre-fire tomosynthesis image set. At this stage, it is
crucial to check if there was patient movement after the lesion coordinates were
obtained. After firing the needle, a post-fire tomosynthesis set or a 2D stereopair
post-fire images may be obtained at the discretion of the radiologist; however, the
stereopair adds more radiation than the tomosynthesis set of images (Fig. 10.26).
The handling of the probe for tissue sampling, the number of fragments required,
the specimen radiographs, the procedure complications, and the post-procedure
care are no different from what has been described for stereotactic biopsy. However,
post-biopsy documentation to ensure the correct sampling with the marker in place
is performed with a single set of tomosynthesis images (Fig. 10.27).
Subtle findings may be hard to locate within the small field of view of the biopsy
equipment. To overcome this problem, before positioning the patient on the stereo-
tactic table, the lesion can be localized using a full-field 2D mammography and an
alphanumeric grid. The alphanumeric grid is used to place a metallic marker (BB)
overlying the skin, which can then be used during the procedure as a landmark to
find the target lesion [32] (Fig. 10.28).
10 Image-Guided Percutaneous Biopsies 193
Fig. 10.25 The Hologic Affirm® Prone Biopsy System uses pre-programmed needle parameters
to display a graphical representation of the selected needle, the target location, the compression
paddle, and the image receptor. In an intuitive fashion, it is possible to see all relevant spatial rela-
tionships: the location of the marked target (green), the length of the needle inside the breast (blue),
the distances between the point of entry of the skin and the beginning (white) and middle (light
gray) of the sampling chamber, as well as the distance between the needle tip and the image recep-
tor (dark gray)
a b c
Fig. 10.27 Comparison of pre- and post-biopsy images to verify sampling accuracy. (a) Pre-
biopsy tomosynthesis slice demonstrating the architectural distortion to be sampled; (b) post-biopsy
tomosynthesis slice showing adequate sampling with the clip at the desired location; and (c) pre-
biopsy 2D mammography for comparison. Note that the architectural distortion is barely seen
without tomosynthesis
a b
Fig. 10.28 (a) Full-field 2D mammography and alphanumeric grid used to locate subtle calcifica-
tions. (b) Magnified view after placing the marker overlying the skin. The BB can later be used as
landmark to find the clustered calcifications
10 Image-Guided Percutaneous Biopsies 195
During breast compression, a minimum thickness of the breast is required to fit the
needle, including its tip and sampling chamber. A minimum thickness of 30 mm is
required for standard VAB or 20 mm for a shortened sampling chamber needle, also
called petite needle (Fig. 10.29).
If there is not enough room for the needle, the stroke margin becomes negative. In
cases where even a petite needle is not enough, the operator can attempt to roll the
I
m shortened sampling chamber
a
g
e
r
e
c
e
p
t
o
r
20mm
30mm
Fig. 10.29 Illustration showing the differences between standard and shortened sampling cham-
ber needles
196 V. C. Zanetta
breast or tape up the breast against the chest wall in order to bulge the breast anteri-
orly through the opening of the paddle to increase thickness. Rarely, very thin breasts
may require adding another biopsy paddle between the posterior aspect of the breast
and the image receptor. In cases where the stroke margin is only slightly negative,
injecting superficial anesthetic to increase the skin wheal can add a few millimeters.
If available, the operator can opt to use a lateral arm extension device to perform the
insertion of the needle between the image receptor and the compression paddle, i.e.,
orthogonal to the compressed breast. In the lateral arm approach, the needle is inserted
horizontally (X) into the breast; thus, the stroke margin is greatly improved (Fig. 10.30).
To avoid unwarranted trauma of the skin, superficial lesions can be sampled with a
shortened sampling chamber needle. In addition, although the software calculates
the coordinates so that the target is right in the center of the sampling chamber,
10 Image-Guided Percutaneous Biopsies 197
I
m
a
g
e
r
e
c
e
p
t
o
r
Proximal edge
of the sampling
chamber
Z+ Z
Fig. 10.31 Illustration showing the position of the sampling chamber and the skin at the calcu-
lated depth (Z) for the target. The operator can further introduce the needle (Z+) so the sampling
chamber is able to sample the lesion at its proximal edge while avoiding the skin
sampling occurs throughout the whole sampling chamber. Taking into account the
length of the sampling chamber and its relationship to the target allows the needle
to advance a few millimeters to avoid skin trauma, while sampling is done at the
beginning of the sampling chamber (Fig. 10.31).
Of all imaging modalities, breast MRI has the highest sensitivity for detecting breast
cancer. Two meta-analyses showed a sensitivity of 90–92% and a specificity of
70–72% [34, 35]. Thus, although MRI is very sensitive, up to 30% of suspicious
findings are in fact benign, and histologic confirmation is required for a definitive
evaluation of suspicious findings (BIRADS 4 and 5). The main indication for breast
MRI-guided procedure is BIRADS 4 and 5 lesions that are not amenable to biopsies
guided by ultrasound or mammography.
198 V. C. Zanetta
MRI-guided biopsy requires a compatible and dedicated breast coil for the proce-
dure, as well as an MRI-compatible vacuum-assisted biopsy device, usually con-
taining an 8–12G needle and an introducer kit. As with stereotactic and DBT-guided
biopsies, the use of VAB is the standard of care, ensuring the procedure is performed
in a timely manner and that adequate samples are retrieved. Before starting the pro-
cedure, MRI safety should be checked, as well as contraindications to intravenous
gadolinium injection. A thorough review of the original diagnostic MR examination
is necessary to be familiar with the lesion’s morphological features and location.
The radiologist uses this information to aid in patient positioning and to select the
best approach to sample the target lesion. Older systems are limited to lateromedial
approach only; however, improvements in design made coils available from multi-
ple vendors that also allow craniocaudal and mediolateral approaches, improving
accessibility to the lesion.
The patient should be positioned in prone position, with the index breast placed
on the breast coil. The breast is compressed between the compression plate and the
biopsy grid, from which the radiologist manipulates the needle during the procedure.
200 V. C. Zanetta
Adequate compression is critical to ensure that the breast is fixed in place while
the procedure is performed; however, care should be taken not to overcompress,
which can interfere with enhancement of the target lesion.
The biopsy grid consists of evenly sized squared holes, one of which will be used
for needle insertion. Some grids have a built-in marker in one of its holes that can
be seen in non-enhanced T1-weighted sequences, while other grids require the
placement of a fiducial marker in one of its holes. Since the marker can be seen on
MR images and its actual location on the grid is known, it is used to identify a par-
ticular hole seen on the MR image and its actual location on the grid plate
(Fig. 10.32).
Pre-contrast images are obtained to check for adequate fat saturation, and the
radiologist attempts to see if the expected lesion location is within the limits of the
grid, using landmarks and the location from the original diagnostic MRI (Fig. 10.33).
Since the grid will be used to locate the needle in the horizontal and vertical planes,
if the expected lesion is not seen within the limits of the grid, the patient can be
repositioned before resuming the procedure.
a b
Fig. 10.32 (a) Breast coil and biopsy grid from Siemens (a) and from General Electric (b). The
fiducial marker is placed in one of the grid holes (arrow) to serve as a landmark for hole identifica-
tion on MR images
a b
Fig. 10.33 (a, b) Non-enhanced sagittal T1 slices showing that the expected location of the lesion
(circle), based on anatomical landmarks, is within the grid boundaries in (b)
10 Image-Guided Percutaneous Biopsies 201
a b
Fig. 10.34 (a) Crosshair placed in the center of target nonmass enhancement and (b) after scroll-
ing back, the adequate grid hole for needle introduction is selected
202 V. C. Zanetta
a b
c d
Fig. 10.35 Biopsy kit. (a) Suros biopsy kit and (b) Bard biopsy kit. Inner plastic obturator (arrow),
outer plastic cannula (arrowhead) tunneled needle guide (double arrowhead) and inner stylet
(curved arrow); (c) detail of Bard tunneled guide plastic and (d) obturator and inner stylet inserted
into the needle guide
anesthesia or bleeding, and occasionally, the coaxial system pushes and displaces
the tissue instead of cutting, which is known as the “snowplow” effect. If needed,
the inner stylet may be reinserted and necessary adjustments performed.
Once the coaxial system with the outer plastic cannula and the inner plastic obtu-
rator is confirmed in satisfactory position, the patient is again removed from the
magnet, the obturator is removed and replaced by the actual VAB needle, and sam-
ples are obtained, usually in a clockwise fashion or according to suitable tailoring of
the sampling chamber toward the lesion (Fig. 10.36).
Usually from 6 to 12 samples are obtained according to the radiologist’s discre-
tion. The VAB needle is then removed and replaced by the plastic obturator, and a
new set of images are obtained to confirm if the expected lesion location has been
sampled. Once adequate sampling has been confirmed, the obturator is removed and
a clip marker is deployed at the biopsy site through the plastic cannula.
Technical issues with MRI-guided biopsy are usually associated with thin breasts
or posterior lesions. MRI guidance allows a minimum thickness of 30 mm for stan-
dard needles and 18 mm for a petite needle. Most of the techniques previously
10 Image-Guided Percutaneous Biopsies 203
Whenever the lesions were missed during sampling, the results are considered dis-
cordant. A thorough review of all available images is required: the original diagnos-
tic examination and the biopsy and post-biopsy images. Morphological features,
size, and location of the target lesion are compared between the studies to ensure
that the suspicious lesion was indeed targeted during the biopsy. Post-biopsy images
may show reduction in size or complete removal of the lesion, especially when VAB
has been performed. If a biopsy marker has been deployed, the radiologist can com-
pare its location in relation to the target lesion, taking into account that migration
may occur. For all procedures, it is very useful to check the proper positioning of the
needle in relation to the target lesion during the procedure. For lesions with calcifi-
cations, the radiologist should obtain sample radiographs to ensure that the
204 V. C. Zanetta
The radiological features of the lesion are suspicious (BIRADS 4 and 5), and the
pathology report is malignant, for instance, a spiculated mass diagnosed as infiltrat-
ing carcinoma, or a segmental enhancement or calcifications diagnosed as DCIS. The
radiologist should communicate the results to the referring physician, and the
patient contacted and referred for appropriate therapy.
The radiological features of the lesions suggest a benign etiology, but the pathology
report is malignant. This can be either related to an incidental finding, inadequate
prior assessment of image features, or the overlap between malignant and benign
image features of some malignant lesions. Management of such cases is identical to
concordant malignant.
The radiological features suggest a benign etiology, and the pathology report is a
benign lesion. A classic example is an oval mass with circumscribed margins clas-
sified as a fibroadenoma. When establishing concordance of benign image features
and a benign histological result, it is important to differentiate benign pathological
entities that are a definitive diagnosis from benign entities that are nonspecific find-
ings. Although the list of definitive and nonspecific diagnosis may be slightly vari-
able within the literature, definitive diagnosis is lesions that have distinct image
10 Image-Guided Percutaneous Biopsies 205
features and are easily correlated with the image findings. Definitive diagnosis
includes fibroadenoma, tubular adenoma, hematoma, abscess, hamartoma (fibroad-
enolipoma), fat necrosis, granulomatous disease, lymph node, myofibroma, and
cyst contents.
On the other hand, nonspecific breast benign results on histopathology might be
less reassuring and require a careful imaging-pathology correlation. Nonspecific
benign entities include fibrocystic changes, apocrine metaplasia, duct hyperplasia,
and stromal fibrosis. Although these lesions, alone or in combination, might be
related to the image findings that led to biopsy, such as suspicious calcifications or
an indistinct mass (e.g., fibrosis), these results are often incidental findings. Thus,
establishing concordance for such entities frequently requires communication
between the radiologist and the interpreting pathologist.
Short-term imaging follow-up protocol after a concordant benign finding is not
consensual. The current NCCN guidelines (2020, version 1) support that either a
6- or 12-month follow-up is appropriate. Some authors suggest that for stereotactic
or US-guided core biopsies, a short-term 6-month follow-up should be recom-
mended to avoid delay in diagnosing a falsely negative biopsy [45, 46], while others
suggest that a short-term follow-up may be appropriate for nonspecific diagnosis
[44, 47, 48]. Recent reports have shown little benefit from short-term follow-up and
that 12-month follow-up for benign concordant lesions is ideal [49–51].
Benign and concordant biopsies guided by MRI may benefit from short-term
follow-up given its technical challenges: MRI-guided biopsy is not a real-time pro-
cedure; specimen radiograph to confirm lesion retrieval is not possible; contrast
wash-out during the procedure and bleeding may limit sampling accuracy. A study
by Hayward et al. showed a 2.4% rate of false-negative results after benign concor-
dant assessment [52]. A short-term MRI follow-up may be useful for reassuring
adequacy of tissue sampling and to avoid delays in false-negative results.
The radiological features suggest a malignant lesion, while the pathology report is
benign and cannot explain the imaging features that lead to biopsy. For instance, a
spiculated mass with microcalcifications cannot be explained by fibrocystic changes
or a fibroadenoma result. A few benign entities can present findings highly sugges-
tive of malignancy, such as granular cell tumor, fat necrosis, diabetic mastopathy,
and granulomatous mastitis. These diagnoses may be concordant but require a care-
ful correlation.
A discordant result is usually related to suboptimal or sampling error, and a thor-
ough review is required to identify possible flaws during sampling. There are sev-
eral reasons that can be related to a discordant result:
–– Inaccurate targeting – wrong location was sampled, which can be related to sub-
tle findings, patient movement, technical issues with the biopsy equipment, or
206 V. C. Zanetta
References
1. Parker SH, Lovin JD, Jobe WE, et al. Stereotactic breast biopsy with a biopsy gun. Radiology.
1990;176(3):741–7. https://doi.org/10.1148/radiology.176.3.2167501.
2. Parker SH, Lovin JD, Jobe WE, Burke BJ, Hopper KD, Yakes WF. Nonpalpable breast lesions:
stereotactic automated large-core biopsies. Radiology. 1991;180(2):403–7. https://doi.
org/10.1148/radiology.180.2.1648757.
3. Parker SH, Jobe WE, Dennis MA, et al. US-guided automated large-core breast biopsy.
Radiology. 1993;187(2):507–11. https://doi.org/10.1148/radiology.187.2.8475299.
4. Liberman L. Percutaneous imaging-guided core breast biopsy. Am J Roentgenol.
2000;174(5):1191–9. https://doi.org/10.2214/ajr.174.5.1741191.
5. Wallis M, Tarvidon A, Helbich T, Schreer I. Guidelines from the European Society of Breast
Imaging for diagnostic interventional breast procedures. Eur Radiol. 2007;17(2):581–8.
https://doi.org/10.1007/s00330-006-0408-x.
6. Chetlen AL, Kasales C, Mack J, Schetter S, Zhu J. Hematoma formation during breast core nee-
dle biopsy in women taking antithrombotic therapy. Am J Roentgenol. 2013;201(1):215–22.
https://doi.org/10.2214/AJR.12.9930.
10 Image-Guided Percutaneous Biopsies 207
7. Portnow LH, Thornton CM, Milch HS, Mango VL, Morris EA, Saphier NB. Biopsy marker
standardization: what’s in a name? Am J Roentgenol. 2019;212(6):1400–5. https://doi.
org/10.2214/AJR.18.20577.
8. Durand MA, Haas BM, Yao X, et al. Early clinical experience with digital breast tomosyn-
thesis for screening mammography. Radiology. 2015;274(1):85–92. https://doi.org/10.1148/
radiol.14131319.
9. Sharma B, Jurgensen-Rauch A, Pace E, et al. Breast implant–associated anaplastic large cell lym-
phoma: review and multiparametric imaging paradigms. Radiographics. 2020;40(3):609–28.
https://doi.org/10.1148/rg.2020190198.
10. Wang M, He X, Chang Y, Sun G, Thabane L. A sensitivity and specificity comparison of fine
needle aspiration cytology and core needle biopsy in evaluation of suspicious breast lesions:
a systematic review and meta-analysis. Breast Edinb Scotl. 2017;31:157–66. https://doi.
org/10.1016/j.breast.2016.11.009.
11. Yu Y-H, Wei W, Liu J-L. Diagnostic value of fine-needle aspiration biopsy for breast mass:
a systematic review and meta-analysis. BMC Cancer. 2012;12(1):41. https://doi.org/10.118
6/1471-2407-12-41.
12. Berg WA, Krebs TL, Campassi C, Magder LS, Sun CC. Evaluation of 14- and 11-gauge direc-
tional, vacuum-assisted biopsy probes and 14-gauge biopsy guns in a breast parenchymal
model. Radiology. 1997;205(1):203–8. https://doi.org/10.1148/radiology.205.1.9314986.
13. Nakano S, Imawari Y, Mibu A, Otsuka M, Oinuma T. Differentiating vacuum-assisted breast
biopsy from core needle biopsy: is it necessary? Br J Radiol. 2018;91(1092) https://doi.
org/10.1259/bjr.20180250.
14. Wen X, Cheng W. Nonmalignant breast papillary lesions at core-needle biopsy: a meta-analysis
of underestimation and influencing factors. Ann Surg Oncol. 2013;20(1):94–101. https://doi.
org/10.1245/s10434-012-2590-1.
15. Rageth CJ, O’Flynn EA, Comstock C, et al. First international consensus conference on
lesions of uncertain malignant potential in the breast (B3 lesions). Breast Cancer Res Treat.
2016;159(2):203–13. https://doi.org/10.1007/s10549-016-3935-4.
16. Krishnamurthy S, Bevers T, Kuerer HM, Smith B, Yang WT. Paradigm shifts in breast care
delivery: impact of imaging in a multidisciplinary environment. AJR Am J Roentgenol.
2017;208(2):248–55. https://doi.org/10.2214/AJR.16.17130.
17. Kettritz U, Rotter K, Schreer I, et al. Stereotactic vacuum-assisted breast biopsy in 2874
patients. Cancer. 2004;100(2):245–51. https://doi.org/10.1002/cncr.11887.
18. Philpotts LE, Shaheen NA, Carter D, Lange RC, Lee CH. Comparison of rebiopsy rates
after stereotactic core needle biopsy of the breast with 11-gauge vacuum suction probe ver-
sus 14-gauge needle and automatic gun. Am J Roentgenol. 1999;172(3):683–7. https://doi.
org/10.2214/ajr.172.3.10063860.
19. Huang XC, Hu XH, Wang XR, et al. A comparison of diagnostic performance of vacuum-
assisted biopsy and core needle biopsy for breast microcalcification: a systematic review
and meta-analysis. Ir J Med Sci 1971. 2018;187(4):999–1008. https://doi.org/10.1007/
s11845-018-1781-6.
20. den Dekker BM, van Diest PJ, de Waard SN, Verkooijen HM, Pijnappel RM. Stereotactic
9-gauge vacuum-assisted breast biopsy, how many specimens are needed? Eur J Radiol.
2019;120:108665. https://doi.org/10.1016/j.ejrad.2019.108665.
21. Stereotactic 11-gauge Vacuum-Assisted Breast Biopsy: Influence of Number of Specimens
on Diagnostic Accuracy - PubMed. Accessed June 21, 2020. https://pubmed.ncbi.nlm.nih.
gov/15273332/
22. Durand MA, Wang S, Hooley RJ, Raghu M, Philpotts LE. Tomosynthesis-detected architec-
tural distortion: management algorithm with radiologic-pathologic correlation. Radiographics.
2016;36(2):311–21. https://doi.org/10.1148/rg.2016150093.
23. Conant EF, Barlow WE, Herschorn SD, et al. Association of digital breast tomosynthesis vs
digital mammography with cancer detection and recall rates by age and breast density. JAMA
Oncol. 2019;5(5):635–42. https://doi.org/10.1001/jamaoncol.2018.7078.
208 V. C. Zanetta
24. Skaane P, Bandos AI, Gullien R, et al. Comparison of digital mammography alone and digi-
tal mammography plus tomosynthesis in a population-based screening program. Radiology.
2013;267(1):47–56. https://doi.org/10.1148/radiol.12121373.
25. Health C for D and R. MQSA National Statistics. FDA. Published online January 6, 2020.
Accessed June 13, 2020. https://www.fda.gov/radiation-emitting-products/mqsa-insights/
mqsa-national-statistics
26. Alshafeiy TI, Nguyen JV, Rochman CM, Nicholson BT, Patrie JT, Harvey JA. Outcome
of architectural distortion detected only at breast tomosynthesis versus 2D mammography.
Radiology. 2018;288(1):38–46. https://doi.org/10.1148/radiol.2018171159.
27. Partyka L, Lourenco AP, Mainiero MB. Detection of mammographically occult architec-
tural distortion on digital breast tomosynthesis screening: initial clinical experience. Am J
Roentgenol. 2014;203(1):216–22. https://doi.org/10.2214/AJR.13.11047.
28. Patel BK, Covington M, Pizzitola VJ, et al. Initial experience of tomosynthesis-guided vacuum-
assisted biopsies of tomosynthesis-detected (2D mammography and ultrasound occult)
architectural distortions. Am J Roentgenol. 2018;210(6):1395–400. https://doi.org/10.2214/
AJR.17.18802.
29. Bahl M, Maunglay M, D’Alessandro HA, Lehman CD. Comparison of upright digital breast
tomosynthesis–guided versus prone stereotactic vacuum-assisted breast biopsy. Radiology.
2018;290(2):298–304. https://doi.org/10.1148/radiol.2018181788.
30. Schrading S, Distelmaier M, Dirrichs T, et al. Digital breast tomosynthesis–guided vacuum-
assisted breast biopsy: initial experiences and comparison with prone stereotactic vacuum-
assisted biopsy. Radiology. 2015;274(3):654–62. https://doi.org/10.1148/radiol.14141397.
31. Ariaratnam NS, Little ST, Whitley MA, Ferguson K. Digital breast Tomosynthesis vacuum
assisted biopsy for Tomosynthesis-detected Sonographically occult lesions. Clin Imaging.
2018;47:4–8. https://doi.org/10.1016/j.clinimag.2017.08.002.
32. Chesebro AL, Chikarmane SA, Ritner JA, Birdwell RL, Giess CS. Troubleshooting to over-
come technical challenges in image-guided breast biopsy. Radiographics. 2017;37(3):705–18.
https://doi.org/10.1148/rg.2017160117.
33. Soo MS, Walsh R, Patton J. Prone table stereotactic breast biopsy: facilitating biopsy of poste-
rior lesions using the arm-through-the-hole technique. Am J Roentgenol. 1998;171(3):615–7.
https://doi.org/10.2214/ajr.171.3.9725284.
34. Zhang Y, Ren H. Meta-analysis of diagnostic accuracy of magnetic resonance imaging
and mammography for breast cancer. J Cancer Res Ther. 2017;13(5):862–8. https://doi.
org/10.4103/jcrt.JCRT_678_17.
35. Peters NHGM, Borel Rinkes IHM, Zuithoff NPA, Mali WPTM, Moons KGM, Peeters
PHM. Meta-analysis of MR imaging in the diagnosis of breast lesions. Radiology.
2008;246(1):116–24. https://doi.org/10.1148/radiol.2461061298.
36. Park VY, Kim MJ, Kim E-K, Moon HJ. Second-look US: how to find breast lesions with
a suspicious MR imaging appearance. Radiographics. 2013;33(5):1361–75. https://doi.
org/10.1148/rg.335125109.
37. Nakano S, Yoshida M, Fujii K, et al. Fusion of MRI and sonography image for breast cancer
evaluation using real-time virtual sonography with magnetic navigation: first experience. Jpn J
Clin Oncol. 2009;39(9):552–9. https://doi.org/10.1093/jjco/hyp087.
38. Mazzei MA, Di Giacomo L, Fausto A, Gentili F, Mazzei FG, Volterrani L. Efficacy of second-
look ultrasound with MR coregistration for evaluating additional enhancing lesions of the breast:
review of the literature. Biomed Res Int. 2018;2018 https://doi.org/10.1155/2018/3896946.
39. Nakano S, Kousaka J, Fujii K, et al. Impact of real-time virtual sonography, a coordinated
sonography and MRI system that uses an image fusion technique, on the sonographic evalu-
ation of MRI-detected lesions of the breast in second-look sonography. Breast Cancer Res
Treat. 2012;134(3):1179–88. https://doi.org/10.1007/s10549-012-2163-9.
40. Nakano S, Yoshida M, Fujii K, et al. Real-time virtual sonography, a coordinated sonogra-
phy and MRI system that uses magnetic navigation, improves the sonographic identification
10 Image-Guided Percutaneous Biopsies 209
Abbreviations
CC Craniocaudal
FDA Food and Drug Administration
LM Lateromedial
MG Mammography
ML Mediolateral
MR Magnetic resonance
MS Magnetic seed
NRW Nonrepositionable wires
NWL Nonwire localization
OR Operation room
PL Preoperative localization
RFID Radiofrequency identification tag
ROLL Radio-guided occult lesion localization
RSL Radioactive seed localization
RW Repositionable wires
SNOLL Sentinel node and occult lesion localization
US Ultrasound
WL Wire localization
11.1 Introduction
Before the procedure, the radiologist reviews relevant imaging findings in order to
become familiar with the target lesion and its topography and proceeds to choose
the best imaging modality for guiding the localization procedure: typically, mam-
mography (MG) or ultrasound (US) and, rarely, magnetic resonance (MR) [8, 9].
Previous discussion with the surgeon to review the PL planning is necessary to
ensure that the chosen procedure will be the most helpful considering the lesion
imaging presentation, its localization in the breast, the technical apparatus available
in the operation room, the surgeon preferences, as well as the radiologist skill and
familiarity with different localization modalities [10].
11 Breast Imaging Preoperative Localization Procedure 213
When performing wire localization (WL), a wire is introduced into the breast with
the aid of a puncture needle (Fig. 11.1) [10]. The wire is then advanced out of the
puncture needle when in the desired position, exposing its hook and anchoring in
the lesion (Fig. 11.1) [19].
When dealing with small breast lesions, the hookwire should ideally penetrate
the center of the lesion, and the maximum distance between the wire and the lesion
should be 1 cm [20, 21]. Larger breast lesions, usually represented by clustered
calcifications, may be best marked with multiple hookwires in order to achieve an
excision with tumor-free resection margins [22]. The preferred imaging modality
used for imaging-guided WL should be the one in which the target lesion is best
visualized while being tolerated by the patient and at an acceptable cost [10].
214 H. D. Skaf et al.
a b
Fig. 11.1 WL. A wire is introduced into the breast with the aid of a puncture needle (a). The
hookwire is then advanced out of the puncture needle, exposing the hook and anchoring itself in
the lesion (b)
The approach chosen for the puncture needle insertion when using a perforated or
marked compression plate for WL should, ideally, result in the shortest distance
from the skin surface to the target lesion with the intention of minimizing the dis-
tance that the needle must travel in the breast, decreasing the chance of needle
deviation from the intended target [23]. In some situations, that approach may not
be the most adequate. Inferior approaches, for example, may require uncomfortable
positioning for the patient and for the radiologist performing the procedure, who
must insert the needle from below, often from a kneeling position. Thus, breast
lesions in the inferior quadrants may be more easily localized from either the medial
or lateral side. Another situation when the shortest approach may not be used is
when the lesion is visualized with greater clarity on another particular MG projec-
tion. Taking the surgical approach into consideration when planning the skin punc-
ture site as well as the needle trajectory for PL is no longer mandatory as tumoral
cell displacement or track seeding is no longer considered a relevant factor in the
rate of local recurrence and overall survival in a context of conservative surgery plus
adjuvant radiotherapy, as many studies have demonstrated [10].
11 Breast Imaging Preoperative Localization Procedure 215
a b c d e f
Fig. 11.2 MG-guided WL of a small non-palpable breast lesion using a marked compression
plate. The first CC projection showed a clip marker (dashed circle) within the fenestrated paddle
view (a). The puncture needle was inserted, oriented by the compression plate markings, perpen-
dicularly to the skin, until it touched the opposite skin in contact with the compressor. The new CC
projection showed the hub of the needle obscuring the clip marker (b). The orthogonal MLO pro-
jection obtained by using a nonfenestrated compression paddle showed the needle extending
through and beyond the clip marker (c). Documentation in two planes after insertion of the wire
and removal of the puncture needle showed the clip marker appropriately positioned at the thick-
ened segment of the wire (d, e). The intraoperative specimen radiograph showed the distal portion
of the hookwire and the clip marker (f)
216 H. D. Skaf et al.
a b c d h
e f g
Fig. 11.3 MG-guided WL using a marked compression plate of a large non-palpable breast lesion.
WL was performed on a cluster of pleomorphic calcifications (dashed circle) (a). The MLO pro-
jection showed the lesion within the fenestrated paddle view (b). In order to achieve complete
excision of the lesion, the area of calcifications was bracketed by placing needles into both ends of
the lesion (c). The needles were inserted perpendicularly to the skin until they touched the oppos-
ing skin in contact with the compressor. The first orthogonal CC projection obtained with a non-
fenestrated compression paddle showed the needles extending through and beyond the lesion
extremities, but further than necessary (d). The depth of the needle insertions was then adjusted by
retracting them, and the new position was confirmed in another CC projection (e). Documentation
in two planes after insertion of the wires and removal of the puncture needles showed the area of
calcifications extending between the thickened segments of the two wires (f, g). The specimen
radiograph obtained after surgery showed the distal portion of the hookwires and the calcifica-
tions (h)
a b
c d
Fig. 11.4 CC and MLO projections of the case shown in Fig. 11.3 before (a, b) and after breast
conservation surgery (c, d). By removing the smallest possible amount of healthy tissue with
adequate surgical margins, conservation breast surgery provides optimal therapeutic results with
good cosmetic outcomes
218 H. D. Skaf et al.
x- and y-coordinates, and advanced to the z-coordinate. The correct needle’s tip
position may be checked using a second stereo pair image, and it should be posi-
tioned within or directly behind the target lesion. The needle is then advanced nearly
1 cm beyond the z-coordinate and the wire is deployed. Lastly, another MG imaging
in two orthogonal views is obtained to document the correct needle positioning.
When performing a stereotactic WL, especially with a hookwire, if the wire is
deployed in the same plane as the initial compression one, for example, a craniocau-
dal compression and a caudal needle insertion, without changing to an orthogonal
plane, it may hook onto tissues deeper ou superficially than the originally intented
targeted point in the z-direction and, consequently, being incorrectly located after
compression release (which is called “the accordion effect”). A positioning error of
a few millimeters in the compressed breast can correspond to a large positioning
error in the non-compressed breast. Some strategies to minimize this effect are to
gently release the breast from compression after deploying the wire [23]. Another is
to obtain a new stereo image in the orthogonal plane, then adjust the depth of needle
insertion based on this image, and then insert the wire. In modern breast biopsy
systems, another possibility is to perform the procedure using lateral needle
approach, in which the needle is inserted orthogonally to the compression plane,
thus eliminating the “accordion effect.”
Additional challenges that may arise during stereotactic WL often are the same
as those encountered during percutaneous biopsy guided by the same method and
will be discussed in more detail in a specific chapter (Fig. 11.5).
US-guided WL is performed when the lesions are visualized on this image modal-
ity, and it should always be considered due to cost-effectiveness and improved
patient comfort, especially when compared to other image modalities like stereotac-
tic guidance.
The procedure starts by positioning the patient similar to that during a percutane-
ous biopsy. The puncture needle should advance through and 1 cm beyond the target
lesion, and then the wire is deployed. Then, distances from lesion to skin and lesion
to wire tip and total inserted wire length should be documented and reported to
guide the surgeon.
Similarly, after MG or stereotactic guided WL and after US-guided procedure,
MG views in two orthogonal planes should be obtained to document the correct
needle insertion (Figs. 11.6, 11.7, and 11.8).
US-guided WL can be adapted for use in the axilla, but it has been infrequently
utilized [26]. WL complications such as pain, hematoma, and adjacent tissue injury
particularly occur in the axilla where sensitive structures (brachial plexus as well as
axillary artery and vein) are nearby [3]. In addition, there is a higher wire migration
and transection risk in the axilla compared to the breast due to arm movement and
ratcheting effect or muscular contraction [27–29].
11 Breast Imaging Preoperative Localization Procedure 219
a b e i
f
c
h
j
Fig. 11.5 WL using a stereotactic table. The MLO MG projection showed a clip marker (dashed
circle) in the right breast (a). Stereo pair (±15°) images obtained based on the scout (0°) image
were used to determine the x-, y-, and z-coordinates by targeting the clip (b). Pre-fire and postfire
stereo pair (±15°) images showed the needle extending through and beyond the clip marker (c, d).
An additional stereotactic image was obtained to confirm the depth of the needle (e). A new stereo
image in the orthogonal plane of this last image was obtained, and the depth of the needle insertion
was adjusted based on this orthogonal image in order to avoid “the accordion effect” (f). After
depth adjustment, the wire was inserted and the puncture needle was carefully removed (g, h). MG
documentation after stereotactic procedure showed the clip marker appropriately positioned at the
thickened segment of the wire (i). The specimen radiograph obtained after surgery showed the
distal portion of the hookwire and the clip marker (j)
MRI-guided WL is usually performed when the lesions are detected only on MRI,
without US or MG imaging correlates. The target lesion may be identified using
contrast-enhanced MRI guidance, in a similar way to MRI-guided percutaneous
biopsy. To locate the lesion, an image is acquired in a sagittal plane associated to a
plastic frame with reference markers (Fig. 11.9). A corresponding external location
of the lesion on the patient’s breast is determined relative to the frame and used to
guide the point of needle insertion. The puncture needle is then placed in the holder
and advanced nearly 1 cm beyond the depth estimated by using an axial reconstruc-
tion obtained from the sagittal acquisition or a new axial acquisition with a smaller
field of view in order to decrease sequence duration and reduce the possibility of
non-lesion characterization due to gadolinium washout (Fig. 11.10).
It is recommended to use wires made from MRI-compatible metals, which are
typically alloys with high titanium and nickel content, to avoid imaging artifacts
[10]. Because these special wires only have minor ferromagnetic properties, the
220 H. D. Skaf et al.
a d e
Fig. 11.6 US-guided WL with a hookwire. US image showing a hypoechoic mass with a clip
marker (dashed circle) (a). The puncture needle was advanced through and 1 cm beyond the lesion,
and then the wire was deployed (b). The distances from lesion to skin and lesion to wire tip are
documented (c). MG documentation in two planes after the procedure showed the lesion appropri-
ately positioned on the thickened segment of the wire (d, e)
a c
d e f
Fig. 11.7 US-guided WL with a hookwire. US performed after chemotherapy showed a non-mass
breast lesion with a clip marker (dashed circle) (a, b). The puncture needle was carefully advanced
through and beyond the lesion, and then the wire was deployed (1: hookwire, 2: thickened segment,
3: wire) (c). MG documentation in two planes obtained after the procedure showed the lesion
appropriately positioned at the distal portion of the thickened segment of the wire (d, e). The speci-
men radiograph obtained after surgery showed the hookwire and the clip marker (f)
11 Breast Imaging Preoperative Localization Procedure 221
a c
Fig. 11.8 US-guided WL with a retractable J-wire. The J-wire (dotted line) was placed adjacent
to a clip marker (arrow) that could be clearly visualized on US (a, b). MG documentation after the
procedure showed the clip marker appropriately positioned at the distal portion of the wire (dashed
circle) (c)
HEAD
PECTORALIS B8 A8
D6 C6 B7 A7
HEAD
FOOT
A1 A2 A3 A4 A5 A6 A7 A8
D5 C5 B6 A6 PECTORALIS
B1 B2 B3 B4 B5 B6 B7 B8
NIPPLE
E2 D4 C4 B5 A5
C1 C2 C3 C4 C5 C6
E1 D3 C3 B4 A4
D1 D2 D3 D4 D5 D6
D2 C2 B3 A3
E1 E2
D1 C1 B2 A2
NEEDLE NEEDLE
NIPPLE
GUIDE GUIDE B1 A1
A B C J F C
D E F I E B FOOT
H I J H D A
Fig. 11.9 To locate a lesion, the breast is imaged on MR while using a frame with reference mark-
ers which show up on the acquired image. The corresponding lesion location, relative to the frame,
is then determined and used to determine the biopsy needle insertion site
222 H. D. Skaf et al.
a b c
d e
Fig. 11.10 WL under contrast-enhanced MRI guidance. To locate the lesion, the sagittal MRI was
acquired associated with a plastic frame. A vitamin D capsule (arrow) was used as a skin marker
to help determine the corresponding external location of the breast lesion relative to the frame (a,
b). MR images acquired after contrast injection showed the target lesion, a focal non-mass
enhancement (dashed circle) (c, d). MR image acquired after insertion of the coaxial needle
showed its correct positioning within the focal non-mass enhancement (e)
susceptibility artifact caused by them produces a signal extinction that can be dis-
tinctly seen without masking relevant findings and fine structures.
Access to an MRI-guided WL is sometimes poor. It is worth mentioning that a
clip marker placed in the cavity after an MRI-guided vacuum-assisted biopsy not
only enables the radiologist to localize the biopsied area once again but also allows
other imaging methods beyond MRI to be utilized in order to perform PL. Therefore,
an originally MRI-visualized lesion can be preoperatively localized using cheaper
imaging guidance, like mammography or ultrasound, for example, by using an MRI
inserted marker.
11 Breast Imaging Preoperative Localization Procedure 223
There are various designs of localization wires that influence the tenacity of it stay-
ing in place, which is also determined by the composition of the breast parenchyma
[25]. In this regard, in patients with fatty breasts, the wire may dislocate more easily
because of the limited stroma for anchorage.
In general, wire variations can be classified into nonrepositionable and reposi-
tionable types, and both have advantages and disadvantages (Figs. 11.11 and 11.12).
A nonrepositionable wire (NRW), such as a hookwire, is more stable than a repo-
sitionable one, preventing movement out of the breast. It also optimizes intraopera-
tive identification of the localized lesion, since the NRW frequently presents a
thickened distal segment that provides a tactile guide to the surgeon. However, this
type allows only one-way forward movement, and once the hook is deployed, it can-
not be drawn back into the puncture needle. Thus, if the hook is improperly placed
or pushed into the breast, it can be permanently displaced away from the tar-
geted area.
A repositionable wire (RW), such as a retractable J-wire, can be withdrawn from
the puncture needle and repositioned if necessary, as many times as needed. Besides
being less stable than a nonrepositionable one due to its design, the absence of the
thickened distal segment on RW can also limit intraoperative identification of the
localized lesion.
As both types have been shown to be effective in localizing breast lesions, the
choice of the needle is largely a matter of service material availability and surgeon’s
and radiologist’s preference [23].
a b
Fig. 11.11 WL. After accurate placement of the puncture needle in the breast, either a nonreposi-
tionable hookwire (a) or a repositionable retractable curved-end wire (or J-wire) (b) can be
inserted. Both types have been shown to be effective in localizing breast lesions
224 H. D. Skaf et al.
a b c d e f g h
Fig. 11.12 Some of the various designs of localization wires. (a–e) represent nonrepositionable
wires, and (f–h) represent repositionable wires
WL has been the standard for PL in breast imaging for decades due to its simplicity
and low cost. However, the available options for performing it have expanded
greatly and now include the use of substances (such as carbon and radiotracers) or
nonwire localization (NWL) devices (such as radioactive and magnetic seeds, radar
reflectors, and radiofrequency identification tags).
Compared with wires, these NWL alternatives, except for radiotracers, can be
placed prior to the surgery date, at the patient’s convenience, in contrast to the WL
device procedures that are performed on the day of surgery. Radioactive seeds can
be placed up to 5–7 days before, and carbon, radar reflectors, magnetic seeds, and
RFID tags have been approved for long-term placement (more than 30 days), with
no restriction on the length of time that they can remain in the breast tissue, which
can be very useful in patients undergoing neoadjuvant systemic therapy.
Consequently, this decoupling of the radiology and surgery schedules results in
fewer delays, minimizes presurgical fasting as the resection can be the first surgery
of the day, reduces the risk of vasovagal syncope, and overall improves patient
satisfaction.
Another advantage of using NWL methods is the absence of a wire partially
outside the breast, improving patient experience, and removing the possibility of
wire migration.
Additionally, when the surgeon makes use of wire localization techniques, the
wire and the healthy tissue along its course are usually retrieved. In contrast, with
NWL, the surgeon keeps a continuous intraoperative target tracking in order to keep
the lesion in the center of the specimen, retrieving a smaller quantity of nontargeted
healthy tissue and improving cosmesis. Besides, wire placement affects the surgical
planning because it must be fully retrieved, sometimes determining the incision site
and increasing unnecessary healthy tissue removal.
11 Breast Imaging Preoperative Localization Procedure 225
There are commercially available carbon suspension products approved by the Food
and Drug Administration (FDA) for tattooing the gastrointestinal tract that can be
used in PL of breast lesions and axillary lymph nodes, although this use is not cur-
rently FDA approved and should be considered off label [30–37].
A sterile solution can be prepared using 4 g of activated charcoal in 100 ml of
0.9% saline solution. And 0.2–0.3 ml of nonionic radiographic contrast medium can
be added when performing MG-guided localization for subsequent image docu-
mentation of correct dispersion, as charcoal is not often identified radiographically
[23]. The charcoal is suspended quickly, so it is important to shake the bottle well
before drawing it into the syringe. Approximately 1–1.5 ml of the solution is injected
into the target lesion or directly proximal to it to achieve sufficient dispersion and
visualization. Then a fine line of the solution extending all the way to the skin is
injected as the needle is withdrawn. Being particulate and not soluble in water, the
carbon solution remains on the path [23, 34–36]. During surgery, the targeted lesion
is localized by visualization of the skin mark and dissection along the carbon track
(Fig. 11.13).
A particular advantage of this PL method is the ease of locating the injection site
during the specimen histopathological examination. However, the coal can render
breast tissues more resistant to microtomy, occulting or distorting the lesion on
microscopy.
There are few reported disadvantages of using carbon for PL. The carbon sus-
pension does not diffuse significantly, but it can rarely spread into the surrounding
tissues, particularly by accidental intraductal injection [23]. In this case, the demar-
cated area can be larger than initially expected, consequently increasing unneces-
sary healthy tissue removal. The surgeon can also remove a larger amount of breast
Fig. 11.13 Several of the methods currently used to localize lesions in the breast may be adapted
for use in the axilla. Axillary lymph node carbon marking permits the surgeon to locate the target
lymph node by visualization of the skin mark and dissection along the carbon trail to the lesion.
(With permissions from Lucas Roskamp Budel, MD)
226 H. D. Skaf et al.
tissue when it faces difficulty to identify the demarcated area intraoperatively, espe-
cially in deeper tissues, where the carbon track is harder to palpate [30].
Carbon marking may be performed immediately after biopsy, obviating a sepa-
rate PL, which is an advantage cost-wise [38]. However, when resection of the
marked biopsy path is not accomplished in 6 months, local granulomatous reactions
can occur and mimic suspicious lesions [39–41]. There are reported cases of mam-
mographic lesions coinciding with previous coal marking sites, whose further
investigation yielded charcoal granulomas.
Sentinel lymph node identification using radiotracers and ROLL can be performed
in combination for the same surgical session in a procedure called sentinel node and
occult lesion localization (SNOLL) [42, 46, 47]. It consists of peritumoral injection
11 Breast Imaging Preoperative Localization Procedure 227
a c d e
f
b
Fig. 11.14 MG-guided ROLL using a marked compression plate. The MLO projection showed a
clip marker (dashed circle) while using a fenestrated paddle (a). The puncture needle was inserted,
oriented by the compression plate markings similar to WL (b). The orthogonal CC projection
obtained with a nonfenestrated compression paddle showed the needle extending through and
beyond the clip marker, but further than necessary (c). The depth of the needle insertion was then
adjusted, and the new position was confirmed in another CC projection. Once in the correct posi-
tion, the radiotracer was injected through the puncture needle (d). The scintigraphic image obtained
after the procedure showed the presence of a focal spot of tracer accumulation corresponding to the
breast localized lesion (e). The specimen radiograph obtained after surgery showed the clip
marker (f)
a b
c d
Fig. 11.15 Stereotactic guided ROLL. The scout (0°) image showed a clip marker (dashed circle)
(a). The stereo pair (±15°) images obtained based on the scout were used to determine the x-, y-,
and z-coordinates by targeting the clip (b). After needle insertion, a new stereo pair (±15°) image
showed the tip of the needle in the correct position; then the radiotracer was injected (c). The scin-
tigraphic image obtained after the procedure showed the presence of a focal spot of tracer accumu-
lation corresponding to the clip (d)
a c d e
b f
Fig. 11.16 MG-guided SNOLL. The MLO projection showed pleomorphic clustered calcifica-
tions (dashed circle) while using the fenestrated paddle (a). The puncture needle was inserted,
oriented by the compression plate markings similar to WL (b). The orthogonal CC projection
obtained with a nonfenestrated compression paddle showed the needle extending through and
beyond the lesion (c). The depth of the needle insertion was then adjusted by retracting it, and the
new position was confirmed in another CC projection. Once in the correct position, the radiotracer
was injected (d). The scintigraphic image obtained after the procedure showed the presence of two
spots of tracer accumulation corresponding to the lesion and the sentinel lymph node in the ipsilat-
eral axilla (e). The specimen radiograph obtained after surgery successfully showed the presence
of the targeted pleomorphic clustered calcifications in it (f)
Cost-wise, at first glance, these current nonwire localization devices may seem
more expensive than wires. Implementing these systems requires investment in the
reusable console and probe, which are the most expensive components, and the
single-use device itself is costlier than a wire. But they may not be unfavorable on a
cost-benefit analysis, as the elimination of operation room delays as well as
increased schedule flexibility may result in cost savings, particularly in a bundled
payment system [52].
a b c f
Fig. 11.17 US-guided SNOLL. MG images (a, b) obtained after chemotherapy showing a focal
asymmetry with a clip marker (dashed circle), more evident with Eklund maneuver (b). US image
showing a hypoechoic mass with a clip marker (arrow) corresponding to the lesion observed on the
mammogram (c). The puncture needle was advanced to the lesion, and the distance from its tip to
skin was documented (d). Once in the correct position, the radiotracer was injected adjacent to the
lesion (e). The scintigraphic image obtained after the procedure showed the presence of two spots
of tracer accumulation corresponding to the localized lesion and the sentinel lymph node in the
ipsilateral axilla (f)
a b c
d e f
Fig. 11.18 Another example of US-guided SNOLL. MG image obtained after chemotherapy
showing a small mass with a clip marker (dashed circle) (a). US image showing a hypoechoic mass
associated with a clip marker (arrow) corresponding to the mammographic lesion (b). The punc-
ture needle (dotted line) was advanced to the lesion, and the distance from its tip to skin was docu-
mented (c, d). Once in the correct position, the radiotracer was injected adjacent to the lesion (e).
The scintigraphic image obtained after the procedure showed the presence of two spots of tracer
accumulation corresponding to the localized lesion and the sentinel lymph node in the ipsilateral
axilla (f)
11 Breast Imaging Preoperative Localization Procedure 231
a b c d
Fig. 11.19 Some schematic examples of current nonwire localization devices, such as iodine
radioactive seed (a), radar reflector (b), magnetic seed (c), and radiofrequency identification tag (d)
a b c
Titanium capsule
Adsorbed I-125
0,8 mm
d 4,5 mm
Fig. 11.20 A digital mammographic craniocaudal view of the right breast containing a post-
biopsy marker alongside a radioactive seed (a). As depicted in the magnified view, despite having
the same radiological density, the seed is thinner and longer than the marker (b). On ultrasound,
the radioactive seed is clearly seen as a linear, echogenic structure, within the targeted breast mass
(c). (With permissions from Fernanda Philadelpho Arantes Pereira, MD, PhD, from Dasa).
Schematic drawing of a radioactive seed example (d)
a b c
Fig. 11.21 Radioactive seed deployment under sonographic guidance of an irregular retroareolar
mass on the left breast (a) as well as an ipsilateral suspicious axillary lymph node (b). Left breast
mediolateral oblique mammographic view showing both lesions associated with correctly posi-
tioned markers (c). (With permissions from Aline Campos, MD, and Carla Tajima, MD, from
BP – A Beneficência Portuguesa de São Paulo)
From the patient’s point of view, those who undergo RSL had fewer vasovagal
reactions at insertion in comparison to those who undergo wire localization, prob-
ably because patients were not fasting for surgery on the day the localization proce-
dure was performed; additionaly, radioactive seeds are usually deployed quicker than
wires are positioned, resulting in less algic stimulus thus decreasing the odds of a
vasovagal reaction [58].
Another benefit of RSL is the possibility to use the same gamma probe, only
adjusting the activity detection setting on the probe, to localize and excise sentinel
11 Breast Imaging Preoperative Localization Procedure 233
a b c d e
Fig. 11.22 Radioactive seed deployment under mammographic guidance using a marked com-
pression plate. Digital mammography mediolateral (a) and craniocaudal (b) views depicting a
post-biopsy marker on the junction of the outer quadrants of the left breast on the site with an
established diagnosis. Digital mammography craniocaudal view using a marked compression plate
showing the radioactive seed insertion needle in a lateral approach with its tip next to post-biopsy
marker (c) and the seed adequate deployment (d, e). (With permissions from Aline Campos, MD,
and Carla Tajima, MD, from BP – A Beneficência Portuguesa de São Paulo)
nodes submitted for technetium 99 m localization, as the 125I seeds emit gamma rays
with a photon energy of 27 keV, while technetium 99 m has a 140-keV activ-
ity signal.
Unlike other nonwire localization techniques, there is no reported depth limita-
tion for detectability when RSL is utilized.
However, any RSL program must follow strict regulations due to the radioactiv-
ity involved and requires massive inter-departmental coordination for its execution.
Therefore, establishing a radioactive seed program can take some time, in contrast
to other nonwire localization programs which can be set up relatively quickly, and
limits the widespread adoption of this localization technique.
Contrary to a migrated biopsy clip, a migrated radioactive seed must be excised
and disposed accordingly due to the radioisotope presence. After the surgery, it is
placed in a lead container and sent back to the nuclear medicine service for safe
disposal. If the seed’s removal takes longer than 5 days, a radiation safety officer
must be notified, and the patient must be monitored until its removal.
Another RSL limitation is that these patients may not undergo MR imaging
while the seed is in place due to the fact that the gamma probe to detect eventually
extruded RSL seeds is not MR compatible (Zone IV), precluding recovering of a
potentially lost seed and limiting long-term RSL use for patients who have MR
follow-up imaging in the neoadjuvant setting.
During radioactive seed localization, it is crucial to not discard any material used
in the procedure until the mammography confirms its placement, which will aid in
the recovery if a seed is lost.
234 H. D. Skaf et al.
This localization technique uses radar technology and was first introduced in 2014,
being composed of a device detected by a specific intraoperative probe that emits
infrared light to excise the area of interest which can be placed under ultrasound or
mammographic guidance in a similar way as a biopsy marker clip (Fig. 11.23).
One of the first radar reflector devices, the SAVI Scout System® (Merit Medical
Systems, USA), measures 12 mm in length, with a 4-mm body and two 4-mm
antennas on each side, including an infrared light detector. Being FDA approved for
long-term placement, it is biocompatible and inert, responding only to signals emit-
ted from the respective reader, and can be implanted at any time prior to surgery.
The reflector is deployed using a 16G needle by withdrawing a release button,
rather than pushing the reflector, in order to prevent antenna bending. After
a c e
b d f
Fig. 11.23 A 12-mm radar reflector with a 4-mm body and two antennas on either side (a).
Console with an attached handpiece probe (b). Mammographic (c), sonographic (e), surgical spec-
imen (d), and MRI (f) appearance of the radar reflector, (© Merit Medical, reprinted with
permission)
11 Breast Imaging Preoperative Localization Procedure 235
displacement, the radiologist uses a console to make sure that an audible signal can
be obtained from the radar reflector. Researchers have successfully detected reflec-
tors at a depth of up to 8 cm; however, in certain circumstances, a radar placed
deeper than 6 cm may not produce a detectable signal through the skin, unlike radio-
active seeds, and additional compression to tissue with the handpiece may be
required [59, 60].
In the OR, the detector console emits power to the infrared light source through
the hand probe, activating it, which results in the reflection of an electromagnetic
wave signal back to the handpiece. The console then processes these signs and pro-
vides visual and audible feedback to the surgeon.
In contrast to the RFID tag and magnetic seed devices, which can cause MRI
susceptibility signal void artifact, the radar reflector causes minimal artifact, and the
patient with the device in place may safely undergo MRI at 3 T or less. However, the
Scout system is costlier than WL or RSL.
Clear surgical margin rates achieved by using radar reflector-guided excision
have ranged from 85% to 93%, and when compared to wire localization surgical
outcomes, researchers found no substantial differences in margin positivity, close
margin, and re-excision rates [19, 60, 61]. Another study comparing the SAVI Scout
system to WGL and RSL found no difference in margin positivity, specimen vol-
ume, or 30-day complication rate but did find that the SAVI Scout device reduced
the overall operating time [62].
Failures related to radar reflector technique are typically due to a lack of signal
from the device: dense objects such as calcified masses, hematomas or wires, as
well as certain plastics, positioned between the reflector and the handpiece, can
interfere with signal detection [61, 63]. Halogen and some older model OR lights
were shown to affect detection of the reflector as well; however, shielding the breast
from these lights or redirecting them while using the handpiece solved these issues.
Besides, LED lights, which are more prevalent than halogen lights in these settings,
did not interfere with radar detection [60].
On top of it, this system cannot be placed in patients with a nickel allergy, as the
radar reflector antennae are made of nitinol, an alloy of nickel and titanium.
In comparison to RSL, radar reflector localization lacks radioactivity and regula-
tory issues, can be implanted for longer periods and is associated with minimum
susceptibility artifact on MRI. However, it should not be placed within or deep to a
hematoma, there is the possibility of an allergic reaction in some patients due to the
presence of nitinol and the Scout system costs more than WL or RSL.
The Magseed® (Endomagnetics, Cambridge, UK) device was FDA cleared in 2016,
being a stainless-steel marker measuring 5 × 1 mm, the smallest among non-
radioactive localization devices. It is deployed under mammogram or ultrasound
guidance, in a similar way to a biopsy marker or RSL, through a preloaded 18G
needle introducer.
236 H. D. Skaf et al.
with the deeper range can be draped for use in a sterile surgical environment and is
reusable, while the one with the smaller range is a pencil-sized, single-use device.
On the reader screen, associated with an audible signal, the distance from the lesion
to the probe can be viewed in millimeters as well as the tag’s unique identifica-
tion number.
There are no clear contraindications to RFID tags nor any adverse effects associ-
ated with their use. The tags have a long history of use similar to those embedded in
livestock, pets, and breast implants as a form of identification.
Maggie et al. found that the clear surgical margin rate associated with RFID tag-
guided excision was 97% in a subset of 33 patients [66].
It is MRI conditional; however, the ferrous and copper material creates a 2.5 cm
susceptibility artifact when imaging is performed using a gradient-echo pulse
sequence in a 3 T MRI unit.
We summarize and compare the main features of the aforementioned localiza-
tion techniques in Table 11.1.
Strengths Cost effective Can be placed No depth No depth limitation Small size Axillary node Pencil size
No depth with MRI limitation Console and probe Axillary node localization surgical probe
limitation guidance Can be placed are the Same used localization available
Can be placed with MRI for sentinel lymph Each tag has a
with MRI guidance node biopsy unique ID
guidance Axillary node number visible
localization on console
Weaknesses Scheduling Potentially Scheduling Radiation safety Requires nonferrous Contains nickel MRI artifact
inflexibility difficult difficulties concerns surgical tools MRI artifact Axillary use is
Presurgical visualization on Radiation Lengthy Process to Largest susceptibility Multiple factors still off label
Patient will be imaging methods safety begin Program artifact on MRI may limit signal
in fasting state Foreign-body concerns Contraindicated in detection on OR
Potential reaction that may Lengthy patients with
discomfort mimic malignancy Process to implanted cardiac
while waiting begin Program devices
for surgery
Breast Imaging Preoperative Localization Procedure
239
240 H. D. Skaf et al.
The radiologist estimates the projection of the tumor on the skin surface, positions
a marker in the corresponding spot, and reports the skin-to-lesion as well as skin-
to–localization device depths, as well as the lesion distance from the nipple and
pectoral muscle. After the procedure, the radiologist must summarize it for the sur-
geon and even consider a call to discuss the procedure, particularly if the localiza-
tion device is not in an ideal position.
After the target tissue is excised, the specimen is labeled for orientation, and
radiographs are obtained to confirm the removal of both the lesion and the localiza-
tion device. Surgical specimens, excised under needle localization guidance, should
be radiographed for several reasons. The radiography verifies that the entire lesion
has been removed, serves as a lesion location guide in the specimen to the patholo-
gist, and certifies that the wire has been removed from the breast. This is an absolute
requirement when localization is performed for calcification, architectural distor-
tion, or asymmetries. If the entire length of the wire is not visualized on the speci-
men radiograph, it is imperative to report this information to the surgeon.
Intraoperative radiography or postoperative imaging is appropriate to identify the
retained fragment [67].
11 Breast Imaging Preoperative Localization Procedure 241
References
23. Dershaw DD. Imaging-guided interventional breast techniques. Springer Science & Business
Media; 2002. p. 31–52.
24. Helvie MA, Ikeda DM, Adler DD. Localization and needle aspiration of breast lesions: com-
plications in 370 cases. AJR Am J Roentgenol. 1991;157(4):711–4.
25. Sylvia H, Dershaw D, Schreer I. Diagnostic breast imaging; 2001. p. 152–60.
26. Woods RW, Camp MS, Durr NJ, Harvey SC. A review of options for localization of axillary
lymph nodes in the treatment of invasive breast cancer. Acad Radiol. 2019;26(6):805–19.
27. Homer MJ. Transection of the localization hooked wire during breast biopsy. Am J Roentgenol.
1983;141(5):929–30.
28. Martinez SR, Gelfand M, Hourani HS, Sorrento JJ, Mohan EP. Cardiac injury during needle
localized surgical breast biopsy. J Surg Oncol. 2003;82(4):261–5.
29. Bristol J, Jones P. Transgression of localizing wire into the pleural cavity prior to mammogra-
phy. Br J Radiol. 1981;54(638):139–40.
30. Choy N, Lipson J, Porter C, Ozawa M, Kieryn A, Pal S, et al. Initial results with preoperative
tattooing of biopsied axillary lymph nodes and correlation to sentinel lymph nodes in breast
cancer patients. Ann Surg Oncol. 2015;22(2):377–82.
31. Patel R, MacKerricher W, Tsai J, Wood L, Allison K, Wapnir I. Pathological confirmation of
pre-chemotherapy biopsied and tattooed axillary lymph nodes; 2018. p. 426–7.
32. Rose A, Collins J, Neerhut P, Bishop C, Mann G. Carbon localisation of impalpable breast
lesions. Breast. 2003;12(4):264–9.
33. Ko K, Han B-K, Jang KM, Choe YH, Shin JH, Yang J-H, et al. The value of ultrasound-guided
tattooing localization of nonpalpable breast lesions. Korean J Radiol. 2007;8(4):295–301.
34. Svane G. A stereotaxic technique for preoperative marking of non-palpable breast lesions.
Acta Radiol Diagn. 1983;24(2):145–51.
35. Langlois SLP, Carter ML. Carbon localisation of impalpable mammographic abnormalities.
Australas Radiol. 1991;35(3):237–41.
36. Canavese G, Catturich A, Vecchio C, Tomei D, Estienne M, Moresco L, et al. Pre-operative
localization of non-palpable lesions in breast cancer by charcoal suspension. Eur J Surg Oncol.
1995;21(1):47–9.
37. Mullen DJ, Eisen RN, Newman RD, Perrone PM, Wilsey JC. The use of carbon marking after
stereotactic large-core-needle breast biopsy. Radiology. 2001;218(1):255–60.
38. Franceschini G, Mason EJ, Grippo C, D’Archi S, D’Angelo A, Scardina L, et al. Image-
guided localization techniques for surgical excision of non-palpable breast lesions: an over-
view of current literature and our experience with preoperative skin tattoo. J Personal Med.
2021;11(2):99.
39. Ruiz-Delgado M, López-Ruiz J, Sáiz-López A. Abnormal mammography and sonography
associated with foreign-body giant-cell reaction after stereotactic vacuum-assisted breast
biopsy with carbon marking. Acta Radiol. 2008;49(10):1112–8.
40. Cavalcanti TCS, Malafaia O, Nassif PAN, Skare TL, Ogata DC, Miguel MT, et al. Non-
palpable breast lesions marked with coal suspension: evaluation of anatomopathological
aspects, viability of interpretation and inflammatory response. Revista do Colegio Brasileiro
de Cirurgioes. 2012;39(6):469–75.
41. de O Salvador GL, Barbieri PP, Maschke L, ALA N, Louveira MH, Budel VM. Charcoal
granuloma mimicking breast cancer: an emerging diagnosis. Acta Radiol Open.
2018;7(12):2058460118815726.
42. Veronesi U. Conservative surgery 23. Breast cancer: innovations in research and management;
2017. p. 247–64.
43. Luini A, Zurrida S, Galimberti V, Paganelli G. Radioguided surgery of occult breast lesions.
Eur J Cancer (Oxford, England: 1990). 1998;34(1):204–5.
44. Grüning T, Brogsitter C, Jones IW, Heales JC. Resolution recovery in planar bone scans: diag-
nostic value in metastatic disease. Nucl Med Commun. 2012;33(12):1307–10.
45. Postma EL, Koffijberg H, Verkooijen H, Witkamp A, Van Den Bosch M, Van Hillegersberg
R. Cost-effectiveness of radioguided occult lesion localization (ROLL) versus wire-guided
11 Breast Imaging Preoperative Localization Procedure 243
localization (WGL) in breast conserving surgery for nonpalpable breast cancer: results from a
randomized controlled multicenter trial. Ann Surg Oncol. 2013;20(7):2219–26.
46. Paganelli G, De Cicco C, Luini A, Cassano E, Pizzamiglio M, Fiorenza M, et al. Radioguided
surgery in non-palpable breast lesions. Eur J Nucl Med Mol Imaging. 1997;24:893.
47. De Cicco C, Trifiro G, Intra M, Marotta G, Ciprian A, Frasson A, et al. Optimised nuclear
medicine method for tumour marking and sentinel node detection in occult primary breast
lesions. Eur J Nucl Med Mol Imaging. 2004;31(3):349–54.
48. Ahmed M, Douek M. Sentinel node and occult lesion localization (SNOLL): a systematic
review. Breast. 2013;22(6):1034–40.
49. Krynyckyi BR, Shim J, Kim CK. Internal mammary chain drainage of breast cancer. Ann Surg.
2004;240(3):557.
50. Price ER, Khoury AL, Esserman LJ, Joe BN, Alvarado MD. Initial clinical experience with an
inducible magnetic seed system for preoperative breast lesion localization. Am J Roentgenol.
2018;210(4):913–7.
51. US Nuclear Regulatory Commission. iodine-125 and palladium-103 low dose rate brachy-
therapy seeds used for localization of non-palpable lesions. Washington, DC: US Government
Printing Office; 2015. p. 27. http://www.nrc.gov/materials/miau/med-use-toolkit/seed-
localization.html
52. Loving VA, Edwards DB, Roche KT, Steele JR, Sapareto SA, Byrum SC, et al. Monte Carlo
simulation to analyze the cost-benefit of radioactive seed localization versus wire localization
for breast-conserving surgery in fee-for-service health care systems compared with account-
able care organizations. Am J Roentgenol. 2014;202(6):1383–8.
53. Jakub JW, Gray RJ, Degnim AC, Boughey JC, Gardner M, Cox CE. Current status of radioac-
tive seed for localization of nonpalpable breast lesions. Am J Surg. 2010;199(4):522–8.
54. McGhan LJ, McKeever SC, Pockaj BA, Wasif N, Giurescu ME, Walton HA, et al. Radioactive
seed localization for nonpalpable breast lesions: review of 1,000 consecutive procedures at a
single institution. Ann Surg Oncol. 2011;18(11):3096–101.
55. Hughes JH, Mason MC, Gray RJ, McLaughlin SA, Degnim AC, Fulmer JT, et al. A multi-site
validation trial of radioactive seed localization as an alternative to wire localization. Breast
J. 2008;14(2):153–7.
56. Sharek D, Zuley ML, Zhang JY, Soran A, Ahrendt GM, Ganott MA. Radioactive seed localiza-
tion versus wire localization for lumpectomies: a comparison of outcomes. Am J Roentgenol.
2015;204(4):872–7.
57. Dryden MJ, Dogan BE, Fox P, Wang C, Black DM, Hunt K, et al. Imaging factors that influ-
ence surgical margins after preoperative 125I radioactive seed localization of breast lesions:
comparison with wire localization. Am J Roentgenol. 2016;206(5):1112–8.
58. Zhang Y, Seely J, Cordeiro E, Hefler J, Thavorn K, Mahajan M, et al. Radioactive seed local-
ization versus wire-guided localization for nonpalpable breast cancer: a cost and operating
room efficiency analysis. Ann Surg Oncol. 2017;24(12):3567–73.
59. SAVI SCOUT reflector and delivery system instructions for use. Cianna Medical. 2016;2019.
https://www.ciannamedical.com/wp-content/uploads/2016/11/Instructions-for-Use-Reflector-
Delivery-System.pdf. Published 2016. Accessed 8 May 2019.
60. Cox CE, Russell S, Prowler V, Carter E, Beard A, Mehindru A, et al. A prospective, single arm,
multi-site, clinical evaluation of a nonradioactive surgical guidance technology for the location
of nonpalpable breast lesions during excision. Ann Surg Oncol. 2016;23(10):3168–74.
61. Mango VL, Wynn RT, Feldman S, Friedlander L, Desperito E, Patel SN, et al. Beyond wires and
seeds: reflector-guided breast lesion localization and excision. Radiology. 2017;284(2):365–71.
62. Srour MK, Kim S, Amersi F, Giuliano AE, Chung A. Comparison of wire localization,
radioactive seed, and Savi scout® radar for management of surgical breast disease. Breast
J. 2020;26(3):406–13.
63. Falcon S, Weinfurtner RJ, Mooney B, Niell BL. SAVI SCOUT® localization of breast lesions
as a practical alternative to wires: outcomes and suggestions for trouble-shooting. Clin
Imaging. 2018;52:280–6.
244 H. D. Skaf et al.
12.1 Introduction
Screening tests are done to detect potential health disorders or diseases in people
who do not have any symptoms of the disease. The goal is early detection to enable
effective treatment or prevention through lifestyle changes. The disease must be
serious and treatable. The exam must be safe, acceptable, and of low cost [1].
For breast cancer, mammography, as a screening test can offer conservative sur-
gery and decrease mortality. Screening mammography detects 2–8 cancers per 1000
mammograms [2], and some guidelines show a 20% of mortality reduction [3].
Periodicity of mammography is controversial, as well as the ideal age to start and
finish scheduled tests. Societies around the world offer different recommendations.
The Brazilian College of Radiology and the Brazilian Society of Breast Cancer
recommends from the age of 40 without a predetermined age for the interruption,
testing women up to the life expectancy of 7 years. The US Preventive Services Task
Force (USPSTF) recommendation (2009 and 2016) is biennial screening mammog-
raphy for women aged 50–74 years. Annual screening mammography of women
aged 40–84 is known to prevent more deaths from breast cancer than biennial
screening of women 50–74 years old. No medical society recommends screening
before 40 years.
On the other hand, mammography is not innocuous. Its main adverse effect is
radiation exposure. Other problems may arise from false-positive results and over-
diagnosis leading to overtreatment.
This type of mammography was used in the initial studies that highlighted the
advantage of screening for breast cancer (Table 12.1). The image needs to be devel-
oped and fixed chemically.
12.4 Tomosynthesis
In some cases, the fibroglandular tissue can hide lesions or hinder the interpretation
of the exam. With tomosynthesis, a moving x-ray source acquires multiple projec-
tions in each incidence and creates thin sections that could help with the masking
problem.
The combined use with mammography can reduce the rate of recall by 30%, but
it increases exposure to radiation, which generates debate about its routine use in
screening.
Magnetic resonance imaging is used regularly to screen for breast cancer in women
at high family risk (>20% lifetime), because MRI has the highest sensitivity for the
detection of invasive as well as of intraductal cancer [7]. On the other hand, MRI
screening has a lower specificity than mammography, and as a result, MRI will
generate more findings judged as uncertain, requiring short-term follow-up or addi-
tional investigations [8].
Kriege et al., in a prospective study, compared the efficacy of mammographic
and MRI screening for breast cancer in women with a positive family history or a
genetic predisposition to breast cancer [9]. Among the women examined by both
methods at the same screening visit, 45 breast cancers were detected (including 6
ductal carcinomas in situ): 32 by MRI (sensitivity, 71.1%) and 18 by mammography
(40%). The sensitivity of MRI was higher than that of mammography, but both the
specificity and positive predictive value of MRI were lower. MRI screening led to
twice as many unnecessary additional examinations as mammography (420 vs. 207)
and three times as many unnecessary biopsies (24 vs. 7) [9].
Despite MRI screening of the breast can detect occult malignancies better than
mammography, several factors must be taken into consideration. MRI is very expen-
sive and is an invasive procedure requiring an intravenous injection of a contrast
agent. It is also contraindicated in women with claustrophobia, pacemakers, or
aneurysm clips. Additionally, low specificity can cause an anxiety regarding the
result of supplemental screening, such as emotional burden, concerns about false-
positive results, or overdiagnosis and aversion to hospitals [10].
Berg et al. observed in the main ACRIN 6666 study that the increase in cancer
detection seen in the MRI sub-study was greater than observed by the addition of
annual ultrasound to mammography [11]. However, given the low rate of 8% of
clinically detected interval cancer rate in the main ACRIN 6666 protocol and given
250 M. T. Garcia et al.
the fact that all interval cancers remained node-negative at diagnosis, it is unclear
that the added cost and reduced tolerability of MRI screening are justified in women
at intermediate risk for breast cancer in lieu of supplemental screening with ultra-
sound. Despite its higher sensitivity, the addition of MRI screening instead of ultra-
sound to mammography in broader populations of women at intermediate risk with
dense breasts may not be appropriate, particularly when the current high false-
positive rates, cost, and reduced tolerability of MRI are considered [11].
12.6 Ultrasound
Unlike mammography and ultrasound that are based on anatomy, molecular breast
imaging (MBI) is a physiologic approach to breast cancer detection. MBI detects
additional foci of occult breast cancer in 9.0% of women with newly diagnosed
breast cancer, has a high sensitivity for detecting high-risk lesions, and detects 98%
of invasive breast cancer and 91% of ductal carcinoma in situ [19]. Furthermore, in
surveillance of high-risk women, breast-specific gamma imaging (BSGI)/MBI
detects occult cancer in up to 16.5 per 1000 women. This modality is increasingly
being used to assess response to treatment in women undergoing neoadjuvant che-
motherapy and for adjunct screening in women with dense breasts. It has been
shown to influence surgical management in nearly a quarter of women with newly
diagnosed breast cancer. The Society of Nuclear Imaging has established clinical
indications and the American College of Radiology has established appropriateness
criteria as well as an accreditation program for MBI. A BIRADS-like lexicon for
MBI has also been described.
12.10 Thermography
A number of new and emerging technologies are being developed for breast cancer
screening and diagnosis internationally. Emerging classes of technology promoted
for breast cancer screening include digital infrared thermal imaging (DITI) – ther-
mography. It is a noninvasive imaging method, which neither emits ionizing radia-
tion nor compresses the breast and operates under differing physiological principles.
DITI aims to detect localized increases in skin temperature, which are thought to
occur as a result of increased vascularization, vasodilation, and recruitment of
inflammatory cells to the site of a developing tumor [30]. Localized differences in
skin temperature are captured by infrared cameras, which produce a heat map of the
breast called a thermogram. Early attempts of thermal imaging technology for
breast cancer detection had poor sensitivity (39%) [31]; however, the recent devel-
opment of high-resolution infrared cameras has generated new interest in the use of
DITI as a tool for breast cancer detection [32].
12.11 Elastography
Screening is one of the main factors that contribute to the reduction of mortality
from breast cancer worldwide. Mammographic screening is a well-known estab-
lished method for all women who are between 50 and 70 years of age and can
reduce mortality from breast cancer by about 25 percent [36]. About the value of
breast cancer screening among women who are under 50 years old, there is no con-
sensus [37]. One of the reasons for the lack of agreement is the difficulty in detect-
ing tumors by mammographic screening in younger women, who have denser
breasts than postmenopausal women [38].
Dense breasts are defined by mammographic appearance. The American College
of Radiology’s (ACR) Breast Imaging Reporting and Data System (BI-RADS) clas-
sifies breasts in four categories based on the fibroglandular breast density [39].
Extremely dense breast tissue is a risk factor for breast cancer and limits the
detection of cancer with mammography. However, there is currently little evidence
that the addition of breast ultrasound or MRI would change the mortality rate.
In J-START (Japan Strategic Anti-cancer Randomized Trial), investigators eval-
uated supplemental ultrasonographic breast screening among Japanese women aged
40–49 years old. 58% of the participants had dense breasts. Cancer-detection rates
were 3.3 per 1000 screenings for mammography alone and 5.0 per 1000 screenings
for mammography plus ultrasonography. The addition of ultrasonographic screen-
ing resulted in an interval-cancer rate of 0.5 per 1000 screenings, as compared with
1.0 per 1000 screenings with mammography alone, and an increase in the false-
positive rate from 8.8% to 12.6% [40].
In a diagnostic setting, magnetic resonance imaging (MRI) is a sensitive method
of breast imaging and is not influenced by breast density. The Dense Tissue and
Early Breast Neoplasm Screening (DENSE) trial is a randomized, controlled trial to
study the effect of supplemental MRI on the incidence of interval cancers in women
with extremely dense breast tissue. The trial assigned 40,373 women between the
ages of 50 and 75 years old with extremely dense breast tissue and randomized to a
group that was invited to undergo supplemental MRI or to a group that received
mammography screening only. The primary outcome was the difference between
groups in the incidence of interval cancers during a 2-year screening period. It was
observed a significantly lower interval-cancer rate in the MRI group (2.5 vs. 5.0 per
1000 screenings). Among the women who were invited to undergo MRI, 59% actu-
ally underwent the procedure. Of the 20 interval cancers diagnosed in the MRI-
invitation group, 4 were diagnosed in the women who had undergone MRI and 16 in
those who had not. A limitation of the trial is that it is not large enough to look at the
effect of MRI screening on breast cancer-specific or overall mortality. This outcome
would require a much larger sample size and longer follow-up [41].
254 M. T. Garcia et al.
A woman is considered at high risk of developing breast cancer if she has a lifetime
risk of more than 20–25% of developing breast cancer based on available risk mod-
els, has a BRCA1 or BRCA2 mutations, or was exposed to therapeutic thoracic
radiation (RT) before age 30. This population needs a special screening due to the
high chances of developing breast cancer.
Kuhl et al. investigated in “the EVA Trial” the contribution of clinical breast
examination (CBE), mammography, ultrasound, and quality-assured breast mag-
netic resonance imaging (MRI), used alone or in different combinations, for screen-
ing women at elevated risk for breast cancer. It was a prospective multicenter
observational cohort study and analyzed 687 asymptomatic women at elevated
familial risk (>20% lifetime). Twenty-seven women were diagnosed with breast
cancer: 11 ductal carcinomas in situ (41%) and 16 invasive cancers (59%). All can-
cers were detected during annual screening; no interval cancer occurred; no cancer
was identified during the half-yearly ultrasound. The cancer yield of ultrasound (6.0
of 1000) and mammography (5.4 of 1000) was equivalent; it increased nonsignifi-
cantly (7.7 of 1000) if both methods were combined. Cancer yield achieved by MRI
alone (14.9 of 1000) was significantly higher; it was not significantly improved by
adding mammography (MRI plus mammography: 16.0 of 1000) and did not change
by adding ultrasound (MRI plus ultrasound: 14.9 of 1000). The positive predictive
value was 39% for mammography, 36% for ultrasound, and 48% for MRI [42].
The National Comprehensive Cancer Network (NCCN) recommends that women
who have a lifetime risk ≥20% should have an annual screening mammography
(begin 10 years prior to when the youngest family member was diagnosed with
breast cancer but not prior to age 30) and also recommends annual breast MRI
(begin 10 years prior to when the youngest family member was diagnosed with
breast cancer, but not prior to age 25) [43].
Special screening has been recommended for women who were exposed to ther-
apeutic thoracic radiation, between the ages of 10 and 30 years. The NCCN recom-
mends an annual screening mammography (begin 8 years after RT but not prior to
age 30) and also recommends an annual breast MRI (begin 8 years after RT but not
prior to age 25) [43].
The upper age limit at which breast cancer screening should be applied is not estab-
lished by guidelines. The woman’s overall state of health should be considered in
any decision to undertake or forgo screening.
12 Screening 255
References
1. The Johns Hopkins University, The Johns Hopkins Hospital, and The Johns Hopkins Health
System Corporation, Johns Hopkins Medicine. Available at: https://www.hopkinsmedicine.
org/. Accessed 10 Sept 2020.
2. Lee CS, Bhargavan-Chatfield M, Burnside ES, Nagy P, Sickles EA. The national mammog-
raphy database: preliminary data. Am. J. Roentgenol [Internet]. American Roentgen Ray
Society; 2016 ;206(4):883–90. Available at: https://doi.org/10.2214/AJR.15.14312.
3. Oeffinger KC, Fontham ETH, Etzioni R, Herzig A, Michaelson JS, Shih Y-CT, et al. Breast
cancer screening for women at average risk: 2015 guideline update from the american cancer
society. JAMA [Internet]. 2015;314(15):1599–614. Available at: https://doi.org/10.1001/
jama.2015.12783.
4. Olsen AH, Agbaje OF, Myles JP, Lynge E, Duffy SW. Overdiagnosis, sojourn time, and
sensitivity in the Copenhagen mammography screening program. Breast J [Internet].
2006;12(4):338–42. Available at: http://doi.wiley.com/10.1111/j.1075-122X.2006.00272.x.
5. Jorgensen KJ, Gotzsche PC. Overdiagnosis in publicly organised mammography screen-
ing programmes: systematic review of incidence trends. BMJ [Internet]. 2009;339(jul09
1):b2587–b2587. Available at: https://www.bmj.com/lookup/doi/10.1136/bmj.b2587.
6. Niell BL, Freer PE, Weinfurtner RJ, Arleo EK, Drukteinis JS. Screening for breast cancer.
Radiol Clin N Am. 2017;55(6):1145–62. https://doi.org/10.1016/j.rcl.2017.06.004. PMID:
28991557.
7. Kuhl CK, Schrading S, Leutner CC, Morakkabati-Spitz N, Wardelmann E, Fimmers R, et al.
Mammography, breast ultrasound, and magnetic resonance imaging for surveillance of women
at high familial risk for breast cancer. J Clin Oncol. 2005;23(33):8469–76.
8. Liberman L, Morris EA, Benton CL, Abramson AF, Dershaw DD. Probably benign lesions
at breast magnetic resonance imaging: preliminary experience in high-risk women. Cancer.
2003;98(2):377–88.
9. Kriege M, Brekelmans CTM, Boetes C, Besnard PE, Zonderland HM, Obdeijn IM, et al.
Efficacy of MRI and mammography for breast-cancer screening in women with a familial or
genetic predisposition. N Engl J Med. 2004;351(5):427–37.
10. de Lange SV, Bakker MF, Monninkhof EM, PHM P, de Koekkoek-Doll PK, Mann RM, et al.
Reasons for (non)participation in supplemental population-based MRI breast screening for
women with extremely dense breasts. Clin Radiol. 2018;73(8):759.e1–9.
11. Berg WA, Zhang Z, Lehrer D, Jong RA, Pisano ED, Barr RG, et al. Detection of breast cancer
with addition of annual screening ultrasound or a single screening MRI to mammography in
women with elevated breast cancer risk. JAMA. 2012;307(13):1394–404.
12. Berg WA. Tailored supplemental screening for breast cancer: what now and what next? AJR
Am J Roentgenol. 2009;192(2):390–9.
13. Thigpen D, Kappler A, Brem R. The role of ultrasound in screening dense breasts—a review of
the literature and practical solutions for implementation. Diagnostics [Internet]. 2018;8(1):20.
Available at: http://www.mdpi.com/2075-4418/8/1/20.
14. Rebolj M, Assi V, Brentnall A, Parmar D, Duffy SW. Addition of ultrasound to mammog-
raphy in the case of dense breast tissue: systematic review and meta-analysis. Br J Cancer.
2018;118(12):1559–70.
15. Sood R, Rositch AF, Shakoor D, Ambinder E, Pool K-L, Pollack E, et al. Ultrasound for breast
cancer detection globally: a systematic review and meta-analysis. J Glob Oncol. [Internet].
2019;(5):1–17. Available at: https://ascopubs.org/doi/10.1200/JGO.19.00127.
16. Forouzanfar MH, Foreman KJ, Delossantos AM, Lozano R, Lopez AD, Murray CJL, et al.
Breast and cervical cancer in 187 countries between 1980 and 2010: a systematic analysis.
Lancet (London, England); 2011;378(9801):1461–84.
17. Kelly KM, Richwald GA. Automated whole-breast ultrasound: advancing the performance of
breast cancer screening. Semin. Ultrasound, CT MRI [Internet]. 2011;32(4):273–80. Available
at: https://linkinghub.elsevier.com/retrieve/pii/S088721711100031X.
256 M. T. Garcia et al.
18. Mendelson EB, Berg WA. Training and standards for performance, interpretation, and structured
reporting for supplemental breast cancer screening. Am J Roentgenol. 2015;204(2):265–8.
19. Huppe AI, Mehta AK, Brem RF. Molecular breast imaging: a comprehensive review. Semin
Ultrasound, CT MRI [Internet]. 2018;39(1):60–9. Available at: https://linkinghub.elsevier.
com/retrieve/pii/S0887217117301105.
20. Mathis KL, Hoskin TL, Boughey JC, Crownhart BS, Brandt KR, Vachon CM, et al. Palpable
presentation of breast cancer persists in the era of screening mammography. J Am Coll Surg
[Internet]. 2010;210(3):314–8. Available at: https://linkinghub.elsevier.com/retrieve/pii/
S1072751509016196.
21. Tabár L, Vitak B, Chen HH, Duffy SW, Yen MF, Chiang CF, et al. The Swedish Two-County
Trial twenty years later. Updated mortality results and new insights from long-term follow-up.
Radiol. Clin. North Am. 2000;38(4):625–51.
22. Vanier A, Leux C, Allioux C, Billon-Delacour S, Lombrail P, Molinié F. Are prognostic fac-
tors more favorable for breast cancer detected by organized screening than by opportunis-
tic screening or clinical diagnosis? A study in Loire-Atlantique (France). Cancer Epidemiol.
[Internet]. 2013;37(5):683–7. Available at: https://linkinghub.elsevier.com/retrieve/pii/
S1877782113001070.
23. Moody-Ayers SY, Wells CK, Feinstein AR. Benign tumors and early detection in
mammography-screened patients of a natural cohort with breast cancer. Arch Intern Med.
American Medical Association;. 2000;160(8):1109–15.
24. Baines CJ, Wall C, Risch HA, Kuin JK, Fan IJ. Changes in breast self-examination behav-
ior in a cohort of 8214 women in the Canadian National Breast Screening Study. Cancer.
1986;57(6):1209–16.
25. Kösters JP, Gøtzsche PC. Regular self-examination or clinical examination for early detection
of breast cancer. Cochrane Database Syst Rev. 2003;2003(2):CD003373.
26. Baxter N, Care CTF on PH. Preventive health care, 2001 update: should women be rou-
tinely taught breast self-examination to screen for breast cancer? CMAJ [Internet].
2001;164(13):1837–46. Available at: https://pubmed.ncbi.nlm.nih.gov/11450279.
27. Hackshaw AK, Paul EA. Breast self-examination and death from breast cancer: a meta-
analysis. Br J Cancer. 2003;88(7):1047–53.
28. Brett J, Austoker J, Ong G. Do women who undergo further investigation for breast screening
suffer adverse psychological consequences? A multi-centre follow-up study comparing dif-
ferent breast creening result groups five months after their last breast screening appointment.
J. Public Health (Bangkok) [Internet]. 1998 1;20(4):396–403. Available at: https://academic.
oup.com/jpubhealth/article-lookup/doi/10.1093/oxfordjournals.pubmed.a024793.
29. MacFarlane ME, Sony SD. Women, breast lump discovery, and associated stress. Health Care
Women Int. 1992;13(1):23–32.
30. Anbar M. Clinical thermal imaging today. IEEE Eng Med Biol Mag. 1998;17(4):25–33.
31. Feig SA, Shaber GS, Schwartz GF, Patchefsky A, Libshitz HI, Edeiken J, et al. Thermography,
mammography, and clinical examination in breast cancer screening. Review of 16,000 studies.
Radiology. 1977;122(1):123–7.
32. Jones BF. A reappraisal of the use of infrared thermal image analysis in medicine. IEEE
Trans Med Imaging [Internet]. 1998;17(6):1019–27. Available at: http://ieeexplore.ieee.org/
document/746635/.
33. Egorov V, Kearney T, Pollak SB, Rohatgi C, Sarvazyan N, Airapetian S, et al. Differentiation of
benign and malignant breast lesions by mechanical imaging. Breast Cancer Res Treat [Internet].
2009;118(1):67–80. Available at: http://link.springer.com/10.1007/s10549-009-0369-2.
34. Garra BS. Imaging and estimation of tissue elasticity by ultrasound. Ultrasound Q
LWW. 2007;23(4):255–68.
35. Moon WK, Chang R-F, Chen C-J, Chen D-R, Chen W-L. Solid breast masses: classification
with computer-aided analysis of continuous US images obtained with probe compression.
Radiology. 2005;236(2):458–64.
12 Screening 257
The clinical complaint referred by the patient can be the first indication of the
appearance of breast cancer and should always be valued. The most common clini-
cal complaint is a palpable lump in the breast, but other complaints such as burning
feeling, pain, and changes in color, texture, and appearance of the skin can also be
mentioned. Despite this fact, it should be emphasized that most palpable complaints
correspond to benign lesions in the breasts.
A breast lump is the most frequent presentation of breast cancer, regardless of the
woman’s age. It can be insidious or fast-growing, depending on the characteristics
of the developing tumor. A breast lump is the second most frequent complaint in the
breast surgeon’s office, after breast pain, and is the most frequent complaint pre-
sented by patients with breast cancer.
A study by Barton et al. showed that 16% of the patients in primary healthcare
offices had breast complaints during a 10-year period, with women under 50 years
old having more complaints (twice as much) when compared to the group of women
over 50 years old, and the diagnosis of cancer was made in 23 of the 372 women
who presented symptoms (6.2%) [1].
Another palpable complaint that can be a symptom of cancer is breast thicken-
ing. This one differs from a mass as it does not have a three-dimensional configura-
tion and presents itself differently from surrounding breast tissues on palpation and
does not have a similar area in the contralateral breast, being asymmetrical.
Thickening is associated with breast cancer about 5% of the time.
The masses and thickenings associated with breast cancer are ill-defined, are
hardened, and may be attached to the skin or muscular plane and may be associated
with changes in the skin and papillae, such as retraction.
Because many breast masses may not exhibit distinctive physical findings, imag-
ing evaluation is necessary in almost all cases to characterize the palpable lesion.
Any woman presenting with a palpable lesion should have a thorough clinical breast
examination, usually by the referring clinician or by a clinical breast specialist, but
the radiologist must also be able to establish a concordance between an imaging
finding and a clinically detected mass [2].
When a suspicious finding is identified, an image-guided biopsy is indicated. It
is preferable to do imaging examinations before biopsy, as changes related to the
biopsy may confuse, alter, obscure, and/or limit image interpretation. The negative
predictive value of mammography with ultrasound (US) in the context of a palpable
mass ranges from 97.4% to 100% [2]. Nevertheless, a negative imaging evaluation
should never overrule a strongly suspicious physical examination finding or vice
versa. Any highly suspicious breast mass detected by imaging or palpation should
undergo biopsy unless there are exceptional clinical circumstances such as the sig-
nificant comorbid conditions presented by the patient.
Diagnostic mammography is indicated for women aged 40 years and over who
present a palpable lump. In several series evaluating palpable breast abnormalities,
the sensitivity of mammography alone was 86–91%, depending on the breast den-
sity (Fig. 13.1). If a clearly benign correlate for a palpable finding (oil cyst,
a b c d
Fig. 13.1 Breast composition on mammography – comparison of breast density: (a) Almost
entirely fatty. (b) Scattered areas of fibroglandular density. (c) Heterogeneously dense. (d)
Extremely dense
13 Diagnostic 261
a b
Fig. 13.3 Mammography and magnetic resonance imaging (MRI). (a) Mammography was nega-
tive for suspicious findings. (b) MRI (T1W post-contrast image) showed an irregular mass with
homogeneous enhancement in the left breast. (c) Second-look ultrasound showed an irregular
hypoechogenic mass with indistinct margins. Histologically proven grade 3 ductal carcinoma in
situ (DCIS)
also assess superficial cutaneous and subcutaneous lesions with the use of high-
frequency transducers and appropriate techniques.
In addition, younger women also tend to have relatively denser breast tissue,
which is associated with decreased mammographic sensitivity [6]. However, in the
event of a suspicious finding, mammography or digital breast tomosynthesis (DBT)
is warranted even in younger women to better delineate disease and identify features
of malignancy that may be seen on mammography or DBT alone.
If a palpable lump can be definitively characterized as benign on US (e.g., simple
cyst, benign lymph node, duct ectasia, lipoma), clinical follow-up is the appropriate
management, and imaging follow-up or tissue sampling is not indicated.
To improve the sensitivity and specificity, the combined use of diagnostic mam-
mography and ultrasonography in palpable lesions has been recommended [2]. The
combination of these two imaging modalities increases true positive rates and
13 Diagnostic 263
a b
Fig. 13.4 A 42-year-old patient screening mammography with tomosynthesis. (a) Conventional
2D mammography was negative for suspicious findings. (b) Tomosynthesis showed an area of
architectural distortion in the outer upper quadrant of the left breast (circle). Histologically proven
grade 2 invasive lobular carcinoma
signs and symptoms and in women recalled from screening has also been shown to
be equivalent to or better than supplemental diagnostic mammographic views in
several studies [11, 14]. DBT can be used with or without spot compression views
in the diagnostic context.
Interpretation time for DBT images is greater than for standard mammography
[14, 15]. Additionally, the dose is increased if standard 2D images are obtained in
addition to DBT images. However, synthesized reconstructed images (a virtual pla-
nar image created from the tomographic data set) may replace the need for a 2D
correlate view, and current data suggest that these synthetic images perform as well
as standard full-field digital images [14, 16].
13 Diagnostic 265
Since 1307 breast lesions have occurred similar to what we now recognize as Paget’s
breast disease, but which were only described as such in 1874 by Sir James Paget.
Clinically, it corresponds to a lesion in the nipple-areola complex (NAC) repre-
sented by eczema, erythema, pruritus, and ulceration. Unlike papillary lesion due to
invasion of the underlying tumor, Paget’s breast disease is characterized by the cell
described as large, pale-staining cells with round or oval nuclei and prominent
nucleoli. It is considered a rare pathology, comprising 1–4% of cases of malignant
breast disease [18].
266 K. B. Carreiro et al.
a b
Fig. 13.5 A 63-year-old patient with Paget’s disease of the right breast. (a) The mammography
showing a diffuse thickening of the nipple-areola complex and fine pleomorphic calcifications in
segmental distribution in the breast extending to the nipple. Histologically proven grade 3 DCIS.
(b) Mammmographic magnification view showing pleomorphic calcifications in segmental
distribution
of 8), and all patients with suspected lesions had a histological confirmation of an
additional malignancy. Among the patients submitted to MRI, 50% had biopsy-
proven suspicious lesions. The remaining four patients had confirmation of malig-
nancy only in the final histological result from surgical specimen analysis. This
work was able to demonstrate the possibility of an adequate surgical scheduling.
The surgical procedures were divided into 21 mastectomies and 17 conservative
surgeries, of which only 4 were converted to mastectomy after the final result. 59%
of patients had disease only in the central breast sector [23].
Although Paget’s disease affects only NAC and is almost exclusively in situ, it
can present invasion of the basement membrane and lymph node involvement, influ-
encing directly the proposed treatment. In addition, associated malignant lesions are
often associated with Paget’s disease, and proper assessment of the rest of the breast
tissue is essential. Patients with suspected lesions on clinical examination are usu-
ally diagnosed with associated malignancy. Even those patients with no palpable
changes should undergo mammography and eventually MRI, to define the extent of
disease and look for additional lesions that need attention during treatment.
268 K. B. Carreiro et al.
Axillary lymph nodes receive drainage from the arm, chest wall, and breast, that is,
any benign or malignant pathology that occurs in these regions may be associated
with local lymphadenopathy. Some neoplastic diseases may also be associated with
the involvement of axillary lymph nodes, even from distant sites, such as primary
tumors of the pancreas, ovary, and bladder, in addition to lymphoma itself. We call
it occult breast carcinoma when there is a metastatic axillary lesion with immuno-
histochemistry pointing to the breast as the primary tumor, with no changes in clini-
cal or radiological examination (Fig. 13.6). The most commonly used radiological
evaluation is mammography and ultrasound, but nowadays MRI shows better per-
formance in detecting the primary tumor.
The incidence of occult breast carcinoma was around 0.3–1.0% between the
1950s and 1980s, as shown by the survey by Ge et al. in which the main radiological
exam for breast investigation was mammography [24]. However, in a 2017 publica-
tion, Hessler et al. published their own statistics based on the National Cancer
Database (NCDB). The NCDB is a clinical oncology database sponsored by the
a b
Fig. 13.6 Occult breast carcinoma in a 34-year-old patient with BRCA1 mutation presenting with
palpable lumps in the left axilla. (a) Mammogram shows multiple enlarged axillary lymph nodes.
No suspicious findings in the breast. (b) MRI (T1W post-contrast image with subtraction) of the
same patient shows multiple round lymph nodes with obliteration of the fatty hilum and heteroge-
neous enhancement. No suspicious enhancements were found in the breast. Histology of axillary
lymph nodes: metastatic triple-negative breast carcinoma
13 Diagnostic 269
American College of Surgeons and the American Cancer Society. In this study, they
found an incidence of 0.1%, with an analysis carried out between the years 2004
and 2013, with 1853 patients [25]. With the evolution of imaging exams and
improved resolution of the images generated by the mammographic and ultrasound
devices, in addition to the beginning of the MRI exams, it was possible to identify
the primary breast lesions that were not possible before.
Vlastos et al. published a survey carried out at MD Anderson of patients treated
at these sites with a metastatic axillary disease, with no specific primary site. Of the
479 identified patients, 45 were considered to be primarily of breast origin. The
inclusion criteria were lymph node biopsy with histological diagnosis of adenocar-
cinoma or undifferentiated or unclassified carcinoma. All patients underwent surgi-
cal treatment, with 13 having mastectomies and 32 breast-conserving surgeries. The
proposed conservative surgery was the resection of the superolateral quadrant of the
breast, which is a denser region with the highest prevalence of breast tumors, com-
prising about 50%. It was possible to identify among the patients who underwent
mastectomy 1 case of invasive tumor and 1 case of Paget’s disease. Among the
patients who underwent conservative surgery, 10 cases of malignant disease were
identified, not stratified as invasive or in situ [26].
Within breast assessment, one factor that makes it difficult to diagnose breast
lesions is the localization in the axillary tail or accessory mammary gland. However,
MRI sensitivity can reach 95%, after negative mammography, ultrasound, and clini-
cal examination, and 71% specificity, as demonstrated in a 2011 study [27].
Currently there are studies that question the existence of ectopic breast tissue in
axillary lymph nodes, which would characterize occult carcinoma in this way, but
with the possibility of definition only after surgery for histological confirma-
tion [28].
Axillary metastasis can arise from the breast or any other extra-mammary site,
requiring histological and immunohistochemical assessment to define the primary
lesion. When compatible with breast origin without clinical evidence on physical
examination, complementary radiological examinations are essential to search for
the primary lesion. The initial workup consists of conventional imaging evaluation
with mammography and ultrasound, and complementation with MRI when conven-
tional imaging is negative. There are studies that include positron emission tomog-
raphy (PET) scan, but this is not part of the initial recommendation for investigation
yet [29].
The aim of mammographic screening is to reduce mortality rates from breast can-
cer through early detection compared to cancers diagnosed clinically at a later
stage. Despite ongoing controversy, the effectiveness of mammographic screening
270 K. B. Carreiro et al.
and the importance of diagnosing breast cancers at an early stage have been empha-
sized [30]. It is known that current screening with 2D digital mammography
(2DDM) detects some cancers that might not cause symptoms during the woman’s
natural life. This has been termed “overdiagnosis,” and the frequency of detecting
such cases has been discussed in the independent review of breast screening.
Screening with DBT will detect even more breast cancers at an earlier stage in
some cases [5].
The accuracy of 2DDM screening is limited because of the effect of overlapping
of normal breast structures and abnormal features on a two-dimensional image. The
mammographic signs of breast cancer may be partially or completely obscured,
particularly in women with a dense glandular parenchymal pattern on mammogra-
phy, leading to delays in the diagnosis of breast cancer.
DBT demonstrates mammographic characteristics of soft-tissue lesions better
than 2DDM, which leads to improved diagnostic accuracy, more accurate workup
of soft-tissue lesions, and better local staging, with performance shown to be even
more beneficial in women with dense breasts. There is improved interpretive perfor-
mance of DBT in the detectability of invasive lobular cancers that present as distor-
tions and spiculations as these lesions are more conspicuous on DBT.
Conversely, well-defined margins of benign lesions are more clearly demon-
strated, potentially eliminating the need for biopsy and reducing recall rates. The
better visibility of the margins of soft-tissue lesions improves the ability of the radi-
ologist to distinguish between benign and malignant masses [5, 8, 31]. Circumscribed
masses may also show a halo sign on DBT. For soft-tissue lesions, DBT improves
the classification of the mammographic features seen on 2DDM and the accuracy of
predicting malignancy.
In the largest retrospective study involving 7060 subjects, the TOMMY trial, the
addition of DBT improved sensitivity in women with dense breasts (93% versus
86% for DBT versus 2DDM, in women with breast density of 50% or more;
p1/40.03). Specificity was significantly improved in all subgroups (57% for 2DDM,
70% for 2DDM + DBT; p < 0.001 in both cases) [9].
Nipple discharge corresponds to the majority of complaints from patients who seek
breast surgeons, with an incidence of 2.5–5% [32–34]. Nipple discharge can be
spontaneous or provoked, uniductal or multiductal, unilateral, or bilateral and can
have several presentations: serous, bloody, milky, greenish, or transparent. The flow
can be secondary to organic or structural causes. Among the organic causes, we can
highlight the postpartum pregnancy cycle, metabolic changes, and the use of medi-
cations. Normally, in these situations, the flow is multiductal and bilateral and with
a dairy aspect. In the case of structural changes, the flow most commonly presents
itself as unilateral, with uni or multiductal secretion output, and can be spontaneous
or provoked [35]. Among the most frequent causes of nipple discharge due to ana-
tomical changes are papilloma (35–48%), carcinoma (5–21%), and ductal ectasia or
13 Diagnostic 271
fibrocystic changes (3–50%), but it varies greatly between studies according to the
type of complaint and clinical and radiological alteration included [32].
The initial assessment of patients with complaints of nipple discharge must con-
tain clinical history and physical examination, in order to better characterize the
flow and identify possible tangible changes. In the case of palpable complaints, it is
possible to perform an examination directed to the site, through ultrasound or mam-
mography. In cases of absence of palpable lesions, there is the benefit of evaluation
by means of MRI [36]. Ductography is an evaluation of the affected duct through
the injection of iodinated contrast and, afterwards, mammography with subareolar
magnification to view possible defects of the ductal tree (Fig. 13.7). It is able to
identify single or multiple lesions, in addition to peripheral lesions when the affected
duct is well identified [33].
Adepoju et al. evaluated 168 cases of patients with spontaneous and uniductal
papillary flow, of which 85% were bloody. Mandatory mammography, physical
examination, and evaluation by complementary ductography and ultrasound, if the
team considered it opportune, were performed before ductal resection to evaluate
the etiology and its imaging correlation. Malignant lesions were diagnosed in 12%
of cases and high-risk lesions in 11%. The mean age of patients with benign changes
was 51 years old, with high-risk lesions 57 years old and malignant lesions 69 years.
The sensitivity and specificity of ultrasound and mammography ranged from 10%
to 35% and 68% to 94%, respectively, in high-risk and malignant lesions.
Ductography, when performed, showed sensitivity and specificity of 75% and
49–53%, respectively, for high-risk and malignant lesions, but showed some limita-
tions as scarcity of places and professionals able to perform the procedure, in
Fig. 13.7 Ductography in a 72-year-old patient with bloody nipple discharge in the left breast
showing a filling defect (circle). Histologically proven intraductal papilloma
272 K. B. Carreiro et al.
addition to anatomical variations that may make it impossible, such as ductal atro-
phy secondary to age [37].
Currently, the use of ductography has been questioned due to the discomfort
potentially caused by duct catheterization and contrast injection, in addition to the
need for a trained staff for the exam. Consequently, Baydoun et al. published an
article evaluating 94 patients retrospectively, who underwent ductography between
January 2005 and October 2010. The indications for the exam consisted of a woman
with a negative image exam but with a persistent or bloody discharge in a 1-month
clinical follow-up, and the biopsy result of an identified lesion was incongruous
with the clinic or in a presurgical evaluation. In case the ductography was not per-
formed properly or if it was negative, the patient was referred for contrast MRI
exam (a total of 12 exams). Seven malignant lesions were identified (5 DCIS and 2
Invasive cancers) and 3 premalignant lesions (atypical hyperplasia). Comparing the
exams, the sensitivity of mammography, ultrasound, and ductography were, respec-
tively, 13%, 73%, and 76%. The low number of MRI performed in this study pre-
cluded the comparison with other modalities, but its sensibility appeared to be
comparable to ductography (Fig. 13.8) [38].
Bahl et al. in a retrospective study evaluated a group of 118 women with papil-
lary flow complaints with at least one of these characteristics: unilateral bloody,
serous, or spontaneous. In these women, an evaluation with contrast MRI was per-
formed after a negative mammography and ultrasound. Four cases of DCIS and two
cases of invasive cancer not previously identified were diagnosed but with two false
negatives cases (two cases of BIRADS 3 biopsied during the follow-up) [39].
Sanders et al. in a retrospective study evaluated 200 women with bloody nipple
discharge with negative mammographic and ultrasound evaluation who would
undergo duct resection. A total of 115 patients were submitted directly to surgery,
and 85 underwent contrast MRI before. In both groups, the incidence of malignant
breast cancer was 7%, and in the MRI group, only one case of false negative was
identified (the final histological result of low-grade ductal carcinoma), but three
malignant lesions were identified in the contralateral breast. The main causes for
a b
Fig. 13.8 A 46-year-old patient with bloody nipple discharge. (a) MRI (post-contrast subtracted
image) shows a linear ductal enhancement in the left breast. (b) Second-look ultrasound depicted
an intraductal mass with vascular flow on Doppler imaging. Biopsy proven Intraductal papilloma
13 Diagnostic 273
If a suspicious mass has been identified on mammography, DBT, MRI, or US, tissue
sampling is warranted except in rare circumstances (e.g., if the patient has comor-
bidities that would contraindicate biopsy). If a lesion is seen equally well on mam-
mography and US, US guidance is preferred because of patient comfort, efficiency,
economy, absence of ionizing radiation, and sampling accuracy due to real-time
visualization of the needle within the lesion.
Although some practices demonstrate very good results using fine-needle aspiration
(FNA) biopsy as the first means of diagnostic evaluation of a palpable breast mass,
especially if a cytopathologist is available to interpret results in real time [16, 40],
larger series demonstrate that core biopsy is superior to FNA in terms of sensitivity,
specificity, and correct histological grading of palpable masses [41]. Core biopsy is
therefore recommended in most cases. Either stereotactic or DBT (x-ray) or US
guidance can be used for core biopsy, especially if the mass is vaguely palpable,
small, deep, or mobile or if there are multiple lesions [40]. Even when a lesion is
clinically palpable, image-guided biopsy has advantages over tissue sampling by
palpation, allowing confirmation of biopsy accuracy and the possibility to place a
post-biopsy marker clip.
274 K. B. Carreiro et al.
palpable lesions than for non-palpable lesions. The study suggests that both FNA
and CNB have good clinical performance. In similar circumstances, the sensitivity
of CNB is better than that of FNA, while their specificities are similar. FNA could
be still considered the first choice to evaluate suspicious non-palpable breast
lesions [42].
Under stereotactic guidance, vacuum-assisted biopsy (VAB) has proven to be
more effective than CNB and less invasive than surgical excision of breast lesions.
VAB is being increasingly utilized as an alternative to stereotactic-guided CNB. The
advantages include biopsy of lesions only visible on DBT/2DDM, low-density
lesions or lesions only identifiable on one view, fewer exposures required, better
technical accuracy in localizing masses, distortions, and asymmetrical densities.
Several studies have shown superior accuracy in lesion targeting and a lower com-
plication rate [5, 44].
The result of the widespread use of MRI in breast diagnostics is an increase in inci-
dental findings. Because of the high sensitivity and lower specificity of breast MRI,
it is imperative that these findings be histologically assessed before any surgical
intervention [41, 45].
The first step to investigate any MRI abnormalities should be a second-look tar-
geted US. The efficacy of the second-look US is reported between 23% and 71%,
with a malignancy detection rate of 15–56% [44, 45]. Larger enhancing masses and
BIRADS 5 (highly suspicious for malignancy) lesions are more readily identified
on a second-look targeted US, with a detection rate estimated between 25% and
62%; however, certain types of MRI lesions, such as non-mass enhancement or
small foci less than10 mm, are less likely to have a US correlate, with a detection
rate ranging from 11% to 42%. In a positive sonographic correlation, it is better to
perform intervention under US guidance instead of MRI, because it is simpler,
faster, and more cost-effective than MRI.
The possible MRI-guided interventions are wire localization of breast lesions
followed by a surgical biopsy, CNB, and VAB [44].
Wire localization was one of the earliest procedures to be attempted under MR
guidance. The technique has been performed freehand and with guidance systems
in open and closed magnets with patients in supine, prone, and prone oblique posi-
tions. The relatively large surgical specimen reduces the chance of sampling error
and compensates for some inaccuracy of needle placement. In a series of 101
patients, Eby et al. reported placement of the needle tip within 10 mm of the target
in 53% of cases and between 11 and 20 mm in 46% of cases with successful exci-
sion in 100% [45].
CNB under MRI guidance also requires an extremely accurate targeting tech-
nique. Postfire sequences should demonstrate the needle within the target. Therefore,
the patient must be scanned with the device in place. To achieve this goal, MRI-
compatible CNB devices must be composed of nonferrous alloys including various
276 K. B. Carreiro et al.
a b
Fig. 13.9 MRI-guided biopsy in a 39-year-old patient with Li-Fraumeni syndrome. (a) Pre-biopsy
image (T1W post-contrast image) shows an irregular mass with heterogeneous enhancement in the
left breast. (b) Post-biopsy image (T1W post-contrast) shows a dark linear structure extending
from the biopsy site to the lateral skin, corresponding to the artifact of the biopsy needle.
Susceptibility artifacts from gas bubbles and tissue marker are also seen on the biopsy site.
Histologic result: grade 1 tubulo lobular carcinoma
amounts of titanium and nickel. As a result of the advances and refinements, MRI-
guided CNB is considered both safe and accurate with published reports of a high
rate of technical success (95–100%) and cancer yields (20–38%) comparable to the
accepted rates reported in mammographic- and sonographic-guided procedures.
The Mammotome (Ethicon Endo-Surgery) is an 11-gauge MRI-compatible VAB
system. It was the first system to be developed and subsequently proved that VAB is
possible under MRI guidance (Fig. 13.9). Clinical studies in Europe have demon-
strated successful VAB in 98% of 341 lesions and have identified important barriers
to the more widespread use of this technology, including needle artifact, tissue dis-
placement during probe insertion, and washout of contrast enhancement during the
procedure. These challenges limited the application of this early technology to
lesions larger than 10 mm in diameter.
DBT-guided breast biopsy has been developed both for use with standard DBT
mammographic equipment and dedicated prone table apparatus, often combined
with stereotaxis. It is essential for lesions seen only with DBT and may be used as
an alternative to stereotactic-guided VAB for both sonographically occult and soft-
tissue lesions with micro-calcifications [44].
References
1. Barton MB, Elmore JG, Fletcher SW. Breast symptoms among women enrolled in a
health maintenance organization: frequency, evaluation, and outcome. Ann Intern Med.
1999;130(8):651–7. https://doi.org/10.7326/0003-4819-130-8-199904200-00005.
13 Diagnostic 277
2. Moy L, et al. ACR appropriateness criteria® palpable breast masses. J Am Coll Radiol.
2017;(5S):S203–S224. https://doi.org/10.1016/j.jacr.2017.02.033.
3. Ciatto S, Houssami N. Breast imaging and needle biopsy in women with clinically evi-
dent breast cancer: does combined imaging change overall diagnostic sensitivity? Breast.
2007;(4):382–6. https://doi.org/10.1016/j.breast.2007.01.007.
4. Murphy IG, et al. Analysis of patients with false negative mammography and symptomatic
breast carcinoma. J Surg Oncol. 2007;16(4):382–6. https://doi.org/10.1002/jso.20801.
5. Michell MJ, Batohi B. Role of tomosynthesis in breast imaging going forward. Clin Radiol.
2018;73(4):358–71. https://doi.org/10.1016/j.crad.2018.01.001.
6. Durfee SM, et al. Sonographic evaluation of clinically palpable breast cancers invisible on
mammography. Breast Journal. 2000;6(4):247–51. https://doi.org/10.1046/j.1524-4741.2000
.99111.x.
7. Gilbert FJ, Pinker-Domenig K. Diagnosis and staging of breast Cancer: when and how to
use mammography, tomosynthesis, ultrasound, contrast-enhanced mmmography, and
magnetic resonance imaging; 2019 Feb 20. In: Hodler J, Kubik-Huch RA, von Schulthess
GK, editors. Diseases of the Chest, Breast, Heart and Vessels 2019–2022: Diagnostic and
Interventional Imaging [Internet]. Cham (CH): Springer; 2019. Chapter 13. https://doi.
org/10.1007/978-3-030-11149-6_13.
8. Greenberg JS, et al. Clinical performance metrics of 3D digital breast tomosynthesis com-
pared with 2D digital mammography for breast cancer screening in community practice. Am J
Roentgenol. 2014;203(3):687–93. https://doi.org/10.2214/AJR.14.12642.
9. Gilbert FJ, et al. The TOMMY trial: a comparison of TOMosynthesis with digital mammog-
raphy in the UK NHS breast screening programme – a multicentre retrospective reading study
comparing the diagnostic performance of digital breast tomosynthesis and digital mammogra-
phy with. Health Technol Assess. 2015;19(4):i–xxv, 1–136. https://doi.org/10.3310/hta19040.
10. Vedantham S, et al. Digital breast tomosynthesis: state of the art1. Radiology.
2015;277(3):663–84. https://doi.org/10.1148/radiol.2015141303.
11. Shetty MK, Shah YP, Sharman RS. Prospective evaluation of the value of combined mam-
mographic and sonographic assessment in patients with palpable abnormalities of the breast. J
Ultrasound Med. 2003;22(3):263–8; quiz 269–70. https://doi.org/10.7863/jum.2003.22.3.263.
12. Lehman CD, Lee AY, Lee CI. Imaging management of palpable breast abnormalities. Am J
Roentgenol. 2014;203(5):1142–53. https://doi.org/10.2214/AJR.14.12725.
13. Dennis MA, et al. Breast biopsy avoidance: the value of normal mammograms and normal
sonograms in the setting of a palpable lump. Radiology. 2001;219(1):186–91 https://doi.
org/10.1148/radiology.219.1.r01ap35186.
14. Bernardi D, et al. Application of breast tomosynthesis in screening: incremental effect on
mammography acquisition and reading time. Br J Radiol. 2012;85(1020):e1174–8. https://doi.
org/10.1259/bjr/19385909.
15. Dang PA, et al. Addition of tomosynthesis to conventional digital mammography: effect on
image interpretation time of screening examinations. Radiology. 2014;270:49–56. https://doi.
org/10.1148/radiol.13130765.
16. Liew PL, et al. Rapid staining and immediate interpretation of fine-needle aspiration cytology
for palpable breast lesions: diagnostic accuracy, mammographic, ultrasonographic and histo-
pathologic correlations. Acta Cytol. 2011;55(1):30–7. https://doi.org/10.1159/000320869.
17. Spick C, et al. Breast MRI used as a problem-solving tool reliably excludes malignancy. Eur J
Radiol. 2015;84(1):61–4. https://doi.org/10.1016/j.ejrad.2014.10.005.
18. Sven J, Kister MD, Cushman D, Haagensen M. Paget’s disease of the breast. Am J Surg.
1970;119(5):606–9. https://doi.org/10.1016/S1776-9817(06)73026-7.
19. Fu W, Mittel VK, Young SC. Paget disease of the breast: analysis of 41 patients. Am J Clin
Oncol. 2001;24(4):397–400. https://doi.org/10.1097/00000421-200108000-00019.
20. Lee HW, et al. Invasive Paget disease of the breast: 20 Years of experience at a single
institution. Human Pathol. Elsevier Inc. 2014;45(12):2480–7. https://doi.org/10.1016/j.
humpath.2014.08.015.
278 K. B. Carreiro et al.
21. Lim HS, et al. Paget disease of the breast: mammographic, US, and MR imaging findings
with pathologic corelation. Radiographics. 2011;31(7):1973–88. https://doi.org/10.1148/
rg.317115070.
22. Zakaria S, et al. Paget’s disease of the breast: accuracy of preoperative assessment. Breast
Cancer Res Treat. 2007;102(2):137–42. https://doi.org/10.1007/s10549-006-9329-2.
23. Morrogh M, et al. MRI identifies otherwise occult disease in select patients with Paget
disease of the nipple. J Am Coll Surg. 2008;206(2):316–21. https://doi.org/10.1016/j.
jamcollsurg.2007.07.046.
24. Ge LP, et al. Clinicopathological characteristics and treatment outcomes of occult breast
cancer: a SEER population-based study. Cancer Manag Res. 2018;10:4381–91. https://doi.
org/10.2147/CMAR.S169019.
25. Hessler LK, et al. Factors influencing management and outcome in patients with occult breast
cancer with axillary lymph node involvement: analysis of the national cancer database. Ann
Surg Oncol. Springer International Publishing,. 2017;24(10):2907–14. https://doi.org/10.1245/
s10434-017-5928-x.
26. Vlastos G, et al. Feasibility of breast preservation in the treatment of occult primary carci-
noma presenting with axillary metastases. Ann Surg Oncol. 2001;8(5):425–31. https://doi.
org/10.1007/s10434-001-0425-6.
27. Lu H, et al. Breast magnetic resonance imaging in patients with occult breast carci-
noma: evaluation on feasibility and correlation with histopathological findings. Chin Med
J. 2011;124(12):1790–5. https://doi.org/10.3760/cma.j.issn.0366-6999.
28. Terada M, et al. Occult breast cancer may originate from ectopic breast tissue present in
axillary lymph nodes. Breast Cancer Res Treat. Springer US,. 2018;172(1):1–7. https://doi.
org/10.1007/s10549-018-4898-4.
29. Wong YP, et al. Occult primary breast carcinoma presented as an axillary mass: a diagnostic
challenge. Malays J Pathol. 2020;42(1):151–5.
30. Brandt KR, et al. Can digital breast tomosynthesis replace conventional diagnostic mammog-
raphy views for screening recalls without calcifications? A comparison study in a simulated
clinical setting. Am J Roentgenol. 2013;200(2):291–8. https://doi.org/10.2214/AJR.12.8881.
31. Gilbert FJ, Tucker L, Young KC. Digital breast tomosynthesis (DBT): a review of the e vidence
for use as a screening tool. Clin Radiol. 2016;71(2):141–50. https://doi.org/10.1016/j.
crad.2015.11.008.
32. Murad TM, Contesso G, Mouriesse H. Nipple discharge from the breast. Ann Surg.
1982;195(3):259–64. https://doi.org/10.1097/00000658-198203000-00003.
33. Dawes LG, et al. Ductography for nipple discharge: no replacement for ductal excision.
Surgery. 1998;124(4):685–91. https://doi.org/10.1067/msy.1998.91362.
34. King TA, et al. A simple approach to nipple discharge. Am Surg. 2000;66(10):960–5; discus-
sion 965–6.
35. Montroni I, et al. Nipple discharge: is its significance as a risk factor for breast cancer fully
understood? Observational study including 915 consecutive patients who underwent selec-
tive duct excision. Breast Cancer Res Treat. 2010;123(3):895–900. https://doi.org/10.1007/
s10549-010-0815-1.
36. Morrogh M, et al. Lessons learned from 416 cases of nipple discharge of the breast. Am J Surg.
Elsevier Inc. 2010;200(1):73–80. https://doi.org/10.1016/j.amjsurg.2009.06.021.
37. Adepoju LJ, et al. The value of clinical characteristics and breast-imaging studies in predicting
a histopathologic diagnosis of cancer or high-risk lesion in patients with spontaneous nipple
discharge. Am J Surg. 2005;190(4):644–6. https://doi.org/10.1016/j.amjsurg.2005.06.032.
38. Bahl M, Gadd MA, Lehman CD. Diagnostic utility of MRI after negative or inconclu-
sive mammography for the evaluation of pathologic nipple discharge. Am J Roentgenol.
2017;209(6):1404–10. https://doi.org/10.2214/AJR.17.18139.
39. Sanders LM, Daigle M. The rightful role of MRI after negative conventional imaging in the
management of bloody nipple discharge. Breast J. 2016;22(2):209–12. https://doi.org/10.1111/
tbj.12551.
13 Diagnostic 279
40. Rosa M, Mohammadi A, Masood S. The value of fine needle aspiration biopsy in the diagnosis
and prognostic assessment of palpable breast lesions. Diagn Cytopathol. 2012;40(1):26–34.
https://doi.org/10.1002/dc.21497.
41. Garg S, et al. A comparative analysis of core needle biopsy and fine-needle aspiration cytology
in the evaluation of palpable and mammographically detected suspicious breast lesions. Diagn
Cytopathol. 2007;35(11):681–9. https://doi.org/10.1002/dc.20721.
42. Wang M, et al. A sensitivity and specificity comparison of fine needle aspiration cytology and
core needle biopsy in evaluation of suspicious breast lesions: a systematic review and meta-
analysis. Breast. 2017;31:157–66. https://doi.org/10.1016/j.breast.2016.11.009.
43. Homesh NA, Issa MA, El-Sofiani HA. The diagnostic accuracy of fine needle aspiration cytol-
ogy versus core needle biopsy for palpable breast lump(s). Saudi Med J. 2005;26(1):42–6.
44. Imschweiler T, et al. MRI-guided vacuum-assisted breast biopsy: comparison with stereotacti-
cally guided and ultrasoundguided techniques. Eur Radiol. 2014;24(1):128–35. https://doi.
org/10.1007/s00330-013-2989-5.
45. Eby PR, Lehman C. MRI-guided breast interventions. In: Seminars in ultrasound, CT and
MRI; 2006;27(4):339–50. https://doi.org/10.1053/j.sult.2006.05.008.
Chapter 14
Preoperative (Breast)
Jonathan Yugo Maesaka , Yedda Nunes Reis , and Jose Roberto Filassi
Newly diagnosed patients with breast cancer should be evaluated clinically and with
imaging exams to guide locoregional surgical treatment (breast and axilla). In this
context, understanding the locoregional extent of the disease assists the physician to
make the best decision for the treatment and will enable the choice of the most
appropriate surgical option, which can be a breast conservative surgery (quadrantec-
tomy/lumpectomy), or a mastectomy; if there is a need for immediate breast recon-
struction or even if there is an ipsilateral or contralateral synchronic disease that
needs to be treated concomitantly or for contralateral breast symmetrization.
When assessing a patient who is a candidate for conservative surgery, the attending
physician uses clinical and imaging parameters for safe decision-making. Important
considerations are made about the tumor size, as well as the extent of the disease, the
presence of multifocality/multicentricity, and involvement of adjacent structures.
Preoperative imaging exams will assist in understanding the true size of the
tumor, so that adequate resection is programmed. Currently, “no ink on tumor” is
the goal for patients with invasive breast disease undergoing conservative surgery,
and in patients with pure ductal carcinoma in situ (DCIS), the recommended margin
is 2 mm. Thus, the surgical approach must be personalized, and there is no longer
contraindication for conservative surgery for multicentric tumors or with extensive
DCIS components. Conservative surgery can be offered as long as the entire tumor
is resected and the aesthetic result is adequate.
Special attention is required for patients with invasive lobular carcinoma (ILC)
subtype. Invasive lobular carcinoma is the second most common histological type of
breast cancer, representing 5–15% of cases, and due to its specific characteristics of
involvement of the breast lobes, without significant inflammatory reaction, it can
bring challenges in the clinical and imaging evaluation. Clinically, in most cases, it
is not possible to identify well-defined palpable masses. This presentation is due to
a slight change in the consistency of the breast. Invasion by malignant cells some-
times occurs insidiously, resulting in little or no desmoplasia. Masses and microcal-
cifications in lobular subtypes are incommon. In addition, lobular carcinoma has a
tendency to multifocal proliferation, with greater challenge in safe delimitation of
the disease. As we will mention further in this chapter, in view of this histological
type, an individualized imaging evaluation must be performed.
14.1.1.1 Mammography
Mammography (MG) is the gold standard imaging method for preoperative breast
assessment and should be performed by all patients diagnosed with breast cancer.
As it is the main test used in screening, the diagnosis is often made based on the
findings of this test.
In addition, mammography is widely used in patients scheduled for non-
oncological aesthetic breast surgery. At this point, it is worth mentioning the
Choosing Wisely statement of the American Society of Plastic Surgeons [1] that
recommends avoiding the routine use of mammograms before breast surgery,
reserving its use for patients with indications for screening. A study carried out by
Sears et al. identified that mammography in patients under 40 years of age at preop-
erative mammoplasty (without other clinical indication) increased the rates of more
exams (such as magnetic resonance imaging (MRI)) and more biopsies, without
objective evidence of their benefit [2].
A characteristic to be mentioned in the use of preoperative mammography is the
interobserver reproducibility. A study carried out by Kim et al. showed an almost
perfect agreement of mammography between different observers, compared to oth-
ers methods such as ultrasound (US) and MRI, when looking for prediction charac-
teristics of an extensive intraductal component [3]. As it is a static method and
easier to interpret, mammography is used by surgeons in the operating room to plan
incisions and tumor location and assess the extent of the disease.
Berg et al. carried out in 2004 a study of the accuracy of MG, US, and MRI tech-
niques in 111 patients [4]. MRI showed greater sensitivity when compared to
MG. An interesting fact is that adding MRI led to a 12% increase in the diagnosis
of new lesions, when compared to the use of MG + US. At the same time, in this
same study, MRI showed a tendency to overestimate the final tumor size. In 2009,
14 Preoperative (Breast) 283
Wasif et al. performed a comparative study between MG, US, and MRI [5]. Of the
61 patients evaluated, 52 were invasive ductal carcinomas. When the final size in
pathology was used as a parameter, the correlation was better with MRI, followed
by US and finally MG, identifying MRI as the exam with the best accuracy in rela-
tion to the pathological size of the lesion.
14.1.1.2 Ultrasound
Ultrasonography is the second most used exam, after mammography, for breast
assessment. It is a widely available test, with a lower cost in relation to MRI, but it
is also worth mentioning that this test depends on the operator and the device used.
Preferably, it should be performed in conjunction with mammography, for a more
complete and global assessment. During the exam, the patient is in the supine posi-
tion, a position similar to that of the surgery, and the characterization of the lesions
can assist in surgical planning, such as the skin distance, distance between lesions,
and distance from the nipple.
Like mammography, ultrasonography also has a moderate correlation coefficient
(0.68) with the pathological size of the tumor and is often due to the presence of
uncalcified DCIS, lobular histologies or tumor isoechogenicity in relation to the
breast parenchyma, especially in dense breasts.
14.1.1.3 Tomosynthesis
Despite the increasingly frequent use in surgical programming, the use of MRI in
preoperative locoregional staging is still controversial. Increased rates of mastec-
tomy, high rates of false-positive findings, and absence of evidence that the use of
MRI has an impact on patient survival are the main points of this controversy.
The objective of using MRI in the preoperative scenario should be the same as
for screening: increased sensitivity. The sensitivity of MRI in detecting multicentric
tumors and contralateral disease reaches 93% and 88%, respectively. Some studies
suggest that this increase in sensitivity is particularly valid in patients with dense
breasts and in lobular tumors. In theory, increased sensitivity should reduce positive
margin rates, decrease reoperation rates, and potentially decrease local recurrence.
However, the results of the studies are not consistent for these benefits and
284 J. Y. Maesaka et al.
The use of 18FDG PET-CT for breast is not routinely recommended. The method
has low sensitivity and low specificity in the local assessment of breast cancer. For
example, it has no capacity to detect lesions smaller than 1 cm, nor does it make the
differential diagnosis with inflammatory diseases or even fibroadenoma [9].
The presence of axillary disease in the context of breast cancer is an important prog-
nostic factor for the disease. Proper assessment of this condition will guide the
proposed therapy for each patient. The purpose of preoperative assessment is to
define whether or not there is axillary involvement and when present, the magnitude
of this involvement, also known as “axillary burden”. For this we can use, in addi-
tion to the physical examination, imaging methods. At the moment, no available
imaging exam has an acceptable negative predictive value to contraindicate surgical
assessment of the axilla when only normal lymph nodes are identified [10].
14.2.1 Indications
surgeon to request tests directed to the axilla, as we will discuss below and also as
explained in Chap. 4. In patients with clinically positive nodes, a scenario in which
the sentinel lymph node is contraindicated, ultrasound-guided fine-needle aspira-
tion (FNA) can confirm cytologically the presence of metastasis. In scenarios where
the physical examination is inconclusive, additional tests should be ordered, and
ultrasound can be of great use in conducting the case. The fact that ultrasound is an
operator-dependent exam must also be taken into account when ordering and inter-
preting the results of the exam.
14.2.2 Methods
14.2.2.1 Ultrasound
Ultrasonography is the most established exam for axillary evaluation. The suspicion
and diagnosis of axillary metastasis can modify the patient’s therapeutic planning:
negative axillae favors the performance of sentinel lymph node biopsy and, thus,
reduces the morbidity associated with axillary dissection. Positive axillae favors the
adoption of neoadjuvant treatment in order to reduce lymph node involvement by
disease and enable axillary surgical de-escalation. Finally, in some cases, it may be
contraindicated to perform the sentinel lymph node technique and guide the pro-
posal for up-front axillary dissection. Such an approach is detailed in Chap. 4.
In the ultrasound evaluation, morphological criteria (concentric or focal cortical
thickening greater than 3 mm, hilar obliteration, increased peri-hilar vasculariza-
tion, round shape) can be used, as well as the size of the lymph node, to identify
suspicious nodes. According to the criterion used, the specificity and sensitivity of
the method may vary. In a systematic review, which included literature from 1980
to 2004, Alvarez and colleagues found that in patients with palpable lymph nodes,
the sensitivity of the ultrasound was 68% on average when using the lymph node
size criteria, with a specificity of 75.2%, whereas if morphological criteria were
used, the sensitivity increased to 71% with a specificity of 96.1% on average. In the
group of patients without palpable lymph nodes, mean sensitivity and specificity
were 60.9% and 75.2%, respectively. However, when using the morphological cri-
teria in these patients, the average sensitivity was 43.9%, with 92.4% specific-
ity [11].
As we can see, there is a variety in the results found in the literature and in the
accuracy of the ultrasound to identify the lymph nodes, and this can be explained by
the variability of the method and criterion used, as well as whether the reference
used was sentinel lymph node biopsy or axillary dissection.
A cost-effective analysis study was conducted by Boughey et al., for the perfor-
mance of FNA guided by ultrasound in all lymph nodes with at least one altered
morphological criterion and compared with the performance of sentinel lymph node
biopsy intraoperatively, without performing axillary ultrasound (standard treat-
ment). As a result, the strategy proved to be cost-effective with a minimal difference
286 J. Y. Maesaka et al.
(US $ 36 dollars) between the strategies, and in 63% of the patients, there were
savings when the ultrasound was performed [12].
In 2017, a study published by Teixeira et al. created a nomogram for predicting
the presence of metastases based on the tumor characteristics and ultrasound image
of the axilla [13]. Using cortical thickening, tumor size, histological type, lesion
location as variables in the breast (quadrant) and the patient’s menopausal status,
the nomogram achieved satisfactory discrimination, with an area under the curve
(AUC) of 0.848.
14.2.2.2 Mammography
Although mammography is the main exam performed for breast assessment, its role
in the preoperative assessment of the axilla is limited [14]. In general, the axilla is
not fully included in the mammographic examination, and the accuracy of its find-
ings is limited. Benign lymph nodes appear on the mammographic exam smaller
than 2 cm in diameter and radiolucent hilar center. Suspicious findings are the
increased size and density of the lymph node.
Most MRI breast protocols cover the entire axillary field and allow a good evalua-
tion of this field. Suspicious findings in axillary lymph nodes on MRI include corti-
cal thickening, loss of fatty hilum, and rounded shape. The presence of perifocal
edema, defined as T2 hyperintensity in the adjacent fatty tissue (“marked T2 prolon-
gation in the surrounding fat”), demonstrated the greatest accuracy among the
descriptors, with a positive predictive value of 100% [15].
14.2.2.4 PET-CT
Another imaging method is the 18-FDG PET-CT, which has a role as a highly sensi-
tive method for detecting distant metastases, even in early tumors. However, its role
in axillary staging is limited: studies have shown a low to moderate sensitivity, and
therefore, it is not recommended for routine axillary assessment [16]. A prospective
trial involving 312 patients demonstrated that, compared to sentinel lymph node
biopsy, the sensitivity of FDG-PET/CT was 24%, with a specificity of 99% [17].
Another classic study by Veronesi et al. demonstrated a sensitivity of 37% [18], and
in a systematic review including 7 studies and 869 patients, PET-CT showed an
average sensitivity of 56% (95% CI 44 to 67%), with an average specificity of 95%
(95% CI 90 to 99%) [19].
14 Preoperative (Breast) 287
14.3.1 Case 1
This is a case of invasive ductal carcinoma in the left breast. What is demonstrated
here is the capacity of MRI in demonstrating larger extension of the tumor, includ-
ing involvement of the nipple (Figs. 14.1 and 14.2).
14.3.2 Case 2
In this case, the mammography was unable to identify the index breast cancer lesion
due to an extremely dense breast, but suspicious axillary lymph nodes were partially
seen. The lesion was identified on breast ultrasound as a hypoechogenic mass. With
MRI, we were able to confirm both the breast and the axillary lesions (Figs. 14.3,
14.4, and 14.5).
14.3.3 Case 3
In this case, all methods were able to accurately identify the breast lesion (Figs. 14.6,
14.7, and 14.8).
14.3.4 Case 4
In this case, suspicions axillary lymph nodes were identified in mammography and
confirmed on ultrasound (Figs. 14.9 and 14.10).
14.3.5 Case 5
References
1. Gutowski K, Gray D. The ASPS choosing wisely list for plastic surgery. Plast Reconstr Surg.
2014;134(4):853–5.
2. Sears ED, Lu YT, Swiatek PR, Chung TT, Kerr EA, Chung KC. Use of preoperative mam-
mography during evaluation for nononcologic breast reduction surgery. JAMA Surg.
2019;154(4):356–8. https://doi.org/10.1001/jamasurg.2018.4875.
3. Kim HR, Jung HK, Ko KH, Kim SJ, Lee KS. Mammography, US, and MRI for preoperative
prediction of extensive intraductal component of invasive breast cancer: interobserver vari-
ability and performances. Clin Breast Cancer. 2016;16(4):305–11. https://doi.org/10.1016/j.
clbc.2016.02.005.
4. Berg WA, Gutierrez L, MS NA, Carter WB, Bhargavan M, Lewis RS, et al. Diagnostic accu-
racy of mammography, clinical examination, US, and MR imaging in preoperative assessment
of breast cancer. Radiology. 2004;233(3):830–49. https://doi.org/10.1148/radiol.2333031484.
5. Wasif N, Garreau J, Terando A, Kirsch D, Mund DF, Giuliano AE. MRI versus ultrasonography
and mammography for preoperative assessment of breast cancer. Am Surg. 2009;75(10):970–5.
6. Eghtedari M, Tsai C, Robles J, Blair SL, Ojeda-Fournier H. Tomosynthesis in breast cancer
imaging: how does it fit into preoperative evaluation and surveillance? Surg Oncol Clin North
Am. 2018;27:33–49. http://dx.doi.org/10.1016/j.soc.2017.07.004.
7. Houssami N, Turner R, Macaskill P, Turnbull LW, McCready DR, Tuttle TM, et al. An individ-
ual person data meta-analysis of preoperative magnetic resonance imaging and breast cancer
recurrence. J Clin Oncol. 2014;32(5):392–401.
8. Sardanelli F, Boetes C, Borisch B, Decker T, Federico M, Gilbert FJ, et al. Magnetic resonance
imaging of the breast: recommendations from the EUSOMA working group. Eur J Cancer.
2010;46(8):1296–316. https://doi.org/10.1016/j.ejca.2010.02.015.
9. Ulaner GA. PET/CT for patients with breast cancer: where is the clinical impact? Am J
Roentgenol. 2019;213:254–65.
10. Rahbar H, Partridge SC, Javid SH, Lehman CD. Imaging axillary lymph nodes in patients with
newly diagnosed breast cancer. Curr Probl in Diagn Radiol. 2012;41:149–58.
11. Alvarez S, Añorbe E, Alcorta P, López F, Alonso I, Cortés J. Role of sonography in the diagno-
sis of axillary lymph node metastases in breast cancer: a systematic review. Am J Roentgenol.
2006;186:1342–8.
14 Preoperative (Breast) 295
12. Boughey JC, Moriarty JP, Degnim AC, Gregg MS, Egginton JS, Long KH. Cost modeling of
preoperative axillary ultrasound and fine-needle aspiration to guide surgery for invasive breast
cancer. Ann Surg Oncol. 2010;17(4):953–8.
13. Akissue de Camargo Teixeira P, Chala LF, Shimizu C, Filassi JR, Maesaka JY, de Barros
N. Axillary lymph node sonographic features and breast tumor characteristics as predictors
of malignancy: a nomogram to predict risk. Ultrasound Med Biol. 2017;43(9):1837–45.
https://doi.org/10.1016/j.ultrasmedbio.2017.05.003.
14. Dialani V, James DF, Slanetz PJ. A practical approach to imaging the axilla. Insights Imaging.
2015;6:217–29.
15. Baltzer PAT, Dietzel M, Burmeister HP, Zoubi R, Gajda M, Camara O, et al. Application
of MR mammography beyond local staging: is there a potential to accurately assess axil-
lary lymph nodes? Evaluation of an extended protocol in an initial prospective study. Am J
Roentgenol. 2011:196–5.
16. Liu Y. Role of FDG PET-CT in evaluation of locoregional nodal disease for initial staging of
breast cancer. World J Clin Oncol. 2014;5:982–9.
17. Pritchard KI, Julian JA, Holloway CMB, McCready D, Gulenchyn KY, George R, et al.
Prospective Study of 2-[ 18F]fluorodeoxyglucose positron emission tomography in the assess-
ment of regional nodal spread of disease in patients with breast cancer: an Ontario Clinical
Oncology Group study. J Clin Oncol. 2012;30(12):1274–9.
18. Veronesi U, De Cicco C, Galimberti VE, Fernandez JR, Rotmensz N, Viale G, et al. A com-
parative study on the value of FDG-PET and sentinel node biopsy to identify occult axillary
metastases. Ann Oncol. 2007;18(3):473–8.
19. Cooper KL, Meng Y, Harnan S, Ward SE, Fitzgerald P, Papaioannou D, et al. Positron emis-
sion tomography (PET) and magnetic resonance imaging (MRI) for the assessment of axillary
lymph node metastases in early breast cancer: Systematic review and economic evaluation.
Health Technol Assess. 2011;15(4):iii–iv, 1–134. https://doi.org/10.3310/hta15040.
Chapter 15
Systemic Staging (Total Body – When,
How)
15.1 Introduction
patients who are candidates for systemic staging by imaging, as well as the choice
of the most appropriate complementary exams to be requested.
The TNM system was proposed by the American Joint Committee on Cancer
(AJCC) and the Union for International Cancer Control (UICC) and is currently
considered the standard staging system. For a most accurate prognostic information,
the eighth edition of the TNM breast cancer classification in use since 2018 includes
histological and molecular characteristics to the anatomical staging previously
used [3].
Currently, it has been frequent the use of anatomical TNM classification criteria,
especially important for the definition of cancer treatments and the use of the prog-
nostic TNM classification (including histological and molecular information) for an
improved definition of the survival impact of these treatments.
The tumor size statement, either through physical examination and complementary
evaluation using imaging methods (cT) or by the anatomopathological measure of
the surgical specimen (pT), is a fundamental step of the TNM classification. The
lymph node involvement of the ipsilateral regional chains of the compromised
breast, either by clinical/imaging (cN) or anatomopathological (pN) evaluation,
associated with the tumor size, make up the main prognostic factors in nonmeta-
static breast cancer and are determinants in the therapeutic choice of this disease
regardless of the tumor subtype [3].
Determining the metastasis existence or absence (M), on the other hand, clarifies
about the virtual incurability of the disease when M1, although great advances have
emerged in the last years as options for palliative therapies which brought signifi-
cant improvements in survival and quality of life. Thus, the accurate recognition of
the systemic metastatic disease is essential to define at the initial diagnosis the pos-
sibility of a treatment strategy with curative potential (definitive local treatment
accompanied by neo/adjuvant systemic therapies) or the need for systemic palliative
treatment to control the metastatic disease [4].
Biomarkers in breast cancer are important because they are able to establish the
prognosis of the disease or, even, to predict the benefit of a specific treatment.
Associated with tumor size and lymph node involvement, the histological grade
15 Systemic Staging (Total Body – When, How) 299
determination is the most important risk factor for disease recurrence after the initial
treatment of a locoregional breast cancer [5]. The main factors that predict the ben-
efit of antihormonal treatment are the positive expression of estrogen or progester-
one receptors (HR-positive), as well as HER2 hyperexpression or amplification
(HER2-positive) which are directly associated with the benefit with anti-HER2
therapies [6, 7]. Nowadays, tests for the tumor gene expression analysis have been
able to refine the prognosis of the initial HR-positive, HER2-negative tumors, and
even helping to select the patient who needs adjuvant chemotherapy [8].
The changes in AJCC staging proposed in its eighth edition included the infor-
mation of histological grade, status of hormone receptors, HER2, and score of the
oncotype DX gene expression test as components of its classification. These changes
are taking into account the advances in the breast cancer treatment that occurred in
the past years and the consequent improvement in the prognosis of certain tumor
subtypes. With this new classification, the ability to predict the survival of breast
cancer patients undergoing conventional treatments has been significantly improved
[3]. However, the prognostic staging is less relevant for choosing which adjuvant
treatment should or should not be indicated.
The TNM classification of locoregional breast cancer (stages I, II, and III) helps to
determine the strategy of local and systemic treatment, since the more extensive the
locoregional disease, the greater the risks of future recurrence and, therefore, the
greater the absolute benefit of a more aggressive and extensive treatment. On the
other hand, the definition of metastatic disease at the initial diagnosis (de novo met-
astatic cancer/stage IV) establishes a virtual incurability and indicates palliative
strategies for its treatment. The failure to identify a metastatic lesion, with the con-
sequent under-staging of the neoplasia, may result in unnecessary treatments, such
as surgical or radiotherapy treatment of locoregional disease or even the indication
of futile neo−/adjuvant treatments for this scenario. In addition, understaging can
also represent inadequate choices for the treatment of metastatic disease [9].
Today, breast cancer represents the malignancy with the highest incidence in the
world, with an estimated 2.3 million people having this diagnosis in 2020 [10]. Of
these, approximately 90–95% are diagnosed as locoregional breast cancer, without
systemic metastases [11]. The chances of systemic involvement are directly associ-
ated with the locoregional involvement extension. It is more likely to identify sys-
temic metastases in more extensive locoregional disease or the more aggressive the
characteristics of this tumor are [9]. Therefore, it is possible to define the need for
300 F. de Almeida Costa and R. D. M. Linck
Unlike initial tumors, those that present with extensive loco-regionally neoplasms
may benefit from the systematic investigation of distant metastases. The incidence
of metastatic lesions to other organs and systems, when asymptomatic, is low even
in these cases. According to Ravaioli et al. [13], in a retrospective evaluation of
1218 cases of breast cancer in clinical stage (CS) III, the prevalence of abnormal
abdominal ultrasound and chest radiography was 6% and 7%, respectively. Myers
et al. [14] presented a meta-analysis of 11 studies, in which 8.3% of the CS III par-
ticipants had bone scintigraphy suggestive of bone metastasis, 2.0% had an abdomi-
nal ultrasound with suspected liver metastasis. and 1.7% had chest x-ray with
lesions suspected for metastasis.
In cases of locally advanced tumors, patients with T3 (≥5 cm) or N positive
(CS ≥ IIB) must undergo a systemic assessment that includes chest computerized
tomography (CT), CT or magnetic resonance imaging (MRI) of the abdomen, and
bone scintigraphy [1, 2, 12]. Exams with less sensitivity for the detection of sys-
temic metastases, such as abdominal ultrasound (US) and chest radiography, are
generally not useful in clinical practice because they have high rates of false nega-
tive [9]. Assessment of central nervous system (CNS) metastasis should be per-
formed exclusively when the patient has suggestive neurological symptoms and
preferably with MRI [4].
15 Systemic Staging (Total Body – When, How) 301
Patients who were suspected of having metastatic lesion on initial exams, whenever
possible, must undergo a biopsy of the metastatic site, since the metastatic diagnosis
is associated with a major impact on therapeutic planning. In addition to confirming
302 F. de Almeida Costa and R. D. M. Linck
The follow-up of the patient with breast cancer begins after the conclusion of
locoregional treatment and neo/adjuvant treatments and should be oriented towards
the monitoring of any side effects associated with the treatment employed, as well
as for the identification of any eventual neoplastic recurrence. Tumor relapse can be
divided into two distinct conditions: when locoregional relapse occurs in the breast
or in regional lymph nodes, which must be promptly identified as it is a potential
curable situation, the reason why it is important to have routine follow-up by imag-
ing exams, usually annual mammography, associated with physical exam [23], and
when systemic relapse occurs, which are virtually incurable. Thus, early identifica-
tion of systemic recurrence will not necessarily be able to change the natural history
of the disease, and early palliative treatments will not be able to translate into
increased chances of a cure or improved survival [24].
The main recommendation for the adequate breast cancer patients’ follow-up is
to carry out regular medical consultations for the active search for signs and symp-
toms suggestive of treatment toxicities and also the possibility of tumor recurrence
[25]. There is no recommendation for routine exams to identify asymptomatic sys-
temic metastases. However, any clinical suspicion should be promptly investigated.
15.7 Conclusion
On the other hand, failure to adequately identify metastatic disease can also lead to
inappropriate choices in the therapeutic planning of these patients. Thus, comple-
mentary exams must be carefully considered.
Patients should always be assessed by anamnesis and physical examination,
looking for signs and symptoms suggestive of metastatic spread, and any evidence
or suspicion of abnormality should be promptly investigated with appropriate exam-
inations. The main sites of metastasis in breast cancer are bones, lymph nodes,
lungs, liver, and central nervous system (the latter especially important in neoplasms
subtype HER2-positive or triple negative). Patients with loco-regionally advanced
tumors, especially those with anatomical staging IIB or more, should undergo rou-
tine imaging assessment when the initial diagnosis is made. The exams indicated for
this evaluation are chest CT, abdominal CT or MRI, and bone scintigraphy. PET-CT
FDG evaluation may be an alternative, especially when the initial exams are
not clear.
Biopsy for histological confirmation of systemic metastasis should be requested
whenever possible, since there is a great impact on therapeutic definitions. The
importance of biopsy of systemic metastases goes beyond the differential diagnosis
between benign conditions or eventually a second primary tumor. In breast cancer,
there is a great impact of immunohistochemical evaluation (related to the expres-
sion of hormone receptors and also of HER2) in the choice between different treat-
ment alternatives. The immunohistochemical profile may, in some cases, differ
between the primary and the metastatic tumor; also, there may be a change in the
immunohistochemical profile after long periods of exposure to cancer treatments.
Patients who are on follow-up after the end of treatment or in adjuvant endocrine
therapy need to be evaluated by trained medical staff in search of signs and symp-
toms suggestive of systemic neoplastic recurrence. When there is a clinical suspi-
cion of systemic metastasis, it necessarily needs to be promptly investigated with
correct and accurate propaedeutics. However, asymptomatic patients with no suspi-
cion of metastatic spread should not be subjected to indiscriminate routine imaging
exams for systemic evaluation.
References
6. Breast E, Trialists C; Group C. Relevance of breast cancer hormone receptors and other
factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised
trials. Lancet [Internet]. 2011;378(9793):771–84. Available at: https://doi.org/10.1016/
S0140-6736(11)60993-8.
7. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, et
al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl
J Med [Internet]. 2005;353(16):1659–72. Available at: http://www.ncbi.nlm.nih.gov/
pubmed/16236737.
8. Andre F, Ismaila N, Stearns V. Use of biomarkers to guide decisions on adjuvant systemic
therapy for women with early-stage invasive breast cancer: ASCO clinical practice guideline
update summary. J Oncol Pract [Internet]. 2019;15(9):495–7. Available at: http://ascopubs.org/
doi/10.1200/JOP.19.00264.
9. Soares G, Pereira A, Vilas Boas M, Vaisberg V, Magalhães M, Linck R, et al. Value of sys-
temic staging in asymptomatic early breast cancer. Rev Bras Ginecol e Obs/RBGO Gynecol
Obstet [Internet]. 2018;40(07):403–9. Available at: http://www.thieme-connect.de/DOI/
DOI?10.1055/s-0038-1666997.
10. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer
statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers
in 185 countries. CA Cancer J Clin [Internet]. 2021:caac.21660. Available at: https://onlineli-
brary.wiley.com/doi/10.3322/caac.21660.
11. Cardoso F, Spence D, Mertz S, Corneliussen-James D, Sabelko K, Gralow J, et al.
Global analysis of advanced/metastatic breast cancer: decade report (2005–2015). Breast
[Internet]. 2018;39:131–8. Available at: https://linkinghub.elsevier.com/retrieve/pii/
S096097761830050X.
12. Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. Early breast
cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann
Oncol. 2019;30(8):1194–220.
13. Ravaioli A, Pasini G, Polselli A, Papi M, Tassinari D, Arcangeli V, et al. Staging of breast
cancer: new recommended standard procedure. Breast Cancer Res Treat. 2002;72(1):53–60.
14. Myers RE, Johnston M, Pritchard K, Levine M, Oliver T, Crump RM, et al. Baseline staging
tests in primary breast cancer: a practice guideline. CMAJ. 2001;164(10):1439–44.
15. Fuster D, Duch J, Paredes P, Velasco M, Muñoz M, Santamaría G, et al. Preoperative stag-
ing of large primary breast cancer with [ 18F]fluorodeoxyglucose positron emission tomog-
raphy/computed tomography compared with conventional imaging procedures. J Clin Oncol.
2008;26(29):4746–51.
16. Groheux D, Giacchetti S, Espié M, Vercellino L, Hamy AS, Delord M, et al. The yield of 18F-
FDG PET/CT in patients with clinical stage IIA, IIB, or IIIA breast cancer: a prospective study.
J Nucl Med. 2011;52(10):1526–34.
17. Carkaci S, Macapinlac HA, Cristofanilli M, Mawlawi O, Rohren E, Gonzalez Angulo AM, et
al. Retrospective study of 18FFDG PET/CT in the diagnosis of inflammatory breast cancer:
preliminary data. J Nucl Med. 2009;50(2):231–8. https://doi.org/10.2967/jnumed.108.056010.
18. Linden HM, Peterson LM, Fowler AM. Clinical potential of estrogen and progesterone recep-
tor imaging. PET Clin [Internet]. 2018;13(3):415–22. Available at: http://www.ncbi.nlm.nih.
gov/pubmed/30100079.
19. Jones EF, Ray KM, Li W, Chien AJ, Mukhtar RA, Esserman LJ, et al. Initial experience of
dedicated breast PET imaging of ER+ breast cancers using [F-18]fluoroestradiol. NPJ Breast
Cancer [Internet]. 2019;5:12. Available at: http://www.ncbi.nlm.nih.gov/pubmed/31016232.
20. Van Poznak C, Somerfield MR, Bast RC, Cristofanilli M, Goetz MP, Gonzalez-Angulo
AM, et al. Use of biomarkers to guide decisions on systemic therapy for women with meta-
static breast cancer: American Society of Clinical Oncology Clinical Practice Guideline. J
Clin Oncol [Internet]. 2015;33(24):2695–704. Available at: http://www.ncbi.nlm.nih.gov/
pubmed/26195705.
15 Systemic Staging (Total Body – When, How) 305
Ana Carolina de Ataíde Góes, Heni Debs Skaf, and Laura Testa
Abbreviations
16.1 Indications
Neoadjuvant systemic therapy (NST) was first introduced in breast cancer treatment
to make surgery possible for inoperable tumors.
As systemic treatment evolved and survival benefits were shown for adjuvant
treatment, the scenario changed, and once a patient has a clear indication of chemo-
therapy, for example, it could be used upfront, making breast conservation possible.
More recently, residual disease in pathological surgical specimens after neoadju-
vant chemotherapy (NCT) is a marker for the need to escalate treatment, both for
HER2 and triple-negative (TN) breast cancer.
The current understanding of the breast medical oncology community is that for
TN disease we should offer NCT for patients with tumor size over 2 cm (some
would say even 1 cm) irrespectively of nodal involvement. For HER2-positive dis-
ease, this is also the case, with some debate on the agents and regimens.
Patients that present with HR-positive breast cancer are usually referred to NCT
when they have locally advanced disease or any node-positive patients who have
other markers of better response to treatment such as Grade 3 or high Ki67
(Table 16.1).
Neoadjuvant endocrine therapy (NET) development followed a different path.
Patients who had postmenopausal locally advanced disease but were not fit for che-
motherapy were offered first tamoxifen with 50% of patients achieving clinical
responses. When aromatase inhibitors became available, they showed improved
rates of response and chances for BCS (breast-conserving surgery).
PCR is not as prognostic for HR (hormone receptor)-positive breast cancer as it
is for TN- or HER2-positive disease, but patients who achieve low Ki67, tumor
staging pT2 or less, and pN0 after 24 weeks of NET have excellent disease-free and
overall survival. These tumor characteristics are evaluated with PEPI (preoperative
endocrine prognostic index) score, and it has been useful. NET was better studied in
postmenopausal patients, so it’s not routinely recommended for premenopausal
HR-positive breast cancer.
Presently there are no established guidelines to best evaluate tumoral response dur-
ing or after NST. However, prior to initiation of chemotherapy, diagnostic imaging
should be performed for both breasts, and, depending on the disease stage and the
presence of symptoms, whole-body imaging should be conducted as well.
Historically, this first evaluation consists of conventional breast imaging (mammog-
raphy and ultrasound), besides physical examination.
Imaging monitoring during NST may help anticipate which patients aren’t expe-
riencing a response in cases of uncertain clinical progression, avoiding unnecessary
chemotherapeutic toxicity. It also permits the oncologist to tailor specific therapy
regimens to be used before or after surgery.
The same imaging modality and protocol should be executed after NST in order
to assist to predict which patients will achieve pCR, as well as estimate residual
tumoral size for the purpose of planning a BCS.
Breast tumor should be clipped with image-detectable marker prior to neoadju-
vant therapy for future preoperative localization and identification of tumor bed.
Similarly, it’s important to clip metastatic axillary lymph node(s). Preferable to use
clip that can be well seen on US for future localization (Fig. 16.1).
However, the surgical planning may dictate the decision whether or not to obtain
posttreatment imaging studies: repeating breast imaging after neoadjuvant therapy
may not be necessary for patients with multicentric disease, extensive lesions, or
another clear contraindication to BCS.
Physical examination is subjective and relies on the physician’s experience, and
its accuracy to determine pCR is inferior to the usual breast imaging methods [1].
Besides, the presence of fibrosis and firm fibroglandular tissue may overestimate the
amount of residual disease, and the absence of a palpable lesion does not exclude
the presence of a remaining tumor, being notably inaccurate for small early-stage
cancers, particularly smaller than 2 cm [2].
a b c
Fig. 16.1 Marker clip (arrows) within breast lesion on different imaging modalities. (a)
Mammography shows a metallic clip. (b) The tissue marker is visible on ultrasound as hyperecho-
genic spots. (c) T1-weighted MR image shows local signal intensity void caused by marker clip
310 A. C. de Ataíde Góes et al.
a b c d
e f
Fig. 16.2 CLM, 58-year-old female. MLO mammography before and after neoadjuvant systemic
therapy shows two partially obscured masses (a), with signs of partial response to therapy charac-
terized by reduced size associated to morphological changes of the lesions, now irregular and
spiculated (b). However, despite the reduction in size after neoadjuvant systemic therapy (c, e), the
cutaneous involvement became more pronounced on post-contrast MR subtracted images (d, f),
emphasizing the intrinsic limitation of mammography in the skin evaluation
In the following sections, we will discuss each imaging modality, its advantages
and limitations, as well as pitfalls and pearls in response evaluation. DCE-MRI
offers the highest diagnostic accuracy in primary tumor therapy response assess-
ment among the currently established imaging methods (mammography and ultra-
sound) [1], and its usage for response evaluation to NST is recommended by the
American College of Radiology and European Society of Breast Imaging
(Fig. 16.2) [3, 4].
Mammography and ultrasound (US) provide suboptimal evaluations in the accu-
rate assessment of response to NST, and such limitations will be further discussed
in this chapter. Predicting pCR is not highly accurate with US or mammography,
correlating with these methods in only 13–25% of cases [5–7].
Despite DCE-MRI more accurately reflects true pathologic tumor size, it is more
expensive, time-consuming, and less widely available and may still overestimate or
underestimate residual disease but only within 1 cm of the final tumor size when
compared to anatomopathological examination of the surgical specimen [8, 9]. The
largest diameter of the lesion in the DCE-MRI showed the highest correlation with
pathology in the evaluation of residual invasive disease after NST [10].
a b c d
e f
Fig. 16.3 GALO, 32-year-old female. MLO mammography before neoadjuvant systemic therapy
shows an indistinct lesion occupying almost the entire left breast, diagnosed as invasive ductal
carcinoma, associated with pleomorphic clustered calcifications (a). After the systemic therapy,
there was partial regression of the lesion; nevertheless, we observed an increase in the extent and
density of the calcifications (b). Post-contrast MRI before treatment characterizes an irregular
mass with heterogeneous enhancement associated with non-mass diffuse enhancement of the left
breast (c, d). After neoadjuvant systemic therapy, there were signs of a partial response character-
ized by a complete regression of the mass lesion with residual non-mass enhancement in multiple
areas (e, f)
312 A. C. de Ataíde Góes et al.
Breast and axillary US should be performed for malignant breast masses for clinical
staging if neoadjuvant therapy is planned, being widely available and
cost-effective.
There are some limitations though, as in physical examination and mammogra-
phy, chemotherapy-induced fibrosis can be difficult to differentiate from residual
disease by US and lesion fragmentation may underestimate its therapeutic response
evaluation (Fig. 16.4).
Keune and colleagues retrospectively evaluated 196 patients diagnosed with
invasive breast carcinoma and concluded that ultrasound was more accurate than
16 Neoadjuvant Systemic Therapy 313
a b c
d e f
Fig. 16.4 BGK, 53-year-old female. Ultrasound of the left breast shows a hypoechoic irregular
mass with angulated margins in the retroareolar region diagnosed as invasive ductal carcinoma (a,
b), characterized on MRI as an irregular mass with heterogeneous enhancement (c). The lesion
was marked with a clip (arrow) before systemic therapy and later characterized on ultrasound as a
smaller hypoechoic irregular lesion (d, e) without corresponding enhancement on MRI (f). The
final surgical specimen yielded a small ductal carcinoma in situ, without invasive component, with
extensive regression signs
a b c e
Fig. 16.5 MAS, 50-year-old female. MLO and CC mammography show an indistinct irregular
mass at the junction of the inner quadrants on the right breast, diagnosed as invasive ductal carci-
noma, as well as two smaller lesions at the upper outer quadrant and round enlarged axillary lymph
nodes (a, b). These lymph nodes were better evaluated by ultrasound, both with thickened cortex
and no visible hilum (c, d). Ultrasound better characterized both lesions in the upper outer quad-
rant which were diagnosed as compromised lymph nodes after core needle biopsy (e). All the
breast lesions were marked with a clip before neoadjuvant systemic therapy for better surgical
planning (f)
Magnetic resonance imaging (MRI) is the most accurate and reproducible method
available to monitor breast cancer response to NST, being an excellent modality to
define the extent of the disease and assist the surgeon in determining the optimal
surgical approach. However, like any method, it has its advantages and limitations,
and in this scenario, it can both overestimate and underestimate the tumor response.
According to the different aims for which NST is applied, MRI can be used for
different purposes. From an oncologist’s perspective, assessing the response to a
16 Neoadjuvant Systemic Therapy 315
specific regimen and measuring changes in the size of the invasive tumor as early as
possible helps to adapt the regimen in case of failure to respond [25, 26]. MRI has
also been used to monitor the degree of response to induction chemotherapy as a
surrogate marker for pCR, that is, to guide further systemic treatment [27]. For a
surgeon, assessing the extent of possible residual disease, including DCIS, is impor-
tant to guide subsequent surgery [28, 29]. Thus, separate strategies might be needed
to determine response to a specific chemotherapy regimen and to determine residual
tumor size after NST.
MRI Technique MRI is often acquired on a scanner of at least 1.5-T using a
dedicated breast coil in the prone position. The imaging protocol usually consists
of fat-suppressed axial T2-weighted images, diffusion-weighted imaging (DWI),
and dynamic contrast-enhanced fat-suppressed T1-weighted images, besides stan-
dard subtraction images [30]. For further improvement of therapy response predic-
tion and monitoring, other advanced imaging approaches are being evaluated.
These include the quantitative dynamic contrast-enhanced imaging, advanced
DWI techniques, and spectroscopic imaging, which are under investigation for the
prediction of neoadjuvant therapy effects [31]. With the advent of artificial intel-
ligence (AI) and machine learning, the large data sets provided by and potentially
extractable from breast MRI make it suitable for AI applications. Recent develop-
ments in predicting the tumor response to the NST show promising results in
objective interpretation of MR images using AI methods in personalized care for
breast cancer patients [32]. However, there still no consensus on its broad
application.
The first step for suitable evaluation is cognizing the factors that affect the accuracy
of MRI in demonstrating residual disease after NST, in order to adopt a tailored
interpretation strategy. These factors include image acquisition parameters, meth-
ods of administration of contrast material, the histologic type of the tumor (ductal
vs lobular), and the status of the tumor’s HR and HER2 receptors. The size of lobu-
lar or HR-positive/ HER2-negative cancers tended to be underestimated on MRI
compared to ductal or other subtypes, respectively (Fig. 16.6) [36]. In cases of
HR-positive cancers, late phases of postcontrast MRI are better to evaluate response.
In addition, factors such as therapy agents and baseline tumor morphology may
modulate the MRI accuracy in identifying residual disease (see below).
To assess response to NST, changes in maximum tumor size, tumor volume, and
enhancement kinetics have been used, as well as functional techniques, including
various DWI, and molecular imaging techniques are being investigated [30]. The
Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 guidelines are stan-
dardized criteria widely used for response assessment [37]. Based upon measure-
ment of a solid tumor in at least the longest diameter, four response categories are
recognized: complete response, partial response, stable disease, and progressive
disease (Table 16.2). Due to its emphasis on changes in lesion size and the unique
a b c
Fig. 16.6 A 50-year-old woman with invasive ductal carcinoma (estrogen receptor-positive and
HER2-negative). (a) Prechemotherapy MRI shows a 6.0 cm irregular enhancing mass (arrows). (b)
MRI obtained after completion of four cycles of doxorubicin/cyclophosphamide followed by four
cycles of docetaxel in the early post-contrast phase (90 seconds) shows an 0.3 cm enhancing lesion
(arrow). (c) An image obtained in the delayed phase (360 seconds) shows the lesion measuring
0.5 cm (arrow). Surgical histopathology revealed a 4.8 cm invasive ductal carcinoma and 5.0 cm
total tumor size (including both invasive tumor and ductal carcinoma in situ). (Reprinted with
permission from Reig et al. [47]. https://doi.org/10.1002/jmri.27145)
16 Neoadjuvant Systemic Therapy 317
Table 16.2 Evaluation of target and nontarget lesions according to Response Evaluation Criteria
in Solid Tumors (RECIST), version 1.1
Type of lesion and response Criteria
Target lesion
Complete response (CR) Disappearance of all target lesions
Partial response (PR) Decrease in the sum of the diameters of the target lesions ≥30%
Progressive disease (PD) Increase in the sum of the diameters of the target lesions ≥20%
Appearance of new lesions
Stable disease Does not meet criteria for CR, PR, or PD
Non-target lesion
Complete response (CR) Disappearance of all non-target lesions
Non-CR/non-PD Persistence of non-target lesions
Progressive disease (PD) Appearance of new lesions
Unequivocal progression of the non-target lesions
features of breast cancer, RECIST may not be reliable in this setting, being criti-
cized for failing to capture meaningful changes in tumor biology [38].
The measurement of the functional tumor volume was a better predictor of
response than the change in the longest diameter, as shown in the American College
of Radiology Imaging Network (ACRIN) 6657 trial [34]. Furthermore, tumor vol-
ume change was found to be a predictor of recurrence-free survival even as early as
after one cycle of chemotherapy (Fig. 16.7) [27].
Changes in time-signal intensity curve analysis or pharmacokinetic parameters
may also be helpful in predicting response [39], with an early decrease in enhance-
ment as an important predictor of response [40, 41].
Likewise, the apparent diffusion coefficient (ADC) has shown promise for
detecting early response to therapy, with an increase in ADC after treatment as a
predictor of response (Fig. 16.8). Although it sounds appealing, the use of DWI as
an exclusive method to visualize residual disease is limited by its lower spatial reso-
lution compared with DCE-MR imaging [42, 43].
The combination of changes in volumetric and functional assessment of the
tumor may improve the specificity of the diagnosis; however, large-scale studies
with standardized image acquisition are needed to implement these parameters as
alternatives to conventional size measurements.
Still, to date there is no uniformly accepted method to obtain response assess-
ment. In order to lead to a more standardized and less subjective evaluation of breast
MRI acquisitions, a proposal for response types follows:
a b
c d
Fig.16.7 A 57-year-old woman with invasive ductal carcinoma. (a) Prechemotherapy maximal
intensity projection (MIP) image of contrast-enhanced MRI shows a 4.5 cm irregular mass (arrow).
(b) Post chemotherapy MIP image of contrast-enhanced MRI shows a 1.7 cm irregular mass
(arrow) [(4.5–1.7)/4.5] × 100 = 62% reduction of the initial tumor diameter, suggestive of partial
response. Computer-aided volumetry (red and yellow color) shows a 95% volume reduction from
27.4 cc (c) to 1.5 cc (d). (Reprinted with permission from Reig et al. [47]. https://doi.org/10.1002/
jmri.27145)
16 Neoadjuvant Systemic Therapy 319
a b c
d e f
Fig. 16.8 A 44-year-old woman with invasive ductal carcinoma. (a) Prechemotherapy contrast-
enhanced subtraction T1-weighted MRI shows an enhancing mass in the left breast (arrow). The
maximal diameter of the mass was measured up to 7.4 cm. (b) After the first cycle of chemother-
apy, the mass was measured up to 5.7 cm (arrow). (c) After the eight cycle of chemotherapy, the
mass is not seen. ADC maps using b = 0 and 750 s/mm2 at baseline (d), post-first cycle (e), and
post-eight cycle chemotherapy timepoints (f) show the signal intensity increases after chemother-
apy. The ADC value for the lowest signal intensity of the tumor was 1.103 × 10−3 mm2/sec,
1.463 × 10−3 mm2/sec, and 2.162 × 10−3 mm2/sec, respectively. %ΔADC was 32.6% at post-first
cycle chemotherapy. The patient underwent surgery, and surgical histopathology revealed no resid-
ual invasive ductal carcinoma. (Reprinted with permission from Reig et al. [47]. https://doi.
org/10.1002/jmri.27145)
invasive disease or no residual invasive and in situ disease in the breast and axilla
[46]. As MRI cannot reliably distinguish between invasive disease and ductal
carcinoma in situ (DCIS), defining pCR as no residual invasive disease will
increase MRI overestimation rates, as MRI identifies residual DCIS that is not
counted in final pathology [47]. It is important to evaluate MRI enhancement in
both early and delayed phases in any patient after NST to reduce underestimation
of risk of residual disease, mainly in patients undergoing antiangiogenic thera-
pies [48]. Although some groups have found that late enhancement may be seen
in HER2+ cancers without residual disease [49], studies evaluating MRI param-
eters by breast cancer subtype are needed to further elucidate these findings.
Lastly, remember that BI-RADS category 6 must be used even in cases of com-
plete response.
–– Partial response: tumor assessment on MRI might be challenging, as NST
causes various histopathological changes in tumor cellularity, causing some
tumors to show concentric shrinkage pattern (usually single mass) [50], while
others may crumble (“fragmentation”) into scattered islands of tumor cells
(Fig. 16.10). In the latter case, a response is present, but the area of residual dis-
ease may remain similar to that prior to treatment. In addition, it is difficult to
determine whether the multinodular enhancing lesions are residual invasive can-
cer, DCIS, or a reactive change after therapy [36]. A shrinkage pattern has also
been found to be associated with tumor molecular subtype [51], with HER2+
tumors more likely to demonstrate concentric shrinkage, and HR-tumors are
320 A. C. de Ataíde Góes et al.
a b
c d
Fig. 16.9 A 38-year-old woman with triple-negative invasive ductal carcinoma of the right breast.
Axial subtraction MRI: Image acquired before the NST demonstrates (a) a right breast mass
(arrow) and (b) pathological lymph nodes (arrow). (c, d) Images after neoadjuvant chemotherapy
show resolution of all lesions characterizing complete response. PCR confirmed after breast-
conserving surgery
Fig. 16.10 Shrinkage patterns after neoadjuvant therapy includes concentric shrinkage and reduc-
tion into residual nodular lesions (fragmentation)
Although MRI performs better than conventional modalities overall, it does have
limits due to false-positive and false-negative results. Studies have found that MRI
underestimated residual disease in 10% of cases and overestimated in 33% of cases
by up to 1.0 cm [52, 53]. The potential clinical impact of overestimation of residual
tumor is the resection of a larger amount of tissue during breast conservation sur-
gery, which may negatively alter cosmetic outcome or influence a decision for
mastectomy. The impact of underestimation of residual tumor is the potential for
an incomplete resection with positive margins and the need for surgical
re-excision.
Thus, it is important to be aware of the following factors [36, 47, 54–56]:
322 A. C. de Ataíde Góes et al.
a b c
d e f
Fig. 16.11 A 31-year-old female with invasive breast carcinoma (not otherwise specified). Pre- (a)
and post-therapy (d) MIP image shows an irregular and indistinct mass that decreased into a frag-
mented pattern after NST. Despite a partial response from the index lesion, new lesions have
appeared on the lower outer quadrant of the right breast (e – circle) and left breast (f - circle), not
shown on prechemotherapy images (b, c), characterizing progressive disease
a b
Fig. 16.12 A 40-year-old female with left invasive mucinous carcinoma (luminal B). (a) MIP
image shows diffuse non-mass enhancement in the left breast that persists after NST (b). Despite
the extent of enhancement in MRI, postoperative histopathologic findings showed an excellent
pathological response (low cellularity)
–– Fibroadenomas and other benign lesions may be stable after therapy, and biopsy
may be required to differentiate them from residual disease.
MRI has a significant role in some lesions that are difficult to assess by other modal-
ities, especially in response to nonmeasurable lesions. In addition to non-mass
enhancement, MRI is also useful in evaluating inflammatory breast cancer and com-
plex cystic lesion.
The diagnosis of IBC is clinical, with a combination of skin changes and breast
enlargement presenting rapidly (few weeks or few months), with or without an
underlying palpable mass. MRI can detect a primary breast lesion in 98% of the
cases, and the most common finding is an index mass or multiple small masses.
Skin abnormalities, such as thickening, edema, and cutaneous masses or enhancing
foci, are also better detected with MRI than other modalities. As the disease is best
seen with MRI, response prediction and residual tumor size correlation with final
pathology are also better with MRI than ultrasound or mammography [57]. Note
that the assessment of the post-therapy response of the skin and nipple-areola com-
plex thickening is limited and should be valued when these structures enhance,
especially in a nodular and irregular pattern (Fig. 16.13).
Furthermore, the exact extent of the tumor in complex solid-cystic lesions may
be difficult to delineate. MRI allows the identification of intratumoral necrosis when
there is a very high signal intensity (similar to water) within the tumor in the fat-
suppressed T2-weighted MRI scans [58]. In these cases, it is important to measure
and follow the entire lesion and also the solid component, as necrotic tumors may
leave residual acellular masses.
324 A. C. de Ataíde Góes et al.
a b
c d
COMPLETE RESIDUAL
PATHOLOGICAL DISEASE
RESPONSE
Fig. 16.13 Skin and nipple assessment of two different patients with inflammatory breast cancer
(MRI T1W CE with subtraction). Images on the left from the same patient who presented global
skin thickening of the right breast (a – arrowhead) that decreased after NST (c). Final pathologic
findings after mastectomy surgery showed complete pathologic response. On the other hand,
images on the right from another patient show diffuse non-mass enhancement with retraction and
enhancement of nipple-areola complex (b – circle) in addition to skin enhancement that persists
after NST (d – arrow), characterizing residual disease
NST can be used to reduce tumor burden and downstage the axilla, with pCR rates
of 50–60% in the axillary lymph nodes (LN) [59]. In patients who are expected to
achieve axillary pCR, omission of axillary LN dissection could prevent morbidity
and complications such as lymphedema; therefore, sentinel LN biopsy (SLNB)
could be an alternative surgical method [60]. Regarding axillary management after
NST, two prospective observational trials – the Alliance Z1071 and SENTINA
16 Neoadjuvant Systemic Therapy 325
trials – suggested that if at least three sentinel LNs were obtained; then SLNB could
be sufficient in patients who become clinically node-negative after NST [59, 61].
However, the overall false-negative rates of the two aforementioned studies were
13–14%, which is above the accepted cutoff value of 10%. Thus, axillary LN dis-
section has been the standard treatment in clinically node-positive breast cancer
after NST due to the lack of consensus in the selection of proper candidates for
SLNB [62] (see Chap. 4).
According to the American College of Radiology (ACR) Appropriateness
Criteria, the most accurate imaging modality in the assessment of residual disease
after NST is MRI for primary breast cancer and US for axillary LN [63].
Although some controversies remain with unsatisfactory false-negative rates, US
is still known to be the most accurate modality in the evaluation of the axillary LN
in NST setting [64, 65].
MRI may also play a role in evaluating LN; however, the sensitivity of post neo-
adjuvant MRI in the detection of persistent LN metastasis is only moderate, ranging
from 61% to 72% [66, 67]. Likewise, axillary regions may be insufficiently included
in MR images.
Suspicious features of axillary LNs include cortical thickening of more than
3 mm, round or irregular shape, or loss of fatty hilum [68] (Fig. 16.14). While a cut
of a value of 3 mm of cortical thickness is used, a small residual metastatic focus
can be missed, or fibrosis and reactive enlargement of 3 mm may be misinterpreted
as residual disease [69].
Note that radiological and pathological response to NST in the breast and axilla
are often correlated, but a proportion of patients (14.4%) have different responses in
the two sites [69] (Fig. 16.15). Therefore, it is recommended that the axilla and the
breast should be viewed and assessed as two separate entities for treatment planning.
326 A. C. de Ataíde Góes et al.
a b
c d
Fig. 16.15 T1W CE MRI in a 60-year-old female with triple-negative invasive ductal carcinoma
in the right breast. Upper images demonstrate baseline MRI: (a) irregular mass in the right breast;
(b) pathologic axillary lymph node (arrow). Bottom images: post-NST MRI shows an increase in
the diameter of the breast mass and evidence of muscular and skin invasion (c), characterizing
progressive breast disease. Even so, there was a reduced lymph node as shown in (d) (arrowheads),
and no axillary metastasis was found at surgery, which impacted the management of the axilla. The
response to NST in the breast and axilla is not always similar, so it is important to evaluate them
separately
References
5. Herrada J, Iyer RB, Atkinson EN, Sneige N, Buzdar AU, Hortobagyi GN. Relative value of
physical examination, mammography, and breast sonography in evaluating the size of the pri-
mary tumor and regional lymph node metastases in women receiving neoadjuvant chemo-
therapy for locally advanced breast carcinoma. Clin Cancer Res. 1997;3(9):1565–9.
6. Vinnicombe SJ, MacVicar AD, Guy RL, Sloane JP, Powles TJ, Knee G, Husband JE. Primary
breast cancer: mammographic changes after neoadjuvant chemotherapy, with pathologic cor-
relation. Radiology. 1996;198(2):333–40. https://doi.org/10.1148/radiology.198.2.8596827.
7. von Minckwitz G, Dan Costa S, Eiermann W, Blohmer JU, Tulusan AH, Jackisch C, Kaufmann
M. Maximized reduction of primary breast tumor size using preoperative chemotherapy with
doxorubicin and docetaxel. J Clin Oncol. 1999;17(7):1999–2005. https://doi.org/10.1200/
jco.1999.17.7.1999.
8. Yeh E, Slanetz P, Kopans DB, Rafferty E, Georgian-Smith D, Moy L, Halpern E, Moore
R, Kuter I, Taghian A. Prospective comparison of mammography, sonography, and MRI in
patients undergoing neoadjuvant chemotherapy for palpable breast cancer. Am J Roentgenol.
2005;184(3):868–77. https://doi.org/10.2214/ajr.184.3.01840868.
9. Rosen EL, Blackwell KL, Baker JA, Soo MS, Bentley RC, Yu D, Samulski TV, Dewhirst
MW. Accuracy of MRI in the detection of residual breast cancer after neoadjuvant chemo-
therapy. Am J Roentgenol. 2003;181(5):1275–82. https://doi.org/10.2214/ajr.181.5.1811275.
10. Scheel JR, Kim E, Partridge SC, et al. MRI, clinical examination, and mammography for pre-
operative assessment of residual disease and pathologic complete response after neoadjuvant
chemotherapy for breast cancer: ACRIN 6657 trial. Am J Roentgenol. 2018;210(6):1376–85.
https://doi.org/10.2214/AJR.17.18323.
11. Huber S, Wagner M, Zuna I, Medl M, Czembirek H, Delorme S. Locally advanced breast
carcinoma: evaluation of mammography in the prediction of residual disease after induction
chemotherapy. Anticancer Res. 2000;20(1B):553–8.
12. Förnvik D, Zackrisson S, Ljungberg O, Svahn T, Timberg P, Tingberg A, Andersson I. Breast
tomosynthesis: accuracy of tumor measurement compared with digital mammography and ultra-
sonography. Acta Radiol. 2010;51(3):240–7. https://doi.org/10.3109/02841850903524447.
13. Adrada BE, Huo L, Lane DL, Arribas EM, Resetkova E, Yang W. Histopathologic correla-
tion of residual mammographic microcalcifications after neoadjuvant chemotherapy for
locally advanced breast cancer. Ann Surg Oncol. 2015;22(4):1111–7. https://doi.org/10.1245/
s10434-014-4113-8.
14. Kuhl CK. MRI of breast tumors. Eur Radiol. 2000;10(1):46–58. https://doi.org/10.1007/
s003300050006.
15. Helvie MA, Joynt LK, Cody RL, Pierce LJ, Adler DD, Merajver SD. Locally advanced
breast carcinoma: accuracy of mammography versus clinical examination in the prediction of
residual disease after chemotherapy. Radiology. 1996;198(2):327–32. https://doi.org/10.1148/
radiology.198.2.8596826.
16. Moskovic EC, Mansi JL, King DM, Murch CR, Smith IE. Mammography in the assessment of
response to medical treatment of large primary breast cancer. Clin Radiol. 1993;47(5):339–44.
https://doi.org/10.1016/S0009-9260(05)81451-5.
17. Patel BK, Hilal T, Covington M, Zhang N, Kosiorek HE, Lobbes M, Northfelt DW, Pockaj
BA. Contrast-enhanced spectral mammography is comparable to MRI in the assess-
ment of residual breast cancer following neoadjuvant systemic therapy. Ann Surg Oncol.
2018;25(5):1350–56. https://doi.org/10.1245/s10434-018-6413-x.
18. Iotti V, Ravaioli S, Vacondio R, Coriani C, Caffarri S, Sghedoni R, et al. Contrast-enhanced
spectral mammography in neoadjuvant chemotherapy monitoring: a comparison with breast
magnetic resonance imaging. Breast Cancer Res. 2017;19(1):106. https://doi.org/10.1186/
s13058-017-0899-1.
19. Chagpar AB, Middleton LP, Sahin AA, et al. Accuracy of physical examination, ultrasonog-
raphy, and mammography in predicting residual pathologic tumor size in patients treated
with neoadjuvant chemotherapy. Ann Surg. 2006;243(2):257–64. https://doi.org/10.1097/01.
sla.0000197714.14318.6f.
328 A. C. de Ataíde Góes et al.
20. Peintinger F, Kuerer HM, Anderson K, et al. Accuracy of the combination of mammography
and sonography in predicting tumor response in breast cancer patients after neoadjuvant chemo-
therapy. Ann Surg Oncol. 2006;13(11):1443–9. https://doi.org/10.1245/s10434-006-9086-9.
21. Ollivier L, Balu-Maestro C, Leclére J. Imaging in evaluation of response to neo-
adjuvant breast cancer treatment. Cancer Imaging. 2005;5(1):27–31. https://doi.
org/10.1102/1470-7330.2005.0009.
22. Roubidoux MA, LeCarpentier GL, Fowlkes JB, Bartz B, Pai D, Gordon SP, Schott AF,
Johnson TD, Carson PL. Sonographic evaluation of early-stage breast cancers that undergo
neoadjuvant chemotherapy. J Ultrasound Med. 2005;24(7):885–95. https://doi.org/10.7863/
jum.2005.24.7.885.
23. Cao X, Xue J, Zhao B. Potential application value of contrast-enhanced ultrasound in neoadju-
vant chemotherapy of breast cancer. Ultrasound Med Biol. 2012;38(12):2065–71. https://doi.
org/10.1016/j.ultrasmedbio.2012.07.027.
24. Alvarez S, Añorbe E, Alcorta P, López F, Alonso I, Cortés J. Role of sonography in the diagno-
sis of axillary lymph node metastases in breast cancer: a systematic review. Am J Roentgenol.
2006;186(5):1342–8. https://doi.org/10.2214/AJR.05.0936.
25. Mano MS, Awada A. Primary chemotherapy for breast cancer: the evidence and the future.
Ann Oncol. 2004;15:1161–71.
26. Maur M, Guarneri V, Frassoldati A, Conte PF. Primary systemic therapy in operable breast
cancer: clinical data and biological fall-out. Ann Oncol. 2006;17 Suppl 5:158–64.
27. Hylton NM, Blume JD, Bernreuter WK, et al. Locally advanced breast cancer: MR imaging
for prediction of response to neoadjuvant chemotherapy—results from ACRIN 6657/I-SPY
TRIAL. Radiology. 2012;263(3):663–72.
28. Pediconi F, Miglio E, Telesca M, et al. Effect of preoperative breast magnetic resonance imaging
on surgical decision making and cancer recurrence rates. Investig Radiol. 2012;47(2):128–35.
29. Pettit K, Swatske ME, Gao F, et al. The impact of breast MRI on surgical decision-making: are
patients at risk for mastectomy? J Surg Oncol. 2009;100(7):553–8.
30. Le-Petross HT, Lim B. Role of MR imaging in neoadjuvant therapy monitoring. Magn Reson
Imaging Clin N Am. 2018;26:207–20.
31. Mann RM, Cho N, Moy L. Breast MRI: state of the art. Radiology. 2019;292:520–36.
32. Codari M, Schiaffino S, Sardanelli F, Trimboli RM. Artificial intelligence for breast MRI in
2008–2018: a systematic mapping review. AJR. 2019;212:280–92.
33. Goorts B, Dreuning KMA, Houwers JB, Kooreman LFS, Boerma EG, Mann RM, et al. MRI-
based response patterns during neoadjuvant chemotherapy can predict pathological (complete)
response in patients with breast cancer. Breast Cancer Res. 2018:20–34.
34. Hylton NM, Gatsonis CA, Rosen MA, Lehman CD, Newitt DC, Partridge SC, et al. Neoadjuvant
chemotherapy for breast cancer: functional tumor volume by MR imaging predicts recurrence-
free survival—results from the ACRIN 6657/CALGB 150007 I-SPY 1 TRIAL. Radiology.
2016;279(1):44–55.
35. Hylton NM. Residual disease after neoadjuvant therapy for breast cancer: can MRI help?
Radiology. 2018;289(2):335–6.
36. Kim TH, Kang DK, Yim H, Jung YS, Kim KS, Kang SY. Magnetic resonance imaging patterns
of tumor regression after neoadjuvant chemotherapy in breast cancer patients: correlation with
pathological response grading system based on tumor cellularity. J Comput Assist Tomogr.
2012;36:200–6.
37. Watanabe H, Okada M, Kaji Y, Satouchi M, Sato Y, Yamabe Y, et al. New response evaluation
criteria in solid tumours – revised RECIST guideline (version 1.1). JPN J Cancer Chemother.
2009;36(13):2495–501.
38. Kuhl CK, Alparslan Y, Schmoee J, Sequeira B, Keulers A, Brümmendorf TH, Keil S. Validity
of RECIST version 1.1 for response assessment in metastatic cancer: a prospective, multi-
reader study. Radiology. 2019;290(3):349–56.
39. Martincich L, Montemurro F, De Rosa G, et al. Monitoring response to primary chemotherapy
in breast cancer using dynamic contrast-enhanced magnetic resonance imaging. Breast Cancer
Res Treat. 2004;83(1):67–76.
16 Neoadjuvant Systemic Therapy 329
40. Ah-See ML, Makris A, Taylor NJ, et al. Early changes in functional dynamics magnetic reso-
nance imaging predict for pathologic response to neoadjuvant chemotherapy in primary breast
cancer. Clin Cancer Res. 2008;14(20):6580–9.
41. Dogan BE, Yuan Q, Bassett R, et al. Comparing the performances of magnetic resonance
imaging size vs pharmacokinetic parameters to predict response to neoadjuvant chemotherapy
and survival in patients with breast cancer. Curr Probl Diagn Radiol. 2019;48(3):235–40.
42. Partridge SC, Zhang Z, Newitt DC, et al. Diffusion-weighted MRI findings predict patho-
logic response in neoadjuvant treatment of breast cancer: the ACRIN 6698 multicenter trial.
Radiology. 2018;289(3):618–27.
43. Chu W, Jin W, Liu D, et al. Diffusion-weighted imaging in identifying breast cancer pathologi-
cal response to neoadjuvant chemotherapy: a meta-analysis. Oncotarget. 2017;9(6):7088–100.
44. Chen CA, Hayward JH, Woodard GA, Ray KM, Starr CJ, Hylton NM, et al. Complete breast
MRI response to neoadjuvant chemotherapy and prediction of pathologic complete response.
J Breast Imaging. 2019;1(3):217–22.
45. Santamaría G, Bargalló X, Fernández PL, Farrús B, Caparrós X, Velasco M. Neoadjuvant systemic
therapy in breast cancer: association of contrast-enhanced MR imaging findings, diffusion-weighted
imaging findings, and tumor subtype with tumor response. Radiology. 2017;283(3):663–72.
46. US Department of Health and Human Services Food and Drug Administration Center for Drug
Evaluation and Research. Guidance for industry: Pathological complete response in neoad-
juvant treatment of highrisk early-stage breast cancer (version 2020). Accessed October 1,
2020. Available at: http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/
Guidances/default.htm
47. Reig B, Heacock L, Lewin A, Cho N, Moy L. Role of MRI to assess response to neoadju-
vant therapy for breast cancer. J Magn Reson Imaging. 2020;52(6). https://doi.org/10.1002/
jmri.27145.
48. Kim SY, Cho N, Park IA, et al. Dynamic contrast-enhanced breast MRI for evaluating residual
tumor size after neoadjuvant chemotherapy. Radiology. 2018;289(2):327–34.
49. Kim Y, Sim SH, Park B, et al. Magnetic resonance imaging (MRI) assessment of residual
breast cancer after neoadjuvant chemotherapy: relevance to tumor subtypes and MRI interpre-
tation threshold. Clin Breast Cancer. 2018;18(6):459–67. e451
50. Negrão EMS, Souza JA, Marques EF, Bitencourt AGV. Breast cancer phenotype influences
MRI response evaluation after neoadjuvant chemotherapy. Eur J Radiol. 2019;120:108701.
51. Ballesio L, Gigli S, Di Pastena F, et al. Magnetic resonance imaging tumor regression shrink-
age patterns after neoadjuvant chemotherapy in patients with locally advanced breast cancer:
correlation with tumor biological subtypes and pathological response after therapy. Tumour
Biol. 2017;39(3):1010428317694540. https://doi.org/10.1177/1010428317694540.
52. Rosen EL, Blackwell KL, Baker JA, et al. Accuracy of MRI in the detection of residual breast
cancer after neoadjuvant chemotherapy. AJR Am J Roentgenol. 2003;181(5):1275–82.
53. Lorenzon M, Zuiani C, Londero V, et al. Assessment of breast cancer response to neoadjuvant
chemotherapy: is volumetric MRI a reliable tool? Eur J Radiol. 2009;71(1):82–8.
54. Schrading S, Kuhl CK. Breast cancer: influence of Taxanes on response assessment with
dynamic contrast-enhanced MR imaging. Radiology. 2015;277(3):687–96.
55. Chen JH, Bahri S, Mehta RS, et al. Impact of factors affecting the residual tumor size diag-
nosed by MRI following neoadjuvant chemotherapy in comparison to pathology. J Surg Oncol.
2014;109(2):158–67.
56. Kopans DB. Breast imaging. 3rd ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007.
57. Shin HJ, Kim HH, Ahn JH, et al. Comparison of mammography, sonography, MRI and clinical
examination in patients with locally advanced or inflammatory breast cancer who underwent
neoadjuvant chemotherapy. Br J Radiol. 2011;84(1003):612–20.
58. Kawashima H, Inokuchi M, Furukawa H, Kitamura S. Triple-negative breast cancer: are the
imaging findings different between responders and nonresponders to neoadjuvant chemother-
apy? Acad Radiol. 2011;18(8):963–9.
59. Boughey JC, Suman VJ, Mittendorf EA, et al. Sentinel lymph node surgery after neoadjuvant
chemotherapy in patients with node-positive breast cancer: the ACOSOG Z1071 (Alliance)
clinical trial. JAMA. 2013;310(14):1455–61.
330 A. C. de Ataíde Góes et al.
For the proper examination to be performed, the radiologist and technologist must be
aware of pertinent aspects of the patient’s history, such as signs and symptoms, locations
of masses, pain, and prior surgeries that the patient must indicate in a diagram. Extensive
surgical procedures, such as mammoplasty, must be shown diagrammatically only,
without the use of skin markers. Smaller scars are marked with pieces of thin wire that
are taped to the skin to correlate with possible mammographic findings (Fig. 17.1), but
it may not always correlate with the surgical excision site located deeper in the breast.
The entire surgical area must be included on at least one mammographic view and, if
necessary, additional views, such as spot compression, magnification, and tangential
and various angulations, may be useful and should be performed in the projection where
the area is better and more completely seen. Diagnostic accuracy increases with aware-
ness of how procedures were performed, knowledge of time since surgery, and breast
density (detection and interpretation are more difficult in dense breasts).
Normal postoperative imaging findings include architectural distortion, increased
density and parenchymal scarring, and decrease in severity over time, more rapidly
L. M. Yano
Hospital Albert Einstein, São Paulo, SP, Brazil
Instituto de Radiologia INRAD, Hospital das Clinicas HCFMUSP, Faculdade de Medicina,
Universidade de Sao Paulo, São Paulo, SP, Brazil
e-mail: [email protected]
M. A. Rudner (*)
Hospital Albert Einstein, São Paulo, SP, Brazil
Instituto de Radiologia INRAD, Hospital das Clinicas HCFMUSP, Faculdade de Medicina,
Universidade de Sao Paulo, São Paulo, SP, Brazil
Hospital Moriah and Prevent Senior, São Paulo, SP, Brazil
e-mail: [email protected]
a b c
Fig. 17.1 Postoperative scarring. Mammography shows thin wire taped to the skin marking the
cutaneous scar to correlate with possible mammographic findings (a, b). Ultrasound depicted a
hypoechoic irregular area with architectural distortion, with different morphologic aspects in dif-
ferent planes, representing a scar (c, d)
and completely in benign biopsies (Fig. 17.2) or aesthetic procedures (Fig. 17.3).
Immediately after the procedure, mammography will show a round/oval mass or
asymmetric soft-tissue density in the postoperative site representing seroma or
hematoma, with or without air in the biopsy cavity. The incidence of postsurgical
fluid collections reflects the surgical technique. Breast edema usually occurs close
to the incision area and may last for the first 1 or 2 months. Outlined by subcutane-
ous fat, linear parenchymal trabeculations extend toward the skin, representing
engorged lymphatics and interstitial fluid. Depending on the extension of the sur-
gery, the breast enlarges, and mammographic compression is more difficult. As the
edema recedes, the breast size also normalizes. Skin thickening and breast edema
are usually seen together and have similar time courses and resolution. Contralateral
asymmetries may be explained by the absence of equivalent tissue on the operative
side and can be mistaken as an abnormality. If there is doubt, sonography can iden-
tify the fluid-filled nature of the mass (Fig. 17.4). Initially, some hematomas show
internal echoes but most soon become anechoic with posterior acoustic enhance-
ment and may present septa (which do not mean complications). The incision can
occasionally be traced from the biopsy cavity to the skin, which is thickened
(Fig. 17.2). Management of a complex mass requires knowledge of the clinical con-
text. If an abscess is not suspected, observation may be preferred [1].
17 Postoperative Breast 333
a b d e
Fig. 17.2 Normal postoperative aspect. Mammogram 1 month after benign biopsy depicts skin
thickening, architectural distortion, increased density, and parenchymal scarring (a, b). Ultrasound
of the same date shows architectural distortion and scarring of the incision from the biopsy cavity
to the skin, which is thickened (c). Late control shows that these findings had diminished over
time (d, e)
In very rare cases, seroma cavities persist 1 year after a benign biopsy [1].
Generally, after a few weeks to 18 months, the fluid collection is gradually reab-
sorbed; the lesion decreases in size and becomes poorly defined. Areas of radiolu-
cency representing fat entrapped by the developing scar are seen interspersed with
soft-tissue density [1]. Scars change appearance in different projections: a spicu-
lated mass like soft tissue in one view may elongate in other projections. After 1 or
2 years, an evolving scar contracts and shrinks as it matures, and architectural dis-
tortion or a spiculated density may be seen. On sequential mammograms, decrease
in size may be barely perceptible or seen in only one projection. It is important to
note that the linear metallic scar marker on the skin will not always be immediately
adjacent to the surgical bed because the skin is pressed away from the underlying
parenchyma. Preoperative mammograms should always be reviewed, fundamen-
tally if a spiculated mass is found far from the marker, and biopsy may be necessary
to eliminate the possibility of cancer. Ultrasound reveals a hypoechoic spiculated
334 L. M. Yano and M. A. Rudner
Fig. 17.3 Increased density and parenchymal scarring after liposuction on the axilla (normal
findings)
a b
Fig. 17.4 Postsurgical seroma/hematoma. Ultrasound depicts the fluid-filled nature of the mass
with the surgical path through the breast tissue up to the skin and hyper-echogenicity of the sur-
rounding parenchyma (a). A 1-year control (b) depicts complete resolution of the fluid collection
with architectural distortion and parenchymal scarring, as well as skin thickening and retraction
mass, and, occasionally, retraction of the scar skin is noted (Figs. 17.1 and 17.5).
History and physical findings in the skin can help distinguish this normal postopera-
tive finding from cancer. Scarring not complicated by fat necrosis is perceived as
induration rather than a mass. The period of change is variable and does not depend
on the type of breast parenchyma, but some aspects can induce the rate of scar for-
mation, such as resection size, postsurgical fluid collection volume, and whether or
17 Postoperative Breast 335
not it was drained. After 3 to 5 years, between 50 and 55% of cases show complete
resolution of the biopsy cavity, leaving no scar or distortion in the underlying breast
parenchyma [2]. The findings should be stable on subsequent mammograms.
To evaluate calcifications at the surgical site, the same morphological and distri-
butional features used in the preoperative period should be applied to classify the
probability of malignancy. In early stages, they may be quite fine, faint, and difficult
to characterize, usually becoming coarser later on. Dystrophic calcifications may
have similar pathogenesis of secretory disease or ductal ectasia, associated with
areas of necrotic tissue, sloughed cells, and cellular detritus, evolving into scars and
in the subcutaneous tissue under surgical incisions. Tangential mammographic
views to the skin can demonstrate their superficial locations. Calcified remnants of
suture material can also be seen, with several millimeters long and quite wide, pre-
senting as knots, branching linear and double tracking calcifications. If the benign
nature of these calcifications cannot be determined, biopsy is indicated [1].
The thickened skin in the incision may be superimposed on the surgical area
causing a mass-like increased density, particularly when a keloid has formed. A
complementary tangential view to the scar permits separation of the skin and paren-
chymal elements, so the surgical bed can be better accessed. Sonographically, a
band of thickened skin will be seen in the incision [1].
Fat necrosis originates from aseptic fat saponification and is associated with all
types of surgical procedures in the breast. It usually appears as a radiolucent lipid-
filled mass. When symptomatic, fat necrosis may present as a lump. In earlier
phases, a hematoma may be present. In all types of imaging methods, it is a com-
mon finding and sometimes a challenging pitfall, manifesting different presenta-
tions that may be indistinguishable from cancer, depending on the stage of the
process (Fig. 17.6). Mammography is pathognomonic if it shows oily cysts or typi-
cal eggshell rim calcifications around a mass with radiolucent center. There is a
wide range of sonographic presentations that include solid mass, complex mass
with mural masses, complex mass with echogenic bands, anechoic mass with pos-
terior acoustic enhancement, anechoic mass with shadowing, or an isoechoic mass.
The margins of the mass vary from well circumscribed to indistinct or spiculated.
336 L. M. Yano and M. A. Rudner
a b c
d e
Fig. 17.6 Fat necrosis. MRI shows an irregular mass, hypointense on axial T1-weighted image
without fat saturation (a) and hyperintense on axial fat-saturated T2-weighted image (b). Axial
contrast-enhanced fat-suppressed T1-weighted MR subtraction image (c) and sagittal contrast-
enhanced fat-suppressed T1-weighted image (d) show homogeneous enhancement. Ultrasound
depicts an irregular, ill-defined, heterogeneous mass (e). Core needle biopsy was consistent with
fat necrosis
a b c
d e
Fig. 17.7 Fat necrosis. Ultrasound shows an oval, circumscribed, and heterogeneous mass (a).
Axial non-fat-saturated T1-weighted (b), axial fat-saturated T2-weighted (c), sagittal contrast-
enhanced fat-suppressed T1-weighted (d), and axial contrast-enhanced fat-suppressed T1-weighted
MR subtraction images (e) show a T1-hyperintense mass with signal loss on the fat-saturated
images and thin discrete peripheral enhancement
a b c
Fig. 17.8 Fat necrosis. Axial T1-weighted image without fat saturation (a), axial fat-saturated
T2- weighted (b), and axial contrast-enhanced fat-suppressed T1-weighted MR subtraction images
(c) show a T1-hyperintense mass with signal loss on the fat-saturated images and thin discrete
peripheral enhancement
338 L. M. Yano and M. A. Rudner
Fig. 17.9 T1-weighted MR images without fat suppression show architectural distortion and scar-
ring on mammoplasty bed (arrows)
enhancement, and thin linear non-mass-like enhancement at the surgical area can all
be expected findings. Non-enhancing areas have a high negative predictive value for
malignancy (88–96%). Postoperative seromas have high signal intensity (fluid-
filled structure signal) on T2-weighted images and smooth, thin rim enhancement.
Benign postoperative changes generally demonstrate more gradual uptake of con-
trast material. Breast hematomas show a varying signal according to the hemoglo-
bin degradation status (high signal on T1-weighted at subacute stage, low signal on
T1/T2-weighted at chronic stage). As they dissipate, fibrous tissue in the site of the
fluid collection can appear as an architectural distortion. Scarring enhancement (a
minimal or small focal area of enhancement or thin linear enhancement) can be seen
for up to 18 months or even longer. At follow-up imaging, the enhancement should
demonstrate stability or progressive decrease [2].
Mammoplasty and mastopexy are different procedures; the central focus of mam-
moplasty is breast volume reduction (most often performed for macromastia) and
for mastopexy is breast shape. Nevertheless, there are many similarities regarding
the techniques used and also postoperative imaging aspects. The multiplicity of
surgical techniques, with different accesses (inframammary and peri-areolar fold
access; peri-areolar, vertical, and horizontal (inverted T); peri-areolar and vertical;
17 Postoperative Breast 339
peri-areolar, vertical and lateral horizontal; isolated peri-areolar), evidence the chal-
lenge to achieve a harmonious and symmetrical aesthetic result, with minimal scar-
ring. The resulting surgery leaves a smaller breast with normal skin visible in the
region of the cleavage. Both surgical procedures may be performed concurrently
with breast conservation therapy. Awareness of normal postoperative imaging fea-
tures of these oncoplastic procedures is important to avoid false-positive results.
The most common surgical procedure involves a circumareolar incision, an
inframammary incision, and a vertical incision between the two (traditional inverted-
T scar, or Wise pattern), with removal of the skin, breast parenchyma, and fat, pre-
dominantly from the lower portion, followed by superior relocation of the nipple
(Fig. 17.9). This will result in elevation of the nipple-areolar complex (NAC) and
reduction of the cutaneous envelope of the breast. With the advent of smaller skin-
incision techniques, new vertical scar techniques feature only one vertical incision
leaving a lollipop-shaped scar, compared with the anchor-shaped appearance of the
inverted-T scar. These smaller skin-incision techniques decrease scarring and dis-
tortion of the inframammary fold, allowing parenchymal rearrangement and pedicle
creation, which maintains blood supply to the NAC [3].
Usually, the nipple is kept attached to the breast ducts, but in very ptotic or very
large breasts, the surgeon may choose to remove the nipple and then graft it back
into the breast, which may increase the risk of nipple necrosis and difficulty breast-
feeding. The new nipple site may have little parenchyma behind it and may result in
a relative shift, resulting in transposition of parenchyma to the dependent portion of
the breast, with nonanatomic distribution, linear strands, parenchymal bands, and
calcifications. Depending on the amount of tissue removed from different areas of
the breast, redistribution of glandular tissue occurs, and breast parenchymal pattern
can be patchy [4]. This fact, associated with scarring, makes comparison with previ-
ous images difficult.
Postoperative mammograms show characteristic skin thickening in the region of
the scars, generally over the lower breast, which is most evident on the mediolateral
oblique or mediolateral view. The lower breast shows architectural distortion, and
its general pattern is distorted from the scar (Fig. 17.10). Fibrous bands can be seen
extending to the repositioned NAC in 20% of the patients, representing scarring
associated with the vascular pedicle (Fig. 17.11) [3]. Fat necrosis or oil cysts are
common findings and may have a characteristic appearance or may be atypical,
forming a palpable mass when symptomatic. Epidermal inclusion cysts can also
form in scars and produce a smooth dense round or oval mass close to the skin sur-
face but not connected to it. They represent epidermal cells displaced into breast
tissue during surgery. Skin calcifications along incisions can be distinguished from
intraparenchymal calcifications or cancer because of their typical skin location.
Periareolar dermal calcifications and thickening related to the repositioned NAC
can be seen; therefore, imaging of the nipple in profile is critical (Fig. 17.12) [3].
Recalls may be higher in this group of patients due to surgical architectural distor-
tions and possible areas of fat necrosis with atypical presentations, but tomosynthe-
sis can resolve some doubts and reduce recall rates.
340 L. M. Yano and M. A. Rudner
Fig. 17.10 Mammoplasty. Mammography shows architectural distortion in the lower quadrants,
and breast overall pattern is distorted from the scar
Fig. 17.11 Mastopexy. Fibrous bands extending to the repositioned NAC, representing scarring
associated with the vascular pedicle
Aesthetic surgery rates are raising worldwide, and the use of silicone breast implants
is the main procedure used in breast augmentation. They were introduced in 1962
by Cronin and Gerow, presenting several modifications in the design, texturing, and
gel cohesiveness over subsequent generations [5]. Approximately 80% is placed for
cosmetic reasons and the other 20% for breast reconstruction after mastectomy [6].
The silicone implants can be positioned via periareolar, axillary, transpapillae, or
inframammary incisions. All implants are placed behind the breast tissue, and sur-
geons will choose the ideal place regarding the pectoralis muscle, which may be
subglandular, subfascial, submuscular, or double plane (Figs. 17.13 and 17.14).
17 Postoperative Breast 341
Breast implants are not lifetime devices [7, 8]. The older the implants, the more
likely they will require removal or substitution [9, 10]. There is no current evidence
that breast augmentation with silicone implants may present increased risk for
developing connective disorders, breast cancer, no proven delay in breast cancer
detection, and no difference in survival or recurrence rates compared with women
with no augmentation [11–18].
Breast implants are classified according to its material, shape, surface texturing, and
number of chambers.
Saline implants are composed of an outer silicone shell and an inner envelope
filled with saline solution. Some are pre-filled, and others can be filled during the
surgical procedure. Single-lumen silicone implants are the most common type and
consist in a silicone elastomer shell filled with silicone made from a synthetic poly-
mer of cross-linked chains of dimethyl siloxane. The inner silicone can have a liq-
uid, gel, or solid consistency depending on the length of the individual chains and
the degree to which these chains are cross-linked.
342 L. M. Yano and M. A. Rudner
a b c
d e f
Fig. 17.13 Submuscular implant. Mammogram (a, b), ultrasound (c, d), and MRI (e, f) show the
pectoralis muscle (arrows) over the implant
Saline and silicone implants are either round or anatomic (teardrop) in shape,
and the outer envelope can be textured or smooth (uncoated). Textured coating sta-
bilizes the implant within the breast pocket, reducing the risk of rotation and result-
ing in lower rates of capsular contraction, decreasing the risk of a secondary
procedure. Early texturized implants were coated with polyurethane foam, but they
were withdrawn from the market due to concerns about in vivo degradation to car-
cinogenic compounds [5]. Therefore, implant shells started to be texturized mechan-
ically, creating pores of different sizes [19].
Double-lumen implants/expanders have two envelopes inside one another, and
each can contain saline or silicone gel. It is common to find saline filling the inner
lumen and silicone in the outer lumen, but the opposite can also occur (Fig. 17.15).
Tissue expanders present different sizes and shapes and may have a smooth or tex-
tured outer surface. They are inserted under the breast skin, autologous tissue, or
pectoralis muscle, immediately following mastectomy reconstruction or in subse-
quent surgery months or years later. As soon as possible, the tissue expander is
slowly inflated through a series of saline solution injections or through a patient-
controlled device, which releases carbon dioxide gas into the expander. The objec-
tive is to stretch the patient’s tissue for insertion of an implant or the patient’s own
tissue as part of the reconstruction process (after mastectomy, to reconstruct injured
or congenitally deformed breasts, or as part of gender reassignment surgery).
17 Postoperative Breast 343
a b c
d e
Fig. 17.14 Subglandular implant. Mammogram (a, b), ultrasound (c), and MRI (d, e) show the
pectoralis muscle (arrows) behind the implant
Stretching may result in breast tissue damage, skin thinning, pain (especially during
saline filling), rupture, and infection.
Motiva implants present a 3D imprinted surface texturing. Radiofrequency iden-
tification transponders are attached to it and can transmit data about the implant
wirelessly. They have a linear radiopaque appearance on mammography and CT
(Figs. 17.16 and 17.17) and produce an important magnetic susceptibility artefact
on MRI (Fig. 17.17).
Stacked implants are two single-lumen, smooth-surface silicone gel-filled
implants placed one on top of the other, firmly joined in the middle. Both have dif-
ferent volumes, the smaller representing 20% of the entire volume and positioned
anteriorly. Each one has its own separate shell so the filling of each portion is
not shared.
344 L. M. Yano and M. A. Rudner
a b c d
e f g h
Fig. 17.15 Double-lumen implants/expander. The inner lumen is filled with saline surrounded by
a smaller outer lumen that contains silicone. Mammogram shows a double contour with a low-
density inner lumen and a high-density outer lumen (a, b). Ultrasound depicts the inner envelope
with undulations that may be mistaken for the stepladder sign (c, d). Axial T1-weighted without fat
suppression (e), axial fat- suppressed T2-weighted (f), axial short T1 inversion-recovery silicone-
excited image (g), and sagittal contrast-enhanced fat-suppressed T1-weighted MR images (h)
demonstrate the inner and outer lumens with different signal intensities, depending on the pulse
sequence
a b c
d e f
Fig. 17.17 Motiva implant. Important magnetic susceptibility artefact on T1-weighted image
without fat suppression (a), short T1 inversion-recovery silicone-excited image (b), fat-suppressed
T2-weighted (c), and contrast-enhanced fat-suppressed T1-weighted MR images (d) related to
Motiva’s transponder. Radiofrequency identification transponder presents a linear radiopaque
appearance on CT (e, f)
a b c d
e f g h
Fig. 17.18 Modified views for implants (Eklund maneuver). Projections of each breast obtained
with routine positioning show the implants contours and only a small amount of breast tissue (a–
d). Projections of each breast after the implant has been displaced posteriorly and the anterior
breast tissue pulled into the compression plates demonstrate more parenchyma (e–h)
implant-displaced views, and only about 80% of the breast tissue is seen, raising the
concern of a possible reduction in detectability of breast (Fig. 17.19) [21–25].
The recommendation for breast cancer screening is the same as that for patients
with no implants. Presurgical and postsurgical mammograms are useful in distin-
guishing benign postoperative changes from other breast pathologies [25]. Limited-
compression views may display a lesion near the implant not evidenced on
implant-displaced views, because masses on the fibrous capsule can be pushed
away from the field of view after Eklund maneuver [22]. Additional views or tomo-
synthesis may be useful for evaluating abnormalities in patients with implants. All
types of percutaneous biopsies and presurgical localization can be performed in the
presence of implants as well, but an informed consent should be obtained, including
implant rupture as a possible complication [24].
To evaluate intracapsular rupture of implants and to detect other complications
such as hematomas and seromas, ultrasound is an important tool. With the exception
17 Postoperative Breast 347
a b c d e f
Fig. 17.19 Reduction in detectability of breast carcinoma in augmentation. Spiculated mass hid-
den by the implant (a, b), the saline filler diminished the risk of overlooking this lesion, but it is
important to note that on the implant-displaced views (c, d), it was very difficult to diagnose the
mass. Tomosynthesis (e) shows the irregular spiculated mass in a very posterior area that could
easily have been lost with a bad positioning. Ultrasound (f) confirmed the irregular mass
of cases with dense breast tissue, in which ultrasound may increase detection of
masses obscured by implants, there is no specific screening protocol that includes
this imaging test. Ultrasound is also helpful as an adjunct to mammography in eval-
uating detected breast masses or palpable findings, as it can evaluate the entire depth
of the breast tissue down to the implant, and it may distinguish breast masses from
silicone granulomas caused by ruptured implants. Ultrasound-guided procedures
also reduce the chance of implant perforation.
The US Food and Drug Administration (FDA) recommends evaluating implant
integrity with magnetic resonance imaging (MRI) 3 years after surgery and every 2
years thereafter to depict silent ruptures (asymptomatic) [3, 26]. MRI is considered
the gold standard in evaluating implant integrity and should be performed whenever
mammography and ultrasound detect any abnormality. Regarding screening, MRI
has the same recommendations according to age and risk factors. A dedicated breast
coil obtains high-resolution images, and it is possible to suppress or emphasize the
signal from water, fat, or silicone [27]. Saline implants follow fluid signal on all
sequences. Silicone appears hypointense on T1-weighted image and hyperintense
on T2-weighted image (Fig. 17.20). The envelope and fibrous capsule have low
signal on all sequences. Like other imaging methods, radial folds and periprosthetic
fluid are normal findings and should not be mistaken for rupture (Fig. 17.21).
On mammography, silicone implant appears near the chest wall and behind the
breast tissue as a dense, smooth, oval, and completely opaque image that partially
obscures the surrounding breast tissue. On the other hand, saline implants are radio-
lucent in the center, surrounded by a dense silicone outer envelope, which makes
wrinkles easily detected, unlike on opaque silicone implants. Wrinkles are normal
findings, accentuated by compression because the envelope is easily folded but are
sometimes related to capsular contraction (Fig. 17.22). Double-lumen implants are
a common type of implant, usually filled with saline in the inner lumen and silicone
in the outer lumen. The subglandular position consists in the implant behind the
348 L. M. Yano and M. A. Rudner
a d
Fig. 17.20 Normal aspects of silicone implants on MRI. Silicone appears hypointense on
T1-weighted image without fat suppression (a) and on short T1 inversion-recovery silicone-excited
images (b) and hyperintense sign on fat-suppressed T2-weighted MR images (c, d)
a b
Fig. 17.21 Fluid inside a radial fold (normal aspect). Axial short T1 inversion-recovery silicone-
excited image (a) and axial fat-suppressed T2-weighted MR image (b)
17 Postoperative Breast 349
a b
Fig. 17.22 Wrinkles on the surface of the implant on mammogram (normal finding)
glandular tissue, overlapping the pectoralis muscle, whose anterior contour will be
underneath the implant on mammography, mostly on the mediolateral oblique view.
On the other hand, in the subpectoral position, the implant is behind the breast tissue
and the pectoralis muscle, which covers the implant [23].
A fibrous capsule is always formed around the implant, usually soft and unde-
tectable by physical examination or mammography, unless it hardens or calcifies
[28]. Dystrophic sheetlike calcifications appear dense, thin, and irregular next to the
implant (Figs. 17.23 and 17.24) [29]. Polyurethane-covered implants are covered
with a spongelike material and, when calcified, produce a typical fine mesh-like
calcification. Implant-displaced views displace the capsular calcifications away
from the implant (Fig. 17.25) [22]. Spot magnification can help differentiate if the
calcifications are in the parenchyma (which may represent postoperative fibrosis or
a more worrisome diagnosis) or in the implant capsule. Sometimes this
350 L. M. Yano and M. A. Rudner
a b
Fig. 17.23 Calcified fibrous capsule of a saline implant after 22 years of placement
differentiation is not possible, and capsular calcifications can be mistaken for malig-
nancy, leading to a false-positive examination [29].
Ultrasound cannot differentiate between silicone or saline implant types, but in
double-lumen implants, it is possible to characterize the inner and outer envelopes
as two distinct chambers. The normal implant (Fig. 17.26) is an anechoic mass with
an echogenic shell, a three-layered appearance, two thin echogenic lines, and an
anechoic line between them (“Oreo cookie sign”) [30]. Reverberation artifacts can
be identified as multiple bands of noise caused by repeat reflections of the beam
between the skin surface and the anterior wall of the implant, characteristically seen
in the near field, parallel to the anterior implant wall, showing the same width as the
17 Postoperative Breast 351
a b
Fig. 17.24 Calcified fibrous capsule of a silicone implant. Dystrophic sheetlike calcifications
appears dense, thin, and irregular next to the implant
a b c d
Fig. 17.25 Capsular calcifications (c, d) displaced away from the implant after Eklund maneuver
352 L. M. Yano and M. A. Rudner
Fig. 17.26 Normal implant on ultrasound. Anechoic mass with an echogenic shell with a three-
layered appearance with two thin echogenic lines (thin arrows) and an anechoic line between them
(“Oreo cookie sign”). Reverberation artifacts (star) and the posterior localization of the seal
(thicker arrow) are also characterized
breast tissue anterior to the implant (Fig. 17.27) [23, 28]. Radial folds are common
findings and represent infoldings of the intact envelope that look like echogenic
lines extending to the periphery of the envelope [30]. The fibrous capsule may be
seen as another echogenic line superficial to the implant shell separated by a small
anechoic line. Implants that have a sponge structure of the outer shell present the
fibrous capsule growth into this coverage; therefore, it cannot be seen. The clue that
helps to evidence the fibrous capsule is the search for a small radial fold, in which
the fibrous capsule is usually lifted from the implant and easy to measure (Fig. 17.28).
17 Postoperative Breast 353
Fig. 17.28 Fibrous capsule. Ultrasound shows small radial folds filled with fluid. In these spots,
fibrous capsule is usually lifted from the implant and easy to measure
Small radial folds and a small amount of periprosthetic fluid are also considered
normal findings (Fig. 17.29) [31].
The main reasons patients decide to remove their implants are suspected silicone-
related health problems; suspected rupture; breast firmness; breast and musculo-
skeletal pain; or other complications. Mammography, ultrasound, and magnetic
resonance imaging after explantation are broad and may simulate malignant tumors.
In this context, the comparison with previous exams and a detailed surgical history
are crucial. The imaging findings range from a nearly normal appearance to
354 L. M. Yano and M. A. Rudner
Fig. 17.30 Explantation. There are no signs of the implant removal, except for the discrete archi-
tectural distortion
a b c e
Fig. 17.31 Explantation. Mammogram (a, b) and CT (d, e) show coarse calcifications in the
residual fibrous capsule after implants removal. On ultrasound (c), the fibrous capsule presents as
a hypoechoic mass next to the thoracic wall with a hyperechoic image in the center that represents
the calcification
17 Postoperative Breast 355
a b c d e
Fig. 17.32 Explantation. Mammogram performed 1 month after explantation depicts architectural
distortion associated with a spiculated mass and calcifications (a, b). Ultrasound of the same date
shows the distortion as a hypoechoic path from the fluid filled implant cavity up to the NAC (c).
Late control mammogram shows an almost completely resolved cavity with a discrete architectural
distortion (d, e)
typically presenting as a sheetlike curvilinear pattern at the chest wall (Fig. 17.34)
but occasionally can show an undetermined pattern, which can be hard to distin-
guish from other breast pathologies (Fig. 17.35) [4, 29, 33].
It is usually difficult to remove all the extracapsular free silicone from a ruptured
implant, without removing too much of the breast [28]. In this case, some free sili-
cone images, infiltrated lymph nodes (Fig. 17.34), or silicone in the residual fibrous
capsule (Fig. 17.36) can be seen [4]. Ultrasound shows the typical snowstorm sign,
and the free silicone signal on MRI should not be misinterpreted as an acute rupture
of new implants [32].
a b
c d
Fig. 17.33 Explantation. Axial T1-weighted without fat suppression (a), axial fat- suppressed
T2-weighted (b), and axial contrast-enhanced fat-suppressed T1-weighted MR images (c) depict
the implant cavity after 2 months of implant removal filled with fluid in a non-collapsed fibrous
capsule, mimicking an abscess. Ultrasound shows a seroma with thin septa (d)
a b c e
d f
a b
a b c d f
Fig. 17.36 Explantation. Mammogram (a–c) shows a dense residual fibrous capsule with an asso-
ciated small spiculated density (arrow). An irregular linear image with posterior shadowing is seen
anteriorly to the implant on ultrasound (d). On axial fat- suppressed T2-weighted (e) and axial
short T1 inversion-recovery silicone-excited MR images (f), this image shows signal compatible
with silicone from previous ruptured implant
a c d
e
b
Fig. 17.37 Infection. Axial fat-suppressed T2-weighted (a) and axial contrast-enhanced fat-
suppressed T1-weighted subtraction image (b) and sagittal contrast-enhanced fat-suppressed
T1-weighted MR images (c) show fibrous capsule thickening with enhancement. Patient had
inflammatory signs with fistulization of purulent content in the upper quadrants which resulted in
skin retraction. Ultrasound depicts a small collection with anfractuous contours next to the implant
and hyper echogenicity of the nearby tissue (d, e)
17 Postoperative Breast 359
a b
c d
Fig. 17.38 Infection. Axial T1-weighted without fat suppression (a), axial fat-suppressed
T2-weighted (b), axial short T1 inversion-recovery silicone-excited (c), and sagittal contrast-
enhanced fat-suppressed T1-weighted MR images (d) show periprosthetic effusion and fibrous
capsule thickening with enhancement
Rotation
Imaging diagnosis can be made by searching for the raised silicone ridge/projec-
tions (Fig. 17.39) used for intraoperative orientation of the implant that usually are
placed between 5 and 7 o’clock, easily visualized on ultrasound. In implants that do
not have an orientation marker, we must look for the posterior seal (Fig. 17.40). If it
is visible on the anterior part of the envelope, it is compatible with rotation
(Fig. 17.41). MRI is a useful tool since it can demonstrate any localization of the
seal, not only anterior or posterior (Fig. 17.42).
Capsular Contraction
Capsular contraction is the most common implant complication [36], with a reported
incidence of more than 70% in older series and about 20% in more recent literature
(lower rates of capsular contracture have been observed with later-generation,
a b
Fig. 17.39 Raised silicone ridge. Ultrasound shows the orientation marker on longitudinal (a) and
transversal position (b) as a hypoechoic periprosthetic layer (arrows)
Fig. 17.40 Posterior seal. Normal positioning of the implant can be confirmed by the posterior
location of the seal that is characterized by its echogenic linear appearance (arrows) separated from
the envelope by a thicker anechoic line
17 Postoperative Breast 361
Fig. 17.41 Rotation. Ultrasound of different implants show the posterior seal with an anterior
location
a b c
Fig. 17.43 Bilateral capsular contraction. Increased anteroposterior diameter due to the spherical
shape. It is very difficult to suggest a bilateral diagnosis only with mammogram. Clinical history
is critical in these cases
a b
Fig. 17.44 Unilateral capsular contraction on ultrasound. Increased anteroposterior diameter due
to the spherical shape (a) is better seen when compared to the other side (b)
a d
Fig. 17.45 Unilateral capsular contraction on MRI. Patient with induration of the right breast.
Axial T1-weighted without fat suppression (a), axial short T1 inversion-recovery silicone-excited
image (b), axial contrast-enhanced fat-suppressed T1-weighted subtraction (c), and sagittal
contrast-enhanced fat-suppressed T1-weighted MR images (d) show bilateral rotation with
increased anteroposterior diameter due to the spherical shape and enhancement of the fibrous cap-
sule of the right implant
364 L. M. Yano and M. A. Rudner
Implant Rupture
Breast implant ruptures consists in partial or total collapse of the envelope, mostly
associated with trauma, closed capsulotomy, and older generation implants [20, 37,
38]. Newer generations of silicone implants are more stable and have rupture rates
estimated in 2.0–3.8% in 6-year follow-up (fifth-generation implants) [3, 5, 8, 38–
41]. Implant type (ruptures in double-lumen and polyurethane-covered implants are
less frequent than in smooth implants) and manufacturers also have influence in risk
factors [5, 36]. Poly Implant Prothèse (PIP) silicone implants have been responsible
for a crisis in this growing aesthetic field, by containing nonmedical industrial sili-
cone and a higher reported rupture rate.
When a saline implant ruptures, the envelope retracts, and deflation is usually
noticeable as the breast becomes smaller [23, 37]. The breast tissue absorbs the
saline solution with no major complications.
The silicone implant usually keeps the original shape and volume if the fibrous
capsule is intact and continues to contain the free silicone (intracapsular rupture).
Intracapsular ruptures account for 77–89% of ruptures, and the patient is usually
asymptomatic. These are also called silent ruptures and occurs in 5% of screening
patients [21].
The collapse of the silicone implant envelope may be associated with a rupture
of the fibrous capsule, with the extrusion of the silicone gel into the breast tissue,
which are not absorbed by the body (extracapsular rupture).
Gel bleed is a situation in which silicone gel leakage is observed through an
apparent intact implant envelope (the existence of undetectable ruptures remains
controversial).
Diagnosis of silicone implant rupture is often difficult based only on clinical
manifestations because they are frequently nonspecific [5, 42]. Reported symptoms
include palpable masses in the axilla, breast, or chest wall (older generations); pain;
or changes in the size, shape, or texture of the breast [3, 28, 43, 44]. Imaging assess-
ment plays a main role in the detection of implant ruptures because physical exami-
nation misses approximately 50% of them [6, 42].
17 Postoperative Breast 365
Mammography
Intracapsular ruptures in implants filled with silicone gel are almost undetectable in
mammograms because the silicone gel is still contained within an intact fibrous
capsule [28, 45]. A flared contour may be related to intracapsular rupture (Fig. 17.46)
but also to capsular contracture or herniation of an intact implant envelope through
a rupture in the surrounding fibrous capsule [44].
Silicone beyond the fibrous capsule is a typical and specific finding of an extra-
capsular rupture (Fig. 17.47) [45, 46]. However, mammograms may show only con-
tour abnormality of the ruptured implant when the extruded silicone is not far
enough from the implant to be seen as a separate finding (Fig. 17.48). Eventually
extracapsular rupture may have no apparent abnormality on mammograms, and the
diagnosis must be confirmed by either ultrasound or MRI [44, 47].
Dense axillary lymph nodes are rare and represent silicone infiltration in the
lymphatics, highly suggestive of an extracapsular rupture [23, 28, 48].
It is impossible to remove all of the free silicone without removing too much of
the breast tissue after removal of an implant with extracapsular rupture. The residual
gel stays within the breast tissue without absorption, making it difficult to correctly
assess a new implant. Radiolucent centered coarse eggshell calcifications of silicone
granulomas may be seen after implant rupture.
In ruptured saline implants, mammography usually shows only a slightly dense
collapsed envelope near the chest wall because the saline fluid is absorbed by
breast tissue.
a b c
Fig. 17.46 Intracapsular silicone implant rupture. The silicone gel is still contained within an
intact fibrous capsule. A flared lobulated contour may be seen on mammogram (a, b). Ultrasound
depicts the stepladder sign (c)
366 L. M. Yano and M. A. Rudner
a b
Fig. 17.47 Extracapsular rupture. Mammogram (a) shows silicone beyond the fibrous capsule
(arrow). Ultrasound depicts the snowstorm sign (echogenic noise), a typical intense echogenic
artifact caused by the slow velocity of sound in the silicone as compared to the surrounding paren-
chyma, obscuring all findings below it (b, c)
Ultrasound
Ultrasound is a useful tool in screening for ruptured breast implants, less expensive
and more cost-effective than MRI, with sensitivity of 25–65%, specificity of
57–98%, and a negative predictive value of 85% [45].
The classic finding in extracapsular rupture of a silicone implant is the snow-
storm sign (echogenic noise), which is a typical intense echogenic artifact caused by
the slow velocity of sound in silicone when compared to the surrounding paren-
chyma that obscures all findings below it (Figs. 17.47 and 17.48) [43, 45, 49, 50].
The snowstorm sign is present when scanned from all angles, whereas edge artifact
may change or disappear. Echogenic noise may also be seen in lymph nodes with
silicone infiltration (Fig. 17.49), gel bleed, and direct silicone or paraffin injections,
which can be so intense that ultrasound is nondiagnostic (Figs. 17.64 and 17.65) [28].
17 Postoperative Breast 367
a b c d g
e f
Fig. 17.48 Extracapsular rupture. Mammogram (a, b) shows contour abnormality due to extruded
silicone that is not far enough from the implant to be seen as a separate finding. It may be difficult
to differentiate from a double contour of periprosthetic effusion. Ultrasound depicts the snowstorm
sign on this topography (c). Axial T1-weighted without fat suppression (d), axial fat-suppressed
T2-weighted (e), and axial short T1 inversion-recovery silicone-excited images (f) demonstrate the
outer silicone next to the implant, and the reconstructed sagittal short T1 inversion-recovery
silicone-excited image (g) shows the discontinuous envelope (arrow)
a b
Fig. 17.49 Snowstorm sign in an intramammary lymph node with silicone infiltration (a), corre-
sponding to the finding with high uptake on PET-CT (b)
The intact fibrous capsule in intracapsular rupture ensures that the silicone is
isolated from the breast parenchyma, so there will be no snowstorm sign, unless
when associated with extracapsular rupture. Instead, stepladder sign may be pres-
ent, which is characterized by multiple thin echogenic lines within the silicone gel
that looks like the steps of a stepladder (Figs. 17.46 and 17.50) [14, 30, 46, 49, 50].
This sign represents the collapsed envelope from ruptured implant contained by the
intact fibrous capsule. False-positive stepladder signs can be caused by radial folds
(which always extend to the implant periphery) (Fig. 17.60) and intact multilumen
implants producing multiple linear echoes [45, 52]. Less specific signs of intracap-
sular rupture are discontinuous envelope lines (Figs. 17.51 and 17.54), diffuse linear
echoes, debris, or diffuse low-level echoes within the implant [50, 53]. When these
signs a re dubious, MRI can confirm the diagnosis [47].
368 L. M. Yano and M. A. Rudner
Fig. 17.50 Stepladder sign. The collapsed envelope from ruptured implant contained by the intact
fibrous capsule, characterized by multiple thin echogenic lines within the silicone gel
sequences. Silicone and water have high signals on T2-weighted images and com-
bined silicone and water images, but signal suppression of either one makes it pos-
sible to distinguish these components [3, 43, 56, 57]. Intravenous contrast is not
necessary for the detection of implant ruptures but may be useful for evaluating
BIA-ALCL, implant-associated infections, and breast cancer in a patient with
implants (known or suspected).
Normal saline implant shows the intact envelope filled with water, which has
high signal on water-specific images but dark on silicone-specific images. MRI is
usually not used to assess integrity of a saline implant because its rupture is clini-
cally diagnosed by an acute reduction in breast size (Fig. 17.52). Normal single-
lumen silicone implant shows high signal from the silicone gel with a smooth oval
border on silicone-specific images. Radial folds in the envelope are dark lines in all
sequences that extend to the periphery of the implant (Fig. 17.53). Multiplanar
acquisitions enable to look at all implant contours and to differentiate them from
ruptured envelopes [57]. Reactive fluid around the implant and water droplets in a
radial fold topographically outside the envelope are common and nonspecific find-
ings [46].
Intracapsular rupture can be characterized by the keyhole or teardrop sign, the
subcapsular line, or the linguine sign [3, 26, 43, 49, 56, 58–60]. The keyhole or
inverted teardrop sign represents silicone outside the implant envelope within a
short radial fold, characterized with hyperintensity on silicone-specific images
(Fig. 17.54). A subcapsular line is a dark line parallel to the implant shell that does
not reach the implant periphery, representing incomplete shell collapse (Fig. 17.55).
The linguine sign is the collapsed shell represented as curvy noodle-shaped dark
lines inside the implant that do not extend to the periphery and is reported as the
a b
c d
Fig. 17.52 Ruptured saline implant. Ultrasound shows a smaller implant when compared to the
other side, with a normal appearance of the reduced implant (a) and only with a small amount of
periprosthetic fluid (b). Axial T1-weighted MR image without fat suppression (c) shows symmet-
ric hypointense signal inside the fibrous capsule. Axial fat-suppressed T2-weighted MR image (d)
presents a reduced implant with a small amount of periprosthetic fluid (arrow)
370 L. M. Yano and M. A. Rudner
a b
Fig. 17.53 Normal radial folds in the envelope on short T1 inversion-recovery silicone-excited
image (a) and fat-suppressed T2-weighted MR images (b), with a small amount of fluid inside the
last one
most sensitive (93%) and specific (65%) finding for rupture (Figs. 17.56 and 17.57)
[26, 28, 47, 58]. Extracapsular rupture is characterized by silicone outside the
fibrous capsule (Fig. 17.48), within the breast parenchyma or axilla, almost always
associated with signs of intracapsular rupture [28, 56]. Severe gel bleed can present
as a thin layer of silicone around the periphery of the implant but with an apparently
intact envelope and fibrous capsule.
Multilumen or multiple implants (Fig. 17.58), stacked implants or implants made
of rare materials, poor positioning of the breast, and a “redo” from a prior rupture
(implant replacement after removal of ruptured implants) are causes of false-
positives when screening for silicone implants rupture [49, 52]. Wrong positioning
may produce artificial bulges by squeezing a part of implant between the coil and
the chest wall. Silicon material extruded from previous rupture may not be com-
pletely removed during an implant replacement surgery and the residual silicone
droplets near the new intact implant may result in misinterpretation as a new extra-
capsular rupture.
a b
c d
Fig. 17.54 Intracapsular rupture. Axial fat-suppressed T2-weighted MR image (a) shows the
absence of periprosthetic fluid. Silicone-specific images (b, c) show silicone outside the implant
envelope within a short radial fold (keyhole sign) and discontinuity of the envelope (arrows).
Ultrasound was only capable to demonstrate the latter finding (d)
Fig. 17.55 Subcapsular line. Silicone-specific images demonstrate dark lines parallel to the
implant shell that do not reach the periphery of the implant, representing incomplete shell collapse
clinical presentation is an abundant and persistent late seroma around the implant
(more than 70%), which presents more than 1 year after implantation [61]. Other
clinical presentations include a solid mass (20%), distant metastases, asymmetry
and breast pain (21%), aggressive capsular contracture (7%), axillary lymphade-
nopathy, skin lesions (7%), fever (7%), and B-type symptoms (5%) [65]. Two-thirds
of the cases are restricted to the fibrous capsule, and in the remaining cases, it pres-
ents as an infiltrative form associated with nodal and systemic extension. These
forms could represent different stages of the same disease with a very different
prognosis: a favorable outcome in cases of disease limited to the fibrous capsule
(>90% remission following capsulectomy) and a more aggressive outcome in
advanced stages [64]. Limited observations suggest that BIA-ALCL is a clinically
indolent disease, but fatal outcomes have revealed the importance of early diagnosis
and treatment, which can be delayed by lack of knowledge and the absence of
unequivocal signs of disease, which often do not allow the identification of early
pathognomonic radiological signs [65].
Radiological signs are often dubious, highlighting either the periprosthetic effu-
sion or breast masses with varying sensitivity and specificity (Table 17.2) [60].
Periprosthetic effusion presents as a “double contour” in a mammogram, indistin-
guishable between seroma, or a mass (Fig. 17.59). Other mammographic findings
are capsular thickening (Fig. 17.60), irregular contours of implants, and masses
emerging from the fibrous capsule to the breast parenchyma (Fig. 17.61). In some
cases, even when the patient has an abnormality in other tests, mammogram can be
normal. Ultrasound can detect effusion and masses that can be complex solid cystic
or even circumscribed and hypoechoic, not necessarily hyper vascularized.
17 Postoperative Breast 373
a b
Fig. 17.56 Linguine sign. The collapsed shell is represented as curvy noodle-shaped dark lines
inside the implant that do not extend to the periphery, better seen on axial T1-weighted MR image
without fat suppression (a) and axial short T1 inversion-recovery silicone-excited image (b).
Computed tomography (CT) also displays the collapsed envelope (c)
a b c d h
e f g
Fig. 17.57 Intracapsular rupture of a double lumen implant. Mammogram shows hypodense
images inside the implant (a, b). Ultrasound depicts a heterogeneous outer lumen and the snow-
storm sign which make impossible to evaluate the inner envelope completely (c, d). Axial
T1-weighted image without fat suppression (e), axial fat- suppressed T2-weighted (f), and axial
and sagittal short T1 inversion-recovery silicone-excited MR images (g, h) show the intact inner
saline lumen and a collapsed outer silicone lumen but with no signs of extracapsular rupture
a b c f g
d e
Fig. 17.58 False-positive rupture. Mammogram show the implant with irregular contour in its
upper aspect (a, b), being very difficult to exclude a diagnosis of rupture. Ultrasound (c) depicts
undulated contours of the implant with a regular thin echogenic line inside the implant that reaches
the periphery of the envelope, but with an angulation and a convexity posterior in its trajectory
(arrows). Axial short T1 inversion-recovery silicone-excited image (d) and axial fat-suppressed
T2-weighted MR image (e) show silicone covering the implant that presents regular contours, with
no signs of rupture. Sagittal short T1 inversion-recovery silicone-excited reconstructed image (f)
and sagittal contrast-enhanced fat-suppressed T1-weighted MR image (g) delimits better the nor-
mal aspect of the implant and depicts another image similar to it, inside the fibrous capsule (arrow).
This patient corrected an aesthetic defect with another silicone implant, placed over the first one.
Knowledge about patient’s medical history is the key to avoid misdiagnosis
17 Postoperative Breast 375
Table 17.2 Sensitivity and specificity for effusion and a detection rate for masses for each
imaging test
Sensitivity for Specificity for detection rate for
Imaging test effusion effusion masses
Mammography 73% 50% ***
Ultrasound 84% 75% 46%
MRI 82% 33% 50%
Computed tomography 55% 83% 50%
Positron emission tomography 38% 83% 64%
(PET-CT)
***Mammogram is unable to distinguish between effusion and masses
Gossypibomas
Gossypibomas are retained surgical material that remains inside the body after sur-
gery, extremely rare in superficial sites (there are few locations of difficult access
that can impair the visibility of a gauze or sponge). Radiologic presentation is broad
and reflects the type of response presented. Acute responses may be exudative, lead-
ing to infection, abscess, and even fistula formation. Chronic responses may present
as aseptic fibrinous, with adhesion/encapsulation, clinically silent or appear within
years after surgery [66].
Mammography pathognomonic finding is the characterization of a radio-opaque
marker (Fig. 17.62). Sponges without radio-opaque markers or markers that are
fragmented or disintegrated may present as a mass with mottled radiolucency, due
to air trapping. Ultrasound may show sponges as a highly echogenic anterior band
376 L. M. Yano and M. A. Rudner
a b
Fig. 17.59 Seroma. Mammography (a, b) shows a “double contour” around the implant consis-
tent with periprosthetic effusion on ultrasound (c)
17 Postoperative Breast 377
a b c e g
d f h
Fig. 17.60 False-positive exam. Mammogram (a, b) shows a discrete double-contour and capsu-
lar thickening (arrow). Ultrasound (c, d) depicts a false-positive stepladder sign caused by radial
folds and a heterogeneous intracapsular mass (star) above them making really hard to characterize
the envelope margin. Axial T1-weighted MR image without fat suppression (e) and axial short T1
inversion-recovery silicone-excited image (f) show the intact envelope being deformed by a intra-
capsular mass. Axial fat- suppressed T2-weighted MR image (g) demonstrates a heterogeneous
mass with enhancement on contrast-enhanced fat-suppressed T1-weighted MR subtraction image
(h). Explantation was performed and the diagnosis was an organized bruise
a b c h
d e f g
Fig. 17.61 Breast implant-associated anaplastic large cell lymphoma. Mammogram shows a dou-
ble contour implant and an irregular density on the lower part of the implant, next to the fibrous
capsule (a–c). Ultrasound depicts an echogenic periprosthetic effusion (f, g) and an irregular ill-
defined mass next to the fibrous capsule (d, e). CT demonstrates irregular margins of the implant
and the periprosthetic effusion (h). CT sagittal reconstruction (i) shows a discrete irregular density
on the lower part of the implant, next to the fibrous capsule. This patient was submitted to a surgi-
cal explantation and the histological result was BIA-ALCL
378 L. M. Yano and M. A. Rudner
with posterior acoustic shadowing that may be associated with fluid collections. On
MRI, a mass with low-signal intensity stripes seen on T2-weighted images is highly
suggestive. In addition, breast augmentation with implants presents diagnostic dif-
ficulties in the evaluation of structures posterior to the implant, both on ultrasound
and on mammography, and MRI is more sensitive in these cases. Strong enhance-
ment on contrast-enhanced sequences is rare and may mimic neoplasm, prompting
biopsy (Fig. 17.63) [66, 67].
a b
Fig. 17.62 Gossypiboma. Mammography (a) shows a circumscribed mass, with typical radio-
opaque marker inside, adjacent to the upper contour of the implant. Ultrasound (b) depicted an
intracapsular complex mass with posterior acoustic shadowing. These findings were consistent
with surgical gauze
a b c
Fig. 17.63 Gossypiboma. MRI shows a circumscribed intracapsular mass, with heterogeneous
signal intensity on axial fat-suppressed T2-weighted image (a) and homogeneous signal intensity
on T1 (b). Sagittal contrast-enhanced fat-suppressed T1-weighted MR image (c) revealed internal
heterogeneous and persistent enhancement, with a small non-enhancing central zone. Surgical
removal of the retained gauze was performed
17 Postoperative Breast 379
a b c
Fig. 17.64 Liquid silicone injection. Mammogram views (a, b) show diffuse multiple radiopaque
masses in different patterns. Ultrasound (c, d) shows the snowstorm sign diffusively, which
obscures all findings below it
17 Postoperative Breast 381
a b c d e
Fig. 17.65 Liquid silicone injection. Patient underwent a second surgery to remove the previously
injected liquid silicone and repair breasts aesthetically with implants placement. Mammogram
(a–d) shows radiopaque irregular densities with tiny punctate images that resemble calcifications,
representing the residual injected silicone. Ultrasound (e) shows the snowstorm sign, adjacent to
the implant contour, which can make it difficult to diagnose a possible rupture
a b
Fig. 17.66 Liquid silicone injection. Mammogram (a, b) shows diffuse multiple radiopaque
masses in different patterns. On CT (c), scattered noncalcified nodular soft-tissue foci is seen
a b
Fig. 17.67 PMMA injection. MRI shows multiple confluents, oval, circumscribed masses,
hypointense on axial fat-suppressed T1-weighted MR image (a) and hyperintense on axial fat- sup-
pressed T2-weighted MR image (b)
PAAG is a polymer synthesized from 2.5% acrylamide and 97.5% water highly
exchangeable with the water molecules of tissue fluids. Thereby a 10% reduction
in volume occurs during the first days, as a consequence of osmotic exchange.
Once considered a nonbiodegradable hydrogel that was nontoxic, non-sensitizing,
and non-teratogenic, with long-term clinical application, many complications
began to be reported, such as pain, induration, displacement, deformation, milk
deposition/galactocele formation, and psychological fear. Some studies suggested
that PAAG may decompose acrylamide monomers under multiple factors, with
carcinogenic and toxic effects on the nervous and reproductive systems. Injection
of approximately 150–200 mL of polyacrylamide gel is made into the retroglandu-
lar space of each breast at the inframammary fold or at the upper region of the
breast. At MRI, PAAG presents hypointense to isointense signal on T1-weighted
images; hyperintense with hypointense peripheral rim on T2-weighted images; and
T2-weighted fat-saturated images are useful because of high water content
(Fig. 17.68) [68, 70].
Mammography may show multiple randomly distributed bizarre densities of
varying shapes and sizes, indistinguishable from the adjacent breast tissues or with
a well-defined oval density (generally on single injection). On sonography, PAAG
injection may be seen as a single collection of globular fluid in a retroglandular pre
pectoral location with variable internal echogenicity similar to the asymptomatic
patients. Other manifestation of acrylate injection is the same of mammogram, mul-
tiple bizarre, and randomly distributed masses. The infected breast may show
marked increase in the size of collection as well as a diffuse increase in its internal
echogenicity to midlevel echoes. The position of the acrylate collections is well
depicted on MRI. Superimposed inflammatory changes increase the intermediate
heterogeneous T1 signal and decrease heterogeneous T2 signal, along with irregular
and thickened rim enhancement. A thin regular rim of delayed enhancement can
also be seen around PAAG collection in asymptomatic patient, similar to the rim
enhancement of breast cysts [68, 70].
384 L. M. Yano and M. A. Rudner
a d
Fig. 17.68 PAAG injection. MRI presents large lobulated masses, with hypointense signal on
axial T1-weighted image without fat suppression (a), hyperintense with hypointense peripheral
rim on fat-suppressed T2-weighted image (b) and a thin peripheral enhancement on contrast-
enhanced fat-suppressed T1-weighted subtraction image (c). Ultrasound (d, e) shows circum-
scribed and heterogeneous masses
Hyaluronic acid (HA) is an absorbable filler, and its aesthetic effects are considered
transitory for its natural and progressive degradability. Despite this significant reab-
sorption, there are some reports of absence of radiological signs of reabsorption
even 24 months after the procedure. Some adverse effects related to the product are
development of masses and breast pain. Infection and abscesses formation are also
described. In spite the potential for additional applications in the future, it increases
the total cost and the risk for development of granulomas.
HA injection determines an increase in breast parenchyma radiodensity on mam-
mography, either diffuse or as multiple radiodense lesions. This finding corresponds
to multiple predominantly anechoic collections with internal echoes of variable
sizes and echogenicity on ultrasound (Figs. 17.69 and 17.70). On MRI, HA collec-
tions appear as well delimited areas with hyperintensity on T2-weighted and hypo-
intensity on T1-weighted images. They may be involved by fibrotic capsules,
assuming suspect appearance.
17 Postoperative Breast 385
a b c
Fig. 17.69 Hyaluronic acid injection. Mammogram (a, b) and tomosynthesis (c) show a slightly
dense asymmetry on superficial planes of the upper quadrants. Ultrasound depicts elongated cystic
images on the subcutaneous fat (d)
386 L. M. Yano and M. A. Rudner
Fig. 17.70 Hyaluronic acid injection. Ultrasound shows multiple cystic images of different sizes,
some of them with thin septa and internal echoes
percentage depends on the methods used to aspirate, prepare, and transplant the fat
and the destination of the graft (in well-vascularized muscles, the survival rate is
higher than in a relatively oxygen-poor environment such as scar tissue from previ-
ous breast surgery) [71]. Most studies estimate that 30–40% of the volume is lost
after the first procedure, which might require subsequent procedures or graft over-
correction [72]. There is no significant difference in the volume or viability of the
fat grafted from different donor sources. FG is considered a safe procedure with a
low number of major complications [73].
Fat grafting is a more targeted augmentation technique than implants, but the two
methods combined are increasingly common. It does not cause hypersensitivity or
foreign-body reactions. The complications related to FG are seroma, under correc-
tion, infection, asymmetry, fat necrosis, and fat embolism [72]. The main diagnostic
difficulties are related to fat necrosis [3, 73]. Patients may be symptomatic, and the
mammographic and US images may be misleading. In such cases, MRI may be
helpful to differentiate between cancer recurrence and liponecrosis, due to fat-
suppressed and post-contrast sequences that demonstrate an unenhanced fat-
containing mass on T1-weighted images [3, 74]. Fat necrosis rate is directly
proportional to the amount of fat injected into the same breast, particularly if per-
formed in a single procedure. It can be prevented by injecting small quantities of fat
in different directions and layers [73].
A wide spectrum of mammographic changes after fat grafting have been reported
in the literature, ranging from benign looking lipid cysts (Fig. 17.71) and architec-
tural distortion of normal breast tissue, to findings suspicious for malignancy such
as clustered microcalcifications, spiculated areas of increased opacity, and focal
masses [3, 75]. Pectoral muscle may appear heterogeneous in density and also pres-
ent low-density strips. On CT or MR images, linear subcutaneous fat stranding can
be seen in the harvesting area. At the site of fat transfer, nonspecific subcutaneous
fat stranding may be seen on CT images, because the injected fat can be difficult to
discern from the native subcutaneous fat layer. If intramuscular injections are per-
formed, lobular foci of fat can be identified. Literature reports no statistically
a b c d
Fig. 17.71 Lipofilling. Mammogram shows a lipid cyst in the palpable area after a fat grafting
procedure to reshape the skin depression from previous oncologic surgery (a–c). On ultrasound,
this area was a subcutaneous complex mass with little shadowing (d)
388 L. M. Yano and M. A. Rudner
significant difference between breast density findings before and after fat injection
and no interference in cancer detection after breast fat grafting [3, 72, 73, 75, 76].
Matrices have been inserted successfully and safely in breast reconstruction and in
cosmetic procedures, with biological or synthetic matrices. Mesh support is used to
restore the lost strength, improving the longevity of ptosis correction in mastopex-
ies. The main objective is to achieve the right balance between persistence, inflam-
mation, biocompatibility, and incorporation without interfering with mammography
or presenting a long-term infection risk. Another advantage is the direct-to-implant
breast reconstruction technique that does not require tissue expansion before implant
insertion, avoids donor site morbidity and lengthy recovery time associated with
autologous flap reconstruction, and substantially reduces operating time, compared
to autologous flap and expander-based breast reconstruction. The high cost of
meshes is a factor to be added when choosing a reconstructive technique, but con-
sidering its good cosmetic outcomes and lower rate of surgical revisions, it can be a
low-cost alternative. The proper selection of patients, considering possible comor-
bidities and risk factors, helps to choose the best reconstructive options for each
one. A low rate of inflammation may not generate the fibrous tissue reaction neces-
sary to prevent recurrent breast ptosis, while higher rates may lead to matrix non-
incorporation and loss.
A biological mesh (acellular dermal matrix – ADM) is a scaffold of dermis pro-
duced from cadaveric human, porcine (Figs. 17.72 and 17.73), bovine, or bovine
pericardium tissue that is stripped of its antigenic cells. It allows rapid host revascu-
larization and cell repopulation, which may favor a better outcome, with statistically
reduced bottoming-out, rippling, capsular contracture of the implant, and
a b c
Fig. 17.72 Porcine biological mesh. Axial T1-weighted without fat suppression (a), fat- sup-
pressed T2-weighted (b) and contrast-enhanced fat-suppressed T1-weighted MR subtraction
images (c) demonstrate a thin intracapsular linear image with hypointense signal in all sequences,
without significant enhancement, which does not cover the entire implant surface, easier to see
because the patient developed a seroma
17 Postoperative Breast 389
Fig. 17.73 Porcine biological mesh. Ultrasound shows a hypoechoic periprosthetic layer repre-
senting the biological membrane (arrows). Usually few months after surgery, it is not still identifi-
able. This is probably due to the high biocompatibility of the biological membrane, causing a mild
fibroblastic reaction with focal tissue integration of the matrix and, thus, appearing less visible
during follow-up
Breast cancer treatment depends on the tumor size and stage (TNM) to establish the
surgical management (lumpectomy or mastectomy) and the need for adjuvant ther-
apy (radiotherapy and/ or systemic therapy). The goals are removing all the cancer
from the breast (tumor-free margins), locoregional control, and eradicate occult
17 Postoperative Breast 391
a b c g
d
f
Fig. 17.75 Synthetic mesh. Mammogram shows meshes almost totally calcified with coarse and
linear patterns (a, b). Ultrasound (c) shows an irregular linear echogenic image, with posterior
acoustic shadow (arrows). Axial T1-weighted without fat suppression (d) axial fat-suppressed
T2-weighted (e), sagittal contrast-enhanced fat-suppressed T1-weighted MR images (f), and 3D
reconstruction (g) depict the mesh as a linear coarse image usually in the interface between the
subcutaneous fat and the fibroglandular tissue, with hypointense signal on T1 sequences and
hyperintense signal on T2 sequences
a b c g
e f
Fig. 17.76 Synthetic mesh. Mammogram (a, b), ultrasound (c) and MRI axial T1-weighted with-
out fat suppression (d), axial fat-suppressed T2-weighted (e), and axial contrast-enhanced fat-
suppressed T1-weighted subtraction images (f) show similar aspects of Fig. 17.75. Sagittal
contrast-enhanced fat-suppressed T1-weighted MR images (g) demonstrate the same aspect shown
on mammogram
392 L. M. Yano and M. A. Rudner
metastatic disease. The treatment plan is based on the tumor TNM classification,
imaging, physical findings, and the patient’s wishes, involving breast radiologists,
breast surgeons, medical oncologists, radiation oncologists, pathologists, and plas-
tic surgeons [79].
Breast-conserving surgery (also known as lumpectomy, setorectomy, partial
mastectomy, or quadrantectomy) is used when the entire tumor can be removed with
a good cosmetic result. These patients usually undergo postsurgical whole-breast
irradiation to control residual microscopic disease.
Mastectomy is chosen when the entire tumor cannot be excised with a good cos-
metic result, for women with contraindications to radiation therapy (i.e., pregnancy,
previous breast radiation therapy, multicentric or diffuse disease, collagen vascular
disease) or if it is the patient’s preference [80].
Both approaches offer equivalent local control disease and identical survival
rates in women with tumors 4 cm or less in diameter and positive or negative axil-
lary lymph nodes, as shown by Protocol B-06 conducted by National Surgical
Adjuvant Breast and Bowel Project (NSABP). Currently, available data suggest that
local recurrence is more related to tumor biology than to surgical technique.
The definition of tumor-free margin has varied among institutions. A recent con-
sensus guideline from the Society of Surgical Oncology (SSO) and the American
Society of Radiation Oncology (ASTRO) has recommended that a negative margin
be defined as “no tumor on ink” based on a review of multiple studies [81].
calcifications at the biopsy site are required to distinguish them from cancer recur-
rence pleomorphic calcifications. When there are no distinguishing features to diag-
nose dystrophic or fat necrosis calcifications, a biopsy should be performed.
Nonspecific microcalcifications that form at or near the biopsy site are a problem.
Unchanged nonspecific calcifications should be monitored because they may repre-
sent either benign findings or incompletely resected tumor. Increasing microcalcifi-
cations are suggestive of breast cancer recurrence and should prompt biopsy unless
they are specific for dystrophic calcifications or fat necrosis [51, 85] (Fig. 17.78).
On MRI, the APBI results in characteristic posttreatment changes, which extend
from the skin to the chest wall. Typically, there is only localized skin thickening in
the APBI area and the absence of the diffuse skin thickening seen with WB-XRT. In
addition, signal voids are common in the postoperative breast after APBI and may
persist up to 25 months after treatment [3].
The incidence of treatment failure is approximately 1% per year. Women who are at
greatest risk for failure include those under age 35 (especially younger than 30) and
treated for invasive cancer with an extensive intraductal component or infiltrating
ductal carcinoma with a large intraductal component; with intraductal carcinoma of
comedo type, intraductal cancer measuring 2.5 cm or greater in diameter, and mul-
ticentric lesions; treated for more than one synchronous cancer in the same breast;
and with angiolymphatic invasion. Microscopic residual disease may not imply a
greater risk of IBTR, but gross residual tumor also has a poor prognosis. Despite the
slightly greater tendency for recurrence in these groups, no risk factor is an absolute
contraindication for breast conservation.
For women who choose lumpectomy, IBTR rates are approximately 5% at 5
years and between 10 and 15% at 10 years after therapy. Invasive IBTR is most
common between 18 months and 7 years after treatment. During this period IBTR
is more common in or around the lumpectomy cavity. After 7 years, IBTR is most
often a random event in any quadrant of the affected breast, not necessarily at the
original site, and is usually unrelated to the original lesion in the breast [86].
Recurrences at the original tumor site are usually caused by original failure to eradi-
cate the cancer and represent true treatment failures and occur earlier than the devel-
opment of a tumor elsewhere in the breast (Fig. 17.79).
On mammography, treatment failures manifest as new pleomorphic calcifica-
tions, scar edges becoming rounder or larger, or masses developing in or around the
lumpectomy cavity [13]. On ultrasound, an IBTR shows a mass with or without
continuity with the surgical scar if it occurs near the original lumpectomy site.
Radiologists must investigate any new mass, because even benign-appearing new
solid masses may represent a new cancer. On MRI, clumped enhancement or an
eccentric residual mass can be seen around the lumpectomy cavity (Fig. 17.80).
396 L. M. Yano and M. A. Rudner
Fig. 17.78 Breast-conserving treatment. Imaging findings of a 54-year-old woman with personal
history of Invasive ductal carcinoma (IDC) in the left breast, treated with breast conserving surgery
7 months ago and radiotherapy. (a) Mammogram shows architectural distortion and cutaneous and
trabecular thickening due to radiation therapy on the left breast. (b) US images show the incision
from the surgical cavity to the skin and cutaneous thickening
17 Postoperative Breast 397
a b
Treatment failures after lumpectomy and WB-XRT are usually treated by sal-
vage mastectomy with or without reconstruction. The choice of repeating lumpec-
tomy without additional radiotherapy or repeating lumpectomy with additional
APBI has little long-term data to conclude on safety and effectiveness [87, 88].
17.7 Mastectomy
Mastectomy is the surgical removal of the mammary gland and is chosen when the
entire tumor cannot be excised with a good cosmetic result, for the women with
contraindications to radiation therapy (i.e., pregnancy, previous breast radiation
therapy, multicentric or diffuse disease, collagen vascular disease), for disease
involving the skin or if preferred by the patient. Even large lesions (more than 5 cm)
or multifocal disease can be treated with breast-conserving surgery if a neoadjuvant
chemotherapy is offered before surgery, and there is a decrease in tumor size
[89–91].
There are various types of mastectomies, and they can be associated or not with
axillary lymph node dissection. In simple mastectomy, the nipple-areolar complex
17 Postoperative Breast 399
(NAC) is removed with an ellipse of skin with underlying breast tissue. In the con-
servative mastectomies (skin-sparing mastectomy and nipple-sparing mastectomy),
the aim is to save as much tissue as possible to provide better aesthetic results in
breast reconstruction. Insufficient removal of the breast tissue may increase the risk
of cancer, and the excessive removal impairs the outcome of the reconstruction.
Residual breast parenchymal tissue should be reported by the radiologist as long-
term surveillance imaging may be indicated. A skin-sparing mastectomy (SSM)
suggests that some of the skin on the breast that would normally have been removed
may remain. It involves complete removal of all breast tissue and the NAC while
preserving the skin envelope and is followed by immediate breast reconstruction.
The postoperative appearance of a skin-sparing mastectomy is variable in terms of
the amount of skin remaining, and imaging will demonstrate a skin flap from the
native skin and subcutaneous fat and either autologous or implant augmentation in
place of the glandular tissue [92]. In case of nipple-sparing mastectomy (NSM,
adeno-mastectomy or subcutaneous mastectomy), the breast tissue is removed as it
is done in a simple mastectomy but with preservation of the nipple-areolar complex.
Some ductal tissue may remain within the nipple itself, as well as in the underlying
tissue, which ensures adequate nipple vascularization. This is usually requested by
patients who are having mastectomy for prophylactic reasons. When this technique
is performed in a cancer treatment setting, tumors should be smaller than <3 cm in
size and more than 2 cm away from the NAC, with negative lymph nodes. There are
no randomized studies comparing the effectiveness of these techniques; however,
retrospective studies have shown an acceptable rate of local recurrence. Typically,
radiotherapy is not routinely performed after a nipple-sparing mastectomy unless
there are pathological indications such as involvement of multiple nodes or a large
tumor. The surgical complication rate of conservative mastectomies is higher when
compared to conventional mastectomies. The main complications are the skin flap
and nipple-areolar complex necrosis, which occur in 3–9% of cases (Fig. 17.81).
Unless there is a medical contraindication to breast reconstruction, patients who
choose mastectomy are always offered breast reconstruction with a tissue expander,
implant, or autologous tissue flap. The breast reconstruction options include an
implant, a latissimus dorsi flap with an implant when significant breast skin has
been lost, or a transverse rectus abdominis myocutaneous (TRAM) flap or one of its
derivative procedures, such as a deep inferior epigastric perforator flap (DIEP).
Imaging of the reconstructed breast is not normally performed after placement of an
expander, implant, or after autologous tissue reconstruction [3].
Reduction mammoplasty may be required in the unaffected contralateral breast
to achieve symmetry with the treated breast. The appearance of breasts recon-
structed with autologous tissue and contralateral normal breasts submitted to reduc-
tion mammoplasty are characteristic and should not be mistaken for cancer.
Recurrence of breast cancer in the mastectomy site without reconstruction is
usually detected by physical examination. Due to the low yield of breast cancer
detection from the small amount of remaining breast tissue, surveillance mammog-
raphy of the mastectomy site is not usually performed. Ultrasound can evaluate
400 L. M. Yano and M. A. Rudner
Fig. 17.82 Recurrence after mastectomy without reconstruction. Imaging findings of a 41-year-
old woman before and after mastectomy. (a) Preoperative CT and US show a solid spiculated mass
involving the skin, confirmed IDC. (b) Postoperative routine CT shows the right mastectomy site
without reconstruction after 10 months and a mass in the axillary tail. (c) US shows a solid spicu-
lated mass and some atypical axillary lymph nodes, core biopsy confirmed new IDC, and meta-
static lymph nodes
After mastectomy, the breast may be reconstructed with autologous tissue, implant,
or a combination of both. The aim of breast reconstruction is to create a new breast
and restore chest symmetry, reducing the psychosocial consequences related to the
mastectomy. There is no ideal technique. It is expected to be effective, fast-
performed, with few complications. The choice of breast reconstruction depends on
402 L. M. Yano and M. A. Rudner
the patient’s goals, medical history, physical examination, and potential need for
adjuvant therapy [93].
Regardless of the chosen technique, cancer may recur in the reconstructed breast.
To detect breast cancer recurrences at a smaller size, radiologists must be familiar
with the range of normal and abnormal imaging appearances of reconstructed
breasts that occur in different surgical techniques.
Implant reconstruction is the most used technique (80% of cases) and requires
placement of a tissue expander in a retropectoral position at the time of mastectomy
(skin-sparing mastectomy) for expansion of the skin. Usually, a saline expander is
used that is gradually filled until the skin is sufficiently stretched and the space is
adequate to hold an appropriately sized implant. Subsequent surgery is done to
replace the tissue expander with a permanent implant behind the pectoral muscle
(Fig. 17.83). Patients with small breasts and little ptosis present more favorable
results since the skin flap completely accommodates the volume of the implant. For
the other hand, women with large and ptotic breasts present less satisfactory results,
often requiring skin reduction, which increases the risk of skin and the nipple-areola
complex necrosis. The need for radiotherapy after mastectomy is not an absolute
contraindication for this type of reconstruction, but it can be associated with poor
outcome, capsular contracture, and even loss of the implant.
The most common form of autologous tissue reconstruction is made with the
transverse rectus abdominis myocutaneous (TRAM) flap (Fig. 17.84). It can be per-
formed as a pedicle (pedicled TRAM) or free flap (muscle-sparing TRAM flap,
DIEP flap, SIEA flap). Another option used when an additional skin is needed to
close the wound or additional soft tissue is required in an implant-based reconstruc-
tion is a latissimus dorsi myocutaneous flap (Fig. 17.85). In all these cases, skin, fat,
and muscle are transferred to the mastectomy site with attachments to vascular
structures and are shaped to form a breast. The choice of flap relies on the tissue
abundance at the donor site and the viability of the vascular pedicle. The method of
choice for vascular mapping (before TRAM and DIEP flaps) is computed tomo-
graphic (CT) angiography. It has an excellent sensitivity (99.6%) and a high positive
predictive value (99.6%) for identifying clinically relevant perforating branches,
allowing a better assessment of perforating vessel size, hemodynamics (maximal
enhancement quantification), and location within the muscle [94]. In recent years,
MR angiography has gained in popularity with similar results to those obtained with
CT angiography [95, 96]. Complications may be related to the donor area or the flap
itself. The most feared flap-related complication is necrosis, in addition to dehis-
cence and hematoma, which are more frequent.
Another form of breast reconstruction involves autologous fat graft injection
(also known as lipofilling) to augment the volume of the reconstructed breast, usu-
ally in combination to other reconstruction form. This procedure transfers mature
adipocytes and adipocyte-derived stem cells (ADSCs) to the defective breast region.
These ADSCs have the ability to stimulate local neoangiogenesis and stimulate
fibroblasts locally, allowing these mature adipocytes to survive and integrate into
the graft-receiving mammary environment. The main disadvantage of this technique
is the impossibility of predicting how much fat will be reabsorbed, often requiring
17 Postoperative Breast 403
Fig. 17.83 Breast reconstruction with double-lumen implant. Imaging findings of a 51-year-old
woman with personal history of IDC in the left breast 4 years ago, treated with nipple-sparing
mastectomy. (a) Mammography showing retropectoral implants, minimal subareolar fibroglandu-
lar parenchyma, and preserved NAC in the left breast. (b) Ultrasound shows the differences
between the single-lumen implant in the right breast (first image) and the expander or double-
lumen implant in the left breast. (c) Axial MR images showing single-lumen silicone implant in the
right breast and double-lumen implant (inner saline compartment; outer silicone gel compartment)
in the left breast
404 L. M. Yano and M. A. Rudner
a e
c f h
Fig. 17.84 Tram-flap reconstruction. Imaging findings of a 41-year-old woman who presented
amorphous clustered calcifications in the screening mammogram, confirmed histologically as
DCIS. (a) Preoperative contrast-enhanced MIP reconstruction image shows two irregular masses
(arrow) and large clumped non-mass enhancement (arrowhead). Mastectomy with a pedicled
TRAM flap reconstruction was performed. Biopsy confirmed IDC, ILC, and DCIS*. (b, c)
Postoperative CT and coronal T1-weighted MR image show the contralateral rectus abdominis
muscle tunneled through the inframammary fold to the mastectomy pocket (arrow). Imaging find-
ings 1 year after mastectomy, chemotherapy, and radiation therapy. (d) Sagittal and axial
T1-weighted MR images show cutaneous thickening and steatonecrosis (arrow) due to radiation
therapy and a thin line separating the fat from the skin of the TRAM flap (arrowhead). Imaging
findings 4 year after treatment. (e) Sagittal and axial T1-weighted MR images show decrease of the
postoperative findings. (f) Mammogram shows autologous flap appearing as fat centrally and cal-
cifications from fat necrosis in the left breast and findings of reduction mammoplasty in the right
breast (made for symmetry). (g) Ultrasound shows the differences between the fat distribution of
the TRAM flap in the left breast and the usual breast parenchyma of the right breast. *IDC invasive
ductal carcinoma, ILC invasive lobular carcinoma, DCIS ductal carcinoma in situ
17 Postoperative Breast 405
more than one procedure to obtain the expected result. The main complication is fat
necrosis and cellulitis. The imaging studies show oil cysts and fat necrosis that are
easily identified on mammography, ultrasound, and MRI. The interaction between
mature adipocytes, ADSC, and the normal mammary cell, as well as the carcinogen
cell, is still unclear [95].
Also, the nipple can be reconstructed out of the skin and tattooed to provide color
similar to the contralateral side.
Mammography is indicated if any breast tissue has been left at the mastectomy
site (with or without implants). Breast cancer recurrences can appear as masses or
suspicious calcifications. The mammographic evaluation is useful in the autologous
tissue reconstruction, especially when there are suspicious physical findings.
Autologous flaps appear radiographically as fat centrally, with or without muscle
fibers around the edges of the flaps. Common findings are calcifications from fat
necrosis, benign dermal calcifications, and calcified hematoma. In patients undergo-
ing radiotherapy after reconstruction, diffuse thickening of the skin and trabeculae
may be seen, usually within the first 6 months after completion of radiotherapy.
Suspicious findings are masses and clusters of microcalcifications. Recurrence is
more likely to occur if the margins were closer to less than 1 cm or if there was prior
lymphovascular invasion. But some studies have shown that mammographic screen-
ing of asymptomatic women does not increase life expectancy and there is no reason
to do so [96].
MR images may be helpful in showing postoperative findings in autologous
reconstructions and to find cancer in the opposite breast. They show that the flap has
fat signal intensity and the pedicle is isointense to pectoralis muscle, with easily
visible fat necrosis, skin thickening, edema, fluid collection, hematoma, or lipofill-
ing changes. The postoperative findings decrease on subsequent MRI studies [3].
406 L. M. Yano and M. A. Rudner
Fig. 17.85 Dorsal-flap reconstruction. Imaging findings of a 53-year-old woman with personal
history of IDC in the left breast 6 years ago with recurrence after 4 years, treated with mastectomy
and latissimus dorsi flap with implant reconstruction and prophylactic nipple-sparing mastectomy
in the contralateral breast. (a) Mammography showing retromuscular implants, autologous flap
appearing as fat centrally in the left breast and minimal subareolar fibroglandular parenchyma, and
preserved NAC in the right breast. (b) Ultrasound shows the differences between the fat distribu-
tion of the TRAM flap in the left breast (arrows) and the usual breast parenchyma of the right
breast (first image). (c) Axial T2-weighted MR image shows major pectoral muscle (arrowhead)
and latissimus dorsi muscle tunneled in the left breast (arrows). (d) Sagittal and axial post-contrast
fat-suppressed T1-weighted MR image show the major pectoral muscle (arrowhead) and a thin line
separating the fat autologous flap (arrow)
17 Postoperative Breast 407
Although the risk for breast cancer is reduced by more than 90% after mastectomy,
it is not eliminated, because the breast tissue is incompletely excised (usually
remaining in the anterolateral aspect of the chest wall and in the axilla). The reported
breast cancer recurrence rate after TRAM flap reconstruction ranges between 4.2%
and 11.7%, (depending on the primary tumor stage, histology and patient’s genetic
risk). Early detection of recurrent tumors in reconstructed breasts may have bene-
fits, but any benefit would come at the cost of many false-positive findings, and a
benefit in terms of patient survival has yet to be proven [97].
The most common finding of recurrent cancer in the reconstructed breast is a
palpable mass and occurs in the skin envelope superficially to the autologous flap
reconstruction. Other signs and symptoms include local pain and tenderness, irregu-
larity of the flap surface, erythematous rash, as well as nodal metastasis. Local
recurrences arise mostly at the medial aspect of the flap because of lymphatic drain-
age to the internal mammary nodes, which are not dissected during mastectomy and
reconstruction. A recurrence can also arise from the chest wall (this one with the
worst prognosis).
The mammographic and sonographic findings may resemble those of the pri-
mary tumor and may include a solid irregular and spiculated mass, distortion,
microcalcifications, and skin thickening. Ultrasound is more sensitive than mam-
mography for detecting occult recurrences [98], especially smaller ones and those
in peripherical location (not included in the mammographic field of view).
MR images show an irregular mass with avid early enhancement and delayed
washout. Nodal recurrence manifests as an increase in the number and size of axil-
lary or internal mammary lymph nodes [99, 100] (Fig. 17.86).
There is not a consensus on the methods for patient follow-up after mastectomy
and breast reconstruction. Many authors recommend performing a physical exami-
nation to detect subcutaneous recurrences and mammography to detect recurrent
tumors in the chest wall. Follow-up with breast MRI may benefit women at high risk
for breast cancer recurrence (histologically aggressive type or genetic susceptibil-
ity). Early detection of recurrence has not been shown to decrease mortality, as most
patients with recurrent breast cancer have metastases when recurrence is diagnosed
[101, 102].
408 L. M. Yano and M. A. Rudner
b c
Fig. 17.86 Recurrence after skin sparing mastectomy (SSM). Imaging findings of a 33-year-old
woman with personal history of DCIS in the right breast 7 years ago, treated with skin sparing
mastectomy. After 5 years she underwent a prophylactic SSM on the left breast. (a) Axial
T1-weighted MR image (anatomical features) and US images show retropectoral implants, absence
of the NAC on the right breast (SSM), and minimal subareolar fibroglandular parenchyma and
preserved NAC on the left breast (NSM). (b) Axial post-contrast fat-suppressed T1-weighted MR
image shows no intramammary suspicious enhancement. (c, d) Axial and sagittal post-contrast
fat-suppressed T1-weighted MR images show an oval, circumscribed axillary mass. (e, f)
Corresponding PET-CT and US images, 7 years after the first treatment. (g) Core biopsy proven
IDC luminal B
17 Postoperative Breast 409
e f
17.10 Conclusion
Patients have a wide range of surgical options and adjuvant therapies. Radiologists
should be familiar with them, including normal and abnormal imaging appearances
from surgical procedures, reconstructed breasts, and radiation therapy changes, as
posttreatment changes can sometimes mimic malignancy or obscure locally recur-
rent breast cancer.
References
1. Mendelson EB. Evaluation of the postoperative breast. Breast Imaging: Curr Status Future
Dir. 1992;30:107–38.
2. Drukteinis JS, Gombos EC, Raza S, et al. MR imaging assessment of the breast after
breast conservation therapy: distinguishing benign from malignant lesions. Radiographics.
2012;32:219–34.
3. Margolis NE, Morley C, Lotfi P, et al. Update on imaging of the postsurgical breast.
Radiographics. 2014;34:642–60.
4. Hayes MK, Gold RH, Bassett LW. Mammographic findings after the removal of breast
implants. AJR Am J Roentgenol. 1993;160:487–90.
5. Collis N, Sharpe DT. Silicone gel-filled breast implant integrity: a retrospective review of 478
consecutively explanted implants. Plast Reconstr Surg. 2000;105:1979–85.
6. Ikeda DM, Borofsky HB, Herfkens RJ, et al. Silicone breast implant rupture: pitfalls of mag-
netic resonance imaging and relative efficacies of magnetic resonance, mammography, and
ultrasound. Plast Reconstr Surg. 1999;104:2054–62.
7. Cicchetti S, Leone MS, Franchelli S, Santi PL. Evaluation of the tolerability of hydrogel
breast implants: a pilot study. Minerva Chir. 2002;57:53–7.
8. Collis N, Litherland J, Enion D, Sharpe DT. Magnetic resonance imaging and explan-
tation investigation of long-term silicone gel implant integrity. Plast Reconstr Surg.
2007;120:1401–6.
9. Kreymerman P, Patrick RJ, Rim A, et al. Guidelines for using breast magnetic resonance
imaging to evaluate implant integrity. Ann Plast Surg. 2009;62:355–7.
10. Marotta JS, Widenhouse CW, Habal MB, Goldberg EP. Silicone gel breast implant failure
and frequency of additional surgeries: analysis of 35 studies reporting examination of more
than 8,000 explants. J Biomed Mater Res. 1999;48:354–64.
11. Janowsky EC, Kupper LL, Hulka BS. Meta-analyses of the relation between silicone breast
implants and the risk of connective-tissue diseases. N Engl J Med. 2000;342:781–90.
12. Kessler DA. The basis of the FDA’s decision on breast implants. N Engl J Med.
1992;326:1713–5.
13. Lipworth L, Holmich LR, McLaughlin JK. Silicone breast implants and connective tissue
disease: no association. Semin Immunopathol. 2011;33:287–94.
14. McLaughlin JK, Lipworth L, Murphy DK, Walker PS. The safety of silicone gel-filled breast
implants: a review of the epidemiologic evidence. Ann Plast Surg. 2007;59:569–80.
15. Muzaffar AR, Rohrich RJ. The silicone gel-filled breast implant controversy: an update. Plast
Reconstr Surg. 2002;109:742–8.
16. Noels EC, Lapid O, Lideman JH, Bastiaanner E. Breast implants and the risk of breast can-
cer: a meta-analysis of cohort studies. Aesthet Surg J. 2015;35:55–62.
17. Spear SL, Mardini S. Alternative filler materials and new implant designs: what’s available
and what’s on the horizon? Clin Plast Surg. 2001;28:435–43.
17 Postoperative Breast 411
18. Tugwell P, Wells G, Peterson J, et al. Do silicone breast implants cause rheumatologic dis-
orders? A systematic review for a court-appointed national science panel. Arthritis Rheum.
2001;44:2477–84.
19. Derby BM, Codner MA. Textured silicone breast implant use in primary augmentation: core
data update and review. Plast Reconstr Surg. 2015;135:113–24.
20. Handel N, Garcia ME, Wixtrom R. Breast implant rupture: causes, incidence, clinical impact,
and management. Plast Reconstr Surg. 2013;132:1128–37.
21. Destouet JM, Monsees BS, Oser RF, et al. Screening mammography in 350 women with
breast implants: prevalence and findings of implant complications. AJR Am J Roentgenol.
1992;159:973–81.
22. Eklund GW, Busby RC, Miller SH, Job JS. Improved imaging of the augmented breast. AJR
Am J Roentgenol. 1988;151:469–73.
23. Ganott MA, Harris KM, Ilkhanipour ZS, Costa-Greco MA. Augmentation mammoplasty:
normal and abnormal findings with mammography and US. Radiographics. 1992;12:281–95.
24. Silverstein MJ, Handel N, Gamagami P. The effect of silicone-gel-filled implants on mam-
mography. Cancer. 1991;68(Suppl):1159–63.
25. Silverstein MJ, Handel N, Gamagami P, et al. Mammographic measurements before and after
augmentation mammaplasty. Plast Reconstr Surg. 1990;86:1126–30.
26. Shah M, Tanna N, Margolies L. Magnetic resonance imaging of breast implants. Top Magn
Reson Imaging. 2014;23:345–53.
27. Monticciolo DL, Nelson RC, Dixon WT, et al. MR detection of leakage from silicone breast
implants: value of a silicone-selective pulse sequence. AJR Am J Roentgenol. 1994;163:51–6.
28. Berg WA, Caskey CI, Hamper UM, et al. Diagnosing breast implant rupture with MR imag-
ing, US, and mammography. Radiographics. 1993;13:1323–36.
29. Young VL, Bartell T, Destouet JM, et al. Calcification of breast implant capsule. South Med
J. 1989;82:1171–3.
30. DeBruhl ND, Gorczyca DP, Ahn CY, Shaw WW, Bassett LW. Silicone breast implants: US
evaluation. Radiology. 1993;189:95–8.
31. Gossner J. Sonography in capsular contracture after breast augmentation: value of estab-
lished criteria, new techniques and directions for research. J Ultrasound. 2017;20:87–9.
32. Ahn CY, Shaw WW, Narayanan K, et al. Residual silicone detection using MRI following
previous breast implant removal: case reports. Aesthet Plast Surg. 1995;19:361–7.
33. Stewart NR, Monsees BS, Destouet JM, Rudloff MA. Mammographic appearance following
implant removal. Radiology. 1992;185:83–5.
34. Henriksen TF, Holmich LR, Fryzek JP, et al. Incidence and severity of short-term complica-
tions after breast augmentation: results from a nationwide breast implant registry. Ann Plast
Surg. 2003;51:531–9.
35. Cortes MIC, Valadez ER, Martinez RDZ, et al. Breast prosthesis syndrome: pathophysiology
and management algorithm. Aesth Plast Surg. 2020;44:1423–37.
36. Feng LJ, Amini SB. Analysis of risk factors associated with rupture of silicone gel breast
implants. Plast Reconstr Surg. 1999;104:955–63.
37. Holmich LR, Friis S, Fryzek JP, Vejborg IM, et al. Incidence of silicone breast implant rup-
ture. Arch Surg. 2003;138:801–6.
38. Bengtson BP, Eaves FF 3rd. High-resolution ultrasound in the detection of silicone gel breast
implant shell failure: background, in vitro studies, and early clinical results. Aesthet Surg
J. 2012;32:157–74.
39. Heden P, Bone B, Murphy DK, et al. Style 410 cohesive silicone breast implants: safety and
effectiveness at 5 to 9 years after implantation. Plast Reconstr Surg. 2006;118:1281–7.
40. Heden P, Bronz G, Elberg JJ, et al. Long-term safety and effectiveness of style 410 highly
cohesive silicone breast implants. Aesthet Plast Surg. 2009;33:430–6.
41. Heden P, Nava MB, van Tetering JP, et al. Prevalence of rupture in Inamed silicone breast
implants. Plast Reconstr Surg. 2006;118:303–12.
412 L. M. Yano and M. A. Rudner
42. Holmich LR, Fryzek JP, Kjoller K, et al. The diagnosis of silicone breast-implant rupture:
clinical findings compared with findings at magnetic resonance imaging. Ann Plast Surg.
2005;54:583–9.
43. Beekman WH, Hage JJ, Taets van Amerongen AH, Mulder JW. Accuracy of ultrasonography
and magnetic resonance imaging in detecting failure of breast implants filled with silicone
gel. Scand J Plast Reconstr Surg Hand Surg. 1999;33:415–8.
44. Rosculet KA, Ikeda DM, Forrest ME, et al. Ruptured gel-filled silicone breast implants:
sonographic findings in 19 cases. AJR Am J Roentgenol. 1992;159:711–6.
45. Chung KC, Wilkins EG, Beil RJ Jr, et al. Diagnosis of silicone gel breast implant rupture by
ultrasonography. Plast Reconstr Surg. 1996;97:104–9.
46. Di Benedetto G, Cecchini S, Grassetti L, et al. Comparative study of breast implant rupture
using mammography, sonography, and magnetic resonance imaging: correlation with surgi-
cal findings. Breast J. 2008;14:532–7.
47. Ahn CY, DeBruhl ND, Gorczyca DP, et al. Comparative silicone breast implant evaluation
using mammography, sonography, and magnetic resonance imaging: experience with 59
implants. Plast Reconstr Surg. 1994;94:620–7.
48. Rivero MA, Schwartz DS, Mies C. Silicone lymphadenopathy involving intramam-
mary lymph nodes: a new complication of silicone mammaplasty. AJR Am J Roentgenol.
1994;162:1089–90.
49. Berg WA, Caskey CI, Hamper UM, et al. Single- and double-lumen silicone breast implant
integrity: prospective evaluation of MR and US criteria. Radiology. 1995;197:45–52.
50. Harris KM, Ganott MA, Shestak KC, et al. Silicone implant rupture: detection with
US. Radiology. 1993;187:761–8.
51. Kuzmiak CM, et al. Mammographic findings of partial breast irradiation. Acad Radiol.
2009;16(7):819–25.
52. Kwek JW, Choi H, Ma J, Miller MJ. Gel-gel double-lumen silicone breast implant: mimic of
intracapsular implant rupture. AJR Am J Roentgenol. 2006;187:W436–7.
53. Palmon LU, Foshager MC, Parantainen H, et al. Ruptured or intact: what can linear echoes
within silicone breast implants tell us? AJR Am J Roentgenol. 1997;168:1595–8.
54. Cher DJ, Conwell JA, Mandel JS. MRI for detecting silicone breast implant rupture: meta-
analysis and implications. Ann Plast Surg. 2001;47:367–80.
55. Holmich LR, Vejborg I, Conrad C, et al. The diagnosis of breast implant rupture: MRI find-
ings compared with findings at explantation. Eur J Radiol. 2005;53:213–25.
56. Gorczyca DP. MR imaging of breast implants. Magn Reson Imaging Clin N Am.
1994;2:659–72.
57. Kim SH, Lipson JA, Moran CJ, et al. Image quality and diagnostic performance of silicone-
specific breast MRI. Magn Reson Imaging. 2013;31:1472–8.
58. Herborn CU, Marincek B, Erfmann D, et al. Breast augmentation and reconstructive surgery:
MR imaging of implant rupture and malignancy. Eur Radiol. 2002;12:2198–206.
59. Soo MS, Kornguth PJ, Walsh R, et al. Intracapsular implant rupture: MR findings of incom-
plete shell collapse. J Magn Reson Imaging. 1997;7:724–30.
60. Adrada BE, Miranda RN, Rauch GM, et al. Breast implant-associated anaplastic large cell
lymphoma: sensitivity, specificity, and findings of imaging studies in 44 patients. Breast
Cancer Res Treat. 2014;147:1–14.
61. Chacko A, Lloyd T. Breast implant-associated anaplastic large cell lymphoma: a pictorial
review. Insights Imaging. 2018;9:683–6.
62. Xu J, Wei S. Breast implant-associated anaplastic large cell lymphoma: review of a distinct
clinicopathologic entity. Arch Pathol Lab Med. 2014;138:842–6.
63. Boer M, Leeuwen FE, Hauptmann M, et al. Breast implants and the risk of anaplastic large-
cell lymphoma in the breast. JAMA Oncol. 2018;4(3):335–41.
64. Forgia DL, Catino A, Fausto A, et al. Diagnostic challenges and potential early indicators of
breast periprosthetic anaplastic large cell lymphoma A case report. Medicine. 2020;99:30.
17 Postoperative Breast 413
65. Miranda RN, Aladily TN, Prince HM, et al. Breast implant-associated anaplastic largecell
lymphoma: long-term follow-up of 60 patients. J Clin Oncol. 2014;32:114–20.
66. Zanetta VC, Rudner MA, et al. Gossypibomas after breast augmentation: an almost-forgotten
diagnosis. Breast J. 2020;26:2125–8.
67. Morris EA, Comstock CE, Lee CH, et al. ACR BI-RADS® magnetic resonance imaging.
ACR BI-RADS® atlas, breast imaging reporting and data system. Reston: American College
of Radiology; 2013.
68. Qian B, Xiong L, Guo K, et al. Comprehensive management of breast augmentation with
polyacrylamide hydrogel injection based on 15 years of experience: a report on 325 cases.
Ann Transl Med. 2020;8(7):475.
69. Grella R, Almadori A, D’Ari A, et al. Management of complication after breast augmentation
with methacrylate. Int J Surg Case Rep. 2015;15:17–20.
70. Teo SY, Wang SC. Radiologic features of polyacrylamide gel mammoplasty.
AJR. 2008;191:W89–95.
71. Khouri RK, Eisenmann-Klein M, Cardoso E, et al. Brava and autologous fat transfer is a
safe and effective breast augmentation alternative: results of a 6-year, 81-patient, prospective
multicenter study. Plast Reconstr Surg. 2012;129:1173–87.
72. Blumenschein AR, Freitas-Junior R, Tuffanin AT, et al. Breast fat grafting: experimental or
established procedure? Rev Bras Cir Plást. 2012;27(4):616–22.
73. Atia AA, Ghareeb FM, Ellabban MG, et al. Clinical and radiological assessment of autolo-
gous fat transfer to the breast. Eur J Plast Surg. 2020;43:139–46.
74. Gigli S, Amabile MI, Pastena FD, et al. Magnetic resonance imaging after breast oncoplastic
surgery: an update. Breast Care. 2017;12:260–5.
75. Veber M, Tourasse C, Toussoun G, et al. Radiographic findings after breast augmentation by
autologous fat transfer. Plast Reconstr Surg. 2011;127(3):1289–99.
76. Rubin JP, Coon D, Zuley M, et al. Mammographic changes after fat transfer to the breast
compared with changes after breast reduction: a blinded study. Plast Reconstr Surg.
2012;129:1029–38.
77. Loo YL, Haider S. The use of porcine acellular dermal matrix in single-stage, implant-based
immediate breast reconstruction: a 2-center retrospective outcome study. Plast Reconstr Surg
Glob Open. 2018;6(8):e1895.
78. Faulkner HR, Shikowitz-Behr L, McLeod M, et al. The use of absorbable mesh in implant-
based breast reconstruction: a 7-year review. Plast Reconstr Surg. 2020;146(6):731e–6e.
79. Bedrosian I, et al. Changes in the surgical management of patients with breast carcinoma
based on preoperative magnetic resonance imaging. Cancer. 2003;98:468–73.
80. Bleicher RJ, et al. Association of routine pretreatment magnetic resonance imaging with time
to surgery, mastectomy rate, and margin status. J Am Coll Surg. 2009;209:180–7.
81. Bleicher RJ, Morrow M. MRI and breast cancer: role in detection, diagnosis, and staging.
Oncology. 2007;21:1521–33.
82. Morrow M. Should routine breast cancer staging include MRI? Nat Clin Pract Oncol.
2009;6:72–3.
83. Sickles EA, Herzog KA. Mammography of the postsurgical breast. AJR Am J Roentgenol.
1981;136:585–8.
84. Godinez J, et al. Breast MRI in the evaluation of eligibility for accelerated partial breast irra-
diation. AJR Am J Roentgenol. 2008;191:272–7.
85. Liberman L, et al. Mammographic features of local recurrence in women who have under-
gone breast-conserving therapy for ductal carcinoma in situ. AJR Am J Roentgenol.
1997;168:489–93.
86. Tuli R, et al. Prognostic indicators following ipsilateral tumor recurrence in patients treated
with breast-conserving therapy. Am J Surg. 2009;198:557–61.
87. Gorechlad JW, et al. Screening for recurrences in patients treated with breast-conserving
surgery: is there a role for MRI? Ann Surg Oncol. 2008;15:1703–9.
414 L. M. Yano and M. A. Rudner
88. Houssami N, Hayes DF. Review of preoperative magnetic resonance imaging (MRI) in breast
cancer: should MRI be performed on all women with newly diagnosed, early stage breast
cancer? CA Cancer J Clin. 2009;59:290–302.
89. Brennan ME, et al. Magnetic resonance imaging screening of the contralateral breast in
women with newly diagnosed breast cancer: systematic review and meta-analysis of incre-
mental cancer detection and impact on surgical management. J Clin Oncol. 2009;27:5640–9.
90. Chen JH, et al. MRI evaluation of pathologically complete response and residual tumors in
breast cancer after neoadjuvant chemotherapy. Cancer. 2008;112:17–26.
91. Dang CM, et al. Increased use of MRI for breast cancer surveillance and staging is not associ-
ated with increased rate of mastectomy. Am Surg. 2009;75:937–40.
92. Pinel-Giroux FM, et al. Breast reconstruction: review of surgical methods and spectrum of
imaging findings. Radiographics. 2013;33:435–53.
93. Clavero JA, et al. MDCT in the preoperative planning of abdominal perforator surgery for
postmastectomy breast reconstruction. AJR Am J Roentgenol. 2008;191(3):670–6.
94. Phillips TJ, et al. Abdominal wall CT angiography: a detailed account of a newly established
preoperative imaging technique. Radiology. 2008;249(1):32–44.
95. Özalp B, Aydinol M. Breast augmentation combining fat injection and breast implants in
patients with atrophied breasts. Ann Plast Surg. 2017;78:623–8.
96. Helvie MA, et al. Mammographic screening of TRAM flap breast reconstructions for detec-
tion of nonpalpable recurrent cancer. Radiology. 2002;224(1):211–6.
97. Medina-Franco H, et al. Factors associated with local recurrence after skin-sparing mas-
tectomy and immediate breast reconstruction for invasive breast cancer. Ann Surg.
2002;235(6):814–9.
98. Edeiken BS, et al. Recurrence in autogenous myocutaneous flap reconstruction after mastec-
tomy for primary breast cancer: US diagnosis. Radiology. 2003;227(2):542–8.
99. Shaikh N, et al. Detection of recurrent breast cancer after TRAM flap reconstruction. Ann
Plast Surg. 2001;47(6):602–7.
100. Vaughan A, et al. Patterns of local breast cancer recurrence after skin-sparing mastectomy
and immediate breast reconstruction. Am J Surg. 2007;194(4):438–43.
101. Lu WL, et al. Impact on survival of early detection of isolated breast recurrences after the pri-
mary treatment for breast cancer: a meta-analysis. Breast Cancer Res Treat. 2009;114:403–12.
102. Seely JM, et al. Breast MRI in the evaluation of locally recurrent or new breast cancer in
the postoperative patient: correlation of morphology and enhancement features with the
BI-RADS category. Acta Radiol. 2007;8:838–45.
Chapter 18
Radiation Therapy
18.1 Introduction
It has been long established that postsurgical radiotherapy reduces the risk of locore-
gional failure. A survival advantage, however, has recently also been demonstrated
[1, 2]. Therefore, some women with breast cancer will need radiation therapy, in
addition to other treatments, as summarized below:
• After breast-conserving surgery (BCS) to reduce locoregional failure in the same
breast or nearby lymph nodes. Breast radiotherapy is recommended in patients
with invasive breast cancer treated with breast-conserving surgery where com-
plete microscopic excision has been achieved, unless life expectancy is less than
3 years due to comorbidities.
• This approach has enormously improved the quality of life and cosmetic out-
come for appropriately selected and treated patients while achieving excellent
long-term survival rates.
• After a mastectomy, especially if the cancer was large (T3/T4), if cancer is found
in many lymph nodes, or if certain surgical margins have cancer such as the skin
or muscle.
• The need for radiotherapy in patients with ductal carcinoma in situ (CDIS) can
be guided by use of the Van Nuys Prognostic Index (VNPI) score that accounts
for tumor size, grade, margin, presence of necrosis, and patient age.
• If cancer has spread to other parts of the body, such as the bones or brain.
18.2.1 Mastectomy
After any type of mastectomy procedure, most of the breast cells are removed.
However, there is a chance of a small amount of breast tissue remaining, and there-
fore the chance of recurrence exists. The rate of recurrence at the chest wall follow-
ing mastectomy is between 5% and 27%.
Recurrence involving the chest wall or skin can frequently be detected on clinical
or breast self-exam, as they are often obvious changes such as palpable masses, skin
thickening, retraction, edema, and redness. The addition of US to clinical exams
may prove to be more accurate than mammography when evaluating a palpable or
visible abnormality, as recurrence tends to be small and close to the skin surface.
Magnetic resonance imaging (MRI) is another imaging tool that can be used to
evaluate the mastectomy site and plays an important role in detecting recurrent
lesions.
For the asymptomatic patients, there is a continuing debate concerning imaging
following mastectomy, because it is said that imaging modalities may not be helpful
after this kind of surgery. So the findings commonly associated with this type of
surgical procedure, followed or not by radiation therapy, will not be addressed in
this chapter.
Breast conservation treatment achieves local tumor control by the surgical removal
of the cancer with margins and is usually followed by radiation therapy. The combi-
nation of conservative surgery and radiation therapy offers the advantage of preserv-
ing the breast, usually with a satisfactory cosmetic result. Given equivalent survival
rates for breast conservation therapy and mastectomy, breast conservation therapy
has become the treatment of choice for early-stage breast cancer.
However, radiologists are faced with increased imaging and diagnostic chal-
lenges when dealing with the conservative treated breast. The treated breasts may be
difficult to position adequately and to compress sufficiently due to surgical
18 Radiation Therapy 417
80,0
70,0
60,0
50,0 Edema
Surgical
20,0
associated
enhancement on
10,0
MRI
0,0 years
0,5 1 2 3 4 5 6
Fig. 18.1 Expected chronological appearance of the surgical findings on the conservatively
treated breast
418 P. de Camargo Moraes
Breast edema and skin thickening are posttreatment findings with similar time
courses for appearance and regression. Typically, post-lumpectomy edema is local-
ized to the area of the incision, and breast edema from radiation therapy usually
encompasses the entire breast.
Breast edema may present as more of an accentuated trabecular pattern or as
overall increased breast density depending on the degree of the edema (Figs. 18.4
and 18.5). The perceived increased density in the irradiated breast may also be
explained by suboptimal exposure because the treated breast often is swollen and
less compressible.
Skin thickening during the period after radiation is secondary to breast edema
from the damage of small vessels. Skin thickening is the most common finding after
breast-conserving therapy, reported in up to 90% of patients. Normal skin thickness
of the breast as seen on mammograms is 2 mm. The skin thickness after radiation
therapy may reach 1 cm or more (Figs. 18.4 and 18.5).
Breast edema and skin thickening are best appreciated when compared with the
contralateral breast or with pretreatment mammograms (Figs. 18.4 and 18.5).
At mammography, maximal breast edema and skin thickening are usually seen
during the first 6 months after completion of radiation therapy. These alterations
then diminish and attain stability within 2–3 years.
18 Radiation Therapy 419
Fig. 18.2 Postoperative seroma in a 62-year-old woman with history of invasive left breast carci-
noma. Mediolateral oblique (a) and craniocaudal (b) mammograms obtained 6 months after radia-
tion therapy show an ill-defined mass in upper outer left breast consistent with postoperative
seroma (arrows). The ultrasound image (c) shows a complex solid-cystic mass. These findings are
consistent with postoperative seroma given history of breast conservation therapy in this area
420 P. de Camargo Moraes
a b c
Fig. 18.3 Postoperative seroma in a 44-year-old woman with history of in situ right breast carci-
noma. Mediolateral (a) and craniocaudal (b) mammograms obtained 6 months after radiation
therapy show an obscured oval mass in upper outer right breast consistent with postoperative
seroma (arrows). Increased breast density and skin thickening can also be seen. The ultrasound
image (c) shows a complex solid-cystic mass with thick walls. These findings are consistent with
postoperative seroma given history of breast conservation therapy in this area
a b
Fig. 18.4 Breast edema and skin thickening due to radiation therapy in a 74-year-old woman with
history of invasive left breast carcinoma. Mediolateral oblique (a) and craniocaudal (b) mammo-
grams of the right and left breasts obtained 1 year after radiation therapy show increased breast
density and skin thickening of the left breast
Edema or skin thickening that increases after stability has been achieved should
alert radiologists to the need for further investigation. The differential diagnoses of
recurrent or worsening breast edema include lymphatic spread of cancer, obstructed
venous drainage, congestive heart failure, and infection.
18 Radiation Therapy 421
a b
Fig. 18.5 Breast edema and skin thickening due to radiation therapy in an 80-year-old woman
with history of invasive left breast carcinoma. Mediolateral oblique (a) and craniocaudal (b) mam-
mograms of right and left breasts obtained 3 years after radiation therapy show increased breast
density and skin thickening of the left breast, which is most prominent in the periareolar area
Fat necrosis of the breast is a benign entity which may be seen after trauma, surgery,
and radiotherapy, among other conditions [6–8]. Clinically, the patients may be
asymptomatic or may present with a palpable lump, skin tethering, and induration.
In imaging studies, the appearance of fat necrosis ranges from typically benign
to worrisome for malignancy, depending on the time at which diagnostic imaging is
performed. This is directly related to whether inflammation or fibrosis is predomi-
nating within the lesion, and correlation with clinical history is very important for
the correct evaluation of these lesions [6].
The classically benign appearing mammographic findings for fat necrosis are the
oil cysts, which are masses with central lucency. These oil cysts may be accompa-
nied by peripheral rim or “eggshell” calcifications (Fig. 18.6).
The presence of calcifications on mammography suggests that most of the calci-
fications will evolve to a dystrophic morphology as the lesions become older. At the
beginning of the calcification process, sometimes we can intercept pleomorphic or
amorphous appearing calcifications on mammography.
Fat necrosis appearing as suspicious noncalcified masses may demonstrate
increased density due to progressive parenchymal fibrosis resulting in an ill-defined,
spiculated mass on mammography, and biopsy may be warranted for the adequate
diagnosis.
Fat necrosis ranges from simple cyst to complex cystic or solid masses on
sonography (Fig. 18.7). As the appearance of fat necrosis can be undetermined
422 P. de Camargo Moraes
Fig. 18.8 Fat necrosis on MRI. T1-weighted nonfat-saturated image (NFS) shows a hyperintense
circumscribed mass with a hypointense rim (arrow). The mass signal is similar to the adjacent fat,
characteristic of fat necrosis. Sagittal T1-enhanced and fat-suppressed and subtraction images
show the fat-containing mass with a non-enhancing thin fibrous rim (arrow)
Fig. 18.10 Mediolateral oblique and craniocaudal mammogram of the right breast shows post-
lumpectomy site as an area of architectural distortion in the upper outer quadrant (arrows). Note
the presence of central lucency and thick, curvilinear spiculations suggestive of surgical scar
Benign calcifications may develop at the postoperative site, with a reported inci-
dence of 28% within the first 6–12 months after radiation therapy.
Dystrophic calcifications generally develop in areas of fat necrosis and are usu-
ally round and coarse and typically large and often have lucent centers (Fig. 18.11).
Suture material left in the breast may also calcify, forming distinctive shapes such
as knot like, rod-shaped, and curvilinear (Fig. 18.12).
On magnification views, these benign forms of calcifications can often be recog-
nized and differentiated from pleomorphic or other suspicious calcifications associ-
ated with malignancy (Fig. 18.13).
At times, however, dystrophic calcifications may simulate malignancy. As previ-
ously described, early calcification of evolving fat necrosis may produce an appear-
ance that is mammographically inconclusive. In such cases, careful inspection of
the previous mammograms may help by showing regression of the calcifications
over time or formation of the calcifications around a radiolucent center of fat, sug-
gesting the benign nature (Fig. 18.14). If calcifications cannot be distinguished from
a possible malignant process radiographically, biopsy should be considered.
426 P. de Camargo Moraes
Fig. 18.11 Dystrophic calcifications in 72-year-old woman with history of right breast carcinoma.
Mediolateral oblique and craniocaudal mammograms of right breast obtained 12 years after
lumpectomy and radiation therapy. Large coarse calcifications (arrows), representing dystrophic
calcifications, are seen within tumor excision site
18.3.1 Mammography
Fig. 18.12 Sutural calcifications in a 91-year-old woman with a history of left breast cancer that
was treated with lumpectomy and radiation therapy. Mediolateral oblique and craniocaudal mam-
mograms of post-lumpectomy and radiation of the left breast show curvilinear and knot-shaped
calcifications. These findings are characteristic of sutural calcifications, which are most commonly
seen in the irradiated breast and are rarely observed after benign breast surgery
Fig. 18.13 Dystrophic calcifications in a 65-year-old woman with history of right breast carci-
noma. Mediolateral oblique and craniocaudal mammograms of right breast obtained 5 years after
lumpectomy and radiation therapy. Coarse calcifications (arrows), representing dystrophic calcifi-
cations, are seen within tumor excision site
Fig. 18.14 Follow-up mammograms help showing the formation of the calcifications around the
area of fat necrosis, suggesting the benign nature
18 Radiation Therapy 429
18.3.2 Tomosynthesis
18.3.3 Ultrasound
Breast ultrasound is a widely used method adjuvant to mammography for the fur-
ther characterization of lesions identified on mammography. It provides additional
information on lesions’ margin, shape, internal echotexture, vascularity, and elastic-
ity [7, 12].
Ultrasonography is also useful in demonstrating the origin of a palpable mass
either within the breast parenchyma or the implant, in cases of breast
reconstruction.
If a lesion has suspicious morphology on any breast imaging method and is vis-
ible on ultrasound, ultrasound-guided biopsy is the procedure of choice. When per-
formed correctly, this procedure is safe and minimally invasive and has a high
diagnostic accuracy, comparable to surgical biopsy.
Fig. 18.15 MRI of post-lumpectomy and radiation therapy of the right breast shows fluid cavity
at the surgical site with smooth, thin rim enhancement (arrows)
18 Radiation Therapy 431
Fig. 18.16 Post-lumpectomy with positive margins MRI was performed before the radiation ther-
apy of the right breast. Fluid cavity at the surgical site with smooth, thin rim enhancement (thin
arrows). Anterior to the seroma, it is possible to identify a suspicious non-mass-like segmental
enhancement (large arrows)
18.4 Conclusion
a b
Fig. 18.17 Mediolateral oblique and craniocaudal mammogram (a) of the left breast after lumpec-
tomy and radiation therapy shows area of architectural distortion at the lumpectomy site in the
retroareolar region, with associated increased density (arrows). Target ultrasound (b) shows irregu-
lar mass with angulated margins. MRI (c) was performed to better access this finding and demon-
strated segmentar enhancement of the area and biopsy was recommended. Histologic result was
steatonecrosis associated with chronic granulomatous inflammatory process with multinucleated
giant foreign body cells
18.5 Summary
The posttreatment alterations include fluid collections, breast edema, skin thicken-
ing, fat necrosis, architectural distortion, and calcifications. There is an expected
chronological appearance for these findings on the conservatively treated breast,
and this is important to know for the correct diagnosis and conduct.
Mammograms and other imaging modalities should be evaluated and compared
with earlier studies to maximize detection of recurrent breast carcinoma while mini-
mizing unnecessary recalls and biopsies.
References
1. Joshi SC, Khan FA, Pant I, Shukla A. Role of radiotherapy in early breast cancer: an overview.
Int J Health Sci (Qassim). 2007;1(2):259.
2. Fisher B, Anderson S, Bryant J, Margolese RG, Deutsch M, Fisher ER, et al. Twenty-year fol-
low-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus
irradiation for the treatment of invasive breast cancer. N Engl J Med. 2002;347(16):1233–41.
3. Chansakul T, Lai KC, Slanetz PJ. The postconservation breast: part 1, expected imaging find-
ings. Am J Roentgenol. 2012;198:321–30.
4. Chansakul T, Lai KC, Slanetz PJ. The postconservation breast: part 2, imaging findings of
tumor recurrence and other long-term sequelae. Am J Roentgenol. 2012;198(2):331–43.
5. Krishnamurthy R, Whitman GJ, Stelling CB, Kushwaha AC. Mammographic findings after
breast conservation therapy. Radiographics. 1999;19(suppl_1):S53–62.
18 Radiation Therapy 433
6. Fernandes Chala L, de Barros N, de Camargo Moraes P, Endo É, Kim SJ, Maciel Pincerato
K, et al. Fat necrosis of the breast: mammographic, sonographic, computed tomography, and
magnetic resonance imaging findings. Curr Probl Diagn Radiol. 2004;33(3):106–26.
7. Tayyab SJ, Adrada BE, Rauch GM, Yang WT. A pictorial review: multimodality imaging of
benign and suspicious features of fat necrosis in the breast. Br J Radiol. 2018;91(1092):2018021.
8. Kerridge WD, Kryvenko ON, Thompson A, Shah BA. Fat necrosis of the breast: a pictorial
review of the mammographic, ultrasound, CT, and MRI findings with histopathologic correla-
tion. Radiol Res Pract. 2015;2015:613139.
9. Günhan-Bilgen I, Oktay A. Mammographic features of local recurrence after conserva-
tive surgery and radiation therapy: comparison with that of the primary tumor. Acta radiol.
2007;48(4):390–7.
10. Robertson C, Ragupathy SKA, Boachie C, Fraser C, Heys SD, MacLennan G, et al. Surveillance
mammography for detecting ipsilateral breast tumour recurrence and metachronous contralat-
eral breast cancer: a systematic review. Eur Radiol. 2011;21(12):2484–91.
11. Sia J, Moodie K, Bressel M, Lau E, Gyorki D, Skandarajah A, et al. A prospective study
comparing digital breast tomosynthesis with digital mammography in surveillance after breast
cancer treatment. Eur J Cancer. 2016;61:122–7.
12. Ramli Hamid MT, Rahmat K, Hamid SA, Kirat Singh SK, Hooi TG. Spectrum of multimo-
dality findings in post-surgical breast cancer imaging. Curr Med Imaging Former Curr Med
Imaging Rev. 2018;15(9):866–72.
13. Drukteinis JS, Gombos EC, Raza S, Chikarmane SA, Swami A, Birdwell RL. MR imaging
assessment of the breast after breast conservation therapy: distinguishing benign from malig-
nant lesions. Radiographics. 2012;32(1):219–34.
14. Margolis NE, Morley C, Lotfi P, Shaylor SD, Palestrant S, Moy L, et al. Update on imaging of
the postsurgical breast. Radiographics. 2014;34(3):642–60.
Chapter 19
Adjuvant Therapy
19.1 Indications
four positive lymph nodes. For patients with negative or less than four positive
lymph nodes, clinical characteristics and tumor pathology need to be evaluated to
assess patients’ risk of recurrence and the benefit of chemotherapy. Features associ-
ated with a higher risk of recurrence include younger age, premenopausal status,
grade 3, positive lymph nodes, presence of angiolymphatic invasion, and low
expression of ER/ PR.
More recently, a helpful tool that can be considered is the use of assays that
evaluate recurrence risk based on gene expression profiles from tumor samples. The
oncotype DX recurrence score (RS) is the most well-validated assay to predict adju-
vant chemotherapy benefit. Oncotype DX RS provided a score based on gene
expression that allows a classification of the risk of recurrence in low (RS < 11),
intermediate (RS 11–25), or high (RS > 25). The TAILORx trial, a phase III trial,
evaluated the use of oncotype DX to predict the benefit of chemotherapy for hor-
mone receptor-positive node-negative breast cancer. Patients with low-risk received
endocrine therapy alone, those with high risk received endocrine therapy plus che-
motherapy, and those with intermediate risk were randomized to endocrine therapy
alone or endocrine therapy plus chemotherapy. Results showed that patients in the
intermediate group can be treated with endocrine therapy alone with satisfactory
outcomes [2]. However, subgroup analysis suggested that women younger than
50 years with a RS of 16–25 might benefit from the addition of chemotherapy [3].
Based on these results, patients with node-negative disease may be spared from
adjuvant chemotherapy if they present a RS less than 16 for women with any age or
less than 26 for those older than 50 years.
HER2 amplification is an important driver of HER2-positive breast cancer.
Adjuvant anti-HER2 therapy is indicated in addition to chemotherapy for HER2-
positive tumors greater than 1 cm or node-positive [1, 4]. Pivotal trials of anti-HER2
therapy did not have a representative population of node-negative tumors smaller
than 1 cm. Nevertheless, anti-HER2 therapy may also be considered for this group
based on a significant risk of relapse described in some studies, especially when
high-risk features are present [5, 6].
Finally, neoadjuvant or adjuvant chemotherapy has a major role for triple-
negative (TN) breast cancer, usually characterized by aggressive behavior and high
recurrence risk. For these patients, chemotherapy should be considered for any
tumor greater than 0.5 cm or with positive lymph node (regardless of primary tumor
size) [1].
Treatment regimens based on anthracyclines and taxanes are standard regimens for
breast cancer patients with an indication of adjuvant chemotherapy. These drugs
may be combined in different ways, depending on local expertise and preference. A
largely used regimen is AC-T, based on doxorubicin and cyclophosphamide,
19 Adjuvant Therapy 437
sequential with paclitaxel [7]. AC is done for four cycles (every 3 weeks or every
2 weeks in the dense dose regimen), followed by weekly paclitaxel for 12 weeks.
However, anthracyclines may lead to some concerning adverse events such as
cardiotoxicity and secondary leukemia. Regimens without anthracyclines constitute
an alternative, especially for patients with contraindications to anthracyclines. In
this way, TC (docetaxel and cyclophosphamide, every 3 weeks, for four to six
cycles) and CMF (cyclophosphamide, methotrexate, 5-fluorouracil, every 4 weeks,
for six cycles) regimens are also common options of adjuvant chemotherapy [7, 8].
In recent years, relevant studies suggested that patients who received neoadju-
vant systemic therapy may have their adjuvant treatment modulated by the response
to neoadjuvant therapy. For patients with triple-negative breast cancer, those who
present residual disease in the surgical specimen after neoadjuvant therapy benefit
from additional adjuvant therapy with capecitabine for 6 months [9].
As anti-HER2 adjuvant therapy, the monoclonal antibody trastuzumab remains
the cornerstone [10]. Adjuvant trastuzumab is used for a year and is started concur-
rently with a taxane. For locally advanced tumors, AC-TH (doxorubicin and cyclo-
phosphamide, followed by paclitaxel and trastuzumab) and TCH (docetaxel,
carboplatin, and trastuzumab) are preferred regimens [11, 12]. Recent studies have
favored non-anthracycline regimens due to a similar efficacy and lower rate of late
toxicities than anthracycline-based regimens in HER2-positive breast cancer [13].
Finally, for early HER2-positive breast cancer, TH (taxane and trastuzumab) regi-
men is an option, which is preferred, especially for node-negative tumors smaller
than 2 cm [14]. An intensification of adjuvant anti-HER2 therapy, with the addition
of pertuzumab to trastuzumab, has a small benefit in disease-free survival and may
be considered for selected high-risk patients [15].
For HER-2 positive breast cancer patients who received neoadjuvant treatment,
the anti-HER2 therapy is continued after surgery for a total duration of 1 year. When
a complete pathological response is observed, the same anti-HER2 therapy is con-
tinued as adjuvant therapy. Otherwise, patients with residual disease have a relevant
disease-free survival gain with the modification of the adjuvant therapy to T-DM1,
an antibody-drug conjugate of trastuzumab and emtansine [16].
As adjuvant endocrine therapy, tamoxifen or an aromatase inhibitor is usually
indicated. Treatment with an aromatase inhibitor is preferred for postmenopausal
women, as the single endocrine therapy or with a switch to tamoxifen [17].
Tamoxifen is the treatment of choice for premenopausal women who will not
receive ovarian suppression. As an alternative, premenopausal women, especially
high-risk patients, may be treated with the addition of ovarian suppression to endo-
crine therapy (tamoxifen or an aromatase inhibitor) [18].
After treatment of localized disease, patients are followed with periodic consult
and physical examination (every 3–6 months). Mammography should be done
annually to detect local recurrence and second primary breast cancer that are ame-
nable to potentially curative treatment. Other routine image tests are not indicated
during follow-up and should be performed in case of suspicion of metastatic disease
based on patients’ signs and symptoms.
438 L. Testa and R. C. Bonadio
References
1. Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. Early breast
cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol.
2019;30(10):1674.
2. Sparano JA, Gray RJ, Makower DF, Pritchard KI, Albain KS, Hayes DF, et al. Adjuvant
chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med.
2018;379(2):111–21.
3. Sparano JA, Gray RJ, Ravdin PM, Makower DF, Pritchard KI, Albain KS, et al. Clinical
and genomic risk to guide the use of adjuvant therapy for breast cancer. N Engl J Med.
2019;380(25):2395–405.
4. Denduluri N, Somerfield MR, Giordano SH. Selection of optimal adjuvant chemotherapy and
targeted therapy for early breast Cancer: ASCO clinical practice guideline focused update
summary. J Oncol Pract. 2018;14(8):508–10.
5. Gonzalez-Angulo AM, Litton JK, Broglio KR, Meric-Bernstam F, Rakkhit R, Cardoso F, et al.
High risk of recurrence for patients with breast cancer who have human epidermal growth factor
receptor 2-positive, node-negative tumors 1 cm or smaller. J Clin Oncol. 2009;27(34):5700–6.
6. Livi L, Meattini I, Saieva C, Franzese C, Di Cataldo V, Greto D, et al. Prognostic value of posi-
tive human epidermal growth factor receptor 2 status and negative hormone status in patients
with T1a/T1b, lymph node-negative breast cancer. Cancer. 2012;118(13):3236–43.
7. Peto R, Davies C, Godwin J, Gray R, Pan HC, Clarke M, et al. Comparisons between differ-
ent polychemotherapy regimens for early breast cancer: meta-analyses of long-term outcome
among 100,000 women in 123 randomised trials. Lancet. 2012;379(9814):432–44.
8. Blum JL, Flynn PJ, Yothers G, Asmar L, Geyer CE, Jacobs SA, et al. Anthracyclines in Early
Breast Cancer: The ABC Trials-USOR 06-090, NSABP B-46-I/USOR 07132, and NSABP
B-49 (NRG Oncology). J Clin Oncol. 2017;35(23):2647–55.
9. Masuda N, Lee SJ, Ohtani S, Im YH, Lee ES, Yokota I, et al. Adjuvant capecitabine for breast
cancer after preoperative chemotherapy. N Engl J Med. 2017;376(22):2147–59.
10. Moja L, Tagliabue L, Balduzzi S, Parmelli E, Pistotti V, Guarneri V, et al. Trastuzumab con-
taining regimens for early breast cancer. Cochrane Database Syst Rev. 2012;4:CD006243.
11. Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE, Davidson NE, et al. Trastuzumab
plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med.
2005;353(16):1673–84.
12. Slamon D, Eiermann W, Robert N, Pienkowski T, Martin M, Press M, et al. Adjuvant trastu-
zumab in HER2-positive breast cancer. N Engl J Med. 2011;365(14):1273–83.
13. van der Voort A, van Ramshorst M, van Werkhoven E, et al. Three-year follow-up of neoad-
juvant chemotherapy with or without anthracyclines in the presence of dual HER2-blockade
for HER2-positive breast cancer (TRAIN-2): a randomized phase III trial. J Clin Oncol.
2020;15_suppl:501.
14. Tolaney SM, Barry WT, Dang CT, Yardley DA, Moy B, Marcom PK, et al. Adjuvant pacli-
taxel and trastuzumab for node-negative, HER2-positive breast cancer. N Engl J Med.
2015;372(2):134–41.
15. von Minckwitz G, Procter M, de Azambuja E, et al. Adjuvant pertuzumab and trastuzumab in
early HER2-positive breast cancer. N Engl J Med. 2017;377(7):122.
16. von Minckwitz G, Huang CS, Mano MS, Loibl S, Mamounas EP, Untch M, et al.
Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med.
2019;380(7):617–28.
17. (EBCTCG) EBCTCG. Aromatase inhibitors versus tamoxifen in early breast cancer: patient-
level meta-analysis of the randomised trials. Lancet. 2015;386(10001):1341–52.
18. Francis PA, Pagani O, Fleming GF, Walley BA, Colleoni M, Láng I, et al. Tailoring adjuvant
endocrine therapy for premenopausal breast cancer. N Engl J Med. 2018;379(2):122–37.
Chapter 20
Follow-Up After Treatment
20.1 Introduction
The most important ways to detect recurrences are still considered [2–4].
Consultations should be held every 3–6 months in the first 3 years, semiannually for
another 2 years, and annually after the fifth year [5].
In anamnesis, one should ask about symptoms of local recurrence or metastasis,
ask about symptoms side by side with the therapy, in addition to reinforcing adher-
ence to it:
Some important topics to be covered include [6, 7]:
• Constitutional symptoms - anorexia, weight loss, fatigue, insomnia.
• Bone health – bone pain, location and characteristic, associated symptoms,
improvement, and worsening factors.
• Pulmonary symptoms – persistent cough, dyspnea.
• Neurological symptoms – headache, nausea, vomiting, visual changes.
• Gastrointestinal symptoms – abdominal pain, change in bowel habits, and char-
acteristics of the stool.
• Genito-urinary symptoms – vaginal bleeding, difficulty urinating.
• Psychological symptoms – depression and anxiety.
• Endocrine/reproductive symptoms – hot flashes, dyspareunia, vaginal dryness,
preservation of fertility.
The physical examination should include a general exam with an assessment of
vital data, cardiac and pulmonary auscultation, and skeletal muscle, abdominal, and
neurological examination. The importance of gynecological follow-up must be rein-
forced, especially for tamoxifen users [6]. Physical analysis of the breasts and arm-
pit must be carried out rigorously, looking for local and lymph node recurrence signs.
In mastectomy cases with reconstruction, special attention should be given to
implants and possible complications related to contractures and or ruptures. In cases
of reconstruction using flaps such as transverse rectus abdominis (TRAM) or great
dorsal attention to steatonecrosis, which, due to its characteristics, can simulate a
recurrence, mammography has typical features that exclude a possible recurrence,
without the need for biopsy.
In the follow-up, evaluate those patients who indicate the need for genetic research
and who may not have been tested during the initial investigation and evalua-
tion phase.
The patients with family cancer history (breast, ovary, colon) and at high risk of
hereditary tumors, such as breast cancer patients before age 50, patients with triple-
negative tumors before age 60, families descended from Jewish Ashkenazis, and
male patients [16].
The BRCA mutation is the most frequent, although other rarer mutations should
be investigated depending on the personal and family history of several types of
cancer, such as Li-Fraumeni and Cowden syndromes.
The performance of these tests and genetic counseling (with a geneticist or
another trained professional) is essential for evaluating other family members. In
this case, with the identification of high-risk individuals, it is possible to establish
risk-reducing measures.
Although it is possible to perform genetic tests on patients not affected by cancer,
these results can often be inconclusive. The ideal is to conduct genetic testing on the
affected individual, and if a particular mutation is identified, it should be investi-
gated on their descendants.
Several observational studies suggest that physical exercise, avoiding obesity, ade-
quate diet, and low alcohol intake decrease breast cancer recurrence risk [17, 18].
High doses of vitamin D at diagnosis, especially in premenopausal patients, have
been associated with a better prognosis; however, no randomized clinical data asso-
ciate vitamin D supplementation with a lower risk of recurrence.
Complementary therapies, such as acupuncture, mindfulness, music therapy, and
yoga, have been studied in breast cancer survivors. Although there is no evidence of
the relationship between these therapies and a lowest recurrence rate, there is a sig-
nificant improvement in the patients’ quality of life [19]. Some studies suggest that
these practices can improve joint pain in patients using aromatase inhibitors and
physical and psychological pain from breast cancer diagnosis and treatment [20].
Symptoms of menopause, such as hot flushes and vaginal dryness, can be due to
age, chemotherapy treatment (in premenopausal patients), and hormone therapy.
20 Follow-Up After Treatment 443
References
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7.
2. de Bock GH, Bonnema J, van der Hage J, et al. Effectiveness of routine visits and routine tests
in detecting isolated locoregional recurrences after treatment for early-stage invasive breast
cancer: a meta-analysis and systematic review. J Clin Oncol. 2004;22:4010.
3. Montgomery DA, Krupa K, Cooke TG. Follow-up in breast cancer: does routine clinical exam-
ination improve outcome? A systematic review of the literature. Br J Cancer. 2007;97:1632.
4. Lu W, de Bock GH, Schaapveld M, et al. The value of routine physical examination in the fol-
low up of women with a history of early breast cancer. Eur J Cancer. 2011;47:676.
5. Runowicz CD, Leach CR, Henry NL, et al. American Cancer Society/American Society of
Clinical Oncology Breast Cancer Survivorship Care Guideline. J Clin Oncol. 2016;34:611.
6. Loomer L, Brockschmidt JK, Muss HB, Saylor G. Postoperative follow-up of patients with
early breast cancer. Patterns of care among clinical oncologists and a review of the literature.
Cancer. 1991;67:55.
7. Pace BW, Tinker MA. Follow-up of patients with breast cancer. Clin Obstet Gynecol.
1994;37:998.
8. Grunfeld E, Noorani H, McGahan L, et al. Surveillance mammography after treatment of
primary breast cancer: a systematic review. Breast. 2002;11:228.
9. Lash TL, Fox MP, Buist DS, et al. Mammography surveillance and mortality in older breast
cancer survivors. J Clin Oncol. 2007;25:3001.
10. Quinn EM, Coveney AP, Redmond HP. Use of magnetic resonance imaging in detection of
breast cancer recurrence: a systematic review. Ann Surg Oncol. 2012;19:3035.
11. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology.
Genetic/familial high-risk assessment: breast and ovarian. https://www.nccn.org/profession-
als/physician_gls/pdf/genetics_bop.pdf. Accessed on 04 Feb 2020.
12. Freedman RA, Keating NL, Partridge AH, et al. Surveillance mammography in older patients
with breast cancer-can we ever stop?: a review. JAMA Oncol. 2017;3:402.
13. Pant S, Shapiro CL. Aromatase inhibitor-associated bone loss: clinical considerations. Drugs.
2008;68:2591.
14. Rojas MP, Telaro E, Russo A, et al. Follow-up strategies for women treated for early breast
cancer. Cochrane Database Syst Rev. 2005:CD001768.
15. Henry NL, Hayes DF, Ramsey SD, et al. Promoting quality and evidence-based care in early-
stage breast cancer follow-up. J Natl Cancer Inst. 2014;106:dju034.
16. National Comprehensive Cancer Network. www.nccn.org
17. Friedenreich CM, Gregory J, Kopciuk KA, et al. Prospective cohort study of lifetime physical
activity and breast cancer survival. Int J Cancer. 2009;124:1954.
18. Kwan ML, Kushi LH, Weltzien E, et al. Alcohol consumption and breast cancer recurrence
and survival among women with early-stage breast cancer: the life after cancer epidemiology
study. J Clin Oncol. 2010;28:4410.
19. Lyman GH, Greenlee H, Bohlke K, et al. Integrative Therapies During and After Breast
Cancer Treatment: ASCO Endorsement of the SIO Clinical Practice Guideline. J Clin Oncol.
2018;36:2647.
20. Park S, Sato Y, Takita Y, et al. Mindfulness-based cognitive therapy for psychological distress,
fear of cancer recurrence, fatigue, spiritual well-being, and quality of life in patients with
breast cancer-a randomized controlled trial. J Pain Symptom Manag. 2020;60:381.
21. Barnabei VM, Cochrane BB, Aragaki AK, et al. Menopausal symptoms and treatment-
related effects of estrogen and progestin in the Women's Health Initiative. Obstet Gynecol.
2005;105:1063.
22. Lesi G, Razzini G, Musti MA, et al. Acupuncture as an integrative approach for the treatment
of hot flashes in women with breast cancer: a prospective multicenter randomized controlled
trial (AcCliMaT). J Clin Oncol. 2016;34:1795.
20 Follow-Up After Treatment 445
23. Atema V, van Leeuwen M, Kieffer JM, et al. Efficacy of internet-based cognitive behavioral
therapy for treatment-induced menopausal symptoms in breast cancer survivors: results of a
randomized controlled trial. J Clin Oncol. 2019;37:809.
24. Avis NE, Crawford S, Manuel J. Psychosocial problems among younger women with breast
cancer. Psychooncology. 2004;13:295.
25. Azim H Jr, Kroman N, Ameye L, et al. Pregnancy following estrogen receptor-positive breast
cancer is safe – results from a large multi-center case-control study. Eur J Cancer. 2012;48S:
European Breast Cancer Conference #21.
Chapter 21
Breast Cancer During Pregnancy
and Lactation
21.1 Introduction
Breast diseases associated with pregnancy are those diagnosed during pregnancy
and 1 year after delivery.
During pregnancy and lactation, complete development of the mammary glands
occurs due to increase in hormone levels (estrogen and progesterone) and is pro-
moted by the placenta. Glandular proliferation, ductal distension, and stromal invo-
lution occur. Such structural changes are manifested clinically by progressive
increase in the volume, firmness, and nodularity of the breasts, making physical
examination difficult. Therefore, imaging evaluation is necessary.
During these periods, the breasts can be affected by diseases directly related to
physiological changes, inflammatory and infectious diseases, juvenile papillomato-
sis, and benign and malignant tumors.
Palpable changes are responsible for initiating investigation in these patients. All
masses found during pregnancy and lactation must be carefully evaluated, as the
diagnosis of injuries or physiological changes secondary to hormonal stimulation
can only be established after a thorough radiological evaluation.
Y. S. Chang
Breast Imaging Section, Centro de Diagnósticos Brasil (CDB – Alliar), São Paulo, SP, Brazil
Instituto de Radiologia INRAD, Hospital das Clinicas HCFMUSP, Faculdade de Medicina,
Universidade de Sao Paulo, São Paulo, SP, Brazil
e-mail: [email protected]
M. A. Rudner (*)
Hospital Albert Einstein, São Paulo, SP, Brazil
Instituto de Radiologia INRAD, Hospital das Clinicas HCFMUSP, Faculdade de Medicina,
Universidade de Sao Paulo, São Paulo, SP, Brazil
Hospital Moriah and Prevent Senior, São Paulo, SP, Brazil
e-mail: [email protected]
a b
Fig. 21.1 Lactational breast parenchyma. Imaging findings of a 35-year-old lactating woman with
family history of breast cancer. Medio lateral (a) and craniocaudal (b) views of screening mam-
mogram show pronounced increase in density of breast parenchyma, which is very dense, hetero-
geneous, nodular, and confluent. US image (c) shows diffusely hyperechoic breast parenchyma
with prominent ducts
21 Breast Cancer During Pregnancy and Lactation 449
21.2.1 Mammography
pregnancy to stage breast cancer with minimal risk to the fetus [6]. However, it is
recommended to avoid having a mammogram during the first trimester of preg-
nancy, proceeding with the evaluation of breast diseases through ultrasound [5].
Several cellular changes occur in the epithelium of the breasts of pregnant or lactat-
ing women. Most of these changes are so pronounced that they can lead to a false
positive diagnosis of carcinoma. Therefore, the cytological diagnosis of breast
lesions during pregnancy and lactation should be done with caution. An experienced
cytopathologist, with knowledge of pregnancy-specific changes, is needed to avoid
false-positive diagnoses.
The American College of Radiology (ACR) recommends the use of MRI only in
situations where the risk-benefit ratio is well established and specifically states that
contrast agents should not be used routinely in pregnant patients, as they cross the
placental barrier and enter fetal circulation [11, 12]. Although there are no reports
of adverse effects on the fetus due to gadolinium, there are few data available that
corroborate the use or not of contrast.
The use of gadolinium does not contraindicate breastfeeding, so interruption is
not recommended. In the first 24 hours, only 0.04% of the injected contrast is pres-
ent in breast milk. Of this amount that is in milk, only 1% is absorbed in the baby’s
gastrointestinal tract [13].
21 Breast Cancer During Pregnancy and Lactation 451
21.3.3 Galactocele
Galactoceles are the most common benign breast lesions in breastfeeding women.
They occur more frequently after the interruption of breastfeeding when the milk is
contained in the breast [15]. They are cysts formed by cuboidal or flattened
a b
Fig. 21.2 Gestational and secretory hyperplasia. Imaging findings of a 33-year-old pregnant
woman (16 weeks) with a palpable mass in the left breast. US images (a, b) show an oval,
hypoechoic with indistinct margin mass). Core biopsy confirmed secretory hyperplasia
452 Y. S. Chang and M. A. Rudner
epithelium containing fluid similar to milk. The cysts result from ductal dilation and
are often enclosed by a fibrous wall of different thicknesses that can be associated
with an inflammatory component. Clinically, it presents as a softened mass that is
painless on palpation, which can often present as a result of complications such as
infection.
In ultrasonography it presents itself in several ways, including simple cysts, cysts
with posterior acoustic shadow, and debris inside (fat particles in suspension, masses
with liquid-fat level (pathognomonic), among other presentations. Due to the vari-
able amount of fat, protein, and water, echogenicity can be heterogeneous and have
suspicious characteristics, such as irregular shape and non-circumscribed margin.
The most important differential diagnosis includes intracystic carcinomas [16, 17]
(Fig. 21.3).
The mammographic aspect of the galactocele depends on its composition, den-
sity, and viscosity of the fluid [16].
Aspiration puncture is a diagnostic and therapeutic procedure, extracting fluid
milk when performed during lactation and thickened milk when obtained from
older lesions after the end of lactation [15].
Infectious diseases of the breast are uncommon during pregnancy, but they occur
relatively frequently during breastfeeding, with an estimated incidence of 6.6–31%
[18]. The most common agent that causes infection is S. aureus, followed by
Streptococcus. The source is the infant’s nose or oropharynx. The infection occurs
due to fissures in the epithelium of the papilla and areola with retrograde dissemina-
tion of the organisms. Milk stasis is an important risk factor, as it represents an
excellent culture medium [15].
Mammography is generally not necessary to investigate mastitis in lactating
women, unless malignancy is suspected. On the other hand, US plays an important
role in the diagnosis and treatment of mastitis, especially in the face of suspected
abscess formation.
Abscesses develop in 5–11% of lactating women with mastitis and usually mani-
fest as complex hypoechogenic masses or irregular anechoic masses, sometimes
with fluid levels, debris, posterior acoustic reinforcement, and increased vascular-
ization. The inflammatory tissue manifests itself as a poorly defined, hypoechogenic
region around the lesion [19] (Fig. 21.4).
Abscesses are usually treated with antibiotic therapy and surgical incision and
drainage, but they can also be treated with needle aspiration or catheter drainage,
especially when smaller than 2.5 cm. US-guided drainage has the advantage of
identifying the necrotic cavity, distinguishing it from inflammatory tissue. The high
rate of recurrence is the most common complication related to abscesses.
21 Breast Cancer During Pregnancy and Lactation 453
Fig. 21.3 Galactocele. Imaging findings of a 37-year-old, 4 months after delivery with a palpable
mass in the left breast. (a) US images show an oval, circumscribed complex solid and cystic mass,
not seen in mammogram (not shown). FNA suggested galactocele. (b) US image of another patient
with a palpable oval, circumscribed, isoechoic heterogeneous mass. (c) Mammogram shows a fat
containing mass (galactocele, marked with a BB)
454 Y. S. Chang and M. A. Rudner
a b
Fig. 21.4 Acute mastitis (puerperal mastitis). Imaging findings of a 33-year-old woman, 6 months
after stop breastfeeding, with erythematous and swollen right breast. US images (a) show an oval
hypoechoic mass, with indistinct margin and internal vascularity at color Doppler (b) in the right
breast (2.7 cm). Core biopsy confirmed acute mastitis (puerperal mastitis)
During lactation, enlarged lymph nodes may appear, both intramammary and axil-
lary, being related to bacterial propagation from the nipple during breastfeeding.
On ultrasound, these lymph nodes have thickened cortical and globular morphol-
ogy in a symmetrical distribution.
Fig. 21.5 Granulomatous mastitis. Imaging findings of a 31-year-old woman, 4 months after
delivery and 2 months after stop breastfeeding, with a palpable mass in the left breast. US images
show a complex solid and cystic mass with indistinct margin (2.6 cm). Core biopsy confirmed
granulomatous mastitis
a b
Fig. 21.6 Lactational adenoma 1 – pregnant. Imaging findings of a 32-year-old pregnant woman
(38 weeks) with a palpable mass in the left breast. (a, b) US shows a solid, oval, circumscribed
mass, parallel, hypoechoic with 3.2 cm. Core biopsy confirmed lactational adenoma
a b
Fig. 21.7 Lactational adenoma 2 – lactating. Imaging findings of a 38-year-old lactating woman
with a palpable mass in the right breast. US image (a) show a solid, irregular, hyperechoic mass
with indistinct margin, parallel, with 3.7 cm, hypervascularized on color doppler (b). Core biopsy
confirmed lactational adenoma
Fibroadenoma is the most common tumor found during pregnancy and lactation,
due to the increase in hormone levels that can induce their growth. For this reason,
preexisting or undiagnosed fibroadenomas can be identified during pregnancy. The
radiological aspects do not differ from the nonpregnant state.
21 Breast Cancer During Pregnancy and Lactation 457
a b
Fig. 21.8 Lactational adenoma 3 – lactating. Imaging findings of a 35-year-old lactating woman
with a palpable mass in the left breast and family history of breast cancer (mother). US images
(a–c) show a solid, oval, circumscribed hypoechoic mass, parallel, with 1.0cm (arrows) associated
with a non-mass heterogeneous lesion (arrowheads). Core biopsy confirmed lactational adenoma.
Control exam 10-months after biopsy didn’t show any lesion
a b
PABC is defined as breast cancer that occurs during pregnancy or within 1 year after
delivery. PABC affects 1 in every 3000–10,000 pregnancies and represents up to 3%
of all malignant breast tumors. In general, it is biologically aggressive, with nega-
tive hormone receptors (estrogen and progesterone) and positive for type 2 recep-
tors for human epidermal growth factor (Her2-neu).
Currently, women have chosen to become pregnant later, after 30 years of age,
which may contribute to an increase in the incidence of PABC [10].
Patients with a palpable lesion complaint are investigated, so it is important that
the clinical examination of the breasts be performed at the beginning of pregnancy
before breast engorgement occurs, which makes its assessment difficult.
PABC patients tend to have larger tumors and are diagnosed at more advanced
stages, which reflects worse prognosis compared to nonpregnant women of the
same age with breast carcinoma.
The sensitivity of mammography to PABC is lower in pregnant or lactating
women due to increased glandular density. As mentioned earlier, US is the most
appropriate radiological method for assessing PABC. More than 90% of women
with PABC present with masses that are easily assessed using US. In addition, it is
useful in assessing lymph node involvement and monitoring the response to neoad-
juvant chemotherapy. However, mammography plays a complementary role and
should always be performed if cancer is suspected, since it is essential for the
assessment of calcifications. The imaging aspects are the same as for nonpregnant
patients [21, 22].
The diagnosis is made preferably by percutaneous biopsy (core), as it is the saf-
est and most accurate method.
Treatment for patients diagnosed with PABC in the first trimester of pregnancy
consists of modified radical mastectomy with adjuvant chemotherapy after the sec-
ond trimester. For patients with a diagnosis established after the second trimester,
neoadjuvant chemotherapy and conservative surgery with radiotherapy after deliv-
ery can be considered (Figs. 21.10 and 21.11).
After making the diagnosis of PABC, one should assess the risk-benefit of the
mother and fetus before the start of treatment. Therapeutic abortion does not
increase patient survival and should not be indicated.
Survival and recurrence of patients with a previous diagnosis of breast cancer do
not appear to be affected by posttreatment pregnancy.
21.8 Conclusions
The diseases that affect the breasts during pregnancy and lactation have similar
aspects to those observed in nonpregnant women. However, the peculiarities in the
presentation of the lesions together with the physiological changes typical of these
21 Breast Cancer During Pregnancy and Lactation 459
a b
c d
Fig. 21.10 PABC lactating. Imaging findings of a 34-year-old lactating woman (left breast prefer-
ential breastfeeding) with a palpable mass in the left breast. Mediolateral (a), craniocaudal (b), and
magnifications views (c) mammography shows fine pleomorphic segmental calcifications in the
left breast, fine pleomorphic segmental calcifications, and skin thickening in the right breast
(arrows in c), atypical right axillary lymph nodes (arrowheads in a). US images (d, e) show diffuse
hyperechoic parenchyma with prominent ductal system, skin thickening, and an extensive non-
mass hypoechoic lesion that occupies the entire right breast and atypical right axillary lymph nodes
(I, II and III levels in c). Core biopsy (right breast) confirmed invasive carcinoma and ductal carci-
noma in situ. Patient has axillary and hepatic metastasis. Vacuum-assisted biopsy of the left breast
confirmed secretory hyperplasia
460 Y. S. Chang and M. A. Rudner
c
a
Fig. 21.11 PABC pregnant. Imaging findings of a 41-year-old pregnant woman (12 weeks twin
pregnancy) with personal history of surgical excision of Phyllodes tumor in the left breast. US
routine images (a, b) show new solid, irregular mass, with angular margin in the right breast. Core
biopsy confirmed invasive carcinoma and ductal carcinoma in situ. Preoperative needle localiza-
tion mammogram (c) shows coarse heterogeneous grouped calcifications (circles in c). Patient
underwent conservative surgery 1 week after the diagnosis with radiation therapy and chemother-
apy after childbirth. No suspicious lesions were seen after 4 years of follow-up
periods must be considered for the correct diagnosis. Delayed diagnosis is the main
cause of worse prognosis commonly found in pregnant or lactating patients with
breast carcinoma.
References
1. Obenauer S, Dammert S. Palpable masses in breast during lactation. Clin Imaging. 2007;31:1–5.
2. Hogge JP, De Paredes ES, Magnant CM, Lage J. Imaging and management of breast masses
during pregnancy and lactation. Breast J. 1999;5:272–83.
3. Ahn BY, Kim HH, Moon WK. Pregnancy and lactation-associated breast cancer: mammo-
graphic and sonographic findings. J Ultrasound Med. 2003;22:491–7.
4. Yang WT, Dryden MJ, Gwyn K, Whitman GJ, Theriault R. Imaging of breast cancer diagnosed
and treated with chemotherapy during pregnancy. Radiology. 2006;239:52–60.
5. Sabate JM, Clotet M, Torrubia S, Gomez A, Guerrero R, de las Heras P, Lerma E. Radiologic
evaluation of breast disorders related to pregnancy and lactation. Radiographics. 2007;27
Suppl 1:S101–24.
6. Robbins J, et al. Accuracy of diagnostic mammography and breast ultrasound during preg-
nancy and lactation. AJR. 2011;196:716–22.
21 Breast Cancer During Pregnancy and Lactation 461
7. Talele AC, Slanetz PJ, Edmister WB, Yeh ED, Kopans DB. The lactating breast: MRI findings
and literature review. Breast J. 2003;9(3):237–40.
8. diFlorio-Alexander, et al. Breast imaging of pregnant and lactating women. J Am Coll Radiol.
2018;15:11S.
9. Greskovich JF Jr, Macklis RM. Radiation therapy in pregnancy: risk calculation and risk mini-
mization. Semin Oncol. 2000;27:633–45.
10. Ayyappan AP, Kulkarni S, Crystal P. Pregnancy-associated breast cancer: spectrum of imaging
appearances. Br J Radiol. 2010;83(990):529–34.
11. Amant F, Deckers S, Van Calsteren K, Loibl S, Halaska M, Brepoels L, Beijnen J, Cardoso F,
Gentilini O, Lagae L, Mir O, Neven P, Ottevanger N, Pans S, Peccatori F, Rouzier R, Senn HJ,
Struikmans H, Christiaens MR, Cameron D, Du Bois A. Breast cancer in pregnancy: recom-
mendations of an international consensus meeting. Eur J Cancer. 2010;46(18):3158–68.
12. Kanal E, Borgstede JP, Barkovich AJ, et al. American College of radiology white paper on MR
safety. AJR Am J Roentgenol. 2002;178:1335–47.
13. Committee opinion – diagnostic imaging during pregnancy and lactation. Obstet Gynecol.
2017;130:4.
14. Mercado CL, Koenigsberg TC, Hamele-Bena D, Smith SJ. Calcifications associated with lac-
tational changes of the breast: mammographic findings with histologic correlation. AJR Am J
Roentgenol. 2002;179:685–9.
15. Scott-Conner CEH. Diagnosing and managing breast disease during pregnancy and lacta-
tion. Medscape Womens Health eJournal. 1997;2(3). Available at: http://www.medscape.com/
viewarticle/408859.
16. Son EJ, Oh KK, Kim EK. Pregnancy-associated breast disease: radiologic features and diag-
nostic dilemmas. Yonsei Med J. 2006;47(1):34–46.
17. Stevens K, Burrell HC, Evans AJ, Sibbering DM. The ultrasound appearances of galactoceles.
Br J Radiol. 1997;70:239–41.
18. Holanda AAR, Gonçalves AKS, Medeiros RD, Oliveira AMG, Maranhão TMO. Ultrasound
findings of the physiological changes and most common breast diseases during pregnancy and
lactation. Radiol Bras. 2016;49(6):389–96.
19. Yu Q, Chang C, Zhang H. Ultrasound imaging characteristics of breast lesions diagnosed dur-
ing pregnancy and lactation. Breastfeed Med. 2019;14(10):712–7.
20. Sumkin JH, Perrone AM, Harris KM, Nath ME, Amortegui AJ, Weinstein BJ. Lactating ade-
noma: US features and literature review. Radiology. 1998;206:271–4.
21. Espinosa LA, Daniel BL, Vidarsson L, Zakhour M, Ikeda DM, Herfkens RJ. The lactating breast:
contrast-enhanced MR imaging of normal tissue and cancer. Radiology. 2005;237:429–36.
22. Taylor D, Lazberger J, Ives A, Wylie E, Saunders C. Reducing delay in the diagnosis of
pregnancy-associated breast cancer: how imaging can help us. J Med Imaging Radiat Oncol.
2011;55(1):33–42.
Index
© The Editor(s) (if applicable) and The Author(s), under exclusive license to 463
Springer Nature Switzerland AG 2022
S. J. Kim Hsieh, E. A. Morris (eds.), Modern Breast Cancer Imaging,
https://doi.org/10.1007/978-3-030-84546-9
464 Index
N O
National comprehensive cancer network Obesity, 20, 21
(NCCN) guidelines, 27, 42, 144 Oil cysts, 339
National surgical adjuvant breast and bowel Oncotype DX assay, 32
project (NSABP), 392 Oncotype DX recurrence score (RS), 436
National Surgical Adjuvant Breast and Bowel Open surgical capsulotomy, 362
Project (NSABP) B-32 trial, 40 Oreo cookie sign, 350
Neoadjuvant chemotherapy (NAC), 151
Neoadjuvant systemic therapy (NST), 308
lymph node evaluation, 324–326 P
mammography interpretation, 310, 312 Palpable changes
MRI interpretation, 314 axillary lymph nodes, 268
assess response, 316 axillary metastasis, 269
challenging lesions, 323, 324 breast lump, 259
complete response, 317 biopsy-proven breast cancer, 265
DCE-MR imaging, 317 breast tomosynthesis, 263
DCIS, 315 DBT, 263
DWI techniques, 315 diagnostic mammography
functional tumor volume, 317 technique, 260
overestimation of residual ductal carcinoma in situ, 262
disease, 322–323 fibroadenoma, 261
partial response, 319 magnetic resonance imaging, 262, 265
progressive disease, 321 negative predictive value, 263
residual tumor, 316 occult breast carcinoma, 268
stable disease, 320 Paget's disease
time-signal intensity curve BIRADS 3 patients, 266
analysis, 317 imaging methods, 266
underestimation of residual nipple areola complex, 265, 266
disease, 322 noninvasive disease, 266
using AI methods, 315 right breast, 267
multimodality response Papillary neoplasms, 49, 50
assessment, 309–310 Parenchymal scarring, 331
NCT indications, 308 Pathologic complete response (pCR), 5
neoadjuvant endocrine therapy (NET), 308 Pathology, breast cancer of
PCR, 308 atypical ductal hyperplasia and low-grade
ultrasound interpretation, 312, 314 carcinoma in situ, 51
Neurological symptoms, 440 benign high-risk lesions and precursors, 50
Next generation sequencing, 62 biomarkers in ductal carcinoma in situ, 53
Nipple-areolar complex (NAC), 265, 266, fibroepithelial lesions, 48, 49
339, 398–399 immunohistochemistry, 61
Nipple-sparing mastectomy (NSM), 399 in situ hybridization, 61
Noninvasive cosmetic surgery, 379 intermediate and high-grade ductal
Non-mass-like enhancement (NME), 430 carcinoma in situ, 51, 52
Non-palpable breast changes, 269–273 invasive carcinoma, 55, 56
image screening, 269–270 macroscopic evaluation, 57, 58
nipple discharge, 270 next generation sequencing, 62
BIRADS 3 biopsied, 272 papillary neoplasms, 49, 50
ductography, 271 pleomorphic lobular carcinoma in situ
ductoscopy, 273 (PLCIS), 52, 53
initial assessment, 271 pre-analytical procedures, 47
MRI, 272 radiological-pathological
Non-radioactive wireless devices correlation, 47, 48
magseed localization, 236 special subtype, 59
radar reflector localization, 234 Percutaneous biopsy, 450
RFID localizer, 236 Periareolar dermal calcifications, 339
468 Index