Download as pdf or txt
Download as pdf or txt
You are on page 1of 72

17

The Breast
chapter Catherine C. Parker, Senthil Damodaran,
Kirby I. Bland, and Kelly K. Hunt

A Brief History of Breast Hormonal and Nonhormonal Local-Regional Recurrence / 587


Cancer Therapy 541 Risk Factors / 555 Breast Cancer Prognosis / 587
Embryology and Functional Risk Assessment Models / 555 Surgical Techniques in
Anatomy of the Breast 543 Risk Management / 556 Breast Cancer Therapy 588
Embryology / 543 BRCA Mutations / 558 Excisional Biopsy With Needle
Functional Anatomy / 544 Epidemiology and Natural Localization / 588
Physiology of the Breast 547 History of Breast Cancer 561 Sentinel Lymph Node Dissection / 590
Breast Development and Function / 547 Epidemiology / 561 Breast Conservation / 591
Pregnancy, Lactation, Natural History / 562 Mastectomy and Axillary Dissection / 591
and Senescence / 548 Histopathology of Breast Cancer 563 Modified Radical Mastectomy / 592
Gynecomastia / 549 Carcinoma In Situ / 563 Reconstruction of the
Invasive Breast Carcinoma / 565 Breast and Chest Wall / 593
Infectious and Inflammatory
Disorders of the Breast 550 Diagnosis of Breast Cancer 567 Nonsurgical Breast
Bacterial Infection / 550 Examination / 567 Cancer Therapies 594
Mycotic Infections / 550 Imaging Techniques / 567 Radiation Therapy / 594
Hidradenitis Suppurativa / 550 Breast Biopsy / 574 Chemotherapy Adjuvant / 594
Mondor’s Disease / 550 Antiestrogen Therapy / 597
Breast Cancer Staging and
Ablative Endocrine Therapy / 598
Common Benign Disorders and Biomarkers 575
Diseases of the Breast 551 Anti-HER2 Therapy / 598
Breast Cancer Staging / 575
Aberrations of Normal Development and Biomarkers / 575 Special Clinical Situations 599
Involution / 551 Nipple Discharge / 599
Overview of Breast Cancer
Pathology of Nonproliferative Axillary Lymph Node Metastases
Therapy 580
Disorders / 552 in the Setting of an Unknown
In Situ Breast Cancer (Stage 0) / 580
Pathology of Proliferative Disorders Primary Cancer / 600
Early Invasive Breast Cancer
Without Atypia / 553 Breast Cancer During Pregnancy / 600
(Stage I, IIA, or IIB) / 582
Pathology of Atypical Proliferative Male Breast Cancer / 600
Advanced Local-Regional Breast Cancer
Diseases / 553 Phyllodes Tumors / 600
(Stage IIIA or IIIB) / 585
Treatment of Selected Benign Breast Inflammatory Breast Carcinoma / 601
Internal Mammary Lymph Nodes / 587
Disorders and Diseases / 554 Rare Breast Cancers / 602
Distant Metastases (Stage IV) / 587
Risk Factors for Breast Cancer 555

A BRIEF HISTORY OF BREAST CANCER THERAPY exactly resembling the animal the crab. Just as the crab has legs
on both sides of his body, so in this disease the veins extending
Breast cancer has captured the attention of surgeons throughout
out from the unnatural growth take the shape of a crab’s legs.
the ages. The Smith Surgical Papyrus (3000–2500 b.c.) is the
We have often cured this disease in its early stages, but after
earliest known document to refer to breast cancer. The cancer
it has reached a large size, no one has cured it. In all operations
was in a man, but the description encompassed most of the
we attempt to excise the tumor in a circle where it borders on
common clinical features. In reference to this cancer, the author
the healthy tissue.”3
concluded, “There is no treatment.”1 There were few other
The Galenic system of medicine ascribed cancers to an
historical references to breast cancer until the first century. In
excess of black bile and concluded that excision of a local
De Medicina, Celsus commented on the value of operations for
bodily outbreak could not cure the systemic imbalance. Theories
early breast cancer: “None of these may be removed but the
espoused by Galen dominated medicine until the Renaissance.
cacoethes (early cancer), the rest are irritated by every method
In 1652, Tulp introduced the idea that cancer was contagious
of cure. The more violent the operations are, the more angry
when he reported an elderly woman and her housemaid who
they grow.”2 In the second century, Galen inscribed his classical
both developed breast cancer (N. Tulp, Observationes medi-
clinical observation: “We have often seen in the breast a tumor
cae 1652). This single incidence was accepted as conclusive
Key Points
1 The breast receives its principal blood supply from per- in women with a known BRCA1 or BRCA2 mutation or
forating branches of the internal mammary artery, lateral women with lobular or ductal carcinoma in situ.
branches of the posterior intercostal arteries, and branches 6 Routine use of screening mammography in women ≥50
from the axillary artery, including the highest thoracic, lat- years of age reduces mortality from breast cancer by 25%.
eral thoracic, and pectoral branches of the thoracoacromial Magnetic resonance imaging (MRI) screening is recom-
artery. mended in women with BRCA mutations and may be con-
2 The axillary lymph nodes usually receive >75% of the sidered in women with a greater than 20% to 25% lifetime
lymph drainage from the breast, and the rest flows through risk of developing breast cancer.
the lymph vessels that accompany the perforating branches 7 Core-needle biopsy is the preferred method for diagnosis
of the internal mammary artery and enters the parasternal of palpable or nonpalpable breast abnormalities.
(internal mammary) group of lymph nodes. 8 When a diagnosis of breast cancer is made, the surgeon
3 Breast development and function are initiated by a variety should determine the clinical stage, histologic characteris-
of hormonal stimuli, with the major trophic effects being tics, and appropriate biomarker levels before initiating
modulated by estrogen, progesterone, and prolactin. local therapy.
4 Benign breast disorders and diseases are related to the nor- 9 Sentinel node dissection is the preferred method for stag-
mal processes of reproductive life and to involution, and ing of the regional lymph nodes in women with clinically
there is a spectrum of breast conditions that ranges from node-negative invasive breast cancer. Axillary dissection
normal to disorder to disease (aberrations of normal devel- may be avoided in women with one to two positive senti-
opment and involution classification). nel nodes who are treated with breast conserving surgery,
5 To calculate breast cancer risk using the Gail model, a whole breast radiation, and systemic therapy.
woman’s risk factors are translated into an overall risk 10 Local-regional and systemic therapy decisions for an indi-
score by multiplying her relative risks from several cat- vidual patient with breast cancer are best made using a
egories. This risk score is then compared with an adjusted multidisciplinary treatment approach. The sequencing of
population risk of breast cancer to determine the wom- therapies is dependent on patient and tumor related factors
an’s individual risk. This model is not appropriate for use including breast cancer subtype.

evidence and started an idea which persisted into the 20th nerve and the thoracodorsal neurovascular bundle with the axil-
century among some lay people. The majority of respected sur- lary contents. In 1943, Haagensen and Stout described the grave
geons considered operative intervention to be a futile and ill- signs of breast cancer, which included: (a) edema of the skin
advised endeavor. The Renaissance and the wars of the 16th and of the breast, (b) skin ulceration, (c) chest wall fixation, (d) an
17th centuries brought developments in surgery, particularly in axillary lymph node >2.5 cm in diameter, and (e) fixed axillary
anatomical understanding. However, there were no new theories lymph nodes. Women with two or more signs had a 42% local
espoused in relation to cancer. Beginning with Morgagni, surgi- recurrence rate and only a 2% 5-year disease-free survival rate.7
cal resections were more frequently undertaken, including some Based on these findings, they declared that women with grave
early attempts at mastectomy and axillary dissection. The 17th signs were beyond cure by radical surgery. In 1948, Patey and
century saw the start of the Age of Enlightenment, which lasted Dyson of the Middlesex Hospital, London, advocated a modi-
until the 19th century. In terms of medicine, this resulted in the fied radical mastectomy for the management of advanced oper-
abandonment of Galen’s humoral pathology, which was repudi- able breast cancer, explaining, “Until an effective general agent
ated by Le Dran, and the subsequent rise in cellular pathology for treatment of carcinoma of the breast is developed, a high
as espoused by Virchow. Le Dran stated that breast cancer was proportion of these cases are doomed to die.”8 Their technique
a local disease that spread by way of lymph vessels to axillary included removal of the breast and axillary lymph nodes with
lymph nodes. When operating on a woman with breast cancer, preservation of the pectoralis major muscle. They showed that
he routinely removed any enlarged axillary lymph nodes.4 removal of the pectoralis minor muscle allowed access to and
In the 19th century, Moore, of the Middlesex Hospital, clearance of axillary lymph node levels I to III.
London, emphasized complete resection of the breast for cancer During the 1970s, there was a transition from the Halsted
and stated that palpable axillary lymph nodes also should be radical mastectomy to the modified radical mastectomy as the
removed.5 In a presentation before the British Medical Asso- surgical procedure most frequently used by American surgeons
ciation in 1877, Banks supported Moore’s concepts and advo- to treat breast cancer. This transition acknowledged that: (a)
cated the resection of axillary lymph nodes even when palpable fewer patients were presenting with advanced local disease
lymphadenopathy was not evident, recognizing that occult with or without the grave signs described by Haagensen, (b)
involvement of axillary lymph nodes was frequently present. In extirpation of the pectoralis major muscle was not essential for
1894, Halsted and Meyer reported their operations for treatment local-regional control in stages I and II breast cancer, and (c)
of breast cancer.6 By demonstrating superior local-regional con- neither the modified radical mastectomy nor the Halsted radi-
trol rates after radical resection, these surgeons established radi- cal mastectomy consistently achieved local-regional control of
cal mastectomy as state-of-the-art treatment for that era. Halsted stage III breast cancer. Radiation therapy was incorporated into
and Meyer advocated complete dissection of axillary lymph the management of advanced breast cancer and demonstrated
542 node levels I to III. Both routinely resected the long thoracic improvements in local-regional control. The National Surgical
Adjuvant Breast and Bowel Project (NSABP) conducted a ran- regimens are superior to cyclophosphamide, methotrexate, and 543
domized trial in the early 1970s to determine the impact of local 5-fluorouracil (CMF), and more recently, that the addition of a
and regional treatments on survival in operable breast cancer. taxane to an anthracycline-based regimen reduces breast cancer
In the B-04 trial, 1665 women were enrolled and stratified by mortality by one-third.11 The overview has also demonstrated
clinical assessment of the axillary lymph nodes. The clinically that tamoxifen is of benefit only in patients with estrogen recep-
node-negative women were randomized into three treatment tor (ER) positive breast cancer and that tamoxifen may decrease
groups: (a) Halsted radical mastectomy; (b) total mastectomy mortality from breast cancer by as much as 30%.13 Importantly,
plus radiation therapy; and (c) total mastectomy alone. Clini- the EBCTCG data have shown that proportional reduction in
cally node-positive women were randomized to Halsted radical risk was not significantly affected by standard clinical and
mastectomy or total mastectomy plus radiation therapy. This pathologic factors such as tumor size, ER status, and nodal

CHAPTER 17 THE BREAST


trial accrued patients between 1971 and 1974, an era that pre- status.14 This underscores the importance of stratification of risk
dated widespread availability of effective systemic therapy for in determining adjuvant therapy decisions in order to minimize
breast cancer and therefore reflect survival associated with local- the toxicities of therapies in those unlikely to benefit, yet real-
regional therapy alone. There were no differences in survival ize the substantial benefits gained in local-regional control and
between the three groups of node-negative women or between survival in those at higher risk.
the two groups of node-positive women. These overall survival Many early randomized clinical trials considered all
equivalence patterns persisted at 25 years of follow-up.9 patients similarly in terms of treatment viewing breast cancer as
The next major advance in the surgical management more of a homogeneous disease. Breast cancer has traditionally
of breast cancer was the development of breast conserving been defined by pathologic determinants using conventional
surgery. Breast conserving surgery and radium treatment light microscopy and basic histologic techniques. In the 1980s,
was first reported by Geoffrey Keynes of St Bartholomew’s immunohistochemistry allowed assessment of the expression
Hospital, London in the British Medical Journal in 1937.10 of individual tumor markers (most commonly proteins) while
Several decades later, the NSABP launched the B-06 trial, a DNA was initially assessed in terms of its ploidy status. Sub-
phase 3 study that randomized 1851 patients to total mastec- sequently, breast cancer specimens have been interrogated at
tomy, lumpectomy alone, or lumpectomy with breast irradia- the level of the DNA by labeling genes of interest and allow-
tion. The results showed no difference in disease-free, distant ing fluorescent dyes to quantify the abundance of a particular
disease-free, and overall survival among the three groups; how- gene and comparing a large number of genes simultaneously in
ever, the omission of radiation therapy resulted in significantly a single breast cancer specimen. Gene expression arrays have
higher rates of ipsilateral breast tumor recurrence in those who shown that breast cancers cluster according to their intrinsic
received lumpectomy alone.11 The B-06 trial excluded patients gene expression patterns into at least five intrinsic subtypes and
who had palpable axillary lymph nodes, and those patients these intrinsic subtypes correlate with breast cancer outcomes.15
randomized to breast conserving surgery had frozen sections Breast cancers are now classified by molecular subtypes and
performed. If on frozen section the margins were involved, the these are being used for risk stratification and decision making
surgeon proceeded to perform a mastectomy, but the patient in terms of local-regional and systemic therapies.
was included in the analysis as having had a breast conserv- Currently, 50% of American women will consult a sur-
ing operation. Furthermore, in B-06, local in-breast recurrences geon regarding breast disease, 25% will undergo breast biopsy
were regarded as “nonevents” in terms of disease-free survival. for diagnosis of an abnormality, and 12% will develop some
Both the NSABP B-04 and B-06 trials were taken to refute the variant of breast cancer. Considerable progress has been made
Halstedian concept that cancer spread throughout a region of the in the integration of surgery, radiation therapy, and systemic
breast to lymphatics and then on to distant sites. Bernard Fisher therapy to control local-regional disease, enhance survival, and
proposed the “alternative hypothesis” that breast cancer was a improve the quality of life of breast cancer survivors. Surgeons
systemic disease at diagnosis and that tumor cells had access to are traditionally the first physician consulted for breast care, and
both the blood and lymphatic systems and that regional lymph it is critical for them to be well trained in all aspects of the breast
nodes were a marker of systemic disease and not a barrier to from embryologic development, to growth and development,
the dissemination of cancer cells. He proposed that host factors to benign and malignant disease processes. This will allow the
were important in the development of metastasis and that varia- greatest opportunity to achieve optimal outcomes for patients
tions in the local-regional approach to breast cancer treatment and their families.
were not likely to substantially impact survival. This idea was
dominant for a number of years but has been challenged by the
Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) EMBRYOLOGY AND FUNCTIONAL
overview analysis, which reported that “the avoidance of recur-
rence in a conserved breast . . . avoids about one breast cancer
ANATOMY OF THE BREAST
death over the next 15 years for every four such recurrences Embryology
avoided,”12 indicating that not all breast cancer is a systemic At the fifth or sixth week of fetal development, two ventral
disease at presentation. bands of thickened ectoderm (mammary ridges, milk lines)
During the 1970s, clinical trials were initiated to determine are evident in the embryo.16 In most mammals, paired breasts
the value of systemic therapy in the postoperative setting as an develop along these ridges, which extend from the base of
adjuvant to surgery. The EBCTCG was established in 1985 to the forelimb (future axilla) to the region of the hind limb
coordinate the meta-analysis of data from randomized clinical (inguinal area). These ridges are not prominent in the human
trials in order to examine the impact of adjuvant treatments embryo and disappear after a short time, except for small
for breast cancer on recurrence and mortality. The EBCTCG portions that may persist in the pectoral region. Accessory
overview has demonstrated that anthracycline containing breasts (polymastia) or accessory nipples (polythelia) may
544 and dysgenesis) and Fleischer’s syndrome (displacement of the
nipples and bilateral renal hypoplasia) may have polymastia as
a component. Accessory axillary breast tissue is uncommon and
usually is bilateral.
Functional Anatomy
The breast is composed of 15 to 20 lobes (Fig. 17-2), which
are each composed of several lobules.17 Fibrous bands of con-
nective tissue travel through the breast (Cooper’s suspensory
ligaments), insert perpendicularly into the dermis, and provide
structural support. The mature female breast extends from the
level of the second or third rib to the inframammary fold at
PART II

the sixth or seventh rib. It extends transversely from the lateral


border of the sternum to the anterior axillary line. The deep or
posterior surface of the breast rests on the fascia of the pecto-
ralis major, serratus anterior, and external oblique abdominal
muscles, and the upper extent of the rectus sheath. The retro-
SPECIFIC CONSIDERATIONS

mammary bursa may be identified on the posterior aspect of the


breast between the investing fascia of the breast and the fascia of
the pectoralis major muscles. The axillary tail of Spence extends
laterally across the anterior axillary fold. The upper outer quad-
rant of the breast contains a greater volume of tissue than do the
other quadrants. The breast has a protuberant conical form. The
Figure 17-1. The mammary milk line. (Visual Art: © 2013.
base of the cone is roughly circular, measuring 10 to 12 cm in
The University of Texas MD Anderson Cancer Center.)
diameter. Considerable variations in the size, contour, and den-
sity of the breast are evident among individuals. The nulliparous
breast has a hemispheric configuration with distinct flattening
occur along the milk line (Fig. 17-1) when normal regression above the nipple. With the hormonal stimulation that accom-
fails. Each breast develops when an ingrowth of ectoderm panies pregnancy and lactation, the breast becomes larger and
forms a primary tissue bud in the mesenchyme. The primary increases in volume and density, whereas with senescence, it
bud, in turn, initiates the development of 15 to 20 secondary assumes a flattened, flaccid, and more pendulous configuration
buds. Epithelial cords develop from the secondary buds and with decreased volume.
extend into the surrounding mesenchyme. Major (lactiferous) Nipple-Areola Complex. The epidermis of the nipple-are-
ducts develop, which open into a shallow mammary pit. Dur- ola complex is pigmented and is variably corrugated. During
ing infancy, a proliferation of mesenchyme transforms the puberty, the pigment becomes darker and the nipple assumes
mammary pit into a nipple. If there is failure of a pit to elevate an elevated configuration. Throughout pregnancy, the areola
above skin level, an inverted nipple results. This congenital
malformation occurs in 4% of infants. At birth, the breasts are
identical in males and females, demonstrating only the pres-
ence of major ducts. Enlargement of the breast may be evi-
dent, and a secretion, historically referred to as witch’s milk,
may be produced. These transitory events occur in response
to maternal hormones that cross the placenta.
The breast remains undeveloped in the female until
puberty, when it enlarges in response to ovarian estrogen and
progesterone, which initiate proliferation of the epithelial
and connective tissue elements. However, the breasts remain
incompletely developed until pregnancy occurs. Absence of
the breast (amastia) is rare and results from an arrest in mam-
mary ridge development that occurs during the sixth fetal week.
Poland’s syndrome consists of hypoplasia or complete absence
of the breast, costal cartilage and rib defects, hypoplasia of the
subcutaneous tissues of the chest wall, and brachysyndactyly.
Breast hypoplasia also may be iatrogenically induced before
puberty by trauma, infection, or radiation therapy. Symmastia
is a rare anomaly recognized as webbing between the breasts
across the midline. Accessory nipples (polythelia) occur in
<1% of infants and may be associated with abnormalities of Figure 17-2. Anatomy of the breast. Tangential and cross-
the urinary and cardiovascular systems. Supernumerary breasts sectional (sagittal) views of the breast and associated chest wall.
may occur in any configuration along the mammary milk line (Reproduced with permission from Bland KI, Copeland EMI: The
but most frequently occur between the normal nipple location Breast: Comprehensive Management of Benign and Malignant
and the symphysis pubis. Turner’s syndrome (ovarian agenesis Diseases, 4th ed. Philadelphia, PA: Elsevier/Saunders; 2009.)
enlarges and pigmentation is further enhanced. The areola con- 545
tains sebaceous glands, sweat glands, and accessory glands,
which produce small elevations on the surface of the areola
(Montgomery’s tubercles). Smooth muscle bundle fibers, which
lie circumferentially in the dense connective tissue and longi-
tudinally along the major ducts, extend upward into the nipple,
where they are responsible for the nipple erection that occurs
with various sensory stimuli. The dermal papilla at the tip of
the nipple contains numerous sensory nerve endings and Meiss-
ner’s corpuscles. This rich sensory innervation is of functional

CHAPTER 17 THE BREAST


importance because the sucking of the infant initiates a chain of
neurohumoral events that results in milk letdown.
Inactive and Active Breast. Each lobe of the breast termi-
nates in a major (lactiferous) duct (2–4 mm in diameter), which
opens through a constricted orifice (0.4–0.7 mm in diameter)
into the ampulla of the nipple (see Fig. 17-2). Immediately
below the nipple-areola complex, each major duct has a dilated Figure 17-4. Active human breast: pregnancy and lactation (160x).
portion (lactiferous sinus), which is lined with stratified squa- The alveolar epithelium becomes conspicuous during the early pro-
mous epithelium. Major ducts are lined with two layers of liferative period. The alveolus is surrounded by cellular connective
cuboidal cells, whereas minor ducts are lined with a single layer tissue. (Used with permission from Dr. Sindhu Menon, Consultant
of columnar or cuboidal cells. Myoepithelial cells of ectoder- Histopathologist and Dr. Rahul Deb, Consultant Histopathologist
mal origin reside between the epithelial cells in the basal lamina and Lead Breast Pathologist, Royal Derby Hospital, Derby, UK.)
and contain myofibrils. In the inactive breast, the epithelium is
sparse and consists primarily of ductal epithelium (Fig. 17-3).
In the early phase of the menstrual cycle, minor ducts are cord-
like with small lumina. With estrogen stimulation at the time of epithelium and accumulation of secretory products in the lumina
ovulation, alveolar epithelium increases in height, duct lumina of the minor ducts. Alveolar epithelium contains abundant endo-
become more prominent, and some secretions accumulate. plasmic reticulum, large mitochondria, Golgi complexes, and
When the hormonal stimulation decreases, the alveolar epithe- dense lysosomes. Two distinct substances are produced by the
lium regresses. alveolar epithelium: (a) the protein component of milk, which is
With pregnancy, the breast undergoes proliferative and synthesized in the endoplasmic reticulum (merocrine secretion);
developmental maturation. As the breast enlarges in response and (b) the lipid component of milk (apocrine secretion), which
to hormonal stimulation, lymphocytes, plasma cells, and eosin- forms as free lipid droplets in the cytoplasm. Milk released in
ophils accumulate within the connective tissues. The minor the first few days after parturition is called colostrum and has
ducts branch and alveoli develop. Development of the alveoli low lipid content but contains considerable quantities of anti-
is asymmetric, and variations in the degree of development bodies. The lymphocytes and plasma cells that accumulate
may occur within a single lobule (Fig. 17-4). With parturition, within the connective tissues of the breast are the source of the
enlargement of the breasts occurs via hypertrophy of alveolar antibody component. With subsequent reduction in the number
of these cells, the production of colostrum decreases and lipid-
rich milk is released.
Blood Supply, Innervation, and Lymphatics. The breast
receives its principal blood supply from: (a) perforating branches
of the internal mammary artery; (b) lateral branches of the poste-
rior intercostal arteries; and (c) branches from the axillary artery,
including the highest thoracic, lateral thoracic, and pectoral
branches of the thoracoacromial artery (Fig. 17-5). The second,
1 third, and fourth anterior intercostal perforators and
branches of the internal mammary artery arborize in the
breast as the medial mammary arteries. The lateral thoracic artery
gives off branches to the serratus anterior, pectoralis major and
pectoralis minor, and subscapularis muscles. It also gives rise to
lateral mammary branches. The veins of the breast and chest wall
follow the course of the arteries, with venous drainage being
toward the axilla. The three principal groups of veins are: (a) per-
forating branches of the internal thoracic vein, (b) perforating
Figure 17-3. Inactive human breast (100x). The epithelium, which branches of the posterior intercostal veins, and (c) tributaries of
is primarily ductal, is embedded in loose connective tissue. Dense the axillary vein. Batson’s vertebral venous plexus, which invests
connective tissue surrounds the terminal duct lobular units (TDLU). the vertebrae and extends from the base of the skull to the sacrum,
(Used with permission from Dr. Sindhu Menon, Consultant Histo- may provide a route for breast cancer metastases to the vertebrae,
pathologist and Dr. Rahul Deb, Consultant Histopathologist and skull, pelvic bones, and central nervous system. Lymph vessels
Lead Breast Pathologist, Royal Derby Hospital, Derby, UK.) generally parallel the course of blood vessels.
546
PART II
SPECIFIC CONSIDERATIONS

Figure 17-5. Arterial supply to the breast, axilla, and chest wall.
(Reproduced with permission from Bland KI, Copeland EMI: The
Breast: Comprehensive Management of Benign and Malignant
Diseases, 4th ed. Philadelphia, PA: Elsevier/Saunders; 2009.)

Lateral cutaneous branches of the third through sixth inter- Figure 17-6. Lymphatic pathways of the breast. Arrows indicate
costal nerves provide sensory innervation of the breast (lateral the direction of lymph flow. (Visual Art: © 2013. The University of
mammary branches) and of the anterolateral chest wall. These Texas MD Anderson Cancer Center.)
branches exit the intercostal spaces between slips of the serratus
anterior muscle. Cutaneous branches that arise from the cervical interpectoral group (Rotter’s lymph nodes), which consists of
plexus, specifically the anterior branches of the supraclavicular one to four lymph nodes that are interposed between the pec-
nerve, supply a limited area of skin over the upper portion of toralis major and pectoralis minor muscles and receive lymph
the breast. The intercostobrachial nerve is the lateral cutane- drainage directly from the breast. The lymph fluid that passes
ous branch of the second intercostal nerve and may be visual-
ized during surgical dissection of the axilla. Resection of the
intercostobrachial nerve causes loss of sensation over the medial
aspect of the upper arm.
The boundaries for lymph drainage of the axilla are not
well demarcated, and there is considerable variation in the posi-
tion of the axillary lymph nodes. The six axillary lymph node
groups recognized by surgeons (Figs. 17-6 and 17-7) are: (a) the
axillary vein group (lateral), which consists of four to six lymph
nodes that lie medial or posterior to the vein and receive most
of the lymph drainage from the upper extremity; (b) the external
mammary group (anterior or pectoral group), which consists of
five to six lymph nodes that lie along the lower border of the
pectoralis minor muscle contiguous with the lateral thoracic
vessels and receive most of the lymph drainage from the lat-
eral aspect of the breast; (c) the scapular group (posterior or
subscapular), which consists of five to seven lymph nodes that
lie along the posterior wall of the axilla at the lateral border of
the scapula contiguous with the subscapular vessels and receive
lymph drainage principally from the lower posterior neck, the
posterior trunk, and the posterior shoulder; (d) the central group,
which consists of three or four sets of lymph nodes that are
embedded in the fat of the axilla lying immediately posterior to
the pectoralis minor muscle and receive lymph drainage both Figure 17-7. Axillary lymph node groups. Level I includes
from the axillary vein, external mammary, and scapular groups lymph nodes located lateral to the pectoralis minor muscle; level II
of lymph nodes, and directly from the breast; (e) the subcla- includes lymph nodes located deep to the pectoralis minor; and
vicular group (apical), which consists of six to twelve sets of level III includes lymph nodes located medial to the pectoralis
lymph nodes that lie posterior and superior to the upper bor- minor. The axillary vein with its major tributaries and the supracla-
der of the pectoralis minor muscle and receive lymph drainage vicular lymph node group are also illustrated. (Visual Art: © 2013.
from all of the other groups of axillary lymph nodes; and (f) the The University of Texas MD Anderson Cancer Center.)
through the interpectoral group of lymph nodes passes directly PHYSIOLOGY OF THE BREAST 547
into the central and subclavicular groups.
As indicated in Fig. 17-7, the lymph node groups are Breast Development and Function
assigned levels according to their anatomic relationship to the Breast development and function are initiated by a variety of
pectoralis minor muscle. Lymph nodes located lateral to or below hormonal stimuli, including estrogen, progesterone, prolactin,
the lower border of the pectoralis minor muscle are referred to as oxytocin, thyroid hormone, cortisol, and growth hormone.17,18
level I lymph nodes, which include the axillary vein, external Estrogen, progesterone, and prolactin especially have profound
mammary, and scapular groups. Lymph nodes located superficial 3 trophic effects that are essential to normal breast develop-
ment and function. Estrogen initiates ductal development,
or deep to the pectoralis minor muscle are referred to as level II
lymph nodes, which include the central and interpectoral groups. whereas progesterone is responsible for differentiation of epithe-
lium and for lobular development. Prolactin is the primary hor-

CHAPTER 17 THE BREAST


Lymph nodes located medial to or above the upper border of the
pectoralis minor muscle are referred to as level III lymph nodes, monal stimulus for lactogenesis in late pregnancy and the
which consist of the subclavicular group. The plexus of lymph postpartum period. It upregulates hormone receptors and stimu-
vessels in the breast arises in the interlobular connective tissue lates epithelial development. Fig. 17-8 depicts the secretion of
and in the walls of the lactiferous ducts and communicates with neurotrophic hormones from the hypothalamus, which is respon-
the subareolar plexus of lymph vessels. Efferent lymph vessels sible for regulation of the secretion of the hormones that affect
from the breast pass around the lateral edge of the pectoralis the breast tissues. The gonadotropins luteinizing hormone (LH)
major muscle and pierce the clavipectoral fascia, ending in the and follicle-stimulating hormone (FSH) regulate the release of
external mammary (anterior, pectoral) group of lymph nodes. estrogen and progesterone from the ovaries. In turn, the release
Some lymph vessels may travel directly to the subscapular (pos- of LH and FSH from the basophilic cells of the anterior pituitary
terior, scapular) group of lymph nodes. From the upper part of the is regulated by the secretion of gonadotropin-releasing hormone
breast, a few lymph vessels pass directly to the subclavicular (api- (GnRH) from the hypothalamus. Positive and negative feedback
cal) group of lymph nodes. The axillary lymph nodes usually effects of circulating estrogen and progesterone regulate the
secretion of LH, FSH, and GnRH. These hormones are respon-
2 receive >75% of the lymph drainage from the breast. The
rest is derived primarily from the medial aspect of the sible for the development, function, and maintenance of breast
breast, flows through the lymph vessels that accompany the per- tissues (Fig. 17-9A). In the female neonate, circulating estrogen
forating branches of the internal mammary artery, and enters the and progesterone levels decrease after birth and remain low
parasternal (internal mammary) group of lymph nodes. throughout childhood because of the sensitivity of

H
-R
F LH
H

DH

GR
ne
TR
mi
y/A
F
CR

pa
Ox
Do

Figure 17-8. Overview of the neuroendocrine con-


trol of breast development and function. ADH =
antidiuretic hormone; CRF = corticotropin-releasing
factor; GRF = growth hormone releasing factor;
LH-RH = luteinizing hormone–releasing hormone;
Oxy = oxytocin; TRH = thyrotropin-releasing hor-
mone. (Reproduced with permission from Bland KI,
Copeland EMI: The Breast: Comprehensive Man-
agement of Benign and Malignant Diseases, 4th ed.
Philadelphia, PA: Elsevier/Saunders; 2009.)
548

A
PART II
SPECIFIC CONSIDERATIONS

Figure 17-9. The breast at different physi-


ologic stages. The central column contains
three-dimensional depictions of microscopic
structures. A. Adolescence. B. Pregnancy.
D C. Lactation. D. Senescence.

the hypothalamic-pituitary axis to negative feedback from these In the first and second trimesters, the minor ducts branch and
hormones. With the onset of puberty, there is a decrease in the develop. During the third trimester, fat droplets accumulate in
sensitivity of the hypothalamic-pituitary axis to negative feed- the alveolar epithelium, and colostrum fills the alveolar and duc-
back and an increase in its sensitivity to positive feedback from tal spaces. In late pregnancy, prolactin stimulates the synthesis
estrogen. These physiologic events initiate an increase in GnRH, of milk fats and proteins.
FSH, and LH secretion and ultimately an increase in estrogen After delivery of the placenta, circulating progesterone
and progesterone secretion by the ovaries, leading to establish- and estrogen levels decrease, permitting full expression of the
ment of the menstrual cycle. At the beginning of the menstrual lactogenic action of prolactin. Milk production and release are
cycle, there is an increase in the size and density of the breasts, controlled by neural reflex arcs that originate in nerve endings
which is followed by engorgement of the breast tissues and epi- of the nipple-areola complex. Maintenance of lactation requires
thelial proliferation. With the onset of menstruation, the breast regular stimulation of these neural reflexes, which results in
engorgement subsides and epithelial proliferation decreases. prolactin secretion and milk letdown. Oxytocin release results
from the auditory, visual, and olfactory stimuli associated with
Pregnancy, Lactation, and Senescence nursing. Oxytocin initiates contraction of the myoepithelial
A dramatic increase in circulating ovarian and placental estro- cells, which results in compression of alveoli and expulsion of
gens and progestins is evident during pregnancy, which initiates milk into the lactiferous sinuses. After weaning of the infant,
striking alterations in the form and substance of the breast (see prolactin and oxytocin release decreases. Dormant milk causes
Fig. 17-9B).17-19 The breast enlarges as the ductal and lobular increased pressure within the ducts and alveoli, which results in
epithelium proliferates, the areolar skin darkens, and the acces- atrophy of the epithelium (Fig. 17-9C). With menopause, there
sory areolar glands (Montgomery’s glands) become prominent. is a decrease in the secretion of estrogen and progesterone by
the ovaries and involution of the ducts and alveoli of the breast.
Table 17-1 549
The surrounding fibrous connective tissue increases in density,
and breast tissues are replaced by adipose tissues (Fig. 17-9D). Pathophysiologic mechanisms of gynecomastia
I. Estrogen excess states
Gynecomastia A. Gonadal origin
Gynecomastia refers to an enlarged breast in the male.20 Physi- 1. True hermaphroditism
ologic gynecomastia usually occurs during three phases of life: 2. Gonadal stromal (nongerminal) neoplasms of
the neonatal period, adolescence, and senescence. Common to the testis
each of these phases is an excess of circulating estrogens in a. Leydig cell (interstitial)
relation to circulating testosterone. Neonatal gynecomastia is b. Sertoli cell
caused by the action of placental estrogens on neonatal breast

CHAPTER 17 THE BREAST


c. Granulosa-theca cell
tissues, whereas in adolescence, there is an excess of estradiol 3. Germ cell tumors
relative to testosterone, and with senescence, the circulating a. Choriocarcinoma
testosterone level falls, which results in relative hyperestrin- b. Seminoma, teratoma
ism. In gynecomastia, the ductal structures of the male breast c. Embryonal carcinoma
enlarge, elongate, and branch with a concomitant increase in B. Nontesticular tumors
epithelium. During puberty, the condition often is unilateral 1. Adrenal cortical neoplasms
and typically occurs between ages 12 and 15 years. In contrast, 2. Lung carcinoma
senescent gynecomastia is usually bilateral. In the nonobese 3. Hepatocellular carcinoma
male, breast tissue measuring at least 2 cm in diameter must C. Endocrine disorders
be present before a diagnosis of gynecomastia may be made. D. Diseases of the liver—nonalcoholic and alcoholic
Mammography and ultrasonography are used to differentiate cirrhosis
breast tissues. Dominant masses or areas of firmness, irregular- E. Nutrition alteration states
ity, and asymmetry suggest the possibility of a breast cancer, II. Androgen deficiency states
particularly in the older male. Gynecomastia generally does not A. Senescence
predispose the male breast to cancer. However, the hypoandro- B. Hypoandrogenic states (hypogonadism)
genic state of Klinefelter’s syndrome (XXY), in which gyneco- 1. Primary testicular failure
mastia is usually evident, is associated with an increased risk of a. Klinefelter’s syndrome (XXY)
breast cancer. Gynecomastia is graded based on the degree of b. Reifenstein’s syndrome
breast enlargement, the position of the nipple with reference to c. Rosewater-Gwinup-Hamwi familial
the inframammary fold, and the degree of breast ptosis and skin gynecomastia
redundancy: Grade I—mild breast enlargement without skin d. Kallmann syndrome
redundancy; Grade IIa—moderate breast enlargement without e. Kennedy’s disease with associated
skin redundancy; Grade IIb—moderate breast enlargement with gynecomastia
skin redundancy; and Grade III—marked breast enlargement f. Eunuchoidal state (congenital anorchia)
with skin redundancy and ptosis. g. Hereditary defects of androgen biosynthesis
Table 17-1 identifies the pathophysiologic mechanisms h. Adrenocorticotropic hormone deficiency
that may initiate gynecomastia: estrogen excess states; andro- 2. Secondary testicular failure
gen deficiency states; pharmacologic causes; and idiopathic a. Trauma
causes. Estrogen excess results from an increase in the secretion b. Orchitis
of estradiol by the testicles or by nontesticular tumors, nutri- c. Cryptorchidism
tional alterations such as protein and fat deprivation, endocrine d. Irradiation
disorders (hyperthyroidism, hypothyroidism), and hepatic dis- C. Renal failure
ease (nonalcoholic and alcoholic cirrhosis). Refeeding gyne- III. Pharmacologic causes
comastia is related to the resumption of pituitary gonadotropin IV. Systemic diseases with idiopathic mechanisms
secretion after pituitary shutdown. Androgen deficiency may
initiate gynecomastia. Concurrently occurring with decreased
circulating testosterone levels is an elevated level of circulating
testosterone-binding globulin, which results in a reduction of spironolactone, antineoplastic agents, diazepam) also have been
free testosterone. This senescent gynecomastia usually occurs implicated. Drugs such as reserpine, theophylline, verapamil,
in men age 50 to 70 years. Hypoandrogenic states can be from tricyclic antidepressants, and furosemide induce gynecomastia
primary testicular failure or secondary testicular failure. Kline- through idiopathic mechanisms.
felter’s syndrome (XXY) is an example of primary testicular When gynecomastia is caused by androgen deficiency,
failure that is manifested by gynecomastia, hypergonadotropic then testosterone administration may cause regression. When it
hypogonadism, and azoospermia. Secondary testicular failure is caused by medications, then these are discontinued if possi-
may result from trauma, orchitis, and cryptorchidism. Renal ble. When endocrine defects are responsible, then these receive
failure, regardless of cause, also may initiate gynecomastia. specific therapy. As soon as gynecomastia is progressive and
Pharmacologic causes of gynecomastia include drugs does not respond to other treatments, surgical therapy is con-
with estrogenic activity (digitalis, estrogens, anabolic steroids, sidered. Techniques include local excision, liposuction or sub-
marijuana) or drugs that enhance estrogen synthesis (human cutaneous mastectomy. Attempts to reverse gynecomastia with
chorionic gonadotropin). Drugs that inhibit the action or syn- danazol have been successful, but the androgenic side effects of
thesis of testosterone (cimetidine, ketoconazole, phenytoin, the drug are considerable.
550 INFECTIOUS AND INFLAMMATORY DISORDERS Zuska’s disease, also called recurrent periductal
OF THE BREAST mastitis, is a condition of recurrent retroareolar infections and
abscesses.22,23 Smoking has been implicated as a risk factor for
Infections in the postpartum period remain proportionately the this condition.24,25 This syndrome is managed symptomatically
most common time for breast infections to occur. Infections of by antibiotics coupled with incision and drainage as necessary.
the breast unrelated to lactation are proportionately less com- Attempts to obtain durable long-term control by wide debride-
mon, however, are still a relatively common presentation to ment of chronically infected tissue and/or terminal duct resec-
breast specialists. The latter are classified as intrinsic (second- tion have been reported and can be curative, but they can also
ary to abnormalities in the breast) or extrinsic (secondary to an be frustrated by postoperative infections.26
infection in an adjacent structure, e.g., skin, thoracic cavity) the
most common being probably periductal mastitis and infected Mycotic Infections
sebaceous cysts, respectively. Fungal infections of the breast are rare and usually involve blas-
PART II

Bacterial Infection tomycosis or sporotrichosis.27 Intraoral fungi that are inoculated


Staphylococcus aureus and Streptococcus species are the into the breast tissue by the suckling infant initiate these infec-
organisms most frequently recovered from nipple discharge tions, which present as mammary abscesses in close proxim-
from an infected breast.17 Typically breast abscesses are seen ity to the nipple-areola complex. Pus mixed with blood may be
expressed from sinus tracts. Antifungal agents can be adminis-
SPECIFIC CONSIDERATIONS

in staphylococcal infections and present with point tenderness,


erythema, and hyperthermia. When these abscesses are related tered for the treatment of systemic (noncutaneous) infections.
to lactation they usually occur within the first few weeks of This therapy generally eliminates the necessity of surgical inter-
breastfeeding. If there is progression of a staphylococcal infec- vention, but occasionally drainage of an abscess, or even partial
tion, this may result in subcutaneous, subareolar, interlobular mastectomy, may be necessary to eradicate a persistent fungal
(periductal), and retromammary abscesses (unicentric or multi- infection. Candida albicans affecting the skin of the breast
centric). Previously almost all breast abscesses were treated by presents as erythematous, scaly lesions of the inframammary
operative incision and drainage, but now the initial approach is or axillary folds. Scrapings from the lesions demonstrate fungal
antibiotics and repeated aspiration of the abscess, usually ultra- elements (filaments and binding cells). Therapy involves the
sound-guided aspiration.21 Operative drainage is now reserved removal of predisposing factors such as maceration and the topi-
for those cases that do not resolve with repeated aspiration and cal application of nystatin.
antibiotic therapy or cases in which there is some other indica- Hidradenitis Suppurativa
tion for incision and drainage (e.g., thinning or necrosis of the Hidradenitis suppurativa of the nipple-areola complex or axilla
overlying skin). Preoperative ultrasonography is effective in is a chronic inflammatory condition that originates within the
delineating the required extent of the drainage procedure. While accessory areolar glands of Montgomery or within the axillary
staphylococcal infections tend to be more localized and may sebaceous glands.27 Women with chronic acne are predisposed
be situated deep in the breast tissues, streptococcal infections to developing hidradenitis. When located in and about the
usually present with diffuse superficial involvement. They are nipple-areola complex, this disease may mimic other chronic
treated with local wound care, including application of warm inflammatory states, Paget’s disease of the nipple, or invasive
compresses, and the administration of IV antibiotics (penicillins breast cancer. Involvement of the axillary skin is often multifo-
or cephalosporins). Breast infections may be chronic, possibly cal and contiguous. Antibiotic therapy with incision and drain-
with recurrent abscess formation. In this situation, cultures are age of fluctuant areas is appropriate treatment. Excision of the
performed to identify acid-fast bacilli, anaerobic and aerobic involved areas may be required. Large areas of skin loss may
bacteria, and fungi. Uncommon organisms may be encountered, necessitate coverage with advancement flaps or split-thickness
and long-term antibiotic therapy may be required. skin grafts.
Biopsy of the abscess cavity wall should be considered at
the time of incision and drainage to rule out underlying breast Mondor’s Disease
cancer in patients where antibiotics and drainage have been Mondor’s disease is a variant of thrombophlebitis that involves
ineffective. the superficial veins of the anterior chest wall and breast.28 In
Nowadays hospital-acquired puerperal infections of the 1939, Mondor described the condition as “string phlebitis,” a
breast are much less common, but nursing women who pres- thrombosed vein presenting as a tender, cord-like structure.29
ent with milk stasis or noninfectious inflammation may still Frequently involved veins include the lateral thoracic vein, the
develop this problem. Epidemic puerperal mastitis is initiated thoracoepigastric vein, and, less commonly, the superficial epi-
by highly virulent strains of methicillin-resistant S aureus that gastric vein. Typically, a woman presents with acute pain in
are transmitted via the suckling neonate and may result in sub- the lateral aspect of the breast or the anterior chest wall. A ten-
stantial morbidity and occasional mortality. Purulent fluid may der, firm cord is found to follow the distribution of one of the
be expressed from the nipple. In this circumstance, breastfeed- major superficial veins. Rarely, the presentation is bilateral, and
ing is stopped, antibiotics are started, and surgical therapy is most women have no evidence of thrombophlebitis in other ana-
initiated. Nonepidemic (sporadic) puerperal mastitis refers to tomic sites. This benign, self-limited disorder is not indicative
involvement of the interlobular connective tissue of the breast of a cancer. When the diagnosis is uncertain, or when a mass is
by an infectious process. The patient develops nipple fissuring present near the tender cord, biopsy is indicated. Therapy for
and milk stasis, which initiates a retrograde bacterial infection. Mondor’s disease includes the liberal use of anti-inflammatory
Emptying of the breast using breast suction pumps shortens the medications and application of warm compresses along the
duration of symptoms and reduces the incidence of recurrences. symptomatic vein. The process usually resolves within 4 to
The addition of antibiotic therapy results in a satisfactory out- 6 weeks. When symptoms persist or are refractory to therapy,
come in >95% of cases. excision of the involved vein segment may be considered.
COMMON BENIGN DISORDERS AND 551
DISEASES OF THE BREAST
Benign breast disorders and diseases encompass a wide range
of clinical and pathologic entities. Surgeons require an in-depth
understanding of benign breast disorders and diseases so that clear
explanations may be given to affected women, appropriate treat-
ment is instituted, and unnecessary long-term follow up is avoided.

Aberrations of Normal Development and


Involution

CHAPTER 17 THE BREAST


The basic principles underlying the aberrations of normal devel-
opment and involution (ANDI) classification of benign breast
conditions are the following: (a) benign breast disorders and
diseases are related to the normal processes of reproductive life
and to involution; (b) there is a spectrum of breast conditions
that ranges from normal to disorder to disease; and (c) the ANDI
classification encompasses all aspects of the breast condition, Figure 17-10. Fibroadenoma (40x). These benign tumors are typi-
cally well circumscribed and are comprised of both stromal and
4 including pathogenesis and the degree of abnormality.30
The horizontal component of Table 17-2 defines ANDI glandular elements. (Used with permission from Dr. Sindhu Menon,
along a spectrum from normal, to mild abnormality (disorder), Consultant Histopathologist and Dr. Rahul Deb, Consultant
to severe abnormality (disease). The vertical component indi- Histopathologist and Lead Breast Pathologist, Royal Derby Hospital,
cates the period during which the condition develops. Derby, UK.)
Early Reproductive Years. Fibroadenomas are seen and pres-
ent symptomatically predominantly in younger women age 15
to 25 years (Fig. 17-10).31 Fibroadenomas usually grow to 1 or fibroadenomas are sometimes found in an older screened popu-
2 cm in diameter and then are stable but may grow to a larger lation. The precise etiology of adolescent breast hypertrophy is
size. Small fibroadenomas (≤1 cm in size) are considered nor- unknown. A spectrum of changes from limited to massive stro-
mal, whereas larger fibroadenomas (≤3 cm) are disorders, and mal hyperplasia (gigantomastia) is seen. Nipple inversion is a
giant fibroadenomas (>3 cm) are disease. Similarly, multiple disorder of development of the major ducts, which prevents nor-
fibroadenomas (more than five lesions in one breast) are very mal protrusion of the nipple. Mammary duct fistulas arise when
uncommon and are considered disease. It is noted that with nipple inversion predisposes to major duct obstruction, leading
the introduction of mammographic screening, asymptomatic to recurrent subareolar abscess and mammary duct fistula.

Table 17-2
ANDI classification of benign breast disorders
  NORMAL DISORDER DISEASE
Early reproductive years Lobular development Fibroadenoma Giant fibroadenoma
(age 15–25 y)  Stromal development Adolescent hypertrophy Gigantomastia
  Nipple eversion Nipple inversion Subareolar abscess
      Mammary duct fistula
Later reproductive years Cyclical changes of Cyclical mastalgia Incapacitating mastalgia
(age 25–40 y) menstruation
    Nodularity  
  Epithelial hyperplasia of Bloody nipple discharge  
pregnancy
Involution (age 35–55 y) Lobular involution Macrocysts —
    Sclerosing lesions  
  Duct involution    
  Dilatation Duct ectasia Periductal mastitis
  Sclerosis Nipple retraction —
  Epithelial turnover Epithelial hyperplasia Epithelial hyperplasia with atypia
ANDI = aberrations of normal development and involution.
Reproduced with permission from Mansel RE, Webster D, Sweetland H: Hughes, Mansel & Webster’s Benign Disorders and Diseases of the Breast, 3rd ed.
London: Elsevier/Saunders; 2009.
552 Later Reproductive Years. Cyclical mastalgia and nodular- Table 17-3
ity usually are associated with premenstrual enlargement of the
breast and are regarded as normal. Cyclical pronounced mastal- Cancer risk associated with benign breast disorders and
gia and severe painful nodularity are viewed differently than are in situ carcinoma of the breast
physiologic discomfort and lumpiness. Painful nodularity that
persists for >1 week of the menstrual cycle is considered a disor- ABNORMALITY RELATIVE RISK
der. In epithelial hyperplasia of pregnancy, papillary projections Nonproliferative lesions of the breast No increased risk
sometimes give rise to bilateral bloody nipple discharge. Sclerosing adenosis No increased risk
Involution. Involution of lobular epithelium is dependent on Intraductal papilloma No increased risk
the specialized stroma around it. However, an integrated invo-
Florid hyperplasia 1.5 to 2-fold
lution of breast stroma and epithelium is not always seen, and
PART II

disorders of the process are common. When the stroma invo- Atypical lobular hyperplasia 4-fold
lutes too quickly, alveoli remain and form microcysts, which are Atypical ductal hyperplasia 4-fold
precursors of macrocysts. The macrocysts are common, often Ductal involvement by cells of 7-fold
subclinical, and do not require specific treatment. Sclerosing atypical ductal hyperplasia
adenosis is considered a disorder of both the proliferative and
Lobular carcinoma in situ 10-fold
SPECIFIC CONSIDERATIONS

the involutional phases of the breast cycle. Duct ectasia (dilated


ducts) and periductal mastitis are other important components Ductal carcinoma in situ 10-fold
of the ANDI classification. Periductal fibrosis is a sequela of Data from Dupont WD, Page DL. Risk factors for breast cancer in
periductal mastitis and may result in nipple retraction. About women with proliferative breast disease, N Engl J Med. 1985 Jan 17;
60% of women ≥70 years of age exhibit some degree of epi- 312(3):146-151.
thelial hyperplasia (Fig. 17-11). Atypical proliferative diseases

include ductal and lobular hyperplasia, both of which display


some features of carcinoma in situ. Women with atypical ductal
or lobular hyperplasia have a fourfold increase in breast cancer
risk (Table 17-3).
Pathology of Nonproliferative Disorders
Of paramount importance for the optimal management of
benign breast disorders and diseases is the histologic differentia-
tion of benign, atypical, and malignant changes.32,33 Determin-
ing the clinical significance of these changes is a problem that
is compounded by inconsistent nomenclature. The classifica-
tion system originally developed by Page separates the various
types of benign breast disorders and diseases into three clini-
cally relevant groups: nonproliferative disorders, proliferative
disorders without atypia, and proliferative disorders with atypia
(Table 17-4). Nonproliferative disorders of the breast account
A
for 70% of benign breast conditions and carry no increased risk

Table 17-4
Classification of benign breast disorders
Nonproliferative disorders of the breast
Cysts and apocrine metaplasia
Duct ectasia
Mild ductal epithelial hyperplasia
Calcifications
Fibroadenoma and related lesions
Proliferative breast disorders without atypia
Sclerosing adenosis
Radial and complex sclerosing lesions
Ductal epithelial hyperplasia
B Intraductal papillomas
Atypical proliferative lesions
Figure 17-11. A. Ductal epithelial hyperplasia. The irregular intra-
Atypical lobular hyperplasia
cellular spaces and variable cell nuclei distinguish this process from
carcinoma in situ. B. Lobular hyperplasia. The presence of alveo- Atypical ductal hyperplasia
lar lumina and incomplete distention distinguish this process from Data from Godfrey SE: Is fibrocystic disease of the breast precancerous?
carcinoma in situ. (Used with permission from Dr. R.L. Hackett.) Arch Pathol Lab Med. 1986 Nov;110(11):991.
for the development of breast cancer. This category includes concordant. Central sclerosis and various degrees of epithelial 553
cysts, duct ectasia, periductal mastitis, calcifications, fibroad- proliferation, apocrine metaplasia, and papilloma formation
enomas, and related disorders. characterize radial scars and complex sclerosing lesions of the
Breast macrocysts are an involutional disorder, have a breast. Lesions up to 1 cm in diameter are called radial scars,
high frequency of occurrence, and are often multiple. Duct ecta- whereas larger lesions are called complex sclerosing lesions.
sia is a clinical syndrome characterized by dilated subareolar Radial scars originate at sites of terminal duct branching where
ducts that are palpable and often associated with thick nipple the characteristic histologic changes radiate from a central area
discharge. Haagensen regarded duct ectasia as a primary event of fibrosis. All of the histologic features of a radial scar are seen
that led to stagnation of secretions, epithelial ulceration, and in the larger complex sclerosing lesions, but there is a greater
leakage of duct secretions (containing chemically irritating fatty disturbance of structure with papilloma formation, apocrine

CHAPTER 17 THE BREAST


acids) into periductal tissue.34 This sequence was thought to pro- metaplasia, and occasionally sclerosing adenosis. Distinguish-
duce a local inflammatory process with periductal fibrosis and ing between a radial scar and invasive breast carcinoma can be
subsequent nipple retraction. An alternative theory considers challenging based on core-needle biopsy sampling. Often the
periductal mastitis to be the primary process, which leads to imaging features of a radial scar (which can be quite similar to
weakening of the ducts and secondary dilatation. It is possible an invasive cancer) will dictate the need for either a vacuum-
that both processes occur and together explain the wide spec- assisted biopsy or surgical excision in order to exclude the pos-
trum of problems seen, which include nipple discharge, nipple sibility of carcinoma.
retraction, inflammatory masses, and abscesses. Mild ductal hyperplasia is characterized by the presence of
Calcium deposits are frequently encountered in the breast. three or four cell layers above the basement membrane. Moder-
Most are benign and are caused by cellular secretions and ate ductal hyperplasia is characterized by the presence of five
debris or by trauma and inflammation. Calcifications that are or more cell layers above the basement membrane. Florid duc-
associated with cancer include microcalcifications, which vary tal epithelial hyperplasia occupies at least 70% of a minor duct
in shape and density and are <0.5 mm in size, and fine, linear lumen. It is found in >20% of breast tissue specimens, is either
calcifications, which may show branching. Fibroadenomas have solid or papillary, and is associated with an increased cancer
abundant stroma with histologically normal cellular elements. risk (see Table 17-3). Intraductal papillomas arise in the major
They show hormonal dependence similar to that of normal ducts, usually in premenopausal women. They generally are
breast lobules in that they lactate during pregnancy and invo- <0.5 cm in diameter but may be as large as 5 cm. A common
lute in the postmenopausal period. Adenomas of the breast are presenting symptom is nipple discharge, which may be serous
well circumscribed and are composed of benign epithelium with or bloody. Grossly, intraductal papillomas are pinkish tan, fri-
sparse stroma, which is the histologic feature that differentiates able, and usually attached to the wall of the involved duct by a
them from fibroadenomas. They may be divided into tubular stalk. They rarely undergo malignant transformation, and their
adenomas and lactating adenomas. Tubular adenomas are seen presence does not increase a woman’s risk of developing breast
in young nonpregnant women, whereas lactating adenomas are cancer (unless accompanied by atypia). However, multiple
seen during pregnancy or during the postpartum period. Ham- intraductal papillomas, which occur in younger women and are
artomas are discrete breast tumors that are usually 2 to 4 cm in less frequently associated with nipple discharge, are susceptible
diameter, firm, and sharply circumscribed. Adenolipomas con- to malignant transformation.
sist of sharply circumscribed nodules of fatty tissue that contain
normal breast lobules and ducts. Pathology of Atypical Proliferative Diseases
Fibrocystic Disease. The term fibrocystic disease is nonspe- The atypical proliferative diseases have some of the features of
cific. Too frequently, it is used as a diagnostic term to describe carcinoma in situ but either lack a major defining feature of car-
symptoms, to rationalize the need for breast biopsy, and to cinoma in situ or have the features in less than fully developed
explain biopsy results. Synonyms include fibrocystic changes, form.34 Atypical ductal hyperplasia (ADH) appears similar to
cystic mastopathy, chronic cystic disease, chronic cystic mas- low grade ductal carcinoma in situ (DCIS) histologically and is
titis, Schimmelbusch’s disease, mazoplasia, Cooper’s disease, composed of monotonous round, cuboidal, or polygonal cells
Reclus’ disease, and fibroadenomatosis. Fibrocystic disease enclosed by basement membrane with rare mitoses. A lesion
refers to a spectrum of histopathologic changes that are best will be considered to be ADH if it is up to 2 or 3 mm in size but
diagnosed and treated specifically. would be called DCIS if it is larger than 3 mm. The diagnosis
can be difficult to establish with core-needle biopsy specimen
Pathology of Proliferative Disorders alone and many cases will require excisional biopsy specimen
Without Atypia for classification. Individuals with a diagnosis of ADH are at
Proliferative breast disorders without atypia include sclerosing increased risk for development of breast cancer and should be
adenosis, radial scars, complex sclerosing lesions, ductal epithe- counseled appropriately regarding risk reduction strategies.
lial hyperplasia, and intraductal papillomas.32,33 Sclerosing ade- In 1978, Haagensen et al described lobular neoplasia, a
nosis is prevalent during the childbearing and perimenopausal spectrum of disorders ranging from atypical lobular hyperplasia
years and has no malignant potential. Histologic changes are to lobular carcinoma in situ (LCIS).35 Atypical lobular hyper-
both proliferative (ductal proliferation) and involutional (stro- plasia (ALH) results in minimal distention of lobular units with
mal fibrosis, epithelial regression). Sclerosing adenosis is char- cells that are similar to those seen in LCIS. The diagnosis of
acterized by distorted breast lobules and usually occurs in the LCIS is made when small monomorphic cells that distend the
context of multiple microcysts, but occasionally presents as a terminal ductal lobular unit are noted. In cases of LCIS, the
palpable mass. Benign calcifications are often associated with acini are full and distended while the overall lobular architec-
this disorder. Sclerosing adenosis can be managed by observa- ture is maintained (Fig. 17-12). Classic LCIS is not associated
tion as long as the imaging features and pathologic findings are with a specific mammographic or palpable abnormality but is
554 guidance. If a mass was noted on initial ultrasound or there is a
residual mass post aspiration, then a tissue specimen is obtained,
usually by core biopsy. When cystic fluid is bloodstained, fluid
can be sent for cytologic examination. A simple cyst is rarely of
concern, but a complex cyst may be the result of an underlying
malignancy. A pneumocystogram can be obtained by injecting
air into the cyst and then obtaining a repeat mammogram. When
this technique is used, the wall of the cyst cavity can be more
carefully assessed for any irregularities.
Fibroadenomas. Most fibroadenomas are self-limiting and
many go undiagnosed, so a more conservative approach is
PART II

reasonable. Careful ultrasound examination with core-needle


biopsy will provide for an accurate diagnosis. Ultrasonogra-
phy may reveal specific features that are pathognomonic for
fibroadenoma, and in a young woman (e.g., under 25 years)
where the risk of breast cancer is already very low a core-
SPECIFIC CONSIDERATIONS

Figure 17-12. Lobular carcinoma in situ (100x). There are small needle biopsy may not be necessary. In patients where biopsy
monomorphic cells that distend the terminal duct lobular unit, with- is performed, the patient is counseled concerning the ultra-
out necrosis or mitoses. (Used with permission from Dr. Sindhu sound and biopsy results, and surgical excision of the fibroad-
Menon, Consultant Histopathologist and Dr. Rahul Deb, Consul- enoma may be avoided. Cryoablation and ultrasound-guided
tant Histopathologist and Lead Breast Pathologist, Royal Derby vacuum-assisted biopsy are approved treatments for fibroad-
Hospital, Derby, UK.) enomas of the breast, especially lesions <3 cm. Larger lesions
are often still best treated by excision. With short-term follow-
up, a significant percentage of fibroadenomas will decrease
in size and will no longer be palpable.38 However, many will
an incidental finding noted on breast biopsy. There is a variant
remain palpable, especially those larger than 2 cm. 39 There-
of LCIS that has been termed pleomorphic LCIS. In the case of
fore, women should be counseled that the options for treat-
pleomorphic LCIS, there can be calcifications or other suspi-
ment include surgical removal, cryoablation, vacuum assisted
cious mammographic changes that dictate the need for biopsy.
biopsy, or observation.
Classic LCIS is not treated with excision as the patient is at
risk for developing invasive breast cancer in either breast and Sclerosing Disorders. The clinical significance of scleros-
therefore the patient is counseled regarding appropriate risk ing adenosis lies in its imitation of cancer. On physical exami-
reduction strategies. Pleomorphic LCIS can be difficult to dis- nation, it may be confused with cancer, by mammography,
tinguish from high-grade DCIS and there are some proponents and at gross pathologic examination. Excisional biopsy and
who have suggested that patients with pleomorphic LCIS be histologic examination are frequently necessary to exclude the
managed similar to those with DCIS with attention to margins diagnosis of cancer. The diagnostic work-up for radial scars
and consideration for radiation therapy in the setting of breast and complex sclerosing lesions frequently involves stereo-
conserving treatment. The use of immunohistochemical stain- tactic biopsy. It usually is not possible to differentiate these
ing for E-cadherin can help to discriminate between LCIS and lesions with certainty from cancer by mammographic features,
DCIS. In lobular neoplasias, such as ALH and LCIS, there is a so a larger tissue biopsy is recommended either by way of
lack of E-cadherin expression, whereas the majority of ductal vacuum-assisted biopsy or an open surgical excisional biopsy.
lesions will demonstrate E-cadherin reactivity. The mammographic appearance of a radial scar or sclerosing
adenosis (mass density with spiculated margins) will usually
Treatment of Selected Benign Breast Disorders lead to an assessment that the results of a core-needle biopsy
and Diseases specimen showing benign disease are discordant with the
Cysts. Because needle biopsy of breast masses may produce radiographic findings.
artifacts that make mammography assessment more difficult, Periductal Mastitis. Painful and tender masses behind the
many multidisciplinary teams prefer to image breast masses nipple-areola complex are aspirated with a 21-gauge needle
before performing either fine-needle aspiration or core-needle attached to a 10-mL syringe. Any fluid obtained is submitted
biopsy.36,37 In practice, however, the first investigation of pal- for culture using a transport medium appropriate for the detec-
pable breast masses may be a needle biopsy, which allows for tion of anaerobic organisms. In the absence of pus, women are
the early diagnosis of cysts. A 21-gauge needle attached to a started on a combination of antibiotics to cover polymicrobial
10-mL syringe is placed directly into the mass, which is fixed infections while awaiting the results of culture. Antibiotics are
by fingers of the nondominant hand. The volume of a typical then continued based on sensitivity tests. Many cases respond
cyst is 5 to 10 mL, but it may be 75 mL or more. If the fluid satisfactorily to antibiotics alone, but when considerable puru-
that is aspirated is not bloodstained, then the cyst is aspirated lent material is present, repeated ultrasound guided aspiration
to dryness, the needle is removed, and the fluid is discarded is performed, and ultimately in a proportion of cases surgical
because cytologic examination of such fluid is not cost effec- treatment is required. Unlike puerperal abscesses, a subareo-
tive. After aspiration, the breast is carefully palpated to exclude lar abscess is usually unilocular and often is associated with
a residual mass. In most cases, however, imaging has been per- a single duct system. Ultrasound will accurately delineate its
formed prior to a needle being introduced into the breast, and extent. In those cases that come to surgery, the surgeon may
indeed the majority of cysts are now aspirated under ultrasound either undertake simple drainage with a view toward formal
Table 17-5 RISK FACTORS FOR BREAST CANCER 555

Treatment of recurrent subareolar sepsis Hormonal and Nonhormonal Risk Factors


Increased exposure to estrogen is associated with an increased
SUITABLE FOR SUITABLE FOR TOTAL risk for developing breast cancer, whereas reducing exposure
FISTULECTOMY DUCT EXCISION is thought to be protective.42-48 Correspondingly, factors that
increase the number of menstrual cycles, such as early men-
Small abscess localized to Large abscess affecting >50%
arche, nulliparity, and late menopause, are associated with
one segment of the areolar circumference
increased risk. Moderate levels of exercise and a longer lacta-
Recurrence involving the Recurrence involving a tion period, factors that decrease the total number of menstrual
same segment different segment cycles, are protective. The terminal differentiation of breast epi-

CHAPTER 17 THE BREAST


Mild or no nipple inversion Marked nipple inversion thelium associated with a full-term pregnancy is also protective,
Patient unconcerned about Patient requests correction of so older age at first live birth is associated with an increased risk
nipple inversion nipple inversion of breast cancer. Finally, there is an association between obesity
and increased breast cancer risk. Because the major source of
Younger patient Older patient
estrogen in postmenopausal women is the conversion of andro-
No discharge from other Purulent discharge from other stenedione to estrone by adipose tissue, obesity is associated
ducts ducts with a long-term increase in estrogen exposure.
No prior fistulectomy Recurrence after fistulectomy Nonhormonal risk factors include radiation exposure.
Modified with permission from Mansel RE, Webster DJT: Benign
Young women who receive mantle radiation therapy for Hodg-
Disorders and Diseases of the Breast: Concepts and Clinical kin’s lymphoma have a breast cancer risk that is 75 times greater
Management, 2nd ed. London: Elsevier/Saunders; 2000. than that of age-matched control subjects. Survivors of the
atomic bomb blasts in Japan during World War II have a very
high incidence of breast cancer, likely because of somatic muta-
tions induced by the radiation exposure. In both circumstances,
radiation exposure during adolescence, a period of active breast
surgery, should the problem recur, or proceed with definitive development, magnifies the deleterious effect. Studies also sug-
surgery. In a woman of childbearing age, simple drainage gest that the risk of breast cancer increases as the amount of
is preferred, but if there is an anaerobic infection, recurrent alcohol a woman consumes increases.49 Alcohol consumption
infection frequently develops. Recurrent abscess with fistula is known to increase serum levels of estradiol. Finally, evidence
is a difficult problem. Treatment of periductal fistula was ini- suggests that long-term consumption of foods with a high fat
tially recommended to be opening up of the fistulous track content contributes to an increased risk of breast cancer by
and allowing it to granulate.40 This approach may still be used, increasing serum estrogen levels.
especially if the fistula is recurrent after previous attempts at
fistulectomy. However, nowadays the preferred initial surgical Risk Assessment Models
treatment is by fistulectomy and primary closure with anti- The average lifetime risk of breast cancer for newborn U.S.
biotic coverage.41 Excision of all the major ducts is an alter- women is 12%.50,51 The longer a woman lives without cancer, the
native option depending on the circumstances (Table 17-5). lower her risk of developing breast cancer. Thus, a woman age
When a localized periareolar abscess recurs at the previous 50 years has an 11% lifetime risk of developing breast cancer,
site and a fistula is present, the preferred operation is fistulec- and a woman age 70 years has a 7% lifetime risk of developing
tomy, which has minimal complications and a high degree of breast cancer. Because risk factors for breast cancer interact,
success. However, when subareolar sepsis is diffused rather evaluating the risk conferred by combinations of risk factors is
than localized to one segment or when more than one fistula is difficult. There are several risk assessment models available to
present, total duct excision is the most expeditious approach. predict the risk of breast cancer. From the Breast Cancer Detec-
The first circumstance is seen in young women with squamous tion Demonstration Project, a mammography screening program
metaplasia of a single duct, whereas the latter circumstance is conducted in the 1970s, Gail et al developed the model most
seen in older women with multiple ectatic ducts. Age is not frequently used in the United States, which incorporates age, age
always a reliable guide, however, and fistula excision is the at menarche, age at first live birth, the number of breast biopsy
preferred initial procedure for localized sepsis irrespective of specimens, any history of atypical hyperplasia, and number of
age. Antibiotic therapy is useful for recurrent infection after first-degree relatives with breast cancer.52 It predicts the cumula-
fistula excision, and a 2- to 4-week course is recommended tive risk of breast cancer according to decade of life. To calculate
before total duct excision. breast cancer risk using the Gail model, a woman’s risk factors
Nipple Inversion. More women request correction of con- are translated into an overall risk score by multiplying her rela-
genital nipple inversion than request correction for the nipple tive risks from several categories (Table 17-6). This risk
inversion that occurs secondary to duct ectasia. Although the 5 score is then compared to an adjusted population risk of
results are usually satisfactory, women seeking correction for breast cancer to determine a woman’s individual or absolute risk.
cosmetic reasons should always be made aware of the surgi- The output is a 5-year risk and a lifetime risk of developing
cal complications of altered nipple sensation, nipple necrosis, breast cancer. A software program incorporating the Gail model
and postoperative fibrosis with nipple retraction. Because nipple is available from the National Cancer Institute at http://bcra.nci.
inversion is a result of shortening of the subareolar ducts, a com- nih.gov/brc. This model was recently modified to more accu-
plete division of these ducts is necessary for permanent correc- rately assess risk in African American women.52,53 There have
tion of the disorder. also been modifications that project individualized absolute
556 Table 17-6
breast cancer and their age at diagnosis. Risk factors that are
less consistently associated with breast cancer (diet, use of oral
Relative risk estimates for the Gail model contraceptives, lactation) or are rare in the general population
(radiation exposure) are not included in either the Gail or Claus
VARIABLE RELATIVE RISK risk assessment model. Other models have been proposed that
Age at menarche (years) account for mammographic breast density in assessing breast
≥14 1.00 cancer risk.54,56
12–13 1.10 Neither the Gail model nor the Claus model accounts for
<12 1.21 the risk associated with mutations in the breast cancer suscepti-
Number of biopsy specimens/history of bility genes BRCA1 and BRCA2 (described in detail in the fol-
benign breast disease, age <50 y lowing section). The BRCAPRO model is a Mendelian model
that calculates the probability that an individual is a carrier of a
PART II

0 1.00
1 1.70 mutation in one of the breast cancer susceptibility genes based
≥2 2.88 on their family history of breast and ovarian cancer.57 The prob-
Number of biopsy specimens/history of ability that an individual will develop breast or ovarian cancer
benign breast disease, age ≥50 y is derived from this mutation probability based on age-specific
0 1.02 incidence curves for both mutation carriers and noncarriers.58
SPECIFIC CONSIDERATIONS

1 1.27 Use of the BRCAPRO model in the clinic is challenging since it


≥2 1.62 requires input of all family history information regarding breast
Age at first live birth (years) and ovarian cancer. The Tyrer-Cuzick model attempts to utilize
<20 y both family history information and individual risk information.
Number of first-degree relatives with It uses the family history to calculate the probability that an
history of breast cancer individual carries a mutation in one of the breast cancer suscep-
0 1.00 tibility genes, and then the risk is adjusted based on personal
1 2.61 risk factors, including age at menarche, parity, age at first live
≥2 6.80 birth, age at menopause, history of atypical hyperplasia or LCIS,
20–24 y height, and body mass index.59 Once a risk model has been uti-
Number of first-degree relatives with lized to assess breast cancer risk, this must be communicated
history of breast cancer to the individual and put into context with competing risk and
0 1.24 medical comorbidities. This information can then be used to
1 2.68 discuss options that are available to the individual for manag-
≥2 5.78 ing risk.
25–29 y
Number of first-degree relatives with Risk Management
history of breast cancer Several important medical decisions may be affected by a wom-
0 1.55 an’s underlying risk of developing breast cancer.60-68 These deci-
1 2.76 sions include when to use postmenopausal hormone replacement
≥2 4.91 therapy, at what age to begin mammography screening or incor-
≥30 y porate magnetic resonance imaging (MRI) screening, when to
Number of first-degree relatives with use tamoxifen to prevent breast cancer, and when to perform
history of breast cancer prophylactic mastectomy to prevent breast cancer. Postmeno-
0 1.93 pausal hormone replacement therapy was widely prescribed in
1 2.83 the 1980s and 1990s because of its effectiveness in controlling
≥2 4.17 the symptoms of estrogen deficiency, namely vasomotor symp-
Reproduced with permission from Armstrong K, Eisen A, Weber B: toms such as hot flashes, night sweats and their associated sleep
Assessing the risk of breast cancer, N Engl J Med. 2000 deprivation, osteoporosis, and cognitive changes. Furthermore,
Feb 24;342(8):564-571.
these hormone supplements were thought to reduce coronary
artery disease as well. Use of combined estrogen and progester-
invasive breast cancer risk for Asian and Pacific Island American one became standard for women who had not undergone hyster-
women. The Gail model is the most widely used model in the ectomy because unopposed estrogen increases the risk of uterine
United States. Gail and colleagues have also described a revised cancer. Concerns of prolonging a woman’s lifetime exposure to
model that includes body weight and mammographic density but estrogen, coupled with conflicting data regarding the impact of
excludes age at menarche.54 these hormones on cardiovascular health, motivated the imple-
Claus et al, using data from the Cancer and Steroid Hor- mentation of large-scale phase 3 clinical trials to definitively
mone Study, a case-control study of breast cancer, developed evaluate the risks vs. benefits of postmenopausal hormone
the other frequently used risk assessment model, which is based replacement therapy. The Women’s Health Initiative (WHI)
on assumptions about the prevalence of high-penetrance breast was therefore designed by the National Institutes of Health as
cancer susceptibility genes.55 Compared with the Gail model, the a series of clinical trials to study the effects of diet, nutritional
Claus model incorporates more information about family his- supplements, and hormones on the risk of cancer, cardiovascular
tory but excludes other risk factors. The Claus model provides disease, and bone health in postmenopausal women. Findings
individual estimates of breast cancer risk according to decade of from primary studies of postmenopausal hormone replacement
life based on presence of first- and second-degree relatives with therapy were released in 2002, demonstrating conclusively that
breast cancer risk is threefold to fourfold higher after >4 years should continue. The use of screening mammography in women 557
of use and there is no significant reduction in coronary artery or <50 years of age is more controversial for several reasons: (a)
cerebrovascular risks. The Collaborative Group on Hormonal breast density is greater, and screening mammography is less
Factors in Breast Cancer combined and reanalyzed data from a likely to detect early breast cancer (i.e., reduced sensitivity);
number of studies totaling 52,705 women with breast cancer and (b) screening mammography results in more false-positive test
108,411 women without breast cancer. They found an increased findings (i.e., reduced specificity), which results in unneces-
risk of breast cancer with every use of estrogen replacement sary biopsy specimens; and (c) younger women are less likely
therapy. They also reported increased risk among current users to have breast cancer (i.e., lower incidence), so fewer young
but not past users and risk increased with increasing duration women will benefit from screening.80,81 In the United States,
of use of hormone replacement therapy.69 Cheblowski et al on a population basis, however, the benefits of screening mam-

CHAPTER 17 THE BREAST


also reported from the WHI study that estrogen + progesterone mography in women between the ages of 40 and 49 years is still
increased the incidence of breast cancer.70 This was con- felt to outweigh the risks; although targeting mammography to
firmed by the Million Women study, which also showed that women at higher risk of breast cancer improves the balance of
the increased risk was substantially greater for the combined risks and benefits and is the approach some health care sys-
estrogen + progesterone replacement therapy than other types tems have taken. In one study of women age 40 to 49 years, an
of hormone replacement therapy.71 abnormal mammography finding was three times more likely
Breast Cancer Screening. Routine use of screening mam- to be cancer in a woman with a family history of breast cancer
mography in women ≥50 years of age has been reported to than in a woman without such a history. Furthermore, as noted
reduce mortality from breast cancer by 25%.72 This reduc- previously in the section Risk Assessment Models, mounting
6 tion comes at an acceptable economic cost. More recently, data regarding mammographic breast density demonstrate an
there has been debate over the potential harms associated with independent correlation with breast cancer risk. Incorporation
breast screening.73 Controversy over the age to initiate screening of breast density measurements into breast cancer risk assess-
mammography is evident in the current recommendations. The ment models appears to be a promising strategy for increasing
U.S. Preventive Services Task Force (USPSTF), the American the accuracy of these tools. Unfortunately, widespread applica-
Cancer Society (ACS), and the National Comprehensive Cancer tion of these modified models is hampered by inconsistencies
Network (NCCN) are three organizations with differing recom- in the reporting of mammographic density. Ultrasonography can
mendations for screening mammography in average risk also be used for breast cancer screening in women with dense
women. The guidelines, however, similarly define high-risk breasts, but there is no data available that the additional cancers
women as those with personal history of breast cancer, history detected with this modality reduce mortality from breast cancer.
of chest radiation at young age, and confirmed or suspected Current recommendations by the United States Preventive
genetic mutation known to increase risk for developing breast Services Task Force are that women undergo biennial mammo-
cancer. The USPSTF recommends biennial screening mammog- graphic screening between the ages of 50 and 74 years.77 The
raphy for women age 50 to 74 years. The USPSTF applies these use of MRI for breast cancer screening is recommended by the
guidelines to asymptomatic women age >40 years who do not ACS for women with a 20% to 25% or greater lifetime risk using
have a preexisting breast cancer or who were not previously risk assessment tools based mainly on family history, BRCA
diagnosed with a high-risk breast lesion, and who are not at high mutation carriers, those individuals who have a family member
risk for breast cancer because of a known underlying genetic with a BRCA mutation who have not been tested themselves,
mutation or history of chest radiation at a young age.74-76 In individuals who received radiation to the chest between the
October 2015, the ACS released updated guidelines stating ages of 10 and 30 years, and those individuals with a history of
average-risk women should start annual screening mammogra- Li-Fraumeni syndrome, Cowden syndrome, or Bannayan-Riley-
phy at 45 years of age. Women age 45 to 54 years should be Ruvalcaba syndrome or those who have a first-degree relative
screened annually, and those 55 years and older should transi- with one of these syndromes. MRI is an extremely sensitive
tion to biennial screening or have the opportunity to continue screening tool that is not limited by the density of the breast
annual screening. Women should have the opportunity to begin tissue as mammography is; however, its specificity is moderate,
annual screening between the ages of 40 and 44 years and leading to more false-positive events and the increased need for
should continue screening as long as their overall health is good biopsy.
and have a life expectancy of 10 years or longer. The ACS does Chemoprevention. Tamoxifen, a selective estrogen receptor
not recommend clinical breast examination for breast cancer modulator, was the first drug shown to reduce the incidence
screening among average-risk women at any age.77 The NCCN of breast cancer in healthy women. There have been four pro-
recommends that average-risk women begin annual screening spective studies published evaluating tamoxifen vs. placebo for
mammograms at ≥40 years of age, along with annual clinical reducing the incidence of invasive breast cancer for women at
breast exams and breast awareness.78 increased risk. The largest trial was the Breast Cancer Preven-
The United Kingdom recently established an independent tion Trial (NSABP P-01), which randomly assigned >13,000
expert panel to review the published literature and estimate women with a 5-year Gail relative risk of breast cancer of 1.66%
the benefits and harms associated with screening women or higher or LCIS to receive tamoxifen or placebo. After a mean
>50 years of age in its national screening program.79 The expert follow-up period of 4 years, the incidence of breast cancer
panel estimated that an invitation to breast screening delivers was reduced by 49% in the group receiving tamoxifen.60 The
about a 20% reduction in breast cancer mortality. At the same decrease was evident only in ER-positive breast cancers with no
time, however, the panel estimated that in women invited to the significant change in ER-negative tumors. The Royal Marsden
screening, about 11% of the cancers diagnosed in their lifetime Hospital Tamoxifen Chemoprevention Trial,78 the Italian Tamox-
constitute overdiagnosis. Despite the overdiagnosis, the panel ifen Prevention Trial,82 and the International Breast Cancer
concluded that breast screening confers significant benefit and Intervention Study I (IBIS-I) trial all83 showed a reduction in
558 ER-positive breast cancers with the use of tamoxifen compared chemoprevention in premenopausal or postmenopausal women
with placebo. There was no effect on mortality; however, the and consideration for raloxifene or exemestane in postmeno-
trials were not powered to assess either breast cancer mortality pausal women who are noted to be at increased risk of breast
or all-cause mortality events. The adverse events were similar in cancer.91,92 The discussion with an individual patient should
all four randomized trials, including an increased risk of endo- include risk assessment and potential risks and benefits with
metrial cancer, thromboembolic events, cataract formation, and each agent.
vasomotor disturbances in individuals receiving tamoxifen.
Risk-Reducing Surgery. A retrospective study of women at
Tamoxifen therapy currently is recommended only for
high risk for breast cancer found that prophylactic mastectomy
women who have a Gail relative risk of 1.66% or higher, who
reduced their risk by >90%.62 However, the effects of prophylac-
are age 35 to 59, women over the age of 60, or women with
tic mastectomy on the long-term quality of life are poorly quan-
a diagnosis of LCIS or atypical ductal or lobular hyperplasia.
tified. A study involving women who were carriers of a breast
In addition, deep vein thrombosis occurs 1.6 times as often,
PART II

cancer susceptibility gene (BRCA) mutation found that the ben-


pulmonary emboli 3.0 times as often, and endometrial cancer
efit of prophylactic mastectomy differed substantially according
2.5 times as often in women taking tamoxifen. The increased
to the breast cancer risk conferred by the mutations. For women
risk for endometrial cancer is restricted to early stage cancers
with an estimated lifetime risk of 40%, prophylactic mastec-
in postmenopausal women. Cataract surgery is required almost
tomy added almost 3 years of life, whereas for women with an
twice as often among women taking tamoxifen. Gail et al sub-
SPECIFIC CONSIDERATIONS

estimated lifetime risk of 85%, prophylactic mastectomy added


sequently developed a model that accounts for underlying risk
>5 years of life.66 Domchek et al evaluated a cohort of BRCA1
of breast cancer as well as comorbidities to determine the net
and 2 mutation carriers who were followed prospectively
risk-benefit ratio of tamoxifen use for chemoprevention.84
and reported on outcomes with risk-reducing surgery.93 They
The NSABP completed a second chemoprevention trial,
found that risk-reducing mastectomy was highly effective at
designed to compare tamoxifen and raloxifene for breast cancer
preventing breast cancer in both BRCA1 and 2 mutation carriers.
risk reduction in high-risk postmenopausal women. Raloxifene,
Risk-reducing salpingo-oophorectomy was highly effective at
another selective estrogen receptor modulator, was selected for
reducing the incidence of ovarian cancer and breast cancer in
the experimental arm in this follow-up prevention trial because
BRCA mutation carriers and was associated with a reduction in
its use in managing postmenopausal osteoporosis suggested
breast cancer-specific mortality, ovarian cancer-specific mor-
that it might be even more effective at breast cancer risk reduc-
tality, and all-cause mortality. While studies of bilateral pro-
tion, but without the adverse effects of tamoxifen on the uterus.
phylactic or risk-reducing mastectomy have reported dramatic
The P-2 trial, the Study of Tamoxifen and Raloxifene (known
reductions in breast cancer incidence among those without
as the STAR trial), randomly assigned 19,747 postmenopausal
known BRCA mutations, there is little data to support a survival
women at high-risk for breast cancer to receive either tamoxi-
benefit. Another consideration is that while most patients are
fen or raloxifene. The initial report of the P-2 trial showed the
satisfied with their decision to pursue risk-reducing surgery,
two agents were nearly identical in their ability to reduce breast
some are dissatisfied with the cosmetic outcomes mostly due to
cancer risk, but raloxifene was associated with a more favor-
reconstructive issues.
able adverse event profile.85 An updated analysis revealed that
raloxifene maintained 76% of the efficacy of tamoxifen in pre-
vention of invasive breast cancer with a more favorable side BRCA Mutations
effect profile. The risk of developing endometrial cancer was BRCA1. Up to 5% of breast cancers are caused by inheritance
significantly higher with tamoxifen use at longer follow-up.86 of germline mutations such as BRCA1 and BRCA2, which
Although tamoxifen has been shown to reduce the incidence are inherited in an autosomal dominant fashion with varying
of LCIS and DCIS, raloxifene did not have an effect on the degrees of penetrance (Table 17-7).94-100 BRCA1 is located on
frequency of these diagnoses. chromosome arm 17q, spans a genomic region of approximately
Aromatase inhibitors (AIs) have been shown to be more 100 kilobases (kb) of DNA, and contains 22 coding exons for
effective than tamoxifen in reducing the incidence of contra- 1863 amino acids. Both BRCA1 and BRCA2 function as tumor-
lateral breast cancers in postmenopausal women receiving AIs suppressor genes, and for each gene, loss of both alleles is
for adjuvant treatment of invasive breast cancer. The MAP.3 required for the initiation of cancer. Data accumulated since
trial was the first study to evaluate an AI as a chemopreventive the isolation of the BRCA1 gene suggest a role in transcription,
agent in postmenopausal women at high risk for breast cancer. cell-cycle control, and DNA damage repair pathways. More
The trial randomized 4560 women to exemestane 25 mg daily than 500 sequence variations in BRCA1 have been identified.
vs. placebo for 5 years. After a median follow-up of 35 months, It now is known that germline mutations in BRCA1 represent
exemestane was shown to reduce invasive breast cancer inci- a predisposing genetic factor in as many as 45% of hereditary
dence by 65%. Side effect profiles demonstrated more grade II breast cancers and in at least 80% of hereditary ovarian cancers.
or higher arthritis and hot flashes in patients taking exemestane.87 Female mutation carriers have been reported to have up to an
The IBIS II trial on the other hand, randomized 3864 postmeno- 85% lifetime risk (for some families) for developing breast
pausal women to either anastrozole, a nonsteroidal aromatase cancer and up to a 40% lifetime risk for developing ovarian
inhibitor, vs. placebo with a further randomization to bisphospho- cancer. The initial families reported had high penetrance and
nate or not based on bone density.88,89 After a median follow-up subsequently the average lifetime risk has been reported to lie
of 5 years, anastrozole reduced the incidence of invasive breast between 60% and 70%. Breast cancer susceptibility in these
cancer by about 50%. The trial also had an initial sub-study families appears as an autosomal dominant trait with high pen-
that looked at the effect of the aromatase inhibitor on cogni- etrance. Approximately 50% of children of carriers inherit the
tive function and reported no adverse effects.90 The American trait. In general, BRCA1-associated breast cancers are invasive
Society of Clinical Oncology recommends tamoxifen for ductal carcinomas, are poorly differentiated, are in the majority
Table 17-7
estimated to be close to 20%. Breast cancer susceptibility in 559
BRCA2 families is an autosomal dominant trait and has a high
Incidence of sporadic, familial, and hereditary breast penetrance. Approximately 50% of children of carriers inherit
cancer the trait. Unlike male carriers of BRCA1 mutations, men with
Sporadic breast cancer 65%–75% germline mutations in BRCA2 have an estimated breast cancer
risk of 6%, which represents a 100-fold increase over the risk in
Familial breast cancer 20%–30%
the general male population. BRCA2-associated breast cancers
Hereditary breast cancer 5%–10% are invasive ductal carcinomas, which are more likely to be
BRCA1 a
45% well differentiated and to express hormone receptors than are
BRCA2 35% BRCA1-associated breast cancers. BRCA2-associated breast

CHAPTER 17 THE BREAST


cancer has a number of distinguishing clinical features, such
p53a (Li-Fraumeni syndrome) 1%
as an early age of onset compared with sporadic cases, a higher
STK11/LKB1 (Peutz-Jeghers syndrome)
a
<1% prevalence of bilateral breast cancer, and the presence of associ-
PTEN (Cowden disease)
a
<1% ated cancers in some affected individuals, specifically ovarian,
MSH2/MLH1a (Muir-Torre syndrome) <1% colon, prostate, pancreatic, gallbladder, bile duct, and stomach
cancers, as well as melanoma. A number of founder mutations
ATM (Ataxia-telangiectasia)
a
<1%
have been identified in BRCA2. The 6174delT mutation is found
Unknown 20% in Ashkenazi Jews with a prevalence of 1.2% and accounts for
a
Affected gene. 60% of ovarian cancer and 30% of early-onset breast cancer
Data from Martin AM, Weber BL: Genetic and hormonal risk factors in patients among Ashkenazi women.106 Another BRCA2 founder
breast cancer, J Natl Cancer Inst. 2000 Jul 19;92(14):1126-1135. mutation, 999del5, is observed in Icelandic and Finnish popula-
tions, while more recently 3036delACAA has been observed in
a number of Spanish families.107-109
hormone receptor negative, and have a triple receptor negative
(immunohistochemical profile: ER-negative, PR-negative, and Identification of BRCA Mutation Carriers. Identifying
HER2-negative) or basal phenotype (based on gene expression hereditary risk for breast cancer is a four-step process that
profiling). BRCA1-associated breast cancers have a number of includes: (a) obtaining a complete, multigenerational family
distinguishing clinical features, such as an early age of onset history, (b) assessing the appropriateness of genetic testing for
compared with sporadic cases; a higher prevalence of bilateral a particular patient, (c) counseling the patient, and (d) interpret-
breast cancer; and the presence of associated cancers in some ing the results of testing.110 Genetic testing should not be offered
affected individuals, specifically ovarian cancer and possibly in isolation, but only in conjunction with patient education and
colon and prostate cancers. counseling, including referral to a genetic counselor. Initial
Several founder mutations have been identified in BRCA1. determinations include whether the individual is an appropriate
The two most common mutations are 185delAG and 5382insC, candidate for genetic testing and whether genetic testing will be
which account for 10% of all the mutations seen in BRCA1. informative for personal and clinical decision-making. A thor-
These two mutations occur at a 10-fold higher frequency ough and accurate family history is essential to this process, and
in the Ashkenazi Jewish population than in non-Jewish the maternal and paternal sides of the family are both assessed
Caucasians. The carrier frequency of the 185delAG mutation because 50% of the women with a BRCA mutation have inher-
in the Ashkenazi Jewish population is 1% and, along with the ited the mutation from their fathers. To help clinicians advise
5382insC mutation, accounts for almost all BRCA1 mutations in women about genetic testing, statistically based models that
this population. Analysis of germline mutations in Jewish and determine the probability that an individual carries a BRCA
non-Jewish women with early-onset breast cancer indicates that mutation have been developed. A method for calculating carrier
20% of Jewish women who develop breast cancer before age probability that has been demonstrated to have acceptable per-
40 years carry the 185delAG mutation. There are founder formance (i.e., both in terms of calibration and discrimination)
BRCA1 mutations in other populations including, among others, such as the Manchester scoring system and BODICEA should
Dutch, Polish, Finnish, and Russian populations.101-105 be used to offer referral to a specialist genetic clinic. A heredi-
tary risk of breast cancer is considered if a family includes Ash-
BRCA2. BRCA2 is located on chromosome arm 13q and spans
kenazi Jewish heritage; a first-degree relative with breast cancer
a genomic region of approximately 70 kb of DNA. The 11.2-kb
before age 50; a history of ovarian cancer at any age in the
coding region contains 26 coding exons.94-100 It encodes a pro-
patient or first- or second-degree relative with ovarian cancer;
tein of 3418 amino acids. The BRCA2 gene bears no homology
breast and ovarian cancer in the same individual; two or more
to any previously described gene, and the protein contains no
first- or second-degree relatives with breast cancer at any age;
previously defined functional domains. The biologic function
patient or relative with bilateral breast cancer; and male breast
of BRCA2 is not well defined, but like BRCA1, it is postulated
cancer in a relative at any age.111 The threshold for genetic test-
to play a role in DNA damage response pathways. BRCA2 mes-
ing is lower in individuals who are members of ethnic groups in
senger RNA also is expressed at high levels in the late G1 and
whom the mutation prevalence is increased.
S phases of the cell cycle. The kinetics of BRCA2 protein regu-
lation in the cell cycle is similar to that of BRCA1 protein, which BRCA Mutation Testing. Appropriate counseling for the
suggests that these genes are coregulated. The mutational spec- individual being tested for a BRCA mutation is strongly rec-
trum of BRCA2 is not as well established as that of BRCA1. ommended, and documentation of informed consent is
To date, >250 mutations have been found. The breast cancer required.110,112 The test that is clinically available for analyzing
risk for BRCA2 mutation carriers is close to 85%, and the life- BRCA mutations is gene sequence analysis. In a family with a
time ovarian cancer risk, while lower than for BRCA1, is still history suggestive of hereditary breast cancer and no previously
560 tested member, the most informative strategy is first to test Currently, there is little documented evidence of genetic dis-
an affected family member. This person undergoes complete crimination resulting from findings of available genetic tests.
sequence analysis of both the BRCA1 and BRCA2 genes. If a
Cancer Prevention for BRCA Mutation Carriers. Risk man-
mutation is identified, relatives are usually tested only for that
agement strategies for BRCA1 and BRCA2 mutation carriers
specific mutation. An individual of Ashkenazi Jewish ancestry
include the following:
is tested initially for the three specific mutations that account
for hereditary breast and ovarian cancer in that population. If 1. Risk-reducing mastectomy and reconstruction
results of that test are negative, it may then be appropriate to 2. Risk-reducing salpingo-oophorectomy
fully analyze the BRCA1 and BRCA2 genes. 3. Intensive surveillance for breast and ovarian cancer
A positive test result is one that discloses the presence of a 4. Chemoprevention
BRCA mutation that interferes with translation or function of the
BRCA protein. A woman who carries a deleterious mutation has Although removal of breast tissue reduces the likeli-
PART II

a breast cancer risk of up to 85% (in some families) as well as hood that BRCA1 and BRCA2 mutation carriers will develop
a greatly increased risk of ovarian cancer. A negative test result breast cancer, mastectomy does not remove all breast tissue,
is interpreted according to the individual’s personal and family and women continue to be at risk because a germline muta-
history, especially whether a mutation has been previously iden- tion is present in any remaining breast tissue. For postmeno-
tified in the family, in which case the woman is generally tested pausal BRCA1 and BRCA2 mutation carriers who have not had
SPECIFIC CONSIDERATIONS

only for that specific mutation. If the mutation is not present, a mastectomy, it may be advisable to avoid hormone replace-
the woman’s risk of breast or ovarian cancer may be no greater ment therapy because no data exist regarding the effect of the
than that of the general population. In addition, no BRCA muta- therapy on the penetrance of breast cancer susceptibility genes.
tion can be passed on to the woman’s children. In the absence Because breast cancers in BRCA mutation carriers have the
of a previously identified mutation, a negative test result in an same mammographic appearance as breast cancers in noncarri-
affected individual generally indicates that a BRCA mutation is ers, a screening mammogram is likely to be effective in BRCA
not responsible for the familial cancer. However, the possibil- mutation carriers, provided it is performed and interpreted by
ity remains of an unusual abnormality in one of these genes an experienced radiologist with a high level of suspicion. Pres-
that cannot yet be identified through clinical testing. It also is ent screening recommendations for BRCA mutation carriers
possible that the familial cancer is indeed caused by an identifi- who do not undergo risk-reducing mastectomy include clinical
able BRCA mutation but that the individual tested had sporadic breast examination every 6 months and mammography every
cancer, a situation known as phenocopy. This is especially pos- 12 months beginning at age 25 years because the risk of breast
sible if the individual tested developed breast cancer close to the cancer in BRCA mutation carriers increases after age 30 years.
age of onset of the general population (age 60 years or older) Recent attention has been focused on the use of MRI for breast
rather than before age 50 years, as is characteristic of BRCA cancer screening in high-risk individuals and known BRCA
mutation carriers. Overall, the false-negative rate for BRCA mutation carriers. MRI appears to be more sensitive at detect-
mutation testing is <5%. Some test results, especially when a ing breast cancer in younger women with dense breasts.113 How-
single base-pair change (missense mutation) is identified, may ever, as noted previously, MRI does lead to the detection of
be difficult to interpret. This is because single base-pair changes benign breast lesions that cannot easily be distinguished from
do not always result in a nonfunctional protein. Thus, missense malignancy, and these false-positive events can result in more
mutations not located within critical functional domains, or interventions, including biopsy specimens. The current recom-
those that cause only minimal changes in protein structure, may mendations from the American Cancer Society are for annual
not be disease associated and are usually reported as indetermi- MRI in women with a 20% to 25% or greater lifetime risk of
nate results. In communicating indeterminate results to women, developing breast cancer (mainly based on family history),
care must be taken to relay the uncertain cancer risk associ- women with a known BRCA1 or BRCA2 mutation, those who
ated with this type of mutation and to emphasize that ongoing have a first-degree relative with a BRCA1 or BRCA2 mutation
research might clarify its meaning. In addition, testing other and have not had genetic testing themselves, women who were
family members with breast cancer to determine if a genetic treated with radiation therapy to the chest between the ages of
variant tracks with their breast cancer may provide clarification 10 and 30 years, and those who have Li-Fraumeni syndrome,
as to its significance. Indeterminate genetic variance currently Cowden syndrome, or Bannayan-Riley-Ruvalcaba syndrome, or
accounts for 12% of the test results. a first-degree relative with one of these syndromes.75,114 Despite
Concern has been expressed that the identification of a 49% reduction in the overall incidence of breast cancer and
hereditary risk for breast cancer may interfere with access to a 69% reduction in the incidence of estrogen receptor positive
affordable health insurance. This concern refers to discrimina- tumors in high-risk women taking tamoxifen reported in the
tion directed against an individual or family based solely on an NSABP P1 trial, there is insufficient evidence to recommend
apparent or perceived genetic variation from the normal human the use of tamoxifen uniformly for BRCA1 mutation carriers.60
genotype. The Health Insurance Portability and Accountability Cancers arising in BRCA1 mutation carriers are usually high
Act of 1996 (HIPAA) made it illegal in the United States for grade and are most often hormone receptor negative. Approxi-
group health plans to consider genetic information as a preexist- mately 66% of BRCA1-associated DCIS lesions are estrogen
ing condition or to use it to deny or limit coverage. Most states receptor negative, which suggests early acquisition of the hor-
also have passed laws that prevent genetic discrimination in the mone-independent phenotype. In the NSABP P1 trial there was
provision of health insurance. In addition, individuals applying a 62% reduction in the incidence of breast cancer in BRCA2
for health insurance are not required to report whether relatives carriers, similar to the overall reduction seen in the P1 trial. In
have undergone genetic testing for cancer risk, only whether contrast, there was no reduction seen in breast cancer incidence
those relatives have actually been diagnosed with cancer. in BRCA1 carriers who started tamoxifen in P1 age 35 years or
older.115 Tamoxifen appears to be more effective at preventing women and is responsible for 14% of the cancer-related deaths 561
estrogen receptor-positive breast cancers. in women.
The risk of ovarian cancer in BRCA1 and BRCA2 muta- Breast cancer was the leading cause of cancer-related
tion carriers ranges from 20% to 40%, which is 10 times higher mortality in women until 1987, when it was surpassed by
than that in the general population. Risk-reducing salpingo- lung cancer. In the 1970s, the probability that a woman in the
oophorectomy is a reasonable prevention option in mutation United States would develop breast cancer at some point in her
carriers. In women with a documented BRCA1 or BRCA2 lifetime was estimated at 1 in 13; in 1980 it was 1 in 11; and in
mutation, consideration for bilateral risk-reducing salpingo- 2004 it was 1 in 8. Cancer registries in Connecticut and upper
oophorectomy should be between the ages of 35 and 40 years New York State document that the age-adjusted incidence of
at the completion of childbearing. Removing the ovaries new breast cancer cases had steadily increased since the mid-

CHAPTER 17 THE BREAST


reduces the risk of ovarian cancer and breast cancer when per- 1940s. The incidence in the United States, based on data from
formed in premenopausal BRCA mutation carriers. Hormone nine SEER registries, has been decreasing by 23% per year
replacement therapy is discussed with the patient at the time since 2000. The increase had been approximately 1% per year
of oophorectomy. The Cancer Genetics Studies Consortium from 1973 to 1980, and there was an additional increase in inci-
recommends yearly transvaginal ultrasound timed to avoid dence of 4% between 1980 and 1987, which was characterized
ovulation and annual measurement of serum cancer antigen by frequent detection of small primary cancers. The increase
125 levels beginning at age 25 years as the best screening in breast cancer incidence occurred primarily in women age
modalities for ovarian carcinoma in BRCA mutation carriers ≥55 years and paralleled a marked increase in the percentage of
who have opted to defer risk-reducing surgery. older women who had mammograms taken. At the same time,
PALB2 (partner and localizer of BRCA2) has recently been incidence rates for regional metastatic disease dropped and
characterized as a potential high-risk gene for breast cancer. breast cancer mortality declined. From 1960 to 1963, 5-year
PALB2 allows nuclear localization of BRCA2 and provides a overall survival rates for breast cancer were 63% and 46% in
scaffold for the BRCA1–PALB2–BRCA2 complex. Analysis by white and African American women, respectively, whereas the
Antoniou et al has suggested that the risk of breast cancer for rates for 1981 to 1983 were 78% and 64%, respectively. For
PALB2 mutation carriers is as high as that of BRCA2 mutation 2002 to 2008 rates were 92% and 78%, respectively.
carriers.116 The absolute risk of breast cancer for PALB2 female There is a 10-fold variation in breast cancer incidence
mutation carriers by 70 years of age ranged from 33% (95% among different countries worldwide. Cyprus and Malta have
CI, 25–44) for those with no family history of breast cancer to the highest age-adjusted mortality for breast cancer (29.6 per
58% (95% CI, 50–66) for those with two or more first-degree 100,000 population), whereas Haiti has the lowest (2.0 deaths
relatives with breast cancer at 50 years of age. The risk of breast per 100,000 population). The United States has an age-adjusted
cancer for female PALB2 mutation carriers, depending on the mortality for breast cancer of 19.0 cases per 100,000 population.
age, was about five to nine times as high compared with the gen- Women living in less industrialized nations tend to have a lower
eral population. While screening with mammogram along with incidence of breast cancer than women living in industrialized
MRI has been suggested for PALB2 mutation carriers starting countries, although Japan is an exception. In the United States,
at age 30 with consideration of risk-reducing mastectomy, there Mormons, Seventh Day Adventists, American Indians, Alaska
is currently insufficient evidence regarding the risk of ovarian natives, Hispanic/Latina Americans, and Japanese and Filipino
cancer and its management. women living in Hawaii have a below-average incidence of
Other hereditary syndromes associated with an increased breast cancer, whereas nuns (due to nulliparity) and Ashkenazi
risk of breast cancer include Cowden disease (PTEN mutations, Jewish women have an above-average incidence.
in which cancers of the thyroid, GI tract, and benign skin and The incidence rates of breast cancer increased in most
subcutaneous nodules are also seen), Li-Fraumeni syndrome countries through the 1990s. Since the estimates for 1990, there
(TP53 mutations, also associated with sarcomas, lymphomas, was an overall increase in incidence rates of approximately
and adrenocortical tumors), hereditary diffuse gastric cancer 0.5% annually. It was predicted that there would be approxi-
syndrome (CDH1 mutations, associated with diffuse gastric mately 1.4 million new cases in 2010. The cancer registries in
cancer and lobular breast cancers), and syndromes of breast China have noted annual increases in incidence of up to 3% to
and melanoma. With the discovery of additional genes related 4%, and in eastern Asia, increases are similar.
to breast cancer susceptibility, panel testing is available for a Data from the SEER program reveal declines in breast
number of genes in addition to BRCA1 and BRCA2. The inter- cancer incidence over the past decade, and this is widely attrib-
pretation of results is complex and is best done with a genetic uted to decreased use of hormone replacement therapy as a con-
counselor. sequence of the Women’s Health Initiative reports.118
Breast cancer burden has well-defined variations by geog-
raphy, regional lifestyle, and racial or ethnic background.119 In
EPIDEMIOLOGY AND NATURAL HISTORY OF general, both breast cancer incidence and mortality are rela-
tively lower among the female populations of Asia and Africa,
BREAST CANCER relatively underdeveloped nations, and nations that have not
Epidemiology adopted Westernized reproductive and dietary patterns. In
Breast cancer is the most common site-specific cancer in women contrast, European and North American women and women
and is the leading cause of death from cancer for women age from heavily industrialized or Westernized countries have a
20 to 59 years. Based on Surveillance, Epidemiology, and End substantially higher breast cancer burden. These international
Results registries (SEER) data, 266,120 new cases were esti- patterns are mirrored in breast cancer incidence and mortality
mated in 2018 with 40,920 estimated deaths attributed to breast rates observed for the racially, ethnically, and culturally diverse
cancers.117 It accounts for 30% of all newly diagnosed cancers in population of the United States.120
562 Although often related, the factors that influence breast Middlesex Hospital 1805-1933 (250 cases)

cancer incidence may differ from those that affect mortality.


Incidence rates are lower among populations that are heavily 100

weighted with women who begin childbearing at young ages 90


86%
and who have multiple full-term pregnancies followed by pro- 80
83%

longed lactation. These are features that characterize many


70
underdeveloped nations and also many eastern nations. Breast 68%
66%
cancer mortality rates should be lower in populations that have a 60

% Survival
lower incidence, but the mortality burden will simultaneously be 54%
50
adversely affected by the absence of effective mammographic 40 44% 41%
screening programs for early detection and diminished access
30
to multidisciplinary cancer treatment programs. These features
PART II

Natural survival
are likely to account for much of the disproportionate mortal- 20 28%
Survival untreated
ity risks that are seen in underdeveloped nations. Similar fac- 10
18% cases
9%
tors probably account for differences in breast cancer burden 3.6% 2% 0.8%
observed among the various racial and ethnic groups within
the United States. Interestingly, breast cancer incidence and
SPECIFIC CONSIDERATIONS

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19
mortality rates rise among second- and third-generation Asian Median
survival
Americans as they adopt Western lifestyles. 2.7 years
Disparities in breast cancer survival among subsets of the Duration of life from onset of symptoms (years)
American population are generating increased publicity because
Figure 17-13. Survival of women with untreated breast cancer
they are closely linked to disparities in socioeconomic status. compared with natural survival. (Reproduced with permission
Poverty rates and proportions of the population that lack health from Bloom HJG, Richardson WW, Harries EJ: Natural history
care insurance are two to three times higher among minority of untreated breast cancer (1805-1933). Comparison of untreated
racial and ethnic groups such as African Americans and His- and treated cases according to histological grade of malignancy,
panic/Latino Americans. These socioeconomic disadvantages Br Med J. 1962 Jul 28;2(5299):213-221.)
create barriers to effective breast cancer screening and result
in delayed breast cancer diagnosis, advanced stage distribu-
tion, inadequacies in comprehensive treatment, and, ultimately, who are likely to share ancestry with African American women
increased mortality rates. Furthermore, the rapid growth in the as a consequence of the Colonial-era slave trade. Interestingly,
Hispanic population is accompanied by increasing problems in male breast cancer also is seen with increased frequency among
health education because of linguistic barriers between physi- both African Americans and Africans.
cians and recently immigrated, non–English-speaking patients.
Recent studies also are documenting inequities in the treatments
Natural History
Bloom and colleagues described the natural history of breast
delivered to minority breast cancer patients, such as increased
cancer based on the records of 250 women with untreated breast
rates of failure to provide systemic therapy, use of sentinel
cancers who were cared for on charity wards in the Middlesex
lymph node dissection, and breast reconstruction. Some of the
Hospital, London, between 1805 and 1933. The median
treatment delivery disparities are related to inadequately con-
survival of this population was 2.7 years after initial diagnosis
trolled comorbidities (such as hypertension and diabetes), which
(Fig.  17-13).122 The 5- and 10-year survival rates for these
are more prevalent in minority populations. However, some
women were 18.0% and 3.6%, respectively. Only 0.8% survived
studies that adjust for these factors report persistent and unex-
for 15 years or longer. Autopsy data confirmed that 95% of these
plained unevenness in treatment recommendations. It is clear
women died of breast cancer, whereas the remaining 5% died
that breast cancer disparities associated with racial or ethnic
of other causes. Almost 75% of the women developed ulcer-
background have a multifactorial cause, and improvements in
ation of the breast during the course of the disease. The longest
outcome will require correction of many public health problems
surviving patient died in the 19th year after diagnosis.
at both the patient and provider levels.
Advances in the ability to characterize breast cancer sub- Primary Breast Cancer. More than 80% of breast cancers
types and the genetics of the disease are now provoking specula- show productive fibrosis that involves the epithelial and stro-
tion regarding possible hereditary influences on breast cancer mal tissues. With growth of the cancer and invasion of the
risk that are related to racial or ethnic ancestry.121 These questions surrounding breast tissues, the accompanying desmoplastic
become particularly compelling when one looks at disparities response entraps and shortens Cooper’s suspensory ligaments to
in breast cancer burden between African Americans and Cau- produce a characteristic skin retraction. Localized edema (peau
casians. Lifetime risk of breast cancer is lower for African d’orange) develops when drainage of lymph fluid from the skin
Americans, yet a paradoxically increased breast cancer mortal- is disrupted. With continued growth, cancer cells invade the
ity risk also is seen. African Americans also have a younger age skin, and eventually ulceration occurs. As new areas of skin
distribution for breast cancer; among women <45 years of age, are invaded, small satellite nodules appear near the primary
breast cancer incidence is highest among African Americans ulceration. The size of the primary breast cancer correlates with
compared to other subsets of the American population. Lastly disease-free and overall survival, but there is a close associa-
and most provocatively, African American women of all ages tion between cancer size and axillary lymph node involvement
have notably higher incidence rates for estrogen receptor- (Fig. 17-14). In general, up to 20% of breast cancer recurrences
negative tumors. These same patterns of disease are seen in con- are local-regional, >60% are distant, and 20% are both local-
temporary female populations of western, sub-Saharan Africa, regional and distant.
nodes adhere to each other and form a conglomerate mass. 563
100
315
Cancer cells may grow through the lymph node capsule and fix
90 * 297 to contiguous structures in the axilla, including the chest wall.
636 173
80 * N - (335)
Typically, axillary lymph nodes are involved sequentially from
177
the low (level I) to the central (level II) to the apical (level III)
Percent survivors

531
70 *
321
142
lymph node groups. Approximately 95% of the women who die
60 * 263
144 Whole series (716) of breast cancer have distant metastases, and traditionally the
50 *
234 N + 1 (198) most important prognostic correlate of disease-free and over-
65
*
40 *
*
all survival was axillary lymph node status (see Fig. 17-14A).
* * N + (381)
30
92 90 Women with node-negative disease had less than a 30% risk of

CHAPTER 17 THE BREAST


recurrence, compared with as much as a 75% risk for women
20 N + >3 (183)
25
with node-positive disease.
10
Distant Metastases. At approximately the 20th cell dou-
bling, breast cancers acquire their own blood supply (neovas-
2 4 6 8 10
cularization). Thereafter, cancer cells may be shed directly into
A Years after mastectomy
the systemic venous blood to seed the pulmonary circulation
via the axillary and intercostal veins or the vertebral column
via Batson’s plexus of veins, which courses the length of the
0.98 vertebral column. These cells are scavenged by natural killer
lymphocytes and macrophages. Successful implantation of
0.95
Proportion of patients with metastases

metastatic foci from breast cancer predictably occurs after the


0.90 x
primary cancer exceeds 0.5 cm in diameter, which corresponds
to the 27th cell doubling. For 10 years after initial treatment,
0.80 distant metastases are the most common cause of death in breast
x x cancer patients. For this reason, conclusive results cannot be
0.70 derived from breast cancer trials until at least 5 to 10 years
x
have elapsed. Although 60% of the women who develop distant
0.60
metastases will do so within 60 months of treatment, metastases
x may become evident as late as 20 to 30 years after treatment
0.50
x of the primary cancer.123 Patients with estrogen receptor nega-
0.40 tive breast cancers are proportionately more likely to develop
xx
recurrence in the first 3 to 5 years, whereas those with estrogen
0.30 receptor positive tumors have a risk of developing recurrence,
xx which drops off more slowly beyond 5 years than is seen with
0.20 ER-negative tumors.124 Recently, a report showed that tumor
10 100
size and nodal status remain powerful predictors of late recur-
Volume (ml) rences compared to more recently developed tools such as the
immunohistochemical score (IHC4) and two gene expression
2 3 4 5 6 7 8 9 10 11
profile tests (Recurrence Score and PAM50).125 Common sites
B Diameter (cm)
of involvement, in order of frequency, are bone, lung, pleura,
Figure 17-14. A. Overall survival for women with breast cancer soft tissues, and liver. Brain metastases are less frequent over-
according to axillary lymph node status. The time periods are years all, although with the advent of adjuvant systemic therapies it
after radical mastectomy. (Reproduced with permission from Vala- has been reported that CNS disease may be seen earlier.126,127
gussa P, Bonadonna G, Veronesi U, et al: Patterns of relapse and There are also reports of factors that are associated with the
survival following radical mastectomy. Analysis of 716 consecutive risk of developing brain metastases.128 For example, they are
patients, Cancer. 1978 Mar;41(3):1170-1178.) B. Risk of metasta- more likely to be seen in patients with triple receptor negative
ses according to breast cancer volume and diameter. (Reproduced breast cancer (ER-negative, PR-negative, and HER2-negative)
with permission from Koscielny S, Tubiana M, Lê MG, et al: Breast or patients with HER2-positive breast cancer who have received
cancer: Relationship between the size of the primary tumour and chemotherapy and HER2-directed therapies.
the probability of metastatic dissemination, Br J Cancer. 1984
Jun;49(6):709-715.)
HISTOPATHOLOGY OF BREAST CANCER
Carcinoma In Situ
Cancer cells are in situ or invasive depending on whether or not
Axillary Lymph Node Metastases. As the size of the pri- they invade through the basement membrane.129,130 Broders’s
mary breast cancer increases, some cancer cells are shed into original description of in situ breast cancer stressed the absence
cellular spaces and transported via the lymphatic network of of invasion of cells into the surrounding stroma and their confine-
the breast to the regional lymph nodes, especially the axillary ment within natural ductal and alveolar boundaries.129 Because
lymph nodes. Lymph nodes that contain metastatic cancer are areas of invasion may be minute, the accurate diagnosis of in
at first ill-defined and soft but become firm or hard with con- situ cancer necessitates the analysis of multiple microscopic sec-
tinued growth of the metastatic cancer. Eventually the lymph tions to exclude invasion. In 1941, Foote and Stewart published
564 a landmark description of LCIS, which distinguished it from and contributes to its diagnosis. The frequency of LCIS in the
DCIS.130 In the late 1960s, Gallagher and Martin published their general population cannot be reliably determined because it usu-
study of whole-breast sections and described a stepwise progres- ally presents as an incidental finding. The average age at diag-
sion from benign breast tissue to in situ cancer and subsequently nosis is 45 years, which is approximately 15 to 25 years younger
to invasive cancer. Before the widespread use of mammography, than the age at diagnosis for invasive breast cancer. LCIS has a
diagnosis of breast cancer was by physical examination. At that distinct racial predilection, occurring 12 times more frequently
time, in situ cancers constituted <6% of all breast cancers, and in white women than in African-American women. Invasive
LCIS was more frequently diagnosed than DCIS by a ratio of breast cancer develops in 25% to 35% of women with LCIS.
>2:1. However, when screening mammography became popular, Invasive cancer may develop in either breast, regardless of
a 14-fold increase in the incidence of in situ cancer (45%) was which breast harbored the initial focus of LCIS, and is detected
demonstrated, and DCIS was more frequently diagnosed than synchronously with LCIS in 5% of cases. In women with a his-
LCIS by a ratio of >2:1. Table 17-8 lists the clinical and patho- tory of LCIS, up to 65% of subsequent invasive cancers are duc-
PART II

logic characteristics of DCIS and LCIS. Multicentricity refers tal, not lobular, in origin. For these reasons, LCIS is regarded
to the occurrence of a second breast cancer outside the breast as a marker of increased risk for invasive breast cancer rather
quadrant of the primary cancer (or at least 4 cm away), whereas than as an anatomic precursor. Individuals should be counseled
multifocality refers to the occurrence of a second cancer within regarding their risk of developing breast cancer and appropriate
the same breast quadrant as the primary cancer (or within 4 cm of risk reduction strategies, including observation with screening,
SPECIFIC CONSIDERATIONS

it). Multicentricity occurs in 60% to 90% of women with LCIS, chemoprevention, and risk-reducing bilateral mastectomy.
whereas the rate of multicentricity for DCIS is reported to be Ductal Carcinoma In Situ. Although DCIS is predominantly
40% to 80%. LCIS occurs bilaterally in 50% to 70% of cases, seen in the female breast, it accounts for 5% of male breast
whereas DCIS occurs bilaterally in 10% to 20% of cases. cancers. Published series suggest a detection frequency of 7% in
Lobular Carcinoma In Situ. LCIS originates from the termi- all biopsy tissue specimens. The term intraductal carcinoma is
nal duct lobular units and develops only in the female breast. frequently applied to DCIS, which carries a high risk for progres-
It is characterized by distention and distortion of the terminal sion to an invasive cancer. Histologically, DCIS is characterized
duct lobular units by cells that are large but maintain a normal by a proliferation of the epithelium that lines the minor ducts,
nuclear to cytoplasmic ratio. Cytoplasmic mucoid globules are resulting in papillary growths within the duct lumina. Early in
a distinctive cellular feature. LCIS may be observed in breast their development, the cancer cells do not show pleomorphism,
tissues that contain microcalcifications, but the calcifications mitoses, or atypia, which leads to difficulty in distinguishing
associated with LCIS typically occur in adjacent tissues. This early DCIS from benign hyperplasia. The papillary growths
neighborhood calcification is a feature that is unique to LCIS (papillary growth pattern) eventually coalesce and fill the duct
lumina so that only scattered, rounded spaces remain between
the clumps of atypical cancer cells, which show hyperchroma-
sia and loss of polarity (cribriform growth pattern). Eventually
Table 17-8
pleomorphic cancer cells with frequent mitotic figures obliterate
Salient characteristics of in situ ductal (DCIS) and the lumina and distend the ducts (solid growth pattern). With
lobular (LCIS) carcinoma of the breast continued growth, these cells outstrip their blood supply and
become necrotic (comedo growth pattern). Calcium deposition
  LCIS DCIS occurs in the areas of necrosis and is a common feature seen
Age (years) 44–47 54–58 on mammography. DCIS is now frequently classified based on
Incidencea 2%–5% 5%–10% nuclear grade and the presence of necrosis (Table 17-9). Based
Clinical signs None Mass, pain, nipple
discharge
Mammographic signs None Microcalcifications Table 17-9
Premenopausal 2/3 1/3 Classification of breast ductal carcinoma in situ (DCIS)
Incidence of synchronous 5% 2%–46% DETERMINING
invasive carcinoma
CHARACTERISTICS
Multicentricity 60%–90% 40%–80%
Bilaterality 50%–70% 10%–20% HISTOLOGIC NUCLEAR
Axillary metastasis 1% 1%–2% SUBTYPE  GRADE NECROSIS DCIS GRADE 
Subsequent carcinomas:     Comedo High Extensive High
Incidence 25%–35% 25%–70% Intermediatea Intermediate Focal or absent Intermediate
Laterality Bilateral Ipsilateral Noncomedo b
Low Absent Low
Interval to diagnosis 15–20 y 5–10 y a
Often a mixture of noncomedo patterns.
b
Solid, cribriform, papillary, or focal micropapillary.
Histologic type Ductal Ductal
Adapted with permission from Koo JS, Kim MJ, Kim EK, et al:
In biopsy specimens of mammographically detected breast lesions.
a
Comparison of immunohistochemical staining in breast papillary
Reproduced with permission from Bland KI, Copeland ED: The Breast: neoplasms of cytokeratin 5/6 and p63 in core needle biopsies and
Comprehensive Management of Benign and Malignant Diseases, 2nd ed. surgical excisions, Appl Immunohistochem Mol Morphol. 2012
Philadelphia, PA: Elsesvier/Saunders; 1998. Mar;20(2):108-115.
on multiple consensus meetings, grading of DCIS has been rec- Current histologic classifications recognize special types of 565
ommended. Although there is no universal agreement on clas- breast cancers (10% of total cases), which are defined by spe-
sification, most systems endorse the use of cytologic grade and cific histologic features. To qualify as a special-type cancer, at
presence or absence of necrosis.131 least 90% of the cancer must contain the defining histologic
The risk for invasive breast cancer is increased nearly features. About 80% of invasive breast cancers are described as
fivefold in women with DCIS.132 The invasive cancers are invasive ductal carcinoma of no special type (NST). These can-
observed in the ipsilateral breast, usually in the same quadrant cers generally have a worse prognosis than special-type cancers.
as the DCIS that was originally detected, which suggests that Foote and Stewart originally proposed the following classifica-
DCIS is an anatomic precursor of invasive ductal carcinoma tion for invasive breast cancer130:
(Fig. 17-15A and B).

CHAPTER 17 THE BREAST


1. Paget’s disease of the nipple
Invasive Breast Carcinoma 2. Invasive ductal carcinoma—Adenocarcinoma with produc-
Invasive breast cancers have been described as lobular or duc-
tive fibrosis (scirrhous, simplex, NST), 80%
tal in origin.128-131 Early classifications used the term lobular
3. Medullary carcinoma, 4%
to describe invasive cancers that were associated with LCIS,
4. Mucinous (colloid) carcinoma, 2%
whereas all other invasive cancers were referred to as ductal.
5. Papillary carcinoma, 2%
6. Tubular carcinoma, 2%
7. Invasive lobular carcinoma, 10%
8. Rare cancers (adenoid cystic, squamous cell, apocrine)

Paget’s disease of the nipple was described in 1874. It fre-


quently presents as a chronic, eczematous eruption of the nipple,
which may be subtle but may progress to an ulcerated, weeping
lesion. Paget’s disease usually is associated with extensive DCIS
and may be associated with an invasive cancer. A palpable mass
may or may not be present. A nipple biopsy specimen will show
a population of cells that are identical to the underlying DCIS
cells (pagetoid features or pagetoid change). Pathognomonic
of this cancer is the presence of large, pale, vacuolated cells
(Paget cells) in the rete pegs of the epithelium. Paget’s disease
may be confused with superficial spreading melanoma. Differ-
entiation from pagetoid intraepithelial melanoma is based on the
presence of S-100 antigen immunostaining in melanoma and
carcinoembryonic antigen immunostaining in Paget’s disease.
A Surgical therapy for Paget’s disease may involve lumpectomy
or mastectomy, depending on the extent of involvement of the
nipple-areolar complex and the presence of DCIS or invasive
cancer in the underlying breast parenchyma.
Invasive ductal carcinoma of the breast with productive
fibrosis (scirrhous, simplex, NST) accounts for 80% of breast
cancers and presents with macroscopic or microscopic axillary
lymph node metastases in up to 25% of screen-detected cases
and up to 60% of symptomatic cases. This cancer occurs most
frequently in perimenopausal or postmenopausal women in the
fifth to sixth decades of life as a solitary, firm mass. It has poorly
defined margins, and its cut surfaces show a central stellate con-
figuration with chalky white or yellow streaks extending into
surrounding breast tissues. The cancer cells often are arranged
in small clusters, and there is a broad spectrum of histologic
types with variable cellular and nuclear grades (Fig. 17-16A
and B). In a large patient series from the SEER database, 75%
of ductal cancers showed estrogen receptor expression.133
B Medullary carcinoma is a special-type breast cancer;
Figure 17-15. Ductal carcinoma in situ (DCIS). A. Craniocau- it accounts for 4% of all invasive breast cancers and is a fre-
dal mammographic view shows a poorly defined mass containing quent phenotype of BRCA1 hereditary breast cancer. Grossly,
microcalcifications. (Used with permission from Dr. Anne Turnbull, the cancer is soft and hemorrhagic. A rapid increase in size
Consultant Radiologist/Director of Breast Screening, Royal Derby may occur secondary to necrosis and hemorrhage. On physi-
Hospital, Derby, UK.) B. Histopathologic preparation of the sur- cal examination, it is bulky and often positioned deep within
gical specimen confirms DCIS with necrosis (100x). (Used with the breast. Bilaterality is reported in 20% of cases. Medullary
permission from Dr. Sindhu Menon, Consultant Histopathologist carcinoma is characterized microscopically by: (a) a dense lym-
and Dr. Rahul Deb, Consultant Histopathologist and Lead Breast phoreticular infiltrate composed predominantly of lymphocytes
Pathologist, Royal Derby Hospital, Derby, UK.) and plasma cells; (b) large pleomorphic nuclei that are poorly
566
PART II
SPECIFIC CONSIDERATIONS

A B

Figure 17-16. Invasive ductal carcinoma with productive fibrosis (scirrhous, simplex, no special type) A. 100x. B. 200x. (Used with permis-
sion from Dr. Sindhu Menon, Consultant Histopathologist and Dr. Rahul Deb, Consultant Histopathologist and Lead Breast Pathologist,
Royal Derby Hospital, Derby, UK.)

differentiated and show active mitosis; and (c) a sheet-like by mammographic screening and usually is diagnosed in the
growth pattern with minimal or absent ductal or alveolar dif- perimenopausal or early menopausal periods. Under low-power
ferentiation. Approximately 50% of these cancers are associated magnification, a haphazard array of small, randomly arranged
with DCIS, which characteristically is present at the periphery tubular elements is seen. In a large SEER database 94% of
of the cancer, and <10% demonstrate hormone receptors. In rare tubular cancers were reported to express estrogen receptor.133
circumstances, mesenchymal metaplasia or anaplasia is noted. Approximately 10% of women with tubular carcinoma or with
Because of the intense lymphocyte response associated with invasive cribriform carcinoma, a special-type cancer closely
the cancer, benign or hyperplastic enlargement of the lymph related to tubular carcinoma, will develop axillary lymph node
nodes of the axilla may contribute to erroneous clinical staging. metastases. However, the presence of metastatic disease in one
Women with this cancer have a better 5-year survival rate than or two axillary lymph nodes does not adversely affect survival.
those with NST or invasive lobular carcinoma. Distant metastases are rare in tubular carcinoma and invasive
Mucinous carcinoma (colloid carcinoma), another spe- cribriform carcinoma. Long-term survival approaches 100%.
cial-type breast cancer, accounts for 2% of all invasive breast Invasive lobular carcinoma accounts for 10% of breast
cancers and typically presents in the older population as a bulky cancers. The histopathologic features of this cancer include
tumor. This cancer is defined by extracellular pools of mucin, small cells with rounded nuclei, inconspicuous nucleoli, and
which surround aggregates of low-grade cancer cells. The cut scant cytoplasm (Fig. 17-17). Special stains may confirm the
surface of this cancer is glistening and gelatinous in quality.
Fibrosis is variable, and when abundant it imparts a firm consis-
tency to the cancer. Over 90% of mucinous carcinomas display
hormone receptors.133 Lymph node metastases occur in 33%
of cases, and 5- and 10-year survival rates are 73% and 59%,
respectively. Because of the mucinous component, cancer cells
may not be evident in all microscopic sections, and analysis
of multiple sections is essential to confirm the diagnosis of a
mucinous carcinoma.
Papillary carcinoma is a special-type cancer of the breast
that accounts for 2% of all invasive breast cancers. It generally
presents in the seventh decade of life and occurs in a dispropor-
tionate number of nonwhite women. Typically, papillary car-
cinomas are small and rarely attain a size of 3 cm in diameter.
These cancers are defined by papillae with fibrovascular stalks
and multilayered epithelium. In a large series from the SEER
database 87% of papillary cancers have been reported to express
estrogen receptor.133 McDivitt and colleagues noted that these
tumors showed a low frequency of axillary lymph node metas- Figure 17-17. Lobular carcinoma (100×). Uniform, relatively
tases and had 5- and 10-year survival rates similar to those for small lobular carcinoma cells are seen arranged in a single-file
mucinous and tubular carcinoma.134 orientation (“Indian file”). (Used with permission from Dr. Sindhu
Tubular carcinoma is another special-type breast cancer Menon, Consultant Histopathologist and Dr. Rahul Deb, Consul-
and accounts for 2% of all invasive breast cancers. It is reported tant Histopathologist and Lead Breast Pathologist, Royal Derby
in as many as 20% of women whose cancers are diagnosed Hospital, Derby, UK.)
presence of intracytoplasmic mucin, which may displace the 567
nucleus (signet-ring cell carcinoma). At presentation, invasive
lobular carcinoma varies from clinically inapparent carcinomas
to those that replace the entire breast with a poorly defined mass.
It is frequently multifocal, multicentric, and bilateral. Because
of its insidious growth pattern and subtle mammographic fea-
tures, invasive lobular carcinoma may be difficult to detect.
Over 90% of lobular cancers express estrogen receptor.133

DIAGNOSIS OF BREAST CANCER

CHAPTER 17 THE BREAST


In ∼30% of cases, the woman discovers a lump in her breast.
Other less frequent presenting signs and symptoms of breast
cancer include: (a) breast enlargement or asymmetry; (b) nipple
changes, retraction, or discharge; (c) ulceration or erythema of
the skin of the breast; (d) an axillary mass; and (e) musculoskel-
etal discomfort. However, up to 50% of women presenting with
breast complaints have no physical signs of breast pathology.
Breast pain usually is associated with benign disease.
Misdiagnosed breast cancer accounts for the greatest num- Figure 17-19. A breast examination record.
ber of malpractice claims for errors in diagnosis and for the
largest number of paid claims. Litigation often involves younger
women, whose physical examination and mammogram may
be misleading. If a young woman (≤45 years) presents with a Palpation. As part of the physical examination, the breast is
palpable breast mass and equivocal mammographic findings, carefully palpated. With the patient in the supine position (see
ultrasound examination and biopsy are used to avoid a delay Fig. 17-18C) the clinician gently palpates the breasts, making
in diagnosis. certain to examine all quadrants of the breast from the sternum
laterally to the latissimus dorsi muscle and from the clavicle
Examination
inferiorly to the upper rectus sheath. The examination is per-
Inspection. The clinician inspects the woman’s breast with formed with the palmar aspects of the fingers, avoiding a grasp-
her arms by her side (Fig. 17-18A), with her arms straight up ing or pinching motion. The breast may be cupped or molded
in the air (Fig. 17-18B), and with her hands on her hips (with in the examiner’s hands to check for retraction. A systematic
and without pectoral muscle contraction).135,136 Symmetry, size, search for lymphadenopathy then is performed. Figure 17-18D
and shape of the breast are recorded, as well as any evidence of shows the position of the patient for examination of the axilla.
edema (peau d’orange), nipple or skin retraction, or erythema. By supporting the upper arm and elbow, the examiner stabi-
With the arms extended forward and in a sitting position, the lizes the shoulder girdle. Using gentle palpation, the clinician
woman leans forward to accentuate any skin retraction. assesses all three levels of possible axillary lymphadenopathy.
Careful palpation of supraclavicular and parasternal sites also is
performed. A diagram of the chest and contiguous lymph node
sites is useful for recording location, size, consistency, shape,
mobility, fixation, and other characteristics of any palpable
breast mass or lymphadenopathy (Fig. 17-19).
Imaging Techniques
Mammography. Mammography has been used in North Amer-
ica since the 1960s, and the techniques used continue to be mod-
ified and improved to enhance image quality.137-140 Conventional
mammography delivers a radiation dose of 0.1 cGy per study.
By comparison, chest radiography delivers 25% of this dose.
However, there is no increased breast cancer risk associated
with the radiation dose delivered with screening mammography.
Screening mammography is used to detect unexpected breast
cancer in asymptomatic women. In this regard, it supplements
history taking and physical examination. With screening mam-
mography, two views of the breast are obtained: the craniocau-
dal (CC) view (Fig. 17-20A,B) and the mediolateral oblique
(MLO) view (Fig. 17-20C,D). The MLO view images the great-
est volume of breast tissue, including the upper outer quadrant
Figure 17-18. Examination of the breast. A. Inspection of the and the axillary tail of Spence. Compared with the MLO view,
breast with arms at sides. B. Inspection of the breast with arms the CC view provides better visualization of the medial aspect
raised. C. Palpation of the breast with the patient supine. D. Palpa- of the breast and permits greater breast compression. Diagnos-
tion of the axilla. tic mammography is used to evaluate women with abnormal
568
PART II
SPECIFIC CONSIDERATIONS

A B

C D

Figure 17-20. A-D. Mammogram of a premenopausal breast with a dense fibroglandular pattern. E-H. Mammogram of a postmenopausal
breast with a sparse fibroglandular pattern. (Used with permission from Dr. Anne Turnbull, Consultant Radiologist/Director of Breast Screening,
Royal Derby Hospital, Derby, UK.)
569

CHAPTER 17 THE BREAST


E F

G H
Figure 17-20. (Continued)

findings such as a breast mass or nipple discharge. In addition overlying tissues (Fig. 17-21C). The compression device mini-
to the MLO and CC views, a diagnostic examination may use mizes motion artifact, improves definition, separates overlying
views that better define the nature of any abnormalities, such as tissues, and decreases the radiation dose needed to penetrate
the 90° lateral and spot compression views. The 90° lateral view the breast. Magnification techniques (×1.5) often are combined
is used along with the CC view to triangulate the exact location with spot compression to better resolve calcifications and the
of an abnormality. Spot compression may be done in any pro- margins of masses. Mammography also is used to guide inter-
jection by using a small compression device, which is placed ventional procedures, including needle localization and needle
directly over a mammographic abnormality that is obscured by biopsy.
570
PART II
SPECIFIC CONSIDERATIONS

A B C

Figure 17-21. Mammogram revealing a small, spiculated mass in the right breast A. A small, spiculated mass is seen in the right breast with
skin tethering (CC view). B. Mass seen on oblique view of the right breast. C. Spot compression mammography view of the cancer seen in
A and B. The spiculated margins of the cancer are accentuated by compression. (Used with permission from Dr. Anne Turnbull, Consultant
Radiologist/Director of Breast Screening, Royal Derby Hospital, Derby, UK.)

Specific mammographic features that suggest a diagnosis mammography in women <50 years of age is more controversial
of breast cancer include a solid mass with or without stellate for previously noted reasons: (a) reduced sensitivity, (b) reduced
features, asymmetric thickening of breast tissues, and clustered specificity, and (c) lower incidence of breast cancer. Because of
microcalcifications. The presence of fine, stippled calcium in the combination of these three reasons, targeting mammography
and around a suspicious lesion is suggestive of breast cancer screening to women <50 years of age, who are at higher risk of
and occurs in as many as 50% of nonpalpable cancers. These breast cancer, improves the balance of risks and benefits and is
microcalcifications are an especially important sign of cancer the approach some health care systems have taken. There are
in younger women, in whom it may be the only mammographic now a number of risk assessment models—as described earlier
abnormality. The clinical impetus for screening mammogra- in this chapter—that can be used to estimate a younger woman’s
phy came from the Health Insurance Plan study and the Breast risk of developing breast cancer and that help assess the risks
Cancer Detection Demonstration Project, which demonstrated and benefits of regular screening.
a 33% reduction in mortality for women after72 screening mam- Screen film mammography has replaced xeromam-
mography. Mammography was more accurate than clinical mography because it requires a lower dose of radiation and
examination for the detection of early breast cancers, providing provides similar image quality. Digital mammography was
a true-positive rate of 90%. Only 20% of women with nonpal- developed to allow the observer to manipulate the degree
pable cancers had axillary lymph node metastases, compared of contrast in the image. This is especially useful in women
with 50% of women with palpable cancers.141 Current guide- with dense breasts and women <50 years of age. Recently,
lines of the National Comprehensive Cancer Network suggest investigators directly compared digital vs. screen film mam-
that normal-risk women ≥20 years of age should have a breast mography in a prospective (DMIST) trial that enrolled over
examination at least every 3 years. Starting at age 40 years, 42,000 women.143 The investigators found that digital and
breast examinations should be performed yearly, and a yearly screen film mammography had similar accuracy; however,
mammogram should be taken.142 Screening mammography in digital mammography was more accurate in women <50 years
women ≥50 years of age has been noted to reduce breast cancer of age, women with mammographically dense breasts, and
mortality by 20% to 25%.72,79 With the increased discussion premenopausal or perimenopausal women. The use of digital
about the potential harms associated with breast screening, the breast tomosynthesis with 3D images has been introduced as
United Kingdom recently established an independent expert an alternative to standard 2D mammography imaging that is
panel to review the published literature and estimate the ben- limited by superimposition of breast parenchyma and breast
efits and harms associated with its national screening program density.144,145 The STORM trial reported that in 7,292 women
for women age >50 years. The panel estimated that in women screened, 3D mammography had a higher cancer detection
invited to screening, about 11% of the cancers diagnosed in rate and fewer false-positive recalls than the standard 2D
their lifetime constitute overdiagnosis. Despite this overdiagno- imaging.146,147 Randomized controlled trials are planned to fur-
sis, the panel concluded that breast screening programs confer ther study tomosynthesis and its role in breast cancer screen-
significant benefit and should continue. The use of screening ing. Standard two-dimensional mammography has limitations,
such as the parenchymal density or superimposition of breast 571
tissue, which obscures cancers or causes normal structures to
appear suspicious reducing the sensitivity of mammography
and increasing the false-positive rates. Digital breast tomo-
synthesis is a technology developed to assist with overcom-
ing these limitations. In digital breast tomosynthesis, multiple
projection images are reconstructed to allow visual review of
thin breast sections, each reconstructed slice as thin as 0.5 mm,
which provides better characterization of noncalcified lesions.
These multiple projection exposures are obtained by a digi-

CHAPTER 17 THE BREAST


tal detector from a mammography X-ray source that moves
through a limited arc angle while the breast is compressed.
Then these projection image data sets are reconstructed using
specific algorithms, which provide the clinical reader a series
of images through the entire breast.148
In 2011, tomosynthesis was approved by the U.S. Food
and Drug Administration (FDA) to be used in combination with
standard digital mammography for breast cancer screening. The
total radiation dose when tomosynthesis is added is about twice
the current dose of digital mammography alone but remains
below the limits set by the FDA.149
The STORM-2 trial reported that synthetic 2D-3D
A
mammography yields similar breast cancer detection as dual-
acquisition 2D-3D mammography with the advantage of reduc-
ing radiation exposure.150
Contrast-enhanced digital mammography (CEDM) was
also approved by the FDA in 2001, which utilizes an iodinated
contrast material and modified digital mammography units for
imaging.148 CEDM has been shown to be feasible and detects
breast cancers at a rate similar to MRI, which has potential to
offer an alternative to MRI.151 The advantages of CEDM over
MRI are that the use of compression limits motion, there is
decrease in cost, decrease in exam time, and there is an option
for patients who are unable to tolerate MRI or who due to vari-
ous reasons cannot have MRI due to incompatibility, such as the
presence of a pacemaker or tissue expanders.148,152
Ductography. The primary indication for ductography is
nipple discharge, particularly when the fluid contains blood.
Radiopaque contrast media is injected into one or more of the
major ducts, and mammography is performed. A duct is gen-
tly enlarged with a dilator, and then a small, blunt cannula is
inserted under sterile conditions into the nipple ampulla. With
the patient in a supine position, 0.1 to 0.2 mL of dilute con-
trast media is injected, and CC and MLO mammographic views
are obtained without compression. Intraductal papillomas are B
seen as small filling defects surrounded by contrast media
(Fig. 17-22). Cancers may appear as irregular masses or as mul- Figure 17-22. Ductogram. Craniocaudal (A) and mediolateral
tiple intraluminal filling defects. oblique (B) mammographic views demonstrate a mass (arrows)
posterior to the nipple and outlined by contrast, which also fills
Ultrasonography. Second only to mammography in fre- the proximal ductal structures. (Used with permission from B.
quency of use for breast imaging, ultrasonography is an impor- Steinbach.)
tant method of resolving equivocal mammographic findings,
defining cystic masses, and demonstrating the echogenic qual-
ities of specific solid abnormalities. On ultrasound examina- of breast lesions. Its findings are highly reproducible, and
tion, breast cysts are well circumscribed, with smooth margins it has a high patient acceptance rate, but it does not reliably
and an echo-free center (Fig. 17-23). Benign breast masses detect lesions that are ≤1 cm in diameter. Ultrasonography can
usually show smooth contours, round or oval shapes, weak also be utilized to image the regional lymph nodes in patients
internal echoes, and well-defined anterior and posterior mar- with breast cancer (Fig. 17-26). The sensitivity of examination
gins (Fig. 17-24). Breast cancer characteristically has irregular for the status of axillary nodes ranges from 35% to 82% and
walls (Fig. 17-25) but may have smooth margins with acous- specificity ranges from 73% to 97%. The features of a lymph
tic enhancement. Ultrasonography is used to guide fine-needle node involved with cancer include cortical thickening, change
aspiration biopsy, core-needle biopsy, and needle localization in shape of the node to more circular appearance, size larger
572
PART II

A
SPECIFIC CONSIDERATIONS

Figure 17-24. Ultrasonography images of benign breast tumors.


A. Fibroadenoma. B. Intraductal papilloma (see arrow). (Used with
permission from Dr. Anne Turnbull, Consultant Radiologist/Director
of Breast Screening, Royal Derby Hospital, Derby, UK.)

than 10 mm, absence of a fatty hilum and hypoechoic internal


B echoes.153
Magnetic Resonance Imaging. In the process of evaluating
magnetic resonance imaging (MRI) as a means of character-
izing mammographic abnormalities, additional breast lesions
have been detected. However, in the circumstance of negative
findings on both mammography and physical examination,
the probability of a breast cancer being diagnosed by MRI
is extremely low. There is current interest in the use of MRI
to screen the breasts of high-risk women and of women with
a newly diagnosed breast cancer. In the first case, women
who have a strong family history of breast cancer or who
carry known genetic mutations require screening at an early
age because mammographic evaluation is limited due to the
increased breast density in younger women. In the second
case, an MRI study of the contralateral breast in women with
C a known breast cancer has shown a contralateral breast cancer
in 5.7% of these women (Fig. 17-27). MRI can also detect
Figure 17-23. Breast cyst. A. Simple cyst. Ultrasound image additional tumors in the index breast (multifocal or multicen-
of the mass shows it to be anechoic with a well-defined back
tric disease) that may be missed on routine breast imaging and
wall, characteristic of a cyst. B. Complex solid and cystic mass.
this may alter surgical decision making (Fig. 17-28). In fact,
C. Complex solid and cystic mass characteristic of intracystic
MRI has been advocated by some for routine use in surgical
papillary tumor. (Used with permission from Dr. Anne Turnbull,
treatment planning based on the fact that additional disease
Consultant Radiologist/Director of Breast Screening, Royal Derby
Hospital, Derby, UK.) can be identified with this advanced imaging modality and the
573

CHAPTER 17 THE BREAST


B
A

C D

Figure 17-25. Ultrasonography images of malignant breast lesions. A. 25 mm irregular mass. B. Ultrasound 30 mm mass anterior to an
implant. C. Ultrasound breast cancer with calcification. D. Ultrasound shows a 9 mm spiculated mass (see arrow) with attenuation. (Used with
permission from Dr. Anne Turnbull, Consultant Radiologist/Director of Breast Screening, Royal Derby Hospital, Derby, UK.)

extent of disease may be more accurately assessed. A random- whole breast irradiation and systemic therapies. There is an
ized trial performed in the United Kingdom (COMICE trial) ongoing trial in the Alliance for Clinical Trials in Oncology
that enrolled 1623 women did not show a decrease in rates of that is randomizing patients to preoperative MRI vs. standard
reoperation in those women randomized to undergo MRI in imaging to assess the impact of MRI on local regional recur-
addition to mammography and ultrasonography (19%) com- rence rates in patients with triple receptor negative and HER2
pared to those undergoing standard breast imaging without positive breast cancers.
MRI (19%).154 Houssami and colleagues performed a meta- The use of dedicated breast coils is mandatory in the MRI
analysis including two randomized trials and seven compara- imaging of the breast. A BIRADS lexicon is assigned to each
tive cohort studies to examine the effect of preoperative MRI examination and an abnormality noted on MRI that is not seen
compared to standard preoperative evaluation on surgical out- on mammography requires a focused ultrasound examination
comes.155 They reported that the use of MRI was associated for further assessment. If the abnormality is not seen on corre-
with increased mastectomy rates. This is problematic because sponding mammogram or ultrasound, then MRI-guided biopsy
there is no evidence that the additional disease detected by is necessary. Some clinical scenarios where MRI may be use-
MRI is of clinical or biologic significance, particularly in ful include the evaluation of a patient who presents with nodal
light of the low local-regional failure rates currently reported metastasis from breast cancer without an identifiable primary
in patients undergoing breast conserving surgery who receive tumor; to assess response to therapy in the setting of neoadjuvant
574
PART II
SPECIFIC CONSIDERATIONS

Figure 17-26. Ultrasonography images of lymph nodes. A. Nor-


mal axillary lymph node (see arrows). B. Indeterminate axillary
lymph node. C. Malignant appearing axillary lymph node. (Used
with permission from Dr. Anne Turnbull, Consultant Radiologist/
B Director of Breast Screening, Royal Derby Hospital, Derby, UK.)

systemic treatment; to select patients for partial breast irradia- cytologic evaluation, core-needle permits the analysis of breast
tion techniques; and evaluation of the treated breast for tumor tissue architecture and allows the pathologist to determine
recurrence. whether invasive cancer is present. This permits the surgeon and
patient to discuss the specific management of a breast cancer
Breast Biopsy before therapy begins. Core-needle biopsy is preferred over
Nonpalpable Lesions. Image-guided breast biopsy specimens open biopsy for nonpalpable breast lesions because a single sur-
are frequently required to diagnose nonpalpable lesions.156 gical procedure can be planned based on the results of the core
biopsy. The advantages of core-needle biopsy include a
Ultrasound localization techniques are used when a mass is
present, whereas stereotactic techniques are used when no mass
7 low complication rate, minimal scarring, and a lower cost
is present (microcalcifications or architectural distortion only). compared with excisional breast biopsy.
The combination of diagnostic mammography, ultrasound or Palpable Lesions. FNA or core biopsy of a palpable breast mass
stereotactic localization, and fine-needle aspiration (FNA) can usually be performed in an outpatient setting.157 A 1.5-in,
biopsy achieves almost 100% accuracy in the preoperative diag- 22-gauge needle attached to a 10-mL syringe or a 14-gauge
nosis of breast cancer. However, although FNA biopsy permits core biopsy needle is used. For FNA, use of a syringe holder

Figure 17-27. MRI examination revealing contral-


ateral breast cancer (see arrows) in a patient diag-
nosed with unilateral breast cancer on mammography
(two arrows). (Used with permission from Dr. Anne
Turnbull, Consultant Radiologist/Director of Breast
Screening, Royal Derby Hospital, Derby, UK.)
575

Figure 17-28. MRI imaging of the breast reveal-


ing multifocal tumors not detected with standard
breast imaging. (Used with permission from Dr. Anne
Turnbull, Consultant Radiologist/Director of Breast
Screening, Royal Derby Hospital, Derby, UK.)

CHAPTER 17 THE BREAST


enables the surgeon performing the FNA biopsy to control the the findings from pathologic examination of the resected pri-
syringe and needle with one hand while positioning the breast mary breast cancer and axillary or other regional lymph nodes.
mass with the opposite hand. After the needle is placed in the Fisher and colleagues found that accurate predictions regarding
mass, suction is applied while the needle is moved back and the occurrence of distant metastases were possible after resec-
forth within the mass. Once cellular material is seen at the hub tion and pathologic analysis of 10 or more levels I and II axillary
of the needle, the suction is released and the needle is with- lymph nodes.160 A frequently used staging system is the TNM
drawn. The cellular material is then expressed onto microscope (tumor, nodes, and metastasis) system. The American Joint
slides. Both air-dried and 95% ethanol–fixed microscopic sec- Committee on Cancer (AJCC) has recently modified the TNM
tions are prepared for analysis. When a breast mass is clinically system for breast cancer to include both anatomic and biologic
and mammographically suspicious, the sensitivity and specific- factors161 (Tables 17-10 and 17-11). Koscielny and colleagues
ity of FNA biopsy approaches 100%. Core-needle biopsy of demonstrated that tumor size correlates with the presence of
palpable breast masses is performed using a 14-gauge needle, axillary lymph node metastases (see Fig. 17-14B). Others have
such as the Tru-Cut needle. Automated devices also are avail- shown an association between tumor size, axillary lymph node
able. Vacuum-assisted core biopsy devices (with 8–10 gauge metastases, and disease-free survival. One of the most important
needles) are commonly utilized with image guidance where predictors of 10- and 20-year survival rates in breast cancer is
between 4 and 12 samples can be acquired at different posi- the number of axillary lymph nodes involved with metastatic
tions within a mass, area of architectural distortion or micro- disease. Routine biopsy of internal mammary lymph nodes is
calcifications. If the target lesion was microcalcifications, the not generally performed; however, it has been reported that in
specimen should be radiographed to confirm appropriate sam- the context of a “triple node” biopsy approach either the internal
pling. A radiopaque marker should be placed at the site of the mammary node or a low axillary node when positive alone
biopsy to mark the area for future intervention. In some cases carried the same prognostic weight. When both nodes were
the entire lesion is removed with the biopsy technique and clip positive, the prognosis declined to the level associated with
placement allows for accurate targeting of the site for surgi- apical node positivity. A double node biopsy of the low axil-
cal resection. Tissue specimens are placed in formalin and then lary node and either the apical or the internal mammary node
processed to paraffin blocks. Although the false-negative rate gave the same maximum prognostic information as a triple node
for core-needle biopsy specimens is very low, a tissue speci- biopsy.162 With the advent of sentinel lymph node dissection
men that does not show breast cancer cannot conclusively rule and the use of preoperative lymphoscintigraphy for localization
out that diagnosis because a sampling error may have occurred. of the sentinel nodes, surgeons have again begun to biopsy the
The clinical, radiographic, and pathologic findings should be internal mammary nodes but in a more targeted manner. The
in concordance. If the biopsy findings do not concur with the 8th edition of the AJCC staging system does allow for staging
clinical and radiographic findings, the multidisciplinary team based on findings from the internal mammary sentinel nodes.163
(including clinician, radiologist, and pathologist) should review Drainage to the internal mammary nodes is more frequent with
the findings and decide whether or not to recommend an image- central and medial quadrant cancers. Clinical or pathologic evi-
guided or open biopsy to be certain that the target lesion has dence of metastatic spread to supraclavicular lymph nodes is
been adequately sampled for diagnosis. no longer considered stage IV disease, but routine scalene or
supraclavicular lymph node biopsy is not indicated.
Biomarkers
BREAST CANCER STAGING AND BIOMARKERS Breast cancer biomarkers are of several types. Risk factor
Breast Cancer Staging biomarkers are those associated with increased cancer risk.164-
The clinical stage of breast cancer is determined primarily 168
These include familial clustering and inherited germline
through physical examination of the skin, breast tissue, and abnormalities, proliferative breast disease with atypia, and
regional lymph nodes (axillary, supraclavicular, and internal mammographic density. Exposure biomarkers are a subset of
mammary).158 However, clinical determination of axillary lymph risk factors that include measures of carcinogen exposure such
node metastases has an accuracy of only 33%. Ultrasound (US) as DNA adducts. Surrogate endpoint biomarkers are biologic
is more sensitive than physical examination alone in determining alterations in tissue that occur between cancer initiation and
axillary lymph node involvement during preliminary staging of development. These biomarkers are used as endpoints in short-
breast carcinoma. FNA or core biopsy of sonographically inde- term chemoprevention trials and include histologic changes,
terminate or suspicious lymph nodes can provide a more defini- indices of proliferation, and genetic alterations leading to
tive diagnosis than US alone.153,159 Pathologic stage combines cancer. Prognostic biomarkers provide information regarding
576 Table 17-10
TNM staging system for breast cancer
Primary tumor (T)
The T classification of the primary tumor is the same regardless of whether it is based on clinical or pathologic criteria, or both. Size
should be measured to the nearest millimeter. If the tumor size is slightly less than or greater than a cutoff for a given T classification,
it is recommended that the size be rounded to the millimeter reading that is closest to the cutoff. For example, a reported size of
1.1 mm is reported as 1 mm, or a size of 2.01 cm is reported as 2.0 cm. Designation should be made with the subscript “c” or “p”
modifier to indicate whether the T classification was determined by clinical (physical examination or radiologic) or pathologic
measurements, respectively. In general, pathologic determination should take precedence over clinical determination of T size.
TX Primary tumor cannot be assessed
PART II

T0 No evidence of primary tumor


Tis (DCIS)* Ductal carcinoma in situ
Tis (Paget) Paget disease of the nipple NOT associated with invasive carcinoma and/or carcinoma in situ (DCIS) in the
underlying breast parenchyma. Carcinomas in the breast parenchyma associated with Paget disease are
categorized based on the size and characteristics of the parenchymal disease, although the presence of Paget
SPECIFIC CONSIDERATIONS

disease should still be noted.


T1 Tumor ≤20 mm in greatest dimension
T1mi Tumor ≤1 mm in greatest dimension
T1a Tumor >1 mm but ≤5 mm in greatest dimension (round any measurement >l.0–1.9 mm to 2 mm).
T1b Tumor >5 mim but ≤10 mm in greatest dimension
T1c Tumor >10 mm but ≤20 mm in greatest dimension
T2 Tumor >20 mm but ≤50 mm in greatest dimension
T3 Tumor >50 mm in greatest dimension
T4 Tumor of any size with direct extension to the chest wall and/or to the skin (ulceration or macroscopic nodules);
invasion of the dermis alone does not qualify as T4
T4a Extension to the chest wall; invasion or adherence to pectoralis muscle in the absence of invasion of chest wall
structures does not qualify as T4
T4b Ulceration and/or ipsilateral macroscopic satellite nodules and/or edema (including peau d’orange) of the skin
that does not meet the criteria for inflammatory carcinoma
T4c Both T4a and T4b are present
T4d Inflammatory carcinoma (see section “Rules for Classification”)
*Note: Lobular carcinoma in situ (LCIS) is a benign entity and is removed from TNM staging in the AJCC Cancer Staging Manual, 8th edition.
Regional lymph nodes—Clinical (N)
cNX* Regional lymph nodes cannot be assessed (e.g., previously removed)
cN0 No regional lymph node metastases (by imaging or clinical examination)
cN1 Metastases to movable ipsilateral Level I, II axillary lymph node(s)
cN1mi** Micrometastases (approximately 200 cells, larger than 0.2 mm, but none larger than 2.0 mm)
cN2 Metastases in ipsilateral Level I, II axillary lymph nodes that are clinically fixed or matted;
or in ipsilateral internal mammary nodes in the absence of axillary lymph node metastases
cN2a Metastases in ipsilateral Level I, II axillary lymph nodes fixed to one another (matted) or to other structures
cN2b Metastases only in ipsilateral internal mammary nodes in the absence of axillary lymph node metastases
cN3 Metastases in ipsilateral infraclavicular (Level III axillary) lymph node(s) with or without Level I, II axillary
lymph node involvement;
or in ipsilateral internal mammary lymph node(s) with Level I, II axillary lymph node metastases;
or metastases in ipsilateral supraclavicular lymph node(s) with or without axillary or internal mammary lymph
cN3a node involvement
cN3b Metastases in ipsilateral infraclavicular lymph node(s)
cN3c Metastases in ipsilateral internal mammary lymph node(s) and axillary lymph node(s)
Metastases in ipsilateral supraclavicular lymph node(s)
Note: (sn) and (f) suffixes should be added to the N category to denote confirmation of metastasis by sentinel node biopsy or fine needle aspiration/core
needle biopsy respectively.
*the cNX category is used sparingly in cases where regional lymph nodes have previously been surgically removed or where there is no documentation of
physical examination of the axilla.
**cN1mi is rarely used but may be appropriate in cases where sentinel node biopsy is performed before tumor resection, most likely to occur in cases
treated with neoadjuvant therapy.

(Continued)
Table 17-10 577

TNM staging system for breast cancer (Continued)


Regional lymph nodes—Pathologic (pN)
pNX Regional lymph nodes cannot be assessed (e.g., not removed for pathological study or previously removed)
pN0 No regional lymph node metastasis identified or ITCs only
pN0(i+) ITCs only (malignant cell clusters no larger than 0.2 mm) in regional lymph node(s)
pN0(mol+) Positive molecular findings by reverse transcriptase polymerase chain reaction (RT-PCR); no ITCs detected
pN1 Micrometastases; or metastases in 1–3 axillary lymph nodes; and/or clinically negative internal mammary nodes
with micrometastases or macrometastases by sentinel lymph node biopsy

CHAPTER 17 THE BREAST


pN1mi Micrometastases (approximately 200 cells, larger than 0.2 mm, but none larger than 2.0 mm)
pN1a Metastases in 1–3 axillary lymph nodes, at least one metastasis larger than 2.0 mm
pN1b Metastases in ipsilateral internal mammary sentinel nodes, excluding ITCs
pN1c pN1a and pNlb combined
pN2 Metastases in 4–9 axillary lymph nodes; or positive ipsilateral internal mammary lymph nodes by imaging in
the absence of axillary lymph node metastases
pN2a Metastases in 4–9 axillary lymph nodes (at least one tumor deposit larger than 2.0 mm)
pN2b Metastases in clinically detected internal mammary lymph nodes with or without microscopic confirmation;
with pathologically negative axillary nodes
pN3 Metastases in 10 or more axillary lymph nodes;
or in infraclavicular (Level III axillary) lymph nodes;
or positive ipsilateral internal mammary lymph nodes by imaging in the presence of one or more positive
Level I, II axillary lymph nodes;
or in more than three axillary lymph nodes and micrometastases or macrometastases by sentinel lymph node
biopsy in clinically negative ipsilateral internal mammary lymph nodes;
pN3a or in ipsilateral supraclavicular lymph nodes
Metastases in 10 or more axillary lymph nodes (at least one tumor deposit larger than 2.0 mm);
pN3b or metastases to the infraclavicular (Level III axillary lymph) nodes
pNla or pN2a in the presence of cN2b (positive internal mammary nodes by imaging);
pN3c or pN2a in the presence of pNlb
Metastases in ipsilateral supraclavicular lymph nodes
Note: (sn) and (f) suffixes should be added to the N category to denote confirmation of metastasis by sentinel node biopsy or FNA/core needle biopsy
respectively, with NO further resection of nodes.
Distant metastasis (M)
M0 No clinical or radiographic evidence of distant metastases*
cM0(i+) No clinical or radiographic evidence of distant metastases in the presence of tumor cells or deposits no larger
than 0.2 mm detected microscopically or by molecular techniques in circulating blood, bone marrow, or other
nonregional nodal tissue in a patient without symptoms or signs of metastases
cM1 Distant metastases detected by clinical and radiographic means
pM1 Any histologically proven metastases in distant organs; or if in non-regional nodes, metastases greater than
0.2 mm
Used with the permission of the American College of Surgeons. Amin MB, Edge SB, Greene FL, et al. (Eds.) AJCC Cancer Staging Manual, 8th Ed. Springer
New York, 2017.

cancer outcome irrespective of therapy, whereas predictive bio- indices of apoptosis and apoptosis modulators such as bcl-2
markers provide information regarding response to therapy.169 and the bax:bcl-2 ratio.
Candidate prognostic and predictive biomarkers and biologic
targets for breast cancer include (a) the steroid hormone recep- Steroid Hormone Receptor Pathway. Hormones play an
tor pathway; (b) growth factors and growth factor receptors important role in the development and progression of breast
such as human epidermal growth factor receptor 2 (HER2)/ cancer. Estrogens, estrogen metabolites, and other steroid hor-
neu, epidermal growth factor receptor (EGFR), transforming mones such as progesterone all have been shown to have an
growth factor, platelet-derived growth factor, and the insulin- effect. Breast cancer risk is related to estrogen exposure over
like growth factor family; (c) indices of proliferation such as time. In postmenopausal women, hormone replacement therapy
proliferating cell nuclear antigen (PCNA) and Ki-67; (d) indi- consisting of estrogen plus progesterone increases the risk of
ces of angiogenesis such as vascular endothelial growth factor breast cancer by 26% compared to placebo.70 Patients with hor-
(VEGF) and the angiogenesis index; (e) the mammalian target mone receptor-positive tumors survive two to three times longer
of rapamycin (mTOR) signaling pathway; (f) tumor-suppressor after a diagnosis of metastatic disease than do patients with hor-
genes such as p53; (g) the cell cycle, cyclins, and cyclin-depen- mone receptor-negative tumors. Patients with tumors negative
dent kinases; (h) the proteasome; (i) the COX-2 enzyme; (j) the for both estrogen receptors and progesterone receptors are not
peroxisome proliferator-activated receptors (PPARs); and (k) considered candidates for hormonal therapy. Tumors positive
578 Table 17-11
cancer specimens. The tumor hormone receptor status should
be ascertained for both premenopausal and postmenopausal
TNM stage groupings patients to identify patients who are most likely to benefit from
endocrine therapy.
Then the stage
When T is... And N is... And M is... group is... Growth Factor Receptors and Growth Factors. Overexpres-
sion of EGFR in breast cancer correlates with estrogen recep-
Tis N0 M0 0 tor–negative status and with p53 overexpression.170-172 Similarly,
T1 N0 M0 IA increased immunohistochemical membrane staining for the
T0 N1mi M0 IB HER2 growth factor receptor in breast cancer is associated with
mutated TP53, Ki67 overexpression, and estrogen receptor–
T1 N1mi M0 IB
negative status. HER2 is a member of the ErbB family of
PART II

T0 N1 M0 IIA growth factor receptors in which ligand binding results in recep-


T1 N1 M0 IIA tor homodimerization and tyrosine phosphorylation by tyrosine
T2 N0 M0 IIA kinase domains within the receptor. Tyrosine phosphorylation
T2 N1 M0 IIB is followed by signal transduction, which results in changes in
cell behavior. An important property of this family of receptors
T3 N0 M0 IIB
SPECIFIC CONSIDERATIONS

is that ligand binding to one receptor type also may result in


T0 N2 M0 IIIA heterodimerization between two different receptor types that are
T1 N2 M0 IIIA coexpressed; this leads to transphosphorylation and transactiva-
T2 N2 M0 IIIA tion of both receptors in the complex (transmodulation). In this
context, the lack of a specific ligand for the HER2/neu receptor
T3 N1 M0 IIIA
suggests that HER2/neu may function solely as a co-receptor,
T3 N2 M0 IIIA modulating signaling by other EGFR family members. HER2/
T4 N0 M0 IIIB neu is both an important prognostic factor and a predictive fac-
T4 N1 M0 IIIB tor in breast cancer.173 When overexpressed in breast cancer,
T4 N2 M0 IIIB
HER2/neu promotes enhanced growth and proliferation, and
increases invasive and metastatic capabilities. Clinical studies
Any T N3 M0 IIIC have shown that patients with HER2/neu–overexpressing breast
Any T Any N M1 IV cancer have poorly differentiated tumors with high prolifera-
Notes: tion rates, positive lymph nodes, decreased hormone receptor
1. T1 includes Tl mi. expression, and an increased risk of recurrence and death due
2. T0 and T1 tumors with nodal micrometastases (N1mi) are staged as to breast cancer.173-177 Routine testing of the primary tumor
Stage IB.
specimen for HER2/neu expression should be performed on
3. T2, T3, and T4 tumors with nodal micrometastases (N1mi) are staged
using the N1 category. all invasive breast cancers. This can be done with immunohis-
4. M0 includes M0(i+). tochemical analysis to evaluate for overexpression of the cell-
5. The designation pM0 is not valid; any M0 is clinical. surface receptor at the protein level or by using fluorescence
6. If a patient presents with M1 disease prior to neoadjuvant systemic in situ hybridization to evaluate for gene amplification. While
therapy, the stage is Stage IV and remains Stage IV regardless of HER2/ERBB2 activation can also be assessed based on mRNA
response to neoadjuvant therapy. expression and reverse transcription polymerase chain reaction
7. Stage designation may be changed if postsurgical imaging studies
(RT-PCR) (Oncotype Dx, Genomic Health), this approach is not
reveal the presence of distant metastases, provided the studies are per-
formed within 4 months of diagnosis in the absence of disease progres- recommended for clinical decision-making because of the high
sion, and provided the patient has not received neoadjuvant therapy. false negative rate.178 Patients whose tumors show HER2 ampli-
8. Staging following neoadjuvant therapy is denoted with a “yc” or “yp” fication or HER2/neu protein overexpression are candidates for
prefix to the T and N classification. There is no anatomic stage group anti-HER2/neu therapy. Trastuzumab (Herceptin) is a recombi-
assigned if there is a complete pathological response (pCR) to neoad- nant humanized monoclonal antibody directed against HER2.
juvant therapy, for example, ypT0ypN0cM0.
Randomized clinical trials have demonstrated that single-agent
Used with the permission of the American College of Surgeons. Amin MB,
Edge SB, Greene FL, et al. (Eds.) AJCC Cancer Staging Manual, 8th Ed.
trastuzumab therapy is well tolerated and active in the treatment
Springer New York, 2017. of women with HER2/neu–overexpressing metastatic breast
cancer.179 Subsequent adjuvant trials demonstrated that trastu-
zumab also was highly effective in the treatment of women with
early-stage breast cancer when used in combination with che-
for estrogen or progesterone receptors have a higher response
motherapy.180-182 Patients who received trastuzumab in combina-
rate to endocrine therapy than tumors that do not express estro-
tion with chemotherapy had between a 40% and 50% reduction
gen or progesterone receptors. The determination of estrogen
in the risk of breast cancer recurrence and approximately a one-
and progesterone receptor status used to require biochemical
third reduction in breast cancer mortality compared with those
evaluation of fresh tumor tissue. Today, however, estrogen and
who received chemotherapy alone.181,183-185
progesterone receptor status can be measured in archived tis-
sue using immunohistochemical techniques. Hormone receptor Indices of Proliferation. PCNA is a nuclear protein asso-
status also can be measured in specimens obtained with fine- ciated with a DNA polymerase whose expression increases in
needle aspiration biopsy or core-needle biopsy, and this can help phase G1 of the cell cycle, reaches its maximum at the G1/S inter-
guide treatment planning. Testing for estrogen and progesterone face, and then decreases through G2.186-189 Immunohistochemical
receptors should be performed on all primary invasive breast staining for PCNA outlines the proliferating compartments in
breast tissue. Good correlation is noted between PCNA expres- expression correlates with axillary lymph node metastases, a 579
sion and (a) cell-cycle distributions seen on flow cytometry poor response to chemotherapy, and decreased overall survival.
based on DNA content, and (b) uptake of bromodeoxyuridine The remaining biomarkers and biologic targets listed ear-
and the proliferation-associated Ki67 antigen. Individual prolif- lier are still in preclinical testing, and clinical trials are evaluat-
eration markers are associated with slightly different phases of ing their importance in breast cancer for both prognostic and
the cell cycle and are not equivalent. PCNA and Ki67 expression predictive purposes.
are positively correlated with p53 overexpression, high S-phase
fraction, aneuploidy, high mitotic index, and high histologic Coexpression of Biomarkers. Selection of optimal therapy
grade in human breast cancer specimens, and are negatively cor- for breast cancer requires both an accurate assessment of prog-
related with estrogen receptor content. Ki67 was included with nosis and an accurate prediction of response to therapy. The
breast cancer markers that are most important in determining

CHAPTER 17 THE BREAST


three other widely measured breast cancer markers (ER, PR,
and HER2) into a panel of four immunohistochemical makers therapy are estrogen receptor, progesterone receptor, and HER2/
(IHC4), which together provided similar prognostic informa- neu. Clinicians evaluate clinical and pathologic staging and the
tion to that in the 21 Gene Recurrence Score (Oncotype DX, expression of estrogen receptor, progesterone receptor, and
Genomic Health).190 While there has been significant interest in HER2/neu in the primary tumor to assess prognosis and assign
using Ki67 as a biomarker, and while the IHC4 panel would be therapy. Adjuvant! Online (http://www.adjuvantonline.com) is
much less expensive than the 21 Gene Recurrence Score, there one of a number of programs available to clinicians that incor-
remain issues regarding reproducibility across laboratories. porates clinical and pathologic factors for an individual patient
and calculates risk of recurrence and death due to breast cancer
Indices of Angiogenesis. Angiogenesis is necessary for the and then provides an assessment of the reduction in risk of
growth and invasiveness of breast cancer and promotes cancer recurrence that would be expected with the use of combination
progression through several different mechanisms, including chemotherapy, endocrine therapy, or both of these. Adjuvant!
delivery of oxygen and nutrients and the secretion of growth- Online was developed using information from the SEER data-
promoting cytokines by endothelial cells.191,192 VEGF induces base, the EBCTCG overview analyses, and results from other
its effect by binding to transmembrane tyrosine kinase recep- individual published trials.198 The website is updated and modi-
tors. Overexpression of VEGF in invasive breast cancer is cor- fied as new information becomes available. Clinicopathologic
related with increased microvessel density and recurrence in factors are used to separate breast cancer patients into broad
node-negative breast cancer. An angiogenesis index has been prognostic groups, and treatment decisions are made on this
developed in which microvessel density (CD31 expression) basis (Table 17-12). Other indices and programs that are vali-
is combined with expression of thrombospondin (a negative dated and used include the Nottingham Prognostic Index, and
modulator of angiogenesis) and p53 expression. Both VEGF PREDICT.199-201 When an approach, which combines prognostic
expression and the angiogenesis index may have prognostic factors is used, up to 70% of early breast cancer patients receive
and predictive significance in breast cancer.193 Bevacizumab adjuvant chemotherapy that is either unnecessary or ineffective.
(a monoclonal antibody to VEGF) was approved by the FDA As described earlier, a wide variety of biomarkers have been
for use in metastatic breast cancer in combination with pacli- shown to individually predict prognosis and response to therapy,
taxel chemotherapy. This approval was based on results from a but they do not improve the accuracy of either the assessment of
phase 3 trial by the Eastern Cooperative Oncology Group. The prognosis or the prediction of response to therapy.
group’s E2100 trial showed that when bevacizumab was added As knowledge regarding cellular, biochemical, and molec-
to paclitaxel chemotherapy, median progression-free survival ular biomarkers for breast cancer have improved, prognostic
increased to 11.3 months from the 5.8 months seen in patients indices have been developed that combine the predictive power
who received paclitaxel alone.194 The results were not repro-
duced in other trials, and the indication for the drug was revoked
by the FDA in 2011. Table 17-12
Indices of Apoptosis. Alterations in programmed cell death Traditional prognostic and predictive factors for
(apoptosis), which may be triggered by p53-dependent or invasive breast cancer
p53-independent factors, may be important prognostic and pre-
dictive biomarkers in breast cancer.195-197 Bcl-2 family proteins TUMOR FACTORS HOST FACTORS
appear to regulate a step in the evolutionarily conserved pathway
Nodal status Age
for apoptosis, with some members functioning as inhibitors of
apoptosis and others as promoters of apoptosis. Bcl-2 is the only Tumor size Menopausal status
oncogene that acts by inhibiting apoptosis rather than by directly Histologic/nuclear grade Family history
increasing cellular proliferation. The death-signal protein bax Lymphatic/vascular invasion Previous breast cancer
is induced by genotoxic stress and growth factor deprivation
Pathologic stage Immunosuppression
in the presence of wild-type (normal) p53 and/or AP-1/fos.
The bax to bcl-2 ratio and the resulting formation of either bax- Hormone receptor status Nutrition
baxhomodimers, which stimulate apoptosis, or bax–bcl-2 het- DNA content (ploidy, S-phase Prior chemotherapy
erodimers, which inhibit apoptosis, represent an intracellular fraction)
regulatory mechanism with prognostic and predictive implica- Extent of intraductal component Prior radiation therapy
tions. In breast cancer, overexpression of bcl-2 and a decrease
HER2/neu expression  
in the bax to bcl-2 ratio correlate with high histologic grade,
the presence of axillary lymph node metastases, and reduced Modified with permission from Ellis N: Inherited Cancer Syndromes.
disease-free and overall survival rates. Similarly, decreased bax New York, NY: Springer-Verlag; 2004.
580 of several individual biomarkers with the relevant clinicopatho-
Table 17-13
logic factors.
Recent technological advances have enabled implemen- Diagnostic studies for breast cancer patients
tation of high throughput gene expression assays in clinical
practice.202 These assays enable detailed stratification of breast   CANCER STAGE
cancer patients for assessment of prognosis and for predic-
tion of response to therapy. The Oncotype DX is a 21-gene   0 I II III IV
RT-PCR–based assay that has been approved for use in newly History & physical X X X X X
diagnosed patients with node-negative, ER-positive breast Complete blood count, platelet     X X X
cancer.203 A recurrence score is generated, and those patients count
with high recurrence scores are likely to benefit from che-
Liver function tests and alkaline     X X X
motherapy, whereas those with low recurrence scores benefit
PART II

phosphatase level
most from endocrine therapy and may not require chemother-
apy. Results from the Trial Assessing Individualized Options Chest radiograph     X X X
for Treatment for breast cancer (TAILORx), designed to pro- Bilateral diagnostic mammograms, X X X X X
spectively validate the use of 21-gene expression assay, have ultrasound as indicated
shown that patients with low recurrence score (0 to 10) have a
SPECIFIC CONSIDERATIONS

Hormone receptor status X X X X X


low rate of local-regional and distant recurrence (98.7%) and
very good overall survival at 5 years (98%) with endocrine HER2/neu expression   X X X X
therapy alone without chemotherapy.204 This study has ran- Bone scan       X X
domly assigned patients with an intermediate recurrence score Abdominal (without or without       X X
(11 to 25) to endocrine therapy alone or to chemotherapy fol- pelvis) computed tomographic
lowed by endocrine therapy. scan or ultrasound or magnetic
Additionally, retrospective analysis has shown that resonance imaging
the 21-gene recurrence score can be used in postmenopausal
Abdominal imaging and bone scanning are indicated for evaluation of
patients with ER-positive tumors and 1 to 3 involved axillary symptoms or abnormal laboratory test results at any presenting stage.
lymph nodes to predict the benefit of chemotherapy in addition Data from NCCN Practice Guidelines in Oncology. Fort Washington,
to endocrine therapy.205 Knowledge of the recurrence score has PA: National Comprehensive Cancer Network, 2006.
been shown to alter treatment recommendations by oncologists,
and patients likewise change their decision to undergo treatment
based on the risk of recurrence.206 The MammaPrint assay uses studies are performed based on the initial stage as presented in
a 70-gene expression profile to assess the risk of distant metas- Table 17-13. Before therapy is initiated, the patient and the sur-
tasis. Mammaprint is FDA approved for use in stage-1 or stage- geon must share a clear perspective on the planned course of
2, node negative, ER-positive or ER-negative breast cancers to treatment. Before initiating local therapy, the surgeon should
identify patients with high or low risk of recurrence. Although determine the clinical stage, histologic characteristics, and
fresh tissue was initially required to perform the assay, it has 8 appropriate biomarker levels.
since been adapted for use in paraffin-embedded tissue sam-
ples. The prospective RASTER study reported that patients In Situ Breast Cancer (Stage 0)
classified as low risk based on MammaPrint had a 97% distant Both LCIS and DCIS may be difficult to distinguish from
recurrence-free interval at five years.207 Results of the prospec- atypical hyperplasia or from cancers with early invasion.60,209-214
tive MINDACT (MicroarrayInNode negative and 1–3 positive Expert pathologic review is required in all cases. Bilateral
lymph node Disease may Avoid ChemoTherapy) trial were mammography is performed to determine the extent of the in
recently reported.208 The study was designed to assess whether situ cancer and to exclude a second cancer. Because LCIS is
the 70-gene expression assay would help avoid chemotherapy in considered a marker for increased risk rather than an inevitable
patients who are considered clinically high risk but categorized precursor of invasive disease, the current treatment options for
as low genomic risk based on the assay. A 5-year rate of distant LCIS include observation, chemoprevention, and bilateral total
metastasis-free survival of more than 92% was identified as the mastectomy. The goal of treatment is to prevent or detect at
cutoff for the benefit of chemotherapy. At 5 years, the rate of an early stage the invasive cancer that subsequently develops
survival without distant metastasis in patients with high clinical in 25% to 35% of these women. There is no benefit to excis-
risk and low genomic risk was 94.7%, meeting the criteria for ing LCIS because the disease diffusely involves both breasts in
noninferiority. However, the rate of disease-free survival and many cases and the risk of developing invasive cancer is equal
overall survival was higher with chemotherapy in the intention for both breasts. The use of tamoxifen as a risk-reduction strat-
to treat population. egy should be considered in women with a diagnosis of LCIS.
Women with DCIS and evidence of extensive disease
(>4 cm of disease or disease in more than one quadrant) usu-
OVERVIEW OF BREAST CANCER THERAPY ally require mastectomy (Fig. 17-29). For women with lim-
Before diagnostic biopsy, the surgeon must consider the possi- ited disease, lumpectomy and radiation therapy are generally
bility that a suspicious mass or mammographic finding may be recommended. For nonpalpable DCIS, needle localization or
a breast cancer. Once a diagnosis of breast cancer is made, the other image-guided techniques are used to guide the surgical
type of therapy offered to a breast cancer patient is determined resection. Specimen mammography is performed to ensure
by the stage of the disease, the biologic subtype, and the general that all visible evidence of cancer is excised. Adjuvant tamoxi-
health status of the individual. Laboratory tests and imaging fen therapy is considered for DCIS patients with ER-positive
disease. The gold standard against which breast conservation 581
therapy for DCIS is evaluated is mastectomy. Women treated
with mastectomy have local recurrence and mortality rates of
<2%. There is no randomized trial comparing mastectomy vs.
breast conserving surgery, and none of the randomized trials
of breast-conserving surgery with or without radiotherapy for
DCIS were powered to show a difference in mortality. Women
treated with lumpectomy and adjuvant radiation therapy in the
initial clinical trials were noted to have a local recurrence rate
that is increased compared to mastectomy. About 45% of these

CHAPTER 17 THE BREAST


recurrences will be invasive cancer when radiation therapy is
not used. The B-17 trial was conducted by the NSABP to assess
the need for radiation in patients treated with breast conserv-
ing surgery for DCIS.215 Patients were randomly assigned to
lumpectomy with radiation or lumpectomy alone, and after
a mean follow-up time of 90 months rates of both ipsilateral
noninvasive and invasive recurrences were significantly lower
in patients who received radiation. However, in the B-17 trial
the margins were not prospectively assessed, and it is estimated
that up to half of the patients may have had tumor at the mar-
gin of resection. The benefit of the addition of radiation over
breast-conserving surgery alone for DCIS has also been dem-
onstrated in several other randomized trials where margins were
prospectively assessed including the European Organization for
Research and Treatment of Cancer (EORTC) protocol 10853;
the United Kingdom, Australia, New Zealand DCIS Trial;
and the Swedish Trial.209,216-218 In 2016, the Society of Surgi-
A
cal Oncology (SSO), American Society for Radiation Oncol-
ogy (ASTRO), and the American Society of Clinical Oncology
(ASCO) established consensus guidelines on margins for
patients with DCIS undergoing breast-conserving surgery.219
Based on a multidisciplinary consensus panel using a meta-
analysis of margin width and ipsilateral breast tumor recur-
rence, a 2-mm margin was determined as adequate width for
DCIS for patients undergoing breast-conserving surgery with
whole-breast radiation therapy.219
Despite the data from randomized trials showing a benefit
in all patient subgroups with the addition of radiation in DCIS,
there has been an interest in trying to define a subset where
radiation could be avoided in order to minimize the cost and
inconvenience associated with radiation. In addition, there have
been several studies published where patients were treated with
excision alone and never developed invasive breast cancer even
at 25 years of follow-up. Silverstein and colleagues were pro-
ponents of avoiding radiation therapy in selected patients with
DCIS who have widely negative margins after surgery.213 They
reported that when greater than 10-mm margins were achieved,
there was no additional benefit from radiation therapy. When
margins were between 1 and 10 mm, there was a relative risk
of local recurrence of 1.49, compared to 2.54 for those with
margins less than 1 mm. These data suggested that appropri-
ately selected patients with DCIS might not require postopera-
tive radiation therapy.
The Eastern Cooperative Oncology Group (ECOG) initi-
B ated a prospective registry trial (ECOG 5194) to identify those
Figure 17-29. Extensive DCIS seen on mammography. A. Exten- patients who could safely undergo breast-conserving surgery
sive calcifications are seen throughout the breast on this cranial without radiation.222 Eligible patients were those with low or
caudal view. B. Magnification view of calcifications. Due to the intermediate grade DCIS measuring 2.5 cm or less who had
extent of the disease the patient is not a good candidate for breast negative margins of at least 3 mm and those with high-grade
conserving surgery. (Used with permission from Dr. Anne Turnbull, DCIS who had tumors measuring 1 cm or less with a negative
Consultant Radiologist/Director of Breast Screening, Royal Derby margin of at least 3 mm. At a median follow-up of 6.2 years,
Hospital, Derby, UK.) patients with low or intermediate grade DCIS had an in-breast
582 recurrence rate of 6.1% while those with high-grade DCIS these patients, tamoxifen could be viewed as treating what, by
had a recurrence rate of 15.3%. Approximately 4% of patients the current standard, would be viewed as inadequate local exci-
developed a contralateral breast cancer during follow-up in both sion of the primary tumor.
the low/intermediate and high-grade groups. This study identi-
fied an acceptable recurrence rate for those patients with low
or intermediate grade DCIS treated with excision alone with a Early Invasive Breast Cancer
margin of at least 3 mm. In contrast, patients with high-grade (Stage I, IIA, or IIB)
DCIS had an unacceptably high local recurrence rate. There have been six prospective randomized trials comparing
The Radiation Therapy Oncology Group (RTOG) initi- breast-conserving surgery to mastectomy in early stage breast
ated the 9804 trial for patients with “good risk” DCIS and cancer, and all have shown equivalent survival rates regardless
randomized them to lumpectomy vs. lumpectomy with whole of the surgical treatment type. One caveat, however, is that the
breast irradiation. Eligible patients were those with unicentric, majority of studies had a restriction of tumor size; most were
PART II

low or intermediate grade DCIS measuring 2.5 cm or less with either 2 cm or 2.5 cm, while the NSABP B-06 trial was 4 cm, and
a margin of 3 mm or greater. The trial was closed early due the NCI trial was up to 5 cm. NSABP B-06, which is the largest
to slow accrual; however, the results for 585 patients were of all the breast conservation trials, compared total mastectomy
recently reported with a median follow-up of 6.46 years.223,224 to lumpectomy with or without radiation therapy in the treatment
The local recurrence rate at 5 years was 0.4% for patients ran- of women with stages I and II breast cancer.227-233 After 5- and
SPECIFIC CONSIDERATIONS

domized to receive radiation and 3.2% for those who did not 8-year follow-up periods, the disease-free (DFS), distant dis-
receive radiation. ease-free, and overall survival (OS) rates for lumpectomy with
Solin et al utilized samples from the ECOG 5194 trial to or without radiation therapy were similar to those observed after
develop a quantitative multigene RT-PCR assay for predict- total mastectomy. However, the incidence of ipsilateral breast
ing recurrence risk in patients with DCIS treated with surgery cancer recurrence was higher in the group not receiving radia-
alone.201 They were able to define low, intermediate, and high tion therapy. These findings supported the use of lumpectomy
risk groups using a DCIS Score. The DCIS Score was able to and radiation therapy in the treatment of stages I and II breast
quantify the risk of recurrence in the breast for both DCIS and cancer and this has since become the preferred method of treat-
invasive events. This tool has recently been evaluated in another ment for women with early stage breast cancer who have uni-
dataset and appears to be a promising tool for clinical use.225 focal disease and who are not known BRCA mutation carriers.
When selecting therapy for patients with DCIS, one must con- Reanalysis of the B-06 study results was undertaken after
sider clinical and pathologic factors, including tumor size, grade, 20 years of follow-up and confirmed that there was no differ-
mammographic appearance, and patient preference. There is no ence in disease-free survival rates after total mastectomy or after
single correct surgical treatment, and many patients will require lumpectomy with or without adjuvant radiation therapy. The
extensive counseling in order to make a decision regarding sur- in-breast recurrence rate was substantially higher in the lumpec-
gical therapy. The role of axillary staging in patients with DCIS tomy alone group (39.2%) compared with the lumpectomy
is limited. One consideration is for patients undergoing mastec- plus adjuvant radiation therapy group (14.3%), confirming the
tomy. Since most lesions are currently diagnosed with needle importance of radiation therapy in the management of patients
core biopsy, there is about a 20% incidence of invasive breast with invasive disease. However, it should be noted that there
cancer on final pathologic assessment of the primary tumor. were several criteria in the B-06 study. There was a specific
Since it is not feasible to perform sentinel node dissection after lymphadenopathy exclusion criteria. Secondly, all patients ran-
mastectomy, most surgeons will recommend the use of sentinel domized to breast-conserving surgery had a frozen section, and
node dissection at the time of mastectomy for DCIS. if the margins were involved, they were converted to mastec-
Results from the NSABP B-24 trial reported a signifi- tomy but were included in the analysis as having had a breast-
cant reduction in local recurrence after 5 years of tamoxifen in conserving operation (on the basis of intention to treat). Finally,
women with ER-positive DCIS. Based on this finding, some in the breast-conserving group recurrences in the treated breast
guidelines have advocated that all patients (women with ER- were considered as a “nonevent.”
positive DCIS without contraindications to tamoxifen therapy) Data from all of the randomized trials where breast con-
should be offered tamoxifen following surgery and radiation servation was performed with or without radiation therapy have
therapy for a duration of 5 years. The B-24 trial revealed a sig- been examined by the EBCTCG.12 At 15 years of follow-up, the
nificant reduction in recurrence with adjuvant tamoxifen therapy absolute reduction in mortality with the use of radiation therapy
for patients with DCIS; however, the results were not initially after lumpectomy was 5.1% in node-negative patients and 7.1%
assessed based on ER status.226 There were 1804 women with in node-positive patients. These data support the concept that
DCIS randomized to lumpectomy and radiation with or without the addition of radiation not only improves local control but
tamoxifen. The rate of breast cancer events was significantly also has an impact on survival. Similar to DCIS, clinicians have
lower in those who received tamoxifen at a median follow-up of sought to identify subgroups of patients who may not benefit
74 months (8.2% vs. 13.4%, P = 0.0009). Subsequently, Allred from the addition of radiation therapy, particularly older patients
and colleagues evaluated 41% of patients with DCIS in the who may have a shorter life expectancy due to medical comor-
NSABP B-24 trial to determine the effect of tamoxifen based bidities. Randomized trials have shown that in selected patients
on ER status measured in the primary tumor.203 They found with small, ER-positive, low-grade tumors, lumpectomy alone
that 76% of women had DCIS that was ER-positive and these without radiation therapy may be appropriate.211,212 The Cancer
women had a greater reduction in ipsilateral breast tumor recur- and Leukemia Group B (CALGB) C9343 trial enrolled women
rence with tamoxifen than did patients with ER-negative DCIS over the age of 70 with T1N0 breast cancer and randomized
(11% vs. 5.2%, P <0.001). However, it should be noted that them to lumpectomy with or without radiation therapy. All
15% of patients in B-24 had tumor at the resection margins. For patients received adjuvant tamoxifen.233a At 5 years, although
there were fewer local recurrences with radiation (1% vs. 4%, are considered equivalent treatments for patients with stages I 583
P <0.001), there were no differences in DFS and OS. While and II breast cancer. Breast conservation is considered for all
long-term follow-up at 10 years showed fewer local recurrences patients because of the important cosmetic advantages and
with radiation (2% vs. 10%), there were no significant differ- equivalent survival outcomes; however, this approach is not
ences in time to distant metastasis, breast cancer–specific sur- advised in women who are known BRCA mutation carriers
vival, or OS between the two groups. A trial similar to CALGB due to the high lifetime risk for development of additional
C9343 was conducted in Canada where they enrolled women breast cancers. Relative contraindications to breast conserva-
age 50 years and older and randomized them to lumpectomy tion therapy include (a) prior radiation therapy to the breast or
with or without radiation. Mean age was 68 years, and 80% of chest wall, (b) persistently positive surgical margins after reex-
women had ER-positive tumors. Again, local recurrence rates cision, (c) multicentric disease, and (d) scleroderma or lupus

CHAPTER 17 THE BREAST


were lower in women who received radiation (0.6% vs. 7.7%, erythematosus.
P <0.001); however, at a median follow-up of 5.6 years, there For most patients with early-stage disease, reconstruc-
were no differences in DFS or OS. The PRIME-2 study enrolled tion can be performed immediately at the time of mastectomy.
women age 65 years or older with ER-positive, node-negative, Immediate reconstruction allows for skin-sparing, thus optimiz-
up to 3 cm breast cancers, who had undergone breast-conserving ing cosmetic outcomes. Skin-sparing mastectomy with immedi-
surgery and were candidates for adjuvant endocrine treatment. ate reconstruction has been popularized over the past decade as
They were assigned to receive whole-breast irradiation or no reports of low local-regional failure rates have been reported
treatment. After a median follow-up of 5 years, ipsilateral breast and reconstructive techniques have advanced. There is a grow-
tumor recurrence was 1.3% with radiation vs. 41% in those ing interest in the use of nipple-areolar sparing mastectomy with
assigned to no radiotherapy. However, no differences in distant reports suggesting the oncologic safety of this approach in early
metastases, contralateral breast cancers, or overall survival were stage breast cancer. Patients who are planned for postmastec-
noted between the groups.234 These studies suggest that radia- tomy radiation therapy may not be ideal candidates for nipple-
tion can be avoided in select older patients with ER-positive, sparing mastectomy because of the effects of radiation on the
early-stage breast cancer. preserved nipple. In addition to providing optimal cosmesis
Accelerated partial breast irradiation (APBI) is also an from preservation of the skin and/or the nipple-areolar complex,
option for carefully selected patients with DCIS and early-stage immediate reconstruction allows patients to wake up with a
breast cancer. Since the majority of recurrences after breast breast mound, which provides some psychological benefit for
conservation occur in or adjacent to the tumor bed, there has the patient. Immediate reconstruction is also more economical
been interest in limiting the radiation to the area of the primary as both the extirpative and reconstructive surgery are combined
tumor bed with a margin of normal tissue. APBI is delivered in in one operation.
an abbreviated fashion (twice daily for 5 days) and at a lower Immediate reconstruction can be performed using implants
total dose compared with the standard course of 5 to 6 weeks of or autologous tissue; tissue flaps commonly used include the
radiation (50 Gy with or without a boost) in the case of whole transverse rectus abdominis myocutaneous flap, deep inferior
breast irradiation. Proponents have suggested that this shortened epigastric perforator flap, and latissimus dorsi flap (with or
course of treatment may increase the feasibility of breast con- without an implant). If postmastectomy radiation therapy is
servation for some women and may improve radiation therapy needed, a tissue expander can be placed at the time of mastec-
compliance. The RTOG 04-13/NSABP B-39 trial is a random- tomy to save the shape of the breast and reduce the amount of
ized comparison of whole breast irradiation to APBI in women skin replacement needed at the time of definitive reconstruc-
with early stage breast cancer. The trial has completed accrual, tion. The expander can be deflated at the initiation of radiation
and it will likely be several years before data are mature to therapy to allow for irradiation of the chest wall and regional
report outcomes between the two radiation treatment strategies. nodal basins. Removal of the tissue expander and definitive
TARGIT is another study that randomized 3451 patients in 33 reconstruction, usually with autologous tissue, can proceed
centers in over 10 countries to intraoperative breast irradiation 6 months to 1 year after completion of radiation therapy.
(IORT) or external beam radiotherapy (EBRT). The prelimi- Axillary lymph node status has traditionally been an
nary results were reported in 2012: with a median follow-up of important determinant in staging and prognosis for women with
2.4 years, use of IORT had a recurrence rate of 3.3% vs. 1.3% early stage breast cancer. Historically, axillary lymph node dis-
with EBRT, a 2% increased recurrence risk.235,236 ASTRO section (ALND) was utilized for axillary staging and regional
developed guidelines for the use of APBI outside of clinical control by removing involved lymph nodes. Randomized tri-
trials based on data reported from published studies.237,238 The als evaluating immediate ALND over ALND performed in a
ASTRO guidelines describe patients “suitable” for APBI to delayed fashion once clinically palpable axillary disease became
include women age 60 years or older with a unifocal, T1, ER- evident have not shown any detriment in survival.9,239 With
positive tumor with no lymphovascular invasion and margins increased mammographic screening and detection of smaller,
of at least 2 mm. They describe a group where there is uncer- node-negative breast cancers, it became clear that routine use
tainty about the appropriateness of APBI (“cautionary” group) of ALND for axillary staging was not necessary in up to 75%
to include patients with invasive lobular histology, a tumor size percent of women with operable breast cancer presenting with
of 2.1 cm to 3 cm, ER-negative disease, focal lymphovascular a negative axilla at the time of screening. Lymphatic mapping
invasion, or margins less than 2 mm. Finally, a group felt to be and sentinel lymph node (SLN) dissection were initially devel-
“unsuitable” for APBI includes those with T3 or T4 disease, oped for assessment of patients with clinically node-negative
ER-negative disease, multifocality, multicentricity, extensive melanoma. Given the changing landscape of newly diagnosed
LVI, or positive margins. breast cancer patients with a clinically node-negative axilla, sur-
Currently, mastectomy with axillary staging and breast geons quickly began to explore the utility of SLN dissection as
conserving surgery with axillary staging and radiation therapy a replacement for ALND in axillary staging.
584 In the early 1990s, David Krag at the University of recurrence, neither immunohistochemical detection of disease
Vermont began performing SLN dissection with injection of a in the SLNs or the bone marrow was statistically significant
radioisotope in the primary tumor site and localizing the SLN on multivariable analysis with clinicopathologic and treatment
node with a handheld gamma probe.240 He was able to identify factors included. The investigators concluded that routine use
a SLN in 18 of 22 patients examined, and the SLN was posi- of immunohistochemistry to detect occult disease in SLNs is
tive in all 7 patients with positive lymph nodes. Giuliano and not warranted.
colleagues initiated a pilot study in 1991 to examine the use of The Z0011 trial was a companion study to Z0010 and was
SLN dissection using blue dye in patients with clinically nega- designed to study the role of completion ALND on survival in
tive nodes. They reported successful identification of a SLN women with positive SLNs. Patients were not eligible if they
in 114 (65.5%) of 174 patients, and in 109 (95.6%), the SLN received neoadjuvant chemotherapy or neoadjuvant hormonal
accurately predicted the status of the axillary nodes.241,242 These therapy or if their treatment plan included mastectomy, lumpec-
studies along with initial work by Doug Reintgen and Charles tomy without radiation, or lumpectomy with APBI. WBI was to
PART II

Cox at the Moffitt Cancer Center and Umberto Veronesi and be administered using standard tangential fields without specific
his colleagues at the European Institute of Oncology in Milan treatment of the axilla or additional fields targeting other nodal
led the way toward validation of the technique in large single basins. Patients with 1 or 2 positive SLNs were randomized to
institution and multicenter studies. completion ALND or no further surgery. Adjuvant systemic
Following validation of the technique of SLN dissection therapy recommendations were left to the treating clinicians.
SPECIFIC CONSIDERATIONS

for staging of the axilla by multiple centers, randomized tri- After median follow-up of 6.3 years, there was no difference
als were initiated in order to determine if SLN dissection could between patients randomized to ALND and those randomized
replace ALND in the contemporary management of breast cancer to no further surgery (SLN only) in terms of OS (91.9% and
patients. The ALMANAC trial randomized 1031 patients with 92.5%, respectively; P = 0.25) or DFS (82.2% and 83.8%,
primary operable breast cancer to SLN dissection vs. standard respectively; P = 0.14). The low local regional failure rates and
axillary surgery. The incidence of lymphedema and sensory similar survival outcomes were recently reported with 10-year
loss for the SLN group was significantly lower than with the follow-up.249,250
standard axillary treatment. At 12 months, drain usage, length The morbidity of SLN dissection alone vs. SLN dissec-
of hospital stay, and time to resumption of normal day-to-day tion with completion ALND has been reported by the ACOSOG
activities after surgery were also statistically significantly lower investigators.251,252 Immediate effects of SLN dissection in the
in the SLN group.221 Z0010 trial included wound infection in 1%, axillary seroma in
The NSABP B-32 trial compared clinically node-negative 7.1%, and axillary hematoma in 1.4%.251 At 6 months following
patients undergoing SLN dissection followed by ALND with surgery, axillary paresthesias were noted in 8.6% of patients,
patients undergoing SLN dissection with ALND only if a SLN decreased range of motion in the upper extremity was reported
was positive for metastatic disease.243 A total of 5611 patients in 3.8%, and 6.9% of patients had a change in the arm circum-
were randomized with a SLN identification rate of 97% and ference of >2 cm on the ipsilateral side, which was reported
a false-negative rate of 9.7%. A total of 26% of these clini- as lymphedema. Younger patients were more likely to report
cally node-negative patients had a positive SLN. Over 60% of paresthesias, whereas increasing age and body mass index were
patients with positive SLNs had no additional positive lymph more predictive of lymphedema. When adverse surgical effects
nodes within the ALND specimen. The B-32 trial and other were examined in the Z0011 trial, patients undergoing SLN
randomized trials demonstrated no difference in DFS, OS, and dissection with ALND had more wound infections, seromas,
local-regional recurrence rates between patients with negative and paresthesias than those women undergoing SLN dissec-
SLNs who had SLN dissection alone compared with those who tion alone. Lymphedema at 1 year after surgery was reported
underwent ALND.244,245 Most important, patients who had SLN by 13% in the SLN plus ALND group but only 2% in the SLN
dissection alone were found to have decreased morbidity (arm dissection alone group. Arm circumference measurements were
swelling and range of motion) and improved quality of life vs. greater at 1 year in patients undergoing SLN dissection plus
patients who underwent ALND.245,246 ALND, but the difference between study groups was not statisti-
The American College of Surgeons Oncology Group cally significant.252 This supports the results published from the
(ACOSOG) initiated the Z0010 and Z0011 trials in order to ALMANAC trial.
evaluate the incidence and prognostic significance of occult Prior to the publication of ACOSOG Z0011, completion
metastases identified in the bone marrow and SLNs (Z0010) ALND was standard of care for patients with positive SLNs.
of early-stage clinically node-negative patients and to evaluate Since the reporting of ACOSOG Z0011, the National Com-
the utility of ALND in patients with clinical T1-2, N0 breast prehensive Cancer Network (NCCN) guidelines now state that
cancer with 1 or 2 positive SLNs for patients treated with there was no OS difference for patients with 1 or 2 positive
breast-conserving surgery and whole breast irradiation (WBI) SLNs treated with breast-conserving surgery who underwent
(Z0011).247,248 completion ALND vs. those who had no further axillary sur-
The Z0010 study enrolled 5539 patients with clinical T1-2 gery. In addition, the American Society of Breast Surgeons
breast cancer planned for breast conserving surgery and WBI.247 issued a consensus statement supporting omission of ALND for
Of these patients, 24% proved to have positive SLNs based on patients who meet Z0011 criteria.253 The results of ACOSOG
standard pathologic assessment, and of the negative SLNs sub- Z0011 have revolutionized management of the axilla and
jected to immunohistochemical staining for cytokeratin, 10.5% changed practice such that selected patients with axillary metas-
proved to have occult metastasis. Of the patients who had bone tasis can now avoid ALND if they have clinical and pathologic
marrow aspiration, 3.0% had immunohistochemically detected features similar to those patients enrolled on Z0011. However,
tumor cells in the bone marrow. Although the presence of dis- there have been some concerns raised about the Z0011 study
ease in the bone marrow identified a population at high risk for that include the fact that the study only recruited about half of
the intended patients and that there was no standardization of determine which patients with a positive SLN are at risk for har- 585
whether or not patients received irradiation to the low axilla boring additional positive non-SLNs in the axilla. These tools
when the radiation oncologist irradiated the breast. These issues can be helpful in determining the likelihood of additional disease
have thus far limited the uptake of the results of Z0011 by some in the axilla and may be used clinically to counsel patients.256
centers. In patients who present with axillary lymphadenopa-
The International Breast Cancer Study Group (IBCSG) thy that is confirmed to be metastatic disease on FNA or core
23-01 trial was similar in design to Z0011 but enrolled only biopsy, SLN dissection is not necessary, and patients can pro-
patients with micrometastases in the SLNs. Patients with SLN ceed directly to ALND or be considered for preoperative sys-
micrometastases were randomized to ALND vs. no further sur- temic therapy (see “Neoadjuvant [Preoperative] Chemotherapy”
gery. Unlike Z0011, the 23-01 trial did not exclude patients under “Nonsurgical Breast Cancer Therapies”). Initially there

CHAPTER 17 THE BREAST


treated with mastectomy. Approximately 9% of patients ran- was controversy about the suitability of SLN dissection in
domized to each study arm underwent mastectomy. The inves- women with larger primary tumors (T3) and those treated with
tigators published the primary and secondary endpoints of the neoadjuvant chemotherapy. The American Society of Clini-
trial showing no differences in OS or local-regional recurrence cal Oncology has included SLN dissection is its guidelines as
between the study arms.254 However, as with the Z0011 trial, appropriate for axillary staging in these patients.257,258 If an SLN
some concerns have been raised regarding the 23-01 study. For cannot be identified, then ALND is generally performed for
example, in the statistics on the primary endpoint, local recur- appropriate staging. However, this is not universally accepted,
rence included contralateral breast cancer and other tumor types and there are as yet no randomized studies that have assessed
as events. No hypothesis was presented as to why the differ- how a patient with a locally advanced cancer at presentation
ence in axillary surgery should impact on either of these events. should be treated if SLN dissection reveals no metastases or
Including these events therefore reduced the power of the study micrometastases after neoadjuvant therapy.
to show a statistical difference between treatment arms. There The ASCO guidelines suggest that adjuvant chemo-
is also concern that the study appears underpowered to show a therapy should be considered for patients with positive lymph
meaningful difference in overall survival. nodes, ER-negative disease, HER2-positive disease, Adju-
Most pathology laboratories perform a more detailed anal- vant! Online mortality greater than 10%, grade 3 node-neg-
ysis of the SLN than is routinely done for axillary nodes recov- ative tumors >5 mm, triple-negative tumors, lymphovascular
ered from a levels I and II dissection. This can include examining invasion, or estimated distant relapse risk of greater than 15%
thin sections of the node with step sectioning at multiple levels at 10 years based on the 21 gene recurrence score assay.259
through the paraffin blocks or performing immunohistochemi- Adjuvant endocrine therapy is considered for women with
cal staining of the SLN for cytokeratin or a combination of these hormone receptor-positive cancers, and an aromatase inhibi-
techniques. The results of ACOSOG Z0010 and NSABP B-32 tor is recommended if the patient is postmenopausal. HER2/
showed no clinically meaningful difference in survival based neu status is determined for all patients with newly diagnosed
on detection of occult metastases in the SLNs using immu- invasive breast cancer and when positive, should be used to
nohistochemical staining and do not support the routine use guide systemic therapy recommendations. The FDA approved
in SLN processing. The type of intraoperative assessment of trastuzumab in November 2006 for use as part of a treatment
SLNs also varies for different clinicians and pathology labo- regimen containing doxorubicin, cyclophosphamide, and pacli-
ratories. Some centers prefer to use touch preparation cyto- taxel for treatment of HER2/neu-positive, node-positive breast
logic analysis of the SLNs, whereas others use frozen-section cancer.181,183 Subsequently, the BCIRG 006 study reported that
analysis, and the sensitivity and specificity of these assays vary giving trastuzumab concurrently with docetaxel and carbopla-
considerably. The GeneSearch Breast Lymph Node Assay is a tin appeared as effective as giving trastuzumab following an
real-time reverse-transcriptase polymerase chain reaction assay anthracycline containing regimen.182,185 In addition to trastu-
that detects breast tumor cell metastasis in lymph nodes through zumab, pertuzumab has also recently been FDA approved for
the identification of the gene expression markers mammaglobin adjuvant use in patients with HER2 amplified breast cancers
and cytokeratin 19. These markers are present in higher lev- with high risk of recurrence.
els in breast tissue and not in nodal tissue (cell type-specific
messenger RNA). The GeneSearch breast lymph node assay Advanced Local-Regional Breast Cancer
generates expression data for genes of interest, which are then (Stage IIIA or IIIB)
evaluated against predetermined criteria to provide a qualitative Women with stage IIIA and IIIB breast cancer have advanced
(positive/negative) result. The assay is designed to detect foci local-regional breast cancer but have no clinically detected
that correspond to metastases that are seen with examination by distant metastases (Fig. 17-30).260 In an effort to provide opti-
standard hematoxylin and eosin staining and measure >0.2 mm. mal local-regional disease-free survival as well as distant dis-
The GeneSearch assay results have been compared with per- ease-free survival for these women, surgery is integrated with
manent-section histologic analysis and frozen-section analy- radiation therapy and chemotherapy (Fig. 17-31). However,
sis of sentinel nodes in a prospective trial, and the assay was it should be noted that these patients have an increased risk
approved by the FDA for the intraoperative assessment of senti- of distant metastasis that is often highlighted by radiological
nel nodes.255 When a positive node is identified intraoperatively evidence when staging PET or CT and bone scans are per-
by touch preparation, frozen-section analysis, or GeneSearch formed. Thus, the paradigm for small screen detected cancers
assay, the surgeon can proceed with immediate ALND. With where cure can be expected in >90% of patients, often by local
the findings of ACOSOG Z0011 that there is not a survival ben- treatment alone, is not appropriate for patients with locally
efit to the use of ALND in selected patients, many surgeons advanced disease.
have abandoned the intraoperative evaluation of SLNs. There Preoperative (also known as neoadjuvant) chemotherapy
are a number of nomograms and predictive models designed to should be considered in the initial management of patients with
586
PART II
SPECIFIC CONSIDERATIONS

A B

Figure 17-30. Locally advanced breast cancer. A. Mammography of the right breast reveals a large tumor with enlarged axillary lymph
nodes. B. Imaging of the left breast is normal. (Used with permission from Dr. Anne Turnbull, Consultant Radiologist/Director of Breast
Screening, Royal Derby Hospital, Derby, UK.)

locally advanced stage III breast cancer, especially those with ipsilateral breast tumor recurrence-free survival rates in this
estrogen receptor negative tumors. Chemotherapy is used to study were 95%. They noted that the ipsilateral breast tumor
maximize distant disease-free survival, whereas radiation ther- recurrence rates increased when patients had clinical N2 or N3
apy is used to maximize local-regional control and disease-free disease, >2 cm of residual disease in the breast at surgery, a
survival. pattern of multifocal residual disease in the breast at surgery,
In selected patients with stage IIIA cancer, preoperative and lymphovascular space invasion in the primary tumor. This
chemotherapy can reduce the size of the primary cancer and study demonstrated that breast-conserving surgery can be used
permit breast-conserving surgery. Investigators from the MD for appropriately selected patients with locally advanced breast
Anderson Cancer Center reported that low local-regional fail- cancer who achieve a good response with preoperative che-
ure rates could be achieved in selected patients with stage III motherapy. However, the Oxford overview of all randomized
disease treated with preoperative chemotherapy followed by studies of neoadjuvant therapy (vs. adjuvant therapy) reported a
breast-conserving surgery and radiation.261 The 5-year actuarial hazard ratio of 1.5 (i.e., 50% increase) in local recurrence rates.

Figure 17-31. Treatment pathways for stage IIIA and stage IIIB breast cancer.
A meta-analysis reported a hazard ratio of 1.3.262 These stud- immediately life threatening disease (or “visceral crisis”). This 587
ies included some patients treated with radiation therapy alone includes not only women with bone or soft tissue metastases
without resection of the primary tumor bed, which results in but also women with limited visceral metastases. Symptoms
higher local failure rates. These findings are important in view per se (e.g., breathlessness) are not in themselves an indication
of the previous findings that the avoidance of recurrence in a for chemotherapy. For example, breathlessness due to a pleural
conserved breast avoids about one breast cancer death over the effusion can be treated with percutaneous drainage, and if the
next 15 years for every four such recurrences avoided.12 The breathlessness is relieved, the patient should be commenced on
German Breast Cancer Group recently reported their local endocrine therapy; if the breathlessness is due to lymphangitic
recurrence rate in 5535 patients in seven studies. With a median spread, then chemotherapy would be the treatment of choice.
of 46 months (range 1–127) follow-up the local recurrence The same approach should be taken to other symptoms such

CHAPTER 17 THE BREAST


rates ranged from 7.6% to 19.5% for T1-T4 tumors and from as pain. Systemic chemotherapy is indicated for women with
6.4% to 17.9% for N0-N3 tumors treated with neoadjuvant hormone receptor-negative cancers, “visceral crisis,” and
therapy.238 For patients with stage IIIA disease who experience hormone-refractory metastases. Women with stage IV breast
minimal response to chemotherapy and for patients with stage cancer may develop anatomically localized problems that will
IIIB breast cancer, preoperative chemotherapy can decrease benefit from individualized surgical or radiation treatment,
the local-regional cancer burden enough to permit subsequent such as brain metastases, pleural effusion, pericardial effusion,
modified radical mastectomy to establish local-regional con- biliary obstruction, ureteral obstruction, impending or existing
trol. In both stages IIIA and IIIB disease, surgery is followed by pathologic fracture of a long bone, spinal cord compression, and
adjuvant radiation therapy. However there is a small percent- painful bone or soft tissue metastases. Bisphosphonates or anti-
age of patients who experience progression of disease during RANKL (receptor activator of nuclear factor kappa-B ligand)
neoadjuvant therapy, and therefore the surgeon should review agent, denosumab, which may be given in addition to chemo-
patients with the oncologist at regular points during the neoad- therapy or endocrine therapy, should be considered in women
juvant regimen. with bone metastases. Whether to perform surgical resection
For selected clinically indolent, ER-positive, locally of the local-regional disease in women with stage IV breast
advanced tumors, primary endocrine therapy may be considered, cancer has been debated after several reports have suggested
especially if the patient has other comorbid conditions. A series that women who undergo resection of the primary tumor have
of 195 patients with ER-positive, locally advanced breast cancer improved survival over those who do not. Khan and associates
treated by endocrine therapy—median age 69 years, median used the National Cancer Data Base to identify patterns of treat-
tumor size 6 cm, median follow-up 61 months—reported a ment in women with metastatic breast cancer and found that
5-year overall survival of 76%, a breast cancer–specific sur- those who had surgical resection with negative margins had a
vival of 86%, and a metastasis-free survival of 77%. The median better prognosis than those women who did not have surgical
time to an alternative treatment was 48 months.263 Given that therapy.267 Gnerlich et al reported similar findings using the
this was a 20-year series, the number of such patients is small SEER database, and there have been several reports subsequent
but should be considered when the clinician is discussing treat- to this study from single institutions that have confirmed these
ment options. Results from the ACOSOG Z1031 trial suggest findings.268 Some have suggested that the finding of improved
that neoadjuvant endocrine therapy is a good option for tumor survival is due to selection bias and that local therapy should be
downstaging in patients with strongly ER-positive tumors. The reserved for palliation of symptoms. A randomized trial through
preoperative endocrine prognostic index (PEPI score) can be ECOG (E2108) was designed to address this question.269 The
calculated based on pathologic findings from surgery following surgical management of patients with stage IV disease should
neoadjuvant endocrine therapy. This can help guide decision- be addressed by obtaining multidisciplinary input and by con-
making regarding the need for systemic chemotherapy in this sidering the treatment goals of each individual patient and the
patient population.264,265 patient’s treating physicians.
Internal Mammary Lymph Nodes
Metastatic disease to internal mammary lymph nodes may be
Local-Regional Recurrence
Women with local-regional recurrence of breast cancer may
occult, may be evident on chest radiograph or CT scan, or may
be separated into two groups: those who have had mastec-
present as a painless parasternal mass with or without skin
tomy and those who have had lumpectomy. Women treated
involvement. There is no consensus regarding the need for
previously with mastectomy undergo surgical resection of
internal mammary lymph node radiation therapy in women who
the local-regional recurrence and appropriate reconstruction.
are at increased risk for occult involvement (cancers involving
Chemotherapy and antiestrogen therapy are considered, and
the medial aspect of the breast, axillary lymph node involve-
adjuvant radiation therapy is given if the chest wall has not pre-
ment) but who show no signs of internal mammary lymph node
viously received radiation therapy or if the radiation oncologist
involvement. Systemic chemotherapy and radiation therapy are
feels that given the time from previous treatment there is scope
indicated in the treatment of grossly involved internal mammary
for further radiation therapy, particularly if this is palliative.
lymph nodes.
Women treated previously with a breast-conservation procedure
Distant Metastases (Stage IV) undergo a mastectomy and appropriate reconstruction. Chemo-
Treatment for stage IV breast cancer is not curative but may therapy and antiestrogen therapy are considered depending of
prolong survival and enhance a woman’s quality of life.266 the hormone receptor status and HER2 status of the tumor.
Endocrine therapies that are associated with minimal toxicity
are preferred to cytotoxic chemotherapy in ER-positive disease. Breast Cancer Prognosis
Appropriate candidates for initial endocrine therapy include Survival rates for women diagnosed with breast cancer in the
women with hormone receptor-positive cancers who do not have United States can be obtained from the SEER Program of the
588 National Cancer Institute. Data have been collected since 1973 provides the best outcome. When the tumor is quite distant from
and are updated at regular intervals. The overall 5-year rela- the central breast, the biopsy incision can be excised separately
tive survival for breast cancer patients from the time period of from the primary mastectomy incision, should a mastectomy be
2003 to 2009 from 18 SEER geographic areas was 89.2%.270 required. Radial incisions in the upper half of the breast are not
The 5-year relative survival by race was reported to be 90.4% recommended because of possible scar contracture resulting in
for white women and 78.7% for black women. The 5-year sur- displacement of the ipsilateral nipple-areola complex. Similarly,
vival rate for patients with localized disease (61% of patients) curvilinear incisions in the lower half of the breast may displace
is 98.6%; for patients with regional disease (32% of patients), the nipple-areolar complex downward.
84.4%; and for patients with distant metastatic disease (5% After excision of a suspicious breast lesion, the specimen
of patients), 24.3%. Breast cancer survival has significantly should be X-rayed to confirm that the lesion has been excised
increased over the past two decades due to improvements in with appropriate margins. The biopsy tissue specimen is ori-
entated for the pathologist using sutures, clips, or dyes. Addi-
PART II

screening and local and systemic therapies. Data from the


American College of Surgeons National Cancer Data Base can tional margins (superior, inferior, medial, lateral, superficial,
also be accessed; this data reports survival based on stage of and deep) may be taken from the surgical bed if the specimen
disease at presentation using the AJCC staging system. X-ray shows the lesion is close to one or more margins. Some
surgeons also take additional shavings from the margins as one
approach to confirm complete excision of the suspicious lesion.
SPECIFIC CONSIDERATIONS

SURGICAL TECHNIQUES IN Electrocautery or absorbable ligatures are used to achieve


BREAST CANCER THERAPY wound hemostasis. Cosmesis may be facilitated by approxima-
tion of the surgical defect using 3-0 absorbable sutures. A run-
Excisional Biopsy With Needle Localization ning subcuticular closure of the skin using 4-0 or 5-0 absorbable
Excisional biopsy implies complete removal of a breast lesion monofilament sutures is performed. Wound drainage is usually
with a margin of normal-appearing breast tissue. In the past, not required.
surgeons would obtain prior consent from the patient, allow- Excisional biopsy with needle or seed localization requires
ing mastectomy if the initial biopsy results confirmed cancer. a preoperative visit to the mammography suite for placement of
Today it is important to consider the options for local therapy a localization wire or a radioactive or magnetic seed that can be
(lumpectomy vs. mastectomy with or without reconstruction) detected intraoperatively with a handheld probe. The lesion can
and the need for nodal assessment with SLN dissection. Needle- also be targeted by sonography in the imaging suite or in the
core biopsy is the preferred diagnostic method, and excisional operating room. The lesion to be excised is accurately localized
biopsy should be reserved for those cases in which the needle by mammography, and the tip of a thin wire hook or a seed is
biopsy results are discordant with the imaging findings or clini- positioned close to the lesion (Fig. 17-33). Using the wire hook
cal examination (Fig. 17-32). In general, circumareolar incisions as a guide, or detection of the seed with a handheld probe, the
can be used to access lesions that are subareolar or within a short surgeon subsequently excises the suspicious breast lesion while
distance of the nipple-areolar complex. Elsewhere in the breast, removing a margin of normal-appearing breast tissue. Before
incisions can be placed along the lines of tension in the skin the patient leaves the operating room, specimen radiography is
that are generally concentric with the nipple-areola complex. In performed to confirm complete excision of the suspicious lesion
the lower half of the breast, the use of radial incisions typically (Fig. 17-34).

Figure 17-32. Lesion to be targeted for excisional


biopsy. A. Craniocaudal view of the left breast
demonstrating 2 lesions (arrows) to be targeted
for needle localization and excision. B. Oblique
view demonstrating target lesions. (Used with
permission from Dr. Anne Turnbull, Consultant
Radiologist/Director of Breast Screening, Royal
A B Derby Hospital, Derby, UK.)
589

CHAPTER 17 THE BREAST


A

B
Figure 17-33. Wire localization procedure. Mammographic
images of hookwire in place targeting lesions for excision in the left C
breast (A) and the right breast (B). (Used with permission from Dr.
Anne Turnbull, Consultant Radiologist/Director of Breast Screen- Figure 17-34. Specimen mammography. Specimen mam-
ing, Royal Derby Hospital, Derby, UK.) mograms demonstrating excision of targeted (A) density,
(B) calcifications, and (C) spiculated mass seen on preoperative
imaging. (Used with permission from Dr. Anne Turnbull, Con-
sultant Radiologist/Director of Breast Screening, Royal Derby
Hospital, Derby, UK.)
590 Sentinel Lymph Node Dissection incision in the lower axilla just below the hairline. After dis-
Sentinel lymph node (SLN) dissection is primarily used to secting through the subcutaneous tissue, the surgeon dissects
assess the regional lymph nodes in women with early breast through the axillary fascia, being mindful to identify blue lym-
cancers who are clinically node-negative by physical examina- phatic channels. Following these channels can lead directly to
tion and imaging studies.271-279 This method also is accurate in the SLN and limit the amount of dissection through the axillary
women with larger tumors (T3 N0), but nearly 75% of tissues. The gamma probe is used to facilitate the dissection and
9 these women will prove to have axillary lymph node to pinpoint the location of the SLN. As the dissection continues,
metastases on histologic examination, and wherever possible it the signal from the probe increases in intensity as the SLN is
is better to identify them preoperatively as this will allow a approached. The SLN also is identified by visualization of blue
definitive procedure for known axillary disease. SLN dissection dye in the afferent lymph vessel and in the lymph node itself.
has also been reported to be accurate for staging of the axilla Before the SLN is removed, a 10-second in vivo radioactivity
count is obtained. After removal of the SLN, a 10-second ex
PART II

after chemotherapy in women with clinically node-negative dis-


ease at initial presentation.280,281 Tan et al in a review and meta- vivo radioactive count is obtained, and the node is then sent to
analysis of 449 cases of SLN biopsy in clinically lymph the pathology laboratory for either permanent- or frozen-section
node-negative disease reported a sensitivity of 93%, giving a analysis. The lowest false-negative rates for SLN dissection
false negative rate of 7% with a negative predictive value of have been obtained when all blue lymph nodes and all lymph
nodes with counts >10% of the 10-second ex vivo count of the
SPECIFIC CONSIDERATIONS

94% and an overall accuracy of 95%.282 Clinical situations


where SLN dissection is not recommended include patients with SLN are harvested (“10% rule”). Based on this, the gamma
inflammatory breast cancers, those with biopsy proven metasta- counter is used before closing the axillary wound to measure
sis, DCIS without mastectomy, or prior axillary surgery. residual radioactivity in the surgical bed. A search is made for
Although limited data are available, SLN dissection appears to additional SLNs if the counts remain high. This procedure is
be safe in pregnancy when performed with radioisotope alone. repeated until residual radioactivity in the surgical bed is less
Evidence from large prospective studies suggests that than 10% of the 10-second ex vivo count of the most radioac-
the combination of intraoperative gamma probe detection of tive SLN and all blue nodes have been removed. Studies have
radioactive colloid and intraoperative visualization of blue dye demonstrated that 98% of all positive SLNs will be recovered
(isosulfan blue dye or methylene blue) is more accurate for with the removal of four SLNs; therefore, it is not necessary to
identification of SLNs than the use of either agent alone. Some remove greater than four SLNs for accurate staging of the axilla.
surgeons use preoperative lymphoscintigraphy, although it is Results from the NSABP B-32 trial showed that the false-
not required for identification of the SLNs. On the day before negative rate for SLN dissection is influenced by tumor loca-
surgery, or the day of surgery, the radioactive colloid is injected tion, type of diagnostic biopsy, and number of SLNs removed
either in the breast parenchyma around the primary tumor or at surgery.243 The authors reported that tumors located in the
prior biopsy site, into the subareolar region, or subdermally in lateral breast were more likely to have a false-negative SLN.
proximity to the primary tumor site. With a 25-gauge needle, This may be explained by difficulty in discriminating the hot
0.5 mCi of 0.2-μm technetium 99m–labeled sulfur colloid is spot in the axilla when the radioisotope has been injected at the
injected for same-day surgery, or a higher dose of 2.5 mCi of primary tumor site in the lateral breast. Those patients who had
technetium-labeled sulfur colloid is administered when the undergone an excisional biopsy before the SLN procedure were
isotope is to be injected on the day before surgery. Subdermal significantly more likely to have a false-negative SLN. This
injections are given in proximity to the cancer site or in the report further confirms that surgeons should use needle biopsy
subareolar location. Later, in the operating room, 3 to 5 mL of for diagnosis whenever possible and reserve excisional biopsy
blue dye is injected either in the breast parenchyma or in the for the rare situations in which needle biopsy findings are non-
subareolar location. It is not recommended that the blue dye be diagnostic or discordant. Finally, removal of a larger number
used in a subdermal injection because this can result in tattoo- of SLNs at surgery appears to reduce the false-negative rate. In
ing of the skin (isosulfan blue dye) or skin necrosis (methylene B-32, the false-negative rate was reduced from 17.7% to 10%
blue). For nonpalpable cancers, the injection of the technetium- when two SLNs were recovered and to 6.9% when three SLNs
labeled sulfur colloid solution can be guided by ultrasound or were removed. Yi and associates reported that the number of
by mammographic guidance. In women who have undergone SLNs that need to be removed for accurate staging is influenced
previous excisional biopsy, the injections are made in the breast by individual patient and primary tumor factors.283
parenchyma around the biopsy cavity but not into the cavity In the B-32 trial, SLNs were identified outside the levels I
itself. Women are told preoperatively that the isosulfan blue and II axillary nodes in 1.4% of cases. This was significantly
dye injection will cause a change in the color of their urine and influenced by the site of radioisotope injection. When a subareo-
that there is a very small risk of allergic reaction to the dye (1 lar or periareolar injection site was used, there were no instances
in 10,000). Anaphylactic reactions have been documented, and of SLNs identified outside the level I or II axilla, compared with
some groups administer a regimen of antihistamine, steroids, a rate of 20% when a peritumoral injection was used. This sup-
and a histamine H-2 receptor antagonist preoperatively as a ports the overall concept that the SLN is the first site of drain-
prophylactic regimen to prevent allergic reactions. The use of age from the lymphatic vessels of the primary tumor. Although
radioactive colloid is safe, and radiation exposure is very low. many patients will have similar drainage patterns from injec-
Sentinel node dissection can be performed in pregnancy with tions given at the primary tumor site and at the subareolar
the radioactive colloid without the use of blue dye. plexus, some patients will have extra-axillary drainage, either
A hand-held gamma counter is used to transcutaneously alone or in combination with axillary node drainage, and this
identify the location of the SLN. This can help to guide place- is best assessed with a peritumoral injection of the radioiso-
ment of the incision. A 3- to 4-cm incision is made in line with tope. Kong et al reported that internal mammary node drain-
that used for an axillary dissection, which is a curved transverse age on preoperative lymphoscintigraphy was associated with
worse distant disease-free survival in early-stage breast cancer indicated, intraoperative assessment of the sentinel node can 591
patients.284 proceed while the segmental mastectomy is being performed.
The use of oncoplastic surgery can be entertained at the
Breast Conservation time of segmental mastectomy or at a later time to improve the
Breast conservation involves resection of the primary breast overall aesthetic outcome. The use of oncoplastic techniques
cancer with a margin of normal-appearing breast tissue, adju- range from a simple reshaping of breast tissue to local tissue
vant radiation therapy, and assessment of regional lymph node rearrangement to the use of pedicled flaps or breast reduction
status.285,286 Resection of the primary breast cancer is alterna- techniques. The overall goal is to achieve the best possible aes-
tively called segmental mastectomy, lumpectomy, partial mas- thetic result. In determining which patients are candidates for
tectomy, wide local excision, and tylectomy. For many women oncoplastic breast surgery, several factors should be considered,
with stage I or II breast cancer, breast-conserving therapy (BCT)

CHAPTER 17 THE BREAST


including the extent of the resection of breast tissue necessary
is preferable to total mastectomy because BCT produces survival to achieve negative margins, the location of the primary tumor
rates equivalent to those after total mastectomy while preserv- within the breast, and the size of the patient’s breast and body
ing the breast.287 Six prospective randomized trials have shown habitus. Oncoplastic techniques are of prime consideration
that overall and disease-free survival rates are similar with BCT when (a) a significant area of breast skin will need to be resected
and mastectomy; however, three of the studies showed higher with the specimen to achieve negative margins; (b) a large vol-
local-regional failure rates in patients undergoing BCT. In two ume of breast parenchyma will be resected resulting in a signifi-
of these studies, there were no clear criteria for histologically cant defect; (c) the tumor is located between the nipple and the
negative margins.285-287 Data from the EBCTCG meta-analysis inframammary fold, an area often associated with unfavorable
revealed that the addition of radiation reduces recurrence by half cosmetic outcomes; or (d) excision of the tumor and closure of
and improves survival at year 15 by about a sixth.288 When all the breast may result in malpositioning of the nipple.
of this information is taken together, BCT is considered to be
oncologically equivalent to mastectomy. Mastectomy and Axillary Dissection
In addition to being equivalent to mastectomy in terms of A skin-sparing mastectomy removes all breast tissue, the
oncologic safety, BCT appears to offer advantages over mas- nipple-areola complex, and scars from any prior biopsy pro-
tectomy with regard to quality of life and aesthetic outcomes. cedures.293,294 There is a recurrence rate of less than 6% to 8%,
BCT allows for preservation of breast shape and skin as well as comparable to the long-term recurrence rates reported with stan-
preservation of sensation, and it provides an overall psychologic dard mastectomy, when skin-sparing mastectomy is used for
advantage associated with breast preservation. patients with Tis to T3 cancers. A total (simple) mastectomy
Breast conservation surgery is currently the standard treat- without skin sparing removes all breast tissue, the nipple-areola
ment for women with stage 0, I, or II invasive breast cancer. complex, and skin. An extended simple mastectomy removes
Women with DCIS require only resection of the primary cancer all breast tissue, the nipple-areola complex, skin, and the level I
and adjuvant radiation therapy without assessment of regional axillary lymph nodes. A modified radical (“Patey”) mastectomy
lymph nodes. When a lumpectomy is performed, a curvilinear removes all breast tissue, the nipple-areola complex, skin, and
incision lying concentric to the nipple-areola complex is made the levels I, II, and III axillary lymph nodes; the pectoralis minor
in the skin overlying the breast cancer when the tumor is in the that was divided and removed by Patey may be simply divided,
upper aspect of the breast. Radial incisions are preferred when giving improved access to level III nodes, and then left in situ,
the tumor is in the lower aspect of the breast. Skin excision is or occasionally the axillary clearance can be performed with-
not necessary unless there is direct involvement of the overlying out dividing pectoralis minor. The Halsted radical mastectomy
skin by the primary tumor. The breast cancer is removed with removes all breast tissue and skin, the nipple-areola complex,
an envelope of normal-appearing breast tissue that is adequate the pectoralis major and pectoralis minor muscles, and the levels
to achieve a cancer-free margin. Significant controversy has I, II, and III axillary lymph nodes. The use of systemic che-
existed on the appropriate margin width for BCT.260 However, motherapy and hormonal therapy as well as adjuvant radiation
recently the SSO and ASTRO developed a consensus statement, therapy for breast cancer have nearly eliminated the need for the
supported by data from a systematic review data, encouraging radical mastectomy.
“no tumor on ink” to be the standard definition of a negative Nipple-areolar sparing mastectomy has been popularized
margin for invasive stages I and II breast cancer in patients who over the last decade especially for risk-reducing mastectomy
undergo breast conserving surgery with whole-breast irradiation. in high risk women. For those patients with a cancer diagno-
The meta-analysis found that increasing the margin width does sis, many consider the following factors for eligibility: tumor
not affect local recurrence rates as long as the inked or transected located more than 2 to 3 cm from the border of the areola,
margin is microscopically negative.289-292 Specimen X-ray should smaller breast size, minimal ptosis, no prior breast surgeries
routinely be performed to confirm the lesion has been excised. with periareolar incisions, body mass index less than 40 kg/m2,
Specimen orientation is performed by the surgeon. Additional no active tobacco use, no prior breast irradiation, and no evi-
margins from the surgical bed are taken as needed to provide dence of collagen vascular disease.
a histologically negative margin. Requests for determination of For a variety of biologic, economic, and psychosocial rea-
ER, PR, and HER2 status are conveyed to the pathologist. sons, some women desire mastectomy rather than breast con-
It is the surgeon’s responsibility to ensure complete servation. Women who are less concerned about cosmesis may
removal of cancer in the breast. Ensuring surgical margins that view mastectomy as the most expeditious and desirable thera-
are free of breast cancer will minimize the chances of local peutic option because it avoids the cost and inconvenience of
recurrence and will enhance cure rates. If negative margins are radiation therapy. Some women whose primary breast cancers
not obtainable with reexcision, mastectomy is required. SLN is cannot be excised with a reasonable cosmetic result or those
performed before removal of the primary breast tumor. When who have extensive microcalcifications are best treated with
592
PART II

Figure 17-35. Modified radical mastectomy: eleva-


tion of skin flaps. Skin flaps are 7 to 8 mm in thick-
ness, inclusive of the skin and telasubcutanea. (Visual
Art: © 2013. The University of Texas MD Anderson
Cancer Center.)
SPECIFIC CONSIDERATIONS

mastectomy. Similarly, women with large cancers that occupy Subsequently, an axillary lymph node dissection is per-
the subareolar and central portions of the breast and women with formed. The most lateral extent of the axillary vein is identified,
multicentric primary cancers also undergo mastectomy. and the areolar tissue of the lateral axillary space is elevated as
the vein is cleared on its anterior and inferior surfaces. The areo-
Modified Radical Mastectomy lar tissues at the junction of the axillary vein and the anterior
A modified radical mastectomy preserves the pectoralis major edge of the latissimus dorsi muscle, which include the lateral
muscle with removal of levels I, II, and III (apical) axillary and subscapular lymph node groups (level I), are cleared. Care
lymph nodes.293 The operation was first described by David is taken to preserve the thoracodorsal neurovascular bundle. The
Patey, a surgeon at St Bartholomew’s Hospital London, who dissection then continues medially with clearance of the central
reported a series of cases where he had removed the pectoralis axillary lymph node group (level II). The long thoracic nerve
minor muscle allowing complete dissection of the level III axil- of Bell is identified and preserved as it travels in the investing
lary lymph nodes while preserving the pectoralis major and the fascia of the serratus anterior muscle. Every effort is made to
lateral pectoral nerve. A modified radical mastectomy permits preserve this nerve because permanent disability with a winged
preservation of the medial (anterior thoracic) pectoral nerve, scapula and shoulder weakness will follow denervation of the
which courses in the lateral neurovascular bundle of the axilla serratus anterior muscle. Patey divided the pectoralis minor and
and usually penetrates the pectoralis minor to supply the lateral removed it to allow access right up to the apex of the axilla.
border of the pectoralis major. Anatomic boundaries of the mod- The pectoralis minor muscle is usually divided at the tendinous
ified radical mastectomy are the anterior margin of the latissi- portion near its insertion onto the coracoid process (Fig. 17-37
mus dorsi muscle laterally, the midline of the sternum medially, inset), which allows dissection of the axillary vein medially to
the subclavius muscle superiorly, and the caudal extension of the costoclavicular (Halsted’s) ligament. Finally, the breast and
the breast 2 to 3 cm inferior to the inframammary fold inferiorly. axillary contents are removed from the surgical bed and are sent
Skin-flap thickness varies with body habitus but ideally is 7 to for pathologic assessment. In his modified radical mastectomy,
8 mm inclusive of skin and telasubcutanea (Fig. 17-35). Once Patey removed the pectoralis minor muscle. Many surgeons
the skin flaps are fully developed, the fascia of the pectoralis now divide only the tendon of the pectoralis minor muscle at
major muscle and the overlying breast tissue are elevated off the its insertion onto the coracoid process while leaving the rest of
underlying musculature, which allows for the complete removal the muscle intact, which still provides good access to the apex
of the breast (Fig. 17-36). of the axilla.

Figure 17-36. Modified radical mastectomy after


resection of breast tissue. The pectoralis major muscle
is cleared of its fascia as the overlying breast is elevated.
The latissimus dorsi muscle is the lateral boundary of the
dissection. (Visual Art: © 2013. The University of Texas
MD Anderson Cancer Center.)
593

CHAPTER 17 THE BREAST


Figure 17-37. Modified radical mastectomy (Patey): axillary lymph node dissection. The dissection proceeds from lateral to medial, with
complete visualization of the anterior and inferior aspects of the axillary vein. Loose areolar tissue at the junction of the axillary vein and the
anterior margin of the latissimus dorsi muscle is swept inferomedially inclusive of the lateral (axillary) lymph node group (level I). Care is
taken to preserve the thoracodorsal artery, vein, and nerve in the deep axillary space. The lateral lymph node group is resected in continuity
with the subscapular lymph node group (level I) and the external mammary lymph node group (level I). Dissection anterior to the axillary
vein allows removal of the central lymph node group (level II) and the apical (subclavicular) lymph node group (level III). The superomedial
limit of this dissection is the clavipectoral fascia (Halsted’s ligament). Inset depicts division of the insertion of the pectoralis minor muscle
at the coracoid process. The surgeon’s finger shields the underlying brachial plexus. (Reproduced with permission from Bland KI, Copeland
EMI: The Breast: Comprehensive Management of Benign and Malignant Diseases, 4th ed. Philadelphia, PA: Elsevier/Saunders; 2009.)

Seromas beneath the skin flaps or in the axilla represent of the wound edges. However, if a more radical removal of skin
the most frequent complication of mastectomy and axillary and subcutaneous tissue is necessary, a pedicled myocutane-
lymph node dissection, reportedly occurring in as many as 30% ous flap from the latissimus dorsi muscle is generally the best
of cases. The use of closed-system suction drainage reduces approach for wound coverage. A skin graft provides functional
the incidence of this complication. Catheters are retained in the coverage that will tolerate adjuvant radiation therapy; however,
wound until drainage diminishes to <30 mL per day. Wound this is not preferred because poor graft adherence may delay
infections occur infrequently after a mastectomy, and the delivery of radiation therapy. Breast reconstruction after risk-
majority are a result of skin-flap necrosis. Cultures of speci- reducing mastectomy or after mastectomy for early-stage breast
mens taken from the infected wound for aerobic and anaerobic cancer may be performed at the same time as the mastectomy.
organisms, debridement, and antibiotic therapy are effective This allows for a skin-sparing mastectomy to be performed,
management. Moderate or severe hemorrhage in the postop- which offers the best overall cosmetic outcomes. Reconstruc-
erative period is rare and is best managed with early wound tion can proceed with an expander/implant reconstruction or
exploration for control of hemorrhage and reestablishment of with autologous tissue such as a pedicled myocutaneous flap
closed-system suction drainage. The incidence of functionally or a free flap using microvascular techniques. In patients with
significant lymphedema after a modified radical mastectomy is locally advanced breast cancer, reconstruction is often delayed
approximately 20% but can be as high as 50% to 60% when until after completion of adjuvant radiation therapy to ensure
postoperative radiation is employed. Extensive axillary lymph that local-regional control of disease is obtained. The expected
node dissection, the delivery of radiation therapy, the presence use of postmastectomy radiotherapy should also be considered
of pathologic lymph nodes, and obesity are predisposing factors. as a reason for delayed reconstruction as radiotherapy to a
Patients should be referred to physical therapy at the earliest reconstructed breast has been reported to result in inferior cos-
signs of lymphedema to prevent progression to the later stages. metic outcomes. Consideration can be made for placement of
The use of individually fitted compressive sleeves and complex a tissue expander to allow for skin-sparing, but this should be
decongestive therapy may be necessary. discussed with the radiation oncologist and other members of
the treatment team. If chest wall coverage is needed to replace
Reconstruction of the Breast and Chest Wall a large skin or soft tissue defect, many different types of myo-
The goals of reconstructive surgery after a mastectomy for breast cutaneous flaps are employed, but the latissimus dorsi and
cancer are wound closure and breast reconstruction, which is the rectus abdominis myocutaneous flaps are most frequently
either immediate or delayed.295 In most cases, wound closure used. The latissimus dorsi myocutaneous flap consists of a skin
after mastectomy is accomplished with simple approximation paddle based on the underlying latissimus dorsi muscle, which
594 is supplied by the thoracodorsal artery with contributions from promising in highly selected low-risk populations, use of APBI
the posterior intercostal arteries. A transverse rectus abdominis should be based on current guidelines or offered in the setting
myocutaneous (TRAM) flap consists of a skin paddle based on of a prospective trial.304
the underlying rectus abdominis muscle, which is supplied by
vessels from the deep inferior epigastric artery. The free TRAM Chemotherapy Adjuvant
flap uses microvascular anastomoses to establish blood supply Chemotherapy. The Early Breast Cancer Trialists’ Collabora-
to the flap. When the bony chest wall is involved with cancer, tive Group overview analysis of adjuvant chemotherapy demon-
resection of a portion of the bony chest wall is indicated. If only strated reductions in the odds of recurrence and death in women
one or two ribs are resected and soft tissue coverage is pro- ≤70 years of age with stage I, IIA, or IIB breast cancer.123,305-309
vided, reconstruction of the bony defect is usually not necessary For those ≥70 years of age, the lack of definitive clinical trial
because scar tissue will stabilize the chest wall. If more than two data regarding adjuvant chemotherapy prevented definitive rec-
ribs are sacrificed, it is advisable to stabilize the chest wall with ommendations. Adjuvant chemotherapy is of minimal benefit
PART II

prosthetic material, which is then covered with soft tissue by to women with negative nodes and cancers ≤0.5 cm in size and
using a latissimus dorsi or TRAM flap. is not recommended. Women with negative nodes and cancers
0.6 to 1.0 cm are divided into those with a low risk of recurrence
and those with unfavorable prognostic features that portend a
NONSURGICAL BREAST CANCER THERAPIES
higher risk of recurrence and a need for adjuvant chemotherapy.
SPECIFIC CONSIDERATIONS

Radiation Therapy Adverse prognostic factors include blood vessel or lymph ves-
Radiation therapy is used for all stages of breast cancer sel invasion, high nuclear grade, high histologic grade, HER2/
depending on whether the patient is undergoing BCT or mas- neu overexpression, and negative hormone receptor status.
tectomy.296-302 Adjuvant radiation for patients with DCIS and American Society of Clinical Oncology guidelines suggest that
early-stage breast cancer have been described previously in this adjuvant chemotherapy should be considered for patients with
chapter. Those women treated with mastectomy who have cancer positive lymph nodes, HER2-positive disease, Adjuvant! Online
at the surgical margins are at sufficiently high risk for local mortality greater than 10%, grade 3 lymph node negative tumors
recurrence to warrant the use of adjuvant radiation therapy to >5 mm, triple-negative tumors, lympho-vascular invasion, or
the chest wall postoperatively. Women with metastatic disease estimated distant relapse risk of greater than 15% at 10 years
involving four or more axillary lymph nodes and premeno- based on 21 gene recurrence score.259 Adjuvant chemotherapy
pausal women with metastatic disease involving one to three is recommended by the NCCN guidelines for women with these
lymph nodes also are at increased risk for recurrence and are unfavorable prognostic features. Table 17-14 lists the frequently
candidates for the use of chest wall and supraclavicular lymph used chemotherapy regimens for breast cancer.
node radiation therapy. In advanced local-regional breast can- For women with hormone receptor-negative cancers
cer (stage IIIA or IIIB), women are at high risk for recurrent that are >1 cm in size, adjuvant chemotherapy is appropriate.
disease after surgical therapy, and adjuvant radiation therapy
is used to reduce the risk of recurrence. Current recommenda-
tions for stages IIIA and IIIB breast cancer are (a) adjuvant Table 17-14
radiation therapy to the breast and supraclavicular lymph nodes
after neoadjuvant chemotherapy and segmental mastectomy Adjuvant chemotherapy regimens for breast cancer
with or without axillary lymph node dissection, (b) adjuvant HER-2 NEGATIVE HER-2 POSITIVE
radiation therapy to the chest wall and supraclavicular lymph
nodes after neoadjuvant chemotherapy and mastectomy with Preferred AC → T + trastuzumab +/−
or without axillary lymph node dissection, and (c) adjuvant Dose dense AC → pertuzumab (T = paclitaxel)
radiation therapy to the chest wall and supraclavicular lymph Paclitaxel every TCH (docetaxel, carboplatin,
nodes after segmental mastectomy or mastectomy with axillary 2 weeks trastuzumab +/− pertuzumab)
lymph node dissection and adjuvant chemotherapy. Data from Dose dense AC → Other Regimens
the EBCTCG has shown improvements in local-regional con- Paclitaxel weekly AC → T + trastuzumab +/−
trol and survival in patients treated with mastectomy and post- TC (T = docetaxel) pertuzumab (T = docetaxel)
mastectomy radiation therapy for one to three positive axillary Other Regimens Docetaxel + cyclophosphamide +
lymph nodes.303 This data is based on clinical trials from the era CMF trastuzumab
of axillary lymph node dissection for staging prior to the routine AC → Docetaxel FEC → Docetaxel +
use of sentinel lymph node dissection. It is likely that the vol- every 3 weeks trastuzumab + pertuzumab
ume of disease in the earlier trials was greater overall than what AC → Paclitaxel weekly FEC → Paclitaxel +
is currently seen in patients who have small volume metastases TAC (T = docetaxel) trastuzumab + pertuzumab
detected at sentinel node dissection. It is important to include Paclitaxel + trastuzumab
all multidisciplinary team members (medical oncology, plastic Paclitaxel + trastuzumab +
surgery, radiation oncology, and surgical oncology) regarding pertuzumab → FEC
the risks and benefits of postmastectomy radiation therapy in Docetaxel + trastuzumab +
patients with one to three positive nodes. pertuzumab → FEC
The use of partial breast irradiation (APBI) for patients A = Adriamycin (doxorubicin); C = cyclophosphamide; E = epirubicin;
treated with breast-conserving surgery has also been previously F = 5-fluorouracil; M = methotrexate; T = Taxane (docetaxel or
described. APBI can be delivered via brachytherapy, external paclitaxel); → = followed by.
beam radiation therapy using 3D conformal radiation, or inten- Data from NCCN Practice Guidelines in Oncology. Fort Washington,
sity-modulated radiation therapy. Although initial results are PA: National Comprehensive Cancer Network, 2006.
However, women with node-negative hormone receptor– and any regional nodal metastases to a specific chemotherapy 595
positive cancers and T1 tumors are candidates for antiestrogen regimen.279 For patients whose tumors remain stable in size or
therapy with or without chemotherapy. Assessment of overall even progress with the initial neoadjuvant chemotherapy regi-
risk using known prognostic factors or additional testing such men, a new regimen may be considered that uses another class
as the 21-gene recurrence score assay can help to guide deci- of agents, although there is no randomized data confirming this
sion making regarding chemotherapy in patients with node- will improve outcome.
negative, ER-positive breast cancer. For special-type cancers After treatment with neoadjuvant chemotherapy, patients
(tubular, mucinous, medullary, etc), which are usually strongly are assessed for clinical and pathologic response to the regimen.
estrogen receptor positive, adjuvant antiestrogen therapy should Patients whose tumors achieve a pathologic complete response
be advised for cancers >1 cm. For women with node-positive to neoadjuvant chemotherapy have been shown to have statisti-

CHAPTER 17 THE BREAST


tumors or with a special-type cancer that is >3 cm, the use of cally improved survival outcomes to those of patients whose
chemotherapy is appropriate; those with hormone receptor- tumors demonstrate only a partial response, remain stable,
positive tumors should receive antiestrogen therapy. or progress on treatment. Researchers at MD Anderson Cancer
For stage IIIA breast cancer, preoperative chemotherapy Center have shown that residual cancer burden (RCB)—
with an anthracycline and taxane-containing regimen followed categorized into four classes, RCB-0 or pathologic complete
by either a modified radical mastectomy or segmental mastec- response, RCB-1, RCB-2, and RCB-3—is predictive of 10-year
tomy with axillary dissection followed by adjuvant radiation relapse-free survival with neoadjuvant chemotherapy in triple
therapy should be considered, especially for estrogen receptor negative, ER-positive, and HER2-positive tumors.313 Patients
negative disease. While the same regimen may be considered who experience progression of disease during neoadjuvant che-
for estrogen receptor positive disease, it is known that these motherapy have the poorest survival.314,315 This means that while
tumors respond less well to chemotherapy with <10% pCR rate patients who achieve a pCR will have a better prognosis based
overall and <3% pCR rate for lobular cancers. Other options on their response to neoadjuvant chemotherapy. Equally other
such as neoadjuvant endocrine therapy followed by local- patients will have a poorer prognosis compared to when they
regional treatment or in some cases primary endocrine therapy started neoadjuvant therapy based on the nonresponse to treat-
may be considered depending on other tumor characteristics and ment. Consequently, the FDA has supported the use of the neo-
the patient’s comorbid conditions and preference. adjuvant platform and pathologic response rates as an endpoint
for mechanism of accelerated approval for new agents in high
Neoadjuvant (Preoperative) Chemotherapy. In the early risk early stage breast cancer, though the short-term endpoints
1970s, the National Cancer Institute in Milan, Italy, initiated two (i.e., pCR) have not been shown to correlate with long-term out-
prospective randomized multimodality clinical trials for women comes (i.e., disease free survival and overall survival).
with T3 or T4 breast cancer.310 The best results were achieved Current NCCN recommendations for treatment of oper-
when surgery was interposed between chemotherapy courses, able advanced local-regional breast cancer are neoadjuvant
with 82% local-regional control and 25% having a 5-year dis- chemotherapy with an anthracycline-containing or taxane-
ease-free survival. The NSABP B-18 trial evaluated the role containing regimen or both, followed by mastectomy or lumpec-
of neoadjuvant chemotherapy in women with operable stages tomy with axillary lymph node dissection if necessary, followed
II and III breast cancer.206 Women entered into this study were by adjuvant radiation therapy. For patients with HER2-positive
randomly assigned to receive either surgery followed by che- breast cancer, trastuzumab and pertuzumab can be combined
motherapy or neoadjuvant chemotherapy followed by surgery. with chemotherapy in the preoperative setting to increase patho-
There was no difference in the 5-year disease-free survival rates logic complete response rates. For inoperable stage IIIA and for
for the two groups, but after neoadjuvant chemotherapy there stage IIIB breast cancer, neoadjuvant chemotherapy is used to
was an increase in the number of lumpectomies performed and decrease the local-regional cancer burden. This may then permit
a decreased incidence of node positivity. It was suggested that subsequent modified radical or radical mastectomy, which is
neoadjuvant chemotherapy be considered for the initial manage- followed by adjuvant radiation therapy.
ment of breast cancers judged too large for initial lumpectomy.
Several prospective clinical trials have evaluated the neo- Nodal Evaluation in Patients Receiving Neoadjuvant
adjuvant approach, and two meta-analyses have been performed, Chemotherapy. The management of the axilla after neoadjuvant
each showing that neoadjuvant vs. adjuvant chemotherapy are chemotherapy has not been specifically addressed in randomized
equivalent in terms of OS.262,311 These analyses also evaluated trials. Standard practice has been to perform an axillary lymph
local-regional recurrence (LRR) and found that there was an node dissection after chemotherapy or to perform a sentinel lymph
increase in LRR rates for patients receiving neoadjuvant chemo- node dissection before chemotherapy for nodal staging before
therapy when radiation therapy was used alone without surgery chemotherapy is initiated. A number of small single-institution
after completion of chemotherapy. Mittendorf and colleagues studies, one multicenter study, and a recent meta-analysis have
evaluated a contemporary series of almost 3000 patients treated explored the use of SLN dissection at the completion of chemo-
with breast conserving surgery and radiation therapy who therapy. The published results from these studies have demon-
received either neoadjuvant or adjuvant chemotherapy for breast strated the feasibility of SLN dissection in breast cancer patients
cancer.312 They found that the risk of LRR was driven by bio- after neoadjuvant chemotherapy. A review of 14 studies with 818
logic factors and disease stage and was not impacted by patients showed a false negative rate of 11% with an overall accu-
10 the timing of chemotherapy delivery. These data high- racy of 94%.280,281,316 While SLN dissection has been accepted for
light the importance of the multidisciplinary management of assessment of the axilla in the clinically node-negative axilla after
patients with breast cancer in achieving the best outcomes. neoadjuvant chemotherapy, clinicians have been slower to adopt
The use of neoadjuvant chemotherapy offers the oppor- this approach for axillary staging after chemotherapy in patients
tunity to observe the response of the intact primary tumor who started with initial node-positive disease. Several clinical
596 trials have been performed to evaluate the accuracy of SLN dis- women with hormone receptor-positive disease who otherwise
section in patients with documented axillary metastases at initial would have to be treated with mastectomy, although long-term
presentation, including ACOSOG Z1071, SENTINA, and SN recurrence rates have not been reported.265 The IMPACT trial
FNAC. ACOSOG Z1071 (Alliance) analyzed women with clini- evaluated neoadjuvant use of tamoxifen or anastrozole or both
cal T0-T4, N1-N2, M0 breast cancer who underwent both SLN in combination in postmenopausal women with ER-positive
surgery and axillary lymph node dissection (ALND).317 The pri- operable or locally advanced breast cancer.321 While there were
mary endpoint was the false-negative rate (FNR) of SLN surgery no significant differences in objective tumor response among
after chemotherapy with clinically node-positive disease with a tamoxifen, anastrozole, or a combination of the two, in patients
prespecified endpoint of 10% considered to be an acceptable rate. who were initially deemed as mastectomy candidates, only 31%
However, the FNR was found to be 12.6%, though it was lower had breast-conserving surgery with tamoxifen, whereas 44%
when dual-agent mapping technique was used and at least three or underwent breast-conserving surgery with anastrozole. Invasive
more SLNs removed.317 The SENTINA and SN FNAC trials had lobular cancers in particular have been shown to respond poorly
PART II

findings similar to Z1071. The results from Z1071 were further to neoadjuvant chemotherapy and may have better response to
analyzed to determine if a clip was placed in the positive node at neoadjuvant endocrine therapy.322-324 A meta-analysis evaluating
initial diagnosis and if the clipped node location at surgery (SLN the response rate and rate of breast conservation surgery with
or ALND) was evaluated. Indeed, this showed that identification the use of neoadjuvant endocrine therapy compared to combi-
of the clipped node during the surgical procedure further decreased nation chemotherapy was recently reported. This meta-analysis
SPECIFIC CONSIDERATIONS

the FNR.318 The results from the ACOSOG Z1071 (Alliance) trial, included nearly 3500 patients across 20 studies.325 Interestingly,
in cases presenting with cN1 disease and at least two SLN resec- aromatase inhibitors had a similar response, and breast conserva-
tions and clipped node was within the SLN specimen, showed that tion rates in comparison with combination chemotherapy albeit
the FNR was 6.8%.318 Caudle et al at MD Anderson Cancer Center with lower toxicity suggest that neoadjuvant endocrine therapy
performed a prospective study of patients with biopsy-confirmed is an appropriate alternative in ER-positive breast cancers.
nodal metastases with a clip placed in the biopsy-proven lymph However, the incidence of complete pathological response was
node, who were treated with neoadjuvant chemotherapy; at the low (<10%) with both approaches. Also, aromatase inhibitors
time of surgery these patients underwent SLN dissection with were associated with significantly higher response and breast
targeting and removal of the clipped node (targeted axillary dis- conservation rates compared with tamoxifen. The ALTER-
section [TAD]).319 TAD includes SLN surgery and selective local- NATE (Alternate Approaches for Clinical Stage II or III Estro-
ization and removal of the clipped node, with the goal to determine gen Receptor Positive Breast Cancer Neoadjuvant Treatment
if pathologic changes in the clipped node accurately reflect the in Postmenopausal Women) trial is currently evaluating neo-
status of the nodal basin, and proposing that TAD improves the adjuvant endocrine therapy with fulvestrant or anastrozole or
FNR compared to SLN surgery alone.319 In patients undergoing in combination.
SLN surgery and ALND (n = 118), the FNR was 10.1% (95% Increasing knowledge of secondary resistance mecha-
CI, 4.2–19.8), and adding evaluation of the clipped node reduced nisms to endocrine therapy and cross talk between ER and the
the FNR to 1.4% (95% CI, 0.03–7.3; P = .03). TAD followed by PI3K/Akt/mTOR pathway have led to the evaluation of PI3K
ALND was performed in 85 patients, with an FNR of 2.0% (1 of pathway inhibitors in combination with endocrine therapy. Post-
50; 95% CI, 0.05–10.7).319 Although the use of dual tracer tech- menopausal women with ER-positive early breast cancers were
nique, retrieval of three or more SLNs, and TAD improve axillary treated with letrozole or letrozole in combination with everoli-
staging after neoadjuvant chemotherapy, there is no long-term data mus, a mTOR inhibitor, in a randomized, phase 2 clinical trial.
about the oncologic safety of omitting ALND in patients who con- Clinical response and antiproliferative response, characterized
vert from cN1 to cN0 disease at this time. by reduction in Ki67, was superior in the combination arm, sug-
gesting that everolimus can increase efficacy of neoadjuvant
Neoadjuvant Endocrine Therapy. While initially used in letrozole.326 The LORLEI study is evaluating the use of taselisib,
elderly women who were deemed poor candidates for surgery a PI3K inhibitor in combination with letrozole compared with
or cytotoxic chemotherapy, neoadjuvant endocrine therapy is letrozole alone. With the approval of CDK 4/6 inhibitors in the
being increasingly evaluated in clinical trials. As clinicians metastatic setting, clinical trials are evaluating the use of CDK
have gained experience with neoadjuvant treatment strategies, inhibitors in combination with neoadjuvant endocrine therapy.
it is now clear from examination of predictors of complete Neoadjuvant anastrozole in combination with palbociclib, a
pathologic response that ER-positive tumors do not shrink in CDK4/6 inhibitor, has been shown to significantly reduce Ki67,
response to chemotherapy as readily as ER-negative tumors.320 suggesting that CDK4/6 inhibition can increase the efficacy of
Indeed, the pCR rate in ER-negative tumors is approximately neoadjuvant endocrine therapy.
three times that of ER-positive tumors. Fisher et al examined With the use of neoadjuvant chemotherapy or endocrine
the results of the NSABP B-14 and B-20 trials and found that, therapy, observation of the response of the intact tumor and/or
as age increased, women obtained less benefit from chemo- nodal metastases to a specific regimen could ultimately help to
therapy. They recommended that factors214 including tumor define which patients will benefit from specific therapies in the
estrogen receptor concentration, nuclear grade, histologic grade, adjuvant setting. In adjuvant trials the primary endpoint is typi-
tumor type, and markers of proliferation should be considered in cally survival, whereas in neoadjuvant trials the endpoints have
these patients before choosing between the use of chemotherapy more often been clinical or pathologic response rates. There are
and hormonal therapy. If in fact the tumor is estrogen-receptor a number of clinical trials underway comparing neoadjuvant
rich, these patients may benefit more from endocrine therapy in chemotherapy and endocrine therapy regimens with pretreat-
the neoadjuvant setting than they might if they received stan- ment and posttreatment biopsy samples obtained from the pri-
dard chemotherapy. Neoadjuvant endocrine therapy has been mary tumors in all of the participants. These samples are being
shown to shrink tumors, enabling breast-conserving surgery in subjected to intensive genomic and proteomic analyses that may
help to define a more personalized or individualized approach to DCIS. With the use of aromatase inhibitors in postmenopausal 597
breast cancer treatment in the future. women, use of adjuvant tamoxifen has increasingly been limited
to premenopausal women.
Antiestrogen Therapy Aromatase Inhibitors. In postmenopausal women, aromatase
Tamoxifen. Within the cytosol of breast cancer cells are spe- inhibitors are now considered first-line therapy in the adjuvant
cific proteins (receptors) that bind and transfer steroid moieties setting. Currently, three third-generation aromatase inhibitors are
into the cell nucleus to exert specific hormonal effects.308,327-331 approved for clinical use: the reversible nonsteroidal inhibitors
The most widely studied hormone receptors are the estrogen anastrozole and letrozole and the irreversible steroidal inhibitor
receptor and progesterone receptor. Hormone receptors are exemestane. While all the aromatase inhibitors have been shown
detectable in >90% of well-differentiated ductal and lobular to have similar efficacy with a similar spectrum of adverse

CHAPTER 17 THE BREAST


invasive cancers. Although the receptor status may remain the effects, the Early Breast Cancer Trialists’ Collaborative Group
same between the primary cancer and metastatic disease in the meta-analyses of 31,920 postmenopausal women with ER-
same patient in the majority of cases, there are instances where positive early breast cancers treated with tamoxifen or aroma-
the status is changed in the metastatic focus; therefore, biopsy tase inhibitors demonstrated that 5 years of aromatase inhibitors
of newly diagnosed metastatic disease should be considered for reduced the rate of recurrence by 30% and 10-year breast cancer
assessment of hormone receptor and HER2 status. mortality by about 15% compared to 5 years of tamoxifen.336-339
After binding to estrogen receptors in the cytosol, tamoxi- The NSABP B42 study evaluated whether an additional 5 years
fen blocks the uptake of estrogen by breast tissue. Clini- of letrozole improved disease-free survival in postmenopausal
cal responses to antiestrogen are evident in >60% of women women who have completed 5 years of tamoxifen or an aromatase
with hormone receptor-positive breast cancers but in <10% inhibitor. After a median follow-up of 6.9 years, while extended
of women with hormone receptor-negative breast cancers. A letrozole significantly improved breast cancer-free interval, no
meta-analysis by the Early Breast Cancer Trialists’ Collabora- improvement in disease-free survival, the primary endpoint, was
tive Group showed that adjuvant therapy with tamoxifen for observed. Recently, the results of the MA-17R study, designed
5 years reduced breast cancer mortality by about a third through to assess the efficacy of adjuvant letrozole for 10 years, were
the first 15 years of follow-up.14 This mortality benefit contin- reported.340 Similar to NSABP B42, extended letrozole improved
ues to be statistically significant in the second and third 5-year disease-free survival without significant improvement in overall
periods (i.e., years 5–9 and 10–15) when the patients are no survival. Patients who are node-positive, have received adjuvant
longer receiving endocrine treatment—the so-called carry-over chemotherapy, with prior receipt of tamoxifen are likely to ben-
effect. The analysis also showed a 39% reduction in the risk efit from long-term use of an aromatase inhibitor.
of cancer in the contralateral breast. The antiestrogens do have The aromatase inhibitors are less likely than tamoxifen to
defined toxicity, including bone pain, hot flashes, nausea, vom- cause endometrial cancer but do lead to changes in bone mineral
iting, and fluid retention. Thrombotic events occur in <3% of density that may result in osteoporosis and an increased rate of
treated women. Cataract surgery is more frequently performed fractures in postmenopausal women. The risk of osteoporosis
in patients receiving tamoxifen. The Stockholm trial showed can be averted by treatment with bisphosphonates. Joint pains
that 5 years of tamoxifen was associated with a significant are a side effect that affects a significant number of patients.
reduction in locoregional recurrences and distant metastasis Node-negative and node-positive breast cancer patients whose
in postmenopausal women with ER-positive breast cancer.332 tumors express hormone receptors should be considered for
However, an increase in endometrial cancers was observed with endocrine therapy in the adjuvant setting. Women with hormone
long-term tamoxifen use. The NSABP B14 trial evaluated 10 receptor–positive cancers achieve significant reduction in risk
years of tamoxifen compared to 5 years.333 However, the study of recurrence of breast cancer and mortality from breast cancer
was terminated based on interim analyses indicating no addi- through the use of endocrine therapies.
tional benefit from tamoxifen beyond 5 years. The ATLAS trial For postmenopausal women with ER-positive, HER2-
also evaluated the use of tamoxifen for 5 years vs. 10 years negative, metastatic breast cancer, available endocrine thera-
in nearly 13,000 women across the world. This study showed pies include nonsteroidal aromatase inhibitors (anastrozole and
that continuing tamoxifen for 10 years vs. 5 years produced a letrozole); steroidal aromatase inhibitors (exemestane); serum
significant reduction in recurrence and mortality.334 Interestingly, ER modulators (tamoxifen or toremifene); ER down-regulators
the benefit was not seen in the second 5 years (i.e., years 5–9) (fulvestrant); progestin (megestrol acetate); androgens
while the patients were on treatment, but it was seen from (fluoxymesterone); and high-dose estrogen (ethinyl estradiol).
years 10 to 15. One reason the NSABP B14 study was led to A third generation nonsteroidal aromatase inhibitor or palbo-
conclude that 10 years of tamoxifen was not beneficial was that ciclib, the CDK 4/6 inhibitor, in combination with letrozole
the follow-up time was shorter. Results of the ATLAS study may be considered as a treatment option for first-line therapy.
were also corroborated by the aTTom study. Similarly, extended Activation of CDK4/CDK6 cell cycle signaling axis has been
adjuvant therapy with letrozole after 5 years of tamoxifen was implicated in mediating endocrine resistance. Consequently,
shown to improve disease-free survival without improvement in PALOMA-1 evaluated the safety and efficacy of palbociclib in
overall survival except in node-positive patients.335 combination with letrozole vs. letrozole alone as first-line treat-
Tamoxifen therapy is also considered for women with ment for patients with ER-positive, HER2-negative advanced
DCIS that is found to be ER-positive. The goals of such ther- breast cancer. Median progression-free survival (PFS) was
apy are to decrease the risk of an ipsilateral recurrence after doubled with the combination compared to letrozole alone
breast conservation therapy for DCIS and to decrease the risk (20.2 months vs. 10.2 months for the letrozole).341 Based on this,
of a primary invasive breast cancer or a contralateral breast the FDA approved palbociclib in combination with letrozole
cancer event. Consequently, tamoxifen is not recommended for for the treatment of postmenopausal women with ER-positive,
patients who have had bilateral mastectomies with ER-positive HER2-negative advanced breast cancer as initial treatment. The
598 benefit of palbociclib in combination with letrozole was sub- Ablative Endocrine Therapy
sequently confirmed in a phase 3 trial (PFS 24.8 months vs. In the past, adrenalectomy and/or hypophysectomy were the pri-
14.5 months for letrozole).342 Two additional CDK4/6 inhibitors, mary endocrine modalities used to treat metastatic breast cancer,
ribociclib and abemaciclib, have been approved for use in com- but today these approaches are seldom used. In women who are
bination with endocrine therapy for patients with hormone premenopausal at diagnosis, ovarian ablation can be accomplished
receptor–positive advanced breast cancer. by oophorectomy or ovarian radiation. Ovarian suppression can
On the other hand, PALOMA-3 compared the combina- be accomplished by the use of gonadotrophin-hormone releasing
tion of palbociclib and fulvestrant to fulvestrant alone in pre- or hormone agonists, such as goserelin or leuprolide. Evaluation of
postmenopausal ER-positive, HER2-negative metastatic breast the combination of goserelin with tamoxifen vs. cyclophospha-
cancer patients, whose disease progressed on prior endocrine mide/methotrexate/fluorouracil chemotherapy in premenopausal
therapy. Premenopausal women also received the GNRH ago- ER-positive early-stage breast cancers showed that relapse-free
PART II

nist, goserelin. The median PFS was 9.2 months for the combi- survival was superior with endocrine therapy combination, with
nation compared to 3.8 months with fulvestrant alone.343 Thus, a similar trend in overall survival.350 Data from the SOFT and
fulvestrant with palbociclib is a potential option for women with TEXT trials on adjuvant endocrine therapy show that exemes-
metastatic breast cancer who have progressed on prior endo- tane plus ovarian suppression significantly reduces recurrences
crine therapy. Additionally, abemaciclib in combination with as compared with tamoxifen plus ovarian suppression.351,352
fulvestrant or as single agent is approved for use in ER-posi-
SPECIFIC CONSIDERATIONS

In these trials, ovarian suppression was achieved with the use of


tive advanced breast cancers previously treated with endocrine the gonadotropin-releasing hormone agonist triptorelin, oopho-
therapy. rectomy, or ovarian irradiation. The disease-free survival was
In premenopausal women with stage IV ER-positive 89% in the tamoxifen plus ovarian suppression group, while it
breast cancer without previous exposure to endocrine therapy, was 93% in exemestane plus ovarian suppression group; how-
initial treatment with tamoxifen or ovarian suppression/ablation ever, there was no significant differences in overall survival. In
plus aromatase inhibitor with or without CDK4/6 inhibitors are the SOFT trial, while tamoxifen plus ovarian suppression was
reasonable options. not superior to tamoxifen alone in terms of disease-free survival,
Activation of the PI3K/mammalian target of rapamycin improved outcomes were observed in ovarian suppression in
(mTOR) signal transduction pathway has also been implicated women with a high risk of recurrence. In women who received
in secondary resistance to estrogen targeting. BOLERO-2 eval- no adjuvant chemotherapy, no meaningful benefit was obtained
uated the use of exemestane in combination with everolimus with ovarian suppression. Thus, ovarian suppression in combi-
in postmenopausal women with ER-positive tumors who had nation with an aromatase inhibitor can be considered in select
progressed or recurred on a nonsteroidal aromatase inhibitor.344 premenopausal women with high-risk features (age <40 years,
An improvement in PFS was observed with combination com- positive lymph nodes) who warranted adjuvant chemotherapy.
pared to exemestane alone (11 vs. 4.1 months) leading to FDA
approval. Similar improvement in PFS was observed with a Anti-HER2 Therapy
combination of tamoxifen and everolimus.345 However, a phase The determination of tumor HER-2 expression or gene ampli-
3 trial of letrozole in combination with temsirolimus, an mTOR fication for all newly diagnosed patients with breast cancer is
inhibitor, did not show any improvement in PFS in aromatase now recommended.353-356 It is used to assist in the selection of
inhibitor–naive metastatic postmenopausal women.346 Trials adjuvant chemotherapy in both node-negative and node-positive
evaluating the adjuvant use of mTOR inhibitors and CDK 4/6 patients. Trastuzumab was initially approved for the treatment
inhibitors are currently in progress. of HER2/neu-positive breast cancer in patients with metastatic
Women whose tumors respond to an endocrine therapy disease. Once efficacy was demonstrated for patients with
with either shrinkage of their breast cancer (objective response) metastatic disease, the NSABP and the North Central Cancer
or long-term stabilization of disease (stable disease) are con- Treatment Group conducted phase 3 trials that evaluated the
sidered to represent “clinical benefit” and should receive addi- impact of adjuvant trastuzumab therapy in patients with early-
tional endocrine therapy at the time of progression because stage breast cancer. After approval from the FDA, these groups
their chances of a further response remain high.294-296 Patients amended their adjuvant trastuzumab trials (B-31 and N9831,
whose tumors progress de novo on an endocrine agent have a respectively), to provide for a joint efficacy analysis. The first
low rate of clinical benefit (<20%) to subsequent endocrine joint interim efficacy analysis demonstrated an improvement in
therapy; the choice of endocrine or chemotherapy should be 3-year disease-free survival from 75% in the control arm to 87%
considered based on the disease site and extent as well as the in the trastuzumab arm (hazard ratio = 0.48, P <.0001). There
patient’s general condition and treatment preference.294 was an accompanying 33% reduction in mortality in the patients
The adjuvant use of aromatase inhibitors and recent who received trastuzumab (hazard ratio = 0.67, P = 0.015).
advances in tumor genome sequencing technologies have The magnitude of reduction in hazard for disease-free survival
enabled the identification of secondary ESR1 mutations.347,348 events crossed prespecified early reporting boundaries, so the
These mutations, typically present in the ligand binding data-monitoring committees for both groups recommended that
domains, lead to ligand-independent activation of the receptor, randomized accrual to the trials be ended, and the results were
mediate resistance to aromatase inhibitors, and are associated subsequently published.181
with shorter survival.349 Reported incidence of these mutations While anthracycline-based adjuvant chemotherapy was
are variable (20%–30%) based on prior exposure to aroma- considered preferable in HER2-positive breast cancer, the
tase inhibitors and are uncommon in primary breast cancers. BCIRG 006 compared the use of anthracycline with taxane and
Clinical trials evaluating novel selective estrogen receptor trastuzumab (AC-TH) versus taxane, carboplatin chemotherapy
degraders with potential activity against these mutations are with trastuzumab (TCH).182 With 10 years of follow-up, no
in progress. statistical significance with regard to disease-free and overall
survival was observed for anthracycline-based chemotherapy. chemotherapy.362 With the use of dual antibody therapy, cur- 599
While anthracycline chemotherapy was numerically superior, rently there is significant interest in identifying patients who
this was accompanied by an increase in the incidence of leu- can avoid chemotherapy and potentially be treated with HER2-
kemia and congestive heart failure. A year of adjuvant trastu- targeted agents alone. The NeoSphere study showed 27%
zumab is considered standard of care. Two years of adjuvant pathologic complete response in HER2-positive, ER-negative,
trastuzumab has been shown to be more effective, although it is breast cancer patients treated with pertuzumab and trastuzumab
associated with more toxicity than 1 year of trastuzumab.357 On alone. Pertuzumab was recently FDA approved in combination
the other hand, the PHARE trial examined 6 months vs. stan- with trastuzumab and chemotherapy in the adjuvant setting in
dard 12 months of trastuzumab. After 3.5 years of follow-up, the HER2 amplified breast cancers with high risk of recurrence.
study failed to demonstrate that 6 months was noninferior com- Approval is based on APHINITY trial showing that the addition

CHAPTER 17 THE BREAST


pared to the standard therapy.358 Patients with HER2-positive of pertuzumab improved invasive disease free survival (7.1%)
tumors benefit if trastuzumab is added to taxane chemotherapy. compared to placebo (8.7%) (HR 0.82, 95% CI: 0.67, 1.00;
Because of overlapping cardiotoxicities, trastuzumab is not usu- p = 0.047). Overall survival data is not mature.
ally given concurrently with anthracyclines. The ExteNET study evaluated the use of neratinib, an
Buzdar and colleagues reported the results of a random- irreversible inhibitor of EGFR, HER2, and HER4, in HER2-
ized neoadjuvant trial of trastuzumab in combination with positive early stage patients who have completed adjuvant
sequential paclitaxel followed by FEC-75 (5-fluorouracil, epi- trastuzumab. A year of neratinib after completion of chemo-
rubicin, cyclophosphamide) vs. the same chemotherapy regimen therapy and trastuzumab-based adjuvant therapy significantly
without trastuzumab in 42 women with early-stage operable improved 2-year disease-free survival, the primary endpoint.363
breast cancer. The pathologic complete response rates in this After two years, invasive disease free survival was 94.2% in
trial increased from 25% to 66.7% when chemotherapy was patients treated with neratinib compared with 91.9% in those
given concurrently with trastuzumab.301 A subsequent report receiving placebo (HR 0.66; 95% CI: 0.49, 0.90, p = 0.008)
that included additional patients treated with concurrent chemo- leading to FDA approval for HER2 amplified breast cancers
therapy and trastuzumab further confirmed the high pathologic following a year of adjuvant trastuzumab.
complete response rates and continued to show that cardiac In addition to amplifications or copy number alterations,
function was preserved.302 activating mutations or single nucleotide variants in HER2
While novel agents have been approved for the treatment have been described (2%).364 Typically observed in ER-positive
of women with metastatic HER2-positive breast cancers, cur- breast cancers, a higher prevalence of HER2 mutations have
rently trastuzumab is the only HER2-targeted agent approved been reported in invasive lobular carcinomas, particularly in
for use in the adjuvant setting. Lapatinib is a dual tyrosine kinase the pleomorphic subtype.365 These mutations, usually exclusive
inhibitor that targets both HER2 and EGFR. It was approved with HER2 amplification, are observed in kinase or extracellular
for use with capecitabine in patients with HER2-positive meta- domains and predict for responses or resistance to HER2-targeting
static disease. Adjuvant lapatinib was shown to be inferior to agents.366,367 A phase 2 trial of neratinib in HER2-mutated meta-
trastuzumab, and the combination of lapatinib with trastuzumab static breast cancers showed a clinical benefit rate of 36% with
did yield a significant improvement in disease-free survival one complete response and one partial response in a heavily pre-
compared to trastuzumab alone. Ado-trastuzumab emtansine treated population. A clinical trial evaluating the combination of
(T-DM1) is approved for HER2-positive metastatic breast neratinib with fulvestrant, in HER2-mutated, ER-positive breast
cancer patients who have previously received trastuzumab cancers, is in progress.
and a taxane either separately or in combination. T-DM1 is an
antibody drug conjugate that incorporates the HER2 targeted
activity of trastuzumab with the cytotoxic activity of DM1, a SPECIAL CLINICAL SITUATIONS
microtubule inhibitory agent leading to apoptosis.359
Pertuzumab is a humanized monoclonal antibody that Nipple Discharge
binds at a different epitope of the HER2 extracellular domain Unilateral Nipple Discharge. Nipple discharge is a finding
(subdomain II) and prevents dimerization of HER2 with other that can be seen in a number of clinical situations. It may be
members of the family, primarily HER3. In the metastatic suggestive of cancer if it is spontaneous, unilateral, localized
setting, it is approved in combination with trastuzumab and to a single duct, present in women ≥40 years of age, bloody,
docetaxel for patients with metastatic HER2-positive breast or associated with a mass. A trigger point on the breast may
cancer who have not received prior HER2-targeted therapy be present so that pressure around the nipple-areolar complex
or chemotherapy for metastatic disease.360 In the neoadjuvant induces discharge from a single duct. In this circumstance,
setting, pertuzumab is approved in combination with trastu- mammography and ultrasound are indicated for further evalu-
zumab and docetaxel in HER2-positive, early stage breast ation. A ductogram also can be useful and is performed by can-
cancers that are greater than 2 cm or node-positive. However, nulating a single discharging duct with a small nylon catheter
this approval is based on improvement in pathologic complete or needle and injecting 1.0 mL of water-soluble contrast solu-
response rate, and not data based on improvement in event free tion. Nipple discharge associated with a cancer may be clear,
or overall survival.361,362 In the NeoSphere trial, neoadjuvant bloody, or serous. Testing for the presence of hemoglobin is
use of pertuzumab with trastuzumab and docetaxel led to nearly helpful, but hemoglobin may also be detected when nipple dis-
a 17% increase in pathologic complete response in the breast charge is secondary to an intraductal papilloma or duct ecta-
(P = .0141).361 While in the TRYPHAENA study, pathologic sia. Definitive diagnosis depends on excisional biopsy of the
complete responses ranging from 57% to 66% were observed with offending duct and any associated mass lesion. A 3.0 lacrimal
neoadjuvant pertuzumab and trastuzumab combination given duct probe can be used to identify the duct that requires exci-
with anthracycline-containing or nonanthracycline-containing sion. Another approach is to inject methylene blue dye within
600 the duct after ductography. The nipple must be sealed with fetus is delivered. A modified radical mastectomy can be per-
collodion or a similar material so that the blue dye does not formed during the first and second trimesters of pregnancy, even
discharge through the nipple but remains within the distended though there is an increased risk of spontaneous abortion after
duct facilitating its localization. Localization with a wire or first-trimester anesthesia. During the third trimester, lumpec-
seed is performed when there is an associated mass that lies tomy with axillary node dissection can be considered if adju-
>2.0 to 3.0 cm from the nipple. vant radiation therapy is deferred until after delivery. Lactation
Bilateral Nipple Discharge. Nipple discharge is suggestive is suppressed. Chemotherapy administered during the first tri-
of a benign condition if it is bilateral and multiductal in origin, mester carries a risk of spontaneous abortion and a 12% risk of
occurs in women ≤39 years of age, or is milky or blue-green. birth defects. There is no evidence of teratogenicity resulting
Prolactin-secreting pituitary adenomas are responsible for bilat- from administration of chemotherapeutic agents in the second
eral nipple discharge in <2% of cases. If serum prolactin levels and third trimesters. For this reason, many clinicians now con-
sider the optimal strategy to be delivery of chemotherapy in the
PART II

are repeatedly elevated, plain radiographs of the sellaturcica are


indicated, and thin section CT scan is required. Optical nerve second and third trimesters as a neoadjuvant approach, which
compression, visual field loss, and infertility are associated with allows local therapy decisions to be made after the delivery of
large pituitary adenomas. the baby. Pregnant women with breast cancer often present at
a later stage of disease because breast tissue changes that occur
Axillary Lymph Node Metastases in the in the hormone-rich environment of pregnancy obscure early
SPECIFIC CONSIDERATIONS

Setting of an Unknown Primary Cancer cancers. However, pregnant women with breast cancer have a
A woman who presents with an axillary lymph node metasta- prognosis, stage by stage, that is similar to that of nonpregnant
sis that is consistent with a breast cancer metastasis has a 90% women with breast cancer.
probability of harboring an occult breast cancer.303 However,
axillary lymphadenopathy is the initial presenting sign in only Male Breast Cancer
1% of breast cancer patients. Fine-needle aspiration biopsy or Fewer than 1% of all breast cancers occur in men.369,370 The inci-
core-needle biopsy can be used to establish the diagnosis when dence appears to be highest among North Americans and the
an enlarged axillary lymph node is identified. When metastatic British, in whom breast cancer constitutes as much as 1.5% of
cancer is found, immunohistochemical analysis may classify all male cancers. Jewish and African-American men have the
the cancer as epithelial, melanocytic, or lymphoid in origin. highest incidence. Male breast cancer is preceded by gyneco-
The presence of hormone receptors (estrogen or progesterone mastia in 20% of men. It is associated with radiation exposure,
receptors) suggests metastasis from a breast cancer but is not estrogen therapy, testicular feminizing syndromes, and Kline-
diagnostic. The search for a primary cancer includes careful felter’s syndrome (XXY). Breast cancer is rarely seen in young
examination of the thyroid, breast, and pelvis, including the males and has a peak incidence in the sixth decade of life. A
rectum. The breast should be examined with diagnostic mam- firm, nontender mass in the male breast requires investigation.
mography, ultrasonography, and MRI to evaluate for an occult Skin or chest wall fixation is particularly worrisome.
primary lesion. Further radiologic and laboratory studies should DCIS makes up <15% of male breast cancer, whereas infil-
include chest radiography and liver function studies. Additional trating ductal carcinoma makes up >85%. Special-type cancers,
imaging of the chest, abdomen, and skeleton may be indicated including infiltrating lobular carcinoma, have occasionally been
if the extent of nodal involvement is consistent with stage III reported. Male breast cancer is staged in the same way as female
breast cancer. Suspicious findings on mammography, ultra- breast cancer, and stage by stage, men with breast cancer have the
sonography, or MRI necessitate breast biopsy. When a breast same survival rate as women. Overall, men do worse because of
cancer is found, treatment consists of an axillary lymph node the more advanced stage of their cancer (stage II, III or IV) at the
dissection with a mastectomy or preservation of the breast fol- time of diagnosis. The treatment of male breast cancer is surgi-
lowed by whole-breast radiation therapy. Chemotherapy and cal, with the most common procedure being a modified radical
endocrine therapy should be considered. mastectomy. SLN dissection has been shown to be feasible and
accurate for nodal assessment in men presenting with a clinically
Breast Cancer During Pregnancy node-negative axilla. Adjuvant radiation therapy is appropriate in
Breast cancer occurs in 1 of every 3000 pregnant women, and cases in which there is a high risk for local-regional recurrence.
axillary lymph node metastases are present in up to 75% of Approximately 80% of male breast cancers are hormone recep-
these women.368 The average age of the pregnant woman with tor–positive, and adjuvant tamoxifen is considered. Systemic che-
breast cancer is 34 years. Fewer than 25% of the breast nodules motherapy is considered for men with hormone receptor-negative
developing during pregnancy and lactation will be cancerous. cancers and for men with large primary tumors, multiple positive
Ultrasonography and needle biopsy specimens are used in the nodes, and locally advanced disease.
diagnosis of these nodules. Mammography is rarely indicated
because of its decreased sensitivity during pregnancy and lac- Phyllodes Tumors
tation; however, the fetus can be shielded if mammography is The nomenclature, presentation, and diagnosis of phyllodes
needed. Approximately 30% of the benign conditions encoun- tumors (including cystosarcoma phyllodes) have posed many
tered will be unique to pregnancy and lactation (galactoceles, problems for surgeons.371 These tumors are classified as benign,
lobular hyperplasia, lactating adenoma, and mastitis or abscess). borderline, or malignant. Borderline tumors have a greater
Once a breast cancer is diagnosed, complete blood count, chest potential for local recurrence.
radiography (with shielding of the abdomen), and liver function Mammographic evidence of calcifications and morpho-
studies are performed. logic evidence of necrosis do not distinguish between benign,
Because of the potential deleterious effects of radiation borderline, and malignant phyllodes tumors. Consequently, it
therapy on the fetus, radiation cannot be considered until the is difficult to differentiate benign phyllodes tumors from the
malignant variant and from fibroadenomas. Phyllodes tumors malignant elements is made, reexcision of the biopsy specimen 601
are usually sharply demarcated from the surrounding breast site to ensure complete excision of the tumor with a 1-cm mar-
tissue, which is compressed and distorted. Connective tissue gin of normal-appearing breast tissue is indicated. Large phyl-
composes the bulk of these tumors, which have mixed gelati- lodes tumors may require mastectomy. Axillary dissection is not
nous, solid, and cystic areas. Cystic areas represent sites of recommended because axillary lymph node metastases rarely
infarction and necrosis. These gross alterations give the gross occur.
cut tumor surface its classical leaf-like (phyllodes) appearance.
The stroma of a phyllodes tumor generally has greater cellular
Inflammatory Breast Carcinoma
Inflammatory breast carcinoma (stage IIIB) accounts for <3% of
activity than that of a fibroadenoma. After microdissection to
breast cancers. This cancer is characterized by the skin changes
harvest clusters of stromal cells from fibroadenomas and from
of brawny induration, erythema with a raised edge, and edema

CHAPTER 17 THE BREAST


phyllodes tumors, molecular biology techniques have shown the
(peau d’orange).372 Permeation of the dermal lymph vessels by
stromal cells of fibroadenomas to be either polyclonal or mono-
cancer cells is seen in skin biopsy specimens. There may be
clonal (derived from a single progenitor cell), whereas those of
an associated breast mass (Fig. 17-39). The clinical differentia-
phyllodes tumors are always monoclonal.
tion of inflammatory breast cancer may be extremely difficult,
Most malignant phyllodes tumors (Fig. 17-38) contain
especially when a locally advanced scirrhous carcinoma invades
liposarcomatous or rhabdomyosarcomatous elements rather than
dermal lymph vessels in the skin to produce peau d’orange and
fibrosarcomatous elements. Evaluation of the number of mitoses
lymphangitis (Table 17-15). Inflammatory breast cancer also
and the presence or absence of invasive foci at the tumor mar-
may be mistaken for a bacterial infection of the breast. More
gins may help to identify a malignant tumor. Small phyllodes
than 75% of women who have inflammatory breast cancer
tumors are excised with a margin of normal-appearing breast
present with palpable axillary lymphadenopathy, and distant
tissue. When the diagnosis of a phyllodes tumor with suspicious
metastases also are frequently present. A PET-CT scan should
be considered at the time of diagnosis to rule out concurrent
metastatic disease. A report of the SEER program described
distant metastases at diagnosis in 25% of white women with
inflammatory breast carcinoma.
Surgery alone and surgery with adjuvant radiation therapy
have produced disappointing results in women with inflamma-
tory breast cancer. However, neoadjuvant chemotherapy with an
anthracycline-containing regimen may affect dramatic regres-
sions in up to 75% of cases. Tumors should be assessed for
HER2 and hormone receptors with treatment dictated based on
receptor status. Modified radical mastectomy is performed after
demonstrated response to systemic therapy to remove residual
cancer from the chest wall and axilla. Adjuvant chemotherapy
may be indicated depending on final pathologic assessment of
the breast and regional nodes. Finally, the chest wall and the
A

Figure 17-38. A. Malignant phyllodes tumor (cystosarcoma- Figure 17-39. Inflammatory breast carcinoma. Stage IIIB cancer
phyllodes). B. Histologic features of a malignant phyllodes tumor of the breast with erythema, skin edema (peau d’orange), nipple
(hematoxylin and eosin stain, ×100). retraction, and satellite skin nodules.
602 Table 17-15
biopsy proven lymph node involvement. Angiosarcomas are
classified as de novo, as postradiation, or as arising in associa-
Inflammatory vs. noninflammatory breast cancer tion with postmastectomy lymphedema. In 1948, Stewart and
Treves described lymphangiosarcoma of the upper extremity in
INFLAMMATORY NONINFLAMMATORY women with ipsilateral lymphedema after radical mastectomy.374
Dermal lymph vessel invasion Inflammatory changes are Angiosarcoma is now the preferred name. The average interval
is present with or without present without dermal between modified radical or radical mastectomy and the devel-
inflammatory changes. lymph vessel invasion. opment of an angiosarcoma is 7 to 10 years. Sixty percent of
Cancer is not sharply Cancer is better delineated. women developing this cancer have a history of adjuvant radia-
delineated. tion therapy. Forequarter amputation may be necessary to palli-
ate the ulcerative complications and advanced lymphedema.
Erythema and edema Erythema is usually confined
PART II

frequently involve >33% to the lesion, and edema is Lymphomas. Primary lymphomas of the breast are rare, and
of the skin over the breast. less extensive. there are two distinct clinicopathologic variants. One type occurs
in women ≤39 years of age, is frequently bilateral, and has the
Lymph node involvement is Lymph nodes are involved in
histologic features of Burkitt’s lymphoma. The second type is
present in >75% of cases. approximately 50% of the
seen in women ≥40 years of age and is usually of the B-cell type.
cases.
SPECIFIC CONSIDERATIONS

Breast involvement by Hodgkin’s lymphoma has been reported.


Distant metastases are more Distant metastases are less An occult breast lymphoma may be diagnosed after detection of
common at the initial common at presentation.  palpable axillary lymphadenopathy. Treatment depends on the
presentation (25% of stage of disease. Lumpectomy or mastectomy may be required.
cases). Axillary dissection for clearance of disease may be necessary.
Modified with permission from Bland KI, Copeland ED: The Breast: Recurrent or progressive local-regional disease is best man-
Comprehensive Management of Benign and Malignant Diseases, 2nd ed. aged by chemotherapy and radiation therapy. The prognosis is
Philadelphia, PA: Elsesvier/Saunders; 1998. favorable, with 5- and 10-year survival rates of 74% and 51%,
respectively. More recently anaplastic large cell lymphoma has
supraclavicular, internal mammary, and axillary lymph node been described in association with breast implants for cosmetic
basins receive adjuvant radiation therapy. This multimodal or reconstructive purposes. This disease is treated with complete
approach results in 5-year survival rates that approach 30%. excision of the implant capsule with any associated soft tissue
Patients with inflammatory breast cancer should be encouraged mass. More advanced cases may require systemic therapy and
to participate in clinical trials. radiation treatment.

Rare Breast Cancers


Squamous Cell (Epidermoid) Carcinoma. Squamous REFERENCES
cell (epidermoid) carcinoma is a rare cancer that arises from Entries highlighted in bright blue are key references.
metaplasia within the duct system and generally is devoid of 1. Breasted JH. The Edwin Smith Surgical Papyrus. University
distinctive clinical or radiographic characteristics.373 Regional of Chicago Press, 1930;405.
metastases occur in 25% of patients, whereas distant metastases 2. Celsus AC. De Medicina (ed Loeb Classical Library Ed).
are rare. Cambridge: Harvard University Press; 1935;131.
3. Beenken SW. History of the therapy of breast cancer. In:
Adenoid Cystic Carcinoma. Adenoid cystic carcinoma is very
Copeland BA, ed. The Breast: Comprehensive Manage-
rare, accounting for <0.1% of all breast cancers. It is typically
ment of Benign and Malignant Disorder. Philadelphia:
indistinguishable from adenoid cystic carcinoma arising in sali- Saunders;2004;5.
vary tissues. These cancers are generally 1 to 3 cm in diameter 4. Le Dran F. Mémoire avec une précis de plusieurs observa-
at presentation and are well circumscribed. Axillary lymph node tions sur le. Mem Acad Roy Chir Paris. 1757;3:1.
metastases are rare, but deaths from pulmonary metastases have 5. Moore C. On the influence of inadequate operations on the
been reported. theory of cancer. R Med Chir Soc. 1867;1:244.
Apocrine Carcinomas. Apocrine carcinomas are well- 6. Halsted WS I. The results of operations for the cure of cancer
of the breast performed at the Johns Hopkins Hospital from
differentiated cancers that have rounded vesicular nuclei and
June, 1889, to January, 1894. Ann Surg. 1894;20:497-555.
prominent nucleoli. There is a very low mitotic rate and little
7. Haagensen CD, Stout AP. Carcinoma of the breast. II-criteria
variation in cellular features. However, apocrine carcinomas of operability. Ann Surg. 1943;118:1032-1051.
may display an aggressive growth pattern. 8. Patey DH, Dyson WH. The prognosis of carcinoma of the
Sarcomas. Sarcomas of the breast are histologically similar breast in relation to the type of operation performed. Br J
to soft tissue sarcomas at other anatomic sites. This diverse Cancer. 1948;2:7-13.
group includes fibrosarcoma, malignant fibrous histiocytoma, 9. Fisher B, Jeong JH, Anderson S, et al. Twenty-five-year
liposarcoma, leiomyosarcoma, malignant schwannoma, rhab- follow-up of a randomized trial comparing radical mas-
tectomy, total mastectomy, and total mastectomy followed
domyosarcoma, osteogenic sarcoma, and chondrosarcoma. The
by irradiation. N Engl J Med. 2002;347:567-575.
clinical presentation is typically that of a large, painless breast
10. Keynes G. Conservative treatment of cancer of the breast.
mass with rapid growth. Diagnosis is by core-needle biopsy or by Br Med J. 1937;2(3):643-666.
open incisional biopsy. Sarcomas are graded based on cellular- 11. Fisher B, Anderson S, Bryant J, et al. Twenty-year follow-up
ity, degree of differentiation, nuclear atypia, and mitotic activity. of a randomized trial comparing total mastectomy, lumpec-
Primary treatment is wide local excision, which may necessitate tomy, and lumpectomy plus irradiation for the treatment of
mastectomy. Axillary dissection is not indicated unless there is invasive breast cancer. N Engl J Med. 2002;347:1233-1241.
12. Clarke M, Collins R, Darby S, et al. Effects of radiotherapy 34. Haagensen CD. Diseases of the Breast. 3rd ed. Philadelphia: 603
and of differences in the extent of surgery for early breast WB Saunders; 1986.
cancer on local recurrence and 15-year survival: an overview 35. Haagensen CD, Lane N, Lattes R, et al. Lobular neoplasia
of the randomised trials. Lancet. 2005;366:2087-2106. (so-called lobular carcinoma in situ) of the breast. Cancer.
13. Peto R, Davies C, Godwin J, et al. Comparisons between dif- 1978;42:737-769.
ferent polychemotherapy regimens for early breast cancer: 36. Gadd MA, Souba WW. Evaluation and treatment of benign
meta-analyses of long-term outcome among 100,000 women breast disorders. In: Bland KI, Copeland EMI, eds. The
in 123 randomised trials. Lancet. 2012;379:432-444. Breast: Comprehensive Management of Benign and Malig-
14. Davies C, Godwin J, Gray R, et al. Relevance of breast cancer nant Diseases. Philadelphia: WB Saunders; 1998:233.
hormone receptors and other factors to the efficacy of adju- 37. Marchant DJ. Benign breast disease. Obstet Gynecol Clin
vant tamoxifen: patient-level meta-analysis of randomised North Am. 2002;29:1-20.

CHAPTER 17 THE BREAST


trials. Lancet. 2011;378:771-784. 38. Nurko J, Mabry CD, Whitworth P, et al. Interim results from
15. Sorlie T, Perou CM, Tibshirani R, et al. Gene expression the FibroAdenoma Cryoablation Treatment Registry. Am J
patterns of breast carcinomas distinguish tumor subclasses Surg. 2005;190:647-651; discussion 651-652.
with clinical implications. Proc Natl Acad Sci U S A. 39. Dixon JM. Conservative management of fibroadenoma of the
2001;98:10869-10874. breast. Br J Surg. 1996;83:1798-1799.
16. Bland KI, Romrell LJ. Congenital and acquired disturbances 40. Atkins HJ. Mammillary fistula. Br Med J. 1955;2:1473-1474.
of breast development and growth. In: Bland KI, Cope- 41. Dixon JM, Thompson AM. Effective surgical treatment for
land EMI, eds. The Breast: Comprehensive Management of mammary duct fistula. Br J Surg. 1991;78:1185-1186.
Benign and Malignant Diseases. Philadelphia: WB Saunders; 42. Bernstein L, Henderson BE, Hanisch R, et al. Physical exer-
1998:214. cise and reduced risk of breast cancer in young women. J
17. Lonnerdal B. Nutritional and physiologic significance of Natl Cancer Inst. 1994;86:1403-1408.
human milk proteins. Am J Clin Nutr. 2003;77:1537S-1543S. 43. Blackburn GL, Copeland T, Khaodhiar L, et al. Diet and
18. Rosenbloom AL. Breast physiology: normal and abnormal breast cancer. J Womens Health (Larchmt). 2003;12:
development and function. In: Bland KI, Copeland EMI, 183-192.
eds. The Breast: Comprehensive Management of Benign and 44. Goss PE, Sierra S. Current perspectives on radiation-induced
Malignant Diseases. Philadelphia: WB Saunders; 1998:38. breast cancer. J Clin Oncol. 1998;16:338-347.
19. Van de Perre P. Transfer of antibody via mother’s milk. 45. Hulka BS. Epidemiologic analysis of breast and gynecologic
Vaccine. 2003;21(24):3374-3376. cancers. Prog Clin Biol Res. 1997;396:17-29.
20. Bland KI, Graves TA. Gynecomastia. In: Bland KI, Copeland 46. Pujol P, Galtier-Dereure F, Bringer J: Obesity and breast
EMI, eds. The Breast: Comprehensive Management of cancer risk. Hum Reprod. 1997;12(1):116-125.
Benign and Malignant Diseases. Philadelphia: WB Saunders; 47. Singletary SE. Rating the risk factors for breast cancer. Ann
1998:153. Surg. 2003;237:474-482.
21. Dixon JM. Outpatient treatment of non-lactational breast 48. Wynder EL, Cohen LA, Muscat JE, et al. Breast cancer:
abscesses. Br J Surg. 1992;79:56-57. weighing the evidence for a promoting role of dietary fat.
22. Furlong AJ, al-Nakib L, Knox WF, et al. Periductal inflamma- J Natl Cancer Inst. 1997;89:766-775.
tion and cigarette smoke. J Am Coll Surg. 1994;179:417-420. 49. Baan R, Straif K, Grosse Y, et al. Carcinogenicity of alcoholic
23. Zuska JJ, Crile G, Jr., Ayres WW. Fistulas of lactifierous beverages. Lancet Oncol. 2007;8:292-293.
ducts. Am J Surg. 1951;81:312-317. 50. Howlader N, Noone AM, Krapcho M, et al. SEER Cancer
24. Dixon JM. Infection in surgical practice. In: Taylor EW, Statistics Review, 1975-2010, National Cancer Institute, based
ed. Breast Surgery. Oxford: Oxford Medical Publications; on November 2012 SEER data submission. Bethesda; 2013.
1992;187. 51. Domchek SM, Eisen A, Calzone K, et al. Application of
25. Dixon JM. Periductal mastitis and duct ectasia: an update. breast cancer risk prediction models in clinical practice. J
Breast. 1998;7:128-130. Clin Oncol. 2003;21:593-601.
26. Dixon JM, Kohlhardt SR, Dillon P. Total duct excision. 52. Gail MH, Brinton LA, Byar DP, et al. Projecting individu-
Breast. 1998;7:216-219. alized probabilities of developing breast cancer for white
27. Frykberg ER, Bland KI. Current concepts on the biology and females who are being examined annually. J Natl Cancer
management of in situ (Tis, stage 0) breast carcinoma. In: Inst. 1989;81:1879-1886.
Bland KI, Copeland EMI, eds. The Breast: Comprehensive 53. Edwards BK, Brown ML, Wingo PA, et al. Annual report
Management of Benign and Malignant Diseases. Philadel- to the nation on the status of cancer, 1975-2002, featuring
phia: WB Saunders; 1998:1020. population-based trends in cancer treatment. J Natl Cancer
28. Camiel MR. Mondor’s disease in the breast. Am J Obstet Inst. 2005;97:1407-1427.
Gynecol. 1985;152:879-881. 54. Chen J, Pee D, Ayyagari R, et al. Projecting absolute invasive
29. Mondor H. Tronculite sous-cutanée subaiguë de la paroi breast cancer risk in white women with a model that includes
thoracique antero-latérale. Mem Acad Chir Paris. 1939; mammographic density. J Natl Cancer Inst. 2006;98:1215-1226.
65:1271. 55. Claus EB, Risch N, Thompson WD. The calculation of breast
30. Hughes LE, Mansel RE, Webster DJ. Aberrations of normal cancer risk for women with a first degree family history of
development and involution (ANDI): a new perspective on ovarian cancer. Breast Cancer Res Treat. 1993;28:115-120.
pathogenesis and nomenclature of benign breast disorders. 56. Kerlikowske K, Ichikawa L, Miglioretti DL, et al. Longitudi-
Lancet. 1987;2:1316-1319. nal measurement of clinical mammographic breast density to
31. Archer F, Omar M. The fine structure of fibro adenoma of the improve estimation of breast cancer risk. J Natl Cancer Inst.
human breast. J Pathol. 1969;99:113-117. 2007;99:386-395.
32. Page DL, Anderson TJ. Diagnostic Histopathology of the 57. Berry DA, Parmigiani G, Sanchez J, et al. Probability of car-
Breast. Edinburgh: Churchill Livingstone; 1987. rying a mutation of breast-ovarian cancer gene BRCA1 based
33. Page DL, Simpson JF. Benign, high-risk, and premalignant on family history. J Natl Cancer Inst. 1997;89:227-238.
lesions of the breast. In: Bland KI, Copeland EMI, eds. The 58. Parmigiani G, Berry D, Aguilar O. Determining carrier prob-
Breast: Comprehensive Management of Benign and Malig- abilities for breast cancer-susceptibility genes BRCA1 and
nant Diseases. Philadelphia: WB Saunders; 1998:191. BRCA2. Am J Hum Genet. 1998;62:145-158.
604 59. Tyrer J, Duffy SW, Cuzick J. A breast cancer prediction screening: updated overview of the Swedish randomised trials.
model incorporating familial and personal risk factors. Stat Lancet. 2002;359:909-919.
Med. 2004;23:1111-1130. 81. Moss S, Thomas I, Evans A, Thomas B, Johns, L. Ran-
60. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxi- domised controlled trial of mammographic screening in
fen for prevention of breast cancer: report of the National women from age 40: results of screening in the first 10 years.
Surgical Adjuvant Breast and Bowel Project P-1 Study. J Br J Cancer. 2005;92:949-954.
Natl Cancer Inst. 1998;90:1371-1388. 82. Veronesi U, Maisonneuve P, Sacchini V, Rotmensz N, Boyle
61. Grodstein F, Stampfer MJ, Colditz GA, et al. Postmeno- P. Tamoxifen for breast cancer among hysterectomised
pausal hormone therapy and mortality. N Engl J Med. women. Lancet. 2002;359:1122-1124.
1997;336:1769-1775. 83. Cuzick J, Forbes JF, Sestak I, et al. Long-term results of
62. Hartmann LC, Schaid DJ, Woods JE, et al. Efficacy of bilat- tamoxifen prophylaxis for breast cancer–96-month follow-up
eral prophylactic mastectomy in women with a family history of the randomized IBIS-I trial. J Natl Cancer Inst. 2007;99:
of breast cancer. N Engl J Med. 1999;340:77-84. 272-282.
PART II

63. Kerlikowske K, Grady D, Rubin SM, et al. Efficacy of screening 84. Gail MH, Costantino JP, Bryant J, et al. Weighing the risks and
mammography. A meta-analysis. JAMA. 1995;273:149-154. benefits of tamoxifen treatment for preventing breast cancer.
64. Rowe TC, Chen GL, Hsiang YH, et al. DNA damage by anti- J Natl Cancer Inst. 1999;91:1829-1846.
tumor acridines mediated by mammalian DNA topoisomer- 85. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of
ase II. Cancer Res. 1986;46:2021-2026. tamoxifen vs. raloxifene on the risk of developing inva-
SPECIFIC CONSIDERATIONS

65. Sakorafas GH. The management of women at high risk for sive breast cancer and other disease outcomes: the NSABP
the development of breast cancer: risk estimation and preven- Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA.
tative strategies. Cancer Treat Rev. 2003;29:79-89. 2006;295:2727-2741.
66. Schrag D, Kuntz KM, Garber JE, et al. Decision analysis— 86. Vogel VG, Costantino JP, Wickerham DL, et al. Update of the
effects of prophylactic mastectomy and oophorectomy on life National Surgical Adjuvant Breast and Bowel Project Study
expectancy among women with BRCA1 or BRCA2 mutations. of Tamoxifen and Raloxifene (STAR) P-2 Trial: Preventing
N Engl J Med. 1997;336:1465-1471. breast cancer. Cancer Prev Res (Phila). 2010;3:696-706.
67. Vogel VG. Management of the high-risk patient. Surg Clin 87. Goss PE, Ingle JN, Ales-Martinez JE, et al. Exemestane for
North Am. 2003;83:733-751. breast-cancer prevention in postmenopausal women. N Engl
68. Wu K, Brown P. Is low-dose tamoxifen useful for the treat- J Med. 2011;364:2381-2391.
ment and prevention of breast cancer? J Natl Cancer Inst. 88. Cuzick J. IBIS II: a breast cancer prevention trial in post-
2003;95:766-767. menopausal women using the aromatase inhibitor anastro-
69. Collaborative Group on Hormonal Factors in Breast Cancer. zole. Expert Rev Anticancer Ther. 2008;8:1377-1385.
Breast cancer and hormone replacement therapy: collabora- 89. Sestak I, Singh S, Cuzick J, et al. Changes in bone mineral
tive reanalysis of data from 51 epidemiological studies of density at 3 years in postmenopausal women receiving anas-
52,705 women with breast cancer and 108,411 women with- trozole and risedronate in the IBIS-II bone substudy: an inter-
out breast cancer. Lancet. 1997;350:1047-1059. national, double-blind, randomised, placebo-controlled trial.
70. Chlebowski RT, Hendrix SL, Langer RD, et al. Influence of Lancet Oncol. 2014;15:1460-1468.
estrogen plus progestin on breast cancer and mammography 90. Jenkins VA, Ambroisine LM, Atkins L, et al. Effects of anas-
in healthy postmenopausal women: the Women’s Health Ini- trozole on cognitive performance in postmenopausal women:
tiative Randomized Trial. JAMA. 2003;289:3243-3253. a randomised, double-blind chemoprevention trial (IBIS II).
71. Beral V. Breast cancer and hormone-replacement therapy in Lancet Oncol. 2008;9:953-961.
the Million Women Study. Lancet. 2003;362:419-427. 91. Nelson HD, Smith ME, Griffin JC, et al. Use of medications
72. Glasziou P, Houssami N. The evidence base for breast cancer to reduce risk for primary breast cancer: a systematic review
screening. Prev Med. 2011;53:100-102. for the U.S. Preventive Services Task Force. Ann Intern Med.
73. Olsen O, Gotzsche PC. Cochrane review on screening for breast 2013;158:604-614.
cancer with mammography. Lancet. 2001;358:1340-1342. 92. Visvanathan K, Hurley P, Bantug E, et al. Use of pharmaco-
74. Siu AL. Screening for Breast Cancer: U.S. Preventive Ser- logic interventions for breast cancer risk reduction: american
vices Task Force recommendation statement. Ann Intern society of clinical oncology clinical practice guideline. J Clin
Med. 2016;164:279-296. Oncol. 2013;31:2942-2962.
75. Oeffinger KC, Fontham ET, Etzioni R, et al. Breast can- 93. Domchek SM, Friebel TM, Singer CF, et al. Association
cer screening for women at average risk: 2015 guide- of risk-reducing surgery in BRCA1 or BRCA2 mutation
line update from the American Cancer Society. JAMA. carriers with cancer risk and mortality. JAMA. 2010;304:
2015;314:1599-1614. 967-975.
76. National Comprehensive Cancer Network. Breast cancer 94. Ford D, Easton DF, Stratton M, et al. Genetic heterogeneity
screening and diagnosis, version 1.2016. Available at: https:// and penetrance analysis of the BRCA1 and BRCA2 genes in
www.nccn.org/professionals/physician_gls/pdf/breast- breast cancer families. The Breast Cancer Linkage Consor-
screening.pdf. Accessed July 9, 2018. tium. Am J Hum Genet. 1998;62:676-689.
77. US Preventive Services Task Force. Screening for breast 95. Gowen LC, Avrutskaya AV, Latour AM, et al. BRCA1
cancer: US Preventive Services Task Force recommenda- required for transcription-coupled repair of oxidative DNA
tion statement. Ann Intern Med. 2009;151:716-726, W-236. damage. Science. 1998;281:1009-1012.
78. Powles TJ, Ashley S, Tidy A, Smith IE, Dowsett M. Twenty- 96. Martin AM, Weber BL. Genetic and hormonal risk factors in
year follow-up of the Royal Marsden randomized, double- breast cancer. J Natl Cancer Inst. 2000;92:1126-1135.
blinded tamoxifen breast cancer prevention trial. J Natl 97. Oddoux C, Struewing JP, Clayton CM, et al. The carrier
Cancer Inst. 2007;99:283-290. frequency of the BRCA2 6174delT mutation among Ash-
79. Independent UK Panel on Breast Cancer Screening. The ben- kenazi Jewish individuals is approximately 1%. Nat Genet.
efits and harms of breast cancer screening: an independent 1996;14:188-190.
review. Lancet. 2012;380:1778-1786. 98. Roa BB, Boyd AA, Volcik K, et al. Ashkenazi Jewish pop-
80. Nyström L, Andersson I, Bjurstam N, Frisell J, Norden- ulation frequencies for common mutations in BRCA1 and
skjöld B, Rutqvist LE. Long-term effects of mammography BRCA2. Nat Genet. 1996;14:185-187.
99. Rosen EM, Fan S, Pestell RG, et al. BRCA1 gene in breast 120. National Cancer Institute. SEER Cancer Statistics Review, 605
cancer. J Cell Physiol. 2003;196:19-41. 1975-2002. Available at: https://seer.cancer.gov/archive/
100. Wooster R, Weber BL. Breast and ovarian cancer. N Engl J csr/1975_2002/. Accessed July 8, 2018.
Med. 2003;348:2339-2347. 121. Fregene A, Newman LA. Breast cancer in sub-Saharan
101. Petrij-Bosch A, Peelen T, van Vliet M, et al. BRCA1 genomic Africa: how does it relate to breast cancer in African-Ameri-
deletions are major founder mutations in Dutch breast cancer can women? Cancer. 2005;103:1540-1550.
patients. Nat Genet. 1997;17:341-345. 122. Bloom HJ, Richardson WW, Harries EJ. Natural history
102. Gorski B, Byrski T, Huzarski T, et al. Founder mutations in of untreated breast cancer (1805-1933). Comparison of
the BRCA1 gene in Polish families with breast-ovarian can- untreated and treated cases according to histological grade of
cer. Am J Hum Genet. 2000;66:1963-1968. malignancy. Br Med J. 1962;2:213-221.
103. Sarantaus L, Huusko P, Eerola H, et al. Multiple founder 123. Early Breast Cancer Trialists’ Collaborative Group. Tamoxi-

CHAPTER 17 THE BREAST


effects and geographical clustering of BRCA1 and BRCA2 fen for early breast cancer. an overview of the randomised
families in Finland. Eur J Hum Genet. 2000;8:757-763. trials. Lancet. 1998;351:1451-1467.
104. Gayther SA, Harrington P, Russell P, et al. Frequently 124. Saphner T, Tormey DC, Gray R. Annual hazard rates of recur-
occurring germ-line mutations of the BRCA1 gene in rence for breast cancer after primary therapy. J Clin Oncol.
ovarian cancer families from Russia. Am J Hum Genet. 1996;14:2738-2746.
1997;60:1239-1242. 125. Sestak I, Dowsett M, Zabaglo L, et al. Factors predicting late
105. Szabo CI, King MC. Population genetics of BRCA1 and recurrence for estrogen receptor-positive breast cancer. J Natl
BRCA2. Am J Hum Genet. 1997;60:1013-1020. Cancer Inst. 2013;105:1504-1511.
106. Abeliovich D, Kaduri L, Lerer I, et al. The founder muta- 126. Gonzalez-Angulo AM, Cristofanilli M, Strom EA, et al.
tions 185delAG and 5382insC in BRCA1 and 6174delT in Central nervous system metastases in patients with high-
BRCA2 appear in 60% of ovarian cancer and 30% of early- risk breast carcinoma after multimodality treatment. Cancer.
onset breast cancer patients among Ashkenazi women. Am J 2004;101:1760-1766.
Hum Genet. 1997;60:505-514. 127. Carey LA, Ewend MG, Metzger R, et al. Central nervous
107. Johannesdottir G, Gudmundsson J, Bergthorsson JT, et al. system metastases in women after multimodality ther-
High prevalence of the 999del5 mutation in Icelandic breast apy for high risk breast cancer. Breast Cancer Res Treat.
and ovarian cancer patients. Cancer Res. 1996;56:3663-3665. 2004;88:273-280.
108. Hakansson S, Johannsson O, Johansson U, et al. Moder- 128. Evans AJ, James JJ, Cornford EJ, et al. Brain metastases from
ate frequency of BRCA1 and BRCA2 germ-line mutations breast cancer: identification of a high-risk group. Clin Oncol
in Scandinavian familial breast cancer. Am J Hum Genet. (R Coll Radiol). 2004;16:345-349.
1997;60:1068-1078. 129. Broders AC. Carcinoma in situ contrasted with benign pen-
109. Infante M, Duran M, Acedo A, et al. The highly prevalent BRCA2 etrating epithelium. JAMA. 1932;99:1670.
mutation c.2808_2811del (3036delACAA) is located in a 130. Foote FW, Stewart FW. Lobular carcinoma in situ: a rare form
mutational hotspot and has multiple origins. Carcinogenesis. of mammary cancer. Am J Pathol. 1941;17:491-496.
2013;34:2505-2511. 131. Consensus conference on the classification of ductal carci-
110. Warner E, Foulkes W, Goodwin P, et al. Prevalence and pen- noma in situ. Hum Pathol. 1997; 28:1221-1225.
etrance of BRCA1 and BRCA2 gene mutations in unselected 132. Recht A, Rutgers EJ, Fentiman IS, et al. The fourth EORTC
Ashkenazi Jewish women with breast cancer. J Natl Cancer DCIS Consensus meeting (Chateau Marquette, Heemskerk,
Inst. 1999;91:1241-1247. The Netherlands, 23-24 January 1998)–conference report.
111. Khatcheressian JL, Hurley P, Bantug E, et al. Breast can- Eur J Cancer. 1998;34:1664-1669.
cer follow-up and management after primary treatment: 133. Dunnwald LK, Rossing MA, Li CI. Hormone receptor status,
American Society of Clinical Oncology clinical practice tumor characteristics, and prognosis: a prospective cohort of
guideline update. J Clin Oncol. 2013;31:961-965. breast cancer patients. Breast Cancer Res. 2007;9:R6.
112. Schneider KA. Genetic counseling for BRCA1/BRCA2 test- 134. McDivitt RW, Boyce W, Gersell D. Tubular carcinoma of the
ing. Genet Test. 1997;1:91-98. breast. Clinical and pathological observations concerning 135
113. Kriege M, Brekelmans CT, Boetes C, et al. Efficacy of MRI cases. Am J Surg Pathol. 1982;6:401-411.
and mammography for breast-cancer screening in women 135. Jatoi I. Screening clinical breast examination. Surg Clin
with a familial or genetic predisposition. N Engl J Med. North Am. 2003;83:789-801.
2004;351:427-437. 136. Rosato FE, Rosato EL. Examination techniques: roles of the
114. Saslow D, Boetes C, Burke W, et al. American Cancer Soci- physician and patient evaluating breast diseases. In: Bland KI,
ety guidelines for breast screening with MRI as an adjunct to Copeland EMI, eds. The Breast: Comprehensive Management
mammography. CA Cancer J Clin. 2007; 57:75-89. of Benign and Malignant Diseases. Philadelphia: WB Saunders;
115. King MC, Wieand S, Hale K, et al. Tamoxifen and breast 1998:615.
cancer incidence among women with inherited mutations in 137. Bassett LW. Breast imaging. In: Bland KI, Copeland EMI,
BRCA1 and BRCA2: National Surgical Adjuvant Breast and eds. The Breast: Comprehensive Management of Benign
Bowel Project (NSABP-P1) Breast Cancer Prevention Trial. and Malignant Diseases. Philadelphia: WB Saunders;
JAMA. 2001;286:2251-2256. 1998:648.
116. Antoniou AC, Foulkes WD, Tischkowitz M. Breast-cancer 138. Fletcher SW, Elmore JG. Clinical practice. Mammographic
risk in families with mutations in PALB2. N Engl J Med. screening for breast cancer. N Engl J Med. 2003;348:1672-1680.
2014;371(17):497-506. 139. Miller AB. Screening and detection. In: Bland KI, Cope-
117. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA land EMI, eds. The Breast: Comprehensive Management of
Cancer J Clin. 2016;66:7-30. Benign and Malignant Diseases. Philadelphia: WB Saunders;
118. Clarke CA, Glaser SL, Uratsu CS, et al. Recent declines in hor- 1998:625.
mone therapy utilization and breast cancer incidence: clinical 140. Schnall MD. Breast MR imaging. Radiol Clin North Am.
and population-based evidence. J Clin Oncol. 2006;24:e49-e50. 2003;41:43-50.
119. Ferlay J, Bray F, Pisani P, Parkin DM. Globocan 2002: 141. Seidman H, Gelb SK, Silverberg E, et al. Survival experience
Cancer Incidence, Mortality and Prevalence Worldwide. in the Breast Cancer Detection Demonstration Project. CA
Lyon, France: IARC Press; 2004. Cancer J Clin. 1987;37:258-290.
606 142. Bevers T, Bibbins-Domingo K, Oeffinger KC, Smith ML. 161. Giuliano AE, et al. Breast cancer—major changes in the
Controversies in breast cancer screening strategies. J Natl American Joint Committee on Cancer eighth edition cancer
Compr Canc Netw. 2016;14:651-653. staging manual. CA Cancer J Clin. 2017;67:290-303.
143. Pisano ED, Gatsonis C, Hendrick E, et al. Diagnostic perfor- 162. Du Toit RS, Locker AP, Ellis IO, Elston CW, Blamey RW.
mance of digital versus film mammography for breast-cancer Evaluation of the prognostic value of triple node biopsy in
screening. N Engl J Med. 2005;353:1773-1783. early breast cancer. Br J Surg. 77:163-167.
144. Helvie MA. Digital mammography imaging: breast tomo- 163. Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL,
synthesis and advanced applications. Radiol Clin North Am. Trotti A, eds. AJCC Cancer Staging Manual. 7th ed. France:
2010;48:917-929. Springer; 2010.
145. Friedewald SM, Rafferty EA, Rose SL, et al. Breast cancer 164. Dillon DA. Molecular markers in the diagnosis and staging
screening using tomosynthesis in combination with digital of breast cancer. Semin Radiat Oncol. 2002;12:305-318.
mammography. JAMA. 2014;311:2499-2507. 165. Esteva FJ, Sahin AA, Cristofanilli M, et al. Molecular prog-
146. Bernardi D, Macaskill P, Pellegrini M, et al. Breast cancer nostic factors for breast cancer metastasis and survival. Semin
PART II

screening with tomosynthesis (3D mammography) with Radiat Oncol. 2002;12:319-328.


acquired or synthetic 2D mammography compared with 2D 166. Haffty BG. Molecular and genetic markers in the localre-
mammography alone (STORM-2): a population-based pro- gional management of breast cancer. Semin Radiat Oncol.
spective study. Lancet Oncol. 2016;17(8):1105-1113. 2002;12:329-340.
147. Ciatto S, Houssami N, Bernardi D, et al. Integration of 3D 167. Morabito A, Magnani E, Gion M, et al. Prognostic and pre-
SPECIFIC CONSIDERATIONS

digital mammography with tomosynthesis for population dictive indicators in operable breast cancer. Clin Breast Can-
breast-cancer screening (STORM): a prospective comparison cer. 2003;3:381-390.
study. Lancet Oncol. 2013;14:583-589. 168. Rogers CE, Loveday RL, Drew PJ, et al. Molecular prog-
148. Helvie MA. Digital mammography imaging: breast tomo- nostic indicators in breast cancer. Eur J Surg Oncol.
synthesis and advanced applications. Radiol Clin North Am. 2002;28:467-478.
2010;48:917-929. 169. Rampaul R, Ellis IO, Robertson JFR. Prognostic indices in
149. Friedewald SM, Rafferty EA, Rose SL, et al. Breast cancer breast cancer. In: Autier BP, Adebamowo C, Anderson BO,
screening using tomosynthesis in combination with digital et al, eds. World Breast Cancer Report. iPRI:2012:
mammography. JAMA. 2014;311:2499-2507. 323-332.
150. Bernardi D, Macaskill P, Pellegrini M, et al. Breast cancer 170. Athanassiadou PP, Veneti SZ, Kyrkou KA, et al. Presence
screening with tomosynthesis (3D mammography) with of epidermal growth factor receptor in breast smears of cyst
acquired or synthetic 2D mammography compared with 2D fluids: relationship to electrolyte ratios and pH concentration.
mammography alone (STORM-2): a population-based pro- Cancer Detect Prev. 1992;16:113-118.
spective study. Lancet Oncol. 2016;17(8):1105-1113. 171. Tsutsumi Y, Naber SP, DeLellis RA, et al. Neu oncogene
151. Jochelson MS, Dershaw DD, Sung JS, et al. Bilateral con- protein and epidermal growth factor receptor are indepen-
trast-enhanced dual-energy digital mammography: feasibility dently expressed in benign and malignant breast tissues. Hum
and comparison with conventional digital mammography and Pathol. 1990;21:750-758.
MR imaging in women with known breast carcinoma. Radi- 172. van de Vijver MJ, Peterse JL, Mooi WJ, et al. Neu-protein
ology. 2013;266(3):743-751. overexpression in breast cancer. Association with com-
152. Jochelson MS, Dershaw DD, Sung JS, et al. Bilateral con- edotype ductal carcinoma in situ and limited prognostic
trast-enhanced dual-energy digital mammography: feasibility value in stage II breast cancer. N Engl J Med. 1988;319:
and comparison with conventional digital mammography and 1239-1245.
MR imaging in women with known breast carcinoma. Radi- 173. Slamon DJ, Clark GM, Wong SG, et al. Human breast cancer:
ology. 2013;266(3):743-751. correlation of relapse and survival with amplification of the
153. Krishnamurthy S, Sneige N, Bedi DG, et al. Role of ultra- HER-2/neu oncogene. Science. 1987;235:177-182.
sound-guided fine-needle aspiration of indeterminate and 174. Gusterson BA, Gelber RD, Goldhirsch A, et al. Prognostic
suspicious axillary lymph nodes in the initial staging of importance of c-erbB-2 expression in breast cancer. Interna-
breast carcinoma. Cancer. 2002;95:982-988. tional (Ludwig) Breast Cancer Study Group. J Clin Oncol.
154. Turnbull L, Brown S, Harvey I, et al. Comparative effective- 1992;10:1049-1056.
ness of MRI in breast cancer (COMICE) trial: a randomised 175. McCann AH, Dervan PA, O’Regan M, et al. Prognostic sig-
controlled trial. Lancet. 2010;375:563-571. nificance of c-erbB-2 and estrogen receptor status in human
155. Houssami N, Turner R, Morrow M. Preoperative magnetic breast cancer. Cancer Res. 1991;51:3296-3303.
resonance imaging in breast cancer: meta-analysis of surgical 176. Slamon DJ, Godolphin W, Jones LA, et al. Studies of the
outcomes. Ann Surg. 2013;257:249-255. HER-2/neu proto-oncogene in human breast and ovarian
156. Robinson DS, Sundaram M. Stereotactic imaging and breast cancer. Science. 1989;244:707-712.
biopsy. In: Bland KI, Copeland EMI, eds. The Breast: Com- 177. Wright C, Angus B, Nicholson S, et al. Expression of
prehensive Management of Benign and Malignant Diseases. c-erbB-2 oncoprotein: a prognostic indicator in human breast
Philadelphia: WB Saunders; 1998:698. cancer. Cancer Res. 1989;49:2087-2090.
157. Wilkinson EJ, Masood S. Cytologic needle samplings of the 178. Dabbs DJ, Klein ME, Mohsin SK, Tubbs RR, Shuai Y,
breast: techniques and end results. In: Bland KI, Copeland EMI, Bhargava R. High false-negative rate of HER2 quantita-
eds. The Breast: Comprehensive Management of Benign and tive reverse transcription polymerase chain reaction of the
Malignant Diseases. Philadelphia: WB Saunders; 1998:705. Oncotype DX test: an independent quality assurance study.
158. Greene FL, Page DL, Fleming ID, eds. Breast. AJCC Cancer J Clin Oncol. 2011;29(32):4279-4285.
Staging Manual. 6th ed. New York: Springer-Verlag: 2002. 179. Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and
159. Britton PD, Goud A, Godward S, et al. Use of ultrasound- safety of trastuzumab as a single agent in first-line treatment
guided axillary node core biopsy in staging of early breast of HER2-overexpressing metastatic breast cancer. J Clin
cancer. Eur Radiol. 2009;19:561-569. Oncol. 20;(3):719-726.
160. Fisher B, Slack NH. Number of lymph nodes examined and 180. Piccart-Gebhart MJ, Proctor M, Leyland-Jones B, et al.
the prognosis of breast carcinoma. Surg Gynecol Obstet. Trastuzumab after adjuvant chemotherapy in HER2-positive
1970;131:79-88. breast cancer. N Engl J Med. 2005;353(16):1659-1672.
181. Romond EH, Perez EA, Bryant J, et al. Trastuzumab therapy for women with early breast cancer. J Clin Oncol. 607
plus adjuvant chemotherapy for operable HER2-positive 2001;19:980-991.
breast cancer. N Engl J Med. 2005;353:1673-1684. 199. Blamey RW, Ellis IO, Pinder SE, et al. Survival of inva-
182. Slamon D, Eiermann W, Robert N, et al. Adjuvant trastu- sive breast cancer according to the Nottingham Prognos-
zumab in HER2-positive breast cancer. N Engl J Med. tic Index in cases diagnosed in 1990-1999. Eur J Cancer.
2011;365:1273-1283. 2007;43:1548-1555.
183. Smith I, Procter M, Gelber RD, et al. 2-year follow-up of 200. Blamey RW, Pinder SE, Ball GR, et al. Reading the prog-
trastuzumab after adjuvant chemotherapy in HER2-posi- nosis of the individual with breast cancer. Eur J Cancer.
tive breast cancer: a randomised controlled trial. Lancet. 2007;43:1545-1547.
2007;369:29-36. 201. Wishart GC, Azzato EM, Greenberg DC, et al. PREDICT:
184. Slamon D, Eiermann W, Robert N, et al. Adjuvant trastu- a new UK prognostic model that predicts survival follow-

CHAPTER 17 THE BREAST


zumab in HER2-positive breast cancer. N Engl J Med. ing surgery for invasive breast cancer. Breast Cancer Res.
2011;365:1273-1283. 2010;12:R1.
185. Dahabreh IJ, Linardou H, Siannis F, et al. Trastuzumab in the 202. Perou CM, Jeffrey SS, van de Rijn M, et al. Distinctive gene
adjuvant treatment of early-stage breast cancer: a systematic expression patterns in human mammary epithelial cells and
review and meta-analysis of randomized controlled trials. breast cancers. Proc Natl Acad Sci U S A. 1999;96:9212-9217.
Oncologist. 2008;13:620-630. 203. Paik S, Shak S, Tang G, et al. A multigene assay to pre-
186. Monaghan P, Perusinghe NP, Nicholson RI, et al. Growth fac- dict recurrence of tamoxifen-treated, node-negative breast
tor stimulation of proliferating cell nuclear antigen (PCNA) cancer. N Engl J Med. 2004:351:2817-2826.
in human breast epithelium in organ culture. Cell Biol Int 204. Sparano JA, Gray RJ, Makower DF, et al. Prospective valida-
Rep. 1991;15:561-570. tion of a 21-gene expression assay in breast cancer. N Engl J
187. Siitonen SM, Isola JJ, Rantala IS, et al. Intratumor varia- Med. 2015;373(21):2005-2014.
tion in cell proliferation in breast carcinoma as determined 205. Albain KS, Barlow WE, Shak S, et al. Prognostic and pre-
by antiproliferating cell nuclear antigen monoclonal anti- dictive value of the 21-gene recurrence score assay in
body and automated image analysis. Am J Clin Pathol. postmenopausal women with node-positive, oestrogen-
1993;99:226-231. receptor-positive breast cancer on chemotherapy: a ret-
188. Tuccari G, Rizzo A, Muscara M, et al. PCNA/cyclin expres- rospective analysis of a randomised trial. Lancet Oncol.
sion in breast carcinomas: its relationships with Ki-67, ER, 2010;11(1):55-65.
PgR immunostainings and clinico-pathologic aspects. Patho- 206. Lo SS, Mumby PB, Norton J, et al. Prospective multicenter
logica. 1993;85:47-55. study of the impact of the 21-gene recurrence score assay on
189. van Dierendonck JH, Wijsman JH, Keijzer R, et al. Cell- medical oncologist and patient adjuvant breast cancer treat-
cyclerelated staining patterns of anti-proliferating cell ment selection. J Clin Oncol. 2010;28(10):1671-1676.
nuclear antigen monoclonal antibodies. Comparison with 207. Drukker CA, Bueno-de-Mesquita JM, Retèl VP, et al. A
BrdUrd labeling and Ki-67 staining. Am J Pathol. 1991;138: prospective evaluation of a breast cancer prognosis sig-
1165-1172. nature in the observational RASTER study. Int J Cancer.
190. Cuzick J, Dowsett M, Pineda S, et al. Prognostic value of a 2013;133(4):929-936.
combined estrogen receptor, progesterone receptor, Ki-67, 208. Cardoso F, van’t Veer LI, Bogaerts J, et al. 70-gene signature
and human epidermal growth factor receptor 2 immunohis- as an aid to treatment decisions in early-stage breast cancer.
tochemical score and comparison with the Genomic Health N Engl J Med. 2016;375:717-729.
recurrence score in early breast cancer. J Clin Oncol. 2011;29: 209. Julien JP, Bijker N, Fentiman IS, et al. Radiotherapy in breast-
4273-4278. conserving treatment for ductal carcinoma in situ: first results
191. Brown LF, Berse B, Jackman RW, et al. Expression of vas- of the EORTC randomised phase III trial 10853. EORTC
cular permeability factor (vascular endothelial growth factor) Breast Cancer Cooperative Group and EORTC Radiotherapy
and its receptors in breast cancer. Hum Pathol. 1995;26:86-91. Group. Lancet. 2000;355:528-533.
192. Gasparini G, Toi M, Gion M, et al. Prognostic significance 210. Lagios MD, Margolin FR, Westdahl PR, et al. Mammo-
of vascular endothelial growth factor protein in node-negative graphically detected duct carcinoma in situ. Frequency of
breast carcinoma. J Natl Cancer Inst. 1997;89:139-147. local recurrence following tylectomy and prognostic effect of
193. Schneider BP, Gray RJ, Radovich M, et al. Prognostic and nuclear grade on local recurrence. Cancer. 1989;63:618-624.
predictive value of tumor vascular endothelial growth factor 211. Rosai J. Borderline epithelial lesions of the breast. Am J Surg
gene amplification in metastatic breast cancer treated with Pathol. 1991;15:209-221.
paclitaxel with and without bevacizumab; results from ECOG 212. Schnitt SJ, Connolly JL, Tavassoli FA, et al. Interobserver
2100 trial. Clin Cancer Res. 2013;19(5):1281-1289. reproducibility in the diagnosis of ductal proliferative
194. Miller K, Wang M, Gralow J, et al. Paclitaxel plus bevaci- breast lesions using standardized criteria. Am J Surg Pathol.
zumab versus paclitaxel alone for metastatic breast cancer. 1992;16:1133-1143.
N Engl J Med. 2007;357:2666-2676. 213. Silverstein MJ, Lagios MD, Groshen S, et al. The influence
195. Allan DJ, Howell A, Roberts SA, et al. Reduction in apop- of margin width on local control of ductal carcinoma in situ
tosis relative to mitosis in histologically normal epithelium of the breast. N Engl J Med. 1999;340:1455-1461.
accompanies fibrocystic change and carcinoma of the pre- 214. Tan-Chiu E. The effect of tamoxifen on benign breast dis-
menopausal human breast. J Pathol. 1992;167:25-32. ease: Findings from the National Surgical Adjuvant Breast
196. Bargou RC, Daniel PT, Mapara MY, et al. Expression of the and Bowel Project (NSABP) breast cancer prevention trial
bcl-2 gene family in normal and malignant breast tissue: low (BCPT). Breast Cancer Res Treat. 2001;69:abstract 7.
bax-alpha expression in tumor cells correlates with resistance 215. Fisher B, Dignam J, Wolmark N, et al. Lumpectomy and radi-
towards apoptosis. Int J Cancer. 1995;60:854-859. ation therapy for the treatment of intraductal breast cancer:
197. Binder C, Marx D, Binder L, Schaurer A, Hiddemann W. findings from National Surgical Adjuvant Breast and Bowel
Expression of Bax in relation to Bcl-2 and other predictive Project B-17. J Clin Oncol. 1998;16:441-452.
parameters in breast cancer. Ann Oncol. 1996;7:129-133. 216. Bijker N, Meijnen P, Peterse JL, et al. Breast-conserving
198. Ravdin PM, Siminoff LA, Davis GJ, et al. Computer treatment with or without radiotherapy in ductal carci-
program to assist in making decisions about adjuvant noma-in-situ: ten-year results of European Organisation for
608 Research and Treatment of Cancer randomized phase III 233. Veronesi U, Cascinelli N, Mariani L, et al. Twenty-year fol-
trial 10853–a study by the EORTC Breast Cancer Coopera- lowup of a randomized study comparing breast-conserving
tive Group and EORTC Radiotherapy Group. J Clin Oncol. surgery with radical mastectomy for early breast cancer.
2006;24:3381-3387. N Engl J Med. 2002;347:1227-1232.
217. Houghton J, George WD, Cuzick J, et al. Radiotherapy and 233a. Hughes KS, Schnaper LA, Berry D, et al: Lumpectomy plus
tamoxifen in women with completely excised ductal carcinoma tamoxifen with or without irradiation in women 70 years of
in situ of the breast in the UK, Australia, and New Zealand: ran- age or older with early breast cancer. N Engl J Med 351:971-
domised controlled trial. Lancet. 2003;362:95-102. 7, 2004.
218. Emdin SO, Granstrand B, Ringberg A, et al. SweDCIS: 234. Kunkler IH, Williams LJ, Jack WJ, Cameron DA, Dixon JM;
Radiotherapy after sector resection for ductal carcinoma PRIME II investigators. Breast-conserving surgery with or
in situ of the breast. Results of a randomised trial in a without irradiation in women aged 65 years or older with
population offered mammography screening. Acta Oncol. early breast cancer (PRIME II): a randomised controlled trial.
2006;45:536-543. Lancet Oncol. 2015;16(3):266-273.
PART II

219. Morrow M, Van Zee KJ, Solin LJ, et al. Society of Surgical Oncol- 235. Vaidya J, Wenz F, Bulsara M, et al. Abstract S4-2: Targeted
ogy–American Society for Radiation Oncology–American Soci- intraoperative radiotherapy for early breast cancer: TARGIT-
ety of Clinical Oncology consensus guideline on margins for A trial-updated analysis of local recurrence and first analysis
breast-conserving surgery with whole-breast irradiation in ductal of survival. Cancer Res. 2012;72:S4-2-S4-2.
carcinoma in situ. J Clin Oncol. 2016;34(33):4040-4046. 236. Corica T, Nowak AK, Saunders CM, et al. Cosmesis and breast-
SPECIFIC CONSIDERATIONS

220. Marinovich ML, Azizi L, Macaskill P, Irwig L, et al. The related quality of life outcomes after intraoperative radiation
Association of Surgical Margins and Local Recurrence in therapy for early breast cancer: a substudy of the TARGIT-A
Women with Ductal Carcinoma In Situ Treated with Breast- trial. Int J Radiat Oncol Biol Phys. 2016;96(1):55-64.
Conserving Therapy: A Meta-Analysis. Ann Surg Oncol. 237. Vaidya JS, Wenz F, Bulsara M, et al. Risk-adapted targeted
2016;23(12):3811-3821. intraoperative radiotherapy versus whole-breast radiotherapy
221. Mansel RE, Fallowfield L, Kissin M, et al. Randomized mul- for breast cancer: 5-year results for local control and over-
ticenter trial of sentinel node biopsy versus standard axillary all survival from the TARGIT-A randomised trial. Lancet.
treatment in operable breast cancer: the ALMANAC Trial. 2014;383:603-613.
J Natl Cancer Inst. 2006;98:599-609. 238. Smith BD, Arthur DW, Buchholz TA, et al. Accelerated par-
222. Hughes LL, Wang M, Page DL, et al. Local excision alone tial breast irradiation consensus statement from the American
without irradiation for ductal carcinoma in situ of the Society for Radiation Oncology (ASTRO). J Am Coll Surg.
breast: a trial of the Eastern Cooperative Oncology Group. 2009;209:269-277.
J Clin Oncol. 2009;27:5319-5324. 239. Fisher B, Redmond C, Fisher ER, et al. Ten-year results of a
223. McCormick B. RTOG 9804: a prospective randomized randomized clinical trial comparing radical mastectomy and
trial for “good risk” ductal carcinoma in situ (DCIS), com- total mastectomy with or without radiation. N Engl J Med.
paring radiation (RT) to observation (OBS). J Clin Oncol. 1985;12:674-681.
2012;30(suppl):abstr 1004. 240. Krag D, Weaver DL, Alex JC, et al. Surgical resection and
224. McCormick B, Winter K, Hudis C, et al. RTOG 9804: a pro- radiolocalization of the sentinel lymph node in breast cancer
spective randomized trial for good-risk ductal carcinoma in using a gamma probe. Surg Oncol. 1993;2:335-339.
situ comparing radiotherapy with observation. J Clin Oncol. 241. Giuliano AE, Dale PS, Turner RR, et al. Improved axillary
2015;33(7):709-715. staging of breast cancer with sentinel lymphadenectomy. Ann
225. Rakovitch E, Nofech-Mozes S, Hanna W, et al. A population- Surg. 1995;222:394-399; discussion 399-401.
based validation study of the DCIS Score predicting recur- 242. Giuliano AE, Kirgan DM, Guenther JM, et al. Lymphatic
rence risk in individuals treated by breast-conserving surgery mapping and sentinel lymphadenectomy for breast cancer.
alone. Breast Cancer Res Treat. 2015;152(2):389-398. Ann Surg. 1994;220:391-398; discussion 398-401.
226. Fisher B, Dignam J, Wolmark N, et al. Tamoxifen in treat- 243. Krag DN, Anderson SJ, Julian TB, et al. Technical outcomes
ment of intraductal breast cancer: National Surgical Adjuvant of sentinel-lymph-node resection and conventional axillary
Breast and Bowel Project B-24 randomised controlled trial. lymph-node dissection in patients with clinically node-
Lancet. 1999;353:1993-2000. negative breast cancer: results from the NSABP B-32 ran-
227. Effects of radiotherapy and surgery in early breast cancer. An domised phase III trial. Lancet Oncol. 2007;8:881-888.
overview of the randomized trials. Early Breast Cancer Trialists’ 244. Veronesi U, Paganelli G, Viale G, et al. A randomized com-
Collaborative Group. N Engl J Med. 1995;333:1444-1455. parison of sentinel-node biopsy with routine axillary dissec-
228. Arriagada R, Le MG, Rochard F, et al. Conservative treatment tion in breast cancer. N Engl J Med. 2003;349:546-553.
versus mastectomy in early breast cancer: patterns of failure 245. Veronesi U, Viale G, Paganelli G, et al. Sentinel lymph node
with 15 years of follow-up data. Institut Gustave-Roussy biopsy in breast cancer: ten-year results of a randomized con-
Breast Cancer Group. J Clin Oncol. 1996;14:1558-1564. trolled study. Ann Surg. 2010;251:595-600.
229. Cooke T, Reeves J, Lanigan A, et al. HER2 as a prognos- 246. Ashikaga T, Krag DN, Land SR, et al. Morbidity results
tic and predictive marker for breast cancer. Ann Oncol. from the NSABP B-32 trial comparing sentinel lymph
2001;12(suppl 1):S23-S28. node dissection versus axillary dissection. J Surg Oncol.
230. Fisher B, Anderson S, Redmond CK, et al. Reanalysis and 2010;102:111-118.
results after 12 years of follow-up in a randomized clinical 247. Giuliano AE, Hawes D, Ballman KV, et al. Association of
trial comparing total mastectomy with lumpectomy with or occult metastases in sentinel lymph nodes and bone marrow
without irradiation in the treatment of breast cancer. N Engl with survival among women with early-stage invasive breast
J Med. 1995;333:1456-1461. cancer. JAMA. 2011;306:385-393.
231. Gump FE, Jicha DL, Ozello L. Ductal carcinoma in situ 248. Giuliano AE, Hunt KK, Ballman KV, et al. Axillary dis-
(DCIS): a revised concept. Surgery. 102:790-795. section vs no axillary dissection in women with invasive
232. Paik S, Bryant J, Tan-Chiu E, et al. HER2 and choice of adju- breast cancer and sentinel node metastasis: a randomized
vant chemotherapy for invasive breast cancer: National Surgi- clinical trial. JAMA. 2011;305:569-575.
cal Adjuvant Breast and Bowel Project Protocol B-15. J Natl 249. Giuliano AE, Ballman KV, McCall L, et al. Effect of axil-
Cancer Inst. 2000;92:1991-1998. lary dissection vs no axillary dissection on 10-year overall
survival among women with invasive breast cancer and senti- exemestane for postmenopausal women with estrogen recep- 609
nel node metastasis: the ACOSOG Z0011 (Alliance) random- tor-rich stage 2 to 3 breast cancer: clinical and biomarker
ized clinical trial. JAMA. 2017;318(10):918-926. outcomes and predictive value of the baseline PAM50-
250. Giuliano AE, Ballman KV, McCall L, et al. Locoregional based intrinsic subtype-ACOSOG Z1031. J Clin Oncol.
recurrence after sentinel lymph node dissection with or with- 2011;29:2342-2349.
out axillary dissection in patients with sentinel lymph node 266. Favret AM, Carlson RW, Goffinet DR, et al. Locally advanced
metastases: long-term follow-up from the American College breast cancer: is surgery necessary? Breast J. 2001;7:131-137.
of Surgeons Oncology Group (Alliance) ACOSOG Z0011 267. Khan SA, Stewart AK, Morrow M. Does aggressive local
randomized trial. Ann Surg. 2016;264:413-420. therapy improve survival in metastatic breast cancer? Sur-
251. Wilke LG, McCall LM, Posther KE, et al. Surgical compli- gery. 2002;132:620-626; discussion 626-627.
cations associated with sentinel lymph node biopsy: results 268. Gnerlich J, Jeffe DB, Deshpande AD, et al. Surgical removal

CHAPTER 17 THE BREAST


from a prospective international cooperative group trial. Ann of the primary tumor increases overall survival in patients
Surg Oncol. 2006;13:491-500. with metastatic breast cancer: analysis of the 1988-2003
252. Lucci A, McCall LM, Beitsch PD, et al. Surgical complica- SEER data. Ann Surg Oncol. 2007;14:2187-2194.
tions associated with sentinel lymph node dissection (SLND) 269. Khan SA, DesJardin ESM. Readdressing the role of surgery
plus axillary lymph node dissection compared with SLND of the primary tumor in de novo stage IV breast cancer. Can-
alone in the American College of Surgeons Oncology Group cer Treat Res. 2018;173:73-88.
Trial Z0011. J Clin Oncol. 2007;25:3657-3663. 270. Howlader N, Cronin KA, Kurian AW, Andridge R. Dif-
253. The American Society of Breast Surgeons position state- ferences in breast cancer survival by molecular subtypes
ment on management of the axilla in patients with invasive in the United States. Cancer Epidemiol Biomarkers Prev.
breast cancer. Available at: http://www.breastsurgeons.org/ 2018;27(6):619-626.
statements/PDF_Statements/Axillary_Management.pdf. 271. Bass SS, Lyman GH, McCann CR, et al. Lymphatic mapping
Accessed February, 26, 2014. and sentinel lymph node biopsy. Breast J. 1999;5(5):288-295.
254. Galimberti V, Cole BF, Zurrida S, et al. Axillary dissection 272. Cox CE, Nguyen K, Gray RJ, et al. Importance of lymphatic
versus no axillary dissection in patients with sentinel-node mapping in ductal carcinoma in situ (DCIS): why map DCIS?
micrometastases (IBCSG 23-01): a phase 3 randomised con- Am Surg. 2001;67:513-519; discussion 519-521.
trolled trial. Lancet Oncol. 2013;14:297-305. 273. Dupont E, Cox C, Shivers S, et al. Learning curves and breast
255. Julian TB, Blumencranz P, Deck K, et al. Novel intraop- cancer lymphatic mapping: institutional volume index. J Surg
erative molecular test for sentinel lymph node metastases Res. 2001;97:92-96.
in patients with early-stage breast cancer. J Clin Oncol. 274. Krag D, Weaver D, Ashikaga T, et al. The sentinel node in
2008;26:3338-3345. breast cancer—a multicenter validation study. N Engl J Med.
256. Van Zee KJ, Manasseh DM, Bevilacqua JL, et al. A nomo- 1988;339:941-946.
gram for predicting the likelihood of additional nodal metas- 275. McMasters KM, Giuliano AE, Ross MI, et al. Sentinel-
tases in breast cancer patients with a positive sentinel node lymphnode biopsy for breast cancer—not yet the standard of
biopsy. Ann Surg Oncol. 2003;10:1140-1151. care. N Engl J Med. 1998;339:990-995.
257. Lyman GH, Giuliano AE, Somerfield MR, et al. American 276. O’Hea BJ, Hill AD, El-Shirbiny AM, et al. Sentinel lymph
Society of Clinical Oncology guideline recommendations for node biopsy in breast cancer: initial experience at Memo-
sentinel lymph node biopsy in early-stage breast cancer. J rial Sloan-Kettering Cancer Center. J Am Coll Surg.
Clin Oncol. 2005:23:7703-7720. 1998;186:423-427.
258. Lyman GH, Somerfield MR, Giuliano AE. Sentinel lymph 277. Souba WW, Bland KI. Indications and techniques for biopsy.
node biopsy for patients with early-stage breast cancer: In: Bland KI, Copeland EMI, eds. The Breast: Comprehen-
American Society of Clinical Oncology Clinical Practice sive Management of Benign and Malignant Diseases. Phila-
guideline update. J Clin Oncol. 2017;35:561-564. delphia: WB Saunders; 1998:802.
259. Henry P, Wasserstein D, Paterson M, Kreder H, Jenkinson R. 278. Veronesi U, Paganelli G, Galimberti V, et al. Sentinel-node
Risk factors for reoperation and mortality after the operative biopsy to avoid axillary dissection in breast cancer with clini-
treatment of tibial plateau fractures in Ontario, 1996-2009. cally negative lymph-nodes. Lancet. 1997;349:1864-1867.
J Orthop Trauma. 2015;29:182-188. 279. Wilke LG, Giuliano A: Sentinel lymph node biopsy in
260. Hortobagyi GN, Singletary SE, et al. Treatment of locally patients with early-stage breast cancer: status of the National
advanced and inflammatory breast cancer. In: Harris JR, et Clinical Trials. Surg Clin North Am. 2003;83:901-910.
al, eds. Diseases of the Breast. Philadelphia: Lippincott Wil- 280. Xing Y, Foy M, Cox DD, et al. Meta-analysis of sentinel
liams Wilkins: 2000:645. lymph node biopsy after preoperative chemotherapy in
261. Chen AM, Meric-Bernstam F, Hunt KK, et al. Breast con- patients with breast cancer. Br J Surg. 2006;93:539-546.
servation after neoadjuvant chemotherapy: the MD Anderson 281. Hunt KK, Yi M, Mittendorf EA, et al. Sentinel lymph node
cancer center experience. J Clin Oncol. 2004;22:2303-2312. surgery after neoadjuvant chemotherapy is accurate and
262. Mieog JS, Van Der Hage JA, Van de Velde CJ. Neoadju- reduces the need for axillary dissection in breast cancer
vant chemotherapy for operable breast cancer. Br J Surg. patients. Ann Surg. 2009;250:558-566.
2007;94:1189-1200. 282. Tan VK, Goh BK, Fook-Chong S, et al. The feasibility and
263. Mathew J, Agrawal A, Asgeirsson KS, et al. Primary endo- accuracy of sentinel lymph node biopsy in clinically noden-
crine therapy in locally advanced breast cancers–the Not- egative patients after neoadjuvant chemotherapy for breast
tingham experience. Breast Cancer Res Treat. 2009;113: cancer—a systematic review and meta-analysis. J Surg
403-407. Oncol. 2011;104:97-103.
264. Ellis MJ, Suman VJ, Hoog J, et al. Ki67 proliferation index 283. Yi M, Meric-Bernstam F, Ross MI, et al. How many sentinel
as a tool for chemotherapy decisions during and after neoad- lymph nodes are enough during sentinel lymph node dissec-
juvant aromatase inhibitor treatment of breast cancer: results tion for breast cancer? Cancer. 2008;113:30-37.
from the American College of Surgeons Oncology Group 284. Kong AL, Tereffe W, Hunt KK, et al. Impact of internal mam-
Z1031 trial (Alliance). J Clin Oncol. 2017;35:1061-1069. mary lymph node drainage identified by preoperative lym-
265. Ellis MJ, Suman VJ, Hoog J, et al. Randomized phase ii phoscintigraphy on outcomes in patients with stage I to III
neoadjuvant comparison between letrozole, anastrozole, and breast cancer. Cancer. 2012;118:6287-6296.
610 285. Fisher B. Lumpectomy (segmental mastectomy and axillary of radiotherapy after mastectomy and axillary surgery on
dissection). In: Bland KI, Copeland EMI, eds. The Breast: 10-year recurrence and 20-year breast cancer mortality:
Comprehensive Management of Benign and Malignant meta-analysis of individual patient data for 8135 women in
Diseases. Philadelphia: WB Saunders; 1998:917. 22 randomised trials. Lancet. 2014;383:2127-2135.
286. Newman LA, Washington TA. New trends in breast conserva- 304. Smith BD, Bellon JR, Blitzblau R, et al. Radiation therapy for
tion therapy. Surg Clin North Am. 2003;83:841-883. the whole breast: Executive summary of an American Society
287. NIH consensus conference. Treatment of early-stage breast for Radiation Oncology (ASTRO) evidence-based guideline.
cancer. JAMA. 1991;265:391-395. Pract Radiat Oncol. 2018;8:145-152.
288. Group EBCTC. Effect of radiotherapy after breast-conserv- 305. Polychemotherapy for early breast cancer: an overview of the
ing surgery on 10-year recurrence and 15-year breast cancer randomised trials. Early Breast Cancer Trialists’ Collabora-
death: meta-analysis of individual patient data for 10 801 tive Group. Lancet. 352:930-942.
women in 17 randomised trials. Lancet. 2011;378:1707-1716. 306. Fisher B, Brown AM, Dimitrov NV, et al. Two months of
289. Houssami N, Macaskill P, Marinovich ML, et al. Meta- doxorubicin-cyclophosphamide with and without interval
PART II

analysis of the impact of surgical margins on local recurrence reinduction therapy compared with 6 months of cyclophos-
in women with early-stage invasive breast cancer treated with phamide, methotrexate, and fluorouracil in positive-node
breast-conserving therapy. Eur J Cancer. 2010;46:3219-3232. breast cancer patients with tamoxifen-nonresponsive tumors:
290. Moran MS, Schnitt SJ Giuliano AE, et al. Society of Surgical results from the National Surgical Adjuvant Breast and Bowel
Oncology–American Society for Radiation Oncology con- Project B-15. J Clin Oncol. 1990;8:1483-1496.
SPECIFIC CONSIDERATIONS

sensus guideline on margins for breast-conserving surgery 307. Kelleher M, Miles D. 21. The adjuvant treatment of breast
with whole-breast irradiation in stages I and II invasive breast cancer. Int J Clin Pract. 2003;57:195-199.
cancer. Int J Radiat Oncol Biol Phys. 2014;88(3):553-564. 308. Loprinzi CL, Thome SD. Understanding the utility of adju-
291. Houssami N, Macaskill P, Marinovich ML, Morrow M. vant systemic therapy for primary breast cancer. J Clin Oncol.
The association of surgical margins and local recurrence in 2001;19:972-979.
women with early-stage invasive breast cancer treated with 309. Wood WC, Budman DR, Korzun AH, et al. Dose and dose
breast-conserving therapy: a meta-analysis. Ann Surg Oncol. intensity of adjuvant chemotherapy for stage II, node-positive
2014;21:717-730. breast carcinoma. N Engl J Med. 1994;330:1253-1259.
292. Moran MS, Schnitt SJ Giuliano AE, et al. Society of Surgi- 310. Bonadonna G, Bignami P, Buzzoni R, et al. New adjuvant trials
cal Oncology–American Society for Radiation Oncology for resectable breast cancer at the Istituto Nazionale Tumori of
consensus guideline on margins for breast-conserving sur- Milan. Recent Results. Cancer Res. 1984;91:210-213.
gery with whole-breast irradiation in stages I and II inva- 311. Mauri D, Pavlidis N, Ioannidis JP. Neoadjuvant versus adju-
sive breast cancer. Int J Radiat Oncol Biol Phys. 2014;88(3): vant systemic treatment in breast cancer: a meta-analysis.
553-564. J Natl Cancer Inst. 2005;97:188-194.
293. Bland KI, Chang HR. Modified radical mastectomy and total 312. Mittendorf EA, Buchholz TA, Tucker SL, et al. Impact of
(simple) mastectomy. In: Bland KI, Copeland EMI, eds. The chemotherapy sequencing on local-regional failure risk in
Breast: Comprehensive Management of Benign and Malig- breast cancer patients undergoing breast-conserving therapy.
nant Diseases. Philadelphia: WB Saunders;1998:881. Ann Surg. 257:173-179.
294. Simmons RM, Adamovich TL. Skin-sparing mastectomy. 313. Symmans WF, Wei C, Gould R, et al. Long-term prognos-
Surg Clin North Am. 2003;83:885-899. tic risk after neoadjuvant chemotherapy associated with
295. McCraw JB, Papp C. Breast reconstruction following mas- residual cancer burden and breast cancer subtype. J Clin
tectomy, In: Bland KI, Copeland EMI, eds. The Breast: Com- Oncol. 2017;35(10):1049-1060.
prehensive Management of Benign and Malignant Diseases. 314. Kuerer HM, Newman LA, Smith TL, et al. Clinical course
Philadelphia: WB Saunders;1998:96. of breast cancer patients with complete pathologic primary
296. Fortin A, Dagnault A, Larochelle M, et al. Impact of locore- tumor and axillary lymph node response to doxorubicin-based
gional radiotherapy in node-positive patients treated by neoadjuvant chemotherapy. J Clin Oncol. 1999;17:460-469.
breast-conservative treatment. Int J Radiat Oncol Biol Phys. 315. Caudle AS, Gonzalez-Angulo AM, Hunt KK, et al. Predictors
2003;56:1013-1022. of tumor progression during neoadjuvant chemotherapy in
297. Hellman S. Stopping metastases at their source. N Engl J breast cancer. J Clin Oncol. 2010;28:1821-1828.
Med. 1997;337:996-997. 316. Mamounas EP, Brown A, Anderson S, et al. Sentinel node
298. Overgaard M, Hansen PS, Overgaard J, et al. Postopera- biopsy after neoadjuvant chemotherapy in breast cancer:
tive radiotherapy in high-risk premenopausal women with results from National Surgical Adjuvant Breast and Bowel
breast cancer who receive adjuvant chemotherapy. Danish Project Protocol B-27. J Clin Oncol. 2005;23:2694-2702.
Breast Cancer Cooperative Group 82b Trial. N Engl J Med. 317. Boughey JC, Suman VJ, Mittendorf EA, et al. Sentinel
1997;337:949-955. lymph node surgery after neoadjuvant chemotherapy in
299. Overgaard M, Jensen MB, Overgaard J, et al. Postopera- patients with node-positive breast cancer: the ACOSOG
tive radiotherapy in high-risk postmenopausal breast-cancer Z1071 (Alliance) clinical trial. JAMA. 2013;310:1455-1461.
patients given adjuvant tamoxifen: Danish Breast Cancer 318. Boughey JC, Ballman KV, Le-Petross HT, et al. Identification
Cooperative Group DBCG 82c randomised trial. Lancet. and resection of clipped node decreases the false-negative
1999;353:1641-1648. rate of sentinel lymph node surgery in patients presenting
300. Ragaz J, Jackson SM, Le N, et al. Adjuvant radiotherapy and with node-positive breast cancer (T0–T4, N1–N2) who
chemotherapy in node-positive premenopausal women with receive neoadjuvant chemotherapy: results from ACOSOG
breast cancer. N Engl J Med. 1997;337:956-962. Z1071 (Alliance). Ann Surg. 2016;263(4):802-807.
301. Recht A, Edge SB. Evidence-based indications for postmas- 319. Caudle AS, Yang WT, Krishnamurthy S, et al. Improved axil-
tectomy irradiation. Surg Clin North Am. 2003;83:995-1013. lary evaluation following neoadjuvant therapy for patients
302. Recht A, Edge SB, Solin LJ, et al. Postmastectomy radio- with node-positive breast cancer using selective evaluation
therapy: clinical practice guidelines of the American Society of clipped nodes: implementation of targeted axillary dissec-
of Clinical Oncology. J Clin Oncol. 2001;19:1539-1569. tion. J Clin Oncol. 2016;34:1072-1078.
303. EBCTCG (Early Breast Cancer Trialists’ Collabora- 320. Guarneri V, Broglio K, Kau SW, et al. Prognostic value of
tive Group); McGale P, Taylor C, Correa C, et al. Effect pathologic complete response after primary chemotherapy in
relation to hormone receptor status and other factors. J Clin 337. Goss PE, Hershman DL, Cheung AM, et al. Effects of adju- 611
Oncol. 24:1037-1044. vant exemestane versus anastrozole on bone mineral density
321. Smith IE, Dowsett M, Ebbs SR, et al. Neoadjuvant treatment for women with early breast cancer (MA.27B): a compan-
of postmenopausal breast cancer with anastrozole, tamoxifen, ion analysis of a randomised controlled trial. Lancet Oncol.
or both in combination: the Immediate Preoperative Anastro- 2014;15(4):474-482.
zole, Tamoxifen, or Combined with Tamoxifen (IMPACT) 338. Smith I, Yardley D, Burris H, et al. Comparative efficacy and
multicenter double-blind randomized trial. J Clin Oncol. safety of adjuvant letrozole versus anastrozole in postmeno-
2005;23(22):5108-5116. pausal patients with hormone receptor-positive, node-positive
322. Delpech Y, Coutant C, Hsu L, et al. Clinical benefit from early breast cancer: final results of the randomized phase III
neoadjuvant chemotherapy in oestrogen receptor-positive Femara Versus Anastrozole Clinical Evaluation (FACE) trial.
invasive ductal and lobular carcinomas. Br J Cancer. J Clin Oncol. 2017;35(10):1041-1048.

CHAPTER 17 THE BREAST


2013;108(2):285-291. 339. Early Breast Cancer Trialists’ Collaborative Group. Aro-
323. Joh JE, Esposito NN, Kiluk JV, et al. Pathologic tumor matase inhibitors versus tamoxifen in early breast cancer:
response of invasive lobular carcinoma to neo-adjuvant che- patient-level meta-analysis of the randomised trials. Lancet.
motherapy. Breast J. 2012;18(6):569-574. 2015;386(10001):1341-1352.
324. Dixon JM, Renshaw L, Dixon J, Thomas J. Invasive lobular 340. Goss PE, Ingle JN, Pritchard KI, et al. Extending aromatase-
carcinoma: response to neoadjuvant letrozole therapy. Breast inhibitor adjuvant therapy to 10 years. N Engl J Med.
Cancer Res Treat. 2011;130:871-877. 2016;375:209-219.
325. Spring LM, Gupta A, Reynolds KL, et al. Neoadjuvant 341. Finn RS, Crown JP, Lang I, et al. The cyclin-dependent
endocrine therapy for estrogen receptor-positive breast can- kinase 4/6 inhibitor palbociclib in combination with letro-
cer: a systematic review and meta-analysis. JAMA Oncol. zole versus letrozole alone as first-line treatment of oestrogen
2016;2(11):1477-1486. receptor-positive, HER2-negative, advanced breast cancer
326. Baselga J, Semiglazov V, van Dam P, et al. Phase II random- (PALOMA-1/TRIO-18): a randomised phase 2 study. Lancet
ized study of neoadjuvant everolimus plus letrozole compared Oncol. 2015;16(1): 25-35.
with placebo plus letrozole in patients with estrogen receptor- 342. Finn RS, Martin M, Rugo HS, et al. Palbociclib and
positive breast cancer. J Clin Oncol. 2009;27:2630-2637. letrozole in advanced breast cancer. N Engl J Med.
327. Baum M, Buzdar A. The current status of aromatase inhibi- 2016;375(20):1925-1936.
tors in the management of breast cancer. Surg Clin North Am. 343. Cristofanilli M, Turnr NC, Bondarenko I, et al. Fulvestrant
83:973-994. plus palbociclib versus fulvestrant plus placebo for treatment
328. Bonneterre J, Thurlimann B, Robertson JF, et al. Anastrozole of hormone-receptor-positive, HER2-negative metastatic
versus tamoxifen as first-line therapy for advanced breast breast cancer that progressed on previous endocrine therapy
cancer in 668 postmenopausal women: results of the Tamoxi- (PALOMA-3): final analysis of the multicentre, double-
fen or Arimidex Randomized Group Efficacy and Tolerability blind, phase 3 randomised controlled trial. Lancet Oncol.
study. J Clin Oncol. 2000;18:3748-3757. 2016;17(4):425-439.
329. Buzdar A, Douma J, Davidson N, et al. Phase III, multicenter, 343a. Goetz, M. P., et al. (2017). MONARCH 3: Abemaciclib As
double-blind, randomized study of letrozole, an aromatase Initial Therapy for Advanced Breast Cancer. J Clin Oncol.
inhibitor, for advanced breast cancer versus megestrol ace- 35(32): 3638-3646.
tate. J Clin Oncol. 2001;19:3357-3366. 343b. Sledge, G. W., Jr., et al. (2017). MONARCH 2: Abemaci-
330. Buzdar AU, Jonat W, Howell A, et al. Anastrozole ver- clib in Combination With Fulvestrant in Women With HR+/
sus megestrol acetate in the treatment of postmenopausal HER2- Advanced Breast Cancer Who Had Progressed
women with advanced breast carcinoma: results of a sur- While Receiving Endocrine Therapy. J Clin Oncol. 35(25):
vival update based on a combined analysis of data from 2875-2884.
two mature phase III trials. Arimidex Study Group. Cancer. 344. Baselga J, Campone M, Piccart M, et al. Everolimus in
1998;83:1142-1152. postmenopausal hormone-receptor-positive advanced breast
331. Campos SM, Winer EP. Hormonal therapy in postmenopausal cancer. N Engl J Med. 2012;366:520-529.
women with breast cancer. Oncology. 2003;64:289-299. 345. Bachelot T, Bourgier C, Cropet C, et al. Randomized phase II
332. Rutqvist LE, Johansson H; Stockholm Breast Cancer trial of everolimus in combination with tamoxifen in patients
Study Group. Long-term follow-up of the randomized with hormone receptor-positive, human epidermal growth
Stockholm trial on adjuvant tamoxifen among postmeno- factor receptor 2-negative metastatic breast cancer with prior
pausal patients with early stage breast cancer. Acta Oncol. exposure to aromatase inhibitors: a GINECO study. J Clin
2007;46(2):133-145. Oncol. 2012;30(22):2718-2724.
333. Fisher B, Dignam J, Bryant J, et al. Five versus more than 346. Wolff AC, Lazar AA, Bondarenko I, et al. Randomized
five years of tamoxifen therapy for breast cancer patients with phase III placebo-controlled trial of letrozole plus oral tem-
negative lymph nodes and estrogen receptor-positive tumors. sirolimus as first-line endocrine therapy in postmenopausal
J Natl Cancer Inst. 1996;88(21):1529-1542. women with locally advanced or metastatic breast cancer.
334. Davies C, Pan H, Godwin J, et al. Long-term effects of J Clin Oncol. 2013;31(2):195-202.
continuing adjuvant tamoxifen to 10 years versus stop- 347. Schiavon G, Hrebien S, Garcia-Murillas I, et al. Analysis of
ping at 5 years after diagnosis of oestrogen receptor-pos- ESR1 mutation in circulating tumor DNA demonstrates evo-
itive breast cancer: ATLAS, a randomised trial. Lancet. lution during therapy for metastatic breast cancer. Sci Transl
2013;381(9869):805-816. Med. 2015;7(313):313ra182.
335. Goss PE, Ingle JN, Martino S, et al. Randomized trial of 348. Robinson DR, Wu YM, Vats P, et al. Activating ESR1 muta-
letrozole following tamoxifen as extended adjuvant therapy tions in hormone-resistant metastatic breast cancer. Nat
in receptor-positive breast cancer: updated findings from Genet. 2013;45(12):1446-1451.
NCIC CTG MA.17. J Natl Cancer Inst. 2005;97:1262-1271. 349. Toy W, Weir H, Razavi P, et al. Activating ESR1 mutations
336. Goss PE, Ingle JN, Pritchard KI, et al. Exemestane versus anas- differentially affect the efficacy of ER antagonists. Cancer
trozole in postmenopausal women with early breast cancer: Discov. 2017;7(3):277-287.
NCIC CTG MA.27—a randomized controlled phase III trial. 350. Jakesz R, Hausmaninger H, Kubista E, et al. Random-
J Clin Oncol. 2013;31(11):1398-1404. ized adjuvant trial of tamoxifen and goserelin versus
612 cyclophosphamide, methotrexate, and fluorouracil: evidence chemotherapy regimens in patients with HER2-positive
for the superiority of treatment with endocrine blockade in pre- early breast cancer: a randomized phase II cardiac safety
menopausal patients with hormone-responsive breast cancer— study (TRYPHAENA). Ann Oncol. 2013;24(9):2278-2284.
Austrian Breast and Colorectal Cancer Study Group Trial 5. 362a. Von Minckwitz, G., et al. (2017). Adjuvant Pertuzumab
J Clin Oncol. 2002;20(24):4621-4627. and Trastuzumab in Early HER2-Positive Breast Cancer.
351. Francis PA, Regan MM, Fleming GF, et al. Adjuvant ovar- N Engl J Med. 377(2): 122-131.
ian suppression in premenopausal breast cancer. N Engl J 363. Chan A, Delaloge S, Holmes FA, et al. Neratinib after
Med. 2015;372(5):436-446. trastuzumab-based adjuvant therapy in patients with
352. Pagani O, Regan MM, Walley BA, et al. Adjuvant exemes- HER2-positive breast cancer (ExteNET): a multicentre,
tane with ovarian suppression in premenopausal breast randomised, double-blind, placebo-controlled, phase 3
cancer. N Engl J Med. 2014;371:107-118. trial. Lancet Oncol. 2016;17(3):367-377.
353. Early Breast Cancer Trialists’ Collaborative Group 364. Bose R, Kavuri SM, Searleman AC, et al. Activating HER2
(EBCTCG). Effects of chemotherapy and hormonal ther- mutations in HER2 gene amplification negative breast cancer.
PART II

apy for early breast cancer on recurrence and 15-year Cancer Discov. 2013;3(2):224-237.
survival: an overview of the randomised trials. Lancet. 365. Lien HC, Chen YL, Juang YL, Jeng YM. Frequent alterations
2005;365:1687-1717. of HER2 through mutation, amplification, or overexpression
354. Paik S, Bryant J, Tan-Chiu E, et al. Real-world performance of in pleomorphic lobular carcinoma of the breast. Breast Can-
HER2 testing—National Surgical Adjuvant Breast and Bowel cer Res Treat. 2015;150:447-455.
SPECIFIC CONSIDERATIONS

Project experience. J Natl Cancer Inst. 2002;94:852-854. 366. Ben-Baruch NE, Bose R, Kavuri SM, Ma CX, Ellis
355. Press MF, Slamon DJ, Flom KJ, et al. Evaluation of HER-2/ MJ. HER2-mutated breast cancer responds to treatment
neu gene amplification and overexpression: comparison with single-agent neratinib, a second-generation HER2/
of frequently used assay methods in a molecularly char- EGFR tyrosine kinase inhibitor. J Natl Compr Canc Netw.
acterized cohort of breast cancer specimens. J Clin Oncol. 2015;13:1061-1064.
2002;20:3095-3105. 367. Gandhi L, Bahleda R, Tolaney SM, et al. Phase I study of
356. Volpi A, De Paola F, Nanni O, et al. Prognostic significance neratinib in combination with temsirolimus in patients with
of biologic markers in node-negative breast cancer patients: human epidermal growth factor receptor 2-dependent and
a prospective study. Breast Cancer Res Treat. 63:181-192. other solid tumors. J Clin Oncol. 2014;32(2):68-75.
357. Goldhirsch A, Gelber RD, Piccart-Gebhart MJ, et al. 2 years 368. Robinson DS, Sundaram M, et al. Carcinoma of the breast
versus 1 year of adjuvant trastuzumab for HER2-positive in pregnancy and lactation. In: Bland KI, Copeland EMI,
breast cancer (HERA): an open-label, randomised controlled eds. The Breast: Comprehensive Management of Benign and
trial. Lancet. 2013;382(9897):1021-1028. Malignant Diseases. Philadelphia: WB Saunders; 1998:1433.
358. Pivot X, Romieu G, Debled M, et al. 6 months versus 369. Giordano SH, Buzdar AU, Hortobagyi GN: Breast cancer in
12 months of adjuvant trastuzumab for patients with HER2- men. Ann Intern Med. 2002;137:678-687.
positive early breast cancer (PHARE): a randomised phase 3 370. Wilhelm MC. Cancer of the male breast. In: Bland KI, Cope-
trial. Lancet Oncol. 2013;14(8):741-748. land EMI, eds. The Breast: Comprehensive Management of
359. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine Benign and Malignant Diseases. Philadelphia: WB Saunders;
for HER2-positive advanced breast cancer. N Engl J Med. 1998:1416.
2012;367:1783-1791. 371. Khan SA, Badve S. Phyllodes tumors of the breast. Curr
360. Baselga J, Cortés J, Kim SB, et al. Pertuzumab plus trastu- Treat Options Oncol. 2001;2:139-147.
zumab plus docetaxel for metastatic breast cancer. N Engl J 372. Chittoor SR, Swain SM. Locally advanced breast cancer:
Med. 2012;366(2):109-119. Role of medical oncology. In: Bland KI, Copeland EMI,
361. Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety eds. The Breast: Comprehensive Management of Benign and
of neoadjuvant pertuzumab and trastuzumab in women Malignant Diseases. Philadelphia: WB Saunders; 1998:1403.
with locally advanced, inflammatory, or early HER2- 373. Mies C. Mammary sarcoma and lymphoma. In: Bland KI,
positive breast cancer (NeoSphere): a randomised multi- Copeland EMI, eds. The Breast: Comprehensive Management
centre, open-label, phase 2 trial. Lancet Oncol. 2012;13(1): of Benign and Malignant Diseases. Philadelphia: WB Saunders;
25-32. 1998:307.
362. Schneeweiss A, Chia S, Hickish T, et al. Pertuzumab 374. Stewart FW, Treves N. Lymphangiosarcoma in postmas-
plus trastuzumab in combination with standard neoad- tectomy lymphedema; a report of six cases in elephantiasis
juvant anthracycline-containing and anthracycline-free chirurgica. Cancer. 1948;1:64-81.

You might also like