Breast Cancer: June 2014
Breast Cancer: June 2014
Breast Cancer: June 2014
net/publication/321477037
Breast Cancer
CITATIONS READS
0 14,822
1 author:
Ahmed El-Sharkawy
Italian National Research Council
14 PUBLICATIONS 35 CITATIONS
SEE PROFILE
Some of the authors of this publication are also working on these related projects:
Unravelling genetic modifiers of Incontinentia Pigmenti and their role in the NF-‐kB pathway View project
All content following this page was uploaded by Ahmed El-Sharkawy on 02 December 2017.
Edited by
Redhwan Ahmed Al-Naggar
Ibrahim Hashim
OMICD Group eBooks
001
eBooks
Notice:
Statements and opinions expressed in the book are these of the individual contributors
and not necessarily those of the editors or publisher. No responsibility is accepted for the
accuracy of information contained in the published chapters. The publisher assumes no
responsibility for any damage or injury to persons or property arising out of the use of any
materials, instructions, methods or ideas contained in the book.
Breast Cancer
Ahmed M ElSharkawy*
*Biochemistry department, Faculty of Science, Alexandria University, Alexandria,
Egypt
*Corresponding author: Ahmed M ElSharkawy, Biochemistry department,
Faculty of Science, Alexandria University, Alexandria, Egypt, Tel: +201000512872;
E-mail: [email protected]
Introduction
Cancer is a group of diseases causing cells in the body to change and proliferate beyond
of control. Most types of cancer cells eventually form a lump or mass called a tumor, and are
named after the part of the body where the tumor originates. Breast cancer begins in the breast
tissue, which contains glands for milk production, called lobules, and the ducts that connect
the lobules to the nipple. The remainder of the breast is made up of fatty, connective, and
lymphatic tissues. Breast cancer typically is detected either during a screening examination,
before symptoms have developed, or after symptoms have developed, when a woman feels
a lump. Most masses seen on a mammogram and most breast lumps turn out to be benign
which are non-cancerous lumps that do not grow uncontrollably, do not spread, and are
not life-threatening. If cancer is suspected after a clinical breast exam or breast imaging, a
microscopic analysis of breast tissue is necessary. This will provide a definitive diagnosis, help
determine the spread (in situ or invasive) and to characterize the pattern of the disease. The
tissue for microscopic analysis can be obtained via a needle or surgical biopsy. Selection of the
type of biopsy is based on individual patient clinical factors, availability of particular biopsy
devices, and resources.
Breast Structure
The breast lies between the second and sixth ribs, from the sternal edge to the edge of the
axilla, and against the pectoralis muscle on the chest wall. Breast tissue also projects into the
axilla as the tail of Spence. For clinical purposes, the breast is divided into four quadrants:
upper inner, upper outer, lower inner and lower outer quadrants. Cancer occurs most often in
the upper outer quadrant.
The breast is composed of 15-20 lobes that radiate from the nipple. Each lobe is surrounded
by fat and fibrous connective tissue and is divided into many lobules. The lobule (sometimes
called the ductal-lobular unit) is the basic structural unit of the breast and is lined by epithelial
cells. Each lobule is subdivided into 10 to 100 alveoli, the milk producing units of the breast.
Milk flows from the alveoli of the lobules into the ducts. The ducts gradually coalesce into 10
to 15 major ducts; each lobe containing one major duct terminating in the nipple.
The most common type of breast cancer is a carcinoma that originates in epithelial cells.
About 85% of breast carcinomas originate within the cells of the ducts (ductal carcinoma); the
remaining 15% begin in the cells that line the lobules (lobular carcinoma). Non-carcinomatous
breast cancers are rare and originate in the connective tissues of the breast (Figure 1).
Breasts also contain blood and lymphatic vessels. Most lymphatic vessels within the
breast lead to axillary lymph nodes. Some also connect to supra- or infraclavicular nodes,
and internal mammary nodes. Cancer cells may enter lymphatic vessels and spread to lymph
nodes. Cancer cells may also enter blood vessels and spread to other parts of the body.
Beneath the tissues of the breast lie the muscles of the chest wall and between the two is the
fascia (a layer of connective tissue). Two layers of suspensory ligaments (Cooper’s ligaments)
link the breast to the fascia, providing support. As these ligaments stretch with age or weight
gain, the breast loses some of its firmness.
1. Chest wall 2. Pectoralis muscles 3. Lobules 4. Nipple 5. Areola 6. Milk duct 7. Fatty
tissue 8. Skin
Breast development
Breast tissue begins to develop around the sixth week in utero with the formation of the
mammary ridge. Also called the mammary fold or milk line, this thickening of the skin extends
from just below the axilla to the inguinal region (groin area). It then recedes before birth,
leaving two primary breast buds on the upper half of the chest. In newborns, the nipples and
beginnings of the milk duct system are present. This rudimentary system remains essentially
at rest until puberty. For females, usually around the ages of 10 or 11, the release of ovary
producing hormones (estrogen and progesterone) stimulates further development of the ducts
and lobules. At the same time, increases in adipose tissue (fatty tissue) and fibrous connective
tissue cause the breasts to enlarge. By adulthood, all glandular elements are fully formed
and breast development is complete. At menopause, the lobules recede, leaving mostly ducts,
adipose and fibrous tissue. Histologically, postmenopausal and prepubertal breasts are very
similar.
Throughout a woman’s life, breast tissue remains sensitive to hormonal changes.
Throughout a woman’s life, breast tissue remains sensitive to hormonal changes, including
those that occur during menstruation, pregnancy and lactation. During each menstrual
cycle, levels of estrogen and progesterone rise, causing ducts and milk glands to enlarge. In
pregnancy, rising hormonal levels cause further branching and differentiation of the ducts and
lobules, accompanied by an increase in adipose tissue and richer blood flow. With lactation,
lobules become dilated and engorged with colostrum and milk. Lactation also increases the
possibility of abnormal nipple discharge, infection and inflammation, although non lactating
women may also experience these types of benign breast changes.
1- Local stage refers to cancers that are confined to the breast (corresponding to stage I and
some stage II cancers in the TNM staging system).
2- Regional stage refers to tumors that have spread to surrounding tissue or nearby lymph
nodes (generally corresponding to stage II or III cancers, depending on size and lymph node
involvement).
3- Distant stage refers to cancers that have metastasized (spread) to distant organs or
lymph nodes above the collar bone (corresponding to stages IIIc and IV).
the rest of the breast tissue. More than 80% of breast cancer cases are discovered when the
woman feels a lump [13]. The earliest breast cancers can be detected by a mammogram [14].
Lumps found in lymph nodes located in the armpits [13] can also indicate breast cancer.
Other signs of breast cancer other than a lump may include thickening of breast tissue, one
breast becoming larger or lower, a nipple changing position or shape or becoming inverted,
skin puckering or dimpling, a rash on or around a nipple, discharge from nipple/s, constant
pain in part of the breast or armpit, and swelling beneath the armpit or around the collarbone
[15]. Pain (“mastodynia”) is an unreliable tool in determining the presence or absence of
breast cancer, but may be indicative of other breast health issues [13,14]. Inflammatory
breast cancer is a particular type of breast cancer which can pose a substantial diagnostic
challenge. Symptoms may resemble a breast inflammation and may include itching, pain,
swelling, nipple inversion, warmth and redness throughout the breast, as well as an orange-
peel texture to the skin referred to as peau d’orange [13] as inflammatory breast cancer
doesn’t show as a lump there is sometimes a delay in diagnosis. Another reported symptom
complication of breast cancer is Paget’s disease of the breast. This syndrome presents as skin
changes resembling eczema, such as redness, discoloration, or mild flaking of the nipple skin.
As Paget’s disease of the breast advances, symptoms may include tingling, itching, increased
sensitivity, burning, and pain. There may also be discharge from the nipple. Approximately
half of women diagnosed with Paget’s disease of the breast also have a lump in the breast.
In rare cases, what initially appears as a fibroadenoma (hard, movable non-cancerous lump)
could in fact be a phyllodes tumor. Phyllodes tumors are formed within the stroma (connective
tissue) of the breast and contain glandular as well as stromal tissue. Phyllodes tumors are
not staged in the usual sense; they are classified on the basis of their appearance under
the microscope as benign, borderline, or malignant. Occasionally, breast cancer presents as
metastatic disease - that is, cancer that has spread beyond the original organ. The symptoms
caused by metastatic breast cancer will depend on the location of metastasis. Common sites of
metastasis include bone, liver, lung and brain [16]. Unexplained weight loss can occasionally
herald an occult breast cancer, as can symptoms of fevers or chills. Bone or joint pains can
sometimes be manifestations of metastatic breast cancer, as can jaundice or neurological
symptoms. These symptoms are called non-specific, meaning they could be manifestations
of many other illnesses. Most symptoms of breast disorders, including most lumps, do not
turn out to represent underlying breast cancer. Fewer than 20% of lumps, for example, are
cancerous, [17] and benign breast diseases such as mastitis and fibroadenoma of the breast
are more common causes of breast disorder symptoms. Nevertheless, the appearance of a
new symptom should be taken seriously by both patients and their doctors, because of the
possibility of an underlying breast cancer at almost any age [13].
Risk Factors
Many of the known breast cancer risk factors such as sex, age, family history, early
menarche, and late menopause, are not modifiable, that is they cannot be changed. However,
other factors associated with increased breast cancer risk, including postmenopausal obesity,
use of combined estrogen and progestin menopausal hormones, cigarette smoking, and
alcohol consumption are modifiable. Many risk factors affect lifetime exposure of breast tissue
to hormones (early menarche, late menopause, obesity, and hormone use). Reproductive
hormones are thought to influence breast cancer risk by increasing cell proliferation, thereby
increasing the likelihood of DNA damage, as well as promotion of cancer growth. Many of
the known risk factors for breast cancer are specifically associated with the ER+/luminal
Principles and Practice of Cancer Prevention and Control 5
Edited by: Redhwan Ahmed Al-Naggar
eBooks
A subtype; less is known about risk factors for ER- and triple negative (basal-like) breast
cancers. Strategies that may help reduce the risk of breast cancer include avoiding weight gain
and obesity, engaging in regular physical activity, and minimizing alcohol intake [18]. Women
who choose to breastfeed for an extended period of time (studies suggest a year or more) may
also reduce their breast cancer risk. Women should consider the increased risk of breast
cancer associated with the use of estrogen and progestin when evaluating treatment options
for menopausal symptoms. Treatment with tamoxifen or raloxifene can also reduce the risk of
breast cancer among women at high risk. Breast cancer risk factors, along with factors that
may decrease the risk of breast cancer, are discussed below.
Personal and family history
Family history of breast cancer: Women (as well as men) with a family history of breast
cancer, especially in a first-degree relative (mother, sister, daughter, father, or brother), are
at increased risk of developing breast cancer; this risk is higher if more than one first-degree
relative developed breast cancer. Compared to women without a family history, risk of breast
cancer is 1.8 times higher for women with one first-degree female relative who has been
diagnosed, nearly 3 times higher for women with two relatives, and nearly 4 times higher for
women with three or more relatives [19]. Risk is further increased when the affected relative
was diagnosed at a young age. It is important to note that the majority of women with one
or more affected first-degree relatives will never develop breast cancer and that most women
who develop breast cancer do not have a family history of the disease. A family history of
ovarian cancer is also associated with increased breast cancer risk in both men and women.
Women with a history of breast or ovarian cancer in their immediate family or in either parent’s
extended family should discuss this with their physicians because it may signal the presence
of a genetic predisposition to cancer.
Genetic predisposition: It is estimated that 5% to 10% of breast cancer cases result from
inherited mutations, including those in the breast cancer susceptibility genes BRCA1 and
BRCA2 [20]. These mutations are present in less than 1% of the general population, but occur
more often in certain ethnic groups such as those of Ashkenazi (Eastern European) Jewish
descent [20]. The estimates of the risk of breast cancer in women with these mutations vary;
by age 70, between 44% and 78% of women with BRCA1 mutations and between 31% and 56%
of women with BRCA2 mutations will develop breast cancer [21,22].
Only about 15%-20% of familial breast cancers are attributed to BRCA1 or BRCA2 gene
mutations [23]. Other inherited conditions associated with smaller increased breast cancer
risk include Li-Fraumeni and Cowden syndromes and a number of more common genetic
mutations [23]. These mutations can be inherited from either parent and by sons as well
as daughters. In addition, low-risk variations in the genetic code may affect breast cancer
risk. Scientists believe that much of the occurrence of breast cancer in families results from
the interaction between lifestyle factors and these low-risk variations that may be shared
within a family [24]. Molecular tests are commercially available to identify some of the BRCA
mutations, as well as many of the family cancer syndromes responsible for inherited forms
of breast cancer; however, the interpretation of these tests and treatment decisions remains
complicated [25]. It is not yet possible to predict if or when women who carry a particular
genetic abnormality will develop breast cancer. Furthermore, tests are not available for all of
the genes that affect breast cancer risk.
Personal history of breast cancer: Women with a history of breast cancer are at increased
risk for developing breast cancer more than once in their lifetime. The risk is higher if the
diagnosis was at a younger age. Women diagnosed with early onset breast cancer (age <40)
have almost a 4.5-fold increased risk of subsequent breast cancer [26]. Genetic predisposition,
such as mutations in BRCA1 and BRCA2 genes, probably contribute to some of the excess
risk of subsequent breast cancers, particularly among women diagnosed at a young age [27].
Lobular carcinoma in situ: This uncommon condition is the result of abnormal cells
forming in the lobules or milk producing glands of the breast. Although LCIS seldom becomes
invasive cancer, women with LCIS are 7 to 12 times more likely to develop invasive cancer in
either breast than women without LCIS [28]. LCIS is not usually apparent on a mammogram
and is typically discovered during a biopsy performed for another reason, such as an abnormal
mammogram. Pure LCIS should be distinguished from DCIS and pleomorphic LCIS, as both
of these conditions are considered precursor lesions for breast cancer and require cancer-
directed therapy.
Benign breast disease: Some types of benign breast conditions are linked to breast cancer
risk. Doctors often categorize these conditions into 3 general groups, reflecting the degree of
risk: non-proliferative lesions, proliferative lesions without atypia (abnormal cells or patterns
of cells), and proliferative lesions with atypia. Non-proliferative lesions are not associated with
overgrowth of breast tissue and have little to no effect on breast cancer risk. Examples of non-
proliferative lesions include fibrosis (also known as fibrocystic changes), simple cysts, and
mild hyperplasia. Proliferative lesions without atypia are associated with a small increase in
the risk of breast cancer (1.5 to 2 times the risk of those who do not have one of these lesions)
and include non-atypical (or usual) ductal hyperplasia and fibroadenoma [29-32]. Proliferative
lesions with atypia are associated with the greatest breast cancer risk four to five times higher
than average risk [29,30,32]. These include Atypical Ductal Hyperplasia (ADH) and Atypical
Lobular Hyperplasia (ALH). Women should keep detailed records of any benign breast biopsy
results, as this information is valuable for risk assessment, screening, and counseling for
chemoprevention and risk-reduction strategies.
Breast density: High breast tissue density (a mammographic indicator of the amount
of breast and connective tissue relative to fatty tissue in the breast) has been shown to be a
strong, independent risk factor for the development of breast cancer [33]. A number of factors
can affect breast density, such as age, menopausal status, the use of certain drugs (such
as menopausal hormone therapy), pregnancy, and genetics. Breast density is influenced by
inherited genetic factors, but decreases with age and is further reduced by pregnancy and
menopause [34,35]. Percent breast density is generally lower among women with higher body
weight because of the higher proportion of fatty tissue [36]. Some drugs affect breast density,
including tamoxifen (decreases density) and combined menopausal hormone therapy (increases
density) [37]. The risk of breast cancer increases with increasing breast density; women with
very high breast density have a four to six fold increased risk of breast cancer compared to
women with the less dense breasts [33,38,39] In addition, mammographic detection of breast
cancer is impaired for dense breast tissue [38]. Some states have laws requiring that women be
informed if they have higher than average breast tissue density, along with the other findings
on their mammogram. In addition, some states also require that women with dense breasts
be told that they may benefit from additional screening [40]. However, at this time there is no
expert consensus about what other tests, if any, should be done in addition to mammograms
to screen for breast cancer in women with dense breasts.
Endogenous hormone levels: Postmenopausal women with high levels of endogenous
Principles and Practice of Cancer Prevention and Control 7
Edited by: Redhwan Ahmed Al-Naggar
eBooks
hormones (estrogen or testosterone produced naturally in the body) have about twice the risk
of developing breast cancer compared to women with the lowest levels [41-43]. High circulating
hormone levels are associated with and may reflect the effects of other breast cancer risk factors,
such as postmenopausal obesity and alcohol use [44]. The relationship in premenopausal
women is less clear, which likely reflects the complexity of measuring hormone levels that
vary during the menstrual cycle. Nevertheless, there is growing evidence linking high levels of
testosterone to breast cancer risk in premenopausal women [45-47]. A recent study reported
that pre- menopausal women in the highest quintiles of total and free testosterone had an 80%
greater breast cancer risk compared to women in the lowest quintiles [45]. Two recent reviews
concluded that high estrogen levels are also associated with a slight increase in breast cancer
risk in premenopausal women [42,48].
Menstrual cycles: Women who have had more menstrual cycles because they started
menstruating before the age of 12 and/or went through menopause after age 55 have a slightly
higher risk of developing breast cancer [49,50]. The increased risk may be due to longer lifetime
exposure to reproductive hormones.
Pregnancy and breast feeding: Younger age at first full-term pregnancy (<30 years) and a
greater number of pregnancies decrease the risk of breast cancer over the long term; however,
there appears to be a transient increase in breast cancer risk following a full-term pregnancy,
particularly among women who are older at first birth [49,51]. Pregnancy-related risk factors
seem to be more strongly related to hormone receptor- positive than hormone receptor-negative
breast cancers [52,53]. Most studies suggest that breastfeeding for a year or more slightly
reduces a woman’s overall risk of breast cancer [54]. The protective effect may be greater for
basal-like breast cancers [54]. Longer duration is associated with greater risk reduction. In a
review of 47 studies conducted in 30 countries, the risk of breast cancer was reduced by 4.3%
for every 12 months of breastfeeding [55]. One possible explanation for this effect may be that
breastfeeding inhibits menstruation, thus reducing the lifetime number of menstrual cycles.
Another possible explanation relates to structural changes that occur in the breast following
lactation and weaning.
Bone mineral density: High bone mineral density in postmenopausal women has been
associated with increased risk for breast cancer in many, but not all, studies; risk appears
to be most strongly related to ER+ disease [56-60]. Bone density is not an independent risk
factor for breast cancer, but a marker for cumulative estrogen exposure [60]. Bone density is
routinely measured to identify women at increased risk for osteoporosis (high bone density
indicates absence of osteoporosis) and may help determine a woman’s risk for developing
breast cancer.
Lifestyle-related factors
Postmenopausal hormone use: Recent use of menopausal hormones (also referred to as
hormone therapy or HT) with combined estrogen and progestin increases the risk of developing
and dying from breast cancer, with higher risk associated with longer use [61,62]. Risk is also
greater for women who start hormone therapy soon after the onset of menopause compared
to those who begin use later [63-65]. The increased risk appears to diminish within 5 years
of discontinuation of hormone use [62,63,66]. Estrogen alone can be prescribed for women
without a uterus, and it is less clear if this therapy increases risk of breast cancer. Updated
results from the Women’s Health Initiative randomized trial found that use of estrogen-only
therapy for an average of 6 years is associated with decreased risk of breast cancer [67];
however, several other observational studies have found a slight increase in risk, particularly
among lean women and for women who began therapy soon after menopause [63,66,68].
Obesity and weight gain: Obesity increases the risk of postmenopausal breast cancer [69].
The risk of postmenopausal breast cancer is about 1.5 times higher in overweight women and
about 2 times higher in obese women than in lean women [70]. Breast cancer risk associated
with excess weight is likely due to high estrogen levels because fat tissue is the largest source
of estrogen in postmenopausal women. Obesity is also a risk factor for type II diabetes, which
some studies have linked to modestly increased risk for postmenopausal breast cancer [71].
The inconsistencies likely reflect the complex relationship between obesity and diabetes,
as well as the fact that diabetic treatments may further affect the risk of developing breast
cancer [72]. In contrast, some studies have found that obesity protects against developing
breast cancer before menopause. A large meta-analysis found that among women ages 40-49,
the risk for developing breast cancer was about 14% lower in overweight women and about
26% lower in obese women compared to women who are normal weight [73]. The underlying
mechanisms for this inverse relationship are not well understood, but the protective effect may
be limited to ER+ breast cancers [52,74,75]. Many studies have looked at whether the timing of
weight gain influences breast cancer risk. Results from a study of more than 80,000 registered
nurses found that women who gained 55 pounds or more after age 18 had almost 50% greater
risk of breast cancer and gaining 22 pounds or more after menopause was associated with an
increased risk of 18%. 102 Although some studies have found weight loss to be associated with
reduced risk, results are not consistent [76-78].
It is more difficult to examine the effect of weight loss on breast cancer because weight loss
is often not sustained.
Physical activity: Growing evidence suggests that women who get regular physical activity
have a 10%-20% lower risk of breast cancer compared to women who are inactive, with stronger
evidence for postmenopausal than premenopausal women [69,79-81]. A recent report from the
Nurses’ Health Study of more than 95,000 women found that increases in physical activity
after menopause lowered breast cancer risk by 10%. 105 The benefit may be due to the effects
of physical activity on body mass, hormones, and energy balance [82].
Diet: Although numerous studies have examined the relationship between food consumption
(including fat, soy, dairy, meat, and fruits and vegetables) and breast cancer, there is no
conclusive evidence that diet influences breast cancer risk [83,84]. A recent meta-analysis of
animal fat intake and breast cancer, which included more than 20,000 breast cancer cases,
concluded there was no association [85]. Similarly, reducing dietary fat in post- menopausal
women did not affect risk of breast cancer in the Women’s Health Initiative dietary intervention.
However, the timing of the exposure may be important, as findings from the Nurses’ Health
Study showed that a high-fat diet during adolescence was associated with a moderate increase
in premenopausal breast cancer risk [86]. It has been suggested that soy consumption may
reduce breast cancer risk, in part because of historically low breast cancer rates among Asian
women. A meta-analysis showed that soy intake was inversely associated with breast cancer
risk in Asian but not Western populations [87]. There is growing evidence that high levels of
fruit and vegetable consumption may be associated with reduced risk of hormone receptor
negative breast cancer [88-90]. The effect of diet on breast cancer risk remains an active
area of research, with studies particularly focusing on timing of exposure, specific dietary
components, and whether risks may differ by tumor hormone receptor status.
Alcohol: Numerous studies have confirmed that alcohol consumption increases the risk
of breast cancer in women by about 7% to 12% for each 10g (roughly one drink) of alcohol
consumed per day [91-93]. The increased risk is dose-dependent and exists regardless of the
type of alcoholic beverage consumed [93]. One of the mechanisms by which alcohol increases
risk of breast cancer is by increasing estrogen and androgen levels [94]. Alcohol use has been
more strongly related with increased risk for ER+ than ER- breast cancers [95,96].
Tobacco: In 2009, the International Agency for Research on Cancer concluded that
there was limited evidence that tobacco smoking causes breast cancer in women based on
a review of 150 studies [97]. A recent meta-analysis by American Cancer Society researchers
found that current smokers had a 12% higher risk of breast cancer than women who never
smoked [98]. Research also suggests that risk may be greater for women who begin smoking
before first childbirth [99,100]. In 2006, the US Surgeon General characterized the evidence
linking secondhand smoke and breast cancer as “suggestive but not sufficient” to infer a
causal relationship [101]. However, a subsequent meta-analysis concluded that there was no
association between secondhand smoke and breast cancer, regardless of the time of onset of
exposure [102]. The results of more recent studies have also failed to show a clear relationship
between passive smoking and breast cancer risk [98,103,104]. Nevertheless, it is clear that
avoiding exposure to secondhand smoke has multiple health benefits.
Oral contraceptive use: Recent use of oral contraceptives may increase the risk of breast
cancer by about 10% to 30%; however, since most studies have looked at older, high-dose
estrogen forms of oral contraceptives, the risk with current, low-dose formulations is not clear
[105]. Women who have stopped using oral contraceptives for 10 years or more have the same
risk as women who never used the pill [105].
Other risk factors
Radiation: The link between radiation exposure and breast cancer has been demonstrated
in studies of atomic bomb survivors and women who have received high-dose radiation therapy
to the chest, particularly for those who were first exposed at younger ages [106,107]. This may
be because breast tissue is most susceptible to carcinogens before it is fully differentiated,
which occurs with first childbirth [108]. Breast cancer is one of the most common types of
second cancers to occur among childhood cancer survivors. Secondary breast cancer is most
strongly associated with high-dose radiation therapy to the chest for women treated between
10 and 30 years of age, such as for Hodgkin lymphoma [109]. Breast cancer risk among
women with such exposure start to rise about 8 years after radiation treatment and continue
to be elevated for more than 25 years [107].
Diethylstilbestrol exposure: From the 1940s through the 1960s, some pregnant women
were given the Drug Diethylstilbestrol (DES) because it was thought to lower the risk of
miscarriage. These women have increased risk (about 30% higher) of developing breast cancer
compared to women who have not taken DES [110]. Some studies have found that women
whose mothers took DES during pregnancy also have a slightly higher risk of breast cancer
[111].
Environmental pollutants: Concerns have been raised among some advocacy groups and
survivors that rising breast cancer incidence in the latter half of the 20th century may have
been caused by environmental pollutants such as organochlorine pesticides. However, studies
to date have found no association between increased concentrations of organochlorines in blood
and fat tissue and breast cancer risk [112-115]. Although animal studies have demonstrated
that prolonged, high-dose exposure to many industrial chemicals can increase mammary
tumor development, it is difficult to determine whether exposure to much lower concentrations
of these chemicals in the general environment - which occur alone or in combination, in air,
drinking water, and consumer products - increases the risk of human breast cancer [116]. In
general, epidemiological studies have not found clear relationships between environmental
pollutants and breast cancer, though these studies have had limited capability to study effects
on population subgroups or to quantify exposures at potentially critical periods of life, such
as adolescence. An association between environmental exposures and breast cancer may be
difficult to quantify because it may reflect an indirect pathway (e.g., an effect of these exposures
on early onset puberty). This continues to be an active area of research.
Occupational exposures: A few occupations have been linked to breast cancer risk.
One study found an increased risk of breast cancer among women employed in commercial
sterilization facilities who were exposed to high levels of ethylene oxide [117]. This chemical
has been shown to cause breast cancer in experimental animals. Night shift work may also
be associated with increased breast cancer risk. Most studies of nurses who work night shifts
and flight attendants who experience circadian rhythm disruption caused by crossing multiple
time zones have found increased risks of breast cancer associated with long-term employment
[118,119]. Animal studies suggest that exposure to light at night causes circadian rhythm
disruption and increases cancer incidence. 146 Some researchers suggest that the increased
risk of breast cancer may be due to decreases in melatonin levels that occur as a result of
exposure to light at night; melatonin may affect estrogen levels, as well as act as a tumor
suppressor [120]. Based on the results of studies in humans and experimental animals, the
International Agency for Research on Cancer concluded in 2007 that shift work, particularly
at night, was probably carcinogenic to humans [121]. Additional studies are needed to confirm
this relationship because shift work at night is a common exposure, involving about 15% to
20% of workers in the US and Europe, and because much of the population in industrialized
countries is exposed to artificial light at night.
the immune system [124]. Abnormal growth factor signaling in the interaction between stromal
cells and epithelial cells can facilitate malignant cell growth [125,126]. In breast adipose tissue,
overexpression of leptin leads to increased cell proliferation and cancer [127]. In the United
States, 10 to 20 percent of patients with breast cancer and patients with ovarian cancer have
a first - or second-degree relative with one of these diseases. The familial tendency to develop
these cancers is called hereditary breast–ovarian cancer syndrome. The best known of these,
the BRCA mutations, confer a lifetime risk of breast cancer of between 60 and 85 percent and
a lifetime risk of ovarian cancer of between 15 and 40 percent. Some mutations associated
with cancer, such as p53, BRCA1 and BRCA2, occur in mechanisms to correct errors in DNA.
These mutations are either inherited or acquired after birth. Presumably, they allow further
mutations, which allow uncontrolled division, lack of attachment, and metastasis to distant
organs [128]. However there is strong evidence of residual risk variation that goes well beyond
hereditary BRCA gene mutations between carrier families. This is caused by unobserved risk
factors [129]. This implicates environmental and other causes as triggers for breast cancers.
The inherited mutation in BRCA1 or BRCA2 genes can interfere with repair of DNA cross links
and DNA double strand breaks (known functions of the encoded protein) [130,131]. These
carcinogens cause DNA damage such as DNA cross links and double strand breaks that often
require repairs by pathways containing BRCA1 and BRCA2 [132,133]. However, mutations
in BRCA genes account for only 2 to 3 percent of all breast cancers [134]. Levin et al., states
that cancer may not be inevitable for all carriers of BRCA1 and BRCA2 mutations [135]. About
half of hereditary breast-ovarian cancer syndromes involve unknown genes. GATA-3 directly
controls the expression of Estrogen Receptor (ER) and other genes associated with epithelial
differentiation, and the loss of GATA-3 leads to loss of differentiation and poor prognosis due
to cancer cell invasion and metastasis [136].
Figure 2: Overview of signal transduction pathways involved in apoptosis. Mutations leading to loss of
apoptosis can lead to tumorigenesis.
do not receive any treatment [13] Depending on age at diagnosis, 20%-40% of women who
undergo mastectomy elect to have breast reconstruction, either with an implant, tissue from
another part of the body, or a combination of the two [137-142]. A woman considering breast
reconstruction should discuss this option with her breast surgeon prior to her mastectomy. The
plastic surgeon and the surgeon performing the mastectomy will work together to coordinate
treatment plans. Some types of reconstruction can begin during the mastectomy itself, so
reconstruction may influence the surgical facility (inpatient versus outpatient) and other
aspects of the mastectomy. Since 1999, the Women’s Health and Cancer Rights Act (WHCRA)
has required group health plans, insurance companies, and health maintenance organizations
that offer mastectomy coverage to also pay for reconstructive surgery after mastectomy.
Breast reconstruction is also covered by Medicare and Medicaid, though Medicaid benefits
vary by state.
Both BCS and mastectomy are usually accompanied by removal of regional lymph nodes
from the armpit to determine if the disease has spread beyond the breast. The presence of any
cancer cells in the lymph nodes will help determine the need for subsequent therapy and the
course it should take. Sentinel Lymph Node Biopsy (SLNB), in which selected lymph nodes are
removed and tested before any others are excised, reduces the need for full axillary lymph node
dissections among most women with no evidence of sentinel lymph node involvement [143].
Furthermore, findings from a recent clinical trial suggest that for some breast cancer patients
treated by lumpectomy and radiation, the axillary lymph node dissection can be avoided even
if cancer cells are found in one or two sentinel lymph nodes [144]. Prior to surgery, patients
should talk with their doctors to determine whether they intend to perform SLNB. If a woman
is eligible for SLNB and wishes to have this procedure, her breast cancer surgery should be
performed at a facility with a medical care team experienced with the technique. SLNB is
widely available in the US Surgery and radiation therapy involving the axillary lymph nodes
can lead to lymphedema, a serious swelling of the arm caused by retention of lymph fluid.
Breast cancer patients who undergo axillary lymph node dissection are about 3 times more
likely to develop lymphedema compared to those who have SLNB [145]. It has been estimated
that about 5% of patients with SLNB and 16%-18% of patients undergoing axillary lymph node
dissection following SLNB will develop clinically measurable lymphedema [146,147]. Some
evidence suggests that upper body exercises may reduce the risk and lessen the severity of
this condition [148].
Radiation therapy
Radiation therapy is the use of high-energy beams or particles to kill cancer cells. Radiation
may be used after potentially curative surgery to destroy cancer cells remaining in the breast,
chest wall, or underarm area. BCS is almost always followed by radiation therapy because it
has been shown to reduce the risk of cancer recurrence by about 50% and the risk of breast
cancer death by about 20%. 214 Although there is a higher risk of local recurrence (cancer
returning to the breast) with BCS, clinical trials with more than 20 years of follow-up data have
confirmed that a woman who chooses BCS and radiation will have the same expected long-
term survival as if she had chosen mastectomy [149-152]. Some mastectomy patients also
require radiation if their tumor is larger than 5 cm or when cancer is found in the lymph nodes.
Radiation can also be used to treat the symptoms of advanced breast cancer, especially when
it has spread to the central nervous system or bones. Radiation therapy may be administered
internally or externally. Some patients are treated with both types of radiation in combination.
The way the radiation therapy is given depends on the type, stage, and location of the tumor,
as well as doctor and patient preference. External beam radiation is the standard type of
radiation for women with breast cancer. Radiation is focused from a machine outside the
body on the area affected by cancer. This usually includes the whole breast and, depending
on the size and extent of the cancer, may include the chest wall and underarm area as well.
External beam radiation therapy is typically administered daily over a period of five to six
weeks; however, in recent studies, shortening the treatment to three weeks (referred to as
Accelerated Breast Irradiation or ABI) appears to be just as effective [153].
Internal radiation therapy, known as brachytherapy, is a form of Accelerated Partial Breast
Irradiation (APBI) which uses a radioactive substance sealed in needles, seeds, wires, or
catheters that are placed directly into or near the cancer. The ability to target radiation therapy
accurately has increased dramatically in recent decades, which has greatly diminished side
effects and can also reduce treatment time [154]. For example, the most common form of
brachytherapy used for breast cancer, intra-cavitary brachytherapy, is given for only five
days [155,156]. However, a recent retrospective study reported that women who were treated
with brachytherapy were more likely to have certain complications and receive a subsequent
mastectomy than those treated with whole breast radiation therapy [157]. Additional follow-
up data are needed to determine the long-term efficacy and risks associated with APBI and
to identify which patients are the best candidates. Clinical trials are also investigating other
forms of APBI that are designed to give radiation to a smaller segment of the breast, also over
a period of five days [158].
Systemic therapy
Systemic therapy is treatment that travels through the bloodstream and affects all parts of
the body, not just the cancer. These cancer drugs are given intravenously or orally. Systemic
therapy includes chemotherapy, hormone therapy, and targeted therapy, all of which work
through different mechanisms. For example, chemo- therapy drugs work by attacking cells
that grow quickly, such as cancer cells. Hormone therapy works by either blocking the body’s
natural hormones or lowering the levels of those hormones, which sometimes act to promote
cancer growth. Newer targeted drugs work by attacking specific parts of cancer cells. Systemic
therapy is treatment that travels through the bloodstream and affects all parts of the body,
not just the cancer.
These cancer drugs are injected into a vein or given by mouth. Systemic therapy includes
chemotherapy, hormone therapy, and targeted therapy, all of which work through different
mechanisms. For example, chemo-therapy drugs work by attacking cells that grow quickly,
such as cancer cells. Hormone therapy works by either blocking the body’s natural hormones
or lowering the levels of those hormones, which sometimes act to promote cancer growth.
Newer targeted drugs work by attacking specific parts of cancer cells.
When systemic treatment is given to patients before surgery, it is called neoadjuvant
therapy. It is often used to shrink the tumor enough to make surgical removal possible or
allow for less extensive surgery (such as BCS in women who would otherwise have required
mastectomy). Neoadjuvant systemic therapy has been found to be as effective as therapy given
after surgery to improve survival, disease progression, and limit recurrence [159]. Systemic
treatment given to patients after surgery is called adjuvant therapy. It is used to kill any
undetected tumor cells that were left behind during surgery or had migrated to other parts of
the body. The use of adjuvant systemic therapy is primarily determined by the tumor stage and
Research
Applications of nanoparticles in breast cancer nanotechnology
Introduction: The use of nanoparticles in cancer therapy is attractive for several reasons:
they exhibit unique pharmacokinetics, including minimal renal filtration; they have high
surface-to-volume ratios enabling modification with various surface functional groups that
home, internalize, or stabilize; and they may be constructed from a wide range of materials
used to encapsulate or solubilize therapeutic agents for drug delivery or to provide unique
optical, magnetic, and electrical properties for imaging and remote actuation. The topology of
a nanoparticle-core, coating, and surface functional groups-makes it particularly amenable
to modular design, whereby features and functional moieties may be interchanged or
combined. Although many functionalities of nanoparticles have been demonstrated, including
some clinically approved drug formulations and imaging agents [174,175], the consolidation
of these into multifunctional nanoparticles capable of targeting, imaging, and delivering
therapeutics is an exciting area of research that holds great promise for cancer therapy in the
future. Figure 3 schematically depicts a hypothetical multifunctional particle that has been
engineered to include many features such as the ability to target tumors, evade uptake by the
Reticuloendothelial System (RES), protect therapeutics that can be released on demand, act
as sensors of tumor responsiveness, and provide image contrast to visualize sites of disease
and monitor disease progression. Some of these features, such as targeting, leverage biological
machinery. Others are derived synthetically and enable external probing or manipulation that
is otherwise not feasible in biological systems. In this chapter, we review both bio-inspired and
synthetic nanoparticle functionalities that have been used in cancer therapy and address both
current efforts and future opportunities to combine these into multifunctional devices.
to achieve increased targeting efficiency, the size should be below 100 nanometers in diameter
and the surface of the nanoparticles should be hydrophilic in nature in order to circumvent
clearance by macrophages. The hydrophilic surface of the nanoparticles provides protection
against plasma protein adsorption to the surface, and this can be made possible by using
hydrophilic polymer coating, like Polyethylene Glycol (PEG), polysaccharides, poloxamines, or
poloxamers or by using block or branched amphiphilic copolymer [181,182]. Passive targeting
system is further classified into (a) leaky vasculature, (b) tumor microenvironment, and (c)
local drug application.
1. Leaky Vasculature: Maeda and Matsumura had first displayed the enhanced
permeation and retention effect by using polymer to form nanoparticles. Concept of enhanced
permeability and retention is based on two factors [183]. (a) The capillary endothelium system
in malignant tissue shows more permeation to macromolecules in comparison to normal tissue
endothelium; this makes circulating polymeric nanoparticles permeable into the tumor. (b)
Tumor lacks lymphatic drainage; hence, more drugs get accumulated in side tumor tissue. By
using a suitable biodegradable polymer, the concentration of drug gets 10 to 100 times higher
than that of free circulating drug.
2. Tumor Microenvironment: Tumor microenvironment provides the advantage of the
passive drug targeting. Active state of chemotherapeutic agent is conjugated with tumor
specific material and administered into the body. When this drug-polymer conjugate reaches
its desired destination, tumor environment converts into more active form. This phenomenon
is called tumor activated prodrug therapy. Mansour et al., had developed an albumin-bound
form of doxorubicin and showed in an in vitro study, that doxorubicin was efficiently cleaved
by matrix metalloproteniase-2 [184].
3. Local Drug Application: Direct application of the chemotherapeutic agent locally to
tumor site prevents systemic toxicity and increased concentration of drug at the tumor site.
Nomura et al., had synthesized intratumoral injection of mitomycin c-dextran conjugate; this
results in increased concentration of anticancer drug at tumor site and decreased systemic
toxicity [19]. Prabha and Labhasetwar had worked on nanoparticles-mediated wild-type p53
gene for breast cancer and observed the sustained and increased antiproliferative effect [185].
Active Targeting: By conjugating the nanoparticles with a drug to desired target site, an
active targeting may be achieved (Figure 6). Active targeting allows the increased accumulation
of the drug in cancer tissue. Directing the nanoparticles to the cancer cell can be done by
the following ways. This approach is basically based on the specific interactions, like lectin
carbohydrate, ligand receptor, and antibody-antigen [186].
BRCA mutation are associated with reduced DNA repair capacity due to the loss of BRCA
function and are hypothesized to rely more heavily on alternate compensatory DNA repair
processes for survival. The BRCA1 and BRCA2 proteins are best known for their important role
in homologous recombination, though BRCA1 has been implicated as having additional roles
in NER and BER [196,197].
PARP plays a central role in BER. It is required for the repair of oxidative damage associated
with breaks in single-strand DNA. If PARP is inhibited, repair-associated breaks result in
replication fork–mediated double-strand break formation, which requires BRCA1- and BRCA2-
associated recombination to resolve [198]. Thus, in tumors lacking intact BRCA function
due to loss of a second allele, chemical inhibition of PARP with a small molecule inhibitor is
postulated to be the “second hit” that renders the cell incapable of survival and mimics genetic
absence of PARP. However, in the host’s somatic tissues that are heterozygous for a BRCA
mutation and that maintain normal BRCA protein function, PARP inhibitors are thought to
have little or no effect.
Figure 7: Two distinct strategies for targeting poly (ADPribose) polymerase in the
treatment of cancer. HR=homologous recombination.
Figure 7: Two distinct strategies for targeting poly (ADPribose) polymerase in the treatment of cancer. HR=homologous recombination.
Drug Name Manufacture Route of Administration Tumors Targeted in Ongoing Clinical Trials Phase
Breast, ovarian, primary peritoneal, fallopian tube, colon,
Veliparib
Abbott Oral prostate, melanoma, HCC, hematologic malignancies/CLL, I,II
ABT-888
lymphoma, advanced solid tumors
BRCA1 and BRCA2 Mutation-associated Breast Cancer: The breast and ovarian cancer
syndromes caused by germline mutations in the BRCA1 and BRCA2 genes account for a
minority of breast cancer overall (approximately 5%), though it was this population that
provided clinical “proof of principle” of chemical synthetic lethality with PARP inhibitors. To
date, the majority of clinical investigation in BRCA mutation–associated cancer has been with
the oral PARP inhibitor olaparib (AstraZeneca), and few data exist for this drug outside of
this population. Fong and colleagues reported a phase I dose escalation study of olaparib in
patients with advanced refractory solid tumors [199]. A total of 60 patients were enrolled,
including 22 patients with known BRCA1 or BRCA2 mutations. Objective responses were
seen in this study, though only in patients with BRCA mutation–associated ovarian, breast, or
prostate cancer. Few patients had breast cancer, however, including 6 with no BRCA mutation
and 3 with documented BRCA2 mutations. No responses were observed in patients lacking
known BRCA mutations. At the 2009 American Society of Clinical Oncology (ASCO) Annual
Meeting, Tutt and colleagues presented the first phase II results of olaparib for the treatment
of women with BRCA1 and BRCA2 mutation–associated stage IIIB–IV breast cancer that had
progressed on at least 1 prior systemic chemotherapy regimen [200]. Two sequential cohorts
of 27 patients were enrolled at dose levels of 100 mg twice daily orally and 400 mg twice daily
orally every 28 days. Patients had received a median of 3 prior chemotherapy regimens for
advanced breast cancer, and 64% and 50% of patients had the triple-negative subtype of
breast cancer in the 100 mg and 400 mg cohorts, respectively. The objective response rate was
41% in the 400 mg cohort and 22% in the 100 mg cohort. One complete response was observed
with the higher dose. The responses seen in both triple-negative (Estrogen Receptor [ER]-
negative, Progesterone Receptor [PR]-negative and Human Epidermal Growth factor [HER] 2/
neu nonover-expressing) and nontriple-negative (but BRCA-mutation carrier) patients were
an important observation and suggest that the selection of patients based on shared DNA
repair defects due to BRCA mutations supersedes phenotypic subtype. To date, the activity
of olaparib in non-BRCA–mutant triple-negative breast cancer remains unclear. Gelmon and
colleagues recently reported results of a phase II study of single-agent olaparib 400 mg orally
twice daily [201]. In a cohort of fifteen BRCA-negatives, triple-negative breast cancer patients
with advanced disease, no objective responses were observed. Following these successes, many
companies are examining the efficacy of their PARP inhibitors in patients with BRCA mutation-
associated breast cancer. The majority of these studies are recruiting patients with advanced
BRCA mutation-associated breast cancer, though BSI-201 (BiPar Sciences/Sanofi-Aventis) is
being investigated in the neoadjuvant setting in combination with gemcitabine Gemzar, Eli Lilly)
and carboplatin for patients with stage I–IIIA BRCA1 and BRCA2 mutation–associated breast
cancer, regardless of breast cancer subtype. Pfizer has also recently launched a randomized
phase II study of cisplatin, with or without PF-01367338, in early stage BRCA1- or BRCA2-
positive patients with residual disease after neoadjuvant chemotherapy. BRCA1 and BRCA2
carriers with triple-negative or ER and/or PR-positive, HER2-negative breast cancer subtypes
are eligible for this study.
Another exciting area of PARP inhibitor development relates to their potential use as
chemoprevention agents in patients with high breast cancer risk. Studies examining this
hypothesis in BRCA mutation carriers are currently in development.
Triple-negative Breast Cancer: Sporadic triple-negative breast cancers share many
pathologic and molecular features with breast cancers caused by hereditary BRCA1 mutations,
including basal-like gene expression, high histologic grade, frequent p53 mutations,
cytogenetic abnormalities, increased genomic instability, and EGFR overexpression [202-
204]. Based on these similarities and the role of BRCA1 in multiple DNA repair pathways,
the hypothesis emerged that sporadic triple-negative breast tumors may possess similar DNA
repair deficiencies and exhibit similar chemosensitivities as BRCA1 mutation-associated breast
tumors. Interestingly, basal-like breast cancer cell lines, like BRCA1-deficient cancer cell
lines, are more sensitive to oxidative DNA damage compared to luminal breast tumor cells or
normal breast epithelial cells, and are deficient in BER [197]. Similar to BRCA1-deficient cells,
basal-like tumor cells also demonstrate increased sensitivity to PARP inhibition, cisplatin, and
gemcitabine, [197,205] and PARP1 has been observed to be overexpressed in triple-negative
breast cancer [206]. These observations provided the rationale to investigate DNA damaging
chemotherapies and PARP inhibitors in triple-negative breast cancer patients. At ASCO 2009,
O’Shaughnessy and colleagues reported the first clinical results exploring the role of PARP
inhibition in the treatment of sporadic triple-negative advanced breast cancer [207]. In this
randomized phase II trial, women with advanced breast cancer were treated with intravenous
gemcitabine 1,000 mg/m2 and intravenous carboplatin area under the concentration of 2 on
days 1 and 8, with or without the PARP inhibitor BSI-201 dosed at 5.6 mg/kg intravenously
on days 1, 4, 8, and 11. A total of 123 women treated with up to 2 prior chemotherapy
regimens for metastatic disease were enrolled. This study demonstrated improvements in the
clinical benefit rate (21% vs 62%; P=0.0002), overall response rate (16% vs 48%; P=0.002),
median progression-free survival (3.3 vs 6.9 months; Hazard Ratio [HR], 0.342; P<0.0001)
and median overall survival (5.7 vs 9.2 months; HR, 0.348; P=0.0005) among patients who
received BSI-201. Prolongation of survival is rarely observed in clinical trials of women with
advanced breast cancer and, as such, these results were received with great enthusiasm in the
oncology community. The overall survival data were updated at the 2009 San Antonio Breast
Cancer Symposium, and they continued to show a significant survival advantage among
women treated with BSI-201 in an intention-to-treat analysis (7.7 vs 12.2 months; HR, 0.50;
P=0.005) [208]. Of particular note, approximately 40% of women treated on the control arm
crossed over to receive BSI-201 at the time of disease progression. In July 2009, a randomized
phase III registration study of gemcitabine and carboplatin, with or without BSI-201, was
launched to examine the safety and efficacy of this combination in a larger patient population
(n=420). Accrual was extremely rapid, with the study closing to accrual in February 2010. A
subsequent study examining this combination in advanced triple-negative breast cancer has
recently been launched in Europe and is testing gemcitabine and carboplatin with 2 different
doses and schedules of BSI-201: 11.2 mg/kg intravenously on days 1 and 8 versus 5.6 mg/kg
Principles and Practice of Cancer Prevention and Control 27
Edited by: Redhwan Ahmed Al-Naggar
eBooks
intravenously on days 1, 4, 8, and 11. Other ongoing PARP inhibitor studies in triple-negative
breast cancer include a phase II study of neoadjuvant gemcitabine, carboplatin, and BSI-201
in women with stage I–IIIA triple-negative breast cancer and a phase I/II study of cisplatin,
with or without olaparib, as neoadjuvant therapy in women with early-stage, triple-negative
breast cancer. Pfizer has recently launched a randomized phase II study of cisplatin with
or without PF-01367338 in early-stage triple-negative breast cancer patients with residual
disease after neoadjuvant chemotherapy. ABT-888 (Veliparib, Abbott) is being investigated in
combination with weekly paclitaxel and every 3 weekly carboplatin followed by doxorubicin
and cyclophosphamide in the neoadjuvant treatment of early breast cancer as part of the
multicenter I-SPY2 TRIAL (Investigation of Serial Studies to Predict Your Therapeutic Response
With Imaging and Molecular Analysis). Data with single-agent BSI-201 in breast cancer, in
particular triple-negative breast cancer, are limited. Likewise, data with single-agent olaparib
in sporadic triple negative breast cancer are also limited. Though PARP inhibitors were
investigated in triple-negative disease based on the hypothesis that this breast cancer subtype
is “BRCA-like,” it is unknown at present whether the activity seen in the BSI-201 study was a
result of a synthetic lethal interaction between a genetic defect characteristic of this subtype
and the PARP inhibitor, a result of enhanced chemosensitivity of gemcitabine and carboplatin
in combination with BSI-201, or possibly a combination of both. It is quite possible that the
mechanism may vary by patient, and identification of a means to distinguish these patients
and their responses is a high priority of research.
References
1. Allred DC (2010) Ductal carcinoma in situ: terminology, classification, and natural history. J Natl Cancer Inst Monogr 2010: 134-
138.
2. Solin LJ, Gray R, Baehner FL, Butler SM, Hughes LL, et al. (2013) A multigene expression assay to predict local recurrence risk
for ductal carcinoma in situ of the breast. J Natl Cancer Inst 105: 701-710.
3. Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FG, et al., (eds.) (2010) AJCC Cancer Staging Manual. (7thedn), Springer,
New York, USA.
4. Young JL Jr, Roffers SD, Ries LAG, Fritz AG, Hurlbut AA (eds.). (2001) SEER Summary Staging Manual - 2000: Codes and Coding
Instructions, National Cancer Institute, NIH Pub. No. 01-4969, Bethesda, MD.
5. Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, et al. (2000) Molecular portraits of human breast tumours. Nature 406:
747-752.
6. Reis-Filho JS, Pusztai L (2011) Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet
378: 1812-1823.
7. Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thürlimann B, et al. (2011) Strategies for subtypes--dealing with the diversity of
breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011.
Ann Oncol 22: 1736-1747.
8. Perou CM, Børresen-Dale AL (2011) Systems biology and genomics of breast cancer. Cold Spring Harb Perspect Biol 3.
9. Blows FM, Driver KE, Schmidt MK, Broeks A, van Leeuwen FE, et al. (2010) Subtyping of breast cancer by immunohistochemistry
to investigate a relationship between subtype and short and long term survival: a collaborative analysis of data for 10,159 cases
from 12 studies. PLoS Med 7: 1000279.
10. Voduc KD, Cheang MC, Tyldesley S, Gelmon K, Nielsen TO, et al. (2010) Breast cancer subtypes and the risk of local and regional
relapse. J Clin Oncol 28: 1684-1691.
11. Cheang MC, Chia SK, Voduc D, Gao D, Leung S, et al. (2009) Ki67 index, HER2 status, and prognosis of patients with luminal B
breast cancer. J Natl Cancer Inst 101: 736-750.
12. Carey LA, Perou CM, Livasy CA, Dressler LG, Cowan D, et al. (2006) Race, breast cancer subtypes, and survival in the Carolina
Breast Cancer Study. JAMA 295: 2492-2502.
13. Merck Manual of Diagnosis and Therapy (2003) Breast Disorders: Breast Cancer. Retrieved 2008-02-05.
14. American Cancer Society (2007) Cancer Facts & Figures 2007. Archived from the original on 10 April 2007. Retrieved 2007-04-26.
16. Lacroix M (2006) Significance, detection and markers of disseminated breast cancer cells. Endocr Relat Cancer 13: 1033-1067.
17. McCann, Executive publisher , Judith A Shilling (2008) Nursing.. Ambler, PA: Lippincott Williams & Wilkins. P: 99. ISBN 978-1-
58255-668-0.
18. Kushi LH, Doyle C, McCullough M, Rock CL, Demark-Wahnefried W, et al. (2012) American Cancer Society Guidelines on nutrition
and physical activity for cancer prevention: reducing the risk of cancer with healthy food choices and physical activity. CA Cancer
J Clin 62: 30-67.
19. Collaborative Group on Hormonal Factors in Breast Cancer (2001) Familial breast cancer: collaborative reanalysis of individual
data from 52 epidemiological studies including 58,209 women with breast cancer and 101,986 women without the disease. Lancet
358:1389-1399.
20. Schwartz GF, Hughes KS, Lynch HT, Fabian CJ, Fentiman IS, et al. (2008) Proceedings of the international consensus conference
on breast cancer risk, genetics, & risk management, April, 2007. Cancer 113: 2627-2637.
21. Antoniou A, Pharoah PD, Narod S, Risch HA, Eyfjord JE, et al. (2003) Average risks of breast and ovarian cancer associated with
BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum
Genet 72: 1117-1130.
22. Chen S, Parmigiani G (2007) Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol 25: 1329-1333.
23. Turnbull C, Rahman N (2008) Genetic predisposition to breast cancer: past, present, and future. Annu Rev Genomics Hum Genet
9: 321-345.
24. Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, et al. (2000) Environmental and heritable factors in the causation of
cancer--analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med 343: 78-85.
25. Robson ME, Storm CD, Weitzel J, Wollins DS, American Society of Clinical Oncology. et al. (2010) American Society of Clinical
Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol 28: 893-901.
26. American Cancer Society. Cancer Facts & Figures 2009. Atlanta: American Cancer Society, 2009.
27. Schottenfeld D, Beebe-Dimmer J Multiple Primary Cancers. In: Schottenfeld D, Fraumeni JF, Jr. (eds.). Cancer Prevention and
Early Detec-tion Third Edition. Oxford University Press, New York USA. 2006: 1269-1280.
28. Kilbride KE, Newman LA (2010) Chapter 25: Lobular carcinoma in situ: Clinical management. In: Harris JR, Lippman ME, Morrow
M, Osborne CK (eds.). Diseases of the Breast. (4th ed), Lippincott Williams & Wilkins.
29. Hartmann LC, Sellers TA, Frost MH, Lingle WL, Degnim AC, et al. (2005) Benign breast disease and the risk of breast cancer. N
Engl J Med 353: 229-237.
30. London SJ, Connolly JL, Schnitt SJ, Colditz GA (1992) A prospective study of benign breast disease and the risk of breast cancer.
JAMA 267: 941-944.
31. Wang J, Costantino JP, Tan-Chiu E, Wickerham DL, Paik S, et al. (2004) Lower-category benign breast disease and the risk of
invasive breast cancer. J Natl Cancer Inst 96: 616-620.
32. Kabat GC, Jones JG, Olson N, Negassa A, Duggan C, et al. (2010) A multi-center prospective cohort study of benign breast
disease and risk of subsequent breast cancer. Cancer Causes Control 21: 821-828.
33. Tamimi RM, Byrne C, Colditz GA, Hankinson SE (2007) Endogenous hormone levels, mammographic density, and subsequent risk
of breast cancer in postmenopausal women. J Natl Cancer Inst 99: 1178-1187.
34. Boyd NF, Dite GS, Stone J, Gunasekara A, English DR, et al. (2002) Heritability of mammographic density, a risk factor for breast
cancer. N Engl J Med 347: 886-894.
35. Ursin G, Lillie EO, Lee E, Cockburn M, Schork NJ, et al. (2009) The relative importance of genetics and environment on
mammographic density. Cancer Epidemiol Biomarkers Prev 18: 102-112.
36. Harris HR, Tamimi RM, Willett WC, Hankinson SE, Michels KB (2011) Body size across the life course, mammographic density,
and risk of breast cancer. Am J Epidemiol 174: 909-918.
37. Boyd NF, Martin LJ, Yaffe MJ, Minkin S (2011) Mammographic density and breast cancer risk: current understanding and future
prospects. Breast Cancer Res 13: 223.
38. Boyd NF, Guo H, Martin LJ, Sun L, Stone J, et al. (2007) Mammographic density and the risk and detection of breast cancer. N
Engl J Med 356: 227-236.
39. Cummings SR, Tice JA, Bauer S, Browner WS, Cuzick J, et al. (2009) Prevention of breast cancer in postmenopausal women:
approaches to estimating and reducing risk. J Natl Cancer Inst 101: 384-398.
40. Brower V (2013) Breast density legislation fueling controversy. J Natl Cancer Inst 105: 510-511.
41. Fuhrman BJ, Schairer C, Gail MH, Boyd-Morin J, Xu X, et al. (2012) Estrogen metabolism and risk of breast cancer in
postmenopausal women. J Natl Cancer Inst 104: 326-339.
42. Key TJ (2011) Endogenous oestrogens and breast cancer risk in premenopausal and postmenopausal women. Steroids 76: 812-
815.
43. Hankinson SE, Eliassen AH (2007) Endogenous estrogen, testosterone and progesterone levels in relation to breast cancer risk.
J Steroid Biochem Mol Biol 106: 24-30.
44. Endogenous Hormones and Breast Cancer Collaborative Group, Key TJ, Appleby PN, Reeves GK, Roddam AW, et al. (2011)
Circulating sex hormones and breast cancer risk factors in postmenopausal women: reanalysis of 13 studies. Br J Cancer 105:
709-722.
45. Zeleniuch-Jacquotte A, Afanasyeva Y, Kaaks R, Rinaldi S, Scarmo S, et al. (2012) Premenopausal serum androgens and breast
cancer risk: a nested case-control study. Breast Cancer Res 14: R32.
46. Dorgan JF, Stanczyk FZ, Kahle LL, Brinton LA (2010) Prospective case-control study of premenopausal serum estradiol and
testosterone levels and breast cancer risk. Breast Cancer Res 12: 98.
47. Kaaks R, Berrino F, Key T, Rinaldi S, Dossus L, et al. (2005) Serum sex steroids in premenopausal women and breast cancer risk
within the European Prospective Investigation into Cancer and Nutrition (EPIC). J Natl Cancer Inst 97: 755-765.
48. Walker K, Bratton DJ, Frost C (2011) Premenopausal endogenous oestrogen levels and breast cancer risk: a meta-analysis. Br J
Cancer 105: 1451-1457.
49. Kelsey JL, Gammon MD, John EM (1993) Reproductive factors and breast cancer. Epidemiol Rev 15: 36-47.
50. Collaborative Group on Hormonal Factors in Breast Cancer (2012) Menarche, menopause, and breast cancer risk: individual
participant meta-analysis, including 118 964 women with breast cancer from 117 epidemiological studies. Lancet Oncol 13: 1141-
1151.
51. Lambe M, Hsieh C, Trichopoulos D, Ekbom A, Pavia M, et al. (1994) Transient increase in the risk of breast cancer after giving
birth. N Engl J Med 331: 5-9.
52. Yang XR, Chang-Claude J, Goode EL, Couch FJ, Nevanlinna H, et al. (2011) Associations of breast cancer risk factors with tumor
subtypes: a pooled analysis from the Breast Cancer Association Consortium studies. J Natl Cancer Inst 103: 250-263.
53. Phipps AI, Buist DS, Malone KE, Barlow WE, Porter PL, et al. (2011) Reproductive history and risk of three breast cancer subtypes
defined by three biomarkers. Cancer Causes Control 22: 399-405.
54. Faupel-Badger JM, Arcaro KF, Balkam JJ, Eliassen AH, Hassiotou F, et al. (2013) Postpartum remodeling, lactation, and breast
cancer risk: summary of a National Cancer Institute-sponsored workshop. J Natl Cancer Inst 105: 166-174.
55. Collaborative Group on Hormonal Factors in Breast Cancer (2002) Breast cancer and breastfeeding: collaborative reanalysis of
individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women
without the disease. Lancet 360: 187-195.
56. Grenier D, Cooke AL, Lix L, Metge C, Lu H, et al. (2011) Bone mineral density and risk of postmenopausal breast cancer. Breast
Cancer Res Treat 126: 679-686.
57. Chen Z, Arendell L, Aickin M, Cauley J, Lewis CE, et al. (2008) Hip bone density predicts breast cancer risk independently of Gail
score: results from the Women’s Health Initiative. Cancer 113: 907-915.
58. Zhang Y, Kiel DP, Kreger BE, Cupples LA, Ellison RC, et al. (1997) Bone mass and the risk of breast cancer among postmenopausal
women. N Engl J Med 336: 611-617.
59. Zmuda JM, Cauley JA, Ljung BM, Bauer DC, Cummings SR, et al. (2001) Bone mass and breast cancer risk in older women:
differences by stage at diagnosis. J Natl Cancer Inst 93: 930-936.
60. Kerlikowske K, Shepherd J, Creasman J, Tice JA, Ziv E, et al. (2005) Are breast density and bone mineral density independent risk
factors for breast cancer? J Natl Cancer Inst 97: 368-374.
61. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, et al. (2002) Risks and benefits of estrogen plus progestin
in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA 288:
321-333.
62. Chlebowski RT, Anderson GL, Gass M, Lane DS, Aragaki AK, et al. (2010) Estrogen plus progestin and breast cancer incidence
and mortality in postmenopausal women. JAMA 304: 1684-1692.
63. Beral V, Reeves G, Bull D, Green J; Million Women Study Collaborators (2011) Breast cancer risk in relation to the interval between
menopause and starting hormone therapy. J Natl Cancer Inst 103: 296-305.
64. Chlebowski RT, Anderson GL (2011) The influence of time from menopause and mammography on hormone therapy-related
breast cancer risk assessment. J Natl Cancer Inst 103: 284-285.
65. Prentice RL, Chlebowski RT, Stefanick ML, Manson JE, Pettinger M, et al. (2008) Estrogen plus progestin therapy and breast
cancer in recently postmenopausal women. Am J Epidemiol 167: 1207-1216.
66. Calle EE, Feigelson HS, Hildebrand JS, Teras LR, Thun MJ, et al. (2009) Postmenopausal hormone use and breast cancer
associations differ by hormone regimen and histologic subtype. Cancer 115: 936-945.
67. Anderson GL, Chlebowski RT, Aragaki AK, Kuller LH, Manson JE, et al. (2012) Conjugated equine oestrogen and breast cancer
incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women’s Health Initiative
randomised placebo-controlled trial. Lancet Oncol 13: 476-486.
68. Bakken K, Fournier A, Lund E, Waaseth M, Dumeaux V, et al. (2011) Menopausal hormone therapy and breast cancer risk: impact
of different treatments. The European Prospective Investigation into Cancer and Nutrition. Int J Cancer 128: 144-156.
69. World Cancer Research Fund/American Institute for Cancer Research (2007) Food, Nutrition, Physical Activity, and the Prevention
of Cancer: a Global Perspective. Washington, DC: AICR.
70. La Vecchia C, Giordano SH, Hortobagyi GN, Chabner B (2011) Overweight, obesity, diabetes, and risk of breast cancer: interlocking
pieces of the puzzle. Oncologist 16: 726-729.
71. Boyle P, Boniol M, Koechlin A, Robertson C, Valentini F, et al. (2012) Diabetes and breast cancer risk: a meta-analysis. Br J Cancer
107: 1608-1617.
72. Goodwin PJ, Thompson AM, Stambolic V (2012) Diabetes, metformin, and breast cancer: lilac time? J Clin Oncol 30: 2812-2814.
73. Nelson HD, Zakher B, Cantor A, Fu R, Griffin J, et al. (2012) Risk factors for breast cancer for women aged 40 to 49 years: a
systematic review and meta-analysis. Ann Intern Med 156: 635-648.
74. Ritte R, Lukanova A, Berrino F, Dossus L, Tjønneland A, et al. (2012) Adiposity, hormone replacement therapy use and breast
cancer risk by age and hormone receptor status: a large prospective cohort study. Breast Cancer Res 14: 76.
75. Rose DP, Vona-Davis L (2010) Interaction between menopausal status and obesity in affecting breast cancer risk. Maturitas 66:
33-38.
76. Eliassen AH, Colditz GA, Rosner B, Willett WC, Hankinson SE (2006) Adult weight change and risk of postmenopausal breast
cancer. JAMA 296: 193-201.
77. Teras LR, Goodman M, Patel AV, Diver WR, Flanders WD, et al. (2011) Weight loss and postmenopausal breast cancer in a
prospective cohort of overweight and obese US women. Cancer Causes Control 22: 573-579.
78. Wolin KY, Colditz GA (2008) Can weight loss prevent cancer? Br J Cancer 99: 995-999.
79. Eliassen AH, Hankinson SE, Rosner B, Holmes MD, Willett WC (2010) Physical activity and risk of breast cancer among
postmenopausal women. Arch Intern Med 170: 1758-1764.
80. Peters TM, Schatzkin A, Gierach GL, Moore SC, Lacey JV Jr, et al. (2009) Physical activity and postmenopausal breast cancer risk
in the NIH-AARP diet and health study. Cancer Epidemiol Biomarkers Prev 18: 289-296.
81. Friedenreich CM, Cust AE (2008) Physical activity and breast cancer risk: impact of timing, type and dose of activity and population
subgroup effects. Br J Sports Med 42: 636-647.
82. Neilson HK, Friedenreich CM, Brockton NT, Millikan RC (2009) Physical activity and postmenopausal breast cancer: proposed
biologic mechanisms and areas for future research. Cancer Epidemiol Biomarkers Prev 18: 11-27.
83. Vera-Ramirez L, Ramirez-Tortosa MC, Sanchez-Rovira P, Ramirez-Tortosa CL, Granados-Principal S, et al. (2013) Impact of diet
on breast cancer risk: a review of experimental and observational studies. Crit Rev Food Sci Nutr 53: 49-75.
84. Thomson CA (2012) Diet and breast cancer: understanding risks and benefits. Nutr Clin Pract 27: 636-650.
85. Alexander DD, Morimoto LM, Mink PJ, Lowe KA (2010) Summary and meta-analysis of prospective studies of animal fat intake and
breast cancer. Nutr Res Rev 23: 169-179.
86. Linos E, Willett WC, Cho E, Frazier L (2010) Adolescent diet in relation to breast cancer risk among premenopausal women.
Cancer Epidemiol Biomarkers Prev 19: 689-696.
87. Wu AH, Yu MC, Tseng CC, Pike MC (2008) Epidemiology of soy exposures and breast cancer risk. Br J Cancer 98: 9-14.
88. Jung S, Spiegelman D, Baglietto L, Bernstein L, Boggs DA, et al. (2013) Fruit and vegetable intake and risk of breast cancer by
hormone receptor status. J Natl Cancer Inst 105: 219-236.
89. Aune D, Chan DS, Vieira AR, Rosenblatt DA, Vieira R, et al. (2012) Fruits, vegetables and breast cancer risk: a systematic review
and meta-analysis of prospective studies. Breast Cancer Res Treat 134: 479-493.
90. Hui C, Qi X, Qianyong Z, Xiaoli P, Jundong Z, et al. (2013) Flavonoids, flavonoid subclasses and breast cancer risk: a meta-
analysis of epidemiologic studies. PLoS One 8: e54318.
91. Hamajima N, Hirose K, Tajima K, Rohan T, Calle EE, et al. (2002) Alcohol, tobacco and breast cancer--collaborative reanalysis
of individual data from 53 epidemiological studies, including 58,515 women with breast cancer and 95,067 women without the
disease. Br J Cancer 87: 1234-1245.
92. Chen WY, Rosner B, Hankinson SE, Colditz GA, Willett WC (2011) Moderate alcohol consumption during adult life, drinking
patterns, and breast cancer risk. JAMA 306: 1884-1890.
93. Allen NE, Beral V, Casabonne D, Kan SW, Reeves GK, et al. (2009) Moderate alcohol intake and cancer incidence in women. J
Natl Cancer Inst 101: 296-305.
94. Singletary KW, Gapstur SM (2001) Alcohol and breast cancer: review of epidemiologic and experimental evidence and potential
mechanisms. JAMA 286: 2143-2151.
95. Li CI, Chlebowski RT, Freiberg M, Johnson KC, Kuller L, et al. (2010) Alcohol consumption and risk of postmenopausal breast
cancer by subtype: the women’s health initiative observational study. J Natl Cancer Inst 102: 1422-1431.
96. Suzuki R, Orsini N, Mignone L, Saji S, Wolk A (2008) Alcohol intake and risk of breast cancer defined by estrogen and progesterone
receptor status--a meta-analysis of epidemiological studies. Int J Cancer 122: 1832-1841.
97. Secretan B, Straif K, Baan R, Grosse Y, El Ghissassi F, et al. (2009) A review of human carcinogens--Part E: tobacco, areca nut,
alcohol, coal smoke, and salted fish. Lancet Oncol 10: 1033-1034.
98. Gaudet MM, Gapstur SM, Sun J, Diver WR, Hannan LM, et al. (2013) Active smoking and breast cancer risk: original cohort data
and meta-analysis. J Natl Cancer Inst 105: 515-525.
99. Luo J, Margolis KL, Wactawski-Wende J, Horn K, Messina C, et al. (2011) Association of active and passive smoking with risk of
breast cancer among postmenopausal women: a prospective cohort study. BMJ 342: 1016.
100. DeRoo LA, Cummings P, Mueller BA (2011) Smoking before the first pregnancy and the risk of breast cancer: a meta-
analysis. Am J Epidemiol 174: 390-402.
101. US Department of Health and Human Services (2006) The Health Consequences of Involuntary Exposure to Tobacco Smoke: A
Report of the Surgeon General. Department of Health and Human Services, Atlanta, GA, USA.
102. Pirie K, Beral V, Peto R, Roddam A, Reeves G, et al. (2008) Passive smoking and breast cancer in never smokers: prospective
study and meta-analysis. Int J Epidemiol 37: 1069-1079.
103. Xue F, Willett WC, Rosner BA, Hankinson SE, Michels KB (2011) Cigarette smoking and the incidence of breast cancer. Arch
Intern Med 171: 125-133.
104. Anderson LN, Cotterchio M, Mirea L, Ozcelik H, Kreiger N (2012) Passive cigarette smoke exposure during various periods of life,
genetic variants, and breast cancer risk among never smokers. Am J Epidemiol 175: 289-301.
105. Collaborative Group on Hormonal Factors in Breast Cancer (1996) Breast cancer and hormonal contraceptives: collaborative
reanalysis of individual data on 53 297 women with breast cancer and 100 239 women without breast cancer from 54
epidemiological studies. Lancet 347: 1713-1727.
106. Preston DL, Mattsson A, Holmberg E, Shore R, Hildreth NG, et al. (2002) Radiation effects on breast cancer risk: a pooled analysis
of eight cohorts. Radiat Res 158: 220-235.
107. Travis LB, Hill DA, Dores GM, Gospodarowicz M, van Leeuwen FE, et al. (2003) Breast cancer following radiotherapy and
chemotherapy among young women with Hodgkin disease. JAMA 290: 465-475.
108. Russo J, Hu YF, Yang X, Russo IH (2000) Developmental, cellular, and molecular basis of human breast cancer. J Natl Cancer
Inst Monogr 17-37.
109. Clemons M, Loijens L, Goss P (2000) Breast cancer risk following irradiation for Hodgkin’s disease. Cancer Treat Rev 26: 291-
302.
110. Titus-Ernstoff L, Hatch EE, Hoover RN, Palmer J, Greenberg ER, et al. (2001) Long-term cancer risk in women given
diethylstilbestrol (DES) during pregnancy. Br J Cancer 84: 126-133.
111. Hoover RN, Hyer M, Pfeiffer RM, Adam E, Bond B, et al. (2011) Adverse health outcomes in women exposed in utero to
diethylstilbestrol. N Engl J Med 365: 1304-1314.
112. Salehi F, Turner MC, Phillips KP, Wigle DT, Krewski D, et al. (2008) Review of the etiology of breast cancer with special attention
to organochlorines as potential endocrine disruptors. J Toxicol Environ Health B Crit Rev 11: 276-300.
113. Calle EE, Frumkin H, Henley SJ, Savitz DA, Thun MJ (2002) Organochlorines and breast cancer risk. CA Cancer J Clin 52: 301-
309.
114. Lopez-Cervantes M, Torres-Sanchez L, Tobias A, Lopez-Carrillo L (2004) Dichlorodiphenyldichloroethane burden and breast
cancer risk: a meta-analysis of the epidemiologic evidence. Environ Health Perspect 112: 207-214.
115. Gammon MD, Wolff MS, Neugut AI, Eng SM, Teitelbaum SL, et al. (2002) Environmental toxins and breast cancer on Long Island.
II. Organochlorine compound levels in blood. Cancer Epidemiol Biomarkers Prev 11: 686-697.
116. Brody JG, Moysich KB, Humblet O, Attfield KR, Beehler GP, et al. (2007) Environmental pollutants and breast cancer: epidemiologic
studies. Cancer 109: 2667-2711.
117. Steenland K, Whelan E, Deddens J, Stayner L, Ward E (2003) Ethylene oxide and breast cancer incidence in a cohort study of
7576 women (United States). Cancer Causes Control 14: 531-539.
118. Jia Y, Lu Y, Wu K, Lin Q, Shen W, et al. (2013) Does night work increase the risk of breast cancer? A systematic review and meta-
119. Kamdar BB, Tergas AI, Mateen FJ, Bhayani NH, Oh J (2013) Night-shift work and risk of breast cancer: a systematic review and
meta-analysis. Breast Cancer Res Treat 138: 291-301.
120. Straif K, Baan R, Grosse Y, Secretan B, El Ghissassi F, et al. (2007) Carcinogenicity of shift-work, painting, and fire-fighting.
Lancet Oncol 8: 1065-1066.
121. International Agency for Research on Cancer (2010) IARC monographs on the evaluation of carcinogenic risks to humans.
Volume 98. Painting, Firefighting, and Shift work. Lyon, France.
122. Adrian Lee, Carlos Arteaga, Ian Smith (2009) 32nd Annual CTRC-AACR San Antonio Breast Cancer Symposium. Sunday
Morning Year-End Review, San Antonio, Texan, USA.
123. Cavalieri E, Chakravarti D, Guttenplan J, Hart E, Ingle J, et al. (2006) Catechol estrogen quinones as initiators of breast and other
human cancers: implications for biomarkers of susceptibility and cancer prevention. Biochim Biophys Acta 1766: 63-78.
124. Farlex (2005) “Immunological Surveilliance”. The Free Dictionary. Retrieved 2008-02-10.
125. Haslam SZ, Woodward TL (2003) Host microenvironment in breast cancer development: epithelial-cell-stromal-cell interactions
and steroid hormone action in normal and cancerous mammary gland. Breast Cancer Res 5: 208-215.
126. Wiseman BS, Werb Z (2002) Stromal effects on mammary gland development and breast cancer. Science 296: 1046-1049.
127. Jardé T, Perrier S, Vasson MP, Caldefie-Chézet F (2011) Molecular mechanisms of leptin and adiponectin in breast cancer. Eur
J Cancer 47: 33-43.
128. Dunning AM, Healey CS, Pharoah PD, Teare MD, Ponder BA, et al. (1999) A systematic review of genetic polymorphisms and
breast cancer risk. Cancer Epidemiol Biomarkers Prev 8: 843-854.
129. Begg CB, Haile RW, Borg A, Malone KE, Concannon P, et al. (2008) Variation of breast cancer risk among BRCA1/2 carriers.
JAMA 299: 194-201.
130. Patel KJ, Yu VP, Lee H, Corcoran A, Thistlethwaite FC, et al. (1998) Involvement of Brca2 in DNA repair. Mol Cell 1: 347-357.
131. Marietta C, Thompson LH, Lamerdin JE, Brooks PJ (2009) Acetaldehyde stimulates FANCD2 monoubiquitination, H2AX
phosphorylation, and BRCA1 phosphorylation in human cells in vitro: implications for alcohol-related carcinogenesis. Mutat Res
664: 77-83.
132. Theruvathu JA, Jaruga P, Nath RG, Dizdaroglu M, Brooks PJ (2005) Polyamines stimulate the formation of mutagenic 1,N2-
propanodeoxyguanosine adducts from acetaldehyde. Nucleic Acids Res 33: 3513-3520.
133. Wooster R, Weber BL (2003) Breast and ovarian cancer. N Engl J Med 348: 2339-2347.
134. Levin B, Lech D, Friedenson B (2012) Evidence that BRCA1- or BRCA2-associated cancers are not inevitable. Mol Med 18:
1327-1337.
135. Kouros-Mehr H, Kim JW, Bechis SK, Werb Z (2008) GATA-3 and the regulation of the mammary luminal cell fate. Curr Opin Cell
Biol 20: 164-170.
136. Saslow D, Hannan J, Osuch J, Alciati MH, Baines C, et al. (2004) Clinical breast examination: practical recommendations for
optimizing performance and reporting. CA Cancer J Clin 54: 327-344.
137. Kruper L, Holt A, Xu XX, Duan L, Henderson K, et al. (2011) Disparities in reconstruction rates after mastectomy: patterns of care
and factors associated with the use of breast reconstruction in Southern California. Ann Surg Oncol 18: 2158-2165.
138. Tseng WH, Stevenson TR, Canter RJ, Chen SL, Khatri VP, et al. (2010) Sacramento area breast cancer epidemiology study: use
of postmastectomy breast reconstruction along the rural-to-urban continuum. Plast Reconstr Surg. 126: 1815-1824.
139. Reuben BC, Manwaring J, Neumayer LA (2009) Recent trends and predictors in immediate breast reconstruction after mastectomy
in the United States. Am J Surg 198: 237-243.
140. Alderman AK, Wei Y, Birkmeyer JD (2006) Use of breast reconstruction after mastectomy following the Women’s Health and
Cancer Rights Act. JAMA 295: 387-388.
141. Rosson GD, Singh NK, Ahuja N, Jacobs LK, Chang DC (2008) Multilevel analysis of the impact of community vs patient factors on
access to immediate breast reconstruction following mastectomy in Maryland. Arch Surg. 143: 1076-1081.
142. Christian CK, Niland J, Edge SB, Ottesen RA, Hughes ME, et al. (2006) A multi-institutional analysis of the socioeconomic
determinants of breast reconstruction: a study of the National Comprehensive Cancer Network. Ann Surg 243: 241-249.
143. Kim T, Giuliano AE, Lyman GH (2006) Lymphatic mapping and sentinel lymph node biopsy in early-stage breast carcinoma: a
metaanalysis. Cancer 106: 4-16.
144. Giuliano AE, Hunt KK, Ballman KV, Beitsch PD, Whitworth PW, et al. (2011) Axillary dissection vs no axillary dissection in women
with invasive breast cancer and sentinel node metastasis: a randomized clinical trial. JAMA 305: 569-575.
145. Tsai RJ, Dennis LK, Lynch CF, Snetselaar LG, Zamba GK, et al. (2009) The risk of developing arm lymphedema among breast
cancer survivors: a meta-analysis of treatment factors. Ann Surg Oncol 16: 1959-1972.
146. Wernicke AG, Shamis M, Sidhu KK, Turner BC, Goltser Y, et al. (2013) Complication rates in patients with negative axillary nodes
10 years after local breast radiotherapy after either sentinel lymph node dissection or axillary clearance. Am J Clin Oncol 36:
12-19.
147. McLaughlin SA, Wright MJ, Morris KT, Sampson MR, Brockway JP, et al. (2008) Prevalence of lymphedema in women with breast
cancer 5 years after sentinel lymph node biopsy or axillary dissection: patient perceptions and precautionary behaviors. J Clin
Oncol 26: 5220-5226.
148. Lawenda BD, Mondry TE, Johnstone PA (2009) Lymphedema: a primer on the identification and management of a chronic
condition in oncologic treatment. CA Cancer J Clin 59: 8-24.
149. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG), Darby S, McGale P, Correa C, Taylor C, et al. (2011) Effect of
radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: meta-analysis of individual
patient data for 10,801 women in 17 randomised trials. Lancet 378: 1707-1716.
150. Fisher B, Anderson S, Bryant J, Margolese RG, Deutsch M, et al. (2002) Twenty-year follow-up of a randomized trial comparing
total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347:
1233-1241.
151. Veronesi U, Cascinelli N, Mariani L, Greco M, Saccozzi R, et al. (2002) Twenty-year follow-up of a randomized study comparing
breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347: 1227-1232.
152. Litière S, Werutsky G, Fentiman IS, Rutgers E, Christiaens MR, et al. (2012) Breast conserving therapy versus mastectomy for
stage I-II breast cancer: 20 year follow-up of the EORTC 10801 phase 3 randomised trial. Lancet Oncol 13: 412-419.
153. Whelan TJ, Pignol JP, Levine MN, Julian JA, MacKenzie R, et al. (2010) Long-term results of hypofractionated radiation therapy
for breast cancer. N Engl J Med 362: 513-520.
154. Beitsch PD, Shaitelman SF, Vicini FA (2011) Accelerated partial breast irradiation. J Surg Oncol 103: 362-368.
155. Shaitelman SF, Vicini FA, Beitsch P, Haffty B, Keisch M.et al. (2010) Five-year outcome of patients classified using the American
Society for Radiation Oncology consensus statement guidelines for the application of accelerated partial breast irradiation: an
analysis of patients treated on the American Society of Breast Surgeons MammoSite Registry Trial. Cancer. 116: 4677-4685.
156. Vicini F, Beitsch P, Quiet C, Gittleman M, Zannis V, et al. Five-year analysis of treatment efficacy and cosmesis by the American
Society of Breast Surgeons Mammo Site Breast Brachytherapy Registry Trial in patients treated with accelerated partial breast
irradiation. Int J Radiat Oncol Biol Phys 79: 808-817.
157. Smith GL, Xu Y, Buchholz TA, Giordano SH, Jiang J, et al. (2012) Association between treatment with brachytherapy vs whole-
breast irradiation and subsequent mastectomy, complications, and survival among older women with invasive breast cancer.
JAMA 307: 1827-1837.
158. Cox JA, Swanson TA (2013) Current modalities of accelerated partial breast irradiation. Nat Rev Clin Oncol 10: 344-356.
159. Mauri D, Pavlidis N, Ioannidis JP (2005) Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis. J
Natl Cancer Inst 97: 188-194.
160. von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, et al. (2012) Definition and impact of pathologic complete
response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol 30: 1796-1804.
161. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG), Davies C, Godwin J, Gray R, Clarke M, et al. (2011) Relevance of
breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised
trials. Lancet 378: 771-784.
162. Davies C, Pan H, Godwin J, Gray R, Arriagada R, et al. (2013) Long-term effects of continuing adjuvant tamoxifen to 10 years
versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomized trial. Lancet 381:
805-816.
163. Theriault RL, Carlson RW, Allred C, Anderson BO, Burstein HJ, et al. (2013) Breast cancer, version 3.2013: featured updates to
the NCCN guidelines. J Natl Compr Canc Netw 11: 753-760.
164. LHRH-agonists in Early Breast Cancer Overview group, Cuzick J, Ambroisine L, Davidson N, Jakesz R, et al. (2007) Use of
luteinising-hormone-releasing hormone agonists as adjuvant treatment in premenopausal patients with hormone-receptor-positive
breast cancer: a meta-analysis of individual patient data from randomised adjuvant trials. Lancet 369: 1711-1723.
165. Dowsett M, Cuzick J, Ingle J, Coates A, Forbes J, et al. (2010) Meta-analysis of breast cancer outcomes in adjuvant trials of
aromatase inhibitors versus tamoxifen. J Clin Oncol 28: 509-518.
166. Burstein HJ, Prestrud AA, Seidenfeld J, Anderson H, Buchholz TA, et al. (2010) American Society of Clinical Oncology clinical
practice guideline: update on adjuvant endocrine therapy for women with hormone receptor-positive breast cancer. J Clin Oncol
28: 3784-3796.
167. Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE Jr, et al. (2005) Trastuzumab plus adjuvant chemotherapy for operable
HER2-positive breast cancer. N Engl J Med 353: 1673-1684.
168. Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, et al. (2007) American Society of Clinical Oncology/College of
American Pathologists Guideline Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer.
Arch Pathol Lab Med 131: 18-43.
169. Baselga J, Cortés J, Kim SB, Im SA, Hegg R, et al. (2012) Pertuzumab plus trastuzumab plus docetaxel for metastatic breast
cancer. N Engl J Med 366: 109-119.
170. Verma S, Miles D, Gianni L, Krop IE, Welslau M, et al. (2012) Trastuzumab emtansine for HER2-positive advanced breast cancer.
N Engl J Med 367: 1783-1791.
171. Cameron D, Casey M, Oliva C, Newstat B, Imwalle B, et al. (2010) Lapatinib plus capecitabine in women with HER-2-positive
advanced breast cancer: final survival analysis of a phase III randomized trial. Oncologist 15: 924-934.
172. Bachelot T, Bourgier C, Cropet C, Ray-Coquard I, Ferrero JM, et al. (2012) Randomized phase II trial of everolimus in combination
with tamoxifen in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast
cancer with prior exposure to aromatase inhibitors: a GINECO study. J Clin Oncol 30: 2718-2724.
173. Baselga J, Semiglazov V, van Dam P, Manikhas A, Bellet M, et al. (2009) Phase II randomized study of neoadjuvant everolimus
plus letrozole compared with placebo plus letrozole in patients with estrogen receptor-positive breast cancer. J Clin Oncol 27:
2630-2637.
174. Harisinghani MG, Weissleder R (2004) Sensitive, noninvasive detection of lymph node metastases. PLoS Med 1: 66.
175. Gordon AN, Fleagle JT, Guthrie D, Parkin DE, Gore ME, et al. (2001) Recurrent epithelial ovarian carcinoma: a randomized phase
III study of pegylated liposomal doxorubicin versus topotecan. J Clin Oncol 19: 3312-3322.
177. Cho K, Wang X, Nie S, Chen ZG, Shin DM (2008) Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res 14:
1310-1316.
178. Sinha R, Kim GJ, Nie S, Shin DM (2006) Nanotechnology in cancer therapeutics: bioconjugated nanoparticles for drug delivery.
Mol Cancer Ther 5: 1909-1917.
179. P Geels, E Eisenhauer, A Bezjak, B Zee, A Day (2000) Palliative effect of chemotherapy: objective tumor response is associated
with symptom improvement in patients with metastatic breast cancer. Journal of Clinical Oncology 18: 2395-2405.
180. Jones A, Harris AL (1998) New developments in angiogenesis: a major mechanism for tumor growth and target for therapy.
Cancer J Sci Am 4: 209-217.
181. Moghimi SM, Hunter AC (2000) Poloxamers and poloxamines in nanoparticle engineering and experimental medicine. Trends
Biotechnol 18: 412-420.
182. Park EK, Lee SB, Lee YM (2005) Preparation and characterization of methoxy poly(ethylene glycol)/poly(epsilon-caprolactone)
amphiphilic block copolymeric nanospheres for tumor-specific folate-mediated targeting of anticancer drugs. Biomaterials 26:
1053-1061.
183. Maeda H, Matsumura Y (1989) Tumoritropic and lymphotropic principles of macromolecular drugs. Crit Rev Ther Drug Carrier
Syst 6: 193-210.
184. Mansour AM, Drevs J, Esser N, Hamada FM, Badary OA, et al. (2003) A new approach for the treatment of malignant melanoma:
enhanced antitumor efficacy of an albumin-binding doxorubicin prodrug that is cleaved by matrix metalloproteinase 2. Cancer
Research 63: 4062-4066.
185. Nomura T, Saikawa A, Morita S, Sakaeda Kakutani T, Yamashita F, et al. (1998) Pharmacokinetic characteristics and therapeutic
effects of mitomycin C-dextran conjugates after intratumoural injection. Journal of Controlled Release 52: 239-252.
186. S Prabha, V Labhasetwar (2004) Nanoparticle-mediated wild-type p53 gene delivery results in sustained antiproliferative activity
in breast cancer cells. Molecular Pharmacology 1: 211-219.
187. Kirpotin DB, Drummond DC, Shao Y, Shalaby MR, Hong K, et al. (2006) Antibody targeting of long-circulating lipidic nanoparticles
does not increase tumor localization but does increase internalization in animal models. Cancer Research 66: 6732-6740.
188. Ford JM, Kastan MB (2008) DNA damage response pathways and cancer. In: Abeloff MD, Armitage JO, Niederhuber JE, Kastan
MB, McKenna WG (eds.). Abeloff’s Clinical Oncology. (4thedn), Churchill Livingston, Philadelphia, PA, USA.
189. Durkacz BW, Omidiji O, Gray DA, Shall S (1980) (ADP-ribose)n participates in DNA excision repair. Nature 283: 593-596.
190. Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG (2010) PARP inhibition: PARP1 and beyond. Nat Rev Cancer 10:
293-301.
191. Arnold Simons, Naomi Dafni, Iris Dotan, Yoram Oron, Dan Canaani, et al. (2001) Establishment of a chemical synthetic lethality
screen in cultured human cells. Genome Res 11: 266-273.
192. Veuger SJ, Curtin NJ, Smith GC, Durkacz BW (2004) Effects of novel inhibitors of poly(ADP-ribose) polymerase-1 and the DNA-
dependent protein kinase on enzyme activities and DNA repair. Oncogene 23: 7322-7329.
193. Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, et al. (2005) Targeting the DNA repair defect in BRCA mutant cells as a
therapeutic strategy. Nature 434: 917-921.
194. Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, et al. (2005) Specific killing of BRCA2-deficient tumours with inhibitors
of poly(ADP-ribose) polymerase. Nature 434: 913-917.
195. Kaelin WG Jr (2005) The concept of synthetic lethality in the context of anticancer therapy. Nat Rev Cancer 5: 689-698.
196. Hartman AR, Ford JM (2002) BRCA1 induces DNA damage recognition factors and enhances nucleotide excision repair. Nat
Genet 32: 180-184.
197. Alli E, Sharma VB, Sunderesakumar P, Ford JM (2009) Defective repair of oxidative dna damage in triple-negative breast cancer
confers sensitivity to inhibition of poly(ADP-ribose) polymerase. Cancer Res 69: 3589-3596.
198. Arnaudeau C, Lundin C, Helleday T (2001) DNA double-strand breaks associated with replication forks are predominantly repaired
by homologous recombination involving an exchange mechanism in mammalian cells. J Mol Biol 307: 1235-1245.
199. Fong PC, Boss DS, Yap TA, Tutt A, Wu P, et al. (2009) Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation
carriers. N Engl J Med 361: 123-134.
200. Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, et al. (2010) Oral poly(ADP-ribose) polymerase inhibitor olaparib in
patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet 376: 235-244.
201. Gelmon KA, Hirte HW, Robidoux A, Tonkin KS, Tischkowitz M, et al. (2010) Can we define tumors that will respond to PARP
inhibitors- A phase II correlative study of olaparib in advanced serous ovarian cancer and triple-negative breast cancer. J Clin
Oncol (ASCO Annual Meeting Abstracts) 28: Abstract 3002.
202. Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, et al. (2003) Repeated observation of breast tumor subtypes in independent
gene expression data sets. Proc Natl Acad Sci USA 100: 8418-8423.
203. Cleator S, Heller W, Coombes RC (2007) Triple-negative breast cancer: therapeutic options. Lancet Oncol 8: 235-244.
204. Schneider BP, Winer EP, Foulkes WD, Garber J, Perou CM, et al. (2008) Triple-negative breast cancer: risk factors to potential
targets. Clin Cancer Res 14: 8010-8018.
205. Hastak K, Alli E, Ford JM (2010) Synergistic chemosensitivity of triple-negative breast cancer cell lines to poly(ADP-Ribose)
polymerase inhibition, gemcitabine, and cisplatin. Cancer Res 70: 7970-7980.
206. J O’Shaughnessy, M Yoffe, C Osborne, J Blum, C Rocha, et al. (2009) Triple negative breast cancer: a phase 2, multi-center,
open-label, randomized trial of gemcitabine/carboplatin (G/C), with or without BSI-201, a PARP inhibitor. Cancer Research 69:
2120.
207. J O’Shaughnessy, C Osborne, J Pippen, M Yoffe, D Patt, et al. (2009) Efficacy of BSI-201, a poly (ADP-ribose) polymerase-1
(PARP1) inhibitor, in combination with gemcitabine/ carboplatin (G/C) in patients with metastatic triple-negative breast cancer
(TNBC): results of a randomized phase II trial. J Clin Oncol (ASCO Annual Meeting Abstracts 27: Abstract 3.
208. J O’Shaughnessy, C Osborne, D Patt, C Rocha, V Ossovskaya, et al. (2009) Final results of a randomized phase II study
demonstrating efficacy and safety of BSI-201, a poly (ADP-Ribose) polymerase (PARP) inhibitor, in combination with gemcitabine/
carboplatin (G/C) in metastatic triple negative breast cancer (TNBC). Cancer Res 69: Abstract 3122.
209. Food, Nutrition, Physical Activity and the Prevention of Cancer: a Global Perspective; American Institute for Cancer Research/
World Cancer Research Fund.
210. Eliassen AH, Hankinson SE, Rosner B, Holmes MD, Willett WC (2010) Physical activity and risk of breast cancer among
postmenopausal women. Arch Intern Med 170: 1758-1764.
211. Zheng JS, Hu XJ, Zhao YM, Yang J, Li D (2013) Intake of fish and marine n-3 polyunsaturated fatty acids and risk of breast cancer:
meta-analysis of data from 21 independent prospective cohort studies. BMJ 346: 3706.
212. Hartmann LC, Schaid DJ, Woods JE, Crotty TP, Myers JL, et al. (1999) Efficacy of bilateral prophylactic mastectomy in women
with a family history of breast cancer. N Engl J Med 340: 77-84.
213. Meijers-Heijboer H, van Geel B, van Putten WL, Henzen-Logmans SC, Seynaeve C, et al. (2001) Breast cancer after prophylactic
bilateral mastectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 345: 159-164.
214. Moyer VA, US Preventive Services Task Force (2014) Risk Assessment, Genetic Counseling, and Genetic Testing for BRCA-
Related Cancer in Women: U.S. Preventive Services Task Force Recommendation Statement. Ann Intern Med 160.
215. Nelson HD, Smith ME, Griffin JC, Fu R (2013) Use of medications to reduce risk for primary breast cancer: a systematic review
for the U.S. Preventive Services Task Force. Ann Intern Med 158: 604-614.
216. Cuzick J, Sestak I, Bonanni B, Costantino JP, Cummings S, et al. (2013) Selective oestrogen receptor modulators in prevention
of breast cancer: an updated meta-analysis of individual participant data. Lancet 381: 1827-1834.
217. Moyer VA (2013) Medications for Risk Reduction of Primary Breast Cancer in Women: U.S. Preventive Services Task Force
Recommendation Statement. Annals of Internal Medicine 159: 698-708.
218. Jack Cuzick, Ivana Sestak, Bernardo Bonanni, Joseph P Costantino, Steve Cummings, et al. (2013) Selective oestrogen receptor
modulators in prevention of breast cancer: an updated meta-analysis of individual participant data. The Lancet 381: 1827-1834.