You are on page 1of 7

810 Breast Cancer Mayo Clin Proc, June 2004, Vol 79

Concise Review for Clinicians

Advances in Screening, Diagnosis, and Treatment of Breast Cancer

BETTY A. MINCEY, MD, AND EDITH A. PEREZ, MD

Breast cancer is the most common cancer in women in the have been evolving. Advances in screening, diagnosis, and
United States; this year, approximately 215,900 new cases treatment of breast cancer continue to influence our ap-
will be diagnosed. Mammography remains the corner- proach to patients with this disease. Many improvements
stone of screening, with technologies such as ultrasonog- have been made as well in supportive care, including in-
raphy and magnetic resonance imaging having an in- creased tolerability of therapy and notable amelioration of
creasingly defined role. Improved risk assessment and disease symptoms.
prevention strategies have been implemented, and current Mayo Clin Proc. 2004;79:810-816
research in these areas includes better identification of
patients at risk, the use of aromatase inhibitors and other AI = aromatase inhibitor; CI = confidence interval; DCIS =
agents to reduce risk, and the use of surrogate markers. ductal carcinoma in situ; ER = estrogen receptor; LCIS =
Breast cancer staging has been optimized recently; also, lobular carcinoma in situ; NSABP = National Surgical Adju-
vant Breast and Bowel Project; PR = progesterone receptor;
local management of breast cancer, adjuvant systemic STAR = Study of Tamoxifen and Raloxifene
therapies, and treatment of patients with advanced disease

B reast cancer is the most common cancer in women in


the United States, with incidence increasing about
0.5% per year. In the year 2004, an estimated 215,900 new
The sensitivity of first mammography ranged from 71% to
96% in studies of a 1-year screening interval.2,3 The posi-
tive predictive value of abnormal mammographic results
cases of invasive breast cancer (stages I-IV) will be diag- requiring biopsy ranged from 12% to 78% in these studies
nosed, and about 40,100 women and 470 men will die of the and increased with age.2,3
disease.1 New strategies to decrease the risk of breast cancer Young women at increased risk of breast cancer devel-
are being studied and implemented. Although breast cancer opment should be advised to undergo mammography be-
is a leading cause of death in women (second only to lung fore age 40 years. Current guidelines suggest that women
cancer), mortality rates have declined recently. This reduc- with BRCA1 or BRCA2 mutations should begin screening
tion in mortality, despite the increase in incidence, is attrib- mammography at age 25 years or 10 years earlier than the
uted to improvements in screening, diagnosis, and treatment. youngest age at which breast cancer has been diagnosed in
their family.4 Although it is intuitive to recommend this
SCREENING approach to women with a strong family history of breast
Despite controversy surrounding recent reports, mammog- cancer and unknown genetic makeup, data are not currently
raphy remains the cornerstone of breast cancer screening. available to quantify benefit for this group. Unfortunately,
Mammographic screening for early detection of breast can- mammography in these groups of young women has a sensi-
cer reduces mortality and is widely recommended as an tivity of only about 33% in most studies. Ultrasonography
integral part of annual preventive health care for all women does not improve this sensitivity and is not recommended for
beginning at age 50 years and continuing for as long as a screening; magnetic resonance imaging of the breast has a
woman’s life expectancy is at least 10 years. For women sensitivity of between 90% and 100% but is expensive and
older than 40 years, most major health organizations rec- has not been endorsed for screening purposes.2,3 Ongoing
ommend annual or semiannual screening mammography.2,3 trials are further exploring these technologies.
The clinical breast examination is an important part of
From the Division of General Internal Medicine (B.A.M.), Multidis- breast cancer screening and surveillance because up to 10%
ciplinary Breast Clinic (B.A.M., E.A.P.), and Division of Hematology of breast cancers may be clinically evident while mam-
and Oncology (E.A.P.), Mayo Clinic College of Medicine, Jackson- mographically occult.2,3 The role of breast self-examination
ville, Fla.
has not been well-defined.
A question-and-answer section appears at the end of this article.
Individual reprints of this article are not available. Address corre- RISK ASSESSMENT
spondence to Edith A. Perez, MD, Division of Hematology and On-
cology, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jack- For some women, the risk of developing breast cancer is
sonville, FL 32224 (e-mail: perez.edith@mayo.edu). high enough to warrant consideration of prevention strate-
Mayo Clin Proc. 2004;79:810-816 810 © 2004 Mayo Foundation for Medical Education and Research

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
Mayo Clin Proc, June 2004, Vol 79 Breast Cancer 811

gies in addition to screening and surveillance. Current Tamoxifen, a selective estrogen receptor (ER) modula-
methods for defining risk include use of the modified tor that has been used for years to treat breast cancer, is
Gail model, use of the Claus model, and determination of the only agent currently approved by the US Food and
BRCA mutation carrier status.4 The modified Gail model Drug Administration to decrease the risk of breast cancer.
combines relative risks associated with age, race, age at Its approval by this agency for prevention of breast cancer
menarche, number of previous breast biopsies, history of was based on the results of the National Surgical Adjuvant
atypical ductal hyperplasia, and family history of breast Breast and Bowel Project (NSABP) P-1 study, in which a
cancer in first-degree relatives for estimation of 5-year and relative risk reduction of approximately 50% was observed
lifetime risks of breast cancer development in an individual for women with a 5-year risk of 1.66% or higher, as calcu-
woman. lated by the modified Gail model (despite notable adverse
The online risk-assessment tool, located at www effects among 13,388 women, including 3 women in the
.breastcancerprevention.com, provides comprehensive in- tamoxifen group who died of pulmonary emboli vs none in
formation approved by the National Cancer Institute and the placebo arm). A meta-analysis of 5 worldwide trials
includes a system of scoring an individual’s risk of devel- that evaluated tamoxifen vs placebo in women at increased
oping breast cancer in the next 5 years and by age 80 years. risk of breast cancer revealed a 38% (95% confidence
A score of 1.66% or higher for the next 5 years indicates interval [CI], 28%-46%; P<.001) relative reduction in risk
high risk. This model is reasonable for estimating risk in of breast cancer development at 5 years of follow-up.6 In
most women but may seriously underestimate risk in terms of absolute numbers, patients in the placebo group
women with strong family histories of breast cancer. For had an approximate 13% risk of developing a ductal carci-
such women, the Claus model, which considers only family noma in situ (DCIS) or invasive breast cancer during the
history, can be used to estimate risk. In appropriate cases, study periods vs 7% of those in the tamoxifen arm. There
genetic counseling and testing for mutations in BRCA1 or was no effect on ER-negative breast cancer but a 48%
BRCA2 genes may be considered. relative decrease in ER-positive disease. However, the risk
None of the risk models consider a history of lobular of endometrial cancer was increased (odds ratio, 2.4; 95%
carcinoma in situ (LCIS), which is primarily regarded as a CI, 1.5-4.0), as was the risk of venous thromboembolism
risk factor rather than a precursor lesion and is associated (odds ratio, 1.9; 95% CI, 1.4-2.7). Survival differences
with an annual risk of approximately 1% for breast cancer between tamoxifen and placebo have not been shown to
development in either breast. date. Ongoing trials are evaluating other prevention strate-
An additional risk factor is mammographic density. gies. The Study of Tamoxifen and Raloxifene (STAR),
Breasts with larger amounts of connective and epithelial NSABP P-2 trial is comparing 5 years of tamoxifen use vs
tissue appear denser than do breasts with more fat. Mam- raloxifene use, another selective ER modulator, with eligi-
mographically dense breast tissue is one of the strongest bility determined as for the NSABP P-1 study by an esti-
established risk factors for breast cancer, conferring a 4- mated baseline risk of breast cancer higher than 1.66% at 5
to 6-fold increased risk for women with the most dense years, based on the modified Gail model. The accrual goal
breast tissue compared with those with the least dense.5 for STAR is 19,000 women, which should be achieved this
Mammographic density also decreases the sensitivity of year. A large prevention study (MAP.3) about to begin at
mammography. The relative importance of mammographic multiple medical centers in the United States and Canada
density as a risk factor and the underlying mechanisms will compare the effectiveness of the aromatase inhibitor
responsible for the increased risk in women with dense (AI) exemestane, either alone or in combination with
breast tissue are topics of ongoing research. celecoxib, with placebo for reducing the risk of breast
cancer in high-risk women.
PREVENTION For women with a substantially elevated risk of breast
Women with a substantially increased risk of breast cancer cancer development, particularly those proved to carry mu-
should be counseled regarding options for breast cancer tations in BRCA1 or BRCA2, surgical options for preven-
prevention. Defining the level of risk that warrants inter- tion should be discussed.4 Available data regarding risk
vention may be difficult and should be done on an indi- reduction with surgical prophylaxis indicate that bilateral
vidual basis, taking into account a woman’s age, comor- mastectomy reduces the risk of breast cancer development
bidities, and her own thoughts about her risk and how it by approximately 90% and that oophorectomy reduces the
affects her quality of life. Decisions regarding interven- risk of breast cancer by approximately 50% if performed
tions such as chemoprevention or surgical prophylaxis before menopause.4 These risk reductions are based on
must be highly individualized and will vary greatly among retrospective, observational studies and on more recent
women with similar risk levels. prospective observational studies of women at high risk.

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
812 Breast Cancer Mayo Clin Proc, June 2004, Vol 79

Three important areas of research in the prevention Sentinel lymph node evaluation has essentially become
setting include better identification and definition of pa- a standard of care as part of the staging of invasive breast
tients at increased risk of breast cancer development, the cancer. Outside of clinical trials, if the sentinel lymph node
use of AIs, and the use of agents that may decrease the risk is negative for tumor by hematoxylin and eosin pathologi-
of hormone receptor–negative disease. Surrogate markers cal evaluation, then axillary node dissection is not per-
for breast cancer risk are being sought so that pilot trials of formed. However, a full axillary node dissection is per-
new agents can be completed with fewer participants in less formed if the sentinel lymph node is positive for tumor.
time than is needed to show actual changes in cancer inci- Ongoing clinical trials are evaluating the feasibility of ob-
dence. An example of a potential surrogate marker is mam- viating full dissection in patients whose sentinel lymph
mographic density. Because it is not only measurable but node is positive for malignancy. Routine evaluation of the
also may be altered potentially by interventions such as sentinel lymph node in the setting of DCIS has not been
selective ER modulators or AIs, mammographic density established, although it is included sometimes in patients
may prove to be a useful surrogate marker for both risk and with multifocal or high-grade DCIS.
risk reduction. A small, ongoing 1-year study (MAP.2) will External beam radiotherapy is a standard component of
determine the effectiveness of exemestane in reducing local treatment of patients with invasive breast cancer
mammographic density. when breast conservation is undertaken.8 For women who
have undergone mastectomy, this therapy is not routinely
BREAST CANCER STAGING recommended unless the tumor is large (>5.0 cm), there is
Most patients presenting with breast cancer have disease chest wall involvement, or more than 3 axillary lymph
localized to the breast or to the breast and axillary lymph nodes are involved. The use of postmastectomy radiation
nodes; however, 40% to 50% eventually may develop therapy for patients with 1 to 3 lymph nodes positive for
metastatic disease. A new staging system became standard malignancy is controversial.
in 2003: the 6th American College of Physicians Staging,7 Generally, radiation therapy after breast conserving sur-
which assigns a stage from 0 to IV on the basis of the gery consists of whole breast external beam radiotherapy
anatomical extent of the tumor, including invasiveness, size, administered over 5 to 6 weeks, but new techniques are
and lymph node or distal involvement. Stage 0 (Tis) breast evaluating partial breast irradiation. Partial breast irradia-
cancers are DCIS (or intraductal) carcinomas. Lobular carci- tion is administered generally to the portion of the breast
noma in situ increasingly is considered a risk factor for that includes the tumor bed and an area of surrounding
subsequent breast cancer, but it is still included as a malig- margin. One advantage of this technique is that it might
nancy in the most recent staging system and is classified as allow the use of higher doses of radiation, shortening treat-
“Tis (LCIS).”7 Stages I through IV are invasive tumors, most ment duration from 5 to 6 weeks to only a few days.2,3
commonly of the infiltrating ductal type. The histological The necessity of radiation therapy is uncertain in
subtype of breast cancer is not part of the staging system but women with small, low- to intermediate-grade DCIS un-
is important for its potential influence on patient outcome. dergoing lumpectomy of a tumor with negative margins.
Several ongoing clinical trials are addressing this issue. For
LOCAL MANAGEMENT OF PRIMARY now, radiation therapy is a standard component of breast
BREAST CANCER conservation in most women with DCIS.
The management of primary breast cancer, whether inva-
sive or in situ, begins with local treatment. The only exclu- ADJUVANT SYSTEMIC THERAPY
sion is LCIS, which is managed as a determinant of in- The goals of adjuvant systemic therapy include preventing
creased risk; in other words, management after a biopsy the recurrence of primary breast cancer and decreasing the
specimen shows LCIS does not require full excision, lump- risk of death from the disease while ameliorating the risks
ectomy, or mastectomy. As a determinant of increased risk of toxicity.9,10 Improvements in this area will depend on
of subsequent breast cancer, LCIS is managed by observa- scientifically rigorous and well-designed clinical trials.
tion (follow-up) and chemoprevention. Most women with Major areas of research include predictors of outcome and
newly diagnosed breast cancer have a choice between response using gene profiling, optimization of existing
breast conservation therapy with lumpectomy and radiation agents, and incorporation of biologically based treatments.
or mastectomy. Although the chance of recurrence is Systemic therapy may consist of antiestrogen hormonal
higher with breast conservation therapy, the effect of both therapy, adjuvant chemotherapy, or both.
therapies on overall survival is the same. Women with Premenopausal or postmenopausal women with DCIS
large tumors or those with multifocal breast cancer often that expresses ER who are treated with lumpectomy with or
are not candidates for breast conservation. without radiation have an approximate 50% relative benefit

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
Mayo Clin Proc, June 2004, Vol 79 Breast Cancer 813

Table 1. Aromatase Inhibitors (AIs) as Adjuvant Therapy for Postmenopausal Women


With Hormonally Responsive Breast Cancer
Randomization Reduction in
Prior Median Relative reduction
recurrence
No. of adjuvant Duration follow-up in contralateral
Study* patients hormones of intervention Treatment (mo) Relative† Absolute‡ breast cancer (%)
ATAC12 9366 None 5y Tamoxifen 47 14%‡ 2.4% at 4 y 38
Anastrozole (P=.03)
Tamoxifen +
anastrozole†
MA.1713 5187 5 y of 5y Placebo 30 43% 6% at 4 y 46
tamoxifen Letrozole (P<.001)
IES14 4742 2 or 3 y of 3 or 2 y§ Tamoxifen 30.6 32% 4.7% at 3 y 56
tamoxifen Exemestane (P<.001)
*ATAC = Arimidex, Tamoxifen Alone or in Combination; IES = International Exemestane Study; MA.17 = Letrozole vs Placebo After 5 Years of
Tamoxifen.
†In favor of AI.
‡Results of the combined arm similar to those of tamoxifen alone.
§3 years after 2 years of tamoxifen, or 2 years after 3 years of tamoxifen.

in decreasing disease recurrence (but only a small absolute been approved for the adjuvant setting, but approval of letro-
benefit, about 1%-2%) from the 5-year use of adjuvant zole and perhaps exemestane for this indication is expected
tamoxifen; AIs currently are being evaluated in clinical this year. Adjuvant AIs for postmenopausal women with
trials for postmenopausal women. However, no evidence at hormonally responsive tumors are summarized in Table 1.12-14
this time suggests that these hormonal agents affect sur- For premenopausal women, one of the central issues
vival of patients with DCIS. Chemotherapy is not recom- related to hormonal therapy is the role of ovarian ablation
mended as adjuvant therapy in the setting of DCIS. as a substitute for, or in combination with, systemic chemo-
The selection of hormonal adjuvant therapy for women therapy. Several ongoing trials are addressing this issue,
with invasive breast cancer depends on the menopausal sta- including the Suppression of Ovarian Function Trial
tus of the patient because the status determines the source of (SOFT), IBCSG 24-02; the Tamoxifen and Exemestane
estrogen production (the ovaries in premenopausal women, Trial (TEXT), IBCSG 25-02; and the Premenopausal En-
peripheral tissues in postmenopausal women). Patients with docrine Responsive Chemotherapy (PERCHE) trial,
resected invasive breast cancer whose tumors express ER IBCSG 26-02; information is available at the Cancer Trials
and/or progesterone receptor (PR) typically receive 5 years Support Unit (CTSU) Web site (www.ctsu.org).
of antiestrogen treatment in the form of tamoxifen; an even Combination chemotherapy is recommended for many
better approach appears to be the use of an AI (such as premenopausal or postmenopausal women with invasive
anastrozole) if the patient is postmenopausal.11,12 Clinical breast cancer who are eligible to receive adjuvant therapy.
trials have shown that continuing to use tamoxifen after 5 Adjuvant chemotherapy should be discussed with patients
years is not helpful (and may be detrimental in terms of diagnosed as having invasive tumors larger than 1.0 cm.9,10
increased tumor relapse). A recent study showed that the AI Many types of chemotherapy approaches are available, and
letrozole improves disease-free survival if used for 5 years in recent data have provided a better understanding of opti-
postmenopausal women who have completed 5 years of mal regimens. Anthracycline (doxorubicin or epirubicin)-
tamoxifen use and who are free of disease.13 Another recent containing regimens are better than cyclophosphamide,
study showed that for postmenopausal women with hormon- methotrexate, 5-fluorouracil type of chemotherapy; thus,
ally responsive tumors, it is better to switch to the AI this latter regimen is used less frequently than it was 5 to
exemestane after 2 to 3 years of tamoxifen use because 10 years ago. Taxanes (paclitaxel or docetaxel) improve
exemestane significantly improves disease-free survival disease-free and overall survival for patients with node-
compared with 5 years of tamoxifen use.14 New clinical trials positive breast cancer (data are pending regarding their
are comparing the efficacies of the following: 5 years of therapeutic ratio in patients with node-negative breast can-
tamoxifen use followed by 5 years of AI use, 5 or even 10 cer). Taxanes may be used either sequentially or concur-
years of AI use, or 2 to 3 years of tamoxifen use followed by rently with the anthracyclines, depending on the agent
AI use for the rest of the 5-year period of hormonal therapy. used. The typical duration of adjuvant systemic chemo-
Three AIs are available for use in the United States: therapy ranges from 2 to 6 months; the duration of older
anastrozole, letrozole, and exemestane; only anastrozole has regimens was 6 months to 2 years. Variations in drug ad-

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
814 Breast Cancer Mayo Clin Proc, June 2004, Vol 79

ministration appear to alter the efficacy of chemotherapy, ultimately develop metastatic disease (although this num-
and newer trials are exploring this strategy. ber is expected to decrease with the institution of better
Treatment of patients with node-negative disease is adjuvant therapies). The goals of systemic chemotherapy in
evolving, and many ongoing trials are attempting to iden- this setting are to ameliorate symptoms of breast cancer,
tify predictors of response and to select patients most likely optimize quality of life, delay disease progression, and
to benefit from different therapies. In general, the same prolong survival.
principles as those outlined for node-positive disease also The decision regarding therapy choice(s) for patients
apply to patients with node-negative breast cancer; how- with metastatic breast cancer depends on several factors,
ever, although the relative improvements with chemo- including prior therapies that were administered in the
therapy are the same irrespective of whether the nodes are adjuvant setting, biological characteristics of the tumor,
involved, the absolute improvements are expected to be sites of involvement, and the patient’s overall clinical con-
lower for patients with node-negative disease because of dition (comorbid conditions, performance status). In gen-
their lower risk of breast cancer recurrence. eral, hormonal approaches are considered appropriate for
If both chemotherapy and hormonal therapy are recom- patients with hormonally responsive tumors (essentially
mended, the chemotherapy is given first to minimize toxic- defined as ER-positive and/or PR-positive) that involve
ity and improve efficacy. bone or soft tissues or even asymptomatic visceral disease.
Otherwise, chemotherapy is the first choice. Two critically
LONG-TERM FOLLOW-UP important biological factors that must be remembered in
The role of routine diagnostic tests in patients who have making treatment decisions are ER and HER2 status, and
undergone primary treatment of resected breast cancer has not all patients diagnosed as having metastatic breast cancer
been established. However, mammography performed every should have their tumor tested for both of these markers
6 months is generally recommended after lumpectomy and (preferably based on a new biopsy, but data from the origi-
radiation until stabilization of the resection scar, and yearly nal tumor are sometimes used) to determine whether the
thereafter. Yearly mammography is generally recommended patient may benefit from targeted therapies against these
for the contralateral breast because of the risk of new primary proteins.
breast cancer in the other breast of approximately 0.5% per
year. Blood tests (including so-called serum tumor markers, Hormonal Therapy
complete blood cell count, chemistry panel) and radiological Although tamoxifen is recommended for premeno-
studies (such as chest radiography, magnetic resonance imag- pausal women with advanced disease, the luteinizing hor-
ing, computed tomography, bone scanning, or positron emis- mone–releasing hormone agonists offer another viable op-
sion tomography) are not recommended routinely. History tion. For postmenopausal women, the AIs have provided a
and physical examination by medical personnel is recom- therapeutic ratio superior to that of tamoxifen and have
mended about every 6 months for the first 5 years after become the first agents of choice. Clinical trials are ongo-
diagnosis for most patients with resected invasive breast can- ing to determine whether there are meaningful differences
cer, and specific diagnostic studies are guided by the clinical in efficacy and tolerability among the 3 available AIs. An
information elicited during those visits or if intervening prob- additional option for treating patients with hormonally re-
lems are identified by the patient. sponsive tumors is fulvestrant, a “pure” antiestrogen that
In patients who will experience a recurrence of breast appears to be slightly better or similar in efficacy to the AI
cancer, approximately 17% of recurrences will be diagnosed anastrozole. Currently, use of fulvestrant is either as a
within 5 years of the initial diagnosis. Patients with breast substitute for AIs or, perhaps more commonly, after dis-
cancer continue to be at risk for metastatic disease for 20 or ease progression while the patient is taking antiaromatase
more years after diagnosis; therefore, clinical vigilance and agents. These hormonal agents may be used in sequence
prompt diagnostic evaluation of suspicious symptoms and but not in combination. If tumors become refractory to
signs are recommended. The most common sites of meta- hormonal therapy, if significant visceral disease is present,
static disease are bone, liver, lungs, skin, and brain, but other or if a tumor has no hormonal receptors, then chemo-
sites such as the peritoneum and retina may be affected by therapy is the treatment of choice.
breast cancer.
Treatment of Patients With HER2-Negative Disease
MANAGEMENT OF LOCALLY The selection of systemic chemotherapy for patients
ADVANCED/METASTATIC DISEASE with HER2-negative breast cancer continues to be a chal-
As alluded to previously, about 40% to 50% of patients lenge because many options are available. Although the
diagnosed as having stage I through III breast cancer may existing approaches do not lead to ultimate cure of the

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
Mayo Clin Proc, June 2004, Vol 79 Breast Cancer 815

disease, management goals include amelioration of symp- used concurrently with anthracyclines because of an in-
toms from the malignancy and improvements in the creased risk of cardiac toxicity. Trastuzumab therapy is
quality and duration of life. Some patients receive single- associated with a small risk of congestive heart failure,
agent treatment, whereas others may benefit from combi- which is managed with otherwise standard therapies for
nation therapies. In general, combination therapies lead to congestive heart failure (angiotensin-converting enzyme
better response rates and time to progression, but not inhibitors, diuretics, β-blockers, digoxin). This cardiac tox-
always to better survival, than do sequential single-agent icity appears to be different from that related to anthra-
approaches. cyclines because it is not dose dependent, appears not to be
No single chemotherapy regimen is best for all pa- associated with identifiable structural cardiac abnormali-
tients; however, many options are available because of ties, and tends to improve promptly with therapy in most
clinical investigations that revealed the antitumor activ- patients.14 Ongoing clinical trials are evaluating the poten-
ity of anthracyclines (doxorubicin, epirubicin), antime- tial role of trastuzumab as part of adjuvant therapy for
tabolites (capecitabine, gemcitabine), and antitubulin HER2-positive resected invasive breast cancer.
agents such as the taxanes (paclitaxel, docetaxel) and
vinorelbine. Overall, individualization of care is critically SUPPORTIVE CARE ISSUES
important. All patients with metastatic breast cancer Many improvements have been made in supportive care,
should seek information regarding available clinical trials, which translate into better tolerability of therapy and
which would be discussed in more detail by the medical marked amelioration of disease symptoms. These advances
oncologist. include newer antiemetics, use of prophylactic growth fac-
More than 50 new agents are being evaluated currently tor support (to maintain neutrophils and hemoglobin), and
in clinical trials, especially agents targeting proteins and bisphosphonates. The 2 bisphosphonates currently avail-
genes believed to be involved in the pathogenesis of breast able, pamidronate and zoledronic acid (others are being
cancer: antiangiogenesis agents (bevacizumab, SU-11248), investigated), are used in the setting of lytic bone me-
antimetabolites (gemcitabine, pemetrexed), topoisomerase tastases to ameliorate pain and decrease the risk of skeletal-
inhibitors (irinotecan), farnesyl transferase inhibitors related events such as fractures.
(tipifarnib), epidermal growth factor receptor inhibitors
(gefitinib, erlotinib, cetuximab, lapatinib), novel taxanes CONCLUSIONS
(ABI-007) and other tubulin-stabilizing agents (ixabepi- Breast cancer management is a rapidly evolving field. New
lone), raf-1 kinase inhibitors (BAY 43.9600), and others. technologies are being incorporated to better understand
Physicians and patients should seek available information the biology of this disease and to help identify the genes
from clinical trials to help improve the outcome of patients involved in prognosis and responsiveness to therapy. These
with advanced breast cancer. findings, added to improved risk assessment and preven-
tion strategies, as well as screening, diagnosis, treatment,
Treatment of Patients With HER2-Positive Disease and supportive care, hold great promise for the future.
Patients with HER2-positive disease (defined as either Education and translational clinical trials will be the key to
positive for HER2 gene amplification by fluorescence in success.
situ hybridization or HER2 3+ positive protein overex-
pression by immunohistochemistry) are treated differently
REFERENCES
than are those with HER2-negative breast cancer, at least in 1. Jemal A, Tiwari RC, Murray T, et al. Cancer statistics, 2004. CA
terms of first-line chemotherapy for metastatic disease. If Cancer J Clin. 2004;54:8-29.
the tumor is ER-positive and/or PR-positive and HER2- 2. Humphrey LL, Helfand M, Chan BKS, Woolf SH. Breast cancer
positive, hormonal therapy alone (without trastuzumab) screening: a summary of the evidence for the U.S. Preventive
Services Task Force. Ann Intern Med. 2002;137(5, pt 1):347-360.
may be appropriate if the patient has only soft tissue 3. Smith RA, Saslow D, Andrews Sawyer K, et al, High-Risk Work
or bone disease and no evidence of rapidly progressive Group, Screening Older Women Work Group, Mammography
metastases. Work Group, Physical Examination Work Group, New Technolo-
Patients with visceral disease or symptomatic progres- gies Work Group, Breast Cancer Advisory Group. American Can-
cer Society guidelines for breast cancer screening: update 2003. CA
sive tumors that are no longer responding to hormones or Cancer J Clin. 2003;53:141-169.
are ER-negative and/or PR-negative should be treated with 4. Mincey BA. Genetics and the management of women at high risk
the anti-HER2 monoclonal antibody trastuzumab added to for breast cancer. Oncologist. 2003;8:466-473.
5. Boyd NF, Martin LJ, Stone J, Greenberg C, Minkin S, Yaffe MJ.
chemotherapy. This recommendation is based on the sur-
Mammographic densities as a marker of human breast cancer risk
vival advantage of this approach compared with chemo- and their use in chemoprevention. Curr Oncol Rep. 2001;3:314-
therapy alone. It is recommended that trastuzumab not be 321.

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
816 Breast Cancer Mayo Clin Proc, June 2004, Vol 79

6. Cuzick J, Powles T, Veronesi U, et al. Overview of the main 2. Which one of the following is the only agent currently
outcomes in breast-cancer prevention trials. Lancet. 2003;361:296- approved by the US Food and Drug Administration
300. to decrease the risk of primary breast cancer
7. Green FL, Page DL, Fleming ID, et al, American Joint Committee
on Cancer, eds. AJCC Cancer Staging Handbook: From the AJCC development?
Cancer Staging Manual. 6th ed. New York, NY: Springer-Verlag; a. Raloxifene
2002. b. Tamoxifen
8. Recht A. Integration of systemic therapy and radiation therapy for
c. Idoxifene
patients with early-stage breast cancer treated with conservative
surgery. Clin Breast Cancer. 2003;4:104-113. d. Alendronate
9. National Institutes of Health Consensus Development Panel. Na- e. Risedronate
tional Institutes of Health Consensus Development Conference
Statement: adjuvant therapy for breast cancer, November 1-3,
3. In which one of the following situations is external
2000. J Natl Cancer Inst. 2001;93:979-989. beam radiotherapy not routinely recommended?
10. Mincey BA, Palmieri FM, Perez EA. Adjuvant therapy for breast a. After lumpectomy for invasive breast cancer
cancer: recommendations for management based on consensus re-
view and recent clinical trials. Oncologist. 2002;7:246-250.
b. After mastectomy in patients with 1 to 3 involved
11. Pritchard KI. The best use of adjuvant endocrine treatments. axillary lymph nodes
Breast. 2003;12:497-508. c. After mastectomy for patients with >3 involved
12. ATAC (Arimidex, Tamoxifen Alone or in Combination) Trialists’ axillary lymph nodes
Group. Anastrozole alone or in combination with tamoxifen versus d. After mastectomy for a primary tumor >5 cm
tamoxifen alone for adjuvant treatment of postmenopausal women e. After lumpectomy for large DCIS (>2.5 cm)
with early-stage breast cancer: results of the ATAC (Arimidex,
Tamoxifen Alone or in Combination) trial efficacy and safety 4. Which one of the following is the current
update analyses. Cancer. 2003;98:1802-1810. recommended duration of adjuvant tamoxifen
13. Goss PE, Ingle JN, Martino S, et al. A randomized trial of letrozole treatment for patients with resected hormonally
in postmenopausal women after five years of tamoxifen therapy for
early-stage breast cancer. N Engl J Med. 2003;349:1793-1802.
responsive breast cancer?
14. Coombes RC, Hall E, Gibson LJ, et al, Intergroup Exemestane a. 6 months
Study. A randomized trial of exemestane after two to three years of b. 1 year
tamoxifen therapy in postmenopausal women with primary breast c. 5 years
cancer. N Engl J Med. 2004;350:1081-1092.
d. 10 years
e. For life
Questions About Breast Cancer 5. Which one of the following proteins is targeted by the
monoclonal antibody trastuzumab?
1. Which one of the following is not included in the Gail a. Raf-1 kinase
model for prediction of breast cancer risk? b. HER1
a. Presence of the BRCA1 gene mutation c. HER2
b. Family history of breast cancer in first-degree d. HER3
relatives e. HER4
c. History of atypical ductal hyperplasia
d. Number of prior breast biopsies Correct answers:
e. Age at menarche 1. a, 2. b, 3. b, 4. c, 5. c

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.

You might also like