Breast Cancer
Synonyms and related keywords: carcinoma of the breast, cancer of the breast, adenocarcinoma of the breast, infiltrating ductal carcinoma, invasive ductal carcinoma, infiltrating lobular carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, DCIS, lobular carcinoma in situ, LCIS, axillary lymph node dissection, ALND, BRCA1, BRCA2, Li-Fraumeni syndrome, Cowden disease, sentinel lymph node biopsy, SLNB, breast-conserving surgery, BCS, lumpectomy, radiation therapy, RT, atypical ductal hyperplasia, ADH, atypical lobular hyperplasia, ALH, radiotherapy, radiation oncology, chemotherapy, hormonal therapy, HT, radical mastectomy, RM, total mastectomy, TM, skin-sparing mastectomy, SSM, quadrantectomy, breast reconstruction, bone marrow micrometastasis, BMM, selective estrogen receptor modulators, SERMS, selective estrogen receptor down-regulators, SERDs, screening mammography, screening mammogram, lymphedema, estrogen-positive tumors, SBLLA syndrome, Peutz-Jeghers syndrome, Muir-Torre syndrome, nipple discharge, peau d'orange sign, skin dimpling, nipple retraction, lupus, Paget disease of the breast, axillary dissection, phantom breast syndrome
INTRODUCTION
For excellent patient education resources, visit eMedicine's Cancer and Tumors Center, Women's Health Center, and Imaging Center. Also, see eMedicine's patient education articles Breast Cancer, Mastectomy, Breast Lumps and Pain, Breast Self-Exam, Mammogram, and Ovarian Cancer.
Problem: Over the past few decades, breast cancer management has undergone significant changes characterized by less aggressive approaches to diagnosis and treatment. Mammogram and ultrasound or stereotactic biopsies have supplanted clinical diagnosis and surgical biopsy for the diagnosis; breast-conserving surgery (BCS) and sentinel lymph node biopsy (SLNB) have successfully replaced the more aggressive radical mastectomy (RM) and axillary lymph node dissection (ALND).
These changes are the results of a century-long experience whereby different models for the disease have been proposed and tested. RM, introduced by Halsted at the end of the last century, was based on the centrifugal model of tumor spread, according to which cancer spread starts locally then moves to the lymphatics and only then invades distant organs. Despite increasingly radical procedures, most patients relapsed with systemic disease.
New paradigms appeared to palliate the deficiency of this model. Both the systemic and the spectrum models acknowledge the role of the blood stream in tumor dissemination independent of lymphatic invasion, but they differ in their explication of the relationship between tumor size and distant metastases.
The systemic model considers breast cancer a systemic disease from its inception, while the spectrum model views breast cancer as a progressive disease in which invasion and metastases are a function of tumor growth and biological transformation. In addition, this model acknowledges that the disease may manifest over a spectrum of biological behaviors, with tumors that are metastatic from the beginning and others that may reach large sizes without dissemination.
Modern trials comparing different forms of locoregional control for patients at the same stage of disease show that variations in local treatment do not result in differences in long-term survival, validating the first premise of these models regarding the inadequacy of local approaches to control the disease in the absence of systemic therapy. In addition, screening mammography results in early detection of breast cancer (average size 1.4 cm vs 2.2 cm for tumors clinically detected) and is associated with a 25% decrease in the mortality rate for breast cancer, thus lending credibility to the spectrum model. This model stresses the importance of both local and systemic treatment.
ALND remains one of the mainstays of breast cancer management because clinical, imaging, or biological methods are insufficient to reliably define nodal status, the most reliable predictor of final outcome. Furthermore, ALND allows local control of the disease and may also improve survival. However, the extent of axillary dissection is still debated. Knowing that the procedure has risks of chronic morbidity in terms of arm mobility and lymphedema, the question is not a trivial one. Results from several prospective studies show that 10 nodes or more should be removed and found negative before declaring the axilla stage N0. This involves a level I dissection and, usually, a level II dissection.
Early detection of breast cancer in the screening mammogram era has raised the question of whether extensive axillary dissection could be replaced by more limited procedures or SLNB. Less invasive techniques, such as positron emission tomography scan and endoscopic axillary clearance, are being evaluated for staging and local treatment.
The indications for postoperative radiation therapy (RT) are being redefined for small estrogen-positive tumors, particularly in postmenopausal women.
Immediate reconstruction after mastectomy became an accepted practice that proved to be safe and compatible with early detection of recurrence. Because breast cancer is being detected at increasingly smaller sizes, plastic surgery will likely play a larger role in the initial treatment of breast cancer.
Frequency: The American Cancer Society estimated that 211,240 new cases of breast cancer (32.2% of all cancers in women) would be diagnosed in 2005 in the United States, making breast cancer the most-diagnosed cancer in women. Male breast cancer is a rare disease and 1690 cases were expected for 2005 in the US. The true incidence rates of breast cancer have been stable from 1987-1996 after a constant increase since 1979 (increase of 1% per year from 1979-1982; 4% per y from 1982-1987). The lack of decline of breast cancer incidence in the 1990s contrasts with a slight decline (decline of 1.3% per y from 1992-1997) of the incidence rate of cancer for all sites. Up to 40,870 cases of breast cancer-related deaths were expected for 2005 in the US.
Based on cancer cases diagnosed from 1995-1997, the probability of developing invasive cancer is 0.44% (1 in 225) for women younger than 39 years, 4.15% (1 in 24) for women aged 40-59 years, and 7.02% (1 in 14) for women aged 60-79 years. The estimated lifetime probability of developing breast cancer is 12.83% (~1 in 8). The likelihood of developing breast cancer is higher in white women than in women of any other racial or ethnic group.
Although the death rate from breast cancer has decreased an average of 2.2% per year from 1990-1997, the recorded number of deaths from breast cancer has remained stable, at approximately 43,000 per year. Deaths dropped to 41,737 in 1998 after reaching the highest number, 43,844, in 1995. Among women aged 20-59 years, breast cancer is the leading cause of death from cancer. However, lung cancer remains the leading cause of death from cancer in women aged 60 years or older.
Etiology: Breast cancer is a heterogeneous disease with no single characterized cause.
Epidemiological studies have identified many risk factors that increase the chance for a woman to develop breast cancer:
- Factors with relative risk greater than 4
- Advanced age
- Being born in North America or northern Europe
- High premenopausal blood insulinlike growth factor (IGF)–1 level
- High postmenopausal blood estrogen level
- History of mother and a sister with breast cancer
- Factors associated with a relative risk of 2-4
- High socioeconomic status
- Age at first full-term pregnancy older than 30 years
- History of cancer in one breast
- Any first-degree relative with a history of breast cancer
- History of a benign proliferative lesion, dysplastic mammographic changes, and a high dose of ionizing radiation to the chest
- Factors associated with a relative risk of 1.1-1.9
- Nulliparity
- Early menarche (age less than 11 y)
- Late menopause (age >55 y)
- Postmenopausal obesity
- High-fat diet/saturated fat–rich diet
- Residence in urban areas and northern United States
- White race - Older than 45 years
- Black race - Younger than 45 years
- History of endometrial or ovarian cancer
- Identified factors with a protective role against breast cancer
- Age at first period older than 15 years
- Breastfeeding for longer than 1 year
- Monounsaturated fat–rich diet
- Physical activity
- Premenopausal obesity
Genetic factors
As with other cancers, breast cancer is the result of multiple genetic changes or mutations. Early mutations may be inherited (eg, mutations of breast stem cells) or acquired (eg, somatic mutations due to ionizing radiation, chemical carcinogens, or oxidative damage).
Estrogens, by their proliferation-promoting effect on the breast epithelium later, increase the chance of DNA replication errors leading to carcinogenic mutations. Indeed, the common denominator to many of these risk factors is their effect on the level and duration of exposure to endogenous estrogenic stimulation.
Early menarche, regular ovulation, and late menopause increase lifetime exposure to estrogens in premenopausal women, while obesity and hormone replacement therapy increase estrogen levels in postmenopausal women. Conversely, late menarche, anovulation, and early menopause (spontaneous or induced) are protective, owing to their effect on lowering the level or shortening the duration of estrogenic exposure.
Lactation and premenopausal obesity are associated with lower estrogen levels as a result of anovulation. For unknown reasons, pregnancy decreases breast tissue susceptibility to somatic mutations; thus, the earlier the first pregnancy, the shorter the susceptibility period.
Hereditary breast cancers have been thought to represent a small proportion (5-10%) of all breast cancers. However, based on new data derived from the comparison of identical and nonidentical twins, up to 27% of breast cancers may be attributed to inherited factors. The mutated genes BRCA1 and BRCA2 are responsible for approximately 30-40% of inherited breast cancers.
The prevalence of BRCA1 in the general population is 0.1%, compared with 20% in the Ashkenazi Jewish population. The gene is encountered in 3% of the unselected breast cancer population and in 70% of women with inherited early-onset breast cancer. Up to 50-87% of women carrying a mutated BRCA1 gene develop breast cancer during their lifetime.
Risks for ovarian and prostate cancers are also increased in carriers of this mutation. BRCA2 mutations are identified in 10-20% of families at high risk for breast and ovarian cancers and in only 2.7% of women with early-onset breast cancer. The lifetime risk of developing breast cancer in female carriers is 25-30%. BRCA2 is also a risk factor for male breast cancer; carriers have a lifetime risk of 6% for developing the cancer. BRCA2 mutations are associated with other types of cancers, such as prostate, pancreatic, fallopian tube, bladder, non-Hodgkin lymphoma, and basal cell carcinoma.
Li-Fraumeni syndrome, characterized by a mutation of TP53, is associated with multiple cancers, including the SBLLA syndrome (sarcoma, breast and brain tumors, leukemia, laryngeal and lung cancer). Cancer susceptibility is transmitted by an autosomal dominant pattern, with penetrance approximating 90% by age 70 years. Li-Fraumeni syndrome is identified in 1% of women with early-onset breast cancer. Bilateral breast cancer is noted in up to 25% of patients.
Cowden disease is a rare genetic syndrome associated with papillomatosis of the lips and oral mucosa, multiple facial trichilemmomas, and acral keratosis. The prevalence rate of breast cancer in women with this disease is 29%. Benign mammary abnormalities (eg, fibroadenomas, fibrocystic lesions, ductal epithelial hyperplasia, nipple malformations) are also more common. Other rare genetic disorders, such as Peutz-Jeghers and Muir-Torre syndromes, are associated with an increased risk of breast cancer.
Pathophysiology: In a normal state, cells proliferate in response to external proliferation-promoting signals to fulfill a function such as replacing lost cells or repairing injured tissues. Once the goal has been reached, a set of proliferation-repressing signals is activated. These signals allow the cells to exit the proliferation cycle (cell cycle) by returning to the dormant state (G0), by differentiating, or by dying (apoptosis). Each of these functions is carried out by a complex system of interacting proteins. Constitutive expression by mutation or another genetic change of any component of the proliferation-promoting system may result in uncontrolled proliferation. The constitutively expressed component is called an oncogene.
Conversely, the loss by mutation or deletion of a proliferation-repressing gene results in an inability to stop the cell cycle and, thereby, continuous proliferation, possibly leading to cancer. The lost gene is called a tumor suppressor gene. Likewise, constitutive expression of antiapoptotic genes may result in immortalization of the cell, paving the way for further genetic changes and eventually cancer formation. Loss of proapoptotic genes may lead to similar results. Thus, autonomous proliferation and immortality shared by all cancers are the final result of successive genetic changes, which may be different from one cancer to another.
Breast cancer is not an exception in that regard. It is the result of multiple genetic changes that are different from those of other malignancies and that confer to this cancer its characteristic phenotype.
Cell-cycle deregulation in breast cancer
Estrogen and progesterone induce cyclin D1 and c-myc expression. Although both sex hormones provide directionality by shifting the CDKI p21 from CDK2 to CDK4, progesterone promotes maturation by inducing p27, while only estrogen allows multiple cycles. Recent studies have reported common amplification of cyclin D1 (a third of breast cancers), inactivation of p16, and mutation of TP53 in breast cancer.
c-myc overexpression is one of the most common genetic alterations encountered in persons with breast cancer (a third of patients). Depending on the availability of its different partners, it may result in proliferation and chromosomal instability (Myc-Max) or differentiation (Myc-Mad), probably by sequestering Myc and reducing its availability. Amplification of the c-myc gene is associated with a poor prognosis and a high S-phase.
Estrogen receptor (ER)–positive breast cancer cells undergo apoptosis after withdrawal of estrogen, suggesting that this hormone functions not only as a mitogen but also as a survival factor. The antiapoptotic factor Bcl-2 is commonly overexpressed in ER-positive breast cancers.
ER negativity is observed in a third of primary breast cancers and a third of recurrences of ER-positive primaries. The ER gene is usually intact with no identifiable deletions or mutations. Although the exact mechanism of this lack of expression is not known, hypermethylation used normally by the genome to silence certain genes is a possible explanation. Methylation of cytosine-rich areas (called CpG islands) of the ER-gene promoter region has been described in the majority of ER-negative breast cancers and in a small fraction of ER-positive breast cancers. Demethylation of these areas with specific agents (eg, 5-azacytidine) restores ER expression and its function in vitro.
A progesterone receptor (PR) is present in approximately 50% of all ER-positive tumors. Its presence depends on the expression of functional ER, which explains its absence in almost all ER-negative breast cancers. The mitogenic effect of progesterone in breast cancer may depend on the induction of local growth hormone production in the hyperplastic mammary epithelium. However, high doses of progestins have proven inhibitory effects on breast cancer growth mediated by the down-regulation of G1-phase CDKs and cyclin D1 leading to cell differentiation.
Regarding adhesion-dependent cell regulation, the transmembrane glycoproteins, ie, epithelial cadherins (E-cadherins), mediate with their extracellular domain cell-to-cell interactions, thus stabilizing the cell in the epithelial tissue. Their intracellular domain interacts with and controls the transcription factors B-catenins. A mutation or the absence of E-cadherins results in cell detachment, increased motility and invasiveness, and release of B-catenins, which up-regulates c-myc expression.
Expression of E-cadherins is down-regulated in breast cancer. Another family of adhesion molecules, the integrins, is involved in cell-to-matrix interactions. Integrins signal through the Fak-Src pathway, which activates PI3K and AKT, resulting in enhanced survival, proliferation, and motility. The main components of this pathway (Fak, PI3K, and AKT) are inhibited by PTEN (ie, phosphatase on chromosome 10 gene product, mutated in Cowden disease), which results in the suppression of survival and apoptosis.
The epidermal growth factor (EGF) receptor family plays a critical role in mammary tumorigenesis. Other than the EGF receptor itself, 3 other members of this family have been described, including c-erb-B2 (HER2, HER2/neu), c-erb-B3, and c-erb-B4; the latter is called a kinase-dead receptor because it does not carry a kinase function on the cytoplasmic domain of the receptor, which is in contrast to the other members of the family.
These receptors interact with many ligands, including EGF, transforming growth factor (TGF)–alpha, hergulin (or Neu differentiation factor), heparin-binding EGF-like growth factor, beta-cellulin, and epiregulin. Upon binding of a ligand with its cognate receptor, a homodimerization or heterodimerization process occurs, followed by autophosphorylation of the intracellular domain and activation of the intrinsic catalytic domain.
Transmission of the signal is operated via phosphorylation of adaptor proteins (GRB2-SOS, Shc, IRS-1/2, STAT) docked or recruited to the cytoplasmic domain of the receptor, followed by activation of the RAS-GTP protein, then 1 of 3 pathways to the nucleus (ie, Raf/MEK/ERK-1/2, MEKK1/MEK4/7/JNK, PI3K/AKT/GSK).
Transmission of the signal from adaptor proteins to the nucleus without mediation by RAS is possible through Fak/Src or Rho/Rac/CDC42 pathways. In addition, phospholipase C-gamma is activated by direct interaction with the phosphorylated c-erb-B-2; however, its intracellular pathway is not fully known. The final result is the induction of transcription factors (ie, Myc, NF-kB, ATF, Ets, AP-1, EIK, SRF) that drive the cell cycle by up-regulating cyclins and inhibiting CDKIs and proapoptotic signals.
Once the biological message has been executed, the complex ligand receptor is internalized and destroyed in the lysosomes. The pattern of heterodimerization, the intracellular pathway used, and the rate of internalization and destruction of the receptor depend on the specific ligand bound to the receptor.
Although c-erb-B2 does not have a specific ligand, its role in the signal transmission from the epidermal growth factor receptor is crucial. Cell lines lacking c-erb-B2 are resistant to the tumorigenic effect of EGF, while those with a kinase-deficient or carboxyl terminal–truncated EGF receptor with intact c-erb-B2 can still execute all the functions of the wild type.
The discovery of the role of HER2 in breast cancer was one of the landmarks in breast cancer research in the last 2 decades. HER2 is overexpressed in 20-30% of breast cancers. Tumor cells overexpressing HER2/neu may have up to 2 million copies of the receptor on their surface compared with 20,000-50,000 copies in normal breast epithelial cells. Because of this abundance of HER2/neu, many heterodimers contain HER2/neu, resulting in potent intracellular signaling and malignant growth.
Despite persistent controversy regarding certain aspects of its biology, prognostic value, and methods of evaluation, HER2 overexpression or amplification is generally accepted to be correlated with a high histologic grade, the absence of hormone receptor expression, aneuploidy, a high proliferation index, tumor size, and a poor clinical outcome. Its role as a predictor of response to chemotherapy and hormonal therapy (HT) is not clearly defined.
Certain clinical studies using tamoxifen showed not only a lack of response, but also a detrimental effect in the group of patients overexpressing HER2. Retrospectively reviewed chemotherapy data showed longer disease-free survival and overall survival in HER2 overexpressors who received high doses of doxorubicin-containing chemotherapy compared with HER2-negative patients. Available data concerning the interaction between cyclophosphamide, methotrexate, and 5-fluorouracil (CMF) chemotherapy and HER2 overexpression are inconclusive and do not allow the formation of final conclusions. Taxanes seem to have high efficacy in patients overexpressing HER2 (relative risk, 65%) compared with the HER2-negative group (relative risk, 35%).
The IGF family consists of IGF-1, IGF-2, IGF receptor-1, IGF receptor-2, and IGF-binding proteins. The members of this family play an important role in normal mammary development and tumorigenesis. Both IGF-1 and IGF-2 bind to IGF receptor-1, whose strong mutagenic effect is synergistic with estrogen. In breast cancer, IGF receptor-1 and IGF-1 and IGF-2 are overexpressed in epithelial cells and stromal cells, respectively. Paradoxically, this overexpression correlates with a good prognosis, perhaps reflecting simple hormone dependence or association. IGF receptor-2 plays a tumor-suppressing role by down-regulating IGF-2.
The TGF-beta family consists of 3 TGF-beta and 2 interdependent serine-threonine kinase receptors. In normal mammary epithelial cells, TGF-beta blocks the expression of cyclin A, an S-phase–promoting protein and (to a lesser degree) cyclins D and E involved in the G1 phase and induces the expression of the CDKI p15, which results in cell-cycle arrest and, potentially, apoptosis. This explains its role in postlactational mammary gland regression.
Expression of TGF-beta in breast cancer is increased and seems to correlate with disease progression rather than tumor suppression. Mutation of TGF-beta receptors (type I and II) or any of the downstream molecules (Smad4) involved in intracellular signal transduction renders breast cancer cells resistant to its suppressive effects. However, its role in promoting angiogenesis and invasion and in suppressing the immune system becomes advantageous for the cancer cells, which acquire a proliferative advantage by losing sensitivity to TGF-beta and developing a way to escape the host immunosurveillance.
Mutated BRCA1 and BRCA2, breast cancer susceptibility genes, are proven risk factors for breast cancer. More than 500 mutations have been described in the BRCA1 gene (band 17q21), and 250 have been described in the BRCA2 gene (band 13q12-13). The mutations occurring at either end of the BRCA1 gene are associated with more aggressive tumors; those occurring at the 5` extremity are associated with breast and ovarian cancers, while those closer to the 3` end are associated with only breast cancer.
The biological function of the BRCA1 gene product is not well known. Accumulated evidence suggests that BRCA1 is a nuclear protein involved in other genes' expression, in cell cycle progression, and in the response to DNA damage. DNA damage results in activation and interaction of BRCA1, BRCA2, RAD51, and TP53 with subsequent expression of p21, which leads to a cell cycle pause until the damage is repaired. The mutation or absence of BRCA1 results in failure to repair the damaged DNA, and the cell cycle continues to accumulate further mutations, eventually leading to tumorigenesis. BRCA2 seems to play a role similar to that of BRCA1 in the cell cycle, other genes' expression, and DNA damage repair.
Clinical: In the past, the great majority of patients presented with a painless palpable mass. Although more than 80% of palpable masses are benign, the decision to observe such lesions should be made only after careful clinical, mammographic, and pathologic workup. Cystic lesions identified clinically or on ultrasound images should be explored using fine-needle aspiration (FNA) biopsy. Nonbloody fluid and complete resolution of the cyst confirm its benign nature. If the fluid is bloody or the cyst does not resolve after aspiration or has a complex appearance on the ultrasound image, a biopsy is indicated.
Other symptoms, such as breast pain or deformity, nipple discharge, and erythema or skin ulceration, occasionally occur. Patients with Paget disease present with a long-standing eczematoid rash of the nipple-areola complex, itching, tenderness, burning, and occasional bloody discharge from the nipple. Skin dimpling, the result of shortening or retraction of the Cooper ligaments induced by the tumor, does not have prognostic value, while the ominous peau d'orange sign reflects the invasion of the subdermal lymphatic plexus and portends a shortened survival.
Symptoms related to distant metastases, such as bone pain, dyspnea, or meningitic syndrome, are encountered in some cases.
In current practice, increasing numbers of breast cancers are mammographically diagnosed in the preclinical stage. Screening mammography has resulted in earlier diagnosis of breast cancer, which has translated in recent years into a 25% improvement in the mortality rate related to breast cancer. Mammographic signs suggestive of cancer include architectural distortions, microcalcifications, or masses. These changes require further evaluation using diagnostic mammograms with or without ultrasound. Biopsies are indicated if these changes are confirmed. Features helpful in the evaluation of palpable breast masses are as follows:
- Malignant masses
- Hard
- Painless: Malignant masses are painful in only 10-15% of patients.
- Irregular
- Possibly fixed to the skin or chest wall
- Skin dimpling
- Nipple retraction
- Bloody discharge
- Benign masses
- Firm, rubbery mass
- Frequently painful
- Regular margins
- Not fixed to skin or chest wall, mobile
- No skin dimpling
- No nipple retraction
- No bloody discharge
- Cysts: No reliable features distinguish cysts from solid masses based on clinical data.
Nipple discharge may be spontaneous or induced, unilateral or bilateral, and have different colors and textures. If the discharge is associated with one or more of the suggestive features, further investigation is necessary. Clinical characteristics of nipple discharges are as follows:
- Malignant discharge
- Unilateral
- Spontaneous
- One duct orifice
- Bloody, serosanguineous, or serous
- Benign discharge
- Bilateral
- Spontaneous or induced
- Multiple duct orifices
- Thick green or yellow, induced and bilateral (duct ectasia)
INDICATIONS
Modified RM is performed in the presence of contraindications to BCS or as a patient preference.
RELEVANT ANATOMY AND CONTRAINDICATIONS
WORKUP
- CT scan of the brain, chest, abdomen, and pelvis: Obtain CT scans if the patient has neurologic symptoms, abnormal chest radiograph results, supraclavicular lymphadenopathy and hepatosplenomegaly, or abnormal liver function test results.
- Skeletal radiograph: Use this for symptomatic areas only.
- Bone scan: Perform a bone scan if any of the following conditions are present:
- Scoring of HER2/neu overexpression using the DAKO HercepTest (DAKO Cytomation; Carpinteria, Calif) follows. The cell membrane staining pattern, the interpretation, and the score are listed.
- Strong complete membrane staining in more than 10% of tumor cells - Interpreted as strongly positive; score of 3+
- Weak-to-moderate complete membrane staining in more than 10% of cells - Interpreted as weakly positive; score of 2+
- Faintly perceptible membrane staining in more than 10% of tumor cells - Interpreted as negative; score of 1+
- No staining or staining in 10% of tumor cells - Interpreted as negative; score of 0
- Cytoplasmic staining of any intensity - Interpreted as negative; score of any
- Image-guided core-needle biopsy
- This is the preferred method for needle biopsy of a nonpalpable lesion. Note that because of sampling error, it carries a higher risk of false-negative findings than open biopsy. Negative or equivocal results in the face of suggestive mammogram findings or residual calcifications should be followed by an open biopsy. False-negative results are encountered in 1-10% of biopsies, with the highest rates occurring with the least-experienced operators.
- For the technique, ultrasound is the method of choice to guide the core-needle biopsy. Stereotactic mammographic guidance is used in lesions not visualized on ultrasound images. Stereotactic core-needle techniques have the advantages of lower complication rates and lower costs, although they cannot be used when the lesion is very close to the chest wall or areola, where open biopsy is the best approach. The radiograph should be compared with the mammogram to ensure that all calcifications are included within the core biopsy specimen.
- Open biopsy with needle localization
- Invasive localization techniques with small radiopaque needles to guide a surgical biopsy are used more commonly than noninvasive techniques.
- For the technique, local anesthesia with or without intravenous sedation is sufficient in most cases. A thin needle and a fine wire with a thickened distal segment are used for immediate preoperative localization of the lesion. The incision may include the wire entry site if the lesion is superficial. A core of tissue along and around the wire is excised (including the lesion easily identified by the previous placement of the thickened segment inside it) and sent en bloc for radiographic evaluation. However, when the wire entry site is located far from the lesion, the incision should be placed directly over the lesion; dissection is then performed to find the wire.
- Once identified, the free end of the wire is pulled up through the incision. A core of tissue around the wire is excised (including the lesion) and sent for radiographic evaluation. Closure should not proceed until radiographic confirmation that the entire lesion was excised.
- Fine-needle aspiration biopsy
- In experienced hands, FNA biopsy may provide a high accuracy rate when combined with physical examination and mammography (sensitivity ~80-98%, specificity ~100%). However, negative results from a palpable lesion cannot exclude carcinoma. Lesions most suited for this procedure are T3 and T4 tumors and axillary or chest wall relapses.
- Owing to the high false-negative rate with FNA in lesions smaller than 1 cm in diameter, another diagnostic procedure should be used. Because false-positive rates are extremely low (less than 2%), positive results are sufficient to plan surgery, without the need for further investigation. However, perioperative frozen sections are necessary to distinguish between invasive and in situ carcinoma and to determine the need for axillary dissection because FNA results cannot be used to make this distinction.
- According to some experts, a negative FNA biopsy finding, a physical examination suggestive of a benign lesion, and a normal mammography result (ie, the triple-negative criteria) are sufficient to stop the workup without further investigation.
- The equipment required is simple, ie, a small needle (22- to 25-gauge) for solid lesions or a larger one (20- to 21-gauge) mounted on a 10-mL syringe with 1-2 mL of air in it for easy visualization of the collected sample, which allows aspiration of a cystic lesion.
- After sterile preparation of the skin, the needle is advanced toward the lesion while stabilized by the fingers of the nondominant hand. Once the needle is in place, a back-and-forth movement of its tip along a 5- to 10-mm track is applied simultaneously with strong suction on the syringe. The suction is released when the collected sample reaches the syringe or the hub of the needle and before withdrawing the needle to prevent contamination with normal tissue.
- If the collected sample is fluid, it should be sent for analysis if (1) it is bloody, (2) a residual mass remains after complete aspiration, or (3) a recurrent cyst is present. If the sample is made of cellular material, it should be expelled on glass slides to make thin smears for cytological analysis.
- Cutting-needle (core-needle) biopsy
- Although the cutting-needle biopsy technique is associated with a higher true-positive rate than the FNA technique, a negative result may reflect sampling error. This biopsy provides a cylinder of tissue for pathological rather than cytological analysis. Determination of ERs and PRs is possible. Its best indications are large tumors and chest wall relapses. Small lesions or those surrounded by fibrocystic tissue are better studied using FNA.
- After applying local anesthesia to the skin, a small nick is made to allow the entry of the large biopsy needle (14 gauge), which can be placed by biopsy gun or by hand while the nondominant hand is stabilizing the lesion. A core sample is harvested and sent for pathological analysis.
- Excision (open) biopsy
- The entire lesion is removed with excisional biopsy, along with a margin of normal breast tissue.
- Local anesthesia, occasionally with intravenous sedation, is sufficient to perform most of these procedures. A curvilinear incision should be placed directly on the tumor mass and oriented in such a way that it could be included within a future mastectomy incision. In extremely lateral or medial lesions, a radial incision placed over the lesion is preferable.
- Once the tumor is removed, the margins should be inked. After hemostasis is achieved, deep breast tissue approximation is performed only if this does not result in deformity of the breast contour. Lastly, the skin is closed with a subcuticular closure.
- I - Ductal
- Intraductal (in situ)
- Invasive with predominant intraductal component: Infiltrating or invasive ductal cancer is the most common breast cancer histologic type, comprising 70-80% of all cases.
- Invasive, not otherwise specified
- Scirrhous
- Tubular
- Medullary with lymphocytic infiltrate
- Mucinous (colloid)
- Papillary
- Inflammatory
- Comedo
- Other
- II - Lobular
- III - Nipple
- IV - Undifferentiated carcinoma
- V - Rare tumor subtypes: The following are not considered typical breast cancers.
- Other terms used
- Ploidy
- Nuclear grade
- Mitoses
- Estrogen receptors
- HER2/neu overexpression
- Distribution
- Necrosis
- Local recurrence
- Prognosis
Mucinous carcinoma is another uncommon histologic type of invasive breast cancer (less than 5%). It is more common in the seventh decade of life and manifests clinically as a palpable mass or mammographically (with increasing frequency) as a poorly defined tumor with rare calcification. Its histologic hallmark is the presence of mucin production occupying more than 90% of the tumor in pure mucinous forms and variable percentages in the mixed forms. The cells are clustered in small islets dispersed in a pool of mucin. DCIS is often present in the vicinity of the tumor. ERs and PRs are positive in 90% and 68% of cases, respectively. HER2/neu overexpression is extremely rare. Patients with pure mucinous carcinoma have a better prognosis than those with mixed forms or other breast cancers of no special type.
- TX - Cannot be assessed
- T0 - No evidence of primary tumor
- Tis - Carcinoma in situ, intraductal carcinoma, LCIS, or Paget disease of the nipple with no associated tumor (Note: Paget disease associated with a tumor is classified according to the size of the tumor.)
- T1 - Tumor 2 cm or smaller in greatest dimension
- T2 - Tumor larger than 2 cm but not larger than 5 cm in greatest dimension
- T3 - Tumor larger than 5 cm in greatest dimension
- T4 - Tumor of any size with direct extension to (a) chest wall or (b) skin, only as described below (Note: Chest wall includes ribs, intercostal muscles, and serratus anterior muscle, but not pectoral muscle.)
- T4a - Extension to chest wall
- T4b - Edema (including peau d'orange) or ulceration of the skin of the breast or satellite skin nodules confined to the same breast
- T4c - Both of the above (T4a and T4b)
- T4d - Inflammatory carcinoma (Note: Inflammatory carcinoma is a clinicopathologic entity characterized by diffuse brawny induration of the skin of the breast with an erysipeloid edge, usually without an underlying palpable mass. Radiologically, a detectable mass and characteristic thickening of the skin may be present over the breast. This clinical presentation is due to tumor embolization of dermal lymphatics with engorgement of superficial capillaries.)
- NX - Cannot be assessed (eg, previously removed)
- N0 - No regional lymph node metastasis
- N1 - Metastasis to movable ipsilateral axillary lymph node(s)
- N2 - Metastasis to ipsilateral axillary lymph node(s) fixed to each other or to other structures
- N3 - Metastasis to ipsilateral internal mammary lymph node(s)
- pNX - Regional lymph nodes cannot be assessed (eg, not removed for pathologic study or removed previously)
- pN0 - No regional lymph node metastasis
- pN1 - Metastasis to movable ipsilateral axillary lymph node(s)
- pN1a - Only micrometastasis (none >0.2 cm)
- pN1b - Metastasis to lymph node(s), any larger than 0.2 cm
- pN1bi - Metastasis in 1-3 lymph nodes, any larger than 0.2 cm and all smaller than 2 cm in greatest dimension
- pN1bii - Metastasis to 4 or more lymph nodes, any larger than 0.2 cm and all smaller than 2 cm in greatest dimension
- pN1biii - Extension of tumor beyond the capsule of a lymph node metastasis, smaller than 2 cm in greatest dimension
- pN1biv - Metastasis to a lymph node 2 cm or larger in greatest dimension
- pN2 - Metastasis to ipsilateral axillary lymph node(s) fixed to each other or to other structures
- pN3 - Metastasis to ipsilateral internal mammary lymph node(s)
TREATMENT
Treatment of in situ disease
Predictors of recurrence in patients undergoing BCS for DCIS are as follows:
- Residual microcalcifications: Risk of relapse is 100%.
- Comedo necrosis
- Positive margins: For margins less than 1 mm, the recurrence rate is 25%. For margins of 1-9 mm, the recurrence rate is 15%. For margins greater than 1 cm, the recurrence rate is 3%.
- Age younger than 50 years
- Bloody discharge
Treatment of invasive disease
Doxorubicin (Adriamycin) and cyclophosphamide (AC) has a threshold effect; thus, doses greater than 60 mg/m2 and 600 mg/m2, respectively, are of no additional benefit. The results of 3 large US trials (NSABP B22 and B25, Cancer and Leukemia Group B 9344) did not support any role for dose intensification of the AC combination. However, 2 other studies, one French (French Adjuvant Study Group -05) and the other Canadian, showed that when epirubicin was escalated in the fluorouracil-epirubicin-cyclophosphamide (FEC) combination, disease-free survival and overall survival were significantly improved in operable breast cancer with positive axillary lymph nodes.
High-dose chemotherapy with stem cell or bone marrow support did not prove superior to standard chemotherapy and is best reserved for clinical trials.
Results from two trials (Cancer and Leukemia Group B 9344 and NSABP-B28) exploring the role of taxanes in the adjuvant setting were encouraging. CALGB-9344 had a 3x2 factorial design with patients randomized to receive doxorubicin at 60, 75, or 90 mg/m2 followed by paclitaxel at 175 mg/m2 or no additional therapy. NSABP-B28 had similar design except that the dose of doxorubicin was not escalated and the dose of paclitaxel was 225 mg/m2. From the first trial it was established unequivocally that doxorubicin dose escalation does not improve outcome while the addition of paclitaxel results in small but statistically significant improvement of both the risk of relapse and the risk of death. In NSABP-B28, relapse free survival was significantly improved by the addition of paclitaxel while the data about survival are not available yet.
In patients with node-negative, early-stage disease (stages I, IIA, and IIB), chemotherapy is not indicated if the tumor is smaller than 0.5 cm, regardless of the histologic subtype. In persons with invasive ductal and lobular carcinomas, tumors measuring 0.6-1 cm require chemotherapy only if they were associated with unfavorable features (eg, angiolymphatic invasion, high S-phase, high nuclear grade, high histologic grade), while all tumors larger than 1 cm require adjuvant chemotherapy alone (hormone-receptor negative) or in combination with tamoxifen (hormone-receptor positive). In patients with other histologic types (ie, tubular, colloid, medullary, adenoid), chemotherapy is indicated only for tumors larger than 3 cm. It may be considered in those from 1-2.9 cm, and it is not indicated for tumors smaller than 1 cm.
In patients with node-positive, early-stage disease, adjuvant chemotherapy with or without adjuvant HT is the mainstay of treatment. Anthracycline-containing chemotherapy, such as doxorubicin (Adriamycin) (60 mg/m2) and cyclophosphamide (600 mg/m2), administered for 4 cycles is the treatment of choice. Many oncologists consider this treatment insufficient and opt for 2 more cycles of the same treatment or 2-4 cycles of paclitaxel (175 mg/m2).
In persons with advanced-stage disease (stage IIIA and IIIB), the same regimen of AC for 4 cycles followed by at least 2 more cycles of the same treatment or 4 cycles of a taxane is recommended after surgical treatment. Neoadjuvant chemotherapy may be offered to patients with this stage.
The role of neoadjuvant chemotherapy is being intensively investigated. The NSABP B-18 trial compared the effect of preoperative AC for 4 cycles to the same regimen postoperatively in subjects with early-stage breast cancer. Pathologic node-negativity of 60% and BCS of 68% could be achieved in the preoperative AC group compared with 42% and 60%, respectively, in the postoperative AC group. Five-year disease-free survival and overall survival rates for those who achieved pathologic complete response were 84% and 87% compared with 72% and 78%, respectively, in patients who had residual disease.
Based on these encouraging results, NSABP started its B-27 trial to address the question related to the role of 4 cycles of preoperative or postoperative docetaxel added to 4 AC cycles. The use of preoperative docetaxel almost doubled the pathologically confirmed complete remission rate compared with the AC arm (25.6% vs 13.7%).
Because the role of neoadjuvant therapy is not fully established, participation in clinical trials should be encouraged. In ER- and/or PR-positive tumors, neoadjuvant tamoxifen or letrozole may achieve the same magnitude of response as chemotherapy but with longer time to response. Although the neoadjuvant approach does not seem to prolong survival, its theoretical advantages include downstaging the tumor, in vivo testing of the chemosensitivity of the tumor, and allowing BCS.
The treatment of metastatic disease is mainly medical. In postmenopausal patients with ER- and/or PR-positive tumors, the use of tamoxifen or an aromatase inhibitor (eg, anastrozole, letrozole, exemestane) is the standard of care for bone disease and limited visceral disease. In many cases, at least a partial response can be achieved. Patients who progress on first-line HT may still respond to second- or third-line HT, aromatase inhibitors, and megestrol acetate, respectively.
ER down-regulators are a promising new class of HT agents that may have some efficacy if the previous treatments fail. In premenopausal women with ER- and/or PR-positive breast cancer, ovarian ablation and tamoxifen are the mainstays of treatment. Ovarian ablation can be achieved by medical (eg, luteinizing hormone-releasing hormone analogues such as goserelin, leuprolide, buserelin, and triptorelin), surgical (ie, bilateral oophorectomy), or RT methods.
Chemotherapy is indicated for patients with advanced visceral disease (visceral crisis) and those with hormone-refractory or hormone-insensitive tumors. The goals of chemotherapy in this setting are palliative and include control of symptoms, control of disease progression, and prolongation of life. The best response rates are obtained with first-line therapy and combination regimens. However, regardless of the response rates achieved with these therapies (including high-dose chemotherapy with autologous stem cell support), survival is not affected in most cases.
In 2001, Slamon and Pegram reported a survival advantage for the combination of chemotherapy and trastuzumab over chemotherapy alone in women with HER2-positive metastatic breast cancer.
Table 1. Single-Agent Chemotherapy for Metastatic Breast Cancer
Drug | Class | Dose/Schedule | Overall Response Rate | Toxicity | Comments |
Paclitaxel | Microtubule-stabilizing | 175 mg/m2 as 3-h infusion q3wk | 21-32% | Myelosuppression, alopecia, neuropathy, myalgias, arthralgias, and allergic reactions | Corticosteroids are used to prevent allergic reactions, especially after first doses. They may be omitted later if no adverse effects occur. |
80-100 mg/m2/wk | 23-53% | Mild and noncumulative myelosuppression; with prolonged use, neuropathy, fatigue, and fluid retention are limiting toxicities | |||
Docetaxel | Microtubule-stabilizing | 75-100 mg/m2 q3wk | 30-68% | Myelosuppression, alopecia, skin reaction, mucositis, and fluid retention | Corticosteroids are used to prevent fluid retention. Initiate diuretics with the first signs of fluid retention. |
35-45 mg/m2/wk for 6 cycles, with 2 wk off | 29-50% | ||||
Doxorubicin | Anthracycline | 45-60 mg/m2 q3wk; not to exceed cumulative dose of 450-500 mg/m2 | 35-50% | Myelosuppression, mucositis, nausea, vomiting, and myocardial dysfunction | Dexrazoxane may be used as a cardioprotectant. |
Epirubicin | Anthracycline | 90 mg/m2 q3wk; not to exceed cumulative dose of 900 mg/m2 | 35-50% | Myelosuppression, mucositis, nausea, vomiting, and myocardial dysfunction | |
Doxil (liposomal encapsulated doxorubicin) | Liposomal anthracycline | 20 mg/m2 IV q3wk | Less cardiotoxicity, neutropenia, and alopecia; stomatitis and hand-foot syndrome | ||
Capecitabine | Oral fluoro-pyrimidine | 2500 mg/m2/d for 2 wk, with 1 wk off | 25-30% | Rash, hand-foot syndrome, mucositis, diarrhea, and mild neutropenia | |
Vinorelbine | Microtubule-assembly inhibitor | 25-30 mg/m2/wk | 35-45% | Myelosuppression, neuropathy, alopecia, constipation, fatigue, and phlebitis | |
Carboplatin | Same as cisplatin | Doses calculated for an area under the curve of 5 or 6. | Neuropathy, nausea, vomiting, kidney dysfunction, and mild myelosuppression |
Predictors of poor response to chemotherapy in patients with metastatic breast cancer are poor performance status, multiple and/or visceral sites of disease, short disease-free intervals, and failure to respond to prior chemotherapy. Because the goals of chemotherapy in patients with metastatic breast cancer are palliative, many authorities recommend the sequential use of single chemotherapeutic agents rather than combinations in order to limit toxicity. Taxanes, anthracyclines, oral fluoropyrimidines, vinorelbine, and gemcitabine are the most effective drugs used in this setting.
Trastuzumab, a monoclonal antibody directed against the extracellular domain of the HER2/neu receptor, has shown significant antitumor activity in patients with metastatic breast cancer overexpressing HER2/neu. Response rates of 30-35% have been observed in patients with metastatic breast cancer who are receiving single-agent trastuzumab as a first-line therapy. The relative risk of death was decreased by 20% with a median follow-up of 30 months when trastuzumab was used in combination with chemotherapy. However, when combined with doxorubicin, a significant increase in cardiotoxicity was noted. For this reason, the current recommendation is to avoid trastuzumab in combination with or after doxorubicin.
The mechanism of action of trastuzumab is still debated. Trastuzumab induces down-regulation of HER2/neu and prevents its heterodimerization, reestablishing breast cancer cell sensitivity to HT and chemotherapy. As a result of this down-regulation, p27 is induced, resulting in cell-cycle arrest in the G1 phase. Furthermore, trastuzumab binds to the receptor at a site where the extracytoplasmic domain (ECD) is usually cleaved by metalloproteinases. The ECD cleavage results in a constitutively active truncated receptor and a more aggressive phenotype; this would be prevented by binding to trastuzumab. Finally, trastuzumab may induce antibody-dependent cytotoxicity, resulting in cell death.
Four large multicenter trials were conducted with trastuzumab in the adjuvant setting in patients with HER2/neu-positive breast cancer (NSABP B31, NCCTG N9831, The HERA trial and BCIRG 06). A combined analysis of the first two trials showed dramatic improvement of DFS in the group randomized to receive trastuzumab with chemotherapy consisting of AC x 4 followed by T x 4.
Radiotherapy
RT reduces the risk of local recurrence and has the potential to decrease long-term mortality from breast cancer. Although certain studies have shown that RT following chemotherapy results in better long-term survival rates than the opposite, recent updates from Bellon et al and the Joint Center randomized trial showed no significant differences between the CT-first and RT-first arms in any endpoint studied. A reduction of approximately 20% in local recurrence correlates with an absolute reduction of approximately 5% in long-term mortality from breast cancer 10-15 years later.
RT to the breast (with or without the supraclavicular area) is indicated after lumpectomy in persons with early-stage breast cancer as an integral part of the treatment plan, and it is indicated after mastectomy in the presence of a large tumor mass (>5 cm), positive margins, and 4 or more lymph nodes positive for disease.
The role of axillary RT after radical ALND is debated. Many experts believe that RT is best avoided after complete dissection of the axilla for level I, II, and III nodes (RM and modified RM). A 6- to 8-fold increase in the incidence of lymphedema in the ipsilateral arm was reported with the combined modality.
Table 2. Adjuvant Chemotherapy Regimens for Breast Cancer
Regimen | Dose and Schedule | Cycle Interval, d | Cycles |
---|---|---|---|
CMF (standard) Cyclophosphamide Methotrexate 5-Fluorouracil | 100 mg/m2/d PO for 14 d 40 mg/m2/d IV days 1 and 8 600 mg/m2/d IV days 1 and 8 | 28 28 28 | 6 6 6 |
CMF (IV; in node-negative patients) Cyclophosphamide Methotrexate 5-Fluorouracil | 600 mg/m2 IV 40 mg/m2 IV 600 mg/m2 IV | 21 21 21 | 12 12 12 |
CAF Cyclophosphamide Doxorubicin (Adriamycin) 5-Fluorouracil | 100 mg/m2/d PO for 14 d 30 mg/m2/d IV days 1 and 8 500 mg/m2/d IV days 1 and 8 | 28 28 28 | 6 6 6 |
CAF Cyclophosphamide Doxorubicin 5-Fluorouracil | 600 mg/m2 IV day 1 60 mg/m2 IV day 1 600 mg/m2/d IV days 1 and 8 | 21-28 21-28 21-28 | 4-6 4-6 4-6 |
AC Doxorubicin Cyclophosphamide | 60 mg/m2 IV day 1 600 mg/m2 IV day 1 | 21 21 | 4 4 |
AC followed by paclitaxel (Taxol) Doxorubicin Cyclophosphamide Paclitaxel | 60 mg/m2 IV day 1 600 mg/m2 IV day 1 175 mg/m2 IV day 1 | 21 21 21 | 4 4 4 (after AC) |
AC followed by CMF Doxorubicin Cyclophosphamide Methotrexate 5-Fluorouracil | 75 mg/m2 IV day 1 600 mg/m2 IV day 1 40 mg/m2 IV day 1 600 mg/m2 IV day 1 | 21 21 21 21 | 4 8 (cycles 5-12) 8 (cycles 5-12) 8 (cycles 5-12) |
Surgical therapy:
Procedures
RM is the en bloc resection of the breast, the overlying skin, the pectoralis muscles, and all of the axillary contents (level I, II, and III dissection). Extended RM is RM with removal of the internal mammary nodes. Both procedures are rarely performed in current practice. In addition to their mutilating effects, they result in functional impairment as a result of neurologic and lymphatic vessel damage. Resection of the involved portion of the muscle is satisfactory even when the tumor abuts or invades the pectoral fascia or when the muscle fascia was violated at the time of biopsy.
Modified RM implies the removal of breast tissue, the underlying fascia of the pectoralis major muscle, and some of the axillary lymph nodes. The nipple-areola complex and the area around the biopsy incision must be removed, but the remainder of the skin of the breast can be preserved.
Total mastectomy removes the entire breast. Both pectoralis muscles and the axillary nodes are preserved.
Skin-sparing mastectomy removes the breast parenchyma, the previous biopsy site(s), and the skin overlying superficial tumors through a periareolar incision. Thus, the breast envelope is preserved, allowing immediate reconstruction with improved cosmetic results. Skin-sparing mastectomy can be coupled with SLNB.
BCS (lumpectomy) removes the tumor with 0.5-1 cm of normal tissue. It is often coupled with ipsilateral axillary lymph node sampling and, increasingly, with SLNB. Quadrantectomy is the removal of the quadrant containing the cancer with the overlying skin and underlying pectoral fascia.
ALND, when performed in a 2-cm primary breast cancer to levels I and II, reduces the probability for regional recurrence from 20% to approximately 3%.
Regarding SLNB, the combined use of a radionuclide and a blue dye increases the detection rate of the SLN. Isosulfan blue (Lymphazurin) is the most commonly used blue dye. Five mL of isosulfan blue are injected into the parenchyma, and the injection site is gently massaged for 3-5 minutes before the SLNB procedure is performed. Technetium sulfur colloid, the radionuclide of choice, is used at a dose of 0.1-1 mCi. The isotope is injected into the parenchyma 1-2 hours before the procedure. The SLN is preoperatively localized using a handheld gamma probe.
Preoperative details: Conduct a thorough preoperative assessment, and discuss with the patient and family the risks related to the cancer itself or to any comorbid conditions that may impact the intended surgery. Focus this discussion on prognostic factors, therapeutic options for the stage of the disease (with their expected benefits and adverse effects), and alternatives to surgery if available.
With young patients or those who desire a reconstruction, also discuss immediate versus delayed plastic surgery. For all patients with locally advanced disease, seek consultation with a medical oncologist to consider the role of neoadjuvant chemotherapy.
Intraoperative details: Whenever BCS is performed, the surgeon has the responsibility to ensure that all involved tissues are removed. Frozen-section studies are performed perioperatively, followed by permanent-section studies. Perform total mastectomy if the margins are not clear or if the specimen contains multicentric disease.
Lumpectomy
Lumpectomy or wide local excision may be performed with the patient under local anesthesia if no axillary node dissection is planned. General anesthesia is preferred for large excisions or if axillary dissection is intended. A long, curvilinear incision is placed directly over the lesion in such a way as to allow its inclusion in a future mastectomy incision if the margins are positive.
A radial incision is better suited for lesions located far laterally or in the areolar area. The dissection is performed around the palpable tumor, which is removed with 1-1.5 cm of grossly normal tissue. In cases of a nonpalpable lesion, remove 2-3 cm of tissue around the localization wire.
Exercise care for proper orientation and margin inking of the specimen to ensure appropriate pathological study of the margins. Reexcision can be performed immediately if instantaneous study results are available, or it can be performed later. Subcuticular closure is performed after appropriate hemostasis and approximation of deep tissues if the latter does not result in major deformity of the breast contour.
Axillary dissection
In axillary dissection, include level I and II lymph nodes and axillary fibrofatty tissue. Level III lymph nodes are preserved unless gross disease is present. The borders of the dissection are the axillary vein and the medial aspect of the pectoralis minor muscle superiorly, the latissimus dorsi muscle laterally, the pectoral muscles and the anterior serratus muscle medially, and the tail of the breast inferiorly.
Make a transverse incision in the lower third of the hair-bearing skin of the axilla; the incision can be extended posteriorly to the latissimus dorsi muscle for larger exposure. The dissection proceeds through the true axillary fat pad and its fascia to allow exposure of major pectoralis and latissimus dorsi muscles. Recognition of anatomical landmarks such as the axillary vein, the thoracodorsal bundle, and the long thoracic nerve help to maintain proper orientation and to preserve important structures.
Low-lying branches of the brachial plexus are preserved after identification of the axillary vein under the pectoralis major muscle medially. The thoracodorsal bundle is identified at its junction with either the axillary vein or the latissimus dorsi muscle. Medial to this bundle, the long thoracic vein is recognized lying on the chest wall.
Retraction is applied on the pectoralis major and latissimus dorsi muscles to ensure good exposure. The fat and the lymph nodes located medial to the thoracodorsal bundle are removed up to the level of the axillary vein, and then the dissection proceeds medially to level II fat and nodes beneath the pectoralis major muscle, where the medial pectoral nerve may be encountered.
Dissection of the axillary fat continues inferiorly along the anterior serratus muscle, where the long thoracic nerve is identified and preserved. Lastly, the axillary fat is released from its attachment to the tail of the breast by knife or electrocautery. Most surgeons drain the axilla after axillary dissection.
Sentinel lymph node biopsy
This procedure can be performed with the patient under local or general anesthesia. The isotope and the blue dye are injected 2 hours and 5 minutes before the procedure, respectively. Through an incision made below the "hot spot" identified by the gamma probe, the surgeon can identify a blue-green lymphatic representing the afferent lymphatic leading to the SLN.
The removed SLN is examined ex vivo using the gamma probe to determine its count, which serves as a reference for a reexamination of the axilla. Remove hot spots with counts exceeding 10% of the hottest ex vivo node. In patients undergoing lumpectomy, perform the SLNB before the lumpectomy; for those undergoing modified RM, perform the procedure during ALND before or after the mastectomy.
Mastectomy
Mastectomy (modified radical or simple) implies removing all the breast tissue and the overlying skin, including the nipple-areola complex, and leaving viable skin flaps. Whether a transverse or an upward-angled incision is chosen, include the nipple-areola complex and previous biopsy incisions in the excised area.
After determining upper and lower endpoints and while the breast is pulled firmly downward, a straight line is drawn to define the upper incision. The lower incision is similarly drawn while the breast is pulled upward.
Skin flaps are started by dissecting through the avascular plane between the subcutaneous fat and the breast tissue and are then followed down by dividing the Cooper ligaments. Perform the dissection medially to the sternum, inferiorly to the inframammary fold, laterally to the latissimus dorsi muscle, and superiorly to the clavicle. Include in the specimen the fascia of the pectoralis major muscle, which constitutes the deep margins of the dissection.
In a simple mastectomy, the dissection is stopped before the axillary fat pad is entered. In a modified RM, and according to the surgeon's preference, the dissection may start with the breast and proceed through the axilla or it may start in the axilla and finish in the breast. Avoid excessive hemostasis to preserve the viability of the flaps. Closure is performed without tension or redundant skin after placement of 2 closed suction drains.
Postoperative details: Offer breast reconstruction to patients with breast cancer as an integral part of the multidisciplinary therapeutic approach. It may be immediate or delayed.
Immediate reconstruction is offered to patients with stage 0, I, or II disease. Its psychological, esthetic, and practical advantages outweigh its disadvantages, which mainly include the need for a multidisciplinary team collaborating during the same operative time and the absence of final histology results during surgery. Complications are similar to those of ablative surgery alone. Immediate reconstruction can be combined with any type of ablative surgery, including modified RM. Delayed reconstruction can be performed in all breast cancer stages. It usually follows chemotherapy and RT.
Two reconstructive techniques are used: implant insertion and autologous graft. Implants are made of a silicone shell filled with silicone gel or saline. Although the surgical procedure itself is short and easy, long-term complications are common and include capsular contracture/wrinkling, skin necrosis, and leakage (33%, 8.5%, and 6%, respectively). Because of this high rate of problems, multiple revisions are required, which can add significantly to the total cost. Implants are indicated for small- to medium-sized breasts, in women with poor general health, and in those with short life expectancy. They are contraindicated in patients who require chest wall RT, owing to the high rate of complications.
Although breast reconstruction using autologous tissue is more complex, its advantages far outweigh those of the implant. Esthetically, breast reconstruction results in a warm, soft texture of the breasts with good imitation of natural ptosis. Results are long-lasting, with a low rate of complications and subsequent procedures. Breast reconstruction is probably the only valid option for patients with partial mastectomy and those who require RT.
The most commonly used flaps are the pedicled latissimus dorsi myocutaneous flap, the thoracodorsal artery perforator skin-flat flap, the pedicled or free transverse rectus abdominis myocutaneous flap, the free deep inferior epigastric perforator skin-flat flap, the superior and inferior gluteus myocutaneous flap, and the free superior gluteal artery perforator skin-flat flap. The choice of the donor site and type of procedure depend not only on local conditions at the donor and recipient sites but also on the surgeon's experience.
COMPLICATIONS
- Mastectomy
- Axillary lymph node dissection
- Injury to or thrombosis of the axillary vein
- Seroma formation
- Lymphedema: The reported prevalence rate of lymphedema is approximately 11%, with extremes ranging from 5-30%. Extensive surgery, RT, and advanced age are recognized risk factors for arm edema. Although the risk may decrease with time, it does not completely disappear.
- Impairment of shoulder movements: Depending on whether the patient has received RT to the axilla, the incidence rate varies from 12-15% (RT) and 7-8% (no RT). Symptoms include decreased range of motion of the shoulder, a problem that may be improved with early participation in a physical therapy program.
- Damage to the brachial plexus, with chronic pain and varying degrees of decreased grip strength occurring in up to 15% of patients and lasting for more than a year after surgery
- Chest wall pain
- Chemotherapy
- Cyclophosphamide: Adverse effects may include hemorrhagic cystitis, and amenorrhea.
- Methotrexate: Adverse effects may include liver toxicity, increased toxicity in the presence of pleural effusion, and ascites.
- Fluorouracil: Adverse effects may include mucositis, hand-foot syndrome, and cerebellar ataxia.
- Doxorubicin: Adverse effects may include myocardial dysfunction, alopecia, nausea, vomiting, mucositis, and neutropenia.
- Paclitaxel: Adverse effects may include myelosuppression, peripheral neuropathy (less common if less than 170 mg/m2 is used), hypersensitivity reaction (premedication with steroids, H1- and H2-receptor blockers), cardiac toxicity, alopecia, mucositis, nausea, vomiting, and typhlitis.
- Docetaxel: Adverse effects may include myelosuppression, mucositis, conjunctivitis, edema due to capillary leak syndrome (>80% of patients if not medicated; less than 10% if premedicated with steroids), hypersensitivity reactions, neurotoxicity (less frequent than with paclitaxel), nausea, vomiting, and alopecia.
- Radiation therapy
- Necrosis of the breast soft tissue, prolonged breast edema, rib fracture (low rates, 1-3%)
- Decreased shoulder mobility (low rates, 1-3%)
- Brachial plexopathy with paresthesia and arm pain (low rates, 1-3%)
- Lymphedema
- Angiosarcoma: The 30-year cumulative risk is lower than 1%, with a peak incidence at 6 years.
- Lung cancer: Ipsilateral lung cancer may occur, with an increased risk in persons who smoke.
- Coronary artery disease: The risk has significantly decreased with newer RT techniques.
- Symptomatic pneumonitis: It is relatively infrequent, affecting 3-6% of women treated with RT for breast cancer. Patients present 3-12 months after competition of RT with dry cough, dyspnea, and low-grade fever. Chest radiographs may reveal intersitial infiltrate in the ipsilateral lung, which can evolve to fibrosis.
- Adverse effects of tamoxifen
- Endometrial cancer: This rare complication occurs in 2 of every 1000 women receiving tamoxifen. Most of these cases are detected at an early stage and are easily cured by surgery. Other than yearly gynecologic examinations in asymptomatic women, the National Cancer Institute and the American Society of Obstetricians and Gynecologists recommend prompt evaluation with endometrial biopsy in women on tamoxifen who experience vaginal bleeding.
- Perimenopausal symptoms: Hot flashes and mood changes may occur in women on tamoxifen and occasionally are very severe, thus posing a serious threat to the woman's quality of life. Selective serotonin uptake inhibitors have been used with some success to treat these symptoms.
- Cataracts: These have also been reported in women receiving tamoxifen, justifying a yearly eye examination.
- Adverse effects of trastuzumab
- Cardiac toxicity: In the phase III trial, trastuzumab alone resulted in cardiac dysfunction in 7% (class III/IV, 1.5%) of cases. The prevalence rate increased to 11% when trastuzumab was administered with paclitaxel (class III/IV, 0%). With the combination of anthracycline and cyclophosphamide, the prevalence rate increased to 28% (class III/IV, 6%). Apparently, the combination of an anthracycline and trastuzumab is particularly cardiotoxic. The mechanism of this dysfunction is yet to be defined.
- Fever, chills, nausea, vomiting, and pain with first infusion: These are relatively common but become infrequent with subsequent infusions.
OUTCOME AND PROGNOSIS
- Tumor smaller than 0.5 cm - Approximately 20%
- Tumor 0.5-0.9 cm - Approximately 20%
- Tumor 1-1.9 cm - 33%
- Tumor 2-2.9 cm - 45%
- Tumor 3-3.9 cm - 52%
- Tumor 4-4.9 cm - 60%
- Tumor larger than 5 cm - 70%
The 5-year survival rate based on tumor size and axillary lymph node status is as follows:
New prognostic or predictive factors
FUTURE AND CONTROVERSIES
- Which selective estrogen receptor modulators will be most effective in adjuvant and prevention settings?
- What is the role of selective estrogen receptor down-regulators?
- Which aromatase inhibitor is the best adjuvant therapy for breast cancer and if these agents are equivalent in terms of efficacy are they different in terms of toxicity?
- Using modern biotechnology (ie, genomics, proteomics), will it be possible to predict the responsiveness to any of these hormonal therapies?
- Will prolonged treatment of aromatase inhibitors beyond 5 years be superior to 5 years of therapy (more than 5 years of tamoxifen is not superior to 5 year) ?
- What is the best sequence to provide these patients with the longest survival possible?
- What are the patterns of resistance to HT, and are there methods available to overcome them?
- Will long-term use of aromatase inhibitors result in an increased prevalence of skeletal or other complications?
Several concepts are being explored in this area.
2 comments:
Cured of TNBC!
I want to take this time out as a cancer survivor to encourage women out there still suffering from this with my story on how i got a cure. The sad news about it is that i was diagnosed on my 36th birthday in 2008 and with stage 3 TNBC which after i made research was a very aggressive form of cancer at that point i decided and told myself i was going to die and that the end has finally come. All my life i never thought of having breast cancer because i was very active and i worked out at the gym several times every week and my diet was okay. In my search for a cure after 6 years of diagnosis and even after chemo which i did twice spending thousands of dollars but to no avail, until a church member told me all about Dr Aleta a herbal doctor that specializes in treating TNBC, who could help me with a permanent cure, i doubted this at first but i later gave it a try following her methods and instructions. It took 3 months and after it all i felt normal but still went for diagnosis and i was clean today i am proud to say i am a cancer survivor no nodes and i am totally free the new diagnosis confirmed it. Do not die in silence or ignorance because of breast cancer just simply reach her on aletedwin@gmail.com don't be shy just speak to her today.
Cathepsin D is a lysosomal aspartyl protease composed of a protein dimer of disulfide-linked heavy and light chains, both produced from a single protein precursor. It is an estrogen-regulated protein associated with tissue breakdown. cathepsin d
Post a Comment