28

Click here to load reader

Systemic Treatment for Metastatic Breast Cancer

  • Upload
    jimdio

  • View
    33

  • Download
    1

Embed Size (px)

DESCRIPTION

medicine

Citation preview

Page 1: Systemic Treatment for Metastatic Breast Cancer

Systemic treatment for metastatic breast cancer: General principlesAuthorDaniel F Hayes, MDSection EditorJulie R Gralow, MDDeputy EditorDon S Dizon, MD, FACPDisclosuresAll topics are updated as new evidence becomes available and our peer review process is complete.Literature review current through: Nov 2012. | This topic last updated: Δεκ 6, 2012.

INTRODUCTION — Although metastatic breast cancer is unlikely to be cured, meaningful improvements in survival have been seen, coincident with the introduction of newer systemic therapies [1-3]. Median overall survival approaches two years, with a range from a few months to many years [4].

The selection of a therapeutic strategy depends upon both tumor biology and clinical factors, with the goal being a tailored approach. Although a subset of patients with oligometastatic disease may benefit from an intensified locoregional approach, most patients with metastatic breast cancer receive systemic medical therapy, consisting of chemotherapy, endocrine therapy, and/or biologic therapies, and supportive care measures [5,6].

General principles of management of metastatic breast cancer are presented here. Details of single agent and combination chemotherapy, endocrine therapy, biologic therapy, and how to select among them, as well as locoregional approaches, osteoclast inhibitors (bisphosphonates and receptor activator of nuclear factor kappa-B [RANK] ligand inhibitors), and supportive care, are discussed separately.

(See "Systemic treatment for metastatic breast cancer: Endocrine therapy".) (See "Systemic treatment for metastatic breast cancer: Single agent

chemotherapy".) (See "Systemic treatment for metastatic breast cancer: Combination

chemotherapy".) (See "Systemic treatment for metastatic breast cancer: Biologic therapy".) (See "Systemic treatment for metastatic breast cancer: Selection of

chemotherapy regimen".) (See "Metastatic breast cancer: Local treatment".) (See "Treatment of metastatic breast cancer in older women".) (See "Osteoclast inhibition in the management of bone metastases from breast

cancer".)

THERAPEUTIC GOALS — The primary goals of systemic treatment for metastatic breast cancer are prolongation of survival, alleviation of symptoms, and maintenance or improvement in quality of life, despite the toxicity associated with treatment [7-9]. The median survival for metastatic breast cancer is 18 to 24 months, though this varies widely based on subtype of tumor, sites of metastatic involvement, and burden of metastatic disease, and some patients experience long-term survival [1-4,10].

Page 2: Systemic Treatment for Metastatic Breast Cancer

No prospective randomized clinical trials have demonstrated that systemic therapy prolongs survival compared to best supportive care alone [7]. However, median survival for patients with metastatic breast cancer appears to have improved over time, a trend which has been attributed to the availability of new, more effective agents, including taxanes, aromatase inhibitors, and trastuzumab [1,2,11-13]. As an example, patients from the British Columbia Breast Cancer Outcomes Database who were diagnosed between 1997 and 2001 had better two-year overall survival than patients diagnosed between 1991 and 1995 (45 versus 34 percent) [1].

The optimal measure of therapeutic efficacy is debated. Overall survival is the gold standard for comparing therapies, but it requires prolonged follow-up and may be diluted by the effects of subsequent treatment. However, no other endpoint, including progression-free survival, time to tumor progression, or objective response rate, has been shown to be a good surrogate for overall survival [14]. Comparisons of objective response rates are often used to determine relative treatment efficacy, but high response rates do not necessarily translate into clinically meaningful increases in survival [14-16]. In addition, symptom relief without measurable disease response and achievement of stable disease as compared to disease progression may be clinically important [17].

TUMOR BIOLOGY AND RISK ASSESSMENT — Biologic markers, such as hormone receptor status, human epidermal growth factor receptor 2 (HER2) overexpression, and tumor burden, have both prognostic and predictive value and are important factors in selecting appropriate treatment.

Disease assessment — Complete evaluation for the extent of metastatic disease includes confirmatory biopsy of suspected lesions and reassessment of molecular markers, particularly estrogen receptor (ER), progesterone receptor (PR), and HER2 overexpression. This is especially important if the primary cancer was deemed negative for ER, PR, and or HER2, since a conversion to positive would dramatically change therapy.

There is a possible discordance of these markers between primary and metastatic disease [18-22]. As an example, in a pooled analysis of two prospective studies, the rates of discordance in ER, PR, and HER2 between the primary and recurrent disease were 13, 28, and 5 percent, respectively [19]. In this study, the number needed to biopsy to alter immediate patient management was 6.3. However, it is not clear that verifying receptor status in new metastases and the resultant management changes will ultimately lead to improvements in patient quality of life and survival.

Prognosis — Clinical factors that predict the rate of progression and survival include the interval between initial therapy and relapse, the number of metastatic sites, the presence or absence of visceral involvement, and biologic markers. As examples:

Relapse-free interval of ≥2 years is more favorable than a shorter time to relapse [23-26].

Patients with metastases involving the chest wall, bones, or lymph nodes may have prolonged progression-free survival, while those with hepatic and/or lymphangitic pulmonary disease have shorter progression-free and overall survival [23,24,27]. Patients with lymphangitic lung metastases, bone marrow

Page 3: Systemic Treatment for Metastatic Breast Cancer

replacement, carcinomatous meningitis, or significant liver metastases are described as having visceral crisis [28]. Patients without visceral crisis but with poor prognostic features represent an intermediate phenotype.

Patients with hormone receptor positivity generally have a more favorable prognosis, and patients with ER- and PR-positive tumors have significantly longer survival than single hormone receptor-positive tumors (ER+/PR- or ER-/PR+) [29]. Patients with either HER2 overexpression or triple (ER, PR, HER2)-negative metastatic breast cancer have a shorter median survival [25,30,31]. The prognosis associated with particular molecular phenotypes is discussed separately. (See "Molecular intrinsic subtypes of breast cancer".)

Other adverse prognostic features include weight loss, poor performance status (table 1), and elevated serum lactic dehydrogenase [23,32]. Age less than 35 years old is a poor prognostic factor for women with early stage breast cancer, but the effect of age on survival after recurrence is not established [25,33]. Similarly, the stage at diagnosis does not reliably indicate prognosis after relapse.

Prediction of response — Hormone receptor status and HER2 overexpression are the most important predictors of treatment response in patients with metastatic breast cancer.

Hormone receptors — Hormone receptor status is the most important predictive factor for response to hormone therapy [34,35]. The likelihood of response is 70 percent in patients with tumors that express both ER and PR, 40 percent in tumors that are ER-positive/PR-negative or ER-negative/PR-positive, and less than 10 percent in ER-negative/PR-negative tumors [36,37]. The molecular basis by which ER status predicts response to endocrine therapy is discussed separately. (See "Hormone receptors in breast cancer: Clinical utility and guideline recommendations to improve test accuracy".)

Additional features that predict responsiveness to hormone therapy in patients with metastatic breast cancer include a long relapse-free interval, isolated bone and soft tissue involvement, and prior response to endocrine therapy [38,39].

There are some data suggesting that patients with ER-positive tumors are less likely to respond to cytotoxic chemotherapy than those with ER-negative tumors [35,40-42].

HER2-overexpression — Overexpression of HER2 predicts response to therapies that target HER2, such as the humanized monoclonal antibody trastuzumab and the small molecule tyrosine kinase inhibitor lapatinib. These drugs are only effective for tumors that have high levels of HER2 expression (3+ by immunohistochemistry) or gene amplification (as detected by fluorescence in situ hybridization [FISH]).

Some evidence suggests that tumors with HER2 overexpression are less responsive to endocrine therapy, although the available data are not sufficient to use HER2 status to predict resistance to endocrine therapy [43,44].

Details of HER2 testing and the role of HER2-status in predicting the response to HER2-directed therapy, chemotherapy, and endocrine therapy are discussed separately. (See "HER2 and predicting response to therapy in breast cancer".)

Page 4: Systemic Treatment for Metastatic Breast Cancer

Chemotherapy response — Consistent predictors of poor chemotherapy response are progression with prior chemotherapy for advanced disease, relapse within 12 months of completing adjuvant chemotherapy, poor performance status, and multiple disease sites, especially visceral involvement [23,24,32,45-52].

Markers of increased cellular proliferation, such as high S-phase fraction by flow cytometry, increased uptake of radiolabeled thymidine, and immunohistochemical staining for the proliferation antigen Ki67, are all associated with higher chemotherapy response rates [53-55]. In contrast, breast cancers that overexpress p-glycoprotein (gp170), a drug efflux pump that mediates multidrug resistance (MDR), or have a mutated p53 gene may be less likely to respond to chemotherapy [56-59]. At present, none of these markers should be used to make clinical decisions.

Another possible approach is the use of chemotherapy sensitivity and resistance assays [60-62]. However, the clinical utility of these assays remains unclear [56,63,64]. The detection and monitoring of circulating tumor cells during treatment for metastatic breast cancer is discussed below. (See 'Circulating tumor cells' below.)

TREATMENT SELECTION — Key points described above are important to keep in mind when selecting treatment for metastatic breast cancer. These include:

The aim of treatment is to palliate symptoms, prolong survival, and maintain quality of life.

Patients with visceral metastases and a short disease-free interval generally have an aggressive phenotype, while patients with soft tissue and bone metastases have a more indolent phenotype.

Hormone receptor status and human epidermal growth factor receptor 2 (HER2) overexpression are important in estimating prognosis and the likelihood of response to therapy.

Hormone receptor (estrogen receptor [ER] and/or progesterone receptor [PR]) status is the major determining factor for response to endocrine therapy.

Likewise, HER2 overexpression is required for response to HER2-directed therapies.

Based upon these principles, selection of treatment can be individualized. Endocrine therapy is best used for patients with hormone receptor-positive breast cancer and not for patients with hormone-negative breast cancer. HER2-directed therapy is only appropriate for patients with tumors that overexpress HER2. Lastly, chemotherapy is indicated for hormone-insensitive metastatic breast cancer (ie, patients with hormone-receptor-negative breast cancer and those with hormone-receptor-positive breast cancer who have become resistant to endocrine therapy). It is less clear is when to use endocrine therapy versus chemotherapy as initial treatment for patients with hormone receptor-positive metastatic breast cancer. Likewise, it is less obvious when to use single agent versus combination chemotherapy, how to best incorporate biologic therapies, and whether combined modalities are of benefit.

Endocrine therapy versus chemotherapy — It is commonly thought that chemotherapy results in higher response rates and more rapid responses than endocrine therapy, and is often used as the initial treatment for patients with hormone receptor-positive

Page 5: Systemic Treatment for Metastatic Breast Cancer

metastatic breast cancer with a poor prognosis, especially those with visceral metastases. A meta-analysis that included eight small randomized trials, all published prior to 1995, compared the response rates for chemotherapy alone with those of endocrine therapy alone [65]. The pooled estimate of response rates showed an advantage for chemotherapy over endocrine therapy (relative risk 1.25, 95% CI 1.01 to 1.54), although the two largest trials had opposite findings [66,67]. No significant difference was seen in overall survival (hazard ratio, HR 0.94, 95% CI 0.79 to 1.12), and on subset analysis, there was no obvious trend to suggest an effect of age, menopausal status, or pattern of metastatic disease on the efficacy of either therapy. There was minimal and contrasting information on quality of life and toxicity.

A major limitation to these findings is that most patients in these trials had tumors of unknown hormone receptor status, since the predictive value of hormone receptor status on response to endocrine therapy was not yet appreciated. Nevertheless, chemotherapy remains the preferred modality for initial treatment of patients with rapidly progressive symptomatic disease or visceral crisis (end-organ dysfunction), given the higher likelihood of achieving a response with chemotherapy.

No survival benefit has been seen when chemotherapy and endocrine therapy were combined.

Sequential single agents versus combination chemotherapy — When compared in a meta-analysis published in 2005, combination chemotherapy was associated with significantly higher overall response rates (odds ratio [OR] 1.28, 95% CI 1.15-1.42) and improved overall survival (hazard ratio [HR] for death 0.88, 95% CI 0.83-0.93), and a significantly lower rate of disease progression (overall hazard ratio HR 0.78, 95% CI 0.73-0.83) [68]. However, this modest survival improvement came with the cost of significantly worse toxicity, including leukopenia, nausea, and vomiting.

The findings of this meta-analysis are limited in that modern chemotherapy agents were not well represented in the included trials and that the rate of cross-over was not known.

Clinical trials with newer agents, such as capecitabine, docetaxel, vinorelbine, and gemcitabine, have either found no difference in overall survival between combination and single agent chemotherapy, or a modest improvement of a few months, and again, worse toxicity [69-71].

Most trials compare single agent therapy to a combination of two drugs, rather than comparing the combination to sequential use of the two single agents. The modest survival benefits favoring combination chemotherapy may not have been seen if crossover to the experimental agent had been allowed. Several trials that directly compared combination versus sequential therapy failed to show a survival benefit for initial combination therapy [72-75].

As an example, Eastern Cooperative Oncology Group (ECOG) trial E1193 randomly assigned 739 patients with metastatic breast cancer to doxorubicin alone, paclitaxel alone, or the combination [72]. Patients receiving single agent therapy crossed over to the alternative agent at the time of progression. Although initial combination therapy with doxorubicin and paclitaxel, compared to either agent alone, was associated with a significantly higher response rate (47 versus 36 and 34 percent)

Page 6: Systemic Treatment for Metastatic Breast Cancer

and time to treatment failure (8 versus 5.8 and 6 months), median overall survival was similar for all three (22, 19, and 22 months, respectively).

Without an appreciable survival advantage with combination chemotherapy, sequential therapy with single agents is often preferred. Toxicity is less with single agent therapy and quality of life appears better [76-78]. For patients in need of quick symptom control or those with rapidly progressive disease or visceral crisis, however, combination therapy may be a more appropriate first-line choice because of the greater likelihood of an objective response in a shorter time.

Adding more agents to combination regimens has not led any further improvements in time to progression or overall survival [79]. In fact, a review of all randomized trials comparing chemotherapy regimens for metastatic breast cancer that were reported between 2000 and 2007 found little evidence of major survival differences among many common chemotherapy regimens [80].

Combining treatment modalities — In theory, combining chemotherapy, biologic therapy, and/or endocrine therapy might have additive efficacy, but it might also lead to increased toxicity. Clinical trials have failed to show a survival advantage for the concurrent administration of chemotherapy and endocrine therapy over either single modality [7,67,81].

However, HER2-directed therapy, namely, trastuzumab and lapatinib, has been successfully combined as individual agents with chemotherapy and endocrine therapy, and with each other.

Biologic therapy — At present, the molecularly targeted agents (or biologic therapy) that have proven efficacy in metastatic breast cancer are the HER2-directed agents, trastuzumab and lapatinib, and bevacizumab. The use of these biologic agents in metastatic breast cancer is discussed in detail separately. (See "Systemic treatment for metastatic breast cancer: Biologic therapy".)

Osteoclast inhibitors — Patients with bone metastases should be treated with osteoclast inhibitors (bisphosphonates or RANK ligand inhibition) as these agents have been shown to reduce the risk of skeletal related events, such as fractures, the need for surgery or radiation to bone, spinal cord compression, and hypercalcemia of malignancy. (See "Osteoclast inhibition in the management of bone metastases from breast cancer".)

TREATMENT ALGORITHMS — The issues discussed above can be summarized and clarified into treatment recommendations for patients with metastatic breast cancer according to the hormone receptor and human epidermal growth factor receptor 2 (HER2) status of their tumors.

Hormone receptor-positive HER2-negative patients — The evidence, despite limitations described above, suggests that chemotherapy has higher response rates than endocrine therapy. Therefore, patients with rapidly progressive, symptomatic disease or visceral metastases with end-organ dysfunction may be best treated with first-line chemotherapy. After chemotherapy response stabilizes (usually four to six months), a switch to maintenance endocrine therapy is a commonly employed

Page 7: Systemic Treatment for Metastatic Breast Cancer

strategy, which can reduce the treatment side effects without compromising overall survival [7,82,83].

Given its generally favorable toxicity profile, first-line endocrine therapy may be more appropriate for patients with asymptomatic or minimally symptomatic disease and those without visceral involvement. If the disease progresses rapidly (within a few months) following initiation of first-line endocrine therapy, chemotherapy is generally recommended as a second-line rather than switching to another endocrine strategy. If time to progression on first-line endocrine therapy is prolonged (greater than six months), then a switch to second-line endocrine therapy at progression is reasonable.

Selection of endocrine therapy is discussed separately. (See "Systemic treatment for metastatic breast cancer: Endocrine therapy".)

Selection of chemotherapy regimen is discussed separately. (See "Systemic treatment for metastatic breast cancer: Selection of chemotherapy regimen".)

Hormone receptor-positive HER2-positive patients — Therapeutic options for these patients include chemotherapy, endocrine therapy, and HER2-directed therapy. HER2 directed therapy has demonstrated improved survival for patients with tumors that overexpress HER2 and thus should be part of first-line therapy for these patients.

Whether it is better to use HER2-directed therapy combined with chemotherapy versus endocrine therapy as first-line treatment is unclear. HER2-directed therapy combined with chemotherapy or endocrine therapy are discussed separately.

(See "Systemic treatment for metastatic breast cancer: Biologic therapy", section on 'HER2-directed therapy'.)

(See "Systemic treatment for metastatic breast cancer: Endocrine therapy".)

Hormone receptor-negative HER2-negative patients — Many patients with triple- (ER-, PR-, HER2-) negative breast cancer have a particularly aggressive subtype, and first-line chemotherapy is recommended. Whether chemotherapy agents are given in combination or sequentially should be determined based on symptoms and location and burden of disease, as well as patient-related factors (ie, preferences, goals, and overall health).

Triple-negative breast cancer is discussed in detail separately. (See "Epidemiology, risk factors and the clinical approach to ER/PR negative, HER2-negative (Triple-negative) breast cancer".)

Selection of chemotherapy regimen is discussed separately. (See "Systemic treatment for metastatic breast cancer: Selection of chemotherapy regimen".)

Hormone receptor-negative HER2-positive patients — The combination of HER2-directed therapy and chemotherapy is recommended for these patients.

Regimens combining HER2-directed therapy with chemotherapy are discussed separately. (See "Systemic treatment for metastatic breast cancer: Biologic therapy", section on 'HER2-directed therapy'.)

Page 8: Systemic Treatment for Metastatic Breast Cancer

MONITORING THERAPY — Careful assessment of the response to therapy will assist in decisions for duration of treatment and in selection of subsequent treatments. However, the best approach for monitoring patients with metastatic breast cancer is not well established.

History and examination — If symptom palliation is the main objective, clinical history alone may suffice to determine the success of therapy. Physical examination may allow response quantitation if disease is easily accessible (eg, chest wall nodules, palpable lymphadenopathy). With a dramatic reduction in symptoms that were clearly disease-related or obvious shrinkage of palpable lesions, serum markers and radiographic tests are likely to be irrelevant.

However, many patients have more subtle disease signs or symptoms that may be confused with treatment toxicity or other nonmalignant conditions. In addition, disease is not measurable by physical examination in nearly half of patients with metastatic breast cancer. In these patients, serial changes in tumor markers or radiographic studies are essential in establishing the response to therapy.

Tumor markers — Serial assay of serum tumor markers (eg, CA15-3 and CA27.29, both products of the MUC-1 gene, and CEA) can aid in response assessment, particularly if disease sites are not assessable by usual criteria [43,84]. Judicious use of serial tumor marker measurements may decrease the need for periodic radiographic evaluation [85].

Guidelines from the American Society of Clinical Oncology (ASCO) expert panel suggest that it is reasonable to evaluate one of the MUC-1 assays and CEA initially in patients with metastatic disease [43]. If the MUC-1 assay is elevated, there is no role for monitoring CEA, but if not, then serial CEA levels may be useful.

Elevated tumor markers may occasionally be spurious. Up to 20 percent of patients successfully treated with systemic therapy may experience a transient increase (marker "flare") during the first one or two months after treatment initiation, presumably due to release of antigen by cytolysis [86,87]. Patients with abnormal liver function may also have falsely elevated marker levels because they are cleared by the liver [86]. CA 15-3 levels may be aberrantly elevated in patients with vitamin B12 deficiency and megaloblastic anemia, as well as in patients with thalassemia or sickle cell disease [88-90].

Radiographic studies — Serial plain radiographs, computed tomography (CT) scan, or magnetic resonance imaging (MRI) can permit assessment of tumor response. Periodic scintigraphic bone scans, while helpful, may also be misleading. Technetium (Tc99) phosphonate accumulates in areas of osteoblastic activity rather than in cancer cells. In a patient experiencing a response to therapy, a "scintigraphic healing flare" may appear as early as two months, and persist for as long as 12 months after initiating therapy [91,92].

Integrated positron emission tomography (PET)/computed tomography (CT) is popular as a whole-body examination in monitoring response to therapy in metastatic breast cancer, as it has demonstrated high sensitivity and specificity in detecting metastatic disease and can reliably assess response to therapy [93-95]. There is also

Page 9: Systemic Treatment for Metastatic Breast Cancer

some evidence that metabolic changes in bone metastases in response to systemic therapy (ie, a change in standardized uptake value [SUV]) can predict response duration or time to progression [96-98]. However, many integrated PET/CT scanners in clinical use provide a limited CT scan primarily for orientational purposes (determining where the PET abnormality is) and not a higher resolution, fine cut, contrast-enhanced CT scan. This should be kept in mind when evaluating response between modalities (standard CT and PET/CT).

Circulating tumor cells — Detection of circulating tumor cells (CTCs) in blood samples of patients with metastatic breast cancer (≥five CTCs) has been shown to be a predictor of progression-free survival (PFS) and overall survival [99-105]. Immunological and RNA-based methods are used to detect CTCs in breast cancer. Although multiple commercially available methods for isolating CTCs exist, the CellSearch system (Veridex Corporation, Warren, NJ) is the only system approved in the US for clinical use to detect the presence of CTCs in patients with breast cancer. In a prospective trial of 177 patients who were beginning a new therapy for metastatic breast cancer [100], elevated CTCs at baseline (defined as ≥5 CTCs per 7.5 mL of blood) compared with the finding of fewer or no detectable CTCs predicted a significantly shorter PFS (3 versus 7 months) and overall survival (10 versus 22 months). Patients with elevated CTCs at the first follow-up visit (within three to five weeks of initiating therapy) also had a worse PFS (2 versus 7 months) and overall survival (8 versus greater than 18 months), whereas those with a decrease in the number of CTCs from baseline had improved PFS and overall survival. Subsequent analyses from this trial and others have suggested that elevated levels of CTCs at any time point during treatment are associated with tumor progression and that CTC levels may reliably estimate disease progression earlier than imaging studies [102,105,106].

An ASCO expert panel on tumor markers in breast cancer, when convened in 2007, concluded that the data were insufficient and needed further validation prior to recommending routine measurement of CTCs in patients with metastatic breast cancer [43]. A prospective randomized trial is underway to test the clinical utility of changing therapy for patients with metastatic breast cancer who have not cleared CTCs after only one cycle of a new, first-line therapy [107].

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, “The Basics” and “Beyond the Basics.” The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on “patient info” and the keyword(s) of interest.)

Page 10: Systemic Treatment for Metastatic Breast Cancer

Beyond the Basics topics (see "Patient information: Treatment of metastatic breast cancer (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS — The goals of systemic treatment for metastatic breast cancer are prolongation of survival, alleviation of symptoms, and maintenance or improvement in quality of life.

Hormone receptor status, human epidermal growth factor receptor 2 (HER2) overexpression, tumor burden, and disease-free interval have prognostic and predictive value and are important determinants in selecting appropriate treatment.

Given the importance of hormone receptor and HER2 status in selecting treatment, hormone receptor and HER2 testing should be repeated upon diagnosis of metastatic breast cancer in case there is discordance in expression between the primary site and metastases, especially if the primary cancer was deemed negative, since a conversion to positive would dramatically change therapy.

For patients with hormone-positive metastatic breast cancer with rapidly progressive, symptomatic disease or visceral metastases with end-organ dysfunction, we recommend first-line treatment with chemotherapy rather than endocrine therapy (Grade 1B). All other patients with hormone-positive metastatic breast cancer should be treated first with endocrine therapy.

We suggest sequential single agent chemotherapy for most patients with metastatic breast cancer who require chemotherapy (Grade 2B). Combination chemotherapy may be preferable for select patients with rapidly progressive disease, visceral crisis, or if quick symptom control is desired.

We recommend that patients with HER2-positive breast cancer, regardless of their hormone receptor status, receive HER2-directed therapy as first-line treatment (Grade 1A). (See "Systemic treatment for metastatic breast cancer: Biologic therapy", section on 'HER2-directed therapy'.)

Careful assessment of the response to therapy will assist in decisions for treatment continuation and in selection of subsequent treatments. Tools that are potentially useful to monitor treatment response include history and physical examination, radiographic imaging, and assay of serum tumor markers and/or circulating tumor cells (CTCs). (See 'Monitoring therapy' above.)

Use of UpToDate is subject to the Subscription and License Agreement.

REFERENCES1. Chia SK, Speers CH, D'yachkova Y, et al. The impact of new

chemotherapeutic and hormone agents on survival in a population-based cohort of women with metastatic breast cancer. Cancer 2007; 110:973.

2. Gennari A, Conte P, Rosso R, et al. Survival of metastatic breast carcinoma patients over a 20-year period: a retrospective analysis based on individual patient data from six consecutive studies. Cancer 2005; 104:1742.

3. Dafni U, Grimani I, Xyrafas A, et al. Fifteen-year trends in metastatic breast cancer survival in Greece. Breast Cancer Res Treat 2010; 119:621.

Page 11: Systemic Treatment for Metastatic Breast Cancer

4. Greenberg PA, Hortobagyi GN, Smith TL, et al. Long-term follow-up of patients with complete remission following combination chemotherapy for metastatic breast cancer. J Clin Oncol 1996; 14:2197.

5. Pagani O, Senkus E, Wood W, et al. International guidelines for management of metastatic breast cancer: can metastatic breast cancer be cured? J Natl Cancer Inst 2010; 102:456.

6. Beslija S, Bonneterre J, Burstein HJ, et al. Third consensus on medical treatment of metastatic breast cancer. Ann Oncol 2009; 20:1771.

7. Stockler M, Wilcken NR, Ghersi D, Simes RJ. Systematic reviews of chemotherapy and endocrine therapy in metastatic breast cancer. Cancer Treat Rev 2000; 26:151.

8. Osoba D. Health-related quality of life as a treatment endpoint in metastatic breast cancer. Can J Oncol 1995; 5 Suppl 1:47.

9. Geels P, Eisenhauer E, Bezjak A, et al. Palliative effect of chemotherapy: objective tumor response is associated with symptom improvement in patients with metastatic breast cancer. J Clin Oncol 2000; 18:2395.

10. Kiely BE, Soon YY, Tattersall MH, Stockler MR. How long have I got? Estimating typical, best-case, and worst-case scenarios for patients starting first-line chemotherapy for metastatic breast cancer: a systematic review of recent randomized trials. J Clin Oncol 2011; 29:456.

11. Giordano SH, Buzdar AU, Smith TL, et al. Is breast cancer survival improving? Cancer 2004; 100:44.

12. Mauri D, Polyzos NP, Salanti G, et al. Multiple-treatments meta-analysis of chemotherapy and targeted therapies in advanced breast cancer. J Natl Cancer Inst 2008; 100:1780.

13. Dawood S, Broglio K, Buzdar AU, et al. Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review. J Clin Oncol 2010; 28:92.

14. Burzykowski T, Buyse M, Piccart-Gebhart MJ, et al. Evaluation of tumor response, disease control, progression-free survival, and time to progression as potential surrogate end points in metastatic breast cancer. J Clin Oncol 2008; 26:1987.

15. Ahmann DL, Schaid DJ, Bisel HF, et al. The effect on survival of initial chemotherapy in advanced breast cancer: polychemotherapy versus single drug. J Clin Oncol 1987; 5:1928.

16. Bruzzi P, Del Mastro L, Sormani MP, et al. Objective response to chemotherapy as a potential surrogate end point of survival in metastatic breast cancer patients. J Clin Oncol 2005; 23:5117.

17. Robertson JF, Howell A, Buzdar A, et al. Static disease on anastrozole provides similar benefit as objective response in patients with advanced breast cancer. Breast Cancer Res Treat 1999; 58:157.

18. Hull DF 3rd, Clark GM, Osborne CK, et al. Multiple estrogen receptor assays in human breast cancer. Cancer Res 1983; 43:413.

19. Amir E, Clemons M, Freedman OC, et al. Tissue confirmation of disease recurrence in patients with breast cancer: Pooled analysis of two large prospective studies (abstract #1007). J Clin Oncol 2010; 28:15s.

20. Simmons C, Miller N, Geddie W, et al. Does confirmatory tumor biopsy alter the management of breast cancer patients with distant metastases? Ann Oncol 2009; 20:1499.

Page 12: Systemic Treatment for Metastatic Breast Cancer

21. Amir E, Clemons M. Should a biopsy be recommended to confirm metastatic disease in women with breast cancer? Lancet Oncol 2009; 10:933.

22. Khasraw M, Brogi E, Seidman AD. The need to examine metastatic tissue at the time of progression of breast cancer: is re-biopsy a necessity or a luxury? Curr Oncol Rep 2011; 13:17.

23. Swenerton KD, Legha SS, Smith T, et al. Prognostic factors in metastatic breast cancer treated with combination chemotherapy. Cancer Res 1979; 39:1552.

24. Hortobagyi GN, Smith TL, Legha SS, et al. Multivariate analysis of prognostic factors in metastatic breast cancer. J Clin Oncol 1983; 1:776.

25. Clark GM, Sledge GW Jr, Osborne CK, McGuire WL. Survival from first recurrence: relative importance of prognostic factors in 1,015 breast cancer patients. J Clin Oncol 1987; 5:55.

26. Harris JR, Hellman S. Observations on survival curve analysis with particular reference to breast cancer treatment. Cancer 1986; 57:925.

27. Robertson JF, Dixon AR, Nicholson RI, et al. Confirmation of a prognostic index for patients with metastatic breast cancer treated by endocrine therapy. Breast Cancer Res Treat 1992; 22:221.

28. Barrios CH, Sampaio C, Vinholes J, Caponero R. What is the role of chemotherapy in estrogen receptor-positive, advanced breast cancer? Ann Oncol 2009; 20:1157.

29. Stuart-Harris R, Shadbolt B, Palmqvist C, Chaudri Ross HA. The prognostic significance of single hormone receptor positive metastatic breast cancer: an analysis of three randomised phase III trials of aromatase inhibitors. Breast 2009; 18:351.

30. Emi Y, Kitamura K, Shikada Y, et al. Metastatic breast cancer with HER2/neu-positive cells tends to have a morbid prognosis. Surgery 2002; 131:S217.

31. Ismail-Khan R, Bui MM. A review of triple-negative breast cancer. Cancer Control 2010; 17:173.

32. Yamamoto N, Watanabe T, Katsumata N, et al. Construction and validation of a practical prognostic index for patients with metastatic breast cancer. J Clin Oncol 1998; 16:2401.

33. Leivonen MK, Kalima TV. Prognostic factors associated with survival after breast cancer recurrence. Acta Oncol 1991; 30:583.

34. Clark GM, Osborne CK, McGuire WL. Correlations between estrogen receptor, progesterone receptor, and patient characteristics in human breast cancer. J Clin Oncol 1984; 2:1102.

35. Rubens RD, Hayward JL. Estrogen receptors and response to endocrine therapy and cytotoxic chemotherapy in advanced breast cancer. Cancer 1980; 46:2922.

36. Osborne CK, Yochmowitz MG, Knight WA 3rd, McGuire WL. The value of estrogen and progesterone receptors in the treatment of breast cancer. Cancer 1980; 46:2884.

37. Bloom ND, Tobin EH, Schreibman B, Degenshein GA. The role of progesterone receptors in the management of advanced breast cancer. Cancer 1980; 45:2992.

38. Santen RJ, Manni A, Harvey H, Redmond C. Endocrine treatment of breast cancer in women. Endocr Rev 1990; 11:221.

Page 13: Systemic Treatment for Metastatic Breast Cancer

39. Epstein RJ. The clinical biology of hormone-responsive breast cancer. Cancer Treat Rev 1988; 15:33.

40. Levine RM, Lippman ME. Relationship between estrogen-receptor proteins and response to chemotherapy in breast cancer. Cancer Treat Rep 1984; 68:573.

41. Lippman ME, Allegra JC, Thompson EB, et al. The relation between estrogen receptors and response rate to cytotoxic chemotherapy in metastatic breast cancer. N Engl J Med 1978; 298:1223.

42. Kiang DT, Frenning DH, Goldman AI, et al. Estrogen receptors and responses to chemotherapy and hormonal therapy in advanced breast cancer. N Engl J Med 1978; 299:1330.

43. Harris L, Fritsche H, Mennel R, et al. American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol 2007; 25:5287.

44. De Laurentiis M, Arpino G, Massarelli E, et al. A meta-analysis on the interaction between HER-2 expression and response to endocrine treatment in advanced breast cancer. Clin Cancer Res 2005; 11:4741.

45. Ahmann DL, Schaid DJ, Ingle JN, et al. A randomized trial of cyclophosphamide, doxorubicin, and prednisone versus cyclophosphamide, 5-fluorouracil, and prednisone in patients with metastatic breast cancer. Am J Clin Oncol 1991; 14:179.

46. Valagussa P, Tancini G, Bonadonna G. Salvage treatment of patients suffering relapse after adjuvant CMF chemotherapy. Cancer 1986; 58:1411.

47. Perez JE, Machiavelli M, Leone BA, et al. Bone-only versus visceral-only metastatic pattern in breast cancer: analysis of 150 patients. A GOCS study. Grupo Oncológico Cooperativo del Sur. Am J Clin Oncol 1990; 13:294.

48. Falkson G, Gelman R, Falkson CI, et al. Factors predicting for response, time to treatment failure, and survival in women with metastatic breast cancer treated with DAVTH: a prospective Eastern Cooperative Oncology Group study. J Clin Oncol 1991; 9:2153.

49. Rabinovich M, Vallejo C, Bianco A, et al. Development and validation of prognostic models in metastatic breast cancer: a GOCS study. Oncology 1992; 49:188.

50. Pronzato P, Bertelli G, Gardin G, et al. Analysis of time to response to chemotherapy in 316 metastatic breast cancer patients. Oncology 1993; 50:460.

51. Aisner J, Cirrincione C, Perloff M, et al. Combination chemotherapy for metastatic or recurrent carcinoma of the breast--a randomized phase III trial comparing CAF versus VATH versus VATH alternating with CMFVP: Cancer and Leukemia Group B Study 8281. J Clin Oncol 1995; 13:1443.

52. Rahman ZU, Frye DK, Buzdar AU, et al. Impact of selection process on response rate and long-term survival of potential high-dose chemotherapy candidates treated with standard-dose doxorubicin-containing chemotherapy in patients with metastatic breast cancer. J Clin Oncol 1997; 15:3171.

53. Rozan S, Vincent-Salomon A, Zafrani B, et al. No significant predictive value of c-erbB-2 or p53 expression regarding sensitivity to primary chemotherapy or radiotherapy in breast cancer. Int J Cancer 1998; 79:27.

54. Hatschek T, Carstensen J, Fagerberg G, et al. Influence of S-phase fraction on metastatic pattern and post-recurrence survival in a randomized mammography screening trial. Breast Cancer Res Treat 1989; 14:321.

Page 14: Systemic Treatment for Metastatic Breast Cancer

55. Amadori D, Volpi A, Maltoni R, et al. Cell proliferation as a predictor of response to chemotherapy in metastatic breast cancer: a prospective study. Breast Cancer Res Treat 1997; 43:7.

56. Mechetner E, Kyshtoobayeva A, Zonis S, et al. Levels of multidrug resistance (MDR1) P-glycoprotein expression by human breast cancer correlate with in vitro resistance to taxol and doxorubicin. Clin Cancer Res 1998; 4:389.

57. Trock BJ, Leonessa F, Clarke R. Multidrug resistance in breast cancer: a meta- analysis of MDR1/gp170 expression and its possible functional significance. J Natl Cancer Inst 1997; 89:917.

58. Clahsen PC, van de Velde CJ, Duval C, et al. p53 protein accumulation and response to adjuvant chemotherapy in premenopausal women with node-negative early breast cancer. J Clin Oncol 1998; 16:470.

59. Formenti SC, Dunnington G, Uzieli B, et al. Original p53 status predicts for pathological response in locally advanced breast cancer patients treated preoperatively with continuous infusion 5-fluorouracil and radiation therapy. Int J Radiat Oncol Biol Phys 1997; 39:1059.

60. Weisenthal LM, Kern DH. Prediction of drug resistance in cancer chemotherapy: the Kern and DiSC assays. Oncology (Williston Park) 1991; 5:93.

61. Tavassoli FA, Cook CB, Pestaner JP. A comparison of two commercially available in vitro chemosensitivity assays. Oncology 1995; 52:413.

62. Elledge RM, Clark GM, Hon J, et al. Rapid in vitro assay for predicting response to fluorouracil in patients with metastatic breast cancer. J Clin Oncol 1995; 13:419.

63. Samson DJ, Seidenfeld J, Ziegler K, Aronson N. Chemotherapy sensitivity and resistance assays: a systematic review. J Clin Oncol 2004; 22:3618.

64. Schrag D, Garewal HS, Burstein HJ, et al. American Society of Clinical Oncology Technology Assessment: chemotherapy sensitivity and resistance assays. J Clin Oncol 2004; 22:3631.

65. Wilcken N, Hornbuckle J, Ghersi D. Chemotherapy alone versus endocrine therapy alone for metastatic breast cancer. Cochrane Database Syst Rev 2003; :CD002747.

66. Taylor SG 4th, Gelman RS, Falkson G, Cummings FJ. Combination chemotherapy compared to tamoxifen as initial therapy for stage IV breast cancer in elderly women. Ann Intern Med 1986; 104:455.

67. A randomized trial in postmenopausal patients with advanced breast cancer comparing endocrine and cytotoxic therapy given sequentially or in combination. The Australian and New Zealand Breast Cancer Trials Group, Clinical Oncological Society of Australia. J Clin Oncol 1986; 4:186.

68. Carrick S, Parker S, Wilcken N, et al. Single agent versus combination chemotherapy for metastatic breast cancer. Cochrane Database Syst Rev 2005; :CD003372.

69. Norris B, Pritchard KI, James K, et al. Phase III comparative study of vinorelbine combined with doxorubicin versus doxorubicin alone in disseminated metastatic/recurrent breast cancer: National Cancer Institute of Canada Clinical Trials Group Study MA8. J Clin Oncol 2000; 18:2385.

70. O'Shaughnessy J, Miles D, Vukelja S, et al. Superior survival with capecitabine plus docetaxel combination therapy in anthracycline-pretreated patients with advanced breast cancer: phase III trial results. J Clin Oncol 2002; 20:2812.

Page 15: Systemic Treatment for Metastatic Breast Cancer

71. Martín M, Ruiz A, Muñoz M, et al. Gemcitabine plus vinorelbine versus vinorelbine monotherapy in patients with metastatic breast cancer previously treated with anthracyclines and taxanes: final results of the phase III Spanish Breast Cancer Research Group (GEICAM) trial. Lancet Oncol 2007; 8:219.

72. Sledge GW, Neuberg D, Bernardo P, et al. Phase III trial of doxorubicin, paclitaxel, and the combination of doxorubicin and paclitaxel as front-line chemotherapy for metastatic breast cancer: an intergroup trial (E1193). J Clin Oncol 2003; 21:588.

73. Alba E, Martín M, Ramos M, et al. Multicenter randomized trial comparing sequential with concomitant administration of doxorubicin and docetaxel as first-line treatment of metastatic breast cancer: a Spanish Breast Cancer Research Group (GEICAM-9903) phase III study. J Clin Oncol 2004; 22:2587.

74. Conte PF, Guarneri V, Bruzzi P, et al. Concomitant versus sequential administration of epirubicin and paclitaxel as first-line therapy in metastatic breast carcinoma: results for the Gruppo Oncologico Nord Ovest randomized trial. Cancer 2004; 101:704.

75. Soto C, Torrecillas L, Reyes S, et al. Capecitabine (X) and taxanes in patients with anthracycline-pretreated metastatic breast cancer: sequential vs. combined therapy results from a MOSG randomized phase III trial (abstract). J Clin Oncol 2006; 24:20s.

76. Bishop JF, Dewar J, Toner GC, et al. Initial paclitaxel improves outcome compared with CMFP combination chemotherapy as front-line therapy in untreated metastatic breast cancer. J Clin Oncol 1999; 17:2355.

77. Joensuu H, Holli K, Heikkinen M, et al. Combination chemotherapy versus single-agent therapy as first- and second-line treatment in metastatic breast cancer: a prospective randomized trial. J Clin Oncol 1998; 16:3720.

78. Heidemann E, Stoeger H, Souchon R, et al. Is first-line single-agent mitoxantrone in the treatment of high-risk metastatic breast cancer patients as effective as combination chemotherapy? No difference in survival but higher quality of life were found in a multicenter randomized trial. Ann Oncol 2002; 13:1717.

79. Jones D, Ghersi D, Wilcken N. Addition of drug/s to a chemotherapy regimen for metastatic breast cancer. Cochrane Database Syst Rev 2006; :CD003368.

80. Wilcken N, Dear R. Chemotherapy in metastatic breast cancer: A summary of all randomised trials reported 2000-2007. Eur J Cancer 2008; 44:2218.

81. Fossati R, Confalonieri C, Torri V, et al. Cytotoxic and hormonal treatment for metastatic breast cancer: a systematic review of published randomized trials involving 31,510 women. J Clin Oncol 1998; 16:3439.

82. Coates A, Gebski V, Bishop JF, et al. Improving the quality of life during chemotherapy for advanced breast cancer. A comparison of intermittent and continuous treatment strategies. N Engl J Med 1987; 317:1490.

83. Muss HB, Case LD, Richards F 2nd, et al. Interrupted versus continuous chemotherapy in patients with metastatic breast cancer. The Piedmont Oncology Association. N Engl J Med 1991; 325:1342.

84. Cheung KL, Evans AJ, Robertson JF. The use of blood tumour markers in the monitoring of metastatic breast cancer unassessable for response to systemic therapy. Breast Cancer Res Treat 2001; 67:273.

Page 16: Systemic Treatment for Metastatic Breast Cancer

85. Robertson JF, Whynes DK, Dixon A, Blamey RW. Potential for cost economies in guiding therapy in patients with metastatic breast cancer. Br J Cancer 1995; 72:174.

86. Stearns V, Yamauchi H, Hayes DF. Circulating tumor markers in breast cancer: accepted utilities and novel prospects. Breast Cancer Res Treat 1998; 52:239.

87. Kiang DT, Greenberg LJ, Kennedy BJ. Tumor marker kinetics in the monitoring of breast cancer. Cancer 1990; 65:193.

88. Symeonidis A, Kouraklis-Symeonidis A, Apostolopoulos D, et al. Increased serum CA-15.3 levels in patients with megaloblastic anemia due to vitamin B12 deficiency. Oncology 2004; 67:359.

89. Symeonidis A, Kouraklis-Symeonidis A, Constantinidou I, et al. Increased CA-15.3 levels in the serum of patients with homozygous beta-thalassaemia and sickle cell/beta-thalassaemia. Br J Haematol 2006; 133:692.

90. Boga C, Ozdogu H, Sezgin N, et al. Serum cancer antigen 15-3 concentrations in patients with sickle cell disease. Br J Haematol 2006; 134:546.

91. Janicek MJ, Hayes DF, Kaplan WD. Healing flare in skeletal metastases from breast cancer. Radiology 1994; 192:201.

92. Vogel CL, Schoenfelder J, Shemano I, et al. Worsening bone scan in the evaluation of antitumor response during hormonal therapy of breast cancer. J Clin Oncol 1995; 13:1123.

93. Ben-Haim S, Israel O. Breast cancer: role of SPECT and PET in imaging bone metastases. Semin Nucl Med 2009; 39:408.

94. Constantinidou A, Martin A, Sharma B, Johnston SR. Positron emission tomography/computed tomography in the management of recurrent/metastatic breast cancer: a large retrospective study from the Royal Marsden Hospital. Ann Oncol 2011; 22:307.

95. Rosen EL, Eubank WB, Mankoff DA. FDG PET, PET/CT, and breast cancer imaging. Radiographics 2007; 27 Suppl 1:S215.

96. Stafford SE, Gralow JR, Schubert EK, et al. Use of serial FDG PET to measure the response of bone-dominant breast cancer to therapy. Acad Radiol 2002; 9:913.

97. Tateishi U, Gamez C, Dawood S, et al. Bone metastases in patients with metastatic breast cancer: morphologic and metabolic monitoring of response to systemic therapy with integrated PET/CT. Radiology 2008; 247:189.

98. Specht JM, Tam SL, Kurland BF, et al. Serial 2-[18F] fluoro-2-deoxy-D- glucose positron emission tomography (FDG-PET) to monitor treatment of bone-dominant metastatic breast cancer predicts time to progression (TTP). Breast Cancer Res Treat 2007; 105:87.

99. Weigelt B, Bosma AJ, Hart AA, et al. Marker genes for circulating tumour cells predict survival in metastasized breast cancer patients. Br J Cancer 2003; 88:1091.

100. Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 2004; 351:781.

101. Cristofanilli M, Hayes DF, Budd GT, et al. Circulating tumor cells: a novel prognostic factor for newly diagnosed metastatic breast cancer. J Clin Oncol 2005; 23:1420.

Page 17: Systemic Treatment for Metastatic Breast Cancer

102. Hayes DF, Cristofanilli M, Budd GT, et al. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res 2006; 12:4218.

103. Wong NS, Kahn HJ, Zhang L, et al. Prognostic significance of circulating tumour cells enumerated after filtration enrichment in early and metastatic breast cancer patients. Breast Cancer Res Treat 2006; 99:63.

104. Dawood S, Broglio K, Valero V, et al. Circulating tumor cells in metastatic breast cancer: from prognostic stratification to modification of the staging system? Cancer 2008; 113:2422.

105. Budd GT, Cristofanilli M, Ellis MJ, et al. Circulating tumor cells versus imaging--predicting overall survival in metastatic breast cancer. Clin Cancer Res 2006; 12:6403.

106. Liu MC, Shields PG, Warren RD, et al. Circulating tumor cells: a useful predictor of treatment efficacy in metastatic breast cancer. J Clin Oncol 2009; 27:5153.

107. Information on SWOG trial 0500 available online at http://clinicaltrials.gov/ct2/show/NCT00382018 (Accessed on April 28, 2011).