Skip to main content
BMJ Clinical Evidence logoLink to BMJ Clinical Evidence
. 2007 Dec 4;2007:0102.

Breast cancer (non-metastatic)

Justin Stebbing 1,#, Geoff Delaney 2,#, Alistair Thompson 3,#
PMCID: PMC2943780  PMID: 19450345

Abstract

Introduction

Breast cancer affects at least 1 in 10 women in the UK, but most present with primary operable disease, which has an 80% 5-year survival rate overall.

Methods and outcomes

We conducted a systematic review and aimed to answer the following clinical questions: What are the effects of interventions after breast-conserving surgery for ductal carcinoma in situ? What are the effects of treatments for primary operable breast cancer? What are the effects of interventions in locally advanced breast cancer (stage IIIB)? We searched: Medline, Embase, The Cochrane Library and other important databases up to February 2006 (BMJ Clinical Evidence reviews are updated periodically, please check our website for the most up-to-date version of this review). We included harms alerts from relevant organisations such as the US Food and Drug Administration (FDA) and the UK Medicines and Healthcare products Regulatory Agency (MHRA).

Results

We found 79 systematic reviews, RCTs, or observational studies that met our inclusion criteria. We performed a GRADE evaluation of the quality of evidence for interventions.

Conclusions

In this systematic review we present information relating to the effectiveness and safety of the following interventions: adding chemotherapy (cyclophosphamide/methotrexate/ fluorouracil and/or anthracycline and/or taxane-based regimens), or hormonal treatment to radiotherapy; adjuvant treatments (aromatase inhibitors, adjuvant anthracycline regimens, tamoxifen); axillary clearance; axillary dissection plus sentinel node dissection; axillary radiotherapy; axillary sampling; combined chemotherapy plus tamoxifen; chemotherapy plus monoclonal antibody (trastuzumab); extensive surgery; high-dose chemotherapy; hormonal treatment; less extensive mastectomy; less than whole breast radiotherapy plus breast conserving surgery; multimodal treatment; ovarian ablation; primary chemotherapy; prolonged adjuvant combination chemotherapy; radiotherapy (after breast-conserving surgery, after mastectomy, plus tamoxifen after breast-conserving surgery, to the internal mammary chain, and to the ipsilateral supraclavicular fossa, and total nodal radiotherapy); sentinel node biopsy; and standard chemotherapy regimens.

Key Points

Breast cancer affects at least 1 in 10 women in the UK, but most present with primary operable disease, which has an 80% 5-year survival rate overall.

In women with ductal carcinoma in situ, radiotherapy reduces local recurrence and invasive carcinoma after breast-conserving surgery, but may not improve survival.

In women with primary operable breast cancer, survival may be increased by full surgical excision, tamoxifen, chemotherapy, radiotherapy, ovarian ablation or trastuzumab (in women who overexpress HER2/neu oncogene).

  • Incomplete excision may increase the risk of local recurrence, but less-extensive mastectomy that excises all local disease is as effective as radical mastectomy at prolonging survival, with better cosmetic results.

  • Axillary clearance (removal of all axillary lymph nodes) achieves local disease control, but has not been shown to increase survival, and can cause arm lymphoedema.

  • Sentinel lymph node biopsy or 4-node sampling may adequately stage the axilla with less morbidity compared with axillary clearance.

  • Adjuvant tamoxifen reduces the risk of recurrence and death in women with oestrogen-positive tumours, but adverse effects begin to outweigh benefit after 5 years of treatment.

  • Primary chemotherapy may facilitate successful breast-conserving surgery instead of mastectomy. Adjuvant combination chemotherapy improves survival compared with no chemotherapy, with greatest benefit likely with anthracycline-based regimens at standard doses for 4-6 months.

  • Radiotherapy decreases recurrence and mortality after breast-conserving surgery. Post-mastectomy radiotherapy for women who are node-positive or at high risk of recurrence decreases recurrence and mortality, but may increase mortality in node-negative women.

  • Adjuvant aromatase inhibitors improve disease-free survival compared with tamoxifen, but their effect on overall survival is unclear.Adjuvant taxoid regimens may improve disease-free survival over standard anthracycline-based therapy.

In women with locally advanced breast cancer, radiotherapy may be as effective as surgery or tamoxifen at increasing survival and local disease control.

  • Adding tamoxifen or ovarian ablation to radiotherapy increases survival compared with radiotherapy alone, but adding chemotherapy may not reduce recurrence or mortality compared with radiotherapy alone.

  • Chemotherapy alone, while widely used, does not improve survival in women with locally advanced breast cancer.

About this condition

Definition

This review examines the effects of treatment for non-metastatic, primary breast cancer. Ductal carcinoma in situ is a non-invasive tumour characterised by the presence of malignant cells in the breast ducts, but with no evidence that they breach the basement membrane and invade into periductal connective tissues. Invasive breast cancer occurs when cancer cells spread beyond the basement membrane, which covers the underlying connective tissue in the breast. This tissue is rich in blood vessels and lymphatic channels capable of carrying cancer cells beyond the breast. Invasive breast cancer can be separated into three main groups: early invasive breast cancer, locally advanced breast cancer, and metastatic breast cancer (see review on breast cancer [metastatic]). Operable breast cancer is apparently restricted to the breast and to local lymph nodes, and can be removed surgically. Although women do not have overt metastases at the time of staging, they remain at risk of local recurrence, and of metastatic spread. They can be divided into those with tumours greater than 4 cm or with multifocal cancers that are usually treated by mastectomy, and those with tumours less than 4 cm cancers that can be treated by breast-conserving surgery. Locally advanced breast cancer is defined according to the TNM staging system of the UICC as stage IIIB (includes T4 a-d; N2 disease, but absence of metastases [see table 1 ]). It is a disease presentation with clinical or histopathological evidence of skin and/or chest-wall involvement, and/or axillary nodes matted together by tumour extension. Metastatic breast cancer is presented in a separate review (see review on breast cancer [metastatic]).

Table 1.

Staging of breast cancer (the individual terms are explained in the glossary) (see text).

  TNM Stage
Non-invasive Tis N0 M0 0
Early invasive T1-2 N0-1 M0 I, II A or B
  T3 N0 M0 II B
Advanced        
Locally advanced Tany N2 M0 III A
  T3 N1-2 M0 III A
  T4 N0-3 M0 III B
  Tany N3 M0 III B
Metastatic Tany Nany M1 IV

Incidence/ Prevalence

Breast cancer affects 1/10-1/11 women in the UK and causes about 21,000 deaths a year. Prevalence is about five times higher, with over 100,000 women in the UK living with breast cancer at any one time. Of the 36,000 new cases of breast cancer each year in England and Wales, most will present with primary operable disease.

Aetiology/ Risk factors

The risk of breast cancer increases with age, doubling every 10 years up to the menopause. Risk factors include an early age at menarche, older age at menopause, older age at birth of first child, family history, atypical hyperplasia, excess alcohol intake, radiation exposure to developing breast tissue, oral contraceptive use, postmenopausal HRT, and postmenopausal obesity. Risk in different countries varies fivefold. The cause of breast cancer in most women is unknown. About 5% of breast cancers can be attributed to mutations in the genes BRCA1 and BRCA2, but the contribution to inherited breast cancer of other genes, including Chk2, ATM, p53, and PTEN, is currently less well established.

Prognosis

Non-metastatic carcinoma of the breast is potentially curable. The risk of relapse depends on various clinicopathological features, of which axillary node involvement, tumour grade, tumour size, and oestrogen receptor status are the most prognostically important. Of women with operable disease, 80% are alive 5 years after diagnosis and treatment (adjuvant treatment is given to most women after surgery). Risk of recurrence is highest during the first 5 years, but the risk remains even 15-20 years after surgery. Recurrence at 10 years, according to one large systematic review, is 60-70% in node-positive women, and 25-30% in node negative women. The prognosis for disease-free survival at 5 years is worse for stage IIIB (33%) than that for stage IIIA (71%). Overall survival at 5 years is 44% for stage IIIB and 84% for stage IIIA. Poor survival and high rates of local recurrence characterise locally advanced breast cancer.

Aims of intervention

To improve survival; to prevent local or regional node recurrence; to obtain prognostic information on the type and extent of tumour and the status of the axillary lymph nodes; to optimise cosmetic results and minimise psychosocial impact; to minimise adverse effects of treatment; and to maximise quality of life.

Outcomes

Survival; rates of local and regional recurrence; rates of mastectomy after breast-conserving treatment; rates of development of metastases; cosmetic outcomes; quality of life; incidence of adverse effects of treatment, including upper-limb lymphoedema.

Methods

BMJ Clinical Evidence search and appraisal January 2007. The following databases were used to identify studies for this review: Medline 1966 to January 2007, Embase 1980 to January 2007, and The Cochrane Database of Systematic Reviews and Cochrane Central Register of Controlled Clinical Trials 2006, Issue 4. Additional searches were carried out using these websites: NHS Centre for Reviews and Dissemination (CRD) — for Database of Abstracts of Reviews of Effects (DARE) and Health Technology Assessment (HTA), Turning Research into Practice (TRIP), and National Institute for Health and Clinical Excellence (NICE). Abstracts of the studies retrieved from the initial search were assessed by an information specialist. Selected studies were then sent to the authors for additional assessment, using pre-determined criteria to identify relevant studies. Study design criteria for evaluation in this review were: published systematic reviews and RCTs in any language, any level of blinding, and containing more than 20 individuals of whom more than 80% were followed up. There was no minimum length of follow-up required to include studies. In addition, we use a regular surveillance protocol to capture harms alerts from organisations such as the US Food and Drug Administration (FDA) and the UK Medicines and Healthcare products Regulatory Agency (MHRA), which are added to the review as required. The authors also identified data by personal communication with experts in the field, and through hand searches. Abstracts of studies that were considered to be fundamental to clinical practice and new developments were included in comments sections. Phase 3 randomised published data are included. We have performed a GRADE evaluation of the quality of evidence for interventions included in this review (see table ).

Table.

GRADE evaluation of interventions for breast cancer (non-metastatic)

Important outcomes Treatment success, mortality, adverse effects
Number of studies (participants) Outcome Comparison Type of evidence Quality Consistency Directness Effect size GRADE Comment
What are the effects of interventions after breast-conserving surgery for ductal carcinoma in situ?
3, 1 paper (2862) Treatment success Radiotherapy v no radiotherapy 4 0 0 0 0 High
3, 1 paper (2862) Mortality Radiotherapy v no radiotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1, 1 paper (1804) Treatment success Tamoxifen plus radiotherapy v placebo 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1, 1 paper (1804) Mortality Tamoxifen plus radiotherapy v placebo 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (523) Treatment success Tamoxifen plus radiotherapy v radiothera py 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
What are the effects of treatments for primary operable breast cancer?
1 (5187 Mortality Adjuvant aromatase inhibitors v placebo 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
2, 2 papers (14068) Treatment success Adjuvant aromatase inhibitors v tamoxifen 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
2, 2 papers (14068) Mortality Adjuvant aromatase inhibitors v tamoxifen 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
47 (18000) Treatment success Adjuvant combination chemotherapy v no chemotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
47 (18000) Mortality Adjuvant combination chemotherapy v no chemotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
not reported (3454) Treatment success Different treatment durations compared with each other ( prolonged adjuvant combination chemotherapy) 4 0 0 0 0 High
not reported (3454) Mortality Different treatment durations compared with each other ( prolonged adjuvant combination chemotherapy) 4 0 0 0 0 High
2 (3877) Treatment success Different doses compared with each other (enhancedose regimens of adjuvant combination chemotherapy) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
2 (3877) Mortality Different doses compared with each other (enhancedose regimens of adjuvant combination chemotherapy) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
11 (5942) Treatment success Anthracycline regimens compared with standard CMF regimens 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
11 (5942) Mortality Anthracycline regimens compared with standard CMF regimens 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
55 (37000) Treatment success Adjuvant tamoxifen v placebo 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
55 (37000) Mortality Adjuvant tamoxifen v placebo 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (3887) Treatment success Durations of treatment compared with each other (adjuvant tamoxifen) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (3887) Mortality Durations of treatment compared with each other (adjuvant tamoxifen) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (3387) Treatment success Trastuzumab v observation (after chemotherapy) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (3387) Mortality Trastuzumab v observation (after chemotherapy) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
2 (3351) Treatment success Trastuzumab v observation (during chemotherapy) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
2 (3351) Mortality Trastuzumab v observation (during chemotherapy) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
6 (at least 3490 women) Treatment success Adjuvant taxoid regimens v standard anthracycline based regimens 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
6 (at least 3490 women) Mortality Adjuvant taxoid regimens v standard anthracycline based regimens 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
5 (2090) Mortality Supraradical, radical, and total mastectomy compared with each other 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
4 (1296) Mortality Radical/total mastectomy v simple mastectomy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (1079) Mortality Radical mastectomy v total mastectomy with or without axillary radiotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
6 (3107) Treatment success Mastectomy v breast conservation 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
6 (4438) Mortality Mastectomy v breast conservation 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (701) Mortality Radical mastectomy v quandrantectomy plus radiotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (1851) Mortality Lumpectomy with or without radiotherapy v total mastectomy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (237) Mortality Modified radical mastectomy v lumpectomy plus axillary dissection plus radiotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (705) Treatment success Different extents of local excision compared with each other 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
12 (2102) Treatment success Ovarian ablation v no ablation 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
12 (2102) Mortality Ovarian ablation v no ablation 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
10 (7311) Treatment success Breast-conserving surgery plus radiotherapy v breast-conserving surgery alone 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
10 (7311) Mortality Breast-conserving surgery plus radiotherapy v breast-conserving surgery alone 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
8 (3531) Treatment success Breast-conserving surgery plus radiotherapy v mastectomy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
7 (3294) Mortality Breast-conserving surgery plus radiotherapy v mastectomy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
3 (2073) Treatment success Breast-conserving surgery plus radiotherapy v breast-conserving surgery plus tamoxifen 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
3 (2071) Mortality Radiotherapy plus tamoxifen v tamoxifen alone (after breast-conserving surgery) 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
36 (10982) Treatment success Radiotherapy after mastectomy v mastectomy alone (in women at high risk of local recurrence) 4 −1 −1 0 0 Low Quality point deducted for incomplete reporting of results. Consistency point deducted for heterogeneity between RCTs
36 (10982) Mortality Radiotherapy after mastectomy v mastectomy alone (in women at high risk of local recurrence) 4 −1 −1 0 0 Low Quality point deducted for incomplete reporting of results. Consistency point deducted for heterogeneity between RCTs
36 (5195) Treatment success Radiotherapy after mastectomy v mastectomy alone (in women not at high risk of local recurrence) 4 −1 −1 0 0 Low Quality point deducted for incomplete reporting of results. Consistency point deducted for heterogeneity between RCTs
36 (5195) Mortality Radiotherapy after mastectomy v mastectomy alone (in women not at high risk of local recurrence) 4 −1 −1 0 0 Low Quality point deducted for incomplete reporting of results. Consistency point deducted for heterogeneity between RCTs
4 (2802) Treatment success Primary chemotherapy v adjuvant chemotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
5 (3216) Mortality Primary chemotherapy v adjuvant chemotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
26 (not reported) Treatment success Radiotherapy after mastectomy v mastectomy alone in women at high risk of local recurrence 4 −2 −1 0 0 Very low Quality points deducted for incomplete reporting of results and for inclusion of RCTs without total nodal radiation. Consistency point deducted for heterogeneity between RCTs
26 (not reported) Mortality Radiotherapy after mastectomy v mastectomy alone in women at high risk of local recurrence 4 −2 −2 0 0 Very low Quality points deducted for incomplete reporting of results and for inclusion of RCTs without total nodal radiation. Consistency point deducted for conflicting results and for heterogeneity between RCTs
1 (466) Mortality Axillary clearance v axillary sampling 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
8 (4370) Treatment success Axillary clearance v axillary radiotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
8 (4370) Mortality Axillary clearance v axillary radiotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (448) Treatment success Standard v dose intensified anthracycline based regimens 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (448) Mortality Standard v dose intensified anthracycline based regimens 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (174) Treatment success FAC regimen (fluorouracil, adriamycin [doxorubicin], and cyclophosphamide) v single agent paclitaxel 4 −2 0 0 0 Low Quality points deducted for sparse data and incomplete reporting of results
1 (174) Mortality FAC regimen (fluorouracil, adriamycin [doxorubicin], and cyclophosphamide) v single agent paclitaxel 4 −2 0 0 0 Low Quality points deducted for sparse data and incomplete reporting of results
1 (101) Treatment success MPEMi (methotrexate, cisplatin, etoposide, mitomycin C), MPEpiE (methotrexate, cisplatin, epirubicin, etoposide), and MPEpiV (methotrexate, cisplatin, epirubicin, vincristine) regimens v each other 4 −2 0 0 0 Low Quality points deducted for sparse data and incomplete reporting of results
2 (2515) Treatment success Sequencing of anthracycline based chemotherapy and docetaxel 4 −2 0 0 0 Low Quality points deducted for incomplete reporting of results and for no long term results
1 (73) Treatment success Intra-arterial v intravenous administration 4 −2 0 0 0 Low Quality points deducted for sparse data and incomplete reporting of results
2 (876) Treatment success Less than whole-breast radiotherapy v whole breast radiotherapy 4 0 −1 0 0 Moderate Consistency point deducted for conflicting results
1 (168) Mortality Less than whole-breast radiotherapy v whole breast radiotherapy 4 −1 −1 0 0 Moderate Quality point deducted for sparse data and conflicting results
1 (270) Treatment success Radiotherapy to the internal mammary chain v nointernal mammary chain irradiation 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (270) Mortality Radiotherapy to the internal mammary chain v nointernal mammary chain irradiation 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (1375) Treatment success Radiotherapy v no radiotherapy to the ipsilateral supraclavicular fossa 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
2 (498) Treatment success Sentinel node biopsy plus total axillary dissection v with sentinel node biopsy alone 4 −2 0 0 0 Low Quality points deducted for incomplete reporting of results and for uncertainty about follow-up
1 (200) Mortality Sentinel node biopsy plus total axillary dissection v with sentinel node biopsy alone 4 −2 0 0 0 Low Quality points deducted for incomplete reporting of results and for uncertainty about follow-up
at least 6 RCTs (at least 3015 women) Mortality High-dose chemotherapy plus autologous stem cell transplantation v conventional chemotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
13 (5064) Adverse effects High-dose chemotherapy plus autologous stem cell transplantation v conventional chemotherapy 4 0 0 0 +2 High Effect size points added for RR greater than 5
What are the effects of interventions in locally advanced breast cancer (stage III B)?
4 (3599) Treatment success Post-operative radiotherapy (in women also receiving postoperative systemic treatment) v no radiotherapy 4 −2 0 −1 0 Low Quality poinst deducted for incomplete reporting of results and for weak methods. Directness points deducted for inclusion of women with different disease severities
4 (3599) Mortality Post-operative radiotherapy (in women also receiving postoperative systemic treatment) v no radiotherapy 4 −2 0 −1 0 Low Quality poinst deducted for incomplete reporting of results and for weak methods. Directness points deducted for inclusion of women with different disease severities
2 (219) Treatment success Surgery alone v radiotherapy alone 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
2 (219) Mortality Surgery v radiotherapy 4 −1 0 0 0 Moderate Quality point deducted for incomplete reporting of results
1 (143) Mortality Low-dose radiotherapy v tamoxifen 4 −2 0 0 0 Low Quality points deducted for sparse data and incomplete reporting of results
3 (580) Treatment success Systemic treatment plus radiotherapy v radiotherapy 4 −1 −1 −1 0 Very low Quality point deducted for incomplete reporting of results. Consistency point deducted for conflicting results. Directness points deducted for inclusion of women with less locally advanced breast cancer
3 (580) Mortality Systemic treatment plus radiotherapy v radiotherapy 4 −1 −1 −1 0 Very low Quality point deducted for incomplete reporting of results. Consistency point deducted for conflicting results. Directness points deducted for inclusion of women with less locally advanced breast cancer
1, 1 publication (107) Treatment success Multimodal treatment v hormone treatment 4 −1 −1 0 +1 Moderate Quality point deducted for sparse data. Consistency point deducted for different results at different end points. Effect size point added for OR 2–5
1 (101) Mortality Adjuvant chemotherapy v primary plus adjuvant chemotherapy 4 −1 0 0 0 Moderate Quality point deducted for sparse data

Type of evidence: 4 = RCT; 2 = Observational Consistency: similarity of results across studies Directness: generalisability of population or outcomes Effect size: based on relative risk or odds ratio

Glossary

Adjuvant treatment

(also known as postoperative chemotherapy) This usually refers to systemic chemotherapy, hormonal treatment, or both, given to people after removal of a primary tumour (in this case, surgery for early breast cancer), with the aim of killing any remaining micrometastatic tumour cells and thus preventing recurrence.

Anthracyclines

These are also known as cytotoxic antibiotics, and are used as adjuvant treatment with radiotherapy. Examples of anthracyclines are aclarubicin, daunorubicin, adriamycin (doxorubicin), epirubicin, and idarubicin.

Axillary clearance

Clearance of level I, II, and usually level III axillary lymph nodes. Level I nodes are lateral to the pectoralis minor muscle, level II nodes are under it, and level III nodes are medial to it at the apex of the axilla.

Axillary radiotherapy

This usually includes irradiation of the supraclavicular fossa. Irradiation of this area incorporates some underlying lung, which increases the risk of radiation pneumonitis. By increasing the volume of the lung irradiated, compared with chest wall or breast radiotherapy alone, the risk of acute pneumonitis is increased.

Axillary sampling

Aims to remove the four largest, most easily palpable axillary lymph nodes for histological examination.

Brachial plexopathy

Damage — usually permanent and often progressive — to the brachial plexus. It may arise from radiotherapy as a delayed or late event. It may also be caused by tumour infiltration.

Brachytherapy

Delivery of a radiation dose over a short distance either from a low-energy x ray source, a low-energy electron source, or a radioactive source.

Breast-conserving surgery

Surgery consisting of lumpectomy (minimal cancer-free margins), wide local excision (wider free margins), or segmental or quadrant resection (usually with wide-free margins).

CMF (classical)

Chemotherapy regimen containing cyclophosphamide, methotrexate, and fluorouracil.

Combination chemotherapy

Two or more cytotoxic drugs given intravenously every 3–4 weeks for 4–6 months.

Disease-free survival

Means being alive with no local or distant recurrence or contralateral disease.

Early invasive breast cancer

(stage I or II) is M0 with T1 or T2 (tumour diameter ≤ 5 cm, no involvement of skin or chest wall) and N0 or N1 (mobile axillary nodes); or M0 with T3 (tumour diameter over 5 cm , no skin or chest wall involvement), but only N0.

FAC

Chemotherapy regimen containing fluorouracil, adriamycin (doxorubicin), and cyclophosphamide.

High-quality evidence

Further research is very unlikely to change our confidence in the estimate of effect.

Less than whole-breast radiotherapy

The delivery of radiotherapy by either temporary internal placement of a radiation source or by external beam treatment to part of the breast (area of wide local excision after breast-conserving surgery).

Locally advanced breast cancer

Operable locally advanced breast cancer (stage III A) is T3 (tumours more than 5 cm) and N1 (non-matted involved axillary nodes). Locally advanced breast cancer (stage III B) is M0 with T4 (skin or chest wall infiltration by tumour), N2 (matted axillary nodes)/N3 (internal mammary node involvement) disease, or both, not classified as non-invasive or early invasive breast cancer. Metastatic breast cancer (stage IV) is M1 (any supraclavicular fossa node involvement or distant metastases to bone, lung, liver, etc.) with any combination of tumour and node parameters.

Low-quality evidence

Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.

Lumpectomy

Removal of tumour with a 1 cm margin (macroscopically) around the palpable cancer.

Milan regimen

A sequential regimen of single agent anthracycline followed by CMF.

Moderate-quality evidence

Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.

Modified radical mastectomy

Modified radical mastectomy is a total mastectomy with removal of all axillary nodes from level I medial to the pectoralis minor, level II underneath the pectoralis minor, and up to the apex and including level III nodes medial to the pectoralis minor but below the axillary vein up to the first rib. Traditionally, a modified radical mastectomy included excision of the pectoralis minor, but most surgeons performing a modified radical mastectomy nowadays preserve the pectoralis minor.

Ovarian ablation

Surgical, medical, or radiation induced suppression of ovarian function in premenopausal women.

Overall objective response rate

The proportion of treated people in whom a complete response (disappearance of all known lesions on 2 separate measurements at least 4 weeks apart), or partial response ( more than 50% reduction in the size of lesions) is observed.

Primary chemotherapy

(also known as neoadjuvant or preoperative chemotherapy) involves the use of chemotherapy to treat breast cancer before loco-regional treatment (surgery and or radiotherapy) to the breast to downstage large primary cancers which would require mastectomy to improve chances of survival.

Quadrantectomy

Tumour excised with ≥ 2 cm of normal surrounding breast tissue and with a segment of breast tissue from the periphery of the breast to the nipple.

Radical mastectomy

Removal of the breast and pectoralis major and minor muscles and axillary contents.

Radiotherapy

Part of initial local and regional treatment. In early stage disease, it may be an adjunct to surgery; in locally advanced disease (T4, N2), it may be the sole locoregional treatment. Radiotherapy may be delivered to the breast or postmastectomy chest wall, as well as to the lymphatic areas of the axilla, supraclavicular fossa, or internal mammary node chain.

Sentinel node biopsy

A procedure whereby the first nodes in the draining lymphatic basin are removed and examined by a pathologist for cancer cells.

Simple mastectomy

Removal of the breast tissue, usually in association with an ellipse of skin which includes the nipple and areolar complex. Dissection continues down to, but does not usually include, the pectoral fascia. It includes removal of the axillary tail of the breast. Lymph nodes are not usually removed other than by an additional procedure.

Supraradical mastectomy

Removal of breast, pectoralis major and minor muscles, axillary contents, and internal mammary chain of nodes.

Systemic therapy

Use of oral or intravenous treatments which affect the whole body.

TNM staging system

See “staging of breast cancer”, above.

Total mastectomy

Surgery to remove the entire breast.

Total nodal irradiation

Radiotherapy to the regional lymph nodes, including supraclavicular, infraclavicular, axillary nodes, and internal mammary nodes in the upper intercostal spaces.

UICC

International Union against Cancer.

Very low-quality evidence

Any estimate of effect is very uncertain.

See review on breast cancer (metastatic).

Disclaimer

The information contained in this publication is intended for medical professionals. Categories presented in Clinical Evidence indicate a judgement about the strength of the evidence available to our contributors prior to publication and the relevant importance of benefit and harms. We rely on our contributors to confirm the accuracy of the information presented and to adhere to describe accepted practices. Readers should be aware that professionals in the field may have different opinions. Because of this and regular advances in medical research we strongly recommend that readers' independently verify specified treatments and drugs including manufacturers' guidance. Also, the categories do not indicate whether a particular treatment is generally appropriate or whether it is suitable for a particular individual. Ultimately it is the readers' responsibility to make their own professional judgements, so to appropriately advise and treat their patients.To the fullest extent permitted by law, BMJ Publishing Group Limited and its editors are not responsible for any losses, injury or damage caused to any person or property (including under contract, by negligence, products liability or otherwise) whether they be direct or indirect, special, incidental or consequential, resulting from the application of the information in this publication.

Contributor Information

Justin Stebbing, Chelsea and Westminister Hospital, London, UK.

Geoff Delaney, Cancer Therapy Centre, Liverpool Hospital, Liverpool, UK.

Professor Alistair Thompson, Department of Surgery and Molecular Oncology, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.

References

  • 1.UICC International Union Against Cancer. TNM classification of malignant tumours, 6th ed. Sobin LH, Wittekind CH, eds. New York: Wiley-Liss, 2002. [Google Scholar]
  • 2.Office for National Statistics. 2005. http://www.statistics.gov.uk/CCI/nugget.asp?ID=575&Pos=&ColRank=1&Rank=374 (last accessed 09 August 2005). [Google Scholar]
  • 3.Easton D, Ford D. Breast and ovarian cancer incidence in BRCA-1 mutation carriers. Am J Hum Genet 1995;56:265–271. [PMC free article] [PubMed] [Google Scholar]
  • 4.Carter CL, Allen C, Henson DE. Relation of tumour size, lymph node status and survival in 24 740 breast cancer cases. Cancer 1989;63:181–187. [DOI] [PubMed] [Google Scholar]
  • 5.Hortobagyi GN, Ames FC, Buzdar AU, et al. Management of stage III primary breast cancer with primary chemotherapy, surgery and radiation therapy. Cancer 1988;62:2507–2516. [DOI] [PubMed] [Google Scholar]
  • 6.Rutqvist LE, Rose C, Cavallin-Stahl E. A systematic overview of radiation therapy effects in breast cancer. Acta Oncol 2003;42:532–545. [DOI] [PubMed] [Google Scholar]
  • 7.Fisher B, Dignam J, Wolmark N, et al. Lumpectomy and radiation therapy for the treatment of intraductal breast cancer: findings of the National Surgical Adjuvant Breast and Bowel Project B-17. J Clin Oncol 1998;16:441–452. [DOI] [PubMed] [Google Scholar]
  • 8.Julien JP, Bijker N, Fentiman IS, et al. Radiotherapy in breast-conserving treatment for ductal carcinoma in situ; first results of EORTC randomised Phase III trial 10853. Lancet 2000;355:528–533. [DOI] [PubMed] [Google Scholar]
  • 9.EORTC Breast Cancer Cooperative Group, EORTC Radiotherapy Group, Bijker, et al. Breast-conserving treatment with or without radiotherapy in ductal carcinoma-in-situ: ten-year results of European Organisation for Research and Treatment of Cancer randomized phase III trial 10853--a study by the EORTC Breast Cancer Cooperative Group and EORTC Radiotherapy Group. J Clin Oncol 2006;24:3381-3387. [DOI] [PubMed] [Google Scholar]
  • 10.Emdin S, Granstrand B, Ringberg A, et al. SweDCIS: Radiotherapy after sector resection for ductal carcinoma in situ of the breast. Results of a randomised trial in a population offered mammography screening. Acta Oncologica 2006;45:536-543. [DOI] [PubMed] [Google Scholar]
  • 11.Bijker N, Peterse JL, Duchateau L, et al. Risk factors for recurrence and metastasis after breast-conserving therapy for ductal carcinoma-in-situ: analysis of European Organization for Research and Treatment of Cancer Trial 10853. J Clin Oncol 2001;19:2263–2271. [DOI] [PubMed] [Google Scholar]
  • 12.Fisher B, Dignam J, Wolmark N, et al. Tamoxifen in treatment of intraductal breast cancer: National Surgical Adjuvant Breast and Bowel Project B-24 randomised controlled trial. Lancet 1999;353:1993–2000. [DOI] [PubMed] [Google Scholar]
  • 13.Allred D, Bryant J, Land S, et al. Estrogen receptor expression as a positive marker of the effectiveness of tamoxifen in the treatment of DCIS: findings from NSABP Protocol B-24 [conference abstract]. San Antonio Breast Cancer Symposium, 2002. [Google Scholar]
  • 14.Houghton J, George WD, Cuzick J, et al. Radiotherapy and tamoxifen in women with completely excised ductal carcinoma in situ of the breast in the UK, Australia, and New Zealand: randomised controlled trial. Lancet 2003;362:95–102. [DOI] [PubMed] [Google Scholar]
  • 15.Goss PE, Ingle JN, Martino S, et al. A randomized trial of letrozole in postmenopausal women after five years of tamoxifen therapy for early-stage breast cancer. N Engl J Med 2003;349:1793–1802. [DOI] [PubMed] [Google Scholar]
  • 16.Baum M, Buzdar A, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early-stage breast cancer: results of the ATAC (Arimidex, Tamoxifen Alone or in Combination) trial efficacy and safety update analyses. Cancer 2003;98:1802–1810. [DOI] [PubMed] [Google Scholar]
  • 17.Baum M, Buzdar AU, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet 2002;359:2131–2139. [DOI] [PubMed] [Google Scholar]
  • 18.Howell A, Cuzick J, Baum M, et al. Results of the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial after completion of 5 years' adjuvant treatment for breast cancer. Lancet 2005;365:60–62. [DOI] [PubMed] [Google Scholar]
  • 19.Coombes RC, Hall E, Gibson LJ, et al. A randomized trial of exemestane after two to three years of tamoxifen therapy in postmenopausal women with primary breast cancer. N Engl J Med 2004;350:1081–1092. [DOI] [PubMed] [Google Scholar]
  • 20.Coombes RC, KilburnLS, Snowdon CF, et al. Survival and safety of exemestane versus tamoxifen after 2-3 year's tamoxifen treatment (Intergroup Exemestane Study): a randomised controlled trial. Lancet 2007;369:559–570. [DOI] [PubMed] [Google Scholar]
  • 21.Howell A. Effect of anastrozole of bone mineral density: 2-years results of the “Arimidex” (anastrozole), tamoxifen, alone or in combination (ATAC) trial. Breast Cancer Res Treat 2003;82:S27 (abstr 129). [Google Scholar]
  • 22.Coleman RE, Banks LM, GirgisSI, et al. Skeletal effects of exemestane on bone-mineral density, bone biomarkers, and fracture incidence in postmenopausal women with early breast cancer participating in the Intergroup Exemestane Study (IES): a randomised controlled study. Lancet Oncol 2007;8:119–127. [DOI] [PubMed] [Google Scholar]
  • 23.Boccardo F, Rubagotti A, Amoroso D, et al. Anastrozole appears to be superior to tamoxifen in women already receiving adjuvant tamoxifen treatment. Breast Cancer Res Treat 2003;82:S6 (abstr 3). [Google Scholar]
  • 24.Early Breast Cancer Trialists' Collaborative Group. Polychemotherapy for early breast cancer: an overview of the randomised trials. Lancet 1998;352:930–942. Search date not reported; primary sources individual patient data from trials that began before 1990, trials identified from lists from national cancer bodies, the International Cancer Research Data Bank, hand searches of conference proceedings and reference lists, and personal contact with investigators. 9752815 [Google Scholar]
  • 25.Bonadonna G, Zambeti M, Valagussa P. Sequential or alternating doxorubicin and CMF regimens in breast cancer with more than three positive nodes. JAMA 1995;273:542–547. [PubMed] [Google Scholar]
  • 26.Fisher B, Anderson S, Wickerham DL, et al. Increased intensification and total dose of cyclophosphamide in a doxorubicin–cyclophosphamide regimen for the treatment of primary breast cancer: findings from national surgical adjuvant breast and bowel project B-22. J Clin Oncol 1997;15:1858–1869. [DOI] [PubMed] [Google Scholar]
  • 27.Wood WC, Budman DR, Korzun AH. Dose and dose intensity of adjuvant chemotherapy for stage II, node-positive breast carcinoma. N Engl J Med 1994;330:1253–1259. [DOI] [PubMed] [Google Scholar]
  • 28.Early Breast Cancer Trialists' Collaborative Group. Tamoxifen for early breast cancer: an overview of the randomised trials. Lancet 1998;351:1451–1467. Search date not reported; primary sources individual patient data from trials that began before 1990, trials identified from lists from national cancer bodies, the International Cancer Research Data Bank, hand searches of conference proceedings and reference lists, and personal contact with investigators. 9605801 [Google Scholar]
  • 29.Swedish Breast Cancer Cooperative Group. Randomised trial of two versus five years of adjuvant tamoxifen for post-menopausal early stage breast cancer. J Natl Cancer Inst 1996;88:1543–1549. [DOI] [PubMed] [Google Scholar]
  • 30.Fisher B, Dignam J, Bryant J, et al. Five versus more than five years of tamoxifen therapy for breast cancer patients with negative lymph nodes and estrogen receptor-positive tumours. J Natl Cancer Inst 1996;88:1529–1542. [DOI] [PubMed] [Google Scholar]
  • 31.Stewart HJ, Forrest AP, Everington D, et al. Randomised comparison of 5 years of adjuvant tamoxifen with continuous therapy for operable breast cancer. Br J Cancer 1996;74:297–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Powles TJ, Hickish T, Kanis JA, et al. Effect of tamoxifen on bone mineral density measured by dual-energy x-ray absorptiometry in healthy premenopausal and postmenopausal women. J Clin Oncol 1996;14:78–84. [DOI] [PubMed] [Google Scholar]
  • 33.Swain SM. Tamoxifen: the long and short of it. J Natl Cancer Inst 1996;88:1510–1512. [DOI] [PubMed] [Google Scholar]
  • 34.Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 2005;353:1659–1672. [DOI] [PubMed] [Google Scholar]
  • 35.Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005;353:1673–1684. [DOI] [PubMed] [Google Scholar]
  • 36.Hortobagyi GN. Trastuzumab in the treatment of breast cancer. N Engl J Med 2005;353:1734–1736. [DOI] [PubMed] [Google Scholar]
  • 37.Bear HD, Anderson S, Brown A, et al. The effect on tumor response of adding sequential preoperative docetaxel to preoperative doxorubicin and cyclophosphamide: preliminary results from National Surgical Adjuvant Breast and Bowel Project Protocol B-27. J Clin Oncol 2003;21:4165–4174. [DOI] [PubMed] [Google Scholar]
  • 38.Henderson IC, Berry DA, Demetri GD, et al. Improved outcomes from adding sequential Paclitaxel but not from escalating Doxorubicin dose in an adjuvant chemotherapy regimen for patients with node-positive primary breast cancer.[comment]. J Clin Oncol 2003;21:976–983. [DOI] [PubMed] [Google Scholar]
  • 39.Martin M, Pienkowski T, Mackey J, et al. Adjuvant docetaxel for node-positive breast cancer.[see comment]. New England Journal of Medicine 2005;352:2302–2313. [DOI] [PubMed] [Google Scholar]
  • 40.Martin M, Rodríguez-Lescure A, Ruiz A, et al. Randomized phase III study of adjuvant chemotherapy for node positive breast cancer comparing 6 cycles of FE90C versus 4 cycles of FE90C followed by 8 weekly paclitaxel administrations: interim efficacy analysis of GEICAM 9906 Trial. San Antonio Breast Cancer Symposium 2005; Abstract 39. [Google Scholar]
  • 41.Roche H, Fumoleau P, Spielmann M, et al. Sequential adjuvant epirubicin-based and docetaxel chemotherapy for node-positive breast cancer patients: the FNCLCC PACS 01 Trial. J Clin Oncol 2006;24:5664–5671. [DOI] [PubMed] [Google Scholar]
  • 42.Jones SE, Savin MA, Holmes FA, et al. Phase III trial comparing doxorubicin plus cyclophosphamide with docetaxel plus cyclophosphamide as adjuvant therapy for operable breast cancer. J Clin Oncol 2006;24:5381–5387. [DOI] [PubMed] [Google Scholar]
  • 43.Early Breast Cancer Trialists' Collaborative Group. Effects of radiotherapy and surgery in early breast cancer: an overview of the randomised trials. N Engl J Med 1995;333:1444–1455. Search date not reported; primary sources individual patient data from trials that began before 1985, trials identified from lists from national cancer bodies, the International Cancer Research Data Bank, hand searches of conference proceedings and reference lists, and personal contact with investigators. [DOI] [PubMed] [Google Scholar]
  • 44.Fisher B, Jeong JH, Anderson S, et al. Twenty-five year follow-up of a randomized trial comparing radical mastectomy, total mastectomy, and total mastectomy followed by irradiation. N Engl J Med 2002;347:567–575. [DOI] [PubMed] [Google Scholar]
  • 45.Morris AD, Morris RD, Wilson JF, et al. Breast conserving therapy versus mastectomy in early stage breast cancer: a meta-analysis of 10 year survival. Cancer J Sci Am 1997;3:6–12. Search date 1995, primary source Medline. [PubMed] [Google Scholar]
  • 46.Veronesi U, Cascinelli N, Mariani L, et al. Twenty-year follow-up of a randomised study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 2002;347:1227–1232. [DOI] [PubMed] [Google Scholar]
  • 47.Fisher B, Andeson S, Bryant J, et al. Twenty-year follow-up of a randomised trial comparing total mastectomy, lumpectomy and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 2002;347:1233–1241. [DOI] [PubMed] [Google Scholar]
  • 48.Poggi MM, Danforth DN, Sciuto LC, et al. Eighteen-year results in the treatment of early breast carcinoma with mastectomy versus breast conservation therapy: the National Cancer Institute randomised trial. Cancer 2003;98:697–702. [DOI] [PubMed] [Google Scholar]
  • 49.Sacchini V, Luini A, Tana S, et al. Quantitative and qualitative cosmetic evaluation after conservative treatment for breast cancer. Eur J Cancer 1991;27:1395–1400. [DOI] [PubMed] [Google Scholar]
  • 50.Smitt NC, Nowels KW, Zdeblick MJ, et al. The importance of the lumpectomy surgical margin status in long-term results of breast conservation. Cancer 1995;76:259–267. [DOI] [PubMed] [Google Scholar]
  • 51.Wazer DE, DiPetrillo T, Schmidt-Ullrich R, et al. Factors influencing cosmetic outcome and complication risk after conservative surgery and radiotherapy for early-stage breast carcinoma. J Clin Oncol 1992;10:356–363. [DOI] [PubMed] [Google Scholar]
  • 52.Abner AL, Recht A, Vicini FA, et al. Cosmetic results after surgery, chemotherapy and radiation therapy for early breast cancer. Int J Radiat Oncol Biol Phys 1991;21:331–338. [DOI] [PubMed] [Google Scholar]
  • 53.Dewar JA, Benhamou S, Benhamou E, et al. Cosmetic results following lumpectomy axillary dissection and radiotherapy for small breast cancers. Radiother Oncol 1988;12:273–280. [DOI] [PubMed] [Google Scholar]
  • 54.Rochefordiere A, Abner A, Silver B, et al. Are cosmetic results following conservative surgery and radiation therapy for early breast cancer dependent on technique? Int J Radiat Oncol Biol Phys 1992;23:925–931. [DOI] [PubMed] [Google Scholar]
  • 55.Sneeuw KA, Aaronson N, Yarnold J, et al. Cosmetic and functional outcomes of breast conserving treatment for early stage breast cancer 1: comparison of patients' ratings, observers' ratings and objective assessments. Radiother Oncol 1992;25:153–159. [DOI] [PubMed] [Google Scholar]
  • 56.Ash DV, Benson EA, Sainsbury JR, et al. Seven year follow-up on 334 patients treated by breast conserving surgery and short course radical postoperative radiotherapy: a report of the Yorkshire Breast Cancer Group. Clin Oncol 1995;7:93–96. [DOI] [PubMed] [Google Scholar]
  • 57.Lindsey I, Serpell JW, Johnson WR, et al. Cosmesis following complete local excision of breast cancer. Aust N Z J Surg 1997;67:428–432. [DOI] [PubMed] [Google Scholar]
  • 58.Touboul E, Belkacemi Y, Lefranc JP, et al. Early breast cancer: influence of type of boost (electrons vs iridium-192 implant) on local control and cosmesis after conservative surgery and radiation therapy. Radiother Oncol 1995;34:105–113. [DOI] [PubMed] [Google Scholar]
  • 59.Halyard MY, Grado GL, Schomber PJ, et al. Conservative therapy of breast cancer: the Mayo Clinic experience. Am J Clin Oncol 1996;19:445–450. [DOI] [PubMed] [Google Scholar]
  • 60.Early Breast Cancer Trialists' Group. Ovarian ablation in early breast cancer: overview of the randomised trials. Lancet 1996;348:1189–1196. Search date not reported; primary sources individual patient data from trials that began before 1990, trials identified from lists from national cancer bodies, the International Cancer Research Data Bank, hand searches of conference proceedings and reference lists, and personal contact with investigators. 8898035 [Google Scholar]
  • 61.Early Breast Cancer Trialists' Collaborative Group (EBCTCG). Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and survival: an overview of the randomised trials. Lancet 2005;366:2087–2106. [DOI] [PubMed] [Google Scholar]
  • 62.Ford HT, Coombes RC, Gazet JC, et al. Long-term follow-up of a randomised trial designed to determine the need for irradiation following conservative surgery for the treatment of invasive breast cancer. Annals of Oncology 2006;17:401–408. [DOI] [PubMed] [Google Scholar]
  • 63.Lee HD, Yoon DS, Koo JY, et al. Breast conserving therapy in stage I & II breast cancer in Korea. Breast Cancer Res Treat 1997;44:193–199. [DOI] [PubMed] [Google Scholar]
  • 64.Fisher B, Bryant J, Dignam JJ, et al. Tamoxifen, radiation therapy, or both for prevention of ipsilateral breast tumor recurrence after lumpectomy in women with invasive breast cancers of one centimeter or less. J Clin Oncol 2002;20:4141–4149. [DOI] [PubMed] [Google Scholar]
  • 65.Steering Committee on Clinical Practice Guidelines for the Care and Treatment of Breast Cancer. A Canadian consensus document. Can Med Assoc J 1998;158(suppl 3):1–84. [PubMed] [Google Scholar]
  • 66.Ragaz J, Olivolto IA, Spinelli J, et al. Locoregional radiation therapy in patients with high-risk breast cancer receiving adjuvant chemotherapy: 20-year results of the British Columbia randomised trial. J Natl Cancer Inst 2005;97:116–126. [DOI] [PubMed] [Google Scholar]
  • 67.Overgaard M, Hansen PS, Overgaard J, et al. Postoperative radiotherapy in high-risk premenopausal women with breast cancer who receive adjuvant chemotherapy. N Engl J Med 1997;337:949–955. [DOI] [PubMed] [Google Scholar]
  • 68.Hojris I, Overgaard M, Christensen JJ, et al. Morbidity and mortality of ischaemic heart disease in high-risk breast-cancer patients after adjuvant postmastectomy systemic treatment with or without radiotherapy: analysis of DBCG 82b and 82c randomised trials. Radiotherapy Committee of the Danish Breast Cancer Cooperative Group. Lancet 1999;354:1425–1430. [DOI] [PubMed] [Google Scholar]
  • 69.Fyles AW, McCready DR, Manchul LA, et al. Tamoxifen with or without breast irradiation in women 50 years of age or older with early breast cancer. N Engl J Med 2004;351:963–970. [DOI] [PubMed] [Google Scholar]
  • 70.Hughes KS, Schnaper LA, Berry D, et al; Cancer and Leukemia Group B; Radiation Therapy Oncology Group; Eastern Cooperative Oncology Group. Lumpectomy plus tamoxifen with or without irradiation in women 70 years of age or older with early breast cancer. N Engl J Med 2004;351:971–977. [DOI] [PubMed] [Google Scholar]
  • 71.Whelan TJ, Levine M, Julian J, et al. The effects of radiation therapy on quality for life of women with breast carcinoma: results of a randomized trial. Cancer 2000;88:2260–2266. [PubMed] [Google Scholar]
  • 72.Rayan G, Dawson LA, Bezjak A, et al. Prospective comparison of breast pain in patients participating in a randomized trial of breast-conserving surgery and tamoxifen with or without radiotherapy. Int J Radiat Oncol Biol Phys 2003;55:154–161. [DOI] [PubMed] [Google Scholar]
  • 73.Winzer KH, Sauer R, Sauerbrei W, et al; German Breast Cancer Study Group. Radiation therapy after breast-conserving surgery; first results of a randomised clinical trial in patients with low risk of recurrence. Eur J Cancer 2004;40:998–1005. [DOI] [PubMed] [Google Scholar]
  • 74.Rutqvist L, Johansson H. Long-term follow-up of the Stockholm randomized trials of postoperative radiation therapy versus adjuvant chemotherapy among 'high risk' pre- and postmenopausal breast cancer patients. Acta Oncologica 2006;45:517–527. [DOI] [PubMed] [Google Scholar]
  • 75.Overgaard M, Jensen MB, Overgaard J, et al. Postoperative radiotherapy in high risk postmenopausal breast cancer patients given adjuvant tamoxifen: Danish Breast Cancer Cooperative Group DBCG 82c randomised trial. Lancet 1999;353:1641–1648. [DOI] [PubMed] [Google Scholar]
  • 76.Ghersi D, Simes J. Draft report of effectiveness of postmastectomy radiotherapy and risk factors for local recurrence in early breast cancer. Report to NHMRC National Breast Cancer Centre, Sydney, 1998. [Google Scholar]
  • 77.O'Rourke S, Gaba MH, Morgan D, et al. Local recurrence after simple mastectomy. Br J Surg 1994;81:386–389. [DOI] [PubMed] [Google Scholar]
  • 78.Fowble B, Gray R, Gilchrist K, et al. Identification of a subset of patients with breast cancer and histologically positive nodes who may benefit from postoperative radiotherapy. J Clin Oncol 1988;6:1107–1117. [DOI] [PubMed] [Google Scholar]
  • 79.Houghton J, Baum M, Haybittle JL. Role of radiotherapy following total mastectomy in patients with early breast cancer: the closed trials working party of the CRC breast cancer trials group. World J Surg 1994;18:117–122. [DOI] [PubMed] [Google Scholar]
  • 80.Gebski V, Lagleva M, Keech A, et al. Survival effects of post-mastectomy adjuvant radiation therapy using biologically equivalent doses: a clinical perspective. J Natl Cancer Inst 2006;98:26–38. [DOI] [PubMed] [Google Scholar]
  • 81.Mauriac L, Durand M, Avril A, et al. Effects of primary chemotherapy in conservative treatment of breast cancer patients with operable tumours larger than 3 cm: results of a randomised trial in a single centre. Ann Oncol 1991;2:347–354. [DOI] [PubMed] [Google Scholar]
  • 82.Scholl SM, Fourquet A, Asselain B, et al. Neoadjuvant versus adjuvant chemotherapy in premenopausal patients with tumours considered too large for breast conserving surgery: preliminary results of a randomised trial. Eur J Cancer 1994;30A:645–652. [DOI] [PubMed] [Google Scholar]
  • 83.Powles TJ, Hickish TF, Makris A, et al. Randomized trial of chemoendocrine therapy started before or after surgery for treatment of primary breast cancer. J Clin Oncol 1995;13:547–552. [DOI] [PubMed] [Google Scholar]
  • 84.Fisher B, Bryant J, Wolmark N, et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol 1998;16:2672–2685. [DOI] [PubMed] [Google Scholar]
  • 85.Van der Hage JA, van de Velde CJ, Julien JP, et al. Pre-operative chemotherapy in primary operable breast cancer: results from the European Organisation for Research and Treatment of Cancer Trial 10902. J Clin Oncol 2001;19:4224–4237. [DOI] [PubMed] [Google Scholar]
  • 86.Mauriac L, MacGrogan G, Avril A, et al. Neoadjuvant chemotherapy for operable breast carcinoma larger than 3 cm: a unicentre randomized trial with a 124-month median follow-up. Institut Bergonie Bordeaux Groupe Sein (IBBGS). Ann Oncol 1999;10:47–52. [DOI] [PubMed] [Google Scholar]
  • 87.Broet P, Scholl S, De la Rochrfordiere A, et al. Short and long term effects on survival in breast cancer patients treated by primary chemotherapy: an updated analysis of a randomised trial. Breast Cancer Res Treat 1999;58:151–156. [DOI] [PubMed] [Google Scholar]
  • 88.Makris A, Powles TJ, Ashley SE, et al. A reduction in the requirements for mastectomy in a randomized trial of neoadjuvant chemoendocrine therapy in primary breast cancer. Ann Oncol 1998;9:1179–1184. [DOI] [PubMed] [Google Scholar]
  • 89.Avril A, Faucher A, Bussieres E, et al. Results of 10 years of a randomized trial of neoadjuvant chemotherapy in breast cancers larger than 3 cm. Chirurgie 1998;123:247–256. [In French] [DOI] [PubMed] [Google Scholar]
  • 90.Chetty U, Jack W, Prescott RJ, et al. Management of the axilla in operable breast cancer treated by breast conservation: a randomised controlled trial. Br J Surg 2000;87:163–169. [DOI] [PubMed] [Google Scholar]
  • 91.Browning C, Redman S, Pillar C, et al. NHMRC National Breast Cancer Centre, Sydney 1998. Lymphoedema: prevalence risk factors and management: a review of research. Search date 1996; primary sources Medline, hand searches of article references, personal contact with key resources of article references, and personal contact with key resources. [Google Scholar]
  • 92.Axelsson CK, Mouridzsen HT, Zedeler K. Axillary dissection at level I and II lymph nodes is important in breast cancer classification. Eur J Cancer 1992;28A:1415–1418. [DOI] [PubMed] [Google Scholar]
  • 93.Kiricuta CI, Tausch J. A mathematical model of axillary lymph node involvement based on 1446 complete axillary dissections in patients with breast carcinoma. Cancer 1992;69:2496–2501. [DOI] [PubMed] [Google Scholar]
  • 94.Steele RJC, Forrest APM, Gibson R, et al. The efficacy of lower axillary sampling in obtaining lymph node status in breast cancer: a controlled randomised trial. Br J Surg 1985;72:368–369. [DOI] [PubMed] [Google Scholar]
  • 95.Therasse P, Mauriac L, Welnicka-Jaskiewicz M, et al. Final results of a randomized phase III trial comparing cyclophosphamide, epirubicin, and fluorouracil with a dose-intensified epirubicin and cyclophosphamide + filgrastim as neoadjuvant treatment in locally advanced breast cancer: an EORTC–NCIC–SAKK multicenter study. J Clin Oncol 2003;21:843–850. [DOI] [PubMed] [Google Scholar]
  • 96.Buzdar AU, Singletary SE, Theriault RL, et al. Prospective evaluation of paclitaxel versus combination chemotherapy with fluorouracil, doxorubicin, and cyclophosphamide as neoadjuvant therapy in patients with operable breast cancer. J Clin Oncol 1999;17:3412–3417. [DOI] [PubMed] [Google Scholar]
  • 97.Cocconi G, Bisagni G, Ceci G, et al. Three new active cisplatin-containing combinations in the neoadjuvant treatment of locally advanced and locally recurrent breast carcinoma: a randomized Phase II trial. Breast Cancer Res Treat 1999;56:125–132. [DOI] [PubMed] [Google Scholar]
  • 98.Smith IC, Heys SD, Hutcheon A, et al. Neoadjuvant chemotherapy in breast cancer: significantly enhanced response with docetaxel. J Clin Oncol 2002;20:1456–1466. [DOI] [PubMed] [Google Scholar]
  • 99.D'Orazio AI, O'Shaughnessy J, Seidman AD. Neoadjuvant docetaxel augments the efficacy of preoperative docetaxel/cyclophosphamide in operable breast cancer: first results of NSABP-27. Clin Breast Cancer 2002;2:266–268. [Google Scholar]
  • 100.Takatsuka Y, Yayoi E, Kobayashi T, et al. Neoadjuvant intra-arterial chemotherapy in locally advanced breast cancer: a prospective randomised study. Jpn J Clin Oncol 1994;24:20–25. [PubMed] [Google Scholar]
  • 101.Webb A, Smith IE, Ahern R. A randomised Phase II trial of pre-operative navelbine/epirubicin (NE) versus navelbine/mitozantrone (NM) versus adriamycin/cyclophosphamide (AC) for early breast cancer. Eur J Cancer 2001;37:174. [Google Scholar]
  • 102.Australian Safety and Efficacy Register of New Interventional Procedures – Surgical, and the Royal Australasian College of Surgeons. A systematic review of intraoperative radiotherapy in early stage breast cancer. 2002, ASERNIP-S Report No. 27. Adelaide, South Australia. Search date 2002; primary sources OVID PreMedline, OVID Medline, Current Contents, Cochrane Library, Embase, UK National Research Register, NIH Clinical Trials Gov, PubMed, and HTA database. [Google Scholar]
  • 103.Ribeiro GG Magee B, Swindell R, et al. The Christie Hospital breast conservation trial: an update at 8 years from inception. Clinical Oncology 1993;5:278–283. [DOI] [PubMed] [Google Scholar]
  • 104.Magee B, Swindell R, Harris M, et al. Prognostic factors for breast recurrence after conservative breast surgery and radiotherapy: results from a randomised trial. Radiother Oncol 1996;39:223–227. [DOI] [PubMed] [Google Scholar]
  • 105.Dodwell DJ, Dyker K Brown J, et al. A randomised study of whole-breast vs tumour-bed irradiation after local excision and axillary dissection for early breast cancer. Clin Oncol 2005;17:618–622. [DOI] [PubMed] [Google Scholar]
  • 106.Coles CE, Moody AM, Wilson CB, et al. Reduction of radiotherapy-induced late complications of early breast cancer: the role of intensity-modulation radiation therapy and partial breast irradiation. Part II – Radiotherapy strategies to reduce radiation-induced late effects. Clin Oncol 2005;17:98–110. [DOI] [PubMed] [Google Scholar]
  • 107.Kaija H, Maunu P. Tangential breast irradiation with or without internal mammary chain irradiation: results of a randomised trial. Radiother Oncol 1995;36:172–176. [DOI] [PubMed] [Google Scholar]
  • 108.Gyenes G, Rutqvist LE, Liedberg A, et al. Long-term cardiac morbidity and mortality in a randomized trial of pre- and postoperative radiation therapy versus surgery alone in primary breast cancer. Radiother Oncol 1998;48:185–190. [DOI] [PubMed] [Google Scholar]
  • 109.Handley R. Carcinoma of the breast. Ann R Coll Surg Engl 1975;57:59–66. [PMC free article] [PubMed] [Google Scholar]
  • 110.Veronesi U, Cascinelli NM, Bufalino R, et al. Risk of internal mammary lymph node metastases and its relevance on prognosis in breast cancer patients. Ann Surg 1983;198:681–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Veronesi U, Valagussa P. Inefficacy of internal mammary node dissection in breast cancer surgery. Cancer 1981;47:170–175. [DOI] [PubMed] [Google Scholar]
  • 112.Early Breast Cancer Trialists' Collaborative Group. Favourable and unfavourable effects on long-term survival of radiotherapy for early breast cancer: an overview of the randomised trials. Lancet 2000;355:1757–1770. Search date not reported; primary sources individual patient data from trials that began before 1990, trials identified from lists from national cancer bodies, the International Cancer Research Data Bank, hand searches of conference proceedings and reference lists, and personal contact with investigators. 10832826 [Google Scholar]
  • 113.Cuzick J, Stewart H, Rutqvist L, et al. Cause-specific mortality in long term survivors of breast cancer who participated in trials of radiotherapy. J Clin Oncol 1994;12:447–453. Search date not reported; primary source cause specific mortality data from unconfounded randomised trials began before 1975 (trial identification methods not reported). [DOI] [PubMed] [Google Scholar]
  • 114.Bates T, Evans RGB. Report of the Independent Review commissioned by The Royal College of Radiologists into brachial plexus neuropathy following radiotherapy for breast cancer. London: Royal College of Radiologists, 1995. [DOI] [PubMed] [Google Scholar]
  • 115.Veronesi U, Paganelli G, Viale G, et al. A randomized comparison of sentinel-node biopsy with routine axillary dissection in breast cancer. N Engl J Med 2003;349:546–553. [DOI] [PubMed] [Google Scholar]
  • 116.Purushotham AD, Upponi S, Klevesath MB, et al. Morbidity after sentinel lymph node biopsy in primary breast cancer: results from a randomized controlled trial. J Clin Oncol 2005;23:4312–4321. [DOI] [PubMed] [Google Scholar]
  • 117.King TA, Fey JV, Van Zee KJ, et al. A prospective analysis of the effect of blue-dye volume on sentinel lymph node mapping success and incidence of allergic reaction in patients with breast cancer. Ann Surg Oncol 2004;11:535–541. [DOI] [PubMed] [Google Scholar]
  • 118.Lyman GH, Giuliano AE, Somerfield MR, et al. American Society of Clinical Oncology Guideline Recommendations for sentinel lymph node biopsy in early-stage breast cancer. J Clin Oncol 2005;23:7703–7720. [DOI] [PubMed] [Google Scholar]
  • 119.Kim T, Giuliano AE, Lyman GH. Lymphatic mapping and sentinel lymph node biopsy in early-stage breast carcinoma. Cancer 2006;106:4–16. [DOI] [PubMed] [Google Scholar]
  • 120.Julian TB, Krag D, Brown A, et al. Preliminary technical results of NSABP B-32, a randomized phase III clinical trial to compare sentinel node resection to conventional axillary dissection in clinically node-negative breast cancer patients. Proc SABCS 2004 (abstr 14). http://www.abstracts2view.com/sabcs/view.php?nu=BCS4L_253 (last accessed: 23 August 2005). [Google Scholar]
  • 121.Mansel RE, Goyal A, Necombe RG, et al: Objective assessment of lymphedema, shoulder function and sensory deficit after sentinel node biopsy for invasive breast cancer: ALMANAC trial. Breast Cancer Res Treat 2004;88:S12 (abstr 15). [Google Scholar]
  • 122.Goyal A, Newcombe RG, Mansel RE. Axillary Lymphatic Mapping Against Nodal Axillary Clearance (ALMANAC) Trialists Group. Clinical relevance of multiple sentinel nodes in patients with breast cancer. Br J Surg 2005;92:438–442. [DOI] [PubMed] [Google Scholar]
  • 123.Farquhar C, Marjoribanks J, Basser R, et al. High dose chemotherapy and autologous bone marrow or stem cell transplantation versus conventional chemotherapy for women with metastatic breast cancer. The Cochrane Database of Systematic Reviews 2006, Issue 4. Chichester, UK: John Wiley & Sons, Ltd. Search date 2004; primary sources Cochrane Breast Cancer Group specialised register, Cochrane Controlled Trials Register, Medline, Embase, Psychinfo, Cinahl, websites of cooperative research groups and American Society of Clinical Oncologists, and reference lists. [Google Scholar]
  • 124.Papaioannou A, Lissaios B, Vasilaros S, et al. Pre- and post-operative chemoendocrine treatment with or without post-operative radiotherapy for locally advanced breast cancer. Cancer 1983;51:1284–1290. [DOI] [PubMed] [Google Scholar]
  • 125.Olson JE, Neuberg D, Pandya KJ, et al. The role of radiotherapy in the management of operable locally advanced breast carcinoma: results of a randomised trial by the Eastern Co-operative Oncology Group. Cancer 1997;79:1138–1149. [PubMed] [Google Scholar]
  • 126.Perloff M, Lesnick G J, Korzun A, et al. Combination chemotherapy with mastectomy or radiotherapy for stage III breast carcinoma: a Cancer and Leukaemia Group B Study. J Clin Oncol 1988;6:261–269. [DOI] [PubMed] [Google Scholar]
  • 127.De Lena M, Varini M, Zucali R, et al. Multimodal treatment for locally advanced breast cancer. Results of chemotherapy–radiotherapy versus chemotherapy–surgery. Cancer Clin Trials 1981;4:229–236. [PubMed] [Google Scholar]
  • 128.Willsher PC, Robertson JF, Armitage NC, et al. Locally advanced breast cancer: long term results of a randomised trial comparing primary treatment with tamoxifen or radiotherapy in post-menopausal women. Eur J Surg Oncol 1996;22:34–37. [DOI] [PubMed] [Google Scholar]
  • 129.Bartelink H, Rubens RD, Van der Schueren E, et al. Hormonal therapy prolongs survival in irradiated locally advanced breast cancer: a European Organisation for Research and Treatment of Cancer randomised Phase III trial. J Clin Oncol 1997;15:207–215. [DOI] [PubMed] [Google Scholar]
  • 130.Koning C, Hart G. Long term follow up of a randomised trial on adjuvant chemotherapy and hormonal therapy in locally advanced breast cancer. Int J Radiat Oncol Biol Phys 1998;41:397–400. [DOI] [PubMed] [Google Scholar]
  • 131.Rodger A, Jack WJL, Hardman PDJ, et al. Locally advanced breast cancer: report of a Phase II study and subsequent Phase III trial. Br J Cancer 1992;65:761–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Willsher PC, Robertson JF, Chan SY, et al. Locally advanced breast cancer: early results of a randomised trial of multimodal therapy versus initial hormone therapy. Eur J Cancer 1997;33:45–49. [DOI] [PubMed] [Google Scholar]
  • 133.Tan SM, Cheung KL, Willsher PC, et al. Locally advanced primary breast cancer: medium term results of a randomised trial of multimodal therapy versus initial hormone therapy. Eur J Cancer 2001;37:2331–2338. [DOI] [PubMed] [Google Scholar]
  • 134.Deo SV, Bhutani M, Shukla NK, et al. Randomized trial comparing neo-adjuvant versus adjuvant chemotherapy in operable locally advanced breast cancer (T4 b N0-2 M0) J Surg Oncol 2003;84:192–197. [DOI] [PubMed] [Google Scholar]
BMJ Clin Evid. 2007 Dec 4;2007:0102.

Radiotherapy

Summary

TREATMENT SUCCESS Compared with no radiotherapy: Radiotherapy after breast-conserving surgery for ductal carcinoma in situ is more effective at 2, 4, 8 and at 10 years at reducing local recurrence and invasive carcinoma ( high-quality evidence ). Compared with tamoxifen plus radiotherapy: Radiotherapy alone and radiotherapy plus tamoxifen seem to be equally effective at 1 year at reducing invasive or ductal carcinoma in situ events in women undergoing local excision ( moderate-quality evidence ). MORTALITY Compared with no radiotherapy: Radiotherapy after breast-conserving surgery for ductal carcinoma in situ is no more effective at 2, 4, 8 or at 10-years at prolonging survival (moderate-quality evidence). ADVERSE EFFECTS Radiotherapy has been associated with an increased risk in contralateral breast cancer at 4 years.

Benefits

Radiotherapy versus no radiotherapy:

We found one systematic review (search date 2001, 2 RCTs), which compared radiotherapy versus no radiotherapy after surgery for ductal carcinoma in situ (DCIS). The first RCT identified by the review (814 women) found no significant difference in survival at 8 years with radiotherapy compared with no radiotherapy (survival 95% with radiotherapy v 94% with no radiotherapy, reported as not significant). It found significant reductions in risk of local recurrence, recurrent DCIS, and invasive carcinoma with radiotherapy compared with no radiotherapy (local recurrence: 12.1% with radiotherapy v 26.8% with no radiotherapy, P less than 0.0005; risk of recurrent DCIS: 8.2% with radiotherapy v 13.4% with no radiotherapy, P = 0.007; risk of invasive carcinoma: 3.9% with radiotherapy v 13.4% with no radiotherapy, P less than 0.0001). The second RCT identified by the review (1002 women) found that surgery plus radiotherapy significantly increased the proportion of women free of local recurrence at 4 years compared with surgery alone (91% with surgery plus radiotherapy v 84% with surgery alone; P = 0.005; HR 0.62, 95% CI 0.44 to 0.87). More women were free of DCIS recurrence after 4 years with radiotherapy, but the difference was not significant (95% with surgery plus radiotherapy v 92% with surgery alone; HR 0.65, 95% CI 0.43 to 1.03). Added radiotherapy significantly reduced invasive recurrence (96% with surgery plus radiotherapy v 92% with surgery alone; HR 0.60, 95% CI 0.37 to 0.97). There was no significant difference between groups in survival at 4 years (survival rates presented graphically; P = 0.94). A subsequent update of this RCT at 10 years' median follow-up found persistence of the benefit of radiotherapy in significantly reducing local recurrence (85% local control with surgery plus radiotherapy v 74% with surgery alone; P less than 0.0001; HR 0.53, 95% CI 0.4 to 0.7). It found no significant difference between groups in survival. One subsequent RCT included 1046 women with screen-detected DCIS. It found that, after a minimum follow-up period of 2 years, surgery plus post-operative radiotherapy significantly reduced recurrence rates compared with surgery alone (local recurrence: 8% with surgery plus radiotherapy v 22% with surgery alone; P less than 0.0001; HR 0.33, 95% CI 0.24 to 0.47). It found no significant difference between groups in survival.

Radiotherapy versus radiotherapy plus tamoxifen:

See benefits of tamoxifen plus radiotherapy.

Harms

Radiotherapy versus no radiotherapy:

The second RCT included in the review found an increase in contralateral breast cancer associated with radiotherapy at 4 years (3% with surgery plus radiotherapy v 1% with surgery alone; HR 2.57, 95% CI 1.24 to 5.33).However, the update at 10-years' follow-up found that there was no longer a statistically significant increase between groups in the risk of contralateral breast cancer (8% with surgery plus radiotherapy v 4% with surgery alone; HR 1.41, 95% CI 0.87 to 2.30).The subsequent RCT did not report on harms.

Radiotherapy versus radiotherapy plus tamoxifen:

See harms of tamoxifen plus radiotherapy.

Comment

Clinical guide:

One RCT has reported a subgroup analysis to assess what impact radiotherapy has after breast-conserving surgery for various pathological sub-groups. The study found that relative risk reduction of radiotherapy was homogeneous across all groups (i.e. that there was a similar hazard reduction for all pathological subgroups, which is statistically significantly different). However, for some subgroups the overall risk of local recurrence at 4-years' follow-up was low (clinging or micropapillary pattern: 3% recurrence with surgery plus radiotherapy v 8% with surgery alone; well differentiated lesions: 7% recurrence with surgery plus radiotherapy v 13% with surgery alone). In good-prognosis subgroups, a case can be made to omit radiotherapy when the overall local control benefit for radiotherapy is low, although this should be done in consultation with the patient, explaining the pros and cons of treatment and the risk of recurrence. Patients undergoing breast-conserving surgery should be referred for discussion on the pros and cons of whole-breast radiotherapy, unless in the context of a clinical trial.

Substantive changes

Radiotherapy (under question on interventions after breast-conserving surgery for ductal carcinoma in situ) One RCT added and one extended 10-year follow-up of a previously included RCT added; benefits and harms data enhanced, categorisation of radiotherapy (reduced recurrence) unchanged (Likely to be beneficial).

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Tamoxifen plus radiotherapy

Summary

TREATMENT SUCCESS Compared with placebo: Adjuvant treatment with tamoxifen seems to be more effective at 6 years at reducing invasive ipsilateral or contralateral breast cancers and breast cancer events in women who have been treated with wide excision and radiotherapy ( moderate-quality evidence ). However, this benefit may be limited to women with oestrogen receptor-positive tumours. Compared with radiotherapy alone: Radiotherapy plus tamoxifen, and radiotherapy alone seem to be equally effective at 1 year at reducing invasive or ductal carcinoma in situ events (moderate-quality evidence). MORTALITY Compared with placebo: Adjuvant treatment with tamoxifen seems to be no more effective at 6 years at reducing overall survival in women who have been treated with wide excision and radiotherapy (moderate-quality evidence).

Benefits

Tamoxifen plus radiotherapy versus placebo:

We found no systematic review but found one RCT (2 publications). The RCT (1804 women with ductal carcinoma in situ [DCIS] treated with wide excision and radiotherapy) compared adjuvant treatment with tamoxifen 20 mg daily versus placebo for 5 years. At a median follow-up of 74 months, there were significantly fewer breast cancer events with tamoxifen than with placebo, and significantly fewer invasive ipsilateral or contralateral breast cancers (breast cancer events: OR 0.63, 95% CI 0.47 to 0.83; invasive ipsilateral or contralateral breast cancers: OR 0.57, 95% CI 0.38 to 0.85). However, there was no significant difference in overall survival (RR 0.88, 95% CI 0.33 to 2.28). A subsequent subgroup analysis found that only women with oestrogen receptor-positive DCIS derived a benefit from tamoxifen.

Tamoxifen plus radiotherapy versus radiotherapy alone:

We found no systematic review but found one RCT. The RCT (1694 women having local excision) compared four treatments in a factorial design: no adjuvant treatment, tamoxifen alone, radiotherapy alone, and tamoxifen plus radiotherapy (See comment on radiotherapy). It found no significant difference between tamoxifen plus radiotherapy and radiotherapy alone in ipsilateral invasive disease, ipsilateral DCIS, and invasive or DCIS events after median follow-up of 1 year (523 women in comparison; ipsilateral invasive disease: HR 1.25, 95% CI 0.43 to 3.61; ipsilateral DCIS: HR 0.75, 95% CI 0.28 to 2.02; invasive or DCIS: 3% in both groups; HR 0.95, 95% CI 0.51 to 1.77).

Harms

Tamoxifen plus radiotherapy versus radiotherapy plus placebo:

The RCT found a higher, but non-significant rate of endometrial cancers associated with tamoxifen (RR 3.4, 95% CI 0.6 to 33.4).

Tamoxifen plus radiotherapy versus radiotherapy alone:

The RCT did not report results comparing harms of tamoxifen plus radiotherapy versus radiotherapy alone. The risks of congenital malformations, or late teratogenic manifestations in adulthood are unknown, and women should be offered choices in all cases concerning continuing with pregnancy or termination. Women are conventionally advised to stop tamoxifen before attempting pregnancy.

Comment

Clinical guide:

With conflicting results from the two published RCTs, it is difficult to recommend the addition of tamoxifen to breast-conserving surgery and radiotherapy in the management of DCIS. Tamoxifen might be considered appropriate if the patient has other risk factors for recurrence — such as they choose to omit radiotherapy, or they have a family history.Tamoxifen does not seem to improve survival if given with radiotherapy for breast conservation, but may reduce local disease recurrence in oestrogen receptor-positive DCIS.

Substantive changes

Tamoxifen plus radiotherapy (under question on interventions after breast-conserving surgery for ductal carcinoma in situ) Existing evidence re-evaluated. Tamoxifen plus radiotherapy (reduced recurrence in women with oestrogen receptor-positive tumours) recategorised from Likely to be beneficial to Unknown effectiveness.

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Adjuvant aromatase inhibitors (anastrozole, letrozole, exemestane)

Summary

TREATMENT SUCCESS Compared with placebo: Letrozole seems to be more effective at 5 years at improving estimated disease-free survival rates in postmenopausal women who have completed 5 years of postoperative tamoxifen therapy ( moderate-quality evidence ). Compared with tamoxifen: Adjuvant aromatase inhibitors (anastrozole and exemestane) are more effective at reducing breast cancer events and at improving disease-free survival in postmenopausal women with early breast cancer (moderate-quality evidence). MORTALITY Compared with placebo: Letrozole seems to be no more effective at 5 years at prolonging estimated overall survival in postmenopausal women who have completed 5 years of postoperative tamoxifen therapy (moderate-quality evidence). Compared with tamoxifen: Exemestane seems to be modestly more effective at improving overall survival in women with oestrogen receptor-positive breast cancer (moderate-quality evidence). ADVERSE EVENTS Adjuvant aromatase inhibitors (letrozole, anastrozole, and exemestane) are more likely to increase the incidence of arthralgia and fractures compared with placebo or tamoxifen, but are less likely to cause thromboembolic and endometrial events compared with tamoxifen.

Benefits

Adjuvant aromatase inhibitors versus placebo:

We found no systematic review but found one RCT. The RCT (5187 postmenopausal women who had completed 5 years of postoperative tamoxifen treatment; see comment below) compared letrozole (2.5 mg orally daily) versus placebo. It found that the estimated disease-free survival rate at 4 years was greater with letrozole versus placebo, although there was no significant difference in estimated overall survival at 4 years (disease-free survival: 93% with letrozole v 87% with placebo, ARR 6.0, 95% CI 2.0 to 10.1; overall survival: 96.0% with letrozole v 93.6% with placebo, ARR 2.4, 95% CI –0.9 to +5.6).

Adjuvant aromatase inhibitors versus tamoxifen:

We found no systematic review but found two RCTs (4 publications), which compared adjuvant aromatase inhibitors versus tamoxifen. The first RCT (3 publications, 9366 postmenopausal women with operable, invasive, non-metastatic breast cancer, who had completed primary surgery and chemotherapy; see comment below) compared anastrozole versus tamoxifen versus a combination of anastrozole and tamoxifen. It found that anastrozole increased disease-free survival at all follow-up points compared with tamoxifen alone (HR at 3 years: 0.83, 95% CI 0.71 to 0.96, P = 0.013; HR at 4 years: 0.86, 95% CI 0.76 to 0.99, P = 0.03; HR at 5 years: 0.87, 95% CI 0.78 to 0.97, P = 0.01). Anastrozole also prolonged time to recurrence, and reduced time to distant metastases at 5 years (time to recurrence: 402 days with anastrozole v 498 days with tamoxifen, HR 0.87, 95% CI 0.78 to 0.97, P = 0.01; distant metastases: 324 days with anastrozole v 375 days with tamoxifen, RR 0.79, 95% CI 0.70 to 0.90, P = 0.0005). It also found that contralateral breast cancer was reduced with anastrozole versus tamoxifen (35 with anastrozole v 59 with tamoxifen; P = 0.01). A combination of tamoxifen and anastrozole did not significantly improve outcomes versus tamoxifen alone at 3 or 4 years (3 years: HR 1.02, 95% CI 0.89 to 1.18, P = 0.8; 4 years: HR 1.08, 95% CI 0.98 to 1.24, P = 0.3). The second RCT (4742 recurrence-free postmenopausal women, who had completed 2–3 years of tamoxifen treatment following surgery for primary, oestrogen receptor-positive breast cancer) compared exemestane versus continuing tamoxifen treatment for the recommended 5 years. It found that exemestane significantly improved disease-free and breast cancer-free survival, and reduced risk of contralateral breast cancer at a median follow-up of 31 months compared with tamoxifen (disease-free survival: HR 0.68, 95% CI 0.56 to 0.82, P less than 0.001, see comment below; breast cancer-free survival: HR 0.63, 95% CI 0.51 to 0.77, P = 0.00001; reduced risk of contralateral breast cancer: HR 0.44, 95% CI 0.20 to 0.98, P = 0.04). It found no significant difference in overall survival between treatments (HR 0.88, 95% CI 0.67 to 1.16; P = 0.41) at the time of analysis.However, subsequent follow-up of this RCT has confirmed a survival advantage for women with oestrogen receptor-positive tumours (HR 0.83, 95% CI 0.69 to 1.00, P = 0.05).

Harms

Adjuvant aromatase inhibitors versus placebo:

The RCT comparing letrozole versus placebo found that hot flushes, arthritis, arthralgia, and myalgia were more common with letrozole (P less than 0.05 for each). Vaginal bleeding was more common with placebo (P = 0.01). The RCT found non-significant increases in osteoporosis and fracture rates with letrozole versus placebo (osteoporosis: 5.8% with letrozole v 4.5% with placebo, P = 0.07; fracture rate: 3.6% with letrozole v 2.9% with placebo, P = 0.24).

Adjuvant aromatase inhibitors versus tamoxifen:

Treatment with anastrozole reduced the incidence of endometrial cancer, thromboembolic events, ischaemic cerebrovascular events, vaginal bleeding, hot flushes, and vaginal discharge compared with tamoxifen, but increased fracture rate and arthralgia (endometrial cancer: OR 0.29, 95% CI 0.11 to 0.80, P = 0.02; thromboembolic events: OR 0.61, 95% CI 0.47 to 0.80, P = 0.0004; ischaemic cerebrovascular events: OR 0.70, 95% CI 0.50 to 0.97, P = 0.03; vaginal bleeding: OR 0.50, 95% CI 0.41 to 0.61, P less than 0.0001; hot flushes: OR 0.80, 95% CI 0.73 to 0.89, P less than 0.0001; vaginal discharge: OR 0.24, 95% CI 0.19 to 0.30, P less than 0.0001; fracture rate: OR 1.49, 95% CI 1.25 to 1.77, P less than 0.0001; arthralgia: OR 1.32, 95% CI 1.19 to 1.47, P less than 0.001). Increases in adverse musculoskeletal events are probably best explained as being due to a “second menopause” induced by lower levels of oestrogen, and appear to be similar to the aches and pains associated with the normal menopause. In a subset of 308 women, there was a median bone mineral loss over 2 years of 4.1% in the lumbar spine and 3.9% in the hip on anastrozole, compared with small increases in bone mineral density on tamoxifen. With a median follow-up of 68 months, this has translated into an increase of fractures from 7.7% to 11.0%, or a 50% relative increase. Similarly, exemestane reduced thromboembolic events, vaginal bleeding, and muscle cramps compared with tamoxifen, but was associated with a higher incidence of arthralgia and diarrhoea (thromboembolic events: 1.0% with exemestane v 1.9% with tamoxifen, P = 0.003; vaginal bleeding: 4.0% with exemestane v 5.5% with tamoxifen, P = 0.05; muscle cramps: 2.8% with exemestane v 4.4% with tamoxifen, P less than 0.001; arthralgia: 5.4% with exemestane v 3.6% with tamoxifen, P = 0.01; diarrhoea: 4.3% with exemestane v 2.3% with tamoxifen, P less than 0.001).In this RCT, a further report found that the increase in survival by the switch therapy was achieved at the expense of some detriment to skeletal health, as bone mineral density was significantly lowered in the exemestane group compared with the tamoxifen group (bone mineral density lowered at the lumbar spine: 2.7%, 95% CI 2% to 3.4%; P less than 0.0001).

Comment

Adjuvant aromatase inhibitors versus placebo:

The RCT comparing letrozole versus placebo was terminated after a median follow-up of 2.4 years owing to the efficacy of the aromatase inhibitor.

Adjuvant aromatase inhibitors versus tamoxifen:

Assessment of hormone receptor status was not routinely performed in some countries, so some women included in the RCT comparing anastrozole versus tamoxifen were hormone receptor-negative. In the RCT comparing continuing tamoxifen treatment with exemestane, adjusting for oestrogen receptor status, nodal status, chemotherapy, and use of HRT did not affect the results,although a significant survival benefit was observed with switching in the oestrogen receptor positive group, at the expense of some loss in bone mineral density.Anastrozole, letrozole, and exemestane, all third-generation aromatase inhibitors, seem to have greater specificity compared with tamoxifen, with a more favourable adverse-effect profile. One small RCT (426 people, 30 months' follow-up) suggests that anastrozole is superior to tamoxifen in women already receiving adjuvant tamoxifen.

Clinical guide:

Aromatase inhibitors have an increased disease-free (but not overall) survival for oestrogen receptor-positive cancers compared with tamoxifen, and have a different safety profile. Aromatase inhibitors may be advantageous for women with a higher risk of disease recurrence, but caution regarding bone density and cardiovascular events remains. The randomised data suggest that there are now three possible strategies that may be better, in postmenopausal women with oestrogen receptor-positive breast cancer, than 5 years of tamoxifen: firstly, tamoxifen for 5 years followed by letrozole for 5 years; secondly, anastrazole for 5 years; thirdly, tamoxifen for 2 to 3 years followed by exemestane for 2 to 3 years.

Substantive changes

Adjuvant aromatase inhibitors (under question on treatments for primary operable breast cancer) Two further follow-up reports of a previously included RCT added; benefits and harms data enhanced, categorisation unchanged (Beneficial).

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Adjuvant combination chemotherapy

Summary

TREATMENT SUCCESS Compared with no chemotherapy: Adjuvant combination chemotherapy is more effective at reducing recurrence and is independent of nodal or menopausal status ( moderate-quality evidence ). Different treatment durations compared with each other: Longer regimens and shorter regimens are equally effective at reducing recurrence rates ( high-quality evidence ). Different doses compared with each other: Enhanced and standard dose chemotherapy regimens seem to be equally effective at improving disease-free survival (moderate-quality evidence). MORTALITY Compared with no chemotherapy: Adjuvant combination chemotherapy is more effective at reducing all-cause death and is independent of nodal or menopausal status (moderate-quality evidence). Different treatment durations compared with each other: Longer regimens and shorter regimens are equally effective at prolonging survival (high-quality evidence). Different doses compared with each other: Enhanced and standard dose chemotherapy regimens seem to be equally effective at prolonging survival (moderate-quality evidence). ADVERSE EFFECTS Chemotherapy has been associated with fatigue, nausea and vomiting, hair loss, bone marrow suppression, neuropathy, and gastrointestinal disturbance. Chemotherapy may impair fertility and ovarian function.

Benefits

Adjuvant combination chemotherapy versus no chemotherapy:

We found one systematic review (search date not reported, 47 RCTs, 18,000 women), which compared prolonged combination chemotherapy versus no chemotherapy. Chemotherapy was associated with significantly lower rates of any kind of recurrence and death from all causes (recurrence: women aged under 50 years, OR 0.65, 95% CI 0.61 to 0.69; women aged 50–69 years, OR 0.80, 95% 0.72 to 0.88; death from all causes: women aged under 50 years, OR 0.73, 95% CI 0.68 to 0.78; women aged 50–69 years, OR 0.89, 95% CI 0.86 to 0.92). Proportional benefits were similar for women with node-negative and node-positive disease. Survival at 10 years according to nodal and age group is summarised (see table 2 ).

Table 2.

Ten year survival with combination chemotherapy versus placebo, according to nodal and age/menopausal status: results of a systematic review of RCTs (see text).

  Control (%) Chemotherapy (%) Absolute benefit (%) SD (%) Significance (two sided)
Age under 50 years
Node +ve 41.4 53.8 +12.4 2.4 P less than 0.0001
Node –ve 71.9 77.6 +5.7 2.1 P = 0.01
           
Age 50–69 years
Node +ve 46.3 48.6 +2.3 1.3 P = 0.001
Node –ve 64.8 71.2 +6.4 2.3 P = 0.0025

SD, standard deviation.

Harms

Acute adverse effects:

Adverse effects include fatigue, nausea and vomiting, hair loss, bone marrow suppression, neuropathy, and gastrointestinal disturbance.

Long-term adverse effects:

Fertility and ovarian function may be permanently affected by chemotherapy, especially in women aged over 40 years, although for some women with hormone dependent cancer, reduced ovarian function may contribute to the benefit of adjuvant treatment. Other potential long-term risks include induction of second cancers (especially haematological malignancies, although the risk is low) and cardiac impairment with cumulative anthracycline dosages. Provided that the cumulative dose of adriamycin (doxorubicin) does not exceed 300–350 mg/m2, the risk of congestive heart failure is lower than 1%.

Comment

The absolute benefits of these regimens need to be balanced against their toxicity for different women. New and highly active cytotoxic agents such as the taxanes are being examined with anthracyclines either in combination or sequence. Alternating sequences of cytotoxic agents may prove an effective way of circumventing acquired drug resistance and thus enhancing the efficacy of a regimen, such as the Milan regimen of single-agent anthracycline followed by standard CMF chemotherapy.

Clinical guide:

Anthracycline based chemotherapy should be used as adjuvant therapy.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Prolonged adjuvant combination chemotherapy (8–12 months v 4–6 months)

Summary

TREATMENT SUCCESS Different treatment durations compared with each other: Longer regimens (8–12 months) and shorter regimens (4–6 months) are equally effective at reducing recurrence rates ( high-quality evidence ). MORTALITY Different treatment durations compared with each other: Longer regimens (8–12 months) and shorter regimens (4–6 months) are equally effective at prolonging survival (high-quality evidence). ADVERSE EFFECTS Chemotherapy has been associated with fatigue, nausea and vomiting, hair loss, bone marrow suppression, neuropathy, and gastrointestinal disturbance. Chemotherapy may impair fertility and ovarian function.

Benefits

Duration of treatment:

We found one systematic review (as above, search date not reported, 11 RCTs, 6104 women), which compared longer regimens (doubling duration of chemotherapy from between 4–6 months to 8–12 months) with shorter regimens. It found no significant additional survival or recurrence benefit from longer treatment duration (recurrence: 754/1747 [43%] with longer duration v 778/1702 [46%] with shorter duration, P = 0.06; deaths: 541/1747 [31%] with longer duration v 526/1702 [31%] with shorter duration; reported as not significant).

Harms

Acute adverse effects:

Adverse effects include fatigue, nausea and vomiting, hair loss, bone marrow suppression, neuropathy, and gastrointestinal disturbance. Prolonged chemotherapy is more likely to be associated with lethargy and haematological toxicity (anaemia and neutropenia), and anthracycline regimens cause complete hair loss.

Long-term adverse effects:

Fertility and ovarian function may be permanently affected by chemotherapy, especially in women aged over 40 years, although for some women with hormone dependent cancer, reduced ovarian function may contribute to the benefit of adjuvant treatment. Other potential long-term risks include induction of second cancers (especially haematological malignancies, although the risk is low) and cardiac impairment with cumulative anthracycline dosages. Provided that the cumulative dose of adriamycin (doxorubicin) does not exceed 300–350 mg/m2, the risk of congestive heart failure is lower than 1%.

Comment

The absolute benefits of these regimens need to be balanced against their toxicity for different women. New and highly active cytotoxic agents such as the taxanes are being examined with anthracyclines either in combination or sequence. Alternating sequences of cytotoxic agents may prove an effective way of circumventing acquired drug resistance and thus enhancing the efficacy of a regimen, such as the Milan regimen of single-agent anthracycline followed by standard CMF chemotherapy.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Enhanced dose regimens of adjuvant combination chemotherapy

Summary

TREATMENT SUCCESS Different doses compared with each other: Enhanced and standard dose chemotherapy regimens seem to be equally effective at improving disease-free survival ( moderate-quality evidence ). MORTALITY Different doses compared with each other: Enhanced and standard dose chemotherapy regimens seem to be equally effective at prolonging survival (moderate-quality evidence). ADVERSE EFFECTS Chemotherapy has been associated with fatigue, nausea and vomiting, hair loss, bone marrow suppression, neuropathy, and gastrointestinal disturbance. Chemotherapy may impair fertility and ovarian function.

Benefits

Different doses:

We found no systematic review, but found two RCTs, which compared enhanced versus standard-dose chemotherapy regimens. The first RCT (2305 women with primary breast cancer) compared four courses of standard doxorubicin–cyclophosphamide versus the same dose of cyclophosphamide delivered in two courses versus double the dose of cyclophosphamide. It found no significant difference between treatments in disease-free survival or overall survival rates (P = 0.30 and P = 0.95). The second RCT (1572 women with node-positive, stage II breast cancer) compared cyclophosphamide (400 mg/m2 of body-surface area), doxorubicin (40 mg/m2 once every 28 days), and fluorouracil (400 mg/m2 twice every 28 days) for six cycles versus 50% higher doses of the three drugs (600 mg cyclophosphamide, 60 mg doxorubicin, and 600 mg fluorouracil), but for only four cycles versus half the total dose used in the other two groups, and at half the dose intensity used in the second group. It found that although moderate- or high-dose intensity significantly increased disease-free survival and overall survival compared with low-dose intensity, there was no significant difference in survival between medium and high doses.

Harms

Acute adverse effects:

Adverse effects include fatigue, nausea and vomiting, hair loss, bone marrow suppression, neuropathy, and gastrointestinal disturbance.

Long-term adverse effects:

Fertility and ovarian function may be permanently affected by chemotherapy, especially in women aged over 40 years, although for some women with hormone dependent cancer, reduced ovarian function may contribute to the benefit of adjuvant treatment. Other potential long-term risks include induction of second cancers (especially haematological malignancies, although the risk is low) and cardiac impairment with cumulative anthracycline dosages. Provided that the cumulative dose of adriamycin (doxorubicin) does not exceed 300–350 mg/m2, the risk of congestive heart failure is lower than 1%.

Comment

The absolute benefits of these regimens need to be balanced against their toxicity for different women. New and highly active cytotoxic agents such as the taxanes are being examined with anthracyclines either in combination or sequence. Alternating sequences of cytotoxic agents may prove an effective way of circumventing acquired drug resistance and thus enhancing the efficacy of a regimen, such as the Milan regimen of single-agent anthracycline followed by standard CMF chemotherapy.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Adjuvant anthracycline chemotherapy

Summary

TREATMENT SUCCESS Anthracycline regimens compared with standard CMF regimens: Adjuvant regimens containing an anthracycline are more effective than a standard multidrug chemotherapy regimen (cyclophosphamide, methotrexate, and fluorouracil) at reducing recurrence ( moderate-quality evidence ). MORTALITY Anthracycline regimens compared with standard CMF regimens: Adjuvant regimens containing an anthracycline are modestly more effective than a standard multidrug chemotherapy regimen (cyclophosphamide, methotrexate, and fluorouracil) at improving 5-year survival (moderate-quality evidence). ADVERSE EFFECTS Chemotherapy has been associated with nausea and vomiting, hair loss, bone marrow suppression, fatigue, and gastrointestinal disturbance. Chemotherapy may impair fertility and ovarian function.

Benefits

Anthracycline regimens versus standard CMF (cyclophosphamide, methotrexate, and fluorouracil) regimen:

We found one systematic review (as above, search date not reported, 11 RCTs, 5942 women), which compared regimens containing anthracycline (including the drugs adriamycin [doxorubicin] or 4-epidoxorubicin) versus standard CMF regimens. It found a significant reduction in recurrence rates in those on anthracycline regimens (P = 0.006), and a modest but significant improvement in 5-year survival (72% with anthracycline v 69% with CMF regimen; P = 0.02).

Harms

Acute adverse effects:

Adverse effects include fatigue, nausea and vomiting, hair loss, bone marrow suppression, neuropathy, and gastrointestinal disturbance. Anthracycline regimens cause complete hair loss.

Long-term adverse effects:

Fertility and ovarian function may be permanently affected by chemotherapy, especially in women aged over 40 years, although for some women with hormone dependent cancer, reduced ovarian function may contribute to the benefit of adjuvant treatment. Other potential long-term risks include induction of second cancers (especially haematological malignancies, although the risk is low) and cardiac impairment with cumulative anthracycline dosages. Provided that the cumulative dose of adriamycin (doxorubicin) does not exceed 300–350 mg/m2, the risk of congestive heart failure is lower than 1%.

Comment

The absolute benefits of these regimens need to be balanced against their toxicity for different women. New and highly active cytotoxic agents such as the taxanes are being examined with anthracyclines either in combination or sequence. Alternating sequences of cytotoxic agents may prove an effective way of circumventing acquired drug resistance and thus enhancing the efficacy of a regimen, such as the Milan regimen of single-agent anthracycline followed by standard CMF chemotherapy.

Clinical guide:

Anthracycline based chemotherapy should be used as adjuvant therapy.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Adjuvant tamoxifen

Summary

TREATMENT SUCCESS Compared with placebo: Adjuvant tamoxifen taken for up to 5 years is more effective at reducing the risk of recurrence in women with oestrogen receptor-positive tumours, irrespective of age, menopausal status, nodal involvement, or the addition of chemotherapy ( moderate-quality evidence ). Compared with adjuvant aromatase inhibitors (anastrozole and exemestane): Tamoxifen is less effective at reducing breast cancer events and at improving disease-free survival in postmenopausal women with early breast cancer (moderate-quality evidence). MORTALITY Compared with placebo: Adjuvant tamoxifen taken for up to 5 years is more effective at reducing mortality in women with oestrogen receptor-positive tumours, irrespective of age, menopausal status, nodal involvement, or the addition of chemotherapy (moderate-quality evidence). Different treatment durations compared with each other: Treatment with tamoxifen for 5 years seems to be more effective than shorter durations (2 years) at improving event-free and overall survival, but there does not seem to be any additional benefit at prolonging treatment beyond 5 years (moderate-quality evidence). Compared with exemestane: Tamoxifen seems to be less effective at improving overall survival in women with oestrogen receptor-positive breast cancer (moderate-quality evidence). ADVERSE EFFECTS Tamoxifen has been associated with a higher incidence of endometrial cancer and thrombotic complications compared with placebo or adjuvant aromatose inhibitors (letrozole, anastrozole, and exemestane) but is less likely to cause arthralgia and fractures. NOTE We found no clinically important results about adjuvant tamoxifen compared with radiotherapy in the treatment of women with primary operable breast cancer.

Benefits

Adjuvant tamoxifen versus placebo:

We found one systematic review (search date not reported, 55 RCTs, 37,000 women), which compared adjuvant tamoxifen with placebo. The review found that taking tamoxifen for a median of 5 years significantly reduced recurrence and mortality compared with placebo (RR for recurrence 0.58; RR for mortality 0.78; CI presented graphically; for both comparisons P less than 0.00001). These benefits seemed to be largely irrespective of age, menopausal status, daily tamoxifen dose (generally 20–40 mg), and of whether chemotherapy had been given to both groups.

Oestrogen receptor and lymph node status:

Subgroup analysis in the systematic review (as above, search date not reported, 55 RCTs, 37,000 women) found that tamoxifen was associated with a greater reduction in the recurrence rate for women with oestrogen receptor-positive versus those with receptor-negative tumours after 5 years (RR for recurrence: 0.5 with oestrogen receptor-positive v 0.94 with oestrogen receptor-negative), and with a slightly greater reduction in the absolute risk of 10-year recurrence in women with node-positive compared with node-negative disease (ARR 15.2% with node-positive v 14.9% with node-negative). Tamoxifen treatment for 5 years was also associated with a greater absolute improvement in 10-year survival in node-positive than node-negative women (see table 3 ).

Table 3.

Survival at 10 years in women treated with tamoxifen for 5 years compared with control treatment (no tamoxifen): results of a systematic review (see text).

  Control (%) Tamoxifen (%) Absolute benefit (%) SD (%) Significance (two sided)
Node +ve 50.5 61.4 +10.9 2.5 P less than 0.00001
Node –ve 73.3 78.9 +5.6 1.3 P less than 0.00001

SD, standard deviation.

Duration of treatment:

The review found significantly greater reductions in the risk of recurrence with increasing duration of adjuvant tamoxifen (RR for recurrence: 0.74 with 5 years of tamoxifen use v 0.88 with 1 year of tamoxifen use; P less than 0.0001). One RCT included in the review (3887 women) compared 2 and 5 years of treatment and found similar results. The RCT found that 5 years of adjuvant tamoxifen therapy improved event-free and overall survival rates versus 2 years of treatment (event-free survival: HR 0.82, 95% CI 0.71 to 0.96; overall survival: 0.82, 95% CI 0.69 to 0.99). The effects of prolonged treatment beyond 5 years are unclear. The largest RCT included in the systematic review (1153 women who had completed 5 years of tamoxifen therapy) compared placebo or 5 more years of tamoxifen. It found that placebo increased disease-free survival versus continued tamoxifen after a further 4-year follow-up (86% with continued tamoxifen v 92% with placebo; P = 0.003), although there was no significant difference in overall survival. Another RCT included in the review found no difference between placebo and continuing tamoxifen beyond 5 years.

Adjuvant tamoxifen versus radiotherapy:

We found no systematic review or RCTs comparing adjuvant tamoxifen versus radiotherapy in women with primary operable breast cancer.

Adjuvant aromatase inhibitors versus tamoxifen:

See benefits of adjuvant aromatase inhibitors.

Harms

One systematic review found an increased risk of endometrial cancer with tamoxifen (average HR 2.58, 95% CI 2.23 to 2.93). For 5 years of tamoxifen treatment, this resulted in a cumulative risk over 10 years of two deaths per 1000 women (95% CI 0 deaths per 1000 women to 4 deaths per 1000 women). There was no evidence of an increased incidence of other cancers, or of non-breast cancer related deaths (i.e. cardiac or vascular), although one extra death per 5000 women years of tamoxifen was attributed to pulmonary embolus. Bone loss was found in premenopausal women (1.4% bone loss a year) but not in postmenopausal women, because of the partial agonist effects of tamoxifen. There were mixed effects on cardiovascular risk, with significant reductions in low-density lipoprotein cholesterol associated with a reduced incidence of myocardial infarction in some studies, but an increased risk of thrombosis. Overall, no effect has been found on non-breast cancer mortality (HR 0.99, 95% CI 0.88 to 1.16).

Adjuvant tamoxifen versus radiotherapy:

See harms of tamoxifen plus radiotherapy. The risks of congenital malformations or late teratogenic manifestations in adulthood are unknown, and women should be offered choices in all cases concerning continuing with pregnancy or termination. Women are conventionally advised to stop tamoxifen before attempting pregnancy.

Adjuvant aromatase inhibitors versus tamoxifen:

See harms of adjuvant aromatase inhibitors.

Comment

The risk : benefit ratio may vary between women, with oestrogen receptor-negative women deriving little benefit. Current clinical practice has been to recommend tamoxifen for only 5 yearsthough we are awaiting results of longer studies using tamoxifen for 10 years.

Adjuvant aromatase inhibitors versus tamoxifen:

See comments of adjuvant aromatase inhibitors.

Clinical guide:

Tamoxifen for 5 years' adjuvant use has clinical benefits in improved survival for oestrogen receptor-positive breast cancer, but endometrial cancer and thrombotic events may cause significant morbidity or mortality.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Chemotherapy plus monoclonal antibody (trastuzumab) in women with overexpressed HER2/neu oncogene

Summary

TREATMENT SUCCESS Compared with observation (during or after chemotherapy): Trastuzumab, started after or during chemotherapy, is more effective at 1 year at increasing disease-free survival in HER2 positive women compared with 2 years of observation ( moderate-quality evidence ). MORTALITY Compared with observation (during or after chemotherapy): Trastuzumab, started after chemotherapy but not during chemotherapy, is more effective at 1 year at increasing overall survival in HER2 positive women compared with 2 years of observation (moderate-quality evidence). ADVERSE EFFECTS Trastuzumab has been associated with cardiac dysfunction.

Benefits

Trastuzumab versus placebo:

We found no systematic review or RCTs.

Trastuzumab versus observation, after chemotherapy:

We found one RCT comparing three treatments: trastuzumab every 3 weeks for 1 year; trastuzumab every 3 weeks for 2 years; or observation. Women completed loco-regional therapy and at least four cycles of primary or adjuvant chemotherapy before randomisation. The RCT found that 1 year of trastuzumab increased disease-free survival (absence of recurrence, contralateral breast cancer, second non-breast malignant disease, or death without prior recurrence) compared with observation at 2 years (5081 women with HER2 positive and either node-positive or node-negative breast cancer; AR of disease-free survival event: 127/1694 [7.5%] with 1 year of trastuzumab v 220/1693 [13.0%] with observation; HR 0.54, 95% CI 0.43 to 0.67). It found no significant difference in overall survival between 1 year of trastuzumab and observation at 2 years (AR of death: 29/1694 [1.7%] with trastuzumab v 37/1693 [2.2%] with observation; HR 0.76, 95% CI 0.47 to 1.23). Outcomes with 2 years of trastuzumab were not reported.

Trastuzumab versus observation, starting during chemotherapy:

We found a pooled analysis of two RCTs, which compared doxorubicin plus cyclophosphamide followed by paclitaxel versus doxorubicin plus cyclophosphamide followed by paclitaxel plus trastuzumab (for 1 year). One of the RCTs also included a group with doxorubicin plus cyclophosphamide followed by paclitaxel followed by trastuzumab, which was excluded from joint analysis. The RCTs found that 1 year of trastuzumab significantly increased disease-free survival and overall survival compared with observation at a median follow-up of 2 years (3351 women with HER2 positive and either node-positive or node-negative breast cancer; AR for disease-free survival events: 133/1672 [8%] with trastuzumab v 261/1679 [16%] with observation; HR 0.48, 95% CI 0.39 to 0.59; AR for death: 62/1672 [4%] with trastuzumab v 92/1679 [6%] with observation; HR 0.67, 95% CI 0.48 to 0.93).

Harms

In women with previous exposure to anthracycline, there is concern about cardiac toxicity associated with trastuzumab therapy. Therefore, the RCTs only included women with normal left ventricular ejection fraction. The RCTs monitored cardiac function (symptoms and left ventricular ejection fraction) during the trials, with specific stopping criteria for cardiotoxicity.

Trastuzumab versus observation, after chemotherapy:

The RCT found that 1 year of trastuzumab significantly increased grade 3 or 4 toxicity and severe congestive heart failure compared with observation (at least one grade 3 or 4 event: 132/1677 [7.9%] with 1 year of trastuzumab v 75/1710 [4.4%] with observation, P less than 0.001; severe congestive heart failure: 9/1677 [0.54%] with 1 year of trastuzumab v 0/1710 [0%] with observation, P = 0.002). It found that left ventricular ejection fraction decreased in a significantly higher proportion of women with trastuzumab than with observation (113/1677 [6.7%] with trastuzumab v 34/1710 [2.0%] with observation; P less than 0.001).

Trastuzumab versus observation, starting during chemotherapy:

The RCT found that 1 year of trastuzumab increased cardiac toxicity compared with observation (New York Heart Association class III or IV congestive heart failure or death from cardiac causes; in the first RCT: 4% with trastuzumab v 1% with observation; second RCT: 3% with trastuzumab v 0% with observation; significance not reported). The RCT found no differences in any other common toxicity criteria (data not reported).

Comment

Clinical guide:

There is no consensus in HER2 positive (either 3+ or FISH positive) individuals about whether to start trastuzumab during or after chemotherapy, and current expert opinion appears to favour continuing the trastuzumab for 1 year with 3-monthly ECHOs to assess cardiac function. Patients with HER 2+ tumours by immunohistochemistry should have the appropriate HER2 status assess by FISH.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Adjuvant taxanes

Summary

TREATMENT SUCCESS Adjuvant taxoid regimens compared with standard anthracycline based regimens: Adjuvant taxoid regimens are more effective at increasing disease-free survival in women with breast cancer who are at high risk of relapse, as defined by node positivity ( moderate-quality evidence ). MORTALITY Adjuvant taxoid regimens compared with standard anthracycline based regimens: Adjuvant taxoid regimens are more effective at improving 5-year overall survival rates in women with node-positive early breast cancer (moderate-quality evidence).

Benefits

We found six RCT phase III trials (1016 to 3121 participants) in women with breast cancer at high risk of relapse, as defined by node positivity, that found a statistically significant disease-free survival benefit, or improved response rate, for adjuvant taxoid regimens compared with standard anthracycline-based therapy in the adjuvant setting. Out of these six studies, four involved docetaxel. The RCTs used a variety of trial designs and comparisons of different regimens. We have reported two of the most robust studies in detail below. The first RCT randomised 1491 women with axillary node-positive breast cancer to six cycles of treatment with either docetaxel plus doxorubicin and cyclophosphamide (TAC) or fluorouracil plus doxorobucin and cyclophosphamide (FAC) as adjuvant chemotherapy after surgery. The primary end-point was disease-free survival. The median follow-up was 55 months. The RCT found that TAC significantly increased the estimated rates of disease-free survival compared with FAC at 5 years (75 % with TAC v 68 % with FAC; P = 0.001). After adjustment for nodal status, TAC was associated with a statistically significant 28% reduction in the risk of relapse compared with FAC (HR 0.72, 95% CI 0.59 to 0.88). The estimated rates of overall survival at 5 years were 87% with TAC compared with 81% with FAC. The RCT found that treatment with TAC resulted in a significant reduction in the risk of death compared with FAC (30% reduction; HR 0.7, 95% CI 0.53 to 0.91; P = 0.008). The second RCT compared six cycles of fluorouracil, epirubicin, and cyclophosphamide (FEC) versus a sequential regimen of three cycles of FEC followed by three cycles of docetaxel (FEC-D) as adjuvant treatment in 1999 women with node-positive early breast cancer. Women who were hormone receptor-positive received tamoxifen for 5 years after chemotherapy. Median follow-up was 60 months. The RCT found that 5-year disease-free survival rates were significantly improved with FEC-D compared with FEC alone (78.4% with FEC-D v 73.2% with FEC; P = 0.012). A multivariate analysis adjusted for prognostic factors found an 18% reduction in the relative risk of relapse with FEC-D (HR 0.82, 95% CI 0.69 to 0.99; P = 0.34). The RCT found that FEC-D significantly improved 5-year overall survival rates compared with FEC (5-year overall survival rates: 90.7% with FEC-D v 86.7% with FEC; HR 0.73, 95% CI 0.56 to 0.94).

Harms

In one RCT, the incidence of grade 3 or 4 neutropenia was significantly higher with docetaxel plus doxorubicin and cyclophosphamide (TAC) compared with fluorouracil plus doxorobucin and cyclophosphamide (FAC) (66% with TAC v 49% with FAC; absolute numbers not reported; P less than 0.001).The RCT found that rates of febrile neutropenia were 25% with TAC compared with 3% with FAC (P less than 0.001), and that grade 3 or 4 infections occurred in 4% of women with TAC compared with 2.% of women with FAC (P = 0.05). It reported that no deaths occurred as a result of infection. It reported that two women in each group died during treatment, and congestive heart failure and acute myeloid leukemia occurred in less than 2% of the women in each group. In another RCT, the incidence of grade 3 to grade 4 neutropenia, and the incidence of nausea and vomiting, were significantly higher with FEC (fluororubacin, epirubicin, and cyclophosphamide) compared with FEC followed by docetaxel (neutropenia on day 21: 33.6% with FEC v 28.1% with FEC-D, P = 0.008; nausea and vomiting grade 3–4: 20.5% v 11.2%, P less than 0.001). It found that docetaxel was associated with more febrile neutropenia in the fourth cycle, stomatitis, oedema, and nail disorders (febrile neutropenia in the 4th cycle: 1% with FEC v 4.6% with FEC-D, P value not reported; stomatitis grade 3–4 cycles 4–6: 2% v 4.0%, P = 0.03; moderate or severe oedema: 0.3% v 4.8%, P less than 0.001; nail disorders: 1% v 10.3%, P less than 0.001). Although rare, it found that there were fewer cardiac events after FEC-D (any cardiac events reported as serious adverse event: 1.3% with FEC v 0.4% with FEC-D, P = 0.03), attributable mainly to the lower anthracycline cumulative dose.

Comment

Clinical guide

: While in England and Wales the National Institute of Clinical Excellence has approved TAC, nearly all clinicians however have indicated their preference to use docetaxel as part of the FEC-T regimen as per the PACS 01 study.The rationale for favouring FEC-T over TAC is based on cost, capacity issues, and data reported in the PACS 01 and BCIRG 001 studies, in particular the rate of haematological toxicities, specifically febrile neutropenia and the need for 7 days of primary prophylactic G-CSF with the TAC regimen. In conclusion, sequential adjuvant chemotherapy with FEC followed by docetaxel significantly improves disease-free and overall survival in node-positive breast cancer patients and has a significantly more favorable safety profile. In other words, FEC followed by docetaxel, FEC-T, buys efficacy with lower toxicity and costs (less G-CSF).

Substantive changes

Adjuvant taxanes (under question on treatments for primary operable breast cancer) New option added. Six RCTs included. Adjuvant taxanes categorised as Likely to be beneficial.

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Less extensive mastectomy

Summary

TREATMENT SUCCESS Mastectomy compared with breast conservation: Mastectomy compared with breast conservation surgery with radiotherapy seem to be equally effective at reducing local or overall recurrence rates ( moderate-quality evidence ). Different extents of local excision compared with each other: We don’t know whether lumpectomy is more effective than quadrantectomy at reducing local recurrence (moderate-quality evidence). MORTALITY Supraradical, radical, and total mastectomy compared with each other: Extensive surgery and less-extensive surgery seem to be equally effective at improving the annual risk of death at 10 years in women with operable breast cancer, but extensive surgery has worse cosmetic outcomes (moderate-quality evidence). Radical compared with total mastectomy: Radical and total mastectomy (with or without axillary radiotherapy) are equally effective at 25 years at improving survival in women with operable breast cancer regardless of nodal status (moderate-quality evidence). Mastectomy compared with breast conservation: Mastectomy and breast conservation surgery (with or without radiotherapy) seem to be equally effective at reducing the annual risk of death at 10 years (moderate-quality evidence). Radical mastectomy compared with quandrantectomy plus radiotherapy: Radical mastectomy, and quandrantectomy plus radiotherapy seem to be equally effective at reducing all-cause mortality at 20 years (moderate-quality evidence). Lumpectomy (with or without radiotherapy) compared with total mastectomy: Lumpectomy (with or without radiotherapy) and total mastectomy seem to be equally effective at reducing mortality at 20 years (moderate-quality evidence). Modified radical mastectomy compared with lumpectomy plus axillary dissection plus radiotherapy: Modified radical mastectomy, and lumpectomy plus axillary dissection plus radiotherapy seem to be equally effective at improving overall survival at 20 years (moderate-quality evidence).

Benefits

Comparisons between supraradical, radical, and total mastectomy:

We found one systematic review (search date not reported, 5 RCTs, 2090 women with operable breast cancer) comparing supraradical mastectomy versus radical mastectomy (2 RCTs), radical versus total mastectomy (2 RCTs), and supraradical versus total mastectomy (1 RCT). It found no significant difference in the annual risk of death over a 10-year period (OR for more extensive v less-extensive surgery 0.98, CI presented graphically; P = 0.7).

Comparisons between radical, total, and simple mastectomy:

We found one systematic review (as above, search date not reported, 4 RCTs, 1296 women with operable breast cancer), which compared radical or total mastectomy versus simple mastectomy. It found no significant difference in the annual risk of death over a 10-year period (OR for more extensive v less-extensive surgery 0.98, CI presented graphically; P = 0.8). Long-term follow-up of one RCT included in the review (1079 women), which compared radical mastectomy versus total mastectomy with or without axillary radiotherapy found no significant difference in survival between total and radical mastectomy, regardless of nodal status at 25-year follow-up (in women with negative nodes: HR for total mastectomy plus radiotherapy v radical mastectomy 1.08, 95% CI 0.91 to 1.28; HR for total mastectomy without radiotherapy v radical mastectomy 1.03, 95% CI 0.87 to 1.23; in women with positive nodes: HR for total mastectomy plus radiotherapy v radical mastectomy 1.06, 95% CI 0.89 to 1.27).

Mastectomy versus breast conservation:

We found two systematic reviews. The first review (search date 1995, 6 RCTs) compared breast conservation versus mastectomy. It found no significant difference in the risk of death at 10 years (5 RCTs, 3006 women; OR breast conservation v mastectomy 0.91, 95% CI 0.78 to 1.05). Where more than half of node-positive women in both mastectomy and breast conservation groups received adjuvant nodal radiotherapy, both groups had similar survival rates. Where fewer than half of node-positive women in both groups received adjuvant nodal radiotherapy, survival was better with breast conservation (OR of death: breast conservation v with mastectomy 0.69, 95% CI 0.49 to 0.97). The second review (search date not reported, 9 RCTs, 5610 women potentially suitable for breast-conserving surgery) compared mastectomy versus breast-conserving surgery (with or without radiotherapy; the proportion of women receiving mastectomy who also received radiotherapy was unclear). It found no significant difference in annual risk of death over 10 years between mastectomy and breast-conserving surgery either with or without radiotherapy (mastectomy v breast conservation alone: 1 RCT, 1432 women, OR 0.97, CI presented graphically, P = 0.4; mastectomy v breast conservation plus radiotherapy: 9 RCTs, 4891 women, OR 1.02, CI presented graphically, P = 0.7). It also found no significant difference in overall rates of recurrence or rates of local recurrence between mastectomy and breast-conserving surgery plus radiotherapy (6 RCTs, 3107 women; OR for overall recurrence mastectomy v breast conservation plus radiotherapy 0.96, 95% CI 0.88 to 1.04; AR for local recurrence 6.2% with mastectomy v 5.9% with breast conservation plus radiotherapy; P value reported as not significant). Three RCTs included in the reviews have now reported 20-year follow-up results. The first RCT (701 women with breast cancer less than 2 cm diameter) found no significant difference between radical mastectomy and quadrantectomy plus radiotherapy for all-cause mortality at 20 years (death rate about 42% in both groups; P = 1.0). The second RCT (1851 women) compared lumpectomy alone, lumpectomy plus radiotherapy, and total mastectomy. It found no significant difference between lumpectomy (with or without radiotherapy) and total mastectomy in mortality at 20 years (HR lumpectomy alone v mastectomy 1.05, 95% CI 0.90 to 1.23; HR lumpectomy plus radiotherapy v mastectomy 0.97, 95% CI 0.77 to 1.06). The third RCT (237 women) found no significant difference between modified radical mastectomy and lumpectomy plus axillary dissection plus radiotherapy in overall survival or in disease-free survival at 20 years (AR for overall survival: 58% with mastectomy v 54% with lumpectomy, P = 0.67; AR for disease-free survival: 67% with mastectomy v 63% with lumpectomy, P = 0.64).

Different extents of local excision in breast conservation:

We found no systematic review but found one RCT (705 women), which compared lumpectomy versus quadrantectomy. There were significantly more local recurrences with lumpectomy than with quadrantectomy (7% with lumpectomy v 2% with quadrantectomy), but a major factor associated with local recurrence in the lumpectomy group was incomplete excision (see comment below). We found no RCTs comparing wide local excision (complete excision microscopically) versus quadrantectomy.

Harms

More extensive surgery results in a poorer cosmetic result. Between 60–90% of women having breast conservation have an excellent or good cosmetic result (median 83%, 95% CI 67% to 87%). The most important factor influencing cosmetic outcome is the volume of tissue excised; the larger the amount of tissue excised, the worse the cosmetic result. The RCT of different extents of local excision in breast conservation found that, in a subset of 148 women, there was a significantly higher rate of poor cosmetic outcome with quadrantectomy (RR quadrantectomy v lumpectomy 3.1, 95% CI 1.2 to 8.1). Only isolated small studies have shown no correlation between extent of surgical excision and cosmesis.

Comment

The link between completeness of excision and local recurrence after breast conservation has been evaluated in 16 centres. In 13 of these, incomplete excision significantly increased the risk of local recurrence compared with complete excision (RR 1.03, 95% CI 1.03 to 1.05). The three centres not reporting increased rates of local recurrence after incomplete excision gave much higher doses of local radiotherapy (65–72 Gy) to people with involved margins. Two centres also used re-excision, and women with involved margins had only focal margin involvement.

Clinical guide:

The main aim of surgical excision is to remove the tumour with microscopically clear margins. The width of what constitutes an adequate margin remains unclear. As a bare minimum, excision should aim to excise all disease with no microscopic extension to the edge of the specimen. The extent of the disease, the size of the breast, the location of the tumour in the breast and the desires of the patient will all play a role in what is then considered appropriate in terms of choice of surgical procedure. Complete excision of breast cancer achieved by less than radical mastectomy or by breast conservation may give an improved cosmetic outcome over more radical surgery without any detriment to outcome.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Ovarian ablation

Summary

TREATMENT SUCCESS Compared with no ablation: Ovarian ablation by irradiation or surgery is more effective at 15 years at improving recurrence-free survival in premenopausal women (irrespective of nodal status) with early breast cancer ( moderate-quality evidence ). MORTALITY Compared with no ablation: Ovarian ablation by irradiation or surgery is more effective at increasing overall survival at 15 years in premenopausal women (irrespective of nodal status) with early breast cancer (moderate-quality evidence).

Benefits

Ovarian ablation versus no ablation:

We found one systematic review (search date not reported, 12 RCTs with at least 15-years' follow-up, 2102 premenopausal women with early breast cancer), which compared ovarian ablation by irradiation or surgery versus no ablation. It found that ovarian ablation by irradiation significantly increased overall survival and recurrence-free survival versus surgery alone after 15 years (overall survival: 52% with ablation v 46% with no ablation, P = 0.001; disease-free survival: 45% with ablation v 39% with no ablation, P = 0.0007). Benefit was independent of nodal status.

Harms

We found no good evidence on long-term adverse effects. Concerns exist about late sequelae of ovarian ablation, especially effects on bone mineral density and cardiovascular risk. Acute adverse effects are likely to be menopausal symptoms.

Comment

Five of the RCTs compared ovarian ablation plus chemotherapy with chemotherapy alone. In these, the absolute benefit of ablation was smaller than in RCTs of ovarian ablation alone. It may be that cytotoxic chemotherapy itself suppresses ovarian function, making the effect of ablation difficult to detect in combined RCTs. When only premenopausal women who did not have chemotherapy were considered, there was a 27% improvement in the odds of recurrence-free survival. RCTs are underway of reversible oophorectomy using gonadotrophin releasing hormone analogues, which would allow preservation of fertility in younger women with oestrogen receptor-positive tumours.

Clinical guide:

Oophorectomy in premenopausal women with oestrogen receptor-positive cancers improves survival independent of node status. The addition of chemotherapy may further improve survival.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Radiotherapy after breast-conserving surgery

Summary

TREATMENT SUCCESS Breast-conserving surgery plus radiotherapy compared with breast-conserving surgery alone: Breast-conserving surgery plus radiotherapy is more effective at 15–20 years at reducing local recurrence ( moderate-quality evidence ). Breast-conserving surgery plus radiotherapy compared with mastectomy: Breast-conserving surgery plus radiotherapy seems to be equally effective as mastectomy at 10 years at reducing recurrences and at improving disease-free survival at 20 years (moderate-quality evidence). MORTALITY Breast-conserving surgery plus radiotherapy compared with breast-conserving surgery alone: Breast-conserving surgery plus radiotherapy is more effective at 15 years at reducing mortality due to breast cancer, and all-cause mortality (moderate-quality evidence). Breast-conserving surgery plus radiotherapy compared with mastectomy: Breast-conserving surgery plus radiotherapy seems to be equally effective as mastectomy at improving overall survival at 10–20 years (moderate-quality evidence). Radical mastectomy compared with quandrantectomy plus radiotherapy: Radical mastectomy seems to be equally effective as quandrantectomy plus radiotherapy at reducing all-cause mortality at 20 years (moderate-quality evidence). Lumpectomy (with or without radiotherapy) compared with total mastectomy: Lumpectomy (with or without radiotherapy), and total mastectomy seem to be equally effective at reducing mortality at 20 years (moderate-quality evidence). Modified radical mastectomy compared with lumpectomy plus axillary dissection plus radiotherapy: Modified radical mastectomy seems to be equally effective as lumpectomy plus axillary dissection plus radiotherapy at improving overall survival at 20 years (moderate-quality evidence). ADVERSE EFFECTS Radiotherapy may rarely be associated with late adverse effects, such as pneumonitis, pericarditis, arm oedema, brachial plexopathy, and radionecrotic rib fracture.

Benefits

Breast-conserving surgery plus radiotherapy versus breast-conserving surgery alone:

We found one systematic review, which compared breast-conserving surgery plus radiotherapy versus breast-conserving surgery alone. Seven RCTs included in the review used megavoltage x rays. The review found that postoperative radiotherapy significantly reduced the risk of breast cancer mortality, all-cause mortality, and isolated local recurrence compared with no radiotherapy (search date not reported, 10 RCTs; 7311 women; 15-year risk of breast cancer mortality: 31% with breast-conserving surgery plus radiotherapy v 36% with breast-conserving surgery alone, rate ratio 0.83, CI presented graphically, P = 0.0002; 15-year risk of all-cause mortality: 35% with breast-conserving surgery plus radiotherapy v 40% with breast-conserving surgery alone, P = 0.005; 5-year risk of recurrence: 7% with breast-conserving surgery plus radiotherapy v 26% with breast-conserving surgery alone, rate ratio 0.31, CI presented graphically, P less than 0.00001). Subgroup analyses suggested that the proportional reduction in local recurrence was similar in women of different ages and tumour characteristics.One RCT included in the review has now reported long-term results with a median follow-up period of 13.7 years. The improvement in local control with the addition of radiotherapy to breast-conserving surgery was confirmed (400 women; 20 year local breast recurrence: 28.6% with breast-conserving surgery plus radiotherapy v 49.8% with breast-conserving surgery and no radiotherapy; HR 0.45 95% CI 0.31 to 0.64; P = 0.0001). It found no significant difference in survival between groups (HR 0.91, 95% CI 0.64 to1.28; P = 0.59).

Breast-conserving surgery plus radiotherapy versus mastectomy:

We found one systematic review (search date not reported, 9 RCTs, 4891 women) and one additional RCT, which compared breast radiotherapy after breast-conserving surgery versus simple or modified radical mastectomy in women with invasive breast cancer. The review found no significant difference in annual risk of death over 10 years (OR 1.02, CI presented graphically; P = 0.7) or annual risk of any recurrence or local recurrence (6 RCTs, 3107 women; overall OR for any recurrence: mastectomy v breast conservation plus radiotherapy 0.96, 95% CI 0.88 to 1.04; AR for local recurrence: 6.2% with radiotherapy after breast conservation surgery v 5.9% with radical mastectomy; P value reported as not significant). Three RCTs included in the review have now reported 20-year follow-up results. The first RCT (701 women with breast cancer less than 2 cm diameter) found no significant difference between radical mastectomy and quadrantectomy plus radiotherapy for all-cause mortality at 20 years (death rate about 42% in both groups; P = 1.0). The second RCT (1851 women) compared lumpectomy alone, lumpectomy plus breast radiotherapy, and total mastectomy. It found no significant difference between lumpectomy plus radiotherapy and total mastectomy for mortality at 20 years (HR lumpectomy plus radiotherapy v mastectomy 0.97, 95% CI 0.77 to 1.06). The third RCT (237 women) found no significant difference between modified radical mastectomy and lumpectomy plus axillary clearance plus radiotherapy in overall survival or disease-free survival at 20 years (AR for overall survival: 58% with mastectomy v 54% with lumpectomy, P = 0.67; AR for disease-free survival: 67% with mastectomy v 63% with lumpectomy, P = 0.64). One additional RCT (187 women) found no significant difference in overall or disease-free survival between breast-conserving surgery plus radiotherapy and modified radical mastectomy at 40 months (overall survival: 94.1% with breast conservation therapy plus radiotherapy v 93.7% with mastectomy; disease-free survival: 93.9% with breast-conserving surgery plus radiotherapy v 89.2% with mastectomy; P less than 0.05 for both comparisons). The incidence of loco-regional recurrence was similar in the two groups (2/76 [2.6%] with breast-conserving surgery plus radiotherapy v 2/111 [1.8%] with mastectomy; significance not reported).

Harms

Breast-conserving surgery plus radiotherapy versus breast-conserving surgery alone:

The review did not perform a separate meta-analysis of non-breast cancer deaths for radiotherapy plus breast-conserving surgery versus breast-conserving surgery alone (see general adverse events of adding radiotherapy to breast surgery below for overall analyses). However, it found that radiotherapy plus breast-conserving surgery reduced all-cause mortality compared with breast-conserving surgery alone (see benefits above).

Breast-conserving surgery plus radiotherapy versus mastectomy:

The review did not report on the harms of breast-conserving surgery plus radiotherapy compared with mastectomy. However, overall, the review found that adding radiotherapy to mastectomy or breast-conserving surgery increased the risk of non-breast cancer death compared with surgery alone (AR 7.7% with radiotherapy v 5.7% with no radiotherapy; OR 1.24, 95% CI 1.09 to 1.42).

Quality of life:

One old prospective study of quality of life after breast-conserving surgery has been reported and is based on an RCT in which participants were randomised to either radiotherapy or no further local treatment after surgery. The study (1984–1989; 837 women) reported that radiotherapy significantly reduced quality of life compared with no radiotherapy at 1 and 2 months (17 item modified Breast Cancer Chemotherapy Questionnaire, higher score indicating better quality of life; mean score change at 1 month: –0.07 with radiotherapy v + 0.21 without radiotherapy; mean score change at 2 months: –0.05 with radiotherapy v + 0.30 without radiotherapy; P = 0.0001 for both time points). Radiotherapy significantly increased breast pain at 6 months and breast skin irritation at 3 months (AR for breast pain: 33% with radiotherapy v 20% without radiotherapy, P = 0.0002; skin irritation: 28% with radiotherapy v 14% without radiotherapy, P = 0.0001). However, there was no significant difference between groups in the risk of breast pain, skin irritation, or upset because of breast appearance at 2 years (AR for breast pain: about 15% for both groups, P reported as not significant; skin irritation: 7% in both groups, P reported as not significant; upset with breast appearance: 5% in both groups, P = 0.62).

General adverse effects of adding radiotherapy to breast surgery:

One systematic overview (search date 2001) found limited data on radiotherapy related morbidity and reported that no conclusions could be drawn. A consensus document published in 1998 (mainly of women having breast-conserving surgery or mastectomy with variation in radiotherapy techniques, doses, and fractionation) reported two severe adverse effects of radiotherapy, namely acute pneumonitis (0.7–7.0%) and pericarditis (0–0.3%), and the following long-term adverse effects: significant arm oedema (1% without axillary clearance), radionecrotic rib fracture (1.1–1.5%), and brachial plexopathy (0–1.8%). The risk and severity of adverse effects increased with volume irradiated, total dose received, dose per fraction, previous surgery (e.g. axillary dissection), and radiotherapy techniques that caused overlap in irradiated tissues. A more recent systematic review found that in adding radiotherapy to surgery (breast-conserving surgery or mastectomy) increased the risk of non-breast cancer mortality (mainly from heart disease and lung cancer) compared with no radiotherapy (search date not reported, 63 RCTs; 32,800 people; 15-year risk of non-breast cancer mortality: 16% with radiotherapy v 15% with no radiotherapy, rate ratio 1.12, CI not reported, P = 0.001; rate ratio for heart disease mortality 1.27, CI not reported, P = 0.0001; rate ratio for lung cancer mortality 1.78, CI not reported, P = 0.0004). Three recent RCTs included in the review, with data beyond 10 years, did not find an excess of cardiac deaths. This may be because great care was taken in these RCTs to reduce the radiotherapy dose to the heart and to measure morbidity and mortality from cardiac disease. Studies assessing cosmetic results have mainly been retrospective, using poorly validated outcomes. The effects of social, psychological, and financial disruption from attending 5–6 weeks of radiotherapy have not been assessed well. There is an extremely low reported incidence of radiotherapy induced malignancy, usually soft tissue sarcomas, in the irradiated breast.

Comment

RCTs comparing breast-conserving surgery with and without radiotherapy, as well as retrospective case series, have found that prognostic factors for local recurrence after breast-conserving surgery include positive tumour margins, an extensive intraduct component, younger age, lymphovascular invasion, histological grade, and systemic therapy. The only consistent independent risk factor is avoiding radiotherapy. One systematic review of radiotherapy effects in breast cancer by the Swedish Council of Technology Assessment in Health Care, which did not carry out meta-analysis, supports the above conclusions for postmastectomy radiotherapy, radiotherapy after breast-conserving surgery for ductal carcinoma in situ, and for the comparability of breast-conserving surgery with radiotherapy and modified radical mastectomy alone for invasive breast cancer for disease-free survival and overall survival. It concluded that there are conflicting data about the effects of breast conservation surgery plus radiotherapy compared with modified radical mastectomy on local recurrence in people with invasive cancer.

Clinical guide

: For women with early breast cancer, there appears to be no survival difference whether breast-conserving surgery and radiotherapy or mastectomy is chosen. Radiotherapy following breast-conserving therapy may substantially reduce local disease recurrence without detriment to survival when modern radiotherapy techniques are used. The pros and cons of these approaches need to be discussed with the patient prior to surgery. The addition of tamoxifen to breast-conserving surgery, with or without radiotherapy, leads to better local control. The best local control outcome is when breast-conserving surgery, radiotherapy and tamoxifen (if oestrogen receptor-positive) are all part of the treatment. Chemotherapy appears to have little effect on local control in the absence of radiotherapy.

Substantive changes

Radiotherapy after breast-conserving surgery (under question on treatments for primary operable breast cancer) One extended follow-up of a previously included RCT added; benefits data enhanced, categorisation of "radiotherapy after breast-conserving surgery (reduced local recurrence and breast cancer mortality compared with breast-conserving surgery alone)" as Beneficial and "radiotherapy plus tamoxifen after breast-conserving surgery (reduced local recurrence rates)" as Likely to be beneficial unchanged.

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Radiotherapy plus tamoxifen after breast-conserving surgery (reduced local recurrence rates)

Summary

TREATMENT SUCCESS Breast conserving surgery plus radiotherapy compared with breast-conserving surgery plus tamoxifen: Radiotherapy plus tamoxifen after breast-conserving surgery is more effective at 5 years at reducing ipsilateral or local recurrences ( moderate-quality evidence ). MORTALITY Radiotherapy plus tamoxifen compared with tamoxifen alone: Radiotherapy plus tamoxifen seems to be equally effective as tamoxifen alone at improving survival in women who have undergone breast-conserving surgery (moderate-quality evidence). ADVERSE EFFECTS Radiotherapy may be associated with late adverse effects, which are rare, including pneumonitis, pericarditis, arm oedema, brachial plexopathy, and radionecrotic rib fracture.

Benefits

Breast-conserving surgery plus radiotherapy versus breast-conserving surgery plus tamoxifen:

We found no systematic review but found three RCTs. The first RCT (1009 women after lumpectomy for node-negative invasive breast cancer 1 cm or less, 80% were aged 50 years or over), which compared three treatments: radiotherapy (started 2 weeks after surgery, 50 Gy over 5 weeks with or without external beam boost), radiotherapy plus tamoxifen, and tamoxifen alone. It found that radiotherapy (with or without tamoxifen) significantly reduced ipsilateral breast cancer recurrence compared with tamoxifen alone after a median follow-up of 87 months (23/332 [7%] with radiotherapy alone v 9/334 [3%] with radiotherapy plus tamoxifen v 45/334 [14%] with tamoxifen alone; HR for radiotherapy alone v tamoxifen alone 0.51, 95% CI 0.31 to 0.84, P = 0.008; HR for radiotherapy plus tamoxifen v tamoxifen alone 0.19, 95% CI 0.09 to 0.39, P less than 0.001; HR for radiotherapy plus tamoxifen v radiotherapy alone 0.37, 95% CI 0.17 to 0.80, P = 0.01). It found no significant difference in survival or other events (tumour recurrence, contralateral breast cancer, other second primary breast cancer, or death with no evidence of cancer) between the three treatments (survival: 312/332 [94%] with radiotherapy alone v 314/334 [94%] with tamoxifen alone v 312/334 [93%] with radiotherapy plus tamoxifen, P = 0.93; events: 61/332 [18%] with radiotherapy alone v 74/334 [22%] with tamoxifen alone v 52/334 [16%] with radiotherapy plus tamoxifen, P = 0.08). The second RCT (769 women with primary breast cancer, aged over 50 years, who had undergone breast-conserving surgery for an invasive adenocarcinoma, tumour diameter 5 cm or less, histologically node-negative unless aged 65 years or over, when a clinically negative axilla was acceptable) compared radiotherapy plus tamoxifen versus tamoxifen alone (20 mg/day for 5 years). It found that radiotherapy plus tamoxifen increased disease-free survival rates and reduced recurrence and rate of relapse in the ipsilateral breast versus tamoxifen alone after 5 years (HR for relapse: 1.7, 95% CI 1.2 to 2.5, P = 0.004; recurrence: 7.7% with tamoxifen alone v 0.6% with radiotherapy plus tamoxifen, HR 8.3, 95% CI 3.3 to 21.1, P less than 0.001; AR for disease-free survival: 84% with tamoxifen alone v 91% with radiotherapy plus tamoxifen, P = 0.004). A subgroup analysis found greater benefits in local relapse rates in women with T1, receptor-positive tumours at 5 years (3.2% with receptor-positive tumours v 7.8% with receptor-negative or T2 tumours). The third RCT (women aged 70 years or over with clinical stage I breast cancer, who had undergone lumpectomy for receptor-positive tumour) compared postoperative radiotherapy versus no radiotherapy in women taking tamoxifen (20 mg/day) for 5 years. It found that radiotherapy reduced local or regional recurrence rate, but found no significant difference in time to mastectomy versus no radiotherapy (recurrence rate: 2/317 [0.6%] with radiotherapy plus tamoxifen v 16/319 [5.0%] with tamoxifen alone, P less than 0.01; time to mastectomy: P = 0.15, no further details reported). There was no significant difference between treatment groups in time to distant metastasis (P = 0.97) or overall survival rates (P = 0.94).

Harms

Breast-conserving surgery plus radiotherapy versus breast-conserving surgery plus tamoxifen:

The first RCT (1009 women after lumpectomy in node-negative invasive breast cancer 1 cm or less) compared radiotherapy, radiotherapy plus tamoxifen, and tamoxifen alone and found no significant difference between treatments in endometrial cancer or other second primary cancers (endometrial cancer: 1/332 [0.3%] with radiotherapy alone v 1/334 [0.3%] with tamoxifen alone v 5/334 [1.5%] with radiotherapy plus tamoxifen, P = 0.12; other second primary cancer: 10/332 [3.0%] with radiotherapy alone v 14/334 [4.2%] with tamoxifen alone v 15/334 [4.5%] with radiotherapy plus tamoxifen, P = 0.65). The third RCT found that more women experienced breast pain and skin fibrosis or retraction following treatment with tamoxifen plus radiotherapy (P less than 0.05 for both outcomes). Physicians rated worse cosmesis, oedema, and skin colour in women who had received radiotherapy in addition to tamoxifen compared with those who received tamoxifen alone (P less than 0.05 for all outcomes).

Quality of life:

A more recent prospective study (1992–2000) assessed whether breast pain affected quality of life in women who received breast-conserving surgery plus tamoxifen plus either radiotherapy or no further treatment. It found no significant difference between groups in pain, physical function, breast symptoms, or global health quality of life at 12 months (measured using the Quality of Life Questionnaire Core 30 Items [QLQ-C30] and breast cancer specific Quality of Life Questionnaire [QLQ-BR23]; mean scores presented graphically; pain: P = 0.33; physical function: P = 0.76; breast symptoms: P = 0.27; quality of life: P = 0.45).

General adverse effects of adding radiotherapy to breast surgery:

One systematic overview (search date 2001) found limited data on radiotherapy related morbidity and reported that no conclusions could be drawn. A consensus document published in 1998 (mainly of women having breast-conserving surgery or mastectomy with variation in radiotherapy techniques, doses, and fractionation) reported two severe adverse effects of radiotherapy, namely acute pneumonitis (0.7–7.0%) and pericarditis (0–0.3%), and the following long-term adverse effects: significant arm oedema (1% without axillary clearance), radionecrotic rib fracture (1.1–1.5%), and brachial plexopathy (0–1.8%). The risk and severity of adverse effects increased with volume irradiated, total dose received, dose per fraction, previous surgery (e.g. axillary dissection), and radiotherapy techniques that caused overlap in irradiated tissues. A more recent systematic review found that in adding radiotherapy to surgery (breast-conserving surgery or mastectomy) increased the risk of non-breast cancer mortality (mainly from heart disease and lung cancer) compared with no radiotherapy (search date not reported, 63 RCTs; 32,800 people; 15-year risk of non-breast cancer mortality: 16% with radiotherapy v 15% with no radiotherapy, rate ratio 1.12, CI not reported, P = 0.001; rate ratio for heart disease mortality 1.27, CI not reported, P = 0.0001; rate ratio for lung cancer mortality 1.78, CI not reported, P = 0.0004). Three recent RCTs included in the review, with data beyond 10 years, did not find an excess of cardiac deaths. This may be because great care was taken in these RCTs to reduce the radiotherapy dose to the heart and to measure morbidity and mortality from cardiac disease. Studies assessing cosmetic results have mainly been retrospective, using poorly validated outcomes. The effects of social, psychological, and financial disruption from attending 5–6 weeks of radiotherapy have not been assessed well. There is an extremely low reported incidence of radiotherapy induced malignancy, usually soft tissue sarcomas, in the irradiated breast.

Comment

One poor quality RCT (361 women with receptor positive breast cancer) compared the effects of radiotherapy versus tamoxifen versus radiotherapy plus tamoxifen on recurrence rates after breast-conserving surgery. It found no significant difference between treatments after a median follow-up of 5.9 years (risk of recurrence: 0.35 with breast-conserving surgery plus radiotherapy v 0.25 with breast-conserving surgery plus tamoxifen v 0.38 with breast-conserving surgery plus radiotherapy plus tamoxifen v 1.00 with breast-conserving surgery alone [reference group]). RCTs comparing breast-conserving surgery with and without radiotherapy, as well as retrospective case series, have found that prognostic factors for local recurrence after breast-conserving surgery include positive tumour margins, an extensive intraduct component, younger age, lymphovascular invasion, histological grade, and systemic therapy. The only consistent independent risk factor is avoiding radiotherapy. One systematic review of radiotherapy effects in breast cancer by the Swedish Council of Technology Assessment in Health Care, which did not carry out meta-analysis, supports the above conclusions for postmastectomy radiotherapy, radiotherapy after breast-conserving surgery for ductal carcinoma in situ, and for the comparability of breast-conserving surgery with radiotherapy and modified radical mastectomy alone for invasive breast cancer for disease-free survival and overall survival. It concluded that there are conflicting data about the effects of breast conservation surgery plus radiotherapy compared with modified radical mastectomy on local recurrence in people with invasive cancer.

Clinical guide:

For women with early breast cancer, there appears to be no survival difference whether breast-conserving surgery and radiotherapy or mastectomy is chosen. Radiotherapy following breast-conserving therapy may substantially reduce local disease recurrence without detriment to survival when modern radiotherapy techniques are used. The pros and cons of these approaches need to be discussed with the patient prior to surgery. The addition of tamoxifen to breast-conserving surgery, with or without radiotherapy, leads to better local control. The best local control outcome is when breast-conserving surgery, radiotherapy and tamoxifen (if oestrogen receptor-positive) are all part of the treatment. Chemotherapy appears to have little effect on local control in the absence of radiotherapy.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Radiotherapy after mastectomy

Summary

TREATMENT SUCCESS Radiotherapy after mastectomy compared with mastectomy alone: Radiotherapy after mastectomy (alone, with axillary clearance, or with axillary sampling) may be more effective at reducing the 5-year risk of isolated local recurrence in women with node-positive disease ( low-quality evidence ). MORTALITY Radiotherapy after mastectomy compared with mastectomy alone: In women with node-positive disease, radiotherapy may be more effective than no radiotherapy after mastectomy plus axillary clearance at reducing the 15-year risk of breast cancer mortality but may be equally effective as radiotherapy alone or after mastectomy plus axillary sampling (low-quality evidence). NOTE Women at high risk of recurrence include those with more axillary node involvement, larger tumours, higher histological grade, lymphovascular invasion, and involvement of tumour margins. ADVERSE EFFECTS Radiotherapy may be associated with late adverse effects, which are rare, including pneumonitis, pericarditis, arm oedema, brachial plexopathy, radionecrotic rib fracture and radiation induced malignancy.

Benefits

Radiotherapy after mastectomy versus mastectomy alone:

We found one systematic review (search date not reported, 36 RCTs; see comment below), which compared mastectomy versus mastectomy followed by radiotherapy to the chest wall. Seven RCTs were of mastectomy alone (5597 women; 3318 node-negative; 2279 node-positive), four RCTs of mastectomy plus axillary sampling (647 women; 449 node-negative; 198 node-positive), and 25 of mastectomy plus axillary clearance (9933 women; 1428 node-negative; 8505 node-positive). In women with node-positive disease, the review found that radiotherapy significantly reduced 5-year risk of isolated local recurrence compared with no radiotherapy after mastectomy plus axillary clearance (6% with radiotherapy v 23% with no radiotherapy; rate ratio 0.28, CI presented graphically; P less than 0.00001), after mastectomy plus axillary sampling (14% with radiotherapy v 50% with no radiotherapy; rate ratio 0.23, CI presented graphically; P less than 0.00001), and after mastectomy alone (12% with radiotherapy v 34% with no radiotherapy; rate ratio 0.34, CI presented graphically; P less than 0.00001). In women with node-positive disease, radiotherapy significantly reduced the 15-year risk of breast cancer mortality compared with no radiotherapy after mastectomy plus axillary clearance (55% with radiotherapy v 60% with no radiotherapy; rate ratio 0.89, CI presented graphically; P = 0.0002). However, there was no significant difference in 15-year risk of breast cancer mortality between radiotherapy and no radiotherapy after mastectomy alone (rate ratio 0.81, CI presented graphically; P = 0.3) or after mastectomy plus axillary sampling (rate ratio 0.92, CI presented graphically; P = 0.2). In women with node-negative disease, the review found that radiotherapy significantly reduced 5-year risk of isolated local recurrence compared with no radiotherapy after mastectomy plus axillary clearance (2.3% with radiotherapy v 6.3% with no radiotherapy; rate ratio 0.41, CI presented graphically; P = 0.0002), after mastectomy plus axillary sampling (6.1% with radiotherapy v 24.5% with no radiotherapy; rate ratio 0.32, CI presented graphically; P less than 0.00001), and after mastectomy alone (6% with radiotherapy v 23% with no radiotherapy; rate ratio OR 0.33, CI presented graphically; P less than 0.00001). In women with node-negative disease, radiotherapy increased 15-year risk of breast cancer mortality compared with no radiotherapy after mastectomy plus axillary clearance (31% with radiotherapy v 28% with no radiotherapy; rate ratio 1.26, CI presented graphically; P = 0.01). However, it found no significant difference in 15-year risk of breast cancer mortality between radiotherapy and no radiotherapy after mastectomy plus axillary sampling (rate ratio 0.88, CI presented graphically; P = 0.4), or after mastectomy alone (rate ratio 0.98, CI presented graphically; P = 0.8).

Radiotherapy after mastectomy versus mastectomy plus adjuvant chemotherapy:

Two RCTs reported in one report compared mastectomy plus radiotherapy versus mastectomy plus chemotherapy (one trial was in premenopausal women [547 participants] and one trial was in postmenopausal women [679 participants]). It found that radiotherapy reduced loco-regional recurrences among both pre and postmenopausal women compared with chemotherapy (relative hazard [RH] radiotherapy group v chemotherapy group: premenopausal women, RH 0.67, 95% CI 0.44 to 1.0, P less than 0.05; postmenopausal women, RH 0.43, 95% CI 0.30 to 0.63, P less than 0.001). Among premenopausal women, it found that distant metastases occurred less frequently in the chemotherapy group compared with the radiotherapy group (HR 1.68, 95% CI 1.3 to 2.2; P less than 0.001) resulting in an improved recurrence-free survival (P = 0.04). It found that overall survival in premenopausal women was better with chemotherapy but the difference between groups was not statistically significant (cumulative survival at 15 years: 50% in chemotherapy group v 44% in radiotherapy group; reported as no significant difference, P value not provided). Among postmenopausal women, it reported that there was no significant difference between the treatment groups in terms of recurrence-free (P = 0.28) or overall survival (P = 0.38).However, the period of enrollment of the RCTs were long (between 1976 to 1990), inclusion criteria and randomisation were modified over the course of the studies, and the chemotherapy regimen used was modfied on a number of occasions. Also, the report noted that the trials had to some extent, a mainly historical interest since some of the regimens tested were no longer relevant to routine medical practice.

Harms

See harms of radiotherapy after breast-conserving surgery. Three RCTs included in the review of total nodal irradiation after mastectomy in high-risk disease found no significant increase in cardiac mortality.

Comment

The RCTs in the large systematic review were heterogeneous, varying in randomisation processes, areas irradiated, use of systemic treatment, radiotherapy doses, fractionation, and treatment schedules. We found little good evidence to identify which women should have postmastectomy radiotherapy to prevent local recurrence. One review of retrospective data found that extent of axillary node involvement, larger tumour size, higher histological grade, presence of lymphovascular invasion, and involvement of tumour margins increased the absolute risk of local recurrence or mortality. One non-systematic review (36 RCTs, 13,199 women) comparing radiotherapy after mastectomy versus no radiotherapy assessed the effects of dose and target volume on overall survival. It found that radiotherapy after mastectomy delivered with an optimal biologically equivalent dose to an appropriate clinical target volume significantly increased overall survival at up to 10 years. We found no evidence that reduction in relative risk of local recurrence was affected by age, nodal status, receptor status, tumour grade, or tumour size, or that the effect of radiotherapy on mortality varied significantly with extent of surgery, type of radiotherapy (megavoltage or orthovoltage), years the RCTs commenced or completed recruitment, or whether systemic treatment was given.

Clinical guide:

Current expert opinion and evidence based treatment guidelines suggest that high-risk patients for loco-regional recurrence should be recommended for radiotherapy. These patients would include those with lymph node positivity (particularly when more than 3 axillary nodes are involved with tumour), positive surgical margins post-mastectomy and larger tumour size (especially more than 5cm). Post-mastectomy radiotherapy should also be considered when patients have lesser nodes involved, or smaller tumours, particularly when there are a number of other adverse features (such as 1–3 nodes, receptor negativity, lymphvascular space invasion), though clinical trials are currently underway to specifically address this question, including the use of radiotherapy in elderly patients.

Substantive changes

Radiotherapy after mastectomy (under question on treatments for primary operable breast cancer) One report of 2 RCTs added; benefits and harms data enhanced, categorisation of 'radiotherapy after mastectomy in women at high risk of local recurrence' as Beneficial and 'radiotherapy after mastectomy in women not at high risk of local recurrence' as Trade off between benefits and harms unchanged.

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Radiotherapy after mastectomy in women not at high risk of local recurrence

Summary

TREATMENT SUCCESS Radiotherapy after mastectomy compared with mastectomy alone: Radiotherapy after mastectomy (alone, with axillary clearance, or with axillary sampling) may be more effective at reducing the 5-year risk of isolated local recurrence in women with node-negative disease ( low-quality evidence ). MORTALITY Radiotherapy after mastectomy compared with mastectomy alone: In women with node-negative disease, radiotherapy may increase the15-year risk of breast cancer mortality compared with no radiotherapy after mastectomy plus axillary clearance but may be equally effective as radiotherapy after mastectomy plus axillary sampling (low-quality evidence). NOTE Women at high risk of recurrence include those with more axillary node involvement, larger tumours, higher histological grade, lymphovascular invasion, and involvement of tumour margins. ADVERSE EFFECTS Radiotherapy may be associated with late adverse effects, which are rare, including pneumonitis, pericarditis, arm oedema, brachial plexopathy, radionecrotic rib fracture and radiation induced malignancy.

Benefits

Radiotherapy after mastectomy versus mastectomy alone:

See benefits of radiotherapy after mastectomy.

Harms

See harms of radiotherapy after breast-conserving surgery. Three RCTs included in the review of total nodal irradiation after mastectomy in high-risk disease found no significant increase in cardiac mortality.

Comment

We found little good evidence to identify which women should have postmastectomy radiotherapy to prevent local recurrence. One review of retrospective data found that extent of axillary node involvement, larger tumour size, higher histological grade, presence of lymphovascular invasion, and involvement of tumour margins increased the absolute risk of local recurrence or mortality. One non-systematic review (36 RCTs, 13,199 women) comparing radiotherapy after mastectomy versus no radiotherapy assessed the effects of dose and target volume on overall survival. It found that radiotherapy after mastectomy delivered with an optimal biologically equivalent dose to an appropriate clinical target volume significantly increased overall survival at up to 10 years. We found no evidence that reduction in relative risk of local recurrence was affected by age, nodal status, receptor status, tumour grade, or tumour size, or that the effect of radiotherapy on mortality varied significantly with extent of surgery, type of radiotherapy (megavoltage or orthovoltage), years the RCTs commenced or completed recruitment, or whether systemic treatment was given.

Clinical guide:

Current expert opinion and evidence based treatment guidelines suggest that high-risk patients for loco-regional recurrence should be recommended for radiotherapy. These patients would include those with lymph node positivity (particularly when more than 3 axillary nodes are involved with tumour), positive surgical margins post-mastectomy and larger tumour size (especially more than 5cm). Post-mastectomy radiotherapy should also be considered when patients have lesser nodes involved, or smaller tumours, particularly when there are a number of other adverse features (such as 1–3 nodes, receptor negativity, lymphvascular space invasion), though clinical trials are currently underway to specifically address this question, including the use of radiotherapy in elderly patients.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Primary chemotherapy

Summary

TREATMENT SUCCESS Primary chemotherapy compared with adjuvant chemotherapy: Primary chemotherapy seems to more effective at reducing mastectomy rates but not at reducing loco-regional recurrences at 4 years ( moderate-quality evidence ). MORTALITY Primary chemotherapy compared with adjuvant chemotherapy: Primary chemotherapy and adjuvant chemotherapy seem to be equally effective at improving overall survival (moderate-quality evidence). ADVERSE EFFECTS Adverse effects of chemotherapy include fatigue, nausea and vomiting, hair loss, bone marrow suppression, neuropathy, and gastrointestinal disturbance. Chemotherapy may impair fertility and ovarian function.

Benefits

Survival:

We found no systematic review but found five RCTs, which compared primary chemotherapy versus adjuvant chemotherapy. The first RCT (272 women with tumours 3 cm or more in whom mastectomy was indicated) compared primary EVMTV (epirubicin, vincristine, mitomycin C, thiotepa, vindesine) chemotherapy versus mastectomy followed by the EVMTV regimen. At an initial median follow-up of 34 months, a significant survival difference was reported in favour of primary chemotherapy (results presented graphically; log rank P = 0.04). However, the final analysis at 124 months showed that the survival improvement was no longer significant, with survival of about 55% in both groups. The second RCT (414 women) compared four cycles of FAC (fluorouracil, adriamycin [doxorubicin], and cyclophosphamide) as primary or adjuvant chemotherapy. At 54-months' follow-up, the primary chemotherapy group had a better overall survival (86% with primary v 68% with adjuvant; P = 0.039); however, a subsequent analysis at 105 months did not show a long-term survival benefit. The third RCT (309 women) compared four cycles of primary MM (mitoxantrone [mitozantrone], methotrexate) chemotherapy followed by surgery and four further cycles of MM versus surgery followed by eight cycles of adjuvant MM. At 48-months' follow-up, there was no difference in survival between the primary and adjuvant groups (84% with primary v 82% with adjuvant; reported as not significant). The fourth, and largest, RCT (National Surgical Adjuvant Breast and Bowel Project [NSABP] 18), in which 1523 women were randomised to four cycles of AC (adriamycin [doxorubicin], cyclophosphamide) as primary or adjuvant chemotherapy, found identical survival rates (67%) in the two groups at 60 months. The fifth RCT (698 women) compared four cycles of fluorouracil, epirubicin, and cyclophosphamide, as primary or adjuvant chemotherapy. It found no significant difference between primary and adjuvant chemotherapy in overall survival (82% with primary v 84% with adjuvant; HR 1.16, 95% CI 0.83 to 1.63), progression-free survival (65% with primary v 70% with adjuvant; HR 1.15, 95% CI 0.89 to 1.48), or loco-regional recurrence at 4 years (21.5% with primary v 17.8% with adjuvant; HR 1.13, 95% CI 0.70 to 1.81).

Mastectomy rates:

We found no systematic review but found three RCTs, which compared mastectomy rates with primary chemotherapy versus adjuvant chemotherapy. The first RCT (309 women receiving MM chemotherapy) found that primary chemotherapy significantly reduced the mastectomy rate compared with adjuvant chemotherapy (13% with primary v 28% with adjuvant; P less than 0.005). The second RCT (1523 women receiving AC chemotherapy) found that breast conservation rates were lower in the adjuvant arm (60% with adjuvant v 67% with primary), although this was not significant. The third RCT assessed 272 women at diagnosis in terms of the recommended surgical procedure, and two of three women who were initially advised to have mastectomy were able to have breast-conserving surgery after primary chemotherapy with FAC.

Harms

We found no evidence that primary chemotherapy has a negative impact on survival. None of the RCTs examining effects on mastectomy rates reported a significantly higher local recurrence rate with primary chemotherapy compared with adjuvant chemotherapy. See harms of adjuvant combination chemotherapy.

Comment

We found no evidence to support the use of primary chemotherapy to improve the chances of survival for operable breast cancers outside the context of an RCT. With an increased number of conservative operations being performed after downstaging by primary chemotherapy for large primary tumours, there are theoretical concerns that this may result in an increased rate of local recurrence. However, primary chemotherapy can lead to a reduction in the requirement for mastectomy and, as such, an increase in breast-conserving surgery and sequential tissue biopsies can be obtained within the context of clinical trials. In the three RCTs of women with operable breast cancer receiving breast-conserving surgery, use of primary chemotherapy has not been associated with a significant increase in the rate of local recurrence.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Total nodal radiotherapy

Summary

TREATMENT SUCCESS Radiotherapy after mastectomy (including total nodal irradiation) compared with mastectomy alone: Radiotherapy (including total nodal irradiation) after mastectomy may be more effective at reducing the 5-year risk of isolated local recurrence in women with node-positive and node-negative disease ( very low-quality evidence ). MORTALITY Radiotherapy after mastectomy (including total nodal irradiation) compared with mastectomy alone: We don't know whether radiotherapy (including total nodal irradiation) after mastectomy is more effective at reducing the 15-year risk of breast cancer mortality in women with node-positive and node-negative disease (very low-quality evidence).

Benefits

We found one systematic review (search date not reported, 36 RCTs of postmastectomy radiotherapy), which included 26 RCTs of total nodal irradiation to the internal mammary chain, supraclavicular fossa, and axilla. It found that postoperative radiotherapy reduced loco-regional recurrence in people with node-negative and node-positive disease. However, it found conflicting evidence on breast cancer mortality. See benefits of radiotherapy after mastectomy. These results should be interpreted with caution because they include some RCTs that did not give total nodal irradiation.The benefits of treating any one particular nodal area has not been addressed adequately to date.

Harms

See harms of radiotherapy to the internal mammary chain; See harms of radiotherapy to the ipsilateral supraclavicular fossa; and See harms of axillary management. Three RCTs included in the review found no increase in cardiac mortality because of radiotherapy.

Comment

Clinical guide:

Total nodal radiotherapy may reduce local recurrence but with an uncertain effect on survival. The RCTs that showed a survival benefit for post-mastectomy radiotherapy usually used nodal irradiation to the internal mammary chain, supraclavicular fossa and axilla as well as chest wall radiation. However, what the relative benefits and risks associated with each of these treatment areas remains unanswered. In terms of risk, the greatest risk of recurrence in most high-risk patients are the chest wall with smaller recurrence risks for supraclavicular fossa, internal mammary chain and axilla. RCTs are currently being conducted to answer some of these questions.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Axillary management

Summary

TREATMENT SUCCESS Axillary clearance compared with axillary radiotherapy: Axillary clearance (level I, II and III dissection) and axillary radiotherapy (regardless of axillary nodal status) seem to be equally effective at 10 years at reducing recurrence ( moderate-quality evidence ). Total axillary dissection plus sentinel node biopsy compared with sentinel node biopsy alone (with axillary node dissection only if the sentinel contains metastases): We don't know whether total axillary dissection plus sentinel node biopsy is more effective at reducing breast cancer events, post-operative pain, arm swelling, seroma formation and loss of sensation ( low-quality evidence ). MORTALITY Axillary clearance compared with axillary sampling: Axillary clearance (level I, II and III dissection) and four node axillary sampling (followed by axillary radiotherapy in women found to be node-positive) seem to be equally effective at improving survival (moderate-quality evidence). Axillary clearance compared with axillary radiotherapy: Axillary clearance (level I, II and III dissection) and axillary radiotherapy (regardless of axillary nodal status) seem to be equally effective at 10 years at reducing the annual risk of mortality (moderate-quality evidence). Total axillary dissection plus sentinel node biopsy compared with sentinel node biopsy alone (with axillary node dissection only if the sentinel contains metastases): We don't know whether total axillary dissection plus sentinel node biopsy is more effective at improving overall survival in women undegoing quadrantectomy (low-quality evidence). NOTE There is consensus that axillary clearance reduces regional recurrence compared with no axillary management. We found no direct information about the effects of radiotherapy in addition to axillary clearance (level I and II or level I, II, or III dissection) in regional disease control. ADVERSE EFFECTS Axillary surgery has been associated with seroma formation, arm swelling, damage to the intercostobrachial nerve and shoulder stiffness.

Benefits

Axillary clearance versus axillary sampling:

We found no systematic review but found one RCT (466 women) in women having breast-conserving surgery. The RCT compared complete axillary clearance (level I, II, and III dissection) versus four node axillary sampling followed by axillary radiotherapy if the nodes were involved. It found that axillary sampling was associated with improved survival compared with axillary clearance, but the difference was not significant (overall survival figures presented graphically, P = 0.2; estimated 5-year survival: 88.6% with axillary sampling v 82.1% with axillary clearance). Rates of node positivity were similar in both groups.

Axillary clearance versus axillary radiotherapy:

We found one systematic review (search date not reported, 8 RCTs, 4370 women) comparing axillary clearance (level I, II, and III dissection) versus axillary radiotherapy (regardless of axillary nodal status). It found no significant difference in annual risk of mortality over 10 years or in recurrence (mortality: OR 0.96, CI presented graphically, P = 0.3; recurrence: OR 1.01, CI not reported). Radiotherapy was associated with fewer isolated local recurrences, but this difference did not reach significance (OR 0.85, CI not reported; P = 0.06.

Axillary clearance alone versus axillary clearance plus radiotherapy:

We found no systematic review or RCTs assessing the effect of radiotherapy in addition to axillary clearance (level I and II, or level I, II, and III dissection) in regional control of disease.

Axillary dissection versus sentinel node biopsy:

See benefits of sentinel node biopsy.

Harms

Axillary clearance versus axillary sampling:

Adverse effects of axillary surgery include seroma formation, arm swelling, damage to the intercostobrachial nerve, and shoulder stiffness. We found one RCT, which compared the morbidity of different axillary procedures. It compared complete axillary clearance (level I, II, and III dissection) versus four node axillary sampling, followed by radiotherapy if the nodes were involved. It found that the rate of arm swelling was higher after clearance than after sampling, whether or not women received postoperative radiotherapy (at 3 years, forearm girth was significantly greater with clearance than with sampling alone [P = 0.005] or sampling plus radiotherapy [P = 0.04]). After removal of axillary drains, 25–50% of women who had had a level I and II, or level I, II, and III axillary clearance developed seromas requiring aspiration. The RCT found that women who received axillary clearance or axillary sampling plus radiotherapy (not to the shoulder joint) had significantly reduced shoulder movement compared with women receiving axillary sampling alone at 6 months (clearance v sampling alone: P = 0.003; sampling plus radiotherapy v sampling alone: P = 0.004). However, by 3 years, the axillary clearance group had improved and was not significantly different from the sampling alone group (P = 0.1).

Arm lymphoedema:

One Australian systematic review (search date 1996) of lymphoedema prevalence, risks, and management found that, although current information is of poor quality, the combination of axillary dissection (to or beyond level II) and axillary radiotherapy was associated with a risk of lymphoedema of 12–60%, with most studies suggesting that at least a third of women are affected. Studies of axillary sampling followed by irradiation found lower rates of lymphoedema (6–32%), and for axillary sampling alone, the rates were lower still (0–21%). Studies of dissection beyond level I found rates of lymphoedema between 0–42%, with most studies reporting a rate of 20–30% 1 year after operation. In women who receive axillary radiotherapy without axillary surgery, the overall lymphoedema rate is about 8%.

Comment

Axillary staging:

Both clearance and sampling provide important prognostic information on which decisions on local and systemic treatment can be based. A decision on axillary management should be based on the risk of involvement of axillary nodes (which varies according to tumour size, grade, and the presence of vascular/lymphatic invasion) and potential treatment related morbidity. Two retrospective cohort studies found that level I dissection accurately assessed axillary lymph node status, providing that at least 10 nodes were removed. One RCT found that a sample of four nodes provided sufficient information to categorise an axilla as histologically positive or negative. Removal of nodes at level I and II, or removal of all nodes below the axillary vein (level I, II, and III), accurately stages the axilla.

Clinical guide:

The probability of axillary lymph node metastasis should be assessed and a selective surgical approach to the axilla (clearance or sampling) employed balancing local disease control and progostic information against morbidity.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Different primary chemotherapy regimens

Summary

TREATMENT SUCCESS Standard compared with dose intensified anthracycline based regimens: A standard based regimen (cyclophosphamide, epirubicin, fluorouracil) and a dose intensified regimen (epirubicin, cyclophosphamide, filgrastim) seem to be equally effective at increasing time to progression in women with locally advanced breast cancer ( moderate-quality evidence ). FAC regimen (fluorouracil, adriamycin [doxorubicin], and cyclophosphamide) compared with single agent paclitaxel: We don't know whether FAC regimens are more effective at improving response rates ( low-quality evidence ). MPEMi (methotrexate, cisplatin, etoposide, mitomycin C), MPEpiE (methotrexate, cisplatin, epirubicin, etoposide), and MPEpiV (methotrexate, cisplatin, epirubicin, vincristine) regimens compared with each other: We don't know which regimen is more effective at improving response rates (low-quality evidence). Sequencing of anthracycline based chemotherapy and docetaxel: Further treatment with docetaxel may be more effective than further treatment with CVAP [cyclophosphamide, adriamycin {doxorubicin}, vincristine, prednisolone]) at improving clinical response rates in women who have acheived complete or partial response to four cycles of CVAP (low-quality evidence). Primary treatment with docetaxel may be more effective than no primary treatment in women who have received four cycles of AC (adriamycin [doxorubicin], cyclophosphamide) at improving clinical complete response rates (low-quality evidence). Intra-arterial compared with intravenous administration: Although intra-arterial epirubicin may be more effective than intravenous epirubicin at improving response rates, this benefit does not lead to an improvement in survival in women with locally advanced breast cancer (low-quality evidence). MORTALITY Standard compared with dose intensified anthracycline based regimens: A standard based (cyclophosphamide, epirubicin, fluorouracil) and a dose intensified anthracycline regimen (epirubicin, cyclophosphamide, filgrastim) seem to be equally effective at increasing time to death, or 5-year survival rates in women with locally advanced breast cancer (moderate-quality evidence). FAC regimen (fluorouracil, adriamycin [doxorubicin], and cyclophosphamide) compared with single agent paclitaxel: We don't know whether FAC regimens are more effective at improving survival (low-quality evidence). NOTE We found no clinically important results about the effects of navelbine based regimens in women with breast cancer.

Benefits

Standard versus dose intensified anthracycline based regimens:

We found no systematic review but found one RCT. The RCT (448 women with locally advanced breast cancer) compared a CEF (cyclophosphamide, epirubicin, fluorouracil) regimen versus a dose intensified ECFi (epirubicin, cyclophosphamide, filgrastim) regimen. It found no significant difference between the regimens in time to progression (recurrence or death) or 5-year survival (median time to progression: 34 months with CEF v 33.7 months with ECFi, P = 0.68; 5-year survival: 53% with CEF v 51% with ECFi, P = 0.94). Complete clinical response rates were similar with both regimens (31% with CEF v 27% with ECFi; P value and RR not reported).

FAC regimen versus paclitaxel:

We found no systematic review but found one RCT. The RCT (174 women in the USA) compared conventional FAC (fluorouracil, adriamycin [doxorubicin], and cyclophosphamide) versus single agent paclitaxel, and found similar response rates in both groups (79% with FAC v 80% with paclitaxel), with no significant difference in survival rates.

Comparison between MPEMi (methotrexate, cisplatin, etoposide, mitomycin C), MPEpiE (methotrexate, cisplatin, epirubicin, etoposide), and MPEpiV (methotrexate, cisplatin, epirubicin, vincristine) regimens:

We found no systematic review but found one RCT. The RCT (101 women) compared three different primary chemotherapy regimens: MPEMi versus MPEpiE versus MPEpiV. It found that the response rate was 89% in all three groups.

Sequencing of anthracycline based chemotherapy and docetaxel:

We found no systematic review but found two RCTs. The first RCT (104 women who had achieved complete or partial clinical response to four cycles of CVAP [cyclophosphamide, adriamycin {doxorubicin}, vincristine, prednisolone]) compared a further four cycles of CVAP versus four cycles of docetaxel. It found that further treatment with docetaxel significantly improved clinical complete response rate compared with further CVAP (clinical complete response rate: 85% with docetaxel v 64% with CVAP; P = 0.03). In the second RCT (2411 people), all women received four cycles of AC (adriamycin [doxorubicin], cyclophosphamide) and were then randomly allocated to three regimens: surgery alone, four cycles of docetaxel followed by surgery, or surgery followed by four cycles of docetaxel. The preliminary results of this RCT found that, at the time of surgery, primary docetaxel improved clinical complete response rate compared with no primary docetaxel (65% with docetaxel v 40% with no docetaxel; P less than 0.001). The final results, which will also examine the effects of adjuvant docetaxel, are awaited.

Navelbine based regimens:

We found no systematic reviews or fully published RCTs (see comment below).

Comparison between routes of administration:

We found no systematic review but found one RCT. The RCT (73 women with locally advanced breast cancer) compared routes of administration of primary chemotherapy. It compared no primary treatment, primary intravenous epirubicin, or primary intra-arterial epirubicin. The RCT found that response rates were higher in women receiving intra-arterial epirubicin compared with intravenous epirubicin (68% with intra-arterial epirubicin v 36% with intravenous epirubicin; P less than 0.05); however, this was not associated with a survival benefit.

Harms

Standard versus dose intensified anthracycline based regimens:

There were similar numbers of serious adverse events requiring hospitalisation in the groups (60 events with CEF v 68 events with ECFi; P value not reported). The dose intensified ECFi regimen increased nausea and vomiting, and induced more grade 3 and 4 anaemia, but there were fewer febrile neutropenic episodes compared with CEF (AR for nausea: 11.8% with CEF v 22.3% with ECFi; vomiting: 10.9% with CEF v 18.8% with ECFi; anaemia: 16.3% with CEF v 50.9% with ECFi; febrile neutropenia: 14% with CEF v 8.4% with ECFi; no P values reported for comparisons).

FAC versus paclitaxel:

In the RCT comparing FAC versus paclitaxel, rates of septic neutropenia (53% with paclitaxel v 21% with FAC) and granulocyte colony stimulating factor usage (56% with paclitaxel v 25% with FAC) were higher in women taking paclitaxel.

Comment

More research is needed to determine the optimal regimen for primary treatment. We found little evidence in the literature comparing different combinations, but anthracycline based combinations probably remain the treatment of choice, with dose intensification not proved to confer additional clinical benefit. Ongoing RCTs are investigating the role of taxane sequencing after anthracycline based treatment (National Surgical Adjuvant Breast and Bowel Project [NSABP] 27) and anthracycline in combination with fluorouracil infusion.

Navelbine based regimens:

We found one RCT (published as an abstract, 147 women), which compared AC, NM (navelbine, mitoxantrone), and NE (navelbine, epirubicin). Response rates were 65% with AC, 73% with NM, and 86% with NE. The time to outcome was not reported. The trial is ongoing, although the NM arm has been stopped because of haematological toxicity.Clinical trials are currently underway addressing different regimens in the primary setting, including the use of taxane-platinum regimens which may be associated with lower long-term toxicities, including leukaemia and cardiac failure when compared to anthracyclines.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Less than whole-breast radiotherapy after breast-conserving surgery

Summary

TREATMENT SUCCESS Less than whole-breast radiotherapy compared with whole breast radiotherapy: We don't know whether less than whole-breast radiotherapy after breast-conserving surgery is more effective at reducing recurrences at 8 years ( moderate-quality evidence ). MORTALITY Less than whole-breast radiotherapy compared with whole breast radiotherapy: We don't know whether tumour bed radiotherapy after breast-conserving surgery is more effective at 8 years at improving survival ( low-quality evidence ).

Benefits

We found one systematic review, one additional RCT, and one subsequent RCT of less than whole-breast radiotherapy after breast-conserving surgery. The review (search date 2002) compared intraoperative radiotherapy versus standard postoperative radiotherapy in women receiving breast-conserving surgery, and found no fully published RCTs (see comment). The additional RCT found that localised-field radiotherapy significantly increased breast recurrence compared with wide-field radiotherapy at 8 years (713 women; 69/353 [20%] with localised-field radiotherapy v 35/355 [10%] with wide-field radiotherapy; P = 0.00008). It found no significant difference in disease-specific survival between localised field radiotherapy and wide field radiotherapy at 8 years (73% with localised-field radiotherapy v 72% with wide-field radiotherapy; P = 0.91). The subsequent RCT found no significant difference in local recurrence, distant recurrence, or survival between tumour bed radiotherapy and whole-breast radiotherapy at median follow-up of 8 years (174 women; local recurrence: 10/84 [12%] with tumour bed radiotherapy v 4/90 [4%] with whole-breast radiotherapy, P = 0.07; distant recurrence: 19/84 [23%] with tumour bed radiotherapy v 24/90 [27%] with radiotherapy, P = 0.70; mortality: 25/84 [30%] with tumour bed radiotherapy v 24/90 [27%] with whole-breast radiotherapy, P = 0.75). The RCT aimed to recruit 400 women, but it closed early because of problems with recruitment; therefore, the RCT may have been underpowered to detect a clinically important difference.

Harms

The additional RCT found that localised-field radiotherapy increased marked fibrosis and marked telangiectasia compared with wide-field radiotherapy (fibrosis: 51/353 [15%] with localised-field radiotherapy v 18/355 [5%] with wide-field radiotherapy; telangiectasia: 116/353 33%] with localised-field radiotherapy v 43/355 [12%] with wide-field radiotherapy; significance assessment not performed). The differences may be related to radiation fractionation, rather than the target field area. The subsequent RCT gave no information on adverse effects.

Comment

The review found one RCT, which was published in abstract form only, making it difficult to assess study quality.The abstract reported similar levels of “good to excellent” cosmetic result with intraoperative radiotherapy plus postoperative radiotherapy and postoperative therapy plus external beam boost radiotherapy. Local cancer recurrence, disease-free survival, and overall survival results were not reported in the abstract. Less than whole-breast radiotherapy may reduce toxicity because less tissue in the breast, chest wall, and thorax is irradiated. It may also be possible to take advantage of the radiobiological effects of irradiating a smaller volume in order to deliver the treatment in a shorter period of time. If an intraoperative technique is used, it may be possible to place radiotherapy dose more accurately within the excision area. There are several techniques under consideration for less than whole-breast radiotherapy: intraoperative brachytherapy; delayed brachytherapy, intraoperative external beam techniques (electrons, low energy x rays), and delayed external beam techniques. We found only two fully published RCTs, but there are several international trials of different technologies underway. Each technique of less than whole-breast radiotherapy will be associated with its own adverse effects. Invasive intraoperative or implantation techniques may require anaesthesia and expose the person to wound infections. Radiotherapy effects on skin, subcutaneous fat, muscle, and ribs will depend on technique, dose, and fractionation. Localisation of a limited volume of irradiation may also be a problem, and various radiological, clinical, and ultrasonographic techniques are advocated to overcome this problem.

Clinical guide:

Studies are currently attempting to address the question as to whether partial breast irradiation may be sufficient treatment for some sub-groups of breast cancer. Until those studies are complete, and the follow-up data mature, it is not possible to recommend partial breast irradiation as an appropriate treatment for breast cancer outside of a properly conducted trial.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Radiotherapy to the internal mammary chain

Summary

RELAPSE Compared with no internal mammary chain irradiation: We don't know whether radiotherapy to the internal mammary chain is more effective at reducing relapses in women treated with breast-conserving surgery and radiotherapy ( moderate-quality evidence ). MORTALITY Compared with no internal mammary chain irradiation: We don't know whether radiotherapy to the internal mammary chain is more effective at improving survival in women treated with breast-conserving surgery and radiotherapy (moderate-quality evidence). NOTE Radiotherapy to the internal mammary chain may increase radiation induced cardiac morbidity.

Benefits

We found no systematic review but found one RCT. The RCT (270 women treated with breast-conserving surgery and radiotherapy) compared internal mammary chain irradiation versus no internal mammary chain irradiation. At a median follow-up of 2.7 years, there was no significant difference in relapse or survival (numbers not reported).

Harms

See harms of radiotherapy after breast-conserving surgery. Radiotherapy to the internal mammary chain is more likely to affect the heart compared with other types of radiotherapy.

Comment

The risk of internal mammary chain node involvement is related to the location and size of the primary tumour and, most importantly, histopathological axillary nodal status. Up to 30% of women with axillary involvement will also exhibit internal mammary chain nodal metastases. Central or medial breast cancers are more likely to metastasise to the internal mammary chain, as are larger tumours and tumours that have axillary node involvement. The risk of internal mammary chain recurrence is low, and after modified radical mastectomy alone is 2%. Modern radiotherapy planning and delivery should involve an assessment of the position and depth of the internal mammary chain nodes to be treated (using computerised tomography or ultrasound), and computer assisted placement, arrangement, and determination of dose distribution; these technologies were unavailable at the time of most RCTs included in the reviews. Recent indirect evidence from RCTs suggests improved survival from nodal irradiation (including radiation to the internal mammary chain) after modified radical mastectomy combined with systemic treatment. Another RCT of internal mammary chain irradiation has started recently (sponsored by the European Organisation for Research and Treatment of Cancer [EORTC]).

Clinical guide:

Internal mammary node chain irradiation remains of uncertain benefit and should be the subject of RCTs.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Radiotherapy to the ipsilateral supraclavicular fossa

Summary

TREATMENT SUCCESS Compared with no radiotherapy: We don't know whether radiotherapy to the chest wall and lymph nodes is more effective at reducing recurrence in the supraclavicular fossa at 123 months in postmenopausal women who have received tamoxifen after mastectomy and are at high risk of local recurrence ( moderate-quality evidence ). NOTE We found no direct information about radiotherapy to the ipsilateral supraclavicular fossa in the treatment of women with breast cancer. Morbidity associated with irradiation of the supraclavicular fossa is rare and, where it occurs, is mild and temporary.

Benefits

We found no systematic review or RCTs on radiotherapy to the ipsilateral supraclavicular fossa. One systematic review (search date not reported) found that postoperative radiotherapy to the chest wall and lymph nodes was associated with reduced loco-regional recurrence: see benefits of radiotherapy after breast-conserving surgery; radiotherapy after mastectomy; and radiotherapy to the internal mammary chain. RCTs indicate reduced recurrence in the supraclavicular fossa following radiotherapy to the peripheral lymphatics. One RCT in postmenopausal women at high risk of local recurrence who received tamoxifen after mastectomy found that radiotherapy (chest wall and total nodal irradiation) was associated with lower recurrence in the supraclavicular fossa at a median follow-up of 123 months (9/689 [1.3%] with radiotherapy v 37/686 [5.4%] with no radiotherapy; CI not reported).

Harms

The acute morbidity of irradiation to the supraclavicular fossa is mild and includes temporary upper oesophagitis in nearly all women. The risk of radiation pneumonitis increases with the volume of lung irradiated. Treatment irradiates the lung apex in addition to any lung included in the breast or chest wall fields. Possible late morbidity includes brachial plexopathy, but this should not exceed 1.8% if attention is paid to limiting the total dose to 50 Gy, limiting the dose per fraction to 2 Gy or less, and avoiding field junction overlaps. Late apical lung fibrosis is common and usually of no clinical importance. Demyelination of the cervical cord is an extremely rare complication of supraclavicular fossa radiotherapy.

Comment

Clinical guide:

There is insufficient evidence to give a definite recommendation for or against supraclavicular fossa node irradiation. In the absence of good quality randomised trial data it might be considered reasonable to include the supraclavicular fossa for patients that are deemed at high risk of involvement. This would be in the group with 3 or more nodes involved in their axillary surgery.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Sentinel node biopsy

Summary

TREATMENT SUCCESS Sentinel node biopsy plus total axillary dissection compared with sentinel node biopsy alone (with axillary node dissection only if the sentinel contains metastases): We don't know whether sentinel node biopsy is more effective at reducing breast cancer events, postoperative pain, arm swelling, seroma formation and loss of sensation ( low-quality evidence ). MORTALITY Sentinel node biopsy plus total axillary dissection compared with sentinel node biopsy alone (with axillary node dissection only if the sentinel contains metastases): We don't know whether sentinel node biopsy is more effective at improving overall survival in women undergoing quadrantectomy (low-quality evidence).

Benefits

Sentinel node biopsy versus axillary procedures (sampling or clearance):

We found two RCTs, which compared sentinel node biopsy plus total axillary dissection versus sentinel node biopsy alone (with axillary node dissection only if the sentinel node contained metastases). In the first RCT, both groups received radiation therapy after surgery for 8 weeks. It found that sentinel node biopsy improved postoperative pain and arm mobility versus axillary dissection and non-significantly reduced breast cancer events (516 women, aged 45–70 years, primary tumour 2 cm or less, undergoing quadrantectomy; no pain at 24 months: 92/100 with sentinel node biopsy v 61/100 with axillary dissection, P value not reported; 80–100% arm mobility: 100/100 with sentinel node biopsy v 79/100 with axillary dissection, P value not reported; breast cancer events: 13/259 with sentinel node biopsy v 21/257 with axillary dissection, P = 0.13; see comment below). It found no significant difference in overall survival between treatments (P = 0.15). The second RCT found that sentinel node biopsy significantly reduced postoperative arm swelling (298 people with early breast cancer; OR for subjective arm swelling 0.30, 95% CI 0.18 to 0.68; P = 0.004) and loss of sensation (AR of no loss of sensation: 34% with sentinel node biopsy v 16% with axillary dissection; OR 2.7, 95% CI 1.5 to 5.0) compared with axillary dissection. Sentinel node biopsy did not significantly reduce seroma formation overall, but did significantly reduce seroma formation in node-negative women, who did not need subsequent axillary dissection (overall risk of seroma formation: 14% with sentinel node biopsy v 21% with axillary dissection, OR 0.60, 95% CI 0.33 to 1.11; risk of seroma formation in node-negative women: 11% with sentinel node biopsy v 24% with axillary dissection, OR 0.37, 95% CI 0.16 to 0.82). It found a trend towards reduced impairment of shoulder mobility and psychological morbidity (Beck Depression Inventory, Global Severity Index of the Brief Symptom Inventory, State-Trait Anxiety Inventory, Mental Adjustment to Cancer Scale) with sentinel node biopsy soon after the operation, but the differences were not consistently significant over different measures, and differences between groups reduced with time.

Harms

One prospective cohort study found that the blue dye used in sentinel lymph node mapping caused some allergic reaction (31/1728 [2%]). The study found a trend toward fewer allergic reactions with smaller volumes of blue dye, although this was not significant.

Sentinel node biopsy versus axillary procedures:

Neither RCT reported on harms specific to sentinel node biopsy.

Comment

We have not included RCTs comparing specific techniques of sentinel node biopsy versus each other. An evidence based guideline from the American Society of Clinical Oncology recommends sentinel lymph node biopsy as an appropriate alternative to axillary dissection for women with early-stage breast cancer and clinically-negative axillary nodes. The guidelines also point out that the clinical significance of isolated cancer cells detected by detailed pathological examination of sentinel nodes is unknown.

Assessment of performance of sentinel node biopsy test:

We found one systematic review (search date not stated, 69 studies, 8059 people), which included studies assessing the diagnostic performance of sentinel node biopsy. It found that 96% of people had successful lymph node mapping (range 41% to 100%) and 42% of people had lymph node involvement (range 17% to 74%). The mean false-negative rate was 8.4% (range 0% to 29%), with a significantly higher false-negative rate in smaller studies (P less than 0.01) and studies with fewer successfully mapped nodes (P = 0.009).

Sentinel node biopsy versus axillary procedures (sampling or clearance):

In the first RCT, which compared sentinel node biopsy versus sentinel node biopsy plus axillary dissection, 36% of women in the sentinel node biopsy group also received axillary dissection due to positive sentinel node status. These women were included in the analysis for the sentinel node group. We found three preliminary publications of two RCTs comparing sentinel node biopsy versus axillary procedures. The first RCT (abstract only, 5611 women with node-negative breast cancer) compared sentinel node resection plus immediate axillary dissection versus sentinel node resection without axillary dissection. Preliminary technical results suggest similar sentinel node identification between treatment groups. The second RCT (Axillary Lymphatic Mapping Against Nodal Axillary Clearance [ALMANAC] study, preliminary results only, women with clinically node-negative invasive breast cancer) compared sentinel node biopsy versus conventional axillary treatment. The results of the randomisation phase have not been fully published, but preliminary results suggest that sentinel node biopsy significantly reduces lymphoedema (circumferential arm measurements) and improves sensory deficit versus standard axillary treatment (clearance or radiotherapy) at 18 months after treatment (P less than 0.0001 for both outcomes). The RCT found no significant difference between treatments in shoulder flexion and abduction, and internal and external rotation of the shoulder. Results of the validation phase of this RCT suggest that indiscriminate removal of axillary nodes may worsen the morbidity of sentinel node biopsy.

Clinical guide:

Axillary sentinel lymph node biopsy or axillary node sampling should be considered a preferential approach to managing the axilla in clinically axillary node-negative breast cancer.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

High-dose chemotherapy

Summary

MORTALITY Compared with conventional chemotherapy: High-dose chemotherapy plus autologous stem cell transplantation seems to be equally effective as conventional chemotherapy at prolonging 3- or 5-year overall survival rates in women with early, poor prognosis breast cancer, multiple positive axillary lymph nodes, and no distant metastasis ( moderate-quality evidence ). ADVERSE EFFECTS Compared with conventional chemotherapy: High-dose chemotherapy plus autologous stem cell transplantation is more likely to increase treatment related deaths compared with conventional chemotherapy ( high-quality evidence ).

Benefits

We found one systematic review (search date 2004, 13 RCTs, 5064 women with early, poor prognosis breast cancer, multiple positive axillary lymph nodes, and no distant metastasis), which compared high-dose chemotherapy plus bone marrow or peripheral blood stem cell autograft versus conventional chemotherapy (see comment below). It found no significant difference between regimens in overall survival at 3, 4, or 5 years (3 years: 5 RCTs, 2465 women, RR 1.01, 95% CI 0.97 to 1.05; 4 years: 3 RCTs, 743 women, RR 1.08, 95% CI 0.98 to 1.19; 5 years: 6 RCTs, 3015 women, RR 1.01, 95% CI 0.96 to 1.05). It found that high-dose chemotherapy significantly increased event-free survival versus conventional chemotherapy at 3 years but not at 5 years (3 years: 5 RCTs, 2465 women, RR 1.12, 95% CI 1.06 to 1.19; 5 years: 6 RCTs, 3017 women, RR 1.05, 95% CI 0.99 to 1.11 )..

Harms

The systematic review found that high-dose chemotherapy significantly increased treatment related mortality compared with conventional chemotherapy (treatment related deaths: 13 RCTs; 65/2535 [2.6%] with high dose v 4/2529 [0.2%] with conventional chemotherapy; RR 8.58, 95% CI 4.13 to 17.80 ).

Comment

Most of the RCTs included in the systematic review have only been published as abstracts or have reported only preliminary results, and reporting of follow-up is incomplete. Further results are awaited. Overall survival rates quoted in the review were predominantly based on results to date and showed no differences in overall survival. The systematic review concluded that there was insufficient evidence to support the routine use of high-dose chemotherapy with autograft for women with early poor prognosis breast cancer.

Substantive changes

High-dose chemotherapy (under question on treatments for primary operable breast cancer) One previously included systematic review updated;benefits and harms data enhanced, categorisation of 'high-dose chemotherapy plus autologous stem cell transplantation' unchanged (Likely to be ineffective or harmful).

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Postoperative radiotherapy (in women also receiving postoperative systemic treatment)

Summary

TREATMENT SUCCESS Post operative radiotherapy in women also receiving postoperative systemic treatment compared with no radiotherapy: Postoperative radiotherapy may be more effective at reducing recurrences or at improving disease-free survival at 10 years in women who also receive chemotherapy or hormone therapy following mastectomy ( very low-quality evidence ). MORTALITY Post operative radiotherapy in women also receiving postoperative systemic treatment compared with no radiotherapy: Postoperative radiotherapy may be more effective at improving overall survival at 10 years in women who also receive chemotherapy or hormone therapy following mastectomy (very low-quality evidence).

Benefits

We found no systematic review but found four RCTs. The first RCT (184 women, including some with stage III B disease, receiving premastectomy and postmastectomy chemoendocrine treatment) compared postmastectomy radiotherapy to the chest wall and regional lymphatics (45–50 Gy in 5 weeks) versus no radiotherapy. However, 43% of the 184 women were excluded and there were more exclusions in the radiotherapy group, and it is impossible to ascertain what percentage of women were stage III B. There were numerous chemotherapy complications, including one death. The RCT found no significant difference in local or distant failures. However, it found that overall crude survival was significantly higher with no radiotherapy compared with radiotherapy (28.7 months with no radiotherapy v 21.7 months with radiotherapy; P less than 0.05). Due to poor study quality, conclusions cannot be drawn from this RCT. The second RCT (332 women who were recurrence-free after modified radical mastectomy and 6 cycles of chemohormone treatment for operable locally advanced breast cancer; 38% stage T4 and 14% N2) compared postoperative radiotherapy versus no further treatment. It found no significant difference in time to relapse or median overall survival (time to relapse: 4.7 years with radiotherapy v 5.2 years with no further treatment, P = 0.68; median overall survival: 8.3 years with radiotherapy v 8.1 years with no further treatment, P = 0.94). Radiotherapy reduced loco-regional sites as first recurrence by 9%. The third RCT (1708 premenopausal women who had undergone mastectomy for stage II or III breast cancer) compared eight cycles of CMF (cyclophosphamide, methotrexate, and fluorouracil) chemotherapy plus radiotherapy to the chest wall and regional lymph nodes versus chemotherapy alone. It found that addition of postoperative radiotherapy to adjuvant chemotherapy after surgery significantly reduced loco-regional recurrence and improved overall and disease-free survival versus chemotherapy alone after 10 years (recurrence: RR 0.27, 95% CI 0.21 to 0.34; overall survival: 54% with radiotherapy plus chemotherapy v 45% with chemotherapy alone; disease-free survival: 48% with radiotherapy plus chemotherapy v 34% with chemotherapy alone; all comparisons reported as significant). The fourth RCT (1375 postmenopausal women, all with high-risk breast cancer, receiving adjuvant tamoxifen) found that postoperative radiotherapy significantly reduced loco-regional recurrence and improved disease-free survival versus no radiotherapy at a median follow-up of 123 months (recurrence: 52/689 [8%] with radiotherapy v 242/686 [35%] without radiotherapy, P less than 0.001; disease-free survival: 36% with radiotherapy v 24% without radiotherapy, P less than 0.001). It found that radiotherapy also improved overall survival versus no radiotherapy at 10 years (survival: 45% with radiotherapy v 36% without radiotherapy; P = 0.03). Subgroup analysis of 189 postmenopausal women with skin invasion found that radiotherapy plus tamoxifen reduced local recurrence and increased overall and disease-free survival after 5 and 10 years (recurrence: 8% with radiotherapy v 34% with tamoxifen alone, significance not provided; 5-year disease-free survival: 41% with radiotherapy v 37% with tamoxifen; 10-year disease-free survival: 23% with radiotherapy v 22% with tamoxifen; 5-year survival: 51% with radiotherapy v 61% with tamoxifen; 10-year survival: 31% with radiotherapy v 27% with tamoxifen, P values not reported). However, the studies used small and retrospective subgroup analyses, making conclusions uncertain.

Harms

The type of harms from radiotherapy for locally advanced breast cancer were similar to those from radiotherapy after mastectomy or breast-conserving surgery (see harms of radiotherapy after breast-conserving surgery. However, in stage III B disease with skin involvement (T4 b, c, d), the skin is usually given a higher dose of radiotherapy. In addition, a higher dose (60 Gy) is often given to more of the breast volume. Acute skin toxicity (including moist desquamation) and late skin toxicity (pigmentation and telangiectasia) are also more likely than in women without skin involvement.

Comment

The lack of good quality, large RCTs addressing directly stage III B breast cancer and the role of radiotherapy render it difficult to draw firm conclusions on its value. The published RCTs are small and have varying approaches to management. It is more difficult to detail the possible benefits of postoperative radiotherapy in women whose locally advanced breast cancers have been rendered operable by systemic treatment and who have had surgery, usually modified radical mastectomy. It is likely that such postoperative radiotherapy will reduce the risk of local (and regional, if nodal areas are irradiated) recurrence. It is not possible to conclude that it will affect survival.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Surgery

Summary

TREATMENT SUCCESS Surgery compared with radiotherapy: Surgery compared with radiotherapy as sole local treatments seem to have equal durations of disease control and remission in women with locally advanced disease (stage III B) rendered operable by prior chemotherapy ( moderate-quality evidence ). MORTALITY Surgery compared with radiotherapy: Surgery compared with radiotherapy as sole local treatments seem to be equally effective at prolonging overall survival at 3 to 4 years in women with locally advanced disease (stage III B) rendered operable by prior chemotherapy (moderate-quality evidence).

Benefits

Surgery alone versus radiotherapy alone:

We found no systematic reviews but found two RCTs, which compared surgery alone with radiotherapy alone as local treatment. In the first RCT (113 women with stage III breast cancer, 67% stage III B), women were given chemotherapy and 81% became operable; then 87 women were randomised to surgery or radiotherapy. After local treatment, a further 2 years of chemotherapy was given. Both groups had a similar duration of disease control (29.2 months with surgery v 24.4 months with radiotherapy; P = 0.5), similar overall median survival (39.3 months with surgery v 39.0 months with radiotherapy), and similar sites of first relapse. In the second RCT (132 women, 91% stage III B, 9% stage III A), all women received chemotherapy before randomisation to either surgery or radiotherapy. The total response rate was 75% in each group. There was no significant difference in the duration of remission (15 months with surgery v 22 months with radiotherapy; P = 0.58). Survival was similar at 4 years (49.1 months with surgery v 52 months with radiotherapy).

Harms

The type of harms from radiotherapy for locally advanced breast cancer were similar to those from radiotherapy after mastectomy or breast-conserving surgery (see harms of radiotherapy after breast-conserving surgery. However, in stage III B disease with skin involvement (T4 b, c, d), the skin is usually given a higher dose of radiotherapy. In addition, a higher dose (60 Gy) is often given to more of the breast volume. Acute skin toxicity (including moist desquamation) and late skin toxicity (pigmentation and telangiectasia) are also more likely than in women without skin involvement.

Comment

From the results of two RCTs, it can be concluded that in terms of overall response (which includes the response from local treatments such as surgery, radiotherapy, or both, and the effects of any initial systemic treatment), duration of that response, and overall survival, there is no advantage of either surgery alone or radiotherapy alone as sole local treatment over the other. It is more difficult to detail the possible benefits of postoperative radiotherapy in women whose locally advanced breast cancers have been rendered operable by systemic treatment and who have had surgery, usually modified radical mastectomy. It is likely that such postoperative radiotherapy will reduce the risk of local (and regional, if nodal areas are irradiated) recurrence. It is not possible to conclude that it will affect survival.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Radiotherapy

Summary

MORTALITY Surgery compared with radiotherapy: We don' t know whether surgery as sole local treatment is more effective at prolonging overall survival in women with locally advanced disease (stage III B) rendered operable by prior chemotherapy ( moderate-quality evidence ).

Benefits

Radiotherapy alone versus surgery alone:

See benefits of surgery alone versus radiotherapy alone.

Harms

The type of harms from radiotherapy for locally advanced breast cancer were similar to those from radiotherapy after mastectomy or breast-conserving surgery (see harms of radiotherapy after breast-conserving surgery. However, in stage III B disease with skin involvement (T4 b, c, d), the skin is usually given a higher dose of radiotherapy. In addition, a higher dose (60 Gy) is often given to more of the breast volume. Acute skin toxicity (including moist desquamation) and late skin toxicity (pigmentation and telangiectasia) are also more likely than in women without skin involvement.

Comment

The lack of good quality, large RCTs addressing directly stage III B breast cancer and the role of radiotherapy render it difficult to draw firm conclusions on its value. The published RCTs are small and have varying approaches to management. From the results of two RCTs, it can be concluded that in terms of overall response (which includes the response from local treatments such as surgery, radiotherapy, or both, and the effects of any initial systemic treatment), duration of that response, and overall survival, there is no advantage of either surgery alone or radiotherapy alone as sole local treatment over the other. It is more difficult to detail the possible benefits of postoperative radiotherapy in women whose locally advanced breast cancers have been rendered operable by systemic treatment and who have had surgery, usually modified radical mastectomy. It is likely that such postoperative radiotherapy will reduce the risk of local (and regional, if nodal areas are irradiated) recurrence. It is not possible to conclude that it will affect survival.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Low-dose radiotherapy

Summary

MORTALITY Low-dose radiotherapy compared with tamoxifen: We don't know whether low-dose radiotherapy (40 Gy in 15 fractions) is more effective at prolonging survival in women with locally advanced breast cancer ( low-quality evidence ). ADVERSE EFFECTS Radiotherapy may rarely be associated with late adverse effects, such as pneumonitis, pericarditis, arm oedema, brachial plexopathy, and radionecrotic rib fracture.

Benefits

Low-dose radiotherapy versus tamoxifen:

We found no systematic review but found one small RCT. The RCT (143 women with locally advanced breast cancer) compared low-dose radiotherapy (40 Gy in 15 fractions) versus tamoxifen 20 mg twice daily. Women were given the alternative treatment on relapse. The RCT found no significant difference in response rates (P = 0.34), duration of response (P = 0.76), or survival (P = 0.38).

Harms

The type of harms from radiotherapy for locally advanced breast cancer were similar to those from radiotherapy after mastectomy or breast-conserving surgery (see harms of radiotherapy after breast-conserving surgery. However, in stage III B disease with skin involvement (T4 b, c, d), the skin is usually given a higher dose of radiotherapy. In addition, a higher dose (60 Gy) is often given to more of the breast volume. Acute skin toxicity (including moist desquamation) and late skin toxicity (pigmentation and telangiectasia) are also more likely than in women without skin involvement.

Comment

The lack of good quality, large RCTs addressing directly stage III B breast cancer and the role of radiotherapy render it difficult to draw firm conclusions on its value. The published RCTs are small and have varying approaches to management. From the results of two RCTs, it can be concluded that in terms of overall response (which includes the response from local treatments such as surgery, radiotherapy, or both, and the effects of any initial systemic treatment), duration of that response, and overall survival, there is no advantage of either surgery alone or radiotherapy alone as sole local treatment over the other. It is more difficult to detail the possible benefits of postoperative radiotherapy in women whose locally advanced breast cancers have been rendered operable by systemic treatment and who have had surgery, usually modified radical mastectomy. It is likely that such postoperative radiotherapy will reduce the risk of local (and regional, if nodal areas are irradiated) recurrence. It is not possible to conclude that it will affect survival.

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Systemic treatment (hormone treatment, chemotherapy or both)

Summary

TREATMENT SUCCESS Systemic treatment plus radiotherapy compared with radiotherapy alone: We don't know whether adding chemotherapy or hormone treatment (ovarian irradiation for premenopausal women, tamoxifen for postmenopausal women) to radiotherapy is more effective at reducing loco-regional recurrences in women with locally advanced breast cancer ( very low-quality evidence ). MORTALITY Systemic treatment plus radiotherapy compared with radiotherapy alone: Adding hormone treatment (ovarian irradiation for premenopausal women, tamoxifen for postmenopausal women) may improve median survival but we don't whether adding chemotherapy improves overall survival in women with locally advanced breast cancer (very low-quality evidence).

Benefits

Systemic treatment plus radiotherapy versus radiotherapy alone:

We found no systematic review but found three RCTs, which compared radiotherapy versus radiotherapy plus systemic treatment (hormone treatment, chemotherapy, or both). The first RCT (410 women, most stage III B) compared four treatments in a factorial design: radiotherapy; radiotherapy plus chemotherapy (CMF [cyclophosphamide, methotrexate, and fluorouracil] for 12 cycles); radiotherapy plus hormone treatment (ovarian irradiation for premenopausal women, tamoxifen for postmenopausal women); and radiotherapy plus chemotherapy plus hormone treatment. Adding chemotherapy (P = 0.0002) or hormone treatment (P = 0.0007) to radiotherapy significantly delayed loco-regional recurrence, and adding both chemotherapy and hormone treatment had the greatest effect on delaying loco-regional recurrence (P = 0.0001). Adding chemotherapy or hormone treatment to radiotherapy reduced loco-regional recurrence at 6 years (AR about 60% with radiotherapy alone v about 50% with radiotherapy plus chemotherapy or hormonal treatment). The effect of adding chemotherapy or hormone treatment to radiotherapy on distant metastases was similar but less marked. Adding hormone treatment to radiotherapy significantly increased median survival, but adding chemotherapy did not significantly increase survival (median survival: 4.3 years with hormone treatment v 3.3 years without hormone treatment after 8 years, HR death 0.75, 95% CI 0.59 to 0.96; median survival: 3.8 years with chemotherapy v 3.6 years without chemotherapy, HR death 0.84, 95% CI 0.66 to 1.08). The second RCT (118 women with stage III B breast cancer) compared three treatments: radiotherapy; radiotherapy plus chemotherapy (CMF for 12 cycles) plus tamoxifen; and chemotherapy (CMF alternating with adriamycin [doxorubicin] and vincristine) followed by radiotherapy and further similar chemotherapy plus tamoxifen. The radiotherapy in the third treatment group delivered a lower dose to the skin and a lower total dose than delivered in the other two treatment groups. After a minimum follow-up of 14 years, the RCT found no significant difference in survival, disease-free survival, or type of first recurrence between groups (figures presented graphically; survival: P = 0.38; disease-free survival: P = 0.26; first recurrence: P = 0.4). The third RCT (52 women with T4 breast cancer) compared an anthracycline chemotherapy regimen before radiotherapy versus similar radiotherapy alone. Chemotherapy plus radiotherapy significantly increased initial loco-regional control rate (complete response defined as no palpable, visible, or radiological evidence of cancer in the breast or regional lymph node areas) compared with radiotherapy alone (complete response: 78.6% with chemotherapy plus radiotherapy v 45.8% with radiotherapy alone; P = 0.03). However, the proportion of women free of loco-regional spread at death or final follow-up was similar (57% with chemotherapy plus radiotherapy v 50% with radiotherapy alone). Overall survival and time to distant recurrence were not significantly different between groups.

Harms

In many RCTs, harms of treatment were not reported (see harms of adjuvant combination chemotherapy).

Comment

The lack of large RCTs and the frequent inclusion of less locally advanced disease (T3) with locally advanced breast cancer (defined here as stage III B) make it difficult to draw conclusions. There is, however, no evidence from the studies using CMF chemotherapy or various regimens incorporating anthracyclines that cytotoxic chemotherapy improves survival, disease-free survival, or long-term loco-regional control in stage III B breast cancer. Chemotherapy is often used in the management of locally advanced breast cancer (as defined here), despite a lack of evidence of measurable benefit. The lack of evidence may in part be due to small sample sizes in RCTs.

Substantive changes

No new evidence

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Multimodal treatment

Summary

RECURRENCE Multimodal treatment compared with hormone treatment: Multimodal treatment (primary chemotherapy, surgery, radiotherapy, and tamoxifen) seems to be more effective than initial treatment with tamoxifen plus salvage treatments upon tumour progression at improving remission rates at 6 months but not at reducing the development of metastases, or controlling disease at 52 months ( low-quality evidence ).

Benefits

Multimodal treatment versus hormone treatment:

We found no systematic review but found one RCT (two publications). The RCT (108 women) compared multimodal treatment (primary chemotherapy, surgery, radiotherapy, and tamoxifen) with initial hormone treatment plus subsequent salvage treatments upon tumour progression. The objective remission after 6 months was higher with multimodal treatment than with tamoxifen alone (31/54 [57%] with multimodal treatment v 19/53 [36%] with tamoxifen alone; OR 2.4, 95% CI 1.1 to 5.0). However, at a median follow-up of 52 months, there was no significant difference in survival, the development of metastases, the time to metastases, or uncontrolled local disease. Women with oestrogen receptor-positive tumours had a higher overall objective response rate (49% with oestrogen receptor-positive v 7% with oestrogen receptor-negative; P value not reported) and increased survival (numbers not reported).

Harms

In many RCTs, harms of treatment were not reported (see harms of adjuvant combination chemotherapy).

Comment

None

Substantive changes

BMJ Clin Evid. 2007 Dec 4;2007:0102.

Chemotherapy plus radiotherapy

Summary

MORTALITY Adjuvant chemotherapy compared with primary plus adjuvant chemotherapy: Adjuvant chemotherapy and primary plus adjuvant chemotherapy are equally effective at 25 months at improving disease-free survival, and overall survival in women also receiving radiotherapy ( moderate-quality evidence ).

Benefits

Adjuvant chemotherapy versus primary plus adjuvant chemotherapy:

We found no systematic review but found one RCT. The RCT (101 women with operable T4bN0–2 breast cancer) compared six cycles of adjuvant CEF (cyclophosphamide, epirubicin, 5-fluorouracil) after surgery (standard or modified radical mastectomy) versus three cycles of primary CEF followed by surgery plus three cycles of adjuvant CEF; both groups received chest wall and total nodal radiotherapy. At a median follow-up of 25 months, there was no significant difference in overall survival or disease-free survival (overall survival: 82% with adjuvant v 76% with primary plus adjuvant chemotherapy, P = 0.42; disease-free survival: 76% with adjuvant v 61% with primary plus adjuvant chemotherapy, P = 0.18).

Harms

In many RCTs, harms of treatment were not reported (see harms of adjuvant combination chemotherapy).

Comment

None

Substantive changes


Articles from BMJ Clinical Evidence are provided here courtesy of BMJ Publishing Group

RESOURCES