你现在的位置: 首页 > 癌症医典(乳腺癌) > 乳腺癌预防
中文版 / English
乳腺癌预防(PDQ®)

哪些是危险人群?

除了性别为女性,年龄增长是乳腺癌的最大风险因素。增加内源性雌激素暴露的生殖因素如初潮提前和绝经延迟会增加乳腺癌风险,绝经后雌孕激素联合使用也会增加乳腺癌风险。未生育和饮酒也与乳腺癌风险增加有关。

有浸润性乳腺癌、原位导管癌或原位小叶癌家族史或个人史,或乳腺活检显示良性增生性疾病的女性患乳腺癌的风险增加。

乳腺密度增加与乳腺癌风险增加有关。其通常具有遗传特性,但也常见于未生育、首次怀孕年龄晚的女性,以及绝经后使用激素和饮酒的女性。

电离辐射暴露,尤其是在青春期或青年期,以及有害基因突变的遗传可增加乳腺癌风险。

参考文献

  • Kotsopoulos J, Chen WY, Gates MA, et al.: Risk factors for ductal and lobular breast cancer: results from the nurses' health study. Breast Cancer Res 12 (6): R106, 2010.
  • Goldacre MJ, Abisgold JD, Yeates DG, et al.: Benign breast disease and subsequent breast cancer: English record linkage studies. J Public Health (Oxf) 32 (4): 565-71, 2010.
  • Kabat GC, Jones JG, Olson N, et al.: A multi-center prospective cohort study of benign breast disease and risk of subsequent breast cancer. Cancer Causes Control 21 (6): 821-8, 2010.
  • Worsham MJ, Raju U, Lu M, et al.: Risk factors for breast cancer from benign breast disease in a diverse population. Breast Cancer Res Treat 118 (1): 1-7, 2009.
  • Breast Cancer Prevention (PDQ®)

    Who Is at Risk?

    Besides female sex, advancing age is the biggest risk factor for breast cancer. Reproductive factors that increase exposure to endogenous estrogen, such as early menarche and late menopause, increase risk, as does the use of combination estrogen-progesterone hormones after menopause. Nulliparity and alcohol consumption also are associated with increased risk.

    Women with a family history or personal history of invasive breast cancer, ductal carcinoma in situ or lobular carcinoma in situ, or a history of breast biopsies that show benign proliferative disease have an increased risk of breast cancer.

    Increased breast density is associated with increased risk. It is often a heritable trait but is also seen more frequently in nulliparous women, women whose first pregnancy occurs late in life, and women who use postmenopausal hormones and alcohol.

    Exposure to ionizing radiation, especially during puberty or young adulthood, and the inheritance of detrimental genetic mutations increase breast cancer risk.

    ReferenceSection

  • Kotsopoulos J, Chen WY, Gates MA, et al.: Risk factors for ductal and lobular breast cancer: results from the nurses' health study. Breast Cancer Res 12 (6): R106, 2010.
  • Goldacre MJ, Abisgold JD, Yeates DG, et al.: Benign breast disease and subsequent breast cancer: English record linkage studies. J Public Health (Oxf) 32 (4): 565-71, 2010.
  • Kabat GC, Jones JG, Olson N, et al.: A multi-center prospective cohort study of benign breast disease and risk of subsequent breast cancer. Cancer Causes Control 21 (6): 821-8, 2010.
  • Worsham MJ, Raju U, Lu M, et al.: Risk factors for breast cancer from benign breast disease in a diverse population. Breast Cancer Res Treat 118 (1): 1-7, 2009.
  • 乳腺癌预防(PDQ®)

    概述

    注:独立的PDQ总结,包括乳腺癌筛查、乳腺癌治疗(成人)、男性乳腺癌治疗、妊娠期乳腺癌治疗,以及癌症筛查和预防研究,可以从相应章节获取。

    有充分证据表明可增加乳腺癌风险的因素

    性别和年龄

    基于确凿证据,性别为女性和年龄增长是乳腺癌发生的主要风险因素。

    影响程度:女性一生患乳腺癌的风险大约是男性的100倍。70岁女性患乳腺癌的短期风险大约是30岁女性的10倍。

  • 研究设计:多项流行病学试验。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 遗传风险

    基于确凿证据,有乳腺癌家族史的女性,特别是一级亲属,患乳腺癌的风险增加。

    影响程度:如果一个一级亲属确诊,风险会增加一倍;如果两个一级亲属确诊,风险会增加五倍。

  • 研究设计:人群研究、队列研究和病例对照研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 基于确凿证据,遗传基因突变的女性患乳腺癌风险增加。

    影响程度:不定,取决于基因突变、家族史和其他影响基因表达的风险因素。

  • 研究设计:队列或病例对照研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 乳腺密度

    有确凿证据表明致密乳腺可增加女性患乳腺癌风险。乳腺密度具有一定的遗传性,但生育行为、药物和饮酒在某种程度上也可以影响乳腺密度。

    影响程度:乳腺致密的女性罹患乳腺癌风险增加,且风险与密度成正相关。相比乳腺密度最低的女性,相对危险度(RR)从乳腺密度稍高女性的1.79倍上升到乳腺密度非常高女性的4.64倍。

  • 研究设计:队列,病例对照研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 有充分证据表明可增加乳腺癌风险的可改变因素

    联合激素治疗

    基于确凿证据,联合激素治疗(HT)(雌孕激素)与乳腺癌发病风险升高相关。

    影响程度:浸润性乳腺癌的发病率大约增加了26%;每超额发生一例乳腺癌所需联合激素治疗的人数为 237。

  • 研究设计:随机对照试验(RCT)。此外,队列研究和生态学研究表明,停止联合使用羟色胺与乳腺癌发病率降低有关。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 电离辐射

    基于确凿证据,暴露于电离辐射与乳腺癌发病风险升高相关,从暴露后10年开始增加并持续终生。乳腺癌发病风险取决于辐射剂量和暴露时的年龄,如果暴露发生在青春期乳房发育时,风险尤其高。

    影响程度:不定,但总体上大约增加了六倍。

  • 研究设计:队列或病例对照研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 肥胖

    基于确凿证据,肥胖与未使用激素治疗的绝经后女性患乳腺癌的风险增加有关。目前尚不清楚减肥是否能降低肥胖女性患乳腺癌的风险。

    影响程度:女性健康行动对85917例绝经后女性的观察性研究发现,体重与乳腺癌有关。将体重超过82.2公斤和低于58.7公斤的女性进行对比,RR为2.85(95%置信区间[CI],1.81–4.49)。

  • 研究设计:病例对照和队列研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 酒精

    基于确凿证据,饮酒与乳腺癌风险增加呈剂量依赖关系。目前尚不确定的是,大量饮酒者减少酒精摄入是否会降低风险。

    影响程度:与非饮酒者相比,每天饮用约4杯酒精饮料的女性的RR为1.32(95%置信区间为1.19-1.45)。每天每喝一杯,RR增加7%(95%CI,5.5%-8.7%)。

  • 研究设计:病例对照和队列研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 有充分证据表明可降低乳腺癌风险的因素

    早孕

    基于确凿证据,20岁以前足月妊娠的女性患乳腺癌风险降低。

    影响程度:与未生育女性或35岁后生育的女性相比,乳腺癌风险降低50%。

  • 研究设计:病例对照和队列研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 母乳喂养

    基于确凿证据,母乳喂养的女性患乳腺癌的风险降低。

    影响程度:每哺乳12个月,乳腺癌的相对风险降低4.3%,此外,每次生育可降低7%的乳腺癌风险。

  • 研究设计:病例对照和队列研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 锻炼

    基于确凿证据,每周高强度运动4小时以上与乳腺癌风险降低有关。

    影响程度:平均RR降低30%至40%。对于体重正常或较轻的绝经前女性,效果可能最显著。

  • 研究设计:前瞻性观察和病例对照研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 子宫切除术后女性使用雌激素:获益

    基于合理证据,既往做过子宫切除术,同时联合雌激素治疗的女性,乳腺癌发病率较低。然而,不同流行病学研究,结果不同。

    影响程度:RCT发现接受雌激素治疗的女性6.8年后乳腺癌发病率比安慰剂组低23%(每年0.27%,服用年限中位数为5.9年,而服用安慰剂的女性为每年0.35%),但在一项观察性研究中,接受雌激素治疗的女性发病率高出30%。两项研究中,两组人群不同的筛查行为可以解释两组之间的差异。

  • 研究设计:一项RCT,观察性研究。
  • 内部效度:一般。
  • 一致性:较差。
  • 外部效度:较差。
  • 子宫切除术后女性使用雌激素:危害

    基于确凿证据,子宫切除术后和绝经后服用雌激素的女性患卒中和心血管疾病的风险增加。

    影响程度:卒中发病率增加39%(RR,1.39;95%CI,1.1-1.77),心血管疾病发病率增加12%(RR,1.12;95%CI,1.01-1.24)。

  • 研究设计:随机对照试验,观察性研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:较差。
  • 有充分证据证明可降低乳腺癌风险的干预措施

    选择性雌激素受体调节剂(SERM):获益

    基于确凿证据,他莫昔芬和雷洛昔芬可降低绝经后女性乳腺癌的发病率,他莫昔芬可降低绝经前高危女性乳腺癌的风险。对他莫昔芬和雷洛昔芬的疗效观察显示,在停用有效治疗数年后,他莫昔芬的疗效持续时间比雷洛昔芬长。

    SERM治疗可减少骨折的发生,主要是雷洛昔芬治疗,而非他莫昔芬治疗。骨折减少主要为椎体骨折减少(减少34%)和非椎体骨折的小幅度减少(7%)。

    影响程度:他莫昔芬在5年的治疗中使乳腺癌高危女性的发病率降低30%-50%,但仅限于雌激素受体阳性(ER阳性)乳腺癌和乳腺导管原位癌(DCIS)。从开始治疗到停用他莫昔芬治疗11年后,ER阳性浸润性乳腺癌的减少率至少维持在此水平约16年。与第0-10年相比,在开始使用他莫昔芬(5年)后的第10-16年之间,效果依然维持。对乳腺癌死亡率没有影响。

  • 研究设计:随机对照试验。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 选择性雌激素受体调节剂:危害

    基于确凿证据,他莫昔芬的治疗会增加子宫内膜癌的风险,这在随访的前5年中很明显。 血栓性血管疾病(即肺栓塞,卒中和深静脉血栓形成)和白内障的患病风险也会增加。 停用他莫昔芬治疗后,许多风险都降低了。 基于确凿证据,雷洛昔芬也可增加静脉肺栓塞和深静脉血栓形成,但不会增加子宫内膜癌风险。

    影响程度:荟萃分析显示子宫内膜癌的相对危险度(RR)为2.4(95%CI,1.5-4.0),静脉血栓栓塞的RR为1.9(95%CI,1.4-2.6)。他莫昔芬和雷洛昔芬对子宫内膜癌的风险比(HR)分别为2.18(95%CI,1.39-3.42)和1.09(95%CI,0.74-1.62)。总的来说,静脉血栓栓塞的HR为1.73(95%CI,1.47–2.05)。50岁以上女性的危害明显高于年轻女性。

  • 研究设计:随机对照试验。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 芳香化酶抑制剂或失活剂:获益

    基于确凿证据,芳香化酶抑制剂或失活剂(AI)可以降低绝经后女性患乳腺癌的风险。

    影响程度:随访35个月(中位随访期)后,35岁及以上的女性至少有一种危险因素(年龄> 60岁,Gail模型5年危险> 1.66%,或行乳腺切除术的DCIS)并且每天服用25 mg的依西美坦可降低浸润性乳腺癌的发生率(HR,0.35; 95%CI,0.18–0.70)。在超过35个月随访的2280名受试者中,有21例癌症患者绝对风险降低。需治疗人数约为100人。

  • 研究设计:一项随机对照试验。
  • 内部效度:良好。
  • 一致性:一项在无乳腺癌病史女性中开展的研究结果与在有乳腺癌病史女性中开展的RCT研究结果一致。
  • 外部效度:对于符合入选标准的女性良好。
  • 芳香化酶抑制剂或失活剂:危害

    根据随访超过35个月的4,560名女性的单项RCT研究的一般证据表明,与安慰剂相比,依西美坦使用与潮热和疲劳有关,但与骨折,骨质疏松或心血管疾病无关。

    影响程度:潮热绝对风险增加8%,疲劳绝对风险增加2%。

  • 研究设计:一项RCT。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:对于符合入选标准的女性良好。
  • 预防性乳房切除术:获益

    基于确凿证据,双侧预防性乳房切除术可以降低有强家族史的女性罹患乳腺癌的风险,并且大多数女性都可以缓解对乳腺癌风险的焦虑。 没有研究探讨同侧乳腺癌手术后进行对侧预防性乳房切除术的女性的乳腺癌结局。

    影响程度:高危女性双侧预防性乳房切除术后患乳腺癌的风险降低了90%,但已发表的研究设计可能会造成结局的高估。

  • 研究设计:从病例对照和队列研究中获得的证据。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 预防性卵巢切除术或卵巢去势治疗:获益

    基于确凿证据,患有BRCA基因突变的绝经前妇女接受预防性卵巢切除术的乳腺癌发病率较低。 同样,卵巢切除术或卵巢去势与正常绝经前女性和因胸部照射导致乳腺癌风险增加的女性乳腺癌发生率降低相关。

    影响程度:乳腺癌的发病率可降低50%,但是已发布的研究设计可能会造成高估。

  • 研究设计:观察性、病例对照和队列研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 预防性卵巢切除术或卵巢去势治疗:危害

    基于确凿证据,卵巢去势可能导致更年期症状的突然出现,如潮热、失眠、焦虑和抑郁。长期影响包括性欲下降、阴道干燥、骨密度降低。

    影响程度:几乎所有的女性都会经历一些睡眠障碍、情绪变化、潮热和骨质疏松,但这些症状的严重程度差别很大。

  • 研究设计:观察性、病例对照和队列研究。
  • 内部效度:良好。
  • 一致性:良好。
  • 外部效度:良好。
  • 参考文献

  • Boyd NF, Martin LJ, Rommens JM, et al.: Mammographic density: a heritable risk factor for breast cancer. Methods Mol Biol 472: 343-60, 2009.
  • McCormack VA, dos Santos Silva I: Breast density and parenchymal patterns as markers of breast cancer risk: a meta-analysis. Cancer Epidemiol Biomarkers Prev 15 (6): 1159-69, 2006.
  • Col: Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet 360 (9328): 187-95, 2002.
  • Cuzick J, Sestak I, Bonanni B, et al.: Selective oestrogen receptor modulators in prevention of breast cancer: an updated meta-analysis of individual participant data. Lancet 381 (9880): 1827-34, 2013.
  • Cuzick J, Sestak I, Cawthorn S, et al.: Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol 16 (1): 67-75, 2015.
  • Goss PE, Ingle JN, Alés-Martínez JE, et al.: Exemestane for breast-cancer prevention in postmenopausal women. N Engl J Med 364 (25): 2381-91, 2011.
  • Maunsell E, Goss PE, Chlebowski RT, et al.: Quality of life in MAP.3 (Mammary Prevention 3): a randomized, placebo-controlled trial evaluating exemestane for prevention of breast cancer. J Clin Oncol 32 (14): 1427-36, 2014.
  • Breast Cancer Prevention (PDQ®)

    Overview

    Note: Separate PDQ summaries on Breast Cancer Screening; Breast Cancer Treatment (Adult); Male Breast Cancer Treatment; Breast Cancer Treatment During Pregnancy; and Levels of Evidence for Cancer Screening and Prevention Studies are also available.

    Factors With Adequate Evidence of Increased Risk of Breast Cancer

    Sex and age

    Based on solid evidence, female sex and increasing age are the major risk factors for the development of breast cancer.

    Magnitude of Effect: Women have a lifetime risk of developing breast cancer that is approximately 100 times the risk for men. The short-term risk of breast cancer in a 70-year-old woman is about ten times that of a 30-year-old woman.

  • Study Design: Many epidemiologic trials.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Inherited risk

    Based on solid evidence, women who have a family history of breast cancer, especially in a first-degree relative, have an increased risk of breast cancer.

    Magnitude of Effect: Risk is doubled if a single first-degree relative is affected; risk is increased fivefold if two first-degree relatives are diagnosed.

  • Study Design: Population studies, cohort studies, and case-control studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Based on solid evidence, women who inherit gene mutations associated with breast cancer have an increased risk.

    Magnitude of Effect: Variable, depending on gene mutation, family history, and other risk factors affecting gene expression.

  • Study Design: Cohort or case-control studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Breast density

    Based on solid evidence, women with dense breasts have an increased risk of breast cancer. This is most often an inherent characteristic, to some extent modifiable by reproductive behavior, medications, and alcohol.

    Magnitude of Effect: Women with dense breasts have increased risk, proportionate to the degree of density. This increased relative risk (RR) ranges from 1.79 for women with slightly increased density to 4.64 for women with very dense breasts, compared with women who have the lowest breast density.

  • Study Design: Cohort, case-control studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Modifiable Factors With Adequate Evidence of Increased Risk of Breast Cancer

    Combination hormone therapy

    Based on solid evidence, combination hormone therapy (HT) (estrogen-progestin) is associated with an increased risk of developing breast cancer.

    Magnitude of Effect: Approximately a 26% increase in incidence of invasive breast cancer; the number needed to produce one excess breast cancer is 237.

  • Study Design: Randomized controlled trials (RCTs). Furthermore, cohort and ecological studies show that cessation of combination HT is associated with a decrease in rates of breast cancer.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Ionizing radiation

    Based on solid evidence, exposure of the breast to ionizing radiation is associated with an increased risk of developing breast cancer, starting 10 years after exposure and persisting lifelong. Risk depends on radiation dose and age at exposure, and is especially high if exposure occurs during puberty, when the breast develops.

    Magnitude of Effect: Variable but approximately a sixfold increase overall.

  • Study Design: Cohort or case-control studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Obesity

    Based on solid evidence, obesity is associated with an increased breast cancer risk in postmenopausal women who have not used HT. It is uncertain whether weight reduction decreases the risk of breast cancer in obese women.

    Magnitude of Effect: The Women's Health Initiative observational study of 85,917 postmenopausal women found body weight to be associated with breast cancer. Comparing women weighing more than 82.2 kg with those weighing less than 58.7 kg, the RR was 2.85 (95% confidence interval [CI], 1.81–4.49).

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Alcohol

    Based on solid evidence, alcohol consumption is associated with increased breast cancer risk in a dose-dependent fashion. It is uncertain whether decreasing alcohol intake by heavy drinkers reduces the risk.

    Magnitude of Effect: The RR for women consuming approximately four alcoholic drinks per day compared with nondrinkers is 1.32 (95% CI, 1.19–1.45). The RR increases by 7% (95% CI, 5.5%–8.7%) for each drink per day.

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Factors With Adequate Evidence of Decreased Risk of Breast Cancer

    Early pregnancy

    Based on solid evidence, women who have a full-term pregnancy before age 20 years have decreased breast cancer risk.

    Magnitude of Effect: 50% decrease in breast cancer, compared with nulliparous women or women who give birth after age 35 years.

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Breast-feeding

    Based on solid evidence, women who breast-feed have a decreased risk of breast cancer.

    Magnitude of Effect: The RR of breast cancer is decreased 4.3% for every 12 months of breast-feeding, in addition to 7% for each birth.

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Exercise

    Based on solid evidence, exercising strenuously for more than 4 hours per week is associated with reduced breast cancer risk.

    Magnitude of Effect: Average RR reduction is 30% to 40%. The effect may be greatest for premenopausal women of normal or low body weight.

  • Study Design: Prospective observational and case-control studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Estrogen use by women with prior hysterectomy: benefits

    Based on fair evidence, women who have undergone a prior hysterectomy and who are treated with conjugated equine estrogen have a lower incidence of breast cancer. However, epidemiological studies yield conflicting results.

    Magnitude of Effect: After 6.8 years, incidence was 23% lower in women treated with estrogen in an RCT (0.27% per year, with a median of 5.9 years of use, compared with 0.35% per year among those taking a placebo), but was 30% higher in women treated with estrogen in an observational study. The difference in these results may be explained by different screening behavior by the women in both studies.

  • Study Design: One RCT, observational studies.
  • Internal Validity: Fair.
  • Consistency: Poor.
  • External Validity: Poor.
  • Estrogen use by women with prior hysterectomy: harms

    Based on solid evidence, women who have undergone hysterectomy and who are taking postmenopausal estrogen have an increased risk of stroke and total cardiovascular disease.

    Magnitude of Effect: There is a 39% increase in the incidence of stroke (RR, 1.39; 95% CI, 1.1–1.77) and a 12% increase in cardiovascular disease (RR, 1.12; 95% CI, 1.01–1.24).

  • Study Design: RCTs, observational studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Poor.
  • Interventions With Adequate Evidence of Decreased Risk of Breast Cancer

    Selective estrogen receptor modulators (SERMs): benefits

    Based on solid evidence, tamoxifen and raloxifene reduce the incidence of breast cancer in postmenopausal women, and tamoxifen reduces the risk of breast cancer in high-risk premenopausal women. The effects observed for tamoxifen and raloxifene show persistence several years after active treatment is discontinued, with longer duration of effect noted for tamoxifen than for raloxifene.

    All fractures were reduced by SERMs, primarily noted with raloxifene but not with tamoxifen. Reductions in vertebral fractures (34% reduction) and small reductions in nonvertebral fractures (7%) were noted.

    Magnitude of Effect: Tamoxifen reduced breast cancer incidence in high-risk women from about 30% to about 50% over 5 years of treatment but only for estrogen receptor–positive (ER-positive) cancer and ductal carcinoma in situ (DCIS). The reduction in ER-positive invasive breast cancer was maintained at about this level for at least 16 years after starting treatment, 11 years after cessation of tamoxifen. There was no loss of effect between years 10 and 16 after starting tamoxifen (for 5 years) compared with years 0 to 10. There was no effect on breast cancer mortality.

  • Study Design: RCTs.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Selective estrogen receptor modulators: harms

    Based on solid evidence, tamoxifen treatment increases the risk of endometrial cancer, which was apparent in the first 5 years of follow-up but not beyond; thrombotic vascular events (i.e., pulmonary embolism, stroke, and deep venous thrombosis); and cataracts. Many of these risks are reduced after active treatment with tamoxifen is discontinued. Based on solid evidence, raloxifene also increases venous pulmonary embolism and deep venous thrombosis but not endometrial cancer.

    Magnitude of Effect: Meta-analysis showed RR of 2.4 (95% CI, 1.5–4.0) for endometrial cancer and 1.9 (95% CI, 1.4–2.6) for venous thromboembolic events. Meta-analysis showed the hazard ratio (HR) for endometrial cancer was 2.18 (95% CI, 1.39–3.42) for tamoxifen and 1.09 (95% CI, 0.74–1.62) for raloxifene. Overall, HR for venous thromboembolic events was 1.73 (95% CI, 1.47–2.05). Harms were significantly higher in women over 50 years than in younger women.

  • Study Design: RCTs.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Aromatase inhibitors or inactivators: benefits

    Based on solid evidence, aromatase inhibitors or inactivators (AIs) reduce the incidence of new breast cancers in postmenopausal women who have an increased risk.

    Magnitude of Effect: After a median follow-up of 35 months, women aged 35 years and older who had at least one risk factor (age >60 years, a Gail 5-year risk >1.66%, or DCIS with mastectomy) and who took 25 mg of exemestane daily had a decreased risk of invasive breast cancer (HR, 0.35; 95% CI, 0.18–0.70). The absolute risk reduction was 21 cancers avoided out of 2,280 participants over 35 months. The number needed to treat was about 100.

  • Study Design: One RCT.
  • Internal Validity: Good.
  • Consistency: One study in women with no history of breast cancer but consistent with RCTs in women with history of breast cancer.
  • External Validity: Good for women who meet inclusion criteria.
  • Aromatase inhibitors or inactivators: harms

    Based on fair evidence from a single RCT of 4,560 women over 35 months, exemestane is associated with hot flashes and fatigue but not with fractures, osteoporosis, or cardiovascular events, compared with placebo.

    Magnitude of Effect: The absolute increase in hot flashes was 8% and the absolute increase in fatigue was 2%.

  • Study Design: One RCT.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good for women who meet inclusion criteria.
  • Prophylactic mastectomy: benefits

    Based on solid evidence, bilateral prophylactic mastectomy reduces the risk of breast cancer in women with a strong family history, and most women experience relief from anxiety about breast cancer risk. There are no studies examining breast cancer outcomes in women who undergo contralateral prophylactic mastectomy after surgery for ipsilateral breast cancer.

    Magnitude of Effect: Breast cancer risk after bilateral prophylactic mastectomy in women at high risk is reduced as much as 90%, but published study designs may have produced an overestimate.

  • Study Design: Evidence obtained from case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Prophylactic oophorectomy or ovarian ablation: benefits

    Based on solid evidence, premenopausal women with BRCA gene mutations who undergo prophylactic oophorectomy have lower breast cancer incidence. Similarly, oophorectomy or ovarian ablation is associated with decreased breast cancer incidence in normal premenopausal women and in women with increased breast cancer risk resulting from thoracic irradiation.

    Magnitude of Effect: Breast cancer incidence is decreased by 50%, but published study designs may have produced an overestimate.

  • Study Design: Observational, case-control, and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • Prophylactic oophorectomy or ovarian ablation: harms

    Based on solid evidence, castration may cause the abrupt onset of menopausal symptoms such as hot flashes, insomnia, anxiety, and depression. Long-term effects include decreased libido, vaginal dryness, and decreased bone mineral density.

    Magnitude of Effect: Nearly all women experience some sleep disturbances, mood changes, hot flashes, and bone demineralization, but the severity of these symptoms varies greatly.

  • Study Design: Observational, case-control, and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.
  • ReferenceSection

  • Boyd NF, Martin LJ, Rommens JM, et al.: Mammographic density: a heritable risk factor for breast cancer. Methods Mol Biol 472: 343-60, 2009.
  • McCormack VA, dos Santos Silva I: Breast density and parenchymal patterns as markers of breast cancer risk: a meta-analysis. Cancer Epidemiol Biomarkers Prev 15 (6): 1159-69, 2006.
  • Col: Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet 360 (9328): 187-95, 2002.
  • Cuzick J, Sestak I, Bonanni B, et al.: Selective oestrogen receptor modulators in prevention of breast cancer: an updated meta-analysis of individual participant data. Lancet 381 (9880): 1827-34, 2013.
  • Cuzick J, Sestak I, Cawthorn S, et al.: Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol 16 (1): 67-75, 2015.
  • Goss PE, Ingle JN, Alés-Martínez JE, et al.: Exemestane for breast-cancer prevention in postmenopausal women. N Engl J Med 364 (25): 2381-91, 2011.
  • Maunsell E, Goss PE, Chlebowski RT, et al.: Quality of life in MAP.3 (Mammary Prevention 3): a randomized, placebo-controlled trial evaluating exemestane for prevention of breast cancer. J Clin Oncol 32 (14): 1427-36, 2014.
  • 乳腺癌预防(PDQ®)

    证据描述

    发病率和死亡率

    2019年美国女性最常诊断的非皮肤恶性肿瘤为乳腺癌,预计有268600例。据我国最新发布的癌症统计数据,我国女性最常诊断的恶性肿瘤为乳腺癌,每年发病约为30.4万例。

    同样在2019年,乳腺癌将导致41,760名妇女死亡,仅次于肺癌,这是导致女性癌症死亡的原因。 乳腺癌也发生在男性中,估计2019年将诊断出2670例新病例。2015年,乳腺癌导致约7万名中国女性死亡。死亡位居我国恶性肿瘤死亡的第5位。

    尽管先前女性乳腺癌发病率呈逐渐上升的趋势,但SEER数据库显示,从2007年到2016年,乳腺癌死亡率每年下降1.8%。中国女性乳腺癌发病率和死亡率均呈上升趋势。2003~2007年女性乳腺癌发病率为23.27/10万,死亡率为4.93/10万;2015年发病率为31.54/10万,死亡率6.67/10万。

    乳腺癌的主要危险因素是年龄的增长。30岁的女性在未来10年中被诊断为乳腺癌的几率是1/250,而70岁的女性为1/27。

    乳腺癌的发病率和死亡率也因地理、文化、人种、种族和社会经济状况的不同而不同。与其他人种相比,白人女性乳腺癌的发病率较高,这可能部分归因于筛查。然而,从2005年到2014年,黑人女性的乳腺癌发病率略有增加,为每年0.3%,导致黑人与白人的发病率趋同。

    乳腺X线筛查可以早期诊断从而早期治疗乳腺癌,进而降低乳腺癌的死亡率。 但是,筛查还发现了一生中不会出现症状的病例,因此筛查会增加乳腺癌的发病率。 (有关更多信息,请参阅PDQ总结中关于乳腺癌筛查的“过度诊断”部分。)

    乳腺癌的病因和发病机制

    乳腺癌的发展伴随着一系列基因突变的发生。

    最初,突变不会改变组织学形态,但是随着突变的累积,会导致增生、非典型性增生、原位癌、最终导致浸润性癌。

    随着年龄的增长,发生突变的体细胞数量会越多,这些突变产生的细胞群就越有可能演变成恶性肿瘤。雌激素和孕激素可能通过生长因子如转化生长因子(TGF)-α引起乳腺细胞的生长和增殖。

    这些激素无论是内源性还是外源性,都可能促进乳腺癌细胞的发育和增殖。

    乳腺癌发病率在全球范围内的不同可以通过遗传学、生育因素、饮食、运动和筛查的差异来解释。 这些因素中的有些因素是可改变的,有观察研究结果证明,日本人移民到美国后在两代人内罹患乳腺癌的风险从较低的日本人风险水平增加到较高的美国人风险水平。

    内源性雌激素

    乳腺癌的许多危险因素表明,长期暴露于内源性雌激素在该疾病的发展中起作用。 与初潮年龄在14岁或14岁以上的女性相比,初潮年龄在11岁或以下的女性患乳腺癌的几率高出约20%。

    绝经较晚的女性也有更高的患病风险。 患乳腺癌的妇女往往具有较高的内源性雌激素和雄激素水平。

    相反,过早绝经的女性患乳腺癌的风险较低。 在接受卵巢去势术后,根据年龄,体重和分娩次数的不同,乳腺癌风险可能降低高达75%,而年轻,偏瘦,未生育女性的风险减少幅度最大。

    单侧卵巢切除也能降低乳腺癌风险,但程度比双侧切除小。

    其他激素的变化对乳腺癌风险也有影响。(有关更多信息,请参阅本总结“有充分证据表明可降低乳腺癌风险的因素”一节中的早孕和母乳喂养部分。)

    内源性雌激素水平,胰岛素水平和肥胖之间的相互作用(均会影响乳腺癌风险)了解较少,但建议采取干预措施降低该风险。 生育危险因素可能与易感基因型相互作用。 例如,在“护士健康研究”中所得到的研究结果,

    仅在没有母亲或姐妹患有乳腺癌的家族史的女性中观察到首次分娩年龄,初潮和绝经与乳腺癌发展之间的关联。

    遗传风险

    有家族史的女性(尤其一级亲属受累时)患乳腺癌的风险增加。

    从数据库,队列研究和病例对照研究得出的以下风险评估模型对这种风险进行了量化:

  • 乳腺癌风险评估工具[Gail模型]。
  • 乳腺癌监测联合会[BCSC]风险计算器。
  • 乳腺癌转诊筛查工具[B-RST]。
  • Hall详细乳腺风险计算器。
  • IBIS乳腺癌风险计算工具。
  • 大约5%的乳腺癌与特定的异常等位基因有关。 (相关更多信息,请参阅PDQ关于乳腺癌和妇科癌症遗传学的总结。)BRCA基因的突变为常染色体显性遗传,并且在致癌方面具有高度外显率,常见在较年轻的女性。

    在具有乳腺癌遗传易感性的人群中,BRCA1或BRCA2基因突变和具有癌症家族史可能会增加患癌风险。

    BRCA1基因突变携带者罹患乳腺癌的终生风险为55%-65%,BRCA2基因突变携带者罹患乳腺癌的终生风险为45%-47%。

    相比之下,在普通人群中,罹患乳腺癌的终生风险为12.4%。

    有些女性遗传了对致突变剂或生长因子的易感性,这也会增加患乳腺癌的风险。

    (有关更多信息,请参阅此总结的“有充分证据表明可增加乳腺癌风险的因素”一节中的“电离辐射暴露”部分。)

    乳腺密度增加

    乳腺X线筛查广泛应用后,揭示乳腺密度存在差异性,且致密乳腺的女性乳腺癌发病率更高。同时乳腺密度也会影响乳腺X线筛查乳腺癌的结果。在由1112对配对的病例对照组成的筛查人群,三项巢式病例对照研究描述了相应风险增加的程度。与乳腺密度不到10%的女性相比,乳腺密度在75%或以上的女性患乳腺癌的风险增加(优势比[OR],4.7;95%置信区间[CI],3.0-7.4),无论癌症是筛查当时发现的(OR,3.5;95%CI,2.0-6.2),还是筛查阴性12个月内检出的(OR,17.8;95%CI,4.8-65.9),都能检测到乳腺癌的风险增加。乳腺癌风险的增加,无论是筛查还是其他方法检测到的乳腺癌风险,都有一定的增加,而且至少持续到研究开始后8年,年轻女性的风险高于年龄大的女性。对于中位年龄在56岁以下的女性,乳腺X线检查显示为乳腺密度为50%或以上的女性中,现场检出的乳腺癌风险增加26%,筛查阴性12个月内检出的乳腺癌增加50%。

    乳腺致密的女性患乳腺癌的风险增加,患乳腺癌的风险与乳房的致密程度成正比。与乳腺密度最低比较,乳腺密度从较高到最高,患乳腺癌的相对风险(RR)从1.79上升至4.64。

    乳腺致密的女性乳腺癌死亡率没有增加。

    有充分证据表明可增加乳腺癌风险的因素

    激素治疗

    1997年对纳入超过150000名女性的51项流行病学研究再分析发现,绝经后激素治疗(HT)与乳腺癌风险增加有关。

    2002年进行的心脏和雌孕激素替代治疗研究结果一致。

    该研究引入2763名平均年龄67岁的绝经后冠心病妇女,随机分为激素替代组和安慰剂组。平均随访6.8年后发现乳腺癌的相对危险度(RR)为1.27(95%CI=0.84-1.94)。尽管无显著统计学意义,但RR估值与2002年发表的美国妇女健康倡议(WHI)报告相一致。

    WHI研究了激素和膳食干预对心脏病和乳腺癌患病风险的影响。

    50-79岁具有完整子宫的女性被随机分配为接受结合雌激素和持续孕激素联合治疗组(n=8506)或安慰剂(n=8102)治疗组。因为HT联合治疗并未降低患冠心病风险,反而增加了卒中和乳腺癌风险,该研究提前终止。所有亚组结果都显示浸润性乳腺癌风险增加(风险比HR=1.24;95%CI=1.02–1.50),但原位乳腺癌风险未增加。与HT联合治疗相关的肿瘤,具有相似的分级、组织学、雌激素受体(ER)、孕激素受体(PR)和HER2/neu的表达,HT联合治疗组淋巴结转移的发生率较高且转移范围较大。

    平均11年的长期随访显示,HT治疗组的乳腺癌特异死亡率较高(分别为25例和12例,每年0.03%和0.01%;HR=1.95;95%CI=1.0-4.04;P=0.049)。HT联合治疗也与乳腺X线检查异常结果的高发生率有关。

    WHI观察性研究与WHI随机对照试验(RCT)平行开展,其招募研究对象为50-79岁的绝经后女性。WHI观察性研究分析评估了已诊断乳腺癌患者接受联合HR治疗的预后,以及从绝经到开始HT治疗之间的时间对乳腺癌风险的影响。经过平均11.3年的随访,雌激素联合孕激素治疗组的女性乳腺癌的年发病率为0.60%,而对照组的发病率为0.42%(HR=1.55;95%CI=1.41-1.70)。HT治疗组和对照组诊断乳腺癌后的生存率相似。HT治疗组的乳腺癌死亡率高于对照,但该差异不具有统计学意义(HR=1.3;94%CI=0.90–1.93)。绝经后立即接受HT治疗的女性患乳腺癌风险最高,随着绝经与开始接受HT联合治疗之间的间期延长,风险降低但持续存在。在乳腺癌确诊后,HT联合治疗组的全因死亡率显著高于对照(HR=1.87;95%CI=1.37-2.54)。总体而言,这些结果与RCT结果一致。

    子宫切除术后的女性无患子宫内膜癌的风险。WHI招募了10739名子宫切除术后、年龄在50-79岁的女性作为研究对象, 随机分组后分别接受马结合雌激素(CEE)或安慰剂治疗。这项试验也因卒中风险增加和风险获益指数无改善而提前终止。

    平均随访6.8年后,CEE组乳腺癌发病率低于安慰剂组(发病率分别为0.26%和0.33%每年;HR=0.77;95%CI=0.59-1.01)。CEE组的总体风险获益指数稍差。

    78%试验参与者接受了长期随访,中位随访时间为11.8年,初步结果保持不变,CEE组乳腺癌发病率同样较低(HR=0.77;95%CI=0.62-0.95),乳腺癌死亡率下降(死亡例数分别为6和16;HR=0.37;95%CI=0.13-0.91)。CEE组的全因死亡率也较低(分别为0.046%和0.076%每年;HR=0.62;95%CI=0.39-0.97)。停止CEE干预后,卒中风险降低。在整个随访期间,冠心病、深静脉血栓形成、卒中、髋部骨折或结直肠癌的发病率均无差异。

    绝经后的前五年内开始接受CEE或安慰剂治疗的女性乳腺癌发病率相近(HR=1.06;95%CI=0.74–1.51)。

    丹麦对1006名绝经早期妇女进行了HT试验,旨在评估其心血管事件结局。407例具有完整子宫的女性接受了联合HT(三相雌二醇和炔诺酮)治疗,95名子宫切除术后的女性接受了雌二醇治疗。对照组(包含407名子宫完整和97名子宫切除的女性)未接受处理。研究10年时,发现可能存在较大偏倚。被分配到HT组的女性中只有一半仍在服用规定的药物,而22%的对照组女性已经开始服用HT。接受HT治疗的女性和对照组乳腺癌的发病率并无差异。

    观察性研究对随机对照试验(RCT)中获得的信息进行了补充。

    英国百万女性研究

    在1996年至2001年间,英国共招募了1084110名50至64岁的女性,并获得了有关HT使用的资料和其他个人资料。对这些女性进行了乳腺癌发病率和死亡率的随访。一半的女性使用过HT。随访2.6年,发生浸润性乳腺癌患者为9364名;随访4.1年,637名死于乳腺癌。招募阶段正使用HT的患者比从未使用HT的患者有较高的风险患乳腺癌(校正后的RR=1.66;95%CI=1.58-1.75;P<0.0001)并死于该疾病(校正后的RR=1.22;95%CI=1.00-1.48;P=0.05)。然而,过去使用过HT的患者,其患乳腺癌的风险并没有增加(分别为1.01[95%CI=0.94–1.09]和1.05[95%CI=0.82–1.34])。仅目前使用雌激素(RR=1.30;95%CI=1.21-1.40;P<0.0001)、联合HT(RR=2.00;95%CI=1.88-2.12;P<0.0001)和替勃龙(RR=1.45;95%CI=1.25-1.68;P<0.0001)的患者发病率显著增加。联合HT治疗组和乳腺癌发生的关联强度显著大于其他类型的HT组(P<0.0001)。

    普吉特海峡癌症监测系统开展了一项基于人群的研究,纳入了965名乳腺癌患者和1007名对照。结果显示,联合HT使用者患浸润性乳腺癌的风险是相应对照的1.7倍,而仅使用雌激素者相关风险未见增高。

    在所有的试验中,联合使用HT和乳腺癌风险增加之间的关系是一致的。而单纯雌激素HT治疗与乳腺癌发病率之间的关系尚不明确,部分研究显示其导致乳腺癌风险增加,但也有研究显示其为保护作用。可能的原因包括:雌激素治疗时间与绝经开始年龄有关,观察性研究中激素用药者和非用药者的常规筛查率不均衡。

    WHI结果发表后,HT治疗在美国等地的使用量急剧下降。对联合HT组参与者进行后续随访,发现在同样的乳腺钼靶筛查率下,治疗后2年内原本升高的乳腺癌风险快速降低。

    美国乳腺癌发病率数据显示,从2002年到2003年,50岁及以上女性的乳腺癌,尤其是ER阳性乳腺癌发病率急剧下降。

    同样,在HT治疗方案使用率较高的多个国家,乳腺癌发病率在相近时间范围内下降,同时处方模式和/或报告的使用率也有所下降。

    一项在接受定期乳腺X线筛查的女性中进行的研究表明,2002年至2003年乳腺癌发病率急剧下降的主要原因是HT方案的使用减少,而不是乳腺X线钼靶检查率下降。

    在2002年至2003年乳腺癌发病率下降之后,美国的乳腺癌发病率趋于稳定。

    电离辐射

    电离辐射与其后罹患乳腺癌之间存在明确的相关性。

    原子弹暴露,结核病导致的频繁X线检查,痤疮、癣、胸腺增大、产后乳腺炎和淋巴瘤导致的放射性治疗等因素导致乳腺癌风险增高。年轻人,特别是青春期阶段的风险更高。据估计,医用射线相关的乳腺癌占所有乳腺癌的比例不足1%。

    但有一些理论提出,对于特定人群,例如AT杂合子人群,更易受到辐射增加乳腺癌风险的影响。

    在携带BRCA1或BRCA2突变的女性中开展的一项大型队列研究显示,胸部X线检查会增加乳腺癌风险(RR=1.54;95%CI=1.1-2.1),特别是在20岁之前曾接受X线检查的女性。

    16岁前接受霍奇金淋巴瘤治疗的女性,在40岁之前罹患乳腺癌的风险高达35%。

    较高放射剂量(对乳腺癌患者剂量中位值为40Gy),且在10-16岁期间接受放疗者的乳腺癌风险较高。

    与继发性白血病的风险不同,随着随访时间的延长,治疗相关乳腺癌的风险并未降低,增高的风险在放疗后可持续超过25年。

    这些研究中,多数患者(85%-100%)在放射野内或放射野边界发生乳腺癌。

    荷兰的一项研究对霍奇金淋巴瘤放疗后至少5年发生乳腺癌的48例女性进行评估,将她们与175例配对的未发生乳腺癌的霍奇金淋巴瘤女性进行比较。发现接受化疗和斗篷野放疗的患者比单独接受斗篷野放疗的患者更不易患乳腺癌,可能是由于化疗可诱导卵巢抑制(RR=0.06;95%CI=0.01–0.45)。

    另一项研究对比了105例放疗相关乳腺癌患者和266例对照,两组之间通过年龄和放疗配对,发现类似的卵巢照射会产生保护作用。

    这些研究提示卵巢激素可能促进了放疗诱导突变后乳腺组织的增生。

    随之而来的另一个问题是,乳腺癌患者行肿瘤切除术和放疗(L-RT)后,再次患乳腺癌或其他恶性肿瘤的风险是否高于乳房切除术后患者?一项研究对比了1029例L-RT患者和1387例乳房切除术后患者,经过中位时间为15年的随访,两组患者的二次恶性肿瘤发病率无差异。

    三项RCT的证据也支持这一结论。一项研究报道了1851例女性随机分组分别接受全乳房切除术、仅肿瘤切除术和L-RT治疗,对侧乳腺癌的发生率分别为8.5%、8.8%和9.4%。

    另一项研究将701例女性患者随机分组,行根治性乳房切除术或保乳术联合术后放疗,发现两组女性的对侧乳腺癌发病率分别为每百人年10.2例和8.7例。

    第三项研究中,1665例女性被随机分配为根治性乳房切除术、全乳房切除术或全乳房切除术联合化疗组,对比三组女性25年结局发现,不同治疗组的对侧乳腺癌发病率无显著差异,总发病率为6%。

    肥胖

    肥胖与乳腺癌风险增高相关,尤其是绝经后未用HT治疗的女性。WHI观察了85917例50-79岁女性,收集了体重和乳腺癌相关危险因素的信息。

    该研究测量了这些女性的身高、体重、腰围和臀围。经过中位时间为34.8个月的随访,共有1030例女性发生浸润性乳腺癌。在从未用过HT的女性中,乳腺癌风险升高与入组时体重和体重指数(BMI)、50岁时BMI、最大BMI、成年和绝经后的体重变化及腰围与臀围相关。体重是最强的预测因素,体重超过82.2kg的女性与体重低于58.6kg的女性相比,RR为2.85(95%CI=1.81-4.49)。

    肥胖、糖尿病和胰岛素水平与乳腺癌风险之间的关联尚未有明确的结果。英国女性心脏与健康研究对60至79岁的151名乳腺癌患者和3690名女性对照进行了研究,发现在非糖尿病女性中,每增加1个单位的log(e)胰岛素水平,年龄校正后的乳腺癌OR为1.34(95%CI=1.02-1.77)。控制可能的混杂因素后,血糖与绝经前/后乳腺癌的关联仍然存在。此外,空腹血糖水平、稳态模型评分(空腹血糖值和胰岛素水平值的乘积除以22.5)、糖尿病、妊娠期糖耐量异常或糖尿病史也与乳腺癌有关。

    酒精摄入

    饮酒会增加患乳腺癌的风险。一项英国的荟萃分析纳入了53项病例对照和队列研究的数据。

    研究显示,与无饮酒史女性相比,每天饮酒35g至44g的女性患乳腺癌的RR为1.32(95%CI=1.19-1.45;P<0.001),每天饮酒45g以上的女性患乳腺癌的RR为1.46(95%CI=1.33-1.61;P<0.001)。每天酒精摄入量增加10g(即一杯酒),乳腺癌的RR增加约7%(95%CI=5.5%-8.7%;P<0.001)。在经过人种、教育、家族史、初潮年龄、身高、体重、BMI、哺乳、口服避孕药、绝经后激素治疗及激素类型、绝经年龄等因素校正后依然得到类似结果。

    有足够证据证明乳腺癌风险降低的因素

    早孕

    分娩后几年内患乳腺癌的风险增加,但之后患乳腺癌的风险长期降低,对较年轻的女性更加明显。

    研究发现,在20岁以前经历足月妊娠的女性患乳腺癌的风险是未生产或35岁以上经历首次足月妊娠女性的一半。

    国际绝经前乳腺癌协作组对来自15项前瞻性队列研究的约890000名女性的数据进行了汇总分析,证实了分娩与乳腺癌风险的相关性。与未生产女性相比,女性在分娩后20年内患ER阳性和ER阴性乳腺癌的风险增加。然而,分娩约24年后,ER阳性乳腺癌的发病风险降低,但ER阴性乳腺癌的发病风险仍然增加。因此,分娩与乳腺癌风险之间的关系是复杂的,受到分娩后的时间长度影响,与不同肿瘤特征相关。

    母乳喂养

    母乳喂养可以降低患乳腺癌的风险。

    对来自30个国家的47项流行病学研究(含50302名乳腺癌女性和96973名对照)的数据再分析后发现,进行过母乳喂养的生育后女性的乳腺癌发病率低于未母乳喂养过的生育后女性。母乳喂养的持续时间与风险降低的程度成正比。

    每分娩一次乳腺癌风险降低7.0%(95%CI=5.0%-9.0%;P<0.0001);此外,每增加12个月的母乳喂养时间,乳腺癌的RR降低4.3%(95%CI=2.9%-5.8%;P<0.0001)。

    运动锻炼

    积极的锻炼可以降低患乳腺癌的风险,尤其是对年轻的生育后女性。

    大量关于体力活动水平与乳腺癌风险之间关系的观察性研究显示,两者呈负相关。

    运动锻炼的RR平均能降低30%至40%,但饮食或乳腺癌遗传易感性等混杂因素的影响未纳入考虑。在挪威开展的一项前瞻性队列纳入了25000多名女性,发现繁重的体力劳动或每周至少4小时的运动与乳腺癌风险降低有关,尤其是在绝经前女性以及等于或低于正常体重的女性中。

    一项非裔美国女性的病例对照研究发现,每周7小时以上剧烈的娱乐性体育活动与乳腺癌发病率的降低有关。

    有充分证据证明有益的干预措施

    选择性雌激素受体调节剂

    乳腺癌辅助治疗临床试验发现他莫昔芬不仅能降低乳腺癌复发率,而且能预防对侧新发的原发性乳腺癌。

    他莫昔芬还能够帮助预防绝经后乳腺癌患者的骨密度降低。

    不良反应包括潮热、静脉血栓栓塞事件和子宫内膜癌等。

    根据这些辅助治疗试验的结果,乳腺癌预防试验(BCPT)将13388例乳腺癌高危患者随机分组,分别接受他莫昔芬或安慰剂治疗。

    由于他莫昔芬组乳腺癌发病率比安慰剂组低49%,该研究提前终止。经过4年随访,他莫昔芬和安慰剂组分别发生了85例和154例浸润性乳腺癌,原位乳腺癌分别为31和59例。他莫昔芬的另一获益为骨折发生率降低,他莫昔芬组和安慰剂组分别为47例和71例。但他莫昔芬组子宫内膜癌的发生较高(分别为33例和14例),血栓事件发生率也较高(分别为99例和70例,其中肺栓塞分别为17例和6例)。

    经过7年随访之后,BCPT更新结果与初步报告相似。

    安慰剂组中有部分女性退出;其中一部分参加了随后的试验,因此有新的女性加入安慰剂组。他莫昔芬的获益和风险与原报告中的并无显著差异,持续的获益是骨折率较低,而子宫内膜癌、血栓形成和白内障手术的风险持续增加。随访7年后,未观察到总死亡率方面的获益(RR=1.10;95%CI=0.85-1.43)。

    另有3项他莫昔芬在乳腺癌一级预防方面的研究已完成。

  • 一项在英国开展的研究
  • 研究对象包括2471名有乳腺癌和/或卵巢癌家族史的女性。随访中位时长近6年时,未观察到他莫昔芬的保护作用(RR=1.06),但随访中位时长达13年后,他莫昔芬组患乳腺癌的风险(HR=0.78;95%CI=0.58–1.04)略低。但他莫昔芬组ER阳性乳腺癌的风险显著降低(HR=0.61;95%CI=0.43-0.86)。
  • 一项在意大利开展的研究
  • 纳入了5408名乳腺癌风险为中低水平,行子宫切除术后的女性。随访中位时间近4年后,未发现他莫昔芬的保护作用。但更长期随访和亚组分析发现,他莫昔芬对激素受体阳性的乳腺癌(RR=0.24;95%CI=0.10-0.59)和期间使用HT治疗的女性(RR=0.43;95%CI=0.20-0.95)具有显著的保护作用。
  • 国际乳腺癌干预研究(IBIS-I)纳入了7152名35-70岁乳腺癌高风险的女性,并随机分为服用他莫昔芬(20mg/天)或安慰剂组,干预时间为5年。
  • 经过中位时间为50个月的随访,他莫昔芬组乳腺癌发病率(浸润性乳腺癌加原位癌的发病率从每千人年6.75例降低到4.6例)比安慰剂组低32%(95%CI=8%-50%)。ER阳性浸润性乳腺癌的RR降幅为31%,ER阴性乳腺癌发病率未降低。此外,研究还发现他莫昔芬组的全因死亡率下降(分别为25例与11例;P=0.028),但作者认为是偶然现象。他莫昔芬对乳腺癌的预防作用在治疗结束后依然持续存在,经过后续中位时间为46个月的随访,发现整个研究期间他莫昔芬组发生乳腺癌的女性比安慰剂组少27%(分别为142例和195例;RR=0.73;95%CI=0.58-0.91)。
  • 对他莫昔芬一级预防试验的荟萃分析显示,乳腺癌的发病率降低了38%,各研究间并无明显的异质性。

    ER阳性乳腺癌降低了48%。子宫内膜癌(总RR=2.4;95%CI=1.5-4.0)和血栓栓塞事件(RR=1.9;95%CI=1.4-2.6)发生率升高。这些一级预防试验均未检测乳腺癌死亡率的变化。

    有导管原位癌(DCIS)病史的女性患对侧乳腺癌的风险增加。美国国家外科辅助乳腺和肠道项目(NSABP)试验B-24评估了他莫昔芬联合L-RT对DCIS患者的益处。接受LR-T治疗的女性被随机分配是否进行他莫昔芬辅助治疗。随访6年后,接受他莫昔芬治疗的女性相较对照组浸润性和原位乳腺癌发生率较低(分别为8.2% 和13.4%;RR=0.63;95%CI=0.47-0.83)。接受他莫昔芬治疗的女性患对侧乳腺癌的风险也较低(RR=0.49;95%CI=0.26-0.87)。

    盐酸雷洛昔芬(易维特)是一种SERM, 它对乳腺有抗雌激素作用,对骨、脂代谢和凝血有雌激素作用。与他莫昔芬不同,它对子宫内膜有抗雌激素作用。

    雷洛昔芬多重结果评估(MORE)试验为一项随机、双盲临床试验,共纳入1994年-1998年间美国180个临床中心的7705例绝经后骨质疏松患者。 研究发现雷洛昔芬可降低椎体骨折的发生。乳腺癌的发生为次要终点。经过中位时间为47个月的随访,接受雷洛昔芬治疗的女性相较对照组患浸润性乳腺癌的风险降低(RR=0.25;95%CI=0.17-0.45)。

    雷洛昔芬与他莫昔芬相似,主要降低ER阳性乳腺癌的风险,而不降低ER阴性乳腺癌风险。雷洛昔芬与他莫昔芬均增加潮热和血栓栓塞事件的发生。但随访47个月后,未发现子宫内膜癌风险增加。

    MORE研究衍生研究,即易维特相关性持续转归(CORE)研究对80%的MORE参与者继续进行评估,在原分组基础之上继续增加4年时间。虽然两项研究之间相差中位时间为10个月的间隔,仅有55%的患者依然按原分组要求服药,但雷洛昔芬组的浸润性乳腺癌发病率仍然较低。与MORE相似,CORE研究的结果主要见于ER阳性浸润性乳腺癌发病风险降低。在MORE联合CORE的8年研究之中,浸润性乳腺癌发病率总体降低66%(HR=0.34;95%CI=0.22-0.50);ER阳性浸润性乳腺癌发病率降低76%(HR=0.24;95%CI=0.15-0.40)。

    雷洛昔芬心脏应用试验为一项随机、安慰剂对照试验,以评估雷洛昔芬对冠状动脉事件和浸润性乳腺癌发病率的影响。与MORE和CORE试验的结果相似,雷洛昔芬可降低浸润性乳腺癌的风险(HR=0.56;95%CI=0.38–0.83)。

    他莫昔芬和雷洛昔芬(STAR)研究(NSABP P-2)在19747例高危女性中对比了他莫昔芬与雷洛昔芬的作用,经过平均3.9年随访,发现2组的浸润性乳腺癌发病率相似,但他莫昔芬组非浸润性乳腺癌发病率低于雷洛昔芬组。子宫癌、静脉栓塞事件、白内障等不良反应的发生率在他莫昔芬组更高,两组缺血性心脏病、卒中或骨折事件的发生无差异。

    接受他莫昔芬治疗的女性中,与治疗相关的性交困难、肌肉骨骼问题和体重增加的症状发生率较低,而接受雷洛昔芬治疗的女性中,血管舒缩、膀胱控制症状、妇科症状和腿部痉挛的发生率较低。

    每1000名女性中结局指标的发生率芳香化酶抑制剂或失活剂(Al)
    他莫昔芬雷洛昔芬RR,95%CI
    浸润性乳腺癌4.34.411.02,0.82-1.28
    非浸润性乳腺癌1.512.111.4,0.98-2.00
    子宫癌21.250.62,0.35–1.08
    静脉血栓栓塞(VTE)3.82.60.7, 0.68–0.99
    白内障12.39.720.79,0.68–0.92
    症状发生率(0-4级)
    首选他莫西芬
    性交困难0.680.78P<0.001
    肌肉及骨骼问题1.11.15P=0.002
    体重增加0.760.82P<0.001
    首选雷洛昔芬
    血管舒缩症状0.960.85P<0.001
    尿失禁0.880.73P<0.001
    腿部抽筋1.10.91P<0.001
    妇科问题0.290.19P<0.001
    CI=置信区间;RR=相对风险;VTE=静脉血栓栓塞。

    芳香化酶抑制剂或失活剂(Al)

    市场上另一大类用于治疗激素敏感性乳腺癌的药物也可能有预防乳腺癌的作用。这些药物干扰肾上腺酶芳香化酶,阻止绝经后女性产生雌激素。阿那曲唑与来曲唑抑制芳香化酶活性,而依西美坦则灭活该酶。这些药物的不良反应包括乏力、关节痛、肌痛和骨密度下降,因此骨折发生率可能升高。

    既往诊断为乳腺癌的妇女在接受AI治疗时复发和新发乳腺癌的风险较低,如下研究显示:

  • 在阿那曲唑,他莫昔芬单独或联合试验中,对阿那曲唑和他莫昔芬作为原发性乳腺癌辅助治疗方案进行了比较,使用阿那曲唑的局部和远处复发率较低(7.1%),他莫昔芬组为8.5%,联合治疗组的复发率较高(9.1%)。
  • 阿那曲唑组对侧原发乳腺癌的发生率降低则更为明显(三组分别为0.4%、1.1%和0.9%)。
  • 在另一项试验中,5187名接受了5年他莫昔芬辅助治疗的女性被随机分配接受来曲唑或安慰剂。
  • 经过中位时间为2.5年的随访,研究因达到了预设有效性终点而终止。接受来曲唑治疗的患者不仅局部和远处癌复发率较低,而且新发对侧乳腺癌的发生率也较安慰剂组低(分别为14例和26例)。
  • 另一项安慰剂对照实验招募了1918名乳腺癌患者,研究了接受他莫昔芬辅助治疗后再接受5年来曲唑治疗的效果。
  • 经过中位时长6.3年的随访后,来曲唑延伸治疗组5年无病生存率为95%(95%CI=93%-96%),而对照组为91%(95%CI=89%-93%)(HR=0.66),但总生存率无差异。两组对侧新发乳腺癌发病率具有显著差异:来曲唑延伸治疗组为21%(95%CI=10%-32%),对照组49%(32%-67%)(HR=0.42)。此外,接受来曲唑治疗的女性患骨痛(分别为18%和14%)、骨折(分别为14%和9%)和新发骨质疏松症(分别为11%和6%)的发生率相比对照组增加。
  • 另一项试验将已经接受过2年他莫昔芬辅助治疗的4742例女性随机分组,继续接受他莫昔芬治疗或改用依西美坦治疗。
  • 经过中位时间为2.4年的随访期,依西美坦组女性的局部或远处转移复发率、对侧原发性乳腺癌的发病风险均低于他莫昔芬组(分别9例与20例)。
  • 芳香化酶抑制剂或失活剂也被证明可以用于乳腺癌高风险女性的预防作用,如下研究所示:

  • 一项乳腺癌预防的RCT试验中,4560例至少有一项危险因素(年龄超过60岁、5年Gail年风险>1.66%,或导管原位癌行乳房切除术后)的女性被随机分组,使用依西美坦或安慰剂。经过中位时间为35个月的随访,发现依西美坦组浸润性乳腺癌的发生率较安慰剂组降低(分别为11例和32例,HR=0.35;95%CI=0.18-0.70;35个月需治疗人数约为100)。依西美坦组不良反应发生率略高于安慰剂组,主要包括潮热(增加8%)、乏力(增加2%)。两组的骨折或心血管事件发生率无差异。
  • 国际乳腺癌干预研究II(IBIS-II)中,将乳腺癌风险较高的3864名绝经后女性随机分组,分别服用阿那曲唑(1mg/天)或安慰剂,干预时长为5年。
  • 高风险的定义因年龄而异,主要由某人群的RR值与一般人群的比值而决定:40至44岁女性中高危个体的RR比值至少为4;45至60岁女性中高危的RR比值至少为2;60至70岁的女性中高危的RR比值至少为1.5。此外,预计10年内患乳腺癌风险至少为5%的女性(根据Tyer-Cuzick模型),以及在6个月内诊断为乳腺导管原位癌并接受单侧乳房切除术治疗的女性也被认为是高风险人群而纳入研究。326名研究对象被随机分配为阿那曲唑组和安慰剂组。经过中位时间为5年的随访后,阿那曲唑组的乳腺癌(浸润性和DCIS)发生率低于安慰剂组(HR=0.47;95%CI=0.32–0.68)。激素受体阳性而非阴性的乳腺癌风险降低。根据7年以上的累积发病率估计,针对IBIS-II资格标准定义的乳腺癌高风险女性中,接受5年治疗至少预防1名乳腺癌发生所需要的最少治疗人数为36名(95%CI=33-44)。接受阿那曲唑治疗的女性比接受安慰剂的女性更有可能出现肌肉骨骼症状,包括关节痛(发生率分别为51%和46%)、关节僵硬(发生率分别为7%和5%)、手足疼痛(发生率分别为9%和8%)和腕管综合征(发生率分别为3%和2%);高血压(发生率分别为5%和3%);血管舒缩症状(发生率分别为57%和49%);干眼症(发生率分别为4%和2%)。手足疼痛与阿那曲唑治疗之间的相关性具有临界统计学意义;以上所有不良反应均与阿那曲唑治疗有显著的关联。
  • 预防性乳房切除术

    一项回顾性队列研究评估了双侧预防性乳房切除术对后续乳腺癌发生率的影响,纳入的患者为家族史评估提示有高-中度乳腺癌风险的女性。

    在BRCA突变状况未知的情况下,90%的女性接受了皮下而非全乳切除术。经过中位时间为14年的随访,425名中危女性的发病风险降低了89%;214名高危女性的风险降低了90%至94%,风险的降低程度也受乳腺癌预期发病率计算方法的影响。中危女性乳腺癌死亡率降低100%,高危女性死亡风险降低81%。由于该研究使用家族史而非基因检测作为风险指标,乳腺癌的风险有可能被高估。

    根据美国国家癌症数据库的数据,患有单侧乳腺疾病(DCIS和早期浸润性乳腺癌)的女性双侧乳腺切除率从1998年的1.9%上升到2011年的11.2%。

    在中等风险人群中,尚无研究评估预防性乳房切除术对于单侧乳腺癌患者的对侧新发乳腺癌的预防效果。

    预防性卵巢切除术

    卵巢去势和卵巢切除术可降低正常女性和胸部照射导致风险增加的女性患乳腺癌的风险。(更多信息请参阅本总结证据描述部分中的内源性雌激素部分。)观察性研究发现,通过预防性卵巢切除术预防卵巢癌发生的BRCA1或BRCA2突变女性中,乳腺癌风险比未行预防性卵巢切除术的突变携带者降低50%。

    但这些观察性研究都受到选择偏倚、患者与对照者的家族关系、卵巢切除术适应证、激素使用信息不全面的影响。一项前瞻性队列研究也有类似的发现,BRCA2突变携带者比BRCA1携带者乳腺癌风险降低的幅度更大。

    相关性不够明确的因素和干预措施

    激素类避孕药

    口服避孕药会导致使用期间的乳腺癌风险小幅增加,但随着时间推移,风险逐渐降低。

    一项设计较佳的病例对照研究发现乳腺癌风险与口服避孕药的规律服用、持续时间和是否近期使用无关。

    另一项病例对照研究发现,在35至64岁的女性中,使用注射或植入式含孕激素的避孕药不会增加患乳腺癌的风险。

    丹麦的一项全国前瞻性队列研究发现,目前正使用或近期使用激素类避孕药的女性比从未使用激素类避孕药的女性患乳腺癌的风险更高。此外,随着药物使用持续时间的延长,患乳腺癌的风险增加。然而,就绝对数量而言,口服避孕药对乳腺癌风险的影响非常小。每7690名使用药物避孕1年的女性中仅新增1例乳腺癌。

    环境因素

    职业、环境或化学暴露都被认为是乳腺癌病因之一。多项荟萃分析描述了多达134种环境化学物质的来源和暴露指标,提示其可能与癌症发生有关。

    一些研究表明,有机氯暴露,如与杀虫剂有关的有机氯暴露,可能与乳腺癌风险增加有关,

    但其他病例对照和巢式病例对照研究未得到类似的结论。

    报道两者之间存在正相关性的一些研究中提到的有机氯的鉴定标准并不一致。其中一些物质有微弱的雌激素效应,但其对乳腺癌风险的影响未得到证实。美国1972年开始禁用双对氯苯基三氯乙烷,1977年开始停止生产多氯联苯。总的来说,支持乳腺癌与特定环境暴露相关性的流行病学和动物研究证据通常较微弱。由于需要考虑的因素较多,任何与乳腺癌或其他癌症的关联结果都可能被诸如多样性、测量误差、回忆和报告偏倚等所混淆。

    相关性未经证实或已经证伪的因素和干预措施

    流产

    曾有研究提出流产是乳腺癌发病因素之一。但观察性研究的结果并不一致;阳性和阴性的结果均存在。但阳性结果的观察性研究往往缺乏严谨性,且女性对社会敏感问题的回应上可能存在偏倚。

    例如,一项研究比较了对流产有不同社会态度的地区中其回忆或报告偏倚的影响。

    美国妇产科医师学会妇科实践委员会的结论是,“最近更严谨的研究表明流产与随后的乳腺癌风险增加之间无因果关系。”

    研究使用了关于流产的前瞻性记录的数据从而避免回忆偏倚的研究,基本显示流产与随后的乳腺癌发生无关。

    膳食

    膳食改变对乳腺癌的影响可能和改变的情况有关。然而,几乎没有证据表明任何形式的饮食改变会影响乳腺癌的发病率。

    很少有随机试验比较不同膳食因素的癌症发病率。相关研究均为观察性,包括随机试验的事后分析。这些分析可能受很多偏倚影响,使得观察结果难以被解释。尤其要注意的是,这里P值和置信区间CI的解释与随机试验中主要终点的计算结果是不同的。

    1975年以前发表的一份生态研究摘要显示,世界范围内,年龄校正后乳腺癌死亡率与人均膳食脂肪消耗量之间存在正相关关系。

    病例对照研究结果并不一致。经过20年后,一份对7项队列研究结果的综合分析发现,饮食脂肪摄入总量与乳腺癌风险之间无关。

    在纳入WHI研究的48835名50-79岁的绝经后女性中进行了一项随机、对照、饮食调整研究。这项干预措施通过增加蔬菜、水果和谷物的摄入,以达到总脂肪摄入量减少20%的目标。经过中位时间为8.1年以上随访,干预组的脂肪摄入量减少了约10%,使得雌二醇和γ-生育酚水平降低,但体重下降未持续。饮食干预组的浸润性乳腺癌发病率较低但无统计学差异,HR为0.91(95%CI=0.83-1.01)。

    在全因死亡率、总死亡率或心血管事件发病率方面干预和对照组无差异。

    为研究水果蔬菜摄入对乳腺癌发生的影响,一项荟萃分析纳入了8项队列研究,包含350000名女性,新发乳腺癌7377名,多种统计模型分析结果均显示二者之间不存在相关性。

    女性健康饮食和生活随机试验

    研究了膳食因素对既往诊断为乳腺癌女性的新发乳腺癌发病率的影响。研究纳入了超过3000名女性并随机分配到两组,一组为增加水果和蔬菜摄入量,增加纤维摄入量,减少脂肪摄入量的强化组,另一组接受“每天5份”膳食指南的对照组。经过平均7.3年的随访,新发原发乳腺癌发生率未见降低、无病生存时间和总生存时间在两组间也未见差异。

    一项在西班牙开展的随机试验

    将心血管病高风险的参与者分配至三种饮食组:添加特级初榨橄榄油的地中海饮食、添加混合坚果的地中海饮食或限制性地中海饮食(经咨询后减少膳食脂肪含量)。主要心血管事件作为该试验项目的主要终点,经膳食调整后发生率降低。

    研究人员还分析了其他终点事件,包括乳腺癌发病率,未报告接受检查的人数的情况下,发生35例浸润性乳腺癌(主要心血管事件发生288例),经过平均时间为4.8年、4.3年和4.2年的随访后,乳腺癌发病率分别为8/1476(0.54%)、10/1285(0.78%)和17/1391(1.22%)。但由于其研究背景,很难确定这些差异的统计意义。

    维生素

    一些临床试验评估了某些微量营养元素对降低乳腺癌风险的作用,其主要结局事件包括心血管疾病和癌症发生。女性健康研究纳入39876例女性,随机分组服用β胡萝卜素或安慰剂,经过2年观察,发现乳腺癌发病率没有差异。

    在该研究中,隔天服用600IU维生素E未见与癌症发生具有关联。

    女性抗氧化心血管研究在8171名女性中,评估了维生素C、维生素E或β-胡萝卜素的摄入与癌症和浸润性乳腺癌发生的关系,发现并无关联。

    两年后,其中5442名女性被随机分配为联合服用1.5mg叶酸、50mg维生素B6和1mg维生素B12或服用安慰剂组。经过7.3年的随访,乳腺癌和各类浸润性癌症的发病率在两组间无明显差异。

    芬维A胺

    是一种维生素A类似物,在之前的临床研究中已被证明可以减少乳腺癌的发生。意大利一项III期临床试验在2972名年龄在30至70岁的女性中比较了接受5年芬维A胺干预治疗与不干预的人群结局,这些女性既往均接受I期乳腺癌切除术或DCIS切除术。经过中位时间为97个月的随访,其对侧乳腺癌(P=0.642)、同侧乳腺癌(P=0.177)、远处转移、非乳腺恶性肿瘤的发生率和和全因死亡率在统计学上无显著差异。

    主动吸烟和被动吸烟

    主动吸烟在乳腺癌病因学中的潜在作用已被研究了30多年,没有明确的证据表明两者之间存在相关性。

    自20世纪90年代中期以来,吸烟与乳腺癌的相关性研究更多关注被动吸烟。

    最近的一项荟萃分析表明,被动吸烟与乳腺癌之间的总体不存在相关性,一些研究中的阳性结果可能是由于方法学的缺陷(如乳腺癌确诊后评估暴露因素)所导致。

    腋下除臭剂/止汗剂

    尽管在非专业出版物上警告女性腋下除臭剂和止汗剂会导致乳腺癌,但没有证据支持这些观点。一项访谈式研究纳入813名乳腺癌患者和793名对照,发现使用止汗剂、除臭剂或刮胡刀与乳腺癌风险之间没有关联。

    与之相反,一项对437名乳腺癌幸存者的研究发现,使用止汗剂/除臭剂并更频繁地刮腋毛的女性癌症诊断的年龄更轻。但其可能的原因之一是,这些女性月经初潮较早或内源性激素水平较高,而这两者是乳腺癌和体毛增加的共同风险因素。

    他汀类药物

    两项设计较佳的RCT试验荟萃分析

    以及RCT加上观察性研究

    未发现他汀类药物增加或降低乳腺癌风险的证据。

    双膦酸盐

    有研究关注口服和静脉注射双膦酸盐治疗高钙血症和骨质疏松症对预防乳腺癌的潜在收益。初步观察性研究表明,使用这些药物大约1至4年的女性乳腺癌发病率较低。

    但患骨质疏松症女性会比骨密度正常的女性患乳腺癌的风险更低,这一情况可能对上述研究结果造成混淆。其他证据来源于在乳腺癌女性病例中开展的研究:这些药物的使用与对侧新发乳腺癌的降低有关。

    在此背景下,开展了两项大型随机安慰剂对照试验。骨折干预试验(FIT)使用阿仑膦酸钠或安慰剂治疗6194名绝经后骨质疏松症女性,经过3.8年后,两组间乳腺癌发病率无差异,分别为1.8%和1.5%(HR=1.24;CI=0.84-1.83)。另一项实验为每年使用1 次唑来膦酸的临床结果和发病率降低试验的关键部位骨折试验(HORIZON-PRT),其对7580名绝经后骨质疏松症女性给予唑来膦酸钠静脉注射治疗或安慰剂治疗,结果发现,在2.8年时,乳腺癌的发病率在两组间无差异,分别为0.8%和0.9%(HR=1.15;CI=0.7-1.89)。

    夜间轮班工作

    2007年,世界卫生组织(WHO)下属的国际癌症研究机构(IARC)将涉及昼夜节律紊乱的轮班工作列为可能的乳腺癌致癌因素。主要证据来自动物研究。当时人类研究的证据有限。

    2013年,一项纳入15项流行病学研究的荟萃分析发现,在曾经上过夜班的女性中,乳腺癌发病率增加的证据并不充分。

    2016年,合并3项新近在英国开展的前瞻性研究(涉及近80万名女性)和其他7项前瞻性研究的结果发现,乳腺癌发病率与夜班工作不存在相关性。即使是长达20年以上的夜间轮班工作,其发病风险比值的置信区间也很窄,(RR=1.01;95%CI=0.93-1.10)。这些结果表明乳腺癌与长期夜间轮班工作不存在任何程度的相关性。

    2003年在英国的亲代研究强调了乳腺癌的风险因素和致病因素。在105000名女性的前瞻性队列中,通过问卷调查获得了基线和20岁时卧室光线水平的信息。经过平均6.1年的随访,观察到1775例乳腺癌。校正包括夜班工作等混杂因素后,发现无证据表明夜间卧室光线亮度与乳腺癌风险有关。夜间光线最亮组相对于最暗组,乳腺癌发病率的HR为1.01(95%CI=0.88–1.15)。

    参考文献

  • American Cancer Society: Cancer Facts and Figures 2019. Atlanta, Ga: American Cancer Society, 2019. Available online. Last accessed December 12, 2019.
  • Howlader N, Noone AM, Krapcho M, et al., eds.: SEER Cancer Statistics Review, 1975-2012. Bethesda, Md: National Cancer Institute, 2015. Also available online. Last accessed February 20, 2020.
  • Altekruse SF, Kosary CL, Krapcho M, et al.: SEER Cancer Statistics Review, 1975-2007. Bethesda, Md: National Cancer Institute, 2010. Also available online. Last accessed December 12, 2019.
  • American Cancer Society: Cancer Facts and Figures 2018. Atlanta, Ga: American Cancer Society, 2018. Available online. Last accessed February 20, 2020.
  • Pfeiffer RM, Mitani A, Matsuno RK, et al.: Racial differences in breast cancer trends in the United States (2000-2004). J Natl Cancer Inst 100 (10): 751-2, 2008.
  • Boone CW, Kelloff GJ, Freedman LS: Intraepithelial and postinvasive neoplasia as a stochastic continuum of clonal evolution, and its relationship to mechanisms of chemopreventive drug action. J Cell Biochem Suppl 17G: 14-25, 1993.
  • Kelloff GJ, Boone CW, Steele VE, et al.: Progress in cancer chemoprevention: perspectives on agent selection and short-term clinical intervention trials. Cancer Res 54 (7 Suppl): 2015s-2024s, 1994.
  • Knabbe C, Lippman ME, Wakefield LM, et al.: Evidence that transforming growth factor-beta is a hormonally regulated negative growth factor in human breast cancer cells. Cell 48 (3): 417-28, 1987.
  • Parkin DM: Cancers of the breast, endometrium and ovary: geographic correlations. Eur J Cancer Clin Oncol 25 (12): 1917-25, 1989.
  • Dunn JE: Breast cancer among American Japanese in the San Francisco Bay area. Natl Cancer Inst Monogr 47: 157-60, 1977.
  • Kliewer EV, Smith KR: Breast cancer mortality among immigrants in Australia and Canada. J Natl Cancer Inst 87 (15): 1154-61, 1995.
  • Brinton LA, Schairer C, Hoover RN, et al.: Menstrual factors and risk of breast cancer. Cancer Invest 6 (3): 245-54, 1988.
  • Collaborative Group on Hormonal Factors in Breast Cancer: Menarche, menopause, and breast cancer risk: individual participant meta-analysis, including 118 964 women with breast cancer from 117 epidemiological studies. Lancet Oncol 13 (11): 1141-51, 2012.
  • Ritte R, Lukanova A, Tjønneland A, et al.: Height, age at menarche and risk of hormone receptor-positive and -negative breast cancer: a cohort study. Int J Cancer 132 (11): 2619-29, 2013.
  • Endogenous Hormones and Breast Cancer Collaborative Group: Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. J Natl Cancer Inst 94 (8): 606-16, 2002.
  • Key TJ, Appleby PN, Reeves GK, et al.: Circulating sex hormones and breast cancer risk factors in postmenopausal women: reanalysis of 13 studies. Br J Cancer 105 (5): 709-22, 2011.
  • Kaaks R, Rinaldi S, Key TJ, et al.: Postmenopausal serum androgens, oestrogens and breast cancer risk: the European prospective investigation into cancer and nutrition. Endocr Relat Cancer 12 (4): 1071-82, 2005.
  • Kaaks R, Berrino F, Key T, et al.: Serum sex steroids in premenopausal women and breast cancer risk within the European Prospective Investigation into Cancer and Nutrition (EPIC). J Natl Cancer Inst 97 (10): 755-65, 2005.
  • Smith PG, Doll R: Late effects of x irradiation in patients treated for metropathia haemorrhagica. Br J Radiol 49 (579): 224-32, 1976.
  • Trichopoulos D, MacMahon B, Cole P: Menopause and breast cancer risk. J Natl Cancer Inst 48 (3): 605-13, 1972.
  • Feinleib M: Breast cancer and artificial menopause: a cohort study. J Natl Cancer Inst 41 (2): 315-29, 1968.
  • Kampert JB, Whittemore AS, Paffenbarger RS: Combined effect of childbearing, menstrual events, and body size on age-specific breast cancer risk. Am J Epidemiol 128 (5): 962-79, 1988.
  • Hirayama T, Wynder EL: A study of the epidemiology of cancer of the breast. II. The influence of hysterectomy. Cancer 15: 28-38, 1962 Jan-Feb.
  • Colditz GA, Kaphingst KA, Hankinson SE, et al.: Family history and risk of breast cancer: nurses' health study. Breast Cancer Res Treat 133 (3): 1097-104, 2012.
  • Miki Y, Swensen J, Shattuck-Eidens D, et al.: A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 266 (5182): 66-71, 1994.
  • Futreal PA, Liu Q, Shattuck-Eidens D, et al.: BRCA1 mutations in primary breast and ovarian carcinomas. Science 266 (5182): 120-2, 1994.
  • Wooster R, Neuhausen SL, Mangion J, et al.: Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science 265 (5181): 2088-90, 1994.
  • Kuchenbaecker KB, Hopper JL, Barnes DR, et al.: Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 317 (23): 2402-2416, 2017.
  • Antoniou A, Pharoah PD, Narod S, et al.: Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet 72 (5): 1117-30, 2003.
  • Chen S, Parmigiani G: Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol 25 (11): 1329-33, 2007.
  • National Cancer Institute: SEER Stat Fact Sheets: Female Breast Cancer. Bethesda, MD: National Cancer Institute. Available online. Last accessed September 26, 2019.
  • Swift M, Morrell D, Massey RB, et al.: Incidence of cancer in 161 families affected by ataxia-telangiectasia. N Engl J Med 325 (26): 1831-6, 1991.
  • Cybulski C, Wokołorczyk D, Jakubowska A, et al.: Risk of breast cancer in women with a CHEK2 mutation with and without a family history of breast cancer. J Clin Oncol 29 (28): 3747-52, 2011.
  • Boyd NF, Guo H, Martin LJ, et al.: Mammographic density and the risk and detection of breast cancer. N Engl J Med 356 (3): 227-36, 2007.
  • Razzaghi H, Troester MA, Gierach GL, et al.: Mammographic density and breast cancer risk in White and African American Women. Breast Cancer Res Treat 135 (2): 571-80, 2012.
  • McCormack VA, dos Santos Silva I: Breast density and parenchymal patterns as markers of breast cancer risk: a meta-analysis. Cancer Epidemiol Biomarkers Prev 15 (6): 1159-69, 2006.
  • Gierach GL, Ichikawa L, Kerlikowske K, et al.: Relationship between mammographic density and breast cancer death in the Breast Cancer Surveillance Consortium. J Natl Cancer Inst 104 (16): 1218-27, 2012.
  • Breast cancer and hormone replacement therapy: collaborative reanalysis of data from 51 epidemiological studies of 52,705 women with breast cancer and 108,411 women without breast cancer. Collaborative Group on Hormonal Factors in Breast Cancer. Lancet 350 (9084): 1047-59, 1997.
  • Hulley S, Furberg C, Barrett-Connor E, et al.: Noncardiovascular disease outcomes during 6.8 years of hormone therapy: Heart and Estrogen/progestin Replacement Study follow-up (HERS II). JAMA 288 (1): 58-66, 2002.
  • Writing Group for the Women's Health Initiative Investigators: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA 288 (3): 321-33, 2002.
  • Chlebowski RT, Anderson GL, Gass M, et al.: Estrogen plus progestin and breast cancer incidence and mortality in postmenopausal women. JAMA 304 (15): 1684-92, 2010.
  • Chlebowski RT, Hendrix SL, Langer RD, et al.: Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women's Health Initiative Randomized Trial. JAMA 289 (24): 3243-53, 2003.
  • Chlebowski RT, Manson JE, Anderson GL, et al.: Estrogen plus progestin and breast cancer incidence and mortality in the Women's Health Initiative Observational Study. J Natl Cancer Inst 105 (8): 526-35, 2013.
  • Anderson GL, Limacher M, Assaf AR, et al.: Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA 291 (14): 1701-12, 2004.
  • LaCroix AZ, Chlebowski RT, Manson JE, et al.: Health outcomes after stopping conjugated equine estrogens among postmenopausal women with prior hysterectomy: a randomized controlled trial. JAMA 305 (13): 1305-14, 2011.
  • Anderson GL, Chlebowski RT, Aragaki AK, et al.: Conjugated equine oestrogen and breast cancer incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women's Health Initiative randomised placebo-controlled trial. Lancet Oncol 13 (5): 476-86, 2012.
  • Schierbeck LL, Rejnmark L, Tofteng CL, et al.: Effect of hormone replacement therapy on cardiovascular events in recently postmenopausal women: randomised trial. BMJ 345: e6409, 2012.
  • Beral V, Reeves G, Bull D, et al.: Breast cancer risk in relation to the interval between menopause and starting hormone therapy. J Natl Cancer Inst 103 (4): 296-305, 2011.
  • Li CI, Malone KE, Porter PL, et al.: Relationship between long durations and different regimens of hormone therapy and risk of breast cancer. JAMA 289 (24): 3254-63, 2003.
  • Chlebowski RT, Anderson GL: The influence of time from menopause and mammography on hormone therapy-related breast cancer risk assessment. J Natl Cancer Inst 103 (4): 284-5, 2011.
  • Prentice RL, Chlebowski RT, Stefanick ML, et al.: Conjugated equine estrogens and breast cancer risk in the Women's Health Initiative clinical trial and observational study. Am J Epidemiol 167 (12): 1407-15, 2008.
  • Chlebowski RT, Kuller LH, Prentice RL, et al.: Breast cancer after use of estrogen plus progestin in postmenopausal women. N Engl J Med 360 (6): 573-87, 2009.
  • Cronin KA, Ravdin PM, Edwards BK: Sustained lower rates of breast cancer in the United States. Breast Cancer Res Treat 117 (1): 223-4, 2009.
  • Ravdin PM, Cronin KA, Howlader N, et al.: The decrease in breast-cancer incidence in 2003 in the United States. N Engl J Med 356 (16): 1670-4, 2007.
  • Parkin DM: Is the recent fall in incidence of post-menopausal breast cancer in UK related to changes in use of hormone replacement therapy? Eur J Cancer 45 (9): 1649-53, 2009.
  • Lambe M, Wigertz A, Holmqvist M, et al.: Reductions in use of hormone replacement therapy: effects on Swedish breast cancer incidence trends only seen after several years. Breast Cancer Res Treat 121 (3): 679-83, 2010.
  • Renard F, Vankrunkelsven P, Van Eycken L, et al.: Decline in breast cancer incidence in the Flemish region of Belgium after a decline in hormonal replacement therapy. Ann Oncol 21 (12): 2356-60, 2010.
  • Farhat GN, Walker R, Buist DS, et al.: Changes in invasive breast cancer and ductal carcinoma in situ rates in relation to the decline in hormone therapy use. J Clin Oncol 28 (35): 5140-6, 2010.
  • DeSantis C, Howlader N, Cronin KA, et al.: Breast cancer incidence rates in U.S. women are no longer declining. Cancer Epidemiol Biomarkers Prev 20 (5): 733-9, 2011.
  • John EM, Kelsey JL: Radiation and other environmental exposures and breast cancer. Epidemiol Rev 15 (1): 157-62, 1993.
  • Evans JS, Wennberg JE, McNeil BJ: The influence of diagnostic radiography on the incidence of breast cancer and leukemia. N Engl J Med 315 (13): 810-5, 1986.
  • Andrieu N, Easton DF, Chang-Claude J, et al.: Effect of chest X-rays on the risk of breast cancer among BRCA1/2 mutation carriers in the international BRCA1/2 carrier cohort study: a report from the EMBRACE, GENEPSO, GEO-HEBON, and IBCCS Collaborators' Group. J Clin Oncol 24 (21): 3361-6, 2006.
  • Bhatia S, Robison LL, Oberlin O, et al.: Breast cancer and other second neoplasms after childhood Hodgkin's disease. N Engl J Med 334 (12): 745-51, 1996.
  • Hancock SL, Tucker MA, Hoppe RT: Breast cancer after treatment of Hodgkin's disease. J Natl Cancer Inst 85 (1): 25-31, 1993.
  • Travis LB, Hill DA, Dores GM, et al.: Breast cancer following radiotherapy and chemotherapy among young women with Hodgkin disease. JAMA 290 (4): 465-75, 2003.
  • Sankila R, Garwicz S, Olsen JH, et al.: Risk of subsequent malignant neoplasms among 1,641 Hodgkin's disease patients diagnosed in childhood and adolescence: a population-based cohort study in the five Nordic countries. Association of the Nordic Cancer Registries and the Nordic Society of Pediatric Hematology and Oncology. J Clin Oncol 14 (5): 1442-6, 1996.
  • van Leeuwen FE, Klokman WJ, Stovall M, et al.: Roles of radiation dose, chemotherapy, and hormonal factors in breast cancer following Hodgkin's disease. J Natl Cancer Inst 95 (13): 971-80, 2003.
  • Obedian E, Fischer DB, Haffty BG: Second malignancies after treatment of early-stage breast cancer: lumpectomy and radiation therapy versus mastectomy. J Clin Oncol 18 (12): 2406-12, 2000.
  • Fisher B, Anderson S, Bryant J, et al.: Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347 (16): 1233-41, 2002.
  • Veronesi U, Cascinelli N, Mariani L, et al.: Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347 (16): 1227-32, 2002.
  • 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 347 (8): 567-75, 2002.
  • Morimoto LM, White E, Chen Z, et al.: Obesity, body size, and risk of postmenopausal breast cancer: the Women's Health Initiative (United States). Cancer Causes Control 13 (8): 741-51, 2002.
  • Wolin KY, Carson K, Colditz GA: Obesity and cancer. Oncologist 15 (6): 556-65, 2010.
  • Lawlor DA, Smith GD, Ebrahim S: Hyperinsulinaemia and increased risk of breast cancer: findings from the British Women's Heart and Health Study. Cancer Causes Control 15 (3): 267-75, 2004.
  • Hamajima N, Hirose K, Tajima K, et al.: Alcohol, tobacco and breast cancer--collaborative reanalysis of individual data from 53 epidemiological studies, including 58,515 women with breast cancer and 95,067 women without the disease. Br J Cancer 87 (11): 1234-45, 2002.
  • Pike MC, Krailo MD, Henderson BE, et al.: 'Hormonal' risk factors, 'breast tissue age' and the age-incidence of breast cancer. Nature 303 (5920): 767-70, 1983.
  • Lambe M, Hsieh C, Trichopoulos D, et al.: Transient increase in the risk of breast cancer after giving birth. N Engl J Med 331 (1): 5-9, 1994.
  • Henderson BE, Pike MC, Ross RK, et al.: Epidemiology and risk factors. In: Bonadonna G, ed.: Breast Cancer: Diagnosis and Management. Chichester, NY: John Wiley & Sons, 1984, pp 15-33.
  • Gail MH, Brinton LA, Byar DP, et al.: Projecting individualized probabilities of developing breast cancer for white females who are being examined annually. J Natl Cancer Inst 81 (24): 1879-86, 1989.
  • Nichols HB, Schoemaker MJ, Cai J, et al.: Breast Cancer Risk After Recent Childbirth: A Pooled Analysis of 15 Prospective Studies. Ann Intern Med 170 (1): 22-30, 2019.
  • Col: Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet 360 (9328): 187-95, 2002.
  • Furberg H, Newman B, Moorman P, et al.: Lactation and breast cancer risk. Int J Epidemiol 28 (3): 396-402, 1999.
  • Bernstein L, Henderson BE, Hanisch R, et al.: Physical exercise and reduced risk of breast cancer in young women. J Natl Cancer Inst 86 (18): 1403-8, 1994.
  • Friedenreich CM: Physical activity and cancer prevention: from observational to intervention research. Cancer Epidemiol Biomarkers Prev 10 (4): 287-301, 2001.
  • Thune I, Brenn T, Lund E, et al.: Physical activity and the risk of breast cancer. N Engl J Med 336 (18): 1269-75, 1997.
  • Adams-Campbell LL, Rosenberg L, Rao RS, et al.: Strenuous physical activity and breast cancer risk in African-American women. J Natl Med Assoc 93 (7-8): 267-75, 2001 Jul-Aug.
  • Nayfield SG, Karp JE, Ford LG, et al.: Potential role of tamoxifen in prevention of breast cancer. J Natl Cancer Inst 83 (20): 1450-9, 1991.
  • Love RR, Barden HS, Mazess RB, et al.: Effect of tamoxifen on lumbar spine bone mineral density in postmenopausal women after 5 years. Arch Intern Med 154 (22): 2585-8, 1994.
  • 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 14 (1): 78-84, 1996.
  • Costantino JP, Kuller LH, Ives DG, et al.: Coronary heart disease mortality and adjuvant tamoxifen therapy. J Natl Cancer Inst 89 (11): 776-82, 1997.
  • McDonald CC, Stewart HJ: Fatal myocardial infarction in the Scottish adjuvant tamoxifen trial. The Scottish Breast Cancer Committee. BMJ 303 (6800): 435-7, 1991.
  • Rutqvist LE, Mattsson A: Cardiac and thromboembolic morbidity among postmenopausal women with early-stage breast cancer in a randomized trial of adjuvant tamoxifen. The Stockholm Breast Cancer Study Group. J Natl Cancer Inst 85 (17): 1398-406, 1993.
  • Fisher B, Costantino JP, Redmond CK, et al.: Endometrial cancer in tamoxifen-treated breast cancer patients: findings from the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-14. J Natl Cancer Inst 86 (7): 527-37, 1994.
  • Bergman L, Beelen ML, Gallee MP, et al.: Risk and prognosis of endometrial cancer after tamoxifen for breast cancer. Comprehensive Cancer Centres' ALERT Group. Assessment of Liver and Endometrial cancer Risk following Tamoxifen. Lancet 356 (9233): 881-7, 2000.
  • Cuzick J, Powles T, Veronesi U, et al.: Overview of the main outcomes in breast-cancer prevention trials. Lancet 361 (9354): 296-300, 2003.
  • Redmond CK, Wickerham DL, Cronin W, et al.: The NSABP breast cancer prevention trial (BCPT): a progress report. [Abstract] Proceedings of the American Society of Clinical Oncology 12: A-78, 69, 1993.
  • Fisher B, Costantino JP, Wickerham DL, et al.: Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 90 (18): 1371-88, 1998.
  • Fisher B, Costantino JP, Wickerham DL, et al.: Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst 97 (22): 1652-62, 2005.
  • Powles T, Eeles R, Ashley S, et al.: Interim analysis of the incidence of breast cancer in the Royal Marsden Hospital tamoxifen randomised chemoprevention trial. Lancet 352 (9122): 98-101, 1998.
  • Veronesi U, Maisonneuve P, Costa A, et al.: Prevention of breast cancer with tamoxifen: preliminary findings from the Italian randomised trial among hysterectomised women. Italian Tamoxifen Prevention Study. Lancet 352 (9122): 93-7, 1998.
  • Cuzick J, Forbes J, Edwards R, et al.: First results from the International Breast Cancer Intervention Study (IBIS-I): a randomised prevention trial. Lancet 360 (9336): 817-24, 2002.
  • Powles TJ, Ashley S, Tidy A, et al.: Twenty-year follow-up of the Royal Marsden randomized, double-blinded tamoxifen breast cancer prevention trial. J Natl Cancer Inst 99 (4): 283-90, 2007.
  • Veronesi U, Maisonneuve P, Rotmensz N, et al.: Tamoxifen for the prevention of breast cancer: late results of the Italian Randomized Tamoxifen Prevention Trial among women with hysterectomy. J Natl Cancer Inst 99 (9): 727-37, 2007.
  • Martino S, Costantino J, McNabb M, et al.: The role of selective estrogen receptor modulators in the prevention of breast cancer: comparison of the clinical trials. Oncologist 9 (2): 116-25, 2004.
  • Cuzick J, Forbes JF, Sestak I, et al.: Long-term results of tamoxifen prophylaxis for breast cancer--96-month follow-up of the randomized IBIS-I trial. J Natl Cancer Inst 99 (4): 272-82, 2007.
  • 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 353 (9169): 1993-2000, 1999.
  • Khovidhunkit W, Shoback DM: Clinical effects of raloxifene hydrochloride in women. Ann Intern Med 130 (5): 431-9, 1999.
  • Cauley JA, Norton L, Lippman ME, et al.: Continued breast cancer risk reduction in postmenopausal women treated with raloxifene: 4-year results from the MORE trial. Multiple outcomes of raloxifene evaluation. Breast Cancer Res Treat 65 (2): 125-34, 2001.
  • Cummings SR, Eckert S, Krueger KA, et al.: The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation. JAMA 281 (23): 2189-97, 1999.
  • Martino S, Cauley JA, Barrett-Connor E, et al.: Continuing outcomes relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst 96 (23): 1751-61, 2004.
  • Grady D, Cauley JA, Geiger MJ, et al.: Reduced incidence of invasive breast cancer with raloxifene among women at increased coronary risk. J Natl Cancer Inst 100 (12): 854-61, 2008.
  • Vogel VG, Costantino JP, Wickerham DL, et al.: Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 295 (23): 2727-41, 2006.
  • Land SR, Wickerham DL, Costantino JP, et al.: Patient-reported symptoms and quality of life during treatment with tamoxifen or raloxifene for breast cancer prevention: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 295 (23): 2742-51, 2006.
  • The ATAC Trialists' Group. Arimidex, tamoxifen alone or in combination: 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 359 (9324): 2131-9, 2002.
  • 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 349 (19): 1793-802, 2003.
  • Goss PE, Ingle JN, Pritchard KI, et al.: Extending Aromatase-Inhibitor Adjuvant Therapy to 10 Years. N Engl J Med 375 (3): 209-19, 2016.
  • 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 350 (11): 1081-92, 2004.
  • Goss PE, Ingle JN, Alés-Martínez JE, et al.: Exemestane for breast-cancer prevention in postmenopausal women. N Engl J Med 364 (25): 2381-91, 2011.
  • Cuzick J, Sestak I, Forbes JF, et al.: Anastrozole for prevention of breast cancer in high-risk postmenopausal women (IBIS-II): an international, double-blind, randomised placebo-controlled trial. Lancet 383 (9922): 1041-8, 2014.
  • Hartmann LC, Schaid DJ, Woods JE, et al.: Efficacy of bilateral prophylactic mastectomy in women with a family history of breast cancer. N Engl J Med 340 (2): 77-84, 1999.
  • Kummerow KL, Du L, Penson DF, et al.: Nationwide trends in mastectomy for early-stage breast cancer. JAMA Surg 150 (1): 9-16, 2015.
  • Rebbeck TR, Levin AM, Eisen A, et al.: Breast cancer risk after bilateral prophylactic oophorectomy in BRCA1 mutation carriers. J Natl Cancer Inst 91 (17): 1475-9, 1999.
  • Kauff ND, Satagopan JM, Robson ME, et al.: Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 346 (21): 1609-15, 2002.
  • Rebbeck TR, Lynch HT, Neuhausen SL, et al.: Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N Engl J Med 346 (21): 1616-22, 2002.
  • Kauff ND, Domchek SM, Friebel TM, et al.: Risk-reducing salpingo-oophorectomy for the prevention of BRCA1- and BRCA2-associated breast and gynecologic cancer: a multicenter, prospective study. J Clin Oncol 26 (8): 1331-7, 2008.
  • Breast cancer and hormonal contraceptives: further results. Collaborative Group on Hormonal Factors in Breast Cancer. Contraception 54 (3 Suppl): 1S-106S, 1996.
  • Marchbanks PA, McDonald JA, Wilson HG, et al.: Oral contraceptives and the risk of breast cancer. N Engl J Med 346 (26): 2025-32, 2002.
  • Strom BL, Berlin JA, Weber AL, et al.: Absence of an effect of injectable and implantable progestin-only contraceptives on subsequent risk of breast cancer. Contraception 69 (5): 353-60, 2004.
  • Mørch LS, Skovlund CW, Hannaford PC, et al.: Contemporary Hormonal Contraception and the Risk of Breast Cancer. N Engl J Med 377 (23): 2228-2239, 2017.
  • Brody JG, Rudel RA, Michels KB, et al.: Environmental pollutants, diet, physical activity, body size, and breast cancer: where do we stand in research to identify opportunities for prevention? Cancer 109 (12 Suppl): 2627-34, 2007.
  • Rodgers KM, Udesky JO, Rudel RA, et al.: Environmental chemicals and breast cancer: An updated review of epidemiological literature informed by biological mechanisms. Environ Res 160: 152-182, 2018.
  • Wolff MS, Toniolo PG, Lee EW, et al.: Blood levels of organochlorine residues and risk of breast cancer. J Natl Cancer Inst 85 (8): 648-52, 1993.
  • Høyer AP, Grandjean P, Jørgensen T, et al.: Organochlorine exposure and risk of breast cancer. Lancet 352 (9143): 1816-20, 1998.
  • Shames LS, Munekata MT, Pike MC: Re: Blood levels of organochlorine residues and risk of breast cancer. J Natl Cancer Inst 86 (21): 1642-3, 1994.
  • Krieger N, Wolff MS, Hiatt RA, et al.: Breast cancer and serum organochlorines: a prospective study among white, black, and Asian women. J Natl Cancer Inst 86 (8): 589-99, 1994.
  • Hunter DJ, Hankinson SE, Laden F, et al.: Plasma organochlorine levels and the risk of breast cancer. N Engl J Med 337 (18): 1253-8, 1997.
  • Laden F, Collman G, Iwamoto K, et al.: 1,1-Dichloro-2,2-bis(p-chlorophenyl)ethylene and polychlorinated biphenyls and breast cancer: combined analysis of five U.S. studies. J Natl Cancer Inst 93 (10): 768-76, 2001.
  • Ward EM, Schulte P, Grajewski B, et al.: Serum organochlorine levels and breast cancer: a nested case-control study of Norwegian women. Cancer Epidemiol Biomarkers Prev 9 (12): 1357-67, 2000.
  • Laden F, Hankinson SE, Wolff MS, et al.: Plasma organochlorine levels and the risk of breast cancer: an extended follow-up in the Nurses' Health Study. Int J Cancer 91 (4): 568-74, 2001.
  • Berry D: Multiplicities in cancer research: ubiquitous and necessary evils. J Natl Cancer Inst 104 (15): 1124-32, 2012.
  • Dickersin K: The existence of publication bias and risk factors for its occurrence. JAMA 263 (10): 1385-9, 1990.
  • Huang Y, Zhang X, Li W, et al.: A meta-analysis of the association between induced abortion and breast cancer risk among Chinese females. Cancer Causes Control 25 (2): 227-36, 2014.
  • Sanderson M, Shu XO, Jin F, et al.: Abortion history and breast cancer risk: results from the Shanghai Breast Cancer Study. Int J Cancer 92 (6): 899-905, 2001.
  • Beral V, Bull D, Doll R, et al.: Breast cancer and abortion: collaborative reanalysis of data from 53 epidemiological studies, including 83,000 women with breast cancer from 16 countries. Lancet 363 (9414): 1007-16, 2004.
  • Melbye M, Wohlfahrt J, Olsen JH, et al.: Induced abortion and the risk of breast cancer. N Engl J Med 336 (2): 81-5, 1997.
  • Rookus MA, van Leeuwen FE: Induced abortion and risk for breast cancer: reporting (recall) bias in a Dutch case-control study. J Natl Cancer Inst 88 (23): 1759-64, 1996.
  • Committee on Gynecologic Practice: ACOG Committee Opinion No. 434: induced abortion and breast cancer risk. Obstet Gynecol 113 (6): 1417-8, 2009.
  • Henderson KD, Sullivan-Halley J, Reynolds P, et al.: Incomplete pregnancy is not associated with breast cancer risk: the California Teachers Study. Contraception 77 (6): 391-6, 2008.
  • Lash TL, Fink AK: Null association between pregnancy termination and breast cancer in a registry-based study of parous women. Int J Cancer 110 (3): 443-8, 2004.
  • Michels KB, Xue F, Colditz GA, et al.: Induced and spontaneous abortion and incidence of breast cancer among young women: a prospective cohort study. Arch Intern Med 167 (8): 814-20, 2007.
  • Wu JQ, Li YY, Ren JC, et al.: Induced abortion and breast cancer: results from a population-based case control study in China. Asian Pac J Cancer Prev 15 (8): 3635-40, 2014.
  • Braüner CM, Overvad K, Tjønneland A, et al.: Induced abortion and breast cancer among parous women: a Danish cohort study. Acta Obstet Gynecol Scand 92 (6): 700-5, 2013.
  • Guo J, Huang Y, Yang L, et al.: Association between abortion and breast cancer: an updated systematic review and meta-analysis based on prospective studies. Cancer Causes Control 26 (6): 811-9, 2015.
  • Carroll KK, Khor HT: Dietary fat in relation to tumorigenesis. Prog Biochem Pharmacol 10: 308-53, 1975.
  • Hunter DJ, Spiegelman D, Adami HO, et al.: Cohort studies of fat intake and the risk of breast cancer--a pooled analysis. N Engl J Med 334 (6): 356-61, 1996.
  • Prentice RL, Caan B, Chlebowski RT, et al.: Low-fat dietary pattern and risk of invasive breast cancer: the Women's Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 295 (6): 629-42, 2006.
  • Howard BV, Van Horn L, Hsia J, et al.: Low-fat dietary pattern and risk of cardiovascular disease: the Women's Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 295 (6): 655-66, 2006.
  • Smith-Warner SA, Spiegelman D, Yaun SS, et al.: Intake of fruits and vegetables and risk of breast cancer: a pooled analysis of cohort studies. JAMA 285 (6): 769-76, 2001.
  • Pierce JP, Natarajan L, Caan BJ, et al.: Influence of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis following treatment for breast cancer: the Women's Healthy Eating and Living (WHEL) randomized trial. JAMA 298 (3): 289-98, 2007.
  • Toledo E, Salas-Salvadó J, Donat-Vargas C, et al.: Mediterranean Diet and Invasive Breast Cancer Risk Among Women at High Cardiovascular Risk in the PREDIMED Trial: A Randomized Clinical Trial. JAMA Intern Med 175 (11): 1752-60, 2015.
  • Estruch R, Ros E, Salas-Salvadó J, et al.: Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med 368 (14): 1279-90, 2013.
  • Lee IM, Cook NR, Manson JE, et al.: Beta-carotene supplementation and incidence of cancer and cardiovascular disease: the Women's Health Study. J Natl Cancer Inst 91 (24): 2102-6, 1999.
  • Lee IM, Cook NR, Gaziano JM, et al.: Vitamin E in the primary prevention of cardiovascular disease and cancer: the Women's Health Study: a randomized controlled trial. JAMA 294 (1): 56-65, 2005.
  • Lin J, Cook NR, Albert C, et al.: Vitamins C and E and beta carotene supplementation and cancer risk: a randomized controlled trial. J Natl Cancer Inst 101 (1): 14-23, 2009.
  • Zhang SM, Cook NR, Albert CM, et al.: Effect of combined folic acid, vitamin B6, and vitamin B12 on cancer risk in women: a randomized trial. JAMA 300 (17): 2012-21, 2008.
  • Costa A, Formelli F, Chiesa F, et al.: Prospects of chemoprevention of human cancers with the synthetic retinoid fenretinide. Cancer Res 54 (7 Suppl): 2032s-2037s, 1994.
  • Veronesi U, De Palo G, Marubini E, et al.: Randomized trial of fenretinide to prevent second breast malignancy in women with early breast cancer. J Natl Cancer Inst 91 (21): 1847-56, 1999.
  • The Health Consequences of Smoking: A Report of the Surgeon General. Atlanta, Ga: U.S. Department of Health and Human Services, CDC, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health, 2004. Also available online. Last accessed February 20, 2020.
  • U.S. Department of Health and Human Services: The Health Consequences of Involuntary Exposure to Tobacco Smoke: A Report of the Surgeon General. Atlanta, Ga: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, Coordinating Center for Health Promotion, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health, 2006. Also available online. Last accessed September 26, 2019.
  • Pirie K, Beral V, Peto R, et al.: Passive smoking and breast cancer in never smokers: prospective study and meta-analysis. Int J Epidemiol 37 (5): 1069-79, 2008.
  • Mirick DK, Davis S, Thomas DB: Antiperspirant use and the risk of breast cancer. J Natl Cancer Inst 94 (20): 1578-80, 2002.
  • McGrath KG: An earlier age of breast cancer diagnosis related to more frequent use of antiperspirants/deodorants and underarm shaving. Eur J Cancer Prev 12 (6): 479-85, 2003.
  • Dale KM, Coleman CI, Henyan NN, et al.: Statins and cancer risk: a meta-analysis. JAMA 295 (1): 74-80, 2006.
  • Bonovas S, Filioussi K, Tsavaris N, et al.: Use of statins and breast cancer: a meta-analysis of seven randomized clinical trials and nine observational studies. J Clin Oncol 23 (34): 8606-12, 2005.
  • Newcomb PA, Trentham-Dietz A, Hampton JM: Bisphosphonates for osteoporosis treatment are associated with reduced breast cancer risk. Br J Cancer 102 (5): 799-802, 2010.
  • Rennert G, Pinchev M, Rennert HS: Use of bisphosphonates and risk of postmenopausal breast cancer. J Clin Oncol 28 (22): 3577-81, 2010.
  • Chlebowski RT, Chen Z, Cauley JA, et al.: Oral bisphosphonate use and breast cancer incidence in postmenopausal women. J Clin Oncol 28 (22): 3582-90, 2010.
  • Cardwell CR, Abnet CC, Veal P, et al.: Exposure to oral bisphosphonates and risk of cancer. Int J Cancer 131 (5): E717-25, 2012.
  • Monsees GM, Malone KE, Tang MT, et al.: Bisphosphonate use after estrogen receptor-positive breast cancer and risk of contralateral breast cancer. J Natl Cancer Inst 103 (23): 1752-60, 2011.
  • Hue TF, Cummings SR, Cauley JA, et al.: Effect of bisphosphonate use on risk of postmenopausal breast cancer: results from the randomized clinical trials of alendronate and zoledronic acid. JAMA Intern Med 174 (10): 1550-7, 2014.
  • Straif K, Baan R, Grosse Y, et al.: Carcinogenicity of shift-work, painting, and fire-fighting. Lancet Oncol 8 (12): 1065-1066, 2007.
  • Kamdar BB, Tergas AI, Mateen FJ, et al.: Night-shift work and risk of breast cancer: a systematic review and meta-analysis. Breast Cancer Res Treat 138 (1): 291-301, 2013.
  • Travis RC, Balkwill A, Fensom GK, et al.: Night Shift Work and Breast Cancer Incidence: Three Prospective Studies and Meta-analysis of Published Studies. J Natl Cancer Inst 108 (12): , 2016.
  • Johns LE, Jones ME, Schoemaker MJ, et al.: Domestic light at night and breast cancer risk: a prospective analysis of 105 000 UK women in the Generations Study. Br J Cancer 118 (4): 600-606, 2018.
  • Breast Cancer Prevention (PDQ®)

    Description of the Evidence

    Incidence and Mortality

    With an estimated 268,600 cases expected, breast cancer will be the most frequently diagnosed nonskin malignancy in U.S. women in 2019.

    Also in 2019, breast cancer will kill an estimated 41,760 women, second only to lung cancer as a cause of cancer mortality in women. Breast cancer also occurs in men, and it is estimated that 2,670 new cases will be diagnosed in 2019.

    Despite a prior long-term trend of gradually increasing breast cancer incidence in women, data from the Surveillance, Epidemiology, and End Results Program show a decrease in breast cancer mortality of 1.8% per year from 2007 to 2016.

    The major risk factor for breast cancer is advancing age. A 30-year-old woman has a 1 in 250 chance of being diagnosed with breast cancer in the next 10 years, whereas a 70-year-old woman has a 1 in 27 chance.

    Breast cancer incidence and mortality risk also vary on the basis of geography, culture, race, ethnicity, and socioeconomic status. Compared with other races, white women have a higher incidence of breast cancer that may be attributable, in part, to screening behavior. However, breast cancer incidence rates increased slightly in black women by 0.3% per year between 2005 and 2014, resulting in the convergence of rates in blacks with those in whites.

    Screening by mammography decreases breast cancer mortality by identifying cases for treatment at an earlier stage. However, screening also identifies more cases than would become symptomatic in a woman’s lifetime, so screening increases breast cancer incidence. (Refer to the Overdiagnosis section in the PDQ summary on Breast Cancer Screening for more information.)

    Etiology and Pathogenesis of Breast Cancer

    Breast cancer develops when a series of genetic mutations occurs.

    Initially, mutations do not change the histologic appearance of the tissue, but accumulated mutations will result in hyperplasia, dysplasia, carcinoma in situ, and eventually, invasive cancer.

    The longer a woman lives, the more somatic mutations occur, and the more likely it is that these mutations will produce populations of cells that will evolve into malignancies. Estrogen and progestin cause growth and proliferation of breast cells that may work through growth factors such as transforming growth factor (TGF)-alpha.

    These hormones, whether endogenous or exogenous, may promote the development and proliferation of breast cancer cells.

    International variation in breast cancer rates may be explained by differences in genetics, reproductive factors, diet, exercise and screening behavior. Some of these factors are modifiable, as evidenced by the observation that Japanese immigrants to the United States increase their breast cancer risk from lower Japanese levels to higher American levels within two generations.

    Endogenous Estrogen

    Many of the risk factors for breast cancer suggest that longer exposure to endogenous estrogen plays a role in the development of the disease. Women who experienced menarche at age 11 years or younger have about a 20% greater chance of developing breast cancer than do those who experienced menarche at age 14 years or older.

    Women who experience late menopause also have an increased risk. Women who develop breast cancer tend to have higher endogenous estrogen and androgen levels.

    Conversely, women who experience premature menopause have a lower risk of breast cancer. Following ovarian ablation, breast cancer risk may be reduced as much as 75% depending on age, weight, and parity, with the greatest reduction for young, thin, nulliparous women.

    The removal of one ovary also reduces the risk of breast cancer but to a lesser degree than does the removal of both ovaries.

    Other hormonal changes also influence breast cancer risk. (Refer to the Early pregnancy and Breast-feeding sections in the Factors With Adequate Evidence of Decreased Risk of Breast Cancer section of this summary for more information.)

    The interaction of endogenous estrogen levels, insulin levels, and obesity—all of which affect breast cancer risk—are poorly understood but suggest strategies for interventions to decrease that risk. It is likely that reproductive risk factors interact with predisposing genotypes. For example, in the Nurses’ Health Study,

    the associations between age at first birth, menarche, and menopause and the development of breast cancer were observed only among women without a family history of breast cancer in a mother or sister.

    Inherited Risk

    Breast cancer risk increases in women with a positive family history, particularly if first-degree relatives are affected.

    The following risk assessment models, derived from databases, cohort, and case-control studies, quantitate this risk:

  • Breast Cancer Risk Assessment Tool [Gail Model].
  • Breast Cancer Surveillance Consortium [BCSC] Risk Calculator.
  • Breast Cancer Referral Screening Tool [B-RST].
  • Hall Detailed Breast Risk Calculator.
  • IBIS Breast Cancer Risk Calculator Tool.
  • Specific abnormal alleles are associated with approximately 5% of breast cancers. (Refer to the PDQ summary on Genetics of Breast and Gynecologic Cancers for more information.) Mutations in BRCA genes are inherited in an autosomal dominant fashion and are highly penetrant in causing cancer, often at a younger age.

    Family history and mutation location within the BRCA1 or BRCA2 gene may contribute to the risk of cancer development among those with an inherited predisposition to breast cancer.

    The lifetime risk of breast cancer is 55% to 65% for BRCA1 mutation carriers and 45% to 47% for BRCA2 mutation carriers.

    In comparison, the lifetime risk of breast cancer is 12.4% in the general population.

    Some women inherit a susceptibility to mutagens or growth factors, which increase breast cancer risk.

    (Refer to the Ionizing radiation exposure section in the Factors With Adequate Evidence of Increased Risk of Breast Cancer section of this summary for more information.)

    Increased Breast Density

    Widespread use of screening mammograms has revealed varying amounts of mammographically dense tissue and that women with a greater proportion of dense tissue have a higher incidence of breast cancer. Mammographic density also confounds the identification of cancers by that technology. The extent of increased risk was described in a report of three nested case-control studies in screened populations with 1,112 matched case-control pairs. Compared with women with density comprising less than 10% of breast tissue, women with density in 75% or more of their breast had an increased risk of breast cancer (odds ratio [OR], 4.7; 95% confidence interval [CI], 3.0–7.4), whether the cancer was detected by screening (OR, 3.5; 95% CI, 2.0–6.2) or detected less than 12 months after a negative screening examination (OR, 17.8; 95% CI, 4.8–65.9). Increased risk of breast cancer, whether detected by screening or other means, persisted for at least 8 years after study entry and was greater in younger women than in older women. For women younger than the median age of 56 years, 26% of all breast cancers and 50% of cancers detected less than 12 months after a negative screening test were identified in women with mammographic breast density of 50% or more.

    Women with dense breasts have increased risk, proportionate to the degree of density. This increased relative risk (RR) ranges from 1.79 for women with slightly increased breast density to 4.64 for women with very dense breasts, compared with women who have the lowest breast density.

    There is no increased risk of breast cancer mortality among women with dense breast tissue.

    Factors With Adequate Evidence of Increased Risk of Breast Cancer

    Hormone therapy

    Based on a 1997 reanalysis of 51 epidemiological studies encompassing more than 150,000 women, hormone therapy (HT) after menopause was shown to be associated with increased breast cancer risk.

    The Heart and Estrogen/Progestin Replacement Study supported this finding in 2002.

    In this study, 2,763 women with coronary heart disease at a mean age of 67 years were randomly assigned to receive either estrogen and progestin therapy or placebo. After a mean follow-up of 6.8 years, the RR for breast cancer was 1.27 (95% CI, 0.84–1.94). Although not statistically significant, the RR estimate is consistent with the much larger Women’s Health Initiative (WHI), also published in 2002.

    The WHI investigated the effect of hormones and dietary interventions on heart disease and breast cancer risk.

    Women aged 50 to 79 years with intact uteri were randomly assigned to receive combined conjugated estrogen with continuous progestin (n = 8,506) or placebo (n = 8,102). The trial was terminated early because combined HT did not decrease coronary heart disease risk but did increase the risk of stroke and breast cancer. An increased rate of invasive breast cancer risk (hazard ratio [HR], 1.24; 95% CI, 1.02–1.50), but not for in situ breast cancer, was observed in all subgroups of women. The combined HT-related cancers had similar grade, histology, and expression of estrogen receptor (ER), progesterone receptor, and HER2/neu, with a trend toward larger size and higher incidence of lymph node metastases in the combined HT group.

    Extended follow-up of a mean of 11 years showed higher breast cancer–specific mortality for the HT group (25 vs. 12 deaths, 0.03% vs. 0.01% per year; HR, 1.95; 95% CI, 1.0–4.04; P = .049). Combined HT was also associated with a higher percentage of abnormal mammograms.

    The WHI observational study was conducted in parallel with the WHI randomized controlled trial (RCT), recruiting postmenopausal women aged 50 to 79 years. An analysis was conducted in the observational study of the WHI to further examine the prognosis of women taking combination HT who were diagnosed with breast cancer and the risks based on time between menopause and initiation of HT. After a mean follow-up of 11.3 years, the annualized incidence of breast cancer among women using estrogen plus progestin was 0.60%, compared with 0.42% among nonusers (HR, 1.55; 95% CI, 1.41–1.70). Survival after the diagnosis of breast cancer was similar for combined HT users and nonusers. Death from breast cancer was higher among combined HT users than among nonusers, but the difference was not statistically significant (HR, 1.3; 94% CI, 0.90–1.93). Risks were highest among women initiating HT at the time of menopause, and risks diminished but persisted with increasing time between menopause and starting combination HT. All-cause mortality after the diagnosis of breast cancer was statistically significantly higher among combined HT users than among nonusers (HR, 1.87; 95% CI, 1.37–2.54.) Overall, these findings were consistent with results from the RCT.

    The WHI also studied women who had previously undergone a hysterectomy and thus were not at risk for endometrial cancer, which is associated with unopposed estrogen therapy. Women aged 50 to 79 years (N = 10,739) were randomly assigned to receive conjugated equine estrogen (CEE) or placebo. This trial was also stopped early because of an increased risk of stroke and no improvement in a global risk-benefit index.

    After an average 6.8 years of follow-up, breast cancer incidence was lower in the group receiving CEE (0.26% per year vs. 0.33%; HR, 0.77; 95% CI, 0.59–1.01). The global risk-benefit index was slightly worse for CEE.

    An extended follow-up for a median of 11.8 years included 78% of the trial participants. Results seen in the initial study persisted, with a similar risk reduction for breast cancer in CEE recipients (HR, 0.77; 95% CI, 0.62–0.95) and a decrease in breast cancer mortality (6 vs. 16 deaths; HR, 0.37; 95% CI, 0.13–0.91). All-cause mortality was also lower in the CEE group (0.046% vs. 0.076% per year; HR, 0.62; 95% CI, 0.39–0.97). After CEE was discontinued, the risk of stroke decreased in the postintervention period. Over the entire follow-up period, there was no difference in the incidence of coronary heart disease, deep vein thrombosis, stroke, hip fracture, or colorectal cancer.

    Breast cancer incidence was similar for women who initiated CEE or placebo within the first 5 years after onset of menopause (HR, 1.06; 95% CI, 0.74–1.51).

    A Danish trial of HT for 1,006 women entering menopause was designed to evaluate cardiovascular outcomes. Combined HT (triphasic estradiol and norethisterone) was given to 407 women with intact uteri, and estradiol was given to 95 women who had undergone hysterectomy. Controls (407 with intact uteri and 97 with hysterectomy) were not treated. At 10 years, there was considerable contamination. Only one-half of the women assigned to the HT group were still taking the prescribed HT, and 22% of the control women had begun HT. Cardiovascular outcomes favored HT-treated women, and there was no difference in breast cancer incidence.

    Observational studies augment the information obtained in RCTs.

    The Million Women Study

    recruited 1,084,110 women aged 50 to 64 years in the United Kingdom between 1996 and 2001 and obtained information about HT use and other personal details. The women were followed for breast cancer incidence and death. One-half of the women had used HT. At 2.6 years of follow up, there were 9,364 invasive breast cancers; at 4.1 years, there were 637 breast cancer deaths. Current users of HT at recruitment were more likely than never-users to develop breast cancer (adjusted RR = 1.66; 95% CI, 1.58–1.75; P < .0001) and to die from the disease (adjusted RR, 1.22; 95% CI, 1.00–1.48; P = .05). Past users of HT were, however, not at an increased risk of incident or fatal breast cancer (1.01 [95% CI, 0.94–1.09] and 1.05 [95% CI, 0.82–1.34], respectively). Incidence was significantly increased for current users of estrogen only (RR, 1.30; 95% CI, 1.21–1.40; P < .0001), combined HT (RR, 2.00; 95% CI, 1.88–2.12; P < .0001), and tibolone (RR, 1.45; 95% CI, 1.25–1.68; P < .0001). The magnitude of the associated risk was substantially greater for combined HT than for other types of HT (P < .0001).

    A population-based survey of 965 women with breast cancer and 1,007 controls was conducted by the Cancer Surveillance System of Puget Sound. It showed that combined HT users had a 1.7-fold increased risk of invasive breast cancer, whereas estrogen-only users did not.

    The association between the use of combined HT and increased breast cancer risk is consistent throughout all the trials. In contrast, the association between estrogen-only HT and breast cancer incidence is confusing because some studies show increased risk and some show protection. It is possible that the timing of estrogen-only HT in relation to the onset of menopause is critical. Furthermore, observational studies may not account for different screening behavior between HT users and nonusers, whereas RCTs, by design, will control that variable.

    Following publication of the WHI results, HT use dropped dramatically in the United States and elsewhere. Follow-up of WHI participants on the combined HT arm demonstrated a rapid decrease in the elevated breast cancer risk of therapy within 2 years, despite similar rates of mammography screening.

    Analysis of changes in breast cancer rates in the United States observed a sharp decline in breast cancer incidence rates from 2002 to 2003 among women aged 50 years and older, especially for ER–positive cancers.

    Similarly, in multiple countries where HT use was high, breast cancer rates decreased in a similar time frame, coincident with decreases in prescribing patterns and/or reported prevalence of use.

    A study among women receiving regular mammography screening supports that the observed sharp decline from 2002 to 2003 in breast cancer incidence was primarily caused by withdrawal of HT rather than declines in mammography rates.

    After the decline in breast cancer incidence from 2002 to 2003, rates in the United States stabilized.

    Ionizing radiation exposure

    A well-established relationship exists between exposure to ionizing radiation and subsequent breast cancer.

    Excess breast cancer risk has been observed in association with atomic bomb exposure, frequent fluoroscopy for tuberculosis, and radiation therapy for acne, tinea, thymic enlargement, postpartum mastitis, and lymphoma. Risk is higher for the young, especially around puberty. An estimate of the risk of breast cancer associated with medical radiology puts the figure at less than 1% of the total.

    However, it has been theorized that certain populations, such as AT heterozygotes, are at an increased risk of breast cancer from radiation exposure.

    A large cohort study of women who carry mutations of BRCA1 or BRCA2 concluded that chest x-rays increase the risk of breast cancer even more (RR, 1.54; 95% CI, 1.1–2.1), especially for women who had x-rays before age 20 years.

    Women treated for Hodgkin lymphoma by age 16 years have a subsequent risk up to 35% of developing breast cancer by age 40 years.

    Higher radiation doses (median dose, 40 Gy in breast cancer cases) and treatment between the ages of 10 and 16 years are associated with higher risk.

    Unlike the risk for secondary leukemia, the risk of treatment-related breast cancer does not abate with duration of follow-up, persisting more than 25 years after treatment.

    In these studies, most patients (85%–100%) who developed breast cancer did so either within the field of radiation or at the margin.

    A Dutch study examined 48 women who developed breast cancer at least 5 years after treatment for Hodgkin disease and compared them with 175 matched female Hodgkin disease patients who did not develop breast cancer. Patients treated with chemotherapy and mantle radiation were less likely to develop breast cancer than were those treated with mantle radiation alone, possibly because of chemotherapy-induced ovarian suppression (RR, 0.06; 95% CI, 0.01–0.45).

    Another study of 105 radiation-associated breast cancer patients and 266 age-matched and radiation-matched controls showed a similar protective effect for ovarian radiation.

    These studies suggest that ovarian hormones promote the proliferation of breast tissue with radiation-induced mutations.

    The question arises whether breast cancer patients treated with lumpectomy and radiation therapy (L-RT) are at higher risk for second breast malignancies or other malignancies than are those treated by mastectomy. Outcomes of 1,029 L-RT patients were compared with outcomes of 1,387 patients who underwent mastectomies. After a median follow-up of 15 years, there was no difference in the risk of second malignancies.

    Further evidence from three RCTs is also reassuring. One report of 1,851 women randomly assigned to undergo total mastectomy, lumpectomy alone, or L-RT showed rates of contralateral breast cancer to be 8.5%, 8.8%, and 9.4%, respectively.

    Another study of 701 women randomly assigned to undergo radical mastectomy or breast-conserving surgery followed by radiation therapy demonstrated the rate of contralateral breast carcinomas per 100 woman-years to be 10.2 versus 8.7, respectively.

    The third study compared 25-year outcomes of 1,665 women randomly assigned to undergo radical mastectomy, total mastectomy, or total mastectomy with radiation. There was no significant difference in the rate of contralateral breast cancer according to treatment group, and the overall rate was 6%.

    Obesity

    Obesity is associated with increased breast cancer risk, especially among postmenopausal women who do not use HT. The WHI observed 85,917 women aged 50 to 79 years and collected information on weight history and known risk factors for breast cancer.

    Height, weight, and waist and hip circumferences were measured. With a median follow-up of 34.8 months, 1,030 of the women developed invasive breast cancer. Among the women who never used HT, increased breast cancer risk was associated with weight at entry, body mass index (BMI) at entry, BMI at age 50 years, maximum BMI, adult and postmenopausal weight change, and waist and hip circumferences. Weight was the strongest predictor, with a RR of 2.85 (95% CI, 1.81–4.49) for women weighing more than 82.2 kg, compared with those weighing less than 58.7 kg.

    The association between obesity, diabetes, and insulin levels with breast cancer risk have been studied but not clearly defined. The British Women’s Heart and Health Study of women aged 60 to 79 years compared 151 women who had a diagnosis of breast cancer with 3,690 women who did not. The age-adjusted OR was 1.34 (95% CI, 1.02–1.77) for each unit increase in log(e) insulin level among nondiabetic women. The association was observed, after adjustment for confounders and for potential mediating factors, for both pre- and postmenopausal breast cancers. In addition, fasting glucose level, homeostatic model assessment score (the product of fasting glucose and insulin levels divided by 22.5), diabetes, and a history of gestational glycosuria or diabetes were also associated with breast cancer.

    Alcohol

    Alcohol consumption increases the risk of breast cancer. A British meta-analysis included individual data from 53 case-control and cohort studies.

    Compared with the RR of breast cancer for women who reported no alcohol consumption, the RR of breast cancer was 1.32 (95% CI, 1.19–1.45; P < .001) for women consuming 35 g to 44 g of alcohol per day and 1.46 (95% CI, 1.33–1.61; P < .001) for those consuming at least 45 g of alcohol per day. The RR of breast cancer increases by about 7% (95% CI, 5.5%–8.7%; P < .001) for each 10 g of alcohol (i.e., one drink) consumed per day. These findings persist after stratification for race, education, family history, age at menarche, height, weight, BMI, breast-feeding, oral contraceptive use, menopausal hormone use and type, and age at menopause.

    Factors With Adequate Evidence of Decreased Risk of Breast Cancer

    Early pregnancy

    Childbirth is followed by an increase in risk of breast cancer for several years, and then a long-term reduction in risk, which is greater for younger women.

    In one study, women who experienced a first full-term pregnancy before age 20 years were half as likely to develop breast cancer as nulliparous women or women whose first full-term pregnancy occurred at age 35 years or older.

    The association of childbirth with breast cancer risk was demonstrated by the International Premenopausal Breast Cancer Collaborative Group, which undertook a pooled analysis of individual-level data from about 890,000 women from 15 prospective cohort studies. When compared with nulliparous women, parous women had an increased risk of developing both ER–positive and ER–negative breast cancer for up to 20 years after childbirth. However, after about 24 years, the risk of developing ER–positive breast cancer decreased, but the risk of developing ER–negative breast cancer remained elevated. Thus, the association between parity and breast cancer risk is complex and appears to be influenced by the time period after childbirth and tumor phenotype.

    Breast-feeding

    Breast-feeding is associated with a decreased risk of breast cancer.

    A reanalysis of individual data from 47 epidemiological studies in 30 countries of 50,302 women with breast cancer and 96,973 controls revealed that breast cancer incidence was lower in parous women who had ever breast-fed than in parous women who had not. It was also proportionate to duration of breast-feeding.

    The RR of breast cancer decreased by 4.3% (95%, CI, 2.9%–5.8%; P < .0001) for every 12 months of breast-feeding in addition to a decrease of 7.0% (95% CI, 5.0%–9.0%; P < .0001) for each birth.

    Exercise

    Active exercise may reduce breast cancer risk, particularly in young parous women.

    Numerous observational studies on the relationship between the level of physical activity and breast cancer risk have shown an inverse relationship.

    The average RR reduction is 30% to 40%, but confounding variables—such as diet or a genetic predisposition to breast cancer—have not been addressed. A prospective study of more than 25,000 Norwegian women found that heavy manual labor or at least 4 hours of exercise per week is associated with decreased breast cancer risk, especially in premenopausal women and those of normal or lower-than-normal body weight.

    In a case-control study of African American women, strenuous recreational physical activity more than 7 hours per week was associated with decreased breast cancer incidence.

    Interventions With Adequate Evidence of Benefit

    Selective estrogen receptor modulators (SERMs)

    Data from adjuvant breast cancer trials using tamoxifen have shown that tamoxifen not only suppresses the recurrence of breast cancer but also prevents new primary contralateral breast cancers.

    Tamoxifen also maintains bone density among postmenopausal women with breast cancer.

    Adverse effects include hot flashes, venous thromboembolic events, and endometrial cancer.

    These adjuvant trial results were the basis for the Breast Cancer Prevention Trial (BCPT) that randomly assigned 13,388 patients at elevated risk of breast cancer to receive tamoxifen or placebo.

    The study was closed early because of a 49% reduction in the incidence of breast cancer for the tamoxifen group (85 vs. 154 invasive breast cancer cases and 31 vs. 59 in situ cases at 4 years). Tamoxifen-treated women also had fewer fractures (47 vs. 71) but more endometrial cancer (33 vs. 14 cases) and thrombotic events (99 vs. 70), including pulmonary emboli (17 vs. 6).

    An update of the BCPT results after 7 years of follow-up demonstrated results similar to those in the initial report.

    There were some dropouts among women in the placebo arm; some of them enrolled in a subsequent trial, so new women were added to the placebo group. Benefits and risks of tamoxifen were not significantly different from those in the original report, with persistent benefit of fewer fractures and persistent increased risk of endometrial cancer, thrombosis, and cataract surgery. No overall mortality benefit was observed after 7 years of follow-up (RR, 1.10; 95% CI, 0.85–1.43).

    Three other trials of tamoxifen for primary prevention of breast cancer have been completed.

  • A study in the United Kingdom
  • focused on 2,471 women at increased breast cancer risk because of their family history of breast and/or ovarian cancer. After a median follow-up of nearly 6 years, no protective effect of tamoxifen was detected (RR, 1.06), but there was a slight reduction in breast cancer risk in the tamoxifen arm (HR, 0.78; 95% CI, 0.58–1.04) at a median of 13 years. However, risk of ER-positive breast cancer was significantly reduced in the treatment arm (HR, 0.61; 95% CI, 0.43–0.86), an effect noted predominantly in the posttreatment period.
  • An Italian study
  • focused on 5,408 women who had undergone hysterectomy and who were described as low to normal risk. After a median follow-up of nearly 4 years, no protective effect of tamoxifen was observed. Longer follow-up and subgroup analysis in this trial found a protective effect of tamoxifen among women at high risk for hormone receptor–positive breast cancer (RR, 0.24; 95% CI, 0.10–0.59) and among women who were taking HT during the trial (RR, 0.43; 95% CI, 0.20–0.95).
  • The International Breast Cancer Intervention Study (IBIS-I) randomly assigned 7,152 women aged 35 to 70 years who were at an increased risk of breast cancer to receive tamoxifen (20 mg/day ) or placebo for 5 years.
  • After a median follow-up of 50 months, fewer tamoxifen-treated women had developed invasive or in situ breast cancer (absolute rate, 4.6 vs. 6.75 per 1,000 woman-years; risk reduction, 32%; 95% CI, 8%–50%). The RR reduction in ER-positive invasive breast cancer was 31%; there was no reduction in ER-negative cancers. There was an excess of all-cause mortality in the tamoxifen group (25 vs. 11; P = .028), which the authors attributed to chance. The beneficial effect of tamoxifen on breast cancer persisted after active treatment, with a median posttherapy follow-up of 46 months; 27% fewer women in the tamoxifen arm developed breast cancer (142 vs. 195 cases, respectively; RR, 0.73, 95% CI, 0.58–0.91).
  • A meta-analysis of these primary prevention tamoxifen trials showed a 38% reduction in the incidence of breast cancer without statistically significant heterogeneity.

    ER-positive tumors were reduced by 48%. Rates of endometrial cancer were increased (consensus RR, 2.4; 95% CI, 1.5–4.0), as were venous thromboembolic events (RR, 1.9; 95% CI, 1.4–2.6). None of these primary prevention trials was designed to detect differences in breast cancer mortality.

    Women with a history of ductal carcinoma in situ (DCIS) are at increased risk for contralateral breast cancer. The National Surgical Adjuvant Breast and Bowel Project (NSABP) trial B-24 addressed their management. Women were randomly assigned to receive L-RT either with or without adjuvant tamoxifen. At 6 years, the tamoxifen-treated women had fewer invasive and in situ breast cancers (8.2% vs. 13.4%; RR, 0.63; 95% CI, 0.47–0.83). The risk of contralateral breast cancer was also lower in women treated with tamoxifen (RR, 0.49; 95% CI, 0.26–0.87).

    Raloxifene hydrochloride (Evista) is a SERM that has antiestrogenic effects on breast and estrogenic effects on bone, lipid metabolism, and blood clotting. Unlike tamoxifen, it has antiestrogenic effects on the endometrium.

    The Multiple Outcomes of Raloxifene Evaluation (MORE) trial was a randomized, double-blind trial that evaluated 7,705 postmenopausal women with osteoporosis from 1994 to 1998 at 180 clinical centers in the United States. Vertebral fractures were reduced. The effect on breast cancer incidence was a secondary endpoint. After a median follow-up of 47 months, the risk of invasive breast cancer decreased in the raloxifene-treated women (RR, 0.25; 95% CI, 0.17–0.45).

    As with tamoxifen, raloxifene reduced the risk of ER-positive breast cancer but not ER-negative breast cancer and was associated with an excess risk of hot flashes and thromboembolic events. No excess risk of endometrial cancer or hyperplasia was observed after 47 months of follow-up.

    An extension of the MORE trial was the Continuing Outcomes Relevant to Evista (CORE) trial, which studied about 80% of MORE participants in their randomly assigned groups for an additional 4 years. Although there was a median 10-month gap between the two studies, and only about 55% of women were adherent to their assigned medications, the raloxifene group continued to experience a lower incidence of invasive ER-positive breast cancer. The overall reduction in invasive breast cancer during the 8 years of MORE and CORE was 66% (HR, 0.34; 95% CI, 0.22–0.50); the reduction for ER-positive invasive breast cancer was 76% (HR, 0.24; 95% CI, 0.15–0.40).

    The Raloxifene Use for the Heart trial was a randomized, placebo-controlled trial to evaluate the effects of raloxifene on incidence of coronary events and invasive breast cancer. As in the MORE and CORE studies, raloxifene reduced the risk of invasive breast cancer (HR, 0.56; 95% CI, 0.38–0.83).

    The Study of Tamoxifen and Raloxifene (STAR) (NSABP P-2) compared tamoxifen and raloxifene in 19,747 high-risk women who were monitored for a mean of 3.9 years. Invasive breast cancer incidence was approximately the same for both drugs, but there were fewer noninvasive cancers in the tamoxifen group. Adverse events of uterine cancer, venous thrombolic events, and cataracts were more common in tamoxifen-treated women, and there was no difference in ischemic heart disease events, strokes, or fractures.

    Treatment-associated symptoms of dyspareunia, musculoskeletal problems, and weight gain occurred less frequently in tamoxifen-treated women, whereas vasomotor flushing, bladder control symptoms, gynecologic symptoms, and leg cramps occurred less frequently in those receiving raloxifene.

    Incidence of Outcomes Per 1,000 WomenAromatase inhibitors or inactivators (Als)
    TamoxifenRaloxifeneRR, 95% CI
    Invasive breast cancer4.34.411.02, 0.82–1.28
    Noninvasive breast cancer1.512.111.4, 0.98–2.00
    Uterine cancer 2.01.250.62, 0.35–1.08
    VTE3.82.60.7, 0.68–0.99
    Cataracts12.39.720.79, 0.68–0.92
    Incidence of Symptoms (0–4 scale)
    Favor Tamoxifen
    Dyspareunia0.680.78P < .001
    Musculoskeletal problems1.101.15P = .002
    Weight gain0.760.82P < .001
    Favor Raloxifene
    Vasomotor symptoms0.960.85P < .001
    Bladder control symptoms0.880.73P < .001
    Leg cramps1.100.91P < .001
    Gynecologic problems0.290.19P < .001
    CI = confidence interval; RR = relative risk; VTE = venous thromboembolism.

    Aromatase inhibitors or inactivators (Als)

    Another class of agents that is commercially available for the treatment of women with hormone-sensitive breast cancer may also prevent breast cancer. These drugs interfere with aromatase, the adrenal enzyme that allows estrogen production in postmenopausal women. Anastrozole and letrozole inhibit aromatase activity, whereas exemestane inactivates the enzyme. Side effects for all three drugs include fatigue, arthralgia, myalgia, decreased bone mineral density, and increased fracture rate.

    Women with a previous diagnosis of breast cancer have a lower risk of recurrence and of new breast cancers when treated with AIs, as shown in the following studies:

  • In the Arimidex, Tamoxifen, Alone or in Combination trial, which compared anastrozole with tamoxifen as adjuvant therapy for primary breast cancer, the rate of locoregional and distant recurrence was lower for anastrozole (7.1% vs. 8.5%) but higher for the combination (9.1%).
  • Anastrozole was also more effective in reducing the incidence of new contralateral breast cancer (0.4% vs. 1.1% vs. 0.9%).
  • In another trial, 5,187 women who received 5 years of adjuvant tamoxifen were randomly assigned to receive either letrozole or placebo.
  • After only 2.5 years of median follow-up, the study was terminated because previously defined efficacy endpoints had been reached. Not only did patients treated with letrozole have a lower incidence of locoregional and distant cancer recurrence, they also had a lower incidence of new contralateral breast cancer (14 vs. 26).
  • Another placebo-controlled trial of 1,918 women with breast cancer examined the effect of extending letrozole treatment for an additional 5 years in women who had received adjuvant tamoxifen followed by 5 years of letrozole.
  • At a median of 6.3 years from study entry, the extended letrozole group had an improved 5-year disease-free survival of 95% (95% CI, 93%–96%) compared with 91% (95% CI, 89%–93%) for the control group (HR, 0.66) but no difference in overall survival. The difference in new contralateral breast cancer diagnoses was statistically significant: 21% (95% CI, 10%–32%) for the extended letrozole group compared with 49% (32%–67%) for the control group (HR, 0.42). Women treated with letrozole had an increased risk of bone pain (18% vs. 14%), bone fracture (14% vs. 9%), and new-onset osteoporosis (11% vs. 6%).
  • A trial randomly assigned 4,742 women who had received 2 years of adjuvant tamoxifen to either continue the tamoxifen or switch to exemestane.
  • After 2.4 years of median follow-up, the exemestane group had a decreased risk of local or metastatic recurrence and a decreased incidence of new contralateral breast cancer (9 vs. 20).
  • Aromatase inhibitors or inactivators also have been shown to prevent breast cancer in women at increased risk, as shown in the following studies:

  • An RCT of primary prevention of breast cancer compared exemestane with placebo in 4,560 women with at least one risk factor (age >60 years, a Gail 5-year risk >1.66%, or a history of DCIS with mastectomy). After 35 months of median follow-up, invasive breast cancer was diagnosed less frequently in the exemestane group (11 vs. 32; HR, 0.35; 95% CI, 0.18–0.70; number needed-to-treat, about 100 for 35 months). Compared with the placebo group, the exemestane-treated women had more hot flashes (increase, 8%) and fatigue (increase, 2%) but no difference in fractures or cardiovascular events.
  • The International Breast Cancer Intervention Study II (IBIS-II) randomly assigned 3,864 postmenopausal women who were at increased risk of developing breast cancer to receive either daily anastrazole (1 mg) or placebo for 5 years.
  • The definition of high risk varied by age and was defined by the RR compared with the general population: women aged 40 to 44 years had to have an RR of at least 4; women aged 45 to 60 years had to have an RR of at least 2; and women aged 60 to 70 years had to have an RR of at least 1.5. Alternatively, women with an estimated 10-year risk of developing breast cancer of at least 5% (per the Tyer-Cuzick model) were eligible for inclusion. Women with DCIS diagnosed within 6 months and treated with unilateral mastectomy were eligible for the trial, and 326 were assigned randomly. After a median follow-up of 5 years, fewer breast cancers (invasive and DCIS) occurred in the anastrazole-treated group than in the placebo group (HR, 0.47; 95% CI, 0.32–0.68). The risk of hormone receptor–positive, but not hormone receptor–negative, breast cancer was reduced. Based on predicted cumulative incidence over 7 years, the number of high-risk women (per the IBIS-II eligibility criteria) needed-to-treat for 5 years to prevent one breast cancer in 7 years of follow-up was estimated to be 36 (95% CI, 33–44). Women treated with anastrazole were more likely than those taking placebo to have musculoskeletal symptoms, including arthralgias (51% vs. 46%), joint stiffness (7% vs. 5%), pain in hand or foot (9% vs. 8%) , and carpal tunnel syndrome (3% vs. 2%); hypertension (5% vs. 3%); vasomotor symptoms (57% vs. 49%); and dry eyes (4% vs. 2%). The association between hand or foot pain with anastrazole treatment was of borderline statistical significance; all other side effects noted above were statistically significantly associated with anastrazole treatment.
  • Prophylactic mastectomy

    A retrospective cohort study evaluated the impact of bilateral prophylactic mastectomy on breast cancer incidence among women at high and moderate risk on the basis of family history.

    BRCA mutation status was not known. Subcutaneous, rather than total, mastectomy was performed in 90% of these women. After a median follow-up of 14 years postsurgery, the risk reduction for the 425 moderate-risk women was 89%; for the 214 high-risk women, it was 90% to 94%, depending on the method used to calculate expected rates of breast cancer. The risk reduction for breast cancer mortality was 100% for moderate-risk women and 81% for high-risk women. Because the study used family history as a risk indicator rather than genetic testing, breast cancer risk may be overestimated.

    The rate of bilateral mastectomy among women with unilateral disease (DCIS and early-stage invasive breast cancer) was reported to have increased from 1.9% in 1998 to 11.2% in 2011 based on data from the U.S. National Cancer Data Base.

    No studies have been done on the benefits of prophylactic mastectomy in the average-risk population to prevent contralateral breast cancer in women with an ipsilateral breast cancer.

    Prophylactic oophorectomy

    Ovarian ablation and oophorectomy are associated with decreased breast cancer risk in normal women and in women with increased risk resulting from thoracic irradiation. (Refer to the Endogenous estrogen section in the Description of the Evidence section of this summary for more information.) Observational studies of women with high breast cancer risk resulting from BRCA1 or BRCA2 gene mutations showed that prophylactic oophorectomy to prevent ovarian cancer was also associated with a 50% decrease in breast cancer incidence.

    These studies are confounded by selection bias, family relationships between patients and controls, indications for oophorectomy, and inadequate information about hormone use. A prospective cohort study had similar findings, with a greater breast cancer risk reduction in BRCA2 mutation carriers than in BRCA1 carriers.

    Factors and Interventions With Inadequate Evidence of an Association

    Hormonal contraceptives

    Oral contraceptives have been associated with a small increased risk of breast cancer in current users that diminishes over time.

    A well-conducted case-control study did not observe an association between breast cancer risk and oral contraceptive use for every use, duration of use, or recency of use.

    Another case-control study found no increased risk of breast cancer associated with the use of injectable or implantable progestin-only contraceptives in women aged 35 to 64 years.

    A nationwide prospective cohort study in Denmark found that women who currently or recently used contemporary hormonal contraceptives had a higher risk of breast cancer than did women who had never used hormonal contraceptives. Moreover, the risk of breast cancer increased with longer duration of hormonal contraceptive use. However, in absolute terms, the effect of oral contraceptives on breast cancer risk was very small. Thus, approximately one extra case of breast cancer would be expected for every 7,690 women using hormonal contraception for 1 year.

    Environmental factors

    Occupational, environmental, or chemical exposures have all been proposed as causes of breast cancer. Meta-analyses, describing up to 134 environmental chemicals, their sources, and biomarkers of their exposures, suggest that they may be associated with cancer.

    Some studies suggest that organochlorine exposures, such as those associated with insecticides, might be associated with an increase in breast cancer risk,

    but other case-control and nested case-control studies do not.

    Studies reporting positive associations have been inconsistent in the identification of responsible organochlorines. Some of these substances have weak estrogenic effects, but their effect on breast cancer risk remains unproven. The use of dichloro-diphenyl-trichloroethane was banned in the United States in 1972, and the production of polychlorinated biphenyls was stopped in 1977. Overall, the epidemiological and animal study evidence that support an association between breast cancer and specific environmental exposures is generally weak. Because so many factors must be considered, any associations with breast cancer or other cancers could be confounded by the analytical problems of multiplicities, measurement challenges, and recall and publication bias.

    Factors and Interventions With Adequate Evidence of Little or No Association

    Abortion

    Abortion has been proposed as a risk factor for breast cancer. Findings from observational studies have varied; some studies showed an association, while other studies did not. Observational studies that support this association were less rigorous and potentially biased because of differential recall by women on a socially sensitive issue.

    For example, the impact of recall or reporting bias was demonstrated in a study that compared regions with different social attitudes on abortion.

    The Committee on Gynecologic Practice of the American College of Obstetricians and Gynecologists has concluded that “more rigorous recent studies demonstrate no causal relationship between induced abortion and a subsequent increase in breast cancer risk.”

    Studies that used prospectively recorded data regarding abortion, thereby avoiding recall bias, largely showed no association with the subsequent development of breast cancer.

    Diet

    Any effect of dietary modifications on breast cancer would likely depend on the type of modification. However, there is little evidence that dietary modifications of any kind have an impact on the incidence of breast cancer.

    There are very few randomized trials in humans comparing cancer incidence for different diets. Most studies are observational—including post hoc analyses of randomized trials—and are subject to biases that may be so large as to render the observation difficult to interpret. In particular, p-values and CIs do not have the same interpretation as when calculated for the primary endpoint in a randomized trial.

    A summary of ecological studies published before 1975 showed a positive correlation between international age-adjusted breast cancer mortality rates and the estimated per capita consumption of dietary fat.

    Results of case-control studies have been mixed. Twenty years later, a pooled analysis of results from seven cohort studies found no association between total dietary fat intake and breast cancer risk.

    A randomized, controlled, dietary modification study was undertaken among 48,835 postmenopausal women aged 50 to 79 years who were also enrolled in the WHI. The intervention promoted a goal of reducing total fat intake by 20% by increasing vegetable, fruit, and grain consumption. The intervention group reduced fat intake by approximately 10% for more than 8.1 years of follow-up, resulting in lower estradiol and gamma-tocopherol levels, but no persistent weight loss. The incidence of invasive breast cancer was numerically, but not statistically lower in the intervention group, with an HR of 0.91 (95% CI, 0.83–1.01).

    There was no difference in all-cause mortality, overall mortality, or the incidence of cardiovascular events.

    With regard to fruit and vegetable intake, a pooled analysis of eight cohort studies including more than 350,000 women with 7,377 incident breast cancers showed little or no association for various assumed statistical models.

    The Women's Healthy Eating and Living Randomized Trial

    examined the effect of diet on the incidence of new primary breast cancers in women previously diagnosed with breast cancer. More than 3,000 women were enrolled and randomly assigned to an intense regimen of increased fruit and vegetable intake, increased fiber intake, and decreased fat intake, or a comparison group receiving printed materials on the “5-A-Day” dietary guidelines. After a mean of 7.3 years of follow-up, there was no reduction in new primary cancers, no difference in disease-free survival, and no difference in overall survival.

    A randomized trial in Spain

    assigned participants who were at high cardiovascular risk to one of three diets: a Mediterranean diet supplemented with extra-virgin olive oil, a Mediterranean diet supplemented with mixed nuts, or a control Mediterranean diet (counseling to reduce dietary fat). The investigators reported a statistically significant reduction in major cardiovascular events, which was the trial’s primary endpoint.

    The investigators also addressed other endpoints, including the incidence of breast cancer, although it is not specified how many were examined. Based on only 35 cases of invasive breast cancer (as compared with 288 major cardiovascular events), the respective rates of breast cancer were 8 of 1,476 (0.54%); 10 of 1,285 (0.78%); and 17 of 1,391 (1.22%) with respective average follow-up durations of 4.8, 4.3, and 4.2 years. The circumstances of the study make it difficult to determine the statistical significance of these differences.

    Vitamins

    The potential role of specific micronutrients for breast cancer risk reduction has been examined in clinical trials, with cardiovascular disease and cancer as outcomes. The Women’s Health Study, a randomized trial with 39,876 women, found no difference in breast cancer incidence at 2 years between women assigned to take either beta carotene or placebo.

    In this same study, no overall effect on cancer was seen in women taking 600 IU of vitamin E every other day.

    The Women’s Antioxidant Cardiovascular Study examined 8,171 women for incidence of total cancer and invasive breast cancer and found no effect for vitamin C, vitamin E, or beta carotene.

    Two years later, a subset of 5,442 women were randomly assigned to take 1.5 mg of folic acid, 50 mg of vitamin B6, and 1 mg of vitamin B12, or placebo. After 7.3 years, there was no difference in the incidence of total invasive cancer or invasive breast cancer.

    Fenretinide

    is a vitamin A analog that has been shown to reduce breast carcinogenesis in preclinical studies. A phase III Italian trial compared the efficacy of a 5-year intervention with fenretinide versus no treatment in 2,972 women, aged 30 to 70 years, with surgically removed stage I breast cancer or DCIS. At a median observation time of 97 months, there were no statistically significant differences in the occurrence of contralateral breast cancer (P = .642), ipsilateral breast cancer (P = .177), incidence of distant metastases, nonbreast malignancies, and all-cause mortality.

    Active and passive cigarette smoking

    The potential role of active cigarette smoking in the etiology of breast cancer has been studied for more than three decades, with no clear-cut evidence of an association.

    Since the mid-1990s, studies of cigarette smoking and breast cancer have more carefully accounted for secondhand smoke exposure.

    A recent meta-analysis suggests that there is no overall association between passive smoking and breast cancer and that study methodology (ascertainment of exposure after breast cancer diagnosis) may be responsible for the apparent risk associations seen in some studies.

    Underarm deodorants/antiperspirants

    Despite warnings to women in lay publications that underarm deodorants and antiperspirants cause breast cancer, there is no evidence to support these concerns. A study based on interviews with 813 women who had breast cancer and 793 controls found no association between the risk of breast cancer and the use of antiperspirants, the use of deodorants, or the use of blade razors before these products were applied.

    In contrast, a study of 437 breast cancer survivors found that women who used antiperspirants/deodorants and shaved their underarms more frequently had cancer diagnosed at a significantly younger age. A possible explanation for this finding is that these women had an earlier menarche or higher levels of endogenous hormones, both known to be risk factors for breast cancer and to increase body hair.

    Statins

    Two well-conducted meta-analyses of RCTs

    and RCTs plus observational studies

    found no evidence that statin use either increases or decreases the risk of breast cancer.

    Bisphosphonates

    Oral and intravenous bisphosphonates for the treatment of hypercalcemia and osteoporosis have been studied for a possible beneficial effect on breast cancer prevention. Initial observational studies suggested that women who used these drugs for durations of approximately 1 to 4 years had a lower incidence of breast cancer.

    These findings are confounded by the fact that women with osteoporosis have lower breast cancer risk than those with normal bone density. Additional evidence came from studies of women with a breast cancer diagnosis; the use of these drugs was associated with fewer new contralateral cancers.

    With this background, two large randomized placebo-controlled trials were done. The Fracture Intervention Trial (FIT) treated 6,194 postmenopausal osteopenic women with either alendronate or placebo and found no difference at 3.8 years in breast cancer incidence, with incidence of 1.8% and 1.5%, respectively (HR, 1.24; CI, 0.84–1.83). The Health Outcomes and Reduced Incidence With Zoledronic Acid Once Yearly-Pivotal Fracture Trial (HORIZON-PRT) examined 7,580 postmenopausal osteoporotic women with either intravenous zoledronate or placebo and found no difference at 2.8 years in breast cancer incidence, with incidence of 0.8% and 0.9%, respectively (HR, 1.15; CI, 0.7–1.89).

    Working night shifts

    In 2007, the World Health Organization’s International Agency for Research on Cancer (IARC) classified shift work that involves circadian disruption as a probable breast carcinogen. The principal evidence was from animal studies. There was limited evidence from human studies at the time.

    In 2013, a meta-analysis of 15 epidemiologic studies concluded that there was weak evidence of an increased incidence of breast cancer among women who had ever worked night shifts.

    In 2016, the results from three recent prospective studies from the United Kingdom, involving nearly 800,000 women, were combined with results from seven other prospective studies and showed no evidence of any association between breast cancer incidence and night shift work. In particular, the confidence intervals for the incidence rate ratios were narrow, even for 20 years or more of night shift work (rate ratio, 1.01; 95% CI, 0.93–1.10). These results exclude a moderate association of breast cancer incidence with long duration of night shift work.

    The U.K. Generations Study was established in 2003 to address risk factors and causes of breast cancer. In a prospective cohort of 105,000 women, information was obtained by questionnaire on bedroom light levels at night at the time of study recruitment and at age 20 years. They followed women for an average of 6.1 years and observed 1,775 breast cancers. Adjusting for potentially confounding factors, including night shift work, they found no evidence that the amount of bedroom light at night was associated with breast cancer risk. For the highest-to-lowest levels of light at night, the HR of breast cancer incidence was 1.01 (95% CI, 0.88–1.15).

    ReferenceSection

  • American Cancer Society: Cancer Facts and Figures 2019. Atlanta, Ga: American Cancer Society, 2019. Available online. Last accessed December 12, 2019.
  • Howlader N, Noone AM, Krapcho M, et al., eds.: SEER Cancer Statistics Review, 1975-2012. Bethesda, Md: National Cancer Institute, 2015. Also available online. Last accessed February 20, 2020.
  • Altekruse SF, Kosary CL, Krapcho M, et al.: SEER Cancer Statistics Review, 1975-2007. Bethesda, Md: National Cancer Institute, 2010. Also available online. Last accessed December 12, 2019.
  • American Cancer Society: Cancer Facts and Figures 2018. Atlanta, Ga: American Cancer Society, 2018. Available online. Last accessed February 20, 2020.
  • Pfeiffer RM, Mitani A, Matsuno RK, et al.: Racial differences in breast cancer trends in the United States (2000-2004). J Natl Cancer Inst 100 (10): 751-2, 2008.
  • Boone CW, Kelloff GJ, Freedman LS: Intraepithelial and postinvasive neoplasia as a stochastic continuum of clonal evolution, and its relationship to mechanisms of chemopreventive drug action. J Cell Biochem Suppl 17G: 14-25, 1993.
  • Kelloff GJ, Boone CW, Steele VE, et al.: Progress in cancer chemoprevention: perspectives on agent selection and short-term clinical intervention trials. Cancer Res 54 (7 Suppl): 2015s-2024s, 1994.
  • Knabbe C, Lippman ME, Wakefield LM, et al.: Evidence that transforming growth factor-beta is a hormonally regulated negative growth factor in human breast cancer cells. Cell 48 (3): 417-28, 1987.
  • Parkin DM: Cancers of the breast, endometrium and ovary: geographic correlations. Eur J Cancer Clin Oncol 25 (12): 1917-25, 1989.
  • Dunn JE: Breast cancer among American Japanese in the San Francisco Bay area. Natl Cancer Inst Monogr 47: 157-60, 1977.
  • Kliewer EV, Smith KR: Breast cancer mortality among immigrants in Australia and Canada. J Natl Cancer Inst 87 (15): 1154-61, 1995.
  • Brinton LA, Schairer C, Hoover RN, et al.: Menstrual factors and risk of breast cancer. Cancer Invest 6 (3): 245-54, 1988.
  • Collaborative Group on Hormonal Factors in Breast Cancer: Menarche, menopause, and breast cancer risk: individual participant meta-analysis, including 118 964 women with breast cancer from 117 epidemiological studies. Lancet Oncol 13 (11): 1141-51, 2012.
  • Ritte R, Lukanova A, Tjønneland A, et al.: Height, age at menarche and risk of hormone receptor-positive and -negative breast cancer: a cohort study. Int J Cancer 132 (11): 2619-29, 2013.
  • Endogenous Hormones and Breast Cancer Collaborative Group: Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. J Natl Cancer Inst 94 (8): 606-16, 2002.
  • Key TJ, Appleby PN, Reeves GK, et al.: Circulating sex hormones and breast cancer risk factors in postmenopausal women: reanalysis of 13 studies. Br J Cancer 105 (5): 709-22, 2011.
  • Kaaks R, Rinaldi S, Key TJ, et al.: Postmenopausal serum androgens, oestrogens and breast cancer risk: the European prospective investigation into cancer and nutrition. Endocr Relat Cancer 12 (4): 1071-82, 2005.
  • Kaaks R, Berrino F, Key T, et al.: Serum sex steroids in premenopausal women and breast cancer risk within the European Prospective Investigation into Cancer and Nutrition (EPIC). J Natl Cancer Inst 97 (10): 755-65, 2005.
  • Smith PG, Doll R: Late effects of x irradiation in patients treated for metropathia haemorrhagica. Br J Radiol 49 (579): 224-32, 1976.
  • Trichopoulos D, MacMahon B, Cole P: Menopause and breast cancer risk. J Natl Cancer Inst 48 (3): 605-13, 1972.
  • Feinleib M: Breast cancer and artificial menopause: a cohort study. J Natl Cancer Inst 41 (2): 315-29, 1968.
  • Kampert JB, Whittemore AS, Paffenbarger RS: Combined effect of childbearing, menstrual events, and body size on age-specific breast cancer risk. Am J Epidemiol 128 (5): 962-79, 1988.
  • Hirayama T, Wynder EL: A study of the epidemiology of cancer of the breast. II. The influence of hysterectomy. Cancer 15: 28-38, 1962 Jan-Feb.
  • Colditz GA, Kaphingst KA, Hankinson SE, et al.: Family history and risk of breast cancer: nurses' health study. Breast Cancer Res Treat 133 (3): 1097-104, 2012.
  • Miki Y, Swensen J, Shattuck-Eidens D, et al.: A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 266 (5182): 66-71, 1994.
  • Futreal PA, Liu Q, Shattuck-Eidens D, et al.: BRCA1 mutations in primary breast and ovarian carcinomas. Science 266 (5182): 120-2, 1994.
  • Wooster R, Neuhausen SL, Mangion J, et al.: Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science 265 (5181): 2088-90, 1994.
  • Kuchenbaecker KB, Hopper JL, Barnes DR, et al.: Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 317 (23): 2402-2416, 2017.
  • Antoniou A, Pharoah PD, Narod S, et al.: Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet 72 (5): 1117-30, 2003.
  • Chen S, Parmigiani G: Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol 25 (11): 1329-33, 2007.
  • National Cancer Institute: SEER Stat Fact Sheets: Female Breast Cancer. Bethesda, MD: National Cancer Institute. Available online. Last accessed September 26, 2019.
  • Swift M, Morrell D, Massey RB, et al.: Incidence of cancer in 161 families affected by ataxia-telangiectasia. N Engl J Med 325 (26): 1831-6, 1991.
  • Cybulski C, Wokołorczyk D, Jakubowska A, et al.: Risk of breast cancer in women with a CHEK2 mutation with and without a family history of breast cancer. J Clin Oncol 29 (28): 3747-52, 2011.
  • Boyd NF, Guo H, Martin LJ, et al.: Mammographic density and the risk and detection of breast cancer. N Engl J Med 356 (3): 227-36, 2007.
  • Razzaghi H, Troester MA, Gierach GL, et al.: Mammographic density and breast cancer risk in White and African American Women. Breast Cancer Res Treat 135 (2): 571-80, 2012.
  • McCormack VA, dos Santos Silva I: Breast density and parenchymal patterns as markers of breast cancer risk: a meta-analysis. Cancer Epidemiol Biomarkers Prev 15 (6): 1159-69, 2006.
  • Gierach GL, Ichikawa L, Kerlikowske K, et al.: Relationship between mammographic density and breast cancer death in the Breast Cancer Surveillance Consortium. J Natl Cancer Inst 104 (16): 1218-27, 2012.
  • Breast cancer and hormone replacement therapy: collaborative reanalysis of data from 51 epidemiological studies of 52,705 women with breast cancer and 108,411 women without breast cancer. Collaborative Group on Hormonal Factors in Breast Cancer. Lancet 350 (9084): 1047-59, 1997.
  • Hulley S, Furberg C, Barrett-Connor E, et al.: Noncardiovascular disease outcomes during 6.8 years of hormone therapy: Heart and Estrogen/progestin Replacement Study follow-up (HERS II). JAMA 288 (1): 58-66, 2002.
  • Writing Group for the Women's Health Initiative Investigators: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA 288 (3): 321-33, 2002.
  • Chlebowski RT, Anderson GL, Gass M, et al.: Estrogen plus progestin and breast cancer incidence and mortality in postmenopausal women. JAMA 304 (15): 1684-92, 2010.
  • Chlebowski RT, Hendrix SL, Langer RD, et al.: Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women's Health Initiative Randomized Trial. JAMA 289 (24): 3243-53, 2003.
  • Chlebowski RT, Manson JE, Anderson GL, et al.: Estrogen plus progestin and breast cancer incidence and mortality in the Women's Health Initiative Observational Study. J Natl Cancer Inst 105 (8): 526-35, 2013.
  • Anderson GL, Limacher M, Assaf AR, et al.: Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA 291 (14): 1701-12, 2004.
  • LaCroix AZ, Chlebowski RT, Manson JE, et al.: Health outcomes after stopping conjugated equine estrogens among postmenopausal women with prior hysterectomy: a randomized controlled trial. JAMA 305 (13): 1305-14, 2011.
  • Anderson GL, Chlebowski RT, Aragaki AK, et al.: Conjugated equine oestrogen and breast cancer incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women's Health Initiative randomised placebo-controlled trial. Lancet Oncol 13 (5): 476-86, 2012.
  • Schierbeck LL, Rejnmark L, Tofteng CL, et al.: Effect of hormone replacement therapy on cardiovascular events in recently postmenopausal women: randomised trial. BMJ 345: e6409, 2012.
  • Beral V, Reeves G, Bull D, et al.: Breast cancer risk in relation to the interval between menopause and starting hormone therapy. J Natl Cancer Inst 103 (4): 296-305, 2011.
  • Li CI, Malone KE, Porter PL, et al.: Relationship between long durations and different regimens of hormone therapy and risk of breast cancer. JAMA 289 (24): 3254-63, 2003.
  • Chlebowski RT, Anderson GL: The influence of time from menopause and mammography on hormone therapy-related breast cancer risk assessment. J Natl Cancer Inst 103 (4): 284-5, 2011.
  • Prentice RL, Chlebowski RT, Stefanick ML, et al.: Conjugated equine estrogens and breast cancer risk in the Women's Health Initiative clinical trial and observational study. Am J Epidemiol 167 (12): 1407-15, 2008.
  • Chlebowski RT, Kuller LH, Prentice RL, et al.: Breast cancer after use of estrogen plus progestin in postmenopausal women. N Engl J Med 360 (6): 573-87, 2009.
  • Cronin KA, Ravdin PM, Edwards BK: Sustained lower rates of breast cancer in the United States. Breast Cancer Res Treat 117 (1): 223-4, 2009.
  • Ravdin PM, Cronin KA, Howlader N, et al.: The decrease in breast-cancer incidence in 2003 in the United States. N Engl J Med 356 (16): 1670-4, 2007.
  • Parkin DM: Is the recent fall in incidence of post-menopausal breast cancer in UK related to changes in use of hormone replacement therapy? Eur J Cancer 45 (9): 1649-53, 2009.
  • Lambe M, Wigertz A, Holmqvist M, et al.: Reductions in use of hormone replacement therapy: effects on Swedish breast cancer incidence trends only seen after several years. Breast Cancer Res Treat 121 (3): 679-83, 2010.
  • Renard F, Vankrunkelsven P, Van Eycken L, et al.: Decline in breast cancer incidence in the Flemish region of Belgium after a decline in hormonal replacement therapy. Ann Oncol 21 (12): 2356-60, 2010.
  • Farhat GN, Walker R, Buist DS, et al.: Changes in invasive breast cancer and ductal carcinoma in situ rates in relation to the decline in hormone therapy use. J Clin Oncol 28 (35): 5140-6, 2010.
  • DeSantis C, Howlader N, Cronin KA, et al.: Breast cancer incidence rates in U.S. women are no longer declining. Cancer Epidemiol Biomarkers Prev 20 (5): 733-9, 2011.
  • John EM, Kelsey JL: Radiation and other environmental exposures and breast cancer. Epidemiol Rev 15 (1): 157-62, 1993.
  • Evans JS, Wennberg JE, McNeil BJ: The influence of diagnostic radiography on the incidence of breast cancer and leukemia. N Engl J Med 315 (13): 810-5, 1986.
  • Andrieu N, Easton DF, Chang-Claude J, et al.: Effect of chest X-rays on the risk of breast cancer among BRCA1/2 mutation carriers in the international BRCA1/2 carrier cohort study: a report from the EMBRACE, GENEPSO, GEO-HEBON, and IBCCS Collaborators' Group. J Clin Oncol 24 (21): 3361-6, 2006.
  • Bhatia S, Robison LL, Oberlin O, et al.: Breast cancer and other second neoplasms after childhood Hodgkin's disease. N Engl J Med 334 (12): 745-51, 1996.
  • Hancock SL, Tucker MA, Hoppe RT: Breast cancer after treatment of Hodgkin's disease. J Natl Cancer Inst 85 (1): 25-31, 1993.
  • Travis LB, Hill DA, Dores GM, et al.: Breast cancer following radiotherapy and chemotherapy among young women with Hodgkin disease. JAMA 290 (4): 465-75, 2003.
  • Sankila R, Garwicz S, Olsen JH, et al.: Risk of subsequent malignant neoplasms among 1,641 Hodgkin's disease patients diagnosed in childhood and adolescence: a population-based cohort study in the five Nordic countries. Association of the Nordic Cancer Registries and the Nordic Society of Pediatric Hematology and Oncology. J Clin Oncol 14 (5): 1442-6, 1996.
  • van Leeuwen FE, Klokman WJ, Stovall M, et al.: Roles of radiation dose, chemotherapy, and hormonal factors in breast cancer following Hodgkin's disease. J Natl Cancer Inst 95 (13): 971-80, 2003.
  • Obedian E, Fischer DB, Haffty BG: Second malignancies after treatment of early-stage breast cancer: lumpectomy and radiation therapy versus mastectomy. J Clin Oncol 18 (12): 2406-12, 2000.
  • Fisher B, Anderson S, Bryant J, et al.: Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347 (16): 1233-41, 2002.
  • Veronesi U, Cascinelli N, Mariani L, et al.: Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347 (16): 1227-32, 2002.
  • 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 347 (8): 567-75, 2002.
  • Morimoto LM, White E, Chen Z, et al.: Obesity, body size, and risk of postmenopausal breast cancer: the Women's Health Initiative (United States). Cancer Causes Control 13 (8): 741-51, 2002.
  • Wolin KY, Carson K, Colditz GA: Obesity and cancer. Oncologist 15 (6): 556-65, 2010.
  • Lawlor DA, Smith GD, Ebrahim S: Hyperinsulinaemia and increased risk of breast cancer: findings from the British Women's Heart and Health Study. Cancer Causes Control 15 (3): 267-75, 2004.
  • Hamajima N, Hirose K, Tajima K, et al.: Alcohol, tobacco and breast cancer--collaborative reanalysis of individual data from 53 epidemiological studies, including 58,515 women with breast cancer and 95,067 women without the disease. Br J Cancer 87 (11): 1234-45, 2002.
  • Pike MC, Krailo MD, Henderson BE, et al.: 'Hormonal' risk factors, 'breast tissue age' and the age-incidence of breast cancer. Nature 303 (5920): 767-70, 1983.
  • Lambe M, Hsieh C, Trichopoulos D, et al.: Transient increase in the risk of breast cancer after giving birth. N Engl J Med 331 (1): 5-9, 1994.
  • Henderson BE, Pike MC, Ross RK, et al.: Epidemiology and risk factors. In: Bonadonna G, ed.: Breast Cancer: Diagnosis and Management. Chichester, NY: John Wiley & Sons, 1984, pp 15-33.
  • Gail MH, Brinton LA, Byar DP, et al.: Projecting individualized probabilities of developing breast cancer for white females who are being examined annually. J Natl Cancer Inst 81 (24): 1879-86, 1989.
  • Nichols HB, Schoemaker MJ, Cai J, et al.: Breast Cancer Risk After Recent Childbirth: A Pooled Analysis of 15 Prospective Studies. Ann Intern Med 170 (1): 22-30, 2019.
  • Col: Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet 360 (9328): 187-95, 2002.
  • Furberg H, Newman B, Moorman P, et al.: Lactation and breast cancer risk. Int J Epidemiol 28 (3): 396-402, 1999.
  • Bernstein L, Henderson BE, Hanisch R, et al.: Physical exercise and reduced risk of breast cancer in young women. J Natl Cancer Inst 86 (18): 1403-8, 1994.
  • Friedenreich CM: Physical activity and cancer prevention: from observational to intervention research. Cancer Epidemiol Biomarkers Prev 10 (4): 287-301, 2001.
  • Thune I, Brenn T, Lund E, et al.: Physical activity and the risk of breast cancer. N Engl J Med 336 (18): 1269-75, 1997.
  • Adams-Campbell LL, Rosenberg L, Rao RS, et al.: Strenuous physical activity and breast cancer risk in African-American women. J Natl Med Assoc 93 (7-8): 267-75, 2001 Jul-Aug.
  • Nayfield SG, Karp JE, Ford LG, et al.: Potential role of tamoxifen in prevention of breast cancer. J Natl Cancer Inst 83 (20): 1450-9, 1991.
  • Love RR, Barden HS, Mazess RB, et al.: Effect of tamoxifen on lumbar spine bone mineral density in postmenopausal women after 5 years. Arch Intern Med 154 (22): 2585-8, 1994.
  • 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 14 (1): 78-84, 1996.
  • Costantino JP, Kuller LH, Ives DG, et al.: Coronary heart disease mortality and adjuvant tamoxifen therapy. J Natl Cancer Inst 89 (11): 776-82, 1997.
  • McDonald CC, Stewart HJ: Fatal myocardial infarction in the Scottish adjuvant tamoxifen trial. The Scottish Breast Cancer Committee. BMJ 303 (6800): 435-7, 1991.
  • Rutqvist LE, Mattsson A: Cardiac and thromboembolic morbidity among postmenopausal women with early-stage breast cancer in a randomized trial of adjuvant tamoxifen. The Stockholm Breast Cancer Study Group. J Natl Cancer Inst 85 (17): 1398-406, 1993.
  • Fisher B, Costantino JP, Redmond CK, et al.: Endometrial cancer in tamoxifen-treated breast cancer patients: findings from the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-14. J Natl Cancer Inst 86 (7): 527-37, 1994.
  • Bergman L, Beelen ML, Gallee MP, et al.: Risk and prognosis of endometrial cancer after tamoxifen for breast cancer. Comprehensive Cancer Centres' ALERT Group. Assessment of Liver and Endometrial cancer Risk following Tamoxifen. Lancet 356 (9233): 881-7, 2000.
  • Cuzick J, Powles T, Veronesi U, et al.: Overview of the main outcomes in breast-cancer prevention trials. Lancet 361 (9354): 296-300, 2003.
  • Redmond CK, Wickerham DL, Cronin W, et al.: The NSABP breast cancer prevention trial (BCPT): a progress report. [Abstract] Proceedings of the American Society of Clinical Oncology 12: A-78, 69, 1993.
  • Fisher B, Costantino JP, Wickerham DL, et al.: Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 90 (18): 1371-88, 1998.
  • Fisher B, Costantino JP, Wickerham DL, et al.: Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst 97 (22): 1652-62, 2005.
  • Powles T, Eeles R, Ashley S, et al.: Interim analysis of the incidence of breast cancer in the Royal Marsden Hospital tamoxifen randomised chemoprevention trial. Lancet 352 (9122): 98-101, 1998.
  • Veronesi U, Maisonneuve P, Costa A, et al.: Prevention of breast cancer with tamoxifen: preliminary findings from the Italian randomised trial among hysterectomised women. Italian Tamoxifen Prevention Study. Lancet 352 (9122): 93-7, 1998.
  • Cuzick J, Forbes J, Edwards R, et al.: First results from the International Breast Cancer Intervention Study (IBIS-I): a randomised prevention trial. Lancet 360 (9336): 817-24, 2002.
  • Powles TJ, Ashley S, Tidy A, et al.: Twenty-year follow-up of the Royal Marsden randomized, double-blinded tamoxifen breast cancer prevention trial. J Natl Cancer Inst 99 (4): 283-90, 2007.
  • Veronesi U, Maisonneuve P, Rotmensz N, et al.: Tamoxifen for the prevention of breast cancer: late results of the Italian Randomized Tamoxifen Prevention Trial among women with hysterectomy. J Natl Cancer Inst 99 (9): 727-37, 2007.
  • Martino S, Costantino J, McNabb M, et al.: The role of selective estrogen receptor modulators in the prevention of breast cancer: comparison of the clinical trials. Oncologist 9 (2): 116-25, 2004.
  • Cuzick J, Forbes JF, Sestak I, et al.: Long-term results of tamoxifen prophylaxis for breast cancer--96-month follow-up of the randomized IBIS-I trial. J Natl Cancer Inst 99 (4): 272-82, 2007.
  • 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 353 (9169): 1993-2000, 1999.
  • Khovidhunkit W, Shoback DM: Clinical effects of raloxifene hydrochloride in women. Ann Intern Med 130 (5): 431-9, 1999.
  • Cauley JA, Norton L, Lippman ME, et al.: Continued breast cancer risk reduction in postmenopausal women treated with raloxifene: 4-year results from the MORE trial. Multiple outcomes of raloxifene evaluation. Breast Cancer Res Treat 65 (2): 125-34, 2001.
  • Cummings SR, Eckert S, Krueger KA, et al.: The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation. JAMA 281 (23): 2189-97, 1999.
  • Martino S, Cauley JA, Barrett-Connor E, et al.: Continuing outcomes relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst 96 (23): 1751-61, 2004.
  • Grady D, Cauley JA, Geiger MJ, et al.: Reduced incidence of invasive breast cancer with raloxifene among women at increased coronary risk. J Natl Cancer Inst 100 (12): 854-61, 2008.
  • Vogel VG, Costantino JP, Wickerham DL, et al.: Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 295 (23): 2727-41, 2006.
  • Land SR, Wickerham DL, Costantino JP, et al.: Patient-reported symptoms and quality of life during treatment with tamoxifen or raloxifene for breast cancer prevention: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 295 (23): 2742-51, 2006.
  • The ATAC Trialists' Group. Arimidex, tamoxifen alone or in combination: 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 359 (9324): 2131-9, 2002.
  • 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 349 (19): 1793-802, 2003.
  • Goss PE, Ingle JN, Pritchard KI, et al.: Extending Aromatase-Inhibitor Adjuvant Therapy to 10 Years. N Engl J Med 375 (3): 209-19, 2016.
  • 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 350 (11): 1081-92, 2004.
  • Goss PE, Ingle JN, Alés-Martínez JE, et al.: Exemestane for breast-cancer prevention in postmenopausal women. N Engl J Med 364 (25): 2381-91, 2011.
  • Cuzick J, Sestak I, Forbes JF, et al.: Anastrozole for prevention of breast cancer in high-risk postmenopausal women (IBIS-II): an international, double-blind, randomised placebo-controlled trial. Lancet 383 (9922): 1041-8, 2014.
  • Hartmann LC, Schaid DJ, Woods JE, et al.: Efficacy of bilateral prophylactic mastectomy in women with a family history of breast cancer. N Engl J Med 340 (2): 77-84, 1999.
  • Kummerow KL, Du L, Penson DF, et al.: Nationwide trends in mastectomy for early-stage breast cancer. JAMA Surg 150 (1): 9-16, 2015.
  • Rebbeck TR, Levin AM, Eisen A, et al.: Breast cancer risk after bilateral prophylactic oophorectomy in BRCA1 mutation carriers. J Natl Cancer Inst 91 (17): 1475-9, 1999.
  • Kauff ND, Satagopan JM, Robson ME, et al.: Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 346 (21): 1609-15, 2002.
  • Rebbeck TR, Lynch HT, Neuhausen SL, et al.: Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N Engl J Med 346 (21): 1616-22, 2002.
  • Kauff ND, Domchek SM, Friebel TM, et al.: Risk-reducing salpingo-oophorectomy for the prevention of BRCA1- and BRCA2-associated breast and gynecologic cancer: a multicenter, prospective study. J Clin Oncol 26 (8): 1331-7, 2008.
  • Breast cancer and hormonal contraceptives: further results. Collaborative Group on Hormonal Factors in Breast Cancer. Contraception 54 (3 Suppl): 1S-106S, 1996.
  • Marchbanks PA, McDonald JA, Wilson HG, et al.: Oral contraceptives and the risk of breast cancer. N Engl J Med 346 (26): 2025-32, 2002.
  • Strom BL, Berlin JA, Weber AL, et al.: Absence of an effect of injectable and implantable progestin-only contraceptives on subsequent risk of breast cancer. Contraception 69 (5): 353-60, 2004.
  • Mørch LS, Skovlund CW, Hannaford PC, et al.: Contemporary Hormonal Contraception and the Risk of Breast Cancer. N Engl J Med 377 (23): 2228-2239, 2017.
  • Brody JG, Rudel RA, Michels KB, et al.: Environmental pollutants, diet, physical activity, body size, and breast cancer: where do we stand in research to identify opportunities for prevention? Cancer 109 (12 Suppl): 2627-34, 2007.
  • Rodgers KM, Udesky JO, Rudel RA, et al.: Environmental chemicals and breast cancer: An updated review of epidemiological literature informed by biological mechanisms. Environ Res 160: 152-182, 2018.
  • Wolff MS, Toniolo PG, Lee EW, et al.: Blood levels of organochlorine residues and risk of breast cancer. J Natl Cancer Inst 85 (8): 648-52, 1993.
  • Høyer AP, Grandjean P, Jørgensen T, et al.: Organochlorine exposure and risk of breast cancer. Lancet 352 (9143): 1816-20, 1998.
  • Shames LS, Munekata MT, Pike MC: Re: Blood levels of organochlorine residues and risk of breast cancer. J Natl Cancer Inst 86 (21): 1642-3, 1994.
  • Krieger N, Wolff MS, Hiatt RA, et al.: Breast cancer and serum organochlorines: a prospective study among white, black, and Asian women. J Natl Cancer Inst 86 (8): 589-99, 1994.
  • Hunter DJ, Hankinson SE, Laden F, et al.: Plasma organochlorine levels and the risk of breast cancer. N Engl J Med 337 (18): 1253-8, 1997.
  • Laden F, Collman G, Iwamoto K, et al.: 1,1-Dichloro-2,2-bis(p-chlorophenyl)ethylene and polychlorinated biphenyls and breast cancer: combined analysis of five U.S. studies. J Natl Cancer Inst 93 (10): 768-76, 2001.
  • Ward EM, Schulte P, Grajewski B, et al.: Serum organochlorine levels and breast cancer: a nested case-control study of Norwegian women. Cancer Epidemiol Biomarkers Prev 9 (12): 1357-67, 2000.
  • Laden F, Hankinson SE, Wolff MS, et al.: Plasma organochlorine levels and the risk of breast cancer: an extended follow-up in the Nurses' Health Study. Int J Cancer 91 (4): 568-74, 2001.
  • Berry D: Multiplicities in cancer research: ubiquitous and necessary evils. J Natl Cancer Inst 104 (15): 1124-32, 2012.
  • Dickersin K: The existence of publication bias and risk factors for its occurrence. JAMA 263 (10): 1385-9, 1990.
  • Huang Y, Zhang X, Li W, et al.: A meta-analysis of the association between induced abortion and breast cancer risk among Chinese females. Cancer Causes Control 25 (2): 227-36, 2014.
  • Sanderson M, Shu XO, Jin F, et al.: Abortion history and breast cancer risk: results from the Shanghai Breast Cancer Study. Int J Cancer 92 (6): 899-905, 2001.
  • Beral V, Bull D, Doll R, et al.: Breast cancer and abortion: collaborative reanalysis of data from 53 epidemiological studies, including 83,000 women with breast cancer from 16 countries. Lancet 363 (9414): 1007-16, 2004.
  • Melbye M, Wohlfahrt J, Olsen JH, et al.: Induced abortion and the risk of breast cancer. N Engl J Med 336 (2): 81-5, 1997.
  • Rookus MA, van Leeuwen FE: Induced abortion and risk for breast cancer: reporting (recall) bias in a Dutch case-control study. J Natl Cancer Inst 88 (23): 1759-64, 1996.
  • Committee on Gynecologic Practice: ACOG Committee Opinion No. 434: induced abortion and breast cancer risk. Obstet Gynecol 113 (6): 1417-8, 2009.
  • Henderson KD, Sullivan-Halley J, Reynolds P, et al.: Incomplete pregnancy is not associated with breast cancer risk: the California Teachers Study. Contraception 77 (6): 391-6, 2008.
  • Lash TL, Fink AK: Null association between pregnancy termination and breast cancer in a registry-based study of parous women. Int J Cancer 110 (3): 443-8, 2004.
  • Michels KB, Xue F, Colditz GA, et al.: Induced and spontaneous abortion and incidence of breast cancer among young women: a prospective cohort study. Arch Intern Med 167 (8): 814-20, 2007.
  • Wu JQ, Li YY, Ren JC, et al.: Induced abortion and breast cancer: results from a population-based case control study in China. Asian Pac J Cancer Prev 15 (8): 3635-40, 2014.
  • Braüner CM, Overvad K, Tjønneland A, et al.: Induced abortion and breast cancer among parous women: a Danish cohort study. Acta Obstet Gynecol Scand 92 (6): 700-5, 2013.
  • Guo J, Huang Y, Yang L, et al.: Association between abortion and breast cancer: an updated systematic review and meta-analysis based on prospective studies. Cancer Causes Control 26 (6): 811-9, 2015.
  • Carroll KK, Khor HT: Dietary fat in relation to tumorigenesis. Prog Biochem Pharmacol 10: 308-53, 1975.
  • Hunter DJ, Spiegelman D, Adami HO, et al.: Cohort studies of fat intake and the risk of breast cancer--a pooled analysis. N Engl J Med 334 (6): 356-61, 1996.
  • Prentice RL, Caan B, Chlebowski RT, et al.: Low-fat dietary pattern and risk of invasive breast cancer: the Women's Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 295 (6): 629-42, 2006.
  • Howard BV, Van Horn L, Hsia J, et al.: Low-fat dietary pattern and risk of cardiovascular disease: the Women's Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 295 (6): 655-66, 2006.
  • Smith-Warner SA, Spiegelman D, Yaun SS, et al.: Intake of fruits and vegetables and risk of breast cancer: a pooled analysis of cohort studies. JAMA 285 (6): 769-76, 2001.
  • Pierce JP, Natarajan L, Caan BJ, et al.: Influence of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis following treatment for breast cancer: the Women's Healthy Eating and Living (WHEL) randomized trial. JAMA 298 (3): 289-98, 2007.
  • Toledo E, Salas-Salvadó J, Donat-Vargas C, et al.: Mediterranean Diet and Invasive Breast Cancer Risk Among Women at High Cardiovascular Risk in the PREDIMED Trial: A Randomized Clinical Trial. JAMA Intern Med 175 (11): 1752-60, 2015.
  • Estruch R, Ros E, Salas-Salvadó J, et al.: Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med 368 (14): 1279-90, 2013.
  • Lee IM, Cook NR, Manson JE, et al.: Beta-carotene supplementation and incidence of cancer and cardiovascular disease: the Women's Health Study. J Natl Cancer Inst 91 (24): 2102-6, 1999.
  • Lee IM, Cook NR, Gaziano JM, et al.: Vitamin E in the primary prevention of cardiovascular disease and cancer: the Women's Health Study: a randomized controlled trial. JAMA 294 (1): 56-65, 2005.
  • Lin J, Cook NR, Albert C, et al.: Vitamins C and E and beta carotene supplementation and cancer risk: a randomized controlled trial. J Natl Cancer Inst 101 (1): 14-23, 2009.
  • Zhang SM, Cook NR, Albert CM, et al.: Effect of combined folic acid, vitamin B6, and vitamin B12 on cancer risk in women: a randomized trial. JAMA 300 (17): 2012-21, 2008.
  • Costa A, Formelli F, Chiesa F, et al.: Prospects of chemoprevention of human cancers with the synthetic retinoid fenretinide. Cancer Res 54 (7 Suppl): 2032s-2037s, 1994.
  • Veronesi U, De Palo G, Marubini E, et al.: Randomized trial of fenretinide to prevent second breast malignancy in women with early breast cancer. J Natl Cancer Inst 91 (21): 1847-56, 1999.
  • The Health Consequences of Smoking: A Report of the Surgeon General. Atlanta, Ga: U.S. Department of Health and Human Services, CDC, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health, 2004. Also available online. Last accessed February 20, 2020.
  • U.S. Department of Health and Human Services: The Health Consequences of Involuntary Exposure to Tobacco Smoke: A Report of the Surgeon General. Atlanta, Ga: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, Coordinating Center for Health Promotion, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health, 2006. Also available online. Last accessed September 26, 2019.
  • Pirie K, Beral V, Peto R, et al.: Passive smoking and breast cancer in never smokers: prospective study and meta-analysis. Int J Epidemiol 37 (5): 1069-79, 2008.
  • Mirick DK, Davis S, Thomas DB: Antiperspirant use and the risk of breast cancer. J Natl Cancer Inst 94 (20): 1578-80, 2002.
  • McGrath KG: An earlier age of breast cancer diagnosis related to more frequent use of antiperspirants/deodorants and underarm shaving. Eur J Cancer Prev 12 (6): 479-85, 2003.
  • Dale KM, Coleman CI, Henyan NN, et al.: Statins and cancer risk: a meta-analysis. JAMA 295 (1): 74-80, 2006.
  • Bonovas S, Filioussi K, Tsavaris N, et al.: Use of statins and breast cancer: a meta-analysis of seven randomized clinical trials and nine observational studies. J Clin Oncol 23 (34): 8606-12, 2005.
  • Newcomb PA, Trentham-Dietz A, Hampton JM: Bisphosphonates for osteoporosis treatment are associated with reduced breast cancer risk. Br J Cancer 102 (5): 799-802, 2010.
  • Rennert G, Pinchev M, Rennert HS: Use of bisphosphonates and risk of postmenopausal breast cancer. J Clin Oncol 28 (22): 3577-81, 2010.
  • Chlebowski RT, Chen Z, Cauley JA, et al.: Oral bisphosphonate use and breast cancer incidence in postmenopausal women. J Clin Oncol 28 (22): 3582-90, 2010.
  • Cardwell CR, Abnet CC, Veal P, et al.: Exposure to oral bisphosphonates and risk of cancer. Int J Cancer 131 (5): E717-25, 2012.
  • Monsees GM, Malone KE, Tang MT, et al.: Bisphosphonate use after estrogen receptor-positive breast cancer and risk of contralateral breast cancer. J Natl Cancer Inst 103 (23): 1752-60, 2011.
  • Hue TF, Cummings SR, Cauley JA, et al.: Effect of bisphosphonate use on risk of postmenopausal breast cancer: results from the randomized clinical trials of alendronate and zoledronic acid. JAMA Intern Med 174 (10): 1550-7, 2014.
  • Straif K, Baan R, Grosse Y, et al.: Carcinogenicity of shift-work, painting, and fire-fighting. Lancet Oncol 8 (12): 1065-1066, 2007.
  • Kamdar BB, Tergas AI, Mateen FJ, et al.: Night-shift work and risk of breast cancer: a systematic review and meta-analysis. Breast Cancer Res Treat 138 (1): 291-301, 2013.
  • Travis RC, Balkwill A, Fensom GK, et al.: Night Shift Work and Breast Cancer Incidence: Three Prospective Studies and Meta-analysis of Published Studies. J Natl Cancer Inst 108 (12): , 2016.
  • Johns LE, Jones ME, Schoemaker MJ, et al.: Domestic light at night and breast cancer risk: a prospective analysis of 105 000 UK women in the Generations Study. Br J Cancer 118 (4): 600-606, 2018.
  • 乳腺癌预防(PDQ®)

    变更总结(09/26/2019)

    本PDQ癌症信息总结经定期审核,新增可用信息时予以更新。本节介绍了截止上述日期本总结的最新更新。

    本总结已经过编辑审核。

    本总结的撰写和维护由 PDQ筛查与预防编辑委员会负责,独立于美国国家癌症研究所(NCI)单独编辑。本总结为独立的文献综述,并不代表NCI或NIH的政策声明。关于总结政策和PDQ编辑委员会的更多信息请参见 关于本PDQ总结和 PDQ NCI综合性癌症数据库页面。

    Breast Cancer Prevention (PDQ®)

    Changes to This Summary (09/26/2019)

    The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.

    Editorial changes were made to this summary.

    This summary is written and maintained by the PDQ Screening and Prevention Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® - NCI's Comprehensive Cancer Database pages.

    乳腺癌预防(PDQ®)

    About This PDQ Summary

    Purpose of This Summary

    This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about breast cancer prevention. It is intended as a resource to inform and assist clinicians who care for cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.

    Reviewers and Updates

    This summary is reviewed regularly and updated as necessary by the PDQ Screening and Prevention Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).

    Board members review recently published articles each month to determine whether an article should:

  • be discussed at a meeting,
  • be cited with text, or
  • replace or update an existing article that is already cited.
  • Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.

    Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.

    Levels of Evidence

    Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Screening and Prevention Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.

    Permission to Use This Summary

    PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”

    The preferred citation for this PDQ summary is:

    PDQ® Screening and Prevention Editorial Board. PDQ Breast Cancer Prevention. Bethesda, MD: National Cancer Institute. Updated . Available at: https://www.cancer.gov/types/breast/hp/breast-prevention-pdq. Accessed . [PMID: 26389323]

    Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.

    Disclaimer

    The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.

    Contact Us

    More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.

    Breast Cancer Prevention (PDQ®)

    About This PDQ Summary

    Purpose of This Summary

    This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about breast cancer prevention. It is intended as a resource to inform and assist clinicians who care for cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.

    Reviewers and Updates

    This summary is reviewed regularly and updated as necessary by the PDQ Screening and Prevention Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).

    Board members review recently published articles each month to determine whether an article should:

  • be discussed at a meeting,
  • be cited with text, or
  • replace or update an existing article that is already cited.
  • Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.

    Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.

    Levels of Evidence

    Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Screening and Prevention Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.

    Permission to Use This Summary

    PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”

    The preferred citation for this PDQ summary is:

    PDQ® Screening and Prevention Editorial Board. PDQ Breast Cancer Prevention. Bethesda, MD: National Cancer Institute. Updated . Available at: https://www.cancer.gov/types/breast/hp/breast-prevention-pdq. Accessed . [PMID: 26389323]

    Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.

    Disclaimer

    The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.

    Contact Us

    More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.

    < 1 2 3 4 5 >
    目录
    章 节
    哪些是危险人群? 概述 证据描述 变更总结(09/26/2019) About This PDQ Summary