这篇总结讨论了女性原发性乳腺上皮细胞癌。 乳房很少出现其他肿瘤如淋巴瘤、肉瘤或黑色素瘤的。 更多关于这些类型癌症的信息,请参阅以下PDQ总结。
乳腺癌也可以发生在男性和儿童中,并且可能出现于妊娠期,但是它很少发生于这些人群。更多信息,请参阅以下PDQ总结:
2020年美国乳腺癌(仅女性)的估计的新发病例和死亡病例:
乳腺癌是美国女性最常见的非皮肤性癌症,且2020年估计有48,530例女性乳腺导管原位癌患者和276,480例浸润性乳腺癌患者。
因此,不到六分之一的被诊断患有乳腺癌的女性死于该疾病。相比之下,据估计,2020年美国约有63,220名女性将死于肺癌。
男性占乳腺癌病例和乳腺癌死亡的1%(更多信息,请参阅有关乳腺癌筛查的PDQ总结中的“特殊人群”部分内容)。
广泛采用筛查可增加特定人群的乳腺癌发病率,并改变检测到的癌症特征,增加低风险癌症、癌前病变和导管原位癌(DCIS)的发病率。(更多信息,请参阅关于乳腺癌筛查的PDQ总结中的“乳腺组织病理评估”中的“导管原位癌(DCIS)”部分内容。) 来自美国
和英国的人群研究
表明,自20世纪70年代以来,DCIS和浸润性乳腺癌的发病率有所增加,这归因于绝经后激素治疗和乳腺X线筛查的广泛应用。 近十年中,女性绝经后激素使用受到了限制,乳腺癌的发病率有所下降,但未降到广泛应用乳房X线筛查之前的水平。
对于大多数癌症来说,年龄增长是最重要的危险因素。其他乳腺癌的危险因素包括以下:
年龄相关的风险评估有助于有乳腺癌家族史女性的筛查策略的讨论和设计。
在所有患有乳腺癌的女性中,5%到10%的可能存在BRCA1和BRCA2基因的胚系突变。
特殊的BRCA1和BRCA2突变更常见于犹太祖系的女性。
BRCA1和BRCA2突变的女性终生患乳腺癌的风险估计为40%至85%。有乳腺癌病史的突变基因携带者患对侧乳腺癌的风险增加,该风险可能高达每年5%。
男性BRCA2突变携带者患乳腺癌的风险也会增加。
BRCA1或BRCA2基因的突变也会增加卵巢癌
或其他原发性癌症的患病风险。
一旦BRCA1或BRCA2突变被确定,其他家庭成员可考虑进行基因咨询和检测。
(更多信息,请参阅有关乳腺癌和妇产科癌症遗传学、乳腺癌预防和乳腺癌筛查的PDQ总结。)
(更多有关增加乳腺癌风险的因素的信息,请参阅有关乳腺癌预防的PDQ总结。)
降低女性乳腺癌患病风险的保护性因素和干预措施包括以下方面:
(更多关于降低乳腺癌风险的因素的信息,请参阅有关乳腺癌预防的PDQ总结。)
临床试验已证实,使用乳房X线检查对无症状的女性进行筛查,无论是否进行临床乳房检查,均可降低乳腺癌的死亡率。 (更多信息,请参阅有关乳腺癌筛查的PDQ总结。)
当怀疑乳腺癌时,一般的患者管理包括以下方面:
以下检测和程序将被用于诊断乳腺癌:
病理上,乳腺癌可以是多中心性和双侧的疾病。双侧乳腺癌更常见于渗入性叶状癌患者中。在诊断后10年,对侧乳房发生原发性乳腺癌的风险在3%到10%之间,尽管内分泌治疗降低了这种风险。
对侧乳腺癌与远期复发风险的增加相关。
当BRCA1/BRCA2突变携带者在40岁前被诊断时,对侧乳腺癌的风险在随后的25年内能够达到约50%。
患有乳腺癌的患者需在诊断时进行双侧的乳房X线检查,以排除同时发生的疾病。为了监测接受保乳手术的患者的同侧乳房的复发或对侧乳房的二次原发性癌症,患者术后需继续进行定期的乳房体格检查和乳房X线检查。
MRI在对侧乳房的筛查和对接受保乳治疗的女性的监测中的作用日益显著。由于乳房X线检查可明确增加乳房隐匿病变的检出率,尽管没有随机对照试验的数据,但选择性使用MRI进行额外筛查的频率逐渐增加。由于MRI阳性病灶中只有25%的恶性肿瘤,因此建议在治疗前进行病理确认。 这种检出率的提高是否提升治疗效果还不得而知。
乳腺癌通常是通过外科手术、放射治疗、化疗和激素治疗的各种组合来治疗的。 治疗的预后和选择可能受以下临床和病理特征的影响(基于常规组织学和免疫组织化学):
乳腺癌分子分型的检测包括以下方面:
根据ER、PR和HER2/neu的检测结果,乳腺癌被归类为以下类型之一:
ER、PR和HER2的状态在确定预后和预测内分泌治疗以及HER2靶向治疗的反应非常重要。美国临床肿瘤学会/美国病理学家学会的共识小组已经发布了相关指南,以帮助那些通过免疫组化来评估ER-PR状态和通过免疫组化与原位杂交技术来评估HER2状态所使用的测试的性能、解释和报告更加标准化。
基因谱检测包括以下内容:
以下试验描述了早期乳腺癌中多基因检测的预后和预测价值:
这项研究发现低风险评分的患者在内分泌治疗5年后,复发率非常低。
在TAILORx研究中,中等复发风险组(复发评分为11-25)中,6907名女性被随机分配到单一内分泌治疗组或内分泌治疗联合化疗组。
其中,3399名单一内分泌治疗组的女性和3312名内分泌治疗联合化疗组的女性根据随机化的治疗进行分析。在中位随访时间90个月后,iDFS的差异、主要研究终点符合预先设定的非劣效标准(P>0.10,在835例事件发生后进行无差异检验),这表明与内分泌治疗联合化疗相比,内分泌治疗具有非劣效性。
来自前瞻性随机临床试验RxPONDER(NCT01272037)的结果,将有助于决定使用内分泌治疗治疗的ER阳性、淋巴结阳性的、进行内分泌治疗的和复发评分低于25的早期乳腺癌患者是否受益于辅助化疗。
许多其他基于基因的检测可能指导早期乳腺癌患者的治疗决策(例如,Microarray50预测因子分析[PAM50],复发风险[ROR]评分,EndoPredict,乳腺癌指数)。
虽然某些罕见的遗传突变,如BRCA1和BRCA2突变,易使女性患上乳腺癌,但BRCA1/BRCA2突变携带者患乳腺癌的预后数据是相互矛盾的。这些女性患对侧乳腺癌的风险更大。 (更多信息,请参阅乳腺癌和妇产癌症PDQ遗传学总结中的BRCA1和BRCA2相关的乳腺癌预后的部分内容。)
慎重考虑后,有严重症状的患者可以接受激素替代治疗。更多信息,请参阅以下PDQ总结:
其他包含与乳腺癌相关信息的PDQ总结包括以下内容:
This summary discusses primary epithelial breast cancers in women. The breast is rarely affected by other tumors such as lymphomas, sarcomas, or melanomas. Refer to the following PDQ summaries for more information on these cancer types:
Breast cancer also affects men and children and may occur during pregnancy, although it is rare in these populations. Refer to the following PDQ summaries for more information:
Estimated new cases and deaths from breast cancer (women only) in the United States in 2020:
Breast cancer is the most common noncutaneous cancer in U.S. women, with an estimated 48,530 cases of female breast ductal carcinoma in situ and 276,480 cases of invasive disease in 2020.
Thus, fewer than one of six women diagnosed with breast cancer die of the disease. By comparison, it is estimated that about 63,220 American women will die of lung cancer in 2020.
Men account for 1% of breast cancer cases and breast cancer deaths (refer to the Special Populations section in the PDQ summary on Breast Cancer Screening for more information).
Widespread adoption of screening increases breast cancer incidence in a given population and changes the characteristics of cancers detected, with increased incidence of lower-risk cancers, premalignant lesions, and ductal carcinoma in situ (DCIS). (Refer to the Ductal carcinoma in situ (DCIS) section in the Pathologic Evaluation of Breast Tissue section in the PDQ summary on Breast Cancer Screening for more information.) Population studies from the United States
and the United Kingdom
demonstrate an increase in DCIS and invasive breast cancer incidence since the 1970s, attributable to the widespread adoption of both postmenopausal hormone therapy and screening mammography. In the last decade, women have refrained from using postmenopausal hormones, and breast cancer incidence has declined, but not to the levels seen before the widespread use of screening mammography.
Increasing age is the most important risk factor for most cancers. Other risk factors for breast cancer include the following:
Age-specific risk estimates are available to help counsel and design screening strategies for women with a family history of breast cancer.
Of all women with breast cancer, 5% to 10% may have a germline mutation of the genes BRCA1 and BRCA2.
Specific mutations of BRCA1 and BRCA2 are more common in women of Jewish ancestry.
The estimated lifetime risk of developing breast cancer for women with BRCA1 and BRCA2 mutations is 40% to 85%. Carriers with a history of breast cancer have an increased risk of contralateral disease that may be as high as 5% per year.
Male BRCA2 mutation carriers also have an increased risk of breast cancer.
Mutations in either the BRCA1 or the BRCA2 gene also confer an increased risk of ovarian cancer
or other primary cancers.
Once a BRCA1 or BRCA2 mutation has been identified, other family members can be referred for genetic counseling and testing.
(Refer to the PDQ summaries on Genetics of Breast and Gynecologic Cancers; Breast Cancer Prevention; and Breast Cancer Screening for more information.)
(Refer to the PDQ summary on Breast Cancer Prevention for more information about factors that increase the risk of breast cancer.)
Protective factors and interventions to reduce the risk of female breast cancer include the following:
(Refer to the PDQ summary on Breast Cancer Prevention for more information about factors that decrease the risk of breast cancer.)
Clinical trials have established that screening asymptomatic women using mammography, with or without clinical breast examination, decreases breast cancer mortality. (Refer to the PDQ summary on Breast Cancer Screening for more information.)
When breast cancer is suspected, patient management generally includes the following:
The following tests and procedures are used to diagnose breast cancer:
Pathologically, breast cancer can be a multicentric and bilateral disease. Bilateral disease is somewhat more common in patients with infiltrating lobular carcinoma. At 10 years after diagnosis, the risk of a primary breast cancer in the contralateral breast ranges from 3% to 10%, although endocrine therapy decreases that risk.
The development of a contralateral breast cancer is associated with an increased risk of distant recurrence.
When BRCA1/BRCA2 mutation carriers were diagnosed before age 40 years, the risk of a contralateral breast cancer reached nearly 50% in the ensuing 25 years.
Patients who have breast cancer will undergo bilateral mammography at the time of diagnosis to rule out synchronous disease. To detect either recurrence in the ipsilateral breast in patients treated with breast-conserving surgery or a second primary cancer in the contralateral breast, patients will continue to have regular breast physical examinations and mammograms.
The role of MRI in screening the contralateral breast and monitoring women treated with breast-conserving therapy continues to evolve. Because an increased detection rate of mammographically occult disease has been demonstrated, the selective use of MRI for additional screening is occurring more frequently despite the absence of randomized, controlled data. Because only 25% of MRI-positive findings represent malignancy, pathologic confirmation before treatment is recommended. Whether this increased detection rate will translate into improved treatment outcome is unknown.
Breast cancer is commonly treated by various combinations of surgery, radiation therapy, chemotherapy, and hormone therapy. Prognosis and selection of therapy may be influenced by the following clinical and pathology features (based on conventional histology and immunohistochemistry):
The use of molecular profiling in breast cancer includes the following:
On the basis of ER, PR, and HER2/neu results, breast cancer is classified as one of the following types:
ER, PR, and HER2 status are important in determining prognosis and in predicting response to endocrine and HER2-directed therapy. The American Society of Clinical Oncology/College of American Pathologists consensus panel has published guidelines to help standardize the performance, interpretation, and reporting of assays used to assess the ER-PR status by immunohistochemistry and HER2 status by immunohistochemistry and in situ hybridization.
Gene profile tests include the following:
The following trials describe the prognostic and predictive value of multigene assays in early breast cancer:
Patients in this study with a low-risk score were found to have very low rates of recurrence at 5 years with endocrine therapy.
In the middle-risk group in the TAILORx study (recurrence score, 11–25), 6,907 women were randomly assigned to endocrine therapy alone or endocrine therapy plus chemotherapy.
Of these, 3,399 women on the endocrine therapy-alone arm and 3,312 women on the endocrine therapy-plus-chemotherapy arm were available for an analysis according to the randomized treatment assignments. After a median follow-up of 90 months, the difference in invasive DFS, the main study endpoint, met the prespecified noninferiority criterion (P > .10 for a test of no difference after 835 events had occurred) suggesting the noninferiority of endocrine therapy compared with endocrine therapy plus chemotherapy.
Results from the prospective, randomized RxPONDER (NCT01272037) trial will help to determine if there is a benefit from adjuvant chemotherapy in patients with ER-positive-, node-positive early breast cancer treated with endocrine therapy, and a recurrence score below 25.
Many other gene-based assays may guide treatment decisions in patients with early breast cancer (e.g., Predictor Analysis of Microarray 50 [PAM50] Risk of Recurrence [ROR] score, EndoPredict, Breast Cancer Index).
Although certain rare inherited mutations, such as those of BRCA1 and BRCA2, predispose women to develop breast cancer, prognostic data on BRCA1/BRCA2 mutation carriers who have developed breast cancer are conflicting. These women are at greater risk of developing contralateral breast cancer. (Refer to the Prognosis of BRCA1- and BRCA2-related breast cancer section of the PDQ Genetics of Breast and Gynecologic Cancers summary for more information.)
After careful consideration, patients with severe symptoms may be treated with hormone replacement therapy. For more information, refer to the following PDQ summaries:
Other PDQ summaries containing information related to breast cancer include the following:
表格1描述了基于肿瘤位置的乳腺癌的组织学分类。
浸润性或侵袭性导管癌是最常见的乳腺癌组织学类型,占所有病例的70%至80%。
肿瘤位置 | 组织学亚类 |
---|---|
癌,NOS | |
导管的 | 导管内的(原位) |
主要成分侵入 | |
侵袭性,NOS | |
粉刺 | |
炎症性 | |
骨髓淋巴细胞浸润 | |
粘液性(胶质性) | |
乳突的 | |
硬癌 | |
管状的 | |
其他 | |
小叶的 | 原位癌为主的浸润性癌 |
侵袭性的 | |
乳头 | 乳腺Paget病,NOS |
伴有导管内癌的Paget病 | |
伴有浸润性导管癌的Paget病 | |
其他 | 未分化癌 |
化生型 | |
NOS=无特殊说明。 |
以下肿瘤亚型发生在乳房,但不被认为是典型的乳腺癌:
Table 1 describes the histologic classification of breast cancer based on tumor location.
Infiltrating or invasive ductal cancer is the most common breast cancer histologic type and comprises 70% to 80% of all cases.
Tumor Location | Histologic Subtype |
---|---|
Carcinoma, NOS | |
Ductal | Intraductal (in situ) |
Invasive with predominant component | |
Invasive, NOS | |
Comedo | |
Inflammatory | |
Medullary with lymphocytic infiltrate | |
Mucinous (colloid) | |
Papillary | |
Scirrhous | |
Tubular | |
Other | |
Lobular | Invasive with predominant in situ component |
Invasive | |
Nipple | Paget disease, NOS |
Paget disease with intraductal carcinoma | |
Paget disease with invasive ductal carcinoma | |
Other | Undifferentiated carcinoma |
Metaplastic | |
NOS = not otherwise specified. |
The following tumor subtypes occur in the breast but are not considered typical breast cancers:
美国癌症联合委员会(AJCC)分期系统为患者在预后方面提供了分期策略。 治疗决策部分根据分期制定,也根据其他临床因素,例如以下因素,其中一些因素也应用于分期:
用于定义生物标志物状态的标准描述如下:
IHC:
ISH(双探针):
ISH(单探针):
AJCC根据TNM(肿瘤、淋巴结、转移)分级来定义乳腺癌分期。
肿瘤的分级由其形态特征决定,如管状形状、核多形性和线粒体计数。
T类别 | T标准 |
---|---|
cN类别 | cN标准 |
pN类别 | pN标准 |
M类别 | M标准 |
G | G定义 |
G | G定义 |
TX | 不可评估的原发肿瘤。 |
T0 | 无原发肿瘤的证据。 |
Tisb | DCIS. |
Tis (Paget) | 与浸润性癌和/或乳腺实质的DCIS无关的乳头Paget病。与Paget病有关的乳腺实质癌根据实质性疾病的大小和特征进行分类,但是应注意到Paget疾病的存在。 |
T1 | 肿瘤最大直径≤20mm。 |
–T1mi | 肿瘤最大直径≤1mm。 |
–T1a | 肿瘤最大直径>1mm但≤5mm(圆周任意测量值>1.0-1.9mm到2,mm)。 |
–T1b | 肿瘤最大直径>5mm但≤10mm。 |
–T1c | 肿瘤最大直径>10mm但≤20mm。 |
T2 | 肿瘤最大直径>20 mm但≤50 mm。 |
T3 | 肿瘤最大直径>50mm。 |
T4 | 累及胸壁和/或皮肤(溃疡或宏观结节)的任意大小的肿瘤;仅侵入真皮不符合T4。 |
–T4a | 累及胸壁;在没有累及胸壁的情况下,侵入或依附于胸肌的不符合T4。 |
–T4b | 不符合炎症性癌标准的皮肤上的溃疡和/或同侧卫星结节和/或水肿(包括橘皮样变)。 |
–T4c | T4a和T4b都存在。 |
–T4d | 炎症性癌(见分类规则)。 |
cN类别 | cN标准 |
cNX c | 无法评估区域淋巴结(例如,以前已被切除)。 |
cN0 | 无区域淋巴结转移(通过成像或临床检查)。 |
cN1 | 活动性好的同侧I、II站淋巴结转移。 |
–cN1mi d | 微转移(约200个细胞,>0.2mm但≤2.0mm)。 |
cN2 | 临床固定的或表面粗糙的同侧I、II级腋下淋巴结的转移; |
或在没有腋下淋巴结转移的情况下,转移至同侧乳腺内部的结节。 | |
–cN2a | 同侧I、II站腋下淋巴结的转移,淋巴结(表面粗糙)融合。 |
–cN2b | 在无腋下淋巴结转移的情况下,在同侧乳腺内部中的转移结节。 |
cN3 | 在同侧锁骨下(腋下III站)淋巴结(s)转移,伴或不伴有l、II站腋下淋巴结转移;或同侧内乳淋巴结转移且伴有I、II站腋下淋巴结转移;或同侧锁骨上淋巴结转移,伴或不伴有腋下或内乳淋巴结转移。 |
cN3a | 同侧锁骨下淋巴结转移。 |
–cN3b | 同侧内乳淋巴结和腋下淋巴结转移。 |
–cN3c | 同侧锁骨上淋巴结转移。 |
pN类别 | pN标准 |
pNX | 无法评估的区域淋巴结(例如,不能切除用于病理研究的或以前被切除的)。 |
pN0 | 无区域淋巴结转移被识别或仅ITCs被识别。 |
–pN0(i+) | 区域淋巴结内仅有ITCs(恶性细胞簇≤0.2mm)。 |
–pN0(mol+) | 通过RT-PCR检测的阳性分子发现;未检测到ITCs。 |
pN1 | 微转移;1-3个腋下淋巴结转移;和/或存在微转移或临床阴性内乳淋巴结微转移或前哨淋巴结活检确认的宏转移。 |
–pN1mi | 微转移(约200个细胞,>0.2mm但≤2.0mm)。 |
–pN1a | 1-3个腋下淋巴结转移,至少有一个转移灶>2.0mm。 |
–pN1b | 同侧内乳前哨淋巴结转移,不包括ITCs。 |
–pN1c | 合并pN1a和pN1b。 |
pN2 | 4-9个腋下淋巴结转移;或通过影像学确诊的同侧内乳淋巴结转移但无腋下淋巴结转移。 |
–pN2a | 4-9个腋下淋巴结转移(至少一个肿瘤沉积>2.0mm)。 |
–pN2b | 临床检测到的内乳淋巴结转移,无论是否有显微确认;且腋下淋巴结病理阴性。 |
pN3 | ≥10个腋下淋巴结转移;或锁骨下(腋窝III级)淋巴结;或通过影像学确诊的同侧内乳区淋巴结转移同时存在一个或多个I、II级腋下淋巴结转移;或>3个腋下淋巴结转移、微转移或通过在临床阴性的同侧内乳淋巴结上进行的前哨淋巴结活检证实的宏转移。 |
–pN3a | ≥10个腋下淋巴结转移(至少1个肿瘤沉积>2.0mm);或锁骨下(III站腋下淋巴)淋巴结转移。 |
–pN3b | pN1a或pN2a且存在cN2b(通过影像学确诊的内乳区淋巴结); |
或pN2a且存在pN1b。 | |
–pN3c | 同侧锁骨上淋巴结转移。 |
M类别 | M标准 |
M0 | 没有往别处转移的临床或影像学证据。 b |
cM0(i+) | 没有远处转移的临床或影像证据时,在无转移症状或迹象的患者中,由显微或分子技术在循环血液、骨髓或其他非区域性淋巴结组织中检测出<0.2mm的肿瘤细胞或沉淀物。 |
cM1 | 通过临床和影像的方法发现的远处转移。 |
pM1 | 任何经组织学证明的在远处器官中的转移;或者,在非局部结节中,转移灶 >0.2mm。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 低组合组织学分级(有利的),SBR评分为3-5分。 |
G2 | 中等的组合组织学分级(适度有利的);SBR评分为6-7分。 |
G3 | 高组合组织学分级(不利的);SBR评分为8-9分。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 核分裂数少 |
G2 | 核分裂数中等 |
G3 | 核分裂数多 |
DCIS=导管原位癌。 | |
a转载需获得AJCC的许可:乳腺,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b小叶原位癌是一种良性实性肿瘤,并在第8版的《AJCC癌症分期手册》中的TNM分期中被移除。 | |
c分类规则--TNM解剖系统是一种编码疾病程度的方法。通过肿瘤(T)、区域淋巴结(N)和远处转移(M)的累及范围进行分期。T、N和M通过临床信息、手术发现和病理信息进行分期。已证实的新辅助治疗对疾病预后的影响程度以及对治疗的反应,需要保证定义中yp前缀和对治疗的反应的使用。使用新辅助疗法不会改变临床(治疗前)分期。根据TNM规则,临床分期的解剖部分通过前缀c(例如cT)标识。此外,新辅助治疗前的临床分期包括使用细针抽吸(FNA)或空芯针穿刺活检和前哨淋巴结活检。这些分别用后缀f和sn表示。FNA或空芯针穿刺活检确诊的淋巴结转移被归类为宏转移(cN1),无论最终病理标本中的肿瘤大小如何。例如,如果在新辅助全身治疗之前,一位患者存在一个1cm大小的原发性病灶但没有可触及的结节,超声引导的腋窝淋巴结FNA活检阳性,该患者将根据其临床(治疗前)分期被归类为cN1(f)并归于IIA期。同样的,如果患者在新辅助全身治疗前有一个阳性的腋下前哨淋巴结,则该肿瘤被归类为cN1(sn)(IIA期)。根据TNM规则,在没有病理T评估(切除原发性肿瘤)的情况下,需标记前缀p(例如pT)以被识别,新辅助治疗前的淋巴结显微评估,甚至是通过完全切除如前哨淋巴结活检进行的评估,仍然被分类为临床(cN)。 | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn)和(f)后缀应分别添加到N类别上,以表示通过前哨淋巴结活检或细针抽吸/空芯针穿刺活检确诊转移。 | |
c 这种cNX类别很少用于之前被手术切除的区域淋巴结或没有进行腋窝体格检查记录的情况。 | |
d cN1mi很少使用,但在肿瘤切除前进行前哨淋巴结活检的情况下可能适用,最有可能发生在接受新辅助治疗的情况下。 | |
ITCs=孤立肿瘤细胞簇;RT-PCR=逆转录酶--聚合酶链反应。 | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn)和(f)后缀应分别添加到N类别上,以表示通过前哨淋巴结活检或细针抽吸/空芯针穿刺活检确认转移,且无更多的淋巴结切除 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b注意,不需要影像研究来决定cM0分类。 | |
SBR=Scarff-Bloom-Richardson分级系统,Nottingham改造。 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. |
cN类别 | cN标准 |
---|---|
pN类别 | pN标准 |
M类别 | M标准 |
G | G定义 |
G | G定义 |
cNX c | 无法评估区域淋巴结(例如,以前已被切除)。 |
cN0 | 无区域淋巴结转移(通过成像或临床检查)。 |
cN1 | 活动性好的同侧I、II站淋巴结转移。 |
–cN1mi d | 微转移(约200个细胞,>0.2mm但≤2.0mm)。 |
cN2 | 临床固定的或表面粗糙的同侧I、II级腋下淋巴结的转移; |
或在没有腋下淋巴结转移的情况下,转移至同侧乳腺内部的结节。 | |
–cN2a | 同侧I、II站腋下淋巴结的转移,淋巴结(表面粗糙)融合。 |
–cN2b | 在无腋下淋巴结转移的情况下,在同侧乳腺内部中的转移结节。 |
cN3 | 在同侧锁骨下(腋下III站)淋巴结(s)转移,伴或不伴有l、II站腋下淋巴结转移;或同侧内乳淋巴结转移且伴有I、II站腋下淋巴结转移;或同侧锁骨上淋巴结转移,伴或不伴有腋下或内乳淋巴结转移。 |
cN3a | 同侧锁骨下淋巴结转移。 |
–cN3b | 同侧内乳淋巴结和腋下淋巴结转移。 |
–cN3c | 同侧锁骨上淋巴结转移。 |
pN类别 | pN标准 |
pNX | 无法评估的区域淋巴结(例如,不能切除用于病理研究的或以前被切除的)。 |
pN0 | 无区域淋巴结转移被识别或仅ITCs被识别。 |
–pN0(i+) | 区域淋巴结内仅有ITCs(恶性细胞簇≤0.2mm)。 |
–pN0(mol+) | 通过RT-PCR检测的阳性分子发现;未检测到ITCs。 |
pN1 | 微转移;1-3个腋下淋巴结转移;和/或存在微转移或临床阴性内乳淋巴结微转移或前哨淋巴结活检确认的宏转移。 |
–pN1mi | 微转移(约200个细胞,>0.2mm但≤2.0mm)。 |
–pN1a | 1-3个腋下淋巴结转移,至少有一个转移灶>2.0mm。 |
–pN1b | 同侧内乳前哨淋巴结转移,不包括ITCs。 |
–pN1c | 合并pN1a和pN1b。 |
pN2 | 4-9个腋下淋巴结转移;或通过影像学确诊的同侧内乳淋巴结转移但无腋下淋巴结转移。 |
–pN2a | 4-9个腋下淋巴结转移(至少一个肿瘤沉积>2.0mm)。 |
–pN2b | 临床检测到的内乳淋巴结转移,无论是否有显微确认;且腋下淋巴结病理阴性。 |
pN3 | ≥10个腋下淋巴结转移;或锁骨下(腋窝III级)淋巴结;或通过影像学确诊的同侧内乳区淋巴结转移同时存在一个或多个I、II级腋下淋巴结转移;或>3个腋下淋巴结转移、微转移或通过在临床阴性的同侧内乳淋巴结上进行的前哨淋巴结活检证实的宏转移。 |
–pN3a | ≥10个腋下淋巴结转移(至少1个肿瘤沉积>2.0mm);或锁骨下(III站腋下淋巴)淋巴结转移。 |
–pN3b | pN1a或pN2a且存在cN2b(通过影像学确诊的内乳区淋巴结); |
或pN2a且存在pN1b。 | |
–pN3c | 同侧锁骨上淋巴结转移。 |
M类别 | M标准 |
M0 | 没有往别处转移的临床或影像学证据。 b |
cM0(i+) | 没有远处转移的临床或影像证据时,在无转移症状或迹象的患者中,由显微或分子技术在循环血液、骨髓或其他非区域性淋巴结组织中检测出<0.2mm的肿瘤细胞或沉淀物。 |
cM1 | 通过临床和影像的方法发现的远处转移。 |
pM1 | 任何经组织学证明的在远处器官中的转移;或者,在非局部结节中,转移灶 >0.2mm。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 低组合组织学分级(有利的),SBR评分为3-5分。 |
G2 | 中等的组合组织学分级(适度有利的);SBR评分为6-7分。 |
G3 | 高组合组织学分级(不利的);SBR评分为8-9分。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 核分裂数少 |
G2 | 核分裂数中等 |
G3 | 核分裂数多 |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn)和(f)后缀应分别添加到N类别上,以表示通过前哨淋巴结活检或细针抽吸/空芯针穿刺活检确诊转移。 | |
c 这种cNX类别很少用于之前被手术切除的区域淋巴结或没有进行腋窝体格检查记录的情况。 | |
d cN1mi很少使用,但在肿瘤切除前进行前哨淋巴结活检的情况下可能适用,最有可能发生在接受新辅助治疗的情况下。 | |
ITCs=孤立肿瘤细胞簇;RT-PCR=逆转录酶--聚合酶链反应。 | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn)和(f)后缀应分别添加到N类别上,以表示通过前哨淋巴结活检或细针抽吸/空芯针穿刺活检确认转移,且无更多的淋巴结切除 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b注意,不需要影像研究来决定cM0分类。 | |
SBR=Scarff-Bloom-Richardson分级系统,Nottingham改造。 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. |
pN类别 | pN标准 |
---|---|
M类别 | M标准 |
G | G定义 |
G | G定义 |
pNX | 无法评估的区域淋巴结(例如,不能切除用于病理研究的或以前被切除的)。 |
pN0 | 无区域淋巴结转移被识别或仅ITCs被识别。 |
–pN0(i+) | 区域淋巴结内仅有ITCs(恶性细胞簇≤0.2mm)。 |
–pN0(mol+) | 通过RT-PCR检测的阳性分子发现;未检测到ITCs。 |
pN1 | 微转移;1-3个腋下淋巴结转移;和/或存在微转移或临床阴性内乳淋巴结微转移或前哨淋巴结活检确认的宏转移。 |
–pN1mi | 微转移(约200个细胞,>0.2mm但≤2.0mm)。 |
–pN1a | 1-3个腋下淋巴结转移,至少有一个转移灶>2.0mm。 |
–pN1b | 同侧内乳前哨淋巴结转移,不包括ITCs。 |
–pN1c | 合并pN1a和pN1b。 |
pN2 | 4-9个腋下淋巴结转移;或通过影像学确诊的同侧内乳淋巴结转移但无腋下淋巴结转移。 |
–pN2a | 4-9个腋下淋巴结转移(至少一个肿瘤沉积>2.0mm)。 |
–pN2b | 临床检测到的内乳淋巴结转移,无论是否有显微确认;且腋下淋巴结病理阴性。 |
pN3 | ≥10个腋下淋巴结转移;或锁骨下(腋窝III级)淋巴结;或通过影像学确诊的同侧内乳区淋巴结转移同时存在一个或多个I、II级腋下淋巴结转移;或>3个腋下淋巴结转移、微转移或通过在临床阴性的同侧内乳淋巴结上进行的前哨淋巴结活检证实的宏转移。 |
–pN3a | ≥10个腋下淋巴结转移(至少1个肿瘤沉积>2.0mm);或锁骨下(III站腋下淋巴)淋巴结转移。 |
–pN3b | pN1a或pN2a且存在cN2b(通过影像学确诊的内乳区淋巴结); |
或pN2a且存在pN1b。 | |
–pN3c | 同侧锁骨上淋巴结转移。 |
M类别 | M标准 |
M0 | 没有往别处转移的临床或影像学证据。 b |
cM0(i+) | 没有远处转移的临床或影像证据时,在无转移症状或迹象的患者中,由显微或分子技术在循环血液、骨髓或其他非区域性淋巴结组织中检测出<0.2mm的肿瘤细胞或沉淀物。 |
cM1 | 通过临床和影像的方法发现的远处转移。 |
pM1 | 任何经组织学证明的在远处器官中的转移;或者,在非局部结节中,转移灶 >0.2mm。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 低组合组织学分级(有利的),SBR评分为3-5分。 |
G2 | 中等的组合组织学分级(适度有利的);SBR评分为6-7分。 |
G3 | 高组合组织学分级(不利的);SBR评分为8-9分。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 核分裂数少 |
G2 | 核分裂数中等 |
G3 | 核分裂数多 |
ITCs=孤立肿瘤细胞簇;RT-PCR=逆转录酶--聚合酶链反应。 | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn)和(f)后缀应分别添加到N类别上,以表示通过前哨淋巴结活检或细针抽吸/空芯针穿刺活检确认转移,且无更多的淋巴结切除 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b注意,不需要影像研究来决定cM0分类。 | |
SBR=Scarff-Bloom-Richardson分级系统,Nottingham改造。 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. |
M类别 | M标准 |
---|---|
G | G定义 |
G | G定义 |
M0 | 没有往别处转移的临床或影像学证据。 b |
cM0(i+) | 没有远处转移的临床或影像证据时,在无转移症状或迹象的患者中,由显微或分子技术在循环血液、骨髓或其他非区域性淋巴结组织中检测出<0.2mm的肿瘤细胞或沉淀物。 |
cM1 | 通过临床和影像的方法发现的远处转移。 |
pM1 | 任何经组织学证明的在远处器官中的转移;或者,在非局部结节中,转移灶 >0.2mm。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 低组合组织学分级(有利的),SBR评分为3-5分。 |
G2 | 中等的组合组织学分级(适度有利的);SBR评分为6-7分。 |
G3 | 高组合组织学分级(不利的);SBR评分为8-9分。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 核分裂数少 |
G2 | 核分裂数中等 |
G3 | 核分裂数多 |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b注意,不需要影像研究来决定cM0分类。 | |
SBR=Scarff-Bloom-Richardson分级系统,Nottingham改造。 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. |
G | G定义 |
---|---|
G | G定义 |
GX | 分级不可被评估。 |
G1 | 低组合组织学分级(有利的),SBR评分为3-5分。 |
G2 | 中等的组合组织学分级(适度有利的);SBR评分为6-7分。 |
G3 | 高组合组织学分级(不利的);SBR评分为8-9分。 |
G | G定义 |
GX | 分级不可被评估。 |
G1 | 核分裂数少 |
G2 | 核分裂数中等 |
G3 | 核分裂数多 |
SBR=Scarff-Bloom-Richardson分级系统,Nottingham改造。 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. |
G | G定义 |
---|---|
GX | 分级不可被评估。 |
G1 | 核分裂数少 |
G2 | 核分裂数中等 |
G3 | 核分裂数多 |
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. |
侵袭性癌症有3个分期组表:
在美国,癌症登记处和临床医生必须使用临床和病理预后分期组表进行报告,对分级、HER2、ER和PR状态进行检测,并报告美国所有浸润性癌症病例的结果。
分期 | TNM |
---|---|
0 | Tis、 N0、 M0 |
IA | T1、 N0、 M0 |
IB | T0、 N1mi、 M0 |
T1、 N1mi、 M0 | |
IIA | T0、 N1、 M0 |
T1、 N1、 M0 | |
T2、 N0、 M0 | |
IIB | T2、 N1、 M0 |
T3、 N0、 M0 | |
IIIA | T0、 N2、 M0 |
T1、 N2、 M0 | |
T2、 N2、 M0 | |
T3、 N1、 M0 | |
T3、 N2、 M0 | |
IIIB | T4、 N0、 M0 |
T4、 N1、 M0 | |
T4、 N2、 M0 | |
IIIC | 任何T (Tis、 T1、 T0、 T2、 T3、 T4; N3、 M0) |
IV | 任何T (Tis、 T1、 T0、 T2、 T3、 T4;任何N = N0、 N1mi、 N1、 N2、 N3、 M1) |
T=原发肿瘤;N=区域淋巴结;M=远处转移。 | |
a转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
注释: | |
1.T1包括T1mi。 | |
2.伴有淋巴结微转移(N1mi)的T0和T1肿瘤为IB期。 | |
3.伴有淋巴结微转移(N1mi)的T2、T3和T4肿瘤为N1期。 | |
4.M0包括M0(i+)。 | |
5.名称pM0无效;任何M0都是临床的。 | |
6.如果患者在接受新辅助系统治疗前列入M1期,则为IV期,无论对新辅助治疗的反应如何,仍保持为IV期。 | |
7.如果患者没有接受新辅助治疗,手术后在无疾病进展的4个月内影像学检查发现存在远处转移,则可改变分期。 | |
8.新辅助治疗后的分期需要在T和N加上yc或ypn前缀。如果对新辅助治疗存在完全的病理欢姐(pCR),例如如ypT0、ypN0、cM0,则不进行解剖分期。 |
临床预后分期在美国用于浸润性乳腺癌患者的临床分类和分期。它使用基于患者的病史、体格检查、影像学结果(临床分期不需要)和活检的TNM信息。
TNM | 分级 | HER2状态 | ER状态 | PR状态 | 分期组 |
---|---|---|---|---|---|
Tis、N0、M0 | 任何(参照表6和表7) | 任何 | 任何 | 任何 | 0 |
T1b、 N0、 M0 | G1 | 阳性 | 阳性 | 阳性 | IA |
阴性 | IA | ||||
T0、 N1mi、 M0 | 阴性 | 阳性 | IA | ||
阴性 | IA | ||||
T1b、 N1mi、 M0 | 阴性 | 阳性 | 阳性 | IA | |
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IB | ||||
G2 | 阳性 | 阳性 | 阳性 | IA | |
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IA | ||||
阴性 | 阳性 | 阳性 | IA | ||
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IB | ||||
G3 | 阳性 | 阳性 | 阳性 | IA | |
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IA | ||||
阴性 | 阳性 | 阳性 | IA | ||
阴性 | IB | ||||
阴性 | 阳性 | IB | |||
阴性 | IB | ||||
T0、 N1c、 M0; T1b、 N1c、 M0; T2、 N0、 M0 | G1 | 阳性 | 阳性 | 阳性 | IB |
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIA | ||||
阴性 | 阳性 | 阳性 | IB | ||
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIA | ||||
G2 | 阳性 | 阳性 | 阳性 | IB | |
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIA | ||||
阴性 | 阳性 | 阳性 | IB | ||
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIB | ||||
G3 | 阳性 | 阳性 | 阳性 | IB | |
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIA | ||||
阴性 | 阳性 | 阳性 | IIA | ||
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIB | ||||
T1b、 N1c、 M0; T2、 N0、 M0 | G1 | 阳性 | 阳性 | 阳性 | IB |
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIB | ||||
阴性 | 阳性 | 阳性 | IIA | ||
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIB | ||||
G2 | 阳性 | 阳性 | 阳性 | IB | |
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIB | ||||
阴性 | 阳性 | 阳性 | IIA | ||
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIIB | ||||
G3 | 阳性 | 阳性 | 阳性 | IB | |
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIB | ||||
阴性 | 阳性 | 阳性 | IIB | ||
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIB | ||||
T0、 N2、 M0; T1b、 N2、 M0; T2、 N2、 M0; T3、 N1d、 M0; T3、 N2、 M0 | G1 | 阳性 | 阳性 | 阳性 | IIA |
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIA | ||||
阴性 | 阳性 | 阳性 | IIA | ||
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIB | ||||
G2 | 阳性 | 阳性 | 阳性 | IIA | |
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIA | ||||
阴性 | 阳性 | 阳性 | IIA | ||
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIB | ||||
G3 | 阳性 | 阳性 | 阳性 | IIB | |
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIA | ||||
阴性 | 阳性 | 阳性 | IIIA | ||
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIC | ||||
T4、 N1c、 M0; T4、 N1c、 M0; T4、N2、M0、 N3、 M0 | G1 | 阳性 | 阳性 | 阳性 | IIIA |
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIB | ||||
阴性 | 阳性 | 阳性 | IIIB | ||
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIC | ||||
G2 | 阳性 | 阳性 | 阳性 | IIIA | |
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIB | ||||
阴性 | 阳性 | 阳性 | IIIB | ||
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIC | ||||
G3 | 阳性 | 阳性 | 阳性 | IIIB | |
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIB | ||||
阴性 | 阳性 | 阳性 | IIIB | ||
阴性 | IIIC | ||||
阴性 | 阳性 | IIIC | |||
阴性 | IIIC | ||||
任何T、任何N、M1 | 任何(参照表6和表7) | 任何 | 任何 | 任何 | IV |
T=原发肿瘤;N=区域淋巴结;M=远处转移。 | |||||
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |||||
b T1包括T1mi。 | |||||
c N1不包括N1mi。T1、N1mi、M0和T0、N1mi、M0癌症预后视为与T1、N0、M0癌症的预后分期相同。 | |||||
d N1包括N1mi。T2、N1,;T3、N1和T4、N1分别包括T2、T3和T4癌症与N1mi癌症分期。 | |||||
注释: | |||||
1.因为N1mi分类需要评估整个的淋巴结,并且不能基于细针穿刺活检或空芯针活检进行分期,因此N1mi只能和临床预后分期一起用于淋巴结已切除但原发性癌症未切除的临床分期,例如在前哨淋巴结活检前接受新辅助化疗或内分泌治疗的情况。 | |||||
2.对于有淋巴结累及但没有原发肿瘤证据(如T0、N1等)或存在乳腺导管原位癌(如Tis、N1等)的病例,则应用从淋巴结的肿瘤获取的分级、人类表皮生长因子受体2 (HER2)、雌激素受体和孕激素受体的信息进行分期。 | |||||
3.对于通过2013年美国临床肿瘤学家协会/美国病理学家学院HER2检测指南中原位杂交(荧光原位杂交或显色原位杂交)检测发现HER2不确定的病例,在病理预后分期组表中,应使用HER2阴性类别进行分期。 | |||||
4.那些提供信息的和大部分已接受了适当的内分泌和/或全身化疗(包括抗her2治疗)治疗的乳腺癌患者人群是这些预后分期组的预后价值的基础。 |
病理预后分期适用于最初接受手术治疗的浸润性乳腺癌患者。它包括所有用于临床分期的信息,手术发现以及手术切除肿瘤后的病理结果。病理预后分期不用于手术切除肿瘤前接受新辅助治疗的患者。
TNM | 分级 | HER2状态 | ER状态 | PR状态 | 分期组 |
---|---|---|---|---|---|
Tis, N0, M0 | 任何(参照表6和表7) | 任何 | 任何 | 任何 | 0 |
T1b、 N1c、 M0; T0、 N1c、 M0; T1b、 N1mi、 M0 | G1 | 阳性 | 阳性 | 阳性 | IA |
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IA | ||||
阴性 | 阳性 | 阳性 | IA | ||
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IA | ||||
G2 | 阳性 | 阳性 | 阳性 | IA | |
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IA | ||||
阴性 | 阳性 | 阳性 | IA | ||
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IB | ||||
G3 | 阳性 | 阳性 | 阳性 | IA | |
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IA | ||||
阴性 | 阳性 | 阳性 | IA | ||
阴性 | IA | ||||
阴性 | 阳性 | IA | |||
阴性 | IB | ||||
T0、 N1c、 M0; T1b、 N1c、 M0; T2、 N0、 M0 | G1 | 阳性 | 阳性 | 阳性 | IA |
阴性 | IB | ||||
阴性 | 阳性 | IB | |||
阴性 | IIA | ||||
阴性 | 阳性 | 阳性 | IA | ||
阴性 | IB | ||||
阴性 | 阳性 | IB | |||
阴性 | IIA | ||||
G2 | 阳性 | 阳性 | 阳性 | IA | |
阴性 | IB | ||||
阴性 | 阳性 | IB | |||
阴性 | IIA | ||||
阴性 | 阳性 | 阳性 | IA | ||
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIA | ||||
G3 | 阳性 | 阳性 | 阳性 | IA | |
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIA | ||||
阴性 | 阳性 | 阳性 | IB | ||
阴性 | IIA | ||||
阴性 | 阳性 | IIA | |||
阴性 | IIA | ||||
T1b、 N1c、 M0; T2、 N0、 M0 | G1 | 阳性 | 阳性 | 阳性 | IA |
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIB | ||||
阴性 | 阳性 | 阳性 | IA | ||
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIB | ||||
G2 | 阳性 | 阳性 | 阳性 | IB | |
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIB | ||||
阴性 | 阳性 | 阳性 | IB | ||
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIB | ||||
G3 | 阳性 | 阳性 | 阳性 | IB | |
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIB | ||||
阴性 | 阳性 | 阳性 | IIA | ||
阴性 | IIB | ||||
阴性 | 阳性 | IIB | |||
阴性 | IIIA | ||||
T0、 N2、 M0; T1b、 N2、 M0; T2、 N2、 M0; T3、 N1d、 M0; T3、 N2、 M0 | G1 | 阳性 | 阳性 | 阳性 | IB |
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIA | ||||
阴性 | 阳性 | 阳性 | IB | ||
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIA | ||||
G2 | 阳性 | 阳性 | 阳性 | IB | |
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIA | ||||
阴性 | 阳性 | 阳性 | IB | ||
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIB | ||||
G3 | 阳性 | 阳性 | 阳性 | IIA | |
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIA | ||||
阴性 | 阳性 | 阳性 | IIB | ||
阴性 | IIIA | ||||
阴性 | 阳性 | IIIA | |||
阴性 | IIIC | ||||
T4、 N1c、 M0; T4、 N1c、 M0; T4、N2、M0、 N3、 M0 | G1 | 阳性 | 阳性 | 阳性 | IIIA |
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIB | ||||
阴性 | 阳性 | 阳性 | IIIA | ||
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIB | ||||
G2 | 阳性 | 阳性 | 阳性 | IIIA | |
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIB | ||||
阴性 | 阳性 | 阳性 | IIIA | ||
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIC | ||||
G3 | 阳性 | 阳性 | 阳性 | IIIB | |
阴性 | IIIB | ||||
阴性 | 阳性 | IIIB | |||
阴性 | IIIB | ||||
阴性 | 阳性 | 阳性 | IIIB | ||
阴性 | IIIC | ||||
阴性 | 阳性 | IIIC | |||
阴性 | IIIC | ||||
任何T、任何N、M1 | 任何(参照表6和表7) | 任何 | 任何 | 任何 | IV |
T=原发肿瘤;N=区域淋巴结;M=远处转移。 | |||||
a 转载需获得AJCC的许可:Breast,修订版。在:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp. 4–96. | |||||
b T1包括T1mi。 | |||||
c N1不包括N1mi。T1、N1mi、M0和T0、N1mi、M0癌症预后视为与T1、N0、M0癌症的预后分期相同。 | |||||
d N1包括N1mi。T2、N1,;T3、N1和T4、N1分别包括T2、T3和T4癌症与N1mi癌症分期。 | |||||
注释: | |||||
1.对于有淋巴结累及但没有原发肿瘤证据(如T0、N1等)或存在乳腺导管原位癌(如Tis、N1等)的病例,则应用从淋巴结的肿瘤获得的分级、人类表皮生长因子受体2 (HER2)、雌激素受体和孕激素受体的信息进行分期。 | |||||
2.对于通过2013年美国临床肿瘤学家协会/美国病理学家学院HER2检测指南中原位杂交(荧光原位杂交或显色原位杂交)检测发现HER2不明确的病例,在病理预后分期组表中,应使用HER2阴性类别进行分期。 | |||||
3.那些提供信息的和大部分已接受了适当的内分泌和/或全身化疗(包括抗her2治疗)治疗的乳腺癌患者人群是这些预后分期组的预后价值的基础。 |
The American Joint Committee on Cancer (AJCC) staging system provides a strategy for grouping patients with respect to prognosis. Therapeutic decisions are formulated in part according to staging categories but also according to other clinical factors such as the following, some of which are included in the determination of stage:
The standards used to define biomarker status are described as follows:
IHC:
ISH (dual probe):
ISH (single probe):
The AJCC has designated staging by TNM (tumor, node, metastasis) classification to define breast cancer.
The grade of the tumor is determined by its morphologic features, such as tubule formation, nuclear pleomorphism, and mitotic count.
T Category | T Criteria |
---|---|
cN Category | cN Criteria |
pN Category | pN Criteria |
M Category | M Criteria |
G | G Definition |
G | G Definition |
TX | Primary tumor cannot be assessed. |
T0 | No evidence of primary tumor. |
Tisb | DCIS. |
Tis (Paget) | Paget disease of the nipple NOT associated with invasive carcinoma and/or DCIS in the underlying breast parenchyma. Carcinomas in the breast parenchyma associated with Paget disease are categorized based on the size and characteristics of the parenchymal disease, although the presence of Paget disease should still be noted. |
T1 | Tumor ≤20 mm in greatest dimension. |
–T1mi | Tumor ≤1 mm in greatest dimension. |
–T1a | Tumor >1 mm but ≤5 mm in greatest dimension (round any measurement >1.0–1.9 mm to 2 mm). |
–T1b | Tumor >5 mm but ≤10 mm in greatest dimension. |
–T1c | Tumor >10 mm but ≤20 mm in greatest dimension. |
T2 | Tumor >20 mm but ≤50 mm in greatest dimension. |
T3 | Tumor >50 mm in greatest dimension. |
T4 | Tumor of any size with direct extension to the chest wall and/or to the skin (ulceration or macroscopic nodules); invasion of the dermis alone does not qualify as T4. |
–T4a | Extension to the chest wall; invasion or adherence to pectoralis muscle in the absence of invasion of chest wall structures does not qualify as T4. |
–T4b | Ulceration and/or ipsilateral macroscopic satellite nodules and/or edema (including peau d'orange) of the skin that does not meet the criteria for inflammatory carcinoma. |
–T4c | Both T4a and T4b are present. |
–T4d | Inflammatory carcinoma (see Rules for Classificationc). |
cN Category | cN Criteria |
cNX c | Regional lymph nodes cannot be assessed (e.g., previously removed). |
cN0 | No regional lymph node metastases (by imaging or clinical examination). |
cN1 | Metastases to movable ipsilateral Level I, II axillary lymph nodes(s). |
–cN1mi d | Micrometastases (approximately 200 cells, >0.2 mm, but ≤2.0 mm). |
cN2 | Metastases in ipsilateral Level I, II axillary lymph nodes that are clinically fixed or matted; |
or in ipsilateral internal mammary nodes in the absence of axillary lymph node metastases. | |
–cN2a | Metastases in ipsilateral Level I, II axillary lymph nodes fixed to one another (matted) or to other structures. |
–cN2b | Metastases only in ipsilateral internal mammary nodes in the absence of axillary lymph node metastases. |
cN3 | Metastases in ipsilateral infraclavicular (Level Ill axillary) lymph node(s) with or without Level l, II axillary lymph node involvement; or in ipsilateral internal mammary lymph node(s) with Level l, II axillary lymph node metastases; or metastases in ipsilateral supraclavicular lymph node(s) with or without axillary or internal mammary lymph node involvement. |
–cN3a | Metastases in ipsilateral infraclavicular lymph node(s). |
–cN3b | Metastases in ipsilateral internal mammary lymph node(s) and axillary lymph node(s). |
–cN3c | Metastases in ipsilateral supraclavicular lymph node(s). |
pN Category | pN Criteria |
pNX | Regional lymph nodes cannot be assessed (e.g., not removed for pathological study or previously removed). |
pN0 | No regional lymph node metastasis identified or ITCs only. |
–pN0(i+) | ITCs only (malignant cell clusters ≤0.2 mm) in regional lymph node(s). |
–pN0(mol+) | Positive molecular findings by RT-PCR; no ITCs detected. |
pN1 | Micrometastases; or metastases in 1–3 axillary lymph nodes; and/or clinically negative internal mammary nodes with micrometastases or macrometastases by sentinel lymph node biopsy. |
–pN1mi | Micrometastases (~200 cells, >0.2 mm, but ≤2.0 mm). |
–pN1a | Metastases in 1–3 axillary lymph nodes, at least one metastasis >2.0 mm. |
–pN1b | Metastases in ipsilateral internal mammary sentinel nodes, excluding ITCs. |
–pN1c | pN1a and pN1b combined. |
pN2 | Metastases in 4–9 axillary lymph nodes; or positive ipsilateral internal mammary lymph nodes by imaging in the absence of axillary lymph node metastases. |
–pN2a | Metastases in 4–9 axillary lymph nodes (at least 1 tumor deposit >2.0 mm). |
–pN2b | Metastases in clinically detected internal mammary lymph nodes with or without microscopic confirmation; with pathologically negative axillary nodes. |
pN3 | Metastases in ≥10 axillary lymph nodes; or in infraclavicular (Level Ill axillary) lymph nodes; or positive ipsilateral internal mammary lymph nodes by imaging in the presence of one or more positive Level l, II axillary lymph nodes; or in >3 axillary lymph nodes and micrometastases or macrometastases by sentinel lymph node biopsy in clinically negative ipsilateral internal mammary lymph nodes; or in ipsilateral supraclavicular lymph nodes. |
–pN3a | Metastases in ≥10 axillary lymph nodes (at least 1 tumor deposit >2.0 mm); or metastases to the infraclavicular (Level III axillary lymph) nodes. |
–pN3b | pN1a or pN2a in the presence of cN2b (positive internal mammary nodes by imaging); |
or pN2a in the presence of pN1b. | |
–pN3c | Metastases in ipsilateral supraclavicular lymph nodes. |
M Category | M Criteria |
M0 | No clinical or radiographic evidence of distant metastases. b |
cM0(i+) | No clinical or radiographic evidence of distant metastases in the presence of tumor cells or deposits ≤0.2 mm detected microscopically or by molecular techniques in circulating blood, bone marrow, or other nonregional nodal tissue in a patient without symptoms or signs of metastases. |
cM1 | Distant metastases detected by clinical and radiographic means. |
pM1 | Any histologically proven metastases in distant organs; or if in nonregional nodes, metastases >0.2 mm. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low combined histologic grade (favorable), SBR score of 3–5 points. |
G2 | Intermediate combined histologic grade (moderately favorable); SBR score of 6–7 points. |
G3 | High combined histologic grade (unfavorable); SBR score of 8–9 points. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low nuclear grade. |
G2 | Intermediate nuclear grade. |
G3 | High nuclear grade. |
DCIS = ductal carcinoma in situ. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
bLobular carcinoma in situ is a benign entity and is removed from TNM staging in the AJCC Cancer Staging Manual, 8th ed. | |
cRules for Classification - The anatomic TNM system is a method for coding extent of disease. This is done by assigning a category of extent of disease for the tumor (T), regional lymph nodes (N), and distant metastases (M). T, N, and M are assigned by clinical means and by adding surgical findings and pathological information to the clinical information. The documented prognostic impact of postneoadjuvant extent of disease and response to therapy warrant clear definitions of the use of the yp prefix and response to therapy. The use of neoadjuvant therapy does not change the clinical (pretreatment) stage. As per TNM rules, the anatomic component of clinical stage is identified with the prefix c (e.g., cT). In addition, clinical staging can include the use of fine-needle aspiration (FNA) or core-needle biopsy and sentinel lymph node biopsy before neoadjuvant therapy. These are denoted with the postscripts f and sn, respectively. Nodal metastases confirmed by FNA or core-needle biopsy are classified as macrometastases (cN1), regardless of the size of the tumor focus in the final pathological specimen. For example, if, prior to neoadjuvant systemic therapy, a patient with a 1 cm primary has no palpable nodes but has an ultrasound-guided FNA biopsy of an axillary lymph node that is positive, the patient will be categorized as cN1 (f) for clinical (pretreatment) staging and is assigned to Stage IIA. Likewise, if the patient has a positive axillary sentinel node identified before neoadjuvant systemic therapy, the tumor is categorized as cN1 (sn) (Stage IIA). As per TNM rules, in the absence of pathological T evaluation (removal of the primary tumor), which is identified with prefix p (e.g., pT), microscopic evaluation of nodes before neoadjuvant therapy, even by complete removal such as sentinel node biopsy, is still classified as clinical (cN). | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn) and (f) suffixes should be added to the N category to denote confirmation of metastasis by sentinel node biopsy or fine-needle aspiration/core needle biopsy, respectively. | |
cThe cNX category is used sparingly in cases where regional lymph nodes have previously been surgically removed or where there is no documentation of physical examination of the axilla. | |
dcN1mi is rarely used but may be appropriate in cases where sentinel node biopsy is performed before tumor resection, most likely to occur in cases treated with neoadjuvant therapy. | |
ITCs = isolated tumor cells; RT-PCR = reverse transcriptase-polymerase chain reaction. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn) and (f) suffixes should be added to the N category to denote confirmation of metastasis by sentinel node biopsy or fine-needle aspiration/core needle biopsy, respectively, with NO further resection of nodes. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
bNote that imaging studies are not required to assign the cM0 category. | |
SBR = Scarff-Bloom-Richardson grading system, Nottingham Modification. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. |
cN Category | cN Criteria |
---|---|
pN Category | pN Criteria |
M Category | M Criteria |
G | G Definition |
G | G Definition |
cNX c | Regional lymph nodes cannot be assessed (e.g., previously removed). |
cN0 | No regional lymph node metastases (by imaging or clinical examination). |
cN1 | Metastases to movable ipsilateral Level I, II axillary lymph nodes(s). |
–cN1mi d | Micrometastases (approximately 200 cells, >0.2 mm, but ≤2.0 mm). |
cN2 | Metastases in ipsilateral Level I, II axillary lymph nodes that are clinically fixed or matted; |
or in ipsilateral internal mammary nodes in the absence of axillary lymph node metastases. | |
–cN2a | Metastases in ipsilateral Level I, II axillary lymph nodes fixed to one another (matted) or to other structures. |
–cN2b | Metastases only in ipsilateral internal mammary nodes in the absence of axillary lymph node metastases. |
cN3 | Metastases in ipsilateral infraclavicular (Level Ill axillary) lymph node(s) with or without Level l, II axillary lymph node involvement; or in ipsilateral internal mammary lymph node(s) with Level l, II axillary lymph node metastases; or metastases in ipsilateral supraclavicular lymph node(s) with or without axillary or internal mammary lymph node involvement. |
–cN3a | Metastases in ipsilateral infraclavicular lymph node(s). |
–cN3b | Metastases in ipsilateral internal mammary lymph node(s) and axillary lymph node(s). |
–cN3c | Metastases in ipsilateral supraclavicular lymph node(s). |
pN Category | pN Criteria |
pNX | Regional lymph nodes cannot be assessed (e.g., not removed for pathological study or previously removed). |
pN0 | No regional lymph node metastasis identified or ITCs only. |
–pN0(i+) | ITCs only (malignant cell clusters ≤0.2 mm) in regional lymph node(s). |
–pN0(mol+) | Positive molecular findings by RT-PCR; no ITCs detected. |
pN1 | Micrometastases; or metastases in 1–3 axillary lymph nodes; and/or clinically negative internal mammary nodes with micrometastases or macrometastases by sentinel lymph node biopsy. |
–pN1mi | Micrometastases (~200 cells, >0.2 mm, but ≤2.0 mm). |
–pN1a | Metastases in 1–3 axillary lymph nodes, at least one metastasis >2.0 mm. |
–pN1b | Metastases in ipsilateral internal mammary sentinel nodes, excluding ITCs. |
–pN1c | pN1a and pN1b combined. |
pN2 | Metastases in 4–9 axillary lymph nodes; or positive ipsilateral internal mammary lymph nodes by imaging in the absence of axillary lymph node metastases. |
–pN2a | Metastases in 4–9 axillary lymph nodes (at least 1 tumor deposit >2.0 mm). |
–pN2b | Metastases in clinically detected internal mammary lymph nodes with or without microscopic confirmation; with pathologically negative axillary nodes. |
pN3 | Metastases in ≥10 axillary lymph nodes; or in infraclavicular (Level Ill axillary) lymph nodes; or positive ipsilateral internal mammary lymph nodes by imaging in the presence of one or more positive Level l, II axillary lymph nodes; or in >3 axillary lymph nodes and micrometastases or macrometastases by sentinel lymph node biopsy in clinically negative ipsilateral internal mammary lymph nodes; or in ipsilateral supraclavicular lymph nodes. |
–pN3a | Metastases in ≥10 axillary lymph nodes (at least 1 tumor deposit >2.0 mm); or metastases to the infraclavicular (Level III axillary lymph) nodes. |
–pN3b | pN1a or pN2a in the presence of cN2b (positive internal mammary nodes by imaging); |
or pN2a in the presence of pN1b. | |
–pN3c | Metastases in ipsilateral supraclavicular lymph nodes. |
M Category | M Criteria |
M0 | No clinical or radiographic evidence of distant metastases. b |
cM0(i+) | No clinical or radiographic evidence of distant metastases in the presence of tumor cells or deposits ≤0.2 mm detected microscopically or by molecular techniques in circulating blood, bone marrow, or other nonregional nodal tissue in a patient without symptoms or signs of metastases. |
cM1 | Distant metastases detected by clinical and radiographic means. |
pM1 | Any histologically proven metastases in distant organs; or if in nonregional nodes, metastases >0.2 mm. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low combined histologic grade (favorable), SBR score of 3–5 points. |
G2 | Intermediate combined histologic grade (moderately favorable); SBR score of 6–7 points. |
G3 | High combined histologic grade (unfavorable); SBR score of 8–9 points. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low nuclear grade. |
G2 | Intermediate nuclear grade. |
G3 | High nuclear grade. |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn) and (f) suffixes should be added to the N category to denote confirmation of metastasis by sentinel node biopsy or fine-needle aspiration/core needle biopsy, respectively. | |
cThe cNX category is used sparingly in cases where regional lymph nodes have previously been surgically removed or where there is no documentation of physical examination of the axilla. | |
dcN1mi is rarely used but may be appropriate in cases where sentinel node biopsy is performed before tumor resection, most likely to occur in cases treated with neoadjuvant therapy. | |
ITCs = isolated tumor cells; RT-PCR = reverse transcriptase-polymerase chain reaction. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn) and (f) suffixes should be added to the N category to denote confirmation of metastasis by sentinel node biopsy or fine-needle aspiration/core needle biopsy, respectively, with NO further resection of nodes. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
bNote that imaging studies are not required to assign the cM0 category. | |
SBR = Scarff-Bloom-Richardson grading system, Nottingham Modification. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. |
pN Category | pN Criteria |
---|---|
M Category | M Criteria |
G | G Definition |
G | G Definition |
pNX | Regional lymph nodes cannot be assessed (e.g., not removed for pathological study or previously removed). |
pN0 | No regional lymph node metastasis identified or ITCs only. |
–pN0(i+) | ITCs only (malignant cell clusters ≤0.2 mm) in regional lymph node(s). |
–pN0(mol+) | Positive molecular findings by RT-PCR; no ITCs detected. |
pN1 | Micrometastases; or metastases in 1–3 axillary lymph nodes; and/or clinically negative internal mammary nodes with micrometastases or macrometastases by sentinel lymph node biopsy. |
–pN1mi | Micrometastases (~200 cells, >0.2 mm, but ≤2.0 mm). |
–pN1a | Metastases in 1–3 axillary lymph nodes, at least one metastasis >2.0 mm. |
–pN1b | Metastases in ipsilateral internal mammary sentinel nodes, excluding ITCs. |
–pN1c | pN1a and pN1b combined. |
pN2 | Metastases in 4–9 axillary lymph nodes; or positive ipsilateral internal mammary lymph nodes by imaging in the absence of axillary lymph node metastases. |
–pN2a | Metastases in 4–9 axillary lymph nodes (at least 1 tumor deposit >2.0 mm). |
–pN2b | Metastases in clinically detected internal mammary lymph nodes with or without microscopic confirmation; with pathologically negative axillary nodes. |
pN3 | Metastases in ≥10 axillary lymph nodes; or in infraclavicular (Level Ill axillary) lymph nodes; or positive ipsilateral internal mammary lymph nodes by imaging in the presence of one or more positive Level l, II axillary lymph nodes; or in >3 axillary lymph nodes and micrometastases or macrometastases by sentinel lymph node biopsy in clinically negative ipsilateral internal mammary lymph nodes; or in ipsilateral supraclavicular lymph nodes. |
–pN3a | Metastases in ≥10 axillary lymph nodes (at least 1 tumor deposit >2.0 mm); or metastases to the infraclavicular (Level III axillary lymph) nodes. |
–pN3b | pN1a or pN2a in the presence of cN2b (positive internal mammary nodes by imaging); |
or pN2a in the presence of pN1b. | |
–pN3c | Metastases in ipsilateral supraclavicular lymph nodes. |
M Category | M Criteria |
M0 | No clinical or radiographic evidence of distant metastases. b |
cM0(i+) | No clinical or radiographic evidence of distant metastases in the presence of tumor cells or deposits ≤0.2 mm detected microscopically or by molecular techniques in circulating blood, bone marrow, or other nonregional nodal tissue in a patient without symptoms or signs of metastases. |
cM1 | Distant metastases detected by clinical and radiographic means. |
pM1 | Any histologically proven metastases in distant organs; or if in nonregional nodes, metastases >0.2 mm. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low combined histologic grade (favorable), SBR score of 3–5 points. |
G2 | Intermediate combined histologic grade (moderately favorable); SBR score of 6–7 points. |
G3 | High combined histologic grade (unfavorable); SBR score of 8–9 points. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low nuclear grade. |
G2 | Intermediate nuclear grade. |
G3 | High nuclear grade. |
ITCs = isolated tumor cells; RT-PCR = reverse transcriptase-polymerase chain reaction. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
b(sn) and (f) suffixes should be added to the N category to denote confirmation of metastasis by sentinel node biopsy or fine-needle aspiration/core needle biopsy, respectively, with NO further resection of nodes. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
bNote that imaging studies are not required to assign the cM0 category. | |
SBR = Scarff-Bloom-Richardson grading system, Nottingham Modification. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. |
M Category | M Criteria |
---|---|
G | G Definition |
G | G Definition |
M0 | No clinical or radiographic evidence of distant metastases. b |
cM0(i+) | No clinical or radiographic evidence of distant metastases in the presence of tumor cells or deposits ≤0.2 mm detected microscopically or by molecular techniques in circulating blood, bone marrow, or other nonregional nodal tissue in a patient without symptoms or signs of metastases. |
cM1 | Distant metastases detected by clinical and radiographic means. |
pM1 | Any histologically proven metastases in distant organs; or if in nonregional nodes, metastases >0.2 mm. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low combined histologic grade (favorable), SBR score of 3–5 points. |
G2 | Intermediate combined histologic grade (moderately favorable); SBR score of 6–7 points. |
G3 | High combined histologic grade (unfavorable); SBR score of 8–9 points. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low nuclear grade. |
G2 | Intermediate nuclear grade. |
G3 | High nuclear grade. |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
bNote that imaging studies are not required to assign the cM0 category. | |
SBR = Scarff-Bloom-Richardson grading system, Nottingham Modification. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. |
G | G Definition |
---|---|
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low combined histologic grade (favorable), SBR score of 3–5 points. |
G2 | Intermediate combined histologic grade (moderately favorable); SBR score of 6–7 points. |
G3 | High combined histologic grade (unfavorable); SBR score of 8–9 points. |
G | G Definition |
GX | Grade cannot be assessed. |
G1 | Low nuclear grade. |
G2 | Intermediate nuclear grade. |
G3 | High nuclear grade. |
SBR = Scarff-Bloom-Richardson grading system, Nottingham Modification. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. |
G | G Definition |
---|---|
GX | Grade cannot be assessed. |
G1 | Low nuclear grade. |
G2 | Intermediate nuclear grade. |
G3 | High nuclear grade. |
aReprinted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. |
There are three stage group tables for invasive cancer:
In the United States, cancer registries and clinicians must use the Clinical and Pathological Prognostic Stage Group tables for reporting. It is expected that testing is performed for grade, HER2, ER, and PR status and that results are reported for all cases of invasive cancer in the United States.
Stage | TNM |
---|---|
0 | Tis, N0, M0 |
IA | T1, N0, M0 |
IB | T0, N1mi, M0 |
T1, N1mi, M0 | |
IIA | T0, N1, M0 |
T1, N1, M0 | |
T2, N0, M0 | |
IIB | T2, N1, M0 |
T3, N0, M0 | |
IIIA | T0, N2, M0 |
T1, N2, M0 | |
T2, N2, M0 | |
T3, N1, M0 | |
T3, N2, M0 | |
IIIB | T4, N0, M0 |
T4, N1, M0 | |
T4, N2, M0 | |
IIIC | Any T (Tis, T1, T0, T2, T3, T4; N3, M0) |
IV | Any T (Tis, T1, T0, T2, T3, T4; Any N = N0, N1mi, N1, N2, N3, M1) |
T = primary tumor; N = regional lymph node; M = distant metastasis. | |
aAdapted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |
Notes: | |
1. T1 includes T1mi. | |
2. T0 and T1 tumors with nodal micrometastases (N1mi) are staged as Stage IB. | |
3. T2, T3, and T4 tumors with nodal micrometastases (N1mi) are staged using the N1 category. | |
4. M0 includes M0(i+). | |
5. The designation pM0 is not valid; any M0 is clinical. | |
6. If a patient presents with M1 disease before receiving neoadjuvant systemic therapy, the stage is Stage IV and remains Stage IV regardless of response to neoadjuvant therapy. | |
7. Stage designation may be changed if postsurgical imaging studies reveal the presence of distant metastases, provided the studies are performed within 4 months of diagnosis in the absence of disease progression, and provided the patient has not received neoadjuvant therapy. | |
8. Staging following neoadjuvant therapy is denoted with a yc or ypn prefix to the T and N classification. There is no anatomic stage group assigned if there is a complete pathological response (pCR) to neoadjuvant therapy, for example, ypT0, ypN0, cM0. |
The Clinical Prognostic Stage is used for clinical classification and staging of patients in the United States with invasive breast cancer. It uses TNM information based on the patient’s history, physical examination, imaging results (not required for clinical staging), and biopsies.
TNM | Grade | HER2 Status | ER Status | PR Status | Stage Group |
---|---|---|---|---|---|
Tis, N0, M0 | Any (refer to Table 6 and Table 7) | Any | Any | Any | 0 |
T1b, N0, M0 | G1 | Positive | Positive | Positive | IA |
Negative | IA | ||||
T0, N1mi, M0 | Negative | Positive | IA | ||
Negative | IA | ||||
T1b, N1mi, M0 | Negative | Positive | Positive | IA | |
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IB | ||||
G2 | Positive | Positive | Positive | IA | |
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IA | ||||
Negative | Positive | Positive | IA | ||
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IB | ||||
G3 | Positive | Positive | Positive | IA | |
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IA | ||||
Negative | Positive | Positive | IA | ||
Negative | IB | ||||
Negative | Positive | IB | |||
Negative | IB | ||||
T0, N1c, M0; T1b, N1c, M0; T2, N0, M0 | G1 | Positive | Positive | Positive | IB |
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIA | ||||
Negative | Positive | Positive | IB | ||
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIA | ||||
G2 | Positive | Positive | Positive | IB | |
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIA | ||||
Negative | Positive | Positive | IB | ||
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIB | ||||
G3 | Positive | Positive | Positive | IB | |
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIA | ||||
Negative | Positive | Positive | IIA | ||
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIB | ||||
T2, N1d, M0; T3, N0, M0 | G1 | Positive | Positive | Positive | IB |
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIB | ||||
Negative | Positive | Positive | IIA | ||
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIB | ||||
G2 | Positive | Positive | Positive | IB | |
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIB | ||||
Negative | Positive | Positive | IIA | ||
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIIB | ||||
G3 | Positive | Positive | Positive | IB | |
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIB | ||||
Negative | Positive | Positive | IIB | ||
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIB | ||||
T0, N2, M0; T1b, N2, M0; T2, N2, M0; T3, N1d, M0; T3, N2, M0 | G1 | Positive | Positive | Positive | IIA |
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIA | ||||
Negative | Positive | Positive | IIA | ||
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIB | ||||
G2 | Positive | Positive | Positive | IIA | |
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIA | ||||
Negative | Positive | Positive | IIA | ||
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIB | ||||
G3 | Positive | Positive | Positive | IIB | |
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIA | ||||
Negative | Positive | Positive | IIIA | ||
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIC | ||||
T4, N0, M0; T4, N1d, M0; T4, N2, M0; Any T, N3, M0 | G1 | Positive | Positive | Positive | IIIA |
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIB | ||||
Negative | Positive | Positive | IIIB | ||
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIC | ||||
G2 | Positive | Positive | Positive | IIIA | |
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIB | ||||
Negative | Positive | Positive | IIIB | ||
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIC | ||||
G3 | Positive | Positive | Positive | IIIB | |
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIB | ||||
Negative | Positive | Positive | IIIB | ||
Negative | IIIC | ||||
Negative | Positive | IIIC | |||
Negative | IIIC | ||||
Any T, Any N, M1 | Any (refer to Table 6 and Table 7) | Any | Any | Any | IV |
T = primary tumor; N = regional lymph node; M = distant metastasis. | |||||
aAdapted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |||||
bT1 includes T1mi. | |||||
cN1 does not include N1mi. T1, N1mi, M0, and T0, N1mi, M0 cancers are included for prognostic staging with T1, N0, M0 cancers of the same prognostic factor status. | |||||
dN1 includes N1mi. T2, T3, and T4 cancers and N1mi are included for prognostic staging with T2, N1; T3, N1; and T4, N1, respectively. | |||||
Notes: | |||||
1. Because N1mi categorization requires evaluation of the entire node, and cannot be assigned on the basis of an fine-needle aspiration or core biopsy, N1mi can only be used with Clinical Prognostic Staging when clinical staging is based on a resected lymph node in the absence of resection of the primary cancer, such as in the situation where sentinel node biopsy is performed before receiving neoadjuvant chemotherapy or endocrine therapy. | |||||
2. For cases with lymph node involvement with no evidence of primary tumor (e.g., T0, N1, etc.) or with breast ductal carcinoma in situ (e.g.,Tis, N1, etc.), the grade, human epidermal growth factor receptor 2 (HER2), estrogen receptor, and progesterone receptor information from the tumor in the lymph node should be used for assigning stage group. | |||||
3. For cases where HER2 is determined to be equivocal by in situ hybridization (fluorescence in situ hybridization or chromogenic in situ hybridization) testing under the 2013 American Society of Clinical Oncologists/College of American Pathologists HER2 testing guidelines, the HER2-negative category should be used for staging in the Pathological Prognostic Stage Group table. | |||||
4. The prognostic value of these Prognostic Stage Groups is based on populations of persons with breast cancer that have been offered and mostly treated with appropriate endocrine and/or systemic chemotherapy (including anti–HER2 therapy). |
The Pathological Prognostic Stage applies to patients with invasive breast cancer initially treated with surgery. It includes all information used for clinical staging, surgical findings, and pathological findings following surgery to remove the tumor. Pathological Prognostic Stage is not used for patients treated with neoadjuvant therapy before surgery to remove the tumor.
TNM | Grade | HER2 Status | ER Status | PR Status | Stage Group |
---|---|---|---|---|---|
Tis, N0, M0 | Any (refer to Table 6 and Table 7) | Any | Any | Any | 0 |
T1b, N0, M0; T0, N1mi, M0; T1b, N1mi, M0 | G1 | Positive | Positive | Positive | IA |
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IA | ||||
Negative | Positive | Positive | IA | ||
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IA | ||||
G2 | Positive | Positive | Positive | IA | |
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IA | ||||
Negative | Positive | Positive | IA | ||
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IB | ||||
G3 | Positive | Positive | Positive | IA | |
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IA | ||||
Negative | Positive | Positive | IA | ||
Negative | IA | ||||
Negative | Positive | IA | |||
Negative | IB | ||||
T0, N1c , M0; T1b, N1c, M0; T2, N0, M0 | G1 | Positive | Positive | Positive | IA |
Negative | IB | ||||
Negative | Positive | IB | |||
Negative | IIA | ||||
Negative | Positive | Positive | IA | ||
Negative | IB | ||||
Negative | Positive | IB | |||
Negative | IIA | ||||
G2 | Positive | Positive | Positive | IA | |
Negative | IB | ||||
Negative | Positive | IB | |||
Negative | IIA | ||||
Negative | Positive | Positive | IA | ||
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIA | ||||
G3 | Positive | Positive | Positive | IA | |
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIA | ||||
Negative | Positive | Positive | IB | ||
Negative | IIA | ||||
Negative | Positive | IIA | |||
Negative | IIA | ||||
T2, N1c, M0; T3, N0, M0 | G1 | Positive | Positive | Positive | IA |
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIB | ||||
Negative | Positive | Positive | IA | ||
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIB | ||||
G2 | Positive | Positive | Positive | IB | |
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIB | ||||
Negative | Positive | Positive | IB | ||
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIB | ||||
G3 | Positive | Positive | Positive | IB | |
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIB | ||||
Negative | Positive | Positive | IIA | ||
Negative | IIB | ||||
Negative | Positive | IIB | |||
Negative | IIIA | ||||
T0, N2, M0; T1b, N2, M0; T2, N2, M0, T3, N1d, M0; T3, N2, M0 | G1 | Positive | Positive | Positive | IB |
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIA | ||||
Negative | Positive | Positive | IB | ||
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIA | ||||
G2 | Positive | Positive | Positive | IB | |
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIA | ||||
Negative | Positive | Positive | IB | ||
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIB | ||||
G3 | Positive | Positive | Positive | IIA | |
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIA | ||||
Negative | Positive | Positive | IIB | ||
Negative | IIIA | ||||
Negative | Positive | IIIA | |||
Negative | IIIC | ||||
T4, N0, M0; T4, N1d, M0; T4, N2, M0; Any T, N3, M0 | G1 | Positive | Positive | Positive | IIIA |
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIB | ||||
Negative | Positive | Positive | IIIA | ||
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIB | ||||
G2 | Positive | Positive | Positive | IIIA | |
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIB | ||||
Negative | Positive | Positive | IIIA | ||
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIC | ||||
G3 | Positive | Positive | Positive | IIIB | |
Negative | IIIB | ||||
Negative | Positive | IIIB | |||
Negative | IIIB | ||||
Negative | Positive | Positive | IIIB | ||
Negative | IIIC | ||||
Negative | Positive | IIIC | |||
Negative | IIIC | ||||
Any T, Any N, M1 | Any (refer to Table 6 and Table 7) | Any | Any | Any | IV |
T = primary tumor; N = regional lymph node; M = distant metastasis. | |||||
aAdapted with permission from AJCC: Breast, revised version. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 4–96. | |||||
bT1 includes T1mi. | |||||
cN1 does not include N1mi. T1, N1mi, M0 and T0, N1mi, M0 cancers are included for prognostic staging with T1, N0, M0 cancers of the same prognostic factor status. | |||||
dN1 includes N1mi. T2, T3, and T4 cancers and N1mi are included for prognostic staging with T2, N1; T3, N1; and T4, N1, respectively. | |||||
Notes: | |||||
1. For cases with lymph node involvement with no evidence of primary tumor (e.g., T0, N1, etc.) or with breast ductal carcinoma in situ (e.g.,Tis, N1, etc.), the grade, human epidermal growth factor receptor 2 (HER2), estrogen receptor, and progesterone receptor information from the tumor in the lymph node should be used for assigning stage group. | |||||
2. For cases where HER2 is determined to be equivocal by in situ hybridization (fluorescence in situ hybridization or chromogenic in situ hybridization) testing under the 2013 American Society of Clinical Oncologists/College of American Pathologists HER2 testing guidelines, the HER2-negative category should be used for staging in the Pathological Prognostic Stage Group table. | |||||
3. The prognostic value of these Prognostic Stage Groups is based on populations of persons with breast cancer that have been offered and mostly treated with appropriate endocrine and/or systemic chemotherapy (including anti–HER2 therapy). |
早期、局限性或可手术的乳腺癌的标准治疗方案可能包括以下内容:
I期、II期、IIIA期和可手术的IIIC期乳腺癌通常需要采用多种方法治疗。诊断性活检和作为主要治疗手段的外科手术应作为两个独立的步骤来完成:
为了指导辅助治疗的选择,需要考虑许多因素,包括肿瘤的分期、分级和分子状态(如ER、PR、HER2/neu或三阴性状态)。
局部治疗方法的选择取决于以下方面:
原发性肿瘤的手术治疗方法包括以下方面:
腋窝的手术分期也应该进行。
上述任何一种选择具有相同的生存率,这是欧洲癌症研究与治疗组织(EORTC)的试验(EORTC-10801)和其他前瞻性随机试验的结果证实的。
同时,一项针对753例患者进行的回顾性研究中,根据激素受体状态将这些患者分为三组(ER阳性或PR阳性;ER阴性及PR阴性,但HER2阳性;三阴性),结果发现,经过标准保乳手术治疗的患者的乳腺疾病控制无差异;然而,目前还没有实质性的数据来支持这一发现。
保乳治疗后乳房的局部复发率较低,并且随所用手术术式(例如,肿块切除术,象限切除术,乳房部分切除术等)而略有不同。 是否需要完全清楚的微观切缘目前仍有争议。
然而,在一个包含33个研究(N=28162例患者)的荟萃分析中,一个多学科小组共识最近使用切缘宽度和同侧乳房肿瘤复发作为一个新共识的主要证据基础,该共识针对的是切缘在接受保乳手术加放射治疗的I期和II期乳腺癌患者。该荟萃分析的结果如下:
对于接受部分乳房切除术的患者,初次手术后切缘如果呈阳性,常常会需要二次手术切除。一项包含235例进行部分乳房切除术的0-III期乳腺癌患者的临床试验,伴或不伴选择性的边缘切除,随机地分配患者进行额外的残腔边缘切除(剃除组)或不进行(非剃除组)。
剃除组患者的切缘阳性率明显低于不剃除组(19% vs.34%,P=0.01),且无瘤切缘的二次手术率更低(10 vs. 21%,P=0.02)。
[证据等级:1iiDiv]
腋窝淋巴结状态仍然是乳腺癌患者预后最重要的预测因素。在大多数浸润性乳腺癌患者中,没有足够的证据表明可以忽略淋巴结分期。一些研究已经尝试定义一个新的分组,其中淋巴结转移的可能性低到可以不行腋窝淋巴结活检。在那些单中心的病例中,T1a肿瘤患者中阳性淋巴结的发生率在9%到16%之间。
另一个研究发现,未进行腋窝淋巴结清扫(ALND)的T1a肿瘤患者的腋窝淋巴结复发率为2%。
[证据等级:3iiiA]
腋窝淋巴结分期有助于判断预后和治疗。SLN活检是在浸润性乳腺癌患者中进行的标准腋窝分期步骤的第一步。SLN被定义为原发肿瘤引流区域淋巴结中的特殊淋巴结,是原发肿瘤发生淋巴结转移所必经的第一批淋巴结;因此,SLN数量往往多于1枚。研究表明,在肿瘤周围或活检腔内或在乳晕区域注入锝Tc99m-标记的硫胶体、或蓝色染料或两者同时注射,引流至腋窝,使得SLN可以在92%至98%的患者中被识别出来。这些报告显示SLN活检和全腋窝淋巴结清扫(ALND)结果的一致性高达97.5%-100%。
由于以下证据,SLN活检是浸润性乳腺癌的标准腋窝手术分期步骤。与腋窝淋巴结清扫术相比,单独进行SLN活检的并发症发病率更低。
证据(SLN活检):
由于以下试验结果,对于有限的SLN阳性乳腺癌患者,通过保乳术或乳房切除术,放疗和全身疗法,无需进行ALND。
证据(对于有限的SLN阳性乳腺癌患者,SLN活检阳性后进行ALND)):
对于需要行ALND的患者,标准的评估通常只涉及I站和II站淋巴结清扫,因此可以切除符合要求数量的淋巴结以进行评估(即至少6-10枚),以降低术后复发率。
对于选择全乳切除的患者,乳房重建手术可与乳房切除手术同步进行(即即刻重建)或在后续的某个时间进行(即延迟重建)。
乳房轮廓可以通过以下方式恢复:
乳房重建后,可以在辅助治疗或局部复发时进行胸壁和区域淋巴结的放疗。 乳房假体重建后的放射治疗可能会影响美观,并且可能会增加包膜纤维化,疼痛或需要假体取出的发生率。
保乳手术后应常规接受放射治疗。放射治疗也适用于乳腺切除术后的高危患者。辅助放射治疗的主要目的是消除残余病灶,从而减少局部复发。
对于接受保乳手术而未接受放射治疗的女性患者,即使腋窝淋巴结阴性,残留乳房的肿瘤复发风险也相当大(>20%)。
虽然所有评估放射治疗在保乳治疗中的作用的试验均显示,局部复发率在统计学上有显著降低,但没有一项试验显示死亡率在统计学上有显著降低。然而,一项大型的荟萃分析显示,保乳联合放疗可显著降低乳腺癌的复发和死亡风险。
因此,证据支持在保乳手术后使用全乳放射治疗。
证据(保乳手术后进行放疗):
早期乳腺癌试验者协同组(EBCTCG)在2011年对17项临床试验进行了荟萃分析,该分析包括了2万多名早期乳腺癌患者,其结果支持保乳手术后行全乳放疗。
[证据等级:1iiA]
考虑到放射剂量和放疗计划,以下方面应被注意:
需要进行更多的研究以确定短程分割放疗是否适用于具有淋巴结瘤荷高的女性。
对累及淋巴结的患者,乳房切除术后常规行区域淋巴结照射;然而,对于接受保乳手术和全乳照射的患者,其作用尚不明确。一项针对1832名女性的随机试验(NCT00005957)显示,保乳手术后局部淋巴结照射和全乳照射可降低复发风险(10年DFS为 82.0% vs.77.0%;HR为0.76;95% CI为0.61-0.94;P =0.01),但不影响生存率(10年OS为 82.8% vs.81.8%;HR为0.91;95% CI为0.72-1.13;P=0.38)。
[证据等级:1iiA]
EORTC试验(NCT00002851)也报告了类似的结果。对于有或无腋窝淋巴结受累的中央或中位原发性肿瘤,或有腋窝淋巴结受累的外位肿瘤的女性,被随机分配接受全乳或胸壁照射,另外进行或不进行区域性淋巴结照射。研究人群中76.1%进行了保乳手术,其余进行了乳房切除术。与未接受局部淋巴结放射治疗的患者相比,接受局部淋巴结放射治疗的患者的OS在10年内未见改善(82.3% vs. 80.7%,P=0.06)。接受局部淋巴结照射的患者与未接受局部淋巴结照射的患者相比,远处DFS有改善(78% vs.75%,P=0.02)。
[证据等级:1iiA]
综合上述两项试验结果的荟萃分析发现,OS上存在微小的统计学显著差异 (HR为0.88;95%置 CI为0.78-0.99;P=0.034;5年时的绝对差异为1.6%)。
术后胸壁及区域淋巴结辅助放疗常规应用于乳房切除术后局部复发风险高的患者。局部复发风险最高的患者有以下一种或多种情况:
在高危人群中,放疗可以减少局部复发,即使是那些接受辅助化疗的患者。
没有任何高危因素的1-3个受累淋巴结的患者局部复发的风险较低,常规辅助放疗在这种情况下的应用价值尚不明确。
证据(1-3个受累淋巴结患者术后放疗):
此外,一项对NSABP试验的分析显示,即使在原发肿瘤较大(>5cm)和腋窝淋巴结阴性的患者中,孤立的局部复发的风险也很低(7.1%),以至于没有必要进行常规的局部放疗。
保乳术后辅助化疗和放疗的最佳顺序已被研究。根据以下研究,在保乳手术后将放疗推迟几个月直至辅助化疗结束,似乎对整体预后没有不利影响。此外,在保乳手术后不久开始化疗,对于有远处转移风险的患者可能是更好的选择。
证据(术后放疗时机):
观察结果如下:
这些研究表明,术后2至7个月延迟放疗对局部复发率没有影响。这些发现在一项荟萃分析中得到了证实。
[证据等级:1iiA]
在一项在接受治疗的HER2/neu阳性乳腺癌患者中开展的、以评估加入曲妥珠单抗获益性的III期临床试验的额外分析中,发现同时接受辅助放疗和曲妥珠单抗的患者的急性不良事件或心脏事件发生率没有增加。
因此,应用曲妥珠单抗的同时,进行放射治疗似乎是安全的,并且避免了放疗开始后的额外延迟。
放射治疗的远期毒性反应是罕见的,可以通过目前的放射传输技术和仔细描绘靶区来最小化其毒性反应。放射的远期效应包括以下方面:
现代放射治疗技术在20世纪90年代被引入,当对左侧胸壁或左侧乳房放射时,可使深层心肌的接受的辐射最小化。心源性死亡率也随之降低。
一个从1973年到1989年美国国家癌症研究所的监测、流行病学、最终结果项目(SEER)数据的分析回顾了接受乳房或胸壁放射的女性的由缺血性心脏病引起的死亡人数,该结果显示,自1980年以来,未发现接受左侧乳房或胸壁放射的女性死于缺血性心脏病的比率增加。
[证据等级:3iB]
肿瘤分期和分子分型决定了是否需要辅助全身治疗及应该选择何种治疗方案。例如,激素受体(ER和/或PR)阳性的患者将接受内分泌治疗。HER2过表达患者应接受辅助曲妥珠单抗治疗,往往还需联合化疗。如果既无HER2过表达,也无激素受体表达,则辅助治疗依赖于化疗,通常还需联合试验性靶向治疗。
一个国际共识小组提出了一个风险分类系统和系统性治疗方案。
这种经过一些修改的分类如下:
亚型 | 治疗方案 | 注释 |
---|---|---|
管状A样 | ||
—激素受体阳性 | 大多数情况下单纯内分泌治疗 | 考虑化疗,如果: |
—HER2阴性 | —高肿瘤负荷(≥4枚阳性淋巴结, T3或更高) | |
–PR>20% | ||
–Ki67水平低 | –3级 | |
管状B样 | ||
—激素受体阳性 | 大多数情况下内分泌治疗加化疗 | |
—HER2阴性 | ||
—Ki67水平高或PR低 | ||
HER2阳性 | 化疗加抗HER2治疗 | 如果激素受体阳性,应采用内分泌治疗 |
肿瘤小且淋巴结阴性可以考虑不进行化疗和抗HER2治疗 | ||
三阴 | 化疗 | 肿瘤小且淋巴结阴性可以考虑不进行化疗 |
HER2=人表皮生长因子受体2;LN =淋巴结;PR=孕激素受体。 | ||
a 来自Senkus等人的修订。 |
最恰当选择治疗方案的基础是了解个体肿瘤复发风险与辅助治疗的短期和长期风险之间的平衡。这种方法允许临床医生帮助患者确定预期的治疗效果是否符合他们的实际情况。以下所述的治疗方案应根据患者和肿瘤的特征来确定。
患者组 | 治疗方案 |
---|---|
绝经前,激素受体阳性(ER或PR) | 无额外的治疗 |
他莫昔芬 | |
他莫昔芬+化疗 | |
卵巢功能抑制+他莫昔芬 | |
卵巢功能抑制+芳香化酶抑制剂 | |
绝经前,激素受体阴性(ER或PR) | 无额外的治疗 |
化疗 | |
绝经后,激素受体阳性(ER或PR) | 无额外的治疗 |
芳香化酶抑制剂或他莫昔芬治疗序贯芳香化酶抑制剂,联合或不联合化疗 | |
绝经后,激素受体阴性(ER或PR) | 无额外的治疗 |
化疗 | |
ER=雌激素受体;PR=孕激素受体。 |
EBCTCG荟萃分析分析了从1976年开始至1989年的11项临床试验,在这些试验中,患者被随机分配接受含有蒽环类药物(如阿霉素或表阿霉素)或CMF(环磷酰胺、甲氨蝶呤和5-FU)治疗方案。比较CMF方案和含蒽环类药物方案的综合分析结果表明,含蒽环类药物的方案对绝经前和绝经后女性都有轻微的优势。
证据(蒽环类方案):
研究结果表明,肿瘤的特征(即淋巴结阳性,HER2/neu过表达型乳腺癌)可能预示着对蒽环类药物的反应性。
证据(HER2/neu扩增的女性患者以蒽环类药物为主的治疗方案):
有几项试验研究了在淋巴结阳性乳腺癌患者,提出在以不含蒽环类药物为基础的辅助化疗方案中添加紫杉类药物(紫杉醇或多西他赛)可以获益。
证据(在以蒽环类药物为基础的方案中添加一种紫杉类药物):
东部肿瘤协作组牵头的组间试验(E1199[NCT00004125])共纳入4950名受试患者,在标准剂量的AC化疗后,对两种方案(单周方案和3周方案)两种药物(多西他赛和紫杉醇)进行比较。
[证据水平:1iiA] 研究结果包括:
历史上,乳腺癌的辅助化疗是每3周进行一次。有研究试图确定减少化疗周期之间的持续时间是否可以改善临床结果。这些研究的总体结果支持对HER2阴性的乳腺癌患者使用剂量密集型化疗。
证据(HER2阴性乳腺癌患者给予高剂量化疗):
多西他赛和环磷酰胺是一种可接受的辅助化疗方案。
证据(多西他赛和环磷酰胺):
开始辅助治疗的最佳时间是不确定的。一项回顾性观察性研究报告如下:
辅助化疗与几种典型的毒性效应有关,这些毒性效应因不同治疗方案中使用的不同药物而不同。常见的毒性效应包括:
严重但不常见的毒性效应包括以下方面:
(请参阅PDQ总结中与治疗有关的恶心及呕吐的内容;有关粘膜炎的信息,请参阅PDQ总结中的化疗和头颈放疗的口腔并发症;有关过早绝经相关症状的信息,请参阅PDQ总结中关于潮热和盗汗的内容。)
然而,使用含蒽环类药物的方案,尤其是那些含有环磷酰胺剂量增加的方案,与5年患急性白血病的累积风险0.2%-1.7%有关。
接受高累积剂量表柔比星(>720mg/m2)和环磷酰胺(>6300mg/m2)的患者,这一风险会增加到4%以上。
据报道,认知障碍发生在一些化疗方案实施后。
然而,关于这一主题的前瞻性随机研究的数据是缺乏的。
EBCTCG荟萃分析显示,接受辅助联合化疗的女性患对侧乳腺癌的年发病率确实降低了20%(标准偏差= 10)。
这种小的比例降低转化为绝对收益,该收益在统计学上微不足道,表明化疗并未增加对侧疾病的风险。此外,该分析表明,在随机分配接受化疗的所有妇女中,归因于其他癌症或血管原因的死亡在统计学上的没有显着增加。
对于HER2/neu阴性的乳腺癌,没有一种辅助化疗方案被认为是标准的或优于其他方案的。首选的方案因机构、地理区域和临床医生的不同而不同。
一些关于辅助化疗益处的最重要数据来自EBCTCG,它每5年回顾分析一次全球乳腺癌试验的数据。在2011年EBCTCG荟萃分析中,使用以蒽环类为基础的辅助化疗方案与未治疗相比,复发风险有显著改善(RR为0.73;95% CI,0.68-0.79),乳腺癌死亡率显著降低(RR为0.79;95% CI,0.72-0.85),总体死亡率显著降低(RR为0.84;95% CI,0.78-0.91),即绝对生存获益为5%。
TNBC被定义为ER、PR和HER2/neu阴性。TNBC对一些最有效的乳腺癌治疗方法不敏感,包括HER2靶向治疗(如曲妥珠单抗)和内分泌治疗(如他莫昔芬或芳香化酶抑制剂)。
剂量-密集或节拍方案联合细胞毒药物化疗,仍是早期TNBC的标准治疗。
证据(TNBC的剂量-密集或节拍方案的新辅助化疗):
铂类药物已成为治疗TNBC的重要药物。然而,在临床试验之外,将它们添加到早期TNBC的治疗中还没有确定的作用。一项试验对28名II期或III期TNBC女性进行了4个周期的顺铂新辅助治疗,结果pCR率为22%。
[证据水平:3iiiDiv]一项随机临床试验CALGB-40603 (NCT00861705),评估了在新辅助方案下,紫杉醇、阿霉素加环磷酰胺化疗方案中加入卡铂的益处。Triple Negative试验(NCT00532727)评估在存在转移的情况下,卡铂与多西他赛的疗效。这些试验将有助于确定铂类药物治疗TNBC的作用。
PARP抑制剂对BRCA突变患者和TNBC患者的疗效正在临床试验中被评估。
PARPs是一组参与多种细胞进程的酶,其中包括DNA修复。由于TNBC与BRCA突变的乳腺癌具有多种相似的临床病理特征,即庇护功能失调的DNA修复机制,因此,PARP抑制剂与依赖于BRCA机制的DNA修复缺失相结合,可能会导致合成致死和细胞死亡的增加。
HER2阳性的早期乳腺癌的标准治疗是一年的辅助曲妥珠单抗治疗。
一些III期临床试验探讨了抗HER2/neu抗体曲妥珠单抗作为HER2过表达癌症患者辅助治疗的作用。研究结果证实了辅助曲妥珠单抗治疗12个月的疗效。
证据(曲妥珠单抗治疗时长):
Herceptin Adjuvant(HERA)(BIG-01-01[NCT00045032])试验检测了在基础治疗结束后使用曲妥珠单抗作为HER2阳性乳腺癌的辅助治疗是否有效。对大多数患者来说,基础治疗包括术前或术后给予含蒽环类药物的化疗方案,加上或不加局部放疗。曲妥珠单抗在基础治疗结束后7周内每3周给予一次。
[证据水平:1iiA]患者被随机分配到三个研究组中的其中一个:
1年曲妥珠单抗组与观察组比较,患者中位年龄为49岁,约33%的患者为淋巴结阴性,接近50%的患者为激素受体(ER、PR)阴性。
一些研究评估了曲妥珠单抗皮下注射在新辅助治疗和辅助治疗中的适用性。
许多研究报告了与辅助曲妥珠单抗相关的心脏事件。主要研究结果包括:
拉帕替尼是一种小分子酪氨酸激酶抑制剂,能够同时抑制表皮生长因子受体和HER2。没有数据支持使用拉帕替尼作为早期HER2/neu阳性乳腺癌辅助治疗的一部分。
证据(反对使用拉帕替尼治疗HER2阳性的早期乳腺癌):
帕妥珠单抗是一种人源化的单克隆抗体,可与HER2受体胞外区域的一个独特的位点结合,能够抑制二聚作用。它与曲妥珠单抗的联合应用已经在术后的随机试验中进行了评估。
证据(帕妥珠单抗):
来那替尼是HER1、HER2和HER4的不可逆酪氨酸激酶抑制剂,已被FDA批准用于早期HER2阳性乳腺癌患者的辅助治疗,以辅助曲妥珠单抗治疗。
证据(来那替尼):
以下有关激素受体阳性疾病女性治疗的章节中展示的许多证据已被一份美国临床肿瘤学协会的指南考虑到,该指南中描述了对这些患者管理的几种治疗方案。
5年的辅助内分泌治疗已被证明可以显著降低局部和远处复发、对侧乳腺癌和乳腺癌死亡的风险。
内分泌治疗的最佳治疗时长尚不清楚,有大量证据支持至少5年的内分泌治疗。一个对88项临床试验的荟萃分析显示,经过5年的内分泌治疗后,62,923名激素受体阳性的乳腺癌患者在诊断后5至20年内有稳定的晚期复发风险。
[证据水平:3iiiD]远处复发的风险与原发肿瘤(T)和淋巴结(N)状态相关,风险范围为10%-41%。
他莫昔芬已被证明对激素受体阳性的乳腺癌女性有益。
证据(他莫昔芬用于激素受体阳性的早期乳腺癌):
EBCTCG荟萃分析和一些大型随机试验已经确定了他莫昔芬的最佳使用时间。
10年的他莫昔芬治疗已被证明优于较短时间的他莫昔芬治疗。
证据(他莫昔芬治疗时间):
ATLAS试验的结果表明,对于服用他莫昔芬5年仍未绝经的女性,继续服用他莫昔芬5年是有益的。
服用他莫昔芬5年后进入绝经期的女性也可以用AIs进行治疗。(更多信息,请参阅本总结中激素受体阳性治疗部分的芳香化酶抑制剂部分内容。)
由于EBCTCG分析的结果,在接受辅助化疗的女性中使用他莫昔芬并不会减弱化疗的益处。
然而,与顺序给药相比,他莫昔芬与化疗同时使用的效果较差。
有证据表明,单纯的卵巢去势并不能有效替代其他的全身疗法。
此外,在化疗和/或他莫昔芬治疗中加入卵巢去势并没有发现预后的明显改善。
证据(他莫昔芬+卵巢抑制):
尽管初步结果总体呈阴性,但亚组分析显示,卵巢抑制对化疗后仍未绝经的女性有益处。然而,8年的随访结果并没有根据是否给予化疗而显示出治疗效果的异质性,尽管在接受化疗的患者中复发更为常见。
在卵巢功能被抑制或切除的绝经前女性中,AIs与他莫昔芬进行了比较。这些研究的结果是相互矛盾的。
证据(绝经前女性AI与他莫昔芬的比较):
在绝经后女性中,AIs与他莫昔芬的顺序使用或作为其替代品已成为多项研究的主题,其结果已在个体患者水平的荟萃分析中进行了总结。
证据(AI vs.他莫昔芬作为绝经后女性的初始治疗):
一些试验和荟萃分析已经检验了在服用他莫昔芬2至3年后改用阿那曲唑或依西美坦完成5年内分泌治疗的效果。
如下所述的证据表明,序贯服用他莫昔芬和AI优于连续服用他莫昔芬5年。
证据(序贯他莫昔芬和AI vs.他莫昔芬 5年):
在一项荟萃分析中,包括来自6个试验的11798名患者,含AI组的10年复发率从19%降至17%(RR, 0.82;95%CI,0.75-0.91;P=0.001)。他莫昔芬组的10年总死亡率为17.5%,而含AI组的10年总死亡率为14.6% (RR,0.82;95%CI,0.73-0.91;P=0.0002)。
[证据等级:1A]
证据表明序贯使用他莫昔芬和AI5年相比单纯使用AI5年并没有获益。
证据(他莫昔芬和AI的序贯使用 vs.AI 5年):
在包括来自试验的12779名患者的荟萃分析中,接受5年AI治疗的组中的7年复发率从14.5%略微降低到13.8%(RR,0.90;95%CI,0.81-0.99;P=0.045)。7年的总死亡率,他莫昔芬后AI组为9.3%,单纯AI组为8.2%(RR,0.89;95%CI,0.78-1.03;P=0.11)。
[证据等级:1A]
如下所述的证据表明,在服用他莫昔芬5年后改用AI比当时停用他莫昔芬要好。
AI治疗的最佳时间是不确定的,且多项试验评估的疗程超过5年。
关于初始AI辅助治疗5年后,继续延长内分泌治疗的证据:
双膦酸盐和地诺单抗均已被评估为早期乳腺癌的辅助疗法;然而,这些药物作为早期乳腺癌辅助治疗的作用尚不清楚。与地诺单抗相比,支持二膦酸盐的证据数量更多,并且有证据支持乳腺癌死亡率-一个更具临床相关性的终点。
证据(二膦酸盐治疗早期乳腺癌):
正在进行的III期试验(NCT01077154)正在研究II期和III期乳腺癌中骨修饰剂地诺单抗的效果。
术前化疗,也称为新辅助化疗,传统上用于局部晚期乳腺癌患者,以减少肿瘤体积达到可手术切除的目的。此外,术前化疗也适用于一些原发性可手术二期或三期乳腺癌患者。2005年进行的多中心随机临床试验的荟萃分析表明,给与新辅助化疗和辅助化疗相同的治疗量,新辅助化疗与辅助化疗具有相同的DFS和OS。
[证据等级:1iiA]
2019年,早期乳腺癌试验者合作小组使用来自10个试验的4756名妇女的个体患者数据进行了荟萃分析,这些试验比较了新辅助化疗和辅助化疗中给予的相同方案。
与辅助治疗相比,新辅助治疗与保乳率增加相关(65%对49%)。新辅助化疗和辅助治疗在远处复发、乳腺癌死亡率或任何原因引起的死亡方面没有差异;然而,新辅助治疗与更高的15年局部复发率相关(21.4%对15.9%;比率,1.37;95% CI,1.17-1.61;P=.001)。
[证据等级:1iiA]
当前关于术前化疗的共识意见建议以蒽环类和紫杉类为基础的治疗,前瞻性研究表明,术前以蒽环类和紫杉烷为基础的治疗比其他方案(例如,单独使用蒽环类)具有更高的缓解率。
[证据等级:1iiDiv]
术前全身治疗的潜在优势是,对于那些局部晚期,无法切除的疾病,明确的局部治疗成功的可能性增加。 通过增加保乳的可能性并提供获得pCR的预后信息,它还可为精心挑选的原发性可手术疾病患者提供益处。 在这些情况下,与残留大量残留疾病的情况相比,可以告知患者复发的风险非常低。
pCR已被用作乳腺癌术前临床试验中长期结果(如DFS、EFS和OS)的替代终点。一项研究(CTNeoBC)对11个术前随机试验(n = 11,955)进行了汇总分析,与仅从乳腺中根除浸润性肿瘤(ypT0/is)相比,pCR提供了更好的预后相关信息,pCR定义为乳腺和腋窝淋巴结中无残留浸润性癌,有或无原位癌(ypT0/is ypN0或ypT0 ypN0)。
pCR在本研究中不能作为改良EFS和OS的替代终点。
[证据等级: 3iiiD]由于pCR与乳腺癌更具侵袭性亚型的个体患者的显著改善的结果有很强的相关性,因此 FDA 支持将pCR作为高危早期乳腺癌患者术前临床试验的终点。
对于术前治疗后腋窝淋巴结组织学阴性的患者,无论术前治疗前的淋巴结状态如何,术后放射治疗也可以省略,从而可以根据患者的具体情况进行治疗。
这种方法的潜在缺点是术前化疗后无法确定准确的病理分期。但是,残留的肿瘤组织可以提供更个体化的预后信息。
由经验丰富的团队对接受术前治疗的患者进行多学科管理对于优化以下方面至关重要:
在开始术前治疗之前,应仔细评估肿瘤的组织学,分级和受体状态。 肿瘤具有纯小叶组织学,低级或高激素受体表达且HER2阴性状态的患者对化学疗法的反应较小,应考虑进行初次手术,尤其是当淋巴结临床阴性时。 即使在这些情况下在手术后进行辅助化疗,也可以避免采用第三代治疗方案(基于蒽环紫杉烷)。
在开始术前治疗之前,应评估乳房和区域淋巴结内的疾病程度。系统性疾病的分期可能包括以下内容:
当需要保乳治疗以确定肿瘤位置并排除多中心疾病时,进行基线乳腺成像。可疑异常通常在开始治疗前进行活检,并在乳腺肿瘤中心放置一个标记。在可能的情况下,在开始全身治疗之前,可以对可疑的腋窝淋巴结进行活检。
在接受术前治疗的患者中,前哨淋巴结(SLN)活检的最佳时机尚未确定。应考虑以下几点:
在考虑术前治疗时,治疗方案包括以下内容:
开始术前治疗后,必须定期对治疗反应进行临床评估。 如果保留乳房是外科手术的目标,则还需要重复影像评估。 在可行的情况下,术前治疗过程中疾病进展的患者可以过渡到非交叉耐药性方案或进行手术。
尽管改用非交叉耐药方案比继续进行相同的疗法可提高pCR率,但尚无明确证据表明采用这种方法可改善其他乳腺癌的预后。
早期试验检验了在辅助治疗中使用蒽环类药物的方案是否会延长术前用药的DFS和OS。证据支持术前使用蒽环类化疗方案比术后使用更高的保乳率,但是术前方案并没有改善生存率。
证据(术前蒽环类药物方案):
为了改善仅用AC观察到的结果,在化疗方案中加入了紫杉烷。以下研究结果支持在基于蒽环类药物的化疗方案中加入紫杉烷治疗HER2阴性乳腺肿瘤。
证据(基于蒽环类-紫杉烷的化疗方案):
然而,在三阴性乳腺癌(TNBC)患者中,联合使用卡铂的蒽环类-紫杉类化疗方案的化疗效果更好。 在将进行手术治疗前加用卡铂视为新的治疗标准之前,有必要进行生存终点的评估和确认生物标志物的反应或耐药性稳定性的研究。
证据(在TNBC患者蒽环类-紫杉烷类化疗方案中加入卡铂):
重要的是,在辅助治疗和转移治疗中的研究结果并未证明在化疗中加入贝伐单抗比单独化疗有OS益处。然而,术前化疗中加入贝伐单抗与pCR率增加以及毒性增加有关,如高血压、心脏毒性、手足综合征和粘膜炎(如NSABP B-40 [NCT00408408和GeparQuinto [NCT00567554])。
[证据等级: 1iiDiv]然而,尚不清楚观察到的适度益处是否会转化为长期生存优势。
辅助治疗成功后,第二阶段研究的初步报告显示,当曲妥珠单抗(一种结合HER2胞外区的单克隆抗体)加入术前蒽环类-紫杉类方案时,pCR率有所提高。
[证据等级:1iiDiv]这已经在第三阶段研究中得到证实。
证据(曲妥珠单抗):
曲妥珠单抗的皮下制剂也已获得批准。
安全性试验(NCT01566721)评估了在HER2阳性乳腺癌的一期至三期中,自我给药与临床医生给药SQ曲妥珠单抗的安全性和耐受性。
化疗同时或依次进行。
一项三期试验(HannaH [NCT 00950300])也证明了术前SQ曲妥珠单抗的药代动力学和疗效不低于静脉注射制剂。这项国际开放性试验(n=596)随机分配患有可手术、局部晚期或炎性HER2阳性乳腺癌的女性接受术前化疗(蒽环类-紫杉类为基础),术前每3周给予SQ或静脉注射曲妥珠单抗。患者接受辅助曲妥珠单抗以完成1年的治疗。
[证据等级:1iiD]各组间的pCR比率相差4.7%(95% CI,4.0-13.4);静脉给药组为40.7%,而SQ给药组为45.4%,表明SQ制剂的非劣效性。EFS和OS是次要终点。六年EFS在两个方面都是65%(HR,0.98;95% CI,0.74-1.29)。两组6年OS的比率为84%(HR,0.94;95% CI,0.61-1.45)。
新的HER2靶向疗法(lapatinib,pertuzumab)也进行了研究。看来HER2受体的双靶治疗使pCR率增高;然而,迄今为止,这种方法还没有显示出生存优势。
帕妥珠单抗是一种人源化单克隆抗体,可与HER2受体胞外域上的独特表位结合并抑制二聚化。 在两项术前临床试验中已对帕妥珠单抗联合曲妥珠单抗联合或不联合化疗进行了评估,以提高曲妥珠单抗和化疗观察到的pCR率。
证据(帕妥珠单抗):
观察到以下结果:
观察到以下结果:
由于这些研究,FDA加快批准使用帕妥珠单抗作为肿瘤大于2 cm或淋巴结阳性的HER2阳性早期乳腺癌女性术前治疗的一部分。
在证实性APHINDY(NCT 01358877)试验(一项针对HER2阳性乳腺癌女性的随机、三期辅助研究)的结果之后,对帕妥珠单抗的FDA批准随后转化为常规批准,该试验证明,与单独使用化疗和曲妥珠单抗相比,使用帕妥珠单抗加曲妥珠单抗的双HER2靶向治疗的组合,侵袭性DFS得到改善。
作为完整治疗方案的一部分,现已批准将帕妥珠单抗与曲妥珠单抗和化疗联合用于局部晚期、炎性或早期HER2阳性乳腺癌(大于2厘米或淋巴结阳性)的新辅助治疗,并与曲妥珠单抗联合化疗用于HER2阳性且具有高复发风险的早期乳腺癌的辅助治疗。
拉帕替尼是一种小分子激酶抑制剂,能够抑制表皮生长因子受体和HER2的双重受体。研究结果不支持术前使用拉帕替尼。
证据(反对使用拉帕替尼治疗HER2阳性的早期乳腺癌):
ALLTO (NCT00490139)三期临床试验提供了更明确的疗效数据,该试验将女性患者随机分配到曲妥珠单抗组或曲妥珠单抗加拉帕替尼组进行辅助治疗。
该试验没有达到它的主要终点DFS。在NeoALTTO试验中,在曲妥珠单抗中加入拉帕替尼后,pCR率增加了一倍,但在中位随访4.5年的ALTTO试验中,这并未转化为生存结果的改善。这表明目前拉帕替尼在术前或辅助治疗中没有作用。
使用Roche和Genentech提供的数据,对598名接受帕妥珠单抗治疗的癌症患者的心脏安全性进行了汇总分析。
[证据等级:3iiiD]
对随机试验(n=6)进行荟萃分析,评价抗HER2单一疗法(曲妥珠单抗或拉帕替尼或帕妥珠单抗)与双重抗HER2疗法(曲妥珠单抗加拉帕替尼或曲妥珠单抗加帕妥珠单抗)的疗效。
[证据等级:3iiiD]
对于激素受体阳性的绝经后女性乳腺癌患者,当因基础疾病或身体状态不是适合化疗时,术前内分泌治疗可能是一种选择。尽管术前激素治疗3至6个月的毒性特征是良好的,但获得的pCR率(1%–8%)远低于未选择人群的化疗报道。
[证据等级:1iDiv]
在这个病人群体中,可能需要更长的术前治疗时间。术前他莫昔芬的总有效率为33%,一些患者在治疗后12个月出现最大反应。
一项对不适于化疗的老年患者术前使用来曲唑4、8或12个月的随机研究表明,治疗持续时间越长,pCR率越高(17.5%对5%对2.5%,趋势的P值<.04)。
[证据等级:1iiDiv]
AIs也与他莫昔芬在术前治疗方面进行了比较。经过3至4个月的术前治疗后,AI组患者的总体客观缓解和保乳率均有统计学意义上的显著提高
或者与他莫昔芬相关的结果相当。
美国外科医师学会肿瘤组的一项试验目前正在比较术前使用阿那曲唑、来曲唑或依西美坦的疗效。
绝经前的激素反应性乳腺癌患者,术前应用内分泌治疗仍在研究中。
一项临床试验表明,对于术前化疗后未获得pCR的患者,使用卡培他滨作为辅助治疗是获益的。
证据(卡培他滨):
证据(T-DM1):
对于术前治疗后仍有残余病变的患者,应考虑采用这些方法并参与新疗法的临床试验。EA1131(NCT02445391)是一项随机的三期临床试验,该试验将术前治疗后残留基底样TNBC的患者随机分配接受铂类化疗或卡培他滨化疗。S1418/BR006(NCT02954874)是一项三期试验,旨在评估术前治疗后残留TNBC (≥1cm浸润性癌或残留结节)患者使用pembrolizumab作为辅助治疗的疗效。
大多数接受术前治疗的女性在保乳术后后进行放射治疗,以降低局部复发的风险。在决定是否实施术后放疗时,应考虑最初临床分期和随后的病理分期。
其他辅助全身治疗可以在术后、辅助放疗期间或完成后进行,包括对激素受体阳性乳腺癌患者的辅助激素治疗和对HER2阳性乳腺癌患者的辅助曲妥珠单抗治疗。(有关更多信息,请参考本摘要早期/局部/可手术乳腺癌部分的激素受体阳性乳腺癌小节。)
在完成一期、二期或三期乳腺癌的一期治疗后,随访的频率和筛查检测是否合适仍存在争议。
来自随机试验的证据表明,与常规体检相比,定期随访骨扫描、肝脏超声检查、胸部x线检查和肝功能血液检查并不能提高生存率或生活质量。
即使这些检查能够早期发现疾病复发,患者的生存率也不会因此受到影响。
基于这些数据,对于I - III期完成治疗的无症状乳腺癌患者,可接受的随访可以限于以下范围:
使用我们的高级临床试验搜索来查找NCI支持的癌症临床试验,这些试验现在正在招募患者。可以通过试验地点、治疗类型、药物名称和其他标准来缩小搜索范围。关于临床试验的一般信息也是可用的。
Standard treatment options for early, localized, or operable breast cancer may include the following:
Stages I, II, IIIA, and operable IIIC breast cancer often require a multimodal approach to treatment. The diagnostic biopsy and surgical procedure that will be used as primary treatment should be performed as two separate procedures:
To guide the selection of adjuvant therapy, many factors including stage, grade, and molecular status of the tumor (e.g., ER, PR, HER2/neu, or triple-negative status) are considered.
Selection of a local therapeutic approach depends on the following:
Options for surgical management of the primary tumor include the following:
Surgical staging of the axilla should also be performed.
Survival is equivalent with any of these options, as documented in the trial of the European Organization for Research and Treatment of Cancer (EORTC) (EORTC-10801) and other prospective randomized trials.
Also, a retrospective study of 753 patients who were divided into three groups based on hormone receptor status (ER positive or PR positive; ER negative and PR negative but HER2/neu positive; and triple negative) found no differences in disease control within the breast in patients treated with standard breast-conserving surgery; however, there are not yet substantive data to support this finding.
The rate of local recurrence in the breast after conservative treatment is low and varies slightly with the surgical technique used (e.g., lumpectomy, quadrantectomy, segmental mastectomy, and others). Whether completely clear microscopic margins are necessary has been debated.
However, a multidisciplinary consensus panel recently used margin width and ipsilateral breast tumor recurrence from a meta-analysis of 33 studies (N = 28,162 patients) as the primary evidence base for a new consensus regarding margins in stage I and stage II breast cancer patients treated with breast-conserving surgery plus radiation therapy. Results of the meta-analysis include the following:
For patients undergoing partial mastectomy, margins may be positive after primary surgery, often leading to re-excision. A clinical trial of 235 patients with stage 0 to III breast cancer who underwent partial mastectomy, with or without resection of selective margins, randomly assigned patients to have additional cavity shave margins resected (shave group) or not (no-shave group).
Patients in the shave group had a significantly lower rate of positive margins than those in the no-shave group (19% vs. 34%, P = .01) and a lower rate of second surgery for clearing margins (10% vs. 21%, P = .02).
[Level of evidence: 1iiDiv]
Axillary node status remains the most important predictor of outcome in breast cancer patients. Evidence is insufficient to recommend that lymph node staging can be omitted in most patients with invasive breast cancer. Several groups have attempted to define a population of women in whom the probability of nodal metastasis is low enough to preclude axillary node biopsy. In these single-institution case series, the prevalence of positive nodes in patients with T1a tumors ranged from 9% to 16%.
Another series reported the incidence of axillary node relapse in patients with T1a tumors treated without axillary lymph node dissection (ALND) was 2%.
[Level of evidence: 3iiiA]
The axillary lymph nodes are staged to aid in determining prognosis and therapy. SLN biopsy is the initial standard axillary staging procedure performed in women with invasive breast cancer. The SLN is defined as any node that receives drainage directly from the primary tumor; therefore, allowing for more than one SLN, which is often the case. Studies have shown that the injection of technetium Tc 99m-labeled sulfur colloid, vital blue dye, or both around the tumor or biopsy cavity, or in the subareolar area, and subsequent drainage of these compounds to the axilla results in the identification of the SLN in 92% to 98% of patients. These reports demonstrate a 97.5% to 100% concordance between SLN biopsy and complete ALND.
Because of the following body of evidence, SLN biopsy is the standard initial surgical staging procedure of the axilla for women with invasive breast cancer. SLN biopsy alone is associated with less morbidity than axillary lymphadenectomy.
Evidence (SLN biopsy):
Because of the following trial results, ALND is unnecessary after a positive SLN biopsy in patients with limited SLN-positive breast cancer treated with breast conservation or mastectomy, radiation, and systemic therapy.
Evidence (ALND after a positive SLN biopsy in patients with limited SLN-positive breast cancer):
For patients who require an ALND, the standard evaluation usually involves only a level I and II dissection, thereby removing a satisfactory number of nodes for evaluation (i.e., at least 6–10), while reducing morbidity from the procedure.
For patients who opt for a total mastectomy, reconstructive surgery may be performed at the time of the mastectomy (i.e., immediate reconstruction) or at some subsequent time (i.e., delayed reconstruction).
Breast contour can be restored by the following:
After breast reconstruction, radiation therapy can be delivered to the chest wall and regional nodes in either the adjuvant or local recurrent disease setting. Radiation therapy after reconstruction with a breast prosthesis may affect cosmesis, and the incidence of capsular fibrosis, pain, or the need for implant removal may be increased.
Radiation therapy is regularly employed after breast-conserving surgery. Radiation therapy is also indicated for high-risk postmastectomy patients. The main goal of adjuvant radiation therapy is to eradicate residual disease thus reducing local recurrence.
For women who are treated with breast-conserving surgery without radiation therapy, the risk of recurrence in the conserved breast is substantial (>20%) even in confirmed axillary lymph node–negative women.
Although all trials assessing the role of radiation therapy in breast-conserving therapy have shown highly statistically significant reductions in local recurrence rate, no single trial has demonstrated a statistically significant reduction in mortality. However, a large meta-analysis demonstrated a significant reduction in risk of recurrence and breast cancer death.
Thus, evidence supports the use of whole-breast radiation therapy after breast-conserving surgery.
Evidence (breast-conserving surgery followed by radiation therapy):
A 2011 meta-analysis of 17 clinical trials performed by the Early Breast Cancer Trialists’ Collaborative Group (EBCTCG), which included over 10,000 women with early-stage breast cancer, supported whole-breast radiation therapy after breast-conserving surgery.
[Level of evidence: 1iiA]
Regarding radiation dosing and schedule, the following has been noted:
Additional studies are needed to determine whether shorter fractionation is appropriate for women with higher nodal disease burden.
Regional nodal irradiation is routinely given postmastectomy to patients with involved lymph nodes; however, its role in patients who have breast-conserving surgery and whole-breast irradiation has been less clear. A randomized trial (NCT00005957) of 1,832 women showed that administering regional nodal irradiation after breast-conserving surgery and whole-breast irradiation reduces the risk of recurrence (10-year DFS, 82.0% vs. 77.0%; HR, 0.76; 95% CI, 0.61–0.94; P = .01) but does not affect survival (10-year OS, 82.8% vs. 81.8%; HR, 0.91; 95% CI, 0.72–1.13; P = .38).
[Level of evidence: 1iiA]
Similar findings were reported from the EORTC trial (NCT00002851). Women with a centrally or medially located primary tumor with or without axillary node involvement, or an externally located tumor with axillary involvement, were randomly assigned to receive whole-breast or thoracic-wall irradiation in addition to regional nodal irradiation or not. Breast-conserving surgery was performed for 76.1% of the study population, and the remaining study population underwent mastectomy. No improvement in OS was seen at 10 years among patients who underwent regional nodal irradiation when compared with patients who did not undergo regional nodal radiation (82.3% vs. 80.7%, P = .06). Distant DFS was improved among patients who underwent regional nodal irradiation when compared with patients who did not undergo regional nodal irradiation (78% vs. 75%, P = .02).
[Level of evidence: 1iiA]
A meta-analysis that combined the results of the two trials mentioned above found a marginally statistically significant difference in OS (HR, 0.88; 95% CI, 0.78–0.99; P = .034; absolute difference, 1.6% at 5 years).
Postoperative chest wall and regional lymph node adjuvant radiation therapy has traditionally been given to selected patients considered at high risk for locoregional failure after mastectomy. Patients at highest risk for local recurrence have one or more of the following:
In this high-risk group, radiation therapy can decrease locoregional recurrence, even among those patients who receive adjuvant chemotherapy.
Patients with one to three involved nodes without any of the high-risk factors are at low risk of local recurrence, and the value of routine use of adjuvant radiation therapy in this setting is unclear.
Evidence (postoperative radiation therapy in patients with one to three involved lymph nodes):
Further, an analysis of NSABP trials showed that even in patients with large (>5 cm) primary tumors and negative axillary lymph nodes, the risk of isolated locoregional recurrence was low enough (7.1%) that routine locoregional radiation therapy was not warranted.
The optimal sequence of adjuvant chemotherapy and radiation therapy after breast-conserving surgery has been studied. Based on the following studies, delaying radiation therapy for several months after breast-conserving surgery until the completion of adjuvant chemotherapy does not appear to have a negative impact on overall outcome. Additionally, initiating chemotherapy soon after breast-conserving surgery may be preferable for patients at high risk of distant dissemination.
Evidence (timing of postoperative radiation therapy):
The following results were observed:
These studies showed that delaying radiation therapy for 2 to 7 months after surgery had no effect on the rate of local recurrence. These findings have been confirmed in a meta-analysis.
[Level of evidence: 1iiA]
In an unplanned analysis of patients treated on a phase III trial evaluating the benefit of adding trastuzumab in HER2/neu–positive breast cancer patients, there was no associated increase in acute adverse events or frequency of cardiac events in patients who received concurrent adjuvant radiation therapy and trastuzumab.
Therefore, delivering radiation therapy concomitantly with trastuzumab appears to be safe and avoids additional delay in radiation therapy treatment initiation.
Late toxic effects of radiation therapy are uncommon and can be minimized with current radiation delivery techniques and with careful delineation of the target volume. Late effects of radiation include the following:
Modern radiation therapy techniques introduced in the 1990s minimized deep radiation to the underlying myocardium when left-sided chest wall or left-breast radiation was used. Cardiac mortality decreased accordingly.
An analysis of the National Cancer Institute’s Surveillance, Epidemiology, and End Results Program (SEER) data from 1973 to 1989 that reviewed deaths caused by ischemic heart disease in women who received breast or chest wall radiation showed that since 1980, no increased death rate resulting from ischemic heart disease in women who received left chest wall or breast radiation was found.
[Level of evidence: 3iB]
Stage and molecular features determine the need for adjuvant systemic therapy and the choice of modalities used. For example, hormone receptor (ER and/or PR)–positive patients will receive hormone therapy. HER2 overexpression is an indication for using adjuvant trastuzumab, usually in combination with chemotherapy. When neither HER2 overexpression nor hormone receptors are present (i.e., triple-negative breast cancer), adjuvant therapy relies on chemotherapeutic regimens, which may be combined with investigational targeted approaches.
An international consensus panel proposed a risk classification system and systemic therapy treatment options.
This classification, with some modification, is described below:
Subtype | Treatment Options | Comments |
---|---|---|
Luminal A–like | ||
– Hormone receptor–positive | Endocrine therapy alone in most cases | Consider chemotherapy if: |
– HER2-negative | – High tumor burden (≥4 LNs, T3 or higher) | |
– PR >20% | ||
– Ki67 low | – Grade 3 | |
Luminal B–like | ||
– Hormone receptor–positive | Endocrine therapy plus chemotherapy in most cases | |
– HER2-negative | ||
– Either Ki67 high or PR low | ||
HER2-positive | Chemotherapy plus anti-HER2 therapy | Use endocrine therapy if also hormone receptor–positive |
May consider omitting chemotherapy plus anti-HER2 for small node-negative tumors | ||
Triple-negative | Chemotherapy | May consider omitting chemotherapy for small node-negative tumors |
HER2 = human epidermal growth factor receptor 2; LN = lymph node; PR = progesterone receptor. | ||
aModified from Senkus et al. |
The selection of therapy is most appropriately based on knowledge of an individual’s risk of tumor recurrence balanced against the short-term and long-term risks of adjuvant treatment. This approach allows clinicians to help individuals determine if the gains anticipated from treatment are reasonable for their situation. The treatment options described below should be modified based on both patient and tumor characteristics.
Patient Group | Treatment Options |
---|---|
Premenopausal, hormone receptor–positive (ER or PR) | No additional therapy |
Tamoxifen | |
Tamoxifen plus chemotherapy | |
Ovarian function suppression plus tamoxifen | |
Ovarian function suppression plus aromatase inhibitor | |
Premenopausal, hormone receptor–negative (ER or PR) | No additional therapy |
Chemotherapy | |
Postmenopausal, hormone receptor–positive (ER or PR) | No additional therapy |
Upfront aromatase inhibitor therapy or tamoxifen followed by aromatase inhibitor with or without chemotherapy | |
Postmenopausal, hormone receptor–negative (ER or PR) | No additional therapy |
Chemotherapy | |
ER = estrogen receptor; PR = progesterone receptor. |
The EBCTCG meta-analysis analyzed 11 trials that began from 1976 to 1989 in which women were randomly assigned to receive regimens containing anthracyclines (e.g., doxorubicin or epirubicin) or CMF (cyclophosphamide, methotrexate, and 5-FU). The result of the overview analysis comparing CMF and anthracycline-containing regimens suggested a slight advantage for the anthracycline regimens in both premenopausal and postmenopausal women.
Evidence (anthracycline-based regimens):
Study results suggest that tumor characteristics (i.e., node-positive breast cancer with HER2/neu overexpression) may predict anthracycline-responsiveness.
Evidence (anthracycline-based regimen in women with HER2/neu amplification):
Several trials have addressed the benefit of adding a taxane (paclitaxel or docetaxel) to an anthracycline-based adjuvant chemotherapy regimen for women with node-positive breast cancer.
Evidence (adding a taxane to an anthracycline-based regimen):
An Eastern Cooperative Oncology Group–led intergroup trial (E1199 [NCT00004125]) involving 4,950 patients compared, in a factorial design, two schedules (weekly and every 3 weeks) of the two drugs (docetaxel vs. paclitaxel) after standard-dose AC chemotherapy given every 3 weeks.
[Level of evidence: 1iiA] Study findings include the following:
Historically, adjuvant chemotherapy for breast cancer was given on an every 3-week schedule. Studies sought to determine whether decreasing the duration between chemotherapy cycles could improve clinical outcomes. The overall results of these studies support the use of dose-dense chemotherapy for women with HER2-negative breast cancer.
Evidence (administration of dose-dense chemotherapy in women with HER2-negative breast cancer):
Docetaxel and cyclophosphamide is an acceptable adjuvant chemotherapy regimen.
Evidence (docetaxel and cyclophosphamide):
The optimal time to initiate adjuvant therapy is uncertain. A retrospective, observational study has reported the following:
Adjuvant chemotherapy is associated with several well-characterized toxic effects that vary according to the individual drugs used in each regimen. Common toxic effects include the following:
Less common, but serious, toxic effects include the following:
(Refer to the PDQ summary on Treatment-Related Nausea and Vomiting; for information on mucositis, refer to the PDQ summary on Oral Complications of Chemotherapy and Head/Neck Radiation; for information on symptoms associated with premature menopause, refer to the PDQ summary on Hot Flashes and Night Sweats.)
The use of anthracycline-containing regimens, however—particularly those containing an increased dose of cyclophosphamide—has been associated with a cumulative risk of developing acute leukemia of 0.2% to 1.7% at 5 years.
This risk increases to more than 4% in patients receiving high cumulative doses of both epirubicin (>720 mg/m2) and cyclophosphamide (>6,300 mg/m2).
Cognitive impairment has been reported to occur after the administration of some chemotherapy regimens.
However, data on this topic from prospective, randomized studies are lacking.
The EBCTCG meta-analysis revealed that women who received adjuvant combination chemotherapy did have a 20% (standard deviation = 10) reduction in the annual odds of developing contralateral breast cancer.
This small proportional reduction translated into an absolute benefit that was marginally statistically significant, but indicated that chemotherapy did not increase the risk of contralateral disease. In addition, the analysis showed no statistically significant increase in deaths attributed to other cancers or to vascular causes among all women randomly assigned to receive chemotherapy.
For HER2/neu–negative breast cancer, there is no single adjuvant chemotherapy regimen that is considered standard or superior to another. Preferred regimen options vary by institution, geographic region, and clinician.
Some of the most important data on the benefit of adjuvant chemotherapy came from the EBCTCG, which reviews data from global breast cancer trials every 5 years. In the 2011 EBCTCG meta-analysis, adjuvant chemotherapy using an anthracycline-based regimen compared with no treatment revealed significant improvement in the risk of recurrence (RR, 0.73; 95% CI, 0.68–0.79), significant reduction in breast cancer mortality (RR, 0.79; 95% CI, 0.72–0.85), and significant reduction in overall mortality (RR, 0.84; 95% CI, 0.78–0.91), which translated into an absolute survival gain of 5%.
TNBC is defined as the absence of staining for ER, PR, and HER2/neu. TNBC is insensitive to some of the most effective therapies available for breast cancer treatment including HER2-directed therapy such as trastuzumab and endocrine therapies such as tamoxifen or the aromatase inhibitors.
Combination cytotoxic chemotherapy administered in a dose-dense or metronomic schedule remains the standard therapy for early-stage TNBC.
Evidence (neoadjuvant chemotherapy on a dose-dense or metronomic schedule for TNBC):
Platinum agents have emerged as drugs of interest for the treatment of TNBC. However, there is no established role for adding them to the treatment of early-stage TNBC outside of a clinical trial. One trial that treated 28 women with stage II or stage III TNBC with four cycles of neoadjuvant cisplatin resulted in a 22% pCR rate.
[Level of evidence: 3iiiDiv] A randomized clinical trial, CALGB-40603 (NCT00861705), evaluated the benefit of carboplatin added to paclitaxel and doxorubicin plus cyclophosphamide chemotherapy in the neoadjuvant setting. The Triple Negative Trial (NCT00532727) is evaluating carboplatin versus docetaxel in the metastatic setting. These trials will help to define the role of platinum agents for the treatment of TNBC.
The PARP inhibitors are being evaluated in clinical trials for patients with BRCA mutations and in TNBC.
PARPs are a family of enzymes involved in multiple cellular processes, including DNA repair. Because TNBC shares multiple clinicopathologic features with BRCA-mutated breast cancers, which harbor dysfunctional DNA repair mechanisms, it is possible that PARP inhibition, in conjunction with the loss of DNA repair via BRCA-dependent mechanisms, would result in synthetic lethality and augmented cell death.
Standard treatment for HER2-positive early breast cancer is 1 year of adjuvant trastuzumab therapy.
Several phase III clinical trials have addressed the role of the anti-HER2/neu antibody, trastuzumab, as adjuvant therapy for patients with HER2-overexpressing cancers. Study results confirm the benefit of 12 months of adjuvant trastuzumab therapy.
Evidence (duration of trastuzumab therapy):
The Herceptin Adjuvant (HERA) (BIG-01-01 [NCT00045032]) trial examined whether the administration of trastuzumab was effective as adjuvant treatment for HER2-positive breast cancer if used after completion of the primary treatment. For most patients, primary treatment consisted of an anthracycline-containing chemotherapy regimen given preoperatively or postoperatively, plus or minus locoregional radiation therapy. Trastuzumab was given every 3 weeks starting within 7 weeks of the completion of primary treatment.
[Level of evidence: 1iiA] Patients were randomly assigned to one of three study arms:
Of the patients in the comparison of 1 year of trastuzumab versus observation group, the median age was 49 years, about 33% had node-negative disease, and nearly 50% had hormone receptor (ER and PR)–negative disease.
Several studies have evaluated the use of subcutaneous (SQ) trastuzumab in the neoadjuvant and adjuvant settings.
Cardiac events associated with adjuvant trastuzumab have been reported in multiple studies. Key study results include the following:
Lapatinib is a small-molecule tyrosine kinase inhibitor that is capable of dual-receptor inhibition of both epidermal growth factor receptor and HER2. There are no data supporting the use of lapatinib as part of adjuvant treatment of early-stage HER2/neu–positive breast cancer.
Evidence (against the use of lapatinib for HER2-positive early breast cancer):
Pertuzumab is a humanized monoclonal antibody that binds to a distinct epitope on the extracellular domain of the HER2 receptor and inhibits dimerization. Its use, in combination with trastuzumab, has been evaluated in a randomized trial in the postoperative setting.
Evidence (pertuzumab):
Neratinib is an irreversible tyrosine kinase inhibitor of HER1, HER2, and HER4, which has been approved by the FDA for the extended adjuvant treatment of patients with early-stage HER2-positive breast cancer, to follow adjuvant trastuzumab-based therapy.
Evidence (Neratinib):
Much of the evidence presented in the following sections on therapy for women with hormone receptor–positive disease has been considered in an American Society of Clinical Oncology guideline that describes several options for the management of these patients.
Five years of adjuvant endocrine therapy has been shown to substantially reduce the risks of locoregional and distant recurrence, contralateral breast cancer, and death from breast cancer.
The optimal duration of endocrine therapy is unclear, with the preponderance of evidence supporting at least 5 years of endocrine therapy. A meta-analysis of 88 clinical trials involving 62,923 women with hormone receptor–positive breast cancer who were disease free after 5 years of endocrine therapy showed a steady risk of late recurrence 5 to 20 years after diagnosis.
[Level of evidence: 3iiiD] The risk of distant recurrence correlated with the original tumor (T) and node (N) status, with risks ranging from 10% to 41%.
Tamoxifen has been shown to be of benefit to women with hormone receptor–positive breast cancer.
Evidence (tamoxifen for hormone receptor–positive early breast cancer):
The optimal duration of tamoxifen use has been addressed by the EBCTCG meta-analysis and by several large randomized trials.
Ten years of tamoxifen therapy has been shown to be superior to shorter durations of tamoxifen therapy.
Evidence (duration of tamoxifen therapy):
The results of the ATLAS trial indicated that for women who remained premenopausal after 5 years of adjuvant tamoxifen, continued tamoxifen for 5 more years was beneficial.
Women who have become menopausal after 5 years of tamoxifen may also be treated with AIs. (Refer to the Aromatase inhibitors section in the Hormone receptor–positive therapy section of this summary for more information.)
Because of the results of an EBCTCG analysis, the use of tamoxifen in women who received adjuvant chemotherapy does not attenuate the benefit of chemotherapy.
However, concurrent use of tamoxifen with chemotherapy is less effective than sequential administration.
Evidence suggests ovarian ablation alone is not an effective substitute for other systemic therapies.
Further, the addition of ovarian ablation to chemotherapy and/or tamoxifen has not been found to significantly improve outcomes.
Evidence (tamoxifen plus ovarian suppression):
Despite overall negative initial results, subgroup analysis suggested a benefit with ovarian suppression in women who underwent chemotherapy and remained premenopausal afterwards. Follow-up results at 8 years, however, did not demonstrate heterogeneity of treatment effect according to whether chemotherapy was administered, although recurrences were more frequent among patients who received chemotherapy.
AIs have been compared with tamoxifen in premenopausal women in whom ovarian function was suppressed or ablated. The results of these studies have been conflicting.
Evidence (comparison of an AI with tamoxifen in premenopausal women):
In postmenopausal women, the use of AIs in sequence with or as a substitute for tamoxifen has been the subject of multiple studies, the results of which have been summarized in an individual patient-level meta-analysis.
Evidence (AI vs. tamoxifen as initial therapy in postmenopausal women):
Several trials and meta-analyses have examined the effect of switching to anastrozole or exemestane to complete a total of 5 years of therapy after 2 to 3 years of tamoxifen.
The evidence, as described below, indicates that sequential tamoxifen and AI is superior to remaining on tamoxifen for 5 years.
Evidence (sequential tamoxifen and AI vs. 5 years of tamoxifen):
In the meta-analysis, which included 11,798 patients from six trials, the 10-year recurrence rate was reduced from 19% to 17% in the AI-containing groups (RR, 0.82; 95% CI, 0.75–0.91; P = .0001). The overall 10-year mortality was 17.5% in the tamoxifen group and 14.6% in the AI-containing group (RR, 0.82; 95% CI, 0.73–0.91; P = .0002).
[Level of evidence: 1A]
The evidence indicates that there is no benefit to the sequential use of tamoxifen and an AI for 5 years over 5 years of an AI.
Evidence (sequential use of tamoxifen and an AI vs. 5 years of an AI):
In the meta-analysis, which included 12,779 patients from the trials, the 7-year recurrence rate was slightly reduced from 14.5% to 13.8% in the groups that received 5 years of an AI (RR, 0.90; 95% CI, 0.81–0.99; P = .045). Overall mortality at 7 years was 9.3% in the tamoxifen-followed-by-AI groups and 8.2% in the AI-alone groups (RR, 0.89; 95% CI, 0.78–1.03; P = .11).
[Level of evidence: 1A]
The evidence, as described below, indicates that switching to an AI after 5 years of tamoxifen is superior to stopping tamoxifen at that time.
The optimal duration of AI therapy is uncertain, and multiple trials have evaluated courses longer than 5 years.
Evidence regarding extension of endocrine therapy beyond 5 years of initial AI-based adjuvant therapy:
Both bisphosphonates and denosumab have been evaluated as adjuvant therapies for early-stage breast cancer; however, the role of these agents as adjuvant therapy for early-stage breast cancer is unclear. Compared with denosumab, the amount of evidence supporting bisphosphonates is greater, and there is evidence supporting breast cancer mortality—an endpoint that is more clinically relevant.
Evidence (bisphosphonates in the treatment of early breast cancer):
An ongoing phase III trial (NCT01077154) is examining the activity of the bone-modifying agent, denosumab, in stage II and stage III breast cancer.
Preoperative chemotherapy, also known as primary or neoadjuvant chemotherapy, has traditionally been administered in patients with locally advanced breast cancer to reduce tumor volume and allow for definitive surgery. In addition, preoperative chemotherapy is being used for some patients with primary operable stage II or stage III breast cancer. A meta-analysis of multiple, randomized clinical trials performed in 2005 demonstrated that preoperative chemotherapy is associated with identical DFS and OS compared with the administration of the same therapy in the adjuvant setting.
[Level of evidence: 1iiA]
In 2019, the Early Breast Cancer Trialists’ Collaborative Group performed a meta-analysis using individual patient data from 4,756 women who participated in 10 trials that compared neoadjuvant chemotherapy with the same regimen given in the adjuvant setting.
Compared with adjuvant therapy, neoadjuvant therapy was associated with an increased frequency of breast conservation (65% vs. 49%). There were no differences between neoadjuvant chemotherapy and adjuvant therapy in distant recurrence, breast cancer mortality, or death from any cause; however, neoadjuvant therapy was associated with higher 15-year local recurrence (21.4% vs. 15.9%; rate ratio, 1.37; 95% CI, 1.17−1.61; P = .001).
[Level of evidence: 1iiA]
Current consensus opinion for use of preoperative chemotherapy recommends anthracycline- and taxane-based therapy, and prospective trials suggest that preoperative anthracycline- and taxane-based therapy is associated with higher response rates than alternative regimens (e.g., anthracycline alone).
[Level of evidence: 1iiDiv]
A potential advantage of preoperative systemic therapy is the increased likelihood of success with definitive local therapy in those presenting with locally-advanced, unresectable disease. It may also offer benefit to carefully selected patients with primary operable disease by enhancing the likelihood of breast conservation and providing prognostic information where pCR is obtained. In these cases, a patient can be informed that there is a very low risk of recurrence compared with a situation in which a large amount of residual disease remains.
pCR has been utilized as a surrogate endpoint for long-term outcomes, such as DFS, EFS, and OS, in preoperative clinical trials in breast cancer. A pooled analysis (CTNeoBC) of 11 preoperative randomized trials (n = 11,955) determined that pCR, defined as no residual invasive cancer in the breast and axillary nodes with presence or absence of in situ cancer (ypT0/is ypN0 or ypT0 ypN0), provided a better association with improved outcomes compared with eradication of invasive tumor from the breast alone (ypT0/is).
pCR could not be validated in this study as a surrogate endpoint for improved EFS and OS.
[Level of evidence: 3iiiD] Because of a strong association of pCR with substantially improved outcomes in individual patients with more aggressive subtypes of breast cancer, the FDA has supported use of pCR as an endpoint in preoperative clinical trials for patients with high-risk, early-stage breast cancer.
Postoperative radiation therapy may also be omitted in a patient with histologically negative axillary nodes after preoperative therapy, irrespective of lymph node status before preoperative therapy, allowing for tailoring of treatment to the individual.
Potential disadvantages with this approach include the inability to determine an accurate pathological stage after preoperative chemotherapy. However, the knowledge of the presence of residual disease may provide more personalized prognostic information, as noted above.
Multidisciplinary management of patients undergoing preoperative therapy by an experienced team is essential to optimize the following:
The tumor histology, grade, and receptor status are carefully evaluated before preoperative therapy is initiated. Patients whose tumors have a pure lobular histology, low grade, or high hormone-receptor expression and HER2-negative status are less likely to respond to chemotherapy and should be considered for primary surgery, especially when the nodes are clinically negative. Even if adjuvant chemotherapy is administered after surgery in these cases, a third-generation regimen (anthracycline-taxane based) may be avoided.
Before beginning preoperative therapy, the extent of the disease within the breast and regional lymph nodes should be assessed. Staging of systemic disease may include the following:
Baseline breast imaging is performed when breast-conserving therapy is desired to identify the tumor location and exclude multicentric disease. Suspicious abnormalities are usually biopsied before beginning treatment and a marker placed at the center of the breast tumor(s). When possible, suspicious axillary nodes may be biopsied before initiation of systemic treatment.
The optimal timing of sentinel lymph node (SLN) biopsy has not been established in patients receiving preoperative therapy. The following points should be considered:
When considering preoperative therapy, treatment options include the following:
Regular clinical assessment of response to therapy is necessary after beginning preoperative therapy. Repeat radiographic assessment is also required if breast conservation is the surgical goal. Patients with progressive disease during preoperative therapy may either transition to a non–cross-resistant regimen or proceed to surgery, if feasible.
Although switching to a non–cross-resistant regimen results in a higher pCR rate than continuing the same therapy, there is no clear evidence that other breast cancer outcomes are improved with this approach.
Early trials examined whether anthracycline-based regimens used in the adjuvant setting would prolong DFS and OS when used in the preoperative setting. The evidence supports higher rates of breast-conserving therapy with the use of a preoperative anthracycline chemotherapy regimen than with postoperative use, but no improvement in survival was noted with the preoperative strategy.
Evidence (preoperative anthracycline-based regimen):
To improve the results observed with AC alone, a taxane was added to the chemotherapy regimen. The following study results support the addition of a taxane to an anthracycline-based chemotherapy regimen for HER2-negative breast tumors.
Evidence (anthracycline-taxane–based chemotherapy regimen):
Promising results have been observed, however, with the addition of carboplatin to anthracycline-taxane combination chemotherapy regimens in patients with triple-negative breast cancer (TNBC). Future definitive studies evaluating survival endpoints and the identification of biomarkers of response or resistance are necessary before the addition of carboplatin to standard preoperative chemotherapy can be considered a new standard of care.
Evidence (adding carboplatin to an anthracycline-taxane–based chemotherapy regimen in patients with TNBC):
Importantly, results of studies in the adjuvant and metastatic settings have not demonstrated an OS benefit with the addition of bevacizumab to chemotherapy versus chemotherapy alone. However, the addition of bevacizumab to preoperative chemotherapy has been associated with an increased pCR rate alongside increased toxicity such as hypertension, cardiac toxicity, hand-foot syndrome, and mucositis (e.g., NSABP B-40 [NCT00408408] and GeparQuinto [NCT00567554]).
[Level of evidence: 1iiDiv] However, it is not clear that the modest benefit observed will translate into a longer term survival advantage.
After the success in the adjuvant setting, initial reports from phase II studies indicated improved pCR rates when trastuzumab, a monoclonal antibody that binds the extracellular domain of HER2, was added to preoperative anthracycline-taxane–based regimens.
[Level of evidence: 1iiDiv] This has been confirmed in phase III studies.
Evidence (trastuzumab):
A subcutaneous formulation of trastuzumab has also been approved.
The SafeHer (NCT01566721) trial evaluated the safety and tolerability of self-administered versus clinician-administered SQ trastuzumab in stage I to stage III HER2-positive breast cancer.
Chemotherapy was administered concurrently or sequentially.
A phase III (HannaH [NCT00950300]) trial also demonstrated that the pharmacokinetics and efficacy of preoperative SQ trastuzumab is noninferior to the IV formulation. This international, open-label trial (n = 596) randomly assigned women with operable, locally advanced, or inflammatory HER2-positive breast cancer to undergo preoperative chemotherapy (anthracycline-taxane–based), with either SQ-administered or IV-administered trastuzumab every 3 weeks before surgery. Patients received adjuvant trastuzumab to complete 1 year of therapy.
[Level of evidence: 1iiD] The pCR rates between the arms differed by 4.7% (95% CI, 4.0–13.4); 40.7% in the IV-administered group versus 45.4% in the SQ-administered group, demonstrating noninferiority for the SQ formulation. EFS and OS were secondary endpoints. Six-year EFS was 65% in both arms (HR, 0.98; 95% CI, 0.74−1.29). Six-year OS was 84% in both arms (HR, 0.94; 95% CI, 0.61−1.45).
Newer HER2-targeted therapies (lapatinib, pertuzumab) have also been investigated. It appears that dual targeting of the HER2 receptor results in an increase in pCR rate; however, no survival advantage has been demonstrated to date with this approach.
Pertuzumab is a humanized monoclonal antibody that binds to a distinct epitope on the extracellular domain of the HER2 receptor and inhibits dimerization. Pertuzumab, in combination with trastuzumab with or without chemotherapy, has been evaluated in two preoperative clinical trials to improve on the pCR rates observed with trastuzumab and chemotherapy.
Evidence (pertuzumab):
The following results were observed:
The following results were observed:
Because of these studies, the FDA-granted accelerated approval for the use of pertuzumab as part of a preoperative treatment for women with early-stage, HER2-positive breast cancer whose tumors are larger than 2 cm or node-positive.
The FDA approval of pertuzumab was subsequently converted to regular approval following the results of the confirmatory APHINITY (NCT01358877) trial, a randomized, phase III, adjuvant study for women with HER2-positive breast cancer, which demonstrated improved invasive DFS with the combination of chemotherapy and dual HER2-targeted therapy with pertuzumab plus trastuzumab compared with chemotherapy and trastuzumab alone.
Pertuzumab is now approved both in combination with trastuzumab and chemotherapy for the neoadjuvant therapy of locally advanced, inflammatory, or early-stage HER2-positive breast cancer, which is larger than 2 cm or node-positive, as part of a complete treatment regimen and in combination with chemotherapy and trastuzumab as adjuvant treatment for HER2-positive early breast cancer at a high risk of recurrence.
Lapatinib is a small-molecule kinase inhibitor that is capable of dual receptor inhibition of both epidermal growth factor receptor and HER2. Study results do not support the use of lapatinib in the preoperative setting.
Evidence (against the use of lapatinib for HER2-positive early breast cancer):
More definitive efficacy data were provided by the phase III ALLTO (NCT00490139) trial that randomly assigned women to receive trastuzumab or trastuzumab plus lapatinib in the adjuvant setting.
The trial did not meet its primary endpoint of DFS. The doubling in pCR rate observed with the addition of lapatinib to trastuzumab in the NeoALTTO trial did not translate into improved survival outcomes in the ALTTO trial at 4.5 years of median follow-up. This indicates that there is currently no role for the use of lapatinib in the preoperative or adjuvant settings.
A pooled analysis of cardiac safety in 598 cancer patients treated with pertuzumab was performed using data supplied by Roche and Genentech.
[Level of evidence: 3iiiD]
A meta-analysis of randomized trials (n = 6) that evaluated the administration of anti-HER2 monotherapy (trastuzumab or lapatinib or pertuzumab) versus dual anti-HER2 therapy (trastuzumab plus lapatinib or trastuzumab plus pertuzumab) was performed.
[Level of evidence: 3iiiD]
Preoperative endocrine therapy may be an option for postmenopausal women with hormone receptor-positive breast cancer when chemotherapy is not a suitable option because of comorbidities or performance status. Although the toxicity profile of preoperative hormonal therapy over the course of 3 to 6 months is favorable, the pCR rates obtained (1%–8%) are far lower than have been reported with chemotherapy in unselected populations.
[Level of Evidence: 1iDiv]
Longer duration of preoperative therapy may be required in this patient population. Preoperative tamoxifen was associated with an overall response rate of 33%, with maximum response occurring up to 12 months after therapy in some patients.
A randomized study of 4, 8, or 12 months of preoperative letrozole in elderly patients who were not fit for chemotherapy indicated that the longer duration of therapy resulted in the highest pCR rate (17.5% vs. 5% vs. 2.5%, P-value for trend < .04).
[Level of Evidence: 1iiDiv]
AIs have also been compared with tamoxifen in the preoperative setting. Overall objective response and breast-conserving therapy rates with 3 to 4 months of preoperative therapy were either statistically significantly improved in the AI-treated women
or comparable to tamoxifen-associated outcomes.
An American College of Surgeons Oncology Group trial is currently comparing the efficacy of anastrozole, letrozole, or exemestane in the preoperative setting.
The use of preoperative endocrine therapy in premenopausal women with hormone-responsive breast cancer remains investigational.
One clinical trial suggested that there is a benefit to using capecitabine as adjuvant therapy in patients who did not obtain a pCR after preoperative chemotherapy.
Evidence (capecitabine):
Evidence (T-DM1):
These approaches and participation in clinical trials of novel therapies should be considered for patients with residual disease after preoperative therapy. EA1131 (NCT02445391) is a randomized phase III clinical trial that randomly assigned patients with residual basal-like TNBC after preoperative therapy to receive platinum-based chemotherapy or capecitabine. S1418/BR006 (NCT02954874) is a phase III trial evaluating the efficacy of pembrolizumab as adjuvant therapy for patients with residual TNBC (≥1 cm invasive cancer or residual nodes) after preoperative therapy.
Radiation therapy is administered after breast conservation in most women who have received preoperative therapy to reduce the risk of locoregional recurrence. Baseline clinical and subsequent pathologic staging should be considered in deciding whether to administer postmastectomy radiation.
Other adjuvant systemic treatments may be administered either postoperatively, during, or after completion of adjuvant radiation, including adjuvant hormonal therapy for patients with hormone receptor-positive disease and adjuvant trastuzumab for those with HER2-positive disease. (Refer to the Hormone receptor–positive breast cancer subsection in the Early/Localized/Operable Breast Cancer section of this summary for more information.)
The frequency of follow-up and the appropriateness of screening tests after the completion of primary treatment for stage I, stage II, or stage III breast cancer remain controversial.
Evidence from randomized trials indicates that periodic follow-up with bone scans, liver sonography, chest x-rays, and blood tests of liver function does not improve survival or quality of life when compared with routine physical examinations.
Even when these tests permit earlier detection of recurrent disease, patient survival is unaffected.
On the basis of these data, acceptable follow-up can be limited to the following for asymptomatic patients who complete treatment for stages I to III breast cancer:
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
根据现有证据,具有治疗意图的多模式治疗是局部晚期或炎性乳腺癌的治疗标准。
局部晚期或炎性乳腺癌的标准治疗方案可能包括以下内容:
初始手术通常限于活检,以确定组织学类型、雌激素受体(ER)和孕激素受体状态以及人表皮生长因子受体2 (HER2/neu)的过表达情况。
初始治疗的标准化疗方案与辅助治疗中使用的方案相同(更多信息参见本总结的术后全身治疗部分),尽管仅在局部晚期疾病患者中进行的试验并未显示出剂量密集化疗的统计学显著优势。
对于对术前化疗有反应的患者,局部治疗可能包括全乳房切除术伴腋窝淋巴结清扫,随后对胸壁和区域淋巴管进行术后放疗。对术前化疗有良好的部分反应或完全反应的患者,可以考虑保乳治疗。
随后的全身治疗可能包括进一步的化疗。激素治疗用于ER阳性或ER未知肿瘤的患者。
虽然下面描述的证据还没有被重复出来,但是它表明局部晚期或炎症性乳腺癌患者应该以治疗为目的来接受治疗。
证据(多模态治疗):
随后的试验已经证实,局部晚期和炎性乳腺癌患者在接受初始化疗时,可能会出现长期DFS。
所有患者都被认为是临床试验的候选对象,以评估新综合疗法中各个组成部分的最合适的管理方案。
使用我们的高级临床试验搜索来查找NCI支持的癌症临床试验,这些试验现在正在招募患者。可以通过试验地点、治疗类型、药物名称和其他标准来缩小搜索范围。关于临床试验的一般信息也是可用的。
On the basis of the available evidence, multimodality therapy delivered with curative intent is the standard of care for patients with locally advanced or inflammatory breast cancer.
The standard treatment options for locally advanced or inflammatory breast cancer may include the following:
Initial surgery is generally limited to biopsy to permit the determination of histology, estrogen receptor (ER) and progesterone receptor levels, and human epidermal growth factor receptor 2 (HER2/neu) overexpression.
The standard chemotherapy regimen for initial treatment is the same as that used in the adjuvant setting (refer to the Postoperative Systemic Therapy section of this summary for more information), although trials done solely in patients with locally advanced disease have not shown a statistically significant advantage to dose-dense chemotherapy.
For patients who respond to preoperative chemotherapy, local therapy may consist of total mastectomy with axillary lymph node dissection followed by postoperative radiation therapy to the chest wall and regional lymphatics. Breast-conserving therapy can be considered for patients with a good partial or complete response to preoperative chemotherapy.
Subsequent systemic therapy may consist of further chemotherapy. Hormone therapy is administered to patients with ER-positive or ER-unknown tumors.
Although the evidence described below has not been replicated, it suggests patients with locally advanced or inflammatory breast cancer should be treated with curative intent.
Evidence (multimodality therapy):
Subsequent trials have confirmed that patients with locally advanced and inflammatory breast cancer can experience long-term DFS when treated with initial chemotherapy.
All patients are considered candidates for clinical trials to evaluate the most appropriate way to administer the various components of new multimodality regimens.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
复发性乳腺癌通常对治疗有反应,尽管疾病在这个阶段很少能够治愈。局部乳腺癌复发的患者可以通过适当的治疗而长期存活。
随着时间的推移,局部复发率已经降低,一项荟萃分析表明,接受保乳手术和放射治疗的患者复发率低于3%。
对于接受乳房切除术治疗的患者,这一比率略高(高达10%)。
9%至25%的局部复发患者在复发时会有远处转移或局部广泛病变。
在治疗复发性乳腺癌之前,需要重新评估疾病的程度。应尽可能获得复发性病变的细胞学或组织学资料。当选择治疗时,考虑复发时和先前治疗时的雌激素受体(ER)状态、孕激素受体(PR)状态和人表皮生长因子受体2 (HER2/neu)状态(如果已知)。
ER状态可能会在复发时发生变化。在癌症和白血病B组(MDA-MBDT-8081)的一项小型研究中,在复发时获取的活检标本中发现,36%原为激素受体阳性的肿瘤变为了受体阴性。
本研究中的患者没有间歇治疗。如果雌激素受体和孕激素受体状态未知,那么在化疗或激素治疗选择上,复发部位、无病间期、对先前治疗的反应和绝经状态是有用的。
局部复发性乳腺癌的治疗选择包括以下内容:
局部复发的患者应考虑接受进一步的局部治疗(如乳房切除术)。在一个研究系列中,保乳术联合化疗后出现浸润性复发患者五年精确复发率为52%。
治疗方案也取决于复发部位,如下所示:
(有关复发性转移性乳腺癌的治疗信息,请参考本摘要的转移性(全身性)疾病部分。)所有复发性乳腺癌患者都被认为是正在进行的临床试验的候选人。
使用我们的高级临床试验搜索来查找NCI支持的癌症临床试验,这些试验现在正在招募患者。可以通过试验地点、治疗类型、药物名称和其他标准来缩小搜索范围。关于临床试验的一般信息也是可用的。
Recurrent breast cancer is often responsive to therapy, although treatment is rarely curative at this stage of disease. Patients with locoregional breast cancer recurrence may become long-term survivors with appropriate therapy.
The rates of locoregional recurrence have been reduced over time, and a meta-analysis suggests a recurrence rate of less than 3% in patients treated with breast-conserving surgery and radiation therapy.
The rates are somewhat higher (up to 10%) for those treated with mastectomy.
Nine percent to 25% of patients with locoregional recurrence will have distant metastases or locally extensive disease at the time of recurrence.
Before treatment for recurrent breast cancer, restaging to evaluate the extent of disease is indicated. Cytologic or histologic documentation of recurrent disease is obtained whenever possible. When therapy is selected, the estrogen-receptor (ER) status, progesterone-receptor (PR) status, and human epidermal growth factor receptor 2 (HER2/neu) status at the time of recurrence and previous treatment are considered, if known.
ER status may change at the time of recurrence. In a single small study by the Cancer and Leukemia Group B (MDA-MBDT-8081), 36% of hormone receptor–positive tumors were found to be receptor negative in biopsy specimens isolated at the time of recurrence.
Patients in this study had no interval treatment. If ER and PR statuses are unknown, then the site(s) of recurrence, disease-free interval, response to previous treatment, and menopausal status are useful in the selection of chemotherapy or hormone therapy.
Treatment options for locoregional recurrent breast cancer include the following:
Patients with locoregional recurrence should be considered for further local treatment (e.g., mastectomy). In one series, the 5-year actuarial rate of relapse for patients treated for invasive recurrence after initial breast conservation and radiation therapy was 52%.
Treatment options also depend on the site of recurrence, as follows:
(Refer to the Metastatic (systemic) disease section of this summary for information about treatment for recurrent metastatic breast cancer.) All patients with recurrent breast cancer are considered candidates for ongoing clinical trials.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
转移性疾病的治疗是姑息性的。治疗的目标包括延长寿命和提高生活质量。尽管据报道总的中位生存期为18至24个月,
存活率因亚型而异。在人表皮生长因子受体2 (HER2)阳性和激素受体阳性转移性乳腺癌患者中观察到的中位结果最长,在三阴性转移性乳腺癌患者中观察到的中位结果较差。
转移性乳腺癌的治疗选择包括:
在许多情况下,这些治疗是按顺序给予的,并以多种组合使用。
应尽可能获得转移性疾病的细胞学或组织学资料,包括雌激素受体(ER)、孕激素受体(PR)和HER2状态的检测。
所有转移性乳腺癌患者都被认为是正在进行的临床试验的候选人。
CDK4和CDK6与激素受体阳性乳腺癌的持续增殖有关,这种乳腺癌对内分泌治疗具有耐药性。美国食品和药物管理局(FDA)已批准CDK抑制剂在晚期激素受体阳性HER2阴性乳腺癌的一线和二线治疗中与内分泌治疗联合使用。目前有三种口服CDK4/6抑制剂:palbociclib、ribociclib和abemaciclib。
总的来说,在内分泌治疗中加入CDK 4/6抑制剂与乳腺癌预后改善相关,一般来说,可以维持或改善生活质量。
证据(一线Palbociclib和内分泌治疗):
另一种CDK4/6抑制剂Ribociclib也已在绝经后和绝经前激素受体阳性HER2阴性复发性或转移性乳腺癌患者的一线治疗中进行了试验。
证据(一线Ribociclib和内分泌治疗):
另一种CDK4/6抑制剂Abemaciclib也已在绝经后激素受体阳性HER2阴性复发或转移性乳腺癌患者的一线治疗中进行了试验。
证据(一线Abemaciclib和内分泌治疗):
证据(二线Palbociclib与内分泌治疗)
MONALEESA-3试验包括接受二线治疗的患者。(有关更多信息,请参考上面关于一线Ribociclib和内分泌治疗的证据。)
证据:
证据(单药细胞周期蛋白依赖性激酶抑制剂治疗):
临床前模型和临床研究表明,mTOR抑制剂可能会克服内分泌耐药。
证据(mTOR抑制剂治疗):
在大约40%的激素受体阳性和HER2阴性乳腺癌中发现PIK3CA激活突变。Alpelisib是一种α-特异性PI3K抑制剂。
证据(alpelisib加内分泌治疗):
在341名受试者中证实了PIK3CA突变。主要终点是PIK3CA突变患者队列中的PFS。
经FDA批准,在接受内分泌治疗后,alpelisib可与氟维司群联合用于晚期PIK3CA突变、激素受体阳性、HER2阴性乳腺癌。
如上所述,由于靶向治疗和内分泌治疗联合应用可带来PFS和OS的优势,所以单纯内分泌治疗不太常用,尤其是在一线治疗中。然而,在选择的病例中,在靶向治疗进展后的一线治疗和后期治疗中,以及在仍认为存在内分泌敏感疾病的病例中使用化疗前,它的使用仍然是合适的。
常用的单剂内分泌治疗包括他莫昔芬、非甾体类AI(来曲唑、阿那曲唑)、甾体类抗AI(依西美坦)和氟维司群。总的来说,患有转移性乳腺癌的绝经前女性会经历卵巢抑制或切除,之后与绝经后女性采用相同的方式治疗。
虽然他莫昔芬多年来一直用于治疗绝经后女性新发转移的ER阳性、PR阳性或ER/PR未知的乳腺癌,但一些随机试验表明,与他莫昔芬相比,AI的具有相当的或更优的反应率和PFS。
[证据等级:1iiDiii]
证据(他莫昔芬和AI治疗):
氟维司群是一种选择性的雌激素受体拮抗剂,已经在晚期或转移性女性乳腺癌患者的一线和二线治疗中得到研究。
证据(一线氟维司群):
证据(二线富维斯塔特):
在两项试验中发现了相互矛盾的结果,这两项试验比较了在激素受体阳性的绝经后复发或转移性乳腺癌患者的一线治疗中联合使用抗雌激素氟维司群(有关该药物的更多信息,请参阅氟维司群一节)和单独使用阿那曲唑与阿那曲唑。
在这两项研究中,在第一天以500毫克负荷剂量给予氟维司群;第15和29天服用250毫克,此后每月服用一次;此外,每天服用1毫克阿那曲唑。西南肿瘤集团(SWOG)试验包括了更多患有转移性乳腺癌的患者;氟维司群和阿那曲唑联合治疗(FACT [NCT00256698])研究招募了更多以前接受过他莫昔芬治疗的患者。
证据(AI和氟维司群联合内分泌治疗):
激素受体阳性转移性乳腺癌的最佳治疗顺序尚不清楚。一般来说,在没有内脏危象的情况下,大多数患者在过渡到化疗前接受基于内分泌的序贯治疗方案。在上述PFS和OS改善的基础上,一线治疗中的CDK4/6抑制剂治疗和内分泌治疗相结合是一个合适的选择。
激素受体阴性乳腺癌的治疗方法是化疗。(更多信息请参阅本总结的化疗部分。)
针对HER2途径的抗体治疗自20世纪90年代开始应用,并彻底改变了HER2阳性转移性乳腺癌的治疗方法。几种HER2靶向药物(如曲妥珠单抗、帕妥珠单抗、曲妥珠单抗-emtansine偶联物、拉帕替尼)已被批准用于治疗本病。
大约20%到25%的乳腺癌过表达HER2/neu。
曲妥珠单抗是一种人源化单克隆抗体,可与HER2/neu受体结合。
在先前接受过细胞毒性化疗且肿瘤过表达HER2/neu的患者中,单用曲妥珠单抗治疗的缓解率为21%。
[证据级别:3iiiDiv]
证据(曲妥珠单抗):
值得注意的是,当与多柔比星联合使用时,曲妥珠单抗具有显著的心脏毒性。
比较多药化疗加曲妥珠单抗与单药化疗的临床试验产生了相互矛盾的结果。
临床试验之外,转移性HER2过表达乳腺癌的标准一线治疗是单药化疗加曲妥珠单抗。
帕妥珠单抗是一种人源化单克隆抗体,与曲妥珠单抗相比,它可以结合到HER2胞外结构域的不同表位。帕妥珠单抗与HER2的结合阻止了与其他配体激活的HER受体的二聚作用,最显著的是HER3。
证据(帕妥珠单抗):
曲妥珠单抗-美坦新偶联物(T-DM1)是一种结合曲妥珠单抗HER2靶向抗肿瘤特性和微管抑制剂DM1细胞毒活性的抗体药物结合物。T-DM1使特异性的细胞内药物传递到HER2过表达的细胞,有可能提高治疗指数,减少正常组织的暴露。
证据(T-DM1):
拉帕替尼是一种口服的HER2/neu和表皮生长因子受体酪氨酸激酶抑制剂。拉帕替尼联合卡培他滨治疗曲妥珠单抗治疗后进展的HER2阳性转移性乳腺癌患者显示出疗效。
证据级别(拉帕替尼):
对于携带生殖系BRCA突变的转移性乳腺癌患者,聚腺苷二磷酸核糖聚合酶(PARP)的口服抑制剂已显示出疗效。BRCA1和BRCA2是通过同源重组修复途径编码DNA修复相关蛋白的抑癌基因。PARP在DNA修复中起着关键性的作用,并已被用于治疗乳腺癌BRCA突变的患者的治疗。
证据(奥拉帕尼):
证据(他拉唑帕尼):
(更多信息请参阅PDQ乳腺癌和妇科癌遗传学总结。)
接受激素治疗且肿瘤进展的患者是细胞毒性化疗的候选者。没有数据表明联合治疗比单药治疗更有益于OS。激素受体阴性肿瘤和内脏转移或症状性疾病的患者也可作为细胞毒性药物化疗的候选者。
在转移性乳腺癌中表现出活性的单一药物包括:
在转移性乳腺癌中显示活性的联合治疗方案包括:
没有数据表明联合治疗比单药治疗更有益于OS。一项东部肿瘤协作组间研究(E-1193)随机分配患者接受紫杉醇和多柔比星的治疗,既给予联合治疗,也给予序贯治疗。
尽管联合治疗组的缓解率和疾病进展时间都更好,但两组的存活率是相同的。
[证据级别:1iiA];
患者的个体治疗选择受以下因素影响:
目前,没有数据支持任何特定治疗方案的优越性。对于转移性疾病复发的患者,可序贯使用单一药物或联合用药。如果有快速进展性疾病或内脏危象的迹象,通常会进行联合化疗。化疗和激素治疗的联合应用并没有显示出这些药物序贯使用的优势。
对17项随机试验的系统回顾发现,在化疗方案中加入一种或多种化疗药物以加强治疗,可改善肿瘤应答,但对OS无影响。
[证据级别:1iiA]
关于化疗持续时间的决定可考虑以下方面:
对患者疾病缓解或稳定的最佳治疗时间进行了多组研究。对于获得对初始治疗完全缓解的患者,两项随机试验显示,与复发时的观察和治疗相比,使用不同的化疗方案立即治疗后,DFS延长。
[证据级别:1iiA]然而,这两项研究都没有显示接受立即治疗的患者的OS有改善; 在其中一项研究中,
在立即接受治疗的组中,存活率实际上更差。同样,当初始治疗后部分缓解或疾病稳定的患者被随机分配接受不同的化疗或观察时,存活率没有差异;
或者用不同的化疗方案,用高剂量和低剂量比较时,存活率没有差异。
[证据级别:1iiA]然而,324例疾病得到控制的患者被随机分配到维持性化疗或观察组。接受维持性化疗(紫杉醇和吉西他滨)的患者在6个月时PFS改善,OS改善。这与不良事件的增加率有关。
[证据级别:1iiA]由于对转移性疾病的治疗没有标准方法,所以需要二线疗法的患者是临床试验的良好候选人。
在III期、随机、安慰剂对照IMpassion130试验(NCT 02425891)中,对激素受体阴性和HER2阴性的晚期乳腺癌患者在一线化疗中添加一种抗程序性死亡配体1 (PD-L1)阳性抗体Atezolizumab进行了评估。
受试者(N=902)按1:1随机接受给 Atezolizumab加白蛋白结合型紫杉醇或安慰剂加白蛋白结合型紫杉醇。受试者根据是/否肝转移、是/否接受过紫杉烷治疗和PD-L1状态(阳性或阴性)进行分层。PD-L1评分≥1%被定义为阳性。共同的主要终点包括PFS和OS,这两个指标在治疗意向人群和PD-L1阳性人群中进行评估(n=369)。
Atezolizumab被FDA批准与蛋白结合型紫杉醇联合应用于局部晚期或转移性三阴性乳腺癌患者,其肿瘤表达PD-L1。
在为选定的患者选择化疗方案时,应考虑蒽环类药物引起心脏毒性效应的可能性。公认的心脏毒性风险因素包括:
在对照研究中,心脏保护药物右雷佐生已被证明可降低多柔比星诱导的心脏毒性的风险。这种药物的使用允许患者接受更高的多柔比星累积剂量,并允许有心脏危险因素的患者能够接受多柔比星。
持续静脉滴注多柔比星也可以降低心脏毒性的风险。
美国临床肿瘤学会指南建议,如果转移性癌症患者接受了累积剂量为300毫克/ m2或更高的多柔比星,当进一步使用蒽环类药物治疗可能获益时,使用右雷佐生。
右雷佐生对接受表柔比星的患者有类似的保护作用。
手术可能适用于特定的患者。例如,如果出现以下问题,患者可能需要手术:
(有关更多信息,请参阅癌症疼痛的PDQ总结;有关胸腔积液和心包积液的信息,请参阅心肺综合征的PDQ总结。)
放射治疗在缓解局部症状性转移中起着重要作用。
外照射治疗的适应症包括:
氯化锶Sr 89是一种全身给药的放射性核素,可用于缓解弥漫性骨转移。
应考虑使用骨修复治疗来降低骨骼疾病患者的骨骼相关事件发病率。
帕米膦酸钠和氯膦酸钠在骨转移患者中的随机试验结果显示骨骼相关事件发病率下降。
[证据级别:1iC]唑来膦酸盐至少和帕米膦酸盐一样有效。
在CALGB-70604[Alliance;NCT00869206]中研究了唑来膦酸盐的最佳给药方案,该方案随机分配1822例患者,其中855例患有转移性乳腺癌,每4周或每12周接受一次唑来膦酸盐。
两组的骨骼相关事件相似,唑来膦酸盐每4周给药组260例患者(29.5%)和唑来膦酸盐每12周给药组253例患者(28.6%)出现至少一个骨骼相关事件(风险差异为-0.3%[单侧95%置信区间,-4%无穷大];非劣性P<0.001)。
[证据级别:1iiD]这项研究表明,每12周延长唑来膦酸盐的给药间隔是一个合理的治疗选择。
单克隆抗体地诺单抗抑制核因子kappaβ配体受体激活剂(RANKL)。比较唑来膦酸盐和地舒单抗治疗骨转移的三期临床试验(NCT 00321464、NCT 0321620和NCT 0330759)的荟萃分析表明,地诺单抗与唑来膦酸盐在降低首次骨相关事件风险方面相似。
(有关双膦酸盐的更多信息,请参阅关于癌症疼痛的PDQ总结。)
贝伐单抗是一种人源化的单克隆抗体,可直接作用于血管内皮生长因子-a的所有亚型。它在转移性乳腺癌治疗中的作用仍有争议。
证据(贝伐单抗治疗转移性乳腺癌):
2011年11月,由于一致发现贝伐单抗只轻微改善了PFS,但没有改善OS,并且考虑到贝伐单抗具有相当大的毒性,FDA撤销了贝伐单抗治疗转移性乳腺癌的批准。
使用高级临床试验搜索引擎,以查找NCI支持的正在招募患者的癌症临床试验。根据试验地点、治疗类型、药物名称和其他标准可以缩小搜索范围。有关临床试验的一般信息也可以获得。
Treatment of metastatic disease is palliative in intent. Goals of treatment include prolonging life and improving quality of life. Although median survival has been reported to be 18 to 24 months overall,
survival varies according to subtype. The longest median outcomes have been observed in patients with human epidermal growth factor receptor 2 (HER2)-positive and hormone receptor–positive metastatic breast cancer, and less favorable outcomes have been observed in patients with triple-negative metastatic breast cancer.
Treatment options for metastatic breast cancer include the following:
In many cases, these therapies are given in sequence and used in various combinations.
Cytologic or histologic documentation of metastatic disease, with testing of estrogen receptor (ER), progesterone receptor, and HER2 status, should be obtained whenever possible.
All patients with metastatic breast cancer are considered candidates for ongoing clinical trials.
CDK4 and CDK6 have been implicated in the continued proliferation of hormone receptor–positive breast cancer resistant to endocrine therapy. CDK inhibitors have been approved by the U.S. Food and Drug Administration (FDA) in combination with endocrine therapy in both first-line and later-line treatment of advanced hormone receptor–positive HER2-negative breast cancer. Three oral CDK4/6 inhibitors are currently available: palbociclib, ribociclib, and abemaciclib.
Overall, the addition of CDK 4/6 inhibitors to endocrine therapy is associated with improved breast cancer outcomes and, in general, either maintained or improved quality of life.
Evidence (first-line palbociclib and endocrine therapy):
Ribociclib, another CDK4/6 inhibitor, has also been tested in the first-line setting for postmenopausal patients and premenopausal patients with hormone receptor–positive and HER2-negative recurrent or metastatic breast cancer.
Evidence (first-line ribociclib and endocrine therapy):
Abemaciclib, another CDK4/6 inhibitor, has also been tested in the first-line setting for postmenopausal patients with hormone receptor–positive and HER2-negative recurrent or metastatic breast cancer.
Evidence (first-line abemaciclib and endocrine therapy):
Evidence (second-line palbociclib and endocrine therapy):
The MONALEESA-3 trial included patients receiving second-line therapy. (Refer to the evidence on first-line ribociclib and endocrine therapy above for more information.)
Evidence:
Evidence (single-agent cyclin-dependent kinase inhibitor therapy):
Preclinical models and clinical studies suggest that mTOR inhibitors might overcome endocrine resistance.
Evidence (mTOR inhibitor therapy):
Activating mutations in PIK3CA are identified in approximately 40% of hormone receptor–positive and HER2-negative breast cancers. Alpelisib is an alpha-specific PI3K inhibitor.
Evidence (alpelisib plus endocrine therapy):
PIK3CA mutations were confirmed in 341 participants. The primary endpoint was PFS in the cohort of patients with PIK3CA mutations.
Alpelisib is approved by the FDA for use in combination with fulvestrant in advanced PIK3CA-mutated, hormone receptor–positive, HER2-negative breast cancer after previous endocrine therapy.
With the PFS and OS advantages associated with combination therapy with targeted agents and endocrine therapy as discussed above, single-agent endocrine therapy is less frequently used, especially in the first-line setting. However, its use remains appropriate in select cases as first-line therapy and in later-line therapy after progression on targeted therapies and before the use of chemotherapy in cases in which endocrine-sensitive disease is still thought to be present.
Commonly used single-agent endocrine therapies include tamoxifen, nonsteroidal AI (letrozole, anastrozole), the steroidal AI exemestane, and fulvestrant. In general, premenopausal women with metastatic breast cancer undergo ovarian suppression or ablation and are treated in the same manner as postmenopausal women.
While tamoxifen has been used for many years in treating postmenopausal women with newly metastatic disease that is ER positive, PR positive, or ER/PR unknown, several randomized trials suggest equivalent or superior response rates and PFS for the AI compared with tamoxifen.
[Level of evidence: 1iiDiii]
Evidence (tamoxifen and AI therapy):
Fulvestrant is a selective estrogen receptor degrader that has been studied in the first-line and second-line setting in women with advanced or metastatic breast cancer.
Evidence (first-line fulvestrant):
Evidence (second-line fulvestrant):
Conflicting results were found in two trials that compared the combination of the antiestrogen fulvestrant (refer to the section on Fulvestrant for more information about this drug) and anastrozole with anastrozole alone in the first-line treatment of hormone receptor–positive postmenopausal patients with recurrent or metastatic disease.
In both studies, fulvestrant was administered as a 500-mg loading dose on day 1; 250 mg was administered on days 15 and 29, and monthly thereafter; plus, 1 mg of anastrozole was administered daily. The Southwest Oncology Group (SWOG) trial included more patients who presented with metastatic disease; the Fulvestrant and Anastrozole Combination Therapy (FACT [NCT00256698]) study enrolled more patients who had previously received tamoxifen.
Evidence (combination endocrine therapy with an AI and fulvestrant):
The optimal sequence of therapies for hormone receptor–positive metastatic breast cancer is not known. In general, in the absence of a visceral crisis, most patients receive sequential endocrine-based regimens before transitioning to chemotherapy. On the basis of the PFS and OS improvements mentioned above, a combination of a CDK4/6 inhibitor therapy and endocrine therapy in the first line is an appropriate choice.
The treatment for hormone receptor–negative breast cancer is chemotherapy. (Refer to the Chemotherapy section of this summary for more information.)
Antibody therapy targeting the HER2 pathway has been used since the 1990s and has revolutionized the treatment of HER2-positive metastatic breast cancer. Several HER2-targeted agents (e.g., trastuzumab, pertuzumab, ado-trastuzumab emtansine, lapatinib) have been approved for treatment of this disease.
Approximately 20% to 25% of patients with breast cancer have tumors that overexpress HER2/neu.
Trastuzumab is a humanized monoclonal antibody that binds to the HER2/neu receptor.
In patients previously treated with cytotoxic chemotherapy whose tumors overexpress HER2/neu, administration of trastuzumab as a single agent resulted in a response rate of 21%.
[Level of evidence: 3iiiDiv]
Evidence (trastuzumab):
Notably, when combined with doxorubicin, trastuzumab is associated with significant cardiac toxicity.
Clinical trials comparing multiagent chemotherapy plus trastuzumab with single-agent chemotherapy have yielded conflicting results.
Outside of a clinical trial, standard first-line treatment for metastatic HER2-overexpressing breast cancer is single-agent chemotherapy plus trastuzumab.
Pertuzumab is a humanized monoclonal antibody that binds to a different epitope at the HER2 extracellular domain than does trastuzumab. The binding of pertuzumab to HER2 prevents dimerization with other ligand-activated HER receptors, most notably HER3.
Evidence (pertuzumab):
Ado-trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that incorporates the HER2-targeted antitumor properties of trastuzumab with the cytotoxic activity of the microtubule-inhibitory agent DM1. T-DM1 allows specific intracellular drug delivery to HER2-overexpressing cells, potentially improving the therapeutic index and minimizing exposure of normal tissue.
Evidence (T-DM1):
Lapatinib is an orally administered tyrosine kinase inhibitor of both HER2/neu and the epidermal growth factor receptor. Lapatinib plus capecitabine has shown activity in patients who have HER2-positive metastatic breast cancer that progressed after treatment with trastuzumab.
Evidence (lapatinib):
For patients with metastatic breast cancer who carry a germline BRCA mutation, the oral inhibitor of poly (adenosine diphosphate-ribose) polymerase (PARP) has shown activity. BRCA1 and BRCA2 are tumor-suppressor genes that encode proteins involved in DNA repair through the homologous recombination repair pathway. PARP plays a critical role in DNA repair and has been studied as therapy for patients with breast cancer who harbor a germline BRCA mutation.
Evidence (olaparib):
Evidence (talazoparib):
(Refer to the PDQ summary on Genetics of Breast and Gynecologic Cancers for more information.)
Patients receiving hormone therapy whose tumors have progressed are candidates for cytotoxic chemotherapy. There are no data suggesting that combination therapy results in an OS benefit over single-agent therapy. Patients with hormone receptor–negative tumors and those with visceral metastases or symptomatic disease are also candidates for cytotoxic agents.
Single agents that have shown activity in metastatic breast cancer include the following:
Combination regimens that have shown activity in metastatic breast cancer include the following:
There are no data suggesting that combination therapy results in an OS benefit over single-agent therapy. An Eastern Cooperative Oncology intergroup study (E-1193) randomly assigned patients to receive paclitaxel and doxorubicin, given both as a combination and sequentially.
Although response rate and time to disease progression were both better for the combination, survival was the same in both groups.
[Level of evidence: 1iiA];
The selection of therapy in individual patients is influenced by the following:
Currently, no data support the superiority of any particular regimen. Sequential use of single agents or combinations can be used for patients who relapse with metastatic disease. Combination chemotherapy is often given if there is evidence of rapidly progressive disease or visceral crisis. Combinations of chemotherapy and hormone therapy have not shown an OS advantage over the sequential use of these agents.
A systematic review of 17 randomized trials found that the addition of one or more chemotherapy drugs to a chemotherapy regimen in the attempt to intensify the treatment improved tumor response but had no effect on OS.
[Level of evidence: 1iiA]
Decisions regarding the duration of chemotherapy may consider the following:
The optimal time for patients with responsive or stable disease has been studied by several groups. For patients who attain a complete response to initial therapy, two randomized trials have shown a prolonged DFS after immediate treatment with a different chemotherapy regimen compared with observation and treatment upon relapse.
[Level of evidence: 1iiA] Neither of these studies, however, showed an improvement in OS for patients who received immediate treatment; in one of these studies,
survival was actually worse in the group that was treated immediately. Similarly, no difference in survival was noted when patients with partial response or stable disease after initial therapy were randomly assigned to receive either a different chemotherapy versus observation
or a different chemotherapy regimen given at higher versus lower doses.
[Level of evidence: 1iiA] However, 324 patients who achieved disease control were randomly assigned to maintenance chemotherapy or observation. Patients who received maintenance chemotherapy (paclitaxel and gemcitabine) had improved PFS at 6 months and improved OS. This was associated with an increased rate of adverse events.
[Level of evidence: 1iiA] Because there is no standard approach for treating metastatic disease, patients requiring second-line regimens are good candidates for clinical trials.
The addition of atezolizumab, an anti-programmed death ligand 1 (PD-L1)–positive antibody, to first-line chemotherapy for patients with hormone receptor–negative and HER2-negative advanced breast cancer was evaluated in the phase III randomized placebo-controlled IMpassion130 trial (NCT02425891).
Participants (N = 902) were randomly assigned 1:1 to atezolizumab plus nanoparticle albumin-bound (nab)-paclitaxel or to placebo plus nab-paclitaxel. Participants were stratified according to the presence of liver metastases (yes/no), receipt of previous taxane therapy (yes/no), and PD-L1 status (positive or negative). PD-L1 score of 1% or greater was defined as positive. Co-primary endpoints included PFS and OS, both of which were evaluated in the intention-to-treat population and in the PD-L1–positive population (n = 369).
Atezolizumab was granted accelerated approval by the FDA for use in combination with protein-bound paclitaxel for patients with unresectable locally advanced or metastatic triple-negative breast cancer whose tumors express PD-L1.
The potential for anthracycline-induced cardiac toxic effects should be considered in the selection of chemotherapeutic regimens for selected patients. Recognized risk factors for cardiac toxicity include the following:
The cardioprotective drug dexrazoxane has been shown to decrease the risk of doxorubicin-induced cardiac toxicity in patients in controlled studies. The use of this agent has permitted patients to receive higher cumulative doses of doxorubicin and has allowed patients with cardiac risk factors to receive doxorubicin.
The risk of cardiac toxicity may also be reduced by administering doxorubicin as a continuous intravenous infusion.
The American Society of Clinical Oncology guidelines suggest the use of dexrazoxane in patients with metastatic cancer who have received a cumulative dose of doxorubicin of 300 mg/m2 or more when further treatment with an anthracycline is likely to be of benefit.
Dexrazoxane has a similar protective effect in patients receiving epirubicin.
Surgery may be indicated for select patients. For example, patients may need surgery if the following issues occur:
(Refer to the PDQ summary on Cancer Pain for more information; refer to the PDQ summary on Cardiopulmonary Syndromes for information about pleural and pericardial effusions.)
Radiation therapy has a major role in the palliation of localized symptomatic metastases.
Indications for external-beam radiation therapy include the following:
Strontium chloride Sr 89, a systemically administered radionuclide, can be administered for palliation of diffuse bony metastases.
The use of bone-modifying therapy to reduce skeletal morbidity in patients with bone metastases should be considered.
Results of randomized trials of pamidronate and clodronate in patients with bony metastatic disease show decreased skeletal morbidity.
[Level of evidence: 1iC] Zoledronate has been at least as effective as pamidronate.
The optimal dosing schedule for zoledronate was studied in CALGB-70604 [Alliance; NCT00869206], which randomly assigned 1,822 patients, 855 of whom had metastatic breast cancer, to receive zoledronic acid every 4 weeks or every 12 weeks.
Skeletal-related events were similar in both groups, with 260 patients (29.5%) in the zoledronate every-4-week dosing group and 253 patients (28.6%) in the zoledronate every-12-week dosing group experiencing at least one skeletal-related event (risk difference of -0.3% [1-sided 95% CI, -4% to infinity]; P < .001 for noninferiority).
[Level of evidence: 1iiD] This study suggests that the longer dosing interval of zoledronate every 12 weeks is a reasonable treatment option.
The monoclonal antibody denosumab inhibits the receptor activator of nuclear factor kappa beta ligand (RANKL). A meta-analysis of three phase III trials (NCT00321464, NCT00321620, and NCT00330759) comparing zoledronate versus denosumab for management of bone metastases suggests that denosumab is similar to zoledronate in reducing the risk of a first skeletal-related event.
(Refer to the PDQ summary on Cancer Pain for more information on bisphosphonates.)
Bevacizumab is a humanized monoclonal antibody directed against all isoforms of vascular endothelial growth factor–A. Its role in the treatment of metastatic breast cancer remains controversial.
Evidence (bevacizumab for metastatic breast cancer):
In November 2011, because of the consistent finding that bevacizumab improved PFS only modestly but did not improve OS, and given bevacizumab’s considerable toxicity profile, the FDA revoked approval of bevacizumab for the treatment of metastatic breast cancer.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
导管原位癌(DCIS)是一种非侵袭性疾病。DCIS可以发展成浸润性癌,但对这种可能性的估计差异很大。一些报告将DCIS纳入乳腺癌统计。预计到2020年,DCIS将占美国所有新诊断的浸润性和非浸润性乳腺肿瘤的18%。
对于通过筛查发现的浸润性和非浸润性肿瘤,DCIS约占所有病例的25%。
自从乳腺钼靶X线检查技术在美国广泛应用以来,诊断DCIS的频率明显增加。很少有DCIS表现为可触及的肿块,超过90%的DCIS仅通过钼靶诊断。
DCIS包括一组异质性组织病理学病变,根据结构模式主要分为以下亚型:
粉刺型乳腺导管原位癌由细胞学上呈恶性的细胞组成,伴有高级别核、多型性和大量中央管腔坏死。粉刺型乳腺导管原位癌的侵袭性更强,伴有浸润性导管癌的可能性更高。
DCIS的治疗方案包括:
在过去,DCIS的常规治疗是乳房切除术。
30%的DCIS为多灶性病变、仅行广泛切除后仍有40%的肿瘤残存、局限性切除可触及的肿瘤后25-50%会复发且其中50%为浸润癌。而行乳房单纯切除后局部及远处复发率仅1%-2%。目前尚无关于乳房单纯切除术与保乳手术联合放疗疗效对比的随机研究数据。
由于保乳手术联合乳腺放射治疗是治疗浸润性癌的有效方法,因此将保乳手术推广到DCIS。为了确定保乳手术加放射治疗是否为治疗DCIS的合理方法,国家外科辅助性乳腺和肠道项目(NSABP)和欧洲癌症研究和治疗组织(EORTC)各自完成了前瞻性随机试验,其中切除活检后局部DCIS和手术切缘阴性的女性被随机分配接受乳腺放射治疗(50 Gy)或者没有进一步的治疗。
证据(保乳手术加乳腺放射治疗):
在这两项研究中,放射治疗的效果在所有评估的风险因素中是一致的。
NSABP-B-17和EORTC-10853试验以及另外两个试验的结果被纳入荟萃分析,显示所有同侧乳腺事件(HR,0.49;95%CI,0.41-0.58;P<0.00001)、同侧浸润性复发(HR,0.50;95%CI,0.32-0.76;P=0.001)和同侧DCIS复发(HR,0.61;95%CI,0.39-0.95;P=0.03)均减少。
[证据级别:1iiD]随访10年后,对乳腺癌死亡率、非乳腺癌死亡率或全因死亡率均无显著影响。
为了确定一组术后可省略放疗的患者,已经开发了几种病理分期系统并对其进行了回顾性检测,但尚未达成一致意见。
Van-Nuys预后指数是一个结合了三个局部复发预测因子(即肿瘤大小、切缘宽度和病理分类)的病理分期系统。采用回顾性分析方法,对333例单纯切除或放射治疗的病例进行分析。
利用预后指数,仅行手术切除的良性病变患者复发率较低(即2%,平均随访79个月)。随后对这些数据进行分析,以确定切缘宽度对局部控制的影响。
切除病灶的患者在各个方向的边缘宽度都在10毫米或以上,仅通过手术,局部复发的可能性极低(4%,平均随访8年)。
这两篇综述都是回顾性的、非对照的,并且有很大的选择偏差。相反,前瞻性NSABP试验未发现任何亚组患者在DCIS的治疗中未从乳腺保乳手术加放射治疗获益。
为了确定他莫昔芬是否能增加局部治疗对DCIS的疗效,NSABP进行了双盲前瞻性试验(NSABP-B-24)。
证据(辅助内分泌治疗):
在诊断出DCIS后,决定采用内分泌疗法通常需要与患者讨论每种药物的潜在获益和副作用。
使用高级临床试验搜索引擎,以查找NCI支持的正在招募患者的癌症临床试验。根据试验地点、治疗类型、药物名称和其他标准可以缩小搜索范围。有关临床试验的一般信息也可以获得。
Ductal carcinoma in situ (DCIS) is a noninvasive condition. DCIS can progress to invasive cancer, but estimates of the probability of this vary widely. Some reports include DCIS in breast cancer statistics. In 2020, DCIS is expected to account for about 18% of all newly diagnosed invasive plus noninvasive breast tumors in the United States.
For invasive and noninvasive tumors detected by screening, DCIS accounts for approximately 25% of all cases.
The frequency of a DCIS diagnosis has increased markedly in the United States since the use of screening mammography became widespread. Very few cases of DCIS present as a palpable mass, with more than 90% being diagnosed by mammography alone.
DCIS comprises a heterogeneous group of histopathologic lesions that have been classified into the following subtypes primarily because of architectural pattern:
Comedo-type DCIS consists of cells that appear cytologically malignant, with the presence of high-grade nuclei, pleomorphism, and abundant central luminal necrosis. Comedo-type DCIS appears to be more aggressive, with a higher probability of associated invasive ductal carcinoma.
Treatment options for DCIS include the following:
In the past, the customary treatment for DCIS was mastectomy.
The rationale for mastectomy included a 30% incidence of multicentric disease, a 40% prevalence of residual tumor at mastectomy after wide excision alone, and a 25% to 50% incidence of in-breast recurrence after limited surgery for palpable tumor, with 50% of those recurrences being invasive carcinoma. The combined local and distant recurrence rate after mastectomy is 1% to 2%. No randomized comparisons of mastectomy versus breast-conserving surgery plus breast radiation therapy are available.
Because breast-conserving surgery combined with breast radiation therapy is successful for invasive carcinoma, this conservative approach was extended to DCIS. To determine whether breast-conserving surgery plus radiation therapy was a reasonable approach to the management of DCIS, the National Surgical Adjuvant Breast and Bowel Project (NSABP) and the European Organisation for Research and Treatment of Cancer (EORTC) have each completed prospective randomized trials in which women with localized DCIS and negative surgical margins after excisional biopsy were randomly assigned to receive either breast radiation therapy (50 Gy) or no further therapy.
Evidence (breast-conserving surgery plus radiation therapy to the breast):
In both studies, the effect of radiation therapy was consistent across all assessed risk factors.
The results of the NSABP-B-17 and EORTC-10853 trials plus two others were included in a meta-analysis that demonstrated reductions in all ipsilateral breast events (HR, 0.49; 95% CI, 0.41–0.58; P < .00001), ipsilateral invasive recurrence (HR, 0.50; 95% CI, 0.32–0.76; P = .001), and ipsilateral DCIS recurrence (HR, 0.61; 95% CI, 0.39–0.95; P = .03).
[Level of evidence: 1iiD] After 10 years of follow-up, there was, however, no significant effect on breast cancer mortality, mortality from causes other than breast cancer, or all-cause mortality.
To identify a favorable group of patients for whom postoperative radiation therapy could be omitted, several pathologic staging systems have been developed and tested retrospectively, but consensus recommendations have not been achieved.
The Van Nuys Prognostic Index is one pathologic staging system that combines three predictors of local recurrence (i.e., tumor size, margin width, and pathologic classification). It was used to retrospectively analyze 333 patients treated with either excision alone or excision and radiation therapy.
Using this prognostic index, patients with favorable lesions who received surgical excision alone had a low recurrence rate (i.e., 2%, with a median follow-up of 79 months). A subsequent analysis of these data was performed to determine the influence of margin width on local control.
Patients whose excised lesions had margin widths of 10 mm or more in every direction had an extremely low probability of local recurrence with surgery alone (4%, with a mean follow-up of 8 years).
Both reviews are retrospective, noncontrolled, and subject to substantial selection bias. In contrast, the prospective NSABP trial did not identify any subset of patients who did not benefit from the addition of radiation therapy to breast-conserving surgery in the management of DCIS.
To determine whether tamoxifen adds to the efficacy of local therapy in the management of DCIS, the NSABP performed a double-blind prospective trial (NSABP-B-24).
Evidence (adjuvant endocrine therapy):
The decision to prescribe endocrine therapy after a diagnosis of DCIS often involves a discussion with the patient about the potential benefits and side effects of each agent.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
定期审查PDQ癌症信息总结,并在获得新信息时进行更新。本节介绍截至上述日期对此总结所做的最新更改。
更新了2020年估计新发病例和死亡病例的统计数据(引用美国癌症协会作为参考1)。
这一节作了广泛的修订。
本总结由PDQ成人治疗编辑委员会编写和维护,编辑委员会独立于NCI。本总结反映了对文献的独立审查,并不代表NCI或NIH的政策声明。关于总结政策和PDQ编辑委员会在维护PDQ总结中的作用的更多信息,可参见本PDQ总结和PDQ® -NCI综合癌症数据库页面。
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.
Updated statistics with estimated new cases and deaths for 2020 (cited American Cancer Society as reference 1).
This section was extensively revised.
This summary is written and maintained by the PDQ Adult Treatment 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.