这份执行摘要回顾了这份关于乳腺癌和妇科癌症遗传学的PDQ总结中涵盖的主题,并超链接到下面描述每个主题证据的详细章节。
提示乳腺癌和妇科癌症发生的遗传因素包括:1)在有乳腺癌家族史的个体中,这些癌症的发病率增加;2)多个家族成员受累于这些或其他癌症;3)与常染色体显性遗传一致的癌症模式。男性和女性都可以遗传和传递常染色体显性的癌症易感性基因。
与家族史相关的其他因素——如生育史、口服避孕药和激素替代使用、早年的辐射暴露史、饮酒和体力活动——能够影响个人患癌症的风险。
已经开发了风险评估模型以明确个人的:1)罹患乳腺癌和/或妇科癌症的终生风险;2) BRCA1或BRCA2发生致病性突变的可能性;3)与Lynch综合症相关的错配修复基因中存在致病性突变的可能性。
乳腺癌和卵巢癌存在于几种常染色体显性遗传的癌症综合症中,尽管它们与生殖系高致病突变最密切相关。其他基因,例如,(与Li-Fraumeni综合症相关),(与Cowden综合症相关),(与弥漫性胃癌和小叶乳腺癌综合征相关),以及(与Peutz-Jeghers综合症相关),都赋予了这些癌症中任何一种或两者较高的外显率的风险。
遗传性子宫内膜癌最常与Lynch综合症相关,该综合症由高外显率的错配修复基因MLH1、MSH2、MSH6、PMS2和EPCAM中的遗传致病性突变引起。结直肠癌(卵巢癌和胃癌,程度较轻)也与Lynch综合症有关。
其他基因,例如,,,和,与存在中等外显率的乳腺癌和/或妇科癌症相关。全基因组搜索在鉴定许多复杂疾病的常见、低外显率易感等位基因方面显示出了希望,其中包括乳腺癌和妇科癌症,但这些发现的临床应用仍不确定。
乳腺癌筛查策略,包括乳腺核磁共振成像和乳腺X线检查,通常是在携带BRCA致病性突变的携带者和乳腺癌风险增加的个体中进行的。一般建议,存在遗传和家族史而导致患病风险增加的个体开始筛查的年龄比一般人群的年龄更早,且筛查的间隔更频繁。有证据表明,乳腺癌筛查策略在早期发现癌症方面具有实用价值。相比之下,目前还没有证据表明,使用癌症抗原125检测和经阴道超声进行妇科癌症筛查能够早期发现癌症。
降低风险的手术,包括降低风险的乳房切除术(RRM)和降低风险的输卵管卵巢切除术(RRSO),已被证明可以显著降低BRCA1和BRCA2致病性突变携带者患乳腺癌和/或卵巢癌的风险,并改善总生存期。化学预防策略,包括使用他莫昔芬和口服避孕药,也在这一人群中进行了研究。他莫昔芬的使用已经被证明可以减少乳腺癌治疗后BRCA1和BRCA2致病性突变携带者患对侧乳腺癌的风险,但原发性癌症预防中有限的数据不足以表明它能减少携带BRCA2致病性突变的健康女性患乳腺癌的风险。使用口服避孕药对卵巢癌风险具有保护作用,包括在BRCA1和BRCA2致病性突变携带者中,当使用1975年以后开发的制剂时,与乳腺癌风险的增加无关。
社会心理因素影响遗传性癌症风险的基因检测的决策和风险管理策略。不同的研究对基因检测的理解差异很大。与检测理解相关的社会心理因素包括癌症特异性痛苦和发生乳腺癌或卵巢癌的感知风险。研究表明,对携带者和非携带者进行基因检测后,他们的痛苦程度都较低,尤其是从长期来看。对RRM和RRSO的理解也因研究而异,且可能受癌症史、年龄、家族史、医疗服务人员的建议、治疗前遗传教育和咨询等因素的影响。患者与家属之间关于乳腺癌和妇科癌症遗传风险的沟通是复杂的;性别、年龄和亲缘程度是影响该信息披露的一些因素。目前正在进行研究,以便更好地了解和处理高危家族中的社会心理和行为问题。
This executive summary reviews the topics covered in this PDQ summary on the genetics of breast and gynecologic cancers, with hyperlinks to detailed sections below that describe the evidence on each topic.
Factors suggestive of a genetic contribution to both breast cancer and gynecologic cancer include 1) an increased incidence of these cancers among individuals with a family history of these cancers; 2) multiple family members affected with these and other cancers; and 3) a pattern of cancers compatible with autosomal dominant inheritance. Both males and females can inherit and transmit an autosomal dominant cancer predisposition gene.
Additional factors coupled with family history—such as reproductive history, oral contraceptive and hormone replacement use, radiation exposure early in life, alcohol consumption, and physical activity—can influence an individual’s risk of developing cancer.
Risk assessment models have been developed to clarify an individual's 1) lifetime risk of developing breast and/or gynecologic cancer; 2) likelihood of having a pathogenic variant in BRCA1 or BRCA2; and 3) likelihood of having a pathogenic variant in one of the mismatch repair genes associated with Lynch syndrome.
Breast and ovarian cancer are present in several autosomal dominant cancer syndromes, although they are most strongly associated with highly penetrant germline pathogenic variants in and . Other genes, such as , (associated with Li-Fraumeni syndrome), (associated with Cowden syndrome), (associated with diffuse gastric and lobular breast cancer syndrome), and (associated with Peutz-Jeghers syndrome), confer a risk to either or both of these cancers with relatively high penetrance.
Inherited endometrial cancer is most commonly associated with Lynch syndrome, a condition caused by inherited pathogenic variants in the highly penetrant mismatch repair genes MLH1, MSH2, MSH6, PMS2, and EPCAM. Colorectal cancer (and, to a lesser extent, ovarian cancer and stomach cancer) is also associated with Lynch syndrome.
Additional genes, such as , , , and , are associated with breast and/or gynecologic cancers with moderate penetrance. Genome-wide searches are showing promise in identifying common, low-penetrance susceptibility alleles for many complex diseases, including breast and gynecologic cancers, but the clinical utility of these findings remains uncertain.
Breast cancer screening strategies, including breast magnetic resonance imaging and mammography, are commonly performed in carriers of BRCA pathogenic variants and in individuals at increased risk of breast cancer. Initiation of screening is generally recommended at earlier ages and at more frequent intervals in individuals with an increased risk due to genetics and family history than in the general population. There is evidence to demonstrate that these strategies have utility in early detection of cancer. In contrast, there is currently no evidence to demonstrate that gynecologic cancer screening using cancer antigen 125 testing and transvaginal ultrasound leads to early detection of cancer.
Risk-reducing surgeries, including risk-reducing mastectomy (RRM) and risk-reducing salpingo-oophorectomy (RRSO), have been shown to significantly reduce the risk of developing breast and/or ovarian cancer and improve overall survival in carriers of BRCA1 and BRCA2 pathogenic variants. Chemoprevention strategies, including the use of tamoxifen and oral contraceptives, have also been examined in this population. Tamoxifen use has been shown to reduce the risk of contralateral breast cancer among carriers of BRCA1 and BRCA2 pathogenic variants after treatment for breast cancer, but there are limited data in the primary cancer prevention setting to suggest that it reduces the risk of breast cancer among healthy female carriers of BRCA2 pathogenic variants. The use of oral contraceptives has been associated with a protective effect on the risk of developing ovarian cancer, including in carriers of BRCA1 and BRCA2 pathogenic variants, with no association of increased risk of breast cancer when using formulations developed after 1975.
Psychosocial factors influence decisions about genetic testing for inherited cancer risk and risk-management strategies. Uptake of genetic testing varies widely across studies. Psychological factors that have been associated with testing uptake include cancer-specific distress and perceived risk of developing breast or ovarian cancer. Studies have shown low levels of distress after genetic testing for both carriers and noncarriers, particularly in the longer term. Uptake of RRM and RRSO also varies across studies, and may be influenced by factors such as cancer history, age, family history, recommendations of the health care provider, and pretreatment genetic education and counseling. Patients' communication with their family members about an inherited risk of breast and gynecologic cancer is complex; gender, age, and the degree of relatedness are some elements that affect disclosure of this information. Research is ongoing to better understand and address psychosocial and behavioral issues in high-risk families.
在美国女性中,乳腺癌是最常诊断的癌症,仅次于非黑色素瘤皮肤癌,是仅次于肺癌的第二大癌症死亡原因。到2020年,预计将有279,100例新发病例(包括2620例男性病例)被确诊,和42,690例死亡病例(包括520例男性死亡)将发生。
乳腺癌的发病率,特别是50岁以后发生的雌激素受体(ER)阳性乳腺癌的发病率正在降低,而且自2003年以来降低的速度更快;这可能与妇女健康倡议(WHI)早期报告后激素替代治疗(HRT)的减少有关。据估计,2015年中国女性乳腺癌新发病例30.4万,死亡人数7.0万。
据估计,2020年美国将有21,750例卵巢癌新发病例,预计将有13,940例死亡。卵巢癌是女性的第五大致死性癌症。卵巢癌在中国城市人群中发病率较高,据估计,2015年城市发病率为为7.22/10万。2015年卵巢癌总死亡人数达2.5万。
据估计,2020年美国子宫内膜癌的新发病例将达到65,620例,预计将有12,590例死亡。据估计,2015年中国女性子宫体癌新发病例6.9万,生存率较高,从2012年到2015年子宫体癌患者年龄标化的5年生存率达72.8%。
(更多有关乳腺癌、卵巢癌和子宫内膜癌发病率、诊断和管理的信息,请参阅乳腺癌治疗[成人]、卵巢上皮癌、输卵管癌、原发性腹膜癌的治疗和子宫内膜癌治疗的PDQ总结。)
这些癌症在有家族史的女性中发病率增加(更多信息,请参阅以下乳腺癌的风险因素、卵巢癌的风险因素和子宫内膜癌的风险因素部分内容),以及对有多个家族成员患乳腺和或卵巢癌的一些家族进行观察,表明了遗传是乳腺癌和卵巢癌的可能风险因素,这符合常染色体显性遗传的癌症易感性。有关家族的正式研究(连锁分析)随后证明了乳腺癌和卵巢癌的常染色体显性遗传倾向,并导致了一些高外显基因被鉴定为许多家族遗传癌症风险的原因。(有关链接分析的更多信息,请参阅PDQ总结中的癌症遗传学概述)。这些基因的致病性突变在普通人群中很少见,估计占乳腺癌和卵巢癌病例总数的不超过5%到10%。看起来,其他的遗传因素可能是这些癌症的病因。
有关一般人群中乳腺癌风险因素的信息,请参阅关于乳腺癌预防的PDQ总结。
乳腺癌的累积风险随着年龄的增长而增加,大多数乳腺癌发生在50岁以后。
乳腺癌(和不同程度的卵巢癌)在具有遗传易感性的女性中往往比散发病例的女性更早发生。
在对成年人群的横断面研究中,5%至10%的女性有患乳腺癌的母亲或姐妹,有患乳腺癌的一级亲属(FDR)或二级亲属的女性大约是这个比例的两倍。
乳腺癌家族史的风险已经在病例对照研究和队列研究中得到评估,使用的是志愿者和基于人群的样本,结果基本一致。
在对38项研究的汇总分析中,由患有乳腺癌的一个FDR引起的乳腺癌相对危险度(RR)为2.1(95%置信区间[CI],2.0-2.3)。
患病亲属的人数、诊断时的年龄、家族成员中双侧或多侧乳腺癌的发生以及男性患病亲属的数量都会增加患病风险。
瑞典家族癌症数据库的一项大规模人群研究证实,母亲或姐妹患有乳腺癌的女性患乳腺癌的风险显著增加。家族中有单一乳腺癌的女性的风险比(HR)为1.8(95% CI,1.8-1.9),而有多个乳腺癌患者家族史的女性的风险比(HR)为2.7(95% CI,2.6-2.9)。家族中有多个乳腺癌患者的女性,其中一例发生在40岁之前,其HR为3.8(95% CI,3.1-4.8)。然而,该研究还发现,如果亲属的年龄在60岁或以上,患乳腺癌的风险也会显著增加,这表明,在家族的任何年龄,患乳腺癌的风险都会有所增加。
另一项针对患有单侧乳腺癌和对侧乳腺癌(CBC)的女性的研究,评估了家庭成员患乳腺癌的风险。
结果显示,在有FDR患乳腺癌的女性中,10年的CBC风险为8.1%。该风险在40岁之前或患有CBC的亲属中更高,在BRCA携带者中接近风险估计的下限。(请参阅本总结中的高外显率乳腺癌和/或妇科癌症易感基因章节中BRCA致病性突变携带者的对侧乳腺癌部分,以了解该人群的癌症风险评估信息。) 当仅对BRCA1/BRCA2、ATM、CHEK2和PALB2的有害突变检测呈阴性的女性进行分析时,这些风险估计仍然保持不变。
一项开展的迄今为止规模最大的关于双胞胎的研究,涉及了80,309对同卵双胞胎和123,382对异卵双胞胎,报告了乳腺癌遗传可能性的估计值为31%(95%置信区间,11%-51%)。
如果一个同卵双胞胎患有乳腺癌,她的双胞胎姐妹患乳腺癌的可能性为28.1%(95% CI,23.9%-32.8%),如果一个异卵双胞胎患有乳腺癌,她的双胞胎姐妹患乳腺癌的概率为19.9% (95% CI,17%-23.2%)。这些估计表明,同卵双胞胎患乳腺癌的风险比异卵双胞胎高10%。然而,即使是同卵双胞胎之间也有很高的不一致率,这表明环境因素在改变乳腺癌风险方面也有作用。
(请参阅本总结中BRCA致病性突变外显率的部分内容,以讨论来自BRCA1/BRCA2致病性突变家族的女性的家族风险,而这些女性本身的家族致病性突变检测结果为阴性。)
一般来说,乳腺癌风险随着月经提前和绝经延迟而增加,并在首次足月妊娠早期降低。在更小的年龄(30岁之前)妊娠时, BRCA1和BRCA2致病性突变携带者患乳腺癌的风险可能会增加,而携带BRCA1致病性突变携带者患乳腺癌的风险更大。
同样,母乳喂养可以降低BRCA1(但不是BRCA2)致病性突变携带者患乳腺癌风险。
关于妊娠对乳腺癌预后的影响,无论是妊娠期诊断的乳腺癌,还是妊娠后诊断的乳腺癌,都与携带BRCA1或BRCA2致病性突变的女性的不良生存结局无关。
产次对BRCA1和BRCA2致病性突变携带者具有保护作用,对40岁前的活产具有额外的保护作用。
生育史也会影响卵巢癌和子宫内膜癌的风险。(更多信息,请参阅本总结中的卵巢癌风险因素和子宫内膜癌风险因素中的生育史部分内容。)
口服避孕药(OC)可能轻微增加长期服用者患乳腺癌的风险,但这是一个短期的作用。在一项对54项研究数据进行的荟萃分析中,与使用OC相关的乳腺癌患病风险与乳腺癌家族史之间的关系没有变化。
OC有时被推荐用于BRCA1和BRCA2致病性突变携带者卵巢癌的预防。(更多信息,请参阅本总结中的卵巢癌风险因素章节中口服避孕药部分内容。) 尽管数据并不完全一致,但一项荟萃分析得出结论,在携带BRCA1/BRCA2致病性突变的人群中,OC的使用不会显著增加乳腺癌风险。
然而,使用1975年以前的OC制剂与乳腺癌风险的增加相关(总结相对风险[SRR],1.47;95%CI,1.06-2.04)。
(更多信息,请参阅本总结中有关BRCA致病性突变携带者的临床管理的生殖因素部分内容。)
存在来自观察性和随机临床试验的有关绝经后激素替代疗法(HRT)与乳腺癌之间关系的数据。对来自51项观察性研究的数据进行的荟萃分析表明,绝经后5年或5年以上使用HRT的女性患乳腺癌的RR为1.35(95% CI,1.21-1.49)。
WHI (NCT00000611),一项对约16,000名绝经后女性进行的随机对照试验,研究了HRT的风险和益处。在这项研究的雌性激素+孕激素组中,超过16,000名女性被随机分配到接受联合HRT或安慰剂,由于健康风险大于益处,该研究提前停止了。
促使研究关闭的不良结果包括总乳腺癌例数(245 vs.185例)和浸润性乳腺癌例数(199 vs.150例)的显著增加(RR,1.24;95% CI,1.02-1.5,P<0.001),和冠心病、中风和肺栓塞风险的增加。类似的发现也出现在英国的前瞻性、观察性的百万女性研究的雌激素-孕激素组。
然而,在WHI研究中,随机分配接受单纯雌激素组和安慰剂组的女性患乳腺癌的风险并没有升高(RR,0.77;95%CI,0.59-1.01)。在这项研究中,需要子宫切除的患者才适合进入单纯雌激素组,并且40%的患者还进行了卵巢切除术,这可能潜在地影响了乳腺癌的风险。
在有乳腺癌家族史的女性中,HRT与乳腺癌风险之间的关系并不一致;一些研究表明,有家族病史的女性患乳腺癌的风险尤其高,而另一些研究则没有发现这些因素之间存在相互作用的证据。
在大型荟萃分析中,使用HRT增加乳腺癌的风险在有家族史和无家族史的受试者之间没有显著差异。
WHI的研究没有报告对乳腺癌家族史的分层分析,也没有对受试者进行BRCA1/BRCA2致病性突变的系统检测。
用于更年期症状的短期激素治疗很少或不会增加乳腺癌风险。
HRT对BRCA1或BRCA2致病性突变携带者乳腺癌风险的影响已经在双侧风险降低卵巢切除术的背景下进行了研究,其中短期替代并不会降低卵巢切除术对乳腺癌风险的保护作用。
(更多信息,请参阅本总结中有关BRCA1/BRCA2致病性突变携带者的激素替代治疗的部分内容。)
使用激素也会影响患子宫内膜癌的风险。(更多信息,请参阅本总结中子宫内膜癌风险因素章节中的激素部分内容。)
在广岛和长崎原子弹爆炸的幸存者以及接受过胸部和上半身放射治疗的女性中所观察到的辐射增加了她们患乳腺癌的风险。这种风险因素对具有乳腺癌遗传易感性的女性的意义尚不清楚。
初步数据表明,辐射敏感性的增加可能是BRCA1或BRCA2致病性突变携带者癌症易感性的一个原因,
并与生殖系ATM和TP53突变相关。
乳腺癌的遗传易感性是通过辐射敏感性的一种机制发生的,这一可能性提出了关于辐射暴露的问题。诊断性辐射暴露,包括乳房x光检查,可能对基因易感女性造成的风险高于一般风险女性。治疗性辐射也可能造成致癌风险。然而,一项对BRCA1和BRCA2致病性突变携带者进行的保乳治疗的队列研究显示,没有证据表明携带者的乳房、肺或骨髓中的辐射敏感性或后遗症有所增加。
这一发现在一项对691名BRCA1/BRCA2相关乳腺癌患者的回顾性研究中得到了证实,这些患者的中位随访时间为8.6年。在整个研究中,没有观察到接受辅助放疗与CBC风险增加之间的联系,包括原发性乳腺癌诊断时年龄在40岁以下的患者子集。
相反,辐射敏感性可能使乳腺癌遗传易感女性的肿瘤对辐射治疗更敏感。研究辐射暴露的影响,包括但不限于,乳房x线摄影在携带BRCA1和BRCA2致病突变的携带者中有相互矛盾的结果。
欧洲的一项大型研究表明,总辐射暴露与风险增加之间存在剂量-效应关系,但这主要是由20岁之前的非乳腺X线照射的辐射暴露造成的。
随后,在一项对1844名BRCA1携带者和502名BRCA2携带者的前瞻性研究中,未发现先前的乳腺X线摄影暴露与乳腺癌风险之间存在显著的相关性;平均的随访时间为5.3年。
(有关辐射的更多信息,请参阅本总结中有关BRCA致病性突变携带者临床管理的乳腺X线摄影部分。)
在一般人群中,每天摄入10g酒精(大约一杯或更少),患乳腺癌的风险就会增加约10%。
先前对BRCA1/BRCA2致病性突变携带者的研究发现,饮酒并没有增加患病风险。
体重增加和超重是公认的乳腺癌风险因素。一般来说,超重的女性绝经后患乳腺癌的风险增加,绝经前患乳腺癌的风险降低。久坐的生活方式也可能是一个风险因素。
这些因素还没有在有乳腺癌家族史的女性或携带致癌性突变基因的人群中得到系统的评估,但一项研究表明,在携带BRCA1和BRCA2致癌性突变基因的人群中,运动可以降低患癌症的风险。
良性乳腺疾病(BBD)是乳腺癌的一个风险因素,不受其他主要风险因素(年龄、月经初潮年龄、首次活产年龄、乳腺癌家族史)的影响。
BBD和乳腺癌家族史之间也可能存在联系。
已被证实,经乳腺X线检查评估的乳腺组织密度增加的女性患乳腺癌的风险也有所增加,
而乳腺密度在其病因学中可能有遗传成分。
其他风险因素,包括那些与乳腺癌仅有微弱关联的因素,以及那些在流行病学研究中与此病不一致的因素(例如,吸烟),可能对特定基因型定义的亚组女性很重要。一项研究
发现,吸烟的BRCA1/BRCA2致病性突变携带者患乳腺癌的风险较低,但一项扩大的随访研究未发现两者之间的联系。
有关一般人群卵巢癌风险因素的信息,请参阅PDQ总结中关于卵巢癌、输卵管癌和原发性腹膜癌预防的内容。
卵巢癌的发病率从30岁到50岁呈线性增长,并在此后继续增长,但速度较慢。30岁之前,上皮性卵巢癌的发病风险很低,即使是在遗传性癌症家族中也是如此。
虽然生育、人口统计学和生活方式影响卵巢癌的风险,但最主要的卵巢癌风险因素是该疾病的家族史。一项对15项已发表研究的大型荟萃分析估计发现,至少有一位患卵巢癌的FDR与卵巢癌相关风险的OR为3.1。
未生育一直与卵巢癌风险增加相关,包括BRCA/BRCA2致病性突变携带者在内,但一项荟萃分析发现,只有在活产4例或4例以上的女性中,风险才会降低。
使用过生育药物的女性,特别是那些仍然没有生育的女性,其患病风险也可能增加。
几项研究已经报道了OC在携带BRCA1/BRCA2致病性突变的人群中使用后降低卵巢癌风险;
BRCA1携带者在输卵管结扎后卵巢癌风险也降低,在手术后风险降低了22%至80%,具有统计学意义。
在BRCA1/BRCA2致病性突变携带者中,母乳喂养超过12个月也可能与卵巢癌发病率降低有关。
另一方面,越来越多的证据表明,绝经后HRT的使用与卵巢癌风险增加有关,特别是在长期使用和连续使用雌激素-孕激素的人群中。
双侧输卵管结扎和子宫切除与降低卵巢癌风险相关,
包括BRCA1/BRCA2致病性突变的携带者。
已被证实存在BRCA1或BRCA2致病性突变的女性选择降低风险的输卵管卵巢切除术(RRSO)后,其患卵巢癌风险降低了90%以上。在相同的人群中,降低风险的卵巢切除术也使随后的乳腺癌风险降低了近50%。
虽然一些研究显示,BRCA2致病性突变对比BRCA1致病性突变对乳腺癌患者的减少更有益处,但其他研究显示BRCA1携带者没有益处。此外,许多研究仍不足以证明其益处。
(有关这些研究的更多信息,请参阅本总结中的RRSO部分。)
在一般人群中,使用OC大于等于4年,可降低约50%的患卵巢癌风险。
大多数但不是全部的研究也支持OC在BRCA1/BRCA2致病性突变携带者中起(卵巢)保护作用。
对包括13,627名BRCA致病性突变携带者在内的18项研究的荟萃分析报告了使用OC与显著降低卵巢癌风险有关(SRR,0.50;95%CI,0.33-0.75)。
(更多信息,请参阅本总结中的化学预防章节中的口服避孕药部分。)
有关一般人群子宫内膜癌风险因素的信息,请参阅PDQ总结有关子宫内膜癌预防的内容。
年龄是子宫内膜癌的重要风险因素。大多数患子宫内膜癌的女性是在绝经后才被诊断出来。只有15%的女性在50岁之前被诊断出子宫内膜癌,不到5%的女性在40岁之前被诊断出子宫内膜癌。
患有Lynch综合症的女性更容易在早期患上子宫内膜癌,诊断的中位年龄为48岁。
虽然高雌激素状态是子宫内膜癌最常见的诱发因素,但家族史在女性患病风险中也起着重要作用。约3%至5%的子宫癌病例可归因于遗传原因,
且主要遗传性子宫内膜癌综合症为Lynch综合症,这是一种常染色体显性遗传疾病,人群患病率为1/300至1/1000。
(更多信息,请参阅PDQ总结中有关结直肠癌遗传学总结中的Lynch综合症部分。)
非Lynch综合症基因也可能构成子宫内膜癌的风险。在一个未经选择的子宫内膜癌队列研究中,进行了多基因的全套检测发现,约3%的患者在非lynch综合症基因中检测出生殖系致病性突变,包括CHEK2、APC、ATM、BARD1、BRCA1、BRCA2、BRIP1、NBN、PTEN和RAD51C。
值得注意的是,非lynch综合症基因中有致病性突变的患者比无致病性突变的患者更有可能有浆液性肿瘤组织。此外,尽管在BRCA1致病性突变携带者中,RRSO治疗后子宫内膜癌的总体风险并未增加,但这些患者患浆液性和浆液样子宫内膜癌的风险增加。
生育因素如多胎产、月经初潮较晚和绝经较早可降低子宫内膜癌的风险,因为雌激素的累积暴露量较低,孕激素的相对暴露量较高。
增加I型子宫内膜癌风险的激素因素得到了较好的理解。所有的子宫内膜癌都以雌激素为主(相对于孕激素)。长期暴露于雌激素或无对抗的雌激素会增加子宫内膜癌的风险。内源性雌激素暴露可导致肥胖、多囊卵巢综合症和不能生育,而外源性雌激素可由服用高剂量的雌激素或他莫昔芬引起。高剂量的雌激素会使子宫内膜癌的风险增加2到20倍,且与使用时间成正比。
他莫昔芬是一种选择性雌激素受体调节剂,在子宫内膜上起雌激素激动剂的作用,而在乳腺组织中起雌激素拮抗剂的作用,并增加子宫内膜癌的风险。
相反,OC、左炔诺孕酮宫内释放系统和雌孕激素联合替代治疗,通过孕激素作用于子宫内膜的抗增殖作用,均可降低子宫内膜癌的风险。
乳腺癌和妇科癌症常染色体显性遗传表现为癌症易感性通过母亲或父亲家族代代相传,具有以下特征:
乳腺癌和卵巢癌是几种常染色体显性癌症综合征的组成部分。与这两种癌症最密切相关的综合征是与BRCA1或BRCA2致病突变相关的综合征。乳腺癌也是Li-Fraumeni综合征和Cowden综合征的共同特征,Li-Fraumeni综合征是由TP53致病性突变引起的,而Cowden综合征是由PTEN致病性突变引起的。
其他可能具有乳腺癌相关特征的遗传综合症包括共济失调毛细血管扩张基因杂合子携带者和Peutz-Jeghers综合症。卵巢癌还与Lynch综合症、基底细胞痣(Gorlin)综合症和多发性内分泌肿瘤I型相关。
Lynch综合症主要与结直肠癌、子宫内膜癌相关,但多项研究表明Lynch综合症患者也可发展为输尿管、肾盂移行细胞癌,胃癌、小肠癌、肝癌、胆管癌、脑癌、乳腺癌、前列腺癌和肾上腺皮质癌,皮脂腺皮肤肿瘤(Muir-Torre综合症)的风险。
在这些常染色体显性癌症综合征的基因中,生殖系致病突变产生了不同的恶性肿瘤临床表型,在某些情况下,还产生了相关的非恶性异常。
提示遗传性癌症易感性的家族特征包括:
图1和图2分别描述了BRCA1和BRCA2致病性突变的一些典型遗传特征。图3描述了一个典型的Lynch综合症家族。(请参考PDQ总结中的癌症遗传学风险评估与咨询中的标准谱系命名图,以获得这些系谱中使用的标准符号的定义。)
BRCA1或BRCA2致病性突变携带者与非携带者乳腺癌和卵巢癌的发病特点没有区别。发生在BRCA1致病突变携带者身上的乳腺癌更有可能是ER阴性、PR阴性、HER2/neu阴性(即三阴性乳腺癌[TNBC]),并具有基底表型。BRCA1相关的卵巢癌更有可能是高级别和浆液性的组织病理学。(有关更多信息,请参阅本总结中的乳腺癌病理和卵巢癌病理部分)
一些病例特征可以区分开Lynch综合症相关致病性突变携带者与非携带者。Lynch综合症子宫内膜癌的显著特征是错配修复(MMR)缺陷,包括存在微卫星不稳定性(MSI),以及缺乏特异性MMR蛋白。除了这些分子变化外,还存在组织学变化,包括肿瘤浸润淋巴细胞、瘤周淋巴细胞、未分化肿瘤组织学、子宫下段起源和同步肿瘤。
当家族史用来评估风险时,必须考虑到家族史的准确性和完整性。报告的家族史可能是错误的,或者一个人可能不知道亲属是否患有癌症。此外,家族规模小和过早死亡可能会限制从家族史中获得的信息。家族中父亲一方的乳腺癌或卵巢癌通常比母亲一方的乳腺癌或卵巢癌涉及更多的远亲,因此获取信息可能更加困难。当将自我报告的信息与独立验证的病例进行比较时,乳腺癌病史的敏感性相对较高,为83%至97%,而卵巢癌的敏感性较低,为60%。
依赖家族史的其他局限性包括收养,女性人数少的家族,限制获取家族史信息,偶然的子宫、卵巢和/或输卵管的摘除。家族史将会演化,因此随着时间的推移,从父母双方更新家族史是很重要的。(更多信息,请参阅PDQ总结中关于癌症遗传风险评估和咨询中家族史的准确性部分内容。)
预测个体一生中患乳腺癌和/或妇科癌症的风险模型是可用的。
此外,存在模型来预测个体在BRCA1、BRCA2或与Lynch综合症相关的MMR基因中致病性突变的可能性。(请参考本总结中有关BRCA1或BRCA2致病性突变可能性预测部分的模型,以获得有关这些模型的更多信息。) 并不是所有的模型都适用于所有的患者。只有当患者的特征和家族史与该模型所基于的研究人群相似时,该模型才是合适的。不同的模型可能会为相同的临床情景提供差异很大的风险评估,而且许多模型尚未进行这些评估的验证。
一般而言,乳腺癌风险评估模型是针对两类人群设计的:1)无乳腺癌或卵巢癌的致病性突变或无强烈的乳腺癌或卵巢癌家族史的女性;2)有乳腺癌或卵巢癌家族史的女性患乳腺癌的风险更高。
为第一种类型的女性设计的模型(例如,Gail模型,它是乳腺癌风险评估工具的基础[BCRAT])
,以及Colditz和Rosner模型
)只要求提供有限的家族史资料(例如,患乳癌的FDR的数目)。为高危女性设计的模型需要更详细的个人和家族乳腺癌和卵巢癌病史信息,包括发病年龄和/或特定乳腺癌易感等位基因的携带状态。后一种模型使用的遗传因素不同,有些模型假设一个风险位点(如Claus模型
)、其他假设两个位点(例如,国际乳腺癌干预研究[IBIS]模型
和BRCAPRO模型
),以及其他假设除了多位点外还有额外多基因成分的研究(例如,乳腺和卵巢疾病发病率分析及携带者估计算法[BOADICEA]模型
)。这些模型在是否包括非遗传风险因素的信息方面也有所不同。三个模型(Gail/BCRAT, Pfeiffer,
和IBIS)包含了非遗传风险因素,但包含的风险因素不同(例如,Pfeiffer模型包括饮酒,而Gail/BCRAT不包括)。这些模型在区分患病的个体和未患癌症的个体方面的能力有限;一个具有高识别率模型的接近1,而一个低识别率的模型接近0.5;目前这些模型的识别率在0.56-0.63之间)。
在评估预测未来癌症的前瞻性研究中,现有的模型通常更准确。
一项对BOADICEA、BRCAPRO、BCRAT和IBIS模型的10年性能的比较分析表明,具有更详细谱系的模型更具优越性,特别是BOADICEA和IBIS模型。
在美国,BRCAPRO,这种Claus模型,
和Gail/BCRAT
广泛应用于临床咨询。根据模型得出的风险估计因患者个体而异。其他一些包含更详细的家族史信息的模型也在使用中,下面将对此进行讨论。
Gail模型是BCRAT的基础,BCRAT是通过拨打癌症信息服务电话1-800-4-CANCER(1-800-422-6237)从美国国家癌症研究所(NCI)获得的一种计算机程序。这个版本的Gail模型只估计了浸润性乳腺癌的风险。Gail/BCRAT模型已被发现在预测每年接受乳腺X线检查的大量白人女性的乳腺癌风险方面相当准确;然而,可靠性因研究队列而异。。
以下人群的风险可能被高估:
Gail/BCRAT模型适用于35岁以上的女性。该模型最初是为白人女性而开发的。
随后,针对美国非裔女性的Gail扩展模型被开发出来,利用来自1600多名患有侵袭性乳腺癌的非裔美国妇女和1600多名对照者的数据,对风险估计进行了校准。
此外,Gail扩展模型纳入了高危单核苷酸多态性和致病性突变;然而,在这些扩展模型中没有软件可以计算风险。
其他用于乳房密度的风险评估模型已经开发出来,但还没有准备好用于临床。
一般来说,Gail/BCRAT模型不应是用于具有以下一种或多种特征的家族的唯一模型:
结合家族史的常用模型包括IBIS、BOADICEA和BRCAPRO模型。IBIS/Tyrer-Cuzick模型融合了遗传和非遗传因素。
一个三代系谱可用于评估个体携带BRCA1/BRCA2致病性突变或假设的低外显率基因的可能性。此外,该模型还考虑了个人风险因素,如胎次、体重指数(BMI);身高;和月经初潮年龄、首次活产年龄、绝经年龄,和使用HRT。遗传和非遗传因素均结合在一起进行风险评估。BOADICEA模型通过检查家族史,并结合BRCA1/BRCA2和非BRCA1/BRCA2基因风险因素来评估乳腺癌风险。
BOADICEA与其他利用BRCA1/BRCA2信息的模型之间最重要的区别在于,BOADICEA除了具有多个位点外,还有一个额外的多基因组分,
这更符合我们对乳腺癌潜在基因的了解。BOADICEA模型也被扩展到包括额外的致病性突变,包括CHEK2、ATM和PALB2。
然而,在独立样本中,这些模型的识别和校准存在显著差异;
IBIS和BOADICEA模型在估计较短且固定时间范围(例如,10年)内的风险时更具可比性,
而不是剩余的终生风险。由于所有的癌症风险评估模型一般都是在较短的时间范围内(如5年或10年)进行验证的,因此固定时间范围的估计可能在临床环境中更准确和更有用,而不是剩余的终身风险。
此外,根据女性的风险因素,提供有关女性个体风险与人群风险的信息的现成模型可能在临床环境中有用(例如,您的疾病风险)。虽然这个工具是应用关于平均风险女性的信息开发的,并不能计算绝对风险估计,但它在向女性提供预防咨询时仍然是有用的。风险评估模型正在大量开发和验证中,以整合遗传和非遗传数据、乳房密度和其他生物标志物。
目前已经建立了两种卵巢癌的风险预测模型。
Rosner模型
包括了绝经年龄、初潮年龄、口服避孕药的使用和输卵管结扎;一致性的统计量为0.60(0.57-0.62)。Pfeiffer模型
包括了使用口服避孕药情况、绝经期激素治疗的使用情况、以及乳腺癌或卵巢癌家族史,具有相似的区分能力0.59(0.56-0.62)。尽管这两种模型都经过了很好的校准,但其微弱的区分能力限制了它们的筛选潜力。
Pfeiffer模型已被用于预测一般人群的子宫内膜癌风险。
对于子宫内膜癌,相对风险模型包括BMI、绝经期激素治疗的使用情况、绝经状态、绝经年龄、吸烟状态和OC的使用情况。模型的区分能力为0.68 (0.66-0.70);它高估了大多数亚组观察到的子宫内膜癌,但低估了BMI指数最高的女性、绝经前女性和接受绝经激素治疗10年或10年以上的女性的患病情况。
相反,MMRpredict、PREMM5(基因突变预测模型)和MMRpro是三种定量预测模型,用于识别可能患有Lynch综合症的个体。
MMRpredict只包括结直肠癌患者,但包括MSI和免疫组化(IHC)肿瘤检测结果。PREMM5是PREMM(1,2,6)的更新,包含与Lynch综合症相关的5个基因,包括PMS2和EPCAM。它包括其他Lynch综合症相关的肿瘤,但不包括肿瘤检测结果。
MMRpro整合了肿瘤检测和生殖系检测结果,但由于它在风险量化过程中包含了受影响和未受影响的个体,因此需要更多的时间。这三种预测模型在识别携带MMR基因致病性突变的结直肠癌患者时,均与传统的Amsterdam和Bethesda标准具有可比性。
然而,由于这些模型是在结直肠癌患者中开发和验证的,这些模型对Lynch综合症的区分能力在子宫内膜癌患者中低于结肠癌患者。
事实上,MSI和IHC在识别致病性突变携带者方面的敏感性和特异性大大高于预测模型,并支持使用分子肿瘤检测来筛查子宫内膜癌患者的Lynch综合症。
表1总结了在临床环境中常用的乳腺癌和妇科癌症风险评估模型的显著特点。这些模型因包括家族史的程度、是否包括非遗传风险因素、是否包括携带者状态和多基因风险(模型的输入)而有所不同。模型在生成的风险估计类型(模型的输出)方面也有所不同。这些因素可能与选择最适合特定个体的模型有关。
模型 | 家族史(输入) | 致病性突变(输入) | 风险因素(输入) | 生成的风险估计(输出) |
---|---|---|---|---|
乳腺癌风险评估模型 | ||||
一般风险女性的模型 | ||||
Gail/BCRAT | 一级亲属(乳腺癌) | 否 | 是 | 乳腺癌 |
Pfeiffer(乳腺) | 一级亲属(乳腺癌、卵巢癌) | 否 | 是 | 乳腺癌 |
Colditz和Rosner | 无 | 否 | 是 | 乳腺癌 |
高风险女性的模型 | ||||
Claus | 多基因(乳腺癌) | 否 | 否 | 乳腺癌 |
BRCAPRO | 多代(乳腺癌、卵巢癌) | BRCA1和BRCA2 | 否 | 乳腺癌;携带BRCA1/BRCA2致病性突变的风险% |
IBIS | 多代(卵巢癌) | BRCA1和BRCA2 | 是 | 乳腺癌;携带BRCA1/BRCA2致病性突变的风险% |
BOADICEA | 多代(胰腺癌、乳腺癌、卵巢癌) | BRCA1和BRCA2 | 无 | 乳腺癌和卵巢癌;携带BRCA1/BRCA2致病性突变的风险% |
卵巢癌风险评估模型 | ||||
一般风险女性的模型 | ||||
Rosner | 无 | 否 | 是 | 卵巢癌 |
Pfeiffer(卵巢) | 一级亲属(乳癌、卵巢癌) | 否 | 是 | 乳腺癌 |
高危女性模型 | ||||
BOADICEA | 多代(胰腺癌、乳腺癌、卵巢癌) | BRCA1和BRCA2 | 否 | 乳腺癌和卵巢癌;携带BRCA1/BRCA2致病性突变的风险% |
子宫内膜癌风险评估模型 | ||||
一般风险女性的模型 | ||||
Pfeiffer(子宫内膜) | 无 | 否 | 是 | 子宫内膜癌 |
PREMM5 | 多代(结肠、子宫内膜和其他Lynch综合症相关的癌症和息肉) | 否 | 否 | 携带MLH1、MSH2、MSH6致病性突变的风险% |
MMRpro | 多代(结肠癌、子宫内膜癌) | 否 | 否 | 携带MLH1、MSH2、MSH6致病性突变的风险% |
MMRpredict | 多代(结肠癌、子宫内膜癌) | 否 | 否 | 携带MLH1、MSH2、MSH6致病性突变的风险% |
BCRAT=乳腺癌风险评估工具;BOADICEA=乳腺和卵巢疾病发病率分析及携带者估计算法;IBIS=国际乳腺癌干预研究;PREMM=基因突变预测模型。 | ||||
a 高危人群指的是那些有特定癌症类型的个人史或家族史的人。 | ||||
b 将多基因作为模型的基本假设。 |
一般风险女性的模型
一般风险女性的模型
高危女性模型
一般风险女性的模型
一些专业机构和专家小组——包括美国临床肿瘤学会、美国国家综合癌症网络(NCCN),美国人类遗传学学会、美国医学遗传学和基因组学学会,国家遗传顾问协会,美国预防服务工作组,妇科肿瘤医师协会-已经制定了临床标准和实践指南,可以帮助卫生保健提供者识别可能具有BRCA1或BRCA2致病突变的个人。
2019年,美国乳腺外科医师协会(American Society of Breast Surgeons)发布了一项建议,让所有乳腺癌患者都可以进行“BRCA1/BRCA2和PALB2,以及其他适合临床情况和家族史的基因”的基因测试。
这一建议是基于一项研究,该研究表明,通过一个扩展的多基因面板,在那些符合或不符合NCCN基因检测指南的乳腺癌患者中发现了类似的致病性突变率。
本研究有重要的方法学挑战需要考虑,包括排除之前检测过的受试者、不确定风险标准报告的准确性、纳入包含不确定管理指南的基因、以及两组人群中致病或可能致病性突变的特定基因的差异。例如,在检测BRCA1/BRCA2突变时,符合和不符合NCCN标准的参与者之间存在显著的统计学差异。其他研究也发现,NCCN标准在预测BRCA1/BRCA2突变时具有良好的敏感性;然而,对许多其他基因的预测了解较少。例如,一项研究表明,NCCN标准能够检测到88.9%的BRCA1/BRCA2致病性突变携带者,
而其他研究发现,如果满足一个以上的NCCN标准,那么阳性预测值会超过10%的阈值(例如,超过两个NCCN标准的阳性预测值为12%)。
随着基因检测的成本持续下降,一旦获得无偏移的证据,则基因检测的适应症,包括对筛查、预防和治疗的成本效益的影响,可能会扩大。
在进行新的癌症诊断时,癌症遗传易感性的基因检测可以指导病人的护理,包括关于手术、化疗和其他生物制剂以及放射治疗的决策。
在高危患者中,基因检测的选择是诊断癌症时有关癌症治疗的共同决策过程的重要组成部分。在这种情况下,可以使用工具来促进有关基因检测的决策。
乳腺癌诊断时进行基因检测的好处包括但不限于:
卵巢癌诊断时进行基因检测的获益包括但不限于以下几点:
子宫内膜癌诊断时进行基因检测的获益包括但不限于:
由于新一代测序的可用性和美国最高法院裁定人类基因不能获得专利,一些临床实验室现在通过多基因面板提供基因检测,其成本相当于单基因测试。即使是对BRCA1和BRCA2的检测,也是对两个基因的有限的面板检测。大约25%的卵巢癌/输卵管癌/腹膜癌是由遗传性疾病引起的。其中,约四分之一(所有卵巢/输卵管/腹膜癌的6%)是由BRCA1和BRCA2以外的基因引起的,包括许多与Fanconi贫血通路相关或与同源重组相关的基因。
在对BRCA1和BRCA2致病性突变检测呈阴性的卵巢癌患者人群中,多基因面板检测可以揭示可操作的致病性突变。
一般而言,多基因面板检测可增加各种人群中非BRCA致病性突变的捕获量。
在未经选择的乳腺癌患者群体中,BRCA1和BRCA2致病性突变的患病率为6.1%,而其他乳腺癌/卵巢癌易感基因致病性突变的患病率为4.6%。
在未经选择的子宫内膜癌患者中,Lynch综合症致病性突变(MLH1、MSH2、EPCAM-MSH2、MSH6和PMS2)的发生率为5.8%;致病性突变在其他可操作基因中的发生率为3.4%。
类似地,在一项对35,409名乳腺癌患者进行的Myriad 25基因面板检测中,9.3%的女性发现了致病性突变。
在这9.3%中,48.5%的女性携带BRCA1或BRCA2的致病性突变。发现的其他致病性突变的乳腺癌基因主要包括CHEK2(11.7%)、ATM(9.7%)和PALB2(9.3%)。面板中其他乳腺癌基因的致病性突变发生率在0.05%到0.31%之间。Lynch综合症中的致病性突变占突变总数的7.0%;3.7%在该面板中的其他基因中发现。在40岁以前诊断为TNBC的女性中,致病性突变的比例更高。对男性乳腺癌患者非BRCA易感基因中识别致病性突变也有类似的趋势。
在之前对BRCA1/BRCA2阴性的女性进行的两项研究中,用多基因面板进行的反射测试在8%到11%的病例中发现了额外基因的致病性突变。
在一项对77,085名乳腺癌患者和6001名卵巢癌患者的研究中,分别有24.1%和30.9%的患者接受了基因检测。在那些被检测的患者中,有7.8%的乳腺癌患者和14.5%的卵巢癌患者发现了致病或可能致病的突变。乳腺癌患者中常见的非BRCA致病性突变包括CHEK2(1.6%)、PALB2(1.0%)、ATM(0.7%)和NBN(0.4%)。在卵巢癌患者中,非BRCA致病性突变包括CHEK2(1.4%)、BRIP1(0.9%)、MSH2(0.8%)和ATM(0.6%)。
目前建议年龄小于60岁的TNBC患者进行BRCA1/BRCA2检测,以指导制定治疗决策的。
一项利用多基因(面板)检测的大型研究包括两个独立的队列,该研究报道,除了BRCA1/BRCA2基因外,其他六个乳腺癌易感基因也与TNBC的更高风险相关。具体而言,除了BRCA1和BRCA2外,BARD1、PALB2和RAD51D中的致病性突变与乳腺癌的发病率均增加了5倍以上有关。
其他三个基因的致病性突变——BRIP1、RAD51C和TP53——都与两倍以上的TNBC风险增加相关。这8个基因的致病性突变在12%的TNBC病例中被报道(8.3%的BRCA1/BRCA2,3.7%的非BRCA1/BRCA2)。该研究在临床试验队列中进行,共有140449人(8753个TNBC病例)接受了21个基因面板(样本A)的基因检测。此外,第二组样本(样本B)使用17个基因面板检测了2143人的基因频率。在样本A中检测的21个基因中,致病性突变的总频率为14.4% (8.4%的BRCA1/BRCA2,6.0%的非BRCA1/BRCA2)。尽管在年龄、种族和家族史方面存在差异,但这两个样本在风险估计方面的结果非常一致。样本A更年轻,人种和种族更多样化,更可能有癌症家族史。这21个基因的致病性突变频率检测结果与白人(总体占14%,BRCA1/BRCA2占7.8%,非BRCA1/BRCA2占6.2%)和非裔美国人(总体占14.6%,BRCA1/BRCA2占9.0%,非BRCA1/BRCA2占5.6%)相似,这支持了非裔美国人TNBC相对于白人的高发病率是由环境因素驱动的。
关于多基因检测有几点需要注意。作为多基因检测的一部分基因可能与不同的乳腺癌风险相关,也可能引起未知的风险。
也有可能找到不确定意义的突变。即使在一个特定的基因中,也可能存在不同的致病性突变。
现在许多中心提供了多基因面板检测,而不只是BRCA1和BRCA2基因面板检测,如果有关于家族史的遗传性乳腺癌和卵巢癌以外的症状,或更重要的是,在一个检测中获得尽可能多的遗传信息,特别是如果有保险的局限性的情况。
(更多有关多基因检测的信息,包括遗传教育和咨询考虑以及研究多基因检测的使用,请参阅PDQ总结中的癌症遗传风险评估和咨询中的多基因检测部分。)
Among women in the United States, breast cancer is the most commonly diagnosed cancer after nonmelanoma skin cancer, and it is the second leading cause of cancer deaths after lung cancer. In 2020, an estimated 279,100 new cases of breast cancer (including 2,620 cases in men) will be diagnosed, and 42,690 deaths (including 520 deaths in men) will occur.
The incidence of breast cancer, particularly for estrogen receptor (ER)–positive cancers occurring after age 50 years, is declining and has declined at a faster rate since 2003; this may be temporally related to a decrease in hormone replacement therapy (HRT) after early reports from the Women’s Health Initiative (WHI).
An estimated 21,750 new cases of ovarian cancer are expected in the United States in 2020, with an estimated 13,940 deaths. Ovarian cancer is the fifth most deadly cancer in women.
An estimated 65,620 new cases of endometrial cancer are expected in the United States in 2020, with an estimated 12,590 deaths.
(Refer to the PDQ summaries on Breast Cancer Treatment [Adult]; Ovarian Epithelial, Fallopian Tube, and Primary Peritoneal Cancer Treatment; and Endometrial Cancer Treatment for more information about breast, ovarian, and endometrial cancer rates, diagnosis, and management.)
A possible genetic contribution to both breast and ovarian cancer risk is indicated by the increased incidence of these cancers among women with a family history (refer to the Risk Factors for Breast Cancer, Risk Factors for Ovarian Cancer, and Risk Factors for Endometrial Cancer sections below for more information), and by the observation of some families in which multiple family members are affected with breast and/or ovarian cancer, in a pattern compatible with an inheritance of autosomal dominant cancer susceptibility. Formal studies of families (linkage analysis) have subsequently proven the existence of autosomal dominant predispositions to breast and ovarian cancer and have led to the identification of several highly penetrant genes as the cause of inherited cancer risk in many families. (Refer to the PDQ summary Cancer Genetics Overview for more information about linkage analysis.) Pathogenic variants in these genes are rare in the general population and are estimated to account for no more than 5% to 10% of breast and ovarian cancer cases overall. It is likely that other genetic factors contribute to the etiology of some of these cancers.
Refer to the PDQ summary on Breast Cancer Prevention for information about risk factors for breast cancer in the general population.
The cumulative risk of breast cancer increases with age, with most breast cancers occurring after age 50 years.
Breast cancer (and ovarian cancer, to a lesser degree) tends to occur at an earlier age in women with a genetic susceptibility than it does in women with sporadic cases.
In cross-sectional studies of adult populations, 5% to 10% of women have a mother or sister with breast cancer, and about twice as many have either a first-degree relative (FDR) or a second-degree relative with breast cancer.
The risk conferred by a family history of breast cancer has been assessed in case-control and cohort studies, using volunteer and population-based samples, with generally consistent results.
In a pooled analysis of 38 studies, the relative risk (RR) of breast cancer conferred by an FDR with breast cancer was 2.1 (95% confidence interval [CI], 2.0–2.2).
Risk increases with the number of affected relatives, age at diagnosis, the occurrence of bilateral or multiple ipsilateral breast cancers in a family member, and the number of affected male relatives.
A large population-based study from the Swedish Family Cancer Database confirmed the finding of a significantly increased risk of breast cancer in women who had a mother or a sister with breast cancer. The hazard ratio (HR) for women with a single breast cancer in the family was 1.8 (95% CI, 1.8–1.9) and was 2.7 (95% CI, 2.6–2.9) for women with a family history of multiple breast cancers. For women who had multiple breast cancers in the family, with one occurring before age 40 years, the HR was 3.8 (95% CI, 3.1–4.8). However, the study also found a significant increase in breast cancer risk if the relative was aged 60 years or older, suggesting that breast cancer at any age in the family carries some increase in risk.
Another study in women with unilateral versus contralateral breast cancer (CBC) evaluated breast cancer risk among family members.
Results indicated that among women with affected FDRs, CBC risk was 8.1% at 10 years. This risk was higher among relatives diagnosed before age 40 years or with CBC, and approached the lower risk estimates among BRCA carriers. (Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section in the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section of this summary for information about cancer risk estimates in that population.) These risk estimates remained unchanged when the analysis was restricted to women who tested negative for a deleterious variant in BRCA1/BRCA2, ATM, CHEK2, and PALB2.
One of the largest studies of twins ever conducted, with 80,309 monozygotic twins and 123,382 dizygotic twins, reported a heritability estimate for breast cancer of 31% (95% CI, 11%–51%).
If a monozygotic twin had breast cancer, her twin sister had a 28.1% probability of developing breast cancer (95% CI, 23.9%–32.8%), and if a dizygotic twin had breast cancer, her twin sister had a 19.9% probability of developing breast cancer (95% CI, 17%–23.2%). These estimates suggest a 10% higher risk of breast cancer for monozygotic twins than for dizygotic twins. However, a high rate of discordance even between monozygotic twins suggests that environmental factors also have a role in modifying breast cancer risk.
(Refer to the Penetrance of BRCA pathogenic variants section of this summary for a discussion of familial risk in women from families with BRCA1/BRCA2 pathogenic variants who themselves test negative for the family pathogenic variant.)
In general, breast cancer risk increases with early menarche and late menopause and is reduced by early first full-term pregnancy. There may be an increased risk of breast cancer in carriers of BRCA1 and BRCA2 pathogenic variants with pregnancy at a younger age (before age 30 y), with a more significant effect seen for carriers of BRCA1 pathogenic variants.
Likewise, breastfeeding can reduce breast cancer risk in carriers of BRCA1 (but not BRCA2) pathogenic variants.
Regarding the effect of pregnancy on breast cancer outcomes, neither diagnosis of breast cancer during pregnancy nor pregnancy after breast cancer seems to be associated with adverse survival outcomes in women who carry a BRCA1 or BRCA2 pathogenic variant.
Parity appears to be protective for carriers of BRCA1 and BRCA2 pathogenic variants, with an additional protective effect for live birth before age 40 years.
Reproductive history can also affect the risk of ovarian cancer and endometrial cancer. (Refer to the Reproductive History sections in the Risk Factors for Ovarian Cancer and Risk Factors for Endometrial Cancer sections of this summary for more information.)
Oral contraceptives (OCs) may produce a slight increase in breast cancer risk among long-term users, but this appears to be a short-term effect. In a meta-analysis of data from 54 studies, the risk of breast cancer associated with OC use did not vary in relationship to a family history of breast cancer.
OCs are sometimes recommended for ovarian cancer prevention in carriers of BRCA1 and BRCA2 pathogenic variants. (Refer to the Oral Contraceptives section in the Risk Factors for Ovarian Cancer section of this summary for more information.) Although the data are not entirely consistent, a meta-analysis concluded that there was no significant increased risk of breast cancer with OC use in carriers of BRCA1/BRCA2 pathogenic variants.
However, use of OCs formulated before 1975 was associated with an increased risk of breast cancer (summary relative risk [SRR], 1.47; 95% CI, 1.06–2.04).
(Refer to the Reproductive factors section in the Clinical Management of Carriers of BRCA Pathogenic Variants section of this summary for more information.)
Data exist from both observational and randomized clinical trials regarding the association between postmenopausal HRT and breast cancer. A meta-analysis of data from 51 observational studies indicated a RR of breast cancer of 1.35 (95% CI, 1.21–1.49) for women who had used HRT for 5 or more years after menopause.
The WHI (NCT00000611), a randomized controlled trial of about 160,000 postmenopausal women, investigated the risks and benefits of HRT. The estrogen-plus-progestin arm of the study, in which more than 16,000 women were randomly assigned to receive combined HRT or placebo, was halted early because health risks exceeded benefits.
Adverse outcomes prompting closure included significant increase in both total (245 vs. 185 cases) and invasive (199 vs. 150 cases) breast cancers (RR, 1.24; 95% CI, 1.02–1.5, P < . 001) and increased risks of coronary heart disease, stroke, and pulmonary embolism. Similar findings were seen in the estrogen-progestin arm of the prospective observational Million Women’s Study in the United Kingdom.
The risk of breast cancer was not elevated, however, in women randomly assigned to estrogen-only versus placebo in the WHI study (RR, 0.77; 95% CI, 0.59–1.01). Eligibility for the estrogen-only arm of this study required hysterectomy, and 40% of these patients also had undergone oophorectomy, which potentially could have impacted breast cancer risk.
The association between HRT and breast cancer risk among women with a family history of breast cancer has not been consistent; some studies suggest risk is particularly elevated among women with a family history, while others have not found evidence for an interaction between these factors.
The increased risk of breast cancer associated with HRT use in the large meta-analysis did not differ significantly between subjects with and without a family history.
The WHI study has not reported analyses stratified on breast cancer family history, and subjects have not been systematically tested for BRCA1/BRCA2 pathogenic variants.
Short-term use of hormones for treatment of menopausal symptoms appears to confer little or no breast cancer risk.
The effect of HRT on breast cancer risk among carriers of BRCA1 or BRCA2 pathogenic variants has been studied in the context of bilateral risk-reducing oophorectomy, in which short-term replacement does not appear to reduce the protective effect of oophorectomy on breast cancer risk.
(Refer to the Hormone replacement therapy in carriers of BRCA1/BRCA2 pathogenic variants section of this summary for more information.)
Hormone use can also affect the risk of developing endometrial cancer. (Refer to the Hormones section in the Risk Factors for Endometrial Cancer section of this summary for more information.)
Observations in survivors of the atomic bombings of Hiroshima and Nagasaki and in women who have received therapeutic radiation treatments to the chest and upper body document increased breast cancer risk as a result of radiation exposure. The significance of this risk factor in women with a genetic susceptibility to breast cancer is unclear.
Preliminary data suggest that increased sensitivity to radiation could be a cause of cancer susceptibility in carriers of BRCA1 or BRCA2 pathogenic variants,
and in association with germline ATM and TP53 variants.
The possibility that genetic susceptibility to breast cancer occurs via a mechanism of radiation sensitivity raises questions about radiation exposure. It is possible that diagnostic radiation exposure, including mammography, poses more risk in genetically susceptible women than in women of average risk. Therapeutic radiation could also pose a carcinogenic risk. A cohort study of carriers of BRCA1 and BRCA2 pathogenic variants treated with breast-conserving therapy, however, showed no evidence of increased radiation sensitivity or sequelae in the breast, lung, or bone marrow of carriers.
This finding was confirmed in a retrospective cohort study of 691 patients with BRCA1/BRCA2-associated breast cancer who were followed up for a median of 8.6 years. No association between receiving adjuvant radiation therapy and increased risk of CBC was observed in the entire cohort, including the subset of patients younger than 40 years at primary breast cancer diagnosis.
Conversely, radiation sensitivity could make tumors in women with genetic susceptibility to breast cancer more responsive to radiation treatment. Studies examining the impact of radiation exposure, including, but not limited to, mammography, in carriers of BRCA1 and BRCA2 pathogenic variants have had conflicting results.
A large European study showed a dose-response relationship of increased risk with total radiation exposure, but this was primarily driven by nonmammographic radiation exposure before age 20 years.
Subsequently, no significant association was observed between prior mammography exposure and breast cancer risk in a prospective study of 1,844 BRCA1 carriers and 502 BRCA2 carriers without a breast cancer diagnosis at time of study entry; average follow-up time was 5.3 years.
(Refer to the Mammography section in the Clinical Management of Carriers of BRCA Pathogenic Variants section of this summary for more information about radiation.)
The risk of breast cancer increases by approximately 10% for each 10 g of daily alcohol intake (approximately one drink or less) in the general population.
Prior studies of carriers of BRCA1/BRCA2 pathogenic variants have found no increased risk associated with alcohol consumption.
Weight gain and being overweight are commonly recognized risk factors for breast cancer. In general, overweight women are most commonly observed to be at increased risk of postmenopausal breast cancer and at reduced risk of premenopausal breast cancer. Sedentary lifestyle may also be a risk factor.
These factors have not been systematically evaluated in women with a positive family history of breast cancer or in carriers of cancer-predisposing pathogenic variants, but one study suggested a reduced risk of cancer associated with exercise among carriers of BRCA1 and BRCA2 pathogenic variants.
Benign breast disease (BBD) is a risk factor for breast cancer, independent of the effects of other major risk factors for breast cancer (age, age at menarche, age at first live birth, and family history of breast cancer).
There may also be an association between BBD and family history of breast cancer.
An increased risk of breast cancer has also been demonstrated for women who have increased density of breast tissue as assessed by mammogram,
and breast density is likely to have a genetic component in its etiology.
Other risk factors, including those that are only weakly associated with breast cancer and those that have been inconsistently associated with the disease in epidemiologic studies (e.g., cigarette smoking), may be important in women who are in specific genotypically defined subgroups. One study
found a reduced risk of breast cancer among carriers of BRCA1/BRCA2 pathogenic variants who smoked, but an expanded follow-up study failed to find an association.
Refer to the PDQ summary on Ovarian, Fallopian Tube, and Primary Peritoneal Cancer Prevention for information about risk factors for ovarian cancer in the general population.
Ovarian cancer incidence rises in a linear fashion from age 30 years to age 50 years and continues to increase, though at a slower rate, thereafter. Before age 30 years, the risk of developing epithelial ovarian cancer is remote, even in hereditary cancer families.
Although reproductive, demographic, and lifestyle factors affect risk of ovarian cancer, the single greatest ovarian cancer risk factor is a family history of the disease. A large meta-analysis of 15 published studies estimated an odds ratio of 3.1 for the risk of ovarian cancer associated with at least one FDR with ovarian cancer.
Nulliparity is consistently associated with an increased risk of ovarian cancer, including among carriers of BRCA/BRCA2 pathogenic variants, yet a meta-analysis identified a risk reduction only in women with four or more live births.
Risk may also be increased among women who have used fertility drugs, especially those who remain nulligravid.
Several studies have reported a risk reduction in ovarian cancer after OC use in carriers of BRCA1/BRCA2 pathogenic variants;
a risk reduction has also been shown after tubal ligation in BRCA1 carriers, with a statistically significant decreased risk of 22% to 80% after the procedure.
Breastfeeding for more than 12 months may also be associated with a reduction in ovarian cancer among carriers of BRCA1/BRCA2 pathogenic variants.
On the other hand, evidence is growing that the use of menopausal HRT is associated with an increased risk of ovarian cancer, particularly in long-time users and users of sequential estrogen-progesterone schedules.
Bilateral tubal ligation and hysterectomy are associated with reduced ovarian cancer risk,
including in carriers of BRCA1/BRCA2 pathogenic variants.
Ovarian cancer risk is reduced more than 90% in women with documented BRCA1 or BRCA2 pathogenic variants who chose risk-reducing salpingo-oophorectomy (RRSO). In this same population, risk-reducing oophorectomy also resulted in a nearly 50% reduction in the risk of subsequent breast cancer.
While some studies have shown more benefit for breast cancer reduction in patients with BRCA2 versus BRCA1 pathogenic variants, others have shown no benefit for BRCA1 carriers. Additionally, many of the studies remain underpowered to demonstrate benefit.
(Refer to the RRSO section of this summary for more information about these studies.)
Use of OCs for 4 or more years is associated with an approximately 50% reduction in ovarian cancer risk in the general population.
A majority of, but not all, studies also support OCs being protective among carriers of BRCA1/BRCA2 pathogenic variants.
A meta-analysis of 18 studies including 13,627 carriers of BRCA pathogenic variants reported a significantly reduced risk of ovarian cancer (SRR, 0.50; 95% CI, 0.33–0.75) associated with OC use.
(Refer to the Oral contraceptives section in the Chemoprevention section of this summary for more information.)
Refer to the PDQ summary on Endometrial Cancer Prevention for information about risk factors for endometrial cancer in the general population.
Age is an important risk factor for endometrial cancer. Most women with endometrial cancer are diagnosed after menopause. Only 15% of women are diagnosed with endometrial cancer before age 50 years, and fewer than 5% are diagnosed before age 40 years.
Women with Lynch syndrome tend to develop endometrial cancer at an earlier age, with the median age at diagnosis of 48 years.
Although the hyperestrogenic state is the most common predisposing factor for endometrial cancer, family history also plays a significant role in a woman’s risk for disease. Approximately 3% to 5% of uterine cancer cases are attributable to a hereditary cause,
with the main hereditary endometrial cancer syndrome being Lynch syndrome, an autosomal dominant genetic condition with a population prevalence of 1 in 300 to 1 in 1,000 individuals.
(Refer to the Lynch Syndrome section in the PDQ summary on Genetics of Colorectal Cancer for more information.)
Non-Lynch syndrome genes may also contribute to endometrial cancer risk. In an unselected endometrial cancer cohort undergoing multigene panel testing, approximately 3% of patients tested positive for a germline pathogenic variant in non-Lynch syndrome genes, including CHEK2, APC, ATM, BARD1, BRCA1, BRCA2, BRIP1, NBN, PTEN, and RAD51C.
Notably, patients with pathogenic variants in non-Lynch syndrome genes were more likely to have serous tumor histology than were patients without pathogenic variants. Furthermore, although the overall risk of endometrial cancer after RRSO was not increased among carriers of BRCA1 pathogenic variants, these patients seemed to have an increased risk of serous and serous-like endometrial cancer.
Reproductive factors such as multiparity, late menarche, and early menopause decrease the risk of endometrial cancer because of the lower cumulative exposure to estrogen and the higher relative exposure to progesterone.
Hormonal factors that increase the risk of type I endometrial cancer are better understood. All endometrial cancers share a predominance of estrogen relative to progesterone. Prolonged exposure to estrogen or unopposed estrogen increases the risk of endometrial cancer. Endogenous exposure to estrogen can result from obesity, polycystic ovary syndrome, and nulliparity, while exogenous estrogen can result from taking unopposed estrogen or tamoxifen. Unopposed estrogen increases the risk of developing endometrial cancer by twofold to twentyfold, proportional to the duration of use.
Tamoxifen, a selective estrogen receptor modulator, acts as an estrogen agonist on the endometrium while acting as an estrogen antagonist in breast tissue, and increases the risk of endometrial cancer.
In contrast, OCs, the levonorgestrel-releasing intrauterine system, and combination estrogen-progesterone hormone replacement therapy all reduce the risk of endometrial cancer through the antiproliferative effect of progesterone acting on the endometrium.
Autosomal dominant inheritance of breast and gynecologic cancers is characterized by transmission of cancer predisposition from generation to generation, through either the mother’s or the father’s side of the family, with the following characteristics:
Breast and ovarian cancer are components of several autosomal dominant cancer syndromes. The syndromes most strongly associated with both cancers are the syndromes associated with BRCA1 or BRCA2 pathogenic variants. Breast cancer is also a common feature of Li-Fraumeni syndrome due to TP53 pathogenic variants and of Cowden syndrome due to PTEN pathogenic variants.
Other genetic syndromes that may include breast cancer as an associated feature include heterozygous carriers of the ataxia telangiectasia gene and Peutz-Jeghers syndrome. Ovarian cancer has also been associated with Lynch syndrome, basal cell nevus (Gorlin) syndrome, and multiple endocrine neoplasia type 1.
Lynch syndrome is mainly associated with colorectal cancer and endometrial cancer, although several studies have demonstrated that patients with Lynch syndrome are also at risk of developing transitional cell carcinoma of the ureters and renal pelvis; cancers of the stomach, small intestine, liver and biliary tract, brain, breast, prostate, and adrenal cortex; and sebaceous skin tumors (Muir-Torre syndrome).
Germline pathogenic variants in the genes responsible for these autosomal dominant cancer syndromes produce different clinical phenotypes of characteristic malignancies and, in some instances, associated nonmalignant abnormalities.
The family characteristics that suggest hereditary cancer predisposition include the following:
Figure 1 and Figure 2 depict some of the classic inheritance features of a BRCA1 and BRCA2 pathogenic variant, respectively. Figure 3 depicts a classic family with Lynch syndrome. (Refer to the Standard Pedigree Nomenclature figure in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for definitions of the standard symbols used in these pedigrees.)
There are no pathognomonic features distinguishing breast and ovarian cancers occurring in carriers of BRCA1 or BRCA2 pathogenic variants from those occurring in noncarriers. Breast cancers occurring in carriers of BRCA1 pathogenic variants are more likely to be ER-negative, progesterone receptor–negative, HER2/neu receptor–negative (i.e., triple-negative breast cancers [TNBC]), and have a basal phenotype. BRCA1-associated ovarian cancers are more likely to be high-grade and of serous histopathology. (Refer to the Pathology of breast cancer and Pathology of ovarian cancer sections of this summary for more information.)
Some pathologic features distinguish carriers of Lynch syndrome–associated pathogenic variants from noncarriers. The hallmark feature of endometrial cancers occurring in Lynch syndrome is mismatch repair (MMR) deficiencies, including the presence of microsatellite instability (MSI), and the absence of specific MMR proteins. In addition to these molecular changes, there are also histologic changes including tumor-infiltrating lymphocytes, peritumoral lymphocytes, undifferentiated tumor histology, lower uterine segment origin, and synchronous tumors.
The accuracy and completeness of family histories must be taken into account when they are used to assess risk. A reported family history may be erroneous, or a person may be unaware of relatives affected with cancer. In addition, small family sizes and premature deaths may limit the information obtained from a family history. Breast or ovarian cancer on the paternal side of the family usually involves more distant relatives than does breast or ovarian cancer on the maternal side, so information may be more difficult to obtain. When self-reported information is compared with independently verified cases, the sensitivity of a history of breast cancer is relatively high, at 83% to 97%, but lower for ovarian cancer, at 60%.
Additional limitations of relying on family histories include adoption; families with a small number of women; limited access to family history information; and incidental removal of the uterus, ovaries, and/or fallopian tubes for noncancer indications. Family histories will evolve, therefore it is important to update family histories from both parents over time. (Refer to the Accuracy of the family history section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information.)
Models to predict an individual’s lifetime risk of developing breast and/or gynecologic cancer are available.
In addition, models exist to predict an individual’s likelihood of having a pathogenic variant in BRCA1, BRCA2, or one of the MMR genes associated with Lynch syndrome. (Refer to the Models for prediction of the likelihood of a BRCA1 or BRCA2 pathogenic variant section of this summary for more information about some of these models.) Not all models can be appropriately applied to all patients. Each model is appropriate only when the patient’s characteristics and family history are similar to those of the study population on which the model was based. Different models may provide widely varying risk estimates for the same clinical scenario, and the validation of these estimates has not been performed for many models.
In general, breast cancer risk assessment models are designed for two types of populations: 1) women without a pathogenic variant or strong family history of breast or ovarian cancer; and 2) women at higher risk because of a personal or family history of breast cancer or ovarian cancer.
Models designed for women of the first type (e.g., the Gail model, which is the basis for the Breast Cancer Risk Assessment Tool [BCRAT])
, and the Colditz and Rosner model
) require only limited information about family history (e.g., number of FDRs with breast cancer). Models designed for women at higher risk require more detailed information about personal and family cancer history of breast and ovarian cancers, including ages at onset of cancer and/or carrier status of specific breast cancer-susceptibility alleles. The genetic factors used by the latter models differ, with some assuming one risk locus (e.g., the Claus model
), others assuming two loci (e.g., the International Breast Cancer Intervention Study [IBIS] model
and the BRCAPRO model
), and still others assuming an additional polygenic component in addition to multiple loci (e.g., the Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm [BOADICEA] model
). The models also differ in whether they include information about nongenetic risk factors. Three models (Gail/BCRAT, Pfeiffer,
and IBIS) include nongenetic risk factors but differ in the risk factors they include (e.g., the Pfeiffer model includes alcohol consumption, whereas the Gail/BCRAT does not). These models have limited ability to discriminate between individuals who are affected and those who are unaffected with cancer; a model with high discrimination would be close to 1, and a model with little discrimination would be close to 0.5; the discrimination of the models currently ranges between 0.56 and 0.63).
The existing models generally are more accurate in prospective studies that have assessed how well they predict future cancers.
An analysis comparing the 10-year performance of the BOADICEA, BRCAPRO, BCRAT, and IBIS models demonstrated superiority of the models with more detailed pedigree inclusion—specifically, BOADICEA and IBIS.
In the United States, BRCAPRO, the Claus model,
and the Gail/BCRAT
are widely used in clinical counseling. Risk estimates derived from the models differ for an individual patient. Several other models that include more detailed family history information are also in use and are discussed below.
The Gail model is the basis for the BCRAT, a computer program available from the National Cancer Institute (NCI) by calling the Cancer Information Service at 1-800-4-CANCER (1-800-422-6237). This version of the Gail model estimates only the risk of invasive breast cancer. The Gail/BCRAT model has been found to be reasonably accurate at predicting breast cancer risk in large groups of white women who undergo annual screening mammography; however, reliability varies depending on the cohort studied.
Risk can be overestimated in the following populations:
The Gail/BCRAT model is valid for women aged 35 years and older. The model was primarily developed for white women.
Extensions of the Gail model for African American women have been subsequently developed to calibrate risk estimates using data from more than 1,600 African American women with invasive breast cancer and more than 1,600 controls.
Additionally, extensions of the Gail model have incorporated high-risk single nucleotide polymorphisms and pathogenic variants; however, no software exists to calculate risk in these extended models.
Other risk assessment models incorporating breast density have been developed but are not ready for clinical use.
Generally, the Gail/BCRAT model should not be the sole model used for families with one or more of the following characteristics:
Commonly used models that incorporate family history include the IBIS, BOADICEA, and BRCAPRO models. The IBIS/Tyrer-Cuzick model incorporates both genetic and nongenetic factors.
A three-generation pedigree is used to estimate the likelihood that an individual carries either a BRCA1/BRCA2 pathogenic variant or a hypothetical low-penetrance gene. In addition, the model incorporates personal risk factors such as parity, body mass index (BMI); height; and age at menarche, first live birth, menopause, and HRT use. Both genetic and nongenetic factors are combined to develop a risk estimate. The BOADICEA model examines family history to estimate breast cancer risk and also incorporates both BRCA1/BRCA2 and non-BRCA1/BRCA2 genetic risk factors.
The most important difference between BOADICEA and the other models using information on BRCA1/BRCA2 is that BOADICEA assumes an additional polygenic component in addition to multiple loci,
which is more in line with what is known about the underlying genetics of breast cancer. The BOADICEA model has also been expanded to include additional pathogenic variants, including CHEK2, ATM, and PALB2.
However, the discrimination and calibration for these models differ significantly when compared in independent samples;
the IBIS and BOADICEA models are more comparable when estimating risk over a shorter fixed time horizon (e.g., 10 years),
than when estimating remaining lifetime risk. As all risk assessment models for cancers are typically validated over a shorter time horizon (e.g., 5 or 10 years), fixed time horizon estimates rather than remaining lifetime risk may be more accurate and useful measures to convey in a clinical setting.
In addition, readily available models that provide information about an individual woman’s risk in relation to the population-level risk depending on her risk factors may be useful in a clinical setting (e.g., Your Disease Risk). Although this tool was developed using information about average-risk women and does not calculate absolute risk estimates, it still may be useful when counseling women about prevention. Risk assessment models are being developed and validated in large cohorts to integrate genetic and nongenetic data, breast density, and other biomarkers.
Two risk prediction models have been developed for ovarian cancer.
The Rosner model
included age at menopause, age at menarche, oral contraception use, and tubal ligation; the concordance statistic was 0.60 (0.57–0.62). The Pfeiffer model
included oral contraceptive use, menopausal hormone therapy use, and family history of breast cancer or ovarian cancer, with a similar discriminatory power of 0.59 (0.56–0.62). Although both models were well calibrated, their modest discriminatory power limited their screening potential.
The Pfeiffer model has been used to predict endometrial cancer risk in the general population.
For endometrial cancer, the relative risk model included BMI, menopausal hormone therapy use, menopausal status, age at menopause, smoking status, and OC use. The discriminatory power of the model was 0.68 (0.66–0.70); it overestimated observed endometrial cancers in most subgroups but underestimated disease in women with the highest BMI category, in premenopausal women, and in women taking menopausal hormone therapy for 10 years or more.
In contrast, MMRpredict, PREMM5 (PREdiction Model for gene Mutations), and MMRpro are three quantitative predictive models used to identify individuals who may potentially have Lynch syndrome.
MMRpredict incorporates only colorectal cancer patients but does include MSI and immunohistochemistry (IHC) tumor testing results. PREMM5 is an update of PREMM(1,2,6) and includes each of the five genes associated with Lynch syndrome, including PMS2 and EPCAM. It accounts for other Lynch syndrome–associated tumors but does not include tumor testing results.
MMRpro incorporates tumor testing and germline testing results, but is more time intensive because it includes affected and unaffected individuals in the risk-quantification process. All three predictive models are comparable to the traditional Amsterdam and Bethesda criteria in identifying individuals with colorectal cancer who carry MMR gene pathogenic variants.
However, because these models were developed and validated in colorectal cancer patients, the discriminative abilities of these models to identify Lynch syndrome are lower among individuals with endometrial cancer than among those with colon cancer.
In fact, the sensitivity and specificity of MSI and IHC in identifying carriers of pathogenic variants are considerably higher than the prediction models and support the use of molecular tumor testing to screen for Lynch syndrome in women with endometrial cancer.
Table 1 summarizes salient aspects of breast and gynecologic cancer risk assessment models that are commonly used in the clinical setting. These models differ by the extent of family history included, whether nongenetic risk factors are included, and whether carrier status and polygenic risk are included (inputs to the models). The models also differ in the type of risk estimates that are generated (outputs of the models). These factors may be relevant in choosing the model that best applies to a particular individual.
Model | Family History (input) | Pathogenic Variants (input) | Risk Factors (input) | Risk Estimate Generated (output) |
---|---|---|---|---|
Breast Cancer Risk Assessment Models | ||||
Models for Average-Risk Women | ||||
Gail/BCRAT | First-degree relatives (breast cancer) | No | Yes | Breast cancer |
Pfeiffer (breast) | First-degree relatives (breast, ovarian cancers) | No | Yes | Breast cancer |
Colditz and Rosner | None | No | Yes | Breast cancer |
Models for High-Risk Women | ||||
Claus | Multigenerational (breast cancer) | No | No | Breast cancer |
BRCAPRO | Multigenerational (breast, ovarian cancers) | BRCA1/BRCA2 | No | Breast cancer; % risk of carrying BRCA1/BRCA2 pathogenic variant |
IBIS | Multigenerational (ovarian cancer) | BRCA1/BRCA2 | Yes | Breast cancer; % risk of carrying BRCA1/BRCA2 pathogenic variant |
BOADICEA | Multigenerational (pancreatic, breast, ovarian cancers) | BRCA1/BRCA2 | No | Breast and ovarian cancer; % risk of carrying BRCA1/BRCA2 pathogenic variant |
Ovarian Cancer Risk Assessment Models | ||||
Models for Average-Risk Women | ||||
Rosner | None | No | Yes | Ovarian cancer |
Pfeiffer (ovarian) | First-degree relatives (breast, ovarian cancers) | No | Yes | Breast cancer |
Models for High-Risk Womena | ||||
BOADICEA | Multigenerational (pancreatic, breast, ovarian cancers) | BRCA1/BRCA2 | No | Breast and ovarian cancer; % risk of carrying BRCA1/BRCA2 pathogenic variant |
Endometrial Cancer Risk Assessment Models | ||||
Models for Average-Risk Women | ||||
Pfeiffer (endometrial) | None | No | Yes | Endometrial cancer |
PREMM5 | Multigenerational (colon, endometrial and other Lynch syndrome–associated cancers and polyps) | No | No | % risk of carrying MLH1, MSH2, MSH6 pathogenic variant |
MMRpro | Multigenerational (colon, endometrial cancers) | No | No | % risk of carrying MLH1, MSH2, MSH6 pathogenic variant |
MMRpredict | Multigenerational (colon, endometrial cancers) | No | No | % risk of carrying MLH1, MSH2, MSH6 pathogenic variant |
BCRAT = Breast Cancer Risk Assessment Tool; BOADICEA = Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm; IBIS = International Breast Cancer Intervention Study; PREMM = PREdiction Model for gene Mutations. | ||||
aHigh risk is defined as those with a personal or family history of the designated cancer type. | ||||
bTakes into account polygenes as an underlying assumption of the model. |
Models for Average-Risk Women
Models for Average-Risk Women
Models for High-Risk Womena
Models for Average-Risk Women
Several professional organizations and expert panels— including the American Society of Clinical Oncology, the National Comprehensive Cancer Network (NCCN), the American Society of Human Genetics, the American College of Medical Genetics and Genomics, the National Society of Genetic Counselors, the U.S. Preventive Services Task Force, and the Society of Gynecologic Oncologists — have developed clinical criteria and practice guidelines that can be helpful to health care providers in identifying individuals who may have a BRCA1 or BRCA2 pathogenic variant.
In 2019, the American Society of Breast Surgeons published a recommendation to make genetic testing for “BRCA1/BRCA2, and PALB2, with other genes as appropriate for the clinical scenario and family history” available to all breast cancer patients.
This recommendation was based on a study that suggested similar pathogenic variant rates identified through an extended multigene panel in patients with breast cancer who did or did not meet the NCCN guidelines for genetic testing.
This study had important methodologic challenges that need to be considered, including exclusion of participants previously tested, uncertain accuracy of the reported risk criteria for study participants, inclusion of genes with uncertain management guidelines, and difference in the specific genes in which pathogenic or likely pathogenic variants were identified across the two groups. For example, there was a statistically significant difference between participants who met and who did not meet NCCN criteria in the detection of BRCA1/BRCA2 variants. Other studies have also found that the NCCN criteria have good sensitivity when predicting BRCA1/BRCA2 variants; however, less is known about many other genes. For example, one study showed that the NCCN criteria were able to detect 88.9% of the BRCA1/BRCA2 pathogenic variant carriers
and others have found that, if more than one NCCN criterion is met, then the positive predictive value does pass the 10% threshold (e.g., 12% for more than two NCCN criteria).
As the cost of genetic testing continues to decrease, the indications for testing, including the cost-benefit impact on screening, prevention, and treatment, may expand once unbiased evidence becomes available.
At the time of a new cancer diagnosis, genetic testing for inherited cancer predisposition may guide patient care including decisions about surgery, chemotherapy and other biologics, and radiation treatment.
Among high-risk patients, the option of genetic testing is an important part of the shared decision-making process regarding cancer treatments at the time of diagnosis. Tools are available to facilitate decision making about genetic testing in this context.
Benefits of offering genetic testing at the time of breast cancer diagnosis include, but are not limited to, the following:
Benefits of offering genetic testing at the time of ovarian cancer diagnosis include, but are not limited to, the following:
Benefits of offering genetic testing at the time of endometrial cancer diagnosis include, but are not limited to, the following:
Since the availability of next-generation sequencing and the Supreme Court of the United States ruling that human genes cannot be patented, several clinical laboratories now offer genetic testing through multigene panels at a cost comparable to that of single-gene testing. Even testing for BRCA1 and BRCA2 is a limited panel test of two genes. Approximately 25% of all ovarian/fallopian tube/peritoneal cancers are caused by a heritable genetic condition. Of these, about one-quarter (6% of all ovarian/fallopian tube/peritoneal cancers) are caused by genes other than BRCA1 and BRCA2, including many genes associated with the Fanconi anemia pathway or otherwise involved with homologous recombination.
In a population of ovarian cancer patients who test negative for BRCA1 and BRCA2 pathogenic variants, multigene panel testing can reveal actionable pathogenic variants.
In general, multigene panel testing increases the yield of non-BRCA pathogenic variants across a variety of populations.
In an unselected population of breast cancer patients, the prevalence of BRCA1 and BRCA2 pathogenic variants was 6.1%, while the prevalence of pathogenic variants in other breast/ovarian cancer–predisposing genes was 4.6%.
In an unselected population of endometrial cancer patients, the prevalence of Lynch syndrome pathogenic variants (MLH1, MSH2, EPCAM-MSH2, MSH6, and PMS2) was 5.8%; the prevalence of pathogenic variants in other actionable genes was 3.4%.
Similarly, in a study of 35,409 women with breast cancer tested with the Myriad 25-gene panel, a pathogenic variant was found in 9.3% of women.
Among that 9.3%, 48.5% of the women carried a pathogenic variant in BRCA1 or BRCA2. The majority of other breast cancer genes with pathogenic variants identified included CHEK2 (11.7%), ATM (9.7%), and PALB2 (9.3%). The prevalence of pathogenic variants in the other breast cancer genes on the panel ranged from 0.05% to 0.31%. Pathogenic variants in Lynch syndrome genes accounted for 7.0% of variants identified; 3.7% were found in other genes included in the panel. The rate of pathogenic variants was higher in women with TNBC diagnosed before age 40 years. A similar trend of identifying pathogenic variants in non-BRCA susceptibility genes in male breast cancer patients has also been described.
In two studies of women who had previously tested negative for BRCA1/BRCA2, reflex testing with a multigene panel identified pathogenic variants in additional genes among 8% to 11% of cases.
In a study of 77,085 patients with breast cancer and 6,001 patients with ovarian cancer, 24.1% and 30.9% had genetic testing, respectively. Of those tested, pathogenic or likely pathogenic variants were identified in 7.8% of patients with breast cancer and 14.5% of patients with ovarian cancer. Prevalent non-BRCA pathogenic variants identified in patients with breast cancer included CHEK2 (1.6%), PALB2 (1.0%), ATM (0.7%), and NBN (0.4%). In patients with ovarian cancer, non-BRCA pathogenic variants included CHEK2 (1.4%), BRIP1 (0.9%), MSH2 (0.8%), and ATM (0.6%).
Women younger than 60 years diagnosed with TNBC are currently recommended to undergo BRCA1/BRCA2 testing to guide treatment decisions.
A large study utilizing multigene (panel) testing comprising two separate cohorts reported that, in addition to BRCA1/BRCA2 genes, six other breast cancer susceptibility genes were also related to a higher risk of TNBC. Specifically, pathogenic variants in BARD1, PALB2, and RAD51D, in addition to BRCA1 and BRCA2, were each associated with more than a fivefold increase in breast cancer.
Pathogenic variants in three other genes —BRIP1, RAD51C, and TP53— were each associated with an increased TNBC risk of more than twofold. Pathogenic variants in these eight genes were reported in 12% of the TNBC cases (8.3% BRCA1/BRCA2, 3.7% non-BRCA1/BRCA2). The study was conducted in a clinical testing cohort of 140,449 individuals (8,753 TNBC cases) who received genetic testing using a 21-gene panel (sample A). In addition, a second sample (sample B) examined gene frequency rates in a pooled consortium of 2,143 individuals using a 17-gene panel. The overall frequency of pathogenic variants in the 21 genes examined in sample A was 14.4% (8.4% BRCA1/BRCA2, 6.0% non-BRCA1/BRCA2). The two samples had very consistent findings with respect to the risk estimates despite differences in age, race/ethnicity, and family history of cancer with sample A being younger, more racially and ethnically diverse, and more likely to have a family history of cancer. The pathogenic variant frequency detection in these 21 genes was also similar for whites (14% overall, 7.8% BRCA1/BRCA2, 6.2% non-BRCA1/BRCA2) and African Americans (14.6% overall, 9.0% BRCA1/BRCA2, 5.6% non-BRCA1/BRCA2) supporting that the higher rates of TNBC in African Americans versus whites is driven by environmental factors.
There are caveats of multigene testing. Genes identified as part of multigene panel testing can be associated with varied breast cancer risk or confer no known risk.
There is also the possibility of finding a variant of uncertain significance. Even within a given gene, there may be differential risks on the basis of specific pathogenic variants.
Many centers now offer a multigene panel test instead of just BRCA1 and BRCA2 testing if there is a concerning family history of syndromes other than hereditary breast and ovarian cancer, or more importantly, to gain as much genetic information as possible with one test, particularly if there may be insurance limitations.
(Refer to the Multigene [panel] testing section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information about multigene testing, including genetic education and counseling considerations and research examining the use of multigene testing.)
携带致病性突变的个体表现出某种疾病的比例被称为外显率。一般来说,与癌症易感性相关的常见遗传突变的外显率低于罕见遗传突变。如图4中所示。对于成年发病的疾病,外显率通常由个体携带者的年龄、性别和器官部位来描述。例如,BRCA1致病性突变的女性携带者的乳腺癌外显率通常在50岁和70岁时被引用。在众多估算外显率的方法中,没有一种是没有潜在偏倚的,因而确定个体携带者患癌症的风险涉及到一定程度的不精确性。
在整个总结中,我们讨论了报告相对和绝对风险的研究。这是两个重要但不同的概念。相对风险(RR)指的是相对于另一组人群的风险估计(例如,暴露于某一风险因素的女性患乳腺癌的风险相对于未暴露于同一风险因素的女性患乳腺癌的风险)。若RR值大于1,则表示在分子中捕获的风险(即暴露的风险)要高于分母中捕捉到的风险(即未暴露的风险)。若RR值小于1,则表示分子中所包含的风险(即暴露的风险)比分母中捕捉到的风险(即未暴露的风险)要低。具有相似相关解释的测量方法包括比值比(OR)、风险比和危险比。
绝对风险评估考虑了有特定结局的人数、可能有结局的人数和时间(个体具有结局风险的时间段),并反映了人群中结局的绝对负担。绝对度量包括风险和概率,并可以在特定的时间段(例如1年、5年)或整个生命周期内表示。累积风险是在一定时期内发生的风险指标。例如,总体终生风险是一种累积风险,通常是根据给定的预期寿命来计算的(例如,80或90岁)。累积风险也可以在其他时间段内呈现(例如,直到50岁)。
较大的相对风险测量并不意味着在人群水平上的实际个体数量会有大的影响,因为疾病结局可能是相当罕见的。例如,吸烟对肺癌的相对风险比对心脏病高得多,但吸烟者和不吸烟者之间的绝对差异对心脏病(更常见的结果)要比肺癌(罕见的结果)大得多。
因此,在评估暴露和生物标记对疾病预防的影响时,必须认识到在权衡给定风险因素的总体影响时相对影响和绝对影响之间的差异。例如,对于许多乳腺癌的风险因素,相对作用的大小是在30%的范围(例如,OR或RR为1.3),这意味着存在风险因素(如饮酒、第一次生育晚龄、使用口服避孕药、绝经后体型)的女性与她们曾有但却没有该风险因素相比,有30%的乳腺癌的相对增加。但是风险的绝对增加是基于疾病的潜在绝对风险。图5和表2显示了1.3范围内的相对风险因素对绝对风险的影响。(参考PDQ《癌症遗传学风险评估和咨询摘要》中的标准谱系命名图,了解这些系谱中使用的标准符号的定义。)如表所示,有乳腺癌家族史的女性从绝对风险因素降低中获益更高。
家族史 | 终生风险(%) | 风险因素修正后的终生风险(%) | 绝对风险差(%) | 相对风险 |
---|---|---|---|---|
低(家族1) | 10.9 | 8.4 | 2.5 | 1.29(增加29%的风险) |
中(家族2) | 21.6 | 16.8 | 4.8 | 1.28(增加28%的风险) |
中/高(家族3) | 27.1 | 21.3 | 5.8 | 1.27(增加27%的风险) |
高(家族4) | 32 | 25.3 | 6.7 | 1.26(增加26%的风险) |
BRCA1致病性突变(家族5) | 53.7 | 44.2 | 9.5 | 1.21(增加21%的风险) |
a 请参见与此表相对应的图5。 |
随着多基因面板检测的日益广泛使用,在新基因中检测到致病性突变的个体中建立的癌症风险管理框架已被描述,
这其中包括了特定年龄、寿命和绝对癌症风险的数据。该框架建议,在这些人的5年癌症风险接近某个水平时即开始进行筛查,该水平是一般人群中的女性开始进行常规筛查的风险水平(对美国乳腺癌患者而言约为1%)。因此,开始筛查的年龄将因基因而异。(更多关于多基因面板检测的信息,请参阅本总结引言部分的多基因[面板]检测部分。)
The proportion of individuals carrying a pathogenic variant who will manifest a certain disease is referred to as penetrance. In general, common genetic variants that are associated with cancer susceptibility have a lower penetrance than rare genetic variants. This is depicted in Figure 4. For adult-onset diseases, penetrance is usually described by the individual carrier's age, sex, and organ site. For example, the penetrance for breast cancer in female carriers of BRCA1 pathogenic variants is often quoted by age 50 years and by age 70 years. Of the numerous methods for estimating penetrance, none are without potential biases, and determining an individual carrier's risk of cancer involves some level of imprecision.
Throughout this summary, we discuss studies that report on relative and absolute risks. These are two important but different concepts. Relative risk (RR) refers to an estimate of risk relative to another group (e.g., risk of an outcome like breast cancer for women who are exposed to a risk factor relative to the risk of breast cancer for women who are unexposed to the same risk factor). RR measures that are greater than 1 mean that the risk for those captured in the numerator (i.e., the exposed) is higher than the risk for those captured in the denominator (i.e., the unexposed). RR measures that are less than 1 mean that the risk for those captured in the numerator (i.e., the exposed) is lower than the risk for those captured in the denominator (i.e., the unexposed). Measures with similar relative interpretations include the odds ratio (OR), hazard ratio, and risk ratio.
Absolute risk measures take into account the number of people who have a particular outcome, the number of people in a population who could have the outcome, and person-time (the period of time during which an individual was at risk of having the outcome), and reflect the absolute burden of an outcome in a population. Absolute measures include risks and rates and can be expressed over a specific time frame (e.g., 1 year, 5 years) or overall lifetime. Cumulative risk is a measure of risk that occurs over a defined time period. For example, overall lifetime risk is a type of cumulative risk that is usually calculated on the basis of a given life expectancy (e.g., 80 or 90 years). Cumulative risk can also be presented over other time frames (e.g., up to age 50 years).
Large relative risk measures do not mean that there will be large effects in the actual number of individuals at a population level because the disease outcome may be quite rare. For example, the relative risk for smoking is much higher for lung cancer than for heart disease, but the absolute difference between smokers and nonsmokers is greater for heart disease, the more-common outcome, than for lung cancer, the more-rare outcome.
Therefore, in evaluating the effect of exposures and biological markers on disease prevention across the continuum, it is important to recognize the differences between relative and absolute effects in weighing the overall impact of a given risk factor. For example, the magnitude is in the range of 30% (e.g., ORs or RRs of 1.3) for many breast cancer risk factors, which means that women with a risk factor (e.g., alcohol consumption, late age at first birth, oral contraceptive use, postmenopausal body size) have a 30% relative increase in breast cancer in comparison with what they would have if they did not have that risk factor. But the absolute increase in risk is based on the underlying absolute risk of disease. Figure 5 and Table 2 show the impact of a relative risk factor in the range of 1.3 on absolute risk. (Refer to the Standard Pedigree Nomenclature figure in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for definitions of the standard symbols used in these pedigrees.) As shown, women with a family history of breast cancer have a much higher benefit from risk factor reduction on an absolute scale.
Family History | Lifetime Risk (%) | Lifetime Risk After Risk Factor Modification (%) | Absolute Risk Difference (%) | Relative Risk |
---|---|---|---|---|
Low (Family 1) | 10.9 | 8.4 | 2.50 | 1.29 (29% increased risk) |
Moderate (Family 2) | 21.6 | 16.8 | 4.80 | 1.28 (28% increased risk) |
Moderate/high (Family 3) | 27.1 | 21.3 | 5.80 | 1.27 (27% increased risk) |
High (Family 4) | 32.0 | 25.3 | 6.70 | 1.26 (26% increased risk) |
BRCA1 pathogenic variant (Family 5) | 53.7 | 44.2 | 9.50 | 1.21 (21% increased risk) |
aRefer to Figure 5, which accompanies this table. |
With the increasing use of multigene panel tests, a framework for cancer risk management among individuals with pathogenic variants detected in novel genes has been described
that incorporates data on age-specific, lifetime, and absolute cancer risks. The framework suggests initiating screening in these individuals at the age when their 5-year cancer risk approaches that at which screening is routinely initiated for women in the general population (approximately 1% for breast cancer in the United States). As a result, the age at which to begin screening will vary depending on the gene. (Refer to the Multigene [panel] testing section in the Introduction section of this summary for more information on multigene panel tests.)
研究发现一些基因与乳腺癌和/或妇科癌症的发生有关。本总结将这些基因分为高外显率、中外显率和低外显率。表3总结了高外显率和中外显率基因。低外显率基因和位点主要包括与癌症易感性相关的多态性。(请参阅本总结的乳腺和/或妇科肿瘤易感基因高外显率、与乳腺和/或妇科肿瘤相关的中外显率基因、低外显率基因和位点部分,以获得更多信息。)
癌症易感性 | 中度外显基因 | 高外显率基因 |
---|---|---|
乳腺癌 | ATM, BRIP1, CHEK2, FANCD2, RAD51C | BRCA1, BRCA2, CDH1, PALB2, PTEN, STK11, TP53 |
卵巢癌 | ATM, BRIP1, EPCAM, MLH1, MSH2, MSH6, RAD51C | BRCA1, BRCA2 |
子宫内膜癌 | EPCAM, MLH1, MSH2, MSH6, PMS2, PTEN | |
a 其他癌症可能与这个表格中的基因有关。 | ||
b 本总结在与乳腺癌和/或妇科癌症相关的中外显率基因部分讨论了其他基因,但外显率未知的基因包括CASP8、TGFB1、Abraxas、RECQL和SMARCA4。 |
Several genes are found to be associated with the development of breast and/or gynecologic cancers. These genes are categorized as high-penetrance, moderate-penetrance, and low-penetrance in this summary. The high- and moderate-penetrance genes are summarized in Table 3. Low-penetrance genes and loci primarily include polymorphisms that have been associated with cancer susceptibility. (Refer to the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes, Moderate-Penetrance Genes Associated With Breast and/or Gynecologic Cancer, and Low-Penetrance Genes and Loci sections of this summary for more information.)
Cancer Susceptibility | Moderate-Penetrance Genes | High-Penetrance Genes |
---|---|---|
Breast cancer | ATM, BRIP1, CHEK2, FANCD2, RAD51C | BRCA1, BRCA2, CDH1, PALB2, PTEN, STK11, TP53 |
Ovarian cancer | ATM, BRIP1, EPCAM, MLH1, MSH2, MSH6, RAD51C | BRCA1, BRCA2 |
Endometrial cancer | EPCAM, MLH1, MSH2, MSH6, PMS2, PTEN | |
aOther cancers may be associated with the genes in this table. | ||
bOther genes discussed in the Moderate-Penetrance Genes Associated With Breast and/or Gynecologic Cancer section of this summary but for which penetrance is unknown include CASP8, TGFB1, Abraxas, RECQL, and SMARCA4. |
流行病学研究明确确立了家族史作为乳腺癌和卵巢癌的重要风险因素。除了性别和年龄外,阳性家族史是乳腺癌最强有力的预测因素。然而,人们早就认识到,在一些家族里,存在遗传性乳腺癌,其特点是发病的年龄早、双侧性,乳腺癌以一种明显的常染色体显性遗传的传递模式(通过母系或父系)在几代人中存在,有时也包括其它器官的肿瘤,尤其是卵巢和前列腺。
现在已知,这些家族中的某些癌症可以用单一癌症易感基因中的特定致病性突变来解释。这些基因一旦改变,就会显著增加患乳腺癌和卵巢癌的风险。尽管这些癌症易感基因非常重要,但据估计,高外显率生殖系致病性突变仅占乳腺癌总体的5%至10%。
1988年的一项研究首次报告了乳腺癌在某些家庭中作为常染色体显性特征分离的定量证据。
通过对有多个患乳腺癌个体的大家族的研究,发现了与乳腺癌遗传易感性相关的基因,并鉴定出几种易感性基因,包括BRCA1、BRCA2、TP53、PTEN/MMAC1和STK11。其他基因,如错配修复基因MLH1、MSH2、MSH6和PMS2,与卵巢癌风险增加有关,但与乳腺癌的相关性并不一致。
1990年,一个乳腺癌易感基因被发现定位于17号染色体的长臂上,区域为17q12-21。
乳腺癌和17q号染色体上的遗传标记之间的联系很快得到了其他人的证实,并观察到在相关家庭中乳腺癌和卵巢癌易感性同时传递的证据。
BRCA1基因随后通过定位克隆的方法得到确认,发现含有24个外显子,编码含1863个氨基酸的蛋白质。BRCA1的生殖系致病性突变与早发性乳腺癌、卵巢癌和输卵管癌相关。(更多信息,请参阅本总结中的BRCA致病性突变外显率部分。) 男性乳腺癌、胰腺癌、睾丸癌和早发性前列腺癌也可能与BRCA1致病性突变有关;
然而,男性乳腺癌、胰腺癌和前列腺癌与BRCA2的致病性突变有更强的相关性。
通过联合研究15个与BRCA1无关的多发性乳腺癌家族,发现了另一个乳腺癌易感基因BRCA2,其位于13号染色体的长臂上。BRCA2的致病性突变与家族中多个乳腺癌病例相关,也与男性乳腺癌、卵巢癌、前列腺癌、黑色素瘤和胰腺癌相关。
(更多信息,请参阅本总结中BRCA致病性突变的外显率部分。) BRCA2是一个大型基因,有27个外显子,编码含有3418个氨基酸的蛋白质。
虽然不是同源基因,但BRCA1和BRCA2都有一个异常大的外显子11和外显子2的转录起始点。和BRCA1一样,BRCA2是一个肿瘤抑制基因。在与BRCA1和BRCA2致病性突变相关的肿瘤中,通常会有野生型等位基因的丢失。
BRCA1和BRCA2的致病性突变似乎只在45%的有多个乳腺癌病例的家族中起作用,而在同时患有乳腺癌和卵巢癌的家族中起作用的比例高达90%。
预测大多数BRCA1和BRCA2致病性突变会产生截断的蛋白产物,从而导致蛋白功能的丧失,尽管一些错义致病性突变会导致功能丧失而不会截断。由于遗传性乳腺癌/卵巢癌是一种常染色体显性遗传疾病,在17号或13号染色体的一个拷贝上有BRCA1或BRCA2致病性突变的人,在另一对染色体上也携带正常的等位基因。在大多数已被研究的致病性突变携带者的乳腺癌和卵巢癌中,等位基因的缺失会导致所有功能的丧失,从而导致BRCA1和BRCA2被归类为肿瘤抑制基因。另外,BRCA1和BRCA2除了作为肿瘤抑制基因的一部分,还参与了细胞内的多种功能,包括同源DNA修复、基因组稳定性、转录调控、蛋白泛素化、染色质重塑和细胞周期控制。
近2000个不同的BRCA1和BRCA2的突变和序列已经被发现。
在一般人群中,大约每400到800个人中就有1人可能携带BRCA1或BRCA2的生殖系致病性突变。
突变所导致的蛋白质缺失或无功能,与癌症风险增加相关,这支持了BRCA1和BRCA2是肿瘤抑制基因的假设。虽然这些致病性突变中一小部分已经在无亲缘关系的家族中被反复发现,但大多数突变在不少的家族中未见报道。
突变筛选方法的敏感性各不相同。在研究实验室中广泛使用的方法,如单链构象多态分析和构象敏感凝胶电泳,遗漏了近三分之一的DNA测序检测到的突变。
此外,包括直接DNA测序在内的大多数技术都没有考虑到大的基因组改变,如易位、逆位、或大的缺失或插入,但对这些技术的测试在商业上是可行的。据信,这种重排导致了12%到18%的BRCA1失活突变,但在BRCA2和德系犹太人(AJ)后裔中较少见。
此外,研究表明,这种重排可能在西班牙裔和加勒比裔人群中更为常见。
BRCA1/BRCA2基因的生殖系致病性突变与60%的乳腺癌终生风险和15%至40%的卵巢癌终生风险相关。没有明确的针对BRCA1或BRCA2的功能检测;因此,核苷酸变化的分类用于预测其功能影响(是有害或无害)所依赖的数据并不不完善。大多数公认的致病性突变导致了蛋白质截断和/或失去重要的功能域。然而,在所有进行BRCA1和BRCA2全序列基因检测的个体中,有10%到15%的个体不会检测到明确的致病性突变,但会有不确定(或未知)意义的突变(VUS)。VUS可能会给咨询带来很大的挑战,尤其是在癌症风险评估和风险管理方面。这类患者的临床管理需要高度个性化,必须考虑到患者的个人和家族癌症史等因素,以及有助于将VUS定性为无害或有害的信息来源。因此,一种改进的分类和报告系统可能具有临床应用价值。
一项由Myriad遗传公司进行的7461个连续全基因序列分析进行的综合分析,描述了3年期间VUS发生的频率。
在没有明确致病性突变的受试者中,13%受试者的VUS定义为“错义突变发生在所分析的内含子区的突变,其临床意义尚未确定,分别截断BRCA1和BRCA2远端氨基酸位置1853和3308的链终止突变,和消除这些蛋白质的正常终止密码子的突变。”将序列突变分类为VUS不是一成不变的的目标。另外6.8%没有明确致病性突变的受试者存在序列的改变,这些改变曾被认为是VUS,但被重新归类为多态性,或偶尔被归类为致病性突变。
VUS的发生频率在美国人群中因种族而异。非裔美国人的VUS发生率最高。
在一项2009年的数据来自Myriad的研究中,非洲血统的个体中有16.5%的人存在VUS,这是所有种族中发生率最高的。VUS在亚洲、中东和西班牙裔人群中出现的频率在10%到14%之间,尽管这些数字是基于有限的样本量得出的。随着时间的推移,在所有种族中,VUS分类的变化率都有所下降,这在很大程度上是改进的突变分类算法的结果。
VUS继续被重新分类,作为附加信息进行编策和解释。
这些信息可能影响患病个人的持续护理。
目前已存在一些鉴别有害的和中性的VUS的方法,其他方法也在开发中
包括综合的方法(见下文)。
对VUS的解释很大程度上得益于对VUS在家庭中的追踪,以确定VUS是否与家庭中的癌症共存。一般而言,在同时具有致病性突变的个体中观察到的VUS,特别是当同一VUS与不同致病性突变一起被识别时,其本身具有危害性的可能性较低,尽管也有罕见的例外。作为临床信息的补充,用于解释VUS的模型已经开发出来,这是基于保守序列、氨基酸生化特性的变化、
合并BRCA1-和BRCA2相关肿瘤的病理特征信息(例如,与BRCA1相关的乳腺癌通常是雌激素受体[ER]阴性的),
和功能研究,以衡量特定的序列变化对BRCA1或BRCA2蛋白活性的影响。
当试图解释一个VUS时,应检查所有可用的信息。
下面提供了有关在具有各种个人癌症病史(不论家族史)的男女样本中发现携带BRCA致病突变的个体百分比的统计资料。这些数据可以帮助确定哪些人最有可能从考虑癌症遗传咨询和遗传测试中受益,但不能取代个性化风险评估,后者可能根据附加的个人和家族史特征显示更高或更低的致病性突变可能性。
在一些病例中,相同的致病性突变在多个明显不相关的家族中被发现。这一观察结果与始祖效应相一致,即在当代人群中发现的致病性突变可以追溯到一个被地理、文化或其他因素隔离的小群体。最值得注意的是,两个特定的BRCA1致病性突变(185delAG和5382insC)和一个BRCA2致病性突变(6174delT)已经被报道在德系犹太人中很常见。然而,在非裔美国人和西班牙裔美国人中也发现了其他始祖人致病性突变。
这些始祖致病性突变的存在对基因检测具有实际意义。许多实验室提供针对特定种族的等位基因的直接检测。这极大地简化了检测的技术,但也不是没有局限性。据报道,德系犹太人人群中非始祖BRCA致病性突变在3%到15%之间。
在一般人群中,发生任何BRCA致病性突变的可能性如下:
在德系犹太人(AJ)个体中,发生任何BRCA致病性突变的可能性如下:
两项针对65岁以下乳腺癌患者的大型美国人群研究检测了BRCA1和BRCA2致病性突变在不同种族中的发生率。乳腺癌患者中BRCA1致病性突变在西班牙裔患者中占3.5%,在非裔美国人中占1.3%至1.4%,在亚裔美国人中占0.5%,在非AJ白人中占2.2%至2.9%,在AJ个人中占8.3%至10.2%。非裔美国人BRCA2致病性突变的发生率为2.6%,白人发生率为2.1%。
一项对有乳腺癌和/或卵巢癌个人史或家族病史的西班牙裔患者的研究,在美国西南部的多家诊所登记,检查了BRCA1和BRCA2致病突变的发生率。在746名患者中有189人(25%)发现了BRCA致病突变(124名BRCA1, 65名BRCA2);
在189个BRCA致病性突变中有21个(11%)是大的重排,其中13个(62%)是BRCA1外显子9-12的缺失。一项未选择的包含810名墨西哥裔乳腺癌患者的队列接受了检测;4.3%的患者有BRCA1致病性突变。在发现的35个致病性突变中,有8个是BRCA1外显子9-12缺失。
在另一个纳入492名患有乳腺癌的西班牙裔女性的群体队列研究中,发现3个患者的BRCA1基因外显子9-12缺失,表明这种突变可能是墨西哥始祖致病性突变,可能代表在美国类似的诊所和群体队列研究中的所有BRCA1致病性突变的10%到12%。在临床队列中,有9个复发性致病性突变,占该队列中观察到的所有突变的53%,提示该人群中存在其他的始祖致病性突变。
一项纳入29例原发性输卵管肿瘤的AJ患者的回顾性研究发现,17%的患者发现生殖系BRCA致病性突变。
另一项纳入108名患有输卵管癌的女性的研究发现,55.6%的犹太女性和26.4%的非犹太女性有致病性突变,(总体占30.6%)。
由于对高级别、转移性、浆液性癌在最初出现时的原发部位分配不够精确,因此对BRCA致病性突变携带者中输卵管癌发生率的估计受到了限制。
人群筛查已经在许多AJ人群中发现了携带者,他们不符合基于家族的检测标准。
这可能会潜在地增加受益于预防策略的人数。一项研究表明,基于美国和英国的数据,对AJ始祖突变进行人群筛选(与基于个人/家族历史的测试相比)是具有成本效益的。
作者使用了一个决策分析模型,通过估计寿命成本和基因检测效果,从而评估成本效益;该模型包括测试前的遗传咨询和遗传测试的费用以及心血管结果的预期风险。该小组进行的其他分析也表明,当测试扩展到包括所有BRCA1、BRCA2、RAD51C、RAD51D和PALB2的致病性突变时,成本效益也很好。
这些研究基于各种假设,其中一些是不精确的(例如,对某些基因的人群患病率估计)。此外,正如作者所承认的,这些类型的努力将需要在整个照护系统中实施临床支持,以使具有致病突变的患者从这些信息中获益。因此,在考虑人口普查工作时,仍然需要大量的资源,这是目前研究工作的重点。由于检出率高度依赖于人群中致病性突变的流行程度,因此尚不清楚这种方法对其他人群(包括其他始祖致病性突变人群)是否适用。另一个未解决的问题是,是否可以为全体人群提供足够的遗传咨询。
一些研究评估了乳腺癌或卵巢癌患者中BRCA1或BRCA2致病性突变的频率。
与增加BRCA1和/或BRCA2致病性突变可能性相关的个人特征包括:
与携带BRCA1和/或BRCA2致病性突变相关的家族史特征包括:
几个专业组织和专家小组,包括美国临床肿瘤学会、国家综合癌症网络(NCCN)、美国人类遗传学学会、美国医学遗传学和基因组学学会、全国遗传顾问协会、美国预防服务工作组、和妇科肿瘤医师协会,已经制定了临床标准和实践指南,可以帮助医疗服务人员识别可能具有BRCA1或BRCA2致病性突变的个体。
许多模型已经被开发以用来预测在个体或家族中识别生殖系BRCA1/BRCA2致病性突变的可能性。这些模型包括使用逻辑回归的模型、采用贝叶斯分析的遗传模型(BRCAPRO和乳腺卵巢疾病发病率分析及携带者估计算法[BOADICEA]),和经验观察,包括Myriad患病率表。
除了BOADICEA,BRCAPRO通常也用于临床的遗传咨询。BRCAPRO 和 BOADICEA 可预测成为携带者的概率,并算出乳腺癌风险的估计值(参见表4)。这些模型的区分度和准确度(用来评估预测模型性能的因素)更高,这是因为这些模型在报告携带者状态上的能力强于他们在预测固定或剩余生命时间风险的能力。
BOADICEA是一个多基因模型,使用复杂的分离分析来检查乳腺癌风险和BRCA1或BRCA2致病性突变的可能性。
即使在有经验的提供者中,预测模型的使用已被证明增加了鉴别哪些患者最有可能是BRCA1/BRCA2致病性突变携带者的能力。
大多数模型不包括在BRCA1和BRCA2谱系中见到的其他癌症,如胰腺癌和前列腺癌。降低个体罹患癌症可能性的干预措施(如卵巢切除术和乳房切除术)可能影响预测BRCA1和BRCA2致病性突变状态的能力。
一项研究表明,遗传风险的预测模型对可获得的家族史数据量很敏感,而在家族信息有限的情况下表现不佳。
BOADICEA正被扩展以纳入额外的风险突变(全基因组关联研究[GWAS]单核苷酸多态性[SNP]),从而更好地预测致病性突变状态,并提高乳腺癌和卵巢癌风险评估的准确性。
在特定的种族群体中,这些模型的表现可能有所不同。BRCAPRO模型最适合一些法裔加拿大家族。
在拉美裔人群中,BRCAPRO模型有不同的结果,BRCAPRO模型和Myriad表格都低估了亚裔美国人中致病性突变携带者的比例。
BOADICEA是在英国女性中开发和验证的。因此,用于总体风险(表1)和遗传风险(表4)的主要模型还没有在大量的人种和种族多样化的女性中开发或验证。在用于评估遗传风险的常用临床模型中,只有Tyrer-Cuzick模型包含非遗传风险因素。
在不同的研究中比较了几种模型的把握度。
对BOADICEA、BRCAPRO、IBIS和eCLAUS四个乳腺癌遗传风险模型进行了评估,以评估它们在预测7352个德国家族中BRCA1/BRCA2致病性突变方面的诊断准确性。
对每个家族中携带致病性突变可能性最高的家族成员进行了BRCA1/BRCA2致病性突变筛查。计算每个模型的携带者概率,并与实际检测到的突变进行比较。BRCAPRO和BOADICEA的诊断准确率明显高于IBIS和eCLAUS。将肿瘤标志物ER、孕激素受体(PR)、人表皮生长因子受体2 (HER2/neu)纳入后,BOADICEA模型的准确性进一步提高。这些生物标志物的加入已被证明可以改善BRCAPRO的性能。
Myriad患病率表 | BRCAPRO | BOADICEA | Tyrer-Cuzick | |
---|---|---|---|---|
方法 | 来自 Myriad Genetics 的经验数据,基于申请表上报告的个人史和家族史 | 统计模型,假设为常染色体显性遗传 | 统计模型,假设多基因风险 | 统计模型,假设为常染色体显性遗传 |
模型的特征 | 先证者可能患有或没患有乳腺癌或卵巢癌 | 先证者可能患有或没患有乳腺癌或卵巢癌 | 先证者可能患有或没患有乳腺癌或卵巢癌 | 先证者必须未患病 |
考虑乳腺癌的年龄诊断为50岁左右 | 考虑乳腺癌和卵巢癌诊断的确切年龄 | 考虑乳腺癌和卵巢癌诊断的确切年龄 | 还包括生育因素和体重指数,以评估乳腺癌的风险 | |
仅当诊断时<50岁且≥1个亲属患病时考虑乳腺癌 | 考虑家族先前的基因检测(即,BRCA1/BRCA2致病性突变阴性的亲属) | 包括所有带有和不带有癌症的FDR和SDR | ||
考虑在任何年龄≥1个亲属的卵巢癌 | 考虑卵巢切除术的状态 | 包括AJ血统 | ||
包括AJ血统 | 包括所有患有和不患有癌症的FDR和SDR | |||
非常容易使用 | 包括AJ血统 | |||
局限性 | 对家族结构的简化/限制性考虑 | 需要电脑软件和耗时的数据输入 | 需要电脑软件和耗时的数据输入 | 专为未患乳腺癌的个体设计 |
只包含FDR和SDR;可能需要改变先证者来更好地捕捉风险并解释父亲遗传的疾病 | ||||
可能高估了双侧乳腺癌的风险 | ||||
早期乳腺癌的发病年龄 | 白人可能比少数族裔表现更好 | 只包含FDR和SDR:可能需要改变先证者以最佳捕获风险 | ||
可能低估了高级别浆液性卵巢癌中BRCA致病性突变的风险,但高估了其他组织学风险 | ||||
AJ=德系犹太人;BOADICEA=乳腺和卵巢疾病发病率分析及携带者估计算法;FDR=一级亲属;SDR =二级亲属。 |
自1996年以来,公众就可以进行BRCA1和BRCA2致病性突变的基因检测。随着越来越多的人进行检测,风险评估模型也得到了改进。反过来,这又为医疗服务人员提供了更好的数据,以评估单个患者携带致病性突变的风险,但风险评估仍然是一门艺术。有一些因素可能会限制提供准确风险评估的能力(如家族规模小、女性缺少、或种族),其中包括个别病人的具体情况(如疾病史或降低手术风险的手术史)。
携带致病性突变的个体表现出该疾病的比例被称为外显率。(更多信息,请参考总结中的遗传性乳腺癌和/或妇科癌症遗传易感性外显率部分。)
许多研究已经估计了BRCA1和BRCA2致病性突变携带者的乳腺癌和卵巢癌外显率。据一致性的估计,乳腺癌和卵巢癌的风险在携带BRCA1致病性突变的人群中要高于携带BRCA2致病性突变的人群。两项大型荟萃分析的结果如表5所示。
一项研究分析了来自22项研究的合并系谱数据,其中涉及289个BRCA1和221个BRCA2致病性突变阳性的个体。这些研究中的指标病例有女性乳腺癌、男性乳腺癌、或卵巢癌,但因家族史而未选择。随后的一项研究结合了以前的研究和其他9个研究的外显率估计,纳入了另外的734个BRCA1和400个BRCA2致病性突变阳性的家族。在这两项荟萃分析中,估计BRCA1致病性突变携带者到70岁时乳腺癌的累积风险为55%至65%,而BRCA2致病性突变携带者的累积风险为45%至47%。BRCA1致病性突变携带者的卵巢癌风险为39%,而BRCA2致病性突变携带者的卵巢癌风险为11%至17%。
研究 | 乳腺癌风险(%)(95% CI) | 卵巢癌风险(%)(95% CI) | ||
---|---|---|---|---|
BRCA1 | BRCA2 | BRCA1 | BRCA2 | |
Antoniou et al.(2003) | 65 (44–78) | 45 (31–56) | 39 (18–54) | 11 (2.4–19) |
Chen et al.(2007) | 55 (50–59) | 47 (42–51) | 39 (34–45) | 17 (13–21) |
Kuchenbaecker et al.(2017) | 72 (65–79) | 69 (61–77) | 44 (36–53) | 17 (11–25) |
CI=置信区间。 | ||||
a 到70岁估计的风险。 | ||||
b 到80岁估计的风险。 |
虽然BRCA1致病性突变携带者到70岁时患癌症的累积风险高于BRCA2致病性突变携带者,但随着BRCA1致病性突变携带者年龄的增长,乳腺癌的相对风险(RR)下降得更多。
对特定个体突变携带者的外显率的研究通常不足以提供稳定的估计,但对德系犹太人致病性突变的大量研究已经开展。一组研究人员从较大的荟萃分析中分析了具有德系犹太人之一的致病性突变的家族子集,发现各个致病性突变的估计外显率与所有携带者的相应估计非常相似。
最近一项对4649名BRCA致病性突变的女性的研究报告显示,BRCA2 6174delT突变的女性乳腺癌的RR明显低于其他BRCA2突变的女性(风险比[HR],0.35;置信区间[CI],0.18-0.69)。
一项研究提供了无症状携带者在不同年龄罹患癌症的10年预期风险。
然而,对单个携带者进行精确的外显率估计是困难的。RAD51C致病性突变携带者的卵巢癌终生风险为5.2%,BRIP1致病性突变携带者的卵巢癌终生风险为5.8%,RAD51D致病性突变携带者的卵巢癌终生风险为12%。这些女性若完成生育,可进行降低风险的输卵管卵巢切除术(RRSO)。
来自BRCA1/BRCA2修饰研究者联盟(CIMBA)的数据,CIMBA纳入了19,581名BRCA1致病性突变携带者和11,900名BRCA2致病性突变携带者,分析了根据致病性突变类型、功能和核苷酸位置估计乳腺癌和卵巢癌的HR。
在这两个基因中都发现了乳腺癌基因簇区和卵巢癌基因簇区。乳腺癌和卵巢癌的发病率风险和诊断年龄因不同类别而异。这些发现需要进一步的评估,然后才能转化为临床实践。
CIMBA组的另一项研究观察了乳腺癌患者的表型,这些患者同时具有BRCA1和BRCA2的致病性突变。
大多数女性携带常见的犹太致病性突变。与相同致病性突变杂合(杂合子对照)的女性相比,BRCA1和BRCA2双杂合的女性比杂合子对照的女性更容易被诊断出患有乳腺癌,且比携带BRCA2杂合子对照的女性更有可能被诊断为卵巢癌,但不是那些携带BRCA1致病性突变的女性。同样,与携带BRCA2的杂合子对照女性相比,携带这两种突变的女性的乳腺癌发病年龄更小,但与携带BRCA1致病性突变的女性相比则没有差异。携带两种突变以及雌激素受体阳性和孕激素受体阳性乳腺癌的女性百分比介于携带 BRCA1 致病性突变的杂合子对照和携带 BRCA2 致病性突变的杂合子对照之间。作者的结论是,同时遗传了BRCA1和BRCA2致病性突变的女性可能与仅遗传了BRCA1突变的携带者进行了相似的管理。
多项研究表明,BRCA致病性突变可能与遗传预测有关。一项研究评估了176个具有BRCA1或BRCA2致病性突变的家族和至少连续两代相同的癌症。先证者被诊断出患有乳腺癌的时间,估计比父母那一代早6.8年,比祖父母那一代早9.8年。
类似的,另一项研究显示,80对母女配对的致病性突变携带者在乳腺癌诊断时的年龄存在差异,但前提是母亲在50岁后被诊断为乳腺癌。
另一项队列研究对106名连续两代携带已知BRCA致病性突变的女性进行了研究,估计在随后几代中发病年龄将提前6至8岁。
RRSO和/或口服避孕药的使用与乳腺癌的风险相关。
(更多信息,请参阅本总结中RRSO部分和口服避孕药部分。) 其他潜在可改变的生殖和激素因素也会影响风险。
乳腺癌和卵巢癌外显率基因修饰物的研究越来越多,但目前还没有临床应用价值。
(更多信息,请参阅BRCA1和BRCA2致病性突变携带者风险的修饰物部分。) 虽然乳腺癌和卵巢癌的平均外显率可能不像最初估计的那么高,但无论从相对值还是绝对值来看,都很重要,尤其是在1940年以后出生的女性中。50岁之前的风险更高,这在最近出生的队列中一直存在的,
还需要更多的研究,以进一步确定潜在的修正因素,从而得出更精确的个人风险预测。对于不太常见的癌症,如胰腺癌,缺乏精确的外显率的估计。
几项大型研究已证实,携带BRCA1和BRCA2致病性突变的携带者患CBC的风险增加,结果相当一致,总结如表6所示。
研究 | BRCA1携带者(%) | BRCA2携带者(%) |
---|---|---|
Graeser et al.(2009) | 18.5 | 13.2 |
Malone et al.(2010) | 20.5 | 15.9 |
van der Kolk et al.(2010) | 34.2 | 29.2 |
Metcalfe et al.(2011) | 23.8 | 18.7 |
Molina-Montes et al.(2014) | 27 | 19 |
Basu et al.(2015) | 25.7 | 19.5 |
van den Broek et al.(2016) | 21.1 | 10.8 |
发表的结果包括德国遗传性乳腺癌和卵巢癌联盟的一项大型研究,该研究评估了已知BRCA1和BRCA2致病性突变家族成员患CBC的风险。在第一例乳腺癌25年后,BRCA1和BRCA2家族的CBC风险都接近50%。在这项研究中,患乳腺癌的风险与年龄成反比,40岁之前首次患乳腺癌的女性患乳腺癌的风险最高。
随后,来自女性环境癌症和辐射流行病学(WECARE)研究(一项大型、基于人群的 CBC 巢式病例对照研究)的结果报告,BRCA1/BRCA2致病性突变携带者患CBC的10年风险为15.9%,而非携带者的风险为4.9%。在这项研究中,风险也与首次诊断的年龄成反比,且比有患有乳腺癌的一级亲属(FDR)的风险高1.8倍。
荷兰的一项对BRCA1/BRCA2家族成员的大型研究报告了来自BRCA1和BRCA2家族的女性患CBC的类似的10年风险(分别为34.2%和29.2%)。
对655名携带BRCA1/BRCA2致病性突变的女性进行保乳或乳房切除术后的比较发现,两组治疗组的CBC发病率都很高,20年随访的发病率超过50%。BRCA1致病性突变的女性的发病率明显高于BRCA2致病性突变的女性,并且在35岁或35岁之前首次发生乳腺癌的女性的发病率也明显高于BRCA2致病性突变的女性。
在一项对810名I期或II期乳腺癌患者的研究中,有149人(18.4%)发展为CBC;15年的精算风险在BRCA1致病性突变携带者中为36.1%,在BRCA2致病性突变携带者中为28.5%。
50岁以前诊断的女性患病风险高于50岁或50岁以后诊断的女性(37.6% vs.16.8%;P=0.003)。此外,在50岁之前被最初诊断为乳腺癌的女性中,患CBC的风险因家族史的不同而不同。有在50岁之前被诊断为乳腺癌的0、1、2或2个以上FDR的女性,患CBC的风险分别为33.4%、39.1%和49.7%。
BRCA1和BRCA2携带者首次乳腺癌后患CBC的风险已在回顾性和前瞻性观察性流行病学研究中得到检验。对这些流行病学研究(18项回顾性和2项前瞻性队列研究)的系统回顾和定量荟萃分析报告了BRCA1携带者患CBC的5年累积风险为15%(95% CI,9.50%-20%),而BRCA2携带者患CBC的5年累积风险为9%(95% CI,5%-14%)。
当单独分析前瞻性研究时,BRCA1携带者的5年累积风险增加到23.4%(95% CI, 9.1%-39.5%),而BRCA2携带者的5年累积风险增加到17.5% (95% CI,9.1%-39.5%)。报告频率的差异可能是固有的,这是由于在回顾系列研究中引入了潜在的偏倚。
同样,一个荷兰的队列研究纳入了6294例(包括200例BRCA1基因携带者和71例BRCA2携带者)在50岁前被诊断患有浸润性乳腺癌的患者,且在平均随访12.5年中,BRCA1携带者患CBC的10年的风险为21.1%(95%CI,15.4%-27.4%),而BRCA2携带者的为10.8%(95%CI,4.7%-19.6%)和非携带者的为5.1%(95%CI,4.5%-5.7%)。
首次乳腺癌诊断的年龄是BRCA1/BRCA2携带者患CBC的10年累积风险的预测指标。具体来说,BRCA1/BRCA2携带者在41岁之前诊断的CBC风险为23.9%(BRCA1,25.5%;BRCA2,17.2%);相比之下,在41至49岁的患者中,被诊断为CBC的风险占12.6%(BRCA1,15.6%;BRCA2,7.2%)。
在一项英国研究中,纳入了506名BRCA1携带者和505名BRCA2携带者,他们均是在任何年龄被诊断为乳腺癌的患者,中位随访时间为7.8年。在这项研究中,BRCA1携带者和BRCA2携带者患CBC的10年风险分别为25.7%和19.5%。
BRCA1和BRCA2携带者较早地被诊断出乳腺癌与较高的CBC风险显著相关,40岁以下的乳腺癌患者20年的患病率为55.4%,而50岁以上的乳腺癌患者20年的患病率为36.4%。此外,与BRCA2携带者相比,BRCA1携带者之间的差异更为明显。
一项国际、多中心、前瞻性队列研究随访了1305名BRCA1和908名BRCA2女性携带者,她们被诊断为乳腺癌(没有任何其他癌症),中位随访时间为4年(范围为2-7年)。
在随访开始时,参与者的中位年龄为47岁(40-55岁)。作者报告了BRCA1携带者在首次乳腺癌诊断20年后的CBC累积风险为40%(95% CI,35%-45%),而BRCA2携带者为26%(95% CI, 20%-33%)。这些20年估计数与表6所报告的10年累积风险估计数一致。
因此,综上所述,尽管在研究设计、研究中心和样本量上存在差异,但在携带BRCA1/BRCA2致病性突变的女性中,关于CBC的数据显示了一些一致的发现:
有关在BRCA致病性突变携带者中使用降低风险手术的信息,请参阅本总结中的降低风险乳腺切除术部分。有关在携带BRCA致病性突变的CBC携带者中使用他莫昔芬作为降低风险策略的信息,请参阅本总结的化疗部分。
最近,两项基于基因登记的研究探讨了BRCA相关的卵巢癌后出现原发性乳腺癌的风险。在一项研究中,164名携带BRCA1/BRCA2的原发性上皮性卵巢癌、输卵管癌或原发性腹膜癌患者被随访观察后续事件。
在诊断为卵巢癌5年后发生乳腺癌的风险低于先前报道的未患病的BRCA1/BRCA2携带者。在这个研究系列中,总生存期主要以卵巢癌相关死亡为主。一项类似的研究比较了与BRCA相关的卵巢癌患者和未患病的携带者的原发性乳腺癌的风险。
卵巢癌患者的2年、5年和10年原发乳腺癌的风险均有统计学意义上的降低。单侧乳腺癌患者在卵巢癌诊断前患CBC的风险也低于非卵巢癌患者,尽管差异无统计学意义。这些研究表明,卵巢癌的治疗,即卵巢切除和以铂为基础的化疗,可能有助于预防随后的乳腺癌。在一项对364名接受BRCA致病性突变检测的上皮性卵巢癌患者的单中心队列研究中,发现135名(37.1%)患者携带一种生殖系BRCA1或BRCA2致病性突变。在这135名BRCA1/BRCA2携带者中,有12人(8.9%)患有乳腺癌。所有乳腺癌分期为0到II期,且诊断如下:乳腺X线检查(7)、可触及的肿块(3)、和在降低风险的乳房切除术中偶然发现的(2)。在平均6.3年的随访中,卵巢癌后患乳腺癌的12人中,3个死于复发性卵巢癌和1个死于转移性乳腺癌。
这些癌症中的大多数是通过乳腺X线检查或临床检查发现的,这表明用其他方式进行额外的乳腺监测或降低风险的手术可能具有可疑的价值。数学模型表明,对于BRCA相关性的卵巢癌患者,乳腺癌筛查应包括乳腺X线摄影和临床乳腺检查。考虑乳腺核磁共振成像(MRI)和/或降低风险的乳房切除术可能对早期卵巢癌女性或长期卵巢癌幸存者有益。
女性乳腺癌和卵巢癌是与BRCA1和BRCA2相关的主要癌症。BRCA致病性突变也增加了患输卵管癌和原发性腹膜癌的风险。一项对BRCA1致病性突变携带者的家族登记的大型研究发现,与普通人群相比,携带BRCA1致病性突变的携带者患输卵管癌的风险是普通人的120倍。
带有完整卵巢的BRCA致病性突变的携带者患原发性腹膜癌的风险有所增加,但仍难以量化,尽管RRSO后20年内的残留风险为3%至4%。
(更多信息,请参阅本总结中卵巢癌部分的RRSO部分。)
胰腺癌、男性乳腺癌和前列腺癌也一直与BRCA致病性突变相关,尤其是与BRCA2相关。在一些研究中还发现了其他癌症。这些癌症与BRCA致病性突变之间的相关性强度更难估计,这是因为在致病性突变携带者中观察到的这些癌症数量较少。
携带BRCA2致病性突变和较小程度上携带BRCA1致病性突变的男性患乳腺癌的风险更高,估计的终生患病风险分别为5%至10%和1%至2%。
携带BRCA2致病性突变和较小程度上携带BRCA1致病性突变的男性患前列腺癌的风险大约增加了3至7倍。
与BRCA2相关的前列腺癌也更具侵袭性。
(更多信息,请参阅PDQ总结中的前列腺癌遗传学部分。)
家族性胰腺癌的研究以及一系列未选择的胰腺癌也支持与BRCA2的相关性,和在较小程度上携带BRCA1的相关性。
总的来说,有3%到15%的有FPC家族可能有生殖系BRCA2致病性突变,随着患病亲属的增多,患病风险也在增加。
类似地,对未选择的胰腺癌的研究报告了BRCA2致病性突变频率在3%到7%之间,而这些数字在AJ后裔中接近10%。
据估计,BRCA2携带者患胰腺癌的终生风险为3%至5%,
而在一般人群中,到70岁估计的终生风险为0.5%。
一项针对1000多名致病性突变携带者的大型单中心的研究发现,与普通人群中的发病率相比,BRCA2携带者患胰腺癌的风险增加了21倍,BRCA1致病性突变携带者患胰腺癌的风险增加了4.7倍。
在一些但并非所有的研究中,其他的与BRCA2致病性突变相关的癌症包括黑色素瘤、胆管癌和头颈癌,但这些风险似乎不大(小于5%的终生风险),而且没有被很好地研究。
癌症部位 | BRCA1 | BRCA2 | ||
---|---|---|---|---|
证据的强度 | 绝对风险程度 | 证据的强度 | 绝对风险程度 | |
乳腺(女) | +++ | 高 | +++ | 高 |
卵巢、卵管、腹膜 | +++ | 高 | +++ | 中等的 |
乳腺(男性) | + | 不明确的 | +++ | 低 |
胰腺 | ++ | 非常低 | +++ | 低 |
前列腺 | + | 不明确的 | +++ | 高 |
a 更多关于BRCA1和BRCA2与前列腺癌关联的信息,请参阅PDQ总结中前列腺癌遗传学部分内容。 | ||||
+++多项研究显示了相关性,并且是相对一致的。 | ||||
++多项研究和优势的证据是积极的。 | ||||
+可能相关,主要是单项研究;较小的有限研究和/或不一致,但倾向于阳性。 |
第一项调查癌症风险的乳腺癌联盟研究,报告了BRCA1携带者中结直肠癌的风险过量(RR, 4.1;95% CI,2.4-7.2)。
这一发现得到了一些但不是所有的基于家族的研究的支持。然而,仅在AJ人群中进行的未经选择的结直肠癌的系列研究显示,没有发现BRCA1或BRCA2致病性突变率的升高。
综上所述,这些数据表明结直肠癌的风险几乎没有增加,而且可能只在特定人群中增加。因此,此时,BRCA1致病性突变的携带者应遵从对结直肠癌人群进行筛查的建议。
在200名患有子宫内膜癌的犹太女性和56名患有子宫乳头状浆液性癌的未被选择的女性中,没有发现遗传性BRCA致病性突变的增加。
(更多信息,请参阅本总结中卵巢癌部分的降低风险输卵管卵巢切除术部分。)
对于那些在家族中分离的BRCA1/BRCA2致病性突变检测阴性的女性,剩余家族风险,存在相互矛盾的证据。一项初步的、基于对353名在家族中分离的BRCA1致病性突变检测呈阴性的女性进行了前瞻性评估的研究发现,在超过6000个人-年的观察期间发生了5例乳腺癌,终生患病风险为6.8%,这个概率与一般人群相似。
一份报告中,家族中BRCA1或BRCA2致病性突变检测呈阴性的女性这种风险可能高达5倍,
随后,他给编辑写了许多信,建议确定偏差是造成这种观察到的过度风险的主要原因。
另有四项分析表明,这一比例大约为过度风险的1.5至2倍。
在一项研究中,报告了两例卵巢癌。
有几项研究涉及了回顾性分析;所有的研究都是基于小的观察病例数,具有不确定的统计意义和临床显著性。
许多其他前瞻性研究的结果都没有发现风险增加。一项针对375名已知家族致病性BRCA1或BRCA2基因突变呈阴性的女性的研究,报道了两种浸润性乳腺癌、两种原位乳腺癌,且未诊断出卵巢癌,平均随访时间为4.9年。预期会有4例浸润性乳腺癌,而只观察到2例。
另一项规模相似但随访时间较长的研究(395名女性和7008人年的随访)也发现,在突变阴性的女性中,乳腺癌风险总体上没有统计学意义的增加(观察到的/预期的[O/E],0.82;95% CI,0.39-1.51),尽管至少有一个患乳腺癌的FDR的女性患乳腺癌的风险增加不显著(O/E, 1.33;95% CI,0.41-2.91)。
一项对来自乳腺癌家族登记处的160个BRCA1和132个BRCA2致病性突变阳性家族的研究发现,没有证据表明这些家族中的非携带者的风险增加。
在一项对722名来自澳大利亚的突变阴性女性的大型研究中,6例浸润性乳腺癌在中位随访6.3年后被观察到,标准化发病率(SIR)并没有显著升高(SIR,1.14;95% CI,0.51-2.53)。
基于可用的数据,似乎已知的家族BRCA1和BRCA2致病性突变检测阴性的女性可以遵从一般人群筛查的指南,除非他们有足够的额外的风险因素,如乳腺非典型增生的个人史或亲属不携带家族致病性突变的乳腺癌家族史。
大多数具有特异性位点的乳腺癌的家族BRCA1/BRCA2检测呈阴性,并没有与Cowden综合症或Li-Fraumeni综合症相一致的特征。
五项使用以人群为基础和临床为基础的方法的研究表明,这些家族的卵巢癌风险没有增加。虽然卵巢癌的风险没有增加,但患乳腺癌的风险仍然很高。
BRCA1和BRCA2的致病性突变导致了乳腺癌和卵巢癌的高风险。然而,该风险在所有的致病性突变携带者中并不相同,而且会因多种因素而不同,包括癌症类型、发病年龄和突变部位。
外显率的这种观察到的突变导致了一种假说,即其他遗传和/或环境因素改变了致病性突变携带者的癌症风险。有越来越多的文献确定了遗传和非遗传因素,这些因素有助于在BRCA1/BRCA2致病性突变家族中观察癌症发病率的变化。
迄今为止,对乳腺癌和卵巢癌风险的基因修饰物进行调查的最大研究来自CIMBA,这是一项大型国际研究,对超过15,000名BRCA1和10,000名BRCA2携带者进行了基因型和表型的数据分析。
利用候选基因分析和GWAS,CIMBA确定了几个与乳腺癌和卵巢癌风险增加和降低相关的位点。一些SNP与乳腺癌亚型相关,例如激素受体和HER2/neu的状态。引起的风险都是适度的,但如果以倍增的方式操作,可能会显著影响BRCA1/BRCA2致病性突变携带者的癌症风险。目前,这些SNP还没有被用于临床决策。
在BRCA1和BRCA2致病性突变家族中都发现了一些基因型与表型的相关性。没有一项研究有足够数量的致病性突变阳性个体来得出明确的结论,而且这些发现可能不足以用于个人风险评估和管理。在25个具有BRCA2致病性突变的家族中,在第11号外显子中发现了一个与核苷酸3035和6629相邻的卵巢癌基因簇区。
一项由乳腺癌联盟收集了164个BRCA2致病性突变家族的研究证实了最初的发现。与在该区域任何一侧有突变的家族相比,卵巢癌基因簇区内的致病性突变与卵巢癌风险的增加和乳腺癌风险的降低有关。
此外,乳腺癌联盟收集了356个具有蛋白截断的BRCA1致病性突变的家族的研究报告,与周围区域相比,中心区域(核苷酸2401-4190)突变的乳腺癌风险更低。3'至4191核苷酸的突变显著降低了卵巢癌的风险。
这些观察结果在随后的研究中得到了普遍的证实。
在德系犹太人中的研究中,可以研究大量的存在相同致病性突变的家族,也发现与基因的3'末端存在BRCA1:5382突变的携带者相比,BRCA1:185delAG突变的携带者的卵巢癌风险更高。
然而,与携带BRCA1:185delAG和BRCA2:6174delT突变的人相比,携带BRCA1:5382insC致病性突变的人患乳腺癌的风险,特别是双侧乳腺癌的风险,以及在同一个体中同时发生乳腺癌和卵巢癌的风险更高。携带BRCA1致病性突变的人患卵巢癌的风险相当高,而携带BRCA2:6174delT致病性突变的人在45岁之前患卵巢癌的风险却不高。
在澳大利亚的一项对122个BRCA1致病性突变家族的研究中,大型基因组重排突变与乳腺癌和卵巢癌的高风险特征相关,包括乳腺癌诊断的年龄更小和双侧乳腺癌的发病率更高。
几项评估了见于BRCA1相关乳腺癌的病理模式的研究表明,其与不良的病理和生物学特征有关。这些发现包括比预期更高的骨髓组织学频率、高组织学分级、坏死区域、小梁生长模式、非整倍性、高S期分数、高有丝分裂指数和频繁的TP53突变。
在一个包含3797名BRCA1致病性突变携带者的大型国际系列研究中,乳腺癌诊断的中位年龄为40岁。
在BRCA1携带者的乳腺肿瘤中,78%为ER阴性;79%是PR阴性;90%是HER2阴性;69%为三阴性。这些发现与多个较小的系列研究结果一致。
此外,ER阴性肿瘤的比例随着乳腺癌诊断年龄的增加而明显下降。
三阴性和基底样亚型癌症之间存在大量但不完全的重叠,这两种亚型在BRCA1相关乳腺癌中都很常见,
尤其是在50岁之前确诊的女性。
一小部分与BRCA1相关的乳腺癌的ER为阳性,这与发病年龄较晚有关。
这些ER阳性的癌症具有介于ER阴性BRCA1癌症和ER阳性的散发乳腺癌之间的临床行为特征,这增加了它们发生独特机制的可能性。
在三阴乳腺癌患者中,生殖系BRCA1致病性突变的患病率显著,在接受临床遗传检测的女性中(因此在很大程度上由于家族史而被选择)和未被选择的三阴性患者中,致病性突变的患病率为9-35%。
值得注意的是,研究表明,在未被选择的三阴乳腺癌患者中,尤其是在50岁之前被诊断的患者中,BRCA1致病性突变的患病率很高。通过12项研究,对1824名三阴性乳腺癌患者进行的大型报告发现,其中有14.6%的患者具有遗传性癌症易感基因的致病性突变。
BRCA1致病性突变占最大比例(8.5%),其次为BRCA2(2.7%),PALB2 (1.2%);以及BARD1、RAD51D、RAD51C和BRIP1(每个基因0.3-0.5%)。在这项研究中,那些在BRCA1/BRCA2或其他遗传性癌症基因中具有致病性突变的患者在更早的年龄被诊断,并且比那些没有致病性突变的患者有更高的肿瘤分级。具体而言,在BRCA1致病性突变携带者中,诊断的平均年龄为44岁,94%为高级别肿瘤。一项研究调查了308名三阴乳腺癌患者;BRCA1致病性突变45例。致病性突变均可见于由于家族史而未被选择的女性(11/58;19%)和有家族病史的女性中(26/111;23%)。
开展了一项基于12项研究的2533名患者的荟萃分析,以评估三阴性乳腺癌高危女性中BRCA1致病性突变的风险。
结果显示,在三阴乳腺癌患者中,BRCA1致病性突变的RR值为5.65(95% CI,4.15-7.69),约有2/9的三阴性乳腺癌患者携带BRCA1致病性突变。有趣的是,有关77例未被选择的三阴性乳腺癌患者的研究中,有15例(19.5%)存在一个生殖系致病性突变或体细胞BRCA1和BRCA2突变,证明复发的风险在那些与BRCA1致病性突变相关的三阴乳腺癌中比在那些非BRCA1相关的三阴乳腺癌中更低;但这项研究因纳入病例少具有局限性。
第二项研究检测了BRCA1相关与非BRCA1相关的三阴乳腺癌的临床结果,结果显示二者无差异,尽管BRCA1相关的乳腺癌患者存在更多脑转移的趋势。在这两项研究中,除了一种BRCA1致病性突变携带者外,所有的BRCA1致病性突变携带者都接受了化疗。
相反,在没有家族史或二次原发性癌的情况下,HER2阳性和年轻并不会增加BRCA1、BRCA2或TP53致病性突变的可能性。
据推测,许多BRCA1肿瘤起源于正常乳腺的基底上皮细胞层,占未选择的浸润性导管癌的3%至15%。如果乳腺基底上皮细胞代表乳腺干细胞,那么野生型BRCA1的调控作用可能部分解释了BRCA1相关乳腺癌在BRCA1功能受损时的侵袭表型。
需要进一步的研究来充分认识这种亚型的乳腺癌在遗传综合症中的重要性。
识别基底样乳腺癌最准确的方法是通过基因表达研究,该方法已被用于将乳腺癌分为生物学上和临床意义上的两类。
这项技术也被证明能在较高比例的病例中正确区分与BRCA1和BRCA2相关的肿瘤与散发肿瘤。
值得注意的是,在通过基因表达阵列研究确定分子表型的一组乳腺肿瘤中,所有BRCA1改变的肿瘤均属于基底肿瘤亚型;
然而,这项技术由于成本高而没有得到常规应用。相反,基底上皮的免疫组织化学标记已被提出用于识别基底样乳腺癌,其中ER、PR和HER2通常为阴性,而细胞角蛋白5/6或表皮生长因子受体染色呈阳性。
基于这些测量蛋白表达的方法,许多研究表明,大多数与BRCA1相关的乳腺癌基底上皮标志物呈阳性。
越来越多的证据表明,浸润前病变是BRCA表型的一个组成部分。乳腺癌联盟最初报告了在BRCA1相关乳腺癌中相对缺乏原位成分,
在随后对BRCA1/BRCA2携带者的两项研究中也发现了这一现象。
然而,在一项对369例导管原位癌(DCIS)病例的研究中,分别检测到BRCA1和BRCA2致病性突变的比例为0.8%和2.4%,仅略低于先前报道的浸润性乳腺癌患者的患病率。
一项对高危临床乳腺癌病例的回顾性研究发现,在73例BRCA相关乳腺癌和146例致病性突变阴性病例中,浸润前病变(尤其是DCIS)的发生率相似。
一项对AJ女性的研究,按她们是否被转到高风险诊所或未被选择进行分层,显示了转诊患者中DCIS和浸润性乳腺癌的患病率与未被选择的受试者中小于三分之一的DCIS病例相似。
同样,关于增生性病灶患病率的数据也不一致,报告的有增加的和减少的患病率。
与浸润性乳腺癌相似,早期诊断的DCIS和/或有乳腺癌和/或卵巢癌家族史的DCIS更可能与BRCA1/BRCA2致病性突变相关。
整体证据表明,DCIS是BRCA1/BRCA2谱的一部分,尤其是BRCA2;然而,因家族史而未被选择的DCIS患者中致病性突变的患病率低于5%。
与BRCA2相关的肿瘤表型比BRCA1的更具异质性,也没有那么明显的特征,尽管它们通常为ER和PR阳性。
一个纳入2392名BRCA2致病性突变携带者的大型国际系列研究发现,只有23%的BRCA2致病性突变携带者的肿瘤呈ER阴性;36%是PR阴性;87%是HER2阴性;16%是三阴性。
一项从12项研究招募的、包含1824名三阴乳腺癌患者的大型报告发现,有2.7%的患者具有BRCA2致病性突变。
(有关本研究的更多信息,请参阅本总结的BRCA1病理学部分。) 来自冰岛的一份报告发现,与散发病例对照相比,BRCA2相关肿瘤的小管形成更少,核多形性更强,有丝分裂率更高;然而,一个单一的BRCA2奠基人致病性突变(999del5)几乎解释了该人群所有的遗传性乳腺癌,因此限制了这一观察的普遍性。
来自北美和欧洲的一个大的病例系列描述了更大比例的BRCA2相关的肿瘤具有持续的边缘推进(一种侵袭模式的组织病理学描述)、更少的小管和更低的有丝分裂计数。
其他报告表明,BRCA2相关肿瘤包括小叶和小管的组织学过多。
总之,与BRCA2致病性突变相关的组织学特征一直不一致。
考虑到BRCA1或BRCA2的生殖系致病性突变导致发生乳腺癌的可能性非常高,很自然地,这些基因也会参与更常见的非遗传性疾病的发展。虽然BRCA1和BRCA2的体细胞突变在散发性乳腺癌中并不常见,但越来越多的证据表明,基因启动子(BRCA1)的高甲基化和杂合性(LOH)缺失是频繁发生的事件。事实上,许多乳腺癌的BRCA1 mRNA水平较低,这可能是由于基因启动子的高甲基化所致。
大约10%到15%的散发性乳腺癌有BRCA1启动子高甲基化,更多的通过其他机制下调BRCA1。基底型乳腺癌(ER阴性、PR阴性、HER2阴性、细胞角蛋白5/6阳性)比其他类型的肿瘤更常见BRCA1失调。
与BRCA1相关的肿瘤特征也与BRCA1启动子的结构甲基化有关。在一项纳入255名无生殖系BRCA1致病性突变而在40岁前被诊断为乳腺癌的女性的研究中,在31%的有多种BRCA1-相关病理特征的肿瘤的女性的外周血中,观察到了BRCA1基因的甲基化(例如,高细胞有丝分裂指数和增长模式,包括多核细胞),相比之下对照组仅观察到不到4%的甲基化。
(更多信息,请参阅BRCA1病理学部分。) 虽然还没有关于BRCA2致病性突变的高甲基化的报道,但是染色体13q上的BRCA2位点是乳腺癌常见的LOH靶点。
针对BRCA1或BRCA2蛋白表达缺失的肿瘤,靶向治疗正在开发中。
具有BRCA1和BRCA2致病性突变的女性卵巢癌更可能是高级别浆液性腺癌,而不太可能是黏液性或交界性肿瘤。
输卵管癌和腹膜癌也是BRCA相关疾病的一部分。
从具有卵巢癌遗传易感性的女性身上摘除的输卵管组织病理学检查显示异常增生病变,提示癌前表型。
据报道,在实施降低风险的手术时,从BRCA致病性突变携带者身上移除的附件中,有2%到11%的癌是隐匿性的。
这些隐匿性病变大部分发生在输卵管,这导致了许多与BRCA相关的卵巢癌可能实际上起源于输卵管的假设。具体来说,输卵管的远端部分(含绒毛)已经被认为是BRCA致病性突变携带者中高级别浆液性癌的一个共同起源,这是基于伞端非常接近卵巢表面、伞端暴露于腹膜腔和伞端较宽的表面积。
由于高级别浆液性癌的多中心起源来自于Müllerian来源的组织,卵巢癌、输卵管癌和腹膜癌的分期现在被国际妇产联合会统一。术语高浆液性卵巢癌可以用来代表高浆液性盆腔癌,以保持语言的一致性。
高级别浆液性卵巢癌具有较高的体细胞TP53突变发生率。
DNA微阵列技术提示,在BRCA1、BRCA2和散发性卵巢癌之间存在不同的致癌分子通路。
此外,数据显示,与BRCA相关的卵巢癌更常转移至内脏,而散发的卵巢癌仍局限于腹膜。
与高级别浆液性癌不同,低级别浆液性卵巢癌不太可能属于BRCA1/BRCA2谱的一部分。
考虑到BRCA1或BRCA2的生殖系突变导致发生卵巢癌的可能性非常高,自然地,这些基因也会参与更常见的非遗传性疾病的发展。虽然BRCA1和BRCA2的体细胞突变在散发的卵巢癌肿瘤中并不常见,
越来越多的证据表明,基因启动子(BRCA1)和LOH (BRCA2)的高甲基化是频繁发生的事件。BRCA1或BRCA2蛋白表达的缺失在卵巢癌中比在乳腺癌中更常见,
并且BRCA1的下调与顺铂敏感性增强和生存期改善有关。
针对BRCA1或BRCA2蛋白表达缺失的肿瘤,靶向治疗正在开发中。
Lynch综合症以常染色体显性遗传为特征,易患右侧结肠癌、子宫内膜癌、卵巢癌和其他结肠外癌症(包括肾盂癌、输尿管癌、小肠癌和胰腺癌)、多种原发性癌症和癌症发病年龄较小。
这种情况是由错配修复(MMR)基因中的生殖系突变引起的,该基因参与DNA错配突变的修复。
MLH1和MSH2基因是Lynch综合症最常见的易感基因,占观察到致病性突变的80%至90%,
其次是MSH6和PMS2。
(有关该综合症的更多信息,请参阅PDQ总结中结直肠癌遗传学中的Lynch综合症部分。)
继结直肠癌之后,子宫内膜癌是Lynch综合症家族的第二典型癌症。即使在Dr. Aldred Scott Warthin所描述的最初的G家族中,也发现许多家族成员患有包括子宫内膜癌在内的结肠外癌。虽然第一版的Amsterdam标准不包括子宫内膜癌,
在1999年,Amsterdam标准进行了修订,将子宫内膜癌纳入与Lynch综合症相关的结肠外肿瘤,以确定有风险的家族。
此外,1997年的Bethesda指南(2004年修订)将子宫内膜癌和卵巢癌列为Lynch综合症相关的癌症,以促进对Lynch综合症的肿瘤检测。
据估计,患有Lynch综合症的女性一生中罹患卵巢癌的风险高达12%,根据已知或怀疑患有Lynch综合症的高风险诊所确定的家族,卵巢癌的RR值在3.6到13之间。
根据Lynch综合症相关的致病性突变,卵巢癌风险可能存在差异。在与PMS2相关的Lynch综合症中,一项对284个家族的研究未能发现卵巢癌风险的增加。
另一项对3119名Lynch综合症致病性突变携带者的前瞻性登记表明,在MLH1、MSH2和MSH6携带者中,卵巢癌的累积风险从10%到17%不等。相比之下,在303年的随访中,67名PMS2致病性突变的女性中有0人患卵巢癌。
总的来说,PMS2致病性突变携带者的病例太少,不足以对卵巢癌的治疗提出明确的建议。Lynch综合症相关的卵巢癌的特征可能包括国际妇产科联合会I期和II期诊断的比例过高(报道的为81.5%)、严重亚型的代表不足(报道的为22.9%),和更好的10年生存率(报道的为80.6%),均是与以人群为基础的系列研究中和BRCA致病性突变携带者相比。
Lynch综合症的乳腺癌风险问题一直存在争议。回顾性研究的结果并不一致,但有几项研究表明,Lynch综合症患者的乳腺癌中有一定比例存在微卫星不稳定性:
其中一项研究对Lynch综合症患者的乳腺癌风险进行了评估,发现其风险并没有升高。
然而,迄今为止最大的前瞻性研究,包含了来自结肠癌家族登记处的446未患病的致病性突变携带者,
随访其10年后,报道的乳腺癌SIR值升高了3.95(95% CI,1.59-8.13;P=0.001)。
随后,该研究小组分析了764名MMR基因致病性突变携带者的数据,这些人之前曾被诊断为结直肠癌。结果显示,结直肠癌术后10年乳腺癌风险为2%(95% CI,1%-4%),SIR为1.76(95% CI, 1.07-2.59)。
来自英国的一个系列研究由临床参考的Lynch综合症组成,经过努力纠正以确定,在157 MLH1携带者中乳腺癌的风险增加了两倍,而在其他MMR突变携带者中则没有增加。
对15项分子肿瘤检测结果的研究中的Lynch综合症患者乳腺癌风险的一项荟萃分析结果显示,在MMR致病性突变携带者中,122例乳腺癌中有62例(51%;95% CI,42%-60%)存在MMR缺陷。此外,在总共21项研究中,对乳腺癌风险的估计显示,将MMR突变携带者与非携带者进行比较的8项研究中,乳腺癌风险增加了2倍至18倍,而13项研究没有观察到乳腺癌风险与Lynch综合症之间存在关联的统计学证据。
随后的一些研究表明,乳腺癌的风险比之前发表的更高,
虽然这并没有得到一致的观察。
通过对325个患有Lynch综合症的加拿大家族(主要包括MLH1和MSH2携带者)的研究,报道的MSH2携带者一生中罹患乳腺癌的累计风险为22%。
同样,在一项对423名Lynch综合症女性的研究中,乳腺癌的风险也有所上升,与MLH1和MSH2致病性突变相比,MSH6和PMS2致病性突变的风险要高得多。
事实上,60岁的乳腺癌风险中,PMS2为37.7%,MSH6为31.1%,MSH2为16.1%,MLH1为15.5%。这些发现与另一项研究的结果一致,该研究纳入了528例存在Lynch综合症相关的致病性突变(包括一种MSH2, MSH6 PMS2, EPCAM)的患者,其中PMS2和MSH6突变更频繁的只存在于乳腺癌患者中,这是与那些只有大肠癌的患者相比(P=2.3×10-5)。
通过对全美1万多名接受基因检测的癌症患者的研究提供了支持MSH6与乳腺癌关联的其他数据。
结果表明,MSH6与乳腺癌相关的优势比(OR)为2.59(95% CI,1.35-5.44)。综上所述,这些研究突出了Lynch综合症患者的风险特征是如何随着越来越多的人接受多基因面板检测而不断演变的,与以前的研究相比,更多的人具有PMS2和MSH6致病性突变。在缺乏明确的风险评估的情况下,Lynch综合症患者需要根据家族史进行乳腺癌筛查。
请参阅其他遗传性乳腺癌和/或妇科癌症的综合征临床管理中的Lynch综合症部分,以了解Lynch综合症的临床管理信息。
乳腺癌也是罕见的LFS的一个组成部分,在LFS中,染色体17p上的TP53基因的生殖系突变已经被证实。TP53位于染色体17p上,编码一种53kd的核磷酸化蛋白,该蛋白与DNA序列结合,在DNA损伤的环境中起着细胞生长和增殖的负调控作用。它也是程序性细胞死亡的活性成分。
TP53基因的失活或蛋白质产物的破坏会导致受损DNA的持续存在和恶性细胞的可能发展。
广泛使用的LFS临床诊断标准最初是由Chompret等人在2001年制定的(称为Chompret标准),
并在2009年根据新的数据进行了修正。
LFS的特点是绝经前乳腺癌合并儿童肉瘤、脑肿瘤、白血病和肾上腺皮质癌。
据认为,TP53的生殖系突变占乳腺癌病例的比例不到1%。
与TP53相关的乳腺癌除了ER阳性、PR阳性或两者同时阳性外,通常还有HER2/neu阳性。
也有证据表明,接受与TP53相关的肿瘤化疗或放疗的患者可能存在与治疗相关的二次恶性肿瘤的风险。
“Li-Fraumeni综合症”这个术语在1982年首次使用,
Li和Fraumen在1988年提出了以下标准,后来成为该综合症的经典定义:
随后在2001年,Chompret等人系统地开发了TP53基因检测的推荐临床标准,将狭义的LFS肿瘤谱定义为肉瘤、脑肿瘤、乳腺癌和肾上腺皮质癌。标准如下:
这些标准在2009年
根据新出现的数据而进行修订,
相关修订如下:
* 2009年Chompret标准将LFS肿瘤谱定义为包括以下癌症:软组织肉瘤、骨肉瘤、脑瘤、绝经前乳腺癌、肾上腺皮质癌、白血病和肺支气管肺泡癌。
2015年,Bougeard等人根据415名致病性突变携带者的数据修订了标准,将31岁前儿童间变性横纹肌肉瘤和乳腺癌的存在作为检测的指征,类似于脉络膜丛癌和肾上腺皮质癌的推荐指标。标准修订如下:
** 2015 Chompret标准将LFS的肿瘤谱定义为包括以下癌症:绝经前乳腺癌、软组织肉瘤、骨肉瘤、中枢神经系统(CNS)肿瘤和肾上腺皮质癌。
在提交给City of Hope实验室进行临床TP53检测的525个样本中,17%(n =91)的样本中发现了生殖系TP53致病性突变。
所有存在TP53致病性突变的家族中至少有一名家族成员患有肉瘤、乳腺癌、脑癌或肾上腺皮质癌(核心癌症)。此外,所有8名脉络丛肿瘤患者都有TP53致病性突变,21名儿童肾上腺皮质癌患者中有14人也有TP53致病性突变。在30至49岁的女性中,没有其他核心癌症家族史的乳腺癌患者没有发现TP53突变。
随后,一大批来自法国的临床患者接受了主要基于2009版Chompret标准的检测,其中包括来自214个家族的415个致病性突变携带者。
在本研究中,43%的携带者有多种恶性肿瘤,首次发病的平均年龄为24.9岁。儿童的肿瘤谱以骨肉瘤、肾上腺皮质癌、中枢神经系统肿瘤和软组织肉瘤为特征(共占23%-30%),而成人的肿瘤谱主要包括乳腺癌(79%的女性)和软组织肉瘤(27%的携带者)。在31岁以下的女性乳腺癌患者中,TP53致病性突变的检出率为6%,没有其他提示LFS的特征。对基因型-表型相关性的评估表明,与所有类型的功能缺失突变(28.5年)或基因组重排(35.8年)相比,具有显性阴性错义突变(21.3年)的患者存在一种临床严重性的阶梯,且其平均发病年龄明显更低。除肾上腺皮质癌外,患病的儿童多存在显性-阴性错义突变。在127名携带致病性乳腺癌突变的女性中,31%的人患上了CBC。有40个肿瘤的受体状态信息,其中55%为HER2阳性,37%为三阳性(即ER阳性、PR阳性和HER2阳性)。在致病性突变携带者中,多重恶性肿瘤的发生率特别高(43%),其中83%是异时性的。64名首次接受肿瘤放疗的患者有治疗记录;其中,19例(30%)在辐射范围内发生26次继发性肿瘤,潜伏期为2-26年(平均10.7年)。
类似的,在国家癌症研究所的LFS研究中,286名TP53致病性突变阳性的个体的结果表明,到70岁时,男性和女性的累积癌症发病率几乎为100%。
他们报告了性别、年龄和癌症类型的显著差异。具体来说,累积癌症发病率在女性31岁时达到50%,男性46岁时达到50%,尽管男性在儿童和成年后期的风险更高。按性别排列的前四种癌症的累积发病率请参见表8。在那些患有一种癌症的患者中,49%的人在平均10年后至少又患上了一种癌症。发生第一和第二种癌症的年龄特异性风险是相当的。
到70岁时累积的癌症风险 | ||
---|---|---|
癌症类型 | 女性(%) | 男性(%) |
乳腺癌 | 54 | - |
软组织肉瘤 | 15 | 22 |
脑癌 | 6 | 19 |
骨肉瘤 | 5 | 11 |
a 改编自Mai等人的文章。 | ||
b 其他恶性肿瘤,如肾上腺皮质癌、白血病和肺支气管肺泡癌,已被认为是LFS癌症谱的一部分。 |
随着多基因(面板)检测的使用越来越多,重要的是要认识到,在没有LFS家族史特征的个体中,TP53的致病性突变被意外地鉴定出来。
在不符合Chompret标准的个人或家族中发现TP53致病性突变的临床意义尚不确定。因此,在考虑个人和家族病史的同时,对意外发现有生殖系TP53致病性突变的个体进行癌症风险解释和确定最佳管理策略仍然很重要。
一项在美国国立卫生研究院临床中队列研究心进行的、每年评估了116名带有生殖系TP53致病性突变的个体的研究,使用了含钆和不含钆的多模式筛查。基线筛查确定了8名患者(6.9%)患有癌症,MRI假阳性率为34.5% (n=40)。
建议每年用乳腺MRI筛查乳腺癌;
对其他癌症的进一步筛查已经得到研究,而且正在进行中。
Cowden和Bannayan-Riley-Ruvalcaba综合症(BRRS)是被统称为PTEN错构瘤综合症的一系列症状的一部分。约85%的Cowden综合症患者和约60%的BRRS患者具有可识别的PTEN致病性突变。
此外,在临床表型非常不同的患者中发现了PTEN的致病性突变。
PTEN错构瘤综合症指的是任何存在PTEN致病性突变的患者,不论其临床表现如何。
PTEN是一种双特异性磷酸酶,可以去除酪氨酸、丝氨酸和苏氨酸中的磷酸基团。PTEN的致病性突变多种多样,包括无义突变、错义突变、移码突变和剪接位点突变。大约40%的突变在编码磷酸酶核心基序的外显子5中发现,并且已经观察到几种复发的致病性突变。
在PTEN的5'端或在磷酸酶核心内具有突变的个体,往往有更多的器官系统受累。
诊断Cowden综合症的标准已被公布,并在随后更新。
这些标准包括主要的、次要的和特殊的标准,其中包括某些黏膜与皮肤的临床表现和成人发生的小脑发育不良性神经节细胞瘤(Lhermitte-Duclos病)。在系统的文献回顾的基础上,一套更新的标准被提出且目前已在国家综合癌症网络(NCCN)的指南中被使用。
与以前的标准相反,作者的结论是,没有足够的证据证明任何特征可以被归类为病原学特征。随着基因检测的增加,特别是多基因面板的使用,Cowden综合症的临床标准将需要与不符合这些标准的生殖系PTEN致病性突变个体的表型相统一。在此之前,Cowden综合症和其他PTEN错构瘤综合症是否将被临床定义或基于基因检测结果仍不明确。美国大学医学遗传学和基因组学(ACMG)表明,推荐为个人史或一级亲属患有1)成人发生的Lhermitte-Duclos疾病或2)已建立的Cowden综合症诊断标准中任何三个主要或次要的诊断标准的个体提供遗传咨询。
详细的建议,包括Cowden综合症的诊断标准,可以在NCCN和ACMG指南中找到。
此外,一种使用临床标准来估计PTEN致病性突变概率的预测模型是可用的;一项成本效益分析表明,如果突变的概率大于10%,生殖系PTEN检测是有成本效益的。
在10年的时间里,国际Cowden协会(ICC)在美国、欧洲和亚洲前瞻性地招募了一系列符合ICC放宽的PTEN检测标准的成人和儿童患者。
大多数人不符合Cowden综合症或BRRS诊断的临床标准。在3399个被招募和检测的个体中,295个先证者(8.8%)和另外73个家族成员被发现含有生殖系PTEN致病性突变。除了乳腺癌、甲状腺癌和子宫内膜癌外,作者得出结论,在癌症风险的基础上,黑色素瘤、肾癌和结直肠癌是生殖系PTEN致病性突变引起的癌症谱的一部分。第二项研究对大约100名具有生殖系PTEN致病性突变的患者进行了研究,证实了这些发现,并提出到70岁时累积癌症风险为85%。
尽管PTEN致病性突变,据估计在20万人中只有1人发生,
只占遗传性乳腺癌的一小部分,但PTEN功能的表征将为信号通路和维持正常细胞生理提供有价值的见解。
据估计,患有Cowden综合症的女性一生中患乳腺癌的风险在25%到50%之间。
其他研究报告的风险高达85%;
然而,在这些研究中存在选择偏倚的问题。与其他形式的遗传性乳腺癌一样,发病年龄通常很小,而且可能是双侧的。
根据研究人群的不同,子宫内膜癌的终生风险估计在19%到28%之间,绝经前发病的风险增加。
由于PTEN致病性突变在人群中的患病率较低,导致Cowden综合症的子宫内膜癌比例较小。没有数据表明PTEN致病性突变与卵巢癌风险的增加有关。皮肤的临床表现包括多发性毛鞘瘤、口腔纤维瘤和乳头状瘤,以及肢端、手掌和足底角化病。通过病史或对皮肤特征的观察,怀疑其为Cowden综合症。中枢神经系统表现为:大头畸形、发育迟缓、小脑发育不良性神经节细胞瘤。
(有关PTEN错构瘤综合症(包括Cowden综合症)的更多信息,请参阅PDQ总结中的结直肠癌遗传学和皮肤癌遗传学的内容。)
E-钙黏素基因CDH1于1998年首次在三个毛利人家族中发现,该家族中有多例弥漫性胃癌(DGC),因此被命名为遗传性弥漫性胃癌(HDGC)。在HDGC家族中已经有多例小叶性乳腺癌的后续报道。
CDH1位于染色体16q22.1上,编码E-钙黏素蛋白,这是一种钙依赖的同嗜性粘附分子,在细胞黏附、细胞极性、细胞信号转导、细胞分化和组织形态的维持等方面起着关键作用。
E-钙黏素与多种连环蛋白结合以稳定胞浆细胞粘附复合体,并维持E-钙黏素与肌动蛋白丝的相互作用。
CDH1的丢失可能由于体细胞突变、LOH或高甲基化而发生,并可能导致人类癌症的去分化和浸润性。
胃切除标本的典型组织病理学表现包括原位印戒细胞和/或印戒细胞呈栅栏状扩散。在所有胃癌中,1%-3%是遗传性胃癌综合症。
HDGC是一种常染色体显性遗传综合症,伴有低分化的浸润性胃腺癌,表现为皮革胃。它是一种高度外显和高度致命的综合症,临床DGC的风险从40%到83%不等。
小叶性乳腺癌的特征是小而均匀的细胞呈“单行”侵犯,在HDGC中,这种风险也增加。尽管浸润性小叶型乳腺癌只占所有乳腺癌的10%至15%,但CDH1致病性突变携带者罹患小叶型乳腺癌的终生风险在30%至50%之间。
筛查CDH1的指南各不相同,但包括一个家族中发生多例DGC、早期DGC或有DGC的家族中发生小叶性乳腺癌。大约25%符合这些标准的家族被发现具有CDH1的致病性突变。
在一些小叶性乳腺癌家族中发现了CDH1致病性突变,但没有发现胃癌。
没有胃癌家族史的CDH1致病性突变个体的管理尚不清楚。
PJS是一种早期发病的常染色体显性遗传疾病,以嘴唇、口周和颊部的黑色素细胞性斑疹为特征,同时伴有多发的胃肠道息肉,包括错构瘤和腺瘤。
在绝大多数PJS家族中,染色体19p13.3上的STK11基因的生殖系致病性突变已被发现。
PJS最常见的癌症是胃肠道癌症。然而,其他器官患恶性肿瘤的风险增加。例如,据估计,乳腺癌的累积风险为32%至54%,
卵巢癌的累积风险为21%(主要是卵巢性索瘤)。
据估计,患胰腺癌的风险比一般人群高出100多倍。
一项系统综述研究发现,PJS患者一生中累积的癌症风险(所有部位加起来)高达93%。
表9显示了这些肿瘤的累积风险。
患有PJS的女性也容易发展为恶性宫颈腺瘤,这是一种罕见的、极具浸润性的宫颈腺癌。
此外,患有PJS的女性一般发展为良性卵巢性索瘤,含有环形小管,而患有PJS的男性更容易发展为睾丸支持细胞瘤;
虽然这两种类型的肿瘤都不是恶性的,但它们可引起与雌激素分泌增加相关的症状。
尽管根据已发表的文献,PJS患者的恶性肿瘤风险非常高,但应该考虑选择和转诊偏倚导致这些风险被高估的可能性。
部位 | 年龄(岁) | 累积风险(%) |
---|---|---|
任何癌症 | 60-70 | 37-93 |
GI癌症 | 60-70 | 38-66 |
妇科癌症 | 60-70 | 13-18 |
根据起源 | ||
胃 | 65 | 29 |
小肠 | 65 | 13 |
结直肠 | 65 | 39 |
胰腺 | 65-70 | 11-36 |
肺 | 65-70 | 44029 |
乳腺 | 60-70 | 32-54 |
子宫 | 65 | 9 |
卵巢 | 65 | 21 |
子宫颈 | 65 | 10 |
睾丸 | 65 | 9 |
GI=胃肠道。 | ||
a 经Macmillan Publishers Ltd许可转载:胃肠病学,版权2010。 | ||
b 除子宫颈和睾丸外,与一般人群相比,所有的累积风险均增加(P<0.05)。 | ||
c GI癌症包括结直肠癌、小肠癌、胃癌、食道癌和胰腺癌。 | ||
d Westerman等人:胃癌不包括胰腺癌。 | ||
e 不包括子宫颈腺瘤或睾丸支持细胞瘤。 |
根据起源
PJS是由位于染色体19p13上的STK11(也称为LKB1)抑癌基因的致病性突变引起的。
与家族性腺瘤样息肉病中出现的腺瘤不同,PJS的息肉是错构瘤。对PJS的错构型息肉和癌症的研究显示等位基因失衡(LOH)与二次突变假说一致,表明STK11是肿瘤抑制基因。
然而,在不使剩余野生型等位基因失活的情况下,杂合STK11剔除小鼠发生错构瘤,表明单倍体不足可能足以导致PJS初始肿瘤的形成。
随后,在STK11+/-小鼠中发生的癌症确实显示LOH;
事实上,表明致病STK11+/-突变复合杂合突变小鼠和致病TP53-/-突变复合纯合突变小鼠的错构瘤和癌症加速发展。
STK11基因的种系突变包括一系列无义、移码和错义突变以及剪接位点突变和大缺失。
大约85%的突变位于表达蛋白质的激酶结构域。没有发现明显的基因型-表型相关性。
高达30%的突变涉及STK11的一个或多个外显子的大缺失,凸显了对PJS疑似病例进行缺失分析的重要性。
STK11已被明确证实会导致PJS。虽然早期使用直接DNA测序的估计显示STK11的致病性突变检出率为50% ,但检测大缺失技术研究发现高达94%的个体符合PJS临床标准的致病性突变。
考虑到这些研究结果,其他主要基因不太可能导致PJS。
由于PJS的癌症高累积风险,在PDQ概要“结直肠癌遗传学”部分的黑斑息肉综合征(PJS)诊断和监测建议表总结了各种筛查建议。
PALB2(BRCA2的配偶子和定位子)与BRCA2蛋白相互作用,并在同源重组和双链DNA修复中起作用。与BRIP1和BRCA2类似,已发现PALB2中的双等位基因致病性突变也会引起范可尼贫血。
多项多群体的家族性和早发性乳腺癌小型研究已经筛选了PALB2致病性突变。
致病性突变患病率介于0.4%至3.9%之间。与BRIP1和CHEK2类似,遗传性乳腺癌家族中PALB2致病性突变的分离不完全。
在559例CBC患者和565例单侧乳腺癌对照者中,0.9%的CBC患者发现致病性(截短性)PALB2致病性突变(RR,5.3;95%CI,1.8-13.2),对照者中无发现。
根据154个携带PALB2功能丧失突变的家庭的数据,该基因可能是遗传性乳腺癌的重要原因,其风险与BRCA2重叠。
这项研究对来自154个携带PALB2致病性突变的家庭的362名家庭成员进行了分析,发现女性携带者到70岁的绝对乳腺癌风险范围为33%(95%CI,24%-44%,无乳腺癌家族史的携带者)至58%(95%CI,50%-66%,有两位及以上FDR患早发性乳腺癌的携带者)。此外,在HER2状态已知的63例乳腺癌病例中,30%患有三阴乳腺癌。芬兰的一项先前的研究报告了PALB2基础致病性突变(c.1592delT),该突变至70岁的乳腺癌风险为40%,
并与三阴乳腺癌的高发病率(54%)和较低的存活率相关。
目前已在多个早发性和家族性乳腺癌患者群体中观察到致病性突变。
通过12项研究招募的1824名因家族史未入选的三阴乳腺癌患者中,有1.2%的患者携带PALB2致病性突变。
(有关该研究的更多信息,请参阅本摘要的BRCA1病理部分。)
在后来波兰的一项对超过12,529名未入选的乳腺癌女性患者和4,702名对照者的研究中,116例患者(0.93%;95%CI,0.76%-1.09%)和10例对照者(0.21%;95%CI,0.08%-0.34%)检测到PALB2致病性突变,乳腺癌OR为4.39(95%CI,2.30-8.37)。
这些研究结果证实,携带PALB2致病性突变的女性到75岁患乳腺癌的风险大幅升高(24%-40%)。PALB2致病性突变携带者的CBC5年累积发病率为10%,BRCA1致病性突变携带者的CBC5年累积发病率为17%,而非BRCA1、BRCA2致病性突变携带为3%。此外,携带PALB2致病性突变的乳腺癌女性患者的10年存活率为48%(95%CI,36.5%-63.2%),而携带BRCA1致病性突变的乳腺癌女性患者的10年存活率为72.0%,在任一基因中未携带BRCA1、BRCA2致病性突变的乳腺癌女性患者的10年存活率为74.7%。在PALB2携带者中,与乳腺肿瘤小于2cm的患者(10年存活率为82.4%)相比,乳腺肿瘤大于等于2cm患者的结局更差(10年存活率为32.4%)。约有三分之一的PALB2致病性突变携带者患有三阴乳腺癌,乳腺癌诊断的平均年龄为53.3岁。
在PALB2致病突变阳性的乳腺癌家族中发现男性乳腺癌患者。
在对115名男性乳腺癌病例的研究中,18名男性患者携带BRCA2致病性突变,另外两名男性患者携带致病性或预测致病性PALB2突变(占研究中种系突变的约10%和总样本的1%-2%)。
在对154个家庭的研究中,与一般人群中相比,PALB2致病性突变男性携带者的乳腺癌RR估计为8.30(95%CI,0.77-88.56;P=.08)。
在确定胰腺肿瘤中存在PALB2致病性突变并在96名家族性胰腺患者中检测到3%的患者携带种系致病性突变后,
许多研究已指出PALB2在胰腺癌中的作用。在81个家族性胰腺癌中,3.7%的患者检测到PALB2致病突变,
94例携带BRCA1/BRCA2致病性突变的阴性乳腺癌患者中有2.1%的患者有胰腺癌的个人或家族史。
在两项相对较小的研究中(一项研究的对象为77例BRCA1/BRCA2致病性突变阴性、有胰腺癌个人或家族史的先证者,其中一半是AJ血统;另一项研究的对象为29名有乳腺癌或卵巢癌个人或家族史的意大利胰腺癌患者),未能检测到任何PALB2致病性突变。
在33名BRCA1/BRCA2阴性,PALB2致病突变阳性的乳腺癌先证者的亲属中观察到胰腺癌增加6倍。
总体而言,观察到的家族性乳腺癌中PALB2致病性突变的患病率相对取决于胰腺癌和卵巢癌个人和家族史,但在所有研究中,观察到的致病突变率低于4%。数据表明乳腺癌的RR可能与BRCA2重叠,特别是在家族史较明显的患者中;因此,在大型研究中改进癌症风险评估仍很重要。此外,其他癌症(如胰腺癌)的风险定义不明确。鉴于人群中PALB2致病性突变的患病率较低,需要更多的数据来确定最佳的检测和适当的管理候选人。
直到20世纪90年代,遗传性乳腺癌和卵巢癌综合征的诊断都是基于临床表现和家族史。如今已经确定了这些综合征的一些相关基因,一些研究试图估计这些群体中的自发性致病突变率(新发致病突变率)。有趣的是,PJS、PTEN错构瘤综合征和LFS都被认为具有较高的自发性致病突变率,10%-30%, 虽然对BRCA基因中新发致病性突变的估计很低,但主要是基于发表的少数病例报告。此外,只有一个病例组的乳腺癌患者接受了BRCA致病性突突变的检测,其中发现了一种新的突突变。具体来说,在这项对193例散发性乳腺癌患者的研究中,检测到17种致病性突变,其中一种被证实是新发致病性突变。因此,根据有限数量的研究,新发致病性突变率似乎较低(≦5%)。同理,对与Lynch综合征相关的MMR基因中新发致病性突变的估计较低,0.9%-5%。然而,这些对BRCA基因和Lynch综合征基因中自发性致病突变率的估计似乎与不同人群中非父系率的估计重叠(0.6%-3.3%),使得这些基因的新发致病突变率相对较低。
Epidemiologic studies have clearly established the role of family history as an important risk factor for both breast and ovarian cancers. After gender and age, a positive family history is the strongest known predictive risk factor for breast cancer. However, it has long been recognized that in some families, there is hereditary breast cancer, which is characterized by an early age of onset, bilaterality, and the presence of breast cancer in multiple generations in an apparent autosomal dominant pattern of transmission (through either the maternal or the paternal lineage), sometimes including tumors of other organs, particularly the ovary and prostate gland.
It is now known that the cancer in some of these families can be explained by specific pathogenic variants in single cancer susceptibility genes. The isolation of several of these genes, which when altered are associated with a significantly increased risk of breast/ovarian cancer, makes it possible to identify individuals at risk. Although such cancer susceptibility genes are very important, highly penetrant germline pathogenic variants are estimated to account for only 5% to 10% of breast cancers overall.
A 1988 study reported the first quantitative evidence that breast cancer segregated as an autosomal dominant trait in some families.
The search for genes associated with hereditary susceptibility to breast cancer has been facilitated by studies of large kindreds with multiple affected individuals and has led to the identification of several susceptibility genes, including BRCA1, BRCA2, TP53, PTEN/MMAC1, and STK11. Other genes, such as the mismatch repair genes MLH1, MSH2, MSH6, and PMS2, have been associated with an increased risk of ovarian cancer, but have not been consistently associated with breast cancer.
In 1990, a susceptibility gene for breast cancer was mapped by genetic linkage to the long arm of chromosome 17, in the interval 17q12-21.
The linkage between breast cancer and genetic markers on chromosome 17q was soon confirmed by others, and evidence for the coincident transmission of both breast and ovarian cancer susceptibility in linked families was observed.
The BRCA1 gene was subsequently identified by positional cloning methods and has been found to contain 24 exons that encode a protein of 1,863 amino acids. Germline pathogenic variants in BRCA1 are associated with early-onset breast cancer, ovarian cancer, and fallopian tube cancer. (Refer to the Penetrance of BRCA pathogenic variants section of this summary for more information.) Male breast cancer, pancreatic cancer, testicular cancer, and early-onset prostate cancer may also be associated with pathogenic variants in BRCA1;
however, male breast cancer, pancreatic cancer, and prostate cancer are more strongly associated with pathogenic variants in BRCA2.
A second breast cancer susceptibility gene, BRCA2, was localized to the long arm of chromosome 13 through linkage studies of 15 families with multiple cases of breast cancer that were not linked to BRCA1. Pathogenic variants in BRCA2 are associated with multiple cases of breast cancer in families, and are also associated with male breast cancer, ovarian cancer, prostate cancer, melanoma, and pancreatic cancer.
(Refer to the Penetrance of BRCA pathogenic variants section of this summary for more information.) BRCA2 is a large gene with 27 exons that encode a protein of 3,418 amino acids.
While not homologous genes, both BRCA1 and BRCA2 have an unusually large exon 11 and translational start sites in exon 2. Like BRCA1, BRCA2 appears to behave like a tumor suppressor gene. In tumors associated with both BRCA1 and BRCA2 pathogenic variants, there is often loss of the wild-type allele.
Pathogenic variants in BRCA1 and BRCA2 appear to be responsible for disease in 45% of families with multiple cases of breast cancer only and in up to 90% of families with both breast and ovarian cancer.
Most BRCA1 and BRCA2 pathogenic variants are predicted to produce a truncated protein product, and thus loss of protein function, although some missense pathogenic variants cause loss of function without truncation. Because inherited breast/ovarian cancer is an autosomal dominant condition, persons with a BRCA1 or BRCA2 pathogenic variant on one copy of chromosome 17 or 13 also carry a normal allele on the other paired chromosome. In most breast and ovarian cancers that have been studied from carriers of pathogenic variants, deletion of the normal allele results in loss of all function, leading to the classification of BRCA1 and BRCA2 as tumor suppressor genes. In addition to, and as part of, their roles as tumor suppressor genes, BRCA1 and BRCA2 are involved in myriad functions within cells, including homologous DNA repair, genomic stability, transcriptional regulation, protein ubiquitination, chromatin remodeling, and cell cycle control.
Nearly 2,000 distinct variants and sequence variations in BRCA1 and BRCA2 have already been described.
Approximately 1 in 400 to 800 individuals in the general population may carry a germline pathogenic variant in BRCA1 or BRCA2.
The variants that have been associated with increased risk of cancer result in missing or nonfunctional proteins, supporting the hypothesis that BRCA1 and BRCA2 are tumor suppressor genes. While a small number of these pathogenic variants have been found repeatedly in unrelated families, most have not been reported in more than a few families.
Variant-screening methods vary in their sensitivity. Methods widely used in research laboratories, such as single-stranded conformational polymorphism analysis and conformation-sensitive gel electrophoresis, miss nearly a third of the variants that are detected by DNA sequencing.
In addition, large genomic alterations such as translocations, inversions, or large deletions or insertions are missed by most of the techniques, including direct DNA sequencing, but testing for these is commercially available. Such rearrangements are believed to be responsible for 12% to 18% of BRCA1 inactivating variants but are less frequently seen in BRCA2 and in individuals of Ashkenazi Jewish (AJ) descent.
Furthermore, studies have suggested that these rearrangements may be more frequently seen in Hispanic and Caribbean populations.
Germline pathogenic variants in the BRCA1/BRCA2 genes are associated with an approximately 60% lifetime risk of breast cancer and a 15% to 40% lifetime risk of ovarian cancer. There are no definitive functional tests for BRCA1 or BRCA2; therefore, the classification of nucleotide changes to predict their functional impact as deleterious or benign relies on imperfect data. The majority of accepted pathogenic variants result in protein truncation and/or loss of important functional domains. However, 10% to 15% of all individuals undergoing genetic testing with full sequencing of BRCA1 and BRCA2 will not have a clearly pathogenic variant detected but will have a variant of uncertain (or unknown) significance (VUS). VUS may cause substantial challenges in counseling, particularly in terms of cancer risk estimates and risk management. Clinical management of such patients needs to be highly individualized and must take into consideration factors such as the patient’s personal and family cancer history, in addition to sources of information to help characterize the VUS as benign or deleterious. Thus an improved classification and reporting system may be of clinical utility.
A comprehensive analysis of 7,461 consecutive full gene sequence analyses performed by Myriad Genetic Laboratories, Inc., described the frequency of VUS over a 3-year period.
Among subjects who had no clearly pathogenic variant, 13% had VUS defined as “missense mutations and mutations that occur in analyzed intronic regions whose clinical significance has not yet been determined, chain-terminating mutations that truncate BRCA1 and BRCA2 distal to amino acid positions 1853 and 3308, respectively, and mutations that eliminate the normal stop codons for these proteins.” The classification of a sequence variant as a VUS is a moving target. An additional 6.8% of subjects with no clear pathogenic variants had sequence alterations that were once considered VUS but were reclassified as a polymorphism, or occasionally as a pathogenic variant.
The frequency of VUS varies by ethnicity within the U.S. population. African Americans appear to have the highest rate of VUS.
In a 2009 study of data from Myriad, 16.5% of individuals of African ancestry had VUS, the highest rate among all ethnicities. The frequency of VUS in Asian, Middle Eastern, and Hispanic populations clusters between 10% and 14%, although these numbers are based on limited sample sizes. Over time, the rate of changes classified as VUS has decreased in all ethnicities, largely the result of improved variant classification algorithms.
VUS continue to be reclassified as additional information is curated and interpreted.
Such information may impact the continuing care of affected individuals.
A number of methods for discriminating deleterious from neutral VUS exist and others are in development
including integrated methods (see below).
Interpretation of VUS is greatly aided by efforts to track VUS in the family to determine if there is cosegregation of the VUS with the cancer in the family. In general, a VUS observed in individuals who also have a pathogenic variant, especially when the same VUS has been identified in conjunction with different pathogenic variants, is less likely to be in itself deleterious, although there are rare exceptions. As an adjunct to the clinical information, models to interpret VUS have been developed, based on sequence conservation, biochemical properties of amino acid changes,
incorporation of information on pathologic characteristics of BRCA1- and BRCA2-related tumors (e.g., BRCA1-related breast cancers are usually estrogen receptor [ER]–negative),
and functional studies to measure the influence of specific sequence variations on the activity of BRCA1 or BRCA2 proteins.
When attempting to interpret a VUS, all available information should be examined.
Statistics regarding the percentage of individuals found to be carriers of BRCA pathogenic variants among samples of women and men with a variety of personal cancer histories regardless of family history are provided below. These data can help determine who might best benefit from a referral for cancer genetic counseling and consideration of genetic testing but cannot replace a personalized risk assessment, which might indicate a higher or lower pathogenic variant likelihood based on additional personal and family history characteristics.
In some cases, the same pathogenic variant has been found in multiple apparently unrelated families. This observation is consistent with a founder effect, wherein a pathogenic variant identified in a contemporary population can be traced to a small group of founders isolated by geographic, cultural, or other factors. Most notably, two specific BRCA1 pathogenic variants (185delAG and 5382insC) and a BRCA2 pathogenic variant (6174delT) have been reported to be common in AJs. However, other founder pathogenic variants have been identified in African Americans and Hispanics.
The presence of these founder pathogenic variants has practical implications for genetic testing. Many laboratories offer directed testing specifically for ethnic-specific alleles. This greatly simplifies the technical aspects of the test but is not without limitations. Nonfounder BRCA pathogenic variants in the AJ population have been reported to be between 3% and 15%.
Among the general population, the likelihood of having any BRCA pathogenic variant is as follows:
Among AJ individuals, the likelihood of having any BRCA pathogenic variant is as follows:
Two large U.S. population-based studies of breast cancer patients younger than 65 years examined the prevalence of BRCA1 and BRCA2 pathogenic variants in various ethnic groups. The prevalence of BRCA1 pathogenic variants in breast cancer patients by ethnic group was 3.5% in Hispanics, 1.3% to 1.4% in African Americans, 0.5% in Asian Americans, 2.2% to 2.9% in non-AJ whites, and 8.3% to 10.2% in AJ individuals. The prevalence of BRCA2 pathogenic variants by ethnic group was 2.6% in African Americans and 2.1% in whites.
A study of Hispanic patients with a personal or family history of breast cancer and/or ovarian cancer, who were enrolled through multiple clinics in the southwestern United States, examined the prevalence of BRCA1 and BRCA2 pathogenic variants. BRCA pathogenic variants were identified in 189 of 746 patients (25%) (124 BRCA1, 65 BRCA2);
21 of the 189 (11%) BRCA pathogenic variants identified were large rearrangements, of which 13 (62%) were the BRCA1 exon 9–12 deletion. An unselected cohort of 810 women of Mexican ancestry with breast cancer were tested; 4.3% had a BRCA pathogenic variant. Eight of the 35 pathogenic variants identified also were the BRCA1 exon 9–12 deletion.
In another population-based cohort of 492 Hispanic women with breast cancer, the BRCA1 exon 9–12 deletion was found in three patients, suggesting that this variant may be a Mexican founder pathogenic variant and may represent 10% to 12% of all BRCA1 pathogenic variants in similar clinic- and population-based cohorts in the United States. Within the clinic-based cohort, there were nine recurrent pathogenic variants, which accounted for 53% of all variants observed in this cohort, suggesting the existence of additional founder pathogenic variants in this population.
A retrospective review of 29 AJ patients with primary fallopian tube tumors identified germline BRCA pathogenic variants in 17%.
Another study of 108 women with fallopian tube cancer identified pathogenic variants in 55.6% of the Jewish women and 26.4% of non-Jewish women (30.6% overall).
Estimates of the frequency of fallopian tube cancer in carriers of BRCA pathogenic variants are limited by the lack of precision in the assignment of site of origin for high-grade, metastatic, serous carcinomas at initial presentation.
Population screening has identified carriers in a number of AJ populations who would not have met criteria for family-based testing.
This could potentially expand the number of individuals who could benefit from preventive strategies. A study has suggested that population screening (compared with personal/family history–based testing) for AJ founder variants is cost-effective on the basis of data from the United States and the United Kingdom.
The authors used a decision-analytic model that estimated lifetime costs and the effects of genetic testing to assess cost-effectiveness; the model included costs of pretest genetic counseling and genetic testing and the anticipated risk of cardiovascular outcomes. Additional analyses conducted by the same group also suggested cost-effectiveness when testing was expanded to include all pathogenic variants in BRCA1, BRCA2, RAD51C, RAD51D, and PALB2.
These studies are based on various assumptions, some of which are imprecise (e.g., population prevalence estimates for some genes). Furthermore, as acknowledged by the authors, these types of efforts would require implementation of clinical support across the care continuum, in order for patients identified with pathogenic variants to benefit from this information. Consequently, there remain significant resource implications as population screening efforts are considered, which are the focus of ongoing research efforts. Because the detection rate is highly dependent on the prevalence of pathogenic variants in a population, it is not clear how applicable this approach would be for other populations, including other founder pathogenic variant populations. Another unanswered question is whether adequate genetic counseling can be provided for whole populations.
Several studies have assessed the frequency of BRCA1 or BRCA2 pathogenic variants in women with breast or ovarian cancer.
Personal characteristics associated with an increased likelihood of a BRCA1 and/or BRCA2 pathogenic variant include the following:
Family history characteristics associated with an increased likelihood of carrying a BRCA1 and/or BRCA2 pathogenic variant include the following:
Several professional organizations and expert panels, including the American Society of Clinical Oncology, the National Comprehensive Cancer Network (NCCN), the American Society of Human Genetics, the American College of Medical Genetics and Genomics, the National Society of Genetic Counselors, the U.S. Preventive Services Task Force, and the Society of Gynecologic Oncologists, have developed clinical criteria and practice guidelines that can be helpful to health care providers in identifying individuals who may have a BRCA1 or BRCA2 pathogenic variant.
Many models have been developed to predict the probability of identifying germline BRCA1/BRCA2 pathogenic variants in individuals or families. These models include those using logistic regression, genetic models using Bayesian analysis (BRCAPRO and Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm [BOADICEA]), and empiric observations, including the Myriad prevalence tables.
In addition to BOADICEA, BRCAPRO is commonly used for genetic counseling in the clinical setting. BRCAPRO and BOADICEA predict the probability of being a carrier and produce estimates of breast cancer risk (refer to Table 4). The discrimination and accuracy (factors used to evaluate the performance of prediction models) of these models are much higher for these models' ability to report on carrier status than for their ability to predict fixed or remaining lifetime risk.
BOADICEA is a polygenetic model that uses complex segregation analysis to examine both breast cancer risk and the probability of having a BRCA1 or BRCA2 pathogenic variant.
Even among experienced providers, the use of prediction models has been shown to increase the power to discriminate which patients are most likely to be carriers of BRCA1/BRCA2 pathogenic variants.
Most models do not include other cancers seen in the BRCA1 and BRCA2 spectrum, such as pancreatic cancer and prostate cancer. Interventions that decrease the likelihood that an individual will develop cancer (such as oophorectomy and mastectomy) may influence the ability to predict BRCA1 and BRCA2 pathogenic variant status.
One study has shown that the prediction models for genetic risk are sensitive to the amount of family history data available and do not perform as well with limited family information.
BOADICEA is being expanded to incorporate additional risk variants (genome-wide association study [GWAS] single nucleotide polymorphisms [SNPs]) to better predict pathogenic variant status and to improve the accuracy of breast cancer and ovarian cancer risk estimates.
The performance of the models can vary in specific ethnic groups. The BRCAPRO model appeared to best fit a series of French Canadian families.
There have been variable results in the performance of the BRCAPRO model among Hispanics, and both the BRCAPRO model and Myriad tables underestimated the proportion of carriers of pathogenic variants in an Asian American population.
BOADICEA was developed and validated in British women. Thus, the major models used for both overall risk (Table 1) and genetic risk (Table 4) have not been developed or validated in large populations of racially and ethnically diverse women. Of the commonly used clinical models for assessing genetic risk, only the Tyrer-Cuzick model contains nongenetic risk factors.
The power of several of the models has been compared in different studies.
Four breast cancer genetic-risk models, BOADICEA, BRCAPRO, IBIS, and eCLAUS, were evaluated for their diagnostic accuracy in predicting BRCA1/BRCA2 pathogenic variants in a cohort of 7,352 German families.
The family member with the highest likelihood of carrying a pathogenic variant from each family was screened for BRCA1/BRCA2 pathogenic variants. Carrier probabilities from each model were calculated and compared with the actual variants detected. BRCAPRO and BOADICEA had significantly higher diagnostic accuracy than IBIS or eCLAUS. Accuracy for the BOADICEA model was further improved when statuses of the tumor markers ER, progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2/neu) were included in the model. The inclusion of these biomarkers has been shown to improve the performance of BRCAPRO.
Myriad Prevalence Tables | BRCAPRO | BOADICEA | Tyrer-Cuzick | |
---|---|---|---|---|
Method | Empiric data from Myriad Genetics based on personal and family history reported on requisition forms | Statistical model, assumes autosomal dominant inheritance | Statistical model, assumes polygenic risk | Statistical model, assumes autosomal dominant inheritance |
Features of the model | Proband may or may not have breast or ovarian cancer | Proband may or may not have breast or ovarian cancer | Proband may or may not have breast or ovarian cancer | Proband must be unaffected |
Considers age of breast cancer diagnosis as <50 y, >50 y | Considers exact age at breast and ovarian cancer diagnosis | Considers exact age at breast and ovarian cancer diagnosis | Also includes reproductive factors and body mass index to estimate breast cancer risk | |
Considers breast cancer in ≥1 affected relative only if diagnosed <50 y | Considers prior genetic testing in family (i.e., BRCA1/BRCA2 pathogenic variant–negative relatives) | Includes all FDR and SDR with and without cancer | ||
Considers ovarian cancer in ≥1 relative at any age | Considers oophorectomy status | Includes AJ ancestry | ||
Includes AJ ancestry | Includes all FDR and SDR with and without cancer | |||
Very easy to use | Includes AJ ancestry | |||
Limitations | Simplified/limited consideration of family structure | Requires computer software and time-consuming data entry | Requires computer software and time-consuming data entry | Designed for individuals unaffected with breast cancer |
Incorporates only FDR and SDR; may need to change proband to best capture risk and to account for disease in the paternal lineage | ||||
May overestimate risk in bilateral breast cancer | ||||
Early age of breast cancer onset | May perform better in whites than minority populations | Incorporates only FDR and SDR; may need to change proband to best capture risk | ||
May underestimate risk of BRCA pathogenic variant in high-grade serous ovarian cancers but overestimate the risk for other histologies | ||||
AJ = Ashkenazi Jewish; BOADICEA = Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm; FDR = first-degree relatives; SDR = second-degree relatives. |
Genetic testing for BRCA1 and BRCA2 pathogenic variants has been available to the public since 1996. As more individuals have undergone testing, risk assessment models have improved. This, in turn, gives providers better data to estimate an individual patient’s risk of carrying a pathogenic variant, but risk assessment continues to be an art. There are factors that might limit the ability to provide an accurate risk assessment (i.e., small family size, paucity of women, or ethnicity) including the specific circumstances of the individual patient (such as history of disease or risk-reducing surgeries).
The proportion of individuals carrying a pathogenic variant who will manifest the disease is referred to as penetrance. (Refer to the Penetrance of Inherited Susceptibility to Hereditary Breast and/or Gynecologic Cancers section of this summary for more information.)
Numerous studies have estimated breast and ovarian cancer penetrance in carriers of BRCA1 and BRCA2 pathogenic variants. Risk of both breast and ovarian cancer is consistently estimated to be higher in carriers of BRCA1 pathogenic variants than in carriers of BRCA2 pathogenic variants. Results from two large meta-analyses are shown in Table 5.
One study analyzed pooled pedigree data from 22 studies involving 289 BRCA1 and 221 BRCA2 pathogenic variant–positive individuals. Index cases from these studies had female breast cancer, male breast cancer, or ovarian cancer but were unselected for family history. A subsequent study combined penetrance estimates from the previous study and nine others that included an additional 734 BRCA1 and 400 BRCA2 pathogenic variant–positive families. The estimated cumulative risks of breast cancer by age 70 years in these two meta-analyses were 55% to 65% for carriers of BRCA1 pathogenic variants and 45% to 47% for carriers of BRCA2 pathogenic variants. Ovarian cancer risks were 39% for carriers of BRCA1 pathogenic variants and 11% to 17% for carriers of BRCA2 pathogenic variants.
Study | Breast Cancer Risk (%) (95% CI) | Ovarian Cancer Risk (%) (95% CI) | ||
---|---|---|---|---|
BRCA1 | BRCA2 | BRCA1 | BRCA2 | |
Antoniou et al. (2003) | 65 (44–78) | 45 (31–56) | 39 (18–54) | 11 (2.4–19) |
Chen et al. (2007) | 55 (50–59) | 47 (42–51) | 39 (34–45) | 17 (13–21) |
Kuchenbaecker et al. (2017) | 72 (65–79) | 69 (61–77) | 44 (36–53) | 17 (11–25) |
CI = confidence interval. | ||||
aRisk estimate calculated up to age 70 years. | ||||
bRisk estimate calculated up to age 80 years. |
While the cumulative risks of developing cancer by age 70 years are higher for carriers of BRCA1 pathogenic variants than for BRCA2 pathogenic variants, the relative risks (RRs) of breast cancer decline more with age in carriers of BRCA1 pathogenic variants.
Studies of penetrance for carriers of specific individual variants are not usually large enough to provide stable estimates, but numerous studies of the Ashkenazi founder pathogenic variants have been conducted. One group of researchers analyzed the subset of families with one of the Ashkenazi founder pathogenic variants from their larger meta-analyses and found that the estimated penetrance for the individual pathogenic variants was very similar to the corresponding estimates among all carriers.
A later study of 4,649 women with BRCA pathogenic variants reported significantly lower RRs of breast cancer in those with the BRCA2 6174delT variant than in those with other BRCA2 variants (hazard ratio [HR], 0.35; confidence interval [CI], 0.18–0.69).
One study provided prospective 10-year risks of developing cancer among asymptomatic carriers at various ages.
Nonetheless, making precise penetrance estimates in an individual carrier is difficult. The lifetime risks of ovarian cancer are 5.2% in carriers of RAD51C pathogenic variants, 5.8% in carriers of BRIP1 pathogenic variants, and 12% in carriers of RAD51D pathogenic variants. Risk-reducing salpingo-oophorectomy (RRSO) may be considered for these patients upon completion of childbearing.
Data from the Consortium of Investigators of Modifiers of BRCA1/BRCA2 (CIMBA), comprising 19,581 carriers of BRCA1 pathogenic variants and 11,900 carriers of BRCA2 pathogenic variants, were analyzed to estimate HRs for breast cancer and ovarian cancer by pathogenic variant type, function, and nucleotide position.
Breast cancer cluster regions and ovarian cancer cluster regions were found in both genes. Risks for incidence of breast cancer and ovarian cancer and age at diagnosis differed by variant class. Further evaluation of these findings is needed before they can be translated into clinical practice.
Another study from the CIMBA group looked at the phenotype of women with breast cancer who had inherited pathogenic variants in both BRCA1 and BRCA2.
The majority of women carried the common Jewish pathogenic variants. Compared with women who were heterozygous for the same pathogenic variant (heterozygote controls), women who were heterozygous for both BRCA1 and BRCA2 were more likely to be diagnosed with breast cancer than women who were heterozygote controls, and more likely to be diagnosed with ovarian cancer than women who were heterozygote controls with BRCA2, but not those with BRCA1 pathogenic variants. Similarly, age at onset of breast cancer was younger in carriers of both variants compared with women who were heterozygote controls with BRCA2, but not compared with those with BRCA1 pathogenic variants. The percentage of women with both variants and estrogen receptor–positive and progesterone receptor–positive breast cancer was intermediate between the heterozygote controls with BRCA1 pathogenic variants and those with BRCA2 pathogenic variants. The authors concluded that women who inherit pathogenic variants in both BRCA1 and BRCA2 may be managed similarly to carriers of only a BRCA1 variant.
Several studies have suggested that BRCA pathogenic variants may be associated with genetic anticipation. One study evaluated 176 families with BRCA1 or BRCA2 pathogenic variants and at least two consecutive generations of the same cancer. The probands’ generations were diagnosed with breast cancer an estimated 6.8 years earlier than the parents’ generations and 9.8 years earlier than the grandparents' generations.
Similarly, another study showed a difference in age at breast cancer diagnosis between 80 mother-and-daughter paired pathogenic variant carriers but only if the mother was diagnosed with breast cancer after age 50 years.
Another cohort study of 106 paired women from two consecutive generations with a known BRCA pathogenic variant in the family estimated a 6- to 8-year earlier age at onset in subsequent generations.
RRSO and/or use of oral contraceptives have been associated with the risk of breast cancer.
(Refer to the RRSO section and the Oral contraceptives section of this summary for more information.) Other potentially modifiable reproductive and hormonal factors can also affect risk.
Genetic modifiers of penetrance of breast cancer and ovarian cancer are increasingly under study but are not clinically useful at this time.
(Refer to the Modifiers of risk in carriers of BRCA1 and BRCA2 pathogenic variants section for more information.) While the average breast cancer and ovarian cancer penetrances may not be as high as initially estimated, they are substantial, both in relative and absolute terms, particularly in women born after 1940. A higher risk before age 50 years has been consistently seen in more recent birth cohorts,
and additional studies will be required to further characterize potential modifying factors to arrive at more precise individual risk projections. Precise penetrance estimates for less common cancers, such as pancreatic cancer, are lacking.
The increased risk of CBC among carriers of BRCA1 and BRCA2 pathogenic variants has been confirmed in several large studies, with fairly consistent results, as summarized in Table 6.
Study | BRCA1 Carriers (%) | BRCA2 Carriers (%) |
---|---|---|
Graeser et al. (2009) | 18.5 | 13.2 |
Malone et al. (2010) | 20.5 | 15.9 |
van der Kolk et al. (2010) | 34.2 | 29.2 |
Metcalfe et al. (2011) | 23.8 | 18.7 |
Molina-Montes et al. (2014) | 27 | 19 |
Basu et al. (2015) | 25.7 | 19.5 |
van den Broek et al. (2016) | 21.1 | 10.8 |
Published results include a large study by the German Consortium for Hereditary Breast and Ovarian Cancer, which estimated the risk of CBC in members of families with known BRCA1 and BRCA2 pathogenic variants. At 25 years after the first breast cancer, the risk of CBC was close to 50% in both BRCA1 and BRCA2 families. The risk was also inversely correlated with age in this study, with the highest risks seen in women whose first breast cancer was before age 40 years.
Subsequently, results from the Women's Environmental Cancer and Radiation Epidemiology (WECARE) study, a large, population-based, nested case-control study of CBC, reported a 10-year risk of CBC of 15.9% among carriers of BRCA1/BRCA2 pathogenic variants and a risk of 4.9% among noncarriers. Risks were also inversely related to age at first diagnosis in this study and were 1.8-fold higher in those with a first-degree relative (FDR) with breast cancer.
A larger study of members of BRCA1/BRCA2 families in the Netherlands reported similar 10-year risks of CBC for women from BRCA1 and BRCA2 families (34.2% and 29.2%, respectively).
A comparison of 655 women with BRCA1/BRCA2 pathogenic variants undergoing either breast-conserving therapy or mastectomy noted that both treatment groups experienced high rates of CBC, exceeding 50% by 20 years of follow-up. Rates were significantly higher among women with BRCA1 pathogenic variants than in women with BRCA2 pathogenic variants, and among women whose first breast cancer occurred at or before age 35 years.
In a study of 810 women with stage I or stage II breast cancer who had a BRCA1 or BRCA2 pathogenic variant identified in the family, 149 (18.4%) developed CBC; the 15-year actuarial risk was 36.1% among carriers of BRCA1 pathogenic variants and 28.5% among carriers of BRCA2 pathogenic variants.
Risks were higher among women diagnosed before age 50 years than among women diagnosed at age 50 years or older (37.6% vs. 16.8%; P = .003). Furthermore, the risk of CBC varied by family history among women whose initial breast cancer was diagnosed before age 50 years. For these women, the CBC risk among those with 0, 1, or 2 or more FDRs with breast cancer diagnosed before age 50 years was 33.4%, 39.1%, and 49.7%, respectively.
The risk of CBC after a first breast cancer in BRCA1 and BRCA2 carriers has been examined in both retrospective and prospective observational epidemiological studies. A systematic review and quantitative meta-analysis of these epidemiologic studies (18 retrospective and 2 prospective cohort studies) reported 5-year cumulative risks of CBC of 15% (95% CI, 9.50%–20%) in BRCA1 carriers and 9% (95% CI, 5%–14%) in BRCA2 carriers.
When the prospective studies were analyzed separately, the 5-year cumulative risk increased to 23.4% (95% CI, 9.1%–39.5%) in BRCA1 carriers and to 17.5% (95% CI, 9.1%–39.5%) in BRCA2 carriers. The discrepancies in the reported frequencies may be inherent due to the potential for biases introduced in retrospective series.
Similarly, in a Dutch cohort of 6,294 patients (including 200 BRCA1 carriers and 71 BRCA2 carriers) with invasive breast cancer diagnosed before age 50 years, and a median follow-up of 12.5 years, the 10-year risks of CBC were 21.1% (95% CI, 15.4%–27.4%) for BRCA1 carriers, 10.8% (95% CI, 4.7%–19.6%) for BRCA2 carriers, and 5.1% (95% CI, 4.5%–5.7%) for noncarriers.
Age at first breast cancer diagnosis was predictive of the 10-year cumulative risk of CBC among BRCA1/BRCA2 carriers only. Specifically, the CBC risk among BRCA1/BRCA2 carriers diagnosed before age 41 years was 23.9% (BRCA1, 25.5%; BRCA2, 17.2%); in contrast, CBC among those diagnosed between 41 and 49 years was 12.6% (BRCA1, 15.6%; BRCA2, 7.2%).
In an English study of 506 BRCA1 carriers and 505 BRCA2 carriers with a diagnosis of breast cancer at any age and median follow-up of 7.8 years, the 10-year risks for CBC were 25.7% for BRCA1 carriers and 19.5% for BRCA2 carriers.
Earlier age at first breast cancer diagnosis for BRCA1 and BRCA2 carriers combined was significantly associated with a higher CBC risk, with a 20-year rate of 55.4% among those younger than 40 years, compared with 36.4% among those older than 50 years. Additionally, differences were more pronounced among BRCA1 carriers compared with BRCA2 carriers.
An international, multicenter, prospective cohort study followed 1,305 BRCA1 and 908 BRCA2 female carriers with a diagnosis of breast cancer (without any other cancers) for a median follow-up time of 4 years (range, 2–7 y).
Participants had a median age of 47 years (range, 40–55 y) at the start of follow-up. The authors reported a cumulative risk of CBC 20 years after the initial breast cancer diagnosis of 40% (95% CI, 35%–45%) for BRCA1 carriers and 26% (95% CI, 20%–33%) for BRCA2 carriers. These 20-year estimates are in line with the 10-year cumulative risk estimates reported in Table 6.
Thus, in summary, despite differences in study design, study sites, and sample sizes, the data on CBC among women with BRCA1/BRCA2 pathogenic variants show several consistent findings:
Refer to the Risk-reducing mastectomy section of this summary for information about the use of risk-reducing surgery in carriers of BRCA pathogenic variants. Refer to the Chemoprevention section of this summary for information about the use of tamoxifen as a risk-reduction strategy for CBC in carriers of BRCA pathogenic variants.
Two genetic registry–based studies have recently explored the risk of primary breast cancer after BRCA-related ovarian cancer. In one study, 164 BRCA1/BRCA2 carriers with primary epithelial ovarian, fallopian tube or primary peritoneal cancer were followed for subsequent events.
The risk of metachronous breast cancer at 5 years after a diagnosis of ovarian cancer was lower than previously reported for unaffected BRCA1/BRCA2 carriers. In this series, overall survival was dominated by ovarian cancer-related deaths. A similar study compared the risk of primary breast cancer in BRCA-related ovarian cancer patients and unaffected carriers.
The 2-year, 5-year, and 10-year risks of primary breast cancer were all statistically significantly lower in patients with ovarian cancer. The risk of CBC among women with a unilateral breast cancer before their ovarian cancer diagnosis was also lower than in women without ovarian cancer, although the difference did not reach statistical significance. These studies suggest that treatment for ovarian cancer, namely oophorectomy and platinum-based chemotherapy, may confer protection against subsequent breast cancer. In a single-institution cohort study of 364 patients with epithelial ovarian cancer who underwent BRCA pathogenic variant testing, 135 (37.1%) were found to carry a germline BRCA1 or BRCA2 pathogenic variant. Of the 135 BRCA1/BRCA2 carriers, 12 (8.9%) developed breast cancer. All breast cancers were stage 0 to stage II and diagnosed as follows: mammogram (7), palpable mass (3), and incidental finding during risk-reducing mastectomy (2). At median follow-up of 6.3 years, of the 12 patients with breast cancer after ovarian cancer, three died of recurrent ovarian cancer and one died of metastatic breast cancer.
The majority of these cancers were detected with mammogram or clinical exam, suggesting additional breast surveillance with other modalities or risk-reducing surgery may be of questionable value. Mathematical modeling suggests that for women with BRCA-associated ovarian cancer, breast cancer screening should consist of mammography and clinical breast exam. The consideration of breast magnetic resonance imaging (MRI) and/or risk-reducing mastectomies may be beneficial for women with early-stage ovarian cancer or for long-term ovarian cancer survivors.
Female breast and ovarian cancers are clearly the dominant cancers associated with BRCA1 and BRCA2. BRCA pathogenic variants also confer an increased risk of fallopian tube and primary peritoneal carcinomas. One large study from a familial registry of carriers of BRCA1 pathogenic variants has found a 120-fold RR of fallopian tube cancer among carriers of BRCA1 pathogenic variants compared with the general population.
The risk of primary peritoneal cancer among carriers of BRCA pathogenic variants with intact ovaries is increased but remains poorly quantified, despite a residual risk of 3% to 4% in the 20 years after RRSO.
(Refer to the RRSO section in the Ovarian cancer section of this summary for more information.)
Pancreatic, male breast, and prostate cancers have also been consistently associated with BRCA pathogenic variants, particularly with BRCA2. Other cancers have been associated in some studies. The strength of the association of these cancers with BRCA pathogenic variants has been more difficult to estimate because of the lower numbers of these cancers observed in carriers of pathogenic variants.
Men with BRCA2 pathogenic variants, and to a lesser extent BRCA1 pathogenic variants, are at increased risk of breast cancer with lifetime risks estimated at 5% to 10% and 1% to 2%, respectively.
Men carrying BRCA2 pathogenic variants, and to a lesser extent BRCA1 pathogenic variants, have an approximately threefold to sevenfold increased risk of prostate cancer.
BRCA2-associated prostate cancer also appears to be more aggressive.
(Refer to the section in the PDQ summary on Genetics of Prostate Cancer for more information.)
Studies of familial pancreatic cancer (FPC) and unselected series of pancreatic cancer have also supported an association with BRCA2, and to a lesser extent, BRCA1.
Overall, it appears that between 3% to 15% of families with FPC may have germline BRCA2 pathogenic variants, with risks increasing with more affected relatives.
Similarly, studies of unselected pancreatic cancers have reported BRCA2 pathogenic variant frequencies between 3% to 7%, with these numbers approaching 10% in those of AJ descent.
The lifetime risk of pancreatic cancer in BRCA2 carriers is estimated to be 3% to 5%,
compared with an estimated lifetime risk of 0.5% by age 70 years in the general population.
A large, single-institution study of more than 1,000 carriers of pathogenic variants found a 21-fold increased risk of pancreatic cancer among BRCA2 carriers and a 4.7-fold increased risk among carriers of BRCA1 pathogenic variants, compared with incidence in the general population.
Other cancers associated with BRCA2 pathogenic variants in some, but not all, studies include melanoma, biliary cancers, and head and neck cancers, but these risks appear modest (<5% lifetime risk) and are less well studied.
Cancer Sites | BRCA1 | BRCA2 | ||
---|---|---|---|---|
Strength of Evidence | Magnitude of Absolute Risk | Strength of Evidence | Magnitude of Absolute Risk | |
Breast (female) | +++ | High | +++ | High |
Ovary, fallopian tube, peritoneum | +++ | High | +++ | Moderate |
Breast (male) | + | Undefined | +++ | Low |
Pancreas | ++ | Very Low | +++ | Low |
Prostate | + | Undefined | +++ | High |
aRefer to the PDQ summary on Genetics of Prostate Cancer for more information about the association of BRCA1 and BRCA2 with prostate cancer. | ||||
+++ Multiple studies demonstrated association and are relatively consistent. | ||||
++ Multiple studies and the predominance of the evidence are positive. | ||||
+ May be an association, predominantly single studies; smaller limited studies and/or inconsistent but weighted toward positive. |
The first Breast Cancer Linkage Consortium study investigating cancer risks reported an excess of colorectal cancer in BRCA1 carriers (RR, 4.1; 95% CI, 2.4–7.2).
This finding was supported by some, but not all, family-based studies. However, unselected series of colorectal cancer that have been exclusively performed in the AJ population have not shown elevated rates of BRCA1 or BRCA2 pathogenic variants.
Taken together, the data suggest little, if any, increased risk of colorectal cancer, and possibly only in specific population groups. Therefore, at this time, carriers of BRCA1 pathogenic variants should adhere to population-screening recommendations for colorectal cancer.
No increased prevalence of hereditary BRCA pathogenic variants was found among 200 Jewish women with endometrial carcinoma or 56 unselected women with uterine papillary serous carcinoma.
(Refer to the Risk-reducing salpingo-oophorectomy section in the Ovarian cancer section of this summary for more information.)
There is conflicting evidence as to the residual familial risk among women who test negative for the BRCA1/BRCA2 pathogenic variant segregating in the family. An initial study based on prospective evaluation of 353 women who tested negative for the BRCA1 pathogenic variant segregating in the family found that five incident breast cancers occurred during more than 6,000 person-years of observation, for a lifetime risk of 6.8%, a rate similar to the general population.
A report that the risk may be as high as fivefold in women who tested negative for the BRCA1 or BRCA2 pathogenic variant in the family
was followed by numerous letters to the editor suggesting that ascertainment biases account for much of this observed excess risk.
Four additional analyses have suggested an approximate 1.5-fold to 2-fold excess risk.
In one study, two cases of ovarian cancer were reported.
Several studies have involved retrospective analyses; all studies have been based on small observed numbers of cases and have been of uncertain statistical and clinical significance.
Results from numerous other prospective studies have found no increased risk. A study of 375 women who tested negative for a known familial pathogenic variant in BRCA1 or BRCA2 reported two invasive breast cancers, two in situ breast cancers, and no ovarian cancers diagnosed, with a mean follow-up of 4.9 years. Four invasive breast cancers were expected, whereas two were observed.
Another study of similar size but longer follow-up (395 women and 7,008 person-years of follow-up) also found no statistically significant overall increase in breast cancer risk among variant-negative women (observed/expected [O/E], 0.82; 95% CI, 0.39–1.51), although women who had at least one FDR with breast cancer had a nonsignificant increased risk (O/E, 1.33; 95% CI, 0.41–2.91).
A study of 160 BRCA1 and 132 BRCA2 pathogenic variant–positive families from the Breast Cancer Family Registry found no evidence for increased risk among noncarriers in these families.
In a large study of 722 variant-negative women from Australia in whom six invasive breast cancers were observed after a median follow-up of 6.3 years, the standardized incidence ratio (SIR) was not significantly elevated (SIR, 1.14; 95% CI, 0.51–2.53).
Based on available data, it appears that women testing negative for known familial BRCA1/BRCA2 pathogenic variants can adhere to general population screening guidelines unless they have sufficient additional risk factors, such as a personal history of atypical hyperplasia of the breast or family history of breast cancer in relatives who do not carry the familial pathogenic variant.
The majority of families with site-specific breast cancer test negative for BRCA1/BRCA2 and have no features consistent with Cowden syndrome or Li-Fraumeni syndrome.
Five studies using population-based and clinic-based approaches have demonstrated no increased risk of ovarian cancer in such families. Although ovarian cancer risk was not increased, breast cancer risk remained elevated.
Pathogenic variants in BRCA1 and BRCA2 confer high risks of breast and ovarian cancers. The risks, however, are not equal in all pathogenic variant carriers and have been found to vary by several factors, including type of cancer, age at onset, and variant position.
This observed variation in penetrance has led to the hypothesis that other genetic and/or environmental factors modify cancer risk in carriers of pathogenic variants. There is a growing body of literature identifying genetic and nongenetic factors that contribute to the observed variation in rates of cancers seen in families with BRCA1/BRCA2 pathogenic variants.
The largest studies investigating genetic modifiers of breast and ovarian cancer risk to date have come from CIMBA, a large international effort with genotypic and phenotypic data on more than 15,000 BRCA1 and 10,000 BRCA2 carriers.
Using candidate gene analysis and GWAS, CIMBA has identified several loci associated both with increased and decreased risk of breast cancer and ovarian cancer. Some of the SNPs are related to subtypes of breast cancer, such as hormone-receptor and HER2/neu status. The risks conferred are all modest but if operating in a multiplicative fashion could significantly impact risk of cancer in carriers of BRCA1/BRCA2 pathogenic variants. Currently, these SNPs are not being tested for or used in clinical decision making.
Some genotype-phenotype correlations have been identified in both BRCA1 and BRCA2 pathogenic variant families. None of the studies have had sufficient numbers of pathogenic variant–positive individuals to make definitive conclusions, and the findings are probably not sufficiently established to use in individual risk assessment and management. In 25 families with BRCA2 pathogenic variants, an ovarian cancer cluster region was identified in exon 11 bordered by nucleotides 3,035 and 6,629.
A study of 164 families with BRCA2 pathogenic variants collected by the Breast Cancer Linkage Consortium confirmed the initial finding. Pathogenic variants within the ovarian cancer cluster region were associated with an increased risk of ovarian cancer and a decreased risk of breast cancer in comparison with families with variants on either side of this region.
In addition, a study of 356 families with protein-truncating BRCA1 pathogenic variants collected by the Breast Cancer Linkage Consortium reported breast cancer risk to be lower with variants in the central region (nucleotides 2,401–4,190) compared with surrounding regions. Ovarian cancer risk was significantly reduced with variants 3’ to nucleotide 4,191.
These observations have generally been confirmed in subsequent studies.
Studies in Ashkenazim, in whom substantial numbers of families with the same pathogenic variant can be studied, have also found higher rates of ovarian cancer in carriers of the BRCA1:185delAG variant, in the 5' end of BRCA1, compared with carriers of the BRCA1:5382insC variant in the 3' end of the gene.
The risk of breast cancer, particularly bilateral breast cancer, and the occurrence of both breast and ovarian cancer in the same individual, however, appear to be higher in carriers of the BRCA1:5382insC pathogenic variant compared with carriers of BRCA1:185delAG and BRCA2:6174delT variants. Ovarian cancer risk is considerably higher in carriers of BRCA1 pathogenic variants, and it is uncommon before age 45 years in carriers of the BRCA2:6174delT pathogenic variant.
In an Australian study of 122 families with a pathogenic variant in BRCA1, large genomic rearrangement variants were associated with higher-risk features in breast and ovarian cancers, including younger age at breast cancer diagnosis and higher incidence of bilateral breast cancer.
Several studies evaluating pathologic patterns seen in BRCA1-associated breast cancers have suggested an association with adverse pathologic and biologic features. These findings include higher than expected frequencies of medullary histology, high histologic grade, areas of necrosis, trabecular growth pattern, aneuploidy, high S-phase fraction, high mitotic index, and frequent TP53 variants.
In a large international series of 3,797 carriers of BRCA1 pathogenic variants, the median age at breast cancer diagnosis was 40 years.
Of breast tumors arising in BRCA1 carriers, 78% were ER-negative; 79% were PR-negative; 90% were HER2-negative; and 69% were triple-negative. These findings were consistent with multiple smaller series.
In addition, the proportion of ER-negative tumors significantly decreased as the age at breast cancer diagnosis increased.
There is considerable, but not complete, overlap between the triple-negative and basal-like subtype cancers, both of which are common in BRCA1-associated breast cancer,
particularly in women diagnosed before age 50 years.
A small proportion of BRCA1-related breast cancers are ER-positive, which are associated with later age of onset.
These ER-positive cancers have clinical behavior features that are intermediate between ER-negative BRCA1 cancers and ER-positive sporadic breast cancers, raising the possibility that there may be a unique mechanism by which they develop.
The prevalence of germline BRCA1 pathogenic variants in women with triple-negative breast cancer is significant, both in women undergoing clinical genetic testing (and thus selected in large part for family history) and in unselected triple-negative patients, with pathogenic variants reported in 9% to 35%.
Notably, studies have demonstrated a high rate of BRCA1 pathogenic variants in unselected women with triple-negative breast cancer, particularly in those diagnosed before age 50 years. A large report of 1,824 patients with triple-negative breast cancer unselected for family history, recruited through 12 studies, identified 14.6% with a pathogenic variant in an inherited cancer susceptibility gene.
BRCA1 pathogenic variants accounted for the largest proportion (8.5%), followed by BRCA2 (2.7%); PALB2 (1.2%); and BARD1, RAD51D, RAD51C and BRIP1 (0.3%–0.5% for each gene). In this study, those with pathogenic variants in BRCA1/BRCA2 or other inherited cancer genes were diagnosed at an earlier age and had higher grade tumors than those without pathogenic variants. Specifically, among carriers of BRCA1 pathogenic variants, the average age at diagnosis was 44 years, and 94% had high-grade tumors. One study examined 308 individuals with triple-negative breast cancer; BRCA1 pathogenic variants were present in 45. Pathogenic variants were seen both in women unselected for family history (11 of 58; 19%) and in those with family history (26 of 111; 23%).
A meta-analysis based on 2,533 patients from 12 studies was conducted to assess the risk of a BRCA1 pathogenic variant in high-risk women with triple-negative breast cancer.
Results indicated that the RR of a BRCA1 pathogenic variant among women with versus without triple-negative breast cancer is 5.65 (95% CI, 4.15–7.69), and approximately two in nine women with triple-negative disease harbor a BRCA1 pathogenic variant. Interestingly, a study of 77 unselected patients with triple-negative breast cancer in which 15 (19.5%) had a germline pathogenic variant or somatic BRCA1/BRCA2 mutation demonstrated a lower risk of relapse in those with BRCA1 pathogenic variant–associated triple-negative breast cancer than in those with non-BRCA1-associated triple-negative breast cancer; this study was limited by its size.
A second study examining clinical outcomes in BRCA1-associated versus non-BRCA1-associated triple-negative breast cancer showed no difference, although there was a trend toward more brain metastases in those with BRCA1-associated breast cancer. In both of these studies, all but one carrier of BRCA1 pathogenic variants received chemotherapy.
In contrast, HER2 positivity and young age alone in the absence of family history or a second primary cancer does not increase the likelihood of a pathogenic variant in BRCA1, BRCA2, or TP53.
It has been hypothesized that many BRCA1 tumors are derived from the basal epithelial layer of cells of the normal mammary gland, which account for 3% to 15% of unselected invasive ductal cancers. If the basal epithelial cells of the breast represent the breast stem cells, the regulatory role suggested for wild-type BRCA1 may partly explain the aggressive phenotype of BRCA1-associated breast cancer when BRCA1 function is damaged.
Further studies are needed to fully appreciate the significance of this subtype of breast cancer within the hereditary syndromes.
The most accurate method for identifying basal-like breast cancers is through gene expression studies, which have been used to classify breast cancers into biologically and clinically meaningful groups.
This technology has also been shown to correctly differentiate BRCA1- and BRCA2-associated tumors from sporadic tumors in a high proportion of cases.
Notably, among a set of breast tumors studied by gene expression array to determine molecular phenotype, all tumors with BRCA1 alterations fell within the basal tumor subtype;
however, this technology is not in routine use due to its high cost. Instead, immunohistochemical markers of basal epithelium have been proposed to identify basal-like breast cancers, which are typically negative for ER, PR, and HER2, and stain positive for cytokeratin 5/6, or epidermal growth factor receptor.
Based on these methods to measure protein expression, a number of studies have shown that the majority of BRCA1-associated breast cancers are positive for basal epithelial markers.
There is growing evidence that preinvasive lesions are a component of the BRCA phenotype. The Breast Cancer Linkage Consortium initially reported a relative lack of an in situ component in BRCA1-associated breast cancers,
also seen in two subsequent studies of BRCA1/BRCA2 carriers.
However, in a study of 369 ductal carcinoma in situ (DCIS) cases, BRCA1 and BRCA2 pathogenic variants were detected in 0.8% and 2.4%, respectively, which is only slightly lower than previously reported prevalence in studies of invasive breast cancer patients.
A retrospective study of breast cancer cases in a high-risk clinic found similar rates of preinvasive lesions, particularly DCIS, among 73 BRCA-associated breast cancers and 146 pathogenic variant–negative cases.
A study of AJ women, stratified by whether they were referred to a high-risk clinic or were unselected, showed similar prevalence of DCIS and invasive breast cancers in referred patients compared with one-third lower DCIS cases among unselected subjects.
Similarly, data about the prevalence of hyperplastic lesions have been inconsistent, with reports of increased and decreased prevalence.
Similar to invasive breast cancer, DCIS diagnosed at an early age and/or with a family history of breast and/or ovarian cancer is more likely to be associated with a BRCA1/BRCA2 pathogenic variant.
Overall evidence suggests DCIS is part of the BRCA1/BRCA2 spectrum, particularly BRCA2; however, the prevalence of pathogenic variants in DCIS patients, unselected for family history, is less than 5%.
The phenotype for BRCA2-related tumors appears to be more heterogeneous and is less well-characterized than that of BRCA1, although they are generally positive for ER and PR.
A large international series of 2,392 carriers of BRCA2 pathogenic variants found that only 23% of tumors arising in carriers of BRCA2 pathogenic variants were ER-negative; 36% were PR-negative; 87% were HER2-negative; and 16% were triple-negative.
A large report of 1,824 patients with triple-negative breast cancer unselected for family history, recruited through 12 studies, identified 2.7% with a BRCA2 pathogenic variant.
(Refer to the BRCA1 pathology section of this summary for more information about this study.) A report from Iceland found less tubule formation, more nuclear pleomorphism, and higher mitotic rates in BRCA2-related tumors than in sporadic controls; however, a single BRCA2 founder pathogenic variant (999del5) accounts for nearly all hereditary breast cancer in this population, thus limiting the generalizability of this observation.
A large case series from North America and Europe described a greater proportion of BRCA2-associated tumors with continuous pushing margins (a histopathologic description of a pattern of invasion), fewer tubules and lower mitotic counts.
Other reports suggest that BRCA2-related tumors include an excess of lobular and tubulolobular histology.
In summary, histologic characteristics associated with BRCA2 pathogenic variants have been inconsistent.
Given that germline pathogenic variants in BRCA1 or BRCA2 lead to a very high probability of developing breast cancer, it was a natural assumption that these genes would also be involved in the development of the more common nonhereditary forms of the disease. Although somatic mutations in BRCA1 and BRCA2 are not common in sporadic breast cancer tumors, there is increasing evidence that hypermethylation of the gene promoter (BRCA1) and loss of heterozygosity (LOH) (BRCA2) are frequent events. In fact, many breast cancers have low levels of the BRCA1 mRNA, which may result from hypermethylation of the gene promoter.
Approximately 10% to 15% of sporadic breast cancers appear to have BRCA1 promoter hypermethylation, and even more have downregulation of BRCA1 by other mechanisms. Basal-type breast cancers (ER negative, PR negative, HER2 negative, and cytokeratin 5/6 positive) more commonly have BRCA1 dysregulation than other tumor types.
BRCA1-related tumor characteristics have also been associated with constitutional methylation of the BRCA1 promoter. In a study of 255 breast cancers diagnosed before age 40 years in women without germline BRCA1 pathogenic variants, methylation of BRCA1 in peripheral blood was observed in 31% of women whose tumors had multiple BRCA1-associated pathological characteristics (e.g., high mitotic index and growth pattern including multinucleated cells) compared with less than 4% methylation in controls.
(Refer to the BRCA1 pathology section for more information.) Although hypermethylation has not been reported for BRCA2 pathogenic variants, the BRCA2 locus on chromosome 13q is the target of frequent LOH in breast cancer.
Targeted therapies are being developed for tumors with loss of BRCA1 or BRCA2 protein expression.
Ovarian cancers in women with BRCA1 and BRCA2 pathogenic variants are more likely to be high-grade serous adenocarcinomas and are less likely to be mucinous or borderline tumors.
Fallopian tube cancer and peritoneal carcinomas are also part of the BRCA-associated disease spectrum.
Histopathologic examinations of fallopian tubes removed from women with a hereditary predisposition to ovarian cancer show dysplastic and hyperplastic lesions that suggest a premalignant phenotype.
Occult carcinomas have been reported in 2% to 11% of adnexa removed from carriers of BRCA pathogenic variants at the time of risk-reducing surgery.
Most of these occult lesions are seen in the fallopian tubes, which has led to the hypothesis that many BRCA-associated ovarian cancers may actually have originated in the fallopian tubes. Specifically, the distal segment of the fallopian tubes (containing the fimbriae) has been implicated as a common origin of the high-grade serous cancers seen in BRCA pathogenic variant carriers, based on the close proximity of the fimbriae to the ovarian surface, exposure of the fimbriae to the peritoneal cavity, and the broad surface area in the fimbriae.
Because of the multicentric origin of high-grade serous carcinomas from Müllerian-derived tissue, staging of ovarian, tubal, and peritoneal carcinomas is now considered collectively by the International Federation of Gynecology and Obstetrics. The term high-grade serous ovarian carcinoma may be used to represent high-grade pelvic serous carcinoma for consistency in language.
High-grade serous ovarian carcinomas have a higher incidence of somatic TP53 mutations.
DNA microarray technology suggests distinct molecular pathways of carcinogenesis between BRCA1, BRCA2, and sporadic ovarian cancer.
Furthermore, data suggest that BRCA-related ovarian cancers metastasize more frequently to the viscera, while sporadic ovarian cancers remain confined to the peritoneum.
Unlike high-grade serous carcinomas, low-grade serous ovarian cancers are less likely to be part of the BRCA1/BRCA2 spectrum.
Given that germline variants in BRCA1 or BRCA2 lead to a very high probability of developing ovarian cancer, it was a natural assumption that these genes would also be involved in the development of the more common nonhereditary forms of the disease. Although somatic mutations in BRCA1 and BRCA2 are not common in sporadic ovarian cancer tumors,
there is increasing evidence that hypermethylation of the gene promoter (BRCA1) and LOH (BRCA2) are frequent events. Loss of BRCA1 or BRCA2 protein expression is more common in ovarian cancer than in breast cancer,
and downregulation of BRCA1 is associated with enhanced sensitivity to cisplatin and improved survival in this disease.
Targeted therapies are being developed for tumors with loss of BRCA1 or BRCA2 protein expression.
Lynch syndrome is characterized by autosomal dominant inheritance of susceptibility to predominantly right-sided colon cancer, endometrial cancer, ovarian cancer, and other extracolonic cancers (including cancer of the renal pelvis, ureter, small bowel, and pancreas), multiple primary cancers, and a young age of onset of cancer.
The condition is caused by germline variants in the mismatch repair (MMR) genes, which are involved in repair of DNA mismatch variants.
The MLH1 and MSH2 genes are the most common susceptibility genes for Lynch syndrome, accounting for 80% to 90% of observed pathogenic variants,
followed by MSH6 and PMS2.
(Refer to the Lynch Syndrome section in the PDQ summary on Genetics of Colorectal Cancer for more information about this syndrome.)
After colorectal cancer, endometrial cancer is the second hallmark cancer of a family with Lynch syndrome. Even in the original Family G, described by Dr. Aldred Scott Warthin, numerous family members were noted to have extracolonic cancers including endometrial cancer. Although the first version of the Amsterdam criteria did not include endometrial cancer,
in 1999, the Amsterdam criteria were revised to include endometrial cancer as extracolonic tumors associated with Lynch syndrome to identify families at risk.
In addition, the Bethesda guidelines in 1997 (revised in 2004) did include endometrial and ovarian cancers as Lynch syndrome–related cancers to prompt tumor testing for Lynch syndrome.
The lifetime risk of ovarian carcinoma in females with Lynch syndrome is estimated to be as high as 12%, and the reported RR of ovarian cancer has ranged from 3.6 to 13, based on families ascertained from high-risk clinics with known or suspected Lynch syndrome.
There may be differences in ovarian cancer risk depending on the Lynch syndrome–associated pathogenic variant. In PMS2-associated Lynch syndrome, one study of 284 families was unable to identify an increased risk of ovarian cancer.
Another prospective registry of 3,119 Lynch syndrome–pathogenic variant carriers described the cumulative risk of ovarian cancer to range from 10% to 17% in MLH1, MSH2, and MSH6 carriers. In contrast, 0 of 67 women with a pathogenic variant in PMS2 developed ovarian cancer in 303 follow-up years.
Overall, there are too few cases of PMS2 pathogenic variant carriers to make definitive recommendations for ovarian cancer management. Characteristics of Lynch syndrome–associated ovarian cancers may include overrepresentation of the International Federation of Gynecology and Obstetrics stages I and II at diagnosis (reported as 81.5%), underrepresentation of serous subtypes (reported as 22.9%), and a better 10-year survival (reported as 80.6%) than reported both in population-based series and in carriers of BRCA pathogenic variants.
The issue of breast cancer risk in Lynch syndrome has been controversial. Retrospective studies have been inconsistent, but several have demonstrated microsatellite instability in a proportion of breast cancers from individuals with Lynch syndrome;
one of these studies evaluated breast cancer risk in individuals with Lynch syndrome and found that it is not elevated.
However, the largest prospective study to date of 446 unaffected carriers of pathogenic variants from the Colon Cancer Family Registry
who were followed for up to 10 years reported an elevated SIR of 3.95 for breast cancer (95% CI, 1.59–8.13; P = .001).
The same group subsequently analyzed data on 764 carriers of MMR gene pathogenic variants with a prior diagnosis of colorectal cancer. Results showed that the 10-year risk of breast cancer following colorectal cancer was 2% (95% CI, 1%–4%) and that the SIR was 1.76 (95% CI, 1.07–2.59).
A series from the United Kingdom composed of clinically referred Lynch syndrome kindreds, with efforts to correct for ascertainment, showed a twofold increased risk of breast cancer in 157 MLH1 carriers but not in carriers of other MMR variants.
Results from a meta-analysis of breast cancer risk in Lynch syndrome among 15 studies with molecular tumor testing results revealed that 62 of 122 breast cancers (51%; 95% CI, 42%–60%) in MMR pathogenic variant carriers were MMR-deficient. In addition, breast cancer risk estimates among a total of 21 studies showed an increased risk of twofold to 18-fold in eight studies that compared MMR variant carriers with noncarriers, while 13 studies did not observe statistical evidence for an association of breast cancer risk with Lynch syndrome.
A number of subsequent studies have suggested the presence of higher breast cancer risks than previously published,
although this has not been consistently observed.
Through a study of 325 Canadian families with Lynch syndrome, primarily encompassing MLH1 and MSH2 carriers, the lifetime cumulative risk for breast cancer among MSH2 carriers was reported to be 22%.
Similarly, breast cancer risks were elevated in a study of 423 women with Lynch syndrome, with substantially higher risks among those with MSH6 and PMS2 pathogenic variants, compared with MLH1 and MSH2 pathogenic variants.
In fact, breast cancer risk to age 60 years was 37.7% for PMS2, 31.1% for MSH6, 16.1% for MSH2, and 15.5% for MLH1. These findings are consistent with another study of 528 patients with Lynch syndrome–associated pathogenic variants (including MLH1, MSH2, MSH6, PMS2, and EPCAM) in which PMS2 and MSH6 variants were much more frequent among patients with only breast cancer, compared with those with only colorectal cancer (P = 2.3 x 10-5).
Additional data to support an association of MSH6 with breast cancer were provided through a study of over 10,000 cancer patients across the United States who had genetic testing.
Findings indicated that MSH6 was associated with breast cancer with an odds ratio (OR) of 2.59 (95% CI, 1.35–5.44). Taken together, these studies highlight how the risk profile among patients with Lynch syndrome is continuing to evolve as more individuals are tested through multigene panel testing, with representation of larger numbers of individuals with PMS2 and MSH6 pathogenic variants compared with prior studies. In the absence of definitive risk estimates, individuals with Lynch syndrome are screened for breast cancer on the basis of family history.
Refer to the Lynch Syndrome section of the Clinical Management of Other Hereditary Breast and/or Gynecologic Cancer Syndromes section of this summary for information about clinical management of Lynch syndrome.
Breast cancer is also a component of the rare LFS, in which germline variants of the TP53 gene on chromosome 17p have been documented. Located on chromosome 17p, TP53 encodes a 53kd nuclear phosphoprotein that binds DNA sequences and functions as a negative regulator of cell growth and proliferation in the setting of DNA damage. It is also an active component of programmed cell death.
Inactivation of the TP53 gene or disruption of the protein product is thought to allow the persistence of damaged DNA and the possible development of malignant cells.
Widely used clinical diagnostic criteria for LFS were originally developed by Chompret et al. in 2001 (called the Chompret Criteria)
and revised in 2009 based on additional emerging data.
LFS is characterized by premenopausal breast cancer in combination with childhood sarcoma, brain tumors, leukemia, and adrenocortical carcinoma.
Germline variants in TP53 are thought to account for fewer than 1% of breast cancer cases.
TP53-associated breast cancer is often HER2/neu-positive, in addition to being ER-positive, PR-positive, or both.
Evidence also exists that patients treated for a TP53-related tumor with chemotherapy or radiation therapy may be at risk of a treatment-related second malignancy.
The term Li-Fraumeni syndrome was used for the first time in 1982,
and the following criteria, which subsequently became the classical definition of the syndrome, were proposed by Li and Fraumeni in 1988 :
Subsequently in 2001, Chompret et al. systematically developed clinical criteria for recommending TP53 genetic testing, with the narrow LFS tumor spectrum defined as sarcoma, brain tumors, breast cancer, and adrenocortical carcinoma. The criteria were as follows:
These criteria were revised in 2009
based on additional emerging data
as follows:
*The 2009 Chompret criteria defined the LFS tumor spectrum as including the following cancers: soft tissue sarcoma, osteosarcoma, brain tumor, premenopausal breast cancer, adrenocortical carcinoma, leukemia, and lung bronchoalveolar cancer.
In 2015, Bougeard et al. revised the criteria based on data from 415 carriers of pathogenic variants, to include the presence of childhood anaplastic rhabdomyosarcoma and breast cancer before age 31 years as an indication for testing, similar to what is recommended for choroid plexus carcinoma and adrenocortical carcinoma. The criteria were revised as follows:
**The 2015 Chompret criteria defined the LFS tumor spectrum as including the following cancers: premenopausal breast cancer, soft tissue sarcoma, osteosarcoma, central nervous system (CNS) tumor, and adrenocortical carcinoma.
Germline TP53 pathogenic variants were identified in 17% (n = 91) of 525 samples submitted to City of Hope laboratories for clinical TP53 testing.
All families with a TP53 pathogenic variant had at least one family member with a sarcoma, breast cancer, brain cancer, or adrenocortical cancer (core cancers). In addition, all eight individuals with a choroid plexus tumor had a TP53 pathogenic variant, as did 14 of the 21 individuals with childhood adrenocortical cancer. In women aged 30 to 49 years who had breast cancer but no family history of other core cancers, no TP53 variants were found.
Subsequently, a large clinical series of patients from France who were tested primarily based on the 2009 version of the Chompret criteria included 415 carriers of pathogenic variants from 214 families.
In this study, 43% of carriers had multiple malignancies, and the mean age at first tumor onset was 24.9 years. The childhood tumor spectrum was characterized by osteosarcomas, adrenocortical carcinomas, CNS tumors, and soft tissue sarcomas (present in 23%–30% collectively), whereas the adult tumor spectrum primarily encompassed breast cancer (79% of females) and soft tissue sarcomas (27% of carriers). The TP53 pathogenic variant detection rate was 6% among females younger than 31 years with breast cancer and no additional features suggestive of LFS. Evaluation of genotype-phenotype correlations indicated a gradient of clinical severity, with a significantly lower mean age at onset among those with dominant-negative missense variants (21.3 years), compared with those with all types of loss-of-function variants (28.5 years) or genomic rearrangements (35.8 years). With the exception of adrenocortical carcinoma, affected children mostly harbored dominant-negative missense pathogenic variants. Among 127 female carriers of pathogenic variants with breast cancer, 31% developed CBC. Receptor status information was available for 40 tumors, which indicated 55% were HER2-positive, and 37% were triple-positive (i.e., ER-positive, PR-positive, and HER2-positive). There was an exceptionally high rate of multiple malignancies (43%) among carriers of pathogenic variants, of which 83% were metachronous. Treatment records were available for 64 carriers who received radiation therapy for treatment of their first tumor; of these, 19 (30%) developed 26 secondary tumors within a radiation field, with a latency of 2 to 26 years (mean, 10.7 y).
Similarly, results of 286 TP53 pathogenic variant–positive individuals in the National Cancer Institute’s LFS Study indicated a cumulative cancer incidence of almost 100% by age 70 years for both males and females.
They reported substantial variations by sex, age, and cancer type. Specifically, cumulative cancer incidence reached 50% by age 31 years in females and age 46 years in males, although male risks were higher in childhood and late adulthood. Cumulative cancer incidence by sex for the top four cancers is included in Table 8. Of those with one cancer, 49% developed at least one additional cancer after a median of 10 years. Age-specific risks for developing first and second cancers were comparable.
Cumulative Cancer Risk by Age 70 Years | ||
---|---|---|
Cancer Type | Females(%) | Males(%) |
Breast cancer | 54 | – |
Soft tissue sarcoma | 15 | 22 |
Brain cancer | 6 | 19 |
Osteosarcoma | 5 | 11 |
aAdapted from Mai et al. | ||
bOther cancers, such as adrenocortical carcinoma, leukemia, and lung bronchoalveolar cancer, have been considered part of the LFS cancer spectrum. |
With the increasing use of multigene (panel) tests, it is important to recognize that pathogenic variants in TP53 are unexpectedly being identified in individuals without a family history characteristic of LFS.
The clinical significance of finding an isolated TP53 pathogenic variant in an individual or family who does not meet the Chompret criteria is uncertain. Consequently, it remains important to interpret cancer risks and determine optimal management strategies for individuals who are unexpectedly found to have a germline TP53 pathogenic variant, while taking into account their personal and family histories.
One cohort study evaluated 116 individuals with a germline TP53 pathogenic variant yearly at the National Institutes of Health Clinical Center using multimodality screening with and without gadolinium. Baseline screening identified a cancer in eight patients (6.9%) with a false-positive rate of 34.5% for MRI (n = 40).
Screening for breast cancer with annual breast MRI is recommended;
additional screening for other cancers has been studied and is evolving.
Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome (BRRS) are part of a spectrum of conditions known collectively as PTEN hamartoma tumor syndromes. Approximately 85% of patients diagnosed with Cowden syndrome, and approximately 60% of patients with BRRS have an identifiable PTEN pathogenic variant.
In addition, PTEN pathogenic variants have been identified in patients with very diverse clinical phenotypes.
The term PTEN hamartoma tumor syndromes refers to any patient with a PTEN pathogenic variant, irrespective of clinical presentation.
PTEN functions as a dual-specificity phosphatase that removes phosphate groups from tyrosine, serine, and threonine. Pathogenic variants of PTEN are diverse, including nonsense, missense, frameshift, and splice-site variants. Approximately 40% of variants are found in exon 5, which encodes the phosphatase core motif, and several recurrent pathogenic variants have been observed.
Individuals with variants in the 5’ end or within the phosphatase core of PTEN tend to have more organ systems involved.
Operational criteria for the diagnosis of Cowden syndrome have been published and subsequently updated.
These included major, minor, and pathognomonic criteria consisting of certain mucocutaneous manifestations and adult-onset dysplastic gangliocytoma of the cerebellum (Lhermitte-Duclos disease). An updated set of criteria based on a systematic literature review has been suggested and is currently utilized in the National Comprehensive Cancer Network (NCCN) guidelines.
Contrary to previous criteria, the authors concluded that there was insufficient evidence for any features to be classified as pathognomonic. With increased utilization of genetic testing, especially the use of multigene panels, clinical criteria for Cowden syndrome will need to be reconciled with the phenotype of individuals with documented germline PTEN pathogenic variants who do not meet these criteria. Until then, whether Cowden syndrome and the other PTEN hamartoma tumor syndromes will be defined clinically or based on the results of genetic testing remains ambiguous. The American College of Medical Genetics and Genomics (ACMG) suggests that referral for genetics consultation be considered for individuals with a personal history of or a first-degree relative with 1) adult-onset Lhermitte-Duclos disease or 2) any three of the major or minor criteria that have been established for the diagnosis of Cowden syndrome.
Detailed recommendations, including diagnostic criteria for Cowden syndrome, can be found in the NCCN and ACMG guidelines.
Additionally, a predictive model that uses clinical criteria to estimate the probability of a PTEN pathogenic variant is available; a cost-effectiveness analysis suggests that germline PTEN testing is cost effective if the probability of a variant is greater than 10%.
Over a 10-year period, the International Cowden Consortium (ICC) prospectively recruited a consecutive series of adult and pediatric patients meeting relaxed ICC criteria for PTEN testing in the United States, Europe, and Asia.
Most individuals did not meet the clinical criteria for a diagnosis of Cowden syndrome or BRRS. Of the 3,399 individuals recruited and tested, 295 probands (8.8%) and an additional 73 family members were found to harbor germline PTEN pathogenic variants. In addition to breast, thyroid, and endometrial cancers, the authors concluded that on the basis of cancer risk, melanoma, kidney cancer, and colorectal cancers should be considered part of the cancer spectra arising from germline PTEN pathogenic variants. A second study of approximately 100 patients with a germline PTEN pathogenic variant confirmed these findings and suggested a cumulative cancer risk of 85% by age 70 years.
Although PTEN pathogenic variants, which are estimated to occur in 1 in 200,000 individuals,
account for a small fraction of hereditary breast cancer, the characterization of PTEN function will provide valuable insights into the signal pathway and the maintenance of normal cell physiology.
Lifetime breast cancer risk is estimated to be between 25% and 50% among women with Cowden syndrome.
Other studies have reported risks as high as 85%;
however, there are concerns regarding selection bias in these studies. As in other forms of hereditary breast cancer, onset is often at a young age and may be bilateral.
Lifetime risk of endometrial cancer is estimated to be between 19% and 28%, depending on the cohort studied, with an increased risk of premenopausal onset.
Because of the low prevalence of PTEN pathogenic variants in the population, the proportion of endometrial cancer attributable to Cowden syndrome is small. There are no data that link PTEN pathogenic variants to an increased risk of ovarian cancer. Skin manifestations include multiple trichilemmomas, oral fibromas and papillomas, and acral, palmar, and plantar keratoses. History or observation of the characteristic skin features raises a suspicion of Cowden syndrome. CNS manifestations include macrocephaly, developmental delay, and dysplastic gangliocytomas of the cerebellum.
(Refer to the PDQ summaries on Genetics of Colorectal Cancer and Genetics of Skin Cancer for more information about PTEN hamartoma tumor syndromes [including Cowden syndrome].)
The E-cadherin gene CDH1 was first described in 1998 in three Maori families with multiple cases of diffuse gastric cancer (DGC), leading to the designation of hereditary diffuse gastric cancer (HDGC). There have been multiple subsequent reports of an excess of lobular breast cancer in HDGC families.
CDH1 is located on chromosome 16q22.1 and encodes the E-cadherin protein, a calcium-dependent homophilic adhesion molecule that plays a key role in cellular adhesion, cell polarity, cell signaling, and maintenance of cellular differentiation and tissue morphology.
E-cadherin binds to various catenins to stabilize the cytoplasmic cell adhesion complex and to maintain the E-cadherin interaction with actin filament.
Loss of CDH1 can occur as a result of somatic mutations, LOH, or hypermethylation, and can result in dedifferentiation and invasiveness in human cancers.
Classic histopathologic findings in gastrectomy specimens include in situ signet ring cells and/or pagetoid spread of signet ring cells. Of all gastric cancers, 1% to 3% are attributed to inherited gastric cancer syndromes.
HDGC is an autosomal dominant syndrome associated with poorly differentiated invasive adenocarcinoma of the stomach presenting as linitis plastica. It is a highly penetrant and highly fatal syndrome, with a risk of clinical DGC ranging from 40% to 83%.
The risk of lobular breast cancer, which is characterized by small uniform cells that tend to invade in “single files,” is also increased in HDGC. Although invasive lobular breast cancer represents only 10% to 15% of all breast cancers, the lifetime risk of lobular breast cancer in carriers of CDH1 pathogenic variants ranges from 30% to 50%.
Guidelines for screening for CDH1 vary but include multiple cases of DGC in a family, early age of DGC, or lobular breast cancer in a family with DGC. Approximately 25% of families meeting these criteria are found to have a pathogenic variant in CDH1.
CDH1 pathogenic variants have been found in some families with lobular breast cancer but no gastric cancer.
The management of individuals with CDH1 pathogenic variants without a family history of gastric cancer is unclear.
PJS is an early-onset autosomal dominant disorder characterized by melanocytic macules on the lips, the perioral region, and buccal region; and multiple gastrointestinal polyps, both hamartomatous and adenomatous.
Germline pathogenic variants in the STK11 gene at chromosome 19p13.3 have been identified in the vast majority of PJS families.
The most common cancers in PJS are gastrointestinal. However, other organs are at increased risk of developing malignancies. For example, the cumulative risks have been estimated to be 32% to 54% for breast cancer
and 21% for ovarian cancer (mainly ovarian sex-cord tumors).
The risk for pancreatic cancer has been estimated to be more than 100-fold higher than that in the general population.
A systematic review found a lifetime cumulative cancer risk, all sites combined, of up to 93% in patients with PJS.
Table 9 shows the cumulative risk of these tumors.
Females with PJS are also predisposed to the development of cervical adenoma malignum, a rare and very aggressive adenocarcinoma of the cervix.
In addition, females with PJS commonly develop benign ovarian sex-cord tumors with annular tubules, whereas males with PJS are predisposed to development of Sertoli-cell testicular tumors;
although neither of these two tumor types is malignant, they can cause symptoms related to increased estrogen production.
Although the risk of malignancy appears to be exceedingly high in individuals with PJS based on the published literature, the possibility that selection and referral biases have resulted in overestimates of these risks should be considered.
Site | Age (y) | Cumulative Risk (%) |
---|---|---|
Any cancer | 60–70 | 37–93 |
GI cancer | 60–70 | 38–66 |
Gynecological cancer | 60–70 | 13–18 |
Per origin | ||
Stomach | 65 | 29 |
Small bowel | 65 | 13 |
Colorectum | 65 | 39 |
Pancreas | 65–70 | 11–36 |
Lung | 65–70 | 7–17 |
Breast | 60–70 | 32–54 |
Uterus | 65 | 9 |
Ovary | 65 | 21 |
Cervix | 65 | 10 |
Testes | 65 | 9 |
GI = gastrointestinal. | ||
aReprinted with permission from Macmillan Publishers Ltd: Gastroenterology , copyright 2010. | ||
bAll cumulative risks were increased compared with the general population (P < .05), with the exception of cervix and testes. | ||
cGI cancers include colorectal, small intestinal, gastric, esophageal, and pancreatic. | ||
dWesterman et al.: GI cancer does not include pancreatic cancer. | ||
eDid not include adenoma malignum of the cervix or Sertoli cell tumors of the testes. |
Per origin
PJS is caused by pathogenic variants in the STK11 (also called LKB1) tumor suppressor gene located on chromosome 19p13.
Unlike the adenomas seen in familial adenomatous polyposis, the polyps arising in PJS are hamartomas. Studies of the hamartomatous polyps and cancers of PJS show allelic imbalance (LOH) consistent with the two-hit hypothesis, demonstrating that STK11 is a tumor suppressor gene.
However, heterozygous STK11 knockout mice develop hamartomas without inactivation of the remaining wild-type allele, suggesting that haploinsufficiency may be sufficient for initial tumor development in PJS.
Subsequently, the cancers that develop in STK11 +/- mice do show LOH;
indeed, compound mutant mice heterozygous for pathogenic variants in STK11 +/- and homozygous for pathogenic variants in TP53 -/- have accelerated development of both hamartomas and cancers.
Germline variants of the STK11 gene represent a spectrum of nonsense, frameshift, and missense variants, and splice-site variants and large deletions.
Approximately 85% of variants are localized to regions of the kinase domain of the expressed protein. No strong genotype-phenotype correlations have been identified.
Up to 30% of variants are large deletions involving one or more exons of STK11, underscoring the importance of deletion analysis in suspected cases of PJS.
STK11 has been unequivocally demonstrated to cause PJS. Although earlier estimates using direct DNA sequencing showed a 50% pathogenic variant detection rate in STK11, studies adding techniques to detect large deletions have found pathogenic variants in up to 94% of individuals meeting clinical criteria for PJS.
Given the results of these studies, it is unlikely that other major genes cause PJS.
The high cumulative risk of cancers in PJS has led to the various screening recommendations summarized in the table of Published Recommendations for Diagnosis and Surveillance of Peutz-Jeghers Syndrome (PJS) in the PDQ summary on Genetics of Colorectal Cancer.
PALB2 (partner and localizer of BRCA2) interacts with the BRCA2 protein and plays a role in homologous recombination and double-stranded DNA repair. Similar to BRIP1 and BRCA2, biallelic pathogenic variants in PALB2 have also been shown to cause Fanconi anemia.
PALB2 pathogenic variants have been screened for in multiple small studies of familial and early-onset breast cancer in multiple populations.
Pathogenic variant prevalence has ranged from 0.4% to 3.9%. Similar to BRIP1 and CHEK2, there was incomplete segregation of PALB2 pathogenic variants in families with hereditary breast cancer.
Among 559 cases with CBC and 565 matched controls with unilateral breast cancer, pathogenic (truncating) PALB2 pathogenic variants were identified in 0.9% of cases and in none of the controls (RR, 5.3; 95% CI, 1.8–13.2).
Data based on 154 families with loss-of-function PALB2 variants suggest that this gene may be an important cause of hereditary breast cancer, with risks that overlap with BRCA2.
In this study, analysis of 362 family members from 154 families with PALB2 pathogenic variants indicated that the absolute risk of female breast cancer by age 70 years ranged from 33% (95% CI, 24%–44%) for those with no family history of breast cancer to 58% (95% CI, 50%–66%) for those with two or more FDRs with early-onset breast cancer. Furthermore, among 63 breast cancer cases in which HER2 status was known, 30% had triple-negative disease. An earlier Finnish study reported on a PALB2 founder pathogenic variant (c.1592delT) that confers a 40% risk of breast cancer to age 70 years
and is associated with a high incidence (54%) of triple-negative disease and lower survival.
Pathogenic variants have been observed in early-onset and familial breast cancer in many populations.
A large report of 1,824 patients with triple-negative breast cancer unselected for family history, recruited through 12 studies, identified 1.2% with a PALB2 pathogenic variant.
(Refer to the BRCA1 pathology section of this summary for more information about this study.)
In a later Polish study of more than 12,529 unselected women with breast cancer and 4,702 controls, PALB2 pathogenic variants were detected in 116 cases (0.93%; 95% CI, 0.76%–1.09%) and 10 controls (0.21%; 95% CI, 0.08%–0.34%), with an OR for breast cancer of 4.39 (95% CI, 2.30–8.37).
The study findings confirm a substantially elevated risk of breast cancer (24%–40%) among women with a PALB2 pathogenic variant up to age 75 years. The 5-year cumulative incidence of CBC was 10% among those with a PALB2 pathogenic variant, compared with 17% among those with a BRCA1 pathogenic variant and 3% among those without a variant in either gene. Furthermore, the 10-year survival for women with a PALB2 pathogenic variant and breast cancer was 48% (95% CI, 36.5%–63.2%), compared with 72.0% among those with a BRCA1 pathogenic variant and 74.7% among those without a variant in either gene. Among PALB2 carriers, breast tumors 2 cm or larger had substantially worse outcomes (32.4% 10-year survival), compared with tumors smaller than 2 cm (82.4% 10-year survival). Approximately one-third of those with a PALB2 pathogenic variant had triple-negative breast cancer, and the average age at breast cancer diagnosis was 53.3 years.
Male breast cancer has been observed in PALB2 pathogenic variant–positive breast cancer families.
In a study of 115 male breast cancer cases in which 18 men had BRCA2 pathogenic variants, an additional two men had either a pathogenic or predicted pathogenic PALB2 variant (accounting for about 10% of germline variants in the study and 1%–2% of the total sample).
The RR of breast cancer for male carriers of PALB2 pathogenic variants compared with that seen in the general population was estimated to be 8.30 (95% CI, 0.77–88.56; P = .08) in the study of 154 families.
After the identification of PALB2 pathogenic variants in pancreatic tumors and the detection of germline pathogenic variants in 3% of 96 familial pancreatic patients,
numerous studies have pointed to a role for PALB2 in pancreatic cancer. PALB2 pathogenic variants were detected in 3.7% of 81 familial pancreatic cancer families
and in 2.1% of 94 BRCA1/BRCA2 pathogenic variant–negative breast cancer patients who had either a personal or family history of pancreatic cancer.
Two relatively small studies—one of 77 BRCA1/BRCA2 pathogenic variant–negative probands with a personal or family history of pancreatic cancer, one-half of whom were of AJ descent, and another study of 29 Italian pancreatic cancer patients with a personal or family history of breast or ovarian cancer—failed to detect any PALB2 pathogenic variants.
A sixfold increase in pancreatic cancer was observed in the relatives of 33 BRCA1/BRCA2-negative, PALB2 pathogenic variant–positive breast cancer probands.
Overall, the observed prevalence of PALB2 pathogenic variants in familial breast cancer varied depending on ascertainment relative to personal and family history of pancreatic and ovarian cancers, but in all studies, the observed pathogenic variant rate was lower than 4%. Data suggest that the RR of breast cancer may overlap with that of BRCA2, particularly in those with a strong family history; thus, it remains important to refine cancer risk estimates in larger studies. Furthermore, the risk of other cancers (e.g., pancreatic) is poorly defined. Given the low PALB2 pathogenic variant prevalence in the population, additional data are needed to define best candidates for testing and appropriate management.
Until the 1990s, the diagnosis of genetically inherited breast and ovarian cancer syndromes was based on clinical manifestations and family history. Now that some of the genes involved in these syndromes have been identified, a few studies have attempted to estimate the spontaneous pathogenic variant rate (de novo pathogenic variant rate) in these populations. Interestingly, PJS, PTEN hamartoma syndromes, and LFS are all thought to have high rates of spontaneous pathogenic variants, in the 10% to 30% range, while estimates of de novo pathogenic variants in the BRCA genes are thought to be low, primarily on the basis of the few case reports published. Additionally, there has been only one case series of breast cancer patients who were tested for BRCA pathogenic variants in which a de novo variant was identified. Specifically, in this study of 193 patients with sporadic breast cancer, 17 pathogenic variants were detected, one of which was confirmed to be a de novo pathogenic variant. As such, the de novo pathogenic variant rate appears to be low and fall into the 5% or less range, based on the limited studies performed. Similarly, estimates of de novo pathogenic variants in the MMR genes associated with Lynch syndrome are thought to be low, in the 0.9% to 5% range. However, these estimates of spontaneous pathogenic variant rates in the BRCA genes and Lynch syndrome genes seem to overlap with the estimates of nonpaternity rates in various populations (0.6%–3.3%), making the de novo pathogenic variant rate for these genes relatively low.
BRCA1、BRCA2和PALB2的致病性突变以及在本摘要“高度外显乳腺癌和/或妇科癌症易感基因”部分讨论的其他罕见综合征相关基因,占乳腺癌家族风险的不到25%。
尽管进行了深入的遗传连锁研究,但似乎没有其他高外显率基因在剩余的多病例家族集群中占很大比例。
然而,目前已确定几种与乳腺癌和/或妇科癌症相关的中度外显基因。CHEK2和ATM等基因导致乳腺癌的终生风险为20%或更高;
同样地,RAD51C、RAD51D和BRIP1等基因导致卵巢癌的风险为5%至10%。
许多这样的基因现在包括在多基因组中,尽管这些发现的临床可行性仍然不确定并仍在研究中。
有大量流行病学研究文献描述了各种基因与乳腺癌风险之间的关联。许多此类研究受到明显的设计限制。或许因此,大多数报告的关联不会在后续研究中重复。本节不是对所有报告的关联的全面回顾。本节描述了编辑认为临床有效的关联,因为多项研究已对其进行了描述,或是有稳定的汇总分析的支持。然而,这些观察的临床效果仍不清楚,因为与这些突变相关的风险通常低于需要进行临床反应的阈值。
范可尼贫血(FA)是一种罕见的遗传性疾病,其特征是骨髓衰竭、恶性肿瘤风险增加和身体异常。迄今为止,已确定16种与FA相关的基因(如表10所示)。除由FANCB中的致病性突变引起外(X连锁隐性遗传),FA主要是常染色体隐性遗传。FANCA占致病性突变的60%至70%,FANCC约占14%,其余基因各占3%或更少。
高度风险基因 |
– BRCA1 (FANCS)a |
– BRCA2 (FANCD1)a |
– PALB2 (FANCN)b |
中度风险基因 |
– BRIP1 (FANCJ/BACH1) |
– FANCD2 |
– RAD51C (FANCO) |
风险不确定或无显著增加风险的基因 |
-FANCA |
– FANCB |
– FANCC |
– FANCE |
– FANCF |
– FANCG (XRCC9) |
– FANCI (KIAA1794) |
– FANCL |
– SLX4 (FANCP) |
– ERCC4 (FANCQ/XPF) |
a 有关BRCA1和BRCA2致病性突变携带者乳腺癌累积风险的信息,请参阅本摘要部分。 |
b 有关PALB2致病性突变携带者乳腺癌累积风险的信息,请参阅本摘要部分。 |
c 中度风险是指有统计学意义,增加了两倍或更低的评估风险。 |
进展性骨髓衰竭通常发生在头十年,患者血小板或白细胞通常会减少。到40至50岁时,骨髓衰竭的发生率为90%。血液系统恶性肿瘤(主要为急性髓性白血病)的发病率为10%至30%,非血液系统恶性肿瘤(实体瘤,主要生长在头颈部,皮肤,胃肠道和生殖道)的发病率为25%至30%。60%至75%的患者身体异常,包括身材矮小、皮肤色素沉着异常、放射线导致的缺陷(包括拇指畸形)、泌尿道、眼睛、耳朵、心脏、胃肠系统和中枢神经系统异常、性腺功能减退以及发育迟缓。
一些FA基因中的突变,主要为BRCA1和BRCA2,其次为PALB2,RAD51基因家族中的RAD51C和BRIP1等,其中,这些基因突变的杂合子可能使携带者易患乳腺癌。鉴于多基因(组)测试的广泛普及,尽管存在不确定的癌症风险,并且缺乏针对许多这样的基因的循证医学管理建议,但仍经常对许多FA基因进行基因检测。
FA基因致病性突变携带者的状态可能会影响生育决定,因为如果父母双方都是同一基因致病性突变的携带者,这些基因中的致病性突变可能导致儿童严重发病。可考虑进行伴侣测试。
BRIP1(也称为BACH1)编码一种螺旋酶,该螺旋酶与BRCA1 C端(BRCT)结构域相互作用。该基因还参与BRCA1依赖性DNA修复和细胞周期检查点功能。BRIP1中的双等位基因致病性突变是导致FA的原因之一,
与BRCA2中双等位基因致病性突变很像。BRIP1失活突变与乳腺癌风险增加相关。在一项研究中,来自BRCA1/BRCA2致病性突变阴性家族的3000多个体被检测出BRIP1突变。1212例乳腺癌患者中有9例发现了致病性突变,而2081例对照者中只有2例发现了致病性突变(P=.003)。乳腺癌的相对危险度(RR)估计为2.0(95%置信区间[CI],1.2-3.2;P=.012)。值得注意的是,在携带BRIP1致病性突变的家族和多例乳腺癌患者中,致病性突变与乳腺癌存在不完全分离,与低度外显等位基因一致,同与CHEK2一致相似。
在一项有3236例卵巢癌患者的病例对照研究中,BRIP1致病性突变更常与卵巢癌风险相关(RR,11.2;95%CI,3.2-34.1)。
CHEK2基因与DNA损伤修复反应通路有关。根据大量研究,CHEK2 1100delC多态性位点似乎是一个罕见的,中度外显的癌症易感等位基因。
一项欧洲研究显示,该致病性突变在对照组占比1.2%,在BRCA1/BRCA2阴性的家族性乳腺癌中占比4.2%,在未经筛选的女性乳腺癌病理中占比1.4%。
在1479名年龄小于50岁的荷兰女性中,发现3.7%的荷兰女性携带CHEK2 1100delc致病性突变。
在其他欧洲国家和美国(致病性突变似乎不太常见)的研究中,包括一项大型前瞻性研究,
在家族性乳腺癌或卵巢癌病例中检测到的CHEK2致病性突变的频率在0%至11%之间;
总的来说,这些研究发现女性患乳腺癌的风险增加了1.5到3倍。
然而,对10项病例对照研究中的近20000名受试者进行的多中心联合分析和再分析证实,致病性突变携带者的乳腺癌发病率显著增加了2.3倍。
随后一项对25项病例对照研究中的29154例患者和37064名对照者进行的汇总分析发现CHEK2 1100DELC杂合子与乳腺癌风险之间存在显著相关性(优势比[OR],2.75;95%CI,2.25–3.36)。未筛选、家族性和早发乳腺癌亚组的OR和CI分别为2.33(1.79-3.05)、3.72(2.61-5.31)和2.78(2.28-3.39)。然而,研究的局限性包括未经亚组分析便合并人群、混合使用基于人群和医院的对照者,以及基于未经调整的估计而得出结果(因为病例和对照只在少数共同因素上匹配);因此,结果应在上述局限的背景下进行解释。
在一系列男性乳腺癌患者中,CHEK2 1100delC突变的确定频率明显高于对照组,表明该突变也与男性乳腺癌风险的增加有关。
两项研究表明,因双侧乳腺癌而确定的先证者家族中与CHEK2 1100delC 致病性突变相关的风险更高。
此外,对1100delC致病性突变携带者的汇总分析显示有乳腺癌家族史的妇女到70岁的乳腺癌风险为42%。
与此类似的是,波兰的一项研究报告称, 根据乳腺癌家族史情况,CHEK2截断致病性突变的乳腺癌风险如下:无家族史:20%;一名二级亲属患乳腺癌:28%;一名一级亲属患乳腺癌:34%;一级和二级亲属患乳腺癌:44%。
此外,荷兰的一项研究表明,与杂合子相比,CHEK2 1100delC突变的纯合子女性患乳腺癌的风险增加了两倍以上。
尽管关于与CHEK2致病性突变相关的乳腺癌以外的癌症存在相互矛盾的报道,但这可能取决于所研究的突变(即错义与截断)或群体类型,并且目前不具有临床效用。
CHEK2突变导致乳腺癌发病的影响程度可能取决于所研究的人群,波兰的突变携带者的患病率可能更高。
波兰的CHEK2突变携带者可能更容易患雌激素受体(ER)阳性乳腺癌。
目前,由于突变患病率低和缺乏临床管理指南,CHEK突变的临床适用性仍然不确定。
荷兰的一项针对86,975人的大型研究报告称,CHEK2 1100delC致病性突变携带者罹患除乳腺癌和结肠癌以外的其它癌症的风险在增加,
尽管需要更多研究以进一步改善这些风险。
(有关更多信息,请参阅PDQ摘要“大肠癌遗传学”部分。)
共济失调毛细血管扩张症(AT)是一种常染色体隐性遗传疾病,其特征是神经系统恶化、毛细血管扩张、免疫缺陷状态和对电离辐射过敏。据估计,总人口的1%可能是ATM突变的杂合子携带者。
该基因中的300多个突变已获鉴定,其中大多数是截断突变。
已显示ATM蛋白在细胞周期控制中起作用。
在体外,AT缺陷细胞对电离辐射和放射模拟药物敏感,遭受辐射后缺乏细胞周期调控特性。
没有足够的证据需要对单个ATM致病突突变(杂合子)携带者进行放射治疗提供建议。
最初的研究是在乳腺癌患者中寻找过量的ATM致病性突变,其结果矛盾,这可能是由于研究设计和突变测试策略所致。
然而,两项大型流行病学研究表明,女性杂合子携带者患乳腺癌的风险在统计意义上增加,其估计RR约为2.0。
一项汇总分析表明,到50岁时乳腺癌风险为6.02%,到80岁时乳腺癌风险为32.83%。
鉴于这些风险,可以考虑增加筛查和其它基于家族史和年龄的建议。
一些研究表明,ATM与卵巢癌之间存在关联,
但是,目前尚无证据表明对风险管理或疾病特征有影响。
国际研究者乳腺癌协会联合会(BCAC)调查了先前研究中发现的单核苷酸多态性(SNP),这些单核苷酸多态性可能与15,000-20,000例病例和15,000-20,000例对照的过度乳腺癌风险相关。两种SNP,CASP8 D302H和TGFB1 L10P均与浸润性乳腺癌相关,其RR分别为0.88(95%CI,0.84-0.92)和1.08(95%CI,1.04-1.11)。
RAD51和RAD51相关基因家族(也称为RAD51旁系同源物)被认为编码通过同源重组并与众多其它DNA修复蛋白(包括BRCA1和BRCA2)相互作用而参与DNA损伤修复的蛋白。 RAD51蛋白在DNA损伤反应的单链退火中起核心作用。 RAD51增长到断裂位点和重组DNA修复依赖于RAD51旁系同源物,只是它们精确的细胞功能尚不清楚。
这些基因的突变被认为会导致DNS损伤反应时RAD51转化灶形成的丢失。
RAD51C是五个与RAD51相关的基因之一,据报道,其与FA类疾病以及家族性乳腺癌和卵巢癌都有关联。 然而,文献的发现相互矛盾。 在对480个以BRCA1和BRCA2致病性突变为阴性的乳腺癌和卵巢癌为特征的德国家庭的研究中,发现RAD51C中有6个单等位基因突变(频率为1.3%)。
另一项研究筛选了286例乳腺癌和/或卵巢癌的BRCA1/BRCA2阴性患者,并且发现RAD51C-G153D中可能存在一种致病突变。
仅在澳大利亚、英国、芬兰和西班牙的非BRCA1/BRCA2卵巢癌和乳腺癌/卵巢癌家族中以及未筛选的卵巢癌病例中,也已报道了RAD51C的致病突变,在这些人中的发生频率范围为0%至3%。
在先前测试过的206种高危犹太女性(包括79名Ashkenazi血统)的犹太常见病原体样本中,检测到两个先前描述且可能致病的错义突变。
另外四项研究无法确定RAD51C基因与遗传性乳腺癌或卵巢癌之间的关联。
除了RAD51C致病突变的携带者外,还有其它RAD51旁系同源物,包括RAD51B、RAD51D、RAD51L1、XRCC2和XRCC3,它们可能与乳腺癌和/或卵巢癌的风险有关,
但是,这些发现的临床意义尚不清楚。在一项针对3,429名卵巢癌患者的病例对照研究中,与对照组(0.11%,P <.001)相比,RAD51C和RAD51D致病突变在卵巢癌病例中更为普遍(0.82%)。
除种系突变之外,还假设RAD51的不同多态性降低了修复DNA缺陷的能力,从而增加了家族性乳腺癌发生的可能。 BRCA1/BRCA2修饰子研究者协会(CIMBA)汇总了来自8,512个BRCA1和BRCA2致病性突变携带者的数据,并发现在RAD51 135G→C SNP中携带BRCA2携带者和CC纯合子的女性罹患乳腺癌的风险增加(危险比,1.17; 95%CI,0.91-1.51)。
多项汇总分析研究了RAD51 135G→C多态性与乳腺癌风险之间的关系。这些汇总分析报告的研究存在重大重叠,所涵盖人群的特征存在显著差异,并且其研究结果存在显著的方法学限制。
对涉及13241例病例和13203例未知BRCA1/BRCA2状态的9项流行病学研究的汇总分析发现:与GG或GC基因型女性相比,携带CC基因型的女性患乳腺癌的风险更高(OR,1.35;95% CI,1.04–1.74)。对14个病例对照研究的汇总分析涉及12,183例病例和10,183例对照,证实只有已知的BRCA2携带者才会增加患病风险(OR,4.92; 95%CI,1.10–21.83)。
另一项对12项研究的汇总分析仅包括对已知BRCA阴性病例的研究,未发现RAD51 135G→C与乳腺癌之间存在关联。
总之,在这些矛盾的数据中,有大量证据表明RAD51C中的种系突变与乳腺癌和卵巢癌之间存在适度的联系。 也有证据表明:在具有纯合CC基因型的女性(尤其是BRCA2携带者)中,RAD51 135G→C基因多态性与乳腺癌之间存在关联。 根据RAD51在维持基因组稳定性中的已知作用,这些关联是合理的。
在三个芬兰乳腺癌家族中发现了brca1相互作用基因Abraxas的致病变异,没有对照组。
尚不清楚这一发现在该人群之外的意义。
通过对高危波兰裔和魁北克裔法裔加拿大家庭的全外显子组测序,发现RECQL基因在两种人群中都有多个罕见的截断型突变。
(有关全外显子组测序的更多信息,请参阅《癌症遗传学概述》 PDQ摘要的“临床测序”部分。)在同一人群中,还在随后的两个验证阶段,从高风险家族以及突变频率高于对照组的其它乳腺癌病例中鉴定该基因中的截断型突变,来自白俄罗斯和德国的病例对照研究调查了最常见的致病突变,即c.1667_1667 + 3delA GTA,发现其与ER阳性乳腺癌有关。仅这项研究中的OR为1.23(95%CI,0.44–3.47; P = .69),但在波兰的一项研究的汇总分析中,OR为2.51(95%CI,1.13-5.57,P = .02)。
尽管研究结果表明,截断型RECQL致病性突变与乳腺癌风险增加有关,但确切的风险程度仍不确定,因此需要进一步研究来确定临床用途。此外,这一发现在这两个人群之外的意义尚不清楚。
SMARCA4编码BRG1,是SWI/SNF染色质重塑复合体的催化亚基,在染色质获得调控基因表达中发挥重要作用。
高钙血症型卵巢小细胞癌(SCCOHT)是一种罕见的侵袭性肿瘤,发病年龄早(40岁之前),而且预后差。
家族聚集有时存在。SCCOHT肿瘤可以是单侧的或双侧的,其组织学特征是有丝分裂活跃的染色小细胞的存在。
已提出包括外科手术、化疗和放疗在内的多模式方法来治疗SCCOHT。
考虑到60%的病例发生高钙血症的副肿瘤现象,跟踪钙水平可用于监测疾病进程。通过广泛的鉴别诊断,包括生殖细胞肿瘤、性索间质肿瘤和未分化癌,SCCOHT被世界卫生组织分类为“杂类肿瘤”,但最近被测序为恶性横纹肌瘤。
通过外显子组测序,大多数SCCOHT病例被发现缺乏功能性SMARCA4/BRG1;实际上,SMARCA4中的致病突变可能是导致SCCOHT的唯一突变。
尽管文献中只有大约300例病例,但三个独立的研究组显示SCCOHT与种系致病突变和SMARCA4基因的体细胞突变有关。在一项针对12名患有SCCOHT的年轻女性的研究中,通过对成对肿瘤和正常样本的测序,在每个病例中发现了失活的双等位基因SMARCA4致病突变。
在其他278个被测序的基因中,只有4个额外的非复发体细胞基因被鉴定出来。免疫组化显示,9例检测病例中有7例SMARCA4蛋白表达缺失,这与肿瘤抑制基因功能一致。在另一项针对12名患者的研究中,下一代测序也发现SMARCA4是唯一的常见突变基因,大多数突变预计会导致蛋白质的截断。
第三项研究包括三个家族,其中通过Sanger测序确认的全外显子组测序在26例病例中,有24例识别出至少一个种系致病突变或体细胞突变。
总的来说,在43种SCCOHT肿瘤中,有38种(88%)显示SMARCA4表达缺失,而在139种其他卵巢肿瘤中,只有1种(0.7%)显示SMARCA4表达缺失。
由于该肿瘤罕见,SMARCA4的外显率尚不清楚。目前还没有关于如何处理的共识,但是SMARCA4已经在目前应用于基因检测的更大的多基因面板上,并且已经为致病性突变携带者提供了降低风险的手术。
Pathogenic variants in BRCA1, BRCA2, PALB2, and the genes involved in other rare syndromes discussed in the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section of this summary account for less than 25% of the familial risk of breast cancer.
Despite intensive genetic linkage studies, there do not appear to be other high-penetrance genes that account for a significant fraction of the remaining multiple-case familial clusters.
However, several moderate-penetrance genes associated with breast and/or gynecologic cancers have been identified. Genes such as CHEK2 and ATM are associated with a 20% or higher lifetime risk of breast cancer;
similarly, genes such as RAD51C, RAD51D, and BRIP1 are associated with a 5% to 10% risk of ovarian cancer.
Many of these genes are now included on multigene panels, although the clinical actionability of these findings remains uncertain and under investigation.
There is a very large literature of genetic epidemiology studies describing associations between various loci and breast cancer risk. Many of these studies suffer from significant design limitations. Perhaps as a consequence, most reported associations do not replicate in follow-up studies. This section is not a comprehensive review of all reported associations. This section describes associations that are believed by the editors to be clinically valid, in that they have been described in several studies or are supported by robust meta-analyses. The clinical utility of these observations remains unclear, however, as the risks associated with these variations usually fall below a threshold that would justify a clinical response.
Fanconi anemia (FA) is a rare, inherited condition characterized by bone marrow failure, increased risk of malignancy, and physical abnormalities. To date, 16 FA-related genes, including , have been identified (as outlined in Table 10). FA is mainly an autosomal recessive condition, except when caused by pathogenic variants in FANCB, which is X-linked recessive. FANCA accounts for 60% to 70% of pathogenic variants, FANCC accounts for approximately 14%, and the remaining genes each account for 3% or fewer.
High-Risk Genes |
– BRCA1 (FANCS)a |
– BRCA2 (FANCD1)a |
– PALB2 (FANCN)b |
Moderate-Risk Genes |
– BRIP1 (FANCJ/BACH1) |
– FANCD2 |
– RAD51C (FANCO) |
Genes With Uncertain or No Significantly Increased Risk |
-FANCA |
– FANCB |
– FANCC |
– FANCE |
– FANCF |
– FANCG (XRCC9) |
– FANCI (KIAA1794) |
– FANCL |
– SLX4 (FANCP) |
– ERCC4 (FANCQ/XPF) |
aRefer to the section of this summary for information about the cumulative risk of breast cancer in carriers of BRCA1 and BRCA2 pathogenic variants. |
bRefer to the section of this summary for information about the cumulative risk of breast cancer in carriers of PALB2 pathogenic variants. |
cModerate risk is defined as a statistically significant, twofold or lower increased risk estimate. |
Progressive bone marrow failure typically occurs in the first decade, with patients often presenting with thrombocytopenia or leucopenia. The incidence of bone marrow failure is 90% by age 40 to 50 years. The incidence is 10% to 30% for hematologic malignancies (primarily acute myeloid leukemia) and 25% to 30% for nonhematologic malignancies (solid tumors, particularly of the head and neck, skin, gastrointestinal [GI] tract, and genital tract). Physical abnormalities, including short stature, abnormal skin pigmentation, radial ray defects (including malformation of the thumbs), abnormalities of the urinary tract, eyes, ears, heart, GI system, and central nervous system, hypogonadism, and developmental delay are present in 60% to 75% of affected individuals.
Variants in some of the FA genes, most notably BRCA1 and BRCA2, but also PALB2, RAD51C (in the RAD51 family of genes), and BRIP1, among others, may predispose to breast cancer in heterozygotes. Given the widespread availability of multigene (panel) tests, genetic testing of many of the FA genes is frequently performed despite uncertain cancer risks and the lack of available evidence-based medical management recommendations for many of these genes.
FA gene pathogenic variant carrier status can have implications for reproductive decision making because pathogenic variants in these genes can lead to serious childhood onset of disease if both parents are carriers of pathogenic variants in the same gene. Partner testing may be considered.
BRIP1 (also known as BACH1) encodes a helicase that interacts with the BRCA1 C-terminal (BRCT) domain. This gene also has a role in BRCA1-dependent DNA repair and cell cycle checkpoint function. Biallelic pathogenic variants in BRIP1 are a cause of FA,
much like such pathogenic variants in BRCA2. Inactivating variants of BRIP1 are associated with an increased risk of breast cancer. In one study, more than 3,000 individuals from BRCA1/BRCA2 pathogenic variant–negative families were examined for BRIP1 variants. Pathogenic variants were identified in 9 of 1,212 individuals with breast cancer but in only 2 of 2,081 controls (P = .003). The relative risk (RR) of breast cancer was estimated to be 2.0 (95% confidence interval [CI], 1.2–3.2; P = .012). Of note, in families with BRIP1 pathogenic variants and multiple cases of breast cancer, there was incomplete segregation of the pathogenic variant with breast cancer, consistent with a low-penetrance allele and similar to that seen with CHEK2.
In a case-control study of 3,236 women with ovarian cancer, BRIP1 pathogenic variants were more frequently associated with ovarian cancer risk (RR, 11.2; 95% CI, 3.2–34.1).
CHEK2 is a gene involved in the DNA damage repair response pathway. Based on numerous studies, a polymorphism, 1100delC, appears to be a rare, moderate-penetrance cancer susceptibility allele.
One study identified the pathogenic variant in 1.2% of the European controls, 4.2% of the European BRCA1/BRCA2-negative familial breast cancer cases, and 1.4% of unselected female breast cancer cases.
In a group of 1,479 Dutch women younger than 50 years with invasive breast cancer, 3.7% were found to have the CHEK2 1100delC pathogenic variant.
In additional European and U.S. (where the pathogenic variant appears to be slightly less common) studies, including a large prospective study,
the frequency of CHEK2 pathogenic variants detected in familial breast or ovarian cancer cases has ranged from 0%
to 11%; overall, these studies have found an approximately 1.5-fold to 3-fold increased risk of female breast cancer.
A multicenter combined analysis and reanalysis of nearly 20,000 subjects from ten case-control studies, however, has verified a significant 2.3-fold excess of breast cancer among carriers of pathogenic variants.
A subsequent meta-analysis based on 29,154 cases and 37,064 controls from 25 case-control studies found a significant association between CHEK2 1100delC heterozygotes and breast cancer risk (odds ratio [OR], 2.75; 95% CI, 2.25–3.36). The ORs and CIs in unselected, familial, and early-onset breast cancer subgroups were 2.33 (1.79–3.05), 3.72 (2.61–5.31), and 2.78 (2.28–3.39), respectively. However, study limitations included pooling of populations without subgroup analysis, using a mix of population-based and hospital-based controls, and basing results on unadjusted estimates (as cases and controls were matched on only a few common factors); therefore, results should be interpreted in the context of these limitations.
In a series of male breast cancer patients, the CHEK2 1100delC variant was significantly more frequently identified than in controls, suggesting that this variant is also associated with an increased risk of male breast cancer.
Two studies have suggested that the risk associated with a CHEK2 1100delC pathogenic variant was stronger in the families of probands ascertained because of bilateral breast cancer.
Furthermore, a meta-analysis of carriers of 1100delC pathogenic variants estimated the risk of breast cancer to be 42% by age 70 years in women with a family history of breast cancer.
Similarly, a Polish study reported that CHEK2 truncating pathogenic variants confer breast cancer risks based on a family history of breast cancer as follows: no family history: 20%; one second-degree relative: 28%; one first-degree relative: 34%; and both first- and second-degree relatives: 44%.
Moreover, a Dutch study suggested that female homozygotes for the CHEK2 1100delC variant have a greater-than-twofold increased breast cancer risk compared with heterozygotes.
Although there have been conflicting reports regarding cancers other than breast cancer associated with CHEK2 pathogenic variants, this may be dependent on variant type (i.e., missense vs. truncating) or population studied and is not currently of clinical utility.
The contribution of CHEK2 variants to breast cancer may depend on the population studied, with a potentially higher variant prevalence in Poland.
Carriers of CHEK2 variants in Poland may be more susceptible to estrogen receptor (ER)–positive breast cancer.
Currently, the clinical applicability of CHEK variants remains uncertain because of low variant prevalence and lack of guidelines for clinical management.
A large Dutch study of 86,975 individuals reported an increased risk of cancers other than breast and colon for carriers of the CHEK2 1100delC pathogenic variant,
although additional studies are needed to further refine these risks.
(Refer to the section in the PDQ summary on Genetics of Colorectal Cancer for more information.)
Ataxia telangiectasia (AT) is an autosomal recessive disorder characterized by neurologic deterioration, telangiectasias, immunodeficiency states, and hypersensitivity to ionizing radiation. It is estimated that 1% of the general population may be heterozygote carriers of ATM variants.
More than 300 variants in the gene have been identified, most of which are truncating variants.
ATM proteins have been shown to play a role in cell cycle control.
In vitro, AT-deficient cells are sensitive to ionizing radiation and radiomimetic drugs, and lack cell cycle regulatory properties after exposure to radiation.
There is insufficient evidence to recommend against radiation therapy in carriers of a single ATM pathogenic variant (heterozygotes).
Initial studies searching for an excess of ATM pathogenic variants among breast cancer patients provided conflicting results, perhaps due to study design and variant testing strategies.
However, two large epidemiologic studies have demonstrated a statistically increased risk of breast cancer among female heterozygote carriers, with an estimated RR of approximately 2.0.
A meta-analysis modeled the risk of breast cancer to be 6.02% by age 50 years and 32.83% by age 80 years.
Given these risks, increased screening and other recommendations based on family history and age may be considered.
Some studies have shown an association between ATM and ovarian cancer,
although, at this time, there is no evidence to suggest an impact on risk management or disease characteristics.
The Breast Cancer Association Consortium (BCAC), an international group of investigators, investigated single nucleotide polymorphisms (SNPs) identified in previous studies as possibly associated with excess breast cancer risk in 15,000 to 20,000 cases and 15,000 to 20,000 controls. Two SNPs, CASP8 D302H and TGFB1 L10P, were associated with invasive breast cancer with RRs of 0.88 (95% CI, 0.84–0.92) and 1.08 (95% CI, 1.04–1.11), respectively.
RAD51 and the family of RAD51-related genes, also known as RAD51 paralogs, are thought to encode proteins that are involved in DNA damage repair through homologous recombination and interaction with numerous other DNA repair proteins, including BRCA1 and BRCA2. RAD51 protein plays a central role in single-strand annealing in the DNA damage response. RAD51 recruitment to break sites and recombinational DNA repair depend on the RAD51 paralogs, although their precise cellular functions are poorly characterized.
Variants in these genes are thought to result in loss of RAD51 focus formation in response to DNA damage.
One of five RAD51-related genes, RAD51C has been reported to be linked to both FA-like disorders and familial breast and ovarian cancers. The literature, however, has produced contradictory findings. In a study of 480 German families characterized by breast and ovarian cancers who were negative for BRCA1 and BRCA2 pathogenic variants, six monoallelic variants in RAD51C were found (frequency of 1.3%).
Another study screened 286 BRCA1/BRCA2-negative patients with breast cancer and/or ovarian cancer and found one likely pathogenic variant in RAD51C-G153D.
RAD51C pathogenic variants have also been reported in Australian, British, Finnish, and Spanish non-BRCA1/BRCA2 ovarian cancer–only and breast/ovarian cancer families, and in unselected ovarian cancer cases, with frequencies ranging from 0% to 3% in these populations.
In a sample of 206 high-risk Jewish women (including 79 of Ashkenazi origin) previously tested for the common Jewish pathogenic variants, two previously described and possibly pathogenic missense variants were detected.
Four additional studies were unable to confirm an association between the RAD51C gene and hereditary breast cancer or ovarian cancer.
In addition to carriers of RAD51C pathogenic variants, there are other RAD51 paralogs, including RAD51B, RAD51D, RAD51L1, XRCC2, and XRCC3, that may be associated with breast and/or ovarian cancer risk,
although the clinical significance of these findings is unknown. In a case-control study of 3,429 ovarian cancer patients, RAD51C and RAD51D pathogenic variants were more commonly found in ovarian cancer cases (0.82%) than in controls (0.11%, P < .001).
In addition to germline variants, different polymorphisms of RAD51 have been hypothesized to have reduced capacity to repair DNA defects, resulting in increased susceptibility to familial breast cancer. The Consortium of Investigators of Modifiers of BRCA1/BRCA2 (CIMBA) pooled data from 8,512 carriers of BRCA1 and BRCA2 pathogenic variants and found evidence of an increased risk of breast cancer among women who were BRCA2 carriers and who were homozygous for CC at the RAD51 135G→C SNP (hazard ratio, 1.17; 95% CI, 0.91–1.51).
Several meta-analyses have investigated the association between the RAD51 135G→C polymorphism and breast cancer risk. There is significant overlap in the studies reported in these meta-analyses, significant variability in the characteristics of the populations included, and significant methodologic limitations to their findings.
A meta-analysis of nine epidemiologic studies involving 13,241 cases and 13,203 controls of unknown BRCA1/BRCA2 status found that women carrying the CC genotype had an increased risk of breast cancer compared with women with the GG or GC genotype (OR, 1.35; 95% CI, 1.04–1.74). A meta-analysis of 14 case-control studies involving 12,183 cases and 10,183 controls confirmed an increased risk only for women who were known BRCA2 carriers (OR, 4.92; 95% CI, 1.10–21.83).
Another meta-analysis of 12 studies included only studies of known BRCA-negative cases and found no association between RAD51 135G→C and breast cancer.
In summary, among this conflicting data is substantial evidence for a modest association between germline variants in RAD51C and breast cancer and ovarian cancer. There is also evidence of an association between polymorphisms in RAD51 135G→C among women with homozygous CC genotypes and breast cancer, particularly among BRCA2 carriers. These associations are plausible given the known role of RAD51 in the maintenance of genomic stability.
Pathogenic variants in the BRCA1-interacting gene Abraxas were found in three Finnish breast cancer families and no controls.
The significance of this finding outside of this population is not yet known.
Through full exome sequencing among high-risk Polish and Quebec-based French Canadian families, the RECQL gene was discovered to harbor multiple rare truncating variants in both populations.
(Refer to the Clinical Sequencing section in the Cancer Genetics Overview PDQ summary for more information about whole-exome sequencing.) In the same populations, truncating variants in this gene were also identified in two subsequent validation phases among additional breast cancer patients from high-risk families, and among additional breast cancer cases in which the variant frequency was higher than that observed among controls. A case-control study from Belarus and Germany looked at the most common pathogenic variant, c.1667_1667+3delA GTA, and found it to be linked to ER-positive breast cancer. The OR in this study alone was 1.23 (95% CI, 0.44–3.47; P = .69), but in a meta-analysis with a Polish study, the OR was 2.51 (95% CI, 1.13–5.57, P = .02).
Although study results suggest that truncating germline RECQL pathogenic variants are associated with an increased risk of breast cancer, the exact magnitude of risk remains uncertain, and future studies are needed to determine clinical usefulness. Furthermore, the significance of this finding outside of these two populations is not yet known.
SMARCA4 encodes BRG1 and is a catalytic subunit of the SWI/SNF chromatin remodeling complex, which plays a major role in rendering chromatin accessible to regulation of gene expression.
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare, aggressive tumor that has an early age at onset (before age 40 y) and a poor prognosis.
Familial clustering is sometimes present. SCCOHT tumors may be unilateral or bilateral and have been characterized histologically by the presence of small hyperchromatic cells with brisk mitotic activity.
A multimodality approach including surgery, chemotherapy, and radiation therapy has been suggested for the treatment of SCCOHT.
Given the paraneoplastic phenomenon of hypercalcemia in 60% of cases, tracking calcium levels is useful in monitoring the course of disease. With a wide range of differential diagnoses including germ cell tumors, sex cord–stromal tumors, and undifferentiated carcinomas, SCCOHT remains classified by the World Health Organization as a "miscellaneous tumor" but more recently has been sequenced to be a malignant rhabdoid tumor.
Through exome sequencing, most cases of SCCOHT have been found to lack functional SMARCA4/BRG1; in fact, pathogenic variants in SMARCA4 may be the sole variants responsible for SCCOHT.
Despite only approximately 300 cases in the literature, three separate research groups showed SCCOHT to be associated with germline pathogenic variants and somatic mutations in the SMARCA4 gene. In one study of 12 young women with SCCOHT, sequencing of paired tumor and normal samples identified inactivating biallelic SMARCA4 pathogenic variants in each case.
Only four additional nonrecurrent somatic genes were identified in any of the other 278 genes sequenced. Immunohistochemistry demonstrated loss of SMARCA4 protein expression in seven of nine tested cases, consistent with a tumor-suppressor gene function. In a second study of another 12 patients, next-generation sequencing also identified SMARCA4 as the only recurrently variant gene, with the majority of variants predicted to result in a truncated protein.
A third study included three families in whom whole-exome sequencing with Sanger sequencing confirmation identified at least one germline pathogenic variant or somatic mutation in 24 of 26 cases.
Overall, 38 of 43 (88%) of SCCOHT tumors showed loss of SMARCA4 expression, in comparison to only 1 of 139 (0.7%) other ovarian tumor types.
Because of the rarity of this tumor, the penetrance of SMARCA4 is unknown. There is currently no consensus for management, yet SMARCA4 is on the larger multigene panels currently available for genetic testing, and risk-reducing surgery has been offered to pathogenic variant carriers.
乳腺癌和妇科癌症多基因易感性的潜在多态性被认为是低外显率的,这一术语通常用于与最小至中度风险相关的序列突变。这与通常与更严重的表型相关的高外显率突变或等位基因形成对比,如导致家族中常染色体显性遗传模式的BRCA1/BRCA2致病性突变以及BRIP1、CHEK2和RAD51C等中度外显率突变。(更多信息请参考本摘要中的高外显率乳腺癌和/或妇科癌症易感基因以及与乳腺癌和/或妇科癌症相关的中外显率基因。)由于这些类型的序列变异(也称为低外显率基因、等位基因、突变和多态性)在一般人群中较为常见,估计它们对癌症风险的总体贡献要远远大于BRCA1和BRCA2致病性突变在人群中的可归因风险。例如,根据隔离分析估计,所有乳腺癌的一半发生在被认为最易感的12%的人群中
BRCA1/BRCA2中没有已知的低外显率突变。最初被认为是低外显率等位基因的BRCA2中的N372H突变尚未在大型综合分析中得到验证。
已尝试两种策略来鉴定导致乳腺癌易感性的低外显率多态性:候选基因和全基因组搜索。两者都涉及流行病学病例对照研究设计。候选基因方法包括根据基因的已知或推测的生物学功能与癌变或器官生理的相关性来选择基因,然后搜索或测试已知的遗传突变与癌症风险的关联。该策略依赖于不完善和不完整的生物学知识,尽管有一些已证实的关联(如下所述),但相对令人失望。
候选基因方法已大部分被全基因组关联研究(GWAS)所取代,在全基因组关联研究中,在基因组中选择并测试了非常多的单核苷酸多态性(SNP)(约100万到500万),大多数情况下不考虑它们可能的生物学功能,而是更均匀地捕获整个基因组中的所有遗传突变。
与评估候选基因和/或等位基因相反,GWAS涉及比较分布在整个基因组中的大量遗传突变。当前的范例使用几组多达500万个SNP,它们被选为基于HapMap和1000基因组工程来捕获基因组中的大部分常见突变。
通过比较大量病例和对照之间的等位基因频率(通常每个病例与对照之间为1,000或更多),并验证受试者复制组的有希望的信号,已获得非常强大的关联统计信号。
即使在生物学上相关的突变也不在测试的SNP组内,在染色体上物理上彼此接近的多个SNP之间的强相关性(连接不平衡)也支持“扫描”基因组中的易感性等位基因。尽管这种SNP之间的相关性使人们无需分析每个SNP即可查询大部分基因组,但当获得经过验证的关联时,通常并不明确许多相关突变中哪一个是有因果关系的。
全基因组搜索在识别许多复杂疾病的常见、低外显率易感性等位基因方面显示了巨大的希望,包括乳腺癌。
第一项研究涉及对家族性乳腺癌病例的初步扫描,然后在两个散发性乳腺癌的大样本集中进行复制,最后一个是乳腺癌协会联盟收集的20,000多个病例和20,000个对照的集合。
已确定五个不同的基因组区域,它们位于FGFR2、TNRC9、MAP3K1和LSP1基因内或附近,或位于染色体8q区域。 8q区域和其它区域可能包含与风险相关的多个独立基因座。随后的全基因组研究已复制这些基因座,并确定了其它基因座。
通过对散发性乳腺癌的大规模研究确定的许多SNP,似乎与雌激素受体(ER)阳性疾病的关联更密切。
但是,有些主要或仅与其它亚型相关,包括三阴性疾病。
可以从已发布的GWAS中获得SNP-性状关联的在线目录,以用于研究性状/疾病相关SNP的基因组特征。
尽管这些位点的遗传突变与乳腺癌和卵巢癌风险之间存在关联的统计证据不计其数,但生物学相关的突变及其导致风险增加的机制尚不清楚,将需要进一步的遗传和功能验征。此外,这些基因座与非常适度的风险相关(通常,优势比[OR] <1.5),并且可能会发现更多的风险突变。尚未发现SNP与乳腺癌的流行病学危险因素之间存在相互作用。
此外,理论模型表明:常见的中度风险SNP在改善个性化风险评估模型方面的潜力有限。
这些模型使用接受者操作特性(ROC)曲线分析来计算曲线下的面积(AUC),作为判别准确性的度量。随后的研究使用ROC曲线分析来检查SNP在超过5500个乳腺癌病例和近6,000个对照的临床数据集中的效用,与传统风险因子模型和同时使用标准风险因子和10个先前确定的SNP的模型相比,该模型具有传统风险因子。添加遗传信息适度地将AUC从58%更改为61.8%,这一结果被认为在临床上并不重要。尽管如此,当包括遗传信息时,仍有32.5%的患者罹患乳腺癌的风险增加了五分之一,而20.4%的患者则将罹患乳腺癌的风险降低了五分之一。这些信息是否具有临床用途,尚不清楚。
关于卵巢癌风险的可用数据更为有限。已经对卵巢癌进行了三个GWAS的分期分析,涉及10,000多个病例和13,000个对照。
与其它GWAS一样,OR中等,通常约为1.2或更低,但暗示了许多与卵巢癌具有合理生物学联系的基因,如BABAM1,其蛋白质与BRCA1形成复合物并可能对其进行调节,而TIRAPR会编码(ADP-核糖)聚合酶,它是BRCA1/BRCA2缺陷型细胞中可能很重要的分子。
最近使用总分评估了许多遗传突变的集体影响。 2015年,欧洲血统女性使用乳腺癌协会联盟(BCAC)的41项研究估算了包含所有已知乳腺癌风险遗传突变或SNP的多基因风险评分(PRS),其中包括33,000多个乳腺癌病例和33,000个对照。
早期估计乳腺癌PRS的尝试包括77个SNP,使最低和最高1%的女性到80岁时终生罹患乳腺癌的风险分别为3.5%和29%。
此后,PRS纳入了额外的遗传变异,并检查了其他乳腺癌相关的结果,包括肿瘤和病理特征、检测方式和对侧乳腺癌(CBC)。
2019年,开发了迄今为止具有最高辨识力的PRS,并在可用的最大GWAS数据集中进行了前瞻性验证(BCAC中进行了79项研究,英国生物库中有190,000多名女性),该研究结合了313种遗传突变的信息并针对ER阳性和ER阴性乳腺癌进行优化。
与中位女性相比,PRS313中最高1%的女性患乳腺癌、患ER阳性乳腺癌和患ER阴性乳腺癌的风险分别为4.04倍、4.37倍和2.78倍。
在80岁以下,PRS313最低和最高1%的女性中,ER阳性乳腺癌的终生绝对风险在2%到31%之间,而ER阴性乳腺癌的绝对风险在0.55%到4%之间。
与原发性乳腺癌的发生有关的常见基因组突变,也与CBC的发生有关。
与最低四分位数相比,在PRS最高四分位数中的女性患CBC的风险增加1.6倍。
此外,还对乳腺癌和卵巢癌的PRS进行了评估,这些女性是BRCA1和BRCA2致病性突变的携带者,并发现可预测这些女性的癌症风险,从而支持了以下假设:一般人群与突变携带者之间共享癌症风险的多基因组成。
在整个乳腺癌PRS中,ER阴性疾病的PRS与BRCA1突变携带者与乳腺癌风险之间的关联最强,而BRCA2突变携带者与乳腺癌整体PRS关联最强。在PRS的第10和第90个百分位数处,BRCA1突变携带者的累积终生风险分别为发生乳腺癌的56%和75%。卵巢癌PRS与BRCA1和BRCA2突变携带者的风险密切相关。对于BRCA2突变携带者,到80岁时,PRS的第10个百分点和第90个百分点的卵巢癌风险分别为6%和19%。作者指出,将PRS纳入风险预测模型可以更好地为该人群的癌症风险管理决策提供依据。
多项研究审查了通过纳入有关已知易感性SNP的信息,并报告了纳入PRS后提高的辨别准确性,可以改善临床乳腺癌风险预测模型的程度。
例如,在一项结合PRS77与临床模型的研究中,用于预测50岁之前的乳腺癌的AUC改善了20%以上。
目前正在进行包括WISDOM和MyPeBs在内的临床试验,以研究PRS在制定筛查决策和了解结果方面的潜在临床效用。
由于PRS只是在欧洲血统的人群中得到了广泛的开发和验证,因此尚不清楚这些PRS在非欧洲人群中的效用和预测准确性。
一项大型研究检查了已知的生殖和生活方式风险因素是否与PRSs相互作用以增加乳腺癌的风险,并且未发现与既定风险因素的相互作用。
除了GWAS询问常见的遗传突变外,基于测序的研究还涉及全基因组或全外显子测序
也鉴定与乳腺癌有关的基因,如XRCC2,这是一种罕见的中等外显率的乳腺癌易感基因 。
(有关完整外显子组测序的更多信息,请参阅关于“癌症遗传学概述”的PDQ摘要中的“临床测序”部分。)
Polymorphisms underlying polygenic susceptibility to breast and gynecologic cancers are considered low penetrance, a term often applied to sequence variants associated with a minimal to moderate risk. This is in contrast to high-penetrance variants or alleles that are typically associated with more severe phenotypes, for example BRCA1/BRCA2 pathogenic variants leading to an autosomal dominant inheritance pattern in a family, and moderate-penetrance variants such as BRIP1, CHEK2, and RAD51C. (Refer to the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes and the Moderate-Penetrance Genes Associated With Breast and/or Gynecologic Cancer sections of this summary for more information.) Because these types of sequence variants (also called low-penetrance genes, alleles, variants, and polymorphisms) are relatively common in the general population, their overall contribution to cancer risk is estimated to be much greater than the attributable risk in the population from pathogenic variants in BRCA1 and BRCA2. For example, it is estimated by segregation analysis that half of all breast cancer occurs in 12% of the population that is deemed most susceptible.
There are no known low-penetrance variants in BRCA1/BRCA2. The N372H variation in BRCA2, initially thought to be a low-penetrance allele, was not verified in a large combined analysis.
Two strategies have attempted to identify low-penetrance polymorphisms leading to breast cancer susceptibility: candidate gene and genome-wide searches. Both involve the epidemiologic case-control study design. The candidate gene approach involves selecting genes based on their known or presumed biological function, relevance to carcinogenesis or organ physiology, and then searching for or testing known genetic variants for an association with cancer risk. This strategy relies on imperfect and incomplete biological knowledge, and, despite some confirmed associations (described below), has been relatively disappointing.
The candidate gene approach has largely been replaced by genome-wide association studies (GWAS) in which a very large number of single nucleotide polymorphisms (SNPs) (approximately 1 million to 5 million) are chosen within the genome and tested, mostly without regard to their possible biological function, but instead to more uniformly capture all genetic variation throughout the genome.
In contrast to assessing candidate genes and/or alleles, GWAS involve comparing a very large set of genetic variants spread throughout the genome. The current paradigm uses sets of as many as 5 million SNPs that are chosen to capture a large portion of common variation within the genome based on the HapMap and the 1000 Genomes Project.
By comparing allele frequencies between a large number of cases and controls, typically 1,000 or more of each, and validating promising signals in replication sets of subjects, very robust statistical signals of association have been obtained.
The strong correlation between many SNPs that are physically close to each other on the chromosome (linkage disequilibrium) allows one to “scan” the genome for susceptibility alleles even if the biologically relevant variant is not within the tested set of SNPs. Although this between-SNP correlation allows one to interrogate the majority of the genome without having to assay every SNP, when a validated association is obtained, it is not usually obvious which of the many correlated variants is causal.
Genome-wide searches are showing great promise in identifying common, low-penetrance susceptibility alleles for many complex diseases, including breast cancer.
The first study involved an initial scan in familial breast cancer cases followed by replication in two large sample sets of sporadic breast cancer, the final being a collection of over 20,000 cases and 20,000 controls from the Breast Cancer Association Consortium.
Five distinct genomic regions were identified that were within or near the FGFR2, TNRC9, MAP3K1, and LSP1 genes or at the chromosome 8q region. The 8q region and others may harbor multiple independent loci associated with risk. Subsequent genome-wide studies have replicated these loci and identified additional ones.
Numerous SNPs identified through large studies of sporadic breast cancer appear to be associated more strongly with estrogen receptor (ER)–positive disease;
however, some are associated primarily or exclusively with other subtypes, including triple-negative disease.
An online catalog is available of SNP-trait associations from published GWAS for use in investigating genomic characteristics of trait/disease-associated SNPs.
Although the statistical evidence for an association between genetic variation at these loci and breast and ovarian cancer risk is overwhelming, the biologically relevant variants and the mechanism by which they lead to increased risk are unknown and will require further genetic and functional characterization. Additionally, these loci are associated with very modest risk (typically, an odds ratio [OR] <1.5), with more risk variants likely to be identified. No interaction between the SNPs and epidemiologic risk factors for breast cancer have been identified.
Furthermore, theoretical models have suggested that common moderate-risk SNPs have limited potential to improve models for individualized risk assessment.
These models used receiver operating characteristic (ROC) curve analysis to calculate the area under the curve (AUC) as a measure of discriminatory accuracy. A subsequent study used ROC curve analysis to examine the utility of SNPs in a clinical dataset of more than 5,500 breast cancer cases and nearly 6,000 controls, using a model with traditional risk factors compared with a model using both standard risk factors and ten previously identified SNPs. The addition of genetic information modestly changed the AUC from 58% to 61.8%, a result that was not felt to be clinically significant. Despite this, 32.5% of patients were in a higher quintile of breast cancer risk when genetic information was included, and 20.4% were in a lower quintile of risk. Whether such information has clinical utility is unclear.
More limited data are available regarding ovarian cancer risk. Three GWAS involving staged analysis of more than 10,000 cases and 13,000 controls have been carried out for ovarian cancer.
As in other GWAS, the ORs are modest, generally about 1.2 or weaker but implicate a number of genes with plausible biological ties to ovarian cancer, such as BABAM1, whose protein complexes with and may regulate BRCA1, and TIRAPR, which codes for a poly (ADP-ribose) polymerase, molecules that may be important in BRCA1/BRCA2-deficient cells.
The collective influence of many genetic variants has more recently been evaluated using an aggregate score. In 2015, a polygenic risk score (PRS) comprising all of the known breast cancer risk genetic variants or SNPs was estimated in women of European ancestry using 41 studies in the Breast Cancer Association Consortium (BCAC), including more than 33,000 breast cancer cases and 33,000 controls.
This early attempt at estimating a PRS for breast cancer included 77 SNPs, which collectively conferred lifetime risks of developing breast cancer by age 80 years of 3.5% and 29% for women in the lowest and highest 1% of the PRS, respectively.
Since then, PRSs incorporating additional genetic variants and examining other breast cancer–related outcomes including tumor and pathological characteristics, mode of detection, and contralateral breast cancer (CBC) have been estimated.
In 2019, the PRS with the highest discriminatory ability to date was developed and prospectively validated in the largest GWAS datasets available (79 studies in BCAC and more than 190,000 women in the U.K. Biobank), which incorporates information on 313 genetic variants and is optimized for ER-positive and ER-negative breast cancer.
Compared with women in the middle quintile, those in the highest 1% of PRS313 had 4.04-, 4.37-, and 2.78-fold risks of developing breast cancer overall, ER-positive disease, and ER-negative disease, respectively.
Lifetime absolute risk of breast cancer by age 80 years for women in the lowest and highest 1% of PRS313 ranged from 2% to 31% for ER-positive breast cancer, while for ER-negative disease, the absolute risks ranged from 0.55% to 4%.
Common genomic variants associated with the development of a first primary breast cancer are also associated with the development of CBC.
Women in the highest quartile of the PRS had a 1.6-fold increased risk of developing CBC compared with the lowest quartile.
Moreover, PRSs of breast and ovarian cancers have been assessed in women who are carriers of BRCA1 and BRCA2 pathogenic variants, and have been found to be predictive of cancer risk in these women, supporting the hypothesis of a shared polygenic component of cancer risk between the general population and variant carriers.
The PRS for ER-negative disease had the strongest association with breast cancer risk in BRCA1 variant carriers, while the strongest association in BRCA2 variant carriers was seen for the overall breast cancer PRS. BRCA1 variant carriers had cumulative lifetime risks of 56% and 75% of developing breast cancer at the 10th and 90th percentile of the PRS, respectively. The ovarian cancer PRS was strongly associated with risk for both BRCA1 and BRCA2 variant carriers. For BRCA2 variant carriers, the ovarian cancer risk was 6% and 19% by age 80 years for those at the 10th and 90th percentile of PRS, respectively. The authors noted that the incorporation of the PRS into risk prediction models may better inform decisions on cancer risk management for this population.
Several studies have examined the extent to which clinical breast cancer risk prediction models can be improved by including information on known susceptibility SNPs, and reporting improved discriminatory accuracy after inclusion of the PRS.
For example, in a study combining PRS77 with clinical models, the AUC for predicting breast cancer before age 50 years improved by more than 20%.
Clinical trials, including WISDOM and MyPeBs, are in progress to study the potential clinical utility of the PRS for making screening decisions and understanding outcomes.
Because PRSs have been largely developed and validated in populations of European ancestry, the utility and prediction accuracy of these PRSs in non-European populations is unknown.
A large study examined whether known reproductive and lifestyle risk factors interact with PRSs to increase breast cancer risk and did not find a multiplicative interaction with established risk factors.
In addition to GWAS interrogating common genetic variants, sequencing-based studies involving whole-genome or whole-exome sequencing
are also identifying genes associated with breast cancer, such as XRCC2, a rare, moderate-penetrance breast cancer susceptibility gene.
(Refer to the Clinical Sequencing section in the PDQ summary on Cancer Genetics Overview for more information about whole-exome sequencing.)
关于减少对乳腺癌或卵巢癌有遗传易感性人群的风险的干预措施的结果,可获得的数据越来越多。
如本摘要其它部分所述,不确定性通常是与家族史阳性或基因检测相关。在这种情况下,个人偏好可能是患者决定降低风险策略的一个重要因素。
有关信息,请参阅PDQ摘要“乳腺癌筛查”部分。在一般人群中进行筛查,并向PDQ摘要汇总“癌症遗传学研究的证据水平”,以获取与筛查和预防相关的证据水平的信息。
在一般人群中,乳房自我检查(BSE)价值的证据有限。在中国上海进行的一项BSE随机研究报告了初步结果。
5年后,与对照组相比,BSE组的乳腺癌死亡率没有下降,被诊断出的乳腺癌也没有明显的分期转变。(有关更多信息,请参阅PDQ摘要“乳腺癌筛查”部分。)
在乳腺癌风险增加的个体中,关于BSE的直接前瞻性证据很少。在加拿大国家乳腺癌筛查研究中,患有乳腺癌的一级亲属(FDRs)的女性的BSE能力得分在统计上显著高于无家族史的女性。在对转诊中心的251位高危女性进行的一项研究中,在距上次筛查不到一年的时间里,有5名乳腺癌患者是在上一次筛查后不到一年的时间里通过自我检查发现的(相比之下,有1名患者是通过临床医生检查发现的,11名患者是通过乳房x光检查发现的)。队列中的妇女接受自我检查指导,但并未说明是否由于计划的自我检查或偶然发现乳房肿块而发现了间隔癌。
在另一组BRCA1/BRCA2致病突变携带者中,9例乳腺癌中有4例在经正常乳房x线摄影检查后被诊断为可扪及的肿块,这进一步表明了自我检查的潜在价值。
癌症遗传学研究协会召集的一个工作组建议“从成年早期(如18至21岁)开始每月进行自我检查,以养成规律的习惯并熟悉乳房组织的正常特征。建议进行自我检查的教育和指导。”
关于临床乳房检查(CBE)的前瞻性数据很少。
癌症遗传学研究协会工作组总结说:“与自我检查一样,临床检查对患有遗传性早期乳腺癌风险的女性尤其重要。 他们建议BRCA1或BRCA2高风险致病突变的女性携带者从25岁至35岁开始接受年度或半年期临床检查。”
在一般人群中,有力的证据表明,定期对50至59岁的女性进行乳腺钼靶筛查可将乳腺癌死亡率降低25%-30%。 (有关更多信息,请参阅PDQ摘要“乳腺癌筛查”部分。)对于40至49岁开始进行乳腺钼靶筛查的女性,在开始筛查后15年,其乳腺癌死亡率降低了17%。
一项对 28000 多名女性进行的队列研究的观察数据表明,乳腺钼靶X线摄影检查对年轻女性的灵敏度较低。在该项研究中,对具有FDR伴乳腺癌年轻女性(30-49岁)的灵敏度最低。对于这些女性,乳腺钼靶X线摄影检查发现的乳腺癌中有 69% 是在首次钼靶X线摄影检查后的 13 个月内诊断出来的。相比之下,对 50 岁以下无家族史女性的灵敏度为 88% (P=0.08)。对于 50 岁及以上的女性,13 个月时的灵敏度为 93%,且不随家族史而变化。
初步数据表明,乳腺钼靶X线摄影检查对于 BRCA1 和 BRCA2 携带者的灵敏度低于非携带者。
随后的观察研究发现,乳腺钼靶X线摄影检查的阳性预测值 (PPV) 随着年龄的增长而增加,在老年女性和有乳腺癌家族史的女性中最高。
较高的 PPV可能与由于乳腺癌发病率增加、较高的灵敏度和/或较高的特异性有关。
一项研究发现,在 28 名女性中(其中 26 名女性携带 BRCA1 致病性突变,2 名女性携带 BRCA2 致病性突变),推进性边缘的出现与乳腺钼靶X线摄影检查的假阴性之间存在关联。髓质组织学特征的推进性边缘与缺乏纤维化反应有关。
此外,快速的肿瘤倍增时间可能导致肿瘤出现时间短。在一项研究中,BRCA1/BRCA2 携带者的平均肿瘤倍增时间为 45 天,而非携带者为 84 天。
另一项评估BRCA致病性突变携带者的钼靶下乳腺密度的研究发现,致病性突变状态与乳腺钼靶检查密度之间没有关联;然而,无论是携带者还是非携带者,乳房密度的增加都与乳腺癌风险的增加有关。
加拿大国家随机乳腺癌筛查研究-2对于普通人群中 50-59 岁的女性每年进行 CBE 加乳腺钼靶X线摄影检查与仅进行 CBE 检查的情况进行了比较。两组均接受了 BSE 的指导。
尽管钼靶X线摄影检查比 CBE 能检测到更小的原发性浸润性肿瘤、更多的浸润性癌和更多的导管原位癌 (DCIS),但 CBE 加乳腺钼靶X线摄影组与 CBE 单独组的乳腺癌死亡率几乎相同,且与其他乳腺癌筛查试验相比具有优势。平均随访 13 年(范围为 11.3-16.0年)后,累积乳腺癌死亡率为 1.02(95% 置信区间 [CI],0.78-1.33)。对这一结果的一个可能解释是对从事 CBE 的卫生专业人员进行了认真的培训与监督。
数字乳腺钼靶X线摄影是指使用数字检测器来查找并记录 x-射线图像。这项技术提高了对比度分辨率,
并已被提议作为提高乳腺钼靶X线摄影灵敏度的一种潜在策略。对6736 名 40 岁及以上女性进行数字乳腺摄影并与常规乳腺摄影的筛查对比发现,两者的乳腺癌检出率没有差异;
然而,数字乳腺钼靶X线摄影检查的召回较少。另一项研究(ACRIN-6652) 比较了 42760 名女性的数字乳腺摄影和平片乳腺摄影检查情况,两种技术的总体诊断准确率相似。
比较ROC曲线后表明,数字乳腺钼靶X线摄影对 50 岁以下女性、胸片显示致密的女性以及绝经前或围绝经期女性则更为准确。
在一项对 251 例携带 BRCA 致病性突变的前瞻性研究中,受试者均接受了关于筛查和降低风险手术的统一建议,在接受BRCA结果后的平均20.2个月,每年的乳腺钼靶X线摄影检查发现6名女性患有乳腺癌。
癌症遗传学研究协会专家组已建议 BRCA1 或 BRCA2 高风险致病性突变的女性携带者“每年进行一次乳腺钼靶X线摄影检查,起始年龄宜为 25 至 35 岁。可能的情况下,应在相同的地方进行乳腺钼靶X线摄影检查,并与之前的胸片进行比较。”
关于使用电离辐射工具与 CBE 或其他非电离辐射工具进行筛查的相对益处及风险的前瞻性研究中得到的数据,将大有裨益。
有一些观察结果引起了人们的担忧,即携带 BRCA 致病性突变的女性可能比未携带致病性突变的女性更容易患上因辐射诱发的乳腺癌。BRCA1 和 BRCA2 蛋白在 DNA 损伤修复的细胞机制中具有重要作用,包括参与修复辐射损伤的细胞机制。一些研究表明,BRCA 突变杂合的细胞具有中等的辐射灵敏度,但这并非一成不变,其会随着实验系统及终点的不同而有所变化。
三项研究未能找到令人信服的证据,证明 BRCA1 和 BRCA2 致病性突变携带者的电离辐射暴露与乳腺癌风险之间存在关联。
相比之下,两项大型国际研究发现,胸片x光或估算的诊断性辐射总暴露量导致乳腺癌风险增加。
一项国际大规模病例对照研究对 1,601 例致病性突变携带者进行了研究,结果表明,曾接受胸部X线检查的女性中,乳腺癌的患病风险增加(风险比[HR],1.54),其中 40 岁及以下、1949年后出生、仅在 20 岁之前接受过X线检查的女性患病风险最高。
该项研究中的一些受试者还被纳入了对来自三个欧洲中心对致病性突变携带者进行的更大规模、更全面的分析中。
在对来自英国、法国和荷兰 1993 例 BRCA1 和 BRCA2 致病性突变携带者的研究中,年龄特异性总诊断辐射照射量(如胸部x线、乳腺 x 射线摄影、荧光透视及计算机断层扫描)估计值来自自我检查的问卷。与从未接触过诊断辐射照射的女性相比,30 岁以前接触过的女性患病风险更高 (HR,1.90; 95% CI,1.20–3.00)。这种风险主要是由20 岁以下女性的非乳腺钼靶X线辐射暴露引起的 (HR,1.62; 95% CI,1.02–2.58)。随后,一项对 1844 例 BRCA1 携带者和 502 例BRCA2 携带者在入组时从未进行乳腺癌诊断的前瞻性研究(平均随访时间为 5.3 年)发现,先前的乳腺钼靶X线辐射暴露与乳腺癌风险之间无显著相关性。
30 岁以下女性的其他亚组分析表明,与乳腺癌风险无关。
随着磁共振成像 (MRI) 在 BRCA1 和 BRCA2 致病性突变携带者中的常规应用,乳腺钼靶X线筛查的任何潜在益处及其潜在风险都必须进行仔细权衡,特别是对年轻女性。
一项研究表明,在携带 BRCA1 和 BRCA2 致病性突变的患者中,最具成本效益的筛查策略可能是从 25 岁开始每年进行一次 MRI,从 30 岁开始交替进行 MRI 和数字乳腺钼靶X线摄影检查(因此,每项检查每年进行一次,但每 6 个月进行一次筛查)。
美国国家综合癌症网络 (NCCN) 目前建议在 25 岁至 29 岁之间每年进行乳腺 MRI 筛查(或考虑X线断层扫描的乳腺钼靶X线摄影检查,仅当 MRI 无法使用时)以及 30 至 75 岁之间每年进行乳腺钼靶X线摄影筛查(考虑断层合成摄影和乳腺 MRI 增强扫描)。
由于乳腺钼靶X线摄影对具有乳腺癌遗传风险的女性相对不灵敏,因此针对高患病风险的女性,包括 BRCA 致病性突变携带者,已经提出并研究了多种筛查方法。许多研究介绍了针对乳腺癌高患病风险女性进行乳腺 MRI 筛查的经验,包括对较大规模多机构试验的介绍。
尽管这些研究存在一些局限性,但他们一致证明乳腺 MRI 在检测遗传性乳腺癌方面比乳腺钼靶X线摄影或超声检查更为灵敏。欲了解六项大型研究的结果,请参见表11-乳腺癌遗传风险女性的 MRI 筛查研究总结。
这些项目中的大多数癌症都是筛查出来的,仅有 6% 的癌症出现在筛查间隔期。MRI 的灵敏度(由研究方法确定)在 71% 到100% 之间。在这些联合研究中,77% 的癌症是通过 MRI 确定的,42% 是通过乳腺钼靶X线摄影确定的。
与其他筛查方式相比,人们对 MRI 的低特异性表示了担忧。 在一项研究中,在进行初次 MRI 筛查后,16.5% 的患者被召回做进一步的评估,另有 7.6% 的患者被建议在 6 个月后接受一次短时间的随访检查。
在随后的几轮筛查中,这些比率显著下降,只有不到 10% 的受试者被召回进行更详细的 MRI,只有不到 3% 的受试者被建议进行短期随访。在另一项磁共振成像用于乳腺筛查 (MARIBS) 的研究中,每年进行额外评估的召回率为 10.7%。
第一项研究的良性活检率在第一轮为 11%,第二轮为 6.6%,第三轮为 4.7%。
在 MARIBS 研究中,总的手术活检率为每 1000 例筛查中有 9 例,尽管这可能会低估了,因为在 MARIBS 计算的分子中并未包括随访超声、组织芯活检和细针穿刺。
MRI 的 PPV 在各个系列中的计算方式有所不同,并有所波动,具体取决于是将所有异常检查项还是仅将导致活检的检查项计入分母中。通常,在大多数系列中,组织采样建议的 PPV(与进一步调查相反)在 50% 的范围内。
这些试验可以证明,在遗传性乳腺癌的检测中,MRI 优于乳腺钼靶X线摄影,参与这些试验(包括每年的 MRI 筛查)的女性患者筛查漏诊癌症的可能性较小。
然而,乳腺钼靶X线摄影可以识别出某些 MRI 无法识别的癌症,尤其是 DCIS。
关于降期,一项筛查研究表明,与两个非随机对照组的女性相比,具有遗传性乳腺癌患病风险的患者更有可能在肿瘤较小,未发生淋巴结转移时被诊断出来。
尽管 MRI 筛查比较灵敏,但一些参加 MRI 检查的女性仍会患上危及生命的乳腺癌。在一项前瞻性研究中,对 51 例 BRCA1致病性突变携带者和 41 例 BRCA2 致病性突变携带者每年进行乳腺钼靶X线摄影及 MRI 筛查(其中 80 例接受过降低风险的卵巢切除术),共检测到 11 例乳腺癌(9 例浸润性和 2 例DCIS)。通过 MRI 首次发现了 6 例癌症;通过乳腺钼靶X线摄影首次发现了 3 例;还有2例是间期癌。所有乳腺癌都发生在 BRCA1 致病性突变携带者中,这表明在卵巢切除术后短期内 BRCA1 相关乳腺癌的患病风险仍然很高。这些结果表明,在 BRCA1 和 BRCA2 致病性突变携带者中,监测和预防策略可能带来不同的结果。
一份结合三项大型研究(MARIBS、一项加拿大研究和一项荷兰MRI 筛查研究)结果显示,当 MRI 联合乳腺摄影检查时,在 BRCA2 致病性突变携带者中检测到的 80% 癌症是 DCIS,或小于 1cm 的浸润性癌。在 BRCA1 致病性突变的携带者中,49% 的癌症是 DCIS 或是小浸润性癌。此外,学者预测,对于 BRCA1 和 BRCA2 致病性突变携带者,增加 MRI检查可降低死亡率。该模型预测乳腺钼靶X线摄影使癌症死亡率降低了42%-47%,MRI 使癌症死亡率降低了 48%-61%,联合筛查使癌症死亡率降低了 50%-62%。
一项对 1997 年至 2006 年间接受 MRI 检查的 BRCA1 / BRCA2致病性突变携带者的另一项研究表明,97% 的癌症为 0 期或 I 期。
2015 年荷兰进行的一项病例对照研究进一步评估了 2308 例高风险患者,包括 706 例具有已知 BRCA 致病性突变的女性,这些女性均接受了乳腺钼靶X线摄影检查,并将其与增加了 MRI 检查的女性进行了比较。
在筛查的患者中,93 例患者被检测出患有 97 种癌症,33 例患者携带 BRCA1 致病性突变,18 例患者携带 BRCA2 致病性突变。在平均 9 年的随访中,MRI 筛查组无转移生存率有所提高(90% vs 77%),但是由于其数量很少,因此在 BRCA1 和 BRCA2 亚组中不具有统计学意义。在整个队列中,经 MRI 筛查的患者更有可能为淋巴结阴性且接受较少的化疗。美国癌症协会和 NCCN 建议每年对有乳腺癌遗传风险的女性进行 MRI 检查。
关于乳腺钼靶X线摄影和 MRI 检查时间的另一个问题是,两者应该同时进行还是以交替方式进行(每年进行一次检查时,每 6 个月进行一次筛查)。一项研究表明,在 BRCA1 和 BRCA2 致病性突变携带者中,最具成本效益的筛选策略可能是从 25 岁开始每年进行 MRI 检查,而从 30 岁开始进行 MRI 和数字乳腺钼靶X线摄影交替检查。
总之,证据均有力地支持乳腺 MRI 检查在乳腺癌监测中对 BRCA1/BRCA2 致病性突变携带者的不可或缺的作用。
系列 | Rijnsburger | Warner | MARIBS | Kuhl | Weinstein | Sardanelli | 总计 | |
---|---|---|---|---|---|---|---|---|
患者例数 N | 总计 | 2157 | 236 | 649 | 687 | 609 | 501 | 4839 |
BRCA1/BRCA2 携带者 | 594 | 236 | 120 | 65 | 44 | 330 | 1,389 | |
筛选集 N | 6253 | 457 | 1,881 | 1,679 | 1,592 | 11,862 | ||
癌症例数 N | 基线 | 22 | 13 | 20 | 10 | 0 | 0 | 65 |
后续 | 97 | 9 | 15 | 17 | 18 | 52 | 208 | |
浸润 | 78 | 16 | 29 | 8 | 11 | 44 | 186 | |
原位 | 19 | 9 | 6 | 9 | 7 | 8 | 58 | |
年发病率 | 10.4/1,000 | 19/1,000 | ||||||
在计划筛查时检测到 | 78 | 21 | 33 | 27 | 18 | 49 | 226 (83%) | |
每种方式检测到的例数-N | 钼靶X线摄像 | 31 | 8 | 14 | 9 | 7 | 25 | 94 (42%) |
MRI | 51 | 17 | 27 | 25 | 12 | 42 | 174 (77%) | |
超声 | 7 | 10 | 3 | 26 | 46 (41%) | |||
随访 | 平均 4.9 年 | 最少 1 年 | 2–7 年 | 平均 29.09 个月 | 2 年 | 3 年 | ||
a 基于接受筛查的前 1909 名女性的数据。 | ||||||||
b 包括仅患有浸润性癌的患者以及同时患有浸润性癌和原位癌的患者。 | ||||||||
c 其中仅有 75 例癌症是在同时接受乳腺钼靶X线摄影和 MRI 筛查的女性身上发现的。 | ||||||||
d 仅限于进行超声检查的研究。 |
原位
在一篇综述中讨论了,几项研究报告了乳腺钼靶X线摄像遗漏却通过超声检查发现的乳腺癌实例。
在一项初步研究中,超声作为乳腺钼靶X线摄像的辅助手段,根据家族史,对 149 名患病风险中度增加的女性进行了检查,根据超声检查结果,发现了一例癌症。还进行了 9 例其他良性病变的活检。一例是由于乳腺钼靶X线摄像与超声均提示异常,其余 8 例基于仅超声提示异常。
一项针对 2809 名具有致密乳腺组织的女性的大型研究 (ACRIN-6666) 表明,超声检查使乳腺癌筛查的检出率从单纯钼靶X线检查的7.6‰ 提高到钼靶X线摄影和超声联合检查的 11.8‰。
但是,超声筛查增加了假阳性率,与 MRI 结合使用的获益有限。在一项对 171 名女性(其中 92% 是 BRCA1 / BRCA2 致病性突变的携带者)同时进行乳腺钼靶X线摄像、MRI 和超声检查的多中心联合研究中,仅超声检查未发现癌症。
超声检查的不确定性包括筛查对死亡率的影响,假阳性结果的发生率及结果,以及有经验的乳腺超声检查医师。
证据级别:未指定
许多其他技术正在积极研究中,包括断层合成摄影、对比增强乳腺X线摄影、热成像和放射性核素扫描。在将这些技术纳入临床实践之前,还需要更多支持证据。
证据级别:未指定
降低风险的乳房切除术 (RRM) 被认为是乳腺癌高危患者的一种治疗选择。外科肿瘤学会认可具有 BRCA1/BRCA2 致病性突变或乳腺癌家族史的女性可以选择 RRM。
历史上,已经进行了全部或简单的乳房切除术,包括去除所有的乳腺组织,包括乳头和乳晕复合体 (NAC)。如果患者感兴趣,可以对手术切除部分同时进行重建。重建的方法包括组织扩张器和基于植入物的重建或自体修复,其中使用患者自身的组织用于再造乳房。可采用许多不同的组织来重建乳房,包括基于背阔肌、腹直肌或臀肌的皮瓣。也可以使用保留肌肉的技术,例如腹壁下动脉深穿支皮瓣,但需要先进的微血管技术。为了提高美容效果,已经采用了保留皮肤的技术,其中通过NAC 切除整个乳房,但保留了乳房的整个皮肤包膜。在进一步的改进中,已经开发了乳头保留技术,其中保留了所有的乳房皮肤和乳头,同时去除了下面的腺体组织。
由于尚无 RRM vs观察的随机、前瞻性试验,因此数据仅限于队列研究和病例对照研究。现有数据表明,RRM 确实可以降低高危患者的乳腺癌发病率,
但是总生存期 (OS) 与原发性乳腺癌发病率的总体风险更为紧密相关。几项研究分析了 RRM 对乳腺癌风险和死亡率的影响。在一项回顾性队列研究中,214 名女性因家族史提示常染色体显性遗传倾向而被认为有遗传风险,其中三名女性在双侧 RRM 后被诊断出患有乳腺癌,平均随访时间为 14 年。
由于预期会发生 37.4 例癌症,因此计算出的风险降低率为 92.0%(95% CI,76.6%-98.3%)。在一项随访亚组分析中,该队列研究的 214 名高危女性中,176 名接受了 BRCA1 和 BRCA2 致病性突变基因检测。在 18 名女性中发现了致病性突变,在平均随访 13.4 年后,没有一名女性患上乳腺癌。
在 RRM 后诊断为乳腺癌的三名女性中,有两人接受了检测,两人均未携带致病性突变。计算得出的致病性突变携带者的风险降低率为 89.5%-100.0% (95% CI, 41.4%-100.0%),这取决于对致病性突变携带者中预期癌症数量的假设,以及未经检测的乳腺癌切除术后患癌症女性的状况。该项回顾性队列研究的结果得到了对 76 例接受 RRM 且进行了平均 2.9 年监测的病原体携带者的前瞻性分析的支持。在这些女性中没有发现乳腺癌,而在接受定期监测的女性中发现了 8 例(RRM 后乳腺癌的 HR 为 0.00[95% CI,0.00-0.36])。
外科终点预防和观察研究组还评估了BRCA1/BRCA2致病性突变的携带者在预防性乳腺切除术(RRM)后乳腺癌风险降低的程度。将105例经双侧RRM致病性突变的携带者与378例未选择手术携带者的乳腺癌发病率进行比较。平均随访6.4年,双侧乳房切除术后乳腺癌风险降低了约90%。
理论模型亦被用来评估RRM在BRCA1和BRCA2致病性突变女性人群中的作用。假设风险降低在90%的范围内,一个模型表明,对于一组携带BRCA1或BRCA2致病性突变的30岁女性,RRM可延长平均预期寿命2.9-5.3年。
使用蒙特卡罗模型进行的计算机模拟生存分析,包括乳腺MRI、乳房钼靶检测、预防性乳腺切除术(RRM)和降低风险的输卵管卵巢切除术(RRSO),并分别评估每种干预措施对BRCA1和BRCA2致病性突变携带者的影响。
结果发现,最有效的策略是40岁时的RRSO和25岁时的RRM,其70岁时的生存率接近普通人群。然而,如果将乳房切除术推迟到40岁,或者用乳腺核磁共振和乳腺钼靶检查代替RRM,则对生存率几乎无明显影响。例如,40岁时采用核磁共振筛查代替RRM,与25岁时行RRM相比,生存率降低3%-5%。
与其他模型一样,许多假设都会造成不确定性;然而,这些研究为女性患者及其难以做决策的临床医生提供更多信息。
另一项针对高危女性的研究显示,RRM有70%的时间权衡价值,即参与者愿意牺牲30%的预期寿命来避免预防性乳腺切除术(RRM)。
本文还对RRM进行了成本效益分析研究。研究结果显示,与监测相比,可降低风险的手术(乳房切除术和卵巢切除术)在挽救生命的生存期方面有成本效益,但对生活质量无明显改善。
尽管这些数据值得关注,对公共政策决策有所帮助。但由于它们包含了无法完全测试的假设,因此不能个体化用于临床诊疗。
如果RRM能有效降低未受累女性的乳腺癌风险,那么对单侧乳腺癌女性患者有何影响?该问题经常在单侧乳腺癌和遗传风险的女性手术讨论决策时出现。本节内容就对侧降低风险的乳房切除术(CRRM)在拟行乳房切除术治疗患者中的作用进行讨论,保乳治疗不在讨论范围内。多项研究表明,单侧乳腺癌患者行CRRM的比例有升高趋势。
在评估单侧乳腺癌患者是否应行CRRM时,首要工作是确定对侧乳腺癌(CBC)的发病风险。
在普通人群中,目前每年乳腺癌治疗后CBC发病率约为0.3%,并且呈下降趋势。
在诊断为乳腺癌的BRCA致病性突变携带者中,发生第二次非相关乳腺癌的风险与初诊时的年龄、生物学和使用的系统治疗有关,但明显高于一般人群。
(有关与该人群中的CBC风险相关的更多信息,请参考“高外显率乳腺癌和/或妇科癌症易感基因”章节的“BRCA致病性突变携带者中的对侧乳腺癌”部分相关内容。)BRCA致病性突变携带者的第一个癌症预后良好,预估第二个乳腺癌无关事件的风险非常重要,有助于告知患者是否接受降低风险的手术,并且有利于提高生存率。
基因检测的时机和对BRCA致病性突变状态的了解可能影响手术决策,或放弃后续手术或影响随诊。因此,对于携带BRCA致病性突变风险增加的个体,应尽可能在术前考虑基因检测。
在一组148例携带BRCA1和BRCA2致病性突变的单侧乳腺癌患者中,其中79人接受了CRRM,,CBC的风险降低了91%,且与降低风险的卵巢切除术无关。在接受CRRM的女性中,其生存率更好,但这一结果可能与未接受手术组中先证癌或异时卵巢癌引起的较高死亡率有关。
欧洲10家医疗中心对550名女性(包括202名BRCA携带者)进行3334个女性年随诊跟踪调查,数据表明RRM非常有效。对终生风险为25%-80%的女性行双侧预防性乳腺切除术,其乳腺癌每年平均预期发病率为1%。在随访期间,这个队列中无人患乳腺癌,尽管预期有34例以上的乳腺癌发生。
对593例BRCA1和BRCA2致病性突变携带者进行回顾性研究,其中105例单侧乳腺癌患者接受了CRRM,10年生存率为89%,而未接受对侧风险降低手术患者为71%(P<0.001)。
这项研究受数个因素影响,如缺乏有关乳腺癌筛查、分级,大部分样本中雌激素受体状态不清。
荷兰的一项队列研究对583名在1980至2011年间确诊为BRCA致病性突变且诊为单侧乳腺癌的患者进行CRRM疗效的评估。
中位随访时间11.4年,242例(42%)患者在确诊后不同时间段接受RRM治疗(其中193例BRCA1致病性突变携带者和49例BRCA2致病性突变携带者)。与观察组相比,RRM组的OS有所改善(HR,0.49;95%CI,0.29-0.82),其中40岁以前确诊,肿瘤分级较低和非三阴性乳腺癌的OS改善最为明显。为了控制手术时机的偏差,作者对原发性癌症诊断2年后无疾病的女性进行了单独评估(HR,0.55;95%CI,0.32–0.95)。此外,接受RRM的患者更可能接受双侧输卵管卵巢切除术和全身化疗,这对生存结果或具有重要意义。
对390例来自已知BRCA1/BRCA2致病性突变家族的早期乳腺癌患者的回顾性研究发现,与选择单侧乳腺切除术的患者相比,双侧乳腺切除术患者的生存率显著提高。
患者的中位随访时间为14.3年(0.1-20.0年)。对诊断年龄、诊断年份、治疗及其他预后因素进行多变量分析,发现CRRM可降低48%乳腺癌相关性死亡率。该项研究规模较小,虽然作者对多个因素进行了调整,但残留的混杂因素或对结果产生影响。
此类研究均受选择的人群进行的相对较小的回顾性研究偏倚的限制。关于潜在的混杂变量方面(如社会经济状况、伴随疾病和获得护理的机会)的数据非常有限。有研究提出,选择接受RRM的女性更健康,因为她们能够忍受更广泛的手术。这一理论得到了一项研究的支持,该研究使用了监测、流行病学和最终结果(SEER)项目的数据,对单侧乳腺癌I-III期女性的CRRM和预后之间的关系进行评估。结果显示全因死亡率和乳腺癌特异性死亡率、非癌症事件死亡率均有所降低,这一发现预计或与CRRM无关。
BRCA致病性突变携带者选择保乳头乳腺切除术(NSM)作为风险降低的手术一直存有争议,因为担心为了保证乳头乳晕复合体(NAC)存活而在术中遗留的乳腺组织增多。然而,是否能留下最小残留组织可能与经验和技术有关。回顾性研究对2007至2014年间在两家医院对BRCA致病性突变携带者的NSM进行分析,201例BRCA致病性突变携带者进行了397个NSM。
这项研究包括未受累和受累的女性。150例RRM患者中,4例(2.7%)意外发现癌症;51例癌症患者中,2例(3.9%)意外发现癌症。平均随访32.6个月(范围1.0-76.0个月),随后发生4例癌症患者中,其中2例伴腋窝复发,1例初始NSM11个月后出现局部和远处复发,1例在乳腺下方发现新癌,保留乳头乳晕复合体(NAC)局部无复发。一项对2005至2013年间的89名BRCA致病性突变携带者进行了177例NSM的研究,报道了相似且较好的局部控制率。63名患者行降低风险的NSM(中位随访时间为26个月,范围11-42个月),26名患者有NSM和乳腺癌既往史(中位随访时间为28个月;范围15-43个月)。有5例需要进一步切除乳头。无局部复发或新诊乳腺癌病例报道。
多项研究对BRCA1或BRCA2致病性突变女性的RRM送检标本的病理组织学描述不一致。在两个系列研究中,37%-46%致病性突变的女性在接受单侧或双侧RRM时发现与乳腺癌风险正相关的增生性病变(原位小叶癌、不典型小叶增生、不典型导管增生和原位性导管癌[DCIS])。
在这些系列中,提示13%-15%患者在预防性切除的乳房中发现先前未疑似的DCIS。在47例已知携带BRCA1或BRCA2致病性突变且行单侧或对侧降低风险的乳腺切除的澳大利亚患者中,有3例(6%)在术中被诊断为乳腺癌。
一般来说,RRM标本的病理组织学结果并不影响治疗。
个人心理因素在未受累女性RRM和单侧乳腺癌患者CRRM的决策中起重要作用。(有关“BRCA携带者的RRM采用率”和“RRM术后心理结果研究”的更多信息,请参考“遗传性乳腺癌和卵巢癌综合征的社会心理问题”章节的“遗传性乳腺癌和卵巢癌的癌症风险管理的社会心理”部分相关内容。)
总结:
在普通人群中,根据胎次、体重和人工绝经时的年龄,切除双侧卵巢可降低75%乳腺癌发病风险。(更多信息,请参考PDQ摘要“乳腺癌预防”部分内容。)梅奥诊所(梅奥医学中心)对680名处于不同家族风险水平的女性进行研究发现,在接受双侧卵巢切除术的60岁以下女性中,所有乳腺癌的风险均有降低。
然而,卵巢切除术后可能会出现一些重要的副作用,如潮热、睡眠障碍、阴道干涩、性交不适,以及增加骨质疏松症和心脏病的风险。可能需要多种策略来消除卵巢切除后导致的不良影响。
降低风险的输卵管-卵巢切除术(RRSO)对乳腺癌影响的循证依据得到不断进展。早期小规模研究提示其具有保护作用。初步回顾性研究支持BRCA致病性突变-阳性的女性行RRSO,可降低乳腺癌和卵巢癌风险。
早期小规模研究包括了
一项对551名BRCA1或BRCA2突变女性的回顾性研究发现,RRSO后乳腺癌(HR,0.47;95%CI,0.29–0.77)和卵巢癌(HR,0.04;95%CI,0.01–0.16)的发病风险显著降低。
一项对170名BRCA1或BRCA2致病性突变女性的前瞻性单一机构研究得出了类似的结果。使用RRSO,卵巢癌、输卵管癌或原发性腹膜癌的HR为0.15(95%CI,0.02–1.31),乳腺癌为0.32(95%CI,0.08–1.2);任一癌症的HR均为0.25(95%CI,0.08–0.74)。
一项对1079名女性进行的前瞻性多中心研究平均随访30-35个月,结果发现虽然RRSO与BRCA1和BRCA2致病性突变携带者乳腺癌风险降低呈相关性,但BRCA2携带者乳腺癌风险降低更为显著(HR,0.28;95%CI,0.08–0.92)。
一项汇总分析对BRCA1/BRCA2致病性突变携带者且行RRSO和乳腺及卵巢/输卵管癌进行了研究,结果显示RRSO与乳腺癌风险降低呈显著相关性(总体:HR,0.49;95%CI,0.37–0.65;BRCA1:HR,0.47;95%CI,0.35–0.64;BRCA2:HR,0.47;95%CI,0.26–0.84)。
然而,在荷兰进行的一项针对822例BRCA1/BRCA2致病性突变携带者的队列研究(荷兰全国范围内进行BRCA携带者筛查)中,未发现RRSO可降低乳腺癌风险(HR,1.09;95%CI,0.67–1.77)。
作者认为,先前的发现或由方法学问题导致,包括癌症引起的检测和永恒人工用时偏差,并通过使用他们自身的队列和应用先前研究中关于计算人时的相同假设对其进行了经验评估。
作为反馈,美国研究人员使用荷兰研究的假设分析了他们的数据,但仍观察到与RRSO和乳腺癌风险成反比。
在676名女性的回顾性队列研究中,乳腺癌诊断时行RRSO的携带者其乳腺癌特异性死亡率风险降低(BRCA1携带者HR,0.38;95%CI,0.19–0.77;BRCA2携带者HR,0.57;95%CI,0.23–1.43)。
随后的一项国际性多机构研究表明,对3722名BRCA1和BRCA2携带者50岁前行卵巢切除术有助于预防BRCA2携带者的乳腺癌发生(HR,0.18;95%CI,0.05-0.63;P=0.007),但对BRCA1携带者则无影响。
考虑到这些结果相互矛盾,另一组17917名女性参与的前瞻性家庭式队列研究得到评估,其中1046名女性被诊断为乳腺癌,中位随访时间为10.7年;未观察到RRSO与乳腺癌之间具有相关性。
一项对2482例BRCA1/BRCA2致病性突变携带者的前瞻性多中心队列研究表明,RRSO可降低全因死亡率(HR,0.40;95%CI,0.26–0.61)、乳腺癌特异性死亡率(HR,0.44;95%CI,0.26–0.76)和卵巢癌特异性死亡率(HR,0.21;95%CI,0.06–0.80)。
随后的汇总分析证实了RRSO对BRCA1和BRCA2致病性突变携带者的全因死亡率(HR,0.32;95%CI,0.27-0.38)的影响,包括既往有/或无乳腺癌病史的携带者。
尽管现有文献中关于RRSO和乳腺癌风险的研究结果并不一致,但总体数据表明具有一定的临床意义,但这种临床意义的程度尚不完全清楚。需要进一步的前瞻性研究来证实这些发现。
有关RRSO对BRCA致病性突变携带者卵巢癌风险影响的更多信息,请参考“卵巢癌”章节的RRSO部分相关内容。
他莫昔芬(一种合成的抗雌激素药物)可增加乳腺细胞生长抑制因子,同时减少乳腺细胞生长刺激因子。国家外科辅助性乳腺癌和肠癌项目乳腺癌预防试验(NSABP-P-1),一项前瞻性的随机、双盲试验比较了高危女性(定义为Gail模型危险评分>1.66,年龄>60岁,或小叶原位癌)服用5年他莫昔芬(20mg/天)和安慰剂的临床疗效。结果证实,他莫昔芬可降低49%侵袭性乳腺癌风险。这种保护作用主要局限于ER阳性的乳腺癌,风险可降低69%。ER阴性乳腺癌的发生率未见明显下降。
他莫昔芬还可降低浸润前乳腺癌的风险。有/或无乳腺癌家族史女性的乳腺癌风险均有所降低。50岁以上女性的子宫内膜癌和血栓性事件的发病率增加。两项欧洲他莫昔芬预防试验的中期数据显示,中位随访分别为48和70 个月,发现他莫昔芬并未降低乳腺癌风险 。
然而,在一项试验中,在使用激素替代疗法(HRT)的亚组中发现乳腺癌风险有降低。
这些试验在研究设计和研究对象上的差异较大。(更多信息,请参考PDQ摘要“乳腺癌预防”部分内容。)
随后,国际乳腺癌干预研究1(IBIS-1)乳腺癌预防试验将7154名35-70岁的女性进行随机分配,接受他莫昔芬或安慰剂治疗5年。本试验的入组条件为具有家族史或异常良性乳腺疾病。中位随访16年,他莫昔芬组乳腺癌风险降低了29%(HR,0.71;95%CI,0.60–0.83)。浸润性ER阳性乳腺癌风险降低43%(HR,0.66;95%CI,0.54–0.81),乳腺导管原位癌风险降低35%(HR,0.65;95%CI,0.43–1.00)。浸润性ER阴性乳腺癌的风险未见降低。
这些结果证实了乳腺癌预防临床试验(P-1)的结果。
NSABP-P-1临床试验的子研究评估了他莫昔芬在35岁以上BRCA1/BRCA2致病性突变携带者中预防乳腺癌的临床意义。BRCA2阳性女性从他莫昔芬中的临床获益程度与无BRCA1/BRCA2致病性突变的女性相同;但是,BRCA1致病性突变的健康女性使用他莫昔芬并不能降低乳腺癌的发病率。鉴于样本中BRCA致病性突变携带者数量较少(8名BRCA1携带者和11名BRCA2携带者),对待这些数据应予以谨慎。
与他莫昔芬用于BRCA1和BRCA2致病性突变携带者的一级预防数据非常有限相比,数项研究发现他莫昔芬可降低对侧乳腺癌的发病风险。
在一项纳入约600例BRCA1/BRCA2致病性突变携带者的研究中,他莫昔芬可降低51%对侧乳腺癌的发病风险。
本报告的一项更新对285例双侧乳腺癌和751例单侧乳腺癌的BRCA1/BRCA2致病性突变携带者(40%的患者纳入初始研究)进行分析。他莫昔芬可降低对侧乳腺癌风险,其中BRCA1致病性突变风险降低50%;BRCA2致病性突变58%。他莫昔芬对接受卵巢切除术的女性似乎未显示临床获益,不过这个亚组的样本量较小。
另一项纳入160名BRCA1/BRCA2致病性突变携带者的研究表明,在乳腺癌根治术和放疗后使用他莫昔芬,可将对侧乳腺癌风险降低69%。
在另一项研究中,从三个家庭队列研究中收集2464名BRCA1/BRCA2致病性突变携带者且既往有乳腺癌病史的病例。通过回顾性和前瞻性数据分析,研究者发现在确诊后接受他莫昔芬辅助治疗的女性中,对侧乳腺癌的发病风险显著降低。在研究人员调整了确诊年龄和首发乳腺癌的ER状况后,这种关联仍然存在。这项研究的一个主要限制是56%的女性缺乏首发乳腺癌的ER状况信息。
这些研究受到回顾性、病例对照设计和缺乏原发性肿瘤ER状态信息的限制。
STAR试验(NSABP-P-2)纳入19000余名女性,并比较了5年雷洛昔芬和他莫昔芬的临床应用在降低浸润性乳腺癌风险方面的疗效。
平均随访时间3.9年,浸润性乳腺癌的发生率无差异;但是,他莫昔芬组的非浸润性乳腺癌较少。雷洛昔芬组的血栓栓塞事件发生率和子宫切除率明显降低。详细的生活质量数据显示,两组之间存在细微差异。
关于BRCA1或BRCA2致病性突变携带者临床疗效的数据尚未提供。(关于选择性ER调节剂和芳香化酶抑制剂在包括绝经后女性等普通人群应用的更多信息,请参阅PDQ摘要“乳腺癌预防”部分内容。)
另一项通过确定双侧乳腺癌患者识别的致病性突变携带者和非携带者的病例对照研究发现,全身辅助化疗降低了致病性突变携带者的CBC风险(RR,0.5;95%CI,0.2–1.0)。他莫昔芬与非显著性风险降低具有正相关性(RR,0.7;95% CI,0.3-1.8)。在非携带者中也发现了类似的风险降低;然而,考虑到携带者中具有较高的绝对CBC风险,辅助治疗或在风险降低方面发挥了更大影响。
他莫昔芬对714例BRCA1致病性突变携带者卵巢癌风险的影响已获研究。所有病例均有乳腺癌病史;结果显示,他莫昔芬与随后卵巢癌的风险增加无相关性(OR,0.78;95% CI,0.46–1.33)。
在普通人群中,初潮早和绝经晚可增加乳腺癌风险,第一胎足月妊娠年龄小可降低风险。(更多信息,请参阅PDQ摘要“乳腺癌预防”部分内容。)在护理健康研究中,还有一些其他因素,如无生育史、姐妹患乳腺癌的家族史。
有乳腺癌家族史的女性,任何年龄段妊娠似乎都会增加患乳腺癌的风险,持续到70岁。
一项研究评估了333名携带BRCA1高危致病性突变的女性患者中的风险调控因素。在已知BRCA1基因致病性突变的女性中,第一胎生产年龄小、三次及以上的胎次均能降低乳腺癌风险。据估计,每多一胎,至多5或5个以上胎次,RR为0.85;然而,高产次似乎与卵巢癌风险增加有关。
在新西兰的一项病例对照研究中,研究人员发现产次对有乳腺癌家族史的女性和没有家族史的女性在乳腺癌风险方面的影响未见差异。
妊娠对乳腺癌风险影响的研究显示,结果较为复杂,与产次的关系不一致。而且,BRCA1和BRCA2致病性突变携带者之间可能存在差异。
在BRCA1致病性突变携带者中,产次与乳腺癌风险降低的相关性较为一致。
值得注意的是,人工流产和自然流产似乎均与乳腺癌风险增加无关。
在普通人群中,包括多个流行病学研究的大规模协作再分析在内的数个研究显示,哺乳可轻微降低乳腺癌风险。
至少有一项研究表明,哺乳可能对BRCA1致病性突变携带者具有保护作用。在一项多中心病例对照研究中,来自多病例家族的685例BRCA1致病性突变的乳腺癌和280例BRCA2致病性突变乳腺癌、965例BRCA突变的非乳腺癌患者,结果显示,在BRCA1致病性突变携带者中,母乳喂养1年或1年以上可降低45%乳腺癌风险。
在BRCA2致病性突变携带者中未观察风险降低。第二项研究未能证实这种相关性。
目前没有一致的证据证明口服避孕药会增加一般人群患乳腺癌的风险。
(更多信息,请参阅PDQ摘要“乳腺癌预防”部分内容。)
尽管有一些小型研究报道,携带BRCA1/BRCA2致病突变人群使用OC会略微增加乳腺癌的患癌风险,
但一项荟萃研究的结论发现,较新的OC制剂与乳腺癌的患癌风险不存在显著相关性。
然而,1975年以前生产的OC与乳腺癌的风险增加相关。
本荟萃分析所纳入的大部分患者来自表12中总结的三项大型研究。
Kotsopoulos 等. (2014 年)a | Brohet 等. (2007年)b | Haile 等. (2006年)a,c | Narod 等. (2002年)a | ||
---|---|---|---|---|---|
研究人数 | 携带BRCA1突变的乳腺癌患者 | N = 2,492 | N = 597 | N = 195; 确诊年龄< 50岁 | N = 981 |
携带BRCA2突变的乳腺癌患者 | 不适用 | N = 249 | N = 128;确诊年龄< 50岁 | N = 330 | |
既往有口服避孕药史 | BRCA1 | 1.18 [CI 1.03–1.36] P = 0.02 | 1.47 [CI 1.13–1.91] | 0.64 [CI 0.35–1.16] | 1.38 [CI 1.11–1.72] P =0.003 |
BRCA2 | 不适用 | 1.49 [Cl 0.8–2.7] | 1.29 [Cl 0.61–2.76] | 0.94 [Cl 0.72–1.24] | |
口服避孕药年龄 <20 岁 | BRCA1 | 1.45 [CI 1.20–1.75] P =0.0001 | 1.41 [Cl 0.99–2.01] | 0.84 [Cl 0.45–1.55] | 1.36 [Cl 1.11–1.67] P =0.003 |
BRCA2 | 不适用 | 1.25 [Cl 0.57–2.74] | 1.64 [Cl 0.77–3.46] | 未报告 | |
总持续时间 | BRCA1 | <5 年:1.14 [CI 0.97–1.35] | <9年:1.51 [Cl 1.1–2.08] | <5年:0.61 [Cl 0.31–1.17] | <10年:1.36 [Cl 1.11–1.67] P = 0.003 |
>5 年:1.22 [CI 1.04–1.49] P = 0.02 | |||||
BRCA2 | 不适用 | <9 年:2.27 [Cl 1.1–4.65] | <5 年:0.79 [Cl 0.26–2.37] | <10 年:0.82 [Cl 0.56–1.91] | |
足月妊娠前使用 | BRCA1 | 不适用 | >4 年:1.49 [Cl 1.05–2.11] | >4 年:0.69 [Cl 0.41–1.16] | 未评估 |
BRCA2 | 不适用 | >4 年:2.58 [Cl 1.21–5.49] | >4 年:2.08 [Cl 1.02–4.25] 每年趋势:1.11; P趋势=0.01 | ||
CI = 置信区间。 | |||||
a报告的风险估计采用95% CI比值比(OR)。 | |||||
b 报告的风险估计采用95% CI危险比(HR) | |||||
c 风险估计受限于40岁以下的BRCA致病突变携带者。 |
在向患者询问避孕方法和预防措施时,需要考虑使用口服避孕药(OC)对乳腺癌和卵巢癌患癌风险的潜在影响以及其他与健康相关的影响。包括BRCA1或BRCA2致病突变携带者之间的潜在差异、年龄和用药时间的影响、以及OC对高外显性早发乳腺癌家族的影响在内的一系列重要的问题仍未解决。
(关于OC的使用和卵巢癌讨论的更多信息,请参考本摘要的“药物预防”章节中的“口服避孕药”部分内容。)
观察性和随机临床试验数据均表明,一般人群使用HRT可增加乳腺癌患病风险
女性健康倡议(WHI)是一项随机对照试验,通过对16000多名绝经后女性进行研究,分析饮食干预和激素治疗降低心脏病、乳腺癌、结直肠癌和骨折发病率方面的风险和益处。 这项研究的雌激素加孕激素组因健康风险超过获益而被提前终止,在该组中,超过16,000名妇女被随机分配接受联合激素疗法或安慰剂治疗。
促使研究终止的不利因素之一是,随机分配接受雌激素和孕激素治疗的女性罹患乳腺癌的总数(245例比185例)和浸润性乳腺癌(199例比150例)均显著增加(RR,1.24;95%CI,1.02-1.50;P<0.001)。
一项后续研究的结果表明,近期乳腺癌发病率的下降,特别是在50-69岁的女性中,主要与雌激素和孕激素联合激素替代疗法的使用减少有关。
与安慰剂组相比,与HRT相关的乳腺癌具有不良的预后特征(临床分期更晚,肿瘤更大),HRT还导致乳腺钼靶检查异常结果增多。
据报道,与绝经后HRT相关的乳腺癌风险有所增加,这结果或许未受乳腺癌家族史的影响。
在荟萃分析中,患癌风险与家族史无相关性。
WHI研究尚未报告乳腺癌家族史的分层分析,也没有对研究对象进行BRCA1/BRCA2致病性突变的系统检测。
在普通人群中,短期使用激素治疗更年期症状似乎很少或几乎没有增加乳腺癌风险。
已有两项研究发现了HRT治疗对BRCA1或BRCA2致病性突变携带者患乳腺癌风险的影响。在一项对462例BRCA1和BRCA2致病性突变携带者的前瞻性研究中,双侧降低风险的输卵管-卵巢切除术(RRSO)(n=155)与乳腺癌风险总体降低显著相关(HR,0.40;95%CI,0.18–0.92)。以未行双侧RRSO或未采用HRT治疗的致病性突变携带者为对照组时,HRT的使用(n=93)对双侧RRSO相关的乳腺癌风险降低无显著相关性(HR,0.37;95%CI,0.14-0.96)。
在一项对472名绝经后患有BRCA1致病性突变的女性进行的病例对照研究中,HRT的使用与乳腺癌风险的总体降低相关(OR,0.58; 95%CI,0.35-0.96; P = .03)。 在接受双侧卵巢切除术的女性和未进行双侧卵巢切除术的女性中,均未观察到风险的显着降低。 单独服用雌激素的女性OR值为0.51(95%CI,0.27–0.98; P = .04),而与雌激素和孕激素的相关性无统计学意义(OR,0.66; 95%CI,0.34–1.27; P = .21)。
一项病例对照研究对432对具有BRCA1致病性突变的绝经后妇女进行了比较,这些妇女有自己的癌症病史,与未患癌的BRCA1携带者进行比较。HRT的使用与乳腺癌发生风险的增加无关(OR,0.80,P=0.24)。
特别是鉴于观察性研究与WHI之间与HRT相关的预计风险有所不同,这些发现应在随机前瞻性研究中得到证实,
但是他们认为BRCA1 / BRCA2致病性突变携带者中的HRT既不会增加乳腺癌的风险,也不会否定卵巢切除术的保护作用。
有关在一般人群中进行筛查的信息,请参阅关于卵巢癌,输卵管癌和原发性腹膜癌筛查的PDQ摘要,以及有关筛查和预防相关证据水平的信息,请参阅PDQ摘要《癌症遗传学研究的证据水平》。 后者还概述了在常规临床实践中被认为适合筛查特定疾病之前必须满足的五个要求。
在一般人群中,卵巢的临床检查在鉴别卵巢癌方面没有特异性和敏感性。没有证据证明卵巢的临床检查(双手盆腔检查)能够让存在卵巢癌遗传风险的女性获益。
循证依据等级:未指定
在一般人群中,在附件包块的术前诊断中,经阴道超声(TVUS)似乎优于经腹超声。 由于绝经前卵巢的周期性月经改变(例如短暂性黄体囊肿),这两种技术在绝经前妇女中的特异性都比绝经后妇女低,从而导致检测结果解读困难。 前列腺癌、肺癌、结直肠癌和卵巢癌随机前瞻性筛查试验(PLCO-1)发现,每年使用TVUS和癌抗原125(CA-125)联合筛查无症状绝经后一般女性人群,不能降低死亡率。
存在卵巢癌遗传风险女性是否会从TVUS筛查中获益,与此相关的临床数据较为有限。一些回顾性研究报道了在高危女性中采用TVUS联合/不联合CA-125筛查卵巢癌的经验。
然而,在高风险标准的定义、筛查的依从性以及检测到的癌症是偶发还是普遍等方面没有一致性。一项最大的已报告的研究纳入888名BRCA1/BRCA2致病突变携带者,每年均采用TVUS和CA-125进行筛查。10名女性诊断卵巢癌,其中5名在诊断卵巢癌前3至10个月的筛查结果正常。另外5例经筛查诊断,诊断前6~14个月筛查结果正常,在这5个病例中,4例是临床IIIB或IV期。
一项类似的研究报告了312名高危女性(152名BRCA1/BRCA2致病突变携带者)每年的TVUS和CA-125结果。
因TVUS和CA-125异常而被发现癌症的4名患者中,均有症状,其中3例属于晚期疾病。在荷兰的一项研究中,每年采用盆腔超声、TVUS和CA-125对BRCA1/BRCA2致病突变携带者进行筛查,但在241例BRCA1/BRCA2致病突变携带者中未发现早期卵巢癌。
在研究过程中发现了3例癌症,均属于晚期临床IIIC期。
最终,一项针对1100名每年接受TVUS和CA-125检查的中高危女性的研究报告显示,13例卵巢肿瘤中有10例通过筛查发现。只有5例属于临床I期和II期。
每半年行TVUS和CA-125筛查的临床意义相关的数据较为有限。
在英国的家族性卵巢癌筛查研究中,对3,563名女性卵巢癌的终生风险估计为10%或更高的女性进行了每年超声和CA-125血清检测,平均筛查了3.2年。在13例筛查出癌症的患者中,有4例为临床I期和II期。前一年接受筛查的女性患IIIC期以上癌症的可能性更小;同时,还有一种趋势,即最佳肿瘤细胞减灭率和更好的总体生存率(OS)。 此外,大多数癌症发生在具有已知卵巢癌易感基因的女性中,从而确定了筛查风险最高的人群。
本项研究的II期将筛查频率提高到每4个月一次;目前这方面的影响尚未得出。
2009年完成了第一项以生存率为主要结果的TVUS和CA-125前瞻性研究。在3532例高危女性筛查中,981例BRCA致病突变携带者,其中49人发展成为卵巢癌。5年和10年生存率分别为58.6%(95%CI,50.9%-66.3%)和36%(95%CI,27-45),携带者和非携带者之间的生存率未见差异。本研究的一个主要局限是缺乏对照组。尽管有一些局限性,这项研究表明,每年采用TVUS和CA-125监测一次,在早期发现肿瘤及生存率方面无明显的临床意义。
如前一节所述,在部分回顾性研究中,TVUS联合血清CA-125对卵巢癌高危女性的筛查能力进行评估。
美国国立卫生研究院(NIH)关于“卵巢癌”的共识声明建议不要对一般人群进行血清CA-125卵巢癌的常规筛查。(更多信息,请参考“前列腺癌、肺癌、结直肠癌和卵巢癌筛查试验:卵巢癌、输卵管癌和原发性腹膜癌筛查”中的“单阈值CA-125水平和TVU”部分相关内容。)然而,NIH共识声明建议有卵巢癌遗传风险的女性从35岁开始,每6至12个月进行一次TVUS和血清CA-125筛查。
癌症遗传学研究联合会特别工作组建议,BRCA1致病突变携带者在25至35岁开始,每年或每半年接受一次TVUS和血清CA-125筛查。
这两条建议仅仅以专家意见和最佳临床判断为基础。
对于携带BRCA1和BRCA2致病性突变以及错配修复(MMR)基因(例如MLH1,MSH2,MSH6,PMS2)的卵巢癌风险高的女性,有效筛查卵巢癌尤为重要。 对于BRCA1致病性突变的携带者,有一种特殊的紧迫感,他们患卵巢癌的终生累积风险可能超过40%。
因此,许多新型卵巢癌生物标志物(单独或联合)有望在未来5-10年内作为卵巢癌筛查策略出现。 尽管这是一个活跃的研究领域,在早期研发中发现多种具有良好应用前景的新型生物标志物,但目前,这些生物标志物单独或联合应用的研究尚不充分,无法证明常规应用于普通人群或高遗传风险女性是合理的。
新型卵巢癌生物标志物的研发需要几个步骤,验证环节尤为重要。国家癌症研究所早期检测研究网络研究人员公布一个有效的框架。
他们指出,癌症筛查计划的目标是早期发现肿瘤和早期成功治疗。判断筛查计划的金标准是,在被筛查的癌症患者中,癌症死亡率是否降低。此外,筛查检测必须具有足够的无创性和廉价性,以便在人群中广泛普及。保持高度的特异性(即少有假阳性结果)对于人群筛查检测至关重要,因为即使是较低的假阳性率,也会导致许多人不得不接受不必要且昂贵的诊断过程和心理压力。有可能需要联合多种癌症生物标志物,才能进行敏感又特异的筛查检测。
此外,临床有用的检测必须具有较高的PPV(一个来自筛查人群中的敏感性、特异性和疾病流行率的参数)。实际上,一个PPV为10%的生物标志物意味着需要10例外科手术来确定一例卵巢癌;其余9例手术将代表假阳性的检测结果。一般来说,因为卵巢癌的发病率与双侧输卵管卵巢切除术有关,卵巢癌研究界认为PPV低于10%的生物标志物在临床上是不可接受的。最后,要时刻谨记,虽然新的生物标志物可能存在于晚期卵巢癌患者的血清中(代表了在生物标志物发展早期分析的大多数病例),但可能在早期疾病患者中检测到,也有可能检测不到,如果筛查在临床上有用的话,这一点很重要。
有研究认为,生物标志物的研发和验证一般分为五个阶段:
最后,要进行生物标志物适合于某特定人群的验证环节,必须在该特定人群中进行评估,而且无需事先选择已知阳性和阴性人群。此外,测试还必须证明其临床实用性,即与测试应用相关的临床获益和风险的良性净平衡。这些可能包括改善健康结局、获得心理社会和经济效益。
假阳性检测结果的潜在影响对于卵巢癌来说是一个独特的挑战。由于对早期疾病缺乏可靠的无创诊断检测,在探查术时可能无法发现有临床意义的早期卵巢癌。
因此,很可能有些患者会认为异常结果并不意味着癌症的存在。只有通过卵巢和输卵管切除术,并进行显微镜检查才能确诊癌。 为了避免不必要的手术和绝经前期女性出现诱导性假阳性结果,需要高度特异性的检测(即假阳性率非常低)。
对75例卵巢癌患者和254名高危对照者(携带BRCA致病突变或有明显乳腺癌和卵巢癌家族史的女性)进行了卵巢癌症状指数预测。
如果女性每月至少报告12次以上的任何预定义症状(腹胀或腹部增大,腹部或盆腔疼痛,进食困难或感觉不快),则症状指数为阳性。CA-125值大于30u/mL被视为异常。控制CA-125水平后,症状指数可独立预测卵巢癌(P<0.05)。结合CA-125升高和阳性症状指数,准确鉴别了89.3%病例。在CA-125未升高的患病 女性中,有50%女性的症状指数与癌症相关。但在无癌症高危对照组中,也有11.8%的症状指数为阳性。作者认为,一个包括CA-125和症状指数的综合指数的表现优于其中任何单独的检测,并且这种策略可作为多步骤筛查程序中的首个筛查项目。在未选择的筛查人群中,需要额外的检测能力验证和确定临床实用性。
对CA-125筛查的一种新型改进是基于一种假设,即随着时间的推移,CA-125水平升高可提供更好的卵巢癌筛查特征,优于简单地根据任意阈值将CA-125分为正常或异常。这种方法采用卵巢癌风险算法(ROCA)的形式实现,该算法是一种研究统计模型,将一系列CA-125检测结果和其他协变量纳入计算中,从而对计算筛查对象中存在卵巢癌的可能性进行估计。根据对斯德哥尔摩卵巢癌筛查检测中5550名平均风险女性的再分析,采用该策略的首份报告显示卵巢癌病例和对照组鉴别的敏感性为99.7%,特异性为83%,PPV为16%。与单纯CA-125检测的2% PPV相比,该策略的PPV测量增加了8倍。
在此报告之后,在一项前瞻性英国卵巢癌筛查试验中,采用ROCA对9233名绝经后女性的33621个系列CA-125值进行研究。
与固定CA-125阈值相比,ROCA的受试者操作曲线下面积从84%升高至93%(P=0.01)。观察结果首次证明,使用这种筛查策略可提高卵巢癌的临床前检测水平。英国的一项13000名50岁及以上普通志愿者的前瞻性研究采用一系列的CA-125值和ROCA,将参与者分为低、中、高风险亚组进行分析。
每组均有其规定的管理策略,包括TVU和每年(低风险)或3个月(中等风险)复查CA-125。使用该方案,ROCA的特异性为99.8%,PPV为19%。
英格兰有两项前瞻性试验使用了ROCA。 英国卵巢癌筛查合作试验(UKCTOCS)将高危女性随机分为1)无筛查,(2)每年超声检查或(3)多模式筛查(N = 202,638;应计完成;随访结束 (2014年),以及针对高危女性的英国家族性卵巢癌筛查研究(UKFOCSS)(应计完成)。 在美国,还有两个使用ROCA的高风险人群正在评估中:癌症遗传学网络ROCA研究(N = 2500;随访已完成;正在进行分析)和《妇科肿瘤学组规程199》(GOG-0199;招募已完成;随访至2011年)。
因此,将提供关于当前该研究筛查策略实用性的其他数据。
在过去十年里,已出现一系列新型卵巢癌生物标志物,如HE4;间皮细胞素;激肽释放酶6、10和11;骨桥蛋白;前列腺素;M-CSF;OVX1;溶血磷脂酸;血管内皮生长因子B7-H4;白细胞介素6和8。
这些都是单项研究,结合CA-125或其他组合方式。大多数研究的样本量相对较小和高选择性,包括在早期生物标志物研发的1和2阶段的已知卵巢癌病例和健康人群对照。这一结果在多个研究中并未得到验证;目前,尚缺乏一个能广泛应用于临床的筛查方案。
最初,血清蛋白的质谱分析与复杂的分析算法相结合,用于发现能够鉴别卵巢癌患者和对照组的蛋白质模式。
这种方法假设该模式本身就足以能鉴别,并且不需要对模式识别的特定蛋白质进行区别。使用类似的实验室工具,对该策略进行了改良,以识别有限数量的特定已知血清标志物,这些标志物可用于替代或与CA-125联用,进行癌症早期检测。
此类研究
一般都是小规模的病例对照研究,受样本量和早期癌症病例数量的限制。需要进一步评估,以确定以这种方式识别的任何其他标志物是否对未选择的临床特定人群的卵巢癌早期检测具有临床实用性。
由于单个生物标志物还没有达到有效筛查检测的标准,因此有研究提出,有必要结合多种卵巢癌生物标志物,以获得满意的筛查结果。该策略采用多重磁珠的平台定量分析六种血清生物标志物(瘦素、催乳素、骨桥蛋白、胰岛素样生长因子II、巨噬细胞抑制因子和CA-125)。
OvaSure旗下有相似的分析方法,后来制造商自愿退出市场。[回复FDA警告信]
本研究病例为术前采集血液的新诊断卵巢癌患者:36例为临床Ⅰ期和Ⅱ期;120例为临床Ⅲ期和Ⅳ期。对照组为年龄匹配的6个月内未发生卵巢癌健康人群。本研究中的病例和对照组在其家族和/或遗传风险状况方面均无明显特征,但被视为高风险人群。首先,对181名对照组和113名卵巢癌患者进行检测,以确定最能区分患者和对照组的生物标志物初始组(训练组)。检测结果组合应用于另外181个对照组和43个卵巢癌病例(测试组)。将早期和晚期卵巢癌合并到训练组和测试组中,采用假设卵巢癌患病率约为50%的公式,操作性能特征报告敏感性为95.3%,特异性为99.4%,PPV为99.3%,阴性预测值为99.2%,如高选择研究人群结果。
为了避免可能使检测性能看起来优于实际情况的偏差,通常不建议在此类分析中结合训练组和测试组人群。
最适合应用的流行率是待筛查未选择人群中的疾病流行率。卵巢癌在普通人群中的发病比例是1/2500。在手稿修正时,作者认为,筛查人群中卵巢癌的患病率为2500分之一(0.04%),重新计算的PPV仅为6.5%。 在此基础上,研究人员撤回了其声称该筛查适合人群的观点。 如果该试验用于卵巢癌风险较高的患者,那么该目标人群的实际患病率可能会高于一般人群中观察到的患病率,但远低于已发表的分析中假定的50%。 修订后的PPV为6.5%,表明大约有15位女性检测为阳性,实际上将患有卵巢癌,只有一小部分患有卵巢癌的患者将处于I或II期。 其余14项阳性测试将代表假阳性,这些女性将面临不必要的焦虑和潜在的疾病诊断程序的风险,包括双侧输卵管卵巢切除术。
根据先前描述的标准,
这种分析方法在研发过程中被归类为第2阶段。虽然这似乎是卵巢癌筛查研究的一种有效途径,但还需要更多的验证,特别是在代表临床筛查人群的未选择人群中。妇科肿瘤学家协会关于这项检测的一份声明指出:“我们认为,在机构审查委员会主持下和在适当的知情同意下进行的研究之外,在向妇女提供该试验之前,还需要进行进一步的研究来验证该检测的有效性。”
许多研究发现,具有乳腺癌和卵巢癌遗传风险的女性在RRSO后卵巢癌的风险降低。(更多信息,请参阅本摘要“乳腺癌”章节中的RRSO部分相关内容。)对551例BRCA1或BRCA2致病性突变女性的回顾性研究发现,双侧卵巢切除术后乳腺癌(HR,0.47;95%CI,0.29–0.77)和卵巢癌(HR,0.04;95%CI, 0.01–0.16)的风险显著降低。
一项针对170名BRCA1或BRCA2致病突变女性的前瞻性单中心研究报道了类似的趋势。
在卵巢切除术中,卵巢癌、输卵管癌或原发性腹膜癌的HR为0.15(95%CI,0.02–1.31),乳腺癌为0.32(95%CI,0.08–1.2);每种癌症的HR均为0.25(95%CI,0.08–0.74)。一项对1079名女性进行的前瞻性多中心研究发现,RRSO可显著降低BRCA1和BRCA2致病突变携带者的卵巢癌风险。这项研究还表明,RRSO与BRCA1和BRCA2致病突变携带者乳腺癌风险降低具有相关性;然而,BRCA2携带者乳腺癌风险的降低更为显著(HR,0.28;95%CI,0.08–0.92)。
在一项以色列的病例对照研究中,双侧卵巢切除术与降低卵巢/腹膜癌风险呈正相关(OR,0.12;95%CI,0.06–0.24)。
对行RRSO的BRCA1/BRCA2致病突变携带者、乳腺癌和卵巢/输卵管癌的所有报告进行汇总分析,证实RRSO可显著降低卵巢或输卵管癌风险(HR,0.21;95%CI,0.12–0.39)。研究还发现乳腺癌风险显著降低(总体:HR,0.49;95%CI,0.37-0.65;BRCA1:HR,0.47;95%CI,0.35-0.64;BRCA2:HR,0.47;95%CI,0.26-0.84)。
随后,一项针对2854对BRCA1或BRCA2致病突变的乳腺癌患者进行的配对病例对照研究显示,与自然绝经(OR,0.81;95%CI,0.62–1.07)相比,手术绝经(OR,0.52;95%CI,0.40–0.66)可显著降低乳腺癌风险。这项研究还报告在自然绝经后进行卵巢切除术女性的乳腺癌风险显著降低(OR,0.13;95%CI,0.02–0.54;P=.006)。
另一项对5783名BRCA1或BRCA2致病突变的女性进行平均随访5.6年的研究报告显示,186名女性中有68人死于卵巢癌、输卵管癌或腹膜癌。双侧卵巢切除术的HR为0.20(95%CI,0.13-0.30;P=0.001)。在无癌症病史的BRCA致病突变携带者中,70岁以下与卵巢切除术相关的全因死亡率HR为0.23(95%CI,0.13-0.39;P<0.001)。
在50名或更多受试者的研究中,发病率约为2.3%-11%。发病率存在差异,或由手术技术、病理处理和RRSO年龄不同导致。在对966名高危女性进行的GOG 199研究中,BRCA1致病突变携带者中隐性癌的发病率最高(4.6%),其次是BRCA2致病突变携带者(3.5%),而非携带者的发病率仅为0.5%。在绝经后女性中,隐性癌的病理诊断率高出4倍
除了降低卵巢癌和乳腺癌的风险外,RRSO还可以显着改善OS以及乳腺癌和卵巢癌的特异性生存率。 一项针对666名具有BRCA1和BRCA2中胚系致病性突变的女性的前瞻性队列研究发现,与无胚系致病突变的女性相比,RRSO女性的总死亡率为0.24(95%CI,0.08-0.71)。
这项研究提供了有关接受RRSO的女性具有生存优势的首个证据。
关于RRSO降低风险程度的研究已说明术中可能发现的隐匿性癌症范围。原发性输卵管癌、原发性腹膜癌和隐性卵巢癌均有报道。数项病例系列研究报道了在接受降低风险卵巢切除术的致病突变携带者中恶性肿瘤的发生率。在50名或更多受试者的研究中,发病率在2.3%-11%。
发病率存在差异,或由手术技术、组织病理处理和RRSO年龄不同导致。在对966名高危女性进行的GOG 199研究中,BRCA1致病突变携带者中隐性癌的发病率最高(4.6%),其次是BRCA2致病突变携带者(3.5%),而非携带者的发病率仅为0.5%。在绝经后女性中,隐匿性病理的诊断率高出4倍。
除了隐性癌,预防切除的输卵管组织中也发现了癌前病变。在一组12例BRCA1致病突变的女性患者中,11例在输卵管上皮中发现增生或异常增生。在部分病例中,具有多灶性病变。
这些病理结果与输卵管癌和原发性腹膜癌患者中的胚系BRCA1和BRCA2致病突变一致。
一项研究表明,早期输卵管癌或输卵管上皮内癌与随后的输卵管、卵巢或腹膜浸润性浆液性癌之间存在因果关系。
(更多信息,请参阅本摘要“卵巢癌病理”部分内容。)
这些发现支持输卵管癌作为遗传性卵巢癌综合征的一部分,在普通人群的所有妇科癌症中比例不到1%,在降低风险的手术时必须切除输卵管。有明确的证据表明,RRSO必须包括常规收集腹腔冲洗液,并认真通过连续切片进行整个附件的综合病理评估。
然而,即使在卵巢切除术后,腹膜仍有发生Müllerian型腺癌的低风险。
Gilda-Radner家族卵巢癌登记处的324名女性中,有6名(1.8%)后来发展成原发性腹膜癌。未注明随访期。
在238名BRCA1/BRCA2致病性突变的克雷顿研究注册者中,有5人后来发展为腹腔内癌(2.1%)。值得注意的是,所有5名女性都有BRCA1致病突变。
一项对1828名BRCA1或BRCA2致病突变女性进行的研究发现,在RRSO后20年,患原发性腹膜癌的风险为4.3%。
关于RRSO术中发现隐性癌的BRCA1和BRCA2致病突变携带者预后的数据有限。在对32例浸润性癌(n=15)和浆液性输卵管上皮内癌(STIC)(n=17)患者的多机构研究中,47%浸润性癌患者在32.5个月复发,总体生存率为73%。
对于有上皮内病变的女性,1名患者(约6%)在43个月时复发,这表明两例患者的疾病发展过程不同。另一项研究证实了STIC病变的恶性可能。243例RRSO良性病变患者中3例(1.2%)随后发展为原发性腹膜癌,9例STIC患者中,有2例(22%)中位随访63个月后发展为高级别盆腔浆液性癌。
考虑到目前卵巢癌筛查的局限性以及BRCA1和BRCA2致病突变携带者患癌的高风险,NCCN指南建议35至40岁之间或生育后的女性,可选择RRSO作为有效的风险降低手段。RRSO的最佳时机具有个体化,但基于致病突变状态评估女性卵巢癌的风险有助于临床决策。在一项对美国BRCA1和BRCA2家族的大型研究中,40岁时BRCA1致病突变携带者卵巢癌的年龄-特异性累积风险为4.7%,BRCA2致病突变携带者为1.9%。
通过对22项BRCA1和BRCA2致病突变携带者的研究进行综合分析后发现,从40岁到50岁,BRCA1致病突变携带者患卵巢癌的风险急剧增加,而携带BRCA2致病突变的风险在50岁前较低,但从50岁到60岁急剧增加。
在卵巢癌患者BRCA致病突变的人群研究中,BRCA2突变患者的发病年龄明显晚于BRCA1突变患者(57.3岁[范围40-72岁]比52.6岁[范围31-78岁])。
总之,与BRCA2致病突变相比,BRCA1致病突变的女性可以早些考虑行RRSO来降低卵巢癌风险;然而,BRCA2突变的女性仍然可以考虑早期行RRSO降低乳腺癌风险。
在BRCA1/BRCA2致病突变携带者行RRSO中是否联合子宫切除术尚存在争议。有人担心,当不采取子宫切除术时,近端输卵管的一小部分会有残留,从而导致残留组织的输卵管癌风险增加。然而,一些研究表明,绝大多数的输卵管癌发生在输卵管的远端或中段,这意味着发生近端输卵管癌的可能性很小。有一些报告表明,在致病突变携带者中,其子宫癌的发病率增加,
而是否会增加浆液性子宫癌的风险尚未得到证实。
一项对857名女性的前瞻性研究表明,子宫癌发病率升高的病例似乎与BRCA1致病突变携带者应用他莫昔芬有关;
随后,有一项研究对4456名携带BRCA1/BRCA2致病突变携带者进行分析,同一组研究对象也证实了这一点。
即便应用他莫昔芬,子宫内膜癌的风险也很小,10年累积风险为2%。
此外,可采用雷洛昔芬(不会增加子宫癌风险)和芳香化酶抑制剂预防绝经后女性患乳腺癌,可尽量减少他莫昔芬使用。因此,根据目前对BRCA致病突变携带者患子宫癌风险的了解,在RRSO时考虑子宫切除以降低子宫癌风险并不是一个特别令人信服的理由。联合子宫切除术确实为拟行激素治疗的BRCA致病突变携带者提供了简化替代方案的优势。子宫切除术后,女性可以单独服用雌激素(不会增加乳腺癌风险),不联合孕激素,从而消除绝经后出血的风险。
研究表明,切除子宫并不是降低风险的必要措施。200例确诊为子宫内膜癌的犹太女性患者和56例未经选择的子宫乳头状浆液性癌患者的研究显示,BRCA致病突变患者的发病率未见升高。
然而,小规模研究报道了子宫乳头状浆液性癌或是BRCA相关疾病中的一种。
在使用他莫昔芬治疗的ER阳性乳腺癌BRCA致病突变携带者中,子宫内膜癌的累积风险或是在该人群咨询降低风险的子宫切除术时需考虑的另一个因素。
对于年轻且未受累的BRCA致病突变携带者,也可以考虑子宫切除术,这些携带者可能更希望采用HRT治疗,但子宫切除术仅能提供一种简化的雌激素替代方案。在BRCA致病突变携带者咨询降低风险的手术的最佳选择时,综合数据表明RRSO术后残留输卵管组织的患癌风险是建议子宫切除术中最难以令人信服的原因。因此,在无他莫昔芬或其他潜在的子宫或宫颈问题的情况下,子宫切除不是BRCA携带者RRSO的常规操作。
对于在手术时处于绝经前的女性来说,手术绝经的症状(如潮热、情绪波动、体重增加和泌尿生殖系统疾病)会严重影响她们的生活质量。为了缓解此类症状,医生经常在术后开一个有时限疗程的全身HRT治疗。(更多信息,请参阅本摘要“BRCA1/BRCA2致病突变携带者中的激素替代疗法”部分内容。)
研究已评估了RRSO对生活质量(QOL)的影响。一项研究调查了846名高危女性,其中44%接受了RRSO,其余56%进行了定期筛查。
在368例BRCA1/BRCA2致病突变携带者中,72%接受了RRSO。接受RRSO或筛查患者或普通人群之间的生活质量评分(通过简表-36评估)无显著差异;然而,行RRSO的女性对乳腺癌和卵巢癌的担忧则较少(P<0.001),对癌症风险的感知更为有利(P<0.05),但内分泌症状更多(P<0.001),性功能更差(P<0.05)。值得注意的是,37%女性在RRSO后使用HRT治疗,其中62%是围绝经期或绝经后女性。
研究人员随后调查了450名绝经前高危女性,选择RRSO(36%)或筛查(64%)。在RRSO组中,47%应用HRT。HRT患者(n=77)的血管舒缩症状少于未应用HRT患者(n=87;P<0.05),但其血管舒缩症状多于筛查组(n=286)。同样,接受RRSO和HRT治疗的女性由于阴道干涩和性交困难而产生的性不适多于筛查组女性(P<0.01)。因此,虽然这些症状可通过使用激素替代疗法改善,但激素替代疗法并非完全有效,有必要进行更多研究来解决这些问题。
RRSO在BRCA1/BRCA2致病突变携带者中的长期非肿瘤作用尚不明确。在普通人群中,RRSO与心血管疾病、痴呆、肺癌死亡率和总死亡率增加有关。
当已有研究对卵巢切除术的患者年龄进行分析,发现最不利的影响因素是在45岁之前行RRSO且不采取雌激素替代疗法的女性。
RRSO可能会增加BRCA1/BRCA2致病突变携带者的代谢综合征风险。
RRSO还能降低该人群的短期死亡率。
RRSO后降低癌症风险的临床获益显而易见,但需要关于长期非肿瘤的利弊方面的进一步数据。
双侧输卵管切除术被认为是降低BRCA致病突变携带者风险的一种临时措施。
目前还没有资料表明输卵管切除术作为一种降低风险方法的有效性。该手术保留了卵巢功能,避免了绝经前患者过早绝经的不良影响。该手术可采用微创方式,双侧卵巢切除术可推迟至患者绝经期。尽管这些数据有力证明,BRCA致病突变携带者的盆腔浆液性癌起源于输卵管,但仍有部分癌症显然起源于卵巢。此外,双侧输卵管切除术可能会让患者产生一种错误的安全感,即已经完全消除了癌症风险,如同接受了双侧输卵管-卵巢切除术。一项对14名BRCA致病突变年轻携带者的小规模研究证明了这一手术具有可行性
然而,在这项研究中,并未对疗效和对卵巢功能的影响进行评估。今后需要前瞻性试验,以确定该手术作为降低风险干预措施的有效性。
在使用蒙特卡罗模拟的马尔可夫统计模型中,在BRCA1/BRCA2致病突变女性的质量调整预期寿命方面,降低风险的输卵管切除术联合延迟卵巢切除术是一种成本效益较高的策略。
另一项模拟卵巢癌风险、RRSO和输卵管切除术影响的研究发现,育龄女性行输卵管切除术和5-10年后行卵巢切除术,其预计卵巢癌风险的差异较小。
当前有前瞻性研究正在对双侧输卵管切除术和延迟卵巢切除术在患者满意度和降低卵巢癌方面的影响进行评估。
在普通人群中,口服避孕药(OC)对卵巢癌有保护作用。
一些研究,包括一项大规模、多中心、病例对照研究,显示口服避孕药的保护作用,
而以色列的一项以人群为基础的研究未能证明其保护作用。
类似的临床价值能否扩展到卵巢癌遗传风险增加的女性中,已有研究对此关注。一项对799名卵巢癌患者(BRCA1或BRCA2致病突变)和2424名对照组(未患卵巢癌,但BRCA1或BRCA2致病突变)的多中心研究显示,使用OCs可显著降低卵巢癌风险(OR,0.56;95%CI,0.45–0.71)。与无Ocs用药史的女性相比,持续应用1年的OR为0.67(95%CI,0.50-0.89)相关,每年应用OC的OR值为0.95(95%CI,0.92–0.97),使用3至5年时,保护作用最大。
这项研究纳入的病例还包括同一作者先前研究的女性,该研究证实了先前研究的结果。
一项以人群为基础的卵巢癌病例对照研究,未发现OC对BRCA1或BRCA2致病突变携带者有保护作用(OR,1.07,OC用药≥5年)。正如预期,OC对非携带者有保护作用(OR,0.53,OC用药≥5年)。
然而,一项对36例BRCA1致病突变携带者进行的以人群为基础的小型病例对照研究发现,在致病突变携带者和非携带者(OR,约0.5)中均有类似的保护作用。
对BRCA1致病突变女性进行的一项更大规模的病例对照研究表明,OC用药5年后临床获益最大,而BRCA2致病突变女性似乎在OC用药3年后获益最大。
一项对来自多个注册中心的受试者进行的多中心研究发现,将147例卵巢癌且伴BRCA1或BRCA2致病突变患者与304例致病突变的携带者但无癌症病史的患者进行对比研究,发现OC对卵巢癌具有保护作用(OR,0.62,OC用药6年以上)。
最后,对18项研究进行汇总分析,包括13627例BRCA致病突变携带者,其中2855例乳腺癌,1503例卵巢癌,结果显示OC用药可显著降低卵巢癌风险(总结RR,0.50;95% CI,0.33–0.75)。结果还提示,随着OC使用时间的延长,风险显著降低(OC使用每增加10年,风险降低36%)。1975年以后配制的OC药应用与乳腺癌风险无关。
(关于该人群中OC的使用和乳腺癌讨论方面的更多信息,请参阅本摘要“生殖系统因素”章节中的“口服避孕药”部分内容。)
有人认为,持续的排卵、反复的创伤和修复卵巢上皮,会增加卵巢癌的风险。在普通人群的流行病学研究中,抑制排卵的生理状态可降低卵巢癌风险。还有研究认为,黄体生成素对卵巢的长期过度刺激在卵巢癌发病机制中具有一定作用。
这些数据大多来源于对普通人群的研究,但一些信息表明,由于遗传倾向,高危女性也存在此类情况。
在普通人群中,与未产妇相比,分娩可降低45%卵巢癌风险。随后妊娠似乎可降低15%卵巢癌风险。
早期对BRCA1/BRCA2致病突变女性的研究表明,分娩可降低卵巢癌风险。
在一项大型病例对照研究中,分娩可显著降低BRCA1致病突变女性的卵巢癌风险,OR为0.67(CI 0.46-0.96)。
每次分娩时,BRCA1致病突变携带者的OR值均为0.87(CI,0.79-0.95)。在同一项研究中,BRCA2致病突变携带者的分娩与卵巢癌风险增加相关;然而,每次分娩OR为1.08(CI,0.90-1.29),无明显趋势。有必要进一步研究BRCA2致病突变携带者的分娩次数和卵巢癌风险之间的关系,但对于BRCA1致病突变携带者来说,每一次分娩均会显著降低卵巢癌的风险,包括散发性卵巢癌。
在普通人群中,母乳喂养可降低卵巢癌风险
关于BRCA致病突变携带者的研究数据较为有限。一项研究发现母乳喂养无保护作用。
在BRCA1或BRCA2致病突变女性进行的病例对照研究表明,输卵管结扎的女性患卵巢癌的风险显著降低(OR,0.39)。这种保护作用仅限于BRCA1致病突变的女性,并且该作用控制OC使用、分娩、乳腺癌病史和种族等方面后仍持续存在。
一项以色列卵巢癌病例对照研究发现,接受妇科手术(输卵管结扎术、子宫切除术、单侧卵巢切除术、卵巢囊肿切除术,不包括双侧卵巢切除术)的女性卵巢癌风险降低了40%-50%。
这种保护机制尚不明确。可能存在的作用机制包括:卵巢血流减少,导致排卵和/或卵巢激素分泌中断;输卵管阻塞,从而阻断潜在致癌物的途径;或到达卵巢的子宫生长因子浓度降低。
(更多信息,请参阅卵巢癌、输卵管癌和原发性腹膜癌预防部分相关内容。)
有关更多信息,请参阅本章节化学预防中口服避孕药部分的相关内容。
有数据表明,携带BRCA致病突变的男性患各种癌症的风险均有所增加,包括男性乳腺癌和前列腺癌(见表7)。
然而,由于信息有限,适用于BRCA致病突变男性携带者管理的临床指南的基础是共识声明和专家意见。
有研究认为,BRCA2相关的前列腺癌与侵袭性疾病表型有关。
特别是,近期两项研究报告了男性BRCA2携带者的前列腺癌中位生存期是4-5年。
此外,其中一项研究中还报告了5年死亡率为60%,而最近欧洲和北美报道的这一数字为2%-8%。
前列腺特异性抗原(PSA)筛选试验的对比随访。这些数据在BRCA1相关的前列腺癌中比较有限,但是最近的一些研究也表明了一种侵袭性疾病的表型。
对BRCA携带者进行PSA筛查的临床意义尚不清楚;然而,有研究认为(基于较小型研究)在前列腺癌诊断中,携带者的PSA水平有可能高于非携带者。
这些研究结果表明,PSA筛查对BRCA致病突变的男性具有潜在的临床实用价值,尤其是侵袭性表型。影响PSA筛查研究的初步结果显示,21名BRCA2携带者在PSA升高的基础上进行活检,阳性预测值(PPV)为47.6%。
对这些男性的筛查发现了有临床意义的前列腺癌,作者认为这些发现可为继续筛查提供了理论依据;然而,这种筛查尚未显示对生存期方面的影响。最终,男性BRCA致病突变的信息有可能为最佳筛查和管理策略提供信息。此外,近期数据表明,胚系BRCA2致病突变是影响前列腺癌生存率的独立预后因素。因此,作者得出结论,由于侵袭性疾病的表型,主动监测可能不是最佳的管理措施。
根据NCCN临床实践指南建议对BRCA致病突变的男性携带者进行乳腺癌筛查。
包括,自35岁起每12个月进行一次乳房自我检查培训和教育以及临床乳房检查。此外,从40岁开始,NCCN建议对BRCA2携带者进行前列腺癌筛查,对BRCA1携带者应考虑进行前列腺癌筛查。
有关更多信息,请参阅本摘要“遗传性乳腺癌和卵巢癌综合征的社会心理问题”中的“产前诊断和孕前基因检测部分”相关内容。
BRCA1相关乳腺肿瘤的不同特征对预后很重要。此外,BRCA1相关性乳腺癌的生长似乎加快,这提示了高增殖指数,而肿瘤大小与淋巴结状态没有预期的相关性。
这些病理特征常提示乳腺癌预后较差,早期研究提示BRCA1致病突变的乳腺癌患者的预后较散发性乳腺癌差。
这些研究特别关注到,BRCA1和BRCA2致病突变携带者中同侧和对侧第二原发乳腺癌的发病率增加。
(有关更多信息,请参阅本摘要的BRCA致病性突变携带者中的对侧乳腺癌。)来自两个中心的496名德系犹太(AJ)乳腺癌患者的回顾性队列研究,比较了56名BRCA1/BRCA2致病突变携带者的相对生存期,随访中位数为116月。 BRCA1致病性突变与疾病特异性生存率较差独立相关。 在接受化疗的女性中未观察到较差的预后。
一项大规模以人群为基础伴少数乳腺癌病例的以色列女性研究发现,BRCA1原始致病突变携带者(n=76)与非携带者(n=1189)的总体生存期无差异。
在一个欧洲队列中也发现了类似结果,在BRCA1相关乳腺癌的无疾病生存期无差异。
一项前瞻性队列研究对3220名来自北美和澳大利亚的乳腺癌患者(包括93名BRCA1携带者和71名BRCA2携带者)进行了平均7.9年的随访,结果显示BRCA1/BRCA2携带者和散发乳腺癌患者的预后无明显差异。
然而,结果是基于20世纪90年代末使用的化疗方案,并没有调整手术方式(肿物切除术vs.乳腺切除术)和卵巢切除术的影响。散发性乳腺癌与遗传性乳腺癌(POSH)前瞻性结果的研究招募了2733名女性,其中12%(n=338)具有BRCA1/BRCA2致病突变。携带者与非携带者的预后无显著差异。
然而,三阴性乳腺癌患者(n=558)的2年总体生存率(HR,0.59;P=0.47)高于非携带者,但5年和10年生存率均无统计学意义。
一组研究人员报道了77名未经选择的三阴性乳腺癌患者的BRCA1/BRCA2检测结果。其中15个(19.5%)有胚系BRCA1(n=11;14%)或BRCA2(n=3;4%)致病突变,或体细胞BRCA1(n=1)突变。BRCA1致病突变携带者确诊癌症的中位年龄为45岁,非携带者确诊癌症的中位年龄为53岁(P=0.005)。值得关注的是,这项研究还表明,与BRCA1致病突变相关的三阴性乳腺癌患者的复发风险低于非BRCA1相关的三阴性乳腺癌患者,但该研究受到规模的限制。
另一项研究显示,与BRCA1相关的乳腺癌与非BRCA1相关的三阴性乳腺癌的临床结果无差异,但与BRCA1相关的乳腺癌的脑转移发生率有上升趋势。在这两项研究中,除了一例BRCA1致病突变携带者外,其他携带者均接受了化疗。
随后,在一项对89名BRCA1携带者和175名三阴性乳腺癌非携带者的研究中,BRCA1致病突变在调整年龄、卵巢切除术和降低风险乳腺切除后并不是一个独立的生存预测因子。
然而,卵巢切除术可显著降低携带者的乳腺癌相关死亡率。
波兰的一项对3345名50岁以下临床I至III期乳腺癌患者进行研究,探讨了BRCA1致病突变对预后的影响。在该队列研究中,233名患者(7%)携带任一种波兰BRCA1原始致病突变(5382insC、C61G或4154delA)。结果显示,BRCA1携带者更年轻,ER和HER2/neu的阴性率较高。10年生存率相似(BRCA1携带者为80.9%,非携带者为82.2%)。卵巢切除术可延长BRCA1携带者生存期(HR,0.30;95% CI,0.12–0.75)。
总之,BRCA1相关肿瘤的预后与散发性肿瘤可能相似,但其临床、病理组织学和分子特征显示出更具侵袭性的表型。未接受化疗的BRCA1致病突变携带者的预后可能更差。然而,由于大多数BRCA1相关是三阴性乳腺癌,因此通常采用辅助化疗。目前,有研究正在明确BRCA1相关乳腺癌是否应该接受不同于散发性肿瘤的临床治疗。(更多信息,请参阅本摘要“BRCA1和BRCA2在系统治疗中作用”相关内容。)
与散发性乳腺癌相比,关于BRCA2相关乳腺癌预后的早期研究并未显示出实质性差异。
一项小规模研究报道了BRCA2致病突变的转移性乳腺癌患者的总体生存率显著升高。
回顾性和前瞻性研究 评估了乳腺癌新辅助化疗(特别是顺铂)用于BRCA1致病突变携带者的临床有效率。
已有回顾性综述,详细介绍了标准化疗的病理完全缓解(pCR)率。在采用蒽环类和紫杉类为基础的标准新辅助化疗方案的患者中,pCR率为40%-60%。
新辅助铂类化合物在BRCA致病突变患者中显示了令人满意的pCR率。一组107名BRCA1致病性突变的波兰女性患者接受了4个周期的顺铂(75 mg/m2,每3周重复),总pCR率为61%
然而,在50名患者的GeparSixto临床试验中,已知BRCA致病突变的个体亚群中,pCR率为66.7%。但是在蒽环类和紫杉烷的新辅助化疗方案中加用铂类药物,并未显示出临床获益。
TNT试验对比了多西紫杉醇与卡铂治疗376例转移性三阴性乳腺癌的临床疗效。29例患者有BRCA1或BRCA2致病性突变。在整个队列研究中,两组患者的客观缓解率(ORR)无差异;然而,BRCA携带者的客观缓解率存在差异。接受多西他赛治疗的致病突变携带者的ORR为33%,接受卡铂治疗组为68%(P=0.03) 。
已有多个临床试验对聚腺苷酸二磷酸核糖基聚合酶(PARP)抑制剂联合/不联合化疗的临床疗效进行研究。BRCA1和BRCA2在同源重组修复双链DNA断裂中较为活跃;PARP参与通过碱基切除修复基因修复单链断裂,以及通过在DNA链上捕获PARP。
2017年,两项III期临床试验对PARP抑制剂在转移性乳腺癌和BRCA致病突变患者中的作用进行研究。在OlympiAD临床试验中,302名患者被随机分为两组,一组给予口服奥拉帕利(300 mg,每日2次),另一组或由医生选择化疗(卡培他滨、艾日布林或长春瑞滨)。奥拉帕利治疗组患者的中位无进展生存期(PFS)从4.2个月延长至7.0个月(HR,0.58;P<0.001)。总生存期是次要终点,发现无显著统计学差异。
EMBRACA临床试验将431名患者随机分为两组,一组给予口服他拉唑帕利(1mg,每日1次),另一组采用卡培他滨、艾日布林、长春瑞滨和吉西他滨治疗。
接受他拉唑帕利治疗患者的中位PFS延长至8.6个月,对照组为5.6个月(HR,0.54;P<0.001)。总体生存期是EMBRACA的α保护终点,在首次报告时,数据尚不成熟,仅报告51%事件(HR,0.76;P=0.105)。在这些结果基础上,美国食品和药物管理局(FDA)已批准他拉唑帕利和奥拉帕利治疗不能手术或转移性乳腺癌患者、胚系BRCA致病性突变的患者。
现有多项研究对多个PARP抑制剂的治疗方案进行评估,包括靶向其他胚系致病突变和体细胞突变。无论早期疾病还是转移癌的临床试验,均对PARP抑制剂单药、PARP抑制剂与其他DNA损伤修复药物联合应用、免疫疗法、和其他靶向治疗进行评估,以提高临床疗效,扩大潜在获益的患者群体。
(有关BRCA相关卵巢癌治疗策略的更多信息,请参阅本摘要“卵巢癌”中“系统治疗”部分相关内容。)
虽然肿瘤切除加放疗已成为早期乳腺癌患者的标准局部治疗,但对于具有遗传性乳腺癌倾向且不选择即刻双侧乳腺切除的女性来说,其临床使用更为复杂。放疗用于BRCA1/BRCA2致病突变,最初曾有诱发肿瘤或引起过度毒性的担忧,但这毫无根据。
尽管如此,第二原发性乳腺癌的发病率仍处于升高趋势,这可能会影响治疗决策。
由于第二原发性乳腺癌的风险增加,在BRCA1致病突变年轻女性中,风险可能高达60%
一些BRCA1/BRCA2致病突变携带者在初次癌症确诊时,会选择双侧乳腺切除术。(更多信息,请参阅本摘要“BRCA致病突变携带者”中“对侧乳腺癌”部分相关内容。)然而,还有一些研究支持采用保乳手术作为原发性肿瘤的合理治疗手段。
据估计,10年后同侧乳腺癌的复发风险为10%-15%,与非携带者相似。
随访时间较长的研究结果表明,15年内同侧乳腺癌的发病率高达24%,主要是同侧第二乳腺癌(而不是原发肿瘤复发)。
尽管各项研究结果并不完全一致,但放疗、化疗、卵巢切除术和他莫昔芬均能降低同侧乳腺癌风险,
与散发性乳腺癌类似。接受保乳手术的女性与接受单侧乳腺切除术的女性患对侧乳腺癌的风险似乎无差异,这表明散射辐射并不会增加对侧乳腺癌风险。
这一发现已得到一项以人群为基础的病例对照研究的支持,该研究调查了55岁以前被确诊为乳腺癌的女性。
所有女性均采用了BRCA1/BRCA2基因分型检测。虽然携带者患对侧乳腺癌的风险是非携带者的四倍,但是首次接受放疗的携带者患对侧乳腺癌的风险并不高于未接受放疗的携带者。(关于放疗和乳腺癌风险的更多信息,请参阅“乳腺钼靶”部分内容。)最后,BRCA1/BRCA2致病性突变携带者选择保乳治疗或乳腺切除术,其15年总生存率无差异。
尽管预后通常较差,但一些研究发现,BRCA致病突变的卵巢癌患者的生存率有所提高。
在以色列进行的一项以人群为基础的全国性病例对照研究发现,与对照组相比,携带BRCA致病突变的卵巢癌患者的3年生存率显著提高。
同一队列的5年随访显示,BRCA1和BRCA2致病突变携带者(54个月)与非携带者(38个月)的生存期均得到改善,这一现象在临床III和IV期卵巢癌及高级别肿瘤的女性中最为明显。
在美国的一项对卵巢癌德系犹太人患者的研究中,结果显示,与无BRCA致病突变卵巢癌德系犹太人女性及两大组别的晚期卵巢癌临床试验的患者相比,BRCA致病突变卵巢癌患者的中位复发间隔时间更长,总体生存期延长。
在美国的一项以医院为基础的回顾性研究中,与散发病例相比,铂类化疗用于BRCA致病突变的卵巢癌德系犹太人具有较佳的临床疗效,采用原发性治疗的临床疗效、无疾病生存期和总体生存期进行评估。
同样,在一项非德系犹太人的BRCA致病突变女性的病例对照研究中也发现了显著的生存优势。
荷兰的一项研究同样显示,112名BRCA1/BRCA2携带者对铂类药物初始化疗的临床反应优于220名散发性卵巢癌。
美国一项以人群为基础的研究显示,BRCA2的总体生存期得到改善,但BRCA1携带者无改善。
然而,该研究仅纳入12例BRCA2致病突变和20例BRCA1致病突变患者。采用卵巢癌基因组图谱对29例高级别浆液性卵巢癌伴已知BRCA2突变患者(20个胚系突变,9个体细胞突变)进行分析,结果显示,与无BRCA致病突变患者相比,有BRCA突变患者的总体生存期和无进展生存期均得到明显改善。BRCA1致病突变与预后无显著相关性。
此外,对26项观察性研究(包括1213例BRCA致病突变携带者和2666例上皮性卵巢癌非携带者)的汇总分析显示,致病突变携带者的生存率更高(BRCA1:HR,0.73;95%CI,0.64-0.84;P<0.001;BRCA2:HR,0.49;95%CI,0.39-0.61;P<0.001)。
因此,卵巢上皮癌伴BRCA1或BRCA2突变患者的5年生存率均高于非携带者,其中BRCA2携带者的预后最佳。一项针对日本患者的研究发现,顺铂治疗BRCA1相关的临床Ⅲ期卵巢癌的临床疗效优于非遗传性癌症。
但是,还有一些研究显示,未发现卵巢癌患者中BRCA致病突变与OS改善相关。
其中最大的一项研究涉及大量未经选择的加拿大和美国病例,均已进行BRCA1和BRCA2致病突变的检测。在3年后,致病突变提示较好的预后,但在10年后,预后无差异。
此外,一项研究表明有家族史卵巢癌患者的生存期更差。
令人信服的数据表明,BRCA致病突变携带者具有短期生存期优势。然而,长期生存尚不明确。德系犹太人卵巢癌伴BRCA1或BRCA2原始致病突变患者的生存率似乎有所提高;
然而,还需要对其他人群进行进一步的大规模研究,以确定这种生存优势是否更广泛地适用于所有BRCA相关的癌症。
遗传性BRCA相关性卵巢癌预后改善的分子机制尚不清楚,但可能与BRCA基因的功能有关。BRCA基因在细胞周期检查点激活和通过同源重组修复受损DNA方面具有重要作用。
除了BRCA以外,还有其他基因保持同源重组,如ATM、BARD1、PALB2、BRIP1、RAD51、BLM、CHEK2和NBN。对大量卵巢癌进行综合基因检测,结果显示,约50%浆液性卵巢癌或有体细胞突变或胚系变异,导致同源重组缺陷。
同源修复的缺陷会损害细胞修复由某些化疗药物(如顺铂)引起的DNA交联的能力。临床前数据显示BRCA1影响乳腺癌和卵巢癌细胞系的化疗敏感性。BRCA1蛋白表达的降低已被证明可以增强顺铂的化疗敏感性。
与散发性肿瘤相比,BRCA相关卵巢癌患者对一线化疗和后续铂类化疗的反应均有所改善,或许有助于改善其预后。
同源重组修复基因缺陷(HRD)的卵巢癌患者,可能由胚系变异或体细胞突变引起。与具有完整同源重组基因的女性患者相比,HRD患者生存期延长。大多数同源重组修复基因变异由BRCA1和BRCA2的体细胞突变或胚系变异组成,其中1/3由其他同源修复基因变异导致。
PARP通路抑制剂已被研究用于治疗BRCA1或BRCA2缺陷的卵巢癌。(关于PARP抑制剂的更多信息,请参考本摘要“BRCA1和BRCA2在系统治疗中的作用”部分内容。)PARP主要通过碱基切除修复来修复单链断裂;BRCA1和BRCA2在同源结合修复双链DNA断裂中发挥重要作用。因此,推测在BRCA1或BRCA2缺陷的肿瘤中,通过PARP抑制剂来阻止碱基切除修复,从而导致细胞死亡,因为两个独立的修复机制被破坏,即合成致死性概念。同一概念还可用于HRD肿瘤。因此,PARP抑制剂可用于除了BRCA基因致病突变以外的其他同源重组缺陷的妇科肿瘤,临床应用具有拓展性。在临床实践中,有多种肿瘤检测方法用于确诊HRD肿瘤,方法和定义各不相同。现有多项关于PARP抑制剂用于HRD卵巢癌的临床研究正在进行。
已有多项研究对PARP抑制剂用于铂类化疗后的卵巢癌探索。口服PARP抑制剂奥拉帕利的I期研究表明,该药用于BRCA1和BRCA2致病突变携带者卵巢癌、乳腺癌和前列腺癌具有较好的耐受性和临床疗效。
II期试验证实了两种不同剂量奥拉帕利治疗BRCA1或BRCA2致病突变的复发性卵巢癌患者具有良好的耐受性和有效性。
奥拉帕利400mg剂量组的总体有效率为33%(11/33);奥拉帕利100mg剂量组13%(3/24)(即每天16粒胶囊),两组患者均每日服药两次。最常见的不良反应是轻微恶心和疲劳。
除了有生殖系BRCA1或BRCA2致病突变的卵巢癌患者,PARP抑制剂也可能对有体细胞BRCA1或BRCA2突变或基因表观遗传沉默的卵巢癌患者有用。
多项II期治疗研究探讨了奥拉帕利用于铂类药物敏感或耐药的复发性卵巢癌患者的临床疗效。在一项单组研究中,奥拉帕利(400mg,每日2次)可用于治疗298种BRCA相关癌症,包括乳腺癌、胰腺癌、前列腺癌和卵巢癌。在接受奥拉帕利治疗的193名卵巢癌患者中,31%有临床缓解,40.4%出现持续至少8周病情稳定。
在之前至少采用三种化疗方案的154名女性中,总有效率约为30%,对铂类药物敏感和耐药的患者的中位有效期分别为8.2个月和8.0个月。
另一项对173例铂敏感患者的研究,将紫杉醇/卡铂联合奥拉帕利与单用紫杉醇/卡铂方案进行对比。奥拉帕立联合组的PFS显著优于对照组(12.2个月比9.6个月)(HR,0.51;95%CI,0.34-0.77),尤其是BRCA致病突变患者的亚组(HR,0.21;95%CI,0.08-0.55)。奥拉帕利联合组与对照组的总体生存期无差异。
与此相反,其他研究提示,BRCA状态不能预测奥拉帕利治疗铂类药物敏感卵巢癌患者的生存优势。一项随机开放式临床试验将90例复发性铂类药物敏感的卵巢癌患者分为奥拉帕利组、西地尼布/奥拉帕利联合组。联合组中位PFS显著延长(17.7个月比9个月)(HR,0.42;95% CI,0.23-0.76)。子集分析显示,在43例BRCA野生型/未知患者中,西地尼布和奥拉帕利联合应用比奥拉帕利单药具有更长的PFS(16.5个月比5.7个月)(HR,0.32;P=0.008)。在47例BRCA致病突变患者中,PFS延长幅度较小(19.4个月比16.5个月)(HR,0.55;P=0.16)。
在另一项研究中,BRCA1/BRCA2致病突变和复发性卵巢癌的女性且在12个月内采用铂类药物治疗的患者,被随机分为阿霉素脂质体(Doxil)组(n=33)、奥拉帕利200mg每天两次组(n = 32)与奥拉帕利400mg每天两次组(n = 32)。这项研究显示组间的主要终点PFS无差异。
值得注意的是,脂质体阿霉素组的临床有效率高于预期。这与其他研究一致,即与BRCA1/BRCA2相关的卵巢癌可能比散发性卵巢癌对脂质体阿霉素更敏感。
另一项研究表明,奥拉帕利用于复发性卵巢癌患者具有显著疗效,其中BRCA1/BRCA2致病突变患者(客观缓解率[ORR],41%)和无BRCA1/BRCA2致病突变患者(ORR,24%)。
本研究强调散发性卵巢癌,特别是高级别浆液性卵巢癌,可能与BRCA1/BRCA2致病突变相关肿瘤具有相似的特性。
作为维持治疗,奥拉帕利可明显改善对铂类药物敏感的复发性卵巢癌的PFS。在一项对265名患者进行的随机对照研究(研究19)中,接受奥拉帕利治疗患者的PFS为8.4个月,而安慰剂组的PFS为4.8个月(HR为0.35;95%CI,0.25-0.49)。
在该队列研究中,136例BRCA致病突变患者与安慰剂组相比,奥拉帕林组临床获益最显著,两组PFS分别为11.2个月和4.3个月(HR,0.18;95%CI,0.1-0.31)。
在整个队列研究中或在BRCA致病突变携带者中,未观察到OS差异。随后探索性分析排除了疾病进展时采用PARP抑制剂治疗的BRCA致病突变患者,最大限度减少对OS的影响。在97名患者中,与安慰剂相比,奥拉帕利可延长OS,HR为0.52(95%CI,0.28–0.97)。
经过5年多随访,较为成熟的研究数据显示,与安慰剂组相比,奥拉帕利组在整个队列中有明显的OS获益趋势,但未达到P<0.0001的预设显著性阈值(29.8个月比27.8个月;HR,0.73;95% CI,0.55-0.96),或在奥拉帕利治疗的BRCA致病突变携带者中(24.5个月比26.6个月;HR,0.62;95% CI,0.41-0.94)。
与安慰剂相比,奥拉帕利片已被证实是一种有效的维持治疗,可用于复发性、铂类药物敏感的卵巢癌和BRCA致病突变人群(SOLO2试验)。奥拉帕利组平均PFS为19.1个月,安慰剂为5.5个月(HR为0.30;95% CI,0.22-0.41)。与每天16粒胶囊相比,奥拉帕利片还具有减负荷优势(每次2片,每日2次)。
奥拉帕利可作为晚期、BRCA相关卵巢癌新发病例在初次治疗后的维持治疗,临床研究显示出其具有显著的临床获益。SOLO-1试验将391例BRCA致病突变的女性随机分为两组:奥拉帕利组(n=260)或安慰剂(n=131)。中位随访41个月后,与安慰剂组相比,奥拉帕利组患者的疾病进展或死亡风险降低70%,估计PFS延长约3年。
3年内,奥拉帕利组260名女性中有102名(39%)出现疾病进展或死亡,安慰剂组131名女性中有96名(73%)出现疾病进展或死亡。28%患者因不良反应而减少剂量,超过一半的患者出现剂量中断。疲劳和恶心是常见的不良反应,这也是减少剂量的原因。
芦卡帕利是PARP-1、-2和-3的小分子抑制剂,2016年12月在美国被批准用于治疗晚期胚系BRCA1/BRCA2相关的卵巢癌。一项II期临床研究发现,在治疗乳腺癌和卵巢癌相关的女性患者时,采用持续给药方式取得的临床疗效优于间歇给药。
随后的Ⅰ期/Ⅱ期剂量-结果研究根据可控毒性选择了芦卡帕利(600mg,每日两次),42例复发性、胚系BRCA相关的高级别浆液性癌患者的临床缓解率为59.5%,这些患者先前接受了2-4种治疗方案。常见的3级不良反应包括疲劳、恶心和贫血。
ARIEL-2 II期研究发现,芦卡帕利可有效用于BRCA突变的复发性、高级别、铂类药物敏感的卵巢癌。同样还可用于BRCA野生型伴杂合性基因缺失(LOH)患者,提示可能是HRD癌。该研究招募了206名女性,其中40名女性有BRCA胚系致病突变或体细胞突变。另有82例为BRCA野生型伴高度LOH。与低LOH亚组(5.2个月)相比,BRCA变异亚组的中位PFS显著延长(12.8个月)(HR,0.27;95%CI,0.16-44),高LOH亚组为(5.8个月)(HR,0.62;95%CI,0.42-0.90)。结果提示,BRCA变异状态和LOH评分可作为HRD的替代指标,均可作为芦卡帕利治疗复发性、铂类药物敏感、高级别卵巢癌患者的预测因子。
一项III期临床试验评估了芦卡帕利与安慰剂用于576例复发性、铂类药物敏感、高级别卵巢癌患者且经二线或以上铂类化疗后患者的临床疗效。研究发现196名BRCA致病突变:130个胚系变异和56个体细胞突变。芦卡帕利组的平均PFS为10.8个月,对照组为5.4个月(HR为0.35;95% CI,0.30-0.45)。BRCA相关性卵巢癌中位PFS最长:芦卡帕利组16.6个月,安慰剂组5.4个月(HR,0.23;95%CI,0.16-0.34)。在HRD癌症患者中,PFS中位数为13.6个月,对照组5.4个月(HR,0.32;95%CI,0.24-0.42)。根据这些数据,作者得出结论,在没有其他HRD或BRCA检测的情况下,单凭铂类药物的敏感性就足以预测芦卡帕利治疗晚期卵巢癌患者的疗效。
尼拉帕利是PARP-1和2的选择性抑制剂。一项I期剂量-结果研究发现,在复发性BRCA相关实体瘤患者中,尼拉帕利(每日300mg)的临床有效率为42%。
在500名铂类药物敏感的复发性卵巢癌患者的队列研究中,234名患者接受尼拉帕利维持治疗,116名患者接受安慰剂(NOVA试验)。
采用尼拉帕利维持治疗的BRCA致病突变携带者(21个月)和HRD阳性的野生型患者(12个月)的PFS较无HRD肿瘤阳性的野生型患者(9个月)有所延长。与先前数据一致,胚系BRCA致病突变患者的PFS最长。无论铂类药物反应或变异状态,以尼拉帕利为基础的维持治疗在经多重治疗的卵巢癌患者取得了广泛的临床疗效。QUADRAⅡ期试验研究分析了463例复发的、可评估的卵巢癌患者应用尼拉帕利的抗肿瘤疗效。该研究患者之前平均接受过四种治疗方法。在铂类药物敏感、HRD阳性的女性中,28%患者出现临床缓解,中位持续时间为9个月。
需要更成熟的数据来确定铂类药物敏感性是否是BRCA致病突变患者采用PARP抑制剂治疗的预测指标,判断PARP抑制剂作为治疗或维持治疗的最佳时机。在对同源修复通路中涉及的多个基因有更多了解的基础上,还可采用HRD状态预测PARP治疗效果。
表13列举了已公布癌症风险评估和遗传咨询、遗传检测和/或遗传性乳腺癌和卵巢癌管理建议的组织机构名称。
组织名称 | 转诊建议 | 风险评估和遗传学咨询建议 | 基因检测建议 | 管理建议 |
---|---|---|---|---|
美国医学遗传学和基因组学学院/国家遗传顾问协会(2015) | 已制定 | 风险评估:已制定 | 未制定 | 未制定 |
遗传学咨询:已制定 | ||||
美国妇产科学院(2017) | 已制定 | 风险评估:已制定 | 已制定 | 已制定 |
遗传学咨询:已制定 | ||||
美国临床肿瘤学会(2015) | 已制定 | 风险评估:一般大纲,非HBOC特异性 | 一般大纲,非HBOC特异性 | 未制定 |
遗传学咨询:已制定 | ||||
欧洲医学肿瘤学会(2016) | 参考其他已公布的指南 | 风险评估:参考其他已公布的指南 | 参考其他已公布的指南 | 已制定 |
遗传学咨询:已制定 | ||||
国家乳腺中心认证计划(2014) | 参考其他已公布的指南 | 风险评估:参考其他已公布的指南 | 检测指征未制定,检测前后的咨询内容已制定。 | 未制定 |
遗传学咨询:已制定 | ||||
全国遗传咨询师协会(2013) | 已制定 | 风险评估:参考其他已公布的指南和模型 | 已制定 | 参考其他已公布的指南 |
遗传学咨询:已制定 | ||||
全国癌症综合网络(2020) | 已制定 | 风险评估:已制定 | 已制定 | 已制定 |
遗传学咨询:已制定 | ||||
妇科肿瘤学会(2015,2017) | 已制定 | 风险评估:已制定 | 已制定 | 已制定 |
遗传学咨询:已制定 | ||||
美国预防服务工作队(2019) | 已制定 | 风险评估:已制定 | 已制定一般术语和参考其他指南 | 已制定一般术语和参考其他指南 |
遗传学咨询:已制定 | ||||
ACMG/NSGC=美国医学遗传学和基因组学学院/国家遗传顾问协会;ACOG=美国妇产科学院;ASCO=美国临床肿瘤学会;ESMO=欧洲医学肿瘤学会;NAPBC=国家乳腺中心认证计划;NCCN=全国癌症综合网络;NSGC=全国遗传咨询师协会;SGO=妇科肿瘤学会;USPSTF=美国预防服务工作队。 | ||||
a USPSTF指南适用于未经癌症诊断的个人。 |
Increasing data are available on the outcomes of interventions to reduce risk in people with a genetic susceptibility to breast cancer or ovarian cancer.
As outlined in other sections of this summary, uncertainty is often considerable regarding the level of cancer risk associated with a positive family history or genetic test. In this setting, personal preferences are likely to be an important factor in patients’ decisions about risk reduction strategies.
Refer to the PDQ summary on Breast Cancer Screening for information on screening in the general population, and to the PDQ summary Levels of Evidence for Cancer Genetics Studies for information on levels of evidence related to screening and prevention.
In the general population, evidence for the value of breast self-examination (BSE) is limited. Preliminary results have been reported from a randomized study of BSE being conducted in Shanghai, China.
At 5 years, no reduction in breast cancer mortality was seen in the BSE group compared with the control group of women, nor was a substantive stage shift seen in breast cancers that were diagnosed. (Refer to the PDQ summary on Breast Cancer Screening for more information.)
Little direct prospective evidence exists regarding BSE in individuals with an increased risk of breast cancer. In the Canadian National Breast Screening Study, women with first-degree relatives (FDRs) with breast cancer had statistically significantly higher BSE competency scores than those without a family history. In a study of 251 high-risk women at a referral center, five breast cancers were detected by self-examination less than a year after a previous screen (as compared with one cancer detected by clinician exam and 11 cancers detected as a result of mammography). Women in the cohort were instructed in self-examination, but it is not stated whether the interval cancers were detected as a result of planned self-examination or incidental discovery of breast masses.
In another series of carriers of BRCA1/BRCA2 pathogenic variants, four of nine incident cancers were diagnosed as palpable masses after a reportedly normal mammogram, further suggesting the potential value of self-examination.
A task force convened by the Cancer Genetics Studies Consortium has recommended “monthly self-examination beginning early in adult life (e.g., by age 18–21 y) to establish a regular habit and allow familiarity with the normal characteristics of breast tissue. Education and instruction in self-examination are recommended.”
Few prospective data exist regarding clinical breast examination (CBE).
The Cancer Genetics Studies Consortium task force concluded, “As with self-examination, the contribution of clinical examination may be particularly important for women at inherited risk of early breast cancer.” They recommended that female carriers of a BRCA1 or BRCA2 high-risk pathogenic variant undergo annual or semiannual clinical examinations beginning at age 25 to 35 years.
In the general population, strong evidence suggests that regular mammography screening of women aged 50 to 59 years leads to a 25% to 30% reduction in breast cancer mortality. (Refer to the PDQ summary on Breast Cancer Screening for more information.) For women who begin mammographic screening at age 40 to 49 years, a 17% reduction in breast cancer mortality is seen, which occurs 15 years after the start of screening.
Observational data from a cohort study of more than 28,000 women suggest that the sensitivity of mammography is lower for young women. In this study, the sensitivity was lowest for younger women (aged 30–49 y) who had an FDR with breast cancer. For these women, mammography detected 69% of breast cancers diagnosed within 13 months of the first screening mammography. By contrast, sensitivity for women younger than 50 years without a family history was 88% (P = .08). For women aged 50 years and older, sensitivity was 93% at 13 months and did not vary by family history.
Preliminary data suggest that mammography sensitivity is lower in BRCA1 and BRCA2 carriers than in noncarriers.
Subsequent observational studies have found that the positive predictive value (PPV) of mammography increases with age and is highest among older women and among women with a family history of breast cancer.
Higher PPVs may be due to increased breast cancer incidence, higher sensitivity, and/or higher specificity.
One study found an association between the presence of pushing margins and false-negative mammograms in 28 women, 26 of whom had a BRCA1 pathogenic variant and two of whom had a BRCA2 pathogenic variant. Pushing margins, characteristic of medullary histology, are associated with an absence of fibrotic reaction.
In addition, rapid tumor doubling times may lead to tumors presenting shortly after an apparently normal study. In one study, mean tumor doubling time in BRCA1/BRCA2 carriers was 45 days, compared with 84 days in noncarriers.
Another study that evaluated mammographic breast density in women with BRCA pathogenic variants found no association between pathogenic variant status and mammographic density; however, in both carriers and noncarriers, increased breast density was associated with increased breast cancer risk.
The randomized Canadian National Breast Screening Study-2 compared annual CBE plus mammography to CBE alone in women aged 50 to 59 years from the general population. Both groups were given instruction in BSE.
Although mammography detected smaller primary invasive tumors, more invasive cancers, and more ductal carcinoma in situ (DCIS) than CBE, the breast cancer mortality rates in the CBE-plus-mammography group and the CBE-alone group were nearly identical, and compared favorably with other breast cancer screening trials. After a mean follow-up of 13 years (range, 11.3–16.0 y), the cumulative breast cancer mortality ratio was 1.02 (95% confidence interval [CI], 0.78–1.33). One possible explanation of this finding was the careful training and supervision of the health professionals performing CBE.
Digital mammography refers to the use of a digital detector to find and record x-ray images. This technology improves contrast resolution
and has been proposed as a potential strategy for improving the sensitivity of mammography. A screening study comparing digital with routine mammography in 6,736 examinations of women aged 40 years and older found no difference in cancer detection rates;
however, digital mammography resulted in fewer recalls. In another study (ACRIN-6652) comparing digital mammography to plain-film mammography in 42,760 women, the overall diagnostic accuracy of the two techniques was similar.
When receiver operating characteristic curves were compared, digital mammography was more accurate in women younger than 50 years, in women with radiographically dense breasts, and in premenopausal or perimenopausal women.
In a prospective study of 251 individuals with BRCA pathogenic variants who received uniform recommendations regarding screening and risk-reducing surgery, annual mammography detected breast cancer in six women at a mean of 20.2 months after receipt of BRCA results.
The Cancer Genetics Studies Consortium task force has recommended for female carriers of a BRCA1 or BRCA2 high-risk pathogenic variant, “annual mammography, beginning at age 25 to 35 years. Mammograms should be done at a consistent location when possible, with prior films available for comparison.”
Data from prospective studies on the relative benefits and risks of screening with an ionizing radiation tool versus CBE or other nonionizing radiation tools would be useful.
Certain observations have led to the concern that carriers of BRCA pathogenic variants may be more prone to radiation-induced breast cancer than women without pathogenic variants. The BRCA1 and BRCA2 proteins are known to be important in cellular mechanisms of DNA damage repair, including those involved in repairing radiation-induced damage. Some studies have suggested intermediate radiation sensitivity in cells that are heterozygous for a BRCA variant, but this is not consistent and varies by experimental system and endpoint.
Three studies have failed to find convincing evidence of an association between ionizing radiation exposure and breast cancer risk in carriers of BRCA1 and BRCA2 pathogenic variants.
In contrast, two large international studies found evidence of an increased breast cancer risk due to chest x-rays or estimates of total exposure to diagnostic radiation.
A large, international, case-control study of 1,601 carriers of pathogenic variants described an increased risk of breast cancer (hazard ratio [HR], 1.54) among women who were ever exposed to chest x-rays, with risk being highest in women aged 40 years and younger, born after 1949, and exposed to x-rays only before age 20 years.
Some of the subjects in this study were also included in a larger, more comprehensive analysis of carriers of pathogenic variants from three European centers.
In that study of 1,993 carriers of BRCA1 and BRCA2 pathogenic variants from the United Kingdom, France, and the Netherlands, age-specific total diagnostic radiation exposure (e.g., chest x-rays, mammography, fluoroscopy, and computed tomography) estimates were derived from self-reported questionnaires. Women exposed before age 30 years had an increased risk (HR, 1.90; 95% CI, 1.20–3.00), compared with those never exposed. This risk was primarily driven by nonmammographic radiation exposure in women younger than 20 years (HR, 1.62; 95% CI, 1.02–2.58). Subsequently, a prospective study of 1,844 BRCA1 carriers and 502 BRCA2 carriers without a breast cancer diagnosis at study entry, with an average follow-up time of 5.3 years, observed no significant association between prior mammography exposure and breast cancer risk.
Additional subgroup analyses in women younger than 30 years demonstrated no association with breast cancer risk.
With the routine use of magnetic resonance imaging (MRI) in carriers of BRCA1 and BRCA2 pathogenic variants, any potential benefit of mammographic screening must be carefully weighed against potential risks, particularly in young women.
One study has suggested that the most cost-effective screening strategy in carriers of BRCA1 and BRCA2 pathogenic variants may be annual MRI beginning at age 25 years, with alternating MRI and digital mammography (so that each test is done annually but screening occurs every 6 months) beginning at age 30 years.
The National Comprehensive Cancer Network (NCCN) currently recommends annual breast MRI screening with contrast (or mammogram with consideration of tomosynthesis, only if MRI is unavailable) between ages 25 and 29 years and annual mammogram (with consideration of tomosynthesis and breast MRI screening with contrast) between ages 30 and 75 years.
Because of the relative insensitivity of mammography in women with an inherited risk of breast cancer, a number of screening modalities have been proposed and investigated in high-risk women, including carriers of BRCA pathogenic variants. Many studies have described the experience with breast MRI screening in women at risk of breast cancer, including descriptions of relatively large multi-institutional trials.
Despite some limitations of these studies, they consistently demonstrate that breast MRI is more sensitive than either mammography or ultrasound for the detection of hereditary breast cancer. The results of six large studies are presented in Table 11, Summary of MRI Screening Studies in Women at Hereditary Risk of Breast Cancer.
Most cancers in these programs were screen detected, with only 6% of cancers presenting in the interval between screenings. The sensitivity of MRI (as defined by the study methodology) ranged from 71% to 100%. Of the combined studies, 77% of cancers were identified by MRI, and 42% were identified by mammography.
Concerns have been raised about the reduced specificity of MRI compared with other screening modalities. In one study, after the initial MRI screen, 16.5% of patients were recalled for further evaluation and an additional 7.6% of patients were recommended to undergo a short-interval follow-up examination at 6 months.
These rates declined significantly during later screening rounds, with fewer than 10% of the subjects recalled for more detailed MRI and fewer than 3% recommended to have short interval follow-up. In a second study, Magnetic Resonance Imaging for Breast Screening (MARIBS), the recall rate for additional evaluation was 10.7% per year.
The benign biopsy rates in the first study were 11% at first round, 6.6% at second round, and 4.7% at third round.
In the MARIBS study, the aggregate surgical biopsy rate was 9 per 1,000 screening episodes, though this may underestimate the burden because follow-up ultrasounds, core-needle biopsies, and fine-needle aspirations have not been included in the numerator of the MARIBS calculation.
The PPV of MRI has been calculated differently in the various series and fluctuates somewhat, depending on whether all abnormal examinations or only the examinations that result in a biopsy are counted in the denominator. Generally, the PPV of a recommendation for tissue sampling (as opposed to further investigation) is in the range of 50% in most series.
These trials appear to establish that MRI is superior to mammography in the detection of hereditary breast cancer, and that women participating in these trials including annual MRI screening were less likely to have a cancer missed by screening.
However, mammography may identify some cancers, particularly DCIS, that are not identified by MRI.
Regarding downstaging, one screening study demonstrated that patients at risk of hereditary breast cancer were more likely to be diagnosed with small tumors and node-negative disease than were women in two nonrandomized control groups.
Despite the apparent sensitivity of MRI screening, some women in MRI-based programs will develop life-threatening breast cancer. In a prospective study of 51 carriers of BRCA1 pathogenic variants and 41 carriers of BRCA2 pathogenic variants screened with yearly mammograms and MRIs (of whom 80 had risk-reducing oophorectomy), 11 breast cancers (9 invasive and 2 DCIS) were detected. Six cancers were first detected on MRI; three were first detected by mammogram; and two were interval cancers. All breast cancers occurred in carriers of BRCA1 pathogenic variants, suggesting a continued high risk of BRCA1-related breast cancer after oophorectomy in the short term. These results suggest that surveillance and prevention strategies may have differing outcomes in carriers of BRCA1 and BRCA2 pathogenic variants.
A publication combining results from three large studies (MARIBS, a Canadian study, and a Dutch MRI screening study) demonstrated that when MRI was added to mammography, 80% of cancers detected in carriers of BRCA2 pathogenic variants were either DCIS or invasive cancers smaller than 1 cm. In carriers of BRCA1 pathogenic variants, 49% of cancers were DCIS or small invasive cancers. In addition, the authors predicted mortality benefits with the addition of MRI for both carriers of BRCA1 and BRCA2 pathogenic variants. The model predicted breast cancer mortality reductions of 42% to 47% for mammography, 48% to 61% for MRI, and 50% to 62% for combined screening.
An additional study examining carriers of BRCA1/BRCA2 pathogenic variants undergoing MRI between 1997 and 2006 has demonstrated that 97% of incident cancers were stage 0 or stage I.
A 2015 Dutch case-control study further evaluated 2,308 high-risk patients, including 706 women with known BRCA pathogenic variants, who were screened with mammogram and compared them with those who had the addition of MRI.
Of the patients screened, 93 patients were detected to have 97 cancers, 33 patients had a BRCA1 pathogenic variant, and 18 patients had a BRCA2 pathogenic variant. With a median follow-up of 9 years, metastases-free survival was improved in the MRI-screened cohort (90% vs. 77%), but it did not reach statistical significance in the BRCA1 and BRCA2 subset because of very small numbers. MRI-screened patients in the entire cohort were more likely to be node-negative and receive less chemotherapy. The American Cancer Society and NCCN have recommended the use of annual MRI screening for women at hereditary risk of breast cancer.
An additional question regarding the timing of mammography and MRI is whether they should be done simultaneously or in an alternating fashion (so that while each test is done annually, screening occurs every 6 months). One study has suggested that the most cost-effective screening strategy in carriers of BRCA1 and BRCA2 pathogenic variants may be annual MRI beginning at age 25 years, with alternating MRI and digital mammography beginning at age 30 years.
In summary, evidence strongly supports the integral role of breast MRI in breast cancer surveillance for carriers of BRCA1/BRCA2 pathogenic variants.
Series | Rijnsburger | Warner | MARIBS | Kuhl | Weinstein | Sardanelli | Totals | |
---|---|---|---|---|---|---|---|---|
N Patients | Overall | 2,157 | 236 | 649 | 687 | 609 | 501 | 4,839 |
BRCA1/BRCA2 Carriers | 594 | 236 | 120 | 65 | 44 | 330 | 1,389 | |
N Screening Episodes | 6,253 | 457 | 1,881 | 1,679 | 1,592 | 11,862 | ||
N Cancers | Baseline | 22 | 13 | 20 | 10 | 0 | 0 | 65 |
Subsequent | 97 | 9 | 15 | 17 | 18 | 52 | 208 | |
Invasive | 78 | 16 | 29 | 8 | 11 | 44 | 186 | |
In situ | 19 | 9 | 6 | 9 | 7 | 8 | 58 | |
Annual Incidence | 10.4/1,000 | 19/1,000 | ||||||
Detected at Planned Screening | 78 | 21 | 33 | 27 | 18 | 49 | 226 (83%) | |
N Detected by Each Modality | Mammography | 31 | 8 | 14 | 9 | 7 | 25 | 94 (42%) |
MRI | 51 | 17 | 27 | 25 | 12 | 42 | 174 (77%) | |
Ultrasound | 7 | 10 | 3 | 26 | 46 (41%) | |||
Follow-up | Median of 4.9 y | Minimum of 1 y | 2–7 y | Median of 29.09 mo | 2 y | 3 y | ||
aBased on the first 1,909 women screened. | ||||||||
bIncludes patients with invasive cancer only and patients with both invasive and in situ cancers. | ||||||||
cIncludes only 75 cancers detected in women who underwent both mammographic and MRI screening. | ||||||||
dRestricted to studies in which ultrasound was performed. |
In situ
Several studies have reported instances of breast cancer detected by ultrasound that were missed by mammography, as discussed in one review.
In a pilot study of ultrasound as an adjunct to mammography in 149 women with moderately increased risk based on family history, one cancer was detected, based on ultrasound findings. Nine other biopsies of benign lesions were performed. One was based on abnormalities on both mammography and ultrasound, and the remaining eight were based on abnormalities on ultrasound alone.
A large study of 2,809 women with dense breast tissue (ACRIN-6666) demonstrated that ultrasound increased the detection rate due to breast cancer screening from 7.6 per 1,000 with mammography alone to 11.8 per 1,000 for combined mammography and ultrasound.
However, ultrasound screening increases false-positive rates and appears to have a limited benefit in combination with MRI. In a multicenter study of 171 women (92% of whom were carriers of BRCA1/BRCA2 pathogenic variants) undergoing simultaneous mammography, MRI, and ultrasound, no cancers were detected by ultrasound alone.
Uncertainties about ultrasound include the effect of screening on mortality, the rate and outcome of false-positive results, and access to experienced breast ultrasonographers.
Level of evidence: None assigned
A number of other techniques are under active investigation, including tomosynthesis, contrast-enhanced mammography, thermography, and radionuclide scanning. Additional evidence is needed before these techniques can be incorporated into clinical practice.
Level of evidence: None assigned
Risk-reducing mastectomy (RRM) is a management option for patients who are considered to be at high risk of developing breast cancer. The Society of Surgical Oncology has endorsed RRM as an option for women with BRCA1/BRCA2 pathogenic variants or strong family histories of breast cancer.
Historically, a total or simple mastectomy has been performed, which includes removal of all of the breast tissue, including the nipple and areolar complex (NAC). If the patient is interested, reconstruction can be performed simultaneously with the ablative portion of the procedure. Options for reconstruction include tissue expander and implant-based reconstructions or autologous reconstructions, in which the patient’s own tissue is used to reconstruct the breast. A number of different tissues can be used to reconstruct the breast, including flaps based on the latissimus dorsi muscle, the transverse rectus abdominis muscle, or the gluteus muscle. Muscle-sparing techniques such as the deep inferior epigastric perforator flap can also be used, but require advanced microvascular techniques. In the interest of improved cosmetic outcomes, skin-sparing techniques have been utilized in which the entire breast is removed with the NAC, but the entire skin envelope of the breast is preserved. In a further refinement, nipple-sparing techniques have been developed in which all of the breast skin and the nipple are preserved while the underlying glandular tissue is removed.
Because there are no randomized, prospective trials of RRM versus observation, data are limited to cohort and case-control studies. The available data demonstrate that RRM does decrease breast cancer incidence in high-risk patients,
but overall survival (OS) correlates more closely with the overall risk from the primary incidence of breast cancer. Several studies have analyzed the impact of RRM on breast cancer risk and mortality. In one retrospective cohort study of 214 women considered to be at hereditary risk by virtue of a family history suggesting an autosomal dominant predisposition, three women were diagnosed with breast cancer after bilateral RRM, with a median follow-up of 14 years.
Because 37.4 cancers were expected, the calculated risk reduction was 92.0% (95% CI, 76.6%–98.3%). In a follow-up subset analysis, 176 of the 214 high-risk women in this cohort study underwent genetic testing for pathogenic variants in BRCA1 and BRCA2. Pathogenic variants were identified in 18 women, none of whom developed breast cancer after a median follow-up of 13.4 years.
Two of the three women diagnosed with breast cancer after RRM were tested, and neither carried a pathogenic variant. The calculated risk reduction among carriers of pathogenic variants was 89.5% to 100.0% (95% CI, 41.4%–100.0%), depending on the assumptions made about the expected numbers of cancers among carriers of pathogenic variants and the status of the untested woman who developed cancer despite mastectomy. The result of this retrospective cohort study has been supported by a prospective analysis of 76 carriers of pathogenic variants who underwent RRM and were monitored prospectively for a mean of 2.9 years. No breast cancers were observed in these women, whereas eight were identified in women who underwent regular surveillance (HR for breast cancer after RRM, 0.00 [95% CI, 0.00–0.36]).
The Prevention and Observation of Surgical Endpoints study group also estimated the degree of breast cancer risk reduction after RRM in carriers of BRCA1/BRCA2 pathogenic variants. The rate of breast cancer in 105 carriers of pathogenic variants who underwent bilateral RRM was compared with that in 378 carriers who did not choose surgery. Bilateral mastectomy reduced the risk of breast cancer by approximately 90% after a mean follow-up of 6.4 years.
Theoretical models have also been utilized to assess the role of RRM in women with pathogenic variants in BRCA1 and BRCA2. Assuming risk reduction in the range of 90%, one model suggests that for a group of women, aged 30 years, with BRCA1 or BRCA2 pathogenic variants, RRM would result in an average increased life expectancy of 2.9 to 5.3 years.
A computer-simulated survival analysis using a Monte Carlo model included breast MRI, mammography, RRM, and risk-reducing salpingo-oophorectomy (RRSO) and examined the impact of each intervention separately on carriers of BRCA1 and BRCA2 pathogenic variants.
The most effective strategy was found to be RRSO at age 40 years and RRM at age 25 years, with survival at age 70 years approaching that of the general population. However, delaying mastectomy until age 40 years, or substituting RRM with screening with breast MRI and mammography, had little impact on survival estimates. For example, replacing RRM with MRI-based screening in women with RRSO at age 40 years led to a 3% to 5% decrement in survival compared with RRM at age 25 years.
As with any models, numerous assumptions cause uncertainty; however, these studies provide additional information for women and their providers who are making these difficult decisions.
Another study of at-risk women showed a 70% time–trade-off value for RRM, indicating that participants were willing to sacrifice 30% of life expectancy to avoid RRM.
A cost-effectiveness analysis study of RRM has also been performed. The investigators concluded that, compared with surveillance, risk-reducing surgery (mastectomy and oophorectomy) is cost-effective with regard to years of life saved, but not for improved quality of life.
While these data are interesting and may be useful for public policy decisions, they cannot be individualized for clinical care because they include assumptions that cannot be fully tested.
If RRM is effective in lowering breast cancer risk in unaffected women, what is its role for women with unilateral breast cancer? This question often arises in discussions about surgical options with women who have unilateral breast cancer and hereditary risks. This section addresses the role of contralateral risk-reducing mastectomy (CRRM) in women being treated with mastectomy and will not discuss breast conservation therapy. Multiple studies have shown an increase in the rate of CRRM in women with unilateral breast cancer.
When the appropriateness of CRRM is being assessed for women with unilateral breast cancer, the first task is to determine the risk of contralateral breast cancer (CBC).
In the general population, current estimates of CBC risk after treatment for breast cancer are approximately 0.3% per year and are declining.
In carriers of BRCA pathogenic variants with a diagnosis of breast cancer, the risk of a second, unrelated breast cancer is related to age at initial diagnosis, biology, and systemic therapies used, but is clearly higher than that in the general population.
(Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section in the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section of this summary for more information about the risk of CBC in this population.) In carriers of BRCA pathogenic variants whose first cancer has an excellent prognosis, estimating the risk of a second, unrelated breast cancer event is important for informing their decision to undergo risk-reducing surgery and has been described in this setting to improve survival.
The timing of genetic testing and knowledge of BRCA pathogenic variant status may influence surgical decision making, may prevent subsequent surgeries, and may influence follow-up care. Therefore, for individuals at increased risk of carrying a BRCA pathogenic variant, it is important that genetic testing be considered in advance of surgery, when possible.
In a group of 148 carriers of BRCA1 and BRCA2 pathogenic variants with unilateral breast cancer, 79 of whom underwent CRRM, the risk of CBC was reduced by 91% and was independent of the effect of risk-reducing oophorectomy. Survival was better among women who underwent CRRM, but this result was likely associated with higher mortality caused by the index cancer or metachronous ovarian cancer in the group not undergoing surgery.
Data from ten European centers on 550 women (including 202 BRCA carriers) with 3,334 woman-years of follow-up indicated that RRM was highly effective. Bilateral RRMs were carried out on women with a lifetime risk of 25% to 80%, with an average expected incidence rate of 1% per year. No breast cancers occurred in this cohort over the follow-up period, though more than 34 breast cancers would have been expected.
A retrospective study of 593 carriers of BRCA1 and BRCA2 pathogenic variants included 105 women with unilateral breast cancer who underwent CRRM and had a 10-year survival rate of 89%, compared with 71% in the group who did not undergo contralateral risk-reducing surgery (P < .001).
This study was limited by several factors, such as the lack of information regarding breast cancer screening, grade, and estrogen receptor status in a large portion of this sample.
A Dutch cohort of 583 patients identified between 1980 and 2011, who had both a BRCA pathogenic variant and a diagnosis of unilateral breast cancer, were evaluated for the effect of CRRM.
With a median follow-up of 11.4 years, 242 (42%) of the patients underwent RRM (193 carriers of BRCA1 pathogenic variants and 49 carriers of BRCA2 pathogenic variants) at differing times after their diagnoses. Improved OS was observed in the RRM group compared with the surveillance group (HR, 0.49; 95% CI, 0.29–0.82), with improvements most pronounced in those diagnosed before age 40 years, with low tumor grade, and non–triple-negative subtype. In an attempt to control for the bias of time to surgery, the authors included a separate evaluation of women who were known to be disease free 2 years after the primary cancer diagnosis (HR, 0.55; 95% CI, 0.32–0.95). Additionally, the group who underwent RRM was more likely to undergo bilateral salpingo-oophorectomy and systemic chemotherapy, which may influence the significance of these survival findings.
A retrospective study of 390 women with early-stage breast cancer who were from families with a known BRCA1/BRCA2 pathogenic variant found a significant improvement in survival for women who underwent bilateral mastectomy compared with those who chose unilateral mastectomy.
Patients were followed for a median of 14.3 years (range, 0.1–20.0 y). A multivariate analysis controlling for age at diagnosis, year of diagnosis, treatment, and other prognostic factors found that CRRM was associated with a 48% reduction in death from breast cancer. This was a relatively small study, and although the authors adjusted for multiple factors, residual confounding factors may have influenced the results.
All of these studies are limited by the biases introduced in relatively small, retrospective studies among very select populations. There is often limited data on potential confounding variables such as socioeconomic status, comorbidities, and access to care. It has been suggested that women who elect to undergo RRM are healthier by virtue of being able to tolerate more extensive surgery. This theory is supported by one study that used Surveillance, Epidemiology, and End Results (SEER) Program data to examine the association between CRRM and outcomes among women with unilateral breast cancer stages I through III. Results showed a reduction in all-cause mortality and breast cancer–specific mortality, and also in noncancer event mortality, a finding that would not be expected to be related to CRRM.
The option of nipple-sparing mastectomy (NSM) in carriers of BRCA pathogenic variants undergoing risk-reducing procedures has been controversial because of concerns about increased breast tissue left behind at surgery to keep the NAC viable. The ability to leave behind minimal residual tissue, however, may be related to experience and technique. In a retrospective review of NSM performed in carriers of BRCA pathogenic variants at two hospitals between 2007 and 2014, NSM was performed on 397 breasts in 201 carriers of BRCA pathogenic variants.
This study included both unaffected and affected women. Incidental cancers were found in 4 of 150 RRM patients (2.7%) and 2 of 51 cancer patients (3.9%). With a mean follow-up of 32.6 months (range, 1.0–76.0 months), there were four subsequent cancer events that included two patients with axillary recurrences, one with a local and distant recurrence 11 months after her original NSM, and one patient who developed a new cancer in the inferior portion of her breast, with no recurrences at the NAC. A study of 177 NSMs performed in 89 carriers of BRCA pathogenic variants between 2005 and 2013 reported similar, excellent local control rates. Sixty-three patients had risk-reducing NSM (median follow-up, 26 months; range, 11–42 months), and 26 patients had NSM and a diagnosis of breast cancer (median follow-up, 28 months; range, 15–43 months). Five patients required further nipple excision. There were no local recurrences or newly diagnosed breast cancers.
Studies describing histopathologic findings in RRM specimens from women with BRCA1 or BRCA2 pathogenic variants have been somewhat inconsistent. In two series, proliferative lesions associated with an increased risk of breast cancer (lobular carcinoma in situ, atypical lobular hyperplasia, atypical ductal hyperplasia, and DCIS) were noted in 37% to 46% of women with pathogenic variants who underwent either unilateral or bilateral RRM.
In these series, 13% to 15% of patients were found to have previously unsuspected DCIS in the prophylactically removed breast. Among 47 cases of risk-reducing bilateral or contralateral mastectomies performed in known carriers of BRCA1 or BRCA2 pathogenic variants from Australia, three (6%) cancers were detected at surgery.
In general, histopathologic findings in RRM specimens do not impact management.
Individual psychological factors play an important role in decision-making about RRM by unaffected women and CRRM in women with unilateral breast cancer. (Refer to the Psychosocial Aspects of Cancer Risk Management for Hereditary Breast and Ovarian Cancer section in the Psychosocial Issues in Inherited Breast and Ovarian Cancer Syndromes section of this summary for information about uptake of RRM in BRCA carriers and the Psychosocial Outcome Studies section for information about psychosocial outcomes of RRM.)
In summary:
In the general population, removal of both ovaries has been associated with a reduction in breast cancer risk of up to 75%, depending on parity, weight, and age at time of artificial menopause. (Refer to the PDQ summary on Breast Cancer Prevention for more information.) A Mayo Clinic study of 680 women at various levels of familial risk found that in women younger than 60 years who had bilateral oophorectomy, the likelihood of breast cancers developing was reduced for all risk groups.
Ovarian ablation, however, is associated with important side effects such as hot flashes, impaired sleep habits, vaginal dryness, dyspareunia, and increased risk of osteoporosis and heart disease. A variety of strategies may be necessary to counteract the adverse effects of ovarian ablation.
The evidence for the effect of RRSO on breast cancer has evolved. Early small studies suggested a protective benefit. Initial retrospective studies supported breast cancer and ovarian cancer risk reduction after RRSO in BRCA pathogenic variant–positive women.
In support of early small studies,
a retrospective study of 551 women with disease-associated BRCA1 or BRCA2 variants found a significant reduction in risk of breast cancer (HR, 0.47; 95% CI, 0.29–0.77) and ovarian cancer (HR, 0.04; 95% CI, 0.01–0.16) after RRSO.
A prospective, single-institution study of 170 women with BRCA1 or BRCA2 pathogenic variants showed a similar trend. With RRSO, the HR was 0.15 (95% CI, 0.02–1.31) for ovarian, fallopian tube, or primary peritoneal cancer, and 0.32 (95% CI, 0.08–1.2) for breast cancer; the HR for either cancer was 0.25 (95% CI, 0.08–0.74).
A prospective, multicenter study of 1,079 women followed up for a median of 30 to 35 months found that while RRSO was associated with reductions in breast cancer risk in both carriers of BRCA1 and BRCA2 pathogenic variants, the risk reduction was more pronounced in BRCA2 carriers (HR, 0.28; 95% CI, 0.08–0.92).
A meta-analysis of all reports of RRSO and breast and ovarian/fallopian tube cancer in carriers of BRCA1/BRCA2 pathogenic variants confirmed that RRSO was associated with a significant reduction in breast cancer risk (overall: HR, 0.49; 95% CI, 0.37–0.65; BRCA1: HR, 0.47; 95% CI, 0.35–0.64; BRCA2: HR, 0.47; 95% CI, 0.26–0.84).
However, a cohort study of 822 carriers of BRCA1/BRCA2 pathogenic variants conducted in the Netherlands, where carrier screening is performed nationwide, did not observe a reduced risk of breast cancer after RRSO (HR, 1.09; 95% CI, 0.67–1.77).
The authors argued that the previous findings were driven by methodological issues including cancer-induced testing bias and immortal person time, and empirically evaluated this by using their own cohort and applying the same assumptions about counting person time from previous studies.
In a response, investigators from the U.S. studies analyzed their data using the assumptions of the Dutch study but still observed an inverse association with RRSO and breast cancer risk.
In a retrospective cohort of 676 women, carriers having an RRSO at the time of breast cancer diagnosis had a reduced risk of breast cancer–specific mortality (HR, 0.38; 95% CI, 0.19–0.77 for BRCA1 carriers and HR, 0.57; 95% CI, 0.23–1.43 for BRCA2 carriers).
A subsequent international, multi-institutional study of 3,722 BRCA1 and BRCA2 carriers using a similar methodology showed that oophorectomy performed before age 50 years was beneficial for preventing breast cancer in BRCA2 carriers (HR, 0.18; 95% CI, 0.05–0.63; P = .007) but not in BRCA1 carriers.
Given these conflicting findings, an additional cohort of 17,917 women who participated in the Prospective Family Study Cohort was assessed with 1,046 women diagnosed with breast cancer and a median follow-up of 10.7 years; no association between RRSO and breast cancer was observed.
A prospective, multicenter, cohort study of 2,482 carriers of BRCA1/BRCA2 pathogenic variants has reported an association of RRSO with a reduction in all-cause mortality (HR, 0.40; 95% CI, 0.26–0.61), breast cancer–specific mortality (HR, 0.44; 95% CI, 0.26–0.76), and ovarian cancer–specific mortality (HR, 0.21; 95% CI, 0.06–0.80).
A subsequent meta-analysis confirmed the impact of RRSO on all-cause mortality (HR, 0.32; 95% CI, 0.27–0.38) in carriers of BRCA1 and BRCA2 pathogenic variants, including those with and without a personal history of breast cancer.
Despite discordant findings regarding RRSO and breast cancer risk in the existing literature, aggregate data suggest that there is a benefit, although the magnitude of this benefit may not be fully understood. Further prospective studies are needed to confirm these findings.
Refer to the RRSO section in the Ovarian cancer section of this summary for more information about the effect of RRSO on ovarian cancer risk in carriers of BRCA pathogenic variants.
Tamoxifen (a synthetic antiestrogen) increases breast-cell growth inhibitory factors and concomitantly reduces breast-cell growth stimulatory factors. The National Surgical Adjuvant Breast and Bowel Project Breast Cancer Prevention Trial (NSABP-P-1), a prospective, randomized, double-blind trial, compared tamoxifen (20 mg/day) with placebo for 5 years in high-risk women (defined by a Gail model risk score >1.66, age >60 y, or lobular carcinoma in situ). Tamoxifen was shown to reduce the risk of invasive breast cancer by 49%. The protective effect was largely confined to ER-positive breast cancer, which was reduced by 69%. The incidence of ER-negative cancer was not significantly reduced.
Similar reductions were noted in the risk of preinvasive breast cancer. Reductions in breast cancer risk were noted both among women with a family history of breast cancer and in those without a family history. An increased incidence of endometrial cancers and thrombotic events occurred among women older than 50 years. Interim data from two European tamoxifen prevention trials did not show a reduction in breast cancer risk with tamoxifen after a median follow-up of 48 months or 70 months,
respectively. In one trial, however, reduction in breast cancer risk was seen among a subgroup who also used hormone replacement therapy (HRT).
These trials varied considerably in study design and populations. (Refer to the PDQ summary on Breast Cancer Prevention for more information.)
Subsequently, the International Breast Cancer Intervention Study 1 (IBIS-1) breast cancer prevention trial randomly assigned 7,154 women between the ages of 35 and 70 years to receive tamoxifen or placebo for 5 years. Eligibility for the trial was based on family history or abnormal benign breast disease. At a median follow-up of 16 years, there was a 29% reduction in risk of breast cancer in the tamoxifen arm (HR, 0.71; 95% CI, 0.60–0.83). There was a 43% reduction in risk for invasive ER-positive breast cancer (HR, 0.66; 95% CI, 0.54–0.81) and a 35% reduction in risk for DCIS (HR, 0.65; 95% CI 0.43–1.00). There was no reduction in risk of invasive ER-negative breast cancer.
These findings confirm those of the Breast Cancer Prevention Trial (P-1).
A substudy of the NSABP-P-1 trial evaluated the effectiveness of tamoxifen in preventing breast cancer in carriers of BRCA1/BRCA2 pathogenic variants older than 35 years. BRCA2-positive women benefited from tamoxifen to the same extent as BRCA1/BRCA2 pathogenic variant–negative participants; however, tamoxifen use among healthy women with BRCA1 pathogenic variants did not appear to reduce breast cancer incidence. These data must be viewed with caution in view of the small number of carriers of pathogenic variants in the sample (8 BRCA1 carriers and 11 BRCA2 carriers).
In contrast to the very limited data on primary prevention in carriers of BRCA1 and BRCA2 pathogenic variants with tamoxifen, several studies have found a protective effect of tamoxifen on the risk of contralateral breast cancer.
In one study involving approximately 600 carriers of BRCA1/BRCA2 pathogenic variants, tamoxifen use was associated with a 51% reduction in contralateral breast cancer.
An update to this report examined 285 carriers of BRCA1/BRCA2 pathogenic variants with bilateral breast cancer and 751 carriers of BRCA1/BRCA2 pathogenic variants with unilateral breast cancer (40% of these patients were included in their initial study). Tamoxifen was associated with a 50% reduction in contralateral breast cancer risk in carriers of BRCA1 pathogenic variants and a 58% reduction in carriers of BRCA2 pathogenic variants. Tamoxifen did not appear to confer benefit in women who had undergone an oophorectomy, although the numbers in this subgroup were quite small.
Another study that involved 160 carriers of BRCA1/BRCA2 pathogenic variants demonstrated that tamoxifen use after the treatment of breast cancer with lumpectomy and radiation was associated with a 69% reduction in the risk of contralateral breast cancer.
In another study, 2,464 carriers of BRCA1/BRCA2 pathogenic variants with a personal history of breast cancer were identified from three family cohorts. Using both retrospective and prospective data, researchers found a significant decrease in the risk of contralateral breast cancer among women who received adjuvant tamoxifen therapy after their diagnosis. This association persisted after researchers adjusted for age at diagnosis and the ER status of the first cancer. A major limitation of this study is the lack of information on ER status of the first breast cancer in 56% of the women.
These studies are limited by their retrospective, case-control designs and the absence of information regarding ER status in the primary tumor.
The STAR trial (NSABP-P-2) included more than 19,000 women and compared 5 years of raloxifene versus tamoxifen in reducing the risk of invasive breast cancer.
There was no difference in incidence of invasive breast cancer at a mean follow-up of 3.9 years; however, there were fewer noninvasive cancers in the tamoxifen group. The incidence of thromboembolic events and hysterectomy was significantly lower in the raloxifene group. Detailed quality-of-life data demonstrate slight differences between the two arms.
Data regarding efficacy in carriers of BRCA1 or BRCA2 pathogenic variants are not available. (Refer to the PDQ summary on Breast Cancer Prevention for more information about the use of selective ER modulators and aromatase inhibitors in the general population, including postmenopausal women.)
Another case-control study of carriers of pathogenic variants and noncarriers identified through ascertainment of women with bilateral breast cancer found that systemic adjuvant chemotherapy reduced CBC risk among carriers of pathogenic variants (RR, 0.5; 95% CI, 0.2–1.0). Tamoxifen was associated with a nonsignificant risk reduction (RR, 0.7; 95% CI, 0.3–1.8). Similar risk reduction was seen in noncarriers; however, given the higher absolute CBC risk in carriers, there is potentially a greater impact of adjuvant treatment in risk reduction.
The effect of tamoxifen on ovarian cancer risk was studied in 714 carriers of BRCA1 pathogenic variants. All subjects had a prior history of breast cancer; use of tamoxifen was not associated with an increased risk of subsequent ovarian cancer (odds ratio [OR], 0.78; 95% CI, 0.46–1.33).
In the general population, breast cancer risk increases with early menarche and late menopause, and is reduced at early first full-term pregnancy. (Refer to the PDQ summary on Breast Cancer Prevention for more information.) In the Nurses’ Health Study, these were risk factors among women who did not have a mother or sister with breast cancer.
Among women with a family history of breast cancer, pregnancy at any age appeared to be associated with an increase in risk of breast cancer, persisting to age 70 years.
One study evaluated risk modifiers among 333 female carriers of a BRCA1 high-risk pathogenic variant. In women with known pathogenic variants of the BRCA1 gene, early age at first live birth and parity of three or more have been associated with a lowered risk of breast cancer. A RR of 0.85 was estimated for each additional birth, up to five or more; however, increasing parity appeared to be associated with an increased risk of ovarian cancer.
In a case-control study from New Zealand, investigators noted no difference in the impact of parity on the risk of breast cancer between women with a family history of breast cancer and those without a family history.
Studies of the effect of pregnancy on breast cancer risk have revealed complex results and the relationship with parity has been inconsistent and may vary between carriers of BRCA1 and BRCA2 pathogenic variants.
Parity has more consistently been associated with a reduced risk of breast cancer in carriers of BRCA1 pathogenic variants.
Of note, neither therapeutic nor spontaneous abortions appear to be associated with an increased breast cancer risk.
In the general population, breastfeeding has been associated with a slight reduction in breast cancer risk in a few studies, including a large collaborative reanalysis of multiple epidemiologic studies,
and at least one study suggests that it may be protective in carriers of BRCA1 pathogenic variants. In a multicenter, case-control study of 685 carriers of BRCA1 pathogenic variants with breast cancer and 280 carriers of BRCA2 pathogenic variants with breast cancer and 965 carriers without breast cancer drawn from multiple-case families, among carriers of BRCA1 pathogenic variants, breastfeeding for 1 year or more was associated with approximately a 45% reduced risk of breast cancer.
No such reduced risk was observed among carriers of BRCA2 pathogenic variants. A second study failed to confirm this association.
There is no consistent evidence that the use of oral contraceptives (OCs) increases the risk of breast cancer in the general population.
(Refer to the PDQ summary on Breast Cancer Prevention for more information.)
Although several smaller studies have reported a slightly increased risk of breast cancer with OC use in carriers of BRCA1/BRCA2 pathogenic variants,
a meta-analysis concluded that the associated risk is not significant with more recent OC formulations.
However, OCs formulated before 1975 were associated with an increased risk of breast cancer.
A large proportion of patients on whom this meta-analysis was based were drawn from three large studies summarized in Table 12.
Kotsopoulos et al. (2014)a | Brohet et al. (2007)b | Haile et al. (2006)a,c | Narod et al. (2002)a | ||
---|---|---|---|---|---|
Study population | BRCA1 carriers with breast cancer | N = 2,492 | N = 597 | N = 195; diagnosis < age 50 y | N = 981 |
BRCA2 carriers with breast cancer | Not applicable | N = 249 | N = 128; diagnosis < age 50 y | N = 330 | |
Ever use OC | BRCA1 | 1.18 [CI 1.03–1.36] P = .02 | 1.47 [CI 1.13–1.91] | 0.64 [CI 0.35–1.16] | 1.38 [CI 1.11–1.72] P = .003 |
BRCA2 | Not applicable | 1.49 [Cl 0.8–2.7] | 1.29 [Cl 0.61–2.76] | 0.94 [Cl 0.72–1.24] | |
Age use <20 y | BRCA1 | 1.45 [CI 1.20–1.75] P = .0001 | 1.41 [Cl 0.99–2.01] | 0.84 [Cl 0.45–1.55] | 1.36 [Cl 1.11–1.67] P = .003 |
BRCA2 | Not applicable | 1.25 [Cl 0.57–2.74] | 1.64 [Cl 0.77–3.46] | Not reported | |
Total duration | BRCA1 | <5 y: 1.14 [CI 0.97–1.35] | <9 y: 1.51 [Cl 1.1–2.08] | <5 y: 0.61 [Cl 0.31–1.17] | <10 y: 1.36 [Cl 1.11–1.67] P = .003 |
>5 y: 1.22 [CI 1.04–1.49] P = .02 | |||||
BRCA2 | Not applicable | <9 y: 2.27 [Cl 1.1–4.65] | <5 y: 0.79 [Cl 0.26–2.37] | <10 y: 0.82 [Cl 0.56–1.91] | |
Use before full-term pregnancy | BRCA1 | Not applicable | >4 y: 1.49 [Cl 1.05–2.11] | >4 y: 0.69 [Cl 0.41–1.16] | Not evaluated |
BRCA2 | Not applicable | >4 y: 2.58 [Cl 1.21–5.49] | >4 y: 2.08 [Cl 1.02–4.25] trend per y: 1.11; P trend = .01 | ||
CI = confidence interval. | |||||
aReports risk estimates in the form of odds ratios with 95% CIs. | |||||
bReports risk estimates in the form of hazard ratios with 95% CIs. | |||||
cRisk estimates restricted to carriers of BRCA pathogenic variants younger than 40 years. |
When patients are counseled about contraceptive options and preventive actions, the potential impact of OC use on the risk of breast cancer and ovarian cancer and other health-related effects of OCs need to be considered. A number of important issues remain unresolved, including the potential differences between carriers of BRCA1 or BRCA2 pathogenic variants, effect of age and duration of exposure, and effect of OCs on families with highly penetrant early-onset breast cancer.
(Refer to the Oral contraceptives section in the Chemoprevention section of this summary for a discussion of OC use and ovarian cancer in this population.)
Both observational and randomized clinical trial data suggest an increased risk of breast cancer associated with HRT in the general population.
The Women’s Health Initiative (WHI) was a randomized controlled trial of approximately 160,000 postmenopausal women that investigated the risks and benefits of dietary interventions and hormone therapy to reduce the incidence of heart disease, breast cancer, colorectal cancer, and fractures. The estrogen-plus-progestin arm of the study, in which more than 16,000 women were randomly assigned to receive combined hormone therapy or placebo, was halted early because health risks exceeded benefits.
One of the adverse outcomes prompting closure was a significant increase in both total (245 vs. 185 cases) and invasive (199 vs. 150) breast cancers (RR, 1.24; 95% CI, 1.02–1.50; P < .001) in women randomly assigned to receive estrogen and progestin.
Results of a follow-up study suggest that the recent reduction in breast cancer incidence, especially among women aged 50 to 69 years, is predominantly related to decrease in use of combined estrogen plus progestin HRT.
HRT-related breast cancers had adverse prognostic characteristics (more advanced stages and larger tumors) compared with cancers occurring in the placebo group, and HRT was also associated with a substantial increase in abnormal mammograms.
Breast cancer risk associated with postmenopausal HRT has been variably reported to be increased or unaffected by a family history of breast cancer;
risk did not vary by family history in the meta-analysis.
The WHI study has not reported analyses stratified on breast cancer family history, and subjects have not been systematically tested for BRCA1/BRCA2 pathogenic variants.
Short-term use of hormones for treatment of menopausal symptoms appears to confer little or no breast cancer risk in the general population.
The effect of HRT on breast cancer risk among carriers of a BRCA1 or BRCA2 pathogenic variant has been examined in two studies. In a prospective study of 462 carriers of BRCA1 and BRCA2 pathogenic variants, bilateral RRSO (n = 155) was significantly associated with breast cancer risk-reduction overall (HR, 0.40; 95% CI, 0.18–0.92). When carriers of pathogenic variants without bilateral RRSO or HRT were used as the comparison group, HRT use (n = 93) did not significantly alter the reduction in breast cancer risk associated with bilateral RRSO (HR, 0.37; 95% CI, 0.14–0.96).
In a matched case-control study of 472 postmenopausal women with BRCA1 pathogenic variants, HRT use was associated with an overall reduction in breast cancer risk (OR, 0.58; 95% CI, 0.35–0.96; P = .03). A nonsignificant reduction in risk was observed both in women who had undergone bilateral oophorectomy and in those who had not. Women taking estrogen alone had an OR of 0.51 (95% CI, 0.27–0.98; P = .04), while the association with estrogen and progesterone was not statistically significant (OR, 0.66; 95% CI, 0.34–1.27; P = .21).
A case-control study of 432 matched pairs of postmenopausal women with a BRCA1 pathogenic variant who had a personal history of cancer were compared with unaffected BRCA1 carriers. The use of HRT was not associated with an increased risk of developing breast cancer (OR, 0.80; P = .24).
Especially given the differences in estimated risk associated with HRT between observational studies and the WHI, these findings should be confirmed in randomized prospective studies,
but they suggest that HRT in carriers of BRCA1/BRCA2 pathogenic variants neither increases breast cancer risk nor negates the protective effect of oophorectomy.
Refer to the PDQ summary on Ovarian, Fallopian Tube, and Primary Peritoneal Cancer Screening for information on screening in the general population and to the PDQ summary Levels of Evidence for Cancer Genetics Studies for information about levels of evidence related to screening and prevention. The latter also outlines the five requirements that must be met before it is considered appropriate to screen for a particular medical condition as part of routine medical practice.
In the general population, clinical examination of the ovaries has neither the specificity nor the sensitivity to reliably identify early ovarian cancer. No data exist regarding the benefit of clinical examination of the ovaries (bimanual pelvic examination) in women at inherited risk of ovarian cancer.
Level of evidence: None assigned
In the general population, transvaginal ultrasound (TVUS) appears to be superior to transabdominal ultrasound in the preoperative diagnosis of adnexal masses. Both techniques have lower specificity in premenopausal women than in postmenopausal women due to the cyclic menstrual changes in premenopausal ovaries (e.g., transient corpus luteum cysts) that can cause difficulty in interpretation. The randomized prospective Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO-1) found no reduction in mortality with the annual use of combined TVUS and cancer antigen 125 (CA-125) in screening asymptomatic postmenopausal women at general-population risk of ovarian cancer.
Data are limited regarding the potential benefit of TVUS in screening women at inherited risk of ovarian cancer. A number of retrospective studies have reported experience with ovarian cancer screening in high-risk women using TVUS with or without CA-125.
However, there is little uniformity in the definition of high-risk criteria and compliance with screening, and in whether cancers detected were incident or prevalent. One of the largest reported studies included 888 carriers of BRCA1/BRCA2 pathogenic variants who were screened annually with TVUS and CA-125. Ten women developed ovarian cancer; five of the ten developed interval cancers after normal screening results within 3 to 10 months before diagnosis. Five of the ten ovarian cancers were screen-detected incident cases, which had normal screening results within 6 to 14 months before diagnosis. Of these five cases, four were stage IIIB or IV.
A similar study reported the results of annual TVUS and CA-125 in a cohort of 312 high-risk women (152 carriers of BRCA1/BRCA2 pathogenic variants).
Of the four cancers that were detected due to abnormal TVUS and CA-125, all four patients were symptomatic, and three had advanced-stage disease. Annual screening of carriers of BRCA1/BRCA2 pathogenic variants with pelvic ultrasound, TVUS, and CA-125 failed to detect early-stage ovarian cancer among 241 carriers of BRCA1/BRCA2 pathogenic variants in a study from the Netherlands.
Three cancers were detected over the course of the study, all advanced stage IIIC disease.
Finally, a study of 1,100 moderate- and high-risk women who underwent annual TVUS and CA-125 reported that ten of 13 ovarian tumors were detected due to screening. Only five of ten were stage I or II.
There are limited data related to the efficacy of semiannual screening with TVUS and CA-125.
In the United Kingdom Familial Ovarian Cancer Screening Study, 3,563 women with an estimated 10% or higher lifetime risk of ovarian cancer were screened with annual ultrasound and serum CA-125 measurements for a mean of 3.2 years. Four of 13 screen-detected cancers were stage I or II. Women screened within the previous year were less likely to have higher than stage IIIC disease; there was also a trend towards better rates of optimal cytoreduction and improved OS. Furthermore, most of the cancers occurred in women with known ovarian cancer susceptibility genes, identifying a cohort at highest cancer risk for consideration of screening.
Phase II of this study increased the frequency of screening to every 4 months; the impact of this is not yet available.
The first prospective study of TVUS and CA-125 with survival as the primary outcome was completed in 2009. Of the 3,532 high-risk women screened, 981 were carriers of BRCA pathogenic variants, 49 of whom developed ovarian cancer. The 5- and 10-year survival was 58.6% (95% CI, 50.9%–66.3%) and 36% (95% CI, 27–45), respectively, and there was no difference in survival between carriers and noncarriers. A major limitation of the study was the absence of a control group. Despite limitations, this study suggests that annual surveillance by TVUS and CA-125 level appear to be ineffective in detecting tumors at an early stage to substantially influence survival.
Serum CA-125 screening for ovarian cancer in high-risk women has been evaluated in combination with TVUS in a number of retrospective studies, as described in the previous section.
The National Institutes of Health (NIH) Consensus Statement on Ovarian Cancer recommended against routine screening of the general population for ovarian cancer with serum CA-125. (Refer to the Prostate, Lung, Colorectal and Ovarian [PLCO] Cancer Screening Trial: Single-threshold CA-125 levels and TVU section in the PDQ summary on Ovarian, Fallopian Tube, and Primary Peritoneal Cancer Screening for more information.) The NIH Consensus Statement did, however, recommend that women at inherited risk of ovarian cancer undergo TVUS and serum CA-125 screening every 6 to 12 months, beginning at age 35 years.
The Cancer Genetics Studies Consortium task force has recommended that female carriers of a BRCA1 pathogenic variant undergo annual or semiannual screening using TVUS and serum CA-125 levels, beginning at age 25 to 35 years.
Both recommendations are based solely on expert opinion and best clinical judgment.
The need for effective ovarian cancer screening is particularly important for women carrying BRCA1 and BRCA2 pathogenic variants, and the mismatch repair (MMR) genes (e.g., MLH1, MSH2, MSH6, PMS2), disorders in which the risk of ovarian cancer is high. There is a special sense of urgency for carriers of BRCA1 pathogenic variants, in whom cumulative lifetime risks of ovarian cancer may exceed 40%.
Thus, it is expected that many new ovarian cancer biomarkers (either singly or in combination) will be proposed as ovarian cancer screening strategies during the next 5 to 10 years. While this is an active area of research with a number of promising new biomarkers in early development, at present, none of these biomarkers alone or in combination have been sufficiently well studied to justify their routine clinical use for screening purposes, either in the general population or in women at increased genetic risk.
Before information related to emerging ovarian cancer biomarkers is addressed, it is important to consider the several steps that are required to develop and, more importantly, validate a new biomarker. One useful framework is that published by the National Cancer Institute Early Detection Research Network investigators.
They indicated that the goal of a cancer-screening program is to detect tumors at an early stage so that treatment is likely to be successful. The gold standard by which such programs are judged is whether the death rate from the cancer for which screening is performed is reduced among those being screened. In addition, the screening test must be sufficiently noninvasive and inexpensive to allow widespread use in the population to be screened. Maintaining high test specificity (i.e., few false-positive results) is essential for a population screening test, because even a low false-positive rate results in many people having to undergo unnecessary and costly diagnostic procedures and psychological stress. It is likely that the use of several such cancer biomarkers in combination will be required for a screening test to be both sensitive and specific.
Furthermore, a clinically useful test must have a high PPV (a parameter derived from sensitivity, specificity, and disease prevalence in the screened population). Practically speaking, a biomarker with a PPV of 10% implies that ten surgical procedures would be required to identify one case of ovarian cancer; the remaining nine surgeries would represent false-positive test findings. In general, the ovarian cancer research community considers biomarkers with a PPV less than 10% to be clinically unacceptable, given the morbidity related to bilateral salpingo-oophorectomy. Finally, it is important to keep in mind that while novel biomarkers may be present in the sera of women with advanced ovarian cancer (who represent most cases analyzed in the early phases of biomarker development), they may or may not be detectable in women with early-stage disease, which is essential if the screening test is to be clinically useful.
It has been suggested that there are five general phases in biomarker development and validation are currently suggested:
Finally, for a validated biomarker test to be considered appropriate for use in a particular population, it must have been evaluated in that specific population without prior selection of known positives and negatives. In addition, the test must demonstrate clinical utility, that is, a positive net balance of benefits and risks associated with the application of the test. These may include improved health outcomes and net psychosocial and economic benefits.
Ovarian cancer poses a unique challenge relative to the potential impact of false-positive test results. There are no reliable noninvasive diagnostic tests for early-stage disease, and clinically significant early-stage cancer may not be grossly visible at the time of exploratory surgery.
Consequently, it is likely that some patients will be reassured that their abnormal test does not indicate the presence of cancer only by having their ovaries and fallopian tubes surgically removed and examined microscopically. High test specificity (i.e., a very low false-positive rate) is required to avoid unnecessary surgery and induction of premature menopause women with in false-positive results.
An ovarian cancer symptom index for predicting the presence of cancer was evaluated in 75 cases and 254 high-risk controls (carriers of BRCA pathogenic variants or women with a strong family history of breast and ovarian cancer).
Women had a positive symptom index if they reported any of the predefined symptoms (bloating or increase in abdominal size, abdominal or pelvic pain, and difficulty eating or feeling full quickly) more than 12 times per month, occurring only within the prior 12 months. CA-125 values greater than 30 U/mL were considered abnormal. The symptom index independently predicted the presence of ovarian cancer, after controlling for CA-125 levels (P < .05). The combination of an elevated CA-125 and a positive-symptom index correctly identified 89.3% of the cases. The symptom index correlated with the presence of cancer in 50% of the affected women who did not have elevated CA-125 levels, but 11.8% of the high-risk controls without cancer also had a positive-symptom index. The authors suggested that a composite index that included both CA-125 and the symptom index had better performance characteristics than either test used alone, and that this strategy might be used as a first screen in a multistep screening program. Additional test performance validation and determination of clinical utility are required in unselected screening populations.
A novel modification of CA-125 screening is based on the hypothesis that rising CA-125 levels over time may provide better ovarian cancer screening performance characteristics than simply classifying CA-125 as normal or abnormal based on an arbitrary cut-off value. This has been implemented in the form of the risk of ovarian cancer algorithm (ROCA), an investigational statistical model that incorporates serial CA-125 test results and other covariates into a computation that produces an estimate of the likelihood that ovarian cancer is present in the screened subject. The first report of this strategy, based on reanalysis of 5,550 average-risk women from the Stockholm Ovarian Cancer screening trial, suggested that ovarian cancer cases and controls could be distinguished with 99.7% sensitivity, 83% specificity, and a PPV of 16%. That PPV represents an eightfold increase over the 2% PPV reported with a single measure of CA-125.
This report was followed by applying the ROCA to 33,621 serial CA-125 values obtained from the 9,233 average-risk postmenopausal women in a prospective British ovarian cancer screening trial.
The area under the receiver operator curve increased from 84% to 93% (P = .01) for ROCA compared with a fixed CA-125 cutoff. These observations represented the first evidence that preclinical detection of ovarian cancer might be improved using this screening strategy. A prospective study of 13,000 normal volunteers aged 50 years and older in England used serial CA-125 values and the ROCA to stratify participants into low, intermediate, and elevated risk subgroups.
Each had its own prescribed management strategy, including TVUS and repeat CA-125 either annually (low risk) or at 3 months (intermediate risk). Using this protocol, ROCA was found to have a specificity of 99.8% and a PPV of 19%.
Two prospective trials in England utilized the ROCA. The United Kingdom Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) randomly assigned normal-risk women to either (1) no screening, (2) annual ultrasound, or (3) multimodal screening (N = 202,638; accrual completed; follow-up ends in 2014), and the U.K. Familial Ovarian Cancer Screening Study (UKFOCSS) targeted high-risk women (accrual completed). There are also two high-risk cohorts using the ROCA under evaluation in the United States: the Cancer Genetics Network ROCA Study (N = 2,500; follow-up complete; analysis underway) and the Gynecologic Oncology Group Protocol 199 (GOG-0199; enrollment complete; follow-up ended in 2011).
Thus, additional data regarding the utility of this currently investigational screening strategy will become available.
A wide array of new candidate ovarian cancer biomarkers has been described during the past decade, e.g., HE4; mesothelin; kallikreins 6, 10, and 11; osteopontin; prostasin; M-CSF; OVX1; lysophosphatidic acid; vascular endothelial growth factor B7-H4; and interleukins 6 and 8.
These have been singly studied, in combination with CA-125, or in various other permutations. Most of the study populations are relatively small and comprise highly selected, known ovarian cancer cases and healthy controls of the type evaluated in early biomarker development phases 1 and 2. Results have not been consistently replicated in multiple studies; presently, none are considered ready for widespread clinical application.
Initially, mass spectroscopy of serum proteins was combined with complex analytic algorithms to identify protein patterns that might distinguish between ovarian cancer cases and controls.
This approach assumed that pattern recognition alone would be sufficient to permit such discrimination, and that identification of the specific proteins responsible for the patterns identified was not required. This strategy was modified, using similar laboratory tools, to identify finite numbers of specific known serum markers that may be used in place of, or in conjunction with, CA-125 measurements for the early detection of cancer.
These studies
have generally been small case-control studies that are limited by sample size and the number of early-stage cancer cases included. Further evaluation is needed to determine whether any additional markers identified in this fashion have clinical utility for the early detection of ovarian cancer in the unselected clinical population of interest.
Because individual biomarkers have not met the criteria for an effective screening test, it has been suggested that it may be necessary to combine multiple ovarian cancer biomarkers to obtain satisfactory screening test results. This strategy was employed to quantitatively analyze six serum biomarkers (leptin, prolactin, osteopontin, insulin-like growth factor II, macrophage inhibitory factor, and CA-125), using a multiplex, bead-based platform.
A similar assay was available commercially under the trade name OvaSure until its voluntary withdrawal from the market by the manufacturer.[Response to FDA Warning Letter]
The cases in this study were newly diagnosed ovarian cancer patients who had blood collected just before surgery: 36 were stage I and II; 120 were stage III and IV. The controls were healthy age-matched individuals who had not developed ovarian cancer within 6 months of blood draw. Neither cases nor controls in this study were well characterized regarding their familial and/or genetic risk status, but they have been suggested to comprise a high-risk population. First, 181 controls and 113 ovarian cancer cases were tested to determine the initial panel of biomarkers that best discriminated between cases and controls (training set). The resulting panel was applied to an additional 181 controls and 43 ovarian cancer cases (test set). Pooling both early- and late-stage ovarian cancer across the combined training and test sets, performance characteristics were reported as a sensitivity of 95.3% and a specificity of 99.4%, with a PPV of 99.3% and a negative predictive value of 99.2%, using a formula that assumed an ovarian cancer prevalence of about 50%, as seen in the highly selected research population.
To avoid biases that may make test performance appear to be better than it really is, combining training populations and test populations in analyses of this sort is generally not recommended.
The most appropriate prevalence to use is the disease prevalence in the unselected population to be screened. The prevalence of ovarian cancer in the general population is 1 in 2,500. In a correction to their manuscript, the authors assumed that the prevalence of ovarian cancer in the screened population was 1 in 2,500 (0.04%) and recalculated the PPV to be only 6.5%. On that basis, the investigators have retracted their claim that this test is suitable for population screening. If this test were used in patients at increased risk of ovarian cancer, the actual prevalence in such a target population is likely to be higher than that observed in the general population, but well below the assumed 50% figure used in the published analysis. This revised PPV of 6.5% indicates that approximately 1 in 15 women with a positive test would in fact have ovarian cancer, and only a fraction of those with ovarian cancer would be stages I or II. The remaining 14 positive tests would represent false-positives, and these women would be at risk of exposure to needless anxiety and potentially morbid diagnostic procedures, including bilateral salpingo-oophorectomy.
Viewed in the context of the criteria previously described,
this assay would be classified as phase 2 in its development. While this appears to be a promising avenue of ovarian cancer screening research, additional validation is required, particularly in an unselected population representative of the clinical screening population of interest. A position statement by the Society of Gynecologic Oncologists regarding this assay indicated “it is our opinion that additional research is needed to validate the test’s effectiveness before offering it to women outside of the context of a research study conducted with appropriate informed consent under the auspices of an institutional review board.”
Numerous studies have found that women with an inherited risk of breast and ovarian cancer have a decreased risk of ovarian cancer after RRSO. (Refer to the RRSO section in the Breast cancer section of this summary for more information.) A retrospective study of 551 women with BRCA1 or BRCA2 pathogenic variants found a significant reduction in risk of breast cancer (HR, 0.47; 95% CI, 0.29–0.77) and ovarian cancer (HR, 0.04; 95% CI, 0.01–0.16) after bilateral oophorectomy.
A prospective, single-institution study of 170 women with BRCA1 or BRCA2 pathogenic variants showed a similar trend.
With oophorectomy, the HR was 0.15 (95% CI, 0.02–1.31) for ovarian, fallopian tube, or primary peritoneal cancer, and 0.32 (95% CI, 0.08–1.2) for breast cancer; the HR for either cancer was 0.25 (95% CI, 0.08–0.74). A prospective multicenter study of 1,079 women who were followed up for a median of 30 to 35 months found that RRSO is highly effective in reducing ovarian cancer risk in carriers of BRCA1 and BRCA2 pathogenic variants. This study also showed that RRSO was associated with reductions in breast cancer risk in both carriers of BRCA1 and BRCA2 pathogenic variants; however, the breast cancer risk reduction was more pronounced in BRCA2 carriers (HR, 0.28; 95% CI, 0.08–0.92).
In a case-control study in Israel, bilateral oophorectomy was associated with reduced ovarian/peritoneal cancer risks (OR, 0.12; 95% CI, 0.06–0.24).
A meta-analysis of all reports of RRSO and breast and ovarian/fallopian tube cancer in carriers of BRCA1/BRCA2 pathogenic variants confirmed that RRSO was associated with a significant reduction in risk of ovarian or fallopian tube cancer (HR, 0.21; 95% CI, 0.12–0.39). The study also found a significant reduction in risk of breast cancer (overall: HR, 0.49; 95% CI, 0.37–0.65; BRCA1: HR, 0.47; 95% CI, 0.35–0.64; BRCA2: HR, 0.47; 95% CI, 0.26–0.84).
Subsequently, a matched case-control study of 2,854 pairs of women with a BRCA1 or BRCA2 pathogenic variant with or without breast cancer showed a greater breast cancer risk reduction with surgical menopause (OR, 0.52; 95% CI, 0.40–0.66) than with natural menopause (OR, 0.81; 95% CI, 0.62–1.07). This study also reported a highly significant reduction in breast cancer risk among women who had an oophorectomy after natural menopause (OR, 0.13; 95% CI, 0.02–0.54; P = .006).
Another study of 5,783 women with BRCA1 or BRCA2 pathogenic variants who were followed up for an average of 5.6 years reported that 68 of 186 women who developed either ovarian, fallopian, or peritoneal cancer had died. The HR for these cancers with bilateral oophorectomy was 0.20 (95% CI, 0.13–0.30; P = .001). In carriers of BRCA pathogenic variants without a history of cancer, the HR for all-cause mortality to age 70 years associated with oophorectomy was 0.23 (95% CI, 0.13–0.39; P < .001).
Among studies with 50 or more subjects, prevalence ranged from 2.3% to 11%. Some of the variation in prevalence is likely due to differences in surgical technique, pathologic handling of the tissues, and age at RRSO. In the GOG 199 study of 966 high-risk women, the incidence of occult cancer was highest among carriers of BRCA1 pathogenic variants (4.6%), followed by carriers of BRCA2 pathogenic variants (3.5%), versus only 0.5% of noncarriers. The odds of an occult pathologic finding was fourfold higher among postmenopausal women.
In addition to a reduction in risk of ovarian and breast cancer, RRSO may also significantly improve OS and breast and ovarian cancer–specific survival. A prospective cohort study of 666 women with germline pathogenic variants in BRCA1 and BRCA2 found an HR for overall mortality of 0.24 (95% CI, 0.08–0.71) in women who had RRSO compared with women who did not.
This study provides the first evidence to suggest a survival advantage among women undergoing RRSO.
Studies on the degree of risk reduction afforded by RRSO have begun to clarify the spectrum of occult cancers discovered at the time of surgery. Primary fallopian tube cancers, primary peritoneal cancers, and occult ovarian cancers have all been reported. Several case series have reported a prevalence of malignant findings among carriers of pathogenic variants undergoing risk-reducing oophorectomy. Among studies with 50 or more subjects, prevalence ranged from 2.3% to 11%.
Some of the variation in prevalence probably results from differences in surgical technique, pathologic handling of the tissues, and age at RRSO. In the GOG 199 study of 966 high-risk women, the incidence of occult cancer was highest in carriers of BRCA1 pathogenic variants (4.6%), followed by carriers of BRCA2 pathogenic variants (3.5%), versus only 0.5% of noncarriers. The odds of an occult pathologic finding was fourfold higher among postmenopausal women.
In addition to occult cancers, premalignant lesions have also been described in fallopian tube tissue removed for prophylaxis. In one series of 12 women with BRCA1 pathogenic variants undergoing risk-reducing surgery, 11 had hyperplastic or dysplastic lesions identified in the tubal epithelium. In several of the cases the lesions were multifocal.
These pathologic findings are consistent with the identification of germline BRCA1 and BRCA2 pathogenic variants in women affected with both tubal and primary peritoneal cancers.
One study suggests a causal relationship between early tubal carcinoma, or tubal intraepithelial carcinoma, and subsequent invasive serous carcinoma of the fallopian tube, ovary, or peritoneum.
(Refer to the Pathology of ovarian cancer section of this summary for more information.)
These findings support the inclusion of fallopian tube cancers, which account for less than 1% of all gynecologic cancers in the general population, as a component of hereditary ovarian cancer syndrome and necessitate removal of the fallopian tubes at the time of risk-reducing surgery. There is clear evidence that RRSO must include routine collection of peritoneal washings and careful adherence to comprehensive pathologic evaluation of the entire adnexa with the use of serial sectioning.
The peritoneum, however, appears to remain at low risk for the development of a Müllerian-type adenocarcinoma, even after oophorectomy.
Of the 324 women from the Gilda Radner Familial Ovarian Cancer Registry who underwent risk-reducing oophorectomy, 6 (1.8%) subsequently developed primary peritoneal carcinoma. No period of follow-up was specified.
Among 238 individuals in the Creighton Registry with BRCA1/BRCA2 pathogenic variants who underwent risk-reducing oophorectomy, 5 subsequently developed intra-abdominal carcinomatosis (2.1%). Of note, all five of these women had BRCA1 pathogenic variants.
A study of 1,828 women with a BRCA1 or BRCA2 pathogenic variant found a 4.3% risk of primary peritoneal cancer at 20 years after RRSO.
Data are limited regarding outcomes of carriers of BRCA1 and BRCA2 pathogenic variants who are found to have occult lesions at the time of RRSO. In a multi-institution study of 32 women with either invasive carcinoma (n = 15) or serous tubal intraepithelial carcinoma (STIC) (n = 17), 47% of women with invasive cancer had a recurrence at a median time of 32.5 months, with an OS rate of 73%.
For women with intraepithelial lesions, one patient (approximately 6%) had a recurrence at 43 months, suggesting a different disease process between the two entities. Another study confirmed the malignant potential of STIC lesions. While 3 of 243 women (1.2%) with benign pathology at RRSO subsequently developed primary peritoneal carcinoma, 2 of 9 women (22%) with STIC developed high-grade pelvic serous carcinoma after a median follow-up time of 63 months.
Given the current limitations of screening for ovarian cancer and the high risk of the disease in carriers of BRCA1 and BRCA2 pathogenic variants, NCCN Guidelines recommend RRSO between the ages of 35 and 40 years or upon completion of childbearing, as an effective risk-reduction option. Optimal timing of RRSO must be individualized, but evaluating a woman's risk of ovarian cancer based on pathogenic variant status can be helpful in the decision-making process. In a large study of U.S. BRCA1 and BRCA2 families, age-specific cumulative risk of ovarian cancer at age 40 years was 4.7% for carriers of BRCA1 pathogenic variants and 1.9% for carriers of BRCA2 pathogenic variants.
In a combined analysis of 22 studies of carriers of BRCA1 and BRCA2 pathogenic variants, risk of ovarian cancer for carriers of BRCA1 pathogenic variants increased most sharply from age 40 years to age 50 years, while the risk for carriers of BRCA2 pathogenic variants was low before age 50 years but increased sharply from age 50 years to age 60 years.
In a population-based study of BRCA pathogenic variants in ovarian cancer patients, patients with BRCA2 variants had a significantly later age of onset than patients with BRCA1 variants (57.3 years [range, 40–72] vs. 52.6 years [range, 31–78]).
In summary, women with BRCA1 pathogenic variants may consider RRSO for ovarian cancer risk reduction at a somewhat earlier age than women with BRCA2 pathogenic variants; however, women with BRCA2 variants may still consider early RRSO for breast cancer risk reduction.
The role of concomitant hysterectomy at the time of RRSO in carriers of BRCA1/BRCA2 pathogenic variants is controversial. There is concern that a small portion of the proximal fallopian tube remains when hysterectomy is not performed, thereby resulting in a residual increased risk of fallopian tube cancer. However, several studies that have examined fallopian tube cancers indicate that the vast majority of these cancers occur in the distal or midportion of the fallopian tube, suggesting that the occurrence of proximal fallopian tube cancer would be a very unlikely event. Some reports have suggested an increased incidence of uterine carcinoma in carriers of pathogenic variants,
whereas others have not confirmed an elevated risk of serous uterine cancer.
A prospective study of 857 women suggested that any increased incidence of uterine cancer appeared to be among carriers of BRCA1 pathogenic variants who used tamoxifen;
this was confirmed by the same group in a later study of 4,456 carriers of BRCA1/BRCA2 pathogenic variants.
Even with tamoxifen use, the excess risk of endometrial cancer was small, with a 10-year cumulative risk of 2%.
In addition, the use of tamoxifen can now be minimized, given the options of raloxifene (which does not increase the risk of uterine cancer) and aromatase inhibitors for breast cancer prevention in postmenopausal women. Therefore, on the basis of the current understanding of the risk of uterine cancer in carriers of BRCA pathogenic variants, there is not a singularly compelling reason to consider hysterectomy at the time of RRSO to reduce the risk of uterine cancer. Concomitant hysterectomy does offer the advantage of simplifying the hormone replacement regimen for carriers of BRCA pathogenic variants who choose to take hormones. After hysterectomy, women can take estrogen alone (which does not increase the risk of breast cancer), without progestins, thereby eliminating the risk of postmenopausal bleeding.
Studies indicate that removal of the uterus is not necessary as a risk-reducing procedure. No increased BRCA pathogenic variant prevalence was seen among 200 Jewish women with endometrial carcinoma or 56 unselected women with uterine papillary serous carcinoma.
However, small studies have reported that uterine papillary serous carcinoma may be part of the BRCA-associated spectrum of disease.
The cumulative risk of endometrial cancer among carriers of BRCA pathogenic variants with ER-positive breast cancer treated with tamoxifen may be an additional factor to consider when counseling this population about risk-reducing hysterectomy.
Hysterectomy might also be considered in young, unaffected carriers of BRCA pathogenic variants who may want to use HRT but for whom hysterectomy would offer a simplified regimen of estrogen alone. In counseling a carrier of a BRCA pathogenic variant about optimal risk-reducing surgical options, aggregate data suggest that the risk from residual tubal tissue after RRSO is the least compelling reason to suggest hysterectomy. Therefore, in the absence of tamoxifen use or other underlying uterine or cervical problems, hysterectomy is not a routine component of RRSO for BRCA carriers.
For women who are premenopausal at the time of surgery, the symptoms of surgical menopause (e.g., hot flashes, mood swings, weight gain, and genitourinary complaints) can cause a significant impairment in their quality of life. To reduce the impact of these symptoms, providers have often prescribed a time-limited course of systemic HRT after surgery. (Refer to the Hormone replacement therapy in carriers of BRCA1/BRCA2 pathogenic variants section of this summary for more information.)
Studies have examined the effect of RRSO on quality of life (QOL). One study examined 846 high-risk women of whom 44% underwent RRSO and 56% had periodic screening.
Of the 368 carriers of BRCA1/BRCA2 pathogenic variants, 72% underwent RRSO. No significant differences were observed in QOL scores (as assessed by the Short Form-36) between those with RRSO or screening or compared with the general population; however, women with RRSO had fewer breast and ovarian cancer worries (P < .001) and more favorable cancer risk perception (P < .05) but more endocrine symptoms (P < .001) and worse sexual functioning (P < .05). Of note, 37% of women used HRT after RRSO, although 62% were either perimenopausal or postmenopausal.
Researchers then examined 450 premenopausal high-risk women who had chosen either RRSO (36%) or screening (64%). Of those in the RRSO group, 47% used HRT. HRT users (n = 77) had fewer vasomotor symptoms than did nonusers (n = 87; P < .05), but they had more vasomotor symptoms than did women in the screening group (n = 286). Likewise, women who underwent RRSO and used HRT had more sexual discomfort due to vaginal dryness and dyspareunia than did those in the screening group (P < .01). Therefore, while such symptoms are improved via HRT use, HRT is not completely effective, and additional research is warranted to address these important issues.
The long-term nononcologic effects of RRSO in carriers of BRCA1/BRCA2 pathogenic variants are unknown. In the general population, RRSO has been associated with increased cardiovascular disease, dementia, death from lung cancer, and overall mortality.
When age at oophorectomy has been analyzed, the most detrimental effect has been seen in women who undergo RRSO before age 45 years and do not take estrogen replacement therapy.
Carriers of BRCA1/BRCA2 pathogenic variants undergoing RRSO may have an increased risk of metabolic syndrome.
RRSO has also been associated with an improvement in short-term mortality in this population.
The benefits related to cancer risk reduction after RRSO are clear, but further data on the long-term nononcologic risks and benefits are needed.
Bilateral salpingectomy has been suggested as an interim procedure to reduce risk in carriers of BRCA pathogenic variants.
There are no data available on the efficacy of salpingectomy as a risk-reducing procedure. The procedure preserves ovarian function and spares the premenopausal patient the adverse effects of a premature menopause. The procedure can be performed using a minimally invasive approach, and a subsequent bilateral oophorectomy could be deferred until the patient approaches menopause. While the data make a compelling argument that some pelvic serous cancers in carriers of BRCA pathogenic variants originate in the fallopian tube, some cancers clearly arise in the ovary. Furthermore, bilateral salpingectomy could give patients a false sense of security that they have eliminated their cancer risk as completely as if they had undergone a bilateral salpingo-oophorectomy. A small study of 14 young carriers of BRCA pathogenic variants documented the procedure as feasible.
However, efficacy and impact on ovarian function was not assessed in this study. Future prospective trials are needed to establish the validity of the procedure as a risk-reducing intervention.
In a statistical Markov model using Monte Carlo simulation, risk-reducing salpingectomy with delayed oophorectomy was a cost-effective strategy considering quality-adjusted life expectancy for women with pathogenic variants in BRCA1/BRCA2.
Another study modeling ovarian cancer risk and effects of RRSO and salpingectomy found that the difference in estimated ovarian cancer risk is small when salpingectomy is performed on women of childbearing age and oophorectomy is performed 5 to 10 years later.
Prospective studies are under way evaluating the impact of bilateral salpingectomy with delayed oophorectomy on patient satisfaction and the reduction in ovarian cancer.
OCs have been shown to have a protective effect against ovarian cancer in the general population.
Several studies, including a large, multicenter, case-control study, showed a protective effect,
while one population-based study from Israel failed to demonstrate a protective effect.
There has been great interest in determining whether a similar benefit extends to women who are at increased genetic risk of ovarian cancer. A multicenter study of 799 ovarian cancer patients with BRCA1 or BRCA2 pathogenic variants, and 2,424 control patients without ovarian cancer but with a BRCA1 or BRCA2 pathogenic variant, showed a significant reduction in ovarian cancer risk with use of OCs (OR, 0.56; 95% CI, 0.45–0.71). Compared with never-use of OCs, duration up to 1 year was associated with an OR of 0.67 (95% CI, 0.50–0.89). The OR for each year of OC use was 0.95 (95% CI, 0.92–0.97), with a maximum observed protection at 3 years to 5 years of use.
This study included women from a prior study by the same authors and confirmed the results of that prior study.
A population-based case-control study of ovarian cancer did not find a protective benefit of OC use in carriers of BRCA1 or BRCA2 pathogenic variants (OR, 1.07 for ≥5 years of use), although they were protective, as expected, among noncarriers (OR, 0.53 for ≥5 years of use).
A small, population-based, case-control study of 36 carriers of BRCA1 pathogenic variants, however, observed a similar protective effect in both carriers of pathogenic variants and noncarriers (OR, approximately 0.5).
A larger case-control study of women with pathogenic variants in BRCA1 demonstrated maximum benefit after 5 years of OC use, while women with pathogenic variants in BRCA2 seemed to reach maximum benefit after 3 years of OC use.
A multicenter study of subjects drawn from numerous registries observed a protective effect of OCs among the 147 carriers of BRCA1 or BRCA2 pathogenic variants, with ovarian cancer compared with the 304 matched carriers of pathogenic variants without cancer (OR, 0.62 for ≥6 years of use).
Finally, a meta-analysis of 18 studies that included 13,627 carriers of BRCA pathogenic variants, 2,855 of whom had breast cancer and 1,503 of whom had ovarian cancer, reported a significantly reduced risk of ovarian cancer (summary RR, 0.50; 95% CI, 0.33–0.75) associated with OC use. The authors also reported significantly higher risk reductions with longer duration of OC use (36% reduction in risk for each additional 10 years of OC use). There was no association with breast cancer risk and use of OC pills formulated after 1975.
(Refer to the Oral contraceptives section in the Reproductive factors section of this summary for a discussion of OC use and breast cancer in this population.)
It has been suggested that incessant ovulation, with repetitive trauma and repair to the ovarian epithelium, increases the risk of ovarian cancer. In epidemiologic studies in the general population, physiologic states that prevent ovulation have been associated with decreased risk of ovarian cancer. It has also been suggested that chronic overstimulation of the ovaries by luteinizing hormone plays a role in ovarian cancer pathogenesis.
Most of these data derive from studies in the general population, but some information suggests the same is true in women at high risk due to genetic predisposition.
Among the general population, parity decreases the risk of ovarian cancer by 45% compared with nulliparity. Subsequent pregnancies appear to decrease ovarian cancer risk by 15%.
Earlier studies of women with BRCA1/BRCA2 pathogenic variants showed that parity decreases the risk of ovarian cancer.
In a large case-control study, parity was associated with a significant reduction in ovarian cancer risk in women with BRCA1 pathogenic variants, OR 0.67 (CI, 0.46–0.96).
For each birth, carriers of BRCA1 pathogenic variants had an OR of 0.87 (CI, 0.79–0.95). In this same study, parity was associated with an increase in ovarian cancer risk in carriers of BRCA2 pathogenic variants; however, there was no significant trend for each birth, OR 1.08 (CI, 0.90–1.29). Further studies are necessary to define the association of parity and risk of ovarian cancer in carriers of BRCA2 pathogenic variants, but for BRCA1 carriers, each live birth significantly decreases risk of ovarian cancer, as it does in sporadic ovarian cancer.
In the general population, breastfeeding is associated with a decrease in ovarian cancer risk.
In carriers of BRCA pathogenic variants, data are limited. One study found no protective effect with breastfeeding.
A case-control study among women with BRCA1 or BRCA2 pathogenic variants demonstrates a significant reduction in risk of ovarian cancer (OR, 0.39) for women who have had a tubal ligation. This protective effect was confined to those women with pathogenic variants in BRCA1 and persists after controlling for OC use, parity, history of breast cancer, and ethnicity.
A case-control study of ovarian cancer in Israel found a 40% to 50% reduced risk of ovarian cancer among women undergoing gynecologic surgeries (tubal ligation, hysterectomy, unilateral oophorectomy, ovarian cystectomy, excluding bilateral oophorectomy).
The mechanism of protection is uncertain. Proposed mechanisms of action include decreased blood flow to the ovary, resulting in interruption of ovulation and/or ovarian hormone production; occlusion of the fallopian tube, thus blocking a pathway for potential carcinogens; or a reduction in the concentration of uterine growth factors that reach the ovary.
(Refer to the PDQ summary on Ovarian, Fallopian Tube, and Primary Peritoneal Cancer Prevention for information relevant to the general population.)
Refer to the Oral contraceptives section in the Chemoprevention section of this summary for more information.
There are data to suggest that men with BRCA pathogenic variants have an increased risk of various cancers including male breast cancer and prostate cancer (refer to Table 7).
However, clinical guidelines to manage male carriers with BRCA pathogenic variants are based on consensus statements and expert opinions because information is limited.
There have been suggestions that BRCA2-associated prostate cancers are associated with aggressive disease phenotype.
Specifically, two recent studies have reported the median survival of male BRCA2 carriers with prostate cancer in the range of 4 to 5 years.
Furthermore, mortality rate was reported as 60% at 5 years in one of these studies, compared with 2% to 8% reported in the recent European and North American
prostate-specific antigen (PSA) screening trials after comparable follow-up. The data have been more limited in BRCA1-associated prostate cancers, however a number of recent studies have suggested an aggressive disease phenotype as well.
The benefits of PSA screening in BRCA carriers are unknown; however, there have been suggestions (based on very small studies) that PSA levels at prostate cancer diagnosis may be higher in carriers than noncarriers.
These findings suggest that PSA screening may be of potential utility in men with BRCA pathogenic variants, especially in view of the aggressive phenotype. Preliminary results of the IMPACT PSA screening study reported a PPV of 47.6% in 21 BRCA2 carriers undergoing biopsy on the basis of elevated PSA.
Because screening these men detected clinically significant prostate cancer, the authors suggest that these findings provide rationale for continued screening in such men; however, a survival benefit from such screening has not been shown. Ultimately, it is possible that information on BRCA pathogenic variant status in men may inform optimal screening and treatment strategies. Furthermore, recent data that the presence of a germline BRCA2 pathogenic variant is an independent prognostic factor for survival in prostate cancer led these authors to conclude that active surveillance may not be the optimal management strategy due to the aggressive disease phenotype.
Screening for male breast cancer in carriers of BRCA pathogenic variants as suggested by the NCCN clinical practice guidelines
includes breast self-exam training and education and clinical breast exam every 12 months starting at age 35 years. Furthermore, beginning at age 40 years, NCCN recommends prostate cancer screening for BRCA2 carriers and the consideration of prostate cancer screening for BRCA1 carriers.
Refer to the Prenatal diagnosis and preimplantation genetic testing section in the Psychosocial Issues in Inherited Breast and Ovarian Cancer Syndromes section of this summary for more information.
The distinct features of BRCA1-associated breast tumors are important in prognosis. In addition, there appears to be accelerated growth in BRCA1-associated breast cancer, which is suggested by high-proliferation indices and absence of the expected correlation of tumor size with lymph node status.
These pathological features are associated with a worse prognosis in breast cancer, and early studies suggested that carriers of BRCA1 pathogenic variants with breast cancer may have a poorer prognosis compared with sporadic cases.
These studies particularly noted an increase in ipsilateral and contralateral second primary breast cancers in carriers of BRCA1 and BRCA2 pathogenic variants.
(Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section of this summary for more information.) A retrospective cohort study of 496 Ashkenazi Jewish (AJ) breast cancer patients from two centers compared the relative survival among 56 carriers of BRCA1/BRCA2 pathogenic variants followed up for a median of 116 months. BRCA1 pathogenic variants were independently associated with worse disease-specific survival. The poorer prognosis was not observed in women who received chemotherapy.
A large population-based study of incident cases of breast cancer among women in Israel failed to find a difference in OS for carriers of BRCA1 founder pathogenic variants (n = 76) compared with noncarriers (n = 1,189).
Similar findings were seen in a European cohort with no differences in disease-free survival in BRCA1-associated breast cancers.
A prospective cohort study of 3,220 women from North America and Australia with incident breast cancer (including 93 BRCA1 carriers and 71 BRCA2 carriers) who were followed up for a mean of 7.9 years reported similar outcomes among BRCA1/BRCA2 carriers and those with sporadic disease.
However, results were based on chemotherapy regimens used in the late 1990s and did not adjust for surgical approach (lumpectomy vs. mastectomy) and effect of oophorectomy. The Prospective Outcomes in Sporadic versus Hereditary breast cancer (POSH) study recruited 2,733 women, 12% (n = 338) of whom had a BRCA1/BRCA2 pathogenic variant. Carriers showed no significant difference in outcome from noncarriers.
However, the cohort of patients with triple-negative breast cancer (n = 558) had a better overall survival than noncarriers at 2 years (HR, 0.59; P = .47), but not a statistically significant difference at 5 and 10 years.
A group of researchers reported the results of BRCA1/BRCA2 testing in 77 unselected patients with triple-negative breast cancer. Of these, 15 (19.5%) had either a germline BRCA1 (n = 11; 14%) or BRCA2 (n = 3; 4%) pathogenic variant or a somatic BRCA1 (n = 1) mutation. The median age at cancer diagnosis was 45 years in carriers of BRCA1 pathogenic variants and 53 years in noncarriers (P = .005). Interestingly, this study also demonstrated a lower risk of relapse in those with triple-negative breast cancer associated with a BRCA1 pathogenic variant than in non-BRCA1-associated triple-negative breast cancer, although this study was limited by its size.
Another study examining clinical outcome in BRCA1-associated versus non–BRCA1-associated triple-negative breast cancer showed no difference, although there was a trend toward more brain metastases in those with BRCA1-associated breast cancer. In both of these studies, all but one carrier of a BRCA1 pathogenic variant received chemotherapy.
Subsequently, in a study of 89 BRCA1 carriers and 175 noncarriers with triple-negative breast cancer, BRCA1 pathogenic variant status was not an independent predictor of survival after adjusting for age, oophorectomy, and risk-reducing mastectomy.
However, carriers who underwent oophorectomy had a significantly lower rate of breast cancer–related death.
A Polish study of 3,345 patients younger than 50 years with stages I through III breast cancer studied the impact of a BRCA1 pathogenic variant on prognosis. In this cohort, 233 patients (7%) carried one of three Polish BRCA1 founder pathogenic variants (5382insC, C61G, or 4154delA). BRCA1 carriers were younger and more frequently ER-negative and HER2/neu-negative. Ten-year survival was similar (80.9% in BRCA1 carriers and 82.2% in noncarriers). Oophorectomy was associated with improved survival in BRCA1 carriers (HR, 0.30; 95% CI 0.12–0.75).
In summary, BRCA1-associated tumors appear to have a prognosis similar to sporadic tumors despite having clinical, histopathologic, and molecular features that indicate a more aggressive phenotype. Carriers of BRCA1 pathogenic variants who do not receive chemotherapy may have a worse prognosis. However, because most BRCA1-associated breast cancers are triple negative, they are usually treated with adjuvant chemotherapy. Work is ongoing to determine whether BRCA1-associated breast cancers should receive different therapy than do sporadic tumors. (Refer to the Role of BRCA1 and BRCA2 in response to systemic therapy section of this summary for more information.)
Early studies of the prognosis of BRCA2-associated breast cancer have not shown substantial differences in comparison with sporadic breast cancer.
A small study reported statistically significant higher OS in carriers of BRCA2 pathogenic variants with metastatic breast cancer.
Retrospective and prospective studies have evaluated the response rate to chemotherapy in carriers of BRCA1 pathogenic variants receiving neoadjuvant chemotherapy for breast cancer, especially when using cisplatin.
Retrospective reviews have been published detailing pathologic complete response (pCR) rates to standard chemotherapies. In patients treated with standard neoadjuvant anthracycline- and taxane-based chemotherapy regimens, pCR rates ranged from 40% to 60%.
Neoadjuvant platinum compounds showed impressive pCR rates in patients with BRCA pathogenic variants. A cohort of 107 Polish women with a BRCA1 pathogenic variant received cisplatin 75 mg/m2 every 3 weeks for four cycles; the overall pCR rate was 61%.
However, in the subset of individuals with known BRCA pathogenic variants in the GeparSixto trial of 50 patients, the pCR rate was 66.7%, but there was no benefit with the addition of a platinum drug to the neoadjuvant chemotherapy regimen that included anthracycline and taxane.
The TNT trial compared docetaxel with carboplatin in 376 patients with metastatic, triple-negative breast cancer. Twenty-nine patients had a pathogenic variant in BRCA1 or BRCA2. There was no difference in the objective response rate (ORR) in the entire cohort between the two arms; however, a difference was noted in the BRCA carriers. The ORR in pathogenic variant carriers who received docetaxel was 33%; the ORR of those patients who received carboplatin was 68% (P = .03).
Multiple trials have evaluated the use of poly (ADP-ribose) polymerase (PARP) inhibition with and without chemotherapy. BRCA1 and BRCA2 are active in the repair of double-stranded DNA breaks by homologous recombination; PARP is involved in the repair of single-stranded breaks by base excision repair, as well as by PARP trapping on the DNA strand.
In 2017, two phase III trials explored PARP inhibitors in patients with metastatic breast cancer and a BRCA pathogenic variant. In the OlympiAD trial, 302 patients were randomly assigned to receive olaparib 300 mg orally twice daily or the physician’s choice of chemotherapy (capecitabine, eribulin, or vinorelbine). Progression-free survival (PFS) was improved from a median of 4.2 months to 7.0 months (HR, 0.58; P < .001) in patients treated with olaparib. OS was a secondary endpoint and no statistically significant difference was identified.
The EMBRACA trial randomly assigned 431 patients to talazoparib 1 mg orally daily versus the physician’s choice of capecitabine, eribulin, vinorelbine, or gemcitabine.
Patients receiving talazoparib had improved PFS by a median of 8.6 months versus 5.6 months (HR, 0.54; P < .001). OS was an alpha-protected endpoint for EMBRACA and, at the time of first report, the data were immature with only 51% of events reported (HR, 0.76; P = .105). On the basis of these results, the U.S. Food and Drug Administration has approved the use of both talazoparib and olaparib for the treatment of patients with inoperable or metastatic breast cancer and who have a germline BRCA pathogenic variant.
Ongoing research is evaluating multiple new strategies with PARP inhibitors to include targeting other germline pathogenic variants and somatic mutations. Trials, both in the early and metastatic settings, are evaluating single-agent PARP inhibitors and combining PARP inhibitors with other DNA damage repair agents, immunotherapies, and other targeted therapies to improve responses, as well as broaden the patient population who may benefit.
(Refer to the Systemic therapy in ovarian cancer treatment section in the Ovarian cancer section of this summary for more information about treatment strategies for BRCA-associated ovarian cancer.)
While lumpectomy plus radiation therapy has become standard local-regional therapy for women with early-stage breast cancer, its use in women with a hereditary predisposition for breast cancer who do not choose immediate bilateral mastectomy is more complicated. Initial concerns about the potential for therapeutic radiation to induce tumors or cause excess toxicity in carriers of BRCA1/BRCA2 pathogenic variants were unfounded.
Despite this, an increased rate of second primary breast cancer exists, which could impact treatment decisions.
Because of the established increased risk of second primary breast cancers, which may be up to 60% in younger women with BRCA1 pathogenic variants,
some carriers of BRCA1/BRCA2 pathogenic variants choose bilateral mastectomy at the time of their initial cancer diagnosis. (Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section of this summary for more information.) However, several studies support the use of breast conservation therapy as a reasonable option to treat the primary tumor.
The risk of ipsilateral recurrence at 10 years has been estimated to be between 10% to 15% and is similar to that seen in noncarriers.
Studies with longer periods of follow-up demonstrate risks of ipsilateral breast events at 15 years to be as high as 24%, largely resulting from ipsilateral second breast cancers (rather than relapse of the primary tumor).
Although not entirely consistent across studies, radiation therapy, chemotherapy, oophorectomy, and tamoxifen are associated with a decreased risk of ipsilateral events,
as is the case in sporadic breast cancer. The risk of contralateral breast cancer does not appear to differ in women undergoing breast conservation therapy versus unilateral mastectomy, suggesting no added risk of contralateral breast cancer from scattered radiation.
This finding is supported by a population-based case-control study of women diagnosed with breast cancer before the age of 55 years.
All women were genotyped for BRCA1/BRCA2. Although there was a significant fourfold risk of contralateral breast cancer in carriers compared with noncarriers, carriers who were exposed to radiation therapy for the first primary were not at increased risk of contralateral breast cancer compared with carriers who were not exposed. (Refer to the Mammography section for more information about radiation and breast cancer risk.) Finally, no difference in OS at 15 years has been seen between carriers of BRCA1/BRCA2 pathogenic variants choosing breast conservation therapy and carriers choosing mastectomy.
Despite generally poor prognostic factors, several studies have found an improved survival among ovarian cancer patients with BRCA pathogenic variants.
A nationwide, population-based, case-control study in Israel found 3-year survival rates to be significantly better for ovarian cancer patients with BRCA founder pathogenic variants, compared with controls.
Five-year follow-up in the same cohort showed improved survival for carriers of both BRCA1 and BRCA2 pathogenic variants (54 months) versus noncarriers (38 months), which was most pronounced for women with stages III and IV ovarian cancer and for women with high-grade tumors.
In a U.S. study of AJ women with ovarian cancer, those with BRCA pathogenic variants had a longer median time to recurrence and an overall improved survival, compared with both AJ women with ovarian cancer who did not have a BRCA pathogenic variant and two large groups of advanced-stage ovarian cancer clinical trial patients.
In a retrospective U.S. hospital-based study, AJ carriers of BRCA pathogenic variants had a better response to platinum-based chemotherapy, as measured by response to primary therapy, disease-free survival, and OS, compared with sporadic cases.
Similarly, a significant survival advantage was seen in a case-control study among women with non-AJ BRCA pathogenic variants.
A study from the Netherlands also showed a better response to platinum-based primary chemotherapy in 112 BRCA1/BRCA2 carriers than in 220 sporadic ovarian cancer patients.
A U.S. population-based study showed improvement in OS in BRCA2, but not in BRCA1, carriers.
However, the study included only 12 carriers of BRCA2 pathogenic variants and 20 carriers of BRCA1 pathogenic variants. Significantly better OS and PFS were observed in 29 high-grade serous ovarian cancer cases with a known BRCA2 variant (20 germline, 9 somatic) from The Cancer Genome Atlas study compared with cases negative for a BRCA pathogenic variant. BRCA1 pathogenic variants were not significantly associated with prognosis.
Furthermore, a pooled analysis of 26 observational studies that included 1,213 carriers of BRCA pathogenic variants and 2,666 noncarriers with epithelial ovarian cancer showed more favorable survival in carriers of pathogenic variants (BRCA1: HR, 0.73; 95% CI, 0.64–0.84; P < .001; BRCA2: HR, 0.49; 95% CI, 0.39–0.61; P < .001).
Thus, 5-year survival in both BRCA1 and BRCA2 carriers with epithelial ovarian cancers was better than that observed in noncarriers, with BRCA2 carriers having the best prognosis. A study in Japanese patients found a survival advantage in stage III BRCA1-associated ovarian cancers treated with cisplatin regimens compared with nonhereditary cancers treated in a similar manner.
In contrast, several studies have not found improved OS among ovarian cancer patients with BRCA pathogenic variants.
The largest of these studies involved a large series of unselected Canadian and U.S. patients who were tested for BRCA1 and BRCA2 pathogenic variants. At 3 years, the presence of a pathogenic variant was associated with a better prognosis, but at 10 years, there was no longer a difference seen in prognosis.
Furthermore, one study suggested that there was worse survival in ovarian cancer patients with a family history.
Compelling data suggest a short-term survival advantage in carriers of BRCA pathogenic variants. However, long-term outcomes are yet to be established. Survival in AJ ovarian cancer patients with BRCA1 or BRCA2 founder pathogenic variants does seem to be improved;
however, further large studies in other populations with appropriate controls are needed to determine whether this survival advantage applies more broadly to all BRCA cancers.
The molecular mechanisms that explain the improved prognosis in hereditary BRCA-associated ovarian cancer are unknown but may be related to the function of BRCA genes. BRCA genes play an important role in cell-cycle checkpoint activation and in the repair of damaged DNA via homologous recombination.
In addition to BRCA, other genes maintain homologous recombination, such as ATM, BARD1, PALB2, BRIP1, RAD51, BLM, CHEK2, and NBN. Comprehensive genetic testing of larger numbers of ovarian cancers has shown that approximately 50% of serous ovarian tumors may have somatic mutations or germline variants leading to a defective homologous recombination.
Deficiencies in homologous repair can impair the cells’ ability to repair DNA cross-links that result from certain chemotherapy agents, such as cisplatin. Preclinical data has demonstrated BRCA1 impacts chemosensitivity in breast cancer and ovarian cancer cell lines. Reduced BRCA1 protein expression has been shown to enhance cisplatin chemosensitivity.
Patients with BRCA-associated ovarian cancer have shown improved responses to both first-line and subsequent platinum-based chemotherapy compared with patients with sporadic cancers, which may contribute to their better outcome.
Women with ovarian cancer whose tumors have homologous recombination repair gene deficiency (HRD), resulting from either germline variants or somatic mutations, have improved survival compared with women with an intact homologous recombination. The majority of homologous recombination repair gene variants consist of somatic mutations or germline variants in BRCA1 and BRCA2, with one-third contributed by variants in other homologous repair genes.
PARP pathway inhibitors have been studied for the treatment of BRCA1- or BRCA2-deficient ovarian cancers. (Refer to the Role of BRCA1 and BRCA2 in response to systemic therapy section in the Treatment Strategies section of this summary for more information about PARP inhibitors.) While PARP is involved in the repair of single-stranded breaks by base excision repair, BRCA1 and BRCA2 are active in the repair of double-stranded DNA breaks by homologous combination. Therefore, it was hypothesized that inhibiting base excision repair with PARP inhibition in BRCA1- or BRCA2-deficient tumors leads to enhanced cell death, as two separate repair mechanisms would be compromised—the concept of synthetic lethality. The same concept may apply to tumors with HRD, and consequently, PARP inhibitors may have expanded use in women whose tumors have any homologous recombination defects beyond pathogenic variants in BRCA genes. In clinical practice, there are different tumor assays available to determine HRD tumors, which vary by method and definition. More study of PARP inhibitors in HRD ovarian cancers is ongoing.
Studies have used PARP inhibitors in ovarian cancer after platinum-based chemotherapy. A phase I study of olaparib, an oral PARP inhibitor, demonstrated tolerability and activity in carriers of BRCA1 and BRCA2 pathogenic variants with ovarian, breast, and prostate cancers.
A phase II trial of two different doses of olaparib demonstrated tolerability and efficacy in recurrent ovarian cancer patients with BRCA1 or BRCA2 pathogenic variants.
The overall response rate was 33% (11 of 33 patients) in the cohort receiving 400 mg twice daily and 13% (3 of 24 patients) in the cohort receiving 100 mg twice daily (i.e., 16 capsules daily). The most frequent side effects were mild nausea and fatigue.
In addition to ovarian cancer patients with germline BRCA1 or BRCA2 pathogenic variants, PARP inhibitors also may be useful in ovarian cancer patients with somatic BRCA1 or BRCA2 mutations or with epigenetic silencing of the genes.
Several phase II treatment studies have explored the efficacy of olaparib in patients with recurrent ovarian cancer, in both platinum-sensitive and platinum-resistant disease. Olaparib at 400 mg twice daily was used in a single-arm study to treat a spectrum of 298 BRCA-associated cancers, including breast, pancreas, prostate, and ovarian. Of the 193 women with ovarian cancer treated with olaparib, 31% had a response, and 40.4% had stable disease that persisted for at least 8 weeks.
Among the 154 women previously treated with at least three lines of chemotherapy, a similar overall response rate of 30% was seen, with comparable median durations of response of 8.2 months for platinum-sensitive disease and 8.0 months for platinum-resistant disease.
Another study of 173 patients with platinum-sensitive disease were treated with paclitaxel/carboplatin plus olaparib versus paclitaxel/carboplatin alone. The PFS was significantly longer in the olaparib group than the control group (12.2 vs. 9.6 months) (HR, 0.51; 95% CI, 0.34–0.77), especially in the subgroup of patients with BRCA pathogenic variants (HR, 0.21; 95% CI, 0.08–0.55). There were no differences in OS between the olaparib and control groups.
In contrast, other studies found that BRCA status did not predict survival advantage in women with platinum-sensitive ovarian cancer treated with olaparib. A randomized open-label trial assigned 90 women with recurrent platinum-sensitive ovarian cancer to either olaparib or cediranib and olaparib. Median PFS was significantly longer with the combination (17.7 mo vs. 9 mo) (HR, 0.42; 95% CI, 0.23–0.76). Subset analysis showed that combination cediranib and olaparib resulted in significantly longer PFS in the 43 BRCA wild-type/unknown patients than did single agent olaparib (16.5 mo vs. 5.7 mo) (HR, 0.32; P = .008) and a smaller trend toward increased PFS in 47 women with BRCA pathogenic variants (19.4 mo vs. 16.5 mo) (HR, 0.55; P = .16).
In another study, women with BRCA1/BRCA2 pathogenic variants and recurrent ovarian cancer within 12 months of a prior platinum-based regimen were randomly assigned to receive liposomal doxorubicin (Doxil) (n = 33) versus olaparib at 200 mg twice daily (n = 32) versus olaparib at 400 mg twice daily (n = 32). This study did not show a difference in PFS between the groups, which was the primary endpoint.
Of interest, the liposomal doxorubicin arm had a higher response rate than anticipated, consistent with other studies demonstrating that BRCA1/BRCA2-associated ovarian cancers may be more sensitive to liposomal doxorubicin than are sporadic ovarian cancers.
Another study demonstrated significant responses to olaparib in recurrent ovarian cancer patients, including patients with a BRCA1/BRCA2 pathogenic variant (objective response rate [ORR], 41%) and patients without a BRCA1/BRCA2 pathogenic variant (ORR, 24%).
This study emphasizes that certain sporadic ovarian cancers, particularly those of high-grade serous histology, may have properties similar to tumors related to a BRCA1/BRCA2 pathogenic variant.
As maintenance treatment, olaparib has shown significantly improved PFS in platinum-sensitive recurrent ovarian cancer. In a randomized controlled study of 265 patients (Study 19), those who received olaparib had a PFS of 8.4 months compared with 4.8 months in those who received the placebo (HR, 0.35; 95% CI, 0.25–0.49).
Within the cohort, the 136 patients with BRCA pathogenic variants demonstrated the most benefit with olaparib compared with placebo, with a PFS of 11.2 versus 4.3 months (HR, 0.18; 95% CI, 0.1–0.31).
There was no OS difference observed in the entire cohort, or in the carriers of BRCA pathogenic variants. A subsequent post hoc exploratory analysis excluded patients with BRCA pathogenic variants who received a PARP inhibitor at the time of progression to minimize the confounding influence on OS. In this group of 97 patients, an improved OS HR of 0.52 (95% CI, 0.28–0.97) was associated with olaparib, compared with placebo.
The more mature Study 19 data, after more than five years of follow-up, showed a trend towards OS benefit but did not meet the a priori significance threshold of P < .0001 with olaparib compared with placebo in the entire cohort (29.8 mo vs. 27.8 mo; HR, 0.73; 95% CI, 0.55–0.96), or among BRCA pathogenic variant carriers treated with olaparib (24.5 mo vs. 26.6 mo; HR, 0.62; 95% CI, 0.41–0.94).
Olaparib tablets have been shown to be effective maintenance therapy, compared with placebo, in a similar population of women with recurrent, platinum-sensitive ovarian cancer and BRCA pathogenic variants (SOLO2 trial). Olaparib resulted in a median PFS of 19.1 months versus 5.5 months for placebo (HR, 0.30; 95% CI, 0.22–0.41). Olaparib tablets offer the advantage of a reduced daily pill burden (two tablets twice daily) compared with 16 capsules daily.
Olaparib has demonstrated significant benefit as maintenance treatment in women with newly diagnosed advanced-stage, BRCA-associated ovarian cancer following response to primary treatment. The SOLO-1 trial randomly assigned 391 women with BRCA pathogenic variants to either olaparib 300 mg twice daily (n = 260) or placebo (n = 131) after primary surgery and platinum-based chemotherapy. After a median follow-up of 41 months, women receiving olaparib had a 70% lower risk of disease progression or death compared with women receiving placebo with an estimated improved PFS of approximately 3 years.
Within 3 years, disease progression or death occurred in 102 of 260 women (39%) in the olaparib group and 96 of 131 women (73%) in the placebo group. Side effects resulted in a dose reduction in 28% of patients and dose interruptions in more than half of patients. Fatigue and nausea were common side effects and reasons for dose reductions.
Rucaparib is a small molecule inhibitor of PARP-1, -2, and -3 and was approved in the United States for the treatment of advanced germline BRCA1/BRCA2-associated ovarian cancer in December 2016. A phase II study found that continuous dosing provided better response rates than intermittent dosing in women with pathogenic BRCA-associated breast and ovarian cancer.
A subsequent phase I/II dose-finding study selected a dose of 600 mg twice daily on the basis of manageable toxicity and a response rate of 59.5% in 42 women with recurrent, germline BRCA-associated, high-grade serous cancer who had received between two and four prior treatment regimens. Common grade 3 toxicities included fatigue, nausea, and anemia.
The ARIEL-2 phase II study found that rucaparib was effective in the treatment of recurrent, high-grade, platinum-sensitive ovarian cancer in women with BRCA variants, but also in BRCA wild-type women with high genomic loss of heterozygosity (LOH), which is a likely marker of HRD cancers. The study enrolled 206 women, of whom 40 had germline pathogenic variants or somatic mutations in BRCA. An additional 82 were BRCA wild-type, but had high LOH. Median PFS was significantly longer in the BRCA variant subgroup (12.8 mo) (HR, 0.27; 95% CI, 0.16–44), and the high LOH subgroup (5.8 mo) (HR, 0.62; 95% CI, 0.42–0.90), compared with the low LOH subgroup (5.2 mo). The authors concluded that both BRCA variant status and LOH score, as a surrogate for HRD, were molecular predictors of rucaparib sensitivity in women with recurrent, platinum-sensitive, high-grade ovarian cancer.
A phase III trial assessed rucaparib versus placebo in 576 women with recurrent, platinum-sensitive, high-grade ovarian cancer after response to second line, or greater, platinum chemotherapy. The study found that 196 women had BRCA pathogenic variants: 130 germline variants and 56 somatic mutations. Median PFS of women in the rucaparib group was 10.8 versus 5.4 months (HR, 0.35; 95% CI, 0.30–0.45). Median PFS was the most prolonged in BRCA-associated ovarian cancer: 16.6 months in the rucaparib group versus 5.4 months in the placebo group (HR, 0.23; 95% CI, 0.16–0.34). In women with HRD cancers, the median PFS was 13.6 versus 5.4 months (HR, 0.32; 95% CI, 0.24–0.42). On the basis of these data, the authors concluded that platinum sensitivity alone was a sufficient marker to predict benefit from rucaparib in women with advanced high-grade ovarian cancer, without requiring additional HRD or BRCA testing.
Niraparib is a selective inhibitor of PARP-1 and -2. A phase I dose-finding study observed a response rate of 42% with 300 mg daily in women with recurrent, BRCA-associated solid tumors.
In a cohort of 500 patients with platinum-sensitive, recurrent ovarian cancer, 234 received niraparib maintenance treatment and 116 received placebo (NOVA trial).
Niraparib maintenance resulted in improved PFS in BRCA pathogenic variant carriers (at 21 mo) and in wild-type patients with HRD positivity (at 12 mo) compared with wild-type patients without HRD tumor positivity (at 9 mo). Consistent with prior data, patients with germline BRCA pathogenic variants had the longest PFS of the three groups. Based upon the broad activity of niraparib maintenance in heavily pretreated women with ovarian cancer, regardless of platinum response or variant status, the QUADRA phase II trial studied the antitumor activity of niraparib in 463 women with recurrent, measurable ovarian cancer. Women had received a median of four prior lines of treatment. Twenty-eight percent of women had an overall response with a median duration of 9 months, which was improved in platinum sensitive, HRD-positive women.
More mature data are necessary to determine whether platinum sensitivity alone is a marker of response to PARP inhibitors in women with BRCA pathogenic variants, and the optimal timing of PARP inhibitors as treatment or as maintenance therapy. HRD status may also be used to predict response to PARP treatment on the basis of a better understanding of the multiple genes involved in homologous repair pathways.
Table 13 lists several organizations that have published recommendations for cancer risk assessment and genetic counseling, genetic testing, and/or management for hereditary breast and ovarian cancer.
Organization | Referral Recommendations | Risk Assessment and Genetic Counseling Recommendations | Genetic Testing Recommendations | Management Recommendations |
---|---|---|---|---|
ACMG/NSGC (2015) | Addressed | Risk Assessment: Addressed | Not addressed | Not addressed |
Genetic Counseling: Addressed | ||||
ACOG (2017) | Addressed | Risk Assessment: Addressed | Addressed | Addressed |
Genetic Counseling: Addressed | ||||
ASCO (2015) | Not addressed | Risk Assessment: General recommendations; not specific to HBOC | General recommendations; not specific to HBOC | Not addressed |
Genetic Counseling: Addressed | ||||
ESMO (2016) | Refers to other published guidelines | Risk Assessment: Refers to other published guidelines | Refers to other published guidelines | Addressed |
Genetic Counseling: Addressed | ||||
NAPBC (2014) | Refers to other published guidelines | Risk Assessment: Refers to other published guidelines | Indications for testing not addressed; components of pretest and posttest counseling addressed | Not addressed |
Genetic Counseling: Addressed | ||||
NSGC (2013) | Addressed | Risk Assessment: Refers to other published guidelines and available models | Addressed | Refers to other published guidelines |
Genetic Counseling: Addressed | ||||
NCCN (2020) | Addressed | Risk Assessment: Addressed | Addressed | Addressed |
Genetic Counseling: Addressed | ||||
SGO (2015, 2017) | Addressed | Risk Assessment: Addressed | Addressed | Addressed |
Genetic Counseling: Addressed | ||||
USPSTFa (2019) | Addressed | Risk Assessment: Addressed | Addressed in general terms and other guidelines referenced | Addressed in general terms and other guidelines referenced |
Genetic Counseling: Addressed | ||||
ACMG/NSGC = American College of Medical Genetics and Genomics/National Society of Genetic Counselors; ACOG = American College of Obstetricians and Gynecologists; ASCO = American Society of Clinical Oncology; ESMO = European Society for Medical Oncology; NAPBC = National Accreditation Program for Breast Centers; NCCN = National Comprehensive Cancer Network; NSGC = National Society of Genetic Counselors; SGO = Society of Gynecologic Oncology; USPSTF = U.S. Preventive Services Task Force. | ||||
aThe USPSTF guidelines apply to individuals without a prior cancer diagnosis. |
研究认为,错配修复基因是林奇综合征的遗传基础,微卫星不稳定性是错配修复基因缺陷的常见分子标志物。约15%散发性结直肠癌显示微卫星不稳定,高达28%散发性子宫内膜癌出现这种分子改变。
微卫星序列不稳定的散发性肿瘤大多数伴MLH1启动子高甲基化。在微卫星序列不稳定的林奇综合征相关肿瘤中,通常会缺失一个或多个错配修复基因相关的蛋白。
某些病理组织学特征强烈提示微卫星序列不稳定表型,包括肿瘤浸润性淋巴细胞、癌周淋巴细胞、未分化癌和子宫下段肿瘤的存在。采用临床标准是肿瘤检测选择标准的一种策略。计算机模型也被用来预测错配修复基因变异的概率,还可以在没有微卫星序列不稳定性或免疫组化信息的情况下使用。
然而,总体来说,当能提供肿瘤组织时,大肠和子宫内膜肿瘤的普遍检测有了进展。(更多信息,请参阅PDQ结直肠癌遗传学中通用肿瘤检测筛查林奇综合征部分的相关内容。)
As mismatch repair genes were identified as the genetic basis of Lynch syndrome, microsatellite instability was identified as a common molecular marker of mismatch repair deficiency. Approximately 15% of sporadic colorectal cancers show microsatellite instability, while up to 28% of sporadic endometrial cancers have this molecular change.
Most frequently, sporadic tumors with microsatellite instability have hypermethylation of the MLH1 promoter. In Lynch syndrome–related tumors showing microsatellite instability, there is typically loss of one or more of the proteins associated with the mismatch repair genes.
Certain histopathologic features are also strongly suggestive of a microsatellite instability phenotype, including the presence of tumor infiltrating lymphocytes, peritumoral lymphocytes, undifferentiated carcinomas, and lower uterine segment tumors. Use of clinical criteria is one strategy of selection criteria for tumor testing. Computer models have also been used to predict the probability of a mismatch repair genetic variant and can be used in the absence of microsatellite instability or immunohistochemistry information.
Overall, however, there is a move towards universal testing of colorectal and endometrial tumors when tumor tissue is available. (Refer to the Universal tumor testing to screen for Lynch syndrome section in the PDQ summary on Genetics of Colorectal Cancer for more information.)
癌症基因检测背景下的社会心理研究有助于定义心理结果,人际和家庭影响以及文化和社区反应。 这种类型的研究还确定鼓励或阻碍筛查和其他健康行为的行为因素。 它可以增强有关降低风险干预措施的决策,评估社会心理干预措施以减少与风险告知和基因检测相关的困扰和/或其他负面后遗症,提供有助于解决伦理问题的数据,并预测对不同人群进行检测的兴趣。
本节讨论遗传性乳腺癌和卵巢癌综合征的社会心理问题。PDQ摘要《大肠癌遗传学》中“遗传性结肠癌综合征的社会心理问题”部分讨论了与林奇综合征相关的妇科癌症的社会心理和筛查问题。
在提供遗传学咨询和基因检测的研究中,比较接受率是一项挑战,因为方法不同,包括所使用的抽样策略、招募设置和通过研究方案为高危人群或亲属进行检测。在2002年之前开展的40项研究进行系统回顾,对基因检测的应用情况进行分析,接受率变化范围较大,从25%到96%不等,平均接受率为59%。
多变量分析结果显示,BRCA1/BRCA2基因检测的接受率与乳腺癌或卵巢癌个人或家族史有关,还与研究的方法学特征有关,包括抽样策略、招募设置,以及研究如何定义实际接受率与检测目的。
其他因素与BRCA1/BRCA2基因检测的接受率呈正相关,但这些结果与其他研究并不一致。与检测呈正相关的心理因素,包括更严重的癌症特异性焦虑、更大的乳腺癌或卵巢癌发病风险。患癌症的亲属越多,越容易接受检测。
表14总结了美国临床和研究队列对基因检测的接受情况。
研究引文 | 研究人数 | 样本量 | 基因检测接受率 | 基因检测接受率的预测指标 | 说明 |
---|---|---|---|---|---|
Schwartz et al. (2005) | 新诊和局部未经治疗的乳腺癌患者,其BRCA1/BRCA2致病性突变风险≥10% | 231 | 177/231(77%)行基因检测 | 决定局部治疗。尚未决定是否接受局部治疗的女性更有可能接受基因检测。 | 免费检测。 |
34/231(15%)有基线访谈,但拒绝基因检测 | |||||
医生推荐检测。有医生推荐基因检测的女性更有可能接受检测。 | 38/177选择在检测结果出来前进行治疗。 | ||||
20/231拒绝基线访谈 | |||||
Kieran et al. (2007) | 2002年至2004年接受GC的妇女 | 250 | 88/250(35%)行基因检测 | 支付基因检测的能力(全部费用或保险不报销的费用)。不接受基因检测的女性是无经济实力支付费用女性的5.5倍。 | 450名女性在研究期间接受了乳腺癌和卵巢癌风险的遗传学咨询。对250名符合条件的女性进行了回访,并邮寄了一份调查问卷。 |
36/88例女性患者返回调查问卷 | |||||
遗传学咨询后回忆风险预估的能力。不接受基因检测的女性是回忆风险预估失败女性的15.5倍。 | 女性患者均有保险。 | ||||
162/250 (65%)符合要求 | |||||
65/162 反馈信息 | |||||
Susswein et al. (2008) | 非洲裔美国妇女和患有乳腺癌的白人妇女 | 768 | 529/768 (69%) 行基因检测 | 种族/民族。与白人女性相比,非裔美国女性接受检测的可能性较小。 | 从临床数据库中获得的样本。当检测不在保险范围内时,检测免费。对诊断时间的影响在非裔美国人(而不是白人)亚组中较为显著。 |
非裔美国女性:77/132(58%)接受基因检测 | |||||
最新诊断。最新确诊的非裔美国女性更容易接受检测。 | |||||
白人女性:452/636(71%)接受基因检测 | |||||
Olaya et al. (2009) | Catharine Wang, PhD, MSc(博士顿大学公共卫生学院) | 213 | 111/213 (52%) 行基因检测 | 既往有乳腺癌病史。有既往史的个人进行检测的机率增加3倍。 | 91.1%(175/213)患者可享受保险。有保险报销患者中,51.4%接受了检测,48.6%未接受检测。在没有保险报销的患者中,41.2%行基因检测,58.9%未行检测。 |
102/213(48%)拒绝基因检测 | 教育水平高。那些高中或以下学历的人群接受检测的人数仅占大学以上学历人数的1/3。 | ||||
Levy et al. (2010) | 20-40岁女性,新诊断为早发乳腺癌。 | 1474 | 446/1,474 (30%) 行基因检测 | 种族/民族。犹太裔女性接受检测的几率是非犹太白人女性的3倍。与非犹太白人女性相比,非裔美国人和西班牙裔女性接受检测的可能性要小得多。 | 样本来自国家商业保险个人数据库。 |
犹太女性:18/32(56%)接受基因检测 | 家庭所在地。生活在南方的女性比生活在东北部的女性更容易接受基因检测。 | ||||
非裔美国女性:10/82(12%)接受基因检测 | 保险类型。有服务点计划的女性比有健康管理组织计划的女性更容易接受检测。 | ||||
最新诊断。2007年确诊的女性接受检测的几率是2004年确诊女性的3.8倍。 | |||||
GC = 遗传咨询;HMO = 健康维护组织。 | |||||
a 自我报告作为数据来源。 | |||||
b 病历资料作为数据来源。 |
一些在美国以外地区开展的研究已对基因检测的接受率进行了分析。
在研究携带BRCA1/BRCA2致病突变的高危亲属的检测接受率时,平均接受率低于50%(36%-48%),女性亲属的接受率高于男性亲属。与高检测接受率相关的因素,多项研究未取得一致结果,通常会包括身为父母及想了解有关儿童风险的信息。
在有关接受BRCA1/BRCA2基因检测预测因子的累积证据评估的综述中,接受基因检测的重要预测因子,包括老年人、德系犹太人(AJ)后裔、未婚、乳腺癌个人史和乳腺癌家族史。在医院环境中招募受试者的研究高于社区招募的研究。需要即刻决定基因检测的接受率要高于延迟决策。
然而,有证据表明,诊断乳腺癌的女性患者,无论是在术前还是术后进行基因咨询,均对基因咨询(包括所收到的信息以及医生建议咨询的力度和时间)结果比较满意。
另有一篇文献
发现BRCA1/BRCA2致病突变患者对基因检测的接受程度与社会心理因素(如对乳腺癌的焦虑和对乳腺癌的感知风险)、人口统计学和医学因素(如乳腺癌或卵巢癌病史,有小孩以及直系亲属(FDR)受累数)相关。具有已知BRCA1/BRCA2致病突变的家庭成员更有可能进行检测;对BRCA1/BRCA2检测有更广泛认知、风险感知度更高、相信乳腺钼靶有利健康以及有较强意愿接受检测的家庭成员更有可能进行检测。
一篇综述分析了种族/种族差异影响BRCA1/BRCA2检测的接受度,非裔美国女性进行基因检测的意愿与以下因素相关:至少有一个直系乳腺癌或卵巢癌亲属,是携带者的风险较高,对自己可能是基因携带者的愧疚感较少。
一篇系统综述发现,与白人相比,包括非裔美国人和拉美裔人在内的某些少数民族群体对基因咨询和检测持有更多的负面看法和更多的担忧。更多非裔美国人和拉美裔人可能相信基因检测会显示他们的种族不如其他群体。此外,结果还提示,非裔美国人和西班牙裔对遗传学在癌症、BRCA状态和基因检测中的重要性方面的认知和了解明显不足。
接受检测的原因,包括希望了解孩子的风险,从不确定性中解脱出来,决定接受筛查或接受降低风险的手术,明确结婚、生育等重要的人生决定。
在非裔美国女性中,接受检测最重要的原因包括促进其他亲属进行基因检测。
医生推荐可能是另一个接受检测的促进因素。在一项对335名正在考虑进行基因检测的女性开展的回顾性研究中,77%女性希望得到遗传学医生提出是否应该进行基因检测的意见,49%女性希望得到家庭医生的意见。
然而,有一些证据表明基因检测转诊的人群多是有乳腺癌或卵巢癌母系家族史。一项对315名加拿大患者的回顾性研究中,与有父系家族史相比,有母系乳腺癌或卵巢癌家族史的患者更容易被医生推荐进行癌症遗传学咨询(95% CI,3.6-6.7)(P<0.001)。
研究发现,医生可能无法充分评估父系家族史。
或者可能低估了父系家族史对遗传风险的重要性。
或者可能低估了父系家族史对遗传风险的重要性。
在研究队列中,在被告知手术治疗时接受BRCA检测的患者比例似乎更高;
然而,其他研究结果表明,基因检测在临床实践中尚未得到充分利用,无法为乳腺癌的治疗决策提供信息。
使用BRCA测试来告知外科治疗决策的障碍包括,没有医生推荐新诊断的患者进行遗传咨询,保险类型(如医疗保险或医疗补助)以及测试时间和协调方面的挑战。
与新诊断乳腺癌患者的常规治疗不同,一项随机临床试验主动提供了快速遗传咨询(术前提供遗传咨询),结果表明,尽管试验组的遗传咨询接受率较高,但并不能转化为更高的遗传检测率、手术前收到检测结果比例以及更高的双侧乳房切除率。
保险覆盖范围是个人决定是否接受基因检测的一个重要考虑因素。(更多信息,请参阅PDQ摘要“癌症遗传风险评估和咨询”中“保险范围”部分内容。)
在非白人群体中,关于遗传咨询和检测的接受度的数据有限,需要进一步研究以明确影响这类人群接受检测的因素。
在某些种族/族裔群体中,BRCA测试的采用似乎有所不同。 一些研究比较了非洲裔美国人和白人女性的接受率。
一项病例对照研究观察了大学为基础的初级保健系统中的女性,有乳腺癌或卵巢癌家族史的非裔美国女性比有类似病史的白人女性更不可能接受BRCA1/BRCA2检测。
在另一项临床研究中,告知乳腺癌患者关于BRCA1/BRCA2的风险,显示非裔美国女性的接受率低于白人女性。
值得注意的是,在这些研究中观察到的种族差异似乎与以下因素无关,包括成本、诊疗途径、携带BRCA1或BRCA2致病突变的风险因素,社会心理因素差异(如风险感知、担忧或对检测的态度)。
几项研究已经评估了参加基因研究计划的非洲裔美国人对BRCA检测的接受程度或“认可度度”。 在来自美国犹他州的一个非洲裔美国人的研究参与者中,有83%接受了BRCA1测试。
年龄被认为是携带者的风险因素,年龄与癌症知识的广泛程度预示携带者对遗传检测的认可度。另一项通过医生和社区转诊的非裔美国女性的研究报道,BRCA1/BRCA2检测的接受率为22%。
接受检测的预测因素,包括携带致病突变的可能性较高、已婚以及对自身癌症风险不太确定的人群。最后,第三项研究从城市癌症筛查诊所招募高危非洲裔美国女性,发现接受BRCA检测的人群了解的乳腺癌遗传学知识更多,检测的主观障碍更少,包括负面情绪、担心歧视和与家庭有关的愧疚感。
虽然这些都是基因检测接受的独立预测因子,但并不能解释不同种族群体对检测接受率上的差异。可以解释这些差异的是,这可能与来自不同人种患者对检测持有的的不同态度和信念有关。
通过调查拉丁美洲和非裔美国人对乳腺癌基因检测的态度,表明他们对检测的了解和认知较为有限,一旦了解情况,他们通常会接受检测;与白人相比,拉丁美洲和非裔美国人对检测的关注相对较多。
例如,在一项对51名风险状态未知的拉丁美洲人进行的定性研究中,有重要发现:尽管参与者对遗传学的了解非常有限,但对遗传性癌症易感性的基因检测非常感兴趣。一个重要障碍是癌症和遗传学家庭讨论的保密性或尴尬性,这一点可通过干预策略加以解决。
另一项对54名具有遗传性乳腺癌风险的拉丁美洲女性进行的定性研究表明,尽管这些女性有兴趣了解更多遗传学咨询的知识,以便为家庭成员获取风险信息,但她们对BRCA1/BRCA2相关内容的了解程度较低。咨询障碍包括生活需求、成本和语言问题。
一项研究采用电话调查了宾夕法尼亚州匹兹堡市中心医疗中心的314名患者,其中50%非裔美国人,研究发现大多数(57%)受访者(非裔美国人和白人)认为基因检测评估疾病风险是个好主意;然而,比起白人,更多的非裔美国人认为基因检测会导致种族歧视(分别为37%和22%),基因研究缺乏道德和破坏自然(分别为20%和11%)。
最后,在一项对佐治亚州萨凡纳222名女性的研究中,大多数女性既无乳腺癌个人史(70%)也无乳腺癌家族史(60%),非洲裔美国女性(约占26%)不太了解乳腺癌基因和基因检测。检测意识还与高收入、高教育水平和有无家族乳腺癌史有关。然而,74%表示愿意接受乳腺癌易感基因检测。
在符合BRCA1/BRCA2致病突变检测标准的146名非裔美国女性的样本中,出生在美国以外的女性出现了更明显的预期负面情绪反应(如恐惧、绝望和对自己能够镇定面对检测缺乏信心)。强烈的乳腺癌特异性焦虑与预期的负面情绪反应、保密性问题以及预期的乳腺癌基因检测对家庭的影响有关。
未来的方向(例如,“我经常考虑我今天的行为将如何影响我的健康”)与该人群乳腺癌基因检测的总体可感知收益相关(n = 140); 然而,未来的方向也被发现与家庭相关的测试弊端正相关,包括家庭内和担心测试对家庭的影响。
供应商讨论和接受BRCA1/BRCA2突变基因检测患者存在种族差异。2007年至2009年间,一项对18至64岁的女性进行的研究发现,即使调整了致病突变风险,非裔美国女性也不太可能接受医生的基因检测建议。不同种族对BRCA1/BRCA2检测费用过高的担忧无差异,仅观察到在检测态度或保险方面的细微差异,这些均不影响检测接受度。
一项对符合BRCA1/BRCA2基因检测条件的乳腺癌或卵巢癌幸存者(N=50)的研究发现,48%被转诊去进行遗传咨询和基因检测和/或已经接受了基因检测。具有较高乳腺癌遗传学认知和更强自信心的个人更有可能接受遗传咨询和检测
在一项针对2009年至2012年50岁以前诊断为浸润性乳腺癌女性进行的研究中,该组患者经佛罗里达州癌症数据系统登记处核实,并符合现有指南规定的BRCA1/BRCA2基因检测要求,非裔美国人更不愿意与医生讨论或接受基因检测。
同一项研究发现,西班牙裔(61%)和非西班牙裔(65%)白人的总体检测率相似。 但是,在主要讲西班牙语的西班牙裔美国人中,其检测率较低(讲西班牙语人群50%,讲英语69%;P=0.0009),并且总体而言,西班牙裔美国人接受基因测试的可能性较小。
然而,这一发现与其他研究并不一致。在乔治亚州和洛杉矶郡对20-79岁患有导管原位癌或浸润性乳腺癌女性进行的一项研究中,通过监测、流行病学和最终结果(SEER)登记,所有女性均符合现有指南的BRCA1/BRCA2基因检测要求,在接受遗传咨询或医生指导的基因检测讨论中未发现种族差异。
有证据表明,拒绝检测的主要原因是没有孩子,这降低了家族进行检测的动机。对检测的负面影响的担忧,包括难以保持保险或对个人健康的担忧。
关于拒绝基因检测或从未接受过检测高危人群的特征,现有数据较有限。由于拒绝测试者可能不愿意参与研究,因此很难获得他们的样本。人们可能因不同时间和接受测试前教育和咨询的不同程度而拒绝检测,因此针对基因检测接受度的不同研究之间很难进行比较。一项研究发现,遗传性乳腺癌/卵巢癌家族中,43%受累和未受累的个人在完成检测基线访谈后拒绝接受检测。大多数拒绝检测的人选择不参加教育课程。拒绝者大多是男性、未婚并且有较少的乳腺癌亲属。与癌症有关的压力越高的患者,其抑郁程度更高。失访患者更易患癌症。
另一项研究观察了少量(n=13)拒绝检测女性患者,携带BRCA1/BRCA2致病突变的概率为25%-50%;这些拒绝检测的女性更有可能没有孩子、有更高的教育程度。研究表明,大多数女性在认真衡量风险和获益后拒绝检测。频繁监测的满意度被认为是大多数女性不接受检测的原因之一。
拒绝咨询和检测的其他原因,还包括没有孩子和在家族中较早的了解乳腺癌/卵巢癌。
第三项研究是英国一项大型多中心研究,评估了拒绝BRCA1/BRCA2检测的34名个体的特征。拒绝检测者比全国接受检测者年轻,拒绝检测的女性在癌症焦虑方面的平均评分较低。尽管78%拒绝检测者/推迟检测者感觉到有疾病风险,但她们认为了解BRCA1/BRCA2致病突变状态会引起一下担忧:
一个更重要的因素是拒绝者担心检测结果的影响,而不是具体的负担,如去遗传学门诊所需的时间和工作请假、家庭和社会义务所花费的时间。
在13个遗传性乳腺癌和卵巢癌家族中,有15%(n=31)的个体接受了遗传教育和咨询,并拒绝检测家族中已有的致病突变,特别报告了家族关系的积极改变,与那些追求检测的人相比,这类人的表现力和凝聚力更强。
BRCA1 / BRCA2致病性突变的检测几乎普遍限于18岁以上的成年人。 几份报告概述了根据儿童医学理解和提供知情同意能力的发展数据推断,对儿童进行成人疾病(例如乳腺癌和卵巢癌检测的风险。
研究表明,接受过BRCA1/BRCA2基因检测的人或已接受检测者的成年后代一般不赞成对未成年人进行检测。
尽管数据有限,但研究表明,男性、非致病突变携带者、母亲无乳腺癌个人史的人群可能更倾向于对未成年人行基因检测。
在对有孩子的家长进行的研究中,只有17%家长表示愿意让自己的孩子接受检测。对未成年人检测的关注点包括心理风险和不够成熟。潜在的获益是具有影响健康行为的能力。
针对儿童进行与乳腺癌、卵巢癌和其他成人疾病相关的基因突变检测遭到了普遍反对,尽管一些研究人员认为有必要对检测的有效性进行验证,但儿童进行BRCA1/BRCA2致病突变检测方面尚无研究数据。
在一项研究中,一组接受基因检测的母亲中有20名儿童(11至17岁)(其中80%以前患有乳腺癌,并且所有人都与他们的孩子讨论过BRCA1 / BRCA2检测)完成了自我报告调查表 他们对癌症的健康信念和态度,与癌症相关的感觉以及行为问题。
约90%孩子认为他们成年后会想要获得癌症风险信息;一半孩子担心自己或家人患癌症。无证据表明孩子有情绪困扰或行为问题。
该研究领域的最新文献表明,风险认知、健康信念、心理状态和个性特征是乳腺癌/卵巢癌基因检测决策的重要因素。在学术中心进行BRCA1/BRCA2检测的许多女性都坚信自己有致病突变,并决定进行基因检测,但对检测的风险或局限性却不甚了解。
在遗传学咨询的研究中,受试者在基线访谈时的大多数心理功能平均评分在正常范围内。
尽管如此,在许多遗传咨询研究中,有一部分受试者表现出焦虑、抑郁 或对癌症的担忧。
识别出这些人群对预防不良后果至关重要。一项针对205名拟行遗传咨询女性的研究,发现与癌症相关的担忧、乳腺癌风险感知、对乳腺癌遗传突变严重程度的感知之间存在相互作用,导致严重的担忧、乳腺癌风险的高感知、对乳腺癌遗传突变严重程度的低感知使女性接受BRCA1/BRCA2检测的几率是其他患者的两倍。
在高风险、癌症、乳腺癌和卵巢癌的配偶以及一般女性中普遍存在高估乳腺癌和卵巢癌遗传风险的倾向。
但高风险和平均风险女性对乳腺癌风险低估也有报道。
这种高估会鼓励人们相信,BRCA1/BRCA2基因检测将比目前已知的信息量更大。一些证据表明,即使咨询也不能劝阻低至中度风险的女性相信BRCA1检测的潜在临床价值。
过高估计乳腺癌和卵巢癌的风险与违背医生推荐的筛查方法有关。
对12项乳腺癌/卵巢癌遗传咨询结果的荟萃分析显示,遗传学咨询提高了风险感知的准确性。
女性似乎是有乳腺癌家族史的家庭主要传播者。
据报道,母婴传播病例多于父系。
可能是与家庭沟通模式有关,误认为乳腺癌风险只能通过母亲传播,在识别更多癌症远亲中,识别父亲家庭史方面遇到更大困难。在一项对2505名参与家庭保健影响临床试验女性的研究中,
不仅发现了父系家族史报告不足,而且与母系家族史相比,女性对父系家族史的乳腺癌风险感知水平明显较低。
鼓励医生和咨询师收集乳腺癌、卵巢癌或其他相关癌症的父系和母系家族史信息。
有报道的乳腺癌或卵巢癌家族史的准确率各不相同;一些研究发现准确率高达90%以上,
还有结果显示,在报告二级或更远亲属的癌症诊断
或癌症发病年龄方面有很多错误。
在报告乳腺癌以外的其他癌症时,检测的准确性较低。在一项研究中,有卵巢癌病史的报告准确率为60%,乳腺癌为83%。
医生和咨询师应注意,在报告假性家族乳腺癌病史时,有少数已发现孟乔森综合征病例。
更为常见的是由于无意的错误或知识上的空白而错误地报告家族癌症史,在某些情况下,这与潜在的母系癌症家族史的知情人早逝有关。
(更多信息,请参阅“癌症遗传风险评估和咨询的家族史”部分内容。
有针对性的写作,
视频、CD-ROM、交互式计算机程序和网站,
以及具有针对性的培训材料
可能是提高对基因检测利弊认识的有效方法。这类补充材料可使遗传学和初级健康医疗人员更有效地利用检测前教育和咨询的时间,并可劝阻无明显风险证据但拟行基因检测的个人。
对乳腺癌风险的咨询通常涉及具有家族病史的个人,这些家族病史可能归因于BRCA1或BRCA2。 但是,它也可能包括具有Li-Fraumeni综合征,共济失调-毛细血管扩张症,Cowden综合征或Peutz-Jeghers综合征家族史的个体。
(更多信息,请参阅本摘要“高外显率乳腺癌和/或妇科癌症易感基因”部分内容。)
携带者的管理策略,可能包括筛查的手段、频率和时机,监测方法,化学预防,降低风险的手术和激素替代疗法(HRT)等。对于携带者来说,保乳治疗和放疗作为癌症治疗手段可能会受到致病突变状态的影响。(更多信息,请参阅本摘要“BRCA致病突变携带者的临床管理”部分内容。)
遗传学咨询,还包括相关的社会心理问题,对计划中的家庭沟通沟通进行讨论,以及提醒其他家庭成员乳腺癌、卵巢癌和其他癌症风险增加的义务。数据表明,成年人对基因检测反应,受其他家庭成员检测结果的影响。
焦虑和焦虑的管理不仅是生活质量的重要因素,而且还有可能因为明显的焦虑而干扰对复杂遗传和医学信息的理解和整合,及对筛查的依从性。
对这些结果的正式、客观的评估有据可查。(更多信息,请参阅本摘要“情绪结果和行为结果”部分内容。)
已出版的BRCA1(以及随后的BRCA2)检测咨询计划,包括了解家族史、评估检测适用性、传达大量有关乳腺癌/卵巢癌遗传和相关医学及社会心理风险和获益的信息,以及关于保密和家庭沟通的专项伦理问题的讨论。
在许多前瞻性检测的候选人中,对参与者的苦恼、对癌症的侵入性想法、处理方式和社会支持进行了评估。在这些项目中,社会心理评估的结果包括怀孕、风险理解、心理调节、家庭和社会功能、生殖系统和健康问题。
荷兰的一项研究发现,受试者接受遗传性癌症综合征相关的癌症遗传咨询时,沟通过程中咨询师提问越多医学问题、提供越多社会心理的信息和越长时间的眼神交流,与较低的咨询满意度相关。咨询师提供的医学信息与受试者满足感和对需求已得到满足的感知密切相关。
许多社会心理结果的研究涉及到专门精选的研究群体,其中一些被用于绘制和克隆BRCA1和BRCA2基因谱。其中一个例子是K2082,一个被广泛研究的家族,来自犹他州的ormon家族的800名成员,其中BRCA1致病突变导致乳腺癌和卵巢癌的发病率增加。还有一项研究发现,即使在乳腺癌研究人群中,对结肠癌和前列腺癌的相关风险、是否需行RRM和RRSO治疗以及存在的社会心理风险的复杂性方面也缺乏完整的认知。
一项纳入21项研究的荟萃分析发现,遗传咨询在增加知识和提高感知风险的准确性方面是有效的。遗传咨询对诸如焦虑、抑郁或癌症特异性焦虑等情感结果和癌症监测活动行为结果的长期影响没有统计学意义。
然而,这些前瞻性研究的特点是,针对癌症的担忧的测量方法不均一,并且与基线变化的影响不一致。
尽管最初有人担心BRCA检测可能会产生不良情绪后果,但多年来的大多数研究表明,携带者和非携带者的心理焦虑程度较低,尤其是长期焦虑程度低。
在一项荟萃分析中,对受试者在收到检测结果后的短期(0-4周)、中期(5-24周)和长期(25-52周)癌症特异性焦虑进行研究,发现携带者在收到检测结果后短期内表现出较高的焦虑水平,并且会在中长时间内恢复到基线水平。
推移,焦虑程度降低。
研究发现,心理焦虑模式受多个因素影响,包括个体的癌症史、开展研究的国家。有癌症疾病史的携带者随着时间的推移,焦虑程度会有小幅度的下降,而没有癌症病史的携带者则没有任何变化。在结果不确定的个体中,没有癌症史的个体的焦虑明显少于有癌症史的个体。在非携带者中,与欧洲和澳大利亚的非携带者相比,美国的非携带者在心理焦虑方面的下降幅度要更加明显。在奥地利进行的一项研究指出,部分亚组受试者经历了更多的焦虑,包括那些年龄较大、新诊癌症病例,或者那些接受过咨询但拒绝接受BRCA检测的人群。
一些研究报告了更长随访期内(即收到结果后超过12个月)的情绪结果,并非上述荟萃分析的报告结果。
在英国的一项研究中,在3年的随访中,携带者和非携带者对癌症相关担忧无差异。
自荟萃分析发表以来,有两项美国研究
报道了收到BRCA检测结果追踪3年以上的女性中也有类似结果。
在一项横断面研究中,
167名接受BRCA检测追踪超过4年的女性报告称,使用癌症风险评估量表多维影响进行测试,她们的基因检测-特异性焦虑水平较低。
在多元回归模型中,致病突变携带者比非携带者更容易经历焦虑。在第二项研究中,
464名女性在基因检测后进行了中位5年(3.4-9.1年)的前瞻性随访。在受累和未受累的受试者中,与已知致病突变家族检测结果为阴性的女性相比,BRCA携带者有显著更高程度的焦虑、不确定性(仅受累人群)、感知压力(仅受累人群)和更少的阳性检测经历(仅未受累)
尽管两项研究
报道了BRCA携带者比非携带者更焦虑,但焦虑程度并未反映出临床上显著的功能障碍。
尽管大多数研究报告BRCA检测结果阳性对心理焦虑的影响相对较小,但其中许多研究是在有乳腺癌或卵巢癌家族史的家庭中进行的,这些家庭曾接受过广泛的检测前遗传咨询。因此,情绪反应可能并不适用于不同情境下检测的个体。例如,接受人口BRCA筛查的个体可能没有癌症家族史。
尽管建议在检测前进行遗传咨询,但当基因检测由非遗传学医生或直接由商业公司发起时往往难以实现。
例如,在加拿大的一项针对2080名犹太女性进行的研究中,她们参加了一项人群为基础的基因筛查研究,以检测犹太后裔家庭中常见的三种BRCA致病突变,但她们并未亲自进行基因咨询,而是在提供DNA样本之前,先得到一本关于BRCA1/BRCA2基因检测的小册子。在基因检测一年后,致病突变呈阳性的女性(n=18)显示出明显的癌症特异性焦虑,而致病突变呈阴性的女性则未观察到焦虑变化。
18名已知有致病突变女性在事件影响量表上的平均焦虑评分为25.3分(范围2-51);18名女性中,有10名(56%)在中度(n=7,评分范围26-43)或重度(n=3,评分范围44+)范围内评分。这个研究尚不明确的是在1年随访中观察到的焦虑增加是由于缺乏亲自遗传咨询,还是由于基线观察的焦虑水平较低,因为研究中的女性处于低风险但因先辈问题而符合检测条件。本项队列研究的后续研究发现,在检测后1-2年内,携带者的焦虑降低,这种变化因采取的风险降低方案而不同。明确的是,接受过降低风险的乳房切除或卵巢切除的患者与未接受两种手术的患者相比,其焦虑程度显著降低。
这一结果得到一个小型定性研究的支持。
同样,通过商业公司进行的直接对消费者(DTC)BRCA检测的影响需要进一步评估。病例研究报告指出,在收到直接基因检测的阳性BRCA结果时,受试者出现了不良的情绪反应,这表明通过公司受检的女性需要做进一步的情绪评估。
只有一项由商业公司开展的研究尝试对BRCA检测在这方面的影响进行评估。
共有32个检测结果阳性的患者(包括16名女性和16名男性)完成了半结构式访谈,这些人均检测到了阿什肯纳兹犹太人常见的三种BRCA原始致病突变的一种。无一例携带者报告极度焦虑,一些人经历了中度焦虑(13%)或最初的失望和焦虑,随着时间的推移而消散(28%)。该研究结果须谨慎解读,因为只有24%(136人中的32人)BRCA致病突变的携带者参与了研究,这有可能存在选择偏倚。
尽管有证据表明,在收到基因检测结果后,短期内焦虑增加,但对阳性携带者来说,任何不良反应都会在12个月内消失。
还需要进一步的研究对检测前未接受基因咨询人群的情绪结果进行评估。
乳腺癌患者在确诊时进行基因咨询和检测以协助制定治疗决策的现象越来越普遍。(更多信息,请参阅本摘要导言部分在癌症诊断时提供基因检测获益部分相关内容)尽管在诊断和手术之间提供快速遗传咨询和基因检测的不良心理影响引起了关注,
其他研究
包括一项随机临床试验
已提供证据表明在新诊断的乳腺癌患者中没有其他不良心理影响。 一项随机对照试验发现,接受快速遗传咨询和检测的患者比接受标准治疗的患者更积极地参与治疗决策。
然而,对20名新诊断乳腺癌患者进行的定性研究发现,这些患者中的部分亚组患者可能更难以面对BRCA检测结果,例如携带者并没有癌症家族史;那些没有可以识别的受累亲属的携带者;以及BRCA 检测结果不确定的高风险女性。
关于癌症易感性基因检测,特别是BRCA1/BRCA2基因检测结果的家庭交流比较复杂。性别似乎是家庭沟通和心理结果的一个重要变量。研究表明,女性携带者比男性携带者更有可能向其他家庭成员(尤其是14-18岁的姐妹和儿童)透露自己的检测结果。
在男性人群中,非携带者比携带者更有可能告诉他们的姐妹和孩子检测结果。BRCA1/BRCA2携带者向姐妹公布检测结果,她们的心理焦虑程度略有下降,相比之下,拒绝向姐妹透露检测结果的女性,其心理焦虑会轻微增加。一项研究发现,男性比女性更难以向家庭成员透露已知的致病突变(90%比70%)。
BRCA1/BRCA2检测结果与亲属的家庭沟通是影响接受检测的另一个因素。与较多远亲相比,更多的研究是关于直系和二级亲属交流。研究探讨了姐妹间关于BRCA1/BRCA2检测结果的交流过程和内容。
研究结果表明两种致病型突变携带者
以及检测结果不确定的女性
与姐妹沟通,为她们提供遗传风险信息。类似的研究结果也见于有些女性将不确定的检测结果透露给其女儿。
在未讨论基因检测结果的亲属中,最重要的原因是受累的女性与其亲属不亲近
或关系较差。
研究发现,BRCA致病突变的女性与母亲、成年姐妹和女儿分享结果的几率高于与父亲、成年兄弟和儿子分享。
一项对检测结果透露给直系亲属4个月后的家庭沟通的研究发现,与年轻女性相比,40岁或40岁以上的女性更有可能将检测结果告知父母。如果BRCA致病突变是通过父系遗传的,参与者也更有可能将结果告知兄弟。
另一项研究发现,检测结果披露主要局限于直系亲属,向远亲透露信息存有一定的难度。
年龄是告知远亲的重要因素,年轻的患者更愿意交流他们的基因检测结果。
此外,一项研究发现,较低的遗传担忧、对基因组信息兴趣浓厚、BRCA1或BRCA2致病突变携带者或从未结婚者愿意与更多的家庭成员交流。
相反,距离诊断时间间隔越长则会与越少的家庭成员交流。
一些深入的定性研究关注了与基因检测家庭交流相关的问题。尽管这些研究的结果可能无法推广到更多的高危人群中,但也说明了在家庭中传递遗传性癌症风险信息所涉及问题的复杂性。
在对一家家族癌症遗传学诊所的妇女进行的15次访谈的基础上,作者得出的结论是,尽管妇女感到有责任与亲戚讨论基因测试,但她们也经历了不确定,互相尊重和孤立的冲突。 妇女在家庭中应向谁通报以及如何向他们通报遗传性癌症和基因检测的决定可能受到妇女履行社会角色的需要与其对自己和他人的责任之间的紧张关系的影响。
另一项对21名参加家族性乳腺癌和卵巢癌遗传学诊所的女性进行的定性研究表明,一些女性可能发现,由于她们自己对癌症的恐惧和担忧,很难与伴侣和某些亲属,特别是兄弟,就遗传性癌症风险进行交流。
这项研究还表明,如何在家庭内部分享遗传风险信息,可能取决于现有的一般癌症沟通规范。例如,家庭成员一般都愿意彼此分享有关癌症的信息,也可能会有选择地避免与某些家庭成员讨论癌症信息,以保护自己或其他亲属不受负面情绪影响,也可能会要求特定亲属充当中间人向其他家庭成员透露。
研究还提到了家庭以外的人的潜在重要性,比如朋友,他们既是遗传性癌症风险信息的知情者,也是帮助处理这些信息支持者。
一项对31位BRCA的致病突变母亲的研究,对母亲向儿童的告知模式进行了探讨。
在选择向子女透露检测结果的人中,子女的年龄是最重要的因素。被告知的儿童中,有50%子女年龄在20-29岁,25%儿童年龄在19岁及19岁以下。儿子和女儿都收到了同等程度的告知。超过70%母亲在得知检测结果后的一周内通知了孩子。选择拒绝与孩子分享母亲中,93%的是由于孩子年龄太小。与年龄较大的儿童相比,13岁以下的儿童被告知结果的可能性更少,这些发现与其他三个研究结果一直。
另一项研究评估了187名接受BRCA1/BRCA2检测母亲在准备与孩子分享BRCA1/BRCA2检测结果的对话时需要的资源。78%母亲对三种或三种以上的资源感兴趣,包括文献(93%)、家庭咨询(86%)、与有经验者交流(79%)和支持小组(54%)。
一项纵向研究纳入了153名自主接受BRCA1和BRCA2致病突变基因检测女性和118名的他们的伴侣,评估检测前和检测后6个月时与基因检测和焦虑相关的交流。
研究发现,大多数夫妇讨论了进行基因检测的决定(98%),大多数检测参与者认为他们的伴侣是支持的,大多数女性向他们的伴侣透露检测结果(97%,n=148)。在检测前讨论中感到伴侣支持的受试者在检测后经历的焦虑较少,伴侣与受试者分享检测前的担心后会感觉更为舒适,并且检测后经历的焦虑更少。6个月跟踪调查显示,22%参与者认为有必要在访谈前一周与伴侣交流检测经验。大多数受试者(72%,n=107)在与伴侣分享问题时感到舒适,5%(n=7)受试者报告了基因检测导致的双方关系紧张。与结果呈阴性或结果不明确的夫妇相比,基因检测结果呈阳性的夫妇更容易出现关系紧张、对伴侣的保护性缓冲以及对相关问题更过的讨论。
一项对561名接受BRCA1/BRCA2基因检测女性的直系亲属的研究发现,尽管接受检测的女性已将结果告知,但22%直系亲属不记得被告知基因检测结果。
男性不太可能回忆起收到的结果(P>0.001)。在那些对接受检测结果能记起的人群中,有10.5%直系亲属忘记结果。对于那些对结果能记起的人,17.9%直系亲属的说法与结果不一致。对于有详细信息的检测结果(无论是真阳性还是真阴性),能回忆起检测结果的准确率更高(P=.029)。然而,不管检测结果如何,直系亲属在得知检测结果前认为的癌症风险更高(分别有74%和53%直系亲属报告他们认为他们的癌症风险高于平均水平,不管是收到检测结果前还是之后)。
近来,已有一些文献对有乳腺癌家族史、正在考虑或已经进行BRCA检测男性的心理影响进行研究。一项对爱尔兰16个高危家庭的22名男性进行的定性研究显示,在这项研究中,有女儿的男性接受的检测多于没有女儿的男性。这些报告提示,他们很少与亲属交流有关疾病的信息,一些报告指出,在女性家庭成员讨论癌症时他们被排除在外。还有一些男性提出,他们往往避免与女儿和其他亲属进行公开讨论。
与此相反,一项对研究纳入了59名BRCA1/BRCA2致病突变阳性男性,发现大多数男性都参与了有关乳腺癌和/或卵巢癌的家庭讨论。然而,只有不到一半的男性参加了关于降低风险手术的家庭讨论。接受BRCA检测的主要原因是担心孩子,并且需要确定他们是否会将致病突变遗传给孩子。在这项研究中,79%男性至少有一个女儿。这些人中,大多数描述了在收到BRCA检测结果后,他们家庭关系得以加强,这有助于家庭沟通并增进理解。
两项研究中的男性均表示担心自己会患上癌症。爱尔兰男人对性器官癌症特别恐惧。
一项研究评估了来自13个遗传性乳腺癌和卵巢癌家族的212名成员,他们接受了遗传咨询,并接受了BRCA1/BRCA2检测,以确定家族中的致病突变。在6-9个月后,未接受检测的成员在家庭表现力和凝聚力方面比接受检测的成员有显著提高。无论检测结果如何,被随机分配到以客户为中心组相比于问题解决基因咨询干预组的患者,冲突显著减少。
许多研究都是基于携带BRCA1/BRCA2致病突变或有明显癌症家族史的女性,来观察女性癌症高风险的心理影响。一些研究还评估了这些女性对伴侣的影响。
加拿大的一项研究评估了59名BRCA1/BRCA2致病突变的女性配偶。所有人都支持配偶接受基因检测的决定,17%的人希望他们能更多地参与基因检测过程。报告基因检测对夫妻关系无影响的配偶具有长期关系(平均时间为27年)。46%配偶表示,他们最担心的是伴侣死于癌症。19%担心他们的配偶会患上癌症,14%担心他们的孩子也会携带BRCA1/BRCA2致病突变。
在美国的一项研究中,118名女性在接受BRCA1和BRCA2致病突变基因检测前和在检测结果发布之后6个月时,其伴侣分别完成一次调查。在6个月时,只有10名伴侣报告说他们未被告知检测结果。91%人报告说,检测并未对双方关系造成压力。那些在检测前乐于分享担忧的伴侣在检测后经历的焦虑相对较少。研究未发现保护性缓冲影响伴侣的焦虑程度。
澳大利亚的一项针对95名乳腺癌和/或卵巢癌高危但未患病的女性(13名致病突变携带者和82名未知突变携带者)及其伴侣的研究表明,尽管大多数男性伴侣的焦虑程度与一般人群相当,但10%男性伴侣的焦虑程度明显高于正常人群,提示需要进一步的临床干预。采取严密监测方式和具有更高乳腺癌风险意识的男性报告更高的焦虑程度。男性和他们的伴侣之间的开放交流以及妻子家族中过去一年内发生癌症相关事件常与较低的焦虑水平相关。当男性被问及他们希望自己和伴侣得到什么样的信息和支持时,57.9%表示他们希望获得更多关于乳腺癌和卵巢癌的信息,32.6%人表示他们希望在处理伴侣的风险方面得到更多的支持。25%男性对如何改善高危女性伴侣的服务提出了建议,包括如何最好地支持伴侣、卫生保健专业人员更多地鼓励她们赴约以及与其他伴侣会面。
一篇文献综述表明,BRCA检测过程可能会使男性伴侣感到不安,特别是那些配偶被确定为携带者的男性。男性伴侣的焦虑似乎与他们对女性乳腺癌风险的认知、缺乏夫妻间的沟通以及对测试过程的疏远感有关。
对男性具有已知的BRCA1和BRCA2致病性突变的家庭的经历进行的文献回顾表明,尽管数据有限,但来自突变阳性家庭的男性比女性更不可能参与各个层面的遗传学交流,包括咨询和检测过程。 男性不太可能了解女性亲属获得的基因检测结果,并且这些家庭中的大多数男性都不会进行自己的基因检测。
一项对荷兰男性遗传BRCA1致病性变异风险增加的研究显示,男性倾向于否认或最小化其风险状况的情感影响,并专注于其女性亲属的医学影响。然而,这些家庭中的男子还提出,他们的女性亲属有相当严重的焦虑。
在另一项关于乳腺癌检测期间男性心理活动的研究中,来自18个不同高危家庭的28名男性(遗传BRCA1/BRCA2致病突变的风险为25%和50%)参与了本项研究。研究的目的是分析携带BRCA1/BRCA2致病突变的男性在进行基因检测时的焦虑。在被调查的男性中,大多数人在检测前的焦虑程度较低;乐观或没有女儿的男性评分最低。大多数致病突变携带者的焦虑和抑郁水平正常,没有内疚感,尽管一些携带者担心他们的女儿患上乳腺癌或卵巢癌,因此焦虑和责任感会增加。非携带者未报告内疚感。
在一项研究中
对检测后受试者是否遵循推荐的筛查指南进行分析。结果显示,超过一半的致病突变男性携带者在公布基因检测结果后未遵守推荐的筛查指南。这一结果与女性BRCA1/BRCA2致病突变携带者一致。
一项多中心英国的队列研究分析了193名个体BRCA1/BRCA2检测的预期结果,其中20%为年龄在28岁-86岁的男性。男性的焦虑程度较低,在携带者和非携带者之间无差异,从基因检测前到3年随访期间也未发生变化。22%致病突变男性携带者接受结直肠癌筛查,44%接受前列腺癌筛查;
然而,这项研究中的男性是否遵循了适合年龄的筛查指南并不清楚。
几项研究探索了BRCA检测结果与高危儿童的沟通。 在所有研究中,向4至25岁的儿童公开的比率约为50%。
一般来说,子女的年龄是决定是否公开检测结果的最重要因素。 在一项针对31位母亲的BRCA检测结果调查中,有50%的孩子被告知年龄在20至29岁之间,而略多于25%的孩子在19岁以下,儿子和女儿被告知人数相等。
同样,在另一项对42名携带BRCA致病突变的女性携带者的研究中,83%年龄18岁以上的子女被告知了检测结果,在13岁或13岁以下的子女中仅有21%被告知了结果。
多项研究对父母收到检测果后向孩子透露的时间进行分析。尽管大多数家长选择结果出来一周至几个月内告诉子女,但仍有部分家长选择延迟告知。
延迟告知的原因,包括等待孩子长大,让父母有时间调整信息,以及等待结果可以当面告知(对于离家在外的成年子女)。
在一项研究中,那些告知13岁以下子女的家长会更加焦虑,而那些没有告知年幼子女的家长焦虑则会少一些。与年轻子女的沟通受年龄、亲子沟通方式等变量的影响
一项研究观察了孩子们对检测结果公布的反应,或是沟通结果对亲子关系的影响。
关于子女对这类信息的理解,在一项研究中,几乎一半父母报告称,子女似乎不理解阳性检测结果的临床意义。根据报告,年龄较大的子女对这些信息有更好的理解。这项研究还显示,48%父母指出他们的孩子至少有一种负面反应,焦虑或担忧(22%),哭泣和恐惧(26%)。然而,值得注意的是,在这项研究中,儿童对检测结果的理解程度和反应仅仅基于父母的看法。此外,考虑到研究的回顾性设计,存在回忆偏差的可能性。不同年龄和性别的子女在情绪反应上无显著差异。最后,65%父母表示他们与孩子的关系未发生改变,而5位父母(22%)表示他们的关系得到了巩固。
有趣的是,一项针对869对母女(女儿年龄6-13岁)的大型多中心研究发现,有乳腺癌家族史或家族性BRCA1/BRCA2致病突变(BCFH+)的女孩与没有家族史的女孩相比,通过母亲的检测结果,能更好进行社会心理调节。
然而,根据母亲报告和对10至13岁女孩的直接评估,BCFH+女孩比没有家族史的同龄人经历了更大的乳腺癌特异性焦虑和更高的乳腺癌感知风险。此外,女儿焦虑程度与母亲密切相关。一项针对11至19岁年龄较大女孩的类似设计的研究发现,女儿对乳腺癌的特殊焦虑程度与感知风险和母亲的焦虑程度相关。与没有乳腺癌家族史的同龄人相比,女儿年龄越大,其自尊心越强。
另一项研究针对187名接受BRCA1/BRCA2检测母亲,评估了她们对基因信息的需求,以便为与孩子分享BRCA1/BRCA2检测结果的对话做准备。78%母亲对三种或三种以上的信息感兴趣,包括文献(93%)、家庭咨询(86%)、与有经验者交流(79%)和支持小组(54%)。
BRCA1/BRCA2致病突变的检测几乎局限于18岁以上的成年人。从儿童的医学理解和提供知情同意能力的发展数据推断,关于儿童患成人高发的疾病(如乳腺癌和卵巢癌)的检测风险在若干报告中已有相关讨论。
关于父母为孩子进行检测以确定是否患有成人遗传性癌症的调查显示,父母更愿意为自己的孩子进行检测,而不是自己接受乳腺癌基因检测,这为检测方提示了潜在冲突。
在美国进行的一项一般人群调查中,71%父母表示,如果他们携带一种易患乳腺癌的致病突变,他们会立刻对一个13岁女儿进行检测,以确定她的乳腺致癌基因状态,他们的意愿是中等、非常或极强的。
针对儿童进行与乳腺癌、卵巢癌和其他成人疾病相关的基因突变检测遭到了普遍反对,尽管一些研究人员认为有必要对检测的有效性进行验证,但儿童进行BRCA1/BRCA2致病突变检测方面尚无研究数据。
在一项研究中,有20名其母亲曾接受基因检测(其中80%有乳腺癌既往史,母亲均和孩子讨论过BRCA1/BRCA2检测)的儿童(11-17岁)完成了关于他们对癌症的健康信念和态度、与癌症有关的感觉和行为问题的自我报告问卷调查
约90%孩子认为他们成年后会想要获得癌症风险信息;一半孩子担心自己或家人患癌症。没有证据表明孩子有情绪困扰或行为问题。
另有一项研究发现,在BRCA1/BRCA2基因检测结果公布后1个月,42名招募的母亲中有53%与她们的8-17岁子女(一名或一名以上子女)讨论过检测结果。孩子的年龄及家庭健康问题沟通方式,并非母亲的致病性突变状态,影响是否将检测结果告知子女。
将致病突变遗传给孩子的可能性或会引起家庭对遗传性乳腺癌和卵巢癌(HBOC)担忧,
一定程度上,部分携带者可能放弃生育。
这些担心也可能促使女性考虑使用产前检查方法,以帮助降低遗传风险。
产前诊断是一个涉及多个方面的术语,用来指为评估胎儿是否存在遗传性疾病而进行的任何医疗措施。方法包括羊水穿刺和绒毛膜取样(CVS)。
这两种方法都有流产风险,一些证据表明使用这些检查或可导致胎儿缺陷。
此外,发现胎儿是遗传缺陷的携带者,可能会带给夫妇一个艰难的抉择继续妊娠还是终止妊娠。替代这些检测的方法是孕前基因检测(PGT),一种在受精卵植入子宫前进行的检测,以确定是否存在遗传性疾病,
从而避免与羊膜穿刺术和CVS相关的潜在危险和终止妊娠。利用基因检测获得的信息,帮助夫妻决定是否受孕。PGT可用于检测遗传性癌症易感基因的致病突变,包括BRCA。
在美国,已有一系列的研究对面对癌症风险授权组织(FORCE)成员对PGT的了解、兴趣(如将考虑使用PGT)和态度进行评估,FORCE是一个关注HBOC高危人群的倡导组织。
第一项研究是对283个成员进行的基于网络的调研,
第二项包括205位出席2007年FORCE大会的成员,
第三项是纳入962例受访者的网络调研。
这些研究记录了较低的意识水平,有20%至32%的研究受访者表示在参与研究之前听说过PGT。
鉴于对PGT的兴趣,第一项研究
发现只有13%女性可能行PGT,而在随后的FORCE研究中,33%受访者指出她们会考虑行PGT。
在第三次基于FORCE的研究(n=962)中,
多变量分析显示,对PGT的兴趣与生育更多孩子的愿望、以前做过任何产前基因检测及以前对PGT的认识有关。对PGT感兴趣的预测因素,包括同意其他有HBOC风险的人应接受PGT;认为PGT对有HBOC风险的人是可接受的;认为PGT信息应提供给有HBOC风险的人;支持无遗传突变的孕育胎儿和消除遗传疾病的PGT有益。相反,那些表示PGT“过度扮演上帝”,并报告说由于宗教原因,他们对PGT的兴趣不高。
目前,尚不清楚FORCE成员对PGT的态度是否代表了大多数BRCA携带者。一项对1081名BRCA携带者的横断面研究显示,其中65%是通过FORCE招募,其余为宾夕法尼亚大学招募。结果显示 大多数携带者支持PGT和产前诊断用于突变携带者(PGT支持率为59%,产前诊断为55.5%),
在那些家庭不完整的人群中,41%BRCA携带者报告说,携带者身份影响了她们未来生孩子的决定。这项研究还显示,21.5%BRCA携带者未婚女性感到更大的结婚压力。
英国人类受精和胚胎学权威机构已批准PGT用于遗传性乳腺癌和卵巢癌。在102名BRCA致病突变的女性中,大多数人支持PGT,但只有38名已成家的女性会考虑是否进行PGT,只有14%女性在考虑未来怀孕时再决定是否采用PGT。
在西班牙进行的一项多中心队列研究中,有77名受试者接受了BRCA检测,61%受试者表示她们会考虑PGT。与PGT感兴趣相关的因素是40岁及以上,且有癌症病史的人群。
在法国,获得多学科产前诊断小组授权的夫妇可以免费进行PGT,这是国家卫生保健系统的一项福利。然而,尚未有BRCA携带者被授权使用PGT。在一项全国性研究针对对490名未患病的BRCA携带者(18-49岁女性;18-69岁男性)进行研究发现,16%表示BRCA检测结果改变了她们正在准备的生育计划
通过对一些受访者的书面评论进行定性分析,主要影响与加提早怀孕时间、可能将致病性突变遗传给后代的负罪感以及未来的生育孩子具有相关性。作为对一个假想场景的回应PGT是现成的,33%参与者报告说,她们将接受PGT。与这一决定相关的因素在调查时尚未有生育计划,在确定BRCA致病性突变的情况下,认为终止妊娠是一个可接受的选择,并且家庭中乳腺癌和/或卵巢癌的病例较少。当被问及对PGT或产前诊断(PND)信息的期望时,85%被调查者认为应该与BRCA检测结果一起提供;45%被调查者认为应该在携带者有生育计划时进行。受访者表示,他们期望这些信息由癌症遗传学家(92%)、妇产科医生(76%)和全科医生(48%)提供。
一项小型(N=25)的定性研究,对育龄期BRCA致病突变阳性的女性进行分析,这些女性在生育前接受了基因检测,评估了她们的BRCA状况如何影响她们对生殖系统的基因检测(PGT和PND)的态度以及对生育的决定。
在本项研究中,决定接受BRCA检测的主要动机是期望控制个人的癌症风险,而不是希望告知未来生育的选择。HBOC严重程度影响了将BRCA病理型突变传给胎儿的担忧,也影响了进行PGT或PND的意愿,根据个人经验而不同。大多数人反对将已知的BRCA致病突变作为终止妊娠的原因。正如先前的研究所观察到的,关于生育选择的认知是多样的;然而,在终止妊娠方面,参与者有一种倾向,认为PGT比PND更容易被接受。关于终止妊娠的PGT与PND利弊的决定主要是由个人偏好和经验决定,并非由道德评判决定。例如,由于需要进行体外受精和服用可能增加癌症风险的激素,以及根据观察其他接受过这种方法的人群的经验,女性不接受PGT。
一项研究针对FORCE和Craigslist(一个公告栏网站)招募的高危男性(N=228)调查了这些问题。
与先前的女性研究相似,在参与调查之前,只有20%的男性知道PGT。 在多变量分析中,与那些从几个预先确定的选项中选择的人(例如,是否有遗传突变的孩子)和那些在宗教信仰下考虑进行PGT的人(而不是健康和安全)相比,那些选择“其他”选项以获得PGT潜在获益的人不太可能说出曾考虑进行PGT。
认识到BRCA1 / BRCA2致病性突变不仅在乳腺癌/卵巢家族中而且在某些种族中都普遍存在,
引起了关于伦理、社会心理和其他影响因素比如种族是确定疾病易感性的因素的重要讨论。担心人们认为一切都是由遗传因素决定和遗传下层阶级的产生。
有人提出了这样的问题:对被挑选出有乳腺癌易感基因群体的影响。有一个困惑就是谁能够给予或禁止这个群体参与他们基因特性相关的研究。这些问题挑战了传统观点,即将知情同意视为个人自主功能。
不断有文献对影响各种文化亚群的独特因素进行分析,显示发展具有文化特异性的遗传咨询和教育方法具有重要意义。
将成员纳入感兴趣的社区(如乳腺癌存活者、倡导者和社区领导人)可能会促进符合文化要求的遗传咨询资料的发展。
一项研究表明,参与任何遗传咨询(文化媒介或标准方法)均可以降低患乳腺癌的风险。
案例研究,论文和研究报告中都描述了由于基因检测对乳腺癌/卵巢癌易感性的出现而产生的伦理问题对人类的影响。 有关遗传风险信息传播的家庭权利和责任问题有望在未来几十年成为伦理和法律上的主要难题。
研究表明,62%受访家庭成员了解家族史,88%受访的遗传性乳腺癌/卵巢癌家庭成员对隐私和保密性格外关注。对三级亲属或远亲患癌症的担忧,与直系亲属或二级亲属的担忧大致相同
只有一半受访的乳腺癌或卵巢癌女性的直系亲属认为,向配偶或直系亲属告知BRCA1/BRCA2检测结果需要获得书面许可。对检测的态度因种族、既往接触过基因信息、年龄、乐观性格和信息类型而有所不同。利他主义是促使部分人群进行基因检测的一个因素。
许多专业团体就知情同意提出了建议。
有一些证据表明,并非所有从业者都具有遵循准则的意识。
研究表明,许多BRCA1 / BRCA2遗传检测同意书并不符合专业团体对应解决的11个领域的建议,并且它们忽略了高度相关的信息点。
在针对有乳腺癌或卵巢癌病史的女性进行的一项研究中,采访发现这些女性在向家庭成员公布基因信息时,对遇到的伦理问题未做充足准备。
这些数据表明,在检测前向家庭成员透露更多信息的,可以减轻告知遗传信息时出现的情绪负担。关于提醒家庭成员遗传性癌症风险方面的伦理问题,将使患者和医疗人员都能受益。(关于癌症遗传学和基因检测伦理知识的更多信息,请参阅癌症遗传学风险评估和咨询及癌症遗传学概述部分的相关内容。)
关于使用决策辅助工具作为常规遗传咨询的辅助手段的文献很少,但仍在不断增长,以帮助患者就癌症风险管理做出明智的决策
一项研究表明,在未患病女性接受6个月的随访后,BRCA1 / BRCA2阳性检测结果后,使用由个体化价值评估和癌症风险管理信息组成的决策辅助措施,可以减少侵入性思维并降低抑郁水平 。 使用决策辅助工具不会改变癌症风险管理的目的和行为。 但是,在受累的女性中,观察到对个人生活和若干决策相关结果产生轻微不利影响。
另一项研究对量身定制的决策辅助(包括价值观说明练习)和一般信息手册进行对比,手册是专门为女性制定卵巢癌风险管理决策而设计。在短期内,与接受小册子的女性相比,接受量身定制决策辅助的女性的决策冲突减少,知识增加,但两组之间的决策结果并无差异。此外,决策辅助似乎并不能改变参与者的癌症风险管理基本决策。
一项针对150名BRCA1/BRCA2致病突变的未患病女性的多地点随机试验,评估了决策辅助对乳腺癌风险管理决策和社会心理结果的影响。在6个月和12个月的随访中,与对照组相比,随机分配到决策辅助组的女性患癌症的焦虑程度较低(6个月时P=0.01,12个月时P=0.05)。
两组的决策冲突评分在基线时相对较低,随着时间的推移而下降;两组之间的评分无统计学差异。
越来越多的研究检查了接受过BRCA1和BRCA2致病性突变基因咨询和测试的个体对癌症风险管理选择的接受程度和依从性。 这些研究的结果报告间表15和表16中。结果因研究而异,包括接受或坚持筛查(乳腺摄影,磁共振成像[MRI],癌症抗原[CA] 125,阴道超声[TVUS])和选择 RRM和RRSO。 研究通常按致病性突变携带者或检测状态报告结果(例如,致病性突变为阳性,致病性突变为阴性或拒绝基因检测)。 遗传风险状况告知后的随访时间在各个研究中也有所不同,范围从12个月到数年不等。
这些研究的结果表明,乳腺筛查通常在告知BRCA1/BRCA2致病突变携带者状态后有所改善;尽管如此,筛查仍不理想。根据近期数据显示,很少有研究将MRI作为筛查手段。卵巢癌的筛查在不同的研究中有着广泛的差异,并且根据筛查检查的类型(即CA-125血清检测与TVUS筛查)也有所不同。然而,卵巢癌筛查似乎并未被BRCA1/BRCA2致病突变携带者广泛接受。RRM的接受率也存在很大差异,或与个人因素(如年龄较轻或有乳腺癌家族史)、社会心理因素(如减少癌症相关焦虑的愿望)、医疗人员的建议、文化或卫生健康系统因素有关。个人选择双侧乳房切除似乎还会受到治疗前的基因教育和咨询的影响,而与基因检测结果无关。
同样,不同研究对RRSO的接受率存在差异,可能受到类似因素的影响,包括年龄、乳腺癌的个人史、卵巢癌的感知风险、文化因素(即国家)和医疗人员的建议。
研究引文 | 研究人数 | RRM接受率 | 乳腺癌钼靶和/或乳腺MRI的接受率 | 随诊时间 | 说明 |
---|---|---|---|---|---|
研究引文 | 研究人数 | RRSO接受率 | 妇科筛查接受率 | 随诊时间 | 说明 |
美国 | |||||
Botkin et al. (2003) | 携带者 (n = 37)a | 携带者 0% | 乳腺钼靶 | 24个月 | |
–携带者57% | |||||
非携带者 (n = 92)a | 非携带者 0% | –非携带者 49% | |||
–拒绝检测20% | |||||
拒绝检测 (n = 15)a | MRI | ||||
–未评估 | |||||
Beattie et al. (2009) | 携带者(n=237) | 携带者 23% | 不适用 | 中位,3.7年 | 既往有乳腺癌病史,RRSO史, 选择RRM的女性年龄多小于60岁。 |
接受RRM中位时间:124天(知晓结果)。 | |||||
O’Neill et al. (2010) | 携带者(n = 146) | 携带者13% | 不适用 | 12个月 | BRCA1/2突变检测结果的会影响携带者行RRM手术决策。 |
Schwartz et al. (2012) | 携带者(n = 108) | 携带者37% | 乳腺钼靶 | 中位, 5.3年 | RRM的预测因素是年龄越小,咨询前癌症压力越大,乳腺癌或卵巢癌的诊断越新,卵巢保存完整。 |
–患病的携带者 92% | |||||
–未患病的携带者 82% | |||||
非携带者 (n = 60)a | 非携带者 0% | –非携带者 66% | |||
–检测结果不明确的患病者 89% | |||||
MRI | |||||
检测结果不明确(n = 206)a | 检测结果不明确6.8% | –患病的携带者 51% | |||
–未患病的携带者 46% | |||||
–非携带者 11% | |||||
检测结果不明确 27% | |||||
Garcia et al. (2013) | 携带者(n = 250) | 携带者44% | 排除RRM术后的女性: | 41个月;范围, 26–66个月 | 乳房监测显着降低从随访的1到5年:乳腺钼靶检查从43%降至7%; MRI 从35%降至3%。 |
乳腺钼靶: | |||||
–携带者43% | |||||
MRI: | |||||
–携带者 35% | |||||
Singh et al. (2013) | 携带者 (n = 136)b | 携带者 42% | 不适用 | 范围, 1–11年 | 一级或二级亲属有乳腺癌病史,至少有一次分娩史,并在2005年后接受过基因检测是决定行RRM的预测因素。 |
全球化 | |||||
Phillips et al. (2006) | 携带者(n = 70) | 携带者 11% | 乳腺钼靶 | 3 年 | |
–携带者 89% | |||||
MRI | |||||
–未评估 | |||||
Metcalfe et al. (2008) | 携带者(n = 2677) | 携带者18%(未患病) | 乳腺钼靶 | 3.9年;范围: 1.5–10.3年 | 不同国家采用风险管理方案的差异很大。 |
–携带者87% | |||||
MRI | 1294名参与者有乳腺癌病史。 | ||||
–携带者 31% | |||||
Julian-Reynier et al. (2011) | 携带者(n = 101) | 携带者 6.9% | 乳腺钼靶 | 5年 | 非携带者继续筛查。 |
–携带者 59% | |||||
–非携带者,年龄 30–39岁, 53% | |||||
非携带者(n = 145) | 非携带者0% | MRI | |||
–携带者 31% | |||||
–非携带者 4.8% | |||||
研究引文 | 研究人数 | RRSO接受率 | 妇科筛查接受率 | 随诊时间 | 说明 |
美国 | |||||
Scheuer et al. (2002) | 携带者(n = 179) | 携带者50.3% | CA-125 | 平均,24.8个月;范围: 1.6–66.0个月 | 接受RRSO的女性年龄较大,更有可能有乳腺癌病史。 |
– 携带者67.6% | |||||
经阴道超声 | |||||
– 携带者 72.9% | |||||
Beattie et al. (2009) | 携带者(n = 240) | 携带者 51% | 不适用 | 平均,3.7年 | 选择RRSP <60岁的女性既往有乳腺癌确诊,也接受了RRM。 |
RRSO的中位时间:123天。 | |||||
O'Neill et al. (2010) | 携带者(n = 146) | 携带者 32% | 不适用 | 12个月 | |
Schwartz et al. (2012) | 携带者(n = 100) | 携带者 65% | CA-125 | 中位,5.3年 | RRSO的预测因子是≥40y,并且乳腺癌诊断>10年。 |
Noncarriers (n = 52) | 非携带者 1.9% | – 携带者 56% | |||
– 非携带者 12% | |||||
–检测未明明确的 33% | |||||
–检测未明明确的 (n = 203) | –检测未明明确的13.3% | TVUS | |||
– 携带者 42% | |||||
– 非携带者20% | |||||
–检测未明明确的 26% | |||||
Garcia et al. (2013) | 携带者 (n = 305) | 携带者74% | 排除RRSO术后的女性: | 41个月;范围:26–66个月 | 随访第1年至第5年,卵巢监测显著降低; CA-125:47%降至2%; TVUS:45%降至2.3% |
CA-125 | |||||
– 携带者47% | |||||
TVUS | |||||
– 携带者 45% | |||||
Mannis et al. (2013) | 携带者 (n = 201)a | 携带者 69.6% | CA-125 | 中位,3.7年 | RRSO和筛查的预测因素,包括BRCA致病性突变携带者,年龄40-49岁,高收入,2个以上孩子,有乳腺癌病史,以及直系亲属患有卵巢癌。 |
– 26.3% | |||||
TVUS | |||||
– 26.3% | |||||
非携带者 (n = 103) | 非携带者 2.0% | 未报告 | |||
检测未明确的n=773)a;59/773,其含义不确定 | 检测未明确的 12.3% | CA-125 | |||
– 10.4% | |||||
TVUS | |||||
– 6.5% | |||||
Singh et al. (2013) | 携带者 (n = 136)b | 携带者 52% | 不适用 | 范围:1–11年 | RRSO的预测因子是直系亲属或二级亲属乳腺癌,母亲病逝于盆腔癌,分娩次数≥1次,年龄≥50岁,2005年后接受过检测。 |
国际 | |||||
Phillips et al. (2006) | 携带者 (n = 70) | 携带者 29% | CA-125 | 3 年 | |
– 携带者 0% | |||||
TVUS | |||||
–携带者 67% | |||||
Friebel et al. (2007) | 携带者 (n = 537) | 携带者 55% | 不适用 | 最少 6个月;中位 36个月 | RRSO最常见于 >40岁的经产妇。 |
Madalinska et al. (2007) | 携带者 (n = 160) | 携带者 74% | 携带者 26% | 12 个月 | 接受RRSO治疗的女性受教育程度较低,认为卵巢癌无法治愈,并且坚信RRSO的优势。 |
未报告妇科筛查的具体方法。 | |||||
Metcalfe et al. (2008) | 携带者 (N = 2,677) | 携带者 57% | 不适用 | 3.9年;范围:1.5–10.3年 | 各国在风险管理手段方面存在较大差异。 |
Julian-Reynier et al. (2011) | 携带者 (n = 101)a | 携带者 42.6% | TVUS | 5年 | RRSO接受率随年龄增长而升高。接受RRSO并未改变人们对乳腺癌风险的认识。非携带者经常会继续筛查。 |
非携带者(n = 145)a | 非携带者 2% | – 非携带者 43.2% | |||
Rhiem et al. (2011) | 携带者 (N = 306) | 携带者 57% | 未评估 | 中位,卵巢癌术后47.8个月 | RRSO时的中位年龄=47岁。RRSO术中发现1例隐性输卵管癌。术后26个月诊断1例腹膜癌。 |
Sidon et al. (2012) | 携带者 (N = 700例)a;386/700有乳腺癌病史 | BRCA1携带者: | 未评估 | 受累的乳腺癌 | > 60岁的女性,RRSO的接受率低(5年接受率为22%)。 > 70岁以上的女性均未进行RRSO。 |
– 54.5% | |||||
BRCA2携带者: | – BRCA1: 中位,2.29;范围, 0.1–11.45年 | ||||
– 45.5% | |||||
所有携带者均无乳腺癌既往史 | – BRCA2: 中位, 1.77;范围, 0.1–11.1年 | ||||
不受乳腺癌影响 | |||||
– 54.2% | |||||
所有携带者有乳腺癌既往史 | – BRCA1: 中位,1.63;范围, 0.1–11.28年 | ||||
– 43.2% | – BRCA2: 中位,1.75;范围, 0.1–8.98年 | ||||
MRI = 核磁共振;RRSO = 降低风险的输卵管-卵巢切除术。 | |||||
a 自我报告作为数据来源。 | |||||
b 病历资料作为数据来源。 | |||||
CA-125 = 癌胚抗原125; RRM = 降低风险的乳腺切除术; TVUS = 经阴道超声。 | |||||
a 自我报告作为数据来源。 | |||||
b 病历资料作为数据来源。 | |||||
c 未指定数据源 |
研究引文 | 研究人数 | RRSO接受率 | 妇科筛查接受率 | 随诊时间 | 说明 |
---|---|---|---|---|---|
美国 | |||||
Scheuer et al. (2002) | 携带者(n = 179) | 携带者50.3% | CA-125 | 平均,24.8个月;范围: 1.6–66.0个月 | 接受RRSO的女性年龄较大,更有可能有乳腺癌病史。 |
– 携带者67.6% | |||||
经阴道超声 | |||||
– 携带者 72.9% | |||||
Beattie et al. (2009) | 携带者(n = 240) | 携带者 51% | 不适用 | 平均,3.7年 | 选择RRSP <60岁的女性既往有乳腺癌确诊,也接受了RRM。 |
RRSO的中位时间:123天。 | |||||
O'Neill et al. (2010) | 携带者(n = 146) | 携带者 32% | 不适用 | 12个月 | |
Schwartz et al. (2012) | 携带者(n = 100) | 携带者 65% | CA-125 | 中位,5.3年 | RRSO的预测因子是≥40y,并且乳腺癌诊断>10年。 |
Noncarriers (n = 52) | 非携带者 1.9% | – 携带者 56% | |||
– 非携带者 12% | |||||
–检测未明明确的 33% | |||||
–检测未明明确的 (n = 203) | –检测未明明确的13.3% | TVUS | |||
– 携带者 42% | |||||
– 非携带者20% | |||||
–检测未明明确的 26% | |||||
Garcia et al. (2013) | 携带者 (n = 305) | 携带者74% | 排除RRSO术后的女性: | 41个月;范围:26–66个月 | 随访第1年至第5年,卵巢监测显著降低; CA-125:47%降至2%; TVUS:45%降至2.3% |
CA-125 | |||||
– 携带者47% | |||||
TVUS | |||||
– 携带者 45% | |||||
Mannis et al. (2013) | 携带者 (n = 201)a | 携带者 69.6% | CA-125 | 中位,3.7年 | RRSO和筛查的预测因素,包括BRCA致病性突变携带者,年龄40-49岁,高收入,2个以上孩子,有乳腺癌病史,以及直系亲属患有卵巢癌。 |
– 26.3% | |||||
TVUS | |||||
– 26.3% | |||||
非携带者 (n = 103) | 非携带者 2.0% | 未报告 | |||
检测未明确的n=773)a;59/773,其含义不确定 | 检测未明确的 12.3% | CA-125 | |||
– 10.4% | |||||
TVUS | |||||
– 6.5% | |||||
Singh et al. (2013) | 携带者 (n = 136)b | 携带者 52% | 不适用 | 范围:1–11年 | RRSO的预测因子是直系亲属或二级亲属乳腺癌,母亲病逝于盆腔癌,分娩次数≥1次,年龄≥50岁,2005年后接受过检测。 |
国际 | |||||
Phillips et al. (2006) | 携带者 (n = 70) | 携带者 29% | CA-125 | 3 年 | |
– 携带者 0% | |||||
TVUS | |||||
–携带者 67% | |||||
Friebel et al. (2007) | 携带者 (n = 537) | 携带者 55% | 不适用 | 最少 6个月;中位 36个月 | RRSO最常见于 >40岁的经产妇。 |
Madalinska et al. (2007) | 携带者 (n = 160) | 携带者 74% | 携带者 26% | 12 个月 | 接受RRSO治疗的女性受教育程度较低,认为卵巢癌无法治愈,并且坚信RRSO的优势。 |
未报告妇科筛查的具体方法。 | |||||
Metcalfe et al. (2008) | 携带者 (N = 2,677) | 携带者 57% | 不适用 | 3.9年;范围:1.5–10.3年 | 各国在风险管理手段方面存在较大差异。 |
Julian-Reynier et al. (2011) | 携带者 (n = 101)a | 携带者 42.6% | TVUS | 5年 | RRSO接受率随年龄增长而升高。接受RRSO并未改变人们对乳腺癌风险的认识。非携带者经常会继续筛查。 |
非携带者(n = 145)a | 非携带者 2% | – 非携带者 43.2% | |||
Rhiem et al. (2011) | 携带者 (N = 306) | 携带者 57% | 未评估 | 中位,卵巢癌术后47.8个月 | RRSO时的中位年龄=47岁。RRSO术中发现1例隐性输卵管癌。术后26个月诊断1例腹膜癌。 |
Sidon et al. (2012) | 携带者 (N = 700例)a;386/700有乳腺癌病史 | BRCA1携带者: | 未评估 | 受累的乳腺癌 | > 60岁的女性,RRSO的接受率低(5年接受率为22%)。 > 70岁以上的女性均未进行RRSO。 |
– 54.5% | |||||
BRCA2携带者: | – BRCA1: 中位,2.29;范围, 0.1–11.45年 | ||||
– 45.5% | |||||
所有携带者均无乳腺癌既往史 | – BRCA2: 中位, 1.77;范围, 0.1–11.1年 | ||||
不受乳腺癌影响 | |||||
– 54.2% | |||||
所有携带者有乳腺癌既往史 | – BRCA1: 中位,1.63;范围, 0.1–11.28年 | ||||
– 43.2% | – BRCA2: 中位,1.75;范围, 0.1–8.98年 | ||||
CA-125 = 癌胚抗原125; RRM = 降低风险的乳腺切除术; TVUS = 经阴道超声。 | |||||
a 自我报告作为数据来源。 | |||||
b 病历资料作为数据来源。 | |||||
c 未指定数据源 |
另一方面,许多致病性突变携带者的女性对RRM很感兴趣,希望将乳腺癌风险降至最低。 在一项对许多以前没有行降低风险手术的未受累妇女的研究中,他们接受了遗传咨询后接受了BRCA1检测,17%携带者(2/12)拟行乳腺切术,33%携带者(4/12)拟行卵巢切除术。
在随后的同一人群研究中,检测出阳性的女性中有35%的女性认为RRM是重要的选择,而有76%的女性认为降低风险的卵巢切除术是重要的选择。一项前瞻性研究评估了80名BRCA致病性突变的荷兰女性在5个时间点(从BRCA之前检测到结果公布后9个月)风险管理偏好的稳定性。46名参与者表示倾向于在基线检查时进行筛查。在25名基线时选择RRM的女性中,22名在检测结果公布9个月后做出同样的选择;但是,未报告实行RRM的病例数。
最初的兴趣并不总是转化为手术的决定。两项不同的研究发现,在结果公布后的一年内,致病性突变携带者的RRM发生率较低,其中一项研究显示携带者中有3%(1/29)和另一项携带者中有9%(3/34)接受过该手术。
在BRCA1大家族成员中,在基线(结果公布前)和BRCA1检测结果公布后1年和2年对癌症筛查和/或降低风险手术的接受率进行了评估。在参与调查的269名男女中,获得了37名女性携带者和92名女性非携带者的完整的数据,年龄均在25岁及25岁以上。在公布检测结果2年后,尽管37名携带者中有4名(10.8%)表示正在考虑RRM。但是无1例女性接受RRM,相比之下,在基线检查前无卵巢切除术史的26名女性中,46%(12/26)女性在检查后2年内进行了卵巢切除术。在25-39岁携带者中,29%(5/17)进行了卵巢切除术,而在40岁及以上携带者中,78%(7/9)进行了卵巢切除术。
在一项评估BRCA结果知晓3个月后接受降低风险手术的研究中,62名女性中有7人接受了RRM,有13人接受了RRSO。检测前接受RRSO的意愿与手术接受率相关。相反,接受RRM的意愿与接受率无关。总体来说,对降低风险手术犹豫不决的原因包括检测复杂的因素,如在无致病性突变的情况下家族史的重要性、对手术过程的担忧、更年期早期、使用激素替代疗法的时间和不确定性。
在英国的一项研究中,在遗传咨询观察过程中收集数据,并在接受遗传咨询后与41名女性进行半结构化访谈。
在遗传咨询中,曾29次提出降低风险手术,术后访谈中35次对此进行了讨论。15名女性表示将来会考虑卵巢切除术,9名女性说她们会考虑乳腺切除术。在访谈中讨论了行卵巢切除术和乳腺切除术的意义。受试者称,降低风险手术对其来讲是一种处理手段,可以(a)履行对其他家庭成员的义务,(b)降低风险,减少对癌症的恐惧。受访者对这类风险管理形式的成本描述如下:
由于以下原因,一些女性选择在未经基因检测的情况下接受RRM和RRSO:
在接受门诊监测的乳腺癌患者的直系亲属中,与对RRM无兴趣的女性相比,对RRM感兴趣和/或接受过手术的女性进行过更多的乳腺癌活检(P<0.05)和更高的客观10年乳腺癌风险预估(P<0.05)。 与那些表示有兴趣但尚未进行手术的妇女和不打算进行手术的妇女相比,随后接受RRM治疗的妇女中,进入诊所时的癌症担忧最高。(P <.001)。
当向刚被诊断出患有乳腺癌的女性提供BRCA检测时,已被证实BRCA检测会影响手术决策,与非携带者相比,携带者更有可能选择双侧乳房切除术。
一项评估435名乳腺癌幸存者中对侧RRM预测因子的研究发现,有16%的患者在转诊接受遗传咨询和BRCA1 / BRCA2基因检测之前已经接受了对侧RRM(连同患病乳房的乳房切除术)。
在遗传学咨询和检测前的对侧RRM预测因素,包括乳腺癌发病年龄较轻、诊断后间隔时间较长、至少有一例直系亲属受累、无全职工作。在公布检测结果的1年内,18%的BRCA1/BRCA2致病性突变检测呈现阳性的女性和2%检测结果为非致病性突变女性接受了对侧RRM。基因检测后的对侧RRM预测因素包括乳腺癌发病年龄较小、遗传咨询前癌症特异性焦虑程度高、BRCA1/BRCA2检测结果阳性。在这项研究中,在基因检测结果公布后1年,对侧RRM与抑郁无相关性。一项回顾性图表式研究对110名双侧乳腺切除术前已行BRCA1/BRCA2基因检测的新诊乳腺癌女性患者进行了评估。BRCA致病性突变携带者比未检测到突变的女性更容易接受双侧乳房切除术(83%比37%;P=0.046)。
在无致病性突变的女性中,对侧RRM的唯一预测因素是已婚(P=0.03)。年龄、种族、产次、疾病分期和生物标志物、乳腺钼靶密度增加和乳腺MRI对初次手术即对侧RRM的决定无影响。
2006至2014年间,在美国11个学术和社区中心对897名年龄在40岁及以下的乳腺癌女性患者进行了一项研究,发现随着时间的推移,行BRCA基因检测的比率有所增加。
诊断后1年内,87%病例进行了BRCA检测。这一数字从2006年的77%上升到2013年的95%。在检测出BRCA致病性突变阳性结果并认为检测结果影响手术决定的女性中(n=88),86%女性接受了双侧乳腺切除术,51%非携带者接受了双侧乳腺切除术(P<0.001)。在未经检测女性中,约1/3人群报告说,医疗人员并未告知需行BRCA检测;然而,根据国家规范指南,所有人都有权根据诊断年龄而进行测试。
对1994年至2002年间接受过单侧或双侧乳房重建术的荷兰女性(N=114)进行回顾性研究,以评估她们对该手术的满意度。
68%病例是BRCA致病性突变的未受累携带者,或有50%遗传家族性BRCA致病性突变风险的人群。60%受访者表示对手术满意,95%会再次选择RRM,80%人会选择同样的乳房重建手术。不到一半的患者报告了一些围手术期或术后并发症、持续的身体不适或一些肢体活动受限。29%病例报告说在手术后女性感知发生了变化,44%报告说他们的性关系发生了不良变化,35%表示她们认为他们的伴侣在性关系中经历了不良变化。然而,10%女性在手术后的性关系出现了积极的变化。与对手术结果满意的患者相比,不满意的患者更可能对手术及其后果感到不太了解,报告更多的并发症和身体不适,感觉乳房不属于自己,并表示不会再选择重建术。那些声称手术为性关系带来负面影响的女性更有可能:
在1997年至2005年间接受RRM治疗的90名瑞典女性接受了术前、术后6个月和术后1年的调查,以评估与健康相关的生活质量、抑郁、焦虑、性行为和身体形象的变化。在这三个时间点,与健康相关的生活质量和抑郁未见显著变化;焦虑随着时间的推移而减少(P=.0004)。超过80%女性在三个时间点均有过亲密关系。本研究在性交快感、不适、习惯和频率方面向性活跃女性提问。在性交频率、习惯或不适方面未见统计学上的显著差异。然而,从基线访谈到术后1年,患者的愉悦感显著降低(P=0.005)。术后1年,48%女性感觉自身吸引力降低,48%女性称有自我意识,44%女性对手术疤痕不满意。
在基因检测前的遗传咨询中,有可能不再对降低风险手术继续讨论。在一项多机构研究中,只有一半的遗传学专家在高危乳腺癌家庭女性咨询时讨论RRM和RRSO,
讨论手术选择能明显提高受试者的满意度,这些信息不会增加患者的焦虑。
鉴于BRCA1或BRCA2致病性突变的女性患卵巢癌的风险增加,收到RRSO信息的女性在手术接受率方面呈较大差异(27%-72%)。
一项研究表明,与选择RRSO或单纯监测相关的临床因素,包括年龄、至少一次分娩以及乳腺癌病史。
在本研究中,是否选择RRSO与乳腺癌或卵巢癌家族史无关。在遗传咨询中,子宫切除术是一种选择,80%已接受RRSO的女性也会选择子宫切除术。
关于癌症风险管理建议(如筛查和降低风险的干预措施)的接受和遵守情况的数据逐渐增多。在一项针对214名有乳腺癌或卵巢癌个人史(n=134)或家族史(n=80)的女性进行的横断面研究中,评估了《国家综合癌症网络指南》所定义的癌症筛查依从性和降低风险的行为。在40岁以上未受累的女性中,有10%人在上一年未行乳房X线或临床乳房检查(CBE),46%未做过乳房自我检查(BSE)。在有乳腺癌或卵巢癌病史的女性中,21%未行乳房X线检查,32%未做过CBE,39%未做过BSE。
在1997至2005年间,有312名女性接受了BRCA病理型突变的咨询和检测,对她们就遗传性乳腺癌和卵巢癌基因检测的看法进行了调查。调查包含降低风险措施的问题,包括筛查和降低风险手术。217名女性(70%)诊为乳腺癌,86名(28%)检测出BRCA1或BRCA2基因的致病性突变呈阳性。没有1例BRCA阳性的女性因检查过程不适拒绝行乳腺钼靶检查,11例BRCA阴性的女性(5.4%)同意这一说法。两组(BRCA阳性和BRCA阴性)均认为降低风险的手术是降低癌症风险和减少担忧的最佳方法,两组中的大多数患者均认为降低风险的乳腺切除术不会太剧烈、太可怕或太难看。
来自英国的一项前瞻性研究调查了1286名年龄在35至49岁之间,有乳腺癌家族病史并参加了多中心筛查计划的女性,对其进行了乳房X光检查的心理影响。112名女性发现了乳房X线检查异常结果并需进一步评估。然而,这些女性并未因为异常发现而出现明显的癌症担忧或消极心理反应。在1174名未发现乳房X线异常结果的女性中,在收到结果后,与基线相比,她们对癌症的担忧减少,消极心理反应更少。在6个月时,整个队列的癌症担忧和乳腺检查的心理不良反应均有所下降。
一项定性研究探讨了了医疗保健专业人员关于提供有关健康保护行为(例如运动和饮食)的信息的观点。7名医学专家和10名遗传咨询师在一个关注小组或单独接受了采访。这项研究报告指出健康保护行为规范信息的内容和范围存在较大差异,一般来说,受访者并不认为在遗传咨询会面的情况下促进此类行为是他们的职责。不过,受访者一致认为,有必要在风险评估的门诊内和其他门诊之间推行统一规范。
并非所有研究都明确指出筛查接受率是否在针对目标人群或特定临床情况的建议指导原则之内,也没有报告可能影响癌症筛查建议的其他变量。 例如,建议具有非典型导管增生史的女性遵循可能与一般人群不同的筛查建议。
在荷兰的一项前瞻性研究发现,在26名BRCA1/BRCA2致病性突变携带者中,选择乳房切除术的14名女性在检测结果公布前以及6个月和12个月后均有较明显的焦虑,高于那些选择监测的12名携带者和53名致病性突变阴性女性。总体来说,接受降低风险的乳房切除术(RRM)的女性焦虑程度有所降低;在1年后,她们的焦虑评分更接近选择监测女性和致病性突变阴性的女性。
有趣的是,选择RRM的女性对检测前的乳房和身体形象的满意度低于选择监测BRCA1/BRCA2致病性突变携带者或非携带者。在接受RRM的女性中,除了1名女性外,其他人都不后悔在检测结果知晓后一年做出的决定,但很多人在身体形象、性兴趣和性功能、自尊方面面临一些困难。认为医生未充分告知RRM的后果。
在5年的随访中,接受过RRM女性的身体形象和性关系的变化都不太乐观,但关于癌症的恐惧也明显降低。
在一项对78名接受降低风险手术女性(包括BRCA1/BRCA2携带者和来自高危家庭但未检测到BRCA1/BRCA2病理型突变的女性)的研究中,在术前2周和术后6个月和12个月对癌症特异性和一般性的焦虑进行评估。
病例包括单独接受RRM或RRSO及同时接受两种手术的女性。在术后1年内,未观察到明显的焦虑。
一项随访研究(平均14年)报告了609名在梅奥医学中心接受RRM的女性的复杂社会心理反应。70%人对RRM表示满意,11%持中立态度,19%人不满意。18%患者认为,如果有再次选择的机会,她们很有可能或肯定不会实施RRM。大约3/4指出,手术减轻了她们对癌症的担忧。一半患者表示她们对身体形象的满意度无变化;16%表示术后身体形象有改善。36%对RRM术后的形象不满意。约1/4女性报告说,RRM对她们的性关系和女性气质产生不良影响,18%女性自尊心受损。与RRM满意度最密切的因素,包括术后对外观的满意度、减压、未行乳房重建或乳房植入物无问题、性关系无改变或得到改善。将医生建议作为选择RRM的主要原因的女性在RRM后往往会感到不满意。
一项对60名接受RRM的健康女性的研究评估了她们在术后4年4个月的平均满意度、身体形象、性功能、侵犯和回避以及当时心理状态。其中76.7%病例有家族史(21.7%)或携带BRCA1或BRCA2致病性突变(55%)。总体来说,97%受访女性对RRM感到满意(17%)或非常满意(80%)。除一名女性外,所有其他参参与者都会向其他女性推荐这种方法。大多数女性(66.7%)报告说手术未影响她们的性生活,31.7%女性报告说性生活变差,76.6%女性报告说无论是否进行重建,身体形象都未见改变或改善。23.3%女性术后自我形象恶化。女性术后平均焦虑水平略高于正常水平,那些认为术后乳腺癌风险仍然较高的女性比那些低风险认知的女性的整体和癌症相关焦虑水平更高。此外,与有不限家族史的女性相比,BRCA1和BRCA2致病性突变携带者以及有乳腺癌和/或卵巢癌家族史的女性,其癌症相关焦虑水平更高。
关于基因检测的结果如何影响癌症诊断时的治疗决策,知之甚少。 两项研究探讨了乳腺癌诊断时的遗传咨询和BRCA1 / BRCA2遗传检测。
其中一项研究发现,诊断时的基因检测明显改变了手术决策,与非携带者相比,致病性突变携带者更多的是选择双侧乳房切除术。在较小的系列中,有48%的具有已知致病性突变的女性和100%的具有已知致病性突变的女性选择了双侧RRM。
在未发现致病性突变的女性中,24%也选择了双侧RRM。4%拒绝检测者也接受了双侧RRM。在致病性突变携带者中,双侧RRM的预测因子包括患者是否告知她们的医生在检测前已推荐BRCA1/BRCA2检测和双侧RRM,以及她们的检测结果是否为阳性
关于基因检测后在确诊时接受RRM女性生活质量结果的数据尚不足。
来自荷兰的一项前瞻性研究就乳腺癌遗传学咨询对女性的长期心理影响进行了评估,如果有指征,还评估了在原发乳腺癌采用放疗时进行的基因检测。在接受遗传学咨询的人中,部分进行了基因检测并选择接受检测结果(n=58),有些人接受了咨询但不符合转诊标准(n=118),还有部分受访者拒绝接受咨询/检测(n=44)。接受放疗的另一组女性未接受咨询(n=182),采用同一种随访方案。在接受放疗的首次咨询后4周、11周、27周和43周,评估患者的心理困扰。四组患者的一般焦虑、抑郁或乳腺癌特异性焦虑均无差异。
对583名有乳腺癌个人和家族史并在1960-1993年间接受对侧RRM的妇女进行了回顾性问卷调查,探讨了对乳房切除术后的总体满意度以及影响对该手术满意和不满意的因素。
术后平均随访10.3年。总体而言,83%受访者对对侧RRM表示满意或非常满意,8%为中立,9%不满意。大多数女性在术后的自尊、压力和情绪稳定性方面出现良好的效果或无明显变化(分别为88%、83%和88%)。尽管总体满意度很高,但仍有33%表示身体形象不佳,26%表示缺乏女性感,23%表示对性关系有负面影响。手术方式也会影响满意度。作者将此差异归因于非预期再手术的高发率,其中皮下乳腺切除术组(43%)与单纯乳腺切除术组(15%)(P<0.0001)。这项研究的局限性主要与参与者进行手术的时间段有关(即是否有手术重建的选择)。
这些女性中无一例行BRCA1/BRCA2致病性突变的基因检测。这项研究表明,虽然大多数女性对对侧RRM满意度较高,但所有女性都至少报告了一种不良影响。
对137例BRCA携带者进行回顾性研究,探讨保留乳头乳晕复合体(NAC)在双侧RRM患者中社会心理的影响
研究发现,由于女性接受的RRM类型不同,她们的身体形象和性生活方面存在显著差异。行NAC的女性对乳房外观更满意(72%比61%),对手术结果更满意(85%比74%),并且比对照组女性有更高的性生活幸福感(68%比52%)。两组患者在癌症相关的焦虑、担忧、抑郁或风险认知方面均无差异。在BRCA携带者中行NAC的肿瘤学结果并不劣于未行保留乳头乳晕复合体的RRM患者。
(更多信息,请参阅RRM部分的相关内容。)
另一项研究比较了1979至1999年间行双侧RRM的195名女性与117名选择筛查乳腺癌高危女性的长期生活质量结果。两组间的社会心理结果均无统计学显著差异。选择手术的人中有84%对他们的决定表示满意。 来自手术和筛查组的女性中,有61%的女性对自己的生活质量非常满意。
在一项针对RRM与即刻乳房重建相关的社会心理结果研究中,61名高危女性(其中27名致病性突变携带者,其他为高危家族史女性),31名有乳腺癌病史,术后3-4年进行评估。
本项研究的设计问题均采用是与否来回答,发现手术是可接受的,83%参与者报告她们的重建手术结果和预期一致,90%患者报告在术前已了解到足够信息,无一例报告后悔手术决定,所有女性均称,如果不得不再做一次,她们仍然会做出同样选择。对结果的满意程度从对乳房形状的74%到对疤痕外观的89%不等。将该组患者与普通人群的生活质量指标(简表36健康调查问卷[SF-36];医院焦虑和抑郁量表问卷评分)进行比较,结果显示该组女性的生活质量未见降低。
对FORCE网站上由21名接受RRM的高风险女性(BRCA1 / BRCA2阳性)发布的病历资料进行的定性研究表明,这些女性对手术的预期和负面反应来自朋友和家人,她们以保持情感支持和自我保护的方式管理信息披露。许多女性对侵袭性乳腺癌的想法和忧虑表示缓解,并对手术的美容效果感到满意。
相比之下,另一项研究则对684名在评估前曾行双侧或对侧RRM(平均9年)的女性的长期社会心理结果进行了研究。
大多数女性(59%)也接受了重建手术。有趣的是,根据利克特量表(Likert scale),85%女性表示对行RRM的决定感到满意或非常满意。然而,在定性访谈中,大量女性陈述了与手术相关的不满或负面社会心理反应。当女性报告仍对乳腺癌风险感到焦虑和/或报告对她们的身体形象、疼痛和性行为有负面影响时,作者将这些反应编码为阴性。79%女性提供负面评论,84%女性提供混合评论(满意和不满意的混合),指出她们对自己的决定要么满意,要么非常满意。接受双侧乳腺切除术的女性比接受对侧乳房切除术的女性发表否定和混杂评论高出一倍。最受关注的焦点是身体形象、乳房植入物问题、术后疼痛和性行为。作者指出,接受对侧手术的患者曾经已进行过癌症治疗,而接受双侧乳腺切除术的患者既往无任何治疗,这可能部分解释了两组患者满意度的差异。这些发现表明,随着时间的推移,女性对RRM的满意度可能会因其复杂的反应而减弱。
在对接受过或正在考虑进行RRM的108名女性进行的定性研究中,超过一半的RRM的女性认为术前咨询心理医生值得考虑。 近三分之二的人认为手术后咨询也是恰当的。 所有考虑拟行RRM的女性都认为术前的心理咨询会有助于决策。
一项回顾性自我管理的调查对40名在35-74岁行RRSO的女性(57.5%女性年龄<50岁)进行分析,该组女性因有卵巢癌家族史而通过安大略省卫生部行手术,结果显示RRSO显著降低了卵巢癌风险。57%将感知风险的降低视为RRSO获益(35%对RRSO获益未予评论),49%报告说她们会再次行RRSO以降低癌症风险。这组女性患者的总体生活质量评分与绝经期女性或参与激素研究的女性一致。
59名接受RRSO的女性在术后24个月进行了生活质量评估。
该组女性的总体生活质量与普通人群和乳腺癌幸存者相似,约20%报告患有抑郁症。在报告阴道干涩和性交不适的女性中,30%更有可能报告对手术不满意。
一项加拿大的前瞻性研究就RRSO对114名已知BRCA1/BRCA2病理型突变女性术前和术后1年的更年期症状和性功能的影响进行评估
不论有何症状报道,对接受RRSO的决定的满意度都很高。那些在手术时处于绝经前的患者(n=75)经历了症状的恶化和性功能的下降。激素替代疗法解决了阴道干燥和性交困难,但没有降低性快感。激素替代疗法也会导致中度到重度潮热。
该组患者的其他研究发现,在一年前接受RRSO治疗的127名女性中(75名BRCA1致病性突变;52名BRCA2致病性突变)中,大多数患者认为RRSO降低了乳腺癌和卵巢癌风险。
这组患者对卵巢癌风险有着广泛的认识。20%BRCA1和BRCA2致病性突变携带者认为经RRSO后卵巢癌风险已经完全消除;其他患者在手术前后对卵巢癌风险会有夸大认识。在这些女性中,一小部分在术后3年左右接受了进一步调查,在这段较长的时间内,她们的风险认知并未明显改变。 这些结果表明,对卵巢癌风险的重要误解是RRSO后可能持续存在。可能需要进行额外的遗传教育和咨询。
一项较大的研究对比了接受定期妇科筛查(GS)的卵巢癌高风险女性与接受RRSO的卵巢癌高风险女性的生活质量。共有846名高危女性,其中44%接受了RRSO,56%选择了GS,均完成了调查问卷,评估了生活质量,癌症癌症特异性焦虑,内分泌症状和性功能。
RRSO组女性的平均术后时间为2.8±1.9年,GS组女性初次高危管理后的平均间隔时间为4.3年。RRSO组和GS组的总体生活质量无统计学差异。与GS组相比,接受RRSO的女性性功能较差,内分泌症状较多,如阴道干涩、性交困难和潮热。接受RRSO治疗的女性患乳腺癌和卵巢癌的焦虑程度较低,对癌症风险的认识更为中肯。
对接受遗传咨询的遗传性乳腺癌和卵巢癌风险女性(N=182)进行调查,了解她们对选择RRSO或定期筛查(PS)(TVUS骨盆检查和CA-125检测,每年2次)控制卵巢癌风险的满意度。
两种选择的总体满意度都非常高,但满意度最高的病例在RRSO组,而不是PS。尚无人口统计学或临床因素能区分满意度。在选择PS的病例中,其决策的矛盾心理较明显。
一项回顾性研究对98例BRCA致病性突变携带者于RRSO术前就术后潜在症状进行遗传学咨询的进行评估。
患者行RRSO手术时的平均年龄为45.5岁(范围,32-63岁)。85%患者在得知携带BRCA突变后进行RRSO,48.0%在手术时未绝经。参与RRSO人群经常报告的术后症状包括阴道干涩症状(52.1%)、性生活的兴趣改变(50.0%)、睡眠障碍(46.7%)、性生活变化(43.9%)和潮热(42.9%)。通常只有阴道干涩和潮热改变最为明显。尽管96%会再次接受手术,但该组患者报告说,手术对他们的性生活(59.2%),冠状动脉心脏疾病(57.1%)的风险,和性别咨询(57.1%)的可用性的影响的讨论将很有帮助。
一项
针对216名自我转诊、高危(BRCA1/BRCA2致病性突变风险>10%)的遗传性乳腺癌家族成员行为的筛查研究,发现了很多筛查方面的问题。即使明知有家族性致病性突变,也不一定就能按照推荐的筛查方法实施。59% 50-64岁女性和83%40-49岁女性在前一年做过乳房X线检查。20%参与者曾做过CA-125检测,31%曾做过盆腔超声或TVUS检查。
针对该组人群进一步分析发现,尽管年轻携带者的乳腺癌筛查接受率较低,对107名获得有效BRCA检测结果的女性研究发现,乳腺癌筛查得到很好的应用。本研究未对低接受率的原因进行探讨。
一项对罹患乳腺癌/卵巢癌高风险女性进行筛查行为的调查表明,医生的推荐是坚持筛查的重要因素。
虽然采取基因检测的动机通常包括期望致病突变携带者将更符合乳腺和/或卵巢筛查的建议,
目前,关于基因检测的参与者是否会随着时间的推移而改变他们的筛查行为以及潜在影响这些行为其他变量(如保险覆盖率和医生的建议或态度)的数据较为有限,有一项英国研究评估了癌症遗传学咨询对筛查行为的影响,主要对来自4个癌症遗传学门诊的293名女性在咨询后进行了随诊12个月。
咨询后,BSE、CBE和乳腺钼靶检查显著增加;但是,在遵守建议方面存在差距:38%女性(35至49岁)在咨询后12个月内未行乳房X线检查。大多数女性均未按照推荐的时间和频率进行BSE检查。
这不仅对于检测呈阳性的女性,而且对于检测结果呈阴性但坚持筛查、收到不确定结果或选择拒收结果的女性来说,都是至关重要的问题。依从性实际上可能会随着遗传测试结果阴性而导致的感知风险的降低而降低。
此外,虽然对癌症相关的担忧与乳腺癌筛查行为之间的联系仍然存在一些疑问,但越来越多的证据似乎支持线性关系,而不是曲线关系。也就是说,在一段时间内,数据并不一致;一些数据支持这样的假设,即轻度到中度的担忧可能会增加依从性,而过度的担忧实际上可能会降低推荐的筛查方法的应用率。还有其他报告支持这样的观点,即线性关系更有可能;也就是说,更多的担忧会增加对筛查建议的依从性。然而,很少有研究跟踪女性在基因测试后评估她们的健康行为。因此,从理论上讲,导致担忧减少的阴性测试结果可能会带来筛查依从性降低。一项大型研究发现,除了BSE,患者对筛查的依从性与一般焦虑或筛查特异性焦虑无关。对于BSE,依从性与检查特异性焦虑呈负相关。
为明确这一潜在问题而开展的进一步研究将强调应进行综合遗传咨询,以讨论后续筛查的必要性。
有一些问题使该领域的研究进一步复杂化,比如基因检测之前,40或50岁以上女性的乳腺钼靶基线依从率。更具体地,基因检测前进行这种检测的女性,对识别这种筛选行为的显着差异能力可能受到对这种筛选行为的高依从性的影响, 在有乳腺癌家族病史且基因检测呈阳性的女性中,更容易发现乳腺钼靶显著的改变。 最后,随着时间的流逝,依存性可能会受到接受基因测试的妇女及其护理人员对许多上述筛查方法的效果的影响,例如年轻女性的乳房X线照片,BSE和卵巢癌筛查(定期阴道超声检查和血清CA- 125种措施)以及预防性干预措施的价值。
在接受乳腺癌和卵巢癌基因检测的女性中,筛选决策和依从性的问题是多个临床试验的主题,也是一个亟需研究的领域。
Psychosocial research in the context of cancer genetic testing helps to define psychological outcomes, interpersonal and familial effects, and cultural and community responses. This type of research also identifies behavioral factors that encourage or impede screening and other health behaviors. It can enhance decision making about risk-reduction interventions, evaluate psychosocial interventions to reduce distress and/or other negative sequelae related to risk notification and genetic testing, provide data to help resolve ethical concerns, and predict the interest in testing of various groups.
This section addresses psychosocial issues in hereditary breast and ovarian cancer syndromes. Psychosocial and screening issues related to gynecologic cancers associated with Lynch syndrome are discussed in the Psychosocial Issues in Hereditary Colon Cancer Syndromes section in the PDQ summary on Genetics of Colorectal Cancer.
Comparison of uptake rates among studies in which counseling and testing were offered is challenging because of differences in methodologies, including the sampling strategy used, the recruitment setting, and testing through a research protocol with high-risk cohorts or kindreds. In a systematic review of 40 studies conducted before 2002 that had assessed genetic testing utilization, uptake rates varied widely and ranged from 25% to 96%, with an average uptake rate of 59%.
Results of multivariate analysis found that BRCA1/BRCA2 genetic testing uptake was associated with having a personal or family history of breast or ovarian cancer, and with methodological features of the studies, including sampling strategies, recruitment settings, and how studies defined actual uptake versus the intention to have testing.
Other factors have been positively correlated with uptake of BRCA1/BRCA2 genetic testing, although these findings are not consistent across all studies. Psychological factors that have been positively correlated with testing uptake include greater cancer-specific distress and greater perceived risk of developing breast or ovarian cancer. Having more cancer-affected relatives also has been correlated with greater testing uptake.
Table 14 summarizes the uptake of genetic testing in clinical and research cohorts in the United States.
Study Citation | Study Population | Sample Size (N) | Uptake of GT | Predictors Associated With Uptake of GT | Comments |
---|---|---|---|---|---|
Schwartz et al. (2005) | Newly diagnosed and locally untreated breast cancer patients with ≥10% risk of having a BRCA1/BRCA2 pathogenic varianta | 231 | 177/231 (77%) underwent GT | Having decided on definitive local treatment. Women who were undecided on a definitive local treatment were more likely to be tested. | Testing was offered free of charge. |
34/231 (15%) had baseline interview but declined GT | |||||
Physician recommendation for testing. Women whose physician had recommended GT were more likely to be tested. | 38/177 chose to proceed with treatment before receiving test results. | ||||
20/231 declined baseline interview | |||||
Kieran et al. (2007) | Women who received GC between 2002 and 2004 | 250 | 88/250 (35%) underwent GT | Ability to pay for GT (entire cost or cost not covered by insurance). Nonuptake was 5.5 times more likely in women who could not afford testing. | 450 women received GC for breast and ovarian cancer risk during study period. 250 women were retrospectively identified as eligible and were mailed a study questionnaire. |
36/88 returned surveys | |||||
Ability to recall risk estimates that were provided post-GC. Nonuptake was 15.5 times more likely in women who could not recall their risk estimates. | All women had some form of insurance. | ||||
162/250 (65%) eligible | |||||
65/162 returned surveys | |||||
Susswein et al. (2008) | African American women and white women with breast cancer | 768 | 529/768 (69%) underwent GT | Race/ethnicity. African American women were less likely to be tested than were white women. | Sample obtained from a clinical database. Testing was offered free of charge when it was not covered by insurance. This effect for time of diagnosis was significant in the African American, but not white, subgroup. |
African American women: 77/132 (58%) underwent GT | |||||
Recent diagnosis. African American women who were recently diagnosed were more likely to be tested. | |||||
White women: 452/636 (71%) underwent GT | |||||
Olaya et al. (2009) | Patients referred for GT between 2001 and 2008b | 213 | 111/213 (52%) underwent GT | Personal history of breast cancer. Having a personal history was associated with 3 times greater odds of being tested. | Insurance coverage for testing was available for 91.1% (175/213) of patients. Of those who had coverage for GT, 51.4% underwent testing and 48.6% did not. Of those without coverage, 41.2% had GT and 58.9% did not. |
102/213 (48%) declined GT | Higher level of education. Those with a high school education or less had one-third the odds of being tested, compared with those with at least some college. | ||||
Levy et al. (2010) | Women aged 20–40 y with newly diagnosed early-onset breast cancer. | 1,474 | 446/1,474 (30%) underwent GT | Race/ethnicity. Women of Jewish ethnicity were 3 times more likely to be tested than were non-Jewish white women. African American and Hispanic women were significantly less likely to receive testing than were non-Jewish white women. | Sample obtained from a national database of commercially insured individuals. |
Jewish women: 18/32 (56%) underwent GT | Home location. Women living in the south were more likely to be tested than were women living in the northeast. | ||||
African American women: 10/82 (12%) underwent GT | Insurance type. Women with point-of-service plans were more likely to be tested than were women with HMO plans. | ||||
Recent diagnosis. Women diagnosed in 2007 were 3.8 times more likely to be tested than were women diagnosed in 2004. | |||||
GC = genetic counseling; HMO = health maintenance organization. | |||||
aSelf-report as data source. | |||||
bMedical records as data source. |
Several studies conducted in non-U.S. settings have examined the uptake of genetic testing.
In studies examining the uptake of testing among at-risk relatives of carriers of BRCA1/BRCA2 pathogenic variants, uptake rates have averaged below 50% (range, 36%–48%), with higher uptake reported among female relatives than in male relatives. Other factors associated with higher uptake of testing were not consistently reported among studies but have most commonly included being a parent and wanting to learn information about a child’s risk.
In reviews that have examined the cumulative evidence concerning the predictors of uptake of BRCA1/BRCA2 genetic testing, important predictors of testing uptake include older age, Ashkenazi Jewish (AJ) heritage, unmarried status, a personal history of breast cancer, and a family history of breast cancer. Studies recruiting participants in hospital settings had significantly higher recruitment rates than did studies recruiting participants in community settings. Studies that required an immediate decision to test, rather than allowing delayed decision making, tended to report higher uptake rates.
However, there is evidence that women diagnosed with breast cancer are equally satisfied with genetic counseling (including information received and strength and timing of physician recommendations for counseling), whether they received genetic counseling before or after their definitive surgery for breast cancer.
Another review
found that uptake of genetic testing for BRCA1/BRCA2 pathogenic variants was related to psychological factors (e.g., anxiety about breast cancer and perceived risk of breast cancer) and demographic and medical factors (e.g., history of breast cancer or ovarian cancer, presence of children, and higher number of affected first-degree relatives [FDRs]). Family members with a known BRCA1/BRCA2 pathogenic variant were more likely to pursue testing; those with more extensive knowledge of BRCA1/BRCA2 testing, heightened risk perceptions, beliefs that mammography would promote health benefit, and high intentions to undergo testing were more likely to follow through with testing.
In a review of racial/ethnic differences that affect uptake of BRCA1/BRCA2 testing, intention to undergo genetic testing in African American women was related to having at least one FDR with breast cancer or ovarian cancer, higher perceived risk of being a carrier, and less anticipatory guilt about the possibility of being a gene carrier.
A systematic review found that certain ethnic minority groups including African Americans and Hispanics had more negative views and greater concerns about genetic counseling and testing when compared with whites. African Americans and Hispanics were more likely to believe genetic testing could be used to show their ethnic group was inferior to other groups. Additionally, African Americans and Hispanics were found to have low awareness and knowledge about the importance of genetics in cancer, BRCA status, and genetic testing.
Reasons cited for following through with testing included a desire to learn about a child's risk, to feel relief from uncertainty, to inform screening or risk-reducing surgery decisions, and to inform important life decisions such as marriage and childbearing.
Among African American women, the most important reason for testing included motivation to help other relatives decide on genetic testing.
Physician recommendation may be another motivator for testing. In a retrospective study of 335 women considering genetic testing, 77% reported that they wanted the opinion of a genetics physician about whether they should be tested, and 49% wanted the opinion of their primary care provider.
However, there is some evidence of referral bias favoring those with a maternal family history of breast cancer or ovarian cancer. In a Canadian retrospective review of 315 patients, those with a maternal family history of breast cancer or ovarian cancer were 4.9 times (95% confidence interval, 3.6–6.7) more likely to be referred for a cancer genetics consultation by their physician than were those with a paternal family history (P < .001).
Studies have found that physicians may not adequately assess paternal family history
or may underestimate the significance of a paternal family history for genetic risk.
Other studies have shown that physician referral of patients who meet U.S. Preventive Services Task Force guidelines for BRCA genetic counseling has been suboptimal.
The uptake of BRCA testing to inform surgical treatment decisions when offered appears to be high in research cohorts;
however, findings from other studies suggest that testing is underutilized in clinical practice to inform breast cancer treatment decisions.
Barriers to the use of BRCA testing to inform surgical treatment decisions, including lack of physician referral of newly diagnosed patients for genetic counseling, type of insurance coverage (such as Medicare or Medicaid), and challenges in the timing and coordination of testing, have been reported.
In a randomized trial that provided proactive rapid genetic counseling (delivery of genetic counseling prior to surgery) compared with usual care for patients with newly diagnosed breast cancer, results suggested that although genetic counseling uptake was higher in the intervention arm, this did not translate into higher rates of genetic testing, receipt of results before surgery, or bilateral mastectomy decisions.
Insurance coverage is an important consideration for individuals deciding whether to undergo genetic testing. (Refer to the Insurance coverage section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information.)
There are limited data on uptake of genetic counseling and testing among nonwhite populations, and further research will be needed to define factors influencing uptake in these populations.
The uptake of BRCA testing appears to vary across some racial/ethnic groups. A few studies have compared uptake rates between African American and white women.
In a case-control study of women who had been seen in a university-based primary care system, African American women with family histories of breast cancer or ovarian cancer were less likely to undergo BRCA1/BRCA2 testing than were white women who had similar histories.
In another study among breast cancer patients who were counseled about BRCA1/BRCA2 risk in a clinical setting, lower uptake was reported among African American women than among white women.
Notably, the racial differences observed in these studies do not appear to be explained by factors related to cost, access to care, risk factors for carrying a BRCA1 or BRCA2 pathogenic variant, or differences in psychosocial factors, including risk perceptions, worry, or attitudes toward testing.
Several studies have examined uptake or “acceptance” of BRCA testing among African Americans enrolled in genetic research programs. Among study enrollees from an African American kindred in Utah, 83% underwent BRCA1 testing.
Age, perceived risk of being a carrier, and more extensive cancer knowledge predicted testing acceptance. Another study that recruited African American women through physician and community referrals reported a BRCA1/BRCA2 testing acceptance rate of 22%.
Predictors of test acceptance included having a higher probability of having a pathogenic variant, being married, and being less certain about one’s cancer risk. Finally, a third study that recruited at-risk African American women from an urban cancer screening clinic found that acceptors of BRCA testing were more knowledgeable about breast cancer genetics and perceived fewer barriers to testing, including negative emotional reactions, stigmatization concerns, and family-related guilt.
While these are independent predictors of genetic testing uptake, they do not explain the disparities in testing uptake across different ethnic groups. What may explain these differences are several attitudes and beliefs held about testing by individuals from diverse populations.
Work examining attitudes toward breast cancer genetic testing in Latino and African American populations indicates limited knowledge and awareness about testing but a generally receptive view once they are informed; in comparison with whites, Latino and African American populations have relatively more concerns about testing.
For example, in a qualitative study with 51 Latino individuals unselected for risk status, important findings included the fact that participants were highly interested in genetic testing for inherited cancer susceptibility, despite very limited knowledge about genetics. One important barrier involved secrecy or embarrassment about family discussions of cancer and genetics, which could be addressed in intervention strategies.
Another qualitative study with 54 Latina women at risk of hereditary breast cancer showed that knowledge about BRCA1/BRCA2 counseling was low, although the women were interested in learning more about counseling to gain risk information for family members. Barriers to counseling included life demands, cost, and language issues.
A telephone survey of 314 patients from an inner-city network of Pittsburgh, Pennsylvania, health centers, 50% of whom were African American, found that most participants (57%) (both African Americans and whites) felt that genetic testing to evaluate disease risk was a good idea; however, more African Americans than whites thought that genetic testing would lead to racial discrimination (37% vs. 22%, respectively) and that genetics research was unethical and tampered with nature (20% vs. 11%, respectively).
Finally, in a study of 222 women in Savannah, Georgia, where most had neither a personal history (70%) nor a family history (60%) of breast cancer, African American women (who comprised 26% of the sample) were less likely to be aware of breast cancer genes and genetic testing. Awareness was also related to higher income, higher education level, and having a family breast cancer history. However, 74% of the entire sample expressed willingness to be tested for breast cancer susceptibility.
In a sample of 146 African American women meeting criteria for BRCA1/BRCA2 pathogenic variant testing, women born outside the United States reported higher levels of anticipated negative emotional reactions (e.g., fear, hopelessness, and lack of confidence that they could emotionally handle testing). Higher levels of breast cancer–specific distress were associated with anticipated negative emotional reactions, confidentiality concerns, and anticipated guilt regarding the family impact of breast cancer genetic testing.
A future orientation (e.g., "I often think about how my actions today will affect my health when I am older") was associated with overall perceived benefits of breast cancer genetic testing in this population (n = 140); however, future orientation was also found to be positively associated with family-related cons of testing, including family guilt and worry regarding the impact of testing on the family.
There are racial differences in provider discussion and patient uptake of genetic testing for variants in BRCA1/BRCA2. A study of women aged 18 to 64 years and diagnosed with invasive breast cancer between 2007 and 2009 found that, even after adjusting for pathogenic variant risk, African American women were less likely to report having received a physician recommendation for genetic testing. There was no difference across all races in concerns that BRCA1/BRCA2 testing was too expensive and only minimal differences in testing attitudes or insurance concerns were found, none of which influenced testing uptake.
A study of breast or ovarian cancer survivors (N = 50) eligible for BRCA1/BRCA2 genetic testing found that 48% were referred for genetic counseling and testing and/or had undergone genetic testing. Individuals with higher breast cancer genetics knowledge and higher self-efficacy were more likely to have engaged in genetic counseling and testing.
In a study of women with invasive breast cancer diagnosed before age 50 years between 2009 and 2012 who were identified through the Florida Cancer Data System state registry and eligible for BRCA1/BRCA2 genetic testing on the basis of existing guidelines, African Americans were less likely to report a discussion with their health care provider and undergo genetic testing.
The same study found similar overall testing rates in Hispanic (61%) and non-Hispanic (65%) whites. However, testing rates were lower among Hispanics who spoke primarily Spanish at home (50% Spanish speaking vs. 69% English speaking; P = .0009), and in general, Hispanics were less likely to have been referred for genetic testing.
However, this finding is not consistent across all studies. In a study of women aged 20 to 79 years with ductal carcinoma in situ or invasive breast cancer identified through the Surveillance, Epidemiology, and End Results (SEER) registry in Georgia and Los Angeles County, all eligible for BRCA1/BRCA2 genetic testing on the basis of existing guidelines, no ethnic differences were detected in receipt of genetic counseling or physician-directed discussion about genetic testing.
There is evidence that primary reasons for declining testing involves being childless, which reduces any family motivations for testing; and concerns about the negative ramifications of testing, including difficulty retaining insurance or concerns about personal health.
Limited data are available about the characteristics of at-risk individuals who decline to be tested or have never been tested. It is difficult to access samples of test decliners because they may be reluctant to participate in research studies. Studies of genetic testing uptake are difficult to compare because people may decline at different points and with different amounts of pretest education and counseling. One study found that 43% of affected and unaffected individuals from hereditary breast/ovarian cancer families who completed a baseline interview regarding testing declined to be tested. Most individuals who declined testing chose not to participate in educational sessions. Decliners were more likely to be male and be unmarried, and have fewer relatives with breast cancer. Decliners who had high levels of cancer-related stress had higher levels of depression. Decliners lost to follow-up were significantly more likely to be affected with cancer.
Another study looked at a small number (n = 13) of women decliners who carried a 25% to 50% probability of harboring a BRCA pathogenic variant; these nontested women were more likely to be childless and to have higher levels of education. This study showed that most women decided not to undergo the test after serious deliberation about the risks and benefits. Satisfaction with frequent surveillance was given as one reason for nontesting by most of these women.
Other reasons for declining included having no children and becoming acquainted with breast/ovarian cancer in the family relatively early in their lives.
A third study evaluated characteristics of 34 individuals who declined BRCA1/BRCA2 testing in a large multicenter study in the United Kingdom. Decliners were younger than a national sample of test acceptors, and female decliners had lower mean scores on a measure of cancer worry. Although 78% of test decliners/deferrers felt that their health was at risk, they reported that learning about their BRCA1/BRCA2 pathogenic variant status would cause them to worry about the following:
Apprehension about the impact of the test result was a more important factor in the decision to decline testing than were concrete burdens such as time required to travel to a genetics clinic and time spent away from work, family, and social obligations.
In 15% (n = 31) of individuals from 13 hereditary breast and ovarian cancer families who underwent genetic education and counseling and declined testing for a documented pathogenic variant in the family, positive changes in family relationships were reported—specifically, greater expressiveness and cohesion—compared with those who pursued testing.
Testing for BRCA1/BRCA2 pathogenic variants has been almost universally limited to adults older than 18 years. The risks of testing children for adult-onset disorders, such as breast and ovarian cancers, as inferred from developmental data on children’s medical understanding and ability to provide informed consent, have been outlined in several reports.
Studies suggest that persons who have undergone BRCA1/BRCA2 genetic testing or who are adult offspring of persons who have had testing are generally not in favor of testing minors.
Although the data are limited, research suggests that males, pathogenic variant noncarriers, and those whose mothers did not have personal histories of breast cancer may be more likely to favor genetic testing in minors in general.
Of those who had minor children at the time the study was conducted, only 17% stated a preference for having their own children tested. Concerns regarding testing of minors included psychological risks and insufficient maturity. Potential benefits included the ability to influence health behaviors.
No data exist on the testing of children for BRCA1/BRCA2 pathogenic variants, although some researchers believe it is necessary to test the validity of assumptions underlying the general prohibition of testing children for genetic variants associated with breast and ovarian cancers and other adult-onset diseases.
In one study, 20 children (aged 11–17 y) of a selected group of mothers undergoing genetic testing (80% of whom previously had breast cancer and all of whom had discussed BRCA1/BRCA2 testing with their children) completed self-report questionnaires on their health beliefs and attitudes toward cancer, feelings related to cancer, and behavioral problems.
Ninety percent of children thought they would want cancer risk information as adults; half worried about themselves or a family member developing cancer. There was no evidence of emotional distress or behavioral problems.
The emerging literature in this area suggests that risk perceptions, health beliefs, psychological status, and personality characteristics are important factors in decision making about breast/ovarian cancer genetic testing. Many women presenting at academic centers for BRCA1/BRCA2 testing arrive with a strong belief that they have a pathogenic variant, having decided they want genetic testing, but possessing little information about the risks or limitations of testing.
Most mean scores of psychological functioning at baseline for subjects in genetic counseling studies were within normal limits.
Nonetheless, a subset of subjects in many genetic counseling studies present with elevated anxiety, depression, or cancer worry.
Identification of these individuals is essential to prevent adverse outcomes. In a study of 205 women pursuing genetic counseling, interactions among cancer worry, breast cancer risk perception, and perceived severity of having a breast cancer genetic variant were found such that those with high worry, high breast cancer risk perception, and low perceived severity were twice as likely to follow through with BRCA1/BRCA2 testing than others.
A general tendency to overestimate inherited risk of breast and ovarian cancer has been noted in at-risk populations, in cancer patients, in spouses of breast and ovarian cancer patients, and among women in the general population.
but underestimation of breast cancer risk in higher-risk and average-risk women also has been reported.
This overestimation may encourage a belief that BRCA1/BRCA2 genetic testing will be more informative than it is currently thought to be. Some evidence exists that even counseling does not dissuade women at low to moderate risk from the belief that BRCA1 testing could be valuable.
Overestimation of both breast and ovarian cancer risk has been associated with nonadherence to physician-recommended screening practices.
A meta-analysis of 12 studies of outcomes of genetic counseling for breast/ovarian cancer showed that counseling improved the accuracy of risk perception.
Women appear to be the prime communicators within families about the family history of breast cancer.
Higher numbers of maternal versus paternal transmission cases are reported,
likely due to family communication patterns, to the misconception that breast cancer risk can only be transmitted through the mother, and to the greater difficulty in recognizing paternal family histories because of the need to identify more distant relatives with cancer. In an analysis of 2,505 women participating in the Family Healthware Impact Trial,
not only was evidence of underreporting of paternal family history identified, but also women reported a lower level of perceived breast cancer risk with a paternal versus maternal breast cancer family history.
Physicians and counselors taking a family history are encouraged to elicit paternal and maternal family histories of breast, ovarian, or other associated cancers.
The accuracy of reported family history of breast or ovarian cancer varies; some studies found levels of accuracy above 90%,
with others finding more errors in the reporting of cancer in second-degree or more distant relatives
or in age of onset of cancer.
Less accuracy has been found in the reporting of cancers other than breast cancer. Ovarian cancer history was reported with 60% accuracy in one study compared with 83% accuracy in breast cancer history.
Providers should be aware that there are a few published cases of Munchausen syndrome in reporting of false family breast cancer history.
Much more common is erroneous reporting of family cancer history due to unintentional errors or gaps in knowledge, related in some cases to the early death of potential maternal informants about cancer family history.
(Refer to the Taking a Family History section of the Cancer Genetics Risk Assessment and Counseling summary for more information.)
Targeted written,
video, CD-ROM, interactive computer programs and websites,
and culturally targeted educational materials
may be effective and efficient methods of increasing knowledge about the pros and cons of genetic testing. Such supplemental materials may allow more efficient use of the time allotted for pretest education and counseling by genetics and primary care providers and may discourage individuals without appropriate indication of risk from seeking genetic testing.
Counseling for breast cancer risk typically involves individuals with family histories that are potentially attributable to BRCA1 or BRCA2. It also, however, may include individuals with family histories of Li-Fraumeni syndrome, ataxia-telangiectasia, Cowden syndrome, or Peutz-Jeghers syndrome.
(Refer to the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section of this summary for more information.)
Management strategies for carriers may involve decisions about the nature, frequency, and timing of screening and surveillance procedures, chemoprevention, risk-reducing surgery, and use of hormone replacement therapy (HRT). The utilization of breast conservation and radiation as cancer therapy for women who are carriers may be influenced by knowledge of pathogenic variant status. (Refer to the Clinical Management of Carriers of BRCA Pathogenic Variants section of this summary for more information.)
Counseling also includes consideration of related psychosocial concerns and discussion of planned family communication and the responsibility to warn other family members about the possibility of having an increased risk of breast, ovarian, and other cancers. Data suggest that individual responses to being tested as adults are influenced by the results status of other family members.
Management of anxiety and distress are important not only as quality-of-life factors, but also because high anxiety may interfere with the understanding and integration of complex genetic and medical information and adherence to screening.
Formal, objective evaluation of these outcomes are well documented. (Refer to the Emotional Outcomes and Behavioral Outcomes sections of this summary for more information.)
Published descriptions of counseling programs for BRCA1 (and subsequently for BRCA2) testing include strategies for gathering a family history, assessing eligibility for testing, communicating the considerable volume of relevant information about breast/ovarian cancer genetics and associated medical and psychosocial risks and benefits, and discussion of specialized ethical considerations about confidentiality and family communication.
Participant distress, intrusive thoughts about cancer, coping style, and social support were assessed in many prospective testing candidates. The psychosocial outcomes evaluated in these programs have included changes in knowledge about the genetics of breast/ovarian cancer after counseling, risk comprehension, psychological adjustment, family and social functioning, and reproductive and health behaviors.
A Dutch study of communication processes and satisfaction levels of counselees going through cancer genetic counseling for inherited cancer syndromes indicated that asking more medical questions (by the counselor), providing more psychosocial information, and longer eye contact by the counselor were associated with lower satisfaction levels. The provision of medical information by the counselor was most highly related to satisfaction and perception that needs have been fulfilled.
Many of the psychosocial outcome studies involve specialized, highly selected research populations, some of which were utilized to map and clone BRCA1 and BRCA2. One such example is K2082, an extensively studied kindred of more than 800 members of a Utah Mormon family in which a BRCA1 pathogenic variant accounts for the observed increased rates of breast and ovarian cancer. A study of the understanding that members of this kindred have about breast/ovarian cancer genetics found that, even in breast cancer research populations, there was incomplete knowledge about associated risks of colon and prostate cancer, the existence of options for RRM and RRSO, and the complexity of existing psychosocial risks.
A meta-analysis of 21 studies found that genetic counseling was effective in increasing knowledge and improved the accuracy of perceived risk. Genetic counseling did not have a statistically significant long-term impact on affective outcomes including anxiety, distress, or cancer-specific worry and the behavioral outcome of cancer surveillance activities.
These prospective studies, however, were characterized by a heterogeneity of measures of cancer-specific worry and inconsistent findings in effects of change from baseline.
Although there were initial concerns about the possibility of adverse emotional consequences from BRCA testing, most studies conducted over the years have shown low levels of psychological distress among both carriers and noncarriers, particularly over the longer term.
In a meta-analysis examining cancer-specific distress over short (0–4 weeks), moderate (5–24 weeks), and long (25–52 weeks) periods of time since the receipt of testing results, carriers were found to demonstrate increased levels of distress shortly after receiving results, with levels returning to baseline within moderate and long periods of time.
In contrast, noncarriers and those with inconclusive results showed reduced levels of distress over time.
Psychological distress patterns were found to vary as a function of several factors, including the cancer history of the individual and the country within which the study was conducted. Carriers with a personal history of cancer experienced small decreases in distress over time, whereas no changes were observed among carriers without a personal history of cancer. Among individuals with inconclusive results, greater decreases in distress were observed among those without a cancer history than among those with a cancer history. Among noncarriers, those in the United States experienced significantly greater decreases in psychological distress than noncarriers from Europe and Australia. A study conducted in Austria noted that certain subgroups of counselees experienced greater distress, including those who were older, had a more recent cancer diagnosis, or those who had received counseling but declined BRCA testing.
Several studies have reported on emotional outcomes over longer follow-up periods (i.e., greater than 12 months after disclosure) than those reported in the meta-analysis described above.
In a U.K. study, cancer-related worry did not differ between carriers and noncarriers at 3 years of follow-up.
Two U.S.-based studies published since the meta-analytic review
have reported similar findings among women who were surveyed more than 3 years after receipt of BRCA test results.
In a cross-sectional study,
167 women who were surveyed more than 4 years after receiving BRCA test results reported low levels of genetic testing–specific concerns, as measured using the Multidimensional Impact of Cancer Risk Assessment Scale.
In multivariate regression models, carriers of pathogenic variants were significantly more likely to experience distress than were noncarriers. In a second study,
464 women were followed prospectively for a median of 5 years (range, 3.4–9.1 y) after testing. Among both affected and unaffected participants, BRCA carriers reported significantly higher levels of distress, uncertainty (affected only), perceived stress (affected only), and lower positive testing experiences (unaffected only) than women who received negative results for a known pathogenic variant in the family.
Although both studies
reported greater distress among BRCA carriers than among noncarriers, the level of distress was not reflective of clinically significant dysfunction.
Although most studies have reported that a positive BRCA test result has a relatively minimal impact on psychological distress, many of these studies were conducted among families with a strong family history of breast or ovarian cancer who underwent extensive pretest genetic counseling. Therefore, emotional responses may not generalize to individuals who test under different contexts. For example, individuals who are tested with population BRCA screening may not have a family history of cancer.
Although pretest genetic counseling is recommended, this is not always done when genetic testing is ordered by nongenetic providers or directly through commercial companies.
For example, in a Canadian study of 2,080 Jewish women who participated in a population-based genetic screening study to test for three BRCA pathogenic variants common in families of Jewish heritage, women were not offered in-person genetic counseling but were given a pamphlet on genetic testing for BRCA1/BRCA2 before they provided a DNA sample. One year after genetic testing, women who were positive for a pathogenic variant (n = 18) showed significant increases in cancer-specific distress, whereas no changes in distress were observed among women who were negative for a pathogenic variant.
The mean distress score on the Impact of Event Scale for the 18 women with a known pathogenic variant was 25.3 (range, 2–51); 10 of 18 women (56%) scored within moderate (26–43) (n = 7) or severe (44+) (n = 3) ranges. It is unclear from this study whether the increase in distress observed at 1 year of follow-up was due to the lack of in-person genetic counseling, or whether the lower levels of distress at baseline observed were because the women in the study were low risk but eligible for testing because of their ancestry. A follow-up study with this cohort found that distress decreased between 1 to 2 years after testing and that changes in distress varied by risk-reduction options undertaken by carriers. Specifically, those who had undergone risk-reducing mastectomy or oophorectomy experienced significant decreases in distress compared with those who did not have either surgery.
Another smaller qualitative study also supports these findings.
Similarly, the impact of direct-to-consumer (DTC) BRCA testing through commercial companies requires further evaluation. Case studies have reported adverse emotional responses after receipt of a positive BRCA result from DTC genetic testing, suggesting the need for further evaluation of the emotional outcomes of women undergoing genetic testing through commercial companies.
Only one study, conducted by a commercial company, has attempted to evaluate the impact of BRCA testing in this context.
A total of 32 individuals (16 women and 16 men) who tested positive for one of three BRCA founder pathogenic variants common in Ashkenazi Jews completed semi-structured interviews. None of the carriers reported extreme anxiety, although some experienced moderate anxiety (13%) or initial disappointment and anxiety that dissipated over time (28%). These findings should be interpreted with caution given that only 24% (32 of 136) of invited carriers of BRCA pathogenic variants participated in the study, raising concerns about selection bias.
Despite evidence of a short-term increase in distress after the receipt of genetic testing results, any adverse responses to a positive carrier status dissipate within 12 months.
Additional research is needed to examine emotional outcomes for those who are not provided genetic counseling before testing.
It is increasingly common for women with breast cancer to pursue genetic counseling and testing at the time of diagnosis to assist with treatment decision making. (Refer to the Benefits of offering genetic testing at the time of cancer diagnosis section in the Introduction section of this summary for more information.) Although concerns have been raised about the adverse psychological implications of offering rapid genetic counseling and testing between diagnosis and surgery,
other studies,
including a randomized trial,
have provided evidence indicating no additional adverse psychological effects in newly diagnosed breast cancer patients. One randomized controlled trial found that patients undergoing rapid genetic counseling and testing felt more actively involved in treatment decision making than those receiving standard care.
However, qualitative research on 20 newly diagnosed breast cancer patients found that some subgroups of these patients may have more difficulty coping with BRCA test results, such as carriers who have no family history of cancer; those who do not have an affected relative with whom they can identify; and higher risk women who receive uninformative negative BRCA results.
Family communication about genetic testing for cancer susceptibility, and specifically about the results of BRCA1/BRCA2 genetic testing, is complex. Gender appears to be an important variable in family communication and psychological outcomes. Studies have documented that female carriers are more likely to disclose their status to other family members (especially sisters and children aged 14–18 y) than are male carriers.
Among males, noncarriers were more likely than carriers to tell their sisters and children the results of their tests. BRCA1/BRCA2 carriers who disclosed their results to sisters had a slight decrease in psychological distress, compared with a slight increase in distress for carriers who chose not to tell their sisters. One study found that men reported greater difficulty disclosing a known pathogenic variant to family members than women (90% vs. 70%).
Family communication of BRCA1/BRCA2 test results to relatives is another factor affecting participation in testing. There have been more studies of communication with FDRs and second-degree relatives than with more distant family members. Studies have investigated the process and content of communication among sisters about BRCA1/BRCA2 test results.
Study results suggest that both carriers of pathogenic variants
and women with uninformative results
communicate with sisters to provide them with genetic risk information. Similar findings were reported in women with uninformative results disclosing test results to their daughters.
Among relatives with whom genetic test results were not discussed, the most important reason given was that the affected women were not close to their relatives
or had a poor relationship with them.
Studies found that women with a BRCA pathogenic variant more often shared their results with their mother and adult sisters and daughters than with their father and adult brothers and sons.
A study that evaluated communication of test results to FDRs at 4 months postdisclosure found that women aged 40 years or older were more likely to inform their parents of test results compared with younger women. Participants also were more likely to inform brothers of their results if the BRCA pathogenic variant was inherited through the paternal line.
Another study found that disclosure was limited mainly to FDRs, and dissemination of information to distant relatives was problematic.
Age was a significant factor in informing distant relatives with younger patients being more willing to communicate their genetic test result.
Additionally, one study found that lower genetic worry, higher interest in genomic information, carrying a BRCA1 or BRCA2 pathogenic variant, or having never been married was associated with communication to more family members.
In contrast, a longer time interval since diagnosis was associated with communication to fewer family members.
A few in-depth qualitative studies have looked at issues associated with family communication about genetic testing. Although the findings from these studies may not be generalizable to the larger population of at-risk persons, they illustrate the complexity of issues involved in conveying hereditary cancer risk information in families.
On the basis of 15 interviews conducted with women attending a familial cancer genetics clinic, the authors concluded that while women felt a sense of duty to discuss genetic testing with their relatives, they also experienced conflicting feelings of uncertainty, respect, and isolation. Decisions about whom in the family to inform and how to inform them about hereditary cancer and genetic testing may be influenced by tensions between women's need to fulfill social roles and their responsibilities toward themselves and others.
Another qualitative study of 21 women who attended a familial breast and ovarian cancer genetics clinic suggested that some women may find it difficult to communicate about inherited cancer risk with their partners and with certain relatives, especially brothers, because of those persons’ own fears and worries about cancer.
This study also suggested that how genetic risk information is shared within families may depend on the existing norms for communicating about cancer in general. For example, family members may be generally open to sharing information about cancer with each other, may selectively avoid discussing cancer information with certain family members to protect themselves or other relatives from negative emotional reactions, or may ask a specific relative to act as an intermediary to disclosure of information to other family members.
The potential importance of persons outside the family, such as friends, as both confidantes about inherited cancer risk information and as sources of support for coping with this information was also noted in the study.
A study of 31 mothers with a documented BRCA pathogenic variant explored patterns of dissemination to children.
Of those who chose to disclose test results to their children, age of offspring was the most important factor. Fifty percent of the children who were told were aged 20 to 29 years and slightly more than 25% of the children were aged 19 years or younger. Sons and daughters were notified in equal numbers. More than 70% of mothers informed their children within a week of learning their test result. Ninety-three percent of mothers who chose not to share their results with their children indicated that it was because their children were too young. These findings were consistent with three other studies showing that children younger than 13 years were less likely to be informed about test results compared with older children.
Another study of 187 mothers undergoing BRCA1/BRCA2 testing evaluated their need for resources to prepare for a facilitated conversation about sharing their BRCA1/BRCA2 testing results with their children. Seventy-eight percent of mothers were interested in three or more resources, including literature (93%), family counseling (86%), talk to prior participants (79%), and support groups (54%).
A longitudinal study of 153 women self-referred for genetic testing for BRCA1 and BRCA2 pathogenic variants and 118 of their partners evaluated communication about genetic testing and distress before testing and at 6 months posttesting.
The study found that most couples discussed the decision to undergo testing (98%), most test participants felt their partners were supportive, and most women disclosed test results to their partners (97%, n = 148). Test participants who felt their partners were supportive during pretest discussions experienced less distress after disclosure, and partners who felt more comfortable sharing concerns with test participants pretest experienced less distress after disclosure. Six-month follow-up revealed that 22% of participants felt the need to talk about the testing experience with their partners in the week before the interview. Most participants (72%, n = 107) reported comfort in sharing concerns with their partners, and 5% (n = 7) reported relationship strain as a result of genetic testing. In couples in which the woman had a positive genetic test result, more relationship strain, more protective buffering of their partners, and more discussion of related concerns were reported than in couples in which the woman had a true-negative or uninformative result.
A study of 561 FDRs of women who had undergone BRCA1/BRCA2 genetic testing found that 22% of FDRs did not recall being informed of the genetic test results despite the women reporting that the results had been shared.
Men were less likely to recall receiving the results (P > .001). Of those with recall about receiving the test results, 10.5% of FDRs did not recall the findings. For those with recall of the results, 17.9% of FDRs had an interpretation that was discordant with the correct results. Accuracy of test results recall was greater for informative test results (those that were either true positive or true negative) (P = .029). However, regardless of the test results, FDRs perceived the cancer risk to be higher before they learned of the findings than after (74% and 53% of FDRs reported that they believed their risk for cancer was greater than average before and after hearing test results, respectively).
There is a small but growing body of literature regarding psychological effects in men who have a family history of breast cancer and who are considering or have had BRCA testing. A qualitative study of 22 men from 16 high-risk families in Ireland revealed that more men in the study with daughters were tested than men without daughters. These men reported little communication with relatives about the illness, with some men reporting being excluded from discussion about cancer among female family members. Some men in the study also reported actively avoiding open discussion with daughters and other relatives.
In contrast, a study of 59 men testing positive for a BRCA1/BRCA2 pathogenic variant found that most men participated in family discussions about breast and/or ovarian cancer. However, fewer than half of the men participated in family discussions about risk-reducing surgery. The main reason given for having BRCA testing was concern for their children and a need for certainty about whether they could have transmitted the pathogenic variant to their children. In this study, 79% of participating men had at least one daughter. Most of these men described how their relationships had been strengthened after receipt of BRCA results, helping communication in the family and greater understanding.
Men in both studies expressed fears of developing cancer themselves. Irish men especially reported fear of cancer in sexual organs.
One study assessed 212 individuals from 13 hereditary breast and ovarian cancer families who received genetic counseling and were offered BRCA1/BRCA2 testing for documented pathogenic variants in the family. Individuals who were not tested were found 6 to 9 months later to have significantly greater increases in family expressiveness and cohesiveness compared with those who were tested. Persons who were randomly assigned to a client-centered versus problem-solving genetic counseling intervention had a significantly greater reduction in conflict, regardless of the test decision.
Many studies have looked at the psychological effects in women of having a high risk of developing cancer, either on the basis of carrying a BRCA1/BRCA2 pathogenic variant or having a strong family history of cancer. Some studies have also examined the effects on the partners of such women.
A Canadian study assessed 59 spouses of women found to have a BRCA1/BRCA2 pathogenic variant. All were supportive of their spouses’ decision to undergo genetic testing and 17% wished they had been more involved in the genetic testing process. Spouses who reported that genetic testing had no impact on their relationship had long-term relationships (mean duration 27 years). Forty-six percent of spouses reported that their major concern was of their partner dying of cancer. Nineteen percent were concerned their spouse would develop cancer and 14% were concerned their children would also be carriers of BRCA1/BRCA2 pathogenic variants.
In a U.S. study, 118 partners of women who underwent genetic testing for pathogenic variants in BRCA1 and BRCA2 completed a survey before testing and then again 6 months after result disclosure. At 6 months, only 10 partners reported that they had not been told of the test result. Ninety-one percent reported that the testing had not caused strain on their relationship. Partners who were comfortable sharing concerns before testing experienced less distress after testing. Protective buffering was not found to impact distress levels of partners.
An Australian study of 95 unaffected women at high risk of developing breast and/or ovarian cancer (13 carriers of pathogenic variants and 82 with unknown variant status) and their partners showed that although the majority of male partners had distress levels comparable to a normative population sample, 10% had significant levels of distress that indicated the need for further clinical intervention. Men with a high monitoring coping style and greater perceived breast cancer risk for their wives reported higher levels of distress. Open communication between the men and their partners and the occurrence of a cancer-related event in the wife’s family in the last year were associated with lower distress levels. When men were asked what kind of information and support they would like for themselves and their partners, 57.9% reported that they would like more information about breast and ovarian cancer, and 32.6% said they would like more support in dealing with their partner's risk. Twenty-five percent of men had suggestions on how to improve services for partners of high-risk women, including strategies on how to best support their partner, greater encouragement from health care professionals to attend appointments, and meeting with other partners.
A review of this literature reported that the BRCA testing process may be distressing for male partners, particularly for those with spouses identified as carriers. Male partner distress appears to be associated with their beliefs about the woman’s breast cancer risk, lack of couple communication, and feelings of alienation from the testing process.
A review of the literature on the experiences of males in families with a known BRCA1 and BRCA2 pathogenic variant reported that while the data are limited, men from variant-positive families are less likely than females to participate in communication regarding genetics at every level, including the counseling and testing process. Men are less likely to be informed of genetic test results received by female relatives, and most men from these families do not pursue their own genetic testing.
A study of Dutch men at increased risk of having inherited a BRCA1 pathogenic variant reported a tendency for the men to deny or minimize the emotional effects of their risk status, and to focus on medical implications for their female relatives. Men in these families, however, also reported considerable distress in relation to their female relatives.
In another study of male psychological functioning during breast cancer testing, 28 men belonging to 18 different high-risk families (with a 25% or 50% risk of having inherited a BRCA1/BRCA2 pathogenic variant) participated. The study purpose was to analyze distress in males at risk of carrying a BRCA1/BRCA2 pathogenic variant who applied for genetic testing. Of the men studied, most had low pretest distress; scores were lowest for men who were optimistic or who did not have daughters. Most carriers of pathogenic variants had normal levels of anxiety and depression and reported no guilt, though some anticipated increased distress and feelings of responsibility if their daughters developed breast or ovarian cancer. None of the noncarriers reported feeling guilty.
In one study,
adherence to recommended screening guidelines after testing was analyzed. In this study, more than half of male carriers of pathogenic variants did not adhere to the screening guidelines recommended after disclosure of genetic test results. These findings are consistent with those for female carriers of BRCA1/BRCA2 pathogenic variants.
A multicenter U.K. cohort study examined prospective outcomes of BRCA1/BRCA2 testing in 193 individuals, of which 20% were men aged 28 to 86 years. Men’s distress levels were low, did not differ among carriers and noncarriers, and did not change from baseline (before genetic testing) to the 3-year follow-up. Twenty-two percent of male carriers of pathogenic variants received colorectal cancer screening and 44% received prostate cancer screening;
however, it is unclear whether men in this study were following age-appropriate screening guidelines.
Several studies have explored communication of BRCA test results to at-risk children. Across all studies, the rate of disclosure to children ranging in age from 4 to 25 years is approximately 50%.
In general, age of offspring was the most important factor in deciding whether to disclose test results. In one study of 31 mothers disclosing their BRCA test results, 50% of the children who were informed of the results were aged 20 to 29 years and slightly more than 25% of the children were aged 19 years or younger. Sons and daughters were notified in equal numbers.
Similarly, in another study of 42 female carriers of BRCA pathogenic variants, 83% of offspring older than age 18 years were told of the results, while only 21% of offspring aged 13 years or younger were told.
Several studies have also looked at the timing of disclosure to children after parents receive their test results. Although the majority of children were told within a week to several months after results disclosure, some parents chose to delay disclosure.
Reasons for delaying disclosure included waiting for the child to get older, allowing time for the parent to adjust to the information, and waiting until results could be shared in person (in the case of adult children living away from home).
In one study, participants who told children younger than 13 years about their carrier status had increased distress, and those who did not tell their young children experienced a slight decrease in distress. Communication with young children was found to be influenced by developmental variables such as age and style of parent/child communication.
One study looked at the reaction of children to results disclosure or the effect on the parent-child relationship of communicating the results.
With regard to offspring’s understanding of the information, almost half of parents from one study reported that their child did not appear to understand the significance of a positive test result, although older children were reported to have a better understanding. This same study also showed that 48% of parents reported at least one negative reaction in their child, ranging from anxiety or concern (22%) to crying and fear (26%). It should be noted, however, that in this study children's level of understanding and reactions to the test result were measured qualitatively and based only on the parents' perception. Also, given the retrospective design of the study, there was a potential for recall bias. There were no significant differences in emotional reaction depending on age or gender of the child. Lastly, 65% of parents reported no change in their relationship with their child, while 5 parents (22%) reported a strengthening of their relationship.
Interestingly, a large multicenter study of 869 mother-daughter pairs (the daughters were aged 6 to 13 y) found that girls with a family history of breast cancer or a familial BRCA1/BRCA2 pathogenic variant (BCFH+) compared with those without such family histories had better psychosocial adjustment by maternal report.
However, based on a combination of maternal report and direct assessment of girls aged 10 to 13 years, BCFH+ girls experienced greater breast cancer–specific distress and a higher perceived risk of breast cancer than their peers without such family histories. Moreover, higher daughter distress was associated with higher maternal distress. A similarly designed study in older girls, aged 11 to 19 years, found that higher breast cancer–specific distress in daughters was associated with perceived risk and maternal distress. This older age group had higher self-esteem than did their peers without a family history of breast cancer.
Another study of 187 mothers undergoing BRCA1/BRCA2 testing evaluated their need for resources to prepare for a facilitated conversation about sharing their BRCA1/BRCA2 testing results with their children. Seventy-eight percent of mothers were interested in three or more resources, including literature (93%), family counseling (86%), talking to prior participants (79%), and support groups (54%).
Testing for BRCA1/BRCA2 has been almost universally limited to adults older than 18 years. The risks of testing children for adult-onset disorders (such as breast and ovarian cancer), as inferred from developmental data on children’s medical understanding and ability to provide informed consent, have been outlined in several reports.
Surveys of parental interest in testing children for adult-onset hereditary cancers suggest that parents are more eager to test their children than to be tested themselves for a breast cancer gene, suggesting potential conflicts for providers.
In a general population survey in the United States, 71% of parents said that it was moderately, very, or extremely likely that if they carried a breast-cancer predisposing pathogenic variant, they would test a 13-year-old daughter now to determine her breast cancer gene status.
To date, no data exist on the testing of children for BRCA1/BRCA2, though some researchers believe it is necessary to test the validity of assumptions underlying the general prohibition of testing of children for breast/ovarian cancer and other adult-onset disease genes.
In one study, 20 children (aged 11–17 y) of a selected group of mothers undergoing genetic testing (80% of whom previously had breast cancer and all of whom had discussed BRCA1/BRCA2 testing with their children) completed self-report questionnaires on their health beliefs and attitudes toward cancer, feelings related to cancer, and behavioral problems.
Ninety percent of children thought they would want cancer risk information as adults; half worried about themselves or a family member developing cancer. There was no evidence of emotional distress or behavioral problems. Another study by this group
found that 1 month after disclosure of BRCA1/BRCA2 genetic test results, 53% of 42 enrolled mothers of children aged 8 to 17 years had discussed their result with one or more of their children. Age of the child rather than pathogenic variant status of the mother influenced whether they were told, as did family health communication style.
The possibility of transmitting a pathogenic variant to a child may pose a concern to families affected by hereditary breast and ovarian cancer (HBOC),
perhaps to the extent that some carriers may avoid childbearing.
These concerns also may prompt women to consider using prenatal testing methods to help reduce the risk of transmission.
Prenatal diagnosis is an encompassing term used to refer to any medical procedure conducted to assess the presence of a genetic disorder in a fetus. Methods include amniocentesis and chorionic villous sampling (CVS).
Both procedures carry some risk of miscarriage and some evidence suggests fetal defects may result from using these tests.
Moreover, discovering the fetus is a carrier for a genetic defect may impose a difficult decision for couples regarding pregnancy continuation or termination. An alternative to these tests is preimplantation genetic testing (PGT), a procedure used to test fertilized embryos for genetic disorders before uterine implantation,
thereby avoiding the potential dangers associated with amniocentesis and CVS and the decision to terminate a pregnancy. Using the information obtained from the genetic testing, potential parents can decide whether or not to implant. PGT can be used to detect pathogenic variants in hereditary cancer predisposing genes, including BRCA.
In the United States, a series of studies has evaluated awareness, interest (e.g., would consider using PGT), and attitudes related to PGT among members of Facing Our Risk of Cancer Empowered (FORCE), an advocacy organization focused on persons at increased risk of HBOC.
The first study was a Web-based survey of 283 members,
the second included 205 attendees of the 2007 annual FORCE conference,
and the third was a Web-based survey of 962 members.
These studies have documented low levels of awareness, with 20% to 32% of study respondents reporting having heard of PGT before study participation.
With respect to interest in PGT, the first study
found only 13% of women would be likely to use PGT, whereas, 33% of respondents in the subsequent FORCE studies reported that they would consider using PGT.
In the third FORCE-based study (n = 962),
multivariable analysis revealed PGT interest was associated with the desire to have more children, having previously had any prenatal genetic test, and previous awareness of PGT. Attitudinal predictors of interest in PGT included agreement that others at risk of HBOC should be offered PGT; the belief that PGT is acceptable for persons at risk of HBOC; the belief that PGT information should be given to individuals at risk of HBOC; and endorsement of PGT benefits of having children without genetic variants and eliminating genetic diseases. Conversely, those who indicated that PGT was “too much like playing God” and reported that they considered PGT in the context of religion, had less interest in PGT.
It is unknown whether the attitudes of FORCE members toward PGT are representative of the majority of BRCA carriers. A cross-sectional study of 1,081 BRCA carriers, 65% of whom were recruited through FORCE and the remainder by the University of Pennsylvania, revealed that a majority of carriers were in favor of offering PGT and prenatal diagnosis to carriers (59% for PGT and 55.5% for prenatal diagnosis).
Of those who indicated that their families were not complete, 41% of BRCA carriers reported that their carrier status impacted their decision about future biological children. This study also revealed that 21.5% of unpartnered BRCA carriers felt more pressure to get married.
The U.K. Human Fertilization and Embryology authority has approved the use of PGT for hereditary breast and ovarian cancer. In a sample of 102 women with a BRCA pathogenic variant, most were supportive of PGT but only 38% of the women who had completed their families would consider it for themselves had PGT been available, and only 14% of women who were contemplating a future pregnancy would consider PGT.
In a study of 77 individuals undergoing BRCA testing as part of a multicenter cohort study in Spain, 61% of respondents reported they would consider PGT. Factors associated with PGT interest were age 40 years and older and had a prior cancer diagnosis.
In France, couples who obtain authorization from a multidisciplinary prenatal diagnosis team may access PGT free of charge as a benefit of their national health care system. However, no BRCA carriers have been authorized to use PGT. In a national study of 490 unaffected carriers of BRCA pathogenic variants of childbearing age (women aged 18–49 y; men aged 18–69 y), 16% stated that BRCA test results had altered their ongoing plans for childbearing.
Upon qualitative analysis of written comments provided by some respondents, the primary impact was related to accelerating the timing of pregnancy, feelings of guilt about possibly passing on the pathogenic variant to offspring, and having future children. In response to a hypothetical scenario in which PGT was readily available, 33% of participants reported that they would undergo PGT. Factors associated with this intention were having no future reproductive plans at the time of the survey, feeling pregnancy termination was an acceptable option in the context of identifying a BRCA pathogenic variant, and having fewer cases of breast and/or ovarian cancer in the family. When presented with questions about expectations about delivery of PGT or prenatal diagnosis (PND) information, 85% of respondents felt it should be provided along with BRCA test results; 45% felt that it should be provided when carriers decide to have children. Respondents stated that they would expect this information to be delivered by cancer geneticists (92%), obstetrician/gynecologists (76%), and general practitioners (48%).
A small (N = 25) qualitative study of women of reproductive age positive for a BRCA pathogenic variant who underwent genetic testing before having children evaluated how their BRCA status influenced their attitudes about reproductive genetic testing (both PGT and PND) and decisions about having children.
In this study, the decision to undergo BRCA testing was primarily motivated by the desire to manage one’s personal cancer risk, rather than a desire to inform future reproductive decisions. The perceived severity of HBOC influenced concerns about passing on a BRCA pathogenic variant to children and also influenced willingness to consider PGT or PND and varied based on personal experience. Most did not believe that a known BRCA pathogenic variant was a reason to terminate a pregnancy. As observed in prior studies, knowledge of reproductive options varied; however, there was a tendency among participants to view PGT as more acceptable than PND with regard to termination of pregnancy. Decisions regarding the pros and cons of PGT versus PND with termination of pregnancy were driven primarily by personal preferences and experiences, rather than by morality judgments. For example, women were deterred from PGT based on the need to undergo in vitro fertilization and to take hormones that might increase cancer risk and based on the observed experiences of others who underwent this procedure.
One study has examined these issues among high-risk men recruited from FORCE and Craigslist (a bulletin board website) (N = 228).
Similar to the previous studies of women, only 20% of men were aware of PGT before survey participation. In a multivariate analysis, those who selected the “other” option for possible benefits of PGT compared with those who selected from several predetermined options (e.g., having children without genetic variants) and those who considered PGT in the context of religion (as opposed to health and safety) were less likely to report that they would ever consider using PGT.
The recognition that BRCA1/BRCA2 pathogenic variants are prevalent, not only in breast/ovarian cancer families but also in some ethnic groups,
has led to considerable discussion of the ethical, psychological, and other implications of having one’s ethnicity be a factor in determination of disease predisposition. Concerns that people will think everything is solely determined by genetic factors and the creation of a genetic underclass
have been voiced. Questions about the impact on the group of being singled out as having genetic vulnerability to breast cancer have been raised. There is also confusion about who gives or withholds permission for the group to be involved in studies of their genetic identity. These issues challenge traditional views on informed consent as a function of individual autonomy.
A growing literature on the unique factors influencing a variety of cultural subgroups suggests the importance of developing culturally specific genetic counseling and educational approaches.
The inclusion of members within the community of interest (e.g., breast cancer survivors, advocates, and community leaders) may enhance the development of culturally tailored genetic counseling materials.
One study showed that participation in any genetic counseling (culturally mediated or standard approaches) reduced perceived risk of developing breast cancer.
The human implications of the ethical issues raised by the advent of genetic testing for breast/ovarian cancer susceptibility are described in case studies,essays, and research reports. Issues about rights and responsibilities in families concerning the spread of information about genetic risk promise to be major ethical and legal dilemmas in the coming decades.
Studies have shown that 62% of studied family members were aware of the family history and that 88% of hereditary breast/ovarian cancer family members surveyed have significant concerns about privacy and confidentiality. Expressed concern about cancer in third-degree relatives, or relatives farther removed, was about the same as that for first- or second-degree relatives of the proband.
Only half of surveyed FDRs of women with breast or ovarian cancer felt that written permission should be required to disclose BRCA1/BRCA2 test results to a spouse or immediate family member. Attitudes toward testing varied by ethnicity, previous exposure to genetic information, age, optimism, and information style. Altruism is a factor motivating genetic testing in some people.
Many professional groups have made recommendations regarding informed consent.
There is some evidence that not all practitioners are aware of or follow these guidelines.
Research shows that many BRCA1/BRCA2 genetic testing consent forms do not fulfill recommendations by professional groups about the 11 areas that should be addressed, and they omit highly relevant points of information.
In a study of women with a history of breast or ovarian cancer, the interviews yielded that the women reported feeling inadequately prepared for the ethical dilemmas they encountered when imparting genetic information to family members.
These data suggest that more preparation about disclosure to family members before testing reduces the emotional burden of disseminating genetic information to family members. Patients and health care providers would benefit from enhanced consideration of the ethical issues of warning family members about hereditary cancer risk. (Refer to the PDQ summaries Cancer Genetics Risk Assessment and Counseling and Cancer Genetics Overview for more information about the ethics of cancer genetics and genetic testing.)
There is a small but growing body of literature on the use of decision aids as an adjunct to standard genetic counseling to assist patients in making informed decisions about cancer risk management.
One study showed that the use of a decision aid consisting of individualized value assessment and cancer risk management information after receiving positive BRCA1/BRCA2 test results was associated with fewer intrusive thoughts and lower levels of depression at the 6-month follow-up in unaffected women. Use of the decision aid did not alter cancer risk management intentions and behaviors. Slightly detrimental effects on well-being and several decision-related outcomes, however, were noted among affected women.
Another study compared responses to a tailored decision aid (including a values-clarification exercise) versus a general information pamphlet intended for women making decisions about ovarian cancer risk management. In the short term, the women receiving the tailored decision aid showed a decrease in decisional conflict and increased knowledge compared with women receiving the pamphlet, but no differences in decisional outcomes were found between the two groups. In addition, the decision aid did not appear to alter the participant’s baseline cancer risk management decisions.
A multisite randomized trial of 150 unaffected women with BRCA1/BRCA2 pathogenic variants assessed the effect of a decision aid on breast cancer risk management decisions and psychosocial outcomes. At 6-month and 12-month follow-up, women randomly assigned to the decision aid had lower levels of cancer-related distress (P = .01 at 6 months and P = .05 at 12 months) than did the control group.
Decisional conflict scores were relatively low at baseline and declined over time in both groups; the scores between the two groups were not statistically different.
An increasing number of studies have examined uptake and adherence to cancer risk management options among individuals who have undergone genetic counseling and testing for BRCA1 and BRCA2 pathogenic variants. Findings from these studies are reported in Table 15 and Table 16. Outcomes vary across studies and include uptake or adherence to screening (mammography, magnetic resonance imaging [MRI], cancer antigen [CA] 125, transvaginal ultrasound [TVUS]) and selection of RRM and RRSO. Studies generally report outcomes by pathogenic variant carrier or testing status (e.g., positive for pathogenic variants, negative for pathogenic variants, or declined genetic testing). Follow-up time after notification of genetic risk status also varied across studies, ranging from 12 months up to several years.
Findings from these studies suggest that breast screening often improves after notification of BRCA1/BRCA2 pathogenic variant carrier status; nonetheless, screening remains suboptimal. Fewer studies have examined adoption of MRI as a screening modality, probably due to the recent availability of efficacy data. Screening for ovarian cancer varied widely across studies, and also varied based on type of screening test (i.e., CA-125 serum testing vs. TVUS screening). However, ovarian cancer screening does not appear to be widely adopted by carriers of BRCA1/BRCA2 pathogenic variants. Uptake of RRM varied widely across studies, and may be influenced by personal factors (such as younger age or having a family history of breast cancer), psychosocial factors (such as a desire for reduction of cancer-related distress), recommendations of the health care provider, and cultural or health care system factors. An individual’s choice to have a bilateral mastectomy also appears to be influenced by pretreatment genetic education and counseling regardless of the genetic test results.
Similarly, uptake of RRSO also varied across studies, and may be influenced by similar factors, including older age, personal history of breast cancer, perceived risk of ovarian cancer, cultural factors (i.e., country), and the recommendations of the health care provider.
Study Citation | Study Population | Uptake of RRM | Uptake of Breast Screening Mammography and/or Breast MRI | Length of Follow-up | Comments |
---|---|---|---|---|---|
Study Citation | Study Population | Uptake of RRSO | Uptake of Gynecologic Screening | Length of Follow-up | Comments |
United States | |||||
Botkin et al. (2003) | Carriers (n = 37)a | Carriers 0% | Mammography | 24 mo | |
– Carriers 57% | |||||
Noncarriers (n = 92)a | Noncarriers 0% | – Noncarriers 49% | |||
– Declined test 20% | |||||
Declined testing (n = 15)a | MRI | ||||
– Not evaluated | |||||
Beattie et al. (2009) | Carriers (n = 237) | Carriers 23% | Not applicable | Mean, 3.7 y | Women opting for RRM were younger than 60 y, had a prior diagnosis of breast cancer, and also underwent RRSO. |
Median time to RRM: 124 days from receiving results. | |||||
O’Neill et al. (2010) | Carriers (n = 146) | Carriers 13% | Not applicable | 12 mo | Intentions at test result disclosure predicted RRM decisions. |
Schwartz et al. (2012) | Carriers (n = 108) | Carriers 37% | Mammography | Mean, 5.3 y | Predictors of RRM were younger age, higher precounseling cancer distress, more recent diagnosis of breast or ovarian cancer, and intact ovaries. |
– Carriers affected 92% | |||||
– Carriers unaffected 82% | |||||
Noncarriers (n = 60)a | Noncarriers 0% | – Noncarriers 66% | |||
– Uninformative affected 89% | |||||
MRI | |||||
Uninformative (n = 206)a | Uninformative 6.8% | – Carriers affected 51% | |||
– Carriers unaffected 46% | |||||
– Noncarriers 11% | |||||
– Uninformative 27% | |||||
Garcia et al. (2013) | Carriers (n = 250) | Carriers 44% | Excluding women post RRM: | 41 months; range, 26–66 mo | Breast surveillance decreased significantly from y 1–5 of follow-up: Mammography 43% to 7%; MRI 35% to 3%. |
Mammography: | |||||
– Carriers 43% | |||||
MRI: | |||||
– Carriers 35% | |||||
Singh et al. (2013) | Carriers (n = 136)b | Carriers 42% | Not applicable | Range, 1–11 y | Predictors of RRM were first- or second-degree relative diseased from breast cancer, having had at least one childbirth, and having undergone testing after 2005. |
International | |||||
Phillips et al. (2006) | Carriers (n = 70) | Carriers 11% | Mammography | 3 y | |
– Carriers 89% | |||||
MRI | |||||
– Not evaluated | |||||
Metcalfe et al. (2008) | Carriers (N = 2,677) | Carriers 18% (unaffected) | Mammography | 3.9 y; range, 1.5–10.3 y | Large differences in uptake of risk management options by country. |
– Carriers 87% | |||||
MRI | 1,294 participants had a personal history of breast cancer. | ||||
– Carriers 31% | |||||
Julian-Reynier et al. (2011) | Carriers (n = 101) | Carriers 6.9% | Mammography | 5 y | Noncarriers often continued screening. |
– Carriers 59% | |||||
– Noncarriers aged 30–39 y 53% | |||||
Noncarriers (n = 145) | Noncarriers 0% | MRI | |||
– Carriers 31% | |||||
– Noncarriers 4.8% | |||||
Study Citation | Study Population | Uptake of RRSO | Uptake of Gynecologic Screening | Length of Follow-up | Comments |
United States | |||||
Scheuer et al. (2002) | Carriers (n = 179) | Carriers 50.3% | CA-125 | Mean, 24.8 mo; range, 1.6–66.0 mo | Women undergoing RRSO were older and more likely to have a personal history of breast cancer. |
– Carriers 67.6% | |||||
TVUS | |||||
– Carriers 72.9% | |||||
Beattie et al. (2009) | Carriers (n = 240) | Carriers 51% | Not applicable | Mean, 3.7 y | Women opting for RRSO <60 y had a prior diagnosis of breast cancer and also underwent RRM. |
Median time to RRSO: 123 days from receiving results. | |||||
O'Neill et al. (2010) | Carriers (n = 146) | Carriers 32% | Not applicable | 12 mo | |
Schwartz et al. (2012) | Carriers (n = 100) | Carriers 65% | CA-125 | Mean, 5.3 y | Predictors of RRSO were being ≥40 y and having received a diagnosis of breast cancer more than 10 y ago. |
Noncarriers (n = 52) | Noncarriers 1.9% | – Carriers 56% | |||
– Noncarriers 12% | |||||
– Uninformative 33% | |||||
Uninformative (n = 203) | Uninformative 13.3% | TVUS | |||
– Carriers 42% | |||||
– Noncarriers 20% | |||||
– Uninformative 26% | |||||
Garcia et al. (2013) | Carriers (n = 305) | Carriers 74% | Excluding women post-RRSO: | 41 mo; range, 26–66 mo | Ovarian surveillance decreased significantly from years 1–5 of follow-up; CA-125: 47% to 2%; TVUS: 45% to 2.3% |
CA-125 | |||||
– Carriers 47% | |||||
TVUS | |||||
– Carriers 45% | |||||
Mannis et al. (2013) | Carriers (n = 201)a | Carriers 69.6% | CA-125 | Median, 3.7 y | Predictors of RRSO and screening included being a carrier of a BRCA pathogenic variant, age 40–49 y, having a higher income, ≥2 children, a personal history of breast cancer, and a first-degree relative with ovarian cancer. |
– 26.3% | |||||
TVUS | |||||
– 26.3% | |||||
Noncarriers (n = 103) | Noncarriers 2.0% | Not reported | |||
Uninformative (n = 773)a; 59/773 with a variant of uncertain significance | Uninformative 12.3% | CA-125 | |||
– 10.4% | |||||
TVUS | |||||
– 6.5% | |||||
Singh et al. (2013) | Carriers (n = 136)b | Carriers 52% | Not applicable | Range, 1–11 y | Predictors of RRSO were first- or second-degree relative with breast cancer, a mother lost to pelvic cancer, having had ≥1 childbirths, age ≥50 y, and having undergone testing after 2005. |
International | |||||
Phillips et al. (2006) | Carriers (n = 70) | Carriers 29% | CA-125 | 3 y | |
– Carriers 0% | |||||
TVUS | |||||
– Carriers 67% | |||||
Friebel et al. (2007) | Carriers (N = 537) | Carriers 55% | Not applicable | Minimum 6 mo; median 36 mo | RRSO greatest in parous women >40 y. |
Madalinska et al. (2007) | Carriers (n = 160) | Carriers 74% | Carriers 26% | 12 mo | Women who underwent RRSO had lower education levels, viewed ovarian cancer as incurable, and believed strongly in the benefits of RRSO. |
Specific method(s) of gynecological screening not reported. | |||||
Metcalfe et al. (2008) | Carriers (N = 2,677) | Carriers 57% | Not applicable | 3.9 y; range, 1.5–10.3 y | Large differences in uptake of risk management options by country. |
Julian-Reynier et al. (2011) | Carriers (n = 101)a | Carriers 42.6% | TVUS | 5 y | RRSO uptake increased with age. Having undergone RRSO did not alter breast cancer risk perception. Noncarriers often continued screening. |
Noncarriers (n = 145)a | Noncarriers 2% | – Noncarriers 43.2% | |||
Rhiem et al. (2011) | Carriers (N = 306) | Carriers 57% | Not evaluated | Mean, 47.8 mo post-oophorectomy | Median age at time of RRSO = 47 y. One occult fallopian tube cancer was detected at the time of RRSO. One peritoneal carcinoma was diagnosed 26 mo post-RRSO. |
Sidon et al. (2012) | Carriers (N = 700)a; 386/700 with personal history of breast cancer | BRCA1 carriers: | Not evaluated | Affected with breast cancer | Uptake of RRSO was lower in women >60 y (22% uptake at 5 y). None of the women >70 y had a RRSO performed. |
– 54.5% | |||||
BRCA2 carriers: | – BRCA1: Mean, 2.29; range, 0.1–11.45 y | ||||
– 45.5% | |||||
All carriers with no personal history of breast cancer | – BRCA2: Mean, 1.77; range, 0.1–11.1 y | ||||
Not affected with breast cancer | |||||
– 54.2% | |||||
All carriers with personal history of breast cancer | – BRCA1: Mean, 1.63; range, 0.1–11.28 y | ||||
– 43.2% | – BRCA2: Mean, 1.75; range, 0.1–8.98 y | ||||
MRI = magnetic resonance imaging; RRSO = risk-reducing salpingo-oophorectomy. | |||||
aSelf-report as data source. | |||||
bMedical records as data source. | |||||
CA-125 = cancer antigen 125; RRM = risk-reducing mastectomy; TVUS = transvaginal ultrasound. | |||||
aSelf-report as data source. | |||||
bMedical records as data source. | |||||
cData source not specified. |
Study Citation | Study Population | Uptake of RRSO | Uptake of Gynecologic Screening | Length of Follow-up | Comments |
---|---|---|---|---|---|
United States | |||||
Scheuer et al. (2002) | Carriers (n = 179) | Carriers 50.3% | CA-125 | Mean, 24.8 mo; range, 1.6–66.0 mo | Women undergoing RRSO were older and more likely to have a personal history of breast cancer. |
– Carriers 67.6% | |||||
TVUS | |||||
– Carriers 72.9% | |||||
Beattie et al. (2009) | Carriers (n = 240) | Carriers 51% | Not applicable | Mean, 3.7 y | Women opting for RRSO <60 y had a prior diagnosis of breast cancer and also underwent RRM. |
Median time to RRSO: 123 days from receiving results. | |||||
O'Neill et al. (2010) | Carriers (n = 146) | Carriers 32% | Not applicable | 12 mo | |
Schwartz et al. (2012) | Carriers (n = 100) | Carriers 65% | CA-125 | Mean, 5.3 y | Predictors of RRSO were being ≥40 y and having received a diagnosis of breast cancer more than 10 y ago. |
Noncarriers (n = 52) | Noncarriers 1.9% | – Carriers 56% | |||
– Noncarriers 12% | |||||
– Uninformative 33% | |||||
Uninformative (n = 203) | Uninformative 13.3% | TVUS | |||
– Carriers 42% | |||||
– Noncarriers 20% | |||||
– Uninformative 26% | |||||
Garcia et al. (2013) | Carriers (n = 305) | Carriers 74% | Excluding women post-RRSO: | 41 mo; range, 26–66 mo | Ovarian surveillance decreased significantly from years 1–5 of follow-up; CA-125: 47% to 2%; TVUS: 45% to 2.3% |
CA-125 | |||||
– Carriers 47% | |||||
TVUS | |||||
– Carriers 45% | |||||
Mannis et al. (2013) | Carriers (n = 201)a | Carriers 69.6% | CA-125 | Median, 3.7 y | Predictors of RRSO and screening included being a carrier of a BRCA pathogenic variant, age 40–49 y, having a higher income, ≥2 children, a personal history of breast cancer, and a first-degree relative with ovarian cancer. |
– 26.3% | |||||
TVUS | |||||
– 26.3% | |||||
Noncarriers (n = 103) | Noncarriers 2.0% | Not reported | |||
Uninformative (n = 773)a; 59/773 with a variant of uncertain significance | Uninformative 12.3% | CA-125 | |||
– 10.4% | |||||
TVUS | |||||
– 6.5% | |||||
Singh et al. (2013) | Carriers (n = 136)b | Carriers 52% | Not applicable | Range, 1–11 y | Predictors of RRSO were first- or second-degree relative with breast cancer, a mother lost to pelvic cancer, having had ≥1 childbirths, age ≥50 y, and having undergone testing after 2005. |
International | |||||
Phillips et al. (2006) | Carriers (n = 70) | Carriers 29% | CA-125 | 3 y | |
– Carriers 0% | |||||
TVUS | |||||
– Carriers 67% | |||||
Friebel et al. (2007) | Carriers (N = 537) | Carriers 55% | Not applicable | Minimum 6 mo; median 36 mo | RRSO greatest in parous women >40 y. |
Madalinska et al. (2007) | Carriers (n = 160) | Carriers 74% | Carriers 26% | 12 mo | Women who underwent RRSO had lower education levels, viewed ovarian cancer as incurable, and believed strongly in the benefits of RRSO. |
Specific method(s) of gynecological screening not reported. | |||||
Metcalfe et al. (2008) | Carriers (N = 2,677) | Carriers 57% | Not applicable | 3.9 y; range, 1.5–10.3 y | Large differences in uptake of risk management options by country. |
Julian-Reynier et al. (2011) | Carriers (n = 101)a | Carriers 42.6% | TVUS | 5 y | RRSO uptake increased with age. Having undergone RRSO did not alter breast cancer risk perception. Noncarriers often continued screening. |
Noncarriers (n = 145)a | Noncarriers 2% | – Noncarriers 43.2% | |||
Rhiem et al. (2011) | Carriers (N = 306) | Carriers 57% | Not evaluated | Mean, 47.8 mo post-oophorectomy | Median age at time of RRSO = 47 y. One occult fallopian tube cancer was detected at the time of RRSO. One peritoneal carcinoma was diagnosed 26 mo post-RRSO. |
Sidon et al. (2012) | Carriers (N = 700)a; 386/700 with personal history of breast cancer | BRCA1 carriers: | Not evaluated | Affected with breast cancer | Uptake of RRSO was lower in women >60 y (22% uptake at 5 y). None of the women >70 y had a RRSO performed. |
– 54.5% | |||||
BRCA2 carriers: | – BRCA1: Mean, 2.29; range, 0.1–11.45 y | ||||
– 45.5% | |||||
All carriers with no personal history of breast cancer | – BRCA2: Mean, 1.77; range, 0.1–11.1 y | ||||
Not affected with breast cancer | |||||
– 54.2% | |||||
All carriers with personal history of breast cancer | – BRCA1: Mean, 1.63; range, 0.1–11.28 y | ||||
– 43.2% | – BRCA2: Mean, 1.75; range, 0.1–8.98 y | ||||
CA-125 = cancer antigen 125; RRM = risk-reducing mastectomy; TVUS = transvaginal ultrasound. | |||||
aSelf-report as data source. | |||||
bMedical records as data source. | |||||
cData source not specified. |
On the other hand, many women found to be pathogenic variant carriers express interest in RRM in hopes of minimizing their risk of breast cancer. In one study of a number of unaffected women with no previous risk-reducing surgery who received results of BRCA1 testing after genetic counseling, 17% of carriers (2 of 12) intended to have mastectomies and 33% (4 of 12) intended to have oophorectomies.
In a later study of the same population, RRM was considered an important option by 35% of women who tested positive, whereas risk-reducing oophorectomy was considered an important option by 76%. A prospective study assessed the stability of risk management preferences over five time points (pre-BRCA testing to 9 months after results disclosure) among 80 Dutch women with a documented BRCA pathogenic variant. Forty-six participants indicated a preference for screening at baseline. Of 25 women who preferred RRM at baseline, 22 indicated the same preference 9 months after test results disclosure; however, it was not reported how many women actually had RRM.
Initial interest does not always translate into the decision for surgery. Two different studies found low rates of RRM among carriers of pathogenic variants in the year after result disclosure, one showing 3% (1 of 29) of carriers and the other 9% (3 of 34) of carriers having had this surgery.
Among members from a large BRCA1 kindred, utilization of cancer screening and/or risk-reducing surgeries was assessed at baseline (before disclosure of results), and at 1 year and 2 years after disclosure of BRCA1 test results. Of the 269 men and women who participated, complete data were obtained on 37 female carriers and 92 female noncarriers, all aged 25 years or older. At 2 years after disclosure of test results, none of the women had undergone RRM, although 4 of the 37 carriers (10.8%) said they were considering the procedure. In contrast, of the 26 women who had not had an oophorectomy before baseline, 46% (12 of 26) had obtained an oophorectomy by 2 years after testing. Of those carriers aged 25 to 39 years, 29% (5 of 17) underwent oophorectomy, while 78% (7 of 9) of the carriers aged 40 years and older had this procedure.
In a study assessing uptake of risk-reducing surgery 3 months after BRCA result disclosure, 7 of 62 women had undergone RRM and 13 of 62 women had undergone RRSO. Intent to undergo RRSO before testing correlated with procedure uptake. In contrast, intent to undergo RRM did not correlate with uptake. Overall, reasons given for indecision about risk-reducing surgery included complex testing factors such as the significance of family history in the absence of a pathogenic variant, concerns over the surgical procedure, and time and uncertainty regarding early menopause and the use of HRT.
In a U.K. study, data were collected during observations of genetic consultations and in semistructured interviews with 41 women after they received genetic counseling.
The option of risk-reducing surgery was raised in 29 consultations and discussed in 35 of the postclinic interviews. Fifteen women said they would consider having an oophorectomy in the future, and nine said they would consider having a mastectomy. The implications of undergoing oophorectomy and mastectomy were discussed in postclinic interviews. Risk-reducing surgery was described by the counselees as providing individuals with a means to (a) fulfill their obligations to other family members and (b) reduce risk and contain their fear of cancer. The costs of this form of risk management were described by the respondents as follows:
A number of women choose to undergo RRM and RRSO without genetic testing because of the following:
Among FDRs of breast cancer patients attending a surveillance clinic, women who expressed an interest in RRM and/or had undergone surgery were found to have significantly more breast cancer biopsies (P < .05) and higher subjective 10-year breast cancer risk estimates (P < .05) than women not interested in RRM. Cancer worry at the time of entry into the clinic was highest among women who subsequently underwent RRM compared with women who expressed interest but had not yet had surgery and women who did not intend to have surgery (P < .001).
BRCA testing, when offered to women newly diagnosed with breast cancer, has been shown to influence surgical decision making in that carriers are more likely to opt for bilateral mastectomy compared with noncarriers.
A study that evaluated predictors of contralateral RRM among 435 breast cancer survivors found that 16% had undergone contralateral RRM (in conjunction with mastectomy of the affected breast) before referral for genetic counseling and BRCA1/BRCA2 genetic testing.
Predictors of contralateral RRM before genetic counseling and testing included younger age at breast cancer diagnosis, more time since diagnosis, having at least one affected FDR, and not being employed full-time. In the year after disclosure of test results, 18% of women who tested positive for a BRCA1/BRCA2 pathogenic variant and 2% of those whose test results were uninformative underwent contralateral RRM. Predictors of contralateral RRM after genetic testing included younger age at breast cancer diagnosis, higher cancer-specific distress before genetic counseling, and having a positive BRCA1/BRCA2 test result. In this study, contralateral RRM was not associated with distress at 1 year after disclosure of genetic test results. A retrospective chart review evaluated uptake of bilateral mastectomies in 110 women who underwent BRCA1/BRCA2 genetic testing before making surgical decisions about the treatment of newly diagnosed breast cancer. Carriers of BRCA pathogenic variants were more likely to undergo bilateral mastectomies than were women in whom no variant was detected (83% vs. 37%; P = .046).
The only predictor of contralateral RRM in women without a pathogenic variant was being married (P = .03). Age, race, parity, disease stage and biomarkers, increased mammographic breast density, and breast MRI did not influence contralateral RRM decisions at the time of primary surgical treatment.
A study conducted from 2006 to 2014 in 11 U.S. academic and community centers of 897 women, aged 40 years and younger at breast cancer diagnosis, found that rates of BRCA genetic testing have increased over time.
Within 1 year after diagnosis, 87% of the sample underwent BRCA testing. The rate increased from 77% of newly diagnosed women tested in 2006 to 95% of women tested in 2013. Among women who tested positive for a pathogenic BRCA variant and stated that testing affected their surgery decisions (n = 88), 86% underwent bilateral mastectomy compared with 51% of noncarriers (P < .001). Among untested women, about one-third reported that they were not told by a health care provider that they were candidates for BRCA testing; yet, according to national guidelines, all were eligible for testing solely on the basis of their age at diagnosis.
Dutch women (N = 114) who had undergone unilateral or bilateral RRM with breast reconstruction between 1994 and 2002 were retrospectively surveyed to determine their satisfaction with the procedure.
Sixty-eight percent were either unaffected carriers of BRCA pathogenic variants or at a 50% risk of having a BRCA pathogenic variant in their family. Sixty percent of respondents indicated that they were satisfied with the procedure, 95% would opt for RRM again, and 80% would opt for the same reconstruction procedure. Less than half reported some perioperative or postoperative complications, ongoing physical complaints, or some physical limitations. Twenty-nine percent reported altered feelings of femininity after the procedure, 44% reported adverse changes in their sexual relationships, and 35% indicated that they believed their partners experienced adverse changes in their sexual relationship. Ten percent of women, however, reported positive changes in their sexual relationship after the procedure. Compared with patients who indicated satisfaction with this procedure, nonsatisfied patients were more likely to feel less informed about the procedure and its consequences, report more complications and physical complaints, feel that their breasts did not belong to their body, and indicate that they would not opt for reconstruction again. Those who reported a negative effect on their sexual relationship were more likely to:
Ninety Swedish women who had undergone RRM between 1997 and 2005 were surveyed before surgery, 6 months after surgery, and 1 year after surgery to evaluate changes in health-related quality of life, depression, anxiety, sexuality, and body image. There were no significant changes in health-related quality of life or depression at the three time points; anxiety decreased over time (P = .0004). More than 80% of women reported having an intimate relationship at all three time points. Women who reported being sexually active were asked to respond to questions about sexual pleasure, discomfort, habit, and frequency of activity. There were no statistically significant differences related to frequency, habit, or discomfort. However, pleasure significantly decreased between baseline and 1 year after surgery (P = .005). At 1 year after surgery, 48% of women reported feeling less attractive, 48% reported feeling self-conscious, and 44% reported dissatisfaction with surgical scars.
Discussion of risk-reducing surgical options may not consistently occur during pretest genetic counseling. In one multi-institutional study, only one-half of genetics specialists discussed RRM and RRSO in consultations with women from high-risk breast cancer families,
despite the fact that discussion of surgical options was significantly associated with meeting counselees’ expectations, and that such information was not associated with increased anxiety.
Given the increased risk of ovarian cancer faced by women with a BRCA1 or BRCA2 pathogenic variant, those who do receive information about RRSO show wide variations in surgery uptake (27%–72%).
A study showed that clinical factors related to choosing RRSO versus surveillance alone are older age, parity of one or more, and a prior breast cancer diagnosis.
In this study, the choice of RRSO was not related to family history of breast or ovarian cancer. Hysterectomy was presented as an option during genetic counseling, and 80% of women who underwent RRSO also elected to have a hysterectomy.
Data are now emerging regarding uptake and adherence to cancer risk management recommendations such as screening and risk-reducing interventions. Cancer screening adherence and risk-reduction behaviors as defined by the National Comprehensive Cancer Network Guidelines were assessed in a cross-sectional study of 214 women with a personal history (n = 134) or family history (n = 80) of breast or ovarian cancer. Among unaffected women older than 40 years, 10% had not had a mammogram or clinical breast examination (CBE) in the previous year and 46% did not practice breast self-examination (BSE). Among women previously affected with breast or ovarian cancer, 21% had not had a mammogram, 32% had not had a CBE, and 39% did not practice BSE.
Three hundred and twelve women who were counseled and tested for BRCA pathogenic variants between 1997 and 2005 responded to a survey regarding their perception of genetic testing for hereditary breast and ovarian cancer. The survey included questions on risk reduction options, including screening and risk-reducing surgeries. Two hundred and seventeen (70%) of the women had been diagnosed with breast cancer, and 86 (28%) tested positive for a pathogenic variant in either the BRCA1 or BRCA2 gene. None of the BRCA-positive women agreed that mammograms are difficult procedures because of the discomfort, while 11 (5.4%) of the BRCA-negative women did agree with this statement. Both groups (BRCA-positive and BRCA-negative) agreed that risk-reducing surgeries provide the best means for lowering cancer risk and worry, and most patients in both groups expressed the belief that risk-reducing mastectomy is not too drastic, too scary, or too disfiguring.
A prospective study from the United Kingdom examined the psychological impact of mammographic screening in 1,286 women aged 35 to 49 years who have a family history of breast cancer and were participants in a multicenter screening program. Mammographic abnormalities that required additional evaluation were detected in 112 women. These women, however, did not show a statistically significant increase in cancer worry or negative psychological consequences as a result of these findings. The 1,174 women who had no mammographic abnormality detected experienced a decrease in cancer worry and a decrease in negative psychological consequences compared with baseline after receipt of their results. At 6 months, the entire cohort had experienced a decrease in measures of cancer worry and psychological consequences of breast screening.
A qualitative study explored health care professionals’ views regarding the provision of information about health protective behaviors (e.g., exercise and diet). Seven medical specialists and ten genetic counselors were interviewed during a focus group or individually. The study reported wide variation in the content and extent of information provided about health-protective behaviors and in general, participants did not consider it their role to promote such behaviors in the context of a genetic counseling session. There was agreement, however, about the need to form consensus about provision of such information both within and across risk assessment clinics.
Not all studies specify whether screening uptake rates fall within recommended guidelines for the targeted population or the specific clinical scenario, nor do they report on other variables that may influence cancer screening recommendations. For example, women who have a history of atypical ductal hyperplasia would be advised to follow screening recommendations that may differ from those of the general population.
A prospective study conducted in the Netherlands found that among 26 carriers of BRCA1/BRCA2 pathogenic variants, the 14 women who chose mastectomy had higher distress both before test result disclosure and 6 and 12 months later, compared with the 12 carriers who chose surveillance and compared with 53 women negative for a pathogenic variant. Overall, however, anxiety declined in women undergoing risk-reducing mastectomy (RRM); at 1 year, their anxiety scores were closer to those of women choosing surveillance and to the scores of women negative for a pathogenic variant.
Interestingly, women opting for RRM had lower pretest satisfaction with their breasts and general body image than carriers who opted for surveillance or noncarriers of BRCA1/BRCA2 pathogenic variants. Of the women who had an RRM, all but one did not regret the decision at 1 year posttest disclosure, but many had difficulties with body image, sexual interest and functioning, and self-esteem. The perception that doctors had inadequately informed them about the consequences of RRM was associated with regret.
At the 5-year follow-up, women who had undergone RRM had less favorable body image and changes in sexual relationships, but also had a significant reduction in the fear of developing cancer.
In a study of 78 women who underwent risk-reducing surgery (including BRCA1/BRCA2 carriers and women who were from high-risk families with no detectable BRCA1/BRCA2 pathogenic variant), cancer-specific and general distress were assessed 2 weeks before surgery and at 6 and 12 months postsurgery.
The sample included women who had RRM and RRSO alone and women who had both surgeries. There was no observable increase in distress over the 1-year period.
Mixed psychosocial outcomes were reported in a follow-up study (mean 14 years) of 609 women who received RRM at the Mayo Clinic. Seventy percent were satisfied with RRM, 11% were neutral, and 19% were dissatisfied. Eighteen percent believed that if they had the choice to make again, they probably or definitely would not have an RRM. About three-quarters said their worry about cancer was diminished by surgery. One-half reported no change in their satisfaction with body image; 16% reported improved body image after surgery. Thirty-six percent said they were dissatisfied with their body image after RRM. About one-quarter of the women reported adverse impact of RRM on their sexual relationships and sense of femininity, and 18% had diminished self-esteem. Factors most strongly associated with satisfaction with RRM were postsurgical satisfaction with appearance, reduced stress, no reconstruction or lack of problems with implants, and no change or improvement in sexual relationships. Women who cited physician advice as the primary reason for choosing RRM tended to be dissatisfied after RRM.
A study of 60 healthy women who underwent RRM measured levels of satisfaction, body image, sexual functioning, intrusion and avoidance, and current psychological status at a mean of 4 years and 4 months postsurgery. Of this group, 76.7% had either a strong family history (21.7%) or carried a BRCA1 or BRCA2 pathogenic variant (55%). Overall, 97% of the women surveyed were either satisfied (17%) or extremely satisfied (80%) with their decision to have RRM, and all but one participant would recommend this procedure to other women. Most women (66.7%) reported that surgery had no impact on their sexual life, although 31.7% reported a worsening sexual life, and 76.6% reported either no change in body image or an improvement in body image, regardless of whether reconstruction was performed. Worsening self-image was reported by 23.3% of women after surgery. Women’s mean distress levels after surgery were only slightly above normal levels, although those women who continued to perceive their postsurgery breast cancer risk as high had higher mean levels of global and cancer-related distress than those who perceived their risk as low. Additionally, carriers of BRCA1 and BRCA2 pathogenic variants and women with a strong family history of breast and/or ovarian cancer had higher mean levels of cancer-related distress than women with a limited family history.
Very little is known about how the results of genetic testing affect treatment decisions at the time of cancer diagnosis. Two studies explored genetic counseling and BRCA1/BRCA2 genetic testing at the time of breast cancer diagnosis.
One of these studies found that genetic testing at the time of diagnosis significantly altered surgical decision making, with more pathogenic variant carriers than noncarriers opting for bilateral mastectomy. Bilateral RRM was chosen by 48% of women with a known pathogenic variant and by 100% of women with a known pathogenic variant in a smaller series
of women undergoing testing at the time of diagnosis. Of women in whom no pathogenic variant was found, 24% also opted for bilateral RRM. Four percent of the test decliners also underwent bilateral RRM. Among carriers of pathogenic variants, predictors of bilateral RRM included whether patients reported that their physicians had recommended BRCA1/BRCA2 testing and bilateral RRM before testing, and whether they received a positive test result.
Data are lacking on quality-of-life outcomes for women who undergo RRM after genetic testing that is performed at the time of diagnosis.
A prospective study from the Netherlands evaluated long-term psychological outcomes of offering women with breast cancer genetic counseling and, if indicated, genetic testing at the onset of breast radiation for treatment of their primary breast cancer. Of those who were approached for counseling, some underwent genetic testing and chose to receive their result (n = 58), some were approached but did not fulfill referral criteria (n = 118), and some declined the option of counseling/testing (n = 44). Another subset of women undergoing radiation therapy was not approached for counseling (n = 182) but was followed using the same measures. Psychological distress was measured at baseline and at 4, 11, 27, and 43 weeks after initial consultation for radiation therapy. No differences were detected in general anxiety, depression or breast cancer–specific distress across all four groups.
A retrospective questionnaire study of 583 women with a personal and family history of breast cancer and who underwent contralateral RRM between 1960 and 1993 measured overall satisfaction after mastectomy and factors influencing satisfaction and dissatisfaction with this procedure.
The mean time of follow-up was 10.3 years after risk-reducing surgery. Overall, 83% of all participants stated they were satisfied or very satisfied, 8% were neutral, and 9% were dissatisfied with contralateral RRM. Most women also reported favorable effects or no change in their self-esteem, level of stress, and emotional stability after surgery (88%, 83%, and 88%, respectively). Despite the high levels of overall satisfaction, 33% reported negative body image, 26% reported a reduced sense of femininity, and 23% reported a negative effect on sexual relationships. The type of surgical procedure also affected levels of satisfaction. The authors attributed this difference to the high rate of unanticipated reoperations in the group of women having subcutaneous mastectomy (43%) versus the group having simple mastectomy (15%) (P < .0001). Limitations to this study are mostly related to the time period during which participants had their surgery (i.e., availability of surgical reconstructive option).
None of these women had genetic testing for pathogenic variants in the BRCA1/BRCA2 genes. Nevertheless, this study shows that while most women in this group were satisfied with contralateral RRM, all women reported at least one adverse outcome.
A retrospective survey of 137 BRCA carriers examined the psychosocial impact of preserving the nipple-areolar complex (NAC) in women with bilateral RRM.
The study found that body image and sexual well-being differed significantly based on the type of RRM the women underwent. Women with NAC preservation were more satisfied with their breasts (72% vs. 61%), were more satisfied with the surgical outcome (85% vs. 74%), and had greater sexual well-being (68% vs. 52%) than women without NAC preservation. No differences in cancer-related distress, anxiety, depression, or risk perceptions were observed between the two groups. Oncologic outcomes of nipple-sparing mastectomy in BRCA carriers have not been inferior to RRM without NAC preservation.
(Refer to the RRM section of this summary for more information.)
Another study compared long-term quality-of-life outcomes in 195 women after bilateral RRM performed between 1979 and 1999 versus 117 women at high risk of breast cancer opting for screening. No statistically significant differences were detected between the groups for psychosocial outcomes. Eighty-four percent of those opting for surgery reported satisfaction with their decision. Sixty-one percent of women from both the surgery and screening groups reported being very much or quite a bit contented with their quality of life.
In a study of psychosocial outcomes associated with RRM and immediate reconstruction, 61 high-risk women (27 carriers of pathogenic variants, others with high-risk family history), 31 of whom had a prior history of breast cancer, were evaluated on average 3 to 4 years after surgery.
The study utilized questions designed to elicit yes versus no responses and found that the surgery was well-tolerated with 83% of participants reporting that the results of their reconstructive surgery were as they expected, 90% reporting that they had received adequate preoperative information, none reporting that they regretted the surgery, and all reporting that they would choose the same route if they had to do it again. Satisfaction with the results ranged from 74% satisfied with the shape of their breasts to 89% satisfied with the appearance of the scarring. Comparison of this group to normative samples on quality-of-life indicators (Short Form 36 Health Survey Questionnaire [SF-36]; Hospital Anxiety and Depression Scale questionnaire scores) indicated no reductions in quality of life in these women.
A qualitative study examining material on the FORCE website posted by 21 high-risk women (BRCA1/BRCA2 positive) undergoing RRM showed that these women anticipated and received negative reactions from friends and family regarding the surgery, and that they managed disclosure in ways to maintain emotional support and self-protection for their decision. Many of the women expressed a relief from intrusive breast cancer thoughts and worry, and were satisfied with the cosmetic result of their surgery.
In contrast, another study examined long-term psychosocial outcomes in 684 women who had had bilateral or contralateral RRM on average 9 years before assessment.
A majority of women (59%) also had reconstructive surgery. Interestingly, based on a Likert scale, 85% of women reported that they were satisfied or very satisfied with their decision to have an RRM. However, in qualitative interviews, a large number of women went on to describe dissatisfaction or negative psychosocial outcomes associated with surgery. The authors coded the responses as negative when women reported still being anxious about their breast cancer risk and/or reported negative feelings about their body image, pain, and sexuality. Seventy-nine percent of the women providing negative comments and 84% of those making mixed comments (mixture of satisfaction and dissatisfaction) responded that they were either satisfied or very satisfied with their decision. Twice as many women with bilateral mastectomy made negative and mixed comments than did women with contralateral mastectomy. The areas of most concern were body image, problems with breast implants, pain after surgery, and sexuality. The authors proposed that those who had undergone contralateral procedures had already been treated for cancer, while those who had undergone bilateral procedures had not been treated previously, and this may partially account for the differences in satisfaction between the two groups. These findings suggest that women's satisfaction with RRM may be tempered by their complex reactions over time.
In a qualitative study of 108 women who underwent or were considering RRM, more than half of those who had RRM felt that presurgical consultation with a psychologist was advisable; nearly two-thirds thought that postsurgical consultation was also appropriate. All of the women who were considering RRM believed that psychological consultation before surgery would facilitate decision-making.
A retrospective self-administered survey of 40 women aged 35 to 74 years at time of RRSO (57.5% were younger than 50 y), who had undergone the procedure through the Ontario Ministry of Health due to a family history of ovarian cancer, found that RRSO resulted in a significant reduction in perceived ovarian cancer risk. Fifty-seven percent identified a decrease in perceived risk as a benefit of RRSO (35% did not comment on RRSO benefits) and 49% reported that they would repeat RRSO to decrease cancer risk. The overall quality-of-life scores were consistent with those published for women who are menopausal or participating in hormone studies.
Quality of life in 59 women who underwent RRSO was assessed at 24 months postprocedure.
Overall quality of life was similar to the general population and breast cancer survivors, with approximately 20% reporting depression. The 30% of subjects reporting vaginal dryness and dyspareunia were more likely to report dissatisfaction with the procedure.
A Canadian prospective study examined the impact of RRSO on menopausal symptoms and sexual functioning before surgery and then 1 year later in a sample of 114 women with known BRCA1/BRCA2 pathogenic variants.
Satisfaction with the decision to undergo RRSO was high regardless of symptoms reported. Those who were premenopausal at the time of surgery (n = 75) experienced a worsening of symptoms and a decline in sexual functioning. HRT addressed vaginal dryness and dyspareunia but not declines in sexual pleasure. HRT also resulted in fewer moderate to severe hot flashes.
Additional work reported by this group found that the majority of the 127 women who had undergone RRSO 1 year previously (75 with BRCA1 pathogenic variants; 52 with BRCA2 pathogenic variants) felt that RRSO reduced their risk of both breast and ovarian cancer.
There was a wide range of risk perceptions for ovarian cancer noted in the group. Twenty percent of carriers of BRCA1 and BRCA2 pathogenic variants thought that their risk of ovarian cancer was completely eliminated; others had an inflated perception of their ovarian cancer risk both before and after surgery. A small group of these women were further surveyed at about 3 years postsurgery, and their risk perceptions did not change significantly during this extended time period. These findings suggest that important misperceptions about ovarian cancer risk may persist after RRSO. Additional genetic education and counseling may be warranted.
A larger study assessed quality of life in women at high risk of ovarian cancer who opted for periodic gynecologic screening (GS) versus those who underwent RRSO. Eight hundred forty-six high-risk women, 44% of whom underwent RRSO and 56% of whom chose GS, completed questionnaires evaluating quality of life, cancer-specific distress, endocrine symptoms, and sexual functioning.
Women in the RRSO group were a mean of 2.8 ±1.9 years from surgery and women in the GS group were a mean of 4.3 years from their first visit to a gynecologist for high-risk management. No statistical difference in overall quality of life was detected between the RRSO and GS groups. When compared with the GS group, women who underwent RRSO had poorer sexual functioning and more endocrine symptoms such as vaginal dryness, dyspareunia, and hot flashes. Women who underwent RRSO experienced lower levels of breast and ovarian cancer distress and had a more favorable perception of cancer risk.
Women (N = 182) at risk of hereditary breast and ovarian cancer referred for genetic counseling were surveyed concerning their satisfaction with their choice of either RRSO or periodic screening (PS) (biannual pelvic examination with TVUS and CA-125 determination) to manage their ovarian cancer risk.
Overall satisfaction with both options was extremely high, but highest among those who chose RRSO over PS. There were no other demographic or clinical factors that distinguished satisfaction level. There was higher decisional ambivalence among those who chose PS.
A retrospective study assessed 98 carriers of BRCA pathogenic variants who underwent RRSO about their preoperative counseling regarding symptoms to expect after surgery.
The mean age at RRSO was 45.5 years (range, 32–63 y). Eighty-five percent pursued RRSO after learning that they harbored a BRCA pathogenic variant, and 48.0% were premenopausal at the time of surgery. Participants reported ‘‘frequent’’ or ‘‘very frequent’’ postsurgical symptoms of vaginal dryness (52.1%), changes in interest in sex (50.0%), sleep disturbances (46.7%), changes in sex life (43.9%), and hot flashes (42.9%). Only vaginal dryness and hot flashes were commonly recalled to have been addressed preoperatively. While 96% would have the surgery again, participants reported that the discussion of the impact of surgery on their sex life (59.2%), risk of coronary heart disease (57.1%), and the availability of sex counseling (57.1%) would have been helpful.
A study
of screening behaviors of 216 self-referred, high-risk (>10% risk of carrying a BRCA1/BRCA2 pathogenic variant) women who are members of hereditary breast cancer families found a range of screening practices. Even the presence of known pathogenic variants in their families was not associated with good adherence to recommended screening practices. Sixty-nine percent of women aged 50 to 64 years and 83% of women aged 40 to 49 years had had a screening mammogram in the previous year. Twenty percent of participants had ever had a CA-125 test and 31% had ever had a pelvic ultrasound or TVUS. Further analysis of this study population
looking specifically at 107 women with informative BRCA test results found good use of breast cancer screening, though the uptake rate in younger carriers is lower. The reason for the lower uptake rate was not explored in this study.
One survey of screening behaviors among women at increased risk of breast/ovarian cancer identified physician recommendations as a significant factor in adherence to screening.
While motivations cited for pursuing genetic testing often include the expectation that carriers of pathogenic variants will be more compliant with breast and/or ovarian screening recommendations,
limited data exist about whether participants in genetic testing alter their screening behaviors over time and about other variables that may influence those behaviors, such as insurance coverage and physician recommendations or attitudes. The impact of cancer genetic counseling on screening behaviors was assessed in a U.K. study of 293 women followed for 12 months postcounseling at four cancer genetics clinics.
BSE, CBE, and mammography were significantly increased after counseling; however, gaps in adherence to recommendations were noted: 38% of women aged 35 to 49 years had not had a mammogram by 12 months postcounseling. BSE was not done by most women at the recommended time and frequency.
This is a critical issue not only for women testing positive, but also for adherence to screening for those testing negative and those who have received indeterminate results or choose not to receive their results. It is possible that adherence actually diminishes with a decrease in the perceived risk that may result from a negative genetic test result.
In addition, while there is still some question regarding the link between cancer-related worry and breast cancer screening behavior, accumulating evidence appears to support a linear rather than a curvilinear relationship. That is, for some time, the data were not consistent; some data supported the hypothesis that mild-to-moderate worry may increase adherence, while excessive worry may actually decrease the utilization of recommended screening practices. Other reports support the notion that a linear relationship is more likely; that is, more worry increases adherence to screening recommendations. Few studies, however, have followed women to assess their health behaviors after genetic testing. Thus, a negative test result leading to decreased worry could theoretically result in decreased screening adherence. A large study found that patient compliance with screening practices was not related to general or screening-specific anxiety—with the exception of BSE, for which compliance is negatively associated with procedure-specific anxiety.
Further research designed to clarify this potential concern would highlight the need for comprehensive genetic counseling to discuss the need for follow-up screening.
Further complicating this area of research are issues such as the baseline rate of mammography adherence among women older than 40 or 50 years before genetic testing. More specifically, the ability to note a significant difference in adherence on this measure may be affected by the high adherence rate to this screening behavior before genetic testing by women undergoing such testing. It may be easier to find significant changes in mammography use among women with a family history of breast cancer who test positive. Finally, adherence over time will likely be affected by how women undergoing genetic testing and their caregivers perceive the efficacy of many of the screening options in question, such as mammography for younger women, BSE, and ovarian cancer screening (periodic vaginal ultrasound and serum CA-125 measurements), along with the value of preventive interventions.
The issue of screening decision-making and adherence among women undergoing genetic testing for breast and ovarian cancer is the subject of several ongoing trials, and an area of much needed ongoing study.
PDQ癌症信息定期评估和及时更新最新内容。这一部分会收录相关内容的最新信息(截至更新日期)。
Added Eoh et al.参考文献181
补充文字说明,一项大型研究探索生殖系统和生活方式的已知风险因素是否与多基因风险评分相互作用,导致乳腺癌风险增加,结果未发现既定风险因素的多重交互作用(引自Rudolph文章。参考文献48)。
已对单纯化疗与BRCA致病性突变的内容进行广泛修正。
修订文本指出,现有多项临床试验对PARP抑制剂在早期癌和转移癌中的应用进行研究,包括对PARP抑制剂单药治疗、PARP抑制剂联合其他DNA损伤修复剂、免疫治疗及其他靶向治疗联合应用以提高临床疗效,以及扩大潜在受益的临床患者群体。
附加文本指出,在一项为新诊乳腺癌患者提供前瞻性快速基因咨询和常规诊疗比较的随机试验中,结果显示尽管干预组的基因咨询接受率较高,但这并不能转化为较高的基因检测率,术前或双侧乳腺切除术决定前的检测结果采纳率(引自Schwartz文章。参考文献31)。
摘要由PDQ癌症遗传学编辑委员会撰写和维护,该委员会独立于NCI。 本篇内容的选取立场公正,不代表NCI和NIH任何政治观点。 有关摘要政策以及PDQ编辑委员会在维护PDQ摘要方面的作用的更多信息,请参见“关于本PDQ摘要”和PDQ®-NCI的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.
Added Eoh et al. as reference 181.
Added text to state that a large study examined whether known reproductive and lifestyle risk factors interact with polygenic risk scores to increase breast cancer risk and did not find a multiplicative interaction with established risk factors (cited Rudolph et al. as reference 48).
The Chemotherapy alone and BRCA pathogenic variants subsection was extensively revised.
Revised text to state that trials, both in the early and metastatic settings, are evaluating single-agent poly (ADP-ribose) polymerase (PARP) inhibitors and combining PARP inhibitors with other DNA damage repair agents, immunotherapies, and other targeted therapies to improve responses, as well as broaden the patient population who may benefit.
Added text to state that in a randomized trial that provided proactive rapid genetic counseling compared with usual care for patients with newly diagnosed breast cancer, results suggested that although genetic counseling uptake was higher in the intervention arm, this did not translate into higher rates of genetic testing, receipt of results before surgery, or bilateral mastectomy decisions (cited Schwartz et al. as reference 31).
This summary is written and maintained by the PDQ Cancer Genetics 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.
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the genetics of breast and gynecologic cancers. It is intended as a resource to inform and assist clinicians who care for cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.
This summary is reviewed regularly and updated as necessary by the PDQ Cancer Genetics Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Genetics of Breast and Gynecologic Cancers are:
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Cancer Genetics Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Cancer Genetics Editorial Board. PDQ Genetics of Breast and Gynecologic Cancers. Bethesda, MD: National Cancer Institute. Updated
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the genetics of breast and gynecologic cancers. It is intended as a resource to inform and assist clinicians who care for cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.
This summary is reviewed regularly and updated as necessary by the PDQ Cancer Genetics Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Genetics of Breast and Gynecologic Cancers are:
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Cancer Genetics Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Cancer Genetics Editorial Board. PDQ Genetics of Breast and Gynecologic Cancers. Bethesda, MD: National Cancer Institute. Updated
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.